Skip to main content
Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
J Virol. 2009 May; 83(9): 4574–4590.
Published online 2009 Feb 25. doi: 10.1128/JVI.01800-08
PMCID: PMC2668474
PMID: 19244337

Suppression of Tetherin-Restricting Activity upon Human Immunodeficiency Virus Type 1 Particle Release Correlates with Localization of Vpu in the trans-Golgi Network

Associated Data

Supplementary Materials

Abstract

Vpu promotes the efficient release of human immunodeficiency virus type 1 (HIV-1) by overcoming the activity of tetherin, a host cell restriction factor that retains assembled virions at the cell surface. In this study, we analyzed the intracellular localization and trafficking of subtype B Vpu in HIV-1-producing human cells. We found that mutations of conserved positively charged residues (R30 and K31) within the putative overlapping tyrosine- and dileucine-based sorting motifs of the Vpu hinge region affected both the accumulation of the protein in the trans-Golgi network (TGN) and its efficient delivery to late endosomal degradative compartments. A functional characterization of this mutant revealed that the mislocalization of Vpu from the TGN correlated with an attenuation of HIV-1 release. Interestingly, clathrin light chain small interfering RNA-directed disruption of Vpu trafficking from the TGN to the endosomal system slightly stimulated Vpu-mediated HIV-1 release and completely restored the activity of the Vpu R30A,K31A mutant. An analysis of the C-terminal deletion mutants of Vpu identified an additional determinant in the second helical structure of the protein, which regulated TGN retention/localization, and further revealed the functional importance of Vpu localization in the TGN. Finally, we show that a large fraction of Vpu colocalizes with tetherin in the TGN and provide evidence that the degree of Vpu colocalization with tetherin in the TGN is important for efficient HIV-1 release. Taken together, our results reveal that Vpu traffics between the TGN and the endosomal system and suggest that the proper distribution of Vpu in the TGN is critical to overcome the restricting activity of tetherin on HIV-1 release.

Human immunodeficiency virus type 1 (HIV-1) viral protein U (Vpu) is an oligomeric type 1 integral membrane protein that is associated with two major functions during HIV-1 infection (1, 16). First, it contributes to CD4 receptor downregulation by targeting newly synthesized CD4 molecules for degradation by the ubiquitin-proteasome system in the endoplasmic reticulum (ER) via a mechanism that most likely involves a dislocation step (2, 19). Second, Vpu expression enhances the release of HIV-1 particles and of widely divergent retroviruses, such as murine leukemia virus, in most human cells by a mechanism that does not rely on a specific interaction with Gag structural proteins (11, 13, 34, 35). Importantly, these Vpu biological activities play an active role in HIV-1 pathogenesis since Vpu-defective simian-HIV-1 strains were found to be less pathogenic in vivo (33).

Vpu enhances the release of retroviral particles by counteracting a human-specific host cell-dominant restriction to particle release in some human cell types (41). This host cell restriction was shown to consist of protein-based factors that caused the retention of fully formed virions on the surfaces of infected cells or in endosomes after endocytosis (21). Indeed, recent studies provided strong evidence that a membrane protein of unknown function called bone marrow stromal antigen-2 (BST-2; also known as CD317 or HMI 1.24), and now designated tetherin, is the host factor that mediates this restriction on HIV-1 and murine leukemia virus particles (22, 39). Accordingly, the effect of Vpu on HIV-1 release is prominent in many human cell types (termed restrictive or Vpu-responsive cells) constitutively expressing high or moderate levels of tetherin, including epithelial cell lines (HeLa), T-cell lines (Jurkat, CEM), and primary T lymphocytes, but is not observed in some human cell lines such as HEK 293, HOS, and HT1080 (termed permissive or Vpu-unresponsive cells) that do not express the restriction factor (22, 39). Indeed, these cells allow efficient viral particle release in the absence of Vpu, and the enhancement of virus production by Vpu is not observed.

The mechanism and the intracellular sites through which Vpu overcomes the antiviral activity of tetherin and enhances the release of HIV-1 particles are not currently precisely defined. Although Vpu appears to act on a host protein that exerts its restricting activity on HIV particle release at the cell surface, the most-studied subtype B Vpu was found to localize predominantly in the trans-Golgi network (TGN) and to a lesser extent in the ER and the recycling endosomes (23, 42). However, in contrast to the prototypical subtype B Vpu, the subtype C Vpu protein was found to localize both at the plasma membrane and in the Golgi complex (23). While ER localization is required for CD4 degradation, the artificial retention of Vpu in the ER appears to interfere with the enhancement of viral particle release, suggesting that Vpu needs to reach a post-ER compartment to mediate this function (32). In that regard, a previous study showed that the expression of dominant negative (DN) Rab11a or DN myosin Vb, which are known to disrupt protein trafficking through the recycling endosomes, can inhibit HIV-1 release in the presence of Vpu, suggesting that a functioning recycling endosome compartment is required (42). Moreover, disruption of the exit from recycling endosomes led to a marked trapping of Vpu in these structures, suggesting that Vpu normally traffics through recycling endosomes. Finally, recent studies that examined the subcellular distribution of Vpu relative to native tetherin have found that the two proteins colocalize within cytoplasmic structures that still remain to be characterized (22, 39). Furthermore, although Vpu had no overt effect on the overall levels of tetherin during conditions of efficient viral particle release (22), its expression downregulated tetherin from the cell surface (39).

In this study, we investigated the intracellular sites through which subtype B Vpu must traffic to counteract the activity of tetherin and perform its viral particle release enhancing function. Notably, we show that Vpu traffics between the TGN and the endosomal system. Furthermore, we provide evidence that the steady-state distribution of Vpu in the TGN is regulated by determinants within the hinge region of the protein as well as by sequences encompassing the second helical structure of the protein cytoplasmic domain. Additionally, we show that a large proportion of Vpu colocalizes with tetherin within the TGN. Importantly, we find that the proper distribution of Vpu in the TGN might represent a critical requirement to overcome the restricting activity of tetherin on HIV-1 particle release.

(This work was performed by M. Dubé in partial fulfillment of the requirements for his doctoral thesis from the University of Laval, Quebec, Ontario, Canada.)

MATERIALS AND METHODS

Antibodies and chemical compounds.

Brefeldin A (BFA), chloroquine (CQ), and rabbit polyclonal anti-actin antibodies were all obtained from Sigma. The anti-Vpu rabbit polyclonal serum was described previously (6). The following antibodies were obtained from commercial sources: mouse anti-CD63 (Hybridoma Bank [NICHD, University of Iowa]), mouse anti-lysosome-associated membrane protein-1 (LAMP1) (Santa Cruz Biotechnology), mouse anti-calreticulin (Santa Cruz Biotechnology), sheep anti-TGN46 (Serotec), mouse anti-Rab5 (BD Biosciences), mouse anti-BST-2 (Novus Biologicals), rabbit anti-clathrin light chains (CLCs) (Millipore), mouse anti-clathrin heavy chains (CHCs) (BD Biosciences), and mouse anti-cation-independent mannose-6-phosphate receptor (CI-MPR) (Abcam). The anti-CD4 (OKT4) and anti-p24 monoclonal antibodies were isolated from the ascitic fluids of BALB/c mice that were injected with the OKT4 or p24 hybridoma (American Type Culture Collection [ATCC], catalog no. CRL-8002 and HB9725, respectively). The human anti-HIV (#153) serum was obtained from an HIV-1-infected individual (15). Anti-green fluorescent protein (GFP) monoclonal antibody, transferrin-Alexa 488 conjugates, and all Alexa 488-, 594-, and 647-conjugated immunoglobulin G antibodies were obtained from Molecular Probes. All reagents were stored according to the manufacturer's instructions.

Cells, transfection, and siRNAs.

HeLa-TZM cells were obtained from the AIDS Research and Reference Program (NIH). HEK 293T and HeLa cells were obtained from ATCC. All cells were maintained as described previously (15). Unless specified, HEK 293T and HeLa cells were transfected by the calcium-phosphate method, and analyses were performed 48 h posttransfection. All specific small interfering RNAs (siRNAs) were synthesized by Dharmacon as 21-mers with UU overhangs. The siRNAs were transfected using Oligofectamine (Invitrogen) according to the manufacturer's instructions. Briefly, siRNAs were preincubated with 15 μl of Oligofectamine and overlaid on cells at 50% confluence. Both siRNAs specific to CLCa (GGAAAGUAAUGGUCCAACA) and CLCb (GGAACCAGCGCCAGAGUGA) were used for CLC depletion at a final concentration of 62.5 nM each. The siRNA specific to CHCs (GCAAUGAGCUGUUUGAAGA) or tetherin (Dharmacon Smartpool, catalogue no. L-011817) and the nontargeting siRNA Scrambled #2 from Dharmacon were used at a final concentration of 125 nM. The siRNA-transfected cells were then transfected with the appropriate proviral construct 72 h post-siRNA transfection using Lipofectamine 2000 (Invitrogen) and processed 24 h later. Empty plasmid DNA was added to each transfection to keep the amount of transfected DNA constant.

Plasmid constructs.

The mammalian expression plasmids SVCMV-vpu wt, SVCMV-vpu-, and SVCMV-CD4 have been described previously (2). HxBH10-vpu wt and HxBH10-vpu- are two isogenic infectious HIV-1 molecular clones that only differ in the expression of Vpu (35). HxBH10-vpu R30A,K31A, HxBH10-vpu Y29A, HxBH10-vpu I32A,L33A, SVCMV-vpu R30A,K31A, SVCMV-vpu I32A,L33A, and SVCMV-vpu Y29A were generated by PCR-based site-directed mutagenesis as described previously (38). The plasmids pEGFP-Rab7 wt, pEGFP-Rab7 N125I, and pEGFP-Rab7 Q67L were kindly provided by Robert Lodge (Université Laval, Quebec, Canada). To construct pEYFP-N1-Vpu wt, Vpu was amplified by PCR from the molecular clone HxBH10-vpu wt using the forward primer 5′-TCAAAGCAGTCTAGAGTACATGTA-3′ and the fusion primer 5′-TCCGCCGCCCCCCAGATCATCAACATC-3′. Enhanced yellow fluorescent protein (EYFP) was amplified by PCR from the mammalian expression plasmid pEYFP-N1 (Clontech) using the fusion primer 5′-GGGGGCGGCGGAGTGAGCAAGGGCGAG-3′ and reverse primer 5′-GATTATGATCTATAGTCGCGGCCGC-3′. The fusion of EYFP to the C-terminal end of Vpu was achieved by a second PCR on the two previously generated fragments using the forward and reverse primers. The resulting PCR product was inserted in pEYFP-N1 within the NheI/NotI restriction sites. The Vpu deletion mutants were generated using a similar strategy. The forward primer mentioned above was used to amplify VpuΔ9, -Δ13, -Δ14, -Δ18, and -Δ23 with the fusion primers 5′-CTCACTCCGCCGCCCCCGTGCCCCATCTCCACCCCCATC-3′, 5′-CTCACTCCGCCGCCCCCCACCCCCATCTCCACAAGTGC-3′, 5′-CTCACTCCGCCGCCCCCCCCCATCTCCACAATGCTGA-3′, 5′-CTCACTCCGCCGCCCCCAAGTGCTGATATTTCTCCTTC-3′, and 5′-CTCACTCCGCCGCCCCCTCCTTCACTCTCATTGCCACTG-3′, respectively. To amplify EYFP, the same reverse primer described above and the fusion primer 5′-GGGGGCGGCGGAGTGAGCAAG-3′ were used. The resulting PCR fragments were inserted in the pEYFP-N1 vector within the NheI/NotI restriction sites. All constructs were confirmed by automatic DNA sequencing.

Pulse-chase and radioimmunoprecipitation experiments.

The pulse-chase experiments were performed as described previously (2). Briefly, transfected HEK 293T cells were pulse labeled for 30 min with 800 μCi/ml of [35S]methionine and [35S]cysteine (Perkin Elmer) and chased in Dulbecco's modified Eagle's medium (DMEM) containing 5% fetal bovine serum. For some experiments, 10 μM BFA was added throughout the labeling and chase periods. At the indicated time periods, radiolabeled cells were lysed in radioimmunoprecipitation assay (RIPA-DOC) buffer (10 mM Tris [pH 7.2], 140 mM NaCl, 8 mM Na2HPO4, 2 mM NaH2PO4, 1% Nonidet P-40, 0.5% sodium dodecyl sulfate [SDS], 1.2 mM deoxycholate) supplemented with a cocktail of protease inhibitors (Protease Inhibitors Complete, Roche Diagnostics). Following lysis, the labeled proteins were immunoprecipitated with the indicated antibodies and analyzed by SDS-polyacrylamide gel electrophoresis (PAGE) and autoradiography (38).

Steady-state detection of proteins by Western blotting.

HeLa or HEK 293T cell transfectants were lysed in RIPA-DOC buffer. The proteins from the lysates were resolved on 10 to 12.5% SDS-PAGE Tricine gels and electroblotted as described elsewhere (2). Western blotting was performed as described previously (10).

Fluorescent and confocal microscopy.

Immunofluorescence procedures were described previously (10). Briefly, immunostaining was performed on HeLa cells transfected with either the HxBH10 proviral DNA constructs or the Vpu-expressing plasmids. Cells were fixed with 4% paraformaldehyde and permeabilized with 0.2% Triton X-100. Next, the cells were incubated with the first antibody and incubated with the appropriate secondary antibody, and for the samples analyzed by fluorescence microscopy, nuclei were stained with DAPI (4′,6-diamidino-2-phenylindole) for 5 min. To increase the EYFP signal, anti-GFP antibodies were used. The cells were examined by conventional epifluorescence micrographs on a Zeiss cell observer system (Zeiss) equipped with an Axiovert 200 M microscope using the 100× oil lens. Where specified, analyses were also performed with an Axiovert 100 M confocal microscope (Zeiss). The images were digitally deconvoluted with the AxioVision 3.1 software, using the nearest-neighbor deconvolution method. The quantitation of Vpu accumulation in the TGN was determined using the Zeiss LSM510 software. The percentage of Vpu accumulating in the TGN was calculated by evaluating the Vpu signal intensity in the TGN, as delimited by the TGN46 marker, relative to the total Vpu signal intensity in the cell. Statistical analysis was performed using an unpaired Student's t test, and values were considered statistically significant at P values of <0.05.

Transferrin uptake assay.

For the transferrin uptake experiments, siRNA-transfected HeLa cells were washed with phosphate-buffered saline (PBS) 96 h post-siRNA transfection. The cells were incubated at 4°C in serum-free DMEM. After 15 min, Alexa-Fluor-488-conjugated transferrin (Molecular Probes) was added and the cells were incubated for 30 min at 4°C. The cells were then washed with PBS and shifted to 37°C in serum-free DMEM for 10 min. The internalization process was blocked by shifting the cells at 4°C. Uninternalized transferrin on the cell membrane was washed away by washing the cells with 150 mM glycine (pH 2.5), and the uptake was determined by flow cytometry.

Viral release assays.

HeLa cells were seeded on 100-mm2 dishes and transfected as described above. The transfectants were metabolically labeled with 200 μCi/ml [35S]methionine-cysteine for 5 h. The virus particles from the supernatants were clarified by centrifugation, filtered through a 45-μm filter, and then pelleted by ultracentrifugation onto a 20% sucrose cushion in PBS for 90 min at 130,000 × g at 4°C. The cells and virions were subsequently lysed in RIPA-DOC buffer. Gag-related products were immunoprecipitated from the cell and virus lysates using a mixture of human anti-HIV (#153) serum and mouse anti-p24 monoclonal antibody. Additionally, Vpu was immunoprecipitated from the cell lysates using a polyclonal rabbit anti-Vpu serum. The radiolabeled proteins were resolved on 10% SDS-PAGE Tricine gels and analyzed by autoradiography. For the steady-state analysis, viral particles were pelleted from the supernatant of the transfected HeLa cells using the procedure described above. The proteins were analyzed by Western blotting using specific antibodies. To measure the infectious virus released from the transfected cells, HeLa-TZM indicator cells were inoculated with an aliquot of virus-containing supernatant. After 48 h, HeLa-TZM cells were lysed and luciferase activity was determined using the Promega luciferase assay system. A statistical analysis was performed using a paired Student's t test, and values were considered statistically significant at P values of <0.05.

Scanning and quantitation.

The scanning of autoradiograms and Western blots was performed on a Storm860 PhosphorImager (Molecular Dynamics) and Duoscan T1200 scanner (AGFA), respectively, followed by densitometric quantitation using the Image Quant 5.0 software (Molecular Dynamics). Viral release efficiency was evaluated by determining the ratio of the viral particle-associated Gag signal over the total Gag signal (virion and cell-associated Gag signals). Statistical analysis was performed using a paired Student's t test, and values were considered statistically significant at P values of <0.05.

RESULTS

Vpu localizes to the TGN and is associated with late endosomes.

Prototypical subtype B Vpu has been previously shown to reside predominantly in a perinuclear region corresponding to the TGN (23, 42). While a significant overlap of Vpu with TGN markers was observed in these studies, it was also apparent that not all of the population of Vpu localized within the TGN. Given that we obtained evidence indicating that Vpu turnover was increased upon exit from the ER (see Fig. S1 in the supplemental material), we hypothesized that Vpu may also traffic through late endosomal compartments. To investigate this, we examined the subcellular localization of native Vpu in HIV-1-producing restrictive HeLa cells using immunostaining and fluorescence microscopy. As reported by others (23, 42), we found that Vpu colocalized predominantly with the TGN marker TGN46. (Fig. (Fig.1A).1A). In addition to its TGN localization, Vpu was also detected in cytoplasmic punctated structures in some Vpu-expressing HeLa cells. These cells displayed a close association of Vpu with the late endosomal marker CD63 but not with the ER marker calreticulin or LAMP1, a type I transmembrane glycoprotein that is associated primarily with lysosomes and late endosomes (Fig. (Fig.1B).1B). Taken together, these results indicate that native Vpu is localized primarily in the TGN but can also be found closely associated with CD63-positive late endosomal structures.

An external file that holds a picture, illustration, etc.
Object name is zjv0090918030001.jpg

Analysis of Vpu subcellular localization. HeLa cells expressing HxBH10-vpu wt were costained with the anti-Vpu serum (red) and anti-TGN46, anti-CD63, anti-LAMP1 or anti-calreticulin antibodies (green). Nuclei were counterstained with DAPI (blue). Cells were observed by deconvolution fluorescence microscopy. Pictures show representative examples of Vpu association with TGN46 (A and B) and with CD63 (B). Enlarged pictures are shown beside panel B. White arrows indicate noticeable examples of punctate colocalization. White bars represent a distance of 10 μm.

Mutations in the hinge region of Vpu affect the subcellular localization of the protein.

The finding that Vpu is localized in the TGN and is associated to some extent with late endosomal compartments suggests that the protein is trafficking through the endosomal network and, as such, might contain motifs and/or domains governing trafficking or the retention of the protein to specific subcellular compartments. Structurally, Vpu consists of two major domains, an N-terminal transmembrane (TM) domain that anchors Vpu in cellular membranes (29, 34, 37) and a cytoplasmic tail consisting of two putative α-helices (α-helix 1, amino acids [aa] 37 to 51, and α-helix 2, aa 57 to 72) separated by a conserved DSGΦXS β-TrCP recognition site phosphorylated by casein kinase II (Fig. (Fig.2A)2A) (9, 30, 31). Upon analysis of our Vpu mutant collection, we identified a mutant of Vpu that was more stable than the Vpu wild type (wt). This mutant, Vpu R30A,K31A, harbored two alanine substitutions at the well-conserved arginine (Arg) and lysine (Lys) basic residues located at positions 30 and 31 in the hinge region between the TM and the cytoplasmic domains of the protein (Fig. (Fig.2A).2A). Interestingly, these two positively charged residues are part of a putative tyrosine-based, YXXΦ sorting signal (Y corresponds to Tyr, the Xs are residues that are highly variable, and Φ corresponds to residues with bulky hydrophobic side chains) and an overlapping dileucine-based motif, (D/E)XXXL(L/I) (D/E corresponds to Asp or Glu, the Xs are residues that are highly variable, and L/I corresponds to Leu or Ile) (Fig. (Fig.2A).2A). In contrast to the Vpu wt, Vpu R30A,K31A levels remained relatively similar throughout a 5-h pulse-chase experiment and were not affected when the protein was prevented from trafficking out of the ER by treatment with BFA, a fungal metabolite that blocks protein sorting from the ER to the Golgi complex (8) (see Fig. S1 in the supplemental material). These results suggested that these highly conserved positively charged amino acid residues (96% conservation throughout the Vpu subtypes) located in the hinge region of Vpu may affect Vpu trafficking and/or stability. In fact, single R30A or K31A mutations were also found to be sufficient to stabilize Vpu (data not shown).

An external file that holds a picture, illustration, etc.
Object name is zjv0090918030002.jpg

Mutations in the hinge region between the TM and cytoplasmic domains of Vpu affect its subcellular localization. (A) Schematic representation of the Vpu structural domains with the amino acid sequence of the hinge region derived from several strains of HIV-1 group M. Representative consensus amino acid sequences of the Vpu hinge region for each HIV-1 subtype are indicated. The red rectangle highlights the overlapping tyrosine-based (YXXΦ) and dileucine-based ([D/E]XXXL[L/I]) sorting motifs. Among sequences of all HIV-1 strains from the HIV databases of the National Institute of Allergy and Infectious Diseases (NIAID) of the National Institutes of Health (NIH; www.hiv.lanl.gov/content/index), 65% of the sequences show RK residues at positions 30 and 31 (red letters), while 96% of the sequences show a conservation of positively charged residues (RR, KR, or KK) at those positions within the hinge region. Yellow circled amino acids indicate phosphoacceptor sites at serines 52 and 56 (bold). −, amino acid identical to Vpu from HxBH10; αH, α-helix. (B) HeLa cells expressing HxBH10-vpu R30A,K31A were costained with the anti-Vpu serum (red) and anti-TGN46, anti-CD63, anti-LAMP1, or anti-calreticulin antibodies (green). Nuclei were counterstained with DAPI (blue). Pictures show representative examples of Vpu R30A,K31A-expressing cells. Enlarged pictures are shown beside the panels. White arrows indicate noticeable examples of punctate colocalization. White bars represent a distance of 10 μm. (C) Quantitation of Vpu wt and Vpu R30A,K31A accumulation in the TGN as determined by the Vpu signal measured in the TGN (region 1) relative to the total Vpu signal (region 2) in the cell. Error bars indicate the standard deviations of the means from the quantitative analysis of at least 25 distinct Vpu-expressing cells.

To investigate the effect of mutations in the hinge region on Vpu trafficking, we compared the subcellular localization of native Vpu wt and Vpu R30A,K31A in HIV-1-producing HeLa cells. As shown in Fig. Fig.2,2, the R30A,K31A mutations decreased very significantly the accumulation of native Vpu in the TGN (∼40% of the total Vpu R30A,K31A is detected in the TGN compared to ∼80% for the Vpu wt [Fig. 2B and C]) while increasing the association of the protein with the late endosomal CD63 marker (Fig. (Fig.2B).2B). Furthermore, in contrast to wt Vpu, we observed a strong colocalization of Vpu R30A,K31A with LAMP1 (compare Fig. Fig.2B2B to Fig. Fig.1B).1B). Importantly, no significant colocalization was observed between Vpu R30A,K31A and calreticulin, thus ruling out the possibility that the decreased accumulation of the mutant protein in the TGN resulted from a sequestration in the ER.

Overall, these findings suggest that mutations at Arg30 and Lys31 within the hinge region of Vpu affect both the accumulation of the protein in the TGN as well as its efficient delivery to late endosomal degradative compartments, indicating that Vpu might traffic between the TGN and the endosomal system.

Vpu traffics through the late endosomal network and is degraded in lysosomes.

To further examine whether Vpu is targeted for degradation in late endosomal structures, we treated Vpu-expressing HEK 293T and HeLa cells with CQ, a lysosomotropic agent that inhibits lysosomal protein degradation by neutralizing the acidic environment of endocytic vesicles (7). As shown in Fig. Fig.3A,3A, the treatment of transfected cells with CQ led to a dose-dependent accumulation of Vpu. Similar results were obtained when Vpu-expressing HEK 293T cells were treated with ammonium chloride, another lysosomotropic weak base that acts as a lysosomal protein degradation inhibitor (data not shown). Furthermore, to provide direct evidence that Vpu is targeted for degradation in lysosomes, we analyzed Vpu degradation and subcellular localization in conditions where we interfered with the function of Rab7, a small GTPase that is found in late endosomes (26) and is implicated in the biogenesis and maintenance of the perinuclear lysosomal compartment (5). For that purpose, we took advantage of constructs encoding enhanced GFP (EGFP)-Rab7 fusion proteins that do not perturb the delivery of cargo to lysosomes (the Rab7 wt and the constitutively active Rab7 Q67L) or, inversely, that interfere with lysosomal targeting and degradation (Rab7 N125I) (5, 26). While the overexpression of EGFP-Rab7 wt or EGFP-Rab7 Q67L did not alter the level of Vpu at steady state, the DN EGFP-Rab7 N125I drastically increased the steady-state levels of Vpu (Fig. (Fig.3B,3B, compare lanes 8 to 10 to lanes 2 to 7). Interestingly, expression of DN EGFP-Rab7 N125I in Vpu+ HIV-1-expressing HeLa cells led to an accumulation of Vpu in Rab7-positive vesicular structures while expression of EGFP-Rab7 wt (Fig. (Fig.3C)3C) or EGFP-Rab7 Q67L did not (data not shown). Importantly, accumulation of Vpu in the TGN was not significantly affected by the overexpression of the DN Rab7 N125I mutant, suggesting that endosome-to-TGN trafficking was not perturbed under these conditions (Fig. (Fig.3D).3D). Altogether, these data indicate that native Vpu traffics through the late endosomal network and undergoes degradation in lysosomes.

An external file that holds a picture, illustration, etc.
Object name is zjv0090918030003.jpg

Abrogation of lysosomal function or targeting increases Vpu stability. (A) Vpu wt-expressing HEK 293T (top panels; SVCMV-vpu+) or HeLa cells (bottom panels; HxBH10-vpu wt) were treated for 20 h with the indicated increasing concentrations of CQ prior to lysis. Vpu was subsequently resolved on SDS-PAGE and detected by Western blotting using anti-Vpu antibodies. Actin served as a loading control. (B) HEK 293T cells were cotransfected with SVCMV-vpu wt and the pEGFP-Rab7 plasmids as indicated. Proteins were resolved on SDS-PAGE and detected by Western blotting using anti-Vpu, anti-actin, and anti-GFP antibodies. Note that the blots in panels A and B are underexposed to clearly show Vpu stabilization upon CQ treatment or pEGFP-Rab7 N125I coexpression. (C) HeLa cells were cotransfected with the HxBH10-vpu wt proviral construct and either the pEGFP-Rab7 wt or pEGFP-Rab7 N125I plasmids. Transfected cells were stained with anti-Vpu rabbit serum (red). Nuclei were counterstained with DAPI (blue). White arrows indicate noticeable colocalization of Vpu with EGFP-Rab7 N125I in vesicular structures. The white bar represents a distance of 20 μm. (D) Quantitation of Vpu accumulation in the TGN was performed as described in the legend to Fig. Fig.22.

Mutations in the hinge region of Vpu interfere with Vpu-mediated enhancement of HIV-1 particle release.

Having shown that the Vpu R30A,K31A mutant displayed defects in lysosomal delivery and degradation as well as in accumulation in the TGN, we examined the implication of these defects on the Vpu-mediated enhancement of viral particle release. Equal DNA amounts of Vpu-defective proviral construct (HxBH10-vpu-) or proviral constructs encoding the Vpu wt (HxBH10-vpu wt) or Vpu R30A,K31A (HxBH10-vpu R30A,K31A) were transfected in restrictive HeLa cells. Forty-eight hours posttransfection, HIV-1-producing cell cultures were metabolically labeled for 5 h and both cells and extracellular supernatants were harvested. Virus particles were further isolated from the supernatants by clarification, filtration, and ultracentrifugation as described in Materials and Methods. Gag protein levels were then analyzed in cell and viral particle lysates by immunoprecipitation using a serum from an HIV-1-positive individual combined with anti-24 polyclonal antibodies. Figure 4A and B reveal that while newly synthesized Vpu wt viral particles were released more efficiently than Vpu-defective particles, the Vpu R30A,K31A mutant virus exhibited a significant (P < 0.001) ∼40% attenuation of viral particle release efficiency compared to that of the Vpu wt virus control. Interestingly, this attenuation was accentuated to ∼80% when viral particle release was evaluated in conditions where the Vpu wt and Vpu R30A,K31A protein levels were comparable (Fig. 4C and D, compare lane 5 to lane 4). In this context, the mislocalization of Vpu R30A,K31A from the TGN was slightly enhanced (data not shown). Importantly, the Vpu R30A,K31A mutant was found to mediate CD4 degradation as efficiently as the Vpu wt (see Fig. S2 in the supplemental material), indicating that these mutations in the hinge region did not affect the overall conformation of the protein but rather specifically interfered with the protein's ability to promote viral particle release. These results suggest that the mislocalization of Vpu outside of the TGN and/or the inefficient delivery of the protein to lysosomal compartments leads to the impairment of Vpu-mediated enhancement of viral particle release. To discriminate between these two possibilities, we examined the effect of disrupting Rab7-mediated transport on the release of HIV-1 particles in HeLa cells. The Vpu-defective proviral construct was cotransfected with Vpu-expressing or control plasmids and the pEGFP-Rab7 wt or pEGFP-Rab7 N125I plasmids. As shown in Fig. 4E and F, the expression of the DN Rab7 N125I mutant stabilized Vpu, as expected, but had no significant effect on the enhancement of viral particle release. Similarly, the abrogation of endocytic vesicle acidification and lysosomal degradation by CQ treatment did not affect the release of Vpu wt HIV-1 particles (data not shown). These findings indicate that the delivery and degradation of Vpu in lysosomes do not represent essential steps in the process underlying the Vpu-mediated enhancement of viral particle release and, consequently, point toward the accumulation of Vpu in the TGN as a possible important requirement for the protein's ability to promote HIV-1 particle release.

An external file that holds a picture, illustration, etc.
Object name is zjv0090918030004.jpg

Mutations in the hinge region of Vpu interfere with the Vpu-mediated enhancement of HIV-1 particle release. (A) HeLa cells were mock transfected (lane 1) or transfected with either the HxBH10-vpu- (lane 2), HxBH10-vpu wt (lane 3), or HxBH10-vpu R30A,K31A (lane 4) proviral plasmids as indicated. Radiolabeled cells and supernatant-containing viral particles were harvested 48 h posttransfection and analyzed for the presence of Vpu and Gag proteins by immunoprecipitation as described in Materials and Methods. (B) Densitometric quantitation of viral release efficiency. Bands corresponding to Gag products in cells and viral particles were scanned by laser densitometry. The release efficiency of HxBH10-vpu wt was arbitrarily set at 100%. Error bars indicate the standard deviations of the means of the results from four separate experiments. (C) HeLa cells were transfected with HxBH10-vpu- alone (lane 1) or with increasing quantities of HxBH10-vpu wt (lanes 2 to 4) or HxBH10-vpu R30A,K31A (lanes 5 to 7). The total amount of proviral DNA transfected in each condition was identical. Cells and supernatant-containing viral particles were harvested 48 h posttransfection, and Gag proteins in cell and virus lysates were analyzed by Western blotting using anti-p24 antibodies. Vpu was detected by Western blotting using anti-Vpu antibodies. The asterisk indicates a nonspecific band. (D) Same as described for panel C except that infectious virions in culture supernatants were measured using HeLa-TZM indicator cells and a chemiluminescence assay in relative light units (RLU), as described in Materials and Methods. The maximal RLU value obtained with HxBH10-vpu wt-expressing cells (lane 4) was arbitrarily set at 100%. Note that the data shown in lanes 4 and 5 correspond to the activities obtained with comparable levels of Vpu wt and Vpu R30A,K31A (panel C). (E) HeLa cells were cotransfected with the HxBH10-vpu- proviral construct, the indicated pEGFP-Rab7 plasmids, and the SVCMV-vpu wt or control constructs. Cells and virus particles were processed and analyzed as described for panel C. (F) Densitometric quantitation of the results shown in panel E. Viral particle release efficiency obtained in cells coexpressing HxBH10-vpu- and SVCMV-vpu wt was arbitrarily set at 100%. Error bars indicate the standard deviations of the means of the results from three separate experiments.

Disruption of Vpu trafficking from the TGN to the endosomal system stimulates HIV-1 particle release.

Clathrin-coated vesicles are major carriers for endocytic cargo and mediate important intracellular trafficking events at the TGN and endosomes. Whereas CHCs provide the structural backbone of the clathrin coat and are critical for all clathrin-mediated trafficking processes recent evidence suggests that CLCs are not required for clathrin-mediated endocytosis but are critical for clathrin-mediated trafficking between the TGN and the endosomal system (25). For instance, CLC depletion by siRNA did not have any influence on the clathrin-mediated endocytosis of transferrin but caused the CI-MPR to cluster near the TGN (25). To obtain direct evidence that the localization of Vpu in the TGN is important for Vpu function on HIV-1 particle release, we assessed the effect of disrupting clathrin-mediated trafficking from the TGN to the endosomal system on the Vpu-mediated enhancement of HIV-1 particle release using siRNA specific for CLCs or CHCs. Equal DNA amounts of proviral constructs defective for Vpu or encoding wt Vpu or the Vpu R30A,K31A mutant were transfected in HeLa cells that were depleted for CLCs beforehand. Viral particle release efficiency was then evaluated 96 h post-siRNA transfection by measuring the levels of Gag associated with cell or viral particle lysates (Fig. (Fig.5A)5A) as well as by evaluating the relative infectivity of the released viral particles using the HeLa-TZM indicator cell line (Fig. (Fig.5B).5B). HeLa cells showed reduced levels of CLCs (approximately a 60% decrease) after the transfection of siRNA specific for CLCs compared to those of the control HeLa cells transfected with a nonspecific scrambled siRNA (Fig. (Fig.5A,5A, compare lanes 4 to 6 to lanes 1 to 3). As previously reported (25), the depletion of CLCs led to a clustering of CI-MPR near the TGN and did not affect the clathrin-mediated endocytosis of transferrin from the cell surface (see Fig. S3A and 3B in the supplemental material). Although the depletion of CLCs did not influence the release of Vpu-defective HIV-1 particles, it had a stimulatory effect on the release of Vpu+ HIV-1 particles. Indeed, a significant ∼30% increase (P = 0.035) in viral particle release efficiency was observed in Vpu-expressing cells transfected with CLC siRNA compared to that of the control (Fig. 5A and B). This stimulation correlated with a stabilization of the Vpu wt (Fig. (Fig.5A,5A, compare lane 2 to lane 5), suggesting that the depletion of CLCs is indeed efficiently retaining Vpu within the TGN and, as such, preventing its trafficking to lysosomes. Interestingly, the defect in viral particle release observed with the Vpu R30A,K31A mutant was completely abrogated under conditions where CLC levels were reduced. Indeed, under these conditions, the release efficiency of the Vpu R30A,K31A mutant was comparable to that of the Vpu wt. Importantly, this abrogation of the viral particle release defect correlated with an increased accumulation of the Vpu R30A,K31A mutant in compartments that costained with the TGN marker TGN46 (Fig. 5C and D). As expected, Vpu R30A,K31A was not increasingly stabilized upon CLC siRNA treatment (Fig. (Fig.5A,5A, compare lane 3 with lane 6) since this mutant is already impaired in its ability to be targeted for degradation in lysosomes. Overall, these findings suggest that this mutant is not impaired in its ability to overcome the restricting activity of tetherin but is rather inefficient at localizing at the TGN. As expected, similar results were obtained when viral particle release was analyzed in HeLa cells that were transfected with CHC siRNA. Under these conditions, the release efficiencies of the Vpu wt- and Vpu R30A,K31A-encoded HIV-1 particles were equivalent (see Fig. S4 in the supplemental material). Taken together, these results further suggest that native Vpu is trafficking between the TGN and the endosomal system. Additionally, they provide strong evidence that the proper distribution of Vpu in the TGN at steady state is a key requirement to overcome the restricting activity of tetherin on HIV-1 particle release.

An external file that holds a picture, illustration, etc.
Object name is zjv0090918030005.jpg

Effect of the depletion of CLCs on the subcellular localization of Vpu and its viral particle release enhancing function. (A) HeLa cells were transfected either with control siRNA (Scrambled) (lanes 1 to 3) or specific siRNA against CLCa and CLCb (CLCs) (lanes 4 to 6). Seventy-two hours post-siRNA transfection, cells were transfected with similar amounts of the HxBH10-vpu- (lanes 1 and 4), HxBH10-vpu wt (lanes 2 and 5), or HxBH10-vpu R30A,K31A (lanes 3 and 6) proviral constructs as indicated. Cells and supernatant-containing viral particles were harvested 96 h post-siRNA transfection. Gag proteins in cell and virus lysates were analyzed by Western blotting using anti-p24 antibodies. Vpu and CLC levels were determined using specific antibodies. Actin served as a loading control. (B) Same as described for panel A except that infectious virions in culture supernatants were measured using HeLa-TZM indicator cells and a chemiluminescence assay in relative light units (RLU) as described in Materials and Methods. The RLU value obtained in HxBH10-vpu wt-expressing cells transfected with the nontargeting control siRNA was arbitrarily set at 100%. Error bars indicate the standard deviations of the means of the results from four independent experiments. (C) HeLa cells were transfected with either the scrambled siRNA or with CLC siRNA. Cells were transfected with the HxBH10-vpu wt or HxBH10-vpu R30A,K31A proviral plasmid 72 h later. Twenty-four hours later, cells were costained for Vpu (red) and TGN46 (green). Nuclei were counterstained with DAPI (blue). The white bar represents a distance of 10 μm. (D) Quantitation of Vpu accumulation in the TGN was performed as described in the legend to Fig. Fig.22.

The membrane proximal tyrosine- and dileucine-based sorting motifs of subtype B Vpu are not functionally active.

Having obtained evidence that Vpu traffics between the TGN and the endosomal system and that mutations at Arg30 and Lys31 in the Vpu hinge region affect this process, we next evaluated whether these residues are part of bona fide tyrosine-based (YXXΦ) or dileucine-based ([D/E]XXXL[L/I]) sorting signals, which might regulate Vpu trafficking between the TGN and late endosomal/lysosomal compartments. Toward this goal, we generated expression plasmids encoding Vpu mutants that harbored substitution mutations at the Tyr residue located at position 29 (SVCMV-vpu Y29A) or at the Ile and Leu residues located at position 32 and 33 (SVCMV-vpu I32A,L33A) (Fig. (Fig.2A)2A) since residues at these positions have been previously shown to be essential for the function of tyrosine- and dileucine-based trafficking signals (3). An analysis of Vpu Y29A and Vpu I32A,L33A subcellular localization did not reveal any marked redistribution of the mutant proteins compared to that of wt Vpu (Fig. 6A and B). Furthermore, mutations at Tyr29 or at Ile32 and Leu33 did not reveal any significant change in the ability of Vpu to enhance HIV-1 particle release (Fig. 6C and D). Moreover, in contrast to Vpu R30A,K31A, the Vpu Y29A and Vpu I32A,L33A mutants did not display any increase in their steady-state levels compared to that of the Vpu wt (Fig. (Fig.6C,6C, compare lanes 4 and 5 with lane 2). These findings indicate that the tyrosine- and the dileucine-based trafficking motifs do not appear to actively regulate Vpu trafficking from the TGN to the endosomal system and, additionally, are not essential for the viral particle release enhancing function of the protein at least in the context of subtype B Vpu. Consequently, the phenotypes observed when the Arg30 and Lys31 residues are mutated might result from a limited activation of these putative trafficking motifs.

An external file that holds a picture, illustration, etc.
Object name is zjv0090918030006.jpg

Mutations in the putative overlapping tyrosine- and dileucine-based trafficking signals do not interfere with Vpu subcellular localization or Vpu-mediated enhancement of HIV-1 particle release. (A) HeLa cells transfected with the SVCMV-vpu wt, SVCMV-vpu Y29A or SVCMV-vpu I32A,L33A plasmid were costained with the anti-Vpu serum (red) and anti-TGN46 antibodies (green) 48 h posttransfection and observed by confocal microscopy. Pictures show representative examples of Vpu localization patterns. The white bar represents a distance of 10 μm. (B) Quantitation of Vpu accumulation in the TGN as determined by the Vpu signal measured in the TGN relative to the total Vpu signal in the cell. Error bars indicate the standard deviations of the means from the quantitative analysis of at least 25 distinct Vpu-expressing cells. (C) HeLa cells were cotransfected with the specified HxBH10 proviral constructs. Cells and supernatant-containing viral particles were analyzed as described in the legend to Fig. Fig.4C.4C. (D) Same as described for panel C except that infectious virions in culture supernatants were measured using HeLa-TZM indicator cells and a chemiluminescence assay. The RLU value obtained in Vpu wt-expressing cells (lane 2) was arbitrarily set at 100%. Error bars indicate the standard deviations of the means of the results from four independent experiments.

Deletion analysis of the Vpu cytosolic tail identifies a determinant important for Vpu retention/localization in the TGN and efficient HIV-1 viral particle release.

To further delineate the region of the cytoplasmic tail that might be important for the subcellular localization of Vpu, we used a Vpu-EYFP reporter system to assess the intracellular localization of Vpu C-terminal deletion mutants by fluorescence microscopy. This approach was previously used by Pacyniak et al. (23) to identify a region within the cytoplasmic domain of subtype B Vpu that might be responsible for the retention of the protein in the TGN. The Vpu-EYFP fusion protein expressed in restrictive HeLa cells demonstrated a subcellular distribution pattern similar to that observed with native Vpu expressed from an infectious molecular clone (compare Fig. Fig.7B7B to Fig. Fig.1A).1A). Notably, ∼87% of the Vpu fluorescence staining was found to localize in a perinuclear region that overlapped with the Golgi marker TGN46 (Fig. 7B and C). No accumulation of wt Vpu-EYFP was observed at the plasma membrane. We next examined the subcellular localization of various Vpu-EYFP deletion mutants (Fig. (Fig.7A).7A). As reported by Pacyniak et al. (23), removal of 9 (VpuΔ9-EYFP) or 13 (VpuΔ13-EYFP) aa from the carboxyl terminus of Vpu had, overall, marginal effects on the localization of the protein (Fig. 7B and C). In contrast, the deletion of the C-terminal 14, 18, and 23 amino acids, which encompass the second helical structure of the Vpu cytoplasmic tail, decreased significantly the accumulation of Vpu-EYFP in the TGN while increasing its detection within cytosolic vesicular structures, which costained in part with the early endosome marker Rab5 and with CD63 (Fig. 7B to D). Interestingly, VpuΔ14, -Δ18, and -Δ23 could also be detected at the plasma membrane in some cells (data not shown).

An external file that holds a picture, illustration, etc.
Object name is zjv0090918030007.jpg

Effect of deletions of the Vpu cytosolic tail on Vpu subcellular localization. (A) Schematic representation of the Vpu-EYFP deletion constructs. Yellow circled amino acids indicate phosphoacceptor sites at serines 52 and 56 (bold). αH, α-helix. (B) HeLa cells transfected with plasmids encoding the indicated Vpu-EYFP deletion mutant were costained with the anti-GFP (green) and anti-TGN46 (red) antibodies. Cells were observed by confocal microscopy. Pictures show representative examples of the localization pattern observed for each deletion mutant. (C) Quantitation of Vpu accumulation in the TGN was performed as described in the legend to Fig. Fig.2.2. (D) HeLa cells expressing Vpu-EYFP or VpuΔ23-EYFP were costained with anti-GFP (green) and anti-CD63 or anti-Rab5 (red) antibodies. Cells were observed by confocal microscopy. Enlarged pictures are shown beside the panels. White arrows indicate noticeable examples of punctate colocalization. All the white bars represent a distance of 10 μm.

We next assessed the effect of Vpu cytoplasmic tail deletion on the ability of the protein to promote the release of the HIV-1 particle. Vpu-EYFP or Vpu-EYFP deletion mutants were cotransfected with the Vpu-defective proviral construct HxBH10-vpu- in HeLa cells, and viral particle release was determined 48 h posttransfection by measuring Gag protein levels in cell and viral particle lysates by Western blotting (Fig. (Fig.8A).8A). In addition, we also determined the viral particle release efficiency by evaluating the relative infectivity of the released viral particles (Fig. (Fig.8B).8B). The results revealed that the VpuΔ9-EYFP and VpuΔ13-EYFP mutants, which were minimally affected in their localization to the TGN, promoted the release of HIV-1 viral particles almost as efficiently as wt Vpu-EYFP (Fig. (Fig.8A8A [compare lanes 3 and 4 to lane 8] and B). In contrast, the VpuΔ14-EYFP, VpuΔ18-EYFP, and VpuΔ23-EYFP deletion mutants, which displayed a significant decrease in the accumulation of the protein in the TGN, showed an attenuation of their viral particle release-enhancing function (Fig. (Fig.8A8A [compare lanes 5 to 7 to lane 8] and B). The removal of the C-terminal 23 amino acids of Vpu abrogated the capacity of the protein to enhance particle release to a level comparable to that of the Vpu-defective HIV-1 virus control (Fig. (Fig.8A,8A, compare lane 7 to lane 2). Overall, these findings suggest that the C-terminal 23 amino acids of Vpu, which contain the second α-helical domain of the Vpu cytoplasmic domain, might be important for the localization/retention of the protein in the TGN. Furthermore, these results provide additional evidence that the proper distribution of Vpu in the TGN is important for the efficient release of viral particles from the cell surface.

An external file that holds a picture, illustration, etc.
Object name is zjv0090918030008.jpg

Effect of deletions of the Vpu cytosolic tail on Vpu-mediated enhancement of viral HIV-1 release. (A) HeLa cells were mock transfected (lane 1) or transfected with HxBH10-vpu- and plasmids encoding EYFP (lane 2) or Vpu-EYFP (lane 8) or the specified Vpu-EYFP deletion mutants (lanes 3 to 7). Cells and supernatants were analyzed as described in the legend to Fig. Fig.4C.4C. EYFP or Vpu-EYFP derivatives were detected using anti-GFP antibodies. (B) Same as described for panel A except that infectious virions in culture supernatants were measured using HeLa-TZM indicator cells and a chemiluminescence assay as described in Materials and Methods. The RLU measured with the Vpu-EYFP wt was arbitrarily set at 100%. Error bars indicate the standard deviations of the means of the results from four independent experiments.

Subtype B Vpu colocalizes with tetherin in the TGN.

Vpu was recently shown to colocalize with tetherin within cytoplasmic structures (22, 40). However, the identity of the intracellular compartments where the two proteins colocalize remains undefined. To characterize the intracellular organelles where Vpu and tetherin reside, we transfected a proviral construct encoding Vpu in HeLa cells, which are known to express tetherin, and determined the subcellular localization of native Vpu and tetherin by immunostaining using specific antibodies. Tetherin localized predominantly within cytoplasmic structures that corresponded at least in part to the TGN (Fig. (Fig.9A).9A). We observed a strong colocalization of Vpu and tetherin in the TGN as shown by the strong costaining of Vpu, tetherin, and TGN46, yet very low if any colocalization was observed in other tetherin-containing subcellular compartments (Fig. (Fig.9A).9A). Similar results were obtained when VpuΔ23-EYFP fusion proteins were expressed in HeLa cells (Fig. (Fig.9B).9B). The quantitation of the proportion of total Vpu that colocalized with tetherin revealed that ∼70% of Vpu (green pixels) overlapped with tetherin (blue pixels) (Fig. (Fig.9C).9C). Having shown that a large proportion of Vpu wt colocalizes with tetherin in the TGN, we next assessed the localization of the Vpu R30A,K31A or the VpuΔ23-EYFP mutants relative to tetherin. Overall, the extent of the colocalization between the Vpu mutants and tetherin was not as extensive as that of the Vpu wt. Although a fraction of the Vpu R30A,K31A and VpuΔ23-EYFP mutants still colocalized with tetherin in the TGN, a significant proportion of the Vpu mutant proteins did not reveal any significant colocalization with tetherin (Fig. (Fig.9A,9A, panels a to d, and B, panels e to h). In fact, whereas ∼70% of the total Vpu wt colocalized with tetherin, only ∼40% of the total Vpu R30A,K31A and VpuΔ23-EYFP mutants displayed such a colocalization (Fig. (Fig.9C),9C), suggesting that the mislocalization of Vpu outside of the TGN decreased the overall degree of Vpu colocalization with tetherin. Importantly, the localization of Vpu did not appear to be influenced by the presence of tetherin since the depletion of tetherin using specific siRNA did not alter the subcellular localization of the wt or Vpu R30A,K31A mutant proteins (data not shown). Taken together, these results suggest a strong correlation between the degree of Vpu colocalization with tetherin in the TGN and the efficiency of viral particle release.

An external file that holds a picture, illustration, etc.
Object name is zjv0090918030009.jpg

Analysis of the subcellular localization of native Vpu and tetherin. HeLa cells expressing HxBH10-vpu-, HxBH10-vpu wt, HxBH10-vpu R30A,K31A (A) or Vpu-EYFP or VpuΔ23-EYFP (B) were costained for Vpu (A) or GFP (B) (green) as well as for tetherin (blue) and TGN46 (red). Cells were observed by confocal microscopy. Enlarged pictures are shown beside panels A and B. White arrows indicate noticeable examples of punctate Vpu localization. White bars represent a distance of 10 μm. (C) Quantitation of the extent of Vpu colocalization with tetherin using the Zeiss LSM510 software. The values (%) represent the fraction of Vpu (green pixels) that overlapped with tetherin (blue pixels) relative to the total Vpu in the cell. Error bars indicate the standard deviations of the means from the quantitative analysis of at least 25 distinct Vpu-expressing cells.

DISCUSSION

In this study, we have analyzed the intracellular localization and trafficking of native subtype B Vpu in HIV-1-producing human cells. The data presented herein provide evidence that native Vpu traffics between the TGN and the endosomal system and that during this process, some Vpu proteins are ultimately delivered to lysosomal compartments where they undergo degradation. Interestingly, mutations of highly conserved positively charged amino acid residues (Arg30 and Lys31) within the hinge region of Vpu were shown to decrease the steady-state localization of the protein in the TGN while increasing the localization of the protein in CD63/LAMP1-positive late endosomal compartments. Furthermore, the disruption of clathrin-mediated trafficking between the TGN and the endosomal system using siRNA directed against CLCs had a stabilizing effect on wild-type Vpu and restored Vpu R30A,K31A distribution in the TGN to Vpu wt levels. Since the Vpu R30A,K31A mutant was found to be less efficiently targeted for degradation in the lysosomes than the Vpu wt, it seems therefore likely that these positively charged amino acids are affecting the efficiency and/or specificity of Vpu trafficking between the TGN and the endosomal system. Interestingly, Arg30 and Lys31 are part of sequences that were recently proposed to encode overlapping tyrosine-based (YXXΦ) and dileucine-based ([D/E]XXXL[L/I]) sorting motifs in subtype C Vpu (27) (Fig. (Fig.2A).2A). Tyrosine- and dileucine-based sorting signals are found in the cytosolic domains of transmembrane proteins and are implicated in endocytosis as well as the targeting of transmembrane proteins to lysosomes and lysosome-related organelles (12, 17). While the Y residue of YXXΦ sorting motifs is essential for function, the Φ position can accommodate several residues with bulky hydrophobic side chains, which depending on their identity can specify the properties of the signal (3). With regard to (D/E)XXXL(L/I) dileucine-based sorting motifs, the acidic residue (D/E) has been previously shown to be important for targeting late endosomes or lysosomes, whereas the first of the two leucines is generally invariant and its substitution with an isoleucine, as in subtype B Vpu (EYRKIL), reduces the potency of the signal (3). Interestingly, in both of these motifs, the X residues have been shown to contribute to the strength and fine specificity of the signal (14). Surprisingly, the mutation of the key Tyr or Ile and Leu residues within these overlapping sorting motifs did not affect the subcellular distribution and stability of Vpu or its enhancing function on HIV-1 particle release, suggesting that at least in the context of subtype B Vpu, these putative sorting motifs are not fully active and do not appear to regulate Vpu trafficking and function. These findings raise the possibility that subtype B Vpu TGN-to-endosome trafficking may not be actively regulated and may indeed occur by bulk flow. These results are in sharp contrast with recent observations made by Ruiz et al., which found that the mutation of the conserved Tyr residue in the hinge region of subtype C Vpu drastically decreased the viral replication of simian-HIV virus encoding subtype C Vpu (27). This same group also showed that the mutation of the second leucine (L39G) of the dileucine-based motif (EYRKLL) of subtype C Vpu leads to a protein that is transported to the cell surface less efficiently, with the majority retained within the Golgi complex (27). It is therefore likely that the distinct requirement of subtype C and subtype B Vpu for tyrosine- and dileucine-based sorting signals is related to the activity of their respective sorting motifs as well as their different patterns of localization. Importantly, the finding that the putative overlapping sorting motifs of subtype B Vpu are not fully active suggests that mutations of the conserved positively charged amino acid residues may have restored to some extent their activities, thus resulting in a change of Vpu subcellular localization and stability. Although it is still conceivable at this point that these conserved, positively charged amino acid residues may regulate Vpu trafficking by a mechanism that is independent from these putative sorting motifs, the fact that current evidence suggests that subtype C Vpu contains conserved functional trafficking signals makes this possibility less likely. Clearly, a more detailed analysis of the functional requirements of these canonical tyrosine- and dileucine-based sorting motifs will be required to fully understand their respective roles in Vpu trafficking and function.

Even though our data suggest that subtype B Vpu traffics to some extent between the TGN and the endosomal system, at steady state, the protein is predominantly localized in the TGN. In this regard, it has been shown that the steady-state distribution of the well-studied endoprotease furin in the TGN is achieved by a mechanism involving two independent signals, which consist of a CK-II-phosphorylated acidic peptide (SDSEEDE, in which the serines can be phosphorylated) and the tetrapeptide YKGL (36). While the phosphorylated acidic domain is responsible for the retention of furin in the TGN, specifically by regulating the transport from late endosomes to the TGN through binding to the adaptor protein PACS1, the YKGL signal is involved in the endosomal retrieval of furin that has escaped to the cell surface (20, 28, 43). TGN38 is another protein that also uses two nonoverlapping signals to mediate its steady-state localization in the TGN (18). In the case of TGN38, the membrane-spanning domain contains a retention signal that localizes the protein in the TGN, while a tyrosine-containing motif is responsible for retrieving escaped TGN38 from the plasma membrane via the recycling endosomes (4, 24). This type of retention/retrieval mechanism establishes an intracellular recycling loop, which allows a predominant Golgi distribution at steady state because the endocytic recycling is fast compared with the exit rate from the Golgi complex to the plasma membrane (18). Interestingly, our deletion analysis of the cytoplasmic tail of subtype B Vpu confirmed previous results from Pacyniak et al. (23), indicating that a domain encompassing the second helical structure of Vpu was important for the retention or localization of the protein in the TGN. However, in contrast to the data obtained by Pacyniak et al., the deletion of these putative retention/localization sequences did not lead to a marked relocalization of the protein to the plasma membrane. In HeLa cells, we rather observed a predominant accumulation of mutant proteins in intracellular vesicles that costained in part with the Rab5 and CD63 endosomal markers, as exemplified by the VpuΔ23-EYFP mutant. This discrepancy is not currently understood but may be related to the distinct cell lines (HeLa versus 293) that were used. While speculative at this juncture, our data raise the possibility that the steady-state TGN distribution of Vpu may be determined by two targeting signals in the cytoplasmic domain of the protein; a signal in the second α-helical domain that ensures the retention/localization of Vpu within the TGN by preventing its exit to more distal compartments and trafficking signals in the hinge region that regulates trafficking between the TGN and the endosomal system and that may also act as a retrieval motif for Vpu proteins that have escaped to other distal compartments. Given the functional importance of Vpu localization in the TGN (as discussed below), the sequence heterogeneity observed at the level of these YXXΦ or (D/E)XXXL(L/I) motifs among different Vpu subtypes (CXXL for subtype D or EXXXIL for subtype B [Fig. [Fig.2A])2A]) may indeed reflect the selective pressure for maintaining trafficking/retrieval signals of different types and strengths in the context of Vpu proteins that are more or less retained in the TGN, as exemplified by subtype B and C Vpu proteins. Clearly more studies will be required to precisely define and dissect these targeting/retention signals and to analyze their individual and combined contributions to Vpu trafficking and steady-state distribution in the TGN.

Our finding that mutations that mislocalize Vpu outside the TGN impair the Vpu-mediated enhancement of HIV-1 particle release suggests that the proper distribution of Vpu in the TGN is critical to overcome the restricting activity of tetherin. This notion is indeed consistent with results from our experiments showing that the retention of the Vpu wt or the Vpu R30A,K31A mutant in TGN-related structures following the disruption of clathrin-mediated TGN-to-endosome trafficking stimulates the Vpu-mediated enhancement of viral particle release. Interestingly, the mutation of the dileucine motif of subtype C Vpu, which resulted in an increased accumulation of the protein in the TGN, led to a similar stimulation of the Vpu-mediated enhancement of viral particle release (27). Thus, the TGN could conceivably represent one key intracellular site from where Vpu overcomes the restricting activity of tetherin on HIV-1 particle release. In support of this model, we have also shown that Vpu colocalizes with tetherin specifically in the TGN: no significant colocalization of subtype B Vpu and tetherin was observed outside the TGN. Furthermore, mutants of Vpu, which were mislocalized from the TGN and impaired in their ability to enhance HIV-1 particle release, displayed a marked decrease in their degree of colocalization with tetherin in the TGN. These findings suggest that a threshold level of the total pool of Vpu needs to be present in the TGN at steady state to encounter tetherin and efficiently overcome its antiviral activity. Since Vpu was recently shown to downregulate tetherin from the cell surface (44), it is therefore conceivable that Vpu overcomes the restricting activity of tetherin at the cell surface by trapping the protein in the TGN.

In conclusion, the results presented herein suggest that native Vpu traffics between the TGN and the endosomal system in HIV-1-producing cells. Importantly, we provide evidence suggesting that the proper distribution of Vpu in the TGN is critical to overcome the restricting activity of tetherin on HIV-1 particle release.

Supplementary Material

[Supplemental material]

Acknowledgments

We thank Robert Lodge for the kind gift of the pEGFP-Rab7 plasmids used in this study as well as René-Pierre Lorgeoux and Alexandre Orthwein for experimental help and for sharing reagents and unpublished data. We also thank present and former members of the Cohen group for helpful discussions. HeLa-TZM cells were obtained from the AIDS Research and Reference Reagent Program (NIH).

This work was supported by grants from the Canadian Institutes of Health Research (CIHR) (MOP-14228) and from the Fonds de la Recherche en Santé du Québec (FRSQ) to E.A.C. M.D. and P.G.-G. are the recipients of studentships from the CIHR strategic training program in cancer research, while J.B. is the recipient of a studentship from the FRSQ. E.A.C. holds the Canada research chair in human retrovirology.

Footnotes

Published ahead of print on 25 February 2009.

Supplemental material for this article may be found at http://jvi.asm.org/.

REFERENCES

1. Binette, J., and E. A. Cohen. 2004. Recent advances in the understanding of HIV-1 Vpu accessory protein functions. Curr. Drug Targets Immune Endocr. Metabol. Disord. 4297-307. [PubMed] [Google Scholar]
2. Binette, J., M. Dube, J. Mercier, and E. A. Cohen. 2007. Requirements for the selective degradation of CD4 receptor molecules by the human immunodeficiency virus type 1 Vpu protein in the endoplasmic reticulum. Retrovirology 475. [PMC free article] [PubMed] [Google Scholar]
3. Bonifacino, J. S., and L. M. Traub. 2003. Signals for sorting of transmembrane proteins to endosomes and lysosomes. Annu. Rev. Biochem. 72395-447. [PubMed] [Google Scholar]
4. Bos, K., C. Wraight, and K. K. Stanley. 1993. TGN38 is maintained in the trans-Golgi network by a tyrosine-containing motif in the cytoplasmic domain. EMBO J. 122219-2228. [PMC free article] [PubMed] [Google Scholar]
5. Bucci, C., P. Thomsen, P. Nicoziani, J. McCarthy, and B. van Deurs. 2000. Rab7: a key to lysosome biogenesis. Mol. Biol. Cell 11467-480. [PMC free article] [PubMed] [Google Scholar]
6. Cohen, E. A., E. F. Terwilliger, J. G. Sodroski, and W. A. Haseltine. 1988. Identification of a protein encoded by the vpu gene of HIV-1. Nature 334532-534. [PubMed] [Google Scholar]
7. de Duve, C., T. de Barsy, B. Poole, A. Trouet, P. Tulkens, and F. Van Hoof. 1974. Commentary. Lysosomotropic agents. Biochem. Pharmacol. 232495-2531. [PubMed] [Google Scholar]
8. Doms, R. W., G. Russ, and J. W. Yewdell. 1989. Brefeldin A redistributes resident and itinerant Golgi proteins to the endoplasmic reticulum. J. Cell Biol. 10961-72. [PMC free article] [PubMed] [Google Scholar]
9. Federau, T., U. Schubert, J. Flossdorf, P. Henklein, D. Schomburg, and V. Wray. 1996. Solution structure of the cytoplasmic domain of the human immunodeficiency virus type 1 encoded virus protein U (Vpu). Int. J. Pept. Protein Res. 47297-310. [PubMed] [Google Scholar]
10. Finzi, A., A. Brunet, Y. Xiao, J. Thibodeau, and E. A. Cohen. 2006. Major histocompatibility complex class II molecules promote human immunodeficiency virus type 1 assembly and budding to late endosomal/multivesicular body compartments. J. Virol. 809789-9797. [PMC free article] [PubMed] [Google Scholar]
11. Gottlinger, H. G., T. Dorfman, E. A. Cohen, and W. A. Haseltine. 1993. Vpu protein of human immunodeficiency virus type 1 enhances the release of capsids produced by gag gene constructs of widely divergent retroviruses. Proc. Natl. Acad. Sci. USA 907381-7385. [PMC free article] [PubMed] [Google Scholar]
12. Gough, N. R., M. E. Zweifel, O. Martinez-Augustin, R. C. Aguilar, J. S. Bonifacino, and D. M. Fambrough. 1999. Utilization of the indirect lysosome targeting pathway by lysosome-associated membrane proteins (LAMPs) is influenced largely by the C-terminal residue of their GYXXphi targeting signals. J. Cell Sci. 1124257-4269. [PubMed] [Google Scholar]
13. Klimkait, T., K. Strebel, M. D. Hoggan, M. A. Martin, and J. M. Orenstein. 1990. The human immunodeficiency virus type 1-specific protein vpu is required for efficient virus maturation and release. J. Virol. 64621-629. [PMC free article] [PubMed] [Google Scholar]
14. Kongsvik, T. L., S. Honing, O. Bakke, and D. G. Rodionov. 2002. Mechanism of interaction between leucine-based sorting signals from the invariant chain and clathrin-associated adaptor protein complexes AP1 and AP2. J. Biol. Chem. 27716484-16488. [PubMed] [Google Scholar]
15. Levesque, K., Y. S. Zhao, and E. A. Cohen. 2003. Vpu exerts a positive effect on HIV-1 infectivity by down-modulating CD4 receptor molecules at the surface of HIV-1-producing cells. J. Biol. Chem. 27828346-28353. [PubMed] [Google Scholar]
16. Malim, M. H., and M. Emerman. 2008. HIV-1 accessory proteins—ensuring viral survival in a hostile environment. Cell Host Microbe 3388-398. [PubMed] [Google Scholar]
17. Marks, M. S., P. A. Roche, E. van Donselaar, L. Woodruff, P. J. Peters, and J. S. Bonifacino. 1995. A lysosomal targeting signal in the cytoplasmic tail of the beta chain directs HLA-DM to MHC class II compartments. J. Cell Biol. 131351-369. [PMC free article] [PubMed] [Google Scholar]
18. Maxfield, F. R., and T. E. McGraw. 2004. Endocytic recycling. Nat. Rev. Mol. Cell Biol. 5121-132. [PubMed] [Google Scholar]
19. Meusser, B., and T. Sommer. 2004. Vpu-mediated degradation of CD4 reconstituted in yeast reveals mechanistic differences to cellular ER-associated protein degradation. Mol. Cell 14247-258. [PubMed] [Google Scholar]
20. Molloy, S. S., L. Thomas, J. K. VanSlyke, P. E. Stenberg, and G. Thomas. 1994. Intracellular trafficking and activation of the furin proprotein convertase: localization to the TGN and recycling from the cell surface. EMBO J. 1318-33. [PMC free article] [PubMed] [Google Scholar]
21. Neil, S. J., S. W. Eastman, N. Jouvenet, and P. D. Bieniasz. 2006. HIV-1 Vpu promotes release and prevents endocytosis of nascent retrovirus particles from the plasma membrane. PLoS Pathog. 2e39. [PMC free article] [PubMed] [Google Scholar]
22. Neil, S. J., T. Zang, and P. D. Bieniasz. 2008. Tetherin inhibits retrovirus release and is antagonized by HIV-1 Vpu. Nature 451425-430. [PubMed] [Google Scholar]
23. Pacyniak, E., M. L. Gomez, L. M. Gomez, E. R. Mulcahy, M. Jackson, D. R. Hout, B. J. Wisdom, and E. B. Stephens. 2005. Identification of a region within the cytoplasmic domain of the subtype B Vpu protein of human immunodeficiency virus type 1 (HIV-1) that is responsible for retention in the Golgi complex and its absence in the Vpu protein from a subtype C HIV-1. AIDS Res. Hum Retrovir. 21379-394. [PubMed] [Google Scholar]
24. Ponnambalam, S., C. Rabouille, J. P. Luzio, T. Nilsson, and G. Warren. 1994. The TGN38 glycoprotein contains two non-overlapping signals that mediate localization to the trans-Golgi network. J. Cell Biol. 125253-268. [PMC free article] [PubMed] [Google Scholar]
25. Poupon, V., M. Girard, V. Legendre-Guillemin, S. Thomas, L. Bourbonniere, J. Philie, N. A. Bright, and P. S. McPherson. 2008. Clathrin light chains function in mannose phosphate receptor trafficking via regulation of actin assembly. Proc. Natl. Acad. Sci. USA 105168-173. [PMC free article] [PubMed] [Google Scholar]
26. Press, B., Y. Feng, B. Hoflack, and A. Wandinger-Ness. 1998. Mutant Rab7 causes the accumulation of cathepsin D and cation-independent mannose 6-phosphate receptor in an early endocytic compartment. J. Cell Biol. 1401075-1089. [PMC free article] [PubMed] [Google Scholar]
27. Ruiz, A., M. S. Hill, K. Schmitt, J. Guatelli, and E. B. Stephens. 2008. Requirements of the membrane proximal tyrosine and dileucine-based sorting signals for efficient transport of the subtype C Vpu protein to the plasma membrane and in virus release. Virology 37858-68. [PMC free article] [PubMed] [Google Scholar]
28. Schafer, W., A. Stroh, S. Berghofer, J. Seiler, M. Vey, M. L. Kruse, H. F. Kern, H. D. Klenk, and W. Garten. 1995. Two independent targeting signals in the cytoplasmic domain determine trans-Golgi network localization and endosomal trafficking of the proprotein convertase furin. EMBO J. 142424-2435. [PMC free article] [PubMed] [Google Scholar]
29. Schubert, U., S. Bour, A. V. Ferrer-Montiel, M. Montal, F. Maldarell, and K. Strebel. 1996. The two biological activities of human immunodeficiency virus type 1 Vpu protein involve two separable structural domains. J. Virol. 70809-819. [PMC free article] [PubMed] [Google Scholar]
30. Schubert, U., P. Henklein, B. Boldyreff, E. Wingender, K. Strebel, and T. Porstmann. 1994. The human immunodeficiency virus type 1 encoded Vpu protein is phosphorylated by casein kinase-2 (CK-2) at positions Ser52 and Ser56 within a predicted alpha-helix-turn-alpha-helix-motif. J. Mol. Biol. 23616-25. [PubMed] [Google Scholar]
31. Schubert, U., T. Schneider, P. Henklein, K. Hoffmann, E. Berthold, H. Hauser, G. Pauli, and T. Porstmann. 1992. Human-immunodeficiency-virus-type-1-encoded Vpu protein is phosphorylated by casein kinase II. Eur. J. Biochem. 204875-883. [PubMed] [Google Scholar]
32. Schubert, U., and K. Strebel. 1994. Differential activities of the human immunodeficiency virus type 1-encoded Vpu protein are regulated by phosphorylation and occur in different cellular compartments. J. Virol. 682260-2271. [PMC free article] [PubMed] [Google Scholar]
33. Stephens, E. B., C. McCormick, E. Pacyniak, D. Griffin, D. M. Pinson, F. Sun, W. Nothnick, S. W. Wong, R. Gunderson, N. E. Berman, and D. K. Singh. 2002. Deletion of the vpu sequences prior to the env in a simian-human immunodeficiency virus results in enhanced Env precursor synthesis but is less pathogenic for pig-tailed macaques. Virology 293252-261. [PubMed] [Google Scholar]
34. Strebel, K., T. Klimkait, F. Maldarelli, and M. A. Martin. 1989. Molecular and biochemical analyses of human immunodeficiency virus type 1 vpu protein. J. Virol. 633784-3791. [PMC free article] [PubMed] [Google Scholar]
35. Terwilliger, E. F., E. A. Cohen, Y. C. Lu, J. G. Sodroski, and W. A. Haseltine. 1989. Functional role of human immunodeficiency virus type 1 vpu. Proc. Natl. Acad. Sci. USA 865163-5167. [PMC free article] [PubMed] [Google Scholar]
36. Thomas, G. 2002. Furin at the cutting edge: from protein traffic to embryogenesis and disease. Nat. Rev. Mol. Cell Biol. 3753-766. [PMC free article] [PubMed] [Google Scholar]
37. Tiganos, E., J. Friborg, B. Allain, N. G. Daniel, X. J. Yao, and E. A. Cohen. 1998. Structural and functional analysis of the membrane-spanning domain of the human immunodeficiency virus type 1 Vpu protein. Virology 25196-107. [PubMed] [Google Scholar]
38. Tiganos, E., X.-J. Yao, J. Friborg, N. Daniel, and E. A. Cohen. 1997. Putative α-helical structures in the human immunodeficiency virus type 1 Vpu protein and CD4 are involved in binding and degradation of the CD4 molecule. J. Virol. 714452-4460. [PMC free article] [PubMed] [Google Scholar]
39. Van Damme, N., D. Goff, C. Katsura, R. L. Jorgenson, R. Mitchell, M. C. Johnson, E. B. Stephens, and J. Guatelli. 2008. 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 3245-252. [PMC free article] [PubMed] [Google Scholar]
40. Van Damme, N., and J. Guatelli. 2008. HIV-1 Vpu inhibits accumulation of the envelope glycoprotein within clathrin-coated, Gag-containing endosomes. Cell. Microbiol. 101040-1057. [PubMed] [Google Scholar]
41. Varthakavi, V., R. M. Smith, S. P. Bour, K. Strebel, and P. Spearman. 2003. Viral protein U counteracts a human host cell restriction that inhibits HIV-1 particle production. Proc. Natl. Acad. Sci. USA 10015154-15159. [PMC free article] [PubMed] [Google Scholar]
42. Varthakavi, V., R. M. Smith, K. L. Martin, A. Derdowski, L. A. Lapierre, J. R. Goldenring, and P. Spearman. 2006. The pericentriolar recycling endosome plays a key role in Vpu-mediated enhancement of HIV-1 particle release. Traffic 7298-307. [PubMed] [Google Scholar]
43. Wan, L., S. S. Molloy, L. Thomas, G. Liu, Y. Xiang, S. L. Rybak, and G. Thomas. 1998. PACS-1 defines a novel gene family of cytosolic sorting proteins required for trans-Golgi network localization. Cell 94205-216. [PubMed] [Google Scholar]

Articles from Journal of Virology are provided here courtesy of American Society for Microbiology (ASM)

-