key: cord-321564-6950p8i9 authors: Wang, Shiu‐Mei; Huang, Kuo‐Jung; Wang, Chin‐Tien title: Severe acute respiratory syndrome coronavirus spike protein counteracts BST2‐mediated restriction of virus‐like particle release date: 2019-07-10 journal: J Med Virol DOI: 10.1002/jmv.25518 sha: doc_id: 321564 cord_uid: 6950p8i9 BST2/tetherin, an interferon‐inducible antiviral factor, can block the cellular release of various enveloped viruses. We previously reported that human coronavirus 229E (HCoV‐229E) infection can alleviate the BST2 tethering of HIV‐1 virions by downregulating cell surface BST2, suggesting that coronaviruses are capable of encoding anti‐BST2 factors. Here we report our new finding that severe acute respiratory syndrome coronavirus (SARS‐CoV) spike (S) glycoprotein, similar to Vpu, is capable of antagonizing the BST2 tethering of SARS‐CoV, HCoV‐229E, and HIV‐1 virus‐like particles via BST2 downregulation. However, unlike Vpu (which downmodulates BST2 by means of proteasomal and lysosomal degradation pathways), BST2 downregulation is apparently mediated by SARS‐CoV S through the lysosomal degradation pathway only. We found that SARS‐CoV S colocalized with both BST2 and reduced cell surface BST2, suggesting an association between SARS‐CoV S and BST2 that targets the lysosomal degradation pathway. According to one recent report, SARS‐CoV ORF7a antagonizes BST2 by interfering with BST2 glycosylation(1). Our data provide support for the proposal that SARS‐CoV and other enveloped viruses are capable of evolving supplementary anti‐BST2 factors in a manner that requires virus replication. Further experiments are required to determine whether the BST2‐mediated restriction of authentic SARS‐CoV virions is alleviated by the SARS‐CoV spike protein. Bone marrow stromal antigen 2 (BST2, also designated as CD317 or tetherin) is a type II integral membrane protein containing a cytoplasmic N-terminal region followed by a spanning transmembrane domain and a carboxy-terminal glycosyl-phosphatidylinositol (GPI) anchor. 1 BST2 is an interferon-inducible gene that functions as an innate defense system against virus infections. It has been described as a host restriction factor capable of impeding the release of several types of enveloped viruses, including retroviruses, 2-7 filoviruses, [8] [9] [10] arenaviruses, 11 influenza, 12 and the Sendai virus. 13 One research team has proposed that BST2 inhibits virus release by tethering nascent virions to cell surfaces via the N-terminal transmembrane domain and C-terminal GPI anchor. 14 Most BST2-restricted enveloped viruses bud directly from cell surfaces, but a small number of enveloped viruses (eg, herpesviruses) are subject to BST2-related restrictions even though their final envelopment entails membranes from TGN and/or endosomal compartments and egression via exocytosis. 15, 16 In a previous study we reported that the human coronavirus 229E (HCoV-229E), whose assembly and budding occurs at the ER-Golgi intermediate compartment and whose virions are released via vesicle exocytosis, [17] [18] [19] is also subject to BST2 inhibition. Results from electron microscopy analyses indicate the presence of HCoV-229E virions on cell surfaces or on the membranes of intracellular vesicles that tend to cluster with BST2. This suggests the BST2-triggered tethering of budding virions to vesicle membranes that remain on cell surfaces at the plasma membrane after exocytosis. 17 BST2 has been described as moderately restricting the release of the hepatitis C virus, whose assembly takes place in the ER and whose release from cells via secretory pathways occurs in a manner similar to that of coronaviruses. 20, 21 Combined, these data support the assumption that enveloped virus budding and release occurring at the plasma membrane or in an intracellular compartment is subject to BST2 blocking. BST2 is a component of innate immune response in the form of restricted enveloped virion release, and many viruses have evolved specific antagonists to counteract BST2 antiviral activity: HIV-1 Vpu, HIV-2 Env, simian immunodeficiency virus (SIV) Nef and Env, Ebola and Sendai virus GP, Kaposi's sarcoma-associated herpesvirus K5, and influenza virus neuraminidase are all capable of antagonizing BST2. [2] [3] [4] [5] 9, 12, 13, 15, 22 Since some of these anti-BST2 viral factors are viral envelope glycoproteins, there is speculation that SARS-CoV spike glycoprotein may possess the property to counteract the BST2 blocking of virus release. Our work is built in part on an earlier finding by another research team that the ORF7a accessory protein (encoded by SARS-CoV) inhibits the BST2 tethering of virions. 23 We also found that the SARS-CoV spike (S) protein is capable of downmodulating BST2, thus mitigating the BST2-mediated restriction of virus-like particle (VLP) release, and suggesting that SARS-CoV and other enveloped viruses are capable of evolving additional anti-BST2 factors. Mammalian expression vectors encoding SARS-CoV M, N, S, and E were provided by G. J. Nabel. 24 BST2 dimerization-defective mutant (BST2/ C3A) was a gift from Klaus Strebel. 25 Plasmid pTRE-HN, kindly provided by Volker Thiel, 26 served as a template to generate PCR product containing HCoV-229E nucldocapsid coding sequence, using a forward primer 5′-CGCAATCGATTCATGAAGGCAGTTGCT-3′ and a reverse supplemented with 10% fetal calf serum (Invitrogen). Confluent cells were trypsinized and seeded onto 10 cm dish plates 24 hours before transfection. For each construct, cells were transfected with 20 μg of plasmid DNA by the calcium phosphate precipitation method; 50 μm chloroquine was added to enhance transfection efficiency. Unless otherwise indicated, 10 μg of each plasmid was used for cotransfection. For HeLa transfection, plasmid DNA was mixed with GenCarrier (Epoch Biolabs) at a ratio of 1 μg to 1 μL; the transfection procedure was performed according to the manufacturer's protocols. Human coronavirus 229E (HCoV-229E) were propagated in HeLa or A549 cells as described previously. 17 detection, a mouse monoclonal antibody was used. 27,28 BST2 was probed with a human BST2 mouse antiserum (ab88523, Abcam) or a rabbit antiserum. 29 Vpu was detected with a rabbit antiserum. 30 The secondary antibody was a sheep antimouse or donkey antirabbit horseradish peroxidase-(HRP) conjugated antibody (Invitrogen). We also used HeLa cells (which constitutively express BST2) to assess the impact of endogenous BST2 on VLP yields and found that BST2 forms stable cysteine-linked dimers. Blocking BST2 dimerization by replacing cysteines C53, C63, and C91 with alanine in the BST2 ectodomain, in turn, blocks the BST2 inhibition of HIV-1 release, suggesting that such dimerization is required for virion release blocking. 25 To test whether BST2 dimerization is also required for N (panel B) , or NL4.3delVpu (panel C) plus a wild-type or mutant BST2 expression vector (BST2/C3A) containing alanine substitutions for three cysteine residues in the BST2 ectodomain. Cells and supernatants were harvested and subjected to western immunoblotting at 24 to 36 hours posttransfection. BST2, bone marrow stromal antigen 2; VLP, virus-like particle further suggesting that BST2 dimerization is required to inhibit coronavirus VLP release. As stated above, several viral membrane glycoproteins such as HIV-2 and SIV Env, as well as Ebola and Sendai virus GP proteins, exert counteractive effects on BST2. We, therefore, attempted to identify anti-BST2 activity associated with the SARS-CoV spike (S) protein. Since BST2 is known to restrict HIV-1 release in the absence of Vpu, we performed tests to determine whether SARS-CoV S counteracts BST2 and therefore supports HIV-1 release. As shown in the upper panel of Figure 4A (lane 2 vs. lanes 3 and 4) , the inhibitory effect of BST2 on NL4.3delVpu virion release decreased in the presence of either SARS-CoV S or Vpu in step with reduced BST2 expression ( Figure 4A, middle panel, lanes 2-4) , suggesting that SARS-CoV S is capable of promoting the release of HIV-1 virus particles from cells via BST2 downregulation. Additional experiments confirmed that SARS-CoV S, like Vpu, is capable of reducing BST2 expression in a dose-dependent manner (Figures 4B and C) . Combined, the data suggest that SARS-CoV S may counteract the BST2-mediated restriction of VLP release. After binding to BST2, Vpu moves BST2 toward lysosomal-and proteasomal-degradation pathways. 2,34-38 Our next task was to examine whether SARS-CoV S mediates BST2 degradation via the same or similar pathway. Transfectants were treated with either MG132 (a proteasome inhibitor) 39 or ammonium chloride (NH 4 Cl, a lysosome inhibitor). 40 We found that BST2 downregulation mediated by SARS-CoV S was not significantly affected when the proteasome function was inhibited, but was noticeably reduced following lysosome function inhibition ( Figure 5, lanes 5-7) . Consistent with previous reports, we observed that proteasome or lysosome function inhibition resulted in markedly reduced Vpu-mediated BST2 downregulation ( Figure 5, lanes 2-4) , suggesting that BST2 downregulation as mediated by SARS-CoV S largely occurs via the lysosomal degradation pathway. Since BST2 largely localizes at cell surfaces (where they tether virions to prevent their release), we tested whether SARS-CoV S antagonizes BST2 via surface BST2 downregulation. 293 cells were cotransfected with BST2 and SARS-CoV S expression vectors. We observed that BST2 and S colocalized in perinuclear areas, but BST2 signals were barely detectable on cell surfaces. Instead, BST2 largely localized in perinuclear areas regardless of whether or not it was coexpressed with S ( Figure 6A ). This is consistent with a previous report that unlike HeLa-endogenous BST2 (which localizes on cell surfaces as well as in the perinuclear area), exogenous BST2 predominantly localizes in perinuclear areas, with little distribution on cell surfaces. 41 Nevertheless, flow cytometry quantification suggests that BST2 cell surface expression is noticeably reduced in 293T cells following SARS-CoV S coexpression ( Figure 6C ). In the case of HeLa cells (which constitutively express BST2), we did found SARS-CoV S colocalized with BST2 in the plasma membrane ( Figure 6B ). BST2 green fluorescence intensity on HeLa cell surfaces decreased slightly following SARS-CoV S coexpression, suggesting that the capacity of SARS-CoV S to counteract the BST2-associated inhibition of virion release was due in part to cell surface BST2 downmodulation. BST2 is capable of inhibiting SARS-CoV and HCoV-229E VLP release ( Figure 1 ). Since the BST2 inhibition of HIV-1 release via virion tethering at cell surfaces is well documented, we used a Vpudeficient HIV-1 virus-producing vector (NL4.3delVpu) as a control in our experiments. As shown in Figure 2 , coronavirus VLPs (similar to those of HIV-1) were tethered to cell surfaces by BST2, and BST2 inhibited SARS-CoV and HCoV-229E VLP release in a BST2 dimerization-dependent manner, similar to HIV-1 ( Figure 3 ). Further, in the same manner, as Vpu, SARS-CoV S facilitated HIV-1 release via BST2 downregulation (Figure 4) . We determined that Vpu downmodulated BST2 via proteasomal and lysosomal degradation pathways and that the predominant lysosomal pathway was mediated by SARS-CoV S ( Figure 5 ). Our confocal microscopy observations suggest that SARS-CoV S colocalized with BST2 at HeLa cell surfaces ( Figure 6 ). SARS-CoV S likely binds to BST2, after which it serves as a target for lysosomal degradation. Vpu is capable of trapping BST2 intracellularly and preventing its recycling back into the plasma membrane. 29, 35, 37, 42, 43 Whether SARS-CoV S similarly counteracts BST2 requires further investigation. The transmembrane protein SARS-CoV ORF7a has been shown to counteract BST2 tethering by interfering with BST2 glycosylation. 23, 44 In addition to the likelihood that BST2-associated restriction of F I G U R E 6 SARS-CoV glycoprotein S colocalizes with BST2. 293 cells (panel A) were cotransfected with BST2 and a SRAS-CoV expression vector. HeLa cells (panel B) were transfected with the SARS-CoV S expression vector. At 24 to 36 hours posttransfection, cells were probed with an anti-BST2 antibody before cell membrane permeabilization. SARS-CoV S was probed with an anti-S polyclonal antiserum. A rhodamineconjugated or FITC-conjugated antirabbit or antimouse antibody served as a secondary antibody. C, SARS-CoV S coexpression reduces BST2 cell surface expression. 293T cells were transfected with BST2 alone (middle panel) or together with a SARS-CoV S expression vector (bottom panel). At 24 to 36 hours posttransfection, cells were fixed and probed with a rabbit anti-BST2 antibody before the permeabilization of cell membranes, followed by a secondary FITC-conjugated antirabbit antibody. Cells then were analyzed by flow cytometry. BST2, bone marrow stromal antigen 2; SARS-CoV, severe acute respiratory syndrome coronavirus SARS-CoV virion release is mitigated by SARS-CoV S, it is possible that a number of enveloped viruses have developed supplementary anti-BST2 factors over time-note that in addition to Vpu, HIV-1 Nef is capable of overcoming BST2 restrictions on virus release under certain conditions. 45 SIV Nef 7 and Env 2 are capable of antagonizing BST2, and influenza neuraminidase 12 and M2 46 proteins both possess anti-BST2 capabilities. Some researchers have suggested that influenza and/or Ebola VLP release, but not virion release, is inhibited by BST2. 11, 47 Due to biosafety requirements, we are currently unable to perform tests to determine whether SARS-CoV S is capable of overcoming BST2 restrictions on SARS-CoV virion release. Bst-2/HM1.24 is a raft-associated apical membrane protein with an unusual topology Simian immunodeficiency virus envelope glycoprotein counteracts tetherin/BST-2/CD317 by intracellular sequestration Species-specific activity of SIV Nef and HIV-1 Vpu in overcoming restriction by tetherin/ BST2 Antagonism to and intracellular sequestration of human tetherin by the human immunodeficiency virus type 2 envelope glycoprotein Tetherin inhibits retrovirus release and is antagonized by HIV-1 Vpu The interferon-induced protein BST-2 restricts HIV-1 release and is downregulated from the cell surface by the viral Vpu protein Nef proteins from simian immunodeficiency viruses are tetherin antagonists Broad-spectrum inhibition of retroviral and filoviral particle release by tetherin Tetherinmediated restriction of filovirus budding is antagonized by the Ebola glycoprotein Dimerization of tetherin is not essential for its antiviral activity against Lassa and Marburg viruses Infectious Lassa virus, but not filoviruses, is restricted by BST-2/tetherin Influenza virus partially counteracts restriction imposed by tetherin/BST-2 Antagonism to human BST-2/tetherin by Sendai virus glycoproteins Tetherin inhibits HIV-1 release by directly tethering virions to cells Molecular mechanism of BST2/tetherin downregulation by K5/MIR2 of Kaposi's sarcomaassociated herpesvirus Herpesvirus assembly and egress BST2/CD317 counteracts human coronavirus 229E productive infection by tethering virions at the cell surface The molecular biology of coronaviruses Virus assembly Hepatitis C virus: assembly and release of virus particles Modulation of hepatitis C virus release by the interferoninduced protein BST-2/tetherin The RING-CH ligase K5 antagonizes restriction of KSHV and HIV-1 particle release by mediating ubiquitin-dependent endosomal degradation of tetherin Severe acute respiratory syndrome coronavirus ORF7a inhibits bone marrow stromal antigen 2 virion tethering through a novel mechanism of glycosylation interference Generation of synthetic severe acute respiratory syndrome coronavirus pseudoparticles: implications for assembly and vaccine production The formation of cysteinelinked dimers of BST-2/tetherin is important for inhibition of HIV-1 virus release but not for sensitivity to Vpu Selective replication of coronavirus genomes that express nucleocapsid protein Identifying epitopes responsible for neutralizing antibody and DC-SIGN binding on the spike glycoprotein of the severe acute respiratory syndrome coronavirus APOBEC3G cytidine deaminase association with coronavirus nucleocapsid protein Vpu enhances HIV-1 virus release in the absence of Bst-2 cell surface down-modulation and intracellular depletion Human immunodeficiency virus type 1 Vpu protein is an oligomeric type I integral membrane protein Self-assembly of severe acute respiratory syndrome coronavirus membrane protein SARS-CoV envelope protein palmitoylation or nucleocapid association is not required for promoting virus-like particle production An interferon-αinduced tethering mechanism inhibits HIV-1 and Ebola virus particle release but is counteracted by the HIV-1 Vpu protein. Cell Host Microbe Vpu directs the degradation of the human immunodeficiency virus restriction factor BST-2/Tetherin via a {beta}TrCP-dependent mechanism Vpu antagonizes BST-2-mediated restriction of HIV-1 release via beta-TrCP and endolysosomal trafficking HIV-1 antagonism of CD317 is species specific and involves Vpu-mediated proteasomal degradation of the restriction factor Antagonism of tetherin restriction of HIV-1 release by Vpu involves binding and sequestration of the restriction factor in a perinuclear compartment HIV-1 Vpu neutralizes the antiviral factor Tetherin/BST-2 by binding it and directing its beta-TrCP2-dependent degradation Differential inhibition of calpain and proteasome activities by peptidyl aldehydes of di-leucine and tri-leucine Ammonium chloride, an inhibitor of phagosomelysosome fusion in macrophages, concurrently induces phagosomeendosome fusion, and opens a novel pathway: studies of a pathogenic mycobacterium and a nonpathogenic yeast The tetherin/BST-2 coiled-coil ectodomain mediates plasma membrane microdomain localization and restriction of particle release Membrane anchoring by a C-terminal tryptophan enables HIV-1 Vpu to displace bone marrow stromal antigen 2 (BST2) from sites of viral assembly HIV-1 Vpu blocks recycling and biosynthetic transport of the intrinsic immunity factor CD317/tetherin to overcome the virion release restriction Structure and intracellular targeting of the SARS-coronavirus Orf7a accessory protein Tetherin antagonism by HIV-1 group M Nef proteins BST-2 restricts IAV release and is countered by the viral M2 protein Influenza virus is not restricted by tetherin whereas influenza VLP production is restricted by tetherin Severe acute respiratory syndrome coronavirus spike protein counteracts BST2-mediated restriction of virus-like particle release