Free access
Virology
Research Article
24 February 2021

The Autophagy-Initiating Protein Kinase ULK1 Phosphorylates Human Cytomegalovirus Tegument Protein pp28 and Regulates Efficient Virus Release

ABSTRACT

Autophagy is a catabolic process contributing to intrinsic cellular defense by degrading viral particles or proteins; however, several viruses hijack this pathway for their own benefit. The role of autophagy during human cytomegalovirus (HCMV) replication has not been definitely clarified yet. Utilizing small interfering RNA (siRNA)-based screening, we observed that depletion of many autophagy-related proteins resulted in reduced virus release, suggesting a requirement of autophagy-related factors for efficient HCMV replication. Additionally, we could show that the autophagy-initiating serine/threonine protein kinase ULK1 as well as other constituents of the ULK1 complex were upregulated at early times of infection and stayed upregulated throughout the replication cycle. We demonstrate that indirect interference with ULK1 through inhibition of the upstream regulator AMP-activated protein kinase (AMPK) impaired virus release. Furthermore, this result was verified by direct abrogation of ULK1 kinase activity utilizing the ULK1-specific kinase inhibitors SBI-0206965 and ULK-101. Analysis of viral protein expression in the presence of ULK-101 revealed a connection between the cellular kinase ULK1 and the viral tegument protein pp28 (pUL99), and we identified pp28 as a novel viral substrate of ULK1 by in vitro kinase assays. In the absence of ULK1 kinase activity, large pp28- and pp65-positive structures could be detected in the cytoplasm at late time points of infection. Transmission electron microscopy demonstrated that these structures represent large perinuclear protein accumulations presumably representing aggresomes. Our results indicate that HCMV manipulates ULK1 and further components of the autophagic machinery to ensure the efficient release of viral particles.
IMPORTANCE The catabolic program of autophagy represents a powerful immune defense against viruses that is, however, counteracted by antagonizing viral factors. Understanding the exact interplay between autophagy and HCMV infection is of major importance since autophagy-related proteins emerged as promising targets for pharmacologic intervention. Our study provides evidence for a proviral role of several autophagy-related proteins suggesting that HCMV has developed strategies to usurp components of the autophagic machinery for its own benefit. In particular, we observed strong upregulation of the autophagy-initiating protein kinase ULK1 and further components of the ULK1 complex during HCMV replication. In addition, both siRNA-mediated depletion of ULK1 and interference with ULK1 protein kinase activity by two chemically different inhibitors resulted in impaired viral particle release. Thus, we propose that ULK1 kinase activity is required for efficient HCMV replication and thus represents a promising novel target for future antiviral drug development.

INTRODUCTION

Autophagy describes a cellular degradative process that is conserved in eukaryotic cells and induced by various stress conditions (1). The molecular machinery underlying this process was first described in yeast, and since then, numerous proteins and their homologs in different species have been found to be important for autophagy (2). The best-studied type of autophagy to date, macroautophagy (referred to here as autophagy), is defined by the involvement of a distinct transport vesicle, the autophagosome (1, 2). The process of autophagy starts at the preautophagosomal structure (PAS), where several proteins locate to and initiate the elongation of an isolation membrane directly around a cargo that undergoes degradation (1). Once the isolation membrane is closed, the resulting autophagosome can fuse with a lysosome to form an autolysosome, where the cargo is finally degraded (1). For a long time, it was assumed that autophagy is exclusively a nonspecific degradation process; however, in the meantime, different autophagic receptor proteins, including p62 (SQSTM1), have been detected (3).
In addition to its role in cellular homeostasis, autophagy has been implicated as a powerful host defense mechanism by targeting and degrading invading pathogens such as viruses (4). On the other hand, however, viruses appear to utilize components of the autophagy machinery to foster their own intracellular growth or cellular egress (5). For herpesviruses, the role and regulation of autophagy during replication are still discussed in a controversial manner, with conflicting results available not only for members of different subfamilies but also for single species (4, 610). Concerning its role in the context of human cytomegalovirus (HCMV) infection, it has been described that autophagy is upregulated immediately upon the entry of virions but then actively counteracted by an interaction of the viral proteins IRS1 and TRS1 with cellular beclin-1 (1113). However, whether autophagy acts in a pro- or antiviral manner during HCMV infection is still controversially discussed (11, 1417). Since autophagy provides membrane material, it is plausible to consider a link between autophagy and late steps of the HCMV replication cycle, such as secondary envelopment, when the newly forming viral particles acquire the final envelope. One protein implicated in this process is the viral tegument protein pp28 (pUL99), which has been shown to be essential for secondary envelopment (18).
ULK1 (unc-51-like kinase 1) is a serine/threonine protein kinase and besides ULK2 is the only protein kinase of the core autophagy machinery (19). It is a key regulator of autophagy that initiates the formation of the autophagosome by phosphorylating numerous proteins, including RB1CC1 (FIP200), ATG13, and beclin-1 (2024). The kinase activity of ULK1 is inversely regulated by the activity of the AMP-activated protein kinase (AMPK) and the mammalian target of rapamycin (mTOR) (21, 22, 2528). In a favorable, nutrient-rich cellular environment, the activity of mTOR is predominant, and ULK1 is phosphorylated by mTOR at different serines, including S758, and therefore remains in an inactive state (2830). In the case of cellular stress such as glucose deprivation, the activity of AMPK becomes predominant, and ULK1 is phosphorylated by AMPK at S317 and S777 as well as other serines (2730). This results in an active state that enables ULK1 to initiate the formation of autophagosomes (2730). In addition to its function as an initiator of autophagy, ULK1 is involved in various cellular pathways. ULK1 has been reported to act as a negative regulator of the stimulator of interferon genes (STING) and thereby contributes to the regulation of innate immunity (31). Additionally, ULK1 has been implicated in endoplasmic reticulum (ER)-to-Golgi apparatus trafficking and axon projection in mice (32, 33).
Utilizing a small interfering RNA (siRNA)-based screening approach, we provide evidence that a large number of autophagy-related proteins are required for efficient replication of HCMV, which includes ULK1 as a key initiator of autophagy. We observe a distinct upregulation of ULK1 expression and phosphorylation at early to late times of the HCMV replication cycle. Via inhibition of ULK1 kinase activity with two chemically different substances, we provide evidence for a novel function of ULK1 during the late phase of HCMV replication and a connection between ULK1 and the viral tegument protein pp28.

RESULTS

Knockdown of multiple autophagy-related genes interferes with HCMV replication.

So far, investigations on the role of autophagy during HCMV infection have mainly focused on a few autophagy-related proteins or viral factors (1117, 34, 35). In order to gain a more complete picture concerning the impact of individual autophagy-related proteins on HCMV replication, we decided to perform an RNA interference (RNAi)-based screening (Fig. 1). For this, primary human foreskin fibroblasts (HFFs) were transfected with either a nontargeting control or one of the 48 individual siRNA mixtures targeting various autophagy-related genes. To avoid interference between transfection and infection, cells were infected at 48 h posttransfection (hpt). Infection was performed with HCMV strain AD169 at a multiplicity of infection (MOI) of 0.1. At 96 h postinfection (hpi), viral genome equivalents in the supernatant were quantitated by TaqMan-based quantitative PCR (qPCR), and the cells from one replicate per sample were lysed and subjected to Western blot analyses (Fig. 1A and C). Thirty-five of the tested siRNAs, including siRNAs targeting the ULK1 complex constituents RB1CC1, ULK1, ATG13, and ULK2, effectively impaired the release of viral genomes into the supernatant, whereas 13 did not have a statistically significant effect compared to the nontargeting control (Fig. 1A). The reduction of viral genomes ranged between approximately 26% for CTSS/CTSD and 66% for CXCR4/UVRAG. Surprisingly, none of the siRNAs tested in this setting enhanced the release of viral genomes into the supernatant. To exclude that the transfection of the nontargeting control had any effect on viral replication and thereby affected the interpretation of the results, we also quantitated viral genomes in the supernatant of cells transfected with increasing amounts of the nontargeting control (6.25 to 100 nM) and compared them to those in mock-transfected samples but observed no difference (Fig. 1B). Regarding the expression levels of different viral marker proteins, we found that only a few siRNAs negatively affected viral gene expression (Fig. 1C). This was clearly the case for siRNAs targeting MTOR, UVRAG, or WDR45 (WIPI4). None of the other siRNAs showed a noticeable effect on viral protein expression, implying that the cause of the reduced virus release shown in Fig. 1A is an impairment of late steps of viral replication, such as tegumentation, secondary envelopment, or viral particle release.
FIG 1
FIG 1 siRNA-mediated knockdown of autophagy-related genes inhibits the release of viral genomes and viral gene expression. HFFs were transfected in triplicates with specific siRNAs or a nontargeting control at a concentration of 25 nM (A and C) or 6.25 to 100 nM (B) 48 h before infection with HCMV strain AD169 (MOI of 0.1). (A and B) At 96 hpi, viral genomes from cell culture supernatants were quantitated by TaqMan-based qPCR. Results are shown as means ± standard deviations (SD) from two different experiments performed in triplicates. Constituents of the ULK1 complex are highlighted in dark gray. Asterisks between the vertical lines indicate statistical significance for the single siRNAs compared to the control. Statistical analysis was performed utilizing Student’s t test. n.s., not significant; *, P < 0.05; **, P < 0.01; ***, P < 0.001. (C) Alternatively, cells were lysed at 96 hpi and analyzed by SDS-PAGE and Western blotting to detect either viral proteins (IE1, pUL44, MCP, and pp28) as an infection control or ULK1. β-Actin served as an internal loading control. The following antibodies were used for detection: anti-IE1 (p63-27), anti-pUL44 (BS510), anti-MCP (28-4), anti-pp28 (41-18), anti-ULK1 (D9D7), and anti-β-actin (AC-15).
We were particularly interested in the role of ULK1 in HCMV infection as it acts as a key initiator of autophagy. Furthermore, ULK1 is the only protein kinase of the core autophagy machinery and may thus serve as a promising drug target. Knockdown of ULK1 resulted in a significant reduction of virus release of approximately 49% (Fig. 1A). Interestingly, the transfection of an siRNA targeting ATG13 also resulted in the depletion of ULK1 and a significant reduction of virus release (Fig. 1A and C, compare lanes 6 and 42). This is consistent with previous reports indicating that ATG13 as part of the ULK1 protein complex affects the stability of ULK1 (22, 25). Taken together, these data indicate that the depletion of many autophagy-related proteins, including ULK1, negatively affects HCMV replication, suggesting that autophagy-related factors may be required for efficient viral replication.

Components of the ULK1 complex and ULK1 phosphorylation are upregulated during HCMV infection.

To gain further insight into the regulation of proteins of the ULK1 complex during HCMV infection, HFFs were infected with HCMV strain AD169 or TB40/E (MOI of 1), harvested throughout the course of infection, and subjected to Western blot analyses (Fig. 2). The viral proteins IE1 (immediate early 1), pUL44, and pp28 (pUL99) were stained as marker proteins for the different phases of the HCMV replication cycle (immediate early, early, and late). The three major components of the ULK1 complex, namely, ULK1, RB1CC1 (FIP200), and ATG13, were clearly upregulated in both AD169- and TB40/E-infected cells (Fig. 2A and B). The same applied to the catalytic subunits of AMPK as well as mTOR, both regulators of ULK1 activity (21, 22, 2528). Epidermal growth factor receptor (EGFR), as an additional control, displayed the opposite trend, as it was downregulated.
FIG 2
FIG 2 Constituents of the ULK1 complex and phosphorylation levels of ULK1 are upregulated during HCMV infection. HFFs were infected with HCMV strain AD169 or TB40/E (MOI of 1), lysed at the indicated time points postinfection in parallel with mock-infected cells, and analyzed by SDS-PAGE and Western blotting. (A and B) Protein levels of constituents of the ULK1 complex (ULK1, RB1CC1, and ATG13) and upstream regulators of ULK1 (mTOR and AMPKα) were analyzed throughout the course of infection. (C) The phosphorylation status of ULK1 at distinct serines (S317, S556, S638, and S758) throughout the course of infection was determined utilizing phospho-specific antibodies. Detection of viral proteins (IE proteins, pUL44, and pp28) served as an infection control, and EGFR and β-actin served as internal loading controls. The following antibodies were used for detection: anti-mTOR, anti-AMPKα, anti-ULK1 (D9D7), anti-RB1CC1, anti-ATG13, anti-EGFR (D38B1), and anti-IE1 (p63-27) (A and B); anti-ULK1 (R600), anti-ULK1-P (S317) (D2B6Y), anti-ULK1-P (S556) (D1H4), anti-ULK1-P (S638), anti-ULK1-P (S758), and anti-IE86 (C); and anti-pUL44 (BS510), anti-pp28 (41-18), and anti-β-actin (AC-15) (A to C).
Since phosphorylation is a major mechanism affecting the activity of ULK1, we next investigated the phosphorylation status of ULK1 in AD169-infected cells using a panel of phospho-specific antibodies (29). Interestingly, the upregulation of ULK1 was not limited to total protein levels but could also be observed concerning the phosphorylation of ULK1 at four distinct serines (S317, S556, S638, and S758) (Fig. 2C). This was most evident at late times of infection, correlating with a distinct mobility shift of ULK1 upon detection with anti-ULK1 antibody R600 (Fig. 2C, top panel, lanes 7 to 10). Surprisingly, since we observed enhanced phosphorylation signals at both S317 and S758, AMPK-dependent activating as well as mTOR-dependent inactivating phosphorylation sites of ULK1 were affected equally (29). These findings in conjunction with previous results suggesting that autophagy is counteracted by HCMV led us to the assumption that ULK1 may play a role during HCMV infection beyond its function in the initiation of autophagy (1113).

Inhibition of ULK1 phosphorylation impairs viral replication.

Intrigued by the finding that both activating as well as inactivating phosphorylation of ULK1 were affected during HCMV infection, we became interested in further elucidating the role of the upstream regulators AMPK and mTOR. One approach was the treatment of AD169-infected HFFs (MOI of 0.5) with different inhibitors of AMPK and mTOR followed by Western blot analyses (Fig. 3). Compound C (10 μM) was used to block AMPK activity, while torin 1 (100 nM) and rapamycin (50 nM) were used as mTOR inhibitors. AG490 (10 μM), an EGFR inhibitor, served as a negative control. Inhibitors were added at either 72 or 96 hpi to circumvent well-known negative effects on HCMV replication upon inhibition of AMPK or mTOR at earlier time points of infection (Fig. 3A) (3639). At 120 hpi, cells were lysed and subjected to sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and Western blotting (Fig. 3B). The addition of the AMPK inhibitor compound C reduced the phosphorylation of ULK1 at the AMPK-specific phosphorylation site S317 by 20 and 70% after treatment for 24 and 48 h (addition at 96 and 72 hpi), respectively (Fig. 3B, lanes 3 and 9). Viral gene expression was hardly affected, although the levels of pp28 appeared to be slightly increased. Total levels of ULK1 protein were reduced after treatment with compound C by 40 and 20%, indicative of either reduced ULK1 upregulation or decreased stability of the protein. Interestingly, the additions of the two mTOR inhibitors torin 1 and rapamycin differed concerning their effects on the phosphorylation of ULK1 at S758 (Fig. 3B, lanes 4, 5, 10, and 11). While rapamycin had no effect, torin 1 efficiently reduced the phosphorylation of ULK1 at S758 by 50 and 60% after addition at 96 and 72 hpi, respectively. Viral gene expression of IE proteins or pUL44 was not affected, but in contrast to compound C, the addition of both inhibitors slightly decreased pp28 levels. Regarding total protein levels of ULK1, torin 1 reduced ULK1 levels by 50 and 40%, whereas rapamycin had no prominent effect.
FIG 3
FIG 3 Inhibition of AMPK and mTOR affects the phosphorylation status of ULK1. To determine the effect of AMPK/mTOR inhibition on the phosphorylation of ULK1, HFFs were infected with HCMV strain AD169 (MOI of 0.5) and treated with inhibitors targeting AMPK (compound C [comp C]), mTOR (torin 1 and rapamycin), or EGFR (AG490) (negative control) beginning at 96 and 72 hpi (treatment periods of 24 and 48 h, respectively). At 120 hpi, cells were lysed and analyzed by SDS-PAGE and Western blotting (WB). (A) Schematic representation of the experimental setup. (B) Total protein levels and the phosphorylation status of ULK1 at an AMPK-specific serine (S317) and an mTOR-specific serine (S758) were determined utilizing either an antibody detecting ULK1 independent of its phosphorylation status or phospho-specific antibodies. The following compounds were added at the indicated concentrations: compound C (10 μM), torin 1 (100 nM), rapamycin (50 nM), and AG490 (10 μM). Signal intensities were quantitated relative to infected control cells (lanes 2 and 8). Detection of viral proteins (IE proteins, pUL44, and pp28) served as an infection control, and β-actin served as an internal loading control. The following antibodies were used for detection: anti-ULK1 (R600), anti-ULK1-P (S317) (D2B6Y), anti-ULK1-P (S758), anti-IE86, anti-pUL44 (BS510), anti-pp28 (41-18), and anti-β-actin (AC-15).
Based on the finding that solely compound C and torin 1 affected the phosphorylation of ULK1 in this setting, both inhibitors were analyzed concerning their effect on HCMV replication. To investigate this, we infected HFFs with AD169 (MOI of 1) and added compound C (10 μM), torin 1 (100 nM), or AG490 (10 μM) at 48 hpi (Fig. 4A). Cell culture supernatants including inhibitors were renewed at 72 and 96 hpi to ensure the integrity of the inhibitors throughout the experiment. At 96 and 120 hpi, supernatants were harvested, and virus release was quantitated via either the determination of viral genomes by qPCR or titration of infectious viral particles (determined as IE1 protein-forming units [IEU] per milliliter) (Fig. 4B and C). The addition of compound C impaired the release of viral genomes into the supernatant more than 10-fold at both investigated time points, and the numbers of infectious particles were significantly reduced as well. In contrast, the effect of torin 1 was less pronounced and reduced the levels of viral genomes and infectious particles to a much lesser extent. Considering that the addition of compound C inhibits ULK1, while torin 1 leads to activation, we speculated that ULK1 possibly acts as a proviral factor during HCMV replication.
FIG 4
FIG 4 Inhibition of AMPK but not mTOR negatively affects the release of both viral genomes and infectious particles. To determine the effect of AMPK/mTOR inhibition on HCMV replication, HFFs were infected in triplicates with HCMV strain AD169 (MOI of 1) and treated with inhibitors targeting AMPK (compound C [comp C]), mTOR (torin 1), or EGFR (AG490) (negative control) beginning at 48 hpi. Inhibitors were renewed at 72 and 96 hpi to ensure the integrity of the inhibitors throughout the experiment. At 96 and 120 hpi, viral genomes (B) and infectious particles (C) from cell culture supernatants (SN) were quantitated by TaqMan-based qPCR and titration (IE1 protein-forming units [IEU] per milliliter), respectively. (A) Schematic representation of the experimental setup. (B and C) Results shown as means ± SD from triplicates. Statistical analysis was performed utilizing Student’s t test. n.s., not significant; *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001. The following compounds were added at the indicated concentrations: compound C (10 μM), torin 1 (100 nM), and AG490 (10 μM).

Direct inhibition of ULK1 kinase activity.

To further analyze the function of ULK1 during HCMV replication, we decided to manipulate the activity of ULK1 directly by using the ULK1 kinase inhibitor SBI-0206965 (40). First, we wanted to confirm the inhibitory activity of SBI-0206965 by both in vitro kinase and cell culture experiments (Fig. 5). For the in vitro kinase assay, HEK293T cells were cotransfected with plasmids for myc-tagged ATG13 (serving as a substrate) in combination with either hemagglutinin (HA)-tagged ULK1 or the kinase-dead mutant HA-ULK1(K46I) (Fig. 5A). After lysis at 2 days posttransfection (dpt), small fractions of the cell lysates were analyzed by Western blotting (input control), and the remainder was subjected to immunoprecipitation (IP) with HA- or myc-specific antibodies. Again, small fractions of the immunoprecipitates were analyzed by Western blotting (precipitation control). The remaining samples were utilized in an in vitro kinase reaction in the presence or absence of SBI-0206965. Subsequent detection of phosphorylated proteins was accomplished via SDS-PAGE, blotting onto a membrane, and autoradiography (in vitro kinase reaction). The coexpression of kinase-active HA-ULK1 and myc-ATG13 clearly resulted in the presence of a second, higher-migrating ATG13 band (myc-ATG13[-P]) in the input control, which was not detectable upon the coexpression of the kinase-dead mutant HA-ULK1(K46I) (Fig. 5A, lanes 1 and 2). The autoradiograph obtained after the in vitro kinase reaction showed distinct phosphorylation signals for HA-ULK1 (autophosphorylation) and myc-ATG13 (Fig. 5A, top panel, lane 2). In line with the results of Egan et al. demonstrating that SBI-0206965 is a potent inhibitor of ULK1 kinase activity, the presence of SBI-0206965 considerably decreased the autophosphorylation of HA-ULK1 as well as the phosphorylation of myc-ATG13 (Fig. 5A, top panel, lane 3) (40).
FIG 5
FIG 5 SBI-0206965 inhibits ULK1-dependent ATG13 phosphorylation in vitro and in cell culture. To investigate the inhibitory effect of SBI-0206965 on ULK1 kinase activity, an in vitro kinase assay and cell culture experiments were performed. (A) For the in vitro kinase assay, HEK293T cells were cotransfected with plasmids coding for HA-tagged ULK1 or a kinase-dead mutant ULK1 (K46I) as a kinase and myc-tagged ATG13 as a substrate. At 2 dpt, HA-ULK1/HA-ULK1(K46I) and myc-ATG13 were immunoprecipitated with monoclonal HA (anti-HA [HA-7])- and myc (anti-myc [MYC 1-9E10.2])-specific antibodies, respectively, and subjected to an in vitro kinase reaction. SBI-0206965 (10 μM) or DMSO was added 30 min prior to the kinase reaction directly to the precipitated proteins and was additionally present during the kinase reaction. Radioactively labeled proteins (i.e., phosphorylated proteins) were visualized on a photostimulable phosphor plate (in vitro kinase reaction). Total protein levels of HA-ULK1, HA-ULK1(K46I), and myc-ATG13 were determined before and after immunoprecipitation to confirm successful expression (input control) and immunoprecipitation (precipitation control), respectively. auto-P, autophosphorylation. (B) For verification of the inhibitory effect of SBI-0206965 in cell culture, HEK293T cells were cotransfected with plasmids coding for HA-tagged ULK1 or a kinase-dead mutant ULK1 (K46I) and myc-tagged ATG13. At 2 dpt, SBI-0206965 (2 or 10 μM) or DMSO was added 1 h prior to lysis and subsequent analysis by SDS-PAGE and Western blotting. As an expression control, total protein levels of HA-ULK1, HA-ULK1(K46I), and myc-ATG13 were analyzed, and the phosphorylation of ATG13 at the ULK1-specific phosphorylation site S318 was determined utilizing a phospho-specific antibody. Signal intensities were quantitated relative to control cells (lane 4). β-Actin served as an internal loading control. The following antibodies were used for detection: anti-HA (HA-7) and anti-myc (MYC 1-9E10.2) (A and B) and anti-ATG13-P (S318) and anti-β-actin (AC-15) (B).
In order to analyze whether SBI-0206965 is cell permeable and inhibits the phosphorylation of ATG13 in analogous cell culture experiments, we cotransfected HEK293T cells with plasmids for HA-ULK1, the kinase-dead mutant HA-ULK1(K46I), and myc-ATG13. One hour prior to lysis, SBI-0206965 or the solvent control dimethyl sulfoxide (DMSO) was added directly to the cell culture supernatant (Fig. 5B). Staining with a phospho-specific antibody that detects ULK1-specific ATG13 phosphorylation at S318 revealed that the coexpression of catalytically active HA-ULK1 and myc-ATG13 resulted in a substantially increased phosphorylation of ATG13 that was not present in samples transfected with the kinase-dead mutant HA-ULK1(K46I) (Fig. 5B, third panel, lanes 4 and 7) (41). The addition of SBI-0206965 at two different concentrations led to a considerable decrease in the phosphorylation signal at S318 by up to 50% in proportion to the concentration of the inhibitor (Fig. 5B, lanes 5 and 6). This experiment demonstrates that the inhibitory effect of SBI-0206965 is evident not only in vitro but also in cell culture experiments.

Inhibition of ULK1 kinase activity negatively affects late steps of viral replication.

We already observed that RNAi-mediated depletion of ULK1 impairs virus release into the cell culture supernatant (Fig. 1A). As a next step, we were interested in the role of ULK1 kinase activity in HCMV replication. Consequently, virus release assays were performed in the absence or presence of either SBI-0206965 or ULK-101, a novel and potent ULK1 inhibitor (Fig. 6A to C) (42). To investigate whether ULK1 kinase activity is important for HCMV replication at the immediate early, early, or late stage of infection, HFFs were infected with AD169 (MOI of 0.1), and the respective inhibitors were added at 1.5, 24, or 48 hpi at different concentrations as indicated (Fig. 6A). At 96 hpi, viral genomes were quantitated in cell culture supernatants by TaqMan-based qPCR. The addition of both inhibitors resulted in the reduced release of viral genomes into the supernatant irrespective of the time point of addition. SBI-0206965 impaired virus release starting at a concentration of 10 μM, whereas ULK-101 proved to be considerably more potent, with significant impairment starting at a concentration of 0.63 μM (Fig. 6A). At a concentration of 20 μM, SBI-0206965 decreased the number of viral genomes in the supernatant by 58 to 93%, while ULK-101 impaired the release of viral genomes by 65 to 89% at a concentration of 1.25 μM. The impact of ULK1 kinase inhibition on the release of viral genomes was less pronounced after the addition of the inhibitors at 48 hpi than after an earlier addition at 1.5 or 24 hpi. However, the fact that the addition of ULK1 inhibitors at 48 hpi was still able to induce a significant impairment of virus release implies that ULK1 kinase activity plays an important proviral role in late events of the viral replication cycle. As a next step, we aimed to analyze whether the reduced release of viral genomes that we observed in the absence of ULK1 kinase activity is limited to a specific HCMV strain, MOI, or time point of analysis. Thus, HFFs were infected with either AD169 or TB40/E (MOI of 0.1 or 1), followed by the addition of SBI-0206965 (20 μM) or ULK-101 (1.25 μM) at 48 hpi. Viral genomes in the cell culture supernatants were quantitated by TaqMan-based qPCR at 96 to 168 hpi (Fig. 6B and C). In AD169-infected cells, the release of viral genomes was reduced by 72 to 91% (MOI of 0.1) and 65 to 83% (MOI of 1) upon inhibition of ULK1 kinase activity (Fig. 6B). Similarly, in TB40/E-infected cells, release was reduced by 61 to 80% (MOI of 0.1) and 36 to 87% (MOI of 1) (Fig. 6C). In conclusion, irrespective of the viral strain, MOI, or time point of analysis, the release of viral genomes into the supernatant was significantly reduced in the absence of ULK1 kinase activity, with one exception. The addition of ULK1 kinase inhibitors did not show a significant impairment after infection with TB40/E at an MOI of 0.1 and analysis at 96 hpi; however, the total number of released viral genomes ranged between 695 and 2,321 on average and was therefore very low (Fig. 6C). To answer the question of whether inhibition of virus release by SBI-0206965 or ULK-101 is due to negative effects on viral gene expression, we investigated the expression pattern of different viral marker proteins throughout the replication cycle in the presence of ULK1 kinase inhibitors (Fig. 6D). Expression of the immediate early and early proteins IE1 and pUL44 was not affected by the addition of ULK1 kinase inhibitors up to 96 hpi. Surprisingly, we observed a modest increase in steady-state levels of the late proteins′ major capsid protein (MCP) and pp28 (pUL99) at 96 hpi in the presence of ULK1 kinase inhibitors. Most strikingly, however, ULK-101 elicited a distinct mobility shift of the phosphoprotein pp28 (Fig. 6D, lanes 14, 15, 19, and 20). In particular, at 96 hpi, a slightly slower-migrating form of pp28 merged with a faster-migrating form in the presence of ULK-101 (Fig. 6D, lane 20). This suggests that phosphorylation of pp28 depends on ULK1 kinase activity. Taken together, our data demonstrate that two chemically distinct ULK1 kinase inhibitors interfere with the release of viral particles without an obvious negative effect on viral gene expression, indicating that ULK1 kinase activity is required for late events of the viral replication cycle.
FIG 6
FIG 6 Inhibition of ULK1 kinase activity inhibits the release of viral genomes and affects the phosphorylation of pp28. To determine the effect of ULK1 inhibition on HCMV replication, the release of viral genomes (A to C) and the expression of viral proteins (D) throughout the course of infection were analyzed. (A to C) HFFs were infected in triplicates with HCMV strain AD169 or TB40/E at the indicated MOIs, and the ULK1-specific inhibitor SBI-0206965 or ULK-101 or DMSO was added at 1.5, 24, and 48 hpi (A) or 48 hpi (B and C) at the indicated concentrations. At 96 hpi (A) or 96 to 168 hpi (B and C), viral genomes from cell culture supernatants were quantitated by TaqMan-based qPCR. Results are shown as means ± SD in a linear scale from two different experiments performed in triplicates (A) or a logarithmic scale from triplicates (B and C). In panel A, asterisks indicate statistical significance for the individual groups compared to DMSO. Statistical analysis was performed utilizing two-way analysis of variance (ANOVA) (A) or Student’s t test (B and C). n.s., not significant; *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001. (D) For analysis of viral protein expression kinetics, HFFs were infected with AD169 (MOI of 0.1), and SBI-0206965, ULK-101, or DMSO was added at 1.5 hpi at the indicated concentrations. Viral protein levels (IE1, pUL44, MCP, and pp28) were determined after lysis of the cells at the indicated time points postinfection and subsequent analysis by SDS-PAGE and Western blotting. β-Actin served as an internal loading control. The following antibodies were used for detection: anti-IE1 (p63-27), anti-pUL44 (BS510), anti-MCP (28-4), anti-pp28 (41-18), and anti-β-actin (AC-15).

The HCMV tegument protein pp28 (pUL99) is phosphorylated upon coexpression of ULK1 in vitro.

Due to the finding that inhibition of ULK1 kinase activity by ULK-101 changes the migration pattern of pp28 in SDS-PAGE gels, we asked the question of whether pp28 acts as a substrate for ULK1. To address this, an in vitro kinase assay was performed. After cotransfection of either HA-ULK1 or the kinase-dead mutant HA-ULK1(K46I) and green fluorescent protein (GFP)-tagged pp28 or GFP as a control, cells were lysed and subjected to immunoprecipitation with the indicated HA- or GFP-specific antibodies (Fig. 7). As evident in the input and precipitation controls, all proteins were expressed and immunoprecipitated to similar extents (Fig. 7A). In the in vitro kinase reaction, a clear phosphorylation signal was detectable for GFP-pp28 only after the coexpression of HA-ULK1, possibly representing direct phosphorylation (Fig. 7B, lane 3). Distinctively, in the absence of coexpressed HA-ULK1 or upon the coexpression of the kinase-dead mutant HA-ULK1(K46I), this phosphorylation signal was not observed (Fig. 7B, lanes 2 and 4). Furthermore, the coexpression of HA-ULK1 and GFP demonstrated that the GFP tag did not serve as a phosphorylation target (Fig. 7B, lane 5).
FIG 7
FIG 7 Coexpression of ULK1 leads to the phosphorylation of pp28 in vitro. To investigate whether pp28 is a substrate of ULK1, an in vitro kinase assay was performed. For this, HEK293T cells were cotransfected with plasmids coding for HA-tagged ULK1 or a kinase-dead mutant ULK1 (K46I) as a kinase and GFP-tagged pp28 as a substrate or the empty vector coding for GFP as a negative control. At 2 dpt, HA-ULK1/HA-ULK1(K46I) and GFP-pp28/GFP were immunoprecipitated with HA (anti-HA [HA-7])- and GFP (anti-GFP [clones 7.1 and 13.1])-specific antibodies, respectively, and subjected to an in vitro kinase reaction. (A) Total protein levels of HA-ULK1, HA-ULK1(K46I), GFP-pp28, and GFP were determined before and after immunoprecipitation to confirm successful expression (input control) and immunoprecipitation (precipitation control), respectively. The following antibodies were used for detection: anti-HA (HA-7) and anti-GFP (clones 7.1 and 13.1). (B) Radioactively labeled proteins (i.e., phosphorylated proteins) were visualized with a photostimulable phosphor plate (in vitro kinase reaction). IgG, immunoglobulin G; hc, heavy chain.

Inhibition of ULK1 kinase activity results in the induction of large cytoplasmic protein aggregates containing pp28 and pp65.

To further characterize the role of ULK1 in HCMV infection, we investigated whether ULK1 kinase activity affects the subcellular localization of pp28, pp65 (pUL83), MCP, and GM130 at late time points of infection (Fig. 8). For this, we infected HFFs with AD169 (MOI of 1) and added DMSO or ULK-101 (1.25 μM) at 48 hpi to exclude any potential immediate early or early effects of ULK1 inhibition. At 96 hpi, cells were fixed; stained for pp28, pp65, MCP, or GM130; and analyzed via fluorescence microscopy. In infected and DMSO-treated samples, pp28 localized primarily next to indentations of the crescent-shaped nuclei, consistent with the described accumulation of pp28 in cytoplasmic virion assembly complexes (cVACs) (Fig. 8A, panels 1 and 3) (43). The tegument protein pp65 localized to cytoplasmic speckles, MCP was detected in nuclear viral replication compartments and cVACs, and the cis-Golgi marker GM130 localized to cVACs (Fig. 8A, panels 5, 7, 9, 11, 13, and 15). In contrast, in the presence of the ULK1 kinase inhibitor ULK-101, pp28 and pp65 localized at large circular structures in the cytoplasm (Fig. 8C, panels 21, 23, 25, and 27, marked by white arrowheads). These structures were also readily visible in bright-field images but not in 4′,6-diamidino-2-phenylindole (DAPI) and MCP stainings, indicating that these structures do not represent accumulations of viral capsids (Fig. 8C, panels 21 to 32). The same applies to the cellular protein GM130, which did not colocalize with the cytoplasmic structures, indicating that cis-Golgi membranes are not involved in their formation (Fig. 8C, panels 33 and 35). To investigate whether the inhibition of ULK1 kinase activity itself results in the formation of these cytoplasmic structures, we analyzed mock-infected HFFs treated with either DMSO or ULK-101 (1.25 μM) (Fig. 8B and D). We found none of these structures in ULK-101-treated cells resembling the ones detected after infection, indicating that their formation is confined to infected cells (Fig. 8C and D, compare, e.g., panels 22 and 24 and panels 38 and 40). The substantial change in the localization pattern of pp28 and pp65 in the presence of ULK-101 implies that ULK1 kinase activity is important for the proper subcellular localization of both proteins at late time points of infection.
FIG 8
FIG 8 Inhibition of ULK1 kinase activity during infection alters the subcellular localization of the tegument proteins pp28 and pp65. To analyze the morphology of HCMV-infected cells after inhibition of ULK1 kinase activity, HFFs were infected with HCMV strain AD169 (MOI of 1) and treated with DMSO (A and B) or the ULK1-specific inhibitor ULK-101 (1.25 μM) (C and D) beginningat 48 hpi. At 96 hpi, cells were fixed with 4% paraformaldehyde (pp28, pp65, and GM130) or methanol (MCP) and immunostained for the tegument proteins pp28 and pp65, the capsid protein MCP, or the cis-Golgi marker GM130 utilizing anti-pp28 (41-18), anti-pp65 (65-33), anti-MCP (28-4), or anti-GM130 (clone 35) as antibodies. Large cytoplasmic accumulations are marked by white arrowheads. 4′,6-Diamidino-2-phenylindole (DAPI) was counterstained to visualize cellular nuclei.
Next, we analyzed ULK-101-treated cells by transmission electron microscopy (TEM) (Fig. 9 and 10). This was done to further characterize the pp28/pp65-positive cytoplasmic accumulations and to investigate whether the altered localization of pp28 leads to an impairment of secondary envelopment as described previously for HCMV mutants containing a manipulated UL99 open reading frame (18). HFFs were infected with AD169 or TB40/E (MOI of 1) followed by the addition of either the solvent control DMSO or ULK-101 (1.25 μM) at 48 hpi. At 96 hpi, samples were prepared by high-pressure freezing, freeze substitution, Epon embedding, and ultrathin sectioning. In both DMSO- and ULK-101-treated cells, typical kidney-shaped nuclei and cVACs (delineated by dashed lines) in nuclear indentations could be observed (Fig. 9A and E and Fig. 10A and E). The cVAC structures and compositions were characteristic of late stages of lytic HCMV infection and contained numerous viral particles (capsids and dense bodies), as typically observed in HCMV infection (Fig. 9B, C, F, and G and Fig. 10B, C, E, and F, marked by arrowheads). Most importantly, in AD169 and TB40/E infection, TEM of ULK-101-treated cells revealed no obvious defect in secondary envelopment, defined as an increased ratio of naked or budding capsids compared to control cells, as the majority of viral particles were enveloped. This suggests that the phosphorylation of pp28 by ULK1 is not essential for secondary envelopment. However, it is noteworthy that in cells infected with either of the virus strains, we observed at least a 2-fold reduction (∼2.7-fold for AD169 and ∼3.6-fold for TB40/E) of viral particles in the cVACs of ULK-101-treated cells (Table 1). Furthermore, TEM confirmed the presence of prominent cytoplasmic structures in the presence of ULK-101 that appeared as circular, electron-dense structures with diameters of between 1 and 7 μm often found in close proximity to the cVACs (Fig. 9H and I and Fig. 10G and H). These structures were clearly distinguishable from accumulations observed in DMSO-treated cells due to their different size, localization, and texture (Fig. 9D). The relatively high electron density and the absence of an enclosing membrane together with the results of immunofluorescence staining (Fig. 8) led us to the assumption that these structures are most likely extraordinarily large protein accumulations containing at least the tegument proteins pp28 and pp65. Higher magnifications of the large cytoplasmic structures in AD169-infected cells treated with ULK-101 revealed that they were often directly associated with vesicles that sometimes contained viral particles or intraluminal vesicles (Fig. 9H and I, marked as V) and membranes (Fig. 9H and I, marked by asterisks). However, the large structures were never completely enclosed by these membranes. In TB40/E-infected cells treated with ULK-101, these accumulations were generally surrounded by numerous DNA-containing, tegumented but nonenveloped capsids (Fig. 10G and H, marked by gray arrowheads). Taken together, our data suggest that ULK1 kinase activity is important to prevent the deregulated accumulation of pp28 and pp65 in the cytoplasm.
FIG 9
FIG 9 Inhibition of ULK1 kinase activity induces large cytoplasmic protein accumulations in AD169-infected HFFs. To further characterize the cytoplasmic accumulations detected by conventional fluorescence microscopy, transmission electron microscopy was performed. Here, HFFs were infected with HCMV strain AD169 (MOI of 1) and treated with DMSO (A to D) or the ULK1-specific inhibitor ULK-101 (1.25 μM) (E to I) beginning at 48 hpi. At 96 hpi, cells were prepared by high-pressure freezing, freeze substitution, Epon embedding, and ultrathin sectioning. (A and E) Overview of cells. cVACs are delineated with dashed lines. Bars, 10 μm. (B and F) Overview of cVACs. Bars, 1 μm. (C and G) Higher magnifications of areas within cVACs. Black arrowheads, virions; white arrowheads, examples of dense bodies. Bars, 1 μm. (D) Higher magnification of small accumulations in the cytoplasm (Cy) often observed in infected cells at the back of the nucleus (Nu) compared to the location of the cVACs. Bar, 1 μm. (H and I) Higher magnifications of large electron-dense accumulations associated with cellular membranes (*) and vesicles (V) in proximity to the cVAC. Bars, 1 μm.
FIG 10
FIG 10 Capsids associate with cytoplasmic protein accumulations in the absence of ULK1 kinase activity in TB40/E-infected HFFs. Here, HFFs were infected with HCMV strain TB40/E (MOI of 1) and treated with DMSO (A to C) or the ULK1-specific inhibitor ULK-101 (1.25 μM) (D to H) beginning at 48 hpi. At 96 hpi, cells were prepared by high-pressure freezing, freeze substitution, Epon embedding, and ultrathin sectioning. (A and D) Overview of cells. cVACs are delineated with dashed lines. Bars, 10 μm. (B and E) Overview of cVACs. Bars, 1 μm. (C and F) Higher magnifications of areas within cVACs. Black arrowheads, virions; gray arrowhead, nonenveloped tegumented capsid; white arrowheads, examples of dense bodies. Bars, 1 μm. (G) Overview of an area with large electron-dense accumulations that are associated with viral nonenveloped tegumented capsids. Bar, 2 μm. (H) Higher magnification of two electron-dense accumulations. Gray arrowheads, examples of nonenveloped tegumented capsids enclosing these accumulations. Bar, 1 μm. Nu, nucleus; Cy, cytoplasm.
TABLE 1
TABLE 1 Quantitation of viral particles (capsids and dense bodies) in cVACs from AD169- or TB40/E-infected HFFs treated with DMSO or ULK-101 at 4 dpi
VirusTreatmentNo. of cVACsNo. of particlesParticle/cVAC ratio
AD169DMSO121,13394.4
 ULK-101517234.4
 
TB40/EDMSO101,322132.2
 ULK-1011347436.5

DISCUSSION

In this study, we provide evidence that ULK1 and other autophagy-related proteins act in a proviral manner to ensure optimal HCMV replication. We could show by siRNA-based screening that the knockdown of many of the investigated genes negatively affected the release of viral genomes and, therefore, viral replication (Fig. 1A). The majority of siRNAs, which had an impact on virus release, did not alter viral gene expression (Fig. 1C). Thus, we presume that autophagy-related proteins affect very late steps of HCMV replication beyond viral gene expression. Importantly, none of the siRNAs led to an enhancement of viral replication, arguing against a dominant antiviral function of individual autophagy-related proteins. However, the knockdown efficiency of the single siRNAs remains to be evaluated. These results extend the observations of Taisne et al. showing that virus release but not viral gene expression is impaired after short hairpin RNA (shRNA)-mediated knockdown of LC3B, ATG5, BECN1, and ULK1 by demonstrating that the knockdown of even more autophagy-related genes impairs virus release but not viral gene expression (44). In contrast to our findings of a negative effect of ATG4B depletion on viral particle release, a recent publication by Zimmermann and colleagues reported that inhibition of ATG4B elicited enhanced viral replication (17). However, the results are difficult to compare because Zimmermann and colleagues used a dominant negative form of ATG4BC74A that was expressed from the viral backbone in the context of HCMV infection, whereas we utilized siRNA-mediated knockdown resulting in the depletion of ATG4B before infection. Furthermore, previous studies showed that ATG4BC74A-overexpressing cells exhibit an accumulation of ATG5- and ULK1-positive intermediate autophagic structures (45). This might affect the outcome of HCMV infection experiments.
In addition to our results of siRNA-mediated depletion experiments suggesting a proviral role of autophagy-related proteins, we were able to demonstrate that ULK1 and other constituents of the ULK1 complex, RB1CC1 (FIP200) and ATG13, were clearly upregulated upon HCMV infection (Fig. 2A and B). Increased levels of ULK1 were observed starting at 24 hpi and culminating during the late phase of the viral replication cycle. This corresponds to the time during infection when autophagy inhibition by TRS1/IRS1 has been reported (1113). At this point, we are not able to distinguish whether the upregulation of ULK1 is the result of a cellular feedback mechanism to counteract viral autophagy inhibition or whether viral functions directly evoke increased ULK1 levels. Even more importantly, we also observed a distinct mobility shift of ULK1 upon separation by SDS-PAGE, suggesting hyperphosphorylation during infection (Fig. 2C). ULK1 is known to act as a target for both AMPK and mTOR, which are considered the main upstream regulators of ULK1 (2830). Of note, previous studies already described the upregulation of both AMPK and mTOR in infected cells, which was confirmed by our experiments (Fig. 2A and B) (38, 46, 47). Consistently, increased phosphorylation of ULK1 could be detected for both S317 and S758, serving as the main target sites for AMPK and mTOR, respectively (Fig. 2C). Since AMPK is supposed to activate, while mTOR inhibits, ULK1, we addressed the consequences of this contradictory scenario by the use of the inhibitory substances compound C and torin 1 (28). The AMPK inhibitor compound C clearly reversed the increased phosphorylation at S317, while the mTOR inhibitor torin 1 abrogated the upregulation of phosphorylation at S758, arguing for specific interference of the used substances (Fig. 3B). The discrepancy of a parallel modification at activating and inhibiting ULK1 phosphorylation sites may be explained by the assumption that differently phosphorylated subpopulations of ULK1 are present in HCMV-infected cells. Considering the major regulatory role of ULK1 during autophagy, we speculated that these modifications were induced by HCMV in order to ensure successful replication. To investigate this, infected HFFs were treated with compound C and torin 1 at 48 hpi in order to block ULK1 activation by AMPK and its inhibition by mTOR during late times of infection, respectively. The subsequent quantitation of genome equivalents and infectious viruses contained in the supernatants revealed a reduction of newly released viral particles (Fig. 4B and C). Since AMPK inhibition evoked predominant effects, we presume that the upregulation and activation of ULK1 by AMPK contribute to the efficient release of viral particles from infected cells.
To further corroborate this and to answer the question of whether the kinase activity of ULK1 is required for HCMV particle release, we utilized the ULK1-specific kinase inhibitors SBI-0206965 and ULK-101 (40, 42). We demonstrate that SBI-0206965 inhibits the phosphorylation of ATG13 dependent on the overexpression of kinase-active ULK1 in vitro as well as in cell culture, thereby confirming previous results (Fig. 5) (40). Furthermore, we observed a dose-dependent negative effect of both inhibitors on virus release (Fig. 6A). Notably, the effect was not limited to a single HCMV strain, MOI, or time point of analysis (Fig. 6B and C). This result indicates that ULK1 kinase activity is important for HCMV replication. We confirmed this observation by analyzing different viral marker proteins that are expressed during the three major phases of HCMV replication by Western blotting in the presence of the ULK1 inhibitors (Fig. 6D). Viral marker proteins were not affected until very late time points of infection, further indicating that viral gene expression does not depend on ULK1 kinase activity. Intriguingly, the inhibition of ULK1 by ULK-101 presumably altered the phosphorylation of the viral tegument protein pp28 (pUL99), which is essential for secondary envelopment (18). Utilizing an in vitro kinase assay, we provide evidence that pp28 could act as a substrate of ULK1 (Fig. 7). Additionally, immunofluorescence analyses showed that pp28, together with pp65, localized to large cytoplasmic structures in the absence of ULK1 kinase activity at late time points of HCMV infection (Fig. 8). Transmission electron microscopy (TEM) identified these structures as large protein aggregates (Fig. 9 and 10). TEM also revealed that there was no obvious defect in secondary envelopment, which was unexpected in light of previously published results (44). Nevertheless, the number of viral particles in the cVACs was reduced in the absence of ULK1 kinase activity (Table 1). In TB40/E infection, the large number of capsids trapped at the protein aggregates could explain why fewer virions are released upon ULK-101 treatment. However, we did not find this phenotype in AD169 infection. This might be explained by the different replication dynamics of the two virus strains and, additionally, by differences in the accumulation kinetics of tegument proteins. In light of the impaired release of genome equivalents and infectivity, it appears that virion morphogenesis is deregulated upon inhibition of ULK1. Whether this is the cause or the consequence of the excessive aggregation of pp28 and pp65, indicating an imbalance between viral protein production and release from the cell in the form of mature virus particles, cannot be concluded from this experiment. However, one possible interpretation could be that the missing phosphorylation of pp28 by ULK1 in the presence of ULK1 inhibitors causes a deregulated aggregation of various viral proteins and, therefore, their reduced availability at the cVAC, making virion maturation less efficient. We consider it possible that ULK1 has additional functions during HCMV replication. However, our results provide evidence for a direct connection between ULK1 kinase activity and the correct localization of pp28 and pp65 at late time points of infection. We propose that ULK1 kinase activity prevents the aggregation of pp28 and pp65 in the cytoplasm at late time points of infection and thus contributes to the efficient release of viral particles.
In summary, our results complement the available evidence that HCMV needs components of the autophagic machinery for optimal viral replication (44). This contrasts with previous reports indicating that autophagy is an antiviral process (1417). A solution for this puzzling discrepancy may be that HCMV has evolved a fine-balanced requirement for autophagy during its replicative cycle. While HCMV may need basal levels of autophagy as well as specific autophagic proteins to ensure optimal viral morphogenesis, the induction of autophagy may result in xenophagy, which restricts viral infection. Thus, both interference with as well as induction of autophagy may constitute targets for inhibition of HCMV replication.

MATERIALS AND METHODS

Oligonucleotides and plasmid constructs.

The expression plasmid encoding HA-ULK1 was generated by amplification of the ULK1 coding sequence from pRK5_myc-ULK1 (a gift from Do-Hyung Kim; Addgene plasmid 31961) (22) utilizing 5′-GCATGAATTCATGTACCCATACGATGTTCCAGATTACGCTGAGCCCGGCCGCGGCGGCACAG-3′ (containing the HA sequence) and 5′-CGTATCTAGATTAGGCACAGATGCCAGTCAGC-3′ as primers and subsequent ligation into pcDNA3 (Invitrogen, Thermo Fisher Scientific, Waltham, MA, USA) with EcoRI and XbaI. An expression plasmid encoding the kinase-dead mutant HA-ULK1(K46I) was generated by site-directed mutagenesis with 5′-GATTTGGAGGTCGCCGTCATCTGCATTAACAAGAAGA-3′ and 5′-TCTTCTTGTTAATGCAGATGACGGCGACCTCCAAATC-3′ as primers and the above-mentioned plasmid pcDNA3_HA-ULK1 as a template. pRK5_myc-ATG13 was a gift from Do-Hyung Kim (Addgene plasmid 31965) (22). The expression plasmid encoding GFP-pp28 was generated by amplification of the pp28 (pUL99) coding sequence from pcDNA3_pp28 utilizing 5′-GCATAAGCTTTGGGTGCCGAACTCTGCAAACG-3′ and 5′-GCATGGATCCTTAAAAGGGCAAGGAGGCGGC-3′ as primers and subsequent ligation into pEGFP-C1 (Clontech/TaKaRa Bio, Saint-Germain-en-Laye, France) with HindIII and BamHI. The template vector pcDNA3_pp28 was generated by amplification of the pp28 (UL99) coding sequence utilizing 5′-GCATGAATTCATGGGTGCCGAACTCTGC-3′ and 5′-CGTATCTAGATTAAAAGGGCAAGGAGGCGGC-3′ as primers and subsequent ligation into pcDNA3 (Invitrogen, Thermo Fisher Scientific) with EcoRI and XbaI.

Antibodies.

The following monoclonal antibodies were used for Western blotting, immunoprecipitation (IP), and indirect immunofluorescence: anti-β-actin (AC-15) (catalog number A1978; Sigma-Aldrich, Taufkirchen, Germany), anti-EGFR (D38B1) (catalog number 4267; Cell Signaling Technology, Frankfurt am Main, Germany), anti-GFP (clones 7.1 and 13.1, a mixture of two monoclonal antibodies) (catalog number 11814460001; Roche Diagnostics, Mannheim, Germany), anti-GM130 (clone 35) (catalog number 610822; BD Biosciences, San Jose, CA, USA), anti-HA (HA-7) (catalog number H9658; Sigma-Aldrich), anti-IE1 (p63-27) (48), anti-MCP (28-4) (49), anti-myc (MYC 1-9E10.2; ATCC, LGC Standards, Wesel, Germany), anti-pp28 (41-18) (50), anti-pp65 (65-33) (kindly provided by W. J. Britt, Birmingham, AL, USA), anti-pUL44 (BS510) (kindly provided by B. Plachter, Mainz, Germany), and anti-ULK1 (D9D7) (catalog number 6439; Cell Signaling Technology). The following polyclonal antibodies were used for Western blotting: anti-AMPKα (catalog number 2532; Cell Signaling Technology), anti-ATG13 (catalog number SAB4200100; Sigma-Aldrich), anti-IE86 (51), anti-mTOR (catalog number 2972; Cell Signaling Technology), anti-RB1CC1 (catalog number 17250-1-AP; Proteintech Europe, Manchester, United Kingdom), and anti-ULK1 (R600) (catalog number 4773; Cell Signaling Technology). The following phospho-specific antibodies were used for Western blotting: polyclonal anti-ATG13-P (S318) (catalog number PAB19948; Abnova, Taipei City, Taiwan), monoclonal anti-ULK1-P (S317) (D2B6Y) (catalog number 12753; Cell Signaling Technology), monoclonal anti-ULK1-P (S556, equivalent to S555 of mouse Ulk1) (D1H4) (catalog number 5869; Cell Signaling Technology), polyclonal anti-ULK1-P (S638) (catalog number 12097; Cell Signaling Technology), and polyclonal anti-ULK1-P (S758, equivalent to S757 of mouse Ulk1) (catalog number 6888; Cell Signaling Technology). As secondary antibodies for Western blotting, horseradish peroxidase (HRP)-conjugated goat anti-mouse or -rabbit antibodies (Dianova, Hamburg, Germany) were used. As a secondary antibody for indirect immunofluorescence, a goat anti-mouse antibody conjugated with Alexa Fluor 488 (Invitrogen, Thermo Fisher Scientific) was used.

Cells, infection, and transfection.

Primary human foreskin fibroblasts (HFFs) were prepared from human foreskin tissue and maintained in minimum essential medium (MEM; Gibco, Thermo Fisher Scientific, Waltham, MA, USA) supplemented with 7% fetal bovine serum (FBS; Sigma-Aldrich, Taufkirchen, Germany) and 350 mg/liter l-glutamine (Sigma-Aldrich) or GlutaMAX (Gibco, Thermo Fisher Scientific). Infection experiments were performed with the HCMV strains AD169 and TB40/E, which were titrated by quantitating IE1 protein-forming units (IEU) as described previously (52). For standard infection, 3 × 105 or 1.2 × 105 HFFs were seeded into 6- or 12-well dishes 1 day prior to inoculation with 1 or 0.5 ml of the viral supernatant. At 1.5 h postinfection (hpi), fresh growth medium was added. Subsequently, infected cells and the resulting viral supernatant were used for analyses.
Human embryonic kidney HEK293T cells were maintained in Dulbecco’s modified Eagle medium (DMEM; Gibco, Thermo Fisher Scientific) supplemented with 10% FBS and 350 mg/liter l-glutamine. Transfection of HEK293T cells was performed utilizing calcium phosphate-DNA precipitates. For standard transfection, 7 × 105 HEK293T cells were seeded into 6-well dishes 1 day prior to transfection with 1 μg plasmid DNA. At 16 h posttransfection (hpt), cells were washed twice with phosphate-buffered saline (PBS) and provided with fresh growth medium. At 48 hpt, cells were used for subsequent analyses.

Compounds.

SBI-0206965 was purchased from either BioVision (Milpitas, CA, USA) (catalog number 9580) or Sigma-Aldrich (Taufkirchen, Germany) (catalog number SML1540), and ULK-101 was obtained from Selleck Chemicals (Munich, Germany) (catalog number S8793). Compound C (dorsomorphin) (catalog number 171260), torin 1 (mTOR inhibitor XI) (catalog number 475991), rapamycin (sirolimus) (catalog number 553210), and AG490 (tyrphostin B42) (catalog number 658401) were purchased from Sigma-Aldrich (Calbiochem, Taufkirchen, Germany). All compounds were dissolved in dimethyl sulfoxide (DMSO), aliquoted, and stored at −20°C or −80°C.

Western blotting and indirect immunofluorescence.

For Western blotting, whole-cell extracts were generated by resuspending cell pellets in PBS, mixing with the Laemmli-based buffer Roti Load 1 (Carl Roth, Karlsruhe, Germany), boiling at 95°C for 10 min, and subsequent sonication. Proteins were separated by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) with 6 to 12.5% polyacrylamide gels and blotted onto nitrocellulose (GE Healthcare, Chicago, IL, USA) or polyvinylidene difluoride (PVDF) (Bio-Rad, Hercules, CA, USA) membranes. Chemiluminescence was detected using an ECL Western blotting detection kit (GE Healthcare) according to the manufacturer’s instructions and digitized with Fusion FX (Vilber Lourmat, Collégien, France) or a Fujifilm LAS-1000 luminescent image analyzer (Fujifilm Europe, Düsseldorf, Germany). Signal intensities were quantitated utilizing Image Lab 6.0, and images were edited in Adobe Photoshop Elements 2018.
For indirect immunofluorescence, HFFs were cultivated at a density of 2.5 × 104 cells/well in μ-slide 8-well dishes (ibidi, Gräfelfing, Germany) with ibiTreat surface modification to ensure reliable adhesion of the cells. Cells were washed once with PBS before fixation with either 4% (wt/vol) paraformaldehyde (PFA) (dissolved in PBS) for 10 min at room temperature or methanol for 15 min at −20°C. Subsequently, cells were washed three times with PBS and, in the case of PFA fixation, permeabilized with 0.2% (vol/vol) Triton X-100 (dissolved in PBS) for 20 min at 4°C and additionally washed five times with PBS. Next, cells were incubated with PBS containing 2 mg/ml gamma globulins from human blood (Sigma-Aldrich) for 30 min at 37°C to block unspecific binding of the antibodies. Next, cells were first incubated with the respective primary antibodies diluted in 1% (vol/vol) FBS (diluted in PBS) for 1.5 h at 37°C and then incubated with secondary goat anti-mouse antibody conjugated with Alexa Fluor 488 (Invitrogen, Thermo Fisher Scientific) diluted 1:400 in 1% (vol/vol) FBS (diluted in PBS) for 45 min at 37°C after three washing steps with PBS. Following another three washing steps with PBS, cells were mounted with Vectashield antifade mounting medium (Vector Laboratories, Burlingame, CA, USA) containing 4′,6-diamidino-2-phenylindole (DAPI) and analyzed with a Zeiss Axio Observer.Z1 instrument with Apotome.2 using a 40× objective. Fluorescent dyes were excited at a wavelength of 475 or 385 nm, and emission was visualized utilizing appropriate filters excluding a signal overlap. Images were edited in ZEN 2.3 lite.

In vitro kinase assay.

One day prior to transfection, 5 × 106 HEK293T cells were seeded into 100-mm dishes. A total of 10 to 20 μg plasmid DNA per construct coding for either the kinase or the substrate protein was cotransfected utilizing calcium phosphate-DNA precipitates. At 2 days posttransfection (dpt), cells were lysed on ice for 20 min with IP buffer (50 mM Tris-HCl [pH 8.0], 150 mM NaCl, 5 mM EDTA, 0.5% [vol/vol] Nonidet P-40 substitute) freshly supplemented with protease inhibitors (1 mM phenylmethylsulfonyl fluoride [PMSF], 2 μg/ml aprotinin, 2 μg/ml leupeptin, 2 μg/ml pepstatin A). Cellular debris was removed by centrifugation at 20,000 × g for 10 min. One-twentieth of the lysate was mixed with the Laemmli-based buffer Roti Load 1 (Carl Roth) and boiled at 95°C for 10 min and served as the input control. The remaining lysate was mixed with protein A-Sepharose beads (Sigma-Aldrich) coupled with the respective antibodies and incubated for 2 h at 4°C in a rotating mixer. Beads were washed twice with IP buffer including protease inhibitors and twice with kinase buffer (20 mM HEPES [pH 7.4], 0.4 mM EDTA, 1 mM EGTA, 5 mM MgCl2, 50 μM dithiothreitol [DTT], 100 μM Na3VO4) including protease inhibitors. Two-sevenths of the protein-loaded beads were mixed with Roti Load 1 and boiled at 95°C for 10 min and served as the precipitation control. If appropriate, beads were preincubated with SBI-0206965 or DMSO for 30 min at 4°C in a rotating mixer. The kinase reaction was performed by mixing the remaining beads with kinase reaction buffer (kinase buffer supplemented with 10 μM ATP, 3 μCi [γ-33P]ATP per sample, and SBI-0206965 or DMSO, if appropriate) followed by incubation at 30°C for 30 min in a microtube shaker. The reaction was stopped by mixing the samples with Roti Load 1 and boiling at 95°C for 10 min. SDS-PAGE and Western blotting were performed as described above. Proteins labeled with [γ-33P]ATP during the kinase reaction were visualized by applying the Western blot membrane onto a photostimulable phosphor plate (Fujifilm) and subsequent scanning (in vitro kinase reaction). Images were edited in Aida software v4.22 and Adobe Photoshop Elements 2018.

Screening of siRNAs.

The siRNAs used in this study were a mixture of four individual siRNAs targeting the same gene (SMARTPool) that were further chemically modified to reduce off-target effects (ON-TARGETplus siRNA). They were purchased from Horizon Discovery (Cambridge, UK) and are as follows: nontargeting control pool (D-001810-10), AMBRA1 (L-029987-01), ATG2A (L-026591-02), ATG2B (L-016822-02), ATG3 (L-015375-00), ATG4A (L-005789-00), ATG4B (L-005786-00), ATG4C (L-005788-00), ATG4D (L-005790-00), ATG5 (L-004374-00), ATG7 (L-020112-00), ATG9A (L-014294-01), ATG9B (L-018082-02), ATG10 (L-019426-01), ATG12 (L-010212-00), ATG13 (L-020765-01), ATG14 (L-020438-01), ATG16L1 (L-021033-01), ATG16L2 (L-026687-01), ATG101 (L-017816-01), BECN1 (L-010552-00), CTSB (L-004266-00), CTSD (L-003649-00), CTSS (L-005844-00), CXCR4 (L-005139-00), GABARAP (L-012368-00), GABARAPL1 (L-014715-00), GABARAPL2 (L-006853-00), MAP1LC3A (L-013579-00), MAP1LC3B (L-012846-00), MAP1LC3C (L-032399-01), MTOR (L-003008-00), PIK3C3 (L-005250-00), PIK3CG (L-005274-00), PIK3R4 (L-005025-00), PRKAA1 (L-005027-00), RB1CC1 (L-021117-00), SQSTM1 (L-010230-00), TMEM74 (L-015923-02), TNFSF10 (L-011524-00), ULK1 (L-005049-00), ULK2 (L-005396-00), UVRAG (L-015465-00), VMP1 (L-015899-01), WDR45 (L-019758-01), WDR45B (L-017119-01), WIPI1 (L-018205-01), WIPI2 (L-020521-01), and ZFYVE1 (L-013086-01). One day after seeding 5 × 104 HFFs into 24-well dishes, the cells were transfected in triplicates with 12.5 pmol of the respective siRNA mixtures (corresponding to a final concentration of 25 nM) utilizing Lipofectamine 2000 transfection reagent (Invitrogen, Thermo Fisher Scientific, Waltham, MA, USA). As a control, the same amount of the ON-TARGETplus nontargeting control pool was used. At 48 hpt, HFFs were infected with the HCMV laboratory strain AD169 at a multiplicity of infection (MOI) of 0.1 and subjected to a virus release assay. At 96 hpi, the cells of one well of each triplicate were additionally lysed and used for Western blot analyses. Statistical analyses were performed with VassarStats (http://www.vassarstats.net/).

Virus release assay, qPCR, and determination of viral titers.

One day prior to infection, 1.2 × 105 HFFs were seeded in triplicates into 12-well dishes. Before infection with either of the HCMV strains AD169 and TB40/E (MOI of 0.1 or 1), cells were washed once with growth medium. After inoculation for 1.5 h, cells were washed twice with growth medium, and 1 ml growth medium was added. At 96 to 168 hpi, the entire supernatant was harvested and centrifuged at 200 × g for 5 min. Infectious particles (determined as IEU per milliliter) in the supernatant were quantitated as described previously (52). For qPCR, 20 μl of the supernatant per sample was mixed with 80 μl of a proteinase K solution (1.65 U/ml proteinase K, 50 mM KCl, 15 mM Tris, 2.5 mM MgCl2, 0.5% [vol/vol] Tween 20 [pH 8.0]) and incubated at 56°C for 1 h and subsequently at 95°C for 5 min. Next, 5 μl of the proteinase K-digested samples was mixed with 2× SsoAdvanced universal probes supermix (Bio-Rad, Hercules, CA, USA), primers (250 nM each) (5′-AAGCGGCCTCTGATAACCAAG-3′ and 5′-GAGCAGACTCTCAGAGGATCGG-3′), and a probe (150 nM) (5′-CATGCAGATCTCCTCAATGCGGCG-3′ [labeled with 6-carboxyfluorescein {FAM} and 6-carboxytetramethylrhodamine {TAMRA}]). qPCR was performed in an AriaMx real-time PCR system (Agilent, Santa Clara, CA, USA) and analyzed with AriaMx software v1.5. Thermal cycling conditions consisted of an initial denaturing step at 95°C (3 min) and 40 amplification cycles at 95°C (10 s) and 60°C (30 s). Alternatively, 5 μl of the proteinase K-digested samples was mixed with 2× TaqMan PCR master mix (Applied Biosystems, Thermo Fisher Scientific, Waltham, MA, USA), primers (375 nM), and a probe (200 nM), and qPCR was performed in an Applied Biosystems 7500 real-time PCR system (Applied Biosystems, Thermo Fisher Scientific). Thermal cycling conditions consisted of two initial steps at 50°C (2 min) and 95°C (10 min) and 40 amplification cycles at 95°C (15 s) and 60°C (60 s). In this case, analysis was performed using 7500 system SDS software v1.4. As a reference for quantitation of genome equivalents, serial 10-fold dilutions (107 to 102) of a plasmid containing the IE1 coding sequence were analyzed in parallel. Statistical analyses were performed with VassarStats (http://www.vassarstats.net/).

Transmission electron microscopy.

HFFs were seeded on sapphire discs (3 mm in diameter; Engineering Office M. Wohlwend GmbH, Sennwald, Switzerland) in μ-slide 8-well dishes (ibidi) at a density of 2.5 × 104 cells/well. One day later, cells were infected with HCMV strain AD169 or TB40/E (MOI of 1), and DMSO or ULK-101 (1.25 μM) was added at 48 hpi. At 96 hpi, EM sample preparation was conducted as described previously (53, 54). In brief, infected HFFs were immobilized by high-pressure freezing (Compact 01; Engineering Office M. Wohlwend GmbH) and subjected to freeze substitution in a substitution medium consisting of acetone with 0.2% osmium tetroxide, 0.1% uranyl acetate, and 5% water (55). After washing and stepwise embedding in epoxy resin (Sigma-Aldrich), ultrathin sections of a 70-nm thickness were prepared, mounted on Formvar-coated single-slot grids (Plano, Wetzlar, Germany), and examined with a JEM-1400 transmission electron microscope (JEOL, Tokyo, Japan) at an accelerating voltage of 120 kV.

ACKNOWLEDGMENTS

We thank Jens von Einem (Ulm, Germany) for providing helpful advice and the GM130 antibody, Paul Walther (Ulm, Germany) for support concerning EM, and Regina Müller (Erlangen, Germany) for excellent technical assistance.
This study was supported by the IZKF Erlangen (project A71 to T.S.), the Deutsche Forschungsgemeinschaft (grant STA357/7-1 to T.S.), and the Kompetenznetzwerk Zytomegalie Baden-Württemberg (KSKV002).

REFERENCES

1.
Parzych KR, Klionsky DJ. 2014. An overview of autophagy: morphology, mechanism, and regulation. Antioxid Redox Signal 20:460–473.
2.
Yang Z, Klionsky DJ. 2010. Mammalian autophagy: core molecular machinery and signaling regulation. Curr Opin Cell Biol 22:124–131.
3.
Lamark T, Svenning S, Johansen T. 2017. Regulation of selective autophagy: the p62/SQSTM1 paradigm. Essays Biochem 61:609–624.
4.
Alexander DE, Leib DA. 2008. Xenophagy in herpes simplex virus replication and pathogenesis. Autophagy 4:101–103.
5.
Kudchodkar SB, Levine B. 2009. Viruses and autophagy. Rev Med Virol 19:359–378.
6.
Graybill C, Morgan MJ, Levin MJ, Lee KS. 2018. Varicella-zoster virus inhibits autophagosome-lysosome fusion and the degradation stage of mTOR-mediated autophagic flux. Virology 522:220–227.
7.
Romeo MA, Masuelli L, Gaeta A, Nazzari C, Granato M, Gilardini Montani MS, Faggioni A, Cirone M. 2019. Impact of HHV-6A and HHV-6B lytic infection on autophagy and endoplasmic reticulum stress. J Gen Virol 100:89–98.
8.
Shao Z, Borde C, Quignon F, Escargueil A, Maréchal V. 2019. Epstein-Barr virus BALF0 and BALF1 modulate autophagy. Viruses 11:1099.
9.
Wen H-J, Yang Z, Zhou Y, Wood C. 2010. Enhancement of autophagy during lytic replication by the Kaposi’s sarcoma-associated herpesvirus replication and transcription activator. J Virol 84:7448–7458.
10.
Yakoub AM, Shukla D. 2015. Basal autophagy is required for herpes simplex virus-2 infection. Sci Rep 5:12985.
11.
Mouna L, Hernandez E, Bonte D, Brost R, Amazit L, Delgui LR, Brune W, Geballe AP, Beau I, Esclatine A. 2016. Analysis of the role of autophagy inhibition by two complementary human cytomegalovirus BECN1/Beclin 1-binding proteins. Autophagy 12:327–342.
12.
Chaumorcel M, Lussignol M, Mouna L, Cavignac Y, Fahie K, Cotte-Laffitte J, Geballe A, Brune W, Beau I, Codogno P, Esclatine A. 2012. The human cytomegalovirus protein TRS1 inhibits autophagy via its interaction with Beclin 1. J Virol 86:2571–2584.
13.
Chaumorcel M, Souquère S, Pierron G, Codogno P, Esclatine A. 2008. Human cytomegalovirus controls a new autophagy-dependent cellular antiviral defense mechanism. Autophagy 4:46–53.
14.
Belzile J-P, Sabalza M, Craig M, Clark E, Morello CS, Spector DH. 2016. Trehalose, an mTOR-independent inducer of autophagy, inhibits human cytomegalovirus infection in multiple cell types. J Virol 90:1259–1277.
15.
Clark AE, Sabalza M, Gordts PLSM, Spector DH. 2018. Human cytomegalovirus replication is inhibited by the autophagy-inducing compounds trehalose and SMER28 through distinctively different mechanisms. J Virol 92:e02015-17.
16.
Mukhopadhyay R, Venkatadri R, Katsnelson J, Arav-Boger R. 2018. Digitoxin suppresses human cytomegalovirus replication via Na+,K+/ATPase α1 subunit-dependent AMP-activated protein kinase and autophagy activation. J Virol 92:e01861-17.
17.
Zimmermann C, Krämer N, Krauter S, Strand D, Sehn E, Wolfrum U, Freiwald A, Butter F, Plachter B. 1 March 2020. Autophagy interferes with human cytomegalovirus genome replication, morphogenesis, and progeny release. Autophagy.
18.
Silva MC, Yu Q-C, Enquist L, Shenk T. 2003. Human cytomegalovirus UL99-encoded pp28 is required for the cytoplasmic envelopment of tegument-associated capsids. J Virol 77:10594–10605.
19.
Mizushima N, Yoshimori T, Ohsumi Y. 2011. The role of Atg proteins in autophagosome formation. Annu Rev Cell Dev Biol 27:107–132.
20.
Chan EYW, Longatti A, McKnight NC, Tooze SA. 2009. Kinase-inactivated ULK proteins inhibit autophagy via their conserved C-terminal domains using an Atg13-independent mechanism. Mol Cell Biol 29:157–171.
21.
Hosokawa N, Hara T, Kaizuka T, Kishi C, Takamura A, Miura Y, Iemura S, Natsume T, Takehana K, Yamada N, Guan J-L, Oshiro N, Mizushima N. 2009. Nutrient-dependent mTORC1 association with the ULK1-Atg13-FIP200 complex required for autophagy. Mol Biol Cell 20:1981–1991.
22.
Jung CH, Jun CB, Ro S-H, Kim Y-M, Otto NM, Cao J, Kundu M, Kim D-H. 2009. ULK-Atg13-FIP200 complexes mediate mTOR signaling to the autophagy machinery. Mol Biol Cell 20:1992–2003.
23.
Zachari M, Ganley IG. 2017. The mammalian ULK1 complex and autophagy initiation. Essays Biochem 61:585–596.
24.
Russell RC, Tian Y, Yuan H, Park HW, Chang Y-Y, Kim J, Kim H, Neufeld TP, Dillin A, Guan K-L. 2013. ULK1 induces autophagy by phosphorylating Beclin-1 and activating VPS34 lipid kinase. Nat Cell Biol 15:741–750.
25.
Ganley IG, Lam DH, Wang J, Ding X, Chen S, Jiang X. 2009. ULK1·ATG13·FIP200 complex mediates mTOR signaling and is essential for autophagy. J Biol Chem 284:12297–12305.
26.
Lee JW, Park S, Takahashi Y, Wang H-G. 2010. The association of AMPK with ULK1 regulates autophagy. PLoS One 5:e15394.
27.
Egan DF, Shackelford DB, Mihaylova MM, Gelino SR, Kohnz RA, Mair W, Vasquez DS, Joshi A, Gwinn DM, Taylor R, Asara JM, Fitzpatrick J, Dillin A, Viollet B, Kundu M, Hansen M, Shaw RJ. 2011. Phosphorylation of ULK1 (hATG1) by AMP-activated protein kinase connects energy sensing to mitophagy. Science 331:456–461.
28.
Egan DF, Kim J, Shaw RJ, Guan K-L. 2011. The autophagy initiating kinase ULK1 is regulated via opposing phosphorylation by AMPK and mTOR. Autophagy 7:643–644.
29.
Kim J, Kundu M, Viollet B, Guan K-L. 2011. AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1. Nat Cell Biol 13:132–141.
30.
Shang L, Chen S, Du F, Li S, Zhao L, Wang X. 2011. Nutrient starvation elicits an acute autophagic response mediated by Ulk1 dephosphorylation and its subsequent dissociation from AMPK. Proc Natl Acad Sci U S A 108:4788–4793.
31.
Konno H, Konno K, Barber GN. 2013. Cyclic dinucleotides trigger ULK1 (ATG1) phosphorylation of STING to prevent sustained innate immune signaling. Cell 155:688–698.
32.
Joo JH, Wang B, Frankel E, Ge L, Xu L, Iyengar R, Li-Harms X, Wright C, Shaw TI, Lindsten T, Green DR, Peng J, Hendershot LM, Kilic F, Sze JY, Audhya A, Kundu M. 2016. Noncanonical role of ULK/ATG1 in ER-to-Golgi trafficking is essential for cellular homeostasis. Mol Cell 62:491–506.
33.
Wang B, Iyengar R, Li-Harms X, Joo JH, Wright C, Lavado A, Horner L, Yang M, Guan J-L, Frase S, Green DR, Cao X, Kundu M. 2018. The autophagy-inducing kinases, ULK1 and ULK2, regulate axon guidance in the developing mouse forebrain via a noncanonical pathway. Autophagy 14:796–811.
34.
McFarlane S, Aitken J, Sutherland JS, Nicholl MJ, Preston VG, Preston CM. 2011. Early induction of autophagy in human fibroblasts after infection with human cytomegalovirus or herpes simplex virus 1. J Virol 85:4212–4221.
35.
Zhao J, Li Z, Wang M, Zhang Z, Ma H, Chang J, Gao D, Wang S. 2013. Manipulation of autophagy by HCMV infection is involved in mTOR and influences the replication of virus. Acta Biochim Biophys Sin (Shanghai) 45:979–981.
36.
Terry LJ, Vastag L, Rabinowitz JD, Shenk T. 2012. Human kinome profiling identifies a requirement for AMP-activated protein kinase during human cytomegalovirus infection. Proc Natl Acad Sci U S A 109:3071–3076.
37.
Clippinger AJ, Maguire TG, Alwine JC. 2011. The changing role of mTOR kinase in the maintenance of protein synthesis during human cytomegalovirus infection. J Virol 85:3930–3939.
38.
McArdle J, Moorman NJ, Munger J. 2012. HCMV targets the metabolic stress response through activation of AMPK whose activity is important for viral replication. PLoS Pathog 8:e1002502.
39.
Moorman NJ, Shenk T. 2010. Rapamycin-resistant mTORC1 kinase activity is required for herpesvirus replication. J Virol 84:5260–5269.
40.
Egan DF, Chun MGH, Vamos M, Zou H, Rong J, Miller CJ, Lou HJ, Raveendra-Panickar D, Yang C-C, Sheffler DJ, Teriete P, Asara JM, Turk BE, Cosford NDP, Shaw RJ. 2015. Small molecule inhibition of the autophagy kinase ULK1 and identification of ULK1 substrates. Mol Cell 59:285–297.
41.
Joo JH, Dorsey FC, Joshi A, Hennessy-Walters KM, Rose KL, McCastlain K, Zhang J, Iyengar R, Jung CH, Suen D-F, Steeves MA, Yang C-Y, Prater SM, Kim D-H, Thompson CB, Youle RJ, Ney PA, Cleveland JL, Kundu M. 2011. Hsp90-Cdc37 chaperone complex regulates Ulk1- and Atg13-mediated mitophagy. Mol Cell 43:572–585.
42.
Martin KR, Celano SL, Solitro AR, Gunaydin H, Scott M, O’Hagan RC, Shumway SD, Fuller P, MacKeigan JP. 2018. A potent and selective ULK1 inhibitor suppresses autophagy and sensitizes cancer cells to nutrient stress. iScience 8:74–84.
43.
Seo JY, Britt WJ. 2006. Sequence requirements for localization of human cytomegalovirus tegument protein pp28 to the virus assembly compartment and for assembly of infectious virus. J Virol 80:5611–5626.
44.
Taisne C, Lussignol M, Hernandez E, Moris A, Mouna L, Esclatine A. 2019. Human cytomegalovirus hijacks the autophagic machinery and LC3 homologs in order to optimize cytoplasmic envelopment of mature infectious particles. Sci Rep 9:4560.
45.
Fujita N, Hayashi-Nishino M, Fukumoto H, Omori H, Yamamoto A, Noda T, Yoshimori T. 2008. An Atg4B mutant hampers the lipidation of LC3 paralogues and causes defects in autophagosome closure. Mol Biol Cell 19:4651–4659.
46.
Hutterer C, Wandinger SK, Wagner S, Müller R, Stamminger T, Zeitträger I, Godl K, Baumgartner R, Strobl S, Marschall M. 2013. Profiling of the kinome of cytomegalovirus-infected cells reveals the functional importance of host kinases Aurora A, ABL and AMPK. Antiviral Res 99:139–148.
47.
Clippinger AJ, Alwine JC. 2012. Dynein mediates the localization and activation of mTOR in normal and human cytomegalovirus-infected cells. Genes Dev 26:2015–2026.
48.
Andreoni M, Faircloth M, Vugler L, Britt WJ. 1989. A rapid microneutralization assay for the measurement of neutralizing antibody reactive with human cytomegalovirus. J Virol Methods 23:157–167.
49.
Waldo FB, Tomana M, Britt WJ, Julian BA, Mestecky J. 1989. Non-specific mesangial staining with antibodies against cytomegalovirus in immunoglobulin-A nephropathy. Lancet i:129–131.
50.
Sanchez V, Sztul E, Britt WJ. 2000. Human cytomegalovirus pp28 (UL99) localizes to a cytoplasmic compartment which overlaps the endoplasmic reticulum-Golgi-intermediate compartment. J Virol 74:3842–3851.
51.
Gebert S, Schmolke S, Sorg G, Flöss S, Plachter B, Stamminger T. 1997. The UL84 protein of human cytomegalovirus acts as a transdominant inhibitor of immediate-early-mediated transactivation that is able to prevent viral replication. J Virol 71:7048–7060.
52.
Scherer M, Otto V, Stump JD, Klingl S, Müller R, Reuter N, Muller YA, Sticht H, Stamminger T. 2016. Characterization of recombinant human cytomegaloviruses encoding IE1 mutants L174P and 1-382 reveals that viral targeting of PML bodies perturbs both intrinsic and innate immune responses. J Virol 90:1190–1205.
53.
Schauflinger M, Villinger C, Mertens T, Walther P, von Einem J. 2013. Analysis of human cytomegalovirus secondary envelopment by advanced electron microscopy. Cell Microbiol 15:305–314.
54.
Read C, Schauflinger M, Nikolaenko D, Walther P, von Einem J. 2019. Regulation of human cytomegalovirus secondary envelopment by a C-terminal tetralysine motif in pUL71. J Virol 93:e02244-18.
55.
Walther P, Ziegler A. 2002. Freeze substitution of high-pressure frozen samples: the visibility of biological membranes is improved when the substitution medium contains water. J Microsc 208:3–10.

Information & Contributors

Information

Published In

cover image Journal of Virology
Journal of Virology
Volume 95Number 624 February 2021
eLocator: 10.1128/jvi.02346-20
Editor: Rozanne M. Sandri-Goldin, University of California, Irvine

History

Received: 8 December 2020
Accepted: 8 December 2020
Published online: 24 February 2021

Permissions

Request permissions for this article.

Keywords

  1. ULK1
  2. autophagy
  3. human cytomegalovirus
  4. tegument protein

Contributors

Authors

Patrick König
Institute of Virology, Ulm University Medical Center, Ulm, Germany
Adriana Svrlanska
Institute of Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
Clarissa Read
Institute of Virology, Ulm University Medical Center, Ulm, Germany
Central Facility for Electron Microscopy, Ulm University, Ulm, Germany
Sabine Feichtinger
Institute of Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
Thomas Stamminger [email protected]
Institute of Virology, Ulm University Medical Center, Ulm, Germany

Editor

Rozanne M. Sandri-Goldin
Editor
University of California, Irvine

Notes

Sabine Feichtinger and Thomas Stamminger contributed equally.

Metrics & Citations

Metrics

Note: There is a 3- to 4-day delay in article usage, so article usage will not appear immediately after publication.

Citation counts come from the Crossref Cited by service.

Citations

If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. Simply select your manager software from the list below and click Download.

View Options

Figures and Media

Figures

Media

Tables

Share

Share

Share the article link

Share with email

Email a colleague

Share on social media

American Society for Microbiology ("ASM") is committed to maintaining your confidence and trust with respect to the information we collect from you on websites owned and operated by ASM ("ASM Web Sites") and other sources. This Privacy Policy sets forth the information we collect about you, how we use this information and the choices you have about how we use such information.
FIND OUT MORE about the privacy policy