Characterization of a lung epithelium specific E-cadherin knock-out model: Implications for obstructive lung pathology

S Post, I H Heijink, L Hesse, H K Koo, F Shaheen, M Fouadi, V N S Kuchibhotla, B N Lambrecht, A J M Van Oosterhout, T L Hackett, M C Nawijn, S Post, I H Heijink, L Hesse, H K Koo, F Shaheen, M Fouadi, V N S Kuchibhotla, B N Lambrecht, A J M Van Oosterhout, T L Hackett, M C Nawijn

Abstract

The airway epithelium regulates responses to aeroallergens, acting as a physical and immunological barrier. In asthma, epithelial barrier function and the expression of adherens junction protein E-cadherin is compromised, but it is unknown whether this is cause or consequence of the disease. We hypothesized that airway epithelial loss of E-cadherin is a critical step in the development of manifestations of asthma. We generated a transgenic mouse model with conditional loss of E-cadherin in lung epithelial cells at birth and onwards. We observed normal lung development at the time of birth in mice lacking E-cadherin in the lung epithelium. However, E-cadherin deficiency led to progressive epithelial damage in mice growing into adulthood, as evidenced by airway epithelial denudation, decreased zonula occludens (ZO)-1 expression, loss of ciliated cells, and enlarged alveolar spaces. In addition, spontaneous goblet cell metaplasia with mucus production was observed. These epithelial changes were accompanied by elevated levels of the epithelial-derived chemokine CCL17, infiltration of eosinophils and dendritic cells, and mucus production. In conclusion, loss of E-cadherin induces features in the lung reminiscent of those observed in asthma, indicating that the disruption of E-cadherin-mediated cell-cell contacts may play a key role in the development of asthma manifestations.

Conflict of interest statement

This study was funded by unrestricted research grants from the Dutch Lung Foundation (Longfonds; 3.2.07.019), Stichting Astma Bestrijding (2014/008), European Respiratory Society (LTRF-118-2012) and the Canadian Institutes for Health Research (260046). We have no non-financial competing interests.

Figures

Figure 1
Figure 1
E-cadherin expression in the lungs of E-cadherin knockout (Cdh1fl/fl Cre+) and wild type (Cdh1fl/fl Cre−) mice (n = 5–7 per group). (A) E-cadherin staining of airway epithelium at day (D)0 (I,IV), week (W) 4 (II,V) and W10 (III,VI) of Cdh1fl/fl Cre−(I-III)/Cre+ (IV-VI) mice. Red arrows indicate epithelial denudation areas. Scale bars: 10 μm. Percentage of E-cadherin positive cell numbers as analyzed by Image-Pro Plus at (B) D0, (C) W4 and (D) W10. mRNA expression of E-cadherin (cdh1) in lung homogenates at (E) W4 and (F) W10. E-cadherin levels were related to the housekeeping genes hprt1 and pgk1 and expressed as 2−ΔCt. Medians are indicated. *p < 0.05, **p < 0.01 and ***p < 0.001 as assessed by the Mann Whitney U test.
Figure 2
Figure 2
Characterization of airway epithelium in E-cadherin knockout (Cdh1fl/fl Cre+) and wild type (Cdh1fl/fl Cre−) mice. Analysis of percent total cells in the epithelium of Cdh1fl/fl Cre−/Cre+ mice at (A) day (D)0, (B) W4 and (C) W10, where the total cell count per 500 µm basement membrane (5–25 counts per lung fragment) per mouse is presented as percentage of the group average. Medians are indicated. (D) Electron microscopy images at W4, magnifications are as indicated. White arrows indicate epithelial denudation areas. (E) Hematoxylin staining in lung tissue of Cdh1fl/fl Cre−/Cre+ mice at W8, magnifications are as indicated. *p < 0.05, **p < 0.01 and ***p < 0.001 between the Cdh1fl/fl Cre+ and Cdh1fl/fl Cre− mice (n = 5–7 per group) as assessed by the Mann Whitney U test.
Figure 3
Figure 3
Characterization of alveoli in E-cadherin knockout (Cdh1fl/fl Cre+) and wild type (Cdh1fl/fl Cre−) mice. (A) E-cadherin staining of alveoli at day (D)0 (I,IV), week (W)4 (II,V) and W10 (III,VI) of Cdh1fl/fl Cre− (I-III)/Cre+ (IV-VI) mice. Scale bars: 10 μm. Measurement of mean linear intercept (Lm) in the lung tissue at (B) D0, (C) W4 and (D) W10. (E) Hematoxylin staining in lung tissue of Cdh1fl/fl Cre−/Cre+ mice at D0, W4 and W10. Scale bars, 100 μm. An average Lm was calculated from 3 randomly selected regions within each lung. Lm = Number of lines × Length of test line/Number of intersections. A greater Lm value therefore indicates increased air-space size.Medians are indicated. *p < 0.05, **p < 0.001 and ***p < 0.0001 between the Cdh1fl/fl Cre+ and Cdh1fl/fl Cre− mice (n = 5–7 per group) as assessed by the Mann Whitney U test.
Figure 4
Figure 4
Loss of ciliated cells and increased mucus production in E-cadherin knockout (Cdh1fl/fl Cre+) and wild type (Cdh1fl/fl Cre−) mice. Analysis of percent ciliated cells at (A) day (D)0, (B) week (W)4 and (C) W10, where the ciliated cell count per mouse was calculated per 500 µm basement membrane (5–25 counts per lung fragment) as percentage of the group average. (D) Electron microscopy images at Week W6. Red arrow indicates loss of ciliated cells. Measurement of percentage alcian blue staining in the airways of Cdh1fl/fl Cre−/Cre+ mice at E) W4, (F) W8 and (G) W10, (H) Representative image of alcian blue staining of Cdh1fl/fl Cre−/Cre+ mice at W8. Scale bars, 10 μm. (I) Percentage of alpha-smooth muscle actin (α-SMA) positive cell numbers as analyzed by Image-Pro Plus in the airways of Cdh1fl/fl Cre−/Cre+ mice at W10. Medians are indicated. *p < 0.05 and **p < p0.01 between the Cdh1fl/fl Cre+ and Cdh1fl/fl Cre− mice (n = 5–7 per group) as assessed by the Mann Whitney U test.
Figure 5
Figure 5
Cytokine responses in E-cadherin knockout (Cdh1fl/fl /Cre+) and wild type (Cdh1fl/fl Cre−) mice.Enzyme-linked immunosorbent assay (ELISA) analysis for (A) Chemokine (C-C motif) ligand 17 (CCL17), (B) Thymic stromal lymphopoietin (TSLP), (C) C-C motif chemokine 11 (CCL11) and (D) Granulocyte-macrophage colony-stimulating factor (GM-CSF) levels in whole lung homogenates at week (W)4. Medians are indicated. **p < 0.01 between the Cdh1fl/fl Cre+ and Cdh1fl/fl Cre− mice (n = 5–7 per group) as assessed by the Mann Whitney U test.
Figure 6
Figure 6
Inflammatory responses in E-cadherin knockout (Cdh1fl/fl Cre+) mice) and wild type (Cdh1fl/fl Cre−) mice. Flow cytometry analysis for inflammatory cells in whole lungs of Cdh1fl/fl Cre−/Cre+ mice at (A) week (W)2 and (E) W4. Dendritic cell (DC) populations at (B) W2 and F) W4 and DC sub-populations (CD103+ conventional (c)DCs, CD11b + DCs and monocyte-derived (mo)DCs) at (C) W2 and (G) W4. Alveolar macrophages at (D) 2 W and (H) 4 W. Medians are indicated. *p < 0.05, **p < 0.01 or p value is as indicated between the Cdh1fl/fl Cre+ and Cdh1fl/fl Cre− mice (n = 5–7 per group) as assessed by the Mann Whitney U test.

References

    1. Goto Y, et al. Dislocation of E-cadherin in the airway epithelium during an antigen-induced asthmatic response. Am. J. Respir. Cell Mol. Biol. 2000;23:712–718. doi: 10.1165/ajrcmb.23.6.4031.
    1. Tam A, Wadsworth S, Dorscheid D, Man SF, Sin DD. The airway epithelium: more than just a structural barrier. Ther. Adv. Respir. Dis. 2011;5:255–273. doi: 10.1177/1753465810396539.
    1. Nawijn MC, Hackett TL, Postma DS, Van Oosterhout AJ, Heijink IH. E-cadherin: gatekeeper of airway mucosa and allergic sensitization. Trends Immunol. 2011;32:248–255. doi: 10.1016/j.it.2011.03.004.
    1. Tunggal JA, et al. E-cadherin is essential for in vivo epidermal barrier function by regulating tight junctions. EMBO J. 2005;24:1146–56. doi: 10.1038/sj.emboj.7600605.
    1. Heijink IH, et al. Down-regulation of E-cadherin in human bronchial epithelial cells leads to epidermal growth factor receptor-dependent Th2 cell-promoting activity. J. Immunol. (Baltimore, Md. 1950) 2007;178:7678–7685. doi: 10.4049/jimmunol.178.12.7678.
    1. de Boer WI, et al. Altered expression of epithelial junctional proteins in atopic asthma: possible role in inflammation. Can. J. Physiol. Pharmacol. 2008;86:105–112. doi: 10.1139/Y08-004.
    1. Hackett TL, et al. Characterization of side population cells from human airway epithelium. Stem Cells. 2008;26:2576–2585. doi: 10.1634/stemcells.2008-0171.
    1. Thomas, B. et al. Ciliary dysfunction and ultrastructural abnormalities are features of severe asthma. J. Allergy Clin. Immunol. 126 (2010).
    1. Hackett NR, et al. The human airway epithelial Basal cell transcriptome. PLoS One. 2011;6:e18378. doi: 10.1371/journal.pone.0018378.
    1. Xiao C, et al. Defective epithelial barrier function in asthma. J. Allergy Clin. Immunol. 2011;128:512–549. doi: 10.1016/j.jaci.2011.05.038.
    1. Looi, K. et al. Effects of human rhinovirus on epithelial barrier integrity and function in children with asthma. Clin. Exp. Allergy. 10.1111/cea.13097 (2018).
    1. Post S, et al. House dust mite-induced calcium signaling instigates epithelial barrier dysfunction and CCL20 production. Allergy. 2013;68:1117–1125.
    1. Heijink IH, Postma DS, Noordhoek JA, Broekema M, Kapus A. House dust mite-promoted epithelial-to-mesenchymal transition in human bronchial epithelium. Am. J. Respir. Cell Mol. Biol. 2010;42:69–79. doi: 10.1165/rcmb.2008-0449OC.
    1. Post S, et al. The composition of house dust mite is critical for mucosal barrier dysfunction and allergic sensitisation. Thorax. 2012;67:488–495. doi: 10.1136/thoraxjnl-2011-200606.
    1. Hackett TL. Epithelial-mesenchymal transition in the pathophysiology of airway remodelling in asthma. Curr. Opin. Allergy Clin. Immunol. 2012;12:53–59. doi: 10.1097/ACI.0b013e32834ec6eb.
    1. Johnson JR, Roos A, Berg T, Nord M, Fuxe J. Chronic respiratory aeroallergen exposure in mice induces epithelial-mesenchymal transition in the large airways. PLoS One. 2011;6:e16175. doi: 10.1371/journal.pone.0016175.
    1. Larue L, Ohsugi M, Hirchenhain J, Kemler R. E-cadherin null mutant embryos fail to form a trophectoderm epithelium. Proc. Natl. Acad. Sci. USA. 1994;91:8263–8267. doi: 10.1073/pnas.91.17.8263.
    1. Holgate ST. The sentinel role of the airway epithelium in asthma pathogenesis. Immunol. Rev. 2011;242:205–219. doi: 10.1111/j.1600-065X.2011.01030.x.
    1. Irwin RS, et al. Spread the word about the journal in 2013: from citation manipulation to invalidation of patient-reported outcomes measures to renaming the Clara cell to new journal features. Chest. 2013;143:1–4. doi: 10.1378/chest.12-2762.
    1. Reynolds SD, Malkinson AM. Clara cell: progenitor for the bronchiolar epithelium. Int. J. Biochem. Cell Biol. 2010;42:1–4. doi: 10.1016/j.biocel.2009.09.002.
    1. Rackley CR, Stripp BR. Building and maintaining the epithelium of the lung. J. Clin. Invest. 2012;122:2724–2730. doi: 10.1172/JCI60519.
    1. Perl AK, Tichelaar JW, Whitsett JA. Conditional gene expression in the respiratory epithelium of the mouse. Transgenic Res. 2002;11:21–29. doi: 10.1023/A:1013986627504.
    1. Wert SE, Glasser SW, Korfhagen TR, Whitsett JA. Transcriptional elements from the human SP-C gene direct expression in the primordial respiratory epithelium of transgenic mice. Dev. Biol. 1993;156:426–443. doi: 10.1006/dbio.1993.1090.
    1. Perl AK, Wert SE, Nagy A, Lobe CG, Whitsett JA. Early restriction of peripheral and proximal cell lineages during formation of the lung. Proc. Natl. Acad. Sci. USA. 2002;99:10482–10487. doi: 10.1073/pnas.152238499.
    1. Kasper M, Behrens J, Schuh D, Muller M. Distribution of E-cadherin and Ep-CAM in the human lung during development and after injury. Histochem. Cell Biol. 1995;103:281–286. doi: 10.1007/BF01457412.
    1. Takeichi M. The cadherins: cell-cell adhesion molecules controlling animal morphogenesis. Development. 1988;102:639–655.
    1. Ceteci F, et al. E-cadherin Controls Bronchiolar Progenitor Cells and Onset of Preneoplastic Lesions in Mice. Neoplasia. 2012;14:1164–1177. doi: 10.1593/neo.121088.
    1. Rawlins EL, et al. The role of Scgb1a1 + Clara cells in the long-term maintenance and repair of lung airway, but not alveolar, epithelium. Cell Stem Cell. 2009;4:525–534. doi: 10.1016/j.stem.2009.04.002.
    1. Mucenski ML, et al. Beta-catenin regulates differentiation of respiratory epithelial cells in vivo. Am. J. Physiol. Lung Cell. Mol. Physiol. 2005;289:L971–L979. doi: 10.1152/ajplung.00172.2005.
    1. Heijink, I. H., van Oosterhout, A. & Kapus, A. EGFR signaling contributes to house dust mite-induced epithelial barrier dysfunction. Eur. Respir. J. Off. J. Eur. Soc. Clin. Respir. Physio, 10.1183/09031936.00125809 (2010).
    1. Nadel JA, Burgel PR. The role of epidermal growth factor in mucus production. Curr. Opin. Pharmacol. 2001;1:254–258. doi: 10.1016/S1471-4892(01)00045-5.
    1. Nakano Y, Van Tho N, Yamada H, Osawa M, Nagao T. Radiological approach to asthma and COPD–the role of computed tomography. Allergol. Int. 2009;58:323–31. doi: 10.2332/allergolint.09-RAI-0124.
    1. Mitsunobu F, et al. Complexity of terminal airspace geometry assessed by computed tomography in asthma. Am. J. Respir. Crit. Care Med. 2003;167:411–417. doi: 10.1164/rccm.2112070.
    1. Hoffman, A. M. & Ingenito, E. P. Alveolar epithelial stem and progenitor cells: emerging evidence for their role in lung regeneration. Curr. Med. Chem. (2012).
    1. Galambos C, Demello DE. Regulation of alveologenesis: clinical implications of impaired growth. Pathology. 2008;40:124–140. doi: 10.1080/00313020701818981.
    1. Perl AK, Riethmacher D, Whitsett JA. Conditional depletion of airway progenitor cells induces peribronchiolar fibrosis. Am. J. Respir. Crit. Care Med. 2011;183:511–521. doi: 10.1164/rccm.201005-0744OC.
    1. Ceteci F, et al. Disruption of Tumor Cell Adhesion Promotes Angiogenic Switch and Progression to Micrometastasis in RAF-Driven Murine Lung Cancer. Cancer Cell. 2007;12:145–159. doi: 10.1016/j.ccr.2007.06.014.
    1. Wan H, et al. Foxa2 regulates alveolarization and goblet cell hyperplasia. Development. 2004;131:953–964. doi: 10.1242/dev.00966.
    1. Heijink IH, et al. Der p, IL-4, and TGF-beta cooperatively induce EGFR-dependent TARC expression in airway epithelium. Am. J. Respir. Cell Mol. Biol. 2007;36:351–359. doi: 10.1165/rcmb.2006-0160OC.
    1. Cowell CF, et al. Loss of cell-cell contacts induces NF-kappaB via RhoA-mediated activation of protein kinase D1. J. Cell. Biochem. 2009;106:714–728. doi: 10.1002/jcb.22067.
    1. Lambrecht, B. N. & Hammad, H. The immunology of asthma. Nat. Immunol. 16 (2014).
    1. Chen G, et al. Foxa2 Programs Th2 Cell-Mediated Innate Immunity in the Developing Lung. J. Immunol. 2010;184:6133–6141. doi: 10.4049/jimmunol.1000223.
    1. Zhang Z, et al. FOXA2 attenuates the epithelial to mesenchymal transition by regulating the transcription of E-cadherin and ZEB2 in human breast cancer. Cancer Lett. 2015;361:240–250. doi: 10.1016/j.canlet.2015.03.008.
    1. Plantinga M, et al. Conventional and monocyte-derived CD11b(+) dendritic cells initiate and maintain T helper 2 cell-mediated immunity to house dust mite allergen. Immunity. 2013;38:322–335. doi: 10.1016/j.immuni.2012.10.016.
    1. Barbato A, et al. Epithelial damage and angiogenesis in the airways of children with asthma. Am. J. Respir. Crit. Care Med. 2006;174:975–981. doi: 10.1164/rccm.200602-189OC.
    1. Ierodiakonou D, et al. E-cadherin gene polymorphisms in asthma patients using inhaled corticosteroids. Eur. Respir. J. Off. J. Eur. Soc. Clin. Respir. Physiol. 2011;38:1044–1052.

Source: PubMed

3
Abonner