1,147
Views
2
CrossRef citations to date
0
Altmetric
Research Paper

Constitutive IFNα/β signaling maintains expression of signaling intermediaries for efficient cytokine responses

, &
Article: e1173804 | Received 09 Feb 2016, Accepted 29 Mar 2016, Published online: 18 May 2016

ABSTRACT

Interferons (IFNs) are a family of immunoregulatory cytokines with important roles in anti-viral and anti-tumor responses. Type I and II IFNs bind distinct receptors and are associated with different stages of the immune response. There is however, considerable crosstalk between these two cytokines with enhancement of IFNγ responses following IFNα/β priming and loss of IFNα/β receptor (IFNAR) resulting in diminished IFNγ responses. In this study, we sought to define the mechanism of crosstalk between the type I and II IFNs. Our previous reports demonstrated reduced expression of the canonically activated transcription factor signal transducer and activator of transcription (STAT)1, in cells lacking the IFNAR α chain (IFNAR1). Therefore, we used microarray analysis to determine whether reconstitution of STAT1 in IFNAR1-deficient cells was sufficient to restore IFNγ responses. We identified several biological pathways, including the MHC class I antigen presentation pathway, in which STAT1 reconstitution was able to significantly rescue IFNγ-mediated gene regulation in Ifnar1−/− cells. Notably, we also found that in addition to low basal expression of STAT1, cells lacking the IFNAR1 also had aberrant expression of multiple other transcription factors and signaling intermediaries. The studies described herein demonstrate that basal and regulated expression of signaling intermediaries is a mechanism for crosstalk between cytokines including type I and II IFNs.

INTRODUCTION

The exposure of cells to cytokines, chemokines and other extracellular signals during pathogenic infection, injury and neoplasia allows for potent, and often synergistic, host immune responses.Citation1,2 IFNs, a family of immunoregulatory cytokines with important roles in anti-pathogen and anti-tumor responses, can synergise with other stimuli, such as tumor necrosis factor (TNF) α and interleukin (IL) 1α for more potent immune responses.Citation3-10 Upon IFN binding, receptor-associated janus activated kinases (JAKs) are phosphorylated leading to activation of latent signal transducer and activator of transcription (STAT) molecules. Pivotal to IFN signaling is the transcription factor STAT1, the loss of which severely impairs IFN-dependent cellular responses.Citation11,12 The canonical, JAK/STAT1-dependent IFNγ signaling pathway has been extensively studied, however more recently alternate signaling pathways triggered by IFNγ such as mitogen activated protein kinase (MAPK) and nuclear factor κB (NFκB) pathways have been identified.Citation13-16

Although specific IFN family members bind to selective receptors and activate distinct signaling pathways, significant crosstalk between these pathways is evident.Citation1-3,6,17,18 Pre-treatment of cells with low levels of type I IFNs (IFNα/β) leads to more potent response to type II IFN (IFNγ) and interestingly loss of the type I IFN receptor (IFNAR) results in diminished type II IFN responses.Citation19,20 Although type I IFNs are secreted in abundance in response to viral infection, uninfected cells also constitutively secrete low concentrations of IFNα/β.Citation21-24 Studies by our laboratory and others, show that cells unable to produce or respond to constitutive IFNα/β have attenuated STAT1 expression. This led to a model of cytokine crosstalk which postulates that priming levels of IFNβ regulate STAT1 expression, which is important for mediating IFNγ responses.Citation23,25,26 Moreover, we have demonstrated that reconstitution of STAT1 into Ifnar1−/− cells could restore IFNγ-induced STAT1 DNA binding and partially rescued IFNγ-dependent anti-viral responses.Citation23 In this study we used global gene expression analysis to examine the contribution of STAT1 to the crosstalk between tonic IFNα/β and IFNγ. Our data revealed that STAT1 alone regulates only a small proportion of IFN crosstalk. Moreover, loss of tonic IFNα/β signaling results in aberrant expression of numerous transcription factors and intracellular signaling molecules, thus tonic IFNα/β signaling regulates the expression of multiple signaling intermediaries involved in cytokine signaling.

RESULTS

STAT1 rescue of IFNγ responsiveness in Ifnar1−/− cells is enhanced at earlier time points

STAT1 is the canonical transcription factor activated in response to IFNγ and we have previously found that its expression is attenuated in Ifnar1−/− cells.Citation23 To determine the extent to which STAT1 reconstitution alone could rescue molecular responses to IFNγ in Ifnar1−/− cells, microarray-based gene expression analysis was performed over a 6 hr time course. Prior to microarray analysis STAT1 expression in wild type, Ifnar1−/−, empty vector-transduced (Ifnar1−/−MSCV) and HA-tagged STAT1α-transduced (Ifnar1−/−HA-STAT1) Ifnar1−/− mouse embryonic fibroblasts (MEFs) was confirmed by western blotting (). Probes in which IFNγ induced a significant (p < 0.05) and greater than 1.2-fold increase or decrease in expression were considered to be regulated by IFNγ. Of the 362 probes which had induced expression in wild type cells following 1 hr of IFNγ treatment, 109 probes (30.1%) were also induced in Ifnar1−/− and/or Ifnar1−/−MSCV cells and Ifnar1−/−HA-STAT1 cells (). In contrast to induced gene expression, only 5 of the 440 probes which showed significant repression in response 1 hr of IFNγ treatment in wild type cells, were also repressed in wild type, Ifnar1−/− and/or Ifnar1−/−MSCV cells and Ifnar1−/−HA-STAT1 cells (). Similar patterns of gene expression were seen at the later time points with 18.2% and 40.3% of probes induced and 3.2% and 2.5% of probes repressed in wild type, Ifnar1−/− and/or Ifnar1−/−MSCV cells and Ifnar1−/−HA-STAT1 cells in response to 3 and 6 hrs of IFNγ treatment respectively ().

Figure 1 Reconstitution of STAT1 in Ifnar1−/− MEFs partially rescues IFNγ responses. (A) Protein lysates from wild type, Ifnar1−/−, Ifnar1−/−MSCV and Ifnar1−/−HA-STAT1 129/C3H MEFs were separated by SDS-PAGE and probed with antibodies specific for STAT1 and actin as a loading control. (B-G) wild type, Ifnar1−/−, Ifnar1−/−MSCV and Ifnar1−/−HA-STAT1 129/C3H MEFs were treated with 100 U/ml IFNγ for 1, 3 and 6 hrs and mRNA expression was assessed by Affymetrix microarray (n = 3). (B-D) Venn diagram of the number of genes that are up-regulated (↑ green) or down-regulated (↓ red) relative to untreated (p < 0.05, >1.2-fold) following (B) 1 hr, (C) 3 hrs or (D) 6 hrs of IFNγ treatment and whether they are specific or common to each cell type. (E-G) Left panel. Heat maps of probes with attenuated IFNγ-dependent gene induction in Ifnar1−/− MEFs relative to wild type MEFs (p > 0.05, <1.2-fold change relative to untreated or >1.2-fold attenuation in change compared to wild type). Right panel. Heat maps of probes with attenuated (as defined above) IFNγ-dependent gene induction in Ifnar1−/− MEFs and Ifnar1−/−MSCV MEFs relative to wild type MEFs and rescued (p < 0.05 and >1.2-fold change relative to untreated and <1.2-fold attenuation in change compared to wild type) in Ifnar1−/−HA-STAT1 MEFs. The 10 attenuated (left panel) and rescued (right panel) probes with the greatest fold change in wild type MEFs following (E) 1 hr, (F) 3 hrs and (G) 6 hrs of IFNγ treatment are displayed. Heatmaps were generated using Java Treeview. Total number of probes with induced (↑) and repressed (↓) expression in wild type MEFs that had attenuated (left panel) or rescued (right panel) expression are listed below heatmaps.

Figure 1 Reconstitution of STAT1 in Ifnar1−/− MEFs partially rescues IFNγ responses. (A) Protein lysates from wild type, Ifnar1−/−, Ifnar1−/−MSCV and Ifnar1−/−HA-STAT1 129/C3H MEFs were separated by SDS-PAGE and probed with antibodies specific for STAT1 and actin as a loading control. (B-G) wild type, Ifnar1−/−, Ifnar1−/−MSCV and Ifnar1−/−HA-STAT1 129/C3H MEFs were treated with 100 U/ml IFNγ for 1, 3 and 6 hrs and mRNA expression was assessed by Affymetrix microarray (n = 3). (B-D) Venn diagram of the number of genes that are up-regulated (↑ green) or down-regulated (↓ red) relative to untreated (p < 0.05, >1.2-fold) following (B) 1 hr, (C) 3 hrs or (D) 6 hrs of IFNγ treatment and whether they are specific or common to each cell type. (E-G) Left panel. Heat maps of probes with attenuated IFNγ-dependent gene induction in Ifnar1−/− MEFs relative to wild type MEFs (p > 0.05, <1.2-fold change relative to untreated or >1.2-fold attenuation in change compared to wild type). Right panel. Heat maps of probes with attenuated (as defined above) IFNγ-dependent gene induction in Ifnar1−/− MEFs and Ifnar1−/−MSCV MEFs relative to wild type MEFs and rescued (p < 0.05 and >1.2-fold change relative to untreated and <1.2-fold attenuation in change compared to wild type) in Ifnar1−/−HA-STAT1 MEFs. The 10 attenuated (left panel) and rescued (right panel) probes with the greatest fold change in wild type MEFs following (E) 1 hr, (F) 3 hrs and (G) 6 hrs of IFNγ treatment are displayed. Heatmaps were generated using Java Treeview. Total number of probes with induced (↑) and repressed (↓) expression in wild type MEFs that had attenuated (left panel) or rescued (right panel) expression are listed below heatmaps.

To determine the genes for which loss of IFNAR1 expression attenuated IFNγ responsiveness, we compared the wild type and Ifnar1−/− microarray data sets and considered probes which lacked a significant, >1.2-fold change in expression or had >1.2-fold attenuation of gene regulation compared to wild type cells as having lost IFNγ responsiveness. In response to 1, 3 and 6 hrs of IFNγ treatment, 85.6%, 93.6% and 75.3% of probes had attenuated gene induction in Ifnar1−/− cells compared to wild type cells respectively (). For IFNγ-dependent gene repression 99.3%, 99.8% and 98.1% of probes had attenuated responses following 1, 3 and 6 hrs of IFNγ treatment (). These data indicate that the vast majority of IFNγ regulated genes are affected by the absence of IFNAR1.

STAT1α reconstitution of Ifnar1−/− cells was considered to have rescued IFNγ responses if the following criteria were met: (i) The gene had lost IFNγ responsiveness in Ifnar1−/− and Ifnar1−/−MSCV cells compared to wild type cells (ii) The gene had wild type-equivalent responsiveness to IFNγ in Ifnar1−/−HA-STAT1 cells (i.e. a significant and >1.2-fold change in expression relative to untreated and <1.2-fold attenuation of gene regulation in Ifnar1−/−HA-STAT1 cells compared to wild type cells). Of the 310 probes in which IFNγ-induced gene expression was attenuated in Ifnar1−/− cells following 1 hr of IFNγ treatment, 43 (13.8%) probes had rescued IFNγ responsiveness in Ifnar1−/−HA-STAT1 cells (). In response to 3 hrs and 6 hrs of IFNγ treatment, induction of gene expression of 62 (8.4%) and 38 (7.4%) probes with attenuated responses in Ifnar1−/− cells was rescued in Ifnar1−/−HA-STAT1 cells respectively (). In contrast to the effects on IFNγ-dependent gene induction, Stat1α reconstitution of Ifnar1−/− cells only rescued gene repression of 0.7%, 1.0% and 0.4% of probes following 1, 3 and 6 hrs of IFNγ treatment respectively. These data indicated that the effect of STAT1 reconstitution on IFNγ-dependent gene regulation was greatest during early phase gene induction.

STAT1 rescues IFNγ-mediated responses to a select range of biological pathways in IFNAR1 cells

Ontological analysis of the microarray data was performed to identify the biological pathways rescued following IFNγ treatment of Ifnar1−/−HA-STAT1 cells. The 2106 genes with attenuated regulation by IFNγ in Ifnar1−/− MEFs over the 6 hr time course of IFNγ treatment were analyzed using MetaCorZe's Genego. Genes with aberrant expression following IFNγ treatment in Ifnar1−/− MEFs were involved in a diverse range of biological pathways, with the most significantly affected including developmental, immunological response, reproduction and apoptosis pathways (). Surprisingly, only one pathway directly associated with STAT signaling was found among the 10 most significantly affected biological pathways. Subsequently, genes with attenuated regulation by IFNγ in Ifnar1−/− MEFs were compared to the genes in which IFNγ responses were significantly rescued by reconstitution of STAT1 using MetaCore's Genego comparative analysis for pathway maps. This analysis revealed, unsurprisingly, that among the 10 pathways most significantly affected by both the loss of IFNAR1 and reconstitution of STAT1 cells were the IFNα/β signaling pathway and multiple JAK/STAT signaling pathways (). Of the 10 most significantly affected pathways, 7 were associated with immune responses with the most significantly affected pathway being the MHC class I antigen presentation pathway (). These data suggested that despite genes involved in a diverse array of biological responses pathways being aberrantly regulated by IFNγ in the absence of IFNAR1, the pathways in which STAT1 reconstitution of IFNAR1 deficient cells rescued IFNγ-mediated gene regulation were enriched for immune response pathways.

Figure 2 Reconstitution of STAT1α in Ifnar1−/− MEFs preferentially rescues immune and inflammatory IFNγ responses. (A-B) Probes from listed as attenuated (A-B) or rescued (B) were analyzed using MetaCore's Genego. The 10 most significantly affected pathways are displayed. (A) Pathway enrichment analysis of attenuated genes. EMT = epithelial-to-mesenchymal transition. (B) Comparative pathway enrichment analysis of attenuated and rescued genes. (C) Heatmaps displaying fold change in expression following 1, 3 or 6 hrs IFNγ treatment of genes involved in antigen presentation via MHC class I for wild type, Ifnar1−/−, Ifnar1−/−MSCV and Ifnar1−/−HA-STAT1 MEFs. Heatmaps were generated using Java Treeview.

Figure 2 Reconstitution of STAT1α in Ifnar1−/− MEFs preferentially rescues immune and inflammatory IFNγ responses. (A-B) Probes from figure 1.E-G listed as attenuated (A-B) or rescued (B) were analyzed using MetaCore's Genego. The 10 most significantly affected pathways are displayed. (A) Pathway enrichment analysis of attenuated genes. EMT = epithelial-to-mesenchymal transition. (B) Comparative pathway enrichment analysis of attenuated and rescued genes. (C) Heatmaps displaying fold change in expression following 1, 3 or 6 hrs IFNγ treatment of genes involved in antigen presentation via MHC class I for wild type, Ifnar1−/−, Ifnar1−/−MSCV and Ifnar1−/−HA-STAT1 MEFs. Heatmaps were generated using Java Treeview.

STAT1-reconstitution of Ifnar1−/− cells rescues IFNγ-inducible protein expression of genes involved in the MHC class I antigen presentation pathway

Several components of the MHC class I-dependent antigen presentation pathway were significantly rescued by STAT1 reconstitution of Ifnar1−/− cells as determined by microarray analysis (). We had previously found that STAT1 reconstitution of Ifnar1−/− cells rescued IFNγ-dependent induction of RNA expression for a key molecule in antigen presentation, β 2 microgloblin (B2M).Citation23 Therefore the MHC class I antigen presentation pathway represented an ideal candidate to validate the attenuation of IFNγ-dependent biological responses in IFNAR1 deficient cells and its rescue by STAT1 reconstitution.

We first examined the IFNγ-dependent induction of molecules involved in the transport of peptides to the endoplasmic reticulum, transporter associated with antigen processing (Tap)1 and Tap2, in Ifnar1−/− cells. In corroboration with the microarray data, we found reduced IFNγ-mediated protein induction of TAP1 expression in Ifnar1−/− MEFs compared to wild type MEFs following exposure to IFNγ for 6 hrs (). There was also attenuation of TAP2 induction in Ifnar1−/− MEFs in response to 6 hrs of IFNγ treatment and a minor effect of loss of IFNAR1 for cells treated for 24 hrs (). Reconstitution of STAT1 in Ifnar1−/−HA-STAT1 cells rescued IFNγ-mediated induction of TAP1 in response to 6 hrs and TAP2 following 6 and 24 hrs of treatment ((). Similar to the TAP molecules, induction of proteasome subunit, β type (PSMB)9, a catalytic component of the immunoproteasome, was attenuated in Ifnar1−/− cells following 6 hrs of treatment (). This attenuated induction of PSMB9 was also evident in Ifnar1−/−MSCV cells following 6 hrs of IFNγ treatment and importantly, was rescued to wild type equivalent levels in Ifnar1−/−HA-STAT1 cells (). Notably, PSMB9 expression reached wild type equivalent levels in all IFNAR1 deficient cell lines by 24 hrs of IFNγ treatment suggesting that, as seen in for multiple genes in the microarray analysis, IFNAR1 expression was required for early, but not late phases responses.

Figure 3. STAT1 reconstitution rescues IFN-induced protein expression of MHC class I antigen presentation proteins in Ifnar1−/− MEFs. (A-C) wild type, Ifnar1, Ifnar1−/−MSCV and Ifnar1−/−HA-STAT1 MEFs were cultured in media alone or treated with 100 U/ml IFNγ for (A) 6 hrs and (B-C) 24 hrs. Protein lysates were separated by SDS-PAGE and probed with antibodies specific for (A) TAP1 (B) TAP2 or (C) PSMB9. Western membranes were re-probed with antibody specific for (A-B) Actin or (C) HSP70 to confirm equivalent loading of proteins. (D-E) wild type, Ifnar1, Ifnar1−/−MSCV and Ifnar1−/−HA-STAT1 MEFs were cultured in media alone or were treated with 5 or 10 U/ml IFNγ for 16 hrs. Cells were analyzed by flow cytometry for expression of the MHC class I molecules (D) H2-Kb and (E) H2-Db. Data represents the IFNγ-induced change in MFI relative to untreated for 11–13 independent experiments and error bars are SEM. Significant changes in MFI relative to untreated (fold increase in MFI) were determined by t-test #p < 0.05, ##p < 0.01, ###p < 0.001. Significant differences in fold increase in MFI relative to wild type were determined by t-test *p < 0.05, **p < 0.01, ***p < 0.001.

Figure 3. STAT1 reconstitution rescues IFN-induced protein expression of MHC class I antigen presentation proteins in Ifnar1−/− MEFs. (A-C) wild type, Ifnar1, Ifnar1−/−MSCV and Ifnar1−/−HA-STAT1 MEFs were cultured in media alone or treated with 100 U/ml IFNγ for (A) 6 hrs and (B-C) 24 hrs. Protein lysates were separated by SDS-PAGE and probed with antibodies specific for (A) TAP1 (B) TAP2 or (C) PSMB9. Western membranes were re-probed with antibody specific for (A-B) Actin or (C) HSP70 to confirm equivalent loading of proteins. (D-E) wild type, Ifnar1, Ifnar1−/−MSCV and Ifnar1−/−HA-STAT1 MEFs were cultured in media alone or were treated with 5 or 10 U/ml IFNγ for 16 hrs. Cells were analyzed by flow cytometry for expression of the MHC class I molecules (D) H2-Kb and (E) H2-Db. Data represents the IFNγ-induced change in MFI relative to untreated for 11–13 independent experiments and error bars are SEM. Significant changes in MFI relative to untreated (fold increase in MFI) were determined by t-test #p < 0.05, ##p < 0.01, ###p < 0.001. Significant differences in fold increase in MFI relative to wild type were determined by t-test *p < 0.05, **p < 0.01, ***p < 0.001.

Finally, we assessed the effects of STAT1 on the ability of IFNγ to regulate cell surface expression of the MHC class I heavy chain on Ifnar1−/− cells. Again, in agreement with the microarray analysis, Ifnar1−/− and Ifnar1−/−MSCV MEFs displayed significantly attenuated IFNγ responses, with minimal changes in H2-Kb and H2Db expression compared to wild type cells (). In contrast, cell surface expression of H2-Kb and H2-Db following both 5 and 10 U/ml treatments of IFNγ was, like wild type cells, significantly induced on Ifnar1−/−HA-STAT1 cells (). Together, these data indicated that STAT1-reconstitution was able to rescue the attenuated IFNγ-dependent regulation of mRNA and protein expression of genes involved in protein processing, peptide transport and cell surface antigen presentation in Ifnar1−/− cells.

Constitutive IFNα/β signaling regulates the expression of multiple signaling molecules

Having found that STAT1 reconstitution of IFNAR1-deficient cells only rescued a proportion (∼15%) of IFNγ regulated genes and that these genes were enriched for involvement in immune and inflammatory responses, we investigated whether constitutive IFNα/β signaling regulated the expression of other transcription factors to modulate IFN crosstalk. We first examined expression of other members of the JAK/STAT signaling pathway known to be regulated by and involved in IFN signaling. In corroboration with our previous qRT-PCR findings, there was attenuated RNA expression of Stat1 and Stat2, but not Socs3, in unstimulated Ifnar1−/− cells, as determined by microarray and qRT-PCR analysis (Stat1 and Socs3) (Supp. Fig. 1A, Supp. Table 1–2).Citation23 Similarly, we validated our previously identified decrease in basal STAT3 protein expression using primary C57BL/6 Ifnar1−/− MEFs and confirmed that the attenuated STAT3 protein expression evident in 129/C3H MEFs lacking IFNAR1 was related to a significant decrease in RNA expression (Supp. Fig. 1A–B).Citation23 Given that both IFNγ and IL-6 can activate STAT3 and that, IL-6 mediated STAT3 DNA binding is attenuated in the absence of IFNAR1,Citation27 we assessed STAT3 activation following IFNγ or IL-6 treatment of Ifnar1−/− cells. In contrast to previous findings,Citation27 we found reduced STAT3 phosphorylation following IL-6 stimulation of Ifnar1−/− cells (). Similarly, IFNγ-mediated STAT3 activation was also attenuated in IFNAR1-deficient cells (). These data indicate that constitutive IFNα/β signaling regulates expression of multiple STAT transcription factors and that this may influence crosstalk with cytokines other than IFNs.

Figure 4. Aberrant expression and activation of signaling intermediaries in unstimulated Ifnar1−/− MEFs. (A) wild type and Ifnar1−/− MEFs were cultured in media alone or treated with 100 U/ml IFNγ or 100 ng/ml IL-6 and 100ng/ml IL-6R for 30mins. Protein lysates were separated by SDS-PAGE and probed with an antibody specific for pSTAT3. Western membrane was re-probed with an antibodies specific for Actin to confirm equivalent loading of proteins. (B) RNA was extracted from untreated wild type and Ifnar1−/− 129/C3H MEFs and mRNA expression of c-Jun, Irf1, Myd88, Nfkbiα and Nfkbie was determined by qRT-PCR. Expression of each gene was normalized to L32 and untreated wild type MEFs. Data represents the mean of 4–6 independent experiments and error bars are SEM. Significance determined by t-test *p < 0.05, **p < 0.01, ***p < 0.001. (C-D) Protein lysates from untreated wild type and Ifnar1−/−MEFs were separated by SDS-PAGE and probed with antibodies specific for (C) c-JUN, and (D) ERK1/2. Western membranes were re-probed with antibodies specific for HSP70 to confirm equivalent loading of proteins.

Figure 4. Aberrant expression and activation of signaling intermediaries in unstimulated Ifnar1−/− MEFs. (A) wild type and Ifnar1−/− MEFs were cultured in media alone or treated with 100 U/ml IFNγ or 100 ng/ml IL-6 and 100ng/ml IL-6R for 30mins. Protein lysates were separated by SDS-PAGE and probed with an antibody specific for pSTAT3. Western membrane was re-probed with an antibodies specific for Actin to confirm equivalent loading of proteins. (B) RNA was extracted from untreated wild type and Ifnar1−/− 129/C3H MEFs and mRNA expression of c-Jun, Irf1, Myd88, Nfkbiα and Nfkbie was determined by qRT-PCR. Expression of each gene was normalized to L32 and untreated wild type MEFs. Data represents the mean of 4–6 independent experiments and error bars are SEM. Significance determined by t-test *p < 0.05, **p < 0.01, ***p < 0.001. (C-D) Protein lysates from untreated wild type and Ifnar1−/−MEFs were separated by SDS-PAGE and probed with antibodies specific for (C) c-JUN, and (D) ERK1/2. Western membranes were re-probed with antibodies specific for HSP70 to confirm equivalent loading of proteins.

To determine the effect that loss of constitutive IFNα/β signaling has on the global transcriptional profile of resting cells, we assessed transcription factor expression in unstimulated wild type and Ifnar1−/− MEFs by microarray analysis. 1264 genes had ≥1.2-fold (p≤0.05) differential basal expression in Ifnar1−/− MEFs compared to wild type MEFs (Supp. Table. 1). Of the 1264 genes with differences in basal expression between wild type and Ifnar1−/− cells, 106 were known transcription factors (Supp. Table. 2). Furthermore, qRT-PCR analysis of intracellular signaling molecules known to crosstalk with the type I IFN signaling pathway confirmed the reduced expression of genes involved in TLR and NF-κB signaling, Myd88, Nfkbia and Nfkbie () in Ifnar1−/− cells ( and Supp. Table. 2).

We also assessed RNA and protein expression of several transcription factors known to regulate IFNγ responses that were not identified by the microarray analysis as having attenuated expression in Ifnar1−/− MEFs. We found significantly reduced RNA expression of the transcription factors c-Jun and Irf1 by qRT-PCR analysis (). As shown in , Ifnar1−/− MEFs also expressed less c-Jun protein than wild type MEFs and thus the decrease in c-Jun mRNA expression observed correlated with a similar decrease in total protein expression. Similarly, although we found no reduction in extracellular signal-regulated kinase (ERK)1/2 expression by microarray analysis, basal expression of ERK1, but not ERK2 was reduced in Ifnar1−/− MEFs (). Overall, these data revealed that the absence of IFNAR1 results in reduced aberrant expression of multiple transcription factors which are involved in a range of cytokine and inflammatory signaling responses.

DISCUSSION

The canonical signal transduction pathways activated by cytokines to modulate cellular responses and gene expression have been extensively investigated. These pathways are comprised of signaling intermediaries such as kinases, G proteins, adaptor proteins and transcription factors that get turned ‘on’ and ‘off’ in the presence of a stimulus. In contrast, the physiological scenario of concomitant exposure to several cytokines and the consequences and mechanisms regulating these interactions are relatively undefined.Citation28 Cytokine crosstalk can occur at the level of the receptor, by regulation of the expression of receptor components or the formation of receptor complexes.Citation19,27,29-31 Crosstalk can also occur at the level of gene promoters as a result of multiple promoter binding sites or chromatin priming.Citation32,33 In this study, we examined the impact that the regulation of the basal levels of intracellular signaling molecules has on cytokine responses.

We had previous established that STAT1 reconstitution of Ifnar1−/− cells rescued IFNγ-dependent STAT1 activation and DNA binding and partially rescued IFNγ-mediated protection from the cytopathic effects of ECMV.Citation23 Using microarray analysis to ascertain the effects of STAT1 reconstitution of IFNAR1-defient cells on a genome wide scale we found that although STAT1 is the canonical transcription factor activated by IFNγ, only up to 15.4% and 3.2% of IFNγ-dependent gene induction and repression were rescued by STAT1 respectively. Moreover, the ability of STAT1 to rescue IFNγ responses diminished over time. The relatively minor effect of STAT1 expression of IFNγ mediated gene repression was unsurprising however, as gene repression by IFNγ is purported to be mediated via STAT1 independent pathwaysCitation34 such as CIITA mediated histone deacetylation.Citation35-37 Also, as STAT1 is induced by IFNγ signaling,Citation38-40 it is plausible that at later time points de novo STAT1 production in Ifnar1−/− cells in response to IFNγ treatment was able to rescue the IFNγ responsiveness of genes at later time points.

Treatment of primary human fibroblasts with both IFNβ and IFNγ synergistically induces expression of genes involved in apoptosis and inflammatory responses.Citation41 Similarly, the biological response pathways which showed attenuated IFNγ responses in Ifnar1−/− cells and for which STAT1 reconstitution significantly rescued IFNγ responsiveness were both enriched for immune responses, apoptosis and alternate cytokine signaling pathways. As expected, our microarray and protein analysis indicated that IFNγ-dependent regulation of the JAK/STAT signaling pathway was significantly affected by the loss of tonic IFNα/β however, interestingly, the MHC class I antigen presentation pathway was the most sensitive to loss of IFNAR1 and was also significantly rescued by STAT1 reconstitution. Expression of a number of genes involved in MHC class I antigen presentation, including Tap1, Psmb9 and MHC class I heavy chain, are known to be mediated by type I and type II IFNs in a STAT1-dependent manner.Citation12,42-45 It was however surprising, given that this regulation is co-operatively mediated by other factors including IRF1, IRF2 and the ISGF3 complex for which basal expression was also reduced ( and Supp. Table 2), that STAT1 alone was able to rescue the IFNγ-dependent regulation of these genes.

Although reconstitution of STAT1 in Ifnar1−/− cells had a major effect on several biological pathways, ∼85% of IFNγ-mediated gene regulation was not rescued. Mediation of cytokine crosstalk by the regulating the expression of signaling intermediaries has also been suggested for STAT4,Citation46,47 IκB,Citation48 IRF9Citation49 and IRF2.Citation50 Our studies, which found that expression of over 100 transcription factors and numerous other signaling intermediaries, including Myd88, IKKγ and MAPK kinases, were decreased in cells that lacked tonic IFNα/β, suggests that rather than being regulated by a single transcription factor or signaling complex, cytokine crosstalk likely involves regulation of the expression of a multitude of signaling molecules.

This level of regulation may enhance specific biological responses when particular sets of transcription factors are involved. For example, although expression of STAT1 had a profound effect on IFNγ dependent regulation of genes involved in antigen presentation, other IFNγ-dependent responses, including regulation of genes involved in development and TLR-dependent immune responses, were not rescued by STAT1 reconstitution of Ifnar1−/- MEFs. As RNA expression of the signaling intermediary Myd88, a fundamental component of the TLR signaling pathway,Citation51,52 was, like STAT1, attenuated in cells lacking constitutive IFNβ signaling, it is plausible that tonic IFNα/β augments TLR responsiveness by regulating the expression of Myd88. Moreover, as expression or activation of transcription factors involved in signaling via other cytokines known to engage in crosstalk with IFNα/β such as IL-6, IL-1 and TNFαCitation6,27,28,53-55 were also revealed by our studies to be attenuated in IFNAR1-deficient cells, it suggest that tonic IFNα/β also regulates responsiveness to these cytokines by this mechanism.

Overall we have demonstrated that, in addition to maintenance of STAT1 expression, constitutive IFNα/β-signaling mediated by IFNAR1 regulates the expression of multiple transcription factors and signaling intermediaries. Moreover, based on the sub-set of biological responses pathways affected by STAT1 expression, we suggest that, the regulation of signaling molecules involved in specific biological pathways or activated by specific cytokines may be a mechanism whereby synergistic biological responses by multiple cytokines are fine-tuned.

MATERIALS AND METHODS

Mice

C57BL/6 J wild-type mice were purchased from the Walter and Eliza Hall Institute of Medical Research or bred in house at the Peter MacCallum Cancer Center. C57BL/6 IFNAR1-deficient (Ifnar1−/−)Citation56 mice were bred in house at the Peter MacCallum Cancer Center. All experiments were performed in accordance with the animal ethics guidelines ascribed by the National Health and Medical Research Council of Australia. All experiments were approved by the Peter MacCallum Cancer Center Animal Ethics Committee.

Reagents and cell culture

Generation of wild type and Ifnar1−/− 129/C3H MEFs and retroviral transduction of Ifnar1−/− 129/C3H MEFs was described previously.Citation23 Briefly, Ifnar1−/− 129/C3H MEFs were transduced with viral supernatants using murine stem cell virus (MSCV) encoding GFP alone or cDNA encoding HA-tagged STAT1 (generous gift from Thomas Decker). Primary MEFs were derived from day 13.5–14.5 wild type or Ifnar1−/− C57BL/6 embryos and used during early passages (passage 2–6). For all culture conditions, cells were maintained at 37°C in Dulbecco's Modified Eagle Medium (DMEM) (JRH Biosciences) supplemented with 10% foetal bovine serum (FBS) (Thermotrace), 2 mM L-glutamine (JRH Biosciences), 50µM 2-mercaptoethanol (Sigma Chemical Co.) and penicillin/streptomycin (Invitrogen Life Technologies Corp.). Recombinant cytokines and receptors utilised in these experiments were IFNγ (Roche, 11276905910), IL-6 (Peprotec, 200-06) and IL-6 receptor (IL-6R) (R & D systems Inc., 227-SR).

Microarray analysis

RNA was extracted from wild type, Ifnar1−/−, Ifnar1−/−MSCV and Ifnar1−/−HA-STAT1 transduced 129/C3H MEFs using Trizol (Invitrogen) according to the manufacturer's instructions. RNA quality was assessed by bioanalyzer using RNA Nano Chip (Agilent technologies) as per the manufacturer's instructions. RNA samples were considered degraded if RNA integrity number (RIN) was below 8.

All reagents (except 100% and 80%v/v Ethanol) were purchased from Affymetrix (Millennium Science) as part of the One-Cycle Target Labeling and Control Reagents (P/N 900493) package and GeneChip® Hybridization control kit (P/N 900454). cDNA preparation, sample hybridization and scanning of the oligonucleotide microarray GeneChip Arrays (Affymetrix) was performed as per manufacturer's instructions. Briefly 1µg RNA was reverse transcribed using Superscript II and T-7 Oligo (dT). Second strand cDNA synthesis, mediated by RNase H was followed by an in vitro transcription (IVT) reaction with T7 RNA polymerase, biotin-11-CTP and biotin-16-UTP to generate biotin labeled cRNA. Purified cRNA was fragmented by mild alkaline treatment, then hybridized to the GeneChip® Mouse Genome 430 2.0 Array Mouse chips (probe arrays) (Affymetrix) and washed using the GeneChip® Fluidics Station 450. Array chips were then stained with Streptavidin Phycoethrin (SAPE) (Molecular Probes) Biotinylated antibody (Vector Labs). Cartridges were loaded into autoloader of the GeneChip® Scanner 3000 7 G laser scanner.

The data (.dat) file, was automatically converted to a cell intensity (.cel) file and the quality of the individual arrays was assessed using GCOS. Microarray CEL files (Affymetrix probe identities and intensities) were converted to annotation files using Affy software (Affy).Citation57 All microarray data was assessed for degradation (degradation plots, Affy) normalized by Robust multi-array (RMA) normalization (Affy), p-values determined by Students t-test using Linear Models for Microarray Analysis (LIMMA) and multiple test corrections for each sample set comparison was performed using Benjamini-Hochberg multiple test correction method.Citation58 Significance cut offs of p ≤ 0.05 and fold change ≥ 1.2 for each comparison were implemented.

Quantitative real time PCR

RNA was extracted using Trizol (Invitrogen) according to the manufacturer's instructions. Alternatively, RNA was extracted using the RNeasy Plus Mini Kit (QIAGEN) as per manufacturer's instructions. cDNA was synthesized from 1 µg RNA using superscript III (Invitrogen) as per the manufacturer's instructions. The abundance of specific genes in the samples was quantitated using the SYBR Green dye detection method (Applied Biosystems, CA, USA). Primers to murine genes were as follows.

c-Jun ACTCCGAGCTGGCATCCA CCCACTGTTAACGTGGTTCATG

Irf1 TGGAGATGTTAGCCCGGACACTTT ACGGTGACAGTGCTGGAGTTATGT

L32 TTCCTGGTCCACAACGTCAAG TGTGAGCGATCTCGGCAC

Myd88 AGCAAGGAATGTGACTTCCAGACCA TGGGAAAGTCCTTCTTCATCGCCT

Nfkbia TGGCCTTCCTCAACTTCCAGAACA TCAGGATCACAGCCAGCTTTCAGA

Nfkbie ACATTGATGTACAGGAGGGCACGA ACAGAGTGGATGAGATGCTGTTGAGG

Socs3 CCTTCAGCTCCAAAAGCGAG GCTCTCCTGCAGCTTGCG

Stat1 CGCGCATGCAACTGGCATATAACT AAGCTCGAACCACTGTGACATCCT

Stat3 AGTTCCTGGCACCTTGGATTGAGA TTGGACTCTTGCACCAATCGGCTA

Threshold cycle numbers (Ct) were measured in the exponential phase for all samples. Relative abundance of sample genes was calculated using the “DDCT method” relative to the L32 control gene. mRNA abundance was normalized to the untreated samples of each genotype or relative to wild type as indicated.

Flow cytometry

Samples were first incubated in FC block in 2% FBS/PBS for 20 minutes. Antibodies against H2Db (28-14-8) and H2Kb (AF6-88.5) were obtained from BD Pharmingen. Viability was determined by absence of Fluoro-gold uptake.

SDS-PAGE and protein gel blotting

Cells were washed once in PBS and lysed in whole cell lysis buffer (0.05% Triton X-100, 50 mM Tris, pH 7.5, 150 mM NaCl, 0.1 mM ethylene diamine tetra-acetic acid, 0.1 mM ethylene glycol tetra-acetic acid and 10% glycerol), containing protease and phosphatase inhibitors (1mg/mL aprotinin,0.5 mg/mL leupeptin, 0.2 mM PMSF, 10 mM NaF, 1 mM Na3VO4, 1 mM β glycero-phosphate) at 4°C for 10 min. Lysates were then cleared by centrifugation. Proteins were separated by SDS-PAGE and transferred to PVDF membranes. Membranes were probed with specific antibodies against Actin (Sigma), HSP70 (kind gift from R. Anderson), STAT1α/β (C terminus) (BD Bioscience), p-STAT3, STAT3, c-JUN, ERK1/2 (Cell Signaling Technology), PSMB9 (Abcam), TAP1 or TAP2 (Santa Cruz Biotechnology). Secondary antibodies were conjugated to horseradish peroxidase and images were visualized by chemiluminescence (ECL, GE Healthcare).

Statistical analysis

Statistical significance was assessed using Microsoft Excel software. For comparisons of means, Student's t-Test (t-test) was used and p-values <0.05 were considered significant. For cluster analysis of microarray probes, k-means organized arrays via Euclidean distance were displayed as logbase2. For cluster analysis of antigen presentation genes, hierarchical organized arrays via Euclidean distance were displayed as logbase2. Cluster analysis was performed using Cluster 3.0Citation59 and visualised using Java Tree View.Citation60 Pathway enrichment analysis and identification of transcription factors was performed using MetaCore from Thomson Reuters (MetaCore Genego, Thomson Reuters, https://portal.genego.com/Genego).

ABBREVIATIONS

B2M=

Beta 2 microgloblin

ERK=

Extracellular signal-regulated kinase

Hrs=

Hours

IFN=

Interferon

IFNAR=

IFNα/β receptor

IFNAR1=

IFNα/β receptor α chain

IL=

Interleukin

IRF=

Interferon regulatory factor

JAK=

Janus activated kinase

MAPK=

Mitogen activated protein kinase

MEF=

Mouse embryonic fibroblast

MSCV=

Murine stem cell virus

NFκB=

Nuclear factor κB

PSMB=

Proteasome subunit,β type

STAT=

Signal transducer and activator of transcription

TAP=

Transporter associated with antigen processing

TGF=

Transforming growth factor

TNF=

Tumor necrosis factor

U/ml=

Units per ml

DISCLOSURE OF POTENTIAL CONFLICTS OF INTEREST

No potential conflicts of interest were disclosed.

Supplemental material

Supplementary_material.zip

Download Zip (315.1 KB)

ACKNOWLEDGMENTS

We thank Natalie Thomson for bioinformatics analysis and QC of microarray data. We also thank Dr Edwin Hawkins, Dr Sabine Hoves and Linda Hii for technical help.

Funding

The research reported in this publication was supported by the Cancer Research Institute (CRI scholarship), a Program Grant from the National Health and Medical Research Council of Australia (NHMRC) and a project grant from the Cancer Council Victoria. RWJ is an NHMRC Senior Principal Research Fellow.

REFERENCES

  • Bartee E, Mohamed MR, McFadden G. Tumor necrosis factor and interferon: cytokines in harmony. Curr Opin Microbiol 2008; 11:378-83; PMID:18595771; http://dx.doi.org/10.1016/j.mib.2008.05.015
  • Paludan SR. Synergistic action of pro-inflammatory agents: cellular and molecular aspects. J Leukoc Biol 2000; 67:18-25; PMID:10647993.
  • Bartee E, Mohamed MR, Lopez MC, Baker HV, McFadden G. The addition of tumor necrosis factor plus β interferon induces a novel synergistic antiviral state against poxviruses in primary human fibroblasts. J Virol 2009; 83:498-511; PMID:18971273; http://dx.doi.org/10.1128/JVI.01376-08
  • Yarilina A, Park-Min KH, Antoniv T, Hu X, Ivashkiv LB. TNF activates an IRF1-dependent autocrine loop leading to sustained expression of chemokines and STAT1-dependent type I interferon-response genes. Nat Immunol 2008; 9:378-87; PMID: 18345002; http://dx.doi.org/10.1038/ni1576
  • Pfeffer LM, Kim JG, Pfeffer SR, Carrigan DJ, Baker DP, Wei L, Homayouni R. Role of nuclear factor-kappaB in the antiviral action of interferon and interferon-regulated gene expression. J Biol Chem 2004; 279:31304-11; PMID:15131130; http://dx.doi.org/10.1074/jbc.M308975200
  • Onozaki K, Urawa H, Tamatani T, Iwamura Y, Hashimoto T, Baba T, Suzuki H, Yamada M, Yamamoto S, Oppenheim JJ, et al. Synergistic interactions of interleukin 1, interferon-β, and tumor necrosis factor in terminally differentiating a mouse myeloid leukemic cell line (M1). Evidence that interferon-β is an autocrine differentiating factor. J Immunol 1988; 140:112-9; PMID:3275716.
  • Gao PQ, Sims SH, Chang DC, Deisseroth AB. Interferon-gamma priming effects in the activation and deactivation of ISGF3 in K562 cells. J Biol Chem 1993; 268:12380-7; PMID:8509377.
  • Improta T, Pine R, Pfeffer LM. Interferon-gamma potentiates the antiviral activity and the expression of interferon-stimulated genes induced by interferon-α in U937 cells. J Interferon Res 1992; 12:87-94; PMID:1315834; http://dx.doi.org/10.1089/jir.1992.12.87
  • Sainz B, Jr, Halford WP. Alpha/Beta interferon and gamma interferon synergize to inhibit the replication of herpes simplex virus type 1. J Virol 2002; 76:11541-50; PMID:12388715; http://dx.doi.org/10.1128/JVI.76.22.11541-11550.2002
  • Mossel EC, Sainz B, Jr, Garry RF, Peters CJ. Synergistic inhibition of SARS-coronavirus replication by type I and type II IFN. Adv Exp Med Biol 2006; 581:503-6; PMID:17037585; http://dx.doi.org/10.1007/978-0-387-33012-9_89
  • Durbin JE, Hackenmiller R, Simon MC, Levy DE. Targeted disruption of the mouse Stat1 gene results in compromised innate immunity to viral disease. Cell 1996; 84:443-50; PMID:8608598; http://dx.doi.org/10.1016/S0092-8674(00)81289-1
  • Meraz MA, White JM, Sheehan KC, Bach EA, Rodig SJ, Dighe AS, Kaplan DH, Riley JK, Greenlund AC, Campbell D, et al. Targeted disruption of the Stat1 gene in mice reveals unexpected physiologic specificity in the JAK-STAT signaling pathway. Cell 1996; 84:431-42; PMID:8608597; http://dx.doi.org/10.1016/S0092-8674(00)81288-X
  • Gil MP, Bohn E, O'Guin AK, Ramana CV, Levine B, Stark GR, Virgin HW, Schreiber RD. Biologic consequences of Stat1-independent IFN signaling. Proc Natl Acad Sci U S A 2001; 98:6680-5; PMID:11390995; http://dx.doi.org/10.1073/pnas.111163898
  • Ramana CV, Gil MP, Schreiber RD, Stark GR. Stat1-dependent and -independent pathways in IFN-gamma-dependent signaling. Trends Immunol 2002; 23:96-101; PMID:11929133; http://dx.doi.org/10.1016/S1471-4906(01)02118-4
  • Bach EA, Aguet M, Schreiber RD. The IFN gamma receptor: a paradigm for cytokine receptor signaling. Annu Rev Immunol 1997; 15:563-91; PMID:9143700; http://dx.doi.org/10.1146/annurev.immunol.15.1.563
  • Gough DJ, Levy DE, Johnstone RW, Clarke CJ. IFNgamma signaling-does it mean JAK-STAT? Cytokine Growth Factor Rev 2008; 19:383-94; PMID:18929502; http://dx.doi.org/10.1016/j.cytogfr.2008.08.004
  • Dezfouli S, Hatzinisiriou I, Ralph SJ. Enhancing CTL responses to melanoma cell vaccines in vivo: synergistic increases obtained using IFNgamma primed and IFNbeta treated B7-1+ B16-F10 melanoma cells. Immunol Cell Biol 2003; 81:459-71; PMID:14636243; http://dx.doi.org/10.1046/j.0818-9641.2003.01189.x
  • Robinson CM, Shirey KA, Carlin JM. Synergistic transcriptional activation of indoleamine dioxygenase by IFN-gamma and tumor necrosis factor-α. J Interferon Cytokine Res 2003; 23:413-21; PMID:13678429; http://dx.doi.org/10.1089/107999003322277829
  • Takaoka A, Mitani Y, Suemori H, Sato M, Yokochi T, Noguchi S, Tanaka N, Taniguchi T. Cross talk between interferon-gamma and -α/β signaling components in caveolar membrane domains. Science 2000; 288:2357-60; PMID:10875919; http://dx.doi.org/10.1126/science.288.5475.2357
  • Phipps-Yonas H, Seto J, Sealfon SC, Moran TM, Fernandez-Sesma A. Interferon-β pretreatment of conventional and plasmacytoid human dendritic cells enhances their activation by influenza virus. PLoS Pathog 2008; 4:e1000193; PMID:18974865; http://dx.doi.org/10.1371/journal.ppat.1000193
  • Hamilton JA, Whitty GA, Kola I, Hertzog PJ. Endogenous IFN-α β suppresses colony-stimulating factor (CSF)-1-stimulated macrophage DNA synthesis and mediates inhibitory effects of lipopolysaccharide and TNF-α. J Immunol 1996; 156:2553-7; PMID:8786318.
  • Vogel SN, Fertsch D. Endogenous interferon production by endotoxin-responsive macrophages provides an autostimulatory differentiation signal. Infect Immun 1984; 45:417-23; PMID:6378797.
  • Gough DJ, Messina NL, Hii L, Gould JA, Sabapathy K, Robertson AP, Trapani JA, Levy DE, Hertzog PJ, Clarke CJ, et al. Functional crosstalk between type I and II interferon through the regulated expression of STAT1. PLoS Biol 2010; 8:e1000361; PMID:20436908; http://dx.doi.org/10.1371/journal.pbio.1000361
  • Nickolaus P, Zawatzky R. Inhibition by interleukin-4 of constitutive β interferon synthesis in mouse macrophages. J Virol 1994; 68:6763-6; PMID:8084010.
  • Karaghiosoff M, Neubauer H, Lassnig C, Kovarik P, Schindler H, Pircher H, McCoy B, Bogdan C, Decker T, Brem G, et al. Partial impairment of cytokine responses in Tyk2-deficient mice. Immunity 2000; 13:549-60; PMID:11070173; http://dx.doi.org/10.1016/S1074-7613(00)00054-6
  • Park C, Li S, Cha E, Schindler C. Immune response in Stat2 knockout mice. Immunity 2000; 13:795-804; PMID:11163195; http://dx.doi.org/10.1016/S1074-7613(00)00077-7
  • Mitani Y, Takaoka A, Kim SH, Kato Y, Yokochi T, Tanaka N, Taniguchi T. Cross talk of the interferon-α/β signalling complex with gp130 for effective interleukin-6 signalling. Genes Cells 2001; 6:631-40; PMID:11473581; http://dx.doi.org/10.1046/j.1365-2443.2001.00448.x
  • Bartee E, McFadden G. Cytokine synergy: an underappreciated contributor to innate anti-viral immunity. Cytokine 2013; 63:237-40; PMID:23693158; http://dx.doi.org/10.1016/j.cyto.2013.04.036
  • Rayamajhi M, Humann J, Penheiter K, Andreasen K, Lenz LL. Induction of IFN-alphabeta enables Listeria monocytogenes to suppress macrophage activation by IFN-gamma. J Exp Med 2010; 207:327-37; PMID:20123961; http://dx.doi.org/10.1084/jem.20091746
  • Ruggiero V, Tavernier J, Fiers W, Baglioni C. Induction of the synthesis of tumor necrosis factor receptors by interferon-gamma. J Immunol 1986; 136:2445-50; PMID:3005411.
  • Aggarwal BB, Eessalu TE, Hass PE. Characterization of receptors for human tumour necrosis factor and their regulation by gamma-interferon. Nature 1985; 318:665-7; PMID:3001529; http://dx.doi.org/10.1038/318665a0
  • Clarke DL, Clifford RL, Jindarat S, Proud D, Pang L, Belvisi M, Knox AJ. TNFalpha and IFNgamma synergistically enhance transcriptional activation of CXCL10 in human airway smooth muscle cells via STAT-1, NF-kappaB, and the transcriptional coactivator CREB-binding protein. J Biol Chem 2010; 285:29101-10; PMID:20833730; http://dx.doi.org/10.1074/jbc.M109.099952
  • Qiao Y, Giannopoulou EG, Chan CH, Park SH, Gong S, Chen J, Hu X, Elemento O, Ivashkiv LB. Synergistic activation of inflammatory cytokine genes by interferon-gamma-induced chromatin remodeling and toll-like receptor signaling. Immunity 2013; 39:454-69; PMID:24012417; http://dx.doi.org/10.1016/j.immuni.2013.08.009
  • Klampfer L, Huang J, Kaler P, Sasazuki T, Shirasawa S, Augenlicht L. STAT1-independent inhibition of cyclooxygenase-2 expression by IFNgamma; a common pathway of IFNgamma-mediated gene repression but not gene activation. Oncogene 2007; 26:2071-81; PMID:17016440; http://dx.doi.org/10.1038/sj.onc.1210015
  • Xu Y, Harton JA, Smith BD. CIITA mediates interferon-gamma repression of collagen transcription through phosphorylation-dependent interactions with co-repressor molecules. J Biol Chem 2008; 283:1243-56; PMID:17991736; http://dx.doi.org/10.1074/jbc.M707180200
  • Li P, Zhao Y, Wu X, Xia M, Fang M, Iwasaki Y, Sha J, Chen Q, Xu Y, Shen A. Interferon gamma (IFN-gamma) disrupts energy expenditure and metabolic homeostasis by suppressing SIRT1 transcription. Nucleic Acids Res 2012; 40:1609-20; PMID:22064865; http://dx.doi.org/10.1093/nar/gkr984
  • Weng X, Cheng X, Wu X, Xu H, Fang M, Xu Y. Sin3B mediates collagen type I gene repression by interferon gamma in vascular smooth muscle cells. Biochem Biophys Res Commun 2014; 447:263-70; PMID:24709079; http://dx.doi.org/10.1016/j.bbrc.2014.03.140
  • Der SD, Zhou A, Williams BR, Silverman RH. Identification of genes differentially regulated by interferon α, β, or gamma using oligonucleotide arrays. Proc Natl Acad Sci U S A 1998; 95:15623-8; PMID:9861020; http://dx.doi.org/10.1073/pnas.95.26.15623
  • Hu X, Herrero C, Li WP, Antoniv TT, Falck-Pedersen E, Koch AE, Woods JM, Haines GK, Ivashkiv LB. Sensitization of IFN-gamma Jak-STAT signaling during macrophage activation. Nat Immunol 2002; 3:859-66; PMID:12172544; http://dx.doi.org/10.1038/ni828
  • Wong LH, Hatzinisiriou I, Devenish RJ, Ralph SJ. IFN-gamma priming up-regulates IFN-stimulated gene factor 3 (ISGF3) components, augmenting responsiveness of IFN-resistant melanoma cells to type I IFNs. J Immunol 1998; 160:5475-84; PMID:9605150.
  • Peng T, Zhu J, Hwangbo Y, Corey L, Bumgarner RE. Independent and cooperative antiviral actions of β interferon and gamma interferon against herpes simplex virus replication in primary human fibroblasts. J Virol 2008; 82:1934-45; PMID:18057251; http://dx.doi.org/10.1128/JVI.01649-07
  • Muller M, Laxton C, Briscoe J, Schindler C, Improta T, Darnell JE, Jr, Stark GR, Kerr IM. Complementation of a mutant cell line: central role of the 91 kDa polypeptide of ISGF3 in the interferon-α and -gamma signal transduction pathways. EMBO J 1993; 12:4221-8; PMID:7693454.
  • Brucet M, Marques L, Sebastian C, Lloberas J, Celada A. Regulation of murine Tap1 and Lmp2 genes in macrophages by interferon gamma is mediated by STAT1 and IRF-1. Genes Immun 2004; 5:26-35; PMID:14735146; http://dx.doi.org/10.1038/sj.gene.6364035
  • Rouyez MC, Lestingi M, Charon M, Fichelson S, Buzyn A, Dusanter-Fourt I. IFN regulatory factor-2 cooperates with STAT1 to regulate transporter associated with antigen processing-1 promoter activity. J Immunol 2005; 174:3948-58; PMID:15778351; http://dx.doi.org/10.4049/jimmunol.174.7.3948
  • Min W, Pober JS, Johnson DR. Kinetically coordinated induction of TAP1 and HLA class I by IFN-gamma: the rapid induction of TAP1 by IFN-gamma is mediated by Stat1 α. J Immunol 1996; 156:3174-83; PMID:8617938.
  • Miyagi T, Gil MP, Wang X, Louten J, Chu WM, Biron CA. High basal STAT4 balanced by STAT1 induction to control type 1 interferon effects in natural killer cells. J Exp Med 2007; 204:2383-96; PMID:17846149; http://dx.doi.org/10.1084/jem.20070401
  • Huang Y, Lei Y, Zhang H, Hou L, Zhang M, Dayton AI. MicroRNA regulation of STAT4 protein expression: rapid and sensitive modulation of IL-12 signaling in human natural killer cells. Blood 2011; 118:6793-802; PMID:22077060; http://dx.doi.org/10.1182/blood-2011-05-356162
  • Auphan N, Di Donato JA, Rosette C, Helmberg A, Karin M. Immunosuppression by glucocorticoids: inhibition of NF-kappa B activity through induction of I kappa B synthesis. Science 1995; 270:286-90; PMID:7569976; http://dx.doi.org/10.1126/science.270.5234.286
  • Weihua X, Hu J, Roy SK, Mannino SB, Kalvakolanu DV. Interleukin-6 modulates interferon-regulated gene expression by inducing the ISGF3 gamma gene using CCAAT/enhancer binding protein-β(C/EBP-β). Biochim Biophys Acta 2000; 1492:163-71; PMID:11004486; http://dx.doi.org/10.1016/S0167-4781(00)00111-1
  • Hayes MP, Zoon KC. Priming of human monocytes for enhanced lipopolysaccharide responses: expression of α interferon, interferon regulatory factors, and tumor necrosis factor. Infect Immun 1993; 61:3222-7; PMID:8335353.
  • von Bernuth H, Picard C, Puel A, Casanova JL. Experimental and natural infections in MyD88- and IRAK-4-deficient mice and humans. Eur J Immunol 2012; 42:3126-35; PMID:23255009; http://dx.doi.org/10.1002/eji.201242683
  • Kawai T, Adachi O, Ogawa T, Takeda K, Akira S. Unresponsiveness of MyD88-deficient mice to endotoxin. Immunity 1999; 11:115-22; PMID:10435584; http://dx.doi.org/10.1016/S1074-7613(00)80086-2
  • Gouwy M, Struyf S, Proost P, Van Damme J. Synergy in cytokine and chemokine networks amplifies the inflammatory response. Cytokine Growth Factor Rev 2005; 16:561-80; PMID:16023396; http://dx.doi.org/10.1016/j.cytogfr.2005.03.005
  • Sharif MN, Tassiulas I, Hu Y, Mecklenbrauker I, Tarakhovsky A, Ivashkiv LB. IFN-α priming results in a gain of proinflammatory function by IL-10: implications for systemic lupus erythematosus pathogenesis. J Immunol 2004; 172:6476-81; PMID:15128840; http://dx.doi.org/10.4049/jimmunol.172.10.6476
  • Reis LF, Ho Lee T, Vilcek J. Tumor necrosis factor acts synergistically with autocrine interferon-β and increases interferon-β mRNA levels in human fibroblasts. J Biol Chem 1989; 264:16351-4; PMID:2550437.
  • Hwang SY, Hertzog PJ, Holland KA, Sumarsono SH, Tymms MJ, Hamilton JA, Whitty G, Bertoncello I, Kola I. A null mutation in the gene encoding a type I interferon receptor component eliminates antiproliferative and antiviral responses to interferons α and β and alters macrophage responses. Proc Natl Acad Sci U S A 1995; 92:11284-8; PMID:7479980; http://dx.doi.org/10.1073/pnas.92.24.11284
  • Irizarry RA, Hobbs B, Collin F, Beazer-Barclay YD, Antonellis KJ, Scherf U, Speed TP. Exploration, normalization, and summaries of high density oligonucleotide array probe level data. Biostatistics 2003; 4:249-64; PMID:12925520; http://dx.doi.org/10.1093/biostatistics/4.2.249
  • Smyth GK. Linear models and empirical bayes methods for assessing differential expression in microarray experiments. Stat Appl Genet Mol Biol 2004; 3:1-25; PMID:16646809.
  • de Hoon MJ, Imoto S, Nolan J, Miyano S. Open source clustering software. Bioinformatics 2004; 20:1453-4; PMID:14871861; http://dx.doi.org/10.1093/bioinformatics/bth078
  • Saldanha AJ. Java Treeview–extensible visualization of microarray data. Bioinformatics 2004; 20:3246-8; PMID:15180930; http://dx.doi.org/10.1093/bioinformatics/bth349

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.