A mouse model of rhinovirus induced asthma exacerbation 5

31 253 0
A mouse model of rhinovirus induced asthma exacerbation 5

Đang tải... (xem toàn văn)

Tài liệu hạn chế xem trước, để xem đầy đủ mời bạn chọn Tải xuống

Thông tin tài liệu

airway and blood cells, TLR3, TLR4 and TLRs7-9 induced interferons are not impaired in well-controlled asthma (Sykes et al., 2013b). Meanwhile, little clinical evidence for deficient IFN responses was reported. Thus, additional studies are needed to resolve the problem. Recently, the hypothesis that Th1 response is not necessary for allergic response initiation is been challenged. Synergic transferring Th1 and Th2 cells to allergen-challenged mice leads to a robust eosinophilic inflammation, with more severe symptoms than Th1 or Th2 cells alone (Randolph et al., 1999). TLR3 were also found to augment the allergic response in asthma pathology (Reuter et al., 2012). Study on RSV-infected mouse model showed that virus-induced type-I IFN infection drives that expression of high-affinity IgE receptor (FcεRI) on cDCs. Activation of FcεRI induced the recruitment of CD4+ T cells that produce IL-13 (Grayson et al., 2007). Besides respiratory virus, other Th1 response stimuli including bacteria infection enhanced Th2 response and atopy symptoms were also reported (Castro et al., 2000). CXCL10, also named IFN-γ induced protein 10 (IP-10), is a Th1 type inflammatory mediator, and increases airway hyper- responsiveness, IL-4-secreting T cells and eosinophilia in allergic mouse model (Medoff et al., 2002). Besides the adaptive immune response, the role of innate immune response components in asthma exacerbation is becoming a new research interest. Virus infection in allergen-challenged mice   83   induced increased expression of eotaxin-1, IL-4 and IL-13 in macrophages ex vivo (Nagarkar et al., 2010). Exacerbated airway hyper-responsiveness and inflammation were associated with increased macrophage and its chemoattractant MCP-1 in experimental infected mice model (Schneider et al., 2013). Though the classically activated macrophage (M1) are considered as an inhibitor for the virus infection and secrete molecules that dampen the Th2 inflammation, evidence have shown that M1 are also contribute to asthma exacerbation induced by respiratory virus (Moreira and Hogaboam, 2011). Animal experiment also showed that iNKT (invariant natural killer T) cells involved in the virus-associated asthma exacerbation, which stimulate macrophages to secrete IL-13 after RSV infection (Holtzman et al., 2009). Meanwhile, several experiments indicate that Th1 components may cooperate with innate immune mediators to worsen the allergic inflammatory symptoms in asthma exacerbation. Interaction between IFN-γ and pulmonary macrophages resulted into more severe airway hyper-responsiveness (Kumar et al., 2012). In vitro co-culture of human monocytic and bronchial epithelial cells leads to the increase of RV-induced MCP-1 and CXCL10 (Korpi-Steiner et al., 2010). In our mouse model, rhinovirus infection enlarged the Th2 response in asthmatic mice, with significantly increased expression of IL-4 and IL-13 in lung. The virus-increased IgE increase is also the evidence of augmented Th2 response. Unfortunately, due to the lack of   84   enough sample and technical errors during experiments, we were unable to obtain Th1 response related data such as the expression of interferon cytokines. Though IgG are considered playing a role in the pathology of asthma by some researchers, experimental allergen challenge on asthmatic and healthy people showed that all four kinds of IgG respond differently when challenged with different allergens, and except IgG4, the other three kinds of whole body allergen-IgG showed no difference between atopic or non-atopic subjects (Hong et al., 1994). Considering the running out of samples and its importance in asthma, we were unable to measure the IgG levels in mouse model. Meanwhile, RV infection induced a significantly increased CXCL10 expression, suggesting that Th1 response may contribute to the mechanism of rhinovirus-induced asthma exacerbation. Besides that, the same increased pattern of MCP-1 and CXCL10 expression may suggest that the involvement of both Th1 and innate immune response in rhinovirus-induced asthma exacerbation. In conclusion, rhinovirus infection increased the Th2 immune response in allergic mouse model, and Th1 and innate immune response may also contribute to the exacerbation. More effort should been made to explore the detail mechanism. 5.4 Further direction and limitations As described in Chapter 1, the “hygiene hypothesis” suggests that a lacking of childhood exposure to infection might increase the   85   susceptibility of certain inflammatory disorders. Thus, a virus infection during early life should prevent the development of asthma. However, in addition to its role as a trigger for asthma exacerbation, viral infections were also considered as the predisposition of development of post-viral asthma and allergic disease. Studies in human showed that children after RSV infection were at an increased risk for developing asthma and allergic sensitization (Sigurs et al., 2000). Rhinovirus infections were also reported to induce a higher risk of childhood asthma than other viruses (Kotaniemi-Syrjänen et al., 2003). Other investigations indicated a relationship between IgE production and respiratory virus infections, with the reported increase in IgE levels in allergen-challenged subjects or atopic subjects (Park et al., 2008; Soto-Quiros et al., 2012). Study on RSV-infected allergic mouse model indicated that the timing of infection might influence the immune response to virus. RSV infection before allergic sensitization dampened the Th2 response, while infection during allergen challenge enhanced the inflammatory response in mouse model (Peebles et al., 2001b; Barends et al., 2004). Pretreatment of Th1/Th2 cytokine in airway epithelial cells also influenced the RSV-induced gene expression, with enhanced expression in Th2-primed cells (Yamada et al., 2011). Comparing with Bartlettʼs group, we reduced the dosage of allergen during challenge. This change has influenced the time point of   86   inflammation peak and exacerbation. Our model induced a fast eosinophilia exacerbation at day 3 after last challenge, which is prior to the day 7 peak in Bartlettʼs model. Meanwhile, our model showed a significantly increased and prior expression of CXCL10 and MCP-1, with significant increase in IL-13 in day 5 and 7 after last challenge. Bartlettʼs group suggested a deficient of Th1 response and augmented Th2 response immediately after infection. However, due to the lack of Th1 related data, it is hard to claim that Th1 response is dampened in our model. But the difference between the two models may indicate an important role of allergen in virus-associated asthma exacerbation. Elevation of serum IgE level was associated with the predisposition of allergic sensitization and persistent wheezing in children, also as an indicator of severe symptoms for asthmatic patients after experimental rhinovirus infection (Naqvi et al., 2007; Zambrano et al., 2003). Anti-IgE treatment conducted in house dust mite and RSV-challenged mice also indicated an IgE-dependent process in airway pathology (Tumas et al., 2001). The results of serum IgE levels in our mouse model indicated that rhinovirus infection amplified the total and OVA-specific IgE increase in allergen challenged mice, markedly on day 7 post challenge (Figure 4.4). The cross-link of antigens, IgE and its high-affinity receptor (FcεRI) is associated with activation of mast cells and basophils and contributes a crucial role in asthma pathogenesis (Koketsu et al., 2013). Recent studies found that the expression of FcεRI on lung dendritic cells after   87   respiratory virus infection is associated with the recruitment of IL-13producing T cells (Benoit and Holtzman, 2010). In our model, the persistently increased mucus overproduction score may also indicate the effect of virus in asthma development and re-exacerbation. Prolonged mucus production and plugging were observed in murine cytomegalovirus-infected allergic mouse (Wu et al., 2008). In mice infected with Sendai virus, IL-13 production by natural killer (NK) T cells and alternatively-activated macrophages is responsible for persistent mucous metaplasia (Kim et al., 2008). Though we got BAL fluid from mice, we were unable to measure the cytokines levels. This may be due to the inappropriate storage of samples and technical mistakes during experiments. Considering the role of timing of infection and the effect of different dosage of allergen on inflammation status, more efforts should be made to explore the role of Th1 immune response and innate immune response in virusassociated asthma exacerbation. The role of prolonged mucus overproduction and IgE increase in susceptibility of re-exacerbation also needs further investigation.   88   Chapter 6 Conclusion   89   In this project, we have successfully established a mouse model of rhinovirus-associated asthma exacerbation with amplified inflammatory cell infiltration, mucus hyper-secretion and airway hyperresponsiveness. The time course of the mRNA levels showed the changes of inflammatory gene expression under the influence of rhinovirus infection. Taken together, rhinovirus infection enhanced the expression of several cytokines and chemokines, which contribute to the exacerbation of asthmatic symptoms. The increase in expression of Th2 cytokines and serum IgE levels indicate that rhinovirus infection increase future risk of asthma exacerbation and may be involved in the development of allergic disease. More efforts should be made to explore the exacerbation, mechanisms and our underlying mouse model virus-associated would facilitate asthma future investigation.   90   Chapter 7 References   91   Aceves, S.S., and Broide, D.H. (2008). Airway fibrosis and angiogenesis due to eosinophil trafficking in chronic asthma. Curr Mol Med 8, 350–358. Aeffner, F., and Davis, I.C. (2012). Respiratory Syncytial Virus Reverses Airway Hyperresponsiveness to Methacholine in OvalbuminSensitized Mice. PLoS ONE 7, e46660. Aikawa, T., Shimura, S., Sasaki, H., Ebina, M., and Takishima, T. (1992). Marked goblet cell hyperplasia with mucus accumulation in the airways of patients who died of severe acute asthma attack. Chest 101, 916–921. Akinbami, L.J., Moorman, J.E., and Liu, X. (2011). Asthma prevalence, health care use, and mortality: United States, 2005-2009. Natl Health Stat Rep. 1–14. American Lung Association (2012). Trends in Asthma Morbidity and Mortality. http://www.lung.org/finding-cures/our-research/trendreports/asthma-trend-report.pdf Amerio, P., Frezzolini, A., Feliciani, C., Verdolini, R., Teofoli, P., De Pità, O., et al. (2003). Eotaxins and CCR3 receptor in inflammatory and allergic skin diseases: therapeutical implications. Curr Drug Targets Inflamm Allergy 2, 81–94. Asosingh, K., Hanson, J.D., Cheng, G., Aronica, M.A., and Erzurum, S.C. (2010). Allergen-induced, eotaxin-rich, proangiogenic bone marrow progenitors: a blood-borne cellular envoy for lung eosinophilia. J Allergy Clin Immunol 125, 918–925. Badger, G.F., Dingle, J.H., Feller, A.E., Hodges, R.G., Jordan, W.S., and Rammelkamp, C.H. (1953). A Study of Illness in a Group of Cleveland Families Ii. Incidence of the Common Respiratory Diseases,. Am J Epidemiol 58, 31–40. Balhara, J., and Gounni, A.S. (2012). The alveolar macrophages in asthma: a double-edged sword. Mucosal Immunol 5, 605–609. Barends, M., Van Oosten, M., De Rond, C.G.H., Dormans, J.A.M.A., Osterhaus, A.D.M.E., Neijens, H.J., et al. (2004). Timing of infection and prior immunization with respiratory syncytial virus (RSV) in RSVenhanced allergic inflammation. J. Infect. Dis. 189, 1866–1872. Bartlett, N.W., and Johnston, S.L. (2008). Rhinoviruses. In Encyclopedia of Virology (Third Edition), Editors-in-Chief: B.W.J. Mahy, and M.H.V. van Regenmortel, eds. (Oxford: Academic Press), pp. 467– 475.   92   Bartlett, N.W., Walton, R.P., Edwards, M.R., Aniscenko, J., Caramori, G., Zhu, J., et al. (2008). Mouse models of rhinovirus-induced disease and exacerbation of allergic airway inflammation. Nat Med 14, 199–204. Bel, E.H. (2004). Clinical phenotypes of asthma. Curr Opin Pulm Med 10, 44–50. Benoit, L.A., and Holtzman, M.J. (2010). New immune pathways from chronic post-viral lung disease. Ann N Acad Sci 1183, 195–210. Van Benten, I.J., KleinJan, A., Neijens, H.J., Osterhaus, A.D., and Fokkens, W.J. (2001). Prolonged nasal eosinophilia in allergic patients after common cold. Allergy 56, 949–956. Berend, N., Salome, C.M., and King, G.G. (2008). Mechanisms of airway hyperresponsiveness in asthma. Respirology 13, 624–631. Berkovich, S., Millian, S.J., and Snyder, R.D. (1970). The association of viral and mycoplasma infections with recurrence of wheezing in the asthmatic child. Ann. Allergy 28, 43–49. Bhardwaj, N., and Ghaffari, G. (2012). Biomarkers for eosinophilic esophagitis: a review. Ann. Allergy Asthma Immunol. 109, 155–159. Bisset, L.R., and Schmid-Grendelmeier, P. (2005). Chemokines and their receptors in the pathogenesis of allergic asthma: progress and perspective. Curr. Opin. Pulm. Med. 11, 35–42. Biswas, S.K., and Mantovani, A. (2010). Macrophage plasticity and interaction with lymphocyte subsets: cancer as a paradigm. Nat. Immunol. 11, 889–896. Bochner, B.S., Hudson, S.A., Xiao, H.Q., and Liu, M.C. (2003). Release of both CCR4-active and CXCR3-active chemokines during human allergic pulmonary late-phase reactions. J. Allergy Clin. Immunol. 112, 930–934. Brannan, J.D., and Lougheed, M.D. (2012). Airway hyperresponsiveness in asthma: mechanisms, clinical significance, and treatment. Front. Physiol. 3, 460. Brar, T., Nagaraj, S., and Mohapatra, S. (2012). Microbes and asthma: the missing cellular and molecular links. Curr. Opin. Pulm. Med. 18, 14–22. Brightling, C.E., Bradding, P., Symon, F.A., Holgate, S.T., Wardlaw, A.J., and Pavord, I.D. (2002). Mast-cell infiltration of airway smooth muscle in asthma. N. Engl. J. Med. 346, 1699–1705.   93   Brightling, C.E., Kaur, D., Berger, P., Morgan, A.J., Wardlaw, A.J., and Bradding, P. (2005a). Differential expression of CCR3 and CXCR3 by human lung and bone marrow-derived mast cells: implications for tissue mast cell migration. J. Leukoc. Biol. 77, 759–766. Brightling, C.E., Ammit, A.J., Kaur, D., Black, J.L., Wardlaw, A.J., Hughes, J.M., et al. (2005b). The CXCL10/CXCR3 axis mediates human lung mast cell migration to asthmatic airway smooth muscle. Am. J. Respir. Crit. Care Med. 171, 1103–1108. Busse, W.W., and Rosenwasser, L.J. (2003). Mechanisms of asthma. J. Allergy Clin. Immunol. 111, S799–804. Castro, M., Chaplin, D.D., Walter, M.J., and Holtzman, M.J. (2000). Could asthma be worsened by stimulating the T-helper type 1 immune response? Am. J. Respir. Cell Mol. Biol. 22, 143–146. Chan, C.-K., Kuo, M.-L., Yeh, K.-W., Ou, L.-S., Chen, L.-C., Yao, T.-C., et al. (2009). Sequential evaluation of serum monocyte chemotactic protein 1 among asymptomatic state and acute exacerbation and remission of asthma in children. J. Asthma 46, 225–228. Chauhan, A.J., Inskip, H.M., Linaker, C.H., Smith, S., Schreiber, J., Johnston, S.L., et al. (2003). Personal exposure to nitrogen dioxide (NO2) and the severity of virus-induced asthma in children. Lancet 361, 1939–1944. Chen, S., Huang, F., Tan, G., Wang, C., Huang, Y., Wang, H., et al. (2009). RNA interference against interleukin-5 attenuates airway inflammation and hyperresponsiveness in an asthma model. J. Zhejiang Univ. Sci. B 10, 22–28. Cheng, C., Ho, W.E., Goh, F.Y., Guan, S.P., Kong, L.R., Lai, W.-Q., et al. (2011). Anti-malarial drug artesunate attenuates experimental allergic asthma via inhibition of the phosphoinositide 3-kinase/Akt pathway. PloS One 6, e20932. Cho, J.Y., Song, D.J., Pham, A., Rosenthal, P., Miller, M., Dayan, S., et al. (2010). Chronic OVA allergen challenged Siglec-F deficient mice have increased mucus, remodeling, and epithelial Siglec-F ligands which are up-regulated by IL-4 and IL-13. Respir. Res. 11, 154. Chung, K.F., Caramori, G., and Adcock, I.M. (2009). Inhaled corticosteroids as combination therapy with beta-adrenergic agonists in airways disease: present and future. Eur. J. Clin. Pharmacol. 65, 853– 871. Cockcroft, D.W., and Davis, B.E. (2006). Mechanisms of airway hyperresponsiveness. J Allergy Clin Immunol 118, 551–559.   94   Collington, S.J., Westwick, J., Williams, T.J., and Weller, C.L. (2010). The function of CCR3 on mouse bone marrow-derived mast cells in vitro. Immunology 129, 115–124. Conti, P., and DiGioacchino, M. (2001). MCP-1 and RANTES are mediators of acute and chronic inflammation. Allergy Asthma Proc. 22, 133–137. Contoli, M., Message, S.D., Laza-Stanca, V., Edwards, M.R., Wark, P.A.B., Bartlett, N.W., et al. (2006). Role of deficient type III interferonlambda production in asthma exacerbations. Nat. Med. 12, 1023–1026. Crimi, E., Spanevello, A., Neri, M., Ind, P.W., Rossi, G.A., and Brusasco, V. (1998). Dissociation between airway inflammation and airway hyperresponsiveness in allergic asthma. Am. J. Respir. Crit. Care Med. 157, 4–9. Dakhama, A., Bramley, A.M., Chan, N.G., McKay, K.O., Schellenberg, R.R., and Hegele, R.G. (1999). Effect of respiratory syncytial virus on subsequent allergic sensitization to ovalbumin in guinea-pigs. Eur. Respir. J. 13, 976–982. DeMeo, D.L., and Weiss, S.T. (2009). Chapter 2 - Epidemiology. In Asthma and COPD (Second Edition), (Oxford: Academic Press), pp. 9– 21. Dhaouadi, T., Sfar, I., Aounallah-Skhiri, H., Jendoubi-Ayed, S., Bouacha, H., Ben Abdallah, T., et al. (2013). MCP-1, CCR2 and CCR5 Polymorphisms in Tunisian Patients with Atopic Asthma. Iran. J. Allergy Asthma Immunol. 12, 29–36. Doull, I.J.M., Lampe, F.C., Smith, S., Schreiber, J., Freezer, N.J., and Holgate, S.T. (1997). Effect of inhaled corticosteroids on episodes of wheezing associated with viral infection in school age children: randomised double blind placebo controlled trial. BMJ 315, 858–862. Drake, K.A., Galanter, J.M., and Burchard, E.G. (2008). Race, Ethnicity and Social Class and the Complex Etiologies of Asthma. Pharmacogenomics 9, 453–462. Dyer, K.D., Percopo, C.M., Fischer, E.R., Gabryszewski, S.J., and Rosenberg, H.F. (2009). Pneumoviruses infect eosinophils and elicit MyD88-dependent release of chemoattractant cytokines and interleukin-6. Blood 114, 2649–2656. Ege, M.J., Mayer, M., Normand, A.-C., Genuneit, J., Cookson, W.O.C.M., Braun-Fahrländer, C., et al. (2011). Exposure to Environmental Microorganisms and Childhood Asthma. N. Engl. J. Med. 364, 701–709.   95   Erickson, S.E., Iribarren, C., Tolstykh, I.V., Blanc, P.D., and Eisner, M.D. (2007). Effect of race on asthma management and outcomes in a large, integrated managed care organization. Arch. Intern. Med. 167, 1846–1852. Evans, C.M., Kim, K., Tuvim, M.J., and Dickey, B.F. (2009). Mucus hypersecretion in asthma: causes and effects. Curr. Opin. Pulm. Med. 15, 4–11. Fahy, J.V. (2002). Goblet cell and mucin gene abnormalities in asthma. Chest 122, 320S–326S. Fahy, J.V., and Dickey, B.F. (2010). Airway Mucus Function and Dysfunction. N. Engl. J. Med. 363, 2233–2247. Falsey, A.R. (2005). Respiratory syncytial virus infection in elderly and high-risk adults. Exp. Lung Res. 31 Suppl 1, 77. Fishbein, A.B., and Fuleihan, R.L. (2012). The hygiene hypothesis revisited: does exposure to infectious agents protect us from allergy? Curr. Opin. Pediatr. 24, 98–102. Fraenkel, D.J., Bardin, P.G., Sanderson, G., Lampe, F., Johnston, S.L., and Holgate, S.T. (1995). Lower airways inflammation during rhinovirus colds in normal and in asthmatic subjects. Am. J. Respir. Crit. Care Med. 151, 879–886. Frank, K.M., Zhou, T., Moreno-Vinasco, L., Hollett, B., Garcia, J.G.N., and Wardenburg, J.B. (2012). Host Response Signature to Staphylococcus aureus Alpha-Hemolysin Implicates Pulmonary Th17 Response. Infect. Immun. 80, 3161–3169. Ganz, T. (2003). Defensins: antimicrobial peptides of innate immunity. Nat. Rev. Immunol. 3, 710–720. Gaudernak, E., Seipelt, J., Triendl, A., Grassauer, A., and Kuechler, E. (2002). Antiviral effects of pyrrolidine dithiocarbamate on human rhinoviruses. J. Virol. 76, 6004–6015. Gehlhar, K., Bilitewski, C., Reinitz-Rademacher, K., Rohde, G., and Bufe, A. (2006). Impaired virus-induced interferon-alpha2 release in adult asthmatic patients. Clin. Exp. Allergy 36, 331–337. Gern, J.E., Vrtis, R., Grindle, K.A., Swenson, C., and Busse, W.W. (2000). Relationship of upper and lower airway cytokines to outcome of experimental rhinovirus infection. Am. J. Respir. Crit. Care Med. 162, 2226–2231. Global Initiative for Asthma (GINA) (2011). Global strategy for asthma management and prevention (updated 2011).   96   Gordon, S. (2003). Alternative activation of macrophages. Nat. Rev. Immunol. 3, 23–35. Gordon, S., and Taylor, P.R. (2005). Monocyte and macrophage heterogeneity. Nat. Rev. Immunol. 5, 953–964. Gotera, J., Giuffrida, M., Mavarez, A., Pons, H., Bermudez, J., Maldonado, M., et al. (2012). Respiratory syncytial virus infection increases regulated on activation normal T cell expressed and secreted and monocyte chemotactic protein 1 levels in serum of patients with asthma and in human monocyte cultures. Ann. Allergy. Asthma. Immunol. 108, 316–320. Gould, H.J., and Sutton, B.J. (2008). IgE in allergy and asthma today. Nat. Rev. Immunol. 8, 205–217. Grayson, M.H., Cheung, D., Rohlfing, M.M., Kitchens, R., Spiegel, D.E., Tucker, J., et al. (2007). Induction of high-affinity IgE receptor on lung dendritic cells during viral infection leads to mucous cell metaplasia. J. Exp. Med. 204, 2759–2769. Green, R.H., Brightling, C.E., McKenna, S., Hargadon, B., Parker, D., Bradding, P., et al. (2002a). Asthma exacerbations and sputum eosinophil counts: a randomised controlled trial. Lancet 360, 1715– 1721. Green, R.M., Custovic, A., Sanderson, G., Hunter, J., Johnston, S.L., and Woodcock, A. (2002b). Synergism between allergens and viruses and risk of hospital admission with asthma: case-control study. BMJ 324, 763. Greve, J.M., Davis, G., Meyer, A.M., Forte, C.P., Yost, S.C., Marlor, C.W., et al. (1989). The major human rhinovirus receptor is ICAM-1. Cell 56, 839–847. Grünberg, K., Timmers, M.C., Smits, H.H., de KLERK, E.P.A., Dick, E.C., Spaan, W.J.M., et al. (1997). Effect of experimental rhinovirus 16 colds on airway hyperresponsiveness to histamine and interleukin-8 in nasal lavage in asthmatic subjects in vivo. Clin. Exp. Allergy 27, 36–45. Grünberg, K., Sharon, R.F., Hiltermann, T.J., Brahim, J.J., Dick, E.C., Sterk, P.J., et al. (2000). Experimental rhinovirus 16 infection increases intercellular adhesion molecule-1 expression in bronchial epithelium of asthmatics regardless of inhaled steroid treatment. Clin. Exp. Allergy 30, 1015–1023. Grünig, G., Warnock, M., Wakil, A.E., Venkayya, R., Brombacher, F., Rennick, D.M., et al. (1998). Requirement for IL-13 Independently of IL4 in Experimental Asthma. Science 282, 2261–2263.   97   Guan, W.-D., Yang, Z.-F., Liu, N., Qin, S., Zhang, F.-X., and Zhu, Y.-T. (2008). In vitro experimental study on the effect of resveratrol against several kinds of respiroviruses. J. Chin. Med. Mater. 31, 1388–1390. Hall, D.J., Bates, M.E., Guar, L., Cronan, M., Korpi, N., and Bertics, P.J. (2005). The Role of p38 MAPK in Rhinovirus-Induced Monocyte Chemoattractant Protein-1 Production by Monocytic-Lineage Cells. J. Immunol. 174, 8056–8063. Hammad, H., and Lambrecht, B.N. (2008). Dendritic cells and epithelial cells: linking innate and adaptive immunity in asthma. Nat. Rev. Immunol. 8, 193–204. Harris, J.R., and Racaniello, V.R. (2003). Changes in rhinovirus protein 2C allow efficient replication in mouse cells. J. Virol. 77, 4773–4780. Harris, J.R., and Racaniello, V.R. (2005). Amino acid changes in proteins 2B and 3A mediate rhinovirus type 39 growth in mouse cells. J. Virol. 79, 5363–5373. Hays, S.R., and Fahy, J.V. (2003). The role of mucus in fatal asthma. Am. J. Med. 115, 68–69. He, L., Hu, C., Guobin, C., Qiuping, Z., Qun, L., Xiaolian, Z., et al. (2004a). Highly up-regulated CXCR3 expression on eosinophils in mice infected with Schistosoma japonicum. Immunology 111, 107–117. He, S.-H., Zheng, J., and Duan, M.-K. (2004b). Induction of mucin secretion from human bronchial tissue and epithelial cells by rhinovirus and lipopolysaccharide. Acta Pharmacol. Sin. 25, 1176–1181. Heikkinen, T., and Järvinen, A. (2003). The common cold. The Lancet 361, 51–59. Hewson, C.A., Haas, J.J., Bartlett, N.W., Message, S.D., Laza-Stanca, V., Kebadze, T., et al. (2010). Rhinovirus induces MUC5AC in a human infection model and in vitro via NF-κB and EGFR pathways. Eur. Respir. 36, 1425–1435. Hiraguchi, Y., Nagao, M., Hosoki, K., Tokuda, R., and Fujisawa, T. (2008). Neutrophil Proteases Activate Eosinophil Function in vitro. Int. Arch. Allergy Immunol. 146 Suppl 1, 16–21. Hofer, F., Gruenberger, M., Kowalski, H., Machat, H., Huettinger, M., Kuechler, E., et al. (1994). Members of the low density lipoprotein receptor family mediate cell entry of a minor-group common cold virus. Proc. Natl. Acad. Sci. 91, 1839–1842. Holgate, S.T. (2008). Pathogenesis of asthma. Clin. Exp. Allergy 38, 872–897.   98   Holtzman, M.J., Byers, D.E., Benoit, L.A., Battaile, J.T., You, Y., Agapov, E., et al. (2009). Immune pathways for translating viral infection into chronic airway disease. Adv. Immunol. 102, 245–276. Homer, R.J., and Elias, J.A. (2000). Consequences of long-term inflammation. Airway remodeling. Clin. Chest Med. 21, 331–343, ix. Hong, C.S., Park, J.W., and Nahm, D.H. (1994). Measurement of IgE and IgG subclass antibodies to whole body antigen and two major allergens (Der fI & Der fII) of Dermatophagoides farinae in normal subjects and asthmatics. Yonsei Med. J. 35, 453–463. Huguenel, E.D., Cohn, D., Dockum, D.P., Greve, J.M., Fournel, M.A., Hammond, L., et al. (1997). Prevention of rhinovirus infection in chimpanzees by soluble intercellular adhesion molecule-1. Am. J. Respir. Crit. Care Med. 155, 1206–1210. Jackson, D.J., Gangnon, R.E., Evans, M.D., Roberg, K.A., Anderson, E.L., Pappas, T.E., et al. (2008). Wheezing rhinovirus illnesses in early life predict asthma development in high-risk children. Am. J. Respir. Crit. Care Med. 178, 667–672. Jackson, D.J., Sykes, A., Mallia, P., and Johnston, S.L. (2011). Asthma exacerbations: origin, effect, and prevention. J. Allergy Clin. Immunol. 128, 1165–1174. Jackson, D.J., Evans, M.D., Gangnon, R.E., Tisler, C.J., Pappas, T.E., Lee, W.-M., et al. (2012). Evidence for a causal relationship between allergic sensitization and rhinovirus wheezing in early life. Am. J. Respir. Crit. Care Med. 185, 281–285. Johnston, N.W., and Sears, M.R. (2006). Asthma exacerbations · 1: Epidemiology. Thorax 61, 722–728. Johnston, N.W., Johnston, S.L., Duncan, J.M., Greene, J.M., Kebadze, T., Keith, P.K., et al. (2005). The September epidemic of asthma exacerbations in children: A search for etiology. J. Allergy Clin. Immunol. 115, 132–138. Johnston, S.L., Pattemore, P.K., Sanderson, G., Smith, S., Lampe, F., Josephs, L., et al. (1995). Community study of role of viral infections in exacerbations of asthma in 9-11 year old children. BMJ 310, 1225– 1229. Kaiko, G.E., Loh, Z., Spann, K., Lynch, J.P., Lalwani, A., Zheng, Z., et al. (2013). Toll-like receptor 7 gene deficiency and early-life Pneumovirus infection interact to predispose toward the development of asthma-like pathology in mice. J. Allergy Clin. Immunol. 131, 1331– 1339.e10.   99   Kato, M., Tsukagoshi, H., Yoshizumi, M., Saitoh, M., Kozawa, K., Yamada, Y., et al. (2011). Different cytokine profile and eosinophil activation are involved in rhinovirus- and RS virus-induced acute exacerbation of childhood wheezing. Pediatr. Allergy Immunol. 22, e87–94. Kay, A.B. (2005). The role of eosinophils in the pathogenesis of asthma. Trends Mol. Med. 11, 148–152. Kelly, J.T., and Busse, W.W. (2008). Host immune responses to rhinovirus: Mechanisms in asthma. J. Allergy Clin. Immunol. 122, 671– 682. Van Kempen, M., Bachert, C., and Van Cauwenberge, P. (1999). An update on the pathophysiology of rhinovirus upper respiratory tract infections. Rhinology 37, 97–103. Kennedy, J.L., Turner, R.B., Braciale, T., Heymann, P.W., and Borish, L. (2012). Pathogenesis of rhinovirus infection. Curr. Opin. Virol. 2, 287–293. Kesson, A.M. (2007). Respiratory virus infections. Paediatr. Respir. Rev. 8, 240–248. Khaled, W.T., Read, E.K.C., Nicholson, S.E., Baxter, F.O., Brennan, A.J., Came, P.J., et al. (2007). The IL-4/IL-13/Stat6 signalling pathway promotes luminal mammary epithelial cell development. Dev. Camb. Engl. 134, 2739–2750. Kibe, A., Inoue, H., Fukuyama, S., Machida, K., Matsumoto, K., Koto, H., et al. (2003). Differential regulation by glucocorticoid of interleukin13-induced eosinophilia, hyperresponsiveness, and goblet cell hyperplasia in mouse airways. Am. J. Respir. Crit. Care Med. 167, 50– 56. Kim, E.Y., Battaile, J.T., Patel, A.C., You, Y., Agapov, E., Grayson, M.H., et al. (2008). Persistent activation of an innate immune response translates respiratory viral infection into chronic lung disease. Nat. Med. 14, 633–640. Kim, H.Y., DeKruyff, R.H., and Umetsu, D.T. (2010). The many paths to asthma: phenotype shaped by innate and adaptive immunity. Nat. Immunol. 11, 577–584. Kinyanjui, M.W., Shan, J., Nakada, E.M., Qureshi, S.T., and Fixman, E.D. (2013). Dose-dependent effects of IL-17 on IL-13-induced airway inflammatory responses and airway hyperresponsiveness. J. Immunol. Baltim. Md 1950 190, 3859–3868.   100   Kloepfer, K.M., and Gern, J.E. (2010). Virus/allergen interactions and exacerbations of asthma. Immunol. Allergy Clin. North Am. 30, 553– 563, vii. Koketsu, R., Yamaguchi, M., Suzukawa, M., Tanaka, Y., Tashimo, H., Arai, H., et al. (2013). Pretreatment with low levels of FcεRIcrosslinking stimulation enhances basophil mediator release. Int. Arch. Allergy Immunol. 161 Suppl 2, 23–31. Kolaczkowska, E., and Kubes, P. (2013). Neutrophil recruitment and function in health and inflammation. Nat. Rev. Immunol. 13, 159–175. Kondo, Y., Matsuse, H., Machida, I., Kawano, T., Saeki, S., Tomari, S., et al. (2004). Effects of primary and secondary low-grade respiratory syncytial virus infections in a murine model of asthma. Clin. Exp. Allergy 34, 1307–1313. Korpi-Steiner, N.L., Valkenaar, S.M., Bates, M.E., Evans, M.D., Gern, J.E., and Bertics, P.J. (2010). Human monocytic cells direct the robust release of CXCL10 by bronchial epithelial cells during rhinovirus infection. Clin. Exp. Allergy 40, 1203–1213. Kotaniemi-Syrjänen, A., Vainionpää, R., Reijonen, T.M., Waris, M., Korhonen, K., and Korppi, M. (2003). Rhinovirus-induced wheezing in infancy--the first sign of childhood asthma? J. Allergy Clin. Immunol. 111, 66–71. Krunkosky, T.M., Fischer, B.M., Martin, L.D., Jones, N., Akley, N.J., and Adler, K.B. (2000). Effects of TNF-alpha on expression of ICAM-1 in human airway epithelial cells in vitro. Signaling pathways controlling surface and gene expression. Am. J. Respir. Cell Mol. Biol. 22, 685– 692. Kumar, R.K., Yang, M., Herbert, C., and Foster, P.S. (2012). Interferonγ, pulmonary macrophages and airway responsiveness in asthma. Inflamm. Allergy Drug Targets 11, 292–297. Kuperman, D.A., Huang, X., Koth, L.L., Chang, G.H., Dolganov, G.M., Zhu, Z., et al. (2002). Direct effects of interleukin-13 on epithelial cells cause airway hyperreactivity and mucus overproduction in asthma. Nat. Med. 8, 885–889. Kurashima, K., Mukaida, N., Fujimura, M., Schröder, J.M., Matsuda, T., and Matsushima, K. (1996). Increase of chemokine levels in sputum precedes exacerbation of acute asthma attacks. J. Leukoc. Biol. 59, 313–316. Kusel, M.M.H., de Klerk, N.H., Kebadze, T., Vohma, V., Holt, P.G., Johnston, S.L., et al. (2007). Early-life respiratory viral infections, atopic   101   sensitization, and risk of subsequent development of persistent asthma. J. Allergy Clin. Immunol. 119, 1105–1110. Kuyper, L.M., Paré, P.D., Hogg, J.C., Lambert, R.K., Ionescu, D., Woods, R., et al. (2003). Characterization of airway plugging in fatal asthma. Am. J. Med. 115, 6–11. Lambert, H.P., and Stern, H. (1972). Infective factors in exacerbations of bronchitis and asthma. Br. Med. J. 3, 323–327. Lambrecht, B.N., and Hammad, H. (2012). The airway epithelium in asthma. Nat. Med. 18, 684–692. Laza-Stanca, V., Stanciu, L.A., Message, S.D., Edwards, M.R., Gern, J.E., and Johnston, S.L. (2006). Rhinovirus Replication in Human Macrophages Induces NF-κB-Dependent Tumor Necrosis Factor Alpha Production. J. Virol. 80, 8248–8258. Leckie, M.J., ten Brinke, A., Khan, J., Diamant, Z., OʼConnor, B.J., Walls, C.M., et al. (2000). Effects of an interleukin-5 blocking monoclonal antibody on eosinophils, airway hyper-responsiveness, and the late asthmatic response. Lancet 356, 2144–2148. Lee, E.Y., Lee, Z.-H., and Song, Y.W. (2009). CXCL10 and autoimmune diseases. Autoimmun. Rev. 8, 379–383. Lewis, T.C., Henderson, T.A., Carpenter, A.R., Ramirez, I.A., McHenry, C.L., Goldsmith, A.M., et al. (2012). Nasal cytokine responses to natural colds in asthmatic children. Clin. Exp. Allergy J. Br. Soc. Allergy Clin. Immunol. 42, 1734–1744. Lindell, D.M., Lane, T.E., and Lukacs, N.W. (2008). CXCL10/CXCR3mediated responses promote immunity to respiratory syncytial virus infection by augmenting dendritic cell and CD8(+) T cell efficacy. Eur. J. Immunol. 38, 2168–2179. Lindenbach, B.D. (2004). Reed & Muench Calculator (Yale University). www.med.yale.edu/micropath/pdf/Infectivity%20calculator.xls Lister, S., Sheppeard, V., Morgan, G., Corbett, S., Kaldor, J., and Henry, R. (2001). February asthma outbreaks in NSW: a case control study. Aust. N. Z. J. Public Health 25, 514–519. Lommatzsch, M. (2012). Airway hyperresponsiveness: new insights into the pathogenesis. Semin. Respir. Crit. Care Med. 33, 579–587. Lowy, R.J. (2003). Influenza virus induction of apoptosis by intrinsic and extrinsic mechanisms. Int. Rev. Immunol. 22, 425–449.   102   Ma, X.-Z., Bartczak, A., Zhang, J., Khattar, R., Chen, L., Liu, M.F., et al. (2010). Proteasome inhibition in vivo promotes survival in a lethal murine model of severe acute respiratory syndrome. J. Virol. 84, 12419–12428. Mackay, D., Haw, S., Ayres, J.G., Fischbacher, C., and Pell, J.P. (2010). Smoke-free legislation and hospitalizations for childhood asthma. N. Engl. J. Med. 363, 1139–1145. Maffey, A.F., Barrero, P.R., Venialgo, C., Fernández, F., Fuse, V.A., Saia, M., et al. (2010). Viruses and atypical bacteria associated with asthma exacerbations in hospitalized children. Pediatr. Pulmonol. 45, 619–625. Mamessier, E., and Magnan, A. (2006). Cytokines in atopic diseases: revisiting the Th2 dogma. Eur. J. Dermatol. EJD 16, 103–113. Martinez, F.O., Sica, A., Mantovani, A., and Locati, M. (2008). Macrophage activation and polarization. Front. Biosci. J. Virtual Libr. 13, 453–461. Masoli, M., Fabian, D., Holt, S., Beasley, R., and Program, G.I. for A. (GINA) (2004). The global burden of asthma: executive summary of the GINA Dissemination Committee Report. Allergy 59, 469–478. Matsukura, S., Stellato, C., Georas, S.N., Casolaro, V., Plitt, J.R., Miura, K., et al. (2001). Interleukin-13 upregulates eotaxin expression in airway epithelial cells by a STAT6-dependent mechanism. Am. J. Respir. Cell Mol. Biol. 24, 755–761. Mattes, J., Yang, M., Siqueira, A., Clark, K., MacKenzie, J., McKenzie, A.N., et al. (2001). IL-13 induces airways hyperreactivity independently of the IL-4R alpha chain in the allergic lung. J. Immunol. Baltim. Md 1950 167, 1683–1692. McConnell, R., Berhane, K., Gilliland, F., Molitor, J., Thomas, D., Lurmann, F., et al. (2003). Prospective study of air pollution and bronchitic symptoms in children with asthma. Am. J. Respir. Crit. Care Med. 168, 790–797. Medoff, B.D., Sauty, A., Tager, A.M., Maclean, J.A., Smith, R.N., Mathew, A., et al. (2002). IFN-gamma-inducible protein 10 (CXCL10) contributes to airway hyperreactivity and airway inflammation in a mouse model of asthma. J. Immunol. Baltim. Md 1950 168, 5278–5286. Melgert, B.N., Postma, D.S., Kuipers, I., Geerlings, M., Luinge, M.A., van der Strate, B.W.A., et al. (2005). Female mice are more susceptible to the development of allergic airway inflammation than male mice. Clin. Exp. Allergy 35, 1496–1503.   103   Message, S.D., and Johnston, S.L. (2001). The immunology of virus infection in asthma. Eur. Respir. J. 18, 1013–1025. Message, S.D., Laza-Stanca, V., Mallia, P., Parker, H.L., Zhu, J., Kebadze, T., et al. (2008). Rhinovirus-induced lower respiratory illness is increased in asthma and related to virus load and Th1/2 cytokine and IL-10 production. Proc. Natl. Acad. Sci. U. S. A. 105, 13562–13567. Miotto, D., Christodoulopoulos, P., Olivenstein, R., Taha, R., Cameron, L., Tsicopoulos, A., et al. (2001). Expression of IFN-gamma-inducible protein; monocyte chemotactic proteins 1, 3, and 4; and eotaxin in TH1- and TH2-mediated lung diseases. J. Allergy Clin. Immunol. 107, 664–670. Molfino, N.A. (2010). Increased vagal airway tone in fatal asthma. Med. Hypotheses 74, 521–523. Monteseirín, J. (2009). Neutrophils and asthma. J. Investig. Allergol. Clin. Immunol. 19, 340–354. Monto, A.S. (2002). The seasonality of rhinovirus infections and its implications for clinical recognition. Clin. Ther. 24, 1987–1997. Monto, A.S. (2003). Epidemiology of viral respiratory infections. Dis. Mon. 49, 160–174. Monto, A.S., Bryan, E.R., and Ohmit, S. (1987). Rhinovirus infections in Tecumseh, Michigan: frequency of illness and number of serotypes. J. Infect. Dis. 156, 43–49. Moon, K.-A., Kim, S.Y., Kim, T.-B., Yun, E.S., Park, C.-S., Cho, Y.S., et al. (2007). Allergen-induced CD11b+ CD11c(int) CCR3+ macrophages in the lung promote eosinophilic airway inflammation in a mouse asthma model. Int. Immunol. 19, 1371–1381. Moreira, A.P., and Hogaboam, C.M. (2011). Macrophages in allergic asthma: fine-tuning their pro- and anti-inflammatory actions for disease resolution. J. Interf. Cytokine Res. 31, 485–491. Mould, A.W., Ramsay, A.J., Matthaei, K.I., Young, I.G., Rothenberg, M.E., and Foster, P.S. (2000). The Effect of IL-5 and Eotaxin Expression in the Lung on Eosinophil Trafficking and Degranulation and the Induction of Bronchial Hyperreactivity. J. Immunol. 164, 2142– 2150. Mukherjee, A.B., and Zhang, Z. (2011). Allergic asthma: influence of genetic and environmental factors. J. Biol. Chem. 286, 32883–32889. Müller, M., Carter, S., Hofer, M.J., and Campbell, I.L. (2010). Review: The chemokine receptor CXCR3 and its ligands CXCL9, CXCL10 and   104   CXCL11 in neuroimmunity--a tale of conflict and conundrum. Neuropathol. Appl. Neurobiol. 36, 368–387. Murray, C.S., Poletti, G., Kebadze, T., Morris, J., Woodcock, A., Johnston, S.L., et al. (2006). Study of modifiable risk factors for asthma exacerbations: virus infection and allergen exposure increase the risk of asthma hospital admissions in children. Thorax 61, 376–382. Myou, S., Leff, A.R., Myo, S., Boetticher, E., Tong, J., Meliton, A.Y., et al. (2003). Blockade of Inflammation and Airway Hyperresponsiveness in Immune-sensitized Mice by Dominant-Negative Phosphoinositide 3Kinase–TAT. J. Exp. Med. 198, 1573–1582. Nagarkar, D.R., Wang, Q., Shim, J., Zhao, Y., Tsai, W.C., Lukacs, N.W., et al. (2009). CXCR2 is required for neutrophilic airways inflammation and hyperresponsiveness in a mouse model of human rhinovirus infection. J. Immunol. Baltim. Md 1950 183, 6698–6707. Nagarkar, D.R., Bowman, E.R., Schneider, D., Wang, Q., Shim, J., Zhao, Y., et al. (2010). Rhinovirus infection of allergen-sensitized and challenged mice induces eotaxin release from functionally polarized macrophages. J. Immunol. Baltim. Md 1950 185, 2525–2535. Nair, P., Gaga, M., Zervas, E., Alagha, K., Hargreave, F.E., OʼByrne, P.M., et al. (2012). Safety and efficacy of a CXCR2 antagonist in patients with severe asthma and sputum neutrophils: a randomized, placebo-controlled clinical trial. Clin. Exp. Allergy 42, 1097–1103. Nakanishi, Y., Nakatsuji, M., Seno, H., Ishizu, S., Akitake-Kawano, R., Kanda, K., et al. (2011). COX-2 inhibition alters the phenotype of tumor-associated macrophages from M2 to M1 in ApcMin/+ mouse polyps. Carcinogenesis 32, 1333–1339. Naqvi, M., Choudhry, S., Tsai, H.-J., Thyne, S., Navarro, D., Nazario, S., et al. (2007). Association between IgE levels and asthma severity among African American, Mexican, and Puerto Rican patients with asthma. J. Allergy Clin. Immunol. 120, 137–143. Newcomb, D.C., Sajjan, U.S., Nagarkar, D.R., Wang, Q., Nanua, S., Zhou, Y., et al. (2008). Human rhinovirus 1B exposure induces phosphatidylinositol 3-kinase-dependent airway inflammation in mice. Am. J. Respir. Crit. Care Med. 177, 1111–1121. Oliver, B.G.G., Lim, S., Wark, P., Laza-Stanca, V., King, N., Black, J.L., et al. (2008). Rhinovirus exposure impairs immune responses to bacterial products in human alveolar macrophages. Thorax 63, 519– 525. Pantano, C., Ather, J.L., Alcorn, J.F., Poynter, M.E., Brown, A.L., Guala, A.S., et al. (2008). Nuclear factor-kappaB Activation in Airway   105   Epithelium Induces Inflammation and Hyperresponsiveness. Am. J. Respir. Crit. Care Med. 177, 959–969. Papadopoulos, N.G., Bates, P.J., Bardin, P.G., Papi, A., Leir, S.H., Fraenkel, D.J., et al. (2000). Rhinoviruses infect the lower airways. J. Infect. Dis. 181, 1875–1884. Papadopoulos, N.G., Papi, A., Meyer, J., Stanciu, L.A., Salvi, S., Holgate, S.T., et al. (2001). Rhinovirus infection up-regulates eotaxin and eotaxin-2 expression in bronchial epithelial cells. Clin. Exp. Allergy 31, 1060–1066. Papadopoulos, N.G., Stanciu, L.A., Papi, A., Holgate, S.T., and Johnston, S.L. (2002). Rhinovirus-induced alterations on peripheral blood mononuclear cell phenotype and costimulatory molecule expression in normal and atopic asthmatic subjects. Clin. Exp. Allergy 32, 537–542. Park, S.S., Kitchens, R.T., and Grayson, M.H. (2008). Single Non-Viral Antigen Exposure During a Paramyxoviral Respiratory Infection is Sufficient to Drive Specific IgE Production. J. Allergy Clin. Immunol. 121, S139–S139. Peebles, R.S., Jr, Sheller, J.R., Collins, R.D., Jarzecka, A.K., Mitchell, D.B., Parker, R.A., et al. (2001a). Respiratory syncytial virus infection does not increase allergen-induced type 2 cytokine production, yet increases airway hyperresponsiveness in mice. J. Med. Virol. 63, 178– 188. Peebles, R.S., Jr, Hashimoto, K., Collins, R.D., Jarzecka, K., Furlong, J., Mitchell, D.B., et al. (2001b). Immune interaction between respiratory syncytial virus infection and allergen sensitization critically depends on timing of challenges. J. Infect. Dis. 184, 1374–1379. Plessa, E., Diakakis, P., Gardelis, J., Thirios, A., Koletsi, P., and Falagas, M.E. (2010). Clinical features, risk factors, and complications among pediatric patients with pandemic influenza A (H1N1). Clin. Pediatr. (Phila.) 49, 777–781. Proust, B., Nahori, M.A., Ruffie, C., Lefort, J., and Vargaftig, B.B. (2003). Persistence of bronchopulmonary hyper-reactivity and eosinophilic lung inflammation after anti-IL-5 or -IL-13 treatment in allergic BALB/c and IL-4Ralpha knockout mice. Clin. Exp. Allergy 33, 119–131. Randolph, D.A., Stephens, R., Carruthers, C.J.L., and Chaplin, D.D. (1999). Cooperation between Th1 and Th2 cells in a murine model of eosinophilic airway inflammation. J. Clin. Invest. 104, 1021–1029.   106   Reddel, H.K., Taylor, D.R., Bateman, E.D., Boulet, L.-P., Boushey, H.A., Busse, W.W., et al. (2009). An official American Thoracic Society/European Respiratory Society statement: asthma control and exacerbations: standardizing endpoints for clinical asthma trials and clinical practice. Am. J. Respir. Crit. Care Med. 180, 59–99. Reed, L.J., and Muench, H. (1938). A Simple Method of Estimating Fifty Per Cent Endpoints,. Am. J. Epidemiol. 27, 493–497. Reuter, S., Dehzad, N., Martin, H., Böhm, L., Becker, M., Buhl, R., et al. (2012). TLR3 but not TLR7/8 ligand induces allergic sensitization to inhaled allergen. J. Immunol. Baltim. Md 1950 188, 5123–5131. Riedel, F., Krause, A., Slenczka, W., and Rieger, C.H.L. (1996). Parainfluenza-3-virus infection enhances allergic sensitization in the guinea-pig. Clin. Exp. Allergy 26, 603–609. Robinson, D.S., Campbell, D.A., Durham, S.R., Pfeffer, J., Barnes, P.J., and Chung, K.F. (2003). Systematic assessment of difficult-to-treat asthma. Eur. Respir. J. 22, 478–483. Rogers, D.F. (2004). Airway mucus hypersecretion in asthma: an undervalued pathology? Curr. Opin. Pharmacol. 4, 241–250. Rollinger, J.M., and Schmidtke, M. (2011). The human rhinovirus: human-pathological impact, mechanisms of antirhinoviral agents, and strategies for their discovery. Med. Res. Rev. 31, 42–92. Rosenberg, H.F., and Domachowske, J.B. (2001). Eosinophils, eosinophil ribonucleases, and their role in host defense against respiratory virus pathogens. J. Leukoc. Biol. 70, 691–698. Rosenberg, H.F., Dyer, K.D., and Domachowske, J.B. (2009). Eosinophils and their interactions with respiratory virus pathogens. Immunol. Res. 43, 128–137. Rosenberg, H.F., Dyer, K.D., and Foster, P.S. (2013). Eosinophils: changing perspectives in health and disease. Nat. Rev. Immunol. 13, 9–22. Saad-El-Din Bessa, S., Abo El-Magd, G.H., and Mabrouk, M.M. (2012). Serum chemokines RANTES and monocyte chemoattractant protein-1 in Egyptian patients with atopic asthma: relationship to disease severity. Arch. Med. Res. 43, 36–41. Saenz, L., Lozano, J.J., Valdor, R., Baroja-Mazo, A., Ramirez, P., Parrilla, P., et al. (2008). Transcriptional regulation by Poly(ADP-ribose) polymerase-1 during T cell activation. BMC Genomics 9, 171.   107   Schleimer, R.P., Kato, A., Kern, R., Kuperman, D., and Avila, P.C. (2007). Epithelium: at the interface of innate and adaptive immune responses. J. Allergy Clin. Immunol. 120, 1279–1284. Schneider, D., Hong, J.Y., Popova, A.P., Bowman, E.R., Linn, M.J., McLean, A.M., et al. (2012). Neonatal rhinovirus infection induces mucous metaplasia and airways hyperresponsiveness. J. Immunol. Baltim. Md 1950 188, 2894–2904. Schneider, D., Hong, J.Y., Bowman, E.R., Chung, Y., Nagarkar, D.R., McHenry, C.L., et al. (2013). Macrophage/epithelial cell CCL2 contributes to rhinovirus-induced hyperresponsiveness and inflammation in a mouse model of allergic airways disease. Am. J. Physiol. Lung Cell. Mol. Physiol. 304, L162–169. Schultz, E.D., Potts, E.N., Mason, S.N., Foster, W.M., and Auten, R.L. (2010). Mast cells mediate hyperoxia-induced airway hyper-reactivity in newborn rats. Pediatr. Res. 68, 70–74. Schwartz, N., Grossman, A., Levy, Y., and Schwarz, Y. (2012). Correlation between eosinophil count and methacholine challenge test in asymptomatic subjects. J. Asthma 49, 336–341. Sears, M.R. (2008). Epidemiology of asthma exacerbations. J. Allergy Clin. Immunol. 122, 662–668. Shore, S.A., and Shapiro, S.D. (2009). Chapter 8 - Asthma and COPD: Animal Models. In Asthma and COPD (Second Edition), (Oxford: Academic Press), pp. 99–109. Siegle, J.S., Hansbro, N., Herbert, C., Yang, M., Foster, P.S., and Kumar, R.K. (2006). Airway hyperreactivity in exacerbation of chronic asthma is independent of eosinophilic inflammation. Am. J. Respir. Cell Mol. Biol. 35, 565–570. Sigurs, N., Bjarnason, R., Sigurbergsson, F., and Kjellman, B. (2000). Respiratory syncytial virus bronchiolitis in infancy is an important risk factor for asthma and allergy at age 7. Am. J. Respir. Crit. Care Med. 161, 1501–1507. Skobeloff, E.M., Spivey, W.H., St Clair, S.S., and Schoffstall, J.M. (1992). The influence of age and sex on asthma admissions. JAMA J. Am. Med. Assoc. 268, 3437–3440. Soto-Quiros, M., Avila, L., Platts-Mills, T.A.E., Hunt, J.F., Erdman, D.D., Carper, H., et al. (2012). High titers of IgE antibody to dust mite allergen and risk for wheezing among asthmatic children infected with rhinovirus. J. Allergy Clin. Immunol. 129, 1499–1505.e5.   108   Spurrell, J.C.L., Wiehler, S., Zaheer, R.S., Sanders, S.P., and Proud, D. (2005). Human airway epithelial cells produce IP-10 (CXCL10) in vitro and in vivo upon rhinovirus infection. Am. J. Physiol. - Lung Cell. Mol. Physiol. 289, L85–L95. Stevens, W., Addo-Yobo, E., Roper, J., Woodcock, A., James, H., Platts-Mills, T., et al. (2011). Differences in both prevalence and titre of specific immunoglobulin E among children with asthma in affluent and poor communities within a large town in Ghana. Clin. Exp. Allergy 41, 1587–1594. Stone, K.D., Prussin, C., and Metcalfe, D.D. (2010). IgE, mast cells, basophils, and eosinophils. J. Allergy Clin. Immunol. 125, S73–80. Strachan, D.P. (2000). Family size, infection and atopy: the first decade of the “hygiene hypothesis.”Thorax 55, S2–10. Summers, C., Rankin, S.M., Condliffe, A.M., Singh, N., Peters, A.M., and Chilvers, E.R. (2010). Neutrophil kinetics in health and disease. Trends Immunol. 31, 318–324. Sykes, A., Macintyre, J., Edwards, M.R., Rosario, A. del, Haas, J., Gielen, V., et al. (2013a). Rhinovirus-induced interferon production is not deficient in well controlled asthma. Thorax thoraxjnl–2012–202909. Sykes, A., Edwards, M.R., Macintyre, J., Del Rosario, A., Gielen, V., Haas, J., et al. (2013b). TLR3, TLR4 and TLRs7-9 Induced Interferons Are Not Impaired in Airway and Blood Cells in Well Controlled Asthma. PloS One 8, e65921. Takaku, Y., Nakagome, K., Kobayashi, T., Hagiwara, K., Kanazawa, M., and Nagata, M. (2011). IFN-γ-inducible protein of 10 kDa upregulates the effector functions of eosinophils through β2 integrin and CXCR3. Respir. Res. 12, 138. Takatsu, K., and Nakajima, H. (2008). IL-5 and eosinophilia. Curr. Opin. Immunol. 20, 288–294. Tan, J., Jing, C., Jacobi, H.H., Reimert, C.M., Millner, A., Quan, S., et al. (2000). CXCR3 expression and activation of eosinophils: role of IFN-gamma-inducible protein-10 and monokine induced by IFN-gamma. J. Immunol. Baltim. Md 1950 165, 1548–1556. Thomson, N.C., Chaudhuri, R., and Livingston, E. (2004). Asthma and cigarette smoking. Eur. Respir. J. 24, 822–833. Thornton, D.J., and Sheehan, J.K. (2004). From mucins to mucus: toward a more coherent understanding of this essential barrier. Proc. Am. Thorac. Soc. 1, 54–61.   109   Thornton, D.J., Rousseau, K., and McGuckin, M.A. (2008). Structure and function of the polymeric mucins in airways mucus. Annu. Rev. Physiol. 70, 459–486. Tsuchiya, K., Siddiqui, S., Risse, P.-A., Hirota, N., and Martin, J.G. (2012). The presence of LPS in OVA inhalations affects airway inflammation and AHR but not remodeling in a rodent model of asthma. Am. J. Physiol. Lung Cell. Mol. Physiol. 303, L54–63. Tumas, D.B., Chan, B., Werther, W., Wrin, T., Vennari, J., Desjardin, N., et al. (2001). Anti-IgE efficacy in murine asthma models is dependent on the method of allergen sensitization. J. Allergy Clin. Immunol. 107, 1025–1033. Tuthill, T.J., Papadopoulos, N.G., Jourdan, P., Challinor, L.J., Sharp, N.A., Plumpton, C., et al. (2003). Mouse respiratory epithelial cells support efficient replication of human rhinovirus. J. Gen. Virol. 84, 2829–2836. Vercelli, D. (2008). Discovering susceptibility genes for asthma and allergy. Nat. Rev. Immunol. 8, 169–182. Voynow, J.A., and Rubin, B.K. (2009). Mucins, mucus, and sputum. Chest 135, 505–512. Wardlaw, A.J., Brightling, C., Green, R., Woltmann, G., and Pavord, I. (2000). Eosinophils in asthma and other allergic diseases. Br. Med. Bull. 56, 985–1003. Wark, P.A.B., Johnston, S.L., Simpson, J.L., Hensley, M.J., and Gibson, P.G. (2002a). Chlamydia pneumoniae immunoglobulin A reactivation and airway inflammation in acute asthma. Eur. Respir. J. 20, 834–840. Wark, P.A.B., Johnston, S.L., Moric, I., Simpson, J.L., Hensley, M.J., and Gibson, P.G. (2002b). Neutrophil degranulation and cell lysis is associated with clinical severity in virus-induced asthma. Eur. Respir. J. 19, 68–75. Wark, P.A.B., Johnston, S.L., Bucchieri, F., Powell, R., Puddicombe, S., Laza-Stanca, V., et al. (2005). Asthmatic bronchial epithelial cells have a deficient innate immune response to infection with rhinovirus. J. Exp. Med. 201, 937–947. Wark, P.A.B., Bucchieri, F., Johnston, S.L., Gibson, P.G., Hamilton, L., Mimica, J., et al. (2007). IFN-gamma-induced protein 10 is a novel biomarker of rhinovirus-induced asthma exacerbations. J. Allergy Clin. Immunol. 120, 586–593. Wenzel, S.E. (2001). The significance of the neutrophil in asthma. Clin. Exp. Allergy Rev. 1, 89–92.   110   Wenzel, S.E. (2006). Asthma: defining of the persistent adult phenotypes. The Lancet 368, 804–813. Wills-Karp, M., Luyimbazi, J., Xu, X., Schofield, B., Neben, T.Y., Karp, C.L., et al. (1998). Interleukin-13: central mediator of allergic asthma. Science 282, 2258–2261. Wu, C.A., Peluso, J.J., Shanley, J.D., Puddington, L., and Thrall, R.S. (2008). Murine Cytomegalovirus Influences Foxj1 Expression, Ciliogenesis, and Mucus Plugging in Mice with Allergic Airway Disease. Am. J. Pathol. 172, 714–724. Yadav, A., Saini, V., and Arora, S. (2010). MCP-1: chemoattractant with a role beyond immunity: a review. Clin. Chim. Acta 411, 1570– 1579. Yamada, Y., Matsumoto, K., Hashimoto, N., Saikusa, M., Homma, T., Yoshihara, S., et al. (2011). Effect of Th1/Th2 cytokine pretreatment on RSV-induced gene expression in airway epithelial cells. Int. Arch. Allergy Immunol. 154, 185–194. Yang, D., Chen, Q., Chertov, O., and Oppenheim, J.J. (2000). Human neutrophil defensins selectively chemoattract naive T and immature dendritic cells. J. Leukoc. Biol. 68, 9–14. Yang, M., Kumar, R.K., Hansbro, P.M., and Foster, P.S. (2012). Emerging roles of pulmonary macrophages in driving the development of severe asthma. J. Leukoc. Biol. 91, 557–569. You, D., Becnel, D., Wang, K., Ripple, M., Daly, M., and Cormier, S.A. (2006). Exposure of neonates to respiratory syncytial virus is critical in determining subsequent airway response in adults. Respir. Res. 7, 107. Yuta, A., Doyle, W.J., Gaumond, E., Ali, M., Tamarkin, L., Baraniuk, J.N., et al. (1998). Rhinovirus infection induces mucus hypersecretion. Am. J. Physiol. 274, L1017–1023. Zambrano, J.C., Carper, H.T., Rakes, G.P., Patrie, J., Murphy, D.D., Platts-Mills, T.A.E., et al. (2003). Experimental rhinovirus challenges in adults with mild asthma: Response to infection in relation to IgE. J. Allergy Clin. Immunol. 111, 1008–1016. Zedan, M., Attia, G., Zedan, M.M., Osman, A., Abo-Elkheir, N., Maysara, N., et al. (2013). Clinical Asthma Phenotypes and Therapeutic Responses. ISRN Pediatr. 2013, 1–7. Zhang, X., Zhang, Y., Tao, B., Wang, D., Cheng, H., Wang, K., et al. (2012). Docking protein Gab2 regulates mucin expression and goblet cell hyperplasia through TYK2/STAT6 pathway. FASEB J. 26, 4603– 4613.   111   Zhen, G., Park, S.W., Nguyenvu, L.T., Rodriguez, M.W., Barbeau, R., Paquet, A.C., et al. (2007). IL-13 and epidermal growth factor receptor have critical but distinct roles in epithelial cell mucin production. Am. J. Respir. Cell Mol. Biol. 36, 244–253. Zhou, J., Perelman, J.M., Kolosov, V.P., and Zhou, X. (2013). Neutrophil elastase induces MUC5AC secretion via protease-activated receptor 2. Mol. Cell. Biochem. 377, 75–85. Zietkowski, Z., Tomasiak-Lozowska, M.M., Skiepko, R., Zietkowska, E., and Bodzenta-Lukaszyk, A. (2010). Eotaxin-1 in exhaled breath condensate of stable and unstable asthma patients. Respir. Res. 11, 110. Zosky, G.R., and Sly, P.D. (2007). Animal models of asthma. Clin. Exp. Allergy 37, 973–988.   112   APPENDIX Sensitization solution per mouse OVA Al(OH)3 Saline 50 μg 2 mg 0.2ml Challenge solution per mouse OVA 1×PBS 25 μg 30 μl Modified Wrightʼs Stain (Liuʼs Stain) Liu A Eosin Y Methylene blue Methanol Liu B Methylene blue Azur I Na2HPO4·12H2O KH2PO4 H2O 0.18g 0.05 g 100 ml 0.7 g 0.6 g 12.6 g 6.25 g 500 ml Red blood cell lysis buffer for BAL fluid cell counts NH4Cl H2O 0.035g 4 ml Diluent buffer for BAL fluid cell counts RPMI BSA   4 ml 0.04g 113   [...]... Viruses and atypical bacteria associated with asthma exacerbations in hospitalized children Pediatr Pulmonol 45, 619–6 25 Mamessier, E., and Magnan, A (2006) Cytokines in atopic diseases: revisiting the Th2 dogma Eur J Dermatol EJD 16, 103–113 Martinez, F.O., Sica, A. , Mantovani, A. , and Locati, M (2008) Macrophage activation and polarization Front Biosci J Virtual Libr 13, 453 –461 Masoli, M., Fabian, D.,... allergen-sensitized and challenged mice induces eotaxin release from functionally polarized macrophages J Immunol Baltim Md 1 950 1 85, 252 5– 253 5 Nair, P., Gaga, M., Zervas, E., Alagha, K., Hargreave, F.E., OʼByrne, P.M., et al (2012) Safety and efficacy of a CXCR2 antagonist in patients with severe asthma and sputum neutrophils: a randomized, placebo-controlled clinical trial Clin Exp Allergy 42, 1097–1103 Nakanishi,... controlled asthma Thorax thoraxjnl–2012–202909 Sykes, A. , Edwards, M.R., Macintyre, J., Del Rosario, A. , Gielen, V., Haas, J., et al (2013b) TLR3, TLR4 and TLRs7-9 Induced Interferons Are Not Impaired in Airway and Blood Cells in Well Controlled Asthma PloS One 8, e 659 21 Takaku, Y., Nakagome, K., Kobayashi, T., Hagiwara, K., Kanazawa, M., and Nagata, M (2011) IFN-γ-inducible protein of 10 kDa upregulates... G., Wang, C., Huang, Y., Wang, H., et al (2009) RNA interference against interleukin -5 attenuates airway inflammation and hyperresponsiveness in an asthma model J Zhejiang Univ Sci B 10, 22–28 Cheng, C., Ho, W.E., Goh, F.Y., Guan, S.P., Kong, L.R., Lai, W.-Q., et al (2011) Anti-malarial drug artesunate attenuates experimental allergic asthma via inhibition of the phosphoinositide 3-kinase/Akt pathway... exposure impairs immune responses to bacterial products in human alveolar macrophages Thorax 63, 51 9– 52 5 Pantano, C., Ather, J.L., Alcorn, J.F., Poynter, M.E., Brown, A. L., Guala, A. S., et al (2008) Nuclear factor-kappaB Activation in Airway   1 05   Epithelium Induces Inflammation and Hyperresponsiveness Am J Respir Crit Care Med 177, 959 –969 Papadopoulos, N.G., Bates, P.J., Bardin, P.G., Papi, A. , Leir,... development of persistent asthma J Allergy Clin Immunol 119, 11 05 1110 Kuyper, L.M., Paré, P.D., Hogg, J.C., Lambert, R.K., Ionescu, D., Woods, R., et al (2003) Characterization of airway plugging in fatal asthma Am J Med 1 15, 6–11 Lambert, H.P., and Stern, H (1972) Infective factors in exacerbations of bronchitis and asthma Br Med J 3, 323–327 Lambrecht, B.N., and Hammad, H (2012) The airway epithelium in asthma. .. human bronchial tissue and epithelial cells by rhinovirus and lipopolysaccharide Acta Pharmacol Sin 25, 1176–1181 Heikkinen, T., and Järvinen, A (2003) The common cold The Lancet 361, 51 59 Hewson, C .A. , Haas, J.J., Bartlett, N.W., Message, S.D., Laza-Stanca, V., Kebadze, T., et al (2010) Rhinovirus induces MUC5AC in a human infection model and in vitro via NF-κB and EGFR pathways Eur Respir 36, 14 25 14 35. .. Crit Care Med 168, 790–797 Medoff, B.D., Sauty, A. , Tager, A. M., Maclean, J .A. , Smith, R.N., Mathew, A. , et al (2002) IFN-gamma-inducible protein 10 (CXCL10) contributes to airway hyperreactivity and airway inflammation in a mouse model of asthma J Immunol Baltim Md 1 950 168, 52 78 52 86 Melgert, B.N., Postma, D.S., Kuipers, I., Geerlings, M., Luinge, M .A. , van der Strate, B.W .A. , et al (20 05) Female mice... Relationship of upper and lower airway cytokines to outcome of experimental rhinovirus infection Am J Respir Crit Care Med 162, 2226–2231 Global Initiative for Asthma (GINA) (2011) Global strategy for asthma management and prevention (updated 2011)   96   Gordon, S (2003) Alternative activation of macrophages Nat Rev Immunol 3, 23– 35 Gordon, S., and Taylor, P.R (20 05) Monocyte and macrophage heterogeneity Nat...Bartlett, N.W., Walton, R.P., Edwards, M.R., Aniscenko, J., Caramori, G., Zhu, J., et al (2008) Mouse models of rhinovirus- induced disease and exacerbation of allergic airway inflammation Nat Med 14, 199–204 Bel, E.H (2004) Clinical phenotypes of asthma Curr Opin Pulm Med 10, 44 50 Benoit, L .A. , and Holtzman, M.J (2010) New immune pathways from chronic post-viral lung disease Ann N Acad Sci 1183, ... functionally polarized macrophages J Immunol Baltim Md 1 950 1 85, 252 5– 253 5 Nair, P., Gaga, M., Zervas, E., Alagha, K., Hargreave, F.E., OʼByrne, P.M., et al (2012) Safety and efficacy of a CXCR2 antagonist... point of   86   inflammation peak and exacerbation Our model induced a fast eosinophilia exacerbation at day after last challenge, which is prior to the day peak in Bartlettʼs model Meanwhile,... Airway and Blood Cells in Well Controlled Asthma PloS One 8, e 659 21 Takaku, Y., Nakagome, K., Kobayashi, T., Hagiwara, K., Kanazawa, M., and Nagata, M (2011) IFN-γ-inducible protein of 10 kDa

Ngày đăng: 01/10/2015, 17:26

Tài liệu cùng người dùng

  • Đang cập nhật ...

Tài liệu liên quan