A defective type 1 response to rhinovirus in atopic asthma ========================================================== * N G Papadopoulos * L A Stanciu * A Papi * S T Holgate * S L Johnston ## Abstract Background: Rhinoviruses (RVs) are the most frequent precipitants of the common cold and asthma exacerbations, but little is known about the immune response to these viruses and its potential implications in the pathogenesis of asthma. Methods: Peripheral blood mononuclear cells (PBMC) from patients with atopic asthma and normal subjects were exposed to live or inactivated RV preparations. Levels of interferon (IFN)γ and interleukins IL-12, IL-10, IL-4, IL-5 and IL-13 were evaluated in the culture supernatants with specific immunoassays. Results: Exposure of PBMC to RVs induced the production of IFNγ, IL-12, IL-10, and IL-13. Cells from asthmatic subjects produced significantly lower levels of IFNγ and IL-12 and higher levels of IL-10 than normal subjects. IL-4 was induced only in the asthmatic group, while the IFNγ/IL-4 ratio was more than three times lower in the asthmatic group. Conclusions: This evidence suggests that the immune response to RVs is not uniquely of a type 1 phenotype, as previously suggested. The type 1 response is defective in atopic asthmatic individuals, with a shift towards a type 2 phenotype in a way similar, but not identical, to their aberrant response to allergens. A defective type 1 immune response to RVs may be implicated in the pathogenesis of virus induced exacerbations of asthma. * rhinovirus * asthma Asthma affects up to 30% of the population in westernised societies, with increasing morbidity and costs.1 Rhinoviruses (RVs), the major common cold viruses, are also the most common trigger of asthma exacerbations.2,3 However, the mechanisms by which RVs provoke asthma are not well understood. A number of studies have focused on the interaction of RVs with airway epithelium,4,5 demonstrating the induction of a local inflammatory response. In contrast, information on the immune response to these viruses is limited, though changes in peripheral blood leucocyte counts during and after in vivo RV infection6 suggest that a systemic immune response to RV does develop. The balance between type 1 and type 2 immune responses in CD4 and CD8 T lymphocytes7 and the proposed roles of type 2 responses in the pathogenesis of asthma8 and of type 1 responses in virus infections9 suggest that the nature of immune responses to RV infections in asthmatic and normal subjects may be a subject of major importance in the pathogenesis of virus induced asthma. Peripheral blood mononuclear cells (PBMC) are activated during RV infection, as indicated by increased production of IL-2 and IFNγ and enhanced NK cytotoxicity after in vitro mitogen stimulation.10 RV16 enters monocytes and airway macrophages in vitro and activates these cells without active replication of the virus,11 inducing non-specific activation of lymphocytes.12 Cytokine responses to RVs in tonsillar cells have been shown to be type 1-like, with large amounts of IL-2 and IFNγ and no IL-4 production.13 These findings are difficult to interpret in the context of allergic asthma, a disease paradigmally connected with type 2 responses. Type 2 responses have been implicated in the pathogenesis of respiratory syncytial virus (RSV) bronchiolitis,14 but responses to RSV have not been examined in asthma. We have previously reported that RV infections result in bronchial CD4 and CD8 lymphocyte and eosinophil infiltration in both normal and asthmatic subjects.15 Others have also reported eosinophil involvement in rhinovirus infections in atopic/asthmatic subjects.16,17 Our study showed persistent eosinophilia in asthmatic but not normal subjects 6–8 weeks after the RV infection.15 These findings led us to hypothesise that the more severe physiological and inflammatory response observed in asthmatic patients relative to normal subjects may result from differences in lymphocyte function, specifically an imbalance between type 1 and type 2 cytokines.18 The proof of this hypothesis would have major implications in our understanding of the pathogenesis of virus induced asthma. We therefore exposed PBMC from normal and atopic asthmatic subjects to RVs and assessed the secretion patterns of a panel of type 1 and type 2 cytokines. The specificity of these responses was evaluated by inactivating the virus or blocking its cellular receptor. ## METHODS ### Subjects Venous blood was drawn from seven subjects with a history of mild to moderate asthma diagnosed by a physician. All were atopic as shown by positive skin prick tests and/or specific IgE to one or more common environmental allergens and a raised total serum IgE (>80 IU/ml). Seven healthy non-atopic subjects were examined in parallel. There were three men and four women in each group and their age distribution was comparable (range 20–57 years). Bronchial provocation with histamine was used to confirm the presence of bronchial hyperresponsiveness in the asthmatic group and the lack of it in the normal subjects. Two asthmatic subjects were receiving low dose inhaled corticosteroids, while the rest of the subjects were not taking any medication at the time of the study. No concomitant diseases were present and no subject had experienced a cold during the previous month. Informed consent was obtained in all cases and the study was approved by the Southampton Hospitals joint ethics subcommittee. ### Virus preparations RV16, a major group subtype,15 was initially obtained from Drs W Busse and E Dick (Madison, WI, USA). RV30, which belongs to the minor RV group, was obtained from the MRC Common Cold Unit (Salisbury, UK). The identity of the viruses was confirmed by neutralisation with specific antisera (ATCC, Rockville, MD, USA). Viruses were propagated in large quantities in Ohio HeLa cells at 33°C in a humidified 5% CO2 incubator and stored at –70°C. For each experiment a new vial was rapidly thawed and used immediately. RV16 was purified for some experiments to remove soluble factors of HeLa cell origin. An equal volume of saturated ammonium sulphate was added to RV16 suspensions and the virus was precipitated by centrifugation. The precipitant was resuspended in PBS, dialysed overnight at 4°C over PBS using a 50 000 kD MCWO membrane (Sigma Chemical Co, Poole, UK), incubated for 1 hour with 50 μl/ml *Staphylococcus* protein A-Sepharose Fast Flow (Sigma), clarified, and stored at –70°C. To study the effect of virus viability and/or receptor binding on the cytokine responses, RV16 was inactivated19 either by (1) exposure to 58°C for 1 hour, (2) exposure to pH 3 for 1 hour at 4°C, or (3) preincubation with 1 mg/ml soluble intercellular adhesion molecule-1 (sICAM-1; kindly donated by P Esmon, Bayer, Berkeley, CA, USA) for 1 hour at room temperature. ### Experimental design PBMC were obtained from whole blood by standard Ficoll centrifugation. They were washed and resuspended in RPMI-1640 medium with Glutamax (Life Technologies, Uxbridge, UK) containing 2.5% FCS and 2.5% human AB serum, without antibiotics, at a final concentration of 2 × 106/ml in 24-well plates. Virus preparations were added at final concentrations ranging from 10 to 0.01 infectious units per cell (multiplicity of infection, MOI). After 1 hour of gentle shaking at room temperature, cultures were placed in a humidified 5% CO2 incubator at 33°C.20 Supernatants were harvested at various time points, clarified by centrifugation, and stored at –70°C until assayed. ### Cytokine assays Levels of IFNγ, IL-12, IL-10, and IL-5 were measured with paired antibodies from R&D Systems (Abingdon, UK) using the manufacturer's recommended concentrations and ELISA protocol. The sensitivity of the assays was 6 pg/ml for IFNγ, 7 pg/ml for IL-12 and IL-10, and 4 pg/ml for IL-5. Commercially available kits were used for the measurement of IL-4 and IL-13 (Biosource, Camarillo, CA, USA). The sensitivities of these assays were <0.27 pg/ml and 12 pg/ml, respectively. ### Statistical analysis Data are expressed as mean (SE). Testing for statistical significance in the time course and dose-response studies was undertaken by analysis of variance (ANOVA). Other comparisons of means were performed using a paired *t* test. ## RESULTS ### Cytokine production in PBMC inoculated with RV16 #### Rhinovirus induction of IFNγ, IL-10 and IL-12 production A dose dependent production of IFNγ, IL-10, and IL-12 in response to RV16 inoculation of PBMC was present in both normal and asthmatic subjects with an effective range of 0.1–10 MOI (fig 1). The induction was statistically significant at all concentrations above 0.1 MOI (p<0.05 versus non-infected controls in all cases). Maximal induction occurred with the highest dose studied (10 MOI) (IFNγ: normal 2500 (810) pg/ml, asthmatic 1717 (426) pg/ml; IL-12: normal 469 (72) pg/ml, asthmatic 337 (101) pg/ml; IL-10 normal 695 (277) pg/ml, asthmatic 1199 (442 pg/ml)). A concentration of 1 MOI also produced significant induction and was used in all subsequent experiments. ![ Figure 1 ](http://thorax.bmj.com/https://thorax.bmj.com/content/thoraxjnl/57/4/328/F1.medium.gif) [ Figure 1 ](http://thorax.bmj.com/content/57/4/328/F1) **Figure 1 ** Dose-response curves of (A) IFNγ, (B) IL-12, and (C) IL-10 production by PBMC in normal (closed symbols) and atopic asthmatic subjects (open symbols) 48 hours after exposure to 0–10 MOI of RV16. Values and error bars are mean (SE). A dose dependent increase in cytokine production was observed for all three cytokines in both groups. #### Time course of RV induced IFNγ, IL-10, and IL-12 production Production of IFNγ, IL-12, and IL-10 in response to RV16 was also time dependent (fig 2). Increasing concentrations of IFNγ and IL-10 were observed at 24 and 48 hours after inoculation, while IL-12 peaked at 24 hours but was still raised at 48 hours. Samples obtained 1 week after inoculation were also available in some experiments. IL-12 was not detectable in those samples, while further increased concentrations of IL-10 were present in the RV infected samples and significant amounts of IFNγ still remained (not shown), suggesting that RV mediated activation persisted in these cultures. ![ Figure 2 ](http://thorax.bmj.com/https://thorax.bmj.com/content/thoraxjnl/57/4/328/F2.medium.gif) [ Figure 2 ](http://thorax.bmj.com/content/57/4/328/F2) **Figure 2 ** Time course of (A) IFNγ, (B) IL-12, and (C) IL-10 production by PBMC of normal and atopic asthmatic subjects 6–48 hours after exposure to 1 MOI of RV16 or medium alone. C– = non-infected cultures from normal subjects, C+ = RV16 infected cultures from normal subjects, A– = non-infected cultures from subjects with atopic asthma, A+ = infected cultures from subjects with atopic asthma. Values are mean (SE). Analysis of variance (ANOVA) was used to compare within and between group values at each time point. Exposure to RV16 resulted in a significant induction of IFNγ, IL-12, and IL-10 production 24 hours after inoculation, increasing further at 48 hours for IFNγ and IL-10. Although this effect was observed in both normal subjects and those with atopic asthma, significant differences were observed between the groups with lower levels of IFNγ and IL-12 and higher levels of IL-10 in asthmatic patients than in normal subjects. **p<0.01, *p<0.05, #0.050.1 in all cases). Inoculations with RV30, a minor group serotype,21 at 1 MOI in parallel cultures resulted in IFNγ production at the same level as RV16 inoculation (372 (81) pg/ml, n=3, p>0.1), indicating that cytokine induction by PBMC in response to RV is not confined to a specific RV serotype. These results also exclude the possibility that the observed induction of IFNγ in response to RV16 was restricted through signalling via ICAM-1, the receptor of RV16, but not of RV30, as similar levels of induction were observed with both serotypes. ### Effect of virus inactivation and receptor blockade Exposure to high temperature or acid pH completely inactivated RV16, as assessed by titration assays on Ohio-HeLa cells (not shown). These pretreatments were also able to markedly inhibit RV16 mediated induction of IFNγ in PBMC (fig 4A), indicating that live virus was necessary to induce the major part of IFNγ production. In contrast, the same pretreatments resulted in only partial reduction of RV mediated IL-10 production, the reductions being non-significant for acid treatment and statistically marginal for heat inactivation (p=0.088, fig 4B). ![ Figure 4 ](http://thorax.bmj.com/https://thorax.bmj.com/content/thoraxjnl/57/4/328/F4.medium.gif) [ Figure 4 ](http://thorax.bmj.com/content/57/4/328/F4) **Figure 4 ** Effects of virus inactivation on production of (A) IFNγ and (B) IL-10 by PBMC exposed to RV16. To inactivate the virus RV16 was exposed to temperatures of 58°C, pH 3, or sICAM-1. IFNγ production was completely abolished by all inactivating treatments (p<0.05 *v* infectious virus preparation in all cases, ANOVA). Only small decreases in RV16 mediated IL-10 production were observed in the same cultures (temperature: 0.053.0.CO;2-D&link_type=DOI) [PubMed](http://thorax.bmj.com/lookup/external-ref?access_num=10223554&link_type=MED&atom=%2Fthoraxjnl%2F57%2F4%2F328.atom) [Web of Science](http://thorax.bmj.com/lookup/external-ref?access_num=000079757900016&link_type=ISI) 21. **Hofer F**, Gruenberger M, Kowalski H, *et al*. Members of the low density lipoprotein receptor family mediate cell entry of a minor-group common cold virus. Proc Natl Acad Sci USA1994;91:1839–42. [Abstract/FREE Full Text](http://thorax.bmj.com/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoicG5hcyI7czo1OiJyZXNpZCI7czo5OiI5MS81LzE4MzkiO3M6NDoiYXRvbSI7czoyNDoiL3Rob3JheGpubC81Ny80LzMyOC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 22. **Papadopoulos NG**, Johnston SL. The acute exacerbation of asthma: Pathogenesis. In: Holgate ST, Boushey HA, Fabbri LM, eds. *Difficult asthma*. London: Martin Dunitz, 1999:183–204. 23. **Levandowski R**, Horohov D. Rhinovirus induces natural killer-like cytotoxic cells and interferon alpha in mononuclear leukocytes. J Med Virol1991;35:116–20. [CrossRef](http://thorax.bmj.com/lookup/external-ref?access_num=10.1016/0166-0934(91)90092-E&link_type=DOI) [PubMed](http://thorax.bmj.com/lookup/external-ref?access_num=1662702&link_type=MED&atom=%2Fthoraxjnl%2F57%2F4%2F328.atom) 24. **Scott P**. IL-12: initiation cytokine for cell-mediated immunity (comment). Science1993;260:496–7. [FREE Full Text](http://thorax.bmj.com/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6MzoiUERGIjtzOjExOiJqb3VybmFsQ29kZSI7czozOiJzY2kiO3M6NToicmVzaWQiO3M6MTI6IjI2MC81MTA3LzQ5NiI7czo0OiJhdG9tIjtzOjI0OiIvdGhvcmF4am5sLzU3LzQvMzI4LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 25. **Orange JS**, Biron CA. An absolute and restricted requirement for IL-12 in natural killer cell IFN-gamma production and antiviral defense. Studies of natural killer and T cell responses in contrasting viral infections. J Immunol1996;156:1138–42. [Abstract](http://thorax.bmj.com/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6ODoiamltbXVub2wiO3M6NToicmVzaWQiO3M6MTA6IjE1Ni8zLzExMzgiO3M6NDoiYXRvbSI7czoyNDoiL3Rob3JheGpubC81Ny80LzMyOC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 26. **Cousens LP**, Peterson R, Hsu S, *et al*. Two roads diverged: interferon alpha/beta- and interleukin 12-mediated pathways in promoting T cell interferon gamma responses during viral infection. J Exp Med1999;189:1315–28. [Abstract/FREE Full Text](http://thorax.bmj.com/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamVtIjtzOjU6InJlc2lkIjtzOjEwOiIxODkvOC8xMzE1IjtzOjQ6ImF0b20iO3M6MjQ6Ii90aG9yYXhqbmwvNTcvNC8zMjguYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 27. **Borish L**, Rosenwasser L. TH1/TH2 lymphocytes: doubt some more. J Allergy Clin Immunol1997;99:161–4. [CrossRef](http://thorax.bmj.com/lookup/external-ref?access_num=10.1016/S0091-6749(97)70090-3&link_type=DOI) [PubMed](http://thorax.bmj.com/lookup/external-ref?access_num=9042039&link_type=MED&atom=%2Fthoraxjnl%2F57%2F4%2F328.atom) [Web of Science](http://thorax.bmj.com/lookup/external-ref?access_num=A1997WH83100002&link_type=ISI) 28. **Hussell T**, Spender LC, Georgiou A, *et al*. Th1 and Th2 cytokine induction in pulmonary T cells during infection with respiratory syncytial virus. J Gen Virol1996;77:2447–55. [Abstract/FREE Full Text](http://thorax.bmj.com/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoidmlyIjtzOjU6InJlc2lkIjtzOjEwOiI3Ny8xMC8yNDQ3IjtzOjQ6ImF0b20iO3M6MjQ6Ii90aG9yYXhqbmwvNTcvNC8zMjguYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 29. **Coyle AJ**, Erard F, Bertrand C, *et al*. Virus-specific CD8+ cells can switch to interleukin 5 production and induce airway eosinophilia. J Exp Med1995;181:1229–33. [Abstract/FREE Full Text](http://thorax.bmj.com/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamVtIjtzOjU6InJlc2lkIjtzOjEwOiIxODEvMy8xMjI5IjtzOjQ6ImF0b20iO3M6MjQ6Ii90aG9yYXhqbmwvNTcvNC8zMjguYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 30. **Zhu Z**, Homer RJ, Wang Z, *et al*. Pulmonary expression of interleukin-13 causes inflammation, mucus hypersecretion, subepithelial fibrosis, physiologic abnormalities, and eotaxin production. J Clin Invest1999;103:779–88. [CrossRef](http://thorax.bmj.com/lookup/external-ref?access_num=10.1172/JCI5909&link_type=DOI) [PubMed](http://thorax.bmj.com/lookup/external-ref?access_num=10079098&link_type=MED&atom=%2Fthoraxjnl%2F57%2F4%2F328.atom) [Web of Science](http://thorax.bmj.com/lookup/external-ref?access_num=000079203100004&link_type=ISI) 31. **Wills-Karp M**, Luyimbazi J, Xu X, *et al*. Interleukin-13: central mediator of allergic asthma. Science1998;282:2258–61. [Abstract/FREE Full Text](http://thorax.bmj.com/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Mzoic2NpIjtzOjU6InJlc2lkIjtzOjEzOiIyODIvNTM5Ny8yMjU4IjtzOjQ6ImF0b20iO3M6MjQ6Ii90aG9yYXhqbmwvNTcvNC8zMjguYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 32. **Panuska JR**, Merolla R, Rebert NA, *et al*. Respiratory syncytial virus induces interleukin-10 by human alveolar macrophages. Suppression of early cytokine production and implications for incomplete immunity. J Clin Invest1995;96:2445–53. 33. **McInnes E**, Collins RA, Taylor G. Cytokine expression in pulmonary and peripheral blood mononuclear cells from calves infected with bovine respiratory syncytial virus. Res Vet Sci1998;64:163–6. [CrossRef](http://thorax.bmj.com/lookup/external-ref?access_num=10.1016/S0034-5288(98)90013-3&link_type=DOI) [PubMed](http://thorax.bmj.com/lookup/external-ref?access_num=9625474&link_type=MED&atom=%2Fthoraxjnl%2F57%2F4%2F328.atom) 34. **Robinson DS**, Tsicopoulos A, Meng Q, *et al*. Increased interleukin-10 messenger RNA expression in atopic allergy and asthma. Am J Respir Cell Mol Biol1996;14:113–7. [CrossRef](http://thorax.bmj.com/lookup/external-ref?access_num=10.1165/ajrcmb.14.2.8630259&link_type=DOI) [PubMed](http://thorax.bmj.com/lookup/external-ref?access_num=8630259&link_type=MED&atom=%2Fthoraxjnl%2F57%2F4%2F328.atom) [Web of Science](http://thorax.bmj.com/lookup/external-ref?access_num=A1996TU36500001&link_type=ISI) 35. **Borish L**, Aarons A, Rumbyrt J, *et al*. Interleukin-10 regulation in normal subjects and patients with asthma. J Allergy Clin Immunol1996;97:1288–96. [CrossRef](http://thorax.bmj.com/lookup/external-ref?access_num=10.1016/S0091-6749(96)70197-5&link_type=DOI) [PubMed](http://thorax.bmj.com/lookup/external-ref?access_num=8648025&link_type=MED&atom=%2Fthoraxjnl%2F57%2F4%2F328.atom) [Web of Science](http://thorax.bmj.com/lookup/external-ref?access_num=A1996VD60700017&link_type=ISI) 36. **Crump C**, Arruda E, Hayden F. Comparative antirhinoviral activities of soluble intercellular adhesion molecule-1 (sICAM-1) and chimeric ICAM-1/immunoglobulin A molecule. Antimicrob Agents Chemother1994;38:1425–7. [Abstract/FREE Full Text](http://thorax.bmj.com/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiYWFjIjtzOjU6InJlc2lkIjtzOjk6IjM4LzYvMTQyNSI7czo0OiJhdG9tIjtzOjI0OiIvdGhvcmF4am5sLzU3LzQvMzI4LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 37. **Fischer JE**, Johnson JE, Kuli-Zade RK, *et al*. Overexpression of interleukin-4 delays virus clearance in mice infected with respiratory syncytial virus. J Virol1997;71:8672–7. [Abstract/FREE Full Text](http://thorax.bmj.com/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoianZpIjtzOjU6InJlc2lkIjtzOjEwOiI3MS8xMS84NjcyIjtzOjQ6ImF0b20iO3M6MjQ6Ii90aG9yYXhqbmwvNTcvNC8zMjguYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 38. **Johnston SL**, Papi A, Monick MM, *et al*. Rhinoviruses induce interleukin-8 mRNA and protein production in human monocytes. J Infect Dis1997;175:323–9. [Abstract/FREE Full Text](http://thorax.bmj.com/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoiamluZmRpcyI7czo1OiJyZXNpZCI7czo5OiIxNzUvMi8zMjMiO3M6NDoiYXRvbSI7czoyNDoiL3Rob3JheGpubC81Ny80LzMyOC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 39. **Gern JE**, Vrtis R, Grindle KA, *et al*. Relationship of upper and lower airway cytokines to outcome of experimental rhinovirus infection. Am J Respir Crit Care Med2000;162:2226–31. [CrossRef](http://thorax.bmj.com/lookup/external-ref?access_num=10.1164/ajrccm.162.6.2003019&link_type=DOI) [PubMed](http://thorax.bmj.com/lookup/external-ref?access_num=11112143&link_type=MED&atom=%2Fthoraxjnl%2F57%2F4%2F328.atom) [Web of Science](http://thorax.bmj.com/lookup/external-ref?access_num=000165794700045&link_type=ISI)