151
|
Tacheva T, Zienolddiny-Narui S, Dimov D, Vlaykova D, Miteva I, Vlaykova T. The Leucocyte Telomere Length, GSTM1 and GSTT1 Null Genotypes and the Risk of Chronic Obstructive Pulmonary Disease. Curr Issues Mol Biol 2022; 44:3757-3769. [PMID: 36005153 PMCID: PMC9406937 DOI: 10.3390/cimb44080257] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/15/2022] [Accepted: 08/17/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Chronic obstructive pulmonary disease (COPD) is characterized by chronic inflammation and oxidative stress, both in the airways and blood, and in other organs. Elevated oxidative stress and inflammation have been reported to affect leucocyte telomere length (LTL). We explored the link between GSTM1 and GSTT1 gene polymorphisms, LTL and COPD risk. For GSTM1 and GSTT1, we genotyped 152 COPD patients and 131 non-affected controls, while for TL, we assessed 91 patients and 88 controls. There was a significant difference in GSTM1 null genotype frequency between the patients and controls (0.59 vs. 0.38, p ≤ 0.000), but such was not found for GSTT1 (p = 0.192). COPD patients carrying the GSTM1 null genotype had shorter telomeres compared to those carrying the non-null genotype (15,720 bp vs. 22,442 bp, p = 0.008); and in controls, the opposite occurred (31,354 bp vs. 17,800 bp, p = 0.020). According to our results GSTM1, but not GSTT1, null genotypes might play role in leucocyte telomere shortening, and thus be involved in the pathogenesis of COPD. Abstract Chronic obstructive pulmonary disease (COPD) is characterized by chronic inflammation and oxidative stress both in the airways and blood and other organs. Elevated oxidative stress and inflammation have been reported to affect leucocyte telomere length (LTL). Glutathione S-transferase (GST) enzymes are a large family of xenobiotic-metabolizing enzymes that utilize different ROS products. We aimed to explore the link between GSTM1 and GSTT1 gene polymorphisms, LTL and COPD risk. For GSTM1, we genotyped 152 COPD patients and 131 non-affected controls; for GSTT1, we genotyped 149 COPD patients and 130 controls. We were able to assess TL for 91 patients and 88 controls. There was a significant difference in the GSTM1 null genotype frequency between the patients and controls (0.59 vs. 0.38, p ≤ 0.000), but such was not found for GSTT1 (p = 0.192). When combining both polymorphisms, we obtained a significantly greater presence of at least one null genotype among patients (0.12 vs. 0.05, p = 0.027). An association between GSTT1 and LTL was not found. COPD patients carrying the GSTM1 null genotype had shorter telomeres compared to those carrying the non-null genotype (15,720 bp vs. 22,442 bp, p = 0.008); as for the controls, it was the opposite (31,354 bp vs. 17,800 bp, p = 0.020). The significance in both groups remained when combining GSTM1 and GSTT1 (COPD (at least one null) 16,409 bp vs. COPD (non-null) 22,092 bp, p = 0.029; control (at least one null) 29,666 bp vs. control (non-null) 16,370 bp, p = 0.027). The total glutathione level in GSTM1 non-null controls was higher compared to the null genotype (15.39 ng/mL vs. 5.53 ng/mL, p = 0.002). In COPD patients, we found no association (p = 0.301). In conclusion, according to our results, GSTM1, but not GSTT1, null genotypes might play a role in leucocyte telomere shortening, and thus be involved in the pathogenesis of COPD.
Collapse
Affiliation(s)
- Tanya Tacheva
- Department of Medical Chemistry and Biochemistry, Medical Faculty, Trakia University, 6000 Stara Zagora, Bulgaria
| | - Shanbeh Zienolddiny-Narui
- Section for Toxicology and Biological Work Environment, National Institute of Occupational Health, NO-036 Oslo, Norway
| | - Dimo Dimov
- Department of Medical Chemistry and Biochemistry, Medical Faculty, Trakia University, 6000 Stara Zagora, Bulgaria
| | - Denitsa Vlaykova
- Department of Medical Chemistry and Biochemistry, Medical Faculty, Trakia University, 6000 Stara Zagora, Bulgaria
| | - Iva Miteva
- Department of Occupational Medicine, Faculty of Public Health, Medical University-Sofia, 1431 Sofia, Bulgaria
| | - Tatyana Vlaykova
- Department of Medical Chemistry and Biochemistry, Medical Faculty, Trakia University, 6000 Stara Zagora, Bulgaria
- Department of Medical Biochemistry, Medical University-Plovdiv, 4002 Plovdiv, Bulgaria
- Correspondence:
| |
Collapse
|
152
|
Lai J, Feng S, Xu S, Liu X. Effects of oral anticoagulant therapy in patients with pulmonary diseases. Front Cardiovasc Med 2022; 9:987652. [PMID: 36035947 PMCID: PMC9399807 DOI: 10.3389/fcvm.2022.987652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 07/26/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundTo evaluate the effect of oral anticoagulants (OACs) therapy, including vitamin K antagonist (VKA) and direct oral anticoagulants (DOAC) in patients with pulmonary diseases.MethodsLiterature from PubMed, MEDLINE, and Cochrane Library were screened until June 2022. Studies assessing OACs for pulmonary hypertension (PH), pulmonary embolism (PE), pulmonary fibrosis (PF), or chronic obstructive pulmonary disease (COPD) were evaluated for inclusion.ResultsOur study indicated that in patients with PH, PE, and COPD, OACs could significantly reduce the mortality risk, and the effects of VKA and DOACs without statistical difference in reducing the risk of recurrent embolism events. In patients with sclerosis-associated pulmonary arterial hypertension (SSc-PAH) or idiopathic pulmonary fibrosis (IPF), vitamin K antagonist (warfarin) significantly increased the mortality risk, while DOACs were not. As for the safety outcome of OACs, existing studies indicate that compared with patients treated with warfarin, the users of DOAC have a lower risk of major bleeding, while there is no statistical significance between them in non-major bleeding events. In current guidelines, the anticoagulation regimen for patients with pulmonary disease has not been defined. The results of our study confirm that DOACs (apixaban, rivaroxaban, dabigatran, and edoxaban) are superior to VKAs in the efficacy and safety outcomes of patients with pulmonary disease.ConclusionsOral anticoagulant therapy brings benefits to patients with PH, PE, or COPD, while the anticoagulation regimen for patients with SSc-PAH or IPF requires serious consideration. Compared with VKA, DOAC is a non-inferior option for anticoagulation in pulmonary disease treatment. Further studies are still needed to provide more reliable evidence about the safety outcome of pulmonary disease anticoagulation.
Collapse
Affiliation(s)
- Jiying Lai
- Department of Critical Care Medicine, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Shenghui Feng
- Queen Mary School, Medical Department, Nanchang University, Nanchang, China
| | - Shuo Xu
- Department of Respiratory and Critical Care Medicine, The Ganzhou People's Hospital, The Affiliated Ganzhou Hospital of Nanchang University, Ganzhou, China
- Shuo Xu
| | - Xin Liu
- Department of Critical Care Medicine, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- *Correspondence: Xin Liu
| |
Collapse
|
153
|
Gregory A, Xu Z, Pratte K, Lee S, Liu C, Chase R, Yun J, Saferali A, Hersh CP, Bowler R, Silverman E, Castaldi PJ, Boueiz A. Clustering-based COPD subtypes have distinct longitudinal outcomes and multi-omics biomarkers. BMJ Open Respir Res 2022; 9:9/1/e001182. [PMID: 35999035 PMCID: PMC9403129 DOI: 10.1136/bmjresp-2021-001182] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 07/31/2022] [Indexed: 11/25/2022] Open
Abstract
Introduction Chronic obstructive pulmonary disease (COPD) can progress across several domains, complicating the identification of the determinants of disease progression. In our previous work, we applied k-means clustering to spirometric and chest radiological measures to identify four COPD-related subtypes: ‘relatively resistant smokers (RRS)’, ‘mild upper lobe-predominant emphysema (ULE)’, ‘airway-predominant disease (AD)’ and ‘severe emphysema (SE)’. In the current study, we examined the associations of these subtypes to longitudinal COPD-related health measures as well as blood transcriptomic and plasma proteomic biomarkers. Methods We included 8266 non-Hispanic white and African-American smokers from the COPDGene study. We used linear regression to investigate cluster associations to 5-year prospective changes in spirometric and radiological measures and to gene expression and protein levels. We used Cox-proportional hazard test to test for cluster associations to prospective exacerbations, comorbidities and mortality. Results The RRS, ULE, AD and SE clusters represented 39%, 15%, 26% and 20% of the studied cohort at baseline, respectively. The SE cluster had the greatest 5-year FEV1 (forced expiratory volume in 1 s) and emphysema progression, and the highest risks of exacerbations, cardiovascular disease and mortality. The AD cluster had the highest diabetes risk. After adjustments, only the SE cluster had an elevated respiratory mortality risk, while the ULE, AD and SE clusters had elevated all-cause mortality risks. These clusters also demonstrated differential protein and gene expression biomarker associations, mostly related to inflammatory and immune processes. Conclusion COPD k-means subtypes demonstrate varying rates of disease progression, prospective comorbidities, mortality and associations to transcriptomic and proteomic biomarkers. These findings emphasise the clinical and biological relevance of these subtypes, which call for more study for translation into clinical practice. Trail registration number NCT00608764.
Collapse
Affiliation(s)
- Andrew Gregory
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Zhonghui Xu
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Katherine Pratte
- Department of Biostatistics, National Jewish Health, Denver, Colorado, USA
| | - Sool Lee
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Congjian Liu
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Robert Chase
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Jeong Yun
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA.,Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Aabida Saferali
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Craig P Hersh
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA.,Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Russell Bowler
- Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, Colorado, USA
| | - Edwin Silverman
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA.,Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Peter J Castaldi
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA.,General Medicine and Primary Care, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Adel Boueiz
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA .,Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| |
Collapse
|
154
|
Ma H, Zhang Q, Zhao Y, Zhang Y, Zhang J, Chen G, Tan Y, Zhang Q, Duan Q, Sun T, Qi C, Li F. Molecular and Clinicopathological Characteristics of Lung Cancer Concomitant Chronic Obstructive Pulmonary Disease (COPD). Int J Chron Obstruct Pulmon Dis 2022; 17:1601-1612. [PMID: 35860812 PMCID: PMC9293488 DOI: 10.2147/copd.s363482] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 06/25/2022] [Indexed: 11/23/2022] Open
Abstract
Introduction Chronic obstructive pulmonary disease (COPD) and lung cancer often coexist, but its pathophysiology and genomics features are still unclear. Methods In this study, we retrospectively collected lung cancer concomitant COPD (COPD-LC) and non-COPD lung cancer (non-COPD-LC) patients, who performed next generation sequencing (NGS) and had clinicopathological information simultaneously. The COPD-LC data from the TCGA cohort were collected to conduct further analysis. Results A total of 51 COPD-LC patients and 88 non-COPD-LC patients were included in the study. Clinicopathological analysis showed that proportion of male gender, older age, and smoking patients were all substantially higher in COPD-LC group than in non-COPD-LC group (all P<0.01). Comparing the genomic data of the two groups in our cohort, COPD-LC had higher mutation frequency of LRP1B (43% vs 9%, P = 0.001), EPHA5 (24% vs 1%, P = 0.002), PRKDC (14% vs 1%, P = 0.039), PREX2 (14% vs 0%, P = 0.012), and FAT1 (14% vs 0%, P = 0.012), which had a relationship with improved tumor immunity. Immunotherapy biomarker of PD-L1 positive expression (62.5% vs 52.0%, P = 0.397) and tumor mutation burden (TMB, median TMB: 7.09 vs 2.94, P = 0.004) also were higher in COPD-LC. In addition, RNA data from TCGA further indicated tumor immunity increased in COPD-LC. Whereas, COPD-LC had lower frequency of EGFR mutation (19% vs 50%, P = 0.013) and EGFR mutant COPD-LC treated with EGFR-TKI had worse progression-free survival (PFS) (HR = 3.52, 95% CI: 1.27–9.80, P = 0.01). Conclusion In this retrospective study, we first explored molecular features of COPD-LC in a Chinese population. Although COPD-LC had lower EGFR mutant frequency and worse PFS with target treatment, high PD-L1 expression and TMB indicated these patients may benefit from immunotherapy.
Collapse
Affiliation(s)
- Hongxia Ma
- Pneumology Department, The Fourth Affiliated Hospital of Xinjiang Medical University, Urumqi, The Xinjiang Uygur Autonomous Region, People's Republic of China
| | - Qian Zhang
- Pneumology Department, The Fourth Affiliated Hospital of Xinjiang Medical University, Urumqi, The Xinjiang Uygur Autonomous Region, People's Republic of China
| | - Yanwen Zhao
- Pneumology Department, The Fourth Affiliated Hospital of Xinjiang Medical University, Urumqi, The Xinjiang Uygur Autonomous Region, People's Republic of China
| | - Yaohui Zhang
- Pneumology Department, The Fourth Affiliated Hospital of Xinjiang Medical University, Urumqi, The Xinjiang Uygur Autonomous Region, People's Republic of China
| | - Jingjing Zhang
- Pneumology Department, The Fourth Affiliated Hospital of Xinjiang Medical University, Urumqi, The Xinjiang Uygur Autonomous Region, People's Republic of China
| | - Guoqing Chen
- Pneumology Department, The Fourth Affiliated Hospital of Xinjiang Medical University, Urumqi, The Xinjiang Uygur Autonomous Region, People's Republic of China
| | - Yuan Tan
- The Medical Department, Jiangsu Simcere Diagnostics Co., Ltd, Nanjing, Jiangsu Province, People's Republic of China.,Nanjing Simcere Medical Laboratory Science Co., Ltd, Nanjing, Jiangsu Province, People's Republic of China.,The State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co., Ltd, Nanjing, Jiangsu Province, People's Republic of China
| | - Qin Zhang
- The Medical Department, Jiangsu Simcere Diagnostics Co., Ltd, Nanjing, Jiangsu Province, People's Republic of China.,Nanjing Simcere Medical Laboratory Science Co., Ltd, Nanjing, Jiangsu Province, People's Republic of China.,The State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co., Ltd, Nanjing, Jiangsu Province, People's Republic of China
| | - Qianqian Duan
- The Medical Department, Jiangsu Simcere Diagnostics Co., Ltd, Nanjing, Jiangsu Province, People's Republic of China.,Nanjing Simcere Medical Laboratory Science Co., Ltd, Nanjing, Jiangsu Province, People's Republic of China.,The State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co., Ltd, Nanjing, Jiangsu Province, People's Republic of China
| | - Tingting Sun
- The Medical Department, Jiangsu Simcere Diagnostics Co., Ltd, Nanjing, Jiangsu Province, People's Republic of China.,Nanjing Simcere Medical Laboratory Science Co., Ltd, Nanjing, Jiangsu Province, People's Republic of China.,The State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co., Ltd, Nanjing, Jiangsu Province, People's Republic of China
| | - Chuang Qi
- The Medical Department, Jiangsu Simcere Diagnostics Co., Ltd, Nanjing, Jiangsu Province, People's Republic of China.,Nanjing Simcere Medical Laboratory Science Co., Ltd, Nanjing, Jiangsu Province, People's Republic of China.,The State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co., Ltd, Nanjing, Jiangsu Province, People's Republic of China
| | - Fengsen Li
- Pneumology Department, The Fourth Affiliated Hospital of Xinjiang Medical University, Urumqi, The Xinjiang Uygur Autonomous Region, People's Republic of China
| |
Collapse
|
155
|
TRAIL protects the immature lung from hyperoxic injury. Cell Death Dis 2022; 13:614. [PMID: 35840556 PMCID: PMC9287454 DOI: 10.1038/s41419-022-05072-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 06/30/2022] [Accepted: 07/05/2022] [Indexed: 02/07/2023]
Abstract
The hyperoxia-induced pro-inflammatory response and tissue damage constitute pivotal steps leading to bronchopulmonary dysplasia (BPD) in the immature lung. The pro-inflammatory cytokines are considered attractive candidates for a directed intervention but the complex interplay between inflammatory and developmental signaling pathways requires a comprehensive evaluation before introduction into clinical trials as studied here for the death inducing ligand TRAIL. At birth and during prolonged exposure to oxygen and mechanical ventilation, levels of TRAIL were lower in tracheal aspirates of preterm infants <29 weeks of gestation which developed moderate/severe BPD. These findings were reproduced in the newborn mouse model of hyperoxic injury. The loss of TRAIL was associated with increased inflammation, apoptosis induction and more pronounced lung structural simplification after hyperoxia exposure for 7 days while activation of NFκB signaling during exposure to hyperoxia was abrogated. Pretreatment with recombinant TRAIL rescued the developmental distortions in precision cut lung slices of both wildtype and TRAIL-/- mice exposed to hyperoxia. Of importance, TRAIL preserved alveolar type II cells, mesenchymal progenitor cells and vascular endothelial cells. In the situation of TRAIL depletion, our data ascribe oxygen toxicity a more injurious impact on structural lung development. These data are not surprising taking into account the diverse functions of TRAIL and its stimulatory effects on NFκB signaling as central driver of survival and development. TRAIL exerts a protective role in the immature lung as observed for the death inducing ligand TNF-α before.
Collapse
|
156
|
Zinellu A, Zinellu E, Pau MC, Carru C, Pirina P, Fois AG, Mangoni AA. A Comprehensive Systematic Review and Meta-Analysis of the Association between the Neutrophil-to-Lymphocyte Ratio and Adverse Outcomes in Patients with Acute Exacerbation of Chronic Obstructive Pulmonary Disease. J Clin Med 2022; 11:jcm11123365. [PMID: 35743436 PMCID: PMC9225466 DOI: 10.3390/jcm11123365] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/10/2022] [Accepted: 06/10/2022] [Indexed: 12/12/2022] Open
Abstract
The neutrophil-to-lymphocyte ratio (NLR) predicts adverse outcomes in stable chronic obstructive pulmonary disease (COPD); however, its prognostic role in acute exacerbations (AECOPD) is less clear. We conducted a systematic review and meta-analysis of the association between the NLR on admission and adverse outcomes (mortality, need for mechanical ventilation, transfer to the intensive care unit, length of stay, pulmonary hypertension, or their combination) in AECOPD by searching PubMed, Web of Science, and Scopus from inception to April 2022. Risk of bias and certainty of evidence were assessed using the Joanna Briggs Institute Critical Appraisal Checklist and the Grades of Recommendation, Assessment, Development, and Evaluation, respectively. In 15 studies (n = 10,038 patients), the NLR was significantly associated with the risk of adverse outcomes (odds ratio = 1.054, 95% CI 1.016 to 1.093, p = 0.005; low certainty of evidence; standard mean difference = 0.82, 95% CI 0.57 to 1.06, p < 0.001; high certainty of evidence). Pooled sensitivity, specificity, and area under the curve were 0.71 (95% CI 0.64 to 0.77), 0.73 (95% CI 0.65 to 0.80), and 0.78 (95% CI 0.74 to 0.81), respectively. In our study, the NLR on admission was significantly associated with adverse outcomes in AECOPD patients, suggesting the potential utility of this biomarker for early risk stratification and management in this group.
Collapse
Affiliation(s)
- Angelo Zinellu
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (A.Z.); (C.C.)
| | - Elisabetta Zinellu
- Clinical and Interventional Pneumology, University Hospital of Sassari (AOU), 07100 Sassari, Italy; (E.Z.); (P.P.); (A.G.F.)
| | - Maria Carmina Pau
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, 07100 Sassari, Italy;
| | - Ciriaco Carru
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (A.Z.); (C.C.)
- Quality Control Unit, University Hospital of Sassari (AOU), 07100 Sassari, Italy
| | - Pietro Pirina
- Clinical and Interventional Pneumology, University Hospital of Sassari (AOU), 07100 Sassari, Italy; (E.Z.); (P.P.); (A.G.F.)
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, 07100 Sassari, Italy;
| | - Alessandro G. Fois
- Clinical and Interventional Pneumology, University Hospital of Sassari (AOU), 07100 Sassari, Italy; (E.Z.); (P.P.); (A.G.F.)
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, 07100 Sassari, Italy;
| | - Arduino A. Mangoni
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia
- Department of Clinical Pharmacology, Flinders Medical Centre, Southern Adelaide Local Health Network, Bedford Park, SA 5042, Australia
- Correspondence:
| |
Collapse
|
157
|
Matarazzo L, Hernandez Santana YE, Walsh PT, Fallon PG. The IL-1 cytokine family as custodians of barrier immunity. Cytokine 2022; 154:155890. [DOI: 10.1016/j.cyto.2022.155890] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 03/31/2022] [Accepted: 04/13/2022] [Indexed: 12/12/2022]
|
158
|
Zhang HH, Zhou XJ, Zhong YS, Ji LT, Yu WY, Fang J, Ying HZ, Li CY. Naringin suppressed airway inflammation and ameliorated pulmonary endothelial hyperpermeability by upregulating Aquaporin1 in lipopolysaccharide/cigarette smoke-induced mice. Biomed Pharmacother 2022; 150:113035. [PMID: 35658207 DOI: 10.1016/j.biopha.2022.113035] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 04/19/2022] [Accepted: 04/25/2022] [Indexed: 11/25/2022] Open
Abstract
Naringin is one of the natural flavonoids extracted from many Chinese medicines. It ameliorates endothelial dysfunctions in atherosclerosis, diabetes, and cardiovascular diseases through free radical scavenging and antioxidant activities. The aim of the present study was to investigate the protective effects of naringin against pulmonary endothelial permeability in addition to airway inflammation in lipopolysaccharide/cigarette smoke (LPS/CS)-induced chronic obstructive pulmonary disease (COPD) mice.The COPD mice were exposed to LPS twice through intranasal inhalation and then to cigarette smoke daily for 6 weeks. The mice were orally administrated with naringin at doses of 40 or 80 mg/kg one hour before cigarette smoke exposure since the first day of the experiment. Naringin significantly alleviated pulmonary histopathological injury, and suppressed inflammatory cell infiltration and cytokine release in bronchoalveolar lavage fluid. Naringin decreased fluorescence intensity of Evans Blue in the lung tissues, and elevated the expression levels of tight junctional proteins. Meanwhile, naringin decreased neutrophil/lymphocyte/platelet counts and MDA content in blood, and upregulated Aquaporin1 (AQP1) in the lung tissues. However, the effect of naringin on airway inflammation and pulmonary endothelial permeability was inhibited in LPS/CS-treatment AQP1 deficiency mice. These results indicated that naringin attenuated LPS/CS-induced airway inflammatory and pulmonary hyperpermeability via upregulating AQP1 expression.
Collapse
Affiliation(s)
- Huan-Huan Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China; Zhejiang Provincial Laboratory of Experimental Animal's & Nonclinical Laboratory Studies, Hangzhou Medical College, Hangzhou, China
| | - Xiao-Jie Zhou
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yu-Sen Zhong
- Zhejiang Provincial Laboratory of Experimental Animal's & Nonclinical Laboratory Studies, Hangzhou Medical College, Hangzhou, China
| | - Li-Ting Ji
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Wen-Ying Yu
- Zhejiang Provincial Laboratory of Experimental Animal's & Nonclinical Laboratory Studies, Hangzhou Medical College, Hangzhou, China
| | - Jie Fang
- Zhejiang Provincial Laboratory of Experimental Animal's & Nonclinical Laboratory Studies, Hangzhou Medical College, Hangzhou, China
| | - Hua-Zhong Ying
- Zhejiang Provincial Laboratory of Experimental Animal's & Nonclinical Laboratory Studies, Hangzhou Medical College, Hangzhou, China.
| | - Chang-Yu Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
159
|
Yang YC, Zhang MY, Liu JY, Jiang YY, Ji XL, Qu YQ. Identification of Ferroptosis-Related Hub Genes and Their Association with Immune Infiltration in Chronic Obstructive Pulmonary Disease by Bioinformatics Analysis. Int J Chron Obstruct Pulmon Dis 2022; 17:1219-1236. [PMID: 35637927 PMCID: PMC9148178 DOI: 10.2147/copd.s348569] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 04/30/2022] [Indexed: 12/15/2022] Open
Abstract
Purpose Ferroptosis and immune infiltration are involved in the pathogenesis of chronic obstructive pulmonary disease (COPD). We aim to identify ferroptosis-related hub genes and analyze their association with immune infiltration in COPD through bioinformatics methods. Materials and Methods The mRNA microarray data of GSE38974 were downloaded from Gene Expression Omnibus to obtain differentially expressed genes (DEGs). The DEGs were intersected with ferroptosis-related genes (FRGs) from FerrDb to obtain differentially expressed FRGs. GO and KEGG enrichment and protein–protein interaction (PPI) analyses of differentially expressed FRGs were conducted in R software and STRING database. The key module and hub genes were screened by Cytoscape software. MiRNAs, transcription factors and signal molecules were predicted in miRNet and NetworkAnalyst. The disease correlation in the Comparative Toxicomics Database (CTD) and the receiver operating characteristic (ROC) curves of hub genes were analyzed. Immune infiltration was evaluated by CIBERSORT algorithm. Spearman correlation analyses were conducted between hub genes and differentially infiltrated immune cells. Results Fifteen differentially expressed FRGs were identified, which were enriched in some terms involving airway inflammatory responses and structural remodeling. Five hub genes were selected including HIF1A, IL6, PTGS2, CDKN1A and ATM. Inference scores in CTD indicated their association with COPD. Two miRNAs, five transcription factors and one signal molecule were predicted. The combination of hub genes could be a fine diagnostic indicator of COPD (AUC: 0.981, CI: 0.940-1.000). Immune infiltration evaluation showed that monocytes and M0 macrophages were upregulated in COPD lung tissues, while CD8 T cells, activated NK cells, M2 macrophages, resting dendritic cells and resting mast cells were downregulated. The hub genes were significantly associated with differentially infiltrated immune cells. Conclusion We identified five ferroptosis-related hub genes (HIF1A, IL6, PTGS2, CDKN1A and ATM) in COPD, and found that they may influence the pathogenesis of COPD by regulating ferroptosis and thus affecting infiltrating immune cells.
Collapse
Affiliation(s)
- Yi-Can Yang
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University; Shandong Key Laboratory of Infectious Respiratory Diseases, Jinan, People's Republic of China
| | - Meng-Yu Zhang
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University; Shandong Key Laboratory of Infectious Respiratory Diseases, Jinan, People's Republic of China
| | - Jian-Yu Liu
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University; Shandong Key Laboratory of Infectious Respiratory Diseases, Jinan, People's Republic of China
| | - Yuan-Yuan Jiang
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital of Shandong University; Shandong Key Laboratory of Infectious Respiratory Diseases, Jinan, People's Republic of China
| | - Xiu-Li Ji
- Department of Pulmonary Disease, Jinan Traditional Chinese Medicine Hospital, Jinan, People's Republic of China
| | - Yi-Qing Qu
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital of Shandong University; Shandong Key Laboratory of Infectious Respiratory Diseases, Jinan, People's Republic of China
| |
Collapse
|
160
|
Calzetta L, Pistocchini E, Ritondo BL, Cavalli F, Camardelli F, Rogliani P. Muscarinic receptor antagonists and airway inflammation: A systematic review on pharmacological models. Heliyon 2022; 8:e09760. [PMID: 35785239 PMCID: PMC9240991 DOI: 10.1016/j.heliyon.2022.e09760] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 05/18/2022] [Accepted: 06/17/2022] [Indexed: 12/05/2022] Open
Abstract
Airway inflammation is crucial in the pathogenesis of many respiratory diseases, including chronic obstructive pulmonary disease (COPD) and asthma. Current evidence supports the beneficial impact of muscarinic receptor antagonists against airway inflammation from bench-to-bedside. Considering the numerous sampling approaches and the ethical implications required to study inflammation in vivo in patients, the use of pre-clinical models is inevitable. Starting from our recently published systematic review concerning the impact of muscarinic antagonists, we have systematically assessed the current pharmacological models of airway inflammation and provided an overview on the advances in in vitro and ex vivo approaches. The purpose of in vitro models is to recapitulate selected pathophysiological parameters or processes that are crucial to the development of new drugs within a controlled environment. Nevertheless, immortalized cell lines or primary airway cells present major limitations, including the inability to fully replicate the conditions of the corresponding cell types within a whole organism. Induced animal models are extensively used in research in the attempt to replicate a respiratory condition reflective of a human pathological state, although considering animal models with spontaneously occurring respiratory diseases may be more appropriate since most of the clinical features are accompanied by lung pathology resembling that of the human condition. In recent years, three-dimensional organoids have become an alternative to animal experiments, also because animal models are unable to fully mimic the complexity of human pulmonary diseases. Ex vivo studies performed on human isolated airways have a superior translational value compared to in vitro and animal models, as they retain the morphology and the microenvironment of the lung in vivo. In the foreseeable future, greater effort should be undertaken to rely on more physiologically relevant models, that provide translational value into clinic and have a direct impact on patient outcomes.
Collapse
Affiliation(s)
- Luigino Calzetta
- Department of Medicine and Surgery, Respiratory Disease and Lung Function Unit, University of Parma, Parma, Italy
- Corresponding author.
| | - Elena Pistocchini
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome “Tor Vergata”, Rome, Italy
| | - Beatrice Ludovica Ritondo
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome “Tor Vergata”, Rome, Italy
| | - Francesco Cavalli
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome “Tor Vergata”, Rome, Italy
| | - Francesca Camardelli
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome “Tor Vergata”, Rome, Italy
| | - Paola Rogliani
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome “Tor Vergata”, Rome, Italy
| |
Collapse
|
161
|
Cazzola M, Ora J, Calzetta L, Rogliani P, Matera MG. Advances in inhaled corticosteroids for the treatment of chronic obstructive pulmonary disease: what is their value today? Expert Opin Pharmacother 2022; 23:917-927. [PMID: 35575510 DOI: 10.1080/14656566.2022.2076592] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION As of today, there is still a need to determine which COPD patients may benefit from ICS therapy, whether ICSs are useful in COPD patients without chronic bronchitis, and whether long-acting bronchodilators can reduce the risk of exacerbations in frequent exacerbators even if ICSs are not used, and whether combination therapy including ICSs is helpful in infrequent exacerbators to optimise the use of ICSs in COPD. Nevertheless, in recent years, a fair amount of evidence has been produced that, at least in part, can help define the role of ICSs in COPD better. AREAS COVERED Herein, the authors provide an overview of current use of ICS in COPD and discuss their value to the current treatment armamentarium. The article includes discussion of which patients will benefit best from the use of ICSs, their potential uses and adverse effects. EXPERT OPINION There is growing agreement on why, in whom, and when ICS therapy can be used in COPD, although the consensus is still lacking because of the heterogeneity of COPD. The use of blood eosinophil counts (BECs) is only helpful in T2 inflammation, while there is a lack of biomarkers indicating the presence of T1 and T17 immunity, which is poorly responsive to ICS. Identifying ICS-sensitive endotypes using specific biomarkers that have yet to be identified and validated is likely to demonstrate that ICSs can influence the natural course of COPD in at least a subset of patients.
Collapse
Affiliation(s)
- Mario Cazzola
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Josuel Ora
- Unit of Respiratory Medicine, "Tor Vergata" Hospital Foundation, Rome, Italy
| | - Luigino Calzetta
- Unit of Respiratory Diseases and Lung Function, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Paola Rogliani
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy.,Unit of Respiratory Medicine, "Tor Vergata" Hospital Foundation, Rome, Italy
| | - Maria Gabriella Matera
- Unit of Pharmacology, Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| |
Collapse
|
162
|
Effective-Component Compatibility of Bufei Yishen Formula III Combined with Electroacupuncture Suppresses Inflammatory Response in Rats with Chronic Obstructive Pulmonary Disease via Regulating SIRT1/NF- κB Signaling. BIOMED RESEARCH INTERNATIONAL 2022; 2022:3360771. [PMID: 35586807 PMCID: PMC9110177 DOI: 10.1155/2022/3360771] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 04/18/2022] [Indexed: 02/05/2023]
Abstract
Objective To explore more efficient treatments for chronic obstructive pulmonary disease (COPD), effective-component compatibility of Bufei Yishen formula III (ECC-BYF III) and electroacupuncture were tested on rats with COPD, and silent information regulator transcript-1 (SIRT1)/nuclear factor-kappaB (NF-κB) signaling was further investigated to interpret the therapy. Methods In total, 70 rats were randomly divided into control (Control), model (Model), aminophylline (APL), ECC-BYF III, electroacupuncture (EA), ECC-BYF III+EA, and sham electroacupuncture (SA) groups. Cigarette smoke exposure combined with repeated bacterial infections was used to establish COPD models in 1-12 weeks. From 13 to 20 weeks, the ECC-BYF III and APL groups received corresponding drugs; the EA group received electroacupuncture therapy, wherein Dazhui (GV 14), Feishu (BL 13), and Shenshu (BL 23) points were selected; the ECC-BYF III+EA group received ECC-BYF III intragastrically combined with electroacupuncture; and the SA group received simulated electroacupuncture (nonacupoint). Pulmonary function, pulmonary histopathology, the expressions of SIRT1/NF-κB signaling, and inflammation-related mRNA and protein were detected. Results Significant deterioration was observed in pulmonary function and pulmonary histopathology in rats with COPD (P < 0.01), and inflammatory state was illustrated by increased levels of interleukin- (IL-) 6 and tumor necrosis factor alpha (TNF-α) and decreased levels of IL-10 (P < 0.01). After the intervention of APL, ECC-BYF III, EA, and ECC-BYF III+EA, both pulmonary function and pulmonary histopathology were improved (P < 0.05 and P < 0.01), whereas the levels of IL-6 and TNF-α were decreased and IL-10 was increased (P < 0.05 and P < 0.01). Additionally, the mRNA expressions of IL-6, TNF-α, NF-κB, and acetylated NF-κBp65 (Ac-NF-κB) were noted to decrease, and SIRT1 and IL-10 were increased (P < 0.05 and P < 0.01); the protein expression of SIRT1 was upregulated, and NF-κBp65 and Ac-NF-κB were downregulated (P < 0.05 and P < 0.01). The effect of ECC-BYF III+EA was better in terms of improving pulmonary function and alleviating inflammation than that of the other treatment groups (P < 0.01 and P < 0.05). Conclusions ECC-BYF III, electroacupuncture, and their combination can suppress inflammation, among which the combination therapy has been proven to be the most effective treatment, and the mechanism may be involved in activating SIRT1/NF-κB signaling.
Collapse
|
163
|
Yousuf AJ, Mohammed S, Carr L, Yavari Ramsheh M, Micieli C, Mistry V, Haldar K, Wright A, Novotny P, Parker S, Glover S, Finch J, Quann N, Brookes CL, Hobson R, Ibrahim W, Russell RJ, John C, Grimbaldeston MA, Choy DF, Cheung D, Steiner M, Greening NJ, Brightling CE. Astegolimab, an anti-ST2, in chronic obstructive pulmonary disease (COPD-ST2OP): a phase 2a, placebo-controlled trial. THE LANCET. RESPIRATORY MEDICINE 2022; 10:469-477. [PMID: 35339234 DOI: 10.1016/s2213-2600(21)00556-7] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 12/03/2021] [Accepted: 12/06/2021] [Indexed: 12/17/2022]
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is a heterogeneous inflammatory airway disease. The epithelial-derived IL-33 and its receptor ST2 have been implicated in airway inflammation and infection. We aimed to determine whether astegolimab, a selective ST2 IgG2 monoclonal antibody, reduces exacerbations in COPD. METHODS COPD-ST2OP was a single-centre, randomised, double-blinded, placebo-controlled phase 2a trial in moderate-to-very severe COPD. Participants were randomly assigned (1:1) with a web-based system to received 490 mg subcutaneous astegolimab or subcutaneous placebo, every 4 weeks for 44 weeks. The primary endpoint was exacerbation rate assessed for 48 weeks assessed with a negative binomial count model in the intention-to-treat population, with prespecified subgroup analysis by baseline blood eosinophil count. The model was the number of exacerbations over the 48-week treatment period, with treatment group as a covariate. Safety was assessed in the whole study population until week 60. Secondary endpoints included Saint George's Respiratory Questionnaire for COPD (SGRQ-C), FEV1, and blood and sputum cell counts. The trial was registered with ClinicalTrials.gov, NCT03615040. FINDINGS The exacerbation rate at 48 weeks in the intention-to-treat analysis was not significantly different between the astegolimab group (2·18 [95% CI 1·59 to 2·78]) and the placebo group (2·81 [2·05 to 3·58]; rate ratio 0·78 [95% CI 0·53 to 1·14]; p=0·19]). In the prespecified analysis stratifying patients by blood eosinophil count, patients with 170 or fewer cells per μL had 0·69 exacerbations (0·39 to 1·21), whereas those with more than 170 cells per μL had 0·83 exacerbations (0·49 to 1·40). For the secondary outcomes, the mean difference between the SGRQ-C in the astegolimab group versus placebo group was -3·3 (95% CI -6·4 to -0·2; p=0·039), and mean difference in FEV1 between the two groups was 40 mL (-10 to 90; p=0·094). The difference in geometric mean ratios between the two groups for blood eosinophil counts was 0·59 (95% CI 0·51 to 0·69; p<0·001) and 0·25 (0·19 to 0·33; p<0·001) for sputum eosinophil counts. Incidence of treatment-emergent adverse events was similar between groups. INTERPRETATION In patients with moderate-to-very severe COPD, astegolimab did not significantly reduce exacerbation rate, but did improve health status compared with placebo. FUNDING Funded by Genentech and National Institute for Health Research Biomedical Research Centres.
Collapse
Affiliation(s)
- Ahmed J Yousuf
- Institute for Lung Health, National Institute for Health Research Biomedical Research Centre Respiratory Medicine, Department of Respiratory Sciences, University of Leicester, Leicester, UK
| | - Seid Mohammed
- Leicester Clinical Trials Unit, College of Life Sciences, University of Leicester, Leicester, UK
| | - Liesl Carr
- Institute for Lung Health, National Institute for Health Research Biomedical Research Centre Respiratory Medicine, Department of Respiratory Sciences, University of Leicester, Leicester, UK
| | - Mohammadali Yavari Ramsheh
- Institute for Lung Health, National Institute for Health Research Biomedical Research Centre Respiratory Medicine, Department of Respiratory Sciences, University of Leicester, Leicester, UK
| | - Claudia Micieli
- Institute for Lung Health, National Institute for Health Research Biomedical Research Centre Respiratory Medicine, Department of Respiratory Sciences, University of Leicester, Leicester, UK
| | - Vijay Mistry
- Institute for Lung Health, National Institute for Health Research Biomedical Research Centre Respiratory Medicine, Department of Respiratory Sciences, University of Leicester, Leicester, UK
| | - Kairobi Haldar
- Institute for Lung Health, National Institute for Health Research Biomedical Research Centre Respiratory Medicine, Department of Respiratory Sciences, University of Leicester, Leicester, UK
| | - Adam Wright
- Institute for Lung Health, National Institute for Health Research Biomedical Research Centre Respiratory Medicine, Department of Respiratory Sciences, University of Leicester, Leicester, UK
| | - Petr Novotny
- Institute for Lung Health, National Institute for Health Research Biomedical Research Centre Respiratory Medicine, Department of Respiratory Sciences, University of Leicester, Leicester, UK
| | - Sarah Parker
- Institute for Lung Health, National Institute for Health Research Biomedical Research Centre Respiratory Medicine, Department of Respiratory Sciences, University of Leicester, Leicester, UK
| | - Sarah Glover
- Institute for Lung Health, National Institute for Health Research Biomedical Research Centre Respiratory Medicine, Department of Respiratory Sciences, University of Leicester, Leicester, UK
| | - Joanne Finch
- Institute for Lung Health, National Institute for Health Research Biomedical Research Centre Respiratory Medicine, Department of Respiratory Sciences, University of Leicester, Leicester, UK
| | - Niamh Quann
- Leicester Clinical Trials Unit, College of Life Sciences, University of Leicester, Leicester, UK
| | - Cassandra L Brookes
- Leicester Clinical Trials Unit, College of Life Sciences, University of Leicester, Leicester, UK
| | - Rachel Hobson
- Leicester Clinical Trials Unit, College of Life Sciences, University of Leicester, Leicester, UK
| | - Wadah Ibrahim
- Institute for Lung Health, National Institute for Health Research Biomedical Research Centre Respiratory Medicine, Department of Respiratory Sciences, University of Leicester, Leicester, UK
| | - Richard J Russell
- Institute for Lung Health, National Institute for Health Research Biomedical Research Centre Respiratory Medicine, Department of Respiratory Sciences, University of Leicester, Leicester, UK
| | - Catherine John
- Department of Health Sciences, University of Leicester, Leicester, UK
| | | | | | | | - Michael Steiner
- Institute for Lung Health, National Institute for Health Research Biomedical Research Centre Respiratory Medicine, Department of Respiratory Sciences, University of Leicester, Leicester, UK
| | - Neil J Greening
- Institute for Lung Health, National Institute for Health Research Biomedical Research Centre Respiratory Medicine, Department of Respiratory Sciences, University of Leicester, Leicester, UK
| | - Christopher E Brightling
- Institute for Lung Health, National Institute for Health Research Biomedical Research Centre Respiratory Medicine, Department of Respiratory Sciences, University of Leicester, Leicester, UK.
| |
Collapse
|
164
|
Shoemark A, Shteinberg M, De Soyza A, Haworth CS, Richardson H, Gao Y, Perea L, Dicker AJ, Goeminne PC, Cant E, Polverino E, Altenburg J, Keir HR, Loebinger MR, Blasi F, Welte T, Sibila O, Aliberti S, Chalmers JD. Characterization of Eosinophilic Bronchiectasis: A European Multicohort Study. Am J Respir Crit Care Med 2022; 205:894-902. [PMID: 35050830 DOI: 10.1164/rccm.202108-1889oc] [Citation(s) in RCA: 92] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Rationale: Bronchiectasis is classically considered a neutrophilic disorder, but eosinophilic subtypes have recently been described. Objectives: To use multiple datasets available through the European Multicentre Bronchiectasis Audit and Research Collaboration to characterize eosinophilic bronchiectasis as a clinical entity focusing on the impact of eosinophils on bronchiectasis exacerbations. Methods: Patients were included from five countries to examine the relationships between blood eosinophil counts and clinical phenotypes after excluding coexisting asthma. 16S rRNA sequencing was used to examine relationships between eosinophil counts and the sputum microbiome. A post hoc analysis of the PROMIS (Inhaled Promixin in the Treatment of Non-Cystic Fibrosis Bronchiectasis) phase 2 trial was used to examine the impact of blood eosinophil counts on exacerbations in patients with Pseudomonas aeruginosa infection. Measurements and Main Results: A relationship between sputum and blood eosinophil counts was demonstrated in two cohorts. In analysis of 1,007 patients from five countries, 22.6% of patients had blood eosinophil counts of ⩾300 cells/μl. Counts of <100 cells/μl were associated with higher bronchiectasis severity and increased mortality. There was no clear relationship with exacerbations. Blood eosinophil counts of ⩾300 cells/μl were associated with both Streptococcus- and Pseudomonas-dominated microbiome profiles. To investigate the relationship of eosinophil counts with exacerbations after controlling for the confounding effects of infection, 144 patients were studied in a clinical trial after treatment with antipseudomonal antibiotics. Compared with patients with blood eosinophil counts of <100 cells/μl (reference), elevated eosinophil counts of 100-299 cells/μl (hazard ratio, 2.38; 95% confidence interval, 1.33-4.25; P = 0.003) and ⩾300 cells/μl (hazard ratio, 3.99; 95% confidence interval, 2.20-7.85; P < 0.0001) were associated with shorter time to exacerbation. Conclusions: Eosinophilic bronchiectasis affects approximately 20% of patients. After accounting for infection status, raised blood eosinophil counts are associated with shortened time to exacerbation.
Collapse
Affiliation(s)
- Amelia Shoemark
- Division of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, United Kingdom.,Royal Brompton Hospital and Imperial College London, London, United Kingdom
| | - Michal Shteinberg
- Pulmonology Institute and Cystic Fibrosis Center, Carmel Medical Center, Haifa, Israel
| | - Anthony De Soyza
- Population and Health Sciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom.,National Institute for Health Research Biomedical Research Centre for Ageing, Freeman Hospital, Newcastle, United Kingdom
| | - Charles S Haworth
- Cambridge Centre for Lung Infection, Royal Papworth Hospital, Cambridge, United Kingdom.,Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Hollian Richardson
- Division of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | - Yonghua Gao
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lidia Perea
- Hospital Clinic of Barcelona, University of Barcelona, Centro de Investigación Biomédica en Red Enfermedades Respiratorias, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Alison J Dicker
- Division of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | - Pieter C Goeminne
- Department of Respiratory Disease, AZ Nikolaas, Sint-Niklaas, Belgium
| | - Erin Cant
- Division of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | - Eva Polverino
- Pneumology Department, Hospital Universitari Vall d'Hebron, Barcelona, Spain.,Thorax Institute, Institute of Biomedical Research August Pi i Sunyer, University of Barcelona, Barcelona, Spain
| | - Josje Altenburg
- Department of Respiratory Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Holly R Keir
- Division of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | | | - Francesco Blasi
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Internal Medicine Department, Respiratory Unit and Cystic Fibrosis Adult Center, Milan, Italy.,Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Tobias Welte
- Department of Pulmonary Medicine and Infectious Diseases, Hannover University School of Medicine, Hannover, Germany
| | - Oriol Sibila
- Hospital Clinic of Barcelona, University of Barcelona, Centro de Investigación Biomédica en Red Enfermedades Respiratorias, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Stefano Aliberti
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy.,Respiratory Unit, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - James D Chalmers
- Division of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, United Kingdom
| |
Collapse
|
165
|
Somayaji R, Chalmers JD. Just breathe: a review of sex and gender in chronic lung disease. Eur Respir Rev 2022; 31:31/163/210111. [PMID: 35022256 DOI: 10.1183/16000617.0111-2021] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 08/20/2021] [Indexed: 01/08/2023] Open
Abstract
Chronic lung diseases are the third leading cause of death worldwide and are increasing in prevalence over time. Although much of our traditional understanding of health and disease is derived from study of the male of the species - be it animal or human - there is increasing evidence that sex and gender contribute to differences in disease risk, prevalence, presentation, severity, treatment approach, response and outcomes. Chronic obstructive pulmonary disease, asthma and bronchiectasis represent the most prevalent and studied chronic lung diseases and have key sex- and gender-based differences which are critical to consider and incorporate into clinical and research approaches. Mechanistic differences present opportunities for therapeutic development whereas behavioural and clinical differences on the part of patients and providers present opportunities for greater education and understanding at multiple levels. In this review, we seek to summarise the sex- and gender-based differences in key chronic lung diseases and outline the clinical and research implications for stakeholders.
Collapse
Affiliation(s)
- Ranjani Somayaji
- Dept of Medicine, Cumming School of Medicine, University of Calgary, Calgary, Canada .,Dept of Microbiology, Immunology and Infectious Disease, University of Calgary, Calgary, Canada.,Dept of Community Health Sciences, University of Calgary, Calgary, Canada
| | - James D Chalmers
- Division of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| |
Collapse
|
166
|
Milara J, Ballester B, de Diego A, Calbet M, Ramis I, Miralpeix M, Cortijo J. The pan-JAK inhibitor LAS194046 reduces neutrophil activation from severe asthma and COPD patients in vitro. Sci Rep 2022; 12:5132. [PMID: 35332239 PMCID: PMC8948298 DOI: 10.1038/s41598-022-09241-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 03/16/2022] [Indexed: 12/19/2022] Open
Abstract
Non-T2 severe asthma and chronic obstructive pulmonary disease (COPD) are airway chronic inflammatory disorders with a poor response to corticosteroids. LAS194046, a novel pan-Janus kinase (JAK) inhibitor, shows inhibitory effects on T2 allergic lung inflammation in rats. In this work we analyze the effects of LAS194046, fluticasone propionate and their combination in neutrophils from non-T2 severe asthma and COPD patients in vitro. Neutrophils from 23 healthy subjects, 23 COPD and 21 non-T2 severe asthma patients were incubated with LAS194046 (0.01 nM–1 µM), fluticasone propionate (0.1 nM–1 µM) or their combination and stimulated with lipopolysaccharide (LPS 1 µM). LAS194046 shows similar maximal % inhibition and potency inhibiting IL-8, MMP-9 and superoxide anion release in neutrophils from healthy, COPD and asthma. Fluticasone propionate suppresses mediator release only in neutrophils from healthy patients. The combination of LAS194046 with fluticasone propionate shows synergistic anti-inflammatory and anti-oxidant effects. The mechanisms involved in the synergistic effects of this combination include the increase of MKP1 expression, decrease of PI3Kδ, the induction of glucocorticoid response element and the decrease of ERK1/2, P38 and JAK2/STAT3 phosphorylation compared with monotherapies. In summary, LAS194046 shows anti-inflammatory effects in neutrophils from COPD and severe non-T2 asthma patients and induces synergistic anti-inflammatory effects when combined with fluticasone propionate.
Collapse
Affiliation(s)
- Javier Milara
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain. .,Pharmacy Unit, Consorcio Hospital General Universitario, Avenida tres cruces s/n, 46014, Valencia, Spain. .,CIBERES, Health Institute Carlos III, Valencia, Spain.
| | - Beatriz Ballester
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain.,CIBERES, Health Institute Carlos III, Valencia, Spain
| | - Alfredo de Diego
- Respiratory Unit, University and Polytechnic La Fe Hospital, Valencia, Spain
| | | | | | | | - Julio Cortijo
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain.,CIBERES, Health Institute Carlos III, Valencia, Spain.,Research and Teaching Unit, University General Hospital Consortium, Valencia, Spain
| |
Collapse
|
167
|
Tiew PY, Mac Aogáin M, Chotirmall SH. The current understanding and future directions for sputum microbiome profiling in chronic obstructive pulmonary disease. Curr Opin Pulm Med 2022; 28:121-133. [PMID: 34839338 DOI: 10.1097/mcp.0000000000000850] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Next-generation sequencing (NGS) has deepened our understanding of the respiratory microbiome in health and disease. The number of microbiome studies employing sputum as an airway surrogate has continued to increase over the past decade to include multiple large multicentre and longitudinal studies of the microbiome in chronic obstructive pulmonary disease (COPD). In this review, we summarize the recent advances to our understanding of the bacteriome, virome and mycobiome in COPD. RECENT FINDINGS Diverse microbiome profiles are reported in COPD. The neutrophilic Haemophilus-predominant bacteriome remains a prominent COPD phenotype, relatively stable over time and during exacerbations. Studies of the virome remain limited but reveal a potential involvement of viruses and bacteriophages particularly during COPD exacerbations and advancing disease severity. Mycobiome signatures, even in stable COPD are associated with poorer clinical outcomes including mortality. SUMMARY The sputum microbiome in COPD is being increasingly recognized for its clinical relevance, even in the stable state. Future studies integrating microbial kingdoms holistically (i.e. bacterial, viral and fungal) will provide deeper insight into its functionality including the relevance of microbial interactions and effect of treatment on microbiome-associated clinical outcomes.
Collapse
Affiliation(s)
- Pei Yee Tiew
- Department of Respiratory and Critical Care Medicine, Singapore General Hospital, Singapore
| | - Micheál Mac Aogáin
- Biochemical Genetics Laboratory, Department of Biochemistry, St. James's Hospital
- Clinical Biochemistry Unit, School of Medicine, Trinity College Dublin, Ireland
| | - Sanjay H Chotirmall
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| |
Collapse
|
168
|
Khan MMKS, Cole AG, Mannino DM. Precision medicine in chronic obstructive pulmonary disease: how far have we come? Curr Opin Pulm Med 2022; 28:115-120. [PMID: 34652296 DOI: 10.1097/mcp.0000000000000837] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW In this review, we will discuss the current status and recent developments in precision medicine in chronic obstructive pulmonary disease (COPD) through the lens of treatable traits. RECENT FINDINGS Although the term 'treatable traits' in the treatment of COPD is relatively recent, this concept has been used for many years if one considers interventions such as long-term oxygen therapy or alpha-1 antitrypsin replacement therapy. Recent advances have included expanding the definition of COPD to include a broader population of people with lower respiratory disease but not meeting the strict criteria for obstruction, advances in imaging to aid in the diagnosis and treatment of COPD, advances in understanding symptoms and exacerbations to define severity, using biomarkers to guide therapy and better understanding and addressing polymorbidity and frailty. In addition, there is a concerted effort to use these concepts to identify COPD patients earlier in the disease process wherein disease modification may be possible. SUMMARY Focusing on subsets of patients with COPD with certain characteristics should lead to better outcomes and fewer adverse effects from treatment. VIDEO ABSTRACT http://links.lww.com/COPM/A30.
Collapse
|
169
|
Haworth JJ, Pitcher CK, Ferrandino G, Hobson AR, Pappan KL, Lawson JLD. Breathing new life into clinical testing and diagnostics: perspectives on volatile biomarkers from breath. Crit Rev Clin Lab Sci 2022; 59:353-372. [PMID: 35188863 DOI: 10.1080/10408363.2022.2038075] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Human breath offers several benefits for diagnostic applications, including simple, noninvasive collection. Breath is a rich source of clinically-relevant biological information; this includes a volatile fraction, where greater than 1,000 volatile organic compounds (VOCs) have been described so far, and breath aerosols that carry nucleic acids, proteins, signaling molecules, and pathogens. Many of these factors, especially VOCs, are delivered to the lung by the systemic circulation, and diffusion of candidate biomarkers from blood into breath allows systematic profiling of organismal health. Biomarkers on breath offer the capability to advance early detection and precision medicine in areas of global clinical need. Breath tests are noninvasive and can be performed at home or in a primary care setting, which makes them well-suited for the kind of public screening program that could dramatically improve the early detection of conditions such as lung cancer. Since measurements of VOCs on breath largely report on metabolic changes, this too aids in the early detection of a broader range of illnesses and can be used to detect metabolic shifts that could be targeted through precision medicine. Furthermore, the ability to perform frequent sampling has envisioned applications in monitoring treatment responses. Breath has been investigated in respiratory, liver, gut, and neurological diseases and in contexts as diverse as infectious diseases and cancer. Preclinical research studies using breath have been ongoing for some time, yet only a few breath-based diagnostics tests are currently available and in widespread clinical use. Most recently, tests assessing the gut microbiome using hydrogen and methane on breath, in addition to tests using urea to detect Helicobacter pylori infections have been released, yet there are many more applications of breath tests still to be realized. Here, we discuss the strengths of breath as a clinical sampling matrix and the technical challenges to be addressed in developing it for clinical use. Historically, a lack of standardized methodologies has delayed the discovery and validation of biomarker candidates, resulting in a proliferation of early-stage pilot studies. We will explore how advancements in breath collection and analysis are in the process of driving renewed progress in the field, particularly in the context of gastrointestinal and chronic liver disease. Finally, we will provide a forward-looking outlook for developing the next generation of clinically relevant breath tests and how they may emerge into clinical practice.
Collapse
|
170
|
Efficacy and safety of modified Bushen Yiqi formulas (MBYF) as an add-on to formoterol and budesonide in the management of COPD: study protocol for a multicentre, double-blind, placebo-controlled, parallel-group, randomized clinical trial: FB-MBYF Trial. Trials 2022; 23:143. [PMID: 35164853 PMCID: PMC8842909 DOI: 10.1186/s13063-022-06057-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 01/27/2022] [Indexed: 11/16/2022] Open
Abstract
Background Inhaled glucocorticoid corticosteroid (ICS), long-acting β2-adrenoceptor agonist (LABA), and other drugs have limited therapeutic effects on COPD with significant individual differences. Traditional Chinese medicine (TCM)-modified Bushen Yiqi formula (MBYF) demonstrates advantages in COPD management in China. This study aims to evaluate the efficacy and safety of MBYF as an add-on to budesonide/formoterol in COPD patients and confirm the related genes affecting the therapeutic effect in the treatment of COPD. Methods In this multicentre, randomised, double-blind, placebo-controlled, parallel-group study, eligible patients with COPD will randomly receive a 360-day placebo or MBYF as an adjuvant to budesonide/formoterol in a 1:1 ratio and be followed up with every 2 months. The primary outcomes will be the frequency, times, and severity of acute exacerbation of COPD (AECOPD), COPD assessment test (CAT) score, and pulmonary function tests (PFTs). The secondary outcomes will include the modified Medical Research Council (mMRC) dyspnoea scale, 6-min walking test (6MWT), BODE index, quantitative scores of syndromes classified in TCM, inflammation indices, and hypothalamic-pituitary-adrenaline (HPA) axis function. We will also test the genotype to determine the relationship between drugs and efficacy. All the data will be recorded in case report forms (CRFs) and analysed by SPSS V.20.0. Discussion A randomized clinical trial design to evaluate the efficacy and safety of MBYF in COPD is described. The results will provide evidence for the combination therapy of modern medicine and TCM medicine, and individual therapy for COPD.Trial registration. Trial registration ID: ChiCTR1900026124, Prospective registration. Supplementary Information The online version contains supplementary material available at 10.1186/s13063-022-06057-7.
Collapse
|
171
|
Ma Y, He X, Liu X, Long Y, Chen Y. Endothelial Microparticles Derived from Primary Pulmonary Microvascular Endothelial Cells Mediate Lung Inflammation in Chronic Obstructive Pulmonary Disease by Transferring microRNA-126. J Inflamm Res 2022; 15:1399-1411. [PMID: 35250291 PMCID: PMC8896043 DOI: 10.2147/jir.s349818] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 02/12/2022] [Indexed: 12/14/2022] Open
Abstract
Background Extracellular vesicles (EVs) are considered to new types of intercellular communication media, and microRNA is one of the most common transferring components of EVs. This study aimed to explore the potential role of endothelial microparticles (EMPs) derived from primary pulmonary microvascular endothelial cells in regulating lung inflammation of chronic obstructive pulmonary disease (COPD) through transferring microRNA-126 (miR-126). Methods EMPs generated from primary pulmonary microvascular endothelial cells were isolated by gradient centrifugation and characterized by transmission electron microscopy, flow cytometry and Western blotting. EMPs were treated to in vitro and in vivo COPD models induced by cigarette smoke extract (CSE). miR-126 mimics or inhibitors were transfected into EMPs by calcium chloride. Pathological changes of lung tissue, mRNA and protein levels of inflammation-related factors were measured to explore the effect of EMPs transferring miR-126 on CSE-induced inflammation. Results Both in vitro and in vivo studies demonstrated that mRNA and protein levels of inflammation-related factors were significantly increased in COPD group, while EMPs could dramatically reverse these increases. In vitro, overexpression of miR-126 in EMPs decreased HMGB1 expression and magnified the decreasing effect of EMPs on inflammation-related factors. Conclusion The present study reveals that EMPs are capable of alleviating lung inflammation and transferring miR-126 can magnify the anti-inflammatory effect of EMPs, which may provide a novel therapeutic alternative for COPD.
Collapse
Affiliation(s)
- Yiming Ma
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital of Central South University, Changsha, People’s Republic of China
| | - Xue He
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital of Central South University, Changsha, People’s Republic of China
| | - Xiangming Liu
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital of Central South University, Changsha, People’s Republic of China
| | - Yingjiao Long
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital of Central South University, Changsha, People’s Republic of China
| | - Yan Chen
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital of Central South University, Changsha, People’s Republic of China
- Correspondence: Yan Chen; Yingjiao Long, Email ;
| |
Collapse
|
172
|
Petris OR, Cojocaru E, Fildan AP, Cojocaru C. COPD and Anticoagulation Therapy: Time for a New Approach? Int J Chron Obstruct Pulmon Dis 2021; 16:3429-3436. [PMID: 34955638 PMCID: PMC8694113 DOI: 10.2147/copd.s340129] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 12/06/2021] [Indexed: 12/11/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is one of the most challenging chronic disease nowadays due to increased morbidity and mortality, despite the multiple new therapies included in the therapeutic scheme. A possible cause may be insufficient approach to thromboembolic risk in these patients, scientific data being so far insufficient and relatively controversial. Areas covered: anticoagulant therapy is used mainly during severe exacerbations. There are data that have shown that therapy with low weight heparins injectable anticoagulants causes not only a reduction in thromboembolic risk but also an improvement in respiratory function parameters. Expert opinion: a number of COPD phenotypes are more prone to procoagulant status and thrombus formation. A layered approach to COPD patients in terms of antithrombotic prophylaxis is needed. Although current published clinical data have not provided irrefutable evidence, possibly due to the relatively heterogeneous approach to inclusion criteria, the frequent identification of autopsy holes in patients with COPD suggests that the high risk of mortality is due to specific bronchopulmonary changes and pulmonary embolism.
Collapse
Affiliation(s)
- Ovidiu Rusalim Petris
- Medical II Department, Faculty of Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, Iasi, 700115, Romania
| | - Elena Cojocaru
- Morpho-Functional Sciences II Department, Faculty of Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, Iasi, 700115, Romania
| | - Ariadna Petronela Fildan
- Internal Medicine 3rd Department, Faculty of Medicine, Ovidius University of Constanta, Constanta, 900527, Romania
| | - Cristian Cojocaru
- Medical III Department, Faculty of Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, Iasi, 700115, Romania
| |
Collapse
|
173
|
Xu X, Huang K, Dong F, Qumu S, Zhao Q, Niu H, Ren X, Gu X, Yu T, Pan L, Yang T, Wang C. The Heterogeneity of Inflammatory Response and Emphysema in Chronic Obstructive Pulmonary Disease. Front Physiol 2021; 12:783396. [PMID: 34950055 PMCID: PMC8689000 DOI: 10.3389/fphys.2021.783396] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 11/17/2021] [Indexed: 11/21/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a heterogeneous disease characterized by chronic inflammation, emphysema, airway remodeling, and altered lung function. Despite the canonical classification of COPD as a neutrophilic disease, blood and airway eosinophilia are found in COPD patients. Identifying the tools to assess eosinophilic airway inflammation in COPD models during stable disease and exacerbations will enable the development of novel anti-eosinophilic treatments. We developed different animal models to mimic the pathological features of COPD. Our results show that eosinophils accumulated in the lungs of pancreatic porcine elastase-treated mice, with emphysema arising from the alveolar septa. A lipopolysaccharide challenge significantly increased IL-17 levels and induced a swift change from a type-2 response to an IL-17-driven inflammatory response. However, lipopolysaccharides can exacerbate cigarette smoking-induced airway inflammation dominated by neutrophil infiltration and airway remodeling in COPD models. Our results suggest that eosinophils may be associated with emphysema arising from the alveolar septa, which may be different from the small airway disease-associated emphysema that is dominated by neutrophilic inflammation in cigarette smoke-induced models. The characterization of heterogeneity seen in the COPD-associated inflammatory signature could pave the way for personalized medicine to identify new and effective therapeutic approaches for COPD.
Collapse
Affiliation(s)
- Xia Xu
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Ke Huang
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Fen Dong
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Shiwei Qumu
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Qichao Zhao
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Hongtao Niu
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Xiaoxia Ren
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Xiaoying Gu
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Tao Yu
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Lin Pan
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Ting Yang
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Chen Wang
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
174
|
Tan ZX, Fu L, Wang WJ, Zhan P, Zhao H, Wang H, Xu DX. Serum CYR61 Is Associated With Airway Inflammation and Is a Potential Biomarker for Severity in Chronic Obstructive Pulmonary Disease. Front Med (Lausanne) 2021; 8:781596. [PMID: 34917638 PMCID: PMC8669148 DOI: 10.3389/fmed.2021.781596] [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: 09/23/2021] [Accepted: 11/08/2021] [Indexed: 12/02/2022] Open
Abstract
Background: Cysteine-rich 61 (CYR61) and inflammation was upregulated in the lungs of patients with chronic obstructive pulmonary disease (COPD). However, the association between CYR61 and inflammation was unclear in COPD patients. This study aimed to analyze the association of serum CYR61 with pulmonary inflammation and lung function indexes in COPD patients. Methods: One hundred and fifty COPD patients and 150 control subjects were enrolled. Serum and pulmonary CYR61 was detected. Lung function indexes were evaluated in COPD patients. Results: Serum CYR61 level was elevated and pulmonary CYR61 expression was upregulated in COPD patients. An increased CYR61 was associated with decreased pulmonary function indexes in COPD patients. Further analyses showed that nuclear factor-kappa B (NF-κB) p65-positive nuclei was elevated in the lungs of COPD patients with high level of CYR61. Accordingly, serum monocyte chemotactic protein (MCP)-1 and tumor necrosis factor α (TNF-α), two downstream inflammatory cytokines of NF-κB pathway, were increased in parallel with CYR61, among which serum MCP-1 and TNF-α were the highest in COPD patients with high level of CYR61. Moreover, a positive correlation, determined by multivariate regression that excluded the influence of age, gender and smoking, was observed between serum CYR61 and inflammatory cytokines in COPD patients. Conclusion: These results provide evidence that an increased CYR61 is associated with pulmonary inflammation and COPD progression. Inflammatory cytokines may be the mediators between CYR61 elevation and COPD progression.
Collapse
Affiliation(s)
- Zhu-Xia Tan
- Department of Toxicology, Anhui Medical University, Hefei, China
- Second Affiliated Hospital, Anhui Medical University, Hefei, China
| | - Lin Fu
- Second Affiliated Hospital, Anhui Medical University, Hefei, China
| | - Wen-Jing Wang
- Second Affiliated Hospital, Anhui Medical University, Hefei, China
| | - Ping Zhan
- Second Affiliated Hospital, Anhui Medical University, Hefei, China
| | - Hui Zhao
- Second Affiliated Hospital, Anhui Medical University, Hefei, China
| | - Hua Wang
- Department of Toxicology, Anhui Medical University, Hefei, China
| | - De-Xiang Xu
- Department of Toxicology, Anhui Medical University, Hefei, China
| |
Collapse
|
175
|
Alcázar-Navarrete B, Díaz-Lopez JM, García-Flores P, Ortega-Antelo M, Aguilar-Cruz I, Ruiz-Rodríguez O, Santiago-Diaz P, Romero Palacios PJ. T2 Biomarkers as Predictors of Exacerbations of Chronic Obstructive Pulmonary Disease. Arch Bronconeumol 2021; 58:595-600. [PMID: 35312535 DOI: 10.1016/j.arbres.2021.11.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 11/01/2021] [Accepted: 11/09/2021] [Indexed: 11/17/2022]
Abstract
INTRODUCTION Type 2 (T2) biomarkers such as blood eosinophil count (BEC) and FeNO have been related to a higher risk of exacerbations in COPD. It is unknown whether combining these biomarkers could be useful in forecasting COPD exacerbations. METHODS COPD patients were enrolled in this prospective, multicenter, observational study and followed up for 1 year, during which BEC were analysed at baseline (V0) while FeNO analyses were performed at baseline (V0), 6 months (V1) and 12 months (V2). The risk of moderate or severe exacerbation during follow up was assessed by Cox regression analysis, and the predictive capacity of both measurements was assessed by ROC curves and the DeLong test. Statistical significance was assumed at P<.05. RESULTS Of the 322 COPD patients initially recruited, 287 were followed up. At baseline, 28.0% were active smokers, and experienced moderate airflow limitation (mean FEV1 56.4%±17.0% predicted). Patients with at least one elevated T2 biomarker (n=125, 42.5%) were at increased risk of COPD exacerbation (HR 1.75, 95% CI 1.25-2.45, P=.001) and of shorter time to first COPD exacerbation. There was no difference between BEC and FeNO regarding the predictive capacity for moderate to severe exacerbation (AUC 0.584 vs 0.576, P=.183) but FeNO predicted severe episodes more accurately than BEC (AUC 0.607 vs 0.539, P<.05). Combining the two biomarkers enhanced the detection of moderate and severe COPD exacerbations. CONCLUSIONS Both eosinophil count and FeNO have limited utility for predicting COPD exacerbations. Combining these T2 biomarkers could enhance the detection of future COPD exacerbations.
Collapse
Affiliation(s)
- Bernardino Alcázar-Navarrete
- Respiratory Department, HU Virgen de las Nieves, Granada, Spain; Facultad de Medicina, Universidad de Granada, Granada, Spain; Centro de Investigación Biomédica en red Enfermedades Respiratorias (CIBERES), Spain.
| | | | | | | | | | - Oliverio Ruiz-Rodríguez
- Respiratory Department, AIG de Medicina, Hospital de Alta Resolución de Loja, Agencia Sanitaria Hospital de Poniente, Loja, Granada, Spain
| | - Pablo Santiago-Diaz
- Cardiology Department, AIG de Medicina, Hospital de Alta Resolución de Loja, Agencia Sanitaria Hospital de Poniente, Loja, Granada, Spain
| | | |
Collapse
|
176
|
Guifu Dihuang Pills Ameliorated Mucus Hypersecretion by Suppressing Muc5ac Expression and Inactivating the ERK-SP1 Pathway in Lipopolysaccharide/Cigarette Smoke-Induced Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:9539218. [PMID: 34777538 PMCID: PMC8580658 DOI: 10.1155/2021/9539218] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 09/29/2021] [Accepted: 10/18/2021] [Indexed: 11/30/2022]
Abstract
Mucus hypersecretion is a hallmark of chronic obstructive pulmonary disease (COPD) and is associated with increasing sputum production and declining pulmonary function. Therefore, reducing mucus secretion can be a new therapeutic opportunity for preventing COPD. The Guifu Dihuang pill (GFDHP) is a classical Chinese medicine and has been used as an immunoregulator for treatment of kidney yang deficiency syndrome, including hypothyroidism, adrenocortical hypofunction, chronic bronchitis, and COPD, for more than 2000 years. However, the protective effects and mechanisms of GFDHP against mucus hypersecretion in COPD remain obscure. The aim of the present study was to explore the inhibitory effects of GFDHP on lipopolysaccharide/cigarette smoke- (LPS/CS-) induced Mucin5ac (Muc5ac) overproduction and airway goblet cell hyperplasia in mice. The mice were randomly assigned into 6 groups: control, model, GFDHP-L, GFDHP-M, GFDHP-H, and dexamethasone. The mice were given LPS twice through intranasal inhalation and then exposed to CS daily for 6 weeks. Three doses of GFDHP were orally administered daily during the last 3 weeks of the experiment. Pulmonary function was examined with an EMKA pulmonary system, and pulmonary hyperpermeability and lung damage were evaluated with an in vivo imaging system. Inflammatory cells and cytokines in bronchoalveolar lavage fluid (BALF) were detected with a cell count analyzer and though ELISA analysis, respectively. Lung pathological changes and airway goblet cell hyperplasia were analyzed with hematoxylin and eosin and Alcian blue periodic acid Schiff staining. The protein expression levels of Muc5ac and extracellular signal-regulated kinase (ERK)-specificity protein1 (SP1) signaling pathway were measured with Western blot and immunohistochemistry. The results demonstrated that GFDHP improved pulmonary function and suppressed mouse pulmonary hyperpermeability and edema. GFDHP suppressed inflammatory cell infiltration and cytokine release in BALF, thereby elevating pulmonary function. It ameliorated lung pathological changes and airway goblet cell hyperplasia, and suppressed expression levels of Muc5ac mRNA and protein and phospho-ERK and SP1 levels in the lung tissues of the COPD mice. In conclusion, GFDHP inhibited mucus hypersecretion induced by LPS/CS by suppressing the activation of the ERK-SP1 pathway.
Collapse
|
177
|
Yang Y, Huang L, Tian C, Qian B. Magnesium isoglycyrrhizinate inhibits airway inflammation in rats with chronic obstructive pulmonary disease. BMC Pulm Med 2021; 21:371. [PMID: 34775946 PMCID: PMC8590971 DOI: 10.1186/s12890-021-01745-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 11/11/2021] [Indexed: 11/16/2022] Open
Abstract
Background Chronic obstructive pulmonary disease (COPD) is a kind of chronic lung diseases with the characteristics of airway remodeling and airflow obstruction. Magnesium isoglycyrrhizinate (MgIG) is an anti-inflammatory glycyrrhizic acid preparation for treating hepatitis. However, whether MgIG can treat other diseases and its action mechanism is still obscure. In this study, we evaluated the anti-inflammatory effect of MgIG in rats with COPD and investigated the underlying mechanisms. Methods Rat model of COPD was constructed by endotracheal-atomized lipopolysaccharide exposure and cigarette smoke induction. Rats were randomly divided into 5 groups: control group, COPD model group, salmeterol fluticasone comparator group, low dose of MgIG group, and high dose of MgIG group. Except for normal control group, the other four groups received sensitization treatment by cigarette smoking and endotracheal-atomization of endotoxin lipopolysaccharide to construct COPD rats model. After model established successfully, the COPD rats in each group received corresponding dose of endotracheal-atomized normal saline, salmeterol fluticasone, and MgIG every day prior to exposure of cigarette smoke from days 30 to 45. Normal control group were treated with normal saline. Finally, All rats were euthanatized. Pulmonary function was measured. Cells in bronchoalveolar lavage fluid were classified, inflammatory factors IL-6 and TNF-α were determined, histopathological analysis was performed by HE staining, and expression of NLRP3 and cleaved caspase-1 in the lung tissue was also determined by Western blotting. Results It showed that MgIG treatment (0.40 or 0.80 mg/kg/day) could recover the weight and the clinical symptoms of rats with COPD, accompanied with lung inflammation infiltration reduction, airway wall attenuation, bronchial mucus secretion reduction. Additionally, MgIG administration reduced inflammatory cells (white blood cells, neutrophils, lymphocytes and monocytes) accumulation in bronchoalveolar lavage fluid and decreased IL-6 and TNF-α production in the serum of COPD rats. Furthermore, MgIG treatment also reduced the expression level of NLRP3 and cleaved caspase-1. Conclusion It indicate that MgIG might be an alternative for COPD treatment, and its mechanism of action might be related to the suppression of NLRP3 inflammasome. Supplementary Information The online version contains supplementary material available at 10.1186/s12890-021-01745-7.
Collapse
Affiliation(s)
- Ye Yang
- Department of Pharmacology and Medicinal Chemistry, Jiangsu Vocational College of Medicine, Yancheng, 224005, Jiangsu, People's Republic of China
| | - Lei Huang
- Department of Pharmacology and Medicinal Chemistry, Jiangsu Vocational College of Medicine, Yancheng, 224005, Jiangsu, People's Republic of China
| | - Chongchong Tian
- Department of Pharmacology and Medicinal Chemistry, Jiangsu Vocational College of Medicine, Yancheng, 224005, Jiangsu, People's Republic of China
| | - Bingjun Qian
- Department of Pharmacology and Medicinal Chemistry, Jiangsu Vocational College of Medicine, Yancheng, 224005, Jiangsu, People's Republic of China.
| |
Collapse
|
178
|
Liu H, Tan X, Liu Z, Ma X, Zheng Y, Zhu B, Zheng G, Hu Y, Fang L, Hong G. Association Between Diet-Related Inflammation and COPD: Findings From NHANES III. Front Nutr 2021; 8:732099. [PMID: 34733875 PMCID: PMC8558221 DOI: 10.3389/fnut.2021.732099] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 09/23/2021] [Indexed: 02/01/2023] Open
Abstract
Background and Aims: Little is known about diet-related inflammation in chronic obstructive pulmonary disease (COPD). In this study, we aimed to explore the association between COPD and dietary inflammatory index (DII) scores in adults over 40 years old. Methods: Data were obtained from the 2013 to 2018 National Health and Nutrition Examination Survey (NHANES). In the present study, 9,929 participants were included and analyzed. The DII score was calculated and divided into tertiles. Logistic regression analysis was performed to determine the odds ratios of DII tertiles. Results: Participants were categorized into COPD (565, 5.69%) and non-COPD groups (9,364, 94.31%) according to interview information. COPD individuals had higher DII scores than non-COPD individuals (0.429 ± 1.809 vs. −0.191 ± 1.791, p < 0.001). The highest DII score tertile included 46.55% of COPD individuals was associated with lower family incomes and education and a higher smoking rate (p < 0.01). The odds ratios (95% CIs) of COPD according to logistic regression were 0.709 (0.512–0.982) for T1 and 0.645 (0.475–0.877) for T2 of the DII score (p = 0.011). Conclusion: Higher DII scores were positively correlated with COPD in participants over 40 years old. These results further support that diet can be used as an intervention strategy for COPD management.
Collapse
Affiliation(s)
- Haiyue Liu
- The Department of Laboratory Medicine, The First Affiliated Hospital of Xiamen University, Xiamen, China.,Xiamen Key Laboratory of Genetic Testing, Xiamen, China
| | - Xilan Tan
- Division of Infection Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Zuheng Liu
- Department of Cardiology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Xiaobo Ma
- The Department of Laboratory Medicine, The First Affiliated Hospital of Xiamen University, Xiamen, China.,Xiamen Key Laboratory of Genetic Testing, Xiamen, China
| | - Yanqing Zheng
- The Department of Laboratory Medicine, The First Affiliated Hospital of Xiamen University, Xiamen, China.,Xiamen Key Laboratory of Genetic Testing, Xiamen, China
| | - Bo Zhu
- The Department of Laboratory Medicine, The First Affiliated Hospital of Xiamen University, Xiamen, China.,Xiamen Key Laboratory of Genetic Testing, Xiamen, China
| | - Gangsen Zheng
- The Department of Laboratory Medicine, The First Affiliated Hospital of Xiamen University, Xiamen, China.,Xiamen Key Laboratory of Genetic Testing, Xiamen, China
| | - Yuehong Hu
- The Department of Laboratory Medicine, The First Affiliated Hospital of Xiamen University, Xiamen, China.,Xiamen Key Laboratory of Genetic Testing, Xiamen, China
| | - Lili Fang
- The Department of Laboratory Medicine, The First Affiliated Hospital of Xiamen University, Xiamen, China.,Xiamen Key Laboratory of Genetic Testing, Xiamen, China
| | - Guolin Hong
- The Department of Laboratory Medicine, The First Affiliated Hospital of Xiamen University, Xiamen, China.,Xiamen Key Laboratory of Genetic Testing, Xiamen, China
| |
Collapse
|
179
|
Keir HR, Chalmers JD. IL-6 trans-signalling: how Haemophilus surfs the NET to amplify inflammation in COPD. Eur Respir J 2021; 58:58/4/2102143. [PMID: 34649972 DOI: 10.1183/13993003.02143-2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 08/06/2021] [Indexed: 12/13/2022]
Affiliation(s)
- Holly R Keir
- Molecular and Clinical Medicine, University of Dundee, Dundee, UK
| | - James D Chalmers
- Molecular and Clinical Medicine, University of Dundee, Dundee, UK
| |
Collapse
|
180
|
Burgoyne RA, Fisher AJ, Borthwick LA. The Role of Epithelial Damage in the Pulmonary Immune Response. Cells 2021; 10:cells10102763. [PMID: 34685744 PMCID: PMC8534416 DOI: 10.3390/cells10102763] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 10/05/2021] [Accepted: 10/12/2021] [Indexed: 12/14/2022] Open
Abstract
Pulmonary epithelial cells are widely considered to be the first line of defence in the lung and are responsible for coordinating the innate immune response to injury and subsequent repair. Consequently, epithelial cells communicate with multiple cell types including immune cells and fibroblasts to promote acute inflammation and normal wound healing in response to damage. However, aberrant epithelial cell death and damage are hallmarks of pulmonary disease, with necrotic cell death and cellular senescence contributing to disease pathogenesis in numerous respiratory diseases such as idiopathic pulmonary fibrosis (IPF), chronic obstructive pulmonary disease (COPD) and coronavirus disease (COVID)-19. In this review, we summarise the literature that demonstrates that epithelial damage plays a pivotal role in the dysregulation of the immune response leading to tissue destruction and abnormal remodelling in several chronic diseases. Specifically, we highlight the role of epithelial-derived damage-associated molecular patterns (DAMPs) and senescence in shaping the immune response and assess their contribution to inflammatory and fibrotic signalling pathways in the lung.
Collapse
Affiliation(s)
- Rachel Ann Burgoyne
- Fibrosis Research Group, Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK;
| | - Andrew John Fisher
- Regenerative Medicine, Stem Cells and Transplantation Theme, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK;
- Institute of Transplantation, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE7 7DN, UK
| | - Lee Anthony Borthwick
- Fibrosis Research Group, Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK;
- Fibrofind, Medical School, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- Correspondence: ; Tel.: +44-191-208-3112
| |
Collapse
|
181
|
Zanthoxylum bungeanum Seed Oil Attenuates LPS-Induced BEAS-2B Cell Activation and Inflammation by Inhibiting the TLR4/MyD88/NF- κB Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:2073296. [PMID: 34603465 PMCID: PMC8486531 DOI: 10.1155/2021/2073296] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/13/2021] [Accepted: 09/08/2021] [Indexed: 11/17/2022]
Abstract
Background Zanthoxylum bungeanum seed oil (ZBSO) is a natural essential oil derived from the seeds of the Chinese medicinal plant Zanthoxylum bungeanum, which has been investigated for antitumor and anti-inflammatory effects. However, little is known regarding the effects of ZBSO in chronic obstructive pulmonary disease (COPD). Methods In this study, lung epithelial cells (BEAS-2B) were induced by lipopolysaccharide (LPS) to establish an in vitro model of COPD, and cytotoxicity was detected by a cell counting kit 8 (CCK-8) assay. Griess test, enzyme-linked immunosorbent assay (ELISA), reverse transcriptase quantitative polymerase chain reaction (RT-qPCR), western blot, immunofluorescence, and molecular docking analyses were used to investigate the effects of ZBSO and its potential mechanisms. Results The results showed that LPS promoted the expression of nitric oxide (NO), reactive oxygen species (ROS), malondialdehyde (MDA), tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), monocyte chemoattractant protein-1 (MCP-1), matrix metalloproteinase-2 (MMP-2), MMP-9, cyclooxygenase-2 (COX-2), and prostaglandin E2 (PGE2), suggesting that LPS can induce inflammation and oxidative stress in BEAS-2B cells. ZBSO inhibits the LPS-induced expression of inflammatory mediators and proinflammatory cytokines in BEAS-2B cells. The molecular docking results indicated that active components in ZBSO could successfully dock with toll-like receptor 4 (TLR4), myeloid differentiation factor 88 (MyD88), and p65. Immunofluorescence and western blot analyses further demonstrated that ZBSO repressed protein expression associated with the TLR4/MyD88/nuclear factor-κB (NF-κB) signaling pathway. Conclusions ZBSO reduced the inflammatory response and oxidative stress induced by LPS by inhibiting the TLR4/MyD88/NF-κB signaling pathway, thereby suppressing COPD. ZBSO may represent a promising therapeutic candidate for COPD treatment.
Collapse
|
182
|
Kammerl IE, Hardy S, Flexeder C, Urmann A, Peierl J, Wang Y, Vosyka O, Frankenberger M, Milger K, Behr J, Koch A, Merl-Pham J, Hauck SM, Pilette C, Schulz H, Meiners S. Activation of immune cell proteasomes in peripheral blood of smokers and COPD patients - implications for therapy. Eur Respir J 2021; 59:13993003.01798-2021. [PMID: 34561290 PMCID: PMC8891681 DOI: 10.1183/13993003.01798-2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 07/19/2021] [Indexed: 11/05/2022]
Abstract
Immune cells contain a specialised type of proteasome, i.e. the immunoproteasome, which is required for intracellular protein degradation. Immunoproteasomes are key regulators of immune cell differentiation, inflammatory activation and autoimmunity. Immunoproteasome function in peripheral immune cells might be altered by smoking and in COPD thereby affecting immune cell responses.We here analysed the expression and activity of proteasome complexes in peripheral blood mononuclear cells (PBMC) isolated from healthy male young smokers as well as from patients with severe COPD and compared them to matching controls. Proteasome expression was upregulated in COPD patients as assessed by RT-qPCR and mass spectrometry-based proteomics analysis. Proteasome activity was quantified using activity-based probes and native gel analysis. We observed distinct activation of immunoproteasomes in the peripheral blood cells of young male smokers and severely ill COPD patients. Native gel analysis and linear regression modeling confirmed robust activation and elevated assembly of 20S proteasomes, which correlated significantly with reduced lung function parameters in COPD patients. The immunoproteasome was distinctly activated in COPD patients upon inflammatory cytokine stimulation of PBMCs in vitro Inhibition of the immunoproteasome reduced proinflammatory cytokine expression in COPD-derived blood immune cells.Given the crucial role of chronic inflammatory signalling and the emerging involvement of autoimmune responses in COPD, therapeutic targeting of the immunoproteasome might represent a novel therapeutic concept for COPD.
Collapse
Affiliation(s)
- Ilona E Kammerl
- Comprehensive Pneumology Center (CPC), University Hospital, Ludwig-Maximilians- University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Sophie Hardy
- Comprehensive Pneumology Center (CPC), University Hospital, Ludwig-Maximilians- University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany.,Cliniques universitaires Saint-Luc, department of pulmonology, and Institute of Experimental and Clinical Research (IREC), Pole of pulmonology, ENT and dermatology, Université catholique de Louvain, Brussels, Belgium
| | - Claudia Flexeder
- Institute of Epidemiology, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Neuherberg, Germany
| | - Andrea Urmann
- Comprehensive Pneumology Center (CPC), University Hospital, Ludwig-Maximilians- University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Julia Peierl
- Comprehensive Pneumology Center (CPC), University Hospital, Ludwig-Maximilians- University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Yuqin Wang
- Comprehensive Pneumology Center (CPC), University Hospital, Ludwig-Maximilians- University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Oliver Vosyka
- Comprehensive Pneumology Center (CPC), University Hospital, Ludwig-Maximilians- University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Marion Frankenberger
- Comprehensive Pneumology Center (CPC), University Hospital, Ludwig-Maximilians- University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany.,Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum Muenchen, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Katrin Milger
- Comprehensive Pneumology Center (CPC), University Hospital, Ludwig-Maximilians- University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany.,Department of Medicine V, University Hospital, LMU, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Jürgen Behr
- Comprehensive Pneumology Center (CPC), University Hospital, Ludwig-Maximilians- University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany.,Department of Medicine V, University Hospital, LMU, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Andrea Koch
- Comprehensive Pneumology Center (CPC), University Hospital, Ludwig-Maximilians- University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany.,Dept. of Pneumology, Teaching Hospital Pyhrn-Eisenwurzen Klinikum Steyr, Austria
| | - Juliane Merl-Pham
- Research Unit Protein Science, Metabolomics and Proteomics Core, Helmholtz Zentrum München, Munich, Germany
| | - Stefanie M Hauck
- Research Unit Protein Science, Metabolomics and Proteomics Core, Helmholtz Zentrum München, Munich, Germany
| | - Charles Pilette
- Cliniques universitaires Saint-Luc, department of pulmonology, and Institute of Experimental and Clinical Research (IREC), Pole of pulmonology, ENT and dermatology, Université catholique de Louvain, Brussels, Belgium
| | - Holger Schulz
- Institute of Epidemiology, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Neuherberg, Germany
| | - Silke Meiners
- Comprehensive Pneumology Center (CPC), University Hospital, Ludwig-Maximilians- University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| |
Collapse
|
183
|
Li T, Zhou HP, Zhou ZJ, Guo LQ, Zhou L. Computed tomography-identified phenotypes of small airway obstructions in chronic obstructive pulmonary disease. Chin Med J (Engl) 2021; 134:2025-2036. [PMID: 34517376 PMCID: PMC8440009 DOI: 10.1097/cm9.0000000000001724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Indexed: 12/02/2022] Open
Abstract
ABSTRACT Chronic obstructive pulmonary disease (COPD) is a heterogeneous disease characteristic of small airway inflammation, obstruction, and emphysema. It is well known that spirometry alone cannot differentiate each separate component. Computed tomography (CT) is widely used to determine the extent of emphysema and small airway involvement in COPD. Compared with the pulmonary function test, small airway CT phenotypes can accurately reflect disease severity in patients with COPD, which is conducive to improving the prognosis of this disease. CT measurement of central airway morphology has been applied in clinical, epidemiologic, and genetic investigations as an inference of the presence and severity of small airway disease. This review will focus on presenting the current knowledge and methodologies in chest CT that aid in identifying discrete COPD phenotypes.
Collapse
Affiliation(s)
- Tao Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, China
- Department of Respiratory Medicine, Xuzhou First People's Hospital, Xuzhou, Jiangsu 221116, China
| | - Hao-Peng Zhou
- Department of Medicine, Jiangsu University School of Medicine, Zhenjiang, Jiangsu 212013, China
| | - Zhi-Jun Zhou
- Institute of Radio Frequency & Optical Electronics-Integrated Circuits, School of Information and Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Li-Quan Guo
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, Jiangsu 215163, China
| | - Linfu Zhou
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, China
- Institute of Integrative Medicine, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| |
Collapse
|
184
|
Cazzola M, Ora J, Cavalli F, Rogliani P, Matera MG. An Overview of the Safety and Efficacy of Monoclonal Antibodies for the Chronic Obstructive Pulmonary Disease. Biologics 2021; 15:363-374. [PMID: 34475751 PMCID: PMC8407524 DOI: 10.2147/btt.s295409] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 08/19/2021] [Indexed: 11/23/2022]
Abstract
Several mAbs have been tested or are currently under clinical evaluation for the treatment of COPD. They can be subdivided into those that aim to block specific pro-inflammatory and pro-neutrophilic cytokines and chemokines, such as TNF-α, IL-1β, CXCL8 and IL-1β, and those that act on T2-mediated inflammation, respectively, by blocking IL-5 and/or its receptor, preventing IL-4 and IL-13 signaling, affecting IL-33 pathway and blocking TSLP. None of these approaches has proved to be effective, probably because in COPD there is no dominant cytokine or chemokine and, therefore, a single mAb cannot be effective on all pathways. With a more in-depth understanding of the numerous pheno/endotypic pathways that play a role in COPD, it may eventually be possible to identify those specific patients in whom some of these cytokines or chemokines might predominate. In this case, it will be possible to implement a personalized treatment, but the use of each mAb will only be reserved for a very limited number of subjects.
Collapse
Affiliation(s)
- Mario Cazzola
- Chair of Respiratory Medicine, Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Josuel Ora
- Division of Respiratory Medicine, University Hospital Tor Vergata, Rome, Italy
| | - Francesco Cavalli
- Chair of Respiratory Medicine, Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Paola Rogliani
- Chair of Respiratory Medicine, Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy.,Division of Respiratory Medicine, University Hospital Tor Vergata, Rome, Italy
| | - Maria Gabriella Matera
- Chair of Pharmacology, Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| |
Collapse
|
185
|
Ancel J, Belgacemi R, Diabasana Z, Perotin JM, Bonnomet A, Dewolf M, Launois C, Mulette P, Deslée G, Polette M, Dormoy V. Impaired Ciliary Beat Frequency and Ciliogenesis Alteration during Airway Epithelial Cell Differentiation in COPD. Diagnostics (Basel) 2021; 11:diagnostics11091579. [PMID: 34573921 PMCID: PMC8469815 DOI: 10.3390/diagnostics11091579] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 08/25/2021] [Accepted: 08/27/2021] [Indexed: 12/19/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a frequent respiratory disease. However, its pathophysiology remains partially elucidated. Epithelial remodeling including alteration of the cilium is a major hallmark of COPD, but specific assessments of the cilium have been rarely investigated as a diagnostic tool in COPD. Here we explore the dysregulation of the ciliary function (ciliary beat frequency (CBF)) and differentiation (multiciliated cells formation in air-liquid interface cultures) of bronchial epithelial cells from COPD (n = 17) and non-COPD patients (n = 15). CBF was decreased by 30% in COPD (11.15 +/- 3.37 Hz vs. 7.89 +/- 3.39 Hz, p = 0.037). Ciliary differentiation was altered during airway epithelial cell differentiation from COPD patients. While the number of multiciliated cells decreased (p < 0.005), the number of primary ciliated cells increased (p < 0.05) and primary cilia were shorter (p < 0.05). Altogether, we demonstrate that COPD can be considered as a ciliopathy through both primary non-motile cilia modifications (related to airway epithelial cell repair and remodeling) and motile cilia function impairment (associated with decrease sputum clearance and clinical respiratory symptoms). These observations encourage considering cilia-associated features in the complex COPD physiopathology and highlight the potential of cilia-derived biomarkers for diagnosis.
Collapse
Affiliation(s)
- Julien Ancel
- Inserm UMR-S1250, P3Cell, Université de Reims Champagne Ardenne, SFR CAP-SANTE, 51092 Reims, France; (J.A.); (R.B.); (Z.D.); (J.-M.P.); (A.B.); (P.M.); (G.D.); (M.P.)
- Department of Respiratory Diseases, Centre Hospitalier Universitaire de Reims, Hôpital Maison Blanche, 51092 Reims, France; (M.D.); (C.L.)
| | - Randa Belgacemi
- Inserm UMR-S1250, P3Cell, Université de Reims Champagne Ardenne, SFR CAP-SANTE, 51092 Reims, France; (J.A.); (R.B.); (Z.D.); (J.-M.P.); (A.B.); (P.M.); (G.D.); (M.P.)
| | - Zania Diabasana
- Inserm UMR-S1250, P3Cell, Université de Reims Champagne Ardenne, SFR CAP-SANTE, 51092 Reims, France; (J.A.); (R.B.); (Z.D.); (J.-M.P.); (A.B.); (P.M.); (G.D.); (M.P.)
| | - Jeanne-Marie Perotin
- Inserm UMR-S1250, P3Cell, Université de Reims Champagne Ardenne, SFR CAP-SANTE, 51092 Reims, France; (J.A.); (R.B.); (Z.D.); (J.-M.P.); (A.B.); (P.M.); (G.D.); (M.P.)
- Department of Respiratory Diseases, Centre Hospitalier Universitaire de Reims, Hôpital Maison Blanche, 51092 Reims, France; (M.D.); (C.L.)
| | - Arnaud Bonnomet
- Inserm UMR-S1250, P3Cell, Université de Reims Champagne Ardenne, SFR CAP-SANTE, 51092 Reims, France; (J.A.); (R.B.); (Z.D.); (J.-M.P.); (A.B.); (P.M.); (G.D.); (M.P.)
- Platform of Cellular and Tissular Imaging (PICT), Université de Reims Champagne Ardenne, 51097 Reims, France
| | - Maxime Dewolf
- Department of Respiratory Diseases, Centre Hospitalier Universitaire de Reims, Hôpital Maison Blanche, 51092 Reims, France; (M.D.); (C.L.)
| | - Claire Launois
- Department of Respiratory Diseases, Centre Hospitalier Universitaire de Reims, Hôpital Maison Blanche, 51092 Reims, France; (M.D.); (C.L.)
| | - Pauline Mulette
- Inserm UMR-S1250, P3Cell, Université de Reims Champagne Ardenne, SFR CAP-SANTE, 51092 Reims, France; (J.A.); (R.B.); (Z.D.); (J.-M.P.); (A.B.); (P.M.); (G.D.); (M.P.)
- Department of Respiratory Diseases, Centre Hospitalier Universitaire de Reims, Hôpital Maison Blanche, 51092 Reims, France; (M.D.); (C.L.)
| | - Gaëtan Deslée
- Inserm UMR-S1250, P3Cell, Université de Reims Champagne Ardenne, SFR CAP-SANTE, 51092 Reims, France; (J.A.); (R.B.); (Z.D.); (J.-M.P.); (A.B.); (P.M.); (G.D.); (M.P.)
- Department of Respiratory Diseases, Centre Hospitalier Universitaire de Reims, Hôpital Maison Blanche, 51092 Reims, France; (M.D.); (C.L.)
| | - Myriam Polette
- Inserm UMR-S1250, P3Cell, Université de Reims Champagne Ardenne, SFR CAP-SANTE, 51092 Reims, France; (J.A.); (R.B.); (Z.D.); (J.-M.P.); (A.B.); (P.M.); (G.D.); (M.P.)
- Department of Biopathology, Centre Hospitalier Universitaire de Reims, Hôpital Maison Blanche, 51092 Reims, France
| | - Valérian Dormoy
- Inserm UMR-S1250, P3Cell, Université de Reims Champagne Ardenne, SFR CAP-SANTE, 51092 Reims, France; (J.A.); (R.B.); (Z.D.); (J.-M.P.); (A.B.); (P.M.); (G.D.); (M.P.)
- Correspondence:
| |
Collapse
|
186
|
Li S, Zhao S, Wu Z, Wang F, Li W. Alteration of immune profiles is associated with pulmonary function and symptoms in patients with chronic obstructive pulmonary disease. Mol Med Rep 2021; 24:742. [PMID: 34435653 PMCID: PMC8430332 DOI: 10.3892/mmr.2021.12382] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 06/10/2021] [Indexed: 02/05/2023] Open
Abstract
Inflammation serves a key role in chronic obstructive pulmonary disease (COPD). However, changes in the immune profiles of patients with COPD remain unclear. The present prospective observational study aimed to determine the expression profiles of immune cells and inflammatory factors of patients with COPD and healthy controls, and to analyze the relationship between immune profiles and smoking history. A total of 140 subjects were enrolled in the present study between September 2018 and April 2019 at West China Hospital of Sichuan University (Chengdu, China). These included 87 patients with stable COPD and 24 patients with acute exacerbated COPD, as well as 29 healthy controls. Baseline characteristics were recorded during the screening period, and levels of immune cells were examined using flow cytometry. In addition, levels of inflammatory factors were measured using ELISAs. The results revealed increased expression of the CD64+/CD14+ mononuclear phagocyte system (MPS) and CD16+CD66+ neutrophils, and decreased expression of CD3+CD4+ T cells and CD3+ CD8+ T cells (all P<0.05) in the peripheral blood of patients with COPD and smokers relative to non-smoking controls. In addition, significant differences were observed in protein levels of IL-6, IL-1β, TNF-α, TGF-α, IFN-γ, IL-8, IL-17A and CRP among the three groups (all P<0.05). Furthermore, the IL-17A, TNF and NF-κB signaling pathways were found to serve a key role in the inflammatory network of COPD. Pearson's correlation analysis revealed a positive correlation between CD3+T lymphocyte counts and pulmonary function, and a negative correlation with smoking and exacerbations. Furthermore, neutrophils and MPS were negatively associated with pulmonary function, and IL-8 was positively associated with cough. There was also a negative association between CRP and IL-17A with pulmonary function but a positive correlation with symptoms and exacerbation. Club cell secretory protein was also negatively associated with emphysema parameters. In conclusion, the present findings revealed significant differences in profiles of immune factors among patients with COPD, smokers and non-smoking controls and their association with clinical characteristics. The clinical trial registration number of the present study is: ChiCTR1800015700 (registered with http://www.chictr.org.cn/index.aspx, 2018/04/16).
Collapse
Affiliation(s)
- Sixiang Li
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Shuang Zhao
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Zhenru Wu
- Pathology Research Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Fangfang Wang
- Hematology Research Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Weimin Li
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
187
|
Martin C, Burgel PR, Roche N. Inhaled Dual Phosphodiesterase 3/4 Inhibitors for the Treatment of Patients with COPD: A Short Review. Int J Chron Obstruct Pulmon Dis 2021; 16:2363-2373. [PMID: 34429594 PMCID: PMC8378910 DOI: 10.2147/copd.s226688] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 07/19/2021] [Indexed: 11/23/2022] Open
Abstract
Current pharmacological treatments for chronic obstructive pulmonary disease (COPD) are mostly limited to inhaled bronchodilators and corticosteroids. Azithromycin can contribute to exacerbation prevention. Roflumilast, a phosphodiesterase (PDE) 4 inhibitor administered orally, also prevents exacerbations in selected patients with chronic bronchitis, recurrent exacerbations, severe airflow limitation and concomitant therapy with long-acting inhaled bronchodilators. This outcome likely results from anti-inflammatory effects since PDE4 is expressed by all inflammatory cell types involved in COPD. The use of this agent is, however, limited by side-effects, particularly nausea and diarrhea. To address remaining unmet needs and enrich therapeutic options for patients with COPD, inhaled dual PDE3/4 inhibitors have been developed, with the aim of enhancing bronchodilation through PDE3 inhibition and modulating inflammation and mucus production though PDE4 inhibition, thus producing a potentially synergistic effect on airway calibre. Experimental preclinical data confirmed these effects in vitro and in animal models. At present, RPL554/ensifentrine is the only agent of this family in clinical development. It decreases sputum markers of both neutrophilic and eosinophilic inflammation in patients with COPD. Clinical Phase II trials confirmed its bronchodilator effect and demonstrated clinically meaningful symptom relief and quality of life improvements in these patients. The safety profile appears satisfactory, with less effects on heart rate and blood pressure than salbutamol and no other side effect. Altogether, these data suggest that ensifentrine could have a role in COPD management, especially in addition to inhaled long-acting bronchodilators with or without corticosteroids since experimental studies suggest potentiation of ensifentrine effects by these agents. However, results from ongoing and future Phase III studies are needed to confirm both beneficial effects and favourable safety profile on a larger scale and assess other outcomes including exacerbations, lung function decline, comorbidities and mortality.
Collapse
Affiliation(s)
- Clémence Martin
- AP-HP Centre, Hôpital Cochin, Service de Pneumologie, Paris, France.,Université de Paris, Institut Cochin, INSERM UMR 1016, Paris, France
| | - Pierre-Régis Burgel
- AP-HP Centre, Hôpital Cochin, Service de Pneumologie, Paris, France.,Université de Paris, Institut Cochin, INSERM UMR 1016, Paris, France
| | - Nicolas Roche
- AP-HP Centre, Hôpital Cochin, Service de Pneumologie, Paris, France.,Université de Paris, Institut Cochin, INSERM UMR 1016, Paris, France
| |
Collapse
|
188
|
Waeijen-Smit K, Reynaert NL, Beijers RJHCG, Houben-Wilke S, Simons SO, Spruit MA, Franssen FME. Alterations in plasma hyaluronic acid in patients with clinically stable COPD versus (non)smoking controls. Sci Rep 2021; 11:15883. [PMID: 34354097 PMCID: PMC8342478 DOI: 10.1038/s41598-021-95030-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 07/20/2021] [Indexed: 12/02/2022] Open
Abstract
Hyaluronic acid (HA) is a key component of the extracellular matrix. HA and its metabolism are suggested to be altered in the lungs of patients with chronic obstructive pulmonary disease (COPD). The present study explored systemic HA, and its metabolic regulators, in patients with clinically stable COPD and smoking and non-smoking controls. Furthermore, associations of HA with acute exacerbations (AECOPD), airway-related hospitalizations, systemic inflammation and cardiovascular risk were studied. In total, 192 patients with moderate to very severe COPD [aged 62.3 y (± SD 7.0)], 84 smoking controls [aged 61.8 y (± 5.7)], and 107 non-smoking controls [aged 60.1 y (± 7.0)] were included. Plasma HA was reduced in patients with COPD compared to non-smoking controls (p = 0.033), but was comparable after adjusting for age and sex. Expression of HAS-3 did not differ between groups, but was substantially less detectable in more patients with COPD than (non)smoking controls (p < 0.001). Expression of HYAL-2 was enhanced in patients with COPD versus smoking (p = 0.019) and non-smoking (p < 0.001) controls, also in the age- and sex- adjusted model (p < 0.001). Plasma HA was not associated with AECOPD, airway-related hospitalizations in the previous year, or systemic inflammation in COPD. Arterial pulse wave velocity explained some of the variance (< 10%) in plasma HA (p = 0.006). Overall, these results indicate that expression of HYAL-2, but not plasma HA nor HAS-3, is enhanced in patients with COPD compared to (non)smoking controls. Furthermore, HA was not associated with clinical outcomes, yet, cardiovascular risk might play a role in its systemic regulation in stable COPD.
Collapse
Affiliation(s)
- Kiki Waeijen-Smit
- Department of Research and Education, Ciro, Horn, The Netherlands. .,Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands.
| | - Niki L Reynaert
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Rosanne J H C G Beijers
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | | | - Sami O Simons
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Martijn A Spruit
- Department of Research and Education, Ciro, Horn, The Netherlands.,Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Frits M E Franssen
- Department of Research and Education, Ciro, Horn, The Netherlands.,Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
| |
Collapse
|
189
|
Yi E, Zhang J, Zheng M, Zhang Y, Liang C, Hao B, Hong W, Lin B, Pu J, Lin Z, Huang P, Li B, Zhou Y, Ran P. Long noncoding RNA IL6-AS1 is highly expressed in chronic obstructive pulmonary disease and is associated with interleukin 6 by targeting miR-149-5p and early B-cell factor 1. Clin Transl Med 2021; 11:e479. [PMID: 34323408 PMCID: PMC8288003 DOI: 10.1002/ctm2.479] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 06/09/2021] [Accepted: 06/14/2021] [Indexed: 11/17/2022] Open
Abstract
Chronic obstructive pulmonary disease is a complex condition with multiple etiologies, including inflammation. We identified a novel long noncoding RNA (lncRNA), interleukin 6 antisense RNA 1 (IL6-AS1), which is upregulated in this disease and is associated with airway inflammation. We found that IL6-AS1 promotes the expression of inflammatory factors, especially interleukin (IL) 6. Mechanistically, cytoplasmic IL6-AS1 acts as an endogenous sponge by competitively binding to the microRNA miR-149-5p to stabilize IL-6 mRNA. Nuclear IL6-AS1 promotes IL-6 transcription by recruiting early B-cell factor 1 to the IL-6 promoter, which increases the methylation of the H3K4 histone and acetylation of the H3K27 histone. We propose a model of lncRNA expression in both the nucleus and cytoplasm that exerts similar effects through differing mechanisms, and IL6-AS1 probably increases inflammation via multiple pathways.
Collapse
Affiliation(s)
- Erkang Yi
- National Center for Respiratory MedicineState Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory DiseaseGuangzhou Institute of Respiratory HealthThe First Affiliated Hospital of Guangzhou Medical University151 Yanjiang Xi RoadGuangzhouGuangdong510000China
| | - Jiahuan Zhang
- National Center for Respiratory MedicineState Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory DiseaseGuangzhou Institute of Respiratory HealthThe First Affiliated Hospital of Guangzhou Medical University151 Yanjiang Xi RoadGuangzhouGuangdong510000China
| | - Mengning Zheng
- National Center for Respiratory MedicineState Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory DiseaseGuangzhou Institute of Respiratory HealthThe First Affiliated Hospital of Guangzhou Medical University151 Yanjiang Xi RoadGuangzhouGuangdong510000China
| | - Yi Zhang
- GMU‐GIBH Joint School of Life SciencesGuangzhou Medical UniversityGuangzhouGuangdong510000China
| | - Chunxiao Liang
- National Center for Respiratory MedicineState Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory DiseaseGuangzhou Institute of Respiratory HealthThe First Affiliated Hospital of Guangzhou Medical University151 Yanjiang Xi RoadGuangzhouGuangdong510000China
| | - Binwei Hao
- National Center for Respiratory MedicineState Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory DiseaseGuangzhou Institute of Respiratory HealthThe First Affiliated Hospital of Guangzhou Medical University151 Yanjiang Xi RoadGuangzhouGuangdong510000China
| | - Wei Hong
- GMU‐GIBH Joint School of Life SciencesGuangzhou Medical UniversityGuangzhouGuangdong510000China
| | - Biting Lin
- GMU‐GIBH Joint School of Life SciencesGuangzhou Medical UniversityGuangzhouGuangdong510000China
| | - Jinding Pu
- National Center for Respiratory MedicineState Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory DiseaseGuangzhou Institute of Respiratory HealthThe First Affiliated Hospital of Guangzhou Medical University151 Yanjiang Xi RoadGuangzhouGuangdong510000China
| | - Zhiwei Lin
- GMU‐GIBH Joint School of Life SciencesGuangzhou Medical UniversityGuangzhouGuangdong510000China
| | - Peiyu Huang
- National Center for Respiratory MedicineState Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory DiseaseGuangzhou Institute of Respiratory HealthThe First Affiliated Hospital of Guangzhou Medical University151 Yanjiang Xi RoadGuangzhouGuangdong510000China
| | - Bing Li
- GMU‐GIBH Joint School of Life SciencesGuangzhou Medical UniversityGuangzhouGuangdong510000China
| | - Yumin Zhou
- National Center for Respiratory MedicineState Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory DiseaseGuangzhou Institute of Respiratory HealthThe First Affiliated Hospital of Guangzhou Medical University151 Yanjiang Xi RoadGuangzhouGuangdong510000China
| | - Pixin Ran
- National Center for Respiratory MedicineState Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory DiseaseGuangzhou Institute of Respiratory HealthThe First Affiliated Hospital of Guangzhou Medical University151 Yanjiang Xi RoadGuangzhouGuangdong510000China
| |
Collapse
|
190
|
Feng C, Xu M, Kang J, Wen F, Chen Y, Zhang J, Xiao W, Zhang Z, Yang L, Huo J, Cao J, Zhao L, Xia S, Yin Y, Wang W. Atypical Pathogen Distribution in Chinese Hospitalized AECOPD Patients: A Multicenter Cross-Sectional Study. Int J Chron Obstruct Pulmon Dis 2021; 16:1699-1708. [PMID: 34135581 PMCID: PMC8200156 DOI: 10.2147/copd.s300779] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 05/09/2021] [Indexed: 02/05/2023] Open
Abstract
Purpose The proportion of atypical pathogens in patient with AECOPD within mainland China is unknown. The objectives of this study were to determine the distribution of atypical pathogens among Chinese patients with AECOPD, to evaluate the clinical characteristics of different atypical pathogen infections, and to compare different detection methods for atypical pathogens. Patients and Methods Specimens were collected from patients with AECOPD from March 2016 to November 2018 at eleven medical institutions in eight cities in China. Double serum, sputum, and urine samples were obtained from 145 patients. Serological and nucleic acid tests were used to assess for Mycoplasma pneumonia and Chlamydia pneumoniae; serological, urinary antigen, and nucleic acid tests were applied to detect Legionella pneumophila. The clinical characteristics of atypical pathogen-positive and -negative groups were also compared. Results The overall positivity rate for Mycoplasma pneumoniae was 20.69% (30/145), with the highest rate being 20.00% (29/145) when determined by passive agglutination.The overall positive rates for Chlamydia pneumoniae and Legionella pneumophila were 29.66% (43/145) and 10.34% (15/145), respectively. The most common serotype of Legionella pneumophila was type 6. The maximum hospitalized body temperature, ratio of eosinophils, C-reactive protein (CRP) level, and procalcitonin (PCT) level of the Mycoplasma pneumoniae-positive group were significantly higher than those of the Mycoplasma pneumoniae-negative group. Patients in the Chlamydia pneumoniae-positive group smoked more, had higher proportions of comorbidities and frequent aggravations in the previous two years than those in the Chlamydia pneumoniae-negative group. Furthermore, the forced expiratory volume in one second to forced vital capacity (FEV1/FVC) ratio assessment of lung function was higher, and the concentration of arterial blood bicarbonate (HCO3 -) was lower in the Legionella pneumophila-positive group than in the Legionella pneumophila-negative group. Conclusion Overall, atypical pathogens play an important role in AECOPD. Regarding the testing method, serological testing is a superior method to nucleic acid testing.
Collapse
Affiliation(s)
- Chenye Feng
- Department of Pulmonary and Critical Care Medicine, Institute of Respiratory Diseases, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Mingtao Xu
- Department of Pulmonary and Critical Care Medicine, Institute of Respiratory Diseases, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Jian Kang
- Department of Pulmonary and Critical Care Medicine, Institute of Respiratory Diseases, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Fuqiang Wen
- Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University and Division of Pulmonary Disease, State Key Laboratory of Biotherapy of China, Chengdu, Sichuan, People's Republic of China
| | - Yahong Chen
- Department of Respiratory and Critical Care Medicine, Peking University, Third Hospital, Beijing, People's Republic of China
| | - Jing Zhang
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Wei Xiao
- Department of Pulmonary Medicine, Qilu Hospital, Shandong University, Jinan, Shandong, People's Republic of China
| | - Zhonghe Zhang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaonign, People's Republic of China
| | - Lan Yang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shanxi, People's Republic of China
| | - Jianmin Huo
- Department of Respiratory, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Jie Cao
- Respiratory Department, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Li Zhao
- Department of Pulmonary and Critical Care Medicine, Institute of Respiratory Diseases, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Shuyue Xia
- Department of Pulmonary Medicine, Central Hospital Affiliated with Shenyang Medical College, Shenyang, People's Republic of China
| | - Yan Yin
- Department of Pulmonary and Critical Care Medicine, Institute of Respiratory Diseases, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Wei Wang
- Department of Pulmonary and Critical Care Medicine, Institute of Respiratory Diseases, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| |
Collapse
|
191
|
Is it time to look beyond bronchodilators and corticosteroids in treating COPD? FUTURE DRUG DISCOVERY 2021. [DOI: 10.4155/fdd-2021-0001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
192
|
Tan WC, Bourbeau J, Nadeau G, Wang W, Barnes N, Landis SH, Kirby M, Hogg JC, Sin DD. High eosinophil counts predict decline in FEV 1: results from the CanCOLD study. Eur Respir J 2021; 57:13993003.00838-2020. [PMID: 33303555 DOI: 10.1183/13993003.00838-2020] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 11/05/2020] [Indexed: 12/13/2022]
Abstract
INTRODUCTION The aim of this study was to examine the association between blood eosinophil levels and the decline in lung function in individuals aged >40 years from the general population. METHODS The study evaluated the eosinophil counts from thawed blood in 1120 participants (mean age 65 years) from the prospective population-based Canadian Cohort of Obstructive Lung Disease (CanCOLD) study. Participants answered interviewer-administered respiratory questionnaires and performed pre-/post-bronchodilator spirometric tests at 18-month intervals; computed tomography (CT) imaging was performed at baseline. Statistical analyses to describe the relationship between eosinophil levels and decline in forced expiratory volume in 1 s (FEV1) were performed using random mixed-effects regression models with adjustments for demographics, smoking, baseline FEV1, ever-asthma and history of exacerbations in the previous 12 months. CT measurements were compared between eosinophil subgroups using ANOVA. RESULTS Participants who had a peripheral eosinophil count of ≥300 cells·µL-1 (n=273) had a greater decline in FEV1 compared with those with eosinophil counts of <150 cells·µL-1 (n=430; p=0.003) (reference group) and 150-<300 cells·µL-1 (n=417; p=0.003). The absolute change in FEV1 was -32.99 mL·year-1 for participants with eosinophil counts <150 cells·µL-1; -38.78 mL·year-1 for those with 150-<300 cells·µL-1 and -67.30 mL·year-1 for participants with ≥300 cells·µL-1. In COPD, higher eosinophil count was associated with quantitative CT measurements reflecting both small and large airway abnormalities. CONCLUSION A blood eosinophil count of ≥300 cells·µL-1 is an independent risk factor for accelerated lung function decline in older adults and is related to undetected structural airway abnormalities.
Collapse
Affiliation(s)
- Wan C Tan
- Centre for Heart Lung Innovation, St Paul's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Jean Bourbeau
- Respiratory Epidemiology and Clinical Research Unit, Research Institute of the McGill University Health Centre, McGill University, Montréal, QC, Canada
| | - Gilbert Nadeau
- Respiratory Medical Affairs, GlaxoSmithKline, Mississauga, ON, Canada
| | - Wendy Wang
- Centre for Heart Lung Innovation, St Paul's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Neil Barnes
- William Harvey Institute, Bart's and the London School of Medicine and Dentistry, and Respiratory Therapy Area, GlaxoSmithKline plc, London, UK
| | - Sarah H Landis
- Real World Evidence and Epidemiology, GlaxoSmithKline plc, Uxbridge, UK
| | - Miranda Kirby
- Centre for Heart Lung Innovation, St Paul's Hospital, University of British Columbia, Vancouver, BC, Canada.,Dept of Physics, Ryerson University, Toronto, ON, Canada
| | - James C Hogg
- Centre for Heart Lung Innovation, St Paul's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Don D Sin
- Centre for Heart Lung Innovation, St Paul's Hospital, University of British Columbia, Vancouver, BC, Canada.,A list of members of the COLD/CanCOLD Collaborative Research Group can be found in the acknowledgements section
| | | | | |
Collapse
|
193
|
The effect of nitrogen dioxide and atmospheric pressure on hospitalization risk for chronic obstructive pulmonary disease in Guangzhou, China. Respir Med 2021; 182:106424. [PMID: 33932714 DOI: 10.1016/j.rmed.2021.106424] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 04/13/2021] [Accepted: 04/16/2021] [Indexed: 01/01/2023]
Abstract
BACKGROUND The relationship between air pollution and meteorological factors on diseases has become a research hotspot recently. Nevertheless, few studies have touched the inferences of nitrogen dioxide (NO2) and atmospheric pressure (AP) on hospitalization risk for chronic obstructive pulmonary disease (COPD). OBJECTIVES To investigate the short-term impact of particulate air pollutants and meteorology factors on hospitalizations for COPD and quantify the corresponding risk burden of hospital admission. METHODS In our study, COPD cases were collected from Guangzhou Panyu Central Hospital (n = 11,979) from Dec of 2013 to Jun 2019. The 24-h average temperature, relative humidity (RH), wind speed (V), AP and other meteorological data were obtained from Guangzhou Meteorological Bureau. Air pollution data were collected from Guangzhou Air Monitoring Station. The influence of different NO2 and AP values on COPD risk was quantified by a distributed lag nonlinear model (DLNM) combined with Poisson Regression and Time Series analysis. RESULTS We found that NO2 had a non-linear relationship with the incidence of COPD, with an approximate "M" type, appearing at the peaks of 126 μg/m³ (RR = 1.32, 95%CI, 1.07 to 1.64) and 168 μg/m³ (RR = 1.21, 95%CI, 0.94 to 1.55), respectively. And the association between AP and COPD incidence exhibited an approximate J-shape with a peak occurring at 1035 hPa (RR = 1.16, 95% CI, 1.02 to 1.31). CONCLUSIONS The nonlinear relationship of NO2 and AP on COPD admission risk in different periods of lag can be used to establish an early warning system for diseases and reduce the possible outbreaks and burdens of COPD in a sensitive population.
Collapse
|
194
|
Waeijen-Smit K, Houben-Wilke S, DiGiandomenico A, Gehrmann U, Franssen FME. Unmet needs in the management of exacerbations of chronic obstructive pulmonary disease. Intern Emerg Med 2021; 16:559-569. [PMID: 33616876 PMCID: PMC7897880 DOI: 10.1007/s11739-020-02612-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 12/17/2020] [Indexed: 12/11/2022]
Abstract
Exacerbations of chronic obstructive pulmonary disease (COPD) are episodes of acute worsening of respiratory symptoms that require additional therapy. These events play a pivotal role in the natural course of the disease and are associated with a progressive decline in lung function, reduced health status, a low physical activity level, tremendous health care costs, and increased mortality. Although most exacerbations have an infectious origin, the underlying mechanisms are heterogeneous and specific predictors of their occurrence in individual patients are currently unknown. Accurate prediction and early diagnosis of exacerbations is essential to develop novel targets for prevention and personalized treatments to reduce the impact of these events. Several potential biomarkers have previously been studied, these however lack specificity, accuracy and do not add value to the available clinical predictors. At present, microbial composition and host-microbiome interactions in the lung are increasingly recognized for their role in affecting the susceptibility to exacerbations, and may steer towards a novel direction in the management of COPD exacerbations. This narrative review describes the current challenges and unmet needs in the management of acute exacerbations of COPD. Exacerbation triggers, biological clusters, current treatment strategies, and their limitations, previously studied biomarkers and prediction tools, the lung microbiome and its role in COPD exacerbations as well as future directions are discussed.
Collapse
Affiliation(s)
- Kiki Waeijen-Smit
- Department of Research and Education, Ciro, Horn, NM, 6085, The Netherlands.
- NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht, The Netherlands.
- Department of Respiratory Medicine, Maastricht University Medical Centre (MUMC+), Maastricht, The Netherlands.
| | - Sarah Houben-Wilke
- Department of Research and Education, Ciro, Horn, NM, 6085, The Netherlands
| | - Antonio DiGiandomenico
- Discovery Microbiome, Microbial Sciences, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, USA
| | - Ulf Gehrmann
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory and Immunology (R&I), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Frits M E Franssen
- Department of Research and Education, Ciro, Horn, NM, 6085, The Netherlands
- NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht, The Netherlands
- Department of Respiratory Medicine, Maastricht University Medical Centre (MUMC+), Maastricht, The Netherlands
| |
Collapse
|
195
|
Peng J, Yu Q, Fan S, Chen X, Tang R, Wang D, Qi D. High Blood Eosinophil and YKL-40 Levels, as Well as Low CXCL9 Levels, are Associated with Increased Readmission in Patients with Acute Exacerbation of Chronic Obstructive Pulmonary Disease. Int J Chron Obstruct Pulmon Dis 2021; 16:795-806. [PMID: 33814903 PMCID: PMC8009765 DOI: 10.2147/copd.s294968] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 03/04/2021] [Indexed: 12/14/2022] Open
Abstract
Background Readmission after hospital discharge is common among patients with acute exacerbation of chronic obstructive pulmonary disease (AECOPD). Predictive biomarkers of readmission would facilitate stratification strategies and individualized prognosis. Therefore, this study aimed to investigate the utility of type 2 biomarkers (eosinophils, periostin, and YKL-40) and a type 1 biomarker (CXCL9) in predicting readmission events in patients with AECOPD. Methods This is a prospective observational study design. Blood levels of eosinophils, periostin, YKL-40, and CXCL9 were measured at admission. The clinical outcomes were 12-month COPD-related readmission, time to COPD-related readmission, and number of 12-month COPD-related readmissions. These outcomes were analyzed using logistic and Cox regression models and Spearman's rank test. Results A total of 123 patients were included, of whom 51 had experienced at least one readmission for AECOPD. High levels of eosinophils (≥200 cells/μL or 2% of the total white blood cell count, adjusted odds ratio [aOR] =3.138, P=0.009) and YKL-40 (≥14.5 ng/mL, aOR =2.840, P=0.015), as well as low CXCL9 levels (≤30.1 ng/mL, aOR =2.551, P=0.028), were associated with an increased COPD-related readmission. The highest relative readmission rate was observed in patients with both high eosinophil and YKL-40 levels. Moreover, high eosinophil and YKL-40 levels were associated with a shorter time to first COPD-related readmission and an increased number of 12-month COPD-related readmissions. Conclusion High blood eosinophil and YKL-40 levels, as well as low CXCL9 levels, have predictive utility for the 12-month COPD-related readmission rate. Using eosinophils and YKL-40 together allows more precise identification of patients at high risk of COPD-related readmission.
Collapse
Affiliation(s)
- Junnan Peng
- Department of Respiratory Medicine, Second Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Qian Yu
- Department of Respiratory Medicine, Second Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Shulei Fan
- Department of Respiratory Medicine, Second Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Xingru Chen
- Department of Respiratory Medicine, Second Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Rui Tang
- Department of Respiratory Medicine, Second Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Daoxin Wang
- Department of Respiratory Medicine, Second Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Di Qi
- Department of Respiratory Medicine, Second Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| |
Collapse
|
196
|
Antunes MA, Lopes-Pacheco M, Rocco PRM. Oxidative Stress-Derived Mitochondrial Dysfunction in Chronic Obstructive Pulmonary Disease: A Concise Review. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6644002. [PMID: 37448755 PMCID: PMC10337713 DOI: 10.1155/2021/6644002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 01/27/2021] [Accepted: 02/26/2021] [Indexed: 08/02/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is a progressive and disabling disorder marked by airflow limitation and extensive destruction of lung parenchyma. Cigarette smoke is the major risk factor for COPD development and has been associated with increased oxidant burden on multiple cell types in the lungs. Elevated levels of reactive oxygen species (ROS) may significantly affect expression of biological molecules, signaling pathways, and function of antioxidant defenses. Although inflammatory cells, such as neutrophils and macrophages, contribute to the release of large quantities of ROS, mitochondrial dysfunction plays a critical role in ROS production due to oxidative phosphorylation. Although mitochondria are dynamic organelles, excess oxidative stress is able to alter mitochondrial function, morphology, and RNA and protein content. Indeed, mitochondria may change their shape by undergoing fusion (regulated by mitofusin 1, mitofusin 2, and optic atrophy 1 proteins) and fission (regulated by dynamin-related protein 1), which are essential processes to maintain a healthy and functional mitochondrial network. Cigarette smoke can induce mitochondrial hyperfusion, thus reducing mitochondrial quality control and cellular stress resistance. Furthermore, diminished levels of enzymes involved in the mitophagy process, such as Parkin (a ubiquitin ligase E3) and the PTEN-induced putative kinase 1 (PINK1), are commonly observed in COPD and correlate directly with faulty removal of dysfunctional mitochondria and consequent cell senescence in this disorder. In this review, we highlight the main mechanisms for the regulation of mitochondrial quality and how they are affected by oxidative stress during COPD development and progression.
Collapse
Affiliation(s)
- Mariana A. Antunes
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
- National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
| | - Miquéias Lopes-Pacheco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Patricia R. M. Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
- National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
- Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSAÚDE/FAPERJ, Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
197
|
Xu X, Hu J, Lyu X, Huang H, Cheng X. Exploring the Interdisciplinary Nature of Precision Medicine:Network Analysis and Visualization. JMIR Med Inform 2021; 9:e23562. [PMID: 33427681 PMCID: PMC7834937 DOI: 10.2196/23562] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 11/02/2020] [Accepted: 12/09/2020] [Indexed: 12/26/2022] Open
Abstract
Background Interdisciplinary research is an important feature of precision medicine. However, the accurate cross-disciplinary status of precision medicine is still unclear. Objective The aim of this study is to present the nature of interdisciplinary collaboration in precision medicine based on co-occurrences and social network analysis. Methods A total of 7544 studies about precision medicine, published between 2010 and 2019, were collected from the Web of Science database. We analyzed interdisciplinarity with descriptive statistics, co-occurrence analysis, and social network analysis. An evolutionary graph and strategic diagram were created to clarify the development of streams and trends in disciplinary communities. Results The results indicate that 105 disciplines are involved in precision medicine research and cover a wide range. However, the disciplinary distribution is unbalanced. Current cross-disciplinary collaboration in precision medicine mainly focuses on clinical application and technology-associated disciplines. The characteristics of the disciplinary collaboration network are as follows: (1) disciplinary cooperation in precision medicine is not mature or centralized; (2) the leading disciplines are absent; (3) the pattern of disciplinary cooperation is mostly indirect rather than direct. There are 7 interdisciplinary communities in the precision medicine collaboration network; however, their positions in the network differ. Community 4, with disciplines such as genetics and heredity in the core position, is the most central and cooperative discipline in the interdisciplinary network. This indicates that Community 4 represents a relatively mature direction in interdisciplinary cooperation in precision medicine. Finally, according to the evolution graph, we clearly present the development streams of disciplinary collaborations in precision medicine. We describe the scale and the time frame for development trends and distributions in detail. Importantly, we use evolution graphs to accurately estimate the developmental trend of precision medicine, such as biological big data processing, molecular imaging, and widespread clinical applications. Conclusions This study can help researchers, clinicians, and policymakers comprehensively understand the overall network of interdisciplinary cooperation in precision medicine. More importantly, we quantitatively and precisely present the history of interdisciplinary cooperation and accurately predict the developing trends of interdisciplinary cooperation in precision medicine.
Collapse
Affiliation(s)
- Xin Xu
- General Medicine Ward, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jiming Hu
- School of Information Management, Wuhan University, Wuhan, China
| | - Xiaoguang Lyu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
| | - He Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xingyu Cheng
- Department of Radiology, Ezhou Central Hospital, Ezhou, China
| |
Collapse
|
198
|
Sunadome H, Sato S, Matsumoto H, Murase K, Kawaguchi T, Tabara Y, Chin K, Matsuda F, Hirai T. Similar distribution of peripheral blood eosinophil counts in European and East Asian populations from investigations of large-scale general population studies: the Nagahama Study. Eur Respir J 2021; 57:57/1/2004101. [PMID: 33402375 DOI: 10.1183/13993003.04101-2020] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 11/09/2020] [Indexed: 11/05/2022]
Affiliation(s)
- Hironobu Sunadome
- Dept of Respiratory Care and Sleep Control Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Susumu Sato
- Dept of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hisako Matsumoto
- Dept of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kimihiko Murase
- Dept of Respiratory Care and Sleep Control Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takahisa Kawaguchi
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yasuharu Tabara
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kazuo Chin
- Dept of Respiratory Care and Sleep Control Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Fumihiko Matsuda
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Toyohiro Hirai
- Dept of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
199
|
Mathioudakis AG, Vanfleteren LEGW, Lahousse L, Higham A, Allinson JP, Gotera C, Visca D, Singh D, Spanevello A. Current developments and future directions in COPD. Eur Respir Rev 2020; 29:29/158/200289. [PMID: 33268439 PMCID: PMC9488623 DOI: 10.1183/16000617.0289-2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 10/27/2020] [Indexed: 11/28/2022] Open
Abstract
The European Respiratory Society journals publish respiratory research and policy documents of the highest quality, offering a platform for the exchange and promotion of scientific knowledge. In this article, focusing on COPD, the third leading cause of death globally, we summarise novel research highlights focusing on the disease's underlying mechanisms, epidemiology and management, with the aim to inform and inspire respiratory clinicians and researchers. Current developments and future directions in COPD: a critical summary of some of the most recent ground-breaking research studies and policy documents from @ERSpublicationshttps://bit.ly/3oW0xDM
Collapse
Affiliation(s)
- Alexander G Mathioudakis
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, The University of Manchester, Manchester, UK .,North West Lung Centre, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Lowie E G W Vanfleteren
- COPD Center, Institute of Medicine, Sahlgrenska University Hospital, University of Gothenburg, Gothenburg, Sweden
| | - Lies Lahousse
- Dept of Bioanalysis, Pharmaceutical Care Unit, Ghent University, Ghent, Belgium
| | - Andrew Higham
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, The University of Manchester, Manchester, UK.,North West Lung Centre, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - James P Allinson
- The Royal Brompton Hospital and The National Heart and Lung Institute, Imperial College London, London, UK
| | - Carolina Gotera
- Dept of Pneumology, IIS-Fundación Jiménez Díaz, ISCIII-CIBERES, Madrid, Spain
| | - Dina Visca
- Division of Pulmonary Rehabilitation, Istituti Clinici Scientifici Maugeri, IRCCS, Tradate, Italy.,Dept of Medicine and Surgery, Respiratory Diseases, University of Insubria, Varese-Como, Italy
| | - Dave Singh
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, The University of Manchester, Manchester, UK.,North West Lung Centre, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK.,Medicines Evaluation Unit, Manchester, UK
| | - Antonio Spanevello
- Division of Pulmonary Rehabilitation, Istituti Clinici Scientifici Maugeri, IRCCS, Tradate, Italy.,Dept of Medicine and Surgery, Respiratory Diseases, University of Insubria, Varese-Como, Italy
| |
Collapse
|
200
|
Lonergan M, Dicker AJ, Crichton ML, Keir HR, Van Dyke MK, Mullerova H, Miller BE, Tal-Singer R, Chalmers JD. Blood neutrophil counts are associated with exacerbation frequency and mortality in COPD. Respir Res 2020; 21:166. [PMID: 32611352 PMCID: PMC7329438 DOI: 10.1186/s12931-020-01436-7] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 06/25/2020] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Identifying patients with COPD at increased risk of poor outcomes is challenging due to disease heterogeneity. Potential biomarkers need to be readily available in real-life clinical practice. Blood eosinophil counts are widely studied but few studies have examined the prognostic value of blood neutrophil counts (BNC). METHODS In a large population-based COPD registry in the East of Scotland (TARDIS: Tayside Allergic and Respiratory Disease Information System), BNC were compared to measures of disease severity and mortality for up to 15 years follow-up. Potential mechanisms of disease modification by BNC were explored in a nested microbiome substudy. RESULTS 178,120 neutrophil counts were obtained from 7220 people (mean follow up 9 years) during stable disease periods. Median BNC was 5200cells/μL (IQR 4000-7000cells/μL). Mortality rates among the 34% of patients with elevated BNCs (defined as 6000-15000cells/μL) at the study start were 80% higher (14.0/100 person years v 7.8/100py, P < 0.001) than those with BNC in the normal range (2000-6000cells/μL). People with elevated BNC were more likely to be classified as GOLD D (46% v 33% P < 0.001), have more exacerbations (mean 2.3 v 1.3/year, P < 0.001), and were more likely to have severe exacerbations (13% vs. 5%, P < 0.001) in the following year. Eosinophil counts were much less predictive of these outcomes. In a sub-cohort (N = 276), patients with elevated BNC had increased relative abundance of Proteobacteria and reduced microbiome diversity. CONCLUSION High BNC may provide a useful indicator of risk of exacerbations and mortality in COPD patients.
Collapse
Affiliation(s)
- Mike Lonergan
- Scottish Centre for Respiratory Research, University of Dundee, Ninewells Hospital and Medical School, Dundee, DD1 9SY, UK
| | - Alison J Dicker
- Scottish Centre for Respiratory Research, University of Dundee, Ninewells Hospital and Medical School, Dundee, DD1 9SY, UK
| | - Megan L Crichton
- Scottish Centre for Respiratory Research, University of Dundee, Ninewells Hospital and Medical School, Dundee, DD1 9SY, UK
| | - Holly R Keir
- Scottish Centre for Respiratory Research, University of Dundee, Ninewells Hospital and Medical School, Dundee, DD1 9SY, UK
| | - Melissa K Van Dyke
- Epidemiology, Value Evidence and Outcomes, GSK R&D, Collegeville, PA, USA
| | - Hana Mullerova
- Epidemiology, Value Evidence and Outcomes, GSK R&D, Uxbridge, UK
| | - Bruce E Miller
- Medical Innovation, Value Evidence and Outcomes, GSK R&D, Collegeville, PA, USA
| | - Ruth Tal-Singer
- Medical Innovation, Value Evidence and Outcomes, GSK R&D, Collegeville, PA, USA
| | - James D Chalmers
- Scottish Centre for Respiratory Research, University of Dundee, Ninewells Hospital and Medical School, Dundee, DD1 9SY, UK.
| |
Collapse
|