1
|
El-Husseini ZW, Karp T, Lan A, Gillett TE, Qi C, Khalenkow D, van der Molen T, Brightling C, Papi A, Rabe KF, Siddiqui S, Singh D, Kraft M, Beghé B, Joubert P, Bossé Y, Sin D, Cordero AH, Timens W, Brandsma CA, Hao K, Nickle DC, Vonk JM, Nawijn MC, van den Berge M, Gosens R, Faiz A, Koppelman GH. Improved Annotation of Asthma Gene Variants with Cell Type Deconvolution of Nasal and Lung Expression Quantitative Trait Loci. Am J Respir Cell Mol Biol 2025; 72:607-614. [PMID: 39836087 DOI: 10.1165/rcmb.2024-0251ma] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 01/21/2025] [Indexed: 01/22/2025] Open
Abstract
Asthma is a genetically complex inflammatory airway disease associated with more than 200 SNPs. However, the functional effects of many asthma-associated SNPs in lung and airway epithelial samples are unknown. Here, we aimed to conduct expression quantitative trait loci (eQTL) analysis using a meta-analysis of nasal and lung samples. We hypothesize that incorporating cell type proportions of airway and lung samples enhances eQTL analysis outcomes. Nasal brush (n = 792) and lung tissue (n = 1,087) samples were investigated separately. Initially, a general eQTL analysis identified genetic variants associated with gene expression levels. Estimated cell type proportions were adjusted based on the Human Lung Cell Atlas. In addition, the presence of significant interaction effects between asthma-associated SNPs and each cell type proportion was explored and considered evidence for cell type-associated eQTL. In nasal brush and lung parenchyma samples, 44 and 116 asthma-associated SNPs were identified as eQTL. Adjusting for cell type proportions revealed eQTL for an additional 17 genes (e.g., FCER1G, CD200R1, and GABBR2) and 16 genes (e.g., CYP2C8, SLC9A2, and SGCD) in nose and lung, respectively. Moreover, we identified eQTL for nine SNPs annotated to genes such as VASP, FOXA3, and PCDHB12 displayed significant interactions with cell type proportions of club, goblet, and alveolar macrophages. Our findings demonstrate increased power for identifying eQTL among asthma-associated SNPs by considering cell type proportion of the bulk RNA-sequencing data from nasal and lung tissues. Integration of cell type deconvolution and eQTL analysis enhances our understanding of asthma genetics and cellular mechanisms, uncovering potential therapeutic targets for personalized interventions.
Collapse
Affiliation(s)
- Zaid W El-Husseini
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
- Groningen Research Institute for Asthma and COPD (GRIAC)
- Molecular Pharmacology, Groningen Research Institute of Pharmacy, Groningen, the Netherlands
| | - Tatiana Karp
- Groningen Research Institute for Asthma and COPD (GRIAC)
- Department of Pulmonary Diseases
| | - Andy Lan
- Respiratory Bioinformatics and Molecular Biology, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Tessa E Gillett
- Groningen Research Institute for Asthma and COPD (GRIAC)
- Department of Pathology and Medical Biology
| | - Cancan Qi
- Microbiome Medicine Center, Division of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Dmitry Khalenkow
- Groningen Research Institute for Asthma and COPD (GRIAC)
- Laboratory of Genome Structure and Ageing, European Research Institute for the Biology of Ageing, University Medical Center Groningen, Groningen, the Netherlands
| | | | - Chris Brightling
- Department of Infection, Immunity, and Inflammation, Institute for Lung Health, University of Leicester, Leicester, United Kingdom
| | - Alberto Papi
- Department of Respiratory Medicine, University of Ferrara, Ferrara, Italy
| | - Klaus F Rabe
- Department of Medicine, Christian Albrechts University Kiel, Kiel and Lungen Clinic Grosshansdorf, Grosshansdorf, Germany (Members of the German Center for Lung Research [DZL])
| | - Salman Siddiqui
- National Heart and Lung Institute, Imperial College and Imperial NIHR Biomedical Research Centre, London, United Kingdom
| | - Dave Singh
- Medicines Evaluation Unit, Manchester University NHS Foundation Hospital Trust, University of Manchester, Manchester, United Kingdom
| | - Monica Kraft
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Bianca Beghé
- Section of Respiratory Diseases, Department of Oncology, Haematology, and Respiratory Diseases, University of Modena and Reggio Emilia, Policlinico di Modena, Modena, Italy
| | - Philippe Joubert
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec City, Québec, Canada
| | - Yohan Bossé
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec City, Québec, Canada
| | - Don Sin
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, British Columbia, Canada; and
| | - Ana H Cordero
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, British Columbia, Canada; and
| | - Wim Timens
- Department of Pathology and Medical Biology
| | - Corry-Anke Brandsma
- Groningen Research Institute for Asthma and COPD (GRIAC)
- Department of Pathology and Medical Biology
| | - Ke Hao
- Merck Research Laboratories, Boston, Massachusetts
| | | | - Judith M Vonk
- Groningen Research Institute for Asthma and COPD (GRIAC)
- Department of Epidemiology, and
| | - Martijn C Nawijn
- Groningen Research Institute for Asthma and COPD (GRIAC)
- Department of Pathology and Medical Biology
| | - Maarten van den Berge
- Groningen Research Institute for Asthma and COPD (GRIAC)
- Department of Pulmonary Diseases
| | - Reinoud Gosens
- Groningen Research Institute for Asthma and COPD (GRIAC)
- Molecular Pharmacology, Groningen Research Institute of Pharmacy, Groningen, the Netherlands
| | - Alen Faiz
- Groningen Research Institute for Asthma and COPD (GRIAC)
- Respiratory Bioinformatics and Molecular Biology, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Gerard H Koppelman
- Groningen Research Institute for Asthma and COPD (GRIAC)
- Department of Pediatric Pulmonology and Pediatric Allergology, Beatrix Children's Hospital, University of Groningen, and
| |
Collapse
|
2
|
Karp T, Faiz A, van Nijnatten J, Kerstjens HAM, Boudewijn I, Kraft M, Vonk JM, Nawijn MC, Heijink IH, Beghé B, Rabe KF, Papi A, Brightling C, Singh D, van der Molen T, Siddiqui S, Christenson S, Guryev V, van den Berge M. Nasal epithelial gene expression identifies relevant asthma endotypes in the ATLANTIS study. Thorax 2024; 79:905-914. [PMID: 39009441 DOI: 10.1136/thorax-2023-221230] [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: 11/23/2023] [Accepted: 06/18/2024] [Indexed: 07/17/2024]
Abstract
INTRODUCTION Asthma is an inflammatory airways disease encompassing multiple phenotypes and endotypes. Several studies suggested gene expression in nasal epithelium to serve as a proxy for bronchial epithelium, being a non-invasive approach to investigate lung diseases. We hypothesised that molecular differences in upper airway epithelium reflect asthma-associated differences in the lower airways and are associated with clinical expression of asthma. METHODS We analysed nasal epithelial gene expression data from 369 patients with asthma and 58 non-asthmatic controls from the Assessment of Small Airways Involvement in Asthma study. Unsupervised hierarchical clustering was performed on asthma-associated genes. Asthma-associated gene signatures were replicated in independent cohorts with nasal and bronchial brushes data by comparing Gene Set Variation Analysis scores between asthma patients and non-asthmatic controls. RESULTS We identified 67 higher expressed and 59 lower expressed genes in nasal epithelium from asthma patients compared with controls (false discovery rate<0.05), including CLCA1, CST1 and POSTN, genes well known to reflect asthma in bronchial airway epithelium. Hierarchical clustering revealed several molecular asthma endotypes with distinct clinical characteristics, including an endotype with higher blood and sputum eosinophils, high fractional exhaled nitric oxide, and more severe small airway dysfunction, as reflected by lower forced expiratory flow at 50%. In an independent cohort, we demonstrated that genes higher expressed in the nasal epithelium reflect asthma-associated changes in the lower airways. CONCLUSION Our results show that the nasal epithelial gene expression profile reflects asthma-related processes in the lower airways. We suggest that nasal epithelium may be a useful non-invasive tool to identify asthma endotypes and may advance personalised management of the disease.
Collapse
Affiliation(s)
- Tatiana Karp
- Groningen Research Institute for Asthma and COPD, University of Groningen, University Medical Center, Groningen, The Netherlands
- Department of Pulmonary Diseases, University of Groningen, University Medical Center, Groningen, The Netherlands
| | - Alen Faiz
- Respiratory Bioinformatics and Molecular Biology, University of Technology Sydney, Ultimo, New South Wales, Australia
| | - Jos van Nijnatten
- Groningen Research Institute for Asthma and COPD, University of Groningen, University Medical Center, Groningen, The Netherlands
- Respiratory Bioinformatics and Molecular Biology, University of Technology Sydney, Ultimo, New South Wales, Australia
| | - Huib A M Kerstjens
- Groningen Research Institute for Asthma and COPD, University of Groningen, University Medical Center, Groningen, The Netherlands
- Department of Pulmonary Diseases, University of Groningen, University Medical Center, Groningen, The Netherlands
| | - Ilse Boudewijn
- Groningen Research Institute for Asthma and COPD, University of Groningen, University Medical Center, Groningen, The Netherlands
- Department of Pulmonary Diseases, University of Groningen, University Medical Center, Groningen, The Netherlands
| | - Monica Kraft
- Samuel Bronfman Department of Medicine, Icahn School of Medicine, Mount Sinai Medical Center, New York, New York, USA
| | - Judith M Vonk
- Groningen Research Institute for Asthma and COPD, University of Groningen, University Medical Center, Groningen, The Netherlands
- Department of Epidemiology, University of Groningen, University Medical Center, Groningen, The Netherlands
| | - Martijn C Nawijn
- Groningen Research Institute for Asthma and COPD, University of Groningen, University Medical Center, Groningen, The Netherlands
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center, Groningen, The Netherlands
| | - Irene H Heijink
- Groningen Research Institute for Asthma and COPD, University of Groningen, University Medical Center, Groningen, The Netherlands
- Department of Pulmonary Diseases, University of Groningen, University Medical Center, Groningen, The Netherlands
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center, Groningen, The Netherlands
| | - Bianca Beghé
- Department of Respiratory Diseases, University of Modena and Reggio Emilia, Modena, Italy
| | - Klaus F Rabe
- Department of Medicine, Christian Albrechts University Kiel, Kiel and LungenClinic, Grosshansdorf, Germany
| | - Alberto Papi
- Section of Respiratory Medicine, Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Chris Brightling
- The Institute for Lung Health, NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
| | - Dave Singh
- Centre for Respiratory Medicine and Allergy, University of Manchester, Manchester University NHS Foundation Trust, Manchester, UK
| | - Thys van der Molen
- Groningen Research Institute for Asthma and COPD, University of Groningen, University Medical Center, Groningen, The Netherlands
- Department of General Practice and Elderly Care Medicine, University of Groningen, University Medical Center, Groningen, The Netherlands
| | - Salman Siddiqui
- Imperial College London National Heart and Lung Institute, London, UK
| | - Stephanie Christenson
- Division of Pulmonary, Critical Care, Allergy, and Sleep Medicine, University of California San Francisco, San Francisco, California, USA
| | - Victor Guryev
- Groningen Research Institute for Asthma and COPD, University of Groningen, University Medical Center, Groningen, The Netherlands
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center, Groningen, The Netherlands
| | - Maarten van den Berge
- Groningen Research Institute for Asthma and COPD, University of Groningen, University Medical Center, Groningen, The Netherlands
- Department of Pulmonary Diseases, University of Groningen, University Medical Center, Groningen, The Netherlands
| |
Collapse
|
3
|
van Nijnatten J, Faiz A, Timens W, Guryev V, Slebos DJ, Klooster K, Hartman JE, Kole T, Choy DF, Chakrabarti A, Grimbaldeston M, Rosenberger CM, Kerstjens H, Brandsma CA, van den Berge M. A bronchial gene signature specific for severe COPD that is retained in the nose. ERJ Open Res 2023; 9:00354-2023. [PMID: 38020574 PMCID: PMC10680034 DOI: 10.1183/23120541.00354-2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 08/09/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction A subset of COPD patients develops advanced disease with severe airflow obstruction, hyperinflation and extensive emphysema. We propose that the pathogenesis in these patients differs from mild-moderate COPD and is reflected by bronchial gene expression. The aim of the present study was to identify a unique bronchial epithelial gene signature for severe COPD patients. Methods We obtained RNA sequencing data from bronchial brushes from 123 ex-smokers with severe COPD, 23 with mild-moderate COPD and 23 non-COPD controls. We identified genes specific to severe COPD by comparing severe COPD to non-COPD controls, followed by removing genes that were also differentially expressed between mild-moderate COPD and non-COPD controls. Next, we performed a pathway analysis on these genes and evaluated whether this signature is retained in matched nasal brushings. Results We identified 219 genes uniquely differentially expressed in severe COPD. Interaction network analysis identified VEGFA and FN1 as the key genes with the most interactions. Genes were involved in extracellular matrix regulation, collagen binding and the immune response. Of interest were 10 genes (VEGFA, DCN, SPARC, COL6A2, MGP, CYR61, ANXA6, LGALS1, C1QA and C1QB) directly connected to fibronectin 1 (FN1). Most of these genes were lower expressed in severe COPD and showed the same effect in nasal brushings. Conclusions We found a unique severe COPD bronchial gene signature with key roles for VEGFA and FN1, which was retained in the upper airways. This supports the hypothesis that severe COPD, at least partly, comprises a different pathology and supports the potential for biomarker development based on nasal brushes in COPD.
Collapse
Affiliation(s)
- Jos van Nijnatten
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, the Netherlands
- University of Groningen University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, Groningen, the Netherlands
- University of Technology Sydney, Respiratory Bioinformatics and Molecular Biology, Sydney, NSW, Australia
| | - Alen Faiz
- University of Groningen University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, Groningen, the Netherlands
- University of Technology Sydney, Respiratory Bioinformatics and Molecular Biology, Sydney, NSW, Australia
- University of Groningen University Medical Center Groningen, Department of Pulmonary Diseases, Groningen, the Netherlands
| | - Wim Timens
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, the Netherlands
- University of Groningen University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, Groningen, the Netherlands
| | - Victor Guryev
- University of Groningen University Medical Center Groningen, European Research Institute for the Biology of Ageing, Groningen, the Netherlands
| | - Dirk-Jan Slebos
- University of Groningen University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, Groningen, the Netherlands
- University of Groningen University Medical Center Groningen, Department of Pulmonary Diseases, Groningen, the Netherlands
| | - Karin Klooster
- University of Groningen University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, Groningen, the Netherlands
- University of Groningen University Medical Center Groningen, Department of Pulmonary Diseases, Groningen, the Netherlands
| | - Jorine E. Hartman
- University of Groningen University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, Groningen, the Netherlands
- University of Groningen University Medical Center Groningen, Department of Pulmonary Diseases, Groningen, the Netherlands
| | - Tessa Kole
- University of Groningen University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, Groningen, the Netherlands
- University of Groningen University Medical Center Groningen, Department of Pulmonary Diseases, Groningen, the Netherlands
| | | | | | | | | | - Huib Kerstjens
- University of Groningen University Medical Center Groningen, Department of Pulmonary Diseases, Groningen, the Netherlands
| | - Corry-Anke Brandsma
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, the Netherlands
- University of Groningen University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, Groningen, the Netherlands
- These authors contributed equally
| | - Maarten van den Berge
- University of Groningen University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, Groningen, the Netherlands
- University of Groningen University Medical Center Groningen, Department of Pulmonary Diseases, Groningen, the Netherlands
- These authors contributed equally
| |
Collapse
|
4
|
Gupta MK, Peng H, Li Y, Xu CJ. The role of DNA methylation in personalized medicine for immune-related diseases. Pharmacol Ther 2023; 250:108508. [PMID: 37567513 DOI: 10.1016/j.pharmthera.2023.108508] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 08/03/2023] [Accepted: 08/08/2023] [Indexed: 08/13/2023]
Abstract
Epigenetics functions as a bridge between host genetic & environmental factors, aiding in human health and diseases. Many immune-related diseases, including infectious and allergic diseases, have been linked to epigenetic mechanisms, particularly DNA methylation. In this review, we summarized an updated overview of DNA methylation and its importance in personalized medicine, and demonstrated that DNA methylation has excellent potential for disease prevention, diagnosis, and treatment in a personalized manner. The future implications and limitations of the DNA methylation study have also been well-discussed.
Collapse
Affiliation(s)
- Manoj Kumar Gupta
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany; TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
| | - He Peng
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany; TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
| | - Yang Li
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany; TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany; Department of Internal Medicine and Radboud Institute for Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Cheng-Jian Xu
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany; TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany; Department of Internal Medicine and Radboud Institute for Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands.
| |
Collapse
|
5
|
Rathnayake SNH, Ditz B, van Nijnatten J, Sadaf T, Hansbro PM, Brandsma CA, Timens W, van Schadewijk A, Hiemstra PS, ten Hacken NHT, Oliver B, Kerstjens HAM, van den Berge M, Faiz A. Smoking induces shifts in cellular composition and transcriptome within the bronchial mucus barrier. Respirology 2023; 28:132-142. [PMID: 36414410 PMCID: PMC10947540 DOI: 10.1111/resp.14401] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 10/07/2022] [Indexed: 11/24/2022]
Abstract
BACKGROUND AND OBJECTIVE Smoking disturbs the bronchial-mucus-barrier. This study assesses the cellular composition and gene expression shifts of the bronchial-mucus-barrier with smoking to understand the mechanism of mucosal damage by cigarette smoke exposure. We explore whether single-cell-RNA-sequencing (scRNA-seq) based cellular deconvolution (CD) can predict cell-type composition in RNA-seq data. METHODS RNA-seq data of bronchial biopsies from three cohorts were analysed using CD. The cohorts included 56 participants with chronic obstructive pulmonary disease [COPD] (38 smokers; 18 ex-smokers), 77 participants without COPD (40 never-smokers; 37 smokers) and 16 participants who stopped smoking for 1 year (11 COPD and 5 non-COPD-smokers). Differential gene expression was used to investigate gene expression shifts. The CD-derived goblet cell ratios were validated by correlating with staining-derived goblet cell ratios from the COPD cohort. Statistics were done in the R software (false discovery rate p-value < 0.05). RESULTS Both CD methods indicate a shift in bronchial-mucus-barrier cell composition towards goblet cells in COPD and non-COPD-smokers compared to ex- and never-smokers. It shows that the effect was reversible within a year of smoking cessation. A reduction of ciliated and basal cells was observed with current smoking, which resolved following smoking cessation. The expression of mucin and sodium channel (ENaC) genes, but not chloride channel genes, were altered in COPD and current smokers compared to never smokers or ex-smokers. The goblet cell-derived staining scores correlate with CD-derived goblet cell ratios. CONCLUSION Smoking alters bronchial-mucus-barrier cell composition, transcriptome and increases mucus production. This effect is partly reversible within a year of smoking cessation. CD methodology can predict goblet-cell percentages from RNA-seq.
Collapse
Affiliation(s)
- Senani N. H. Rathnayake
- University of Technology Sydney, Respiratory Bioinformatics and Molecular Biology (RBMB), School of Life SciencesSydneyNew South WalesAustralia
- The University of Sydney, Respiratory Cellular and Molecular Biology (RCMB), Woolcock Institute of Medical ResearchSydneyNew South WalesAustralia
| | - Benedikt Ditz
- Department of Pulmonary DiseasesUniversity of Groningen, University Medical Center GroningenGroningenthe Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPDGroningenthe Netherlands
| | - Jos van Nijnatten
- University of Technology Sydney, Respiratory Bioinformatics and Molecular Biology (RBMB), School of Life SciencesSydneyNew South WalesAustralia
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPDGroningenthe Netherlands
- Department of Pathology & Medical BiologyUniversity of Groningen, University Medical Center GroningenGroningenthe Netherlands
| | - Tayyaba Sadaf
- University of Technology Sydney, Respiratory Bioinformatics and Molecular Biology (RBMB), School of Life SciencesSydneyNew South WalesAustralia
- Centre for InflammationCentenary Institute, and the University of Technology Sydney, Faculty of ScienceSydneyNew South WalesAustralia
| | - Philip M. Hansbro
- Centre for InflammationCentenary Institute, and the University of Technology Sydney, Faculty of ScienceSydneyNew South WalesAustralia
| | - Corry A. Brandsma
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPDGroningenthe Netherlands
- Department of Pathology & Medical BiologyUniversity of Groningen, University Medical Center GroningenGroningenthe Netherlands
| | - Wim Timens
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPDGroningenthe Netherlands
- Department of Pathology & Medical BiologyUniversity of Groningen, University Medical Center GroningenGroningenthe Netherlands
| | | | - Peter S. Hiemstra
- Department of PulmonologyLeiden University Medical CenterLeidenthe Netherlands
| | - Nick H. T. ten Hacken
- Department of Pulmonary DiseasesUniversity of Groningen, University Medical Center GroningenGroningenthe Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPDGroningenthe Netherlands
| | - Brian Oliver
- The University of Sydney, Respiratory Cellular and Molecular Biology (RCMB), Woolcock Institute of Medical ResearchSydneyNew South WalesAustralia
| | - Huib A. M. Kerstjens
- Department of Pulmonary DiseasesUniversity of Groningen, University Medical Center GroningenGroningenthe Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPDGroningenthe Netherlands
| | - Maarten van den Berge
- Department of Pulmonary DiseasesUniversity of Groningen, University Medical Center GroningenGroningenthe Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPDGroningenthe Netherlands
| | - Alen Faiz
- University of Technology Sydney, Respiratory Bioinformatics and Molecular Biology (RBMB), School of Life SciencesSydneyNew South WalesAustralia
- The University of Sydney, Respiratory Cellular and Molecular Biology (RCMB), Woolcock Institute of Medical ResearchSydneyNew South WalesAustralia
- Department of Pulmonary DiseasesUniversity of Groningen, University Medical Center GroningenGroningenthe Netherlands
| |
Collapse
|
6
|
Rathnayake SNH, Ditz B, Willemse BWM, Timens W, Kooistra W, Heijink IH, Oliver BGG, van den Berge M, Faiz A. Longitudinal Effects of 1-Year Smoking Cessation on Human Bronchial Epithelial Transcriptome. Chest 2023:S0012-3692(23)00158-7. [PMID: 36716955 DOI: 10.1016/j.chest.2022.12.050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 12/19/2022] [Indexed: 01/30/2023] Open
Affiliation(s)
- Senani N H Rathnayake
- Respiratory Bioinformatics and Molecular Biology Group, Ultimo, NSW, Australia; School of Life Sciences, The University of Technology Sydney, Sydney, NSW, Australia; Respiratory Cellular and Molecular Biology Group, Woolcock Institute of Medical Research, The University of Sydney, Sydney, NSW, Australia
| | - B Ditz
- Division of Paediatric Pulmonology, Beatrix Children's Hospital, Groningen Research Institute for Asthma and COPD, Groningen, the Netherlands; Department of Pulmonary Diseases, Groningen, the Netherlands
| | - Brigitte W M Willemse
- University of Technology Sydney; the Department of Paediatrics, Ultimo, NSW, Australia
| | - Wim Timens
- Division of Paediatric Pulmonology, Beatrix Children's Hospital, Groningen Research Institute for Asthma and COPD, Groningen, the Netherlands; Department of Pathology & Medical Biology, University Medical Centre Groningen, University of Groningen, Groningen, the Netherlands
| | - Wierd Kooistra
- Division of Paediatric Pulmonology, Beatrix Children's Hospital, Groningen Research Institute for Asthma and COPD, Groningen, the Netherlands; Department of Pathology & Medical Biology, University Medical Centre Groningen, University of Groningen, Groningen, the Netherlands
| | - Irene H Heijink
- Division of Paediatric Pulmonology, Beatrix Children's Hospital, Groningen Research Institute for Asthma and COPD, Groningen, the Netherlands; Department of Pulmonary Diseases, Groningen, the Netherlands; Department of Pathology & Medical Biology, University Medical Centre Groningen, University of Groningen, Groningen, the Netherlands
| | - Brian G G Oliver
- School of Life Sciences, The University of Technology Sydney, Sydney, NSW, Australia; Respiratory Cellular and Molecular Biology Group, Woolcock Institute of Medical Research, The University of Sydney, Sydney, NSW, Australia
| | - Maarten van den Berge
- Division of Paediatric Pulmonology, Beatrix Children's Hospital, Groningen Research Institute for Asthma and COPD, Groningen, the Netherlands; Department of Pulmonary Diseases, Groningen, the Netherlands
| | - Alen Faiz
- Respiratory Bioinformatics and Molecular Biology Group, Ultimo, NSW, Australia; School of Life Sciences, The University of Technology Sydney, Sydney, NSW, Australia; Respiratory Cellular and Molecular Biology Group, Woolcock Institute of Medical Research, The University of Sydney, Sydney, NSW, Australia; Division of Paediatric Pulmonology, Beatrix Children's Hospital, Groningen Research Institute for Asthma and COPD, Groningen, the Netherlands; Department of Pulmonary Diseases, Groningen, the Netherlands.
| |
Collapse
|
7
|
Dapas M, Thompson EE, Wentworth-Sheilds W, Clay S, Visness CM, Calatroni A, Sordillo JE, Gold DR, Wood RA, Makhija M, Khurana Hershey GK, Sherenian MG, Gruchalla RS, Gill MA, Liu AH, Kim H, Kattan M, Bacharier LB, Rastogi D, Altman MC, Busse WW, Becker PM, Nicolae D, O’Connor GT, Gern JE, Jackson DJ, Ober C. Multi-omic association study identifies DNA methylation-mediated genotype and smoking exposure effects on lung function in children living in urban settings. PLoS Genet 2023; 19:e1010594. [PMID: 36638096 PMCID: PMC9879483 DOI: 10.1371/journal.pgen.1010594] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 01/26/2023] [Accepted: 12/23/2022] [Indexed: 01/14/2023] Open
Abstract
Impaired lung function in early life is associated with the subsequent development of chronic respiratory disease. Most genetic associations with lung function have been identified in adults of European descent and therefore may not represent those most relevant to pediatric populations and populations of different ancestries. In this study, we performed genome-wide association analyses of lung function in a multiethnic cohort of children (n = 1,035) living in low-income urban neighborhoods. We identified one novel locus at the TDRD9 gene in chromosome 14q32.33 associated with percent predicted forced expiratory volume in one second (FEV1) (p = 2.4x10-9; βz = -0.31, 95% CI = -0.41- -0.21). Mendelian randomization and mediation analyses revealed that this genetic effect on FEV1 was partially mediated by DNA methylation levels at this locus in airway epithelial cells, which were also associated with environmental tobacco smoke exposure (p = 0.015). Promoter-enhancer interactions in airway epithelial cells revealed chromatin interaction loops between FEV1-associated variants in TDRD9 and the promoter region of the PPP1R13B gene, a stimulator of p53-mediated apoptosis. Expression of PPP1R13B in airway epithelial cells was significantly associated the FEV1 risk alleles (p = 1.3x10-5; β = 0.12, 95% CI = 0.06-0.17). These combined results highlight a potential novel mechanism for reduced lung function in urban youth resulting from both genetics and smoking exposure.
Collapse
Affiliation(s)
- Matthew Dapas
- Department of Human Genetics, University of Chicago, Chicago Illinois, United States of America
| | - Emma E. Thompson
- Department of Human Genetics, University of Chicago, Chicago Illinois, United States of America
| | | | - Selene Clay
- Department of Human Genetics, University of Chicago, Chicago Illinois, United States of America
| | | | | | - Joanne E. Sordillo
- Department of Population Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Diane R. Gold
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Robert A. Wood
- Department of Pediatrics, Johns Hopkins University Medical Center, Baltimore, Maryland, United States of America
| | - Melanie Makhija
- Division of Allergy and Immunology, Ann & Robert H. Lurie Children’s Hospital, Chicago, Illinois, United States of America
| | - Gurjit K. Khurana Hershey
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
- Division of Asthma Research, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Michael G. Sherenian
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
- Division of Asthma Research, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Rebecca S. Gruchalla
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Michelle A. Gill
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Andrew H. Liu
- Department of Allergy and Immunology, Children’s Hospital Colorado, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Haejin Kim
- Department of Medicine, Henry Ford Health System, Detroit, Michigan, United States of America
| | - Meyer Kattan
- Columbia University College of Physicians and Surgeons, New York, New York, United States of America
| | - Leonard B. Bacharier
- Monroe Carell Jr. Children’s Hospital at Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Deepa Rastogi
- Children’s National Health System, Washington, District of Columbia, United States of America
| | - Matthew C. Altman
- Department of Allergy and Infectious Diseases, University of Washington, Seattle, Washington, United States of America
| | - William W. Busse
- Department of Pediatrics and Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Patrice M. Becker
- National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, United States of America
| | - Dan Nicolae
- Department of Statistics, University of Chicago, Chicago, Illinois, United States of America
| | - George T. O’Connor
- Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - James E. Gern
- Department of Pediatrics and Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Daniel J. Jackson
- Department of Pediatrics and Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Carole Ober
- Department of Human Genetics, University of Chicago, Chicago Illinois, United States of America
| |
Collapse
|
8
|
The establishment of COPD organoids to study host-pathogen interaction reveals enhanced viral fitness of SARS-CoV-2 in bronchi. Nat Commun 2022; 13:7635. [PMID: 36496442 PMCID: PMC9735280 DOI: 10.1038/s41467-022-35253-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 11/22/2022] [Indexed: 12/13/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is characterised by airflow limitation and infective exacerbations, however, in-vitro model systems for the study of host-pathogen interaction at the individual level are lacking. Here, we describe the establishment of nasopharyngeal and bronchial organoids from healthy individuals and COPD that recapitulate disease at the individual level. In contrast to healthy organoids, goblet cell hyperplasia and reduced ciliary beat frequency were observed in COPD organoids, hallmark features of the disease. Single-cell transcriptomics uncovered evidence for altered cellular differentiation trajectories in COPD organoids. SARS-CoV-2 infection of COPD organoids revealed more productive replication in bronchi, the key site of infection in severe COVID-19. Viral and bacterial exposure of organoids induced greater pro-inflammatory responses in COPD organoids. In summary, we present an organoid model that recapitulates the in vivo physiological lung microenvironment at the individual level and is amenable to the study of host-pathogen interaction and emerging infectious disease.
Collapse
|
9
|
Delhove J, Alawami M, Macowan M, Lester SE, Nguyen PT, Jersmann HPA, Reynolds PN, Roscioli E. Organotypic sinonasal airway culture systems are predictive of the mucociliary phenotype produced by bronchial airway epithelial cells. Sci Rep 2022; 12:19225. [PMID: 36357550 PMCID: PMC9648462 DOI: 10.1038/s41598-022-23667-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 11/03/2022] [Indexed: 11/12/2022] Open
Abstract
Differentiated air-liquid interface models are the current standard to assess the mucociliary phenotype using clinically-derived samples in a controlled environment. However, obtaining basal progenitor airway epithelial cells (AEC) from the lungs is invasive and resource-intensive. Hence, we applied a tissue engineering approach to generate organotypic sinonasal AEC (nAEC) epithelia to determine whether they are predictive of bronchial AEC (bAEC) models. Basal progenitor AEC were isolated from healthy participants using a cytological brushing method and differentiated into epithelia on transwells until the mucociliary phenotype was observed. Tissue architecture was assessed using H&E and alcian blue/Verhoeff-Van Gieson staining, immunofluorescence (for cilia via acetylated α-tubulin labelling) and scanning electron microscopy. Differentiation and the formation of tight-junctions were monitored over the culture period (day 1-32) by quantifying trans-epithelial electrical resistance. End point (day 32) tight junction protein expression was assessed using Western blot analysis of ZO-1, Occludin-1 and Claudin-1. Reverse transcription qPCR-array was used to assess immunomodulatory and autophagy-specific transcript profiles. All outcome measures were assessed using R-statistical software. Mucociliary architecture was comparable for nAEC and bAEC-derived cultures, e.g. cell density P = 0.55, epithelial height P = 0.88 and cilia abundance P = 0.41. Trans-epithelial electrical resistance measures were distinct from day 1-14, converged over days 16-32, and were statistically similar over the entire culture period (global P < 0.001). This agreed with end-point (day 32) measures of tight junction protein abundance which were non-significant for each analyte (P > 0.05). Transcript analysis for inflammatory markers demonstrated significant variation between nAEC and bAEC epithelial cultures, and favoured increased abundance in the nAEC model (e.g. TGFβ and IL-1β; P < 0.05). Conversely, the abundance of autophagy-related transcripts were comparable and the range of outcome measures for either model exhibited a considerably more confined uncertainty distribution than those observed for the inflammatory markers. Organotypic air-liquid interface models of nAEC are predictive of outcomes related to barrier function, mucociliary architecture and autophagy gene activity in corresponding bAEC models. However, inflammatory markers exhibited wide variation which may be explained by the sentinel immunological surveillance role of the sinonasal epithelium.
Collapse
Affiliation(s)
- Juliette Delhove
- grid.1010.00000 0004 1936 7304Adelaide Medical School, University of Adelaide, Adelaide, SA Australia ,grid.1694.aRespiratory and Sleep Medicine, Women’s and Children’s Hospital, Adelaide, SA Australia
| | - Moayed Alawami
- grid.1010.00000 0004 1936 7304Adelaide Medical School, University of Adelaide, Adelaide, SA Australia ,grid.460761.20000 0001 0323 4206Respiratory Department, Lyell McEwin Hospital, Adelaide, SA Australia
| | - Matthew Macowan
- grid.1010.00000 0004 1936 7304Adelaide Medical School, University of Adelaide, Adelaide, SA Australia ,grid.1002.30000 0004 1936 7857Department of Immunology and Pathology, Monash University, Melbourne, VIC Australia
| | - Susan E. Lester
- grid.1010.00000 0004 1936 7304Adelaide Medical School, University of Adelaide, Adelaide, SA Australia ,grid.278859.90000 0004 0486 659XDepartment of Rheumatology, The Queen Elizabeth Hospital, Adelaide, SA Australia
| | - Phan T. Nguyen
- grid.1010.00000 0004 1936 7304Adelaide Medical School, University of Adelaide, Adelaide, SA Australia ,grid.416075.10000 0004 0367 1221Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, SA Australia
| | - Hubertus P. A. Jersmann
- grid.1010.00000 0004 1936 7304Adelaide Medical School, University of Adelaide, Adelaide, SA Australia ,grid.416075.10000 0004 0367 1221Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, SA Australia
| | - Paul N. Reynolds
- grid.1010.00000 0004 1936 7304Adelaide Medical School, University of Adelaide, Adelaide, SA Australia ,grid.416075.10000 0004 0367 1221Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, SA Australia
| | - Eugene Roscioli
- grid.1010.00000 0004 1936 7304Adelaide Medical School, University of Adelaide, Adelaide, SA Australia ,grid.416075.10000 0004 0367 1221Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, SA Australia ,Adelaide Health and Medical Science, Building, Corner of North Terrace and George St, Adelaide, SA 5005 Australia
| |
Collapse
|
10
|
Johnson RK, Manke J, Campbell M, Armstrong M, Boorgula MP, Pinheiro G, Santana CVN, Mathias RA, Barnes KC, Cruz A, Reisdorph N, Figueiredo CA. Lipid mediators are detectable in the nasal epithelium and differ by asthma status in female subjects. J Allergy Clin Immunol 2022; 150:965-971.e8. [PMID: 35304161 PMCID: PMC9475490 DOI: 10.1016/j.jaci.2022.02.026] [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: 11/05/2021] [Revised: 01/13/2022] [Accepted: 02/24/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Lipid mediators, bioactive products of polyunsaturated fatty acid metabolism, contribute to inflammation initiation and resolution in allergic diseases; however, their presence in lung-related biosamples has not been fully described. OBJECTIVE We aimed to quantify lipid mediators in the nasal airway epithelium and characterize preliminary associations with asthma. METHODS Using liquid chromatography-mass spectrometry, we conducted a pilot study to quantify 56 lipid mediators from nasal epithelial samples collected from 11 female participants of an outpatient asthma clinic and community controls (aged 30-55 years). We examined the presence of each compound using descriptive statistics to test whether lipid mediators could distinguish subjects with asthma (n = 8) from control subjects (n = 3) using linear regression and partial least squares discriminant analysis. RESULTS Fifteen lipid mediators were detectable in all samples, including resolvin (Rv) D5 (RvD5), with the highest median concentrations (in pg/μg protein) of 13-HODE (126.481), 15-HETE (32.869), and 13-OxoODE (13.251). From linear regression adjusted for age, prostaglandin E2 (PGE2) had a trend (P < .1) for higher concentrations in patients with severe asthma compared to controls (mean difference, 0.95; 95% confidence interval, -0.04 to 1.95). Asthma patients had higher scores on principal component 3 compared to controls (mean difference, 2.42; 95% confidence interval, 0.89 to 3.96), which represented lower levels of proresolving 15-HEPE, 19,20-DiHDPA, RvD5, 14-HDHA, 17-HDHA, and 13-HOTrE. Most of these compounds were best at discriminating asthma cases from controls in partial least squares discriminant analysis. CONCLUSION Lipid mediators are detectable in the nasal epithelium, and their levels distinguish asthma cases from controls.
Collapse
Affiliation(s)
- Randi K Johnson
- Division of Biomedical Informatics and Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colo.
| | - Jonathan Manke
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colo
| | - Monica Campbell
- Division of Biomedical Informatics and Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colo
| | - Michael Armstrong
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colo
| | - Meher Preethi Boorgula
- Division of Biomedical Informatics and Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colo
| | - Gabriela Pinheiro
- Federal University of Bahia and Fundação Program for Control of Asthma in Bahia (ProAR), Salvador, Brazil
| | - Cinthia Vila Nova Santana
- Federal University of Bahia and Fundação Program for Control of Asthma in Bahia (ProAR), Salvador, Brazil
| | - Rasika A Mathias
- Department of Medicine, Division of Allergy & Clinical Immunology, Johns Hopkins University, Baltimore, Md
| | - Kathleen C Barnes
- Division of Biomedical Informatics and Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colo
| | - Alvaro Cruz
- Federal University of Bahia and Fundação Program for Control of Asthma in Bahia (ProAR), Salvador, Brazil
| | - Nichole Reisdorph
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colo
| | - Camila A Figueiredo
- Federal University of Bahia and Fundação Program for Control of Asthma in Bahia (ProAR), Salvador, Brazil; Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| |
Collapse
|
11
|
Johansen MD, Mahbub RM, Idrees S, Nguyen DH, Miemczyk S, Pathinayake P, Nichol K, Hansbro NG, Gearing LJ, Hertzog PJ, Gallego-Ortega D, Britton WJ, Saunders BM, Wark PA, Faiz A, Hansbro PM. Increased SARS-CoV-2 Infection, Protease, and Inflammatory Responses in Chronic Obstructive Pulmonary Disease Primary Bronchial Epithelial Cells Defined with Single-Cell RNA Sequencing. Am J Respir Crit Care Med 2022; 206:712-729. [PMID: 35549656 PMCID: PMC9799113 DOI: 10.1164/rccm.202108-1901oc] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 05/12/2022] [Indexed: 01/01/2023] Open
Abstract
Rationale: Patients with chronic obstructive pulmonary disease (COPD) develop more severe coronavirus disease (COVID-19); however, it is unclear whether they are more susceptible to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and what mechanisms are responsible for severe disease. Objectives: To determine whether SARS-CoV-2 inoculated primary bronchial epithelial cells (pBECs) from patients with COPD support greater infection and elucidate the effects and mechanisms involved. Methods: We performed single-cell RNA sequencing analysis on differentiated pBECs from healthy subjects and patients with COPD 7 days after SARS-CoV-2 inoculation. We correlated changes with viral titers, proinflammatory responses, and IFN production. Measurements and Main Results: Single-cell RNA sequencing revealed that COPD pBECs had 24-fold greater infection than healthy cells, which was supported by plaque assays. Club/goblet and basal cells were the predominant populations infected and expressed mRNAs involved in viral replication. Proteases involved in SARS-CoV-2 entry/infection (TMPRSS2 and CTSB) were increased, and protease inhibitors (serpins) were downregulated more so in COPD. Inflammatory cytokines linked to COPD exacerbations and severe COVID-19 were increased, whereas IFN responses were blunted. Coexpression analysis revealed a prominent population of club/goblet cells with high type 1/2 IFN responses that were important drivers of immune responses to infection in both healthy and COPD pBECs. Therapeutic inhibition of proteases and inflammatory imbalances reduced viral titers and cytokine responses, particularly in COPD pBECs. Conclusions: COPD pBECs are more susceptible to SARS-CoV-2 infection because of increases in coreceptor expression and protease imbalances and have greater inflammatory responses. A prominent cluster of IFN-responsive club/goblet cells emerges during infection, which may be important drivers of immunity. Therapeutic interventions suppress SARS-CoV-2 replication and consequent inflammation.
Collapse
Affiliation(s)
- Matt D. Johansen
- Faculty of Science, School of Life Sciences, Centre for Inflammation, Centenary Institute, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Rashad M. Mahbub
- Faculty of Science, School of Life Sciences, Centre for Inflammation, Centenary Institute, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Sobia Idrees
- Faculty of Science, School of Life Sciences, Centre for Inflammation, Centenary Institute, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Duc H. Nguyen
- Faculty of Science, School of Life Sciences, Centre for Inflammation, Centenary Institute, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Stefan Miemczyk
- Faculty of Science, School of Life Sciences, Centre for Inflammation, Centenary Institute, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Prabuddha Pathinayake
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia
| | - Kristy Nichol
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia
| | - Nicole G. Hansbro
- Faculty of Science, School of Life Sciences, Centre for Inflammation, Centenary Institute, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Linden J. Gearing
- Department of Molecular and Translational Sciences, School of Clinical Sciences at Monash Health, Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Monash University, Clayton, Victoria, Australia
| | - Paul J. Hertzog
- Department of Molecular and Translational Sciences, School of Clinical Sciences at Monash Health, Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Monash University, Clayton, Victoria, Australia
| | - David Gallego-Ortega
- Faculty of Engineering and Information Technology, School of Biomedical Engineering, Centre for Single Cell Technology, University of Technology Sydney, Ultimo, New South Wales, Australia
- Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
- St. Vincent’s Clinical School, Faculty of Medicine, University of New South Wales Sydney, Kensington, New South Wales, Australia; and
| | - Warwick J. Britton
- Centenary Institute, University of Sydney and Department of Clinical Immunology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Bernadette M. Saunders
- Faculty of Science, School of Life Sciences, Centre for Inflammation, Centenary Institute, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Peter A. Wark
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia
| | - Alen Faiz
- Faculty of Science, School of Life Sciences, Centre for Inflammation, Centenary Institute, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Philip M. Hansbro
- Faculty of Science, School of Life Sciences, Centre for Inflammation, Centenary Institute, University of Technology Sydney, Sydney, New South Wales, Australia
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia
| |
Collapse
|
12
|
Early transcriptional responses of bronchial epithelial cells to whole cigarette smoke mirror those of in-vivo exposed human bronchial mucosa. Respir Res 2022; 23:227. [PMID: 36056356 PMCID: PMC9440516 DOI: 10.1186/s12931-022-02150-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 08/16/2022] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Despite the well-known detrimental effects of cigarette smoke (CS), little is known about the complex gene expression dynamics in the early stages after exposure. This study aims to investigate early transcriptomic responses following CS exposure of airway epithelial cells in culture and compare these to those found in human CS exposure studies. METHODS Primary bronchial epithelial cells (PBEC) were differentiated at the air-liquid interface (ALI) and exposed to whole CS. Bulk RNA-sequencing was performed at 1 h, 4 h, and 24 h hereafter, followed by differential gene expression analysis. Results were additionally compared to data retrieved from human CS studies. RESULTS ALI-PBEC gene expression in response to CS was most significantly changed at 4 h after exposure. Early transcriptomic changes (1 h, 4 h post CS exposure) were related to oxidative stress, xenobiotic metabolism, higher expression of immediate early genes and pro-inflammatory pathways (i.e., Nrf2, AP-1, AhR). At 24 h, ferroptosis-associated genes were significantly increased, whereas PRKN, involved in removing dysfunctional mitochondria, was downregulated. Importantly, the transcriptome dynamics of the current study mirrored in-vivo human studies of acute CS exposure, chronic smokers, and inversely mirrored smoking cessation. CONCLUSION These findings show that early after CS exposure xenobiotic metabolism and pro-inflammatory pathways were activated, followed by activation of the ferroptosis-related cell death pathway. Moreover, significant overlap between these transcriptomic responses in the in-vitro model and human in-vivo studies was found, with an early response of ciliated cells. These results provide validation for the use of ALI-PBEC cultures to study the human lung epithelial response to inhaled toxicants.
Collapse
|
13
|
van den Berge M, Faiz A. Transcriptome Based Signatures: The Future Biomarkers in Obstructive Pulmonary Diseases Such as Asthma and COPD? Am J Respir Crit Care Med 2021; 205:139-140. [PMID: 34793289 PMCID: PMC8787239 DOI: 10.1164/rccm.202110-2353ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Maarten van den Berge
- University of Groningen, University Medical Center Groningen, Department of Pulmonary Diseases, Groningen, Netherlands.,University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, Netherlands;
| | - Alen Faiz
- University of Technology Sydney, 1994, Respiratory Bioinformatics and Molecular Biology (RBMB), School of Life Sciences, Sydney, New South Wales, Australia
| |
Collapse
|
14
|
Soliai MM, Kato A, Helling BA, Stanhope CT, Norton JE, Naughton KA, Klinger AI, Thompson EE, Clay SM, Kim S, Celedón JC, Gern JE, Jackson DJ, Altman MC, Kern RC, Tan BK, Schleimer RP, Nicolae DL, Pinto JM, Ober C. Multi-omics colocalization with genome-wide association studies reveals a context-specific genetic mechanism at a childhood onset asthma risk locus. Genome Med 2021; 13:157. [PMID: 34629083 PMCID: PMC8504130 DOI: 10.1186/s13073-021-00967-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 09/10/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Genome-wide association studies (GWASs) have identified thousands of variants associated with asthma and other complex diseases. However, the functional effects of most of these variants are unknown. Moreover, GWASs do not provide context-specific information on cell types or environmental factors that affect specific disease risks and outcomes. To address these limitations, we used an upper airway epithelial cell (AEC) culture model to assess transcriptional and epigenetic responses to rhinovirus (RV), an asthma-promoting pathogen, and provide context-specific functional annotations to variants discovered in GWASs of asthma. METHODS Genome-wide genetic, gene expression, and DNA methylation data in vehicle- and RV-treated upper AECs were collected from 104 individuals who had a diagnosis of airway disease (n=66) or were healthy participants (n=38). We mapped cis expression and methylation quantitative trait loci (cis-eQTLs and cis-meQTLs, respectively) in each treatment condition (RV and vehicle) in AECs from these individuals. A Bayesian test for colocalization between AEC molecular QTLs and adult onset asthma and childhood onset asthma GWAS SNPs, and a multi-ethnic GWAS of asthma, was used to assign the function to variants associated with asthma. We used Mendelian randomization to demonstrate DNA methylation effects on gene expression at asthma colocalized loci. RESULTS Asthma and allergic disease-associated GWAS SNPs were specifically enriched among molecular QTLs in AECs, but not in GWASs from non-immune diseases, and in AEC eQTLs, but not among eQTLs from other tissues. Colocalization analyses of AEC QTLs with asthma GWAS variants revealed potential molecular mechanisms of asthma, including QTLs at the TSLP locus that were common to both the RV and vehicle treatments and to both childhood onset and adult onset asthma, as well as QTLs at the 17q12-21 asthma locus that were specific to RV exposure and childhood onset asthma, consistent with clinical and epidemiological studies of these loci. CONCLUSIONS This study provides evidence of functional effects for asthma risk variants in AECs and insight into RV-mediated transcriptional and epigenetic response mechanisms that modulate genetic effects in the airway and risk for asthma.
Collapse
Affiliation(s)
- Marcus M Soliai
- Departments of Human Genetics, University of Chicago, Chicago, IL, USA.
- Committee on Genetics, Genomics and Systems Biology, University of Chicago, Chicago, IL, USA.
| | - Atsushi Kato
- Departments of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Britney A Helling
- Departments of Human Genetics, University of Chicago, Chicago, IL, USA
| | | | - James E Norton
- Departments of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | - Aiko I Klinger
- Departments of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Emma E Thompson
- Departments of Human Genetics, University of Chicago, Chicago, IL, USA
| | - Selene M Clay
- Departments of Human Genetics, University of Chicago, Chicago, IL, USA
| | - Soyeon Kim
- Division of Pediatric Pulmonary Medicine, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
| | - Juan C Celedón
- Division of Pediatric Pulmonary Medicine, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
| | - James E Gern
- Department of Pediatrics, University of Wisconsin, School of Medicine and Public Health, Madison, WI, 53706, USA
| | - Daniel J Jackson
- Department of Pediatrics, University of Wisconsin, School of Medicine and Public Health, Madison, WI, 53706, USA
| | - Matthew C Altman
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, WA, USA
- Systems Immunology Program, Benaroya Research Institute, Seattle, WA, USA
| | - Robert C Kern
- Department of Otolaryngology-Head and Neck Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Bruce K Tan
- Department of Otolaryngology-Head and Neck Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Robert P Schleimer
- Departments of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Dan L Nicolae
- Department of Statistics, University of Chicago, Chicago, IL, USA
| | - Jayant M Pinto
- Department of Surgery, University of Chicago, Chicago, IL, USA
| | - Carole Ober
- Departments of Human Genetics, University of Chicago, Chicago, IL, USA.
- Committee on Genetics, Genomics and Systems Biology, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
15
|
Citgez E, van der Palen J, van der Valk P, Kerstjens HAM, Brusse-Keizer M. Stability in eosinophil categorisation during subsequent severe exacerbations of COPD. BMJ Open Respir Res 2021; 8:8/1/e000960. [PMID: 34376399 PMCID: PMC8354268 DOI: 10.1136/bmjresp-2021-000960] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 06/27/2021] [Indexed: 12/03/2022] Open
Abstract
Background The blood eosinophil count has been shown to be a promising biomarker for establishing personalised treatment strategies to reduce corticosteroid use, either inhaled or systemic, in chronic obstructive pulmonary disease (COPD). Eosinophil levels seem relatively stable over time in stable state, but little is known whether this is also true in subsequent severe acute exacerbations of COPD (AECOPD). Aims and objectives To determine the stability in eosinophil categorisation between two subsequent severe AECOPDs employing frequently used cut-off levels. Methods During two subsequent severe AECOPDs, blood eosinophil counts were determined at admission to the hospital in 237 patients in the Cohort of Mortality and Inflammation in COPD Study. The following four cut-off levels were analysed: absolute counts of eosinophils ≥0.2×10⁹/L (200 cells/µL) and ≥0.3×10⁹/L (300 cells/µL) and relative eosinophil percentage of ≥2% and ≥3% of total leucocyte count. Categorisations were considered stable if during the second AECOPD their blood eosinophil status led to the same classification: eosinophilic or not. Results Depending on the used cut-off, the overall stability in eosinophil categorisation varied between 70% and 85% during two subsequent AECOPDs. From patients who were eosinophilic at the first AECOPD, 34%–45% remained eosinophilic at the subsequent AECOPD, while 9%–21% of patients being non-eosinophilic at the first AECOPD became eosinophilic at the subsequent AECOPD. Conclusions The eosinophil variability leads to category changes in subsequent AECOPDs, which limits the eosinophil categorisation stability. Therefore, measurement of eosinophils at each new exacerbation seems warranted.
Collapse
Affiliation(s)
- Emanuel Citgez
- Department of Pulmonary Medicine, Medisch Spectrum Twente, Enschede, The Netherlands .,Department of Research Methodology, Measurement, and Data analysis, University of Twente, Enschede, The Netherlands
| | - Job van der Palen
- Department of Research Methodology, Measurement, and Data analysis, University of Twente, Enschede, The Netherlands.,Medical School Twente, Medisch Spectrum Twente, Enschede, The Netherlands
| | - Paul van der Valk
- Department of Pulmonary Medicine, Medisch Spectrum Twente, Enschede, The Netherlands
| | - Huib A M Kerstjens
- Department of Pulmonary Medicine, and Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | | |
Collapse
|
16
|
Faiz A, Rathnayake SNH, Ten Hacken NHT, Guryev V, van den Berge M, Pouwels SD. Single-nucleotide polymorphism rs2070600 regulates AGER splicing and the sputum levels of the COPD biomarker soluble receptor for advanced glycation end-products. ERJ Open Res 2021; 7:00947-2020. [PMID: 34195255 PMCID: PMC8236754 DOI: 10.1183/23120541.00947-2020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 04/24/2021] [Indexed: 11/05/2022] Open
Abstract
The COPD susceptibility SNP rs2070600 affects the levels of the COPD biomarker sRAGE in sputum as well as splicing of AGER. Moreover, @PouwelsScience et al. demonstrate large differences in sRAGE levels between serum and sputum. https://bit.ly/3t0pJtK.
Collapse
Affiliation(s)
- Alen Faiz
- Respiratory Bioinformatics and Molecular Biology Group, University of Technology Sydney, Sydney, Australia
| | - Senani N H Rathnayake
- Respiratory Bioinformatics and Molecular Biology Group, University of Technology Sydney, Sydney, Australia
| | - Nick H T Ten Hacken
- Dept of Pulmonary Diseases, University Medical Center Groningen, Groningen, The Netherlands
| | - Victor Guryev
- European Research Institute for the Biology of Ageing, Groningen, The Netherlands
| | - Maarten van den Berge
- Dept of Pulmonary Diseases, University Medical Center Groningen, Groningen, The Netherlands.,GRIAC Research Institute, University of Groningen, Groningen, The Netherlands
| | - Simon D Pouwels
- Dept of Pulmonary Diseases, University Medical Center Groningen, Groningen, The Netherlands.,GRIAC Research Institute, University of Groningen, Groningen, The Netherlands.,Dept of Pathology and Medical Biology, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
17
|
Reddy KD, Lan A, Boudewijn IM, Rathnayake SNH, Koppelman GH, Oliver BG, van den Berge M, Faiz A. Current-Smoking alters Gene Expression and DNA Methylation in the Nasal Epithelium of Asthmatics. Am J Respir Cell Mol Biol 2021; 65:366-377. [PMID: 33989148 DOI: 10.1165/rcmb.2020-0553oc] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Current-smoking contributes to worsened asthma prognosis, more severe symptoms and limits the beneficial effects of corticosteroids. As the nasal epithelium can reflect smoking-induced changes in the lower airways, it is a relevant source to investigate changes in gene expression and DNA methylation. This study explores gene expression and DNA methylation changes in current and ex-smokers with asthma. Matched gene expression and epigenome-wide DNA methylation samples collected from nasal brushings of 55 patients enrolled in a clinical trial investigation of current and ex-smoker asthma patients were analysed. Differential gene expression and DNA methylation analyses were conducted comparing current- vs ex-smokers. Expression quantitative trait methylation (eQTM) analysis was completed to explore smoking relevant genes by CpG sites that differ between current and ex-smokers. To investigate the relevance of the smoking-associated DNA methylation changes for the lower airways, significant CpG sites were explored in bronchial biopsies from patients who had stopped smoking. 809 genes and 18,814 CpG sites were differentially associated with current-smoking in the nose. The cis-eQTM analysis uncovered 171 CpG sites whose methylation status associated with smoking-related gene expression, including AHRR, ALDH3A1, CYP1A1 and CYP1B1. Methylation status of CpG sites altered by current-smoking reversed with one-year smoking cessation. We confirm current-smoking alters epigenetic patterns and affects gene expression in the nasal epithelium of asthma patients, which is partially reversible in bronhcial biopsies after smoking cessation. We demonstrate the ability to discern molecular changes in the nasal epithelium, presenting this as a tool in future investigations into disease-relevant effects of tobacco smoke.
Collapse
Affiliation(s)
- Karosham D Reddy
- Woolcock Institute of Medical Research, 104349, Cell Biology, Glebe, New South Wales, Australia.,University of Technology Sydney, 1994, School of Life Sciences, Ultimo, New South Wales, Australia;
| | - Andy Lan
- University of Groningen, 3647, Department of Pulmonary Diseases, Groningen, Netherlands.,University of Groningen, 3647, GRIAC Research Institute, Groningen, Netherlands
| | - Ilse M Boudewijn
- University of Groningen, 3647, Department of Pulmonary Diseases, Groningen, Netherlands.,University of Groningen, 3647, GRIAC Research Insitute, Groningen, Netherlands
| | - Senani N H Rathnayake
- University of Technology Sydney, 1994, Respiratory Bioinformatics and Molecular Biology (RBMB) group,, Sydney, New South Wales, Australia
| | - Gerard H Koppelman
- University of Groningen, 3647, University Medical Center Groningen, Department of Pulmonology and Pediatric Allergy, Beatrix Children's Hospital, Groningen, Netherlands.,University of Groningen, 3647, University Medical Center Groningen, GRIAC research institute, Groningen, Netherlands
| | - Brian G Oliver
- Woolcock Institute of Medical Research, 104349, Glebe, New South Wales, Australia.,University of Technology Sydney, 1994, School of Medical and Molecular Biosciences, Sydney, New South Wales, Australia
| | - Maarten van den Berge
- University of Groningen, 3647, University Medical Center, Department of Pulmonary Diseases, Groningen, Netherlands.,University Medical Center Göttingen, 84922, Groningen Research Institute for Asthma and COPD (GRIAC), Gottingen, Germany
| | - Alen Faiz
- University of Technology Sydney, 1994, Respiratory Bioinformatics and Molecular Biology (RBMB), School of Life Sciences, Sydney, New South Wales, Australia
| | | |
Collapse
|
18
|
Awatade NT, Wong SL, Capraro A, Pandzic E, Slapetova I, Zhong L, Turgutoglu N, Fawcett LK, Whan RM, Jaffe A, Waters SA. Significant functional differences in differentiated Conditionally Reprogrammed (CRC)- and Feeder-free Dual SMAD inhibited-expanded human nasal epithelial cells. J Cyst Fibros 2021; 20:364-371. [PMID: 33414087 DOI: 10.1016/j.jcf.2020.12.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 12/16/2020] [Accepted: 12/20/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Patient-derived airway cells differentiated at Air Liquid Interface (ALI) are valuable models for Cystic fibrosis (CF) precision therapy. Different culture expansion methods have been established to extend expansion capacity of airway basal cells, while retaining functional airway epithelium physiology. Considerable variation in response to CFTR modulators is observed in cultures even within the same CFTR genotype and despite the use of similar ALI culture techniques. We aimed to address culture expansion method impact on differentiation. METHODS Nasal epithelial brushings from 14 individuals (CF=9; non-CF=5) were collected, then equally divided and expanded under conditional reprogramming culture (CRC) and feeder-serum-free "dual-SMAD inhibition" (SMADi) methods. Expanded cells from each culture were differentiated with proprietary PneumaCult™-ALI media. Morphology (Immunofluorescence), global proteomics (LC-MS/MS) and function (barrier integrity, cilia motility, and ion transport) were compared in CRCALI and SMADiALI under basal and CFTR corrector treated (VX-809) conditions. RESULTS No significant difference in the structural morphology or baseline global proteomics profile were observed. Barrier integrity and cilia motility were significantly different, despite no difference in cell junction morphology or cilia abundance. Epithelial Sodium Channels and Calcium-activated Chloride Channel activity did not differ but CFTR mediated chloride currents were significantly reduced in SMADiALI compare to their CRCALI counterparts. CONCLUSION Alteration of cellular physiological function in vitro were more prominent than structural and differentiation potential in airway ALI. Since initial expansion culture conditions significantly influence CFTR activity, this could lead to false conclusions if data from different labs are compared against each other without specific reference ranges.
Collapse
Affiliation(s)
- Nikhil T Awatade
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia; Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC), University of New South Wales and Sydney Children's Hospital, Sydney, NSW, Australia
| | - Sharon L Wong
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia; Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC), University of New South Wales and Sydney Children's Hospital, Sydney, NSW, Australia
| | - Alexander Capraro
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia; Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC), University of New South Wales and Sydney Children's Hospital, Sydney, NSW, Australia
| | - Elvis Pandzic
- Biomedical Imaging Facility, University of New South Wales, Sydney, NSW, Australia
| | - Iveta Slapetova
- Biomedical Imaging Facility, University of New South Wales, Sydney, NSW, Australia
| | - Ling Zhong
- Bioanalytical Mass Spectrometry Facility, University of New South Wales, Sydney, NSW, Australia
| | - Nihan Turgutoglu
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia; Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC), University of New South Wales and Sydney Children's Hospital, Sydney, NSW, Australia
| | - Laura K Fawcett
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia; Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC), University of New South Wales and Sydney Children's Hospital, Sydney, NSW, Australia; Department of Respiratory Medicine, Sydney Children's Hospital, Sydney, NSW, Australia
| | - Renee M Whan
- Biomedical Imaging Facility, University of New South Wales, Sydney, NSW, Australia
| | - Adam Jaffe
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia; Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC), University of New South Wales and Sydney Children's Hospital, Sydney, NSW, Australia; Department of Respiratory Medicine, Sydney Children's Hospital, Sydney, NSW, Australia
| | - Shafagh A Waters
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia; Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC), University of New South Wales and Sydney Children's Hospital, Sydney, NSW, Australia; Department of Respiratory Medicine, Sydney Children's Hospital, Sydney, NSW, Australia.
| |
Collapse
|
19
|
Pouwels SD, Wiersma VR, Fokkema IE, Berg M, Ten Hacken NHT, van den Berge M, Heijink I, Faiz A. Acute cigarette smoke-induced eQTL affects formyl peptide receptor expression and lung function. Respirology 2020; 26:233-240. [PMID: 33078507 PMCID: PMC7983955 DOI: 10.1111/resp.13960] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 09/23/2020] [Accepted: 09/24/2020] [Indexed: 02/07/2023]
Abstract
Background and objective Cigarette smoking is one of the most prevalent causes of preventable deaths worldwide, leading to chronic diseases, including chronic obstructive pulmonary disease (COPD). Cigarette smoke is known to induce significant transcriptional modifications throughout the respiratory tract. However, it is largely unknown how genetic profiles influence the smoking‐related transcriptional changes and how changes in gene expression translate into altered alveolar epithelial repair responses. Methods We performed a candidate‐based acute cigarette smoke‐induced eQTL study, investigating the association between SNP and differential gene expression of FPR family members in bronchial epithelial cells isolated 24 h after smoking and after 48 h without smoking. The effects FPR1 on lung epithelial integrity and repair upon damage in the presence and absence of cigarette smoke were studied in CRISPR‐Cas9‐generated lung epithelial knockout cells. Results One significant (FDR < 0.05) inducible eQTL (rs3212855) was identified that induced a >2‐fold change in gene expression. The minor allele of rs3212855 was associated with significantly higher gene expression of FPR1, FPR2 and FPR3 upon smoking. Importantly, the minor allele of rs3212855 was also associated with lower lung function. Alveolar epithelial FPR1 knockout cells were protected against CSE‐induced reduction in repair capacity upon wounding. Conclusion We identified a novel smoking‐related inducible eQTL that is associated with a smoke‐induced increase in the expression of FPR1, FPR2 and FPR3, and with lowered lung function. in vitro FPR1 down‐regulation protects against smoke‐induced reduction in lung epithelial repair.
Collapse
Affiliation(s)
- Simon D Pouwels
- Department of Pathology and Medical Biology, University Medical Center Groningen (UMCG), University of Groningen, Groningen, The Netherlands.,Department of Pulmonology, University Medical Center Groningen (UMCG), University of Groningen, Groningen, The Netherlands.,Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen (UMCG), University of Groningen, Groningen, The Netherlands
| | - Valerie R Wiersma
- Department of Hematology, Cancer Research Center Groningen, University Medical Center Groningen (UMCG), University of Groningen, Groningen, The Netherlands
| | - Immeke E Fokkema
- Department of Pathology and Medical Biology, University Medical Center Groningen (UMCG), University of Groningen, Groningen, The Netherlands
| | - Marijn Berg
- Department of Pathology and Medical Biology, University Medical Center Groningen (UMCG), University of Groningen, Groningen, The Netherlands.,Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen (UMCG), University of Groningen, Groningen, The Netherlands
| | - Nick H T Ten Hacken
- Department of Pulmonology, University Medical Center Groningen (UMCG), University of Groningen, Groningen, The Netherlands
| | - Maarten van den Berge
- Department of Pulmonology, University Medical Center Groningen (UMCG), University of Groningen, Groningen, The Netherlands.,Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen (UMCG), University of Groningen, Groningen, The Netherlands
| | - Irene Heijink
- Department of Pathology and Medical Biology, University Medical Center Groningen (UMCG), University of Groningen, Groningen, The Netherlands.,Department of Pulmonology, University Medical Center Groningen (UMCG), University of Groningen, Groningen, The Netherlands.,Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen (UMCG), University of Groningen, Groningen, The Netherlands
| | - Alen Faiz
- Respiratory Bioinformatics and Molecular Biology, University of Technology Sydney, Sydney, NSW, Australia
| |
Collapse
|
20
|
Ghosh B, Park B, Bhowmik D, Nishida K, Lauver M, Putcha N, Gao P, Ramanathan M, Hansel N, Biswal S, Sidhaye VK. Strong correlation between air-liquid interface cultures and in vivo transcriptomics of nasal brush biopsy. Am J Physiol Lung Cell Mol Physiol 2020; 318:L1056-L1062. [PMID: 32233789 PMCID: PMC7272738 DOI: 10.1152/ajplung.00050.2020] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 03/24/2020] [Accepted: 03/24/2020] [Indexed: 02/07/2023] Open
Abstract
Air-liquid interface (ALI) cultures are ex vivo models that are used extensively to study the epithelium of patients with chronic respiratory diseases. However, the in vitro conditions impose a milieu different from that encountered in the patient in vivo, and the degree to which this alters gene expression remains unclear. In this study we employed RNA sequencing to compare the transcriptome of fresh brushings of nasal epithelial cells with that of ALI-cultured epithelial cells from the same patients. We observed a strong correlation between cells cultured at the ALI and cells obtained from the brushed nasal epithelia: 96% of expressed genes showed similar expression profiles, although there was greater similarity between the brushed samples. We observed that while the ALI model provides an excellent representation of the in vivo airway epithelial transcriptome for mechanistic studies, several pathways are affected by the change in milieu.
Collapse
Affiliation(s)
- Baishakhi Ghosh
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Bongsoo Park
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Debarshi Bhowmik
- Department of Biology, Johns Hopkins University, Baltimore, Maryland
| | - Kristine Nishida
- Department of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Molly Lauver
- Department of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Nirupama Putcha
- Department of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Peisong Gao
- Division of Allergy and Clinical Immunology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Murugappan Ramanathan
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Nadia Hansel
- Department of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Shyam Biswal
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Venkataramana K Sidhaye
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
- Department of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| |
Collapse
|
21
|
Genetic profiling for disease stratification in chronic obstructive pulmonary disease and asthma. Curr Opin Pulm Med 2020; 25:317-322. [PMID: 30762612 DOI: 10.1097/mcp.0000000000000568] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW In asthma and chronic obstructive pulmonary disease (COPD), the movement towards genetic profiling with a push towards 'personalized medicine' has been hindered by complex environment--gene interactions and lack of tools to identify clear causal genetic traits. In this review, we will discuss the need for genetic profiling in asthma and COPD, what methods are currently used in the clinics and the recent finding using new sequencing methods. RECENT FINDINGS Over the past 10-15 years, genome-wide association studies analysis of common variants has provide little in the way of new genetic profiling markers for asthma and COPD. Whole exome/genome sequencing has provided a new method to identify lowly abundant alleles, which might have a much higher impact. Although, low population numbers due to high costs has hindered early studies, recent studies have reached genome wide significance. SUMMARY The use of genetic profiling of COPD in the clinic is current limited to the identification of Alpha-1 antitrypsin deficiency, while being absent in asthma. Advances in sequencing technology provide new avenues to identify disease causes or therapy response altering variants that in the short-term will allow for the development of screening procedures for disease to identify patients at risk of developing asthma or COPD.
Collapse
|
22
|
Bartel S, La Grutta S, Cilluffo G, Perconti G, Bongiovanni A, Giallongo A, Behrends J, Kruppa J, Hermann S, Chiang D, Pfaffl MW, Krauss‐Etschmann S. Human airway epithelial extracellular vesicle miRNA signature is altered upon asthma development. Allergy 2020; 75:346-356. [PMID: 31386204 DOI: 10.1111/all.14008] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 06/18/2019] [Accepted: 07/05/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND miRNAs are master regulators of signaling pathways critically involved in asthma and are transferred between cells in extracellular vesicles (EV). We aimed to investigate whether the miRNA content of EV secreted by primary normal human bronchial epithelial cells (NHBE) is altered upon asthma development. METHODS NHBE cells were cultured at air-liquid interface and treated with interleukin (IL)-13 to induce an asthma-like phenotype. EV isolations by precipitation from basal culture medium or apical surface wash were characterized by nanoparticle tracking analysis, transmission electron microscopy, and Western blot, and EV-associated miRNAs were identified by a RT-qPCR-based profiling. Significant candidates were confirmed in EVs isolated by size-exclusion chromatography from nasal lavages of children with mild-to-moderate (n = 8) or severe asthma (n = 9), and healthy controls (n = 9). RESULTS NHBE cells secrete EVs to the apical and basal side. 47 miRNAs were expressed in EVs and 16 thereof were significantly altered in basal EV upon IL-13 treatment. Expression of miRNAs could be confirmed in EVs from human nasal lavages. Of note, levels of miR-92b, miR-210, and miR-34a significantly correlated with lung function parameters in children (FEV1 FVC%pred and FEF25-75%pred ), thus lower sEV-miRNA levels in nasal lavages associated with airway obstruction. Subsequent ingenuity pathway analysis predicted the miRNAs to regulate Th2 polarization and dendritic cell maturation. CONCLUSION Our data indicate that secretion of miRNAs in EVs from the airway epithelium, in particular miR-34a, miR-92b, and miR-210, might be involved in the early development of a Th2 response in the airways and asthma.
Collapse
Affiliation(s)
- Sabine Bartel
- Early Life Origins of Chronic Lung Disease Research Center Borstel, Leibniz Lung Center, Member of the German Center for Lung Research (DZL) and the Airway Research Center North (ARCN) Borstel Germany
- Department of Pathology and Medical Biology GRIAC Research Institute, University of Groningen, University Medical Center Groningen Groningen The Netherlands
| | - Stefania La Grutta
- Institute for Research and Biomedical Innovation (IRIB) National Research Council Palermo Italy
| | - Giovanna Cilluffo
- Institute for Research and Biomedical Innovation (IRIB) National Research Council Palermo Italy
| | - Giovanni Perconti
- Institute for Research and Biomedical Innovation (IRIB) National Research Council Palermo Italy
| | - Antonella Bongiovanni
- Institute for Research and Biomedical Innovation (IRIB) National Research Council Palermo Italy
| | - Agata Giallongo
- Institute for Research and Biomedical Innovation (IRIB) National Research Council Palermo Italy
| | - Jochen Behrends
- Core Facility Fluorescence Cytometry Research Center Borstel, Leibniz Lung Center Borstel Germany
| | - Jochen Kruppa
- Institute of Biometry and Clinical Epidemiology Charité ‐ Universitätsmedizin Berlin, Humboldt‐Universität zu Berlin, Berlin Institute of Health Berlin Germany
- Berlin Institute of Health (BIH) Berlin Germany
| | - Stefanie Hermann
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan Technical University of Munich Munich Germany
| | - Dapi Chiang
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan Technical University of Munich Munich Germany
| | - Michael W. Pfaffl
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan Technical University of Munich Munich Germany
| | - Susanne Krauss‐Etschmann
- Early Life Origins of Chronic Lung Disease Research Center Borstel, Leibniz Lung Center, Member of the German Center for Lung Research (DZL) and the Airway Research Center North (ARCN) Borstel Germany
- Institute for Experimental Medicine Christian‐Albrechts‐Universität zu Kiel Kiel Germany
| |
Collapse
|
23
|
Qi C, Jiang Y, Yang IV, Forno E, Wang T, Vonk JM, Gehring U, Smit HA, Milanzi EB, Carpaij OA, Berg M, Hesse L, Brouwer S, Cardwell J, Vermeulen CJ, Acosta-Pérez E, Canino G, Boutaoui N, van den Berge M, Teichmann SA, Nawijn MC, Chen W, Celedón JC, Xu CJ, Koppelman GH. Nasal DNA methylation profiling of asthma and rhinitis. J Allergy Clin Immunol 2020; 145:1655-1663. [PMID: 31953105 DOI: 10.1016/j.jaci.2019.12.911] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 11/27/2019] [Accepted: 12/17/2019] [Indexed: 10/25/2022]
Abstract
BACKGROUND Epigenetic signatures in the nasal epithelium, which is a primary interface with the environment and an accessible proxy for the bronchial epithelium, might provide insights into mechanisms of allergic disease. OBJECTIVE We aimed to identify and interpret methylation signatures in nasal epithelial brushes associated with rhinitis and asthma. METHODS Nasal epithelial brushes were obtained from 455 children at the 16-year follow-up of the Dutch Prevention and Incidence of Asthma and Mite Allergy birth cohort study. Epigenome-wide association studies were performed on children with asthma, rhinitis, and asthma and/or rhinitis (AsRh) by using logistic regression, and the top results were replicated in 2 independent cohorts of African American and Puerto Rican children. Significant CpG sites were related to environmental exposures (pets, active and passive smoking, and molds) during secondary school and were correlated with gene expression by RNA-sequencing (n = 244). RESULTS The epigenome-wide association studies identified CpG sites significantly associated with rhinitis (n = 81) and AsRh (n = 75), but not with asthma. We significantly replicated 62 of 81 CpG sites with rhinitis and 60 of 75 with AsRh, as well as 1 CpG site with asthma. Methylation of cg03565274 was negatively associated with AsRh and positively associated with exposure to pets during secondary school. DNA methylation signals associated with AsRh were mainly driven by specific IgE-positive subjects. DNA methylation related to gene transcripts that were enriched for immune pathways and expressed in immune and epithelial cells. Nasal CpG sites performed well in predicting AsRh. CONCLUSIONS We identified replicable DNA methylation profiles of asthma and rhinitis in nasal brushes. Exposure to pets may affect nasal epithelial methylation in relation to asthma and rhinitis.
Collapse
Affiliation(s)
- Cancan Qi
- Department of Pediatric Pulmonology and Pediatric Allergy, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Gronigen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Yale Jiang
- Division of Pulmonary Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pa; Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pa; School of Medicine, Tsinghua University, Beijing, China
| | - Ivana V Yang
- Department of Medicine, University of Colorado, Aurora, Colo
| | - Erick Forno
- Division of Pulmonary Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pa; Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pa
| | - Ting Wang
- Division of Pulmonary Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pa; Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pa
| | - Judith M Vonk
- Gronigen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Department of Epidemiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Ulrike Gehring
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, The Netherlands
| | - Henriëtte A Smit
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Edith B Milanzi
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, The Netherlands
| | - Orestes A Carpaij
- Gronigen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Department of Pulmonary Diseases, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Marijn Berg
- Gronigen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Department of Pathology & Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Laura Hesse
- Gronigen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Department of Pathology & Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Sharon Brouwer
- Gronigen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Department of Pathology & Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | | | - Cornelis J Vermeulen
- Gronigen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Department of Pulmonary Diseases, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Edna Acosta-Pérez
- Behavioral Sciences Research Institute, University of Puerto Rico, San Juan, Puerto Rico
| | - Glorisa Canino
- Behavioral Sciences Research Institute, University of Puerto Rico, San Juan, Puerto Rico
| | - Nadia Boutaoui
- Division of Pulmonary Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pa; Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pa
| | - Maarten van den Berge
- Gronigen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Department of Pulmonary Diseases, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Sarah A Teichmann
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom; Theory of Condensed Matter Group, Cavendish Laboratory/Department of Physics, University of Cambridge, Cambridge, United Kingdom
| | - Martijn C Nawijn
- Gronigen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Department of Pathology & Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Wei Chen
- Division of Pulmonary Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pa; Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pa
| | - Juan C Celedón
- Division of Pulmonary Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pa; Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pa
| | - Cheng-Jian Xu
- Department of Pediatric Pulmonology and Pediatric Allergy, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Gronigen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Department of Gastroenterology, Hepatology and Endocrinology, Centre for Individualised Infection Medicine, CiiM, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany; TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
| | - Gerard H Koppelman
- Department of Pediatric Pulmonology and Pediatric Allergy, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Gronigen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
24
|
Boudewijn IM, Lan A, Faiz A, Cox CA, Brouwer S, Schokker S, Vroegop SJ, Nawijn MC, Woodruff PG, Christenson SA, Hagedoorn P, Frijlink HW, Choy DF, Brouwer U, Wisman M, Postma DS, Fingleton J, Beasley R, van den Berge M, Guryev V. Nasal gene expression changes with inhaled corticosteroid treatment in asthma. Allergy 2020; 75:191-194. [PMID: 31230369 DOI: 10.1111/all.13952] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 06/01/2019] [Accepted: 06/14/2019] [Indexed: 12/15/2022]
Affiliation(s)
- Ilse M. Boudewijn
- Department of Pulmonary Diseases University Medical Center Groningen, University of Groningen Groningen The Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen University of Groningen Groningen The Netherlands
| | - Andy Lan
- Department of Pulmonary Diseases University Medical Center Groningen, University of Groningen Groningen The Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen University of Groningen Groningen The Netherlands
- Department of Pathology, Section Medical Biology, University Medical Center Groningen University of Groningen Groningen The Netherlands
| | - Alen Faiz
- Department of Pulmonary Diseases University Medical Center Groningen, University of Groningen Groningen The Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen University of Groningen Groningen The Netherlands
- Department of Pathology, Section Medical Biology, University Medical Center Groningen University of Groningen Groningen The Netherlands
| | - Claire A. Cox
- Department of Pulmonary Diseases University Medical Center Groningen, University of Groningen Groningen The Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen University of Groningen Groningen The Netherlands
| | - Sharon Brouwer
- Department of Pathology, Section Medical Biology, University Medical Center Groningen University of Groningen Groningen The Netherlands
| | - Siebrig Schokker
- Department of Pulmonary Diseases Martini Hospital Groningen the Netherlands
| | | | - Martijn C. Nawijn
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen University of Groningen Groningen The Netherlands
- Department of Pathology, Section Medical Biology, University Medical Center Groningen University of Groningen Groningen The Netherlands
| | - Prescott G. Woodruff
- Department of Medicine, Division of Pulmonary, Critical Care, Allergy, and Sleep Medicine University of California San Francisco California USA
- Cardiovascular Research Institute University of California San Francisco California USA
| | - Stephanie A. Christenson
- Department of Medicine, Division of Pulmonary, Critical Care, Allergy, and Sleep Medicine University of California San Francisco California USA
- Cardiovascular Research Institute University of California San Francisco California USA
| | - Paul Hagedoorn
- Department of Pharmaceutical Technology and Biopharmacy University of Groningen Groningen The Netherlands
| | - Henderik W. Frijlink
- Department of Pharmaceutical Technology and Biopharmacy University of Groningen Groningen The Netherlands
| | | | - Uilke Brouwer
- Department of Pathology, Section Medical Biology, University Medical Center Groningen University of Groningen Groningen The Netherlands
| | - Marissa Wisman
- Department of Pathology, Section Medical Biology, University Medical Center Groningen University of Groningen Groningen The Netherlands
| | - Dirkje S. Postma
- Department of Pulmonary Diseases University Medical Center Groningen, University of Groningen Groningen The Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen University of Groningen Groningen The Netherlands
| | - James Fingleton
- The Medical Research Institute of New Zealand Wellington New Zealand
- The Capital & Coast District Health Board Wellington New Zealand
- Victoria University of Wellington New Zealand
| | - Richard Beasley
- The Medical Research Institute of New Zealand Wellington New Zealand
- The Capital & Coast District Health Board Wellington New Zealand
- Victoria University of Wellington New Zealand
| | - Maarten van den Berge
- Department of Pulmonary Diseases University Medical Center Groningen, University of Groningen Groningen The Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen University of Groningen Groningen The Netherlands
| | - Victor Guryev
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen University of Groningen Groningen The Netherlands
- European Research Institute for the Biology of Ageing University Medical Center Groningen, University of Groningen Groningen The Netherlands
| |
Collapse
|
25
|
Imkamp K, Bernal V, Grzegorzcyk M, Horvatovich P, Vermeulen CJ, Heijink IH, Guryev V, Kerstjens HAM, van den Berge M, Faiz A. Gene network approach reveals co-expression patterns in nasal and bronchial epithelium. Sci Rep 2019; 9:15835. [PMID: 31676779 PMCID: PMC6825243 DOI: 10.1038/s41598-019-50963-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 09/13/2019] [Indexed: 12/20/2022] Open
Abstract
Nasal gene expression profiling is a new approach to investigate the airway epithelium as a biomarker to study the activity and treatment responses of obstructive pulmonary diseases. We investigated to what extent gene expression profiling of nasal brushings is similar to that of bronchial brushings. We performed genome wide gene expression profiling on matched nasal and bronchial epithelial brushes from 77 respiratory healthy individuals. To investigate differences and similarities among regulatory modules, network analysis was performed on correlated, differentially expressed and smoking-related genes using Gaussian Graphical Models. Between nasal and bronchial brushes, 619 genes were correlated and 1692 genes were differentially expressed (false discovery rate <0.05, |Fold-change|>2). Network analysis of correlated genes showed pro-inflammatory pathways to be similar between the two locations. Focusing on smoking-related genes, cytochrome-P450 pathway related genes were found to be similar, supporting the concept of a detoxifying response to tobacco exposure throughout the airways. In contrast, cilia-related pathways were decreased in nasal compared to bronchial brushes when focusing on differentially expressed genes. Collectively, while there are substantial differences in gene expression between nasal and bronchial brushes, we also found similarities, especially in the response to the external factors such as smoking.
Collapse
Affiliation(s)
- Kai Imkamp
- University of Groningen, University Medical Center Groningen, Department of Pulmonology, Groningen, The Netherlands. .,University of Groningen, University Medical Center Groningen, GRIAC (Groningen Research Institute for Asthma and COPD), Groningen, The Netherlands.
| | - Victor Bernal
- University of Groningen, Bernoulli Institute (JBI), Groningen, The Netherlands.,University of Groningen, Department of Pharmacy, Analytical Biochemistry, Groningen, The Netherlands
| | - Marco Grzegorzcyk
- University of Groningen, Bernoulli Institute (JBI), Groningen, The Netherlands
| | - Peter Horvatovich
- University of Groningen, Department of Pharmacy, Analytical Biochemistry, Groningen, The Netherlands
| | - Cornelis J Vermeulen
- University of Groningen, University Medical Center Groningen, Department of Pulmonology, Groningen, The Netherlands.,University of Groningen, University Medical Center Groningen, GRIAC (Groningen Research Institute for Asthma and COPD), Groningen, The Netherlands
| | - Irene H Heijink
- University of Groningen, University Medical Center Groningen, Department of Pulmonology, Groningen, The Netherlands.,University of Groningen, University Medical Center Groningen, GRIAC (Groningen Research Institute for Asthma and COPD), Groningen, The Netherlands.,University of Groningen, University Medical Center Groningen, Department of Pathology & Medical Biology, section Medical Biology, Groningen, The Netherlands
| | - Victor Guryev
- University of Groningen, University Medical Center Groningen, GRIAC (Groningen Research Institute for Asthma and COPD), Groningen, The Netherlands.,European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Huib A M Kerstjens
- University of Groningen, University Medical Center Groningen, Department of Pulmonology, Groningen, The Netherlands.,University of Groningen, University Medical Center Groningen, GRIAC (Groningen Research Institute for Asthma and COPD), Groningen, The Netherlands
| | - Maarten van den Berge
- University of Groningen, University Medical Center Groningen, Department of Pulmonology, Groningen, The Netherlands.,University of Groningen, University Medical Center Groningen, GRIAC (Groningen Research Institute for Asthma and COPD), Groningen, The Netherlands
| | - Alen Faiz
- University of Groningen, University Medical Center Groningen, Department of Pulmonology, Groningen, The Netherlands.,University of Groningen, University Medical Center Groningen, GRIAC (Groningen Research Institute for Asthma and COPD), Groningen, The Netherlands.,University of Groningen, University Medical Center Groningen, Department of Pathology & Medical Biology, section Medical Biology, Groningen, The Netherlands.,University of Technology Sydney, Respiratory Bioinformatics and Molecular Biology (RBMB), School of life sciences, Sydney, Australia.,Woolcock Emphysema Centre, Woolcock Institute of Medical Research, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
26
|
Zuo H, Faiz A, van den Berge M, Mudiyanselage SNHR, Borghuis T, Timens W, Nikolaev VO, Burgess JK, Schmidt M. Cigarette smoke exposure alters phosphodiesterases in human structural lung cells. Am J Physiol Lung Cell Mol Physiol 2019; 318:L59-L64. [PMID: 31664853 DOI: 10.1152/ajplung.00319.2019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Cigarette smoke (CS), a highly complex mixture containing more than 4,000 compounds, causes aberrant cell responses leading to tissue damage around the airways and alveoli, which underlies various lung diseases. Phosphodiesterases (PDEs) are a family of enzymes that hydrolyze cyclic nucleotides. PDE inhibition induces bronchodilation, reduces the activation and recruitment of inflammatory cells, and the release of various cytokines. Currently, the selective PDE4 inhibitor roflumilast is an approved add-on treatment for patients with severe chronic obstructive pulmonary disease with chronic bronchitis and a history of frequent exacerbations. Additional selective PDE inhibitors are being tested in preclinical and clinical studies. However, the effect of chronic CS exposure on the expression of PDEs is unknown. Using mRNA isolated from nasal and bronchial brushes and lung tissues of never smokers and current smokers, we compared the gene expression of 25 PDE coding genes. Additionally, the expression and distribution of PDE3A and PDE4D in human lung tissues was examined. This study reveals that chronic CS exposure modulates the expression of various PDE members. Thus, CS exposure may change the levels of intracellular cyclic nucleotides and thereby impact the efficiency of PDE-targeted therapies.
Collapse
Affiliation(s)
- Haoxiao Zuo
- University of Groningen, Department of Molecular Pharmacology, Groningen, The Netherlands.,University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands.,Institute of Experimental Cardiovascular Research, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Alen Faiz
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands.,University of Groningen, Department of Pulmonary Diseases, University Medical Center Groningen, Groningen, The Netherlands.,Emphysema Center, Woolcock Institute of Medical Research, The University of Sydney, Glebe, New South Wales, Australia.,Faculty of Science, University of Technology Sydney, Respiratory Bioinformatics and Molecular Biology, Ultimo, New South Wales, Australia
| | - Maarten van den Berge
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands.,University of Groningen, Department of Pulmonary Diseases, University Medical Center Groningen, Groningen, The Netherlands
| | | | - Theo Borghuis
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands.,University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands
| | - Wim Timens
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands.,University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands
| | - Viacheslav O Nikolaev
- Institute of Experimental Cardiovascular Research, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany.,German Center for Cardiovascular Research, Hamburg, Germany
| | - Janette K Burgess
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands.,University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands
| | - Martina Schmidt
- University of Groningen, Department of Molecular Pharmacology, Groningen, The Netherlands.,University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands
| |
Collapse
|
27
|
Faiz A, van den Berge M, Vermeulen CJ, Ten Hacken NHT, Guryev V, Pouwels SD. AGER expression and alternative splicing in bronchial biopsies of smokers and never smokers. Respir Res 2019; 20:70. [PMID: 30971245 PMCID: PMC6456980 DOI: 10.1186/s12931-019-1038-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 03/29/2019] [Indexed: 02/07/2023] Open
Abstract
Cigarette smoking is one of the major risk factors for the development of chronic obstructive pulmonary disease (COPD). Evidence is accumulating that Receptor for Advanced Glycation-End products (RAGE)-signaling is a key pathway in the pathophysiology of COPD. To date, it is unknown how smoking affects RAGE expression. In the current study, we investigated the effect of smoking on AGER, the gene encoding RAGE, expression and on alternative splicing of AGER. To this end, we conducted RNA-Seq on bronchial biopsies for asymptomatic smokers (n = 36) and never smokers (n = 40). Total AGER gene expression was accessed using DESeq2, while alternative splicing was investigated by measuring the number of specific split reads spanning exon-exon junctions and the total split reads. One of the major isoforms of RAGE is endogenous soluble (es) RAGE, an anti-inflammatory decoy receptor, making up for approximately 10% of the total amount of soluble (s)RAGE. We found that smokers show decreased total gene expression of AGER in bronchial biopsies, while the relative abundance of the esRAGE isoform is increased. Furthermore, no difference in the serum levels of total sRAGE were observed between smokers and non-smokers. Our data indicates that smoking initiates a protective anti-inflammatory mechanism with decreased expression of the pro-inflammatory gene AGER and increased relative abundance of the anti-inflammatory isoform esRAGE.
Collapse
Affiliation(s)
- Alen Faiz
- Department of Pathology & Medical Biology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands.,Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, Univesity of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands.,Department of Pulmonary Diseases, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Maarten van den Berge
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, Univesity of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands.,Department of Pulmonary Diseases, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Cornelis J Vermeulen
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, Univesity of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands.,Department of Pulmonary Diseases, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Nick H T Ten Hacken
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, Univesity of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands.,Department of Pulmonary Diseases, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Victor Guryev
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, Univesity of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands.,European Research Institute for the Biology of Ageing, University of Groningen, Groningen, The Netherlands
| | - Simon D Pouwels
- Department of Pathology & Medical Biology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands. .,Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, Univesity of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands.
| |
Collapse
|
28
|
Morrow JD, Chase RP, Parker MM, Glass K, Seo M, Divo M, Owen CA, Castaldi P, DeMeo DL, Silverman EK, Hersh CP. RNA-sequencing across three matched tissues reveals shared and tissue-specific gene expression and pathway signatures of COPD. Respir Res 2019; 20:65. [PMID: 30940135 PMCID: PMC6446359 DOI: 10.1186/s12931-019-1032-z] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Accepted: 03/25/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Multiple gene expression studies have been performed separately in peripheral blood, lung, and airway tissues to study COPD. We performed RNA-sequencing gene expression profiling of large-airway epithelium, alveolar macrophage and peripheral blood samples from the same subset of COPD cases and controls from the COPDGene study who underwent bronchoscopy at a single center. Using statistical and gene set enrichment approaches, we sought to improve the understanding of COPD by studying gene sets and pathways across these tissues, beyond the individual genomic determinants. METHODS We performed differential expression analysis using RNA-seq data obtained from 63 samples from 21 COPD cases and controls (includes four non-smokers) via the R package DESeq2. We tested associations between gene expression and variables related to lung function, smoking history, and CT scan measures of emphysema and airway disease. We examined the correlation of differential gene expression across the tissues and phenotypes, hypothesizing that this would reveal preserved and private gene expression signatures. We performed gene set enrichment analyses using curated databases and findings from prior COPD studies to provide biological and disease relevance. RESULTS The known smoking-related genes CYP1B1 and AHRR were among the top differential expression results for smoking status in the large-airway epithelium data. We observed a significant overlap of genes primarily across large-airway and macrophage results for smoking and airway disease phenotypes. We did not observe specific genes differentially expressed in all three tissues for any of the phenotypes. However, we did observe hemostasis and immune signaling pathways in the overlaps across all three tissues for emphysema, and amyloid and telomere-related pathways for smoking. In peripheral blood, the emphysema results were enriched for B cell related genes previously identified in lung tissue studies. CONCLUSIONS Our integrative analyses across COPD-relevant tissues and prior studies revealed shared and tissue-specific disease biology. These replicated and novel findings in the airway and peripheral blood have highlighted candidate genes and pathways for COPD pathogenesis.
Collapse
Affiliation(s)
- Jarrett D Morrow
- Channing Division of Network Medicine, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, MA, 02115, USA.
| | - Robert P Chase
- Channing Division of Network Medicine, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, MA, 02115, USA
| | - Margaret M Parker
- Channing Division of Network Medicine, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, MA, 02115, USA
| | - Kimberly Glass
- Channing Division of Network Medicine, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, MA, 02115, USA
| | - Minseok Seo
- Channing Division of Network Medicine, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, MA, 02115, USA
| | - Miguel Divo
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Caroline A Owen
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Peter Castaldi
- Channing Division of Network Medicine, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, MA, 02115, USA
| | - Dawn L DeMeo
- Channing Division of Network Medicine, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, MA, 02115, USA.,Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Edwin K Silverman
- Channing Division of Network Medicine, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, MA, 02115, USA.,Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Craig P Hersh
- Channing Division of Network Medicine, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, MA, 02115, USA.,Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
| |
Collapse
|
29
|
Pouwels SD, Klont F, Kwiatkowski M, Wiersma VR, Faiz A, van den Berge M, Horvatovich P, Bischoff R, Ten Hacken NHT. Reply to Biswas: Acute and Chronic Effects of Cigarette Smoking on sRAGE. Am J Respir Crit Care Med 2019; 199:806-807. [PMID: 30589564 PMCID: PMC6423096 DOI: 10.1164/rccm.201812-2257le] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
| | - Frank Klont
- 1 University of Groningen Groningen, the Netherlands and
| | | | | | - Alen Faiz
- 1 University of Groningen Groningen, the Netherlands and.,2 University of Technology Sydney Sydney, Australia
| | | | | | | | | |
Collapse
|