ABSTRACT
The nuclear factor kappa B (NF-κB) pathway is known to integrate signaling associated with very diverse intra- and extracellular stressors, including virus infections, and triggers a powerful (proinflammatory) response through the expression of NF-κB-regulated genes. Typically, the NF-κB pathway collects and transduces threatening signals at the cell surface or in the cytoplasm leading to nuclear import of activated NF-κB transcription factors. In the current work, we demonstrate that the swine alphaherpesvirus pseudorabies virus (PRV) induces a peculiar mode of NF-κB activation known as “inside-out” NF-κB activation. We show that PRV triggers the DNA damage response (DDR) and that this DDR response drives NF-κB activation since inhibition of the nuclear ataxia telangiectasia-mutated (ATM) kinase, a chief controller of DDR, abolished PRV-induced NF-κB activation. Initiation of the DDR-NF-κB signaling axis requires viral protein synthesis but occurs before active viral genome replication. In addition, the initiation of the DDR-NF-κB signaling axis is followed by a virus-induced complete shutoff of NF-κB-dependent gene expression that depends on viral DNA replication. In summary, the results presented in this study reveal that PRV infection triggers a noncanonical DDR-NF-κB activation signaling axis and that the virus actively inhibits the (potentially antiviral) consequences of this pathway, by inhibiting NF-κB-dependent gene expression.
IMPORTANCE The NF-κB signaling pathway plays a critical role in coordination of innate immune responses that are of vital importance in the control of infections. The current report generates new insights into the interaction of the alphaherpesvirus pseudorabies virus (PRV) with the NF-κB pathway, as they reveal that (i) PRV infection leads to NF-κB activation via a peculiar “inside-out” nucleus-to-cytoplasm signal that is triggered via the DNA damage response (DDR), (ii) the DDR-NF-κB signaling axis requires expression of viral proteins but is initiated before active PRV replication, and (iii) late viral factor(s) allow PRV to actively and efficiently inhibit NF-κB-dependent (proinflammatory) gene expression. These data suggest that activation of the DDR-NF-κB during PRV infection is host driven and that its potential antiviral consequences are actively inhibited by the virus.
KEYWORDS: DDR, DNA damage response, NF-κB, herpes, pseudorabies virus
INTRODUCTION
Pseudorabies virus (PRV) is a porcine alphaherpesvirus and constitutes the etiological agent of the multiorgan syndrome termed Aujeszky’s disease (1). Alphaherpesvirinae represent the largest subfamily of Herpesviridae and include a.o. human herpes simplex viruses (HSV-1 and HSV-2) and varicella zoster virus (VZV) and several animal viruses, including equine herpesvirus 1 (EHV-1), bovine herpesvirus 1 (BHV-1), and feline herpesvirus 1 (FeHV-1) (2). Alphaherpesviruses display complex and fine-balanced interactions with the cells and immune system of their natural host, and PRV is often used as a model organism to study general alphaherpesvirus-host interactions (3).
The nuclear factor kappa B (NF-κB) signaling pathway is a leading regulator of innate immunity and inflammation and is activated by endangering and/or infectious stimuli, culminating in the activation of NF-κB/Rel transcription factors that trigger expression of a variety of genes, including several inflammatory genes (4). An accurate regulation of NF-κB-driven responses is imperative and is reflected by the serious outcomes caused by NF-κB-associated pathogenesis, including cancer and autoimmune and cardiovascular diseases, among several others (5–8). Pathogen recognition and cellular detection of proinflammatory cytokines and/or a wide diversity of intracellular stressors are molecular triggers of NF-κB. Triggering signals are located typically at the cell surface and/or in the cytoplasm and lead to activation of the inhibitory kappa B kinase (IKK) complex that contains the IKKβ kinase. The IKKβ kinase phosphorylates inhibitory kappa B (IκB) proteins (particularly IκBα) that are associated with NF-κB and retain it in the cytoplasm. Phosphorylation tags the IκB proteins for ubiquitin-dependent proteasomal degradation, thereby releasing NF-κB and allowing it to translocate to the nucleus and drive expression of the genes that harbor NF-κB-responsive promoters (9, 10).
Recently, our lab reported that PRV triggers persistent activation of NF-κB in infected porcine epithelial cells, although this does not appear to result in the expression of NF-κB-driven genes (11). PRV infection causes progressive IκBα protein degradation starting from 4 hours postinfection (hpi), which is accompanied by nuclear import of the p65/RelA subunit of NF-κB. The trigger for NF-κB activation during PRV infection was found to be largely independent of IKKβ kinase activity, in contrast to canonical activation of NF-κB, suggesting a noncanonical pathway involved in PRV-mediated NF-κB activation (11), although the nature of the trigger of NF-κB activation during PRV remained unclear.
In the current report, we aimed at generating new insights in the stage of PRV replication and the cellular pathway that are involved in NF-κB activation during PRV infection. We show that (i) NF-κB activation precedes viral DNA synthesis, (ii) PRV-induced NF-κB is triggered via a peculiar “inside-out” signaling initiated by double-stranded DNA damage (DDR) in an ataxia telangiectasia-mutated (ATM)-dependent way, (iii) inhibition of the DDR-NF-κB signaling axis does not suppress virus replication in cell culture, and (iv) treatment of PRV-infected cells with the DNA replication inhibitor phosphonoacetic acid does not prevent NF-κB activation but releases NF-κB transcriptional activity indicating that viral late factor(s) inhibit expression of NF-κB-dependent genes. Altogether, these data shed new light on the interaction of alphaherpesviruses with the NF-κB signaling axis and how this may modulate the host innate immune response against these pathogens.
RESULTS
NF-κB activation by PRV does not rely on generation of infectious virus and precedes viral DNA replication.
We showed earlier (i) that PRV infection triggers persistent activation of NF-κB but (ii) that PRV inhibits NF-κB-dependent gene expression (11). Hence, in the context of PRV infection and the current work, activation of NF-κB refers to permanent degradation of IκBα and nuclear import of NF-κB p65 but is not associated with productive transcription of NF-κB-induced genes. The lytic infection cycle of alphaherpesviruses can be subdivided in two stages. During the prereplicative phase, input genomes serve as the template for the expression of immediate early and early viral genes, which encode mainly proteins involved in the modulation of viral gene expression and in genome replication. Genome replication initiates the second phase of the replication cycle, including the expression of viral late genes that encode mostly structural proteins or factors involved in virus particle production and egress from infected cells. We reported before that, in the presence of the viral DNA polymerase inhibitor phosphonoacetic acid (PAA), a clear IκBα Western blot band could be observed in PRV-infected cells (11). The fact that the IκBα protein was detected in PAA-treated PRV-infected cells may indicate that PAA treatment prevents IκBα degradation and hence that the trigger for NF-κB activation is a PAA-sensitive late event in PRV replication. However, an alternative explanation for the IκBα protein in PAA-treated PRV-infected cells may be that PAA addition releases the PRV-induced inhibition of NF-κB-dependent gene expression, resulting in expression of NF-κB-driven genes, which include IκBα (12–14). To more carefully address whether late events in PRV replication are involved in NF-κB activation or rather in suppression of NF-κB-dependent gene expression, we made use of a PRV strain expressing a synthetic green fluorescent protein (GFP)-tagged VP26 capsid protein (PRV-GS443) (15), which allows us to visually correlate timing of NF-κB activation with late events in the virus life cycle like formation and nucleo-cytoplasmic shuttling of PRV capsids. First, we confirmed that PRV-GS443, like wild-type (WT) PRV (11), triggers gradual and persistent IκBα degradation between 4 and 6 hpi and that PAA treatment of PRV-GS443-infected cells resulted in a clear IκBα Western blot band (Fig. 1A). Importantly, at 6 hpi, when most of the IκBα protein pool was already degraded by PRV infection, subcellular location of the VP26-GFP capsid protein showed a nearly complete restriction to the nuclei of infected cells (Fig. 1B), suggesting that virus maturation processes, like capsid tegumentation and envelope acquisition that occur in the cytoplasm, are likely not involved in PRV-induced NF-κB activation. This finding was further supported by assessment of extracellular infectious virus titers. As shown in Fig. 1C, infectious virus titers started to increase only after 6 hpi, supporting the idea that the production of infectious virus particles is not involved in NF-κB activation by PRV. To assess whether viral genome replication may serve as a trigger for NF-κB activation, quantitative PCR (qPCR) assays were performed to quantify intracellular PRV genomes at different time points postinoculation (at 2, 4, 6, 8, and 12 hpi), using PAA as negative control for virus genome replication (Fig. 1D). The exponential replication of genomes started from 4 hpi onward, indicating that NF-κB activation might occur before or early during the replicative phase of infection. Initiation of PRV genome replication and NF-κB activation around 4 hpi also corresponded with the timing of detectable expression of viral late proteins by Western blotting (Fig. 1A).
In an attempt to further disentangle viral genome replication from viral late protein expression as putative triggers of NF-κB activation (of which both are inhibited by PAA), late protein expression was blocked by addition of the protein translation inhibitor cycloheximide (CHX) for 4 h starting at 4 hpi, which is just before the onset of viral genome replication and detectable late viral protein expression, and assessed for its effect on IκBα protein degradation. Interestingly, PRV-infected cells treated with CHX from 4 hpi still showed full IκBα degradation at 8 hpi, whereas expression of viral late proteins (e.g., gB and gD) was completely abrogated (Fig. 1E). In line with this result, addition of CHX at 4 hpi did not inhibit nuclear translocation of the p65 subunit of NF-κB (Fig. 1F). These data indicate that the expression of viral late proteins is not required for NF-κB activation in PRV-infected cells, although addition of the viral DNA replication inhibitor PAA rescues IκBα protein levels.
Fascinatingly, addition of both inhibitors, namely, PAA and CHX, resulted in almost complete degradation of IκBα protein levels in PRV-infected cells (Fig. 1G). In line with this finding, the addition of PAA alone resulted in cytoplasmic localization of NF-κB p65, whereas NF-κB p65 was mostly nuclear upon addition of both PAA and CHX (Fig. 1H). These results indicate that the IκBα protein band and the cytoplasmic localization of NF-κB p65 in PAA-treated PRV-infected cells are not caused by an impaired NF-κB activation in the presence of PAA but instead by de novo expression of IκBα (which subsequently recycles NF-κB p65 from the nucleus to the cytoplasm) due to a release of the PRV-induced inhibition of NF-κB-dependent gene expression. Combined, these data therefore show (i) that the trigger for NF-κB activation during PRV infection does not depend on viral genome replication or late viral protein production and (ii) that PRV encodes one or more PAA-sensitive inhibitors of NF-κB-dependent gene expression.
PRV triggers the DNA damage response, and PRV-induced NF-κB activation depends on the activation of the double-stranded DNA (dsDNA) damage kinase ATM.
Several steps of the virus replication cycle take place during the initial 4-h postinoculation step when NF-κB activation is triggered, including virus attachment, virus entry, genome delivery to the host cell nucleus, the expression of viral (immediate) early genes, and preparation of the viral genome for active replication (3).
Since we showed earlier that UV-inactivated PRV does not trigger NF-κB activation (11), NF-κB activation does not appear to rely on the very early stages of virus infection, from virus attachment up to viral genome delivery in the nucleus. Taking this into consideration, we posit that the cell nucleus represents a conceivable source of NF-κB activation possibly through stress associated with PRV infection. Although nuclear activation of NF-κB, so-called “inside-out” NF-κB activation from the nucleus to the cytoplasm, does not represent the most typical NF-κB signaling cascade, induction of NF-κB can be a result of the DNA damage response (DDR) in the cell. Such activation relies on initiation of the DDR-associated ATM kinase in response to dsDNA breaks or oxidative stress (16, 17). To test the possible involvement of this pathway in PRV-induced NF-κB activation, we first tested whether PRV infection triggers the dsDNA DDR during the initial 4 h of infection through Western blot analysis of histone H2A.X phosphorylation at residue Ser139 (γ-H2A.X), a hallmark of DDR activation (18, 19). Our data indicate that PRV infection indeed triggers the DDR between 2 and 4 hpi, corresponding to the same time interval at which the NF-κB pathway is activated (Fig. 2A). Importantly, CHX treatment of PRV-infected cells from the onset of infection or the use of UV-inactivated PRV did not trigger detectable H2A.X phosphorylation (Fig. 2A and 2B), indicating that virus entry and incoming linear viral genomes, exhibiting exposed dsDNA ends in the nucleus, do not trigger a detectable DDR response. As shown in Fig. 2A, phosphorylation of H2A.X increases substantially from 4 to 8 hpi, suggesting that increasing virus DNA replication and/or the consequences of viral replication on host chromatin may further boost DDR activation. In line with this idea, phosphorylated H2A.X levels were substantially reduced at 8 hpi when PRV-infected cells were treated with the viral DNA replication inhibitor PAA (Fig. 2C). Despite the reduction, H2A.X phosphorylation was still noticeable in the presence of PAA, indicating that PRV-induced DDR activation is initiated before active viral DNA replication, corresponding with the timing of NF-κB activation. Addition of CHX at 4 hpi, thereby blocking viral late protein expression but only partially suppressing viral genome replication (Fig. 2D), did not substantially reduce H2A.X phosphorylation at 8 hpi (Fig. 2E), indicating that viral DNA replication itself, rather than viral late protein production, leads to the increased H2A.X phosphorylation between 4 hpi and 8 hpi. Immunofluorescence assays showed that phosphorylated H2A.X colocalizes with host chromatin during late stages of infection (Fig. 2F). Phosphorylated H2A.X is involved in the maintenance of DNA repair factors at the damaged DNA foci (19). The fact that γH2A.X associates mainly with marginalized chromatin rather than with expanded viral replication compartments (VRC) (Fig. 2F) indicates that the DDR occurs predominantly at the host chromatin or that γH2A.X molecules are redirected from VRC to host chromatin.
PRV-induced NF-κB activation is also peculiar since, in comparison to canonical NF-κB activation, IκBα degradation and p65 nuclear translocation occur relatively slowly (hours rather than minutes) and persist for a long time (several hours versus minutes to a few hours) (Fig. 1A) (11). Interestingly, it has been reported that doxorubicin, an agent that activates the DDR by generating dsDNA breaks, also triggers relatively slow and persistent NF-κB activation (20). In line with this concept, we observed that doxorubicin triggered potent H2A.X phosphorylation which correlated with a progressive and irreversible IκBα degradation (Fig. 2G), reminiscent of what we observed for PRV infection and further in line with a potential role for the dsDNA DDR in PRV-induced NF-κB activation.
To investigate whether the DDR is responsible for NF-κB activation during PRV infection, we used the selective ATM kinase inhibitor KU60019, a more potent and selective derivative of the widely used ATM inhibitor KU55933 (21, 22). KU60019 efficiently prevented H2A.X phosphorylation in PRV-infected cells (and doxorubicin-treated cells), indicating that the ATM kinase is the main driver of the PRV-induced DDR (Fig. 3A). Importantly, we found that different doses of the ATM inhibitor (1, 3, and 10 μM) were capable of efficiently preventing IκBα degradation in PRV-infected cells, indicating that the ATM kinase is a central regulator of NF-κB activation triggered by PRV infection (Fig. 3B). Of note and as a control, the ATM inhibitor exclusively repressed IκBα degradation in response to PRV infection or doxorubicin treatment but not upon canonical NF-κB activation via addition of tumor necrosis factor alpha (TNF-α) (Fig. 3C and 3D). As an additional control, the ATM inhibitor also prevented IκBα degradation in PRV-infected cells that were treated with the protein translation inhibitor CHX from 4 hpi to 8 hpi (Fig. 3E), confirming that the IκBα protein band detected in KU60019-treated PRV-infected cells corresponds to nondegraded preexisting IκBα rather than replenished IκBα. The addition of KU60019 to cells infected with two additional PRV wild-type strains (PRV Kaplan and PRV NIA3) showed similar phenotypes, i.e., full inhibition of H2A.X phosphorylation and substantial inhibition of IκBα degradation (Fig. 3F), indicating that the results are not PRV strain dependent.
A time course analysis showed that the ATM inhibitor very effectively represses IκBα degradation at 6 hpi, during the starting time interval of IκBα degradation from 4 to 6 hpi (Fig. 3G). At later time points of infection and particularly somewhere from 8 to 12 hpi, the ATM inhibitor KU60019 is unable to completely block the massive DDR caused by progressive PRV infection, illustrated by incomplete inhibition of H2A.X phosphorylation and IκBα degradation (Fig. 3G). In agreement with the IκBα degradation data, migration of NF-κB p65 to infected cell nuclei was considerably reduced with KU60019, although again not abolished completely (Fig. 3H). As controls, KU60019 inhibited nuclear transport of NF-κB p65 upon doxorubicin but not upon TNF-α treatment (Fig. 3H).
Combined, these results indicate that the ATM kinase is the main, if not the only, driver of NF-κB activation during PRV infection. In conclusion, PRV infection triggers the DNA damage response which in turn leads to NF-κB activation.
Potential causes of PRV-induced DDR and NF-κB activation.
The fact that PRV induces NF-κB activation through a pathway involved in sensing and repair of dsDNA damage led us to first investigate if the consequences of virus replication on the integrity of the cell nucleus may contribute to the DDR-dependent NF-κB activation. To this end, we analyzed nucleus enlargement by measuring nuclear areas at the nuclei medial section and visually assessed chromatin marginalization (the displacement of the host genome to the borders of the nucleus) (Fig. 4A). Our data show that a clear nuclear size increase and chromatin marginalization can be observed at 8 hpi. However, at 4 hpi, when the NF-κB pathway has already been triggered, no significant increase in nuclear size and only a modest chromatin marginalization could be observed (Fig. 4A), with the latter probably representing the very primary VRC. In addition, although PAA treatment does not prevent NF-κB activation, it virtually abolishes chromatin marginalization and nuclear size increase (Fig. 4A). Hence, these data indicate that PRV-induced NF-κB activation does not correlate with nuclear expansion and deformation.
Our data indicate that PRV-induced NF-κB activation precedes viral genome replication and depends on viral protein expression, which suggests that NF-κB is triggered in a time window that encompasses circularization of the PRV genome, the production of (immediate) early proteins, and the preparation of the viral genome for replication. With regard to the preparation of the viral genome for replication, the viral helicase/primase complex (UL5/UL8/UL52), which unwinds and primes viral DNA, is essential for the ensuing DNA polymerization. To assess its potential involvement in PRV-induced DDR and NF-κB activation, we made use of a potent inhibitor of the helicase/primase complex of alphaherpesviruses, pritelivir (BAY-57-1293) (23–25). As expected, pritelivir strongly suppressed PRV genome replication (Fig. 4B). As described above for PAA, to disentangle the potential effect of pritelivir on NF-κB activation from its effect on expression of the viral late factor(s) that inhibits NF-κB-dependent gene expression, CHX was either added or not to pritelivir-treated cells at 4 hpi. Based on Fig. 4C, it can be concluded that, just like PAA, pritelivir does not inhibit NF-κB activation but does prevent expression of the late viral factor(s) that inhibit NF-κB-driven gene expression. This conclusion indicates that the source of NF-κB activation precedes the activity of the viral helicase/primase. Unfortunately, we were not able to assess earlier events associated with virus genome replication intermediates, in particular a possible involvement of the nuclease activity of UL12, the single-stranded DNA (ssDNA) binding capacity of ICP8, or the role of UL9 binding to the origin of replication (Ori) sites (23, 26), due to the lack of (reliable) tools in PRV with which to conduct these assays.
Alternatively, we investigated the putative role of the two viral serine/threonine protein kinases US3 and UL13, as they are expressed with early kinetics, do not have a direct role in genome replication but are expressed within the infected cell nucleus at early time points, and have catalytic activity that could potentially drive the DDR. In addition, the UL13 homologs of gammaherpesviruses Epstein Barr virus (BGLF4) and gammaherpesvirus-68 (ORF36) have been reported to trigger the DDR (27, 28), and the UL13 homolog of different herpesviruses, including HSV-1, has been shown to activate Tip60, an acetyltransferase involved in ATM activation (28). Both US3 and UL13 are indeed expressed in a PAA-independent way (Fig. 4D). However, infections with PRV mutants lacking US3 or UL13 did not show differences in H2A.X phosphorylation or IκBα degradation at 8 hpi (Fig. 4E). In addition, in the presence of PAA, US3null and UL13null PRV triggered reduced (but still noticeable) H2A.X phosphorylation as observed for WT PRV (data not shown). Hence, both viral kinases can be excluded as triggers of the PRV-induced DDR-NF-κB signaling axis.
Overall, our data show that DDR activation and consequent NF-κB activation are triggered at a prereplicative stage of PRV infection, before the activity of the viral helicase/primase, but require expression of (immediate) early viral proteins different from US3 and UL13.
The ATM kinase is not required for PRV replication in cell culture.
We reported earlier that preventing NF-κB activation during PRV infection does not affect virus protein expression and production of extracellular infectious virus in cell culture (11). Here, we identified the DDR as an upstream event of NF-κB activation during PRV infection. However, in addition to NF-κB activation, the DDR can have different downstream effects that may affect virus replication efficiency. Therefore, we investigated the effect of ATM kinase inhibition on viral genome replication, viral protein production, and infectious virus production. Quantitative PCR analysis of intracellular viral DNA, flow cytometry assessment of late viral protein expression (gB, gD, and VP26-GFP), and virus titer determination revealed that inhibition of ATM activity did not affect viral genome replication, viral protein expression, or infectious virus production (Fig. 5).
Altogether, these results indicate that the DDR-NF-κB signaling axis does not contribute to PRV replication in cell culture. This discovery suggests that triggering the ATM-dependent DDR and the ensuing NF-κB activation by PRV may constitute a host response to the early phases of virus replication rather than a process that is deliberately triggered by the virus for its own benefit.
The DDR-NF-κB signaling axis results in NF-κB-dependent gene expression, which is inhibited in PRV-infected cells.
We showed earlier that PRV-induced NF-κB activation does not result in the expression of NF-κB-dependent genes (11). Since our current data show that NF-κB activation during PRV infection occurs via a noncanonical DDR-dependent mechanism, we wondered whether this lack of NF-κB-dependent gene expression is a natural outcome of the DDR-NF-κB signaling axis (in swine testicle [ST] cells) or whether it indicates that PRV actively inhibits the (proinflammatory) gene expression outcome of this pathway. To address this question, we again made use of the DDR inducer doxorubicin, which triggers activation of the DDR-NF-κB signaling axis in a similar manner as PRV. Like PRV, doxorubicin triggers persistent IκBα degradation until at least 24 h posttreatment (Fig. 2G) and persistent nuclear translocation of NF-κB p65 (Fig. 6A) in ST cells, strongly resembling PRV-induced activation of NF-κB. We used real-time quantitative PCR (RT-qPCR) to assess the expression of different NF-κB-driven genes (IκBα, A20, TNF-α, and IL-6 genes) in doxorubicin-stimulated cells and PRV-infected cells. For PRV-infected cells, we treated the cells or not with the DNA polymerase inhibitor PAA, as our earlier data suggested that PRV may encode PAA-sensitive inhibitor(s) of NF-κB-dependent gene expression. TNF-α-stimulated cells were taken as a positive control for the induction of NF-κB-dependent gene expression. Notably, all four genes included in the analysis showed a transcriptional upregulation in response to doxorubicin versus the mock-treated condition but not in response to PRV infection (Fig. 6B). Importantly, addition of PAA rescued NF-κB-dependent gene expression in PRV-infected cells to levels largely comparable to those observed in doxorubicin-treated cells (Fig. 6B). These data indicate that the DDR-NF-κB signaling axis does lead to productive NF-κB-dependent gene expression but that it is actively inhibited by one or more PAA-sensitive viral factors in PRV-infected cells.
These data also suggest that in doxorubicin-treated cells, the persistent lack of IκBα signal as detected via Western blot (and consequent NF-κB p65 nuclear translocation as detected by confocal microscopy) is a result of continuous degradation of replenished IκBα, whereas in PRV-infected cells, there is no need for continuous degradation of IκBα due to the inhibition of NF-κB-dependent gene expression. To confirm this idea, we performed additional assays in the absence or presence of the 26S proteasome inhibitor MG132. Western blot results showed that proteasome inhibition allowed the recovery of a detectable IκBα protein in doxorubicin-treated cells even after complete IκBα degradation (from 8 to 12 h), whereas in PRV-infected cells, once IκBα reached undetectable levels, it remained undetectable when MG132 was added (Fig. 6C). This finding confirms that in doxorubicin-treated cells, IκBα is expressed and IκBα transcripts undergo successful translation into protein but are instantly degraded, most likely due to incessant ATM activation. In PRV-infected cells, however, PAA-sensitive inhibitor(s) of NF-κB-dependent gene expression prevent replenishment of IκBα. As an alternative explanation, it is possible that NF-κB-dependent gene expression does occur but that the virion host shutoff (vhs) protein of PRV, encoded by the UL41 gene, degrades the resulting transcripts, thereby preventing IκBα replenishment. Using a UL41null PRV strain (PRV ΔUL41) and a UL41rescue strain (PRV ΔUL41R), we found that both viruses trigger IκBα degradation and neither virus leads to IκBα replenishment, even at 16 hpi (Fig. 6D), indicating that vhs/UL41 is not involved in the lack of detectable NF-κB-dependent transcripts in PRV-infected cells.
To narrow down the time window during which the viral inhibitor of NF-κB-dependent gene expression is produced, CHX was added at different time points postinoculation of PAA-treated PRV-infected cells. Figure 6E shows that a considerable amount of replenished IκBα protein could be detected at 8 hpi only when CHX was added from 6 hpi, but not earlier (at 4 or 5 hpi). Since IκBα protein degradation and subsequent p65 nuclear translocation occur between 4 and 6 hpi in PRV-infected cells (Fig. 1A) (11), these data indicate that the viral NF-κB repressor acts immediately after nuclear import of NF-κB, thereby enabling the complete shutoff of NF-κB-dependent gene expression observed during PRV infection (Fig. 6B).
Combined, these data show that the DDR-NF-κB signaling axis leads to NF-κB-dependent gene expression but that this gene expression is actively inhibited by PRV late factor(s).
DISCUSSION
In the current study, we report that pseudorabies virus (PRV) infection of host cells triggers the DNA damage response (DDR) and that this in turn leads to persistent activation of the NF-κB pathway. This DDR-NF-κB signaling axis is initiated at a prereplication stage and depends on the expression of (immediate) early viral genes. Furthermore, PRV very efficiently inhibits NF-κB-dependent gene expression via one or more viral late factor(s) that are expressed relatively early in infection (between 4 hpi and 6 hpi). A schematic representation of the interaction between PRV and the DDR-NF-κB pathway is shown in Fig. 7. Since neither inhibition of the DDR (current report) nor knockdown of NF-κB p65 (11) affects PRV replication in cell culture, these data suggest that activation of the DDR-NF-κB pathway is a host-driven process that is not beneficial for the virus and that PRV encodes one or more viral factor(s) to efficiently impair the (proinflammatory) consequences of this signaling axis.
This is the first report unequivocally connecting the DDR with NF-κB activation in the context of virus infection. Nevertheless, indications in other reports suggest that different viruses may trigger a similar DDR-NF-κB signaling axis. For example, the betaherpesvirus human cytomegalovirus (HCMV) controls interleukin-8 (IL-8) expression through the viral UL76 protein involving ATM and NF-κB activation (29). Another DNA virus, human polyomavirus 2 or John Cunninghamvirus (JCV) promotes nuclear translocation of IKKγ/NEMO for recruitment of activated ATM kinase back to the cytoplasm (30). In addition, paramyxoviruses have been shown to promote an ATM-dependent phosphorylation of the NF-κB subunit p65 at Ser276 which enhances IRF7/RIG-I-dependent IFN production (31). This so-called inside-out ATM-dependent NF-κB activation has been associated with the shuttling of ATM to the cytoplasm through IKKγ/NEMO, although alternative IKK-independent pathways have been described (32–34). The finding that PRV potently triggers this ATM-dependent NF-κB pathway may therefore represent an interesting model to explore this still poorly understood mode of NF-κB activation further mechanistically.
This report also reveals ATM-dependent DDR activation during PRV infection, which is something that was reported previously for the alphaherpesviruses HSV-1 and HSV-2 (35, 36). Interestingly, other DDR pathways controlled by ATR and DNA-dependent protein kinase, catalytic subunit (DNA-PKcs) kinases seem to be inhibited by the HSV-1 ICP0 protein. More concretely, ICP0 is involved in the mislocalization of the ATR interacting protein (ATRIP) and in DNA-PKcs degradation (37, 38), suggesting that in agreement with the current observations in PRV infection, ATM is the main driver of the DDR to alphaherpesvirus infections. It has been proposed that the DDR to HSV enables the recruitment of host cell DNA repair factors to virus replication factories to enhance virus replication (35, 39, 40). In contrast, some DNA repair proteins activated by the DDR, such as RNF8 and RNF168, are recruited very early in infection to incoming genomes to limit virus replication, a host antiviral mechanism counteracted by the ICP0 protein (41, 42). Up to date, there is no clear consensus about the possible implications of the DDR on HSV-1 replication fitness and different hypotheses have been proposed (35, 36, 39, 40, 43–46). Our current data on PRV infection indicate that inactivation of ATM kinase does not reduce the viral replication capacity (e.g., viral genome production, late protein expression, and extracellular virus titers), at least in cell culture.
Although the exact mechanism of DDR-NF-κB activation during PRV infection is unclear, our data indicate that the trigger takes place before the activities of the helicase/primase complex and DNA polymerase. These observations fit with what was previously described in HSV-1 infection where the MRN complex that plays a crucial role in ATM-dependent DDR activation upon sensing dsDNA breaks interacts with the HSV-1 UL12 exonuclease (26). UL12 and ICP8 are proposed as a two-subunit recombinase that participates in the primary initiation of alphaherpesvirus genome replication before the activity of the helicase/primase. Future research will reveal whether the activity of PRV UL12 and ICP8 are involved in PRV-induced DDR-NF-κB activation.
DDR-dependent NF-κB activation by PRV infection takes place very early in infection before viral genome replication, which is a phenotype consistent with what has been reported for HSV-1 infection (45, 46). Amici et al. proposed a biphasic induction of the NF-κB pathway consisting of a first wave caused immediately after infection, which probably does not involve viral gene expression, and a second and more powerful wave of activation that requires active virus replication in infected cells (47). Structural virion components have been reported to play a role in NF-κB activation immediately after infection, such as the viral glycoproteins gB, gD, gH, and gL (48, 49) and the tegument protein UL37 (50). In PRV infection, using IκBα degradation as a readout, we identified only one wave of activation that requires viral protein expression, although an earlier wave associated with PRV entry that may not lead to substantial IκBα degradation cannot be formally excluded. Expression of the immediately early proteins ICP4 and ICP27 has been reported to play a crucial role in HSV-1-dependent NF-κB activation (51). Particularly, the N-terminal region of HSV-1 ICP27 has been reported to trigger NF-κB activation via the Jun N-terminal protein kinase (JNK) (52). In PRV infection, viral protein expression until at least 2 hpi is not sufficient to trigger NF-κB, which suggests that the expression of the sole immediate early PRV protein IE180 (a homolog of HSV-1 ICP4) does not cause NF-κB activation. Unfortunately, we could not draw meaningful conclusions from infection of cells with a PRV mutant lacking UL54 expression (the homolog of HSV-1 ICP27) given that infection was massively delayed compared with PRV WT infection (data not shown). The defective replication of PRV ΔUL54 is supported by the complete dysregulation of the viral gene transcription program observed for this mutant (53). In general, removal of viral regulators of viral gene expression will cause an impaired expression of other early proteins (e.g., those involved in viral genome replication) complicating interpretation of the data. Alternatively, Taddeo et al., showed that HSV-1 activates the NF-κB pathway through the protein kinase R (PKR) (54). In conclusion, the time window of NF-κB activation for HSV-1 and PRV seems to correlate, but this report for the first time connects the activation of the NF-κB pathway with triggering of the ATM-dependent DDR during alphaherpesvirus infection.
PRV-induced NF-κB activation is aberrant, consisting of a persistent and irreversible IκBα degradation and nuclear localization of NF-κB (11), which is similar to reports for HSV-1 (47, 51). This persistent NF-κB activation caused by both viruses relies on the failure of autoregulatory mechanisms involved in the repression of the pathway upon activation (e.g., IκBα replenishment) and is achieved by inhibition of the transcriptional activity of NF-κB (current report, 11, 51). Curiously, the closely related alphaherpesvirus varicella zoster virus (VZV) inhibits the NF-κB pathway via a different strategy, based on the persistent stabilization of the IκBα protein in the cytoplasm of infected cells, accompanied by a retention of NF-κB in the cytoplasm after a transient nuclear translocation (55–57). The results shown in this report indicate that NF-κB-dependent gene expression can be released by the addition of PAA, indicating that a late factor(s) encoded by PRV is actively repressing NF-κB immediately upon its nuclear translocation. The HSV-1-dependent inhibition of NF-κB transcription activity might be distinct from that operating in PRV infection, given that PAA treatment does not cause IκBα replenishment during HSV-1 infection (52). It has been proposed that HSV-1-dependent shutoff of NF-κB-dependent gene expression is achieved through the active relocalization of NF-κB molecules from κB binding sites on the host genome to viral gene promoters (47, 58). Nevertheless, some cellular and viral events in HSV-1 infection, such as apoptosis and virus egress, have still been associated with NF-κB transcriptional activity in infected cells (59–61). In any way, independently of the mechanism(s) used, alphaherpesviruses effectively evade NF-κB-dependent gene expression in infected cells, suggesting that productive NF-κB pathway activation may represent a strong barrier toward efficient virus replication and spread, especially in vivo.
The fact that persistent ATM-dependent DDR, e.g., as caused by doxorubicin, drives continuous IκBα degradation, including both the pre-existing and the replenished pool of IκBα, leads us to hypothesize that the ATM-dependent NF-κB pathway is an unstoppable signaling stream that effectively overcomes the negative regulatory mechanisms that are meant to repress the NF-κB pathway upon initial activation. This idea is unusual, as these self-regulated inhibitory strategies of NF-κB are functional when NF-κB is activated via the canonical pathway (11, 12). The irreversible and powerful DDR activation elicited throughout the PRV lytic infection likely triggers a similar incessant ATM-dependent NF-κB activation. This activation might explain why PRV may have evolved a mechanism to generally shut down NF-κB-dependent gene expression, since alternative strategies to interfere with NF-κB activation, such as the overexpression/activation of physiological repressors of the pathway, likely would be unable to prevent ATM-dependent persistent NF-κB activation.
In conclusion, the data provided in the current study show that PRV-induced NF-κB activation depends on a peculiar ATM-dependent DDR pathway involved in the maintenance of cellular genome integrity and regulation of cell survival upon DNA injury. Our data suggest that DDR activation by PRV may be part of the cellular response to infection and does not provide an evident benefit for the virus, at least in cell culture, which may explain our observation that PRV potently suppresses subsequent NF-κB-dependent gene expression. Altogether, the interaction of PRV with the DDR-NF-κB signaling axis indicates a viral subversion of the very potent NF-κB signaling axis in the early control of alphaherpesvirus infections by the host.
MATERIALS AND METHODS
Cell cultures and viruses.
Swine testicle (ST) cells (ATCC CRL-1746) were cultured in modified Eagle’s medium (MEM) supplemented with 10% inactivated fetal bovine serum (FBS), 100 U/ml penicillin, 0.1 mg/ml streptomycin, 50 μg/ml gentamicin, and 1 mM sodium pyruvate (all from Gibco, Thermo Fisher Scientific) (ST medium).
The wild-type (WT) PRV strains used in the present study were PRV WT Becker (62), PRV WT Kaplan (63), and PRV WT NIA3 (64). The PRV-GS443 strain, a kind gift from G. A. Smith (Northwestern University, USA) was generated from the PRV WT Becker backbone by fusing the green fluorescent protein (GFP) open reading frame to the PRV UL35 gene, resulting in the expression of GFP-tagged PRV capsid protein VP26 (VP26-GFP) (15). The PRV mutant viruses PRV ΔUS3 NIA (M118) (65) and PRV ΔUL13 NIA (M137) (66), which lacked either US3 or UL13 expression, respectively, were a kind gift from the ID-DLO Institute (Lelystad, The Netherlands). PRV ΔUL41 Kaplan and PRV ΔUL41 rescue Kaplan viruses (67) were kindly provided by T. C. Mettenleiter. Infections with wild-type PRV strains PRV ΔUL41 and ΔUL41 rescue viruses and PRV-GS443 were all performed at a multiplicity of infection (MOI) of 10 PFUs per cell. Infections with PRV ΔUS3 and PRV ΔUL13 viruses were carried out using an MOI of 20 PFU/cell. PRV infections were always performed on confluent cell monolayers.
Chemicals and cytokines.
The viral DNA polymerase inhibitor phosphonoacetic acid (PAA) was purchased from Sigma-Aldrich (catalog number 284270). The alphaherpesvirus helicase/primase inhibitor pritelivir (BAY-57-1293) was obtained from Selleckchem (catalog number S7546). Cycloheximide (CHX) was purchased from Sigma (catalog number C1988). The 26S proteasome inhibitor MG132 was purchased from Sigma-Aldrich (catalog number M7449). The ATM kinase inhibitor KU60019 was obtained from Selleckchem (catalog number S1570). Doxorubicin hydrochloride was purchased from Thermo Fisher Scientific (catalog number BP25161). Purified recombinant porcine TNF-α was obtained from R&D System (catalog number 690-T).
Antibodies targeting PRV proteins.
The mouse monoclonal antibodies used were directed against gB (clone 1C11; 1:100 for Western blotting (WB), 1:50 for immunofluorescence (IF), and 1:20 for FC), gD (clone 13D12; 1:100 for WB and 1:20 for FC), gE (clone 18E8; 1:100 for WB) (68), US3 (1:100 for WB, 1:50 for IF) (69), and VP5 (clone 3C10; 1:2,000 for WB) (70). Anti-IE180 (1:1,000 for WB) (71), anti-UL13 (1:1,000 for WB) (72), and anti-UL41/vhs (1:10,000 for WB) antibodies were rabbit polyclonal antisera. Antibodies against US3, VP5, and UL13 proteins were generous gifts from Lynn Enquist (Princeton University, NJ, USA). Anti-IE180 antiserum was kindly provided by Enrique Tabarés (Autónoma University of Madrid, Madrid, Spain). The antibody used for detection of UL41/vhs was a kind gift from Thomas C. Mettenleiter (Friedrich-Loeffler Insititute, Insel-Reims, Germany).
Antibodies targeting cellular proteins.
The antibodies used for the detection of proteins associated with NF-κB and the DNA damage response pathways were all purchased from Cell Signaling Technology (CST) and included mouse anti-IκBα (clone L35A5; 1:1,000 for WB; catalog number 4814), mouse anti-NF-κB p65 (clone L8F6; 1:1,000 for WB, 1:400 for IF; catalog number 6956), rabbit anti-H2A.X (clone D17A3; 1:1,000 for WB, 1:50 for IF; catalog number 7631), and rabbit anti-phospho-H2A.X Ser139 (1:1,000 for WB and 1:500 for IF; catalog number 2577). The rabbit anti-α-tubulin antibody was purchased from Abcam (1:2,000 for WB; catalog number ab15246).
Western blot analysis.
Cell lysates were collected in ice-cold 1× RIPA buffer (made from 10× radioimmunoprecipitation assay (RIPA) buffer, catalog number ab156034, Abcam) containing a protease inhibitor cocktail (cOmplete mini EDTA free; catalog number 11836170001; Roche). A phosphatase inhibitor cocktail (PhosSTOP; catalog number 04906845001; Roche) was added to the lysis buffer only when the detection of phosphorylated residues was performed. Lysates were incubated at 4°C for 20 min prior to storage at −20°C. SDS-PAGE and Western blotting procedures were described in detail in reference 73. A total of 5% (wt/vol) nonfat dry milk diluted in 0.1% PBS-Tween 20 (PBS-T) was typically used as the blocking solution and then incubated for 1 h at room temperature (RT). However, the blocking buffer used for the detection of phospho-proteins, as well as for detection with certain commercial antibodies, consisted of 5% (wt/vol) bovine serum albumin (BSA; catalog number 160069; MP Biomedicals) diluted in PBS-T and incubated for 1 h at RT. Blots were incubated overnight at 4°C with primary antibodies diluted in the corresponding blocking buffer. After primary antibody incubation, blots were washed three times with PBS-T (10 min each). Incubation with horseradish peroxidase (HRP)-conjugated secondary antibodies, either with goat anti-IgG mouse-HRP (1:2,,000; catalog number P0447; Dako) or with goat anti-IgG rabbit-HRP (1:3000; catalog number P0448; Dako) was performed for 1 h at room temperature. After three washing steps with PBS-T, protein bands were detected by chemiluminescence using a ChemiDoc imaging device (Bio-Rad). The Pierce enhanced chemiluminescence (ECL) substrate (Thermo Scientific), ECL Plus substrate (GE Health Care), or SuperSignal West Femto maximum sensitivity substrate (Thermo Scientific) were used for protein band detection according to protein abundance in the cell lysate and antibody sensitivity.
Immunofluorescence assays.
Cell monolayers were rinsed once with sterile PBS (supplemented with calcium and magnesium; complete PBS) and then fixed with 4% paraformaldehyde for 15 min at room temperature. Fixed cells were incubated for 1 h at 37°C with blocking/permeabilizing buffer (0.3% Triton X-100 and 5% inactivated FBS diluted in PBS). Later, primary antibodies diluted in incubation buffer (0.3% Triton X-100 and 1% [wt/vol] BSA diluted in PBS) were incubated overnight at 4°C. After three washing steps with PBS, cell monolayers were incubated for 1 h at 37°C with fluorochrome-conjugated goat anti-IgG mouse secondary antibodies (1:200; Invitrogen, Thermo Fisher Scientific). Finally, cells were incubated with Hoechst 33342 (1:200; catalog number H3570; Invitrogen, Thermo Fisher Scientific) diluted in PBS for 10 min at room temperature for cell nuclei counterstaining. All fluorescence images were taken using a Leica SPE confocal microscope (Leica) or a Leica Thunder fluorescence microscope (Leica). Fluorescent microscopy images were analyzed using ImageJ imaging software (NIH, USA).
Flow cytometry.
Cells were detached by incubation for 5 min at 37°C in an undiluted Accutase protease solution (StemPro Accutase cell dissociation reagent; catalog number A1110501; Thermo Fisher Scientific). Protease activity was inhibited by adding inactivated FBS up to 15%. Cells were fixed and permeabilized using a Cytofix/Cytoperm solution (catalog 554714BD; BD Biosciences) at 4°C for 20 min, and then fixed cells were washed with 1× BD Perm/Wash buffer (catalog number 554714; BD Biosciences). Primary and secondary antibodies were diluted in 1× BD Perm/Wash buffer (100 μl of antibody solution per 1 million cells) and incubated at 4°C for 40 min. Primary anti-gB and anti-gD antibodies were used in a 1:20 dilution, and the allophycocyanin (APC)-conjugated goat anti-IgG mouse secondary antibody was used in a 1:200 dilution (Invitrogen; Thermo Fisher Scientific). A total of 10,000 cells were analyzed per sample, using a NovoCyte flow cytometer (ACEA Biosciences). NovoExpress software was used for flow cytometry data analysis and representation.
DNA isolation for intracellular PRV genome quantification.
Total DNA was isolated from c.a. 2 million cells. First, cell monolayers were washed twice with sterile complete PBS to remove the vast majority of extracellular virus. In addition, cell monolayers were treated with sodium citrate buffer (pH 3.0; 40 mM sodium citrate, 10 mM KCl, and 135 mM NaCl) for 2 min at room temperature. After a sodium citrate buffer incubation, cells were rinsed three times with complete PBS before cells were harvested. Cell pellets were frozen at −20°C before the DNA isolation was started. DNA isolation was carried out using the DNeasy blood and tissue kit (catalog number 69504; Qiagen) according to the manufacturer’s protocol. DNA yields were diluted in 200 μl, from which 5 μl was used as the template for real-time quantitative PCR (RT-qPCR).
RNA isolation and reverse transcription for mRNA quantification.
Total RNA was isolated using the RNeasy mini kit (catalog number 74104; Qiagen) by following instructions provided by the manufacturer. RNA yields were subjected to DNase I treatment (RNase free; catalog number M0303S; New England BioLabs) for 10 min at 37°C. DNase I reaction was inhibited by addition of EDTA (up to 5 mM EDTA; catalog number AM9260G; Invitrogen) and by incubating samples at 75°C for 10 min. A total of 500 ng of DNA-free RNA was used as the substrate for reverse transcription (RT) using the iScript cDNA synthesis kit (catalog number 1708891; Bio-Rad) according to the manufacturer’s procedure. The one-step RT reaction was set by 5 min at 25°C (priming), 20 min at 46°C (reverse transcription), and 1 min at 95°C (RT inhibition). The RT reaction was carried out in a total volume of 20 μl, from which 1 μl was used as the template for RT-qPCR analysis.
Quantitative PCR (qPCR).
Primer DNA oligonucleotides used for qPCR were synthesized by Integrative DNA Technologies (IDT). When primer sequences were not obtained from the literature, Primer-BLAST software (NIH, USA) was used for primer design. qPCR was performed using SYBR green PCR master mix (catalog number 4309155; Thermo Fisher Scientific). qPCRs were performed in a final volume of 20 μl on MicroAmp Fast optical 96-well reaction plates (catalog number 4346906; Thermo Fisher Scientific) using a StepOnePlus real-time PCR system (catalog number 4376600; Thermo Fisher Scientific). Every sample was measured in duplicate. qPCR data were analyzed by the double delta threshold cycle method.
Intracellular PRV genome quantification.
PRV genomes were quantified by amplification of sequences belonging to two different PRV open reading frame sequences, namely, us3 and ul27, obtained from the PRV reference sequence (Suid herpesvirus 1, complete genome; GenBank accession number NC_006151.1). Cellular genome quantification targeting the porcine beta-2-microglobulin (B2M) gene (Chromosome 1, Sus scrofa isolate TJ Tabasco breed Duroc, NC_010443.5) allowed normalization of viral genome quantification. Primer sequences for intracellular PRV genome quantification assays can be found in Table 1.
TABLE 1.
Target gene | GenBank accession no. | Sequence of (5′–3′): |
|
---|---|---|---|
Forward primer | Reverse primer | ||
PRV UL27 | NC_006151.1 | GTC YGT GAA GCG GTT CGT GAT | ACA AGT TCA AGG CCC ACA TCT AC |
PRV US3 | NC_006151.1 | GAC GGG GGC TTT CCT GAT TTA | GTA TCT CAT CAG CGG AAG GGC |
B2M | NC_010443.5 | AAA CGG AAA GCC AAA TTA CC | ATC CAC AGC GTT AGG AGT GA |
NF-κB-dependent gene expression.
The relative expression of the porcine (Sus scrofa) target genes TNFA (encoding TNF-α), IL-6 (encoding IL-6), NFKBIA (encoding IκBα), and TNFAIP3 (encoding A20) was normalized to the level of expression 28S rRNA gene expression. Primer sequences used for this analysis are described in Table 2.
TABLE 2.
Target gene | GenBank accession no. | Reference | Sequence of (5′–3′): |
|
---|---|---|---|---|
Forward primer (5′–3′) | Reverse primer (5′–3′) | |||
TNFA (encoding TNF-α) | NM_214022.1 | 74 | ACT GCA CTT CGA GGT TAT CGG | GGC GAC GGG CTT ATC TGA |
IL6 (encoding IL-6) | NM_214399.1 | 75 | TTC ACC TCT CCG GAC AAA ACT | TCT GCC ATG ACC TCC TTG CTG T |
NFKBIA (encoding IκBα) | NM_001005150.1 | 11 | AAG CAC TCG GAT ACA GCA GC | AGT CGT CAT AGG GCA GCT CA |
TNFIAP3 (encoding A20) | NM_001267890.1 | 11 | CCT GTT CAG CGA GAC TAC GG | 5′-AAC GTC CTG GTG ACG TTC TG-3′ |
28 rRNA | 76 | GGG CCG AAA CGA TCT CAA CC | GCC GGG CTT ACC CAT T |
Extracellular virus titer determination.
Confluent cell monolayers seeded on 24-well plates were infected at a multiplicity of infection of 10 PFU/cell. At 2 hpi, cell monolayers were rinsed twice with complete PBS and incubated with sodium citrate buffer (pH 3.0; 40 mM sodium citrate, 10 mM KCl, and 135 mM NaCl) for 2 min at room temperature to inactivate remaining virus inoculum. Two additional washing steps with complete PBS were carried out before adding fresh cell culture medium. At the indicated time points, PRV-infected cell supernatants were collected and immediately frozen at −80°C. Extracellular titers were determined by infection of confluent ST cell monolayers seeded in 96-well plates with 10-fold serial dilution of the PRV-infected cell supernatants, using four experimental repeats. The PRV-associated cytopathic effect was used as a readout for infection. Extracellular virus titers were calculated in PFU per milliliter (PFU/ml).
Quantification of cell nuclei areas.
Areas of cell nuclei were determined using ImageJ imaging software (NIH, USA) by contour determination of Hoechst-positive structures (corresponding to nuclear DNA) from confocal microscopy pictures taken with a 63× objective. Nuclear areas of 20 cells were determined per sample, and 3 independent repeats were performed.
ACKNOWLEDGMENTS
N.R. is supported by a Ph.D. grant from Research Foundation Flanders (F.W.O.-Vlaanderen, grant number FWO.SPB.2019.0043.01). This research was supported by grants from the F.W.O.-Vlaanderen (grant numbers G017615 and G.019617N) and the Special Research Fund of Ghent University (G.O.A. grant 01G01317 and grant BOFBAS2018000301)
We thank Thomas Mettenleiter (Friedrich-Loeffler Institute, Germany), Lynn Enquist (Princeton University, USA), Greg Smith (Northwestern University, USA), and the ID-DLO Institute (The Netherlands) for virus strains. We thank our colleague Cliff Van Waesberghe for the excellent technical assistance provided throughout the development of this study.
We declare no conflict of interest.
N.R. and H.W.F. designed the research. N.R. performed the experiments. N.R. and H.W.F. analyzed the results, interpreted the data, made the figures, and wrote the manuscript.
Contributor Information
Herman W. Favoreel, Email: herman.favoreel@ugent.be.
Rozanne M. Sandri-Goldin, University of California, Irvine
REFERENCES
- 1.Mettenleiter TC. 2000. Aujeszky’s disease (pseudorabies) virus: the virus and molecular pathogenesis–state of the art, June 1999. Vet Res 31:99–115. 10.1051/vetres:2000110. [DOI] [PubMed] [Google Scholar]
- 2.Roizmann B, Desrosiers RC, Fleckenstein B, Lopez C, Minson AC, Studdert MJ. 1992. The family Herpesviridae: an update. Arch Virol 123:425–449. 10.1007/BF01317276. [DOI] [PubMed] [Google Scholar]
- 3.Pomeranz LE, Reynolds AE, Hengartner CJ. 2005. Molecular biology of pseudorabies virus: impact on neurovirology and veterinary medicine. Microbiol Mol Biol Rev 69:462–500. 10.1128/MMBR.69.3.462-500.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Hayden MS, West AP, Ghosh S. 2006. NF-kappaB and the immune response. Oncogene 25:6758–6780. 10.1038/sj.onc.1209943. [DOI] [PubMed] [Google Scholar]
- 5.Zhang Q, Lenardo MJ, Baltimore D. 2017. 30 Years of NF-κB: a blossoming of relevance to human pathobiology. Cell 168:37–57. 10.1016/j.cell.2016.12.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Gilmore T, Gapuzan ME, Kalaitzidis D, Starczynowski D. 2002. Rel/NF-kappa B/I kappa B signal transduction in the generation and treatment of human cancer. Cancer Let 181:1–9. 10.1016/S0304-3835(01)00795-9. [DOI] [PubMed] [Google Scholar]
- 7.Valen G, Yan ZQ, Hansson GK. 2001. Nuclear factor kappa-B and the heart. J Am Coll Cardiol 38:307–314. 10.1016/S0735-1097(01)01377-8. [DOI] [PubMed] [Google Scholar]
- 8.Hayashi T, Faustman D. 2000. Defective function of the proteasome in autoimmunity: involvement of impaired NF-kappaB activation. Diabetes Technol Ther 2:415–428. 10.1089/15209150050194288. [DOI] [PubMed] [Google Scholar]
- 9.Hayden MS, Ghosh S. 2008. Shared principles in NF-kappaB signaling. Cell 132:344–362. 10.1016/j.cell.2008.01.020. [DOI] [PubMed] [Google Scholar]
- 10.Gilmore TD. 2006. Introduction to NF-kappaB: players, pathways, perspectives. Oncogene 25:6680–6684. 10.1038/sj.onc.1209954. [DOI] [PubMed] [Google Scholar]
- 11.Romero N, Van Waesberghe C, Favoreel HW. 2020. Pseudorabies virus infection of epithelial cells leads to persistent but aberrant activation of the NF-κB pathway, inhibiting hallmark NF-κB-induced proinflammatory gene expression. J Virol 94:e00196-20. 10.1128/JVI.00196-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Brown K, Park S, Kanno T, Franzoso G, Siebenlist U. 1993. Mutual regulation of the transcriptional activator NF-kappa B and its inhibitor, I kappa B-alpha. Proc Natl Acad Sci USA 90:2532–2536. 10.1073/pnas.90.6.2532. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.de Martin R, Vanhove B, Cheng Q, Hofer E, Csizmadia V, Winkler H, Bach FH. 1993. Cytokine-inducible expression in endothelial cells of an I kappa B alpha-like gene is regulated by NF kappa B. EMBO J 12:2773–2779. 10.1002/j.1460-2075.1993.tb05938.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Le Bail O, Schmidt-Ullrich R, Israël A. 1993. Promoter analysis of the gene encoding the I kappa B-alpha/MAD3 inhibitor of NF-kappa B: positive regulation by members of the rel/NF-kappa B family. EMBO J 12:5043–5049. 10.1002/j.1460-2075.1993.tb06197.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Smith GA, Gross SP, Enquist LW. 2001. Herpesviruses use bidirectional fast-axonal transport to spread in sensory neurons. Proc Natl Acad Sci USA 98:3466–3470. 10.1073/pnas.061029798. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Janssens S, Tschopp J. 2006. Signals from within: the DNA-damage-induced NF-kappaB response. Cell Death Differ 13:773–784. 10.1038/sj.cdd.4401843. [DOI] [PubMed] [Google Scholar]
- 17.Shiloh Y, Ziv Y. 2013. The ATM protein kinase: regulating the cellular response to genotoxic stress, and more. Nat Rev Mol Cell Biol 14:197–210. 10.1038/nrm3546. [DOI] [PubMed] [Google Scholar]
- 18.Burma S, Chen BP, Murphy M, Kurimasa A, Chen DJ. 2001. ATM phosphorylates histone H2AX in response to DNA double-strand breaks. J Biol Chem 276:42462–42467. 10.1074/jbc.C100466200. [DOI] [PubMed] [Google Scholar]
- 19.Turinetto V, Giachino C. 2015. Multiple facets of histone variant H2AX: a DNA double-strand-break marker with several biological functions. Nucleic Acids Res 43:2489–2498. 10.1093/nar/gkv061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Ho WC, Dickson KM, Barker PA. 2005. Nuclear factor-kappaB induced by doxorubicin is deficient in phosphorylation and acetylation and represses nuclear factor-kappaB-dependent transcription in cancer cells. Cancer Res 65:4273–4281. 10.1158/0008-5472.CAN-04-3494. [DOI] [PubMed] [Google Scholar]
- 21.Golding SE, Rosenberg E, Valerie N, Hussaini I, Frigerio M, Cockcroft XF, Chong WY, Hummersone M, Rigoreau L, Menear KA, O'Connor MJ, Povirk LF, van Meter T, Valerie K. 2009. Improved ATM kinase inhibitor KU-60019 radiosensitizes glioma cells, compromises insulin, AKT and ERK prosurvival signaling, and inhibits migration and invasion. Mol Cancer Ther 8:2894–2902. 10.1158/1535-7163.MCT-09-0519. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Jin MH, Oh DY. 2019. ATM in DNA repair in cancer. Pharmacol Ther 203:107391. 10.1016/j.pharmthera.2019.07.002. [DOI] [PubMed] [Google Scholar]
- 23.Weller SK, Coen DM. 2012. Herpes simplex viruses: mechanisms of DNA replication. Cold Spring Harb Perspect Biol 4:a013011. 10.1101/cshperspect.a013011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Field HJ, Biswas S. 2011. Antiviral drug resistance and helicase-primase inhibitors of herpes simplex virus. Drug Resist Updat 14:45–51. 10.1016/j.drup.2010.11.002. [DOI] [PubMed] [Google Scholar]
- 25.Wald A, Corey L, Timmler B, Magaret A, Warren T, Tyring S, Johnston C, Kriesel J, Fife K, Galitz L, Stoelben S, Huang ML, Selke S, Stobernack HP, Ruebsamen-Schaeff H, Birkmann A. 2014. Helicase-primase inhibitor pritelivir for HSV-2 infection. N Engl J Med 370:201–210. 10.1056/NEJMoa1301150. [DOI] [PubMed] [Google Scholar]
- 26.Balasubramanian N, Bai P, Buchek G, Korza G, Weller SK. 2010. Physical interaction between the herpes simplex virus type 1 exonuclease, UL12, and the DNA double-strand break-sensing MRN complex. J Virol 84:12504–12514. 10.1128/JVI.01506-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Tarakanova VL, Leung-Pineda V, Hwang S, Yang C, Matatall CW, Basson M, Sun R, Piwnica-Worms H, Sleckman BP, Virgin HW. 2007. Gamma-herpesvirus kinase actively initiates a DNA damage response by inducing phosphorylation of H2AX to foster viral replication. Cell Host Microbe 1:275–286. 10.1016/j.chom.2007.05.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Li R, Zhu J, Xie Z, Liao G, Liu J, Chen MR, Hu S, Woodard C, Lin J, Taverna SD, Desai P, Ambinder RF, Hayward GS, Qian J, Zhu H, Hayward SD. 2011. Conserved herpesvirus kinases target the DNA damage response pathway and TIP60 histone acetyltransferase to promote virus replication. Cell Host Microbe 10:390–400. 10.1016/j.chom.2011.08.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Costa H, Nascimento R, Sinclair J, Parkhouse RM. 2013. Human cytomegalovirus gene UL76 induces IL-8 expression through activation of the DNA damage response. PLoS Pathog 9:e1003609. 10.1371/journal.ppat.1003609. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.White MK, Bellizzi A, Ibba G, Pietropaolo V, Palamara AT, Wollebo HS. 2017. The DNA damage response promotes polyomavirus JC infection by nucleus to cytoplasm NF- kappaB activation. Virol J 14:31. 10.1186/s12985-017-0707-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Fang L, Choudhary S, Tian B, Boldogh I, Yang C, Ivanciuc T, Ma Y, Garofalo RP, Brasier AR. 2015. Ataxia telangiectasia mutated kinase mediates NF-κB serine 276 phosphorylation and interferon expression via the IRF7-RIG-I amplification loop in paramyxovirus infection. J Virol 89:2628–2642. 10.1128/JVI.02458-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Huang TT, Wuerzberger-Davis SM, Seufzer BJ, Shumway SD, Kurama T, Boothman DA, Miyamoto S. 2000. NF-kappaB activation by camptothecin. A linkage between nuclear DNA damage and cytoplasmic signaling events. J Biol Chem 275:9501–9509. 10.1074/jbc.275.13.9501. [DOI] [PubMed] [Google Scholar]
- 33.Wu ZH, Shi Y, Tibbetts RS, Miyamoto S. 2006. Molecular linkage between the kinase ATM and NF-kappaB signaling in response to genotoxic stimuli. Science 311:1141–1146. 10.1126/science.1121513. [DOI] [PubMed] [Google Scholar]
- 34.Miyamoto S. 2011. Nuclear initiated NF-κB signaling: NEMO and ATM take center stage. Cell Res 21:116–130. 10.1038/cr.2010.179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Lilley CE, Carson CT, Muotri AR, Gage FH, Weitzman MD. 2005. DNA repair proteins affect the lifecycle of herpes simplex virus 1. Proc Natl Acad Sci USA 102:5844–5849. 10.1073/pnas.0501916102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Shirata N, Kudoh A, Daikoku T, Tatsumi Y, Fujita M, Kiyono T, Sugaya Y, Isomura H, Ishizaki K, Tsurumi T. 2005. Activation of ataxia telangiectasia-mutated DNA damage checkpoint signal transduction elicited by herpes simplex virus infection. J Biol Chem 280:30336–30341. 10.1074/jbc.M500976200. [DOI] [PubMed] [Google Scholar]
- 37.Wilkinson DE, Weller SK. 2006. Herpes simplex virus type I disrupts the ATR-dependent DNA-damage response during lytic infection. J Cell Sci 119:2695–2703. 10.1242/jcs.02981. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Parkinson J, Lees-Miller SP, Everett RD. 1999. Herpes simplex virus type 1 immediate-early protein vmw110 induces the proteasome-dependent degradation of the catalytic subunit of DNA-dependent protein kinase. J Virol 73:650–657. 10.1128/JVI.73.1.650-657.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Wilkinson DE, Weller SK. 2004. Recruitment of cellular recombination and repair proteins to sites of herpes simplex virus type 1 DNA replication is dependent on the composition of viral proteins within prereplicative sites and correlates with the induction of the DNA damage response. J Virol 78:4783–4796. 10.1128/jvi.78.9.4783-4796.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Taylor TJ, Knipe DM. 2004. Proteomics of herpes simplex virus replication compartments: association of cellular DNA replication, repair, recombination, and chromatin remodeling proteins with ICP8. J Virol 78:5856–5866. 10.1128/JVI.78.11.5856-5866.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Lilley CE, Chaurushiya MS, Boutell C, Landry S, Suh J, Panier S, Everett RD, Stewart GS, Durocher D, Weitzman MD. 2010. A viral E3 ligase targets RNF8 and RNF168 to control histone ubiquitination and DNA damage responses. EMBO J 29:943–955. 10.1038/emboj.2009.400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Lilley CE, Chaurushiya MS, Boutell C, Everett RD, Weitzman MD. 2011. The intrinsic antiviral defense to incoming HSV-1 genomes includes specific DNA repair proteins and is counteracted by the viral protein ICP0. PLoS Pathog 7:e1002084. 10.1371/journal.ppat.1002084. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Mertens ME, Knipe DM. 2021. Herpes simplex virus 1 manipulates host cell antiviral and proviral DNA damage responses. mBio 12:e03552-20. 10.1128/mBio.03552-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Edwards TG, Bloom DC, Fisher C. 2018. The ATM and Rad3-related (ATR) protein kinase pathway is activated by herpes simplex virus 1 and required for efficient viral replication. J Virol 92:e01884-17. 10.1128/JVI.01884-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Alekseev O, Donegan WE, Donovan KR, Limonnik V, Azizkhan-Clifford J. 2020. HSV-1 hijacks the host DNA damage response in corneal epithelial cells through ICP4-mediated activation of ATM. Invest Ophthalmol Vis Sci 61:39. 10.1167/iovs.61.6.39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Gregory DA, Bachenheimer SL. 2008. Characterization of mre11 loss following HSV-1 infection. Virology 373:124–136. 10.1016/j.virol.2007.12.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Amici C, Rossi A, Costanzo A, Ciafrè S, Marinari B, Balsamo M, Levrero M, Santoro MG. 2006. Herpes simplex virus disrupts NF-kappaB regulation by blocking its recruitment on the IkappaBalpha promoter and directing the factor on viral genes. J Biol Chem 281:7110–7117. 10.1074/jbc.M512366200. [DOI] [PubMed] [Google Scholar]
- 48.Leoni V, Gianni T, Salvioli S, Campadelli-Fiume G. 2012. Herpes simplex virus glycoproteins gH/gL and gB bind Toll-like receptor 2, and soluble gH/gL is sufficient to activate NF-κB. J Virol 86:6555–6562. 10.1128/JVI.00295-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Sciortino MT, Medici MA, Marino-Merlo F, Zaccaria D, Giuffrè-Cuculletto M, Venuti A, Grelli S, Mastino A. 2008. Involvement of HVEM receptor in activation of nuclear factor kappaB by herpes simplex virus 1 glycoprotein D. Cell Microbiol 10:2297–2311. 10.1111/j.1462-5822.2008.01212.x. [DOI] [PubMed] [Google Scholar]
- 50.Liu X, Fitzgerald K, Kurt-Jones E, Finberg R, Knipe DM. 2008. Herpesvirus tegument protein activates NF-kappaB signaling through the TRAF6 adaptor protein. Proc Natl Acad Sci USA 105:11335–11339. 10.1073/pnas.0801617105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Patel A, Hanson J, McLean TI, Olgiate J, Hilton M, Miller WE, Bachenheimer SL. 1998. Herpes simplex type 1 induction of persistent NF-kappa B nuclear translocation increases the efficiency of virus replication. Virology 247:212–222. 10.1006/viro.1998.9243. [DOI] [PubMed] [Google Scholar]
- 52.Hargett D, Rice S, Bachenheimer SL. 2006. Herpes simplex virus type 1 ICP27-dependent activation of NF-kappaB. J Virol 80:10565–10578. 10.1128/JVI.01119-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Csabai Z, Takács IF, Snyder M, Boldogkői Z, Tombácz D. 2017. Evaluation of the impact of ul54 gene-deletion on the global transcription and DNA replication of pseudorabies virus. Arch Virol 162:2679–2694. 10.1007/s00705-017-3420-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Taddeo B, Luo TR, Zhang W, Roizman B. 2003. Activation of NF-kappaB in cells productively infected with HSV-1 depends on activated protein kinase R and plays no apparent role in blocking apoptosis. Proc Natl Acad Sci USA 100:12408–12413. 10.1073/pnas.2034952100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Jones JO, Arvin AM. 2006. Inhibition of the NF-kappaB pathway by varicella-zoster virus in vitro and in human epidermal cells in vivo. J Virol 80:5113–5124. 10.1128/JVI.01956-05. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Sloan E, Henriquez R, Kinchington PR, Slobedman B, Abendroth A. 2012. Varicella-zoster virus inhibition of the NF-κB pathway during infection of human dendritic cells: role for open reading frame 61 as a modulator of NF-κB activity. J Virol 86:1193–1202. 10.1128/JVI.06400-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Whitmer T, Malouli D, Uebelhoer LS, DeFilippis VR, Früh K, Verweij MC. 2015. The ORF61 protein encoded by simian varicella virus and varicella-zoster virus inhibits NF-κB signaling by interfering with IκBα degradation. J Virol 89:8687–8700. 10.1128/JVI.01149-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Rong BL, Libermann TA, Kogawa K, Ghosh S, Cao LX, Pavan-Langston D, Dunkel EC. 1992. HSV-1-inducible proteins bind to NF-kappa B-like sites in the HSV-1 genome. Virology 189:750–756. 10.1016/0042-6822(92)90599-k. [DOI] [PubMed] [Google Scholar]
- 59.Goodkin ML, Ting AT, Blaho JA. 2003. NF-kappaB is required for apoptosis prevention during herpes simplex virus type 1 infection. J Virol 77:7261–7280. 10.1128/jvi.77.13.7261-7280.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Taddeo B, Zhang W, Lakeman F, Roizman B. 2004. Cells lacking NF-kappaB or in which NF-kappaB is not activated vary with respect to ability to sustain herpes simplex virus 1 replication and are not susceptible to apoptosis induced by a replication-incompetent mutant virus. J Virol 78:11615–11621. 10.1128/JVI.78.21.11615-11621.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Hadigal SR, Agelidis AM, Karasneh GA, Antoine TE, Yakoub AM, Ramani VC, Djalilian AR, Sanderson RD, Shukla D. 2015. Heparanase is a host enzyme required for herpes simplex virus-1 release from cells. Nat Commun 6:6985. 10.1038/ncomms7985. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Becker CH. 1967. Zur primären Schädigung vegetativer Ganglien nach Infektion mit dem Herpes suis Virus bei verschiedenen Tierarten. Experientia 23:209–210. 10.1007/BF02136291. [DOI] [PubMed] [Google Scholar]
- 63.Kaplan AS, Vatter AE. 1959. A comparison of herpes simplex and pseudorabies viruses. Virology 7:394–407. 10.1016/0042-6822(59)90068-6. [DOI] [PubMed] [Google Scholar]
- 64.McFerran JB, Dow C, McCracken RM. 1979. Experimental studies in weaned pigs with three vaccines against Aujeszky’s disease. Comp Immunol Microbiol Infect Dis 2:327–334. 10.1016/0147-9571(79)90020-1. [DOI] [PubMed] [Google Scholar]
- 65.Kimman TG, de Wind N, Oei-Lie N, Pol JM, Berns AJ, Gielkens AL. 1992. Contribution of single genes within the unique short region of Aujeszky’s disease virus (suid herpesvirus type 1) to virulence, pathogenesis and immunogenicity. J Gen Virol 73:243–251. 10.1099/0022-1317-73-2-243. [DOI] [PubMed] [Google Scholar]
- 66.de Wind N, Domen J, Berns A. 1992. Herpesviruses encode an unusual protein-serine/threonine kinase which is nonessential for growth in cultured cells. J Virol 66:5200–5209. 10.1128/JVI.66.9.5200-5209.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Klopfleisch R, Klupp BG, Fuchs W, Kopp M, Teifke JP, Mettenleiter TC. 2006. Influence of pseudorabies virus proteins on neuroinvasion and neurovirulence in mice. J Virol 80:5571–5576. 10.1128/JVI.02589-05. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Nauwynck HJ, Pensaert MB. 1995. Effect of specific antibodies on the cell-associated spread of pseudorabies virus in monolayers of different cell types. Arch Virol 140:1137–1146. 10.1007/BF01315422. [DOI] [PubMed] [Google Scholar]
- 69.Olsen LM, Ch'ng TH, Card JP, Enquist LW. 2006. Role of pseudorabies virus Us3 protein kinase during neuronal infection. J Virol 80:6387–6398. 10.1128/JVI.00352-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Ch'ng TH, Enquist LW. 2005. Efficient axonal localization of alphaherpesvirus structural proteins in cultured sympathetic neurons requires viral glycoprotein E. J Virol 79:8835–8846. 10.1128/JVI.79.14.8835-8846.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Gómez-Sebastián S, Tabarés E. 2004. Negative regulation of herpes simplex virus type 1 ICP4 promoter by IE180 protein of pseudorabies virus. J Gen Virol 85:2125–2130. 10.1099/vir.0.80119-0. [DOI] [PubMed] [Google Scholar]
- 72.Van Cleemput J, Koyuncu OO, Laval K, Engel EA, Enquist LW. 2021. CRISPR/Cas9-constructed pseudorabies virus mutants reveal the importance of UL13 in alphaherpesvirus escape from genome silencing. J Virol 95:e02286-20. 10.1128/JVI.02286-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Deruelle M, Geenen K, Nauwynck HJ, Favoreel HW. 2007. A point mutation in the putative ATP binding site of the pseudorabies virus US3 protein kinase prevents Bad phosphorylation and cell survival following apoptosis induction. Virus Res 128:65–70. 10.1016/j.virusres.2007.04.006. [DOI] [PubMed] [Google Scholar]
- 74.Meissonnier GM, Pinton P, Laffitte J, Cossalter AM, Gong YY, Wild CP, Bertin G, Galtier P, Oswald IP. 2008. Immunotoxicity of aflatoxin B1: impairment of the cell-mediated response to vaccine antigen and modulation of cytokine expression. Toxicol Appl Pharmacol 231:142–149. 10.1016/j.taap.2008.04.004. [DOI] [PubMed] [Google Scholar]
- 75.Devriendt B, Gallois M, Verdonck F, Wache Y, Bimczok D, Oswald IP, Goddeeris BM, Cox E. 2009. The food contaminant fumonisin B(1) reduces the maturation of porcine CD11R1(+) intestinal antigen presenting cells and antigen-specific immune responses, leading to a prolonged intestinal ETEC infection. Vet Res 40:40. 10.1051/vetres/2009023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Tombácz D, Tóth JS, Petrovszki P, Boldogkoi Z. 2009. Whole-genome analysis of pseudorabies virus gene expression by real-time quantitative RT-PCR assay. BMC Genomics 10:491. 10.1186/1471-2164-10-491. [DOI] [PMC free article] [PubMed] [Google Scholar]