Elsevier

Virus Research

Volume 160, Issues 1–2, September 2011, Pages 367-373
Virus Research

Comparison of innate immune responses to pathogenic and putative non-pathogenic hantaviruses in vitro

https://doi.org/10.1016/j.virusres.2011.07.013Get rights and content

Abstract

Hantaviruses are human pathogens that cause hemorrhagic fever with renal syndrome or hantavirus cardiopulmonary syndrome. The mechanisms accounting for the differences in virulence between pathogenic and non-pathogenic hantaviruses are not well known. We have examined the pathogenesis of different hantavirus groups by comparing the innate immune responses induced in the host cell following infection by pathogenic (Sin Nombre, Hantaan, and Seoul virus) and putative non-pathogenic (Prospect Hill, Tula, and Thottapalayam virus) hantaviruses. Pathogenic hantaviruses were found to replicate more efficiently in interferon-competent A549 cells than putative non-pathogenic hantaviruses. The former also suppressed the expression of the interferon-β and myxovirus resistance protein genes, while the transcription level of both genes increased rapidly within 24 h post-infection in the latter. In addition, the induction level of interferon correlated with the activation level of interferon regulatory factor-3. Taken together, these results suggest that the observed differences are correlated with viral pathogenesis and further indicate that pathogenic and putative non-pathogenic hantaviruses differ in terms of early interferon induction via activation of the interferon regulatory factor-3 in infected host cells.

Highlights

► Pathogenic hantaviruses cause hemorrhagic fever with renal syndrome (HFRS) in Eurasia and hantavirus cardiopulmonary syndrome (HCPS) in the Americas. However, not all hantaviruses are human pathogens. ► We have examined the pathogenesis of different hantavirus groups by comparing the innate immune responses induced in the host cell following infection by pathogenic (Sin Nombre, Hantaan, and Seoul virus) and putative non-pathogenic (Prospect Hill, Tula, and Thottapalayam virus) hantaviruses. ► The degree of innate immune inhibition may play an important role to distinguish pathogenic and non-pathogenic hantaviruses.

Introduction

Hantavirus is a genus of rodent-borne, lipid-enveloped, negative-sense, single-stranded RNA viruses belonging to the family Bunyaviridae. Viruses of this family are characterized by a trisegmented genome, designated according to size as the large (L), medium (M), and small (S) segments, respectively, which encode four proteins: the RNA-dependent RNA polymerase, two envelope glycoproteins (Gn and Gc), and the nucleocapsid protein (N) (Elliott et al., 1991). Hantaviruses are broadly classified into Old World and New World categories based on geographic distribution. The transmission of hantaviruses to humans occurs via inhalation of contaminated aerosolized excreta from rodents (Nuzum et al., 1988). Pathogenic hantaviruses cause hemorrhagic fever with renal syndrome (HFRS) in Eurasia (Old World hantavirus) and hantavirus cardiopulmonary syndrome (HCPS) in the Americas (New World hantavirus). However, not all hantaviruses are human pathogens; for example, there is no known association between the Prospect Hill, Tula, and Thottapalayam virus, and human disease (Carey et al., 1971, Lee et al., 1982b, Lee et al., 1985, Plyusnin et al., 1994, Song et al., 2007, Vapalahti et al., 1996).

Virus infection elicits a cascade of rapid innate immune responses in the host that are aimed at blocking viral replication through the activation of an early defense mechanism. Activation of the innate immune responses generally leads to the induction of type-I Interferon (IFN-α/β) as one of the first lines of defense against viral infection (Bonjardim, 2005, Conzelmann, 2005). However, IFN-α/β is not only synthesized in response to viral infection, but also induced at low levels in the absence of viral infection (Taniguchi and Takaoka, 2001). The regulation of IFN responses may differ among virus species, cell lines, signaling pathways, and integrin receptors.

The expression of IFN-β requires the activation of transcription factors, such as IFN regulatory factor (IRF)-3 and nuclear factor kappa B (NF-κB), both of which play a major role in the induction of IFN by viruses in the cytoplasm (Bonjardim, 2005, Haller et al., 2006, Hiscott, 2007). Viral infection triggers the phosphorylation and dimerization of IRF-3 and its subsequent translocation to the nucleus where it initiates IFN-β gene expression. Secreted IFN-β binds to the type-I IFN receptors (IFNARs) and initiates activation of the IFN signaling pathway. The expression of IFN-stimulated genes (ISGs), including 2′,5′ oligoadenylate synthetases (2′,5′-OAS), protein kinase R (PKR), and myxovirus resistance protein (Mx), is activated via Janus kinase/signal transducers and activators of the transcription (JAK-STAT) pathway (Borden et al., 2007, Randall and Goodbourn, 2008).

In the study reported here, we have compared innate immune responses to the pathogenic hantaviruses (PaV), including Sin Nombre virus (SNV), Hantaan virus (HTNV), and Seoul virus (SEOV), and the putative non-pathogenic hantaviruses (NPaV), including Prospect Hill virus (PHV), Tula virus (TULV), and Thottapalayam virus (TPMV) (Table 1). Our results provide interesting insights into the differences in human virulence among hantavirus strains and demonstrate that infection with NPaV may induce a strong innate immune response through the activation of such effectors as IFN-β and the MxA gene during the early post-infection period.

Section snippets

Cells and plasmid

Vero E6 (African green monkey epithelial kidney cell, ATCC C1008 CRL-1586) and A549 (Human epithelial lung cell, ATCC CCL-185) cells were maintained in Dulbecco's Modified Eagle's Medium (DMEM; Lonza) and RPMI-1640 (Lonza, Basel, Switzerland) supplemented with 5% fetal bovine serum (FBS), 10 mM HEPES buffer, 2 mM l-glutamine, and 50 ng/ml gentamycin. The pEGFP-c1-hIRF3 plasmid was kindly provided by Dr. Man-Seong Park (College of Medicine, Hallym University, Korea).

Viruses

The SNV (strain Convict Creek

Differential efficiencies of hantavirus replication in Vero E6 and A549 cells

The replication of the PaV and NPaV hantaviruses in Vero E6 and A549 cells was determined on each post-infection day up to post-infection day 7 (Fig. 1). Vero E6 cells, which are IFN-deficient cells, are unable to produce type-I IFN because of a chromosomal defect (Diaz et al., 1988, Wathelet et al., 1992), whereas A549 cells are IFN-competent cells. In Vero E6 cells, both PaV and NPaV grew with similar kinetics and replication efficiency, yielding titers of 1 × 105 to 1 × 106 PFU/ml within 7 days

Discussion

Hantaviruses are distributed worldwide and occur in both pathogenic (etiologic agents of HFRS and HCPS) and non-pathogenic hantaviruses. The differences between pathogenic and non-pathogenic hantaviruses are currently unclear. Interestingly, infection with pathogenic and non-pathogenic hantaviruses differentially activate the innate immunity in vitro (Alff et al., 2006, Geimonen et al., 2002, Handke et al., 2009, Kraus et al., 2004, Spiropoulou et al., 2007), and this difference in the innate

Acknowledgment

This work was supported in part by the Brain Korea 21 Project of the Ministry of Education and Human Resources Development, Republic of Korea.

References (51)

  • P.J. Alff et al.

    The NY-1 hantavirus Gn cytoplasmic tail coprecipitates TRAF3 and inhibits cellular interferon responses by disrupting TBK1–TRAF3 complex formation

    Journal of Virology

    (2008)
  • E.C. Borden et al.

    Interferons at age 50: past, current and future impact on biomedicine

    Nature Reviews Drug Discovery

    (2007)
  • D.E. Carey et al.

    Thottapalayam virus: a presumptive arbovirus isolated from a shrew in India

    Indian Journal of Medical Research

    (1971)
  • K.K. Conzelmann

    Transcriptional activation of alpha/beta interferon genes: interference by nonsegmented negative-strand RNA viruses

    Journal of Virology

    (2005)
  • M.O. Diaz et al.

    Homozygous deletion of the alpha- and beta 1-interferon genes in human leukemia and derived cell lines

    Proceedings of the National Academy of Sciences of the United States of America

    (1988)
  • R.M. Elliott et al.

    Bunyaviridae genome structure and gene expression

    Current Topics in Microbiology and Immunology

    (1991)
  • M. Frese et al.

    Inhibition of bunyaviruses, phleboviruses, and hantaviruses by human MxA protein

    Journal of Virology

    (1996)
  • I.N. Gavrilovskaya et al.

    Cellular entry of hantaviruses which cause hemorrhagic fever with renal syndrome is mediated by beta3 integrins

    Journal of Virology

    (1999)
  • E. Geimonen et al.

    Pathogenic and nonpathogenic hantaviruses differentially regulate endothelial cell responses

    Proceedings of the National Academy of Sciences of the United States of America

    (2002)
  • N. Grandvaux et al.

    Transcriptional profiling of interferon regulatory factor 3 target genes: direct involvement in the regulation of interferon-stimulated genes

    Journal of Virology

    (2002)
  • O. Haller et al.

    Mx proteins: mediators of innate resistance to RNA viruses

    Revue Scientifique et Technique

    (1998)
  • W. Handke et al.

    Hantaan virus triggers TLR3-dependent innate immune responses

    Journal of Immunology

    (2009)
  • D. Holzinger et al.

    Induction of MxA gene expression by influenza A virus requires type I or type III interferon signaling

    Journal of Virology

    (2007)
  • J. Hooper

    Virus detection and identification with serological tests

  • K.M. Jaaskelainen et al.

    Tula and Puumala hantavirus NSs ORFs are functional and the products inhibit activation of the interferon-beta promoter

    Journal of Medical Virology

    (2007)
  • Cited by (20)

    • Hantavirus entry: Perspectives and recent advances

      2019, Advances in Virus Research
      Citation Excerpt :

      The Gn cytoplasmic tail is 110 amino acids long and carries a single structured domain, a zinc finger, which likely plays a role in interactions with the N protein during virus assembly (Estrada et al., 2009, 2011; Wang et al., 2010), possibly in a manner functionally analogous to the matrix proteins of other viruses (Hepojoki et al., 2010a,b). The Gn cytoplasmic tails of virulent hantaviruses can also block the type I interferon response to limit the host immune response to infection (Alff et al., 2008; Shim et al., 2011). Crystal structures of the pre-fusion ectodomain of Gc proteins from multiple hantaviruses reveal that Gc is a class II fusion protein similar in structure to the glycoproteins of flaviviruses, alphaviruses, Rubella virus, and phleboviruses (Guardado-Calvo and Rey, 2017; Guardado-Calvo et al., 2016; Willensky et al., 2016).

    • Infection of human airway epithelial cells by different subtypes of Dobrava-Belgrade virus reveals gene expression patterns corresponding to their virulence potential

      2016, Virology
      Citation Excerpt :

      Lung tissue cells, as a first line of defense after inhalation of hantavirus particles, decide about the further progression of disease. Moreover, A549 cells have been widely used as an in vitro model system for hantavirus infections including gene expression profiling (Lee et al., 2011; Oelschlegel et al., 2007; Popugaeva et al., 2012; Shim et al., 2011; Stoltz and Klingström, 2010). Our study was conducted by an analysis of the cellular gene expression profiles via whole-genome gene expression microarray and quantitative real-time PCR (qPCR) techniques.

    • Shikonin induces cell cycle arrest in human gastric cancer (AGS) by early growth response 1 (Egr1)-mediated p21 gene expression

      2014, Journal of Ethnopharmacology
      Citation Excerpt :

      Blots were visualized using an enhanced chemiluminescence western blotting detection kit (SantaCruz Biotechnology, CA, USA). Immunostaining for the indicated proteins was performed as described (Shim et al., 2011). AGS cells were fixed with 4% paraformaldehyde for 30 min.

    View all citing articles on Scopus

    The findings and conclusions in this report are those of the authors and do not necessarily represent the views of CDC.

    View full text