
Ebolaviruses Associated with Differential Pathogenicity Induce Distinct Host Responses in Human Macrophages
Judith Olejnik
Adriana Forero
Laure R Deflubé
Adam J Hume
Whitney A Manhart
Andrew Nishida
Andrea Marzi
Michael G Katze
Hideki Ebihara
Angela L Rasmussen
Elke Mühlberger
Address correspondence to Elke Mühlberger,muehlber@bu.edu.
Present address: Hideki Ebihara, Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA; Angela L. Rasmussen, Center for Infection and Immunity, Columbia University, New York, New York, USA.
Citation Olejnik J, Forero A, Deflubé LR, Hume AJ, Manhart WA, Nishida A, Marzi A, Katze MG, Ebihara H, Rasmussen AL, Mühlberger E. 2017. Ebolaviruses associated with differential pathogenicity induce distinct host responses in human macrophages. J Virol 91:e00179-17.https://doi.org/10.1128/JVI.00179-17.
Corresponding author.
Roles
Received 2017 Feb 6; Accepted 2017 Mar 8; Prepublished 2017 Mar 22; Collection date 2017 Jun 1.
ABSTRACT
Ebola virus (EBOV) and Reston virus (RESTV) are members of theEbolavirus genus which greatly differ in their pathogenicity. While EBOV causes a severe disease in humans characterized by a dysregulated inflammatory response and elevated cytokine and chemokine production, there are no reported disease-associated human cases of RESTV infection, suggesting that RESTV is nonpathogenic for humans. The underlying mechanisms determining the pathogenicity of different ebolavirus species are not yet known. In this study, we dissected the host response to EBOV and RESTV infection in primary human monocyte-derived macrophages (MDMs). As expected, EBOV infection led to a profound proinflammatory response, including strong induction of type I and type III interferons (IFNs). In contrast, RESTV-infected macrophages remained surprisingly silent. Early activation of IFN regulatory factor 3 (IRF3) and NF-κB was observed in EBOV-infected, but not in RESTV-infected, MDMs. In concordance with previous results, MDMs treated with inactivated EBOV and Ebola virus-like particles (VLPs) induced NF-κB activation mediated by Toll-like receptor 4 (TLR4) in a glycoprotein (GP)-dependent manner. This was not the case in cells exposed to live RESTV, inactivated RESTV, or VLPs containing RESTV GP, indicating that RESTV GP does not trigger TLR4 signaling. Our results suggest that the lack of immune activation in RESTV-infected MDMs contributes to lower pathogenicity by preventing the cytokine storm observed in EBOV infection. We further demonstrate that inhibition of TLR4 signaling abolishes EBOV GP-mediated NF-κB activation. This finding indicates that limiting the excessive TLR4-mediated proinflammatory response in EBOV infection should be considered as a potential supportive treatment option for EBOV disease.
IMPORTANCE Emerging infectious diseases are a major public health concern, as exemplified by the recent devastating Ebola virus (EBOV) outbreak. Different ebolavirus species are associated with widely varying pathogenicity in humans, ranging from asymptomatic infections for Reston virus (RESTV) to severe disease with fatal outcomes for EBOV. In this comparative study of EBOV- and RESTV-infected human macrophages, we identified key differences in host cell responses. Consistent with previous data, EBOV infection is associated with a proinflammatory signature triggered by the surface glycoprotein (GP), which can be inhibited by blocking TLR4 signaling. In contrast, infection with RESTV failed to stimulate a strong host response in infected macrophages due to the inability of RESTV GP to stimulate TLR4. We propose that disparate proinflammatory host signatures contribute to the differences in pathogenicity reported for ebolavirus species and suggest that proinflammatory pathways represent an intriguing target for the development of novel therapeutics.
KEYWORDS: chemokines, cytokines, Ebola virus, filovirus, host response, interferons, macrophages, Reston virus, Toll-like receptor 4, Toll-like receptors
INTRODUCTION
Ebolaviruses are subdivided into five distinct species whose members not only show significant molecular differences but also vary in terms of virulence and pathogenicity. Ebola virus (EBOV), which belongs to theZaire ebolavirus species, shows the highest pathogenicity in humans, with case fatality rates ranging from 40 to 90%. The EBOV Makona variant caused the recent unprecedented outbreak in West Africa (1). EBOV disease is characterized by an uncontrolled systemic infection, leading to high viral titers, coagulation abnormalities, vascular leakage, a dysregulated immune response as reflected by elevated cytokine and chemokine levels, and organ failure (2–9). In contrast, Reston virus (RESTV), which belongs to theReston ebolavirus species, has not yet been associated with human disease. Individuals exposed to RESTV-infected animals seroconverted without showing any signs of disease, suggesting that RESTV is strongly attenuated in humans (10). Compared to EBOV, RESTV is either nonpathogenic or less pathogenic in different nonhuman primate species (11,12), and it causes no or less severe disease in IFNAR−/− and STAT1−/− mice (13–16), suggesting that RESTV is generally less pathogenic across species. EBOV is known to efficiently suppress the type I interferon (IFN) response and expresses at least two IFN antagonists, the viral protein 35 (VP35) and VP24 (reviewed in references17 and18). RESTV VP35 and VP24 have also been shown to antagonize the type I IFN response, albeit less efficiently (19–24). The primaryin vivo target cells of ebolaviruses are thought to be cells of the mononuclear phagocyte system, including monocytes, macrophages, and myeloid dendritic cells (9,25). Primary human monocyte-derived macrophages (MDMs) are activated upon EBOV infectionin vitro, as shown by the induction and secretion of cytokines reported in various studies (6,9,26–30). Cytokine induction was observed at early time points postinfection in MDMs infected with live or inactivated virus or with virus-like particles (VLPs). It has been reported that this activation is triggered by the interaction of the EBOV glycoprotein (GP) with Toll-like receptor 4 (TLR4) (6,27–33). In addition, GP has been shown to interact with C-type lectin domain family 4 member G (CLEC4G/LSECtin) on monocyte-derived dendritic cells, leading to the induction of an inflammatory response (34). A hallmark of severe EBOV disease and a predictor of fatal outcome is the induction of a strong inflammatory response as shown by elevated levels of inflammatory cytokines and chemokines (2–6,8,35). It is not known if RESTV infection elicits an inflammatory response in humans.
In this study, we uncovered differences in the host response of human MDMs to infection with the highly pathogenic EBOV and the presumably nonpathogenic RESTV. While EBOV infection of MDMs led to a profound proinflammatory response, including IFN regulatory factor 3 (IRF3) and NF-κB activation and the induction of type I and type III IFNs, RESTV failed to activate similar immune signaling in infected cells. We confirmed previous reports showing that the EBOV surface GP induces cytokine responses via TLR4-mediated signaling and showed that this is not the case for RESTV GP, offering an explanation for the weak cytokine responses. Finally, we showed that blocking TLR4 signaling mutes the EBOV GP-mediated inflammatory response.
RESULTS
RESTV infection is not associated with strong changes in gene expression.
To compare the host response to infection with highly pathogenic and presumably nonpathogenic ebolaviruses, human MDMs were infected with purified, endotoxin-free EBOV or RESTV particles at a multiplicity of infection (MOI) of 5 to 7. As controls, cells were treated with lipopolysaccharide (LPS) or were mock infected. Samples were prepared at different time points starting at 6 h up to 4 days postinfection (dpi). At the desired time points, total cellular RNA was isolated for transcriptional analysis (RNA sequencing [RNA-Seq] and quantitative reverse transcription-PCR [qRT-PCR]) and cell supernatant cytokine concentrations were determined by Luminex analysis. Immunofluorescence analysis was performed to visualize infected cells.
We detected viral inclusions indicative of established infection by immunofluorescence analysis in both EBOV- and RESTV-infected MDMs at comparable rates (about 65%) at 1 dpi (Fig. 1A andB). About 90% of the cells were infected with EBOV or RESTV at 4 dpi (data not shown). For a quantitative comparison of infection, we analyzed VP35 mRNA levels. Alignment of viral RNA reads generated by RNA-Seq and qRT-PCR analysis was performed on mRNA isolated from EBOV- and RESTV-infected MDMs. While the amount of VP35 mRNA was lower in RESTV-infected MDMs than in EBOV-infected cells at 6 h postinfection (hpi), the observed difference was not statistically significant (Fig. 1C andD). Consistent with the immunofluorescence data, EBOV and RESTV VP35 mRNA levels were similar at 1 dpi (Fig. 1A,C, andD).
FIG 1.
MDMs are infected with EBOV and RESTV at comparable rates. (A) MDMs grown in chamber slides were infected with EBOV or RESTV or left uninfected (Mock) and examined by immunofluorescence analysis at 1 dpi using antibodies directed against EBOV NP or RESTV NP (red). Cell nuclei were stained with DAPI (blue). Infection was performed in five separate donors in independent experiment; representative images are shown. (B) Infection rate was determined by counting infected MDMs from five different donors as described for panel A. (C) RNA-Seq analysis was performed with samples generated from three different donors in independent experiments. Reads mapping to the VP35 sequence in the EBOV or RESTV genomes were counted using HTseq-count. Horizontal bars represent mean values; each symbol represents an individual donor. EBOV and RESTV VP35 expression levels were compared using two-tailedt test (GraphPad Prism 5 software), and no statistically significant differences were determined (P > 0.05 for all samples). (D) qRT-PCR analysis of EBOV and RESTV VP35 mRNA. Experiments were performed in duplicate using MDMs obtained from the same 3 individual donors as used for RNA-Seq in panel C. EBOV and RESTV VP35 expression levels were compared using two-tailedt test. *,P ≤ 0.05.
The RNA-Seq data set was used to compare host gene expression profiles across samples. Statistical analysis of differential host gene expression showed significant changes in the expression of hundreds of genes in EBOV-infected MDMs at 1 and 2 dpi (Fig. 2A). In stark contrast to the strong transcriptional response to EBOV infection, only a few cellular genes were differentially expressed (DE) in RESTV-infected MDMs at these time points. The DE genes induced in RESTV-infected MDMs early and late in infection overlapped with those identified in EBOV-infected cells; about one-third of the DE genes identified in RESTV infection were also identified as DE in EBOV infection (Fig. 2B). However, the fold change in gene expression was generally less in the RESTV-infected samples than in the EBOV-infected samples, indicating a weak host response to RESTV infection. The overall gene expression pattern observed in EBOV infection was strikingly similar to that observed in LPS-treated MDMs (Fig. 2B). To better understand the functional role of genes affected by ebolavirus infection and LPS stimulation, genes identified as DE were clustered on the basis of their coexpression. This hierarchical clustering revealed four modules of gene expression (Fig. 2C). Module 1 (light blue; 572 genes) contained innate immune effector genes and genes encoding chemokines involved in immune cell recruitment (Table 1). Genes within this module were found to be profoundly upregulated in EBOV infection and LPS stimulation, whereas only a subset of these genes was impacted by RESTV infection. Similar observations were made for module 2 genes (dark blue; 1,297 genes), which are involved in antigen presentation and innate immune cellular responses (Table 1). Significant downregulation in genes involved in the epithelial tissue integrity, G-protein-coupled receptor (GPCR) signaling, and cell cycle control (grouped within module 3 (939 genes; orange) and module 4 (805 genes; red) was observed in both EBOV infection and LPS treatment (Fig. 2C;Table 1). In contrast, RESTV infection was not correlated with strong inhibition of these pathways (Fig. 2C). These data are in line with previous studies and confirm that human MDMs are strongly activated upon EBOV infection (6,9,26–30). Functional enrichment analysis was performed on global transcriptomic data using Ingenuity Pathway Analysis (IPA), an interactive software package that uses proprietary algorithms to determine functional enrichment of large, complex omics data sets such as those produced by mRNA-Seq. IPA is based on a manually curated knowledge base that links gene expression to biological functions that have been experimentally demonstrated and described in peer-reviewed articles. Functional enrichment showed a strong activation of multiple cytokine signaling pathways after EBOV infection (Fig. 3). The same overall pattern was also detected after LPS treatment, again demonstrating a striking similarity in the EBOV- and LPS-mediated responses (Fig. 3). Many of these pathways contain genes known to be involved in potent inflammatory responses such as TLR4 signaling and proinflammatory cytokine production in response to LPS. Enrichment for these pathways was not found during RESTV infection (Fig. 3). Taken together, these data suggest that RESTV fails to efficiently activate human MDMs.
FIG 2.
EBOV infection and LPS stimulation induce similar transcriptional changes in MDMs that are absent in RESTV infection. (A) Number of highly DE genes following infection or LPS stimulation relative to time-matched mock-treated samples of MDMs generated from three separate donors in independent experiments. Differential gene expression cutoff was set to |log2FC| >1, and aP value of ≤0.05 was calculated using a generalized linear model with subsequent Benjamini-Hochberg correction. (B) Distribution of DE genes following infection with EBOV or RESTV or LPS treatment. A Venn diagram shows the overlap of DE genes at 6 hpi and 1 and 2 dpi. (C) Hierarchical clustering of the union of 3,574 DE genes based on Euclidean distances reveals distinct modules of gene expression in infected and LPS-stimulated MDMs. The heat map represents the average expression intensities for each condition and time point relative to the average of time-matched mock-infected samples. Each module is identified by a unique color, represented on the left of the heat map. Bar graphs represent canonical pathways associated with genes identified in each of the modules.
TABLE 1.
Functional enrichment analysis of DE genes following EBOV or RESTV infection or LPS stimulationa
Module | Ingenuity canonical pathway(s) | −log (P value) |
---|---|---|
Module 1 (588 transcripts, light blue) | Granulocyte adhesion and diapedesis | 1.87E + 01 |
Role of hypercytokinemia/hyperchemokinemia in the pathogenesis of influenza | 1.73E + 01 | |
Agranulocyte adhesion and diapedesis | 1.68E + 01 | |
Role of cytokines in mediating communication between immune cells | 1.65E + 01 | |
Communication between innate and adaptive immune cells | 1.54E + 01 | |
Differential regulation of cytokine production in IECs by IL-17A and IL-17F | 1.32E + 01 | |
Differential regulation of cytokine production in Mf and Th cells by IL-17A and IL-17F | 1.32E + 01 | |
Role of pattern recognition receptors in recognition of bacteria and viruses | 1.31E + 01 | |
Interferon signaling | 1.31E + 01 | |
Activation of IRF by cytosolic pattern recognition receptors | 1.28E + 01 | |
Module 2 (1,342 transcripts, dark blue) | Death receptor signaling | 1.20E + 01 |
Antigen presentation pathway | 1.14E + 01 | |
Type I diabetes mellitus signaling | 7.85E + 00 | |
Cross talk between dendritic cells and natural killer cells | 7.58E + 00 | |
Dendritic cell maturation | 7.51E + 00 | |
TREM1 signaling | 7.45E + 00 | |
NF-κB signaling | 6.70E + 00 | |
Protein ubiquitination pathway | 6.61E + 00 | |
GM-CSF signaling | 6.50E + 00 | |
Role of macrophages, fibroblasts, and endothelial cells in rheumatoid arthritis | 6.26E + 00 | |
Module 3 (938 transcripts, orange) | EIF2 signaling | 7.42E + 00 |
Cell cycle control of chromosomal replication | 7.24E + 00 | |
Breast cancer regulation by stathmin 1 | 7.08E + 00 | |
Cell cycle: G2/M DNA damage checkpoint regulation | 6.31E + 00 | |
Remodeling of epithelial adherens junctions | 6.26E + 00 | |
Role of CHK proteins in cell cycle checkpoint control | 5.74E + 00 | |
Mitotic roles of Polo-like kinase | 5.63E + 00 | |
Gap junction signaling | 5.23E + 00 | |
GDP-mannose biosynthesis | 4.40E + 00 | |
Role of BRCA1 in DNA damage response | 4.12E + 00 | |
Module 4 (805 transcripts, red) | CDP-diacylglycerol biosynthesis I | 3.87E + 00 |
Phosphatidylglycerol biosynthesis II (nonplastidic) | 3.60E + 00 | |
Gαi signaling | 3.15E + 00 | |
Mismatch repair in eukaryotes | 2.78E + 00 | |
Eicosanoid signaling | 2.26E + 00 | |
G-protein-coupled receptor signaling | 2.22E + 00 | |
cAMP-mediated signaling | 2.14E + 00 | |
Leukotriene biosynthesis | 1.98E + 00 | |
Antiproliferative role of TOB in T cell signaling | 1.98E + 00 | |
The visual cycle | 1.90E + 00 |
The 10 most significantly enriched IPA canonical pathways for each gene expression module are shown. Colors refer to colors inFig. 2C. Significance is considered a −log10P value greater than 1.3 as determined by the Fisher exact test.
FIG 3.
EBOV and LPS, but not RESTV, activate similar immune pathways in MDMs. Ingenuity Pathway Analysis (IPA) was used to assess enrichment of canonical pathways in the data sets obtained from each infectious/stimulation condition across time as described in the legend toFig. 2. TheP values are represented by the diameters of circles with more significant pathway enrichment as determined by the Fisher exact test correlating with bigger diameters. “Ratio” represents the number of genes found to be DE relative to the total number of genes that represent the pathway, with darker red color correlating with a greater number of pathway-associated DE genes. Pathway enrichment is reported as −log10P value, where values greater than 1.3 are considered significant. Gray X's represent a lack of enrichment for a canonical pathway.
RESTV infection is not associated with an IFN signature.
The EBOV VP35 and VP24 proteins counteract the host IFN response (17,18), yet the top upregulated genes in EBOV-infected MDMs at 1 dpi included type I and III IFN genes, as well as IFN-stimulated genes (ISGs) (Fig. 4A). The genes for C-X-C motif chemokine ligand 10 (CXCL10/IP10) and C-C motif chemokine ligand 5 (CCL5/RANTES), both regulated by IFN regulatory factor 3 (IRF3), were also strongly upregulated in EBOV-infected cells at 1 dpi (Fig. 4A). Similar to EBOV infection, LPS treatment led to upregulation of ISGs as well as CXCL10 and CCL5. However, we were not able to detect type I or type III IFN gene expression at any time point investigated for LPS-treated samples (Fig. 4A). In agreement with an overall lack of changes in gene expression (Fig. 2), RESTV infection led to minimal changes in the expression levels of IFN genes and ISGs (Fig. 4A).
FIG 4.
Infection with EBOV, but not RESTV, induces a type I and type III IFN response in MDMs. (A) MDMs were either infected with EBOV or RESTV, stimulated with LPS, or left untreated, and cellular RNA was isolated at the indicated time points. The heat map represents the average expression intensities for type I and type III IFN genes and selected ISGs in MDMs from three separate donors in independent experiments. Each column represents an experimental condition and time point, and each row represents an individual gene. Upregulated genes are represented in red, and downregulated genes are represented in blue. (B) Luminex analysis of supernatants from MDMs infected with EBOV or RESTV. As controls, cells were stimulated with LPS or left untreated (Mock). Time points analyzed by RNA-Seq (6 h, 1 day [1d], or 2d as for panel A) were examined using samples from four separate donors, including the corresponding supernatants from the three donors used to generate the RNA-Seq data for all conditions. Extended analysis of late time points postinfection (3d and 4d) were performed with samples from two donors (in gray). Each donor is represented by a different symbol. Horizontal bars represent mean values. Statistically significant differences from time-matched Mock samples as determined by one-way ANOVA are indicated by asterisks (***,P ≤ 0.0001; **,P ≤ 0.001; *,P ≤ 0.05). 3d and 4d time points were not included in the statistical analysis.
Secretion of selected corresponding proteins was confirmed by Luminex analysis of the supernatants of infected cells. IFN-α secretion was exclusively observed in supernatants from EBOV-infected MDMs, whereas both LPS treatment and EBOV infection led to significant secretion of CXCL10 (Fig. 4B). Consistent with the RNA-Seq data, neither IFN-α nor CXCL10 was detected in the supernatants of RESTV-infected MDMs (Fig. 4B). Since it is conceivable that RESTV infection induces a delayed response at late stages of infection, we included additional time points in the Luminex analysis for two donors. In contrast to the case with EBOV, no IFN-α or CXCL10 secretion was detected in RESTV-infected MDMs up to 4 dpi (Fig. 4B, gray symbols). The lack of expression of these genes in RESTV-infected cells up to 4 dpi was confirmed by qRT-PCR analysis (data not shown). Taken together, these data show that RESTV infection of MDMs does not lead to a delayed host cell response at late stages of infection.
Expression of type I IFNs, CXCL10, and CCL5 requires the activation and nuclear translocation of the transcription factor IRF3 (reviewed in reference36). To examine whether IRF3 was activated and translocated into the nuclei of infected MDMs, we performed coimmunofluorescence analysis using antibodies directed against IRF3 and the nucleoproteins (NPs) of RESTV or EBOV. The nucleoproteins localize in large viral inclusions within the cytoplasm of the infected cells (37). In line with previous observations, viral inclusions were detected only at later time points postinfection, when viral protein expression was high (Fig. 5A) (37,38). IRF3 remained in the cytoplasm of RESTV-infected cells at all time points analyzed, whereas nuclear translocation of IRF3 was detected in a significant number of EBOV-infected cells as early as 30 min postinfection (Fig. 5). These data suggest that early events that trigger the activation of IRF3 and subsequent induction of IFN genes in EBOV-infected MDMs are absent in RESTV infection.
FIG 5.
RESTV infection does not induce nuclear IRF3 translocation. (A) MDMs grown in chamber slides were infected with EBOV or RESTV, fixed at the indicated times postinfection, and examined by immunofluorescence analysis using antibodies directed against EBOV NP or RESTV NP (red) and IRF3 (green). Cell nuclei were stained with DAPI (blue). The experiment was performed twice with cells obtained from two separate donors; representative images are shown. A representative image of mock-infected cells for one time point and one antibody combination is shown. (B) The percentage of cells (+SEM) showing nuclear IRF3 localization was determined by counting at least 100 infected MDMs from two different donors used in two independent experiments. Gray bar, mock infected; black bars, EBOV infected; white bars, RESTV infected. Statistically significant differences compared to Mock 30 min as determined by one-way ANOVA are indicated by asterisks (***,P ≤ 0.0001).
EBOV, but not RESTV, profoundly activates NF-κB signaling pathways.
A key regulator of cytokine and chemokine expression is nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) (reviewed in reference39). We observed profound upregulation of genes involved in NF-κB signaling in EBOV-infected and LPS-treated MDMs, whereas this group of genes was only moderately affected by RESTV infection (Fig. 6A). Consistent with previous reports (6,9,26–30), robust expression of proinflammatory mediators was observed in EBOV-infected MDMs, including interleukin-6 (IL-6), tumor necrosis factor alpha (TNF-α), CXCL8 (IL-8), CCL3 (MIP1α), and CCL4 (MIP1β) (Fig. 6B). In contrast, RESTV-infected MDMs remained remarkably silent. The anti-inflammatory Epstein-Barr virus-induced gene 3 (EBI3), also known as the gene for interleukin-27 subunit β (IL-27β), was the only DE cytokine gene with a >2-fold upregulation in RESTV-infected cells. A few chemokine genes were found to be upregulated, including the genes for CCL20 (MIP3α), CCL19 (MIP3β), CCL13, CXCL6, and CXCL8 (Fig. 6B). The differential expression of these genes was not unique for RESTV infection; all of them were also DE in EBOV-infected MDMs (Fig. 6B). Despite robust upregulation at the mRNA level, only moderate amounts of the proinflammatory cytokines IL-6 and TNF-α were secreted from EBOV-infected cells from some donors, compared to high levels after LPS stimulation as shown by Luminex analysis (Fig. 6C). Due to high donor variability, the observed changes in EBOV-infected cells were not statistically significant. Neither IL-6 nor TNF-α was detected in the supernatants of RESTV-infected cells at any time point analyzed. In line with the RNA data, the chemokine CXCL8 was secreted from both EBOV- and RESTV-infected MDMs (Fig. 6C). The biological relevance of CXCL8 secretion in RESTV infection remains to be determined.
FIG 6.
Differences in NF-κB activation in EBOV- and RESTV-infected MDMs. (A) MDMs were either infected with EBOV or RESTV, stimulated with LPS, or left untreated, and cellular mRNA was isolated at the indicated time points and used for RNA-Seq analysis. The heat map represents the average expression intensities for 137 genes either involved in NF-κB signal transduction or being NF-κB target genes, as identified by the GeneCards database, relative to the time-matched mock-treated samples of MDMs generated from three separate donors in independent experiments. Each column represents an experimental condition and time point, and each row represents an individual gene. Upregulated genes are represented in red and downregulated genes are represented in blue. (B) Average gene expression intensity for proinflammatory cytokine genes, relative to time-matched mock-treated samples. (C) Luminex analysis of supernatants from MDMs infected with EBOV or RESTV. As controls, cells were stimulated with LPS or left untreated (Mock). Time points analyzed by RNA-Seq (6 h, 1d, and 2d as in panel A) were examined using samples from four separate donors, including the corresponding supernatants from the three donors used to generate the RNA-Seq data for all conditions. Later time points (3 and 4 dpi) were examined using samples from 2 individual donors (shown in gray). Each donor is represented by a different symbol. Horizontal bars represent mean values. Statistically significant differences to time-matched Mock samples were determined by one-way ANOVA (***,P ≤ 0.0001; **,P ≤ 0.001; *,P ≤ 0.05). 3d and 4d time points were not included in the statistical analysis.
An early event of canonical NF-κB activation is the nuclear translocation of the NF-κB subunit p65, which we examined by coimmunofluorescence analysis of infected or LPS-treated cells. In contrast to EBOV-infected or LPS-treated cells, nuclear accumulation of p65 was rarely observed in RESTV-infected cells (Fig. 7). At all time points analyzed, a few cells showed nuclear p65 staining in RESTV-infected samples, but this was also the case in noninfected control cells. This supports the RNA-Seq and Luminex results, which indicate that RESTV infection does not elicit a robust proinflammatory response in human MDMs. Similar to the case with IRF3, we observed nuclear translocation of p65 in EBOV-infected MDMs as early as 30 min postinfection but not at late time points (Fig. 7). These data were supported by Western blotting showing degradation of NF-κB inhibitor alpha (NFKBIA, IκBα) in EBOV-infected MDMs starting at 30 min postinfection, with a peak at 1 hpi, but not at later time points (Fig. 8A). As a control, cells were treated with LPS or TNF-α, which led to IκBα degradation as early as 15 min after treatment. IκBα prevents the nuclear translocation of NF-κB proteins, and its degradation is a crucial step in the NF-κB pathway. Controlled activation of NF-κB is achieved by a negative-feedback loop restoring IκBα levels after initial activation. The reappearance of IκBα at later time points suggests that NF-κB is not constitutively activated in EBOV-infected MDMs, which is in line with the p65 nuclear translocation data shown inFig. 7. We next performed DNA binding assays to examine if the various NF-κB subunits were able to bind to their DNA target sequences in EBOV-infected cells. MDMs were infected with EBOV, left untreated, or stimulated with LPS, and nuclear lysates were prepared at various time points. DNA binding of NF-κB subunit p65 was detected in EBOV-infected cells at 1 hpi (Fig. 8B). A similar pattern was observed for p50, although less pronounced (Fig. 8B). DNA binding of p52, c-Rel, or RelB subunits was not detected (data not shown). Together, our data show that EBOV infection of MDMs leads to early activation of canonical NF-κB signaling, whereas noncanonical signaling pathways are not activated. Compared to the case with LPS-treated samples, NF-κB activation seems to be moderate in EBOV-infected cells and only marginal in RESTV-infected MDMs.
FIG 7.
EBOV, but not RESTV, infection induces p65 nuclear translocation in MDMs. (A) MDMs grown in chamber slides were infected with EBOV or RESTV, treated with LPS, or left noninfected (Mock). At the indicated time points, cells were examined by immunofluorescence analysis using antibodies targeting EBOV NP or RESTV NP (red) and p65 (green). Cell nuclei were stained with DAPI (blue). A representative image of mock-infected cells for one time point and one antibody combination is shown. The experiment was performed four times with cells from four separate donors; representative images are shown. (B) The percentage of cells (+SEM) showing nuclear p65 localization was determined by counting at least 100 infected MDMs from four different donors used in four independent experiments. Light gray bars, mock infected; dark gray bars, LPS treated; black bars, EBOV infected; white bars, RESTV infected. Statistically significant differences from Mock 30 min as determined by one-way ANOVA are indicated (***,P ≤ 0.0001).
FIG 8.
EBOV activates canonical NF-κB signaling in MDMs. (A) MDMs were infected with EBOV, stimulated with LPS or TNF-α, or left untreated (Mock), and whole-cell lysates were prepared at the indicated time points. Western blot analysis was performed using antibodies directed against β-actin and IκBα. The experiment was performed in triplicate using MDMs from three different donors. A representative result is shown. (B) MDMs were infected with EBOV, stimulated with LPS, or left untreated (Mock). Nuclear lysates were prepared at the indicated time points and analyzed using an NF-κB subunit DNA binding assay. Raji cell extract was used as a positive control for DNA binding. Light gray bars, mock infected; dark gray bars, LPS treated; black bars, EBOV infected; checkered bars, Raji control. The experiment was performed in triplicate using MDMs from three different donors. Bars represent mean values (+SEM). Statistically significant differences to corresponding time-matched Mock samples as determined by one-way ANOVA are indicated (*,P ≤ 0.05).
Neither RESTV nor EBOV is able to inhibit NF-κB activation.
The lack of an inflammatory response in RESTV-infected MDMs could be due either to the lack of activating stimuli or to the inhibitory function of viral proteins that block antiviral signaling pathways, such as VP35 and VP24. To discriminate between these two possibilities, we analyzed LPS-mediated NF-κB activation at later stages of infection, when the viral proteins are expressed at high levels. MDMs were infected with RESTV or EBOV and at 1 dpi treated with LPS. LPS stimulation after RESTV infection led to significant changes in gene expression compared to RESTV-infected, untreated cells (Fig. 9A, compare RESTV versus Mock to RESTV+LPS versus RESTV) or noninfected, untreated cells (Fig. 9A, compare RESTV versus Mock to RESTV+LPS versus Mock). The gene expression profile was similar to the response to LPS alone, representing a proinflammatory signature including activation of TLR signaling and cytokine-regulated pathways (Fig. 9A, red and orange modules;Table 2). In contrast, LPS treatment after EBOV infection of MDMs resulted in minor changes in the gene expression pattern (Fig. 9A, compare EBOV versus Mock to EBOV+LPS versus EBOV) as EBOV infection alone already induced a proinflammatory response similar to LPS (Fig. 9A, EBOV versus Mock and LPS versus Mock).
FIG 9.
Neither EBOV nor RESTV interferes with LPS-mediated NF-κB activation. (A) LPS treatment induces significant gene expression in RESTV-infected cells. MDMs generated from three different donors were infected with EBOV or RESTV or left uninfected and at 1 dpi treated with LPS for 6 h. Cellular mRNA was isolated and subjected to RNA-Seq analysis. The heat map represents the average expression intensities of the union of 4311 DE genes for each of the specified treatment conditions (left, EBOV or RESTV infection or LPS treatment compared to noninfected, untreated cells [Mock]; middle: EBOV or RESTV infection + LPS stimulation compared to infected cells only; right, EBOV or RESTV infection + LPS compared to Mock. Each column represents an experimental condition, and each row represents individual genes. (B) MDMs grown in chamber slides were infected with EBOV or RESTV or left uninfected and at 1 dpi were stimulated with LPS for 30 min where indicated. Immunofluorescence analysis was performed using antibodies directed against EBOV NP or RESTV NP (red) and p65 (green). Cell nuclei were stained with DAPI (blue). The experiment was performed three times with cells from three separate donors. Representative images are shown. EBOV NP staining is shown for Mock samples; no difference was observed for RESTV NP staining. (C) The percentage of cells (+SEM) showing nuclear p65 localization was determined by counting at least 100 infected MDMs from 3 different donors. Statistical analysis was performed using one-way ANOVA (***,P ≤ 0.0001). (D) MDMs were infected with EBOV or RESTV and at 1 dpi were stimulated with LPS for 6 h or 1 day. Cell supernatants were analyzed by Luminex assay. As controls, cells were infected with EBOV or RESTV for the indicated times without subsequent LPS stimulation, stimulated with LPS, or left untreated (Mock) for the indicated times. No statistically significant differences were observed between LPS-treated cells and cells infected with EBOV or RESTV prior to LPS treatment as determined with one-way ANOVA (P > 0.05). The experiment was performed three times using MDMs from 3 individual donors; each donor is represented by a different symbol. Horizontal bars represent mean values.
TABLE 2.
Functional enrichment analysis of DE genes following LPS stimulation of EBOV- or RESTV-infected cellsa
Module | Ingenuity canonical pathway(s) | −log (P value) |
---|---|---|
1,765 transcripts, red | Death receptor signaling | 1.58E + 01 |
Type I diabetes mellitus signaling | 1.08E + 01 | |
Induction of apoptosis by HIV1 | 1.07E + 01 | |
TWEAK signaling | 1.03E + 01 | |
Toll-like receptor signaling | 1.01E + 01 | |
Role of pattern recognition receptors in recognition of bacteria and viruses | 1.00E + 01 | |
TNFR1 signaling | 9.95E + 00 | |
Unfolded protein response | 9.94E + 00 | |
Role of PKR in interferon induction and antiviral response | 9.87E + 00 | |
Interferon signaling | 9.76E + 00 | |
Granulocyte adhesion and diapedesis | 2.22E + 01 | |
285 transcripts, orange | Agranulocyte adhesion and diapedesis | 2.02E + 01 |
Role of hypercytokinemia/hyperchemokinemia in the pathogenesis of influenza | 1.94E + 01 | |
Role of cytokines in mediating communication between immune cells | 1.91E + 01 | |
Differential regulation of cytokine production in intestinal epithelial cells by IL-17A and IL-17F | 1.69E + 01 | |
Differential regulation of cytokine production in macrophages and T helper cells by IL-17A and IL-17F | 1.66E + 01 | |
Communication between innate and adaptive immune cells | 1.51E + 01 | |
Cross talk between dendritic cells and natural killer cells | 1.01E + 01 | |
Activation of IRF by cytosolic pattern recognition receptors | 9.60E + 00 | |
Pathogenesis of multiple sclerosis | 9.55E + 00 | |
Mismatch repair in eukaryotes | 7.79E + 00 | |
2261 transcripts, blue | Role of CHK proteins in cell cycle checkpoint control | 7.16E + 00 |
Role of BRCA1 in DNA damage response | 6.86E + 00 | |
Cell cycle control of chromosomal replication | 5.61E + 00 | |
ATM signaling | 4.62E + 00 | |
Estrogen-mediated S-phase entry | 4.46E + 00 | |
Cell cycle: G1/S checkpoint regulation | 4.13E + 00 | |
Antiproliferative role of TOB in T cell signaling | 4.11E + 00 | |
Mitotic roles of Polo-like kinase | 3.95E + 00 | |
DNA double-strand break repair by homologous recombination | 3.87E + 00 |
The 10 most significantly enriched IPA canonical pathways for each gene expression module are shown. Colors refer to colors inFig. 9A. Significance is considered a −log10P value greater than 1.3 as determined by the Fisher exact test.
As shown inFig. 7, only a minor fraction of EBOV- and RESTV-infected cells showed nuclear accumulation of p65 at 1 dpi. However, when the infected cells were treated with LPS at 1 dpi, p65 shuttled into the nucleus in almost all cells, indicating that neither RESTV nor EBOV infection interfered with LPS-induced NF-κB activation (Fig. 9B andC). This was confirmed by Luminex analysis, showing that LPS treatment of RESTV-infected cells elicited a robust cytokine and chemokine secretion (Fig. 9D). These data suggest that the lack of cytokine and chemokine induction in RESTV-infected MDMs cannot be attributed to the inhibition of immune-modulatory signaling pathways. Rather, RESTV fails to activate these pathways in human MDMs.
RESTV GP does not activate TLR4-mediated responses.
Previous studies showed that the EBOV surface protein GP is sufficient to induce a proinflammatory response via TLR4 activation (30,32,33). Since our data indicate that RESTV infection does not lead to the activation of MDMs, we next investigated if RESTV GP lacks the ability to induce a proinflammatory response. We first tested gamma-irradiated EBOV and RESTV particles for their ability to induce p65 translocation in MDMs in the absence of viral replication. p65 was translocated into the nuclei of MDMs stimulated with gamma-irradiated EBOV particles but not in cells treated with inactivated RESTV particles (Fig. 10A andB).
FIG 10.
EBOV GP, but not RESTV GP, induces TLR4-mediated p65 nuclear translocation. (A, D, and E) MDMs grown on coverslips were treated as indicated and examined for p65 nuclear translocation by immunofluorescence analysis using a p65 antibody (green). Cell nuclei were stained with DAPI (blue). Experiments were performed three times using MDMs from three separate donors. Shown are representative images. (A) MDMs were treated with gamma-irradiated EBOV or RESTV or were mock treated for 1 h. (B) The percentage of cells (+SEM) showing nuclear p65 localization was determined by counting at least 100 cells from three different donors. Statistically significant differences from mock-treated samples as determined by one-way ANOVA are indicated (***,P ≤ 0.0001). (C) Western blot analysis of eVLPs and rVLPs using antibodies against EBOV GP1, RESTV GP1, and EBOV VP40. The EBOV VP40 antibody cross-reacts with RESTV VP40 at a lower affinity and was used to visualize both EBOV and RESTV VP40 proteins. Protein bands were visualized using the Li-Cor Odyssey system. E, EBOV proteins; R, RESTV proteins. (D) MDMs were treated with eVLPs or rVLPs for 1 h. Top row, GP-containing VLPs; middle row, GP-free VLPs; bottom row, VLPs containing the heterologous GP. (E) MDMs were treated for 3 h with LPS-RS or left untreated before stimulation with LPS or VLPs for 1 h. (F) The percentage of cells (+SEM) showing nuclear p65 localization was determined by counting at least 100 cells from three different donors. Light gray bars, untreated; dark gray bars, LPS treated; black bars, treated with VLPs containing EBOV GP; hatched bars, treated with VLPs without GP; white bars, treated with VLPs containing RESTV GP. Statistically significant differences of LPS-RS-treated samples from corresponding untreated samples as determined by one-way ANOVA are indicated (***,P ≤ 0.0001).
As mentioned above, we observed slight differences in EBOV and RESTV RNA levels at 6 hpi (Fig. 1C andD) which might be due to differences in the viral replication kinetics as reported previously (13,40,41). In addition, it has been shown recently that the particle-to-PFU ratio of EBOV stocks influenced disease outcome in an animal model (42). In order to exclude the possibility that the observed differences in the host response of MDMs to infection with EBOV or RESTV are due to differences in the replication kinetics of the two viruses or to differences in the particle-to-PFU ratio of EBOV and RESTV stocks, we next employed Ebola and Reston VLPs to determine potential differences in the ability of RESTV and EBOV GP to induce an inflammatory response in MDMs. VLPs were produced in 293T cells transfected with a mixture of expression plasmids encoding EBOV or RESTV NP, matrix protein VP40, and GP. To produce GP-free VLPs, the GP expression plasmids were omitted. The presence of EBOV and RESTV GP in VLP preparations was confirmed by Western blotting (Fig. 10C). When MDMs were stimulated with VLPs, p65 translocation was observed only in cells treated with Ebola virus VLPs (eVLPs) containing EBOV GP and not in cells treated with eVLPs lacking GP or in cells treated with Reston virus VLPs (rVLPs) with or without RESTV GP (Fig. 10D andF, bars 5, 7, 9, and 10). To confirm that the observed differences were attributed to GP and not to other VLP components, MDMs were stimulated with chimeric VLPs (eVLPs containing RESTV GP and vice versa). Nuclear p65 shuttling was observed with rVLPs containing EBOV GP but not with eVLPs containing RESTV GP, confirming that RESTV GP is not able to induce a proinflammatory response in MDMs (Fig. 10D andF, bars 8 and 11).
To examine if EBOV GP-mediated NF-κB activation could be blocked by inhibiting the TLR4 signaling pathway, MDMs were pretreated with LPS-RS, a competitive LPS antagonist fromRhodobacter sphaeroides which binds to TLR4 without inducing signaling (43,44). LPS-RS pretreatment abolished nuclear p65 translocation mediated by VLPs containing EBOV GP, indicating that the EBOV GP-triggered activation of NF-κB in MDMs is, indeed, mediated by TLR4 signaling and can be blocked by TLR4 antagonists (Fig. 10E andF, bars 5, 6, 11, and 12). Together, our data suggest that the observed differences in the proinflammatory response to EBOV and RESTV infection can be attributed to differences in GP-mediated TLR4 activation in MDMs.
DISCUSSION
In this study, we showed that infection of human MDMs with RESTV is not associated with a strong immune response, in contrast to activation of NF-κB- and IRF3-mediated cytokine and chemokine responses triggered by TLR4 activation in MDMs infected with the highly pathogenic EBOV. This suggests that higher virulence and pathogenicity might be correlated with a greater activation of immune responses in this cell type. A previous study analyzing the cytokine response to filovirus infection in human monocytes and MDMs reported no significant differences between RESTV- and EBOV-infected cells (27). We previously showed increased expression of antiviral genes, including IFN response genes, in RESTV-infected human hepatocarcinoma (Huh7) cells compared to EBOV-infected cells (41). Besides cell-specific differences, various infection conditions might account for the observed discrepancies. It has been shown previously that gene expression patterns can be obscured by using unpurified EBOV virus stocks for infection presumably due to the presence of soluble factors in the inoculum (45). Compared to unpurified inoculum, EBOV infections with purified virus yielded strikingly different results in the gene expression profiles of the infected cells (45). While virus-containing cell supernatants cleared from cell debris by low-speed centrifugation were used as inocula in the previous RESTV studies (27,41), all infections in the present study were performed with sucrose cushion-purified, endotoxin-free virus stocks for both EBOV and RESTV.
In our study, the EBOV and RESTV infection rates in MDMs were similar at 1 dpi, as shown by immunofluorescence analysis and quantification of viral mRNA (Fig. 1). However, at early time points of infection (6 hpi), the amount of RESTV mRNA was slightly smaller than the amount of EBOV mRNA in infected cells. This could be attributed either to differences in the transcription and replication kinetics, as reported previously (13,40,41), or to differences in the amount of input virus. When MDMs were infected with EBOV at an MOI of 0.1, they displayed a cytokine and IFN response similar to but weaker than seen in cells infected with EBOV at a high MOI (data not shown). In contrast, MDMs infected with RESTV at an MOI of 5 to 7 remained silent, suggesting that potential differences in the infection rates do not account for the observed differences in the host response. Even at late times postinfection (3 and 4 dpi), and despite robust viral replication, RESTV-infected MDMs did not show the proinflammatory signature characteristic of EBOV infection. Further, our data with inactivated viral particles and VLPs clearly indicate that the observed differences between EBOV and RESTV can be attributed to differences in the ability of EBOV and RESTV GP to activate TLR4.
The surface glycoproteins of various viruses, including members of the orderMononegavirales, have been shown to induce a TLR4-mediated inflammatory response (30,32,33,46–50). Recognition of bacterial LPS, the best-studied TLR4 ligand, is a complex process and requires the interaction of four proteins, including CD14 and MD-2 (51). Both have been shown to be required for TLR4 activation mediated by the fusion protein F of respiratory syncytial virus (RSV), potentially mediated by interaction between the hydrophobic fusion peptide of RSV F and MD-2 (49,50). Similar to our results, TLR4 activation by RSV F protein was inhibited by LPS-RS, which competitively blocks MD-2 binding of the lipid A region of LPS (52,53). Future studies will show if hydrophobic regions within EBOV GP interact with components of the TLR4 signaling complex to induce inflammatory responses.
EBOV GP-mediated cytokine induction in monocyte-derived dendritic cells is triggered not only by TLR4 but also by C-type lectin domain family 4 member G (CLEC4G/LSECtin) (34). Expression of LSECtin on MDMs depends on IL-4 stimulation (54), and unstimulated MDMs, as used in the presented study, do not express LSECtin (54). This is consistent with our data which suggest that TLR4 activation is mainly responsible for the observed inflammatory response in naive MDMs, as inhibition of TLR4 prevented GP-mediated NF-κB activation.
Dengue virus (DENV) nonstructural protein 1 (NS1) has also been identified as a viral TLR4 agonist. NS1 not only triggers a TLR4-dependent proinflammatory response but also induces vascular leakage (55). Intriguingly, mice treated with LPS-RS prior to infection with DENV showed significantly less capillary leakage (55). Our data show that the EBOV-triggered proinflammatory response was also antagonized by LPS-RS, opening up new treatment options for EBOV disease by blocking TLR4 activation during infection. Indeed, a recent study provided evidence that controlling the excessive inflammatory response induced by EBOV infection could be a viable strategy to treat EBOV disease (56).
LPS-mediated TLR4 signaling activates NF-κB via the adaptor protein corresponding to myeloid differentiation primary response gene 88 (MyD88), leading to cytokine and chemokine production. Endocytosis of TLR4 is not required for MyD88-dependent signaling (51,57,58). In contrast, activation of IRF3-dependent genes, including those for type I and III IFNs, CCL5, and CXCL10, through Toll-like receptor adaptor molecule 2 (TICAM2/TRAM) and Toll-like receptor adaptor molecule 1 (TICAM1/TRIF) requires endocytosis of the TLR4 complex (59–61). We observed both p65 and IRF3 nuclear translocation within 30 min of EBOV infection, suggesting that EBOV triggers both MyD88- and TRIF/TRAM-dependent signaling pathways early in the viral replication cycle. Activation of these two signaling pathways was also observed in murine dendritic cells after stimulation with EBOV-like particles (62). A recent EBOV entry study showed that the viral particles are quickly internalized but are only released into the cytoplasm after a 30-min lag period, during which they traffic to the endolysosome, where fusion occurs (63,64). The prolonged presence of EBOV in the endosome might allow for efficient activation of endocytosed TLR4 complexes, leading to activation of IRF3. We did not observe type I or type III IFN induction in LPS-treated cells, although CXCL10, CCL5, and ISGs were upregulated. This is in line with previous reports showing that although IRF3 is activated in LPS-treated human MDMs, LPS stimulation alone is not sufficient for full activation of the type I IFN response (65,66). Proinflammatory cytokines, including IL-6 and TNF-α, were secreted at much higher levels in LPS-treated MDMs than in EBOV-infected cells in our study. This suggests that MyD88-dependent pathways are preferentially activated in LPS-treated cells, while TRIF/TRAM-mediated signaling is activated to a lesser extent. Our data indicate that the balance between MyD88- and TRIF/TRAM-mediated signaling might be different in EBOV infection than in LPS treatment. The upregulation of genes involved in IFN signaling might play a crucial role in EBOV pathogenesis. Thus, a strong IFN signature was found to be associated with fatal outcome in the recent EBOV outbreak in West Africa (67).
Surprisingly, there was no upregulation of IFN or ISGs in RESTV-infected human MDMs. This suggests that IFN induction, if at all, only moderately contributes to the restriction of RESTV infection in primary cells. Our data are consistent with reports showing that mice lacking a functional IFN system did not become severely ill when infected with RESTV (13,14,16,68). However, similar to EBOV, RESTV is equipped with functional IFN antagonists, so one might assume that antiviral IFN responses are involved in controlling RESTV infection. We have previously shown that RESTV is less capable than EBOV of counteracting IFN responses in Huh7 cells (41), suggesting a complex correlation between IFN antagonism and virus control in specific cell types. Along those lines, studies on the ebolavirus IFN antagonists VP35 and VP24 provide evidence that RESTV VP35 and VP24 might be less efficient in counteracting the IFN response (19,23,24,69–71). It is likely that multiple factors contribute to the reduced pathogenicity of RESTV, and our data indicate that the lack of TLR4 activation might be one of them. A dysregulated induction of a TLR4-mediated proinflammatory response can lead to severe tissue and organ damage, which is a hallmark of EBOV infection (72). Thus, the lack of TLR4 activation in RESTV infection might be highly beneficial for the host because it minimizes inflammation and cell damage, as observed in an IFNAR knockout mouse model using recombinant EBOV expressing RESTV GP (13).
Activation of TLR4 induces a massive upregulation of cellular genes, which may also lead to the production of proviral factors in infected cells. Indeed, it has been recently reported that TLR4-mediated upregulation of SOCS3 led to enhanced EBOV budding (33). Intriguingly, SOCS3 was one of the few genes upregulated in both EBOV- and RESTV-infected MDMs (Fig. 6B). It remains to be determined whether RESTV GP binds to TLR4 and triggers a moderate host response, including the upregulation of genes such as SOCS3, or if RESTV GP fails to interact with TLR4 and expression of these genes is mediated by another signaling pathway. Interestingly, a previous study on the interaction of EBOV GP with TLR4 showed that GP lacking the mucin-like domain still binds to TLR4 but does not induce TLR4 signaling (32).
Taken together, our results show remarkable differences in the host response to EBOV and RESTV infection in primary human MDMs. Future studies focusing on the differences in EBOV- and RESTV-mediated immune activation in different cell types and tissues and across different species will help to define the correlates of protection against RESTV infection and deepen our understanding of filovirus pathogenesis.
MATERIALS AND METHODS
Cells and viruses.
VeroE6 (ATCC CRL-1586) and 293T cells (ATCC CRL-3216) were obtained from the American Type Culture collection (ATCC). Cells were cultured in Dulbecco's modified Eagle medium (DMEM) supplemented with 10% fetal calf serum (FCS), penicillin (50 U/ml), and streptomycin (50 mg/ml). EBOV (Kikwit isolate, GenBank accession numberAY354458.1) and RESTV (Pennsylvania isolate, GenBank accession numberAF522874.1) stocks were grown in VeroE6 cells and purified using ultracentrifugation through a 20% sucrose cushion. Virus titers were determined in VeroE6 cells by 50% tissue culture infectious dose (TCID50) assay for EBOV and focus-forming assay for RESTV. Gamma irradiation of EBOV and RESTV virus stocks was performed with a 10-megarad dose on dry ice by following standard operating procedures (SOPs) approved by the local Institutional Biosafety Committee (IBC). All work with EBOV and RESTV was performed under biosafety level 4 (BSL4) conditions at the Integrated Research Facility at the Rocky Mountain Laboratories, Division of Intramural Research, NIAID, NIH, Hamilton, MT, by following IBC-approved SOPs.
Generation of MDMs.
MDMs were generated from leukopaks (NY Biologics Inc.) or apheresed peripheral blood mononuclear cells (PBMCs; NIH Clinics Center, Department of Transfusion Medicine) using Ficoll separation (GE Healthcare) and isolation of CD14+ monocytes by magnetic bead selection (Milteny Biotech). A total of 1.5 × 106 or 3 × 105 CD14-selected monocytes were seeded into 6-well or 24-well culture plates (with or without coverslips) or at 2 × 105 cells per well of a chamber slide (μ-Slide [8 well; ibidi] or Lab-Tek II chamber slide system [8 well; Nunc]) in RPMI medium without serum and allowed to adhere. Nonattached cells were removed, and RPMI medium supplemented with penicillin (50 U/ml), streptomycin (50 mg/ml), HEPES (10 mM), 5% FCS, and 5% human AB serum (Atlanta Biologicals) was added. Monocytes were incubated for 6 to 8 days to ensure differentiation into MDMs. MDMs were CD14+ and CD11b+ as confirmed by flow cytometry (data not shown). To ensure similar experimental conditions, cells from a single donor were used in a single experiment. To account for donor variations, experiments were repeated multiple times using cells obtained from different donors as noted in the figure legends. Throughout the figures, each donor is represented by a donor-specific symbol.
Generation of VLPs.
For generation of GP-containing VLPs, 3 × 106 293T cells seeded in T75 cell culture flasks were transfected with 3 μg of each expression plasmid encoding EBOV or RESTV VP40, nucleoprotein (NP), and GP using the CalPhos mammalian transfection kit (Clontech) according to the manufacturer's protocol. NP was included because it has been demonstrated to increase VLP release (73,74). The GP plasmids were replaced with 3 μg of green fluorescent protein (GFP) expression plasmid to produce VLPs lacking GP. Two days posttransfection, cell supernatants containing the VLPs were harvested and purified by ultracentrifugation through a 20% sucrose cushion. VLP protein concentration was determined using the quick-start Bradford protein assay (Bio-Rad).
Endotoxin test.
Purified, gamma-irradiated virus stocks and VLP preparations were tested for the presence of endotoxins using the Pyrogent Plus gel clotLimulus amoebocyte lysate (LAL) assay (Lonza) according to the manufacturer's protocol. Samples were found to be below the detection limit, while positive controls demonstrated that the tests were valid.
Infection and treatment of cells with viral particles, VLPs, LPS, or TNF-α.
MDMs from the same donors were infected with EBOV or RESTV at an MOI of 5 to 7. Cells were incubated at 37°C until the time of sample preparation. Equal amounts of viral particles were used for the treatment with gamma-irradiated virus. As controls, cells were left untreated or were treated with 100 ng/ml of LPS (ultrapure LPS; InvivoGen) or 20 ng/ml of TNF-α (Miltenyi Biotech) for the desired times. For VLP experiments, MDMs were treated with 1 μg/ml of VLPs for 1 h at 37°C and fixed. For LPS-RS experiments, MDMs were pretreated with 10 μg/ml of LPS-RS (ultrapure; InvivoGen) for 3 h at 37°C. Following addition of 1 ng/ml of LPS or 1 μg/ml of VLPs directly to the medium, cells were incubated for 1 h at 37°C and fixed.
RNA sample preparation.
Total RNA from 1.5 × 106 (6-well format) or 3 × 105 (24-well format) MDMs was isolated at the desired time points using TRIzol reagent (Invitrogen) according to the manufacturer's protocol and IBC-approved SOPs. RNA concentration was determined using a NanoDrop 1000 spectrophotometer (Thermo Scientific). For VP35 mRNA quantification by qRT-PCR, mRNA was purified from total RNA using the Poly(A)Purist kit (Ambion) according to the manufacturer's protocol.
Library preparation, RNA sequencing, and processing.
Libraries for mRNA sequencing were constructed using the Kapa stranded mRNA-Seq kit (Kapa Biosystems) using RNA samples from 3 separate donors. Libraries were quality controlled and quantified using the BioAnalyzer 2100 (Agilent Technologies, Inc.) and QuBit (Invitrogen) systems. Libraries were clonally amplified and sequenced on an Illumina NextSeq 500 to achieve a target density of approximately 200,000 to 220,000 clusters/mm2 on the flow cell with dual indexed paired-end sequencing at a 76-bp length using NextSeq 500 NCS v1.3 software. Raw reads (76 bp) had their adaptor sequences removed. General quality control of the raw reads was performed using FastQC. rRNA reads were removed via mapping by Bowtie (v2.1.0) using an index of human, mouse, and rat rRNA sequences (75). On average ∼10% of the reads from each sample were identified as mapping to rRNA and were removed from downstream analysis. Reads were also aligned to the genomes of the EBOV (Kikwit isolate, GenBank accession numberAY354458.1) and RESTV (Pennsylvania isolate, GenBank accession numberAF522874.1) using Bowtie2. Additionally, reads mapping to VP35 regions in either genome were counted using HTseq-count. Following removal of viral reads, the remaining reads were then mapped against a human reference genome (hg19, build GRCh37, from the UCSC genome browser using STAR [v2.4.0h1]) (76). Quantitative gene counts were produced from this alignment using the Python package HT-Seq utilizing the human annotation associated with the genome (77).
Differential expression analysis.
Normalization and differential expression analysis were carried out using R (v3.1.3) and the software package edgeR (v3.10.2) (78). Prior to differential analysis expression, genes with no counts were removed and counts across samples were normalized using the weighted trimmed mean of M-values. Remaining genes without at least three samples with counts were additionally removed leaving 14,288 genes with an average of ∼10 M reads per sample. Differentially expressed genes were determined between treatments relative to time-matched and donor-matched samples using a generalized linear model implemented in edgeR. Cutoffs for differential gene expression were defined as an absolute fold change of 2 and aP value of ≤0.05 after false-discovery rate adjustment using the Benjamini-Hochberg multiple-testing correction (79).
Functional enrichment analysis.
Functional analysis of DE genes was done using Ingenuity Pathway Analysis (IPA; Ingenuity Systems). IPA assesses enrichment of biological pathways and functional categories using a proprietary, manually curated knowledgebase derived from peer-reviewed published studies. IPA functional enrichment was calculated using a right-tailed Fisher exact test with a threshold of significance set at aP value of 0.05. Pathway enrichment is reported as (−log10P value) as determined by the Fisher exact test. Additional functional gene annotation was obtained from the human gene database, GeneCards (http://www.genecards.org) (v4.0.5) (80).
qRT-PCR analysis.
Five nanograms of mRNA was analyzed by quantitative reverse transcription-PCR (qRT-PCR) using the QuantiFast SYBR green RT-PCR kit (Qiagen). qRT-PCR-based quantification of VP35 mRNA was performed using a primer pair that binds to both the EBOV and RESTV VP35 genes without mismatches. Validated QuantiTect primer assays (Qiagen) for β2-microglobulin were used as an endogenous control for normalization. qRT-PCR was performed in a CFX96 real-time PCR cycler (Bio-Rad). Fold expression levels compared to those of noninfected samples collected at each corresponding time point were quantified using the threshold cycle (ΔΔCT) method (Bio-Rad CFX Manager 1.5 software). Statistical analysis was performed with a two-tailedt test using GraphPad Prism 5 software.
Luminex analysis.
At the desired time points after infection or treatment, 1 ml of supernatant was collected from 1.5 × 106 (6-well format) or 3 × 105 (24-well format) MDMs. For inactivation, samples were exposed to 10 megarads of gamma irradiation on dry ice by following IBC-approved SOPs. Samples were clarified by low-speed centrifugation and analyzed using the Procarta human cytokine assay (Affymetrix). Mean fluorescence intensity was measured to calculate final concentration in pictograms per milliliter using Bioplex200 and Bioplex Manager 5 software (Bio-Rad). Statistical analysis of data was performed using one-way analysis of variance (ANOVA) using GraphPad Prism 5 software.
Immunofluorescence analysis.
A total of 2 × 105 MDMs seeded per well of μ-Slide (ibidi) or Lab-Tek II (Nunc) 8-well chamber slides or 3 × 105 MDMs seeded on coverslips were infected or treated as described above. At the desired time points, cells were fixed with 4% paraformaldehyde overnight and removed from the BSL4 facility by following IBC-approved SOPs. Cells were permeabilized with acetone-methanol (1:1) at −20°C for 10 min. Primary antibodies include a mouse anti-EBOV NP antibody (gift from G. Olinger, formerly USAMRIID, Virology Division, Frederick, MD), a mouse anti-RESTV NP antibody, and a rabbit anti-IRF3 antibody and a rabbit anti-p65 antibody (both Santa Cruz). Secondary antibodies were conjugated with Alexa Fluor 495 or Alexa Fluor 488 (Molecular Probes). 4′,6-Diamidino-2-phenylindole (DAPI) (Sigma-Aldrich) was used for nucleus staining. At least 100 cells per sample were counted to determine infection rates or percentage of cells with nuclear localization of p65 or IRF3. GraphPad Prism 5 software was used to calculate standard errors of the means (SEM) and to perform statistical analysis of data using one-way ANOVA for p65 and IRF3 localization.
Western blot analysis.
At the desired time points, 1.5 × 106 (6-well format) MDMs were scraped into phosphate-buffered saline (PBS) and pelleted by low-speed centrifugation. Whole-cell extracts were prepared in cell extraction buffer (Biosource) complemented with protease inhibitor mix (complete; Roche) and serine/threonine phosphatase inhibitor (Calyculin A; CST) according to the manufacturer's protocol. Cell lysates were transferred to fresh tubes containing 2× SDS sample buffer. Inactivation of EBOV or RESTV and corresponding controls was performed by incubating the samples at 95°C for 10 min by following IBC-approved SOPs. Samples were subjected to Western blot analysis using antibodies against IκBα (Santa Cruz) and β-actin (Abcam). Secondary antibodies (Dianova) were conjugated with horseradish peroxidase. Detection was performed using SuperSignal chemiluminescent substrate (Pierce/Thermo Scientific) according to the manufacturer's protocol. For VLP analysis, an aliquot of each VLP preparation was mixed with 2× SDS sample buffer and subjected to Western blot analysis using antibodies against EBOV GP1, RESTV GP1 (IBT Bioservices), and EBOV VP40. The EBOV VP40 antibody cross-reacts with RESTV VP40. The secondary antibody was conjugated to IRDye800 (Li-Cor), and protein bands were visualized using the Odyssey imaging system and software (Li-Cor).
DNA binding assay.
At the desired time points, nuclear lysates from 6 × 106 MDMs were prepared using the NucBuster protein extraction kit (Novagen). Protein concentration was determined using the bicinchoninic acid (BCA) protein assay kit (Pierce/Thermo Scientific), and 5 μg of protein per sample was analyzed using the TransAM NF-κB family kit assay (Active Motif) according to the manufacturer's instructions. Statistical analysis of data was performed using one-way ANOVA with GraphPad Prism 5 software.
Accession number(s).
Raw and normalized sequencing data files and differentially expressed genes were deposited in the public Gene Expression Omnibus (GEO) database (http://www.ncbi.nlm.nih.gov/geo/) under accession numberGSE84188.
ACKNOWLEDGMENTS
We are indebted to H. Feldmann, F. Feldmann, E. Haddock, and members of the Laboratory of Virology for training of J.O. and L.R.D. and support with conducting BSL4 experiments at the Rocky Mountain Laboratories, NIAID, NIH, Hamilton, MT. We thank S. D'Souza and T. Kepler (both Boston University) for help with the statistical analysis, G. Viglianti (Boston University) and G. Olinger (formerly USAMRIID, Virology Division, Frederick, MD), for providing material, T. Geijtenbeek and B. Wevers (Vrije University Medical Center, Amsterdam, Netherlands) for technical advice, and J. Pacheco (Boston University) for excellent technical assistance.
REFERENCES
- 1.Rougeron V, Feldmann H, Grard G, Becker S, Leroy EM. 2015. Ebola and Marburg haemorrhagic fever. J Clin Virol64:111–119. doi: 10.1016/j.jcv.2015.01.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.McElroy AK, Harmon JR, Flietstra TD, Campbell S, Mehta AK, Kraft CS, Lyon MG, Varkey JB, Ribner BS, Kratochvil CJ, Iwen PC, Smith PW, Ahmed R, Nichol ST, Spiropoulou CF. 27June2016. Kinetic analysis of biomarkers in a cohort of US patients with Ebola virus disease. Clin Infect Dis doi: 10.1093/cid/ciw334. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Ruibal P, Oestereich L, Lüdtke A, Becker-Ziaja B, Wozniak DM, Kerber R, Korva M, Cabeza-Cabrerizo M, Bore JA, Koundouno FR, Duraffour S, Weller R, Thorenz A, Cimini E, Viola D, Agrati C, Repits J, Afrough B, Cowley LA, Ngabo D, Hinzmann J, Mertens M, Vitoriano I, Logue CH, Boettcher JP, Pallasch E, Sachse A, Bah A, Nitzsche K, Kuisma E, Michel J, Holm T, Zekeng E-G, García-Dorival I, Wölfel R, Stoecker K, Fleischmann E, Strecker T, Di Caro A, Avšič-Županc T, Kurth A, Meschi S, Mély S, Newman E, Bocquin A, Kis Z, Kelterbaum A, Molkenthin P, Carletti F, Portmann J, Wolff S, Castilletti C, Schudt G, Fizet A, Ottowell LJ, Herker E, Jacobs T, Kretschmer B, Severi E, Ouedraogo N, Lago M, Negredo A, Franco L, Anda P, Schmiedel S, Kreuels B, Wichmann D, Addo MM, Lohse AW, De Clerck H, Nanclares C, Jonckheere S, Van Herp M, Sprecher A, Xiaojiang G, Carrington M, Miranda O, Castro CM, Gabriel M, Drury P, Formenty P, Diallo B, Koivogui L, Magassouba N, Carroll MW, Günther S, Muñoz-Fontela C. 2016. Unique human immune signature of Ebola virus disease in Guinea. Nature533:100–104. doi: 10.1038/nature17949. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Villinger F, Rollin PE, Brar SS, Chikkala NF, Winter J, Sundstrom JB, Zaki SR, Swanepoel R, Ansari AA, Peters CJ. 1999. Markedly elevated levels of interferon (IFN)-gamma, IFN-alpha, interleukin (IL)-2, IL-10, and tumor necrosis factor-alpha associated with fatal Ebola virus infection. J Infect Dis179(Suppl):S188–S191. [DOI] [PubMed] [Google Scholar]
- 5.Baize S, Leroy EM, Georges-Courbot MC, Capron M, Lansoud-Soukate J, Debré P, Fisher-Hoch SP, McCormick JB, Georges AJ, Debre P. 1999. Defective humoral responses and extensive intravascular apoptosis are associated with fatal outcome in Ebola virus-infected patients. Nat Med5:423–426. doi: 10.1038/7422. [DOI] [PubMed] [Google Scholar]
- 6.Gupta M, Mahanty S, Ahmed R, Rollin PE. 2001. Monocyte-derived human macrophages and peripheral blood mononuclear cells infected with Ebola virus secrete MIP-1alpha and TNF-alpha and inhibit poly-IC-induced IFN-alpha in vitro. Virology284:20–25. doi: 10.1006/viro.2001.0836. [DOI] [PubMed] [Google Scholar]
- 7.Leroy EM, Baize S, Debre P, Lansoud-Soukate J, Mavoungou E. 2001. Early immune responses accompanying human asymptomatic Ebola infections. Clin Exp Immunol124:453–460. doi: 10.1046/j.1365-2249.2001.01517.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Wauquier N, Becquart P, Padilla C, Baize S, Leroy EM. 2010. Human fatal Zaire Ebola virus infection is associated with an aberrant innate immunity and with massive lymphocyte apoptosis. PLoS Negl Trop Dis4(10):e837. doi: 10.1371/journal.pntd.0000837. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Bixler S, Goff A. 2015. The role of cytokines and chemokines in filovirus infection. Viruses7:5489–5507. doi: 10.3390/v7102892. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Miranda MEG, Miranda NLJ. 2011. Reston ebolavirus in humans and animals in the Philippines: a review. J Infect Dis204(Suppl):S757–S760. [DOI] [PubMed] [Google Scholar]
- 11.Fisher-Hoch SP, Brammer TL, Trappier SG, Hutwagner LC, Farrar BB, Ruo SL, Brown BG, Hermann LM, Perez-Oronoz GI, Goldsmith CS, Hanes MA, McCormick JB. 1992. Pathogenic potential of filoviruses: role of geographic origin of primate host and virus strain. J Infect Dis166:753–763. doi: 10.1093/infdis/166.4.753. [DOI] [PubMed] [Google Scholar]
- 12.Geisbert TW, Strong JE, Feldmann H. 2015. Considerations in the use of nonhuman primate models of Ebola virus and Marburg virus infection. J Infect Dis212(Suppl):S91–S97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Groseth A, Marzi A, Hoenen T, Herwig A, Gardner D, Becker S, Ebihara H, Feldmann H. 2012. The Ebola virus glycoprotein contributes to but is not sufficient for virulence in vivo. PLoS Pathog8:e1002847. doi: 10.1371/journal.ppat.1002847. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Brannan JM, Froude JW, Prugar LI, Bakken RR, Zak SE, Daye SP, Wilhelmsen CE, Dye JM. 2015. Interferon α/β receptor-deficient mice as a model for Ebola virus disease. J Infect Dis212(Suppl 2):S282–S294. doi: 10.1093/infdis/jiv215. [DOI] [PubMed] [Google Scholar]
- 15.de Wit E, Munster VJ, Metwally SA, Feldmann H. 2011. Assessment of rodents as animal models for Reston ebolavirus. J Infect Dis204(Suppl):S968–S972. doi: 10.1093/infdis/jir330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Raymond J, Bradfute S, Bray M. 2011. Filovirus infection of STAT-1 knockout mice. J Infect Dis204(Suppl):S986–S990. [DOI] [PubMed] [Google Scholar]
- 17.Messaoudi I, Amarasinghe GK, Basler CF. 2015. Filovirus pathogenesis and immune evasion: insights from Ebola virus and Marburg virus. Nat Rev Microbiol13:663–676. doi: 10.1038/nrmicro3524. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Rasmussen AL.2016. Host factors in Ebola infection. Annu Rev Genomics Hum Genet17:333–351. doi: 10.1146/annurev-genom-083115-022446. [DOI] [PubMed] [Google Scholar]
- 19.Basler CF, Mikulasova A, Martinez-Sobrido L, Paragas J, Muhlberger E, Bray M, Klenk HD, Palese P, Garcia-Sastre A, Mühlberger E, García-Sastre A. 2003. The Ebola virus VP35 protein inhibits activation of interferon regulatory factor 3. J Virol77:7945–7956. doi: 10.1128/JVI.77.14.7945-7956.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Reid SP, Valmas C, Martinez O, Sanchez FM, Basler CF. 2007. Ebola virus VP24 proteins inhibit the interaction of NPI-1 subfamily karyopherin alpha proteins with activated STAT1. J Virol81:13469–13477. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Leung DW, Prins KC, Borek DM, Farahbakhsh M, Tufariello JM, Ramanan P, Nix JC, Helgeson LA, Otwinowski Z, Honzatko RB, Basler CF, Amarasinghe GK. 2010. Structural basis for dsRNA recognition and interferon antagonism by Ebola VP35. Nat Struct Mol Biol17:165–172. doi: 10.1038/nsmb.1765. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Kimberlin CR, Bornholdt ZA, Li S, Woods VL, MacRae IJ, Saphire EO. 2010. Ebolavirus VP35 uses a bimodal strategy to bind dsRNA for innate immune suppression. Proc Natl Acad Sci U S A107:314–319. doi: 10.1073/pnas.0910547107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Guito JC, Albariño CG, Chakrabarti AK, Towner JS. 2017. Novel activities by ebolavirus and marburgvirus interferon antagonists revealed using a standardized in vitro reporter system. Virology501:147–165. doi: 10.1016/j.virol.2016.11.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Schwarz TM, Edwards MR, Diederichs A, Alinger JB, Leung DW, Amarasinghe GK, Basler CF. 14December2016. VP24-karyopherin alpha binding affinities differ betweenEbolavirus species influencing interferon inhibition and VP24 stability. J Virol doi: 10.1128/JVI.01715-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Olejnik J, Ryabchikova E, Corley RB, Mühlberger E. 2011. Intracellular events and cell fate in filovirus infection. Viruses3:1501–1531. doi: 10.3390/v3081501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Martinez O, Johnson JC, Honko A, Yen B, Shabman RS, Hensley LE, Olinger GG, Basler CF. 2013. Ebola virus exploits a monocyte differentiation program to promote its entry. J Virol87:3801–3814. doi: 10.1128/JVI.02695-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Ströher U, West E, Bugany H, Klenk H-D, Schnittler H-J, Feldmann H. 2001. Infection and activation of monocytes by Marburg and Ebola viruses. J Virol75:11025–11033. doi: 10.1128/JVI.75.22.11025-11033.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Hensley LE, Young HA, Jahrling PB, Geisbert TW. 2002. Proinflammatory response during Ebola virus infection of primate models: possible involvement of the tumor necrosis factor receptor superfamily. Immunol Lett80:169–179. doi: 10.1016/S0165-2478(01)00327-3. [DOI] [PubMed] [Google Scholar]
- 29.Wahl-Jensen V, Kurz S, Feldmann F, Buehler LK, Kindrachuk J, Defilippis V, da Silva Correia J, Früh K, Kuhn JH, Burton DR, Feldmann H. 2011. Ebola virion attachment and entry into human macrophages profoundly effects early cellular gene expression. PLoS Negl Trop Dis5:e1359. doi: 10.1371/journal.pntd.0001359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Escudero-Pérez B, Volchkova VA, Dolnik O, Lawrence P, Volchkov VE. 2014. Shed GP of Ebola virus triggers immune activation and increased vascular permeability. PLoS Pathog10:e1004509. doi: 10.1371/journal.ppat.1004509. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Wahl-Jensen V, Kurz SK, Hazelton PR, Schnittler HJ, Stroher U, Burton DR, Feldmann H. 2005. Role of Ebola virus secreted glycoproteins and virus-like particles in activation of human macrophages. J Virol79:2413–2419. doi: 10.1128/JVI.79.4.2413-2419.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Okumura A, Pitha PM, Yoshimura A, Harty RN. 2010. Interaction between Ebola virus glycoprotein and host Toll-like receptor 4 leads to induction of proinflammatory cytokines and SOCS1. J Virol84:27–33. doi: 10.1128/JVI.01462-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Okumura A, Rasmussen AL, Halfmann P, Feldmann F, Yoshimura A, Feldmann H, Kawaoka Y, Harty RN, Katze MG. 2015. Suppressor of cytokine signaling 3 is an inducible host factor that regulates virus egress during Ebola virus infection. J Virol89:10399–10406. doi: 10.1128/JVI.01736-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Zhao D, Han X, Zheng X, Wang H, Yang Z, Liu D, Han K, Liu J, Wang X, Yang W, Dong Q, Yang S, Xia X, Tang L, He F. 2016. The myeloid LSECtin is a DAP12-coupled receptor that is crucial for inflammatory response induced by Ebola virus glycoprotein. PLoS Pathog12:e1005487. doi: 10.1371/journal.ppat.1005487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Baize S, Leroy EM, Georges AJ, Georges-Courbot M-CC, Capron M, Bedjabaga I, Lansoud-Soukate J, Mavoungou E. 2002. Inflammatory responses in Ebola virus-infected patients. Clin Exp Immunol128:163–168. doi: 10.1046/j.1365-2249.2002.01800.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Honda K, Taniguchi T. 2006. IRFs: master regulators of signalling by Toll-like receptors and cytosolic pattern-recognition receptors. Nat Rev Immunol6:644–658. doi: 10.1038/nri1900. [DOI] [PubMed] [Google Scholar]
- 37.Nelson EV, Schmidt KM, Deflubé LR, Doğanay S, Banadyga L, Olejnik J, Hume AJ, Ryabchikova E, Ebihara H, Kedersha N, Ha T, Mühlberger E. 2016. Ebola virus does not induce stress granule formation during infection and sequesters stress granule proteins within viral inclusions. J Virol90:7268–7284. doi: 10.1128/JVI.00459-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Hoenen T, Shabman RS, Groseth A, Herwig A, Weber M, Schudt G, Dolnik O, Basler CF, Becker S, Feldmann H. 2012. Inclusion bodies are a site of ebolavirus replication. J Virol86:11779–11788. doi: 10.1128/JVI.01525-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Lawrence T.2009. The nuclear factor NF-kappaB pathway in inflammation. Cold Spring Harb Perspect Biol1(6):a001651. doi: 10.1101/cshperspect.a001651. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Boehmann Y, Enterlein S, Randolf A, Mühlberger E. 2005. A reconstituted replication and transcription system for Ebola virus Reston and comparison with Ebola virus Zaire. Virology332:406–417. doi: 10.1016/j.virol.2004.11.018. [DOI] [PubMed] [Google Scholar]
- 41.Kash JC, Mu E, Carter V, Grosch M, Perwitasari O, Proll SC, Thomas MJ, Weber F, Klenk HD, Katze MG, Muhlberger E, Mühlberger E. 2006. Global suppression of the host antiviral response by Ebola- and Marburgviruses: increased antagonism of the type I interferon response is associated with enhanced virulence. J Virol80:3009–3020. doi: 10.1128/JVI.80.6.3009-3020.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Alfson KJ, Avena LE, Beadles MW, Staples H, Nunneley JW, Ticer A, Dick EJ, Owston MA, Reed C, Patterson JL, Carrion R, Griffiths A. 2015. Particle-to-PFU ratio of Ebola virus influences disease course and survival in cynomolgus macaques. J Virol89:6773–6781. doi: 10.1128/JVI.00649-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Golenbock DT, Hampton RY, Qureshi N, Takayama K, Raetz CRH. 1991. Lipid A-like molecules that antagonize the effects of endotoxins on human monocytes. J Biol Chem266:19490–19498. [PubMed] [Google Scholar]
- 44.Kutuzova GD, Albrecht RM, Erickson CM, Qureshi N. 2001. Diphosphoryl lipid A from Rhodobacter sphaeroides blocks the binding and internalization of lipopolysaccharide in RAW 264.7 cells. J Immunol167:482–489. doi: 10.4049/jimmunol.167.1.482. [DOI] [PubMed] [Google Scholar]
- 45.Hartman AL, Ling L, Nichol ST, Hibberd ML. 2008. Whole-genome expression profiling reveals that inhibition of host innate immune response pathways by Ebola virus can be reversed by a single amino acid change in the VP35 protein. J Virol82:5348–5358. doi: 10.1128/JVI.00215-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Melchjorsen J.2013. Learning from the messengers: innate sensing of viruses and cytokine regulation of immunity—clues for treatments and vaccines. Viruses5:470–527. doi: 10.3390/v5020470. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Boehme KW, Compton T. 2004. Innate sensing of viruses by Toll-like receptors. J Virol78:7867–7873. doi: 10.1128/JVI.78.15.7867-7873.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Georgel P, Jiang Z, Kunz S, Janssen E, Mols J, Hoebe K, Bahram S, Oldstone MBA, Beutler B. 2007. Vesicular stomatitis virus glycoprotein G activates a specific antiviral Toll-like receptor 4-dependent pathway. Virology362:304–313. doi: 10.1016/j.virol.2006.12.032. [DOI] [PubMed] [Google Scholar]
- 49.Kurt-Jones EA, Popova L, Kwinn L, Haynes LM, Jones LP, Tripp RA, Walsh EE, Freeman MW, Golenbock DT, Anderson LJ, Finberg RW. 2000. Pattern recognition receptors TLR4 and CD14 mediate response to respiratory syncytial virus. Nat Immunol1:398–401. doi: 10.1038/80833. [DOI] [PubMed] [Google Scholar]
- 50.Rallabhandi P, Phillips RL, Boukhvalova MS, Pletneva LM, Shirey KA, Gioannini TL, Weiss JP, Chow JC, Hawkins LD, Vogel SN, Blanco JCG. 2012. Respiratory syncytial virus fusion protein-induced Toll-like receptor 4 (TLR4) signaling is inhibited by the TLR4 antagonists Rhodobacter sphaeroides lipopolysaccharide and eritoran (E5564) and requires direct interaction with MD-2. mBio3:e00218-12. doi: 10.1128/mBio.00218-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Lu Y-C, Yeh W-C, Ohashi PS. 2008. LPS/TLR4 signal transduction pathway. Cytokine42:145–151. doi: 10.1016/j.cyto.2008.01.006. [DOI] [PubMed] [Google Scholar]
- 52.Kim HM, Park BS, Kim J-I, Kim SE, Lee J, Oh SC, Enkhbayar P, Matsushima N, Lee H, Yoo OJ, Lee J-O. 2007. Crystal structure of the TLR4-MD-2 complex with bound endotoxin antagonist Eritoran. Cell130:906–917. doi: 10.1016/j.cell.2007.08.002. [DOI] [PubMed] [Google Scholar]
- 53.Park BS, Song DH, Kim HM, Choi B-S, Lee H, Lee J-O. 2009. The structural basis of lipopolysaccharide recognition by the TLR4-MD-2 complex. Nature458:1191–1195. doi: 10.1038/nature07830. [DOI] [PubMed] [Google Scholar]
- 54.Dominguez-Soto A, Aragoneses-Fenoll L, Martin-Gayo E, Martinez-Prats L, Colmenares M, Naranjo-Gomez M, Borras FE, Munoz P, Zubiaur M, Toribio ML, Delgado R, Corbi AL. 2007. The DC-SIGN-related lectin LSECtin mediates antigen capture and pathogen binding by human myeloid cells. Blood109:5337–5345. doi: 10.1182/blood-2006-09-048058. [DOI] [PubMed] [Google Scholar]
- 55.Modhiran N, Watterson D, Muller DA, Panetta AK, Sester DP, Liu L, Hume DA, Stacey KJ, Young PR. 2015. Dengue virus NS1 protein activates cells via Toll-like receptor 4 and disrupts endothelial cell monolayer integrity. Sci Transl Med7:304ra142. doi: 10.1126/scitranslmed.aaa3863. [DOI] [PubMed] [Google Scholar]
- 56.Rialdi A, Campisi L, Zhao N, Lagda AC, Pietzsch C, Ho JSY, Martinez-Gil L, Fenouil R, Chen X, Edwards M, Metreveli G, Jordan S, Peralta Z, Munoz-Fontela C, Bouvier N, Merad M, Jin J, Weirauch M, Heinz S, Benner C, van Bakel H, Basler C, Garcia-Sastre A, Bukreyev A, Marazzi I. 2016. Topoisomerase 1 inhibition suppresses inflammatory genes and protects from death by inflammation. Science352:aad7993. doi: 10.1126/science.aad7993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Latz E, Visintin A, Lien E, Fitzgerald KA, Monks BG, Kurt-Jones EA, Golenbock DT, Espevik T. 2002. Lipopolysaccharide rapidly traffics to and from the Golgi apparatus with the Toll-like receptor 4-MD-2-CD14 complex in a process that is distinct from the initiation of signal transduction. J Biol Chem277:47834–47843. doi: 10.1074/jbc.M207873200. [DOI] [PubMed] [Google Scholar]
- 58.Kagan JC, Medzhitov R. 2006. Phosphoinositide-mediated adaptor recruitment controls Toll-like receptor signaling. Cell125:943–955. doi: 10.1016/j.cell.2006.03.047. [DOI] [PubMed] [Google Scholar]
- 59.Fitzgerald KA, Rowe DC, Barnes BJ, Caffrey DR, Visintin A, Latz E, Monks B, Pitha PM, Golenbock DT. 2003. LPS-TLR4 signaling to IRF-3/7 and NF-kappaB involves the toll adapters TRAM and TRIF. J Exp Med198:1043–1055. doi: 10.1084/jem.20031023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Yamamoto M, Sato S, Hemmi H, Uematsu S, Hoshino K, Kaisho T, Takeuchi O, Takeda K, Akira S. 2003. TRAM is specifically involved in the Toll-like receptor 4-mediated MyD88-independent signaling pathway. Nat Immunol4:1144–1150. doi: 10.1038/ni986. [DOI] [PubMed] [Google Scholar]
- 61.Kagan JC, Su T, Horng T, Chow A, Akira S, Medzhitov R. 2008. TRAM couples endocytosis of Toll-like receptor 4 to the induction of interferon-β. Nat Immunol9:361–368. doi: 10.1038/ni1569. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Ayithan N, Bradfute SB, Anthony SM, Stuthman KS, Dye JM, Bavari S, Bray M, Ozato K. 2014. Ebola virus-like particles stimulate type I interferons and proinflammatory cytokine expression through the toll-like receptor and interferon signaling pathways. J Interferon Cytokine Res34(2):79–89. doi: 10.1089/jir.2013.0035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Mingo RM, Simmons JA, Shoemaker CJ, Nelson EA, Schornberg KL, D'Souza RS, Casanova JE, White JM. 2015. Ebola virus and severe acute respiratory syndrome coronavirus display late cell entry kinetics: evidence that transport to NPC1+ endolysosomes is a rate-defining step. J Virol89:2931–2943. doi: 10.1128/JVI.03398-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Simmons JA, D'Souza RS, Ruas M, Galione A, Casanova JE, White JM. 14October2015. The Ebolavirus glycoprotein directs fusion through NPC1+ endolysosomes. J Virol doi: 10.1128/JVI.01828-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Reimer T, Brcic M, Schweizer M, Jungi TW. 2008. poly(I:C) and LPS induce distinct IRF3 and NF-kappaB signaling during type-I IFN and TNF responses in human macrophages. J Leukoc Biol83:1249–1257. doi: 10.1189/jlb.0607412. [DOI] [PubMed] [Google Scholar]
- 66.Solis M, Romieu-Mourez R, Goubau D, Grandvaux N, Mesplede T, Julkunen I, Nardin A, Salcedo M, Hiscott J. 2007. Involvement of TBK1 and IKKepsilon in lipopolysaccharide-induced activation of the interferon response in primary human macrophages. Eur J Immunol37:528–539. doi: 10.1002/eji.200636090. [DOI] [PubMed] [Google Scholar]
- 67.Liu X, Speranza E, Muñoz-Fontela C, Haldenby S, Rickett NY, Garcia-Dorival I, Fang Y, Hall Y, Zekeng E-G, Lüdtke A, Xia D, Kerber R, Krumkamp R, Duraffour S, Sissoko D, Kenny J, Rockliffe N, Williamson ED, Laws TR, N′Faly M, Matthews DA, Günther S, Cossins AR, Sprecher A, Connor JH, Carroll MW, Hiscox JA. 2017. Transcriptomic signatures differentiate survival from fatal outcomes in humans infected with Ebola virus. Genome Biol18:4. doi: 10.1186/s13059-016-1137-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Bray M, Davis K, Geisbert T, Schmaljohn C, Huggins J. 1999. A mouse model for evaluation of prophylaxis and therapy of Ebola hemorrhagic fever. J Infect Dis179(Suppl):S248–S258. [DOI] [PubMed] [Google Scholar]
- 69.Leung DW, Shabman RS, Farahbakhsh M, Prins KC, Borek DM, Wang T, Mühlberger E, Basler CF, Amarasinghe GK. 2010. Structural and functional characterization of Reston Ebola virus VP35 interferon inhibitory domain. J Mol Biol399:347–357. doi: 10.1016/j.jmb.2010.04.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Zhang APP, Bornholdt ZA, Liu T, Abelson DM, Lee DE, Li S, Woods VL, Saphire EO. 2012. The ebola virus interferon antagonist VP24 directly binds STAT1 and has a novel, pyramidal fold. PLoS Pathog8:e1002550. doi: 10.1371/journal.ppat.1002550. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Pappalardo M, Juliá M, Howard MJ, Rossman JS, Michaelis M, Wass MN. 2016. Conserved differences in protein sequence determine the human pathogenicity of Ebolaviruses. Sci Rep6:23743. doi: 10.1038/srep23743. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Martins K, Cooper C, Warren T, Wells J, Bell T, Raymond J, Stuthman K, Benko J, Garza N, van Tongeren S, Donnelly G, Retterer C, Dong L, Bavari S. 2015. Characterization of clinical and immunological parameters during ebola virus infection of rhesus macaques. Viral Immunol28:32–41. doi: 10.1089/vim.2014.0085. [DOI] [PubMed] [Google Scholar]
- 73.Licata JM, Johnson RF, Han Z, Harty RN. 2004. Contribution of ebola virus glycoprotein, nucleoprotein, and VP24 to budding of VP40 virus-like particles. J Virol78:7344–7351. doi: 10.1128/JVI.78.14.7344-7351.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Kallstrom G, Warfield KL, Swenson DL, Mort S, Panchal RG, Ruthel G, Bavari S, Aman MJ. 2005. Analysis of Ebola virus and VLP release using an immunocapture assay. J Virol Methods127:1–9. doi: 10.1016/j.jviromet.2005.02.015. [DOI] [PubMed] [Google Scholar]
- 75.Langmead B, Salzberg SL. 2012. Fast gapped-read alignment with Bowtie 2. Nat Methods9:357–359. doi: 10.1038/nmeth.1923. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, Batut P, Chaisson M, Gingeras TR. 2013. STAR: ultrafast universal RNA-seq aligner. Bioinformatics29:15–21. doi: 10.1093/bioinformatics/bts635. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Anders S, Pyl PT, Huber W. 2015. HTSeq—a Python framework to work with high-throughput sequencing data. Bioinformatics31:166–169. doi: 10.1093/bioinformatics/btu638. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Robinson MD, McCarthy DJ, Smyth GK. 2010. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics26:139–140. doi: 10.1093/bioinformatics/btp616. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Benjamini Y, Hochberg Y. 1995. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J R Stat Soc57:289–300. [Google Scholar]
- 80.Rebhan M, Chalifa-Caspi V, Prilusky J, Lancet D. 1998. GeneCards: a novel functional genomics compendium with automated data mining and query reformulation support. Bioinformatics14:656–664. doi: 10.1093/bioinformatics/14.8.656. [DOI] [PubMed] [Google Scholar]