1
|
Beilleau G, Stalder H, Almeida L, Oliveira Esteves BI, Alves MP, Schweizer M. The Pestivirus RNase E rns Tames the Interferon Response of the Respiratory Epithelium. Viruses 2024; 16:1908. [PMID: 39772215 PMCID: PMC11680131 DOI: 10.3390/v16121908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/06/2024] [Accepted: 12/08/2024] [Indexed: 01/11/2025] Open
Abstract
Bovine viral diarrhea virus (BVDV), a pestivirus in the family Flaviviridae, is a major livestock pathogen. Horizontal transmission leads to acute transient infections via the oronasal route, whereas vertical transmission might lead to the birth of immunotolerant, persistently infected animals. In both cases, BVDV exerts an immunosuppressive effect, predisposing infected animals to secondary infections. Erns, an immunomodulatory viral protein, is present on the envelope of the virus and is released as a soluble protein. In this form, it is taken up by cells and, with its RNase activity, degrades single- and double-stranded (ds) RNA, thus preventing activation of the host's interferon system. Here, we show that Erns of the pestiviruses BVDV and Bungowannah virus effectively inhibit dsRNA-induced IFN synthesis in well-differentiated airway epithelial cells cultured at the air-liquid interface. This activity was observed independently of the side of entry, apical or basolateral, of the pseudostratified, polarized cell layer. Virus infection was successful from both surfaces but was inefficient, requiring several days of incubation. Virus release was almost exclusively restricted to the apical side. This confirms that primary, well-differentiated respiratory epithelial cells cultured at the air-liquid interface are an appropriate model to study viral infection and innate immunotolerance in the bovine respiratory tract. Furthermore, evidence is presented that Erns might contribute to the immunosuppressive effect observed after BVDV infections, especially in persistently infected animals.
Collapse
Affiliation(s)
- Guillaume Beilleau
- Institute of Virology and Immunology, Länggass-Str. 122, CH-3001 Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, CH-3001 Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, CH-3012 Bern, Switzerland
| | - Hanspeter Stalder
- Institute of Virology and Immunology, Länggass-Str. 122, CH-3001 Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, CH-3001 Bern, Switzerland
| | - Lea Almeida
- Institute of Virology and Immunology, Länggass-Str. 122, CH-3001 Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, CH-3001 Bern, Switzerland
| | - Blandina I. Oliveira Esteves
- Institute of Virology and Immunology, Länggass-Str. 122, CH-3001 Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, CH-3001 Bern, Switzerland
| | - Marco P. Alves
- Institute of Virology and Immunology, Länggass-Str. 122, CH-3001 Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, CH-3001 Bern, Switzerland
- Multidisciplinary Center for Infectious Diseases, University of Bern, CH-3012 Bern, Switzerland
| | - Matthias Schweizer
- Institute of Virology and Immunology, Länggass-Str. 122, CH-3001 Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, CH-3001 Bern, Switzerland
| |
Collapse
|
2
|
Sekiya T, Murakami K, Isohama Y. Seihaito, a Kampo medicine, attenuates IL-13-induced mucus production and goblet cell metaplasia. J Pharmacol Sci 2024; 155:21-28. [PMID: 38677782 DOI: 10.1016/j.jphs.2024.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 02/02/2024] [Accepted: 02/13/2024] [Indexed: 04/29/2024] Open
Abstract
Goblet cell hyperplasia and increased mucus production are features of airway diseases, including asthma, and excess airway mucus often worsens these conditions. Even steroids are not uniformly effective in mucus production in severe asthma, and new therapeutic options are needed. Seihaito is a Japanese traditional medicine that is used clinically as an antitussive and expectorant. In the present study, we examined the effect of Seihaito on goblet cell differentiation and mucus production. In in vitro studies, using air-liquid interface culture of guinea-pig tracheal epithelial cells, Seihaito inhibited IL-13-induced proliferation of goblet cells and MUC5AC, a major component of mucus production. Seihaito suppressed goblet cell-specific gene expression, without changing ciliary cell-specific genes, suggesting that it inhibits goblet cell differentiation. In addition, Seihaito suppressed MUC5AC expression in cells transfected with SPDEF, a transcription factor activated by IL-13. Furthermore, Seihaito attenuated in vivo goblet cell proliferation and MUC5AC mRNA expression in IL-13-treated mouse lungs. Collectively, these findings demonstrated that Seihaito has an inhibitory effect on goblet cell differentiation and mucus production, which is at least partly due to the inhibition of SPDEF.
Collapse
Affiliation(s)
- Tomoki Sekiya
- Laboratory of Applied Pharmacology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| | - Kazuhito Murakami
- Laboratory of Applied Pharmacology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| | - Yoichiro Isohama
- Laboratory of Applied Pharmacology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan.
| |
Collapse
|
3
|
Romanova OA, Klein OI, Sytina EV, Rudyak SG, Patsaev TD, Tenchurin TH, Grigorchuk AY, Demina TS, Chvalun SN, Panteleyev AA. Fibroblasts and polymer composition are essential for bioengineering of airway epithelium on nonwoven scaffolds. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2024; 35:851-868. [PMID: 38310545 DOI: 10.1080/09205063.2024.2310370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 12/19/2024] [Indexed: 02/06/2024]
Abstract
To make tissue engineering a truly effective tool, it is necessary to understand how the patterns of specific tissue development are modulated by and depend on the artificial environment. Even the most advanced approaches still do not fully meet the requirements of practical engineering of tracheobronchial epithelium. This study aimed to test the ability of the synthetic and natural nonwoven scaffolds to support the formation of morphological sound airway epithelium including the basement membrane (BM). We also sought to identify the potential role of fibroblasts in this process. Our results showed that nonwoven scaffolds are generally suitable for producing well-differentiated tracheobronchial epithelium (with cilia and goblet cells), while the structure and functionality of the equivalents appeared to be highly dependent on the composition of the scaffolds. Unlike natural scaffolds, synthetic ones supported the formation of the epithelium only when epithelial cells were cocultured with fibroblasts. Fibroblasts also appeared to be obligatory for basal lamina formation, regardless of the type of the nonwoven material used. However, even in the presence of fibroblasts, the synthetic scaffolds were unable to support the formation of the epithelium and of the BM (in particular, basal lamina) as effectively as the natural scaffolds did.
Collapse
Affiliation(s)
| | - Olga I Klein
- NRC Kurchatov Institute, Moscow, Russian Federation
- The Federal Research Center "Fundamentals of Biotechnology" of the Russian Academy of Sciences, Bach Institute of Biochemistry
| | | | - Stanislav G Rudyak
- Pirogov Russian National Research Medical University, Moscow, Russian Federation
| | | | | | | | - Tatiana S Demina
- Enikolopov Institute of Synthetic Polymeric Materials, Russian Academy of Sciences, Moscow, Russian Federation
| | - Sergey N Chvalun
- NRC Kurchatov Institute, Moscow, Russian Federation
- Enikolopov Institute of Synthetic Polymeric Materials, Russian Academy of Sciences, Moscow, Russian Federation
| | - Andrey A Panteleyev
- NRC Kurchatov Institute, Moscow, Russian Federation
- A.V. Vishnevsky Institute of Surgery, Moscow, Russian Federation
| |
Collapse
|
4
|
Gandhi NN, Inzana TJ, Rajagopalan P. Bovine Airway Models: Approaches for Investigating Bovine Respiratory Disease. ACS Infect Dis 2023; 9:1168-1179. [PMID: 37257116 DOI: 10.1021/acsinfecdis.2c00618] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Bovine respiratory disease (BRD) is a multifactorial condition where different genera of bacteria, such as Mannheimia haemolytica, Histophilus somni, Pasteurella multocida, and Mycoplasma bovis, and viruses, like bovine respiratory syncytial virus, bovine viral diarrhea virus, and bovine herpes virus-1, infect the lower respiratory tract of cattle. These pathogens can co-infect cells in the respiratory system, thereby making specific treatment very difficult. Currently, the most common models for studying BRD include a submerged tissue culture (STC), where monolayers of epithelial cells are typically covered either in cellular or spent biofilm culture medium. Another model is an air-liquid interface (ALI), where epithelial cells are exposed on their apical side and allowed to differentiate. However, limited work has been reported on the study of three-dimensional (3D) bovine models that incorporate multiple cell types to represent the architecture of the respiratory tract. The roles of different defense mechanisms in an infected bovine respiratory system, such as mucin production, tight junction barriers, and the production of antimicrobial peptides in in vitro cultures require further investigation in order to provide a comprehensive understanding of the disease pathogenesis. In this report, we describe the different aspects of BRD, including the most implicated pathogens and the respiratory tract, which are important to incorporate in disease models assembled in vitro. Although current advancements of bovine respiratory cultures have led to knowledge of the disease, 3D multicellular organoids that better recapitulate the in vivo environment exhibit potential for future investigations.
Collapse
Affiliation(s)
- Neeti N Gandhi
- Department of Chemical Engineering, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Thomas J Inzana
- College of Veterinary Medicine, Long Island University, Brookville, New York 11548, United States
| | - Padmavathy Rajagopalan
- Department of Chemical Engineering, Virginia Tech, Blacksburg, Virginia 24061, United States
| |
Collapse
|
5
|
In Vitro Characteristics of Canine Primary Tracheal Epithelial Cells Maintained at an Air-Liquid Interface Compared to In Vivo Morphology. Int J Mol Sci 2023; 24:ijms24054987. [PMID: 36902418 PMCID: PMC10003254 DOI: 10.3390/ijms24054987] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/28/2023] [Accepted: 03/03/2023] [Indexed: 03/08/2023] Open
Abstract
Culturing respiratory epithelial cells at an air-liquid interface (ALI) represents an established method for studies on infection or toxicology by the generation of an in vivo-like respiratory tract epithelial cellular layer. Although primary respiratory cells from a variety of animals have been cultured, an in-depth characterization of canine tracheal ALI cultures is lacking despite the fact that canines are a highly relevant animal species susceptible to various respiratory agents, including zoonotic pathogens such as severe acute respiratory coronavirus 2 (SARS-CoV-2). In this study, canine primary tracheal epithelial cells were cultured under ALI conditions for four weeks, and their development was characterized during the entire culture period. Light and electron microscopy were performed to evaluate cell morphology in correlation with the immunohistological expression profile. The formation of tight junctions was confirmed using transepithelial electrical resistance (TEER) measurements and immunofluorescence staining for the junctional protein ZO-1. After 21 days of culture at the ALI, a columnar epithelium containing basal, ciliated and goblet cells was seen, resembling native canine tracheal samples. However, cilia formation, goblet cell distribution and epithelial thickness differed significantly from the native tissue. Despite this limitation, tracheal ALI cultures could be used to investigate the pathomorphological interactions of canine respiratory diseases and zoonotic agents.
Collapse
|
6
|
Lee DF, Thompson CL, Baynes RE, Enomoto H, Smith GW, Chambers MA. Development and evaluation of a bovine lung-on-chip (bLOC) to study bovine respiratory diseases. IN VITRO MODELS 2022; 1:333-346. [PMID: 36660607 PMCID: PMC9383688 DOI: 10.1007/s44164-022-00030-z] [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] [Received: 04/29/2022] [Revised: 07/25/2022] [Accepted: 08/01/2022] [Indexed: 01/22/2023]
Abstract
Purpose Current air-liquid interface (ALI) models of bovine proximal airways have their limitations. They do not simulate blood flow necessary to mimic systemic drug administration, and repeated sampling requires multiple, independent cultures. A bovine lung-on-chip (bLOC) would overcome these limitations, providing a convenient and cost-effective model for pharmacokinetic or pathogenicity studies. Methods Bovine pulmonary arterial endothelial cells seeded into the endothelial channel of an Emulate Lung-Chip were interfaced with bovine bronchial epithelial cells in the epithelial channel. Cells were cultured at ALI for up to 21 days. Differentiation was assessed by mucin quantification, phase-contrast light microscopy and immunofluorescence of cell-specific markers in fixed cultures. Barrier integrity was determined by FITC-labelled dextran 3-5 kDa permeability. To evaluate the model, endothelial-epithelial transport of the antibiotic drug, danofloxacin, was followed using liquid chromatography-mass spectrometry, with the aim of replicating data previously determined in vivo. Results bLOC cultures secreted quantifiable mucins, whilst cilia formation was evident in the epithelial channel. Barrier integrity of the model was demonstrated by resistance to FITC-Dextran 3-5 kDa permeation. Bronchial epithelial and endothelial cell-specific markers were observed. Close to plasma, representative PK data for danofloxacin was observed in the endothelial channel; however, danofloxacin in the epithelial channel was mostly below the limit of quantification. Conclusion A co-culture model of the bovine proximal airway was successfully generated, with potential to replace in vivo experimentation. With further optimisation and characterisation, the bLOC may be suitable to perform drug pharmacokinetic studies for bovine respiratory disease (BRD), and other applications.
Collapse
Affiliation(s)
- Diane F. Lee
- School of Veterinary Medicine, University of Surrey, Guildford, UK
- Now at Sussex Drug Discovery Centre, University of Sussex, Falmer, UK
| | - Clare L. Thompson
- Centre for Predictive In Vitro Models, School of Engineering and Materials Science, Queen Mary University of London, London, UK
| | - Ronald E. Baynes
- College of Veterinary Medicine, North Carolina State University, Raleigh, NC USA
| | - Hiroko Enomoto
- College of Veterinary Medicine, North Carolina State University, Raleigh, NC USA
| | | | - Mark A. Chambers
- School of Veterinary Medicine, University of Surrey, Guildford, UK
| |
Collapse
|
7
|
Luengen AE, Cheremkhina M, Gonzalez-Rubio J, Weckauf J, Kniebs C, Uebner H, Buhl EM, Taube C, Cornelissen CG, Schmitz-Rode T, Jockenhoevel S, Thiebes AL. Bone Marrow Derived Mesenchymal Stromal Cells Promote Vascularization and Ciliation in Airway Mucosa Tri-Culture Models in Vitro. Front Bioeng Biotechnol 2022; 10:872275. [PMID: 35782511 PMCID: PMC9247357 DOI: 10.3389/fbioe.2022.872275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 05/18/2022] [Indexed: 11/13/2022] Open
Abstract
Patients suffering from irresectable tracheal stenosis often face limited treatment options associated with low quality of life. To date, an optimal tracheal replacement strategy does not exist. A tissue-engineered tracheal substitute promises to overcome limitations such as implant vascularization, functional mucociliary clearance and mechanical stability. In order to advance a tracheal mucosa model recently developed by our group, we examined different supporting cell types in fibrin-based tri-culture with primary human umbilical vein endothelial cells (HUVEC) and primary human respiratory epithelial cells (HRE). Bone marrow-derived mesenchymal stromal cells (BM-MSC), adipose-derived mesenchymal stromal cells (ASC) and human nasal fibroblasts (HNF) were compared regarding their ability to promote mucociliary differentiation and vascularization in vitro. Three-dimensional co-cultures of the supporting cell types with either HRE or HUVEC were used as controls. Mucociliary differentiation and formation of vascular-like structures were analyzed by scanning electron microscopy (SEM), periodic acid Schiff’s reaction (PAS reaction), two-photon laser scanning microscopy (TPLSM) and immunohistochemistry. Cytokine levels of vascular endothelial growth factor (VEGF), epidermal growth factor (EGF), interleukin-6 (IL6), interleukin-8 (IL8), angiopoietin 1, angiopoietin 2, fibroblast growth factor basic (FGF-b) and placenta growth factor (PIGF) in media supernatant were investigated using LEGENDplex™ bead-based immunoassay. Epithelial morphology of tri-cultures with BM-MSC most closely resembled native respiratory epithelium with respect to ciliation, mucus production as well as expression and localization of epithelial cell markers pan-cytokeratin, claudin-1, α-tubulin and mucin5AC. This was followed by tri-cultures with HNF, while ASC-supported tri-cultures lacked mucociliary differentiation. For all supporting cell types, a reduced ciliation was observed in tri-cultures compared to the corresponding co-cultures. Although formation of vascular-like structures was confirmed in all cultures, vascular networks in BM-MSC-tri-cultures were found to be more branched and extended. Concentrations of pro-angiogenic and inflammatory cytokines, in particular VEGF and angiopoietin 2, revealed to be reduced in tri-cultures compared to co-cultures. With these results, our study provides an important step towards a vascularized and ciliated tissue-engineered tracheal replacement. Additionally, our tri-culture model may in the future contribute to an improved understanding of cell-cell interactions in diseases associated with impaired mucosal function.
Collapse
Affiliation(s)
- Anja E. Luengen
- Department of Biohybrid and Medical Textiles (BioTex), AME - Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Aachen, Germany
- Aachen-Maastricht Institute for Biobased Materials, Faculty of Science and Engineering, Maastricht University, Brightlands Chemelot Campus, Geleen, Netherlands
| | - Maria Cheremkhina
- Department of Biohybrid and Medical Textiles (BioTex), AME - Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Aachen, Germany
- Aachen-Maastricht Institute for Biobased Materials, Faculty of Science and Engineering, Maastricht University, Brightlands Chemelot Campus, Geleen, Netherlands
| | - Julian Gonzalez-Rubio
- Department of Biohybrid and Medical Textiles (BioTex), AME - Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Aachen, Germany
- Aachen-Maastricht Institute for Biobased Materials, Faculty of Science and Engineering, Maastricht University, Brightlands Chemelot Campus, Geleen, Netherlands
| | - Jan Weckauf
- Department of Biohybrid and Medical Textiles (BioTex), AME - Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Aachen, Germany
- Aachen-Maastricht Institute for Biobased Materials, Faculty of Science and Engineering, Maastricht University, Brightlands Chemelot Campus, Geleen, Netherlands
| | - Caroline Kniebs
- Department of Biohybrid and Medical Textiles (BioTex), AME - Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Aachen, Germany
- Aachen-Maastricht Institute for Biobased Materials, Faculty of Science and Engineering, Maastricht University, Brightlands Chemelot Campus, Geleen, Netherlands
| | - Hendrik Uebner
- Department of Pulmonary Medicine, University Medical Center Essen—Ruhrlandklinik, Essen, Germany
| | - E. Miriam Buhl
- Institute of Pathology, Electron Microscopy Facility, RWTH Aachen University Hospital, Aachen, Germany
| | - Christian Taube
- Department of Pulmonary Medicine, University Medical Center Essen—Ruhrlandklinik, Essen, Germany
| | - Christian G. Cornelissen
- Department of Biohybrid and Medical Textiles (BioTex), AME - Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Aachen, Germany
- Clinic for Pneumology and Internal Intensive Care Medicine (Medical Clinic V), RWTH Aachen University Hospital, Aachen, Germany
| | - Thomas Schmitz-Rode
- Department of Biohybrid and Medical Textiles (BioTex), AME - Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Aachen, Germany
| | - Stefan Jockenhoevel
- Department of Biohybrid and Medical Textiles (BioTex), AME - Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Aachen, Germany
- Aachen-Maastricht Institute for Biobased Materials, Faculty of Science and Engineering, Maastricht University, Brightlands Chemelot Campus, Geleen, Netherlands
- *Correspondence: Stefan Jockenhoevel, ; Anja Lena Thiebes,
| | - Anja Lena Thiebes
- Department of Biohybrid and Medical Textiles (BioTex), AME - Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Aachen, Germany
- Aachen-Maastricht Institute for Biobased Materials, Faculty of Science and Engineering, Maastricht University, Brightlands Chemelot Campus, Geleen, Netherlands
- *Correspondence: Stefan Jockenhoevel, ; Anja Lena Thiebes,
| |
Collapse
|
8
|
Jung O, Tung YT, Sim E, Chen YC, Lee E, Ferrer M, Song MJ. Development of human-derived, three-dimensional respiratory epithelial tissue constructs with perfusable microvasculature on a high-throughput microfluidics screening platform. Biofabrication 2022; 14. [PMID: 35166694 PMCID: PMC10053540 DOI: 10.1088/1758-5090/ac32a5] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 10/22/2021] [Indexed: 11/12/2022]
Abstract
The COVID-19 pandemic has highlighted the need for human respiratory tract-based assay platforms for efficient discovery and development of antivirals and disease-modulating therapeutics. Physiologically relevant tissue models of the lower respiratory tract (LRT), including the respiratory bronchioles and the alveolar sacs, are of high interest because they are the primary site of severe SARS-CoV-2 infection and are most affected during the terminal stage of COVID-19. Current epithelial lung models used to study respiratory viral infections include lung epithelial cells at the air-liquid interface (ALI) with fibroblasts and endothelial cells, but such models do not have a perfusable microvascular network to investigate both viral infectivity and viral infection-induced thrombotic events. Using a high throughput, 64-chip microfluidic plate-based platform, we have developed two novel vascularized, LRT multi-chip models for the alveoli and the small airway. Both models include a perfusable microvascular network consisting of human primary microvascular endothelial cells, fibroblasts and pericytes. The established biofabrication protocols also enable the formation of differentiated lung epithelial layers at the ALI on top of the vascularized tissue bed. We validated the physiologically relevant cellular composition, architecture and perfusion of the vascularized lung tissue models using fluorescence microscopy, flow cytometry, and electrical resistance measurements. These vascularized, perfusable microfluidic lung tissue on high throughput assay platforms will enable the development of respiratory viral infection and disease models for research investigation and drug discovery.
Collapse
Affiliation(s)
- Olive Jung
- 3D Tissue Bioprinting Laboratory, Department of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, United States of America.,Biomedical Ultrasonics, Biotherapy and Biopharmaceuticals Laboratory, Institute of Biomedical Engineering, University of Oxford, Oxford, United Kingdom
| | - Yen-Ting Tung
- 3D Tissue Bioprinting Laboratory, Department of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, United States of America
| | - Esther Sim
- 3D Tissue Bioprinting Laboratory, Department of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, United States of America
| | - Yu-Chi Chen
- 3D Tissue Bioprinting Laboratory, Department of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, United States of America
| | - Emily Lee
- 3D Tissue Bioprinting Laboratory, Department of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, United States of America
| | - Marc Ferrer
- 3D Tissue Bioprinting Laboratory, Department of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, United States of America
| | - Min Jae Song
- 3D Tissue Bioprinting Laboratory, Department of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, United States of America
| |
Collapse
|
9
|
Chai J, Capik SF, Kegley B, Richeson JT, Powell JG, Zhao J. Bovine respiratory microbiota of feedlot cattle and its association with disease. Vet Res 2022; 53:4. [PMID: 35022062 PMCID: PMC8756723 DOI: 10.1186/s13567-021-01020-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 12/06/2021] [Indexed: 12/15/2022] Open
Abstract
Bovine respiratory disease (BRD), as one of the most common and costly diseases in the beef cattle industry, has significant adverse impacts on global food security and the economic stability of the industry. The bovine respiratory microbiome is strongly associated with health and disease and may provide insights for alternative therapy when treating BRD. The niche-specific microbiome communities that colonize the inter-surface of the upper and the lower respiratory tract consist of a dynamic and complex ecological system. The correlation between the disequilibrium in the respiratory ecosystem and BRD has become a hot research topic. Hence, we summarize the pathogenesis and clinical signs of BRD and the alteration of the respiratory microbiota. Current research techniques and the biogeography of the microbiome in the healthy respiratory tract are also reviewed. We discuss the process of resident microbiota and pathogen colonization as well as the host immune response. Although associations between the microbiota and BRD have been revealed to some extent, interpreting the development of BRD in relation to respiratory microbial dysbiosis will likely be the direction for upcoming studies, which will allow us to better understand the importance of the airway microbiome and its contributions to animal health and performance.
Collapse
Affiliation(s)
- Jianmin Chai
- Division of Agriculture, Department of Animal Science, University of Arkansas, Fayetteville, AR, 72701, USA
| | - Sarah F Capik
- Texas A&M AgriLife Research and Department of Veterinary Pathobiology, Texas A&M College of Veterinary Medicine and Biomedical Sciences, Canyon, TX, 79015, USA
| | - Beth Kegley
- Division of Agriculture, Department of Animal Science, University of Arkansas, Fayetteville, AR, 72701, USA
| | - John T Richeson
- Department of Agricultural Sciences, West Texas A&M University, Canyon, TX, 79016, USA
| | - Jeremy G Powell
- Division of Agriculture, Department of Animal Science, University of Arkansas, Fayetteville, AR, 72701, USA
| | - Jiangchao Zhao
- Division of Agriculture, Department of Animal Science, University of Arkansas, Fayetteville, AR, 72701, USA.
| |
Collapse
|
10
|
Archer F, Bobet-Erny A, Gomes M. State of the art on lung organoids in mammals. Vet Res 2021; 52:77. [PMID: 34078444 PMCID: PMC8170649 DOI: 10.1186/s13567-021-00946-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 05/04/2021] [Indexed: 02/08/2023] Open
Abstract
The number and severity of diseases affecting lung development and adult respiratory function have stimulated great interest in developing new in vitro models to study lung in different species. Recent breakthroughs in 3-dimensional (3D) organoid cultures have led to new physiological in vitro models that better mimic the lung than conventional 2D cultures. Lung organoids simulate multiple aspects of the real organ, making them promising and useful models for studying organ development, function and disease (infection, cancer, genetic disease). Due to their dynamics in culture, they can serve as a sustainable source of functional cells (biobanking) and be manipulated genetically. Given the differences between species regarding developmental kinetics, the maturation of the lung at birth, the distribution of the different cell populations along the respiratory tract and species barriers for infectious diseases, there is a need for species-specific lung models capable of mimicking mammal lungs as they are of great interest for animal health and production, following the One Health approach. This paper reviews the latest developments in the growing field of lung organoids.
Collapse
Affiliation(s)
- Fabienne Archer
- UMR754, IVPC, INRAE, EPHE, Univ Lyon, Université Claude Bernard Lyon 1, 69007, Lyon, France.
| | - Alexandra Bobet-Erny
- UMR754, IVPC, INRAE, EPHE, Univ Lyon, Université Claude Bernard Lyon 1, 69007, Lyon, France
| | - Maryline Gomes
- UMR754, IVPC, INRAE, EPHE, Univ Lyon, Université Claude Bernard Lyon 1, 69007, Lyon, France
| |
Collapse
|
11
|
The specific features of the developing T cell compartment of the neonatal lung are a determinant of respiratory syncytial virus immunopathogenesis. PLoS Pathog 2021; 17:e1009529. [PMID: 33909707 PMCID: PMC8109812 DOI: 10.1371/journal.ppat.1009529] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 05/10/2021] [Accepted: 04/05/2021] [Indexed: 11/19/2022] Open
Abstract
The human respiratory syncytial virus (RSV) is a major cause of severe lower respiratory tract infections in infants, possibly due to the properties of the immature neonatal pulmonary immune system. Using the newborn lamb, a classical model of human lung development and a translational model of RSV infection, we aimed to explore the role of cell-mediated immunity in RSV disease during early life. Remarkably, in healthy conditions, the developing T cell compartment of the neonatal lung showed major differences to that seen in the mature adult lung. The most striking observation being a high baseline frequency of bronchoalveolar IL-4-producing CD4+ and CD8+ T cells, which declined progressively over developmental age. RSV infection exacerbated this pro-type 2 environment in the bronchoalveolar space, rather than inducing a type 2 response per se. Moreover, regulatory T cell suppressive functions occurred very early to dampen this pro-type 2 environment, rather than shutting them down afterwards, while γδ T cells dropped and failed to produce IL-17. Importantly, RSV disease severity was related to the magnitude of those unconventional bronchoalveolar T cell responses. These findings provide novel insights in the mechanisms of RSV immunopathogenesis in early life, and constitute a major step for the understanding of RSV disease severity.
Collapse
|
12
|
Uprety T, Sreenivasan CC, Bhattarai S, Wang D, Kaushik RS, Li F. Isolation and development of bovine primary respiratory cells as model to study influenza D virus infection. Virology 2021; 559:89-99. [PMID: 33862336 DOI: 10.1016/j.virol.2021.04.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/30/2021] [Accepted: 04/05/2021] [Indexed: 02/06/2023]
Abstract
Influenza D virus (IDV) is a novel type of influenza virus that infects and causes respiratory illness in bovines. Lack of host-specific in vitro model that can recapitulate morphology and physiology of in vivo airway epithelial cells has impeded the study of IDV infection. Here, we established and characterized bovine primary respiratory epithelial cells from nasal turbinate, soft palate, and trachea of the same calf. All three cell types showed characteristics peculiar of epithelial cells, polarized into apical-basolateral membrane, and formed tight junctions. Furthermore, these cells expressed both α-2,3- and α-2,6-linked sialic acids with α-2,3 linkage being more abundant. IDV strains replicated to high titers in these cells, while influenza A and B viruses exhibited moderate to low titers, with influenza C virus replication not detected. These findings suggest that bovine primary airway epithelial cells can be utilized to model infection biology and pathophysiology of IDV and other respiratory pathogens.
Collapse
Affiliation(s)
- Tirth Uprety
- M. H. Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, KY, 40546, USA
| | - Chithra C Sreenivasan
- M. H. Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, KY, 40546, USA
| | - Shaurav Bhattarai
- Department of Biology and Microbiology, South Dakota State University, Brookings, SD, 57007, USA
| | - Dan Wang
- M. H. Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, KY, 40546, USA
| | - Radhey S Kaushik
- Department of Biology and Microbiology, South Dakota State University, Brookings, SD, 57007, USA.
| | - Feng Li
- M. H. Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, KY, 40546, USA.
| |
Collapse
|
13
|
Strässle M, Laloli L, Gultom M, V'kovski P, Stoffel MH, Crespo Pomar S, Chanfon Bätzner A, Ebert N, Labroussaa F, Dijkman R, Jores J, Thiel V. Establishment of caprine airway epithelial cells grown in an air-liquid interface system to study caprine respiratory viruses and bacteria. Vet Microbiol 2021; 257:109067. [PMID: 33862331 DOI: 10.1016/j.vetmic.2021.109067] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/06/2021] [Indexed: 11/29/2022]
Abstract
Respiratory diseases negatively impact the global goat industry, but are understudied. There is a shortage of established and biological relevant in vitro or ex vivo assays to study caprine respiratory infections. Here, we describe the establishment of an in vitro system based on well-differentiated caprine airway epithelial cell (AEC) cultures grown under air liquid interface conditions as an experimental platform to study caprine respiratory pathogens. The functional differentiation of the AEC cultures was monitored and confirmed by light and immunofluorescence microscopy, scanning electron microscopy and examination of histological sections. We validated the functionality of the platform by studying Influenza D Virus (IDV) infection and Mycoplasma mycoides subsp. capri (Mmc) colonization over 5 days, including monitoring of infectious agents by titration and qPCR as well as colour changing units, respectively. The inoculation of caprine AEC cultures with IDV showed that efficient viral replication takes place, and revealed that IDV has a marked cell tropism for ciliated cells. Furthermore, AEC cultures were successfully infected with Mmc using a multiplicity of infection of 0.1 and colonization was monitored over several days. Altogether, these results demonstrate that our newly-established caprine AEC cultures can be used to investigate host-pathogen interactions of caprine respiratory pathogens.
Collapse
Affiliation(s)
- Marina Strässle
- Institute of Virology and Immunology (IVI), Bern, Switzerland; Institute of Veterinary Bacteriology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Länggass-Str. 122, PO Box 3001, Bern, Switzerland; Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Laura Laloli
- Institute of Virology and Immunology (IVI), Bern, Switzerland; Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland; Graduate School for Biomedical Science, University of Bern, Bern, Switzerland; Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Mitra Gultom
- Institute of Virology and Immunology (IVI), Bern, Switzerland; Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland; Graduate School for Biomedical Science, University of Bern, Bern, Switzerland; Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Philip V'kovski
- Institute of Virology and Immunology (IVI), Bern, Switzerland; Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Michael H Stoffel
- Division of Veterinary Anatomy, Vetsuisse Faculty, University of Bern, Länggass-Str. 120, PO Box 3001, Bern, Switzerland
| | - Silvia Crespo Pomar
- Institute of Veterinary Bacteriology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Länggass-Str. 122, PO Box 3001, Bern, Switzerland
| | - Astrid Chanfon Bätzner
- Institute of Animal Pathology (COMPATH), Vetsuisse Faculty, University of Bern, Länggass-Str. 122, PO Box 3001, Bern, Switzerland
| | - Nadine Ebert
- Institute of Virology and Immunology (IVI), Bern, Switzerland; Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Fabien Labroussaa
- Institute of Veterinary Bacteriology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Länggass-Str. 122, PO Box 3001, Bern, Switzerland
| | - Ronald Dijkman
- Institute of Virology and Immunology (IVI), Bern, Switzerland; Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland; Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Joerg Jores
- Institute of Veterinary Bacteriology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Länggass-Str. 122, PO Box 3001, Bern, Switzerland.
| | - Volker Thiel
- Institute of Virology and Immunology (IVI), Bern, Switzerland; Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| |
Collapse
|
14
|
Preliminary Study on the Development of In Vitro Human Respiratory Epithelium Using Collagen Type I Scaffold as a Potential Model for Future Tracheal Tissue Engineering. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app11041787] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Pathological conditions of the tracheal epithelium, such as postoperative injuries and chronic conditions, often compromise the functionality of the respiratory epithelium. Although replacement of the respiratory epithelium using various types of tracheal transplantation has been attempted, there is no predictable and dependable replacement method that holds for safe and practicable long-term use. Therefore, we used a tissue engineering approach for ex vivo regeneration of the respiratory epithelium (RE) construct. Collagen type I was isolated from sheep tendon and it was fabricated in a three-dimensional (3D) scaffold format. Isolated human respiratory epithelial cells (RECs) and fibroblasts from nasal turbinate were co-cultured on the 3D scaffold for 48 h, and epithelium maturation was allowed for another 14 days in an air–liquid interface culture system. The scanning electron microscope results revealed a fabricated porous-structure 3D collagen scaffold. The scaffold was found to be biocompatible with RECs and fibroblasts and allows cells attachment, proliferation, and migration. Immunohistochemical analysis showed that the seeded RECs and fibroblasts were positive for expression of cytokeratin 14 and collagen type I markers, respectively, indicating that the scaffold supports the native phenotype of seeded cells over a period of 14 days. Although a longer maturation period is needed for ciliogenesis to occur in RECs, the findings suggest that the tissue-engineered RE construct is a potential candidate for direct use in tracheal epithelium replacement or tracheal tube reengineering.
Collapse
|
15
|
O'Boyle N, Berry CC, Davies RL. Differentiated ovine tracheal epithelial cells support the colonisation of pathogenic and non-pathogenic strains of Mannheimia haemolytica. Sci Rep 2020; 10:14971. [PMID: 32917945 PMCID: PMC7486916 DOI: 10.1038/s41598-020-71604-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 08/07/2020] [Indexed: 11/15/2022] Open
Abstract
Mannheimia haemolytica is the primary bacterial species associated with respiratory disease of ruminants. A lack of cost-effective, reproducible models for the study of M. haemolytica pathogenesis has hampered efforts to better understand the molecular interactions governing disease progression. We employed a highly optimised ovine tracheal epithelial cell model to assess the colonisation of various pathogenic and non-pathogenic M. haemolytica isolates of bovine and ovine origin. Comparison of single representative pathogenic and non-pathogenic ovine isolates over ten time-points by enumeration of tissue-associated bacteria, histology, immunofluorescence microscopy and scanning electron microscopy revealed temporal differences in adhesion, proliferation, bacterial cell physiology and host cell responses. Comparison of eight isolates of bovine and ovine origin at three key time-points (2 h, 48 h and 72 h), revealed that colonisation was not strictly pathogen or serotype specific, with isolates of serotype A1, A2, A6 and A12 being capable of colonising the cell layer regardless of host species or disease status of the host. A trend towards increased proliferative capacity by pathogenic ovine isolates was observed. These results indicate that the host-specific nature of M. haemolytica infection may result at least partially from the colonisation-related processes of adhesion, invasion and proliferation at the epithelial interface.
Collapse
Affiliation(s)
- Nicky O'Boyle
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Catherine C Berry
- Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Robert L Davies
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
16
|
Cai Y, Varasteh S, van Putten JPM, Folkerts G, Braber S. Mannheimia haemolytica and lipopolysaccharide induce airway epithelial inflammatory responses in an extensively developed ex vivo calf model. Sci Rep 2020; 10:13042. [PMID: 32747652 PMCID: PMC7400546 DOI: 10.1038/s41598-020-69982-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 07/20/2020] [Indexed: 01/27/2023] Open
Abstract
Pulmonary infection is associated with inflammation and damage to the bronchial epithelium characterized by an increase in the release of inflammatory factors and a decrease in airway barrier function. Our objective is to optimize a method for the isolation and culture of primary bronchial epithelial cells (PBECs) and to provide an ex vivo model to study mechanisms of epithelial airway inflammation. PBECs were isolated and cultured from the airways of calves in a submerged cell culture and liquid-liquid interface system. A higher yield and cell viability were obtained after stripping the epithelium from the bronchial section compared to cutting the bronchial section in smaller pieces prior to digestion. Mannheimia haemolytica and lipopolysaccharide (LPS) as stimulants increased inflammatory responses (IL-8, IL-6 and TNF-α release), possibly, by the activation of "TLR-mediated MAPKs and NF-κB" signaling. Furthermore, M. haemolytica and LPS disrupted the bronchial epithelial layer as observed by a decreased transepithelial electrical resistance and zonula occludens-1 and E-cadherin expression. An optimized isolation and culture method for calf PBECs was developed, which cooperated with animal use Replacement, Reduction and Refinement (3R's) principle, and can also contribute to the increased knowledge and development of effective therapies for other animal and humans (childhood) respiratory diseases.
Collapse
Affiliation(s)
- Yang Cai
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Soheil Varasteh
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Jos P M van Putten
- Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, The Netherlands
| | - Gert Folkerts
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Saskia Braber
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
17
|
Luengen AE, Kniebs C, Buhl EM, Cornelissen CG, Schmitz-Rode T, Jockenhoevel S, Thiebes AL. Choosing the Right Differentiation Medium to Develop Mucociliary Phenotype of Primary Nasal Epithelial Cells In Vitro. Sci Rep 2020; 10:6963. [PMID: 32332878 PMCID: PMC7181704 DOI: 10.1038/s41598-020-63922-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 04/06/2020] [Indexed: 11/08/2022] Open
Abstract
In vitro differentiation of airway epithelium is of interest for respiratory tissue engineering and studying airway diseases. Both applications benefit from the use of primary cells to maintain a mucociliated phenotype and thus physiological functionality. Complex differentiation procedures often lack standardization and reproducibility. To alleviate these shortfalls, we compared differentiation behavior of human nasal epithelial cells in four differentiation media. Cells were differentiated at the air-liquid interface (ALI) on collagen-coated inserts. Mucociliary differentiation status after five weeks was analyzed by electron microscopy, histology and immunohistochemistry. The amount of ciliation was estimated and growth factor concentrations were evaluated using ELISA. We found that retinoic-acid-supplemented mixture of DMEM and Airway Epithelial Cell Growth Medium gave most promising results to obtain ciliated and mucus producing nasal epithelium in vitro. We discovered the balance between retinoic acid (RA), vascular endothelial growth factor (VEGF), epidermal growth factor (EGF) and fibroblast growth factor β (FGF-β) to be relevant for differentiation. We could show that low VEGF, EGF and FGF-β concentrations in medium correspond to absent ciliation in specific donors. Therefore, our results may in future facilitate donor selection and non-invasive monitoring of ALI cultures and by this contribute to improved standardization of epithelial in vitro culture.
Collapse
Affiliation(s)
- Anja E Luengen
- Department of Biohybrid and Medical Textiles (BioTex), AME - Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Forckenbeckstraße 55, 52074, Aachen, Germany.
- Aachen-Maastricht Institute for Biobased Materials, Faculty of Science and Engineering, Maastricht University, Brightlands Chemelot Campus, 6167 RD, Geleen, The Netherlands.
| | - Caroline Kniebs
- Department of Biohybrid and Medical Textiles (BioTex), AME - Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Forckenbeckstraße 55, 52074, Aachen, Germany
- Aachen-Maastricht Institute for Biobased Materials, Faculty of Science and Engineering, Maastricht University, Brightlands Chemelot Campus, 6167 RD, Geleen, The Netherlands
| | - Eva Miriam Buhl
- Institute of Pathology, Electron Microscopy Facility, RWTH Aachen University Hospital, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Christian G Cornelissen
- Department of Biohybrid and Medical Textiles (BioTex), AME - Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Forckenbeckstraße 55, 52074, Aachen, Germany
- Clinic for Pneumology and Internal Intensive Care Medicine (Medical Clinic V), RWTH Aachen University Hospital, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Thomas Schmitz-Rode
- Department of Biohybrid and Medical Textiles (BioTex), AME - Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Forckenbeckstraße 55, 52074, Aachen, Germany
| | - Stefan Jockenhoevel
- Department of Biohybrid and Medical Textiles (BioTex), AME - Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Forckenbeckstraße 55, 52074, Aachen, Germany.
- Aachen-Maastricht Institute for Biobased Materials, Faculty of Science and Engineering, Maastricht University, Brightlands Chemelot Campus, 6167 RD, Geleen, The Netherlands.
| | - Anja Lena Thiebes
- Department of Biohybrid and Medical Textiles (BioTex), AME - Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Forckenbeckstraße 55, 52074, Aachen, Germany
- Aachen-Maastricht Institute for Biobased Materials, Faculty of Science and Engineering, Maastricht University, Brightlands Chemelot Campus, 6167 RD, Geleen, The Netherlands
| |
Collapse
|
18
|
Rock Inhibitor Y-27632 Enables Feeder-Free, Unlimited Expansion of Sus scrofa domesticus Swine Airway Stem Cells to Facilitate Respiratory Research. Stem Cells Int 2019; 2019:3010656. [PMID: 31871466 PMCID: PMC6906834 DOI: 10.1155/2019/3010656] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 10/29/2019] [Indexed: 12/21/2022] Open
Abstract
Current limitations in the efficacy of treatments for chronic respiratory disorders position them as prospective regenerative medicine therapeutic targets. A substantial barrier to these ambitions is that research requires large numbers of cells whose acquisition is hindered by the limited availability of human tissue samples leading to an overreliance on physiologically dissimilar rodents. The development of cell culture strategies for airway cells from large mammals will more effectively support the transition from basic research to clinical therapy. Using readily available porcine lungs, we isolated conducting airway tissue and subsequently a large number of porcine airway epithelial cells (pAECs) using a digestion and mechanical scraping technique. Cells were cultured in a variety of culture media formulations, both foetal bovine serum-containing and serum-free media, in air (21%) and physiological (2%) oxygen tension and in the presence and absence of Rho kinase inhibitor Y-27362 (RI). Cell number at isolation and subsequent population doublings were determined; cells were characterised during culture and following differentiation by immunofluorescence, histology, and IL-8 ELISA. Cells were positive for epithelial markers (pan-cytokeratin and E-cadherin) and negative for fibroblastic markers (vimentin and smooth muscle actin). Supplementation of cultures with Y-27632 allowed for unlimited expansion whilst sustaining an epithelial phenotype. Early passage pAECs readily produced differentiated air-liquid interface (ALI) cultures with a capacity for mucociliary differentiation retained after substantial expansion, strongly modulated by the culture condition applied. Primary pAECs will be a useful tool to further respiratory-oriented research whilst RI-expanded pAECs are a promising tool, particularly with further optimisation of culture conditions.
Collapse
|
19
|
Sudaryatma PE, Mekata H, Kubo M, Subangkit M, Goto Y, Okabayashi T. Co-infection of epithelial cells established from the upper and lower bovine respiratory tract with bovine respiratory syncytial virus and bacteria. Vet Microbiol 2019; 235:80-85. [PMID: 31282382 DOI: 10.1016/j.vetmic.2019.06.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 06/06/2019] [Accepted: 06/12/2019] [Indexed: 11/17/2022]
Abstract
Bovine respiratory disease complex is a major disease affecting the global cattle industry. Multiple infections by viruses and bacteria increase disease severity. Previously, we reported that bovine respiratory syncytial virus (BRSV) infection increases adherence of Pasteurella multocida to human respiratory and bovine kidney epithelial cells. To examine the interaction between the virus and bacteria in bovine respiratory cells, we generated respiratory epithelial cell lines from bovine trachea (bTEC), bronchus (bBEC), and lung (bLEC). Although all established cell lines were infected by BRSV and P. multocida susceptibility differed according to site of origin. The cells derived from the lower respiratory tract (bBEC and bLEC) were significantly more susceptible to BRSV than those derived from the upper respiratory tract (bTEC). Pre-infection of bBEC and bLEC with BRSV increased adherence of P. multocida; this was not the case for bTEC. These results indicate that BRSV may reproduce better in the lower respiratory tract and encourage adherence of bacteria. Thus, we identify one possible mechanism underlying severe pneumonia.
Collapse
Affiliation(s)
- Putu Eka Sudaryatma
- Graduate School of Medicine and Veterinary Medicine, University of Miyazaki, Miyazaki, Japan; Department of Veterinary Science, Faculty of Agriculture, University of Miyazaki, Miyazaki, Japan
| | - Hirohisa Mekata
- Center for Animal Disease Control, University of Miyazaki, Miyazaki, Japan; Organization for Promotion of Tenure Track University of Miyazaki, Miyazaki, Japan
| | - Meiko Kubo
- Miyakonojo Meat Inspection Centre Miyazaki Prefecture Government, Miyazaki, Japan
| | - Mawar Subangkit
- Graduate School of Medicine and Veterinary Medicine, University of Miyazaki, Miyazaki, Japan; Department of Veterinary Science, Faculty of Agriculture, University of Miyazaki, Miyazaki, Japan
| | - Yoshitaka Goto
- Department of Veterinary Science, Faculty of Agriculture, University of Miyazaki, Miyazaki, Japan; Center for Animal Disease Control, University of Miyazaki, Miyazaki, Japan
| | - Tamaki Okabayashi
- Department of Veterinary Science, Faculty of Agriculture, University of Miyazaki, Miyazaki, Japan; Center for Animal Disease Control, University of Miyazaki, Miyazaki, Japan.
| |
Collapse
|
20
|
Pathogenic Mannheimia haemolytica Invades Differentiated Bovine Airway Epithelial Cells. Infect Immun 2019; 87:IAI.00078-19. [PMID: 30962401 PMCID: PMC6529648 DOI: 10.1128/iai.00078-19] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 04/01/2019] [Indexed: 12/14/2022] Open
Abstract
The Gram-negative bacterium Mannheimia haemolytica is the primary bacterial species associated with bovine respiratory disease (BRD) and is responsible for significant economic losses to livestock industries worldwide. Healthy cattle are frequently colonized by commensal serotype A2 strains, but disease is usually caused by pathogenic strains of serotype A1. For reasons that are poorly understood, a transition occurs within the respiratory tract and a sudden explosive proliferation of serotype A1 bacteria leads to the onset of pneumonic disease. Very little is known about the interactions of M. haemolytica with airway epithelial cells of the respiratory mucosa which might explain the different abilities of serotype A1 and A2 strains to cause disease. In the present study, host-pathogen interactions in the bovine respiratory tract were mimicked using a novel differentiated bovine bronchial epithelial cell (BBEC) infection model. In this model, differentiated BBECs were inoculated with serotype A1 or A2 strains of M. haemolytica and the course of infection followed over a 5-day period by microscopic assessment and measurement of key proinflammatory mediators. We have demonstrated that serotype A1, but not A2, M. haemolytica invades differentiated BBECs by transcytosis and subsequently undergoes rapid intracellular replication before spreading to adjacent cells and causing extensive cellular damage. Our findings suggest that the explosive proliferation of serotype A1 M. haemolytica that occurs within the bovine respiratory tract prior to the onset of pneumonic disease is potentially due to bacterial invasion of, and rapid proliferation within, the mucosal epithelium. The discovery of this previously unrecognized mechanism of pathogenesis is important because it will allow the serotype A1-specific virulence determinants responsible for invasion to be identified and thereby provide opportunities for the development of new strategies for combatting BRD aimed at preventing early colonization and infection of the bovine respiratory tract.
Collapse
|
21
|
Romanova OA, Tenchurin TH, Demina TS, Sytina EV, Shepelev AD, Rudyak SG, Klein OI, Krasheninnikov SV, Safronova EI, Kamyshinsky RA, Mamagulashvili VG, Akopova TA, Chvalun SN, Panteleyev AA. Non-woven bilayered biodegradable chitosan-gelatin-polylactide scaffold for bioengineering of tracheal epithelium. Cell Prolif 2019; 52:e12598. [PMID: 30900363 PMCID: PMC6536443 DOI: 10.1111/cpr.12598] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 01/02/2019] [Accepted: 01/29/2019] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVES The conversion of tissue engineering into a routine clinical tool cannot be achieved without a deep understanding of the interaction between cells and scaffolds during the process of tissue formation in an artificial environment. Here, we have investigated the cultivation conditions and structural features of the biodegradable non-woven material in order to obtain a well-differentiated human airway epithelium. MATERIALS AND METHODS The bilayered scaffold was fabricated by electrospinning technology. The efficiency of the scaffold has been evaluated using MTT cell proliferation assay, histology, immunofluorescence and electron microscopy. RESULTS With the use of a copolymer of chitosan-gelatin-poly-l-lactide, a bilayered non-woven scaffold was generated and characterized. The optimal structural parameters of both layers for cell proliferation and differentiation were determined. The basal airway epithelial cells differentiated into ciliary and goblet cells and formed pseudostratified epithelial layer on the surface of the scaffold. In addition, keratinocytes formed a skin equivalent when seeded on the same scaffold. A comparative analysis of growth and differentiation for both types of epithelium was performed. CONCLUSIONS The structural parameters of nanofibres should be selected experimentally depending on polymer composition. The major challenges on the way to obtain the well-differentiated equivalent of respiratory epithelium on non-woven scaffold include the following: the balance between scaffold permeability and thickness, proper combination of synthetic and natural components, and culture conditions sufficient for co-culturing of airway epithelial cells and fibroblasts. For generation of skin equivalent, the lack of diffusion is not so critical as for pseudostratified airway epithelium.
Collapse
Affiliation(s)
- Olga A. Romanova
- Kurchatov Complex of NBICS TechnologiesNRC Kurchatov InstituteMoscowRussian Federation
| | - Timur H. Tenchurin
- Kurchatov Complex of NBICS TechnologiesNRC Kurchatov InstituteMoscowRussian Federation
| | - Tatiana S. Demina
- Enikolopov Institute of Synthetic Polymeric Materials, Russian Academy of SciencesMoscowRussian Federation
- Sechenov First Moscow State Medical UniversityMoscowRussian Federation
| | - Elena V. Sytina
- Kurchatov Complex of NBICS TechnologiesNRC Kurchatov InstituteMoscowRussian Federation
| | - Alexey D. Shepelev
- Kurchatov Complex of NBICS TechnologiesNRC Kurchatov InstituteMoscowRussian Federation
| | - Stanislav G. Rudyak
- Emanuel Institute of Biochemical Physics, Russian Academy of SciencesMoscowRussian Federation
| | - Olga I. Klein
- Kurchatov Complex of NBICS TechnologiesNRC Kurchatov InstituteMoscowRussian Federation
| | | | | | - Roman A. Kamyshinsky
- Kurchatov Complex of NBICS TechnologiesNRC Kurchatov InstituteMoscowRussian Federation
| | | | - Tatiana A. Akopova
- Enikolopov Institute of Synthetic Polymeric Materials, Russian Academy of SciencesMoscowRussian Federation
| | - Sergey N. Chvalun
- Kurchatov Complex of NBICS TechnologiesNRC Kurchatov InstituteMoscowRussian Federation
| | - Andrey A. Panteleyev
- Kurchatov Complex of NBICS TechnologiesNRC Kurchatov InstituteMoscowRussian Federation
| |
Collapse
|
22
|
Toor A, Culibrk L, Singhera GK, Moon KM, Prudova A, Foster LJ, Moore MM, Dorscheid DR, Tebbutt SJ. Transcriptomic and proteomic host response to Aspergillus fumigatus conidia in an air-liquid interface model of human bronchial epithelium. PLoS One 2018; 13:e0209652. [PMID: 30589860 PMCID: PMC6307744 DOI: 10.1371/journal.pone.0209652] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 12/10/2018] [Indexed: 12/31/2022] Open
Abstract
Aspergillus fumigatus (A. fumigatus) is a wide-spread fungus that is a potent allergen in hypersensitive individuals but also an opportunistic pathogen in immunocompromised patients. It reproduces asexually by releasing airborne conidiospores (conidia). Upon inhalation, fungal conidia are capable of reaching the airway epithelial cells (AECs) in bronchial and alveolar tissues. Previous studies have predominantly used submerged monolayer cultures for studying this host-pathogen interaction; however, these cultures do not recapitulate the mucocililary differentiation phenotype of the in vivo epithelium in the respiratory tract. Thus, the aim of this study was to use well-differentiated primary human bronchial epithelial cells (HBECs) grown at the air-liquid interface (ALI) to determine their transcriptomic and proteomic responses following interaction with A. fumigatus conidia. We visualized conidial interaction with HBECs using confocal laser scanning microscopy (CLSM), and applied NanoString nCounter and shotgun proteomics to assess gene expression changes in the human cells upon interaction with A. fumigatus conidia. Western blot analysis was used to assess the expression of top three differentially expressed proteins, CALR, SET and NUCB2. CLSM showed that, unlike submerged monolayer cultures, well-differentiated ALI cultures of primary HBECs were estimated to internalize less than 1% of bound conidia. Nevertheless, transcriptomic and proteomic analyses revealed numerous differentially expressed host genes; these were enriched for pathways including apoptosis/autophagy, translation, unfolded protein response and cell cycle (up-regulated); complement and coagulation pathways, iron homeostasis, nonsense mediated decay and rRNA binding (down-regulated). CALR and SET were confirmed to be up-regulated in ALI cultures of primary HBECs upon exposure to A. fumigatus via western blot analysis. Therefore, using transcriptomics and proteomics approaches, ALI models recapitulating the bronchial epithelial barrier in the conductive zone of the respiratory tract can provide novel insights to the molecular response of bronchial epithelial cells upon exposure to A. fumigatus conidia.
Collapse
Affiliation(s)
- Amreen Toor
- Experimental Medicine, University of British Columbia, Vancouver, Canada
- Centre for Heart Lung Innovation, University of British Columbia and St. Paul’s Hospital, Vancouver, Canada
| | - Luka Culibrk
- Centre for Heart Lung Innovation, University of British Columbia and St. Paul’s Hospital, Vancouver, Canada
| | - Gurpreet K. Singhera
- Centre for Heart Lung Innovation, University of British Columbia and St. Paul’s Hospital, Vancouver, Canada
| | - Kyung-Mee Moon
- Department of Biochemistry & Molecular Biology, University of British Columbia, Vancouver, Canada
| | - Anna Prudova
- Department of Biochemistry & Molecular Biology, University of British Columbia, Vancouver, Canada
| | - Leonard J. Foster
- Department of Biochemistry & Molecular Biology, University of British Columbia, Vancouver, Canada
| | - Margo M. Moore
- Department of Biological Sciences, Simon Fraser University, Burnaby, Canada
| | - Delbert R. Dorscheid
- Centre for Heart Lung Innovation, University of British Columbia and St. Paul’s Hospital, Vancouver, Canada
- Department of Medicine, Division of Respiratory Medicine, University of British Columbia, Vancouver, Canada
| | - Scott J. Tebbutt
- Centre for Heart Lung Innovation, University of British Columbia and St. Paul’s Hospital, Vancouver, Canada
- Department of Medicine, Division of Respiratory Medicine, University of British Columbia, Vancouver, Canada
- Prevention of Organ Failure (PROOF) Centre of Excellence, Vancouver, Canada
| |
Collapse
|
23
|
Temporal differentiation of bovine airway epithelial cells grown at an air-liquid interface. Sci Rep 2018; 8:14893. [PMID: 30291311 PMCID: PMC6173764 DOI: 10.1038/s41598-018-33180-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 09/21/2018] [Indexed: 12/21/2022] Open
Abstract
There is an urgent need to develop improved, physiologically-relevant in vitro models of airway epithelia with which to better understand the pathological processes associated with infection, allergies and toxicological insults of the respiratory tract of both humans and domesticated animals. In the present study, we have characterised the proliferation and differentiation of primary bovine bronchial epithelial cells (BBECs) grown at an air-liquid interface (ALI) at three-day intervals over a period of 42 days from the introduction of the ALI. The differentiated BBEC model was highly representative of the ex vivo epithelium from which the epithelial cells were derived; a columnar, pseudostratified epithelium that was highly reflective of native airway epithelium was formed which comprised ciliated, goblet and basal cells. The hallmark defences of the respiratory tract, namely barrier function and mucociliary clearance, were present, thus demonstrating that the model is an excellent mimic of bovine respiratory epithelium. The epithelium was fully differentiated by day 21 post-ALI and, crucially, remained healthy and stable for a further 21 days. Thus, the differentiated BBEC model has a three-week window which will allow wide-ranging and long-term experiments to be performed in the fields of infection, toxicology or general airway physiology.
Collapse
|
24
|
O’Boyle N, Sutherland E, Berry CC, Davies RL. Optimisation of growth conditions for ovine airway epithelial cell differentiation at an air-liquid interface. PLoS One 2018; 13:e0193998. [PMID: 29518140 PMCID: PMC5843276 DOI: 10.1371/journal.pone.0193998] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 02/22/2018] [Indexed: 11/18/2022] Open
Abstract
Respiratory tract infections are of significant concern in the agriculture industry. There is a requirement for the development of well-characterised in vitro epithelial cell culture models in order to dissect the diverse molecular interactions occurring at the host-pathogen interface in airway epithelia. We have analysed key factors that influence growth and differentiation of ovine tracheal epithelial cells in an air-liquid interface (ALI) culture system. Cellular differentiation was assessed at 21 days post-ALI, a time-point which we have previously shown to be sufficient for differentiation in standard growth conditions. We identified a dose-dependent response to epidermal growth factor (EGF) in terms of both epithelial thickening and ciliation levels. Maximal ciliation levels were observed with 25 ng ml-1 EGF. We identified a strict requirement for retinoic acid (RA) in epithelial differentiation as RA exclusion resulted in the formation of a stratified squamous epithelium, devoid of cilia. The pore-density of the growth substrate also had an influence on differentiation as high pore-density inserts yielded higher levels of ciliation and more uniform cell layers than low pore-density inserts. Differentiation was also improved by culturing the cells in an atmosphere of sub-ambient oxygen concentration. We compared two submerged growth media and observed differences in the rate of proliferation/expansion, barrier formation and also in terminal differentiation. Taken together, these results indicate important differences between the response of ovine tracheal epithelial cells and other previously described airway epithelial models, to a variety of environmental conditions. These data also indicate that the phenotype of ovine tracheal epithelial cells can be tailored in vitro by precise modulation of growth conditions, thereby yielding a customisable, potential infection model.
Collapse
Affiliation(s)
- Nicky O’Boyle
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Erin Sutherland
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Catherine C. Berry
- Institute of Molecular Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Robert L. Davies
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|