1
|
Marshall CL, Mostafa D, Hemshehkar M, Lao Y, Balshaw R, Spicer V, Mookherjee N. Biological Sex Is an Effect Modifier of Allergen-Mediated Alteration of the Lung Proteome. J Proteome Res 2024; 23:4203-4215. [PMID: 39214566 DOI: 10.1021/acs.jproteome.4c00025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Asthma exhibits a distinct sex bias in the disease prevalence, severity, and response to therapy. However, sex-related differences in alterations of the lung proteome mediated by aeroallergens critical in asthma, such as house dust mites (HDM), remain unknown. In this study, we define sex-related differences in the lung proteome using an HDM-challenged mouse model by 1D LC-MS/MS. Sex-disaggregated data analysis showed that 406 proteins were uniquely altered in females, 273 proteins were uniquely altered in males, and 414 proteins were altered in both females and males in response to HDM. In a linear mixed model analysis, sex modified the HDM exposure effect for 163 proteins, i.e., a significant sex:exposure interaction was identified in 84 proteins in females and 35 proteins in males. Of these, 12 proteins showed a significant sex effect in both female and male lungs. We further selected 3 proteins Tjp1, Lamtor1, and G3BP2 for independent confirmation studies. Our findings detail the sex-specific lung proteome in response to an aeroallergen critical in asthma and demonstrate that sex is a significant effect modifier of HDM response. These results will serve as a valuable resource for delineating sex-specific mechanisms in aeroallergen-driven responses in asthma research.
Collapse
Affiliation(s)
- Courtney Lynn Marshall
- Department of Immunology, Rady Faculty of Health Sciences, University of Manitoba, Mercedes-Benz, Winnipeg R3E0T5, Canada
- Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, Rady Faculty of Health Sciences, University of Manitoba, Mercedes-Benz, Winnipeg R3E3P4, Canada
| | - Dina Mostafa
- Department of Immunology, Rady Faculty of Health Sciences, University of Manitoba, Mercedes-Benz, Winnipeg R3E0T5, Canada
- Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, Rady Faculty of Health Sciences, University of Manitoba, Mercedes-Benz, Winnipeg R3E3P4, Canada
| | - Mahadevappa Hemshehkar
- Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, Rady Faculty of Health Sciences, University of Manitoba, Mercedes-Benz, Winnipeg R3E3P4, Canada
| | - Ying Lao
- Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, Rady Faculty of Health Sciences, University of Manitoba, Mercedes-Benz, Winnipeg R3E3P4, Canada
| | - Robert Balshaw
- George and Fay Yee Centre for Healthcare Innovation, University of Manitoba, Mercedes-Benz, Winnipeg R3E0T6, Canada
| | - Victor Spicer
- Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, Rady Faculty of Health Sciences, University of Manitoba, Mercedes-Benz, Winnipeg R3E3P4, Canada
| | - Neeloffer Mookherjee
- Department of Immunology, Rady Faculty of Health Sciences, University of Manitoba, Mercedes-Benz, Winnipeg R3E0T5, Canada
- Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, Rady Faculty of Health Sciences, University of Manitoba, Mercedes-Benz, Winnipeg R3E3P4, Canada
| |
Collapse
|
2
|
Menson KE, Coleman SRM. Reprint of: Smoking and pulmonary health in women: A narrative review and behavioral health perspective. Prev Med 2024:108113. [PMID: 39198081 DOI: 10.1016/j.ypmed.2024.108113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/01/2024]
Abstract
OBJECTIVE Cigarette smoking prevalence has declined slower among women than men, and smoking-related pulmonary disease (PD) has risen among women. Given these trends, there is a critical need to understand and mitigate PD risk among women who smoke. The purpose of this narrative review and commentary is to highlight important evidence from the literature on smoking and PD among women. METHODS This review focuses broadly on examining cigarette smoking and PD among women within six topic areas: (1) demographic characteristics and prevalence of smoking, (2) smoking behavior, (3) lung cancer, (4) obstructive PD, (5) diagnostic and treatment disparities, and (6) gaps in the literature and potential directions for future research and treatment. RESULTS Growing evidence indicates that compared to men, women are at increased risk for developing smoking-related PD and poorer PD outcomes. Gender disparities in smoking-related PD may be largely accounted for by genetic differences and sex hormones contributing to PD pathogenesis and presentation, smoking behavior, nicotine dependence, and pathogen/carcinogen clearance. Moreover, gender disparities in smoking-related PD may be exacerbated by important social determinants (e.g., women with less formal education and those from minoritized groups may be at especially high risk for poor PD outcomes due to higher rates of smoking). CONCLUSION Rising rates of smoking-related PD among women risk widening diagnostic and treatment disparities. Ongoing research is needed to explore potentially complex relationships between sex, gender, and smoking-related PD processes and outcomes, and to improve smoking-cessation and PD treatment for women.
Collapse
Affiliation(s)
- Katherine E Menson
- Division of Pulmonary and Critical Care Medicine, University of Vermont, Burlington, VT, USA; Larner College of Medicine, University of Vermont, Burlington, VT, USA.
| | - Sulamunn R M Coleman
- Department of Psychiatry, University of Vermont, Burlington, VT, USA; Vermont Center on Behavior and Health, University of Vermont, Burlington, VT, USA
| |
Collapse
|
3
|
Menson KE, Coleman SRM. Smoking and pulmonary health in women: A narrative review and behavioral health perspective. Prev Med 2024; 185:108029. [PMID: 38851402 DOI: 10.1016/j.ypmed.2024.108029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 05/24/2024] [Accepted: 06/04/2024] [Indexed: 06/10/2024]
Abstract
OBJECTIVE Cigarette smoking prevalence has declined slower among women than men, and smoking-related pulmonary disease (PD) has risen among women. Given these trends, there is a critical need to understand and mitigate PD risk among women who smoke. The purpose of this narrative review and commentary is to highlight important evidence from the literature on smoking and PD among women. METHODS This review focuses broadly on examining cigarette smoking and PD among women within six topic areas: (1) demographic characteristics and prevalence of smoking, (2) smoking behavior, (3) lung cancer, (4) obstructive PD, (5) diagnostic and treatment disparities, and (6) gaps in the literature and potential directions for future research and treatment. RESULTS Growing evidence indicates that compared to men, women are at increased risk for developing smoking-related PD and poorer PD outcomes. Gender disparities in smoking-related PD may be largely accounted for by genetic differences and sex hormones contributing to PD pathogenesis and presentation, smoking behavior, nicotine dependence, and pathogen/carcinogen clearance. Moreover, gender disparities in smoking-related PD may be exacerbated by important social determinants (e.g., women with less formal education and those from minoritized groups may be at especially high risk for poor PD outcomes due to higher rates of smoking). CONCLUSION Rising rates of smoking-related PD among women risk widening diagnostic and treatment disparities. Ongoing research is needed to explore potentially complex relationships between sex, gender, and smoking-related PD processes and outcomes, and to improve smoking-cessation and PD treatment for women.
Collapse
Affiliation(s)
- Katherine E Menson
- Division of Pulmonary and Critical Care Medicine, University of Vermont, Burlington, VT, USA; Larner College of Medicine, University of Vermont, Burlington, VT, USA.
| | - Sulamunn R M Coleman
- Department of Psychiatry, University of Vermont, Burlington, VT, USA; Vermont Center on Behavior and Health, University of Vermont, Burlington, VT, USA
| |
Collapse
|
4
|
Li CX, Chen H, Zounemat-Kermani N, Adcock IM, Sköld CM, Zhou M, Wheelock ÅM. Consensus clustering with missing labels (ccml): a consensus clustering tool for multi-omics integrative prediction in cohorts with unequal sample coverage. Brief Bioinform 2023; 25:bbad501. [PMID: 38205966 PMCID: PMC10782800 DOI: 10.1093/bib/bbad501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 11/14/2023] [Accepted: 12/01/2023] [Indexed: 01/12/2024] Open
Abstract
Multi-omics data integration is a complex and challenging task in biomedical research. Consensus clustering, also known as meta-clustering or cluster ensembles, has become an increasingly popular downstream tool for phenotyping and endotyping using multiple omics and clinical data. However, current consensus clustering methods typically rely on ensembling clustering outputs with similar sample coverages (mathematical replicates), which may not reflect real-world data with varying sample coverages (biological replicates). To address this issue, we propose a new consensus clustering with missing labels (ccml) strategy termed ccml, an R protocol for two-step consensus clustering that can handle unequal missing labels (i.e. multiple predictive labels with different sample coverages). Initially, the regular consensus weights are adjusted (normalized) by sample coverage, then a regular consensus clustering is performed to predict the optimal final cluster. We applied the ccml method to predict molecularly distinct groups based on 9-omics integration in the Karolinska COSMIC cohort, which investigates chronic obstructive pulmonary disease, and 24-omics handprint integrative subgrouping of adult asthma patients of the U-BIOPRED cohort. We propose ccml as a downstream toolkit for multi-omics integration analysis algorithms such as Similarity Network Fusion and robust clustering of clinical data to overcome the limitations posed by missing data, which is inevitable in human cohorts consisting of multiple data modalities. The ccml tool is available in the R language (https://CRAN.R-project.org/package=ccml, https://github.com/pulmonomics-lab/ccml, or https://github.com/ZhoulabCPH/ccml).
Collapse
Affiliation(s)
- Chuan-Xing Li
- Respiratory Medicine Unit, Department of Medicine Solna & Centre for Molecular Medicine, Karolinska Institutet
| | - Hongyan Chen
- School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, China
| | - Nazanin Zounemat-Kermani
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United Kingdom
- Data Science Institute, Imperial College London, London, United Kingdom
| | - Ian M Adcock
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United Kingdom
- Data Science Institute, Imperial College London, London, United Kingdom
| | - C Magnus Sköld
- Respiratory Medicine Unit, Department of Medicine Solna & Centre for Molecular Medicine, Karolinska Institutet
- Department of Respiratory Medicine and Allergy, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Meng Zhou
- School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, China
| | - Åsa M Wheelock
- Respiratory Medicine Unit, Department of Medicine Solna & Centre for Molecular Medicine, Karolinska Institutet
- Department of Respiratory Medicine and Allergy, Karolinska University Hospital Solna, Stockholm, Sweden
| | | |
Collapse
|
5
|
Rezaeeyan H, Arabfard M, Rasouli HR, Shahriary A, Gh BFNM. Evaluation of common protein biomarkers involved in the pathogenesis of respiratory diseases with proteomic methods: A systematic review. Immun Inflamm Dis 2023; 11:e1090. [PMID: 38018577 PMCID: PMC10659759 DOI: 10.1002/iid3.1090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 09/22/2023] [Accepted: 11/04/2023] [Indexed: 11/30/2023] Open
Abstract
AIM Respiratory disease (RD) is one of the most common diseases characterized by lung dysfunction. Many diagnostic mechanisms have been used to identify the pathogenic agents of responsible for RD. Among these, proteomics emerges as a valuable diagnostic method for pinpointing the specific proteins involved in RD pathogenesis. Therefore, in this study, for the first time, we examined the protein markers involved in the pathogenesis of chronic obstructive pulmonary disease (COPD), idiopathic pulmonary fibrosis (IPF), asthma, bronchiolitis obliterans (BO), and chemical warfare victims exposed to mustard gas, using the proteomics method as a systematic study. MATERIALS AND METHODS A systematic search was performed up to September 2023 on several databases, including PubMed, Scopus, ISI Web of Science, and Cochrane. In total, selected 4246 articles were for evaluation according to the criteria. Finally, 119 studies were selected for this systematic review. RESULTS A total of 13,806 proteins were identified, 6471 in COPD, 1603 in Asthma, 5638 in IPF, three in BO, and 91 in mustard gas exposed victims. Alterations in the expression of these proteins were observed in the respective diseases. After evaluation, the results showed that 31 proteins were found to be shared among all five diseases. CONCLUSION Although these 31 proteins regulate different factors and molecular pathways in all five diseases, they ultimately lead to the regulation of inflammatory pathways. In other words, the expression of some proteins in COPD and mustard-exposed patients increases inflammatory reactions, while in IPF, they cause lung fibrosis. Asthma, causes allergic reactions due to T-cell differentiation toward Th2.
Collapse
Affiliation(s)
- Hadi Rezaeeyan
- Chemical Injuries Research Center, Systems Biology and Poisonings InstituteBaqiyatallah University of Medical SciencesTehranIran
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion MedicineIranian Blood Transfusion Organization (IBTO)TehranIran
| | - Masoud Arabfard
- Chemical Injuries Research Center, Systems Biology and Poisonings InstituteBaqiyatallah University of Medical SciencesTehranIran
| | - Hamid R. Rasouli
- Trauma Research CenterBaqiyatallah University of Medical SciencesTehranIran
| | - Alireza Shahriary
- Chemical Injuries Research Center, Systems Biology and Poisonings InstituteBaqiyatallah University of Medical SciencesTehranIran
| | - B. Fatemeh Nobakht M. Gh
- Chemical Injuries Research Center, Systems Biology and Poisonings InstituteBaqiyatallah University of Medical SciencesTehranIran
| |
Collapse
|
6
|
Gea J, Enríquez-Rodríguez CJ, Agranovich B, Pascual-Guardia S. Update on metabolomic findings in COPD patients. ERJ Open Res 2023; 9:00180-2023. [PMID: 37908399 PMCID: PMC10613990 DOI: 10.1183/23120541.00180-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 08/15/2023] [Indexed: 11/02/2023] Open
Abstract
COPD is a heterogeneous disorder that shows diverse clinical presentations (phenotypes and "treatable traits") and biological mechanisms (endotypes). This heterogeneity implies that to carry out a more personalised clinical management, it is necessary to classify each patient accurately. With this objective, and in addition to clinical features, it would be very useful to have well-defined biological markers. The search for these markers may either be done through more conventional laboratory and hypothesis-driven techniques or relatively blind high-throughput methods, with the omics approaches being suitable for the latter. Metabolomics is the science that studies biological processes through their metabolites, using various techniques such as gas and liquid chromatography, mass spectrometry and nuclear magnetic resonance. The most relevant metabolomics studies carried out in COPD highlight the importance of metabolites involved in pathways directly related to proteins (peptides and amino acids), nucleic acids (nitrogenous bases and nucleosides), and lipids and their derivatives (especially fatty acids, phospholipids, ceramides and eicosanoids). These findings indicate the relevance of inflammatory-immune processes, oxidative stress, increased catabolism and alterations in the energy production. However, some specific findings have also been reported for different COPD phenotypes, demographic characteristics of the patients, disease progression profiles, exacerbations, systemic manifestations and even diverse treatments. Unfortunately, the studies carried out to date have some limitations and shortcomings and there is still a need to define clear metabolomic profiles with clinical utility for the management of COPD and its implicit heterogeneity.
Collapse
Affiliation(s)
- Joaquim Gea
- Respiratory Medicine Department, Hospital del Mar – IMIM, Barcelona, Spain
- MELIS Department, Universitat Pompeu Fabra, Barcelona, Spain
- CIBERES, ISCIII, Barcelona, Spain
| | - César J. Enríquez-Rodríguez
- Respiratory Medicine Department, Hospital del Mar – IMIM, Barcelona, Spain
- MELIS Department, Universitat Pompeu Fabra, Barcelona, Spain
| | - Bella Agranovich
- Rappaport Institute for Research in the Medical Sciences, Technion University, Haifa, Israel
| | - Sergi Pascual-Guardia
- Respiratory Medicine Department, Hospital del Mar – IMIM, Barcelona, Spain
- MELIS Department, Universitat Pompeu Fabra, Barcelona, Spain
- CIBERES, ISCIII, Barcelona, Spain
| |
Collapse
|
7
|
Merikallio H, Pincikova T, Kotortsi I, Karimi R, Li CX, Forsslund H, Mikko M, Nyrén S, Lappi-Blanco E, Wheelock ÅM, Kaarteenaho R, Sköld MC. Mucins 3A and 3B Are Expressed in the Epithelium of Human Large Airway. Int J Mol Sci 2023; 24:13546. [PMID: 37686350 PMCID: PMC10487631 DOI: 10.3390/ijms241713546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/25/2023] [Accepted: 08/29/2023] [Indexed: 09/10/2023] Open
Abstract
Aberrant mucus secretion is a hallmark of chronic obstructive pulmonary disease (COPD). Expression of the membrane-tethered mucins 3A and 3B (MUC3A, MUC3B) in human lung is largely unknown. In this observational cross-sectional study, we recruited subjects 45-65 years old from the general population of Stockholm, Sweden, during the years 2007-2011. Bronchial mucosal biopsies, bronchial brushings, and bronchoalveolar lavage fluid (BALF) were retrieved from COPD patients (n = 38), healthy never-smokers (n = 40), and smokers with normal lung function (n = 40). Protein expression of MUC3A and MUC3B in bronchial mucosal biopsies was assessed by immunohistochemical staining. In a subgroup of subjects (n = 28), MUC3A and MUC3B mRNAs were quantified in bronchial brushings using microarray. Non-parametric tests were used to perform correlation and group comparison analyses. A value of p < 0.05 was considered statistically significant. MUC3A and MUC3B immunohistochemical expression was localized to ciliated cells. MUC3B was also expressed in basal cells. MUC3A and MUC3B immunohistochemical expression was equal in all study groups but subjects with emphysema had higher MUC3A expression, compared to those without emphysema. Smokers had higher mRNA levels of MUC3A and MUC3B than non-smokers. MUC3A and MUC3B mRNA were higher in male subjects and correlated negatively with expiratory air flows. MUC3B mRNA correlated positively with total cell concentration and macrophage percentage, and negatively with CD4/CD8 T cell ratio in BALF. We concluded that MUC3A and MUC3B in large airways may be a marker of disease or may play a role in the pathophysiology of airway obstruction.
Collapse
Affiliation(s)
- Heta Merikallio
- Research Unit of Biomedicine and Internal Medicine, University of Oulu, 90570 Oulu, Finland; (H.M.)
- Center of Internal Medicine and Respiratory Medicine, Medical Research Center Oulu, University Hospital of Oulu, 90220 Oulu, Finland
| | - Terezia Pincikova
- Respiratory Medicine Unit, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden
- Stockholm CF-Center, Albatross, K56, Karolinska University Hospital Huddinge, 141 86 Stockholm, Sweden
| | - Ioanna Kotortsi
- Respiratory Medicine Unit, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden
- Department of Respiratory Medicine and Allergy, Karolinska University Hospital, 171 77 Stockholm, Sweden
| | - Reza Karimi
- Respiratory Medicine Unit, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Chuan-Xing Li
- Respiratory Medicine Unit, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Helena Forsslund
- Respiratory Medicine Unit, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Mikael Mikko
- Respiratory Medicine Unit, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Sven Nyrén
- Department of Molecular Medicine and Surgery, Division of Radiology, Karolinska Institutet, Karolinska University Hospital Solna, 171 76 Stockholm, Sweden
| | - Elisa Lappi-Blanco
- Cancer and Translational Medicine Research Unit, Department of Pathology, University Hospital of Oulu, Oulu University, 90220 Oulu, Finland
| | - Åsa M. Wheelock
- Respiratory Medicine Unit, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden
- Department of Respiratory Medicine and Allergy, Karolinska University Hospital, 171 77 Stockholm, Sweden
| | - Riitta Kaarteenaho
- Research Unit of Biomedicine and Internal Medicine, University of Oulu, 90570 Oulu, Finland; (H.M.)
- Center of Internal Medicine and Respiratory Medicine, Medical Research Center Oulu, University Hospital of Oulu, 90220 Oulu, Finland
| | - Magnus C. Sköld
- Respiratory Medicine Unit, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden
- Department of Respiratory Medicine and Allergy, Karolinska University Hospital, 171 77 Stockholm, Sweden
| |
Collapse
|
8
|
Chen Z, Wang J, Wang H, Yao Y, Deng H, Peng J, Li X, Wang Z, Chen X, Xiong W, Wang Q, Zhu T. Machine learning reveals sex differences in clinical features of acute exacerbation of chronic obstructive pulmonary disease: A multicenter cross-sectional study. Front Med (Lausanne) 2023; 10:1105854. [PMID: 37056727 PMCID: PMC10086189 DOI: 10.3389/fmed.2023.1105854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 02/09/2023] [Indexed: 03/30/2023] Open
Abstract
Introduction Intrinsically, chronic obstructive pulmonary disease (COPD) is a highly heterogonous disease. Several sex differences in COPD, such as risk factors and prevalence, were identified. However, sex differences in clinical features of acute exacerbation chronic obstructive pulmonary disease (AECOPD) were not well explored. Machine learning showed a promising role in medical practice, including diagnosis prediction and classification. Then, sex differences in clinical manifestations of AECOPD were explored by machine learning approaches in this study. Methods In this cross-sectional study, 278 male patients and 81 female patients hospitalized with AECOPD were included. Baseline characteristics, clinical symptoms, and laboratory parameters were analyzed. The K-prototype algorithm was used to explore the degree of sex differences. Binary logistic regression, random forest, and XGBoost models were performed to identify sex-associated clinical manifestations in AECOPD. Nomogram and its associated curves were established to visualize and validate binary logistic regression. Results The predictive accuracy of sex was 83.930% using the k-prototype algorithm. Binary logistic regression revealed that eight variables were independently associated with sex in AECOPD, which was visualized by using a nomogram. The AUC of the ROC curve was 0.945. The DCA curve showed that the nomogram had more clinical benefits, with thresholds from 0.02 to 0.99. The top 15 sex-associated important variables were identified by random forest and XGBoost, respectively. Subsequently, seven clinical features, including smoking, biomass fuel exposure, GOLD stages, PaO2, serum potassium, serum calcium, and blood urea nitrogen (BUN), were concurrently identified by three models. However, CAD was not identified by machine learning models. Conclusions Overall, our results support that the clinical features differ markedly by sex in AECOPD. Male patients presented worse lung function and oxygenation, less biomass fuel exposure, more smoking, renal dysfunction, and hyperkalemia than female patients with AECOPD. Furthermore, our results also suggest that machine learning is a promising and powerful tool in clinical decision-making.
Collapse
Affiliation(s)
- Zhihong Chen
- Respiratory Medicine and Critical Care Medicine, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Jiajia Wang
- Rheumatology Medicine, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hanchao Wang
- Respiratory Medicine and Critical Care Medicine, and Preclinical Research Center, Suining Central Hospital, Suining, China
| | - Yu Yao
- Respiratory Medicine and Critical Care Medicine, and Preclinical Research Center, Suining Central Hospital, Suining, China
| | - Huojin Deng
- Respiratory Medicine and Critical Care Medicine, ZhuJiang Hospital of Southern Medical University, Guangzhou, China
| | - Junnan Peng
- Respiratory Medicine and Critical Care Medicine, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xinglong Li
- Respiratory Medicine and Critical Care Medicine, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhongruo Wang
- Department of Mathematics, University of California, Davis, CA, United States
| | - Xingru Chen
- Respiratory Medicine and Critical Care Medicine, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wei Xiong
- Respiratory Medicine and Critical Care Medicine, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qin Wang
- Respiratory Medicine and Critical Care Medicine, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Tao Zhu
- Respiratory Medicine and Critical Care Medicine, and Preclinical Research Center, Suining Central Hospital, Suining, China
| |
Collapse
|
9
|
Reddy KD, Oliver BGG. Sexual dimorphism in chronic respiratory diseases. Cell Biosci 2023; 13:47. [PMID: 36882807 PMCID: PMC9993607 DOI: 10.1186/s13578-023-00998-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 02/23/2023] [Indexed: 03/09/2023] Open
Abstract
Sex differences in susceptibility, severity, and progression are prevalent for various diseases in multiple organ systems. This phenomenon is particularly apparent in respiratory diseases. Asthma demonstrates an age-dependent pattern of sexual dimorphism. However, marked differences between males and females exist in other pervasive conditions such as chronic obstructive pulmonary disease (COPD) and lung cancer. The sex hormones estrogen and testosterone are commonly considered the primary factors causing sexual dimorphism in disease. However, how they contribute to differences in disease onset between males and females remains undefined. The sex chromosomes are an under-investigated fundamental form of sexual dimorphism. Recent studies highlight key X and Y-chromosome-linked genes that regulate vital cell processes and can contribute to disease-relevant mechanisms. This review summarises patterns of sex differences in asthma, COPD and lung cancer, highlighting physiological mechanisms causing the observed dimorphism. We also describe the role of the sex hormones and present candidate genes on the sex chromosomes as potential factors contributing to sexual dimorphism in disease.
Collapse
Affiliation(s)
- Karosham Diren Reddy
- Respiratory and Cellular Molecular Biology Group, Woolcock Institute of Medical Research, Glebe, NSW, 2037, Australia.
- School of Life Science, University of Technology Sydney, Ultimo, NSW, 2007, Australia.
| | - Brian Gregory George Oliver
- Respiratory and Cellular Molecular Biology Group, Woolcock Institute of Medical Research, Glebe, NSW, 2037, Australia
- School of Life Science, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| |
Collapse
|
10
|
Gea J, Enríquez-Rodríguez CJ, Pascual-Guardia S. Metabolomics in COPD. Arch Bronconeumol 2023; 59:311-321. [PMID: 36717301 DOI: 10.1016/j.arbres.2022.12.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 11/28/2022] [Accepted: 12/07/2022] [Indexed: 01/20/2023]
Abstract
The clinical presentation of chronic obstructive pulmonary disease (COPD) is highly heterogeneous. Attempts have been made to define subpopulations of patients who share clinical characteristics (phenotypes and treatable traits) and/or biological characteristics (endotypes), in order to offer more personalized care. Assigning a patient to any of these groups requires the identification of both clinical and biological markers. Ideally, biological markers should be easily obtained from blood or urine, but these may lack specificity. Biomarkers can be identified initially using conventional or more sophisticated techniques. However, the more sophisticated techniques should be simplified in the future if they are to have clinical utility. The -omics approach offers a methodology that can assist in the investigation and identification of useful markers in both targeted and blind searches. Specifically, metabolomics is the science that studies biological processes involving metabolites, which can be intermediate or final products. The metabolites associated with COPD and their specific phenotypic and endotypic features have been studied using various techniques. Several compounds of particular interest have emerged, namely, several types of lipids and derivatives (mainly phospholipids, but also ceramides, fatty acids and eicosanoids), amino acids, coagulation factors, and nucleic acid components, likely to be involved in their function, protein catabolism, energy production, oxidative stress, immune-inflammatory response and coagulation disorders. However, clear metabolomic profiles of the disease and its various manifestations that may already be applicable in clinical practice still need to be defined.
Collapse
Affiliation(s)
- Joaquim Gea
- Servicio de Neumología, Hospital del Mar - IMIM, Barcelona, Spain; Dpt. MELIS, Universitat Pompeu Fabra, Barcelona, Spain; CIBERES, ISCIII, Barcelona, Spain.
| | - César J Enríquez-Rodríguez
- Servicio de Neumología, Hospital del Mar - IMIM, Barcelona, Spain; Dpt. MELIS, Universitat Pompeu Fabra, Barcelona, Spain
| | - Sergi Pascual-Guardia
- Servicio de Neumología, Hospital del Mar - IMIM, Barcelona, Spain; Dpt. MELIS, Universitat Pompeu Fabra, Barcelona, Spain; CIBERES, ISCIII, Barcelona, Spain
| |
Collapse
|
11
|
Li CX, Gao J, Sköld CM, Wheelock ÅM. miRNA-mRNA-protein dysregulated network in COPD in women. Front Genet 2022; 13:1010048. [PMID: 36468026 PMCID: PMC9712209 DOI: 10.3389/fgene.2022.1010048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 10/10/2022] [Indexed: 10/14/2023] Open
Abstract
Rationale: Chronic obstructive pulmonary disease (COPD) is a complex disease caused by a multitude of underlying mechanisms, and molecular mechanistic modeling of COPD, especially at a multi-molecular level, is needed to facilitate the development of molecular diagnostic and prognostic tools and efficacious treatments. Objectives: To investigate the miRNA-mRNA-protein dysregulated network to facilitate prediction of biomarkers and disease subnetwork in COPD in women. Measurements and Results: Three omics data blocks (mRNA, miRNA, and protein) collected from BAL cells from female current-smoker COPD patients, smokers with normal lung function, and healthy never-smokers were integrated with miRNA-mRNA-protein regulatory networks to construct a COPD-specific dysregulated network. Furthermore, downstream network topology, literature annotation, and functional enrichment analysis identified both known and novel disease-related biomarkers and pathways. Both abnormal regulations in miRNA-induced mRNA transcription and protein translation repression play roles in COPD. Finally, the let-7-AIFM1-FKBP1A pathway is highlighted in COPD pathology. Conclusion: For the first time, a comprehensive miRNA-mRNA-protein dysregulated network of primary immune cells from the lung related to COPD in females was constructed to elucidate specific biomarkers and disease pathways. The multi-omics network provides a new molecular insight from a multi-molecular aspect and highlights dysregulated interactions. The highlighted let-7-AIFM1-FKBP1A pathway also indicates new hypotheses of COPD pathology.
Collapse
Affiliation(s)
- Chuan Xing Li
- Respiratory Medicine Unit, Department of Medicine, Centre for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Jing Gao
- Respiratory Medicine Unit, Department of Medicine, Centre for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Heart and Lung Centre, Department of Pulmonary Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - C. Magnus Sköld
- Respiratory Medicine Unit, Department of Medicine, Centre for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Stockholm, Sweden
| | - Åsa M. Wheelock
- Respiratory Medicine Unit, Department of Medicine, Centre for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
12
|
Lamichhane R, Patial S, Saini Y. Higher susceptibility of males to bleomycin-induced pulmonary inflammation is associated with sex-specific transcriptomic differences in myeloid cells. Toxicol Appl Pharmacol 2022; 454:116228. [PMID: 36087614 PMCID: PMC9793441 DOI: 10.1016/j.taap.2022.116228] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/24/2022] [Accepted: 09/02/2022] [Indexed: 12/30/2022]
Abstract
Idiopathic pulmonary fibrosis, a condition with likely genetic and environmental etiology, is relatively more prevalent with poor prognosis in human males. However, the underlying mechanisms for these gender-associated differences in the severity of fibrosis remain unknown. Here, we tested the hypothesis that the transcriptomic repertoire of myeloid cells determines the higher susceptibility of male mice to bleomycin (BLM)-induced lung fibrosis. Adult mice were oropharyngeally challenged with saline or BLM. Lung injury, inflammation, and fibrosis outcomes were assessed, and airspace myeloid-cells were subjected to RNA-sequencing. As compared with the female mice, the male mice manifested significantly increased lung injury, inflammation, proinflammatory cytokines (IL-6, IL-1β, IL-7, and IP-10), and fibrosis in response to BLM challenge. Interestingly, several pro-inflammatory and extracellular matrix-associated genes were significantly up-regulated in male myeloid-cells compared to female myeloid-cells in the saline-control group. Similarly, BLM challenge resulted in greater pro-inflammatory and pro-fibrotic transcriptomic changes in male compared to female myeloid cells. On the other hand, anti-inflammatory and regulatory cytokine, Il10 and Ifng respectively, were uniquely upregulated in BLM-challenged female but not in male myeloid cells when compared to their respective saline-control groups. Further, cross-sex bone marrow transplantation experiments revealed that male hematopoietic progenitor cells (HPCs) increased the granulocytic infiltration in female mice while female HPCs decreased the granulocytic infiltration in male mice post-BLM challenge. These findings suggest that there are inherent transcriptomic differences between the male and female lung myeloid cells and that the pro-inflammatory nature of male myeloid cells is sufficient to increase the susceptibility of female mice to BLM-induced inflammation.
Collapse
|
13
|
Godbole S, Bowler RP. Metabolome Features of COPD: A Scoping Review. Metabolites 2022; 12:621. [PMID: 35888745 PMCID: PMC9324381 DOI: 10.3390/metabo12070621] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/16/2022] [Accepted: 06/20/2022] [Indexed: 02/03/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a complex heterogeneous disease state with multiple phenotypic presentations that include chronic bronchitis and emphysema. Although COPD is a lung disease, it has systemic manifestations that are associated with a dysregulated metabolome in extrapulmonary compartments (e.g., blood and urine). In this scoping review of the COPD metabolomics literature, we identified 37 publications with a primary metabolomics investigation of COPD phenotypes in human subjects through Google Scholar, PubMed, and Web of Science databases. These studies consistently identified a dysregulation of the TCA cycle, carnitines, sphingolipids, and branched-chain amino acids. Many of the COPD metabolome pathways are confounded by age and sex. The effects of COPD in young versus old and male versus female need further focused investigations. There are also few studies of the metabolome's association with COPD progression, and it is unclear whether the markers of disease and disease severity are also important predictors of disease progression.
Collapse
Affiliation(s)
- Suneeta Godbole
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Russell P. Bowler
- Division of Medicine, National Jewish Health, Denver, CO 80206, USA;
| |
Collapse
|
14
|
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: 20] [Impact Index Per Article: 10.0] [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
|
15
|
Siegfried JM. Sex and Gender Differences in Lung Cancer and Chronic Obstructive Lung Disease. Endocrinology 2022; 163:6470418. [PMID: 34927202 DOI: 10.1210/endocr/bqab254] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Indexed: 11/19/2022]
Abstract
Two highly prevalent pulmonary diseases, lung cancer and chronic obstructive lung disease (COPD), show both sex and gender differences in their presentations and outcomes. Sex differences are defined as biological differences associated with the male vs female genotype, and gender differences are defined as behavioral or social differences that primarily arise because of gender identity. The incidence of both lung cancer and COPD has increased dramatically in women over the past 50 years, and both are associated with chronic pulmonary inflammation. Development of COPD is also a risk factor for lung cancer. In this review, the main differences in lung cancer and COPD biology observed between men and women will be summarized. Potential causative factors will be discussed, including the role of estrogen in promoting pro-growth and inflammatory phenotypes which may contribute to development of both lung cancer and COPD. Response of the innate and adaptive immune system to estrogen is a likely factor in the biology of both lung cancer and COPD. Estrogen available from synthesis by reproductive organs as well as local pulmonary estrogen synthesis may be involved in activating estrogen receptors expressed by multiple cell types in the lung. Estrogenic actions, although more pronounced in women, may also have importance in the biology of lung cancer and COPD in men. Effects of estrogen are also timing and context dependent; the multiple cell types that mediate estrogen action in the lungs may confer both positive and negative effects on disease processes.
Collapse
Affiliation(s)
- Jill M Siegfried
- Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15260, USA
| |
Collapse
|
16
|
Kalafatis D, Löfdahl A, Näsman P, Dellgren G, Wheelock ÅM, Elowsson Rendin L, Sköld M, Westergren-Thorsson G. Distal Lung Microenvironment Triggers Release of Mediators Recognized as Potential Systemic Biomarkers for Idiopathic Pulmonary Fibrosis. Int J Mol Sci 2021; 22:ijms222413421. [PMID: 34948231 PMCID: PMC8704101 DOI: 10.3390/ijms222413421] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/03/2021] [Accepted: 12/11/2021] [Indexed: 12/27/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive fibrotic lung disease with an unmet need of biomarkers that can aid in the diagnostic and prognostic assessment of the disease and response to treatment. In this two-part explorative proteomic study, we demonstrate how proteins associated with tissue remodeling, inflammation and chemotaxis such as MMP7, CXCL13 and CCL19 are released in response to aberrant extracellular matrix (ECM) in IPF lung. We used a novel ex vivo model where decellularized lung tissue from IPF patients and healthy donors were repopulated with healthy fibroblasts to monitor locally released mediators. Results were validated in longitudinally collected serum samples from 38 IPF patients and from 77 healthy controls. We demonstrate how proteins elevated in the ex vivo model (e.g., MMP7), and other serum proteins found elevated in IPF patients such as HGF, VEGFA, MCP-3, IL-6 and TNFRSF12A, are associated with disease severity and progression and their response to antifibrotic treatment. Our study supports the model’s applicability in studying mechanisms involved in IPF and provides additional evidence for both established and potentially new biomarkers in IPF.
Collapse
Affiliation(s)
- Dimitrios Kalafatis
- Respiratory Medicine Unit, Department of Medicine Solna and Center for Molecular Medicine, Karolinska Institutet, SE-171 76 Stockholm, Sweden; (Å.M.W.); (M.S.)
- Correspondence: ; Tel.: +46-72-3416617
| | - Anna Löfdahl
- Department of Experimental Medical Science, Lung Biology, Lund University, SE-221 84 Lund, Sweden; (A.L.); (L.E.R.); (G.W.-T.)
| | - Per Näsman
- Center for Safety Research, KTH, Royal Institute of Technology, SE-100 44 Stockholm, Sweden;
| | - Göran Dellgren
- Department of Cardiothoracic Surgery and Transplant Institute, Sahlgrenska University Hospital, SE-413 45 Gothenburg, Sweden;
| | - Åsa M. Wheelock
- Respiratory Medicine Unit, Department of Medicine Solna and Center for Molecular Medicine, Karolinska Institutet, SE-171 76 Stockholm, Sweden; (Å.M.W.); (M.S.)
- Department of Respiratory Medicine and Allergy, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| | - Linda Elowsson Rendin
- Department of Experimental Medical Science, Lung Biology, Lund University, SE-221 84 Lund, Sweden; (A.L.); (L.E.R.); (G.W.-T.)
| | - Magnus Sköld
- Respiratory Medicine Unit, Department of Medicine Solna and Center for Molecular Medicine, Karolinska Institutet, SE-171 76 Stockholm, Sweden; (Å.M.W.); (M.S.)
- Department of Respiratory Medicine and Allergy, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| | - Gunilla Westergren-Thorsson
- Department of Experimental Medical Science, Lung Biology, Lund University, SE-221 84 Lund, Sweden; (A.L.); (L.E.R.); (G.W.-T.)
| |
Collapse
|
17
|
Abstract
There are many differences between men and women in risk factor vulnerability and impact, symptom development, presentation, clinical manifestations, and outcomes of chronic obstructive pulmonary disease (COPD). These differences have been inadequately explored, which is of particular concern in view of the delay in diagnosis and treatment that occurs for both men and women, and the need to use all therapies in a targeted and tailored way for best clinical gain and least harm. Women are generally underrepresented in randomized trials, as are non-tobacco-related phenotypes of COPD. We must garner the information available to better describe optimal treatment approaches.
Collapse
Affiliation(s)
- Christine Jenkins
- Respiratory Group, The George Institute for Global Health, Sydney, Level 5, 1 King Street, Newtown, New South Wales 2042 Australia; UNSW Medicine and Health, UNSW Sydney, NSW, Australia; Concord Clinical School, University of Sydney, NSW, Australia.
| |
Collapse
|
18
|
Kokelj S, Östling J, Georgi B, Fromell K, Ekdahl KN, Olsson HK, Olin AC. Smoking induces sex-specific changes in the small airway proteome. Respir Res 2021; 22:234. [PMID: 34429114 PMCID: PMC8385797 DOI: 10.1186/s12931-021-01825-6] [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: 03/02/2021] [Accepted: 07/12/2021] [Indexed: 12/31/2022] Open
Abstract
Introduction Cigarette smoke triggers many cellular and signaling responses in the lung and the resulting inflammation plays a central role in smoke-related lung diseases, such as COPD. We explored the effects of smoking on the small airway proteome in samples obtained by collection of exhaled particles with the aim to identify specific proteins dysregulated by smoking. Methods Exhaled particles were obtained from 38 current smokers, 47 former smokers and 22 healthy controls with the PExA method. 120 ng of sample was collected from individual subjects and analyzed with the SOMAscan proteomics platform. General linear model-based statistics were performed. Results Two hundred and three proteins were detected in at least half of 107 total samples. Active smoking exerted a significant impact on the protein composition of respiratory tract lining fluid (RTLF), with 81 proteins altered in current smokers compared to never smokers (p < 0.05, q < 0.124). Among the proteins most clearly discriminating between current and never smokers were sRAGE, FSTL3, SPOCK2 and protein S, all of them being less abundant in current smokers. Analysis stratified for sex unveiled sex differences with more pronounced proteomic alterations due to active smoking in females than males. Proteins whose abundance was altered by active smoking in women were to a larger extent related to the complement system. The small airway protein profile of former smokers appeared to be more similar to that observed in never smokers. Conclusions The study shows that smoking has a strong impact on protein expression in the small airways, and that smoking affects men and women differently, suggesting PExA sampling combined with high sensitivity protein analysis offers a promising platform for early detection of COPD and identification of novel COPD drug targets. Supplementary Information The online version contains supplementary material available at 10.1186/s12931-021-01825-6.
Collapse
Affiliation(s)
- Spela Kokelj
- Occupational and Environmental Medicine, School of Public Health and Community Medicine, Inst. of Medicine, Sahlgrenska Academy, University of Gothenburg, Box 414, 405 30, Gothenburg, Sweden.
| | - Jörgen Östling
- PExA AB, Gothenburg, Sweden.,Translational Science and Experimental Medicine, Research and Early Development, Respiratory and Immunology, AstraZeneca, BioPharmaceuticals R&D, Gothenburg, Sweden
| | - Benjamin Georgi
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory and Immunology, AstraZeneca, BioPharmaceuticals R&D, Gothenburg, Sweden
| | - Karin Fromell
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Kristina Nilsson Ekdahl
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.,Linnaeus Centre for Biomaterials Chemistry, Linnaeus University, Kalmar, Sweden
| | - Henric K Olsson
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory and Immunology, AstraZeneca, BioPharmaceuticals R&D, Gothenburg, Sweden
| | - Anna-Carin Olin
- Occupational and Environmental Medicine, School of Public Health and Community Medicine, Inst. of Medicine, Sahlgrenska Academy, University of Gothenburg, Box 414, 405 30, Gothenburg, Sweden
| |
Collapse
|
19
|
Serban KA, Pratte KA, Bowler RP. Protein Biomarkers for COPD Outcomes. Chest 2021; 159:2244-2253. [PMID: 33434499 PMCID: PMC8213963 DOI: 10.1016/j.chest.2021.01.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 12/14/2020] [Accepted: 01/01/2021] [Indexed: 12/15/2022] Open
Abstract
COPD is a clinically heterogeneous syndrome characterized by injury to airways, airspaces, and lung vasculature and usually caused by tobacco smoke and/or air pollution exposure. COPD is also independently associated with nonpulmonary comorbidities (eg, cardiovascular disease) and malignancies (eg, GI, bladder), suggesting a role for systemic injury. Since not all those with exposure develop COPD, there has been a search for plasma and lung biomarkers that confer increased cross-sectional and longitudinal risk. This search typically focuses on clinically relevant COPD outcomes such as FEV1, FEV1 decline, CT measurements of emphysema, or exacerbation frequency. The rapid advances in omics technology and the molecular phenotyping of COPD cohorts now permit large-scale evaluation of genetic, transcriptomic, proteomic, and metabolic biomarkers. This review focuses on protein biomarkers associated with clinically relevant COPD outcomes. The prototypic COPD protein biomarker is alpha-1 antitrypsin; however, this biomarker only accounts for 1% to 5% of COPD. This article reviews and summarizes the evidence for other validated biomarkers for each COPD outcome, and discusses their advantages, weaknesses, and required regulatory steps to move the biomarker from the bench into clinic. Although we highlight the emergence of many novel biomarkers (eg, fibrinogen, soluble receptor for advanced glycation, surfactant protein D, club cell secretory protein), there is increasing evidence that individual biomarkers only explain a fraction of the increased COPD risk and that multiple biomarker panels are needed to completely explain clinical variation and risk in individuals and populations.
Collapse
Affiliation(s)
- Karina A Serban
- National Jewish Health, Denver; University of Colorado, Anschutz Medical Campus, Aurora, CO.
| | | | - Russell P Bowler
- National Jewish Health, Denver; University of Colorado, Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
20
|
Gillenwater LA, Kechris KJ, Pratte KA, Reisdorph N, Petrache I, Labaki WW, O’Neal W, Krishnan JA, Ortega VE, DeMeo DL, Bowler RP. Metabolomic Profiling Reveals Sex Specific Associations with Chronic Obstructive Pulmonary Disease and Emphysema. Metabolites 2021; 11:161. [PMID: 33799786 PMCID: PMC7999201 DOI: 10.3390/metabo11030161] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/25/2021] [Accepted: 03/08/2021] [Indexed: 12/12/2022] Open
Abstract
Susceptibility and progression of lung disease, as well as response to treatment, often differ by sex, yet the metabolic mechanisms driving these sex-specific differences are still poorly understood. Women with chronic obstructive pulmonary disease (COPD) have less emphysema and more small airway disease on average than men, though these differences become less pronounced with more severe airflow limitation. While small studies of targeted metabolites have identified compounds differing by sex and COPD status, the sex-specific effect of COPD on systemic metabolism has yet to be interrogated. Significant sex differences were observed in 9 of the 11 modules identified in COPDGene. Sex-specific associations by COPD status and emphysema were observed in 3 modules for each phenotype. Sex stratified individual metabolite associations with COPD demonstrated male-specific associations in sphingomyelins and female-specific associations in acyl carnitines and phosphatidylethanolamines. There was high preservation of module assignments in SPIROMICS (SubPopulations and InteRmediate Outcome Measures In COPD Study) and similar female-specific shift in acyl carnitines. Several COPD associated metabolites differed by sex. Acyl carnitines and sphingomyelins demonstrate sex-specific abundances and may represent important metabolic signatures of sex differences in COPD. Accurately characterizing the sex-specific molecular differences in COPD is vital for personalized diagnostics and therapeutics.
Collapse
Affiliation(s)
- Lucas A. Gillenwater
- Computational Bioscience Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
| | - Katerina J. Kechris
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
| | - Katherine A. Pratte
- Division of Medicine, National Jewish Health, Denver, CO 80206, USA; (K.A.P.); (I.P.); (R.P.B.)
| | - Nichole Reisdorph
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
| | - Irina Petrache
- Division of Medicine, National Jewish Health, Denver, CO 80206, USA; (K.A.P.); (I.P.); (R.P.B.)
- School of Medicine, University of Colorado, Aurora, CO 80045, USA
| | - Wassim W. Labaki
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI 48109, USA;
| | - Wanda O’Neal
- Cystic Fibrosis/Pulmonary Research and Treatment Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jerry A. Krishnan
- Breathe Chicago Center, University of Illinois at Chicago, Chicago, IL 60608, USA;
| | - Victor E. Ortega
- Center for Precision Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA;
| | - Dawn L. DeMeo
- Channing Division of Network Medicine, and Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Russell P. Bowler
- Division of Medicine, National Jewish Health, Denver, CO 80206, USA; (K.A.P.); (I.P.); (R.P.B.)
- School of Medicine, University of Colorado, Aurora, CO 80045, USA
| |
Collapse
|
21
|
Chiang AJ, Thanabalasuriar A, Boo CC. Proteomics: An advanced tool to unravel the role of alveolar macrophages in respiratory diseases. Int J Biochem Cell Biol 2021; 134:105966. [PMID: 33677070 DOI: 10.1016/j.biocel.2021.105966] [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] [Received: 08/16/2020] [Revised: 02/05/2021] [Accepted: 03/03/2021] [Indexed: 02/08/2023]
Abstract
As we learn more about chronic lung diseases, we are seeing that an unbalanced immune system plays a key role in disease pathogenesis. Innate immune cells, particularly tissue-resident macrophages, are important navigators of immunity, both during infection and in non-communicable lung disease. In the lung, alveolar macrophages are considered some of the most critical and diverse immune cells, yet despite an array of studies over the years, alveolar macrophages remain poorly understood. In this review, we highlight the importance of alveolar macrophages in health and disease, and discuss how proteomics can be used to elucidate mechanistic information and identify potential targets for therapy development.
Collapse
Affiliation(s)
- Abby J Chiang
- Dynamic Omics, Antibody Discovery and Protein Engineering, R&D AstraZeneca, Gaithersburg, MD 20878, USA
| | | | - Chelsea C Boo
- Dynamic Omics, Antibody Discovery and Protein Engineering, R&D AstraZeneca, Gaithersburg, MD 20878, USA.
| |
Collapse
|
22
|
Merikallio H, Kaarteenaho R, Lindén S, Padra M, Karimi R, Li CX, Lappi-Blanco E, Wheelock ÅM, Sköld MC. Smoking-associated increase in mucins 1 and 4 in human airways. Respir Res 2020; 21:239. [PMID: 32948202 PMCID: PMC7499856 DOI: 10.1186/s12931-020-01498-7] [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: 10/29/2019] [Accepted: 08/31/2020] [Indexed: 12/18/2022] Open
Abstract
Rationale Smoking-related chronic obstructive pulmonary disease (COPD) is associated with dysregulated production of mucus. Mucins (MUC) are important both for mucus secretion and epithelial defense. We have examined the distribution of MUC1 and MUC4 in the airway epithelial cells of never-smokers and smokers with and without COPD. Methods Mucosal biopsies and bronchial wash samples were obtained by bronchoscopy from age- and sex-matched COPD-patients (n = 38; GOLD I-II/A-B), healthy never-smokers (n = 40) and current smokers with normal lung function (n = 40) from the Karolinska COSMIC cohort (NCT02627872). Cell-specific expressions of MUC1, MUC4 and regulating factors, i.e., epithelial growth factor receptor (EGFR) 1 and 2, were analyzed by immunohistochemistry. Soluble MUC1 was measured by quantitative immunodetection on slot blot. Results The levels of cell-bound MUC1 expression in basal cells and in soluble MUC1 in bronchial wash were increased in smokers, regardless of airway obstruction. Patients with chronic bronchitis had higher MUC1 expression. The expression of MUC4 in cells with goblet cell phenotype was increased in smokers. The expression of EGFR2, but not that of EGFR1, was higher in never-smokers than in smokers. Conclusions Smoking history and the presence of chronic bronchitis, regardless of airway obstruction, affect both cellular and soluble MUC1 in human airways. Therefore, MUC1 may be a novel marker for smoking- associated airway disease.
Collapse
Affiliation(s)
- Heta Merikallio
- Respiratory Medicine Unit, Department of Medicine Solna and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden. .,Research Unit of Internal Medicine, University of Oulu, Oulu, Finland. .,Medical Research Center Oulu, Oulu University Hospital, Oulu, Finland.
| | - Riitta Kaarteenaho
- Research Unit of Internal Medicine, University of Oulu, Oulu, Finland.,Medical Research Center Oulu, Oulu University Hospital, Oulu, Finland
| | - Sara Lindén
- Department of Biomedical Chemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Médea Padra
- Department of Biomedical Chemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Reza Karimi
- Respiratory Medicine Unit, Department of Medicine Solna and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Respiratory Medicine and Allergy, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Chuan-Xing Li
- Respiratory Medicine Unit, Department of Medicine Solna and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Elisa Lappi-Blanco
- Department of Pathology, Center for Cancer Research and Translational Medicine, Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Åsa M Wheelock
- Respiratory Medicine Unit, Department of Medicine Solna and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Magnus C Sköld
- Respiratory Medicine Unit, Department of Medicine Solna and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Respiratory Medicine and Allergy, Karolinska University Hospital Solna, Stockholm, Sweden
| |
Collapse
|
23
|
Brandsma C, Van den Berge M, Hackett T, Brusselle G, Timens W. Recent advances in chronic obstructive pulmonary disease pathogenesis: from disease mechanisms to precision medicine. J Pathol 2020; 250:624-635. [PMID: 31691283 PMCID: PMC7216938 DOI: 10.1002/path.5364] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 10/18/2019] [Accepted: 11/01/2019] [Indexed: 12/22/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a devastating lung disease with a high personal and societal burden. Exposure to toxic particles and gases, including cigarette smoke, is the main risk factor for COPD. Together with smoking cessation, current treatment strategies of COPD aim to improve symptoms and prevent exacerbations, but there is no disease-modifying treatment. The biggest drawback of today's COPD treatment regimen is the 'one size fits all' pharmacological intervention, mainly based on disease severity and symptoms and not the individual's disease pathology. To halt the worrying increase in the burden of COPD, disease management needs to be advanced with a focus on personalized treatment. The main pathological feature of COPD includes a chronic and abnormal inflammatory response within the lungs, which results in airway and alveolar changes in the lung as reflected by (small) airways disease and emphysema. Here we discuss recent developments related to the abnormal inflammatory response, ECM and age-related changes, structural changes in the small airways and the role of sex-related differences, which are all relevant to explain the individual differences in the disease pathology of COPD and improve disease endotyping. Furthermore, we will discuss the most recent developments of new treatment strategies using biologicals to target specific pathological features or disease endotypes of COPD. © 2020 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Corry‐Anke Brandsma
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical BiologyGroningenThe Netherlands
- University of Groningen, University Medical Center GroningenGroningen Research Institute for Asthma and COPD (GRIAC)GroningenThe Netherlands
| | - Maarten Van den Berge
- University of Groningen, University Medical Center GroningenGroningen Research Institute for Asthma and COPD (GRIAC)GroningenThe Netherlands
- University of Groningen, University Medical Center Groningen, Department of Pulmonary DiseasesGroningenThe Netherlands
| | - Tillie‐Louise Hackett
- Centre for Heart Lung InnovationUnive rsity of British ColumbiaVancouverCanada
- Department of Anesthesiology, Pharmacology and TherapeuticsUniversity of British ColumbiaVancouverCanada
| | - Guy Brusselle
- Department of Respiratory MedicineGhent University HospitalGhentBelgium
- Department of Epidemiology and Respiratory MedicineErasmus Medical Center RotterdamRotterdamThe Netherlands
| | - Wim Timens
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical BiologyGroningenThe Netherlands
- University of Groningen, University Medical Center GroningenGroningen Research Institute for Asthma and COPD (GRIAC)GroningenThe Netherlands
| |
Collapse
|
24
|
Han MK. Chronic Obstructive Pulmonary Disease in Women: A Biologically Focused Review with a Systematic Search Strategy. Int J Chron Obstruct Pulmon Dis 2020; 15:711-721. [PMID: 32280209 PMCID: PMC7132005 DOI: 10.2147/copd.s237228] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 03/10/2020] [Indexed: 01/06/2023] Open
Abstract
Purpose Evidence suggests that chronic obstructive pulmonary disease (COPD) symptoms and progression may differ between men and women. However, limited information is currently available on the pathophysiological and biological factors that may underlie these sex-related differences. The objective of this review is to systematically evaluate reports of potential sex-related differences, including genetic, pathophysiological, structural, and other biological factors, that may influence COPD development, manifestation, and progression in women. Patients and Methods A PubMed literature search was conducted from inception until January 2020. Original reports of genetic, hormonal, and physiological differences, and biological influences that could contribute to COPD development, manifestation, and progression in women were included. Results Overall, 491 articles were screened; 29 articles met the inclusion criteria. Results from this analysis demonstrated between-sex differences in inflammatory, immune, genetic, structural, and physiological factors in patients with COPD. Conclusion Various biological differences are observed between men and women with COPD including differences in inflammatory and metabolic pathways related to obesity and fat distribution, immune cell function and autophagy, extent and distribution of emphysema and airway wall remodeling. An enhanced understanding of these differences has the potential to broaden our understanding of how COPD develops and progresses, thereby providing an opportunity to ultimately improve diagnosis, treatment, and monitoring of COPD in both men and women.
Collapse
Affiliation(s)
- MeiLan K Han
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
25
|
Zhang WZ, Rice MC, Hoffman KL, Oromendia C, Barjaktarevic IZ, Wells JM, Hastie AT, Labaki WW, Cooper CB, Comellas AP, Criner GJ, Krishnan JA, Paine R, Hansel NN, Bowler RP, Barr RG, Peters SP, Woodruff PG, Curtis JL, Han MK, Ballman KV, Martinez FJ, Choi AM, Nakahira K, Cloonan SM, Choi ME. Association of urine mitochondrial DNA with clinical measures of COPD in the SPIROMICS cohort. JCI Insight 2020; 5:133984. [PMID: 31895696 DOI: 10.1172/jci.insight.133984] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 12/26/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUNDMitochondrial dysfunction, a proposed mechanism of chronic obstructive pulmonary disease (COPD) pathogenesis, is associated with the leakage of mitochondrial DNA (mtDNA), which may be detected extracellularly in various bodily fluids. Despite evidence for the increased prevalence of chronic kidney disease in COPD subjects and for mitochondrial dysfunction in the kidneys of murine COPD models, whether urine mtDNA (u-mtDNA) associates with measures of disease severity in COPD is unknown.METHODSCell-free u-mtDNA, defined as copy number of mitochondrially encoded NADH dehydrogenase-1 (MTND1) gene, was measured by quantitative PCR and normalized to urine creatinine in cell-free urine samples from participants in the Subpopulations and Intermediate Outcome Measures in COPD Study (SPIROMICS) cohort. Urine albumin/creatinine ratios (UACR) were measured in the same samples. Associations between u-mtDNA, UACR, and clinical disease parameters - including FEV1 % predicted, clinical measures of exercise tolerance, respiratory symptom burden, and chest CT measures of lung structure - were examined.RESULTSU-mtDNA and UACR levels were measured in never smokers (n = 64), smokers without airflow obstruction (n = 109), participants with mild/moderate COPD (n = 142), and participants with severe COPD (n = 168). U-mtDNA was associated with increased respiratory symptom burden, especially among smokers without COPD. Significant sex differences in u-mtDNA levels were observed, with females having higher u-mtDNA levels across all study subgroups. U-mtDNA associated with worse spirometry and CT emphysema in males only and with worse respiratory symptoms in females only. Similar associations were not found with UACR.CONCLUSIONU-mtDNA levels may help to identify distinct clinical phenotypes and underlying pathobiological differences in males versus females with COPD.TRIAL REGISTRATIONThis study has been registered at ClinicalTrials.gov ( NCT01969344).FUNDINGUS NIH, National Heart, Lung and Blood Institute, supplemented by contributions made through the Foundation for the NIH and the COPD Foundation from AstraZeneca/MedImmune, Bayer, Bellerophon Therapeutics, Boehringer-Ingelheim Pharmaceuticals Inc., Chiesi Farmaceutici S.p.A., Forest Research Institute Inc., GlaxoSmithKline, Grifols Therapeutics Inc., Ikaria Inc., Novartis Pharmaceuticals Corporation, Nycomed GmbH, ProterixBio, Regeneron Pharmaceuticals Inc., Sanofi, Sunovion, Takeda Pharmaceutical Company, and Theravance Biopharma and Mylan.
Collapse
Affiliation(s)
- William Z Zhang
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, New York, New York, USA.,New York-Presbyterian Hospital, Weill Cornell Medicine, New York, New York, USA
| | - Michelle C Rice
- Division of Nephrology and Hypertension, Joan and Sanford I. Weill Department of Medicine, and
| | - Katherine L Hoffman
- Department of Healthcare Policy and Research, Division of Biostatistics and Epidemiology, Weill Cornell Medicine, New York, New York, USA
| | - Clara Oromendia
- Department of Healthcare Policy and Research, Division of Biostatistics and Epidemiology, Weill Cornell Medicine, New York, New York, USA
| | - Igor Z Barjaktarevic
- Division of Pulmonary and Critical Care Medicine, UCLA Medical Center, Los Angeles, California, USA
| | - J Michael Wells
- University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Annette T Hastie
- Pulmonary, Critical Care, Allergy, and Immunologic Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Wassim W Labaki
- Division of Pulmonary and Critical Care Medicine, University of Michigan Health System, Ann Arbor, Michigan, USA
| | - Christopher B Cooper
- Division of Pulmonary and Critical Care Medicine, UCLA Medical Center, Los Angeles, California, USA
| | - Alejandro P Comellas
- Division of Pulmonary and Critical Care, University of Iowa, Iowa City, Iowa, USA
| | - Gerard J Criner
- Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| | - Jerry A Krishnan
- Division of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Robert Paine
- Division of Respiratory, Critical Care, and Occupational Pulmonary Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Nadia N Hansel
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Russell P Bowler
- Division of Pulmonary, Critical Care Medicine, National Jewish Health, Denver, Colorado, USA
| | - R Graham Barr
- Columbia University Medical Center, New York, New York, USA
| | - Stephen P Peters
- Pulmonary, Critical Care, Allergy, and Immunologic Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Prescott G Woodruff
- Division of Pulmonary and Critical Care Medicine, UCSF, School of Medicine, San Francisco, California, USA
| | - Jeffrey L Curtis
- Division of Pulmonary and Critical Care Medicine, University of Michigan Health System, Ann Arbor, Michigan, USA.,Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, Michigan, USA
| | - Meilan K Han
- Division of Pulmonary and Critical Care Medicine, University of Michigan Health System, Ann Arbor, Michigan, USA
| | - Karla V Ballman
- Department of Healthcare Policy and Research, Division of Biostatistics and Epidemiology, Weill Cornell Medicine, New York, New York, USA
| | - Fernando J Martinez
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, New York, New York, USA.,New York-Presbyterian Hospital, Weill Cornell Medicine, New York, New York, USA
| | - Augustine Mk Choi
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, New York, New York, USA.,New York-Presbyterian Hospital, Weill Cornell Medicine, New York, New York, USA
| | - Kiichi Nakahira
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, New York, New York, USA
| | - Suzanne M Cloonan
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, New York, New York, USA
| | - Mary E Choi
- New York-Presbyterian Hospital, Weill Cornell Medicine, New York, New York, USA.,Division of Nephrology and Hypertension, Joan and Sanford I. Weill Department of Medicine, and
| | | |
Collapse
|
26
|
Tam A, Tanabe N, Churg A, Wright JL, Hogg JC, Sin DD. Sex differences in lymphoid follicles in COPD airways. Respir Res 2020; 21:46. [PMID: 32033623 PMCID: PMC7006095 DOI: 10.1186/s12931-020-1311-8] [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: 10/17/2019] [Accepted: 01/30/2020] [Indexed: 01/20/2023] Open
Abstract
Background Female smokers have increased risk for chronic obstructive pulmonary disease (COPD) compared with male smokers who have a similar history of cigarette smoke exposure. Tertiary lymphoid follicles are often found in the lungs of patients with severe COPD but sex-related differences have not been previously investigated. We determined the impact of female sex hormones on chronic cigarette smoke-induced expression of lymphoid aggregates in mice with COPD-like pathologies. Methods Lymphoid aggregate counts, total aggregate cross-sectional area and foamy macrophage counts were determined morphometrically in male, female, and ovariectomized mice exposed to air or cigarette smoke for 6 months. B-cell activating factor (BAFF) protein expression and markers of oxidative stress were evaluated in mouse lung tissues by immunofluorescence staining and gene expression analyses. Quantitative histology was performed on lung tissue sections of human COPD lungs to evaluate follicle formation. Results Lymphoid follicle and foamy macrophage counts as well as the total follicle cross-sectional area were differentially increased in lung tissues of female mice compared to male mice, and these differences were abolished by ovariectomy. These lymphoid aggregates were positive for CD45, CD20, CD21 and BAFF expression. Differential increases in Mmp12 and Cxcl2 gene expression correlated with an increase in foamy macrophages in parenchymal tissues of female but not male mice after smoke exposure. Parenchymal tissues from female mice failed to induce antioxidant-related genes in response to smoke exposure, and this effect was restored by ovariectomy. 3-nitrotyrosine, a stable marker of oxidative stress, positively correlated with Mmp12 and Cxcl2 gene expression. Hydrogen peroxide induced BAFF protein in mouse macrophage cell line. In human lung tissues, female smokers with severe COPD demonstrated increased numbers of lymphoid follicles compared with males. Conclusions Chronic smoke exposure increases the risk of lymphoid aggregate formation in female mice compared with male mice, which is mediated female sex hormones and BAFF expression in an oxidative environment.
Collapse
Affiliation(s)
- Anthony Tam
- Centre for Heart Lung Innovation, St. Paul's Hospital, & Department of Medicine, Vancouver, British Columbia, Canada
| | - Naoya Tanabe
- Centre for Heart Lung Innovation, St. Paul's Hospital, & Department of Medicine, Vancouver, British Columbia, Canada.,Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Andrew Churg
- Department of Pathology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Joanne L Wright
- Department of Pathology, University of British Columbia, Vancouver, British Columbia, Canada
| | - James C Hogg
- Centre for Heart Lung Innovation, St. Paul's Hospital, & Department of Medicine, Vancouver, British Columbia, Canada
| | - Don D Sin
- Centre for Heart Lung Innovation, St. Paul's Hospital, & Department of Medicine, Vancouver, British Columbia, Canada.
| |
Collapse
|
27
|
Ray JL, Fletcher P, Burmeister R, Holian A. The role of sex in particle-induced inflammation and injury. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2019; 12:e1589. [PMID: 31566915 DOI: 10.1002/wnan.1589] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 08/19/2019] [Accepted: 08/28/2019] [Indexed: 12/17/2022]
Abstract
The use of engineered nanomaterials within various applications such as medicine, electronics, and cosmetics has been steadily increasing; therefore, the rate of occupational and environmental exposures has also increased. Inhalation is an important route of exposure to nanomaterials and has been shown to cause various respiratory diseases in animal models. Human lung disease frequently presents with a sex/gender-bias in prevalence or severity, but investigation of potential sex-differences in the adverse health outcomes associated with nanoparticle inhalation is greatly lacking. Only ~20% of basic research in the general sciences use both male and female animals and a substantial percentage of these do not address differences between sexes within their analyses. This has prevented researchers from fully understanding the impact of sex-based variables on health and disease, particularly the pathologies resulting from the inhalation of particles. The mechanisms responsible for sex-differences in respiratory disease remain unclear, but could be related to a number of variables including sex-differences in hormone signaling, lung physiology, or respiratory immune function. By incorporating sex-based analysis into respiratory nanotoxicology and utilizing human data from other relevant particles (e.g., asbestos, silica, particulate matter), we can improve our understanding of sex as a biological variable in nanoparticle exposures. This article is categorized under: Toxicology and Regulatory Issues in Nanomedicine > Toxicology of Nanomaterials.
Collapse
Affiliation(s)
- Jessica L Ray
- Center for Environmental Health Sciences, Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, Montana
| | - Paige Fletcher
- Center for Environmental Health Sciences, Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, Montana
| | - Rachel Burmeister
- Center for Environmental Health Sciences, Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, Montana
| | - Andrij Holian
- Center for Environmental Health Sciences, Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, Montana
| |
Collapse
|
28
|
Raherison-Semjen C. [Vulnerability of women to tobacco: The broncho-pulmonary consequences (asthma, COPD)]. Rev Mal Respir 2019; 36:1032-1037. [PMID: 31540739 DOI: 10.1016/j.rmr.2019.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 11/20/2018] [Indexed: 11/30/2022]
Abstract
Smoking remains common, with an exposure that begins early during pregnancy. It induces epigenetic changes, with a trans-generational transmission. Smoking increases the risk of uncontrolled asthma during childhood and adult life. Asthma is also associated with increased risk of a decline of lung function and chronic obstructive pulmonary disease (COPD). Women are more at risk of developing early and severe COPD. The mechanisms are currently poorly known.
Collapse
Affiliation(s)
- C Raherison-Semjen
- Inserm, pôle cardiothoracique, service des maladies respiratoires, U1219 BPH Bordeaux Population Health Research Center, université de Bordeaux, CHU de Bordeaux, avenue Magellan, 33604 Pessac, France.
| |
Collapse
|
29
|
Gut-Gobert C, Cavaillès A, Dixmier A, Guillot S, Jouneau S, Leroyer C, Marchand-Adam S, Marquette D, Meurice JC, Desvigne N, Morel H, Person-Tacnet C, Raherison C. Women and COPD: do we need more evidence? Eur Respir Rev 2019; 28:28/151/180055. [PMID: 30814138 PMCID: PMC9488562 DOI: 10.1183/16000617.0055-2018] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 09/21/2018] [Indexed: 01/20/2023] Open
Abstract
The increasingly female face of chronic obstructive pulmonary disease (COPD) prevalence among women has equalled that of men since 2008, due in part to increased tobacco use among women worldwide and exposure to biomass fuels. This finding is supported by a number of characteristics. There is evidence of susceptibility to smoking and other airborne contaminants, along with epidemiological and phenotypic manifestations. COPD has thus become the leading cause of death in women in the USA. The clinical presentation is characterised by increasingly pronounced dyspnoea with a marked tendency towards anxiety and depression, undernutrition, nonsmall cell lung cancer (especially adenocarcinoma) and osteoporosis. Quality of life is also more significantly impacted. The theories advanced to explain these differences involve the role played by oestrogens, impaired gas exchange in the lungs and smoking habits. While these differences require appropriate therapeutic responses (smoking cessation, pulmonary rehabilitation, long-term oxygen therapy), barriers to the treatment of women with COPD include greater under-diagnosis than in men, fewer spirometry tests and medical consultations. Faced with this serious public health problem, we need to update and adapt our knowledge to the epidemiological changes. The face of COPD is increasingly female. We need more evidence and a change in how the disease is managed. http://ow.ly/zueL30mWqlS
Collapse
Affiliation(s)
- Christophe Gut-Gobert
- G.E.T.B.O. (Groupe d'Etude de la Thrombose de Bretagne Occidentale), Université Européenne de Bretagne, Université de Brest, EA3878, IFR148, Hôpital La Cavale Blanche, Département de Médecine Interne et Pneumologie, Brest, France
| | - Arnaud Cavaillès
- Institut du Thorax, CHU de Nantes, Dept of Pulmonology, Nantes, France
| | - Adrien Dixmier
- Dept of Pulmonology, Orléans Regional Hospital, Orléans, France
| | - Stéphanie Guillot
- Unité d'Explorations Fonctionnelles Respiratoires, CHRU Rennes, Rennes, France
| | - Stéphane Jouneau
- Service de Pneumologie, Hôpital Pontchaillou, Rennes, France.,IRSET UMR 1085, Université de Rennes 1, Rennes, France
| | - Christophe Leroyer
- G.E.T.B.O. (Groupe d'Etude de la Thrombose de Bretagne Occidentale), Université Européenne de Bretagne, Université de Brest, EA3878, IFR148, Hôpital La Cavale Blanche, Département de Médecine Interne et Pneumologie, Brest, France
| | - Sylvain Marchand-Adam
- Université François Rabelais Faculté de Médecine de Tours, Inserm 1100, CHRU de Tours Service de Pneumologie, Tours, France
| | - David Marquette
- Dept of Pulmonary Medicine, Centre Hospitalier Bretagne Atlantique, Vannes, France
| | - Jean-Claude Meurice
- Dept of Pulmonology Centre Hospitalier de l'Université de Poitiers, Poitiers, France
| | | | - Hugues Morel
- Dept of Pulmonology, Orléans Regional Hospital, Orléans, France
| | | | - Chantal Raherison
- Service des Maladies Respiratoires, CHU Bordeaux, Epicene U1219 Université de Bordeaux, Bordeaux, France
| |
Collapse
|
30
|
Ward LJ, Olausson P, Li W, Yuan XM. Proteomics and multivariate modelling reveal sex-specific alterations in distinct regions of human carotid atheroma. Biol Sex Differ 2018; 9:54. [PMID: 30594242 PMCID: PMC6311011 DOI: 10.1186/s13293-018-0217-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 12/12/2018] [Indexed: 01/05/2023] Open
Abstract
Background Atherosclerotic lesions are comprised of distinct regions with different proteomic profiles. Men and women develop differences in lesion phenotype, with lesions from women generally being more stable and less prone to rupture. We aimed to investigate the differences in proteomic profiles between sexes, including distinct lesion regions, to identify altered proteins that contribute to these differences observed clinically. Methods Carotid endarterectomy samples (ten men/ten women) were obtained, and intraplaque biopsies from three distinct regions (internal control, fatty streak and plaque) were analysed by tandem-mass spectrometry. Multivariate statistical modelling, using orthogonal partial least square-discriminant analysis, was used to discriminate the proteomes between men and women. Results Multivariate discriminant modelling revealed proteins from 16 functional groups that displayed sex-specific associations. Additional statistics revealed ten proteins that display region-specific alterations when comparing sexes, including proteins related to inflammatory response, response to reactive oxygen species, complement activation, transport and blood coagulation. Transport protein afamin and blood coagulation proteins antithrombin-III and coagulation factor XII were significantly increased in plaque region from women. Inflammatory response proteins lysozyme C and phospholipase A2 membrane-associated were significantly increased in plaque region from men. Limitations with this study are the small sample size, limited patient information and lack of complementary histology to control for cell type differences between sexes. Conclusions This pilot study, for the first time, utilises a multivariate proteomic approach to investigate sexual dimorphism in human atherosclerotic tissue, and provides an essential proteomic platform for further investigations to help understand sexual dimorphism and plaque vulnerability in atherosclerosis. Electronic supplementary material The online version of this article (10.1186/s13293-018-0217-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Liam J Ward
- Obstetrics and Gynaecology, Department of Clinical and Experimental Medicine, Linköping University, SE-581 85, Linköping, Sweden. .,Occupational and Environmental Medicine, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden.
| | - Patrik Olausson
- Pain and Rehabilitation Centre, Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Wei Li
- Obstetrics and Gynaecology, Department of Clinical and Experimental Medicine, Linköping University, SE-581 85, Linköping, Sweden
| | - Xi-Ming Yuan
- Occupational and Environmental Medicine, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| |
Collapse
|
31
|
Cagnone M, Salvini R, Bardoni A, Fumagalli M, Iadarola P, Viglio S. Searching for biomarkers of chronic obstructive pulmonary disease using proteomics: The current state. Electrophoresis 2018; 40:151-164. [PMID: 30216498 DOI: 10.1002/elps.201800305] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 09/10/2018] [Accepted: 09/10/2018] [Indexed: 12/24/2022]
Abstract
Detection of proteins which may be potential biomarkers of disorders represents a big step forward in understanding the molecular mechanisms that underlie pathological processes. In this context proteomics plays the important role of opening a path for the identification of molecular signatures that can potentially assist in early diagnosis of several clinical disturbances. Aim of this report is to provide an overview of the wide variety of proteomic strategies that have been applied to the investigation of chronic obstructive pulmonary disease (COPD), a severe disorder that causes an irreversible damage to the lungs and for which there is no cure yet. The results in this area published over the past decade show that proteomics indeed has the ability of monitoring alterations in expression profiles of proteins from fluids/tissues of patients affected by COPD and healthy controls. However, these data also suggest that proteomics, while being an attractive tool for the identification of novel pathological mediators of COPD, remains a technique mainly generated and developed in research laboratories. Great efforts dedicated to the validation of these biological signatures will result in the proof of their clinical utility.
Collapse
Affiliation(s)
- Maddalena Cagnone
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Italy
| | - Roberta Salvini
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Italy
| | - Anna Bardoni
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Italy
| | - Marco Fumagalli
- Department of Biology and Biotechnologies "L.Spallanzani", Biochemistry Unit, University of Pavia, Italy
| | - Paolo Iadarola
- Department of Biology and Biotechnologies "L.Spallanzani", Biochemistry Unit, University of Pavia, Italy
| | - Simona Viglio
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Italy
| |
Collapse
|
32
|
The neutrophil-mobilizing cytokine interleukin-26 in the airways of long-term tobacco smokers. Clin Sci (Lond) 2018; 132:959-983. [PMID: 29780024 PMCID: PMC6365630 DOI: 10.1042/cs20180057] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 03/26/2018] [Accepted: 04/03/2018] [Indexed: 02/08/2023]
Abstract
Long-term tobacco smokers with chronic obstructive pulmonary disease (COPD) or chronic bronchitis display an excessive accumulation of neutrophils in the airways; an inflammation that responds poorly to established therapy. Thus, there is a need to identify new molecular targets for the development of effective therapy. Here, we hypothesized that the neutrophil-mobilizing cytokine interleukin (IL)-26 (IL-26) is involved in airway inflammation amongst long-term tobacco smokers with or without COPD, chronic bronchitis or colonization by pathogenic bacteria. By analyzing bronchoalveolar lavage (BAL), bronchail wash (BW) and induced sputum (IS) samples, we found increased extracellular IL-26 protein in the airways of long-term smokers in vivo without further increase amongst those with clinically stable COPD. In human alveolar macrophages (AM) in vitro, the exposure to water-soluble tobacco smoke components (WTC) enhanced IL-26 gene and protein. In this cell model, the same exposure increased gene expression of the IL-26 receptor complex (IL10R2 and IL20R1) and nuclear factor κ B (NF-κB); a proven regulator of IL-26 production. In the same cell model, recombinant human IL-26 in vitro caused a concentration-dependent increase in the gene expression of NF-κB and several pro-inflammatory cytokines. In the long-term smokers, we also observed that extracellular IL-26 protein in BAL samples correlates with measures of lung function, tobacco load, and several markers of neutrophil accumulation. Extracellular IL-26 was further increased in long-term smokers with exacerbations of COPD (IS samples), with chronic bronchitis (BAL samples ) or with colonization by pathogenic bacteria (IS and BW samples). Thus, IL-26 in the airways emerges as a promising target for improving the understanding of the pathogenic mechanisms behind several pulmonary morbidities in long-term tobacco smokers.
Collapse
|
33
|
Li CX, Wheelock CE, Sköld CM, Wheelock ÅM. Integration of multi-omics datasets enables molecular classification of COPD. Eur Respir J 2018; 51:13993003.01930-2017. [PMID: 29545283 DOI: 10.1183/13993003.01930-2017] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 03/08/2018] [Indexed: 01/06/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is an umbrella diagnosis caused by a multitude of underlying mechanisms, and molecular sub-phenotyping is needed to develop molecular diagnostic/prognostic tools and efficacious treatments.The objective of these studies was to investigate whether multi-omics integration improves the accuracy of molecular classification of COPD in small cohorts.Nine omics data blocks (comprising mRNA, micro RNA, proteomes and metabolomes) collected from several anatomical locations from 52 female subjects were integrated by similarity network fusion (SNF). Multi-omics integration significantly improved the accuracy of group classification of COPD patients from healthy never-smokers and from smokers with normal spirometry, reducing required group sizes from n=30 to n=6 at 95% power. Seven different combinations of four to seven omics platforms achieved >95% accuracy.For the first time, a quantitative relationship between multi-omics data integration and accuracy of data-driven classification power has been demonstrated across nine omics data blocks. Integrating five to seven omics data blocks enabled 100% correct classification of COPD diagnosis with groups as small as n=6 individuals, despite strong confounding effects of current smoking. These results can serve as guidelines for the design of future systems-based multi-omics investigations, with indications that integrating five to six data blocks from several molecular levels and anatomical locations suffices to facilitate unsupervised molecular classification in small cohorts.
Collapse
Affiliation(s)
- Chuan-Xing Li
- Respiratory Medicine Unit, Dept of Medicine and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Craig E Wheelock
- Integrative Molecular Phenotyping Laboratory, Division of Physiological Chemistry II, Dept of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - C Magnus Sköld
- Respiratory Medicine Unit, Dept of Medicine and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Lung-Allergy Clinic, Karolinska University Hospital, Stockholm, Sweden
| | - Åsa M Wheelock
- Respiratory Medicine Unit, Dept of Medicine and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
34
|
Long-term smoking alters abundance of over half of the proteome in bronchoalveolar lavage cell in smokers with normal spirometry, with effects on molecular pathways associated with COPD. Respir Res 2018. [PMID: 29514648 PMCID: PMC5842534 DOI: 10.1186/s12931-017-0695-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background Smoking represents a significant risk factor for many chronic inflammatory diseases, including chronic obstructive pulmonary disease (COPD). Methods To identify dysregulation of specific proteins and pathways in bronchoalveolar lavage (BAL) cells associated with smoking, isobaric tags for relative and absolute quantitation (iTRAQ)-based shotgun proteomics analyses were performed on BAL cells from healthy never-smokers and smokers with normal lung function from the Karolinska COSMIC cohort. Multivariate statistical modeling, multivariate correlations with clinical data, and pathway enrichment analysis were performed. Results Smoking exerted a significant impact on the BAL cell proteome, with more than 500 proteins representing 15 molecular pathways altered due to smoking. The majority of these alterations occurred in a gender-independent manner. The phagosomal- and leukocyte trans endothelial migration (LTM) pathways significantly correlated with FEV1/FVC as well as the percentage of CD8+ T-cells and CD8+CD69+ T-cells in smokers. The correlations to clinical parameters in healthy never-smokers were minor. Conclusion The significant correlations of proteins in the phagosome- and LTM pathways with activated cytotoxic T-cells (CD69+) and the level of airway obstruction (FEV1/FVC) in smokers, both hallmarks of COPD, suggests that these two pathways may play a role in the molecular events preceding the development of COPD in susceptible smokers. Both pathways were found to be further dysregulated in COPD patients from the same cohort, thereby providing further support to this hypothesis. Given that not all smokers develop COPD in spite of decades of smoking, it is also plausible that some of the molecular pathways associated with response to smoking exert protective mechanisms to smoking-related pathologies in resilient individuals. Trial registration ClinicalTrials.gov identifier NCT02627872; Retrospectively registered on December 9, 2015. Electronic supplementary material The online version of this article (10.1186/s12931-017-0695-6) contains supplementary material, which is available to authorized users.
Collapse
|
35
|
Yang M, Kohler M, Heyder T, Forsslund H, Garberg HK, Karimi R, Grunewald J, Berven FS, Nyrén S, Magnus Sköld C, Wheelock ÅM. Proteomic profiling of lung immune cells reveals dysregulation of phagocytotic pathways in female-dominated molecular COPD phenotype. Respir Res 2018. [PMID: 29514663 PMCID: PMC5842633 DOI: 10.1186/s12931-017-0699-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Background Smoking is the main risk factor for chronic obstructive pulmonary disease (COPD). Women with COPD who smoke experienced a higher risk of hospitalization and worse decline of lung function. Yet the mechanisms of these gender-related differences in clinical presentations in COPD remain unknown. The aim of our study is to identify proteins and molecular pathways associated with COPD pathogenesis, with emphasis on elucidating molecular gender difference. Method We employed shotgun isobaric tags for relative and absolute quantitation (iTRAQ) proteome analyses of bronchoalveolar lavage (BAL) cells from smokers with normal lung function (n = 25) and early stage COPD patients (n = 18). Multivariate modeling, pathway enrichment analysis, and correlation with clinical characteristics were performed to identify specific proteins and pathways of interest. Results More pronounced alterations both at the protein- and pathway- levels were observed in female COPD patients, involving dysregulation of the FcγR-mediated phagocytosis-lysosomal axis and increase in oxidative stress. Alterations in pathways of the phagocytosis-lysosomal axis associated with a female-dominated COPD phenotype correlated well with specific clinical features: FcγR-mediated phagocytosis correlated with FEV1/FVC, the lysosomal pathway correlated with CT < −950 Hounsfield Units (HU), and regulation of actin cytoskeleton correlated with FEV1 and FEV1/FVC in female COPD patients. Alterations observed in the corresponding male cohort were minor. Conclusion The identified molecular pathways suggest dysregulation of several phagocytosis-related pathways in BAL cells in female COPD patients, with correlation to both the level of obstruction (FEV1/FVC) and disease severity (FEV1) as well as emphysema (CT < −950 HU) in women. Trial registration No.: NCT02627872, retrospectively registered on December 9, 2015. Electronic supplementary material The online version of this article (10.1186/s12931-017-0699-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mingxing Yang
- Respiratory Medicine Unit, Department of Medicine Solna & Center for Molecular Medicine, Karolinska Institutet, Lung Research Lab L4:01, SE-171 76, Stockholm, Sweden.
| | - Maxie Kohler
- Respiratory Medicine Unit, Department of Medicine Solna & Center for Molecular Medicine, Karolinska Institutet, Lung Research Lab L4:01, SE-171 76, Stockholm, Sweden
| | - Tina Heyder
- Respiratory Medicine Unit, Department of Medicine Solna & Center for Molecular Medicine, Karolinska Institutet, Lung Research Lab L4:01, SE-171 76, Stockholm, Sweden
| | - Helena Forsslund
- Respiratory Medicine Unit, Department of Medicine Solna & Center for Molecular Medicine, Karolinska Institutet, Lung Research Lab L4:01, SE-171 76, Stockholm, Sweden
| | - Hilde K Garberg
- Proteomics Unit (PROBE), Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Reza Karimi
- Respiratory Medicine Unit, Department of Medicine Solna & Center for Molecular Medicine, Karolinska Institutet, Lung Research Lab L4:01, SE-171 76, Stockholm, Sweden
| | - Johan Grunewald
- Respiratory Medicine Unit, Department of Medicine Solna & Center for Molecular Medicine, Karolinska Institutet, Lung Research Lab L4:01, SE-171 76, Stockholm, Sweden
| | - Frode S Berven
- Proteomics Unit (PROBE), Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Sven Nyrén
- Department of Molecular Medicine and Surgery, Division of Radiology, Karolinska Institutet, Karolinska University Hospital, Solna, Stockholm, Sweden
| | - C Magnus Sköld
- Respiratory Medicine Unit, Department of Medicine Solna & Center for Molecular Medicine, Karolinska Institutet, Lung Research Lab L4:01, SE-171 76, Stockholm, Sweden
| | - Åsa M Wheelock
- Respiratory Medicine Unit, Department of Medicine Solna & Center for Molecular Medicine, Karolinska Institutet, Lung Research Lab L4:01, SE-171 76, Stockholm, Sweden.
| |
Collapse
|
36
|
Che KF, Kaarteenaho R, Lappi-Blanco E, Levänen B, Sun J, Wheelock Å, Palmberg L, Sköld CM, Lindén A. Interleukin-26 Production in Human Primary Bronchial Epithelial Cells in Response to Viral Stimulation: Modulation by Th17 cytokines. Mol Med 2017; 23:247-257. [PMID: 28853490 DOI: 10.2119/molmed.2016.00064] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 08/21/2017] [Indexed: 12/22/2022] Open
Abstract
Interleukin (IL)-26 is abundant in human airways and this cytokine is involved in the local immune response to a bacterial stimulus in vivo. Specifically, local exposure to the toll-like receptor (TLR) 4 agonist endotoxin does increase IL-26 in human airways and this cytokine potentiates chemotactic responses in human neutrophils. In addition to T-helper (Th) 17 cells, alveolar macrophages can produce IL-26, but it remains unknown whether this cytokine can also be produced in the airway mucosa per se in response to a viral stimulus. Here, we evaluated whether this is the case using primary bronchial epithelial cells from the airway epithelium in vitro, and exploring the signaling mechanisms involved, including the modulatory effects of additional Th17 cytokines. Finally, we assessed IL-26 and its archetype signaling responses in healthy human airways in vivo. We found increased transcription and release of IL-26 protein after stimulation with the viral-related double stranded (ds) RNA polyinosinic-polycytidylic acid (poly-IC) and showed that this IL-26 release involved mitogen-activated protein (MAP) kinases and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). The release of IL-26 in response to a viral stimulus was modulated by additional Th17 cytokines. Moreover, there was transcription of IL26 mRNA and expression of the protein in epithelial cells of bronchial brush and tissue biopsies respectively after harvest in vivo. In addition, the extracellular IL-26 protein concentrations in bronchoalveolar lavage (BAL) samples did correlate with increased epithelial cell transcription of an archetype intracellular signaling molecule downstream of the IL-26-receptor complex, STAT1, in the bronchial brush biopsies. Thus, our study suggests that viral stimulation causes the production of IL-26 in lining epithelial cells of human airway structural cells that constitute a critical immune barrier and that this production is modulated by Th17 cytokines.
Collapse
Affiliation(s)
- Karlhans Fru Che
- Unit for Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, SE-171 77 Stockholm, Sweden
| | - Riitta Kaarteenaho
- Unit of Medicine and Clinical Research, Pulmonary Division, University of Eastern Finland and Center of Medicine and Clinical Research, Division of Respiratory Medicine, Kuopio University Hospital, Kuopio, Finland
| | - Elisa Lappi-Blanco
- Department of Pathology, Center for Cancer Research and Translational Medicine, University of Oulu, Oulu, Finland
| | - Bettina Levänen
- Unit for Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, SE-171 77 Stockholm, Sweden
| | - Jitong Sun
- Unit for Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, SE-171 77 Stockholm, Sweden
| | - Åsa Wheelock
- Respiratory Medicine Unit. Center for Molecular Medicine, Department of Medicine Solna, Karolinska Institutet, SE-171 76 Stockholm
| | - Lena Palmberg
- Unit for Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, SE-171 77 Stockholm, Sweden
| | - C Magnus Sköld
- Respiratory Medicine Unit. Center for Molecular Medicine, Department of Medicine Solna, Karolinska Institutet, SE-171 76 Stockholm.,Lung Allergy Clinic, Karolinska University Hospital Solna, Stockholm, SE-171 76 Stockholm, Sweden
| | - Anders Lindén
- Unit for Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, SE-171 77 Stockholm, Sweden.,Lung Allergy Clinic, Karolinska University Hospital Solna, Stockholm, SE-171 76 Stockholm, Sweden
| |
Collapse
|
37
|
Naz S, Kolmert J, Yang M, Reinke SN, Kamleh MA, Snowden S, Heyder T, Levänen B, Erle DJ, Sköld CM, Wheelock ÅM, Wheelock CE. Metabolomics analysis identifies sex-associated metabotypes of oxidative stress and the autotaxin-lysoPA axis in COPD. Eur Respir J 2017. [PMID: 28642310 PMCID: PMC5898938 DOI: 10.1183/13993003.02322-2016] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a heterogeneous disease and a leading cause of mortality and morbidity worldwide. The aim of this study was to investigate the sex dependency of circulating metabolic profiles in COPD. Serum from healthy never-smokers (healthy), smokers with normal lung function (smokers), and smokers with COPD (COPD; Global Initiative for Chronic Obstructive Lung Disease stages I–II/A–B) from the Karolinska COSMIC cohort (n=116) was analysed using our nontargeted liquid chromatography–high resolution mass spectrometry metabolomics platform. Pathway analyses revealed that several altered metabolites are involved in oxidative stress. Supervised multivariate modelling showed significant classification of smokers from COPD (p=2.8×10−7). Sex stratification indicated that the separation was driven by females (p=2.4×10−7) relative to males (p=4.0×10−4). Significantly altered metabolites were confirmed quantitatively using targeted metabolomics. Multivariate modelling of targeted metabolomics data confirmed enhanced metabolic dysregulation in females with COPD (p=3.0×10−3) relative to males (p=0.10). The autotaxin products lysoPA (16:0) and lysoPA (18:2) correlated with lung function (forced expiratory volume in 1 s) in males with COPD (r=0.86; p<0.0001), but not females (r=0.44; p=0.15), potentially related to observed dysregulation of the miR-29 family in the lung. These findings highlight the role of oxidative stress in COPD, and suggest that sex-enhanced dysregulation in oxidative stress, and potentially the autotaxin–lysoPA axis, are associated with disease mechanisms and/or prevalence. Oxidative stress and the autotaxin–lysoPA axis evidence sex-associated metabotypes in the serum of COPD patientshttp://ow.ly/kAeE309MpdI
Collapse
Affiliation(s)
- Shama Naz
- Division of Physiological Chemistry 2, Dept of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Johan Kolmert
- Division of Physiological Chemistry 2, Dept of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden.,Division of Experimental Asthma and Allergy Research, Institute of Environmental Medicine, Karolinska Instituet, Stockholm, Sweden
| | - Mingxing Yang
- Respiratory Medicine Unit, Dept of Medicine Solna and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Stacey N Reinke
- Division of Physiological Chemistry 2, Dept of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Muhammad Anas Kamleh
- Division of Physiological Chemistry 2, Dept of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Stuart Snowden
- Division of Physiological Chemistry 2, Dept of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Tina Heyder
- Respiratory Medicine Unit, Dept of Medicine Solna and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Bettina Levänen
- Respiratory Medicine Unit, Dept of Medicine Solna and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - David J Erle
- Division of Pulmonary and Critical Care Medicine, Dept of Medicine and Lung Biology Center, University of California San Francisco, San Francisco, CA, USA
| | - C Magnus Sköld
- Respiratory Medicine Unit, Dept of Medicine Solna and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Åsa M Wheelock
- Respiratory Medicine Unit, Dept of Medicine Solna and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Both authors contributed equally
| | - Craig E Wheelock
- Division of Physiological Chemistry 2, Dept of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden .,Both authors contributed equally
| |
Collapse
|
38
|
Fei X, Bao W, Zhang P, Zhang X, Zhang G, Zhang Y, Zhou X, Zhang M. Inhalation of progesterone inhibits chronic airway inflammation of mice exposed to ozone. Mol Immunol 2017; 85:174-184. [PMID: 28279894 DOI: 10.1016/j.molimm.2017.02.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 02/09/2017] [Accepted: 02/09/2017] [Indexed: 10/20/2022]
Abstract
Chronic ozone exposure leads to a model of mice with lung inflammation, emphysema and oxidative stress. Progesterone plays an important role in attenuating the neuroinflammation. We assume that progesterone will reduce the chronic airway inflammation exposed to ozone and evaluate whether combination of progesterone with glucocorticoids results in synergistic effects. C57/BL6 mice were exposed to ozone (2.5ppm, 3h) 12 times over 6 weeks, and were administered with progesterone (0.03 or 0.3mg/L; inhaled) alone or combined with budesonide (BUD) (0.2g/L) after each exposure until the tenth week. Mice were studied 24h after final exposure, cells and inflammatory mediators were assessed in bronchoalveolar lavage fluid (BALF) and lungs used for evaluation of glucocorticoids receptors (GR), p38 mitogen-activated protein kinase (MAPK) phosphorylation and nuclear transcription factor κB (NF-κB) activation. Exposure to ozone resulted in a marked lung neutrophilia. Moreover, in ozone-exposed group, the levels of oxidative stress-related interleukin (IL)-1β, IL-6, IL-8, IL-17A, activated NF-κB and p38MAPK, airway inflammatory cells infiltration density, mean linear intercept (Lm) were greatly increased, FEV25 and glucocorticoids receptors (GR) were markedly decreased. Comparable to BUD, progesterone treatment dose-dependently led to a significant reduction of IL-1β, IL-6, IL-8, IL-17A, activated NF-κB and p38MAPK, and an increase of FEV25 and GR. Progesterone combined with BUD resulted in dramatic changes, compared to monotherapy of BUD or progesterone. Therefore, these results demonstrate that chronic ozone exposure has profound airway inflammatory effects counteracted by progesterone and progesterone acts synergistically with glucocorticoids in attenuating the airway inflammation dose-dependently.
Collapse
Affiliation(s)
- Xia Fei
- Department of Respiratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Wuping Bao
- Department of Respiratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Pengyu Zhang
- Department of Respiratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xue Zhang
- Department of Respiratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Guoqing Zhang
- Department of Respiratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yingying Zhang
- Department of Respiratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xin Zhou
- Department of Respiratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Min Zhang
- Department of Respiratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
39
|
Karimi R, Tornling G, Forsslund H, Mikko M, Wheelock ÅM, Nyrén S, Sköld CM. Differences in regional air trapping in current smokers with normal spirometry. Eur Respir J 2017; 49:49/1/1600345. [DOI: 10.1183/13993003.00345-2016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Accepted: 10/13/2016] [Indexed: 12/20/2022]
Abstract
We investigated regional air trapping on computed tomography in current smokers with normal spirometry. It was hypothesised that presence of regional air trapping may indicate a specific manifestation of smoking-related changes.40 current smokers, 40 patients with chronic obstructive pulmonary disease (COPD), and 40 healthy never- smokers underwent computed tomography scans. Regional air trapping was assessed on end-expiratory scans and emphysema, micronodules and bronchial wall thickening on inspiratory scans. The ratio of expiratory and inspiratory mean lung attenuation (E/I) was calculated as a measure of static (fixed) air trapping.Regional air trapping was present in 63% of current smokers, in 45% of never smokers and in 8% of COPD patients (p<0.001). Current smokers with and without regional air trapping had E/I ratio of 0.81 and 0.91, respectively (p<0.001). Forced expiratory volume in 1 s (FEV1) was significantly higher and emphysema less frequent in current smokers with regional air trapping.Current smokers with regional air trapping had higher FEV1 and less emphysema on computed tomography. In contrast, current smokers without regional air trapping resembled COPD. Our results highlight heterogeneity among smokers with normal spirometry and may contribute to early detection of smoking related structural changes in the lungs.
Collapse
|
40
|
Forsslund H, Yang M, Mikko M, Karimi R, Nyrén S, Engvall B, Grunewald J, Merikallio H, Kaarteenaho R, Wahlström J, Wheelock ÅM, Sköld CM. Gender differences in the T-cell profiles of the airways in COPD patients associated with clinical phenotypes. Int J Chron Obstruct Pulmon Dis 2016; 12:35-48. [PMID: 28053515 PMCID: PMC5191844 DOI: 10.2147/copd.s113625] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
T lymphocytes are believed to play an important role in the pathogenesis of chronic obstructive pulmonary disease (COPD). How T cells are recruited to the lungs and contribute to the inflammatory process is largely unknown. COPD is a heterogeneous disease, and discriminating disease phenotypes based on distinct molecular and cellular pathways may provide new approaches for individualized diagnosis and therapies. Bronchoalveolar lavage (BAL) and blood samples were obtained from 40 never-smokers, 40 smokers with normal lung function, and 38 COPD patients. T-cell chemokine receptor expression was analyzed with flow cytometry, and soluble BAL cytokines and chemokines were measured using a cytokine multiplex assay. Correlations with gender and clinical characteristics including lung imaging were investigated using multivariate modeling. Th1/Tc1- and Th2/Tc2-associated soluble analytes and T-cell chemokine receptors were analyzed as cumulative Th1/Tc1 and Th2/Tc2 immune responses. A higher expression of chemokine receptor CCR5 on CD8+ T cells in BAL and higher percentage of CXCR3+CD8+ T cells in blood was found in female smokers with COPD compared to those without COPD. CCR5 expression on CD4+ and CD8+ T cells was lower in BAL from male smokers with COPD compared to those without COPD. Among female smokers with COPD, Th1/Tc1 immune response was linked to BAL macrophage numbers and goblet cell density, and Th2/Tc2 response was associated with the measures of emphysema on high-resolution computed tomography. The highly gender-dependent T-cell profile in COPD indicates different links between cellular events and clinical manifestations in females compared to males. Our findings may reveal mechanisms of importance for the difference in clinical course in female COPD patients compared to males.
Collapse
Affiliation(s)
- Helena Forsslund
- Department of Medicine Solna and Centre for Molecular Medicine, Respiratory Medicine Unit
| | - Mingxing Yang
- Department of Medicine Solna and Centre for Molecular Medicine, Respiratory Medicine Unit
| | - Mikael Mikko
- Department of Medicine Solna and Centre for Molecular Medicine, Respiratory Medicine Unit
| | - Reza Karimi
- Department of Medicine Solna and Centre for Molecular Medicine, Respiratory Medicine Unit
| | - Sven Nyrén
- Department of Molecular Medicine and Surgery, Karolinska Institutet and Karolinska University Hospital Solna, Stockholm, Sweden
| | - Benita Engvall
- Department of Medicine Solna and Centre for Molecular Medicine, Respiratory Medicine Unit
| | - Johan Grunewald
- Department of Medicine Solna and Centre for Molecular Medicine, Respiratory Medicine Unit
| | - Heta Merikallio
- Department of Medicine Solna and Centre for Molecular Medicine, Respiratory Medicine Unit; Respiratory Research Unit and Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Riitta Kaarteenaho
- Respiratory Research Unit and Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland; Unit of Medicine and Clinical Research, Pulmonary Division, University of Eastern Finland; Center for Medicine and Clinical Research, Division of Respiratory Medicine, Kuopio University Hospital, Kuopio, Finland
| | - Jan Wahlström
- Department of Medicine Solna and Centre for Molecular Medicine, Respiratory Medicine Unit
| | - Åsa M Wheelock
- Department of Medicine Solna and Centre for Molecular Medicine, Respiratory Medicine Unit
| | - C Magnus Sköld
- Department of Medicine Solna and Centre for Molecular Medicine, Respiratory Medicine Unit
| |
Collapse
|
41
|
Kjellqvist S, Klose C, Surma MA, Hindy G, Mollet IG, Johansson A, Chavaux P, Gottfries J, Simons K, Melander O, Fernandez C. Identification of Shared and Unique Serum Lipid Profiles in Diabetes Mellitus and Myocardial Infarction. J Am Heart Assoc 2016; 5:JAHA.116.004503. [PMID: 27899364 PMCID: PMC5210412 DOI: 10.1161/jaha.116.004503] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background Diabetes mellitus (DM) and cardiovascular disease are associated with dyslipidemia, but the detailed lipid molecular pattern in both diseases remains unknown. Methods and Results We used shotgun mass spectrometry to determine serum levels of 255 molecular lipids in 316 controls, 171 DM, and 99 myocardial infarction (MI) events from a cohort derived from the Malmö Diet and Cancer study. Orthogonal projections to latent structures analyses were conducted between the lipids and clinical parameters describing DM or MI. Fatty acid desaturases (FADS) and elongation of very long chain fatty acid protein 5 (ELOVL5) activities were estimated by calculating product to precursor ratios of polyunsaturated fatty acids in complex lipids. FADS genotypes encoding these desaturases were then tested for association with lipid levels and ratios. Differences in the levels of lipids belonging to the phosphatidylcholine and triacylglyceride (TAG) classes contributed the most to separating DM from controls. TAGs also played a dominating role in discriminating MI from controls. Levels of C18:2 fatty acids in complex lipids were lower both in DM and MI versus controls (DM, P=0.004; MI, P=6.0E‐06) at least due to an acceleration in the metabolic flux from C18:2 to C20:4 (eg, increased estimated ELOVL5: DM, P=0.02; MI, P=0.04, and combined elongase‐desaturase activities: DM, P=3.0E‐06; MI, P=2.0E‐06). Minor allele carriers of FADS genotypes were associated with increased levels of C18:2 (P≤0.007) and lower desaturase activity (P≤0.002). Conclusions We demonstrate a possible relationship between decreased levels of C18:2 in complex lipids and DM or MI. We thereby highlight the importance of molecular lipids in the pathogenesis of both diseases.
Collapse
Affiliation(s)
- Sanela Kjellqvist
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | | | | | - George Hindy
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Inês G Mollet
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Anna Johansson
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Patrick Chavaux
- Department of Nutrition, Faculté de Médecine et de Pharmacie de la Timone, Aix Marseille University, Marseille, France
| | - Johan Gottfries
- Department of Chemistry and Molecular Biology, University of Gothenburg, Sweden
| | | | - Olle Melander
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | | |
Collapse
|
42
|
Balgoma D, Yang M, Sjödin M, Snowden S, Karimi R, Levänen B, Merikallio H, Kaarteenaho R, Palmberg L, Larsson K, Erle DJ, Dahlén SE, Dahlén B, Sköld CM, Wheelock ÅM, Wheelock CE. Linoleic acid-derived lipid mediators increase in a female-dominated subphenotype of COPD. Eur Respir J 2016; 47:1645-56. [DOI: 10.1183/13993003.01080-2015] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 01/13/2016] [Indexed: 12/11/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a leading cause of mortality; however, the role of inflammatory mediators in its pathobiology remains unclear. The aim of this study was to investigate the influence of gender in COPD on lipid mediator levels.Bronchoalveolar lavage fluid (BALF) and serum were obtained from healthy never-smokers, smokers and COPD patients (Global Initiative for Chronic Obstructive Lung Disease stage I–II/A–B) (n=114). 94 lipid mediators derived from the cytochrome-P450, lipoxygenase, and cyclooxygenase pathways were analysed by liquid chromatography-mass spectrometry.Multivariate modelling identified a 9-lipid panel in BALF that classified female smokers with COPD from healthy female smokers (p=6×10−6). No differences were observed for the corresponding male population (p=1.0). These findings were replicated in an independent cohort with 92% accuracy (p=0.005). The strongest drivers were the cytochrome P450-derived epoxide products of linoleic acid (leukotoxins) and their corresponding soluble epoxide hydrolase (sEH)-derived products (leukotoxin-diols). These species correlated with lung function (r=0.87; p=0.0009) and mRNA levels of enzymes putatively involved in their biosynthesis (r=0.96; p=0.003). Leukotoxin levels correlated with goblet cell abundance (r=0.72; p=0.028).These findings suggest a mechanism by which goblet cell-associated cytochrome-P450 and sEH activity produce elevated leukotoxin-diol levels, which play a putative role in the clinical manifestations of COPD in a female-dominated disease sub-phenotype.
Collapse
|
43
|
Sandberg A, Buschmann V, Kapusta P, Erdmann R, Wheelock ÅM. Use of Time-Resolved Fluorescence To Improve Sensitivity and Dynamic Range of Gel-Based Proteomics. Anal Chem 2016; 88:3067-74. [DOI: 10.1021/acs.analchem.5b03805] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- AnnSofi Sandberg
- Respiratory
Medicine Unit, Department of Medicine, Center for Molecular Medicine, Karolinska Institutet, 171 76 Stockholm, Sweden
| | | | - Peter Kapusta
- PicoQuant GmbH, Rudower Chaussee 29, 124 89 Berlin, Germany
| | - Rainer Erdmann
- PicoQuant GmbH, Rudower Chaussee 29, 124 89 Berlin, Germany
| | - Åsa M. Wheelock
- Respiratory
Medicine Unit, Department of Medicine, Center for Molecular Medicine, Karolinska Institutet, 171 76 Stockholm, Sweden
- Hikari Bio AB, Franzéngatan
39, 112 16 Stockholm, Sweden
| |
Collapse
|
44
|
Sathish V, Prakash Y. Sex Differences in Pulmonary Anatomy and Physiology. SEX DIFFERENCES IN PHYSIOLOGY 2016:89-103. [DOI: 10.1016/b978-0-12-802388-4.00006-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
|
45
|
Reynisdottir G, Olsen H, Joshua V, Engström M, Forsslund H, Karimi R, Sköld CM, Nyren S, Eklund A, Grunewald J, Catrina AI. Signs of immune activation and local inflammation are present in the bronchial tissue of patients with untreated early rheumatoid arthritis. Ann Rheum Dis 2015; 75:1722-7. [PMID: 26530319 DOI: 10.1136/annrheumdis-2015-208216] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2015] [Accepted: 09/20/2015] [Indexed: 12/26/2022]
Abstract
OBJECTIVES Events in the lungs might contribute to generation of anticitrullinated protein antibodies (ACPA) in rheumatoid arthritis (RA). We investigated if signs of immune activation are present in bronchial biopsies and bronchoalveolar lavage (BAL) of patients with early-untreated RA without clinical signs of lung involvement. METHODS Twenty-four patients with RA with symptom duration <1 year and naïve to disease-modifying antirheumatic drugs were subjected to bronchoscopy where BAL and mucosal bronchial biopsies were retrieved. For comparison, 15 bronchial biopsies and 79 BAL samples from healthy volunteers were available. Histological examination was performed to evaluate lymphocyte infiltration, presence of immune cells (T and B cells, plasma cells, dendritic cells and macrophages) and immune activation markers. Cell composition of BAL samples was analysed by differential counting and T cell subsets by flow cytometry. RESULTS Lymphocyte infiltration was more frequently found in ACPA-positive patients (50%) as compared with ACPA-negative patients (17%) and controls (13%). Germinal centres, B cells and plasma cells were only found in ACPA-positive patients. The frequency of T cells in bronchial biopsies of patients with ACPA-positive RA was positively associated with expression of immune activation markers. BAL samples of patients with ACPA-positive, but not ACPA-negative, RA had significantly higher relative numbers of lymphocytes and expressed higher levels of activation markers compared with controls. CONCLUSIONS Signs of immune cell accumulation and activation are present both in the bronchial tissue and in BAL of untreated patients with early RA without concomitant lung disease, strengthening the role of the lung compartment as an important player in ACPA-positive RA.
Collapse
Affiliation(s)
- Gudrun Reynisdottir
- Rheumatology Unit, Department of Medicine, Solna, Karolinska University Hospital and Institutet, Stockholm, Sweden
| | - Helga Olsen
- Division of Respiratory Medicine, Department of Medicine, Solna, Center for Molecular Medicine, Karolinska University Hospital, Solna, Karolinska Institutet, Stockholm, Sweden
| | - Vijay Joshua
- Rheumatology Unit, Department of Medicine, Solna, Karolinska University Hospital and Institutet, Stockholm, Sweden
| | - Marianne Engström
- Rheumatology Unit, Department of Medicine, Solna, Karolinska University Hospital and Institutet, Stockholm, Sweden
| | - Helena Forsslund
- Division of Respiratory Medicine, Department of Medicine, Solna, Center for Molecular Medicine, Karolinska University Hospital, Solna, Karolinska Institutet, Stockholm, Sweden
| | - Reza Karimi
- Division of Respiratory Medicine, Department of Medicine, Solna, Center for Molecular Medicine, Karolinska University Hospital, Solna, Karolinska Institutet, Stockholm, Sweden
| | - C Magnus Sköld
- Division of Respiratory Medicine, Department of Medicine, Solna, Center for Molecular Medicine, Karolinska University Hospital, Solna, Karolinska Institutet, Stockholm, Sweden
| | - Sven Nyren
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Anders Eklund
- Division of Respiratory Medicine, Department of Medicine, Solna, Center for Molecular Medicine, Karolinska University Hospital, Solna, Karolinska Institutet, Stockholm, Sweden
| | - Johan Grunewald
- Division of Respiratory Medicine, Department of Medicine, Solna, Center for Molecular Medicine, Karolinska University Hospital, Solna, Karolinska Institutet, Stockholm, Sweden
| | - Anca I Catrina
- Rheumatology Unit, Department of Medicine, Solna, Karolinska University Hospital and Institutet, Stockholm, Sweden
| |
Collapse
|
46
|
Pinkerton KE, Harbaugh M, Han MK, Jourdan Le Saux C, Van Winkle LS, Martin WJ, Kosgei RJ, Carter EJ, Sitkin N, Smiley-Jewell SM, George M. Women and Lung Disease. Sex Differences and Global Health Disparities. Am J Respir Crit Care Med 2015; 192:11-6. [PMID: 25945507 PMCID: PMC4511423 DOI: 10.1164/rccm.201409-1740pp] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 05/05/2015] [Indexed: 12/29/2022] Open
Abstract
There is growing evidence that a number of pulmonary diseases affect women differently and with a greater degree of severity than men. The causes for such sex disparity is the focus of this Blue Conference Perspective review, which explores basic cellular and molecular mechanisms, life stages, and clinical outcomes based on environmental, sociocultural, occupational, and infectious scenarios, as well as medical health beliefs. Owing to the breadth of issues related to women and lung disease, we present examples of both basic and clinical concepts that may be the cause for pulmonary disease disparity in women. These examples include those diseases that predominantly affect women, as well as the rising incidence among women for diseases traditionally occurring in men, such as chronic obstructive pulmonary disease. Sociocultural implications of pulmonary disease attributable to biomass burning and infectious diseases among women in low- to middle-income countries are reviewed, as are disparities in respiratory health among sexual minority women in high-income countries. The implications of the use of complementary and alternative medicine by women to influence respiratory disease are examined, and future directions for research on women and respiratory health are provided.
Collapse
Affiliation(s)
| | - Mary Harbaugh
- Public Advisory Roundtable of the American Thoracic Society, New York, New York
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Sex steroid signaling: implications for lung diseases. Pharmacol Ther 2015; 150:94-108. [PMID: 25595323 DOI: 10.1016/j.pharmthera.2015.01.007] [Citation(s) in RCA: 115] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 01/09/2015] [Indexed: 12/12/2022]
Abstract
There is increasing recognition that sex hormones (estrogen, progesterone, and testosterone) have biological and pathophysiological actions in peripheral, non-reproductive organs, including the lung. Clinically, sex differences in the incidence, morbidity and mortality of lung diseases such as asthma, chronic obstructive pulmonary disease (COPD), pulmonary fibrosis, lung cancer and pulmonary hypertension have been noted, although intrinsic sex differences vs. the roles of sex steroids are still not well-understood. Accordingly, it becomes important to ask the following questions: 1) Which sex steroids are involved? 2) How do they affect different components of the lung under normal circumstances? 3) How does sex steroid signaling change in or contribute to lung disease, and in this regard, are sex steroids detrimental or beneficial? As our understanding of sex steroid signaling in the lung improves, it is important to consider whether such information can be used to develop new therapeutic strategies to target lung diseases, perhaps in both sexes or in a sex-specific manner. In this review, we focus on the basics of sex steroid signaling, and the current state of knowledge regarding how they influence structure and function of specific lung components across the life span and in the context of some important lung diseases. We then summarize the potential for sex steroids as useful biomarkers and therapeutic targets in these lung diseases as a basis for future translational research in the area of gender and individualized medicine.
Collapse
|
48
|
Aryal S, Diaz-Guzman E, Mannino DM. Influence of sex on chronic obstructive pulmonary disease risk and treatment outcomes. Int J Chron Obstruct Pulmon Dis 2014; 9:1145-54. [PMID: 25342899 PMCID: PMC4206206 DOI: 10.2147/copd.s54476] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD), one of the most common chronic diseases and a leading cause of death, has historically been considered a disease of men. However, there has been a rapid increase in the prevalence, morbidity, and mortality of COPD in women over the last two decades. This has largely been attributed to historical increases in tobacco consumption among women. But the influence of sex on COPD is complex and involves several other factors, including differential susceptibility to the effects of tobacco, anatomic, hormonal, and behavioral differences, and differential response to therapy. Interestingly, nonsmokers with COPD are more likely to be women. In addition, women with COPD are more likely to have a chronic bronchitis phenotype, suffer from less cardiovascular comorbidity, have more concomitant depression and osteoporosis, and have a better outcome with acute exacerbations. Women historically have had lower mortality with COPD, but this is changing as well. There are also differences in how men and women respond to different therapies. Despite the changing face of COPD, care providers continue to harbor a sex bias, leading to underdiagnosis and delayed diagnosis of COPD in women. In this review, we present the current knowledge on the influence of sex on COPD risk factors, epidemiology, diagnosis, comorbidities, treatment, and outcomes, and how this knowledge may be applied to improve clinical practices and advance research.
Collapse
Affiliation(s)
- Shambhu Aryal
- Division of Pulmonary, Allergy and Critical Care Medicine, Duke University, Durham, NC, USA
| | - Enrique Diaz-Guzman
- Division of Pulmonary, Allergy and Critical Care, University of Alabama, Birmingham, AL, USA
| | - David M Mannino
- Department of Preventive Medicine and Environmental Health, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
49
|
Forsslund H, Mikko M, Karimi R, Grunewald J, Wheelock ÅM, Wahlström J, Sköld CM. Distribution of T-cell subsets in BAL fluid of patients with mild to moderate COPD depends on current smoking status and not airway obstruction. Chest 2014; 145:711-722. [PMID: 24264182 DOI: 10.1378/chest.13-0873] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND COPD is characterized by chronic inflammation. CD8+ T cells and CD4+ T cells have both been implicated in the inflammatory response. We investigated whether the lymphocyte and T-cell subpopulations in BAL differ between patients with COPD who are current smokers and those who are ex-smokers. METHODS Forty never smokers, 40 smokers with normal lung function, and 38 patients with COPD, GOLD (Global Initiative for Chronic Obstructive Pulmonary Disease) stage I-II (27 smokers and 11 ex-smokers) underwent BAL. Using flow cytometry, cells were analyzed from BAL and blood for T-cell subsets, B cells, natural killer cells, and natural killer T (NKT)-like cells. The differentiation status of CD4+ T cells was also determined. RESULTS Smokers with or without COPD had higher percentages of CD8+ T cells and NKT-like cells in BAL than did never smokers and ex-smokers with COPD. Most of the NKT-like cells were CD8+. In contrast, the percentages of CD4+ T cells were lower in the smoking than in the nonsmoking groups. In blood, the frequency of CD4+ T cells was increased in the two smoking groups. Current smokers also had increased numbers of activated (CD69+) naive and effector CD4+ T cells in BAL compared with nonsmokers, particularly in patients with COPD. In male smokers with COPD, the percentage of CD8+ T cells in BAL positively correlated with the number of cigarettes per day. CONCLUSIONS Current smoking status has a greater impact than airway obstruction on the distribution of T-cell subsets in BAL of patients with mild to moderate COPD. This fact must be considered when the role of T cells in COPD is evaluated. Our results stress the importance of subgrouping patients with COPD in terms of smoking.
Collapse
Affiliation(s)
- Helena Forsslund
- Respiratory Medicine Unit, Department of Medicine Solna and Centre for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Solna, Stockholm, Sweden.
| | - Mikael Mikko
- Respiratory Medicine Unit, Department of Medicine Solna and Centre for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Solna, Stockholm, Sweden
| | - Reza Karimi
- Respiratory Medicine Unit, Department of Medicine Solna and Centre for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Solna, Stockholm, Sweden
| | - Johan Grunewald
- Respiratory Medicine Unit, Department of Medicine Solna and Centre for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Solna, Stockholm, Sweden
| | - Åsa M Wheelock
- Respiratory Medicine Unit, Department of Medicine Solna and Centre for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Solna, Stockholm, Sweden
| | - Jan Wahlström
- Respiratory Medicine Unit, Department of Medicine Solna and Centre for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Solna, Stockholm, Sweden
| | - C Magnus Sköld
- Respiratory Medicine Unit, Department of Medicine Solna and Centre for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Solna, Stockholm, Sweden
| |
Collapse
|
50
|
Wheelock ÅM, Wheelock CE. Trials and tribulations of 'omics data analysis: assessing quality of SIMCA-based multivariate models using examples from pulmonary medicine. MOLECULAR BIOSYSTEMS 2014; 9:2589-96. [PMID: 23999822 DOI: 10.1039/c3mb70194h] [Citation(s) in RCA: 233] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Respiratory diseases are multifactorial heterogeneous diseases that have proved recalcitrant to understanding using focused molecular techniques. This trend has led to the rise of 'omics approaches (e.g., transcriptomics, proteomics) and subsequent acquisition of large-scale datasets consisting of multiple variables. In 'omics technology-based investigations, discrepancies between the number of variables analyzed (e.g., mRNA, proteins, metabolites) and the number of study subjects constitutes a major statistical challenge. The application of traditional univariate statistical methods (e.g., t-test) to these "short-and-wide" datasets may result in high numbers of false positives, while the predominant approach of p-value correction to account for these high false positive rates (e.g., FDR, Bonferroni) are associated with significant losses in statistical power. In other words, the benefit in decreased false positives must be counterbalanced with a concomitant loss in true positives. As an alternative, multivariate statistical analysis (MVA) is increasingly being employed to cope with 'omics-based data structures. When properly applied, MVA approaches can be powerful tools for integration and interpretation of complex 'omics-based datasets towards the goal of identifying biomarkers and/or subphenotypes. However, MVA methods are also prone to over-interpretation and misuse. A common software used in biomedical research to perform MVA-based analyses is the SIMCA package, which includes multiple MVA methods. In this opinion piece, we propose guidelines for minimum reporting standards for a SIMCA-based workflow, in terms of data preprocessing (e.g., normalization, scaling) and model statistics (number of components, R2, Q2, and CV-ANOVA p-value). Examples of these applications in recent COPD and asthma studies are provided. It is expected that readers will gain an increased understanding of the power and utility of MVA methods for applications in biomedical research.
Collapse
Affiliation(s)
- Åsa M Wheelock
- Respiratory Medicine Unit, Department of Medicine, and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.
| | | |
Collapse
|