Allergy Asthma Immunol Res.  2015 Nov;7(6):525-533. 10.4168/aair.2015.7.6.525.

Models of Respiratory Infections: Virus-Induced Asthma Exacerbations and Beyond

Affiliations
  • 1Section of Respiratory Medicine, University of Ferrara, Ferrara, Italy. ppa@unife.it
  • 2Department of Respiratory Diseases, Fondazione Maugeri, Tradate, University of Varese, Italy.

Abstract

Respiratory infections are one of the main health problems worldwide. They are a challenging field of study due to an intricate relationship between the pathogenicity of microbes and the host's defenses. To better understand mechanisms of respiratory infections, different models have been developed. A model is the reproduction of a disease in a system that mimics human pathophysiology. For this reason, the best models should closely resemble real-life conditions. Thus, the human model is the best. However, human models of respiratory infections have some disadvantages that limit their role. Therefore, other models, including animal, in vitro, and mathematical ones, have been developed. We will discuss advantages and limitations of available models and focus on models of viral infections as triggers of asthma exacerbations, viral infections being one of the most frequent causes of exacerbating disease. Future studies should focus on the interrelation of various models.

Keyword

Respiratory tract infections; theoretical models; animal models; in vitro diagnostic devices; mathematical computing

MeSH Terms

Animals
Asthma*
Humans
Mathematical Computing
Models, Animal
Models, Theoretical
Reagent Kits, Diagnostic
Reproduction
Respiratory Tract Infections*
Virulence
Reagent Kits, Diagnostic

Reference

1. DeVincenzo JP, Wilkinson T, Vaishnaw A, Cehelsky J, Meyers R, Nochur S, et al. Viral load drives disease in humans experimentally infected with respiratory syncytial virus. Am J Respir Crit Care Med. 2010; 182:1305–1314.
2. You SY, Jwa HJ, Yang EA, Kil HR, Lee JH. Effects of methylprednisolone pulse therapy on refractory Mycoplasma pneumoniae pneumonia in children. Allergy Asthma Immunol Res. 2014; 6:22–26.
3. Mallia P, Contoli M, Caramori G, Pandit A, Johnston SL, Papi A. Exacerbations of asthma and chronic obstructive pulmonary disease (COPD): focus on virus induced exacerbations. Curr Pharm Des. 2007; 13:73–97.
4. Johnston SL, Pattemore PK, Sanderson G, Smith S, Lampe F, Josephs L, et al. Community study of role of viral infections in exacerbations of asthma in 9-11 year old children. BMJ. 1995; 310:1225–1229.
5. Nicholson KG, Kent J, Ireland DC. Respiratory viruses and exacerbations of asthma in adults. BMJ. 1993; 307:982–986.
6. Papadopoulos NG, Johnston SL. Rhinoviruses as pathogens of the lower respiratory tract. Can Respir J. 2000; 7:409–414.
7. Papadopoulos NG, Psarras S. Rhinoviruses in the pathogenesis of asthma. Curr Allergy Asthma Rep. 2003; 3:137–145.
8. Message SD, Laza-Stanca V, Mallia P, Parker HL, Zhu J, Kebadze T, et al. 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. 2008; 105:13562–13567.
9. Fraenkel DJ, Bardin PG, Sanderson G, Lampe F, Johnston SL, Holgate ST. Lower airways inflammation during rhinovirus colds in normal and in asthmatic subjects. Am J Respir Crit Care Med. 1995; 151:879–886.
10. Zhu J, Message SD, Qiu Y, Mallia P, Kebadze T, Contoli M, et al. Airway inflammation and illness severity in response to experimental rhinovirus infection in asthma. Chest. 2014; 145:1219–1229.
11. Mallia P, Message SD, Gielen V, Contoli M, Gray K, Kebadze T, et al. Experimental rhinovirus infection as a human model of chronic obstructive pulmonary disease exacerbation. Am J Respir Crit Care Med. 2011; 183:734–742.
12. Bem RA, Domachowske JB, Rosenberg HF. Animal models of human respiratory syncytial virus disease. Am J Physiol Lung Cell Mol Physiol. 2011; 301:L148–L156.
13. Acosta A, Norazmi MN, Hernandez-Pando R, Alvarez N, Borrero R, Infante JF, et al. The importance of animal models in tuberculosis vaccine development. Malays J Med Sci. 2011; 18:5–12.
14. Bartlett NW, Walton RP, Edwards MR, Aniscenko J, Caramori G, Zhu J, et al. Mouse models of rhinovirus-induced disease and exacerbation of allergic airway inflammation. Nat Med. 2008; 14:199–204.
15. Nagarkar DR, Bowman ER, Schneider D, Wang Q, Shim J, Zhao Y, et al. Rhinovirus infection of allergen-sensitized and -challenged mice induces eotaxin release from functionally polarized macrophages. J Immunol. 2010; 185:2525–2535.
16. Hong JY, Chung Y, Steenrod J, Chen Q, Lei J, Comstock AT, et al. Macrophage activation state determines the response to rhinovirus infection in a mouse model of allergic asthma. Respir Res. 2014; 15:63.
17. Mori H, Parker NS, Rodrigues D, Hulland K, Chappell D, Hincks JS, et al. Differences in respiratory syncytial virus and influenza infection in a house-dust-mite-induced asthma mouse model: consequences for steroid sensitivity. Clin Sci (Lond). 2013; 125:565–574.
18. Clarke DL, Davis NH, Majithiya JB, Piper SC, Lewis A, Sleeman MA, et al. Development of a mouse model mimicking key aspects of a viral asthma exacerbation. Clin Sci (Lond). 2014; 126:567–580.
19. Wright JL, Cosio M, Churg A. Animal models of chronic obstructive pulmonary disease. Am J Physiol Lung Cell Mol Physiol. 2008; 295:L1–L15.
20. Beasley V, Joshi PV, Singanayagam A, Molyneaux PL, Johnston SL, Mallia P. Lung microbiology and exacerbations in COPD. Int J Chron Obstruct Pulmon Dis. 2012; 7:555–569.
21. Churg A, Sin DD, Wright JL. Everything prevents emphysema: are animal models of cigarette smoke-induced chronic obstructive pulmonary disease any use? Am J Respir Cell Mol Biol. 2011; 45:1111–1115.
22. Sajjan U, Ganesan S, Comstock AT, Shim J, Wang Q, Nagarkar DR, et al. Elastase- and LPS-exposed mice display altered responses to rhinovirus infection. Am J Physiol Lung Cell Mol Physiol. 2009; 297:L931–L944.
23. Meshi B, Vitalis TZ, Ionescu D, Elliott WM, Liu C, Wang XD, et al. Emphysematous lung destruction by cigarette smoke. The effects of latent adenoviral infection on the lung inflammatory response. Am J Respir Cell Mol Biol. 2002; 26:52–57.
24. Drannik AG, Pouladi MA, Robbins CS, Goncharova SI, Kianpour S, Stämpfli MR. Impact of cigarette smoke on clearance and inflammation after Pseudomonas aeruginosa infection. Am J Respir Crit Care Med. 2004; 170:1164–1171.
25. Pang B, Hong W, West-Barnette SL, Kock ND, Swords WE. Diminished ICAM-1 expression and impaired pulmonary clearance of nontypeable Haemophilus influenzae in a mouse model of chronic obstructive pulmonary disease/emphysema. Infect Immun. 2008; 76:4959–4967.
26. Keller CE, Elliott TB, Bentzel DE, Mog SR, Shoemaker MO, Knudson GB. Susceptibility of irradiated B6D2F1/J mice to Klebsiella pneumoniae administered intratracheally: a pulmonary infection model in an immunocompromised host. Comp Med. 2003; 53:397–403.
27. Marquette CH, Wermert D, Wallet F, Copin MC, Tonnel AB. Characterization of an animal model of ventilator-acquired pneumonia. Chest. 1999; 115:200–209.
28. Garau J, Gomez L. Pseudomonas aeruginosa pneumonia. Curr Opin Infect Dis. 2003; 16:135–143.
29. Luna CM, Baquero S, Gando S, Patrón JR, Morato JG, Sibila O, et al. Experimental severe Pseudomonas aeruginosa pneumonia and antibiotic therapy in piglets receiving mechanical ventilation. Chest. 2007; 132:523–531.
30. Shultz LD, Brehm MA, Bavari S, Greiner DL. Humanized mice as a preclinical tool for infectious disease and biomedical research. Ann N Y Acad Sci. 2011; 1245:50–54.
31. Dupuit F, Gaillard D, Hinnrasky J, Mongodin E, de Bentzmann S, Copreni E, et al. Differentiated and functional human airway epithelium regeneration in tracheal xenografts. Am J Physiol Lung Cell Mol Physiol. 2000; 278:L165–L176.
32. Mongodin E, Bajolet O, Cutrona J, Bonnet N, Dupuit F, Puchelle E, et al. Fibronectin-binding proteins of Staphylococcus aureus are involved in adherence to human airway epithelium. Infect Immun. 2002; 70:620–630.
33. Brehm MA, Cuthbert A, Yang C, Miller DM, DiIorio P, Laning J, et al. Parameters for establishing humanized mouse models to study human immunity: analysis of human hematopoietic stem cell engraftment in three immunodeficient strains of mice bearing the IL2rgamma(null) mutation. Clin Immunol. 2010; 135:84–98.
34. Strowig T, Rongvaux A, Rathinam C, Takizawa H, Borsotti C, Philbrick W, et al. Transgenic expression of human signal regulatory protein alpha in Rag2-/-gamma(c)-/- mice improves engraftment of human hematopoietic cells in humanized mice. Proc Natl Acad Sci U S A. 2011; 108:13218–13223.
35. White RL. What in vitro models of infection can and cannot do. Pharmacotherapy. 2001; 21:292S–301S.
36. Terajima M, Yamaya M, Sekizawa K, Okinaga S, Suzuki T, Yamada N, et al. Rhinovirus infection of primary cultures of human tracheal epithelium: role of ICAM-1 and IL-1beta. Am J Physiol. 1997; 273:L749–L759.
37. Subauste MC, Jacoby DB, Richards SM, Proud D. Infection of a human respiratory epithelial cell line with rhinovirus. Induction of cytokine release and modulation of susceptibility to infection by cytokine exposure. J Clin Invest. 1995; 96:549–557.
38. Spannhake EW, Reddy SP, Jacoby DB, Yu XY, Saatian B, Tian J. Synergism between rhinovirus infection and oxidant pollutant exposure enhances airway epithelial cell cytokine production. Environ Health Perspect. 2002; 110:665–670.
39. Edwards MR, Johnson MW, Johnston SL. Combination therapy: synergistic suppression of virus-induced chemokines in airway epithelial cells. Am J Respir Cell Mol Biol. 2006; 34:616–624.
40. Papi A, Papadopoulos NG, Degitz K, Holgate ST, Johnston SL. Corticosteroids inhibit rhinovirus-induced intercellular adhesion molecule-1 up-regulation and promoter activation on respiratory epithelial cells. J Allergy Clin Immunol. 2000; 105:318–326.
41. Contoli M, Message SD, Laza-Stanca V, Edwards MR, Wark PA, Bartlett NW, et al. Role of deficient type III interferon-lambda production in asthma exacerbations. Nat Med. 2006; 12:1023–1026.
42. Papadopoulos NG, Stanciu LA, Papi A, Holgate ST, Johnston SL. A defective type 1 response to rhinovirus in atopic asthma. Thorax. 2002; 57:328–332.
43. Mathur SK, Fichtinger PS, Kelly JT, Lee WM, Gern JE, Jarjour NN. Interaction between allergy and innate immunity: model for eosinophil regulation of epithelial cell interferon expression. Ann Allergy Asthma Immunol. 2013; 111:25–31.
44. Shin SH, Ye MK, Kim JK. Effects of fungi and eosinophils on mucin gene expression in rhinovirus-infected nasal epithelial cells. Allergy Asthma Immunol Res. 2014; 6:149–155.
45. Papi A, Contoli M, Adcock IM, Bellettato C, Padovani A, Casolari P, et al. Rhinovirus infection causes steroid resistance in airway epithelium through nuclear factor κB and c-Jun N-terminal kinase activation. J Allergy Clin Immunol. 2013; 132:1075–1085.e6.
46. Contoli M, Ito K, Papi A. Effects of RV568, a narrow spectrum kinase inhibitor, on HRV16 replication and cytokine production in airway epithelial cells obtained from COPD patients. In : American Thoracic Society International Conference Abstracts of American Thoracic Society 2011 International Conference; 2011 May 13-18; Denver. New York (NY): American Thoracic Society;2011. p. A2766.
47. Contoli M, Ito K, Papi A. Effects of RV1088, a narrow spectrum kinase inhibitor, on HRV16 replication and cytokine production in airway epithelial cells obtained from COPD patients. In : American Thoracic Society International Conference Abstracts of American Thoracic Society 2011 International Conference; 2011 May 13-18; Denver. New York (NY): American Thoracic Society;2011. p. A3368.
48. Morehead RS, Pinto SJ. Ventilator-associated pneumonia. Arch Intern Med. 2000; 160:1926–1936.
49. Lyte M, Ernst S. Catecholamine induced growth of gram negative bacteria. Life Sci. 1992; 50:203–212.
50. Smythe MA, Melendy S, Jahns B, Dmuchowski C. An exploratory analysis of medication utilization in a medical intensive care unit. Crit Care Med. 1993; 21:1319–1323.
51. Freestone PP, Hirst RA, Sandrini SM, Sharaff F, Fry H, Hyman S, et al. Pseudomonas aeruginosa-catecholamine inotrope interactions: a contributory factor in the development of ventilator-associated pneumonia? Chest. 2012; 142:1200–1210.
52. European Agency for the Evaluation of Medicinal Products (GB). CPMP/EWP/2655/99. Points to Consider on Pharmacokinetics and Pharmacodynamics in the Development of Antibacterial Medicinal Products. http://www.ema.europa.eu/docs/en_GB/document_library/Scientific_guideline/2009/09/WC500003420.pdf.
53. Takahashi M, Washio T, Suzuki N, Igeta K, Yamashita S. The species differences of intestinal drug absorption and first-pass metabolism between cynomolgus monkeys and humans. J Pharm Sci. 2009; 98:4343–4353.
54. Craig WA. Pharmacokinetic/pharmacodynamic parameters: rationale for antibacterial dosing of mice and men. Clin Infect Dis. 1998; 26:1–10.
55. Gloede J, Scheerans C, Derendorf H, Kloft C. In vitro pharmacodynamic models to determine the effect of antibacterial drugs. J Antimicrob Chemother. 2010; 65:186–201.
56. Wang L, Wismer MK, Racine F, Conway D, Giacobbe RA, Berejnaia O, et al. Development of an integrated semi-automated system for in vitro pharmacodynamic modelling. J Antimicrob Chemother. 2008; 62:1070–1077.
57. Mackowiak PA, Marling-Cason M, Cohen RL. Effects of temperature on antimicrobial susceptibility of bacteria. J Infect Dis. 1982; 145:550–553.
58. Brown MR, Williams P. Influence of substrate limitation and growth phase on sensitivity to antimicrobial agents. J Antimicrob Chemother. 1985; 15:Suppl A. 7–14.
59. Tuomanen E, Cozens R, Tosch W, Zak O, Tomasz A. The rate of killing of Escherichia coli by beta-lactam antibiotics is strictly proportional to the rate of bacterial growth. J Gen Microbiol. 1986; 132:1297–1304.
60. Lorian V. In vitro simulation of in vivo conditions: physical state of the culture medium. J Clin Microbiol. 1989; 27:2403–2406.
61. Brook I. Inoculum effect. Rev Infect Dis. 1989; 11:361–368.
62. Czock D, Keller F. Mechanism-based pharmacokinetic-pharmacodynamic modeling of antimicrobial drug effects. J Pharmacokinet Pharmacodyn. 2007; 34:727–751.
63. Getto P, Kimmel M, Marciniak-Czochra A. Modelling and analysis of dynamics of viral infection of cells and of interferon resistance. J Math Anal Appl. 2008; 344:821–850.
64. Keeling MJ, Danon L. Mathematical modelling of infectious diseases. Br Med Bull. 2009; 92:33–42.
65. Moghadas SM. Gaining insights into human viral diseases through mathematics. Eur J Epidemiol. 2006; 21:337–342.
66. Yang W, Sun C, Arino J. Global analysis for a general epidemiological model with vaccination and varying population. J Math Anal Appl. 2010; 372:208–223.
67. Brass AL, Huang IC, Benita Y, John SP, Krishnan MN, Feeley EM, et al. The IFITM proteins mediate cellular resistance to influenza A H1N1 virus, West Nile virus, and dengue virus. Cell. 2009; 139:1243–1254.
68. Everitt AR, Clare S, Pertel T, John SP, Wash RS, Smith SE, et al. IFITM3 restricts the morbidity and mortality associated with influenza. Nature. 2012; 484:519–523.
69. Baraldo S, Contoli M, Bazzan E, Turato G, Padovani A, Marku B, et al. Deficient antiviral immune responses in childhood: distinct roles of atopy and asthma. J Allergy Clin Immunol. 2012; 130:1307–1314.
70. Barbato A, Turato G, Baraldo S, Bazzan E, Calabrese F, Panizzolo C, et al. Epithelial damage and angiogenesis in the airways of children with asthma. Am J Respir Crit Care Med. 2006; 174:975–981.
Full Text Links
  • AAIR
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr