Summary

Translational 3D-Cell Culture Model to Assess Hyperoxia Effects on Human Neonatal Airway Epithelial Cells

Published: July 12, 2024
doi:

Summary

We describe a protocol to establish an air-liquid interface (ALI) culture model utilizing neonatal tracheal airway epithelial cells (nTAEC) and perform physiologically relevant hyperoxia exposure to study the effect of atmospheric-induced oxidative stress on cells derived from the developing neonatal airway surface epithelium.

Abstract

The preterm neonatal airway epithelium is constantly exposed to environmental stressors. One of these stressors in neonates with lung disease includes oxygen (O2) tension higher than the ambient atmosphere – termed hyperoxia (>21% O2). The effect of hyperoxia on the airway depends on various factors, including the developmental stage of the airway, the degree of hyperoxia, and the duration of exposure, with variable exposures potentially leading to unique phenotypes. While there has been extensive research on the effect of hyperoxia on neonatal lung alveolarization and airway hyperreactivity, little is known about the short and long-term underlying effect of hyperoxia on human neonatal airway epithelial cells. A major reason for this is the scarcity of an effective in vitro model to study human neonatal airway epithelial development and function. Here, we describe a method for isolating and expanding human neonatal tracheal airway epithelial cells (nTAECs) utilizing human neonatal tracheal aspirates and culturing these cells in air-liquid interface (ALI) culture. We demonstrate that nTAECs form a mature polarized cell-monolayer in ALI culture and undergo mucociliary differentiation. We also present a method for moderate hyperoxia exposure of the cell monolayer in ALI culture using a specialized incubator. Additionally, we describe an assay to measure cellular oxidative stress following hyperoxia exposure in ALI culture using fluorescent quantification, which confirms that moderate hyperoxia exposure induces cellular oxidative stress but does not cause significant cell membrane damage or apoptosis. This model can potentially be used to simulate clinically relevant hyperoxia exposure encountered by neonatal airways in the Neonatal Intensive Care Unit (NICU) and used to study the short and long-lasting effects of O2 on neonatal airway epithelial programming. Studies using this model could be utilized to explore ways to mitigate early-life oxidative injury to developing airways, which is implicated in the development of long-term airway diseases in former premature infants.

Introduction

Therapeutic oxygen (O2) is one of the most used therapies in the neonatal intensive care unit (NICU)1. Consequently, hyperoxia exposure (>21% O2) is a common atmospheric stressor encountered by neonates with and without significant lung disease. Lung responses to hyperoxia can vary depending on the intensity and/or duration of exposure and the anatomical location, cell type, and stage of lung development2,3,4,5,6. The bulk of the research in neonatal hyperoxia lung injury has been focused on the effect of hyperoxia exposure in the context of postnatal alveolarization to model bronchopulmonary dysplasia (BPD) – the most common chronic lung disease affecting preterm infants6,7,8,9,10,11. BPD severity is classified by the amount of respiratory support, and O2 needed at 36 weeks post-menstrual age9. Most babies with BPD improve clinically over time as the lungs continue to grow, with the majority weaning off respiratory support before their first birthday12,13. Regardless of the severity of BPD after birth, significant morbidities that affect former preterm babies include a 5-fold increased risk of preschool wheezing, asthma, recurrent respiratory infections throughout childhood, and early onset chronic obstructive pulmonary disease12,14,15,16,17,18,19. The effect of hyperoxia on long-term airway disease and pulmonary infections in preterm infants has been investigated using in vitro and in vivo animal models20,21,22,23,24. However, most of these models focused on the role of mesenchymal tissue, alveolar epithelium, and airway smooth muscle25,26,27,28.

The airway surface epithelium lines the entire path of the respiratory system, extending from the trachea down to the terminal bronchiole, ending just before the level of the alveoli29. Airway basal cells are the primary stem cells in the airway epithelium, with the capacity to differentiate into the entire repertoire of mature airway epithelial lineages, which include ciliated and secretory cells (club cells: non-mucus producing, and goblet cells: mucus-producing)29,30,31,32. Cell culture studies in the context of neonatal hyperoxic lung injury have mostly used adult human or mouse cancer cell lines33,34. Additionally, most in vitro experiments have used submerged culture systems, which do not permit differentiation of the cells into a mucociliary airway epithelium resembling the in vivo airway epithelium in humans35. Consequently, there is a gap in knowledge regarding the effects of hyperoxia-induced lung injury in developing airway epithelial cells of human neonates. One reason is the scarcity of translational models to study the effects of atmospheric exposure on human neonatal airway epithelium. Hyperoxic lung injury early in life can lead to long-term airway disease and increased risk of infection, resulting in life-altering consequences in former preterm infants14,36,37. In non-surviving infants with severe BPD, airway surface epithelium has distinct abnormalities, including goblet cell hyperplasia and disordered ciliary development, denoting abnormal mucociliary clearance and increased epithelial height compromising airway caliber38. In the last decade, there has been increased interest in culturing primary airway epithelial cells at the air-liquid interface (ALI) to study postnatal airway epithelial development39,40,41,42. However, ALI models of neonatal airway epithelial cells have not been used in the context of atmospheric redox perturbation models such as hyperoxia exposure.

Using a previously published method39, we have utilized neonatal tracheal aspirate samples obtained from intubated neonates in the NICU and successfully isolated and expanded primary neonatal tracheal airway epithelial cells (nTAECs). We have utilized inhibitors of Rho, Smad, Glycogen synthase kinase (GSK3), and mammalian target of rapamycin (mTOR) signaling to increase the expansion capacity and delay senescence in these cells, as described previously39,42, which allows for efficient and later passaging of nTAECs. The protocol describes methods for establishing 3D ALI cultures using nTAECs and performing hyperoxia exposure on the nTAEC monolayers. Rho and Smad inhibition is used for the first 7 days of ALI culture (ALI days 0 to 7), after which these inhibitors are removed from the differentiation media for the rest of the ALI culture duration. The apical surface of the ALI-cultured airway epithelial cell monolayer stays exposed to the environment43, which enables atmospheric perturbation studies and closely resembles the pathobiology of a developing neonatal airway exposed to hyperoxia in vivo. The concentration of O2 used in previous cell culture studies (regardless of immortalized or primary cells) of neonatal hyperoxic lung injury varies significantly (ranging from 40% to 95%), as does the duration of exposure (ranging from 15 min to 10 days)36,44,45,46,47. For this study, the ALI cell monolayer was exposed to 60% O2 for 7 days from ALI day 7 to 14 (after removal of Rho/Smad inhibitors from the differentiation media). Hyperoxia exposure was performed during the early-mid phase of mucociliary differentiation (ALI day 7 to 14) as opposed to the fully differentiated mature epithelium and thus simulates the in vivo developing airway epithelium in preterm infants. This exposure strategy minimizes the risk of acute O2 toxicity (which is expected with higher concentrations of O2) while still exerting oxidative stress within a physiologically relevant range and resembles the critical window of transition from the relatively hypoxic intrauterine environment to a hyperoxic external environment in preterm human neonates.

Protocol

Neonatal tracheal aspirate samples were collected only after informed consent from parents, and the protocol used for collection, transport, and storage has been approved by the Institutional Review Board (IRB) of the University of Oklahoma Health Sciences Center (IRB 14377). 1. Preparation for isolation, passaging, and ALI culture of nTAEC Media preparation Bronchial epithelial airway medium (BLEAM) with inhibitors (BLEAM-I): Prepare 500 mL (1 bottle, …

Representative Results

To isolate nTAECs, we collected tracheal aspirates from intubated neonates in the NICU and transported the aspirates on ice to the lab for further processing (Figure 1A). After seeding the tracheal aspirate samples in airway epithelial growth medium (BLEAM-I containing Rho/Smad, GSK3, and mTOR inhibitors), cuboidal cells appeared within 7-10 days. By 14 days, the cells were 50%-60% confluent, and around 21 days post-plating, the cells were densely packed and…

Discussion

The protocol described here details a method for the collection and processing of neonatal tracheal aspirate samples from intubated neonates in the NICU with subsequent isolation and expansion of live nTAECs from these samples using previously established methods39. Furthermore, we have described a method for culturing nTAECs on ALI and characterizing their differentiation into a polarized mucociliary airway epithelium as a function of time via measurement of TEER, FITC-dextran assay, immunofluore…

Declarações

The authors have nothing to disclose.

Acknowledgements

This work is supported by funding from Presbyterian Health Foundation (PHF) and Oklahoma Shared Clinical and Translational Resources (U54GM104938 with an Institutional Development Award (IDeA) from NIGMS) to AG. We would like to thank Dr. Paul LeRou and Dr. Xingbin Ai at Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, for providing neonatal donor cells used in some of the experiments. Figures were created with Biorender. Statistical analysis was performed with GraphPad Prism.

Materials

10% Buffered Formalin Fisher Scientific 23-426796
1X PBS (Phosphate Buffered Saline) Solution, pH 7.4 Gibco 10010049
A 83-01 Tocris 29-391-0
ALI Transwell Inserts, 6.5mm Corning 3470
Anti-Acetylated Tubulin antibody, Mouse monoclonal Sigma T7451
Anti-alpha Tubulin antibody Abcam ab7291
Anti-Cytokeratin 5 antibody Abcam ab53121
BronchiaLife Epithelial Airway Medium (BLEAM) LifeLine Cell Technology LL-0023
CHIR 99021 Tocris 44-231-0
Cleaved caspase-3 antibody Cell signaling 9664T
SCGB1A1 or Club Cell Protein (CC16) Human, Rabbit Polyclonal Antibody BioVendor R&D RD181022220-01
CM-H2DCFDA (General Oxidative Stress Indicator) Thermo Scientific C6827
Corning Cell Culture Treated T25 Flasks Corning 430639
Corning U-Shaped Cell Culture T75 Flasks Corning 430641U
CyQUANT LDH Cytotoxicity Assay Thermo Scientific C20300
DAPI Solution (1 mg/mL) Fisher Scientific EN62248
Dimethyl sulfoxide [DMSO] Hybri-Max Sigma D2650
Distilled water Gibco 15230162
EVOM Manual for TEER Measurement World Precision Instrument EVM-MT-03-01
FBS (Fetal Bovine Serum) Gibco 10082147
Fluorescein Isothiocyanate Dextran (average mol wt 10,000) Fisher Scientific F0918100MG
Fluorescein isothiocyanate–dextran (average mol wt 20,00) Sigma FD20-100MG
Goat Anti-Mouse IgG(H+L), Human ads-HRP Southern Biotech 1031-05
Goat anti-Mouse IgG2b Cross-Adsorbed Secondary Antibody, Alexa Fluor 488 Invitrogen A-21141
Goat anti-Rabbit IgG (H+L) Highly Cross-Adsorbed Secondary Antibody, Alexa Fluor 546 Invitrogen A-11035
Goat Anti-Rabbit IgG(H+L), Mouse/Human ads-HRP Southern Biotech 4050-05
HBTEC Air-Liquid Interface (ALI) Differentiation Medium LifeLine Cell Technology LM-0050
HEPES Lonza CC-5024
Heracell VIOS 160i Tri-Gas CO2 Incubator, 165 L Thermo Scientific 51030411
High-Capacity cDNA Reverse Transcription Kit Thermo Scientific 4368814
HLL supplement LifeLine Cell Technology LS-1001
ImageJ NIH N/A imagej.nih.gov/ij/
Invivogen Normocin – Antimicrobial Reagent Fisher Scientific NC9273499
L-Glutamine LifeLine Cell Technology LS-1013
Normal Goat Serum Gibco PCN5000
Normocin Invivogen ant-nr-05
p63 antibody Santa Cruz Biotechnology sc-25268
ProLong Gold Antifade Mountant Invitrogen P36930
PureLink RNA Mini Kit Thermo Scientific 12183025
RAPAMYCIN Thermo Scientific AAJ62473MC
TaqMan Fast Advanced Master Mix Thermo Scientific 4444964
Taqman Gene Exression Assays: 18S rRNA Thermo Scientific Hs99999901_s1
Taqman Gene Exression Assays: CAT Thermo Scientific Hs00156308_m1
Taqman Gene Exression Assays: FOXJ1 Thermo Scientific Hs00230964_m1
Taqman Gene Exression Assays: GAPDH Thermo Scientific Hs02786624_g1
Taqman Gene Exression Assays: GPX1 Thermo Scientific Hs00829989_gH
Taqman Gene Exression Assays: GPX2 Thermo Scientific Hs01591589_m1
Taqman Gene Exression Assays: GPX3 Thermo Scientific Hs01078668_m1
Taqman Gene Exression Assays: KRT5 Thermo Scientific Hs00361185_m1
Taqman Gene Exression Assays: MUC5AC Thermo Scientific Hs01365616_m1
Taqman Gene Exression Assays: SCGB1A1 Thermo Scientific Hs00171092_m1
Taqman Gene Exression Assays: SOD1 Thermo Scientific Hs00533490_m1
Taqman Gene Exression Assays: SOD2 Thermo Scientific Hs00167309_m1
Thermo Scientific Nalgene Rapid-Flow Sterile Disposable Filter Units with PES Membrane (0.22 μm pores, 500 ml) Thermo Scientific 5660020
TM-1 Combined Supplement LifeLine Cell Technology LS-1055
Total caspase-3 antibody Cell signaling 14220S
Triton X-100 Sigma 9036-19-5
Trypsin-EDTA (0.05%), Phenol red Gibco 25300062
Y-27632 2 HCl Tocris 12-541-0

Referências

  1. Torres-Cuevas, I., et al. Oxygen and oxidative stress in the perinatal period. Redox Biol. 12, 674-681 (2017).
  2. Hanidziar, D., Robson, S. C. Hyperoxia and modulation of pulmonary vascular and immune responses in COVID-19. Am J Physiol-Lung Cell Mol Physiol. 320 (1), L12-L16 (2021).
  3. Amarelle, L., Quintela, L., Hurtado, J., Malacrida, L. Hyperoxia and lungs: What we have learned from animal models. Front Med. 8, 606678 (2021).
  4. Mach, W. J., Thimmesch, A. R., Pierce, J. T., Pierce, J. D. Consequences of hyperoxia and the toxicity of oxygen in the lung. Nursing Res Pract. 2011, 260482 (2011).
  5. Kallet, R. H., Matthay, M. A. Hyperoxic acute lung injury. Respir Care. 58 (1), 123-141 (2013).
  6. Penkala, I. J., et al. Age-dependent alveolar epithelial plasticity orchestrates lung homeostasis and regeneration. Cell Stem Cell. 28 (10), 1775-1789.e1775 (2021).
  7. Thébaud, B., et al. Bronchopulmonary dysplasia. Nat Rev Dis Primers. 5 (1), 78 (2019).
  8. Ibrahim, J., Bhandari, V. The definition of bronchopulmonary dysplasia: an evolving dilemma. Pediatr Res. 84 (5), 586-588 (2018).
  9. Higgins, R. D., et al. Bronchopulmonary Dysplasia: Executive summary of a workshop. J Pediatr. 197, 300-308 (2018).
  10. Collins, J. J. P., Tibboel, D., de Kleer, I. M., Reiss, I. K. M., Rottier, R. J. The future of bronchopulmonary Dysplasia: Emerging pathophysiological concepts and potential new avenues of treatment. Front Med (Lausanne). 4, 61 (2017).
  11. Northway, W. H., Rosan, R. C., Porter, D. Y. Pulmonary disease following respirator therapy of hyaline-membrane disease. Bronchopulmonary dysplasia. N Engl J Med. 276 (7), 357-368 (1967).
  12. Collaco, J. M., McGrath-Morrow, S. A. Bronchopulmonary dysplasia as a determinant of respiratory outcomes in adult life. Pediatr Pulmonol. 56 (11), 3464-3471 (2021).
  13. Tepper, R. S., Morgan, W. J., Cota, K., Taussig, L. M. Expiratory flow limitation in infants with bronchopulmonary dysplasia. J Pediatr. 109 (6), 1040-1046 (1986).
  14. Davidson, L. M., Berkelhamer, S. K. Bronchopulmonary Dysplasia: Chronic lung disease of infancy and long-term pulmonary outcomes. J Clin Med. 6 (1), 4 (2017).
  15. Filbrun, A. G., Popova, A. P., Linn, M. J., McIntosh, N. A., Hershenson, M. B. Longitudinal measures of lung function in infants with bronchopulmonary dysplasia. Pediatr Pulmonol. 46 (4), 369-375 (2011).
  16. Stoll, B. J., et al. Neonatal outcomes of extremely preterm infants from the NICHD Neonatal Research Network. Pediatrics. 126 (3), 443-456 (2010).
  17. Colin, A. A., McEvoy, C., Castile, R. G. Respiratory morbidity and lung function in preterm infants of 32 to 36 weeks’ gestational age. Pediatrics. 126 (1), 115-128 (2010).
  18. Doyle, L. W., Ranganathan, S., Cheong, J. Bronchopulmonary dysplasia and expiratory airflow at 8 years in children born extremely preterm in the post-surfactant era. Thorax. 78 (5), 484-488 (2022).
  19. Doyle, L. W., et al. Ventilation in extremely preterm infants and respiratory function at 8 Years. N Engl J Med. 377 (4), 329-337 (2017).
  20. Brozmanova, M., Hanacek, J., Tatar, M., Strapkova, A., Szepe, P. Effects of hyperoxia and allergic airway inflammation on cough reflex intensity in guinea pigs. Physiol Res. 51 (5), 529-536 (2002).
  21. Burghardt, J. S., Boros, V., Biggs, D. F., Olson, D. M. Lipid mediators in oxygen-induced airway remodeling and hyperresponsiveness in newborn rats. Am J Respir Crit Care Med. 154 (4 Pt 1), 837-842 (1996).
  22. Mazurek, H., Haouzi, P., Belaguid, A., Marchal, F. Persistent increased lung response to methacholine after normobaric hyperoxia in rabbits. Respir Physiol. 99 (2), 199-204 (1995).
  23. Onugha, H., et al. Airway hyperreactivity is delayed after mild neonatal hyperoxic exposure. Neonatology. 108 (1), 65-72 (2015).
  24. Regal, J. F., Lawrence, B. P., Johnson, A. C., Lojovich, S. J., O’Reilly, M. A. Neonatal oxygen exposure alters airway hyper-responsiveness but not the response to allergen challenge in adult mice. Pediatr Allergy Immunol. 25 (2), 180-186 (2014).
  25. Mayer, C. A., et al. CPAP-induced airway hyper-reactivity in mice is modulated by hyaluronan synthase-3. Pediatr Res. 92 (3), 685-693 (2022).
  26. Ganguly, A., Martin, R. J. Vulnerability of the developing airway. Respir Physiol Neurobiol. 270, 103263 (2019).
  27. Yee, M., Buczynski, B. W., O’Reilly, M. A. Neonatal hyperoxia stimulates the expansion of alveolar epithelial type II cells. Am J Respir Cell Mol Biol. 50 (4), 757-766 (2014).
  28. Balasubramaniam, V., Mervis, C. F., Maxey, A. M., Markham, N. E., Abman, S. H. Hyperoxia reduces bone marrow, circulating, and lung endothelial progenitor cells in the developing lung: implications for the pathogenesis of bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol. 292 (5), L1073-L1084 (2007).
  29. Crystal, R. G., Randell, S. H., Engelhardt, J. F., Voynow, J., Sunday, M. E. Airway epithelial cells. Proc Am Thoracic Soc. 5 (7), 772-777 (2008).
  30. Whitsett, J. A. Airway epithelial differentiation and mucociliary clearance. Ann Am Thor Soc. 15 (Supplement_3), S143-S148 (2018).
  31. Davis, J. D., Wypych, T. P. Cellular and functional heterogeneity of the airway epithelium. Mucosal Immunol. 14 (5), 978-990 (2021).
  32. Whitsett, J. A., Kalin, T. V., Xu, Y., Kalinichenko, V. V. Building and regenerating the lung cell by cell. Physiol Rev. 99 (1), 513-554 (2019).
  33. Maeda, H., et al. Involvement of miRNA-34a regulated Krüppel-like factor 4 expression in hyperoxia-induced senescence in lung epithelial cells. Respir Res. 23 (1), 340 (2022).
  34. Zhu, Y., Mosko, J. J., Chidekel, A., Wolfson, M. R., Shaffer, T. H. Effects of xenon gas on human airway epithelial cells during hyperoxia and hypothermia. J Neonatal Perinatal Med. 13 (4), 469-476 (2020).
  35. Cao, X., et al. Invited review: human air-liquid-interface organotypic airway tissue models derived from primary tracheobronchial epithelial cells-overview and perspectives. In Vitro Cell Dev Biol – Animal. 57 (2), 104-132 (2021).
  36. Garcia, D., et al. Short exposure to hyperoxia causes cultured lung epithelial cell mitochondrial dysregulation and alveolar simplification in mice. Pediatr Res. 90 (1), 58-65 (2020).
  37. Crist, A. P., Hibbs, A. M. Prematurity-associated wheeze: current knowledge and opportunities for further investigation. Pediatr Res. 94 (1), 74-81 (2022).
  38. Lee, R. M., O’Brodovich, H. Airway epithelial damage in premature infants with respiratory failure. Am Rev Respir Dis. 137 (2), 450-457 (1988).
  39. Amonkar, G. M., et al. Primary culture of tracheal aspirate-derived human airway basal stem cells. STAR Protoc. 3 (2), 101390 (2022).
  40. Shui, J. E., et al. Prematurity alters the progenitor cell program of the upper respiratory tract of neonates. Sci Rep. 11 (1), 10799 (2021).
  41. Hillas, J., et al. Nasal airway epithelial repair after very preterm birth. ERJ Open Res. 7 (2), 00913-02020 (2021).
  42. Lu, J., et al. Rho/SMAD/mTOR triple inhibition enables long-term expansion of human neonatal tracheal aspirate-derived airway basal cell-like cells. Pediatr Res. 89 (3), 502-509 (2020).
  43. Jiang, D., Schaefer, N., Chu, H. W. Air-liquid interface culture of human and mouse airway epithelial cells. Meth Mol Biol. 1809, 91-109 (2018).
  44. You, K., et al. Moderate hyperoxia induces senescence in developing human lung fibroblasts. Am J Physiol Lung Cell Mol Physiol. 317 (5), L525-L536 (2019).
  45. Wang, H., et al. Severity of neonatal hyperoxia determines structural and functional changes in developing mouse airway. Am J Physiol Lung Cell Mol Physiol. 307 (4), L295-L301 (2014).
  46. Ravikumar, P., et al. α-Klotho protects against oxidative damage in pulmonary epithelia. Am J Physi-Lung Cell Mol Physiol. 307 (7), L566-L575 (2014).
  47. Hartman, W. R., et al. Oxygen dose responsiveness of human fetal airway smooth muscle cells. Am J Physiol Lung Cell Mol Physiol. 303 (8), L711-L719 (2012).
  48. Cadena-Herrera, D., et al. Validation of three viable-cell counting methods: Manual, semi-automated, and automated. Biotechnol Rep. 7, 9-16 (2015).
  49. Bodas, M., et al. Cigarette smoke activates NOTCH3 to promote goblet cell differentiation in human airway epithelial cells. Am J Respir Cell Mol Biol. 64 (4), 426-440 (2021).
  50. Srinivasan, B., et al. TEER measurement techniques for in vitro barrier model systems. J Lab Autom. 20 (2), 107-126 (2015).
  51. Jakubowski, W., Bartosz, G. 2,7-dichlorofluorescin oxidation and reactive oxygen species: what does it measure. Cell Biol Int. 24 (10), 757-760 (2000).
  52. Oksvold, M. P., Skarpen, E., Widerberg, J., Huitfeldt, H. S. Fluorescent histochemical techniques for analysis of intracellular signaling. J Histochem Cytochem. 50 (3), 289-303 (2002).
  53. Hewitt, R. J., et al. Lung extracellular matrix modulates KRT5(+) basal cell activity in pulmonary fibrosis. Nat Commun. 14 (1), 6039 (2023).
  54. Hawkins, F. J., et al. Derivation of airway basal stem cells from human pluripotent stem cells. Cell Stem Cell. 28 (1), 79-95.e78 (2021).
  55. Zou, X. L., et al. Down-expression of Foxj1 on airway epithelium with impaired cilia architecture in non-cystic fibrosis bronchiectasis implies disease severity. Clin Respir J. 17 (5), 405-413 (2023).
  56. Li, X., et al. Low CC16 mRNA expression levels in bronchial epithelial cells are associated with asthma severity. Am J Respir Crit Care Med. 207 (4), 438-451 (2023).
  57. Li, T., Wang, Y., Huang, S., Tang, H. The regulation mechanism of MUC5AC secretion in airway of obese asthma. Cell Mol Biol (Noisy-le-grand). 68 (7), 153-159 (2022).
  58. Nekooki-Machida, Y., Hagiwara, H. Role of tubulin acetylation in cellular functions and diseases. Med Mol Morphol. 53 (4), 191-197 (2020).
  59. Creagh, E. M., Conroy, H., Martin, S. J. Caspase-activation pathways in apoptosis and immunity. Immunol Rev. 193, 10-21 (2003).
  60. Abo, K. M., et al. Air-liquid interface culture promotes maturation and allows environmental exposure of pluripotent stem cell-derived alveolar epithelium. JCI Insight. 7 (6), e155589 (2022).
  61. Guénette, J., Breznan, D., Thomson, E. M. Establishing an air-liquid interface exposure system for exposure of lung cells to gases. Inhal Toxicol. 34 (3-4), 80-89 (2022).
  62. Zhao, C., et al. Age-related STAT3 signaling regulates severity of respiratory syncytial viral infection in human bronchial epithelial cells. bioRxiv. , (2023).
  63. Lochbaum, R., et al. Retinoic acid signalling adjusts tight junction permeability in response to air-liquid interface conditions. Cellular Signalling. 65, 109421 (2020).
  64. Jain, R., et al. Temporal relationship between primary and motile ciliogenesis in airway epithelial cells. Am J Respir Cell Mol Biol. 43 (6), 731-739 (2010).
  65. You, Y., et al. Role of f-box factor foxj1 in differentiation of ciliated airway epithelial cells. Am J Physiol Lung Cell Mol Physiol. 286 (4), L650-L657 (2004).
  66. Pan, J., You, Y., Huang, T., Brody, S. L. RhoA-mediated apical actin enrichment is required for ciliogenesis and promoted by Foxj1. J Cell Sci. 120 (Pt 11), 1868-1876 (2007).
  67. Teape, D., et al. Hyperoxia impairs intraflagellar transport and causes dysregulated metabolism with resultant decreased cilia length. Am J Physiol Lung Cell Mol Physiol. 324 (3), L325-L334 (2023).
  68. Hall, C. B. Respiratory syncytial virus and parainfluenza virus. N Engl J Med. 344 (25), 1917-1928 (2001).
  69. Guillien, A., et al. Determinants of immunoglobulin G responses to respiratory syncytial virus and rhinovirus in children and adults. Front Immunol. 15, 1355214 (2024).
  70. Ito, K., Daly, L., Coates, M. An impact of age on respiratory syncytial virus infection in air-liquid-interface culture bronchial epithelium. Front Med (Lausanne). 10, 1144050 (2023).
  71. Zimmermann, P., Curtis, N. Why does the severity of COVID-19 differ with age?: Understanding the mechanisms underlying the age gradient in outcome following SARS-CoV-2 infection. Pediatr Infect Dis J. 41 (2), e36-e45 (2022).
  72. Leung, C., Wadsworth, S. J., Yang, S. J., Dorscheid, D. R. Structural and functional variations in human bronchial epithelial cells cultured in air-liquid interface using different growth media. Am J Physiol Lung Cell Mol Physiol. 318 (5), L1063-L1073 (2020).
  73. Dieperink, H. I., Blackwell, T. S., Prince, L. S. Hyperoxia and apoptosis in developing mouse lung mesenchyme. Pediatric research. 59 (2), 185-190 (2006).
  74. Looi, K., et al. Preterm birth: Born too soon for the developing airway epithelium. Paediatr Respir Revi. 31, 82-88 (2019).
  75. Hilgendorff, A., Reiss, I., Ehrhardt, H., Eickelberg, O., Alvira, C. M. Chronic lung disease in the preterm infant. Lessons learned from animal models. Am J Respir Cell Mol Biol. 50 (2), 233-245 (2014).
This article has been published
Video Coming Soon
Keep me updated:

.

Citar este artigo
Carter, C. M., Mathias, M. M., Bailey-Downs, L., Tipple, T. E., Vitiello, P. F., Walters, M. S., Ganguly, A. Translational 3D-Cell Culture Model to Assess Hyperoxia Effects on Human Neonatal Airway Epithelial Cells. J. Vis. Exp. (209), e65913, doi:10.3791/65913 (2024).

View Video