Hierarchical effects of pro-inflammatory cytokines on the post-influenza susceptibility to pneumococcal coinfection

Stefanie Duvigneau, Niharika Sharma-Chawla, Alessandro Boianelli, Sabine Stegemann-Koniszewski, Van Kinh Nguyen, Dunja Bruder, Esteban A Hernandez-Vargas, Stefanie Duvigneau, Niharika Sharma-Chawla, Alessandro Boianelli, Sabine Stegemann-Koniszewski, Van Kinh Nguyen, Dunja Bruder, Esteban A Hernandez-Vargas

Abstract

In the course of influenza A virus (IAV) infections, a secondary bacterial infection frequently leads to serious respiratory conditions provoking high hospitalization and death tolls. Although abundant pro-inflammatory responses have been reported as key contributing factors for these severe dual infections, the relative contributions of cytokines remain largely unclear. In the current study, mathematical modelling based on murine experimental data dissects IFN-γ as a cytokine candidate responsible for impaired bacterial clearance, thereby promoting bacterial growth and systemic dissemination during acute IAV infection. We also found a time-dependent detrimental role of IL-6 in curtailing bacterial outgrowth which was not as distinct as for IFN-γ. Our numerical simulations suggested a detrimental effect of IFN-γ alone and in synergism with IL-6 but no conclusive pathogenic effect of IL-6 and TNF-α alone. This work provides a rationale to understand the potential impact of how to manipulate temporal immune components, facilitating the formulation of hypotheses about potential therapeutic strategies to treat coinfections.

Figures

Figure 1. Experimental scheme.
Figure 1. Experimental scheme.
(a) C57BL/6 J wildtype mice were intranasally infected with a sub-lethal dose of IAV (A/PR8/34) followed by bacterial infection with the S. pneumoniae strain T4 on day 7. Bronchoalveolar lavage (BAL), post-lavage lung and blood were collected at the indicated time points post secondary bacterial infection (hpi). (b) The infection groups were single viral infection (IAV), single bacterial infection (T4) and coinfection (IAV+T4). (c) The bacterial burden, viral titers and cytokine concentrations were determined as the experimental readouts.
Figure 2. Organ-wide bacterial burden in the…
Figure 2. Organ-wide bacterial burden in the single and coinfected animals.
Bacterial titers in single T4 infected and IAV+T4 coinfected mice were determined in (a) post-lavage lung, (b) BAL and (c) blood. All experiments were performed in groups of 4–7 WT C57BL/6 J mice, raw data can be found in the Supplementary Fig. S1. Statistical analysis was performed using the Mann-Whitney test. Asterisks indicate significant differences between single and coinfected mice: *p < 0.05; **p < 0.01.
Figure 3. Absolute numbers of alveolar macrophages…
Figure 3. Absolute numbers of alveolar macrophages (AM) in post-lavage lungs of single and coinfected animals.
Statistical analysis was performed using the Mann-Whitney test. Asterisks indicate significant differences between single and coinfected mice: **p 

Figure 4. Pro-inflammatory cytokine profiles of the…

Figure 4. Pro-inflammatory cytokine profiles of the BAL of coinfected and single T4 infected mice.

Figure 4. Pro-inflammatory cytokine profiles of the BAL of coinfected and single T4 infected mice.
Protein concentrations of (a) IFN-γ, (b) TNF-α, (c) IL-6 and (d) MCP-1 were determined in the BAL fluid at the indicated time points after secondary T4 infection on day 7 post IAV or single T4 infection on day 7 post PBS treatment. Raw data can be found in the Supplementary Fig. S2. Statistical analysis was performed using the Mann-Whitney test. Asterisks indicate significant differences between single and coinfected mice: *p < 0.05; **p < 0.01.

Figure 5. Simulations for the coinfection model…

Figure 5. Simulations for the coinfection model M6.

In silico neutralizations of the different pro-inflammatory…
Figure 5. Simulations for the coinfection model M6.
In silico neutralizations of the different pro-inflammatory responses in model M6 are presented in the panel (a). The time-dependent contributions of pro-inflammatory cytokines to the impairment of bacterial clearance by the function fx are depicted in the panel (b). When the mathematical function fx is 1 means that there is no impairment to the bacterial clearance.
Figure 4. Pro-inflammatory cytokine profiles of the…
Figure 4. Pro-inflammatory cytokine profiles of the BAL of coinfected and single T4 infected mice.
Protein concentrations of (a) IFN-γ, (b) TNF-α, (c) IL-6 and (d) MCP-1 were determined in the BAL fluid at the indicated time points after secondary T4 infection on day 7 post IAV or single T4 infection on day 7 post PBS treatment. Raw data can be found in the Supplementary Fig. S2. Statistical analysis was performed using the Mann-Whitney test. Asterisks indicate significant differences between single and coinfected mice: *p < 0.05; **p < 0.01.
Figure 5. Simulations for the coinfection model…
Figure 5. Simulations for the coinfection model M6.
In silico neutralizations of the different pro-inflammatory responses in model M6 are presented in the panel (a). The time-dependent contributions of pro-inflammatory cytokines to the impairment of bacterial clearance by the function fx are depicted in the panel (b). When the mathematical function fx is 1 means that there is no impairment to the bacterial clearance.

References

    1. Ballinger M. N. & Standiford T. J. Postinfluenza bacterial pneumonia: host defenses gone awry. Journal of interferon & cytokine research: the official journal of the International Society for Interferon and Cytokine Research 30, 643–652 (2010).
    1. Taubenberger J. K. & Morens D. M. The pathology of influenza virus infections. Annual review of Pathology 3, 499–522 (2008).
    1. McCullers J. A. & Rehg J. E. Lethal synergism between influenza virus and Streptococcus pneumoniae: characterization of a mouse model and the role of platelet-activating factor receptor. The Journal of Infectious Diseases 186, 341–350 (2002).
    1. McCullers J. A. Insights into the interaction between influenza virus and pneumococcus. Clinical Microbiology Reviews 19, 571–582 (2006).
    1. McCullers J. A. The co-pathogenesis of influenza viruses with bacteria in the lung. Nature Reviews Microbiology 12, 252–262 (2014).
    1. Ortqvist A., Hedlund J. & Kalin M. Streptococcus pneumoniae: Epidemiology, Risk Factors, and Clinical Features. Seminars in Respiratory and Critical Care Medicine 1, 563–574 (2005).
    1. Mina M. J., McCullers J. A. & Klugman K. P. Live attenuated influenza vaccine enhances colonization of Streptococcus pneumoniae and Staphylococcus aureus in mice. mBio 5, e01040-13- (2014).
    1. Pasman L. The complication of coinfection. Yale Journal of Biology and Medicine 85, 127–132 (2012).
    1. Gatanaga H., Yasuoka A., Kikuchi Y., Tachikawa N. & Oka S. Influence of Prior HIV-1 Infection on the Development of Chronic Hepatitis B Infection. European Journal of Clinical Microbiology & Infectious Diseases 19, 237–239 (2000).
    1. Goulding J. et al.. Lowering the threshold of lung innate immune cell activation alters susceptibility to secondary bacterial superinfection. Journal of Infectious Diseases 204, 1086–1094 (2011).
    1. Shahangian A. et al.. Type I IFNs mediate development of postinfluenza bacterial pneumonia in mice. Journal of Clinical Investigation 119, 1910–1920 (2009).
    1. Sun K. & Metzger D. W. Inhibition of pulmonary antibacterial defense by interferon-gamma during recovery from influenza infection. Nature Medicine 14, 558–564 (2008).
    1. Damjanovic D., Lai R., Jeyanathan M., Hogaboam C. M. & Xing Z. Marked improvement of severe lung immunopathology by influenza-associated pneumococcal superinfection requires the control of both bacterial replication and host immune responses. American Journal of Pathology 183, 868–880 (2013).
    1. Cauley L. S. & Vella A. T. Why is coinfection with influenza virus and bacteria so difficult to control? Discovery Medicine 19, 33–40 (2015).
    1. Marchuk G. I. Mathematical modelling of immune response in infectious diseases, vol. 395 (Springer Science & Business Media, 1997).
    1. Möhler L., Flockerzi D., Sann H. & Reichl U. Mathematical model of influenza A virus production in large-scale microcarrier culture. Biotechnology and Bioengineering 90, 46–58 (2005).
    1. Baccam P., Beauchemin C., Macken C. a., Hayden F. G. & Perelson A. S. Kinetics of influenza A virus infection in humans. Journal of Virology 80, 7590–7599 (2006).
    1. Beauchemin C. A. & Handel A. A review of mathematical models of influenza A infections within a host or cell culture: lessons learned and challenges ahead. BMC Public Health 11 Suppl 1, S7 (2011).
    1. Mitchell H. et al.. Higher level of replication efficiency of 2009 (H1N1) pandemic influenza virus than those of seasonal and avian strains: kinetics from epithelial cell culture and computational modeling. Journal of Virology 85, 1125–1135 (2011).
    1. Smith A. M., McCullers J. A. & Adler F. R. Mathematical model of a three-stage innate immune response to a pneumococcal lung infection. Journal of Theoretical Biology 276, 106–116 (2011).
    1. Boianelli A. et al.. Modeling Influenza Virus Infection: A Roadmap for Influenza Research Viruses, 7(10), 5274–5304 (2015).
    1. Hancioglu B., Swigon D. & Clermont G. A dynamical model of human immune response to influenza A virus infection. Journal of Theoretical Biology 246, 70–86 (2007).
    1. Lee H. Y. et al.. Simulation and prediction of the adaptive immune response to influenza A virus infection. Journal of Virology 83, 7151–7165 (2009).
    1. Saenz R. A. et al.. Dynamics of Influenza Virus Infection and Pathology. Journal of Virology 84, 3974–3983 (2010).
    1. Miao H. et al.. Quantifying the Early Immune Response and Adaptive Immune Response Kinetics in Mice Infected with Influenza A Virus. Journal of Virology 84, 6687–6698 (2010).
    1. Smith A. M. et al.. Effect of 1918 PB1-F2 expression on influenza A virus infection kinetics. PLoS Computational Biology 7, e1001081 (2011).
    1. Canini L. & Carrat F. Population modeling of influenza A/H1N1 virus kinetics and symptom dynamics. Journal of Virology 85, 2764–2770 (2011).
    1. Pawelek K. A. et al.. Modeling Within-Host Dynamics of Influenza Virus Infection Including Immune Responses. PLoS Comput Biol 8, e1002588 (2012).
    1. Hernandez-Vargas E. A. & Meyer-Hermann M. Innate Immune System Dynamics to Influenza Virus. In 8th IFAC Symposium on Biological and Medical Systems 45, 260–265 (2012).
    1. Dobrovolny H. M., Reddy M. B., Kamal M. a., Rayner C. R. & Beauchemin C. a. a. Assessing Mathematical Models of Influenza Infections Using Features of the Immune Response. PLoS ONE 8, e57088 (2013).
    1. Hernandez-Vargas E. A. et al.. Effects of aging on influenza virus infection dynamics. Journal of Virology 88, 4123–4131 (2014).
    1. Boianelli A., Sharma-Chawla N., Bruder D. & Hernandez-Vargas E. A. Oseltamivir PK/PD modeling and simulation to evaluate treatment strategies against influenza-pneumococcus coinfection. Frontiers in Cellular and Infection Microbiology 6, 60 (2016).
    1. Smith A. M. et al.. Kinetics of coinfection with influenza A virus and Streptococcus pneumoniae. PLoS Pathogens 9, e1003238 (2013).
    1. Stegemann S. et al.. Increased Susceptibility for Superinfection with Streptococcus pneumoniae during Influenza Virus Infection Is Not Caused by TLR7-Mediated Lymphopenia. PLoS ONE 4, e4840 (2009).
    1. Ke R., Lewin S. R., Elliott J. H. & Perelson A. S. Modeling the Effects of Vorinostat In Vivo Reveals both Transient and Delayed HIV Transcriptional Activation and Minimal Killing of Latently Infected Cells. PLOS Pathogens 11, e1005237 (2015).
    1. Ghoneim H. E., Thomas P. G. & McCullers J. A. Depletion of Alveolar Macrophages during Influenza Infection Facilitates Bacterial Superinfections. The Journal of Immunology 191, 1250–1259 (2013).
    1. Sharma-Chawla N. et al.. Influenza A virus infection predisposes hosts to secondary infection with different Streptococcus pneumoniae serotypes with similar outcome but serotype-specific manifestation. Infection and Immunity 00422-16 (2016).
    1. Szade A. et al.. Effect of crossing C57BL/6 and FVB mouse strains on basal cytokine expression. Mediators of Inflammation 2015 (2015).
    1. Hussell T. & Cavanagh M. M. The innate immune rheostat: influence on lung inflammatory disease and secondary bacterial pneumonia. Biochemical Society transactions 37, 811–813 (2009).
    1. McNamee L. A. & Harmsen A. G. Both influenza-induced neutrophil dysfunction and neutrophil-independent mechanisms contribute to increased susceptibility to a secondary Streptococcus pneumoniae infection. Infection and Immunity 74, 6707–6721 (2006).
    1. Didierlaurent A. et al.. Sustained desensitization to bacterial Toll-like receptor ligands after resolutionof respiratory influenza infection. The Journal of Experimental Medicine 205, 323–329 (2008).
    1. Van der Poll T. et al.. Interleukin-6 gene-deficient mice show impaired defense against pneumococcal pneumonia. The Journal of Infectious Diseases 176, 439–444 (1997).
    1. Norrby-Teglund A., Pauksens K., Norgren M. & Holm S. E. Correlation between serum TNF alpha and IL-6 levels and severity of group A streptococcal infections. Scandinavian Journal of Infectious Diseases 27, 125–130 (1995).
    1. Kirby A. C., Raynes J. G. & Kaye P. M. The role played by tumor necrosis factor during localized and systemic infection with Streptococcus pneumoniae. Journal of Infectious Diseases 191, 1538–1547 (2005).
    1. Ellis G. T. et al.. TRAIL + monocytes and monocyte-related cells cause lung damage and thereby increase susceptibility to influenza-Streptococcus pneumoniae coinfection. EMBO reports 16, 1203–1218 (2015).
    1. Hussell T., Pennycook A. & Openshaw P. J. M. Inhibition of tumor necrosis factor reduces the severity of virus-specific lung immunopathology. European Journal of Immunology 31, 2566–2573 (2001).
    1. La Gruta N. L., Kedzierska K., Stambas J. & Doherty P. C. A question of self-preservation: immunopathology in influenza virus infection. Immunology and Cell Biology 85, 85–92 (2007).
    1. Marcos-Ramiro B., Garcia-Weber D. & Millan J. TNF-induced endothelial barrier disruption: Beyond actin and Rho. Thrombosis and Haemostasis 112, 1088–1102 (2014).
    1. Wiley J. A., Cerwenka A., Harkema J. R., Dutton R. W. & Harmsen A. G. Production of interferon-gamma by influenza hemagglutinin-specific CD8 effector T cells influences the development of pulmonary immunopathology. The American Journal of Pathology 158, 119–130 (2001).
    1. Yamada M. et al.. Interferon-gamma production by neutrophils during bacterial pneumonia in mice. American Journal of Respiratory and Critical Care Medicine 183, 1391–1401 (2011).
    1. Gomez J. C. et al.. Mechanisms of interferon-gamma production by neutrophils and its function during streptococcus pneumoniae pneumonia. American Journal of Respiratory Cell and Molecular Biology 52, 349–364 (2015).
    1. Storn R. & Price K. Differential evolution–a simple and efficient heuristic for global optimization over continuous spaces. Journal of Global Optimization 341–359 (1997).
    1. Nguyen V. K., Binder S. C., Boianelli A., Meyer-Hermann M. & Hernandez-Vargas E. A. Ebola virus infection modeling and identifiability problems. Frontiers in Microbiology 6, 1–11 (2015).
    1. Burnham K. P. & Anderson D. R. Model selection and multimodel inference: a practical information-theoretic approach (Springer Science & Business Media, 2002).
    1. Davison A. C. & Hinkley D. V. Bootstrap Methods and Their Application (Cambridge Univ, 1997).
    1. Xue H., Miao H. & Wu H. Sieve estimation of constant and time-varying coefficients in nonlinear ordinary differential equation models by considering both numerical error and measurement error. The Annals of Statistics 38, 2351–2387 (2010).
    1. Jaafoura S. et al.. Progressive contraction of the latent HIV reservoir around a core of less-differentiated CD4 (+) memory T Cells. Nature Communications 5, 1–8 (2014).
    1. Xia X. & Moog C. Identifiability of nonlinear systems with application to HIV/AIDS models. IEEE Transactions on Automatic Control 48, 330–336 (2003).
    1. Wu H., Zhu H., Miao H. & Perelson A. S. Parameter identifiability and estimation of HIV/AIDS dynamic models. Bulletin of Mathematical Biology 70, 785–799 (2008).
    1. Miao H., Xia X., Perelson A. S. & Wu H. On Identifiability of Nonlinear ODE Models and Applications in Viral Dynamics. Plus Review 53, 3–39 (2011).
    1. Nguyen V.-K. & Hernandez-Vargas E. A. Identifiability Challenges in Mathematical Models of Viral Infectious Diseases. IFAC-PapersOnLine 48, 2–7 (2015).
    1. Raue A. et al.. Structural and practical identifiability analysis of partially observed dynamical models by exploiting the profile likelihood. Bioinformatics 25, 1923–1929 (2009).
    1. Scheller J., Chalaris A., Schmidt-Arras D. & Rose-John S. The pro- and anti-inflammatory properties of the cytokine interleukin-6. Biochimica et Biophysica Acta 1813, 878–888 (2011).

Source: PubMed

3
Předplatit