Çevik, r. e., cesarec, m., da silva filipe, a., licastro, d...

49
Çevik, R. E., Cesarec, M., Da Silva Filipe, A., Licastro, D., McLauchlan, J. and Marcello, A. (2017) Hepatitis C virus NS5A targets the nucleosome assembly protein NAP1L1 to control the innate cellular response. Journal of Virology, 91(18), e00880-17. (doi:10.1128/JVI.00880-17) This is the author’s final accepted version. There may be differences between this version and the published version. You are advised to consult the publisher’s version if you wish to cite from it. http://eprints.gla.ac.uk/143947/ Deposited on: 28 August 2017 Enlighten Research publications by members of the University of Glasgow http://eprints.gla.ac.uk

Upload: others

Post on 07-Mar-2021

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Çevik, R. E., Cesarec, M., Da Silva Filipe, A., Licastro, D ...eprints.gla.ac.uk/143947/1/143947.pdf1 1 Hepatitis C virus NS5A targets the nucleosome assembly protein 2 NAP1L1 to

Çevik, R. E., Cesarec, M., Da Silva Filipe, A., Licastro, D., McLauchlan, J.

and Marcello, A. (2017) Hepatitis C virus NS5A targets the nucleosome

assembly protein NAP1L1 to control the innate cellular response. Journal

of Virology, 91(18), e00880-17. (doi:10.1128/JVI.00880-17)

This is the author’s final accepted version.

There may be differences between this version and the published version.

You are advised to consult the publisher’s version if you wish to cite from

it.

http://eprints.gla.ac.uk/143947/

Deposited on: 28 August 2017

Enlighten – Research publications by members of the University of Glasgow

http://eprints.gla.ac.uk

Page 2: Çevik, R. E., Cesarec, M., Da Silva Filipe, A., Licastro, D ...eprints.gla.ac.uk/143947/1/143947.pdf1 1 Hepatitis C virus NS5A targets the nucleosome assembly protein 2 NAP1L1 to

1

Hepatitis C virus NS5A targets the nucleosome assembly protein 1 NAP1L1 to control the innate cellular response 2 3 1,4,#Recep Emrah Çevik, 1,#Mia Cesarec, 2Ana Da Silva Filipe, 3Danilo 4 Licastro, 2John McLauchlan and 1Alessandro Marcello 5 6 1Molecular Virology Laboratory, International Centre for Genetic Engineering 7 and Biotechnology (ICGEB), Trieste, Italy 8 2MRC-University of Glasgow Centre for Virus Research, Glasgow, United 9 Kingdom 10 3CBM, Trieste, Italy 11 4Current address: The Scientific and Technological Research Council of 12 Turkey, Ankara, Turkey. 13 14 15 #These authors contributed equally to this work. 16 17 18 19 20 21 22 23 Corresponding Author: 24 25 Alessandro Marcello, PhD 26 Head, Laboratory of Molecular Virology 27 International Centre for Genetic Engineering and Biotechnology (ICGEB) 28 Padriciano, 99 29 34149 Trieste, ITALY 30 tel: +39 040 375 7384 (office) 7385 (lab) 31 fax: +39 040 226555 32 http://www.icgeb.org/molecular-virology.html 33

34

Page 3: Çevik, R. E., Cesarec, M., Da Silva Filipe, A., Licastro, D ...eprints.gla.ac.uk/143947/1/143947.pdf1 1 Hepatitis C virus NS5A targets the nucleosome assembly protein 2 NAP1L1 to

2

Abstract 35

Hepatitis C virus (HCV) is a single-stranded positive-sense RNA hepatotropic 36 virus. Despite cellular defenses, HCV is able to replicate in hepatocytes and 37 to establish a chronic infection that could lead to severe complications and 38 hepatocellular carcinoma. An important player in subverting the host 39 response to HCV infection is the viral non-structural protein NS5A that, in 40 addition to its role in replication and assembly, targets several pathways 41 involved in the cellular response to viral infection. Several unbiased screens 42 identified the nucleosome-assembly protein 1-like 1 (NAP1L1) as an 43 interaction partner of HCV NS5A. Here we confirm this interaction and map it 44 to the C-terminus of NS5A of both genotype 1 and 2. NS5A sequesters 45 NAP1L1 in the cytoplasm blocking its nuclear translocation. However, only 46 NS5A from genotype 2 HCV, but not from genotype 1, targets NAP1L1 for 47 proteosomal-mediated degradation. NAP1L1 is a nuclear chaperone involved 48 in chromatin remodeling and we demonstrate the NAP1L1-dependent 49 regulation of specific pathways involved in cellular responses to viral infection 50 and cell survival. Among those we show that lack of NAP1L1 leads to a 51 decrease of RELA protein levels and a strong defect of IRF3 TBK1/IKKε-52 mediated phosphorylation leading to inefficient RIG-I and TLR3 responses. 53 Hence, HCV is able to modulate the host cell environment by targeting 54 NAP1L1 through NS5A. 55

56

Page 4: Çevik, R. E., Cesarec, M., Da Silva Filipe, A., Licastro, D ...eprints.gla.ac.uk/143947/1/143947.pdf1 1 Hepatitis C virus NS5A targets the nucleosome assembly protein 2 NAP1L1 to

3

Importance 57 Viruses have evolved to replicate and to overcome antiviral countermeasures 58 of the infected cell. The hepatitis C virus is capable of establishing a life-long 59 chronic infection in the liver, which could develop into cirrhosis and cancer. 60 Chronic viruses are particularly able to interfere with the cellular antiviral 61 pathways by several different mechanisms. In this study we identify a novel 62 cellular target of the viral non-structural protein NS5A and demonstrate its role 63 in antiviral signaling. This factor, called nucleosome-assembly protein like 1 64 (NAP1L1), is a nuclear chaperone involved in the remodeling of chromatin 65 during transcription. When depleted, specific signaling pathways leading to 66 antiviral effectors are affected. Therefore, we provide both evidence for a 67 novel strategy of virus evasion from cellular immunity and a novel role for a 68 cellular protein, which has not been described to date. 69

70

71

Keywords: HCV; NAP1L1; innate immunity; IRF3, NF-kB; RIG-I; TLR3 72 73

Page 5: Çevik, R. E., Cesarec, M., Da Silva Filipe, A., Licastro, D ...eprints.gla.ac.uk/143947/1/143947.pdf1 1 Hepatitis C virus NS5A targets the nucleosome assembly protein 2 NAP1L1 to

4

Introduction 74 HCV is a member of the Flaviviridae family, genus Hepacivirus, with a 75

single-stranded positive-polarity RNA genome of approximately 9.6 kb (1). 76 Seven viral genotypes (1 to 7) have been identified with important differences 77 in geographical distribution, pathogenesis and response to treatment (2). The 78 HCV life cycle is entirely cytoplasmic and involves entry, uncoating and 79 translation of the viral RNA to a polyprotein that is processed by proteolysis 80 into 3 structural (Core, E1 and E2) and 7 non-structural proteins (p7, NS2, 81 NS3, NS4A, NS4B, NS5A, NS5B). Viral RNA translation, replication and 82 subsequent steps of particle assembly and release takes place associated 83 with remodelled intracellular membranes and lipid droplets (LD) (3-5). Non-84 structural proteins are required for RNA replication, with NS3-NS4A being the 85 helicase/proteinase and NS5B being the viral RNA-dependent RNA 86 polymerase. In addition, HCV non-structural proteins have also been 87 implicated in perturbing cell signalling and mediating immune evasion. Among 88 them, NS5A has been implicated in subverting several cellular pathways and 89 as a candidate viral oncogene. 90

NS5A is a 447aa (gt-2a) phosphoprotein that is found associated to ER 91 membranes through an N-terminal amphipathic helix (aa 1-27). The rest of the 92 polypeptide is hydrophilic and consists of a large amino-terminal domain I 93 followed by two smaller, more variable domains II and III. Domains I and II are 94 essential for viral genome replication while domain III is dispensable for 95 genome replication, but is required for viral particle assembly via interaction 96 with Core (6-8). A cluster of phosphorylated Serine residues at position 97 452/454/457 (strain JFH1 gt-2a) at the very C-terminal end of NS5A is 98 responsible for this interaction. In addition to a direct role in HCV genome 99 replication and assembly, NS5A makes an important contribution to 100 modulating the host cell environment. NS5A is a promiscuous protein and 101 interacts with several host factors thus affecting different signalling pathways 102 that control cell cycle, apoptosis and the interferon response to viral infection 103 (reviewed in: (9, 10)). 104

Chronic infection with HCV is a major risk factor for the development of 105 hepatocellular carcinoma (HCC) (11). The leading hypothesis is that 106 malignant transformation of hepatocytes occurs through increased liver cell 107

Page 6: Çevik, R. E., Cesarec, M., Da Silva Filipe, A., Licastro, D ...eprints.gla.ac.uk/143947/1/143947.pdf1 1 Hepatitis C virus NS5A targets the nucleosome assembly protein 2 NAP1L1 to

5

turnover induced by chronic liver injury and regeneration, in a context of 108 inflammation and oxidative DNA damage. However, increasing experimental 109 evidence suggests that HCV might also contribute to malignant transformation 110 of hepatocytes through the direct action of viral proteins on cellular 111 transformation pathways (12). Liver-specific expression in transgenic mice of 112 the full viral polyprotein (Core to NS5B) at low levels, comparable to those 113 found in patients, has been shown to induce HCC without inflammation (13). 114 Mice transgenic for NS5A alone may also develop liver cancer, depending on 115 the genetic background of the mice (14, 15). Furthermore, expression of 116 NS5A in NIH3T3 fibroblasts promoted anchorage-independent growth and 117 tumour formation in nude mice (16, 17). These data support a direct role of 118 HCV proteins, and NS5A in particular, in the development of HCC. 119

In a recent attempt to identify host factors that associate with a number 120 of innate immune-modulating viral proteins, Pichlmair et al. screened novel 121 proteins that interact with HCV NS5A (18). Careful inspection of the data led 122 to the unexpected observation that NS5A interacted with the human 123 nucleosome assembly protein-like 1 (NAP1L1 or hNAP1). The interaction of 124 NS5A with NAP1L1, or with the highly homologous NAP1L4, was 125 independently identified in at least another three independent reports (19-21). 126

NAP1L1 was originally identified in HeLa cells as the human homolog 127 of the yeast nucleosome assembly protein 1 (NAP-1) (22). NAP1L1 is a 128 391aa polypeptide characterized by nuclear import/export sequences and 129 histone and protein binding domains (23). NAP1L1 is a chaperone and 130 nucleo-cytoplasmic shuttling factor that facilitates the delivery and 131 incorporation of two histone H2A-H2B dimers to complete the nucleosome 132 (reviewed in (24-26)). NAP1L1 has been involved in the regulation of 133 transcription, cell-cycle progression, incorporation and exchange of histone 134 variants and promotion of nucleosome sliding. In addition, NAP1L1 has been 135 shown to interact with several host and viral factors including the coactivator 136 p300 and E2 of papillomaviruses (27-29). NAP1L1 interacts with the human 137 immunodeficiency virus type 1 (HIV-1) Tat transactivator and enhances HIV-1 138 trans-activation (30, 31). NAP1L1 family proteins are localized in the 139 cytoplasm, but inhibition of nuclear export results in their accumulation in the 140 nuclei indicating a shuttling activity that has been implicated also in the 141

Page 7: Çevik, R. E., Cesarec, M., Da Silva Filipe, A., Licastro, D ...eprints.gla.ac.uk/143947/1/143947.pdf1 1 Hepatitis C virus NS5A targets the nucleosome assembly protein 2 NAP1L1 to

6

delivery of histones to the nucleus as part of their chaperoning activity. 142 NAP1L1 has been involved in the process of nucleosome depletion during 143 embryonic stem cell differentiation (32). Knockdown of NAP1L1 enhanced the 144 differentiation of iPSC into functional cardiomyocytes (33). NAP1L1 145 expression has also been shown to be elevated in several cancers (34-36). 146

In this work we confirm the interaction of NS5A with NAP1L1 and its 147 co-localization in the cytoplasm of cells replicating HCV. The interaction could 148 be mapped to the carboxy-terminus of domain III of NS5A, which is shared 149 among all HCV genotypes. However, only NS5A from genotype 2, but not 150 from genotype 1, targets NAP1L1 for proteosomal-mediated degradation. 151 RNAseq analysis of NAP1L1-depleted cells shows dis-regulation of a number 152 of genes from pathways of innate immunity and cell survival. Among those, 153 we show that depletion of NAP1L1 leads to the down-modulation of NF-κB 154 and to a strong down-regulation of IRF3 phosphorylation mediated by the 155 kinase TBK1/IKKε. Both the RIG-I and TLR3 pathways were affected by 156 NAP1L1 depletion. We conclude that HCV is able to modulate the host cell 157 environment by targeting NAP1L1 through NS5A. We believe this may 158 represent a novel strategy deployed by HCV to evade from cellular antiviral 159 responses and possibly to maintain a chronic infection thus contributing to the 160 development of HCC. 161 162 163 164

165

Page 8: Çevik, R. E., Cesarec, M., Da Silva Filipe, A., Licastro, D ...eprints.gla.ac.uk/143947/1/143947.pdf1 1 Hepatitis C virus NS5A targets the nucleosome assembly protein 2 NAP1L1 to

7

Results 166 167 HCV NS5A binds NAP1L1 168

In independent reports, the non-structural protein 5A (NS5A) from the 169 human hepatitis C virus (HCV) has been repeatedly found to interact with the 170 nucleosome assembly protein NAP1L1 (18-21). However, such reports didn’t 171 map the interaction or conducted additional functional studies. To confirm the 172 interaction we generated a full-length flag-tagged f-NS5A (N-terminus) derived 173 from JFH1 genotype 2a (gt2a). Co-transfection of f-NS5A with HA-NAP1L1 in 174 HEK 293T cells followed by anti-flag affinity chromatography resulted in the 175 detection of HA-NAP1L1 (Figure 1A). As a positive control for the interaction 176 we used the human immunodeficiency virus Tat protein, which has been 177 previously described to bind NAP1L1 (30, 31). To study the interaction of 178 NS5A with NAP1L1 at the endogenous levels we took advantage of the JFH1 179 subgenomic replicon (SGR-JFH1/Luc), which efficiently replicates in 180 hepatocytes. As shown in Figure 1B, the interaction was preserved, albeit with 181 a very low efficiency of immunoprecipitation, which could be explained by an 182 effect of NS5A on the stability of the protein (see below). Furthermore, 183 extensive colocalization of the two proteins was observed in the cytoplasm 184 (Figure 1C). Interestingly, the extent of the co-localization increased from 185 approximately 10% of NS5A expressing cells at 48 hpe to more than 70% at 186 72 hpe (not shown). We repeated the experiment with a sub-genomic replicon 187 that expresses also HCV Core (Luc-JFH1ΔE1/E2) (37). In this case NS5A 188 drives the localization of NAP1L1 to subcellular locations reminiscent of lipid 189 droplets (Figure 1C, lower panels). However, the interaction between NS5A 190 and Core was not required for the NS5A to bind NAP1L1, as demonstrated in 191 the experiments conducted in the absence of Core (Figure 1C top panels and 192 Figure 1AB). 193 194 HCV NS5A from gt2 mediates NAP1L1 proteosomal degradation 195

In order to investigate if the interaction with NAP1L1 was shared by 196 NS5A from other HCV genotypes we performed an IP with genotype 1 (gt1) 197 Con1- and H77-derived proteins. Surprisingly, NS5A from gt1 isolates was 198 able to IP higher levels of NAP1L1 compared to JFH1 NS5A (Figure 2A). 199

Page 9: Çevik, R. E., Cesarec, M., Da Silva Filipe, A., Licastro, D ...eprints.gla.ac.uk/143947/1/143947.pdf1 1 Hepatitis C virus NS5A targets the nucleosome assembly protein 2 NAP1L1 to

8

Furthermore, co-transfection of HA-NAP1L1 with increasing amounts of JFH1 200 NS5A (gt2) resulted in a decrease of NAP1L1 levels, while Con1 NS5A (gt1) 201 did not (Figure 2B). Hence, NS5A from JFH1 (gt2), but not those from Con1 202 or H77 (gt1), resulted in the degradation of NAP1L1. Treatment with the 203 inhibitor MG132 partially rescued the JFH1 NS5A degradation of NAP1L1 204 indicating proteasome involvement (Figure 2C). Finally, cycloheximide 205 treatment of SGR-JFH1/Luc transfected cells showed a progressive decrease 206 of endogenous NAP1L1, although less efficiently than transfecting NS5A 207 alone (Figure 2B), possibly related to the different levels of NS5A expression 208 in the two experimental conditions. Degradation of NS5A could be rescued by 209 MG132, further highlighting the targeting of NAP1L1 for proteosomal-210 mediated degradation also at physiological levels during HCV replication 211 (Figure 2D). The major difference between JFH1-derived NS5A and those of 212 the other HCV genotypes resides in a 20 amino acids insertion at the carboxy-213 terminal end of the protein (Figure 2E), which also accounts for the 214 differences of molecular weight observed (Figure 2A). 215

We conclude that NAP1L1 binding is shared among NS5A derived from 216 g1 and gt2 genotypes, but gt2 NS5A has the additional feature of being able 217 to target NAP1L1 for proteosomal-mediated degradation. 218

219 The carboxy-terminus of NS5A is required for NAP1L1 binding 220

In order to map the interaction, NS5A from JFH1 was divided in three 221 domains: Domain I (D1, nt. 2003-2189), Domain II (D2, nt. 2226-2314), 222 Domain III (D3, nt. 2328-2442) all linked in various combinations to the N-223 terminal amphipatic helix for membrane tethering, and flag-tagged at the N-224 terminus (Figure 3A). As shown in Figure 3B, only constructs maintaining D3 225 could interact with NAP1L1. Therefore, NS5A was further truncated from the 226 C-terminus to generate proteins with progressive deletions. With this 227 approach we found that deletions starting from the acidic motif 458-EEDD-461 228 (AH-D3.5 in Figure 3C) completely abolished the interaction with NAP1L1. 229 Since the acidic motif is a perfect consensus for casein kinase 2 mediated 230 Serine phosphorylation (CK2: S-D/E-X-E/D) we reasoned that NAP1L1 231 interacts with the C-terminal cluster of Serines already implicated in the 232 interaction with HCV Core (6-8). To this end, we tested both the delB deletion 233

Page 10: Çevik, R. E., Cesarec, M., Da Silva Filipe, A., Licastro, D ...eprints.gla.ac.uk/143947/1/143947.pdf1 1 Hepatitis C virus NS5A targets the nucleosome assembly protein 2 NAP1L1 to

9

mutant of aa 2419-2433 (residues are numbered according to the positions 234 within the original JFH1 polyprotein corresponding to aa 443-457 of NS5A) (7) 235 and the triple mutant of cluster 3B (CL3B/SA or m2) (corresponding to 236 S2428/2430/2433A, aa S452/454/457A of NS5A) (6). Co-IP analysis showed 237 loss of interaction for both mutants (Figure 3D) further suggesting that these 238 residues are crucial for NAP1L1 interaction. To further demonstrate that the 239 co-localization requires the interaction of NS5A with NAP1L1 through the C-240 terminal Serine-rich region we exploited two sub-genomic constructs SGR-241 delB, and SGR-m2 (6, 7). NS5A from both constructs localized in clusters in 242 the cytoplasm, like the wild-type replicon, but did not co-localize with NAP1L1, 243 which remained diffused in the cytoplasm (Figure 3E). We conclude that 244 NAP1L1 binds to the same cluster of serine residues at the carboxy-terminus 245 of NS5A as the HCV core protein. 246 247 NAP1L1 is not required for HCV replication and infectivity in Huh7 cells 248

In order to assess the potential role of NAP1L1 in HCV replication we 249 overexpressed the EYFP-tagged version of NAP1L1 in Huh7-Lunet cells by 250 lentiviral vector (LV) transduction. NAP1L1-EYFP had the expected 251 cytoplasmic localization and co-localized with NS5A when cells were 252 transfected with SGR-JFH1 RNA (Figure 4A). However, there was no 253 difference between cells overexpressing NAP1L1-EYFP or EYFP alone in the 254 levels of luciferase from SGR-JFH/Luc, which is a measure of HCV genome 255 replication (Figure 4B). Next, we efficiently depleted NAP1L1 by lentivectors 256 delivering a specific shRNA (Figure 4C). Again, Huh7-Lunet cells replicated 257 SGR-JFH/Luc in conditions of NAP1L1 depletion as well as in mock 258 conditions (Figure 4D). To further confirm these results, we introduced full-259 length genomic HCV JFH1 RNA into Huh7-lunet cells, which had been treated 260 with shRNA as above. Cell culture supernatants at the indicated time points 261 were used to infect naïve Huh7.5 cells to measure infectivity. As shown in 262 Figure 4E, also HCV infectivity was not affected by NAP1L1 depletion in 263 Huh7-lunet cells. Finally, we investigated the replication of HCV SGR-JFH/luc 264 carrying the m2 mutation of NS5A. As shown in Figure 4F, wild type and m2 265 replicons replicated equally well in Huh7-Lunet cells, compared to the non-266 replicative control mutated in the polymerase NS5B GND. 267

Page 11: Çevik, R. E., Cesarec, M., Da Silva Filipe, A., Licastro, D ...eprints.gla.ac.uk/143947/1/143947.pdf1 1 Hepatitis C virus NS5A targets the nucleosome assembly protein 2 NAP1L1 to

10

We took the inverse approach to investigate the effect of NS5A on 268 NAP1L1 activity. In physiological conditions in cell culture NAP1L1 is found 269 predominantly in the cytoplasm (Figure 1C). However, incubation of cells with 270 the nuclear export inhibitor Leptomycin B (LMB) results in the accumulation of 271 NAP1L1 in the nucleus (Figure 5A) (31). Therefore, we transfected cells with 272 SGR-JFH1 and its mutant m2 and monitored the localization of endogenous 273 NAP1L1 in the nucleus. As shown in Figure 5A & 5B, SGR-driven NS5A, but 274 not the m2 mutant, significantly inhibited nuclear localization of NAP1L1. To 275 further analyze this phenotype in the context of HCV gt1 we engineered the 276 corresponding m2 mutations also in Con1-derived NS5A (see Figure 2E for 277 an alignment of the carboxyterminus of NS5A from the different genotypes). 278 As shown in Figure 5C, Con1-derived NS5A m2 lost the ability to interact with 279 NAP1L1 as expected. Interestingly, both JFH1 and Con1-derived NS5A 280 equally inhibited nuclear translocation of NAP1L1 (Figure 5D & 5E). 281

We conclude that NAP1L1 is not directly involved in HCV replication 282 and infectivity, but NS5A from both genotypes affect NAP1L1 nuclear 283 localization, possibly by sequestering NAP1L1 in the cytoplasm. Hence, we 284 set to investigate the nuclear activity of NAP1L1. 285 286 Transcriptome analysis of NAP1L1-depleted cells 287 NAP1L1 is a nucleosome chaperone involved in several nuclear processes 288 including transcription. Genome-wide analysis of yNAP1-deleted 289 Saccharomyces cerevisiae showed that about 10% of all yeast open reading 290 frames changed the transcription levels more than 2-fold (38). To investigate 291 the transcriptome of hepatocytes we depleted NAP1L1 with shNAP1L1 292 (Figure 4C). Differential analysis of the shNAP1L1 trascriptome versus cells 293 transduced with the control shRNA (shCTRL) showed significant up-regulation 294 of 144 genes (fold change ≥ 2) and down-regulation of 358 genes (fold 295 change ≤ 2) with a false discovery rate of less than 0.05 (data derived from 296 the most stringent DESEQ2 statistical analysis of Supplementary Table 1, 297 which also shows the EDGR statistical analysis of the same data for 298 comparison). These numbers correspond to approximately 1% of the total 299 reads of the analysis (46623 reads) suggesting a good degree of specificity 300 for the genes regulated by NAP1L1. To validate the sequencing data we re-301

Page 12: Çevik, R. E., Cesarec, M., Da Silva Filipe, A., Licastro, D ...eprints.gla.ac.uk/143947/1/143947.pdf1 1 Hepatitis C virus NS5A targets the nucleosome assembly protein 2 NAP1L1 to

11

tested a number of modulated genes by RT PCR as shown in Figure 6A. 302 Ingenuity pathway analysis of down-regulated genes indicated that top 303 canonical pathways involved in cancer and signaling were mostly affected 304 (Supplementary Table 1). Interestingly, we also noticed down-modulation of 305 interferon-stimulated genes (ISG) such as IFITM3, GBP2 and UBD as well as 306 genes involved in interferon (IFN) transcriptional activation such as RELA 307 (p65 subunit of NF-κB), c-Jun and GEF2 (39, 40). To investigate if the 308 expression of the NAP1L1 target genes is reduced in HCV infected cells we 309 conducted a meta-analysis of published data obtained in a similar setting (41). 310 We found an overlap of 40 genes between the two analysis with 5 up-311 regulated genes, 27 down-regulated genes and 8 genes showing opposed 312 regulation. Interestingly, among the overlapping down-regulated genes we 313 found again genes implicated in innate immunity regulation such as RELB, c-314 Jun, and GEF2 (Supplementary Table 2). We also compared Huh7 cells 315 stably replicating SGR JFH-1 with the homologous cured cells for the 316 expression of some NAP1L1 regulated genes (not shown). We found that two 317 significantly down-regulated genes in the context of NAP1L1 depletion such 318 as GEF2 and IFTM1 where also down-regulated in conditions that favor HCV 319 replication. However, other genes were not affected (CFL2 and HEPACAM2) 320 or showed opposite regulation (UBD). 321

In order to explore the impact of NAP1L1 depletion on the interferon 322 response pathway we explored IFNα-mediated induction of IFITM3, GBP2 323 and UBD (as well as IFIT1, IFIT3, OASL, IL8 and CXCL11, not shown). We 324 found that these genes were not impaired in their IFN-dependent induction by 325 NAP1L1 depletion, ruling out a specific role for NAP1L1 in ISG transcriptional 326 activation (Figure 6B). Finally, in order to assess the effect of NAP1L1 327 depletion on the induction of IFNβ we needed a different cell line from Huh7-328 derived cells. To this end we used U2OS cells that maintain an intact IFN-329 signaling pathway following poly(I:C) transfection, compared to cells more 330 permissive to HCV such as Huh7-Lunet or Huh7.5 (Figure 6C) (42, 43). 331 However, as shown in Figure 5D and 5E, the IFNβ response to poly(I:C) 332 transfection in U2OS cells was completely obliterated in the absence of 333 NAP1L1, both at the mRNA and protein level. Similar results were also 334 obtained in the context of vesicular stomatitis virus infection (not shown). 335

Page 13: Çevik, R. E., Cesarec, M., Da Silva Filipe, A., Licastro, D ...eprints.gla.ac.uk/143947/1/143947.pdf1 1 Hepatitis C virus NS5A targets the nucleosome assembly protein 2 NAP1L1 to

12

These data indicate that NAP1L1 is involved in regulating pathways that lead 336 to interferon induction. 337 338 Involvement of NAP1L1 in the induction of IFN 339 Pattern recognition receptors (PRR) like RIG-I/MDA5 and TLR3 respond to 340 viral RNA agonists as well as to poly(I:C) by activating transcription factors 341 such as NF-κB and IRF3. We observed previously that the NF-κB subunit 342 RELA mRNA levels were reduced in the context of NAP1L1 depletion (Figure 343 6A) and we confirmed this also at the protein level (Figure 7A and 344 quantification Figure 7B). Following activation, RELA translocates into the 345 nucleus as a phosphorylated protein. As shown in Figure 7A (quantification in 346 Figure 7C), phosphorylation of RELA occurs 1-4 hours post polyI:C induction. 347 In conditions of NAP1L1 depletion, albeit in conditions of reduced RELA 348 protein content, phosphorylation occurs normally up to 4 hours post-349 treatment, when a slight decrease was consistently observed (Figure 7A and 350 quantification in Figure 7C). Nuclear translocation of RELA showed a 351 significant decrease in the context of NAP1L1 depletion following polyI:C 352 induction (Figure 7D and 7E). At variance with RELA, IRF3 protein levels 353 were not changed by NAP1L1 depletion (Figure 7A). However, IRF3 354 phosphorylation was profoundly affected (Figure 7A and quantification in 355 Figure 7C) as well as IRF3 nuclear translocation (Figure 7F and 7G). Finally 356 we wished to recapitulate the phenotype of NAP1L1 depletion using HCV 357 NS5A. Wild-type NS5A inhibits TBK1-mediated activation of the IFN response 358 (Figure 7H), but the activity was rescued by the NS5A mutant that cannot bind 359 NAP1L1. These results suggest that NAP1L1 depletion regulates the innate 360 immune response by down-modulating RELA protein levels and by inhibiting 361 IRF3 phosphorylation. 362 363 Molecular basis of NAP1L1 depletion on IRF3 phosphorylation 364 IRF3 phosphorylation is the consequence of a complex series of molecular 365 events (see diagram in Figure 8A). PRRs such as RIG-I recognize the RNA 366 agonist in the cytoplasm and bind the adaptor protein IPS-1/MAVS to trigger 367 the downstream kinases TBK1/IKKε, which then phosphorylate IRF3. TLR3 368 instead recognizes the RNA agonist within endosomes and signals through 369

Page 14: Çevik, R. E., Cesarec, M., Da Silva Filipe, A., Licastro, D ...eprints.gla.ac.uk/143947/1/143947.pdf1 1 Hepatitis C virus NS5A targets the nucleosome assembly protein 2 NAP1L1 to

13

the adaptor TRIF to induce IRF3 phosphorylation. Since NAP1L1 depletion 370 could affect each of these steps, we proceeded to dissect the whole signaling 371 pathway. First, we took advantage of the constitutively active phosphomimetic 372 IRF3-5D (44). Consistent with our interpretation, induction of IFNβ by IRF3-5D 373 was not affected by NAP1L1 depletion (Figures 8B & 8C). Conversely, 374 depletion of NAP1L1 resulted in the inhibition of each step of the RIG-I 375 pathway, from MAVS down to TBK1/IKKε kinases. With a similar approach, 376 we could demonstrate that the TLR3 adaptor protein TRIF activity is inhibited 377 by NAP1L1 depletion (Figure 9A). To further investigate this pathway we 378 reconstituted the TLR3 pathway in Huh7-lunet cells depleted for NAP1L1 379 (Figures 9B & 9C). Stimulation with exogenous p(I:C) induced high levels of 380 the ISG IFIT1 (interferon induced protein with tetratricopeptide repeats 1), 381 which was severely affected by NAP1L1 depletion (Figure 9D). We conclude 382 that NAP1L1 controls both arms of innate sensing at the IRF3 crossroad. 383 384 385

386

Page 15: Çevik, R. E., Cesarec, M., Da Silva Filipe, A., Licastro, D ...eprints.gla.ac.uk/143947/1/143947.pdf1 1 Hepatitis C virus NS5A targets the nucleosome assembly protein 2 NAP1L1 to

14

Discussion 387 388 The co-evolution of viruses with their host results in a number of defense 389 strategies and countermeasures. Particularly for chronic infections, where the 390 virus persists for long periods of time, a delicate equilibrium is established to 391 permit limited virus replication in the context of a permissive cellular 392 environment. HCV is highly successful at establishing a chronic infection, with 393 about 80% of patients that become chronically infected. In this work we 394 describe several lines of evidence that identify NAP1L1 as a key cellular 395 effector of innate sensing and propose a novel mechanism that the virus 396 deploys to subvert innate immunity in infected hepatocytes. 397 First, we confirmed that NAP1L1 is a bona fide interactor of NS5A. These 398 data are in support of a series of independent observations from other groups 399 that indicated NAP1L1 and/or NAP1L4 as binding partners for NS5A, but 400 failed to identify a functional role (18-21). We mapped the interaction at the 401 extreme carboxy-terminus of NS5A, in a conserved motif encompassing three 402 Serine residues that have been implicated in the interaction of NS5A with 403 Core, which is essential for virus assembly, but dispensable for HCV genome 404 replication (Figure 4F) (6-8). Interestingly, NAP1L1 has also been identified as 405 a binding partner of Core in the same proteomic screenings that identified it 406 as a binding partner of NS5A (18, 21). Indeed, in the presence of Core, we 407 could visualize NAP1L1 on the surface of lipid droplets together with NS5A 408 and Core. However, interaction with Core appears not to be essential for 409 NAP1L1 and NS5A interaction, since experiments conducted in the absence 410 of Core showed efficient interaction and co-localization. Therefore, Core and 411 NAP1L1 bind independently the same region of NS5A. Unfortunately, 412 mutagenesis of the binding motif in NS5A results in the disruption of both 413 Core and NAP1L1 interactions and in a defect in assembly for viruses 414 generated with these mutations, thus precluding their use unless these two 415 interactions are uncoupled, if at all possible. 416 Next we questioned the functional role of the interaction. We discovered that 417 NS5A from genotype 2 was able to bind and degrade NAP1L1 through a 418 proteosomal-dependent mechanism. NS5A from genotype 1 was also able to 419 bind efficiently, but unable to degrade NAP1L1. Furthermore, wild-type NS5A 420

Page 16: Çevik, R. E., Cesarec, M., Da Silva Filipe, A., Licastro, D ...eprints.gla.ac.uk/143947/1/143947.pdf1 1 Hepatitis C virus NS5A targets the nucleosome assembly protein 2 NAP1L1 to

15

from both genotypes, but not the mutated version defective for NAP1L1 421 binding, inhibited the nuclear re-localization of NAP1L1. It is well established 422 that acutely infected patients respond well to IFN therapy while in chronically 423 infected ones the response to IFN is variable and depends on the viral 424 genotype (39, 45). Patients infected with HCV genotype 2 and 3 show a better 425 response compared to genotypes 1 and 4, which correlates with higher levels 426 hepatic ISG expression in HCV genotype 1 and 4 infected patient liver before 427 therapy (46-49). The ability of different genotypes to subvert the innate 428 response has been ascribed to the genetic variability of NS3 and NS5A, which 429 could affect their known activities in targeting innate immunity effectors such 430 as MAVS (NS3) or PKR and possibly NAP1L1 (NS5A). For example, the 431 levels of MAVS cleavage in vivo showed a positive correlation with the 432 decrease of the interferon response (50). In that report, HCV genotypes 2 and 433 3 were more efficient than genotype 1 and 4 in MAVS cleavage and blockage 434 of the endogenous IFN system, which determines the response to the 435 treatment with pegylated IFN and ribavirin. Therefore we could speculate that 436 also the differential ability of NS5A from genotype 1 (binding of NAP1L1) and 437 2 (binding and degradation of NAP1L1) contributes to the observed responses 438 following IFN treatment. However, the interaction of NS5A and NAP1L1 439 appears functionally dominant over JFH1 dependent degradation. In fact, 440 NS5A from the two genotypes were observed to be equally efficient in 441 blocking NAP1L1 translocation into the nucleus (Figure 5C and D). 442 Depletion of NAP1L1 by shRNA, which recapitulates NS5A-mediated 443 inhibition, resulted in the modulation of several genes at the transcription 444 level. In particular, we noticed down-modulation of interferon-stimulated genes 445 (ISG), such as GBP2, IFITM3 and UBD, and genes involved in the 446 transcriptional activation of IFNβ such as RELA, the p65 subunit of NFκB. 447 Indeed, depletion of NAP1L1 strongly affected polyI:C mediated induction of 448 IFNβ, while no effect was observed for IFNβ induction of ISGs. These findings 449 restrict the functional role of NAP1L1 on HCV to the modulation of the innate 450 sensing of the virus in infected cells. It is worthwhile noting that the latter 451 experiments were conducted in U2OS cells, which are competent for the 452 interferon response (43). Huh7-derived cell lines adapted for HCV growth are 453 instead defective for the sensing of HCV replication (see Figure 5C) (42, 43). 454

Page 17: Çevik, R. E., Cesarec, M., Da Silva Filipe, A., Licastro, D ...eprints.gla.ac.uk/143947/1/143947.pdf1 1 Hepatitis C virus NS5A targets the nucleosome assembly protein 2 NAP1L1 to

16

Hence, NS5A control of NAP1L1 results in the inhibition of the cellular innate 455 response pathway leading to IFNβ transcription, which is appreciable only in 456 cells that maintain this pathway active. This observation clearly explains why 457 we failed to observe any effect of NAP1L1 overexpression or depletion on 458 HCV replication and infectivity in Huh7 derived cells. 459 HCV infection triggers a number of innate immune pathways (39). The 5’-ppp 460 and the poly U/UC sequence of viral RNA are potent activators of RIG-I 461 signaling through MAVS/IPS-1 leading to the activation of the transcription 462 factors IRF3 and NF-κB, which in turn drive transcription of IFNβ. HCV 463 infection is also monitored in the host by the Toll-like receptors (TLRs). Viral 464 RNA activates TLR3 and signals are transduced through the TIR-domain 465 containing adapter-inducing IFNβ (TRIF) leading to activation of the 466 transcription factors IRF3 and NFκB for the induction of innate immunity (51, 467 52). Another recently described sensor protein for HCV is the antiviral protein 468 kinase R (PKR). Kinase-independent PKR signaling activates specific ISGs 469 and IFNβ early during HCV infection (53). This signaling induces protein–470 protein interactions between PKR and MAVS, which have been previously 471 described as a signaling adaptor protein also for PKR (54-56). Interestingly, 472 all these pathways converge on the activation of transcription factors NFκB 473 and/or IRF3. We found that depletion of NAP1L1 not only results in a 474 significant reduction of the mRNA and protein levels of NFκB, but also 475 severely impairs IRF3 phosphorylation. Furthermore, nuclear translocation of 476 both NFκB and IRF3 following polyI:C stimulation of RIG-I is reduced when 477 NAP1L1 is depleted. Therefore, NAP1L1 affects a step leading to IRF3 478 phosphorylation, a conclusion that is further substantiated by an experiment 479 where IFNβ expression induced by a constitutively active phosphorylated form 480 of IRF3 (IRF3-5D) remains unaffected by NAP1L1 depletion, which rules out 481 inhibitory effects downstream of IRF3 phosphorylation. In order to understand 482 at which step NAP1L1 depletion was inhibiting the pathway upstream of IRF3 483 we proceeded to dissect the major RIG-I dependent axis leading to IFNβ 484 expression. We could consistently observe that depletion of NAP1L1 reduces 485 INFβ expression induced by activated RIG-I, MAVS, TBK1 and IKKε. Hence, 486 we can hypothesize that NAP1L1 depletion affects the RIG-I pathway at the 487 level of TBK1/IKKε phosphorylation of IRF3. As mentioned above, at this step 488

Page 18: Çevik, R. E., Cesarec, M., Da Silva Filipe, A., Licastro, D ...eprints.gla.ac.uk/143947/1/143947.pdf1 1 Hepatitis C virus NS5A targets the nucleosome assembly protein 2 NAP1L1 to

17

converge all three pathways of IFNβ activation by HCV: RIG-I, TLR3 and 489 PKR. Indeed, the TLR3 pathway was also inhibited by NAP1L1 depletion. 490 Finally, to demonstrate that NS5A targeting of NAP1L1 is sufficient to inhibit 491 this phosphorylation step we show that wild type NS5A, but not NS5A mutants 492 defective for NAP1L1 binding, are able to inhibit IFNβ induction by TBK1. 493 The master viral regulator of the HCV immune evasion program is the HCV 494 NS3/4A protease. To regulate innate immune signaling, NS3/4A utilizes its 495 protease domain to cleave key innate immune signaling adaptor proteins such 496 as MAVS (57-60) and TRIF (61, 62). However, hepatitis A virus, a 497 hepatotropic virus, which does not usually become chronic, encodes a 498 protease that also cleaves MAVS (63). Thus, MAVS/TRIF cleavage is 499 probably necessary but not sufficient for viral chronicity. HCV also regulates 500 PKR activity during viral infection. HCV has several PKR-inactivation 501 strategies that probably contribute to viral persistence in addition to NS3-502 NS4A cleavage of MAVS, which depend on the activity of NS5A and E2 (64-503 66). In this work we add another mechanism that could be deployed by HCV 504 to subvert the host response to infection. We could not fully recapitulate the 505 functional role of NAP1L1 in the infectious HCV life cycle due to a number of 506 limitations of our experimental tools. First, mutations in NS5A that abolish 507 NAP1L1 binding are not compatible with a fully infectious virus (6-8). Second, 508 Huh7-derived cells that support HCV replication are impaired in the interferon 509 response (Figure 6C). Third, Infection with full-length HCV would lead to 510 several additional mechanisms of inhibition of the interferon response in 511 addition to the NS5A/NAP1L1 axis, such as NS3/4A targeting MAVS and 512 TRIF or NS2 inhibiting TBK/IKKε (39, 67). However, notwithstanding the 513 limitations outlined above, we could clearly identify NAP1L1 as a target for 514 HCV NS5A and define its novel role in the innate response. To note, a very 515 recent report confirmed the interaction of HCV NS5A with NAP1L1 and 516 showed some effect of NAP1L1 depletion on viral replication in the context of 517 cells stably harboring a SGR HCV (68). It is possible that chronically 518 replicating HCV is somehow more sensitive to NAP1L1 depletion. 519 NAP1L1 is a cytoplasmic protein unless stimulated to translocate into the 520 nucleus. Therefore we initially hypothesized a direct participation of NAP1L1 521 in the TBK1/IKKε kinase complex that phosphorylates IRF3. However, we 522

Page 19: Çevik, R. E., Cesarec, M., Da Silva Filipe, A., Licastro, D ...eprints.gla.ac.uk/143947/1/143947.pdf1 1 Hepatitis C virus NS5A targets the nucleosome assembly protein 2 NAP1L1 to

18

failed to immunoprecipitate NAP1L1 together with TBK1 and IKKε (not 523 shown). Most probably, the activity of NAP1L1 is at the transcriptional level 524 instead, as we observed for the down-modulation of NFκB. NAP1L1 depletion 525 and/or sequestration in the cytoplasm would result in the decrease of an as 526 yet unknown cellular factor that promotes TBK1/IKKε phosphorylation of IRF3. 527 Targeting general transcription factors to subvert innate sensing is not 528 unusual. Several examples of viral proteins that target host cell transcription 529 have been described. NSs from La Crosse encephalitis virus acts 530 downstream of IRF3 by specifically inhibiting RNA polymerase II (RNAPII) 531 mediated transcription by proteasomal degradation of the RBP1 subunit (69). 532 Other Bunyaviruses interfere with RNAPII CTD Ser2 phosphorylation or target 533 TFIIH (70-72). The NS1 of influenza A H3N2 subtype mimics a histone tail 534 and suppresses hPAF1C-mediated transcriptional elongation of a subset of 535 inducible genes involved in the antiviral response (73). Therefore, targeting 536 transcription appears a generalized strategy to fine-tune transcriptional 537 programs triggered by infection, which puts the cell in the optimal state to 538 overcome the invaders’ attack. HCV makes no exception and we demonstrate 539 here that by targeting NAP1L1, it is able to control a subset of host genes, 540 including key components of the antiviral innate sensing. It will be important to 541 investigate at the transcriptional level the mechanism of action of NAP1L1 and 542 to identify the factor(s) involved in TBK1/IKKε IRF3 phosphorylation that are 543 down-modulated when NAP1L1 is depleted. 544 545

546

Page 20: Çevik, R. E., Cesarec, M., Da Silva Filipe, A., Licastro, D ...eprints.gla.ac.uk/143947/1/143947.pdf1 1 Hepatitis C virus NS5A targets the nucleosome assembly protein 2 NAP1L1 to

19

Materials and Methods 547 548 Cells and viruses 549 The human hepatocarcinoma Huh7 cell line, its derivative Huh7-lunet kindly 550 provided by Ralf Bartenschlager (University of Heidelberg, Germany) (74), the 551 HEK293T cell line and the osteosarcoma cell line U2OS were cultured in 552 Dulbecco’s modified Eagle’s medium (DMEM) supplemented with 10% fetal 553 bovine serum (FBS) and antibiotics. Cell cultures were maintained at 37 °C 554 under 5% CO2. Cells were routinely tested for mycoplasma contamination. 555 In vitro transcribed JFH1 RNA was introduced into Huh7-lunet cells by 556 electroporation as described (75). The supernatants from cells electroporated 557 with JFH1 RNA were removed at required time points and used to infect 558 monolayers of naive Huh7.5 cells. Infected cells were detected at three days 559 post-inoculation by indirect immunofluorescence using a polyclonal NS5A 560 antiserum. The tissue culture 50% infectious dose (TCID50) was determined 561 by limiting dilution assay (76). 562 IFNα was obtained from the Biotechnology Development Group of the ICGEB. 563 564 Plasmids 565 Plasmid pJFH1 was provided by T. Wakita (77). Plasmids encoding SGR-566 JFH1/Luc and the non-replicating control SGR-JFH1/Luc-GND were 567 described previously (78). Plasmid pFKLuc-JFH/ΔE1-E2 was provided by R. 568 Bartenschlager (37), plasmid SGR-JFH/m2 was provided by T. Masaki (6) 569 and plasmid SGR-JFH1/delB by T. Tellinghuisen (7). The pHA-NAP1L1 and 570 pNAP1L1-EYFP expression vectors were described previously (30, 31). 571 pFLAG CMV-2 (Sigma-Aldrich) was used as backbone for all NS5A 572 constructs. Flag tagged NS5A from HCV JFH1, Con1 and H77 were cloned 573 by PCR of the corresponding HCV genotypes. Mutagenesis of NS5A (JFH1 574 and Con1) was performed by PCR, detailed information is available on 575 request. The reporter plasmid carrying the firefly luciferase (Fluc) gene under 576 the control of the IFNβ promoter (pIFNβ-Luc) was provided by J. Jung (44). T. 577 Fujita kindly provided FLAG-tagged expression vectors for RIG-I, RIG-I-N, 578 TBK1, IKKε, IPS-1/MAVS and HA-IRF3-5D. The control pCMV-Renilla was 579 from Promega. 580

Page 21: Çevik, R. E., Cesarec, M., Da Silva Filipe, A., Licastro, D ...eprints.gla.ac.uk/143947/1/143947.pdf1 1 Hepatitis C virus NS5A targets the nucleosome assembly protein 2 NAP1L1 to

20

581 Lentivector production and shRNA delivery 582 Overexpression of NAP1-EYFP and control EYFP was obtained by lentivector 583 transduction on a pWPI backbone with blasticidin resistance (BLR) kindly 584 provided by D. Trono. Lentiviral silencing vectors were derived from pLKO.1 585 TRC (Addgene). The control short-hairpin RNA (shRNA) was the pLKO.1 586 scramble from Addgene while for NAP1L1 targeting a specific targeting 587 sequence was designed and cloned into pLKO.1 TRC (shNAP1L1) using the 588 following oligonucleotides: 589 5’-ccggcctattctgaagcacttgaaactgcagtttcaagtgcttcagaataggtttttg -3’ and 590 5’- ggataagacttcgtgaactttgacgtcaaagttcacgaagtcttatccaaaaacttaa -3’. 591 A LV for TLR3 reconstitution together with its GFP control was obtained from 592 Sam Wilson (MRC – University of Glasgow Centre for Virus Research). 593 Packaging in HEK 293T was performed according to standard procedures 594 using the packaging plasmid psPAX2 and pMD2.G (Addgene). Cells’ 595 supernatants were filtered and kept at -80 °C in small aliquots until use. 596 597 In vitro transcription and electroporation of HCV SGR RNA 598 The HCV SGR constructs were linearized with XbaI and treated with mung-599 bean nuclease as described previously (79). RNA was transcribed in vitro 600 from linearized constructs using the MEGAscript T7 kit (Ambion). Synthesized 601 RNA was treated with DNase I and transfected into cells by electroporation. 602 PolyI:C (polyinosinic : polycytidylic acid sodium salt; Invivogen) was also 603 transfected into cells by lipofection (Lipofectamine PLUS, Life Technologies) 604 according to manufacturer’s instructons. 605 606 Western-blotting, immunoprecipitation and immunofluorescence 607 Indirect immunofluorescence analysis (IF) and Western-blotting (WB) were 608 performed essentially as previously described (80). Immunoprecipitation (IP) 609 was performed by lysis of cells in RIPA buffer (50 mM Tris HCl pH7.5, 150 610 mM NaCl, 1% NP-40, 1% SDS, 1M PMSF, 1mM EDTA and proteinase 611 inhibitors (cOmplete Mini, Roche). Lysates were cleared by centrifugation and 612 incubated with anti-FLAG M2 agarose beads (Sigma-Aldrich), or with anti-613 NAP1L1 Ab/IgG control and protein A/G agarose beads, washed several 614

Page 22: Çevik, R. E., Cesarec, M., Da Silva Filipe, A., Licastro, D ...eprints.gla.ac.uk/143947/1/143947.pdf1 1 Hepatitis C virus NS5A targets the nucleosome assembly protein 2 NAP1L1 to

21

times in RIPA and eluted in SDS-PAGE sample buffer. The following 615 antibodies were used in this study: a sheep polyclonal against NS5A kindly 616 provided by M. Harris (1:200 IF; 1:2000 WB) (81); a rabbit polyclonal against 617 human NAP1L1 (Ab33076, Abcam) (1:200 IF; 1:1000 WB; 1:100 IP); a rabbit 618 polyclonal against human IRF3 kindly provided by T. Fujita (1:100 IF); a rabbit 619 monoclonal against human IRF3 (#4302 Cell Signaling) (1:500 WB); a rabbit 620 monoclonal against phosphorylated human IRF3 (#4947 Cell Signaling) 621 (1:500 WB); a rabbit monoclonal against human NF-κB p65/RELA (#8242 Cell 622 Signaling) (1:100 IF; 1:1000 WB); a rabbit monoclonal against phosphorylated 623 human NF-κB p65/RELA (#3033 Cell Signaling) (1:1000 WB); a mouse 624 monoclonal against human β-actin conjugated with peroxidase (A3854 Sigma-625 Aldrich) (1:10000 WB); a mouse monoclonal against the FLAG tag (F1804 626 Sigma-Aldrich) (1:1000 WB); a mouse monoclonal against the HA tag 627 conjugated with peroxidase (H6533 Sigma-Aldrich) (1:10000 WB). Secondary 628 antibodies conjugated with AlexaFluor 488/594 were from Life Technologies 629 (1:500 IF) and peroxidase conjugates from Dako (1:5000 WB). 630 631 Luciferase assay and real-time quantitative reverse transcription PCR 632 Luciferase assays were conducted essentially as described previously (79, 633 80). For real-time quantitative reverse transcription PCR (qPCR) total cellular 634 RNA was extracted with the isol-RNA reagent (5 Prime) and treated with 635 DNAse I (Life Technologies). 500 ng were then reverse-transcribed with 636 random primers and M-MLV Reverse Transcriptase (Life Technologies). 637 Quantification of mRNA was obtained by real-time PCR using the Kapa Sybr 638 fast qPCR kit on a CFX96 Bio-Rad thermocycler. Primers for amplification are 639 available upon request. 640 641 Transcriptome analysis by RNAseq 642 Huh7-lunet cells were transduced with shNAP1L1 or shCTRL in triplicate and 643 incubated with puromicin for 3 days. Total RNA (Isol RNA lysis, Reagent 5 644 PRIME, Hamburg, DE) was extracted. Quality of extracted RNA was checked 645 by gel electrophoresis (ribosomal 18S and 28S), spectrophotometric analysis 646 (260/280>1.8) and Agilent bioanalyzer (RNA integrity number, RINt8). A 647

Page 23: Çevik, R. E., Cesarec, M., Da Silva Filipe, A., Licastro, D ...eprints.gla.ac.uk/143947/1/143947.pdf1 1 Hepatitis C virus NS5A targets the nucleosome assembly protein 2 NAP1L1 to

22

cDNA library of polyA-containing mRNA molecules was prepared (TruSeq, 648 Illumina) and sequenced on the Illumina Platform (Hiseq2000 4-plex run, 50 649 bp reads, about 30M reads/sample) at IGA Technology Services (Udine, 650 Italy). Raw data were subjected to quality control (FastQC) and mapped 651 against the human genome RNA reference from NCBI using CLCbio sotware. 652 The Bioconductor packages DESeq2 version 1.4.5 (82) and EdgeR (83) 653 version 3.6.2 in the framework of R software version 3.1.0 were used to 654 perform the differential gene expression analysis of mRNAseq data. Both the 655 packages are based on the negative binomial distribution (NB) to model the 656 gene reads counts and shrinkage estimator to estimate the per-gene NB 657 dispersion parameters. Specifically, we used rounded gene counts as input 658 and we estimated the per-gene NB dispersion parameter using the function 659 DESeq for DESEQ2 while, for edgeR we used the function calcNormFactors 660 with the default parameters. To detect outlier data after normalization we used 661 the R packages arrayQualityMetrix (84) and before testing differential gene 662 expression we dropped all genes with normalized counts below 14 to improve 663 testing power while maintaining type I error rates. Estimated p-values for each 664 gene were adjusted using the Benjamini-Hochberg method (85). Genes with 665 adjusted P<0.05 and absolute Logarithmic base 2 fold change > 1 were 666 selected. Data were finally analysed with the Ingenuity Pathway Analysis 667 software. The significance values for the canonical pathway across the 668 dataset shown in the Supplementary Table 1 are calculated by the Fisher’s 669 exact test right-tailed. The significance indicates the probability of association 670 of molecules from our dataset with the canonical pathway by random chance 671 alone. 672 673 Statistics 674 Three independent experiments in triplicate repeats were conducted for each 675 condition examined, unless otherwise indicated in the figure legends. Mean 676 values are shown with standard deviation and p-values, measured with a 677 paired two-tailed t-test. Only significant p-values are indicated by the asterisks 678 above the graphs (p<0.01 = ** highly significant; p<0.05 = * significant). 679 Where asterisks are missing the differences are calculated as non-significant. 680 681

Page 24: Çevik, R. E., Cesarec, M., Da Silva Filipe, A., Licastro, D ...eprints.gla.ac.uk/143947/1/143947.pdf1 1 Hepatitis C virus NS5A targets the nucleosome assembly protein 2 NAP1L1 to

23

Acknowledgments 682 Work on flaviviruses in A.M.’s laboratory is supported by the Beneficientia 683 Stiftung. The funders had no role in study design, data collection and 684 interpretation, or the decision to submit the work for publication. 685 686 References 687 1. Choo QL, Kuo G, Weiner AJ, Overby LR, Bradley DW, Houghton 688

M. 1989. Isolation of a cDNA clone derived from a blood-borne non-A, 689 non-B viral hepatitis genome. Science 244:359-362. 690

2. Smith DB, Bukh J, Kuiken C, Muerhoff AS, Rice CM, Stapleton JT, 691 Simmonds P. 2014. Expanded classification of hepatitis C virus into 7 692 genotypes and 67 subtypes: updated criteria and genotype assignment 693 web resource. Hepatology 59:318-327. 694

3. Miyanari Y, Atsuzawa K, Usuda N, Watashi K, Hishiki T, Zayas M, 695 Bartenschlager R, Wakita T, Hijikata M, Shimotohno K. 2007. The 696 lipid droplet is an important organelle for hepatitis C virus production. 697 Nat Cell Biol 9:1089-1097. 698

4. Romero-Brey I, Merz A, Chiramel A, Lee JY, Chlanda P, Haselman 699 U, Santarella-Mellwig R, Habermann A, Hoppe S, Kallis S, Walther 700 P, Antony C, Krijnse-Locker J, Bartenschlager R. 2013. Three-701 dimensional architecture and biogenesis of membrane structures 702 associated with hepatitis C virus replication. PLoS Pathog 8:e1003056. 703

5. Boulant S, Targett-Adams P, McLauchlan J. 2007. Disrupting the 704 association of hepatitis C virus core protein with lipid droplets 705 correlates with a loss in production of infectious virus. J Gen Virol 706 88:2204-2213. 707

6. Masaki T, Suzuki R, Murakami K, Aizaki H, Ishii K, Murayama A, 708 Date T, Matsuura Y, Miyamura T, Wakita T, Suzuki T. 2008. 709 Interaction of hepatitis C virus nonstructural protein 5A with core 710 protein is critical for the production of infectious virus particles. J Virol 711 82:7964-7976. 712

7. Tellinghuisen TL, Foss KL, Treadaway J. 2008. Regulation of 713 hepatitis C virion production via phosphorylation of the NS5A protein. 714 PLoS Pathog 4:e1000032. 715

Page 25: Çevik, R. E., Cesarec, M., Da Silva Filipe, A., Licastro, D ...eprints.gla.ac.uk/143947/1/143947.pdf1 1 Hepatitis C virus NS5A targets the nucleosome assembly protein 2 NAP1L1 to

24

8. Appel N, Zayas M, Miller S, Krijnse-Locker J, Schaller T, Friebe P, 716 Kallis S, Engel U, Bartenschlager R. 2008. Essential role of domain 717 III of nonstructural protein 5A for hepatitis C virus infectious particle 718 assembly. PLoS Pathog 4:e1000035. 719

9. Macdonald A, Harris M. 2004. Hepatitis C virus NS5A: tales of a 720 promiscuous protein. J Gen Virol 85:2485-2502. 721

10. Ross-Thriepland D, Harris M. 2015. Hepatitis C virus NS5A: 722 enigmatic but still promiscuous 10 years on! J Gen Virol 96:727-738. 723

11. IARC. 2012. Hepatitits C virus. A review of human carcinogens Part B: 724 Biological agents IARC Working Group on the Evaluation of 725 Carcinogenic Risks to Humans:135-168. 726

12. McGivern DR, Lemon SM. 2011. Virus-specific mechanisms of 727 carcinogenesis in hepatitis C virus associated liver cancer. Oncogene 728 30:1969-1983. 729

13. Lerat H, Honda M, Beard MR, Loesch K, Sun J, Yang Y, Okuda M, 730 Gosert R, Xiao SY, Weinman SA, Lemon SM. 2002. Steatosis and 731 liver cancer in transgenic mice expressing the structural and 732 nonstructural proteins of hepatitis C virus. Gastroenterology 122:352-733 365. 734

14. Wang AG, Lee DS, Moon HB, Kim JM, Cho KH, Choi SH, Ha HL, 735 Han YH, Kim DG, Hwang SB, Yu DY. 2009. Non-structural 5A protein 736 of hepatitis C virus induces a range of liver pathology in transgenic 737 mice. J Pathol 219:253-262. 738

15. Majumder M, Steele R, Ghosh AK, Zhou XY, Thornburg L, Ray R, 739 Phillips NJ, Ray RB. 2003. Expression of hepatitis C virus non-740 structural 5A protein in the liver of transgenic mice. FEBS Lett 555:528-741 532. 742

16. Gale M, Jr., Kwieciszewski B, Dossett M, Nakao H, Katze MG. 743 1999. Antiapoptotic and oncogenic potentials of hepatitis C virus are 744 linked to interferon resistance by viral repression of the PKR protein 745 kinase. J Virol 73:6506-6516. 746

17. Ghosh AK, Steele R, Meyer K, Ray R, Ray RB. 1999. Hepatitis C 747 virus NS5A protein modulates cell cycle regulatory genes and 748 promotes cell growth. J Gen Virol 80 ( Pt 5):1179-1183. 749

Page 26: Çevik, R. E., Cesarec, M., Da Silva Filipe, A., Licastro, D ...eprints.gla.ac.uk/143947/1/143947.pdf1 1 Hepatitis C virus NS5A targets the nucleosome assembly protein 2 NAP1L1 to

25

18. Pichlmair A, Kandasamy K, Alvisi G, Mulhern O, Sacco R, Habjan 750 M, Binder M, Stefanovic A, Eberle CA, Goncalves A, 751 Burckstummer T, Muller AC, Fauster A, Holze C, Lindsten K, 752 Goodbourn S, Kochs G, Weber F, Bartenschlager R, Bowie AG, 753 Bennett KL, Colinge J, Superti-Furga G. 2012. Viral immune 754 modulators perturb the human molecular network by common and 755 unique strategies. Nature 487:486-490. 756

19. de Chassey B, Navratil V, Tafforeau L, Hiet MS, Aublin-Gex A, 757 Agaugue S, Meiffren G, Pradezynski F, Faria BF, Chantier T, Le 758 Breton M, Pellet J, Davoust N, Mangeot PE, Chaboud A, Penin F, 759 Jacob Y, Vidalain PO, Vidal M, Andre P, Rabourdin-Combe C, 760 Lotteau V. 2008. Hepatitis C virus infection protein network. Mol Syst 761 Biol 4:230. 762

20. Chung HY, Gu M, Buehler E, MacDonald MR, Rice CM. 2014. Seed 763 sequence-matched controls reveal limitations of small interfering RNA 764 knockdown in functional and structural studies of hepatitis C virus 765 NS5A-MOBKL1B interaction. J Virol 88:11022-11033. 766

21. Ramage HR, Kumar GR, Verschueren E, Johnson JR, Von Dollen 767 J, Johnson T, Newton B, Shah P, Horner J, Krogan NJ, Ott M. 768 2015. A combined proteomics/genomics approach links hepatitis C 769 virus infection with nonsense-mediated mRNA decay. Mol Cell 57:329-770 340. 771

22. Ishimi Y, Kikuchi A. 1991. Identification and molecular cloning of 772 yeast homolog of nucleosome assembly protein I which facilitates 773 nucleosome assembly in vitro. J Biol Chem 266:7025-7029. 774

23. Park YJ, Luger K. 2006. The structure of nucleosome assembly 775 protein 1. Proc Natl Acad Sci U S A 103:1248-1253. 776

24. Park YJ, Luger K. 2006. Structure and function of nucleosome 777 assembly proteins. Biochem Cell Biol 84:549-558. 778

25. Loyola A, Almouzni G. 2004. Histone chaperones, a supporting role 779 in the limelight. Biochim Biophys Acta 1677:3-11. 780

26. Tyler JK. 2002. Chromatin assembly. Cooperation between histone 781 chaperones and ATP-dependent nucleosome remodeling machines. 782 Eur J Biochem 269:2268-2274. 783

Page 27: Çevik, R. E., Cesarec, M., Da Silva Filipe, A., Licastro, D ...eprints.gla.ac.uk/143947/1/143947.pdf1 1 Hepatitis C virus NS5A targets the nucleosome assembly protein 2 NAP1L1 to

26

27. Asahara H, Tartare-Deckert S, Nakagawa T, Ikehara T, Hirose F, 784 Hunter T, Ito T, Montminy M. 2002. Dual roles of p300 in chromatin 785 assembly and transcriptional activation in cooperation with nucleosome 786 assembly protein 1 in vitro. Mol Cell Biol 22:2974-2983. 787

28. Rehtanz M, Schmidt HM, Warthorst U, Steger G. 2004. Direct 788 interaction between nucleosome assembly protein 1 and the 789 papillomavirus E2 proteins involved in activation of transcription. Mol 790 Cell Biol 24:2153-2168. 791

29. Sharma N, Nyborg JK. 2008. The coactivators CBP/p300 and the 792 histone chaperone NAP1 promote transcription-independent 793 nucleosome eviction at the HTLV-1 promoter. Proc Natl Acad Sci U S 794 A 105:7959-7963. 795

30. Vardabasso C, Manganaro L, Lusic M, Marcello A, Giacca M. 2008. 796 The histone chaperone protein Nucleosome Assembly Protein-1 797 (hNAP-1) binds HIV-1 Tat and promotes viral transcription. 798 Retrovirology 5:8. 799

31. De Marco A, Dans PD, Knezevich A, Maiuri P, Pantano S, Marcello 800 A. 2010. Subcellular localization of the interaction between the human 801 immunodeficiency virus transactivator Tat and the nucleosome 802 assembly protein 1. Amino Acids 38:1583-1593. 803

32. Li Z, Gadue P, Chen K, Jiao Y, Tuteja G, Schug J, Li W, Kaestner 804 KH. 2012. Foxa2 and H2A.Z mediate nucleosome depletion during 805 embryonic stem cell differentiation. Cell 151:1608-1616. 806

33. Gong H, Yan Y, Fang B, Xue Y, Yin P, Li L, Zhang G, Sun X, Chen 807 Z, Ma H, Yang C, Ding Y, Yong Y, Zhu Y, Yang H, Komuro I, Ge J, 808 Zou Y. 2014. Knockdown of nucleosome assembly protein 1-like 1 809 induces mesoderm formation and cardiomyogenesis via notch 810 signaling in murine-induced pluripotent stem cells. Stem Cells 32:1759-811 1773. 812

34. Nagata T, Takahashi Y, Ishii Y, Asai S, Nishida Y, Murata A, 813 Koshinaga T, Fukuzawa M, Hamazaki M, Asami K, Ito E, Ikeda H, 814 Takamatsu H, Koike K, Kikuta A, Kuroiwa M, Watanabe A, Kosaka 815 Y, Fujita H, Miyake M, Mugishima H. 2003. Transcriptional profiling in 816

Page 28: Çevik, R. E., Cesarec, M., Da Silva Filipe, A., Licastro, D ...eprints.gla.ac.uk/143947/1/143947.pdf1 1 Hepatitis C virus NS5A targets the nucleosome assembly protein 2 NAP1L1 to

27

hepatoblastomas using high-density oligonucleotide DNA array. 817 Cancer Genet Cytogenet 145:152-160. 818

35. Kidd M, Modlin IM, Mane SM, Camp RL, Eick G, Latich I. 2006. The 819 role of genetic markers--NAP1L1, MAGE-D2, and MTA1--in defining 820 small-intestinal carcinoid neoplasia. Ann Surg Oncol 13:253-262. 821

36. Schimmack S, Taylor A, Lawrence B, Alaimo D, Schmitz-822 Winnenthal H, Buchler MW, Modlin IM, Kidd M. 2014. A mechanistic 823 role for the chromatin modulator, NAP1L1, in pancreatic 824 neuroendocrine neoplasm proliferation and metastases. Epigenetics 825 Chromatin 7:15. 826

37. Koutsoudakis G, Kaul A, Steinmann E, Kallis S, Lohmann V, 827 Pietschmann T, Bartenschlager R. 2006. Characterization of the 828 early steps of hepatitis C virus infection by using luciferase reporter 829 viruses. J Virol 80:5308-5320. 830

38. Ohkuni K, Shirahige K, Kikuchi A. 2003. Genome-wide expression 831 analysis of NAP1 in Saccharomyces cerevisiae. Biochem Biophys Res 832 Commun 306:5-9. 833

39. Horner SM, Gale M, Jr. 2013. Regulation of hepatic innate immunity 834 by hepatitis C virus. Nat Med 19:879-888. 835

40. Chiang HS, Zhao Y, Song JH, Liu S, Wang N, Terhorst C, Sharpe 836 AH, Basavappa M, Jeffrey KL, Reinecker HC. 2013. GEF-H1 837 controls microtubule-dependent sensing of nucleic acids for antiviral 838 host defenses. Nat Immunol 15:63-71. 839

41. Luna JM, Scheel TK, Danino T, Shaw KS, Mele A, Fak JJ, 840 Nishiuchi E, Takacs CN, Catanese MT, de Jong YP, Jacobson IM, 841 Rice CM, Darnell RB. 2015. Hepatitis C virus RNA functionally 842 sequesters miR-122. Cell 160:1099-1110. 843

42. Sumpter R, Jr., Loo YM, Foy E, Li K, Yoneyama M, Fujita T, Lemon 844 SM, Gale M, Jr. 2005. Regulating intracellular antiviral defense and 845 permissiveness to hepatitis C virus RNA replication through a cellular 846 RNA helicase, RIG-I. J Virol 79:2689-2699. 847

43. Miorin L, Albornoz A, Baba MM, D'Agaro P, Marcello A. 2012. 848 Formation of membrane-defined compartments by tick-borne 849

Page 29: Çevik, R. E., Cesarec, M., Da Silva Filipe, A., Licastro, D ...eprints.gla.ac.uk/143947/1/143947.pdf1 1 Hepatitis C virus NS5A targets the nucleosome assembly protein 2 NAP1L1 to

28

encephalitis virus contributes to the early delay in interferon signaling. 850 Virus Res 163:660-666. 851

44. Lin R, Genin P, Mamane Y, Hiscott J. 2000. Selective DNA binding 852 and association with the CREB binding protein coactivator contribute to 853 differential activation of alpha/beta interferon genes by interferon 854 regulatory factors 3 and 7. Mol Cell Biol 20:6342-6353. 855

45. Jaeckel E, Cornberg M, Wedemeyer H, Santantonio T, Mayer J, 856 Zankel M, Pastore G, Dietrich M, Trautwein C, Manns MP, German 857 Acute Hepatitis CTG. 2001. Treatment of acute hepatitis C with 858 interferon alfa-2b. N Engl J Med 345:1452-1457. 859

46. Sarasin-Filipowicz M, Oakeley EJ, Duong FH, Christen V, 860 Terracciano L, Filipowicz W, Heim MH. 2008. Interferon signaling 861 and treatment outcome in chronic hepatitis C. Proc Natl Acad Sci U S 862 A 105:7034-7039. 863

47. Chen L, Borozan I, Feld J, Sun J, Tannis LL, Coltescu C, 864 Heathcote J, Edwards AM, McGilvray ID. 2005. Hepatic gene 865 expression discriminates responders and nonresponders in treatment 866 of chronic hepatitis C viral infection. Gastroenterology 128:1437-1444. 867

48. Asselah T, Bieche I, Narguet S, Sabbagh A, Laurendeau I, Ripault 868 MP, Boyer N, Martinot-Peignoux M, Valla D, Vidaud M, Marcellin P. 869 2008. Liver gene expression signature to predict response to pegylated 870 interferon plus ribavirin combination therapy in patients with chronic 871 hepatitis C. Gut 57:516-524. 872

49. Dill MT, Duong FH, Vogt JE, Bibert S, Bochud PY, Terracciano L, 873 Papassotiropoulos A, Roth V, Heim MH. 2011. Interferon-induced 874 gene expression is a stronger predictor of treatment response than 875 IL28B genotype in patients with hepatitis C. Gastroenterology 876 140:1021-1031. 877

50. Bellecave P, Sarasin-Filipowicz M, Donze O, Kennel A, 878 Gouttenoire J, Meylan E, Terracciano L, Tschopp J, Sarrazin C, 879 Berg T, Moradpour D, Heim MH. 2010. Cleavage of mitochondrial 880 antiviral signaling protein in the liver of patients with chronic hepatitis C 881 correlates with a reduced activation of the endogenous interferon 882 system. Hepatology 51:1127-1136. 883

Page 30: Çevik, R. E., Cesarec, M., Da Silva Filipe, A., Licastro, D ...eprints.gla.ac.uk/143947/1/143947.pdf1 1 Hepatitis C virus NS5A targets the nucleosome assembly protein 2 NAP1L1 to

29

51. Li K, Li NL, Wei D, Pfeffer SR, Fan M, Pfeffer LM. 2012. Activation of 884 chemokine and inflammatory cytokine response in hepatitis C virus-885 infected hepatocytes depends on Toll-like receptor 3 sensing of 886 hepatitis C virus double-stranded RNA intermediates. Hepatology 887 55:666-675. 888

52. Wang N, Liang Y, Devaraj S, Wang J, Lemon SM, Li K. 2009. Toll-889 like receptor 3 mediates establishment of an antiviral state against 890 hepatitis C virus in hepatoma cells. J Virol 83:9824-9834. 891

53. Arnaud N, Dabo S, Akazawa D, Fukasawa M, Shinkai-Ouchi F, 892 Hugon J, Wakita T, Meurs EF. 2011. Hepatitis C virus reveals a novel 893 early control in acute immune response. PLoS Pathog 7:e1002289. 894

54. Kanazawa N, Kurosaki M, Sakamoto N, Enomoto N, Itsui Y, 895 Yamashiro T, Tanabe Y, Maekawa S, Nakagawa M, Chen CH, 896 Kakinuma S, Oshima S, Nakamura T, Kato T, Wakita T, Watanabe 897 M. 2004. Regulation of hepatitis C virus replication by interferon 898 regulatory factor 1. J Virol 78:9713-9720. 899

55. Kumar A, Yang YL, Flati V, Der S, Kadereit S, Deb A, Haque J, Reis 900 L, Weissmann C, Williams BR. 1997. Deficient cytokine signaling in 901 mouse embryo fibroblasts with a targeted deletion in the PKR gene: 902 role of IRF-1 and NF-kappaB. EMBO J 16:406-416. 903

56. McAllister CS, Samuel CE. 2009. The RNA-activated protein kinase 904 enhances the induction of interferon-beta and apoptosis mediated by 905 cytoplasmic RNA sensors. J Biol Chem 284:1644-1651. 906

57. Foy E, Li K, Sumpter R, Jr., Loo YM, Johnson CL, Wang C, Fish 907 PM, Yoneyama M, Fujita T, Lemon SM, Gale M, Jr. 2005. Control of 908 antiviral defenses through hepatitis C virus disruption of retinoic acid-909 inducible gene-I signaling. Proc Natl Acad Sci U S A 102:2986-2991. 910

58. Foy E, Li K, Wang C, Sumpter R, Jr., Ikeda M, Lemon SM, Gale M, 911 Jr. 2003. Regulation of interferon regulatory factor-3 by the hepatitis C 912 virus serine protease. Science 300:1145-1148. 913

59. Meylan E, Curran J, Hofmann K, Moradpour D, Binder M, 914 Bartenschlager R, Tschopp J. 2005. Cardif is an adaptor protein in 915 the RIG-I antiviral pathway and is targeted by hepatitis C virus. Nature 916 437:1167-1172. 917

Page 31: Çevik, R. E., Cesarec, M., Da Silva Filipe, A., Licastro, D ...eprints.gla.ac.uk/143947/1/143947.pdf1 1 Hepatitis C virus NS5A targets the nucleosome assembly protein 2 NAP1L1 to

30

60. Baril M, Racine ME, Penin F, Lamarre D. 2009. MAVS dimer is a 918 crucial signaling component of innate immunity and the target of 919 hepatitis C virus NS3/4A protease. J Virol 83:1299-1311. 920

61. Li K, Foy E, Ferreon JC, Nakamura M, Ferreon AC, Ikeda M, Ray 921 SC, Gale M, Jr., Lemon SM. 2005. Immune evasion by hepatitis C 922 virus NS3/4A protease-mediated cleavage of the Toll-like receptor 3 923 adaptor protein TRIF. Proc Natl Acad Sci U S A 102:2992-2997. 924

62. Lin R, Lacoste J, Nakhaei P, Sun Q, Yang L, Paz S, Wilkinson P, 925 Julkunen I, Vitour D, Meurs E, Hiscott J. 2006. Dissociation of a 926 MAVS/IPS-1/VISA/Cardif-IKKepsilon molecular complex from the 927 mitochondrial outer membrane by hepatitis C virus NS3-4A proteolytic 928 cleavage. J Virol 80:6072-6083. 929

63. Yang Y, Liang Y, Qu L, Chen Z, Yi M, Li K, Lemon SM. 2007. 930 Disruption of innate immunity due to mitochondrial targeting of a 931 picornaviral protease precursor. Proc Natl Acad Sci U S A 104:7253-932 7258. 933

64. Gale MJ, Jr., Korth MJ, Tang NM, Tan SL, Hopkins DA, Dever TE, 934 Polyak SJ, Gretch DR, Katze MG. 1997. Evidence that hepatitis C 935 virus resistance to interferon is mediated through repression of the 936 PKR protein kinase by the nonstructural 5A protein. Virology 230:217-937 227. 938

65. Taylor DR, Shi ST, Romano PR, Barber GN, Lai MM. 1999. Inhibition 939 of the interferon-inducible protein kinase PKR by HCV E2 protein. 940 Science 285:107-110. 941

66. Noguchi T, Satoh S, Noshi T, Hatada E, Fukuda R, Kawai A, Ikeda 942 S, Hijikata M, Shimotohno K. 2001. Effects of mutation in hepatitis C 943 virus nonstructural protein 5A on interferon resistance mediated by 944 inhibition of PKR kinase activity in mammalian cells. Microbiol Immunol 945 45:829-840. 946

67. Kaukinen P, Sillanpaa M, Nousiainen L, Melen K, Julkunen I. 2013. 947 Hepatitis C virus NS2 protease inhibits host cell antiviral response by 948 inhibiting IKKepsilon and TBK1 functions. J Med Virol 85:71-82. 949

Page 32: Çevik, R. E., Cesarec, M., Da Silva Filipe, A., Licastro, D ...eprints.gla.ac.uk/143947/1/143947.pdf1 1 Hepatitis C virus NS5A targets the nucleosome assembly protein 2 NAP1L1 to

31

68. Goonawardane N, Gebhardt A, Bartlett C, Pichlmair A, Harris M. 950 2017. Phosphorylation of serine 225 in hepatitis C virus NS5A 951 regulates protein-protein interactions. J Virol doi:10.1128/JVI.00805-17. 952

69. Verbruggen P, Ruf M, Blakqori G, Overby AK, Heidemann M, Eick 953 D, Weber F. 2011. Interferon antagonist NSs of La Crosse virus 954 triggers a DNA damage response-like degradation of transcribing RNA 955 polymerase II. J Biol Chem 286:3681-3692. 956

70. Le May N, Dubaele S, Proietti De Santis L, Billecocq A, Bouloy M, 957 Egly JM. 2004. TFIIH transcription factor, a target for the Rift Valley 958 hemorrhagic fever virus. Cell 116:541-550. 959

71. Le May N, Mansuroglu Z, Leger P, Josse T, Blot G, Billecocq A, 960 Flick R, Jacob Y, Bonnefoy E, Bouloy M. 2008. A SAP30 complex 961 inhibits IFN-beta expression in Rift Valley fever virus infected cells. 962 PLoS Pathog 4:e13. 963

72. Thomas D, Blakqori G, Wagner V, Banholzer M, Kessler N, Elliott 964 RM, Haller O, Weber F. 2004. Inhibition of RNA polymerase II 965 phosphorylation by a viral interferon antagonist. J Biol Chem 966 279:31471-31477. 967

73. Marazzi I, Ho JS, Kim J, Manicassamy B, Dewell S, Albrecht RA, 968 Seibert CW, Schaefer U, Jeffrey KL, Prinjha RK, Lee K, Garcia-969 Sastre A, Roeder RG, Tarakhovsky A. 2012. Suppression of the 970 antiviral response by an influenza histone mimic. Nature 483:428-433. 971

74. Friebe P, Boudet J, Simorre JP, Bartenschlager R. 2005. Kissing-972 loop interaction in the 3' end of the hepatitis C virus genome essential 973 for RNA replication. J Virol 79:380-392. 974

75. Hope RG, McLauchlan J. 2000. Sequence motifs required for lipid 975 droplet association and protein stability are unique to the hepatitis C 976 virus core protein. J Gen Virol 81:1913-1925. 977

76. Lindenbach BD, Evans MJ, Syder AJ, Wolk B, Tellinghuisen TL, 978 Liu CC, Maruyama T, Hynes RO, Burton DR, McKeating JA, Rice 979 CM. 2005. Complete replication of hepatitis C virus in cell culture. 980 Science 309:623-626. 981

77. Wakita T, Pietschmann T, Kato T, Date T, Miyamoto M, Zhao Z, 982 Murthy K, Habermann A, Krausslich HG, Mizokami M, 983

Page 33: Çevik, R. E., Cesarec, M., Da Silva Filipe, A., Licastro, D ...eprints.gla.ac.uk/143947/1/143947.pdf1 1 Hepatitis C virus NS5A targets the nucleosome assembly protein 2 NAP1L1 to

32

Bartenschlager R, Liang TJ. 2005. Production of infectious hepatitis 984 C virus in tissue culture from a cloned viral genome. Nat Med 11:791-985 796. 986

78. Targett-Adams P, McLauchlan J. 2005. Development and 987 characterization of a transient-replication assay for the genotype 2a 988 hepatitis C virus subgenomic replicon. J Gen Virol 86:3075-3080. 989

79. Bartolomei G, Cevik RE, Marcello A. 2011. Modulation of hepatitis C 990 virus replication by iron and hepcidin in Huh7 hepatocytes. J Gen Virol 991 92:2072-2081. 992

80. Albornoz A, Carletti T, Corazza G, Marcello A. 2014. The Stress 993 Granule Component TIA-1 Binds Tick-Borne Encephalitis Virus RNA 994 and Is Recruited to Perinuclear Sites of Viral Replication To Inhibit Viral 995 Translation. J Virol 88:6611-6622. 996

81. Macdonald A, Crowder K, Street A, McCormick C, Saksela K, 997 Harris M. 2003. The hepatitis C virus non-structural NS5A protein 998 inhibits activating protein-1 function by perturbing ras-ERK pathway 999 signaling. J Biol Chem 278:17775-17784. 1000

82. Love MI, Huber W, Anders S. 2014. Moderated estimation of fold 1001 change and dispersion for RNA-seq data with DESeq2. Genome Biol 1002 15:550. 1003

83. McCarthy DJ, Smyth GK. 2009. Testing significance relative to a fold-1004 change threshold is a TREAT. Bioinformatics 25:765-771. 1005

84. Kauffmann A, Gentleman R, Huber W. 2009. arrayQualityMetrics--a 1006 bioconductor package for quality assessment of microarray data. 1007 Bioinformatics 25:415-416. 1008

85. Benjamini Y, Hochberg Y. 1995. Controlling the false discovery rate: 1009 a practical and powerful approach to multiple testing. Journal of the 1010 Royal Statistical Society, Series B 57:289-300. 1011

1012 1013

Page 34: Çevik, R. E., Cesarec, M., Da Silva Filipe, A., Licastro, D ...eprints.gla.ac.uk/143947/1/143947.pdf1 1 Hepatitis C virus NS5A targets the nucleosome assembly protein 2 NAP1L1 to

33

Figure Legends 1014 1015 Figure 1 – HCV NS5A binds NAP1L1. 1016 A) Co-immune precipitation of flag-tagged NS5A and HA-NAP1L1 in HEK 1017 293T cells. Transfected cells were lysed, co-IP with anti-FLAG agarose beads 1018 and blotted against α-FLAG or α-HA antibodies as indicated. A plasmid 1019 encoding for HIV-1 flag-tagged Tat was used as positive control. 1020 B) NS5A interacts with endogenous NAP1L1 during HCV replication. Huh7-1021 Lunet cells were electroporated with sub-genomic SGR-JFH1/Luc or mock 1022 transfected. At 72 hpe cell lysates were incubated with α-NAP1L1 antibodies 1023 or with matching irrelevant IgGs. Input and co-IP samples were then immuno-1024 blotted with α-NAP1L1 and α-NS5A antibodies as indicated (IgH, 1025 Immunoglobulin heavy-chain). 1026 C) NS5A and NAP1L1 co-localize during HCV replication. Huh7-Lunet cells 1027 were either mock electroporated or treated with the sub-genomic HCV 1028 replicon SGR-JFH1/Luc or with SGR-FK-Luc-JFH1/ΔE1-E2 and fixed at 72 1029 hpe. Indirect immunofluorescence analysis was performed with α-NAP1L1 1030 (green) and α-NS5A (red) antibodies and corresponding fluorescent 1031 secondary antibodies (scale bar 10 μm). Colocalization is shown in the merge 1032 channel (Pearson’s correlation coefficient of 0.658 for SGR-JFH1/Luc and 1033 0.731 for SGR-FK-Luc-JFH1/ΔE1-E2). The inset shows a high magnification 1034 image. Diffused, cytoplasmic localization of NAP1L1 in mock cells is also 1035 shown for comparison (above). 1036 1037 Figure 2 – HCV JFH1 NS5A targets NAP1L1 for proteasome-mediated 1038 degradation. 1039 A) HCV NS5A from Con1 and H77 interact with NAP1L1. HEK293T cells were 1040 transfected with flag-tagged NS5A from HCV genotypes as indicated. 1041 Transfected cells were lysed, co-IP with α -FLAG agarose beads and blotted 1042 against α-FLAG or α-HA antibodies. 1043 B) HCV NS5A from JFH1 degrades NAP1L1. 293T cells were transfected with 1044 equal amount of HA-NAP1L1 (5 μg) and increasing amount of flag tagged 1045 NS5A from JFH1 or from Con1 (0, 2.5, 5 and 10 μg). After 24 hours cell 1046 lysates were immunoblotted with α-FLAG, α-HA, and α-Actin antibodies. 1047

Page 35: Çevik, R. E., Cesarec, M., Da Silva Filipe, A., Licastro, D ...eprints.gla.ac.uk/143947/1/143947.pdf1 1 Hepatitis C virus NS5A targets the nucleosome assembly protein 2 NAP1L1 to

34

C) HCV NS5A-mediated degradation is proteasome dependent. HEK293T 1048 cells were co-transfected with HA-NAP1L1 and flag-NS5A, treated with the 1049 MG132 (5μM) for 14 h, and lysed. Samples were run and immunoblotted with 1050 α-HA, α-FLAG and α-Actin antibodies. 1051 D) HCV replication induces NAP1L1 degradation. Huh7-Lunet cells were 1052 electroporated with SGR-JFH1/Luc RNA or mock transfected. Cells were 1053 treated with 200 μM Cycloheximide (CHX) for 1 and 3 hours and lysed. 1054 Samples were run and immunoblotted with α-HA, α-FLAG and α-Actin 1055 antibodies. 1056 E) CLUSTAL O (1.2.4) multiple sequence alignment NS5A from different HCV 1057 genotypes. The asterisk character (*) indicates all the sequences that share 1058 the same amino acid. Gaps are indicated by the minus character (-). The 1059 cluster of Serines implcated in NS5A binding are boxed. 1060 1061 Figure 3 – NAP1L1 interacts with the extreme carboxy-terminus of NS5A. 1062 A) Diagram of NS5A and mutants produced in this work. The full-length of 1063 JFH1 NS5A is shown with the flag-tag at the N-terminus, the amphipathic 1064 helix (AH) for membrane tethering, domains I, II and III with the low-1065 complexity sequences (LCS) I and II. AH-fused domain III (AH-D3-FL) is 1066 deleted from the C-terminus in 6 fragments (AH-D3.1-6). The amino acid 1067 sequence of the C-terminal region of NS5A encompassing S452/454/457A 1068 (Serines are in bold) is also shown, together with the sequences of the 1069 deletion mutant NS5A-delB and the triple S>A mutant NS5A-m2. 1070 B) NAP1L1 binds Domain III of NS5A. HEK293T cells were transfected with 1071 the indicated domains fused to AH together with HA-NAP1L1. Transfected 1072 cells were lysed, co-IP with anti-FLAG agarose beads and blotted against α-1073 FLAG or α-HA antibodies. White asterisks (*) indicate the position of the 1074 NS5A mutants that are distinguishable from the heavy (IgH) and light (IgL) 1075 immunoglobulin chains. 1076 C) NAP1L1 binds the extreme C-terminus of NS5A. HEK293T cells were 1077 transfected with expression plasmids encoding for NS5A Domain III fused to 1078 AH carrying progressive deletions from the C-terminus as indicated in the 1079 diagram (Figure 3A). Co-IP was conducted as in Figure 3B. 1080 D) NS5A mutants delB and m2 do not bind NAP1L1. HEK293T were 1081

Page 36: Çevik, R. E., Cesarec, M., Da Silva Filipe, A., Licastro, D ...eprints.gla.ac.uk/143947/1/143947.pdf1 1 Hepatitis C virus NS5A targets the nucleosome assembly protein 2 NAP1L1 to

35

transfected with expression plasmids for NS5A mutants delB and m2 1082 described in Figure 3A. Co-IP was conducted as in Figure 3B. 1083 E) NS5A mutants delB and m2 lose colocalization with NS5A. Huh7-Lunet 1084 cells were electroporated with SGR-JFH1/Luc and with mutants SGR-1085 JFH1/Luc_m2 and SGR-JFH1/Luc_delB and fixed at 72 hpe. Indirect 1086 immunofluorescence analysis was performed with α-NAP1L1 (green) and α-1087 NS5A (red) antibodies and corresponding fluorescent secondary antibodies 1088 (scale bar 10 μm). Colocalization is shown in the merge channel. 1089 1090 Figure 4 – HCV does not require NAP1L1 for replication and infectivity 1091 A) NS5A and NAP1L1-EYFP co-localize during HCV replication. Huh7-Lunet 1092 cells were transduced with a lentiviral vector (LV) expressing NAP1L1-EYFP 1093 and then either mock electroporated or treated with the sub-genomic HCV 1094 replicon SGR-JFH1/Luc and fixed at 72 hpe. Indirect immunofluorescence 1095 analysis was performed with α-NS5A (red) (scale bar 10 μm). 1096 B) NAP1L1 overexpression does not affect HCV genome replication. Huh7-1097 Lunet cells were transduced with a lentiviral vector (LV) expressing EYFP or 1098 NAP1L1-EYFP at an efficiency >85% on average as measured by 1099 cytofluorimetric analysis. Cells were then electroporated with the HCV SGR 1100 JFH1/Luc RNA and luciferase monitored at the indicated time points. Values 1101 are normalized to the luciferase signal at 4 hours post-electroporation. 1102 Average of 3 independent replicates are shown with standard deviations. 1103 C) Depletion of NAP1L1 by shRNA. Huh7-Lunet cells were transduced with 1104 LV expressing shRNA targeting NAP1L1 (shNAP1L1) or non-targeting control 1105 (shCTRL). After selection with puromycin, cells were electroporated with HCV 1106 SGR JFH1/Luc RNA and protein levels detected by WB. 1107 D) Depletion of NAP1L1 does not affect HCV genome replication. Huh7-Lunet 1108 cells were treated as in Figure 4C and the luciferase signal measured as in 1109 Figure 4B. 1110 E) Depletion of NAP1L1 does not affect HCV infectivity. Huh7-Lunet cells 1111 treated with shRNAs as in Figure 4C were electroporated with full-length HCV 1112 JFH1 RNA. At the indicated time points, the supernatant was collected and 1113 used to infect naïve Huh7.5 cells at various dilutions. The 50% tissue culture 1114 infectious dose (TCID50) was then calculated counting cells stained with the 1115

Page 37: Çevik, R. E., Cesarec, M., Da Silva Filipe, A., Licastro, D ...eprints.gla.ac.uk/143947/1/143947.pdf1 1 Hepatitis C virus NS5A targets the nucleosome assembly protein 2 NAP1L1 to

36

NS5A antiserum. 1116 F) Mutagenesis of the NAP1L1 binding site of NS5A does not affect HCV 1117 genome replication. Huh7-Lunet cells were electroporated with HCV SGR 1118 JFH1/Luc RNA from wt, the m2 mutant of NS5A or the GND mutant of NS5B, 1119 which is replication defective. Luciferase was measured as in Figure 4B. 1120 1121 Figure 5 – The interaction with HCV NS5A inhibits NAP1L1 nuclear 1122 translocation. 1123 A) SGR-JFH1 NS5A inhibits NAP1L1 nuclear localization. Huh7-Lunet cells 1124 were electroporated with HCV SGR-JFH1/Luc RNA or the mutant replicon 1125 SGR-JFH1/m2 or control SGR-JFH1/GND as indicated. At 63 h.p.e., cells 1126 were treated with 150 nM LMB for 9 hours. Cells were then fixed and stained 1127 for NS5A and NAP1L1. Scale bar = 10 μm. 1128 B) Quantification of SGR-JFH1 NS5A inhibition of NAP1L1 nuclear 1129 localization. 300 cells treated as in Figure 5A were visually scored for NAP1L1 1130 nuclear localization in the presence of NS5A. The investigator was blinded to 1131 the group allocation during visual counting. Average of 3 independent 1132 replicates are shown with standard deviations. 1133 C) Con1 NS5A mutant m2 does not bind NAP1L1. HEK293T were transfected 1134 with expression plasmids for Con1 NS5A and m2. Co-IP was conducted as in 1135 Figure 3B. 1136 D) Both JFH1 and Con1-derived NS5A inhibit nuclear translocation of 1137 NAP1L1. Huh7-Lunet cells were transfected with expression plasmids for 1138 NS5A from JFH1 or Con1 and their respective m2 mutants. Cells were treated 1139 with LMB as described above, fixed and stained for ectopic flag-tagged NS5A 1140 and endogenous NAP1L1. Scale bar = 10 μm. 1141 E) Quantification of NS5A inhibition of NAP1L1 nuclear localization. Cells 1142 treated as in Figure 5C were visually scored as described above (Figure 5B). 1143 1144 Figure 6 – NAP1L1 is involved in the innate immunity response. 1145 A) Whole-genome transcriptome analysis in NAP1L1-depleted cells. Huh7-1146 Lunet cells were treated with shNAP1L1/shCTRL followed by RNAseq 1147 analysis. Black bars show the levels of 15 down-regulated genes (fold change 1148 ≤ -2) and 2 up-regulated genes (fold change ≥ 2), which were further validated 1149

Page 38: Çevik, R. E., Cesarec, M., Da Silva Filipe, A., Licastro, D ...eprints.gla.ac.uk/143947/1/143947.pdf1 1 Hepatitis C virus NS5A targets the nucleosome assembly protein 2 NAP1L1 to

37

by qRT-PCR (grey bars) normalized for β-actin. 1150 B) Induction of ISG genes is not NAP1L1-dependent. Huh7-Lunet cells were 1151 treated with 1000 U/ml of IFNα for 8 hours. UBD, GBP2, IFITM3 and GAPDH 1152 mRNA was measured by qRT-PCR normalized for β-actin in triplicate 1153 independent experiments. Shown are fold changes over basal, non induced 1154 levels ± SD. 1155 C) IFN induction following poly(I:C) transfection in different cell lines. U2OS, 1156 Huh7-lunet and Huh7.5 were were transfected with 1 μg poly(I:C) for 8 hours. 1157 IFNβ mRNA levels were measured by qRT-PCR, normalized for β-actin and 1158 plotted against mock (lipofectamine). Average of 3 independent replicates are 1159 shown with standard deviations. 1160 D) NAP1L1 depletion affects the induction of IFNβ mRNA by poly(I:C). U2OS 1161 cells treated with shNAP1L1/shCTRL for 3 days were transfected with 1 μg 1162 poly(I:C) for 8 hours. IFNβ mRNA levels were measured as above. 1163 E) NAP1L1 depletion affects the induction of IFNβ by poly(I:C). U2OS cells 1164 were treated as above (Figure 6C). Secreted IFNβ protein was measured by a 1165 commercial ELISA in triplicate, quantified against a standard curve and 1166 plotted. 1167 1168 Figure 7 – NAP1L1 controls RELA levels and IRF3 activation. 1169 A) NAP1L1 depletion affects RELA levels and IRF3 phosphorylation. U2OS 1170 cells were transduced with LV for shNAP1L1 or shCTRL and subsequently 1171 transfected with 1 μg poly(I:C) using lipofectamine (lipo). Protein levels as 1172 indicated were monitored by WB at 1-2-4 hours post transfection of poly(I:C). 1173 B) NAP1L1 depletion decreases RELA protein levels. Blots as in Figure 7A 1174 were quantified to measure RELA and IRF3 protein levels using ImageJ. 1175 Shown is the ratio shNAP1L1/shCTRL in cells not transfected with poly(I:C). 1176 Average of 3 independent replicates are shown with standard deviations. 1177 C) NAP1L1 depletion affects IRF3 phosphorylation. Blots as in Figure 7A 1178 were quantified to measure RELA and IRF3 phosphorylation levels using 1179 ImageJ. Shown is the ration phosphorylated/total protein in cells transfected 1180 with poly(I:C). Average of 3 independent replicates are shown with standard 1181 deviations. 1182 D) NAP1L1 depletion reduces RELA nuclear translocation. U2OS cells were 1183

Page 39: Çevik, R. E., Cesarec, M., Da Silva Filipe, A., Licastro, D ...eprints.gla.ac.uk/143947/1/143947.pdf1 1 Hepatitis C virus NS5A targets the nucleosome assembly protein 2 NAP1L1 to

38

transduced with LV for shNAP1L1 or shCTRL and subsequently transfected 1184 with 1 μg poly(I:C) for 8 hours. Cells were then fixed and stained for RELA. 1185 E) NAP1L1 depletion reduces RELA nuclear translocation. Around 500 cells 1186 from the experiment shown in Figure 7D were counted for each condition to 1187 calculate the percentage of RELA nuclear translocation. Average of 3 1188 independent replicates are shown with standard deviations. 1189 F) NAP1L1 depletion reduces IRF3 nuclear translocation. U2OS cells were 1190 transduced with LV for shNAP1L1 or shCTRL and subsequently transfected 1191 with 1 μg poly(I:C) for 8 hours. Cells were then fixed and stained for IRF3. 1192 G) NAP1L1 depletion reduces IRF3 nuclear translocation. Around 500 cells 1193 from the experiment shown in Figure 7F were counted for each condition to 1194 calculate the percentage of IRF3 nuclear translocation. Average of 3 1195 independent replicates are shown with standard deviations. 1196 H) HCV NS5A inhibits TBK1-mediated activation of IFNβ. HEK 293T cells 1197 were transfected with expression vectors for FLAG-tagged TBK1, NS5A or the 1198 mutants NS5A-m2 together with a reporter plasmid carrying the firefly 1199 luciferase (Fluc) gene under the control of the IFNβ promoter (pIFNβ-Luc) and 1200 the control pCMV-Renilla. Relative light units (RLUs) of luciferase activity was 1201 measured in quintuplicate independent experiments, normalized for Renilla, 1202 and represented as fold change over mock ± SD. 1203 1204 Figure 8 – NAP1L1 controls IRF3 phosphorylation at the TBK1/IKKε 1205 level. 1206 A) Schematic representation of the RIG-I and TLR3 pathways. Both lead to 1207 activation of NF-κB and phosphorylation of IRF3 through MAVS/TBK1/IKKe or 1208 TRIF, respectively. NF-κB and pIRF3 translocate to the nucleus and activate 1209 IFNβ and other ISGs. 1210 B) NAP1L1 does not affect constitutive IRF3-5D activity. Huh7-Lunet cells 1211 were transduced with LV for shNAP1L1 or shCTRL and subsequently 1212 transfected with an expression vector for IRF3-5D, the reporter IFNβ-Luc and 1213 the Renilla control. Cell lysates were blotted as indicated. 1214 C) NAP1L1 does not affect constitutive IRF3-5D activity. Luciferase activity of 1215 cells from the experiment shown in Figure 7H was measured in triplicate 1216 independent experiments, normalized for Renilla. Average values are shown 1217

Page 40: Çevik, R. E., Cesarec, M., Da Silva Filipe, A., Licastro, D ...eprints.gla.ac.uk/143947/1/143947.pdf1 1 Hepatitis C virus NS5A targets the nucleosome assembly protein 2 NAP1L1 to

39

with standard deviations. 1218 D) Depletion of NAP1L1 affects TBK1/IKKε-mediated activation of IFNβ. HEK 1219 293T cells were transduced with LV for shNAP1L1 or shCTRL and 1220 subsequently transfected with expression vectors for FLAG-tagged RIG-I, 1221 IPS-1/MAVS, TBK1 and IKKε together with the reporter IFNβ-Luc and the 1222 Renilla control. Cell lysates were blotted with anti-FLAG as indicated. 1223 Luciferase activity was measured in triplicate independent experiments, 1224 normalized for Renilla, and represented as fold change over mock ± SD. 1225 1226 Figure 9 – NAP1L1 controls the TLR3 pathway. 1227 A) Depletion of NAP1L1 affects TRIF-mediated activation of IFNβ. Huh7-1228 Lunet cells were transduced with LV for shNAP1L1 or shCTRL and 1229 subsequently transfected with expression vectors for TRIF together with the 1230 reporter IFNβ-Luc and the Renilla control. Luciferase activity was measured in 1231 triplicate independent experiments, normalized for Renilla, and represented 1232 as fold change over mock ± SD. 1233 B) Depletion of NAP1L1 by LV shRNA treatment. Huh7-Lunet cells were 1234 transduced with LV for shNAP1L1 or shCTRL and then with LV expressing 1235 TLR3 or EGFP as control. 50 μg of Poly(I:C) was added to the medium for 24 1236 hours. IFNβ mRNA levels for NAP1L1 were measured by qRT-PCR, 1237 normalized for β-actin and plotted against mock. Average of 3 independent 1238 replicates are shown with standard deviations. 1239 C) Reconstitution of the TLR3 pathway. Cells were processed and TLR3 1240 mRNA quantified as in Figure 9B above. 1241 D) Depletion of NAP1L1 affects TLR3 signaling. Cells were processed and 1242 IFIT1 mRNA quantified as in Figure 9B above. 1243 1244 1245

Page 41: Çevik, R. E., Cesarec, M., Da Silva Filipe, A., Licastro, D ...eprints.gla.ac.uk/143947/1/143947.pdf1 1 Hepatitis C virus NS5A targets the nucleosome assembly protein 2 NAP1L1 to

Cevik - Figure 1

NS5A

A

NAP1L1

Tat

NAP1L1

IP DFlag IB DHA

DHA

DFlag

DFlag

Lysate

Lysate

IgH

DIgG DNAP1L1

IgH

NAP1L1

NS5A

IB DNAP1L1

IB DNS5A

B IP

NAP1L1 NS5A merge

NAP1L1 NS5A merge

C NAP1L1

JF

H1/L

uc

FK

-Lu

c-JFH

1/ΔE1

-E2

NS5A

Tat

IP DFlag IB DFlag

IgL

IgH in

set

inset

NS5A merge

Page 42: Çevik, R. E., Cesarec, M., Da Silva Filipe, A., Licastro, D ...eprints.gla.ac.uk/143947/1/143947.pdf1 1 Hepatitis C virus NS5A targets the nucleosome assembly protein 2 NAP1L1 to

NS5A

A

NAP1L1

MG132 + - -

E-actin

MG132 + - - - - - - +

CHX (h) 0 0 1 3 1 3 3 3

mock HCV

mo

ck

HC

V

E-actin

NS5A

C

Cevik - Figure 2

IB DHA

IP DFlag IB DHA

IB DFlag

NAP1L1

D

NS5A

NAP1L1

E-actin

- -

B NS5A-JFH1 NS5A-Con1

NAP1L1

NAP1L1

IP DFlag IB DFlag

NS5A (JFH1)

IgH NS5A (Con1, H77)

Lysate

NS5A (JFH1)

NS5A (Con1, H77)

JFH-1 GSASSMPPLEGEPGDPDLESDQVELQPPPQGGGVAPGSGSGSWSTCSEED--DTTVCC Con1 ESYSSMPPLEGEPGDPDLS--------------------DGSWSTVSEEA-SEDVVCC H77 ESYSSMPPLEGEPGDPDLS--------------------DGSWSTVSSGADTEDVVCC gt3 ESCSSMPPLEGEPGDPDLS--------------------CDSWSTVSDSE-EQSVVCC gt4 GSYSSMPPLEGEPGDPDLT--------------------SDSWSTVSGS---EDVVCC gt5 ASYSSMPPLEGEPGDPDLS--------------------SGSWSTVSGE---DNVVCC gt6 GSFSSMPPLEGEPGDPDLS--------------------TGSWSTVSEE---DDVVCC gt7 ISFSSMPPLEGEPGDPDLS--------------------DGSWSTVSTR---SDVICC * *************** **** * **

E

Page 43: Çevik, R. E., Cesarec, M., Da Silva Filipe, A., Licastro, D ...eprints.gla.ac.uk/143947/1/143947.pdf1 1 Hepatitis C virus NS5A targets the nucleosome assembly protein 2 NAP1L1 to

Cevik - Figure 3

A Domain I Domain II Domain III

LCS I LCS II

AH Flag-

SDQVELQPPPQGGGVAPGSGSGSWSTCSEEDDTTVCC

AH-D3.1 AH-D3.2 AH-D3.3 AH-D3.4

AH-D3.5 AH-D3.6

SDQVELQPPPQGG_______________EEDDTTVCC

SDQVELQPPPQGGGVAPGSGSGAWATCAEEDDTTVCC

delB:

m2:

NAP1L1

NAP1L1

IP DFlag IB DHA

IP DFlag IB DFlag

Lysate IB DHA

*

* *

*

* *

IgH

IgL

NS5A

B

NAP1L1

C

NS5A

DHA

IP DFlag IB DHA

DFlag

NAP1L1 NS5A Merge

JF

H1/L

uc_d

elB

JF

H1/L

uc_m

2

E

JF

H1/L

uc

D

NAP1L1

NS5A

IP DFlag IB DHA

IP DFlag IB DFlag

NS5A IgH

Lysate

NS5A IgH

DHA

DFlag Lysate

IP DFlag IB DFlag

NAP1L1

NAP1L1

Page 44: Çevik, R. E., Cesarec, M., Da Silva Filipe, A., Licastro, D ...eprints.gla.ac.uk/143947/1/143947.pdf1 1 Hepatitis C virus NS5A targets the nucleosome assembly protein 2 NAP1L1 to

Cevik – Figure 4

B

shCTRL

24 48 72

shNAP1L1

24 48 72

NAP1L1

NS5A

β-actin

hpt

24 48 720

20

40

60

80

100

hours post-transfection

Luci

fera

se (f

old

chan

ge)

shNAP1L1shCTRL

24 48 720

1×1004

2×1004

3×1004

hours post-infection

Infe

ctiv

ity (T

CID5

0/m

l)

shNAP1L1shCTRL

24 48 720

10

20

30

40

hours post-transfection

Luci

fera

se (f

old

chan

ge)

NAP1L1-EYFPEYFP

A

D

E

0 24 48 720

50

100

150

200

hours post-transfection

Luci

fera

se (f

old

chan

ge)

JFH1 wtJFH1 GND

JFH1 m2

F

NAP1L1-EYFP NS5A merge

C

JF

H1/L

uc

mo

ck

Page 45: Çevik, R. E., Cesarec, M., Da Silva Filipe, A., Licastro, D ...eprints.gla.ac.uk/143947/1/143947.pdf1 1 Hepatitis C virus NS5A targets the nucleosome assembly protein 2 NAP1L1 to

SGR-JFH1

SGR-m2

0

25

50

75

100

% n

ucle

ar N

AP1

L1in

NS5

A+

cells

*

A" B"NAP1L1" NS5A" Merge"SG

R0JFH1/Luc"

+"LM

B"SG

R0JFH1/m2"

+"LM

B"

NS5A JF

H1 wt

NS5A JF

H1 m2

NS5A C

on1 wt

NS5A C

on1 m2

0

25

50

75

100

% n

ucle

ar N

AP1

L1in

NS5

A+

cells * *

SGR0JFH1/GND"

+LMB"

SGR0JFH1/GND""

D" NAP1L1" NS5A" Merge"

NS5A"JF

H1"wt"

NS5A"JF

H1"m2"

NS5A"Con

1"wt"

NS5A"Con

1"m2"

E"

C"

Cevik&–&Figure&5&

NAP1L1"IP"αFlag"IB"αHA"

NS5A"IgH"

IP"αFlag"IB"αFlag"

input" IP"

Page 46: Çevik, R. E., Cesarec, M., Da Silva Filipe, A., Licastro, D ...eprints.gla.ac.uk/143947/1/143947.pdf1 1 Hepatitis C virus NS5A targets the nucleosome assembly protein 2 NAP1L1 to

Cevik – Figure 6

shCTRL

shNAP1L

1

shCTRL

shNAP1L

1

shCTRL

shNAP1L

1

shCTRL

shNAP1L

102468

101214

fold

cha

nge

ISG

mRN

A

UBD GBP2 IFITM3 GAPDH

HEPACAM

ADH1BCDK2

GEF2RELA

E2F2

CASP7

TGFB1

CCND1 C3CROT

GBP2CFL2

IFITM3

ADAM9

NFKB2UBD

NAP1L1

-20-18-16-14-12-10

-8-6-4-202468

10fo

ld c

hang

e (s

hCTR

L/sh

NAP1

L1)

RNAseqQ-RT-PCR

A

B D E

C

Page 47: Çevik, R. E., Cesarec, M., Da Silva Filipe, A., Licastro, D ...eprints.gla.ac.uk/143947/1/143947.pdf1 1 Hepatitis C virus NS5A targets the nucleosome assembly protein 2 NAP1L1 to

TBK1

TBK1+NS5A

wt

TBK1+NS5A

m2

0

50

100

fold

cha

nge

(RLU

s) **

shCTRL

shNAP1L

10

20

40

60

% n

ucle

ar IR

F3

**

shCTRL

shNAP1L

10

10

20

30

% n

ucle

ar R

ELA *

Cevik - Figure 7

A

pIRF3

E-actin

IRF3

shNAP1L1 shCTRL

lipo p(I:C)

hours 1 2 4 1 2 4 1 2 4 1 2 4

NAP1L1

pRELA

RELA

B lipo p(I:C)

F G

sh

CT

RL

sh

NA

P1L

1

p(I:C)

mock

RELA

p(I:C) lipo

sh

CT

RL

sh

NA

P1L

1

IRF3

shCTRL

shNAP1L

1

shCTRL

shNAP1L

10.0

0.5

1.0

1.5

prot

ein

leve

ls/s

hCTR

L

IRF3 RELA

**

shCTRL

shNAP1L

1

shCTRL

shNAP1L

1

shCTRL

shNAP1L

1

shCTRL

shNAP1L

1

shCTRL

shNAP1L

1

shCTRL

shNAP1L

10.0

0.5

1.0

1.5

2.0

phos

phor

ylat

ed/to

tal

IRF3 RELA

**

*

*

1h 1h2h 2h4h 4hC D E

H

Page 48: Çevik, R. E., Cesarec, M., Da Silva Filipe, A., Licastro, D ...eprints.gla.ac.uk/143947/1/143947.pdf1 1 Hepatitis C virus NS5A targets the nucleosome assembly protein 2 NAP1L1 to

Cevik - Figure 8

C

shCTRL

shNAP1L

10

20

40

60

80

100

fold

cha

nge

(RLU

s)

IRF3-5D

E-actin

IRF3-5D

NAP1L1

- + - +

B A

shCTRL

shNAP1L

1

shCTRL

shNAP1L

1

shCTRL

shNAP1L

1

shCTRL

shNAP1L

1

shCTRL

shNAP1L

10

50

100

150

200

fold

cha

nge

(RLU

s) ** **

****

**

RIG-IN MAVS TBK1 IKK� TBK1/IKK�

RIG-I

MAVS

TBK1/IKKε IRF3

IRF3-P

IFN

NF-κB

TRIF

TLR3

D

NF-κB

E-actin

flag

Page 49: Çevik, R. E., Cesarec, M., Da Silva Filipe, A., Licastro, D ...eprints.gla.ac.uk/143947/1/143947.pdf1 1 Hepatitis C virus NS5A targets the nucleosome assembly protein 2 NAP1L1 to

Cevik - Figure 9

C

B A

D

shCTRL

shNAP1L

10

200

400

600fo

ld c

hang

e (R

LUs)

TRIF

**

shCTRL EGFP

shNAP1L

1 EGFP

shCTRL TLR3

shNAP1L

1 TLR3

shCTRL EGFP

shNAP1L

1 EGFP

shCTRL TLR3

shNAP1L

1 TLR3

0

5000

10000

fold

cha

nge

(TLR

3 m

RNA)

mock p(I:C)

shCTRL E

GFP

shNAP1L

1 EGFP

shCTRL TLR3

shNAP1L

1 TLR3

shCTRL EGFP

shNAP1L

1 EGFP

shCTRL TLR3

shNAP1L

1 TLR3

0

10

20

30

40

50

fold

cha

nge

(IFIT

1 m

RNA)

mock

**

p(I:C)

shCTRL EGFP

shNAP1L

1 EGFP

shCTRL TLR3

shNAP1L

1 TLR3

shCTRL EGFP

shNAP1L

1 EGFP

shCTRL TLR3

shNAP1L

1 TLR3

0.0

0.5

1.0

1.5

fold

cha

nge

(NAP

1L1

mRN

A) mock

**

p(I:C)

** ** **