1
|
Marriott H, Duchesne M, Moitra S, Okoye I, Gerla L, Mayers I, Moolji J, Adatia A, Lacy P. Upper Airway Alarmin Cytokine Expression in Asthma of Different Severities. J Clin Med 2024; 13:3721. [PMID: 38999286 PMCID: PMC11242732 DOI: 10.3390/jcm13133721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 06/21/2024] [Accepted: 06/24/2024] [Indexed: 07/14/2024] Open
Abstract
Background: The secretion of alarmin cytokines by epithelial cells, including thymic stromal lymphopoietin (TSLP), interleukin (IL)-25, and IL-33, initiates inflammatory cascades in asthma. However, alarmin cytokine expression in the upper airways in asthma remains largely unknown. Methods: We recruited 40 participants with asthma into four groups as per the Global Initiative for Asthma (GINA) steps (10 in each group of GINA 1/2, 3, 4, and 5). Cells were derived from nasal, buccal, and throat brushings. Intracellular cytokine expression (TSLP, IL-25, and IL-33) was assessed by flow cytometry in cytokeratin 8+ (Ck8+) epithelial cells immediately following collection. Results: TSLP was significantly increased (p < 0.001) in GINA 5 patients across nasal, buccal, and throat Ck8+ epithelial cells, while IL-25 was elevated in nasal and throat samples (p < 0.003), and IL-33 levels were variable, compared with GINA 1-4 patients. Individual GINA subgroup comparison showed that TSLP levels in nasal samples from GINA 5 patients were significantly (p = 0.03) elevated but did not differ between patients with and without nasal comorbidities. IL-25 and IL-33 (obtained from nasal, buccal, and throat samples) were not significantly different in individual groups. Conclusions: Our study demonstrates for the first time that Ck8+ nasal epithelial cells from GINA 5 asthma patients express elevated levels of TSLP.
Collapse
Affiliation(s)
- Hazel Marriott
- Division of Pulmonary Medicine, Department of Medicine, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Marc Duchesne
- Division of Pulmonary Medicine, Department of Medicine, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Subhabrata Moitra
- Division of Pulmonary Medicine, Department of Medicine, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Isobel Okoye
- Division of Pulmonary Medicine, Department of Medicine, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Luke Gerla
- Division of Pulmonary Medicine, Department of Medicine, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Irvin Mayers
- Division of Pulmonary Medicine, Department of Medicine, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Jalal Moolji
- Division of Pulmonary Medicine, Department of Medicine, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Adil Adatia
- Division of Pulmonary Medicine, Department of Medicine, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Paige Lacy
- Division of Pulmonary Medicine, Department of Medicine, University of Alberta, Edmonton, AB T6G 2R3, Canada
| |
Collapse
|
2
|
Rowe T, Davis W, Wentworth DE, Ross T. Differential interferon responses to influenza A and B viruses in primary ferret respiratory epithelial cells. J Virol 2024; 98:e0149423. [PMID: 38294251 PMCID: PMC10878268 DOI: 10.1128/jvi.01494-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 12/02/2023] [Indexed: 02/01/2024] Open
Abstract
Influenza B viruses (IBV) cocirculate with influenza A viruses (IAV) and cause periodic epidemics of disease, yet antibody and cellular responses following IBV infection are less well understood. Using the ferret model for antisera generation for influenza surveillance purposes, IAV resulted in robust antibody responses following infection, whereas IBV required an additional booster dose, over 85% of the time, to generate equivalent antibody titers. In this study, we utilized primary differentiated ferret nasal epithelial cells (FNECs) which were inoculated with IAV and IBV to study differences in innate immune responses which may result in differences in adaptive immune responses in the host. FNECs were inoculated with IAV (H1N1pdm09 and H3N2 subtypes) or IBV (B/Victoria and B/Yamagata lineages) and assessed for 72 h. Cells were analyzed for gene expression by quantitative real-time PCR, and apical and basolateral supernatants were assessed for virus kinetics and interferon (IFN), respectively. Similar virus kinetics were observed with IAV and IBV in FNECs. A comparison of gene expression and protein secretion profiles demonstrated that IBV-inoculated FNEC expressed delayed type-I/II IFN responses and reduced type-III IFN secretion compared to IAV-inoculated cells. Concurrently, gene expression of Thymic Stromal Lymphopoietin (TSLP), a type-III IFN-induced gene that enhances adaptive immune responses, was significantly downregulated in IBV-inoculated FNECs. Significant differences in other proinflammatory and adaptive genes were suppressed and delayed following IBV inoculation. Following IBV infection, ex vivo cell cultures derived from the ferret upper respiratory tract exhibited reduced and delayed innate responses which may contribute to reduced antibody responses in vivo.IMPORTANCEInfluenza B viruses (IBV) represent nearly one-quarter of all human influenza cases and are responsible for significant clinical and socioeconomic impacts but do not pose the same pandemic risks as influenza A viruses (IAV) and have thus received much less attention. IBV accounts for greater severity and deaths in children, and vaccine efficacy remains low. The ferret can be readily infected with human clinical isolates and demonstrates a similar course of disease and immune responses. IBV, however, generates lower antibodies in ferrets than IAV following the challenge. To determine whether differences in initial innate responses following infection may affect the development of robust adaptive immune responses, ferret respiratory tract cells were isolated, infected with IAV/IBV, and compared. Understanding the differences in the initial innate immune responses to IAV and IBV may be important in the development of more effective vaccines and interventions to generate more robust protective immune responses.
Collapse
Affiliation(s)
- Thomas Rowe
- Centers for Disease Control and Prevention, Influenza Division, Atlanta, Georgia, USA
- Center for Vaccines and Immunology, University of Georgia, Athens, Georgia, USA
| | - William Davis
- Centers for Disease Control and Prevention, Influenza Division, Atlanta, Georgia, USA
| | - David E. Wentworth
- Centers for Disease Control and Prevention, Influenza Division, Atlanta, Georgia, USA
| | - Ted Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
3
|
Cuthbertson L, Löber U, Ish-Horowicz JS, McBrien CN, Churchward C, Parker JC, Olanipekun MT, Burke C, McGowan A, Davies GA, Lewis KE, Hopkin JM, Chung KF, O'Carroll O, Faul J, Creaser-Thomas J, Andrews M, Ghosal R, Piatek S, Willis-Owen SAG, Bartolomaeus TUP, Birkner T, Dwyer S, Kumar N, Turek EM, William Musk A, Hui J, Hunter M, James A, Dumas ME, Filippi S, Cox MJ, Lawley TD, Forslund SK, Moffatt MF, Cookson WOC. Genomic attributes of airway commensal bacteria and mucosa. Commun Biol 2024; 7:171. [PMID: 38347162 PMCID: PMC10861553 DOI: 10.1038/s42003-024-05840-3] [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: 01/30/2023] [Accepted: 01/22/2024] [Indexed: 02/15/2024] Open
Abstract
Microbial communities at the airway mucosal barrier are conserved and highly ordered, in likelihood reflecting co-evolution with human host factors. Freed of selection to digest nutrients, the airway microbiome underpins cognate management of mucosal immunity and pathogen resistance. We show here the initial results of systematic culture and whole-genome sequencing of the thoracic airway bacteria, identifying 52 novel species amongst 126 organisms that constitute 75% of commensals typically present in heathy individuals. Clinically relevant genes encode antimicrobial synthesis, adhesion and biofilm formation, immune modulation, iron utilisation, nitrous oxide (NO) metabolism and sphingolipid signalling. Using whole-genome content we identify dysbiotic features that may influence asthma and chronic obstructive pulmonary disease. We match isolate gene content to transcripts and metabolites expressed late in airway epithelial differentiation, identifying pathways to sustain host interactions with microbiota. Our results provide a systematic basis for decrypting interactions between commensals, pathogens, and mucosa in lung diseases of global significance.
Collapse
Affiliation(s)
- Leah Cuthbertson
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Ulrike Löber
- Max Delbrück Center for Molecular Medicine (MDC), 13125, Berlin, Germany
- Experimental and Clinical Research Center, A Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Lindenberger Weg 80, 13125, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site, 10785, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117, Berlin, Germany
| | - Jonathan S Ish-Horowicz
- National Heart and Lung Institute, Imperial College London, London, UK
- Department of Mathematics, Imperial College London, London, UK
| | - Claire N McBrien
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Colin Churchward
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Jeremy C Parker
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | - Conor Burke
- Department of Respiratory Medicine, Connolly Hospital, Dublin, Ireland
| | - Aisling McGowan
- Department of Respiratory Medicine, Connolly Hospital, Dublin, Ireland
| | - Gwyneth A Davies
- Population Data Science and Health Data Research UK BREATHE Hub, Swansea University Medical School, Swansea University, Swansea, UK
- College of Medicine, Institute of Life Science, Swansea University, Swansea, UK
| | - Keir E Lewis
- College of Medicine, Institute of Life Science, Swansea University, Swansea, UK
- Respiratory Medicine, Hywel Dda University Health Board, Llanelli, UK
| | - Julian M Hopkin
- College of Medicine, Institute of Life Science, Swansea University, Swansea, UK
| | - Kian Fan Chung
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Orla O'Carroll
- Department of Respiratory Medicine, Connolly Hospital, Dublin, Ireland
| | - John Faul
- Department of Respiratory Medicine, Connolly Hospital, Dublin, Ireland
| | - Joy Creaser-Thomas
- College of Medicine, Institute of Life Science, Swansea University, Swansea, UK
| | - Mark Andrews
- Respiratory Medicine, Hywel Dda University Health Board, Llanelli, UK
| | - Robin Ghosal
- Respiratory Medicine, Hywel Dda University Health Board, Llanelli, UK
| | - Stefan Piatek
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | - Theda U P Bartolomaeus
- Max Delbrück Center for Molecular Medicine (MDC), 13125, Berlin, Germany
- Experimental and Clinical Research Center, A Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Lindenberger Weg 80, 13125, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site, 10785, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117, Berlin, Germany
| | - Till Birkner
- Max Delbrück Center for Molecular Medicine (MDC), 13125, Berlin, Germany
- Experimental and Clinical Research Center, A Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Lindenberger Weg 80, 13125, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117, Berlin, Germany
| | - Sarah Dwyer
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Nitin Kumar
- Host-Microbiota Interactions Laboratory, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Elena M Turek
- National Heart and Lung Institute, Imperial College London, London, UK
| | - A William Musk
- School of Population and Global Health, The University of Western Australia, Perth, WA, Australia
- Busselton Population Medical Research Institute, Sir Charles Gairdner Hospital, Perth, WA, Australia
- Department of Respiratory Medicine Sir Charles Gairdner Hospital, Perth, WA, Australia
| | - Jennie Hui
- School of Population and Global Health, The University of Western Australia, Perth, WA, Australia
- Busselton Population Medical Research Institute, Sir Charles Gairdner Hospital, Perth, WA, Australia
| | - Michael Hunter
- School of Population and Global Health, The University of Western Australia, Perth, WA, Australia
- Busselton Population Medical Research Institute, Sir Charles Gairdner Hospital, Perth, WA, Australia
| | - Alan James
- School of Population and Global Health, The University of Western Australia, Perth, WA, Australia
- Department of Respiratory Medicine Sir Charles Gairdner Hospital, Perth, WA, Australia
- Department of Pulmonary Physiology and Sleep Medicine, Sir Charles Gairdner Hospital, Perth, WA, Australia
| | - Marc-Emmanuel Dumas
- National Heart and Lung Institute, Imperial College London, London, UK
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- U1283 INSERM / UMR8199 CNRS, Institut Pasteur de Lille, Lille University Hospital, European Genomic Institute for Diabetes, University of Lille, Lille, France
- McGill Genome Centre, McGill University, Montréal, QC, Canada
| | - Sarah Filippi
- Department of Mathematics, Imperial College London, London, UK
| | - Michael J Cox
- University of Birmingham College of Medical and Dental Sciences, 150183, Institute of Microbiology and Infection, Birmingham, UK
| | - Trevor D Lawley
- Host-Microbiota Interactions Laboratory, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Sofia K Forslund
- Max Delbrück Center for Molecular Medicine (MDC), 13125, Berlin, Germany.
- Experimental and Clinical Research Center, A Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Lindenberger Weg 80, 13125, Berlin, Germany.
- DZHK (German Centre for Cardiovascular Research), Partner Site, 10785, Berlin, Germany.
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117, Berlin, Germany.
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Structural and Computational Biology Unit, 69117, Heidelberg, Germany.
| | - Miriam F Moffatt
- National Heart and Lung Institute, Imperial College London, London, UK.
| | | |
Collapse
|
4
|
Karahashi Y, Cueno ME, Kamio N, Takahashi Y, Takeshita I, Soda K, Maruoka S, Gon Y, Sato S, Imai K. Fusobacterium nucleatum putatively affects the alveoli by disrupting the alveolar epithelial cell tight junction, enlarging the alveolar space, and increasing paracellular permeability. Biochem Biophys Res Commun 2023; 682:216-222. [PMID: 37826945 DOI: 10.1016/j.bbrc.2023.10.026] [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: 10/04/2023] [Accepted: 10/07/2023] [Indexed: 10/14/2023]
Abstract
Fusobacterium nucleatum (Fn) is abundant in the human oral cavity and has been associated with periodontal disease, which in-turn has been linked to respiratory disease development. Tight junctions (TJs) line the airway and alveoli surfaces serving as a first line of defense against multiple pathogens. Fn has already been linked to respiratory diseases, however, how Fn affects the alveolar TJ was not fully elucidated. Here, we designed and analyzed a TJ network, grew Fn cells and inoculated it in vitro (16HBE and primary cells) and in vivo (mice lung), measured transepithelial electrical resistance, performed RT-PCR, checked for in vitro cell and mice lung permeability, and determined air space size through morphometric measurements. We found that Fn can potentially affect TJs proteins that are directly exposed to the alveolar surface. Additionally, Fn could possibly cause neutrophil accumulation and an increase in alveolar space. Moreover, Fn putatively may cause an increase in paracellular permeability in the alveoli.
Collapse
Affiliation(s)
- Yukihiro Karahashi
- Department of Periodontology, Nihon University School of Dentistry, Tokyo, 101-8310, Japan; Department of Microbiology and Immunology, Nihon University School of Dentistry, Tokyo, 101-8310, Japan
| | - Marni E Cueno
- Department of Microbiology and Immunology, Nihon University School of Dentistry, Tokyo, 101-8310, Japan
| | - Noriaki Kamio
- Department of Microbiology and Immunology, Nihon University School of Dentistry, Tokyo, 101-8310, Japan
| | - Yuwa Takahashi
- Department of Microbiology and Immunology, Nihon University School of Dentistry, Tokyo, 101-8310, Japan
| | - Ikuko Takeshita
- Division of Respiratory Medicine, Department of Internal Medicine, Nihon University School of Medicine, Tokyo, 173-8610, Japan
| | - Kaori Soda
- Division of Respiratory Medicine, Department of Internal Medicine, Nihon University School of Medicine, Tokyo, 173-8610, Japan
| | - Shuichiro Maruoka
- Division of Respiratory Medicine, Department of Internal Medicine, Nihon University School of Medicine, Tokyo, 173-8610, Japan
| | - Yasuhiro Gon
- Division of Respiratory Medicine, Department of Internal Medicine, Nihon University School of Medicine, Tokyo, 173-8610, Japan
| | - Shuichi Sato
- Department of Periodontology, Nihon University School of Dentistry, Tokyo, 101-8310, Japan
| | - Kenichi Imai
- Department of Microbiology and Immunology, Nihon University School of Dentistry, Tokyo, 101-8310, Japan.
| |
Collapse
|
5
|
Hangjin L, Junting Y, Yiqin W, Hui Q, Shen Y, Jizhe W. Culture expansion of primary human nasal epithelial cells (NEC) isolated with a nasal scraping spoon. J Int Med Res 2023; 51:3000605231207759. [PMID: 37917806 PMCID: PMC10623993 DOI: 10.1177/03000605231207759] [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: 04/22/2023] [Accepted: 09/21/2023] [Indexed: 11/04/2023] Open
Abstract
OBJECTIVE To obtain high-purity nasal epithelial cells (NEC) while avoiding the irritation experienced by patients during nasal biopsies. METHODS This prospective, observational study enrolled patients undergoing surgical treatment for nasal septum deviation. After general anaesthesia, a novel nasal scraping spoon was used to collect epithelial cells from the mid-part of the inferior turbinate. The cells were evenly plated on six-well plates coated with rat tail collagen. The morphology and growth of the cells were observed at different time-points using an inverted phase-contrast microscope. Immunofluorescent staining of cytokeratin 18 was used to identify NEC. Ki67 staining was used to check cell viability. RESULTS This study collected samples from 19 patients during a short procedure. No postoperative complications were observed. Cell samples ranging from 8.31 × 105 to 2.04 × 106 cells/sample were obtained. The culture model was suitable for primary NEC culture as demonstrated by the faster proliferation (5-7 days). There was no fungal or bacterial contamination. Immunofluorescent staining confirmed the presence and proliferative activity of NEC in the cultures. CONCLUSION A novel nasal scraping spoon provided an easy sampling method, avoided nasal injuries and psychological barriers to sampling and sufficient viable NEC to establish primary cultures.
Collapse
Affiliation(s)
- Li Hangjin
- Department of Otolaryngology, Head and Neck Surgery, Second Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Yin Junting
- Department of Otolaryngology, Head and Neck Surgery, Second Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Wang Yiqin
- Department of Otolaryngology, Head and Neck Surgery, Second Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Qu Hui
- Department of Otolaryngology, Head and Neck Surgery, Second Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Yu Shen
- Dalian University of Technology, Dalian, Liaoning Province, China
| | - Wang Jizhe
- Department of Otolaryngology, Head and Neck Surgery, Second Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| |
Collapse
|
6
|
Otter CJ, Fausto A, Tan LH, Weiss SR, Cohen NA. Infection of Primary Nasal Epithelial Cells Grown at an Air-Liquid Interface to Characterize Human Coronavirus-Host Interactions. J Vis Exp 2023:10.3791/64868. [PMID: 37811957 PMCID: PMC10811614 DOI: 10.3791/64868] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2023] Open
Abstract
Three highly pathogenic human coronaviruses (HCoVs) - SARS-CoV (2002), MERS-CoV (2012), and SARS-CoV-2 (2019) - have emerged and caused significant public health crises in the past 20 years. Four additional HCoVs cause a significant portion of common cold cases each year (HCoV-NL63, -229E, -OC43, and -HKU1), highlighting the importance of studying these viruses in physiologically relevant systems. HCoVs enter the respiratory tract and establish infection in the nasal epithelium, the primary site encountered by all respiratory pathogens. We use a primary nasal epithelial culture system in which patient-derived nasal samples are grown at an air-liquid interface (ALI) to study host-pathogen interactions at this important sentinel site. These cultures recapitulate many features of the in vivo airway, including the cell types present, ciliary function, and mucus production. We describe methods to characterize viral replication, host cell tropism, virus-induced cytotoxicity, and innate immune induction in nasal ALI cultures following HCoV infection, using recent work comparing lethal and seasonal HCoVs as an example1. An increased understanding of host-pathogen interactions in the nose has the potential to provide novel targets for antiviral therapeutics against HCoVs and other respiratory viruses that will likely emerge in the future.
Collapse
Affiliation(s)
- Clayton J Otter
- Department of Microbiology, University of Pennsylvania; Penn Center for Research on Coronaviruses and Other Emerging Pathogens, Perelman School of Medicine, University of Pennsylvania
| | - Alejandra Fausto
- Department of Microbiology, University of Pennsylvania; Penn Center for Research on Coronaviruses and Other Emerging Pathogens, Perelman School of Medicine, University of Pennsylvania
| | - Li Hui Tan
- Otorhinolaryngology-Head and Neck Surgery, University of Pennsylvania; Corporal Michael J. Crescenz VA Medical Center
| | - Susan R Weiss
- Department of Microbiology, University of Pennsylvania; Penn Center for Research on Coronaviruses and Other Emerging Pathogens, Perelman School of Medicine, University of Pennsylvania
| | - Noam A Cohen
- Otorhinolaryngology-Head and Neck Surgery, University of Pennsylvania; Corporal Michael J. Crescenz VA Medical Center;
| |
Collapse
|
7
|
Escher A, Kieninger E, Groof SD, Savas ST, Schneiter M, Tschanz SA, Frenz M, Latzin P, Casaulta C, Müller L. In Vitro Effect of Combined Hypertonic Saline and Salbutamol on Ciliary Beating Frequency and Mucociliary Transport in Human Nasal Epithelial Cells of Healthy Volunteers and Patients with Cystic Fibrosis. J Aerosol Med Pulm Drug Deliv 2023; 36:171-180. [PMID: 37196208 DOI: 10.1089/jamp.2022.0026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2023] Open
Abstract
Background: Inhalation of hypertonic saline (HS) is standard of care in patients with cystic fibrosis (CF). However, it is unclear if adding salbutamol has-besides bronchodilation-further benefits, for example, on the mucociliary clearance. We assessed this in vitro by measuring the ciliary beating frequency (CBF) and the mucociliary transport rate (MCT) in nasal epithelial cells (NECs) of healthy volunteers and patients with CF. Aims: To investigate the effect of HS, salbutamol, and its combination on (muco)ciliary activity of NECs in vitro, and to assess potential differences between healthy controls and patients with CF. Methods: NECs obtained from 10 healthy volunteers and 5 patients with CF were differentiated at the air-liquid interface and aerosolized with 0.9% isotonic saline ([IS] control), 6% HS, 0.06% salbutamol, or combined HS and salbutamol. CBF and MCT were monitored over 48-72 hours. Results: In NECs of healthy controls, the absolute CBF increase was comparable for all substances, but CBF dynamics were different: HS increased CBF slowly and its effect lasted for an extended period, salbutamol and IS increased CBF rapidly and the effect subsided similarly fast, and HS and salbutamol resulted in a rapid and long-lasting CBF increase. Results for CF cells were comparable, but less pronounced. Similar to CBF, MCT increased after the application of all the tested substances. Conclusion: CBF and MCT of NECs of healthy participants and CBF of patients with CF increased upon treatment with aerosolized IS, HS, salbutamol, or HS and salbutamol, showing a relevant effect for all tested substances. The difference in the CBF dynamics can be explained by the fact that the properties of the mucus are changed differently by different saline concentrations.
Collapse
Affiliation(s)
- Anaïs Escher
- Division of Paediatric Respiratory Medicine and Allergology, Department of Paediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Elisabeth Kieninger
- Division of Paediatric Respiratory Medicine and Allergology, Department of Paediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Susan De Groof
- Division of Paediatric Respiratory Medicine and Allergology, Department of Paediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Sibel T Savas
- Division of Paediatric Respiratory Medicine and Allergology, Department of Paediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Martin Schneiter
- Institute of Applied Physics, University of Bern, Bern, Switzerland
- Institute of Anatomy, University of Bern, Bern, Switzerland
| | | | - Martin Frenz
- Institute of Applied Physics, University of Bern, Bern, Switzerland
| | - Philipp Latzin
- Division of Paediatric Respiratory Medicine and Allergology, Department of Paediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Carmen Casaulta
- Division of Paediatric Respiratory Medicine and Allergology, Department of Paediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Loretta Müller
- Division of Paediatric Respiratory Medicine and Allergology, Department of Paediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| |
Collapse
|
8
|
Otter C, Fausto A, Tan L, Khosla A, Cohen N, Weiss S. Infection of primary nasal epithelial cells differentiates among lethal and seasonal human coronaviruses. Proc Natl Acad Sci U S A 2023; 120:e2218083120. [PMID: 37023127 PMCID: PMC10104492 DOI: 10.1073/pnas.2218083120] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 02/27/2023] [Indexed: 04/07/2023] Open
Abstract
The nasal epithelium is the initial entry portal and primary barrier to infection by all human coronaviruses (HCoVs). We utilize primary human nasal epithelial cells grown at air-liquid interface, which recapitulate the heterogeneous cellular population as well as mucociliary clearance functions of the in vivo nasal epithelium, to compare lethal [Severe acute respiratory syndrome (SARS)-CoV-2 and Middle East respiratory syndrome-CoV (MERS-CoV)] and seasonal (HCoV-NL63 and HCoV-229E) HCoVs. All four HCoVs replicate productively in nasal cultures, though replication is differentially modulated by temperature. Infections conducted at 33 °C vs. 37 °C (reflective of temperatures in the upper and lower airway, respectively) revealed that replication of both seasonal HCoVs (HCoV-NL63 and -229E) is significantly attenuated at 37 °C. In contrast, SARS-CoV-2 and MERS-CoV replicate at both temperatures, though SARS-CoV-2 replication is enhanced at 33 °C late in infection. These HCoVs also diverge significantly in terms of cytotoxicity induced following infection, as the seasonal HCoVs as well as SARS-CoV-2 cause cellular cytotoxicity as well as epithelial barrier disruption, while MERS-CoV does not. Treatment of nasal cultures with type 2 cytokine IL-13 to mimic asthmatic airways differentially impacts HCoV receptor availability as well as replication. MERS-CoV receptor DPP4 expression increases with IL-13 treatment, whereas ACE2, the receptor used by SARS-CoV-2 and HCoV-NL63, is down-regulated. IL-13 treatment enhances MERS-CoV and HCoV-229E replication but reduces that of SARS-CoV-2 and HCoV-NL63, reflecting the impact of IL-13 on HCoV receptor availability. This study highlights diversity among HCoVs during infection of the nasal epithelium, which is likely to influence downstream infection outcomes such as disease severity and transmissibility.
Collapse
Affiliation(s)
- Clayton J. Otter
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
- Penn Center for Research on Coronaviruses and Other Emerging Pathogens, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Alejandra Fausto
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
- Penn Center for Research on Coronaviruses and Other Emerging Pathogens, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Li Hui Tan
- Otorhinolaryngology-Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
- Department of Surgery, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA19104
| | - Alisha S. Khosla
- Otorhinolaryngology-Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
- Department of Surgery, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA19104
| | - Noam A. Cohen
- Otorhinolaryngology-Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
- Department of Surgery, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA19104
| | - Susan R. Weiss
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
- Penn Center for Research on Coronaviruses and Other Emerging Pathogens, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| |
Collapse
|
9
|
Silva S, Bicker J, Falcão A, Fortuna A. Air-liquid interface (ALI) impact on different respiratory cell cultures. Eur J Pharm Biopharm 2023; 184:62-82. [PMID: 36696943 DOI: 10.1016/j.ejpb.2023.01.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 09/24/2022] [Accepted: 01/19/2023] [Indexed: 01/23/2023]
Abstract
The intranasal route has been receiving greater attention from the scientific community not only for systemic drug delivery but also for the treatment of pulmonary and neurological diseases. Along with it, drug transport and permeability studies across the nasal mucosa have exponentially increased. Nevertheless, the translation of data from in vitro cell lines to in vivo studies is not always reliable, due to the difficulty in generating an in vitro model that resembles respiratory human physiology. Among all currently available methodologies, the air-liquid interface (ALI) method is advantageous to promote cell differentiation and optimize the morphological and histological characteristics of airway epithelium cells. Cells grown under ALI conditions, in alternative to submerged conditions, appear to provide relevant input for inhalation and pulmonary toxicology and complement in vivo experiments. Different methodologies and a variety of materials have been used to induce ALI conditions in primary cells and numerous cell lines. Until this day, with only exploratory results, no consensus has been reached regarding the validation of the ALI method, hampering data comparison. The present review describes the most adequate cell models of airway epithelium and how these models are differently affected by ALI conditions. It includes the evaluation of cellular features before and after ALI, and the application of the method in primary cell cultures, commercial 3D primary cells, cell lines and stem-cell derived models. A variety of these models have been recently applied for pharmacological studies against severe acute respiratory syndrome-coronavirus(-2) SARS-CoV(-2), namely primary cultures with alveolar type II epithelium cells and organotypic 3D models. The herein compiled data suggest that ALI conditions must be optimized bearing in mind the type of cells (nasal, bronchial, alveolar), their origin and the objective of the study.
Collapse
Affiliation(s)
- Soraia Silva
- Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal; CIBIT - Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal
| | - Joana Bicker
- Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal; CIBIT - Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal
| | - Amílcar Falcão
- Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal; CIBIT - Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal
| | - Ana Fortuna
- Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal; CIBIT - Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
10
|
Rebuli ME, Stanley Lee A, Nurhussien L, Tahir UA, Sun WY, Kimple AJ, Ebert CS, Almond M, Jaspers I, Rice MB. Nasal biomarkers of immune function differ based on smoking and respiratory disease status. Physiol Rep 2023; 11:e15528. [PMID: 36780897 PMCID: PMC9925276 DOI: 10.14814/phy2.15528] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 11/04/2022] [Accepted: 11/13/2022] [Indexed: 02/15/2023] Open
Abstract
Respiratory biomarkers have the potential to identify airway injury by revealing inflammatory processes within the respiratory tract. Currently, there are no respiratory biomarkers suitable for clinical use to identify patients that warrant further diagnostic work-up, counseling, and treatment for toxic inhalant exposures or chronic airway disease. Using a novel, noninvasive method of sampling the nasal epithelial lining fluid, we aimed to investigate if nasal biomarker patterns could distinguish healthy nonsmoking adults from active smokers and those with chronic upper and lower airway disease in this exploratory study. We compared 28 immune mediators from healthy nonsmoking adults (n = 32), former smokers with COPD (n = 22), chronic rhinosinusitis (CRS) (n = 22), and smoking adults without airway disease (n = 13). Using ANOVA, multinomial logistic regressions, and weighted gene co-expression network analysis (WGCNA), we determined associations between immune mediators and each cohort. Six mediators (IL-7, IL-10, IL-13, IL-12p70, IL-15, and MCP-1) were lower among disease groups compared to healthy controls. Participants with lower levels of IL-10, IL-12p70, IL-13, and MCP-1 in the nasal fluid had a higher odds of being in the COPD or CRS group. The cluster analysis identified groups of mediators that correlated with disease status. Specifically, the cluster of IL-10, IL-12p70, and IL-13, was positively correlated with healthy and negatively correlated with COPD groups, and two clusters were correlated with active smoking. In this exploratory study, we preliminarily identified groups of nasal mucosal mediators that differed by airway disease and smoking status. Future prospective, age-matched studies that control for medication use are needed to validate these patterns and determine if nasosorption has diagnostic utility for upper and lower airway disease or injury.
Collapse
Affiliation(s)
- Meghan E. Rebuli
- Department of Pediatrics and Curriculum in Toxicology and Environmental MedicineUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
- Center for Environmental Medicine, Asthma and Lung BiologyUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Anna Stanley Lee
- Department of Medicine, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - Lina Nurhussien
- Department of Medicine, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - Usman A. Tahir
- Department of Medicine, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - Wendy Y. Sun
- Department of Medicine, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - Adam J. Kimple
- Department of Otolaryngology‐Head and Neck SurgeryUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Charles S. Ebert
- Department of Otolaryngology‐Head and Neck SurgeryUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Martha Almond
- Center for Environmental Medicine, Asthma and Lung BiologyUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Ilona Jaspers
- Department of Pediatrics and Curriculum in Toxicology and Environmental MedicineUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
- Center for Environmental Medicine, Asthma and Lung BiologyUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Mary B. Rice
- Department of Medicine, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| |
Collapse
|
11
|
Delhove J, Alawami M, Macowan M, Lester SE, Nguyen PT, Jersmann HPA, Reynolds PN, Roscioli E. Organotypic sinonasal airway culture systems are predictive of the mucociliary phenotype produced by bronchial airway epithelial cells. Sci Rep 2022; 12:19225. [PMID: 36357550 PMCID: PMC9648462 DOI: 10.1038/s41598-022-23667-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 11/03/2022] [Indexed: 11/12/2022] Open
Abstract
Differentiated air-liquid interface models are the current standard to assess the mucociliary phenotype using clinically-derived samples in a controlled environment. However, obtaining basal progenitor airway epithelial cells (AEC) from the lungs is invasive and resource-intensive. Hence, we applied a tissue engineering approach to generate organotypic sinonasal AEC (nAEC) epithelia to determine whether they are predictive of bronchial AEC (bAEC) models. Basal progenitor AEC were isolated from healthy participants using a cytological brushing method and differentiated into epithelia on transwells until the mucociliary phenotype was observed. Tissue architecture was assessed using H&E and alcian blue/Verhoeff-Van Gieson staining, immunofluorescence (for cilia via acetylated α-tubulin labelling) and scanning electron microscopy. Differentiation and the formation of tight-junctions were monitored over the culture period (day 1-32) by quantifying trans-epithelial electrical resistance. End point (day 32) tight junction protein expression was assessed using Western blot analysis of ZO-1, Occludin-1 and Claudin-1. Reverse transcription qPCR-array was used to assess immunomodulatory and autophagy-specific transcript profiles. All outcome measures were assessed using R-statistical software. Mucociliary architecture was comparable for nAEC and bAEC-derived cultures, e.g. cell density P = 0.55, epithelial height P = 0.88 and cilia abundance P = 0.41. Trans-epithelial electrical resistance measures were distinct from day 1-14, converged over days 16-32, and were statistically similar over the entire culture period (global P < 0.001). This agreed with end-point (day 32) measures of tight junction protein abundance which were non-significant for each analyte (P > 0.05). Transcript analysis for inflammatory markers demonstrated significant variation between nAEC and bAEC epithelial cultures, and favoured increased abundance in the nAEC model (e.g. TGFβ and IL-1β; P < 0.05). Conversely, the abundance of autophagy-related transcripts were comparable and the range of outcome measures for either model exhibited a considerably more confined uncertainty distribution than those observed for the inflammatory markers. Organotypic air-liquid interface models of nAEC are predictive of outcomes related to barrier function, mucociliary architecture and autophagy gene activity in corresponding bAEC models. However, inflammatory markers exhibited wide variation which may be explained by the sentinel immunological surveillance role of the sinonasal epithelium.
Collapse
Affiliation(s)
- Juliette Delhove
- grid.1010.00000 0004 1936 7304Adelaide Medical School, University of Adelaide, Adelaide, SA Australia ,grid.1694.aRespiratory and Sleep Medicine, Women’s and Children’s Hospital, Adelaide, SA Australia
| | - Moayed Alawami
- grid.1010.00000 0004 1936 7304Adelaide Medical School, University of Adelaide, Adelaide, SA Australia ,grid.460761.20000 0001 0323 4206Respiratory Department, Lyell McEwin Hospital, Adelaide, SA Australia
| | - Matthew Macowan
- grid.1010.00000 0004 1936 7304Adelaide Medical School, University of Adelaide, Adelaide, SA Australia ,grid.1002.30000 0004 1936 7857Department of Immunology and Pathology, Monash University, Melbourne, VIC Australia
| | - Susan E. Lester
- grid.1010.00000 0004 1936 7304Adelaide Medical School, University of Adelaide, Adelaide, SA Australia ,grid.278859.90000 0004 0486 659XDepartment of Rheumatology, The Queen Elizabeth Hospital, Adelaide, SA Australia
| | - Phan T. Nguyen
- grid.1010.00000 0004 1936 7304Adelaide Medical School, University of Adelaide, Adelaide, SA Australia ,grid.416075.10000 0004 0367 1221Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, SA Australia
| | - Hubertus P. A. Jersmann
- grid.1010.00000 0004 1936 7304Adelaide Medical School, University of Adelaide, Adelaide, SA Australia ,grid.416075.10000 0004 0367 1221Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, SA Australia
| | - Paul N. Reynolds
- grid.1010.00000 0004 1936 7304Adelaide Medical School, University of Adelaide, Adelaide, SA Australia ,grid.416075.10000 0004 0367 1221Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, SA Australia
| | - Eugene Roscioli
- grid.1010.00000 0004 1936 7304Adelaide Medical School, University of Adelaide, Adelaide, SA Australia ,grid.416075.10000 0004 0367 1221Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, SA Australia ,Adelaide Health and Medical Science, Building, Corner of North Terrace and George St, Adelaide, SA 5005 Australia
| |
Collapse
|
12
|
Otter CJ, Fausto A, Tan LH, Cohen NA, Weiss SR. Infection of primary nasal epithelial cells differentiates among lethal and seasonal human coronaviruses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.10.17.512617. [PMID: 36299422 PMCID: PMC9603826 DOI: 10.1101/2022.10.17.512617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The nasal epithelium is the initial entry portal and primary barrier to infection by all human coronaviruses (HCoVs). We utilize primary nasal epithelial cells grown at air-liquid interface, which recapitulate the heterogeneous cellular population as well as mucociliary clearance functions of the in vivo nasal epithelium, to compare lethal (SARS-CoV-2 and MERS-CoV) and seasonal (HCoV-NL63 and HCoV-229E) HCoVs. All four HCoVs replicate productively in nasal cultures but diverge significantly in terms of cytotoxicity induced following infection, as the seasonal HCoVs as well as SARS-CoV-2 cause cellular cytotoxicity as well as epithelial barrier disruption, while MERS-CoV does not. Treatment of nasal cultures with type 2 cytokine IL-13 to mimic asthmatic airways differentially impacts HCoV replication, enhancing MERS-CoV replication but reducing that of SARS-CoV-2 and HCoV-NL63. This study highlights diversity among HCoVs during infection of the nasal epithelium, which is likely to influence downstream infection outcomes such as disease severity and transmissibility.
Collapse
Affiliation(s)
- Clayton J. Otter
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn Center for Research on Coronaviruses and Other Emerging Pathogens, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alejandra Fausto
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn Center for Research on Coronaviruses and Other Emerging Pathogens, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Li Hui Tan
- Department of Otorhinolaryngology-Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
| | - Noam A. Cohen
- Department of Otorhinolaryngology-Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
| | - Susan R. Weiss
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn Center for Research on Coronaviruses and Other Emerging Pathogens, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
13
|
Moreau M, Fisher J, Andersen ME, Barnwell A, Corzine S, Ranade A, McMullen PD, Slattery SD. NAM-based Prediction of Point-of-contact Toxicity in the Lung: A Case Example With 1,3-dichloropropene. Toxicology 2022; 481:153340. [PMID: 36183849 DOI: 10.1016/j.tox.2022.153340] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 07/13/2022] [Accepted: 09/27/2022] [Indexed: 11/27/2022]
Abstract
Time, cost, ethical, and regulatory considerations surrounding in vivo testing methods render them insufficient to meet existing and future chemical safety testing demands. There is a need for the development of in vitro and in silico alternatives to replace traditional in vivo methods for inhalation toxicity assessment. Exposures of differentiated airway epithelial cultures to gases or aerosols at the air-liquid interface (ALI) can assess tissue responses and in vitro to in vivo extrapolation can align in vitro exposure levels with in-life exposures expected to give similar tissue exposures. Because the airway epithelium varies along its length, with various regions composed of different cell types, we have introduced a known toxic vapor to five human-derived, differentiated, in vitro airway epithelial cell culture models-MucilAir of nasal, tracheal, or bronchial origin, SmallAir, and EpiAlveolar-representing five regions of the airway epithelium-nasal, tracheal, bronchial, bronchiolar, and alveolar. We have monitored toxicity in these cultures 24hours after acute exposure using an assay for transepithelial conductance (for epithelial barrier integrity) and the lactate dehydrogenase (LDH) release assay (for cytotoxicity). Our vapor of choice in these experiments was 1,3-dichloropropene (1,3-DCP). Finally, we have developed an airway dosimetry model for 1,3-DCP vapor to predict in vivo external exposure scenarios that would produce toxic local tissue concentrations as determined by in vitro experiments. Measured in vitro points of departure (PoDs) for all tested cell culture models were similar. Calculated rat equivalent inhaled concentrations varied by model according to position of the modeled tissue within the airway, with nasal respiratory tissue being the most proximal and most sensitive tissue, and alveolar epithelium being the most distal and least sensitive tissue. These predictions are qualitatively in accordance with empirically determined in vivo PoDs. The predicted PoD concentrations were close to, but slightly higher than, PoDs determined by in vivo subchronic studies.
Collapse
Affiliation(s)
- Marjory Moreau
- ScitoVation, LLC, 6 Davis Drive, Suite 146, Durham, North Carolina, 27709, USA
| | - Jeff Fisher
- ScitoVation, LLC, 6 Davis Drive, Suite 146, Durham, North Carolina, 27709, USA
| | - Melvin E Andersen
- ScitoVation, LLC, 6 Davis Drive, Suite 146, Durham, North Carolina, 27709, USA
| | - Asayah Barnwell
- ScitoVation, LLC, 6 Davis Drive, Suite 146, Durham, North Carolina, 27709, USA
| | - Sage Corzine
- ScitoVation, LLC, 6 Davis Drive, Suite 146, Durham, North Carolina, 27709, USA
| | - Aarati Ranade
- ScitoVation, LLC, 6 Davis Drive, Suite 146, Durham, North Carolina, 27709, USA
| | - Patrick D McMullen
- ScitoVation, LLC, 6 Davis Drive, Suite 146, Durham, North Carolina, 27709, USA
| | - Scott D Slattery
- ScitoVation, LLC, 6 Davis Drive, Suite 146, Durham, North Carolina, 27709, USA.
| |
Collapse
|
14
|
Suzuki M, Cooksley C, Suzuki T, Ramezanpour M, Nakazono A, Nakamaru Y, Homma A, Vreugde S. TLR Signals in Epithelial Cells in the Nasal Cavity and Paranasal Sinuses. FRONTIERS IN ALLERGY 2022; 2:780425. [PMID: 35387020 PMCID: PMC8974762 DOI: 10.3389/falgy.2021.780425] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 11/01/2021] [Indexed: 12/24/2022] Open
Abstract
The respiratory tract is constantly at risk of invasion by microorganisms such as bacteria, viruses, and fungi. In particular, the mucosal epithelium of the nasal cavity and paranasal sinuses is at the very forefront of the battles between the host and the invading pathogens. Recent studies have revealed that the epithelium not only constitutes a physical barrier but also takes an essential role in the activation of the immune system. One of the mechanisms equipped in the epithelium to fight against microorganisms is the Toll-like receptor (TLR) response. TLRs recognize common structural components of microorganisms and activate the innate immune system, resulting in the production of a plethora of cytokines and chemokines in the response against microbes. As the epithelia-derived cytokines are deeply involved in the pathogenesis of inflammatory conditions in the nasal cavity and paranasal sinuses, such as chronic rhinosinusitis (CRS) and allergic rhinitis (AR), the molecules involved in the TLR response may be utilized as therapeutic targets for these diseases. There are several differences in the TLR response between nasal and bronchial epithelial cells, and knowledge of the TLR signals in the upper airway is sparse compared to that in the lower airway. In this review, we provide recent evidence on TLR signaling in the upper airway, focusing on the expression, regulation, and responsiveness of TLRs in human nasal epithelial cells (HNECs). We also discuss how TLRs in the epithelium are involved in the pathogenesis of, and possible therapeutic targeting, for CRS and AR.
Collapse
Affiliation(s)
- Masanobu Suzuki
- Department of Otolaryngology-Head and Neck Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Clare Cooksley
- Department of Surgery-Otorhinolaryngology Head and Neck Surgery, Central Adelaide Local Health Network and the University of Adelaide, Adelaide, SA, Australia
| | - Takayoshi Suzuki
- Department of Otolaryngology-Head and Neck Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Mahnaz Ramezanpour
- Department of Surgery-Otorhinolaryngology Head and Neck Surgery, Central Adelaide Local Health Network and the University of Adelaide, Adelaide, SA, Australia
| | - Akira Nakazono
- Department of Otolaryngology-Head and Neck Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Yuji Nakamaru
- Department of Otolaryngology-Head and Neck Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Akihiro Homma
- Department of Otolaryngology-Head and Neck Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Sarah Vreugde
- Department of Surgery-Otorhinolaryngology Head and Neck Surgery, Central Adelaide Local Health Network and the University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
15
|
Sivarajan R, Kessie DK, Oberwinkler H, Pallmann N, Walles T, Scherzad A, Hackenberg S, Steinke M. Susceptibility of Human Airway Tissue Models Derived From Different Anatomical Sites to Bordetella pertussis and Its Virulence Factor Adenylate Cyclase Toxin. Front Cell Infect Microbiol 2021; 11:797491. [PMID: 35059325 PMCID: PMC8765404 DOI: 10.3389/fcimb.2021.797491] [Citation(s) in RCA: 3] [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: 10/18/2021] [Accepted: 12/06/2021] [Indexed: 11/13/2022] Open
Abstract
To study the interaction of human pathogens with their host target structures, human tissue models based on primary cells are considered suitable. Complex tissue models of the human airways have been used as infection models for various viral and bacterial pathogens. The Gram-negative bacterium Bordetella pertussis is of relevant clinical interest since whooping cough has developed into a resurgent infectious disease. In the present study, we created three-dimensional tissue models of the human ciliated nasal and tracheo-bronchial mucosa. We compared the innate immune response of these models towards the B. pertussis virulence factor adenylate cyclase toxin (CyaA) and its enzymatically inactive but fully pore-forming toxoid CyaA-AC-. Applying molecular biological, histological, and microbiological assays, we found that 1 µg/ml CyaA elevated the intracellular cAMP level but did not disturb the epithelial barrier integrity of nasal and tracheo-bronchial airway mucosa tissue models. Interestingly, CyaA significantly increased interleukin 6, interleukin 8, and human beta defensin 2 secretion in nasal tissue models, whereas tracheo-bronchial tissue models were not significantly affected compared to the controls. Subsequently, we investigated the interaction of B. pertussis with both differentiated primary nasal and tracheo-bronchial tissue models and demonstrated bacterial adherence and invasion without observing host cell type-specific significant differences. Even though the nasal and the tracheo-bronchial mucosa appear similar from a histological perspective, they are differentially susceptible to B. pertussis CyaA in vitro. Our finding that nasal tissue models showed an increased innate immune response towards the B. pertussis virulence factor CyaA compared to tracheo-bronchial tissue models may reflect the key role of the nasal airway mucosa as the first line of defense against airborne pathogens.
Collapse
Affiliation(s)
- Rinu Sivarajan
- Chair of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Würzburg, Germany
| | | | - Heike Oberwinkler
- Chair of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Niklas Pallmann
- Chair of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Thorsten Walles
- Department of Thoracic Surgery, University Medicine Magdeburg, Magdeburg, Germany
| | - Agmal Scherzad
- Department of Oto-Rhino-Laryngology, Plastic, Aesthetic and Reconstructive Head and Neck Surgery, University Hospital Würzburg, Würzburg, Germany
| | - Stephan Hackenberg
- Department of Oto-Rhino-Laryngology – Head and Neck Surgery, Rheinisch-Westfälische Technische Hochschule Aachen (RWTH) Aachen University Hospital, Aachen, Germany
| | - Maria Steinke
- Chair of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Würzburg, Germany
- Translational Center Regenerative Therapies, Fraunhofer Institute for Silicate Research ISC, Würzburg, Germany
- *Correspondence: Maria Steinke,
| |
Collapse
|
16
|
Rebuli ME, Brocke SA, Jaspers I. Impact of inhaled pollutants on response to viral infection in controlled exposures. J Allergy Clin Immunol 2021; 148:1420-1429. [PMID: 34252446 PMCID: PMC8569906 DOI: 10.1016/j.jaci.2021.07.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 07/01/2021] [Accepted: 07/06/2021] [Indexed: 12/09/2022]
Abstract
Air pollutants are a major source of increased risk of disease, hospitalization, morbidity, and mortality worldwide. The respiratory tract is a primary target of potential concurrent exposure to both inhaled pollutants and pathogens, including viruses. Although there are various associative studies linking adverse outcomes to co- or subsequent exposures to inhaled pollutants and viruses, knowledge about causal linkages and mechanisms by which pollutant exposure may alter human respiratory responses to viral infection is more limited. In this article, we review what is known about the impact of pollutant exposure on antiviral host defense responses and describe potential mechanisms by which pollutants can alter the viral infection cycle. This review focuses on evidence from human observational and controlled exposure, ex vivo, and in vitro studies. Overall, there are a myriad of points throughout the viral infection cycle that inhaled pollutants can alter to modulate appropriate host defense responses. These alterations may contribute to observed increases in rates of viral infection and associated morbidity and mortality in areas of the world with high ambient pollution levels or in people using tobacco products. Although the understanding of mechanisms of interaction is advancing through controlled in vivo and in vitro exposure models, more studies are needed because emerging infectious pathogens, such as severe acute respiratory syndrome coronavirus 2, present a significant threat to public health.
Collapse
Affiliation(s)
- Meghan E Rebuli
- Curriculum in Toxicology & Environmental Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC; Center for Environmental Medicine, Asthma and Lung Biology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC; Department of Pediatrics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Stephanie A Brocke
- Curriculum in Toxicology & Environmental Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Ilona Jaspers
- Curriculum in Toxicology & Environmental Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC; Center for Environmental Medicine, Asthma and Lung Biology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC; Department of Pediatrics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC.
| |
Collapse
|
17
|
Mastalerz M, Dick E, Chakraborty AA, Hennen E, Schamberger AC, Schröppel A, Lindner M, Hatz R, Behr J, Hilgendorff A, Schmid O, Staab-Weijnitz CA. Validation of in vitro models for smoke exposure of primary human bronchial epithelial cells. Am J Physiol Lung Cell Mol Physiol 2021; 322:L129-L148. [PMID: 34668416 DOI: 10.1152/ajplung.00091.2021] [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] [Indexed: 11/22/2022] Open
Abstract
RATIONALE The bronchial epithelium is constantly challenged by inhalative insults including cigarette smoke (CS), a key risk factor for lung disease. In vitro exposure of bronchial epithelial cells using CS extract (CSE) is a widespread alternative to whole CS (wCS) exposure. However, CSE exposure protocols vary considerably between studies, precluding direct comparison of applied doses. Moreover, they are rarely validated in terms of physiological response in vivo and the relevance of the findings is often unclear. METHODS We tested six different exposure settings in primary human bronchial epithelial cells (phBECs), including five CSE protocols in comparison with wCS exposure. We quantified cell-delivered dose and directly compared all exposures using expression analysis of 10 well-established smoke-induced genes in bronchial epithelial cells. CSE exposure of phBECs was varied in terms of differentiation state, exposure route, duration of exposure, and dose. Gene expression was assessed by quantitative Real-Time PCR (qPCR) and Western Blot analysis. Cell type-specific expression of smoke-induced genes was analyzed by immunofluorescent analysis. RESULTS Three surprisingly dissimilar exposure types, namely chronic CSE treatment of differentiating phBECs, acute CSE treatment of submerged basal phBECs, and wCS exposure of differentiated phBECs performed best, resulting in significant upregulation of seven (chronic CSE) and six (acute wCS, acute submerged CSE exposure) out of 10 genes. Acute apical or basolateral exposure of differentiated phBECs with CSE was much less effective despite similar doses used. CONCLUSIONS Our findings provide guidance for the design of human in vitro CS exposure models in experimental and translational lung research.
Collapse
Affiliation(s)
- Michal Mastalerz
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Elisabeth Dick
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Ashesh Anjankumar Chakraborty
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Elisabeth Hennen
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Andrea C Schamberger
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Andreas Schröppel
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | | | - Rudolf Hatz
- Thoraxchirurgisches Zentrum, Klinik für Allgemeine, Viszeral-, Transplantations-, Gefäß- und Thoraxchirurgie, Klinikum Großhadern, Ludwig-Maximilians-Universität (LMU), Munich, Germany
| | - Jürgen Behr
- Medizinische Klinik und Poliklinik V, Klinikum der Ludwig-Maximilians-Universität (LMU), Munich, Germany, Member of the German Center for Lung Research (DZL)
| | - Anne Hilgendorff
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Otmar Schmid
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Claudia A Staab-Weijnitz
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| |
Collapse
|
18
|
Turek EM, Cox MJ, Hunter M, Hui J, James P, Willis-Owen SAG, Cuthbertson L, James A, Musk AW, Moffatt MF, Cookson WOCM. Airway microbial communities, smoking and asthma in a general population sample. EBioMedicine 2021; 71:103538. [PMID: 34425308 PMCID: PMC8387768 DOI: 10.1016/j.ebiom.2021.103538] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 07/27/2021] [Accepted: 07/29/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Normal airway microbial communities play a central role in respiratory health but are poorly characterized. Cigarette smoking is the dominant global environmental influence on lung function, and asthma has become the most prevalent chronic respiratory disease worldwide. Both conditions have major microbial components that are incompletely defined. METHODS We investigated airway bacterial communities in a general population sample of 529 Australian adults. Posterior oropharyngeal swabs were analyzed by sequencing of the 16S rRNA gene. The microbiota were characterized according to their prevalence, abundance and network memberships. FINDINGS The microbiota were similar across the general population, and were strongly organized into co-abundance networks. Smoking was associated with diversity loss, negative effects on abundant taxa, profound alterations to network structure and expansion of Streptococcus spp. By contrast, the asthmatic microbiota were selectively affected by an increase in Neisseria spp. and by reduced numbers of low abundance but prevalent organisms. INTERPRETATION Our study shows that the healthy airway microbiota in this population were contained within a highly structured ecosystem, suggesting balanced relationships between the microbiome and human host factors. The marked abnormalities in smokers may contribute to chronic obstructive pulmonary disease (COPD) and lung cancer. The narrow spectrum of abnormalities in asthmatics encourages investigation of damaging and protective effects of specific bacteria. FUNDING The study was funded by the Asmarley Trust and a Wellcome Joint Senior Investigator Award to WOCC and MFM (WT096964MA and WT097117MA). The Busselton Healthy Ageing Study is supported by the Government of Western Australia (Office of Science, Department of Health) the City of Busselton, and private donations.
Collapse
Affiliation(s)
- Elena M Turek
- National Heart and Lung Institute, Centre for Genomic Medicine, Imperial College London SW3 6LY, United Kingdom
| | - Michael J Cox
- National Heart and Lung Institute, Centre for Genomic Medicine, Imperial College London SW3 6LY, United Kingdom
| | - Michael Hunter
- School of Population and Global Health, University of Western Australia, Australia; Busselton Population Medical Research Institute, Western Australia, Australia
| | - Jennie Hui
- School of Population and Global Health, University of Western Australia, Australia; Busselton Population Medical Research Institute, Western Australia, Australia; PathWest Laboratory Medicine, Queen Elizabeth II Medical Centre, Western Australia, Australia
| | - Phillip James
- National Heart and Lung Institute, Centre for Genomic Medicine, Imperial College London SW3 6LY, United Kingdom
| | - Saffron A G Willis-Owen
- National Heart and Lung Institute, Centre for Genomic Medicine, Imperial College London SW3 6LY, United Kingdom
| | - Leah Cuthbertson
- National Heart and Lung Institute, Centre for Genomic Medicine, Imperial College London SW3 6LY, United Kingdom
| | - Alan James
- Busselton Population Medical Research Institute, Western Australia, Australia; Department of Pulmonary Physiology, Sir Charles Gairdner Hospital, UWA Medical School, University of Western Australia, Australia
| | - A William Musk
- School of Population and Global Health, University of Western Australia, Australia; Busselton Population Medical Research Institute, Western Australia, Australia; Department of Respiratory Medicine Sir Charles Gairdner Hospital, UWA Medical School, University of Western Australia, Australia
| | - Miriam F Moffatt
- National Heart and Lung Institute, Centre for Genomic Medicine, Imperial College London SW3 6LY, United Kingdom.
| | - William O C M Cookson
- National Heart and Lung Institute, Centre for Genomic Medicine, Imperial College London SW3 6LY, United Kingdom.
| |
Collapse
|
19
|
Cho HJ, Ha JG, Lee SN, Kim CH, Wang DY, Yoon JH. Differences and similarities between the upper and lower airway: focusing on innate immunity. Rhinology 2021; 59:441-450. [PMID: 34339483 DOI: 10.4193/rhin21.046] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The nose is the first respiratory barrier to external pathogens, allergens, pollutants, or cigarette smoke, and vigorous immune responses are triggered when external pathogens come in contact with the nasal epithelium. The mucosal epithelial cells of the nose are essential to the innate immune response against external pathogens and transmit signals that modulate the adaptive immune response. The upper and lower airways share many physiological and immunological features, but there are also numerous differences. It is crucial to understand these differences and their contribution to pathophysiology in order to optimize treatments for inflammatory diseases of the respiratory tract. This review summarizes important differences in the embryological development, histological features, microbiota, immune responses, and cellular subtypes of mucosal epithelial cells of the nose and lungs.
Collapse
Affiliation(s)
- H-J Cho
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Korea.,Global Research Laboratory for Allergic Airway Disease, Yonsei University College of Medicine, Seoul, Korea.,The Airway Mucus Institute, Yonsei University College of Medicine, Seoul, Korea
| | - J G Ha
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Korea
| | - S N Lee
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Korea 2 Global Research Laboratory for Allergic Airway Disease, Yonsei University College of Medicine, Seoul, Korea
| | - C-H Kim
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Korea.,The Airway Mucus Institute, Yonsei University College of Medicine, Seoul, Korea
| | - D-Y Wang
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - J-H Yoon
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Korea.,Global Research Laboratory for Allergic Airway Disease, Yonsei University College of Medicine, Seoul, Korea.,The Airway Mucus Institute, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
20
|
Bukowy-Bieryłło Z. Long-term differentiating primary human airway epithelial cell cultures: how far are we? Cell Commun Signal 2021; 19:63. [PMID: 34044844 PMCID: PMC8159066 DOI: 10.1186/s12964-021-00740-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 04/16/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Human airway epithelial (HAE) cellular models are widely used in applicative studies of the airway physiology and disease. In vitro expanded and differentiated primary HAE cells collected from patients seem to be an accurate model of human airway, offering a quicker and cheaper alternative to the induced pluripotent stem cell (iPSCs) models. However, the biggest drawback of primary HAE models is their limited proliferative lifespan in culture. Much work has been devoted to understand the factors, which govern the HAE cell proliferation and differentiation, both in vivo and in vitro. Here, I have summarized recent achievements in primary HAE culture, with the special emphasis on the models of conditionally reprogrammed cells (CRC), which allow longer in vitro proliferation and differentiation of HAE cells. The review compares the CRC HAE technique variants (feeder culture or HAE mono-culture), based on recently published studies exploiting this model. The advantages and limitations of each CRC HAE model variant are summarized, along with the description of other factors affecting the CRC HAE culture success (tissue type, sampling method, sample quality). CONCLUSIONS CRC HAE cultures are a useful technique in respiratory research, which in many cases exceeds the iPSCs and organoid culture methods. Until the current limitations of the iPSCs and organoid culture methods will be alleviated, the primary CRC HAE cultures might be a useful model in respiratory research. Airway epithelium (AE) is a type of tissue, which lines the whole length of human airways, from the nose to the bronchi. Improper functioning of AE causes several human airway disorders, such as asthma, chronic obstructive pulmonary disease (COPD) or cystic fibrosis (CF). Much work has been devoted to finding the best scientific model of human AE, in order to learn about its functioning in health and disease. Among the popular AE models are the primary in vitro cultured AE cells collected from human donors. Unfortunately, such human AE (HAE) cells do not easily divide (expand) in vitro; this poses a large logistic and ethical problem for the researchers. Here, I summarize recent achievements in the methods for in vitro culture of human AE cells, with special emphasis on the conditionally reprogrammed cell (CRC) models, which allow longer and more effective expansion of primary human AE cells in vitro. The review describes how the specific chemicals used in the CRC models work to allow the increased HAE divisions and compares the effects of the different so-far developed variants of the CRC HAE culture. The review also pinpoints the areas which need to be refined, in order to maximize the usefulness of the CRC AE cultures from human donors in research on human airway disorders. Video abstract.
Collapse
|
21
|
Nasal Epithelial Cell-Based Models for Individualized Study in Cystic Fibrosis. Int J Mol Sci 2021; 22:ijms22094448. [PMID: 33923202 PMCID: PMC8123210 DOI: 10.3390/ijms22094448] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 04/22/2021] [Accepted: 04/23/2021] [Indexed: 12/11/2022] Open
Abstract
The emergence of highly effective CFTR modulator therapy has led to significant improvements in health care for most patients with cystic fibrosis (CF). For some, however, these therapies remain inaccessible due to the rarity of their individual CFTR variants, or due to a lack of biologic activity of the available therapies for certain variants. One proposed method of addressing this gap is the use of primary human cell-based models, which allow preclinical therapeutic testing and physiologic assessment of relevant tissue at the individual level. Nasal cells represent one such tissue source and have emerged as a powerful model for individual disease study. The ex vivo culture of nasal cells has evolved over time, and modern nasal cell models are beginning to be utilized to predict patient outcomes. This review will discuss both historical and current state-of-the art use of nasal cells for study in CF, with a particular focus on the use of such models to inform personalized patient care.
Collapse
|
22
|
From Submerged Cultures to 3D Cell Culture Models: Evolution of Nasal Epithelial Cells in Asthma Research and Virus Infection. Viruses 2021; 13:v13030387. [PMID: 33670992 PMCID: PMC7997270 DOI: 10.3390/v13030387] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 02/22/2021] [Accepted: 02/24/2021] [Indexed: 12/18/2022] Open
Abstract
Understanding the response to viral infection in the context of respiratory diseases is of significant importance. Recently, there has been more focus on the role of the nasal epithelium in disease modeling. Here, we provide an overview of different submerged, organotypic 3D and spheroid cell culture models of nasal epithelial cells, which were used to study asthma and allergy with a special focus on virus infection. In detail, this review summarizes the importance, benefits, and disadvantages of patient-derived cell culture models of nasal- and bronchial epithelial cells, including a comparison of these cell culture models and a discussion on why investigators should consider using nasal epithelial cells in their research. Exposure experiments, simple virus transduction analyses as well as genetic studies can be performed in these models, which may provide first insights into the complexity of molecular signatures and may open new doors for drug discovery and biomarker research.
Collapse
|
23
|
Kim MD, Baumlin N, Dennis JS, Yoshida M, Kis A, Aguiar C, Schmid A, Mendes E, Salathe M. Losartan reduces cigarette smoke-induced airway inflammation and mucus hypersecretion. ERJ Open Res 2021; 7:00394-2020. [PMID: 33532463 PMCID: PMC7836504 DOI: 10.1183/23120541.00394-2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 09/17/2020] [Indexed: 11/05/2022] Open
Abstract
The aim was to determine whether losartan reduces cigarette smoke (CS)-induced airway inflammation and mucus hypersecretion in an in vitro model and a small clinical trial. Primary human bronchial epithelial cells (HBECs) were differentiated at the air-liquid interface (ALI) and exposed to CS. Expression of transforming growth factor (TGF)-β1 and the mucin MUC5AC, and expression or activity of matrix metalloproteinase (MMP)-9 were measured after CS exposure. Parameters of mucociliary clearance were evaluated by measuring airway surface liquid volumes, mucus concentrations, and conductance of cystic fibrosis transmembrane conductance regulator (CFTR) and large conductance, Ca2+-activated and voltage-dependent potassium (BK) channels. Nasal cells were collected from study participants and expression of MUC5AC, TGF-β1, and MMP-9 mRNAs was measured before and after losartan treatment. In vitro, CS exposure of HBECs caused a significant increase in mRNA expression of MUC5AC and TGF-β1 and MMP-9 activity and decreased CFTR and BK channel activities, thereby reducing airway surface liquid volumes and increasing mucus concentrations. Treatment of HBECs with losartan rescued CS-induced CFTR and BK dysfunction and caused a significant decrease in MUC5AC expression and mucus concentrations, partially by inhibiting TGF-β signalling. In a prospective clinical study, cigarette smokers showed significantly reduced mRNA expression levels of MUC5AC, TGF-β1, and MMP-9 in the upper airways after 2 months of losartan treatment. Our findings suggest that losartan may be an effective therapy to reduce inflammation and mucus hypersecretion in CS-induced chronic airway diseases.
Collapse
Affiliation(s)
- Michael D Kim
- Dept of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, USA.,These authors contributed equally
| | - Nathalie Baumlin
- Dept of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, USA.,These authors contributed equally
| | - John S Dennis
- Dept of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Makoto Yoshida
- Dept of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Adrian Kis
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Carolina Aguiar
- Dept of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Andreas Schmid
- Dept of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Eliana Mendes
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Matthias Salathe
- Dept of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, USA.,Division of Pulmonary, Critical Care and Sleep Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
24
|
Miya C, Cueno ME, Suzuki R, Maruoka S, Gon Y, Kaneko T, Yonehara Y, Imai K. Porphyromonas gingivalis gingipains potentially affect MUC5AC gene expression and protein levels in respiratory epithelial cells. FEBS Open Bio 2020; 11:446-455. [PMID: 33332733 PMCID: PMC7876492 DOI: 10.1002/2211-5463.13066] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 11/02/2020] [Accepted: 12/15/2020] [Indexed: 01/08/2023] Open
Abstract
Porphyromonas gingivalis (Pg) is a periodontopathic pathogen that may affect MUC5AC‐related mucus hypersecretion along airway epithelial cells. Here, we attempted to establish whether Pg virulence factors (lipopolysaccharide, FimA fimbriae, gingipains) affect MUC5AC in immortalized and primary bronchial cells. We report that MUC5AC gene expression and protein levels are affected by Pg culture supernatant, but not by lipopolysaccharide or FimA fimbriae. Cells treated with either Pg single (Kgp or Rgp) or double (Kgp/Rgp) mutants had altered levels of MUC5AC gene expression and protein levels, and MUC5AC staining of double mutant‐treated mouse lung cells showed that MUC5AC protein levels were unaffected. Taken together, we propose that Pg gingipains may be the primary virulence factor that influences both MUC5AC gene expression and protein levels.
Collapse
Affiliation(s)
- Chihiro Miya
- Department of Oral and Maxillofacial Surgery II, Nihon University School of Dentistry, Tokyo, Japan.,Department of Microbiology, Nihon University School of Dentistry, Tokyo, Japan
| | - Marni E Cueno
- Department of Microbiology, Nihon University School of Dentistry, Tokyo, Japan
| | - Ryuta Suzuki
- Department of Oral and Maxillofacial Surgery II, Nihon University School of Dentistry, Tokyo, Japan.,Department of Microbiology, Nihon University School of Dentistry, Tokyo, Japan
| | - Shuichiro Maruoka
- Department of Internal Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Yasuhiro Gon
- Department of Internal Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Tadayoshi Kaneko
- Department of Oral and Maxillofacial Surgery II, Nihon University School of Dentistry, Tokyo, Japan
| | - Yoshiyuki Yonehara
- Department of Oral and Maxillofacial Surgery II, Nihon University School of Dentistry, Tokyo, Japan
| | - Kenichi Imai
- Department of Microbiology, Nihon University School of Dentistry, Tokyo, Japan
| |
Collapse
|
25
|
Bianchi M, Sivarajan R, Walles T, Hackenberg S, Steinke M. Susceptibility of primary human airway epithelial cells to Bordetella pertussis adenylate cyclase toxin in two- and three-dimensional culture conditions. Innate Immun 2020; 27:89-98. [PMID: 33317363 PMCID: PMC7780358 DOI: 10.1177/1753425920979354] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The human pathogen Bordetella pertussis targets the respiratory epithelium and causes whooping cough. Its virulence factor adenylate cyclase toxin (CyaA) plays an important role in the course of infection. Previous studies on the impact of CyaA on human epithelial cells have been carried out using cell lines derived from the airways or the intestinal tract. Here, we investigated the interaction of CyaA and its enzymatically inactive but fully pore-forming toxoid CyaA-AC– with primary human airway epithelial cells (hAEC) derived from different anatomical sites (nose and tracheo-bronchial region) in two-dimensional culture conditions. To assess possible differences between the response of primary hAEC and respiratory cell lines directly, we included HBEC3-KT in our studies. In comparative analyses, we studied the impact of both the toxin and the toxoid on cell viability, intracellular cAMP concentration and IL-6 secretion. We found that the selected hAEC, which lack CD11b, were differentially susceptible to both CyaA and CyaA-AC–. HBEC3-KT appeared not to be suitable for subsequent analyses. Since the nasal epithelium first gets in contact with airborne pathogens, we further studied the effect of CyaA and its toxoid on the innate immunity of three-dimensional tissue models of the human nasal mucosa. The present study reveals first insights in toxin–cell interaction using primary hAEC.
Collapse
Affiliation(s)
- Maria Bianchi
- Chair of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Germany
| | - Rinu Sivarajan
- Chair of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Germany
| | - Thorsten Walles
- Department of Thoracic Surgery, University Medicine Magdeburg, Germany
| | - Stephan Hackenberg
- Department of Oto-Rhino-Laryngology, Plastic, Aesthetic and Reconstructive Head and Neck Surgery, University Hospital Würzburg, Germany
| | - Maria Steinke
- Chair of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Germany
| |
Collapse
|
26
|
Laulajainen‐Hongisto A, Lyly A, Hanif T, Dhaygude K, Kankainen M, Renkonen R, Donner K, Mattila P, Jartti T, Bousquet J, Kauppi P, Toppila‐Salmi S. Genomics of asthma, allergy and chronic rhinosinusitis: novel concepts and relevance in airway mucosa. Clin Transl Allergy 2020; 10:45. [PMID: 33133517 PMCID: PMC7592594 DOI: 10.1186/s13601-020-00347-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 09/25/2020] [Indexed: 12/14/2022] Open
Abstract
Genome wide association studies (GWASs) have revealed several airway disease-associated risk loci. Their role in the onset of asthma, allergic rhinitis (AR) or chronic rhinosinusitis (CRS), however, is not yet fully understood. The aim of this review is to evaluate the airway relevance of loci and genes identified in GWAS studies. GWASs were searched from databases, and a list of loci associating significantly (p < 10-8) with asthma, AR and CRS was created. This yielded a total of 267 significantly asthma/AR-associated loci from 31 GWASs. No significant CRS -associated loci were found in this search. A total of 170 protein coding genes were connected to these loci. Of these, 76/170 (44%) showed bronchial epithelial protein expression in stained microscopic figures of Human Protein Atlas (HPA), and 61/170 (36%) had a literature report of having airway epithelial function. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) annotation analyses were performed, and 19 functional protein categories were found as significantly (p < 0.05) enriched among these genes. These were related to cytokine production, cell activation and adaptive immune response, and all were strongly connected in network analysis. We also identified 15 protein pathways that were significantly (p < 0.05) enriched in these genes, related to T-helper cell differentiation, virus infection, JAK-STAT signaling pathway, and asthma. A third of GWAS-level risk loci genes of asthma or AR seemed to have airway epithelial functions according to our database and literature searches. In addition, many of the risk loci genes were immunity related. Some risk loci genes also related to metabolism, neuro-musculoskeletal or other functions. Functions overlapped and formed a strong network in our pathway analyses and are worth future studies of biomarker and therapeutics.
Collapse
Affiliation(s)
- Anu Laulajainen‐Hongisto
- Department of Otorhinolaryngology–Head and Neck SurgeryUniversity of Helsinki and Helsinki University HospitalP.O.Box 263Kasarmikatu 11‐1300029 HUSHelsinkiFinland
- Laboratory of Cellular and Molecular ImmunologyInstitute of Microbiology of the Czech Academy of SciencesPragueCzech Republic
| | - Annina Lyly
- Department of Otorhinolaryngology–Head and Neck SurgeryUniversity of Helsinki and Helsinki University HospitalP.O.Box 263Kasarmikatu 11‐1300029 HUSHelsinkiFinland
- Skin and Allergy HospitalUniversity of Helsinki and Helsinki University HospitalHelsinkiFinland
| | | | | | - Matti Kankainen
- HUS Diagnostic CenterHelsinki University HospitalHelsinkiFinland
- Hematology Research Unit HelsinkiDepartment of HematologyHelsinki University Hospital Comprehensive Cancer CenterHelsinkiFinland
- Translational Immunology Research Program and Department of Clinical ChemistryUniversity of HelsinkiHelsinkiFinland
| | - Risto Renkonen
- Haartman InstituteUniversity of HelsinkiHelsinkiFinland
- HUS Diagnostic CenterHelsinki University HospitalHelsinkiFinland
| | - Kati Donner
- Hematology Research Unit HelsinkiDepartment of HematologyHelsinki University Hospital Comprehensive Cancer CenterHelsinkiFinland
| | - Pirkko Mattila
- Haartman InstituteUniversity of HelsinkiHelsinkiFinland
- Hematology Research Unit HelsinkiDepartment of HematologyHelsinki University Hospital Comprehensive Cancer CenterHelsinkiFinland
| | - Tuomas Jartti
- Department of Pediatrics and Adolescent MedicineTurku University Hospital and University of TurkuTurkuFinland
| | - Jean Bousquet
- Université MontpellierMontpellierFrance
- MACVIA‐FranceMontpellierFrance
- Corporate Member of Freie Universität BerlinHumboldt‐Universität Zu BerlinBerlin Institute of HealthComprehensive Allergy CenterDepartment of Dermatology and AllergyCharité–Universitätsmedizin BerlinBerlinGermany
| | - Paula Kauppi
- Skin and Allergy HospitalUniversity of Helsinki and Helsinki University HospitalHelsinkiFinland
| | - Sanna Toppila‐Salmi
- Skin and Allergy HospitalUniversity of Helsinki and Helsinki University HospitalHelsinkiFinland
- Haartman InstituteUniversity of HelsinkiHelsinkiFinland
| |
Collapse
|
27
|
Rayner RE, Wellmerling J, Osman W, Honesty S, Alfaro M, Peeples ME, Cormet-Boyaka E. In vitro 3D culture lung model from expanded primary cystic fibrosis human airway cells. J Cyst Fibros 2020; 19:752-761. [PMID: 32565193 DOI: 10.1016/j.jcf.2020.05.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 04/13/2020] [Accepted: 05/23/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND In vitro cystic fibrosis (CF) models are crucial for understanding the mechanisms and consequences of the disease. They are also the gold standard for pre-clinical efficacy studies of current and novel CF drugs. However, few studies have investigated expansion and differentiation of primary CF human bronchial epithelial (CF-HBE) cells. Here we describe culture conditions to expand primary CF airway cells while preserving their ability to differentiate into 3D epithelial cultures expressing functional cystic fibrosis transmembrane conductance regulator (CFTR) ion channels that responds to CFTR modulators. METHODS Primary CF airway cells were expanded using PneumaCultTM-Ex Plus (StemCell Technologies) medium with no feeder cells or added Rho kinase (ROCK) inhibitor. Differentially passaged CF-HBE cells at the air-liquid interface (ALI) were characterized phenotypically and functionally in response to the CFTR corrector drug VX-661 (Tezacaftor). RESULTS CF-HBE primary cells, expanded up to six passages (~25 population doublings), differentiated into 3D epithelial cultures as evidenced by trans-epithelial electrical resistance (TEER) of >400 Ohms∙cm2 and presence of pseudostratified columnar ciliated epithelium with goblet cells. However, up to passage five cells from most donors showed increased CFTR-mediated short-circuit currents when treated with the corrector drug, VX-661. Ciliary beat frequency (CBF) also increased with the corrector VX-661. CONCLUSIONS CF donor-derived airway cells can be expanded without the use of feeder cells or additional ROCK inhibitor, and still achieve optimal 3D epithelial cultures that respond to CFTR modulators. The study of rare CF mutations could benefit from cell expansion and could lead to the design of personalized medicine/treatments.
Collapse
Affiliation(s)
- Rachael E Rayner
- Department of Veterinary Biosciences, The Ohio State University, 1925 Coffey Road, Columbus, OH 43210, USA
| | - Jack Wellmerling
- Department of Veterinary Biosciences, The Ohio State University, 1925 Coffey Road, Columbus, OH 43210, USA
| | - Wissam Osman
- Department of Veterinary Biosciences, The Ohio State University, 1925 Coffey Road, Columbus, OH 43210, USA
| | - Sean Honesty
- Department of Veterinary Biosciences, The Ohio State University, 1925 Coffey Road, Columbus, OH 43210, USA
| | - Maria Alfaro
- Institute for Genomic Medicine, Abigail Wexner Research Institute, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH 43205, USA
| | - Mark E Peeples
- Center for Vaccines and Immunity, Abigail Wexner Research Institute, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH 43205, USA; Department of Pediatrics, The Ohio State University College of Medicine, 700 Children's Drive, Columbus, OH 43205, USA
| | - Estelle Cormet-Boyaka
- Department of Veterinary Biosciences, The Ohio State University, 1925 Coffey Road, Columbus, OH 43210, USA.
| |
Collapse
|
28
|
Mavin E, Verdon B, Carrie S, Saint-Criq V, Powell J, Kuttruff CA, Ward C, Garnett JP, Miwa S. Real-time measurement of cellular bioenergetics in fully differentiated human nasal epithelial cells grown at air-liquid-interface. Am J Physiol Lung Cell Mol Physiol 2020; 318:L1158-L1164. [PMID: 32267720 PMCID: PMC7347273 DOI: 10.1152/ajplung.00414.2019] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Shifts in cellular metabolic phenotypes have the potential to cause disease-driving processes in respiratory disease. The respiratory epithelium is particularly susceptible to metabolic shifts in disease, but our understanding of these processes is limited by the incompatibility of the technology required to measure metabolism in real-time with the cell culture platforms used to generate differentiated respiratory epithelial cell types. Thus, to date, our understanding of respiratory epithelial metabolism has been restricted to that of basal epithelial cells in submerged culture, or via indirect end point metabolomics readouts in lung tissue. Here we present a novel methodology using the widely available Seahorse Analyzer platform to monitor real-time changes in the cellular metabolism of fully differentiated primary human airway epithelial cells grown at air-liquid interface (ALI). We show increased glycolytic, but not mitochondrial, ATP production rates in response to physiologically relevant increases in glucose availability. We also show that pharmacological inhibition of lactate dehydrogenase is able to reduce glucose-induced shifts toward aerobic glycolysis. This method is timely given the recent advances in our understanding of new respiratory epithelial subtypes that can only be observed in vitro through culture at ALI and will open new avenues to measure real-time metabolic changes in healthy and diseased respiratory epithelium, and in turn the potential for the development of novel therapeutics targeting metabolic-driven disease phenotypes.
Collapse
Affiliation(s)
- Emily Mavin
- Institute of Cellular Medicine, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Bernard Verdon
- Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Sean Carrie
- Institute of Health and Society, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Vinciane Saint-Criq
- Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Jason Powell
- Institute of Cellular Medicine, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | | | - Chris Ward
- Institute of Cellular Medicine, Newcastle University, Newcastle Upon Tyne, United Kingdom.,Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - James P Garnett
- Institute of Cellular Medicine, Newcastle University, Newcastle Upon Tyne, United Kingdom.,Immunology and Respiratory Diseases Research, Boehringer Ingelheim Pharma, Biberach an der Riss, Germany
| | - Satomi Miwa
- Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle Upon Tyne, United Kingdom
| |
Collapse
|
29
|
Gilpin DF, McGown KA, Gallagher K, Bengoechea J, Dumigan A, Einarsson G, Elborn JS, Tunney MM. Electronic cigarette vapour increases virulence and inflammatory potential of respiratory pathogens. Respir Res 2019; 20:267. [PMID: 31847850 PMCID: PMC6918581 DOI: 10.1186/s12931-019-1206-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 10/04/2019] [Indexed: 12/20/2022] Open
Abstract
INTRODUCTION Bacteria have been extensively implicated in the development of smoking related diseases, such as COPD, by either direct infection or bacteria-mediated inflammation. In response to the health risks associated with tobacco exposure, the use of electronic cigarettes (e-cigs) has increased. This study compared the effect of e-cig vapour (ECV) and cigarette smoke (CSE) on the virulence and inflammatory potential of key lung pathogens (Haemophilus influenzae, Streptococcus pneumoniae, Staphylococcus aureus and Pseudomonas aeruginosa). METHODS Biofilm formation, virulence in the Galleria mellonella infection model, antibiotic susceptibility and IL-8/TNF-α production in A549 cells, were compared between bacteria exposed to ECV, CSE and non-exposed bacteria. RESULTS Statistically significant increases in biofilm and cytokine secretion were observed following bacterial exposure to either ECV or CSE, compared to non-exposed bacteria; the effect of exposure to ECV on bacterial phenotype and virulence was comparable, and in some cases greater, than that observed following CSE exposure. Treatment of A549 cells with cell signaling pathway inhibitors prior to infection, did not suggest that alternative signaling pathways were being activated following exposure of bacteria to either ECV or CSE. CONCLUSIONS These findings therefore suggest that ECV and CSE can induce changes in phenotype and virulence of key lung pathogens, which may increase bacterial persistence and inflammatory potential.
Collapse
Affiliation(s)
- Deirdre F Gilpin
- Halo Research Group, School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast, Northern Ireland, BT9 7BL, UK.
| | - Katie-Ann McGown
- Halo Research Group, School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast, Northern Ireland, BT9 7BL, UK
| | - Kevin Gallagher
- Halo Research Group, School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast, Northern Ireland, BT9 7BL, UK
| | - Jose Bengoechea
- Centre for Experimental Medicine, Queen's University Belfast, 97 Lisburn Road, Belfast, Northern Ireland, BT9 7BL, UK
| | - Amy Dumigan
- Centre for Experimental Medicine, Queen's University Belfast, 97 Lisburn Road, Belfast, Northern Ireland, BT9 7BL, UK
| | - Gisli Einarsson
- Halo Research Group, School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast, Northern Ireland, BT9 7BL, UK
| | - J Stuart Elborn
- Centre for Experimental Medicine, Queen's University Belfast, 97 Lisburn Road, Belfast, Northern Ireland, BT9 7BL, UK
| | - Michael M Tunney
- Halo Research Group, School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast, Northern Ireland, BT9 7BL, UK
| |
Collapse
|
30
|
Cell-Type-Specific Transcription of Innate Immune Regulators in response to HMPV Infection. Mediators Inflamm 2019; 2019:4964239. [PMID: 31686982 PMCID: PMC6803734 DOI: 10.1155/2019/4964239] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Revised: 06/18/2019] [Accepted: 08/24/2019] [Indexed: 12/17/2022] Open
Abstract
Human metapneumovirus (HMPV) may cause severe respiratory disease. The early innate immune response to viruses like HMPV is characterized by induction of antiviral interferons (IFNs) and proinflammatory immune mediators that are essential in shaping adaptive immune responses. Although innate immune responses to HMPV have been comprehensively studied in mice and murine immune cells, there is less information on these responses in human cells, comparing different cell types infected with the same HMPV strain. The aim of this study was to characterize the HMPV-induced mRNA expression of critical innate immune mediators in human primary cells relevant for airway disease. In particular, we determined type I versus type III IFN expression in human epithelial cells and monocyte-derived macrophages (MDMs) and dendritic cells (MDDCs). In epithelial cells, HMPV induced only low levels of IFN-β mRNA, while a robust mRNA expression of IFN-λs was found in epithelial cells, MDMs, and MDDCs. In addition, we determined induction of the interferon regulatory factors (IRFs) IRF1, IRF3, and IRF7 and critical inflammatory cytokines (IL-6, IP-10, and IL-1β). Interestingly, IRF1 mRNA was predominantly induced in MDMs and MDDCs. Overall, our results suggest that for HMPV infection of MDDCs, MDMs, NECs, and A549 cells (the cell types examined), cell type is a strong determinator of the ability of HMPV to induce different innate immune mediators. HMPV induces the transcription of IFN-β and IRF1 to higher extents in MDMs and MDDCs than in A549s and NECs, whereas the induction of type III IFN-λ and IRF7 is considerable in MDMs, MDDCs, and A549 epithelial cells.
Collapse
|
31
|
Groves HE, Guo-Parke H, Broadbent L, Shields MD, Power UF. Characterisation of morphological differences in well-differentiated nasal epithelial cell cultures from preterm and term infants at birth and one-year. PLoS One 2018; 13:e0201328. [PMID: 30517096 PMCID: PMC6281239 DOI: 10.1371/journal.pone.0201328] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 11/13/2018] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Innate immune responses of airway epithelium are important defences against respiratory pathogens and allergens. Newborn infants are at greater risk of severe respiratory infections compared to older infants, while premature infants are at greater risk than full term infants. However, very little is known regarding human neonatal airway epithelium immune responses and whether age-related morphological and/or innate immune changes contribute to the development of airway disease. METHODS We collected nasal epithelial cells from 41 newborn infants (23 term, 18 preterm) within 5 days of birth. Repeat sampling was achieved for 24 infants (13 term, 11 preterm) at a median age of 12.5 months. Morphologically- and physiologically-authentic well-differentiated primary paediatric nasal epithelial cell (WD-PNEC) cultures were generated and characterised using light microscopy and immunofluorescence. RESULTS WD-PNEC cultures were established for 15/23 (65%) term and 13/18 (72%) preterm samples at birth, and 9/13 (69%) term and 8/11 (73%) preterm samples at one-year. Newborn and infant WD-PNEC cultures demonstrated extensive cilia coverage, mucous production and tight junction integrity. Newborn WD-PNECs took significantly longer to reach full differentiation and were noted to have much greater proportions of goblet cells compared to one-year repeat WD-PNECs. No differences were evident in ciliated/goblet cell proportions between term- and preterm-derived WD-PNECs at birth or one-year old. CONCLUSION We describe the successful generation of newborn-derived WD-PNEC cultures and their revival from frozen. We also compared the characteristics of WD-PNECs derived from infants born at term with those born prematurely at birth and at one-year-old. The development of WD-PNEC cultures from newborn infants provides a powerful and exciting opportunity to study the development of airway epithelium morphology, physiology, and innate immune responses to environmental or infectious insults from birth.
Collapse
Affiliation(s)
- Helen E. Groves
- Wellcome Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Sciences, The Queen’s University of Belfast, Belfast, Northern Ireland, United Kingdom
| | - Hong Guo-Parke
- Wellcome Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Sciences, The Queen’s University of Belfast, Belfast, Northern Ireland, United Kingdom
| | - Lindsay Broadbent
- Wellcome Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Sciences, The Queen’s University of Belfast, Belfast, Northern Ireland, United Kingdom
| | - Michael D. Shields
- Wellcome Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Sciences, The Queen’s University of Belfast, Belfast, Northern Ireland, United Kingdom
- Department of Paediatric Respiratory Medicine, Royal Belfast Hospital for Sick Children, Belfast, Northern Ireland, United Kingdom
| | - Ultan F. Power
- Wellcome Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Sciences, The Queen’s University of Belfast, Belfast, Northern Ireland, United Kingdom
| |
Collapse
|
32
|
Castellani S, Di Gioia S, di Toma L, Conese M. Human Cellular Models for the Investigation of Lung Inflammation and Mucus Production in Cystic Fibrosis. Anal Cell Pathol (Amst) 2018; 2018:3839803. [PMID: 30581723 PMCID: PMC6276497 DOI: 10.1155/2018/3839803] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 09/04/2018] [Accepted: 09/23/2018] [Indexed: 12/12/2022] Open
Abstract
Chronic inflammation, oxidative stress, mucus plugging, airway remodeling, and respiratory infections are the hallmarks of the cystic fibrosis (CF) lung disease. The airway epithelium is central in the innate immune responses to pathogens colonizing the airways, since it is involved in mucociliary clearance, senses the presence of pathogens, elicits an inflammatory response, orchestrates adaptive immunity, and activates mesenchymal cells. In this review, we focus on cellular models of the human CF airway epithelium that have been used for studying mucus production, inflammatory response, and airway remodeling, with particular reference to two- and three-dimensional cultures that better recapitulate the native airway epithelium. Cocultures of airway epithelial cells, macrophages, dendritic cells, and fibroblasts are instrumental in disease modeling, drug discovery, and identification of novel therapeutic targets. Nevertheless, they have to be implemented in the CF field yet. Finally, novel systems hijacking on tissue engineering, including three-dimensional cocultures, decellularized lungs, microfluidic devices, and lung organoids formed in bioreactors, will lead the generation of relevant human preclinical respiratory models a step forward.
Collapse
Affiliation(s)
- Stefano Castellani
- Laboratory of Regenerative and Experimental Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Sante Di Gioia
- Laboratory of Regenerative and Experimental Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Lorena di Toma
- Laboratory of Regenerative and Experimental Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Massimo Conese
- Laboratory of Regenerative and Experimental Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| |
Collapse
|
33
|
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: 1.5] [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
|
34
|
Giovannini-Chami L, Paquet A, Sanfiorenzo C, Pons N, Cazareth J, Magnone V, Lebrigand K, Chevalier B, Vallauri A, Julia V, Marquette CH, Marcet B, Leroy S, Barbry P. The "one airway, one disease" concept in light of Th2 inflammation. Eur Respir J 2018; 52:13993003.00437-2018. [PMID: 30190271 DOI: 10.1183/13993003.00437-2018] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 08/13/2018] [Indexed: 12/19/2022]
Abstract
In line with the pathophysiological continuum described between nose and bronchus in allergic respiratory diseases, we assessed whether nasal epithelium could mirror the Type 2 T-helper cell (Th2) status of bronchial epithelium.Nasal and bronchial cells were collected by brushing from healthy controls (C, n=13), patients with allergic rhinitis and asthma (AR, n=12), and patients with isolated allergic rhinitis (R, n=14). Cellular composition was assessed by flow cytometry, gene expression was analysed by RNA sequencing and Th2, Type 17 T-helper cell (Th17) and interferon (IFN) signatures were derived from the literature.Infiltration by polymorphonuclear neutrophils (PMN) in the nose excluded 30% of the initial cohort. All bronchial samples from the AR group were Th2-high. The gene expression profile of nasal samples from the AR group correctly predicted the paired bronchial sample Th2 status in 71% of cases. Nevertheless, nasal cells did not appear to be a reliable surrogate for the Th2 response, in particular due to a more robust influence of the IFN response in 14 out of 26 nasal samples. The Th2 scores in the nose and bronchi correlated with mast cell count (both p<0.001) and number of sensitisations (p=0.006 and 0.002), while the Th17 scores correlated with PMN count (p=0.006 and 0.003).The large variability in nasal cell composition and type of inflammation restricts its use as a surrogate for assessing bronchial Th2 inflammation in AR patients.
Collapse
Affiliation(s)
- Lisa Giovannini-Chami
- Pediatric Pulmonology and Allergology Dept, Hôpitaux Pédiatriques de Nice CHU-Lenval, Université Côte d'Azur, Nice, France.,Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, Université Côte d'Azur, Sophia Antipolis, France.,These authors contributed equally to this work
| | - Agnès Paquet
- Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, Université Côte d'Azur, Sophia Antipolis, France.,These authors contributed equally to this work
| | - Céline Sanfiorenzo
- Pulmonology Dept, FHU Oncoage, CHU de Nice, Université Côte d'Azur, Nice, France
| | - Nicolas Pons
- Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, Université Côte d'Azur, Sophia Antipolis, France
| | - Julie Cazareth
- Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, Université Côte d'Azur, Sophia Antipolis, France
| | - Virginie Magnone
- Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, Université Côte d'Azur, Sophia Antipolis, France
| | - Kévin Lebrigand
- Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, Université Côte d'Azur, Sophia Antipolis, France
| | - Benoit Chevalier
- Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, Université Côte d'Azur, Sophia Antipolis, France
| | - Ambre Vallauri
- Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, Université Côte d'Azur, Sophia Antipolis, France
| | - Valérie Julia
- Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, Université Côte d'Azur, Sophia Antipolis, France
| | | | - Brice Marcet
- Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, Université Côte d'Azur, Sophia Antipolis, France
| | - Sylvie Leroy
- Pulmonology Dept, FHU Oncoage, CHU de Nice, Université Côte d'Azur, Nice, France.,These authors contributed equally to this work
| | - Pascal Barbry
- Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, Université Côte d'Azur, Sophia Antipolis, France.,These authors contributed equally to this work
| |
Collapse
|
35
|
Roberts N, Al Mubarak R, Francisco D, Kraft M, Chu HW. Comparison of paired human nasal and bronchial airway epithelial cell responses to rhinovirus infection and IL-13 treatment. Clin Transl Med 2018; 7:13. [PMID: 29721720 PMCID: PMC5931947 DOI: 10.1186/s40169-018-0189-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 04/22/2018] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Because of its advantage as a minimally invasive procedure, nasal brushings have been increasingly used and proposed as a valuable approach to study lower airway diseases in lieu of bronchial epithelial cells. However, there is limited or conflicting evidence pertaining to whether nasal samples can be surrogates to bronchial samples. The goal of the present study is to test whether nasal epithelial cells have similar antiviral and inflammatory responses to IL-13 treatment and rhinovirus infection, a condition mimicking virally induced asthma exacerbation. Nasal and bronchial airway epithelial cells taken from the same patient were cultured under submerged and air-liquid interface (ALI) culture in the absence or presence of rhinovirus and IL-13 treatment. Inflammatory cytokines IP-10 and eotaxin-3, antiviral gene Mx1 and viral levels were measured. RESULTS In the absence of IL-13 treatment, nasal and bronchial cells showed a similar IP-10 response in both ALI and submerged cultures. Under the ALI culture, short term (e.g., 3 days) IL-13 treatment had a minimal effect on viral and Mx1 levels in both cell types. However, prolonged (e.g., 14 days) IL-13 treatments in both cell types decreased viral load and Mx1 expression. Under the submerged culture, IL-13 treatment in both cell types has minimal effects on viral load, IP-10 and Mx1. IL-13-induced eotaxin-3 production was similar in both types of cells under either submerged or ALI culture, which was not affected by viral infection. CONCLUSIONS Our data suggest that nasal epithelial cells could serve as a surrogate to bronchial epithelial cells in future studies aimed at defining the role of type 2 cytokine IL-13 in regulating pro-inflammatory and antiviral responses.
Collapse
Affiliation(s)
- Nicole Roberts
- Department of Medicine, National Jewish Health, 1400 Jackson Street, Room A639, Denver, CO, 80206, USA
| | - Reem Al Mubarak
- Department of Medicine, National Jewish Health, 1400 Jackson Street, Room A639, Denver, CO, 80206, USA
| | - David Francisco
- Department of Medicine, University of Arizona, Tucson, AZ, USA
| | - Monica Kraft
- Department of Medicine, University of Arizona, Tucson, AZ, USA
| | - Hong Wei Chu
- Department of Medicine, National Jewish Health, 1400 Jackson Street, Room A639, Denver, CO, 80206, USA.
| |
Collapse
|
36
|
Schögler A, Blank F, Brügger M, Beyeler S, Tschanz SA, Regamey N, Casaulta C, Geiser T, Alves MP. Characterization of pediatric cystic fibrosis airway epithelial cell cultures at the air-liquid interface obtained by non-invasive nasal cytology brush sampling. Respir Res 2017; 18:215. [PMID: 29282053 PMCID: PMC5745630 DOI: 10.1186/s12931-017-0706-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 12/18/2017] [Indexed: 12/16/2022] Open
Abstract
Background In vitro systems of primary cystic fibrosis (CF) airway epithelial cells are an important tool to study molecular and functional features of the native respiratory epithelium. However, undifferentiated CF airway cell cultures grown under submerged conditions do not appropriately represent the physiological situation. A more advanced CF cell culture system based on airway epithelial cells grown at the air-liquid interface (ALI) recapitulates most of the in vivo-like properties but requires the use of invasive sampling methods. In this study, we describe a detailed characterization of fully differentiated primary CF airway epithelial cells obtained by non-invasive nasal brushing of pediatric patients. Methods Differentiated cell cultures were evaluated with immunolabelling of markers for ciliated, mucus-secreting and basal cells, and tight junction and CFTR proteins. Epithelial morphology and ultrastructure was examined by histology and transmission electron microscopy. Ciliary beat frequency was investigated by a video-microscopy approach and trans-epithelial electrical resistance was assessed with an epithelial Volt-Ohm meter system. Finally, epithelial permeability was analysed by using a cell layer integrity test and baseline cytokine levels where measured by an enzyme-linked immunosorbent assay. Results Pediatric CF nasal cultures grown at the ALI showed a differentiation into a pseudostratified epithelium with a mucociliary phenotype. Also, immunofluorescence analysis revealed the presence of ciliated, mucus-secreting and basal cells and tight junctions. CFTR protein expression was observed in CF (F508del/F508del) and healthy cultures and baseline interleukin (IL)-8 and IL-6 release were similar in control and CF ALI cultures. The ciliary beat frequency was 9.67 Hz and the differentiated pediatric CF epithelium was found to be functionally tight. Conclusion In summary, primary pediatric CF nasal epithelial cell cultures grown at the ALI showed full differentiation into ciliated, mucus-producing and basal cells, which adequately reflect the in vivo properties of the human respiratory epithelium. Electronic supplementary material The online version of this article (10.1186/s12931-017-0706-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Aline Schögler
- Department of Clinical Research, University of Bern, Bern, Switzerland.,Division of Respiratory Medicine, University Children's Hospital of Bern, Bern, Switzerland
| | - Fabian Blank
- Department of Clinical Research, University of Bern, Bern, Switzerland.,Department of Pulmonary Medicine, University Hospital of Bern, Bern, Switzerland
| | - Melanie Brügger
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland.,Institute of Virology and Immunology, Federal Department of Home Affairs, Mittelhäusern, Switzerland.,Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Seraina Beyeler
- Department of Clinical Research, University of Bern, Bern, Switzerland.,Department of Pulmonary Medicine, University Hospital of Bern, Bern, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | | | | | - Carmen Casaulta
- Division of Respiratory Medicine, University Children's Hospital of Bern, Bern, Switzerland
| | - Thomas Geiser
- Department of Pulmonary Medicine, University Hospital of Bern, Bern, Switzerland
| | - Marco P Alves
- Department of Clinical Research, University of Bern, Bern, Switzerland. .,Division of Respiratory Medicine, University Children's Hospital of Bern, Bern, Switzerland. .,Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland. .,Institute of Virology and Immunology, Federal Department of Home Affairs, Mittelhäusern, Switzerland. .,Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland.
| |
Collapse
|
37
|
Forero A, Fenstermacher K, Wohlgemuth N, Nishida A, Carter V, Smith EA, Peng X, Hayes M, Francis D, Treanor J, Morrison J, Klein SL, Lane A, Katze MG, Pekosz A. Evaluation of the innate immune responses to influenza and live-attenuated influenza vaccine infection in primary differentiated human nasal epithelial cells. Vaccine 2017; 35:6112-6121. [PMID: 28967519 DOI: 10.1016/j.vaccine.2017.09.058] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 09/13/2017] [Accepted: 09/19/2017] [Indexed: 01/08/2023]
Abstract
The host innate immune response to influenza virus is a key determinant of pathogenic outcomes and long-term protective immune responses against subsequent exposures. Here, we present a direct contrast of the host responses in primary differentiated human nasal epithelial cell (hNEC) cultures following infection with either a seasonal H3N2 influenza virus (WT) or the antigenically-matched live-attenuated vaccine (LAIV) strain. Comparison of the transcriptional profiles obtained 24 and 36h post-infection showed that the magnitude of gene expression was greater in LAIV infected relative to that observed in WT infected hNEC cultures. Functional enrichment analysis revealed that the antiviral and inflammatory responses were largely driven by type III IFN induction in both WT and LAIV infected cells. However, the enrichment of biological pathways involved in the recruitment of mononuclear leukocytes, antigen-presenting cells, and T lymphocytes was uniquely observed in LAIV infected cells. These observations were reflective of the host innate immune responses observed in individuals acutely infected with influenza viruses. These findings indicate that cell-intrinsic type III IFN-mediated innate immune responses in the nasal epithelium are not only crucial for viral clearance and attenuation, but may also play an important role in the induction of protective immune responses with live-attenuated vaccines.
Collapse
Affiliation(s)
- Adriana Forero
- Department of Microbiology, University of Washington, Seattle, WA, USA
| | - Katherine Fenstermacher
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Nicholas Wohlgemuth
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Andrew Nishida
- Department of Microbiology, University of Washington, Seattle, WA, USA
| | - Victoria Carter
- Department of Microbiology, University of Washington, Seattle, WA, USA
| | - Elise A Smith
- Department of Microbiology, University of Washington, Seattle, WA, USA
| | - Xinxia Peng
- Department of Microbiology, University of Washington, Seattle, WA, USA
| | - Melissa Hayes
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Doreen Francis
- Department of Internal Medicine, University of Rochester, School of Medicine and Dentistry, Rochester, New York, USA
| | - John Treanor
- Department of Internal Medicine, University of Rochester, School of Medicine and Dentistry, Rochester, New York, USA
| | - Juliet Morrison
- Department of Microbiology, University of Washington, Seattle, WA, USA
| | - Sabra L Klein
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Andrew Lane
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Michael G Katze
- Department of Microbiology, University of Washington, Seattle, WA, USA; Washington National Primate Research Center, University of Washington, Seattle, WA, USA
| | - Andrew Pekosz
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA; Department of Environmental Health Sciences, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA.
| |
Collapse
|
38
|
Novel freeze-dried DDA and TPGS liposomes are suitable for nasal delivery of vaccine. Int J Pharm 2017; 533:179-186. [PMID: 28887219 DOI: 10.1016/j.ijpharm.2017.09.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 08/31/2017] [Accepted: 09/05/2017] [Indexed: 11/21/2022]
Abstract
There is a pressing need for effective needle-free vaccines that are stable enough for use in the developing world and stockpiling. The inclusion of the cationic lipid DDA and the PEG-containing moiety TPGS into liposomes has the potential to improve mucosal delivery. The aim of this study was to develop stable lyophilized cationic liposomes based on these materials suitable for nasal antigen delivery. Liposomes containing DDA and TPGS were developed. Size and zeta potential measurements, ex vivo, CLSM cell penetration study and cell viability investigations were made. Preliminary immunisation and stability studies using ovalbumin were performed. The liposomes exhibited suitable size and charge for permeation across nasal mucosa. DDA and TPGS increased tissue permeation in ex vivo studies and cell uptake with good cell viability. The liposomes improved immune response both locally and vaginally when compared to i.m administration or control liposomes delivered nasally. Additionally, the lyophilized products demonstrated good stability in terms of Tg, size and antigen retention. This study has shown that the novel liposomes have potential for development as a mucosal vaccine delivery system. Furthermore, the stability of the lyophilized liposomes offers potential additional benefits in terms of thermal stability over liquid formats.
Collapse
|
39
|
Liu SC, Lu HH, Fan HC, Wang HW, Chen HK, Lee FP, Yu CJ, Chu YH. The identification of the TRPM8 channel on primary culture of human nasal epithelial cells and its response to cooling. Medicine (Baltimore) 2017; 96:e7640. [PMID: 28767579 PMCID: PMC5626133 DOI: 10.1097/md.0000000000007640] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND It has been proposed that the transient receptor potential (TRP) channel Melastatin 8 (TRPM8) is a cold-sensing TRP channel. However, its presence and its role in the nasal cavity have not yet been fully studied. METHODS Immunohistology was used to study TRPM8 receptors in both the nasal mucosa tissue and the primary cultures of human nasal cells. Cells from primary cultures were immunostained with antibodies to TRPM8, mucin, cytokeratin (CK)-14, CK-18, and vimentin. Western blotting and real-time polymerase chain reaction (PCR) were used to determine the physiological role of TRPM8 in mucus production in the nasal cavity, with and without its agonist and antagonist. RESULTS The TRPM8 is clearly present in the epithelium, mucous glands, and vessels. No obvious TRPM8-immunoreactive cells were detected in the connective tissue. Immunostaining of cytospin preparations showed that epithelial cells test positive for CK-14, CK-18, TRPM8, and mucin 5AC (MUC5AC). Fibroblastic cells are stained negative for TRPM8. Secreted mucins in the cultured supernatant are detected after exposure to menthol and moderate cooling to 24°C. Both induce a statistically significant increase in the level of MUC5AC mRNA and mucin production. BCTC, a TRPM8 antagonist, has a statistically significant inhibitory effect on MUC5AC mRNA expression and MUC5AC protein production that is induced by menthol and moderate cooling to 24°C. CONCLUSIONS The study demonstrates that TRPM8 is present in the nasal epithelium. When it is activated by moderate cooling to 24°C or menthol, TRPM8 induces the secretion of mucin. This study shows that TRPM8 channels are important regulators of mucin production. Therefore, TRPM8 antagonists could be used to treat refractory rhinitis.
Collapse
Affiliation(s)
- Shao-Cheng Liu
- Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University
| | - Hsuan-Hsuan Lu
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University
- Department of Internal Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University
| | - Hueng-Chuen Fan
- Department of Pediatrics, Tungs’ Taichung Metro Harbor Hospital
| | - Hsing-Won Wang
- Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University
- Department of Otolaryngology-Head and Neck Surgery, Shuang Ho Hospital, Taipei, Taiwan, Republic of China
| | - Hang-Kang Chen
- Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center
| | - Fei-Peng Lee
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University
| | - Chong-Jen Yu
- Department of Internal Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University
| | - Yueng-Hsiang Chu
- Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center
| |
Collapse
|
40
|
Talikka M, Martin F, Sewer A, Vuillaume G, Leroy P, Luettich K, Chaudhary N, Peck MJ, Peitsch MC, Hoeng J. Mechanistic Evaluation of the Impact of Smoking and Chronic Obstructive Pulmonary Disease on the Nasal Epithelium. CLINICAL MEDICINE INSIGHTS-CIRCULATORY RESPIRATORY AND PULMONARY MEDICINE 2017; 11:1179548417710928. [PMID: 28620266 PMCID: PMC5466113 DOI: 10.1177/1179548417710928] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 04/04/2017] [Indexed: 12/27/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is one of the major causes of chronic morbidity and mortality worldwide. The development of markers of COPD onset is hampered by the lack of accessibility to the primary target tissue, and there is a need to consider other sample sources as surrogates for biomarker research. Airborne toxicants pass through the nasal epithelium before reaching the lower airways, and the similarity with bronchial histology makes it an attractive surrogate for lower airways. In this work, we describe the transcriptomics findings from the nasal epithelia of subjects enrolled in a clinical study focusing on the identification of COPD biomarkers. Transcriptomic data were analyzed using the biological network approach that enabled us to pinpoint the biological processes affected in the upper respiratory tract in response to smoking and mild-to-moderate COPD. Our results indicated that nasal and lower airway immune responses were considerably different in COPD subjects and caution should be exercised when using upper airway samples as a surrogate for the lower airway. Nevertheless, the network approach described here could present a sensitive means of identifying smokers at risk of developing COPD.
Collapse
Affiliation(s)
- Marja Talikka
- Philip Morris Products SA and Research & Development (R&D), Philip Morris International, Neuchâtel, Switzerland
| | - Florian Martin
- Philip Morris Products SA and Research & Development (R&D), Philip Morris International, Neuchâtel, Switzerland
| | - Alain Sewer
- Philip Morris Products SA and Research & Development (R&D), Philip Morris International, Neuchâtel, Switzerland
| | - Grégory Vuillaume
- Philip Morris Products SA and Research & Development (R&D), Philip Morris International, Neuchâtel, Switzerland
| | - Patrice Leroy
- Philip Morris Products SA and Research & Development (R&D), Philip Morris International, Neuchâtel, Switzerland
| | - Karsta Luettich
- Philip Morris Products SA and Research & Development (R&D), Philip Morris International, Neuchâtel, Switzerland
| | - Nveed Chaudhary
- Philip Morris Products SA and Research & Development (R&D), Philip Morris International, Neuchâtel, Switzerland
| | - Michael J Peck
- Philip Morris Products SA and Research & Development (R&D), Philip Morris International, Neuchâtel, Switzerland
| | - Manuel C Peitsch
- Philip Morris Products SA and Research & Development (R&D), Philip Morris International, Neuchâtel, Switzerland
| | - Julia Hoeng
- Philip Morris Products SA and Research & Development (R&D), Philip Morris International, Neuchâtel, Switzerland
| |
Collapse
|
41
|
Huang CC, Wang CH, Fu CH, Huang CC, Chang PH, Chen YW, Wu CC, Wu PW, Lee TJ. Association between cigarette smoking and interleukin-17A expression in nasal tissues of patients with chronic rhinosinusitis and asthma. Medicine (Baltimore) 2016; 95:e5432. [PMID: 27893686 PMCID: PMC5134879 DOI: 10.1097/md.0000000000005432] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Cigarette smoke plays a substantial role in the development of airway inflammatory diseases, including asthma and chronic rhinosinusitis (CRS). Interleukin (IL)-17A might contribute to cigarette smoke-related inflammation of the airway. This study aimed to investigate the association between cigarette smoking and IL-17A expression in the nasal tissues of patients with CRS and asthma.We prospectively recruited 24 patients (13 smokers, 11 nonsmokers) with CRS and asthma and 6 patients with asthma but without CRS (control group) in a tertiary medical center. Nasal mucosa was obtained as part of the nasal surgery. Protein and mRNA levels of IL-17A in the nasal tissues were determined by immunostaining and real-time polymerase chain reaction.The number of unexpected emergency clinic visits for acute asthma attacks were higher among smokers than among nonsmokers. Interleukin-17A protein and mRNA levels in the nasal tissues of smokers were greater compared to those in the nasal tissues of nonsmokers (P = 0.02 both) and control patients (P = 0.05 and 0.04, respectively).Cigarette smoking was associated with an increase in the number of unexpected emergency clinic visits due to acute asthma attack and in the expression of IL-17A in the nasal tissues of patients with airway inflammatory diseases.
Collapse
Affiliation(s)
- Chien-Chia Huang
- Division of Rhinology, Department of Otolaryngology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University
| | - Chun-Hua Wang
- Department of Thoracic Medicine, Chang Gung Memorial Hospital and Chang Gung Memorial Hospital and Chang Gung University, Taoyuan
| | - Chia-Hsiang Fu
- Division of Rhinology, Department of Otolaryngology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University
| | - Chi-Che Huang
- Division of Rhinology, Department of Otolaryngology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University
| | - Po-Hung Chang
- Division of Rhinology, Department of Otolaryngology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University
| | - Yi-Wei Chen
- Division of Rhinology, Department of Otolaryngology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan
| | - Chia-Chen Wu
- Division of Rhinology, Department of Otolaryngology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan
| | - Pei-Wen Wu
- Division of Rhinology, Department of Otolaryngology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan
- Department of Otolaryngology–Head and Neck Surgery, Chang Gung Memorial Hospital and Chang Gung University, Keelung, Taiwan
| | - Ta-Jen Lee
- Division of Rhinology, Department of Otolaryngology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan
| |
Collapse
|
42
|
Schramm F, Lange M, Hoppmann P, Heutelbeck A. Cytotoxicity of carbon nanohorns in different human cells of the respiratory system. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2016; 79:1085-1093. [PMID: 27924712 DOI: 10.1080/15287394.2016.1219594] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
One of the new synthetic carbon-based nanomaterials is carbon nanohorns (CNH). A potential risk for employees of production processes is an unintentional intake of these nanomaterials via inhalation. Once taken up, nanoparticles might interact with cells of different tissues as well as with intercellular substances. These interactions may have far-reaching consequences for human health. Currently, many gaps in available information on the CNH toxicological profile remain. The aim of this study was to determine the cytotoxicity of CNH particles on human epithelial cells of the respiratory system with special consideration given to different particle sizes. In all cell lines, cell viability was reduced after 24 h of exposure up to 60% and metabolic activity as evidenced by mitochondrial activity was lowered to 9% at a concentration of 1 g/L. The three respiratory cell lines differed in their sensitivity. The most robust cells were the bronchial epithelial cells. Further, particle size fractions induced different adverse effect strength, whereby no correlation between particle size fraction and toxicity was found. These findings demonstrate the need for further information regarding the behavior and effect strength of nanomaterial. To avoid the production of new harmful materials, a more comprehensive integration of results from toxicity studies in the development processes of engineered nanomaterials is recommended not only from an occupational viewpoint but also from an environmental perspective.
Collapse
Affiliation(s)
- Franziska Schramm
- a Department of Occupational, Social and Environmental Medicine , Georg-August-University Göttingen , Göttingen , Germany
| | - Martina Lange
- a Department of Occupational, Social and Environmental Medicine , Georg-August-University Göttingen , Göttingen , Germany
| | - Pia Hoppmann
- a Department of Occupational, Social and Environmental Medicine , Georg-August-University Göttingen , Göttingen , Germany
| | - Astrid Heutelbeck
- a Department of Occupational, Social and Environmental Medicine , Georg-August-University Göttingen , Göttingen , Germany
| |
Collapse
|
43
|
Williams MTS, de Courcey F, Comer D, Elborn JS, Ennis M. Bronchial epithelial cell lines and primary nasal epithelial cells from cystic fibrosis respond differently to cigarette smoke exposure. J Cyst Fibros 2015; 15:467-72. [PMID: 26651594 DOI: 10.1016/j.jcf.2015.11.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Revised: 10/22/2015] [Accepted: 11/03/2015] [Indexed: 12/27/2022]
Abstract
The effects of cigarette smoke extract (CSE) on airway epithelial cells (AECs) from cystic fibrosis (CF) and non-cystic fibrosis (non-CF) individuals are not fully understood. It has been suggested that CSE modulates inflammatory cytokine release from AECs by modulating the epidermal growth factor receptor (EGFR) pathway; these pathways could reveal novel therapeutic targets. We compared the effect of CSE pre-incubation on IL-8 release from CF and non-CF bronchial epithelial cell lines, and separately, with primary nasal epithelial cells (NECs) retrieved from CF and non-CF individuals. We also determined if the EGFR pathway regulates IL-8 release by LPS or cytomix in non-CF and CF AECs at baseline and following CSE exposure. CF and non-CF cell lines, NECs derived from both CF patients (R117H heterozygous and F508del homozygous), and from healthy subjects, were cultured in the presence or absence of CSE, and subsequently exposed to inflammatory stimuli. In cell lines CSE significantly reduced IL-8 release following inflammatory challenge. Conversely, CSE pre-treatment was pro-inflammatory in primary NECs. In NECs from control subjects, CSE increased cytomix and LPS induced IL-8 release, and for the R117H heterozygous NEC cultures, CSE enhanced basal IL-8 release. Cytomix and LPS induced IL-8 release from F508del homozygous NEC cultures was further heightened following CSE pre-treatment. EGFR inhibition mitigated IL-8 release from immortalised and primary non-CF and CF AECs, suggesting that constitutive and CSE elicited IL-8 release from AECs is partly regulated via the EGFR pathway. This study demonstrates the importance of the EGFR cascade in the regulation of constitutive and CSE induced inflammatory mediator release from immortalised and primary AECs. Moreover, it clearly highlights the significance of using primary cells to confirm results obtained from immortalised cell studies, as these model systems may respond very differently to the stimuli under investigation.
Collapse
Affiliation(s)
- Mark Thomas Shaw Williams
- Centre for Infection and Immunity, Queen's University Belfast, Belfast BT9 7AE, Northern Ireland, United Kingdom; Institute for Infection, Immunity and Inflammation, University of Glasgow, Glasgow, G12 8TA Scotland, United Kingdom.
| | - Francine de Courcey
- Centre for Infection and Immunity, Queen's University Belfast, Belfast BT9 7AE, Northern Ireland, United Kingdom
| | - David Comer
- Centre for Infection and Immunity, Queen's University Belfast, Belfast BT9 7AE, Northern Ireland, United Kingdom
| | - Joseph S Elborn
- Centre for Infection and Immunity, Queen's University Belfast, Belfast BT9 7AE, Northern Ireland, United Kingdom
| | - Madeleine Ennis
- Centre for Infection and Immunity, Queen's University Belfast, Belfast BT9 7AE, Northern Ireland, United Kingdom
| |
Collapse
|
44
|
Alves MP, Schögler A, Ebener S, Vielle NJ, Casaulta C, Jung A, Moeller A, Geiser T, Regamey N. Comparison of innate immune responses towards rhinovirus infection of primary nasal and bronchial epithelial cells. Respirology 2015; 21:304-12. [DOI: 10.1111/resp.12692] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 07/14/2015] [Accepted: 08/26/2015] [Indexed: 12/27/2022]
Affiliation(s)
- Marco P. Alves
- Department of Clinical ResearchUniversity Hospital of Bern Bern Switzerland
- Division of Pediatric Respiratory MedicineUniversity Children's Hospital Bern Switzerland
| | - Aline Schögler
- Department of Clinical ResearchUniversity Hospital of Bern Bern Switzerland
- Graduate School for Cellular and Biomedical SciencesUniversity Hospital of Bern Bern Switzerland
- Division of Pediatric Respiratory MedicineUniversity Children's Hospital Bern Switzerland
| | - Simone Ebener
- Department of Clinical ResearchUniversity Hospital of Bern Bern Switzerland
- Graduate School for Cellular and Biomedical SciencesUniversity Hospital of Bern Bern Switzerland
- Division of Pediatric Respiratory MedicineUniversity Children's Hospital Bern Switzerland
| | - Nathalie J. Vielle
- Department of Clinical ResearchUniversity Hospital of Bern Bern Switzerland
- Division of Pediatric Respiratory MedicineUniversity Children's Hospital Bern Switzerland
| | - Carmen Casaulta
- Division of Pediatric Respiratory MedicineUniversity Children's Hospital Bern Switzerland
| | - Andreas Jung
- Division of Respiratory MedicineUniversity Children's Hospital Zürich Switzerland
| | - Alexander Moeller
- Division of Respiratory MedicineUniversity Children's Hospital Zürich Switzerland
| | - Thomas Geiser
- Department of Clinical ResearchUniversity Hospital of Bern Bern Switzerland
- Division of Respiratory MedicineUniversity Hospital of Bern Bern Switzerland
| | - Nicolas Regamey
- Department of Clinical ResearchUniversity Hospital of Bern Bern Switzerland
- Children's Hospital Lucerne Lucerne Switzerland
| |
Collapse
|
45
|
Hox V, Maes T, Huvenne W, Van Drunen C, Vanoirbeek JA, Joos G, Bachert C, Fokkens W, Ceuppens JL, Nemery B, Hellings PW. A chest physician's guide to mechanisms of sinonasal disease. Thorax 2015; 70:353-8. [DOI: 10.1136/thoraxjnl-2014-205520] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
46
|
Hawley B, Schaeffer J, Poole JA, Dooley GP, Reynolds S, Volckens J. Differential response of human nasal and bronchial epithelial cells upon exposure to size-fractionated dairy dust. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2015; 78:583-94. [PMID: 25965193 PMCID: PMC4430335 DOI: 10.1080/15287394.2015.1015699] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Exposure to organic dusts is associated with increased respiratory morbidity and mortality in agricultural workers. Organic dusts in dairy farm environments are complex, polydisperse mixtures of toxic and immunogenic compounds. Previous toxicological studies focused primarily on exposures to the respirable size fraction; however, organic dusts in dairy farm environments are known to contain larger particles. Given the size distribution of dusts from dairy farm environments, the nasal and bronchial epithelia represent targets of agricultural dust exposures. In this study, well-differentiated normal human bronchial epithelial cells and human nasal epithelial cells were exposed to two different size fractions (PM10 and PM>10) of dairy parlor dust using a novel aerosol-to-cell exposure system. Levels of proinflammatory transcripts (interleukin [IL]-8, IL-6, and tumor necrosis factor [TNF]-α) were measured 2 h after exposure. Lactate dehydrogenase (LDH) release was also measured as an indicator of cytotoxicity. Cell exposure to dust was measured in each size fraction as a function of mass, endotoxin, and muramic acid levels. To our knowledge, this is the first study to evaluate the effects of distinct size fractions of agricultural dust on human airway epithelial cells. Our results suggest that both PM10 and PM>10 size fractions elicit a proinflammatory response in airway epithelial cells and that the entire inhalable size fraction needs to be considered when assessing potential risks from exposure to agricultural dusts. Further, data suggest that human bronchial cells respond differently to these dusts than human nasal cells, and therefore that the two cell types need to be considered separately in airway cell models of agricultural dust toxicity.
Collapse
Affiliation(s)
- Brie Hawley
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA 80523
| | - Joshua Schaeffer
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA 80523
- High Plains Intermountain Center for Agricultural Health and Safety; 154B EHB Department of Environmental and Radiological Health Sciences, Colorado State University, Ft. Collins, CO 80523-1681
| | - Jill A. Poole
- High Plains Intermountain Center for Agricultural Health and Safety; 154B EHB Department of Environmental and Radiological Health Sciences, Colorado State University, Ft. Collins, CO 80523-1681
- Pulmonary, Critical Care, Sleep & Allergy Division; Department of Internal Medicine, University of Nebraska Medical Center, 985990 The Nebraska Medical Center, Omaha, NE 68198-5990
| | - Gregory P. Dooley
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA 80523
| | - Stephen Reynolds
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA 80523
- High Plains Intermountain Center for Agricultural Health and Safety; 154B EHB Department of Environmental and Radiological Health Sciences, Colorado State University, Ft. Collins, CO 80523-1681
| | - John Volckens
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA 80523
- Department of Mechanical Engineering, Colorado State University, Fort Collins, CO, USA 80523
| |
Collapse
|
47
|
Stokes AB, Kieninger E, Schögler A, Kopf BS, Casaulta C, Geiser T, Regamey N, Alves MP. Comparison of three different brushing techniques to isolate and culture primary nasal epithelial cells from human subjects. Exp Lung Res 2014; 40:327-32. [PMID: 25058379 DOI: 10.3109/01902148.2014.925987] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
PURPOSE Primary nasal epithelial cells are used for diagnostic purposes in clinical routine and have been shown to be good surrogate models for bronchial epithelial cells in studies of airway inflammation and remodeling. We aimed at comparing different instruments allowing isolation of nasal epithelial cells. METHODS Primary airway epithelial cell cultures were established using cells acquired from the inferior surface of the middle turbinate of both nostrils. Three different instruments to isolate nasal cells were used: homemade cytology brush, nasal swab, and curette. Cell count, viability, time until a confluent cell layer was reached, and success rate in establishing cell cultures were evaluated. A standard numeric pain intensity scale was used to assess the acceptability of each instrument. RESULTS Sixty healthy adults (median with interquartile range [IQR] age of 31 [26-37] years) participated in the study. Higher number of cells (×10(5) cells/ml) was obtained using brushes (9.8 [5.9-33.5]) compared to swabs (2.4 [1.5-3.9], p < 0.0001) and curettes (5.5 [4.4-6.9], p < 0.01). Cell viability was similar between groups. Cells obtained by brushes had the fastest growth rate, and the success rate in establishing primary cell cultures was highest with brushes (90% vs. 65% for swabs and 70% for curettes). Pain was highest with curettes (VAS score 4.0 [3.0-5.0] out of 10). The epithelial phenotype of the cultures was confirmed through cytokeratin and E-cadherin staining. CONCLUSIONS All three types of instruments allow collection and growth of human nasal epithelial cells with good acceptability to study participants. The most efficient instrument is the nasal brush.
Collapse
Affiliation(s)
- Andrea B Stokes
- 1Division of Paediatric Respiratory Medicine, University Children's Hospital , Bern , Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Bardet G, Achard S, Loret T, Desauziers V, Momas I, Seta N. A model of human nasal epithelial cells adapted for direct and repeated exposure to airborne pollutants. Toxicol Lett 2014; 229:144-9. [PMID: 24960057 DOI: 10.1016/j.toxlet.2014.05.023] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 05/04/2014] [Accepted: 05/27/2014] [Indexed: 12/28/2022]
Abstract
Airway epithelium lining the nasal cavity plays a pivotal role in respiratory tract defense and protection mechanisms. Air pollution induces alterations linked to airway diseases such as asthma. Only very few in vitro studies to date have succeeded in reproducing physiological conditions relevant to cellular type and chronic atmospheric pollution exposure. We therefore, set up an in vitro model of human Airway Epithelial Cells of Nasal origin (hAECN) close to real human cell functionality, specifically adapted to study the biological effects of exposure to indoor gaseous pollution at the environmental level. hAECN were exposed under air-liquid interface, one, two, or three-times at 24 h intervals for 1 h, to air or formaldehyde (200 μg/m(3)), an indoor air gaseous pollutant. All experiments were ended at day 4, when both cellular viability and cytokine production were assessed. Optimal adherence and confluence of cells were obtained 96 h after cell seeding onto collagen IV-precoated insert. Direct and repeated exposure to formaldehyde did not produce any cellular damage or IL-6 production change, although weak lower IL-8 production was observed only after the third exposure. Our model is significantly better than previous ones due to cell type and the repeated exposure protocol.
Collapse
Affiliation(s)
- Gaëlle Bardet
- Université Paris Descartes, EA 4064, Laboratoire de Santé Publique et Environnement, 4, Avenue de l'Observatoire, 75006 Paris, France; Agence de l'Environnement et de la Maîtrise de l'Energie, Angers, France.
| | - Sophie Achard
- Université Paris Descartes, EA 4064, Laboratoire de Santé Publique et Environnement, 4, Avenue de l'Observatoire, 75006 Paris, France.
| | - Thomas Loret
- Université Paris Descartes, EA 4064, Laboratoire de Santé Publique et Environnement, 4, Avenue de l'Observatoire, 75006 Paris, France.
| | - Valérie Desauziers
- Centre des Matériaux des Mines d'Alès, Ecole des Mines d'Alès, Pau, France.
| | - Isabelle Momas
- Université Paris Descartes, EA 4064, Laboratoire de Santé Publique et Environnement, 4, Avenue de l'Observatoire, 75006 Paris, France.
| | - Nathalie Seta
- Université Paris Descartes, EA 4064, Laboratoire de Santé Publique et Environnement, 4, Avenue de l'Observatoire, 75006 Paris, France; AP-HP, Hôpital Bichat, Biochimie, Paris, France.
| |
Collapse
|
49
|
Calzetta L, Rogliani P, Cazzola M, Matera MG. Advances in asthma drug discovery: evaluating the potential of nasal cell sampling and beyond. Expert Opin Drug Discov 2014; 9:595-607. [PMID: 24749518 DOI: 10.1517/17460441.2014.909403] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Inhaled corticosteroid anti-inflammatory therapy is effective at controlling disease symptoms of asthma, but a subset of patients remains symptomatic despite optimal treatment, creating a clear unmet medical need. Moreover, none of the currently available drugs for asthma are really disease-modifying or curative. Although murine models of asthma, based on transgenic and knockout animals, may offer an integrated pathophysiological system for studying the characteristics of airway inflammation and hyperresponsiveness, these alterations are noteworthily different compared with those observed in asthmatic patients. Since a clear functional and inflammatory relationship between the nasal mucosa and bronchial tissue in patients suffering from asthma and allergic rhinitis has been recognized, using preclinical models based on human nasal cells sampling might support a prompt and effective anti-inflammatory drug discovery in asthma. AREAS COVERED The authors provide a review, which discusses the potential role of nasal cell sampling and its application in advanced drug discovery for asthma. The contents range from the similarities and differences between asthma and allergic rhinitis up to artificial airway models based on sophisticated human lung-on-a-chip devices. EXPERT OPINION Nasal cell sampling and processing have reached a great potential in asthma drug discovery. The authors believe that models of asthma, which are based on human nasal cells, can provide valuable indications of proof of pharmacological and potential therapeutic efficacy in both preclinical and early clinical settings.
Collapse
Affiliation(s)
- Luigino Calzetta
- IRCCS, San Raffaele Pisana Hospital, Department of Pulmonary Rehabilitation , Rome , Italy
| | | | | | | |
Collapse
|
50
|
Comer DM, Elborn JS, Ennis M. Inflammatory and cytotoxic effects of acrolein, nicotine, acetylaldehyde and cigarette smoke extract on human nasal epithelial cells. BMC Pulm Med 2014; 14:32. [PMID: 24581246 PMCID: PMC3945717 DOI: 10.1186/1471-2466-14-32] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Accepted: 02/25/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Cigarette smoke induces a pro-inflammatory response in airway epithelial cells but it is not clear which of the various chemicals contained within cigarette smoke (CS) should be regarded as predominantly responsible for these effects. We hypothesised that acrolein, nicotine and acetylaldehyde, important chemicals contained within volatile cigarette smoke in terms of inducing inflammation and causing addiction, have immunomodulatory effects in primary nasal epithelial cell cultures (PNECs). METHODS PNECs from 19 healthy subjects were grown in submerged cultures and were incubated with acrolein, nicotine or acetylaldehyde prior to stimulation with Pseudomonas aeruginosa lipopolysaccharide (PA LPS). Experiments were repeated using cigarette smoke extract (CSE) for comparison. IL-8 was measured by ELISA, activation of NF-κB by ELISA and Western blotting, and caspase-3 activity by Western blotting. Apoptosis was evaluated using Annexin-V staining and the terminal transferase-mediated dUTP nick end-labeling (TUNEL) method. RESULTS CSE was pro-inflammatory after a 24 h exposure and 42% of cells were apoptotic or necrotic after this exposure time. Acrolein was pro-inflammatory for the PNEC cultures (30 μM exposure for 4 h inducing a 2.0 fold increase in IL-8 release) and also increased IL-8 release after stimulation with PA LPS. In contrast, nicotine had anti-inflammatory properties (0.6 fold IL-8 release after 50 μM exposure to nicotine for 24 h), and acetylaldehyde was without effect. Acrolein and nicotine had cellular stimulatory and anti-inflammatory effects respectively, as determined by NF-κB activation. Both chemicals increased levels of cleaved caspase 3 and induced cell death. CONCLUSIONS Acrolein is pro-inflammatory and nicotine anti-inflammatory in PNEC cultures. CSE induces cell death predominantly by apoptotic mechanisms.
Collapse
Affiliation(s)
- David M Comer
- Centre for Infection and Immunity, Health Sciences Building, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University of Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
- Respiratory Department, Belfast City Hospital, Lisburn Road, Belfast BT9 7AB, UK
| | - Joseph Stuart Elborn
- Centre for Infection and Immunity, Health Sciences Building, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University of Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
- Respiratory Department, Belfast City Hospital, Lisburn Road, Belfast BT9 7AB, UK
| | - Madeleine Ennis
- Centre for Infection and Immunity, Health Sciences Building, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University of Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| |
Collapse
|