Skip directly to site content Skip directly to page options Skip directly to A-Z link Skip directly to A-Z link Skip directly to A-Z link
Volume 12, Number 1—January 2006
THEME ISSUE
Influenza
Pathogenesis

Antiviral Response in Pandemic Influenza Viruses

Author affiliation: *Mount Sinai School of Medicine, New York, New York, USA

Cite This Article

Abstract

The outcome of viral infections depends on a complex set of interactions between the viruses and their hosts. Particularly, viral infection triggers specific signaling programs within the infected cells that results in substantial changes in host gene expression. While some of these changes might be beneficial for viral replication, others represent the induction of a host antiviral response. In this respect, viruses have evolved genes that counteract this initial innate antiviral response. These viral-host interactions shape the subsequent phases of the disease and influence the adaptive immune response. In influenza viruses, the nonstructural protein 1 inhibits the interferon-mediated antiviral response. The regulatory activities of this viral protein play a major role in the pathogenicity of influenza virus and appear partially responsible for the ability of influenza viruses to infect multiple animal species, which likely contributes to the generation of new pandemic viruses in humans.

Coevolution of pathogens with their hosts has resulted in the shaping of the host immune system. A major component of this system is the innate immune response, which includes all the host barriers and responses with broad specificity against pathogens. The innate immune response not only represents the first barrier against infection but also provides the appropriate signals required for the subsequent adaptive cellular and humoral immune responses to develop. The type I interferon (IFNα/β) response constitutes a critical element of the innate immune system that is particularly important in the battle against viral pathogens. Secretion of IFNα/β results in the induction of a cellular antiviral response involving the transcriptional upregulation of >100 genes (1).

Despite the host's sophisticated immune system, viruses continue to successfully infect them and cause disease and, in some cases, death. The success of viruses is explained, at least in part, by the acquisition of viral genes during evolution that antagonize the host immune response. Viral-encoded IFNα/β antagonists are of particular interest, since they appear to be present in most animal viruses. We detail how influenza viruses evade the host innate immunity, with particular emphasis on the IFNα/β response, and the implications of this immune evasion in pandemic influenza.

IFNα/β Antiviral Response

Animal cells that sense viral infection respond almost immediately by synthesizing and secreting IFNα/β. The IFNα/β genes include IFNβ and many closely related IFNα genes. Most cells have intracellular sensors of viral products that, when activated, initiate a signaling cascade that results in transcriptional induction of the IFNβ gene. The nature of these sensors has remained unknown until recently, when 2 putative RNA helicases, RIG-I and MDA-5, were identified as sensors for viral dsRNA generated in the cytoplasm during viral infection (24). Binding to dsRNA by these proteins may result in initiation of helicase activity, concomitant with a conformational change that leads to recruiting additional cellular factors, including the recently identified IPS-1/MAVS protein (5,6). As a result, different cellular kinases, including the IRF3 kinases TBK1 and IKKε, become activated. Activated IRF3, together with NF-κB and AP-1, accumulate in the nucleus, bind to the IFNβ promoter, and stimulate transcription. While cytoplasmic viral dsRNA is one of the viral molecules that trigger this cascade, other viral products and other cellular sensor molecules also likely participate in the induction of IFNβ. IFNα/β induction is also stimulated by the presence of viral RNA and DNA in the endosome through the action of TLR3, TLR7, TLR8, or TLR9 (7). Different cell subtypes appear to employ different mechanisms to recognize viral products (8).

Once IFNα/β has been synthesized, it is secreted and binds to the IFNα/β receptor. All IFNαs and IFNβ bind to the same receptor and as a result, the cytoplasmic kinases JAK1 and TYK2 become activated and phosphorylate the STAT1 and STAT2 molecules. This process promotes generation of the ISGF3 transcription factor, a complex of STAT1, STAT2, and IRF9 that accumulates in the nucleus. Nuclear ISGF3 binds to promoters that contain interferon-stimulated response elements and stimulates the transcriptional induction of antiviral genes, including MxA, PKR, OAS, ADAR, PML, p56, and many others (9). These IFN-stimulated genes inhibit viral replication by many different mechanisms, including binding to viral nucleocapsids, translation inhibition, RNA degradation, RNA editing, and apoptosis induction. Moreover, secreted IFNα/β promotes the generation of robust cellular and humoral immunity (10,11). In general, the IFNα/β response has a complex regulation that involves positive and negative feed-back mechanisms, some of which are still unknown.

Nonstructural Protein 1 of Influenza Virus

Although IFNα/β was first described as a factor with antiviral activity secreted by cells treated with partially heat-inactivated influenza A viruses (12), it was also recognized early on that influenza viruses are poor IFNα/β inducers (13). This is because influenza viruses, like many other viruses, encode mechanisms to evade and antagonize the IFNα/β response (14). In the case of influenza A virus, this IFNα/β antagonistic function is encoded by the nonstructural protein 1 (NS1) gene.

NS1 of influenza A viruses is encoded by the unspliced mRNA derived from the shortest RNA segment of the 8 viral RNA segments. The protein is the most abundant nonstructural viral protein expressed in influenza A virus–infected cells. The development of reverse genetics techniques to manipulate the influenza virus genome made it possible to generate NS1 mutant viruses, including a recombinant influenza A virus lacking the NS1 gene (15). The NS1 knockout influenza A virus, delNS1, was replication defective in most cells and hosts, except for those lacking a functional IFNα/β system. Most remarkable, delNS1 virus was highly attenuated in mice but replicated and caused disease in STAT1 knockout mice, which lack one of the key transactivator molecules needed for the IFNα/β response (15). These results indicate that NS1 is required to overcome the IFNα/β response during influenza A virus infection.

The basis of the IFNα/β antagonistic properties of the NS1 of influenza A virus relies on its ability to prevent IFNβ synthesis; this explains the poor IFNβ–inducing properties of influenza A viruses (16,17). In the absence of NS1, influenza A virus becomes a high IFNα/β–inducing virus, and induction of high levels of IFNα/β results in inhibition of replication of delNS1 virus. NS1, by virtue of its dsRNA binding properties, is likely to sequester viral dsRNA produced during viral infection, which prevents recognition of this dangerous molecule by cellular sensors. This model of action is consistent with the ability of NS1 expression to prevent activation of transcription factors involved in the induction of IFNα/β synthesis, including IRF3 (16). Moreover, dsRNA binding is required for optimal inhibition of IFNβ production by NS1 (18). Similar results were obtained with the NS1 of influenza B virus (19,20). However, interactions of NS1 with cellular proteins also likely contribute to its IFNα/β antagonistic functions (21). NS1 of influenza A virus, but not of influenza B virus, inhibits cellular factors involved in mRNA processing (22,23); this function might also play a role in inhibiting IFNα/β production by influenza A virus (24). Finally, NS1 has also been shown to have IFNα/β inhibitory properties at a post-IFNα/β synthesis level. The NS1 of both influenza A and B viruses prevents the activation of the translation inhibitory and IFN inducible protein PKR (25,26); the NS1 of influenza B virus inhibits the activity of ISG15 (27), an IFN-inducible protein that enhances the IFN-mediated antiviral response.

Role of NS1 Gene

Influenza A viruses can infect many different animal species, such as different birds (e.g., waterfowl, chickens, turkeys), horses, pigs and humans, but also cross species, with avian strains infecting mammalian species, including humans. This property is especially critical during human pandemics that are characterized by novel antigenic determinants. These determinants derive from avian strains for which no immunity exists in most human population, which results in higher illness and death rates. The factors involved in the ability of an avian influenza virus strain, or of a reassortant virus containing avian antigenic determinants, to infect and propagate in humans are poorly understood; this lack of knowledge hampers our ability to predict the pandemic potential of avian influenza virus strains circulating in birds. Although the receptor specificity of the hemagglutinin protein is a factor that appears to be important for human adaptation of avian strains, other poorly understood factors also participate in this adaptation (28). With respect to NS1, viral strains from different animal hosts likely have NS1 genes adapted to antagonize the IFNα/β system of their specific host species. This was the case when the NS1 gene of the human influenza A virus that caused the 1918 H1N1 pandemic was compared with the NS1 gene of the mouse-adapted H1N1 influenza A virus strain WSN. Replacement of the NS1 gene of WSN virus with that of the 1918 virus resulted in an attenuated virus in mice, but this virus more efficiently inhibited the IFNα/β system in human cells (21,29). This inhibition might be explained by specific interactions of NS1 with host factors that have different sequences depending on the host, with NS1 of a mouse-adapted strain interacting better with murine factors than with human factors, and vice versa. If this is a general property of NS1 from different influenza virus strains, an avian strain would require adaptation of its NS1 gene to efficiently antagonize the human IFNα/β system. Alternatively, an avian strain would require acquisition by reassortment of an NS1 gene from a human strain to efficiently infect and propagate in humans.

Because mutations that affect NS1 function also have a profound effect on viral pathogenicity, highly pathogenic influenza virus strains may have an NS1 gene with particularly strong IFNα/β antagonistic properties. Moreover, the ability of NS1 to attenuate the activation of different transcription factors during viral infections has implications beyond the inhibition of IFNα/β synthesis. For instance, expression of many other cytokines and molecules involved in activation of dendritic cell function also appear to be regulated by NS1 (30). In this respect, the NS1 of the highly pathogenic avian H5N1 viruses circulating in poultry and waterfowl in Southeast Asia might be responsible for an enhanced proinflammatory cytokine response (especially TNFa) induced by these viruses in human macrophages (31,32). High levels of proinflammatory cytokines are likely to play an important role in the unusual lethality of these viruses in humans. Fortunately, infection with these viruses appears to be rare and the viruses have not been able to efficiently propagate from human to human.

Other Influenza Antagonists of Host Response

Although delNS1 influenza virus is a high inducer of IFNα/β, partial UV inactivation of this virus results in even higher induction of IFNα/β (33). These results suggest the presence of additional viral genes besides NS1 that attenuate IFNα/β production during viral infection and that become inactivated by UV. The viral polymerase, possibly through its endonuclease "cap-snatching" activity, might be responsible for this anti- IFNα/β activity (33). Further experimentation will be required to evaluate this hypothesis. In any case, the presence of multiple viral genes that cooperatively antagonize the IFNα/β response is not uncommon among the different virus families.

Influenza A virus encodes a second nonstructural polypeptide in virus-infected cells, the PB1-F2 protein (34). This protein is encoded by an alternative open reading frame of the PB1 RNA segment, which also directs the synthesis of the PB1 protein, a critical component of the viral polymerase. The PB1-F2 protein localizes to the mitochondria of the infected cells (35) where it interacts with 2 components of the mitochondrial permeability transition pore complex, ANT3 and VDCA1, that are thought to play a major role in apoptosis control (36). As a result, expression of PB1-F2 sensitizes cells to apoptosis. This process might constitute an important immune evasion strategy. Thus, a PB1-F2-knockout influenza A virus induced less cell death than the wild-type virus in infected human monocytes, which suggests that expression of PB1-F2 affects immune cell function during viral infection (34). Although several influenza A virus strains that lack PB1-F2 occur naturally, PB1-F2 likely contributes to viral pathogenicity and might have an important role in determining the severity of pandemic influenza.

NS1 as Target for Antivirals and Vaccines

Our knowledge of NS1 function might be applied in the near future to select for new antiviral compounds against influenza virus. Predictably, small molecules that interfere with the ability of NS1 to bind dsRNA or prevent IFNα/β production will also enhance the host innate immunity against influenza virus, resulting in faster viral clearance. In addition, recombinant influenza viruses with impaired NS1 function might represent efficient live attenuated vaccines against influenza. These viruses can be grown in IFNα/β-deficient substrates to high titers, but they are attenuated in the host (37). Moreover, since the inhibitory effects of NS1 attenuate aspects of both innate and adaptive immunity, NS1 mutant viruses appear to be intrinsically more immunogenic (38). Recombinant influenza viruses with modified NS1 genes have been developed and have proven to be attenuated and immunogenic in different animal models. These modified viruses might be used in the future as the basis of live vaccines against epidemic and pandemic influenza (3740).

Dr García-Sastre is a professor in the department of microbiology at Mount Sinai School of Medicine in New York. His studies provided the first description and molecular analysis of a viral-encoded IFN antagonist among negative-strand RNA viruses.

Top

Acknowledgment

This research was supported by the National Institutes of Health and Department of Defense.

Top

References

  1. Der  SD, Zhou  A, Williams  BR, Silverman  RH. Identification of genes differentially regulated by interferon alpha, beta, or gamma using oligonucleotide arrays. Proc Natl Acad Sci U S A. 1998;95:156238. DOIPubMedGoogle Scholar
  2. Yoneyama  M, Kikuchi  M, Natsukawa  T, Shinobu  N, Imaizumi  T, Miyagishi  M, The RNA helicase RIG-I has an essential function in double-stranded RNA-induced innate antiviral responses. Nat Immunol. 2004;5:7307. DOIPubMedGoogle Scholar
  3. Andrejeva  J, Childs  KS, Young  DF, Carlos  TS, Stock  N, Goodbourn  S, The V proteins of paramyxoviruses bind the IFN-inducible RNA helicase, mda-5, and inhibit its activation of the IFN-beta promoter. Proc Natl Acad Sci U S A. 2004;101:172649. DOIPubMedGoogle Scholar
  4. Foy  E, Li  K, Sumpter  R Jr, Loo  YM, Johnson  CL, Wang  C, Control of antiviral defenses through hepatitis C virus disruption of retinoic acid-inducible gene-I signaling. Proc Natl Acad Sci U S A. 2005;102:298691. DOIPubMedGoogle Scholar
  5. Kawai  T, Takahashi  K, Sato  S, Coban  C, Kumar  H, Kato  H, IPS-1, an adaptor triggering RIG-I- and Mda5-mediated type I interferon induction. Nat Immunol. 2005;6:9818. DOIPubMedGoogle Scholar
  6. Seth  RB, Sun  L, Ea  CK, Chen  ZJ. Identification and characterization of MAVS, a mitochondrial antiviral signaling protein that activates NF-kappaB and IRF3. Cell. 2005;122:66982. DOIPubMedGoogle Scholar
  7. Barchet  W, Cella  M, Colonna  M. Plasmacytoid dendritic cells-virus experts of innate immunity. Semin Immunol. 2005;17:25361. DOIPubMedGoogle Scholar
  8. Kato  H, Sato  S, Yoneyama  M, Yamamoto  M, Uematsu  S, Matsui  K, Cell type-specific involvement of RIG-I in antiviral response. Immunity. 2005;23:1928. DOIPubMedGoogle Scholar
  9. Stark  GR, Kerr  IM, Williams  BR, Silverman  RH, Schreiber  RD. How cells respond to interferons. Annu Rev Biochem. 1998;67:22764. DOIPubMedGoogle Scholar
  10. Le Bon  A, Schiavoni  G, D'Agostino  G, Gresser  I, Belardelli  F, Tough  DF. Type I interferons potently enhance humoral immunity and can promote isotype switching by stimulating dendritic cells in vivo. Immunity. 2001;14:46170. DOIPubMedGoogle Scholar
  11. Biron  CA. Interferons alpha and beta as immune regulators—a new look. Immunity. 2001;14:6614. DOIPubMedGoogle Scholar
  12. Isaacs  A, Lindenmann  J. Virus interference. 1. The interferon. Proc R Soc Lond B Biol Sci. 1957;147:25867. DOIPubMedGoogle Scholar
  13. Isaacs  A, Burke  DC. Mode of action of interferon. Nature. 1958;182:10734. DOIPubMedGoogle Scholar
  14. Weber  F, Kochs  G, Haller  O. Inverse interference: how viruses fight the interferon system. Viral Immunol. 2004;17:498515. DOIPubMedGoogle Scholar
  15. García-Sastre  A, Egorov  A, Matassov  D, Brandt  S, Levy  DE, Durbin  JE, Influenza A virus lacking the NS1 gene replicates in interferon-deficient systems. Virology. 1998;252:32430. DOIPubMedGoogle Scholar
  16. Talon  J, Horvath  CM, Polley  R, Basler  CF, Muster  T, Palese  P, Activation of interferon regulatory factor 3 is inhibited by the influenza A virus NS1 protein. J Virol. 2000;74:798996. DOIPubMedGoogle Scholar
  17. Wang  X, Li  M, Zheng  H, Muster  T, Palese  P, Beg  AA, Influenza A virus NS1 protein prevents the activation of NF-κB and induction of type I IFN. J Virol. 2000;74:1156673. DOIPubMedGoogle Scholar
  18. Donelan  N, Basler  CF, García-Sastre  A. A recombinant influenza A virus expressing an RNA-binding defective NS1 protein induces high levels of beta interferon and is attenuated in mice. J Virol. 2003;77:1325766. DOIPubMedGoogle Scholar
  19. Dauber  B, Heins  G, Wolff  T. The influenza B virus nonstructural NS1 protein is essential for efficient viral growth and antagonizes beta interferon induction. J Virol. 2004;78:186572. DOIPubMedGoogle Scholar
  20. Donelan  NR, Dauber  B, Wang  X, Basler  CF, Wolff  T, García-Sastre  A. The N- and C-terminal domains of the NS1 protein of influenza B virus can independently inhibit IRF-3 and beta interferon promoter activation. J Virol. 2004;78:1157482. DOIPubMedGoogle Scholar
  21. Geiss  GK, Salvatore  M, Tumpey  TM, Carter  VS, Wang  X, Basler  CF, Cellular transcriptional profiling in influenza A virus infected lung epithelial cells: the role of the nonstructural NS1 protein in the evasion of the host innate defense and its potential contribution to pandemic influenza. Proc Natl Acad Sci U S A. 2002;99:1073641. DOIPubMedGoogle Scholar
  22. Fortes  P, Beloso  A, Ortín  J. Influenza virus NS1 protein inhibits pre-mRNA splicing and blocks mRNA nucleocytoplasmic transport. EMBO J. 1994;13:70412.PubMedGoogle Scholar
  23. Lu  Y, Qian  XY, Krug  RM. The influenza virus NS1 protein: a novel inhibitor of pre-mRNA splicing. Genes Dev. 1994;8:181728. DOIPubMedGoogle Scholar
  24. Krug  RM, Yuan  W, Noah  DL, Latham  AG. Intracellular warfare between human influenza viruses and human cells: the roles of the viral NS1 protein. Virology. 2003;309:1819. DOIPubMedGoogle Scholar
  25. Lu  Y, Wambach  M, Katze  MG, Krug  RM. Binding of the influenza virus NS1 protein to double-stranded RNA inhibits the activation of the protein kinase that phosphorylates the elF-2 translation initiation factor. Virology. 1995;214:2228. DOIPubMedGoogle Scholar
  26. Wang  W, Krug  RM. The RNA-binding and effector domains of the viral NS1 protein are conserved to different extents among influenza A and B viruses. Virology. 1996;223:4150. DOIPubMedGoogle Scholar
  27. Yuan  W, Krug  RM. Influenza B virus NS1 protein inhibits conjugation of the interferon (IFN)-induced ubiquitin-like ISG15 protein. EMBO J. 2001;20:36271. DOIPubMedGoogle Scholar
  28. Webster  RG, Hulse  DJ. Microbial adaptation and change: avian influenza. Rev Sci Tech. 2004;23:45365.PubMedGoogle Scholar
  29. Basler  CF, Reid  AH, Dybing  JK, Janczewski  TA, Fanning  TG, Zheng  H, Sequence of the 1918 pandemic influenza virus nonstructural gene (NS) segment and characterization of recombinant viruses bearing the 1918 NS genes. Proc Natl Acad Sci U S A. 2001;98:274651. DOIPubMedGoogle Scholar
  30. López  CB, García-Sastre  A, Williams  BRG, Moran  TM. Type I interferon induction pathway, but not released interferon, participates in the maturation of dendritic cells induced by negative-strand RNA viruses. J Infect Dis. 2003;187:112636. DOIPubMedGoogle Scholar
  31. Cheung  CY, Poon  LL, Lau  AS, Luk  W, Lau  YL, Shortridge  KF, Induction of proinflammatory cytokines in human macrophages by influenza A (H5N1) viruses: a mechanism for the unusual severity of human disease? Lancet. 2002;360:18317. DOIPubMedGoogle Scholar
  32. Seo  SH, Hoffmann  E, Webster  RG. Lethal H5N1 influenza viruses escape host anti-viral cytokine responses. Nat Med. 2002;8:9504. DOIPubMedGoogle Scholar
  33. Marcus  PI, Rojek  JM, Sekellick  MJ. Interferon induction and/or production and its suppression by influenza A viruses. J Virol. 2005;79:288090. DOIPubMedGoogle Scholar
  34. Chen  W, Calvo  PA, Malide  D, Gibbs  J, Schubert  U, Bacik  I, A novel influenza A virus mitochondrial protein that induces cell death. Nat Med. 2001;7:130612. DOIPubMedGoogle Scholar
  35. Gibbs  JS, Malide  D, Hornung  F, Bennink  JR, Yewdell  JW. The influenza A virus PB1-F2 protein targets the inner mitochondrial membrane via a predicted basic amphipathic helix that disrupts mitochondrial function. J Virol. 2003;77:721424. DOIPubMedGoogle Scholar
  36. Zamarin  D, García-Sastre  A, Xiao  X, Wang  R, Palese  P. Influenza virus PB1-F2 protein induces cell death though mitochondrial ANT3 and VDAC1. PLoS Pathog. 2005;1:e4. Epub 2005 Sep 30. DOIPubMedGoogle Scholar
  37. Talon  J, Salvatore  M, O'Neill  RE, Nakaya  Y, Zheng  H, Muster  T, Influenza A and B viruses expressing altered NS1 proteins: a vaccine approach. Proc Natl Acad Sci U S A. 2000;97:430914. DOIPubMedGoogle Scholar
  38. Ferko  B, Stasakova  J, Romanova  J, Kittel  C, Sereinig  S, Katinger  H, Immunogenicity and protection efficacy of replication-deficient influenza A viruses with altered NS1 genes. J Virol. 2004;78:1303745. DOIPubMedGoogle Scholar
  39. Solórzano  A, Webby  RJ, Lager  KM, Janke  BH, García-Sastre  A, Richt  JA. Mutations in the NS1 protein of swine influenza virus impair anti-interferon activity and confer attenuation in pigs. J Virol. 2005;79:753543. DOIPubMedGoogle Scholar
  40. Quinlivan  M, Zamarin  D, García-Sastre  A, Cullinane  A, Chambers  T, Palese  P. Attenuation of equine influenza viruses through truncations of the NS1 protein. J Virol. 2005;79:84319. DOIPubMedGoogle Scholar

Top

Cite This Article

DOI: 10.3201/eid1201.051186

Table of Contents – Volume 12, Number 1—January 2006

EID Search Options
presentation_01 Advanced Article Search – Search articles by author and/or keyword.
presentation_01 Articles by Country Search – Search articles by the topic country.
presentation_01 Article Type Search – Search articles by article type and issue.

Top

Comments

Please use the form below to submit correspondence to the authors or contact them at the following address:

Adolfo García-Sastre, Mount Sinai School of Medicine, Department of Microbiology, Box 1124, 1 Gustave L. Levy Place, New York, NY 10028, USA; fax: 212-534-1684

Send To

10000 character(s) remaining.

Top

Page created: February 16, 2012
Page updated: February 16, 2012
Page reviewed: February 16, 2012
The conclusions, findings, and opinions expressed by authors contributing to this journal do not necessarily reflect the official position of the U.S. Department of Health and Human Services, the Public Health Service, the Centers for Disease Control and Prevention, or the authors' affiliated institutions. Use of trade names is for identification only and does not imply endorsement by any of the groups named above.
file_external