INTRODUCTION
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) belongs to the
Coronaviridae family of coronaviruses (CoVs) in the order
Nidovirales. The viruses are enveloped, single-stranded viruses with a positive-sense RNA genome ~30 kb in length that cause respiratory, hepatic, enteric, and neurological diseases in diverse avian species and a wide range of mammals, including humans (
1). CoVs currently in current or recent circulation in human populations include human coronavirus-229E (HCoV-229E), HCoV-OC43, HCoV-HKU1, and HCoV-NL63 and the two emerging CoV strains, severe acute respiratory syndrome coronavirus (SARS-CoV) and Middle East respiratory syndrome coronavirus (MERS-CoV) (
2,
3). SARS-CoV-2, the most recently identified HCoV, resulted from the zoonotic transfer from an animal reservoir, resulting in a worldwide pandemic at considerable cost to human health and economic stability (
4).
CoVs, like other viruses, are subject to the antiviral effects of the innate immune response. The response consists of a large number of interferon-stimulated genes (ISGs) that interfere with virus replication through a variety of mechanisms (
5 - 9). While the viruses encode proteins that dampen interferon (IFN) responses and evade various innate immune mechanisms, it is clear that the innate immune response plays an important role in limiting virus replication and disease pathogenesis, as demonstrated by the increased pathogenicity in patients with mutations in the pathway (
10 - 13). CoVs are subjected to restriction from several ISGs. The zinc finger antiviral protein (ZAP) restricts SARS-CoV-2 replication by targeting CpG dinucleotides in viral RNA sequences (
14). IFIT1, IFIT3, and IFIT5 with tetratricopeptide repeats inhibit viral protein translation by targeting the 2′-O methyltransferase activity, reducing the proliferation and virulence of SARS-CoV and SARS-CoV-2 (
15 - 17). Viperin, an antiviral protein that restricts a broad range of viruses, inhibits the replication of porcine epidemic diarrhea virus, a member of CoV family, by interacting with the viral nucleocapsid (N) protein (
18). The bone marrow stromal cell antigen 2 (BST-2) restricts SARS-CoV-2 and HCoV-229E replication by tethering progeny virions to the cellular surface and intracellular membranes, preventing their release (
19 - 21). OAS1 was identified in genetic screens and population genetic studies, has been demonstrated to restrict SARS-CoV-2 infection, and plays a role in SARS-CoV-2 pathogenesis (
22 - 25). IFN-induced transmembrane proteins (IFITMs) have been shown to both inhibit and enhance the entry of HCoV-OC43 and SARS-CoV-2 (
26 - 30). Lymphocyte antigen 6E (LY6E) restricts CoVs block virus entry by interrupting fusion of the viral envelope with the cellular plasma membranes (
31 - 33). The IFNγ-inducible lysosomal thiolreductase (GILT) and CD74 have been found to suppress the entry of SARS-CoV-1 into lysosomes (
22,
34).
The receptor transporter protein 4 (RTP4) encoded by black flying fox bats, as well as several other bat species, has been shown to be a potent IFN-inducible inhibitor of the replication of Zika virus, West Nile virus, hepatitis C virus, yellow fever virus, and dengue flaviviruses (
35,
36). RTP4 orthologs of their natural host were less potent against particular flaviviruses than from nonnatural host species suggesting that the viruses have evolved to escape restriction (
35,
36). In humans, RTP4 belongs to a gene family consisting of four members (RTP1, RTP2, RTP3, and RTP4) that serve as chaperones that facilitate the transport of G protein-coupled receptors to the plasma membrane (
37 - 39). RTP1 and RTP2 are specifically expressed on olfactory neurons where they are involved in the functional expression of odorant receptors (
39 - 42), while RTP3 and RTP4 colocalize with bitter taste receptors (
43) and serve to regulate opioid and taste receptors (
43). Human RTP4 (hRTP4) is a widely expressed (
7,
44,
45) 247 amino acid protein consisting of an amino-terminal (N-terminal) zinc finger domain (ZFD), an intrinsic disordered variable region and a single carboxy-terminal (C-terminal) transmembrane domain (TM). In humans, RTP4 is up-regulated in the endometrium and corpus luteum in early pregnancy (
46). Previous studies have shown that bat RTP4 is localized to the Golgi apparatus and the endoplasmic reticulum (
41). Bat RTP4 inhibits flavivirus replication by targeting viral double-stranded RNA (ds RNA), preventing genome amplification and virion production (
35).
To identify novel ISGs that restrict HCoV replication, we analyzed candidate IFN-inducible genes for their ability to interfere with HCoV-OC43 and SARS-CoV-2 replication. Of the genes that were tested, the most potent inhibitor was hRTP4. The protein prevented viral RNA synthesis and was active against the ancestral Wuhan SARS-CoV-2 as well as the Omicron variants. The protein associated with viral RNA to target the replication phase of viral infection, preventing viral RNA synthesis and the production of virion proteins. Like the bat protein, the N-terminal domain was required for antiviral activity, while the C-terminal was dispensable. RTP4 was induced in SARS-CoV-2-infected K1-hACE2 and K18-hACE2 transgenic mice, but the protein was inactive against the viruses. This study extends the antiviral activity of hRTP4 to a more general inhibitor of both flavivirus and coronavirus replication.
DISCUSSION
We show here that hRTP4 is a potent inhibitor of coronavirus replication that is active against SARS-CoV-2 and the related coronavirus HCoV-OC43. hRTP4 inhibited the synthesis of HCoV-OC43 viral RNAs resulting in a block to the production of viral proteins. The protein formed complexes with the viral RNA in infected cells and mutated proteins that failed to bind viral RNA did not inhibit virus replication. The N-terminal domain of the protein was required for antiviral activity, while the C-terminal transmembrane domain was dispensable, similar to what was found for flavivirus inhibition by the bat homolog paRTP4 (
35). Infection of K1-hACE2 and K18-hACE2 transgenic mice with SARS-CoV-2 induced the expression of mRTP4 yet the murine homolog did not restrict coronavirus replication. These findings extend the role of hRTP4 as a restriction factor that acts on of several classes of RNA viruses.
Boys et al. first identified RTP4 as an ISG with antiviral activity, showing that bat RTP4 was a potent inhibitor of flavivirus replication (
35). They suggested that RTP4 could be involved in a Red Queen conflict with flaviviruses, in which diversification of both hosts and viruses has yielded a complex pattern of antiviral specificity of mammalian RTP4 orthologs. In their study, hRTP4 inhibited the Entebbe bat virus replication, a member of flavivirus family but did not appreciably inhibit HCoV-OC43, a finding that differs from ours. The explanation for this difference is not clear but may have been the result of the relatively low level at which the human homolog was expressed in transfected cells (6% that of the bat RTP4). In other studies, hRTP4 was also found to moderately inhibit the replication of norovirus in HG23 cells (
53) and yellow fever virus in STAT1-deficient human fibroblasts (
7).
Although the murine protein lacked activity against the coronaviruses tested, its expression in mice was induced by SARS-CoV-2 infection, most likely a result of type-I IFN which is present at high levels in SARS-CoV-2 infected mice. The IFN-inducibility of mRTP4 suggests that it has an antiviral role, although the viruses that it targets are not yet determined. Several reports have implicated RTP4 as playing a role in the innate immune response to virus infection (
54). The protein was found to be induced in the brains of mice infected with chikungunya and Newcastle disease virus (
55,
56). The induction was dependent upon toll-like receptors and the adaptor proteins myeloid differentiation primary response 88 (MyD88) and toll/interleukin-1 receptor domain-containing adaptor inducing IFN-β (TRIF) (
55). RTP4 knock-out mice were found to support increased levels of West Nile virus (
57). The other RTP family members, RTP1, RTP2, and RTP3, could not be tested for antiviral activity as they were unstable in transfected cells. Their genes were not IFN-inducible and thus not likely to have antiviral function.
In CHME3 cells, hRTP4 decreased the viral genomic and subgenomic HCoV-OC43 RNAs by about 1,000-fold and resulted in the absence of the viral N protein in infected cells. We found that hRTP4 was associated with the viral RNA and the association was mediated by the N-terminal ZFD. These results are consistent with those of Boys et al. who found that bat RTP4 binds the flavivirus dsRNA viral replication intermediate, disrupting the viral replicase complex (
35). The association was found to prevent the association of the viral polymerase NS5 and viral helicase NS3, altering binding of the viral polymerase to viral RNA in the replication complex (
35). Whether RTP4 has specificity for binding to viral RNAs is not clear.
The findings reported here extend the antiviral activity of RTP4 to another positive-stranded RNA virus family. Its potent activity against SARS-CoV-2 and induction upon infection suggests that it is an important restriction factor that could play a role in disease pathogenesis. Given the profound differences in pathogenicity in COVID-19 severity, the protein could be an important factor contributing to these differences. It does not appear that the virus has a means to avoid the antiviral effects of RTP4. It will be of interest to measure levels of the protein in the cells of SARS-CoV-2-infected individuals and to understand the mechanism by which the protein acts with such broad antiviral activity.
MATERIALS AND METHODS
Mice
K1-hACE2 [B6.129S2(Cg)ACE2tm1(ACE2)Dwnt/J] and K18-hACE2 [B6.Cg-Tg(K18-ACE2)2Prlmn/J] transgenic mice were purchased from The Jackson Laboratory and bred in-house. Animal experiments were done under the protocol approved by the NYU Langone Institutional Animal Care and Use Committee (#170304) according to the standards set by the Animal Welfare Act.
Virus stock preparation
SARS-CoV-2 WA1/2020 P1 virus stock (BEI Resources, NR-52281) was grown on Vero E6 cells by infection at an MOI of 0.01. At 2 h post-infection, input virus was removed and fresh medium was added. After 3 days, the virus-containing supernatant was harvested, filtered through a 0.45 µm filter, and frozen in aliquots at −80°C. Virus titers were determined by plaque assay on Vero E6 cells. The P1 stocks of Omicron BA.1 and BA.2 (BEI Resources, NR-56781) were generated by inoculating ACE2.TMPRSS2.Vero E6 cells at an MOI of 0.1. The P1 stock was expanded by a second round of replication on ACE2.TMPRSS2.Vero E6 cells infected at an MOI of 0.01. After 2 days, the supernatant was collected, filtered, and frozen at −80°C.
MRC5 cells were seeded at a density of 2 × 106 /mL in 100 mm dishes. The following day, the cells were infected with 3 × 106 PFU/mL of HCoV-OC43 (ATCC strain VR-1558) and incubated at 33°C for 4 days at which time 90–100% cells showed a cytopathic effect. The culture supernatant and infected cells were harvested, centrifuged at 1000 × g for 5 minutes, and the supernatant was stored at −80°C. The virus was titered by limiting dilution on MRC5 cells. Infection was scored by cytopathic effect 7 dpi in quadruplicate wells and expressed TCID50/mL defined as the dose at which two out of four quadruplicate wells exhibited a cytopathic effect.
Lentiviral vector stock for ACE2 expressing lentiviral vector pLenti.ACE2 was prepared by calcium-phosphate transfection of 293T cells with pLenti.ACE2 (
58) and expression plasmids pRSV-Rev, pMDL-X, and VSV-G.
Mouse infections
Six- to eight-week-old K1-hACE2 and K18-hACE2 transgenic mice were anesthetized with by intraperitoneal injection of ketamine and xylazine and infected intranasally with 2 × 104 PFU SARS-CoV-2 WA1/2020 or the same volume of PBS. At 2 dpi, the mice were killed and the lungs harvested and homogenized in Lysing Matrix D Tubes (MP Biomedicals, Irvine, California, USA) with a FastPrep-24 5G homogenizer (MP Biomedicals). The homogenates were clarified by brief centrifugation and RNA was prepared using a Quick-RNA MiniPrep kit (Zymo Research, Irvine, California, USA).
Cells
MRC5, CHME3, 293T, and Vero E6 cells were cultured in Dulbecco’s modified Eagle medium (DMEM) supplemented with 10% fetal bovine serum (FBS) and 1% penicillin/streptomycin and incubated at 37°C under 5% CO
2. To prepare primary human monocytes, PBMCs were purified by Ficoll density gradient centrifugation from Leukopaks provided by the New York Blood Center. Monocytes were purified by plastic adherence and cultured in RPMI containing 10 mM HEPES, 24 µg/mL gentamicin, and 5% heat inactivated pooled human serum. THP-1 cells were cultured in RPMI medium supplemented with 10% FBS and 1% penicillin/streptomycin and incubated at 37°C under 5% CO
2. To establish ACE2 expressing CHME3 stable cell lines, CHME3 cells were transduced with pLenti.ACE2 vector stock and after 2 days, cloned at limiting dilution in medium containing 1 µg/mL
puromycin. The cell clones were stained with anti-ACE2 antibody (NOVUS) and Alexa Fluor 594-conjugated goat anti-mouse IgG antibody (BioLegend, Eugene, Oregon, USA) and screened by flow cytometry to choose a clone that expressed high levels of cell surface ACE2. The data were analyzed with FlowJo software. To establish CHME3 ISG stable cell lines, CHME3 cells were transduced with corresponding ISG lentiviral vector stock and then selected in medium containing 1 µg/mL puromycin and screened by flow cytometry.
Plasmid construction
hRTP4 expressing lentiviral vector was generated by amplifying an hRTP4 cDNA with a forward primer containing a Spe-I restriction enzyme site and reverse primer containing 2X FLAG tag and Sal-I restriction enzyme site. The amplicon was cleaved with Spe-I and Sal-I and cloned into pLenti6.3/V5-DEST-GFP in place of GFP. hRTP4 TM domain truncation was generated using existing hRTP4 plasmid construct with a forward primer containing a Spe-I restriction enzyme site and reverse primer containing 2X FLAG tag and Sal-I restriction enzyme site. Mutations in the ZFDs were introduced by overlap extension PCR and mutations of the charged residues and the human to murine RTP4 mutants were introduced by quick change site-directed mutagenesis. Expression vectors for hRTP1, hRTP2, hRTP3, and mRTP4 were generated by amplifying the corresponding cDNA sequences (GenScript, Piscataway, New Jersey, USA) with a forward primer containing a Spe-I site and reverse primer containing 2X FLAG tag and Sal-I site and ligating to pLenti6.3/V5-DEST. The sequences of all plasmid constructs were confirmed by sequencing. The primer sequences of the constructs are shown in
Table S4.
HCoV-OC43 viral replication assay
CHME3 cell lines expressing different ISGs were seeded into black 96-well plates at 90% confluency. The next day, the cells were infected with HCoV-OC43 at an MOI of 0.01 and incubated at 33°C. At 48 h post-infection, the cells were fixed with 4% PFA for 20 minutes and permeabilized by treatment for 15 minutes in 0.05% Triton X-100/PBS. The cells were blocked for 1 h in 4% FBS and then stained overnight at 4°C with anti-OC43-N antibody (Sigma-Aldrich, Damstadt, Germany). The antibody was then removed by three washes with PBS, and the cells were stained for 1 h with Alexa Fluor 594 (Invitrogen, Eugene, Oregon, USA) and DAPI at room temperature. Images were analyzed using a Cell Insight CX7 LZR high-content screening platform with HCS Navigator software for DAPI and Alexa Fluor 647.
Western blotting
Transfected cells were lysed in buffer containing 10 mM Tris HCl, pH 7.5, 150 mM NaCl, 2 mM EDTA, 0.5% NP-40, and protease inhibitor cocktail and lysate protein concentrations were determined. The cell lysates (40 µg) were separated by SDS-PAGE, transferred to polyvinylidene difluoride membranes, and probed with mouse anti-FLAG antibody (Sigma-Aldrich), mouse anti-coronavirus OC43-N antibody (Sigma-Aldrich), rabbit anti-RTP4 antibody (Epigentek, Farmingdale, New York, USA), and anti-GAPDH antibody (Life Technologies, Carlsbad, California, USA) followed by goat anti-mouse HRP-conjugated second antibody (Sigma-Aldrich). The blots were washed and visualized using luminescent HRP substrate (Millipore, Burlington, MA, USA) on an iBright CL1000 imaging system.
RTP4: RNA complex pull-down
CHME3 cells were transiently transfected with hRTP4-2F expression vector by lipofection with Lipofectamine 2000 (Invitrogen) and then infected with HCoV-OC43 at an MOI of 5. After 2 days, the transfected cells were lysed in NP-40 lysis buffer (10 mM Tris HCl, pH 7.5, 150 mM NaCl, 2 mM EDTA, 0.5% NP-40, and protease inhibitor cocktail). The cell lysates (10 µg) were incubated for 1 h at 4°C with 20 µL anti-FLAG M2 magnetic beads (Sigma-Aldrich). The beads were pulled down in a magnetic separator and washed with buffer containing 50 mM Tris HCl, pH 7.5, and 150 mM NaCl. Bound protein complexes were eluted in buffer containing 0.1 M Glycine HCl, pH 3.0. The eluted proteins were analyzed on an immunoblot probed with mouse anti-FLAG antibody (Sigma-Aldrich) followed by goat anti-mouse HRP-conjugated second antibody (Sigma-Aldrich). RNA that had been pulled down with the RTP4 was isolated by phenol extraction followed by ethanol precipitation. Genomic and subgenomic HCoV-OC43 RNAs that have been pulled down were quantified by qRT-PCR.
Transfection of 293T cells and gene expression in vitro
293T cells were seeded at 2 × 105 cells/well in 6-well plate were transiently transfected with 4 µg of expression vectors along with the empty vector control using Lipofectamine 2000 (Invitrogen). After 2 days, the transfected cells were lysed in NP-40 lysis buffer and the cell lysates were analyzed on an immunoblot probed with mouse anti-FLAG antibody (Sigma-Aldrich) and anti-GAPDH antibody (Life Technologies) followed by goat anti-mouse HRP-conjugated second antibody (Sigma-Aldrich).
HCoV-OC43 and SARS-CoV-2 infection in vitro
CHME3 cells (2 × 105) were transfected with RTP4 expression vectors using Lipofectamine 2000. One dpi, the cells were infected with HCoV-OC43 at an MOI of 0.5 and 5 and incubated at 33°C. The cells were lysed on days 1 and 3 and the lysates were analyzed on an immunoblot probed with mouse anti-coronavirus OC43-N antibody (Sigma-Aldrich) followed by goat anti-mouse HRP-conjugated second antibody (Sigma-Aldrich). The infectivity for experiments was quantified by qRT-PCR. All HCoV-OC43 infections were done at Biosafety Level 2 (BSL2). SARS-CoV-2 (MOI = 0.1), Omicron BA.1, and BA.2 (MOI = 0.05, 0.5) infections in a BSL3 facility according to institutional guidelines provided by the NYU Langone and Institutional Animal Care and Use Committee according to the standards set by the Animal Welfare Act.
Quantitative RT-PCR
RNA was isolated from virus-infected CHME3 cells using Quick-RNA MiniPrep kit (Zymo Research). qRT-PCR analysis was done with TaqMan Fast Virus 1-Step Master Mix using primers in Orf1ab to amplify genomic HCoV-OC43 RNA and primers in the N gene to amplify subgenomic HCoV-OC43 RNA. SARS-CoV-2 genomic and subgenomic RNA levels were determined in SARS-CoV-2-infected ACE2.CHME3 cells and lung. Primer sequences and probe are detailed in
Table S5. Relative RNA copy numbers were calculated by the comparative C
T method with GAPDH as the internal control.
Measurement of ISGs
RNA was prepared from 200 µL homogenized lung using the Quick-RNA MiniPrep kit (Zymo Research). cDNA was reverse-transcribed using Transcriptor RT (Roche, Mannheim, Germany) with random hexamers. qPCR was performed using PowerUp SYBR Green Master Mix (Applied Biosystems, Waltham, Massachusetts, USA) with a typical three-step PCR protocol. The PCR was set for 40 cycles of 95°C/15 s, 60°C/30 s, and 95°C/15 s. Signals were normalized to GAPDH, and quantification of relative gene expression was relative to untreated controls with comparative C
T method. Primer sequences are detailed in
Table S5.
Quantification and statistical analysis
Statistical analyses were determined using GraphPad Prism, and statistical significance was determined with the two-tailed, unpaired t-test or two-way ANOVA. All the experiments were performed in duplicates or triplicates. Confidence intervals are shown as the mean ± SD or SEM (ns, not significant; *P ≤ 0.05, **P ≤ 0.01, ***P ≤ 0.001, ****P ≤ 0.0001).