1
|
Ishii Y, Orr JC, El Mdawar MB, de Pilger DRB, Pearce DR, Lazarus KA, Graham RE, Nikolić MZ, Ketteler R, Carragher NO, Janes SM, Hynds RE. Compound screening in human airway basal cells identifies Wnt pathway activators as potential pro-regenerative therapies. J Cell Sci 2025; 138:jcs263487. [PMID: 40065746 DOI: 10.1242/jcs.263487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 03/04/2025] [Indexed: 04/15/2025] Open
Abstract
Regeneration of the airway epithelium restores barrier function and mucociliary clearance following lung injury and infection. The mechanisms regulating the proliferation and differentiation of tissue-resident airway basal stem cells remain incompletely understood. To identify compounds that promote human airway basal cell proliferation, we performed phenotype-based compound screening of 1429 compounds (from the ENZO and Prestwick Chemical libraries) in 384-well format using primary cells transduced with lentiviral luciferase. A total of 17 pro-proliferative compounds were validated in independent donor cell cultures, including the antiretroviral therapy agent abacavir and several Wnt signalling pathway-activating compounds. The effects of compounds on proliferation were further explored in colony formation and 3D organoid assays. Structurally and functionally related compounds that more potently induced Wnt pathway activation were investigated. One such compound, 1-azakenpaullone, induced Wnt target gene activation and basal cell proliferation in mice. Our results demonstrate the pro-proliferative effect of small-molecule Wnt pathway activators on airway basal cells. These findings contribute to the rationale to develop novel approaches to modulate Wnt signalling during airway epithelial repair.
Collapse
Affiliation(s)
- Yuki Ishii
- Lungs for Living Research Centre, UCL Respiratory, University College London, London WC1E 6JF, UK
| | - Jessica C Orr
- Lungs for Living Research Centre, UCL Respiratory, University College London, London WC1E 6JF, UK
- Epithelial Cell Biology in ENT Research Group, Developmental Biology and Cancer Department, UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1DZ, UK
| | - Marie-Belle El Mdawar
- Lungs for Living Research Centre, UCL Respiratory, University College London, London WC1E 6JF, UK
| | | | - David R Pearce
- UCL Cancer Institute, University College London, London WC1N 6DD, UK
| | - Kyren A Lazarus
- Lungs for Living Research Centre, UCL Respiratory, University College London, London WC1E 6JF, UK
| | - Rebecca E Graham
- Edinburgh Cancer Research, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, UK
- Cancer Research UK Scotland Centre, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Marko Z Nikolić
- Lungs for Living Research Centre, UCL Respiratory, University College London, London WC1E 6JF, UK
| | - Robin Ketteler
- MRC Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK
| | - Neil O Carragher
- Edinburgh Cancer Research, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, UK
- Cancer Research UK Scotland Centre, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Sam M Janes
- Lungs for Living Research Centre, UCL Respiratory, University College London, London WC1E 6JF, UK
| | - Robert E Hynds
- Epithelial Cell Biology in ENT Research Group, Developmental Biology and Cancer Department, UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1DZ, UK
- UCL Cancer Institute, University College London, London WC1N 6DD, UK
| |
Collapse
|
2
|
Yarlagadda T, Carey A, Bryan E, Huygens F, Yarlagadda P, Maresco-Pennisi D, Coleman A, Cervin A, Spann K. The response of nasal epithelial cells exposed to novel Lactobacillus and alpha-haemolytic Streptococcus isolated from the upper respiratory tract of children. J Appl Microbiol 2025; 136:lxaf071. [PMID: 40107866 DOI: 10.1093/jambio/lxaf071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 01/27/2025] [Accepted: 03/17/2025] [Indexed: 03/22/2025]
Abstract
AIMS To investigate the response of primary nasal epithelial cells (NECs) to novel alpha haemolytic Streptococcus and lactobacilli strains, isolated from the upper respiratory tract of children. METHODS AND RESULTS Submerged cultures of NECs from healthy adult donors were exposed to either novel strains; Lactobacillus rhamnosus D3189, D3160, Streptococcus salivarius D3837; or commercially available probiotic strains L. rhamnosus LB21, S. salivarius K12; or a pathogenic strain (S. pneumoniae 49619). Cytotoxicity (measured through lactate dehydrogenase release) and cytokine release were quantified 24 hours post-exposure. Exposure to novel and commercially available strains did not induce the production of IFN-β, IFN-λ1/3, IL-1β, IL-6, IL-8, or TNF-α production or the release of LDH. Conversely, the pathogenic strain S. pneumoniae 49 619 significantly elevated the expression of IL-1β, IL-8, TNF-α, and LDH in NECs. CONCLUSIONS The findings within this study highlight the non-pathogenic nature of these novel strains and support further investigation of the potential to develop nasally administered probiotics.
Collapse
Affiliation(s)
- Tejasri Yarlagadda
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Queensland University of Technology, Queensland 4000, Australia
| | - Alison Carey
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Queensland University of Technology, Queensland 4000, Australia
| | - Emily Bryan
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Queensland University of Technology, Queensland 4000, Australia
- Faculty of Medicine, University of Queensland Centre for Clinical Research, Queensland 4000, Australia
| | - Flavia Huygens
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Queensland University of Technology, Queensland 4000, Australia
| | - Prasad Yarlagadda
- School of Engineering, University of Southern Queensland, Queensland 4300, Australia
| | - Diane Maresco-Pennisi
- Faculty of Medicine, University of Queensland Centre for Clinical Research, Queensland 4000, Australia
| | - Andrea Coleman
- Faculty of Medicine, University of Queensland Centre for Clinical Research, Queensland 4000, Australia
| | - Anders Cervin
- Faculty of Medicine, University of Queensland Centre for Clinical Research, Queensland 4000, Australia
| | - Kirsten Spann
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Queensland University of Technology, Queensland 4000, Australia
| |
Collapse
|
3
|
Zhao Y, Patel J, Fan J, Wang X, Chen L, Li Y, Luo Z. Integrated analysis reveals that EGR1 promotes epithelial IL33 production in T2 asthma. J Transl Med 2025; 23:203. [PMID: 39966984 PMCID: PMC11837401 DOI: 10.1186/s12967-025-06116-y] [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: 11/04/2024] [Accepted: 01/08/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Airway epithelial cells constitute the first line of defense against external noxious stimuli and play crucial roles in the release of epithelial inflammatory cytokines (IL33, IL25 and TSLP), initiating airway allergic inflammatory diseases such as asthma. IL33 plays critical physiological processes in T2-endotype asthma. However, the mechanisms by which allergen exposure triggers IL33 release from airway epithelial cells remain unclear. METHODS Integrated bioinformatic analysis and transcriptional analysis of bulk RNA-seq and single cell RNA-seq (scRNA-seq) data were used to identify core genes and determine the internal gene network associated with IL33. The expression of EGR1 was subsequently analyzed in vitro in the BEAS-2B cell line and in vivo in a house dust mite (HDM)-induced mouse asthma model. The functional experiments of EGR1 were investigated in vitro via siRNA knockdown and over-expressed plasmid. Chromatin immunoprecipitation (ChIP)-PCR and dual-luciferase reporter assay validation were subsequently performed to investigate the mechanisms by which EGR1 regulates IL33 secretion. RESULTS Bulk RNA-seq and scRNA-seq data identified EGR1 as an epithelial cell-derived gene implicated in IL33 expressions in asthma. The comprehensive analysis of multiple datasets indicated that the high EGR1 expression in epithelial cells may suggest a mechanistic basis of T2-endotype childhood asthma. Moreover, we verified that the expressions of EGR1 in airway epithelial cells were elevated both in vitro and in vivo asthma models. EGR1 regulated the production of IL33. Ultimately, ChIP and luciferase reporter assays confirmed that transcription factor EGR1 directly regulate the transcription of IL33 mRNA. CONCLUSIONS Our integrated bioinformatic analysis elucidated that EGR1 directly regulates the production of IL33 in T2-asthma and provide insights underlying the progression of asthma.
Collapse
Affiliation(s)
- Yan Zhao
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China
- China International Science and Technology Cooperation base of Child development and Critical Disorders, Chongqing, China
| | - Jenil Patel
- Department of Epidemiology, Human Genetics and Environmental Sciences, The University of Texas Health Science Center at Houston (UTHealth Houston) School of Public Health, Dallas, TX, USA
| | - Jinhua Fan
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Xinyang Wang
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Lin Chen
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yuanyuan Li
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China
| | - Zhengxiu Luo
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.
- Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China.
- China International Science and Technology Cooperation base of Child development and Critical Disorders, Chongqing, China.
- Department of Respiratory Medicine, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Children's Hospital of Chongqing Medical University, No.136 Zhongshan 2nd Road, Yuzhong District, Chongqing, 400010, China.
| |
Collapse
|
4
|
Liu YB, Tan XH, Yang HH, Yang JT, Zhang CY, Jin L, Yang NSY, Guan CX, Zhou Y, Liu SK, Xiong JB. Wnt5a-mediated autophagy contributes to the epithelial-mesenchymal transition of human bronchial epithelial cells during asthma. Mol Med 2024; 30:93. [PMID: 38898476 PMCID: PMC11188189 DOI: 10.1186/s10020-024-00862-3] [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: 02/24/2024] [Accepted: 06/12/2024] [Indexed: 06/21/2024] Open
Abstract
BACKGROUND The epithelial-mesenchymal transition (EMT) of human bronchial epithelial cells (HBECs) is essential for airway remodeling during asthma. Wnt5a has been implicated in various lung diseases, while its role in the EMT of HBECs during asthma is yet to be determined. This study sought to define whether Wnt5a initiated EMT, leading to airway remodeling through the induction of autophagy in HBECs. METHODS Microarray analysis was used to investigate the expression change of WNT5A in asthma patients. In parallel, EMT models were induced using 16HBE cells by exposing them to house dust mites (HDM) or interleukin-4 (IL-4), and then the expression of Wnt5a was observed. Using in vitro gain- and loss-of-function approaches via Wnt5a mimic peptide FOXY5 and Wnt5a inhibitor BOX5, the alterations in the expression of the epithelial marker E-cadherin and the mesenchymal marker protein were observed. Mechanistically, the Ca2+/CaMKII signaling pathway and autophagy were evaluated. An autophagy inhibitor 3-MA was used to examine Wnt5a in the regulation of autophagy during EMT. Furthermore, we used a CaMKII inhibitor KN-93 to determine whether Wnt5a induced autophagy overactivation and EMT via the Ca2+/CaMKII signaling pathway. RESULTS Asthma patients exhibited a significant increase in the gene expression of WNT5A compared to the healthy control. Upon HDM and IL-4 treatments, we observed that Wnt5a gene and protein expression levels were significantly increased in 16HBE cells. Interestingly, Wnt5a mimic peptide FOXY5 significantly inhibited E-cadherin and upregulated α-SMA, Collagen I, and autophagy marker proteins (Beclin1 and LC3-II). Rhodamine-phalloidin staining showed that FOXY5 resulted in a rearrangement of the cytoskeleton and an increase in the quantity of stress fibers in 16HBE cells. Importantly, blocking Wnt5a with BOX5 significantly inhibited autophagy and EMT induced by IL-4 in 16HBE cells. Mechanistically, autophagy inhibitor 3-MA and CaMKII inhibitor KN-93 reduced the EMT of 16HBE cells caused by FOXY5, as well as the increase in stress fibers, cell adhesion, and autophagy. CONCLUSION This study illustrates a new link in the Wnt5a-Ca2+/CaMKII-autophagy axis to triggering airway remodeling. Our findings may provide novel strategies for the treatment of EMT-related diseases.
Collapse
Affiliation(s)
- Yu-Biao Liu
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan, 410078, China
| | - Xiao-Hua Tan
- Experimental Center of Medical Morphology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| | - Hui-Hui Yang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan, 410078, China
| | - Jin-Tong Yang
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan, 410078, China
| | - Chen-Yu Zhang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan, 410078, China
| | - Ling Jin
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan, 410078, China
| | - Nan-Shi-Yu Yang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan, 410078, China
| | - Cha-Xiang Guan
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan, 410078, China
| | - Yong Zhou
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan, 410078, China
| | - Shao-Kun Liu
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
- Research Unit of Respiratory Disease, Central South University, Changsha, Hunan, 410011, China.
- Clinical Medical Research Center for Pulmonary and Critical Care Medicine in Hunan Province, Changsha, Hunan, 410011, China.
- Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, Hunan, 410011, China.
| | - Jian-Bing Xiong
- Department of Emergency, Xiangya Hospital, Central South University, Changsha, Hunan, 410078, China.
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410078, China.
| |
Collapse
|
5
|
Szczesny B, Boorgula MP, Chavan S, Campbell M, Johnson RK, Kammers K, Thompson EE, Cox MS, Shankar G, Cox C, Morin A, Lorizio W, Daya M, Kelada SNP, Beaty TH, Doumatey AP, Cruz AA, Watson H, Naureckas ET, Giles BL, Arinola GA, Sogaolu O, Falade AG, Hansel NN, Yang IV, Olopade CO, Rotimi CN, Landis RC, Figueiredo CA, Altman MC, Kenny E, Ruczinski I, Liu AH, Ober C, Taub MA, Barnes KC, Mathias RA. Multi-omics in nasal epithelium reveals three axes of dysregulation for asthma risk in the African Diaspora populations. Nat Commun 2024; 15:4546. [PMID: 38806494 PMCID: PMC11133339 DOI: 10.1038/s41467-024-48507-7] [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: 05/18/2023] [Accepted: 05/02/2024] [Indexed: 05/30/2024] Open
Abstract
Asthma has striking disparities across ancestral groups, but the molecular underpinning of these differences is poorly understood and minimally studied. A goal of the Consortium on Asthma among African-ancestry Populations in the Americas (CAAPA) is to understand multi-omic signatures of asthma focusing on populations of African ancestry. RNASeq and DNA methylation data are generated from nasal epithelium including cases (current asthma, N = 253) and controls (never-asthma, N = 283) from 7 different geographic sites to identify differentially expressed genes (DEGs) and gene networks. We identify 389 DEGs; the top DEG, FN1, was downregulated in cases (q = 3.26 × 10-9) and encodes fibronectin which plays a role in wound healing. The top three gene expression modules implicate networks related to immune response (CEACAM5; p = 9.62 × 10-16 and CPA3; p = 2.39 × 10-14) and wound healing (FN1; p = 7.63 × 10-9). Multi-omic analysis identifies FKBP5, a co-chaperone of glucocorticoid receptor signaling known to be involved in drug response in asthma, where the association between nasal epithelium gene expression is likely regulated by methylation and is associated with increased use of inhaled corticosteroids. This work reveals molecular dysregulation on three axes - increased Th2 inflammation, decreased capacity for wound healing, and impaired drug response - that may play a critical role in asthma within the African Diaspora.
Collapse
Affiliation(s)
- Brooke Szczesny
- Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Meher Preethi Boorgula
- Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Sameer Chavan
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Monica Campbell
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Randi K Johnson
- Department of Epidemiology, Colorado School of Public Health, Aurora, CO, USA
- Quantitative Sciences Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kai Kammers
- Departments of Human Genetics, University of Chicago, Chicago, IL, USA
| | - Emma E Thompson
- Division of Allergy and Infectious Diseases, Dept of Medicine, University of Washington, Seattle, WA, USA
| | - Madison S Cox
- Division of Allergy and Infectious Diseases, Dept of Medicine, University of Washington, Seattle, WA, USA
| | - Gautam Shankar
- Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Corey Cox
- Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Andréanne Morin
- Departments of Human Genetics, University of Chicago, Chicago, IL, USA
| | - Wendy Lorizio
- Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Michelle Daya
- Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Samir N P Kelada
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
- Marsico Lung Institute, University of North Carolina, Chapel Hill, NC, USA
| | - Terri H Beaty
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Ayo P Doumatey
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Alvaro A Cruz
- Fundacao ProAR and Federal University of Bahia, Salvador, Bahia, Brazil
| | - Harold Watson
- Faculty of Medical Sciences, The University of the West Indies, Queen Elizabeth Hospital, St. Michael, Bridgetown, Barbados
| | | | - B Louise Giles
- Departments of Pediatrics, University of Chicago, Chicago, IL, USA
| | - Ganiyu A Arinola
- Department of Immunology, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Olumide Sogaolu
- Department of Medicine, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Adegoke G Falade
- Department of Pediatrics, University of Ibadan, and University College Hospital, Ibadan, Nigeria
| | - Nadia N Hansel
- Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Ivana V Yang
- Departments of Biomedical Informatics and Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | | | - Charles N Rotimi
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - R Clive Landis
- Edmund Cohen Laboratory for Vascular Research, George Alleyne Chronic Disease Research Centre, Caribbean Institute for Health Research, The University of the West Indies, Cave Hill Campus, Wanstead, Barbados
| | - Camila A Figueiredo
- Federal University of Bahia and Funda. Program for Control of Asthma in Bahia (ProAR), Salvador, Brazil
- Instituto de Ciências de Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - Matthew C Altman
- Systems Immunology Program, Benaroya Research Institute, Seattle, WA, 98101, USA
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, WA, 98109, USA
| | - Eimear Kenny
- Center for Genomic Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ingo Ruczinski
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Andrew H Liu
- Department of Pediatrics, Childrens Hospital Colorado and University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Carole Ober
- Departments of Human Genetics, University of Chicago, Chicago, IL, USA
| | - Margaret A Taub
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Kathleen C Barnes
- Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA.
| | - Rasika A Mathias
- Department of Medicine, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
6
|
Kicic-Starcevich E, Hancock DG, Iosifidis T, Agudelo-Romero P, Caparros-Martin JA, Karpievitch YV, Silva D, Turkovic L, Le Souef PN, Bosco A, Martino DJ, Kicic A, Prescott SL, Stick SM. Airway epithelium respiratory illnesses and allergy (AERIAL) birth cohort: study protocol. FRONTIERS IN ALLERGY 2024; 5:1349741. [PMID: 38666051 PMCID: PMC11043573 DOI: 10.3389/falgy.2024.1349741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 04/01/2024] [Indexed: 04/28/2024] Open
Abstract
Introduction Recurrent wheezing disorders including asthma are complex and heterogeneous diseases that affect up to 30% of all children, contributing to a major burden on children, their families, and global healthcare systems. It is now recognized that a dysfunctional airway epithelium plays a central role in the pathogenesis of recurrent wheeze, although the underlying mechanisms are still not fully understood. This prospective birth cohort aims to bridge this knowledge gap by investigating the influence of intrinsic epithelial dysfunction on the risk for developing respiratory disorders and the modulation of this risk by maternal morbidities, in utero exposures, and respiratory exposures in the first year of life. Methods The Airway Epithelium Respiratory Illnesses and Allergy (AERIAL) study is nested within the ORIGINS Project and will monitor 400 infants from birth to 5 years. The primary outcome of the AERIAL study will be the identification of epithelial endotypes and exposure variables that influence the development of recurrent wheezing, asthma, and allergic sensitisation. Nasal respiratory epithelium at birth to 6 weeks, 1, 3, and 5 years will be analysed by bulk RNA-seq and DNA methylation sequencing. Maternal morbidities and in utero exposures will be identified on maternal history and their effects measured through transcriptomic and epigenetic analyses of the amnion and newborn epithelium. Exposures within the first year of life will be identified based on infant medical history as well as on background and symptomatic nasal sampling for viral PCR and microbiome analysis. Daily temperatures and symptoms recorded in a study-specific Smartphone App will be used to identify symptomatic respiratory illnesses. Discussion The AERIAL study will provide a comprehensive longitudinal assessment of factors influencing the association between epithelial dysfunction and respiratory morbidity in early life, and hopefully identify novel targets for diagnosis and early intervention.
Collapse
Affiliation(s)
| | - David G. Hancock
- Wal-yan RespiratoryResearch Centre, Telethon Kids Institute, Perth, WA, Australia
- Department of Respiratory and Sleep Medicine, Perth Children’s Hospital, Nedlands, WA, Australia
- School of Medicine, The University of Western Australia, Nedlands, WA, Australia
| | - Thomas Iosifidis
- Wal-yan RespiratoryResearch Centre, Telethon Kids Institute, Perth, WA, Australia
- School of Population Health, Curtin University, Bentley, WA, Australia
- Centre for Cell Therapy and Regenerative Medicine, School of Medicine, The University of Western Australia, Nedlands, WA, Australia
| | - Patricia Agudelo-Romero
- Wal-yan RespiratoryResearch Centre, Telethon Kids Institute, Perth, WA, Australia
- European Virus Bioinformatics Centre, Jena, Germany
| | | | | | - Desiree Silva
- School of Medicine, The University of Western Australia, Nedlands, WA, Australia
- Telethon Kids Institute, Perth, WA, Australia
- Department of Paediatrics and Neonatology, Joondalup Health Campus, Joondalup, WA, Australia
- School of Medicine and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| | | | - Peter N. Le Souef
- Wal-yan RespiratoryResearch Centre, Telethon Kids Institute, Perth, WA, Australia
- Department of Respiratory and Sleep Medicine, Perth Children’s Hospital, Nedlands, WA, Australia
| | - Anthony Bosco
- School of Population Health, Curtin University, Bentley, WA, Australia
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, AZ, United States
- Department of Immunobiology, The University of Arizona College of Medicine, Tucson, AZ, United States
| | - David J. Martino
- Wal-yan RespiratoryResearch Centre, Telethon Kids Institute, Perth, WA, Australia
| | - Anthony Kicic
- Wal-yan RespiratoryResearch Centre, Telethon Kids Institute, Perth, WA, Australia
- School of Population Health, Curtin University, Bentley, WA, Australia
- Centre for Cell Therapy and Regenerative Medicine, School of Medicine, The University of Western Australia, Nedlands, WA, Australia
| | - Susan L. Prescott
- School of Medicine, The University of Western Australia, Nedlands, WA, Australia
- European Virus Bioinformatics Centre, Jena, Germany
| | - Stephen M. Stick
- Wal-yan RespiratoryResearch Centre, Telethon Kids Institute, Perth, WA, Australia
- Department of Respiratory and Sleep Medicine, Perth Children’s Hospital, Nedlands, WA, Australia
- Centre for Cell Therapy and Regenerative Medicine, School of Medicine, The University of Western Australia, Nedlands, WA, Australia
| |
Collapse
|
7
|
Russell RJ, Boulet LP, Brightling CE, Pavord ID, Porsbjerg C, Dorscheid D, Sverrild A. The airway epithelium: an orchestrator of inflammation, a key structural barrier and a therapeutic target in severe asthma. Eur Respir J 2024; 63:2301397. [PMID: 38453256 PMCID: PMC10991852 DOI: 10.1183/13993003.01397-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 02/15/2024] [Indexed: 03/09/2024]
Abstract
Asthma is a disease of heterogeneous pathology, typically characterised by excessive inflammatory and bronchoconstrictor responses to the environment. The clinical expression of the disease is a consequence of the interaction between environmental factors and host factors over time, including genetic susceptibility, immune dysregulation and airway remodelling. As a critical interface between the host and the environment, the airway epithelium plays an important role in maintaining homeostasis in the face of environmental challenges. Disruption of epithelial integrity is a key factor contributing to multiple processes underlying asthma pathology. In this review, we first discuss the unmet need in asthma management and provide an overview of the structure and function of the airway epithelium. We then focus on key pathophysiological changes that occur in the airway epithelium, including epithelial barrier disruption, immune hyperreactivity, remodelling, mucus hypersecretion and mucus plugging, highlighting how these processes manifest clinically and how they might be targeted by current and novel therapeutics.
Collapse
Affiliation(s)
- Richard J Russell
- Institute for Lung Health, NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
| | | | - Christopher E Brightling
- Institute for Lung Health, NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
| | - Ian D Pavord
- Respiratory Medicine, NIHR Oxford Biomedical Research Centre, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Celeste Porsbjerg
- Department of Respiratory Medicine and Infectious Diseases, Bispebjerg Hospital, Copenhagen University, Copenhagen, Denmark
| | - Del Dorscheid
- Centre for Heart Lung Innovation, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Asger Sverrild
- Department of Respiratory Medicine and Infectious Diseases, Bispebjerg Hospital, Copenhagen University, Copenhagen, Denmark
| |
Collapse
|
8
|
Yarlagadda T, Zhu Y, Snape N, Carey A, Bryan E, Maresco-Pennisi D, Coleman A, Cervin A, Spann K. Lactobacillus rhamnosus dampens cytokine and chemokine secretion from primary human nasal epithelial cells infected with rhinovirus. J Appl Microbiol 2024; 135:lxae018. [PMID: 38268489 DOI: 10.1093/jambio/lxae018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 01/15/2024] [Accepted: 01/22/2024] [Indexed: 01/26/2024]
Abstract
AIMS To investigate the effect of Lactobacillus rhamnosus on viral replication and cellular response to human rhinovirus (HRV) infection, including the secretion of antiviral and inflammatory mediators from well-differentiated nasal epithelial cells (WD-NECs). METHODS AND RESULTS The WD-NECs from healthy adult donors (N = 6) were cultured in vitro, exposed to different strains of L. rhamnosus (D3189, D3160, or LB21), and infected with HRV (RV-A16) after 24 h. Survival and adherence capacity of L. rhamnosus in a NEC environment were confirmed using CFSE-labelled isolates, immunofluorescent staining, and confocal microscopy. Shed virus and viral replication were quantified using TCID50 assays and RT-qPCR, respectively. Cytotoxicity was measured by lactate dehydrogenase (LDH) activity. Pro-inflammatory mediators were measured by multiplex immunoassay, and interferon (IFN)-λ1/3 was measured using a standard ELISA kit. Lactobacillus rhamnosus was able to adhere to and colonize WD-NECs prior to the RV-A16 infection. Lactobacillus rhamnosus did not affect shed RV-A16, viral replication, RV-A16-induced IFN-λ1/3 production, or LDH release. Pre-exposure to L. rhamnosus, particularly D3189, reduced the secretion of RV-A16-induced pro-inflammatory mediators by WD-NECs. CONCLUSIONS These findings demonstrate that L. rhamnosus differentially modulates RV-A16-induced innate inflammatory immune responses in primary NECs from healthy adults.
Collapse
Affiliation(s)
- Tejasri Yarlagadda
- Centre for Immunology and Infection Control, Queensland University of Technology, Brisbane 4000, Australia
| | - Yanshan Zhu
- School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia 4072, Australia
| | - Natale Snape
- University of Queensland Frazer Institute, Woolloongabba 4102, Australia
| | - Alison Carey
- Centre for Immunology and Infection Control, Queensland University of Technology, Brisbane 4000, Australia
| | - Emily Bryan
- Centre for Immunology and Infection Control, Queensland University of Technology, Brisbane 4000, Australia
- Faculty of Medicine, University of Queensland Centre for Clinical Research, Herston 4006, Australia
| | - Diane Maresco-Pennisi
- Faculty of Medicine, University of Queensland Centre for Clinical Research, Herston 4006, Australia
| | - Andrea Coleman
- Faculty of Medicine, University of Queensland Centre for Clinical Research, Herston 4006, Australia
| | - Anders Cervin
- Faculty of Medicine, University of Queensland Centre for Clinical Research, Herston 4006, Australia
| | - Kirsten Spann
- Centre for Immunology and Infection Control, Queensland University of Technology, Brisbane 4000, Australia
| |
Collapse
|
9
|
Huang GX, Hallen NR, Lee M, Zheng K, Wang X, Mandanas MV, Djeddi S, Fernandez D, Hacker J, Ryan T, Bergmark RW, Bhattacharyya N, Lee S, Maxfield AZ, Roditi RE, Buchheit KM, Laidlaw TM, Gern JE, Hallstrand TS, Ray A, Wenzel SE, Boyce JA, Gutierrez-Arcelus M, Barrett NA. Increased epithelial mTORC1 activity in chronic rhinosinusitis with nasal polyps. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.13.562288. [PMID: 37904989 PMCID: PMC10614789 DOI: 10.1101/2023.10.13.562288] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Background The airway epithelium plays a central role in the pathogenesis of chronic respiratory diseases such as asthma and chronic rhinosinusitis with nasal polyps (CRSwNP), but the mechanisms by which airway epithelial cells (EpCs) maintain inflammation are poorly understood. Objective We hypothesized that transcriptomic assessment of sorted airway EpCs across the spectrum of differentiation would allow us to define mechanisms by which EpCs perpetuate airway inflammation. Methods Ethmoid sinus EpCs from adult patients with CRS were sorted into 3 subsets, bulk RNA sequenced, and analyzed for differentially expressed genes and pathways. Single cell RNA-seq (scRNA-seq) datasets from eosinophilic and non-eosinophilic CRSwNP and bulk RNA-seq of EpCs from mild/moderate and severe asthma were assessed. Immunofluorescent staining and ex vivo functional analysis of sinus EpCs were used to validate our findings. Results Analysis within and across purified EpC subsets revealed an enrichment in glycolytic programming in CRSwNP vs CRSsNP. Correlation analysis identified mammalian target of rapamycin complex 1 (mTORC1) as a potential regulator of the glycolytic program and identified EpC expression of cytokines and wound healing genes as potential sequelae. mTORC1 activity was upregulated in CRSwNP, and ex vivo inhibition demonstrated that mTOR is critical for EpC generation of CXCL8, IL-33, and CXCL2. Across patient samples, the degree of glycolytic activity was associated with T2 inflammation in CRSwNP, and with both T2 and non-T2 inflammation in severe asthma. Conclusions Together, these findings highlight a metabolic axis required to support epithelial generation of cytokines critical to both chronic T2 and non-T2 inflammation in CRSwNP and asthma.
Collapse
Affiliation(s)
- George X. Huang
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital; Boston, MA
- Department of Medicine, Harvard Medical School; Boston, MA
| | - Nils R. Hallen
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital; Boston, MA
- Department of Medicine, Harvard Medical School; Boston, MA
| | - Minkyu Lee
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital; Boston, MA
- Department of Medicine, Harvard Medical School; Boston, MA
| | - Kelly Zheng
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital; Boston, MA
- Department of Medicine, Harvard Medical School; Boston, MA
| | - Xin Wang
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital; Boston, MA
- Department of Medicine, Harvard Medical School; Boston, MA
| | | | - Sarah Djeddi
- Division of Immunology, Boston Children’s Hospital; Boston, MA
| | | | - Jonathan Hacker
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital; Boston, MA
- Department of Medicine, Harvard Medical School; Boston, MA
| | - Tessa Ryan
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital; Boston, MA
- Department of Medicine, Harvard Medical School; Boston, MA
| | - Regan W. Bergmark
- Department of Otolaryngology, Head and Neck Surgery, Brigham and Women’s Hospital; Boston, MA
| | - Neil Bhattacharyya
- Department of Otolaryngology, Head and Neck Surgery, Massachusetts Eye and Ear Infirmary; Boston, MA
| | - Stella Lee
- Department of Otolaryngology, Head and Neck Surgery, Brigham and Women’s Hospital; Boston, MA
| | - Alice Z. Maxfield
- Department of Otolaryngology, Head and Neck Surgery, Brigham and Women’s Hospital; Boston, MA
| | - Rachel E. Roditi
- Department of Otolaryngology, Head and Neck Surgery, Brigham and Women’s Hospital; Boston, MA
| | - Kathleen M. Buchheit
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital; Boston, MA
- Department of Medicine, Harvard Medical School; Boston, MA
| | - Tanya M. Laidlaw
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital; Boston, MA
- Department of Medicine, Harvard Medical School; Boston, MA
| | - James E. Gern
- Division of Allergy, Immunology, and Rheumatology, University of Wisconsin School of Medicine and Public Health; Madison, WI
| | - Teal S. Hallstrand
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington Medical Center; Seattle, WA
| | - Anuradha Ray
- Department of Immunology, University of Pittsburgh; Pittsburgh, PA
| | - Sally E. Wenzel
- Department of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh Medical Center; Pittsburgh, PA
| | - Joshua A. Boyce
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital; Boston, MA
- Department of Medicine, Harvard Medical School; Boston, MA
| | - Maria Gutierrez-Arcelus
- Division of Immunology, Boston Children’s Hospital; Boston, MA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard; Cambridge, MA
| | - Nora A. Barrett
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital; Boston, MA
- Department of Medicine, Harvard Medical School; Boston, MA
| |
Collapse
|
10
|
Murphy RC, Lai Y, Altman MC, Barrow KA, Dill-McFarland KA, Liu M, Hamerman JA, Lacy-Hulbert A, Piliponsky AM, Ziegler SF, Altemeier WA, Debley JS, Gharib SA, Hallstrand TS. Rhinovirus infection of the airway epithelium enhances mast cell immune responses via epithelial-derived interferons. J Allergy Clin Immunol 2023; 151:1484-1493. [PMID: 36708815 PMCID: PMC10257743 DOI: 10.1016/j.jaci.2022.12.825] [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: 03/16/2022] [Revised: 12/15/2022] [Accepted: 12/22/2022] [Indexed: 01/27/2023]
Abstract
BACKGROUND Mast cells (MCs) within the airway epithelium in asthma are closely related to airway dysfunction, but cross talk between airway epithelial cells (AECs) and MCs in asthma remains incompletely understood. Human rhinovirus (RV) infections are key triggers for asthma progression, and AECs from individuals with asthma may have dysregulated antiviral responses. OBJECTIVE We utilized primary AECs in an ex vivo coculture model system to examine cross talk between AECs and MCs after epithelial rhinovirus infection. METHODS Primary AECs were obtained from 11 children with asthma and 10 healthy children, differentiated at air-liquid interface, and cultured in the presence of laboratory of allergic diseases 2 (LAD2) MCs. AECs were infected with rhinovirus serogroup A 16 (RV16) for 48 hours. RNA isolated from both AECs and MCs underwent RNA sequencing. Direct effects of epithelial-derived interferons on LAD2 MCs were examined by real-time quantitative PCR. RESULTS MCs increased expression of proinflammatory and antiviral genes in AECs. AECs demonstrated a robust antiviral response after RV16 infection that resulted in significant changes in MC gene expression, including upregulation of genes involved in antiviral responses, leukocyte activation, and type 2 inflammation. Subsequent ex vivo modeling demonstrated that IFN-β induces MC type 2 gene expression. The effects of AEC donor phenotype were small relative to the effects of viral infection and the presence of MCs. CONCLUSIONS There is significant cross talk between AECs and MCs, which are present in the epithelium in asthma. Epithelial-derived interferons not only play a role in viral suppression but also further alter MC immune responses including specific type 2 genes.
Collapse
Affiliation(s)
- Ryan C Murphy
- Division of Pulmonary, Critical Care, and Sleep Medicine, Seattle, Wash; Center for Lung Biology, University of Washington, Seattle, Wash.
| | - Ying Lai
- Division of Pulmonary, Critical Care, and Sleep Medicine, Seattle, Wash; Center for Lung Biology, University of Washington, Seattle, Wash
| | - Matthew C Altman
- Division of Allergy and Infectious Disease, Department of Medicine, Seattle, Wash; Immunology Program, Benaroya Research Institute, Seattle, Wash
| | - Kaitlyn A Barrow
- Division of Pulmonary and Sleep Medicine, Seattle Children's Hospital, Department of Pediatrics, Seattle, Wash; Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Wash
| | | | - Matthew Liu
- Division of Pulmonary, Critical Care, and Sleep Medicine, Seattle, Wash; Center for Lung Biology, University of Washington, Seattle, Wash
| | | | | | - Adrian M Piliponsky
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Wash
| | | | - William A Altemeier
- Division of Pulmonary, Critical Care, and Sleep Medicine, Seattle, Wash; Center for Lung Biology, University of Washington, Seattle, Wash
| | - Jason S Debley
- Division of Pulmonary and Sleep Medicine, Seattle Children's Hospital, Department of Pediatrics, Seattle, Wash; Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Wash
| | - Sina A Gharib
- Division of Pulmonary, Critical Care, and Sleep Medicine, Seattle, Wash; Center for Lung Biology, University of Washington, Seattle, Wash
| | - Teal S Hallstrand
- Division of Pulmonary, Critical Care, and Sleep Medicine, Seattle, Wash; Center for Lung Biology, University of Washington, Seattle, Wash
| |
Collapse
|
11
|
Kicic-Starcevich E, Hancock DG, Iosifidis T, Agudelo-Romero P, Caparros-Martin JA, Silva D, Turkovic L, Le Souef PN, Bosco A, Martino DJ, Kicic A, Prescott SL, Stick SM. Airway Epithelium Respiratory Illnesses and Allergy (AERIAL) birth cohort: study protocol. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.04.29.23289314. [PMID: 37205501 PMCID: PMC10187351 DOI: 10.1101/2023.04.29.23289314] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Introduction Recurrent wheezing disorders including asthma are complex and heterogeneous diseases that affect up to 30% of all children, contributing to a major burden on children, their families, and global healthcare systems. It is now recognized that a dysfunctional airway epithelium plays a central role in the pathogenesis of recurrent wheeze, although the underlying mechanisms are still not fully understood. This prospective birth cohort aims to bridge this knowledge gap by investigating the influence of intrinsic epithelial dysfunction on the risk for developing respiratory disorders and the modulation of this risk by maternal morbidities, in utero exposures, and respiratory exposures in the first year of life. Methods and Analysis The Airway Epithelium Respiratory Illnesses and Allergy (AERIAL) study is nested within the ORIGINS Project and will monitor 400 infants from birth to five years. The primary outcome of the AERIAL study will be the identification of epithelial endotypes and exposure variables that influence the development of recurrent wheezing, asthma, and allergic sensitisation. Nasal respiratory epithelium at birth to six weeks, one, three, and five years will be analysed by bulk RNA-seq and DNA methylation sequencing. Maternal morbidities and in utero exposures will be identified on maternal history and their effects measured through transcriptomic and epigenetic analyses of the amnion and newborn epithelium. Exposures within the first year of life will be identified based on infant medical history as well as on background and symptomatic nasal sampling for viral PCR and microbiome analysis. Daily temperatures and symptoms recorded in a study-specific Smartphone App will be used to identify symptomatic respiratory illnesses. Ethics and Dissemination Ethical approval has been obtained from Ramsey Health Care HREC WA-SA (#1908). Results will be disseminated through open-access peer-reviewed manuscripts, conference presentations, and through different media channels to consumers, ORIGINS families, and the wider community.
Collapse
|
12
|
Wang X, Sima Y, Zhao Y, Zhang N, Zheng M, Du K, Wang M, Wang Y, Hao Y, Li Y, Liu M, Piao Y, Liu C, Tomassen P, Zhang L, Bachert C. Endotypes of chronic rhinosinusitis based on inflammatory and remodeling factors. J Allergy Clin Immunol 2023; 151:458-468. [PMID: 36272582 DOI: 10.1016/j.jaci.2022.10.010] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 09/29/2022] [Accepted: 10/05/2022] [Indexed: 11/17/2022]
Abstract
BACKGROUND Previous studies on the endotyping of chronic rhinosinusitis (CRS) that were based on inflammatory factors have broadened our understanding of the disease. However, the endotype of CRS combined with inflammatory and remodeling features has not yet been clearly elucidated. OBJECTIVE We sought to identify the endotypes of patients with CRS according to inflammatory and remodeling factors. METHODS Forty-eight inflammatory and remodeling factors in the nasal mucosal tissues of 128 CRS patients and 24 control subjects from northern China were analyzed by Luminex, ELISA, and ImmunoCAP. Sixteen factors were used to perform the cluster analysis. The characteristics of each cluster were analyzed using correlation analysis and validated by immunofluorescence staining. RESULTS Patients were classified into 5 clusters. Clusters 1 and 2 showed non-type 2 signatures with low biomarker concentrations, except for IL-19 and IL-27. Cluster 3 involved a low type 2 endotype with the highest expression of neutrophil factors, such as granulocyte colony-stimulating factor, IL-8, and myeloperoxidase, and remodeling factors, such as matrix metalloproteinases and fibronectin. Cluster 4 exhibited moderate type 2 inflammation. Cluster 5 exhibited high type 2 inflammation, which was associated with relatively higher levels of neutrophil and remodeling factors. The proportion of CRS with nasal polyps, asthma, allergies, anosmia, aspirin sensitivity, and the recurrence of CRS increased from clusters 1 to 5. CONCLUSION Diverse inflammatory mechanisms result in distinct CRS endotypes and remodeling profiles. The explicit differentiation and accurate description of these endotypes will guide targeted treatment decisions.
Collapse
Affiliation(s)
- Xiangdong Wang
- Department of Otorhinolaryngology Head and Neck Surgery, Department of Allergy, Beijing Tongren Hospital, Capital Medical University, Beijing, China; Beijing Institute of Otolaryngology, Beijing Laboratory of Allergic Diseases and Beijing Key Laboratory of Nasal Diseases, Key Laboratory of Otolaryngology Head and Neck Surgery, Ministry of Education, Capital Medical University, Beijing, China
| | - Yutong Sima
- Department of Otorhinolaryngology Head and Neck Surgery, Department of Allergy, Beijing Tongren Hospital, Capital Medical University, Beijing, China; Beijing Institute of Otolaryngology, Beijing Laboratory of Allergic Diseases and Beijing Key Laboratory of Nasal Diseases, Key Laboratory of Otolaryngology Head and Neck Surgery, Ministry of Education, Capital Medical University, Beijing, China
| | - Yan Zhao
- Department of Otorhinolaryngology Head and Neck Surgery, Department of Allergy, Beijing Tongren Hospital, Capital Medical University, Beijing, China; Beijing Institute of Otolaryngology, Beijing Laboratory of Allergic Diseases and Beijing Key Laboratory of Nasal Diseases, Key Laboratory of Otolaryngology Head and Neck Surgery, Ministry of Education, Capital Medical University, Beijing, China
| | - Nan Zhang
- Upper Airways Research Laboratory, Department of Oto-Rhino-Laryngology, Ghent University Hospital, Ghent, Belgium
| | - Ming Zheng
- Department of Otorhinolaryngology Head and Neck Surgery, Department of Allergy, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Kun Du
- Department of Otorhinolaryngology Head and Neck Surgery, Department of Allergy, Beijing Tongren Hospital, Capital Medical University, Beijing, China; Beijing Institute of Otolaryngology, Beijing Laboratory of Allergic Diseases and Beijing Key Laboratory of Nasal Diseases, Key Laboratory of Otolaryngology Head and Neck Surgery, Ministry of Education, Capital Medical University, Beijing, China
| | - Min Wang
- Department of Otorhinolaryngology Head and Neck Surgery, Department of Allergy, Beijing Tongren Hospital, Capital Medical University, Beijing, China; Beijing Institute of Otolaryngology, Beijing Laboratory of Allergic Diseases and Beijing Key Laboratory of Nasal Diseases, Key Laboratory of Otolaryngology Head and Neck Surgery, Ministry of Education, Capital Medical University, Beijing, China
| | - Yue Wang
- Department of Otorhinolaryngology Head and Neck Surgery, Department of Allergy, Beijing Tongren Hospital, Capital Medical University, Beijing, China; Beijing Institute of Otolaryngology, Beijing Laboratory of Allergic Diseases and Beijing Key Laboratory of Nasal Diseases, Key Laboratory of Otolaryngology Head and Neck Surgery, Ministry of Education, Capital Medical University, Beijing, China
| | - Yun Hao
- Department of Otorhinolaryngology Head and Neck Surgery, Department of Allergy, Beijing Tongren Hospital, Capital Medical University, Beijing, China; Beijing Institute of Otolaryngology, Beijing Laboratory of Allergic Diseases and Beijing Key Laboratory of Nasal Diseases, Key Laboratory of Otolaryngology Head and Neck Surgery, Ministry of Education, Capital Medical University, Beijing, China
| | - Ying Li
- Department of Otorhinolaryngology Head and Neck Surgery, Department of Allergy, Beijing Tongren Hospital, Capital Medical University, Beijing, China; Beijing Institute of Otolaryngology, Beijing Laboratory of Allergic Diseases and Beijing Key Laboratory of Nasal Diseases, Key Laboratory of Otolaryngology Head and Neck Surgery, Ministry of Education, Capital Medical University, Beijing, China
| | | | - Yingshi Piao
- Department of Pathology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Chengyao Liu
- Department of Otorhinolaryngology Head and Neck Surgery, Department of Allergy, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Peter Tomassen
- Upper Airways Research Laboratory, Department of Oto-Rhino-Laryngology, Ghent University Hospital, Ghent, Belgium
| | - Luo Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, Department of Allergy, Beijing Tongren Hospital, Capital Medical University, Beijing, China; Beijing Institute of Otolaryngology, Beijing Laboratory of Allergic Diseases and Beijing Key Laboratory of Nasal Diseases, Key Laboratory of Otolaryngology Head and Neck Surgery, Ministry of Education, Capital Medical University, Beijing, China; Research Unit of Diagnosis and Treatment of Chronic Nasal Diseases, Chinese Academy of Medical Sciences, Beijing, China.
| | - Claus Bachert
- Upper Airways Research Laboratory, Department of Oto-Rhino-Laryngology, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
13
|
Ehrhardt B, El-Merhie N, Kovacevic D, Schramm J, Bossen J, Roeder T, Krauss-Etschmann S. Airway remodeling: The Drosophila model permits a purely epithelial perspective. FRONTIERS IN ALLERGY 2022; 3:876673. [PMID: 36187164 PMCID: PMC9520053 DOI: 10.3389/falgy.2022.876673] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 08/22/2022] [Indexed: 11/16/2022] Open
Abstract
Airway remodeling is an umbrella term for structural changes in the conducting airways that occur in chronic inflammatory lung diseases such as asthma or chronic obstructive pulmonary disease (COPD). The pathobiology of remodeling involves multiple mesenchymal and lymphoid cell types and finally leads to a variety of hardly reversible changes such as hyperplasia of goblet cells, thickening of the reticular basement membrane, deposition of collagen, peribronchial fibrosis, angiogenesis and hyperplasia of bronchial smooth muscle cells. In order to develop solutions for prevention or innovative therapies, these complex processes must be understood in detail which requires their deconstruction into individual building blocks. In the present manuscript we therefore focus on the role of the airway epithelium and introduce Drosophila melanogaster as a model. The simple architecture of the flies’ airways as well as the lack of adaptive immunity allows to focus exclusively on the importance of the epithelium for the remodeling processes. We will review and discuss genetic and environmentally induced changes in epithelial structures and molecular responses and propose an integrated framework of research for the future.
Collapse
Affiliation(s)
- Birte Ehrhardt
- Division of Early Life Origins of Chronic Lung Diseases, Research Center Borstel, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Borstel, Germany
| | - Natalia El-Merhie
- Division of Early Life Origins of Chronic Lung Diseases, Research Center Borstel, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Borstel, Germany
| | - Draginja Kovacevic
- Division of Early Life Origins of Chronic Lung Diseases, Research Center Borstel, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Borstel, Germany
| | - Juliana Schramm
- Division of Early Life Origins of Chronic Lung Diseases, Research Center Borstel, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Borstel, Germany
| | - Judith Bossen
- Division of Molecular Physiology, Institute of Zoology, Christian-Albrechts University Kiel, Kiel, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Kiel, Germany
| | - Thomas Roeder
- Division of Molecular Physiology, Institute of Zoology, Christian-Albrechts University Kiel, Kiel, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Kiel, Germany
| | - Susanne Krauss-Etschmann
- Division of Early Life Origins of Chronic Lung Diseases, Research Center Borstel, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Borstel, Germany
- Institute of Experimental Medicine, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
- Correspondence: Susanne Krauss-Etschmann
| |
Collapse
|
14
|
Zhu Y, Chew KY, Wu M, Karawita AC, McCallum G, Steele LE, Yamamoto A, Labzin LI, Yarlagadda T, Khromykh AA, Wang X, Sng JDJ, Stocks CJ, Xia Y, Kollmann TR, Martino D, Joensuu M, Meunier FA, Balistreri G, Bielefeldt-Ohmann H, Bowen AC, Kicic A, Sly PD, Spann KM, Short KR. Ancestral SARS-CoV-2, but not Omicron, replicates less efficiently in primary pediatric nasal epithelial cells. PLoS Biol 2022; 20:e3001728. [PMID: 35913989 PMCID: PMC9371332 DOI: 10.1371/journal.pbio.3001728] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 08/11/2022] [Accepted: 06/24/2022] [Indexed: 01/02/2023] Open
Abstract
Children typically experience more mild symptoms of Coronavirus Disease 2019 (COVID-19) when compared to adults. There is a strong body of evidence that children are also less susceptible to Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infection with the ancestral viral isolate. However, the emergence of SARS-CoV-2 variants of concern (VOCs) has been associated with an increased number of pediatric infections. Whether this is the result of widespread adult vaccination or fundamental changes in the biology of SARS-CoV-2 remain to be determined. Here, we use primary nasal epithelial cells (NECs) from children and adults, differentiated at an air-liquid interface to show that the ancestral SARS-CoV-2 replicates to significantly lower titers in the NECs of children compared to those of adults. This was associated with a heightened antiviral response to SARS-CoV-2 in the NECs of children. Importantly, the Delta variant also replicated to significantly lower titers in the NECs of children. This trend was markedly less pronounced in the case of Omicron. It is also striking to note that, at least in terms of viral RNA, Omicron replicated better in pediatric NECs compared to both Delta and the ancestral virus. Taken together, these data show that the nasal epithelium of children supports lower infection and replication of ancestral SARS-CoV-2, although this may be changing as the virus evolves.
Collapse
Affiliation(s)
- Yanshan Zhu
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Keng Yih Chew
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Melanie Wu
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Anjana C. Karawita
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Georgina McCallum
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Lauren E. Steele
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Ayaho Yamamoto
- Child Health Research Centre, The University of Queensland, South Brisbane, Queensland, Australia
| | - Larisa I. Labzin
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Tejasri Yarlagadda
- Centre for Immunology and Infection Control, Faculty of Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Alexander A. Khromykh
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia
- Australian Infectious Diseases Research Centre, Global Virus Network Centre of Excellence, Brisbane, Queensland, Australia
| | - Xiaohui Wang
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Julian D. J. Sng
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Claudia J. Stocks
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Yao Xia
- School of Science, Edith Cowan University, Joondalup, Western Australia, Australia
- School of Biomedical Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Tobias R. Kollmann
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, The University of Western Australia, Perth, Western Australia, Australia
| | - David Martino
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, The University of Western Australia, Perth, Western Australia, Australia
| | - Merja Joensuu
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland, Australia
| | - Frédéric A. Meunier
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Giuseppe Balistreri
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
- Department of Virology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Helle Bielefeldt-Ohmann
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia
- Australian Infectious Diseases Research Centre, Global Virus Network Centre of Excellence, Brisbane, Queensland, Australia
| | - Asha C. Bowen
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, The University of Western Australia, Perth, Western Australia, Australia
- Department of Infectious Diseases, Perth Children’s Hospital, Nedlands, Perth, Western Australia, Australia
- Wesfarmers Centre for Vaccines and Infectious Diseases, Telethon Kids Institute, The University of Western Australia, Perth, Western Australia, Australia
- Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
| | - Anthony Kicic
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, The University of Western Australia, Perth, Western Australia, Australia
- Occupation and Environment, School of Public Health, Curtin University, Perth, Western Australia, Australia
- Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia, Perth, Western Australia, Australia
| | - Peter D. Sly
- Child Health Research Centre, The University of Queensland, South Brisbane, Queensland, Australia
- Australian Infectious Diseases Research Centre, Global Virus Network Centre of Excellence, Brisbane, Queensland, Australia
| | - Kirsten M. Spann
- Centre for Immunology and Infection Control, Faculty of Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Kirsty R. Short
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia
- Australian Infectious Diseases Research Centre, Global Virus Network Centre of Excellence, Brisbane, Queensland, Australia
| |
Collapse
|
15
|
Do AR, Ko DY, Kim J, Bak SH, Lee KY, Yoon D, Shin C, Kim S, Kim WJ, Won S. Genome-Wide Association Study of Airway Wall Thickening in a Korean Chronic Obstructive Pulmonary Disease Cohort. Genes (Basel) 2022; 13:genes13071258. [PMID: 35886039 PMCID: PMC9318537 DOI: 10.3390/genes13071258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 06/23/2022] [Accepted: 06/28/2022] [Indexed: 02/04/2023] Open
Abstract
Airway wall thickening (AWT) plays an important pathophysiological role in airway diseases such as chronic obstructive pulmonary disease (COPD). There are only a few studies on the genetic components contributing to AWT in the Korean population. This study aimed to identify AWT-related single-nucleotide polymorphisms (SNPs) using a genome-wide association study (GWAS). We performed GWAS for AWT using the CODA and KUCOPD cohorts. Thereafter, a meta-analysis was performed. Airway wall thickness was measured using automatic segmentation software. The AWT at an internal perimeter of 10 mm (AWT-Pi10) was calculated by the square root of the theoretical airway wall area using the full-width-half-maximum method. We identified a significant SNP (rs11648772, p = 1.41 × 10-8) located in LINC02127, near SALL1. This gene is involved in the inhibition of epithelial-mesenchymal transition in glial cells, and it affects bronchial wall depression in COPD patients. Additionally, we identified other SNPs (rs11970854, p = 1.92 × 10-6; rs16920168, p = 5.29 × 10-6) involved in airway inflammation and proliferation and found that AWT is influenced by these genetic variants. Our study helps identify the genetic cause of COPD in an Asian population and provides a potential basis for treatment.
Collapse
Affiliation(s)
- Ah Ra Do
- Interdisciplinary Program of Bioinformatics, Seoul National University, Seoul 08826, Korea;
| | - Do Yeon Ko
- Environmental Health Center, Department of Internal Medicine, Kangwon National University, Chuncheon 25948, Korea; (D.Y.K.); (J.K.)
| | - Jeeyoung Kim
- Environmental Health Center, Department of Internal Medicine, Kangwon National University, Chuncheon 25948, Korea; (D.Y.K.); (J.K.)
| | - So Hyeon Bak
- Department of Radiology, Kangwon National University Hospital, Kangwon National University School of Medicine, Chuncheon 24341, Korea;
| | - Ki Yeol Lee
- Department of Radiology, Korea University Ansan Hospital, Ansan 15355, Korea;
| | - Dankyu Yoon
- Department of Chronic Disease Convergence Research, Division of Allergy and Respiratory Disease Research, National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju 28159, Korea;
| | - Chol Shin
- Institute for Human Genomic Study, College of Medicine, Korea University, Seoul 08826, Korea;
- Department of Internal Medicine, Division of Pulmonary Sleep and Critical Care Medicine, Korea University Ansan Hospital, Ansan 15355, Korea
| | - Soriul Kim
- Institute for Human Genomic Study, College of Medicine, Korea University, Seoul 08826, Korea;
- Correspondence: (S.K.); (W.J.K.); (S.W.); Tel.: +82-31-412-5603 (S.K.); +82-33-258-9303 (W.J.K.), +82-2-880-2714 (S.W.)
| | - Woo Jin Kim
- Environmental Health Center, Department of Internal Medicine, Kangwon National University, Chuncheon 25948, Korea; (D.Y.K.); (J.K.)
- Correspondence: (S.K.); (W.J.K.); (S.W.); Tel.: +82-31-412-5603 (S.K.); +82-33-258-9303 (W.J.K.), +82-2-880-2714 (S.W.)
| | - Sungho Won
- Interdisciplinary Program of Bioinformatics, Seoul National University, Seoul 08826, Korea;
- Department of Public Health Sciences, School of Public Health, Seoul National University, Seoul 08826, Korea
- Institute of Health and Environment, Seoul National University, Seoul 08826, Korea
- RexSoft Inc., Seoul 08826, Korea
- Correspondence: (S.K.); (W.J.K.); (S.W.); Tel.: +82-31-412-5603 (S.K.); +82-33-258-9303 (W.J.K.), +82-2-880-2714 (S.W.)
| |
Collapse
|
16
|
Kalesinskas L, Gupta S, Khatri P. Increasing reproducibility, robustness, and generalizability of biomarker selection from meta-analysis using Bayesian methodology. PLoS Comput Biol 2022; 18:e1010260. [PMID: 35759523 PMCID: PMC9269905 DOI: 10.1371/journal.pcbi.1010260] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 07/08/2022] [Accepted: 05/29/2022] [Indexed: 01/07/2023] Open
Abstract
A major limitation of gene expression biomarker studies is that they are not reproducible as they simply do not generalize to larger, real-world, heterogeneous populations. Frequentist multi-cohort gene expression meta-analysis has been frequently used as a solution to this problem to identify biomarkers that are truly differentially expressed. However, the frequentist meta-analysis framework has its limitations-it needs at least 4-5 datasets with hundreds of samples, is prone to confounding from outliers and relies on multiple-hypothesis corrected p-values. To address these shortcomings, we have created a Bayesian meta-analysis framework for the analysis of gene expression data. Using real-world data from three different diseases, we show that the Bayesian method is more robust to outliers, creates more informative estimates of between-study heterogeneity, reduces the number of false positive and false negative biomarkers and selects more generalizable biomarkers with less data. We have compared the Bayesian framework to a previously published frequentist framework and have developed a publicly available R package for use.
Collapse
Affiliation(s)
- Laurynas Kalesinskas
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, California, United States of America
- Center for Biomedical Informatics Research, Department of Medicine, Stanford University, Stanford, California, United States of America
- Department of Biomedical Data Science, School of Medicine, Stanford University, Stanford, California, United States of America
| | - Sanjana Gupta
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, California, United States of America
- Center for Biomedical Informatics Research, Department of Medicine, Stanford University, Stanford, California, United States of America
| | - Purvesh Khatri
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, California, United States of America
- Center for Biomedical Informatics Research, Department of Medicine, Stanford University, Stanford, California, United States of America
- * E-mail:
| |
Collapse
|
17
|
Gwatimba A, Rosenow T, Stick SM, Kicic A, Iosifidis T, Karpievitch YV. AI-Driven Cell Tracking to Enable High-Throughput Drug Screening Targeting Airway Epithelial Repair for Children with Asthma. J Pers Med 2022; 12:jpm12050809. [PMID: 35629232 PMCID: PMC9146422 DOI: 10.3390/jpm12050809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/08/2022] [Accepted: 05/13/2022] [Indexed: 11/16/2022] Open
Abstract
The airway epithelium of children with asthma is characterized by aberrant repair that may be therapeutically modifiable. The development of epithelial-targeting therapeutics that enhance airway repair could provide a novel treatment avenue for childhood asthma. Drug discovery efforts utilizing high-throughput live cell imaging of patient-derived airway epithelial culture-based wound repair assays can be used to identify compounds that modulate airway repair in childhood asthma. Manual cell tracking has been used to determine cell trajectories and wound closure rates, but is time consuming, subject to bias, and infeasible for high-throughput experiments. We therefore developed software, EPIC, that automatically tracks low-resolution low-framerate cells using artificial intelligence, analyzes high-throughput drug screening experiments and produces multiple wound repair metrics and publication-ready figures. Additionally, unlike available cell trackers that perform cell segmentation, EPIC tracks cells using bounding boxes and thus has simpler and faster training data generation requirements for researchers working with other cell types. EPIC outperformed publicly available software in our wound repair datasets by achieving human-level cell tracking accuracy in a fraction of the time. We also showed that EPIC is not limited to airway epithelial repair for children with asthma but can be applied in other cellular contexts by outperforming the same software in the Cell Tracking with Mitosis Detection Challenge (CTMC) dataset. The CTMC is the only established cell tracking benchmark dataset that is designed for cell trackers utilizing bounding boxes. We expect our open-source and easy-to-use software to enable high-throughput drug screening targeting airway epithelial repair for children with asthma.
Collapse
Affiliation(s)
- Alphons Gwatimba
- Wal-Yan Respiratory Research Centre, Telethon Kids Institute, University of Western Australia, Perth, WA 6009, Australia; (T.R.); (S.M.S.); (A.K.); (T.I.); (Y.V.K.)
- School of Computer Science and Software Engineering, University of Western Australia, Nedlands, WA 6009, Australia
- Correspondence:
| | - Tim Rosenow
- Wal-Yan Respiratory Research Centre, Telethon Kids Institute, University of Western Australia, Perth, WA 6009, Australia; (T.R.); (S.M.S.); (A.K.); (T.I.); (Y.V.K.)
- Centre for Microscopy, Characterisation and Analysis, University of Western Australia, Nedlands, WA 6009, Australia
| | - Stephen M. Stick
- Wal-Yan Respiratory Research Centre, Telethon Kids Institute, University of Western Australia, Perth, WA 6009, Australia; (T.R.); (S.M.S.); (A.K.); (T.I.); (Y.V.K.)
- Division of Paediatrics, Medical School, University of Western Australia, Nedlands, WA 6009, Australia
- Department of Respiratory and Sleep Medicine, Perth Children’s Hospital, Nedlands, WA 6009, Australia
- Centre for Cell Therapy and Regenerative Medicine, School of Medicine, University of Western Australia, Nedlands, WA 6009, Australia
| | - Anthony Kicic
- Wal-Yan Respiratory Research Centre, Telethon Kids Institute, University of Western Australia, Perth, WA 6009, Australia; (T.R.); (S.M.S.); (A.K.); (T.I.); (Y.V.K.)
- Division of Paediatrics, Medical School, University of Western Australia, Nedlands, WA 6009, Australia
- Centre for Cell Therapy and Regenerative Medicine, School of Medicine, University of Western Australia, Nedlands, WA 6009, Australia
- School of Population Health, Curtin University, Bentley, WA 6102, Australia
| | - Thomas Iosifidis
- Wal-Yan Respiratory Research Centre, Telethon Kids Institute, University of Western Australia, Perth, WA 6009, Australia; (T.R.); (S.M.S.); (A.K.); (T.I.); (Y.V.K.)
- Centre for Cell Therapy and Regenerative Medicine, School of Medicine, University of Western Australia, Nedlands, WA 6009, Australia
- School of Population Health, Curtin University, Bentley, WA 6102, Australia
| | - Yuliya V. Karpievitch
- Wal-Yan Respiratory Research Centre, Telethon Kids Institute, University of Western Australia, Perth, WA 6009, Australia; (T.R.); (S.M.S.); (A.K.); (T.I.); (Y.V.K.)
- School of Biomedical Sciences, University of Western Australia, Nedlands, WA 6009, Australia
| |
Collapse
|
18
|
Jin M, Watkins S, Larriba Y, Wallace C, St. Croix C, Zhou X, Zhao J, Peddada S, Wenzel SE. Real-time imaging of asthmatic epithelial cells identifies migratory deficiencies under type-2 conditions. J Allergy Clin Immunol 2022; 149:579-588. [PMID: 34547368 PMCID: PMC8821171 DOI: 10.1016/j.jaci.2021.08.027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 08/05/2021] [Accepted: 08/27/2021] [Indexed: 02/03/2023]
Abstract
BACKGROUND The epithelium is increasingly recognized as a pathologic contributor to asthma and its phenotypes. Although delayed wound closure by asthmatic epithelial cells is consistently observed, underlying mechanisms remain poorly understood, partly due to difficulties in studying dynamic physiologic processes involving polarized multilayered cell systems. Although type-2 immunity has been suggested to play a role, the mechanisms by which repair is diminished are unclear. OBJECTIVES This study sought to develop and utilize primary multilayered polarized epithelial cell systems, derived from patients with asthma, to evaluate cell migration in response to wounding under type-2 and untreated conditions. METHODS A novel wounding device for multilayered polarized cells, along with time-lapse live cell/real-time confocal imaging were evaluated under IL-13 and untreated conditions. The influence of inhibition of 15 lipoxygenase (15LO1), a type-2 enzyme, on the process was also addressed. Cell migration patterns were analyzed by high-dimensional frequency modulated Möbius for statistical comparisons. RESULTS IL-13 stimulation negatively impacts wound healing by altering the total speed, directionality, and acceleration of individual cells. Inhibition 15LO1 partially improved the wound repair through improving total speed. CONCLUSIONS Migration abnormalities contributed to markedly slower wound closure of IL-13 treated cells, which was modestly reversed by 15LO1 inhibition, suggesting its potential as an asthma therapeutic target. These novel methodologies offer new ways to dynamically study cell movements and identify contributing pathologic processes.
Collapse
Affiliation(s)
- Mingzhu Jin
- Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, USA,Department of Rhinology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Simon Watkins
- Center for Biologic Imaging, School of Medicine, University of Pittsburgh, Pittsburgh, USA
| | - Yolanda Larriba
- Department of Statistics and Operations Research, Universidad de Valladolid, Valladolid, Spain
| | - Callen Wallace
- Center for Biologic Imaging, School of Medicine, University of Pittsburgh, Pittsburgh, USA
| | - Claudette St. Croix
- Center for Biologic Imaging, School of Medicine, University of Pittsburgh, Pittsburgh, USA
| | - Xiuxia Zhou
- Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, USA
| | - Jinming Zhao
- Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, USA
| | - Shyamal Peddada
- Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, USA
| | - Sally E. Wenzel
- Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, USA,Asthma and Environmental Lung Health Institute @UPMC, Pittsburgh, USA
| |
Collapse
|
19
|
Watkinson RL, Looi K, Laing IA, Cianferoni A, Kicic A. Viral Induced Effects on a Vulnerable Epithelium; Lessons Learned From Paediatric Asthma and Eosinophilic Oesophagitis. Front Immunol 2021; 12:773600. [PMID: 34912343 PMCID: PMC8666438 DOI: 10.3389/fimmu.2021.773600] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 11/05/2021] [Indexed: 01/07/2023] Open
Abstract
The epithelium is integral to the protection of many different biological systems and for the maintenance of biochemical homeostasis. Emerging evidence suggests that particular children have epithelial vulnerabilities leading to dysregulated barrier function and integrity, that resultantly contributes to disease pathogenesis. These epithelial vulnerabilities likely develop in utero or in early life due to various genetic, epigenetic and environmental factors. Although various epithelia are uniquely structured with specific function, prevalent allergic-type epithelial diseases in children potentially have common or parallel disease processes. These include inflammation and immune response dysregulation stemming from atypical epithelial barrier function and integrity. Two diseases where aetiology and pathogenesis are potentially linked to epithelial vulnerabilities include Paediatric Asthma and Eosinophilic Oesophagitis (EoE). For example, rhinovirus C (RV-C) is a known risk factor for paediatric asthma development and is known to disrupt respiratory epithelial barrier function causing acute inflammation. In addition, EoE, a prevalent atopic condition of the oesophageal epithelium, is characterised by similar innate immune and epithelial responses to viral injury. This review examines the current literature and identifies the gaps in the field defining viral-induced effects on a vulnerable respiratory epithelium and resulting chronic inflammation, drawing from knowledge generated in acute wheezing illness, paediatric asthma and EoE. Besides highlighting the importance of epithelial structure and barrier function in allergic disease pathogenesis regardless of specific epithelial sub-types, this review focuses on the importance of examining other parallel allergic-type disease processes that may uncover commonalities driving disease pathogenesis. This in turn may be beneficial in the development of common therapeutics for current clinical management and disease prevention in the future.
Collapse
Affiliation(s)
- Rebecca L Watkinson
- Division of Paediatrics, Medical School, The University of Western Australia, Nedlands, WA, Australia.,Wal-Yan Respiratory Research Centre, Telethon Kids Institute, Perth, WA, Australia
| | - Kevin Looi
- Wal-Yan Respiratory Research Centre, Telethon Kids Institute, Perth, WA, Australia.,School of Public Health, Curtin University, Bentley, WA, Australia
| | - Ingrid A Laing
- Division of Paediatrics, Medical School, The University of Western Australia, Nedlands, WA, Australia.,Wal-Yan Respiratory Research Centre, Telethon Kids Institute, Perth, WA, Australia
| | - Antonella Cianferoni
- Pediatrics Department, Perlman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Anthony Kicic
- Wal-Yan Respiratory Research Centre, Telethon Kids Institute, Perth, WA, Australia.,School of Public Health, Curtin University, Bentley, WA, Australia.,Centre for Cell Therapy and Regenerative Medicine, School of Medicine, The University of Western Australia, Nedlands, WA, Australia
| |
Collapse
|
20
|
Dysregulated Notch Signaling in the Airway Epithelium of Children with Wheeze. J Pers Med 2021; 11:jpm11121323. [PMID: 34945795 PMCID: PMC8707470 DOI: 10.3390/jpm11121323] [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: 09/06/2021] [Revised: 11/16/2021] [Accepted: 11/29/2021] [Indexed: 11/17/2022] Open
Abstract
The airway epithelium of children with wheeze is characterized by defective repair that contributes to disease pathobiology. Dysregulation of developmental processes controlled by Notch has been identified in chronic asthma. However, its role in airway epithelial cells of young children with wheeze, particularly during repair, is yet to be determined. We hypothesized that Notch is dysregulated in primary airway epithelial cells (pAEC) of children with wheeze contributing to defective repair. This study investigated transcriptional and protein expression and function of Notch in pAEC isolated from children with and without wheeze. Primary AEC of children with and without wheeze were found to express all known Notch receptors and ligands, although pAEC from children with wheeze expressed significantly lower NOTCH2 (10-fold, p = 0.004) and higher JAG1 (3.5-fold, p = 0.002) mRNA levels. These dysregulations were maintained in vitro and cultures from children with wheeze displayed altered kinetics of both NOTCH2 and JAG1 expression during repair. Following Notch signaling inhibition, pAEC from children without wheeze failed to repair (wound closure rate of 76.9 ± 3.2%). Overexpression of NOTCH2 in pAEC from children with wheeze failed to rescue epithelial repair following wounding. This study illustrates the involvement of the Notch pathway in airway epithelial wound repair in health and disease, where its dysregulation may contribute to asthma development.
Collapse
|
21
|
Bhaker S, Portelli MA, Rakkar K, Shaw D, Johnson S, Brightling C, Sayers I. Human bronchial epithelial cells from patients with asthma have an altered gene expression profile. ERJ Open Res 2021; 8:00625-2021. [PMID: 35198626 PMCID: PMC8859501 DOI: 10.1183/23120541.00625-2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 11/10/2021] [Indexed: 11/22/2022] Open
Abstract
Asthma is a multifactorial disease presenting with wheeze and shortness of breath, and is known to be exacerbated by triggers such as pollen, house dust mites and viral infection. In the lung, the bronchial epithelium is recognised as a central driver of airway structural changes, including epithelial goblet cell hyperplasia and metaplasia, which are features of asthma. Bronchial epithelial cells (BECs) isolated from patients with asthma and cultured in vitro have altered barrier properties [1], elevated expression of remodelling factors [2] and defective repair [3]. Interestingly, genome-wide association studies (GWAS) of asthma have implicated a number of genes that are known to be expressed and functional in the airway epithelium, including IL33, IL1RL1, TSLP and MUC5AC [4]. To identify the molecular mechanisms underlying altered BECs phenotype in asthma patients, several studies have completed transcriptomic analyses using bronchial brush samples. Two recent meta-analyses [5, 6] suggested that alterations in chemical stimulus, extracellular region, pathways in cancer and arachidonic acid metabolism were features of the bronchial epithelium in the lungs of patients with asthma, and included 78 up- and 75 down-regulated genes [5]. While useful, a key question is how much the airway environment of a patient is driving this differential gene expression profile (GEP) and how much is intrinsic to the BECs themselves? To answer this question, we completed transcriptomic analyses of BECs cultured two-dimensionally through multiple passages in the laboratory that had originally been isolated from control subjects without disease or patients with asthma. An attrition rate (for successful culture) of 54% and 42% was observed in the asthma and control populations respectively. Gene changes observed in asthma bronchial epithelial cells are maintained following repeated culture, presenting with an exaggerated response to viral infection and immune responses as well as having differences in the rate of cell division and replicationhttps://bit.ly/3Cq2xKf
Collapse
|
22
|
Ben Hamouda S, Miglino MA, de Sá Schiavo Matias G, Beauchamp G, Lavoie JP. Asthmatic Bronchial Matrices Determine the Gene Expression and Behavior of Smooth Muscle Cells in a 3D Culture Model. FRONTIERS IN ALLERGY 2021; 2:762026. [PMID: 35387054 PMCID: PMC8974673 DOI: 10.3389/falgy.2021.762026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 10/18/2021] [Indexed: 11/17/2022] Open
Abstract
Asthma is associated with increased deposition and altered phenotype of airway smooth muscle (ASM) cells. However, little is known about the processes responsible for these changes. It has been suggested that alterations of the extracellular matrix (ECM) contribute to the remodeling of ASM cells in asthma. Three-dimensional matrices allow the in vitro study of complex cellular responses to different stimuli in a close-to-natural environment. Thus, we investigated the ultrastructural and genic variations of ASM cells cultured on acellular asthmatic and control bronchial matrices. We studied horses, as they spontaneously develop a human asthma-like condition (heaves) with similarities to chronic pulmonary changes observed in human asthma. Primary bronchial ASM cells from asthmatic (n = 3) and control (n = 3) horses were cultured on decellularized bronchi from control (n = 3) and asthmatic (n = 3) horses. Each cell lineage was used to recellularize six different bronchi for 41 days. Histomorphometry on HEPS-stained-recellularized matrices revealed an increased ASM cell number in the control cell/control matrix (p = 0.02) and asthmatic cell/control matrix group (p = 0.04) compared with the asthmatic cell/asthmatic matrix group. Scan electron microscopy revealed a cell invasion of the ECM. While ASM cells showed high adhesion and proliferation processes on the control ECM, the presence of senescent cells and cellular debris in the asthmatic ECM with control or asthmatic ASM cells suggested cell death. When comparing asthmatic with control cell/matrix combinations by targeted next generation sequencing, only AGC1 (p = 0.04), MYO10 (p = 0.009), JAM3 (p = 0.02), and TAGLN (p = 0.001) were differentially expressed out of a 70-gene pool previously associated with smooth muscle remodeling. To our knowledge, this is the first attempt to evaluate the effects of asthmatic ECM on an ASM cell phenotype using a biological bronchial matrix. Our results indicate that bronchial ECM health status contributes to ASM cell gene expression and, possibly, its survival.
Collapse
Affiliation(s)
- Selma Ben Hamouda
- Faculty of Veterinary Medicine, Université de Montréal, Quebec City, QC, Canada
| | - Maria Angélica Miglino
- School of Veterinary Medicine and Animal Sciences, University of São Paulo, São Paulo, Brazil
| | | | - Guy Beauchamp
- Faculty of Veterinary Medicine, Université de Montréal, Quebec City, QC, Canada
| | - Jean-Pierre Lavoie
- Faculty of Veterinary Medicine, Université de Montréal, Quebec City, QC, Canada
- *Correspondence: Jean-Pierre Lavoie
| |
Collapse
|
23
|
Fang L, Roth M. Airway Wall Remodeling in Childhood Asthma-A Personalized Perspective from Cell Type-Specific Biology. J Pers Med 2021; 11:jpm11111229. [PMID: 34834581 PMCID: PMC8625708 DOI: 10.3390/jpm11111229] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/12/2021] [Accepted: 11/17/2021] [Indexed: 12/16/2022] Open
Abstract
Airway wall remodeling is a pathology occurring in chronic inflammatory lung diseases including asthma, chronic obstructive pulmonary disease, and fibrosis. In 2017, the American Thoracic Society released a research statement highlighting the gaps in knowledge and understanding of airway wall remodeling. The four major challenges addressed in this statement were: (i) the lack of consensus to define “airway wall remodeling” in different diseases, (ii) methodologic limitations and inappropriate models, (iii) the lack of anti-remodeling therapies, and (iv) the difficulty to define endpoints and outcomes in relevant studies. This review focuses on the importance of cell-cell interaction, especially the bronchial epithelium, in asthma-associated airway wall remodeling. The pathology of “airway wall remodeling” summarizes all structural changes of the airway wall without differentiating between different pheno- or endo-types of asthma. Indicators of airway wall remodeling have been reported in childhood asthma in the absence of any sign of inflammation; thus, the initiation event remains unknown. Recent studies have implied that the interaction between the epithelium with immune cells and sub-epithelial mesenchymal cells is modified in asthma by a yet unknown epigenetic mechanism during early childhood.
Collapse
|
24
|
Nowak JK, Dworacka M, Gubaj N, Dossimov A, Dossimov Z, Walkowiak J. Expression profiling of ileal mucosa in asthma reveals upregulation of innate immunity and genes characteristic of Paneth and goblet cells. Allergy Asthma Clin Immunol 2021; 17:82. [PMID: 34332619 PMCID: PMC8325823 DOI: 10.1186/s13223-021-00584-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 07/21/2021] [Indexed: 11/10/2022] Open
Abstract
Background The expression profiles of the intestinal mucosa have not been comprehensively investigated in asthma. We aimed to explore this in the Correlated Expression and Disease Association Research (CEDAR) patient cohort. Methods Differential expression analysis of ileal, transverse colon, and rectal biopsies were supplemented by a comparison of transcriptomes from platelets and leukocytes subsets, including CD4+, CD8+, CD14+, CD15+, and CD19+ cells. Asthma patients (n = 15) and controls (n = 15) had similar age (p = 0.967), body mass index (p = 0.870), similar numbers of females (80%) and smoking rates (13.3%). Results Significant differential expression was found in the ileum alone, and not in any other cell/tissue types. More genes were found to be overexpressed (1,150) than under-expressed (380). The most overexpressed genes included Fc Fragment of IgG Binding Protein (FCGBP, logFC = 3.01, pFDR = 0.015), Mucin 2 (MUC2, logFC = 2.78, pFDR = 0.015), and Alpha 1B Defensin (DEFA1B, logFC = 2.73, pFDR = 0.024). Gene ontology implicated the immune system, including interleukins 4 and 13, as well as antimicrobial peptides in this overexpression. There was concordance of gene over- (STAT1, XBP1) and underexpression (NELF, RARA) in asthma and Crohn’s disease ileum when our results were compared to another dataset (p = 3.66 × 10–7). Conclusion Ileal mucosa in asthma exhibits a specific transcriptomic profile, which includes the overexpression of innate immune genes, mostly characteristic of Paneth and goblet cells, in addition to other changes that may resemble Crohn’s disease. Supplementary Information The online version contains supplementary material available at 10.1186/s13223-021-00584-9.
Collapse
Affiliation(s)
- Jan K Nowak
- Department of Pediatric Gastroenterology and Metabolic Diseases, Poznan University of Medical Sciences, ul. Szpitalna 27/33, 60-572, Poznan, Poland.
| | - Marzena Dworacka
- Department of Pharmacology, Poznan University of Medical Sciences, Poznan, Poland
| | - Nazgul Gubaj
- Department of Pediatric Diseases No. 2, West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan
| | - Arystan Dossimov
- Department of Pediatric Diseases No. 2, West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan
| | - Zhumabek Dossimov
- Department of Pediatric Diseases No. 2, West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan
| | - Jarosław Walkowiak
- Department of Pediatric Gastroenterology and Metabolic Diseases, Poznan University of Medical Sciences, ul. Szpitalna 27/33, 60-572, Poznan, Poland
| |
Collapse
|
25
|
Yang Y, Jia M, Ou Y, Adcock IM, Yao X. Mechanisms and biomarkers of airway epithelial cell damage in asthma: A review. CLINICAL RESPIRATORY JOURNAL 2021; 15:1027-1045. [PMID: 34097803 DOI: 10.1111/crj.13407] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 05/17/2021] [Accepted: 06/03/2021] [Indexed: 12/15/2022]
Abstract
Bronchial asthma is a heterogeneous disease with complex pathological mechanisms representing different phenotypes, including severe asthma. The airway epithelium is a major site of complex pathological changes in severe asthma due, in part, to activation of inflammatory and immune mechanisms in response to noxious agents. Current imaging procedures are unable to accurately measure epithelial and airway remodeling. Damage of airway epithelial cells occurs is linked to specific phenotypes and endotypes which provides an opportunity for the identification of biomarkers reflecting epithelial, and airway, remodeling. Identification of patients with more severe epithelial disruption using biomarkers may also provide personalised therapeutic opportunities and/or markers of successful therapeutic intervention. Here, we review the evidence for ongoing epithelial cell dysregulation in the pathogenesis of asthma, the sentinel role of the airway epithelium and how understanding these molecular mechanisms provides the basis for the identification of candidate biomarkers for asthma prediction, prevention, diagnosis, treatment and monitoring.
Collapse
Affiliation(s)
- Yuemei Yang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Man Jia
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yingwei Ou
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Department of Emergency Medical, Zhejiang Province People's Hospital, Zhejiang, China
| | - Ian M Adcock
- Airway Disease Section, National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, UK
| | - Xin Yao
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
26
|
Hillas J, Evans DJ, Ang S, Iosifidis T, Garratt LW, Hemy N, Kicic-Starcevich E, Simpson SJ, Kicic A. Nasal airway epithelial repair after very preterm birth. ERJ Open Res 2021; 7:00913-2020. [PMID: 34109241 PMCID: PMC8181665 DOI: 10.1183/23120541.00913-2020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 03/27/2021] [Indexed: 12/03/2022] Open
Abstract
Preterm birth rates are increasing and now account for >11% of global births. Simultaneously, advances in neonatal care have led to increased survival of lower gestation neonates. A complication of preterm birth, and the biggest determinant of survival, is lung and airway immaturity. After preterm birth, the immature respiratory system is exposed to pro-inflammatory stimuli like injury from resuscitation and oxygen toxicity. The airway epithelium, the physical barrier between insults and the airways, is particularly vulnerable to injury. If epithelial barrier integrity cannot be restored rapidly following damage (i.e. via aberrant repair), the respiratory system is left unprotected, increasing the risk of infection, inflammation and tissue damage. Altered epithelial repair may play an important role in the ongoing respiratory health problems experienced by preterm survivors, including severe respiratory infections throughout early life, or low and declining lung function [1–3]. Deficits are further exacerbated in those with bronchopulmonary dysplasia (BPD). The mechanisms contributing to ongoing respiratory problems are currently unknown, although probably begin in early life. Until now, understanding the role of the preterm epithelial barrier has been limited by a lack of appropriate cellular models. Our study aimed to assess the reparative capacity of the airway epithelium in survivors of preterm birth and its association with early life outcomes, with the hypothesis that preterm airway epithelial cells have an abnormal repair mechanism. Nasal epithelialcells from very preterm infants have a functional defect in their ability to repair beyond the first year of life, and failed repair may be associated with antenatal steroid exposurehttps://bit.ly/39OFJs7
Collapse
Affiliation(s)
- Jessica Hillas
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Nedlands, Australia
| | - Denby J Evans
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Nedlands, Australia.,Occupation and Environment, School of Public Health, Curtin University, Bentley, Australia
| | - Sherlynn Ang
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Nedlands, Australia
| | - Thomas Iosifidis
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Nedlands, Australia.,Occupation and Environment, School of Public Health, Curtin University, Bentley, Australia.,Centre for Cell Therapy and Regenerative Medicine, University of Western Australia, Nedlands, Australia
| | - Luke W Garratt
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Nedlands, Australia
| | - Naomi Hemy
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Nedlands, Australia
| | | | - Shannon J Simpson
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Nedlands, Australia.,School of Physiotherapy and Exercise Science, Curtin University, Bentley, Australia.,These authors contributed equally
| | - Anthony Kicic
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Nedlands, Australia.,Occupation and Environment, School of Public Health, Curtin University, Bentley, Australia.,Centre for Cell Therapy and Regenerative Medicine, University of Western Australia, Nedlands, Australia.,Dept of Respiratory and Sleep Medicine, Perth Children's Hospital, Nedlands, Australia.,These authors contributed equally
| |
Collapse
|
27
|
Bonner K, Scotney E, Saglani S. Factors and mechanisms contributing to the development of preschool wheezing disorders. Expert Rev Respir Med 2021; 15:745-760. [PMID: 33881953 DOI: 10.1080/17476348.2021.1913057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
INTRODUCTION Half of all children will experience an episode of wheezing by their sixth birthday and acute episodes of wheezing in preschool children account for the majority of all childhood hospital admissions for wheeze. Recurrent preschool wheezing associates with early loss of lung function and a life-long impact on lung health. AREAS COVERED We reviewed the literature on PubMed from August 2010-2020 focussing on factors associated with wheeze inception and persistence, paying specific attention to mechanistic studies that have investigated the impact of early life exposures in shaping immune responses in children with underlying susceptibility to wheezing. In particular, the role of early allergen sensitization, respiratory infections, and the impact of the environment on shaping the airway microbiome and resulting immune responses are discussed. EXPERT OPINION There is an abundance of associative data showing the role of in utero and postnatal factors influencing wheeze onset and persistence. However, mechanistic and stratified, biomarker-based interventional studies that confirm these associations are now needed if we are to impact the significant healthcare burden resulting from preschool wheezing disorders.
Collapse
Affiliation(s)
- Katie Bonner
- Inflammation, Repair & Development Section, National Heart & Lung Institute, Imperial College London, London, UK.,Department of Paediatric Respiratory Medicine, Royal Brompton Hospital, London, UK
| | - Elizabeth Scotney
- Inflammation, Repair & Development Section, National Heart & Lung Institute, Imperial College London, London, UK.,Department of Paediatric Respiratory Medicine, Royal Brompton Hospital, London, UK
| | - Sejal Saglani
- Inflammation, Repair & Development Section, National Heart & Lung Institute, Imperial College London, London, UK.,Department of Paediatric Respiratory Medicine, Royal Brompton Hospital, London, UK
| |
Collapse
|
28
|
McErlean P, Kelly A, Dhariwal J, Kirtland M, Watson J, Ranz I, Smith J, Saxena A, Cousins DJ, Van Oosterhout A, Solari R, Edwards MR, Johnston SL, Lavender P. Profiling of H3K27Ac Reveals the Influence of Asthma on the Epigenome of the Airway Epithelium. Front Genet 2020; 11:585746. [PMID: 33362848 PMCID: PMC7758344 DOI: 10.3389/fgene.2020.585746] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 11/03/2020] [Indexed: 12/17/2022] Open
Abstract
Background Asthma is a chronic airway disease driven by complex genetic–environmental interactions. The role of epigenetic modifications in bronchial epithelial cells (BECs) in asthma is poorly understood. Methods We piloted genome-wide profiling of the enhancer-associated histone modification H3K27ac in BECs from people with asthma (n = 4) and healthy controls (n = 3). Results We identified n = 4,321 (FDR < 0.05) regions exhibiting differential H3K27ac enrichment between asthma and health, clustering at genes associated predominately with epithelial processes (EMT). We identified initial evidence of asthma-associated Super-Enhancers encompassing genes encoding transcription factors (TP63) and enzymes regulating lipid metabolism (PTGS1). We integrated published datasets to identify epithelium-specific transcription factors associated with H3K27ac in asthma (TP73) and identify initial relationships between asthma-associated changes in H3K27ac and transcriptional profiles. Finally, we investigated the potential of CRISPR-based approaches to functionally evaluate H3K27ac-asthma landscape in vitro by identifying guide-RNAs capable of targeting acetylation to asthma DERs and inducing gene expression (TLR3). Conclusion Our small pilot study validates genome-wide approaches for deciphering epigenetic mechanisms underlying asthma pathogenesis in the airways.
Collapse
Affiliation(s)
- Peter McErlean
- Peter Gorer Department of Immunobiology, King's College London, London, United Kingdom.,Asthma UK Centre in Allergic Mechanisms of Asthma, London, United Kingdom
| | - Audrey Kelly
- Peter Gorer Department of Immunobiology, King's College London, London, United Kingdom.,Asthma UK Centre in Allergic Mechanisms of Asthma, London, United Kingdom
| | - Jaideep Dhariwal
- Asthma UK Centre in Allergic Mechanisms of Asthma, London, United Kingdom.,Airway Disease Infection Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Max Kirtland
- Peter Gorer Department of Immunobiology, King's College London, London, United Kingdom.,Asthma UK Centre in Allergic Mechanisms of Asthma, London, United Kingdom
| | - Julie Watson
- Peter Gorer Department of Immunobiology, King's College London, London, United Kingdom.,Asthma UK Centre in Allergic Mechanisms of Asthma, London, United Kingdom
| | - Ismael Ranz
- Peter Gorer Department of Immunobiology, King's College London, London, United Kingdom.,Asthma UK Centre in Allergic Mechanisms of Asthma, London, United Kingdom
| | - Janet Smith
- GlaxoSmithKline Allergic Inflammation Discovery Performance Unit, Respiratory Therapy Area, Stevenage, United Kingdom
| | - Alka Saxena
- Genomics Platform, Biomedical Research Centre at Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - David J Cousins
- Asthma UK Centre in Allergic Mechanisms of Asthma, London, United Kingdom.,National Institute for Health Research (NIHR) Respiratory Biomedical Research Unit, Department of Infection, Immunity & Inflammation, Leicester Institute for Lung Health, University of Leicester, Leicester, United Kingdom
| | - Antoon Van Oosterhout
- GlaxoSmithKline Allergic Inflammation Discovery Performance Unit, Respiratory Therapy Area, Stevenage, United Kingdom
| | - Roberto Solari
- Asthma UK Centre in Allergic Mechanisms of Asthma, London, United Kingdom.,Airway Disease Infection Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Michael R Edwards
- Asthma UK Centre in Allergic Mechanisms of Asthma, London, United Kingdom.,Airway Disease Infection Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Sebastian L Johnston
- Asthma UK Centre in Allergic Mechanisms of Asthma, London, United Kingdom.,Airway Disease Infection Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Paul Lavender
- Peter Gorer Department of Immunobiology, King's College London, London, United Kingdom.,Asthma UK Centre in Allergic Mechanisms of Asthma, London, United Kingdom
| |
Collapse
|
29
|
Andersson CK, Iwasaki J, Cook J, Robinson P, Nagakumar P, Mogren S, Fleming L, Bush A, Saglani S, Lloyd CM. Impaired airway epithelial cell wound-healing capacity is associated with airway remodelling following RSV infection in severe preschool wheeze. Allergy 2020; 75:3195-3207. [PMID: 32578219 DOI: 10.1111/all.14466] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 04/06/2020] [Accepted: 04/20/2020] [Indexed: 01/02/2023]
Abstract
BACKGROUND Respiratory syncytial virus (RSV) causes exacerbations of asthma and preschool wheeze (PSW). However, the anti-viral and repair responses of the bronchial epithelium in children with severe therapy-resistant asthma (STRA) and PSW are poorly understood. METHODS Children with STRA (age 12 [6-16] years), PSW (age 2 [1-5] years) and non-asthmatic controls (age 7 [2-14] years) underwent bronchoscopy with endobronchial brushings and biopsies. Anti-viral, wound injury responses were quantified in biopsies and primary bronchial epithelial cells (PBECs) in response to RSV, poly(I:C), house dust mite (HDM) or IL-33 using RT-qPCR, Luminex and live cell imaging. Collagen deposition and tissue expression of epithelial growth factor receptor (EGFR), IL-33 and receptor ST2 were investigated in bronchial biopsies. RESULTS PBECs from STRA and PSW had increased TLR3 gene expression and increased secretion of anti-viral and pro-inflammatory cytokines (IFN-γ, IL-6 and IL-13) in response to RSV compared to controls. Exposure of PBECs to concomitant TLR3 agonist poly(I:C) and HDM resulted in a significant reduction in epithelial cell proliferation in PSW compared to controls. Wound-healing was also impaired in PSW compared to controls at baseline and following IL-33 stimulation. In addition, tissue EGFR expression was significantly reduced in PSW and correlated with collagen deposition in endobronchial biopsies. CONCLUSIONS Despite increased anti-viral responses, preschool children with severe wheeze had impaired airway epithelial proliferative responses following damage. This might be connected to the low expression of EGFR in PSW which may affect epithelial function and contribute to asthma pathogenesis.
Collapse
Affiliation(s)
- Cecilia K. Andersson
- Inflammation, Repair and Development Section National Heart and Lung InstituteImperial College London
- Respiratory Cell Biology Lund University Lund Sweden
| | - Jua Iwasaki
- Inflammation, Repair and Development Section National Heart and Lung InstituteImperial College London
| | - James Cook
- Inflammation, Repair and Development Section National Heart and Lung InstituteImperial College London
- Respiratory Paediatricsthe Royal Brompton and Harefield NHS Trust London UK
| | - Polly Robinson
- Inflammation, Repair and Development Section National Heart and Lung InstituteImperial College London
- Respiratory Paediatricsthe Royal Brompton and Harefield NHS Trust London UK
| | - Prasad Nagakumar
- Inflammation, Repair and Development Section National Heart and Lung InstituteImperial College London
- Respiratory Paediatricsthe Royal Brompton and Harefield NHS Trust London UK
| | - Sofia Mogren
- Respiratory Cell Biology Lund University Lund Sweden
| | - Louise Fleming
- Respiratory Paediatricsthe Royal Brompton and Harefield NHS Trust London UK
| | - Andrew Bush
- Respiratory Paediatricsthe Royal Brompton and Harefield NHS Trust London UK
| | - Sejal Saglani
- Inflammation, Repair and Development Section National Heart and Lung InstituteImperial College London
- Respiratory Paediatricsthe Royal Brompton and Harefield NHS Trust London UK
| | - Clare M. Lloyd
- Inflammation, Repair and Development Section National Heart and Lung InstituteImperial College London
| |
Collapse
|
30
|
Airway Epithelial Dysfunction in Asthma: Relevant to Epidermal Growth Factor Receptors and Airway Epithelial Cells. J Clin Med 2020; 9:jcm9113698. [PMID: 33217964 PMCID: PMC7698733 DOI: 10.3390/jcm9113698] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/11/2020] [Accepted: 11/13/2020] [Indexed: 12/20/2022] Open
Abstract
Airway epithelium plays an important role as the first barrier from external pathogens, including bacteria, viruses, chemical substances, and allergic components. Airway epithelial cells also have pivotal roles as immunological coordinators of defense mechanisms to transfer signals to immunologic cells to eliminate external pathogens from airways. Impaired airway epithelium allows the pathogens to remain in the airway epithelium, which induces aberrant immunological reactions. Dysregulated functions of asthmatic airway epithelium have been reported in terms of impaired wound repair, fragile tight junctions, and excessive proliferation, leading to airway remodeling, which contributes to aberrant airway responses caused by external pathogens. To maintain airway epithelium integrity, a family of epidermal growth factor receptors (EGFR) have pivotal roles in mechanisms of cell growth, proliferation, and differentiation. There are extensive studies focusing on the relation between EGFR and asthma pathophysiology, which describe airway remodeling, airway hypermucus secretion, as well as immunological responses of airway inflammation. Furthermore, the second EGFR family member, erythroblastosis oncogene B2 (ErbB2), has been recognized to be involved with impaired wound recovery and epithelial differentiation in asthmatic airway epithelium. In this review, the roles of the EGFR family in asthmatic airway epithelium are focused on to elucidate the pathogenesis of airway epithelial dysfunction in asthma.
Collapse
|
31
|
The Utility of Resolving Asthma Molecular Signatures Using Tissue-Specific Transcriptome Data. G3-GENES GENOMES GENETICS 2020; 10:4049-4062. [PMID: 32900903 PMCID: PMC7642926 DOI: 10.1534/g3.120.401718] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
An integrative analysis focused on multi-tissue transcriptomics has not been done for asthma. Tissue-specific DEGs remain undetected in many multi-tissue analyses, which influences identification of disease-relevant pathways and potential drug candidates. Transcriptome data from 609 cases and 196 controls, generated using airway epithelium, bronchial, nasal, airway macrophages, distal lung fibroblasts, proximal lung fibroblasts, CD4+ lymphocytes, CD8+ lymphocytes from whole blood and induced sputum samples, were retrieved from Gene Expression Omnibus (GEO). Differentially regulated asthma-relevant genes identified from each sample type were used to identify (a) tissue-specific and tissue-shared asthma pathways, (b) their connection to GWAS-identified disease genes to identify candidate tissue for functional studies, (c) to select surrogate sample for invasive tissues, and finally (d) to identify potential drug candidates via connectivity map analysis. We found that inter-tissue similarity in gene expression was more pronounced at pathway/functional level than at gene level with highest similarity between bronchial epithelial cells and lung fibroblasts, and lowest between airway epithelium and whole blood samples. Although public-domain gene expression data are limited by inadequately annotated per-sample demographic and clinical information which limited the analysis, our tissue-resolved analysis clearly demonstrated relative importance of unique and shared asthma pathways, At the pathway level, IL-1b signaling and ERK signaling were significant in many tissue types, while Insulin-like growth factor and TGF-beta signaling were relevant in only airway epithelial tissue. IL-12 (in macrophages) and Immunoglobulin signaling (in lymphocytes) and chemokines (in nasal epithelium) were the highest expressed pathways. Overall, the IL-1 signaling genes (inflammatory) were relevant in the airway compartment, while pro-Th2 genes including IL-13 and STAT6 were more relevant in fibroblasts, lymphocytes, macrophages and bronchial biopsies. These genes were also associated with asthma in the GWAS catalog. Support Vector Machine showed that DEGs based on macrophages and epithelial cells have the highest and lowest discriminatory accuracy, respectively. Drug (entinostat, BMS-345541) and genetic perturbagens (KLF6, BCL10, INFB1 and BAMBI) negatively connected to disease at multi-tissue level could potentially repurposed for treating asthma. Collectively, our study indicates that the DEGs, perturbagens and disease are connected differentially depending on tissue/cell types. While most of the existing literature describes asthma transcriptome data from individual sample types, the present work demonstrates the utility of multi-tissue transcriptome data. Future studies should focus on collecting transcriptomic data from multiple tissues, age and race groups, genetic background, disease subtypes and on the availability of better-annotated data in the public domain.
Collapse
|
32
|
Azithromycin Partially Mitigates Dysregulated Repair of Lung Allograft Small Airway Epithelium. Transplantation 2020; 104:1166-1176. [PMID: 31985728 DOI: 10.1097/tp.0000000000003134] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
BACKGROUND Dysregulated airway epithelial repair following injury is a proposed mechanism driving posttransplant bronchiolitis obliterans (BO), and its clinical correlate bronchiolitis obliterans syndrome (BOS). This study compared gene and cellular characteristics of injury and repair in large (LAEC) and small (SAEC) airway epithelial cells of transplant patients. METHODS Subjects were recruited at the time of routine bronchoscopy posttransplantation and included patients with and without BOS. Airway epithelial cells were obtained from bronchial and bronchiolar brushing performed under radiological guidance from these patients. In addition, bronchial brushings were also obtained from healthy control subjects comprising of adolescents admitted for elective surgery for nonrespiratory-related conditions. Primary cultures were established, monolayers wounded, and repair assessed (±) azithromycin (1 µg/mL). In addition, proliferative capacity as well as markers of injury and dysregulated repair were also assessed. RESULTS SAEC had a significantly dysregulated repair process postinjury, despite having a higher proliferative capacity than large airway epithelial cells. Addition of azithromycin significantly induced repair in these cells; however, full restitution was not achieved. Expression of several genes associated with epithelial barrier repair (matrix metalloproteinase 7, matrix metalloproteinase 3, the integrins β6 and β8, and β-catenin) were significantly different in epithelial cells obtained from patients with BOS compared to transplant patients without BOS and controls, suggesting an intrinsic defect. CONCLUSIONS Chronic airway injury and dysregulated repair programs are evident in airway epithelium obtained from patients with BOS, particularly with SAEC. We also show that azithromycin partially mitigates this pathology.
Collapse
|
33
|
Do AN, Chun Y, Grishina G, Grishin A, Rogers AJ, Raby BA, Weiss ST, Vicencio A, Schadt EE, Bunyavanich S. Network study of nasal transcriptome profiles reveals master regulator genes of asthma. J Allergy Clin Immunol 2020; 147:879-893. [PMID: 32828590 DOI: 10.1016/j.jaci.2020.07.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 06/19/2020] [Accepted: 07/01/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Nasal transcriptomics can provide an accessible window into asthma pathobiology. OBJECTIVE Our goal was to move beyond gene signatures of asthma to identify master regulator genes that causally regulate genes associated with asthma phenotypes. METHODS We recruited 156 children with severe persistent asthma and controls for nasal transcriptome profiling and applied network-based and probabilistic causal methods to identify severe asthma genes and their master regulators. We then took the same approach in an independent cohort of 190 adults with mild/moderate asthma and controls to identify mild/moderate asthma genes and their master regulators. Comparative analysis of the master regulator genes followed by validation testing in independent children with severe asthma (n = 21) and mild/moderate asthma (n = 154) was then performed. RESULTS Nasal gene signatures for severe persistent asthma and for mild/moderate persistent asthma were identified; both were found to be enriched in coexpression network modules for ciliary function and inflammatory response. By applying probabilistic causal methods to these gene signatures and validation testing in independent cohorts, we identified (1) a master regulator gene common to asthma across severity and ages (FOXJ1); (2) master regulator genes of severe persistent asthma in children (LRRC23, TMEM231, CAPS, PTPRC, and FYB); and (3) master regulator genes of mild/moderate persistent asthma in children and adults (C1orf38 and FMNL1). The identified master regulators were statistically inferred to causally regulate the expression of downstream genes that modulate ciliary function and inflammatory response to influence asthma. CONCLUSION The identified master regulator genes of asthma provide a novel path forward to further uncovering asthma mechanisms and therapy.
Collapse
Affiliation(s)
- Anh N Do
- Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Yoojin Chun
- Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Galina Grishina
- Division of Allergy and Immunology, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Alexander Grishin
- Division of Allergy and Immunology, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Angela J Rogers
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, Calif
| | - Benjamin A Raby
- Division of Pulmonary Medicine, Children's Hospital Boston, Boston, Mass
| | - Scott T Weiss
- Channing Division of Network Medicine, Brigham and Women's Hospital, and Harvard Medical School, Boston, Mass; Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, and Harvard Medical School, Boston, Mass
| | - Alfin Vicencio
- Division of Pulmonary Medicine, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Eric E Schadt
- Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Supinda Bunyavanich
- Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY; Division of Allergy and Immunology, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY.
| |
Collapse
|
34
|
Wang Q, Sundar IK, Li D, Lucas JH, Muthumalage T, McDonough SR, Rahman I. E-cigarette-induced pulmonary inflammation and dysregulated repair are mediated by nAChR α7 receptor: role of nAChR α7 in SARS-CoV-2 Covid-19 ACE2 receptor regulation. Respir Res 2020; 21:154. [PMID: 32552811 PMCID: PMC7301079 DOI: 10.1186/s12931-020-01396-y] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 05/14/2020] [Indexed: 12/13/2022] Open
Abstract
Electronic cigarette (e-cig) vaping is increasing rapidly in the United States, as e-cigs are considered less harmful than combustible cigarettes. However, limited research has been conducted to understand the possible mechanisms that mediate toxicity and pulmonary health effects of e-cigs. We hypothesized that sub-chronic e-cig exposure induces inflammatory response and dysregulated repair/extracellular matrix (ECM) remodeling, which occur through the α7 nicotinic acetylcholine receptor (nAChRα7). Adult wild-type (WT), nAChRα7 knockout (KO), and lung epithelial cell-specific KO (nAChRα7 CreCC10) mice were exposed to e-cig aerosol containing propylene glycol (PG) with or without nicotine. Bronchoalveolar lavage fluids (BALF) and lung tissues were collected to determine e-cig induced inflammatory response and ECM remodeling, respectively. Sub-chronic e-cig exposure with nicotine increased inflammatory cellular influx of macrophages and T-lymphocytes including increased pro-inflammatory cytokines in BALF and increased SARS-Cov-2 Covid-19 ACE2 receptor, whereas nAChRα7 KO mice show reduced inflammatory responses associated with decreased ACE2 receptor. Interestingly, matrix metalloproteinases (MMPs), such as MMP2, MMP8 and MMP9, were altered both at the protein and mRNA transcript levels in female and male KO mice, but WT mice exposed to PG alone showed a sex-dependent phenotype. Moreover, MMP12 was increased significantly in male mice exposed to PG with or without nicotine in a nAChRα7-dependent manner. Additionally, sub-chronic e-cig exposure with or without nicotine altered the abundance of ECM proteins, such as collagen and fibronectin, significantly in a sex-dependent manner, but without the direct role of nAChRα7 gene. Overall, sub-chronic e-cig exposure with or without nicotine affected lung inflammation and repair responses/ECM remodeling, which were mediated by nAChRα7 in a sex-dependent manner.
Collapse
Affiliation(s)
- Qixin Wang
- Department of Environmental Medicine, University of Rochester Medical Center, Box 850, 601 Elmwood Avenue, Rochester, NY, 14642, USA
| | - Isaac K Sundar
- Department of Environmental Medicine, University of Rochester Medical Center, Box 850, 601 Elmwood Avenue, Rochester, NY, 14642, USA
| | - Dongmei Li
- Department of Clinical and Translational Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Joseph H Lucas
- Department of Environmental Medicine, University of Rochester Medical Center, Box 850, 601 Elmwood Avenue, Rochester, NY, 14642, USA
| | - Thivanka Muthumalage
- Department of Environmental Medicine, University of Rochester Medical Center, Box 850, 601 Elmwood Avenue, Rochester, NY, 14642, USA
| | - Samantha R McDonough
- Department of Environmental Medicine, University of Rochester Medical Center, Box 850, 601 Elmwood Avenue, Rochester, NY, 14642, USA
| | - Irfan Rahman
- Department of Environmental Medicine, University of Rochester Medical Center, Box 850, 601 Elmwood Avenue, Rochester, NY, 14642, USA.
| |
Collapse
|
35
|
Wang Q, Sundar I, Li D, Lucas J, Muthumalage T, McDonough S, Rahman I. E-cigarette-Induced Pulmonary Inflammation and Dysregulated Repair are Mediated by nAChR α7 Receptor: Role of nAChR α7 in ACE2 Covid-19 receptor regulation. RESEARCH SQUARE 2020:rs.2.23829. [PMID: 32702718 PMCID: PMC7336696 DOI: 10.21203/rs.2.23829/v2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Electronic cigarette (e-cig) vaping is increasing rapidly in the United States, as e-cigs are considered less harmful than combustible cigarettes. However, limited research has been conducted to understand the possible mechanism that mediate, toxicity and pulmonary health effects of e-cigs. We hypothesized that sub-chronic e-cig exposure induces inflammatory response and dysregulated repair/extracellular matrix (ECM) remodeling, which occur through the α7 nicotinic acetylcholine receptor (nAChR α7). Adult wild-type (WT), nAChRα7 knockout (KO), and lung epithelial cell-specific KO (nAChRα7 CreCC10) mice were exposed to e-cig aerosol containing propylene glycol (PG) with or without nicotine. Bronchoalveolar lavage fluids (BALF) and lungs tissues were collected to determine e-cig induced inflammatory response and ECM remodeling, respectively. Sub-chronic e-cig exposure with nicotine increased the inflammatory cellular influx of macrophages and T-lymphocytes including increased pro-inflammatory cytokines in BALF and increased ACE2 Covid-19 receptor, whereas nAChR α7 KO mice show reduced inflammatory responses associated with decreased ACE2 receptor. Interestingly, matrix metalloproteinases (MMPs), such as MMP2, MMP8, and MMP9 were altered both at the protein and mRNA transcript levels in female and male, but WT mice exposed to PG alone showed a sex-dependent phenotype. Moreover, MMP12 was increased significantly in male mice exposed to PG with or without nicotine in a nAChR α7-dependent manner. Additionally, sub-chronic e-cig exposure with or without nicotine altered the abundance of ECM proteins, such as collagen and fibronectin significantly in a sex-dependent manner, but without the direct role of nAChR α7 gene. Overall, sub-chronic e-cig exposure with or without nicotine affected lung inflammation and repair responses/ECM remodeling, which were mediated by nAChR α7 in a sex-dependent manner.
Collapse
|
36
|
Iosifidis T, Sutanto EN, Buckley AG, Coleman L, Gill EE, Lee AH, Ling KM, Hillas J, Looi K, Garratt LW, Martinovich KM, Shaw NC, Montgomery ST, Kicic-Starcevich E, Karpievitch YV, Le Souëf P, Laing IA, Vijayasekaran S, Lannigan FJ, Rigby PJ, Hancock RE, Knight DA, Stick SM, Kicic A. Aberrant cell migration contributes to defective airway epithelial repair in childhood wheeze. JCI Insight 2020; 5:133125. [PMID: 32208383 DOI: 10.1172/jci.insight.133125] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 03/04/2020] [Indexed: 12/13/2022] Open
Abstract
Abnormal wound repair has been observed in the airway epithelium of patients with chronic respiratory diseases, including asthma. Therapies focusing on repairing vulnerable airways, particularly in early life, present a potentially novel treatment strategy. We report defective lower airway epithelial cell repair to strongly associate with common pre-school-aged and school-aged wheezing phenotypes, characterized by aberrant migration patterns and reduced integrin α5β1 expression. Next generation sequencing identified the PI3K/Akt pathway as the top upstream transcriptional regulator of integrin α5β1, where Akt activation enhanced repair and integrin α5β1 expression in primary cultures from children with wheeze. Conversely, inhibition of PI3K/Akt signaling in primary cultures from children without wheeze reduced α5β1 expression and attenuated repair. Importantly, the FDA-approved drug celecoxib - and its non-COX2-inhibiting analogue, dimethyl-celecoxib - stimulated the PI3K/Akt-integrin α5β1 axis and restored airway epithelial repair in cells from children with wheeze. When compared with published clinical data sets, the identified transcriptomic signature was also associated with viral-induced wheeze exacerbations highlighting the clinical potential of such therapy. Collectively, these results identify airway epithelial restitution via targeting the PI3K-integrin α5β1 axis as a potentially novel therapeutic avenue for childhood wheeze and asthma. We propose that the next step in the therapeutic development process should be a proof-of-concept clinical trial, since relevant animal models to test the crucial underlying premise are unavailable.
Collapse
Affiliation(s)
- Thomas Iosifidis
- Division of Pediatrics and.,Centre for Cell Therapy and Regenerative Medicine, School of Medicine, University of Western Australia, Nedlands, Western Australia, Australia.,Telethon Kids Institute Respiratory Research Centre, Perth, Western Australia, Australia
| | - Erika N Sutanto
- Telethon Kids Institute Respiratory Research Centre, Perth, Western Australia, Australia
| | - Alysia G Buckley
- Centre of Microscopy, Characterisation and Analysis, University of Western Australia, Nedlands, Western Australia, Australia
| | - Laura Coleman
- Division of Pediatrics and.,Telethon Kids Institute Respiratory Research Centre, Perth, Western Australia, Australia
| | - Erin E Gill
- Center for Microbial Diseases Research, University of British Columbia, Vancouver, British Columbia, Canada
| | - Amy H Lee
- Center for Microbial Diseases Research, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kak-Ming Ling
- Division of Pediatrics and.,Telethon Kids Institute Respiratory Research Centre, Perth, Western Australia, Australia
| | - Jessica Hillas
- Telethon Kids Institute Respiratory Research Centre, Perth, Western Australia, Australia
| | - Kevin Looi
- Telethon Kids Institute Respiratory Research Centre, Perth, Western Australia, Australia
| | - Luke W Garratt
- Telethon Kids Institute Respiratory Research Centre, Perth, Western Australia, Australia
| | - Kelly M Martinovich
- Division of Pediatrics and.,Telethon Kids Institute Respiratory Research Centre, Perth, Western Australia, Australia
| | - Nicole C Shaw
- Division of Pediatrics and.,Telethon Kids Institute Respiratory Research Centre, Perth, Western Australia, Australia
| | - Samuel T Montgomery
- Division of Pediatrics and.,Telethon Kids Institute Respiratory Research Centre, Perth, Western Australia, Australia
| | | | - Yuliya V Karpievitch
- Telethon Kids Institute Respiratory Research Centre, Perth, Western Australia, Australia
| | - Peter Le Souëf
- Division of Pediatrics and.,Telethon Kids Institute Respiratory Research Centre, Perth, Western Australia, Australia
| | - Ingrid A Laing
- Division of Pediatrics and.,Telethon Kids Institute Respiratory Research Centre, Perth, Western Australia, Australia
| | | | - Francis J Lannigan
- School of Medicine, Notre Dame University, Fremantle, Western Australia, Australia
| | - Paul J Rigby
- Centre of Microscopy, Characterisation and Analysis, University of Western Australia, Nedlands, Western Australia, Australia
| | - Robert Ew Hancock
- Center for Microbial Diseases Research, University of British Columbia, Vancouver, British Columbia, Canada.,Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Darryl A Knight
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia.,Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, Canada
| | - Stephen M Stick
- Division of Pediatrics and.,Centre for Cell Therapy and Regenerative Medicine, School of Medicine, University of Western Australia, Nedlands, Western Australia, Australia.,Telethon Kids Institute Respiratory Research Centre, Perth, Western Australia, Australia.,Department of Respiratory and Sleep Medicine, Perth Children's Hospital, Nedlands, Western Australia, Australia
| | - Anthony Kicic
- Division of Pediatrics and.,Centre for Cell Therapy and Regenerative Medicine, School of Medicine, University of Western Australia, Nedlands, Western Australia, Australia.,Telethon Kids Institute Respiratory Research Centre, Perth, Western Australia, Australia.,Department of Respiratory and Sleep Medicine, Perth Children's Hospital, Nedlands, Western Australia, Australia.,School of Public Health, Curtin University, Bentley, Western Australia, Australia
| | | | | |
Collapse
|
37
|
Persson C. Humoral First-Line Mucosal Innate Defence in vivo. J Innate Immun 2020; 12:373-386. [PMID: 32203966 DOI: 10.1159/000506515] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 02/11/2020] [Indexed: 12/14/2022] Open
Abstract
Based on observations in vivo in guinea-pig and human airways, this review presents plasma exudation as non-sieved transmission of bulk plasma across an unperturbed mucosa that maintains its normal barrier functions. Several steps have led to the present understanding of plasma exudation as a non-injurious response to mucosal challenges. The implication of a swift appearance of all circulating multipotent protein systems (also including antimicrobial peptides that now are viewed as being exclusively produced by local cells) on challenged, but intact, mucosal surfaces cannot be trivial. Yet, involvement of early plasma exudation responses in innate mucosal immunology has dwelled below the radar. Admittedly, exploration of physiological plasma exudation mechanisms requires in vivo approaches beyond mouse studies. Plasma exudation also lacks the specificity that is a hallmark of biological revelations. These aspects separate plasma exudation from mainstream progress in immunology. The whole idea, presented here, thus competes with strong paradigms currently entertained in the accepted research front. The present focus on humoral innate immunity in vivo further deviates from most discussions, which concern cell-mediated innate defence. Indeed, plasma exudation has emerged as sole in vivo source of major mucosal defence proteins that now are viewed as local cell produce. In conclusion, this review highlights opportunities for complex actions and interactions provided by non-sieved plasma proteins/peptides on the surface of intact mucosal barriers in vivo.
Collapse
Affiliation(s)
- Carl Persson
- Laboratory Medicine, University Hospital of Lund, Lund, Sweden,
| |
Collapse
|
38
|
Nie X, Wei J, Hao Y, Tao J, Li Y, Liu M, Xu B, Li B. Consistent Biomarkers and Related Pathogenesis Underlying Asthma Revealed by Systems Biology Approach. Int J Mol Sci 2019; 20:ijms20164037. [PMID: 31430856 PMCID: PMC6720652 DOI: 10.3390/ijms20164037] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Revised: 08/14/2019] [Accepted: 08/17/2019] [Indexed: 12/13/2022] Open
Abstract
Asthma is a common chronic airway disease worldwide. Due to its clinical and genetic heterogeneity, the cellular and molecular processes in asthma are highly complex and relatively unknown. To discover novel biomarkers and the molecular mechanisms underlying asthma, several studies have been conducted by focusing on gene expression patterns in epithelium through microarray analysis. However, few robust specific biomarkers were identified and some inconsistent results were observed. Therefore, it is imperative to conduct a robust analysis to solve these problems. Herein, an integrated gene expression analysis of ten independent, publicly available microarray data of bronchial epithelial cells from 348 asthmatic patients and 208 healthy controls was performed. As a result, 78 up- and 75 down-regulated genes were identified in bronchial epithelium of asthmatics. Comprehensive functional enrichment and pathway analysis revealed that response to chemical stimulus, extracellular region, pathways in cancer, and arachidonic acid metabolism were the four most significantly enriched terms. In the protein-protein interaction network, three main communities associated with cytoskeleton, response to lipid, and regulation of response to stimulus were established, and the most highly ranked 6 hub genes (up-regulated CD44, KRT6A, CEACAM5, SERPINB2, and down-regulated LTF and MUC5B) were identified and should be considered as new biomarkers. Pathway cross-talk analysis highlights that signaling pathways mediated by IL-4/13 and transcription factor HIF-1α and FOXA1 play crucial roles in the pathogenesis of asthma. Interestingly, three chemicals, polyphenol catechin, antibiotic lomefloxacin, and natural alkaloid boldine, were predicted and may be potential drugs for asthma treatment. Taken together, our findings shed new light on the common molecular pathogenesis mechanisms of asthma and provide theoretical support for further clinical therapeutic studies.
Collapse
Affiliation(s)
- Xiner Nie
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, China
| | - Jinyi Wei
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, China
| | - Youjin Hao
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, China
| | - Jingxin Tao
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, China
| | - Yinghong Li
- School of Biological Information, Chongqing University of Posts and Telecommunications, Chongqing 400065, China
| | - Mingwei Liu
- College of Laboratory Medicine, Chongqing Medical University, Chongqing 400046, China
| | - Boying Xu
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, China
| | - Bo Li
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, China.
| |
Collapse
|
39
|
Singh A, Shannon CP, Kim YW, Yang CX, Balshaw R, Cohen Freue GV, Gauvreau GM, FitzGerald JM, Boulet LP, O'Byrne PM, Tebbutt SJ. Novel Blood-based Transcriptional Biomarker Panels Predict the Late-Phase Asthmatic Response. Am J Respir Crit Care Med 2019; 197:450-462. [PMID: 29087730 DOI: 10.1164/rccm.201701-0110oc] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE The allergen inhalation challenge is used in clinical trials to test the efficacy of new treatments in attenuating the late-phase asthmatic response (LAR) and associated airway inflammation in subjects with allergic asthma. However, not all subjects with allergic asthma develop the LAR after allergen inhalation. Blood-based transcriptional biomarkers that can identify such individuals may help in subject recruitment for clinical trials as well as provide novel molecular insights. OBJECTIVES To identify blood-based transcriptional biomarker panels that can predict an individual's response to allergen inhalation challenge. METHODS We applied RNA sequencing to total RNA from whole blood (n = 36) collected before and after allergen challenge and generated both genome-guided and de novo datasets: genes, gene-isoforms (University of California, Santa Cruz, UCSC Genome Browser), Ensembl, and Trinity. Candidate biomarker panels were validated using the NanoString platform in an independent cohort of 33 subjects. MEASUREMENTS AND MAIN RESULTS The Trinity biomarker panel consisting of known and novel biomarker transcripts had an area under the receiver operating characteristic curve of greater than 0.70 in both the discovery and validation cohorts. The Trinity biomarker panel was useful in predicting the response of subjects that elicited different responses (accuracy between 0.65 and 0.71) and subjects that elicit a dual response (accuracy between 0.70 and 0.75) upon repeated allergen inhalation challenges. CONCLUSIONS Interestingly, the biomarker panel containing novel transcripts successfully validated compared with panels with known, well-characterized genes. These biomarker-blood tests may be used to identify subjects with asthma who develop the LAR, and may also represent members of novel molecular mechanisms that can be targeted for therapy.
Collapse
Affiliation(s)
- Amrit Singh
- 1 Centre for Heart Lung Innovation, St. Paul's Hospital.,3 Department of Pathology and Laboratory Medicine.,2 Prevention of Organ Failure Centre of Excellence, Vancouver, British Columbia, Canada
| | - Casey P Shannon
- 2 Prevention of Organ Failure Centre of Excellence, Vancouver, British Columbia, Canada
| | - Young Woong Kim
- 1 Centre for Heart Lung Innovation, St. Paul's Hospital.,2 Prevention of Organ Failure Centre of Excellence, Vancouver, British Columbia, Canada
| | - Chen Xi Yang
- 1 Centre for Heart Lung Innovation, St. Paul's Hospital.,2 Prevention of Organ Failure Centre of Excellence, Vancouver, British Columbia, Canada
| | - Robert Balshaw
- 4 Centre for Healthcare Innovation, University of Manitoba, Winnipeg, Manitoba, Canada
| | | | - Gail M Gauvreau
- 6 Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - J Mark FitzGerald
- 8 Division of Respiratory Medicine, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada.,7 Vancouver Coastal Health Research Institute, Vancouver General Hospital, Vancouver, British Columbia, Canada; and
| | | | - Paul M O'Byrne
- 6 Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Scott J Tebbutt
- 1 Centre for Heart Lung Innovation, St. Paul's Hospital.,8 Division of Respiratory Medicine, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada.,2 Prevention of Organ Failure Centre of Excellence, Vancouver, British Columbia, Canada
| |
Collapse
|
40
|
Inoue H, Hattori T, Zhou X, Etling EB, Modena BD, Trudeau JB, Holguin F, Wenzel SE. Dysfunctional ErbB2, an EGF receptor family member, hinders repair of airway epithelial cells from asthmatic patients. J Allergy Clin Immunol 2019; 143:2075-2085.e10. [PMID: 30639343 PMCID: PMC6556416 DOI: 10.1016/j.jaci.2018.11.046] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 11/26/2018] [Accepted: 11/30/2018] [Indexed: 01/11/2023]
Abstract
BACKGROUND Genetic and genomic data increasingly point to the airway epithelium as critical to asthma pathogenesis. Epithelial growth factor (EGF) family members play a fundamental role in epithelial differentiation, proliferation, and repair. Although expression of erythroblastosis oncogene B2 (ErbB2) mRNA, an EGF family receptor, was reported to be lower in asthmatic patients, little is understood about its functional role. OBJECTIVE We sought to determine whether decreased ErbB2 activation in freshly isolated human airway epithelial cells (HAECs) from asthmatic patients associated with impaired wound closure in vitro. METHODS An in vitro scratch-wound model of air-liquid interface cultured and freshly isolated HAECs were compared between HAECs from healthy control subjects (HCs) and asthmatic patients in relation to ErbB2. RESULTS Freshly brushed HAECs from asthmatic patients had impaired ErbB2 activation compared with those from HCs. In an in vitro scratch-wound model, HAECs from asthmatic patients showed delayed wound closure compared with HAECs from HCs. Cell proliferation, as assessed based on [3H] thymidine incorporation after wounding, and expression or activation of ErbB2 and cyclin D1 at the leading edge of the wound were lower in HAECs from asthmatic patients and HCs. A selective ErbB2 tyrosine kinase inhibitor, mubritinib, impaired wound closure and decreased cyclin D1 expression in healthy HAECs, with less effect on cells from asthmatic patients, supporting diminished activity in asthmatic patients. CONCLUSION These results implicate a primary defect in the ErbB2 pathway as constraining epithelial repair processes in asthmatic patients. Restoration of homeostatic ErbB2 function should be considered a novel asthma therapeutic target.
Collapse
Affiliation(s)
- Hideki Inoue
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pa; Division of Pulmonary and Allergy Medicine, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Takeshi Hattori
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pa
| | - Xiuxia Zhou
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pa
| | - Emily B Etling
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pa
| | - Brian D Modena
- Molecular and Experimental Medicine, Scripps Research Institute, La Jolla, Calif; Department of Allergy, Asthma and Immunology, Scripps Health, San Diego, Calif
| | - John B Trudeau
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pa
| | - Fernando Holguin
- Division of Medicine-Pulmonary Sciences & Critical Care, University of Colorado School of Medicine, Aurora, Colo
| | - Sally E Wenzel
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pa.
| |
Collapse
|
41
|
Schagen J, Sly PD, Fantino E. Characterizing well-differentiated culture of primary human nasal epithelial cells for use in wound healing assays. J Transl Med 2018; 98:1478-1486. [PMID: 30089850 DOI: 10.1038/s41374-018-0100-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 05/31/2018] [Accepted: 06/05/2018] [Indexed: 12/18/2022] Open
Abstract
The nasal epithelium is the initial contact between the external environment and the respiratory tract and how it responds to noxious stimuli and repairs epithelial damage is important. Growing airway epithelial cells in culture at air-liquid interface allows for a physiologically relevant model of the human upper airways. The aim of the present study was to characterize human primary nasal epithelial cells grown at the air-liquid interface and establish a model for use in wound healing assays. This study determined the time required for full differentiation of nasal epithelial cells in an air-liquid interface culture to be at least 7 weeks using the standardized B-ALI media. Also, a model was established that studied the response to wounding and the effect of EGFR inhibition on this process. Nasal epithelial cultures from healthy subjects were differentiated at air-liquid interface and manually wounded. Wounds were monitored over time to complete closure using a time lapse imaging microscope with cultures identified to have a rate of wound healing above 2.5%/h independent of initial wound size. EGFR inhibition caused the rate of wound healing to drop a significant 4.6%/h with there being no closure of the wound after 48 h. The robust model established in this study will be essential for studying factors influencing wound healing, including host disease status and environmental exposures in the future.
Collapse
Affiliation(s)
- Johanna Schagen
- Children's Lung, Environment and Asthma Research Team, Centre for Children's Health Research, The University of Queensland, Brisbane, Australia
| | - Peter D Sly
- Children's Lung, Environment and Asthma Research Team, Centre for Children's Health Research, The University of Queensland, Brisbane, Australia.
| | - Emmanuelle Fantino
- Children's Lung, Environment and Asthma Research Team, Centre for Children's Health Research, The University of Queensland, Brisbane, Australia
| |
Collapse
|
42
|
Gelfand EW, Schedel M. Molecular Endotypes Contribute to the Heterogeneity of Asthma. Immunol Allergy Clin North Am 2018; 38:655-665. [PMID: 30342586 DOI: 10.1016/j.iac.2018.06.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Diagnosis and management of asthma is commonly implemented based on clinical assessment. Although these nonmolecular biomarkers have been useful, limited resolution of the heterogeneity among asthmatic patients and little information regarding the underlying pathobiology of disease in individuals have been provided. Molecular endotying using global transcriptome expression profiling associated with clinical features of asthma has improved our understanding of disease mechanisms, risk assessment of asthma exacerbations, and treatment responses, especially in patients with type 2 inflammation. Further advances in establishing pathobiological subgroups, bioactive pathways, and true disease endotypes hold potential for a more personalized medical approach in asthmatic patients.
Collapse
Affiliation(s)
- Erwin W Gelfand
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, 1400 Jackson Street, Denver, CO 80206, USA
| | - Michaela Schedel
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, 1400 Jackson Street, Denver, CO 80206, USA.
| |
Collapse
|
43
|
Abstract
Asthma has been the most common chronic disease in children that places a major burden for affected people and their families.An integrated analysis of microarrays studies was performed to identify differentially expressed genes (DEGs) in childhood asthma compared with normal control. We also obtained the differentially methylated genes (DMGs) in childhood asthma according to GEO. The genes that were both differentially expressed and differentially methylated were identified. Functional annotation and protein-protein interaction network construction were performed to interpret biological functions of DEGs. We performed q-RT-PCR to verify the expression of selected DEGs.One DNA methylation and 3 gene expression datasets were obtained. Four hundred forty-one DEGs and 1209 DMGs in childhood asthma were identified. Among which, 16 genes were both differentially expressed and differentially methylated in childhood asthma. Natural killer cell mediated cytotoxicity pathway, Jak-STAT signaling pathway, and Wnt signaling pathway were 3 significantly enriched pathways in childhood asthma according to our KEGG enrichment analysis. The PPI network of top 20 up- and downregulated DEGs consisted of 822 nodes and 904 edges and 2 hub proteins (UBQLN4 and MID2) were identified. The expression of 8 DEGs (GZMB, FGFBP2, CLC, TBX21, ALOX15, IL12RB2, UBQLN4) was verified by qRT-PCR and only the expression of GZMB and FGFBP2 was inconsistent with our integrated analysis.Our finding was helpful to elucidate the underlying mechanism of childhood asthma and develop new potential diagnostic biomarker and provide clues for drug design.
Collapse
Affiliation(s)
| | | | - Yu-Hua Mu
- Department of General Surgery, Rizhao People's Hospital, Rizhao, China
| | | |
Collapse
|
44
|
Xie B, Laxman B, Hashemifar S, Stern R, Gilliam TC, Maltsev N, White SR. Chemokine expression in the early response to injury in human airway epithelial cells. PLoS One 2018; 13:e0193334. [PMID: 29534074 PMCID: PMC5849294 DOI: 10.1371/journal.pone.0193334] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 02/08/2018] [Indexed: 12/22/2022] Open
Abstract
Basal airway epithelial cells (AEC) constitute stem/progenitor cells within the central airways and respond to mucosal injury in an ordered sequence of spreading, migration, proliferation, and differentiation to needed cell types. However, dynamic gene transcription in the early events after mucosal injury has not been studied in AEC. We examined gene expression using microarrays following mechanical injury (MI) in primary human AEC grown in submersion culture to generate basal cells and in the air-liquid interface to generate differentiated AEC (dAEC) that include goblet and ciliated cells. A select group of ~150 genes was in differential expression (DE) within 2-24 hr after MI, and enrichment analysis of these genes showed over-representation of functional categories related to inflammatory cytokines and chemokines. Network-based gene prioritization and network reconstruction using the PINTA heat kernel diffusion algorithm demonstrated highly connected networks that were richer in differentiated AEC compared to basal cells. Similar experiments done in basal AEC collected from asthmatic donor lungs demonstrated substantial changes in DE genes and functional categories related to inflammation compared to basal AEC from normal donors. In dAEC, similar but more modest differences were observed. We demonstrate that the AEC transcription signature after MI identifies genes and pathways that are important to the initiation and perpetuation of airway mucosal inflammation. Gene expression occurs quickly after injury and is more profound in differentiated AEC, and is altered in AEC from asthmatic airways. Our data suggest that the early response to injury is substantially different in asthmatic airways, particularly in basal airway epithelial cells.
Collapse
Affiliation(s)
- Bingqing Xie
- Department of Human Genetics, University of Chicago, Chicago, IL, United States of America
- Illinois Institute of Technology, Chicago, IL, United States of America
| | - Bharathi Laxman
- Department of Medicine, University of Chicago, Chicago, IL, United States of America
| | - Somaye Hashemifar
- Department of Human Genetics, University of Chicago, Chicago, IL, United States of America
- Toyota Technological Institute at Chicago, Chicago, IL, United States of America
| | - Randi Stern
- Department of Medicine, University of Chicago, Chicago, IL, United States of America
| | - T. Conrad Gilliam
- Department of Human Genetics, University of Chicago, Chicago, IL, United States of America
| | - Natalia Maltsev
- Department of Human Genetics, University of Chicago, Chicago, IL, United States of America
| | - Steven R. White
- Department of Medicine, University of Chicago, Chicago, IL, United States of America
| |
Collapse
|
45
|
Looi K, Buckley AG, Rigby PJ, Garratt LW, Iosifidis T, Zosky GR, Larcombe AN, Lannigan FJ, Ling KM, Martinovich KM, Kicic-Starcevich E, Shaw NC, Sutanto EN, Knight DA, Kicic A, Stick SM. Effects of human rhinovirus on epithelial barrier integrity and function in children with asthma. Clin Exp Allergy 2018; 48:513-524. [PMID: 29350877 DOI: 10.1111/cea.13097] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 11/13/2017] [Accepted: 11/21/2017] [Indexed: 11/29/2022]
Abstract
BACKGROUND Bronchial epithelial tight junctions (TJ) have been extensively assessed in healthy airway epithelium. However, no studies have yet assessed the effect of human rhinovirus (HRV) infection on the expression and resultant barrier function in epithelial tight junctions (TJ) in childhood asthma. OBJECTIVES To investigate the impact of HRV infection on airway epithelial TJ expression and barrier function in airway epithelial cells (AECs) of children with and without asthma. Furthermore, to test the hypothesis that barrier integrity and function is compromised to a greater extent by HRV in AECs from asthmatic children. METHODS Primary AECs were obtained from children with and without asthma, differentiated into air-liquid interface (ALI) cultures and infected with rhinovirus. Expression of claudin-1, occludin and zonula occluden-1 (ZO-1) was assessed via qPCR, immunocytochemistry (ICC), in-cell western (ICW) and confocal microscopy. Barrier function was assessed by transepithelial electrical resistance (TER; RT ) and permeability to fluorescent dextran. RESULTS Basal TJ gene expression of claudin-1 and occludin was significantly upregulated in asthmatic children compared to non-asthmatics; however, no difference was seen with ZO-1. Interestingly, claudin-1, occludin and ZO-1 protein expression was significantly reduced in AEC of asthmatic children compared to non-asthmatic controls suggesting possible post-transcriptional inherent differences. HRV infection resulted in a transient dissociation of TJ and airway barrier integrity in non-asthmatic children. Although similar dissociation of TJ was observed in asthmatic children, a significant and sustained reduction in TJ expression concurrent with both a significant decrease in TER and an increase in permeability in asthmatic children was observed. CONCLUSION This study demonstrates novel intrinsic differences in TJ gene and protein expression between AEC of children with and without asthma. Furthermore, it correlates directly the relationship between HRV infection and the resultant dissociation of epithelial TJ that causes a continued altered barrier function in children with asthma.
Collapse
Affiliation(s)
- K Looi
- School of Paediatrics and Child Health, University of Western Australia, Nedlands, WA, Australia.,Telethon Kids Institute, University of Western Australia, Subiaco, WA, Australia
| | - A G Buckley
- Centre for Microscopy, Characterisation and Analysis (CMCA), University of Western Australia, Crawley, WA, Australia
| | - P J Rigby
- Centre for Microscopy, Characterisation and Analysis (CMCA), University of Western Australia, Crawley, WA, Australia
| | - L W Garratt
- School of Paediatrics and Child Health, University of Western Australia, Nedlands, WA, Australia.,Telethon Kids Institute, University of Western Australia, Subiaco, WA, Australia
| | - T Iosifidis
- School of Paediatrics and Child Health, University of Western Australia, Nedlands, WA, Australia.,Centre for Cell Therapy and Regenerative Medicine, University of Western Australia, Nedlands, WA, Australia
| | - G R Zosky
- School of Medicine, Faculty of Health, University of Tasmania, Hohart, TAS, Australia
| | - A N Larcombe
- Telethon Kids Institute, University of Western Australia, Subiaco, WA, Australia.,Occupation and Environment, School of Public Health, Curtin University, Perth, WA, Australia
| | - F J Lannigan
- School of Paediatrics and Child Health, University of Western Australia, Nedlands, WA, Australia.,School of Medicine, Notre Dame University, Fremantle, WA, Australia
| | - K-M Ling
- Telethon Kids Institute, University of Western Australia, Subiaco, WA, Australia
| | - K M Martinovich
- Telethon Kids Institute, University of Western Australia, Subiaco, WA, Australia
| | - E Kicic-Starcevich
- Telethon Kids Institute, University of Western Australia, Subiaco, WA, Australia.,Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, WA, Australia
| | - N C Shaw
- Telethon Kids Institute, University of Western Australia, Subiaco, WA, Australia
| | - E N Sutanto
- Telethon Kids Institute, University of Western Australia, Subiaco, WA, Australia.,Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, WA, Australia
| | - D A Knight
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia.,Priority Research Centre for Asthma and Respiratory Disease, Hunter Medical Research Institute, Newcastle, NSW, Australia.,Department of Anaesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| | - A Kicic
- School of Paediatrics and Child Health, University of Western Australia, Nedlands, WA, Australia.,Telethon Kids Institute, University of Western Australia, Subiaco, WA, Australia.,Centre for Cell Therapy and Regenerative Medicine, University of Western Australia, Nedlands, WA, Australia.,Occupation and Environment, School of Public Health, Curtin University, Perth, WA, Australia.,Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, WA, Australia
| | - S M Stick
- School of Paediatrics and Child Health, University of Western Australia, Nedlands, WA, Australia.,Telethon Kids Institute, University of Western Australia, Subiaco, WA, Australia.,Centre for Cell Therapy and Regenerative Medicine, University of Western Australia, Nedlands, WA, Australia.,Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, WA, Australia
| |
Collapse
|
46
|
Buckley AG, Looi K, Iosifidis T, Ling KM, Sutanto EN, Martinovich KM, Kicic-Starcevich E, Garratt LW, Shaw NC, Lannigan FJ, Larcombe AN, Zosky G, Knight DA, Rigby PJ, Kicic A, Stick SM. Visualisation of Multiple Tight Junctional Complexes in Human Airway Epithelial Cells. Biol Proced Online 2018; 20:3. [PMID: 29434527 PMCID: PMC5793437 DOI: 10.1186/s12575-018-0070-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 01/22/2018] [Indexed: 01/30/2023] Open
Abstract
Background Apically located tight junctions in airway epithelium perform a fundamental role in controlling macromolecule migration through paracellular spaces. Alterations in their expression may lead to disruptions in barrier integrity, which subsequently facilitates entry of potential bacterial and other pathogens into the host. Furthermore, there is emerging evidence that the barrier integrity of the airway in certain airway inflammatory diseases may be altered. However, there is little consensus on the way this is assessed and measured and the type of cells used to achieve this. Methods Here, we assessed four fixation methods including; (i) 4% (v/v) paraformaldehyde; (ii) 100% methanol; (iii) acetone or; (iv) 1:1 methanol: acetone. Pre-extraction with Triton X-100 was also performed and assessed on cells prior to fixation with either methanol or paraformaldehyde. Cells were also permeabilized with 0.1% (v/v) Saponin in 1× TBS following fixation and subsequently stained for tight junction proteins. Confocal microscopy was then used to visualise, compare and evaluate staining intensity of the tight junctional complexes in order to determine a standardised workflow of reproducible staining. Results Positive staining was observed following methanol fixation for claudin-1 and ZO-1 tight junction proteins but no staining was detected for occludin in 16HBE14o- cells. Combinatorial fixation with methanol and acetone also produced consistent positive staining for both occludin and ZO-1 tight junction proteins in these cells. When assessed using primary cells cultured at air-liquid interface, similar positive staining for claudin-1 and ZO-1 was observed following methanol fixation, while similar positive staining for occludin and ZO-1 was observed following the same combinatorial fixation with methanol and acetone. Conclusions The present study demonstrates the importance of a personalised approach to optimise staining for the visualisation of different tight junction proteins. Of significance, the workflow, once optimised, can readily be translated into primary airway epithelial cell air-liquid interface cultures where it can be used to assess barrier integrity in chronic lung diseases.
Collapse
Affiliation(s)
- Alysia G Buckley
- 1Centre of Microscopy, Characterisation and Analysis, The University of Western Australia, Crawley, Western Australia 6009 Australia
| | - Kevin Looi
- 2School of Paediatrics and Child Health, The University of Western Australia, Nedlands, Western Australia 6009 Australia
| | - Thomas Iosifidis
- 2School of Paediatrics and Child Health, The University of Western Australia, Nedlands, Western Australia 6009 Australia.,3Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia, Nedlands, Western Australia 6009 Australia
| | - Kak-Ming Ling
- 4Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Crawley, Western Australia 6009 Australia
| | - Erika N Sutanto
- 4Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Crawley, Western Australia 6009 Australia.,5Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, Western Australia 6001 Australia
| | - Kelly M Martinovich
- 4Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Crawley, Western Australia 6009 Australia
| | - Elizabeth Kicic-Starcevich
- 4Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Crawley, Western Australia 6009 Australia
| | - Luke W Garratt
- 2School of Paediatrics and Child Health, The University of Western Australia, Nedlands, Western Australia 6009 Australia
| | - Nicole C Shaw
- 4Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Crawley, Western Australia 6009 Australia
| | - Francis J Lannigan
- School of Medicine, Notre Dame University, Fremantle, Western Australia 6160 Australia
| | - Alexander N Larcombe
- 4Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Crawley, Western Australia 6009 Australia
| | - Graeme Zosky
- 4Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Crawley, Western Australia 6009 Australia.,7School of Medicine, Faculty of Health, University of Tasmania, Hobart, Tasmania 7000 Australia
| | - Darryl A Knight
- 8School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales Australia.,9Priority Research Centre for Asthma and Respiratory Disease, Hunter Medical Research Institute, Newcastle, New South Wales Australia.,10Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, Canada
| | - Paul J Rigby
- 1Centre of Microscopy, Characterisation and Analysis, The University of Western Australia, Crawley, Western Australia 6009 Australia
| | - Anthony Kicic
- 2School of Paediatrics and Child Health, The University of Western Australia, Nedlands, Western Australia 6009 Australia.,3Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia, Nedlands, Western Australia 6009 Australia.,4Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Crawley, Western Australia 6009 Australia.,5Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, Western Australia 6001 Australia.,11School of Public Health, Curtin University, Bentley, Western Australia 6102 Australia.,12Telethon Kids Institute, Subiaco, Perth, Western Australia 6008 Australia
| | - Stephen M Stick
- 2School of Paediatrics and Child Health, The University of Western Australia, Nedlands, Western Australia 6009 Australia.,3Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia, Nedlands, Western Australia 6009 Australia.,4Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Crawley, Western Australia 6009 Australia.,5Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, Western Australia 6001 Australia
| |
Collapse
|
47
|
Martinovich KM, Iosifidis T, Buckley AG, Looi K, Ling KM, Sutanto EN, Kicic-Starcevich E, Garratt LW, Shaw NC, Montgomery S, Lannigan FJ, Knight DA, Kicic A, Stick SM. Conditionally reprogrammed primary airway epithelial cells maintain morphology, lineage and disease specific functional characteristics. Sci Rep 2017; 7:17971. [PMID: 29269735 PMCID: PMC5740081 DOI: 10.1038/s41598-017-17952-4] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 12/04/2017] [Indexed: 01/19/2023] Open
Abstract
Current limitations to primary cell expansion led us to test whether airway epithelial cells derived from healthy children and those with asthma and cystic fibrosis (CF), co-cultured with an irradiated fibroblast feeder cell in F-medium containing 10 µM ROCK inhibitor could maintain their lineage during expansion and whether this is influenced by underlying disease status. Here, we show that conditionally reprogrammed airway epithelial cells (CRAECs) can be established from both healthy and diseased phenotypes. CRAECs can be expanded, cryopreserved and maintain phenotypes over at least 5 passages. Population doublings of CRAEC cultures were significantly greater than standard cultures, but maintained their lineage characteristics. CRAECs from all phenotypes were also capable of fully differentiating at air-liquid interface (ALI) and maintained disease specific characteristics including; defective CFTR channel function cultures and the inability to repair wounds. Our findings indicate that CRAECs derived from children maintain lineage, phenotypic and importantly disease-specific functional characteristics over a specified passage range.
Collapse
Affiliation(s)
- Kelly M Martinovich
- Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Crawley, Western Australia, Australia
| | - Thomas Iosifidis
- School of Paediatrics and Child Health, The University of Western Australia, Crawley, Western Australia, Australia.,Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia, Nedlands, Western Australia, Australia
| | - Alysia G Buckley
- Centre of Microscopy, Characterisation and Analysis, The University of Western Australia, Crawley, Western Australia, Australia
| | - Kevin Looi
- Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Crawley, Western Australia, Australia
| | - Kak-Ming Ling
- Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Crawley, Western Australia, Australia
| | - Erika N Sutanto
- Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Crawley, Western Australia, Australia
| | - Elizabeth Kicic-Starcevich
- Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Crawley, Western Australia, Australia
| | - Luke W Garratt
- Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Crawley, Western Australia, Australia
| | - Nicole C Shaw
- Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Crawley, Western Australia, Australia
| | - Samuel Montgomery
- Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Crawley, Western Australia, Australia
| | - Francis J Lannigan
- School of Paediatrics and Child Health, The University of Western Australia, Crawley, Western Australia, Australia
| | - Darryl A Knight
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia.,Priority Research Centre for Asthma and Respiratory Disease, Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, Canada
| | - Anthony Kicic
- Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Crawley, Western Australia, Australia. .,School of Paediatrics and Child Health, The University of Western Australia, Crawley, Western Australia, Australia. .,Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia, Nedlands, Western Australia, Australia. .,Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, Western Australia, Australia. .,Occupation and Environment, School of Public Health, Curtin University, Perth, Western Australia, Australia.
| | - Stephen M Stick
- Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Crawley, Western Australia, Australia.,School of Paediatrics and Child Health, The University of Western Australia, Crawley, Western Australia, Australia.,Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia, Nedlands, Western Australia, Australia.,Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, Western Australia, Australia
| |
Collapse
|
48
|
Jones AC, Bosco A. Using Network Analysis to Understand Severe Asthma Phenotypes. Am J Respir Crit Care Med 2017; 195:1409-1411. [PMID: 28569573 DOI: 10.1164/rccm.201612-2572ed] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Anya C Jones
- 1 Telethon Kids Institute The University of Western Australia Perth, Australia
| | - Anthony Bosco
- 1 Telethon Kids Institute The University of Western Australia Perth, Australia
| |
Collapse
|
49
|
Loffredo LF, Abdala-Valencia H, Anekalla KR, Cuervo-Pardo L, Gottardi CJ, Berdnikovs S. Beyond epithelial-to-mesenchymal transition: Common suppression of differentiation programs underlies epithelial barrier dysfunction in mild, moderate, and severe asthma. Allergy 2017; 72:1988-2004. [PMID: 28599074 DOI: 10.1111/all.13222] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/05/2017] [Indexed: 01/11/2023]
Abstract
BACKGROUND Epithelial barrier dysfunction is a central feature in the pathogenesis of allergic disease. Epithelial-to-mesenchymal transition (EMT) has been proposed as one mechanism afflicting barrier in asthma. However, genes and pathways involved in aberrant epithelial-mesenchymal signaling, and their relationship to asthma severity, are poorly understood. METHODS We used unbiased gene network analysis to evaluate functional convergence in epithelial gene expression signatures across multiple public access transcriptomics datasets of human asthma, followed by text mining to evaluate functional marker relevance of discovered genes. We objectively confirmed these findings in epithelial brushings and primary asthmatic epithelial cells cultured in different biological contexts. RESULTS We found a striking suppression of epithelial differentiation in asthma, overrepresented by insufficiency in insulin and Notch signaling, but with the absence of conventional EMT markers. We identified EFNB2, FGFR1, FGFR2, INSR, IRS2, NOTCH2, TLE1, and NTRK2 as novel markers central to dysregulation of epithelial-mesenchymal signaling, but surprisingly overlooked in asthma research. We found that this "core" signature of asthma is shared by mild, moderate, and severe forms of disease, progressing with severity. Loss of epithelial differentiation induced by insulin deprivation in normal human bronchial epithelial cells cultured in organotypic conditions closely approximated gene expression in asthmatic epithelial brushings. CONCLUSIONS The comparative analysis of publically available transcriptomes demonstrated that epithelial barrier dysfunction in asthma is characterized by persistent underlying de-differentiation program with complex etiology. The lasting alteration of the asthmatic epithelial cell transcriptome implicates regulation involving metabolism and epigenetics, beyond EMT driven by injury and repair in chronic inflammation.
Collapse
Affiliation(s)
- L. F. Loffredo
- Division of Allergy and Immunology; Department of Medicine; Northwestern University Feinberg School of Medicine; Chicago IL USA
| | - H. Abdala-Valencia
- Division of Pulmonary and Critical Care; Department of Medicine; Northwestern University Feinberg School of Medicine; Chicago IL USA
| | - K. R. Anekalla
- Division of Pulmonary and Critical Care; Department of Medicine; Northwestern University Feinberg School of Medicine; Chicago IL USA
| | - L. Cuervo-Pardo
- Division of Allergy and Immunology; Department of Medicine; Northwestern University Feinberg School of Medicine; Chicago IL USA
| | - C. J. Gottardi
- Division of Pulmonary and Critical Care; Department of Medicine; Northwestern University Feinberg School of Medicine; Chicago IL USA
| | - S. Berdnikovs
- Division of Allergy and Immunology; Department of Medicine; Northwestern University Feinberg School of Medicine; Chicago IL USA
| |
Collapse
|
50
|
Fang F, Pan J, Li Y, Li Y, Feng X, Wang J. Identification of potential transcriptomic markers in developing asthma: An integrative analysis of gene expression profiles. Mol Immunol 2017; 92:38-44. [PMID: 29031950 DOI: 10.1016/j.molimm.2017.09.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Revised: 09/24/2017] [Accepted: 09/30/2017] [Indexed: 12/14/2022]
Abstract
The goal of this study was to identify potential transcriptomic markers in developing asthma by an integrative analysis of multiple public microarray data sets. Using the R software and bioconductor packages, we performed a statistical analysis to identify differentially expressed (DE) genes in asthma, and further performed functional interpretation (enrichment analysis and co-expression network construction) and classification quality evaluation of the DE genes identified. 3 microarray datasets (192 cases and 91 controls in total) were collected for this analysis. 62 DE genes were identified in asthma, among which 43 genes were up-regulated and 19 genes were down-regulated. The up-regulated gene with the highest Log2 Fold Change (LFC) was CLCA1 (LFC=2.81). The down-regulated gene with the highest absolute LFC was BPIFA1 (LFC=-1.45). Enrichment analysis revealed that those DE genes strongly associated with proteolysis, retina homeostasis, humoral immune response, and salivary secretion. A support vector machine classifier (asthma versus healthy control) was also trained based on DE genes. In conclusion, the consistently DE genes identified in this study are suggested as candidate transcriptomic markers for asthma diagnosis, and provide novel insights into the pathogenesis of asthma.
Collapse
Affiliation(s)
- Fang Fang
- Institute of Pediatric Research, Children's Hospital of Soochow University, 92 Zhongnan Street, Suzhou 215025, China.
| | - Jian Pan
- Institute of Pediatric Research, Children's Hospital of Soochow University, 92 Zhongnan Street, Suzhou 215025, China
| | - Yanhong Li
- Institute of Pediatric Research, Children's Hospital of Soochow University, 92 Zhongnan Street, Suzhou 215025, China
| | - Yiping Li
- Institute of Pediatric Research, Children's Hospital of Soochow University, 92 Zhongnan Street, Suzhou 215025, China
| | - Xing Feng
- Institute of Pediatric Research, Children's Hospital of Soochow University, 92 Zhongnan Street, Suzhou 215025, China
| | - Jian Wang
- Institute of Pediatric Research, Children's Hospital of Soochow University, 92 Zhongnan Street, Suzhou 215025, China.
| |
Collapse
|