Elsevier

Cytokine

Volume 62, Issue 1, April 2013, Pages 151-159
Cytokine

Seasonal and pandemic influenza H1N1 viruses induce differential expression of SOCS-1 and RIG-I genes and cytokine/chemokine production in macrophages

https://doi.org/10.1016/j.cyto.2013.01.018Get rights and content

Abstract

Background

Infection with pandemic (pdm) A/H1N1 virus induces high levels of pro-inflammatory mediators in blood and lungs of experimental animals and humans.

Methods

To compare the involvement of seasonal A/PR/8/34 and pdm A/H1N1 virus strains in the regulation of inflammatory responses, we analyzed the changes in the whole-genome expression induced by these strains in macrophages and A549 epithelial cells. We also focused on the functional implications (cytokine production) of the differential induction of suppressors of cytokine signaling (SOCS)-1, SOCS-3, retinoid-inducible gene (RIG)-I and interferon receptor 1 (IFNAR1) genes by these viral strains in early stages of the infection.

Results

We identified 130 genes differentially expressed by pdm A/H1N1 and A/PR/8/34 infections in macrophages. mRNA levels of SOCS-1 and RIG-I were up-regulated in macrophages infected with the A/PR/8/34 but not with pdm A/H1N1 virus. mRNA levels of SOCS-3 and IFNAR1 induced by A/PR/8/34 and pdm A/H1N1 strains in macrophages, as well as in A549 cells were similar. We found higher levels of IL-6, TNF-α, IL-10, CCL3, CCL5, CCL4 and CXCL8 (p < 0.05) in supernatants from cultures of macrophages infected with the pdm A/H1N1 virus compared to those infected with the A/PR/8/34 strain, coincident with the lack of SOCS-1 and RIG-I expression. In contrast, levels of INF-α were higher in cultures of macrophages 48 h after infection with the A/PR/8/34 strain than with the pdm A/H1N1 virus.

Conclusions

These findings suggest that factors inherent to the pdm A/H1N1 viral strain may increase the production of inflammatory mediators by inhibiting SOCS-1 and modifying the expression of antiviral immunity-related genes, including RIG-I, in human macrophages.

Highlights

Hypercytokinemia contributes to the severity of influenza A virus infection. ► SOCS-1 and RIG-I are differentially induced by the A/H1N1 and seasonal strains. ► The pandemic A/H1N1 strain induces an overproduction of inflammatory mediators. ► The inflammatory-antiviral responses are regulated by viral-host related factors.

Introduction

The 2009 outbreak of swine-origin influenza A/H1N1 [1], [2] continues to affect many countries, having caused over 18,000 deaths worldwide [3]. A growing body of evidence supports the hypothesis that the development of severe pneumonia in patients with pandemic (pdm) A/H1N1 infection is associated with increased immune activation and immune complex deposition [4], [5]. Therefore, it is of great importance to understand the factors that determine the development of severe disease. In this regard, high levels of pro-inflammatory cytokines and chemokines have been detected in peripheral blood and lung tissue from patients with severe pneumonia associated to the pdm A/H1N1 infection [4], [6]. Even though cytokines, chemokines, and growth factors are required to control influenza virus infection, their overproduction in an uncontrolled inflammatory response can lead to lung tissue damage [4], [6], [7]. The suppressors of cytokine signaling (SOCS) are a family of proteins that down-regulate cytokine signaling [8], [9], [10], [11] by negatively regulating JAK/STAT-mediated signal transduction [9], [10], [11]. Experimental infection of A549 epithelial cells with seasonal influenza A virus up-regulates the expression of SOCS-1 and SOCS-3. These proteins regulate the immune response against influenza A viruses through a retinoid-inducible gene (RIG)-I/mitochondrial antiviral signaling protein (MAVS)/interferon (alpha and beta) receptor 1 (IFNAR1)-dependent pathway [12].

On the other hand, the combination of gene segments from North American and Eurasian swine lineages of the pdm A/H1N1 virus [13] create unique molecular structures [14], [15] that could regulate host immune responses differently than seasonal influenza strains, and contribute to the particular clinical presentation of pandemic influenza infections.

We therefore hypothesized that immune feedback or regulatory mechanisms that normally control host inflammatory responses to pathogens may be absent or impaired in severe pdm A/H1N1 infection, due to the virus itself.

To establish if particular patterns of gene expression and alterations of immune regulation are attributable to specific viral factors, we analyzed the whole genome expression patterns induced by the pdm A/H1N1 and A/PR/8/34 strains through microarray analysis of infected human macrophages and A549 cells. In addition, we performed in vitro assays of macrophages and A549 cells in order to evaluate the differences between the pdm A/H1N1 and A/PR/8/34 in their capacity to induce SOCS-1, SOCS-3, and the antiviral response molecule RIG-I, as well as the production of pro-inflammatory cytokines, chemokines and growth factors.

Section snippets

Ethics statement

The Institutional Review Board of the National Institute of Respiratory Diseases (INER) reviewed and approved this protocol (protocol number B27-10), under which all subjects were recruited. All subjects provided written informed consent, and authorized the storage of their samples at INER repositories for this and future studies.

Seasonal and pandemic A/H1N1 influenza virus isolation, identification, and propagation

Influenza pdm A/H1N1 virus isolates were obtained from patients with severe pneumonia, who signed an informed consent letter, during the 2009 outbreak in Mexico City,

Differential expression of antiviral immune response-related genes is induced by pandemic A/H1N1 and A/PR/8/34 strains

In order to compare the transcriptional profiles induced by different influenza A virus strains, we compared genome-wide expression induced by infection (10 h) with the pdm A/H1N1 and A/PR/8/34 viruses in macrophages and A549 cells using the exon array 1.0 ST. We also analyzed the gene expression profile of macrophages without virus infection (Mock at 10 h of infection) and the results were used as basal expression to determine the differences induced in the gene expression provoked by the

Discussion

In previous studies, elevated levels of pro-inflammatory cytokines and chemokines in blood and lung from patients with severe pneumonia associated with pdm A/H1N1 virus have been identified, suggesting that hypercytokinemia may contribute to the severity of the disease [4], [6].

Here, we analyzed the ability of pdm A/H1N1 and A/PR8/34 viruses to induce specific gene expression patterns in infected macrophages and A549 epithelial cells. These analyses allowed us to identify 130 genes

Acknowledgement

This work was supported by The Mexican National Council of Science and Technology (CONACYT) Grants [127002, 142364 CB-2010-155382 and E-1105].

References (37)

  • R. Perez-Padilla et al.

    Pneumonia and respiratory failure from swine-origin influenza A (H1N1) in Mexico

    N Eng J Med

    (2009)
  • Writing Committee of the WHO Consultation on Clinical Aspects of Pandemic (H1N1) 2009 Influenza, E. Bautista, T....
  • J. Zúñiga et al.

    Inflammatory profiles in severe pneumonia associated to the pandemic influenza A/H1N1 virus isolated in Mexico City

    Autoimmunity

    (2011)
  • A.C. Monsalvo et al.

    Severe pandemic 2009 H1N1 influenza disease due to pathogenic immune complexes

    Nat Med

    (2011)
  • J.F. Bermejo-Martin et al.

    Host adaptive immunity deficiency in severe pandemic influenza

    Crit Care

    (2010)
  • D. Kobasa et al.

    Aberrant innate immune response in lethal infection of macaques with the 1918 influenza virus

    Nature

    (2007)
  • T.A. Endo et al.

    A new protein containing an SH2 domain that inhibits JAK kinases

    Nature

    (2005)
  • A. Yoshimura et al.

    SOCS proteins, cytokine signalling and immune regulation

    Nat Rev Immunol

    (2007)
  • Cited by (30)

    • Influence of adjuvants on the amount, specificity and functional activity of antibody response to human influenza vaccine in mice

      2021, Molecular Immunology
      Citation Excerpt :

      The percentage of IgGs that recognized the globular head relative to total IIV was estimated dividing the optical density obtained from an ELISA assay using the globular head as the source of antigen vs. the optical density from a parallel ELISA assay using the complete vaccine as the antigen multiplied by 100. Human Influenza A H1N1pdm09 virus isolated at Instituto Nacional de Enfermedades Respiratorias (INER) during the 2009 outbreak in México (Ramirez-Martinez et al., 2013; Zuniga et al., 2011) was propagated in MDCK cells and the whole supernatant batch was aliquoted and frozen in liquid nitrogen until use. The virus was titrated to obtain the batch’s TCID50 (Kawaoka and Neumann, 2012; Spackman, 2012).

    • Galectins regulate the inflammatory response in airway epithelial cells exposed to microbial neuraminidase by modulating the expression of SOCS1 and RIG1

      2015, Molecular Immunology
      Citation Excerpt :

      SOCS1, a JAK binding protein, belongs to the SOCS family that is responsible for key negative feedback mechanisms to prevent uncontrolled inflammation (Croker et al., 2008). Upon IAV challenge, SOCS1 expression is upregulated through a RIG1/MAVS/IFNAR1-dependent pathway (Pothlichet et al., 2008), which suppresses expression of inflammatory cytokines, e.g., IL-6, TNF-α, IL-10, CCL3, CCL5, CCL4 and CXCL8, to prevent the cytokine storm (Ramirez-Martinez et al., 2013). Thus, upregulation of SOCS1 provides a negative feedback mechanism to set inflammation under control after pathogen clearance.

    • Circulating levels of miR-150 are associated with poorer outcomes of A/H1N1 infection

      2015, Experimental and Molecular Pathology
      Citation Excerpt :

      miRNAs have an important role in the regulation of many processes including inflammation in different clinical conditions (Perera and Ray, 2007). It is well known that uncontrolled immune inflammatory responses may contribute to the pathogenesis of severe lung damage in mice and humans with pandemic A/H1N1 virus infection (Zuniga et al., 2011; Bautista et al., 2013; Ubags et al., 2014; Ramirez-Martinez et al., 2013). In the current study we analyzed the circulating miRNA profiles and inflammatory mediators circulating levels in patients with different clinical presentations of the A/H1N1 infection and the most striking findings were: 1) high circulating levels of miR-150 are associated with severe A/H1N1 illness; 2) in experimental infection assays pandemic A/H1N1 induces higher levels of miR-150 than seasonal H3N2 strain; 3) miR-29c, miR-145 and miR-22 are differentially expressed in patients with severe A/H1N1 disease whereas miR-210, miR-126 and miRT-222 are downregulated in individuals exposed to the A/H1N1 virus; 4) there is a correlation between circulating levels of miR-150 with different pro-inflammatory mediators and growth factors including: IL-1b, IFN-γ, CXCL8 and G-CSF, particularly in critically ill A/H1N1 patients and 5) PI3K–Akt–mTOR, apoptosis, cancer and antiviral pathways are the most associated with the differentially expressed miRNAs in A/H1N1 infection.

    View all citing articles on Scopus
    1

    These authors contributed equally to this work.

    View full text