Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Letter
  • Published:

Muc5b is required for airway defence

Abstract

Respiratory surfaces are exposed to billions of particulates and pathogens daily. A protective mucus barrier traps and eliminates them through mucociliary clearance (MCC)1,2. However, excessive mucus contributes to transient respiratory infections and to the pathogenesis of numerous respiratory diseases1. MUC5AC and MUC5B are evolutionarily conserved genes that encode structurally related mucin glycoproteins, the principal macromolecules in airway mucus1,3. Genetic variants are linked to diverse lung diseases4,5,6, but specific roles for MUC5AC and MUC5B in MCC, and the lasting effects of their inhibition, are unknown. Here we show that mouse Muc5b (but not Muc5ac) is required for MCC, for controlling infections in the airways and middle ear, and for maintaining immune homeostasis in mouse lungs, whereas Muc5ac is dispensable. Muc5b deficiency caused materials to accumulate in upper and lower airways. This defect led to chronic infection by multiple bacterial species, including Staphylococcus aureus, and to inflammation that failed to resolve normally7. Apoptotic macrophages accumulated, phagocytosis was impaired, and interleukin-23 (IL-23) production was reduced in Muc5b−/− mice. By contrast, in mice that transgenically overexpress Muc5b, macrophage functions improved. Existing dogma defines mucous phenotypes in asthma and chronic obstructive pulmonary disease (COPD) as driven by increased MUC5AC, with MUC5B levels either unaffected or increased in expectorated sputum1,8. However, in many patients, MUC5B production at airway surfaces decreases by as much as 90%9,10,11. By distinguishing a specific role for Muc5b in MCC, and by determining its impact on bacterial infections and inflammation in mice, our results provide a refined framework for designing targeted therapies to control mucin secretion and restore MCC.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Purchase on Springer Link

Instant access to full article PDF

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Muc5b is required for survival and particle clearance.
Figure 2: Muc5b deficiency causes severe upper airway obstruction.
Figure 3: Infection is the cause of death in Muc5b−/− mice.
Figure 4: Muc5b maintains functioning lung macrophage populations.

Similar content being viewed by others

References

  1. Fahy, J. V. & Dickey, B. F. Airway mucus function and dysfunction. N. Engl. J. Med. 363, 2233–2247 (2010)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Button, B. et al. A periciliary brush promotes the lung health by separating the mucus layer from airway epithelia. Science 337, 937–941 (2012)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  3. Young, H. W. et al. Central role of Muc5ac expression in mucous metaplasia and its regulation by conserved 5′ elements. Am. J. Respir. Cell Mol. Biol. 37, 273–290 (2007)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. The Collaborative Study on the Genetics of Asthma. A genome-wide search for asthma susceptibility loci in ethnically diverse populations. Nature Genet. 15, 389–392 (1997)

  5. Seibold, M. A. et al. A common MUC5B promoter polymorphism and pulmonary fibrosis. N. Engl. J. Med. 364, 1503–1512 (2011)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Kamio, K. et al. Promoter analysis and aberrant expression of the MUC5B gene in diffuse panbronchiolitis. Am. J. Respir. Crit. Care Med. 171, 949–957 (2005)

    Article  PubMed  Google Scholar 

  7. Janssen, W. J. et al. Fas determines differential fates of resident and recruited macrophages during resolution of acute lung injury. Am. J. Respir. Crit. Care Med. 184, 547–560 (2011)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Thornton, D. J., Rousseau, K. & McGuckin, M. A. Structure and function of the polymeric mucins in airways mucus. Annu. Rev. Physiol. 70, 459–486 (2008)

    Article  CAS  PubMed  Google Scholar 

  9. Woodruff, P. G. et al. T-helper type 2-driven inflammation defines major subphenotypes of asthma. Am. J. Respir. Crit. Care Med. 180, 388–395 (2009)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Innes, A. L. et al. Epithelial mucin stores are increased in the large airways of smokers with airflow obstruction. Chest 130, 1102–1108 (2006)

    Article  CAS  PubMed  Google Scholar 

  11. Ordoñez, C. L. et al. Mild and moderate asthma is associated with airway goblet cell hyperplasia and abnormalities in mucin gene expression. Am. J. Respir. Crit. Care Med. 163, 517–523 (2001)

    Article  PubMed  Google Scholar 

  12. Kawakubo, M. et al. Natural antibiotic function of a human gastric mucin against helicobacter pylori infection. Science 305, 1003–1006 (2004)

    Article  ADS  CAS  PubMed  Google Scholar 

  13. Johansson, M. E. et al. The inner of the two Muc2 mucin-dependent mucus layers in colon is devoid of bacteria. Proc. Natl Acad. Sci. USA 105, 15064–15069 (2008)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  14. Van der Sluis, M. et al. Muc2-deficient mice spontaneously develop colitis, indicating that MUC2 is critical for colonic protection. Gastroenterology 131, 117–129 (2006)

    Article  CAS  PubMed  Google Scholar 

  15. Velcich, A. et al. Colorectal cancer in mice genetically deficient in the mucin Muc2. Science 295, 1726–1729 (2002)

    Article  ADS  CAS  PubMed  Google Scholar 

  16. Hasnain, S. Z. et al. Muc5ac: a critical component mediating the rejection of enteric nematodes. J. Exp. Med. 208, 893–900 (2011)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Preciado, D. et al. Muc5b is the predominant mucin glycoprotein in chronic otitis media fluid. Pediatr. Res. 68, 231–236 (2010)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Labandeira-Rey, M. et al. Staphylococcus aureus panton-valentine leukocidin causes necrotizing pneumonia. Science 315, 1130–1133 (2007)

    Article  ADS  CAS  PubMed  Google Scholar 

  19. Stark, M. A. et al. Phagocytosis of apoptotic neutrophils regulates granulopoiesis via IL-23 and IL-17. Immunity 22, 285–294 (2005)

    Article  CAS  PubMed  Google Scholar 

  20. Patnode, M. L. et al. Galactose-6-o-sulfotransferases are not required for the generation of siglec-F ligands in leukocytes or lung tissue. J. Biol. Chem. 288, 26533–26545 (2013)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Moser, C. et al. β-defensin 1 contributes to pulmonary innate immunity in mice. Infect. Immun. 70, 3068–3072 (2002)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Manitz, M. P. et al. Loss of S100a9 (Mrp14) results in reduced interleukin-8-induced CD11b surface expression, a polarized microfilament system, and diminished responsiveness to chemoattractants in vitro. Mol. Cell. Biol. 23, 1034–1043 (2003)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Flo, T. H. et al. Lipocalin 2 mediates an innate immune response to bacterial infection by sequestrating iron. Nature 432, 917–921 (2004)

    Article  ADS  CAS  PubMed  Google Scholar 

  24. Ward, P. P., Mendoza-Meneses, M., Cunningham, G. A. & Conneely, O. M. Iron status in mice carrying a targeted disruption of lactoferrin. Mol. Cell. Biol. 23, 178–185 (2003)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. O'Riordan, T. G., Zwang, J. & Smaldone, G. C. Mucociliary clearance in adult asthma. Am. Rev. Respir. Dis. 146, 598–603 (1992)

    Article  CAS  PubMed  Google Scholar 

  26. Goodman, R. M., Yergin, B. M., Landa, J. F., Golivanux, M. H. & Sackner, M. A. Relationship of smoking history and pulmonary function tests to tracheal mucous velocity in nonsmokers, young smokers, ex-smokers, and patients with chronic bronchitis. Am. Rev. Respir. Dis. 117, 205–214 (1978)

    CAS  PubMed  Google Scholar 

  27. Rowe, S. M., Miller, S. & Sorscher, E. J. Cystic fibrosis. N. Engl. J. Med. 352, 1992–2001 (2005)

    Article  CAS  PubMed  Google Scholar 

  28. Talbot, T. R. et al. Asthma as a risk factor for invasive pneumococcal disease. N. Engl. J. Med. 352, 2082–2090 (2005)

    Article  CAS  PubMed  Google Scholar 

  29. Lee, T. A., Weaver, F. M. & Weiss, K. B. Impact of pneumococcal vaccination on pneumonia rates in patients with COPD and asthma. J. Gen. Intern. Med. 22, 62–67 (2007)

    Article  PubMed  PubMed Central  Google Scholar 

  30. Hendley, J. O. Clinical practice. Otitis media. N. Engl. J. Med. 347, 1169–1174 (2002)

    Article  PubMed  Google Scholar 

  31. Yu, D. et al. An efficient recombination system for chromosome engineering in Escherichia coli. Proc. Natl Acad. Sci. USA 97, 5978–5983 (2000)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  32. Zhu, Y. et al. Munc13-2−/− baseline secretion defect reveals source of oligomeric mucins in mouse airways. J. Physiol. (Lond.) 586, 1977–1992 (2008)

    Article  CAS  Google Scholar 

  33. Roy, M. G. et al. Mucin production during prenatal and postnatal murine lung development. Am. J. Respir. Cell Mol. Biol. 44, 755–760 (2011)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Sisson, J. H., Stoner, J. A., Ammons, B. A. & Wyatt, T. A. All-digital image capture and whole-field analysis of ciliary beat frequency. J. Microsc. 211, 103–111 (2003)

    Article  MathSciNet  CAS  PubMed  Google Scholar 

  35. Voronina, V. A. et al. Inactivation of chibby affects function of motile airway cilia. J. Cell Biol. 185, 225–233 (2009)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Coote, K., Nicholls, A., Atherton, H. C., Sugar, R. & Danahay, H. Mucociliary clearance is enhanced in rat models of cigarette smoke and lipopolysaccharide-induced lung disease. Exp. Lung Res. 30, 59–71 (2004)

    Article  CAS  PubMed  Google Scholar 

  37. You, Y., Richer, E. J., Huang, T. & Brody, S. L. Growth and differentiation of mouse tracheal epithelial cells: Selection of a proliferative population. Am. J. Physiol. Lung Cell. Mol. Physiol. 283, L1315–L1321 (2002)

    Article  CAS  PubMed  Google Scholar 

  38. Dillard, P., Wetsel, R. A. & Drouin, S. M. Complement c3a regulates muc5ac expression by airway clara cells independently of th2 responses. Am. J. Respir. Crit. Care Med. 175, 1250–1258 (2007)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Matsui, H. et al. Evidence for periciliary liquid layer depletion, not abnormal ion composition, in the pathogenesis of cystic fibrosis airways disease. Cell 95, 1005–1015 (1998)

    Article  CAS  PubMed  Google Scholar 

  40. Okada, S. F. et al. Voltage-dependent anion channel-1 (vdac-1) contributes to atp release and cell volume regulation in murine cells. J. Gen. Physiol. 124, 513–526 (2004)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Weinberg, E. D. Iron withholding: a defense against infection and neoplasia. Physiol. Rev. 64, 65–102 (1984)

    Article  CAS  PubMed  Google Scholar 

  42. Weinberg, E. D. Nutritional immunity. Host's attempt to withold iron from microbial invaders. J. Am. Med. Assoc. 231, 39–41 (1975)

    Article  CAS  Google Scholar 

  43. Weinberg, E. D. Iron and susceptibility to infectious disease. Science 184, 952–956 (1974)

    Article  ADS  CAS  PubMed  Google Scholar 

  44. Evans, S. E. et al. Stimulated innate resistance of lung epithelium protects mice broadly against bacteria and fungi. Am. J. Respir. Cell Mol. Biol. 42, 40–50 (2010)

    Article  CAS  PubMed  Google Scholar 

  45. DeSantis, T. Z. et al. Greengenes, a chimera-checked 16S rRNA gene database and workbench compatible with ARB. Appl. Environ. Microbiol. 72, 5069–5072 (2006)

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  46. Drancourt, M. et al. 16s ribosomal DNA sequence analysis of a large collection of environmental and clinical unidentifiable bacterial isolates. J. Clin. Microbiol. 38, 3623–3630 (2000)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Moghaddam, S. J. et al. Haemophilus influenzae lysate induces aspects of the chronic obstructive pulmonary disease phenotype. Am. J. Respir. Cell Mol. Biol. 38, 629–638 (2008)

    Article  CAS  PubMed  Google Scholar 

  48. Steinkamp, J. A., Wilson, J. S., Saunders, G. C. & Stewart, C. C. Phagocytosis: flow cytometric quantitation with fluorescent microspheres. Science 215, 64–66 (1982)

    Article  ADS  CAS  PubMed  Google Scholar 

  49. Lozupone, C. et al. Widespread colonization of the lung by Tropheryma whipplei in HIV infection. Am. J. Respir. Crit. Care Med. 187, 1110–1117 (2013)

    Article  PubMed  PubMed Central  Google Scholar 

  50. Moghaddam, S. J. et al. Promotion of lung carcinogenesis by chronic obstructive pulmonary disease-like airway inflammation in a k-ras-induced mouse model. Am. J. Respir. Cell Mol. Biol. 40, 443–453 (2009)

    Article  CAS  PubMed  Google Scholar 

  51. Evans, C. M. et al. Mucin is produced by clara cells in the proximal airways of antigen-challenged mice. Am. J. Respir. Cell Mol. Biol. 31, 382–394 (2004)

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank F. Ttofali, D. Harper, D. Raclawska, V. Mdoe, C. Ramsey and J. Parker-Thornburg for their assistance. We also thank K. Naff and the MD Anderson Cancer Center Department of Veterinary Medicine and Surgery for support in animal care. This work was supported by the National Institutes of Health Grants R01 HL080396 (C.M.E.); R01 AA008769 (J.H.S.), R01 HL109517 (W.J.J.); R01 HL114381 (P.M.H.); R01 HL097000 (B.F.D.), P01 HL108808, P01 HL110873, P50 HL107168, P30 DK065988 (R.C.B.), Medical Research Council Grant G1000450 (D.J.T.) and Cystic Fibrosis Foundation Grants 06IO (C.M.E.) and RDP R026-CR11 (R.C.B.). Additional support was provided by National Institutes of Health Cancer Center Support Grants CA016672 for the MD Anderson Cancer Center and CA046934 for the University of Colorado transgenic mouse facilities; by CA016086 for the UNC Biomedical Research Imaging Center Small Animal Imaging Facility, and for the UNC Michael Hooker Microscopy Facility funded by an anonymous private donor.

Author information

Authors and Affiliations

Authors

Contributions

M.G.R., A.L.-B. and A.A.F designed and performed survival, histological, particle clearance, inflammation, and infectious agent identification experiments, performed and collected data for S. aureus infection, macrophage and neutrophil identification experiments, and cytokine analyses. A.M.W., R.C.K., C.E.H. and D.A.S. generated and assisted in studies in Scgb1a1-Muc5b mice. M.M.M., R.M.B, I.R., A.S.B., M.G.B., W.K.O., K.A.T., S.C.F., A.C.-G., J.M.D. and C.A.L. performed infectious pneumonia and infectious agent identification experiments. S.N.A., L.K.B., A.S.S. and Y.M.P. constructed and generated Muc5b knockout mice. S.E.E., M.M.D.G., S.J.M. and M.J.T. assisted in the design and performance of inflammation studies. B.R.G., R.A., H.K.-Q. and M.R.B. assisted in the design and performance of hypoxemia studies. J.H.S., S.M.D. and B.R.G. assisted in the design and performance of tracheal and nasal mucociliary function studies. D.J.T. and K.R. provided purified MUC5B protein. W.J.J. and L.B. designed and performed macrophage activation and apoptosis assays. I.V.Y, P.M.H., P.G.W., C.W.D., R.C.B and B.F.D. assisted in the analysis and interpretation of data, and C.W.D. provided Muc5b antisera. C.M.E. designed the study, analysed data, and wrote the manuscript.

Corresponding author

Correspondence to Christopher M. Evans.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Extended data figures and tables

Extended Data Figure 1 Generation and characterization of Muc5ac- and Muc5b-mutant mice.

a, Muc5b-knockout mice were generated by flanking exon 1 with loxP sites, selecting embryonic stem cells for neomycin resistance, and crossing allele-positive mice with germline active FLP and Cre deleter strains. b, Muc5b transgenic mice were generated by inserting a full-length Muc5b genomic DNA (gDNA) transgene driven by the airway specific rat Scgb1a1 promoter. c, Muc5b transgene expression throughout the conducting airways (top part, bronchi; bottom part, bronchioles). Note the lack of Muc5ac induction in Scgb1a1-Muc5b mouse airways (top right). d, Immunoblot analysis of wild-type (WT) and Scgb1a1-Muc5b mouse lung homogenates. e, Pulmonary MCC in Scgb1a1-Muc5b mice. f, Loss of Muc5ac and retention of Muc5b in ovalbumin sensitized and challenged Muc5ac−/− airways. Tg, transgenic.

Extended Data Figure 2 Pathways for the clearance of bacteria and particles from the lower airways.

Inhaled agents such as bacteria (yellow) impact upon epithelium-lined surfaces in conducting airways or deposit in alveoli. Bacteria trapped in surface and glandular mucous secretions (green) are eliminated directly by MCC (1) or are killed and then eliminated by MCC (2). Bacteria that evade these mechanisms or deposit more deeply in the lungs are detected and phagocytosed by dendritic cells (DCs) and macrophages (3; present in the airways and alveoli, but shown only in the latter here for clarity). DCs and macrophages secrete IL-23 and facilitate innate anti-bacterial immune responses through innate myeloid- and lymphoid-cell (4), and monocyte and granulocyte recruitment (5). Recruited inflammatory cells accumulate in the lungs and kill bacteria through phagocytic, exocytic and extracellular trap-mediated mechanisms (6). To resolve inflammation, recruited leukocytes undergo apoptosis and are phagocytosed by macrophages, which then repress IL-23 production (7), undergo apoptosis (8) and are eliminated by MCC. In the studies presented here, we demonstrate that these defences are critically dependent upon the expression of Muc5b.

Extended Data Figure 3 Increased Muc5ac production in Muc5b−/− mice.

a, Steady-state Muc5ac messenger RNA levels in Muc5b+/+ and Muc5b−/− mice (age 12 weeks, n = 3 per group). *P = 0.019, one-tailed t-test. b, c, AB-PAS and Muc5ac staining (arrows) in Muc5b+/+ and Muc5b−/− mice. Note the increased PAS (pink) and lack of AB (blue or purple) positivity in Muc5b−/− airway goblet cells.

Extended Data Figure 4 Otitis media and upper airway congestion in Muc5b−/− mice.

a, b, Micro-CT reveals dark and radiopaque middle-ear cavities (red arrows) in Muc5b+/+ and Muc5b−/− mice, respectively. c, Mass of accumulated mucus plugs removed from nasal cavities at the indicated time points. Plug mass was compared to age using Pearson’s product–moment correlation (ρ). d, Hair in the middle ear of a Muc5b−/− mouse. The ventral side of the tympanic bulla was removed to visualize middle-ear content. White arrowhead identifies encased hair fragments. Et, Eustachian tube; tm, tympanic membrane.

Extended Data Figure 5 Further characterization of bacteria in Muc5b−/− mouse lungs.

a–e, Bacteria grown from lungs under microaerophilic conditions. a, b, Total growth in 12-week-old Muc5b−/− and Muc5b+/+ mice, and in moribund and control mice. ce, Bacterial genera, staphylococcal species and S. aureus incidence. f–h, Streptococcal group and S. acidominimus growth in mouse lungs. f, Streptococci identified by taxonomic classifications were assessed for abundance (g) and prevalence (h). Symbols identify samples from the lungs of individual Muc5b+/+ (blue) and Muc5b−/− (red) mice at 12-week-old (open circles) and moribund or cage-mate control cohorts (filled circles). Data were analysed by t-tests in a, b, ANOVA in c, d, g, and using chi-squared tests in e, h. P < 0.05 was considered significant. Total numbers of mice in each group are listed as denominators below e and h. ND, none detected; NS, not significant.

Extended Data Figure 6 Systemic polymicrobial infection in Muc5b−/− mice.

a–e, Bacteria from spleen homogenates obtained from moribund Muc5b−/− mice and Muc5b+/+ controls were identified by 16SrRNA gene sequencing. a, Genera in spleens from Muc5b−/− mice (red circles). Staphylococcus was identified in the spleen of a single Muc5b+/+ control (blue circle), but its incidence was 20% compared to 80% in Muc5b−/− mice (b). c, Sequence identification of staphylococci present. d, e, Prevalence and abundance of streptococci. Data were analysed by ANOVA in a, c, e, and using chi-squared tests in b, d. P < 0.05 was considered significant. Total numbers of mice in each group are listed as denominators below b and d.

Extended Data Figure 7 Effects of airway mucins on infection and growth of S. aureus.

a–e, USA300 infection (107 CFU intranasally) in Muc5b+/+ and Muc5b−/− mice. a, Changes in body weights during the first week in Muc5b+/+ animals (blue circles, n = 7), Muc5b−/− animals that recovered fully from infection (open red circles, n = 4), and Muc5b−/− animals that did not survive to 21 days (filled red circles, n = 6). b–e, On day 21 (surviving mice), or on the day in which a >15% loss of body weight was discovered (non-surviving mice), lungs and spleens were cultured. One non-survivor was found to have died in its cage and not cultured. Incidence of positive cultures (b) and numbers of CFUs (c) in infected Muc5b+/+ and Muc5b−/− lungs. d, e, Spleen-culture incidences and numbers of CFUs. Error bars, s.e.m. in a, c, e. Data were analysed using Student’s t-test in a (day 7) and c, and Fisher’s exact test in b and d. Because all values for Muc5b+/+ mice are zeroes in e, Mann–Whitney and Wilcoxon ranked sums tests were used. For all statistical analyses, P < 0.05 was considered significant. f, USA300 growth in medium containing MUC5AC or MUC5B (2–250 µg ml−1). Purified mucin stocks were solubilized in urea. Separate growth curves were thus also measured in serially diluted urea at matched concentrations (1.2–150 mM). Each line represents mean growth at a single concentration of mucin or urea over time. Error bars, s.d. from triplicate experiments.

Extended Data Figure 8 Effects of Muc5b expression on pulmonary inflammation.

a, Lung lavage leukocytes from Muc5b+/+ (blue, n = 15, 23, 7), Scgb1a1-Muc5b (green, n = 11, 18, 5), and Muc5b−/− (red, n = 12, 5, 5) mice in the steady state at ages 3, 6 and 12 months and moribund Muc5b−/− animals (red; n = 8, various ages) and Muc5b+/+ cagemates (blue, n = 6). *P < 0.05 by ANOVA (timecourse studies) and by Student’s t-test (moribund Muc5b−/− versus Muc5b+/+ cage mates). b, Three-month-old Muc5b+/+ (blue, n = 7), Scgb1a1-Muc5b (green, n = 3) and Muc5b−/− (red, n = 3) mice were challenged intratracheally with 5 × 107 CFU USA300. Inflammation was assessed 24 h post infection. No significant differences in macrophages or neutrophils were seen among groups. Error bars, s.e.m. c, Young (3 months of age) and ageing (6–7 months of age) Muc5b+/+ (blue), Scgb1a1-Muc5b (green) and Muc5b−/− (red) mice were exposed to 2–5 × 107CFU of live or heat-killed Staphylococcus USA300, or to saline (SAL) as indicated. Lung lavage leukocytes were assessed 7 days post challenge. Ageing Muc5b−/− mice died before lavage sampling. Infected Muc5ac−/− mice showed no differences in inflammation compared to Muc5b+/+ or C57BL/6J (not shown). Error bars, s.e.m. Numbers in brackets, n mice. *P < 0.05 across genotypes by ANOVA and within genotypes by Student’s t-test. Tg, Scgb1a1-Muc5b transgenic.

Extended Data Figure 9 Effects of Muc5b expression on pulmonary macrophages.

a, Microsphere phagocytosis in Muc5b+/+ and Muc5b−/− mouse lung macrophages. Data are percentages of cells containing microspheres per mouse. Error bars, s.e.m. *P < 0.05 between groups as determined by Student’s t-test. b, Muc5b was not detected in lung lavage macrophages from allergic wild-type mice. Data are representative of five mice. Scale bar, 5 μm. c, Accumulation of efferocytotic macrophages in Muc5b−/− lung tissues. Neutrophils in lung tissues from Muc5b+/+ and Muc5b−/− mice were immunolabelled and stained with DAB (brown). Asterisk in lower right panel identifies whole neutrophils contained within pulmonary macrophages. Arrows identify areas in c shown at high magnification. Scale bar, 50 μm in low-magnification and 15 μm in high-magnification images.

Extended Data Table 1 Cytokines and chemokines in mouse lung lavage fluid

Supplementary information

Mucociliary transport in Muc5b+/+ mTEC cultures

Microsphere transport measured by video microscopy. Time-lapse video speed is 300x. (MPG 503 kb)

Mucociliary transport in Muc5b-/- mTEC cultures

Microsphere transport measured by video microscopy. Time-lapse video speed is 300x. (MPG 503 kb)

Labored breathing pattern of a moribund Muc5b-/- mouse before tracheostomy

An isoflurane anesthetized Muc5b-/- mouse displays deep and extended ventilatory patterns. (AVI 9397 kb)

Quiet tidal breathing pattern of a moribund Muc5b-/- mouse immediately after tracheostomy

Ventilation following tracheostomy is shallow and rapid. Note isoflurane administration through the tracheal cannula. (AVI 11064 kb)

PowerPoint slides

Source data

Rights and permissions

Reprints and permissions

About this article

Cite this article

Roy, M., Livraghi-Butrico, A., Fletcher, A. et al. Muc5b is required for airway defence. Nature 505, 412–416 (2014). https://doi.org/10.1038/nature12807

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature12807

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research