Research Article
16 December 2014

The HIV-1 gp120 CD4-Bound Conformation Is Preferentially Targeted by Antibody-Dependent Cellular Cytotoxicity-Mediating Antibodies in Sera from HIV-1-Infected Individuals

ABSTRACT

Recent studies have linked antibody Fc-mediated effector functions with protection or control of human immunodeficiency type 1 (HIV-1) and simian immunodeficiency (SIV) infections. Interestingly, the presence of antibodies with potent antibody-dependent cellular cytotoxicity (ADCC) activity in the Thai RV144 vaccine trial was suggested to correlate with decreased HIV-1 acquisition risk. These antibodies recently were found to recognize HIV envelope (Env) epitopes exposed upon Env-CD4 interaction. CD4 downregulation by Nef and Vpu, as well as Vpu-mediated BST-2 antagonism, were reported to modulate exposure of those CD4-induced HIV-1 Env epitopes and were proposed to play a role in reducing the susceptibility of infected cells to ADCC mediated by this class of antibodies. Here, we report the high prevalence of antibodies recognizing CD4-induced HIV-1 Env epitopes in sera from HIV-1-infected individuals, which correlated with their ability to mediate ADCC responses against HIV-1-infected cells, exposing these Env epitopes at the cell surface. Furthermore, our results indicate that Env variable regions V1, V2, V3, and V5 do not represent a major determinant for ADCC responses mediated by sera from HIV-1-infected individuals. Altogether, these findings suggest that HIV-1 tightly controls the exposure of certain Env epitopes at the surface of infected cells in order to prevent elimination by Fc-effector functions.
IMPORTANCE Here, we identified a particular conformation of HIV-1 Env that is specifically targeted by ADCC-mediating antibodies present in sera from HIV-1-infected individuals. This observation suggests that HIV-1 developed sophisticated mechanisms to minimize the exposure of these epitopes at the surface of infected cells.

INTRODUCTION

The IgG class of antibodies (Abs) can mediate cellular cytotoxic effector functions, such as Ab-dependent cell-mediated cytotoxicity (ADCC), viral inhibition (ADCVI), or phagocytosis (ADCP). These immune responses are driven by the engagement of the Ab Fc region with a family of proteins, known as Fcγ receptors (FcγR), at the surface of effector immune cells (1). In the case of ADCC, cross-linking of FcγRIII (CD16) leads to the activation of the associated ITAM-containing subunits CD3ζ and/or FcεRIγ, which promotes the effector cells (e.g., NK cells, macrophages, or neutrophils) to perform a cytotoxic attack on the target cell (2, 3). Interestingly, there is increasing evidence that ADCC plays a role in protecting against or controlling different viral infections (46). Accordingly, Fc-mediated effector functions were reported to correlate with decreased viral loads or rate of disease progression in both human immunodeficiency type 1 (HIV-1) and simian immunodeficiency virus (SIV) infections (714). Additionally, it was recently suggested that ADCC could apply a significant immune pressure on HIV-1 (15), which further supports a role for this effector function in vivo. The analysis of the correlates of protection in the RV144 vaccine trial suggested that increased ADCC activity was linked with decreased HIV-1 acquisition (16). Interestingly, Abs with potent ADCC activity were isolated from some RV144 vaccinees (17). However, little is known regarding the cellular and viral determinants that govern the susceptibility of HIV-1-infected cells to ADCC killing by Abs normally elicited during the course of HIV-1 infection.
We recently reported that HIV-1 envelope (Env) interaction with the CD4 receptor at the surface of infected cells was critical for efficient ADCC activity mediated by monoclonal Abs (MAbs) targeting CD4-induced (CD4i) Env epitopes (18). Our initial findings were recently corroborated by others (19). Importantly, we reported that multiple MAbs with potent ADCC activity isolated from RV144 vaccinees also recognized Env CD4i epitopes in a manner similar to that of the well-characterized inner domain recognizing A32 Ab (18, 20). Studies by other groups suggested that Abs recognizing variable regions 1 and 2 (V1V2) elicited in some RV144 vaccinees could mediate Fc effector functions (21, 22). However, little is known as to the relative contribution of inner domain CD4i to the overall Fc-mediated effector functions present in sera from HIV-1-infected individuals.

MATERIALS AND METHODS

Cells.

293T human embryonic kidney and HOS cell lines (obtained from the ATCC and NIH AIDS Research and Reference Reagent Program, respectively) were grown at 37°C and 5% CO2 in Dulbecco's modified Eagle's medium (Invitrogen) containing 10% fetal bovine serum (Sigma) and 100 μg/ml of penicillin-streptomycin (Wysent). CEM.NKr cells (obtained from David Evans, Harvard Medical School) were grown at 37°C and 5% CO2 in RPMI 1640 (Invitrogen) containing 10% fetal bovine serum and 100 μg/ml primocine (Invitrogen).
Ficoll density gradient isolated and cryopreserved human peripheral blood mononuclear cells (PBMCs) from healthy donors were thawed and kept at 37°C and 5% CO2 in RPMI 1640 containing 10% FBS and 100 μg/ml penicillin-streptomycin for at least 16 h before subsequent experiments.

Plasmids and site-directed mutagenesis.

Mutations were introduced into the previously described pNL43-ADA-GFP.IRES.Nef proviral vector (23). To generate env mutants, the SalI-BamHI fragment of pNL43-ADA-GFP.IRES.Nef was subcloned in a pUC19 intermediate before being subjected to site-directed mutagenesis using the QuikChange II XL protocol (Stratagene). The mutated insert was then cloned back into pNL43-ADA-GFP.IRES.Nef. Mutations in nef were introduced by a two-step PCR strategy using primers having 18-nucleotide overlaps and cloned back into the proviral construct using XhoI and NcoI restriction sites. All mutations were confirmed by Sanger DNA sequencing. The codon-optimized pcDNA3.1-HIV-1YU2 ΔV1V2V3V5 expression construct was made by replacing the sequence encoding residues 124 to 198 from the V1/V2 loop with a sequence encoding a GG linker and the sequence encoding residues 302 to 323 from the V3 loop with a sequence encoding a GGSGSG linker (24). ΔV5 was made by replacing residues 460 to 465 with a GSG linker into pcDNA3.1-HIV-1YU2 ΔV1V2V3. The D368R mutation was introduced into pcDNA3.1-HIV-1YU2 V1V2V3V5 by site-directed mutagenesis as described above.

Sera from HIV-infected individuals.

Informed consent was obtained from all study participants (the Montreal Primary HIV Infection Cohort [25, 26] and the Canadian Cohort of HIV-Infected Slow Progressors [2729]), and research adhered to the ethical guidelines of CRCHUM. Sera were collected during Ficoll isolation of PBMCs and conserved at −80°C. Serum aliquots were heat inactivated for 30 min at 56°C and stored at 4°C until they were used in subsequent experiments. A random-number generator (GraphPad QuickCalcs) was used to randomly select a number of sera from each cohort.

Purification of recombinant HIV-1 gp120 glycoproteins.

FreeStyle 293F cells (Invitrogen) were grown in FreeStyle 293F medium (Invitrogen) to a density of 1 × 106 cells/ml at 37°C with 8% CO2 with regular agitation (125 rpm). Cells were transfected with a pCDNA3.1 plasmid encoding codon-optimized His6-tagged wild-type (wt) or mutant HIV-1 YU2 gp120 using the 293Fectin reagent as directed by the manufacturer (Invitrogen). One week later, cells were pelleted and discarded. The supernatants were filtered (0.22-μm-pore-size filter) (Corning), and the gp120 glycoproteins were purified by nickel affinity columns according to the manufacturer's instructions (Invitrogen). The gp120 preparations were dialyzed against phosphate-buffered saline (PBS) and stored in aliquots at −80°C. To assess purity, recombinant proteins were loaded on SDS-PAGE gels and stained with Coomassie blue.

Cell-based ELISA.

Detection of trimeric Env at the surface of HOS cells was performed by cell-based ELISA, as previously described (18, 30, 31). Briefly, HOS cells were seeded in 96-well plates (2 × 104 cells per well) and transfected the next day with a cytoplasmic tail-deleted HIV-1 EnvYU2 variant alone or together with a human CD4 expressor using a standard polyethylenimine transfection method. Two days later, transfected cells were washed and then incubated with 1:1,000 dilutions of sera from HIV-infected cells or 1 μg/ml of relevant MAbs. Env-specific IgGs were detected using a horseradish peroxidase (HRP)-conjugated anti-human IgG-specific secondary Ab (Pierce) with a TriStar LB 941 luminometer (Berthold Technologies).

Viral production and infection.

Vesicular stomatitis virus G (VSV-G)-pseudotyped NL4.3 green fluorescent protein (GFP)-encoding ADA-Env-based viruses were produced as previously described (18). Briefly, our panel of pNL4.3-GFP-ADA-based HIV-1 proviral vectors and VSV-G-encoding plasmid were cotransfected in 293T cells by standard calcium phosphate transfection. Two days after transfection, cell supernatants were harvested, clarified by low-speed centrifugation (5 min at 1,200 rpm), and concentrated by ultracentrifugation for 1 h at 4°C at 143,260 × g over a 20% sucrose cushion. Pellets were harvested in fresh RPMI, and aliquots were stored at −80°C until use. Viral preparations were normalized before infection according to reverse transcriptase activity or using a standard 50% tissue culture infectious dose (TCID50) procedure using TZM-bl cells (32). Viruses were then used to infect approximately 20% to 30% of CEM.NKr cells by spin infection at 800 × g for 1 h in 96-well plates at 25°C.

Flow cytometry analysis of cell surface staining, ADCC responses, and binding competition assays.

For cell surface staining, infected or mock-infected CEM.NKr cells were incubated for 20 min at room temperature 48 h postinfection with 1 μg/ml OKT4 (anti-CD4 Ab; 14-0048-82; eBioscience), 2 μg/ml BST-2 (sc-99191; Santa Cruz), 1 μg/ml 2G12 (AB002; Polymun), 1 μg/ml A32 Ab (kindly given by J. Robinson), or a 1:1,000 final dilution of serum from participants in PBS. Cells were then washed once with PBS and stained with 1 μg/ml goat anti-mouse and anti-rabbit (Alexa Fluor 594; Invitrogen) or anti-human (Alexa Fluor 647; Invitrogen) secondary Abs for 15 min in PBS. After one more PBS washing, cells were fixed in a 2% PBS-formaldehyde solution.
Measurement of serum- and A32-mediated ADCC was performed with a previously described assay (18, 33). Briefly, CEM.NKr-infected cells were stained with viability (AquaVivid; Invitrogen) and cellular (cell proliferation dye eFluor670; eBioscience) markers and used as target cells. PBMC effectors cells, stained with another cellular marker (cell proliferation dye eFluor450; eBioscience), were then mixed at an effector/target (E/T) ratio of 10:1 in 96-well V-bottom plates (Corning). A 1:1,000 final dilution of serum or 5 μg/ml of the A32 MAb was added to appropriate wells. Cocultures were centrifuged for 1 min at 300 × g and incubated at 37°C for 5 to 6 h before being fixed in a 2% PBS-formaldehyde solution containing 5 × 104 flow cytometry particles/ml (AccuCount blank particles; 5.3 μm; Spherotech). Samples were analyzed on an LSRII cytometer (BD Biosciences) and acquisition was set to acquire 1,000 particles, which allows the calculation of relative cell counts. Data analysis was performed using FlowJo vX.0.7 (Tree Star). The percentage of cytotoxicity was calculated with the following formula: (relative count of GFP+ cells in targets plus effectors) − (relative count of GFP+ cells in targets plus effectors plus A32 or serum)/(relative count of GFP+ cells in targets) according to our previously described gating strategy (18, 33).
For serum adsorption and gp120 competition assays, serum dilutions from HIV-1-infected individuals were preincubated for 30 min at room temperature with purified soluble D368R gp120 ΔV1V2V3V5 at a concentration of 83.3 pmol/μl of serum. This concentration was determined by assaying the dose-dependent reduction in gp120-coated cell staining (see Fig. S4 in the supplemental material), as previously reported (33).

RESULTS

Env-CD4 interaction enhances recognition of HIV-1-infected cells by sera from HIV-1-infected individuals.

HIV-1 accessory proteins Nef and Vpu are known to modulate cell surface levels of CD4 (34, 35). In addition to its role in CD4 degradation, Vpu also antagonizes a restriction factor, Tetherin/BST-2, which normally inhibits retroviral release (36, 37). Viruses lacking Vpu remain trapped at the cell surface, resulting in an accumulation of exposed Env (18, 19). Therefore, Nef and Vpu can indirectly modulate Env-CD4 interaction at the surface of infected cells through CD4 and BST-2 downregulation (18). Accordingly, we recently reported that cells infected with viruses defective for both Nef and Vpu present enhanced levels of CD4 and Env at the cell surface, resulting in the exposure of Env CD4i epitopes (18). To address whether these epitopes were recognized at the surface of infected cells by sera from HIV-1-infected individuals, we infected CEM.NKr cells with a panel of NL4.3-GFP ADA-Env-based viruses encoding either wild-type (wt) or Nef- and Vpu-defective accessory proteins as previously described (18). Two days postinfection, infected cells were stained with sera from HIV-infected individuals and then fluorescently labeled (Fig. 1). Interestingly, as we previously reported for the ADCC-mediating A32 Ab (18), we observed that a threshold of both CD4 and Env must be reached at the cell surface, and Env must be able to engage with CD4 in order for Env to be detected by A32 or sera from HIV-1-infected individuals (Fig. 1 and 2; also see Fig. S1 and S2 in the supplemental material). The majority of the sera recognized more efficiently cells infected with a virus lacking Nef and Vpu than its wild-type counterpart. Cells infected with viruses lacking Nef and Vpu (i.e., presenting high levels of CD4 and Env at the cell surface [Fig. 2A; also see Fig. S2]) but where the ability of Env to interact with CD4 was dramatically decreased by a CD4-binding site mutation (D368R) (38, 39) were poorly recognized by sera from HIV-1-infected individuals. Of note, the decreased recognition of the D368R Env variant by sera from HIV-1-infected individuals was not due to decreased levels of CD4 and/or Env at the surface of cells infected with HIV-1 lacking nef and vpu (see Fig. S2).
FIG 1
FIG 1 Env-CD4 interaction is required for efficient recognition of infected cells by sera from HIV-1-infected individuals. CEM.NKr cells infected with a panel of VSV-G-pseudotyped NL4.3 GFP ADA-Env-based viruses expressing the wild type (wt) or a CD4-binding site (D368R) Env variant, lacking Nef (Nef−) or expressing a Nef variant (L166A-L168A) defective for CD4 downregulation (34) (NefAA) or lacking Vpu (Vpu−) or both Nef and Vpu (Nef− Vpu−), were stained at 48 h postinfection with sera from 30 HIV-infected individuals and then fluorescently labeled with an Alexa Fluor 647-conjugated anti-human IgG secondary Ab. (A) Histograms depicting representative staining of infected (GFP+) cells by serum from one HIV and one HIV+ donor. (B) Fold increase of staining relative to mock infection for all tested sera. Data shown are the results from two different experiments, and error bars depict the standard errors of the means (SEM). Statistical significance was tested using paired one-way analyses of variance (ANOVA) (*, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001).
FIG 2
FIG 2 Env-CD4 interaction modulates the exposure of the Env ADCC-mediating A32 epitope at the surface of infected cells. CEM.NKr cells infected with a panel of VSV-G-pseudotyped NL4.3 GFP ADA-Env-based viruses expressing the wild type (wt) or a CD4-binding site (D368R) Env variant, lacking Nef (Nef−) or expressing a Nef variant (L166A-L168A) defective for CD4 downregulation (34) (NefAA) or lacking Vpu (Vpu−) or both Nef and Vpu (Nef− Vpu−), were stained at 48 h postinfection for surface CD4 levels (A) or Env A32 epitope exposure (B). Data shown are the results from at least three different experiments, and error bars depict SEM. Statistical significance was tested using paired one-way ANOVA (*, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001).
The impact of Env-CD4 interaction on the binding of serum IgGs from 163 HIV-infected individuals was also evaluated by cell-based ELISA, as previously described (18, 30, 31). Briefly, HOS cells were transfected with a cytoplasmic tail-deleted HIV-1 EnvYU2 variant alone or with a human CD4 expressor as reported previously (18, 30). Two days later, transfected cells were washed and then incubated with 1:1,000 dilutions of sera from HIV-infected individuals. Env-specific IgGs were detected using an anti-human IgG-specific secondary Ab. Interestingly, the recognition of the Env trimer by sera from all of the clinical categories tested was significantly increased by the coexpression of CD4, while the outer domain, recognizing Ab 2G12, was not affected (see Fig. S3 in the supplemental material), suggesting that sera from HIV-1-infected individuals contain a significant portion of CD4i Abs, as previously proposed (40).

The ability of sera from HIV-infected individuals to mediate ADCC requires Env-CD4 interaction.

We previously described that Env-CD4 interaction modulates the susceptibility of HIV-1-infected cells to ADCC by CD4i Abs (18). However, whether this was also the case for sera from a large number of HIV-1-infected individuals remains unknown. Therefore, we sought to determine if the overall ADCC activity potential in sera from HIV-infected individuals would be associated with efficient Env-CD4 interaction. Using infected CEM.NKr cells as described above, we measured serum-mediated ADCC with our previously described fluorescence-activated cell sorting (FACS)-based ADCC assay (18, 33). Thus, we determined the ability of 48 randomly chosen sera (including those tested in Fig. 1B) from HIV-infected individuals to mediate ADCC against infected cells (Fig. 3). Strikingly, while cells infected with wt virus were slightly more susceptible to ADCC killing than mock-infected cells, those infected with a virus lacking Nef and Vpu were dramatically more susceptible to ADCC (Fig. 3A). Interestingly, the slight increase in ADCC killing of cells lacking Nef was similar to that obtained with cells infected with a Nef variant (L166A-L168A) unable to downregulate CD4 from the cell surface (34), further stressing the importance of CD4 downregulation to protect infected cells from ADCC. However, the increase in ADCC observed with cells lacking Nef or Vpu alone was significantly lower than ADCC levels reached with cells infected with viruses lacking both accessory proteins. Moreover, the ADCC potential of different sera correlated with their ability to recognize Env at the surface of cells infected with viruses lacking both Nef and Vpu (Fig. 3B), suggesting that the efficient recognition of HIV-1-infected cells by sera is required for their ability to mediate ADCC. Finally, introducing the CD4-binding site D368R mutation in Env dramatically decreased the sensitivity of infected cells to killing by ADCC (Fig. 3A and C), further stressing the importance of Env-CD4 interaction for exposure of epitopes recognized by ADCC-mediating Abs normally elicited in the course of HIV-1 infection.
FIG 3
FIG 3 Env-CD4 interaction modulates susceptibility of infected cells to ADCC killing mediated by sera from HIV-1-infected individuals. (A) CEM.NKr cells infected with a panel of VSV-G-pseudotyped NL4.3 GFP ADA-based viruses expressing the wild type (wt) or a CD4-binding site (D368R) Env variant, lacking Nef (Nef−) or expressing a Nef variant (L166A-L168A) defective for CD4 downregulation (34) (NefAA) or lacking Vpu (Vpu−) or both Nef and Vpu (Nef− Vpu−), were used at 48 h postinfection as target cells in our FACS-based ADCC assay (18, 33) to determine their susceptibility to sera from HIV-1-infected individuals to mediate cell lysis by PBMCs from healthy donors. Data shown are the results from three different experiments (median ± interquartile range). (B) A positive correlation was observed between the staining intensity of sera from HIV-1-infected individuals on cells infected with Nef- and Vpu-defective virus and their ability to mediate ADCC. (C) Paired values of ADCC mediated by sera from HIV-1-infected individuals and those of cells infected by HIV-1 viruses lacking both Nef and Vpu and encoding a wt or D368R Env variant. Statistical significance was tested using paired one-way ANOVA (A) or a paired t test (*, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001) (C).

Antibodies targeting the V1V2V3 and V5 gp120 variable regions do not play a major role in HIV+ serum-mediated ADCC.

Recent reports indicate that various vaccine-elicited Abs targeting conserved (CD4i) or variable epitopes (V1V2) as well as rare broadly neutralizing Abs can mediate ADCC against HIV-1-infected cells (17, 18, 21, 22, 41). To investigate whether, aside from gp120 CD4-induced antibodies, epitopes recognized by anti-gp41, variable regions, or quaternary-dependent antibodies contributed to serum-mediated ADCC responses, we designed an antibody competition assay using purified, soluble gp120 lacking variable regions V1, V2, V3, and V5 while presenting a D368R mutation (ΔV1V2V3V5 D368R) making it unable to bind cell surface CD4 (33). Of note, variable region 4 (V4) could not be removed without affecting the structural integrity of the protein (42 and data not shown). The preincubation of sera with either full-length or ΔV1V2V3V5 D368R gp120 recombinant proteins captured anti-Env antibodies and prevented the recognition of gp120-coated cells by serum Abs (see Fig. S4 in the supplemental material). Interestingly, this was also observed at the surface of HIV-1-infected cells where competition with the ΔV1V2V3V5 recombinant gp120 almost completely abrogated cell surface staining of cells infected with virus unable to express Nef and Vpu (Fig. 4A) and correlated with decreased ADCC activity (Fig. 4B), indicating that a recombinant protein lacking the V1V2V3V5 variable regions is sufficient to absorb the majority of ADCC activity present in sera from HIV-1-infected individuals.
FIG 4
FIG 4 A recombinant gp120 protein lacking V1V2V3 and V5 variable regions is sufficient to adsorb the majority of ADCC activity present in sera from HIV-infected individuals. CEM.NKr cells infected with wt or Nef- and Vpu-defective VSV-G-pseudotyped NL4.3 GFP ADA-based viruses were used at 48 h postinfection for surface staining (A) or FACS-based ADCC assay (B) using sera from HIV-1-infected individuals preincubated in the absence or presence of 83.3 pmol/μl ΔV1V2V3V5 D368R serum for 30 min at room temperature. Data shown are representative of at least two different experiments. Statistical significance was tested using paired t test (**, P < 0.01; ns, not significant).

DISCUSSION

Renewed interest in Fc-mediated functions, such as ADCC, stems in part from correlations in controlling or preventing HIV-1 infection. Fc-mediated effector functions were found to inversely correlate with viral loads or decreased disease progression in SIV-infected macaques (79) as well as in HIV-1-infected individuals (1014). Furthermore, the analysis of immune protection correlates in the recent RV144 vaccine trial suggested that high levels of ADCC-mediating Abs correlated with decreased HIV-1 acquisition when combined with low plasma IgA anti-Env Ab levels (16, 43), warranting further studies on the viral determinants modulating ADCC.
Recent observations suggested that Vpu antagonism of BST2 was sufficient to protect HIV-infected cells from ADCC (44, 45). In this study, we found no significant increase in the susceptibility of cells infected with a virus lacking Vpu to serum-mediated ADCC (Fig. 3). However, in agreement with previous work done with CD4i Abs (18), decreasing Env-CD4 interaction by introducing an Env CD4 binding site mutation (D368R) in the context of a virus lacking both Nef and Vpu was sufficient to dramatically diminish killing of infected cells by serum-mediated ADCC (Fig. 3C). This is also supported by our soluble gp120 competition assay, suggesting that gp120 CD4i Abs represent the major determinant of serum-mediated ADCC in HIV-1-infected individuals (Fig. 4), since this recombinant protein could not absorb Abs directed against gp41, quaternary-dependent Abs, or V1V2V3 and V5 variable regions. In this study, we only tested sera from HIV-1-infected individuals (not from vaccinees); therefore, we do not exclude the possibility that additional types of ADCC-mediating Abs, such as anti-V1V2, elicited through vaccination, could have therapeutic utility and/or help in preventing infection.
Furthermore, stratifying the patient sera in clinical disease progression rates (classic, rapid progressors or long-term nonprogressors, as described previously [25, 26]) did not reveal any significant differences between them as to the requirement of Env-CD4 interaction to promote serum-mediated ADCC against infected cells (see Table S1 in the supplemental material).
Recent observations suggested that the angle of approach of the Ab is important in order to mediate ADCC (46). Whether anti-gp120 CD4i Abs bind Env with an angle of approach that promotes the recruitment of Fc-bearing effector cells is unclear but warrants further studies.
Altogether, these data suggest that ADCC-mediating Abs are elicited in the course of natural HIV-1 infection and that a significant proportion of them require Env to interact with CD4 in order to be effective at mediating ADCC. This supports a major role of CD4i Env epitopes in the susceptibility of infected cells to ADCC mediated by sera from HIV-1-infected individuals. Our data suggest that HIV-1, via Nef and Vpu, tightly controls cell surface levels of CD4 and Env in order to limit the exposure of potential epitopes recognized by ADCC-mediating Abs elicited in the course of natural HIV-1 infection. Therefore, targeting the ability of Vpu and Nef to downregulate CD4 and BST-2 or strategies aimed at modifying Env conformation to expose CD4i epitopes could render HIV-1-infected cells susceptible to ADCC and have therapeutic utility.

ACKNOWLEDGMENTS

We thank Nathalie Brassard, Stéphanie Matte, and the CRCHUM Flow Cytometry Platform for technical assistance, as well as Mario Legault for cohort coordination. We are thankful for the subjects' participation and collaboration.
This work was supported by a Canada Foundation for Innovation Program Leader grant, by CIHR operating grants 119334 and 134117, by FRQS Establishment of Young Scientist grant 26702 to A.F., by CIHR catalyst grant 126630 to A.F. and M.R., and by the FRQS AIDS and Infectious Diseases Network. A.F. is the recipient of a Canada Research Chair on Retroviral Entry. M.V. was supported by CIHR doctoral research award 291485, and J.R. is the recipient of CIHR fellowship award 135349.
The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
We have no conflicts of interest to report.

Supplemental Material

File (zjv999099899so1.pdf)
ASM does not own the copyrights to Supplemental Material that may be linked to, or accessed through, an article. The authors have granted ASM a non-exclusive, world-wide license to publish the Supplemental Material files. Please contact the corresponding author directly for reuse.

REFERENCES

1.
Cohen-Solal JF, Cassard L, Fridman WH, Sautes-Fridman C. 2004. Fc gamma receptors. Immunol Lett 92:199–205.
2.
Lanier LL. 2008. Up on the tightrope: natural killer cell activation and inhibition. Nat Immunol 9:495–502.
3.
Lanier LL. 2005. NK cell recognition. Annu Rev Immunol 23:225–274.
4.
Kohl S. 1991. Role of antibody-dependent cellular cytotoxicity in defense against herpes simplex virus infections. Rev Infect Dis 13:108–114.
5.
Beltran D, Lopez-Verges S. 2014. NK cells during dengue disease and their recognition of dengue virus-infected cells. Front Immunol 5:192.
6.
Jegaskanda S, Weinfurter JT, Friedrich TC, Kent SJ. 2013. Antibody-dependent cellular cytotoxicity is associated with control of pandemic H1N1 influenza virus infection of macaques. J Virol 87:5512–5522.
7.
Banks ND, Kinsey N, Clements J, Hildreth JE. 2002. Sustained antibody-dependent cell-mediated cytotoxicity (ADCC) in SIV-infected macaques correlates with delayed progression to AIDS. AIDS Res Hum Retrovir 18:1197–1205.
8.
Sun Y, Asmal M, Lane S, Permar SR, Schmidt SD, Mascola JR, Letvin NL. 2011. Antibody-dependent cell-mediated cytotoxicity in simian immunodeficiency virus-infected rhesus monkeys. J Virol 85:6906–6912.
9.
Alpert MD, Harvey JD, Lauer WA, Reeves RK, Piatak M Jr, Carville A, Mansfield KG, Lifson JD, Li W, Desrosiers RC, Johnson RP, Evans DT. 2012. ADCC develops over time during persistent infection with live-attenuated SIV and is associated with complete protection against SIV(mac)251 challenge. PLoS Pathog 8:e1002890.
10.
Baum LL, Cassutt KJ, Knigge K, Khattri R, Margolick J, Rinaldo C, Kleeberger CA, Nishanian P, Henrard DR, Phair J. 1996. HIV-1 gp120-specific antibody-dependent cell-mediated cytotoxicity correlates with rate of disease progression. J Immunol 157:2168–2173.
11.
Forthal DN, Landucci G, Haubrich R, Keenan B, Kuppermann BD, Tilles JG, Kaplan J. 1999. Antibody-dependent cellular cytotoxicity independently predicts survival in severely immunocompromised human immunodeficiency virus-infected patients. J Infect Dis 180:1338–1341.
12.
Ljunggren K, Moschese V, Broliden PA, Giaquinto C, Quinti I, Fenyo EM, Wahren B, Rossi P, Jondal M. 1990. Antibodies mediating cellular cytotoxicity and neutralization correlate with a better clinical stage in children born to human immunodeficiency virus-infected mothers. J Infect Dis 161:198–202.
13.
Chung AW, Navis M, Isitman G, Wren L, Silvers J, Amin J, Kent SJ, Stratov I. 2011. Activation of NK cells by ADCC antibodies and HIV disease progression. J Acquir Immune Defic Syndr 58:127–131.
14.
Mabuka J, Nduati R, Odem-Davis K, Peterson D, Overbaugh J. 2012. HIV-specific antibodies capable of ADCC are common in breastmilk and are associated with reduced risk of transmission in women with high viral loads. PLoS Pathog 8:e1002739.
15.
Chung AW, Isitman G, Navis M, Kramski M, Center RJ, Kent SJ, Stratov I. 2011. Immune escape from HIV-specific antibody-dependent cellular cytotoxicity (ADCC) pressure. Proc Natl Acad Sci U S A 108:7505–7510.
16.
Haynes BF, Gilbert PB, McElrath MJ, Zolla-Pazner S, Tomaras GD, Alam SM, Evans DT, Montefiori DC, Karnasuta C, Sutthent R, Liao HX, DeVico AL, Lewis GK, Williams C, Pinter A, Fong Y, Janes H, DeCamp A, Huang Y, Rao M, Billings E, Karasavvas N, Robb ML, Ngauy V, de Souza MS, Paris R, Ferrari G, Bailer RT, Soderberg KA, Andrews C, Berman PW, Frahm N, De Rosa SC, Alpert MD, Yates NL, Shen X, Koup RA, Pitisuttithum P, Kaewkungwal J, Nitayaphan S, Rerks-Ngarm S, Michael NL, Kim JH. 2012. Immune-correlates analysis of an HIV-1 vaccine efficacy trial. N Engl J Med 366:1275–1286.
17.
Bonsignori M, Pollara J, Moody MA, Alpert MD, Chen X, Hwang KK, Gilbert PB, Huang Y, Gurley TC, Kozink DM, Marshall DJ, Whitesides JF, Tsao CY, Kaewkungwal J, Nitayaphan S, Pitisuttithum P, Rerks-Ngarm S, Kim JH, Michael NL, Tomaras GD, Montefiori DC, Lewis GK, Devico A, Evans DT, Ferrari G, Liao HX, Haynes BF. 2012. Antibody-dependent cellular cytotoxicity-mediating antibodies from an HIV-1 vaccine efficacy trial target multiple epitopes and preferentially use the VH1 gene family. J Virol 86:11521–11532.
18.
Veillette M, Desormeaux A, Medjahed H, Gharsallah NE, Coutu M, Baalwa J, Guan Y, Lewis G, Ferrari G, Hahn BH, Haynes BF, Robinson JE, Kaufmann DE, Bonsignori M, Sodroski J, Finzi A. 2014. Interaction with cellular CD4 exposes HIV-1 envelope epitopes targeted by antibody-dependent cell-mediated cytotoxicity. J Virol 88:2633–2644.
19.
Pham TN, Lukhele S, Hajjar F, Routy JP, Cohen EA. 2014. HIV Nef and Vpu protect HIV-infected CD4+ T cells from antibody-mediated cell lysis through down-modulation of CD4 and BST2. Retrovirology 11:15.
20.
Ferrari G, Pollara J, Kozink D, Harms T, Drinker M, Freel S, Moody MA, Alam SM, Tomaras GD, Ochsenbauer C, Kappes JC, Shaw GM, Hoxie JA, Robinson JE, Haynes BF. 2011. An HIV-1 gp120 envelope human monoclonal antibody that recognizes a C1 conformational epitope mediates potent antibody-dependent cellular cytotoxicity (ADCC) activity and defines a common ADCC epitope in human HIV-1 serum. J Virol 85:7029–7036.
21.
Liao HX, Bonsignori M, Alam SM, McLellan JS, Tomaras GD, Moody MA, Kozink DM, Hwang KK, Chen X, Tsao CY, Liu P, Lu X, Parks RJ, Montefiori DC, Ferrari G, Pollara J, Rao M, Peachman KK, Santra S, Letvin NL, Karasavvas N, Yang ZY, Dai K, Pancera M, Gorman J, Wiehe K, Nicely NI, Rerks-Ngarm S, Nitayaphan S, Kaewkungwal J, Pitisuttithum P, Tartaglia J, Sinangil F, Kim JH, Michael NL, Kepler TB, Kwong PD, Mascola JR, Nabel GJ, Pinter A, Zolla-Pazner S, Haynes BF. 2013. Vaccine induction of antibodies against a structurally heterogeneous site of immune pressure within HIV-1 envelope protein variable regions 1 and 2. Immunity 38:176–186.
22.
Pollara J, Bonsignori M, Moody MA, Liu P, Alam SM, Hwang KK, Gurley TC, Kozink DM, Armand LC, Marshall DJ, Whitesides JF, Kaewkungwal J, Nitayaphan S, Pitisuttithum P, Rerks-Ngarm S, Robb ML, O'Connell RJ, Kim JH, Michael NL, Montefiori DC, Tomaras GD, Liao HX, Haynes BF, Ferrari G. 2014. HIV-1 vaccine-induced C1 and V2 Env-specific antibodies synergize for increased antiviral activities. J Virol 88:7715–7726.
23.
Richard J, Sindhu S, Pham TN, Belzile JP, Cohen EA. 2010. HIV-1 Vpr up-regulates expression of ligands for the activating NKG2D receptor and promotes NK cell-mediated killing. Blood 115:1354–1363.
24.
Kwon YD, Finzi A, Wu X, Dogo-Isonagie C, Lee LK, Moore LR, Schmidt SD, Stuckey J, Yang Y, Zhou T, Zhu J, Vicic DA, Debnath AK, Shapiro L, Bewley CA, Mascola JR, Sodroski JG, Kwong PD. 2012. Unliganded HIV-1 gp120 core structures assume the CD4-bound conformation with regulation by quaternary interactions and variable loops. Proc Natl Acad Sci U S A 109:5663–5668.
25.
Fontaine J, Chagnon-Choquet J, Valcke HS, Poudrier J, Roger M, Montreal Primary HIVI, Long-Term Non-Progressor Study Group. 2011. High expression levels of B lymphocyte stimulator (BLyS) by dendritic cells correlate with HIV-related B-cell disease progression in humans. Blood 117:145–155.
26.
Fontaine J, Coutlee F, Tremblay C, Routy JP, Poudrier J, Roger M, Montreal Primary HIVI, Long-Term Nonprogressor Study Group. 2009. HIV infection affects blood myeloid dendritic cells after successful therapy and despite nonprogressing clinical disease. J Infect Dis 199:1007–1018.
27.
Peretz Y, Ndongala ML, Boulet S, Boulassel MR, Rouleau D, Cote P, Longpre D, Routy JP, Falutz J, Tremblay C, Tsoukas CM, Sekaly RP, Bernard NF. 2007. Functional T cell subsets contribute differentially to HIV peptide-specific responses within infected individuals: correlation of these functional T cell subsets with markers of disease progression. Clin Immunol 124:57–68.
28.
Kamya P, Boulet S, Tsoukas CM, Routy JP, Thomas R, Cote P, Boulassel MR, Baril JG, Kovacs C, Migueles SA, Connors M, Suscovich TJ, Brander C, Tremblay CL, Bernard N, Canadian Cohort of HIVISP. 2011. Receptor-ligand requirements for increased NK cell polyfunctional potential in slow progressors infected with HIV-1 coexpressing KIR3DL1*h/*y and HLA-B*57. J Virol 85:5949–5960.
29.
International HIV Controllers Study, Pereyra F, Jia X, McLaren PJ, Telenti A, de Bakker PI, Walker BD, Ripke S, Brumme CJ, Pulit SL, Carrington M, Kadie CM, Carlson JM, Heckerman D, Graham RR, Plenge RM, Deeks SG, Gianniny L, Crawford G, Sullivan J, Gonzalez E, Davies L, Camargo A, Moore JM, Beattie N, Gupta S, Crenshaw A, Burtt NP, Guiducci C, Gupta N, Gao X, Qi Y, Yuki Y, Piechocka-Trocha A, Cutrell E, Rosenberg R, Moss KL, Lemay P, O'Leary J, Schaefer T, Verma P, Toth I, Block B, Baker B, Rothchild A, Lian J, Proudfoot J, Alvino DM, Vine S, Addo MM, Allen TM, Altfeld M, Henn MR, Le Gall S, et al. 2010. The major genetic determinants of HIV-1 control affect HLA class I peptide presentation. Science 330:1551–1557.
30.
Veillette M, Coutu M, Richard J, Batraville L-A, Désormeaux A, Roger M, Finzi A. 2014. Conformational evaluation of HIV-1 trimeric envelope glycoproteins using a cell-based ELISA assay. J Vis Exp 2014:e51995–e51995.
31.
Desormeaux A, Coutu M, Medjahed H, Pacheco B, Herschhorn A, Gu C, Xiang SH, Mao Y, Sodroski J, Finzi A. 2013. The highly conserved layer-3 component of the HIV-1 gp120 inner domain is critical for CD4-required conformational transitions. J Virol 87:2549–2562.
32.
Wei X, Decker JM, Liu H, Zhang Z, Arani RB, Kilby JM, Saag MS, Wu X, Shaw GM, Kappes JC. 2002. Emergence of resistant human immunodeficiency virus type 1 in patients receiving fusion inhibitor (T-20) monotherapy. Antimicrob Agents Chemother 46:1896–1905.
33.
Richard J, Veillette M, Batraville LA, Coutu M, Chapleau JP, Bonsignori M, Bernard N, Tremblay C, Roger M, Kaufmann DE, Finzi A. 2014. Flow cytometry-based assay to study HIV-1 gp120 specific antibody-dependent cellular cytotoxicity responses. J Virol Methods 208C:107–114.
34.
Schwartz O, Dautry-Varsat A, Goud B, Marechal V, Subtil A, Heard JM, Danos O. 1995. Human immunodeficiency virus type 1 Nef induces accumulation of CD4 in early endosomes. J Virol 69:528–533.
35.
Willey RL, Maldarelli F, Martin MA, Strebel K. 1992. Human immunodeficiency virus type 1 Vpu protein induces rapid degradation of CD4. J Virol 66:7193–7200.
36.
Neil SJ, Zang T, Bieniasz PD. 2008. Tetherin inhibits retrovirus release and is antagonized by HIV-1 Vpu. Nature 451:425–430.
37.
Van Damme N, Goff D, Katsura C, Jorgenson RL, Mitchell R, Johnson MC, Stephens EB, Guatelli J. 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 3:245–252.
38.
Brand D, Srinivasan K, Sodroski J. 1995. Determinants of human immunodeficiency virus type 1 entry in the CDR2 loop of the CD4 glycoprotein. J Virol 69:166–171.
39.
Kwong PD, Wyatt R, Robinson J, Sweet RW, Sodroski J, Hendrickson WA. 1998. Structure of an HIV gp120 envelope glycoprotein in complex with the CD4 receptor and a neutralizing human antibody. Nature 393:648–659.
40.
Decker JM, Bibollet-Ruche F, Wei X, Wang S, Levy DN, Wang W, Delaporte E, Peeters M, Derdeyn CA, Allen S, Hunter E, Saag MS, Hoxie JA, Hahn BH, Kwong PD, Robinson JE, Shaw GM. 2005. Antigenic conservation and immunogenicity of the HIV coreceptor binding site. J Exp Med 201:1407–1419.
41.
Falkowska E, Le KM, Ramos A, Doores KJ, Lee JH, Blattner C, Ramirez A, Derking R, van Gils MJ, Liang CH, McBride R, von Bredow B, Shivatare SS, Wu CY, Chan-Hui PY, Liu Y, Feizi T, Zwick MB, Koff WC, Seaman MS, Swiderek K, Moore JP, Evans D, Paulson JC, Wong CH, Ward AB, Wilson IA, Sanders RW, Poignard P, Burton DR. 2014. Broadly neutralizing HIV antibodies define a glycan-dependent epitope on the prefusion conformation of gp41 on cleaved envelope trimers. Immunity 40:657–668.
42.
Yuan T, Li J, Zhang MY. 2013. HIV-1 envelope glycoprotein variable loops are indispensable for envelope structural integrity and virus entry. PLoS One 8:e69789.
43.
Tomaras GD, Ferrari G, Shen X, Alam SM, Liao HX, Pollara J, Bonsignori M, Moody MA, Fong Y, Chen X, Poling B, Nicholson CO, Zhang R, Lu X, Parks R, Kaewkungwal J, Nitayaphan S, Pitisuttithum P, Rerks-Ngarm S, Gilbert PB, Kim JH, Michael NL, Montefiori DC, Haynes BF. 2013. Vaccine-induced plasma IgA specific for the C1 region of the HIV-1 envelope blocks binding and effector function of IgG. Proc Natl Acad Sci U S A 110:9019–9024.
44.
Alvarez RA, Hamlin RE, Monroe A, Moldt B, Hotta MT, Rodriguez Caprio G, Fierer DS, Simon V, Chen BK. 2014. HIV-1 Vpu antagonism of tetherin inhibits antibody-dependent cellular cytotoxic responses by natural killer cells. J Virol.
45.
Arias JF, Heyer LN, von Bredow B, Weisgrau KL, Moldt B, Burton DR, Rakasz EG, Evans DT. 2014. Tetherin antagonism by Vpu protects HIV-infected cells from antibody-dependent cell-mediated cytotoxicity. Proc Natl Acad Sci U S A.
46.
Acharya P, Tolbert WD, Gohain N, Wu X, Yu L, Liu T, Huang W, Huang CC, Do Kwon Y, Louder RK, Luongo TS, McLellan JS, Pancera M, Yang Y, Zhang B, Flinko R, Foulke JS Jr, Sajadi MM, Kamin-Lewis R, Robinson JE, Martin L, Kwong PD, Guan Y, DeVico AL, Lewis GK, Pazgier M. 2014. Structural definition of an antibody-dependent cellular cytotoxicity (ADCC) response implicated in reduced risk for HIV-1 infection. J Virol 88:12895–12906.

Information & Contributors

Information

Published In

cover image Journal of Virology
Journal of Virology
Volume 89Number 11 January 2015
Pages: 545 - 551
Editor: G. Silvestri
PubMed: 25339767

History

Received: 1 October 2014
Accepted: 16 October 2014
Published online: 16 December 2014

Permissions

Request permissions for this article.

Contributors

Authors

Maxime Veillette
Centre de Recherche du CHUM, Université de Montréal, Montreal, Quebec, Canada
Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montreal, Quebec, Canada
Mathieu Coutu
Centre de Recherche du CHUM, Université de Montréal, Montreal, Quebec, Canada
Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montreal, Quebec, Canada
Jonathan Richard
Centre de Recherche du CHUM, Université de Montréal, Montreal, Quebec, Canada
Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montreal, Quebec, Canada
Laurie-Anne Batraville
Centre de Recherche du CHUM, Université de Montréal, Montreal, Quebec, Canada
Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montreal, Quebec, Canada
Olina Dagher
Centre de Recherche du CHUM, Université de Montréal, Montreal, Quebec, Canada
Nicole Bernard
Department of Experimental Medicine, McGill University, Montreal, Quebec, Canada
Cécile Tremblay
Centre de Recherche du CHUM, Université de Montréal, Montreal, Quebec, Canada
Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montreal, Quebec, Canada
Daniel E. Kaufmann
Centre de Recherche du CHUM, Université de Montréal, Montreal, Quebec, Canada
Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montreal, Quebec, Canada
Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, Massachusetts, USA
Michel Roger
Centre de Recherche du CHUM, Université de Montréal, Montreal, Quebec, Canada
Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montreal, Quebec, Canada
Andrés Finzi
Centre de Recherche du CHUM, Université de Montréal, Montreal, Quebec, Canada
Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montreal, Quebec, Canada
Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada

Editor

G. Silvestri
Editor

Notes

Address correspondence to Andrés Finzi, [email protected].

Metrics & Citations

Metrics

Note:

  • For recently published articles, the TOTAL download count will appear as zero until a new month starts.
  • There is a 3- to 4-day delay in article usage, so article usage will not appear immediately after publication.
  • Citation counts come from the Crossref Cited by service.

Citations

If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. For an editable text file, please select Medlars format which will download as a .txt file. Simply select your manager software from the list below and click Download.

View Options

Figures

Tables

Media

Share

Share

Share the article link

Share with email

Email a colleague

Share on social media

American Society for Microbiology ("ASM") is committed to maintaining your confidence and trust with respect to the information we collect from you on websites owned and operated by ASM ("ASM Web Sites") and other sources. This Privacy Policy sets forth the information we collect about you, how we use this information and the choices you have about how we use such information.
FIND OUT MORE about the privacy policy