HIV-1 acute infection env glycomutants designed from 3D ... · In order to be as close as possible...

13
HIV-1 acute infection env glycomutants designed from 3D model: effects on processing, antigenicity, and neutralization sensitivity Fre ´de ´ric Reynard, Ahmed Fatmi, Bernard Verrier, and Fre ´de ´ric Bedin * FRE 2736 CNRS-bioMe ´rieux, IFR128 Biosciences, CERVI, Lyon, France Received 9 February 2004; returned to author for revision 5 March 2004; accepted 22 March 2004 Abstract The human immunodeficiency virus type 1 (HIV-1) envelope protein (Env) has evolved to limit its overall immunogenicity by extensive glycosylation. Only a few studies dealing with glycosylation sites have taken into account available 3D data in a global approach. We compared primary env sequences from patients with acute HIV-1 infection. Conserved N-glycosylation sites were placed on the gp120-3D model. Based on vicinity, we defined glycosylation clusters. According to these clusters, we engineered plasmids encoding deglycosylated gp160 mutants. We also constructed mutants corresponding to nonclustered glycans or to the full deglycosylation of the V1 or V2 loop. After in vitro expression, mutants were tested for functionality. We also compared the inhibition of pseudotyped particles infection by human- neutralizing sera. Generally, clustered and nonclustered mutants were affected similarly. Silencing of more than one glycan had deleterious effects, independently of the type of sugar removed. However, some mutants were moderately affected by glycans removal suggesting a distinct role for these N-glycans. Additionally, compared to the wild-type pseudovirus, two of these mutants were neutralized at higher sera dilutions strengthening the importance of the location of specific N-glycans in limiting the neutralizing response. These results could guide the selection of env mutants with the fewest antigenic and functional alterations but with enhanced neutralization sensitivity. D 2004 Elsevier Inc. All rights reserved. Keywords: HIV-1; Env glycoprotein; N-glycans; Neutralization; Env processing; Acute infection Introduction The envelope glycoprotein (Env) of human immunode- ficiency virus type 1 (HIV-1) promotes binding of virus to target cells through CD4/chemokine receptors complex and triggers events leading to fusion between viral and cellular membranes. During and after translation, the envelope precursor gp160 folds in endoplasmic reticulum and acquires the ability to oligomerize before being exported to the Golgi apparatus. In the Golgi, gp160 is proteolytically cleaved to create two noncovalently associated subunits, the gp120 (surface protein) and gp41 (transmembrane protein). The gp120/gp41 association is labile and may dissociate (or ‘‘shed’’) spontaneously (McKeating et al., 1991). The gp120 consists of a core that is mostly conserved among different strains and highly variable surface-exposed loops. Gp160 (about 850 residues) contains about 28 sites for N-linked carbohydrates attachment. Nearly 50% of the gp120 mass is due to carbohydrate residues and a broad variety of both high-mannose and complex-type have been identified (Leonard et al., 1990; Zhu et al., 2000). Such extensive glycosylation is very unusual regarding to the envelope glycoproteins of others viruses. As a comparison, the F and H glycoproteins of measles virus have, respec- tively, 3 and 5 glycosylation sites for 550 and 617 residues (Cattaneo and Rose, 1993). Sugars play an important role in structural folding of nascent proteins in endoplasmic retic- ulum, in precursor cleavage in the early Golgi apparatus, and consequently modulate the biological properties and the infectivity of the virus (Benjouad et al., 1992; Fischer et al., 1996). Nevertheless, N-glycans appear to be dispensable for maintaining gp120 native structure once formed (Fennie and Lasky, 1989; Li et al., 1993). Glycosylation is also a mean of diversifying proteins without recourse of the genome and a growing body of evidence demonstrates that it modulates 0042-6822/$ - see front matter D 2004 Elsevier Inc. All rights reserved. doi:10.1016/j.virol.2004.03.022 * Corresponding author. FRE 2736 CNRS-bioMe ´rieux, CERVI, 21 Avenue Tony Garnier, 69007 Lyon, France. Fax: +33-437-28-24-11. E-mail address: [email protected] (F. Bedin). www.elsevier.com/locate/yviro Virology 324 (2004) 90 – 102

Transcript of HIV-1 acute infection env glycomutants designed from 3D ... · In order to be as close as possible...

  • www.elsevier.com/locate/yviro

    Virology 324 (2004) 90–102

    HIV-1 acute infection env glycomutants designed from 3D model:

    effects on processing, antigenicity, and neutralization sensitivity

    Frédéric Reynard, Ahmed Fatmi, Bernard Verrier, and Frédéric Bedin*

    FRE 2736 CNRS-bioMérieux, IFR128 Biosciences, CERVI, Lyon, France

    Received 9 February 2004; returned to author for revision 5 March 2004; accepted 22 March 2004

    Abstract

    The human immunodeficiency virus type 1 (HIV-1) envelope protein (Env) has evolved to limit its overall immunogenicity by extensive

    glycosylation. Only a few studies dealing with glycosylation sites have taken into account available 3D data in a global approach. We

    compared primary env sequences from patients with acute HIV-1 infection. Conserved N-glycosylation sites were placed on the gp120-3D

    model. Based on vicinity, we defined glycosylation clusters. According to these clusters, we engineered plasmids encoding deglycosylated

    gp160 mutants. We also constructed mutants corresponding to nonclustered glycans or to the full deglycosylation of the V1 or V2 loop. After

    in vitro expression, mutants were tested for functionality. We also compared the inhibition of pseudotyped particles infection by human-

    neutralizing sera. Generally, clustered and nonclustered mutants were affected similarly. Silencing of more than one glycan had deleterious

    effects, independently of the type of sugar removed. However, some mutants were moderately affected by glycans removal suggesting a

    distinct role for these N-glycans. Additionally, compared to the wild-type pseudovirus, two of these mutants were neutralized at higher sera

    dilutions strengthening the importance of the location of specific N-glycans in limiting the neutralizing response. These results could guide

    the selection of env mutants with the fewest antigenic and functional alterations but with enhanced neutralization sensitivity.

    D 2004 Elsevier Inc. All rights reserved.

    Keywords: HIV-1; Env glycoprotein; N-glycans; Neutralization; Env processing; Acute infection

    Introduction

    The envelope glycoprotein (Env) of human immunode-

    ficiency virus type 1 (HIV-1) promotes binding of virus to

    target cells through CD4/chemokine receptors complex and

    triggers events leading to fusion between viral and cellular

    membranes.

    During and after translation, the envelope precursor

    gp160 folds in endoplasmic reticulum and acquires the

    ability to oligomerize before being exported to the Golgi

    apparatus. In the Golgi, gp160 is proteolytically cleaved to

    create two noncovalently associated subunits, the gp120

    (surface protein) and gp41 (transmembrane protein). The

    gp120/gp41 association is labile and may dissociate (or

    ‘‘shed’’) spontaneously (McKeating et al., 1991). The

    0042-6822/$ - see front matter D 2004 Elsevier Inc. All rights reserved.

    doi:10.1016/j.virol.2004.03.022

    * Corresponding author. FRE 2736 CNRS-bioMérieux, CERVI, 21

    Avenue Tony Garnier, 69007 Lyon, France. Fax: +33-437-28-24-11.

    E-mail address: [email protected] (F. Bedin).

    gp120 consists of a core that is mostly conserved among

    different strains and highly variable surface-exposed loops.

    Gp160 (about 850 residues) contains about 28 sites for

    N-linked carbohydrates attachment. Nearly 50% of the

    gp120 mass is due to carbohydrate residues and a broad

    variety of both high-mannose and complex-type have been

    identified (Leonard et al., 1990; Zhu et al., 2000). Such

    extensive glycosylation is very unusual regarding to the

    envelope glycoproteins of others viruses. As a comparison,

    the F and H glycoproteins of measles virus have, respec-

    tively, 3 and 5 glycosylation sites for 550 and 617 residues

    (Cattaneo and Rose, 1993). Sugars play an important role in

    structural folding of nascent proteins in endoplasmic retic-

    ulum, in precursor cleavage in the early Golgi apparatus,

    and consequently modulate the biological properties and the

    infectivity of the virus (Benjouad et al., 1992; Fischer et al.,

    1996). Nevertheless, N-glycans appear to be dispensable for

    maintaining gp120 native structure once formed (Fennie and

    Lasky, 1989; Li et al., 1993). Glycosylation is also a mean

    of diversifying proteins without recourse of the genome and

    a growing body of evidence demonstrates that it modulates

  • F. Reynard et al. / Virology 324 (2004) 90–102 91

    the exposure of the Env protein to immune surveillance in

    patients or in experimentally infected animals, acting like a

    shield (Cheng-Mayer et al., 1999; Schonning et al., 1998;

    Wei et al., 2003a, 2003b).

    The envelope glycoprotein is the target of virus-neutral-

    izing antibodies; however, it does not elicit efficient neu-

    tralizing response in infected people. The high number of N-

    glycans could partially explain why broad neutralizing

    antibodies directed against envelope glycoprotein have not

    been frequently identified.

    Several reports have described individual substitutions of

    N-glycosylation sites located on domains implied in the

    resistance to antibody-mediated neutralization. These

    domains include V1/V2 or V3 loops that mask CD4 binding

    sites or epitopes induced after binding of the CD4 (CD4-

    induced epitopes). Except for the gp41 moiety and for the

    gp120 variable loops, few systematic has been made,

    especially regarding the spatial location of N-glycans (John-

    son et al., 2001; Desrosiers et al., 1998; Hemming et al.,

    1996; Lee et al., 1992; Ogert et al., 2001). In addition,

    studies were mainly conducted on SIV or HIV-1 T-cell line-

    adapted viruses (TCLA). It is now well accepted that this

    type of virus is antigenically faraway from primary clinical

    isolates and typically more sensitive to antibodies neutral-

    ization (Moore et al., 1995).

    In order to be as close as possible to the virus that has

    founded the infection, we chose to base our study on gp120

    sequences from HIV-1 acute infection primary isolates. It

    was recently postulated that the ‘‘glycan shield’’ was evolv-

    ing during the course of infection in the face of a changing

    antibody repertoire (Wei et al., 2003a). We made the

    hypothesis that at the time of acute infection, the well-

    conserved glycosylation sites would be located in specific

    regions at the surface of the protein and serve to protect

    against humoral immune response gp120 epitopes critical

    for HIV-1 transmission. According to the 3D-model data,

    we mutated potential glycosylation sites to remove sugars

    on these different regions.

    In this approach, the major obstacle was to increase

    immunogenicity without altering the oligomeric gp120:

    we monitored the effects on Env processing and function;

    we also tested the neutralizing activity of human sera on

    pseudotyped particles harboring mutated envelopes. These

    data could be useful for rationalizing the design of new Env

    immunogens that would induce a strong broad range anti-

    body response against the HIV-1.

    Results

    Rational of deglycosylation constructs

    To identify conserved N-linked glycosylation sites of

    HIV-1 gp120, 14 primary env sequences obtained during

    acute infection and before seroconversion were aligned

    (Fig. 1). These sequences were generated from patients

    infected sexually (eight homosexuals, six heterosexuals).

    The sequence alignment revealed the presence of 10 well-

    conserved N-glycosylation sites (defined by the NX{-P}T/

    S sequon), spread all over the gp120 amino acids sequen-

    ces (N44, N88, N109, N123, N148, N201, N243, N253,

    N289, and N301/307). The N185 site was present in all

    sequences from homosexually transmitted isolates. Two

    other potential sites (N136, N142) were conserved among

    env sequences from heterosexually transmitted isolates. The

    133 env sequence retained for mutants construction has

    been isolated from a patient contaminated by the homo-

    sexual route; consequently, the N185 site was included in

    the study. Due to the systematic presence of at least one

    sugar at the close positions N231/N237, the N231 site was

    also included. The N136/N142 sites, as well as the N81

    site, were not considered as they were absent from the 133

    env sequence. Due to their proximity on 133 sequence, the

    N-glycan sites N180/N185 (as N201/N208) are removed

    simultaneously.

    To analyze the 3D distribution of the selected N-glyco-

    sylation sites on the 133 sequence, we used the gp120

    model based on the X-ray crystallography data (Kwong et

    al., 1998). Each potential site was highlighted on the 3D

    model. The selected sites were arbitrarily grouped into three

    nonoverlapping ‘‘regions’’ (or ‘‘cluster’’) based both on the

    proximity of each site with the others (Fig. 2). The size of

    each cluster was roughly delimited by a 20 Å circular area,

    considered as the typical antibody footprint (Pantophlet et

    al., 2003). The first region (colored in red; A region; Fig.

    2A), constituted by sites N44, N231, and N243, was located

    approximately on the bridging sheet, which is implied in

    receptor and coreceptor binding. The second region, pre-

    sented in blue (B region; Fig. 2B), grouped the positions

    N148, N180/N185, N201/N208. This cluster was separated

    from the first one by several residues (Q227, L262, and

    P279) critical for coreceptor binding (Rizzuto et al., 1998).

    Two out of four glycosylation sites in the B cluster (N180

    and N185) also belonged to the 2G12 neutralization epitope

    (Scanlan et al., 2002). The third region (C region; Fig. 2C),

    represented in yellow (N109, N253 and N289), was located

    on the gp120 silent face, which is well exposed at the

    surface of the envelope oligomer but does not induce

    antibodies. Among the remaining positions, the N88 and

    N301/307 sites were not retained for our study, as they

    belonged to the inner domain of the gp120 that faces up the

    gp41 moiety. This domain is poorly accessible in Env

    oligomers and does not induce neutralizing antibodies. As

    the N123 site (colored in green) remained alone, it was

    grouped with sites in the neighborhood at positions N109

    and N289 (C cluster) or N148 (B cluster) and defined the

    ‘‘D’’ mutants. All of four were roughly located near the

    base of the V3 loop. These mutants were considered as a

    separate group.

    The clusters definition was validated by checking con-

    served N-glycans location on 3D model for the 13 others

    primary gp120 sequences (data not shown).

  • Fig. 1. Positions of the potential glycosylation sites on 14 gp120 primary sequences from patients with HIV-1 acute infection. The amino acids numbering of N

    mutations corresponds to the HIV-1HXB2 gp120 sequence deleted for a part of C1 and C5 regions and for the V1V2 loop as described by Kwong et al. (1998).

    The code number of the patients and the contamination route, homosexual (HM) or heterosexual (HT), identify each sequence. The position of the missing

    V1V2 loop is indicated by a triangle.

    F. Reynard et al. / Virology 324 (2004) 90–10292

    For each region, several deglycosylation mutants were

    engineered (Table 1); in a series of mutants, all the N-

    glycan attachment sites belonging to the same region were

    removed (g4A, g5B/g6B, g5C, respectively, for the A, B,

    and C clusters). As several intermediates were generated

    for this purpose, they were also included in the study

    (g1A–g3A, g1B–g4B, g1C–g4C). In parallel, the effects

    induced by two (g1), three (g2), or four mutations (g3),

    but not located in the same region, were evaluated (Fig.

    2D). The deglycosylation of the V1 loop (g4) or the V2

    loop (g5) are modifications that improve the capacity of

    the Env protein to induce neutralizing antibodies or its

    sensitivity to neutralizing sera (Reitter and Desrosiers,

    1998; Quinones-Kochs et al., 2002). We made these

    mutations on the 133 env sequence to complete our study

    because the crystallized core-gp120 is devoid of the V1/

    V2 loops. For all the constructs, the potential N-glycosyl-

    ation sites were removed by replacing the asparagine

    residue (N) in the NX{-P}T/S sequons by a glutamine

    residue. Glutamine was chosen because of its structural

    similarity with asparagine differing by only a single

    methylene group. In addition, a minimum of two nucleo-

    tides changes are necessary to revert to the original

    asparagine codon. All the constructs were checked by

    sequencing both strands to exclude mutations induced by

    PCR during mutagenesis.

  • Fig. 2. (A–C) Positions of the mutated N-glycosylation sites on the 133 gp120 sequence according to the gp120 crystallized model viewed on three different

    angles. Each asparagine from the canonical motif NX{-P}T/S is highlighted by different colors according to its respective region. The amino acid numbering

    corresponds to the variable loops deleted envelope sequence. The missing V1/V2 structure is schematized by a gray oval. The 20 Å bar represents

    approximately the size of an antibody footprint. (D) Positions of asparagine residues according to the nonclustered mutants definition viewed on two different

    angles.

    F. Reynard et al. / Virology 324 (2004) 90–102 93

    Influence of N-glycans removal on gp160 cleavage and

    gp120 shedding

    To determine the effect of cluster deglycosylation on

    envelope processing and particularly on cleavage, 293T

    cells were transfected and both the lysates and the cell

    culture supernatants were analyzed by an anti-gp120 im-

    munoblot (Fig. 3). Luminescence scanning was used to

    detect the immunoreactive bands on immunoblots on cell

    lysates. As the Rev protein is necessary to Env synthesis,

    we checked that it was present at the same level in all

    lysates (data not shown). All constructs were expressed at

    a comparable order of magnitude (Fig. 3). The mobility of

    the gp160 and gp120 gene products expressed from g1A–

    g2A, g1B, g1C–g2C, and g1D constructs appeared to be

    close to the one of the wild-type proteins. For these

    mutants, only one or two glycosylation sites were re-

    moved. The gp160 of all others mutants migrated slightly

    faster than wild-type gp160. This result confirmed the loss

    of glycans caused by mutagenesis; the glycosylation of one

    N-glycan attachment site is predicted to contribute approx-

    imately 2–3 kDa to the apparent molecular mass of a

    protein.

    For the mutants with one N-glycan removed (i.e., g1A,

    g2A, g1B, g1C, g1D, but not g2C), gp120 was detected in

    cell lysates. Gp120 was also detected for g3C (two muta-

    tions), g4 (three mutations), and g3B (four mutations). For

    g2B, g4B, g6B, g2C, g4C, g5C, and g2D, the gp120 was

    exclusively detected in the culture supernatants: a particu-

    larly important shedding caused absence of detectable

    gp120 in cell lysates. An Env bearing one unique mutation

    at N109 or N148 was affected similarly indicating the

    leading role of these mutations in gp120/gp41 dissociation

    (cf. g2C; data not shown for N148). For the remaining

    mutants, no gp120 was detected both in the lysate and the

    supernatant; for these mutants, the gp160 cleavage was

    severely impaired. The removal of the three potential sites

    present on the V1 loop (g4) had no effect on the cleavage

    rate compared to the native protein. Cleavage on nonclus-

    tered mutants was affected in the same way as the clustered

    mutants. Additionally, the effects were apparently indepen-

    dent of the nature of the removed sugars (complex or high-

    mannose type, cf. Table 1).

    Influence of N-glycans removal on cell membrane

    presentation and incorporation in pseudovirion

    Cell-to-cell fusion assays on GHOST-CCR5 cells were

    performed (Fig. 4A). This type of experiment was carried

    out with 293T cells expressing the different mutants. The

  • Table 1

    Glycosylation status of the engineered mutants

    Region Mutants Positions of

    mutated N-glycan

    attachment sites

    gp120 regions

    mutated

    N-glycans

    removed

    WT no-mutation

    wild type

    – –

    anti no-mutation

    insert in reverse

    orientation

    – –

    (A) Red g1A 44 C2 (V1V2 stem) 1C

    g2A 243 V4 1C

    g3A 243–231 V4–V4 1C1M

    g4A 44–231–243 C2–V4–V4 2C1M

    (B) Blue g1B 180/185 C3 2M

    g2B 180/185–148 C3–V3 1C2M

    g3B 180/185–201/208 C3–C3 1C2M1U

    g4B 180/185–148–

    201/208

    C3–V3–C3 1C3M1U

    g5B 180/185–201/

    208–231

    C3–C3–V4 1C3M1U

    g6B 180/185–148–

    201/208–243

    C3–V3–C3–V4 2C3M1U

    (C)Yellow g1C 289 C4 1M

    g2C 109 C2 1H

    g3C 289–253 C4–V4 1M1C

    g4C 289–109 C4–C2 1M1H

    g5C 289–253–109 C4–V4–C2 1M1C1H

    (D) Green g1D 123 C2 1C

    g2D 123–148 C2–V3 2C

    g3D 289–109–123 C4–C2–C2 1C1M1H

    Misc. g1 289–123 C4–C2 1C1M

    g2 289–123–243 C4–C2–V4 2C1M

    g3 289–123–243–

    180/185

    C4–C2–V4–C3 2C3M

    g4 V1 deglycosylated V1 2C1U

    g5 V2 deglycosylated V2 2C

    For each mutant, location, number, and nature of sugar removed are

    indicated. C: complex-type sugar, M: high-mannose type sugar, H: hybrid

    type sugar, and U: unknown type sugar. The numbering of N-glycan

    positions is according to Fig. 1.

    F. Reynard et al. / Virology 324 (2004) 90–10294

    fusion capacity was dependent on the correct processing of

    the Env glycoprotein and on its exportation to the cellular

    membrane. The number of syncytia decreased with the

    number of mutations, correlating with the gp160 cleavage

    impairment and edifice instability as described in previous

    section. Compared to the wild-type Env, the loss of one or

    Fig. 3. gp160 and gp120 immunoblots after 293T cells transfection. Detection was

    weight standard of migration (kDa). To normalize sample loading, an anti-h-actinsupernatant.

    sometimes two glycosylation sites (g3A, g3C, and g1) had

    little impact on syncytia formation. This biological assay is

    more sensitive than an immunoblot, and we noticed a fusion

    capacity even for some mutants for which no gp120 was

    detected on blot (i.e., g3A, g1 or g5, and in a lesser extent,

    g5B and g2). No fusion was observed for g4A, g5C, g3D

    (three mutations), g3 (five mutations), and g6B (6 muta-

    tions). For g6B and g5C, the gp120 was detected by

    Western blot in supernatant but not in cell lysate, suggesting

    that the protein was exported to the cell surface and the

    gp120 heavily shed. Apparently, the remaining gp120/gp41

    was not able to induce cell-to-cell fusion. Unexpectedly, the

    mutants g4 and g3B with three and four mutations, respec-

    tively, had a fusion capacity comparable to the wt. Glyco-

    sylations located near or within the V1/V2 or the V3

    variable loops could play an important role in coreceptor

    usage and CD4 dependence (Kolchinsky et al., 2001;

    Pollakis et al., 2001). We tested fusion on CD4+–CXCR4+

    cells and CD4�–CCR5+ cells (GHOST-CXCR4 and HOS-

    CCR5, respectively). In our hands, no mutant proteins

    induced fusion on these cells (data not shown).

    The exportation of Env to the cell membrane does not

    presume its incorporation into virions. In order to know if

    the mutant proteins could be incorporated with preserved

    functionality into viral particles, we generated pseudotyped

    particles with the different mutants and tested the capacity

    of these pseudoviruses to infect several GHOST cell lines

    (Fig. 4B). The envelopes for which no or few cell-to-cell

    fusion occurred showed no infection, neither on GHOST-

    CCR5 nor on GHOST-CXCR4. The results for g4B, g2C,

    g4C, g2D, and g5 were more ambiguous because no

    infection was noticed although cell-to-cell fusion assay gave

    syncytia. For mutants g3A and g1, the level of infection was

    unexpectedly weak considering their important fusion ca-

    pacity. These results were reproducible and indicated a

    difference between Env presented to the cell surface and

    Env incorporated in enveloped pseudovirions.

    Immunoblots carried out on pseudoparticles after sedi-

    mentation by ultracentrifugation on sucrose cushion con-

    firmed the previous results (Fig. 4C); the gp120 and the

    gp41 were detected only on pseudoviruses that infected

    indicator cells the most efficiently. For g2C and g4C, the

    gp41, but not the gp120, was detected.

    made with a polyclonal antiserum directed against gp120. MW: molecular

    monoclonal antibody was used (A; bottom). L: cell lysates; S: cell culture

  • Fig. 4. Env exportation to cell membrane, incorporation in pseudoparticles, and infectivity. (A) Cell-to-cell fusion assay. 293T cells expressing the different

    mutant genes were cocultivated with GHOST-CCR5 cells. (� ): fusion as on negative control, that is, < 5 syncytia/10 fields; (++): fusion equivalent to wildtype, that is, 60–100 syncytia/10 fields; (+) and (+/� ): intermediate levels of fusion, that is, 20–50 and 5–15 syncytia/10 fields, respectively. Resultscorrespond to the average of two independent experiments conducted in duplicate. (B) GHOST cells infections by pseudoparticles. Pseudotyped particles were

    incubated with GHOST-CCR5 cells and subsequent luminescence was read in cell lysates. Results correspond to the average of two experiments made in

    triplicate. (C) Purified pseudoparticles immunoblotted with a polyclonal antibody directed against gp120 (top) and gp41(bottom). Samples were normalized

    after p24 quantification.

    F. Reynard et al. / Virology 324 (2004) 90–102 95

    Influence of N-glycan removal on sCD4 binding

    The ability of Env to bind CD4 is an important feature

    that could be used to give information about the general

    conformation and the functionality of the mutants. Antigen-

    capture ELISAs were carried out on both cell lysates and

    culture supernatants of transfected 293T cells (Fig. 5). The

    samples were the same with those used for the Western

    blots. These ELISA were revealed with soluble CD4

    (sCD4)/anti-CD4 antibody. Mutants with important alter-

    ations such as g4A, g5B, g3D, or g2–g3 have not been

    taken into account. Compared with the gp120 detected in

    lysates, the signal in culture supernatants reflected both the

    cleavage efficiency and the instability of the gp120–gp41

    association. These results confirmed Western blot experi-

    ments (Fig. 3) but with more sensitivity. Taken as a whole,

    the removal of several glycosylation sites had little impact

    on sCD4 binding. However, for g6B-6 mutations (and in a

    lesser extent, for g3B-4 mutations), the decrease in super-

    natant signal was probably attributable to antigenicity

    disturbance because the relative amount of gp120 detected

    on blot was equivalent to gp120 detected for g1B or g4B

    (Fig. 3). When ELISA with serial dilutions of sCD4 were

    performed, some mutants with one mutation had a relative

    affinity better than the wild-type protein; the sCD4 half-

    maximum binding was 0.05 Ag/ml for mutants g2A, g1B,and g1C, and 0.15 Ag/ml for wild-type (data not shown).Removal of N243, N180/185, or N289 improved sCD4

    binding.

    In order to compare the respective effects of mutations on

    both sCD4 binding and overall antigenicity, another ELISA

    was carried out with the autologous serum from patient

    #133 (Fig. 5). The results were comparable to those

    obtained on the sCD4-ELISA. Mutations had little impact

    on sera recognition compared to native protein. No signif-

    icant difference was noticed between clustered and non-

    clustered mutants. Surprisingly, although the g2C, g4C, g5C

    mutants were relatively well recognized by serum, no signal

    was monitored for sCD4 binding probably because muta-

    tions affected mainly CD4 binding site. For g1 and g3A,

    despite the lack of gp120 detected in Western blot (Fig. 3),

    we found a little but significant signal in supernatant and

    lysate indicating that these mutants were cleaved and

    exportated.

    Neutralizing activity against pseudoparticles bearing Env

    glycomutants

    To ascertain the influence of the glycosylation on sensi-

    tivity to antibody-mediated neutralization, four sera speci-

    men from HIV-1-infected patients were tested for their

    ability to neutralize infectious pseudotyped particles in an

    in vitro neutralization assay using GHOST CD4+–CCR5+

    cell line. One serum was taken from patient #133 two years

    after the acute phase, and the three others corresponded to

    titrated neutralizing sera from a previous study (Burrer et al.,

    2001). These sera had a measurable neutralizing activity

    against several primary isolates. The results are displayed in

  • Fig. 5. Accessibility of the CD4 binding site. ELISAwas carried out both on lysates (A) and culture supernatants (B) from transient expression in 293T cells.

    Detection was made with sCD4/anti-CD4 antibody and with the autologous serum from patient #133. Results correspond to the average of two independent

    experiments conducted in duplicate. The threshold (horizontal line) was defined as 3 times the background read on the negative samples.

    F. Reynard et al. / Virology 324 (2004) 90–10296

    Table 2 as 50% inhibitory titer (IT50). The results were

    compared to those obtained for the parental env gene. The

    four neutralizing sera showed a greater inhibitory titer for

    two glycomutants than for the wild type; CCR5-mediated

    Table 2

    Neutralization of pseudoviruses with 1:2 serial dilutions of neutralizing and

    nonneutralizing human sera

    Pseudovirions Neutralizing sera

    1 2 3 4 5

    g2A 570 180 60 1300 20

    g1B 820 150 60 1300 80

    g3B 1350 1120 250 1600 80

    g1C 510 120 45 400 80

    g3C 490 150 25 800 50

    g1D 640 160 250 1300 80

    g1 1090 300 180 700 10

    g4 2500 2000 800 1600 30

    wt 720 210 45 750 80

    Values correspond to serum dilution required to yield z 50% reduction ofinfection by 8.104–105 RLU-equivalent promoting infection of pseudo-

    typed viruses. Results correspond to the average of two experiments

    performed in triplicate. The column #1 corresponds to the autologous serum

    from patient #133. The columns #2–4 correspond to titrated HIV-1 human-

    neutralizing sera. The column #5 corresponds to an HIV-1 negative serum.

    infection was neutralized more efficiently (Table 2, columns

    #1 to #4). These two mutants corresponded to the complete

    deglycosylation of the V1-loop (g4) and to four mutations in

    the blue cluster (g3B). The results displayed in the previous

    section showed that these mutants were structurally and

    functionally barely affected despite a large number of N-

    glycans removed. For g1D (one mutation in D cluster) and

    for g1 (two mutations but not in a cluster), a notable

    neutralization effect for some sera was noticed. Only a

    weak neutralizing activity was detected for HIV-1 negative

    serum (10 < IT50 < 80; column #5).

    Discussion

    In the present report, we aimed at the determination of

    the biological properties of Env glycomutants designed

    from the available 3D structure. We used HIV-1 acute

    infection primary isolates (PHI) to work with env sequences

    from the early stage of the disease, as close as possible to

    the transmission event. At this stage, viruses are still

    homogenous in sequence (Delwart et al., 2002). Moreover,

    the glycan shield could evolve during the course of infection

  • F. Reynard et al. / Virology 324 (2004) 90–102 97

    by an escape process (Wei et al., 2003a) and it seemed

    judicious to focus on primary sequences isolated early after

    the infection. Alignment of 14 PHI gp120 allowed us to

    select conserved glycosylation sites, which could be

    grouped into four regions of occupancy when laid on a

    3D model. We hypothesized that such a conservation and

    clustering could reflect a role in shielding crucial gp120

    epitopes during HIV-1 transmission. To check this hypoth-

    esis, we engineered gp120 glycomutants with progressive

    deglycosylation of each region. Each mutant was tested for

    both its functional and antigenic properties and compared to

    the wild-type protein.

    The number of sequences analyzed was not sufficient to

    be sure that the positions we chose as conserved are indeed

    found in all HIV-1 isolates, but we can assume that these

    positions are at least well conserved. Moreover, the ana-

    lyzed sequences were almost exclusively from B clade

    except two of them belonging to E and G clades (sequen-

    ces #355AHT and #365AHT, respectively). Even if some

    cohorts of acute seroconverters have been extensively

    studied (Delwart et al., 2002; Freel et al., 2003; Hu et

    al., 2000), they are few complete env sequences from PHI

    available in HIV-1 sequences databases. A comparison

    between our 14 gp120 PHI sequences and two other ones

    found in databases (GeneBank accession nos. U32396 and

    AF310108) showed that the N-glycan sites we selected

    were actually present in these two PHI sequences (data not

    shown). The repartition of the different conserved potential

    glycosylation sites on the 3D model showed that N-glycans

    could be located in specific regions. These regions are

    virtual because of the arbitrary nature of our clusters: as the

    available crystallized gp120 is partially deleted and its

    association with the sCD4 and 17b Fab probably induced

    conformational changes compared to gp120 alone, we

    considered alternate possibilities to enlarge the definition

    of each region. We chose to restrain the number of

    deglycosylation to six because it had been previously

    proposed that Env can tolerate around five mutations in

    combination without disturbance of the infectivity (Ohgi-

    moto et al., 1998; Reitter and Desrosiers, 1998).

    Transient expression of the env glycomutants in the 293T

    cells and subsequent Western blots analysis showed that

    cleavage of the precursor gp160 into gp120 and gp41 was

    affected by the number of N-glycans removed. Previous

    studies have shown that removing of one N-glycan has no

    impact on biosynthesis and functionality of gp160, but more

    mutations (3–5) have major effects (Dirckx et al., 1990;

    Fenouillet et al., 1994). In our hands, if one—or exception-

    ally more mutations (g3B, g3C, and g4)—had generally

    little impact on processing, the accumulation of three to six

    mutations entailed a dramatic decrease in gp160 cleavage

    and gp120/gp41 cohesion. This could be explained by the

    misfolding of the partially deglycosylated protein or the

    inefficient trafficking from endoplasmic reticulum to trans-

    Golgi, where the cleavage by cellular endoproteases occurs

    (Moulard and Decroly, 2000). These results are compatible

    with previously published data (Johnson et al., 2001;

    Quinones-Kochs et al., 2002; Reitter and Desrosiers,

    1998). Both the number and the specific location of the

    silenced N-glycans seemed to have additive effects. If some

    N-glycans (especially at positions N109 and N148) play a

    foreground role, others seem to be more accessory in terms

    of functionality and antigenicity, notably those located on

    the V1 variable loop and some in the B (e.g., N180/N185 or

    N201/N208) or in the C cluster (N289, N253).

    The positive results in cell-to-cell fusion assays indicated

    that sufficient level of functional Env was expressed on the

    cell surface. In this assay, fusion observed on GHOST-

    CCR5 cells was inversely related to the number of N-

    glycans removed. Previous studies have shown that all N-

    glycans on V1 or V2 loops on HIV-1 primary isolate could

    be mutated without affecting expression on the cell surface.

    However, mutations reduced (four or five sites) or sup-

    pressed (six sites) fusion activity (Quinones-Kochs et al.,

    2002). The N-glycans on the V1/V2 stem and V3 loops have

    often been described as playing an important role in HIV-1

    strains coreceptor usage (Nakayama et al., 1998; Pollakis et

    al., 2001). In our experimental conditions, no cell-to-cell

    fusion was observed for the mutations N44 (V1/V2 stem)

    and N148 (V3) when GHOST-CXCR4 cells were used as

    indicators.

    In order to characterize the effects of clustered N-glycans

    mutagenesis on CD4 binding site, sCD4 binding was tested

    by ELISA. It was barely affected by mutations. For some

    mutants (g2C, g4C, and g5C), mutation at position N109

    played an unfavorable role on sCD4 binding compared to

    the results obtained on some constructs without mutation at

    this position (g1C and g3C). Mutation N109 is located in

    the C2 domain, 30 residues upstream the V3 loop. It has

    been previously reported that N109Q substitution in HIV-1

    is critical for the intracellular process of gp160, for the

    ability of gp120 to bind to CD4, and for the infectivity by

    reduction in the amount of gp120 incorporated in virions. It

    has been proposed that such impairment could be due to

    interactions disturbance between C2 and V3 regions (Willey

    and Martin, 1993). Interestingly, the glycosylation site at

    N109 is exceptionally well conserved in all Env primate

    retroviruses (Kwong et al., 1998). Although g2C and g4C

    were dramatically affected for sCD4 binding, they induced

    syncytia formation in cell-to-cell fusion experiment on

    GHOST CD4+–CCR5+. It could be tempting to conclude

    that these mutants have acquired CD4 independence; how-

    ever, cell-to-cell fusion assay on HOS CD4�–CCR5+ did

    not show any syncytia.

    Results obtained with pseudovirions indicated that the

    incorporation efficiency depends on the number of glycans

    removed. The correct processing of the precursor gp160

    seems required for Env incorporation into viral particles

    (Dubay et al., 1995; McCune et al., 1988). The uncleaved

    form of the HIV-1LAI Env is highly presented at the cell

    surface but particles following budding from the cell surface

    are devoid of gp160 precursor (Moulard et al., 1999); other

  • F. Reynard et al. / Virology 324 (2004) 90–10298

    data suggested that the entire gp120 with an adequate

    conformation is required for Env complex incorporation

    into virions (Li and Perez, 1997).

    The mutants g3A and g1, despite cleavage alteration,

    induced a high number of syncytia and promoted infec-

    tious pseudoparticles formation. These results suggested

    that the few gp160 that is correctly processed at the cell

    membrane kept their functionality and could be incorpo-

    rated into virions. The differences observed between fusion

    assay and infectivity assay for mutants g2C and g4C could

    be also imputed to the low density of functional Env found

    on cell membranes, which has repercussions on virion

    envelope composition. Detection of gp41, but not gp120,

    on pseudovirions g2C and g4C provided the confirmation

    of N109 influence upon Env edifice stability; the not yet

    shedded gp120/gp41 edifices present at the cell surface

    (which induced fusion in cell-to-cell fusion assay) would

    be incorporated into pseudovirions, but once in pseudovi-

    rions, the gp120 would shed. The cell-to-cell fusion assay

    is a very sensitive test for monitoring Env functionality

    because in theory, a single functional Env oligomer must

    be sufficient to promote membranes fusion on susceptible

    cells and therefore to lead to the formation of a detectable

    fusion event. This could explain why some mutants

    showed cell fusion although the gp120 could not be

    detected by immunoblot.

    With one unique mutation at position N44, the mutant

    g1Awas correctly processed. Unexpectedly, a weak level of

    cell-to-cell fusion and infection was observed. Previous

    studies suggested that the glycan located at N44 influences

    the spacial location of the V1/V2 loop of HIV-1ADAglycoprotein and consequently modulates the accessibility

    of the coreceptor binding site (Kolchinsky et al., 2001). We

    propose that for g1A, the mutation relocates V1/V2 the

    same way but resulting in a reduced accessibility of the

    CCR5 binding site.

    Early studies showed that the acquisition or the removal

    of glycans in variable loops of HIV-1 or SIV modify the

    sensitivity of these viruses to neutralization (Bolmstedt et

    al., 1996; Chackerian et al., 1997; Matsushita et al., 1988).

    In our hands, the most remarkable results were obtained on

    mutants devoid of three N-glycans in V1 or 4 N-glycans in

    C3 (g4 and g3B, respectively). It has been suggested that the

    density of the envelope spikes on the surface of virions can

    influence relative sensitivity to antibody-mediated neutrali-

    zation. If mutations result in a decrease in spikes incorpo-

    ration or in a high level of spontaneous shedding, then the

    increased neutralization sensitivity could reflect a reduction

    in the number of antibodies required to inhibit mutant

    virions (Burton et al., 2000). However, immunoblots made

    on pseudotyped particles showed that neutralization sensi-

    tivity was independent on the intensity of the gp120 signal.

    For V1 deglycosylation (g4, three mutations), there is

    apparently a few differences comparing wt Env. However,

    g4 pseudotyped particles are more sensitive to human-

    neutralizing sera. The V1/V2 loop is a bulky domain that

    emerges from the gp120 core; moreover, V1 loop is struc-

    tured by three disulfide bridges: this particular topology

    could explain why deglycosylation led to small differences

    in structure and processing comparing parental glycopro-

    tein. However, a complete deglycosylation of the V2 loop

    (g5, 2 mutations) caused a drastic impairment for gp160

    cleavage, conformation, and CD4 binding. Both structures

    have complex-type sugar linked to asparagine (and an extra

    glycan not yet characterized, for V1). Apparently, the

    chemical nature of glycans does not explain differences

    observed between the two mutants.

    Thus, we can hypothesize that some sugars play an

    essential role during Env biosynthesis and processing

    (e.g., sugars linked at residues N109 and N148); the

    silencing of these sugars entails defects on cleavage or

    gp120/gp41 stability. A second type of sugar plays an

    accessory role in envelope processing but probably serves

    to shield protein against antibodies by evolving during the

    course of the disease. The sugars removed from g3C, g4,

    and g3B mutants would belong to this second type. This

    hypothesis agrees with a previous study showing that N-

    glycans important for infectivity were not randomly distrib-

    uted on HIV-1HXB2 gp120, and consequently, the role of the

    remaining N-glycans would be to evade the host immune

    response (Lee et al., 1992). Interestingly, N-glycan sites

    silenced on g4 and g3B are separated by few residues (less

    than eight amino acids) and could be considered as redun-

    dant. Moreover, the asparagine residues mutated for g3C or

    g3B locate close together on 3D model. This special

    topology strengthens the hypothesis of evolving and adapt-

    able positions for N-glycans shield in face of the antibody

    repertoire.

    Recent results suggested that high-mannose residues

    defining 2G12 epitope play a foreground role in DC-SIGN

    or related lectins binding (Sanders et al., 2002). The 2G12

    human monoclonal antibody neutralizes HIV-1 in vitro and

    protects macaque in association with others antibodies in

    passive immunization experiment from Simian-HIV-1

    (SHIV) virus challenge (Mascola et al., 2000; Trkola et

    al., 1996). The N-glycans N180–N185 from the B region

    take part in 2G12 epitope. They are silenced in g3B mutant.

    Thus, we can also hypothesize that in this area, the presence

    of a patch of N-glycans has a critical importance on virus–

    cell interactions. Consequently, antibodies directed against

    this region of Env could interfere with lectins binding and

    thus could allow better viruses neutralization.

    The differences between the mutants and wt protein are

    not the result of gross defect in expression or processing of

    the envelope complex because the majority of mutants were

    cleaved and exported to the cell surface. The cleavage and

    the conformation disturbance are apparently independent of

    clustering mutations or glycan type. However, results can be

    modulated in function of the respective position of each

    glycan removed on the gp120, underlining the different role

    of each sugar in the Env protein characteristics. Although

    our integrated approach, taking into account the 3D disper-

  • F. Reynard et al. / Virology 324 (2004) 90–102 99

    sion of N-glycans on gp120 from PHI, failed to clearly

    demonstrate a cluster-specific role of N-glycans in Env

    function and immunogenicity, it has led us to identify new

    potential immunogens. If the criteria in the choice of a Env

    immunogen suitable for neutralizing antibodies induction

    are based on the preservation of the structure as much as

    possible, but with an enhanced exposition of critical epito-

    pes and correlatively enhanced neutralization, the g3B and

    g4 mutants are good candidates.

    Materials and methods

    Sequence data

    Nucleotide sequences coding for the entire gp160 env

    region were generated from DNA PBMC from patients

    with acute HIV-1 infection. These sequences were de-

    scribed in a previous study (Ataman-Onal et al., 1999).

    They were named 133 AHM, 146 AHM, 153 AHM, 159

    AHM, 160 AHM, 374 AHM, 384 AHM, 385 AHM

    (homosexual transmission), 120 AHT, 309 AHT, 355

    AHT, 365 AHT, 373 AHT, and 426 AHT (heterosexual

    transmission). All sequences were aligned with CLUS-

    TALW (MacVector software, Oxford Molecular Group).

    The 3D model was based on the coordinates for the HIV-

    CD4-17bFab antibody complex deposited in the Protein

    Data Bank as entry IG9M. Molecules were drawn with

    PDBwiever (Glaxo-Smithkline).

    Site-directed mutagenesis

    Wild-type 133 env sequence was cloned into the Xho1–

    Xba1 sites of the pCI expression vector (Promega, Madison,

    USA) using standard protocols. To remove potential N-

    glycosylation sites, asparagine to glutamine substitutions

    were carried out using the QuickChange site-directed mu-

    tagenesis kit (Stratagene, La Jolla, USA). All mutant con-

    structs were checked by sequencing both strands on a 377

    automatic sequencer (Perkin-ABI, Boston, USA) with the

    PRISM Big Dye terminator V.02 (Perkin-ABI). Plasmid

    DNA was prepared using the NucleobondPC500 kit

    (Macherey-Nagel, Düren, Germany).

    Cells transfection and pseudovirions generation

    293T cells were used for transfection experiments and

    were cultivated in MEM (Gibco-LifeTech, Gaithersburg,

    USA) supplemented with 10% fetal calf serum, 1% nones-

    sential amino acids and 1% Na-pyruvate (Gibco-LifeTech).

    Cells (8.105) were cotransfected with 0.2 Ag pCI-rev and 1Ag of the different pCI-env constructs using the Lipofect-AMINE Kit (BRL, Rockville, USA). The env gene cloned

    in reverse orientation in the same vector (pCI-anti) was used

    as a negative control. Cells were harvested 24 h (for cell-to-

    cell fusion assay) or 48 h later (for both immunoblots and

    ELISA). Alternatively, pseudovirions stocks with wild-type

    or env mutants were generated as previously described with

    the help of the env-deficient viral reporter vector pNL43-

    LucR-E- (Park et al., 1998). Supernatants containing pseu-

    doviruses were harvested 48 h post-transfection and frozen

    at �80 jC. The amount of virus particles was determined byVidas-p24 tests (bioMerieux, Marcy-l’Etoile, France).

    In vitro cell-to-cell fusion assay and infectivity assay

    Twenty-four hours post-transfection, 293T cells were

    harvested and replated at 105 cells in a 12-well plate. The

    unseeded cells were used to check gp120 expression by an

    immunoblot. Cells were left to re-adhere during 6 h, then

    GHOST-CD4+CCR5+,GHOST-CD4+CXCR4+, or HOS-

    CD4�CCR5+ cells were added (106 cells/well) and incubat-

    ed during 16 h. The cells were fixed 15 min in 0.5%

    glutaraldehyde, colored in May–Grünwald/Giemsa stain,

    and syncytia were observed with an optical microscope.

    Alternatively, 106 GHOST cells were infected by pseudovi-

    ruses (50 ng p24/wells). After 12 h of incubation, inoculum

    was replaced by 1 ml of fresh culture media. After 72 h, the

    cells were lysed and luciferase activity was measured with

    the Luciferase Assay Kit (Promega) on a luminometer (TD

    20/20, Turner).

    Western blot

    The same protocol was used for both cell lysates, culture

    supernatants, and pseudoparticles pellets. Forty-height hours

    post-transfection, cells were harvested, washed once with

    PBS1x, lysed in TBS1x/NP40 1%, and finally cleared by

    centrifugation (15000 g/10 min). The culture supernatants

    were cleared of the cell debris by centrifugation (15000 g/10

    min). The pseudoparticles (equivalent to 500 ng of p24) were

    pelleted 3 h at 20 000 g on a 20% sucrose cushion and then

    lysed by 50 Al of TBS1x/NP40 1%. The proteins were heat-denatured in presence of h-mercaptoethanol and separatedon a 8% SDS-polyacrylamide gel. After electro-transfer, the

    membrane (Hybon-P; Amersham-Pharmacia, UK) was sat-

    urated in PBS1x/Tween 200.1% nonfat dry milk 3% and

    incubated overnight at 4 jC with a goat anti-gp120 poly-clonal antibody (Biogenesis ref. 5000–0557, Poole, UK; 1

    Ag/ml). Alternatively, an anti-gp41 monoclonal antibody(F240, NIH AIDS Research and Reference Reagent Pro-

    gram, Rockville, USA) was used at 1 Ag/ml. After extensivewashing, the membrane was incubated for 45 min at room

    temperature with peroxydase-conjugated antibodies (Jack-

    son, West Grove, USA; 0.1 Ag/ml). Detection was performedusing the ECL reagent (Amersham-Pharmacia; UK). Sam-

    ples were normalized by densitometry on a Typhoon 8600

    reader (Amersham-Pharmacia, UK) based on the detection of

    the Rev and Actin proteins on immunoblots with monoclonal

    antibodies against Rev (1F6D1B7; bioMérieux, Marcy-

    L’Etoile, France) and against human actin (A4700; Sigma-

    Aldrich, St. Louis, USA).

  • rology 324 (2004) 90–102

    ELISA

    Env ELISA-capture was carried out on cell-culture

    supernatants and cell lysates processed as described above.

    The D7324 polyclonal antibody (Aalto, Dublin, Ireland)

    was used as capture antibody as previously described

    (Brand et al., 2000). After normalization of total protein

    concentration with the BCA protein assay kit (Pierce,

    Rockford, USA), cell lysates were diluted at 1:10 in

    TBS/NP40 1%/New-Born Calf Serum 2%. For detection,

    the autologous HIV + serum from #133 patient (B clade;

    hôpital de la Croix-Rousse; Lyon, France) was used at 1/

    100 dilution. Alternatively, soluble CD4 (sCD4, NIH AIDS

    Research and Reference Reagent Program) was used at a

    final concentration of 0.5 Ag/ml. The soluble CD4 wasdetected with the BAF 379 monoclonal antibody (R&D,

    Abington, UK) directed against its V4 loop. Each result

    corresponded to the mean of two independent experiments

    made in duplicate.

    Neutralization assay

    Supernatant containing pseudotyped particles were pre-

    incubated for 1 h at 37 jC with four serial human-neutralizing sera or one nonneutralizing human serum

    (Burrer et al., 2001) before infection of GHOST-CCR5

    cells in triplicate wells in 96-well plates. The volume of

    supernatant loaded in each well corresponded to the quan-

    tity of pseudotyped particles necessary to promote 80000–

    100000 relative light units (RLU) after infection of per-

    missive cells. After an overnight incubation, the medium

    was replaced and incubation took place during additional 2

    days. After cell lysis, the infectivity was measured by using

    LucLite Plus kit (Perkin–Elmer) and luminescence was

    read on a TopCount apparatus (Packard Biosciences).

    Neutralizing titers were expressed as the reciprocal of the

    plasma dilution required to protect 50% of cells from

    pseudoviruses infection.

    F. Reynard et al. / Vi100

    Acknowledgments

    We thank Dr. Christophe Guillon and Dr. Yasemine

    Ataman-Önal for helpful discussion and manuscript reading.

    The sCD4 and the monoclonal antibody F240 (Dr. Posner)

    were obtained through the NIH AIDS Research and

    Reference Reagent Program. We thank C. Moog (INSERM

    U544, Strasbourg) for neutralizing and control sera. This

    work was supported by ANRS (#2003/006) and bioMérieux

    SA.

    References

    Ataman-Onal, Y., Coiffier, C., Giraud, A., Babic-Erceg, A., Biron, F.,

    Verrier, B., 1999. Comparison of complete env gene sequences from

    individuals with symptomatic primary HIV type 1 infection. AIDS Res.

    Hum. Retroviruses 15 (11), 1035–1039.

    Benjouad, A., Gluckman, J.C., Rochat, H., Montagnier, L., Bahraoui, E.,

    1992. Influence of carbohydrate moieties on the immunogenicity of

    human immunodeficiency virus type 1 recombinant gp160. J. Virol.

    66 (4), 2473–2483.

    Bolmstedt, A., Sjolander, S., Hansen, J.E., Akerblom, L., Hemming, A.,

    Hu, S.L., Morein, B., Olofsson, S., 1996. Influence of N-linked glycans

    in V4–V5 region of human immunodeficiency virus type 1 glycopro-

    tein gp160 on induction of a virus-neutralizing humoral response. J.

    Acquired Immune Defic. Syndr. Hum. Retrovirol. 12 (3), 213–220.

    Brand, D., Lemiale, F., Thibault, G., Verrier, B., Lebigot, S., Roingeard, P.,

    Buzelay, L., Brunet, S., Barin, F., 2000. Antigenic properties of recom-

    binant envelope glycoproteins derived from T-cell-line-adapted isolates

    or primary human immunodeficiency virus isolates and their relation-

    ship to immunogenicity. Virology 271 (2), 350–362.

    Burrer, R., Salmon-Ceron, D., Richert, S., Pancino, G., Spiridon, G., Haes-

    sig, S., Roques, V., Barre-Sinoussi, F., Aubertin, A.M., Moog, C., 2001.

    Immunoglobulin G (IgG) and IgA, but also nonantibody factors, ac-

    count for in vitro neutralization of human immunodeficiency virus

    (HIV) type 1 primary isolates by serum and plasma of HIV-infected

    patients. J. Virol. 75 (11), 5421–5424.

    Burton, D.R., Williamson, R.A., Parren, P.W., 2000. Antibody and virus:

    binding and neutralization. Virology 270 (1), 1–3.

    Cattaneo, R., Rose, J.K., 1993. Cell fusion by the envelope glycoproteins

    of persistent measles viruses which caused lethal human brain disease.

    J. Virol. 67 (3), 1493–1502.

    Chackerian, B., Rudensey, L.M., Overbaugh, J., 1997. Specific N-linked

    and O-linked glycosylation modifications in the envelope V1 domain of

    simian immunodeficiency virus variants that evolve in the host alter

    recognition by neutralizing antibodies. J. Virol. 71 (10), 7719–7727.

    Cheng-Mayer, C., Brown, A., Harouse, J., Luciw, P.A., Mayer, A.J., 1999.

    Selection for neutralization resistance of the simian/human immunode-

    ficiency virus SHIVSF33A variant in vivo by virtue of sequence

    changes in the extracellular envelope glycoprotein that modify N-linked

    glycosylation. J. Virol. 73 (7), 5294–5300.

    Delwart, E., Magierowska, M., Royz, M., Foley, B., Peddada, L., Smith,

    R., Heldebrant, C., Conrad, A., Busch, M., 2002. Homogeneous qua-

    sispecies in 16 out of 17 individuals during very early HIV-1 primary

    infection. AIDS 16 (2), 189–195.

    Desrosiers, R.C., Lifson, J.D., Gibbs, J.S., Czajak, S.C., Howe, A.Y.,

    Arthur, L.O., Johnson, R.P., 1998. Identification of highly attenuat-

    ed mutants of simian immunodeficiency virus. J. Virol. 72 (2),

    1431–1437.

    Dirckx, L., Lindemann, D., Ette, R., Manzoni, C., Moritz, D., Mous, J.,

    1990. Mutation of conserved N-glycosylation sites around the CD4-

    binding site of human immunodeficiency virus type 1 GP120 affects

    viral infectivity. Virus Res. 18 (1), 9–20.

    Dubay, J.W., Dubay, S.R., Shin, H.J., Hunter, E., 1995. Analysis of the

    cleavage site of the human immunodeficiency virus type 1 glycoprotein:

    requirement of precursor cleavage for glycoprotein incorporation. J.

    Virol. 69 (8), 4675–4682.

    Fennie, C., Lasky, L.A., 1989. Model for intracellular folding of the human

    immunodeficiency virus type 1 gp120. J. Virol. 63 (2), 639–646.

    Fenouillet, E., Gluckman, J.C., Jones, I.M., 1994. Functions of HIV enve-

    lope glycans. Trends Biochem. Sci. 19 (2), 65–70.

    Fischer, P.B., Karlsson, G.B., Dwek, R.A., Platt, F.M., 1996. N-butyldeox-

    ynojirimycin-mediated inhibition of human immunodeficiency virus

    entry correlates with impaired gp120 shedding and gp41 exposure. J.

    Virol. 70 (10), 7153–7160.

    Freel, S.A., Fiscus, S.A., Pilcher, C.D., Menezes, P., Giner, J., Patrick, E.,

    Lennox, J.L., Hicks, C.B., Jr. J.J., Shugars, D.C., 2003. Envelope di-

    versity, coreceptor usage and syncytium-inducing phenotype of HIV-1

    variants in saliva and blood during primary infection. AIDS 17 (14),

    2025–2033.

    Hemming, A., Gram, G.J., Bolmstedt, A., Losman, B., Hansen, J.E., Ricks-

    ten, A., Olofsson, S., 1996. Conserved N-linked oligosaccharides of the

  • F. Reynard et al. / Virology 324 (2004) 90–102 101

    C-terminal portion of human immunodeficiency virus type 1 gp120 and

    viral susceptibility to neutralizing antibodies. Arch. Virol. 141 (11),

    2139–2151.

    Hu, Q.X., Barry, A.P., Wang, Z.X., Connolly, S.M., Peiper, S.C., Green-

    berg, M.L., 2000. Evolution of the human immunodeficiency virus type

    1 envelope during infection reveals molecular corollaries of specificity

    for coreceptor utilization and AIDS pathogenesis. J. Virol. 74 (24),

    11858–11872.

    Johnson, W.E., Sauvron, J.M., Desrosiers, R.C., 2001. Conserved, N-

    linked carbohydrates of human immunodeficiency virus type 1 gp41

    are largely dispensable for viral replication. J. Virol. 75 (23),

    11426–11436.

    Kolchinsky, P., Kiprilov, E., Bartley, P., Rubinstein, R., Sodroski, J., 2001.

    Loss of a single N-linked glycan allows CD4-independent human im-

    munodeficiency virus type 1 infection by altering the position of the

    gp120 V1/V2 variable loops. J. Virol. 75 (7), 3435–3443.

    Kwong, P.D., Wyatt, R., Robinson, J., Sweet, R.W., Sodroski, J., Hendrick-

    son, W.A., 1998. Structure of an HIV gp120 envelope glycoprotein in

    complex with the CD4 receptor and a neutralizing human antibody.

    Nature 393 (6686), 648–659.

    Lee, W.R., Syu, W.J., Du, B., Matsuda, M., Tan, S., Wolf, A., Essex, M.,

    Lee, T.H., 1992. Nonrandom distribution of gp120 N-linked glycosyl-

    ation sites important for infectivity of human immunodeficiency virus

    type 1. Proc. Natl. Acad. Sci. U.S.A. 89 (6), 2213–2217.

    Leonard, C.K., Spellman, M.W., Riddle, L., Harris, R.J., Thomas, J.N.,

    Gregory, T.J., 1990. Assignment of intrachain disulfide bonds and char-

    acterization of potential glycosylation sites of the type 1 recombinant

    human immunodeficiency virus envelope glycoprotein (gp120)

    expressed in Chinese hamster ovary cells. J. Biol. Chem. 265 (18),

    10373–10382.

    Li, Y.Y., Perez, L.G., 1997. The entire SU subunit is required for the

    incorporation of the HIV-1 envelope glycoprotein complex into virions.

    Virus Genes 14 (3), 211–223.

    Li, Y., Luo, L., Rasool, N., Kang, C.Y., 1993. Glycosylation is necessary

    for the correct folding of human immunodeficiency virus gp120 in CD4

    binding. J. Virol. 67 (1), 584–588.

    Mascola, J.R., Stiegler, G., VanCott, T.C., Katinger, H., Carpenter, C.B.,

    Hanson, C.E., Beary, H., Hayes, D., Frankel, S.S., Birx, D.L., Lewis,

    M.G., 2000. Protection of macaques against vaginal transmission of a

    pathogenic HIV-1/SIV chimeric virus by passive infusion of neutraliz-

    ing antibodies. Nat. Med. 6 (2), 207–210.

    Matsushita, S., Robert-Guroff, M., Rusche, J., Koito, A., Hattori, T., Hosh-

    ino, H., Javaherian, K., Takatsuki, K., Putney, S., 1988. Characteriza-

    tion of a human immunodeficiency virus neutralizing monoclonal

    antibody and mapping of the neutralizing epitope. J. Virol. 62 (6),

    2107–2114.

    McCune, J.M., Rabin, L.B., Feinberg, M.B., Lieberman, M., Kosek, J.C.,

    Reyes, G.R., Weissman, I.L., 1988. Endoproteolytic cleavage of gp160

    is required for the activation of human immunodeficiency virus. Cell 53

    (1), 55–67.

    McKeating, J.A., McKnight, A., Moore, J.P., 1991. Differential loss of

    envelope glycoprotein gp120 from virions of human immunodeficiency

    virus type 1 isolates: effects on infectivity and neutralization. J. Virol.

    65 (2), 852–860.

    Moore, J.P., Cao, Y., Qing, L., Sattentau, Q.J., Pyati, J., Koduri, R., Rob-

    inson, J. , Barbas III, C.F., Burton, D.R., Ho, D.D. 1995. Primary

    isolates of human immunodeficiency virus type 1 are relatively resistant

    to neutralization by monoclonal antibodies to gp120, and their neutral-

    ization is not predicted by studies with monomeric gp120. J. Virol. 69

    (1), 101–109.

    Moulard, M., Decroly, E., 2000. Maturation of HIV envelope glycoprotein

    precursors by cellular endoproteases. Biochim. Biophys. Acta 1469 (3),

    121–132.

    Moulard, M., Hallenberger, S., Garten, W., Klenk, H.D., 1999. Processing

    and routage of HIV glycoproteins by furin to the cell surface. Virus Res.

    60 (1), 55–65.

    Nakayama, E.E., Shioda, T., Tatsumi, M., Xin, X., Yu, D., Ohgimoto, S.,

    Kato, A., Sakai, Y., Ohnishi, Y., Nagai, Y., 1998. Importance of the N-

    glycan in the V3 loop of HIV-1 envelope protein for CXCR-4- but not

    CCR-5-dependent fusion. FEBS Lett. 426 (3), 367–372.

    Ogert, R.A., Lee, M.K., Ross, W., Buckler-White, A., Martin, M.A., Cho,

    M.W., 2001. N-linked glycosylation sites adjacent to and within the V1/

    V2 and the V3 loops of dual tropic human immunodeficiency virus type

    1 isolate DH12 gp120 affect coreceptor usage and cellular tro-

    pism. J. Virol. 75 (13), 5998–6006.

    Ohgimoto, S., Shioda, T., Mori, K., Nakayama, E.E., Hu, H., Nagai, Y.,

    1998. Location-specific, unequal contribution of the N glycans in

    simian immunodeficiency virus gp120 to viral infectivity and removal

    of multiple glycans without disturbing infectivity. J. Virol. 72 (10),

    8365–8370.

    Pantophlet, R., Ollmann Saphire, E., Poignard, P., Parren, P.W., Wilson,

    I.A., Burton, D.R., 2003. Fine mapping of the interaction of neutral-

    izing and nonneutralizing monoclonal antibodies with the CD4 bind-

    ing site of human immunodeficiency virus type 1 gp120. J. Virol. 77

    (1), 642–658.

    Park, E.J., Vujcic, L.K., Anand, R., Theodore, T.S., Quinnan Jr., G.V.,

    1998. Mutations in both gp120 and gp41 are responsible for the broad

    neutralization resistance of variant human immunodeficiency virus type

    1 MN to antibodies directed at V3 and non-V3 epitopes. J. Virol. 72 (9),

    7099–7107.

    Pollakis, G., Kang, S., Kliphuis, A., Chalaby, M.I., Goudsmit, J., Paxton,

    W.A., 2001. N-linked glycosylation of the HIV type-1 gp120 envelope

    glycoprotein as a major determinant of CCR5 and CXCR4 coreceptor

    utilization. J. Biol. Chem. 276 (16), 13433–13441.

    Quinones-Kochs, M.I., Buonocore, L., Rose, J.K., 2002. Role of N-linked

    glycans in a human immunodeficiency virus envelope glycoprotein:

    effects on protein function and the neutralizing antibody response.

    J. Virol. 76 (9), 4199–4211.

    Reitter, J.N., Desrosiers, R.C., 1998. Identification of replication-competent

    strains of simian immunodeficiency virus lacking multiple attachment

    sites for N-linked carbohydrates in variable regions 1 and 2 of the

    surface envelope protein. J. Virol. 72 (7), 5399–5407.

    Rizzuto, C.D., Wyatt, R., Hernandez-Ramos, N., Sun, Y., Kwong, P.D.,

    Hendrickson, W.A., Sodroski, J., 1998. A conserved HIV gp120 gly-

    coprotein structure involved in chemokine receptor binding. Science

    280 (5371), 1949–1953.

    Sanders, R.W., Vesanen, M., Schuelke, N., Master, A., Schiffner, L., Kalya-

    naraman, R., Paluch, M., Berkhout, B., Maddon, P.J., Olson, W.C., Lu,

    M., Moore, J.P., 2002. Stabilization of the soluble, cleaved, trimeric

    form of the envelope glycoprotein complex of human immunodeficien-

    cy virus type 1. J. Virol. 76 (17), 8875–8889.

    Scanlan, C.N., Pantophlet, R., Wormald, M.R., Ollmann Saphire, E.,

    Stanfield, R., Wilson, I.A., Katinger, H., Dwek, R.A., Rudd, P.M.,

    Burton, D.R., 2002. The broadly neutralizing anti-human immunode-

    ficiency virus type 1 antibody 2G12 recognizes a cluster of alpha1!2mannose residues on the outer face of gp120. J. Virol. 76 (14),

    7306–7321.

    Schonning, K., Joost, M., Gram, G.J., Machuca, R., Nielsen, C., Niel-

    sen, J.O., Hansen, J.E., 1998. Chemokine receptor polymorphism and

    autologous neutralizing antibody response in long-term HIV-1 infec-

    tion. J. Acquired Immune Defic. Syndr. Hum. Retrovirol. 18 (3),

    195–202.

    Trkola, A., Purtscher, M., Muster, T., Ballaun, C., Buchacher, A., Sullivan,

    N., Srinivasan, K., Sodroski, J., Moore, J.P., Katinger, H., 1996. Human

    monoclonal antibody 2G12 defines a distinctive neutralization epitope

    on the gp120 glycoprotein of human immunodeficiency virus type 1.

    J. Virol. 70 (2), 1100–1108.

    Wei, X., Decker, J.M., Wang, S., Hui, H., Kappes, J.C., Wu, X., Sal-

    azar-Gonzalez, J.F., Salazar, M.G., Kilby, J.M., Saag, M.S., Komar-

    ova, N.L., Nowak, M.A., Hahn, B.H., Kwong, P.D., Shaw, G.M.,

    2003a. Antibody neutralization and escape by HIV-1. Nature 422

    (6929), 307–312.

    Wei, X., Decker, J.M., Wang, S., Hui, H., Kappes, J.C., Wu, X., Salazar-

    Gonzalez, J.F., Salazar, M.G., Michael Kilby, J., Saag, M.S., Komarova,

  • F. Reynard et al. / Virology 324 (2004) 90–102102

    N.L., Nowak, M.A., Hahn, B.H., Kwong, P.D., Shaw, G.M., 2003b.

    Corrigendum: antibody neutralization and escape by HIV-1. Nature 423

    (6936), 197.

    Willey, R.L., Martin, M.A., 1993. Association of human immunodeficiency

    virus type 1 envelope glycoprotein with particles depends on interac-

    tions between the third variable and conserved regions of gp120. J.

    Virol. 67 (6), 3639–3643.

    Zhu, X., Borchers, C., Bienstock, R.J., Tomer, K.B., 2000. Mass spectro-

    metric characterization of the glycosylation pattern of HIV-gp120

    expressed in CHO cells. Biochemistry 39 (37), 11194–11204.

    HIV-1 acute infection env glycomutants designed from 3D model: effects on processing, antigenicity, and neutralization sensitivityIntroductionResultsRational of deglycosylation constructsInfluence of N-glycans removal on gp160 cleavage and gp120 sheddingInfluence of N-glycans removal on cell membrane presentation and incorporation in pseudovirionInfluence of N-glycan removal on sCD4 bindingNeutralizing activity against pseudoparticles bearing Env glycomutants

    DiscussionMaterials and methodsSequence dataSite-directed mutagenesisCells transfection and pseudovirions generationIn vitro cell-to-cell fusion assay and infectivity assayWestern blotELISANeutralization assay

    AcknowledgementsReferences