Skip to main content
Erschienen in: Virology Journal 1/2016

Open Access 01.12.2016 | Research

HSV-2 glycoprotein gD targets the CC domain of tetherin and promotes tetherin degradation via lysosomal pathway

verfasst von: Yalan Liu, Mei Li, Di Zhang, Mudan Zhang, Qinxue Hu

Erschienen in: Virology Journal | Ausgabe 1/2016

Abstract

Background

HSV-2 is the major cause of genital herpes. We previously demonstrated that the host viral restriction factor tetherin restricts HSV-2 release and is antagonized by several HSV-2 glycoproteins. However, the mechanisms underlying HSV-2 glycoproteins mediated counteraction of tetherin remain unclear. In this study, we investigated whether tetherin restricts the cell-to-cell spread of HSV-2 and the mechanisms underlying HSV-2 gD mediated antagonism of tetherin.

Methods

Infectious center assays were used to test whether tetherin could affect cell-to-cell spread of HSV-2. Coimmunoprecipitation assays were performed to map the tetherin domains required for HSV-2 gD-mediated downregulation. Immunoflurence assays were performed to detect the accumulation of tetherin in lysosomes or proteasomes. All experiments were repeated for at least three times and the data were performed statistical analysis.

Results

1) Tetherin restricts cell-to-cell spread of HSV-2; 2) HSV-2 gD specifically interacts with the CC domain of tetherin; 3) HSV-2 gD promotes tetherin to the lysosomal degradation pathway.

Conclusions

Tetherin not only restricts HSV-2 release but also its cell-to-cell spread. In turn, HSV-2 gD targets the CC domain of tetherin and promotes its degradation in the lysosome. Findings in this study have increased our understanding of tetherin restriction and viral countermeasures.
Abkürzungen
CC
Coil-coiled
CT
Cytoplasmic tail
gD
Glycoprotein D
GPI
Glycophosphatidylinositol
GUD
Genital ulcer disease
HIV
Human immunodeficiency virus
HSV
Herpes simplex virus
MOI
Multiplicity of infection
TM
Transmembrane
WT
Wild-type

Background

Tetherin is an interferon (IFN)-inducible innate restriction factor involved in the host defense against the release of envelope viruses [1, 2]. Based on the deduced amino acid sequence, tetherin is characterized as a type II membrane protein with a Mr of approximately 24 kDa, while its mature form is a 30-36-kDa, heterogeneously glycosylated, dimeric, type II integral membrane protein, presumably due to heterogeneity of glycosylation during post-translational modification, comprising a small cytoplasmic tail (CT) domain, a trans-membrane (TM) domain, a glycophosphatidylinositol (GPI) membrane anchor at the C-terminus, and a long disulfide-rich coiled coil structure (CC) predicted for the extracellular domain [3, 4]. The mechanism by which different viruses counteract tetherin is distinct. To date, a number of viral proteins including Vpu and Env of HIV, Env and Nef of SIV, K5 of KSHV, Ebola GP, gM/VHS of HSV-1 and gB/gD/gH/gL of HSV-2 have been revealed to counteract the restrictive properties of tetherin [1, 512]. Except a recent report that Ebola GP may overcome tetherin restriction by blocking an interaction between VP40 and tetherin [13], the counteraction generally depends on the interaction between an viral antagonist and a specific domain of tetherin [14]. For instance, HIV-1 Vpu targets the TM domain of tetherin for the subsequent antagonism of tetherin function [1, 2, 1519]; HIV-2 and tantalus SIV (SIVtan) Env interacts with the ectodomain of tetherin [20, 21], while other SIVs employ the Nef protein to target the cytoplasmic domain of tetherin [6, 8, 22]. The mechanisms underlying HIV-1 counteracts tetherin have been intensively investigated, but less attention has been paid to understanding those mediated by other viruses.
Herpes simplex virus 2 (HSV-2) is a DNA virus sexually transmitted and causes persistent infection that cannot be eliminated [23]. HSV-2 is the leading cause of genital ulcer disease (GUD) throughout the world and associated with human immunodeficiency virus 1 (HIV-1) acquisition [2428]. HSV-2 transmission occurs via cell-free and direct cell-to-cell spread [29]. The HSV-2 glycoprotein D (gD), a major component of the virion envelope, was previously revealed to be essential for viral fusion and plays an important role in the cell-to-cell spread of the virus in permissive cells [30, 31].
Our previous study has revealed that tetherin functions as a restriction factor to inhibit HSV-2 release and that several glycoproteins including gD downregulate the expression of tetherin [11]. In this study, we investigated whether tetherin restricts the cell-to-cell spread of HSV-2 and the mechanisms underlying HSV-2 gD mediated antagonism of tetherin.

Results

Tetherin restricts cell-to-cell spread of HSV-2

Spread of HSV-2 progeny virus can occur both by release of mature infectious virus particles into the extracellular medium and by viral cell-to-cell spread. Having demonstrated that the release of cell-free HSV-2 progeny virions was restricted by tetherin [11], we further asked whether tetherin could affect cell-to-cell spread of HSV-2 by using infectious center assay. Firstly, the plaque formation on HeLa monolayer with or without anti-HSV-2 antibody treatment was tested. The area of plaques was relatively uniform in the presence of anti-HSV-2 antibody, while some small spots existed in the samples without anti-HSV-2 antibody (Fig. 1a and b). The following experiments were all performed under the anti-HSV-2 antibody treatment. Subsequently, the HeLa monolayers pretreated with tetherin or control siRNA were examined. Western blot analysis indicated that siRNA knockdown of tetherin efficiently reduced the expression of tetherin (Fig. 1c). As shown in Fig. 1d and e, compared to control siRNA-pretreated HeLa cells, the plaque size was modestly increased upon tetherin siRNA pretreatment. Accordingly, two polarized epithelial cell types (HaCaT and ARPE-19) [32, 33] were used in the infectious center assay. As shown in Fig. 1f and g, the plaque size was significantly reduced on the pBST2 transfected HaCaT and ARPE-19 monolayers, confirming an interference with the cell-to-cell spread of HSV-2 in the presence of tetherin. Taken together, the reduced size of HSV-2 plaques by tetherin informs that tetherin at least partially inhibits the direct cell-to-cell spread of HSV-2 during plaque formation.

HSV-2 gD specifically interacts with the CC domain of tetherin

The interaction between HSV gD expressed in the progeny virus producing cells and the neighbouring host cells is essential for cell-to-cell spread. We previously demonstrated that several glycoproteins of HSV-2, including gD, can downregulate the expression of tetherin. To map the tetherin domains required for HSV-2 gD-mediated downregulation, we constructed a panel of deletion mutants of tetherin (Fig. 2a) and performed coimmunoprecipitation assays using cells transiently expressing wild type tetherin or its mutants. The expression of tetherin mutants was confirmed by western blot using an anti-tetherin antibody (Fig. 2b). Subsequently, the precleared cell lysates from the transfected cells were incubated with an anti-flag antibody or an isotype control antibody. The precipitates were analyzed by western blot using the antibody against tetherin. The tetherin mutants delTM, delCT and delGPI but not delCC were specifically coimmunoprecipitated by the flag antibody (Fig. 2c, d, e and f, left panels). Co-IP experiments were also performed by pulling down with the antibody against tetherin followed by western blot with the flag antibody. The antibody against tetherin was able to specifically precipitate the immune complexes that contained gD-flag and delTM, delCT or delGPI (Fig. 2c, d, e and f, right panels), indicating that only the CC domain of tetherin is required for the physical interaction with HSV-2 gD. Taken together, these data suggest that tetherin is down-regulated through a specific interaction between HSV-2 gD and the CC domain of tetherin.

HSV-2 gD promotes tetherin to the lysosomal degradation pathway

The known antagonists of tetherin can lead the degradation of tetherin in late endosomes, lysosomes or proteasomes [34]. To examine whether the gD mediated-downregulation of tetherin undergoes lysosomal or proteasomal degradation pathway, HeLa cells were transfected with pcDNA3.1 or gD expressing plasmid followed by cultivation in the presence of a mixture of lysosome protease inhibitors (LPI; containing leupeptin, pepstatin A, and E64d) or the proteasome protease inhibitor (PPI; containing MG132). The paralleled HeLa cells transfected with gM expressing plasmid were included as the control. At 24 hours post transfection, cells were processed for immunofluorescence staining for subcellular localization of tetherin, gD/gM and a lysosome marker cathepsin D or a proteasome marker 20S proteasome. As shown in Fig. 3a and c, colocalization of gD, tetherin and cathepsin D was seen in gD expressing cells treated with lysosomal protease inhibitors, whereas tetherin and cathepsin D did not apparently colocalize in the pcDNA3.1- or gM-transfected cells. Tetherin colocalized with the 20S proteasome in the presence or absence of HSV-2 gD (Fig. 3b and d), implying that tetherin likely physiologically undergoes proteasomal degradation in the absence of HSV-2 gD. Western blot assay demonstrated that the lysosome protease inhibitors (LPI) rescued tetherin from degradation in the gD-transfected cells (Fig. 3e). To confirm that the gD mediated-downregulation of tetherin undergoes lysosomal degradation, we used a lysosome enrichment kit (Thermo) to isolate and enrich the intact lysosomes from cells transfected with pcDNA3.1 or plasmids expressing gD. The prepared cell extract was ultracentrifuged by density gradient centrifugation. We collected the corresponding band and detected the finally harvested lysosome pellets by western blot. As shown in Fig. 3f, the lysosome marker LAMP1, tetherin and gD all existed in the separated samples from gD-transfected cells. In contrast, only LAMP1 existed in the samples from pcDNA3.1 transfected cells. These data together indicate that the gD mediated-downregulation of tetherin undergoes lysosomal degradation.

Discussion

Tetherin has been shown to inhibit viral release and direct cell-to-cell spread of HIV-1 [3537]. However, whether tetherin restricts cell-to-cell spread of other viruses is less clear. We previously showed that tetherin restricts viral release of HSV-2. Here we demonstrate that tetherin restricts the cell-to-cell spread of HSV-2, suggesting that such restriction may be physiologically relevant for viruses that are capable of cell to cell transmission. Interestingly, HSV-1 can use heparan sulfate (HS) as an entry receptor for infection of host cells [38]. However, during a productive infection, the HS moieties on parent cells can trap newly exiting viral progenies and inhibit their release and cell to cell spread. It has been revealed that heparanase (HPSE), a HS-degrading enzyme of the host, can translocate to the cell surface upon HSV-1 infection, leading to the removal of HS to facilitate viral release [39]. In agreement, potent HPSE inhibitor can successfully decrease cell-to-cell spread of HSV-1 and HSV-2 [40]. Although beyond the scope of this current study, it will be interesting to determine whether there is a similar enzyme or mechanism which can degrade tetherin to aid virus egress.
To counteract host restriction, viruses have evolved various evasion strategies. For instance, although tetherin efficiently inhibits the release of a number of enveloped virus particles by “tethering” them to the cell surface, different viral antagonists employ different mechanisms to counteract the restriction by tetherin. Current understanding how viruses counteract the restrictive property of tetherin is largely limited to the findings from retroviruses. In general, the counteraction is dependent of the interaction between viral antagonists and different domains of tetherin [14]. In the current study, we demonstrated that HSV-2 gD specifically interacts with the CC domain of tetherin.
There are several proposed mechanisms for the final fate of tetherin, including lysosomal degradation, proteasomal degradation, and/or sequestration/retargeting of tetherin to the trans-Golgi network [16, 21, 4144]. Our previous study demonstrated that the levels of cell surface and total cellular tetherin were decreased in the presence of HSV-2 glycoproteins, implying that tetherin is likely counteracted by viral glycoproteins via a degradation pathway. In the current study, immunofluorescence images revealed that tetherin colocalized with the lysosome marker cathepsin D in the presence of HSV-2 gD, and western blot assay showed that gD and tetherin exist in the isolated lysosomes of gD-transfected cells, suggesting that gD promotes the degradation of tetherin in lysosomal pathway. Surprisingly, we found that the colocalization of tetherin and proteasome marker 20S proteasome was independent of the presence of gD, suggesting that tetherin itself, at least in HeLa cells, may physiologically undergo proteasomal degradation in the absence of HSV-2 gD. Although we cannot exclude other possibility, one possible explanation is that the proteasomal degradation may be cell-type dependent. Conjugation of proteins to ubiquitin is the hallmark of 26S proteasome mediated degradation, which is composed of a central 20S core containing catalytic subunits. The colocalization of tetherin and 20S proteasome implies the involvement of ubiquitinylation. Our data together demonstrated that tetherin indeed undergoes the lysosomal degradation in the presence of HSV-2 gD.

Conclusions

We demonstrate that tetherin not only inhibits HSV-2 release but also restricts its cell-to-cell spread. In turn, HSV-2 gD targets the CC domain of tetherin and promotes its degradation in the lysosome. Findings in this study have increased our understanding of tetherin restriction and viral countermeasures.

Methods

Virus and cells

Human cervical epithelial cell line HeLa, human keratinocyte cell line HaCaT, African green monkey kidney cell line Vero and embryonic kidney cell line 293 T were grown in Dulbecco’s modified Eagle’s medium (DMEM; Gibco) containing 10 % FBS, 100 Units/ml penicillin and 100 Units/ml streptomycin at 37 °C in 5 % CO2. Human retinal pigment epithelial cell line ARPE-19 was grown in DMEM/F-12 medium (50:50) supplemented with 10 % FBS, 100 Units/ml penicillin and 100 Units/ml streptomycin at 37 °C in 5 % CO2. HSV-2 (G strain) was obtained from LGC standards and propagated in Vero cells. Virus stock was stored at −80 °C before used for infection.

Plasmids

The open reading frames (ORF) of human tetherin amplified from pBST2 (Origene) was subcloned into pcDNA3.1(−) to construct the plasmid pWT (expressing full-length human tetherin). A panel of deletion mutants of tetherin were made and subcloned into pcDNA3.1(−), named delCT expressing 20–180 aa of human tetherin, delTM expressing 45–180 aa of human tetherin, delCC expressing human tetherin with the deleption of 100–154 aa and delGPI expressing 1–161 aa of human tetherin, respectively. The delTM without secretory leader peptide is not efficient for tetherin mutant to anchor on the cell surface membrane, we therefore fused the secretory leader peptide MDAMKRGLCCVLLLCGAVFVSPSQE from tissue plasminogen activator to tetherin codons 45–180 as described previously [45]. The glycoprotein gD and gM fused to flag tag were cloned into pcDNA3.1 (+) (named pgD-flag and pgM-flag) as described previously. The construction, cloning and propagation of plasmids were carried out using standard techniques. All constructs were verified by DNA sequencing (Sunny Biotechnology Co. Ltd, Shanghai, China).

siRNA mediated knockdown of tetherin

Tetherin siRNAs (SI02777054) and control siRNA (1027281) were purchased from Qiagen. HeLa cells were plated onto 6-well culture dishes overnight. Hela monolayers were transfected with tetherin siRNA or control siRNA using HiPerFect Transfection Reagent (301704; Qiagen) according to the manufacturer's instructions.

Infectious center assay

Infectious center assay was carried out as previously described [46]. In brief, HeLa, HaCaT and ARPE-19 cells plated in 6-well plates at 50 % confluence were exposed to HSV-2 at an MOI of 5 at 37 °C. After 90 min of incubation, cells were washed once with PBS and then treated with 0.1 M citrate buffer (pH 3.0) for 1 min to inactivate extracellular virus particles. The monolayer was then washed twice with PBS to remove the low-pH buffer, and cells were placed in growth medium supplemented with anti-HSV-2 antibody (PAB13979; Abnova) at a dilution of 1:1000 to neutralize extracellular HSV-2. After a total incubation of 5.5 h, the infected cells were detached with trypsin-EDTA, resuspended in growth medium, and ~100 cells were plated onto 50 % confluent monolayers of uninfected HaCaT and ARPE-19 cells transfected with pBST2 or pcDNA3.1, or of uninfected HeLa cells pretreated with tetherin or control siRNA. Cells were maintained in growth medium containing anti-HSV-2 polyclonal antibody. After 2 days, the cells were fixed and stained for HSV-2 antigens, followed by the addition of the peroxidase-conjugated secondary antibody and substrates. Plaques were photographed and the plaque areas were compared.

Co-immunoprecipitation

Co-immunoprecipitation assay was carried out using Pierce Crosslink Immunoprecipitation Kit (26147; Thermo scientific) according to the manufacturer's instructions. For co-immunoprecipitations of gD-flag and tetherin mutants, mouse monoclonal antibody against FLAG (F1804; Sigma) and rabbit polyclonal antibody to tetherin (11721; NIH AIDS Research and Reference Reagent Program) were used for pull-down experiments. These two antibodies were also used for western blot analysis of the immunoprecipitates and the lysates.

Western blot

Prepared cell lysates were resolved by 12 % SDS-PAGE and transferred to 0.45 μm polyvinylidene difluoride membranes (Millipore). Nonspecific binding was blocked using 5 % non-fat milk in PBS overnight at 4 °C. The membrane was incubated with primary antibody against tetherin (rabbit polyclonal antibody; 11721; NIH AIDS Research and Reference Reagent Program) at a dilution of 1:5000, FLAG (mouse monoclonal antibody; F1804; Sigma) at a dilution of 1:3000, and β-actin (mouse monoclonal antibody; sc-81178; Santa Cruz) at a dilution of 1:500, for 1 h at 37 °C. The membrane was washed five times with 0.1 % Tween 20/PBS, followed by incubation for 1 h with HRP conjugated goat anti–rabbit secondary antibody (1:10,000; BA1054, Boster) or HRP conjugated goat anti–mouse secondary antibody (1:10,000; BA1050, Boster). After five washes with 0.1 % Tween-20/PBS, the bands were visualized by exposure to FluorChem HD2 Imaging System (Alpha Innotech) after the addition of chemiluminescent substrate (SuperSignal® West Dura Extended Duration Substrate; 34075; Thermo Scientific Pierce).

Immunofluorescence

To detect the accumulation of tetherin in lysosomes or proteasomes, transfected cells were cultured in complete medium in the presence of a mixture of lysosome protease inhibitors (containing leupeptin, pepstatin A and E64d; Sigma) for 16 h or a proteasome inhibitor (MG132; Sigma) for 12 h as previously described [16]. Transfected cells on 35 mm glass bottom culture dishes were washed twice with phosphate-buffered saline (PBS), followed by fixation with 4 % (w/v) cold paraformaldehyde for 30 min at room temperature. Cells were permeabilized with PBST (PBS-0.2 % (v/v) Triton X-100) for 10 min at room temperature and then blocked with PBS-2 % (w/v) BSA for 1 h at room temperature. Cells were incubated for 1 h at 37 °C with mouse monoclonal antibody against FLAG (F1804; Sigma) at a dilution of 1:200 and followed by incubation for 1 h at 37 °C with Cy3-conjugated goat anti-mouse secondary antibody (Beyotime, China) at a dilution of 1:200 in PBS-2 % (w/v) BSA. Thereafter, cells were incubated for 1 h at 37 °C with mouse monoclonal antibody against tetherin (H00000684-M15, Abnova) at a dilution of 1:200 and rabbit polyclonal antibody against cathepsin D (sc-10725; Santa Cruz) (or 20S proteasome α7/α8 (sc-67344; Santa Cruz)) at a dilution of 1:100 in PBS-2 % (w/v) BSA, followed by incubation for 1 h at 37 °C with Cy5-conjugated goat anti-mouse secondary antibody (Beyotime, China), FITC-conjugated goat anti-rabbit secondary antibody (Beyotime, China) at a dilution of 1:200 in PBS-2 % (w/v) BSA. Cells were washed three times after each incubation with PBS and then twice with distilled water. Nuclei were dyed by Hoechst 33258 (Invitrogen). Stained cells were analyzed using confocal microscopy (PerkinElmer UltraView VoX).

Statistical analysis

All experiments were repeated for at least three times. Data are presented as mean ± SD unless otherwise specified. The difference of mean value was analyzed by a paired Student’s t-test. P < 0.05 was considered statistically significant.

Acknowledgements

This work was supported by National Natural Science Foundation of China Grants 31570165 (to Y.L.).

Funding

National Natural Science Foundation of China Grants (31570165).

Availability of data and materials

The authors agree to the terms of the BioMed Central Copyright and License Agreement and Open data policy and declare that the reproducible materials described in the manuscript freely available to any scientist wishing to use them.

Authors’ contributions

YL designed the study and performed the major experiments and statistical analysis, drafted and revised the manuscript. ML carried out the construction of plasmids and coIP assay. DZ and MZ participated in the statistical analysis and revised the manuscript. QH participated in the study design and manuscript drafting. All authors read and approved the final manuscript.

Competing interests

The authors declare that they have no competing interests.
This study does not contain any individual person’s data.
Not applicable.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Neil SJ, Zang T, Bieniasz PD. Tetherin inhibits retrovirus release and is antagonized by HIV-1 Vpu. Nature. 2008;451:425–30.CrossRefPubMed Neil SJ, Zang T, Bieniasz PD. Tetherin inhibits retrovirus release and is antagonized by HIV-1 Vpu. Nature. 2008;451:425–30.CrossRefPubMed
2.
Zurück zum Zitat Van Damme N, Goff D, Katsura C, Jorgenson RL, Mitchell R, Johnson MC, et al. The interferon-induced protein BST-2 restricts HIV-1 release and is downregulated from the cell surface by the viral Vpu protein. Cell Host Microbe. 2008;3:245–52.CrossRefPubMedPubMedCentral Van Damme N, Goff D, Katsura C, Jorgenson RL, Mitchell R, Johnson MC, et al. The interferon-induced protein BST-2 restricts HIV-1 release and is downregulated from the cell surface by the viral Vpu protein. Cell Host Microbe. 2008;3:245–52.CrossRefPubMedPubMedCentral
3.
Zurück zum Zitat Ohtomo T, Sugamata Y, Ozaki Y, Ono K, Yoshimura Y, Kawai S, et al. Molecular cloning and characterization of a surface antigen preferentially overexpressed on multiple myeloma cells. Biochem Biophys Res Commun. 1999;258:583–91.CrossRefPubMed Ohtomo T, Sugamata Y, Ozaki Y, Ono K, Yoshimura Y, Kawai S, et al. Molecular cloning and characterization of a surface antigen preferentially overexpressed on multiple myeloma cells. Biochem Biophys Res Commun. 1999;258:583–91.CrossRefPubMed
4.
Zurück zum Zitat Kupzig S, Korolchuk V, Rollason R, Sugden A, Wilde A, Banting G. Bst-2/HM1.24 is a raft-associated apical membrane protein with an unusual topology. Traffic. 2003;4:694–709.CrossRefPubMed Kupzig S, Korolchuk V, Rollason R, Sugden A, Wilde A, Banting G. Bst-2/HM1.24 is a raft-associated apical membrane protein with an unusual topology. Traffic. 2003;4:694–709.CrossRefPubMed
5.
Zurück zum Zitat Bartee E, McCormack A, Fruh K. Quantitative membrane proteomics reveals new cellular targets of viral immune modulators. PLoS Pathog. 2006;2:e107.CrossRefPubMedPubMedCentral Bartee E, McCormack A, Fruh K. Quantitative membrane proteomics reveals new cellular targets of viral immune modulators. PLoS Pathog. 2006;2:e107.CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat Jia B, Serra-Moreno R, Neidermyer W, Rahmberg A, Mackey J, Fofana IB, et al. Species-specific activity of SIV Nef and HIV-1 Vpu in overcoming restriction by tetherin/BST2. PLoS Pathog. 2009;5:e1000429.CrossRefPubMedPubMedCentral Jia B, Serra-Moreno R, Neidermyer W, Rahmberg A, Mackey J, Fofana IB, et al. Species-specific activity of SIV Nef and HIV-1 Vpu in overcoming restriction by tetherin/BST2. PLoS Pathog. 2009;5:e1000429.CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Mansouri M, Viswanathan K, Douglas JL, Hines J, Gustin J, Moses AV, et al. Molecular mechanism of BST2/tetherin downregulation by K5/MIR2 of Kaposi's sarcoma-associated herpesvirus. J Virol. 2009;83:9672–81.CrossRefPubMedPubMedCentral Mansouri M, Viswanathan K, Douglas JL, Hines J, Gustin J, Moses AV, et al. Molecular mechanism of BST2/tetherin downregulation by K5/MIR2 of Kaposi's sarcoma-associated herpesvirus. J Virol. 2009;83:9672–81.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Zhang F, Wilson SJ, Landford WC, Virgen B, Gregory D, Johnson MC, et al. Nef proteins from simian immunodeficiency viruses are tetherin antagonists. Cell Host Microbe. 2009;6:54–67.CrossRefPubMedPubMedCentral Zhang F, Wilson SJ, Landford WC, Virgen B, Gregory D, Johnson MC, et al. Nef proteins from simian immunodeficiency viruses are tetherin antagonists. Cell Host Microbe. 2009;6:54–67.CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Kaletsky RL, Francica JR, Agrawal-Gamse C, Bates P. Tetherin-mediated restriction of filovirus budding is antagonized by the Ebola glycoprotein. Proc Natl Acad Sci U S A. 2009;106:2886–91.CrossRefPubMedPubMedCentral Kaletsky RL, Francica JR, Agrawal-Gamse C, Bates P. Tetherin-mediated restriction of filovirus budding is antagonized by the Ebola glycoprotein. Proc Natl Acad Sci U S A. 2009;106:2886–91.CrossRefPubMedPubMedCentral
10.
Zurück zum Zitat Blondeau C, Pelchen-Matthews A, Mlcochova P, Marsh M, Milne RS, Towers GJ. Tetherin restricts herpes simplex virus 1 and is antagonized by glycoprotein M. J Virol. 2013;87:13124–33.CrossRefPubMedPubMedCentral Blondeau C, Pelchen-Matthews A, Mlcochova P, Marsh M, Milne RS, Towers GJ. Tetherin restricts herpes simplex virus 1 and is antagonized by glycoprotein M. J Virol. 2013;87:13124–33.CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Liu Y, Luo S, He S, Zhang M, Wang P, Li C, et al. Tetherin restricts HSV-2 release and is counteracted by multiple viral glycoproteins. Virology. 2015;475:96–109.CrossRefPubMed Liu Y, Luo S, He S, Zhang M, Wang P, Li C, et al. Tetherin restricts HSV-2 release and is counteracted by multiple viral glycoproteins. Virology. 2015;475:96–109.CrossRefPubMed
12.
Zurück zum Zitat Zenner HL, Mauricio R, Banting G, Crump CM. Herpes simplex virus 1 counteracts tetherin restriction via its virion host shutoff activity. J Virol. 2013;87:13115–23.CrossRefPubMedPubMedCentral Zenner HL, Mauricio R, Banting G, Crump CM. Herpes simplex virus 1 counteracts tetherin restriction via its virion host shutoff activity. J Virol. 2013;87:13115–23.CrossRefPubMedPubMedCentral
13.
Zurück zum Zitat Gustin JK, Bai Y, Moses AV, Douglas JL: Ebola Virus Glycoprotein Promotes Enhanced Viral Egress by Preventing Ebola VP40 From Associating With the Host Restriction Factor BST2/Tetherin. J Infect Dis 2015, 212 Suppl 2:S181–190. Gustin JK, Bai Y, Moses AV, Douglas JL: Ebola Virus Glycoprotein Promotes Enhanced Viral Egress by Preventing Ebola VP40 From Associating With the Host Restriction Factor BST2/Tetherin. J Infect Dis 2015, 212 Suppl 2:S181–190.
14.
Zurück zum Zitat Evans DT, Serra-Moreno R, Singh RK, Guatelli JC. BST-2/tetherin: a new component of the innate immune response to enveloped viruses. Trends Microbiol. 2010;18:388–96.CrossRefPubMedPubMedCentral Evans DT, Serra-Moreno R, Singh RK, Guatelli JC. BST-2/tetherin: a new component of the innate immune response to enveloped viruses. Trends Microbiol. 2010;18:388–96.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Dube M, Roy BB, Guiot-Guillain P, Binette J, Mercier J, Chiasson A, et al. Antagonism of tetherin restriction of HIV-1 release by Vpu involves binding and sequestration of the restriction factor in a perinuclear compartment. PLoS Pathog. 2010;6:e1000856.CrossRefPubMedPubMedCentral Dube M, Roy BB, Guiot-Guillain P, Binette J, Mercier J, Chiasson A, et al. Antagonism of tetherin restriction of HIV-1 release by Vpu involves binding and sequestration of the restriction factor in a perinuclear compartment. PLoS Pathog. 2010;6:e1000856.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Iwabu Y, Fujita H, Kinomoto M, Kaneko K, Ishizaka Y, Tanaka Y, et al. HIV-1 accessory protein Vpu internalizes cell-surface BST-2/tetherin through transmembrane interactions leading to lysosomes. J Biol Chem. 2009;284:35060–72.CrossRefPubMedPubMedCentral Iwabu Y, Fujita H, Kinomoto M, Kaneko K, Ishizaka Y, Tanaka Y, et al. HIV-1 accessory protein Vpu internalizes cell-surface BST-2/tetherin through transmembrane interactions leading to lysosomes. J Biol Chem. 2009;284:35060–72.CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat 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.CrossRefPubMedPubMedCentral 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.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Guo F, Liang C. Transmembrane interactions of HIV-1 Vpu and tetherin. Curr HIV Res. 2012;10:292–7.CrossRefPubMed Guo F, Liang C. Transmembrane interactions of HIV-1 Vpu and tetherin. Curr HIV Res. 2012;10:292–7.CrossRefPubMed
19.
Zurück zum Zitat Rollason R, Dunstan K, Billcliff PG, Bishop P, Gleeson P, Wise H, et al. Expression of HIV-1 Vpu leads to loss of the viral restriction factor CD317/Tetherin from lipid rafts and its enhanced lysosomal degradation. PLoS One. 2013;8:e75680.CrossRefPubMedPubMedCentral Rollason R, Dunstan K, Billcliff PG, Bishop P, Gleeson P, Wise H, et al. Expression of HIV-1 Vpu leads to loss of the viral restriction factor CD317/Tetherin from lipid rafts and its enhanced lysosomal degradation. PLoS One. 2013;8:e75680.CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Le Tortorec A, Neil SJ. Antagonism to and intracellular sequestration of human tetherin by the human immunodeficiency virus type 2 envelope glycoprotein. J Virol. 2009;83:11966–78.CrossRefPubMedPubMedCentral Le Tortorec A, Neil SJ. Antagonism to and intracellular sequestration of human tetherin by the human immunodeficiency virus type 2 envelope glycoprotein. J Virol. 2009;83:11966–78.CrossRefPubMedPubMedCentral
21.
Zurück zum Zitat Gupta RK, Mlcochova P, Pelchen-Matthews A, Petit SJ, Mattiuzzo G, Pillay D, et al. Simian immunodeficiency virus envelope glycoprotein counteracts tetherin/BST-2/CD317 by intracellular sequestration. Proc Natl Acad Sci U S A. 2009;106:20889–94.CrossRefPubMedPubMedCentral Gupta RK, Mlcochova P, Pelchen-Matthews A, Petit SJ, Mattiuzzo G, Pillay D, et al. Simian immunodeficiency virus envelope glycoprotein counteracts tetherin/BST-2/CD317 by intracellular sequestration. Proc Natl Acad Sci U S A. 2009;106:20889–94.CrossRefPubMedPubMedCentral
22.
Zurück zum Zitat Zhang F, Landford WN, Ng M, Mcnatt MW, Bieniasz PD, Hatziioannou T. SIV Nef proteins recruit the AP-2 complex to antagonize Tetherin and facilitate virion release. PLoS Pathog. 2011;7:e1002039.CrossRefPubMedPubMedCentral Zhang F, Landford WN, Ng M, Mcnatt MW, Bieniasz PD, Hatziioannou T. SIV Nef proteins recruit the AP-2 complex to antagonize Tetherin and facilitate virion release. PLoS Pathog. 2011;7:e1002039.CrossRefPubMedPubMedCentral
23.
Zurück zum Zitat Corey L, Adams HG, Brown ZA, Holmes KK. Genital herpes simplex virus infections: clinical manifestations, course, and complications. Ann Intern Med. 1983;98:958–72.CrossRefPubMed Corey L, Adams HG, Brown ZA, Holmes KK. Genital herpes simplex virus infections: clinical manifestations, course, and complications. Ann Intern Med. 1983;98:958–72.CrossRefPubMed
24.
Zurück zum Zitat Beyrer C, Jitwatcharanan K, Natpratan C, Kaewvichit R, Nelson KE, Chen CY, et al. Molecular methods for the diagnosis of genital ulcer disease in a sexually transmitted disease clinic population in northern Thailand: predominance of herpes simplex virus infection. J Infect Dis. 1998;178:243–6.CrossRefPubMed Beyrer C, Jitwatcharanan K, Natpratan C, Kaewvichit R, Nelson KE, Chen CY, et al. Molecular methods for the diagnosis of genital ulcer disease in a sexually transmitted disease clinic population in northern Thailand: predominance of herpes simplex virus infection. J Infect Dis. 1998;178:243–6.CrossRefPubMed
25.
Zurück zum Zitat Nilsen A, Kasubi MJ, Mohn SC, Mwakagile D, Langeland N, Haarr L. Herpes simplex virus infection and genital ulcer disease among patients with sexually transmitted infections in Dar es Salaam, Tanzania. Acta Derm Venereol. 2007;87:355–9.CrossRefPubMed Nilsen A, Kasubi MJ, Mohn SC, Mwakagile D, Langeland N, Haarr L. Herpes simplex virus infection and genital ulcer disease among patients with sexually transmitted infections in Dar es Salaam, Tanzania. Acta Derm Venereol. 2007;87:355–9.CrossRefPubMed
26.
Zurück zum Zitat Leichliter JS, Lewis DA, Sternberg M, Habel MA, Paz-Bailey G. Health care seeking among men with genital ulcer disease in South Africa: correlates and relationship to human immunodeficiency virus-1 and herpes simplex virus type 2 detection and shedding. Sex Transm Dis. 2011;38:865–70.CrossRefPubMed Leichliter JS, Lewis DA, Sternberg M, Habel MA, Paz-Bailey G. Health care seeking among men with genital ulcer disease in South Africa: correlates and relationship to human immunodeficiency virus-1 and herpes simplex virus type 2 detection and shedding. Sex Transm Dis. 2011;38:865–70.CrossRefPubMed
27.
Zurück zum Zitat Mehta SD, Moses S, Parker CB, Agot K, Maclean I, Bailey RC. Circumcision status and incident herpes simplex virus type 2 infection, genital ulcer disease, and HIV infection. AIDS. 2012;26:1141–9.CrossRefPubMedPubMedCentral Mehta SD, Moses S, Parker CB, Agot K, Maclean I, Bailey RC. Circumcision status and incident herpes simplex virus type 2 infection, genital ulcer disease, and HIV infection. AIDS. 2012;26:1141–9.CrossRefPubMedPubMedCentral
28.
Zurück zum Zitat Serwadda D, Gray RH, Sewankambo NK, Wabwire-Mangen F, Chen MZ, Quinn TC, et al. Human immunodeficiency virus acquisition associated with genital ulcer disease and herpes simplex virus type 2 infection: a nested case-control study in Rakai, Uganda. J Infect Dis. 2003;188:1492–7.CrossRefPubMed Serwadda D, Gray RH, Sewankambo NK, Wabwire-Mangen F, Chen MZ, Quinn TC, et al. Human immunodeficiency virus acquisition associated with genital ulcer disease and herpes simplex virus type 2 infection: a nested case-control study in Rakai, Uganda. J Infect Dis. 2003;188:1492–7.CrossRefPubMed
29.
Zurück zum Zitat Corey L, Spear PG. Infections with herpes simplex viruses (1). N Engl J Med. 1986;314:686–91.CrossRefPubMed Corey L, Spear PG. Infections with herpes simplex viruses (1). N Engl J Med. 1986;314:686–91.CrossRefPubMed
30.
Zurück zum Zitat Spear PG, Eisenberg RJ, Cohen GH. Three classes of cell surface receptors for alphaherpesvirus entry. Virology. 2000;275:1–8.CrossRefPubMed Spear PG, Eisenberg RJ, Cohen GH. Three classes of cell surface receptors for alphaherpesvirus entry. Virology. 2000;275:1–8.CrossRefPubMed
33.
Zurück zum Zitat Wisner T, Brunetti C, Dingwell K, Johnson DC. The extracellular domain of herpes simplex virus gE is sufficient for accumulation at cell junctions but not for cell-to-cell spread. J Virol. 2000;74:2278–87.CrossRefPubMedPubMedCentral Wisner T, Brunetti C, Dingwell K, Johnson DC. The extracellular domain of herpes simplex virus gE is sufficient for accumulation at cell junctions but not for cell-to-cell spread. J Virol. 2000;74:2278–87.CrossRefPubMedPubMedCentral
34.
Zurück zum Zitat Tokarev A, Skasko M, Fitzpatrick K, Guatelli J. Antiviral activity of the interferon-induced cellular protein BST-2/tetherin. AIDS Res Hum Retroviruses. 2009;25:1197–210.CrossRefPubMedPubMedCentral Tokarev A, Skasko M, Fitzpatrick K, Guatelli J. Antiviral activity of the interferon-induced cellular protein BST-2/tetherin. AIDS Res Hum Retroviruses. 2009;25:1197–210.CrossRefPubMedPubMedCentral
35.
Zurück zum Zitat Kuhl BD, Sloan RD, Donahue DA, Bar-Magen T, Liang C, Wainberg MA. Tetherin restricts direct cell-to-cell infection of HIV-1. Retrovirology. 2010;7:115.CrossRefPubMedPubMedCentral Kuhl BD, Sloan RD, Donahue DA, Bar-Magen T, Liang C, Wainberg MA. Tetherin restricts direct cell-to-cell infection of HIV-1. Retrovirology. 2010;7:115.CrossRefPubMedPubMedCentral
36.
Zurück zum Zitat Giese S, Marsh M. Tetherin can restrict cell-free and cell-cell transmission of HIV from primary macrophages to T cells. PLoS Pathog. 2014;10:e1004189.CrossRefPubMedPubMedCentral Giese S, Marsh M. Tetherin can restrict cell-free and cell-cell transmission of HIV from primary macrophages to T cells. PLoS Pathog. 2014;10:e1004189.CrossRefPubMedPubMedCentral
37.
Zurück zum Zitat Casartelli N, Sourisseau M, Feldmann J, Guivel-Benhassine F, Mallet A, Marcelin AG, et al. Tetherin restricts productive HIV-1 cell-to-cell transmission. PLoS Pathog. 2010;6:e1000955.CrossRefPubMedPubMedCentral Casartelli N, Sourisseau M, Feldmann J, Guivel-Benhassine F, Mallet A, Marcelin AG, et al. Tetherin restricts productive HIV-1 cell-to-cell transmission. PLoS Pathog. 2010;6:e1000955.CrossRefPubMedPubMedCentral
38.
39.
Zurück zum Zitat Hadigal SR, Agelidis AM, Karasneh GA, Antoine TE, Yakoub AM, Ramani VC, et al. Heparanase is a host enzyme required for herpes simplex virus-1 release from cells. Nat Commun. 2015;6:6985.CrossRefPubMedPubMedCentral Hadigal SR, Agelidis AM, Karasneh GA, Antoine TE, Yakoub AM, Ramani VC, et al. Heparanase is a host enzyme required for herpes simplex virus-1 release from cells. Nat Commun. 2015;6:6985.CrossRefPubMedPubMedCentral
40.
Zurück zum Zitat Nyberg K, Ekblad M, Bergstrom T, Freeman C, Parish CR, Ferro V, et al. The low molecular weight heparan sulfate-mimetic, PI-88, inhibits cell-to-cell spread of herpes simplex virus. Antiviral Res. 2004;63:15–24.CrossRefPubMed Nyberg K, Ekblad M, Bergstrom T, Freeman C, Parish CR, Ferro V, et al. The low molecular weight heparan sulfate-mimetic, PI-88, inhibits cell-to-cell spread of herpes simplex virus. Antiviral Res. 2004;63:15–24.CrossRefPubMed
41.
Zurück zum Zitat Janvier K, Roy N, Berlioz-Torrent C. Role of the endosomal ESCRT machinery in HIV-1 Vpu-induced down-regulation of BST2/tetherin. Curr HIV Res. 2012;10:315–20.CrossRefPubMed Janvier K, Roy N, Berlioz-Torrent C. Role of the endosomal ESCRT machinery in HIV-1 Vpu-induced down-regulation of BST2/tetherin. Curr HIV Res. 2012;10:315–20.CrossRefPubMed
42.
Zurück zum Zitat Goffinet C, Allespach I, Homann S, Tervo HM, Habermann A, Rupp D, et al. HIV-1 antagonism of CD317 is species specific and involves Vpu-mediated proteasomal degradation of the restriction factor. Cell Host Microbe. 2009;5:285–97.CrossRefPubMed Goffinet C, Allespach I, Homann S, Tervo HM, Habermann A, Rupp D, et al. HIV-1 antagonism of CD317 is species specific and involves Vpu-mediated proteasomal degradation of the restriction factor. Cell Host Microbe. 2009;5:285–97.CrossRefPubMed
43.
Zurück zum Zitat Mitchell RS, Katsura C, Skasko MA, Fitzpatrick K, Lau D, Ruiz A, et al. Vpu antagonizes BST-2-mediated restriction of HIV-1 release via beta-TrCP and endo-lysosomal trafficking. PLoS Pathog. 2009;5:e1000450.CrossRefPubMedPubMedCentral Mitchell RS, Katsura C, Skasko MA, Fitzpatrick K, Lau D, Ruiz A, et al. Vpu antagonizes BST-2-mediated restriction of HIV-1 release via beta-TrCP and endo-lysosomal trafficking. PLoS Pathog. 2009;5:e1000450.CrossRefPubMedPubMedCentral
44.
Zurück zum Zitat Skasko M, Tokarev A, Chen CC, Fischer WB, Pillai SK, Guatelli J. BST-2 is rapidly down-regulated from the cell surface by the HIV-1 protein Vpu: evidence for a post-ER mechanism of Vpu-action. Virology. 2011;411:65–77.CrossRefPubMedPubMedCentral Skasko M, Tokarev A, Chen CC, Fischer WB, Pillai SK, Guatelli J. BST-2 is rapidly down-regulated from the cell surface by the HIV-1 protein Vpu: evidence for a post-ER mechanism of Vpu-action. Virology. 2011;411:65–77.CrossRefPubMedPubMedCentral
45.
Zurück zum Zitat Hu S, Pang X, Li J, Cen S, Jin Q, Guo F. The role of the structural domains of human BST-2 in inhibiting the release of xenotropic murine leukemia virus-related virus. Biochem Biophys Res Commun. 2012;428:17–23.CrossRefPubMed Hu S, Pang X, Li J, Cen S, Jin Q, Guo F. The role of the structural domains of human BST-2 in inhibiting the release of xenotropic murine leukemia virus-related virus. Biochem Biophys Res Commun. 2012;428:17–23.CrossRefPubMed
46.
Zurück zum Zitat Jenssen H, Sandvik K, Andersen JH, Hancock RE, Gutteberg TJ. Inhibition of HSV cell-to-cell spread by lactoferrin and lactoferricin. Antiviral Res. 2008;79:192–8.CrossRefPubMed Jenssen H, Sandvik K, Andersen JH, Hancock RE, Gutteberg TJ. Inhibition of HSV cell-to-cell spread by lactoferrin and lactoferricin. Antiviral Res. 2008;79:192–8.CrossRefPubMed
Metadaten
Titel
HSV-2 glycoprotein gD targets the CC domain of tetherin and promotes tetherin degradation via lysosomal pathway
verfasst von
Yalan Liu
Mei Li
Di Zhang
Mudan Zhang
Qinxue Hu
Publikationsdatum
01.12.2016
Verlag
BioMed Central
Erschienen in
Virology Journal / Ausgabe 1/2016
Elektronische ISSN: 1743-422X
DOI
https://doi.org/10.1186/s12985-016-0610-7

Weitere Artikel der Ausgabe 1/2016

Virology Journal 1/2016 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Neu im Fachgebiet Innere Medizin

22.04.2024 | DGIM 2024 | Kongressbericht | Nachrichten

Krebspatienten impfen: Was? Wen? Und wann nicht?

22.04.2024 | DGIM 2024 | Kongressbericht | Nachrichten

Nierenultraschall: Tipps vom Profi

22.04.2024 | DGIM 2024 | Kongressbericht | Nachrichten

„KI sieht, was wir nicht sehen“

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.