1
|
Daneshdoust D, He K, Wang QE, Li J, Liu X. Modeling respiratory tract diseases for clinical translation employing conditionally reprogrammed cells. CELL INSIGHT 2024; 3:100201. [PMID: 39391007 PMCID: PMC11462205 DOI: 10.1016/j.cellin.2024.100201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 09/06/2024] [Accepted: 09/08/2024] [Indexed: 10/12/2024]
Abstract
Preclinical models serve as indispensable tools in translational medicine. Specifically, patient-derived models such as patient-derived xenografts (PDX), induced pluripotent stem cells (iPSC), organoids, and recently developed technique of conditional reprogramming (CR) have been employed to reflect the host characteristics of diseases. CR technology involves co-culturing epithelial cells with irradiated Swiss-3T3-J2 mouse fibroblasts (feeder cells) in the presence of a Rho kinase (ROCK) inhibitor, Y-27632. CR technique facilitates the rapid conversion of both normal and malignant cells into a "reprogrammed stem-like" state, marked by robust in vitro proliferation. This is achieved without reliance on exogenous gene expression or viral transfection, while maintaining the genetic profile of the parental cells. So far, CR technology has been used to study biology of diseases, targeted therapies (precision medicine), regenerative medicine, and noninvasive diagnosis and surveillance. Respiratory diseases, ranking as the third leading cause of global mortality, pose a significant burden to healthcare systems worldwide. Given the substantial mortality and morbidity rates of respiratory diseases, efficient and rapid preclinical models are imperative to accurately recapitulate the diverse spectrum of respiratory conditions. In this article, we discuss the applications and future potential of CR technology in modeling various respiratory tract diseases, including lung cancer, respiratory viral infections (such as influenza and Covid-19 and etc.), asthma, cystic fibrosis, respiratory papillomatosis, and upper aerodigestive track tumors. Furthermore, we discuss the potential utility of CR in personalized medicine, regenerative medicine, and clinical translation.
Collapse
Affiliation(s)
- Danyal Daneshdoust
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Kai He
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Division of Medical Oncology, Department of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Qi-En Wang
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Department of Radiation Oncology, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Jenny Li
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Department of Pathology, Wexner Medical Center, Ohio State University, Columbus, OH, USA
| | - Xuefeng Liu
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Departments of Pathology, Urology, and Radiation Oncology, Wexner Medical Center, Ohio State University, Columbus, OH, USA
| |
Collapse
|
2
|
McCarron A, Ling KM, Montgomery ST, Martinovich KM, Cmielewski P, Rout-Pitt N, Kicic A, Parsons D, Donnelley M. Lentiviral vector gene therapy and CFTR modulators show comparable effectiveness in cystic fibrosis rat airway models. Gene Ther 2024:10.1038/s41434-024-00480-y. [PMID: 39183346 DOI: 10.1038/s41434-024-00480-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 08/08/2024] [Accepted: 08/13/2024] [Indexed: 08/27/2024]
Abstract
Mutation-agnostic treatments such as airway gene therapy have the potential to treat any individual with cystic fibrosis (CF), irrespective of their CF transmembrane conductance regulator (CFTR) gene variants. The aim of this study was to employ two CF rat models, Phe508del and CFTR knockout (KO), to assess the comparative effectiveness of CFTR modulators and lentiviral (LV) vector-mediated gene therapy. Cells were isolated from the tracheas of rats and used to establish air-liquid interface (ALI) cultures. Phe508del rat ALIs were treated with the modulator combination, elexacaftor-tezacaftor-ivacaftor (ETI), and separate groups of Phe508del and KO tracheal epithelial cells were treated with LV-CFTR followed by differentiation at ALI. Ussing chamber measurements were performed to assess CFTR function. ETI-treated Phe508del ALI cultures demonstrated CFTR function that was 59% of wild-type level, while gene-addition therapy restored Phe508del to 68% and KO to 47% of wild-type level, respectively. Our findings show that rat Phe508del-CFTR protein can be successfully rescued with ETI treatment, and that CFTR gene-addition therapy provides significant CFTR correction in Phe508del and KO ALI cultures to levels that were comparable to ETI. These findings highlight the potential of an LV vector-based gene therapy for the treatment of CF lung disease.
Collapse
Affiliation(s)
- Alexandra McCarron
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia.
- Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia.
- Department of Respiratory and Sleep Medicine, Women's and Children's Hospital, North Adelaide, SA, Australia.
| | - Kak-Ming Ling
- Wal-Yan Respiratory Research Centre, Telethon Kids Institute, Nedlands, WA, Australia
- School of Population Health, Curtin University, Bentley, WA, Australia
| | - Samuel T Montgomery
- Wal-Yan Respiratory Research Centre, Telethon Kids Institute, Nedlands, WA, Australia
- School of Population Health, Curtin University, Bentley, WA, Australia
| | - Kelly M Martinovich
- Wal-Yan Respiratory Research Centre, Telethon Kids Institute, Nedlands, WA, Australia
- Centre for Child Health Research, University of Western Australia, Crawley, WA, Australia
| | - Patricia Cmielewski
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
- Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
- Department of Respiratory and Sleep Medicine, Women's and Children's Hospital, North Adelaide, SA, Australia
| | - Nathan Rout-Pitt
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
- Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
- Department of Respiratory and Sleep Medicine, Women's and Children's Hospital, North Adelaide, SA, Australia
| | - Anthony Kicic
- Wal-Yan Respiratory Research Centre, Telethon Kids Institute, Nedlands, WA, Australia
- School of Population Health, Curtin University, Bentley, WA, Australia
- Department of Respiratory and Sleep, Perth Children's Hospital, Nedlands, WA, Australia
- Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia and Harry Perkins Institute of Medical Research, Nedlands, WA, Australia
| | - David Parsons
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
- Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
- Department of Respiratory and Sleep Medicine, Women's and Children's Hospital, North Adelaide, SA, Australia
| | - Martin Donnelley
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
- Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
- Department of Respiratory and Sleep Medicine, Women's and Children's Hospital, North Adelaide, SA, Australia
| |
Collapse
|
3
|
Sun L, Walls SA, Dang H, Quinney NL, Sears PR, Sadritabrizi T, Hasegawa K, Okuda K, Asakura T, Chang X, Zheng M, Mikami Y, Dizmond FU, Danilova D, Zhou L, Deshmukh A, Cholon DM, Radicioni G, Rogers TD, Kissner WJ, Markovetz MR, Guhr Lee TN, Gutay MI, Esther CR, Chua M, Grubb BR, Ehre C, Kesimer M, Hill DB, Ostrowski LE, Button B, Gentzsch M, Robinson C, Olivier KN, Freeman AF, Randell SH, O'Neal WK, Boucher RC, Chen G. Dysregulated Airway Host Defense in Hyper IgE Syndrome due to STAT3 Mutations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.14.607930. [PMID: 39211176 PMCID: PMC11361074 DOI: 10.1101/2024.08.14.607930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Rationale Hyper IgE syndrome (STAT3-HIES), also known as Job's syndrome, is a rare immunodeficiency disease typically caused by dominant-negative STAT3 mutations. STAT3-HIES syndrome is characterized by chronic pulmonary infection and inflammation, suggesting impairment of pulmonary innate host defense. Objectives To identify airway epithelial host defense defects consequent to STAT3 mutations that, in addition to reported mutant STAT3 immunologic abnormalities, produce pulmonary infection. Methods STAT3-HIES sputum was evaluated for biochemical/biophysical properties. STAT3-HIES excised lungs were harvested for histology; bronchial brush samples were collected for RNA sequencing and in vitro culture. A STAT3-HIES-specific mutation (R382W), expressed by lentiviruses, and a STAT3 knockout, generated by CRISPR/Cas9, were maintained in normal human bronchial epithelia under basal or inflammatory (IL1β) conditions. Effects of STAT3 deficiency on transcriptomics, and epithelial ion channel, secretory, antimicrobial, and ciliary functions were assessed. Measurements and Main Results Mucus concentrations and viscoelasticity were increased in STAT3-HIES sputum. STAT3-HIES excised lungs exhibited mucus obstruction and elevated IL1β expression. STAT3 deficiency impaired CFTR-dependent fluid and mucin secretion, inhibited expression of antimicrobial peptides, cytokines, and chemokines, and acidified airway surface liquid at baseline and post-IL1β exposure in vitro. Notably, mutant STAT3 suppressed IL1R1 expression. STAT3 mutations also inhibited ciliogenesis in vivo and impaired mucociliary transport in vitro, a process mediated via HES6 suppression. Administration of a γ-secretase inhibitor increased HES6 expression and improved ciliogenesis in STAT3 R382W mutant cells. Conclusions STAT3 dysfunction leads to multi-component defects in airway epithelial innate defense, which, in conjunction with STAT3-HIES immune deficiency, contributes to chronic pulmonary infection.
Collapse
|
4
|
Evans DJ, Hillas JK, Iosifidis T, Simpson SJ, Kicic A, Agudelo-Romero P. Transcriptomic analysis of primary nasal epithelial cells reveals altered interferon signalling in preterm birth survivors at one year of age. Front Cell Dev Biol 2024; 12:1399005. [PMID: 39114569 PMCID: PMC11303191 DOI: 10.3389/fcell.2024.1399005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 06/21/2024] [Indexed: 08/10/2024] Open
Abstract
Introduction: Many survivors of preterm birth (<37 weeks gestation) have lifelong respiratory deficits, the drivers of which remain unknown. Influencers of pathophysiological outcomes are often detectable at the gene level and pinpointing these differences can help guide targeted research and interventions. This study provides the first transcriptomic analysis of primary nasal airway epithelial cells in survivors of preterm birth at approximately 1 year of age. Methods: Nasal airway epithelial brushings were collected, and primary cell cultures established from term (>37 weeks gestation) and very preterm participants (≤32 weeks gestation). Ex vivo RNA was collected from brushings with sufficient cell numbers and in vitro RNA was extracted from cultured cells, with bulk RNA sequencing performed on both the sample types. Differential gene expression was assessed using the limma-trend pipeline and pathway enrichment identified using Reactome and GO analysis. To corroborate gene expression data, cytokine concentrations were measured in cell culture supernatant. Results: Transcriptomic analysis to compare term and preterm cells revealed 2,321 genes differentially expressed in ex vivo samples and 865 genes differentially expressed in cultured basal cell samples. Over one third of differentially expressed genes were related to host immunity, with interferon signalling pathways dominating the pathway enrichment analysis and IRF1 identified as a hub gene. Corroboration of disrupted interferon release showed that concentrations of IFN-α2 were below measurable limits in term samples but elevated in preterm samples [19.4 (76.7) pg/ml/µg protein, p = 0.03]. IFN-γ production was significantly higher in preterm samples [3.3 (1.5) vs. 9.4 (17.7) pg/ml/µg protein; p = 0.01] as was IFN-β [7.8 (2.5) vs. 13.6 (19.5) pg/ml/µg protein, p = 0.01]. Conclusion: Host immunity may be compromised in the preterm nasal airway epithelium in early life. Altered immune responses may lead to cycles of repeated infections, causing persistent inflammation and tissue damage which can have significant impacts on long-term respiratory function.
Collapse
Affiliation(s)
- Denby J. Evans
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Nedlands, WA, Australia
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute and The University of Western Australia, Crawley, WA, Australia
- School of Population Health, Curtin University, Bentley, WA, Australia
| | - Jessica K. Hillas
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Nedlands, WA, Australia
| | - Thomas Iosifidis
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Nedlands, WA, Australia
- School of Population Health, Curtin University, Bentley, WA, Australia
- Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia and Harry Perkins Institute of Medical Research, Nedlands, WA, Australia
| | - Shannon J. Simpson
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Nedlands, WA, Australia
- School of Allied Health, Curtin University, Bentley, WA, Australia
| | - Anthony Kicic
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Nedlands, WA, Australia
- School of Population Health, Curtin University, Bentley, WA, Australia
- Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia and Harry Perkins Institute of Medical Research, Nedlands, WA, Australia
- Department of Respiratory and Sleep Medicine, Perth Children’s Hospital, Nedlands, WA, Australia
| | - Patricia Agudelo-Romero
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Nedlands, WA, Australia
- School of Molecular Science, University of Western Australia, Nedlands, WA, Australia
- European Virus Bioinformatics Centre, Jena, Thuringia, Germany
| |
Collapse
|
5
|
Elbialy A, Kappala D, Desai D, Wang P, Fadiel A, Wang SJ, Makary MS, Lenobel S, Sood A, Gong M, Dason S, Shabsigh A, Clinton S, Parwani AV, Putluri N, Shvets G, Li J, Liu X. Patient-Derived Conditionally Reprogrammed Cells in Prostate Cancer Research. Cells 2024; 13:1005. [PMID: 38920635 PMCID: PMC11201841 DOI: 10.3390/cells13121005] [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: 03/17/2024] [Revised: 05/31/2024] [Accepted: 06/03/2024] [Indexed: 06/27/2024] Open
Abstract
Prostate cancer (PCa) remains a leading cause of mortality among American men, with metastatic and recurrent disease posing significant therapeutic challenges due to a limited comprehension of the underlying biological processes governing disease initiation, dormancy, and progression. The conventional use of PCa cell lines has proven inadequate in elucidating the intricate molecular mechanisms driving PCa carcinogenesis, hindering the development of effective treatments. To address this gap, patient-derived primary cell cultures have been developed and play a pivotal role in unraveling the pathophysiological intricacies unique to PCa in each individual, offering valuable insights for translational research. This review explores the applications of the conditional reprogramming (CR) cell culture approach, showcasing its capability to rapidly and effectively cultivate patient-derived normal and tumor cells. The CR strategy facilitates the acquisition of stem cell properties by primary cells, precisely recapitulating the human pathophysiology of PCa. This nuanced understanding enables the identification of novel therapeutics. Specifically, our discussion encompasses the utility of CR cells in elucidating PCa initiation and progression, unraveling the molecular pathogenesis of metastatic PCa, addressing health disparities, and advancing personalized medicine. Coupled with the tumor organoid approach and patient-derived xenografts (PDXs), CR cells present a promising avenue for comprehending cancer biology, exploring new treatment modalities, and advancing precision medicine in the context of PCa. These approaches have been used for two NCI initiatives (PDMR: patient-derived model repositories; HCMI: human cancer models initiatives).
Collapse
Affiliation(s)
- Abdalla Elbialy
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (A.E.)
- Computational Oncology Unit, The University of Chicago Comprehensive Cancer Center, 900 E 57th Street, KCBD Bldg., STE 4144, Chicago, IL 60637, USA
| | - Deepthi Kappala
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (A.E.)
| | - Dhruv Desai
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (A.E.)
| | - Peng Wang
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (A.E.)
| | - Ahmed Fadiel
- Computational Oncology Unit, The University of Chicago Comprehensive Cancer Center, 900 E 57th Street, KCBD Bldg., STE 4144, Chicago, IL 60637, USA
| | - Shang-Jui Wang
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (A.E.)
- Department of Radiation Oncology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Mina S. Makary
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (A.E.)
- Division of Vascular and Interventional Radiology, Department of Radiology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Scott Lenobel
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (A.E.)
- Division of Musculoskeletal Imaging, Department of Radiology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Akshay Sood
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (A.E.)
- Department of Urology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Michael Gong
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (A.E.)
- Department of Urology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Shawn Dason
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (A.E.)
- Department of Urology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Ahmad Shabsigh
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (A.E.)
- Department of Urology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Steven Clinton
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (A.E.)
| | - Anil V. Parwani
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (A.E.)
- Departments of Pathology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Nagireddy Putluri
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Gennady Shvets
- School of Applied and Engineering Physics, Cornell University, Ithaca, NY 14850, USA
| | - Jenny Li
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (A.E.)
- Departments of Pathology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Xuefeng Liu
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (A.E.)
- Departments of Pathology, Urology, and Radiation Oncology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
6
|
Zhang G, Le Souëf P. The influence of modern living conditions on the human microbiome and potential therapeutic opportunities for allergy prevention. World Allergy Organ J 2024; 17:100857. [PMID: 38235259 PMCID: PMC10793171 DOI: 10.1016/j.waojou.2023.100857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 12/01/2023] [Accepted: 12/09/2023] [Indexed: 01/19/2024] Open
Abstract
Modern living conditions and the recent surge in global urbanization have transformed the human microbiome. This transformation is believed to be a significant factor in the recent spike of common chronic inflammatory diseases like asthma and allergies worldwide, evident in both developed and developing nations. Immigrants from less developed regions who settle in highly urbanized and affluent areas present an ideal demographic for research. Investigating immigrant populations can yield valuable insights, particularly when studying microbiome changes that occur as individuals transition from areas with low asthma prevalence to regions with a high prevalence of the condition. The application of prebiotics and probiotics as potential treatments for asthma and allergies faces challenges. This is due to the complex interplay of numerous factors that contribute to their aetiology. Exploring the interaction between the human microbiome and potential epigenetic changes in specific populations, such as immigrants adapting to new, urbanized environments, may offer crucial insights. Such research could underscore the role of prebiotics and probiotics in preventing allergic conditions. Recognizing the changes in the human microbiome in the context of a Western/modern environment might be essential in addressing the increasing prevalence of allergic diseases. Persistent research in this domain is pivotal for devising effective interventions such as dietary supplementation with prebiotics and probiotics.
Collapse
Affiliation(s)
- Guicheng Zhang
- School of Population Health, Curtin University, Perth, 6102, Western Australia, Australia
- School of Biomedical Sciences, The University of Western Australia, Perth, Western Australia 6008, Australia
| | - Peter Le Souëf
- School of Population Health, Curtin University, Perth, 6102, Western Australia, Australia
- School of Medicine, The University of Western Australia, Perth, Western Australia 6008, Australia
| |
Collapse
|
7
|
Awatade NT, Reid AT, Nichol KS, Budden KF, Veerati PC, Pathinayake PS, Grainge CL, Hansbro PM, Wark PAB. Comparison of commercially available differentiation media on cell morphology, function, and anti-viral responses in conditionally reprogrammed human bronchial epithelial cells. Sci Rep 2023; 13:11200. [PMID: 37433796 DOI: 10.1038/s41598-023-37828-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 06/28/2023] [Indexed: 07/13/2023] Open
Abstract
Primary air liquid interface (ALI) cultures of bronchial epithelial cells are used extensively to model airway responses. A recent advance is the development of conditional reprogramming that enhances proliferative capability. Several different media and protocols are utilized, yet even subtle differences may influence cellular responses. We compared the morphology and functional responses, including innate immune responses to rhinovirus infection in conditionally reprogrammed primary bronchial epithelial cells (pBECs) differentiated using two commonly used culture media. pBECs collected from healthy donors (n = 5) were CR using g-irradiated 3T3 fibroblasts and Rho Kinase inhibitor. CRpBECs were differentiated at ALI in either PneumaCult (PN-ALI) or bronchial epithelial growth medium (BEGM)-based differentiation media (BEBM:DMEM, 50:50, Lonza)-(AB-ALI) for 28 days. Transepithelial electrical resistance (TEER), immunofluorescence, histology, cilia activity, ion channel function, and expression of cell markers were analyzed. Viral RNA was assessed by RT-qPCR and anti-viral proteins quantified by LEGENDplex following Rhinovirus-A1b infection. CRpBECs differentiated in PneumaCult were smaller and had a lower TEER and cilia beat frequency compared to BEGM media. PneumaCult media cultures exhibited increased FOXJ1 expression, more ciliated cells with a larger active area, increased intracellular mucins, and increased calcium-activated chloride channel current. However, there were no significant changes in viral RNA or host antiviral responses. There are distinct structural and functional differences in pBECs cultured in the two commonly used ALI differentiation media. Such factors need to be taken into consideration when designing CRpBECs ALI experiments for specific research questions.
Collapse
Affiliation(s)
- Nikhil T Awatade
- School of Medicine and Public Health, University of Newcastle, Callaghan, NSW, Australia.
- Immune Health Program, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, NSW, Australia.
| | - Andrew T Reid
- School of Medicine and Public Health, University of Newcastle, Callaghan, NSW, Australia
- Asthma and Breathing Research Program, Hunter Medical Research Institute University of Newcastle, New Lambton Heights, NSW, Australia
| | - Kristy S Nichol
- School of Medicine and Public Health, University of Newcastle, Callaghan, NSW, Australia
- Immune Health Program, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, NSW, Australia
| | - Kurtis F Budden
- School of Medicine and Public Health, University of Newcastle, Callaghan, NSW, Australia
- Immune Health Program, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, NSW, Australia
| | - Punnam Chander Veerati
- School of Medicine and Public Health, University of Newcastle, Callaghan, NSW, Australia
- Asthma and Breathing Research Program, Hunter Medical Research Institute University of Newcastle, New Lambton Heights, NSW, Australia
| | - Prabuddha S Pathinayake
- School of Medicine and Public Health, University of Newcastle, Callaghan, NSW, Australia
- Immune Health Program, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, NSW, Australia
| | - Christopher L Grainge
- School of Medicine and Public Health, University of Newcastle, Callaghan, NSW, Australia
- Asthma and Breathing Research Program, Hunter Medical Research Institute University of Newcastle, New Lambton Heights, NSW, Australia
- Dept of Respiratory and Sleep Medicine, John Hunter Hospital, New Lambton Heights, NSW, Australia
| | - Philip M Hansbro
- Immune Health Program, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, NSW, Australia
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, NSW, Australia
| | - Peter A B Wark
- School of Medicine and Public Health, University of Newcastle, Callaghan, NSW, Australia.
- Immune Health Program, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, NSW, Australia.
- Asthma and Breathing Research Program, Hunter Medical Research Institute University of Newcastle, New Lambton Heights, NSW, Australia.
- Dept of Respiratory and Sleep Medicine, John Hunter Hospital, New Lambton Heights, NSW, Australia.
| |
Collapse
|
8
|
Dai H, Zheng Q, Shan S, Lei Y, Zhang J, Zhong C, Xu Y, Liu Z, Ren T. A novel type of serum‑free medium for culturing human airway epithelium cells. Exp Ther Med 2023; 25:239. [PMID: 37114176 PMCID: PMC10126810 DOI: 10.3892/etm.2023.11938] [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: 12/01/2022] [Accepted: 03/16/2023] [Indexed: 04/29/2023] Open
Abstract
The present study aimed to evaluate the ability of a novel serum-free medium (SFM) to culture human airway epithelium cells (hAECs). hAECs were cultured in the novel SFM as the experimental group in the PneumaCult-Ex medium and Dulbecco's modified eagle medium (DMEM) and fetal bovine serum (FBS) as the control groups. Cell morphology, proliferative capacity, differentiation capacity and expression levels of basal cell markers were assessed accordingly in both culture systems. Optical microscope photos of hAECs were collected for cell morphology assessment. Cell Counting Kit-8 assay was conducted to assess the proliferation ability, and an air-liquid interface (ALI) assay was conducted to assess the differentiation capacity. Markers for proliferating basal and differentiated cells were relatively identified by immunohistochemical and immunofluorescent analysis. The results show that whether grown in the novel SFM or Ex medium, hAECs exhibited similar morphology at every passage, whereas cells could hardly form colonies in the DMEM + FBS group. Cells typically exhibited cobblestone shape, while a proportion of them in the novel SFM at late passage exhibited a larger shape. White vesicles appeared in the cytoplasm of some control cells at the later stage of culture. Basal cell markers (P63+KRT5+KI67+CC10-) for proliferating ability were found in the hAECs cultured by the novel SFM and Ex medium. hAECs at passage 3 cultured in the novel SFM and Ex medium both had the capacity to differentiate into ciliated cells (acetylated tubulin+), goblet cells (MUC5AC+) and club cells (CC10+) in the ALI culture assay. In conclusion, the novel SFM was capable of culturing hAECs. The hAECs cultured by the novel SFM could proliferate and differentiate in vitro. The novel SFM does not change the morphological characteristics or biomarkers of hAECs. The novel SFM has the potential for the amplification of hAECs for scientific research and clinical application.
Collapse
Affiliation(s)
- Haotian Dai
- Department of Respiratory Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, P.R. China
| | - Qi Zheng
- Department of Respiratory Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, P.R. China
| | - Shan Shan
- Department of Respiratory Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, P.R. China
| | - Yuqiong Lei
- Department of Respiratory Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, P.R. China
| | - Jingyuan Zhang
- Department of Respiratory Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, P.R. China
| | - Cheng Zhong
- Department of Respiratory Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, P.R. China
| | - Yongle Xu
- Department of Respiratory Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, P.R. China
| | - Zeyu Liu
- Department of Respiratory Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, P.R. China
- Correspondence to: Professor Tao Ren or Dr Zeyu Liu, Department of Respiratory Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Xuhui, Shanghai 200233, P.R. China
| | - Tao Ren
- Department of Respiratory Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, P.R. China
- Correspondence to: Professor Tao Ren or Dr Zeyu Liu, Department of Respiratory Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Xuhui, Shanghai 200233, P.R. China
| |
Collapse
|
9
|
Stricker S, Ziegahn N, Karsten M, Boeckel T, Stich-Boeckel H, Maske J, Rugo E, Balazs A, Millar Büchner P, Dang-Heine C, Schriever V, Eils R, Lehmann I, Sander LE, Ralser M, Corman VM, Mall MA, Sawitzki B, Roehmel J. RECAST: Study protocol for an observational study for the understanding of the increased REsilience of Children compared to Adults in SARS-CoV-2 infecTion. BMJ Open 2023; 13:e065221. [PMID: 37068896 PMCID: PMC10111194 DOI: 10.1136/bmjopen-2022-065221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/19/2023] Open
Abstract
INTRODUCTION The SARS-CoV-2 pandemic remains a threat to public health. Soon after its outbreak, it became apparent that children are less severely affected. Indeed, opposing clinical manifestations between children and adults are observed for other infections. The SARS-CoV-2 outbreak provides the unique opportunity to study the underlying mechanisms. This protocol describes the methods of an observational study that aims to characterise age dependent differences in immune responses to primary respiratory infections using SARS-CoV-2 as a model virus and to assess age differences in clinical outcomes including lung function. METHODS AND ANALYSIS The study aims to recruit at least 120 children and 60 adults that are infected with SARS-CoV-2 and collect specimen for a multiomics analysis, including single cell RNA sequencing of nasal epithelial cells and peripheral blood mononuclear cells, mass cytometry of whole blood samples and nasal cells, mass spectrometry-based serum and plasma proteomics, nasal epithelial cultures with functional in vitro analyses, SARS-CoV-2 antibody testing, sequencing of the viral genome and lung function testing. Data obtained from this multiomics approach are correlated with medical history and clinical data. Recruitment started in October 2020 and is ongoing. ETHICS AND DISSEMINATION The study was reviewed and approved by the Ethics Committee of Charité - Universitätsmedizin Berlin (EA2/066/20). All collected specimens are stored in the central biobank of Charité - Universitätsmedizin Berlin and are made available to all participating researchers and on request. TRIAL REGISTRATION NUMBER DRKS00025715, pre-results publication.
Collapse
Affiliation(s)
- Sebastian Stricker
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Niklas Ziegahn
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Martin Karsten
- Karsten, Rugo, Wagner, Paediatric Practice, Berlin, Germany
| | - Thomas Boeckel
- Boeckel, Haverkaemper, Paediatric Practice and Practice for Paediatric Cardiology, Berlin, Germany
| | | | - Jakob Maske
- Maske, Pankok, Paediatric Practice, Berlin, Germany
| | - Evelyn Rugo
- Karsten, Rugo, Wagner, Paediatric Practice, Berlin, Germany
| | - Anita Balazs
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Pamela Millar Büchner
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Chantip Dang-Heine
- Clinical Study Center (CSC), Berlin Institute of Health at Charité, Berlin, Germany
| | - Valentin Schriever
- Department of Paediatric Neurology, Charité Universitätsmedizin Berlin, Berlin, Germany
- Center for Chronically Sick Children (Sozialpädiatrisches Zentrum, SPZ), Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Roland Eils
- Center for Digital Health, Berlin Institute of Health at Charité, Berlin, Germany
- Molecular Epidemiology Unit, Berlin Institute of Health at Charité, Berlin, Germany
| | - Irina Lehmann
- Center for Digital Health, Berlin Institute of Health at Charité, Berlin, Germany
- German Center for Lung Research, Giessen, Germany
| | - Leif E Sander
- Department of Infectious Diseases and Respiratory Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Markus Ralser
- Department of Biochemistry, Charité Universitätsmedizin Berlin, Berlin, Germany
- Molecular Biology of Metabolism Laboratory, The Francis Crick Institute, London, UK
| | - Victor M Corman
- Institute of Virology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Marcus A Mall
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
- German Center for Lung Research, Giessen, Germany
| | - Birgit Sawitzki
- Berlin Institute of Health, Berlin, Germany
- Institute of Medical Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Jobst Roehmel
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
10
|
Antunes KH, Singanayagam A, Williams L, Faiez TS, Farias A, Jackson MM, Faizi FK, Aniscenko J, Kebadze T, Chander Veerati P, Wood L, Bartlett NW, Duarte de Souza AP, Johnston SL. Airway-delivered short-chain fatty acid acetate boosts antiviral immunity during rhinovirus infection. J Allergy Clin Immunol 2023; 151:447-457.e5. [PMID: 36216081 DOI: 10.1016/j.jaci.2022.09.026] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 08/03/2022] [Accepted: 09/09/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Microbiota are recognized to play a major role in regulation of immunity through release of immunomodulatory metabolites such as short-chain fatty acids (SCFAs). Rhinoviruses (RVs) induce upper respiratory tract illnesses and precipitate exacerbations of asthma and chronic obstructive pulmonary disease through poorly understood mechanisms. Local interactions between SCFAs and antiviral immune responses in the respiratory tract have not been previously investigated. OBJECTIVE We sought to investigate whether pulmonary metabolite manipulation through lung-delivered administration of SCFAs can modulate antiviral immunity to RV infection. METHODS We studied the effects of intranasal administration of the SCFAs acetate, butyrate, and propionate on basal expression of antiviral signatures, and of acetate in a mouse model of RV infection and in RV-infected lung epithelial cell lines. We additionally assessed the effects of acetate, butyrate, and propionate on RV infection in differentiated human primary bronchial epithelial cells. RESULTS Intranasal acetate administration induced basal upregulation of IFN-β, an effect not observed with other SCFAs. Butyrate induced RIG-I expression. Intranasal acetate treatment of mice increased interferon-stimulated gene and IFN-λ expression during RV infection and reduced lung virus loads at 8 hours postinfection. Acetate ameliorated virus-induced proinflammatory responses with attenuated pulmonary mucin and IL-6 expression observed at day 4 and 6 postinfection. This interferon-enhancing effect of acetate was confirmed in human bronchial and alveolar epithelial cell lines. In differentiated primary bronchial epithelial cells, butyrate treatment better modulated IFN-β and IFN-λ gene expression during RV infection. CONCLUSIONS SCFAs augment antiviral immunity and reduce virus load and proinflammatory responses during RV infection.
Collapse
Affiliation(s)
- Krist Helen Antunes
- Laboratory of Clinical and Experimental Immunology - Pontifical Catholic University of Rio Grande do Sul, Porto Alegre; National Heart and Lung Institute and, Department of Infectious Disease, Imperial College London, London
| | - Aran Singanayagam
- Centre for Molecular Bacteriology and Infection, Department of Infectious Disease, Imperial College London, London
| | - Lily Williams
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Newcastle
| | - Tasnim Syakirah Faiez
- Centre for Molecular Bacteriology and Infection, Department of Infectious Disease, Imperial College London, London
| | - Ana Farias
- National Heart and Lung Institute and, Department of Infectious Disease, Imperial College London, London
| | - Millie M Jackson
- Centre for Molecular Bacteriology and Infection, Department of Infectious Disease, Imperial College London, London
| | - Fatima K Faizi
- Centre for Molecular Bacteriology and Infection, Department of Infectious Disease, Imperial College London, London
| | - Julia Aniscenko
- National Heart and Lung Institute and, Department of Infectious Disease, Imperial College London, London
| | - Tatiana Kebadze
- National Heart and Lung Institute and, Department of Infectious Disease, Imperial College London, London
| | | | - Lisa Wood
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Newcastle
| | - Nathan W Bartlett
- National Heart and Lung Institute and, Department of Infectious Disease, Imperial College London, London; School of Biomedical Sciences and Pharmacy, The University of Newcastle, Newcastle
| | - Ana Paula Duarte de Souza
- Laboratory of Clinical and Experimental Immunology - Pontifical Catholic University of Rio Grande do Sul, Porto Alegre; National Heart and Lung Institute and, Department of Infectious Disease, Imperial College London, London.
| | - Sebastian L Johnston
- National Heart and Lung Institute and, Department of Infectious Disease, Imperial College London, London; Asthma UK Centre in Allergic Mechanisms of Asthma, London.
| |
Collapse
|
11
|
Landwehr KR, Hillas J, Mead-Hunter R, King A, O'Leary RA, Kicic A, Mullins BJ, Larcombe AN. Biodiesel feedstock determines exhaust toxicity in 20% biodiesel: 80% mineral diesel blends. CHEMOSPHERE 2023; 310:136873. [PMID: 36252896 DOI: 10.1016/j.chemosphere.2022.136873] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 10/04/2022] [Accepted: 10/11/2022] [Indexed: 06/16/2023]
Abstract
To address climate change concerns, and reduce the carbon footprint caused by fossil fuel use, it is likely that blend ratios of renewable biodiesel with commercial mineral diesel fuel will steadily increase, resulting in biodiesel use becoming more widespread. Exhaust toxicity of unblended biodiesels changes depending on feedstock type, however the effect of feedstock on blended fuels is less well known. The aim of this study was to assess the impact of biodiesel feedstock on exhaust toxicity of 20% blended biodiesel fuels (B20). Primary human airway epithelial cells were exposed to exhaust diluted 1/15 with air from an engine running on conventional ultra-low sulfur diesel (ULSD) or 20% blends of soy, canola, waste cooking oil (WCO), tallow, palm or cottonseed biodiesel in diesel. Physico-chemical exhaust properties were compared between fuels and the post-exposure effect of exhaust on cellular viability and media release was assessed 24 h later. Exhaust properties changed significantly between all fuels with cottonseed B20 being the most different to both ULSD and its respective unblended biodiesel. Exposure to palm B20 resulted in significantly decreased cellular viability (96.3 ± 1.7%; p < 0.01) whereas exposure to soy B20 generated the greatest number of changes in mediator release (including IL-6, IL-8 and TNF-α, p < 0.05) when compared to air exposed controls, with palm B20 and tallow B20 closely following. In contrast, canola B20 and WCO B20 were the least toxic with only mediators G-CSF and TNF-α being significantly increased. Therefore, exposure to palm B20, soy B20 and tallow B20 were found to be the most toxic and exposure to canola B20 and WCO B20 the least. The top three most toxic and the bottom three least toxic B20 fuels are consistent with their unblended counterparts, suggesting that feedstock type greatly impacts exhaust toxicity, even when biodiesel only comprises 20% of the fuel.
Collapse
Affiliation(s)
- Katherine R Landwehr
- Occupation, Environment and Safety, School of Population Health, Curtin University, PO Box U1987, Perth, 6845, Western Australia, Australia; Respiratory Environmental Health, Wal-yan Respiratory Research Centre, Telethon Kids Institute, Perth Children's Hospital, Nedlands, Perth, 6009, Western Australia, Australia.
| | - Jessica Hillas
- Respiratory Environmental Health, Wal-yan Respiratory Research Centre, Telethon Kids Institute, Perth Children's Hospital, Nedlands, Perth, 6009, Western Australia, Australia
| | - Ryan Mead-Hunter
- Occupation, Environment and Safety, School of Population Health, Curtin University, PO Box U1987, Perth, 6845, Western Australia, Australia
| | - Andrew King
- Fluid Dynamics Research Group, School of Civil and Mechanical Engineering, Curtin University, Perth, Western Australia, Australia
| | - Rebecca A O'Leary
- Department of Primary Industries and Regional Development, Perth, 6151, Western Australia, Australia
| | - Anthony Kicic
- Occupation, Environment and Safety, School of Population Health, Curtin University, PO Box U1987, Perth, 6845, Western Australia, Australia; Respiratory Environmental Health, Wal-yan Respiratory Research Centre, Telethon Kids Institute, Perth Children's Hospital, Nedlands, Perth, 6009, Western Australia, Australia; Department of Respiratory and Sleep Medicine, Perth Children's Hospital, Nedlands, Perth, 6009, Western Australia, Australia; Centre for Cell Therapy and Regenerative Medicine, The University of Western Australia, Perth, 6009, Western Australia, Australia
| | - Benjamin J Mullins
- Occupation, Environment and Safety, School of Population Health, Curtin University, PO Box U1987, Perth, 6845, Western Australia, Australia
| | - Alexander N Larcombe
- Occupation, Environment and Safety, School of Population Health, Curtin University, PO Box U1987, Perth, 6845, Western Australia, Australia; Respiratory Environmental Health, Wal-yan Respiratory Research Centre, Telethon Kids Institute, Perth Children's Hospital, Nedlands, Perth, 6009, Western Australia, Australia
| |
Collapse
|
12
|
Laucirica DR, Stick SM, Garratt LW, Kicic A. Bacteriophage: A new therapeutic player to combat neutrophilic inflammation in chronic airway diseases. Front Med (Lausanne) 2022; 9:1069929. [PMID: 36590945 PMCID: PMC9794625 DOI: 10.3389/fmed.2022.1069929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 11/29/2022] [Indexed: 12/15/2022] Open
Abstract
Persistent respiratory bacterial infections are a clinical burden in several chronic inflammatory airway diseases and are often associated with neutrophil infiltration into the lungs. Following recruitment, dysregulated neutrophil effector functions such as increased granule release and formation of neutrophil extracellular traps (NETs) result in damage to airway tissue, contributing to the progression of lung disease. Bacterial pathogens are a major driver of airway neutrophilic inflammation, but traditional management of infections with antibiotic therapy is becoming less effective as rates of antimicrobial resistance rise. Bacteriophages (phages) are now frequently identified as antimicrobial alternatives for antimicrobial resistant (AMR) airway infections. Despite growing recognition of their bactericidal function, less is known about how phages influence activity of neutrophils recruited to sites of bacterial infection in the lungs. In this review, we summarize current in vitro and in vivo findings on the effects of phage therapy on neutrophils and their inflammatory mediators, as well as mechanisms of phage-neutrophil interactions. Understanding these effects provides further validation of their safe use in humans, but also identifies phages as a targeted neutrophil-modulating therapeutic for inflammatory airway conditions.
Collapse
Affiliation(s)
- Daniel R. Laucirica
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, The University of Western Australia, Nedlands, WA, Australia
| | - Stephen M. Stick
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, The University of Western Australia, Nedlands, WA, Australia
- Department of Respiratory and Sleep Medicine, Perth Children’s Hospital, Nedlands, WA, Australia
- Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia and Harry Perkins Institute of Medical Research, Nedlands, WA, Australia
| | - Luke W. Garratt
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, The University of Western Australia, Nedlands, WA, Australia
| | - Anthony Kicic
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, The University of Western Australia, Nedlands, WA, Australia
- Department of Respiratory and Sleep Medicine, Perth Children’s Hospital, Nedlands, WA, Australia
- Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia and Harry Perkins Institute of Medical Research, Nedlands, WA, Australia
- School of Population Health, Curtin University, Bentley, WA, Australia
| |
Collapse
|
13
|
Kelly NA, Shontz KM, Bergman M, Manning AM, Reynolds SD, Chiang T. Biobanked tracheal basal cells retain the capacity to differentiate. Laryngoscope Investig Otolaryngol 2022; 7:2119-2125. [PMID: 36544928 PMCID: PMC9764751 DOI: 10.1002/lio2.925] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 08/19/2022] [Accepted: 09/07/2022] [Indexed: 12/24/2022] Open
Abstract
Objective While airway epithelial biorepositories have established roles in the study of bronchial progenitor stem (basal) cells, the utility of a bank of tracheal basal cells from pediatric patients, who have or are suspected of having an airway disease, has not been established. In vitro study of these cells can enhance options for tracheal restoration, graft design, and disease modeling. Development of a functional epithelium in these settings is a key measure. The aim of this study was the creation a tracheal basal cell biorepository and assessment of recovered cells. Methods Pediatric patients undergoing bronchoscopy were identified and endotracheal brush (N = 29) biopsies were collected. Cells were cultured using the modified conditional reprogramming culture (mCRC) method. Samples producing colonies by day 14 were passaged and cryopreserved. To explore differentiation potential, cells were thawed and differentiated using the air-liquid interface (ALI) method. Results No adverse events were associated with biopsy collection. Of 29 brush biopsies, 16 (55%) were successfully cultured to passage 1/cryopreserved. Samples with higher initial cell yields were more likely to achieve this benchmark. Ten unique donors were then thawed for analysis of differentiation. The average age was 2.2 ± 2.2 years with five donors (50%) having laryngotracheal pathology. Nine donors (90%) demonstrated differentiation capacity at 21 days of culture, as indicated by detection of ciliated cells (ACT+) and mucous cells (MUC5B+). Conclusion Pediatric tracheal basal cells can be successfully collected and cryopreserved. Recovered cells retain the ability to differentiate into epithelial cell types in vitro. Level of Evidence Level 3.
Collapse
Affiliation(s)
- Natalie A. Kelly
- Department of OtolaryngologyNationwide Children's HospitalColumbusOhioUSA
| | - Kimberly M. Shontz
- Center for Regenerative MedicineAbigail Wexner Research Institute at Nationwide Children's HospitalColumbusOhioUSA
| | - Maxwell Bergman
- Department of Otolaryngology‐Head and Neck SurgeryThe Ohio State Wexner Medical CenterColumbusOhioUSA
| | - Amy M. Manning
- Department of OtolaryngologyNationwide Children's HospitalColumbusOhioUSA
- Department of Otolaryngology‐Head and Neck SurgeryThe Ohio State Wexner Medical CenterColumbusOhioUSA
| | - Susan D. Reynolds
- Center for Perinatal MedicineAbigail Wexner Research Institute at Nationwide Children's HospitalColumbusOhioUSA
| | - Tendy Chiang
- Department of OtolaryngologyNationwide Children's HospitalColumbusOhioUSA
- Center for Regenerative MedicineAbigail Wexner Research Institute at Nationwide Children's HospitalColumbusOhioUSA
- Department of Otolaryngology‐Head and Neck SurgeryThe Ohio State Wexner Medical CenterColumbusOhioUSA
| |
Collapse
|
14
|
Landwehr KR, Nabi MN, Rasul MG, Kicic A, Mullins BJ. Biodiesel Exhaust Toxicity with and without Diethylene Glycol Dimethyl Ether Fuel Additive in Primary Airway Epithelial Cells Grown at the Air-Liquid Interface. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2022; 56:14640-14648. [PMID: 36177943 DOI: 10.1021/acs.est.2c03806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Biodiesel usage is increasing steadily worldwide as the push for renewable fuel sources increases. The increased oxygen content in biodiesel fuel is believed to cause decreased particulate matter (PM) and increased nitrous oxides within its exhaust. The addition of fuel additives to further increase the oxygen content may contribute to even further benefits in exhaust composition. The aim of this study was to assess the toxicity of 10% (v/v) diethylene glycol dimethyl ether (DGDME) added as a biodiesel fuel additive. Primary human airway epithelial cells were grown at the air-liquid interface and exposed to diluted exhaust from an engine running on either grapeseed, bran, or coconut biodiesel or the same three biodiesels with 10% (v/v) DGDME added to them; mineral diesel and air were used as controls. Exhaust properties, culture permeability, epithelial cell damage, and IL-6 and IL-8 release were measured postexposure. The fuel additive DGDME caused a decrease in PM and nitrous oxide concentrations. However, exhaust exposure with DGDME also caused decreased permeability, increased epithelial cell damage, and increased release of IL-6 and IL-8 (p < 0.05). Despite the fuel additive having beneficial effects on the exhaust properties of the biodiesel, it was found to be more toxic.
Collapse
Affiliation(s)
- Katherine R Landwehr
- Occupation, Environment & Safety, School of Population Health, Curtin University, Perth, Western Australia 6102, Australia
- Respiratory Environmental Health, Telethon Kids Institute, Perth, Western Australia 6009, Australia
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Perth, Western Australia 6009, Australia
| | - Md Nurun Nabi
- School of Engineering and Technology, Fuel and Energy Research Group, Central Queensland University, Perth, Western Australia 6000, Australia
| | - Mohammad G Rasul
- School of Engineering and Technology, Fuel and Energy Research Group, Central Queensland University, Rockhampton, Queensland 4701, Australia
| | - Anthony Kicic
- Occupation, Environment & Safety, School of Population Health, Curtin University, Perth, Western Australia 6102, Australia
- Respiratory Environmental Health, Telethon Kids Institute, Perth, Western Australia 6009, Australia
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Perth, Western Australia 6009, Australia
- Department of of Respiratory and Sleep Medicine, Perth Children's Hospital, Perth, Western Australia 6009, Australia
- Centre for Cell Therapy and Regenerative Medicine, The University of Western Australia, Perth, Western Australia 6009, Australia
| | - Benjamin J Mullins
- Occupation, Environment & Safety, School of Population Health, Curtin University, Perth, Western Australia 6102, Australia
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Perth, Western Australia 6009, Australia
- St. John of God Hospital, Subiaco, Western Australia 6008, Australia
| |
Collapse
|
15
|
Guo D, Zhang L, Wang X, Zheng J, Lin S. Establishment methods and research progress of livestock and poultry immortalized cell lines: A review. Front Vet Sci 2022; 9:956357. [PMID: 36118350 PMCID: PMC9478797 DOI: 10.3389/fvets.2022.956357] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 08/10/2022] [Indexed: 11/13/2022] Open
Abstract
An infinite cell line is one of the most favored experimental tools and plays an irreplaceable role in cell-based biological research. Primary cells from normal animal tissues undergo a limited number of divisions and subcultures in vitro before they enter senescence and die. On the contrary, an infinite cell line is a population of non-senescent cells that could proliferate indefinitely in vitro under the stimulation of external factors such as physicochemical stimulation, virus infection, or transfer of immortality genes. Cell immortalization is the basis for establishing an infinite cell line, and previous studies have found that methods to obtain immortalized cells mainly included physical and chemical stimulations, heterologous expression of viral oncogenes, increased telomerase activity, and spontaneous formation. However, some immortalized cells do not necessarily proliferate permanently even though they can extend their lifespan compared with primary cells. An infinite cell line not only avoids the complicated process of collecting primary cell, it also provides a convenient and reliable tool for studying scientific problems in biology. At present, how to establish a stable infinite cell line to maximize the proliferation of cells while maintaining the normal function of cells is a hot issue in the biological community. This review briefly introduces the methods of cell immortalization, discusses the related progress of establishing immortalized cell lines in livestock and poultry, and compares the characteristics of several methods, hoping to provide some ideas for generating new immortalized cell lines.
Collapse
|
16
|
Lv X, Dong M, Tang W, Qin J, Wang W, Li M, Teng F, Yi L, Dong J, Wei Y. Ferroptosis, novel therapeutics in asthma. Biomed Pharmacother 2022; 153:113516. [DOI: 10.1016/j.biopha.2022.113516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 08/02/2022] [Accepted: 08/03/2022] [Indexed: 11/26/2022] Open
|
17
|
Landwehr KR, Hillas J, Mead-Hunter R, King A, O'Leary RA, Kicic A, Mullins BJ, Larcombe AN. Toxicity of different biodiesel exhausts in primary human airway epithelial cells grown at air-liquid interface. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 832:155016. [PMID: 35381248 DOI: 10.1016/j.scitotenv.2022.155016] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/25/2022] [Accepted: 03/30/2022] [Indexed: 06/14/2023]
Abstract
Biodiesel is created through the transesterification of fats/oils and its usage is increasing worldwide as global warming concerns increase. Biodiesel fuel properties change depending on the feedstock used to create it. The aim of this study was to assess the different toxicological properties of biodiesel exhausts created from different feedstocks using a complex 3D air-liquid interface (ALI) model that mimics the human airway. Primary human airway epithelial cells were grown at ALI until full differentiation was achieved. Cells were then exposed to 1/20 diluted exhaust from an engine running on Diesel (ULSD), pure or 20% blended Canola biodiesel and pure or 20% blended Tallow biodiesel, or Air for control. Exhaust was analysed for various physio-chemical properties and 24-h after exposure, ALI cultures were assessed for permeability, protein release and mediator response. All measured exhaust components were within industry safety standards. ULSD contained the highest concentrations of various combustion gases. We found no differences in terms of particle characteristics for any of the tested exhausts, likely due to the high dilution used. Exposure to Tallow B100 and B20 induced increased permeability in the ALI culture and the greatest increase in mediator response in both the apical and basal compartments. In contrast, Canola B100 and B20 did not impact permeability and induced the smallest mediator response. All exhausts but Canola B20 induced increased protein release, indicating epithelial damage. Despite the concentrations of all exhausts used in this study meeting industry safety regulations, we found significant toxic effects. Tallow biodiesel was found to be the most toxic of the tested fuels and Canola the least, both for blended and pure biodiesel fuels. This suggests that the feedstock biodiesel is made from is crucial for the resulting health effects of exhaust exposure, even when not comprising the majority of fuel composition.
Collapse
Affiliation(s)
- Katherine R Landwehr
- Occupation, Environment and Safety, School of Population Health, Curtin University, PO Box U1987, Perth 6845, Western Australia, Australia; Respiratory Environmental Health, Wal-yan Respiratory Research Centre, Telethon Kids Institute, Perth Children's Hospital, Nedlands, Perth 6009, Western Australia, Australia.
| | - Jessica Hillas
- Respiratory Environmental Health, Wal-yan Respiratory Research Centre, Telethon Kids Institute, Perth Children's Hospital, Nedlands, Perth 6009, Western Australia, Australia
| | - Ryan Mead-Hunter
- Occupation, Environment and Safety, School of Population Health, Curtin University, PO Box U1987, Perth 6845, Western Australia, Australia
| | - Andrew King
- Fluid Dynamics Research Group, School of Civil and Mechanical Engineering, Curtin University, Perth, Western Australia, Australia
| | - Rebecca A O'Leary
- Department of Primary Industries and Regional Development, Perth 6151, Western Australia, Australia
| | - Anthony Kicic
- Occupation, Environment and Safety, School of Population Health, Curtin University, PO Box U1987, Perth 6845, Western Australia, Australia; Respiratory Environmental Health, Wal-yan Respiratory Research Centre, Telethon Kids Institute, Perth Children's Hospital, Nedlands, Perth 6009, Western Australia, Australia; Department of Respiratory and Sleep Medicine, Perth Children's Hospital, Nedlands, Perth 6009, Western Australia, Australia; Centre for Cell Therapy and Regenerative Medicine, The University of Western Australia, Perth 6009, Western Australia, Australia
| | - Benjamin J Mullins
- Occupation, Environment and Safety, School of Population Health, Curtin University, PO Box U1987, Perth 6845, Western Australia, Australia
| | - Alexander N Larcombe
- Occupation, Environment and Safety, School of Population Health, Curtin University, PO Box U1987, Perth 6845, Western Australia, Australia; Respiratory Environmental Health, Wal-yan Respiratory Research Centre, Telethon Kids Institute, Perth Children's Hospital, Nedlands, Perth 6009, Western Australia, Australia
| |
Collapse
|
18
|
Williams TC, Loo SL, Nichol KS, Reid AT, Veerati PC, Esneau C, Wark PAB, Grainge CL, Knight DA, Vincent T, Jackson CL, Alton K, Shimkets RA, Girkin JL, Bartlett NW. IL-25 blockade augments antiviral immunity during respiratory virus infection. Commun Biol 2022; 5:415. [PMID: 35508632 PMCID: PMC9068710 DOI: 10.1038/s42003-022-03367-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 04/13/2022] [Indexed: 12/12/2022] Open
Abstract
IL-25 is implicated in the pathogenesis of viral asthma exacerbations. However, the effect of IL-25 on antiviral immunity has yet to be elucidated. We observed abundant expression and colocalization of IL-25 and IL-25 receptor at the apical surface of uninfected airway epithelial cells and rhinovirus infection increased IL-25 expression. Analysis of immune transcriptome of rhinovirus-infected differentiated asthmatic bronchial epithelial cells (BECs) treated with an anti-IL-25 monoclonal antibody (LNR125) revealed a re-calibrated response defined by increased type I/III IFN and reduced expression of type-2 immune genes CCL26, IL1RL1 and IL-25 receptor. LNR125 treatment also increased type I/III IFN expression by coronavirus infected BECs. Exogenous IL-25 treatment increased viral load with suppressed innate immunity. In vivo LNR125 treatment reduced IL-25/type 2 cytokine expression and increased IFN-β expression and reduced lung viral load. We define a new immune-regulatory role for IL-25 that directly inhibits virus induced airway epithelial cell innate anti-viral immunity.
Collapse
Affiliation(s)
- Teresa C Williams
- The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Su-Ling Loo
- The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Kristy S Nichol
- The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Andrew T Reid
- The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Punnam C Veerati
- The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Camille Esneau
- The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Peter A B Wark
- The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
- Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, NSW, Australia
| | - Christopher L Grainge
- The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
- Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, NSW, Australia
| | - Darryl A Knight
- The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
- UBC Providence Health Care Research Institute, Vancouver, BC, Canada
- Department of Anaesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| | - Thomas Vincent
- Abeome Corporation/Lanier Biotherapeutics, Athens, GA, USA
| | | | - Kirby Alton
- Abeome Corporation/Lanier Biotherapeutics, Athens, GA, USA
| | | | - Jason L Girkin
- The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Nathan W Bartlett
- The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia.
| |
Collapse
|
19
|
Laucirica DR, Schofield CJ, McLean SA, Margaroli C, Agudelo‐Romero P, Stick SM, Tirouvanziam R, Kicic A, Garratt LW. Pseudomonas aeruginosa
modulates neutrophil granule exocytosis in an
in vitro
model of airway infection. Immunol Cell Biol 2022; 100:352-370. [PMID: 35318736 PMCID: PMC9544492 DOI: 10.1111/imcb.12547] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 03/03/2022] [Accepted: 03/21/2022] [Indexed: 12/24/2022]
Abstract
A population of neutrophils recruited into cystic fibrosis (CF) airways is associated with proteolytic lung damage, exhibiting high expression of primary granule exocytosis marker CD63 and reduced phagocytic receptor CD16. Causative factors for this population are unknown, limiting intervention. Here we present a laboratory model to characterize responses of differentiated airway epithelium and neutrophils following respiratory infection. Pediatric primary airway epithelial cells were cultured at the air–liquid interface, challenged individually or in combination with rhinovirus (RV) and Pseudomonas aeruginosa, then apically washed with medical saline to sample epithelial infection milieus. Cytokine multiplex analysis revealed epithelial antiviral signals, including IP‐10 and RANTES, increased with exclusive RV infection but were diminished if P. aeruginosa was also present. Proinflammatory signals interleukin‐1α and β were dominant in P. aeruginosa infection milieus. Infection washes were also applied to a published model of neutrophil transmigration into the airways. Neutrophils migrating into bacterial and viral–bacterial co‐infection milieus exhibited the in vivo CF phenotype of increased CD63 expression and reduced CD16 expression, while neutrophils migrating into milieus of RV‐infected or uninfected cultures did not. Individually, bacterial products lipopolysaccharide and N‐formylmethionyl‐leucyl‐phenylalanine and isolated cytokine signals only partially activated this phenotype, suggesting that additional soluble factors in the infection microenvironment trigger primary granule release. Findings identify P. aeruginosa as a trigger of acute airway inflammation and neutrophil primary granule exocytosis, underscoring potential roles of airway microbes in prompting this neutrophil subset. Further studies are required to characterize microbes implicated in primary granule release, and identify potential therapeutic targets.
Collapse
Affiliation(s)
- Daniel R Laucirica
- Faculty of Health and Medical Sciences University of Western Australia Nedlands WA Australia
- Wal‐Yan Respiratory Research Centre Telethon Kids Institute University of Western Australia Nedlands WA Australia
| | - Craig J Schofield
- Wal‐Yan Respiratory Research Centre Telethon Kids Institute University of Western Australia Nedlands WA Australia
| | - Samantha A McLean
- Wal‐Yan Respiratory Research Centre Telethon Kids Institute University of Western Australia Nedlands WA Australia
| | - Camilla Margaroli
- Department of Medicine Division of Pulmonary, Allergy and Critical Care Medicine University of Alabama at Birmingham Birmingham AL USA
- Program in Protease and Matrix Biology University of Alabama at Birmingham Birmingham AL USA
| | - Patricia Agudelo‐Romero
- Wal‐Yan Respiratory Research Centre Telethon Kids Institute University of Western Australia Nedlands WA Australia
| | - Stephen M Stick
- Faculty of Health and Medical Sciences University of Western Australia Nedlands WA Australia
- Wal‐Yan Respiratory Research Centre Telethon Kids Institute University of Western Australia Nedlands WA Australia
- Department of Respiratory and Sleep Medicine Perth Children’s Hospital Nedlands WA Australia
| | - Rabindra Tirouvanziam
- Department of Pediatrics Emory University Atlanta GA USA
- Center for CF and Airways Disease Research Children’s Healthcare of Atlanta Atlanta GA USA
| | - Anthony Kicic
- Faculty of Health and Medical Sciences University of Western Australia Nedlands WA Australia
- Wal‐Yan Respiratory Research Centre Telethon Kids Institute University of Western Australia Nedlands WA Australia
- Department of Respiratory and Sleep Medicine Perth Children’s Hospital Nedlands WA Australia
- Occupation and Environment School of Public Health Curtin University Bentley WA Australia
| | - Luke W Garratt
- Wal‐Yan Respiratory Research Centre Telethon Kids Institute University of Western Australia Nedlands WA Australia
| | | | | |
Collapse
|
20
|
Wong SL, Awatade NT, Astore MA, Allan KM, Carnell MJ, Slapetova I, Chen PC, Setiadi J, Pandzic E, Fawcett LK, Widger JR, Whan RM, Griffith R, Ooi CY, Kuyucak S, Jaffe A, Waters SA. Molecular Dynamics and Theratyping in Airway and Gut Organoids Reveal R352Q-CFTR Conductance Defect. Am J Respir Cell Mol Biol 2022; 67:99-111. [PMID: 35471184 PMCID: PMC9273222 DOI: 10.1165/rcmb.2021-0337oc] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
A significant challenge to making targeted cystic fibrosis transmembrane conductance regulator (CFTR) modulator therapies accessible to all individuals with cystic fibrosis (CF) are many mutations in the CFTR gene that can cause CF, most of which remain uncharacterized. Here, we characterized the structural and functional defects of the rare CFTR mutation R352Q, with a potential role contributing to intrapore chloride ion permeation, in patient-derived cell models of the airway and gut. CFTR function in differentiated nasal epithelial cultures and matched intestinal organoids was assessed using an ion transport assay and forskolin-induced swelling assay, respectively. CFTR potentiators (VX-770, GLPG1837, and VX-445) and correctors (VX-809, VX-445, with or without VX-661) were tested. Data from R352Q-CFTR were compared with data of 20 participants with mutations with known impact on CFTR function. R352Q-CFTR has residual CFTR function that was restored to functional CFTR activity by CFTR potentiators but not the corrector. Molecular dynamics simulations of R352Q-CFTR were carried out, which indicated the presence of a chloride conductance defect, with little evidence supporting a gating defect. The combination approach of in vitro patient-derived cell models and in silico molecular dynamics simulations to characterize rare CFTR mutations can improve the specificity and sensitivity of modulator response predictions and aid in their translational use for CF precision medicine.
Collapse
Affiliation(s)
- Sharon L Wong
- University of New South Wales, 7800, School of Women's and Children's Health, Faculty of Medicine, Sydney, New South Wales, Australia.,University of New South Wales, 7800, Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC), Sydney, New South Wales, Australia
| | - Nikhil T Awatade
- University of New South Wales, 7800, School of Women's and Children's Health, Faculty of Medicine, Sydney, New South Wales, Australia.,University of New South Wales, 7800, Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC), Sydney, New South Wales, Australia
| | - Miro A Astore
- The University of Sydney, 4334, School of Physics, Sydney, New South Wales, Australia
| | - Katelin M Allan
- University of New South Wales, 7800, School of Women's and Children's Health, Faculty of Medicine, Sydney, New South Wales, Australia.,University of New South Wales, 7800, Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC), Sydney, New South Wales, Australia
| | - Michael J Carnell
- University of New South Wales, 7800, Biomedical Imaging Facility, Mark Wainwright Analytical Centre, Sydney, New South Wales, Australia
| | - Iveta Slapetova
- University of New South Wales, 7800, Biomedical Imaging Facility, Mark Wainwright Analytical Centre, Sydney, New South Wales, Australia
| | - Po-Chia Chen
- The University of Sydney, 4334, School of Physics, Sydney, New South Wales, Australia
| | - Jeffry Setiadi
- The University of Sydney, 4334, School of Physics, Sydney, New South Wales, Australia
| | - Elvis Pandzic
- University of New South Wales, 7800, Biomedical Imaging Facility, Mark Wainwright Analytical Cen, Sydney, New South Wales, Australia
| | - Laura K Fawcett
- University of New South Wales, 7800, School of Women's and Children's Health, Faculty of Medicine, Sydney, New South Wales, Australia.,University of New South Wales, 7800, Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC), Sydney, New South Wales, Australia.,Sydney Children's Hospital Randwick, 63623, Department of Respiratory Medicine, Randwick, New South Wales, Australia
| | - John R Widger
- University of New South Wales, 7800, School of Women's and Children's Health, Faculty of Medicine, Sydney, New South Wales, Australia.,University of New South Wales, 7800, Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC), Sydney, New South Wales, Australia.,Sydney Children's Hospital Randwick, 63623, Department of Respiratory Medicine, Randwick, New South Wales, Australia
| | - Renee M Whan
- University of New South Wales, 7800, Biomedical Imaging Facility, Mark Wainwright Analytical Centre, Sydney, New South Wales, Australia
| | - Renate Griffith
- University of New South Wales, 7800, School of Chemistry, Sydney, New South Wales, Australia
| | - Chee Y Ooi
- Sydney Children's Hospital Randwick, Gastroenterology, Sydney, New South Wales, Australia
| | - Serdar Kuyucak
- The University of Sydney, 4334, School of Physics, Sydney, New South Wales, Australia
| | - Adam Jaffe
- Sydney Children`s Hospital, Respiratory Medicine, Sydney, New South Wales, Australia.,University of New South Wales, 7800, School of Women`s and Children`s Health, Sydney, New South Wales, Australia
| | - Shafagh A Waters
- Sydney Children's Hospital, Department of Respiratory Medicine, Sydney, New South Wales, Australia.,Univeristy of New South Wales, School of Women's and Children's Health, Sydney, New South Wales, Australia;
| |
Collapse
|
21
|
Martinovich KM, Kicic A, Stick SM, Johnsen RD, Fletcher S, Wilton SD. Investigating the Implications of CFTR Exon Skipping Using a Cftr Exon 9 Deleted Mouse Model. Front Pharmacol 2022; 13:868863. [PMID: 35392567 PMCID: PMC8981082 DOI: 10.3389/fphar.2022.868863] [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: 02/03/2022] [Accepted: 03/07/2022] [Indexed: 11/13/2022] Open
Abstract
Introduction: Severity and disease progression in people with Cystic Fibrosis (CF) is typically dependent on their genotype. One potential therapeutic strategy for people with specific mutations is exon skipping with antisense oligonucleotides (AO). CFTR exon 9 is an in-frame exon and hence the exclusion of this exon would excise only 31 amino acids but not alter the reading frame of the remaining mRNA. Splice mutations 1209 + 1 G > C and 1209 + 2 T > G were documented to cause CFTR exon 9 skipping and these variants were reported to manifest as a milder CF disease, therefore exon 9 skipping could be beneficial for people with class I mutations that affect exon 9 such as p.Trp401X. While the impact of exon 9 skipping on gene expression and cellular pathways can be studied in cells in vitro, trace amount of full-length normal or mutated material could confound the evaluation. To overcome this limitation, the impact of CFTR exon 9 skipping on disease phenotype and severity is more effectively evaluated in a small animal model. It was hypothesised that antisense oligonucleotide-mediated skipping this particular exon could result in a "mild mouse CF phenotype". Methods: Cftr exon 9 deleted mice were generated using homologous recombination. Survival of homozygous (Cftr Δ9/Δ9 ) and heterozygous (Cftr Δ9/+ ) mice was compared to that of other CF mouse models, and lung and intestinal organ histology examined for any pathologies. Primary airway epithelial cells (pAECs) were harvested from Cftr Δ9/Δ9 mice and cultured at the Air Liquid Interface for CFTR functional assessment using Ussing Chamber analysis. Results: A Cftr Δ9/Δ9 mouse model presented with intestinal obstructions, and at time of weaning (21 days). Cftr Δ9/Δ9 mice had a survival rate of 83% that dropped to 38% by day 50. Histological sections of the small intestine from Cftr Δ9/Δ9 mice showed more goblet cells and mucus accumulation than samples from the Cftr Δ9/+ littermates. Airway epithelial cell cultures established from Cftr Δ9/Δ9 mice were not responsive to forskolin stimulation. Summary: The effect of Cftr exon 9 deletion on Cftr function was assessed and it was determined that the encoded Cftr isoform did not result in a milder "mouse CF disease phenotype," suggesting that Cftr exon 9 is not dispensable, although further investigation in human CF pAECs would be required to confirm this observation.
Collapse
Affiliation(s)
- Kelly M Martinovich
- School of Medicine, The University of Western Australia, Perth, WA, Australia.,Telethon Kids Institute, Wal-yan Respiratory Research Centre, Perth, WA, Australia.,Centre for Molecular Medicine and Innovative Therapeutics, Health Futures Institute, Murdoch University, Murdoch, WA, Australia
| | - Anthony Kicic
- School of Medicine, The University of Western Australia, Perth, WA, Australia.,Telethon Kids Institute, Wal-yan Respiratory Research Centre, Perth, WA, Australia.,Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia, Perth, WA, Australia.,Department of Respiratory and Sleep Medicine, Perth Childrens Hospital, Nedlands, WA, Australia.,School of Population Health, Curtin University, Bentley, WA, Australia
| | - Stephen M Stick
- School of Medicine, The University of Western Australia, Perth, WA, Australia.,Telethon Kids Institute, Wal-yan Respiratory Research Centre, Perth, WA, Australia.,Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia, Perth, WA, Australia.,Department of Respiratory and Sleep Medicine, Perth Childrens Hospital, Nedlands, WA, Australia
| | - Russell D Johnsen
- Centre for Molecular Medicine and Innovative Therapeutics, Health Futures Institute, Murdoch University, Murdoch, WA, Australia.,Perron Institute for Neurological and Translational Sciences, Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, WA, Australia
| | - Sue Fletcher
- Centre for Molecular Medicine and Innovative Therapeutics, Health Futures Institute, Murdoch University, Murdoch, WA, Australia.,Perron Institute for Neurological and Translational Sciences, Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, WA, Australia.,PYC Therapeutics, Perth, WA, Australia
| | - Steve D Wilton
- Centre for Molecular Medicine and Innovative Therapeutics, Health Futures Institute, Murdoch University, Murdoch, WA, Australia.,Perron Institute for Neurological and Translational Sciences, Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, WA, Australia
| |
Collapse
|
22
|
Analysis of the diagnosis of Japanese patients with primary ciliary dyskinesia using a conditional reprogramming culture. Respir Investig 2022; 60:407-417. [PMID: 35305968 DOI: 10.1016/j.resinv.2022.02.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 02/13/2022] [Accepted: 02/15/2022] [Indexed: 11/24/2022]
Abstract
BACKGROUND Primary ciliary dyskinesia (PCD) is diagnosed through multiple methods, including transmission electron microscopy (TEM), a high-speed video microscopy analysis (HSVA), immunofluorescence (IF), and genetic testing. A primary cell culture has been recommended to avoid the misdiagnosis of secondary ciliary dyskinesia derived from infection or inflammation and improve diagnostic accuracy. However, primary cells fail to differentiate into ciliated cells through repeated passages. The conditional reprogramming culture (CRC) method, a combination of a Rho-kinase inhibitor and fibroblast feeder cells, has been applied to cystic fibrosis. The goal of this study was to evaluate the value of CRC in diagnosing PCD in Japanese patients. METHODS Eleven patients clinically suspected of having PCD were included. Airway epithelial cells were obtained from an endobronchial forceps biopsy and cultured at the air-liquid interface (ALI) combined with CRC. Ciliary movement, ultrastructure, and mutated ciliary protein evaluation were performed using HSVA, TEM, and IF, respectively. Genetic testing was performed on some patients. RESULTS CRC yielded dense and well-differentiated ciliated cells with a high success rate (∼90%). In patients with PCD, the ciliary ultrastructure phenotype (outer dynein arm defects or normal ultrastructure) and IF findings (absence of the mutated ciliary protein) were confirmed after CRC. In DNAH11-mutant cases with normal ultrastructure by TEM, the HSVA revealed stiff and hyperfrequent ciliary beating with low bending capacity in CRC-expanded cells, thereby supporting the diagnosis. CONCLUSIONS CRC could be a potential tool for improving diagnostic accuracy and contributing to future clinical and basic research in PCD.
Collapse
|
23
|
Ekanger CT, Zhou F, Bohan D, Lotsberg ML, Ramnefjell M, Hoareau L, Røsland GV, Lu N, Aanerud M, Gärtner F, Salminen PR, Bentsen M, Halvorsen T, Ræder H, Akslen LA, Langeland N, Cox R, Maury W, Stuhr LEB, Lorens JB, Engelsen AST. Human Organotypic Airway and Lung Organoid Cells of Bronchiolar and Alveolar Differentiation Are Permissive to Infection by Influenza and SARS-CoV-2 Respiratory Virus. Front Cell Infect Microbiol 2022; 12:841447. [PMID: 35360113 PMCID: PMC8964279 DOI: 10.3389/fcimb.2022.841447] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/10/2022] [Indexed: 12/13/2022] Open
Abstract
The ongoing coronavirus disease 2019 (COVID-19) pandemic has led to the initiation of unprecedented research efforts to understand the pathogenesis mediated by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). More knowledge is needed regarding the cell type-specific cytopathology and its impact on cellular tropism. Furthermore, the impact of novel SARS-CoV-2 mutations on cellular tropism, alternative routes of entry, the impact of co-infections, and virus replication kinetics along the respiratory tract remains to be explored in improved models. Most applied virology models are not well suited to address the remaining questions, as they do not recapitulate the histoarchitecture and cellular composition of human respiratory tissues. The overall aim of this work was to establish from single biopsy specimens, a human adult stem cell-derived organoid model representing the upper respiratory airways and lungs and explore the applicability of this model to study respiratory virus infection. First, we characterized the organoid model with respect to growth pattern and histoarchitecture, cellular composition, and functional characteristics. Next, in situ expression of viral entry receptors, including influenza virus-relevant sialic acids and SARS-CoV-2 entry receptor ACE2 and TMPRSS2, were confirmed in organoids of bronchiolar and alveolar differentiation. We further showed successful infection by pseudotype influenza A H7N1 and H5N1 virus, and the ability of the model to support viral replication of influenza A H7N1 virus. Finally, successful infection and replication of a clinical isolate of SARS-CoV-2 were confirmed in the organoids by TCID50 assay and immunostaining to detect intracellular SARS-CoV-2 specific nucleocapsid and dsRNA. The prominent syncytia formation in organoid tissues following SARS-CoV-2 infection mimics the findings from infected human tissues in situ. We conclude that the human organotypic model described here may be particularly useful for virology studies to evaluate regional differences in the host response to infection. The model contains the various cell types along the respiratory tract, expresses respiratory virus entry factors, and supports successful infection and replication of influenza virus and SARS-CoV-2. Thus, the model may serve as a relevant and reliable tool in virology and aid in pandemic preparedness, and efficient evaluation of antiviral strategies.
Collapse
Affiliation(s)
- Camilla Tvedt Ekanger
- Department of Biomedicine, Faculty of Medicine, University of Bergen, Bergen, Norway
- Centre for Cancer Biomarkers, University of Bergen (CCBIO), Department of Clinical Medicine, Bergen, Norway
- The Influenza Centre, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Fan Zhou
- The Influenza Centre, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Dana Bohan
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, United States
| | - Maria Lie Lotsberg
- Department of Biomedicine, Faculty of Medicine, University of Bergen, Bergen, Norway
- Centre for Cancer Biomarkers, University of Bergen (CCBIO), Department of Clinical Medicine, Bergen, Norway
| | - Maria Ramnefjell
- Centre for Cancer Biomarkers, University of Bergen (CCBIO), Department of Clinical Medicine, Bergen, Norway
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Laurence Hoareau
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
- Department of Pediatrics, Haukeland University Hospital, Bergen, Norway
| | - Gro Vatne Røsland
- Department of Biomedicine, Faculty of Medicine, University of Bergen, Bergen, Norway
- Centre for Cancer Biomarkers, University of Bergen (CCBIO), Department of Clinical Medicine, Bergen, Norway
| | - Ning Lu
- Department of Biomedicine, Faculty of Medicine, University of Bergen, Bergen, Norway
- Centre for Cancer Biomarkers, University of Bergen (CCBIO), Department of Clinical Medicine, Bergen, Norway
| | - Marianne Aanerud
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
- Department of Thoracic Medicine, Haukeland University Hospital, Bergen, Norway
| | - Fabian Gärtner
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
- Department of Thoracic Medicine, Haukeland University Hospital, Bergen, Norway
| | - Pirjo Riitta Salminen
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
- Section of Cardiothoracic Surgery, Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| | - Mariann Bentsen
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
- Department of Pediatrics, Haukeland University Hospital, Bergen, Norway
| | - Thomas Halvorsen
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
- Department of Pediatrics, Haukeland University Hospital, Bergen, Norway
| | - Helge Ræder
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
- Department of Pediatrics, Haukeland University Hospital, Bergen, Norway
| | - Lars A. Akslen
- Centre for Cancer Biomarkers, University of Bergen (CCBIO), Department of Clinical Medicine, Bergen, Norway
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Nina Langeland
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Rebecca Cox
- The Influenza Centre, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
- Department of Microbiology, Haukeland University Hospital, Bergen, Norway
| | - Wendy Maury
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, United States
| | | | - James B. Lorens
- Department of Biomedicine, Faculty of Medicine, University of Bergen, Bergen, Norway
- Centre for Cancer Biomarkers, University of Bergen (CCBIO), Department of Clinical Medicine, Bergen, Norway
| | - Agnete S. T. Engelsen
- Department of Biomedicine, Faculty of Medicine, University of Bergen, Bergen, Norway
- Centre for Cancer Biomarkers, University of Bergen (CCBIO), Department of Clinical Medicine, Bergen, Norway
- *Correspondence: Agnete S. T. Engelsen,
| |
Collapse
|
24
|
Shaw NC, Kicic A, Fletcher S, Wilton SD, Stick SM, Schultz A. Primary Nasal Epithelial Cells as a Surrogate Cell Culture Model for Type-II Alveolar Cells to Study ABCA-3 Deficiency. Front Med (Lausanne) 2022; 9:827416. [PMID: 35265641 PMCID: PMC8899037 DOI: 10.3389/fmed.2022.827416] [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: 12/02/2021] [Accepted: 01/24/2022] [Indexed: 11/13/2022] Open
Abstract
ATP Binding Cassette Subfamily A Member 3 (ABCA-3) is a lipid transporter protein highly expressed in type-II alveolar (AT-II) cells. Mutations in ABCA3 can result in severe respiratory disease in infants and children. To study ABCA-3 deficiency in vitro, primary AT-II cells would be the cell culture of choice although sample accessibility is limited. Our aim was to investigate the suitability of primary nasal epithelial cells, as a surrogate culture model for AT-II cells, to study ABCA-3 deficiency. Expression of ABCA3, and surfactant protein genes, SFTPB and SFTPC, was detected in primary nasal epithelial cells but at a significantly lower level than in AT-II cells. ABCA-3, SP-B, and SP-C were detected by immunofluorescence microscopy in primary nasal epithelial cells. However, SP-B and SP-C were undetectable in primary nasal epithelial cells using western blotting. Structurally imperfect lamellar bodies were observed in primary nasal epithelial cells using transmission electron microscopy. Functional assessment of the ABCA-3 protein demonstrated that higher concentrations of doxorubicin reduced cell viability in ABCA-3 deficient nasal epithelial cells compared to controls in an assay-dependent manner. Our results indicate that there may be a role for primary nasal epithelial cell cultures to model ABCA-3 deficiency in vitro, although additional cell culture models that more effectively recapitulate the AT-II phenotype may be required.
Collapse
Affiliation(s)
- Nicole C Shaw
- Faculty of Health and Medical Sciences, The University of Western Australia, Perth, WA, Australia.,Wal-yan Respiratory Research Centre, Telethon Kids Institute, The University of Western Australia, Perth, WA, Australia
| | - Anthony Kicic
- Faculty of Health and Medical Sciences, The University of Western Australia, Perth, WA, Australia.,Wal-yan Respiratory Research Centre, Telethon Kids Institute, The University of Western Australia, Perth, WA, Australia.,Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia, Perth, WA, Australia.,Department of Respiratory and Sleep Medicine, Perth Children's Hospital, Perth, WA, Australia.,Occupation and Environment, School of Public Health, Curtin University, Perth, WA, Australia
| | - Sue Fletcher
- Centre for Neuromuscular and Neurological Disorders, Perron Institute for Neurological and Translational Sciences, The University of Western Australia, Perth, WA, Australia.,Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, WA, Australia
| | - Stephen D Wilton
- Centre for Neuromuscular and Neurological Disorders, Perron Institute for Neurological and Translational Sciences, The University of Western Australia, Perth, WA, Australia.,Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, WA, Australia
| | - Stephen M Stick
- Faculty of Health and Medical Sciences, The University of Western Australia, Perth, WA, Australia.,Wal-yan Respiratory Research Centre, Telethon Kids Institute, The University of Western Australia, Perth, WA, Australia.,Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia, Perth, WA, Australia.,Department of Respiratory and Sleep Medicine, Perth Children's Hospital, Perth, WA, Australia
| | - André Schultz
- Faculty of Health and Medical Sciences, The University of Western Australia, Perth, WA, Australia.,Wal-yan Respiratory Research Centre, Telethon Kids Institute, The University of Western Australia, Perth, WA, Australia.,Department of Respiratory and Sleep Medicine, Perth Children's Hospital, Perth, WA, Australia
| |
Collapse
|
25
|
Schmidt H, Gutjahr L, Sauter A, Zech F, Nchioua R, Stenger S, Frick M, Kirchhoff F, Dietl P, Wittekindt OH. Serially passaged, conditionally reprogrammed nasal epithelial cells as a model to study epithelial functions and SARS-CoV-2 infection. Am J Physiol Cell Physiol 2022; 322:C591-C604. [PMID: 35196166 PMCID: PMC8977148 DOI: 10.1152/ajpcell.00363.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Primary airway epithelial cells (pAECs) cultivated at air-liquid interface (ALI) conditions are widely used as surrogates for human in vivo epithelia. To extend the proliferative capacity and to enable serially passaging of pAECs, conditional reprogramming (cr) has been employed in recent years. However, ALI epithelia derived from cr cells often display functional changes with increasing passages. This highlights the need for thorough validation of the ALI cultures for the respective application. In our study, we evaluated the use of serially passaged cr nasal epithelial cells (crNECs) as a model to study SARS-CoV-2 infection and effects on ion and water transport. NECs were obtained from healthy individuals and cultivated as ALI epithelia derived from passages 1, 2, 3, and 5. We compared epithelial differentiation, ion and water transport, and infection with SARS-CoV-2 between passages. Our results show that epithelia maintained major differentiation characteristics and physiological ion and water transport properties through all passages. However, the frequency of ciliated cells, short circuit currents reflecting epithelial Na+ channel (ENaC) and cystic fibrosis transmembrane conductance regulator (CFTR) activity and expression of aquaporin 3 and 5 decreased gradually over passages. crNECs also expressed SARS-CoV-2 receptors angiotensin converting enzyme 2 (ACE2) and transmembrane serin2 protease 2 (TMPRSS2) across all passages and allowed SARS-CoV-2 replication in all passages. In summary, we provide evidence that passaged crNECs provide an appropriate model to study SARS-CoV-2 infection and also epithelial transport function when considering some limitations that we defined herein.
Collapse
Affiliation(s)
- Hanna Schmidt
- Department of Pediatric and Adolescent Medicine, Ulm University Medical Cente, Ulm, Germany.,Institute of General Physiology, Ulm University, Ulm, Germany
| | - Lara Gutjahr
- Institute of General Physiology, Ulm University, Ulm, Germany
| | | | - Fabian Zech
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Rayhane Nchioua
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Steffen Stenger
- Institute of Medical Microbiology and Hygiene, Ulm University Medical Center, Ulm, Germany
| | - Manfred Frick
- Institute of General Physiology, Ulm University, Ulm, Germany
| | - Frank Kirchhoff
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Paul Dietl
- Institute of General Physiology, Ulm University, Ulm, Germany
| | | |
Collapse
|
26
|
Tran E, Shi T, Li X, Chowdhury AY, Jiang D, Liu Y, Wang H, Yan C, Wallace WD, Lu R, Ryan AL, Marconett CN, Zhou B, Borok Z, Offringa IA. Development of human alveolar epithelial cell models to study distal lung biology and disease. iScience 2022; 25:103780. [PMID: 35169685 PMCID: PMC8829779 DOI: 10.1016/j.isci.2022.103780] [Citation(s) in RCA: 11] [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: 01/25/2021] [Revised: 10/27/2021] [Accepted: 01/12/2022] [Indexed: 11/25/2022] Open
Abstract
Many acute and chronic diseases affect the distal lung alveoli. Alveolar epithelial cell (AEC) lines are needed to better model these diseases. We used de-identified human remnant transplant lungs to develop a method to establish AEC lines. The lines grow well in 2-dimensional (2D) culture as epithelial monolayers expressing lung progenitor markers. In 3-dimensional (3D) culture with fibroblasts, Matrigel, and specific media conditions, the cells form alveolar-like organoids expressing mature AEC markers including aquaporin 5 (AQP5), G-protein-coupled receptor class C group 5 member A (GPRC5A), and surface marker HTII280. Single-cell RNA sequencing of an AEC line in 2D versus 3D culture revealed increased cellular heterogeneity and induction of cytokine and lipoprotein signaling in 3D organoids. Our approach yields lung progenitor lines that retain the ability to differentiate along the alveolar cell lineage despite long-term expansion and provides a valuable system to model and study the distal lung in vitro.
Collapse
Affiliation(s)
- Evelyn Tran
- Department of Surgery, Keck School of Medicine, University of Southern California (USC), Los Angeles, CA 90033, USA
- USC Norris Comprehensive Cancer Center, Keck School of Medicine, USC, Los Angeles, CA 90033, USA
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, USC, Los Angeles, CA 90033, USA
| | - Tuo Shi
- Department of Surgery, Keck School of Medicine, University of Southern California (USC), Los Angeles, CA 90033, USA
- USC Norris Comprehensive Cancer Center, Keck School of Medicine, USC, Los Angeles, CA 90033, USA
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, USC, Los Angeles, CA 90033, USA
| | - Xiuwen Li
- USC Norris Comprehensive Cancer Center, Keck School of Medicine, USC, Los Angeles, CA 90033, USA
- Department of Translational Genomics, Keck School of Medicine, USC, Los Angeles, CA 90033, USA
| | - Adnan Y. Chowdhury
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, USC, Los Angeles, CA 90033, USA
| | - Du Jiang
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, USC, Los Angeles, CA 90033, USA
| | - Yixin Liu
- Hastings Center for Pulmonary Research and Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Keck School of Medicine, USC, Los Angeles, CA 90033, USA
| | - Hongjun Wang
- Hastings Center for Pulmonary Research and Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Keck School of Medicine, USC, Los Angeles, CA 90033, USA
| | - Chunli Yan
- Department of Surgery, Keck School of Medicine, University of Southern California (USC), Los Angeles, CA 90033, USA
- USC Norris Comprehensive Cancer Center, Keck School of Medicine, USC, Los Angeles, CA 90033, USA
| | - William D. Wallace
- Department of Pathology, Keck School of Medicine, USC, Los Angeles, CA 90033, USA
| | - Rong Lu
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, USC, Los Angeles, CA 90033, USA
| | - Amy L. Ryan
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, USC, Los Angeles, CA 90033, USA
- Hastings Center for Pulmonary Research and Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Keck School of Medicine, USC, Los Angeles, CA 90033, USA
| | - Crystal N. Marconett
- Department of Surgery, Keck School of Medicine, University of Southern California (USC), Los Angeles, CA 90033, USA
- USC Norris Comprehensive Cancer Center, Keck School of Medicine, USC, Los Angeles, CA 90033, USA
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, USC, Los Angeles, CA 90033, USA
| | - Beiyun Zhou
- USC Norris Comprehensive Cancer Center, Keck School of Medicine, USC, Los Angeles, CA 90033, USA
- Hastings Center for Pulmonary Research and Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Keck School of Medicine, USC, Los Angeles, CA 90033, USA
| | - Zea Borok
- USC Norris Comprehensive Cancer Center, Keck School of Medicine, USC, Los Angeles, CA 90033, USA
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, USC, Los Angeles, CA 90033, USA
- Hastings Center for Pulmonary Research and Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Keck School of Medicine, USC, Los Angeles, CA 90033, USA
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Ite A. Offringa
- Department of Surgery, Keck School of Medicine, University of Southern California (USC), Los Angeles, CA 90033, USA
- USC Norris Comprehensive Cancer Center, Keck School of Medicine, USC, Los Angeles, CA 90033, USA
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, USC, Los Angeles, CA 90033, USA
| |
Collapse
|
27
|
Morgan R, Manfredi C, Easley KF, Watkins LD, Hunt WR, Goudy SL, Sorscher EJ, Koval M, Molina SA. A medium composition containing normal resting glucose that supports differentiation of primary human airway cells. Sci Rep 2022; 12:1540. [PMID: 35087167 PMCID: PMC8795386 DOI: 10.1038/s41598-022-05446-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 01/10/2022] [Indexed: 02/07/2023] Open
Abstract
Primary cells isolated from the human respiratory tract are the state-of-the-art for in vitro airway epithelial cell research. Airway cell isolates require media that support expansion of cells in a basal state to maintain the capacity for differentiation as well as proper cellular function. By contrast, airway cell differentiation at an air-liquid interface (ALI) requires a distinct medium formulation that typically contains high levels of glucose. Here, we expanded and differentiated human basal cells isolated from the nasal and conducting airway to a mature mucociliary epithelial cell layer at ALI using a medium formulation containing normal resting glucose levels. Of note, bronchial epithelial cells expanded and differentiated in normal resting glucose medium showed insulin-stimulated glucose uptake which was inhibited by high glucose concentrations. Normal glucose containing ALI also enabled differentiation of nasal and tracheal cells that showed comparable electrophysiological profiles when assessed for cystic fibrosis transmembrane conductance regulator (CFTR) function and that remained responsive for up to 7 weeks in culture. These data demonstrate that normal glucose containing medium supports differentiation of primary nasal and lung epithelial cells at ALI, is well suited for metabolic studies, and avoids pitfalls associated with exposure to high glucose.
Collapse
Affiliation(s)
- Rachel Morgan
- Center for Cystic Fibrosis and Airways Disease Research, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, 205 Whitehead Building, 615 Michael Street, Atlanta, GA, 30322, USA
| | - Candela Manfredi
- Center for Cystic Fibrosis and Airways Disease Research, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Division of Pulmonary, Allergy & Immunology, Cystic Fibrosis, and Sleep, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Kristen F Easley
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, 205 Whitehead Building, 615 Michael Street, Atlanta, GA, 30322, USA
| | - Lionel D Watkins
- Center for Cystic Fibrosis and Airways Disease Research, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, 205 Whitehead Building, 615 Michael Street, Atlanta, GA, 30322, USA
| | - William R Hunt
- Center for Cystic Fibrosis and Airways Disease Research, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, 205 Whitehead Building, 615 Michael Street, Atlanta, GA, 30322, USA
| | - Steven L Goudy
- Division of Pulmonary, Allergy & Immunology, Cystic Fibrosis, and Sleep, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Eric J Sorscher
- Center for Cystic Fibrosis and Airways Disease Research, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Division of Pulmonary, Allergy & Immunology, Cystic Fibrosis, and Sleep, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Michael Koval
- Center for Cystic Fibrosis and Airways Disease Research, Emory University School of Medicine, Atlanta, GA, 30322, USA.
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, 205 Whitehead Building, 615 Michael Street, Atlanta, GA, 30322, USA.
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| | - Samuel A Molina
- Center for Cystic Fibrosis and Airways Disease Research, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, 205 Whitehead Building, 615 Michael Street, Atlanta, GA, 30322, USA
| |
Collapse
|
28
|
Dysregulated Notch Signaling in the Airway Epithelium of Children with Wheeze. J Pers Med 2021; 11:jpm11121323. [PMID: 34945795 PMCID: PMC8707470 DOI: 10.3390/jpm11121323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 11/16/2021] [Accepted: 11/29/2021] [Indexed: 11/17/2022] Open
Abstract
The airway epithelium of children with wheeze is characterized by defective repair that contributes to disease pathobiology. Dysregulation of developmental processes controlled by Notch has been identified in chronic asthma. However, its role in airway epithelial cells of young children with wheeze, particularly during repair, is yet to be determined. We hypothesized that Notch is dysregulated in primary airway epithelial cells (pAEC) of children with wheeze contributing to defective repair. This study investigated transcriptional and protein expression and function of Notch in pAEC isolated from children with and without wheeze. Primary AEC of children with and without wheeze were found to express all known Notch receptors and ligands, although pAEC from children with wheeze expressed significantly lower NOTCH2 (10-fold, p = 0.004) and higher JAG1 (3.5-fold, p = 0.002) mRNA levels. These dysregulations were maintained in vitro and cultures from children with wheeze displayed altered kinetics of both NOTCH2 and JAG1 expression during repair. Following Notch signaling inhibition, pAEC from children without wheeze failed to repair (wound closure rate of 76.9 ± 3.2%). Overexpression of NOTCH2 in pAEC from children with wheeze failed to rescue epithelial repair following wounding. This study illustrates the involvement of the Notch pathway in airway epithelial wound repair in health and disease, where its dysregulation may contribute to asthma development.
Collapse
|
29
|
Landwehr KR, Hillas J, Mead-Hunter R, Brooks P, King A, O'Leary RA, Kicic A, Mullins BJ, Larcombe AN. In Vitro primary human airway epithelial whole exhaust exposure. MethodsX 2021; 8:101561. [PMID: 34754823 PMCID: PMC8563817 DOI: 10.1016/j.mex.2021.101561] [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: 08/13/2021] [Accepted: 10/23/2021] [Indexed: 11/06/2022] Open
Abstract
The method outlined in this article is a customization of the whole exhaust exposure method generated by Mullins et al. (2016) using reprogrammed primary human airway epithelial cells as described by Martinovich et al. (2017). It has been used successfully to generate recently published data (Landwehr et al. 2021). The goal was to generate an exhaust exposure model where exhaust is collected from a modern engine, real-world exhaust concentrations are used and relevant tissues exposed to assess the effects of multiple biodiesel exposures. Exhaust was generated, gently vacuumed into a dilution chamber where it was diluted 1/15 with air and then vacuumed into an incubator containing the primary cell cultures for exposure. Exhaust physico-chemical properties including combustion gas concentrations and particle spectra were then analyzed using a combustion gas analyzer and a Universal Scanning Mobility Particle Sizer. 24 h after exposure, cellular viability and mediator release were measured using Annexin-V/PI staining and meditator multiplexing kits respectively. This method was generated to test biodiesel exhaust exposures but can be easily adapted for any type of engine exhaust exposure or even potentially other respirable environmental exposures such as woodsmoke. The main customization points for this method are:Exhaust generated by a diesel engine equipped with EURO VI exhaust after treatment devices including diesel particulate filter and diesel oxidation catalyst. The generated exhaust was diluted 1/15 with air to replicate real world exposure concentrations. Used primary human airway epithelial cells obtained from bronchoscope brushings from multiple volunteers and reprogrammed to allow multiple, comparative exposures from the same individual.
Collapse
Affiliation(s)
- Katherine R Landwehr
- Occupation, Environment and Safety, School of Population Health, Curtin University, PO Box U1987, Perth, Western Australia 6845, Australia.,Respiratory Environmental Health, Wal-yan Respiratory Research Centre, Telethon Kids Institute, Perth Children's Hospital, Nedlands, Perth, Western Australia 6009, Australia
| | - Jessica Hillas
- Respiratory Environmental Health, Wal-yan Respiratory Research Centre, Telethon Kids Institute, Perth Children's Hospital, Nedlands, Perth, Western Australia 6009, Australia
| | - Ryan Mead-Hunter
- Occupation, Environment and Safety, School of Population Health, Curtin University, PO Box U1987, Perth, Western Australia 6845, Australia
| | - Peter Brooks
- School of Science, Technology and Engineering, University of the Sunshine Coast, Sippy Downs, Queensland, Australia
| | - Andrew King
- Fluid Dynamics Research Group, School of Civil and Mechanical Engineering, Curtin University, Perth, Western Australia, Australia
| | - Rebecca A O'Leary
- Department of Primary Industries and Regional Development, Perth, Western Australia 6151, Australia
| | - Anthony Kicic
- Occupation, Environment and Safety, School of Population Health, Curtin University, PO Box U1987, Perth, Western Australia 6845, Australia.,Respiratory Environmental Health, Wal-yan Respiratory Research Centre, Telethon Kids Institute, Perth Children's Hospital, Nedlands, Perth, Western Australia 6009, Australia.,Department of Respiratory and Sleep Medicine, Perth Children's Hospital, Nedlands, Perth, Western Australia 6009, Australia.,Centre for Cell Therapy and Regenerative Medicine, The University of Western Australia, Perth, Western Australia 6009, Australia
| | - Benjamin J Mullins
- Occupation, Environment and Safety, School of Population Health, Curtin University, PO Box U1987, Perth, Western Australia 6845, Australia
| | - Alexander N Larcombe
- Occupation, Environment and Safety, School of Population Health, Curtin University, PO Box U1987, Perth, Western Australia 6845, Australia.,Respiratory Environmental Health, Wal-yan Respiratory Research Centre, Telethon Kids Institute, Perth Children's Hospital, Nedlands, Perth, Western Australia 6009, Australia
| | | | | |
Collapse
|
30
|
Landwehr KR, Hillas J, Mead-Hunter R, Brooks P, King A, O'Leary RA, Kicic A, Mullins BJ, Larcombe AN. Fuel feedstock determines biodiesel exhaust toxicity in a human airway epithelial cell exposure model. JOURNAL OF HAZARDOUS MATERIALS 2021; 420:126637. [PMID: 34329109 DOI: 10.1016/j.jhazmat.2021.126637] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 07/02/2021] [Accepted: 07/10/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Biodiesel is promoted as a sustainable replacement for commercial diesel. Biodiesel fuel and exhaust properties change depending on the base feedstock oil/fat used during creation. The aims of this study were, for the first time, to compare the exhaust exposure health impacts of a wide range of biodiesels made from different feedstocks and relate these effects with the corresponding exhaust characteristics. METHOD Primary airway epithelial cells were exposed to diluted exhaust from an engine running on conventional diesel and biodiesel made from Soy, Canola, Waste Cooking Oil, Tallow, Palm and Cottonseed. Exhaust properties and cellular viability and mediator release were analysed post exposure. RESULTS The exhaust physico-chemistry of Tallow biodiesel was the most different to diesel as well as the most toxic, with exposure resulting in significantly decreased cellular viability (95.8 ± 6.5%) and increased release of several immune mediators including IL-6 (+223.11 ± 368.83 pg/mL) and IL-8 (+1516.17 ± 2908.79 pg/mL) above Air controls. In contrast Canola biodiesel was the least toxic with exposure only increasing TNF-α (4.91 ± 8.61). CONCLUSION This study, which investigated the toxic effects for the largest range of biodiesels, shows that exposure to different exhausts results in a spectrum of toxic effects in vitro when combusted under identical conditions.
Collapse
Affiliation(s)
- Katherine R Landwehr
- Occupation, Environment and Safety, School of Population Health, Curtin University, PO Box U1987, Perth 6845, Western Australia, Australia; Respiratory Environmental Health, Wal-yan Respiratory Research Centre, Telethon Kids Institute, Perth Children's Hospital, Nedlands, Perth 6009, Western Australia, Australia.
| | - Jessica Hillas
- Respiratory Environmental Health, Wal-yan Respiratory Research Centre, Telethon Kids Institute, Perth Children's Hospital, Nedlands, Perth 6009, Western Australia, Australia
| | - Ryan Mead-Hunter
- Occupation, Environment and Safety, School of Population Health, Curtin University, PO Box U1987, Perth 6845, Western Australia, Australia
| | - Peter Brooks
- School of Science, Technology and Engineering, University of the Sunshine Coast, Sippy Downs, Queensland, Australia
| | - Andrew King
- Fluid Dynamics Research Group, School of Civil and Mechanical Engineering, Curtin University, Perth, Western Australia, Australia
| | - Rebecca A O'Leary
- Department of Primary Industries and Regional Development, Perth 6000, Western Australia, Australia
| | - Anthony Kicic
- Occupation, Environment and Safety, School of Population Health, Curtin University, PO Box U1987, Perth 6845, Western Australia, Australia; Respiratory Environmental Health, Wal-yan Respiratory Research Centre, Telethon Kids Institute, Perth Children's Hospital, Nedlands, Perth 6009, Western Australia, Australia; Department of Respiratory and Sleep Medicine, Perth Children's Hospital, Nedlands, Perth 6009, Western Australia, Australia; Centre for Cell Therapy and Regenerative Medicine, The University of Western Australia, Perth 6009, Western Australia, Australia
| | - Benjamin J Mullins
- Occupation, Environment and Safety, School of Population Health, Curtin University, PO Box U1987, Perth 6845, Western Australia, Australia
| | - Alexander N Larcombe
- Occupation, Environment and Safety, School of Population Health, Curtin University, PO Box U1987, Perth 6845, Western Australia, Australia; Respiratory Environmental Health, Wal-yan Respiratory Research Centre, Telethon Kids Institute, Perth Children's Hospital, Nedlands, Perth 6009, Western Australia, Australia
| |
Collapse
|
31
|
Three-Dimensional Airway Spheroids and Organoids for Cystic Fibrosis Research. JOURNAL OF RESPIRATION 2021. [DOI: 10.3390/jor1040022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cystic fibrosis (CF) is an autosomal recessive multi-organ disease caused by mutations in the CF Transmembrane Conductance Regulator (CFTR) gene, with morbidity and mortality primacy related to the lung disease. The CFTR protein, a chloride/bicarbonate channel, is expressed at the apical side of airway epithelial cells and is mainly involved in appropriate ion and fluid transport across the epithelium. Although many animal and cellular models have been developed to study the pathophysiological consequences of the lack/dysfunction of CFTR, only the three-dimensional (3D) structures termed “spheroids” and “organoids” can enable the reconstruction of airway mucosa to model organ development, disease pathophysiology, and drug screening. Airway spheroids and organoids can be derived from different sources, including adult lungs and induced pluripotent stem cells (iPSCs), each with its advantages and limits. Here, we review the major features of airway spheroids and organoids, anticipating that their potential in the CF field has not been fully shown. Further work is mandatory to understand whether they can accomplish better outcomes than other culture conditions of airway epithelial cells for CF personalized therapies and tissue engineering aims.
Collapse
|
32
|
Lee DDH, Cardinale D, Nigro E, Butler CR, Rutman A, Fassad MR, Hirst RA, Moulding D, Agrotis A, Forsythe E, Peckham D, Robson E, Smith CM, Somavarapu S, Beales PL, Hart SL, Janes SM, Mitchison HM, Ketteler R, Hynds RE, O'Callaghan C. Higher throughput drug screening for rare respiratory diseases: readthrough therapy in primary ciliary dyskinesia. Eur Respir J 2021; 58:13993003.00455-2020. [PMID: 33795320 PMCID: PMC8514977 DOI: 10.1183/13993003.00455-2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 03/01/2021] [Indexed: 11/17/2022]
Abstract
BACKGROUND Development of therapeutic approaches for rare respiratory diseases is hampered by the lack of systems that allow medium-to-high-throughput screening of fully differentiated respiratory epithelium from affected patients. This is a particular problem for primary ciliary dyskinesia (PCD), a rare genetic disease caused by mutations in genes that adversely affect ciliary movement and consequently mucociliary transport. Primary cell culture of basal epithelial cells from nasal brush biopsies followed by ciliated differentiation at the air-liquid interface (ALI) has proven to be a useful tool in PCD diagnostics but the technique's broader utility, including in pre-clinical PCD research, has been restricted by the limited number of basal cells that can be expanded from such biopsies. METHODS We describe an immunofluorescence screening method, enabled by extensive expansion of basal cells from PCD patients and the directed differentiation of these cells into ciliated epithelium in miniaturised 96-well transwell format ALI cultures. As proof-of-principle, we performed a personalised investigation in a patient with a rare and severe form of PCD (reduced generation of motile cilia), in this case caused by a homozygous nonsense mutation in the MCIDAS gene. RESULTS Initial analyses of ciliary ultrastructure, beat pattern and beat frequency in the 96-well transwell format ALI cultures indicate that a range of different PCD defects can be retained in these cultures. The screening system in our proof-of-principal investigation allowed drugs that induce translational readthrough to be evaluated alone or in combination with nonsense-mediated decay inhibitors. We observed restoration of basal body formation but not the generation of cilia in the patient's nasal epithelial cells in vitro. CONCLUSION: Our study provides a platform for higher throughput analyses of airway epithelia that is applicable in a range of settings and suggests novel avenues for drug evaluation and development in PCD caused by nonsense mutations.
Collapse
Affiliation(s)
- Dani Do Hyang Lee
- UCL Great Ormond Street Institute of Child Health, London, UK
- D.D.H. Lee and D. Cardinale contributed equally
| | - Daniela Cardinale
- UCL Great Ormond Street Institute of Child Health, London, UK
- D.D.H. Lee and D. Cardinale contributed equally
| | - Ersilia Nigro
- Lungs for Living Research Centre, UCL Respiratory, Division of Medicine, University College London, London, UK
| | - Colin R Butler
- Lungs for Living Research Centre, UCL Respiratory, Division of Medicine, University College London, London, UK
| | - Andrew Rutman
- Centre for PCD Diagnosis and Research, Dept of Respiratory Sciences, University of Leicester, Leicester, UK
| | - Mahmoud R Fassad
- Ciliary Disease Section, Genetics and Genomic Medicine Research and Teaching Dept, UCL Great Ormond Street Institute of Child Health, London, UK
- Dept of Human Genetics, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Robert A Hirst
- Centre for PCD Diagnosis and Research, Dept of Respiratory Sciences, University of Leicester, Leicester, UK
| | - Dale Moulding
- Developmental Biology and Cancer, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Alexander Agrotis
- MRC Laboratory for Molecular Cell Biology, University College London, London, UK
| | - Elisabeth Forsythe
- Ciliary Disease Section, Genetics and Genomic Medicine Research and Teaching Dept, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Daniel Peckham
- Leeds Institute for Medical Research, University of Leeds, Leeds, UK
| | - Evie Robson
- Leeds Institute for Medical Research, University of Leeds, Leeds, UK
| | - Claire M Smith
- UCL Great Ormond Street Institute of Child Health, London, UK
| | | | - Philip L Beales
- Ciliary Disease Section, Genetics and Genomic Medicine Research and Teaching Dept, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Stephen L Hart
- Ciliary Disease Section, Genetics and Genomic Medicine Research and Teaching Dept, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Sam M Janes
- Lungs for Living Research Centre, UCL Respiratory, Division of Medicine, University College London, London, UK
| | - Hannah M Mitchison
- Ciliary Disease Section, Genetics and Genomic Medicine Research and Teaching Dept, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Robin Ketteler
- MRC Laboratory for Molecular Cell Biology, University College London, London, UK
| | - Robert E Hynds
- Lungs for Living Research Centre, UCL Respiratory, Division of Medicine, University College London, London, UK
- UCL Cancer Institute, University College London, London, UK
- R.E. Hynds and C. O'Callaghan contributed equally to this article as lead authors and supervised the work
| | - Christopher O'Callaghan
- UCL Great Ormond Street Institute of Child Health, London, UK
- Centre for PCD Diagnosis and Research, Dept of Respiratory Sciences, University of Leicester, Leicester, UK
- R.E. Hynds and C. O'Callaghan contributed equally to this article as lead authors and supervised the work
| |
Collapse
|
33
|
Iszatt JJ, Larcombe AN, Chan HK, Stick SM, Garratt LW, Kicic A. Phage Therapy for Multi-Drug Resistant Respiratory Tract Infections. Viruses 2021; 13:v13091809. [PMID: 34578390 PMCID: PMC8472870 DOI: 10.3390/v13091809] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/08/2021] [Accepted: 09/09/2021] [Indexed: 12/30/2022] Open
Abstract
The emergence of multi-drug resistant (MDR) bacteria is recognised today as one of the greatest challenges to public health. As traditional antimicrobials are becoming ineffective and research into new antibiotics is diminishing, a number of alternative treatments for MDR bacteria have been receiving greater attention. Bacteriophage therapies are being revisited and present a promising opportunity to reduce the burden of bacterial infection in this post-antibiotic era. This review focuses on the current evidence supporting bacteriophage therapy against prevalent or emerging multi-drug resistant bacterial pathogens in respiratory medicine and the challenges ahead in preclinical data generation. Starting with efforts to improve delivery of bacteriophages to the lung surface, the current developments in animal models for relevant efficacy data on respiratory infections are discussed before finishing with a summary of findings from the select human trials performed to date.
Collapse
Affiliation(s)
- Joshua J. Iszatt
- Occupation, Environment and Safety, School of Population Health, Curtin University, Perth 6845, Australia; (J.J.I.); (A.N.L.)
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Perth 6009, Australia; (S.M.S.); (L.W.G.)
| | - Alexander N. Larcombe
- Occupation, Environment and Safety, School of Population Health, Curtin University, Perth 6845, Australia; (J.J.I.); (A.N.L.)
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Perth 6009, Australia; (S.M.S.); (L.W.G.)
| | - Hak-Kim Chan
- Advanced Drug Delivery Group, Sydney Pharmacy School, University of Sydney, Camperdown 2006, Australia;
| | - Stephen M. Stick
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Perth 6009, Australia; (S.M.S.); (L.W.G.)
- Department of Respiratory and Sleep Medicine, Perth Children’s Hospital, Perth 6009, Australia
- Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, Harry Perkins Institute of Medical Research, The University of Western Australia, Nedlands 6009, Australia
| | - Luke W. Garratt
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Perth 6009, Australia; (S.M.S.); (L.W.G.)
| | - Anthony Kicic
- Occupation, Environment and Safety, School of Population Health, Curtin University, Perth 6845, Australia; (J.J.I.); (A.N.L.)
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Perth 6009, Australia; (S.M.S.); (L.W.G.)
- Department of Respiratory and Sleep Medicine, Perth Children’s Hospital, Perth 6009, Australia
- Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, Harry Perkins Institute of Medical Research, The University of Western Australia, Nedlands 6009, Australia
- Correspondence:
| |
Collapse
|
34
|
Han F, Li S, Yang Y, Bai Z. Interleukin-6 promotes ferroptosis in bronchial epithelial cells by inducing reactive oxygen species-dependent lipid peroxidation and disrupting iron homeostasis. Bioengineered 2021; 12:5279-5288. [PMID: 34402724 PMCID: PMC8806540 DOI: 10.1080/21655979.2021.1964158] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Asthma occurs accompanied by the ferroptosis in bronchial epithelial cells, during which Interleukin-6 (IL-6) plays a key role. However, the associations between IL-6, ferroptosis and asthma have not been reported. Bronchial epithelial cells BEAS-2B cells were induced by different concentrations of IL-6 and cell viability was detected by MTT assay. The TBARS production rate was detected by corresponding kit. The expression of oxidative stress-related indexes was detected by ELISA. The Iron Assay Kits detected total iron levels and ferrous ion (Fe2+) levels. Labile iron pool assay was used to detect the cell unstable iron pool. The expression of ferroptosis-related proteins was detected by Western blot. To further examine the mechanism of action, ferroptosis inhibitor Ferrostatin 1 (Fer-1), antioxidant NAC, and the iron supplement Fe were added. We found that IL-6 decreased the activity, promoted lipid peroxidation, disrupted iron homeostasis of BEAS-2B cells, and induced iron death in bronchial epithelial BEAS-2B cells. However, pretreatment with Ferrostatin-1 (Fer-1) and antioxidant NAC partially reversed the effect of IL-6 on lipid peroxidation and ferroptosis in BEAS-2B cells, while Fe augmented the effect. Overall, IL-6 promotes ferroptosis in bronchial epithelial cells by inducing reactive oxygen species (ROS)-dependent lipid peroxidation and disrupting iron homeostasis.
Collapse
Affiliation(s)
- Fei Han
- The Key Laboratory Of Industrial Biotechnology, Ministry Of Education, School Of Biotechnology, jiangnan university, Jiangsu, China.,National Engineering Laboratory For Cereal Fermentation Technology, Jiangnan University, Jiangsu, China.,Jiangsu Provincial Research Center For Bioactive Product Processing Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Shijie Li
- The Key Laboratory Of Industrial Biotechnology, Ministry Of Education, School Of Biotechnology, jiangnan university, Jiangsu, China.,National Engineering Laboratory For Cereal Fermentation Technology, Jiangnan University, Jiangsu, China.,Jiangsu Provincial Research Center For Bioactive Product Processing Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Yankun Yang
- The Key Laboratory Of Industrial Biotechnology, Ministry Of Education, School Of Biotechnology, jiangnan university, Jiangsu, China.,National Engineering Laboratory For Cereal Fermentation Technology, Jiangnan University, Jiangsu, China.,Jiangsu Provincial Research Center For Bioactive Product Processing Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Zhonghu Bai
- The Key Laboratory Of Industrial Biotechnology, Ministry Of Education, School Of Biotechnology, jiangnan university, Jiangsu, China.,National Engineering Laboratory For Cereal Fermentation Technology, Jiangnan University, Jiangsu, China.,Jiangsu Provincial Research Center For Bioactive Product Processing Technology, Jiangnan University, Wuxi, Jiangsu, China
| |
Collapse
|
35
|
McLean SA, Cullen L, Gardam DJ, Schofield CJ, Laucirica DR, Sutanto EN, Ling KM, Stick SM, Peacock CS, Kicic A, Garratt LW. Cystic Fibrosis Clinical Isolates of Aspergillus fumigatus Induce Similar Muco-inflammatory Responses in Primary Airway Epithelial Cells. Pathogens 2021; 10:pathogens10081020. [PMID: 34451484 PMCID: PMC8399118 DOI: 10.3390/pathogens10081020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/03/2021] [Accepted: 08/11/2021] [Indexed: 11/16/2022] Open
Abstract
Aspergillus is increasingly associated with lung inflammation and mucus plugging in early cystic fibrosis (CF) disease during which conidia burden is low and strains appear to be highly diverse. It is unknown whether clinical Aspergillus strains vary in their capacity to induce epithelial inflammation and mucus production. We tested the hypothesis that individual colonising strains of Aspergillus fumigatus would induce different responses. Ten paediatric CF Aspergillus isolates were compared along with two systemically invasive clinical isolates and an ATCC reference strain. Isolates were first characterised by ITS gene sequencing and screened for antifungal susceptibility. Three clusters (A-C) of Aspergillus isolates were identified by ITS. Antifungal susceptibility was variable, particularly for itraconazole. Submerged CF and non-CF monolayers as well as differentiated primary airway epithelial cell cultures were incubated with conidia for 24 h to allow germination. None of the clinical isolates were found to significantly differ from one another in either IL-6 or IL-8 release or gene expression of secretory mucins. Clinical Aspergillus isolates appear to be largely homogenous in their mucostimulatory and immunostimulatory capacities and, therefore, only the antifungal resistance characteristics are likely to be clinically important.
Collapse
Affiliation(s)
- Samantha A. McLean
- Wal-Yan Respiratory Research Centre, Telethon Kids Institute, University of Western Australia, Crawley 6009, Australia; (S.A.M.); (C.J.S.); (D.R.L.); (E.N.S.); (K.-M.L.); (S.M.S.); (A.K.)
| | - Leilani Cullen
- Faculty of Health and Medical Sciences, University of Western Australia, Crawley 6009, Australia; (L.C.); (C.S.P.)
| | - Dianne J. Gardam
- PathWest Laboratory Medicine WA, Fiona Stanley Hospital, Murdoch 6150, Australia;
| | - Craig J. Schofield
- Wal-Yan Respiratory Research Centre, Telethon Kids Institute, University of Western Australia, Crawley 6009, Australia; (S.A.M.); (C.J.S.); (D.R.L.); (E.N.S.); (K.-M.L.); (S.M.S.); (A.K.)
| | - Daniel R. Laucirica
- Wal-Yan Respiratory Research Centre, Telethon Kids Institute, University of Western Australia, Crawley 6009, Australia; (S.A.M.); (C.J.S.); (D.R.L.); (E.N.S.); (K.-M.L.); (S.M.S.); (A.K.)
- Faculty of Health and Medical Sciences, University of Western Australia, Crawley 6009, Australia; (L.C.); (C.S.P.)
| | - Erika N. Sutanto
- Wal-Yan Respiratory Research Centre, Telethon Kids Institute, University of Western Australia, Crawley 6009, Australia; (S.A.M.); (C.J.S.); (D.R.L.); (E.N.S.); (K.-M.L.); (S.M.S.); (A.K.)
| | - Kak-Ming Ling
- Wal-Yan Respiratory Research Centre, Telethon Kids Institute, University of Western Australia, Crawley 6009, Australia; (S.A.M.); (C.J.S.); (D.R.L.); (E.N.S.); (K.-M.L.); (S.M.S.); (A.K.)
- Faculty of Health and Medical Sciences, University of Western Australia, Crawley 6009, Australia; (L.C.); (C.S.P.)
| | - Stephen M. Stick
- Wal-Yan Respiratory Research Centre, Telethon Kids Institute, University of Western Australia, Crawley 6009, Australia; (S.A.M.); (C.J.S.); (D.R.L.); (E.N.S.); (K.-M.L.); (S.M.S.); (A.K.)
- Faculty of Health and Medical Sciences, University of Western Australia, Crawley 6009, Australia; (L.C.); (C.S.P.)
- Department of Respiratory and Sleep Medicine, Perth Children’s Hospital, Nedlands 6009, Australia
| | - Christopher S. Peacock
- Faculty of Health and Medical Sciences, University of Western Australia, Crawley 6009, Australia; (L.C.); (C.S.P.)
| | - Anthony Kicic
- Wal-Yan Respiratory Research Centre, Telethon Kids Institute, University of Western Australia, Crawley 6009, Australia; (S.A.M.); (C.J.S.); (D.R.L.); (E.N.S.); (K.-M.L.); (S.M.S.); (A.K.)
- Faculty of Health and Medical Sciences, University of Western Australia, Crawley 6009, Australia; (L.C.); (C.S.P.)
- Department of Respiratory and Sleep Medicine, Perth Children’s Hospital, Nedlands 6009, Australia
- Occupation and Environment, School of Public Health, Curtin University, Bentley 6102, Australia
| | - Luke W. Garratt
- Wal-Yan Respiratory Research Centre, Telethon Kids Institute, University of Western Australia, Crawley 6009, Australia; (S.A.M.); (C.J.S.); (D.R.L.); (E.N.S.); (K.-M.L.); (S.M.S.); (A.K.)
- Correspondence:
| | | | | |
Collapse
|
36
|
Girkin J, Loo SL, Esneau C, Maltby S, Mercuri F, Chua B, Reid AT, Veerati PC, Grainge CL, Wark PAB, Knight D, Jackson D, Demaison C, Bartlett NW. TLR2-mediated innate immune priming boosts lung anti-viral immunity. Eur Respir J 2021; 58:13993003.01584-2020. [PMID: 33303547 DOI: 10.1183/13993003.01584-2020] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 11/27/2020] [Indexed: 01/07/2023]
Abstract
BACKGROUND We assessed whether Toll-like receptor (TLR)2 activation boosts the innate immune response to rhinovirus infection, as a treatment strategy for virus-induced respiratory diseases. METHODS We employed treatment with a novel TLR2 agonist (INNA-X) prior to rhinovirus infection in mice, and INNA-X treatment in differentiated human bronchial epithelial cells derived from asthmatic-donors. We assessed viral load, immune cell recruitment, cytokines, type I and III interferon (IFN) production, as well as the lung tissue and epithelial cell immune transcriptome. RESULTS We show, in vivo, that a single INNA-X treatment induced innate immune priming characterised by low-level IFN-λ, Fas ligand, chemokine expression and airway lymphocyte recruitment. Treatment 7 days before infection significantly reduced lung viral load, increased IFN-β/λ expression and inhibited neutrophilic inflammation. Corticosteroid treatment enhanced the anti-inflammatory effects of INNA-X. Treatment 1 day before infection increased expression of 190 lung tissue immune genes. This tissue gene expression signature was absent with INNA-X treatment 7 days before infection, suggesting an alternate mechanism, potentially via establishment of immune cell-mediated mucosal innate immunity. In vitro, INNA-X treatment induced a priming response defined by upregulated IFN-λ, chemokine and anti-microbial gene expression that preceded an accelerated response to infection enriched for nuclear factor (NF)-κB-regulated genes and reduced viral loads, even in epithelial cells derived from asthmatic donors with intrinsic delayed anti-viral immune response. CONCLUSION Airway epithelial cell TLR2 activation induces prolonged innate immune priming, defined by early NF-κB activation, IFN-λ expression and lymphocyte recruitment. This response enhanced anti-viral innate immunity and reduced virus-induced airway inflammation.
Collapse
Affiliation(s)
- Jason Girkin
- Viral Immunology and Respiratory Disease group, University of Newcastle, Newcastle, Australia.,Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia.,These authors contributed equally
| | - Su-Ling Loo
- Viral Immunology and Respiratory Disease group, University of Newcastle, Newcastle, Australia.,Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia.,These authors contributed equally
| | - Camille Esneau
- Viral Immunology and Respiratory Disease group, University of Newcastle, Newcastle, Australia.,Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia
| | - Steven Maltby
- Viral Immunology and Respiratory Disease group, University of Newcastle, Newcastle, Australia.,Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia
| | | | - Brendon Chua
- Dept of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Andrew T Reid
- Viral Immunology and Respiratory Disease group, University of Newcastle, Newcastle, Australia.,Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia
| | - Punnam Chander Veerati
- Viral Immunology and Respiratory Disease group, University of Newcastle, Newcastle, Australia.,Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia
| | - Chris L Grainge
- Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia.,Dept of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, Australia
| | - Peter A B Wark
- Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia.,Dept of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, Australia
| | - Darryl Knight
- Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia
| | - David Jackson
- Dept of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | | | - Nathan W Bartlett
- Viral Immunology and Respiratory Disease group, University of Newcastle, Newcastle, Australia .,Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia
| |
Collapse
|
37
|
Hillas J, Evans DJ, Ang S, Iosifidis T, Garratt LW, Hemy N, Kicic-Starcevich E, Simpson SJ, Kicic A. Nasal airway epithelial repair after very preterm birth. ERJ Open Res 2021; 7:00913-2020. [PMID: 34109241 PMCID: PMC8181665 DOI: 10.1183/23120541.00913-2020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 03/27/2021] [Indexed: 12/03/2022] Open
Abstract
Preterm birth rates are increasing and now account for >11% of global births. Simultaneously, advances in neonatal care have led to increased survival of lower gestation neonates. A complication of preterm birth, and the biggest determinant of survival, is lung and airway immaturity. After preterm birth, the immature respiratory system is exposed to pro-inflammatory stimuli like injury from resuscitation and oxygen toxicity. The airway epithelium, the physical barrier between insults and the airways, is particularly vulnerable to injury. If epithelial barrier integrity cannot be restored rapidly following damage (i.e. via aberrant repair), the respiratory system is left unprotected, increasing the risk of infection, inflammation and tissue damage. Altered epithelial repair may play an important role in the ongoing respiratory health problems experienced by preterm survivors, including severe respiratory infections throughout early life, or low and declining lung function [1–3]. Deficits are further exacerbated in those with bronchopulmonary dysplasia (BPD). The mechanisms contributing to ongoing respiratory problems are currently unknown, although probably begin in early life. Until now, understanding the role of the preterm epithelial barrier has been limited by a lack of appropriate cellular models. Our study aimed to assess the reparative capacity of the airway epithelium in survivors of preterm birth and its association with early life outcomes, with the hypothesis that preterm airway epithelial cells have an abnormal repair mechanism. Nasal epithelialcells from very preterm infants have a functional defect in their ability to repair beyond the first year of life, and failed repair may be associated with antenatal steroid exposurehttps://bit.ly/39OFJs7
Collapse
Affiliation(s)
- Jessica Hillas
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Nedlands, Australia
| | - Denby J Evans
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Nedlands, Australia.,Occupation and Environment, School of Public Health, Curtin University, Bentley, Australia
| | - Sherlynn Ang
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Nedlands, Australia
| | - Thomas Iosifidis
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Nedlands, Australia.,Occupation and Environment, School of Public Health, Curtin University, Bentley, Australia.,Centre for Cell Therapy and Regenerative Medicine, University of Western Australia, Nedlands, Australia
| | - Luke W Garratt
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Nedlands, Australia
| | - Naomi Hemy
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Nedlands, Australia
| | | | - Shannon J Simpson
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Nedlands, Australia.,School of Physiotherapy and Exercise Science, Curtin University, Bentley, Australia.,These authors contributed equally
| | - Anthony Kicic
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Nedlands, Australia.,Occupation and Environment, School of Public Health, Curtin University, Bentley, Australia.,Centre for Cell Therapy and Regenerative Medicine, University of Western Australia, Nedlands, Australia.,Dept of Respiratory and Sleep Medicine, Perth Children's Hospital, Nedlands, Australia.,These authors contributed equally
| |
Collapse
|
38
|
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
|
39
|
Wu M, Hong G, Chen Y, Ye L, Zhang K, Cai K, Yang H, Long X, Gao W, Li H. Personalized drug testing in a patient with non-small-cell lung cancer using cultured cancer cells from pleural effusion. J Int Med Res 2021; 48:300060520955058. [PMID: 32954884 PMCID: PMC7509736 DOI: 10.1177/0300060520955058] [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] [Indexed: 12/03/2022] Open
Abstract
Objective Patients with non-small-cell lung cancer (NSCLC) and primary or acquired resistance do not respond to targeted drugs. We explored whether cancer cells can be cultured from liquid biopsies from patients with primary resistance to tyrosine kinase inhibitors (TKIs). We aimed to predict patients’ responses to drugs according to in vitro drug testing results. Methods Cancer cell cultures were established from the pleural effusion of a patient with TKI-resistant NSCLC using a conditional reprogramming technique. Phenotypic drug sensitivity tests were performed using the Cell Counting Kit-8 assay. We tested individual drugs and compared the synergistic and inhibitory effects of drug combinations. Results The results of our in vitro sensitivity test using the combination of cisplatin and pemetrexed were correlated with the patient’s response. Conclusion This represents the first successful report of predictive testing for combination therapy in patients with epidermal growth factor receptor-mutant NSCLC and primary TKI resistance. This strategy should be applicable to both chemotherapies and targeted therapies, and it will significantly improve the clinical treatment and management of patients with NSCLC and primary or acquired resistance to targeted therapies, as well as patients lacking targetable mutations.
Collapse
Affiliation(s)
- Ming Wu
- Wuhan University Shenzhen Institute, Shenzhen, Guangdong, China
| | - Guodai Hong
- Shenzhen Luohu People's Hospital, Shenzhen, Guangdong, China
| | - Yu Chen
- Wuhan University Shenzhen Institute, Shenzhen, Guangdong, China
| | - Lina Ye
- Wuhan University Shenzhen Institute, Shenzhen, Guangdong, China
| | - Kang Zhang
- Wuhan University Shenzhen Institute, Shenzhen, Guangdong, China
| | - Kaihong Cai
- Wuhan University Shenzhen Institute, Shenzhen, Guangdong, China
| | - Huadong Yang
- Wuhan University Shenzhen Institute, Shenzhen, Guangdong, China
| | - Xiang Long
- Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Wenbin Gao
- Shenzhen Luohu People's Hospital, Shenzhen, Guangdong, China
| | - Hui Li
- Wuhan University Shenzhen Institute, Shenzhen, Guangdong, China
| |
Collapse
|
40
|
Campbell DR, Senger CN, Ryan AL, Magin CM. Engineering Tissue-Informed Biomaterials to Advance Pulmonary Regenerative Medicine. Front Med (Lausanne) 2021; 8:647834. [PMID: 33898484 PMCID: PMC8060451 DOI: 10.3389/fmed.2021.647834] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 03/09/2021] [Indexed: 11/13/2022] Open
Abstract
Biomaterials intentionally designed to support the expansion, differentiation, and three-dimensional (3D) culture of induced-pluripotent stem cells (iPSCs) may pave the way to cell-based therapies for chronic respiratory diseases. These conditions are endured by millions of people worldwide and represent a significant cause of morbidity and mortality. Currently, there are no effective treatments for the majority of advanced lung diseases and lung transplantation remains the only hope for many chronically ill patients. Key opinion leaders speculate that the novel coronavirus, COVID-19, may lead to long-term lung damage, further exacerbating the need for regenerative therapies. New strategies for regenerative cell-based therapies harness the differentiation capability of human iPSCs for studying pulmonary disease pathogenesis and treatment. Excitingly, biomaterials are a cell culture platform that can be precisely designed to direct stem cell differentiation. Here, we present a closer look at the state-of-the-art of iPSC differentiation for pulmonary engineering, offer evidence supporting the power of biomaterials to improve stem cell differentiation, and discuss our perspective on the potential for tissue-informed biomaterials to transform pulmonary regenerative medicine.
Collapse
Affiliation(s)
- Donald R. Campbell
- Department of Bioengineering, Denver, Anschutz Medical Campus, University of Colorado, Aurora, CO, United States
| | - Christiana N. Senger
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Hastings Center for Pulmonary Research, University of Southern California, Los Angeles, CA, United States
| | - Amy L. Ryan
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Hastings Center for Pulmonary Research, University of Southern California, Los Angeles, CA, United States
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA, United States
| | - Chelsea M. Magin
- Department of Bioengineering, Denver, Anschutz Medical Campus, University of Colorado, Aurora, CO, United States
- Department of Pediatrics, Anschutz Medical Campus, University of Colorado, Aurora, CO, United States
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO, United States
| |
Collapse
|
41
|
Ballesteros OR, Brooks PT, Haastrup EK, Fischer-Nielsen A, Munthe-Fog L, Svalgaard JD. Adipose-Derived Stromal/Stem Cell Culture: Effects of Different Concentrations of Human Platelet Lysate in Media. Cells Tissues Organs 2021; 209:257-265. [PMID: 33752213 DOI: 10.1159/000513604] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 12/06/2020] [Indexed: 11/19/2022] Open
Abstract
Adipose-derived stromal/stem cells (ASCs) are being tested as a possible treatment for a wide range of diseases to exploit the immunomodulatory and regenerative potential demonstrated in vitro. Pooled human platelet lysate (pHPL) has replaced fetal bovine serum (FBS) as the preferred growth supplement because of its xeno-free origin and improved cell proliferation. Much has been done toward reducing the concentration of pHPL required when expanding ASCs. However, little is known on how increasing the concentration of pHPL affects ASC potency, which could lead to changes with possible beneficial applications. This study investigated the effect of 5, 10, or 20% pHPL in culture media on ASC proliferation and phenotypic marker expression, including chemokine receptors CXCR2, CXCR3, CXCR4, and VLA-4. Adipogenic and osteogenic properties, as well as immunosuppressive properties, including the ability to induce indoleamine-pyrrole 2,3-dioxygenase 1 (IDO1) and suppress T cell proliferation, were also examined. We observed a significant increase in cell yield (approximately 2-fold) and a corresponding reduction in population doubling time and cell volume when doubling the concentration of pHPL in the growth media. ASCs maintained expression of phenotypic surface markers CD73, CD90, and CD105 and were negative for CD45 and CD31. The ability to induce IDO1 and suppress T cell proliferation was observed as well. Adipogenesis and osteogenesis, however, seem to be increased at higher concentrations of pHPL (20% > 10% > 5%), while expression of chemokine receptors CXCR2 and CXCR3 was lower. In conclusion, increasing the pHPL concentration to 20% could be used to optimize culture conditions when producing cells for clinical treatments and may even be used to enhance beneficial ASC properties depending on the desired therapeutic effect.
Collapse
Affiliation(s)
- Olga R Ballesteros
- Department of Clinical Immunology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Patrick T Brooks
- Department of Clinical Immunology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Eva K Haastrup
- Department of Clinical Immunology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Anne Fischer-Nielsen
- Department of Clinical Immunology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Lea Munthe-Fog
- Department of Clinical Immunology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Jesper D Svalgaard
- Department of Clinical Immunology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark,
| |
Collapse
|
42
|
Allan KM, Farrow N, Donnelley M, Jaffe A, Waters SA. Treatment of Cystic Fibrosis: From Gene- to Cell-Based Therapies. Front Pharmacol 2021; 12:639475. [PMID: 33796025 PMCID: PMC8007963 DOI: 10.3389/fphar.2021.639475] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 01/27/2021] [Indexed: 12/11/2022] Open
Abstract
Prognosis of patients with cystic fibrosis (CF) varies extensively despite recent advances in targeted therapies that improve CF transmembrane conductance regulator (CFTR) function. Despite being a multi-organ disease, extensive lung tissue destruction remains the major cause of morbidity and mortality. Progress towards a curative treatment strategy that implements a CFTR gene addition-technology to the patients’ lungs has been slow and not yet developed beyond clinical trials. Improved delivery vectors are needed to overcome the body’s defense system and ensure an efficient and consistent clinical response before gene therapy is suitable for clinical care. Cell-based therapy–which relies on functional modification of allogenic or autologous cells ex vivo, prior to transplantation into the patient–is now a therapeutic reality for various diseases. For CF, pioneering research has demonstrated proof-of-principle for allogenic transplantation of cultured human airway stem cells into mouse airways. However, applying a cell-based therapy to the human airways has distinct challenges. We review CF gene therapies using viral and non-viral delivery strategies and discuss current advances towards autologous cell-based therapies. Progress towards identification, correction, and expansion of a suitable regenerative cell, as well as refinement of pre-cell transplant lung conditioning protocols is discussed.
Collapse
Affiliation(s)
- Katelin M Allan
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Sydney, Australia.,Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC), University of New South Wales and Sydney Children's Hospital, Sydney, Australia
| | - Nigel Farrow
- Respiratory and Sleep Medicine, Women's and Children's Health Network, Adelaide, Australia.,Robinson Research Institute, The University of Adelaide, Adelaide, Australia.,Adelaide Medical School, The University of Adelaide, Adelaide, Australia
| | - Martin Donnelley
- Respiratory and Sleep Medicine, Women's and Children's Health Network, Adelaide, Australia.,Robinson Research Institute, The University of Adelaide, Adelaide, Australia.,Adelaide Medical School, The University of Adelaide, Adelaide, Australia
| | - Adam Jaffe
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Sydney, Australia.,Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC), University of New South Wales and Sydney Children's Hospital, Sydney, Australia.,Department of Respiratory Medicine, Sydney Children's Hospital, Sydney, Australia
| | - Shafagh A Waters
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Sydney, Australia.,Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC), University of New South Wales and Sydney Children's Hospital, Sydney, Australia.,Department of Respiratory Medicine, Sydney Children's Hospital, Sydney, Australia
| |
Collapse
|
43
|
Villamizar O, Waters SA, Scott T, Grepo N, Jaffe A, Morris KV. Mesenchymal Stem Cell exosome delivered Zinc Finger Protein activation of cystic fibrosis transmembrane conductance regulator. J Extracell Vesicles 2021; 10:e12053. [PMID: 33532041 PMCID: PMC7825549 DOI: 10.1002/jev2.12053] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 12/13/2020] [Accepted: 12/16/2020] [Indexed: 02/06/2023] Open
Abstract
Cystic fibrosis is a genetic disorder that results in a multi-organ disease with progressive respiratory decline which leads to premature death. Mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene disrupts the capacity of the protein to function as a channel, transporting chloride ions and bicarbonate across epithelial cell membranes. Small molecule treatments targeted at potentiating or correcting CFTR have shown clinical benefits, but are only effective for a small percentage of individuals with specific CFTR mutations. To overcome this limitation, we engineered stromal-derived mesenchymal stem cells (MSC) and HEK293 cells to produce exosomes containing a novel CFTR Zinc Finger Protein fusion with transcriptional activation domains VP64, P65 and Rta to target the CFTR promoter (CFZF-VPR) and activate transcription. Treatment with CFZF-VPR results in robust activation of CFTR transcription in patient derived Human Bronchial Epithelial cells (HuBEC). We also find that CFZF-VPR can be packaged into MSC and HEK293 cell exosomes and delivered to HuBEC cells to potently activate CFTR expression. Connexin 43 appeared to be required for functional release of CFZF-VPR from exosomes. The observations presented here demonstrate that MSC derived exosomes can be used to deliver a packaged zinc finger activator to target cells and activate CFTR. The novel approach presented here offers a next-generation genetic therapy that may one day prove effective in treating patients afflicted with Cystic fibrosis.
Collapse
Affiliation(s)
- Olga Villamizar
- Center for Gene Therapy City of Hope-Beckman Research Institute at the City of Hope Duarte California USA
| | - Shafagh A Waters
- Faculty of Medicine School of Women's & Children's Health University of New South Wales (UNSW) Sydney NSW Australia.,Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC) Faculty of Medicine University of New South Wales Sydney NSW Australia.,Department of Respiratory Medicine Sydney Children's Hospital Sydney NSW Australia
| | - Tristan Scott
- Center for Gene Therapy City of Hope-Beckman Research Institute at the City of Hope Duarte California USA
| | - Nicole Grepo
- Center for Gene Therapy City of Hope-Beckman Research Institute at the City of Hope Duarte California USA
| | - Adam Jaffe
- Faculty of Medicine School of Women's & Children's Health University of New South Wales (UNSW) Sydney NSW Australia.,Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC) Faculty of Medicine University of New South Wales Sydney NSW Australia.,Department of Respiratory Medicine Sydney Children's Hospital Sydney NSW Australia
| | - Kevin V Morris
- Center for Gene Therapy City of Hope-Beckman Research Institute at the City of Hope Duarte California USA.,School of Medical Science Griffith University, Gold Coast Campus 1 Parklands Dr Southport QLD Australia
| |
Collapse
|
44
|
Ng RN, Tai AS, Chang BJ, Stick SM, Kicic A. Overcoming Challenges to Make Bacteriophage Therapy Standard Clinical Treatment Practice for Cystic Fibrosis. Front Microbiol 2021; 11:593988. [PMID: 33505366 PMCID: PMC7829477 DOI: 10.3389/fmicb.2020.593988] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 12/08/2020] [Indexed: 12/17/2022] Open
Abstract
Individuals with cystic fibrosis (CF) are given antimicrobials as prophylaxis against bacterial lung infection, which contributes to the growing emergence of multidrug resistant (MDR) pathogens isolated. Pathogens such as Pseudomonas aeruginosa that are commonly isolated from individuals with CF are armed with an arsenal of protective and virulence mechanisms, complicating eradication and treatment strategies. While translation of phage therapy into standard care for CF has been explored, challenges such as the lack of an appropriate animal model demonstrating safety in vivo exist. In this review, we have discussed and provided some insights in the use of primary airway epithelial cells to represent the mucoenvironment of the CF lungs to demonstrate safety and efficacy of phage therapy. The combination of phage therapy and antimicrobials is gaining attention and has the potential to delay the onset of MDR infections. It is evident that efforts to translate phage therapy into standard clinical practice have gained traction in the past 5 years. Ultimately, collaboration, transparency in data publications and standardized policies are needed for clinical translation.
Collapse
Affiliation(s)
- Renee N. Ng
- School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia
- Wal-yan Respiratory Research Center, Telethon Kids Institute, The University of Western Australia, Crawley, WA, Australia
| | - Anna S. Tai
- Department of Respiratory Medicine, Sir Charles Gairdner Hospital, Perth, WA, Australia
- Institute for Respiratory Health, School of Medicine, The University of Western Australia, Perth, WA, Australia
| | - Barbara J. Chang
- The Marshall Center for Infectious Diseases Research and Training, School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia
| | - Stephen M. Stick
- Wal-yan Respiratory Research Center, Telethon Kids Institute, The University of Western Australia, Crawley, WA, Australia
- Department of Respiratory and Sleep Medicine, Perth Children’s Hospital, Perth, WA, Australia
- Center for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia and Harry Perkins Institute of Medical Research, Perth, WA, Australia
| | - Anthony Kicic
- Wal-yan Respiratory Research Center, Telethon Kids Institute, The University of Western Australia, Crawley, WA, Australia
- Department of Respiratory and Sleep Medicine, Perth Children’s Hospital, Perth, WA, Australia
- Center for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia and Harry Perkins Institute of Medical Research, Perth, WA, Australia
- Occupation and the Environment, School of Public Health, Curtin University, Perth, WA, Australia
| |
Collapse
|
45
|
Awatade NT, Wong SL, Capraro A, Pandzic E, Slapetova I, Zhong L, Turgutoglu N, Fawcett LK, Whan RM, Jaffe A, Waters SA. Significant functional differences in differentiated Conditionally Reprogrammed (CRC)- and Feeder-free Dual SMAD inhibited-expanded human nasal epithelial cells. J Cyst Fibros 2021; 20:364-371. [PMID: 33414087 DOI: 10.1016/j.jcf.2020.12.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 12/16/2020] [Accepted: 12/20/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Patient-derived airway cells differentiated at Air Liquid Interface (ALI) are valuable models for Cystic fibrosis (CF) precision therapy. Different culture expansion methods have been established to extend expansion capacity of airway basal cells, while retaining functional airway epithelium physiology. Considerable variation in response to CFTR modulators is observed in cultures even within the same CFTR genotype and despite the use of similar ALI culture techniques. We aimed to address culture expansion method impact on differentiation. METHODS Nasal epithelial brushings from 14 individuals (CF=9; non-CF=5) were collected, then equally divided and expanded under conditional reprogramming culture (CRC) and feeder-serum-free "dual-SMAD inhibition" (SMADi) methods. Expanded cells from each culture were differentiated with proprietary PneumaCult™-ALI media. Morphology (Immunofluorescence), global proteomics (LC-MS/MS) and function (barrier integrity, cilia motility, and ion transport) were compared in CRCALI and SMADiALI under basal and CFTR corrector treated (VX-809) conditions. RESULTS No significant difference in the structural morphology or baseline global proteomics profile were observed. Barrier integrity and cilia motility were significantly different, despite no difference in cell junction morphology or cilia abundance. Epithelial Sodium Channels and Calcium-activated Chloride Channel activity did not differ but CFTR mediated chloride currents were significantly reduced in SMADiALI compare to their CRCALI counterparts. CONCLUSION Alteration of cellular physiological function in vitro were more prominent than structural and differentiation potential in airway ALI. Since initial expansion culture conditions significantly influence CFTR activity, this could lead to false conclusions if data from different labs are compared against each other without specific reference ranges.
Collapse
Affiliation(s)
- Nikhil T Awatade
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia; Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC), University of New South Wales and Sydney Children's Hospital, Sydney, NSW, Australia
| | - Sharon L Wong
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia; Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC), University of New South Wales and Sydney Children's Hospital, Sydney, NSW, Australia
| | - Alexander Capraro
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia; Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC), University of New South Wales and Sydney Children's Hospital, Sydney, NSW, Australia
| | - Elvis Pandzic
- Biomedical Imaging Facility, University of New South Wales, Sydney, NSW, Australia
| | - Iveta Slapetova
- Biomedical Imaging Facility, University of New South Wales, Sydney, NSW, Australia
| | - Ling Zhong
- Bioanalytical Mass Spectrometry Facility, University of New South Wales, Sydney, NSW, Australia
| | - Nihan Turgutoglu
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia; Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC), University of New South Wales and Sydney Children's Hospital, Sydney, NSW, Australia
| | - Laura K Fawcett
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia; Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC), University of New South Wales and Sydney Children's Hospital, Sydney, NSW, Australia; Department of Respiratory Medicine, Sydney Children's Hospital, Sydney, NSW, Australia
| | - Renee M Whan
- Biomedical Imaging Facility, University of New South Wales, Sydney, NSW, Australia
| | - Adam Jaffe
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia; Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC), University of New South Wales and Sydney Children's Hospital, Sydney, NSW, Australia; Department of Respiratory Medicine, Sydney Children's Hospital, Sydney, NSW, Australia
| | - Shafagh A Waters
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia; Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC), University of New South Wales and Sydney Children's Hospital, Sydney, NSW, Australia; Department of Respiratory Medicine, Sydney Children's Hospital, Sydney, NSW, Australia.
| |
Collapse
|
46
|
Novel Antioxidant Therapy with the Immediate Precursor to Glutathione, γ-Glutamylcysteine (GGC), Ameliorates LPS-Induced Cellular Stress in In Vitro 3D-Differentiated Airway Model from Primary Cystic Fibrosis Human Bronchial Cells. Antioxidants (Basel) 2020; 9:antiox9121204. [PMID: 33266084 PMCID: PMC7760366 DOI: 10.3390/antiox9121204] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/25/2020] [Accepted: 11/25/2020] [Indexed: 01/21/2023] Open
Abstract
Systemic glutathione deficiency, inflammation, and oxidative stress are hallmarks of cystic fibrosis (CF), an inherited disease that causes persistent lung infections and severe damage to the respiratory system and many of the body organs. Improvements to current antioxidant therapeutic strategies are needed. The dietary supplement, γ-glutamylcysteine (GGC), which is the immediate precursor to glutathione, rapidly boosts cellular glutathione levels following a single dose in healthy individuals. Efficacy of GGC against oxidative stress induced by Pseudomonas aeruginosa, which is a common and chronic pathogen infecting lungs of CF patients, remains unassessed. Primary mucocilliary differentiated airway (bronchial and/or nasal) epithelial cells were created from four individuals with CF. Airway oxidative stress and inflammation was induced by P. aeruginosa lipopolysaccharide (LPS). Parameters including global proteomics alterations, cell redox state (glutathione, oxidative stress), pro-inflammatory mediators (IL-8, IDO-1), and cellular health (membrane integrity, stress granule formation, cell metabolic viability) were assayed under six experimental conditions: (1) Mock, (2) LPS-challenged (3) therapeutic, (4) prophylactic (5) therapeutic and prophylactic and (6) GGC alone. Proteomic analysis identified perturbation of several pathways related to cellular respiration and stress responses upon LPS challenge. Most of these were resolved when cells were treated with GGC. While GGC did not resolve LPS-induced IL-8 and IDO-1 activity, it effectively attenuated LPS-induced oxidative stress and stress granule formation, while significantly increasing total intracellular glutathione levels, metabolic viability and improving epithelial cell barrier integrity. Both therapeutic and prophylactic treatments were successful. Together, these findings indicate that GGC has therapeutic potential for treatment and prevention of oxidative stress-related damage to airways in cystic fibrosis.
Collapse
|
47
|
McCarron A, Parsons D, Donnelley M. Animal and Cell Culture Models for Cystic Fibrosis: Which Model Is Right for Your Application? THE AMERICAN JOURNAL OF PATHOLOGY 2020; 191:228-242. [PMID: 33232694 DOI: 10.1016/j.ajpath.2020.10.017] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 10/01/2020] [Accepted: 10/23/2020] [Indexed: 01/18/2023]
Abstract
Over the past 30 years, a range of cystic fibrosis (CF) animal models have been generated for research purposes. Different species, including mice, rats, ferrets, rabbits, pigs, sheep, zebrafish, and fruit flies, have all been used to model CF disease. While access to such a variety of animal models is a luxury for any research field, it also complicates the decision-making process when it comes to selecting the right model for an investigation. The purpose of this review is to provide a guide for selecting the most appropriate CF animal model for any given application. In this review, the characteristics and phenotypes of each animal model are described, along with a discussion of the key considerations that must be taken into account when choosing a suitable animal model. Available in vitro systems of CF are also described and can offer a useful alternative to using animal models. Finally, the future of CF animal model generation and its use in research are speculated upon.
Collapse
Affiliation(s)
- Alexandra McCarron
- Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia; Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia; Department of Respiratory and Sleep Medicine, Women's and Children's Hospital, North Adelaide, South Australia, Australia.
| | - David Parsons
- Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia; Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia; Department of Respiratory and Sleep Medicine, Women's and Children's Hospital, North Adelaide, South Australia, Australia
| | - Martin Donnelley
- Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia; Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia; Department of Respiratory and Sleep Medicine, Women's and Children's Hospital, North Adelaide, South Australia, Australia
| |
Collapse
|
48
|
Montgomery ST, Stick SM, Kicic A. An adapted novel flow cytometry methodology to delineate types of cell death in airway epithelial cells. J Biol Methods 2020; 7:e139. [PMID: 33204742 PMCID: PMC7666329 DOI: 10.14440/jbm.2020.336] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 09/30/2020] [Accepted: 10/30/2020] [Indexed: 01/01/2023] Open
Abstract
Current methodologies to measure apoptotic and necrotic cell death using flow cytometry do not adequately differentiate between the two. Here, we describe a flow cytometry methodology adapted to airway epithelial cells (AEC) to sufficiently differentiate apoptotic and necrotic AEC. Specifically, cell lines and primary AEC (n = 12) were permeabilized or infected with rhinovirus 1b (RV1b) over 48 h. Cell death was then measured via annexin V/propidium iodide (A5/PI) or annexin V/TO-PRO-3 (A5/TP3) staining using a novel flow cytometry and gating methodology adapted to AEC. We show that A5/PI staining could not sufficiently differentiate between types of cell death following RV1b infection of primary AEC. However, A5/TP3 staining was able to distinguish six cell death populations (viable, necrotic, debris, A5+ apoptotic, A5– apoptotic, apoptotic bodies) after permeabilization or infection with RV1b, with phenotypic differences were observed in apoptotic populations. Collectively, using a staining and gating strategy never adapted to AEC, A5/TP3 could accurately differentiate and quantify viable, necrotic, and apoptotic AEC following RV1b infection.
Collapse
Affiliation(s)
- Samuel T Montgomery
- Faculty of Medicine and Health Science, University of Western Australia, Western Australia 6009, Australia
| | - Stephen M Stick
- Faculty of Medicine and Health Science, University of Western Australia, Western Australia 6009, Australia.,Telethon Kids Institute, University of Western Australia, Western Australia 6009, Australia.,Department of Respiratory and Sleep Medicine, Perth Children's Hospital, Western Australia 6009, Australia.,Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, University of Western Australia, Western Australia 6009, Australia
| | - Anthony Kicic
- Faculty of Medicine and Health Science, University of Western Australia, Western Australia 6009, Australia.,Telethon Kids Institute, University of Western Australia, Western Australia 6009, Australia.,Department of Respiratory and Sleep Medicine, Perth Children's Hospital, Western Australia 6009, Australia.,School of Public Health, Curtin University, Western Australia 6102, Australia.,Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, University of Western Australia, Western Australia 6009, Australia
| |
Collapse
|
49
|
Liu X, Mondal AM. Conditional cell reprogramming for modeling host-virus interactions and human viral diseases. J Med Virol 2020; 92:2440-2452. [PMID: 32478897 PMCID: PMC7586785 DOI: 10.1002/jmv.26093] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 05/26/2020] [Accepted: 05/28/2020] [Indexed: 01/08/2023]
Abstract
Conventional cancer and transformed cell lines are widely used in cancer biology and other fields within biology. These cells usually have abnormalities from the original tumor itself, but may also develop abnormalities due to genetic manipulation, or genetic and epigenetic changes during long-term passages. Primary cultures may maintain lineage functions as the original tissue types, yet they have a very limited life span or population doubling time because of the nature of cellular senescence. Primary cultures usually have very low yields, and the high variability from any original tissue specimens, largely limiting their applications in research. Animal models are often used for studies of virus infections, disease modeling, development of antiviral drugs, and vaccines. Human viruses often need a series of passages in vivo to adapt to the host environment because of variable receptors on the cell surface and may have intracellular restrictions from the cell types or host species. Here, we describe a long-term cell culture system, conditionally reprogrammed cells (CRCs), and its applications in modeling human viral diseases and drug discovery. Using feeder layer coculture in presence of Y-27632 (conditional reprogramming, CR), CRCs can be obtained and rapidly propagated from surgical specimens, core or needle biopsies, and other minimally invasive or noninvasive specimens, for example, nasal cavity brushing. CRCs preserve their lineage functions and provide biologically relevant and physiological conditions, which are suitable for studies of viral entry and replication, innate immune responses of host cells, and discovery of antiviral drugs. In this review, we summarize the applications of CR technology in modeling host-virus interactions and human viral diseases including severe acute respiratory syndrome coronavirus-2 and coronavirus disease-2019, and antiviral discovery.
Collapse
Affiliation(s)
- Xuefeng Liu
- Department of Pathology, Center for Cell ReprogrammingGeorgetown University Medical CenterWashingtonDC
- Department of Oncology, Lombardi Comprehensive Cancer CenterGeorgetown University Medical CenterWashingtonDC
| | - Abdul M. Mondal
- Department of Pathology, Center for Cell ReprogrammingGeorgetown University Medical CenterWashingtonDC
- Department of Oncology, Lombardi Comprehensive Cancer CenterGeorgetown University Medical CenterWashingtonDC
| |
Collapse
|
50
|
Lee RE, Miller SM, Mascenik TM, Lewis CA, Dang H, Boggs ZH, Tarran R, Randell SH. Assessing Human Airway Epithelial Progenitor Cells for Cystic Fibrosis Cell Therapy. Am J Respir Cell Mol Biol 2020; 63:374-385. [PMID: 32437238 DOI: 10.1165/rcmb.2019-0384oc] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Cystic fibrosis (CF) is caused by loss-of-function mutations in the CFTR (CF transmembrane regulator) gene. Pharmacologic therapies directed at CFTR have been developed but are not effective for mutations that result in little or no mRNA or protein expression. Cell therapy is a potential mutation-agnostic approach to treatment. One strategy is to harvest human bronchial epithelial cells (HBECs) for gene addition or genetic correction, followed by expansion and engraftment. This approach will require cells to grow extensively while retaining their ability to reconstitute CFTR activity. We hypothesized that conditionally reprogrammed cell (CRC) technology, namely growth in the presence of irradiated feeder cells and a Rho kinase inhibitor, would enable expansion while maintaining cell capacity to express functional CFTR. Our goal was to compare expression of the basal cell marker NGFR (nerve growth factor receptor) and three-dimensional bronchosphere colony-forming efficiency (CFE) in early- and later-passage HBECs grown using nonproprietary bronchial epithelial growth medium or the CRC method. Cell number and CFTR activity were determined in a competitive repopulation assay employing chimeric air-liquid interface cultures. HBECs expanded using the CRC method expressed the highest NGFR levels, had the greatest 3D colony-forming efficiency at later passage, generated greater cell numbers in chimeric cultures, and most effectively reconstituted CFTR activity. In our study, the HBEC air-liquid interface model, an informative testing platform proven vital for the development of other CF therapies, illustrated that cells grown by CRC technology or equivalent methods may be useful for cell therapy of CF.
Collapse
Affiliation(s)
- Rhianna E Lee
- Marsico Lung Institute/Cystic Fibrosis Center and.,Department of Cell Biology and Physiology, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | | | | | | | - Hong Dang
- Marsico Lung Institute/Cystic Fibrosis Center and
| | | | - Robert Tarran
- Marsico Lung Institute/Cystic Fibrosis Center and.,Department of Cell Biology and Physiology, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Scott H Randell
- Marsico Lung Institute/Cystic Fibrosis Center and.,Department of Cell Biology and Physiology, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|