Skip to main content

Advertisement

Log in

Cross-roads in the lung: immune cells and tissue interactions as determinants of allergic asthma

  • Singapore Immunology Network
  • Published:
Immunologic Research Aims and scope Submit manuscript

Abstract

Allergic asthma is a chronic disease of the lung characterized by underlying Th2- and IgE-mediated inflammation, structural alterations of the bronchial wall, and airway hyperresponsiveness. Initial allergic sensitization and later development of chronic disease are determined by close interactions between lung structural cells and the resident and migratory immune cells in the lung. Epithelial cells play a crucial role in allergic sensitization by directly influencing dendritic cells induction of tolerant or effector T cells and production of type 2 cytokines by innate immune cells. During chronic disease, the bronchial epithelium, stroma, and smooth muscle become structurally and functionally altered, contributing to the perpetuation of tissue remodeling. Thus, targeting tissue-driven pathology in addition to inflammation may increase the effectiveness of asthma treatment.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1

Similar content being viewed by others

References

  1. Seibold MA, Schwartz DA. The lung: the natural boundary between nature and nurture. Annu Rev Physiol. 2011;17(73):457–78.

    Article  CAS  Google Scholar 

  2. Al-Muhsen S, Johnson JR, Hamid Q. Remodeling in asthma. J Allergy Clin Immunol. 2011;128(3):451–62; quiz 63–4.

    Google Scholar 

  3. Panettieri RA Jr, Covar R, Grant E, Hillyer EV, Bacharier L. Natural history of asthma: persistence versus progression-does the beginning predict the end? J Allergy Clin Immunol. 2008;121(3):607–13.

    Google Scholar 

  4. Curotto de Lafaille MA, Lafaille JJ, Graca L. Mechanisms of tolerance and allergic sensitization in the airways and the lungs. Curr Opin Immunol. 2010;22(5):616–22.

    Google Scholar 

  5. Curotto de Lafaille MA, Lafaille JJ. Natural and adaptive foxp3+ regulatory T cells: more of the same or a division of labor? Immunity 2009;30(5):626–35.

    Google Scholar 

  6. Hammad H, Lambrecht BN. Dendritic cells and airway epithelial cells at the interface between innate and adaptive immune responses. Allergy. 2011;66(5):579–87.

    Article  PubMed  CAS  Google Scholar 

  7. Holgate ST. The sentinel role of the airway epithelium in asthma pathogenesis. Immunol Rev. 2011;242(1):205–19.

    Article  PubMed  CAS  Google Scholar 

  8. Eisele NA. Host defense and the airway epithelium: frontline responses that protect against bacterial invasion and pneumonia. J Pathogens 2011;2011.

  9. Hong KU, Reynolds SD, Watkins S, Fuchs E, Stripp BR. In vivo differentiation potential of tracheal basal cells: evidence for multipotent and unipotent subpopulations. Am J Physiol Lung Cell Mol Physiol. 2004;286(4):L643–9.

    Article  PubMed  CAS  Google Scholar 

  10. Schoch KG, Lori A, Burns KA, Eldred T, Olsen JC, Randell SH. A subset of mouse tracheal epithelial basal cells generates large colonies in vitro. Am J Physiol Lung Cell Mol Physiol. 2004;286(4):L631–42.

    Article  PubMed  CAS  Google Scholar 

  11. Stripp BR. Hierarchical organization of lung progenitor cells: is there an adult lung tissue stem cell? Proc Am Thorac Soc. 2008;5(6):695–8.

    Article  PubMed  Google Scholar 

  12. Plopper CG, Nishio SJ, Alley JL, Kass P, Hyde DM. The role of the nonciliated bronchiolar epithelial (Clara) cell as the progenitor cell during bronchiolar epithelial differentiation in the perinatal rabbit lung. Am J Respir Cell Mol Biol. 1992;7(6):606–13.

    PubMed  CAS  Google Scholar 

  13. Reynolds SD, Giangreco A, Power JHT, Stripp BR. Neuroepithelial bodies of pulmonary airways serve as a reservoir of progenitor cells capable of epithelial regeneration. Am J Pathol. 2000;156(1):269–78.

    Google Scholar 

  14. Giangreco A, Reynolds SD, Stripp BR. Terminal bronchioles harbor a unique airway stem cell population that localizes to the bronchoalveolar duct junction. Am J Pathol. 2002;161(1):173–82.

    Article  PubMed  Google Scholar 

  15. Kim CF, Jackson EL, Woolfenden AE, Lawrence S, Babar I, Vogel S, Crowley D, Bronson RT, Jacks T. Identification of bronchioalveolar stem cells in normal lung and lung cancer. Cell. 2005;121(6):823–35.

    Article  PubMed  CAS  Google Scholar 

  16. Parker D, Prince A. Innate immunity in the respiratory epithelium. Am J Respir Cell Mol Biol. 2011;45(2):189–201.

    Article  PubMed  CAS  Google Scholar 

  17. Proud D, Leigh R. Epithelial cells and airway diseases. Immunol Rev. 2011;242(1):186–204.

    Article  PubMed  CAS  Google Scholar 

  18. Saenz SA, Taylor BC, Artis D. Welcome to the neighborhood: epithelial cell-derived cytokines license innate and adaptive immune responses at mucosal sites. Immunol Rev. 2008;226:172–90.

    Article  PubMed  CAS  Google Scholar 

  19. Liew FY, Pitman NI, McInnes IB. Disease-associated functions of IL-33: the new kid in the IL-1 family. Nat Rev Immunol. 2010;10(2):103–10.

    Article  PubMed  CAS  Google Scholar 

  20. Liu YJ. TSLP in epithelial cell and dendritic cell cross talk. Adv Immunol. 2009;101:1–25.

    Article  PubMed  CAS  Google Scholar 

  21. He R, Geha RS. Thymic stromal lymphopoietin. Ann N Y Acad Sci. 2010;1183:13–24.

    Article  PubMed  CAS  Google Scholar 

  22. Holgate ST, Lackie P, Wilson S, Roche W, Davies D. Bronchial epithelium as a key regulator of airway allergen sensitization and remodeling in asthma. Am J Respir Crit Care Med. 2000;162(3 Pt 2):S113–7.

    PubMed  CAS  Google Scholar 

  23. Wills-Karp M, Nathan A, Page K, Karp CL. New insights into innate immune mechanisms underlying allergenicity. Mucosal Immunol. 2010;3(2):104–10.

    Article  PubMed  CAS  Google Scholar 

  24. Hammad H, Chieppa M, Perros F, Willart MA, Germain RN, Lambrecht BN. House dust mite allergen induces asthma via Toll-like receptor 4 triggering of airway structural cells. Nat Med. 2009;15(4):410–6.

    Article  PubMed  CAS  Google Scholar 

  25. Trompette A, Divanovic S, Visintin A, Blanchard C, Hegde RS, Madan R, Thorne PS, Wills-Karp M, Gioannini TL, Weiss JP, Karp CL. Allergenicity resulting from functional mimicry of a Toll-like receptor complex protein. Nature. 2009;457(7229):585–8.

    Article  PubMed  CAS  Google Scholar 

  26. Davis BK, Wen H, Ting JP. The inflammasome NLRs in immunity, inflammation, and associated diseases. Annu Rev Immunol. 2011;23(29):707–35.

    Article  CAS  Google Scholar 

  27. Dostert C, Petrilli V, Van Bruggen R, Steele C, Mossman BT, Tschopp J. Innate immune activation through Nalp3 inflammasome sensing of asbestos and silica. Science. 2008;320(5876):674–7.

    Article  PubMed  CAS  Google Scholar 

  28. Kool M, Willart MA, van Nimwegen M, Bergen I, Pouliot P, Virchow JC, Rogers N, Osorio F, Reis ESC, Hammad H, Lambrecht BN. An unexpected role for uric acid as an inducer of T helper 2 cell immunity to inhaled antigens and inflammatory mediator of allergic asthma. Immunity. 2011;34(4):527–40.

    Article  PubMed  CAS  Google Scholar 

  29. Yin J, Xu K, Zhang J, Kumar A, Yu FS. Wound-induced ATP release and EGF receptor activation in epithelial cells. J Cell Sci. 2007;120(Pt 5):815–25.

    Article  PubMed  CAS  Google Scholar 

  30. Idzko M, Hammad H, van Nimwegen M, Kool M, Willart MA, Muskens F, Hoogsteden HC, Luttmann W, Ferrari D, Di Virgilio F, Virchow JC Jr, Lambrecht BN. Extracellular ATP triggers and maintains asthmatic airway inflammation by activating dendritic cells. Nat Med. 2007;13(8):913–9.

    Article  PubMed  CAS  Google Scholar 

  31. Kouzaki H, Iijima K, Kobayashi T, O’Grady SM, Kita H. The danger signal, extracellular ATP, is a sensor for an airborne allergen and triggers IL-33 release and innate Th2-type responses. J Immunol. 2011;186(7):4375–87.

    Article  PubMed  CAS  Google Scholar 

  32. Hammad H, Lambrecht BN. Dendritic cells and epithelial cells: linking innate and adaptive immunity in asthma. Nat Rev Immunol. 2008;8(3):193–204.

    Article  PubMed  CAS  Google Scholar 

  33. Ovrevik J, Lag M, Holme JA, Schwarze PE, Refsnes M. Cytokine and chemokine expression patterns in lung epithelial cells exposed to components characteristic of particulate air pollution. Toxicology. 2009;259(1–2):46–53.

    Article  PubMed  CAS  Google Scholar 

  34. Reibman J, Hsu Y, Chen LC, Bleck B, Gordon T. Airway epithelial cells release MIP-3alpha/CCL20 in response to cytokines and ambient particulate matter. Am J Respir Cell Mol Biol. 2003;28(6):648–54.

    Article  PubMed  CAS  Google Scholar 

  35. Sung SS, Fu SM, Rose CE Jr, Gaskin F, Ju ST, Beaty SR. A major lung CD103 (alphaE)-beta7 integrin-positive epithelial dendritic cell population expressing Langerin and tight junction proteins. J Immunol. 2006;176(4):2161–72.

    PubMed  CAS  Google Scholar 

  36. Bleck B, Tse DB, Curotto de Lafaille MA, Zhang F, Reibman J. Diesel exhaust particle-exposed human bronchial epithelial cells induce dendritic cell maturation and polarization via thymic stromal lymphopoietin. J Clin Immunol. 2008;28(2):147–56.

    Article  PubMed  Google Scholar 

  37. Barnes PJ. The cytokine network in asthma and chronic obstructive pulmonary disease. J Clin Invest. 2008;118(11):3546–56.

    Article  PubMed  CAS  Google Scholar 

  38. Eisenbarth SC, Piggott DA, Huleatt JW, Visintin I, Herrick CA, Bottomly K. Lipopolysaccharide-enhanced, toll-like receptor 4-dependent T helper cell type 2 responses to inhaled antigen. J Exp Med. 2002;196(12):1645–51.

    Article  PubMed  CAS  Google Scholar 

  39. Asokananthan N, Graham PT, Stewart DJ, Bakker AJ, Eidne KA, Thompson PJ, Stewart GA. House dust mite allergens induce proinflammatory cytokines from respiratory epithelial cells: the cysteine protease allergen, Der p 1, activates protease-activated receptor (PAR)-2 and inactivates PAR-1. J Immunol. 2002;169(8):4572–8.

    PubMed  CAS  Google Scholar 

  40. King C, Brennan S, Thompson PJ, Stewart GA. Dust mite proteolytic allergens induce cytokine release from cultured airway epithelium. J Immunol. 1998;161(7):3645–51.

    PubMed  CAS  Google Scholar 

  41. Reibman J, Hsu Y, Chen LC, Kumar A, Su WC, Choy W, Talbot A, Gordon T. Size fractions of ambient particulate matter induce granulocyte macrophage colony-stimulating factor in human bronchial epithelial cells by mitogen-activated protein kinase pathways. Am J Respir Cell Mol Biol. 2002;27(4):455–62.

    PubMed  CAS  Google Scholar 

  42. Stampfli MR, Wiley RE, Neigh GS, Gajewska BU, Lei XF, Snider DP, Xing Z, Jordana M. GM-CSF transgene expression in the airway allows aerosolized ovalbumin to induce allergic sensitization in mice. J Clin Invest. 1998;102(9):1704–14.

    Article  PubMed  CAS  Google Scholar 

  43. Finkelman FD, Hogan SP, Hershey GK, Rothenberg ME, Wills-Karp M. Importance of cytokines in murine allergic airway disease and human asthma. J Immunol. 2010;184(4):1663–74.

    Article  PubMed  CAS  Google Scholar 

  44. Simeonova PP, Toriumi W, Kommineni C, Erkan M, Munson AE, Rom WN, Luster MI. Molecular regulation of IL-6 activation by asbestos in lung epithelial cells: role of reactive oxygen species. J Immunol. 1997;159(8):3921–8.

    PubMed  CAS  Google Scholar 

  45. Neveu WA, Allard JB, Dienz O, Wargo MJ, Ciliberto G, Whittaker LA, Rincon M. IL-6 is required for airway mucus production induced by inhaled fungal allergens. J Immunol. 2009;183(3):1732–8.

    Article  PubMed  CAS  Google Scholar 

  46. Neveu WA, Bernardo E, Allard JL, Nagaleekar V, Wargo MJ, Davis RJ, Iwakura Y, Whittaker LA, Rincon M. Fungal allergen beta-glucans trigger p38 mitogen-activated protein kinase-mediated IL-6 translation in lung epithelial cells. Am J Respir Cell Mol Biol. 2011;45(6):1133–41.

    Article  PubMed  CAS  Google Scholar 

  47. Yokoyama A, Kohno N, Fujino S, Hamada H, Inoue Y, Fujioka S, Ishida S, Hiwada K. Circulating interleukin-6 levels in patients with bronchial asthma. Am J Respir Crit Care Med. 1995;151(5):1354–8.

    PubMed  CAS  Google Scholar 

  48. Neveu WA, Allard JL, Raymond DM, Bourassa LM, Burns SM, Bunn JY, Irvin CG, Kaminsky DA, Rincon M. Elevation of IL-6 in the allergic asthmatic airway is independent of inflammation but associates with loss of central airway function. Respir Res. 2010;11:28.

    Article  PubMed  CAS  Google Scholar 

  49. Medoff BD, Thomas SY, Luster AD. T cell trafficking in allergic asthma: the ins and outs. Annu Rev Immunol. 2008;26:205–32.

    Article  PubMed  CAS  Google Scholar 

  50. Vlahakis NE, Schroeder MA, Limper AH, Hubmayr RD. Stretch induces cytokine release by alveolar epithelial cells in vitro. Am J Physiol. 1999;277(1 Pt 1):L167–73.

    PubMed  CAS  Google Scholar 

  51. Wu W, Samet JM, Ghio AJ, Devlin RB. Activation of the EGF receptor signaling pathway in airway epithelial cells exposed to Utah Valley PM. Am J Physiol Lung Cell Mol Physiol. 2001;281(2):L483–9.

    PubMed  CAS  Google Scholar 

  52. Garofalo R, Sabry M, Jamaluddin M, Yu RK, Casola A, Ogra PL, Brasier AR. Transcriptional activation of the interleukin-8 gene by respiratory syncytial virus infection in alveolar epithelial cells: nuclear translocation of the RelA transcription factor as a mechanism producing airway mucosal inflammation. J Virol. 1996;70(12):8773–81.

    PubMed  CAS  Google Scholar 

  53. Bautista MV, Chen Y, Ivanova VS, Rahimi MK, Watson AM, Rose MC. IL-8 regulates mucin gene expression at the posttranscriptional level in lung epithelial cells. J Immunol. 2009;183(3):2159–66.

    Article  PubMed  CAS  Google Scholar 

  54. Berin MC, Eckmann L, Broide DH, Kagnoff MF. Regulated production of the T helper 2-type T-cell chemoattractant TARC by human bronchial epithelial cells in vitro and in human lung xenografts. Am J Respir Cell Mol Biol. 2001;24(4):382–9.

    PubMed  CAS  Google Scholar 

  55. Heijink IH, Marcel Kies P, van Oosterhout AJ, Postma DS, Kauffman HF, Vellenga E. Der p, IL-4, and TGF-beta cooperatively induce EGFR-dependent TARC expression in airway epithelium. Am J Respir Cell Mol Biol. 2007;36(3):351–9.

    Article  PubMed  CAS  Google Scholar 

  56. Zhou B, Comeau MR, De Smedt T, Liggitt HD, Dahl ME, Lewis DB, Gyarmati D, Aye T, Campbell DJ, Ziegler SF. Thymic stromal lymphopoietin as a key initiator of allergic airway inflammation in mice. Nat Immunol. 2005;6(10):1047–53.

    Article  PubMed  CAS  Google Scholar 

  57. Al-Shami A, Spolski R, Kelly J, Keane-Myers A, Leonard WJ. A role for TSLP in the development of inflammation in an asthma model. J Exp Med. 2005;202(6):829–39.

    Article  PubMed  CAS  Google Scholar 

  58. Liu YJ, Soumelis V, Watanabe N, Ito T, Wang YH, Malefyt Rde W, Omori M, Zhou B, Ziegler SF. TSLP: an epithelial cell cytokine that regulates T cell differentiation by conditioning dendritic cell maturation. Annu Rev Immunol. 2007;25:193–219.

    Article  PubMed  CAS  Google Scholar 

  59. Bleck B, Tse DB, Gordon T, Ahsan MR, Reibman J. Diesel exhaust particle-treated human bronchial epithelial cells upregulate Jagged-1 and OX40 ligand in myeloid dendritic cells via thymic stromal lymphopoietin. J Immunol. 2010;185(11):6636–45.

    Article  PubMed  CAS  Google Scholar 

  60. Nakano H, Free ME, Whitehead GS, Maruoka S, Wilson RH, Nakano K, Cook DN. Pulmonary CD103(+) dendritic cells prime Th2 responses to inhaled allergens. Mucosal Immunol. 2012;5(1):53–65.

    Google Scholar 

  61. Fort MM, Cheung J, Yen D, Li J, Zurawski SM, Lo S, Menon S, Clifford T, Hunte B, Lesley R, Muchamuel T, Hurst SD, Zurawski G, Leach MW, Gorman DM, Rennick DM. IL-25 induces IL-4, IL-5, and IL-13 and Th2-associated pathologies in vivo. Immunity. 2001;15(6):985–95.

    Article  PubMed  CAS  Google Scholar 

  62. Saenz SA, Noti M, Artis D. Innate immune cell populations function as initiators and effectors in Th2 cytokine responses. Trends Immunol. 2010;31(11):407–13.

    Article  PubMed  CAS  Google Scholar 

  63. Koyasu S, Moro K. Type 2 innate immune responses and the natural helper cell. Immunology. 2011;132(4):475–81.

    Article  PubMed  CAS  Google Scholar 

  64. Neill DR, Wong SH, Bellosi A, Flynn RJ, Daly M, Langford TK, Bucks C, Kane CM, Fallon PG, Pannell R, Jolin HE, McKenzie AN. Nuocytes represent a new innate effector leukocyte that mediates type-2 immunity. Nature. 2010;464(7293):1367–70.

    Article  PubMed  CAS  Google Scholar 

  65. Monticelli LA, Sonnenberg GF, Abt MC, Alenghat T, Ziegler CG, Doering TA, Angelosanto JM, Laidlaw BJ, Yang CY, Sathaliyawala T, Kubota M, Turner D, Diamond JM, Goldrath AW, Farber DL, Collman RG, Wherry EJ, Artis D. Innate lymphoid cells promote lung-tissue homeostasis after infection with influenza virus. Nat Immunol. 2011;12(11):1045–54.

    Google Scholar 

  66. Mjosberg JM, Trifari S, Crellin NK, Peters CP, van Drunen CM, Piet B, Fokkens WJ, Cupedo T, Spits H. Human IL-25- and IL-33-responsive type 2 innate lymphoid cells are defined by expression of CRTH2 and CD161. Nat Immunol. 2011;12(11):1055–62.

    Article  PubMed  CAS  Google Scholar 

  67. Li M, Li Q, Yang G, Kolosov VP, Perelman JM, Zhou XD. Cold temperature induces mucin hypersecretion from normal human bronchial epithelial cells in vitro through a transient receptor potential melastatin 8 (TRPM8)-mediated mechanism. J Allergy Clin Immunol. 2011;128(3):626–34 e1–5.

    Google Scholar 

  68. Park J-A, Drazen JM, Tschumperlin DJ. The chitinase-like protein YKL-40 is secreted by airway epithelial cells at base line and in response to compressive mechanical stress. J Biol Chem. 2010;285(39):29817–25.

    Google Scholar 

  69. Shiomi T, Tschumperlin DJ, Park JA, Sunnarborg SW, Horiuchi K, Blobel CP, Drazen JM. TNF-alpha-converting enzyme/a disintegrin and metalloprotease-17 mediates mechanotransduction in murine tracheal epithelial cells. Am J Respir Cell Mol Biol. 2011;45(2):376–85.

    Article  PubMed  CAS  Google Scholar 

  70. Allahverdian S, Harada N, Singhera GK, Knight DA, Dorscheid DR. Secretion of IL-13 by airway epithelial cells enhances epithelial repair via HB-EGF. Am J Respir Cell Mol Biol. 2008;38(2):153–60.

    Article  PubMed  CAS  Google Scholar 

  71. Grainge CL, Lau LC, Ward JA, Dulay V, Lahiff G, Wilson S, Holgate S, Davies DE, Howarth PH. Effect of bronchoconstriction on airway remodeling in asthma. N Engl J Med. 2011;364(21):2006–15.

    Article  PubMed  CAS  Google Scholar 

  72. Murray CS. Can inhaled corticosteroids influence the natural history of asthma? Curr Opin Allergy Clin Immunol. 2008;8(1):77–81.

  73. Mohapatra SS, Boyapalle S. Epidemiologic, experimental, and clinical links between respiratory syncytial virus infection and asthma. Clin Microbiol Rev. 2008;21(3):495–504.

    Article  PubMed  CAS  Google Scholar 

  74. Miller AL, Bowlin TL, Lukacs NW. Respiratory syncytial virus-induced chemokine production: linking viral replication to chemokine production in vitro and in vivo. J Infect Dis. 2004;189(8):1419–30.

    Article  PubMed  CAS  Google Scholar 

  75. Schwarze J, Hamelmann E, Bradley KL, Takeda K, Gelfand EW. Respiratory syncytial virus infection results in airway hyperresponsiveness and enhanced airway sensitization to allergen. J Clin Invest. 1997;100(1):226–33.

    Article  PubMed  CAS  Google Scholar 

  76. Han J, Takeda K, Gelfand EW. The role of RSV infection in asthma initiation and progression: findings in a mouse model. Pulm Med. 2011;2011:748038.

    PubMed  Google Scholar 

  77. Kristjansson S, Bjarnarson SP, Wennergren G, Palsdottir AH, Arnadottir T, Haraldsson A, Jonsdottir I. Respiratory syncytial virus and other respiratory viruses during the first 3 months of life promote a local TH2-like response. J Allergy Clin Immunol. 2005;116(4):805–11.

    Article  PubMed  CAS  Google Scholar 

  78. Qiao J, Li A, Jin X. TSLP from RSV-stimulated rat airway epithelial cells activates myeloid dendritic cells. Immunol Cell Biol. 2011;89(2):231–8.

    Article  PubMed  CAS  Google Scholar 

  79. Chang YJ, Kim HY, Albacker LA, Lee HH, Baumgarth N, Akira S, Savage PB, Endo S, Yamamura T, Maaskant J, Kitano N, Singh A, Bhatt A, Besra GS, van den Elzen P, Appelmelk B, Franck RW, Chen G, DeKruyff RH, Shimamura M, Illarionov P, Umetsu DT. Influenza infection in suckling mice expands an NKT cell subset that protects against airway hyperreactivity. J Clin Invest. 2011;121(1):57–69.

    Article  PubMed  CAS  Google Scholar 

  80. Dahl ME, Dabbagh K, Liggitt D, Kim S, Lewis DB. Viral-induced T helper type 1 responses enhance allergic disease by effects on lung dendritic cells. Nat Immunol. 2004;5(3):337–43.

    Article  PubMed  CAS  Google Scholar 

  81. Evans SM, Blyth DI, Wong T, Sanjar S, West MR. Decreased distribution of lung epithelial junction proteins after intratracheal antigen or lipopolysaccharide challenge: correlation with neutrophil influx and levels of BALF sE-cadherin. Am J Respir Cell Mol Biol. 2002;27(4):446–54.

    PubMed  CAS  Google Scholar 

  82. Tillie-Leblond I, Gosset P, Le Berre R, Janin A, Prangere T, Tonnel AB, Guery BP. Keratinocyte growth factor improves alterations of lung permeability and bronchial epithelium in allergic rats. Eur Respir J. 2007;30(1):31–9.

    Google Scholar 

  83. Holgate ST. Epithelium dysfunction in asthma. J Allergy Clin Immunol. 2007;120(6):1233–44; quiz 45–6.

    Google Scholar 

  84. Xiao C, Puddicombe SM, Field S, Haywood J, Broughton-Head V, Puxeddu I, Haitchi HM, Vernon-Wilson E, Sammut D, Bedke N, Cremin C, Sones J, Djukanovic R, Howarth PH, Collins JE, Holgate ST, Monk P, Davies DE. Defective epithelial barrier function in asthma. J Allergy Clin Immunol. 2011;128(3):549–56 e1–12.

    Google Scholar 

  85. Basuroy S, Seth A, Elias B, Naren AP, Rao R. MAPK interacts with occludin and mediates EGF-induced prevention of tight junction disruption by hydrogen peroxide. Biochem J. 2006;393(Pt 1):69–77.

    Google Scholar 

  86. Tsao PN, Wei SC, Wu MF, Huang MT, Lin HY, Lee MC, Lin KM, Wang IJ, Kaartinen V, Yang LT, Cardoso WV. Notch signaling prevents mucous metaplasia in mouse conducting airways during postnatal development. Development. 2011;138(16):3533–43.

    Article  PubMed  CAS  Google Scholar 

  87. Wan H, Kaestner KH, Ang SL, Ikegami M, Finkelman FD, Stahlman MT, Fulkerson PC, Rothenberg ME, Whitsett JA. Foxa2 regulates alveolarization and goblet cell hyperplasia. Development. 2004;131(4):953–64.

    Article  PubMed  CAS  Google Scholar 

  88. Chen G, Korfhagen TR, Xu Y, Kitzmiller J, Wert SE, Maeda Y, Gregorieff A, Clevers H, Whitsett JA. SPDEF is required for mouse pulmonary goblet cell differentiation and regulates a network of genes associated with mucus production. J Clin Invest. 2009;119(10):2914–24.

    PubMed  CAS  Google Scholar 

  89. Park KS, Korfhagen TR, Bruno MD, Kitzmiller JA, Wan H, Wert SE, Khurana Hershey GK, Chen G, Whitsett JA. SPDEF regulates goblet cell hyperplasia in the airway epithelium. J Clin Invest. 2007;117(4):978–88.

    Article  PubMed  CAS  Google Scholar 

  90. Chen G, Wan H, Luo F, Zhang L, Xu Y, Lewkowich I, Wills-Karp M, Whitsett JA. Foxa2 programs Th2 cell-mediated innate immunity in the developing lung. J Immunol. 2010;184(11):6133–41.

    Article  PubMed  CAS  Google Scholar 

  91. Park SW, Verhaeghe C, Nguyenvu LT, Barbeau R, Eisley CJ, Nakagami Y, Huang X, Woodruff PG, Fahy JV, Erle DJ. Distinct roles of FOXA2 and FOXA3 in allergic airway disease and asthma. Am J Respir Crit Care Med. 2009;180(7):603–10.

    Article  PubMed  CAS  Google Scholar 

  92. Zhen G, Park SW, Nguyenvu LT, Rodriguez MW, Barbeau R, Paquet AC, Erle DJ. IL-13 and epidermal growth factor receptor have critical but distinct roles in epithelial cell mucin production. Am J Respir Cell Mol Biol. 2007;36(2):244–53.

    Article  PubMed  CAS  Google Scholar 

  93. Maeda Y, Dave V, Whitsett JA. Transcriptional control of lung morphogenesis. Physiol Rev. 2007;87(1):219–44.

    Article  PubMed  CAS  Google Scholar 

  94. Bohinski RJ, Di Lauro R, Whitsett JA. The lung-specific surfactant protein B gene promoter is a target for thyroid transcription factor 1 and hepatocyte nuclear factor 3, indicating common factors for organ-specific gene expression along the foregut axis. Mol Cell Biol. 1994;14(9):5671–81.

    Article  PubMed  CAS  Google Scholar 

  95. DeFelice M, Silberschmidt D, DiLauro R, Xu Y, Wert SE, Weaver TE, Bachurski CJ, Clark JC, Whitsett JA. TTF-1 phosphorylation is required for peripheral lung morphogenesis, perinatal survival, and tissue-specific gene expression. J Biol Chem. 2003;278(37):35574–83.

    Article  PubMed  CAS  Google Scholar 

  96. Maeda Y, Chen G, Xu Y, Haitchi HM, Du L, Keiser AR, Howarth PH, Davies DE, Holgate ST, Whitsett JA. Airway epithelial transcription factor NK2 homeobox 1 inhibits mucous cell metaplasia and Th2 inflammation. Am J Respir Crit Care Med. 2011;184(4):421–9.

    Article  PubMed  CAS  Google Scholar 

  97. Rock JR, Randell SH, Hogan BL. Airway basal stem cells: a perspective on their roles in epithelial homeostasis and remodeling. Dis Model Mech. 2010;3(9-10):545–56.

    Google Scholar 

  98. Whitsett JA, Haitchi HM, Maeda Y. Intersections between pulmonary development and disease. Am J Respir Crit Care Med. 2011;184(4):401–6.

    Article  PubMed  CAS  Google Scholar 

  99. Patel AC, Brody SL, Stappenbeck TS, Holtzman MJ. Tracking cell lineage to rediscover (again) the switch from ciliated to mucous cells. Am J Respir Cell Mol Biol. 2011;44(3):261–3.

    Article  PubMed  CAS  Google Scholar 

  100. Turner J, Roger J, Fitau J, Combe D, Giddings J, Heeke GV, Jones CE. Goblet cells are derived from a FOXJ1-expressing progenitor in a human airway epithelium. Am J Respir Cell Mol Biol. 2011;44(3):276–84.

    Article  PubMed  CAS  Google Scholar 

  101. Kondo M, Tamaoki J, Takeyama K, Isono K, Kawatani K, Izumo T, Nagai A. Elimination of IL-13 reverses established goblet cell metaplasia into ciliated epithelia in airway epithelial cell culture. Allergol Int. 2006;55(3):329–36.

    Article  PubMed  CAS  Google Scholar 

  102. Rankin JA, Picarella DE, Geba GP, Temann UA, Prasad B, DiCosmo B, Tarallo A, Stripp B, Whitsett J, Flavell RA. Phenotypic and physiologic characterization of transgenic mice expressing interleukin 4 in the lung: lymphocytic and eosinophilic inflammation without airway hyperreactivity. Proc Natl Acad Sci USA 1996;93(15):7821–5.

    Google Scholar 

  103. Zhu Z, Homer RJ, Wang Z, Chen Q, Geba GP, Wang J, Zhang Y, Elias JA. Pulmonary expression of interleukin-13 causes inflammation, mucus hypersecretion, subepithelial fibrosis, physiologic abnormalities, and eotaxin production. J Clin Invest. 1999;103(6):779–88.

    Article  PubMed  CAS  Google Scholar 

  104. White SR, Martin LD, Stern R, Laxman B, Marroquin BA. Expression of IL-4/IL-13 receptors in differentiating human airway epithelial cells. Am J Physiol Lung Cell Mol Physiol. 2010;299(5):L681–93.

    Article  PubMed  CAS  Google Scholar 

  105. Zhou Y, McLane M, Levitt RC. Th2 cytokines and asthma. Interleukin-9 as a therapeutic target for asthma. Respir Res. 2001;2(2):80–4.

    Article  PubMed  CAS  Google Scholar 

  106. Townsend JM, Fallon GP, Matthews JD, Smith P, Jolin EH, McKenzie NA. IL-9-deficient mice establish fundamental roles for IL-9 in pulmonary mastocytosis and goblet cell hyperplasia but not T cell development. Immunity. 2000;13(4):573–83.

    Article  PubMed  CAS  Google Scholar 

  107. Vermeer PD, Harson R, Einwalter LA, Moninger T, Zabner J. Interleukin-9 induces goblet cell hyperplasia during repair of human airway epithelia. Am J Respir Cell Mol Biol. 2003;28(3):286–95.

    Article  PubMed  CAS  Google Scholar 

  108. Kearley J, Erjefalt JS, Andersson C, Benjamin E, Jones CP, Robichaud A, Pegorier S, Brewah Y, Burwell TJ, Bjermer L, Kiener PA, Kolbeck R, Lloyd CM, Coyle AJ, Humbles AA. IL-9 governs allergen-induced mast cell numbers in the lung and chronic remodeling of the airways. Am J Respir Crit Care Med. 2011;183(7):865–75.

    Article  PubMed  CAS  Google Scholar 

  109. Song KS, Lee WJ, Chung KC, Koo JS, Yang EJ, Choi JY, Yoon JH. Interleukin-1 beta and tumor necrosis factor-alpha induce MUC5AC overexpression through a mechanism involving ERK/p38 mitogen-activated protein kinases-MSK1-CREB activation in human airway epithelial cells. J Biol Chem. 2003;278(26):23243–50.

    Article  PubMed  CAS  Google Scholar 

  110. Bry K, Whitsett JA, Lappalainen U. IL-1beta disrupts postnatal lung morphogenesis in the mouse. Am J Respir Cell Mol Biol. 2007;36(1):32–42.

    Article  PubMed  CAS  Google Scholar 

  111. Dinarello CA. Biologic basis for interleukin-1 in disease. Blood. 1996;87(6):2095–147.

    PubMed  CAS  Google Scholar 

  112. Kondo Y, Yoshimoto T, Yasuda K, Futatsugi-Yumikura S, Morimoto M, Hayashi N, Hoshino T, Fujimoto J, Nakanishi K. Administration of IL-33 induces airway hyperresponsiveness and goblet cell hyperplasia in the lungs in the absence of adaptive immune system. Int Immunol. 2008;20(6):791–800.

    Article  PubMed  CAS  Google Scholar 

  113. Sakashita M, Yoshimoto T, Hirota T, Harada M, Okubo K, Osawa Y, Fujieda S, Nakamura Y, Yasuda K, Nakanishi K, Tamari M. Association of serum interleukin-33 level and the interleukin-33 genetic variant with Japanese cedar pollinosis. Clin Exp Allergy. 2008;38(12):1875–81.

    Article  PubMed  CAS  Google Scholar 

  114. Ishikawa Y, Yoshimoto T, Nakanishi K. Contribution of IL-18-induced innate T cell activation to airway inflammation with mucus hypersecretion and airway hyperresponsiveness. Int Immunol. 2006;18(6):847–55.

    Article  PubMed  CAS  Google Scholar 

  115. Ma Y, Zhang B, Tang RK, Liu Y, Peng GG. Interleukin-18 promoter polymorphism and asthma risk: a meta-analysis. Mol Biol Rep. 2012;39(2):1371–6.

    Google Scholar 

  116. Harada M, Obara K, Hirota T, Yoshimoto T, Hitomi Y, Sakashita M, Doi S, Miyatake A, Fujita K, Enomoto T, Taniguchi M, Higashi N, Fukutomi Y, Nakanishi K, Nakamura Y, Tamari M. A functional polymorphism in IL-18 is associated with severity of bronchial asthma. Am J Respir Crit Care Med. 2009;180(11):1048–55.

    Google Scholar 

  117. Kuperman DA, Huang X, Koth LL, Chang GH, Dolganov GM, Zhu Z, Elias JA, Sheppard D, Erle DJ. Direct effects of interleukin-13 on epithelial cells cause airway hyperreactivity and mucus overproduction in asthma. Nat Med. 2002;8(8):885–9.

    PubMed  CAS  Google Scholar 

  118. Kuperman D, Schofield B, Wills-Karp M, Grusby MJ. Signal transducer and activator of transcription factor 6 (Stat6)-deficient mice are protected from antigen-induced airway hyperresponsiveness and mucus production. J Exp Med. 1998;187(6):939–48.

    Article  PubMed  CAS  Google Scholar 

  119. Yu H, Li Q, Kolosov VP, Perelman JM, Zhou X. Interleukin-13 induces mucin 5AC production involving STAT6/SPDEF in human airway epithelial cells. Cell Commun Adhes. 2010;17(4–6):83–92.

    Article  PubMed  CAS  Google Scholar 

  120. Mattes J, Yang M, Siqueira A, Clark K, MacKenzie J, McKenzie AN, Webb DC, Matthaei KI, Foster PS. IL-13 induces airways hyperreactivity independently of the IL-4R alpha chain in the allergic lung. J Immunol. 2001;167(3):1683–92.

    PubMed  CAS  Google Scholar 

  121. Lee PJ, Zhang X, Shan P, Ma B, Lee CG, Homer RJ, Zhu Z, Rincon M, Mossman BT, Elias JA. ERK1/2 mitogen-activated protein kinase selectively mediates IL-13-induced lung inflammation and remodeling in vivo. J Clin Invest. 2006;116(1):163–73.

    Article  PubMed  CAS  Google Scholar 

  122. Blease K, Schuh JM, Jakubzick C, Lukacs NW, Kunkel SL, Joshi BH, Puri RK, Kaplan MH, Hogaboam CM. Stat6-deficient mice develop airway hyperresponsiveness and peribronchial fibrosis during chronic fungal asthma. Am J Pathol. 2002;160(2):481–90.

    Article  PubMed  CAS  Google Scholar 

  123. Holgate ST, Lackie PM, Davies DE, Roche WR, Walls AF. The bronchial epithelium as a key regulator of airway inflammation and remodelling in asthma. Clin Exp Allergy. 1999;29(Suppl 2):90–5.

    Article  PubMed  Google Scholar 

  124. Puddicombe SM, Polosa R, Richter A, Krishna MT, Howarth PH, Holgate ST, Davies DE. Involvement of the epidermal growth factor receptor in epithelial repair in asthma. FASEB J. 2000;14(10):1362–74.

    Google Scholar 

  125. Le Cras TD, Acciani TH, Mushaben EM, Kramer EL, Pastura PA, Hardie WD, Korfhagen TR, Sivaprasad U, Ericksen M, Gibson AM, Holtzman MJ, Whitsett JA, Hershey GK. Epithelial EGF receptor signaling mediates airway hyperreactivity and remodeling in a mouse model of chronic asthma. Am J Physiol Lung Cell Mol Physiol. 2011;300(3):L414–21.

    Article  PubMed  CAS  Google Scholar 

  126. Hart LA, Krishnan VL, Adcock IM, Barnes PJ, Chung KF. Activation and localization of transcription factor, nuclear factor-kappaB, in asthma. Am J Respir Crit Care Med. 1998;158(5 Pt 1):1585–92.

    PubMed  CAS  Google Scholar 

  127. Barnes PJ. Pathophysiology of asthma. Br J Clin Pharmacol. 1996;42(1):3–10.

    Article  PubMed  CAS  Google Scholar 

  128. Nissen RM, Yamamoto KR. The glucocorticoid receptor inhibits NFkappaB by interfering with serine-2 phosphorylation of the RNA polymerase II carboxy-terminal domain. Genes Dev. 2000;14(18):2314–29.

    Article  PubMed  CAS  Google Scholar 

  129. Cheng DS, Han W, Chen SM, Sherrill TP, Chont M, Park GY, Sheller JR, Polosukhin VV, Christman JW, Yull FE, Blackwell TS. Airway epithelium controls lung inflammation and injury through the NF-kappa B pathway. J Immunol. 2007;178(10):6504–13.

    PubMed  CAS  Google Scholar 

  130. Pantano C, Ather JL, Alcorn JF, Poynter ME, Brown AL, Guala AS, Beuschel SL, Allen GB, Whittaker LA, Bevelander M, Irvin CG, Janssen-Heininger YM. Nuclear factor-kappaB activation in airway epithelium induces inflammation and hyperresponsiveness. Am J Respir Crit Care Med. 2008;177(9):959–69.

    Article  PubMed  CAS  Google Scholar 

  131. Broide DH, Lawrence T, Doherty T, Cho JY, Miller M, McElwain K, McElwain S, Karin M. Allergen-induced peribronchial fibrosis and mucus production mediated by IkappaB kinase beta-dependent genes in airway epithelium. Proc Natl Acad Sci USA 2005;102(49):17723–8.

    Google Scholar 

  132. Chapoval SP, Al-Garawi A, Lora JM, Strickland I, Ma B, Lee PJ, Homer RJ, Ghosh S, Coyle AJ, Elias JA. Inhibition of NF-kappaB activation reduces the tissue effects of transgenic IL-13. J Immunol. 2007;179(10):7030–41.

    PubMed  CAS  Google Scholar 

  133. Ogawa H, Azuma M, Muto S, Nishioka Y, Honjo A, Tezuka T, Uehara H, Izumi K, Itai A, Sone S. IkappaB kinase beta inhibitor IMD-0354 suppresses airway remodelling in a Dermatophagoides pteronyssinus-sensitized mouse model of chronic asthma. Clin Exp Allergy. 2011;41(1):104–15.

    Article  PubMed  CAS  Google Scholar 

  134. Thuault S, Tan EJ, Peinado H, Cano A, Heldin CH, Moustakas A. HMGA2 and Smads co-regulate SNAIL1 expression during induction of epithelial-to-mesenchymal transition. J Biol Chem. 2008;283(48):33437–46.

    Article  PubMed  CAS  Google Scholar 

  135. Logan CY, Nusse R. The Wnt signaling pathway in development and disease. Annu Rev Cell Dev Biol. 2004;20:781–810.

    Article  PubMed  CAS  Google Scholar 

  136. Giangreco A, Lu L, Vickers C, Teixeira VH, Groot KR, Ilieva EV, George J, Nicholson AG, Sage EK, Watt FM, Janes SM. beta- catenin determines upper airway progenitor cell fate and pre-invasive squamous lung cancer progression by modulating epithelial-to-mesenchymal transition. J Pathol. 2012;226(4):575–87.

    Google Scholar 

  137. Yook JI, Li XY, Ota I, Fearon ER, Weiss SJ. Wnt-dependent regulation of the E-cadherin repressor snail. J Biol Chem. 2005;280(12):11740–8.

    Article  PubMed  CAS  Google Scholar 

  138. 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(1):e16175.

    Article  PubMed  CAS  Google Scholar 

  139. Hackett TL, Warner SM, Stefanowicz D, Shaheen F, Pechkovsky DV, Murray LA, Argentieri R, Kicic A, Stick SM, Bai TR, Knight DA. Induction of epithelial-mesenchymal transition in primary airway epithelial cells from patients with asthma by transforming growth factor-beta1. Am J Respir Crit Care Med. 2009;180(2):122–33.

    Article  PubMed  CAS  Google Scholar 

  140. Doerner AM, Zuraw BL. TGF-beta1 induced epithelial to mesenchymal transition (EMT) in human bronchial epithelial cells is enhanced by IL-1beta but not abrogated by corticosteroids. Respir Res. 2009;10:100.

    Article  PubMed  CAS  Google Scholar 

  141. Perng DW, Chang KT, Su KC, Wu YC, Chen CS, Hsu WH, Tsai CM, Lee YC. Matrix metalloprotease-9 induces transforming growth factor-beta(1) production in airway epithelium via activation of epidermal growth factor receptors. Life Sci. 2011;89(5–6):204–12.

    Article  PubMed  CAS  Google Scholar 

  142. Vermeer PD, Denker J, Estin M, Moninger TO, Keshavjee S, Karp P, Kline JN, Zabner J. MMP9 modulates tight junction integrity and cell viability in human airway epithelia. Am J Physiol Lung Cell Mol Physiol. 2009;296(5):L751–62.

    Article  PubMed  CAS  Google Scholar 

  143. Davey A, McAuley DF, O’Kane CM. Matrix metalloproteinases in acute lung injury: mediators of injury and drivers of repair. Eur Respir J. 2011;38(4):959–70.

    Google Scholar 

  144. Wilson JW, Li X. The measurement of reticular basement membrane and submucosal collagen in the asthmatic airway. Clin Exp Allergy. 1997;27(4):363–71.

    Article  PubMed  CAS  Google Scholar 

  145. Mauad T, Xavier AC, Saldiva PH, Dolhnikoff M. Elastosis and fragmentation of fibers of the elastic system in fatal asthma. Am J Respir Crit Care Med. 1999;160(3):968–75.

    PubMed  CAS  Google Scholar 

  146. Mauad T, Silva LF, Santos MA, Grinberg L, Bernardi FD, Martins MA, Saldiva PH, Dolhnikoff M. Abnormal alveolar attachments with decreased elastic fiber content in distal lung in fatal asthma. Am J Respir Crit Care Med. 2004;170(8):857–62.

    Article  PubMed  Google Scholar 

  147. Mukhopadhyay S, Sypek J, Tavendale R, Gartner U, Winter J, Li W, Page K, Fleming M, Brady J, O’Toole M, Macgregor DF, Goldman S, Tam S, Abraham W, Williams C, Miller DK, Palmer CN. Matrix metalloproteinase-12 is a therapeutic target for asthma in children and young adults. J Allergy Clin Immunol. 2010;126(1):70–6 e16.

    Google Scholar 

  148. Gounni AS. The high-affinity IgE receptor (FcepsilonRI): a critical regulator of airway smooth muscle cells? Am J Physiol Lung Cell Mol Physiol. 2006;291(3):L312–21.

    Google Scholar 

  149. Bara I, Ozier A, Tunon de Lara JM, Marthan R, Berger P. Pathophysiology of bronchial smooth muscle remodelling in asthma. Eur Respir J. 2010;36(5):1174–84.

    Google Scholar 

  150. Koziol-White CJ, Panettieri RA Jr. Airway smooth muscle and immunomodulation in acute exacerbations of airway disease. Immunol Rev. 2011;242(1):178–85.

    Article  PubMed  CAS  Google Scholar 

  151. Tliba O, Panettieri RA Jr. Noncontractile functions of airway smooth muscle cells in asthma. Annu Rev Physiol. 2009;71:509–35.

    Article  PubMed  CAS  Google Scholar 

  152. Castro M, Rubin A, Laviolette M, Hanania NA, Armstrong B, Cox G. Persistence of effectiveness of bronchial thermoplasty in patients with severe asthma. Ann Allergy Asthma Immunol. 2011;107(1):65–70.

    Article  PubMed  CAS  Google Scholar 

  153. Cox G, Thomson NC, Rubin AS, Niven RM, Corris PA, Siersted HC, Olivenstein R, Pavord ID, McCormack D, Chaudhuri R, Miller JD, Laviolette M. Asthma control during the year after bronchial thermoplasty. N Engl J Med. 2007;356(13):1327–37.

    Article  PubMed  CAS  Google Scholar 

  154. Thomson NC, Rubin AS, Niven RM, Corris PA, Siersted HC, Olivenstein R, Pavord ID, McCormack D, Laviolette M, Shargill NS, Cox G. Long-term (5 year) safety of bronchial thermoplasty: Asthma Intervention Research (AIR) trial. BMC Pulm Med. 2011;11:8.

    Article  PubMed  Google Scholar 

  155. Halayko AJ, Tran T, Ji SY, Yamasaki A, Gosens R. Airway smooth muscle phenotype and function: interactions with current asthma therapies. Curr Drug Targets. 2006;7(5):525–40.

    Article  PubMed  CAS  Google Scholar 

  156. Begueret H, Berger P, Vernejoux JM, Dubuisson L, Marthan R, Tunon-de-Lara JM. Inflammation of bronchial smooth muscle in allergic asthma. Thorax. 2007;62(1):8–15.

    Article  PubMed  CAS  Google Scholar 

  157. Nguyen TT, Ward JP, Hirst SJ. beta1-Integrins mediate enhancement of airway smooth muscle proliferation by collagen and fibronectin. Am J Respir Crit Care Med. 2005;171(3):217–23.

    Article  PubMed  Google Scholar 

  158. Dekkers BG, Schaafsma D, Nelemans SA, Zaagsma J, Meurs H. Extracellular matrix proteins differentially regulate airway smooth muscle phenotype and function. Am J Physiol Lung Cell Mol Physiol. 2007;292(6):L1405–13.

    Article  PubMed  CAS  Google Scholar 

  159. Hirst SJ, Twort CH, Lee TH. Differential effects of extracellular matrix proteins on human airway smooth muscle cell proliferation and phenotype. Am J Respir Cell Mol Biol. 2000;23(3):335–44.

    PubMed  CAS  Google Scholar 

  160. Freyer AM, Johnson SR, Hall IP. Effects of growth factors and extracellular matrix on survival of human airway smooth muscle cells. Am J Respir Cell Mol Biol. 2001;25(5):569–76.

    PubMed  CAS  Google Scholar 

  161. Chan V, Burgess JK, Ratoff JC, O’Connor BJ, Greenough A, Lee TH, Hirst SJ. Extracellular matrix regulates enhanced eotaxin expression in asthmatic airway smooth muscle cells. Am J Respir Crit Care Med. 2006;174(4):379–85.

    Article  PubMed  CAS  Google Scholar 

  162. Dekkers BG, Maarsingh H, Meurs H, Gosens R. Airway structural components drive airway smooth muscle remodeling in asthma. Proc Am Thorac Soc. 2009;6(8):683–92.

    Article  PubMed  CAS  Google Scholar 

  163. Peng Q, Lai D, Nguyen TT, Chan V, Matsuda T, Hirst SJ. Multiple beta 1 integrins mediate enhancement of human airway smooth muscle cytokine secretion by fibronectin and type I collagen. J Immunol. 2005;174(4):2258–64.

    PubMed  CAS  Google Scholar 

  164. Johnson PR, Burgess JK, Ge Q, Poniris M, Boustany S, Twigg SM, Black JL. Connective tissue growth factor induces extracellular matrix in asthmatic airway smooth muscle. Am J Respir Crit Care Med. 2006;173(1):32–41.

    Article  PubMed  CAS  Google Scholar 

  165. Benayoun L, Druilhe A, Dombret MC, Aubier M, Pretolani M. Airway structural alterations selectively associated with severe asthma. Am J Respir Crit Care Med. 2003;167(10):1360–8.

    Article  PubMed  Google Scholar 

  166. Regamey N, Ochs M, Hilliard TN, Muhlfeld C, Cornish N, Fleming L, Saglani S, Alton EW, Bush A, Jeffery PK, Davies JC. Increased airway smooth muscle mass in children with asthma, cystic fibrosis, and non-cystic fibrosis bronchiectasis. Am J Respir Crit Care Med. 2008;177(8):837–43.

    Article  PubMed  Google Scholar 

  167. Woodruff PG, Dolganov GM, Ferrando RE, Donnelly S, Hays SR, Solberg OD, Carter R, Wong HH, Cadbury PS, Fahy JV. Hyperplasia of smooth muscle in mild to moderate asthma without changes in cell size or gene expression. Am J Respir Crit Care Med. 2004;169(9):1001–6.

    Article  PubMed  Google Scholar 

  168. Johnson PR, Roth M, Tamm M, Hughes M, Ge Q, King G, Burgess JK, Black JL. Airway smooth muscle cell proliferation is increased in asthma. Am J Respir Crit Care Med. 2001;164(3):474–7.

    PubMed  CAS  Google Scholar 

  169. Fairbank NJ, Connolly SC, Mackinnon JD, Wehry K, Deng L, Maksym GN. Airway smooth muscle cell tone amplifies contractile function in the presence of chronic cyclic strain. Am J Physiol Lung Cell Mol Physiol. 2008;295(3):L479–88.

    Article  PubMed  CAS  Google Scholar 

  170. Hasaneen NA, Zucker S, Cao J, Chiarelli C, Panettieri RA, Foda HD. Cyclic mechanical strain-induced proliferation and migration of human airway smooth muscle cells: role of EMMPRIN and MMPs. FASEB J. 2005;19(11):1507–9.

    Google Scholar 

  171. Bosse Y, Stankova J, Rola-Pleszczynski M. Transforming growth factor-beta1 in asthmatic airway smooth muscle enlargement: is fibroblast growth factor-2 required? Clin Exp Allergy 2010;40(5):710–24.

    Google Scholar 

  172. Enomoto Y, Orihara K, Takamasu T, Matsuda A, Gon Y, Saito H, Ra C, Okayama Y. Tissue remodeling induced by hypersecreted epidermal growth factor and amphiregulin in the airway after an acute asthma attack. J Allergy Clin Immunol. 2009;124(5):913–20 e1–7.

    Google Scholar 

  173. Aravamudan B, Thompson M, Pabelick C, Prakash YS. Brain derived neurotrophic factor induces proliferation of human airway smooth muscle cells. J Cell Mol Med. 2011. doi:10.1111/j.1582-4934.2011.01356.x.

  174. Walker TR, Moore SM, Lawson MF, Panettieri RA Jr, Chilvers ER. Platelet-derived growth factor-BB and thrombin activate phosphoinositide 3-kinase and protein kinase B: role in mediating airway smooth muscle proliferation. Mol Pharmacol. 1998;54(6):1007–15.

    PubMed  CAS  Google Scholar 

  175. Johnson JR, Swirski FK, Gajewska BU, Wiley RE, Fattouh R, Pacitto SR, Wong JK, Stampfli MR, Jordana M. Divergent immune responses to house dust mite lead to distinct structural-functional phenotypes. Am J Physiol Lung Cell Mol Physiol. 2007;293(3):L730–9.

    Article  PubMed  CAS  Google Scholar 

  176. Cho JY, Miller M, Baek KJ, Han JW, Nayar J, Lee SY, McElwain K, McElwain S, Friedman S, Broide DH. Inhibition of airway remodeling in IL-5-deficient mice. J Clin Invest. 2004;113(4):551–60.

    PubMed  CAS  Google Scholar 

  177. Doherty TA, Soroosh P, Khorram N, Fukuyama S, Rosenthal P, Cho JY, Norris PS, Choi H, Scheu S, Pfeffer K, Zuraw BL, Ware CF, Broide DH, Croft M. The tumor necrosis factor family member LIGHT is a target for asthmatic airway remodeling. Nat Med. 2011;17(5):596–603.

    Article  PubMed  CAS  Google Scholar 

  178. Hastie AT, Moore WC, Meyers DA, Vestal PL, Li H, Peters SP, Bleecker ER. Analyses of asthma severity phenotypes and inflammatory proteins in subjects stratified by sputum granulocytes. J Allergy Clin Immunol. 2010;125(5):1028–36 e13.

    Google Scholar 

  179. Bradding P. Mast cell regulation of airway smooth muscle function in asthma. Eur Respir J. 2007;29(5):827–30.

    Google Scholar 

  180. Brightling CE, Bradding P, Symon FA, Holgate ST, Wardlaw AJ, Pavord ID. Mast-cell infiltration of airway smooth muscle in asthma. N Engl J Med. 2002;346(22):1699–705.

    Article  PubMed  Google Scholar 

  181. Sutcliffe A, Kaur D, Page S, Woodman L, Armour CL, Baraket M, Bradding P, Hughes JM, Brightling CE. Mast cell migration to Th2 stimulated airway smooth muscle from asthmatics. Thorax. 2006;61(8):657–62.

    Article  PubMed  CAS  Google Scholar 

  182. Hollins F, Kaur D, Yang W, Cruse G, Saunders R, Sutcliffe A, Berger P, Ito A, Brightling CE, Bradding P. Human airway smooth muscle promotes human lung mast cell survival, proliferation, and constitutive activation: cooperative roles for CADM1, stem cell factor, and IL-6. J Immunol. 2008;181(4):2772–80.

    PubMed  CAS  Google Scholar 

  183. Galli SJ, Tsai M. Mast cells in allergy and infection: versatile effector and regulatory cells in innate and adaptive immunity. Eur J Immunol. 2010;40(7):1843–51.

    Article  PubMed  CAS  Google Scholar 

  184. Chhabra J, Li YZ, Alkhouri H, Blake AE, Ge Q, Armour CL, Hughes JM. Histamine and tryptase modulate asthmatic airway smooth muscle GM-CSF and RANTES release. Eur Respir J. 2007;29(5):861–70.

    Google Scholar 

  185. Gordon JR, Galli SJ. Release of both preformed and newly synthesized tumor necrosis factor alpha (TNF-alpha)/cachectin by mouse mast cells stimulated via the Fc epsilon RI. A mechanism for the sustained action of mast cell-derived TNF-alpha during IgE-dependent biological responses. J Exp Med. 1991;174(1):103–7.

    Article  PubMed  CAS  Google Scholar 

  186. Burgess JK, Blake AE, Boustany S, Johnson PR, Armour CL, Black JL, Hunt NH, Hughes JM. CD40 and OX40 ligand are increased on stimulated asthmatic airway smooth muscle. J Allergy Clin Immunol. 2005;115(2):302–8.

    Article  PubMed  CAS  Google Scholar 

  187. Croft M, So T, Duan W, Soroosh P. The significance of OX40 and OX40L to T-cell biology and immune disease. Immunol Rev. 2009;229(1):173–91.

    Article  PubMed  CAS  Google Scholar 

  188. So T, Croft M. Cutting edge: OX40 inhibits TGF-beta- and antigen-driven conversion of naive CD4 T cells into CD25+ Foxp3+ T cells. J Immunol. 2007;179(3):1427–30.

    PubMed  CAS  Google Scholar 

  189. Vu MD, Xiao X, Gao W, Degauque N, Chen M, Kroemer A, Killeen N, Ishii N, Chang Li X. OX40 costimulation turns off Foxp3+ Tregs. Blood. 2007;110(7):2501–10.

    Article  PubMed  CAS  Google Scholar 

  190. Tregoning JS, Schwarze J. Respiratory viral infections in infants: causes, clinical symptoms, virology, and immunology. Clin Microbiol Rev. 2010;23(1):74–98.

    Article  PubMed  CAS  Google Scholar 

  191. Papadopoulos NG, Xepapadaki P, Mallia P, Brusselle G, Watelet JB, Xatzipsalti M, Foteinos G, van Drunen CM, Fokkens WJ, D’Ambrosio C, Bonini S, Bossios A, Lotvall J, van Cauwenberge P, Holgate ST, Canonica GW, Szczeklik A, Rohde G, Kimpen J, Pitkaranta A, Makela M, Chanez P, Ring J, Johnston SL. Mechanisms of virus-induced asthma exacerbations: state-of-the-art. A GA2LEN and InterAirways document. Allergy. 2007;62(5):457–70.

    Article  PubMed  CAS  Google Scholar 

  192. Busse WW, Lemanske RF Jr, Gern JE. Role of viral respiratory infections in asthma and asthma exacerbations. Lancet. 2010;376(9743):826–34.

    Article  PubMed  Google Scholar 

  193. Folkerts G, Busse WW, Nijkamp FP, Sorkness R, Gern JE. Virus-induced airway hyperresponsiveness and asthma. Am J Respir Crit Care Med. 1998;157(6 Pt 1):1708–20.

    PubMed  CAS  Google Scholar 

  194. Monick MM, Cameron K, Staber J, Powers LS, Yarovinsky TO, Koland JG, Hunninghake GW. Activation of the epidermal growth factor receptor by respiratory syncytial virus results in increased inflammation and delayed apoptosis. J Biol Chem. 2005;280(3):2147–58.

    Article  PubMed  CAS  Google Scholar 

  195. Monick MM, Powers LS, Hassan I, Groskreutz D, Yarovinsky TO, Barrett CW, Castilow EM, Tifrea D, Varga SM, Hunninghake GW. Respiratory syncytial virus synergizes with Th2 cytokines to induce optimal levels of TARC/CCL17. J Immunol. 2007;179(3):1648–58.

    PubMed  CAS  Google Scholar 

  196. Erb-Downward JR, Thompson DL, Han MK, Freeman CM, McCloskey L, Schmidt LA, Young VB, Toews GB, Curtis JL, Sundaram B, Martinez FJ, Huffnagle GB. Analysis of the lung microbiome in the “healthy” smoker and in COPD. PLoS ONE. 2011;6(2):e16384.

    Article  PubMed  CAS  Google Scholar 

  197. Hilty M, Burke C, Pedro H, Cardenas P, Bush A, Bossley C, Davies J, Ervine A, Poulter L, Pachter L, Moffatt MF, Cookson WO. Disordered microbial communities in asthmatic airways. PLoS ONE. 2010;5(1):e8578.

    Article  PubMed  CAS  Google Scholar 

  198. Huang YJ, Nelson CE, Brodie EL, Desantis TZ, Baek MS, Liu J, Woyke T, Allgaier M, Bristow J, Wiener-Kronish JP, Sutherland ER, King TS, Icitovic N, Martin RJ, Calhoun WJ, Castro M, Denlinger LC, Dimango E, Kraft M, Peters SP, Wasserman SI, Wechsler ME, Boushey HA, Lynch SV. Airway microbiota and bronchial hyperresponsiveness in patients with suboptimally controlled asthma. J Allergy Clin Immunol. 2011;127(2):372–81 e1–3.

    Google Scholar 

  199. Ichinohe T, Pang IK, Kumamoto Y, Peaper DR, Ho JH, Murray TS, Iwasaki A. Microbiota regulates immune defense against respiratory tract influenza A virus infection. Proc Natl Acad Sci USA 2011;108(13):5354–9.

    Google Scholar 

Download references

Acknowledgments

MACL laboratory is funded by the Agency for Science, Technology and Research, Singapore.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Maria Alicia Curotto de Lafaille.

Additional information

Lakshmi Ramakrishna and Victor Christoff de Vries contributed equally.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Ramakrishna, L., de Vries, V.C. & Curotto de Lafaille, M.A. Cross-roads in the lung: immune cells and tissue interactions as determinants of allergic asthma. Immunol Res 53, 213–228 (2012). https://doi.org/10.1007/s12026-012-8296-4

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12026-012-8296-4

Keywords

Navigation