1887

Abstract

In order to gain further insight into the early virus–host interactions associated with highly pathogenic avian influenza virus infections in chickens, genome-wide expression profiling of chicken lung and brain was carried out at 24 and 72 h post-inoculation (h p.i.). For this purpose two recombinant H5N3 viruses were utilized, each possessing a polybasic HA cleavage site but differing in pathogenicity. The original rH5N3 P0 virus, which has a low-pathogenic phenotype, was passaged six times through chickens to give rise to the derivative rH5N3 P6 virus, which is highly pathogenic (Diederich S, Berhane Y, Embury-Hyatt C, Hisanaga T, Handel K 2015;89:10724–10734). The gene-expression profiles in lung were similar for both viruses, although they varied in magnitude. While both viruses produced systemic infections, differences in clinical disease progression and viral tissue loads, particularly in brain, where loads of rH5N3 P6 were three orders of magnitude higher than rH5N3 P0 at 72 .p.i., were observed. Although genes associated with gene ontology (GO) categories INFα and INFβ biosynthesis, regulation of innate immune response, response to exogenous dsRNA, defence response to virus, positive regulation of NF-κB import into the nucleus and positive regulation of immune response were up-regulated in rH5N3 P0 and rH5N3 P6 brains, fold changes were higher for rH5N3 P6. The additional up-regulation of genes associated with cytokine production, inflammasome and leukocyte activation, and cell–cell adhesion detected in rH5N3 P6 versus rH5N3 P0 brains, suggested that the balance between antiviral and pro-inflammatory innate immune responses leading to acute CNS inflammation might explain the observed differences in pathogenicity.

Loading

Article metrics loading...

/content/journal/jgv/10.1099/jgv.0.000801
2017-06-01
2024-03-19
Loading full text...

Full text loading...

/deliver/fulltext/jgv/98/6/1245.html?itemId=/content/journal/jgv/10.1099/jgv.0.000801&mimeType=html&fmt=ahah

References

  1. Diederich S, Berhane Y, Embury-Hyatt C, Hisanaga T, Handel K et al. Hemagglutinin-neuraminidase balance influences the virulence phenotype of a recombinant H5N3 influenza A virus possessing a polybasic HA0 cleavage site. J Virol 2015; 89:10724–10734 [View Article][PubMed]
    [Google Scholar]
  2. Kuribayashi S, Sakoda Y, Kawasaki T, Tanaka T, Yamamoto N et al. Excessive cytokine response to rapid proliferation of highly pathogenic avian influenza viruses leads to fatal systemic capillary leakage in chickens. PLoS One 2013; 8:e68375 [View Article][PubMed]
    [Google Scholar]
  3. Ranaware PB, Mishra A, Vijayakumar P, Gandhale PN, Kumar H et al. Genome wide host gene expression analysis in chicken lungs infected with avian influenza viruses. PLoS One 2016; 11:e0153671 [View Article][PubMed]
    [Google Scholar]
  4. Rebel JM, Peeters B, Fijten H, Post J, Cornelissen J et al. Highly pathogenic or low pathogenic avian influenza virus subtype H7N1 infection in chicken lungs: small differences in general acute responses. Vet Res 2011; 42:10 [View Article][PubMed]
    [Google Scholar]
  5. Suzuki K, Okada H, Itoh T, Tada T, Mase M et al. Association of increased pathogenicity of Asian H5N1 highly pathogenic avian influenza viruses in chickens with highly efficient viral replication accompanied by early destruction of innate immune responses. J Virol 2009; 83:7475–7486 [View Article][PubMed]
    [Google Scholar]
  6. Uchida Y, Watanabe C, Takemae N, Hayashi T, Oka T et al. Identification of host genes linked with the survivability of chickens infected with recombinant viruses possessing H5N1 surface antigens from a highly pathogenic avian influenza virus. J Virol 2012; 86:2686–2695 [View Article][PubMed]
    [Google Scholar]
  7. Gottlieb TA, Ivanov IE, Adesnik M, Sabatini DD. Actin microfilaments play a critical role in endocytosis at the apical but not the basolateral surface of polarized epithelial cells. J Cell Biol 1993; 120:695–710 [View Article][PubMed]
    [Google Scholar]
  8. Nayak DP, Balogun RA, Yamada H, Zhou ZH, Barman S. Influenza virus morphogenesis and budding. Virus Res 2009; 143:147–161 [View Article][PubMed]
    [Google Scholar]
  9. Ohkura T, Momose F, Ichikawa R, Takeuchi K, Morikawa Y. Influenza A virus hemagglutinin and neuraminidase mutually accelerate their apical targeting through clustering of lipid rafts. J Virol 2014; 88:10039–10055 [View Article][PubMed]
    [Google Scholar]
  10. Sun X, Whittaker GR. Role of the actin cytoskeleton during influenza virus internalization into polarized epithelial cells. Cell Microbiol 2007; 9:1672–1682 [View Article][PubMed]
    [Google Scholar]
  11. Elo HA, Kulomaa MS, Tuohimaa PJ. Progesterone-independent avidin induction in chick tissues caused by tissue injury and inflammation. Acta Endocrinol 1979; 90:743–752 [View Article][PubMed]
    [Google Scholar]
  12. Tuohimaa P, Joensuu T, Isola J, Keinänen R, Kunnas T et al. Development of progestin-specific response in the chicken oviduct. Int J Dev Biol 1989; 33:125–134[PubMed]
    [Google Scholar]
  13. Matulova M, Rajova J, Vlasatikova L, Volf J, Stepanova H et al. Characterization of chicken spleen transcriptome after infection with Salmonella enterica serovar enteritidis. PLoS One 2012; 7:e48101 [View Article][PubMed]
    [Google Scholar]
  14. Rychlik I, Elsheimer-Matulova M, Kyrova K. Gene expression in the chicken caecum in response to infections with non-typhoid Salmonella. Vet Res 2014; 45:119 [View Article][PubMed]
    [Google Scholar]
  15. Korpela J. Chicken macrophages synthesize and secrete avidin in culture. Eur J Cell Biol 1984; 33:105–111[PubMed]
    [Google Scholar]
  16. Haller O, Staeheli P, Schwemmle M, Kochs G. Mx GTPases: dynamin-like antiviral machines of innate immunity. Trends Microbiol 2015; 23:154–163 [View Article][PubMed]
    [Google Scholar]
  17. Verhelst J, Parthoens E, Schepens B, Fiers W, Saelens X. Interferon-inducible protein Mx1 inhibits influenza virus by interfering with functional viral ribonucleoprotein complex assembly. J Virol 2012; 86:13445–13455 [View Article][PubMed]
    [Google Scholar]
  18. Ewald SJ, Kapczynski DR, Livant EJ, Suarez DL, Ralph J et al. Association of Mx1 Asn631 variant alleles with reductions in morbidity, early mortality, viral shedding, and cytokine responses in chickens infected with a highly pathogenic avian influenza virus. Immunogenetics 2011; 63:363–375 [View Article][PubMed]
    [Google Scholar]
  19. Ko JH, Jin HK, Asano A, Takada A, Ninomiya A et al. Polymorphisms and the differential antiviral activity of the chicken Mx gene. Genome Res 2002; 12:595–601 [View Article][PubMed]
    [Google Scholar]
  20. Sironi L, Williams JL, Moreno-Martin AM, Ramelli P, Stella A et al. Susceptibility of different chicken lines to H7N1 highly pathogenic avian influenza virus and the role of Mx gene polymorphism coding amino acid position 631. Virology 2008; 380:152–156 [View Article][PubMed]
    [Google Scholar]
  21. Li XY, Qu LJ, Yao JF, Yang N. Skewed allele frequencies of an Mx gene mutation with potential resistance to avian influenza virus in different chicken populations. Poult Sci 2006; 85:1327–1329[PubMed] [CrossRef]
    [Google Scholar]
  22. Schusser B, Reuter A, von der Malsburg A, Penski N, Weigend S et al. Mx is dispensable for interferon-mediated resistance of chicken cells against influenza A virus. J Virol 2011; 85:8307–8315 [View Article][PubMed]
    [Google Scholar]
  23. Liniger M, Summerfield A, Zimmer G, Mccullough KC, Ruggli N. Chicken cells sense influenza A virus infection through MDA5 and CARDIF signaling involving LGP2. J Virol 2012; 86:705–717 [View Article][PubMed]
    [Google Scholar]
  24. Hayashi T, Watanabe C, Suzuki Y, Tanikawa T, Uchida Y et al. Chicken MDA5 senses short double-stranded RNA with implications for antiviral response against avian influenza viruses in chicken. J Innate Immun 2014; 6:58–71 [View Article][PubMed]
    [Google Scholar]
  25. Barber MR, Aldridge JR, Webster RG, Magor KE. Association of RIG-I with innate immunity of ducks to influenza. Proc Natl Acad Sci USA 2010; 107:5913–5918 [View Article][PubMed]
    [Google Scholar]
  26. Mibayashi M, Martínez-Sobrido L, Loo YM, Cárdenas WB, Gale M et al. Inhibition of retinoic acid-inducible gene I-mediated induction of beta interferon by the NS1 protein of influenza A virus. J Virol 2007; 81:514–524 [View Article][PubMed]
    [Google Scholar]
  27. Fitzgerald KA. The interferon inducible gene: viperin. J Interferon Cytokine Res 2011; 31:131–135 [View Article][PubMed]
    [Google Scholar]
  28. Goossens KE, Karpala AJ, Rohringer A, Ward A, Bean AG. Characterisation of chicken viperin. Mol Immunol 2015; 63:373–380 [View Article][PubMed]
    [Google Scholar]
  29. Severa M, Coccia EM, Fitzgerald KA. Toll-like receptor-dependent and -independent viperin gene expression and counter-regulation by PRDI-binding factor-1/BLIMP1. J Biol Chem 2006; 281:26188–26195 [View Article][PubMed]
    [Google Scholar]
  30. Oshiumi H, Mifsud EJ, Daito T. Links between recognition and degradation of cytoplasmic viral RNA in innate immune response. Rev Med Virol 2016; 26:90–101 [View Article][PubMed]
    [Google Scholar]
  31. Penski N, Härtle S, Rubbenstroth D, Krohmann C, Ruggli N et al. Highly pathogenic avian influenza viruses do not inhibit interferon synthesis in infected chickens but can override the interferon-induced antiviral state. J Virol 2011; 85:7730–7741 [View Article][PubMed]
    [Google Scholar]
  32. Hu J, Hu Z, Mo Y, Wu Q, Cui Z et al. The PA and HA gene-mediated high viral load and intense innate immune response in the brain contribute to the high pathogenicity of H5N1 avian influenza virus in mallard ducks. J Virol 2013; 87:11063–11075 [View Article][PubMed]
    [Google Scholar]
  33. Rue CA, Susta L, Cornax I, Brown CC, Kapczynski DR et al. Virulent Newcastle disease virus elicits a strong innate immune response in chickens. J Gen Virol 2011; 92:931–939 [View Article][PubMed]
    [Google Scholar]
  34. Kobayashi KS, van den Elsen PJ. NLRC5: a key regulator of MHC class I-dependent immune responses. Nat Rev Immunol 2012; 12:813–820 [View Article][PubMed]
    [Google Scholar]
  35. Chau BN, Cheng EH, Kerr DA, Hardwick JM. Aven, a novel inhibitor of caspase activation, binds Bcl-xL and Apaf-1. Mol Cell 2000; 6:31–40 [View Article][PubMed]
    [Google Scholar]
  36. Guo H, Callaway JB, Ting JP. Inflammasomes: mechanism of action, role in disease, and therapeutics. Nat Med 2015; 21:677–687 [View Article][PubMed]
    [Google Scholar]
  37. Göbel TW, Schneider K, Schaerer B, Mejri I, Puehler F et al. IL-18 stimulates the proliferation and IFN-gamma release of CD4+ T cells in the chicken: conservation of a Th1-like system in a nonmammalian species. J Immunol 2003; 171:1809–1815 [View Article][PubMed]
    [Google Scholar]
  38. Lee SM, Gardy JL, Cheung CY, Cheung TK, Hui KP et al. Systems-level comparison of host-responses elicited by avian H5N1 and seasonal H1N1 influenza viruses in primary human macrophages. PLoS One 2009; 4:e8072 [View Article][PubMed]
    [Google Scholar]
  39. Pauli EK, Schmolke M, Wolff T, Viemann D, Roth J et al. Influenza A virus inhibits type I IFN signaling via NF-kappaB-dependent induction of SOCS-3 expression. PLoS Pathog 2008; 4:e1000196 [View Article][PubMed]
    [Google Scholar]
  40. Pothlichet J, Chignard M, Si-Tahar M. Cutting edge: innate immune response triggered by influenza A virus is negatively regulated by SOCS1 and SOCS3 through a RIG-I/IFNAR1-dependent pathway. J Immunol 2008; 180:2034–2038 [View Article][PubMed]
    [Google Scholar]
  41. Vijayakumar P, Mishra A, Ranaware PB, Kolte AP, Kulkarni DD et al. Analysis of the crow lung transcriptome in response to infection with highly pathogenic H5N1 avian influenza virus. Gene 2015; 559:77–85 [View Article][PubMed]
    [Google Scholar]
  42. Lucas S-M, Rothwell NJ, Gibson RM. The role of inflammation in CNS injury and disease. Br J Pharmacol 2006; 147:S232–S240 [View Article]
    [Google Scholar]
  43. Spackman E, Senne DA, Myers TJ, Bulaga LL, Garber LP et al. Development of a real-time reverse transcriptase PCR assay for type A influenza virus and the avian H5 and H7 hemagglutinin subtypes. J Clin Microbiol 2002; 40:3256–3260 [View Article][PubMed]
    [Google Scholar]
  44. Yang M, Berhane Y, Salo T, Li M, Hole K et al. Development and application of monoclonal antibodies against avian influenza virus nucleoprotein. J Virol Methods 2008; 147:265–274 [View Article][PubMed]
    [Google Scholar]
  45. Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J Royal Stat Soc B 1995; 57:289–300
    [Google Scholar]
  46. Benjamin Y, Yekutieli D. The control of false discovery rate in multiple testing under dependency. Annals Stat 2001; 29:1165–1188 [CrossRef]
    [Google Scholar]
  47. Ashburner M, Ball CA, Blake JA, Botstein D, Butler H et al. Gene ontology: tool for the unification of biology. Nat Genet 2000; 25:25–29 [View Article]
    [Google Scholar]
  48. da Huang W, Sherman BT, Lempicki RA. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc 2009; 4:44–57 [View Article][PubMed]
    [Google Scholar]
http://instance.metastore.ingenta.com/content/journal/jgv/10.1099/jgv.0.000801
Loading
/content/journal/jgv/10.1099/jgv.0.000801
Loading

Data & Media loading...

Supplements

Supplementary File 1

PDF
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error