Next Article in Journal
Picornavirus 3C Proteins Intervene in Host Cell Processes through Proteolysis and Interactions with RNA
Next Article in Special Issue
Quantification of Hepatitis E Virus ORF2 Protein by a Novel Sandwich ELISA
Previous Article in Journal
HPV16 Intratypic Variants in Head and Neck Cancers: A North American Perspective
Previous Article in Special Issue
High Hepatitis E Virus (HEV) Seroprevalence and No Evidence of HEV Viraemia in Vietnamese Blood Donors
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Virus–Host Protein Interaction Network of the Hepatitis E Virus ORF2-4 by Mammalian Two-Hybrid Assays

1
Laboratory of Liver Infectious Diseases, Department of Diagnostic Sciences, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
2
VIB-UGent Center for Medical Biotechnology, Department of Biomolecular Medicine, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
*
Authors to whom correspondence should be addressed.
Viruses 2023, 15(12), 2412; https://doi.org/10.3390/v15122412
Submission received: 14 September 2023 / Revised: 1 December 2023 / Accepted: 4 December 2023 / Published: 12 December 2023
(This article belongs to the Special Issue Hepatitis E: Molecular Virology, Pathogenesis, and Treatment)

Abstract

:
Throughout their life cycle, viruses interact with cellular host factors, thereby influencing propagation, host range, cell tropism and pathogenesis. The hepatitis E virus (HEV) is an underestimated RNA virus in which knowledge of the virus–host interaction network to date is limited. Here, two related high-throughput mammalian two-hybrid approaches (MAPPIT and KISS) were used to screen for HEV-interacting host proteins. Promising hits were examined on protein function, involved pathway(s), and their relation to other viruses. We identified 37 ORF2 hits, 187 for ORF3 and 91 for ORF4. Several hits had functions in the life cycle of distinct viruses. We focused on SHARPIN and RNF5 as candidate hits for ORF3, as they are involved in the RLR-MAVS pathway and interferon (IFN) induction during viral infections. Knocking out (KO) SHARPIN and RNF5 resulted in a different IFN response upon ORF3 transfection, compared to wild-type cells. Moreover, infection was increased in SHARPIN KO cells and decreased in RNF5 KO cells. In conclusion, MAPPIT and KISS are valuable tools to study virus–host interactions, providing insights into the poorly understood HEV life cycle. We further provide evidence for two identified hits as new host factors in the HEV life cycle.

1. Introduction

During its life cycle, viruses interact with a variety of host proteins for their survival, pathogenesis and virus spread. Unraveling virus–host interactions at the protein level aids in the understanding of a virus’ life cycle, its molecular virology and immunology. Moreover, insight into these complex interactions will indicate potential antiviral targets and therefore, protein–protein interactions (PPIs) offer a wide range of possibilities to develop new and more effective therapies.
Many technologies to discover PPIs have been developed, ranging from two-hybrid systems to mass spectrometry, phage display and protein chip technology; all function in high-throughput formats [1,2]. Two described analogous high-throughput mammalian two-hybrid assays are the MAmmalian Protein–protein Interaction Trap (MAPPIT) and the KInase Substrate Sensor (KISS) [2,3]. MAPPIT and KISS are complementation assays based on (parts) of the JAK-STAT signaling pathway. In MAPPIT, a bait protein of interest is fused to a type I transmembrane cytokine receptor that is mutated at its STAT recruitment sites. A prey protein is fused to the cytoplasmic portion of another cytokine receptor containing intact STAT docking sites. Upon the functional association of bait and prey, the JAK proteins can phosphorylate the prey-linked moiety in trans, leading to subsequent STAT binding, dimerization and translocation to the nucleus to induce reporter gene activation. In KISS, a bait protein of interest is fused to the tyrosine kinase 2 (TYK2) domain, which after interaction with the prey is able to phosphorylate the prey-linked moiety. This results in the same downstream signaling as with MAPPIT, but it overcomes the tethering of the bait moiety to the plasma membrane. The advantage of those mammalian systems is that they allow for temporal, spatial and functional modulation and that necessary co-factors or regulatory proteins to study those dynamics of PPIs are present. Moreover, proteins can undergo the proper modifications in their native cellular environment, which is often necessary to allow interactions. The sensitivity and specificity of the described methods are comparable to that of other well-known assays such as Yeast-Two-Hybrid (Y2H) [4]. Both MAPPIT and KISS have shown in the past to be robust methods for detecting PPIs for a variety of applications, including in the field of virus research [5,6].
In this study, both systems were used to identify novel protein binding partners for the proteins of the hepatitis E virus (HEV). HEV is a positive-sense, single-stranded RNA virus and currently the major cause of acute hepatitis worldwide, affecting about 20 million people each year [7]. No specific therapy is available today and a vaccine is only licensed in China [8], highlighting the need for new treatment options. HEV is a virus infecting a range of species, but variants infecting humans can mainly be classified into four genotypes (gts), which differ in geographical distribution, route of transmission and pathogenicity [9]. Gt-1 and gt-2 solely infect humans, are predominant in developing countries and mostly result in self-limiting infections. Nevertheless, mortality in pregnant women reaches up to 25%. Gt-3 and gt-4 infections are mostly zoonotic and are predominant in the industrialized world. Chronic gt-3 and gt-4 infections have been described in immunocompromised patients, which represent an important risk group.
The viral genome translates into three or four proteins, depending on viral genotype (gt). ORF1 encodes a nonstructural polyprotein consisting of eight putative domains and is important for viral replication [10]. Whether this polyprotein is cleaved into several non-structural proteins by either a viral and/or cellular proteases is still a matter of debate. ORF2 and ORF3 are translated from a bicistronic subgenomic replicon. Different forms of ORF2 are produced during the life cycle: an infectious ORF2 that is the component of viral particles, as well as secreted/glycosylated ORF2 and cleaved ORF2 [11,12]. The ORF3 protein is a small phosphoprotein playing a role in multiple cellular processes such as modulating immune responses and is important for viral egress [13,14,15]. A fourth ORF is only observed in gt-1 HEV and is expressed under cellular stress conditions and influences viral replication [16].
The continued effort to define the interactions between the virus and host in general and in particular for HEV, will certainly lead to information about the life cycle of a virus that is far from understood. Previous work on this topic has already led to interesting findings, but overall our knowledge remains limited. To better understand the HEV–host interface as a whole and to compare different genotypes, we generated a protein–protein interaction network by subjecting ORF2 and ORF3 of both gt-1 and gt-3 and ORF4 of gt-1, to two mammalian two-hybrid assays utilizing a prey library of 15,000 proteins. We used bioinformatic databases to analyze these interactions and found significant enriched processes in relation to the respective protein(s). Two proteins from the hit lists, SHARPIN and RNF5, were selected for their interaction with ORF3 and their effect on the ubiquitination status of target proteins involved in the antiviral interferon signaling pathway. Using knockout (KO) cells for the respective proteins, we show they influence HEV-induced interferon (IFN) signaling. Moreover, HEV infection is affected in the KO cells.

2. Materials and Methods

2.1. High Throughput Screens

The MAPPIT and KISS bait were constructed as follows: the coding sequences of both gt-1 ORF2-4 were amplified from a fecal sample of a HEV Sar-55-infected liver-chimeric mouse, and gt-3 ORF2-3 were amplified from the gt-3 Kernow-C1 passage 6 plasmid. These were fused to the EpoR-LR-F3 chimeric receptor in the pSEL (+2L) vector via SalI-NotI restriction cloning (MAPPIT) or N-terminally to a fragment of human TYK2 in the pSVSport vector via EcoRI/MfeI-NotI restriction cloning (KISS). Primers are listed in Table 1. The obtained products were sequence-verified. The construction of prey constructs and the reverse transfection method was performed as described previously [2,3,17]. High-throughput MAPPIT and KISS screens were performed in microarray and retested in luciferase assay as described previously, using a prey library generated using the hORFeome v8.1 and ORFeome Collaboration clones (Center for Cancer Systems Biology, Boston, MA, USA) [17,18,19,20].

2.2. PPI Analysis

Jvenn was used to create Venn diagrams of overlapping proteins [21].
The Database for Annotation, Visualization and Integrated Discovery (DAVID) was used to perform a functional annotation analysis regarding the different GO terms (biological process, molecular function, cellular compartment). DAVID’s modified Fisher Exact p-value was set to 0.05, as a p-value equal or smaller to this number is considered as strongly enriched in the annotation category. Additionally, functional annotation clustering for the terms Functional Annotation and Gene ontology was performed with default settings and a medium classification stringency.

2.3. CRISPR-Cas9 Knockout of SHARPIN and RNF5

Single-guide RNAs (sgRNA) targeting the DNA sequence of SHARPIN and RNF5 were designed using the CRISPR gRNA design tool of Atum https://www.atum.bio/eCommerce/cas9/input (accessed on March–April 2020). Oligo pairs encoding the guide sequences were annealed and ligated into the sgRNA expressing plasmid, pX458 (kindly provided by Feng Zhang, Addgene plasmid #48138) [22]. The sequences of the oligo pairs are represented in Table 2. PLC3 cells were transfected with the CRISPR plasmids using lipofectamine 2000 (Thermo Fisher Scientific, Waltham, MA, USA). In brief, 3 × 105 cells were seeded per well in 6-well plates and transfected with 2500 ng DNA and 12.5 µL lipofectamine. Two days after transfection, single-cell sorting of GFP+ cells was performed using a BD FACS Aria III cell sorter (Becton Dickinson, NJ, USA). Clonal cell populations were expanded and checked for absence of protein expression by Western blot.

2.4. HEV ORF3 Transfection and IFN qRT-PCR Assays

An HEV ORF3 plasmid was constructed as follows. The ORF3 sequence was amplified from the pSK-E2 cDNA clone with following primers: 5′-aagcttaccatgaataacatgtcttttgctgcgcccatgggttc-3′ and 5′-gcggccgcttagcggcgcggccccag-3′. Purified PCR products were cloned into pcDNA3.1 using HindIII and NotI restriction sites and resulting construct was verified by sanger sequencing.
PLC3WT and PLC3SHARPIN-KO and PLC3RNF5-KO were maintained at 37 °C and 5% CO2 in Dulbecco’s Modified Eagle’s Medium (DMEM) (Thermo Fisher Scientific, Waltham, MA, USA) supplemented with 10% inactivated FCS (Biowest, Nuaillé, France), 2 mM L-glutamine (Thermo Fisher Scientific, Waltham, MA, USA), 1% non-essential amino acids (Thermo Fisher Scientific, Waltham, MA, USA), 100 U/mL of penicillin and 100 µg/mL streptomycin (Thermo Fisher Scientific, Waltham, MA, USA). The ORF3 expressing plasmid or mock control were transfected using Lipofectamine 2000 (Thermo Fisher Scientific, Waltham, MA, USA), with a final amount of 2500 ng DNA and 9 µL lipofectamine/well. Tested conditions were performed in duplicate. Two days after MOCK or ORF3 transfection, cells were transfected with poly(I:C) (Invivogen, San Diego, CA, USA) to stimulate the RLR pathway as described before [14]. The end concentration of poly(I:C) used was 1 µg/mL. Sixteen hours post stimulation, cells were lysed for Western blot analysis and RNA extraction for downstream RT-qPCR. RNA was extracted using RNeasy plus mini kit (Qiagen, Hilden, Germany).

2.5. HEV Infectious Virus Production

The gt-3 HEV Kernow-C1 Passage 6 (p6) plasmid (GenBank accession number JQ679013) was linearized with MluI (New England Biolabs, Ipswich, MA, USA). Capped RNA was produced either using T7 RiboMAX Express Large Scale RNA production (Promega, Madison, WI, USA), followed by a ScriptCap m7G capping system (Cellscript, Madison, WI, USA) or using a T7 mMESSAGE mMACHINE kit (Thermo Fisher Scientific, Waltham, MA, USA). Ten micrograms of capped RNA was electroporated into 9 × 106 PLC3 cells using a Gene Pulser (Bio-Rad, Hercules, CA, USA). Viral particles were harvested from centrifuged and filtered (0.22 µm) supernatant, and concentrated using Amicon ultra centrifugal Filters (100 kDa) (Merck, Darmstadt, Germany).

2.6. HEV Infection and HEV ORF2 Immunofluorescence Staining

PLC3WT, PLC3SHARPIN-KO and PLC3RNF5-KO were seeded in black 96-well plates and exposed to a dilution of HEV preparation with an MOI of 22,000, defined as 1 IU/cell, and IU (International unit) was quantified by qPCR as described previously [23]. To determine the infectivity titer, cells were fixed by methanol, followed by incubation with 0.5% Triton X-100 and blocking with PBS containing 10% goat serum. HEV ORF2 staining was performed using a mouse ORF2-specific monoclonal antibody (1E6, Merck, Darmstadt, Germany) in combination with an Alexa Fluor 488-conjugated goat anti-mouse antibody. Nuclei were stained with DAPI (4′,6-diamidino-2-phenylindole). Microscopy was performed using a Leica TSC-SPE confocal microscope (10× or 20× objective) (Wetzlar, Germany). Infectivity of the respective well was calculated as a percentage with automated cell counting as well as ORF2 fluorescence with ImageJ software (v1.54d).

2.7. SDS-PAGE and Western Blot

Cells were lysed with RIPA Lysis and Extraction buffer (Thermo Fisher Scientific, Waltham, MA, USA), supplemented with Halt Protease and Phosphatase Inhibitor Cocktail (Thermo Fisher Scientific, Waltham, MA, USA). Lysates were denatured in the presence of Bolt LDS sample buffer and Bolt Reducing agent (both Thermo Fisher Scientific, Waltham, MA, USA) at 70 °C for 10 min. Samples were subjected to 12% SDS-PAGE and transferred to a polyvinylidene difluoride membrane (PVDF, 0.2 µm pore size; Thermo Fisher Scientific, Waltham, MA, USA). Detection of the targeted proteins was performed by specific antibodies and corresponding horseradish peroxidase-conjugated secondary antibodies (Table 3). Immunoblots were incubated with SuperSignal™ West Femto Maximum Sensitivity Substrate kit (Thermo Fisher Scientific, Waltham, MA, USA) and imaged on an ImageQuant LAS4000 chemiluminescent imaging system (GE Healthcare, Diegem, Belgium).

2.8. Reverse Transcription Quantitative PCR (RT-qPCR)

Total RNA was extracted from cultured cells using the RNeasy plus mini kit (Qiagen, Hilden, Germany). cDNA synthesis was performed with SuperScript III reverse transcriptase (Thermo Fisher Scientific, Waltham, MA, USA). Target and reference gene transcripts were detected using pre-designed Taqman primer-probe assays (Thermo Fisher Scientific, Waltham, MA, USA). All used assays are listed in Table 4. cDNA was diluted 2.5× (target genes) or 10× (reference genes) and 2 µL was used as input for qPCR reaction using TaqMan Fast Advanced Master mix (Thermo Fisher Scientific, Waltham, MA, USA). Cycles of quantification were generated on a LightCycler 480 (Roche, Bazel, Switzerland) using the second-derivative maximum method. Calibrated normalized relative quantities (CNRQs) were calculated for each target gene with qBasePlus Software v3.1 (CellCarta, Montreal, QC, CA), using the most stable reference genes as selected by GeNorm analysis in the qBasePlus Software.

2.9. Statistical Analyses

Statistical analysis was performed using GraphPad Prism 8.0.1 software. Statistical significance of differences between groups was evaluated with t-tests.

3. Results

3.1. MAPPIT and KISS Are Functional Tools to Study HEV–Host Protein–protein Interactions

ORF2 and ORF3 from both gt-1 and gt-3 and ORF4 (gt-1) were used as bait in the two related assays MAPPIT and KISS.
Using this approach, we had four constructs for ORF2: gt-1 ORF2 MAPPIT, gt-1 ORF2 KISS, gt-3 ORF2 MAPPIT and gt-3 ORF2 KISS. Microarrays were performed and revealed 37 hits in total for this protein (Figure 1A). These were uncovered solely with the MAPPIT constructs, as the KISS assay failed to reveal any interactions. Comparing both genotypes, we found that two proteins were shared, specifically CA12 and ADCY3 (Figure 1A) (Appendix A Table A1).
Likewise, four constructs were created for ORF3: gt-1 ORF3 MAPPIT, gt-1 ORF3 KISS, gt-3 ORF3 MAPPIT and gt-3 ORF3 KISS, revealing 187 hits in total (Figure 1B). Most hits were shared between the KISS assay of the two different genotypes, and this initial screening showed an overlap of 25 hits between gt-1 and gt-3 (Figure 1B) (Appendix A Table A1).
For ORF4, combining MAPPIT and KISS revealed 91 hits, of which 11 are shared between the two assays (Figure 1C) (Appendix A Table A1).
To obtain a general overview of all the HEV hits discovered with MAPPIT and KISS, we compiled all the different hits per HEV protein, irrespective of the used method or genotype, and looked whether there are particular hits shared between HEV ORF2, ORF3 and ORF4, as HEV proteins often interact with each other during the viral life cycle. Following this approach, the Venn diagram shows that a substantial amount of hits were shared between both ORF3 and ORF4, STUB1 was shared between ORF2 and ORF4; and ORF2 and ORF3 shared FTL, TMEM154 and FCGR2A (Figure 1D) (Appendix A Table A1).

3.2. Functional Annotation Clustering for the HEV Proteins

To gain a comprehensive overview of the hits and relate biological relevance to those identified, we first performed gene ontology (GO) annotation for each protein with the DAVID bioinformatics tool.
For ORF2, five significant clusters could be observed for biological processes (BP) (Figure 2A, Appendix A Table A2), all containing two proteins or 5.4% of the ORF2 dataset. From the cellular compartment (CC) clusters, it is obvious that almost half of the amount of hits are associated with the (plasma) membrane, and about 14% is associated with the cell surface (Figure 2B, Appendix A Table A2). Three clusters could be observed regarding the molecular function (MF) annotation, being TPR-domain binding, carbonate dehydratase activity and hydro-lyase activity, which all contained two proteins or 5.4% of the dataset (Figure 2B, Appendix A Table A2).
For ORF3, 28 BP clusters were observed, with among them processes such as viral entry into host cells, transmembrane transport, GPCR signaling and immune response (Figure 3A, Appendix A Table A3). As with the ORF2 hits, a high proportion of the total ORF3 hit list reside in membranes or plasma membranes (Figure 3B, Appendix A Table A3).
For ORF4, 13 BP clusters could be observed with regulation of alternative mRNA splicing, with the positive regulation of proteolysis and negative regulation of transcription among the most significant ones (Figure 4A, Appendix A Table A4). More than half of the proteins are clustered in the cellular compartment cytoplasm, but a great deal of proteins also cluster to the nucleus. Regarding the MF ontology, 88% of the hits have protein-binding capacities, and about 15% are (m)RNA-binding proteins (Figure 4C, Appendix A Table A4).
As mentioned in the previous section, HEV proteins often interact with each other and therefore, we were interested to see if the identified candidate interactors would cluster together in certain (upregulated) GO terms. Functional annotation clustering of the total HEV hit list was performed in DAVID and revealed 22 clusters (Appendix A Table A5). We could again observe that most of the proteins are observed in a membrane cluster. The second most enriched cluster involved proteins important for viral entry or receptor activity. Proteins related to GPCR signaling were again observed. Cluster 5 and 6 relate to ion channel terms and may be related to the function of ORF3 as an ion channel. Inflammatory response and immunity also emerged and molecular functions relating to post-translational modifications such as protein phosphorylation and ubiquitination are also present.
Additionally, we also performed a search for hits that have previously been described in the context of viral infections (Appendix A Table A6). Specifically, we found that 15 of the 37 ORF2-interacting proteins identified with MAPPIT and KISS are in some way involved in viral infections. Likewise, 108 of the 187 identified ORF3-interacting host proteins and 33 ORF4-interacting protein hits have previously been described to play a role in viral infections.
To further scrutinize the proteins, we focused on hits that were identified either for both genotypes, or in both assays, or shared among HEV proteins. The selection was further narrowed down by investigating if potential hits are known to be important in viral infections (Appendix A Table A6). One of the hits we observed in the clusters of protein ubiquitination is SHARPIN or SHANK-Associated RH Domain Interactor. In our analysis, we observed this as a hit of gt-1 ORF3, identified by both MAPPIT and KISS assay, and as a hit of ORF4, also identified by both MAPPIT and KISS assay. We speculated that gt-3 ORF3 could also bind SHARPIN. Therefore, we performed a retest of the gt-3 ORF3 in the KISS configuration, as this proved the most effective for this genotype in our initial analysis. Using this approach, we could confirm that gt-3 ORF3 also bound SHARPIN (Table 5).
In the same cluster of protein ubiquitination, we identified the Ring Finger Protein 5 (RNF5). During our analysis, this appeared a candidate hit of gt-1 ORF3. Along with the experiments we performed for SHARPIN, we speculated that gt-3 ORF3 could also bind RNF5. Upon retest, we could indeed confirm the interaction of gt-3 ORF3 with RNF5 (Table 5).

3.3. SHARPIN Affects the Induction of Interferon upon ORF3 Transfection

SHARPIN forms the linear ubiquitin chain assembly complex (LUBAC) which mainly targets proteins in the Retinoic acid-Inducible Gene I (RIG-I)-like receptor (RLR)-Mitochondrial Antiviral Signaling Protein (MAVS) Pathway.
To understand the roles of this protein in the context of HEV infection, a CRISPR-Cas9 knockout (KO) cell line was generated (Figure 5A). We used PLC3 cells, a subclone of PLC/PRF/5, that are permissive to HEV replication [11].
To understand a potential interaction of this protein with ORF3, we expressed this protein in PLC3WT and PLC3SHARPIN-KO (Figure 5A) and investigated its effect on the induction of IFN, since LUBAC has been shown to regulate signaling downstream of TLR3, RIG-I and MDA5. According to what was previously published [14], we could show that ORF3 also boosted poly (I:C) mediated-type I IFN induction in PLC3 cells. Viral protein-transfected cells showed a response twice as high as the mock control (p = 0.017), while unstimulated cells had an absent or only very minimal response (Figure 5B, left panel). Strikingly, in the SHARPIN KO, the ORF3-induced enhancement of type I IFN was abolished (p = 0.24) and rather tended to show an inverse effect (Figure 5B, left panel). Likewise, we investigated the type III IFN response. Although the trend seemed to be the same as for type I IFN, we could not show a significant induction of type III IFN upon ORF3 addition in PLC3WT cells (p = 0.13) (Figure 5B, right panel). We next wondered if these changes in interferon response could have an impact on HEV infection. To this end, WT and SHARPIN-KO cells were infected and the level of infection was measured by immunostaining 6 days later. Strikingly, infection increased from 17% in WT cells to 25% in the SHARPIN-KO cells (p = 0.0357) (Figure 5C). Together, these results show SHARPIN has an effect on the ORF3-mediated IFN induction, while infection is stimulated in the KO cells.

3.4. RNF5 Interferes with IFN Induction upon ORF3 Transfection

The Ring Finger Protein 5 (RNF5) is a membrane-bound ubiquitin ligase that is known to inhibit IFN type I in the context of virus infections. To investigate a potential effect of RNF5 in the HEV life cycle by interacting with ORF3, we performed a similar experiments as for SHARPIN above. PLC3RNF5-KO cells were generated and successful KO was verified by Western blot; ORF3 was expressed in these cells and compared to PLC3WT (Figure 6A). ORF3 enhanced the type I IFN response upon poly I:C stimulation (p = 0.022), but this was not observed in the RNF5-KO cells. Moreover, the trend seems to be that IFN-β diminishes upon ORF3 transfection in these cells (p = 0.28) (Figure 6B, left panel). For type III IFN responses, we observed the same trend: there was an increase upon ORF3 transfection in WT cells (p = 0.023), which was not observed in the KO cells (p = 0.26) (Figure 6B, right panel).
Furthermore, we checked if there is an influence on HEV infection in PLC3RNF5-KO cells compared to PLC3WT. Cells were infected and the amount of infection was measured by immunostaining 6 days later. Infection reduced from 15% in the WT population to 6% in the KO population (Figure 6C). These results show that RNF5 interferes with the ORF3increased IFN response and influences HEV infection.

4. Discussion

In this study, we used two PPI methods to identify interactions of the HEV ORF2-4 to aid in the urgent need to better understand the HEV pathogenesis, life cycle and virus–host interactions. Some HEV PPI studies have been performed in the past, most of them using yeast-two-hybrid and affinity purification in combination with mass spectrometry [24,25,26,27,28,29]. One method can only detect a certain fraction of all PPIs, illustrating that in order to comprehensively map the interactome of one particular protein of interest, it is important to use different methods and consider them as complementary rather than as independent assays [30]. The advantage of the MAPPIT and KISS screenings used in this study is their mammalian background and high-throughput capability [2,3,19]. We validated this approach for HEV and identified 37 candidate interactors for ORF2, 187 for ORF3 and 91 for ORF4. We did not investigate ORF1 due to the controversy and complexity about the processing of this protein, and the studies conducted so far with this protein have always used the individual domains as bait [31,32]. Additionally, ORF1 interacts with JAK2 [32], which means it could bind the JAK2 proteins that are constitutively associated with the chimeric MAPPIT receptor, posing an additional difficulty.
For ORF2, our candidate interactors were identified only in the MAPPIT configuration. In initial tests of the KISS screening, positive controls that bind the Tyk2 domain did not result in acceptable signals and a pre-screen with known ORF2 interactions did not reveal any hits.It is unclear why this happened. It could point to a technical issue of the assay but it is more likely that it is attributed to the nature of the HEV ORF2 protein, as this is the only one of the HEV proteins that failed. In MAPPIT, the bait moiety is tethered to the plasma membrane, whereas in KISS, the bait can shuttle in the cytosol. It is known that ORF2 translocates to the nucleus and reticular compartments; therefore, it is possible that by residing in these subcellular compartments, the construct is hidden for potential interactions with prey proteins in the cytosol [33]. Before screening, we anticipated that the subcellular localization of HEV proteins could impede some analyses, especially since it is known, for example, that replication of viruses can induce membrane rearrangements to shield for immune responses [34]. For this reason, we decided to perform screenings with both methods.
In our 37 identified ORF2 hits, we could confirm four previously described interactions of this HEV protein: HSP90, TMBIM4, FTL and CYB5A [24,26]. Only two hits were shared between gt-1 and gt-3 in our assay, being CA12 and ADCY3 (Figure 1A); the former protein is part of the zinc metalloenzymes catalyzing the reversible hydration of carbon dioxide, whereas ADCY3 catalyzes the formation of the secondary messenger cyclic adenosine monophosphate (cAMP). Gene ontology annotation analysis revealed that most of the identified ORF2 hits are membrane-associated proteins, which is in agreement with what was published in another ORF2 screen using Y2H [26]. Looking at the biological processes, there were two proteins that were often observed: FLOT1 and COLEC12 (Figure 2A). FLOT1 is an integral membrane component of caveolae and plays a role in vesicle trafficking and cell morphology. Additionally, flotillin-dependent endocytosis is an alternative pathway, independent of clathrin or dynamin that could in theory be used by viruses. However, mechanisms remain unclear and many studies report no involvement of the flotillin in virus entry, including for hepatitis C virus or hepatitis A virus (HAV) [35,36,37]. However, FLOT1 is present in HAV exosomes [38], which might also be the case for HEV, and a potential interaction with FLOT1 might be of importance during membrane fusion in the process of viral release [39]. COLEC12 is a member of the C-lectin family: it is a scavenger receptor that may function as pattern recognition molecule (PRM) by binding carbohydrate antigens on microorganisms (particularly bacteria) to facilitate their removal. However, no clear studies have yet confirmed this role in the context of viral infection, except one study showing no binding of the SARS-CoV-2 spike to COLEC12, while it did bind other related humoral pattern recognition molecules [40]. The expression of COLEC-12 in human hepatocytes was confirmed in a study involving the hepatitis C virus, but the role of the protein was not further investigated [41].
For ORF3, we confirmed the previously described interactions with TSG101, NAT1, MAT1A, HSPA8, HPX, FTL, ALDOB, C151, FGB, APOH and AMBP and an additional 176 hits that were not previously described. Strikingly, the most significant biological process we observed was the one containing proteins involved in ‘viral entry into host cells’ (Figure 3A). EFNB2 is a receptor for Hendra and Nipah virus [42], CD4 for human immunodeficiency virus [43], CXADR is the coxsackievirus and adenovirus receptor [44], and BSG has been shown to be involved in infection of measles and human cytomegalovirus [45,46]. AGTR1 also plays a role during SARS-CoV-2 endocytosis and NECTIN-4 is a receptor for measles virus [47,48]. These proteins mostly bind a viral protein to mediate viral entry, so it is intriguing why ORF3 is observed as an interacting partner of these proteins. Only HAVCR1 is a phosphatidylserine receptor that mediates the entry of a variety of viruses that acquire PS in their viral envelope through a process called ‘apoptotic mimicry’ [49]. We very recently studied the role of this protein in the entry of HEV and could demonstrate the involvement of PS acquired in the enveloped form of the HEV virions [50]. The observation of all these viral receptors might indicate that the role of ORF3 during entry of enveloped hepatitis E virus is of more importance than we think and should be further studied. We also observed some BPs and MFs related to ion transmembrane transport (Figure 3), which could be related to the function of ORF3 as a functional ion channel in the release of virions [13]. We also observed some BPs regarding the immune response. For ORF3, it was previously described that it can interfere with cellular host defenses and its interferon-induced effectors, sometimes by inhibiting IFN induction, or by induction of IFN [14,51,52]. Most of the proteins here were also membrane proteins (Figure 3B). Intriguingly, we observed an annotation term of extracellular exosome, containing 31 proteins. It is well known that ORF3 uses the exosome pathway, a process orchestrated by the interaction of ORF3 and TSG101 to dock virions in the multivesicular bodies leading to viral release [53].
For ORF4, only two previous studies have been performed to screen for interacting proteins [16,54]. The hits we observed during our MAPPIT and KISS analysis were not reported before. We observed that ORF3 and ORF4 shared 10 protein hits. Previously, it was reported that ORF4 forms a protein complex with the RdRp, helicase and X, of which the assembly is inhibited by interaction with ORF3 [16]. It is possible ORF3 and ORF4 form another complex together with host proteins to exert this and/or other functions. Interestingly, TSG101 seems to be one of the proteins that is shared between ORF3 and ORF4. Another multivesicular body protein, CHMP4C, interacted with ORF4, illustrating it might be worthwhile to study this interaction in the context of gt-1 virus assembly and release. For ORF4 BPs, we could also observe proteins related to virus responses. Both ADAR and PRKRA (PACT), part of the RNA sensing pathway, were observed to interact with ORF4. ADAR has both pro- and antiviral activities, one of them by limiting the amount of dsRNA and thereby inhibiting PKR activation. Furthermore, PKR is activated by PACT and its activation inhibits cellular mRNA translation through the phosphorylation of EIF2A [55,56]. It is intriguing that ORF4 binds both these proteins. It was also reported that ORF4 is a target of the proteasome due to ubiquitination of Lysine at the 51st amino acid position [16]. Several proteins in our hit list were linked to a (poly)ubiquitination status (Figure 4), most of them are (part of) E3 ubiquitin ligases (complex). FBXW11 and BTRC are even part of the same complex. It would be interesting to see if one of the observed interactors is able to ligate ubiquitination to ORF4. On the other hand, these proteins often ubiquitinate target proteins involved in innate defense pathways such as NF-kB pathways. Additionally, SHARPIN and TRAF1 were also identified as ORF4 interactors, and previous work illustrated that TRAF1 is a target of the LUBAC complex, consisting of HOIP, HOIL-1L and SHARPIN, to enhance NF-kB activation. On the contrary, it was also reported that TRAF1 sequesters LUBAC to limit ubiquitination of another target NEMO, thereby limiting NF-kB activation [57]. It remains to be elucidated if a similar process could be observed upon ORF4 interaction.
Many of the proteins we discovered as HEV-interacting proteins were already described to be implicated in other viral infections (Appendix A Table A6). This clearly indicates the relevance of the interactome we generated in this work. For an abundance of these host proteins, this role was linked to an increased or decreased expression upon viral infection or relates to data generated by other PPI-screens. The specific role that the identified host protein played in the viral life cycle was not necessarily further investigated. For other host proteins, including the already-mentioned HAVCR1, ADAR, EFNB2, CXADR and NECTIN4, their role has already been more thoroughly studied, making a potential relationship to the HEV life cycle more easy. A disadvantage of the here-applied PPI methods, but by extension of similar approaches such as yeast-two-hybrid, is that they rely on interactions between proteins that are artificially brought together in the same cell by ectopic expression. Confirming biological relevance of the uncovered interactions is therefore of the utmost importance. We selected SHARPIN and RNF5 for further characterization.
Initially, we only observed an interaction of both SHARPIN and RNF5 with gt-1 ORF3. By retesting the constructs in a second MAPPIT and KISS screening experiment, we could confirm the interaction for both ORF3 (Table 5). Again this illustrates that additional hits may be found upon repetition of a certain PPI method. That we confirmed the interaction with gt-3 for both SHARPIN and RNF5 is not unusual: amino acid sequence homology between the used gt-1 Sar55 strain and gt-3 Kernow-C1 p6 strain is more than 80%, but gt-specific regions exist. Similarly, Geng et al., identified 32 shared human proteins interacting with ORF3 from human isolates of gt-1 and gt-4, and with ORF3 from a gt-3 rabbit isolate [28].
SHARPIN, together with HOIL-1 and HOIP, forms LUBAC, the only complex known so far able to polyubiquitinate target proteins in a linear fashion. Studies regarding this complex in viral infections have led to opposing data. For Sendai virus (SeV) and Vesicular stomatitis virus (VSV), it was shown that LUBAC inhibits virus-induced interferon, whereas a study using norovirus showed that parts of the LUBAC complex are essential for the induction of both type I and type III IFN [58,59,60]. Here, we also investigated both the type I and type III IFN response. In addition, it is known that HEV can induce both types as well [61,62,63]. In agreement with the previous literature, we could confirm that ORF3 enhances poly (I:C)-mediated type I IFN induction 1.9–2.3 fold compared to the mock control (Figure 5B and Figure 6B) [14]. Remarkably, when PLC3SHARPIN-KO cells were transfected with ORF3, the poly(I:C)-mediated induction of IFN diminished (Figure 5B). It is thought the ORF3-mediated IFN enhancement is caused by RIG-I activation through K63-linked ubiquitination. Two ubiquitin ligases can mediate this activation: TRIM25 and Riplet [14]. Interestingly, TRIM25 has been described as a target for LUBAC [60]. During SeV, LUBAC suppresses RIG-I ubiquitination and activation by inducing TRIM25 degradation, thereby suppressing subsequent type I IFN production. However, our data would suggest an inverse relationship as absence of SHARPIN/LUBAC might suppress IFN induction upon ORF3 (Figure 5B). Still, since opposing effects of LUBAC (components) have been described for a variety of viruses, the targets may be differentially regulated depending on the virus of interest. These effects were mainly seen upon type I IFN induction, but the trend seems to be the same for type III IFN induction. Interestingly, HEV infection is slightly higher in SHARPIN-KO cells compared to control (Figure 5C), and might be related to the lower level of ORF3-mediated IFN induction in these cells.
Future research should explore which exact proteins belonging to the RLR-MAVS pathway leading to IFN induction are targeted by SHARPIN/LUBAC in the context of HEV. Taking things together, a protein complex between RIG-I, TRIM25, SHARPIN and ORF3 might be a possibility, as mentioned above. Additionally, LUBAC can also target NEMO to attenuate IFN response upon VSV infection, via disruption of the MAVS-TRAF3 complex. The hepatitis B virus induces the protein Parkin to recruit LUBAC to MAVS and disrupt downstream signaling [59,64].
A second candidate interactor of ORF3 that we selected was RNF5, especially because this protein has also been described to influence IFN induction in context of viral infections. For example, the V protein of Newcastle disease virus and PB1 protein of Influenza A virus recruit RNF5 to polyubiquitinate and degrade MAVS, which inhibit type I IFN production [65,66]. Next to MAVS, the protein STING has also been described as a target for RNF5. SeV induces the ubiquitination and degradation of STING by RNF5 affecting IFN signaling [67]. We showed that IFN induction is affected in RNF5-KO cells upon ORF3 transfection (Figure 6B). We also show that HEV infection is affected in these KO cells, with a significantly lower infection level compared to wild-type cells (Figure 6C). It would be interesting to speculate if HEV (ORF3) could interfere with the IFN induction by recruiting RNF5 to its target proteins. One previous study showed that HEV does not induce MAVS degradation; further research should explore whether STING would be a more reasonable target in case of HEV infection [61].

5. Conclusions

In conclusion, our data demonstrate that MAPPIT and KISS are valuable tools to probe HEV–host protein interactions. Moreover, our obtained hit list for HEV ORF2-4 provides a wealth of information to find new host factors implicated in the HEV life cycle, which remains far from understood. Confirmation of biological relevance of obtained hits in high-throughput PPI-methods is important. We specifically identified SHARPIN and RNF5 as interactors for both gt-1 and gt-3 ORF3 and showed they interfere with IFN signaling and HEV infection in general.

Author Contributions

Conceptualization, L.C., I.L., J.T. and P.M.; methodology, L.C., I.L., J.T. and P.M.; investigation, L.C., I.L., K.W., A.D.M. and F.V.H.; writing—original draft preparation, L.C. and P.M.; writing—review and editing, L.C., I.L., K.W., J.T. and P.M.; supervision, J.T. and P.M.; funding acquisition, J.T. and P.M. All authors have read and agreed to the published version of the manuscript.

Funding

P.M. was supported by the Special Research Fund of Ghent University (BOFEXP2017001002), P.M. and J.T. were supported by the Research Foundation-Flanders (FWO-Vlaanderen; Excellence of Science (EOS) projects VirEOS and VirEOS2.0).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Supporting data in Appendix A are available on request.

Acknowledgments

We would like to thank Suzanne U. Emerson (NIH, United States) for providing the p6 clone, and Laurence Cocquerel (Institut Pasteur, Lille) for the PLC3 cells. We acknowledge the CORE Flow Cytometry from the Faculty of Medicine and Health Sciences at Ghent University for use and support of the flow cytometry and sorting devices.

Conflicts of Interest

The authors declare no conflict of interest.

Appendix A

Table A1. Overview of hits in common between MAPPIT and KISS screens.
Table A1. Overview of hits in common between MAPPIT and KISS screens.
ScreenShared Protein Hits
Gt-1 ORF2 MAPPIT|Gt-3 ORF2 MAPPITCA12, ADCY3
Gt-1 ORF3 MAPPIT|Gt-1 ORF3 KISSSHARPIN
Gt-1 ORF3 KISS|Gt-3 ORF3 KISSTMEM108, FAM171B, MCAM, TREML1, GPRC5D, SPN, MAG, HS1BP3, OPALIN, BSG, ERMAP, BTN3A2, CRB3, NUMB, PVRL4, TMEM184A, PTH1R, VSIG8, SLC7A8, PTGER4, SURF4
Gt-1 ORF3 MAPPIT|Gt-3 ORF3 KISSCX3CL1, CYBRD1, HAVCR1
Gt-3 ORF3 MAPPIT|Gt-3 ORF3 KISSNAT1, MAT1A, HSPA8, HPX
Gt-1 ORF3 KISS|Gt-3 ORF3 KISS|Gt-3 MAPPITTSG101
ORF4 MAPPIT|ORF4 KISSSHARPIN, C9orf169, MSI2, C3orf56, CELF5, TRAF1, LPXN, RBFOX1, THAP8, FAM168A, THAP11
ORF2|ORF3TMEM154, FCGR2A, FTL
ORF3|ORF4FAM92A1, RBPMS, SHARPIN, SMN2, C9orf169, RCHY1, OLIG1, TSG101, CCDC170, METTL16
ORF2|ORF4STUB1
Table A2. GO analysis of hits identified during MAPPIT and KISS screens against the HEV ORF2 protein.
Table A2. GO analysis of hits identified during MAPPIT and KISS screens against the HEV ORF2 protein.
Annotation CategoryGO TermGene Name
Biological ProcessGO:0097531~mast cell migrationSTAT5B, KITLG
GO:0044857~plasma membrane raft organizationCOLEC12, FLOT1
GO:0060355~positive regulation of cell adhesion molecule productionCOLEC12, FLOT1
GO:0061684~chaperone-mediated autophagyHSP90AA1, STUB1
GO:0071360~cellular response to exogenous dsRNACOLEC12, FLOT1
Cellular CompartmentGO:0016020~membraneCA12, COLEC12, CYB5A, HSP90AA1, CMTM6, TOMM34, ADCY3, GBA2, EDA2R, TMEM154, KITLG, FUBP3, TMBIM4, CA4, FLOT1, C5ORF60, LGALS8, FTL
GO:0016021~integral component of MembraneCA12, COLEC12, CYB5A, CMTM6, TOMM34, NDUFB1, ADCY3, TNFRSF10B, GBA2, EDA2R, TMEM154, KITLG, FCGR2A, TMBIM4, IGDCC3, CA4, C5ORF60, TIGIT
GO:0005886~plasma membraneCA12, COLEC12, HSP90AA1, ANXA4, CMTM6, ADCY3, TNFRSF10B, GBA2, EDA2R, KITLG, FCGR2A, RAB42, CAPNS2, IGDCC3, CA4, FLOT1, TIGIT
GO:0009986~cell surface TGIT, TNFRSF10B, ANXA4, CA4, HSP90AA1
Molecular FunctionGO:0030911~TPR domain bindingHSP90AA1, STUB1
GO:0004089~carbonate dehydratase activityCA12, CA4
GO:0016836~hydro-lyase activityCA12, CA4
Table A3. GO analysis of hits identified during MAPPIT and KISS screens against the HEV ORF3 protein.
Table A3. GO analysis of hits identified during MAPPIT and KISS screens against the HEV ORF3 protein.
Annotation CategoryGO TermGene Name
Biological ProcessGO:0046718~viral entry into host cellEFNB2, CD4, CXADR, BSG, AGTR1, NECTIN4, HAVCR1
GO:1901379~regulation of potassium ion transmembrane transportKCNE1, KCNIP1, KCNIP2, KCNAB3
GO:0007166~cell surface receptor
signaling pathway
SPN, CD4, FCGR2A, ADGRF5, BSG, PTH2R, PTH1R, ADRB2, MS4A12, FCGR2B, IL27RA
GO:0007155~cell adhesionFGB, CD151, AMBP, SEMA4D, MCAM, CD99L2, CX3CL1, EFNB2, EFNB1, MAG, CD4, BSG, SSPN, NECTIN4
GO:0006955~immune responsePTGER4, SEMA4D, IL1R2, CXCR6, CX3CL1, IL27RA, SPN, CD4, CD7, FCGR2B, ICOS, CAMP, CMKLR1
GO:0007188~adenylate cyclase-
modulating G-protein coupled
receptor signaling pathway
PTGER4, FPR1, PTH2R, PTH1R, ADRB2
GO:0007204~positive regulation of cytosolic calcium ion concentrationPTGER4, FPR1, AGTR1, LPAR1, S1PR3, CXCR6, CMKLR1
GO:0030168~platelet activationFGB, DGKG, TREML1, ENTPD2, PDPN
GO:0050863~regulation of T cell activationSPN, CD4, TREX1
GO:0043031~negative regulation of macrophage activationADGRF5, FCGR2B, CD200
GO:0006935~chemotaxisSPN, FPR1, CXCR6, PLP2, CX3CL1, CMKLR1
GO:0050776~regulation of immune responseSPN, FCGR2A, FCGR2B, CD200
GO:0098915~membrane repolarization
during ventricular cardiac muscle cell action potential
KCNE1, KCNE4, SCN2B
GO:0007189~adenylate cyclase-activating G-protein coupled receptor signaling pathwayPTGER4, ADGRF5, LPAR1, S1PR3, PTH1R, ADRB2
GO:0097623~potassium ion export across plasma membraneKCNE1, KCNIP2, KCNE4
GO:0019722~calcium-mediated signalingTREML1, CD4, AGTR1, CXCR6, MCOLN1
GO:0007186~G-protein coupled receptor signaling pathwayENTPD2, FZD7, LPAR1, FPR1, MRGPRF, GPBAR1, PTH2R, CXCR6, PTH1R, CX3CL1, GPR173, ADGRF5, AGTR1, APLNR, S1PR3, GPRC5D, CMKLR1
GO:1904659~glucose transmembrane transportSLC2A12, SLC2A5, SLC2A6
GO:0007200~phospholipase C-activating G-protein coupled receptor
signaling pathway
FPR1, AGTR1, PTH1R, CMKLR1
GO:0001817~regulation of cytokine
Production
BTN3A3, BTN3A2, ERMAP, LITAF
GO:0055085~transmembrane transportSLC44A5, SLCO1B1, GJA3, FXYD2, CYBRD1, RNF5, SLC2A6
GO:0060326~cell chemotaxisAGTR1, LPAR1, CXCR6, CX3CL1
GO:0006556~S-adenosylmethionine
biosynthetic process
MAT1A, METTL16
GO:0071347~cellular response to
interleukin-1
FGB, MMP2, CAMP, CX3CL1
GO:0020027~hemoglobin metabolic
process
HPX, AMBP
GO:0032690~negative regulation of
interleukin-1 alpha production
IL1R2, CX3CL1
GO:0071277~cellular response to
calcium ion
SYT5, NEUROD2, BRAF, MCOLN1
GO:0031295~T cell costimulationEFNB2, EFNB1, ICOS
Cellular CompartmentGO:0005886~plasma membraneDGKG, MRGPRF, NCMAP, IL27RA, GPR173, GJA3, BSG, MS4A12, CMKLR1, ENTPD2, CXADR, IL1R2, SYTL1, SLC9B2, MAG, AGTR1, APLNR, KCNE1, TSG101, TTYH3, CD151, GYPB, SLC22A1, FPR1, LPAR1, LYPD3, CD99L2, ADRB2, CBARP, EFNB2, SLCO1B1, EFNB1, EPB41L2, EPB41L3, ICOS, TMEM184A, SYT5, HSPA8, MPIG6B, AMBP, CAV2, MCAM, FZD7, SURF4, CYBRD1, TNFRSF10A, BRAF, TREML1, LRFN4, FXYD2, SSPN, CD200, SCN2B, SLC44A5, PTH1R, SLC2A5, SLC6A1, LITAF, SLC2A6, CX3CL1, SPN, HAVCR1, ERMAP, C11ORF24, FGB, OPALIN, MMP2, SLC2A12, GPBAR1, PTH2R, BTN3A3, BTN3A2, KCNAB3, SLC7A8, ADGRF5, NUMB, GPRC5D, PTGER4, PLPPR1, STAU2, PACC1, CXCR6, ACVR1B, RNF5, SLITRK3, PDPN, S1PR3, CACNG1, CACNG2, RELT, LAIR1, JAM2, SEMA4D, KCNIP1, KCNIP2, KIR3DL2, MCOLN1, CD4, FCGR2A, PIK3IP1, CD6, CD7, RAB9A, PLP2, FCGR2B, NECTIN4, CRB3
GO:0016021~integral component of
membrane
ZNF451, DPY19L2P2, MRGPRF, IL27RA, GPR173, SYNPR, GJA3, BSG, MS4A12, VSIG8, CMKLR1, STARD3, ENTPD2, IL1R2, SLC9B2, MAG, AGTR1, APLNR, KCNE1, RTN1, CD151, KCNE4, GYPB, SLC22A1, FPR1, LPAR1, LYPD3, SLC38A11, CD99L2, ADRB2, CBARP, SLCO1B1, EPB41L3, ICOS, C16ORF54, TMEM184A, SYT5, MPIG6B, CAV2, RHBDD1, TREX1, MCAM, FZD7, SURF4, CYBRD1, TNFRSF10A, DNAJC14, TREML1, LRFN4, FAM163A, CD200, SLC44A5, PTH1R, SLC2A5, SLC6A1, LITAF, SLC2A6, CX3CL1, SPN, TMEM108, HAVCR1, ERMAP, C11ORF24, OPALIN, SLC2A12, GPBAR1, PTH2R, BTN3A3, C1ORF162, BTN3A2, SYP, SLC7A8, ADGRF5, RFT1, GPRC5D, PTGER4, PLPPR1, SLC43A3, PACC1, TMEM72, CXCR6, RNF5, PDPN, RELT, LAIR1, JAM2, RPRM, CMTM4, TMEM176B, KIR3DL2, MCOLN1, TMEM154, CD4, FCGR2A, PIK3IP1, CD6, LPCAT4, CD7, FAM171B, PLP2, FCGR2B, NECTIN4, CRB3
GO:0005887~integral component of
plasma membrane
CD151, PLPPR1, PACC1, GYPB, SLC22A1, LPAR1, ADRB2, CXCR6, PTH1R, SLC6A1, ACVR1B, SLC2A5, NCMAP, IL27RA, EFNB2, SPN, EFNB1, SLCO1B1, GJA3, PDPN, BSG, S1PR3, CACNG1, JAM2, CMKLR1, CXADR, SEMA4D, CAV2, PTH2R, KIR3DL2, MCOLN1, MAG, CD4, FCGR2A, SLC7A8, CD6, SSPN, AGTR1, APLNR, FCGR2B, CD200
GO:0009986~cell surfaceFGB, KCNE1, CD151, AMBP, PACC1, LPAR1, CD99L2, TNFRSF10A, SLC6A1, ACVR1B, CX3CL1, SPN, TREML1, EFNB1, LRFN4, CD6, APOH, ADGRF5, SLITRK3, CACNG2, HAVCR1, CD200, JAM2
GO:0016020~membraneLC44A5, DGKG, PTH1R, SLC6A1, SLC2A5, SLC2A6, CX3CL1, SPN, SYNPR, BSG, H4C1, VSIG8, CMKLR1, H3C8, ENTPD2, IL1R2, SLC2A12, BTN3A3, SYP, C1ORF162, BTN3A2, DNAJC7, ADGRF5, AGTR1, VDAC3, FTL, PTGER4, CD151, STAU2, SLC22A1, TMEM72, FPR1, SLC38A11, ADRB2, ACVR1B, EFNB2, SLCO1B1, PDPN, RPRM, C16ORF54, HSPA8, CMTM4, CAV2, FZD7, CYBRD1, DNAJC14, MCOLN1, TMEM154, TREML1, CD4, CD6, LPCAT4, FAM163A, AGO2, CD7, FAM171B, PLP2, CD200
GO:0016323~basolateral plasma membraneSLCO1B1, CXADR, SLC7A8, SLC43A3, SLC22A1, PDPN, BSG, NUMB, PTH1R, SLC9B2
GO:0043005~neuron projectionSYNPR, CXADR, PLPPR1, RTN1, STAU2, SMN2, BRAF, SYP, SLC6A1, MARK4, CX3CL1, CD200
GO:0045121~membrane raftEFNB1, MAG, CD4, KCNE1, CXADR, CAV2, PDPN, BSG, TNFRSF10A
GO:0042383~sarcolemmaSTAC, CAV2, BSG, SSPN, CACNG1, SLC2A5
GO:0016324~apical plasma membraneKCNE1, SLC7A8, KCNE4, SLC22A1, PDPN, CYBRD1, PTH1R, ADRB2, SLC2A5, CRB3, SLC9B2
GO:0033270~paranode region of axonMAG, EPB41L3, NCMAP
GO:0036477~somatodendritic compartmentTMEM108, CACNG2, JAM2
GO:0009897~external side of plasma membraneFGB, SPN, CD4, CD6, MCAM, BTN3A3, CXCR6, BTN3A2, FCGR2B, ERMAP, IL27RA
GO:0070062~extracellular exosomeTSG101, TTYH3, SLC2A5, CST4, SPN, RAB43, EFNB1, HPX, CYSRT1, APOH, EPB41L2, BSG, H4C1, CAMP, FGB, H3C8, HSPA8, ENTPD2, AMBP, SYTL1, CYBRD1, DNAJC7, FXYD2, AGO2, VDAC3, RAB9A, ALDOB, NECTIN4, GPRC5D, CRB3, FTL
GO:0043235~receptor complexGPBAR1, PTH1R, ADRB2, GPRC5D, MCOLN1, ACVR1B, IL27RA
GO:0005925~focal adhesionEFNB2, HSPA8, TLE2, CD151, CAV2, MCAM, EPB41L2, BSG, NUMB, CD99L2
GO:0030672~synaptic vesicle membraneSYT5, SYNPR, SYP, CBARP, SLC9B2
GO:0044297~cell bodyCXADR, BRAF, FCGR2B, CD200
GO:0031528~microvillus membraneSLC7A8, PDPN, SYTL1
GO:0008076~voltage-gated potassium channel complexKCNE1, KCNIP1, KCNIP2, KCNAB3
GO:0098982~GABA-ergic synapseLRFN4, SLITRK3, LPAR1, SLC6A1
GO:0072562~blood microparticleFGB, HSPA8, HPX, PRSS1, AMBP
GO:0030425~dendriteEFNB2, HSPA8, SHARPIN, RTN1, KCNIP1, KCNIP2, AGO2, TMEM108, MARK4
Molecular FunctionGO:0005515~protein bindingZNF451, IL27RA, RAB43, SYNPR, CYSRT1, RUSC1, BSG, BANF1, MS4A12, H4C1, PDK1, CMKLR1, STARD3, TLE2, ENTPD2, CXADR, IL1R2, SYTL1, MAT1A, KCTD4, SLC9B2, NPAS4, AGTR1, APLNR, VDAC3, FTL, NAA60, ZNF792, KCNE1, TSG101, RTN1, CD151, KCNE4, GYPB, CUL2, SLC22A1, FPR1, LPAR1, SLC38A11, CD99L2, ADRB2, EFNB2, EFNB1, HPX, SHARPIN, APOH, EPB41L2, EPB41L3, SMN2, DMAP1, ICOS, MARK4, C16ORF54, HSPA8, NIBAN3, MPIG6B, AMBP, RBPMS, CAV2, STAC, RHBDD1, FZD7, SURF4, CYBRD1, TNFRSF10A, BRAF, TREML1, KIF18A, LRFN4, FAM163A, AGO2, CIBAR1, CIBAR2, CD200, SLC44A5, PTH1R, SLC2A5, SLC6A1, LITAF, SLC2A6, CX3CL1, SPN, TMEM108, HAVCR1, ERMAP, C11ORF24, TGIF1, FGB, H3C8, STRBP, MMP2, PTH2R, OLIG1, BTN3A2, SYP, KCNAB3, DNAJC7, SLC7A8, GFOD1, ADGRF5, RFT1, NUMB, ALDOB, GPRC5D, PTGER4, PLPPR1, SLC43A3, STAU2, PACC1, TMEM72, RCHY1, ACVR1B, RNF5, CST4, SLITRK3, PDPN, HS1BP3, S1PR3, CACNG1, CACNG2, RELT, LAIR1, JAM2, RPRM, KRTAP10-5, SEMA4D, KCNIP1, CMTM4, KCNIP2, TMEM176B, CIMAP1A, KIR3DL2, MCOLN1, DHRS2, TMEM154, CD4, FCGR2A, PIK3IP1, CD6, CD7, CCDC170, RAB9A, PLP2, FCGR2B, NECTIN4, CRB3
GO:0015459~potassium channel regulator activityKCNE1, KCNIP1, KCNIP2, KCNE4, ADRB2, KCNAB3
GO:0001618~virus receptor activityEFNB2, CD4, CXADR, BSG, NECTIN4, HAVCR1
GO:0044325~ion channel bindingKCNE1, KCNIP1, STAC, KCNIP2, KCNE4, KCNAB3, CBARP
GO:0022857~transmembrane transporter activitySLC44A5, SLCO1B1, SLC7A8, SLC43A3, SLC22A1, SLC2A5, SLC2A6
GO:1902282~voltage-gated potassium channel activity involved in ventricular
cardiac muscle cell action potential
repolarization
KCNE1, KCNE4, SCN2B
GO:0005355~glucose transmembrane transporter activitySLC2A12, SLC2A5, SLC2A6
GO:0004888~transmembrane signaling receptor activitySPN, TREML1, CD4, FCGR2A, SEMA4D, FCGR2B, IL27RA
GO:0042802~identical protein bindingZNF792, SLC22A1, ADRB2, SLC6A1, RNF5, LITAF, SHARPIN, CYSRT1, APOH, SMN2, BANF1, KRTAP10-5, CXADR, SEMA4D, KCNIP2, CYBRD1, TNFRSF10A, BRAF, SYP, MAT1A, MCOLN1, SLC9B2, CD4, CD6, ALDOB, NECTIN4, FTL
GO:0005102~receptor bindingFGB, MAG, CXADR, SEMA4D, PIK3IP1, PDPN, BTN3A3, BTN3A2, ERMAP, CX3CL1
GO:0004991~parathyroid hormone
receptor activity
PTH2R, PTH1R
GO:0038023~signaling receptor activityCD4, SEMA4D, BSG, CD7, APLNR, TNFRSF10A, CMKLR1
GO:0044877~macromolecular complex bindingNPAS4, KCNE1, TSG101, SHARPIN, KCNIP2, CUL2, ADRB2, FCGR2B, RNF5
GO:0004930~G-protein coupled receptor activityFZD7, LPAR1, FPR1, MRGPRF, PTH2R, CXCR6, ADRB2, GPR173, ADGRF5, AGTR1, APLNR, S1PR3, GPRC5D, CMKLR1
GO:0008289~lipid bindingDGKG, STARD3, CD4, APOH, S1PR3, MCOLN1
GO:0042803~protein homodimerization
activity
STARD3, NAA60, TSG101, AMBP, RBPMS, CAV2, TREX1, PTH1R, ADRB2, RCHY1, MAG, CD4, BANF1
Table A4. GO analysis of hits identified during MAPPIT and KISS screens against the HEV ORF4 protein.
Table A4. GO analysis of hits identified during MAPPIT and KISS screens against the HEV ORF4 protein.
Annotation CategoryGO TermGene Name
Biological ProcessGO:0000381~regulation of alternative mRNA splicing, via spliceosomeRBFOX1, RBFOX2, CELF4, CELF5
GO:0045862~positive regulation of
Proteolysis
FBXW11, STUB1, BTRC
GO:0045892~negative regulation of transcription, DNA-templetedLBH, RBFOX2, HEY1, FBXW11, EPC1, BTRC, WTIP, GATAD2A
GO:0009615~response to virusRPS15A, PRKRA, ADAR, CCT5
GO:0043161~proteasome-mediated
ubiquitin-dependent protein
catabolic process
SHARPIN, FBXW11, STUB1, BTRC, KLHL20
GO:0006397~mRNA processingRBFOX1, RBFOX2, CELF4, SMN2, ADAR
GO:0016567~protein ubiquitinationFBXW11, MKRN3, STUB1, BTRC, LZTR1, RCHY1, KLHL20
GO:0007010~cytoskeleten organizationDES, ZMYM6, WTIP, KLHL20
GO:0016236~macroautophagyTSG101, CHMP4C, ZFYVE1
GO:0000209~protein polyubiquitinationFBXW11, MKRN3, STUB1, BTRC
GO:0042753~positive regulation of
circadian rhythm
FBXW11, BTRC
GO:0035455~response to interferon-alphaADAR, KLHL20
GO:0000122~negative regulation of transcription from RNA polymerase II
promotor
TSG101, HEY1, DDX20, ZBTB42, EPC1, ZNF3, MEIS2, GATAD2A, THAP11
Cellular CompartmentGO:0005737~cytoplasmITSN2, HSPB6, CELF4, CELF5, MSI1, ADAR, MSI2, PPP1CC, LBH, HEY1, EPC1, METTL16, LZTS2, BTRC, CEP55, SH3GLB2, RBFOX1, RBFOX2, DYNLT3, TRAF1, PRKAR1B, PSME1, BLZF1, TSG101, FBLIM1, CAMK2A, DDX20, C10ORF88, DAZ3, ZBTB42, RCHY1, KLC1, SAV1, RPS15A, GNA11, SMN2, LPXN, CTAG1B, PDLIM5, ZC3H14, PIBF1, CCT5, CARD9, KLHL20, DES, IMPDH1, PRKRA, STUB1, CIBAR1
GO:0005829~cytosolITSN2, TSG101, HSPB6, FBLIM1, CAMK2A, DDX20, MSI2, RCHY1, KLC1, SAV1, KRT83, PPP1CC, RPS15A, SHARPIN, SMN2, CEP72, PLXNA1, LPXN, LZTS2, BTRC, PDLIM5, CCT5, SH3GLB2, RBFOX2, RBPMS, FBXW11, KRTAP10-9, CARD9, TRAF1, KLHL20, THAP11, DES, PRKAR1B, DDAH2, IMPDH1, PRKRA, CHMP4C, PSME1, STUB1
GO:0005634~nucleusHSPB6, CAMK2A, NAB2, DDX20, CELF4, CELF5, DAZ3, ZBTB42, MSI1, ADAR, ZNF3, RCHY1, SAV1, PPP1CC, LBH, HEY1, ZMYM6, SMN2, EPC1, LPXN, METTL16, BTRC, ZC3H14, PIBF1, RBFOX1, RBFOX2, FBXW11, DYNLT3, OLIG1, WTIP, MEIS2, GATAD2A, DES, IMPDH1, PRKRA, STUB1, CIBAR1, BLZF1
GO:0090543~Flemming bodyTSG101, CHMP4C, CEP55
GO:0000151~ubiquitin ligase complexSHARPIN, FBXW11, STUB1, RCHY1
GO:0005771~multivesicular bodyTSG101, PRKAR1B, CHMP4C
GO:0030018~Z-discDES, SMN2, STUB1, PDLIM5
GO:0005813~centrosomeITSN2, FBXW11, CEP72, LZTS2, PIBF1, CCT5, CEP55
GO:0000776~kinetochorePPP1CC, FBXW11, DYNLT3, CHMP4C
GO:0005654~nucleoplasmSH3GLB2, RBFOX2, RBPMS, CAMK2A, NAB2, DDX20, CELF4, ZBTB42, ADAR, RCHY1, GATAD2A, THAP11, PPP1CC, RPS15A, HEY1, PRKRA, SMN2, PSME1, CEP72, PLXNA1, EPC1, STUB1, BTRC, BLZF1
GO:1990904~ribonucleoprotein complexCELF4, MKRN3, CELF5, ZC3H14
GO:0030496~midbodyPPP1CC, CHMP4C, LZTS2, CEP55
GO:0032797~SMN complexDDX20, SMN2
GO:0097504~Gemini of coiled bodiesDDX20, SMN2
Molecular FunctionGO:0005515~protein bindingITSN2, NAB2, IHH, CELF5, MSI1, ADAR, MSI2, ZFYVE1, PPP1CC, LBH, CYSRT1, HEY1, EPC1, LZTS2, BTRC, SMCO1, CEP55, LINC02875, SH3GLB2, RBFOX1, RBFOX2, FBXW11, DYNLT3, OLIG1, TRAF1, THAP8, PRKAR1B, DDAH2, PPP1R3B, CHMP4C, PSME1, PRR23E, NUP54, METTL27, BLZF1, TSG101, FBLIM1, CAMK2A, MOSPD2, DDX20, C10ORF88, DAZ3, ZBTB42, ZNF3, RCHY1, KLC1, KRT83, SAV1, FAM168A, RPS15A, SHARPIN, GNA11, MGAT5B, SMN2, CEP72, LPXN, CTAG1B, PDLIM5, ZC3H14, PIBF1, CCT5, RBPMS, KRTAP10-9, MIIP, CARD9, WTIP, MEIS2, GATAD2A, KLHL20, THAP11, DES, PRKRA, CCDC170, CORO6, MKRN3, NMUR2, STUB1, LZTR1, CIBAR1
GO:0042802~identical protein bindingSH3GLB2, DYNLT3, CARD9, CAMK2A, NAB2, C10ORF88, MSI1, TRAF1, MSI2, ZNF3, SAV1, SHARPIN, DES, CYSRT1, IMPDH1, PRKRA, SMN2, MKRN3, NUP54, CTAG1B, PIBF1, CEP55
GO:0003723~RNA bindingRBFOX1, RBFOX2, RBPMS, DDX20, CELF4, CELF5, ALG13, ADAR, MSI1, MSI2, PPP1CC, RPS15A, IMPDH1, PRKRA, SMN2, MKRN3, METTL16, ZC3H14
GO:0003729~mRNA bindingRBFOX1, RBFOX2, RBPMS, CELF4, CELF5, MSI1, MSI2
GO:0004842~ubiquitin-protein
transferase activity
SHARPIN, FBXW11, STUB1, TRAF1, BTRC, RCHY1, KLHL20
GO:0042803~protein homodimerization activityTSG101, RBPMS, HSPB6, PRKRA, CARD9, CAMK2A, CHMP4C, STUB1, RCHY1
GO:0046983~protein dimerization activityHEY1, FBXW11, OLIG1, BTRC
Table A5. Functional annotation clustering of the HEV ORF candidate interactors.
Table A5. Functional annotation clustering of the HEV ORF candidate interactors.
Annotation cluster 1Enrichment score: 10.14Amount of hits
GOTERM_CC_DIRECTplasma membrane138
UP_KW_CELLULAR_COMPONENTcell membrane103
UP_KW_CELLULAR_COMPONENTMembrane170
Annotation cluster 2Enrichment score: 2.38Amount of hits
GOTERM_BP_DIRECTviral entry into host cell7
UP_KW_MOLECULAR_FUNCTIONhost cell receptor for virus entry6
GOTERM_MF_DIRECTvirus receptor activity6
Annotation cluster 3Enrichment score: 1.9Amount of hits
GOTERM_BP_DIRECTmiRNA loading onto RISC involved in gene silencing by miRNA3
UP_KW_BIOLOGICAL_PROCESSRNA-mediated gene silencing5
GOTERM_BP_DIRECTpre-miRNA processing3
GOTERM_MF_DIRECTdouble-stranded RNA binding5
Annotation cluster 4Enrichment score: 1.72Amount of hits
GOTERM_MF_DIRECTmRNA binding11
UP_KW_MOLECULAR_FUNCTIONRNA-binding21
GOTERM_MF_DIRECTRNA binding27
Annotation cluster 5Enrichment score: 1.7Amount of hits
UP_KW_MOLECULAR_FUNCTIONvoltage-gated channel8
GOTERM_BP_DIRECTmembrane repolarization during ventricular cardiac muscle cell action potential3
GOTERM_MF_DIRECTvoltage-gated potassium channel activity involved in ventricular cardiac muscle cell action potential repolarization3
GOTERM_BP_DIRECTregulation of heart rate by cardiac conduction3
Annotation cluster 6Enrichment score: 1.62Amount of hits
GOTERM_MF_DIRECTpotassium channel regulator activity6
GOTERM_BP_DIRECTregulation of potassium ion transmembrane transport4
GOTERM_MF_DIRECTion channel binding8
UP_KW_MOLECULAR_FUNCTIONvoltage-gated channel8
GOTERM_BP_DIRECTpotassium ion export across plasma membrane3
UP_KW_BIOLOGICAL_PROCESSIon transport17
UP_KW_BIOLOGICAL_PROCESSPotassium transport6
UP_KW_MOLECULAR_FUNCTIONIon channel11
UP_KW_MOLECULAR_FUNCTIONPotassium channel4
GOTERM_CC_DIRECTvoltage-gated potassium channel complex4
GOTERM_MF_DIRECTvoltage-gated potassium channel activity3
GOTERM_BP_DIRECTpotassium ion transmembrane transport3
Annotation cluster 7Enrichment score: 1.53Amount of hits
UP_KW_MOLECULAR_FUNCTIONreceptor43
GOTERM_BP_DIRECTpositive regulation of cytosolic calcium ion concentration8
UP_KW_MOLECULAR_FUNCTIONG-protein coupled receptor19
UP_KW_MOLECULAR_FUNCTIONtransducer20
GOTERM_BP_DIRECTG-protein coupled receptor signaling pathway20
GOTERM_MF_DIRECTG-protein coupled receptor activity16
Annotation cluster 8Enrichment score: 1.5Amount of hits
GOTERM_MF_DIRECTglucose transmembrane transporter activity3
UP_KW_BIOLOGICAL_PROCESSsugar transport4
GOTERM_BP_DIRECTglucose transmembrane transport3
Annotation cluster 9Enrichment score: 1.04Amount of hits
GOTERM_BP_DIRECTchemotaxis7
GOTERM_BP_DIRECTchemokine-mediated signaling pathway3
GOTERM_BP_DIRECTinflammatory response8
Annotation cluster 10Enrichment score: 0.96Amount of hits
UP_KW_MOLECULAR_FUNCTIONdevelopmental protein21
UP_KW_MOLECULAR_FUNCTIONdevelopmental protein21
UP_KW_BIOLOGICAL_PROCESSdifferentiation16
Annotation cluster 11Enrichment score: 0.94Amount of hits
GOTERM_BP_DIRECTpositive regulation of proteolysis3
GOTERM_BP_DIRECTpost-translational protein modification4
GOTERM_BP_DIRECTSCF-dep. proteasomal ubiquitin-dep. protein catabolic process4
GOTERM_MF_DIRECTubiquitin-protein transferase activity8
GOTERM_BP_DIRECTprotein polyubiquitination6
GOTERM_BP_DIRECTproteasome-mediated ubiquitin-dep. protein catabolic process6
GOTERM_MF_DIRECTubiquitin protein ligase activity7
Annotation cluster 12Enrichment score: 0.79Amount of hits
GOTERM_MF_DIRECTprotein binding involved in protein folding4
GOTERM_BP_DIRECTprotein refolding3
UP_KW_MOLECULAR_FUNCTIONchaperone7
GOTERM_MF_DIRECTunfolded protein binding5
GOTERM_BP_DIRECTprotein folding5
UP_KW_BIOLOGICAL_PROCESSstress response3
GOTERM_MF_DIRECTATPase activity6
Annotation cluster 13Enrichment score: 0.68Amount of hits
GOTERM_CC_DIRECTubiquitin ligase complex5
GOTERM_MF_DIRECTubiquitin-protein transferase activity8
GOTERM_BP_DIRECTprotein destabilization3
GOTERM_BP_DIRECTproteasome-mediated ubiquitin-dep. protein catabolic process6
GOTERM_BP_DIRECTprotein ubiquitination10
UP_KW_BIOLOGICAL_PROCESSUbl conjugation pathway13
GOTERM_MF_DIRECTubiquitin protein ligase activity7
GOTERM_BP_DIRECTubiquitin-dependent protein catabolic process5
Annotation cluster 14Enrichment score: 0.62Amount of hits
GOTERM_BP_DIRECTcell projection organization4
GOTERM_CC_DIRECTciliary basal body5
UP_KW_BIOLOGICAL_PROCESScilium biogenesis/degradation5
GOTERM_CC_DIRECTcentriole4
GOTERM_BP_DIRECTcilium assembly4
Annotation cluster 15Enrichment score: 0.59Amount of hits
GOTERM_BP_DIRECTregulation of alternative mRNA splicing, via spliceosome4
GOTERM_BP_DIRECTmRNA processing6
UP_KW_BIOLOGICAL_PROCESSmRNA processing8
UP_KW_BIOLOGICAL_PROCESSmRNA splicing6
GOTERM_BP_DIRECTRNA splicing4
Annotation cluster 16Enrichment score: 0.59Amount of hits
UP_KW_BIOLOGICAL_PROCESSdifferentiation16
UP_KW_BIOLOGICAL_PROCESSspermatogenesis6
GOTERM_BP_DIRECTcell differentiation9
GOTERM_BP_DIRECTspermatogenesis6
Annotation cluster 17Enrichment score: 0.34Amount of hits
GOTERM_BP_DIRECTmicrotubule-based movement3
UP_KW_CELLULAR_COMPONENTmicrotubule6
UP_KW_MOLECULAR_FUNCTIONmotor protein3
Annotation cluster 18Enrichment score: 0.26Amount of hits
GOTERM_CC_DIRECTmidbody5
UP_KW_BIOLOGICAL_PROCESScell division7
GOTERM_BP_DIRECTcell cycle6
UP_KW_BIOLOGICAL_PROCESSmitosis5
GOTERM_BP_DIRECTcell division6
UP_KW_BIOLOGICAL_PROCESScell cycle8
Annotation cluster 19Enrichment score: 0.21Amount of hits
UP_KW_BIOLOGICAL_PROCESSadaptive immunity9
UP_KW_BIOLOGICAL_PROCESSimmunity13
GOTERM_BP_DIRECTadaptive immune response6
Annotation cluster 20Enrichment score: 0.08Amount of hits
GOTERM_MF_DIRECTkinase activity4
GOTERM_BP_DIRECTprotein phosphorylation7
UP_KW_MOLECULAR_FUNCTIONkinase9
UP_KW_MOLECULAR_FUNCTIONserine/threonine-protein kinase4
GOTERM_MF_DIRECTprotein serine/threonine kinase activity4
Annotation cluster 21Enrichment score: 0.08Amount of hits
GOTERM_CC_DIRECTextracellular region28
GOTERM_CC_DIRECTextracellular space25
UP_KW_CELLULAR_COMPONENTsecreted23
Annotation cluster 22Enrichment score: 0.05Amount of hits
UP_KW_MOLECULAR_FUNCTIONrepressor11
GOTERM_BP_DIRECTpositive regulation of transcription from RNA polymerase II promoter15
GOTERM_BP_DIRECTregulation of transcription, DNA-templated12
GOTERM_MF_DIRECTtranscriptional activator activity, RNA polymerase II transcription regulatory region sequence-specific binding6
GOTERM_CC_DIRECTchromatin10
UP_KW_BIOLOGICAL_PROCESStranscription regulation26
UP_KW_MOLECULAR_FUNCTIONDNA-binding22
UP_KW_BIOLOGICAL_PROCESStranscription26
GOTERM_MF_DIRECTRNA polymerase II transcription factor activity, sequence-specific DNA binding12
GOTERM_MF_DIRECTRNA polymerase II core promoter proximal region sequence-specific DNA binding11
GOTERM_MF_DIRECTsequence-specific double-stranded DNA binding4
GOTERM_MF_DIRECTtranscription factor activity, sequence-specific DNA binding4
GOTERM_BP_DIRECTregulation of transcription from RNA polymerase II promoter13
Table A6. Described functions in viral infection of HEV ORF candidate interactors.
Table A6. Described functions in viral infection of HEV ORF candidate interactors.
Gene SymbolHEV Interacting ProteinFunction in Viral InfectionReference
UBR2ORF2
-
Human immunodeficiency virus 1 (HIV-1) infection was decreased in UBR2-depleted cells
[68]
FTLORF2, ORF3
-
Interacts with several proteins and domains of HEV (ORF1, macro, protease, helicase, ORF3). It was proposed that macro domain prevents ferritin from entering into circulation and helping attenuating host immune response
-
In vitro overexpression of FTL can enhance Hepatitis B virus (HBV) attachment
[26,28,69,70]
TNFRSF10BORF2
-
Expression reduced in liver tissue from chronic HBV and HBV infected cell lines
-
HBV X protein degrades TNFRSF10B via macroautophagy-mediated degradation, allowing survival of virus-infected cells
-
Hantavirus inhibits TRAIL-mediated killing of infected cells by downregulating TNFRSF10B
-
HBV core protein inhibits TRAIL-induced apoptosis of hepatocytes by blocking TNFRSF10B expression
-
Hepatitis C virus (HCV) upregulates TNFRSF10B, sensitizes host cells to TRAIL-induced apoptosis
[71,72,73]
TOMM34ORF2
-
Upregulated in HCV-induced hepatocellular carcinoma (HCC)
-
Upregulated in HIV-infected CD4 T-cells
-
Increased phosphorylation in Dengue Virus (DENV)-2 infected cells
[74,75,76]
TIGITORF2
-
Functionally linked to IL-10 expression, known to promote virus persistence in vitro
-
TIGIT modulation alters phenotype and cytokine profile of T cells during Influenza and chronic Lymphocytic choriomeningitis virus (LCMV)
-
Regulates NK cell function in chronic HBV infection
-
Blocking TIGIT enhances NK cell activity against HIV-infected T cells
[77,78,79]
FCGR2AORF2, ORF3
-
Variants are associated with thrombocytopenia in DENV
-
Variants are associated with severe Respiratory syncytial virus (RSV) in Argentina
-
Enhanced FCGR2A signaling in HIV viremic controllers
[80,81]
ANXA4ORF2
-
Downregulated in human monocytic cells infected with DENV
-
Upregulated in a proteomic study analyzing saliva from COVID-19 patients
-
Higher levels of serum ANXA4 in HCV-related HCC
[82,83,84]
CMTM6ORF2
-
Involved in the PLD1 upregulation after entecavir treatment during HBV infection, may be a mechanism of hepatocytes to subvert immune surveillance
[85]
TTC1ORF2
-
Upregulated in A549 cells after Influenza virus
[86]
CAPNS2ORF2
-
Downregulated Human papillomavirus 16 (HPV16) infection
-
Upregulated during Human cytomegalovirus (HCMV) infection
[87,88]
PSMA5ORF2
-
Recognizes oxidized ApoB (together with PSMA6) that is induced upon HCV infection, which is known to enhance lipid accumulation and promote viral production
-
Increased in PBMCs from COVID-19
[89,90]
FUBP3ORF2
-
Knockdown leads to decreased Japanese encephalitis virus (JEV), co-localizes with viral proteins and assists in viral replication complex
-
Host restriction factor in Porcine epidemic diarrhea virus (PEDV) infection, degrades PEDV-encoded nucleocapsid via E3 ubiquitin ligase recruitment, and regulates IFN-1 signaling pathway by interacting with TNF-TRAF3 expression
-
Interacts with enterovirus EV71 5′UTR and knockdown and leads to decreased EV71 IRES activity; might therefore serve as IRES regulator
[91,92,93]
LGALS8ORF2
-
Controls autophagy during adenovirus cell entry through viral capsid. PPXY motif in this protein is critical for efficient evasion of autophagic sequestration. Depletion of LGALS8 rescues infectivity of a PPXY-mutant virus
[94]
STUB1ORF2, ORF4
-
Targeted by the SUMO-interacting motif of EBNA1 to maintain Epstein–Barr Virus (EBV) latency
-
Bovine ephemeral fever (BEFV) triggers RACK1 expression, which upregulates STUB1, thereby promoting MAVS degradation and IFN type I inhibition
-
STAT4 can interact with STUB1 to block RIG-I in RNA virus infection
[95,96,97]
STAT5BORF2
-
Reduction in STAT5B expression in HIV-infected patients
-
STAT5B activation promotes virus amplification of insect viruses, whereas suppression inhibits apoptosis and reduces virus accumulation, linked with JAK-STAT pathway
-
Target of SARS-CoV-2 encoded miRNAs
[98,99,100,101]
ADRB2ORF3
-
Upregulated by NK cells in early mouse CMV infection
-
Downregulates innate immune response and reduces host resistance to viral infection
[102,103]
PDPNORF3
-
Incorporated into HIV released from HEK293T, required for efficient binding to attachment factor CLEC-2
[104]
IL27RAORF3
-
IL-27 enables optimal antiviral immunity early and late after persistent virus infection
[105,106]
ICOSORF3
-
Enhances all basic T cell responses
-
Antibody responses affected for LCMV, vesicular stomatitis virus (VSV) and influenza in ICOS-knockout mice
[107]
SLITRK3ORF3
-
Downregulated in cerebrospinal fluid in patients with varicella zoster virus (VZV) meningitis
-
Downregulated in HCV+ liver tissues (cirrhotic state) compared to normal adjacent tissues
[108,109]
FCGR2BORF3
-
Upregulated upon HBsAg-anti-HBs complex treatment in dendritic cells from an HBsAg transgenic mouse model and peripheral B cells from patients with chronic hepatitis B
-
Upregulation in patients with mild COVID-19 compared to asymptomatic cases
-
Simian virus vaccine-induced Abs form virus-specific immune complexes upon reaction with challenge virus and interact with FCGR2B to downregulate innate immune and inflammatory responses
[110,111,112]
CD7ORF3
-
Downregulation of CD7 in combination with activated CD8+ T-cell population might be attributed to acute EBV infection
-
Important for the HIV-1 fusion process that results in syncytium formation
-
HTLV-I results in loss of CD7 expression, further inducing Akt/Bad phosphorylation, which generates resistance to apoptosis
-
Upregulated upon IBDV infection
[113,114,115,116]
SLC9B2ORF3
-
Upregulated in cells treated with SARS-CoV-2 nucleocapsid protein
[117]
LPAR1ORF3
-
Ligand (LPA) concentrations are upregulated upon HCV or SARS-CoV-2, binding of LPA to LPAR1 restricts type I and type III IFN.
[118]
RBPMSORF3, ORF4
-
Identified as a host stimulatory factor for West Nile Virus (WNV) by RNAi screen.
-
Interacts with EBV proteins EBNA3B, BALF4 and BDLF4 and acts as latent–lytic switch in EBV by negatively interacting with STAT3
[119,120]
CMKLR1ORF3
-
Co-receptor for several SIV strains and HIV-1 strain
-
Lower levels of CMKLR1 in HBV HCC tissues
[121,122]
APLNRORF3
-
Alternative coreceptor with CD4 for HIV-1
[123]
SLCO1B1ORF3
-
Significant association with survival in HBV-mediated HCC
[124]
PRRS1ORF3
-
Related to genetic variant and pancreatitis in HEV-infected individual
-
Protein associated with the Pseudorabies virion
[125,126]
EPB41L3ORF3
-
Associated with asthma in RSV latent infection
-
Differentially expressed in blood of older individuals that might predispose to severe cases of COVID-19 with vascular and immune alterations
[127,128]
CACNG1ORF3
-
Expression levels downregulated in SH-SY5Y cells infected with VZV
[129]
DNAJC14ORF3
-
Pivotal cellular cofactor for replication and maintenance of pestiviruses, interacts with viral protein NS2
-
Interacts with members of the Flaviviridae family and inhibits viral replication
[130,131]
SLC2A5ORF3
-
Differentially expressed after SARS-CoV-2 infection
[132]
RTN1ORF3
-
Previous interaction with HEV ORF1 and RdRp
-
Identified as a regulator of HCV propagation
[133]
STARD3ORF3
-
Host dependency factor for HCV, affects entry and replication
[134]
CXCR6ORF3
-
Co-receptor for HIV-1 and HIV-2
-
Influences inflammatory response in context of influenza virus
-
NK cell memory of viruses depends on this receptor
-
Effector T cells in the liver express more CXCR6
[135,136,137,138]
CST4ORF3
-
Upregulated in nasal aspirates from influenza A virus (IAV)-infected children
[139]
FPR1ORF3
-
Upregulated after mono-HIV and HCV infection
-
Higher expression in HTLV-1 patients
-
Differentially expressed in bronchoalveolar lavage from severe-COVID-19 patients
[140,141,142]
TMEM176BORF3
-
Protective host factor in coronavirus infection by controlling inflammasome activation
[143]
RNF5ORF3
-
V protein of Newcastle disease virus (NDV) or PB1 of IAV recruit RNF5 to polyubiquitinate and degrade MAVS, inhibiting type I IFN
-
Sendai Virus and pseudorabies virus recruit RNF5 to inhibit STING-mediated antiviral immunity
[66,67,144,145]
CXADRORF3
-
Coxsackievirus and adenovirus receptor,
[44]
ZAR1LORF3
-
Upregulated in ZIKV-infected JEG-3 cells.
[146]
PDK1ORF3
-
JEV infection of neurons results in upregulation of PDK1 abundance and promotes neuronal apoptosis, which contributes to JEV morbidity and mortality
-
mRNA of PDKs are elevated upon HCV infection and result in promoting serine and glycine synthesis, thereby contributing to a favorable environment for HCV replication
[147,148]
TGIF1ORF3
-
downregulation in HCV-infected patients in context of liver fibrosis
-
upregulated upon SARS-CoV-2 infection
-
Bovine viral diarrhea virus-infected cells have reduced TGIF1 expression, which might cause gastrulation defects and embryonic lethality
[149,150,151]
FZD7ORF3
-
Overexpressed in HCC tumors, mostly related to chronic hepatitis B virus infection
[152]
SHARPINORF3, ORF4
-
PRRSV NSP1 impairs interaction between HOIP and SHARPIN, thereby reducing LUBAC-dependent ubiquitination of NEMO and suppressing NF-kB activation
-
LUBAC inhibits virus-induced interferon in case of Sendai virus and VSV infection
-
LUBAC is essential for induction of interferon in case of human norovirus infection
[58,59,153,154]
TNFRSF10AORF3
-
Identified as HIV-1 restriction factor in cell culture
-
Upregulation of TNFRSF10A (and related receptor and associated ligand) upon RSV infection; linked with apoptosis in early viral replicative cycle
[155,156,157]
SCN2BORF3
-
Identified as druggable gene for viral hemorrhagic septicemia virus (VHSV)
-
Differentially expressed in HIV-infected individuals suffering from HIV-encephalitis and proposed as potential therapeutic target
[158,159]
OCT1ORF3
-
Downregulated protein abundance and mRNA levels in HCV-infected liver samples.
[160]
PLPPR1ORF3
-
Upregulated expression in cells expressing flavivirus capsid proteins
[161]
ADGRF5ORF3
-
Upregulated in glioblastoma cells susceptible to infection with a strain of measles virus
[162]
ENTPD2ORF3
-
Reduced levels in COVID-19 patients
[163]
SLC38A11ORF3
-
Differentially expressed in influenza-stimulated B-cells
[164]
SLC44A5ORF3
-
Interacts with RSV NS1, also identified with MAPPIT and KISS
[5]
EPB41L2ORF3
-
Enriched in proteomic study of HCV core + 1/ARFP protein, possibly related to cirrhosis
-
Identified in a proteomic screen as a cellular factor incorporated during Nipah virus budding
[165,166]
GPBAR1ORF3
-
Zebrafish orthologue involved in antiviral activity against spring viremia of carp virus (SVCV)
-
Downregulated expression in relation to SARS-CoV-2, but upregulated in influenza, SARS-CoV-1, RSV and rhinovirus
-
Identified as ISG with increased expression after viral infection. Deficiency results in increased VSV and Herpe simplex virus (HSV)-1 replication, probably by facilitating IFN production through Akt/IRF3
[167,168,169]
LITAFORF3
-
Interacts with NS26 of grass carp reovirus (GCRV)
-
Alters the subcellular localization of frog virus 3 75L to late endosomes/lysosomes and co-localizes with LITAF.
-
Upregulation in chickens infected with highly pathogenic avian influenza virus, but downregulated in ducks (who show a milder infection compared to chickens)
[170,171,172]
APOMORF3
-
Levels of serum APOM in patients with HBV are elevated compared to healthy individuals; APOM promotor activity, mRNA and protein expression simulated in HBV-transfected cells, and reduces expression of HBV S and E proteins and synthesis of viral DNA
-
APOM is associated with HCV particles, as identified by affinity purification–mass spectrometry, and is required for HCV production; interacts with HCV E2 protein
[173,174]
SYPORF3
-
Borna disease virus (BDV) induces decrease in expression of SYP, followed by loss of cortical neurons
-
HSV-1-infected neurons have reduced SYP expression
-
Lower immunoreactivity of SYP in the cortex resulting from feline immunodeficiency virus co- and superinfection
[175,176,177]
NEUROD2ORF3
-
Downregulated in Zika virus (ZIKV)-infected brains having neurodevelopmental phenotypes
[178]
RCHY1ORF3, ORF4
-
Interacts with SARS-unique domain (SUD) and papain-like protease (PLpro), association increases RCHY1 stability and increases its ubiquitination function and degradation of p53
-
Host factor restricting IAV infection by modulating ubiquitination of nucleoproteins
-
ORF3 protein of porcine circovirus type 2 competes with p53 in binding RCHY1 to deregulate p53 and apoptosis
-
Interacts with C-terminal domain of the measles virus phosphoprotein in Y2H, thereby stabilizing RCHY2 by preventing its ubiquitination
[179,180,181,182]
S1PR3ORF3
-
S1PR3 mRNA overexpressed in EBV-positive nasopharyngeal carcinoma (NPC) patient-derived xenografts and primary NPC tissues; knockdown suppresses AKT activation and Sphingosine-1-phosphate induced migration of NPC cells
[183]
ACVR1BORF3
-
HCMV-induced miRNA (miRUL148D) targets ACVR1B, specifically inhibits ACVR1B expression in latently infected cells to limit pro-inflammatory cytokine production
[184]
EFNB1ORF3
-
Receptor for Cedar virus
-
Strongly upregulated after infectious pancreatic necrosis virus (IPNV) infection of Chinook salmon embryonic cells
[185]
DNAJC7ORF3
-
Fowl adenovirus serotype 4 (FAdV-4) PPI method shows interaction HSP70 and co-factor DNAJC7, which negatively regulate viral replication. Hexon protein interaction with HSP70 and assistance of DNAJC7 results in suppression of Hexon protein through the autophagy pathway
-
The cap protein of Porcine circovirus type 2 interacts with DNAJC7 and allows one to inhibit PKR activation
[186,187]
CAV2ORF3
-
Together with other caveolae-associated proteins incorporated into Tiger frog virus (TFV), they might be a restriction factor that affects viral release
-
Host factor of the RSV matrix protein; siRNA knockdown reduces RSV virus production
-
Identified as a protein, together with other proteins associated with lipid rafts, associated with HCV-induced autophagosomes (to enhance viral replication)
[188,189,190]
VDAC3ORF3
-
HBV X protein binds VDAC3 and alters mitochondrial membrane potential. It also activates transcription factors including STAT3 and NFkB
-
Interacts with EV71 2B protein, to enhance mitochondrial reactive oxygen species generation, which promotes viral replication.
[191,192]
SPNORF3
-
IAV binds to human neutrophils (PMNs), mediated by glycoproteins including SPN
-
SPN-deficient CD4+ T-cells have an enhanced susceptibility to HIV
[193,194]
MAGORF3
-
Binds HIV-1 gp120; may serve as receptor in peripheral nerve and be associated with neuropathy
-
Binds VZV glycoprotein to mediate entry of neurotropic herpesviruses
[195,196]
BSGORF3
-
Involved in infection of measles and HCMV
[45,46]
BTN3A2ORF3
-
Polymorphisms of this gene may alter susceptibility to HCV infection
[197]
CRB3ORF3
-
CRB3 is part of complex (CRB3-PALS1-PATJ). The SARS-CoV-2 E protein competes with CRB3 for binding PALS1, which might play a role in disrupting lung epithelium in SARS patients
[198]
NUMBORF3
-
ZIKV induces degradation of the NUMB protein, which might contribute to neurogenesis
-
The HBV e antigen and its precursors can interact with NUMB to impair stability of p53 and suppress its dependent apoptosis, thereby contributing to hepatocellular carcinoma
[199,200]
NECTIN4ORF3
-
Receptor for measles virus
[48]
TSG101ORF3, ORF4
-
Well known as interaction partner of ORF3 to mediate viral release
[15]
PTH1RORF3
-
HTLV-1 infection upregulates PTH1R in lymphocytes, indicating a potential autocrine role for PTHrP
[201]
PTGER4ORF3
-
Receptor for PGE2, which is a modulator for viral infections
[202]
SURF4ORF3
-
involved in the replication of HCV by interacting with the HCV NS4B protein
[203]
CX3CL1ORF3
-
upregulated upon HEV infection in primary human hepatocytes
-
HIV Tat protein suppresses CX3CL1-CX3CR1 axis in microglia, which serve as viral reservoirs and producers of neurotoxins
-
Differentially expressed in HVB-infected hepatoma cells
[204,205,206]
CYBRD1ORF3
-
SNPs in CYBRD1 related with higher prevalence of HCC in patients with chronic HCV
[207]
HAVCR1ORF3
-
Well-known receptor for enveloped and non-enveloped viruses by binding phosphatidylserine, including HEV
[49,50]
AGTR1ORF3
-
During SARS coronavirus-2/SARS-CoV-2 infection, it is able to recognize and internalize the complex formed by secreted ACE2 and SARS-CoV-2 spike protein through DNM2/dynamin 2-dependent endocytosis
[47]
BANF1ORF3
-
Antiviral effector against vaccinia virus due to its DNA-binding activity, vaccina’s B1 kinase can inactivate BANF1 by phosphorylation
-
Interferes with HSV DNA replication
-
HBV inhibits BANF1, by downregulation of miR-203 by the X protein
[208,209,210]
BRAFORF3
-
Cellular interactor of NS3 from bluetongue virus
-
Dysregulated expression in Merker cell polyomavirus positive samples from non-small cell lung cancer
[211,212]
BTN3A3ORF3
-
Polymorphisms of this gene may alter susceptibility to HCV infection
-
Potent inhibitor of avian influenza A virus (but not human IAV) by restricting its replication
[197,213]
CAMPORF3
-
Inhibits DENV-2 at the stage of entry into cells by binding E protein
-
Inhibits infection of recombinant virus pseudotypes with Ebola virus (EBOV) glycoprotein and wild-type EBOV by targeting the endosomal entry step
-
Differentially regulated after IAV infection
[214,215,216]
CD6ORF3
-
Overexpressed in CD8+ T-cells in lymphoid tissues and BAL from chronic SIV infection
[217]
CUL2ORF3
-
NLRC5 recruits CUL2 to ubiquitinate NS3 protease and degrade it, thereby restricting DENV infection
-
CUL2 knockdown inhibited SAMHD1 loss during CMV infection, which functions as a restriction factor
-
Vif protein of Bovine immunodeficiency virus (BIV) interacts with CUL2 to degrade restrictive bovine A3 (APOBEC3) proteins
[218,219,220]
AGO2ORF3
-
Upon SARS-CoV-2 infection, associates with viral miRNA-like small RNA, CoV2-miR-O7a, and may repress mRNAs, such as BATF2, to evade the IFN response
-
Inhibition of AGO2 results in decreases encephalitic alphavirus replication
-
AGO2 knockout promotes Seneca Virus A replication
-
H5N1 influenza viral infection reduces AGO2 distribution in A549 cells, to promote IFN-β expression and increase host survival
[221,222,223,224]
FXYD2ORF3
-
Upregulated in HIV-2-infected cells, downregulated in HIV-1-infected PBMCs
[225]
H3C8ORF3
-
Histones mobilized during HSV-1 infection and relates histone dynamics to assembly of viral chromatin
-
VZV IE63 protein interacts with ASF1 and increases its binding to H3C8, which may help regulating transcription of viral or cellular genes during infection
[226,227]
H4C1ORF3
-
Binds to IAV and aggregates viral particles; potentiates IAV induced neutrophil respiratory burst responses
-
TIP60 inhibits HBV replication and localized HBV cccDNA chromatin complex catalyzed H4C1 acetylation. This all might regulate HBV chromatin structure
[228,229]
IL1R2ORF3
-
May play a crucial role in innate and adaptive immune responses in pathogenic avian influenza virus
-
High expression positively correlated with viral shedding time in SARS-CoV-2 patients
[230,231]
KIF18AORF3
-
Expression of KIF18A is increased in host cells after IAV infection, which provide a beneficial role in replication and a potential therapeutic target
[232]
KIR3DL2ORF3
-
Could be associated with immunity against HBV infection
-
Expression may be associated with more rapid progression of chronic HCV fibrosis in patients
[233,234]
LAIR1ORF3
-
Chronic EBV infection is associated with low LAIR expression on NK cells
-
LAIR knockout mice show enhanced airway inflammation and increased neutrophil and lymphocyte recruitment after RSV infection
[235,236]
MMP2ORF3
-
Together with MMP9, it mediates sCD100 release, which has an important role in regulating intrahepatic anti-HBV CD8 T-cell responses
-
Increased in long-term experimental HSV encephalitis
[237]
NAA60ORF3
-
N-terminal acetyltransferase has proviral properties during IAV infection by interfering with interferon alpha signaling
[238]
NPAS4ORF3
-
Expression level of this transcription factor severely decreased after ZIKV infection of fetal murine neurons
[239]
PIWIL4ORF3
-
Promotes HIV-1 latency by imposing repressive marks at the HIV-1 5′ long terminal repeat
[240]
RAB43ORF3
-
Important for HSV-1 assembly
[241]
RAB9AORF3
-
Differentially expressed in lymphoblastoid cell lines after EBV infection compared to resting B cells
-
Mediator of HPV16 trans-golgi network transit during infection
-
Required for replication of HIV-1, filoviruses and measles virus, probably associated with late-endosome-to plasma membrane vesicular transpart in viral assembly
[242,243,244]
SEMA4DORF3
-
miR-125b (which is downregulated in J avian leukosis virus) and its target SEMA4D play an important role in aggressive growth of HP45 cells induced by avian leukosis viruses
-
loss of SEMA4D expression might play an important role in dysfunctional immunity in HIV-1 infection
-
IFN-alpha enhanced SEMA4D/Plexin-B1/B2 interactions plays an important role in promoting NK functions in chronic hepatitis C patients
[245,246,247]
STAU2ORF3
-
STAU2 interacts with NS1 from avian influenza virus to promote viral replication by enhancing NS1 mRNA nuclear export
[248]
TREX1ORF3
-
3′-to 5′ DNA exonuclease, digests excess HIV DNA to inhibit IFN expression
[249]
MSI2ORF4
-
Promotes translation of multiple IRES-containing oncogenes as well as HCV IRES; inhibition of MSI2-RNA binding reduced HCV IRES activity, viral replication and liver hyperplasia in humanized mice
[250]
CAMK2AORF4
-
HBV replication suppresses CAMK2 activity, and CAMK2 upregulation suppresses HBV replication from cccDNA via AMPK and the AKT/mTOR pathway
-
EV71 VP1 protein activates CAMK2A to phosphorylate vimentin, which is required for virus replication
-
African swine fever virus results in activation of CAMK2A and phosphorylation of vimentin, which may be necessary for cage formation surrounding virus factories
[251,252,253]
DDAH2ORF4
-
DDAH2 induces nitric oxide and Drp1 activation, which blocks innate immune responses mediated by MAVS upon viral infection
-
Differentially expressed protein after North-American type Porcine reproductive and respiratory syndrome virus (PRRSV) infection
[254,255]
LBHORF4
-
LMP1 from EBV plays a fundamental role in NPC; LBH is downregulated in NPC tissues. LBH normally induces NPC cell cycle arrest and growth of tumors by downregulating NFkB transcriptional activity
[256]
TRAF1ORF4
-
Interacts with HEV RdRp
-
Lost from HIV- or LCMV-specific CD8 T cells in chronic phase of infection
-
TRAF1 expression may be upregulated by EBV LMP1 protein in NPC
[32,257,258]
DYNLT3ORF4
-
Interacts with L2 protein of HPV and is most likely required for transport purposes towards nucleus
[259]
GATAD2AORF4
-
Circular GATAD2A promotes replication of H1N1 IAV by inhibiting autophagy
[260]
PPP1CCORF4
-
Gene 7 of transmissible gastroenteritis virus (TGEV) mediates binding to PPP1CC and may counteract host antiviral response
[261]
CCT5ORF4
-
Cap protein of porcine circovirus type 2 (PCV2) could directly degrade CCT5
-
Interacts with avian influenza nucleoprotein (NP). Expression elevated upon infection, promotes nuclear export of NP and viral polymerase activity
[262,263]
ZFYVE1ORF4
-
Binds SARS-CoV-2 NSP6, mediates the replication organelle–lipid droplet association required to sustain viral replication
-
Required for HCV genome replication, probably by the formation of omegasomes that are used by HCV to generate double-membrane vesicles
[264,265]
DDX20ORF4
-
Interacts with EBNA2 and EBNA3C proteins from EBV. It is suggested EBNA2 targets spliceosome complex to release SMN proteins after binding DDX20. EBNA3C binding to DDX20 stabilizes the protein to form a complex with p53 to block transcriptional activity and apoptosis
-
Interacts with HIV-1 Vpr protein
[266]
HEY1ORF4
-
Increased expression in two human thyroid cancer lines transfected with EBNA2 from EBV
[267]
CEP72ORF4
-
Differentially expressed in HBV-derived HCC tumors
[268]
CEP55ORF4
-
Upregulated after HTLV-1 and Bovine leukemia virus (BLV) infection
[269]
KLHL20ORF4
-
KLHL20 polyubiquitinates SERINC5, which determines its expression on the plasma membrane, where it is packaged into HIV-1 virions for viral inhibition or downregulation by Nef via lysosomal degradation
[270]
NAB2ORF4
-
Early growth response gene 1 (egr1) can repress HSV-1 early gene expression through the recruitment of co-repressor NAB2
[271]
RBFOX2ORF4
-
Identified as hub gene in HCC patients with HBV or/and HCV infection
[272]
SH3GLB2ORF4
-
SH3GLB2 regulates lung homeostasis and recovery from severe IAV infection
[273]
PSME1ORF4
-
Expression level decreased in PRRSV-infected alveolar macrophages
[274]
MSI1ORF4
-
Interacts with ZIKV genome and promotes viral replication
[275]
FBXW11ORF4
-
Vpu interacts with tetherin to direct its FBXW11-dependent proteasomal degradation, thereby alleviating the blockade to the release of infectious virions
-
NSs eliminates the antiviral kinase PKR by recruitment of SCF-type E3 ubiquitin ligases containing FBXW11 and beta-TRCP1 as substrate recognition subunits. This antagonism of PKR by NSs (nonstructural protein) is essential for efficient RVFV replication in mammalian cells
[276,277]
BTRCORF4
-
EBV miR-BART10-3p targets BTRC and facilitates epithelial–mesenchymal transition of NPC
-
Interacts with HIV-1 Vpu and connects CD4 to the proteolytic machinery for degradation
[278,279]
RPS15AORF4
-
Previously identified as interactor of IRES like element in gt-1 HEV
-
HBV-encoded X antigen upregulates RPS15A. This stimulates cell growth and tumor formation, potentially participating in HCC development
[280,281]
PRKAR1BORF4
-
Upregulated after duck HBV infection in vitro
[282]
ADARORF4
-
Responsible for editing of Hepatitis delta virus (HDV) RNA to produce antigen-L, with key role in assembly of viral particles
-
HCV, VSV, Measles virus, and HIV-1 are all targets of ADAR
-
Has proviral or antiviral effects, editing-dependent or -independent
-
Enhances viral replication through suppression of EIF2AK2/PKR activation
[55,283]
CARD9ORF4
-
Upregulated in HEV-infected Huh-7 cells.
-
Recognition of viral RNA (VSV or EMCV) by RIG-I induces CARD9 and inflammasome signaling to produce IL-1β
-
CARD-deficient mice have impaired cytokine production upon LACV and CVB3 infection
[284,285,286]
PRKRAORF4
-
ORF virus encoded protein OV20.0 interacts with PRKRA, blocking PACT-mediated PKR activation
-
Us11 protein of HSV-1 binds PACT to inhibit PKR activation
-
VP35 of Marburg virus interacts with PACT but is not necessarily related to PKR inhibition
[287,288,289]
DESORF4
-
Interacts with the rabies virus matrix protein, which positively regulates virus infection
-
Theiler murine encephalitis virus (TMEV) virions associate with DES and vimentin. Following infection, the filament network rearranged into a shell like structure surrounding viral inclusion
-
HIV-1 protease cleaves DES besides myosin heavy chain, tropomyosin and an actin fraction, showing it can cause alterations in muscle cell ultrastructure that may be of relevance in infected individuals
[290,291,292]
IMPDH1ORF4
-
Knockdown suppresses human norovirus replication
[293]
ZMYM6ORF4
-
Downregulated in cells expressing HPV-derived oncoprotein E7
[294]
CHMP4CORF4
-
Plays a role in the integrity of the recycling endosomal network, unveiled through the dependence of HSV1 on these membranes
-
Plays a role in HIV-1 budding
-
Depletion reduces flavivirus propagation
[295,296,297]
BDV = Borna disease virus, BEFV = Bovine ephemeral fever, BIV = Bovine immunodeficiency virus, BLV = Bovine leukemia virus, DENV = Dengue virus, EBOV = Ebola virus, EBV = Epstein–Barr virus, EV = Enterovirus, FAdV = Fowl adenovirus, GCRV = grass carp reovirus, HBV = Hepatitis B virus, HCC = Hepatocellular carcinoma, HCMV = Human cytomegalovirus, HCV = Hepatitis C virus, HIV = Human immunodeficiency virus, HPV = Human papillomavirus, HSV = Herpes simplex virus, HTLV = Human T-lymphotropic virus, IAV = Influenza A virus, IPNV = infectious pancreatic necrosis virus, JEV = Japanese encephalitis virus, LCMV = Lymphocytic choriomeningitis virus, NDV = Newcastle disease virus, NPC = Nasopharyngeal carcinoma, PCV = Porcine circovirus, PEDV = Porcine epidemic diarrhea virus, PRRSV = Porcine reproductive and respiratory syndrome virus, RSV = Respiratory syncytial virus, SIV = Simian immunodeficiency virus, SVCV = spring viremia of carp virus, TFV = Tiger frog virus, TGEV = Transmissible gastroenteritis virus, TMEV = Theiler murine encephalitis virus, VHSV = Viral hemorrhagic septicemia virus, VSV = Vesicular stomatitis virus, VZV = Varicella zoster virus, WNV = West Nile virus, ZIKV = Zika virus.

References

  1. Titeca, K.; Lemmens, I.; Tavernier, J.; Eyckerman, S. Discovering cellular protein-protein interactions: Technological strategies and opportunities. Mass Spectrom. Rev. 2019, 38, 79–111. [Google Scholar] [CrossRef] [PubMed]
  2. Eyckerman, S.; Verhee, A.; der Heyden, J.V.; Lemmens, I.; Ostade, X.V.; Vandekerckhove, J.; Tavernier, J. Design and application of a cytokine-receptor-based interaction trap. Nat. Cell Biol. 2001, 3, 1114–1119. [Google Scholar] [CrossRef] [PubMed]
  3. Lievens, S.; Gerlo, S.; Lemmens, I.; De Clercq, D.J.; Risseeuw, M.D.; Vanderroost, N.; De Smet, A.S.; Ruyssinck, E.; Chevet, E.; Van Calenbergh, S.; et al. Kinase Substrate Sensor (KISS), a mammalian in situ protein interaction sensor. Mol. Cell. Proteom. MCP 2014, 13, 3332–3342. [Google Scholar] [CrossRef] [PubMed]
  4. Rolland, T.; Tasan, M.; Charloteaux, B.; Pevzner, S.J.; Zhong, Q.; Sahni, N.; Yi, S.; Lemmens, I.; Fontanillo, C.; Mosca, R.; et al. A proteome-scale map of the human interactome network. Cell 2014, 159, 1212–1226. [Google Scholar] [CrossRef] [PubMed]
  5. Van Royen, T.; Sedeyn, K.; Moschonas, G.D.; Toussaint, W.; Vuylsteke, M.; Van Haver, D.; Impens, F.; Eyckerman, S.; Lemmens, I.; Tavernier, J.; et al. An Unexpected Encounter: Respiratory Syncytial Virus Nonstructural Protein 1 Interacts with Mediator Subunit MED25. J. Virol. 2022, 96, e0129722. [Google Scholar] [CrossRef] [PubMed]
  6. Van Schoubroeck, B.; Van Acker, K.; Dams, G.; Jochmans, D.; Clayton, R.; Berke, J.M.; Lievens, S.; Van der Heyden, J.; Tavernier, J. MAPPIT as a high-throughput screening assay for modulators of protein-protein interactions in HIV and HCV. Methods Mol. Biol. 2012, 812, 295–307. [Google Scholar] [CrossRef]
  7. Webb, G.W.; Dalton, H.R. Hepatitis E: An expanding epidemic with a range of complications. Clin. Microbiol. Infect. Off. Publ. Eur. Soc. Clin. Microbiol. Infect. Dis. 2020, 26, 828–832. [Google Scholar] [CrossRef]
  8. Zhu, F.C.; Zhang, J.; Zhang, X.F.; Zhou, C.; Wang, Z.Z.; Huang, S.J.; Wang, H.; Yang, C.L.; Jiang, H.M.; Cai, J.P.; et al. Efficacy and safety of a recombinant hepatitis E vaccine in healthy adults: A large-scale, randomised, double-blind placebo-controlled, phase 3 trial. Lancet 2010, 376, 895–902. [Google Scholar] [CrossRef]
  9. Sayed, I.M.; Vercouter, A.S.; Abdelwahab, S.F.; Vercauteren, K.; Meuleman, P. Is hepatitis E virus an emerging problem in industrialized countries? Hepatology 2015, 62, 1883–1892. [Google Scholar] [CrossRef]
  10. Kenney, S.P.; Meng, X.J. Hepatitis E Virus Genome Structure and Replication Strategy. Cold Spring Harb. Perspect. Med. 2019, 9. [Google Scholar] [CrossRef]
  11. Montpellier, C.; Wychowski, C.; Sayed, I.M.; Meunier, J.C.; Saliou, J.M.; Ankavay, M.; Bull, A.; Pillez, A.; Abravanel, F.; Helle, F.; et al. Hepatitis E Virus Lifecycle and Identification of 3 Forms of the ORF2 Capsid Protein. Gastroenterology 2018, 154, 211–223.e8. [Google Scholar] [CrossRef] [PubMed]
  12. Yin, X.; Ying, D.; Lhomme, S.; Tang, Z.; Walker, C.M.; Xia, N.; Zheng, Z.; Feng, Z. Origin, antigenicity, and function of a secreted form of ORF2 in hepatitis E virus infection. Proc. Natl. Acad. Sci. USA 2018, 115, 4773–4778. [Google Scholar] [CrossRef] [PubMed]
  13. Ding, Q.; Heller, B.; Capuccino, J.M.; Song, B.; Nimgaonkar, I.; Hrebikova, G.; Contreras, J.E.; Ploss, A. Hepatitis E virus ORF3 is a functional ion channel required for release of infectious particles. Proc. Natl. Acad. Sci. USA 2017, 114, 1147–1152. [Google Scholar] [CrossRef] [PubMed]
  14. Nan, Y.; Ma, Z.; Wang, R.; Yu, Y.; Kannan, H.; Fredericksen, B.; Zhang, Y.J. Enhancement of interferon induction by ORF3 product of hepatitis E virus. J. Virol. 2014, 88, 8696–8705. [Google Scholar] [CrossRef] [PubMed]
  15. Nagashima, S.; Takahashi, M.; Jirintai, S.; Tanaka, T.; Nishizawa, T.; Yasuda, J.; Okamoto, H. Tumour susceptibility gene 101 and the vacuolar protein sorting pathway are required for the release of hepatitis E virions. J. Gen. Virol. 2011, 92, 2838–2848. [Google Scholar] [CrossRef] [PubMed]
  16. Nair, V.P.; Anang, S.; Subramani, C.; Madhvi, A.; Bakshi, K.; Srivastava, A.; Shalimar; Nayak, B.; Ranjith Kumar, C.T.; Surjit, M. Endoplasmic Reticulum Stress Induced Synthesis of a Novel Viral Factor Mediates Efficient Replication of Genotype-1 Hepatitis E Virus. PLoS Pathog. 2016, 12, e1005521. [Google Scholar] [CrossRef]
  17. Lievens, S.; Van der Heyden, J.; Masschaele, D.; De Ceuninck, L.; Petta, I.; Gupta, S.; De Puysseleyr, V.; Vauthier, V.; Lemmens, I.; De Clercq, D.J.; et al. Proteome-scale Binary Interactomics in Human Cells. Mol. Cell. Proteom. MCP 2016, 15, 3624–3639. [Google Scholar] [CrossRef]
  18. Masschaele, D.; Gerlo, S.; Lemmens, I.; Lievens, S.; Tavernier, J. KISS: A Mammalian Two-Hybrid Method for In Situ Analysis of Protein-Protein Interactions. Methods Mol. Biol. 2018, 1794, 269–278. [Google Scholar] [CrossRef]
  19. Lievens, S.; Vanderroost, N.; Van der Heyden, J.; Gesellchen, V.; Vidal, M.; Tavernier, J. Array MAPPIT: High-throughput interactome analysis in mammalian cells. J. Proteome Res. 2009, 8, 877–886. [Google Scholar] [CrossRef]
  20. Masschaele, D.; De Ceuninck, L.; Wauman, J.; Defever, D.; Stenner, F.; Lievens, S.; Peelman, F.; Tavernier, J. RNF41 interacts with the VPS52 subunit of the GARP and EARP complexes. PLoS ONE 2017, 12, e0178132. [Google Scholar] [CrossRef]
  21. Bardou, P.; Mariette, J.; Escudie, F.; Djemiel, C.; Klopp, C. jvenn: An interactive Venn diagram viewer. BMC Bioinform. 2014, 15, 293. [Google Scholar] [CrossRef] [PubMed]
  22. Ran, F.A.; Hsu, P.D.; Wright, J.; Agarwala, V.; Scott, D.A.; Zhang, F. Genome engineering using the CRISPR-Cas9 system. Nat. Protoc. 2013, 8, 2281–2308. [Google Scholar] [CrossRef] [PubMed]
  23. Sayed, I.M.; Verhoye, L.; Cocquerel, L.; Abravanel, F.; Foquet, L.; Montpellier, C.; Debing, Y.; Farhoudi, A.; Wychowski, C.; Dubuisson, J.; et al. Study of hepatitis E virus infection of genotype 1 and 3 in mice with humanised liver. Gut 2017, 66, 920–929. [Google Scholar] [CrossRef] [PubMed]
  24. Zheng, Z.Z.; Miao, J.; Zhao, M.; Tang, M.; Yeo, A.E.; Yu, H.; Zhang, J.; Xia, N.S. Role of heat-shock protein 90 in hepatitis E virus capsid trafficking. J. Gen. Virol. 2010, 91, 1728–1736. [Google Scholar] [CrossRef]
  25. Shen, Q.; Zhang, W.; Kang, Y.; Chen, Y.; Cui, L.; Yang, Z.; Hua, X. HEV-Capsid Protein Interacts With Cytochrome P4502C8 and Retinol-Binding Protein 4. Hepat. Mon. 2011, 11, 913–917. [Google Scholar] [CrossRef] [PubMed]
  26. Tian, Y.; Huang, W.; Yang, J.; Wen, Z.; Geng, Y.; Zhao, C.; Zhang, H.; Wang, Y. Systematic identification of hepatitis E virus ORF2 interactome reveals that TMEM134 engages in ORF2-mediated NF-kappaB pathway. Virus Res. 2017, 228, 102–108. [Google Scholar] [CrossRef]
  27. Zhang, L.; Tian, Y.; Wen, Z.; Zhang, F.; Qi, Y.; Huang, W.; Zhang, H.; Wang, Y. Asialoglycoprotein receptor facilitates infection of PLC/PRF/5 cells by HEV through interaction with ORF2. J. Med. Virol. 2016, 88, 2186–2195. [Google Scholar] [CrossRef]
  28. Geng, Y.; Yang, J.; Huang, W.; Harrison, T.J.; Zhou, Y.; Wen, Z.; Wang, Y. Virus host protein interaction network analysis reveals that the HEV ORF3 protein may interrupt the blood coagulation process. PLoS ONE 2013, 8, e56320. [Google Scholar] [CrossRef]
  29. Tyagi, S.; Surjit, M.; Roy, A.K.; Jameel, S.; Lal, S.K. The ORF3 protein of hepatitis E virus interacts with liver-specific alpha1-microglobulin and its precursor alpha1-microglobulin/bikunin precursor (AMBP) and expedites their export from the hepatocyte. J. Biol. Chem. 2004, 279, 29308–29319. [Google Scholar] [CrossRef]
  30. Venkatesan, K.; Rual, J.F.; Vazquez, A.; Stelzl, U.; Lemmens, I.; Hirozane-Kishikawa, T.; Hao, T.; Zenkner, M.; Xin, X.; Goh, K.I.; et al. An empirical framework for binary interactome mapping. Nat. Methods 2009, 6, 83–90. [Google Scholar] [CrossRef]
  31. LeDesma, R.; Nimgaonkar, I.; Ploss, A. Hepatitis E Virus Replication. Viruses 2019, 11, 719. [Google Scholar] [CrossRef] [PubMed]
  32. Ojha, N.K.; Lole, K.S. Hepatitis E virus ORF1 encoded non structural protein-host protein interaction network. Virus Res. 2016, 213, 195–204. [Google Scholar] [CrossRef] [PubMed]
  33. Hervouet, K.; Ferrie, M.; Ankavay, M.; Montpellier, C.; Camuzet, C.; Alexandre, V.; Dembele, A.; Lecoeur, C.; Foe, A.T.; Bouquet, P.; et al. An Arginine-Rich Motif in the ORF2 capsid protein regulates the hepatitis E virus lifecycle and interactions with the host cell. PLoS Pathog. 2022, 18, e1010798. [Google Scholar] [CrossRef] [PubMed]
  34. Netherton, C.; Moffat, K.; Brooks, E.; Wileman, T. A guide to viral inclusions, membrane rearrangements, factories, and viroplasm produced during virus replication. Adv. Virus Res. 2007, 70, 101–182. [Google Scholar] [CrossRef] [PubMed]
  35. Mercer, J.; Schelhaas, M.; Helenius, A. Virus entry by endocytosis. Annu. Rev. Biochem. 2010, 79, 803–833. [Google Scholar] [CrossRef] [PubMed]
  36. Matsuda, M.; Suzuki, R.; Kataoka, C.; Watashi, K.; Aizaki, H.; Kato, N.; Matsuura, Y.; Suzuki, T.; Wakita, T. Alternative endocytosis pathway for productive entry of hepatitis C virus. J. Gen. Virol. 2014, 95, 2658–2667. [Google Scholar] [CrossRef] [PubMed]
  37. Rivera-Serrano, E.E.; Gonzalez-Lopez, O.; Das, A.; Lemon, S.M. Cellular entry and uncoating of naked and quasi-enveloped human hepatoviruses. eLife 2019, 8, e43983. [Google Scholar] [CrossRef]
  38. Feng, Z.; Hensley, L.; McKnight, K.L.; Hu, F.; Madden, V.; Ping, L.; Jeong, S.H.; Walker, C.; Lanford, R.E.; Lemon, S.M. A pathogenic picornavirus acquires an envelope by hijacking cellular membranes. Nature 2013, 496, 367–371. [Google Scholar] [CrossRef]
  39. Raab-Traub, N.; Dittmer, D.P. Viral effects on the content and function of extracellular vesicles. Nat. Reviews. Microbiol. 2017, 15, 559–572. [Google Scholar] [CrossRef]
  40. Stravalaci, M.; Pagani, I.; Paraboschi, E.M.; Pedotti, M.; Doni, A.; Scavello, F.; Mapelli, S.N.; Sironi, M.; Perucchini, C.; Varani, L.; et al. Recognition and inhibition of SARS-CoV-2 by humoral innate immunity pattern recognition molecules. Nat. Immunol. 2022, 23, 275–286. [Google Scholar] [CrossRef]
  41. Dansako, H.; Yamane, D.; Welsch, C.; McGivern, D.R.; Hu, F.; Kato, N.; Lemon, S.M. Class A scavenger receptor 1 (MSR1) restricts hepatitis C virus replication by mediating toll-like receptor 3 recognition of viral RNAs produced in neighboring cells. PLoS Pathog. 2013, 9, e1003345. [Google Scholar] [CrossRef] [PubMed]
  42. Bonaparte, M.I.; Dimitrov, A.S.; Bossart, K.N.; Crameri, G.; Mungall, B.A.; Bishop, K.A.; Choudhry, V.; Dimitrov, D.S.; Wang, L.F.; Eaton, B.T.; et al. Ephrin-B2 ligand is a functional receptor for Hendra virus and Nipah virus. Proc. Natl. Acad. Sci. USA 2005, 102, 10652–10657. [Google Scholar] [CrossRef] [PubMed]
  43. Wilen, C.B.; Tilton, J.C.; Doms, R.W. HIV: Cell binding and entry. Cold Spring Harb. Perspect. Med. 2012, 2. [Google Scholar] [CrossRef] [PubMed]
  44. Excoffon, K. The coxsackievirus and adenovirus receptor: Virological and biological beauty. FEBS Lett. 2020, 594, 1828–1837. [Google Scholar] [CrossRef] [PubMed]
  45. Vanarsdall, A.L.; Pritchard, S.R.; Wisner, T.W.; Liu, J.; Jardetzky, T.S.; Johnson, D.C. CD147 Promotes Entry of Pentamer-Expressing Human Cytomegalovirus into Epithelial and Endothelial Cells. mBio 2018, 9, e00781-18. [Google Scholar] [CrossRef] [PubMed]
  46. Watanabe, A.; Yoneda, M.; Ikeda, F.; Terao-Muto, Y.; Sato, H.; Kai, C. CD147/EMMPRIN acts as a functional entry receptor for measles virus on epithelial cells. J. Virol. 2010, 84, 4183–4193. [Google Scholar] [CrossRef] [PubMed]
  47. Yeung, M.L.; Teng, J.L.L.; Jia, L.; Zhang, C.; Huang, C.; Cai, J.P.; Zhou, R.; Chan, K.H.; Zhao, H.; Zhu, L.; et al. Soluble ACE2-mediated cell entry of SARS-CoV-2 via interaction with proteins related to the renin-angiotensin system. Cell 2021, 184, 2212–2228.e12. [Google Scholar] [CrossRef]
  48. Muhlebach, M.D.; Mateo, M.; Sinn, P.L.; Prufer, S.; Uhlig, K.M.; Leonard, V.H.; Navaratnarajah, C.K.; Frenzke, M.; Wong, X.X.; Sawatsky, B.; et al. Adherens junction protein nectin-4 is the epithelial receptor for measles virus. Nature 2011, 480, 530–533. [Google Scholar] [CrossRef]
  49. Amara, A.; Mercer, J. Viral apoptotic mimicry. Nat. Rev. Microbiol. 2015, 13, 461–469. [Google Scholar] [CrossRef]
  50. Corneillie, L.; Lemmens, I.; Montpellier, C.; Ferrie, M.; Weening, K.; Van Houtte, F.; Hanoulle, X.; Cocquerel, L.; Amara, A.; Tavernier, J.; et al. The phosphatidylserine receptor TIM1 promotes infection of enveloped hepatitis E virus. Cell. Mol. Life Sci. CMLS 2023, 80, 326. [Google Scholar] [CrossRef]
  51. Dong, C.; Zafrullah, M.; Mixson-Hayden, T.; Dai, X.; Liang, J.; Meng, J.; Kamili, S. Suppression of interferon-alpha signaling by hepatitis E virus. Hepatology 2012, 55, 1324–1332. [Google Scholar] [CrossRef] [PubMed]
  52. He, M.; Wang, M.; Huang, Y.; Peng, W.; Zheng, Z.; Xia, N.; Xu, J.; Tian, D. The ORF3 Protein of Genotype 1 Hepatitis E Virus Suppresses TLR3-induced NF-kappaB Signaling via TRADD and RIP1. Sci. Rep. 2016, 6, 27597. [Google Scholar] [CrossRef] [PubMed]
  53. Ju, X.; Ding, Q. Hepatitis E Virus Assembly and Release. Viruses 2019, 11, 539. [Google Scholar] [CrossRef] [PubMed]
  54. Subramani, C.; Nair, V.P.; Anang, S.; Mandal, S.D.; Pareek, M.; Kaushik, N.; Srivastava, A.; Saha, S.; Shalimar; Nayak, B.; et al. Host-Virus Protein Interaction Network Reveals the Involvement of Multiple Host Processes in the Life Cycle of Hepatitis E Virus. mSystems 2018, 3. [Google Scholar] [CrossRef]
  55. Samuel, C.E. Adenosine deaminases acting on RNA (ADARs) are both antiviral and proviral. Virology 2011, 411, 180–193. [Google Scholar] [CrossRef] [PubMed]
  56. Garcia, M.A.; Gil, J.; Ventoso, I.; Guerra, S.; Domingo, E.; Rivas, C.; Esteban, M. Impact of protein kinase PKR in cell biology: From antiviral to antiproliferative action. Microbiol. Mol. Biol. Rev. MMBR 2006, 70, 1032–1060. [Google Scholar] [CrossRef] [PubMed]
  57. Edilova, M.I.; Abdul-Sater, A.A.; Watts, T.H. TRAF1 Signaling in Human Health and Disease. Front. Immunol. 2018, 9, 2969. [Google Scholar] [CrossRef]
  58. MacDuff, D.A.; Baldridge, M.T.; Qaqish, A.M.; Nice, T.J.; Darbandi, A.D.; Hartley, V.L.; Peterson, S.T.; Miner, J.J.; Iwai, K.; Virgin, H.W. HOIL1 Is Essential for the Induction of Type I and III Interferons by MDA5 and Regulates Persistent Murine Norovirus Infection. J. Virol. 2018, 92. [Google Scholar] [CrossRef]
  59. Belgnaoui, S.M.; Paz, S.; Samuel, S.; Goulet, M.L.; Sun, Q.; Kikkert, M.; Iwai, K.; Dikic, I.; Hiscott, J.; Lin, R. Linear ubiquitination of NEMO negatively regulates the interferon antiviral response through disruption of the MAVS-TRAF3 complex. Cell Host Microbe 2012, 12, 211–222. [Google Scholar] [CrossRef]
  60. Inn, K.S.; Gack, M.U.; Tokunaga, F.; Shi, M.; Wong, L.Y.; Iwai, K.; Jung, J.U. Linear ubiquitin assembly complex negatively regulates RIG-I- and TRIM25-mediated type I interferon induction. Mol. Cell 2011, 41, 354–365. [Google Scholar] [CrossRef]
  61. Yin, X.; Li, X.; Ambardekar, C.; Hu, Z.; Lhomme, S.; Feng, Z. Hepatitis E virus persists in the presence of a type III interferon response. PLoS Pathog. 2017, 13, e1006417. [Google Scholar] [CrossRef] [PubMed]
  62. Sooryanarain, H.; Heffron, C.L.; Meng, X.J. The U-Rich Untranslated Region of the Hepatitis E Virus Induces Differential Type I and Type III Interferon Responses in a Host Cell-Dependent Manner. mBio 2020, 11. [Google Scholar] [CrossRef] [PubMed]
  63. Wu, X.; Dao Thi, V.L.; Liu, P.; Takacs, C.N.; Xiang, K.; Andrus, L.; Gouttenoire, J.; Moradpour, D.; Rice, C.M. Pan-Genotype Hepatitis E Virus Replication in Stem Cell-Derived Hepatocellular Systems. Gastroenterology 2018, 154, 663–674.e667. [Google Scholar] [CrossRef] [PubMed]
  64. Khan, M.; Syed, G.H.; Kim, S.J.; Siddiqui, A. Hepatitis B Virus-Induced Parkin-Dependent Recruitment of Linear Ubiquitin Assembly Complex (LUBAC) to Mitochondria and Attenuation of Innate Immunity. PLoS Pathog. 2016, 12, e1005693. [Google Scholar] [CrossRef] [PubMed]
  65. Zeng, Y.; Xu, S.; Wei, Y.L.; Zhang, X.G.; Wang, Q.; Jia, Y.N.; Wang, W.B.; Han, L.; Chen, Z.S.; Wang, Z.X.; et al. The PB1 protein of influenza A virus inhibits the innate immune response by targeting MAVS for NBR1-mediated selective autophagic degradation. PLoS Pathog. 2021, 17, e1009300. [Google Scholar] [CrossRef]
  66. Sun, Y.; Zheng, H.; Yu, S.; Ding, Y.; Wu, W.; Mao, X.; Liao, Y.; Meng, C.; Ur Rehman, Z.; Tan, L.; et al. Newcastle Disease Virus V Protein Degrades Mitochondrial Antiviral Signaling Protein To Inhibit Host Type I Interferon Production via E3 Ubiquitin Ligase RNF5. J. Virol. 2019, 93. [Google Scholar] [CrossRef] [PubMed]
  67. Zhong, B.; Zhang, L.; Lei, C.Q.; Li, Y.; Mao, A.P.; Yang, Y.; Wang, Y.Y.; Zhang, X.L.; Shu, H.B. The Ubiquitin Ligase RNF5 Regulates Antiviral Responses by Mediating Degradation of the Adaptor Protein MITA. Immunity 2009, 30, 397–407. [Google Scholar] [CrossRef]
  68. Boso, G.; Tasaki, T.; Kwon, Y.T.; Somia, N.V. The N-end rule and retroviral infection: No effect on integrase. Virol. J. 2013, 10, 233. [Google Scholar] [CrossRef]
  69. Ojha, N.K.; Lole, K.S. Hepatitis E virus ORF1 encoded macro domain protein interacts with light chain subunit of human ferritin and inhibits its secretion. Mol. Cell. Biochem. 2016, 417, 75–85. [Google Scholar] [CrossRef]
  70. Hao, Z.; Zheng, L.; Kluwe, L.; Huang, W. Ferritin light chain and squamous cell carcinoma antigen 1 are coreceptors for cellular attachment and entry of hepatitis B virus. Int. J. Nanomed. 2012, 7, 827–834. [Google Scholar] [CrossRef]
  71. Du, J.; Liang, X.; Liu, Y.; Qu, Z.; Gao, L.; Han, L.; Liu, S.; Cui, M.; Shi, Y.; Zhang, Z.; et al. Hepatitis B virus core protein inhibits TRAIL-induced apoptosis of hepatocytes by blocking DR5 expression. Cell Death Differ. 2009, 16, 219–229. [Google Scholar] [CrossRef] [PubMed]
  72. Sola-Riera, C.; Gupta, S.; Maleki, K.T.; Gonzalez-Rodriguez, P.; Saidi, D.; Zimmer, C.L.; Vangeti, S.; Rivino, L.; Leo, Y.S.; Lye, D.C.; et al. Hantavirus Inhibits TRAIL-Mediated Killing of Infected Cells by Downregulating Death Receptor 5. Cell Rep. 2019, 28, 2124–2139.e6. [Google Scholar] [CrossRef]
  73. Shin, G.C.; Kang, H.S.; Lee, A.R.; Kim, K.H. Hepatitis B virus-triggered autophagy targets TNFRSF10B/death receptor 5 for degradation to limit TNFSF10/TRAIL response. Autophagy 2016, 12, 2451–2466. [Google Scholar] [CrossRef] [PubMed]
  74. Geiss, G.K.; Bumgarner, R.E.; An, M.C.; Agy, M.B.; van’t Wout, A.B.; Hammersmark, E.; Carter, V.S.; Upchurch, D.; Mullins, J.I.; Katze, M.G. Large-scale monitoring of host cell gene expression during HIV-1 infection using cDNA microarrays. Virology 2000, 266, 8–16. [Google Scholar] [CrossRef] [PubMed]
  75. Toraih, E.A.; Alrefai, H.G.; Hussein, M.H.; Helal, G.M.; Khashana, M.S.; Fawzy, M.S. Overexpression of heat shock protein HSP90AA1 and translocase of the outer mitochondrial membrane TOM34 in HCV-induced hepatocellular carcinoma: A pilot study. Clin. Biochem. 2019, 63, 10–17. [Google Scholar] [CrossRef] [PubMed]
  76. Wongtrakul, J.; Thongtan, T.; Pannengpetch, S.; Wikan, N.; Kantamala, D.; Kumrapich, B.; Suwan, W.; Smith, D.R. Phosphoproteomic analysis of dengue virus infected U937 cells and identification of pyruvate kinase M2 as a differentially phosphorylated phosphoprotein. Sci. Rep. 2020, 10, 14493. [Google Scholar] [CrossRef] [PubMed]
  77. Holder, K.A.; Burt, K.; Grant, M.D. TIGIT blockade enhances NK cell activity against autologous HIV-1-infected CD4(+) T cells. Clin. Transl. Immunol. 2021, 10, e1348. [Google Scholar] [CrossRef] [PubMed]
  78. Schorer, M.; Rakebrandt, N.; Lambert, K.; Hunziker, A.; Pallmer, K.; Oxenius, A.; Kipar, A.; Stertz, S.; Joller, N. TIGIT limits immune pathology during viral infections. Nat. Commun. 2020, 11, 1288. [Google Scholar] [CrossRef]
  79. Wang, J.; Hou, H.; Mao, L.; Wang, F.; Yu, J.; Luo, Y.; Lin, Q.; Sun, Z. TIGIT Signaling Pathway Regulates Natural Killer Cell Function in Chronic Hepatitis B Virus Infection. Front. Med. 2021, 8, 816474. [Google Scholar] [CrossRef]
  80. Alvarez, R.A.; Maestre, A.M.; Law, K.; Durham, N.D.; Barria, M.I.; Ishii-Watabe, A.; Tada, M.; Kapoor, M.; Hotta, M.T.; Rodriguez-Caprio, G.; et al. Enhanced FCGR2A and FCGR3A signaling by HIV viremic controller IgG. JCI Insight 2017, 2, e88226. [Google Scholar] [CrossRef]
  81. Alagarasu, K.; Bachal, R.V.; Damle, I.; Shah, P.S.; Cecilia, D. Association of FCGR2A p.R131H and CCL2 c.-2518 A>G gene variants with thrombocytopenia in patients with dengue virus infection. Hum. Immunol. 2015, 76, 819–822. [Google Scholar] [CrossRef]
  82. Martínez-Betancur, V.; Martínez-Gutierrez, M. Proteomic profile of human monocytic cells infected with dengue virus. Asian Pac. J. Trop. Bio. 2016, 6, 914–923. [Google Scholar] [CrossRef]
  83. Saad, Z.M.; Fouad, Y.; Ali, L.H.; Hassanin, T.M. Clinical Significance of Annexin A4 as a Biomarker in the Early Diagnosis of Hepatocellular Carcinoma. Asian Pac. J. Cancer Prev. APJCP 2020, 21, 2661–2665. [Google Scholar] [CrossRef] [PubMed]
  84. Munoz-Prieto, A.; Rubic, I.; Gonzalez-Sanchez, J.C.; Kules, J.; Martinez-Subiela, S.; Ceron, J.J.; Bernal, E.; Torres-Cantero, A.; Vicente-Romero, M.R.; Mrljak, V.; et al. Saliva changes in composition associated to COVID-19: A preliminary study. Sci. Rep. 2022, 12, 10879. [Google Scholar] [CrossRef] [PubMed]
  85. Yamamoto, Y.; Kakizaki, M.; Shimizu, T.; Carreras, J.; Chiba, T.; Chamoto, K.; Kagawa, T.; Aoki, T.; Nakamura, N.; Ando, K.; et al. PD-L1 is induced on the hepatocyte surface via CKLF-like MARVEL transmembrane domain-containing protein 6 up-regulation by the anti-HBV drug Entecavir. Int. Immunol. 2020, 32, 519–531. [Google Scholar] [CrossRef] [PubMed]
  86. Coombs, K.M.; Berard, A.; Xu, W.; Krokhin, O.; Meng, X.; Cortens, J.P.; Kobasa, D.; Wilkins, J.; Brown, E.G. Quantitative proteomic analyses of influenza virus-infected cultured human lung cells. J. Virol. 2010, 84, 10888–10906. [Google Scholar] [CrossRef] [PubMed]
  87. Zhang, Y.; Ma, W.; Mo, X.; Zhao, H.; Zheng, H.; Ke, C.; Zheng, W.; Tu, Y.; Zhang, Y. Erratum: Differential expressed genes in ECV304 Endothelial-like Cells infected with Human Cytomegalovirus. Afr. Health Sci. 2013, 13, 864–879. [Google Scholar] [CrossRef]
  88. Kang, S.D.; Chatterjee, S.; Alam, S.; Salzberg, A.C.; Milici, J.; van der Burg, S.H.; Meyers, C. Effect of Productive Human Papillomavirus 16 Infection on Global Gene Expression in Cervical Epithelium. J. Virol. 2018, 92. [Google Scholar] [CrossRef]
  89. Alfaro, E.; Diaz-Garcia, E.; Garcia-Tovar, S.; Zamarron, E.; Mangas, A.; Galera, R.; Lopez-Collazo, E.; Garcia-Rio, F.; Cubillos-Zapata, C. Upregulated Proteasome Subunits in COVID-19 Patients: A Link with Hypoxemia, Lymphopenia and Inflammation. Biomolecules 2022, 12, 442. [Google Scholar] [CrossRef]
  90. Wang, B.; Zhu, Y.; Yu, C.; Zhang, C.; Tang, Q.; Huang, H.; Zhao, Z. Hepatitis C virus induces oxidation and degradation of apolipoprotein B to enhance lipid accumulation and promote viral production. PLoS Pathog. 2021, 17, e1009889. [Google Scholar] [CrossRef]
  91. Dong, S.; Kong, N.; Wang, C.; Li, Y.; Sun, D.; Qin, W.; Zhai, H.; Zhai, X.; Yang, X.; Ye, C.; et al. FUBP3 Degrades the Porcine Epidemic Diarrhea Virus Nucleocapsid Protein and Induces the Production of Type I Interferon. J. Virol. 2022, 96, e0061822. [Google Scholar] [CrossRef] [PubMed]
  92. Tsai, F.J.; Lin, C.W.; Lai, C.C.; Lan, Y.C.; Lai, C.H.; Hung, C.H.; Hsueh, K.C.; Lin, T.H.; Chang, H.C.; Wan, L.; et al. Kaempferol inhibits enterovirus 71 replication and internal ribosome entry site (IRES) activity through FUBP and HNRP proteins. Food Chem. 2011, 128, 312–322. [Google Scholar] [CrossRef] [PubMed]
  93. Xu, P.; Tong, W.; Chen, Y.M. FUSE binding protein FUBP3 is a potent regulator in Japanese encephalitis virus infection. Virol. J. 2021, 18, 224. [Google Scholar] [CrossRef] [PubMed]
  94. Montespan, C.; Marvin, S.A.; Austin, S.; Burrage, A.M.; Roger, B.; Rayne, F.; Faure, M.; Campell, E.M.; Schneider, C.; Reimer, R.; et al. Multi-layered control of Galectin-8 mediated autophagy during adenovirus cell entry through a conserved PPxY motif in the viral capsid. PLoS Pathog. 2017, 13, e1006217. [Google Scholar] [CrossRef] [PubMed]
  95. Zhang, Y.; Hou, P.; He, D.C.; Wang, H.; He, H. RACK1 degrades MAVS to promote bovine ephemeral fever virus replication via upregulating E3 ubiquitin ligase STUB1. Vet. Microbiol. 2021, 257, 109096. [Google Scholar] [CrossRef] [PubMed]
  96. Zhao, K.; Zhang, Q.; Li, X.; Zhao, D.; Liu, Y.; Shen, Q.; Yang, M.; Wang, C.; Li, N.; Cao, X. Cytoplasmic STAT4 Promotes Antiviral Type I IFN Production by Blocking CHIP-Mediated Degradation of RIG-I. J. Immunol. 2016, 196, 1209–1217. [Google Scholar] [CrossRef] [PubMed]
  97. Wang, Y.; Du, S.; Zhu, C.; Wang, C.; Yu, N.; Lin, Z.; Gan, J.; Guo, Y.; Huang, X.; He, Y.; et al. STUB1 is targeted by the SUMO-interacting motif of EBNA1 to maintain Epstein-Barr Virus latency. PLoS Pathog. 2020, 16, e1008447. [Google Scholar] [CrossRef]
  98. Pericle, F.; Pinto, L.A.; Hicks, S.; Kirken, R.A.; Sconocchia, G.; Rusnak, J.; Dolan, M.J.; Shearer, G.M.; Segal, D.M. HIV-1 infection induces a selective reduction in STAT5 protein expression. J. Immunol. 1998, 160, 28–31. [Google Scholar] [CrossRef]
  99. Zhang, Y.; Li, B.X.; Mao, Q.Z.; Zhuo, J.C.; Huang, H.J.; Lu, J.B.; Zhang, C.X.; Li, J.M.; Chen, J.P.; Lu, G. The JAK-STAT pathway promotes persistent viral infection by activating apoptosis in insect vectors. PLoS Pathog. 2023, 19, e1011266. [Google Scholar] [CrossRef]
  100. Aydemir, M.N.; Aydemir, H.B.; Korkmaz, E.M.; Budak, M.; Cekin, N.; Pinarbasi, E. Computationally predicted SARS-CoV-2 encoded microRNAs target NFKB, JAK/STAT and TGFB signaling pathways. Gene Rep. 2021, 22, 101012. [Google Scholar] [CrossRef]
  101. Prost, S.; Le Dantec, M.; Auge, S.; Le Grand, R.; Derdouch, S.; Auregan, G.; Deglon, N.; Relouzat, F.; Aubertin, A.M.; Maillere, B.; et al. Human and simian immunodeficiency viruses deregulate early hematopoiesis through a Nef/PPARgamma/STAT5 signaling pathway in macaques. J. Clin. Investig. 2008, 118, 1765–1775. [Google Scholar] [CrossRef]
  102. Diaz-Salazar, C.; Bou-Puerto, R.; Mujal, A.M.; Lau, C.M.; von Hoesslin, M.; Zehn, D.; Sun, J.C. Cell-intrinsic adrenergic signaling controls the adaptive NK cell response to viral infection. J. Exp. Med. 2020, 217, e20190549. [Google Scholar] [CrossRef]
  103. Wieduwild, E.; Girard-Madoux, M.J.; Quatrini, L.; Laprie, C.; Chasson, L.; Rossignol, R.; Bernat, C.; Guia, S.; Ugolini, S. beta2-adrenergic signals downregulate the innate immune response and reduce host resistance to viral infection. J. Exp. Med. 2020, 217, e20190554. [Google Scholar] [CrossRef]
  104. Chaipan, C.; Steffen, I.; Tsegaye, T.S.; Bertram, S.; Glowacka, I.; Kato, Y.; Schmokel, J.; Munch, J.; Simmons, G.; Gerardy-Schahn, R.; et al. Incorporation of podoplanin into HIV released from HEK-293T cells, but not PBMC, is required for efficient binding to the attachment factor CLEC-2. Retrovirology 2010, 7, 47. [Google Scholar] [CrossRef]
  105. Amsden, H.; Kourko, O.; Roth, M.; Gee, K. Antiviral Activities of Interleukin-27: A Partner for Interferons? Front. Immunol. 2022, 13, 902853. [Google Scholar] [CrossRef]
  106. Harker, J.A.; Wong, K.A.; Dallari, S.; Bao, P.; Dolgoter, A.; Jo, Y.; Wehrens, E.J.; Macal, M.; Zuniga, E.I. Interleukin-27R Signaling Mediates Early Viral Containment and Impacts Innate and Adaptive Immunity after Chronic Lymphocytic Choriomeningitis Virus Infection. J. Virol. 2018, 92. [Google Scholar] [CrossRef]
  107. Bertram, E.M.; Tafuri, A.; Shahinian, A.; Chan, V.S.; Hunziker, L.; Recher, M.; Ohashi, P.S.; Mak, T.W.; Watts, T.H. Role of ICOS versus CD28 in antiviral immunity. Eur. J. Immunol. 2002, 32, 3376–3385. [Google Scholar] [CrossRef]
  108. Yeh, M.M.; Boukhar, S.; Roberts, B.; Dasgupta, N.; Daoud, S.S. Genomic variants link to hepatitis C racial disparities. Oncotarget 2017, 8, 59455–59475. [Google Scholar] [CrossRef] [PubMed]
  109. Liu, H.; Wang, J.; Zhang, Y.; Gu, J.; Wang, Y.; Yan, Y.; Pan, D.; Sun, Z. Cerebrospinal fluid proteomics in meningitis patients with reactivated varicella zoster virus. Immun. Inflamm. Dis. 2023, 11, e1038. [Google Scholar] [CrossRef] [PubMed]
  110. Yao, X.; Wang, X.Y.; Zhao, C.; Sun, S.H.; Meng, Z.F.; Zhang, J.M.; Xu, J.Q.; Xie, Y.H.; Yuan, Z.H.; Wen, Y.M. Transcriptional analysis of immune-related genes in dendritic cells from hepatitis B surface antigen (HBsAg)-positive transgenic mice and regulation of Fc gamma receptor IIB by HBsAg-anti-HBs complex. J. Med. Virol. 2011, 83, 78–87. [Google Scholar] [CrossRef] [PubMed]
  111. Haslbauer, J.D.; Savic Prince, S.; Stalder, A.K.; Matter, M.S.; Zinner, C.P.; Jahn, K.; Obermann, E.; Hanke, J.; Leuzinger, K.; Hirsch, H.H.; et al. Differential Gene Expression of SARS-CoV-2 positive Bronchoalveolar Lavages: A Case Series. Pathobiol. J. Immunopathol. Mol. Cell. Biol. 2023, 10, 000532057. [Google Scholar] [CrossRef] [PubMed]
  112. Smith, A.J.; Wietgrefe, S.W.; Shang, L.; Reilly, C.S.; Southern, P.J.; Perkey, K.E.; Duan, L.; Kohler, H.; Muller, S.; Robinson, J.; et al. Live simian immunodeficiency virus vaccine correlate of protection: Immune complex-inhibitory Fc receptor interactions that reduce target cell availability. J. Immunol. 2014, 193, 3126–3133. [Google Scholar] [CrossRef] [PubMed]
  113. Weisberger, J.; Cornfield, D.; Gorczyca, W.; Liu, Z. Down-regulation of pan-T-cell antigens, particularly CD7, in acute infectious mononucleosis. Am. J. Clin. Pathol. 2003, 120, 49–55. [Google Scholar] [CrossRef] [PubMed]
  114. Sato, A.I.; Balamuth, F.B.; Ugen, K.E.; Williams, W.V.; Weiner, D.B. Identification of CD7 glycoprotein as an accessory molecule in HIV-1-mediated syncytium formation and cellfree infection. J. Immunol. 1994, 152, 5142–5152. [Google Scholar] [CrossRef] [PubMed]
  115. Akl, H.; Badran, B.M.; Zein, N.E.; Bex, F.; Sotiriou, C.; Willard-Gallo, K.E.; Burny, A.; Martiat, P. HTLV-I infection of WE17/10 CD4+ cell line leads to progressive alteration of Ca2+ influx that eventually results in loss of CD7 expression and activation of an antiapoptotic pathway involving AKT and BAD which paves the way for malignant transformation. Leukemia 2007, 21, 788–796. [Google Scholar] [CrossRef] [PubMed]
  116. Han, D.; Tai, Y.; Hua, G.; Yang, X.; Chen, J.; Li, J.; Deng, X. Melanocytes in black-boned chicken have immune contribution under infectious bursal disease virus infection. Poult. Sci. 2021, 100, 101498. [Google Scholar] [CrossRef]
  117. Lu, Y.; Ye, Z.; Liu, X.; Zhou, L.; Ding, X.; Hou, Y. Role of SARS-CoV-2 nucleocapsid protein in affecting immune cells and insights on its molecular mechanisms. Exp. Ther. Med. 2023, 26, 504. [Google Scholar] [CrossRef] [PubMed]
  118. Zhang, C.; Li, W.; Lei, X.; Xie, Z.; Qi, L.; Wang, H.; Xiao, X.; Xiao, J.; Zheng, Y.; Dong, C.; et al. Targeting lysophospholipid acid receptor 1 and ROCK kinases promotes antiviral innate immunity. Sci. Adv. 2021, 7, eabb5933. [Google Scholar] [CrossRef]
  119. Krishnan, M.N.; Ng, A.; Sukumaran, B.; Gilfoy, F.D.; Uchil, P.D.; Sultana, H.; Brass, A.L.; Adametz, R.; Tsui, M.; Qian, F.; et al. RNA interference screen for human genes associated with West Nile virus infection. Nature 2008, 455, 242–245. [Google Scholar] [CrossRef]
  120. Li, C.W.; Jheng, B.R.; Chen, B.S. Investigating genetic-and-epigenetic networks, and the cellular mechanisms occurring in Epstein-Barr virus-infected human B lymphocytes via big data mining and genome-wide two-sided NGS data identification. PLoS ONE 2018, 13, e0202537. [Google Scholar] [CrossRef]
  121. Haberl, E.M.; Feder, S.; Pohl, R.; Rein-Fischboeck, L.; Durholz, K.; Eichelberger, L.; Wanninger, J.; Weiss, T.S.; Buechler, C. Chemerin Is Induced in Non-Alcoholic Fatty Liver Disease and Hepatitis B-Related Hepatocellular Carcinoma. Cancers 2020, 12, 2967. [Google Scholar] [CrossRef] [PubMed]
  122. Samson, M.; Edinger, A.L.; Stordeur, P.; Rucker, J.; Verhasselt, V.; Sharron, M.; Govaerts, C.; Mollereau, C.; Vassart, G.; Doms, R.W.; et al. ChemR23, a putative chemoattractant receptor, is expressed in monocyte-derived dendritic cells and macrophages and is a coreceptor for SIV and some primary HIV-1 strains. Eur. J. Immunol. 1998, 28, 1689–1700. [Google Scholar] [CrossRef]
  123. Cayabyab, M.; Hinuma, S.; Farzan, M.; Choe, H.; Fukusumi, S.; Kitada, C.; Nishizawa, N.; Hosoya, M.; Nishimura, O.; Messele, T.; et al. Apelin, the natural ligand of the orphan seven-transmembrane receptor APJ, inhibits human immunodeficiency virus type 1 entry. J. Virol. 2000, 74, 11972–11976. [Google Scholar] [CrossRef] [PubMed]
  124. Nong, W.; Ma, L.; Lan, B.; Liu, N.; Yang, H.; Lao, X.; Deng, Q.; Huang, Z. Comprehensive Identification of Bridge Genes to Explain the Progression from Chronic Hepatitis B Virus Infection to Hepatocellular Carcinoma. J. Inflamm. Res. 2021, 14, 1613–1624. [Google Scholar] [CrossRef] [PubMed]
  125. Lehmann, J.; Muresan, S.; Weber, S.N.; Lammert, F.; Krawczyk, M. Acute Pancreatitis in the Setting of Hepatitis E Virus (Genotype 3) Infection and Compound CLDN2-PRSS1 Risk Variants. Pancreas 2020, 49, e91–e93. [Google Scholar] [CrossRef] [PubMed]
  126. Delva, J.L.; Daled, S.; Van Waesberghe, C.; Almey, R.; Jansens, R.J.J.; Deforce, D.; Dhaenens, M.; Favoreel, H.W. Proteomic Comparison of Three Wild-Type Pseudorabies Virus Strains and the Attenuated Bartha Strain Reveals Reduced Incorporation of Several Tegument Proteins in Bartha Virions. J. Virol. 2022, 96, e0115822. [Google Scholar] [CrossRef]
  127. Tsai, C.H.; Wu, A.C.; Chiang, B.L.; Yang, Y.H.; Hung, S.P.; Su, M.W.; Chang, Y.J.; Lee, Y.L. CEACAM3 decreases asthma exacerbations and modulates respiratory syncytial virus latent infection in children. Thorax 2020, 75, 725–734. [Google Scholar] [CrossRef]
  128. de Almeida Chuffa, L.G.; Freire, P.P.; Dos Santos Souza, J.; de Mello, M.C.; de Oliveira Neto, M.; Carvalho, R.F. Aging whole blood transcriptome reveals candidate genes for SARS-CoV-2-related vascular and immune alterations. J. Mol. Med. 2022, 100, 285–301. [Google Scholar] [CrossRef]
  129. Wu, S.; Yang, S.; Ou, M.; Chen, J.; Huang, J.; Xiong, D.; Sun, W.; Xiao, L. Transcriptome Analysis Reveals the Role of Cellular Calcium Disorder in Varicella Zoster Virus-Induced Post-Herpetic Neuralgia. Front. Mol. Neurosci. 2021, 14, 665931. [Google Scholar] [CrossRef]
  130. Isken, O.; Postel, A.; Bruhn, B.; Lattwein, E.; Becher, P.; Tautz, N. CRISPR/Cas9-Mediated Knockout of DNAJC14 Verifies This Chaperone as a Pivotal Host Factor for RNA Replication of Pestiviruses. J. Virol. 2019, 93. [Google Scholar] [CrossRef]
  131. Yi, Z.; Sperzel, L.; Nurnberger, C.; Bredenbeek, P.J.; Lubick, K.J.; Best, S.M.; Stoyanov, C.T.; Law, L.M.; Yuan, Z.; Rice, C.M.; et al. Identification and characterization of the host protein DNAJC14 as a broadly active flavivirus replication modulator. PLoS Pathog. 2011, 7, e1001255. [Google Scholar] [CrossRef] [PubMed]
  132. Goncalves, R.L.; de Souza, G.A.P.; de Souza Terceti, M.; de Castro, R.F.G.; de Mello Silva, B.; Novaes, R.D.; Malaquias, L.C.C.; Coelho, L.F.L. Integrative transcriptome analysis of SARS-CoV-2 human-infected cells combined with deep learning algorithms identifies two potential cellular targets for the treatment of coronavirus disease. Braz. J. Microbiol. Publ. Braz. Soc. Microbiol. 2023, 54, 53–68. [Google Scholar] [CrossRef] [PubMed]
  133. Tripathi, L.P.; Kambara, H.; Chen, Y.A.; Nishimura, Y.; Moriishi, K.; Okamoto, T.; Morita, E.; Abe, T.; Mori, Y.; Matsuura, Y.; et al. Understanding the biological context of NS5A-host interactions in HCV infection: A network-based approach. J. Proteome Res. 2013, 12, 2537–2551. [Google Scholar] [CrossRef] [PubMed]
  134. Stoeck, I.K.; Lee, J.Y.; Tabata, K.; Romero-Brey, I.; Paul, D.; Schult, P.; Lohmann, V.; Kaderali, L.; Bartenschlager, R. Hepatitis C Virus Replication Depends on Endosomal Cholesterol Homeostasis. J. Virol. 2018, 92. [Google Scholar] [CrossRef] [PubMed]
  135. Paust, S.; Gill, H.S.; Wang, B.Z.; Flynn, M.P.; Moseman, E.A.; Senman, B.; Szczepanik, M.; Telenti, A.; Askenase, P.W.; Compans, R.W.; et al. Critical role for the chemokine receptor CXCR6 in NK cell-mediated antigen-specific memory of haptens and viruses. Nat. Immunol. 2010, 11, 1127–1135. [Google Scholar] [CrossRef]
  136. Liao, F.; Alkhatib, G.; Peden, K.W.; Sharma, G.; Berger, E.A.; Farber, J.M. STRL33, A novel chemokine receptor-like protein, functions as a fusion cofactor for both macrophage-tropic and T cell line-tropic HIV-1. J. Exp. Med. 1997, 185, 2015–2023. [Google Scholar] [CrossRef]
  137. Ashhurst, A.S.; Florido, M.; Lin, L.C.W.; Quan, D.; Armitage, E.; Stifter, S.A.; Stambas, J.; Britton, W.J. CXCR6-Deficiency Improves the Control of Pulmonary Mycobacterium tuberculosis and Influenza Infection Independent of T-Lymphocyte Recruitment to the Lungs. Front. Immunol. 2019, 10, 339. [Google Scholar] [CrossRef]
  138. Pirozyan, M.R.; Nguyen, N.; Cameron, B.; Luciani, F.; Bull, R.A.; Zekry, A.; Lloyd, A.R. Chemokine-Regulated Recruitment of Antigen-Specific T-Cell Subpopulations to the Liver in Acute and Chronic Hepatitis C Infection. J. Infect. Dis. 2019, 219, 1430–1438. [Google Scholar] [CrossRef]
  139. Teran, L.M.; Ruggeberg, S.; Santiago, J.; Fuentes-Arenas, F.; Hernandez, J.L.; Montes-Vizuet, A.R.; Xinping, L.; Franz, T. Immune response to seasonal influenza A virus infection: A proteomic approach. Arch. Med. Res. 2012, 43, 464–469. [Google Scholar] [CrossRef]
  140. Li, S.Y.; Zhang, Z.N.; Jiang, Y.J.; Fu, Y.J.; Shang, H. Transcriptional insights into the CD8(+) T cell response in mono-HIV and HCV infection. J. Transl. Med. 2020, 18, 96. [Google Scholar] [CrossRef]
  141. Santana, B.B.; Queiroz, M.A.F.; Cerveira, R.A.; Rodrigues, C.M.; da Silva Graca Amoras, E.; da Costa, C.A.; de Sousa, M.S.; Ishak, R.; Goulart, L.R.; Vallinoto, A.C.R. Low Annexin A1 level in HTLV-1 infected patients is a potential biomarker for the clinical progression and diagnosis of HAM/TSP. BMC Infect. Dis. 2021, 21, 219. [Google Scholar] [CrossRef] [PubMed]
  142. Shaath, H.; Vishnubalaji, R.; Elkord, E.; Alajez, N.M. Single-Cell Transcriptome Analysis Highlights a Role for Neutrophils and Inflammatory Macrophages in the Pathogenesis of Severe COVID-19. Cells 2020, 9, 2374. [Google Scholar] [CrossRef] [PubMed]
  143. Duhalde Vega, M.; Olivera, D.; Gastao Davanzo, G.; Bertullo, M.; Noya, V.; Fabiano de Souza, G.; Primon Muraro, S.; Castro, I.; Arevalo, A.P.; Crispo, M.; et al. PD-1/PD-L1 blockade abrogates a dysfunctional innate-adaptive immune axis in critical beta-coronavirus disease. Sci. Adv. 2022, 8, eabn6545. [Google Scholar] [CrossRef] [PubMed]
  144. Zhong, B.; Zhang, Y.; Tan, B.; Liu, T.T.; Wang, Y.Y.; Shu, H.B. The E3 ubiquitin ligase RNF5 targets virus-induced signaling adaptor for ubiquitination and degradation. J. Immunol. 2010, 184, 6249–6255. [Google Scholar] [CrossRef] [PubMed]
  145. Kong, Z.; Yin, H.; Wang, F.; Liu, Z.; Luan, X.; Sun, L.; Liu, W.; Shang, Y. Pseudorabies virus tegument protein UL13 recruits RNF5 to inhibit STING-mediated antiviral immunity. PLoS Pathog. 2022, 18, e1010544. [Google Scholar] [CrossRef]
  146. Chen, Z.L.; Yin, Z.J.; Qiu, T.Y.; Chen, J.; Liu, J.; Zhang, X.Y.; Xu, J.Q. Revealing the characteristics of ZIKV infection through tissue-specific transcriptome sequencing analysis. BMC Genom. 2022, 23, 697. [Google Scholar] [CrossRef]
  147. Chakraborty, S.; Sen, E.; Basu, A. Pyruvate dehydrogenase kinase 1 promotes neuronal apoptosis upon Japanese encephalitis virus infection. IBRO Neurosci. Rep. 2022, 13, 410–419. [Google Scholar] [CrossRef]
  148. Jung, G.S.; Jeon, J.H.; Choi, Y.K.; Jang, S.Y.; Park, S.Y.; Kim, S.W.; Byun, J.K.; Kim, M.K.; Lee, S.; Shin, E.C.; et al. Pyruvate dehydrogenase kinase regulates hepatitis C virus replication. Sci. Rep. 2016, 6, 30846. [Google Scholar] [CrossRef]
  149. Dawood, R.M.; El-Meguid, M.A.; Ibrahim, M.K.; Bader El Din, N.G.; Barakat, A.; El-Wakeel, K.; Alla, M.; Wu, G.Y.; El Awady, M.K. Dysregulation of fibrosis related genes in HCV induced liver disease. Gene 2018, 664, 58–69. [Google Scholar] [CrossRef]
  150. Schmidt, N.; Lareau, C.A.; Keshishian, H.; Ganskih, S.; Schneider, C.; Hennig, T.; Melanson, R.; Werner, S.; Wei, Y.; Zimmer, M.; et al. The SARS-CoV-2 RNA-protein interactome in infected human cells. Nat. Microbiol. 2021, 6, 339–353. [Google Scholar] [CrossRef]
  151. Villalba, M.; Fredericksen, F.; Otth, C.; Olavarria, V. Transcriptomic analysis of responses to cytopathic bovine viral diarrhea virus-1 (BVDV-1) infection in MDBK cells. Mol. Immunol. 2016, 71, 192–202. [Google Scholar] [CrossRef] [PubMed]
  152. Merle, P.; de la Monte, S.; Kim, M.; Herrmann, M.; Tanaka, S.; Von Dem Bussche, A.; Kew, M.C.; Trepo, C.; Wands, J.R. Functional consequences of frizzled-7 receptor overexpression in human hepatocellular carcinoma. Gastroenterology 2004, 127, 1110–1122. [Google Scholar] [CrossRef] [PubMed]
  153. Ikeda, F.; Deribe, Y.L.; Skanland, S.S.; Stieglitz, B.; Grabbe, C.; Franz-Wachtel, M.; van Wijk, S.J.; Goswami, P.; Nagy, V.; Terzic, J.; et al. SHARPIN forms a linear ubiquitin ligase complex regulating NF-kappaB activity and apoptosis. Nature 2011, 471, 637–641. [Google Scholar] [CrossRef] [PubMed]
  154. Jing, H.; Fang, L.; Ding, Z.; Wang, D.; Hao, W.; Gao, L.; Ke, W.; Chen, H.; Xiao, S. Porcine Reproductive and Respiratory Syndrome Virus nsp1alpha Inhibits NF-kappaB Activation by Targeting the Linear Ubiquitin Chain Assembly Complex. J. Virol. 2017, 91. [Google Scholar] [CrossRef] [PubMed]
  155. Schoggins, J.W.; Wilson, S.J.; Panis, M.; Murphy, M.Y.; Jones, C.T.; Bieniasz, P.; Rice, C.M. A diverse range of gene products are effectors of the type I interferon antiviral response. Nature 2011, 472, 481–485. [Google Scholar] [CrossRef] [PubMed]
  156. McLaren, P.J.; Gawanbacht, A.; Pyndiah, N.; Krapp, C.; Hotter, D.; Kluge, S.F.; Gotz, N.; Heilmann, J.; Mack, K.; Sauter, D.; et al. Identification of potential HIV restriction factors by combining evolutionary genomic signatures with functional analyses. Retrovirology 2015, 12, 41. [Google Scholar] [CrossRef] [PubMed]
  157. Kotelkin, A.; Prikhod’ko, E.A.; Cohen, J.I.; Collins, P.L.; Bukreyev, A. Respiratory syncytial virus infection sensitizes cells to apoptosis mediated by tumor necrosis factor-related apoptosis-inducing ligand. J. Virol. 2003, 77, 9156–9172. [Google Scholar] [CrossRef]
  158. Ebrahimpour Gorji, A.; Roudbari, Z.; Ebrahimpour Gorji, F.; Sadeghi, B. Computational study of zebrafish immune-targeted microarray data for prediction of preventive drug candidates. Vet. Res. Forum Int. Q. J. 2021, 12, 87–93. [Google Scholar] [CrossRef]
  159. Wang, S.; Ding, X.; Li, Z.; Rao, F.; Xu, H.; Lu, J.; Ma, X.; Zhang, M.; Xie, Z. Comprehensive analyses identify potential biomarkers for encephalitis in HIV infection. Sci. Rep. 2023, 13, 18418. [Google Scholar] [CrossRef]
  160. Drozdzik, M.; Lapczuk-Romanska, J.; Wenzel, C.; Skalski, L.; Szelag-Pieniek, S.; Post, M.; Syczewska, M.; Kurzawski, M.; Oswald, S. Protein Abundance of Drug Transporters in Human Hepatitis C Livers. Int. J. Mol. Sci. 2022, 23, 7947. [Google Scholar] [CrossRef]
  161. Airo, A.M.; Felix-Lopez, A.; Mancinelli, V.; Evseev, D.; Lopez-Orozco, J.; Shire, K.; Paszkowski, P.; Frappier, L.; Magor, K.E.; Hobman, T.C. Flavivirus Capsid Proteins Inhibit the Interferon Response. Viruses 2022, 14, 968. [Google Scholar] [CrossRef] [PubMed]
  162. Ammour, Y.; Susova, O.; Krasnov, G.; Nikolaeva, E.; Varachev, V.; Schetinina, Y.; Gavrilova, M.; Mitrofanov, A.; Poletaeva, A.; Bekyashev, A.; et al. Transcriptome Analysis of Human Glioblastoma Cells Susceptible to Infection with the Leningrad-16 Vaccine Strain of Measles Virus. Viruses 2022, 14, 2433. [Google Scholar] [CrossRef] [PubMed]
  163. Molinero, M.; Gomez, S.; Benitez, I.D.; Vengoechea, J.J.; Gonzalez, J.; Polanco, D.; Gort-Paniello, C.; Moncusi-Moix, A.; Garcia-Hidalgo, M.C.; Perez-Pons, M.; et al. Multiplex protein profiling of bronchial aspirates reveals disease-, mortality- and respiratory sequelae-associated signatures in critically ill patients with ARDS secondary to SARS-CoV-2 infection. Front. Immunol. 2022, 13, 942443. [Google Scholar] [CrossRef] [PubMed]
  164. Quach, H.Q.; Goergen, K.M.; Grill, D.E.; Haralambieva, I.H.; Ovsyannikova, I.G.; Poland, G.A.; Kennedy, R.B. Virus-specific and shared gene expression signatures in immune cells after vaccination in response to influenza and vaccinia stimulation. Front. Immunol. 2023, 14, 1168784. [Google Scholar] [CrossRef] [PubMed]
  165. Vrazas, V.; Moustafa, S.; Makridakis, M.; Karakasiliotis, I.; Vlahou, A.; Mavromara, P.; Katsani, K.R. A Proteomic Approach to Study the Biological Role of Hepatitis C Virus Protein Core+1/ARFP. Viruses 2022, 14, 1694. [Google Scholar] [CrossRef] [PubMed]
  166. Johnston, G.P.; Bradel-Tretheway, B.; Piehowski, P.D.; Brewer, H.M.; Lee, B.N.R.; Usher, N.T.; Zamora, J.L.R.; Ortega, V.; Contreras, E.M.; Teuton, J.R.; et al. Nipah Virus-Like Particle Egress Is Modulated by Cytoskeletal and Vesicular Trafficking Pathways: A Validated Particle Proteomics Analysis. mSystems 2019, 4, e00194-19. [Google Scholar] [CrossRef] [PubMed]
  167. Xiong, F.; Cao, L.; Wu, X.M.; Chang, M.X. The function of zebrafish gpbar1 in antiviral response and lipid metabolism. Dev. Comp. Immunol. 2021, 116, 103955. [Google Scholar] [CrossRef]
  168. Smith, T.; Rohaim, M.A.; Munir, M. Mapping molecular gene signatures mediated by SARS-COV-2 and large-scale and genome-wide transcriptomics comparative analysis among respiratory viruses of medical importance. Mol. Cell. Probes 2022, 64, 101820. [Google Scholar] [CrossRef]
  169. Xiong, Q.; Huang, H.; Wang, N.; Chen, R.; Chen, N.; Han, H.; Wang, Q.; Siwko, S.; Liu, M.; Qian, M.; et al. Metabolite-Sensing G Protein Coupled Receptor TGR5 Protects Host From Viral Infection Through Amplifying Type I Interferon Responses. Front. Immunol. 2018, 9, 2289. [Google Scholar] [CrossRef]
  170. Lu, J.; Wang, H.; Zhang, Y.; Li, Y.; Lu, L. Grass carp reovirus NS26 interacts with cellular lipopolysaccharide-induced tumor necrosis factor-alpha factor, LITAF. Virus Genes 2016, 52, 789–796. [Google Scholar] [CrossRef]
  171. Eaton, H.E.; Ferreira Lacerda, A.; Desrochers, G.; Metcalf, J.; Angers, A.; Brunetti, C.R. Cellular LITAF interacts with frog virus 3 75L protein and alters its subcellular localization. J. Virol. 2013, 87, 716–723. [Google Scholar] [CrossRef] [PubMed]
  172. Kuchipudi, S.V.; Tellabati, M.; Sebastian, S.; Londt, B.Z.; Jansen, C.; Vervelde, L.; Brookes, S.M.; Brown, I.H.; Dunham, S.P.; Chang, K.C. Highly pathogenic avian influenza virus infection in chickens but not ducks is associated with elevated host immune and pro-inflammatory responses. Vet. Res. 2014, 45, 118. [Google Scholar] [CrossRef] [PubMed]
  173. Gu, J.G.; Zhu, C.L.; Cheng, D.Z.; Xie, Y.; Liu, F.; Zhou, X. Enchanced levels of apolipoprotein M during HBV infection feedback suppresses HBV replication. Lipids Health Dis. 2011, 10, 154. [Google Scholar] [CrossRef] [PubMed]
  174. Cai, H.; Yao, W.; Huang, J.; Xiao, J.; Chen, W.; Hu, L.; Mai, R.; Liang, M.; Chen, D.; Jiang, N.; et al. Apolipoprotein M, identified as a novel hepatitis C virus (HCV) particle associated protein, contributes to HCV assembly and interacts with E2 protein. Antivir. Res. 2020, 177, 104756. [Google Scholar] [CrossRef]
  175. Gonzalez-Dunia, D.; Watanabe, M.; Syan, S.; Mallory, M.; Masliah, E.; De La Torre, J.C. Synaptic pathology in Borna disease virus persistent infection. J. Virol. 2000, 74, 3441–3448. [Google Scholar] [CrossRef] [PubMed]
  176. Afkhami-Goli, A.; Liu, S.H.; Zhu, Y.; Antony, J.M.; Arab, H.; Power, C. Dual lentivirus infection potentiates neuroinflammation and neurodegeneration: Viral copassage enhances neurovirulence. J. Neurovirology 2009, 15, 139–152. [Google Scholar] [CrossRef] [PubMed]
  177. Piacentini, R.; Li Puma, D.D.; Ripoli, C.; Marcocci, M.E.; De Chiara, G.; Garaci, E.; Palamara, A.T.; Grassi, C. Herpes Simplex Virus type-1 infection induces synaptic dysfunction in cultured cortical neurons via GSK-3 activation and intraneuronal amyloid-beta protein accumulation. Sci. Rep. 2015, 5, 15444. [Google Scholar] [CrossRef] [PubMed]
  178. Fujimura, K.; Guise, A.J.; Nakayama, T.; Schlaffner, C.N.; Meziani, A.; Kumar, M.; Cheng, L.; Vaughan, D.J.; Kodani, A.; Van Haren, S.; et al. Integrative systems biology characterizes immune-mediated neurodevelopmental changes in murine Zika virus microcephaly. iScience 2023, 26, 106909. [Google Scholar] [CrossRef]
  179. Ma-Lauer, Y.; Carbajo-Lozoya, J.; Hein, M.Y.; Muller, M.A.; Deng, W.; Lei, J.; Meyer, B.; Kusov, Y.; von Brunn, B.; Bairad, D.R.; et al. p53 down-regulates SARS coronavirus replication and is targeted by the SARS-unique domain and PLpro via E3 ubiquitin ligase RCHY1. Proc. Natl. Acad. Sci. USA 2016, 113, E5192–E5201. [Google Scholar] [CrossRef]
  180. Chen, H.; Gao, X.; Zhao, S.; Bao, C.; Ming, X.; Qian, Y.; Zhou, Y.; Jung, Y.S. Pirh2 restricts influenza A virus replication by modulating short-chain ubiquitination of its nucleoprotein. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 2022, 36, e22537. [Google Scholar] [CrossRef]
  181. Opriessnig, T.; Karuppannan, A.K.; Halbur, P.G.; Calvert, J.G.; Nitzel, G.P.; Matzinger, S.R.; Meng, X.J. Porcine circovirus type 2a or 2b based experimental vaccines provide protection against PCV2d/porcine parvovirus 2 co-challenge. Vaccine 2020, 38, 1975–1981. [Google Scholar] [CrossRef] [PubMed]
  182. Chen, M.; Cortay, J.C.; Logan, I.R.; Sapountzi, V.; Robson, C.N.; Gerlier, D. Inhibition of ubiquitination and stabilization of human ubiquitin E3 ligase PIRH2 by measles virus phosphoprotein. J. Virol. 2005, 79, 11824–11836. [Google Scholar] [CrossRef] [PubMed]
  183. Lee, H.M.; Lo, K.W.; Wei, W.; Tsao, S.W.; Chung, G.T.Y.; Ibrahim, M.H.; Dawson, C.W.; Murray, P.G.; Paterson, I.C.; Yap, L.F. Oncogenic S1P signalling in EBV-associated nasopharyngeal carcinoma activates AKT and promotes cell migration through S1P receptor 3. J. Pathol. 2017, 242, 62–72. [Google Scholar] [CrossRef] [PubMed]
  184. Lau, B.; Poole, E.; Krishna, B.; Sellart, I.; Wills, M.R.; Murphy, E.; Sinclair, J. The Expression of Human Cytomegalovirus MicroRNA MiR-UL148D during Latent Infection in Primary Myeloid Cells Inhibits Activin A-triggered Secretion of IL-6. Sci. Rep. 2016, 6, 31205. [Google Scholar] [CrossRef] [PubMed]
  185. Pryce, R.; Azarm, K.; Rissanen, I.; Harlos, K.; Bowden, T.A.; Lee, B. A key region of molecular specificity orchestrates unique ephrin-B1 utilization by Cedar virus. Life Sci. Alliance 2020, 3, e201900578. [Google Scholar] [CrossRef] [PubMed]
  186. Cao, J.; Liu, S.; Liu, M.; Wang, S.; Bi, Z.; Fan, W.; Shi, Z.; Song, S.; Yan, L. Hsp70 Inhibits the Replication of Fowl Adenovirus Serotype 4 by Suppressing Viral Hexon with the Assistance of DnaJC7. J. Virol. 2022, 96, e0080722. [Google Scholar] [CrossRef]
  187. Lv, Q.; Wang, T.; Liu, S.; Zhu, Y. Porcine circovirus type 2 exploits cap to inhibit PKR activation through interaction with Hsp40. Vet. Microbiol. 2021, 252, 108929. [Google Scholar] [CrossRef]
  188. He, J.; Zheng, Y.W.; Lin, Y.F.; Mi, S.; Qin, X.W.; Weng, S.P.; He, J.G.; Guo, C.J. Caveolae Restrict Tiger Frog Virus Release in HepG2 cells and Caveolae-Associated Proteins Incorporated into Virus Particles. Sci. Rep. 2016, 6, 21663. [Google Scholar] [CrossRef]
  189. Kipper, S.; Hamad, S.; Caly, L.; Avrahami, D.; Bacharach, E.; Jans, D.A.; Gerber, D.; Bajorek, M. New host factors important for respiratory syncytial virus (RSV) replication revealed by a novel microfluidics screen for interactors of matrix (M) protein. Mol. Cell. Proteom. MCP 2015, 14, 532–543. [Google Scholar] [CrossRef]
  190. Kim, J.Y.; Wang, L.; Lee, J.; Ou, J.J. Hepatitis C Virus Induces the Localization of Lipid Rafts to Autophagosomes for Its RNA Replication. J. Virol. 2017, 91. [Google Scholar] [CrossRef]
  191. Waris, G.; Huh, K.W.; Siddiqui, A. Mitochondrially associated hepatitis B virus X protein constitutively activates transcription factors STAT-3 and NF-kappa B via oxidative stress. Mol. Cell. Biol. 2001, 21, 7721–7730. [Google Scholar] [CrossRef] [PubMed]
  192. Cheng, M.L.; Wu, C.H.; Chien, K.Y.; Lai, C.H.; Li, G.J.; Liu, Y.Y.; Lin, G.; Ho, H.Y. Enteroviral 2B Interacts with VDAC3 to Regulate Reactive Oxygen Species Generation That Is Essential to Viral Replication. Viruses 2022, 14, 1717. [Google Scholar] [CrossRef] [PubMed]
  193. Srinivas, R.V.; Su, T.; Trimble, L.A.; Lieberman, J.; Ardman, B. Enhanced susceptibility to human immunodeficiency virus infection in CD4+ T lymphocytes genetically deficient in CD43. AIDS Res. Hum. Retroviruses 1995, 11, 1015–1021. [Google Scholar] [CrossRef] [PubMed]
  194. Rothwell, S.W.; Wright, D.G. Characterization of influenza A virus binding sites on human neutrophils. J. Immunol. 1994, 152, 2358–2367. [Google Scholar] [CrossRef] [PubMed]
  195. Suenaga, T.; Satoh, T.; Somboonthum, P.; Kawaguchi, Y.; Mori, Y.; Arase, H. Myelin-associated glycoprotein mediates membrane fusion and entry of neurotropic herpesviruses. Proc. Natl. Acad. Sci. USA 2010, 107, 866–871. [Google Scholar] [CrossRef] [PubMed]
  196. van den Berg, L.H.; Sadiq, S.A.; Lederman, S.; Latov, N. The gp120 glycoprotein of HIV-1 binds to sulfatide and to the myelin associated glycoprotein. J. Neurosci. Res. 1992, 33, 513–518. [Google Scholar] [CrossRef] [PubMed]
  197. Ampuero, J.; del Campo, J.A.; Rojas, L.; Garcia-Lozano, R.J.; Buti, M.; Sola, R.; Forns, X.; Moreno-Otero, R.; Andrade, R.; Diago, M.; et al. Fine-mapping butyrophilin family genes revealed several polymorphisms influencing viral genotype selection in hepatitis C infection. Genes Immun. 2015, 16, 297–300. [Google Scholar] [CrossRef] [PubMed]
  198. Teoh, K.T.; Siu, Y.L.; Chan, W.L.; Schluter, M.A.; Liu, C.J.; Peiris, J.S.; Bruzzone, R.; Margolis, B.; Nal, B. The SARS coronavirus E protein interacts with PALS1 and alters tight junction formation and epithelial morphogenesis. Mol. Biol. Cell 2010, 21, 3838–3852. [Google Scholar] [CrossRef]
  199. He, J.; Yang, L.; Chang, P.; Yang, S.; Wang, Y.; Lin, S.; Tang, Q.; Zhang, Y. Zika Virus Induces Degradation of the Numb Protein Required through Embryonic Neurogenesis. Viruses 2023, 15, 1258. [Google Scholar] [CrossRef]
  200. Liu, D.; Cui, L.; Wang, Y.; Yang, G.; He, J.; Hao, R.; Fan, C.; Qu, M.; Liu, Z.; Wang, M.; et al. Hepatitis B e antigen and its precursors promote the progress of hepatocellular carcinoma by interacting with NUMB and decreasing p53 activity. Hepatology 2016, 64, 390–404. [Google Scholar] [CrossRef]
  201. Nadella, M.V.; Shu, S.T.; Dirksen, W.P.; Thudi, N.K.; Nadella, K.S.; Fernandez, S.A.; Lairmore, M.D.; Green, P.L.; Rosol, T.J. Expression of parathyroid hormone-related protein during immortalization of human peripheral blood mononuclear cells by HTLV-1: Implications for transformation. Retrovirology 2008, 5, 46. [Google Scholar] [CrossRef] [PubMed]
  202. Sander, W.J.; O’Neill, H.G.; Pohl, C.H. Prostaglandin E(2) As a Modulator of Viral Infections. Front. Physiol. 2017, 8, 89. [Google Scholar] [CrossRef]
  203. Kong, L.; Aoyagi, H.; Yang, Z.; Ouyang, T.; Matsuda, M.; Fujimoto, A.; Watashi, K.; Suzuki, R.; Arita, M.; Yamagoe, S.; et al. Surfeit 4 Contributes to the Replication of Hepatitis C Virus Using Double-Membrane Vesicles. J. Virol. 2020, 94. [Google Scholar] [CrossRef] [PubMed]
  204. Duan, M.; Yao, H.; Cai, Y.; Liao, K.; Seth, P.; Buch, S. HIV-1 Tat disrupts CX3CL1-CX3CR1 axis in microglia via the NF-kappaBYY1 pathway. Curr. HIV Res. 2014, 12, 189–200. [Google Scholar] [CrossRef] [PubMed]
  205. Kondo, Y.; Kimura, O.; Tanaka, Y.; Ninomiya, M.; Iwata, T.; Kogure, T.; Inoue, J.; Sugiyama, M.; Morosawa, T.; Fujisaka, Y.; et al. Differential Expression of CX3CL1 in Hepatitis B Virus-Replicating Hepatoma Cells Can Affect the Migration Activity of CX3CR1+ Immune Cells. J. Virol. 2015, 89, 7016–7027. [Google Scholar] [CrossRef] [PubMed]
  206. Todt, D.; Friesland, M.; Moeller, N.; Praditya, D.; Kinast, V.; Bruggemann, Y.; Knegendorf, L.; Burkard, T.; Steinmann, J.; Burm, R.; et al. Robust hepatitis E virus infection and transcriptional response in human hepatocytes. Proc. Natl. Acad. Sci. USA 2020, 117, 1731–1741. [Google Scholar] [CrossRef] [PubMed]
  207. Wroblewska, A.; Woziwodzka, A.; Rybicka, M.; Bielawski, K.P.; Sikorska, K. Polymorphisms Related to Iron Homeostasis Associate with Liver Disease in Chronic Hepatitis C. Viruses 2023, 15, 1710. [Google Scholar] [CrossRef] [PubMed]
  208. Mishra, A.K.; Hossain, M.M.; Sata, T.N.; Yadav, A.K.; Zadran, S.; Sah, A.K.; Nayak, B.; Shalimar; Venugopal, S.K. Hepatitis B Virus X Protein Inhibits the Expression of Barrier To Autointegration factor1 via Upregulating miR-203 Expression in Hepatic Cells. Microbiol. Spectr. 2023, 11, e0123522. [Google Scholar] [CrossRef]
  209. Wiebe, M.S.; Traktman, P. Poxviral B1 kinase overcomes barrier to autointegration factor, a host defense against virus replication. Cell Host Microbe 2007, 1, 187–197. [Google Scholar] [CrossRef]
  210. Jamin, A.; Thunuguntla, P.; Wicklund, A.; Jones, C.; Wiebe, M.S. Barrier to auto integration factor becomes dephosphorylated during HSV-1 Infection and Can Act as a host defense by impairing viral DNA replication and gene expression. PLoS ONE 2014, 9, e100511. [Google Scholar] [CrossRef]
  211. Kundlacz, C.; Pourcelot, M.; Fablet, A.; Amaral Da Silva Moraes, R.; Leger, T.; Morlet, B.; Viarouge, C.; Sailleau, C.; Turpaud, M.; Gorlier, A.; et al. Novel Function of Bluetongue Virus NS3 Protein in Regulation of the MAPK/ERK Signaling Pathway. J. Virol. 2019, 93. [Google Scholar] [CrossRef] [PubMed]
  212. Lasithiotaki, I.; Antoniou, K.M.; Derdas, S.P.; Sarchianaki, E.; Symvoulakis, E.K.; Psaraki, A.; Spandidos, D.A.; Stathopoulos, E.N.; Siafakas, N.M.; Sourvinos, G. The presence of Merkel cell polyomavirus is associated with deregulated expression of BRAF and Bcl-2 genes in non-small cell lung cancer. Int. J. Cancer 2013, 133, 604–611. [Google Scholar] [CrossRef] [PubMed]
  213. Pinto, R.M.; Bakshi, S.; Lytras, S.; Zakaria, M.K.; Swingler, S.; Worrell, J.C.; Herder, V.; Hargrave, K.E.; Varjak, M.; Cameron-Ruiz, N.; et al. BTN3A3 evasion promotes the zoonotic potential of influenza A viruses. Nature 2023, 619, 338–347. [Google Scholar] [CrossRef] [PubMed]
  214. Alagarasu, K.; Patil, P.S.; Shil, P.; Seervi, M.; Kakade, M.B.; Tillu, H.; Salunke, A. In-vitro effect of human cathelicidin antimicrobial peptide LL-37 on dengue virus type 2. Peptides 2017, 92, 23–30. [Google Scholar] [CrossRef] [PubMed]
  215. Yu, Y.; Cooper, C.L.; Wang, G.; Morwitzer, M.J.; Kota, K.; Tran, J.P.; Bradfute, S.B.; Liu, Y.; Shao, J.; Zhang, A.K.; et al. Engineered Human Cathelicidin Antimicrobial Peptides Inhibit Ebola Virus Infection. iScience 2020, 23, 100999. [Google Scholar] [CrossRef] [PubMed]
  216. LeMessurier, K.S.; Lin, Y.; McCullers, J.A.; Samarasinghe, A.E. Antimicrobial peptides alter early immune response to influenza A virus infection in C57BL/6 mice. Antivir. Res. 2016, 133, 208–217. [Google Scholar] [CrossRef] [PubMed]
  217. Enyindah-Asonye, G.; Nwankwo, A.; Rahman, M.A.; Hunegnaw, R.; Hogge, C.; Helmold Hait, S.; Ko, E.J.; Hoang, T.; Robert-Guroff, M. Overexpression of CD6 and PD-1 Identifies Dysfunctional CD8(+) T-Cells During Chronic SIV Infection of Rhesus Macaques. Front. Immunol. 2019, 10, 3005. [Google Scholar] [CrossRef] [PubMed]
  218. Hao, J.; Li, J.; Zhang, Z.; Yang, Y.; Zhou, Q.; Wu, T.; Chen, T.; Wu, Z.; Zhang, P.; Cui, J.; et al. NLRC5 restricts dengue virus infection by promoting the autophagic degradation of viral NS3 through E3 ligase CUL2 (cullin 2). Autophagy 2023, 19, 1332–1347. [Google Scholar] [CrossRef]
  219. Hyeon, S.; Lee, M.K.; Kim, Y.E.; Lee, G.M.; Ahn, J.H. Degradation of SAMHD1 Restriction Factor Through Cullin-Ring E3 Ligase Complexes During Human Cytomegalovirus Infection. Front. Cell. Infect. Microbiol. 2020, 10, 391. [Google Scholar] [CrossRef]
  220. Zhang, W.; Wang, H.; Li, Z.; Liu, X.; Liu, G.; Harris, R.S.; Yu, X.F. Cellular requirements for bovine immunodeficiency virus Vif-mediated inactivation of bovine APOBEC3 proteins. J. Virol. 2014, 88, 12528–12540. [Google Scholar] [CrossRef]
  221. Pawlica, P.; Yario, T.A.; White, S.; Wang, J.; Moss, W.N.; Hui, P.; Vinetz, J.M.; Steitz, J.A. SARS-CoV-2 expresses a microRNA-like small RNA able to selectively repress host genes. Proc. Natl. Acad. Sci. USA 2021, 118, e2116668118. [Google Scholar] [CrossRef] [PubMed]
  222. Madsen, C.; Hooper, I.; Lundberg, L.; Shafagati, N.; Johnson, A.; Senina, S.; de la Fuente, C.; Hoover, L.I.; Fredricksen, B.L.; Dinman, J.; et al. Small molecule inhibitors of Ago2 decrease Venezuelan equine encephalitis virus replication. Antivir. Res. 2014, 112, 26–37. [Google Scholar] [CrossRef] [PubMed]
  223. Wu, X.; Wang, S.; Li, C.; Shi, J.; Peng, Z.; Liu, C.; Han, H.; Ma, Y.; Zheng, L.; Xu, S.; et al. CRISPR/Cas9-Mediated Knockout of the Dicer and Ago2 Genes in BHK-21 Cell Promoted Seneca Virus A Replication and Enhanced Autophagy. Front. Cell. Infect. Microbiol. 2022, 12, 865744. [Google Scholar] [CrossRef] [PubMed]
  224. Wang, S.; Sun, X.; Yi, C.; Zhang, D.; Lin, X.; Sun, X.; Chen, H.; Jin, M. AGO2 Negatively Regulates Type I Interferon Signaling Pathway by Competition Binding IRF3 with CBP/p300. Front. Cell. Infect. Microbiol. 2017, 7, 195. [Google Scholar] [CrossRef]
  225. Devadas, K.; Biswas, S.; Haleyurgirisetty, M.; Wood, O.; Ragupathy, V.; Lee, S.; Hewlett, I. Analysis of Host Gene Expression Profile in HIV-1 and HIV-2 Infected T-Cells. PLoS ONE 2016, 11, e0147421. [Google Scholar] [CrossRef]
  226. Conn, K.L.; Hendzel, M.J.; Schang, L.M. The differential mobilization of histones H3.1 and H3.3 by herpes simplex virus 1 relates histone dynamics to the assembly of viral chromatin. PLoS Pathog. 2013, 9, e1003695. [Google Scholar] [CrossRef]
  227. Ambagala, A.P.; Bosma, T.; Ali, M.A.; Poustovoitov, M.; Chen, J.J.; Gershon, M.D.; Adams, P.D.; Cohen, J.I. Varicella-zoster virus immediate-early 63 protein interacts with human antisilencing function 1 protein and alters its ability to bind histones h3.1 and h3.3. J. Virol. 2009, 83, 200–209. [Google Scholar] [CrossRef]
  228. Hsieh, I.N.; White, M.; Hoeksema, M.; Deluna, X.; Hartshorn, K. Histone H4 potentiates neutrophil inflammatory responses to influenza A virus: Down-modulation by H4 binding to C-reactive protein and Surfactant protein D. PLoS ONE 2021, 16, e0247605. [Google Scholar] [CrossRef]
  229. Nishitsuji, H.; Ujino, S.; Harada, K.; Shimotohno, K. TIP60 Complex Inhibits Hepatitis B Virus Transcription. J. Virol. 2018, 92. [Google Scholar] [CrossRef]
  230. Truong, A.D.; Tran, H.T.T.; Nguyen, H.T.; Chu, N.T.; Hong, Y.H.; Lillehoj, H.S.; Dang, H.V.; Song, K.D. Molecular and functional characterization of chicken interleukin 1 receptor 2 (chIL-1R2). Poult. Sci. 2023, 102, 102399. [Google Scholar] [CrossRef]
  231. Li, Z.; Li, Y.; Sun, R.; Li, S.; Chen, L.; Zhan, Y.; Xie, M.; Yang, J.; Wang, Y.; Zhu, A.; et al. Longitudinal virological changes and underlying pathogenesis in hospitalized COVID-19 patients in Guangzhou, China. Sci. China. Life Sci. 2021, 64, 2129–2143. [Google Scholar] [CrossRef]
  232. Cho, Y.B.; Hong, S.; Kang, K.W.; Kang, J.H.; Lee, S.M.; Seo, Y.J. Selective and ATP-competitive kinesin KIF18A inhibitor suppresses the replication of influenza A virus. J. Cell. Mol. Med. 2020, 24, 5463–5475. [Google Scholar] [CrossRef]
  233. Sorgho, P.A.; Martinson, J.J.; Djigma, F.W.; Yonli, A.T.; Nagalo, B.M.; Compaore, T.R.; Obiri-Yeboah, D.; Diarra, B.; Sombie, H.K.; Zongo, A.W.; et al. Insights into the Interplay between KIR Gene Frequencies and Chronic HBV Infection in Burkina Faso. Mediterr. J. Hematol. Infect. Dis. 2018, 10, e2018060. [Google Scholar] [CrossRef]
  234. Podhorzer, A.; Dirchwolf, M.; Machicote, A.; Belen, S.; Montal, S.; Paz, S.; Fainboim, H.; Podesta, L.G.; Fainboim, L. The Clinical Features of Patients with Chronic Hepatitis C Virus Infections Are Associated with Killer Cell Immunoglobulin-Like Receptor Genes and Their Expression on the Surface of Natural Killer Cells. Front. Immunol. 2017, 8, 1912. [Google Scholar] [CrossRef]
  235. Aoukaty, A.; Lee, I.F.; Wu, J.; Tan, R. Chronic active Epstein-Barr virus infection associated with low expression of leukocyte-associated immunoglobulin-like receptor-1 (LAIR-1) on natural killer cells. J. Clin. Immunol. 2003, 23, 141–145. [Google Scholar] [CrossRef]
  236. Kumawat, K.; Geerdink, R.J.; Hennus, M.P.; Roda, M.A.; van Ark, I.; Leusink-Muis, T.; Folkerts, G.; van Oort-Jansen, A.; Mazharian, A.; Watson, S.P.; et al. LAIR-1 Limits Neutrophilic Airway Inflammation. Front. Immunol. 2019, 10, 842. [Google Scholar] [CrossRef]
  237. Martinez-Torres, F.J.; Wagner, S.; Haas, J.; Kehm, R.; Sellner, J.; Hacke, W.; Meyding-Lamade, U. Increased presence of matrix metalloproteinases 2 and 9 in short- and long-term experimental herpes simplex virus encephalitis. Neurosci. Lett. 2004, 368, 274–278. [Google Scholar] [CrossRef]
  238. Ahmed, F.; Husain, M. Human N-Alpha-Acetyltransferase 60 Promotes Influenza A Virus Infection by Dampening the Interferon Alpha Signaling. Front. Immunol. 2021, 12, 771792. [Google Scholar] [CrossRef]
  239. Alpuche-Lazcano, S.P.; Saliba, J.; Costa, V.V.; Campolina-Silva, G.H.; Marim, F.M.; Ribeiro, L.S.; Blank, V.; Mouland, A.J.; Teixeira, M.M.; Gatignol, A. Profound downregulation of neural transcription factor Npas4 and Nr4a family in fetal mice neurons infected with Zika virus. PLoS Neglected Trop. Dis. 2021, 15, e0009425. [Google Scholar] [CrossRef]
  240. He, Z.; Jing, S.; Yang, T.; Chen, J.; Huang, F.; Zhang, W.; Peng, Z.; Liu, B.; Ma, X.; Wu, L.; et al. PIWIL4 Maintains HIV-1 Latency by Enforcing Epigenetically Suppressive Modifications on the 5’ Long Terminal Repeat. J. Virol. 2020, 94. [Google Scholar] [CrossRef]
  241. Zenner, H.L.; Yoshimura, S.; Barr, F.A.; Crump, C.M. Analysis of Rab GTPase-activating proteins indicates that Rab1a/b and Rab43 are important for herpes simplex virus 1 secondary envelopment. J. Virol. 2011, 85, 8012–8021. [Google Scholar] [CrossRef] [PubMed]
  242. Baik, S.Y.; Yun, H.S.; Lee, H.J.; Lee, M.H.; Jung, S.E.; Kim, J.W.; Jeon, J.P.; Shin, Y.K.; Rhee, H.S.; Kimm, K.C.; et al. Identification of stathmin 1 expression induced by Epstein-Barr virus in human B lymphocytes. Cell Prolif. 2007, 40, 268–281. [Google Scholar] [CrossRef] [PubMed]
  243. Day, P.M.; Thompson, C.D.; Schowalter, R.M.; Lowy, D.R.; Schiller, J.T. Identification of a role for the trans-Golgi network in human papillomavirus 16 pseudovirus infection. J. Virol. 2013, 87, 3862–3870. [Google Scholar] [CrossRef] [PubMed]
  244. Murray, J.L.; Mavrakis, M.; McDonald, N.J.; Yilla, M.; Sheng, J.; Bellini, W.J.; Zhao, L.; Le Doux, J.M.; Shaw, M.W.; Luo, C.C.; et al. Rab9 GTPase is required for replication of human immunodeficiency virus type 1, filoviruses, and measles virus. J. Virol. 2005, 79, 11742–11751. [Google Scholar] [CrossRef] [PubMed]
  245. Ren, C.; Xie, R.; Yao, Y.; Yu, M.; Chang, F.; Xing, L.; Zhang, Y.; Liu, Y.; Wang, S.; Farooque, M.; et al. MiR-125b Suppression Inhibits Apoptosis and Negatively Regulates Sema4D in Avian Leukosis Virus-Transformed Cells. Viruses 2019, 11, 728. [Google Scholar] [CrossRef]
  246. Eriksson, E.M.; Milush, J.M.; Ho, E.L.; Batista, M.D.; Holditch, S.J.; Keh, C.E.; Norris, P.J.; Keating, S.M.; Deeks, S.G.; Hunt, P.W.; et al. Expansion of CD8+ T cells lacking Sema4D/CD100 during HIV-1 infection identifies a subset of T cells with decreased functional capacity. Blood 2012, 119, 745–755. [Google Scholar] [CrossRef]
  247. He, Y.; Guo, Y.; Fan, C.; Lei, Y.; Zhou, Y.; Zhang, M.; Ye, C.; Ji, G.; Ma, L.; Lian, J.; et al. Interferon-alpha-Enhanced CD100/Plexin-B1/B2 Interactions Promote Natural Killer Cell Functions in Patients with Chronic Hepatitis C Virus Infection. Front. Immunol. 2017, 8, 1435. [Google Scholar] [CrossRef] [PubMed]
  248. Wang, Q.; Zhang, Q.; Zheng, M.; Wen, J.; Li, Q.; Zhao, G. Viral-Host Interactome Analysis Reveals Chicken STAU2 Interacts With Non-structural Protein 1 and Promotes the Replication of H5N1 Avian Influenza Virus. Front. Immunol. 2021, 12, 590679. [Google Scholar] [CrossRef]
  249. Yan, N.; Regalado-Magdos, A.D.; Stiggelbout, B.; Lee-Kirsch, M.A.; Lieberman, J. The cytosolic exonuclease TREX1 inhibits the innate immune response to human immunodeficiency virus type 1. Nat. Immunol. 2010, 11, 1005–1013. [Google Scholar] [CrossRef]
  250. Yeh, D.W.; Zhao, X.; Siddique, H.R.; Zheng, M.; Choi, H.Y.; Machida, T.; Narayanan, P.; Kou, Y.; Punj, V.; Tahara, S.M.; et al. MSI2 promotes translation of multiple IRES-containing oncogenes and virus to induce self-renewal of tumor initiating stem-like cells. Cell Death Discov. 2023, 9, 141. [Google Scholar] [CrossRef]
  251. Kim, J.; Kwon, H.; Kalsoom, F.; Sajjad, M.A.; Lee, H.W.; Lim, J.H.; Jung, J.; Chwae, Y.J.; Park, S.; Shin, H.J.; et al. Ca(2+)/Calmodulin-Dependent Protein Kinase II Inhibits Hepatitis B Virus Replication from cccDNA via AMPK Activation and AKT/mTOR Suppression. Microorganisms 2022, 10, 498. [Google Scholar] [CrossRef] [PubMed]
  252. Haolong, C.; Du, N.; Hongchao, T.; Yang, Y.; Wei, Z.; Hua, Z.; Wenliang, Z.; Lei, S.; Po, T. Enterovirus 71 VP1 activates calmodulin-dependent protein kinase II and results in the rearrangement of vimentin in human astrocyte cells. PLoS ONE 2013, 8, e73900. [Google Scholar] [CrossRef] [PubMed]
  253. Stefanovic, S.; Windsor, M.; Nagata, K.I.; Inagaki, M.; Wileman, T. Vimentin rearrangement during African swine fever virus infection involves retrograde transport along microtubules and phosphorylation of vimentin by calcium calmodulin kinase II. J. Virol. 2005, 79, 11766–11775. [Google Scholar] [CrossRef] [PubMed]
  254. Huang, S.; Li, Z.; Wu, Z.; Liu, C.; Yu, M.; Wen, M.; Zhang, L.; Wang, X. DDAH2 suppresses RLR-MAVS-mediated innate antiviral immunity by stimulating nitric oxide-activated, Drp1-induced mitochondrial fission. Sci. Signal. 2021, 14, eabc7931. [Google Scholar] [CrossRef] [PubMed]
  255. Xiao, S.; Wang, Q.; Jia, J.; Cong, P.; Mo, D.; Yu, X.; Qin, L.; Li, A.; Niu, Y.; Zhu, K.; et al. Proteome changes of lungs artificially infected with H-PRRSV and N-PRRSV by two-dimensional fluorescence difference gel electrophoresis. Virol. J. 2010, 7, 107. [Google Scholar] [CrossRef] [PubMed]
  256. Liu, Q.; Guan, X.; Lv, J.; Li, X.; Wang, Y.; Li, L. Limb-bud and Heart (LBH) functions as a tumor suppressor of nasopharyngeal carcinoma by inducing G1/S cell cycle arrest. Sci. Rep. 2015, 5, 7626. [Google Scholar] [CrossRef] [PubMed]
  257. Wang, C.; McPherson, A.J.; Jones, R.B.; Kawamura, K.S.; Lin, G.H.; Lang, P.A.; Ambagala, T.; Pellegrini, M.; Calzascia, T.; Aidarus, N.; et al. Loss of the signaling adaptor TRAF1 causes CD8+ T cell dysregulation during human and murine chronic infection. J. Exp. Med. 2012, 209, 77–91. [Google Scholar] [CrossRef]
  258. Siegler, G.; Meyer, B.; Dawson, C.; Brachtel, E.; Lennerz, J.; Koch, C.; Kremmer, E.; Niedobitek, E.; Gonnella, R.; Pilch, B.Z.; et al. Expression of tumor necrosis factor receptor-associated factor 1 in nasopharyngeal carcinoma: Possible upregulation by Epstein-Barr virus latent membrane protein 1. Int. J. Cancer 2004, 112, 265–272. [Google Scholar] [CrossRef]
  259. Schneider, M.A.; Spoden, G.A.; Florin, L.; Lambert, C. Identification of the dynein light chains required for human papillomavirus infection. Cell. Microbiol. 2011, 13, 32–46. [Google Scholar] [CrossRef]
  260. Yu, T.; Ding, Y.; Zhang, Y.; Liu, Y.; Li, Y.; Lei, J.; Zhou, J.; Song, S.; Hu, B. Circular RNA GATAD2A promotes H1N1 replication through inhibiting autophagy. Vet. Microbiol. 2019, 231, 238–245. [Google Scholar] [CrossRef]
  261. Cruz, J.L.; Sola, I.; Becares, M.; Alberca, B.; Plana, J.; Enjuanes, L.; Zuniga, S. Coronavirus gene 7 counteracts host defenses and modulates virus virulence. PLoS Pathog. 2011, 7, e1002090. [Google Scholar] [CrossRef] [PubMed]
  262. Yang, X.; Yang, W.; Zhang, W.; Li, J.; Yang, G.; Zhao, S.; Zheng, Y. Cap Is the Protease of the Porcine Circovirus 2. Viruses 2022, 14, 1550. [Google Scholar] [CrossRef] [PubMed]
  263. Zhang, X.; Lin, X.; Qin, C.; Huang, K.; Sun, X.; Zhao, L.; Jin, M. Avian Chaperonin Containing TCP1 Subunit 5 Supports Influenza A Virus Replication by Interacting With Viral Nucleoprotein, PB1, and PB2 Proteins. Front. Microbiol. 2020, 11, 538355. [Google Scholar] [CrossRef] [PubMed]
  264. Ricciardi, S.; Guarino, A.M.; Giaquinto, L.; Polishchuk, E.V.; Santoro, M.; Di Tullio, G.; Wilson, C.; Panariello, F.; Soares, V.C.; Dias, S.S.G.; et al. The role of NSP6 in the biogenesis of the SARS-CoV-2 replication organelle. Nature 2022, 606, 761–768. [Google Scholar] [CrossRef] [PubMed]
  265. Mohl, B.P.; Bartlett, C.; Mankouri, J.; Harris, M. Early events in the generation of autophagosomes are required for the formation of membrane structures involved in hepatitis C virus genome replication. J. Gen. Virol. 2016, 97, 680–693. [Google Scholar] [CrossRef] [PubMed]
  266. He, L.; Yang, J.; Hao, Y.; Yang, X.; Shi, X.; Zhang, D.; Zhao, D.; Yan, W.; Bie, X.; Chen, L.; et al. DDX20: A Multifunctional Complex Protein. Molecules 2023, 28, 7198. [Google Scholar] [CrossRef] [PubMed]
  267. Golden, S.; Yu, X.M.; Odorico, S.; Jain, V.; Marin, A.; Ma, S.; Kenney, S.; Chen, H. The Epstein-Barr virus EBNA2 protein induces a subset of NOTCH target genes in thyroid cancer cell lines but fails to suppress proliferation. Surgery 2017, 161, 195–201. [Google Scholar] [CrossRef] [PubMed]
  268. Ye, C.; Zhang, X.; Chen, X.; Cao, Q.; Zhang, X.; Zhou, Y.; Li, W.; Hong, L.; Xie, H.; Liu, X.; et al. Multiple novel hepatocellular carcinoma signature genes are commonly controlled by the master pluripotency factor OCT4. Cell. Oncol. 2020, 43, 279–295. [Google Scholar] [CrossRef]
  269. Ashrafi, F.; Ghezeldasht, S.A.; Ghobadi, M.Z. Identification of joint gene players implicated in the pathogenesis of HTLV-1 and BLV through a comprehensive system biology analysis. Microb. Pathog. 2021, 160, 105153. [Google Scholar] [CrossRef]
  270. Li, S.; Li, R.; Ahmad, I.; Liu, X.; Johnson, S.F.; Sun, L.; Zheng, Y.H. Cul3-KLHL20 E3 ubiquitin ligase plays a key role in the arms race between HIV-1 Nef and host SERINC5 restriction. Nat. Commun. 2022, 13, 2242. [Google Scholar] [CrossRef]
  271. Bedadala, G.R.; Pinnoji, R.C.; Hsia, S.C. Early growth response gene 1 (Egr-1) regulates HSV-1 ICP4 and ICP22 gene expression. Cell Res. 2007, 17, 546–555. [Google Scholar] [CrossRef]
  272. Cai, Y.; Xia, J.; Wang, N.; Zhou, H. Identification of prognostic alternative splicing signatures in hepatitis B or/and C viruses related hepatocellular carcinoma. Genomics 2020, 112, 3396–3406. [Google Scholar] [CrossRef] [PubMed]
  273. Fino, K.K.; Yang, L.; Silveyra, P.; Hu, S.; Umstead, T.M.; DiAngelo, S.; Halstead, E.S.; Cooper, T.K.; Abraham, T.; Takahashi, Y.; et al. SH3GLB2/endophilin B2 regulates lung homeostasis and recovery from severe influenza A virus infection. Sci. Rep. 2017, 7, 7262. [Google Scholar] [CrossRef] [PubMed]
  274. Liu, Q.; Yu, Y.Y.; Wang, H.Y.; Wang, J.F.; He, X.J. The IFN-gamma-induced immunoproteasome is suppressed in highly pathogenic porcine reproductive and respiratory syndrome virus-infected alveolar macrophages. Vet. Immunol. Immunopathol. 2020, 226, 110069. [Google Scholar] [CrossRef]
  275. Chavali, P.L.; Stojic, L.; Meredith, L.W.; Joseph, N.; Nahorski, M.S.; Sanford, T.J.; Sweeney, T.R.; Krishna, B.A.; Hosmillo, M.; Firth, A.E.; et al. Neurodevelopmental protein Musashi-1 interacts with the Zika genome and promotes viral replication. Science 2017, 357, 83–88. [Google Scholar] [CrossRef] [PubMed]
  276. Mangeat, B.; Gers-Huber, G.; Lehmann, M.; Zufferey, M.; Luban, J.; Piguet, V. HIV-1 Vpu neutralizes the antiviral factor Tetherin/BST-2 by binding it and directing its beta-TrCP2-dependent degradation. PLoS Pathog. 2009, 5, e1000574. [Google Scholar] [CrossRef] [PubMed]
  277. Kainulainen, M.; Lau, S.; Samuel, C.E.; Hornung, V.; Weber, F. NSs Virulence Factor of Rift Valley Fever Virus Engages the F-Box Proteins FBXW11 and beta-TRCP1 To Degrade the Antiviral Protein Kinase PKR. J. Virol. 2016, 90, 6140–6147. [Google Scholar] [CrossRef] [PubMed]
  278. Yan, Q.; Zeng, Z.; Gong, Z.; Zhang, W.; Li, X.; He, B.; Song, Y.; Li, Q.; Zeng, Y.; Liao, Q.; et al. EBV-miR-BART10-3p facilitates epithelial-mesenchymal transition and promotes metastasis of nasopharyngeal carcinoma by targeting BTRC. Oncotarget 2015, 6, 41766–41782. [Google Scholar] [CrossRef]
  279. Margottin, F.; Bour, S.P.; Durand, H.; Selig, L.; Benichou, S.; Richard, V.; Thomas, D.; Strebel, K.; Benarous, R. A novel human WD protein, h-beta TrCp, that interacts with HIV-1 Vpu connects CD4 to the ER degradation pathway through an F-box motif. Mol. Cell 1998, 1, 565–574. [Google Scholar] [CrossRef]
  280. Kumar, S.; Verma, R.; Saha, S.; Agrahari, A.K.; Shukla, S.; Singh, O.N.; Berry, U.; Anurag; Maiti, T.K.; Asthana, S.; et al. RNA-Protein Interactome at the Hepatitis E Virus Internal Ribosome Entry Site. Microbiol. Spectr. 2023, 11, e0282722. [Google Scholar] [CrossRef]
  281. Lian, Z.; Liu, J.; Li, L.; Li, X.; Tufan, N.L.; Wu, M.C.; Wang, H.Y.; Arbuthnot, P.; Kew, M.; Feitelson, M.A. Human S15a expression is upregulated by hepatitis B virus X protein. Mol. Carcinog. 2004, 40, 34–46. [Google Scholar] [CrossRef] [PubMed]
  282. Yan, L.; Qu, S.; Liu, G.; Liu, L.; Yu, Y.; Ding, G.; Zhao, Y.; Li, Y.; Xie, Y.; Zhang, J.; et al. Comparative Transcriptomic Analysis of Primary Duck Hepatocytes Provides Insight into Differential Susceptibility to DHBV Infection. PLoS ONE 2016, 11, e0149702. [Google Scholar] [CrossRef] [PubMed]
  283. Pfaller, C.K.; George, C.X.; Samuel, C.E. Adenosine Deaminases Acting on RNA (ADARs) and Viral Infections. Annu. Rev. Virol. 2021, 8, 239–264. [Google Scholar] [CrossRef] [PubMed]
  284. Monteiro, J.T.; Schon, K.; Ebbecke, T.; Goethe, R.; Ruland, J.; Baumgartner, W.; Becker, S.C.; Lepenies, B. The CARD9-Associated C-Type Lectin, Mincle, Recognizes La Crosse Virus (LACV) but Plays a Limited Role in Early Antiviral Responses against LACV. Viruses 2019, 11, 303. [Google Scholar] [CrossRef] [PubMed]
  285. Poeck, H.; Bscheider, M.; Gross, O.; Finger, K.; Roth, S.; Rebsamen, M.; Hannesschlager, N.; Schlee, M.; Rothenfusser, S.; Barchet, W.; et al. Recognition of RNA virus by RIG-I results in activation of CARD9 and inflammasome signaling for interleukin 1 beta production. Nat. Immunol. 2010, 11, 63–69. [Google Scholar] [CrossRef] [PubMed]
  286. Sun, C.; Zhang, X.; Yu, Y.; Li, Z.; Xie, Y. CARD9 mediates T cell inflammatory response in Coxsackievirus B3-induced acute myocarditis. Cardiovasc. Pathol. Off. J. Soc. Cardiovasc. Pathol. 2020, 49, 107261. [Google Scholar] [CrossRef]
  287. Peters, G.A.; Khoo, D.; Mohr, I.; Sen, G.C. Inhibition of PACT-mediated activation of PKR by the herpes simplex virus type 1 Us11 protein. J. Virol. 2002, 76, 11054–11064. [Google Scholar] [CrossRef]
  288. Hume, A.; Muhlberger, E. Marburg Virus Viral Protein 35 Inhibits Protein Kinase R Activation in a Cell Type-Specific Manner. J. Infect. Dis. 2018, 218, S403–S408. [Google Scholar] [CrossRef]
  289. Tseng, Y.Y.; Liao, G.R.; Sen, G.C.; Lin, F.Y.; Hsu, W.L. Regulation of PACT-Mediated Protein Kinase Activation by the OV20.0 Protein of Orf Virus. J. Virol. 2015, 89, 11619–11629. [Google Scholar] [CrossRef]
  290. Zhang, W.; Liu, Y.; Li, M.; Zhu, J.; Li, X.; Luo, T.R.; Liang, J. Host Desmin Interacts with RABV Matrix Protein and Facilitates Virus Propagation. Viruses 2023, 15, 434. [Google Scholar] [CrossRef]
  291. Nedellec, P.; Vicart, P.; Laurent-Winter, C.; Martinat, C.; Prevost, M.C.; Brahic, M. Interaction of Theiler’s virus with intermediate filaments of infected cells. J. Virol. 1998, 72, 9553–9560. [Google Scholar] [CrossRef] [PubMed]
  292. Shoeman, R.L.; Sachse, C.; Honer, B.; Mothes, E.; Kaufmann, M.; Traub, P. Cleavage of human and mouse cytoskeletal and sarcomeric proteins by human immunodeficiency virus type 1 protease. Actin, desmin, myosin, and tropomyosin. Am. J. Pathol. 1993, 142, 221–230. [Google Scholar] [PubMed]
  293. Dang, W.; Yin, Y.; Wang, Y.; Wang, W.; Su, J.; Sprengers, D.; van der Laan, L.J.W.; Felczak, K.; Pankiewicz, K.W.; Chang, K.O.; et al. Inhibition of Calcineurin or IMP Dehydrogenase Exerts Moderate to Potent Antiviral Activity against Norovirus Replication. Antimicrob. Agents Chemother. 2017, 61. [Google Scholar] [CrossRef] [PubMed]
  294. Na Rangsee, N.; Yanatatsaneejit, P.; Pisitkun, T.; Somparn, P.; Jintaridth, P.; Topanurak, S. Host proteome linked to HPV E7-mediated specific gene hypermethylation in cancer pathways. Infect. Agents Cancer 2020, 15, 7. [Google Scholar] [CrossRef]
  295. Russell, T.; Samolej, J.; Hollinshead, M.; Smith, G.L.; Kite, J.; Elliott, G. Novel Role for ESCRT-III Component CHMP4C in the Integrity of the Endocytic Network Utilized for Herpes Simplex Virus Envelopment. mBio 2021, 12. [Google Scholar] [CrossRef]
  296. von Schwedler, U.K.; Stuchell, M.; Muller, B.; Ward, D.M.; Chung, H.Y.; Morita, E.; Wang, H.E.; Davis, T.; He, G.P.; Cimbora, D.M.; et al. The protein network of HIV budding. Cell 2003, 114, 701–713. [Google Scholar] [CrossRef]
  297. Tabata, K.; Arimoto, M.; Arakawa, M.; Nara, A.; Saito, K.; Omori, H.; Arai, A.; Ishikawa, T.; Konishi, E.; Suzuki, R.; et al. Unique Requirement for ESCRT Factors in Flavivirus Particle Formation on the Endoplasmic Reticulum. Cell Rep. 2016, 16, 2339–2347. [Google Scholar] [CrossRef]
Figure 1. Venn diagrams depicting HEV ORF interactors identified by MAPPIT and KISS analyses. (A) Interactors of the ORF2 protein identified by MAPPIT analyses. (B) Interactors of the ORF3 protein identified by both MAPPIT and KISS analyses. (C) Interactors of ORF4 protein identified by both MAPPIT and KISS analyses. (D) Venn diagram shows distinct and overlapping interacting proteins of all HEV proteins analyzed.
Figure 1. Venn diagrams depicting HEV ORF interactors identified by MAPPIT and KISS analyses. (A) Interactors of the ORF2 protein identified by MAPPIT analyses. (B) Interactors of the ORF3 protein identified by both MAPPIT and KISS analyses. (C) Interactors of ORF4 protein identified by both MAPPIT and KISS analyses. (D) Venn diagram shows distinct and overlapping interacting proteins of all HEV proteins analyzed.
Viruses 15 02412 g001
Figure 2. Gene ontology (GO) annotation of the ORF2 hit list. GO annotation for the ORF2 interacting proteins of both gt-1 and gt-3 as identified by MAPPIT analysis; different categories are depicted: biological process (A), cellular compartment (B) and molecular function (C). Red circles indicate the number of proteins belonging to each term and shown as percentage of the total ORF2 hit list (=37). p-value was set to equal to or smaller than 0.05 (indicated by dashed line).
Figure 2. Gene ontology (GO) annotation of the ORF2 hit list. GO annotation for the ORF2 interacting proteins of both gt-1 and gt-3 as identified by MAPPIT analysis; different categories are depicted: biological process (A), cellular compartment (B) and molecular function (C). Red circles indicate the number of proteins belonging to each term and shown as percentage of the total ORF2 hit list (=37). p-value was set to equal to or smaller than 0.05 (indicated by dashed line).
Viruses 15 02412 g002
Figure 3. Gene ontology annotation of the ORF3 hit list. GO annotation for the ORF3 interacting proteins of both gt-1 and gt-3 as identified by MAPPIT and KISS analyses. Different categories are depicted: biological process (A), cellular compartment (B) and molecular function (C). Red circles indicate the number of proteins belonging to each term and shown as percentage of the total ORF3 hit list (=187). p-value was set to equal to or smaller than 0.05 (indicated by dashed line).
Figure 3. Gene ontology annotation of the ORF3 hit list. GO annotation for the ORF3 interacting proteins of both gt-1 and gt-3 as identified by MAPPIT and KISS analyses. Different categories are depicted: biological process (A), cellular compartment (B) and molecular function (C). Red circles indicate the number of proteins belonging to each term and shown as percentage of the total ORF3 hit list (=187). p-value was set to equal to or smaller than 0.05 (indicated by dashed line).
Viruses 15 02412 g003
Figure 4. Gene ontology annotation of the ORF4 hit list. GO annotation for the gt-1 ORF4 interacting proteins as identified by MAPPIT and KISS analyses, different categories are depicted: biological process (A), cellular compartment (B) and molecular function (C). Red circles indicate the number of proteins belonging to each term and shown as percentage of the total ORF4 hit list (=91). p-value was set to equal to or smaller than 0.05 (indicated by dashed line).
Figure 4. Gene ontology annotation of the ORF4 hit list. GO annotation for the gt-1 ORF4 interacting proteins as identified by MAPPIT and KISS analyses, different categories are depicted: biological process (A), cellular compartment (B) and molecular function (C). Red circles indicate the number of proteins belonging to each term and shown as percentage of the total ORF4 hit list (=91). p-value was set to equal to or smaller than 0.05 (indicated by dashed line).
Viruses 15 02412 g004
Figure 5. SHARPIN affects the induction of IFN response. (A) PLC3WT and PLC3SHARPIN-KO cells (indicated, respectively, with + and −), are mock- or HEV ORF3-transfected (indicated, respectively, with − and +). Successful knockout and ORF3 transfection verified by Western blot. B-actin is displayed as a control. Two replicates of each condition are displayed. (B) PLC3WT and PLC3SHARPIN-KO cells were mock- or HEV ORF3-transfected and two days later transfected with poly (I:C). IFN-β (left) and IFN-λ mRNA levels measured by qRT-PCR. Transcripts normalized to reference genes (ATP5B, CYC1, YWHAZ, HPRT1, RPL30) and scaled to PLC3WT—MOCK (ORF3-). (C) PLC3WT and PLC3SHARPIN-KO cells infected with HEV (MOI 22000) and infection levels analyzed 6 days later by HEV ORF2 immunostaining. Representative confocal images on the right. Nuclei in blue and ORF2 in green. Results are represented as mean ± SEM compiled from three independent experiments. Statistical significance calculated by (multiple) t-test. Differences were considered as statistically significant when p < 0.05 (*).
Figure 5. SHARPIN affects the induction of IFN response. (A) PLC3WT and PLC3SHARPIN-KO cells (indicated, respectively, with + and −), are mock- or HEV ORF3-transfected (indicated, respectively, with − and +). Successful knockout and ORF3 transfection verified by Western blot. B-actin is displayed as a control. Two replicates of each condition are displayed. (B) PLC3WT and PLC3SHARPIN-KO cells were mock- or HEV ORF3-transfected and two days later transfected with poly (I:C). IFN-β (left) and IFN-λ mRNA levels measured by qRT-PCR. Transcripts normalized to reference genes (ATP5B, CYC1, YWHAZ, HPRT1, RPL30) and scaled to PLC3WT—MOCK (ORF3-). (C) PLC3WT and PLC3SHARPIN-KO cells infected with HEV (MOI 22000) and infection levels analyzed 6 days later by HEV ORF2 immunostaining. Representative confocal images on the right. Nuclei in blue and ORF2 in green. Results are represented as mean ± SEM compiled from three independent experiments. Statistical significance calculated by (multiple) t-test. Differences were considered as statistically significant when p < 0.05 (*).
Viruses 15 02412 g005
Figure 6. RNF5 affects the induction of IFN response. (A) PLC3WT and PLC3RNF5-KO cells (indicated, respectively, with + and −), are mock- or HEV ORF3-transfected (indicated, respectively, with − and +). Successful knockout and ORF3 transfection verified by Western blot (upper band in the RNF5 blot is aspecific). B-actin is displayed as a control. Two replicates of each condition are displayed. (B) PLC3WT and PLC3RNF5-KO cells were mock- or HEVORF3-transfected and two days later transfected with poly (I:C). IFN-β (left) and IFN-λ mRNA levels measured by qRT-PCR. Transcripts normalized to reference genes (ATP5B, CYC1 and YWHAZ) and scaled to PLC3WT—MOCK (ORF3-). (C) PLC3WT and PLC3RNF5-KO cells infected with HEV (MOI 22,000) and infection levels analyzed 6 days later by HEV ORF2 immunostaining. Representative confocal images on the right. Nuclei in blue and ORF2 in green. Results represented as mean ± SEM from three independent experiments. Statistical significance calculated by (multiple) t-test. The level of significance is indicated with p < 0.01 (**) or p < 0.05 (*).
Figure 6. RNF5 affects the induction of IFN response. (A) PLC3WT and PLC3RNF5-KO cells (indicated, respectively, with + and −), are mock- or HEV ORF3-transfected (indicated, respectively, with − and +). Successful knockout and ORF3 transfection verified by Western blot (upper band in the RNF5 blot is aspecific). B-actin is displayed as a control. Two replicates of each condition are displayed. (B) PLC3WT and PLC3RNF5-KO cells were mock- or HEVORF3-transfected and two days later transfected with poly (I:C). IFN-β (left) and IFN-λ mRNA levels measured by qRT-PCR. Transcripts normalized to reference genes (ATP5B, CYC1 and YWHAZ) and scaled to PLC3WT—MOCK (ORF3-). (C) PLC3WT and PLC3RNF5-KO cells infected with HEV (MOI 22,000) and infection levels analyzed 6 days later by HEV ORF2 immunostaining. Representative confocal images on the right. Nuclei in blue and ORF2 in green. Results represented as mean ± SEM from three independent experiments. Statistical significance calculated by (multiple) t-test. The level of significance is indicated with p < 0.01 (**) or p < 0.05 (*).
Viruses 15 02412 g006
Table 1. PCR primers for amplification of ORF3 for MAPPIT and KISS assays.
Table 1. PCR primers for amplification of ORF3 for MAPPIT and KISS assays.
Primer NameSequence (5′ → 3′)
Gt-1 ORF2 MAPPIT FWgtcgacgagctccggatccatgcgccctcggcctattttg
Gt-1 ORF2 MAPPIT RVgcggccgccaaataaactataactcccgagttttacccaccttcatc
Gt-1 ORF2 KISS FWcaattgaccatgcgccctcggcctattttg
Gt-1 ORF2 KISS RVgcggccgctaactcccgagttttacccaccttcatcttaaggcgctg
Gt-1 ORF3 MAPPIT FWgtcgacgagctccggatccatgaataacatgtcttttgctgcgcccatg
Gt-1 ORF3 MAPPIT RVgcggccgcggagcgaccgcggttagc
Gt-1 ORF3 KISS FWcaattgaccatgaataacatgtcttttgctgcgcccatgggttc
Gt-1 ORF3 KISS RVgcggccgcgcggcgcggccccagctg
Gt-1 ORF4 MAPPIT FWgtcgacgagctccggatccatgttgcgcggacagcaaatc
Gt-1 ORF4 MAPPIT RVgcggccgcttagctcacatacatccgcagggcag
Gt-1 ORF4 KISS FWcaattgaccatgttgcgcggacagcaaatc
Gt-1 ORF4 KISS RVgcggccgcgctcacatacatccgcagggcag
Gt-3 ORF2 MAPPIT FWgtcgacgagctccggatccaccatgtgccctagggttg
Gt-3 ORF2 MAPPIT RVgcggccgcttaagactcccgggttttgcctacc
Gt-3 ORF2 KISS FWcaattgaccatgtgccctagggttgttc
Gt-3 ORF2 KISS RVgcggccgcagactcccgggttttgcctacc
Gt-3 ORF3 MAPPIT FWgtcgacgagctccggatccaccatgggatcaccatgtgccctagg
Gt-3 ORF3 MAPPIT RVgcggccgctcaacggcgcagccccagc
Gt-3 ORF3 KISS FWcaattgaccatgggatcaccatgtgccctagggttg
Gt-3 ORF3 KISS RVgcggccgcacggcgcagccccagctg
Restriction sites are underlined.
Table 2. Sequences of oligos used for annealing and cloning into pX548.
Table 2. Sequences of oligos used for annealing and cloning into pX548.
Gene TargetedOligo NameSequence (5′ → 3′)
SHARPINSHARPIN_ FWCACCGCCTAGTCCGAGGTGCCACCG
SHARPIN_ RVAAACCGGTGGCACCTCGGACTAGGC
RNF5RNF5_ FWCACCGAAGCCCCCGGTATCACCAAA
RNF5_ RVAAACTTTGGTGATACCGGGGGCTTC
Table 3. List of antibodies used for Western blot.
Table 3. List of antibodies used for Western blot.
AntibodyReference
Primary antibodiesSHARPINMAB8100, R&D systems, Mineapolis, MN, USA
RNF5PA5-31793, Invitrogen, Waltham, MA, USA
ORF3Bs-0212R, Bioss, Woburn, MA, USA
β-ACTINBA3R, Invitrogen, Waltham, MA, USA
Secondary antibodiesSheep anti-mouse HRP-linkedNA931, Cytiva, Amersham, UK
Donkey anti-rabbit HRP-linkedNA934, Cytiva, Amersham, UK
Table 4. List of predesigned primer/probe pairs used for RT-qPCR.
Table 4. List of predesigned primer/probe pairs used for RT-qPCR.
Gene NameAssay ID
IFNB1Hs01077958_s1
IFNL2/3Hs04193049_gH
HPRT1Hs99999909_m1
RPL30Hs00265497_m1
CYC1Hs00357717_m1
YWHAZHs01122445_g1
ATP5BHs00969569_m1
Table 5. Overview of the interaction of SHARPIN and RNF5 with gt-1 and gt-3 ORF3 in MAPPIT and KISS.
Table 5. Overview of the interaction of SHARPIN and RNF5 with gt-1 and gt-3 ORF3 in MAPPIT and KISS.
ConstructEntrez_IDValue PIBValue BPBP/PIBBP/BIPLowest Value (Min BP/PIB and BP/BIP)SCORE
Gt-1 ORF3 MAPPITSHARPIN1.1782.0969.961.261.2+
Gt-1 ORF3 KISSSHARPIN1857796,44442942.242.2+
Gt-3 ORF3 KISSSHARPIN1857187,953101.225.425.4+
Gt-1 ORF3 KISSRNF53114250,91780.633.833.8+
Gt-3 ORF3 KISSRNF53114365,420117.419.419.4+
Results are compiled from the luciferase reporter read-out. The positive score of an interaction is related to the lowest value of BP/PIB and BP/BIP. In MAPPIT, a fold induction of a specific bait–prey interaction (BP) is calculated by dividing the average of EPO stimulated and non-stimulated controls. The obtained value of the specific BP is then compared to a fold induction of a bait with irrelevant prey interaction (BIP) and to a fold induction of an irrelevant bait and prey interaction (PIB). The BIP in this case was 1.34. To score an interaction as positive, the obtained lowest value of PB/BIP and PB/PIB needs to be above a threshold of 9. In KISS, stimulation is not possible and the average value of the bait–prey interaction is divided by the average of the BIP or PIB and needs to be above a threshold of 5. BIP in this case was 7414. The chosen thresholds depend on positive and negative reference sets and in this case relate to a false positivity rate of 2%.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Corneillie, L.; Lemmens, I.; Weening, K.; De Meyer, A.; Van Houtte, F.; Tavernier, J.; Meuleman, P. Virus–Host Protein Interaction Network of the Hepatitis E Virus ORF2-4 by Mammalian Two-Hybrid Assays. Viruses 2023, 15, 2412. https://doi.org/10.3390/v15122412

AMA Style

Corneillie L, Lemmens I, Weening K, De Meyer A, Van Houtte F, Tavernier J, Meuleman P. Virus–Host Protein Interaction Network of the Hepatitis E Virus ORF2-4 by Mammalian Two-Hybrid Assays. Viruses. 2023; 15(12):2412. https://doi.org/10.3390/v15122412

Chicago/Turabian Style

Corneillie, Laura, Irma Lemmens, Karin Weening, Amse De Meyer, Freya Van Houtte, Jan Tavernier, and Philip Meuleman. 2023. "Virus–Host Protein Interaction Network of the Hepatitis E Virus ORF2-4 by Mammalian Two-Hybrid Assays" Viruses 15, no. 12: 2412. https://doi.org/10.3390/v15122412

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop