Skip to main content

Main menu

  • Home
  • Articles
    • Current Issue
    • Next in The JI
    • Archive
    • Brief Reviews
    • Pillars of Immunology
    • Translating Immunology
    • Most Read
    • Top Downloads
    • Annual Meeting Abstracts
  • COVID-19/SARS/MERS Articles
  • Info
    • About the Journal
    • For Authors
    • Journal Policies
    • Influence Statement
    • For Advertisers
  • Editors
  • Submit
    • Submit a Manuscript
    • Instructions for Authors
    • Journal Policies
  • Subscribe
    • Journal Subscriptions
    • Email Alerts
    • RSS Feeds
    • ImmunoCasts
  • More
    • Most Read
    • Most Cited
    • ImmunoCasts
    • AAI Disclaimer
    • Feedback
    • Help
    • Accessibility Statement
  • Other Publications
    • American Association of Immunologists
    • ImmunoHorizons

User menu

  • Subscribe
  • Log in

Search

  • Advanced search
The Journal of Immunology
  • Other Publications
    • American Association of Immunologists
    • ImmunoHorizons
  • Subscribe
  • Log in
The Journal of Immunology

Advanced Search

  • Home
  • Articles
    • Current Issue
    • Next in The JI
    • Archive
    • Brief Reviews
    • Pillars of Immunology
    • Translating Immunology
    • Most Read
    • Top Downloads
    • Annual Meeting Abstracts
  • COVID-19/SARS/MERS Articles
  • Info
    • About the Journal
    • For Authors
    • Journal Policies
    • Influence Statement
    • For Advertisers
  • Editors
  • Submit
    • Submit a Manuscript
    • Instructions for Authors
    • Journal Policies
  • Subscribe
    • Journal Subscriptions
    • Email Alerts
    • RSS Feeds
    • ImmunoCasts
  • More
    • Most Read
    • Most Cited
    • ImmunoCasts
    • AAI Disclaimer
    • Feedback
    • Help
    • Accessibility Statement
  • Follow The Journal of Immunology on Twitter
  • Follow The Journal of Immunology on RSS

CCR5-Reactive Antibodies in Seronegative Partners of HIV-Seropositive Individuals Down-Modulate Surface CCR5 In Vivo and Neutralize the Infectivity of R5 Strains of HIV-1 In Vitro

Lucia Lopalco, Claudia Barassi, Claudia Pastori, Renato Longhi, Samuele E. Burastero, Giuseppe Tambussi, Francesco Mazzotta, Adriano Lazzarin, Mario Clerici and Antonio G. Siccardi
J Immunol March 15, 2000, 164 (6) 3426-3433; DOI: https://doi.org/10.4049/jimmunol.164.6.3426
Lucia Lopalco
*Department of Biological and Technological Research and Infectious Diseases Clinic, San Raffaele Scientific Institute, Milano, Italy;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Claudia Barassi
*Department of Biological and Technological Research and Infectious Diseases Clinic, San Raffaele Scientific Institute, Milano, Italy;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Claudia Pastori
*Department of Biological and Technological Research and Infectious Diseases Clinic, San Raffaele Scientific Institute, Milano, Italy;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Renato Longhi
†Istituto Biocatalisi e Riconoscimento Molecolare, Consiglio Nazionale delle Ricerche, Milano, Italy;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Samuele E. Burastero
*Department of Biological and Technological Research and Infectious Diseases Clinic, San Raffaele Scientific Institute, Milano, Italy;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Giuseppe Tambussi
*Department of Biological and Technological Research and Infectious Diseases Clinic, San Raffaele Scientific Institute, Milano, Italy;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Francesco Mazzotta
‡Malattie Infettive, Ospedale S.M. Annunziata, Firenze, Italy;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Adriano Lazzarin
*Department of Biological and Technological Research and Infectious Diseases Clinic, San Raffaele Scientific Institute, Milano, Italy;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Mario Clerici
§Cattedra di Immunologia, Università di Milano, Milano, Italy; and
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Antonio G. Siccardi
¶Dipartimento di Biologia e Genetica per le Scienze Mediche, Milano, Italy
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Exposure to HIV does not necessarily results in infection. Because primary HIV infection is associated with CCR5-tropic HIV variants (R5), CCR5-specific Abs in the sera of HIV-seronegative, HIV-exposed individuals (ESN) might be associated with protection against infection. We analyzed sera from ESN, their HIV-infected sexual partners (HIV+), and healthy controls (USN) searching for CCR5-specific Abs, studying whether incubation of PBMC with sera could prevent macrophage inflammatory protein 1β (Mip1β) (natural ligand of CCR5) binding to CCR5. Results showed that Mip1β binding to CCR5 was not modified by sera of either 40 HIV+ or 45 USN but was greatly reduced by sera of 6/48 ESN. Binding inhibition was due to Abs reactive with CCR5. The CCR5-specific Abs neutralized the infectivity of primary HIV isolates obtained from the corresponding HIV+ partners and of R5-primary HIV strains, but not that of CXCR4-tropic or amphitropic HIV strains. Immunoadsorption on CCR5-transfected, but not on CXCR4-transfected, cells removed CCR5-specific and virus-neutralizing Abs. Epitope mapping on purified CCR5-specific Abs showed that these Abs recognize a conformational epitope in the first cysteine loop of CCR5 (aa 89–102). Affinity-purified anti-CCR5-peptide neutralized the infectivity of R5 strains of HIV-1. Anti-CCR5 Abs inhibited Mip1β-induced chemotaxis of PBMC from healthy donors. PBMC from two ESN (with anti-CCR5 Abs) were CCR5-negative and could not be stimulated by Mip1β in chemotaxis assays. These results contribute to clarifying the phenomenon of immunologic resistance to HIV and may have implications for the development of a protective vaccine.

During the last few years, documentary evidence of the existence of individuals who remain HIV-seronegative and are apparently uninfected despite multiple exposures to HIV-1 (exposed seronegative, ESN)3 has been produced by a number of groups, suggesting that it is possible to achieve some degree of resistance to the virus. Some ESN show signs of unconventional HIV-related immunity, including 1) Abs to cellular Ags, including HLA class I and CD4 (1, 2, 3), 2) IgA-mediated and mucosally confined HIV humoral immune responses (4, 5), 3) HIV-specific systemic cell-mediated immunity (6, 7, 8) as well as production of type 1 cytokines (9, 10, 11) and β-chemokines (12) by HIV peptide-stimulated PBMC, and 4) HIV-specific CD8+ T lymphocyte-mediated cytotoxicity directed toward early HIV proteins (13, 14, 15).

Anti-cell Abs and T cell-mediated immunity have been associated with protection in primate models of lentivirus infection (16, 17, 18), but the role of these immune responses in humans is still unclear. More recently, it was shown that the cells of some ESN couldn’t be infected in vitro by M-tropic (R5) strains of HIV because they lack the essential CCR5 coreceptor (19, 20). In the CCR5 gene, at least two mutations (20, 21) have been associated with total or partial resistance to infection by M-tropic R5 strains of HIV, and one mutation was associated with slowing progression of the disease (22). R5 strains (23) account for most of the transmission of HIV infections (particularly sexually transmitted infections) and are associated with the earlier phases of the disease (24). Because CCR5-defective individuals were described to have normal inflammatory and immune reactions, CCR5 has been interpreted as a redundant molecule in adults, and has thus become an important potential target for blocking drugs (25) and immune modulation (26, 27, 28). Therefore, we searched for anti-CCR5 autoantibodies in the sera of ESN individuals on the assumption that, if such Abs exist, they are unlikely to affect immune function but may interfere with the HIV coreceptor function of CCR5.

Materials and Methods

Study population

Two different cohorts of monogamous couples discordant for HIV serostatus were selected for the study. A first cohort was enrolled in Milan (3 homosexual and 37 heterosexual couples); a second group was enrolled in Florence (2 homosexual and 6 heterosexual couples). The 48 ESN subjects included in the study were 26 heterosexual females, 17 heterosexual males, and 5 homosexual males (passive recipients). Written informed consent was obtained from all the participants. The inclusion criterion was a history of penetrative sexual intercourse without condom, at least twice per week and for at least 2 years with no other known risk factors during that period or afterward. HIV-1 infection was excluded because of the absence of anti HIV-1 Abs (ELISA and Western blot) and viral DNA (PCR). Seronegativity was confirmed 6 mo after enrollment. Forty-five serum samples from unexposed, sexually active, seronegative individuals (USN) (29 females, 16 males) were used as negative controls. Forty serum samples from HIV-seropositive partners of ESN subjects were also analyzed. After enrollment in the protocol, counseling was offered to all of the couples and many (but not all) switched to safe-sex practices. The ESN were characterized for the presence of CCR5-Δ32 alleles (courtesy of Drs. V. Nardese and P. Lusso), anti-CD4 Abs, and HIV-specific IgA (3, 5).

Purification of CD4+ cells

PBMC from healthy blood donors were isolated by Ficoll-Hypaque centrifugation and stimulated for 3 days with PHA (3 μg/ml) (Sigma-Aldrich, Steinheim, Germany) and rIL-2 (100 U/ml) (Amersham, Buchinghamshire, U.K.). CD4+ cells were purified from activated PBMC by immunoadsorption to anti-CD4 magnetic beads (Oxoid, Hampshire, U.K.). Purified CD4+ cells were stimulated for 3 days with IL-2 before the macrophage inflammatory protein 1β (Mip1β) binding assay. PBMC were characterized on the basis of the presence of CCR5-Δ32 alleles; only PBMC from donors not carrying the CCR5-Δ32 allele were used.

Mip1β binding assay

The assay was performed as described (29). Briefly, 106 purified CD4+ cells in 200 μl of RPMI 1640 (Life Technologies, Milan, Italy) (containing 0.05 M NaN3, 1% BSA, and 25 mM HEPES) were incubated with appropriate dilutions of sera and/or Ig-enriched fractions; after 45 min of incubation, 125I-Mip1β (DuPont-NEN, Mechelem, Belgium) was added (final concentration, 0.1 nM, 0.2 μCi), and the cells were further incubated for 2 h on ice. Unbound radioactivity was separated by centrifugation on a two-step gradient (30) in 0.3-ml tubes (Nunc, Roskilde, Denmark) as follows: the lower layer consisted of FCS containing 10% sucrose; the upper layer consisted of 80% silicone (Sigma-Aldrich) and 20% mineral oil (Sigma-Aldrich). The bound radioactivity in the cell pellets was measured in a gamma counter. Serum samples were diluted 1:10 (five replicas for each sample). A specificity control consisting of a 100-fold excess of unlabeled Mip1β was included in all experiments. The binding of the 125I-Mip1β to activated CD4+ cells ranged between 1000 and 6000 cpm. The cut-off value was set at 12% (three SD above the mean value of the 45 USN serum samples).

Anti-CCR5 Ab assay

Anti-CCR5 Abs bound to CCR5-positive cells were revealed by 0.5 μCi/ml of 125I-sheep anti human Ig F(ab′)2 Ab fragments (Amersham). Activated CD4+ T cells were treated with human sera diluted 1/10 and then processed as described for the Mip1β binding assay. The cut-off value (1065 cpm) was established as 3 SD above the mean of 10 USN serum samples. Cellular suspensions from CXCR4- or CCR5-transfected U87 cells monolayers pretreated with human sera diluted 1/10 were processed as described above. A pool of 10 USN sera was used as negative control, while 0.1 nM 125I-Mip1β and the anti-CCR5 mouse mAb 2D7 (31) were used as positive controls. The mAb was used at 0.3 μg/ml and revealed by 0.5 μCi/ml 125I-sheep anti-mouse IgG F(ab′)2 Ab fragments (Amersham).

Chemotaxis assay

PBMC from one healthy donor (selected for high expression of CCR5) were activated with PHA and IL-2 for 3 days (see above) in the presence of two concentrations of purified Igs (250 and 62 μg/ml) from ESN34, ESN55, and USN5. Then, 3 × 105 activated PBMC in 50 μl of RPMI 1640 medium containing 0.3% human serum albumin were placed in the upper chamber of 5-μm pore size bare filter Transwell (Costar, Europe, Amsterdam, The Netherlands). Chemotaxis was conducted in the presence of 1.5 μg/ml of Mip1β (placed in the lower chamber). The transwells were incubated for 2 h at 37°C; cells that migrated from the upper to the lower chamber were then quantified by FACS analysis. PBMC from ESN34 and ESN55 were also used in chemotaxis assays to evaluate the capacity of ESN PBMC to migrate in the presence of Mip1β. The results were expressed as chemotaxis index, which represents the fold increase in the number of migrated cells in response to Mip1β over the spontaneous cell migration in control medium.

C-C chemokines assay

Serum chemokines concentrations (RANTES, Mip1α, and Mip1β) were determined with commercial ELISA kits (R&D Systems, Minneapolis, MN).

Ig purification

Anti-human polyvalent Ig-coupled Agarose (Sigma-Aldrich) was used to purify total Ig from the sera of ESN and USN. Briefly, 100 μl of serum were incubated overnight at 4°C in columns containing 5 ml of anti-human Ig-agarose. After recovering the column washout (Ig-depleted fraction), the columns were washed six times in phosphate buffer (0.01 M with 0.5 M NaCl). Bound Igs were eluted with glycine/NaCl 0.2 M, and the eluted fractions neutralized with 1 M Tris (Ig-enriched fraction). Ig-enriched and Ig-depleted fractions were concentrated on Ultrafree-15 Biomax 30 membranes (Millipore, Bedford, MA) with a cut-off of 30 kDa and dialyzed against RPMI 1640. Ig concentration was determined by ELISA using commercial Igs as standard and adjusted to 2.5 mg/ml (corresponding to a serum dilution of 1:10). The Ig-depleted fractions were diluted by the same factor.

Virus isolation and titration

HIV was isolated from the PBMC of HIV-seropositive partners by cocultivation with PHA-stimulated PBMC of two USN. Cultures were maintained until increasing levels of HIV-p24 Ag were detected in two consecutive determinations. The infectivity (ID50) of each virus isolate was determined on PBMC from one single donor as follows: six replicas (150 μl) of 5-fold serial dilutions (from 1:5 to 1:3, 125) of virus were added to six wells of a round-bottom Microtiter plate (Nunc) containing 105 resting PBMC in 75 μl of medium, incubated for 2 h, washed, and resuspended in RPMI 1640 medium containing PHA (3 μg/ml) and 10 U/ml rIL-2. HIV-1 p24 Ag was titrated after 5, 7, and 9 days of culture by standard assays (32). ID50 titers were defined as the reciprocal of the virus dilution yielding 50% positive wells (Reed-Muench calculation).

Phenotypic characterization of viral coreceptor usage

Each virus isolate was used to infect U87 human glioma cell lines (provided by Dr. P. Lusso) expressing one of the following chemokine receptors: CCR1, CCR2B, CCR3, CCR5, or CXCR4. Then, 1 ml of virus-containing culture supernatant was incubated with 8 × 104 U87 cells in 12-wells plates for 4 h at 37°C. Cells were washed twice and incubated in RPMI 1640 medium supplemented with 10% FCS. Cell cultures were observed daily for cytopathic effect. Culture supernatants, collected from each well 2, 5, and 7 days after infection, were analyzed for HIV-1 p24 Ag.

Virus neutralization assays

The “resting cell assay” was performed according to Zolla-Pazner (33). Briefly, 2 × 105 resting PBMC were added to 75 μl of serial dilutions of Ig-enriched fractions from ESN or USN; after 1 h incubation, 75 μl of a virus dilution (ID50 adjusted to 20) was added. The cultures were incubated for another 2 h, washed, and resuspended in PHA and IL-2-containing medium. HIV-1 p24 Ag in the supernatants was determined on days 7 and 9. Percent neutralization was calculated relatively to a nontreated control.

For the “activated PBMC assay,” the cells were cultured in medium containing PHA and IL2 for 48 h before the neutralization assay.

Mip1β ELISA assay

First, 100 μl of recombinant human Mip1β (1 mg/ml) (R&D Systems) was coated on 96 Microplate wells in 50 mM NaHCO3-Na2CO3 buffer (2 h at 37°C). After overcoating with PBS buffer plus 10% BSA and 1% Tween 20 (1 h at 37°C), serum from ESN and USN in various dilutions (from 1/5 to 1/40) were added (1 h at 37°C). Goat anti-human Mip1β IgG Abs at 300–200-100–50 pg/ml were used as positive control. After four washes in PBS buffer, 100 μl of 1:1000 peroxidase-conjugated rabbit anti-human IgG or rabbit anti-goat IgG (Dako, Copenhagen, Denmark) were added to the wells and incubated (30 min at 37°C). The wells were washed seven times in PBS; substrate buffer containing o-phenylenediamine plus H2O2 was then added, and absorbance was determined at 492 nm.

Adsorption of purified Igs on U87 cell lines

Ig-enriched fractions from ESN and USN were adsorbed on monolayers of CXCR4- or CCR5-transfected U87 cell lines obtained by seeding 8 × 104 cells in Microtiter wells; 100 μl of Ig-enriched fractions (2.5 mg Ig/ml) were immunoadsorbed on the cell monolayers (10 min at 37°C); the liquid was then collected and passed to a second cell monolayer (10 min at 37°C). The procedure was repeated 10 times.

Synthesis of peptides and preparation of peptide/beads

Peptides were synthesized by the solid phase F-moc method (34) using an Applied Biosystem model 433 A peptide synthetizer (Foster City, CA). After the peptide assembly, the side chain-protected peptidyl resin was deblocked as previously described (35) and purified to apparent homogeneity by reverse-phase chromatography. An extra-sequence cysteine was added to peptides 1, 3, and 4 to obtain conformationally cyclic peptides. These peptides were treated overnight with a 5-fold excess of oxidized glutathione and purified by reverse-phase chromatography.

Coupling of CCR5 peptides to tosyl-activated Dynabeads M280 (Dynal, Oslo, Norway) was obtained following standard procedures. Briefly, 3 × 107 beads were incubated with 9 μg of CCR5 peptides in 50 mM borate buffer pH 9.5 (16 h at 37°C). After four washes in PBS, peptide/beads were ready for use.

Affinity-purification of Abs on peptide/beads

Binding of anti-CCR5 specific Igs to peptide/beads was obtained by incubating 9 μg Igs to 9 μg peptide/beads for 1 h at 4°C. Igs were eluted in 0.5 M acetic acid, dialyzed against RPMI 1640 medium, then tested in Mip1β binding assays and/or in HIV neutralization assays. To establish if the region recognized by anti-CCR5 Abs corresponds to a conformational epitope, the specific peptide/beads were incubated with 10 mM of 2-ME, and subsequently with 30 mM of N-ethyl maleimide (final concentration, 30 mM) for 60 min before Ab binding.

Results

Inhibition of Mip1β binding to surface CCR5 by sera of ESN

Unlike RANTES and Mip1α (the other two CCR5-binding β-chemokines that inhibit infection by R5 strains of HIV), Mip1β binds exclusively to CCR5. For this reason, a Mip1β radio-binding assay to measure the presence and function of CCR5 on cell surfaces was recently introduced (29). We used a similar radio-binding inhibition assay to search for anti-CCR5 Abs in sera of ESN and USN individuals as well as in sera of HIV-infected patients. A total of 133 serum samples from 45 USN, 40 HIV-seropositives, and 48 ESN were assayed (at a 1:10 dilution; five replicas for each serum) to investigate whether they could inhibit Mip1β binding to purified CD4+ cells. The ability of ESN sera to inhibit Mip1β binding to purified CD4+ cells was higher than that observed with sera from either USN or HIV-positives, and the difference was statistically significant (t = 1.939; p < 0. 05). The arithmetic means ± SE of the inhibition values were 1.42 ± 0. 7 (USN), 1.23 ± 0.06 (HIV-seropositives), and 5.2 ± 2.2 (ESN). The sera of six ESN (ESN9, 31, 32, 34, 53, 55) had values well above the cut-off (mean + 3 SE of USN controls) (Fig. 1⇓).

           FIGURE 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 1.

Inhibition of radiolabeled Mip1β binding to CD4+ cells by sera of six ESN (•). Means (♦) ± 3 SE of the values obtained with the sera of 45 USN controls (used to establish the cut-off), 48 ESN, and 40 HIV-seropositives are shown. Sera were employed at 1:10 dilution.

To exclude the possibility that the observed inhibition may have been due to high serum concentrations of β-chemokines, we measured the concentration of RANTES, Mip1α, and Mip1β in the six ESN sera in which Mip1β-inhibiting activity was detected and in the CD4+ sera of 10 USN control. The concentrations of all three chemokines were comparable between ESN and USN sera (data not shown).

Inhibition of Mip1β binding to CCR5 by sera of ESN is mediated by Abs

The sera of the six ESN that inhibited Mip1β binding to CCR5 and of three USN were purified by affinity chromatography (on agarose-bound anti-human Ig Abs) to obtain Ig-enriched and Ig-depleted fractions. Both fractions were then tested in the radio-binding assay. Fig. 2⇓A shows that Mip1β-binding inhibition was associated with the Ig-enriched fraction in all ESN samples, whereas no activity was observed in the Ig-depleted fractions. Both the Ig-enriched and Ig-depleted fractions from USN sera were devoid of inhibitory activity. The data suggest the Mip1β-binding inhibition to be due to Abs. The dose-dependence of the binding inhibition is shown in Fig. 2⇓B. In all but one case (ESN55), significant inhibition was observed at a concentration of 62.5 μg (corresponding to a serum dilution of 1:40); all fractions lost their inhibitory activity at a concentration of 15.6 μg (corresponding to a serum dilution of 1:160).

           FIGURE 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 2.

A, Inhibition of radiolabeled Mip1β binding to CD4+ cells by Ig-enriched and Ig-depleted fractions of ESN and USN sera. Both kinds of fractions were used at a concentration corresponding to a 1/10 dilution of sera. B, Dose dependence of the inhibition by Ig-enriched fractions.

Anti-CCR5 and not anti-Mip1β Abs are responsible for the inhibitory activity

To exclude the possibility that the inhibitory Abs could be directed against Mip1β (rather than against CCR5), we set up an ELISA to detect anti-Mip1β Abs. Mip1β-binding Abs could not be detected in either the whole sera or Ig-enriched fractions of either ESN or 10 USN (data not shown), although the ELISA was sensitive enough to detect concentrations as little as 100 pg/ml of anti-Mip1β Abs. Therefore, it is likely that the observed inhibition is due to Abs interacting with CCR5 and blocking Mip1β binding to its receptor, possibly by steric hindrance.

Anti-CCR5 Abs were demonstrated in five of five of the Mip1β-inhibiting sera by a similar radio-binding assay, which employs CD4+ T cells or CCR5-transfected U87 cells and radiolabeled anti-human Ig secondary Abs. A strong increase in binding was observed when CCR5-transfected U87 cells were compared with CD4+ T cells, as shown in Fig. 3⇓, A and B.

           FIGURE 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 3.

Anti-CCR5 Abs efficiently bind CCR5-positive cells and inhibit Mip1β-induced PBMC chemotaxis. Ab binding to (A) CD4+ T cells and (B) CCR5-transfected U87 cells. The values in B are subtracted of the values obtained with CXCR4-transfected U87 cell lines. The mAb 2D7 and Mip1β were used as positive controls. C, Anti-CCR5 Abs from ESN34 and ESN55 inhibit PBMC chemotactic response in a dose-dependent manner. Igs from USN5 were used as control. PBMC from ESN34 and ESN55 did not migrate in the chemotactic assay in response to Mip1β.

Anti-CCR5 Abs inhibit the chemotactic response to Mip1β

To evaluate whether the cells of ESN in whom anti-CCR5 Abs are detected are functionally CCR5-negative, purified Ig fractions from ESN34, ESN55, and from one control (USN5) were used to inhibit the chemotactic response induced by Mip1β on PBMC from healthy controls. The results show that anti-CCR5 Abs from ESN34 and ESN55 reduced chemotaxis in a dose-dependent manner (Fig. 3⇑C). Additionally, whereas control PBMCs migrated in response to Mip1β, PBMCs from ESN34 and ESN55 were not responsive to Mip1β (Fig. 3⇑C). These results indicate that anti-CCR5 Abs are functionally active as they suppress chemotaxis and suggest that, in vivo, these Abs down-regulate CCR5 expression.

Lack of correlation between anti-CCR5 Abs and other immune correlates of protection

No correlation was found between the presence of anti-CCR5 Abs and other immune correlates of protection previously associated with the ESN status. ESN were tested for the following correlates and resulted negative (data not shown): 1) the presence of a homozygous CCR5 deletion (CCR5-Δ32), 2) anti-CD4 Abs (anti-CD4 Abs were only detected in ESN53), or 3) serum HIV-specific IgA. Moreover, no correlation was found between anti-CCR5 Abs and gender: ESN55, 32, and 53 were males from heterosexual couples, the other three were females. Interestingly, the anti-CCR5 response is persistent in time: in the four cases tested (ESN34, 32, 53, and 55), anti-CCR5 titers persisted in sera obtained after 2 years of safe sex (data not shown).

Anti-CCR5 Abs inhibit the infectivity of R5 strains of HIV

The data reported above demonstrate that anti-CCR5 Abs can be found in the sera of ESN, and that these Abs prevent binding of Mip1β to CCR5. Because CCR5 is the main coreceptor for R5 strains of HIV, we verified whether anti-CCR5 Abs could also inhibit HIV-1 binding to CCR5. Thus, we assayed the activity of Ig fractions from five of the ESN in whom anti-CCR5 Abs were observed (ESN31, 32, 34, 53, and 55) in HIV-1 neutralization assays using primary virus isolates derived from seropositive patients. A number of primary isolates were analyzed for coreceptor usage; three isolates were chosen: HIV36 (R5 strain that uses CCR5 as its coreceptor), HIV26 (X4 strain that uses CXCR4 as its coreceptor), and HIV45 (amphitropic R3, R5, X4 strain that uses CCR3, CCR5, and CXCR4 as coreceptors). Additionally, the isolates from four seropositive partners (all using CCR5 as coreceptor) were used in the neutralization assay. Ig-enriched fractions from all five ESN inhibited infection by the R5 strain in a dose-dependent manner (Fig. 4⇓A); these fractions had no inhibitory effect on the other two HIV-1 strains (data not shown). The purified Ig fractions of four ESN (ESN34, 53, 31, and 55) inhibit the replication of the primary isolate derived from the corresponding four HIV-seropositive partners (Fig. 4⇓, B–E).

           FIGURE 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 4.

Neutralization of HIV-1 primary isolates by Ig-enriched fractions from ESN and USN sera. Five different isolates from HIV-seropositive partners (HIV36, 34, 53, 31, and 55) were used to infect PBMC. Infectivity reduction of the R5 strain HIV36 by Igs from all five patients is shown in A. The neutralizing activity of Igs isolated from sera of four ESN (ESN31, 34, 53, and 55) against primary isolates (R5 strains) from the corresponding four HIV+ partners is shown in B–E.

These data are consistent with the activity of Abs reactive with CCR5 and blocking the infectivity of CCR5-restricted viruses rather than with the activity of Abs reacting with HIV-1 epitopes involved in the interaction with CCR5, because in the latter case the amphitropic virus would also have been inhibited.

Evidence that anti-CCR5 Abs are also responsible for the neutralizing activity on the R5 strains was sought by immunoadsorption: Ig-enriched fractions from sera of three ESN and three USN were adsorbed on either CCR5- or CXCR4-transfected U87 cells. Adsorption on CCR5-positive cells (but not on CXCR4-positive cells) reduced both the virus-neutralizing activity (Fig. 5⇓A) and the Mip1β-binding inhibitory activity (Fig. 5⇓B). In particular, 1) in the neutralization assay, the concentration of unabsorbed fractions giving 50% inhibition (IC50), as well as that of CXCR4-adsorbed fractions, was ∼30 μg/assay, whereas the IC50 of CCR5-adsorbed fractions was >200 μg/assay; 2) Mip1β-binding inhibition was ∼82% in both unabsorbed and CXCR4-adsorbed fractions, but was 57% in CCR5-adsorbed fractions. ESN inhibiting sera could also neutralize another primary R5 isolate (HIV53) in both resting and activated PBMC neutralization assays (data not shown). These data are consistent with the hypothesis that the Abs removed by adsorption on CCR5 are the same responsible for the neutralization of HIV-1 R5 strains.

           FIGURE 5.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 5.

Immunoadsorption of Mip1β-inhibiting and HIV-neutralizing Abs. Ig-enriched fractions of sera from ESN34 and USN1 were adsorbed on either CCR5-transfected or CXCR4-transfected U87 cells. Adsorbed sera were assayed in the neutralization assay (A) and in the Mip1β binding assay (B). CCR5-adsorbed fractions, but not CXCR4-adsorbed fractions, of ESN34 lost activity in both assays.

CCR5 expression on PBMC from Ab-positive ESN individuals

The issue of in vivo coexistence of anti-CCR5 Abs and CCR5 Ag on the surface of cells was addressed by studying the CCR5 phenotype of purified CD4+ cells from two patients (ESN55 and ESN34) and from four USN controls using the Mip1β binding assay. No binding could be detected for the two patients, while all the USN exhibited normal binding values (range, 2000–3500 cpm; data not shown). To evaluate CCR5 expression on the cell surface, purified, preactivated CD4+ cells from ESN55 and ESN34 and one USN control were stained with a CCR5-specific Ab (mAb 2D7) and tested in a flow cytometry assay, which employs FITC-labeled anti-mouse Ig secondary Abs. Results (Fig. 6⇓) showed that, whereas CCR5 was normally detected on lymphocytes of the USN, no CCR5 expression was observed on the cells of the two ESN individuals.

           FIGURE 6.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 6.

In vivo down-modulation of CCR5 by anti-CCR5 Abs. Mouse anti-CCR5 Ab (2D7) binding on activated CD4+ cells from ESN55, ESN34, and USN8 (positive control). An isotype control was used as negative control (not shown).

Epitope mapping of anti-CCR5 Abs

Anti-CCR5 Abs were tested on a panel of synthetic peptides covering the complete sequence of the extra membrane region of CCR5 (Table I⇓). Specific Ab binding to peptide 3 (aa 89–102), corresponding to the second external domain (first cystein loop) of CCR5, was observed. As shown in Fig. 7⇓A, the Abs are highly selective for this epitope as no binding was observed on a panel of other peptides (peptides 1, 2, 4, and 5). Binding of the anti-CCR5 Abs from ESN sera to peptide 3 was abolished by addition of 2-ME and N-ethyl maleimide, which cause reduction and alkylation of the cystein loop (Fig. 7⇓A), suggesting that Ab recognition requires the maintenance of the epitope tertiary structure.

           FIGURE 7.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 7.

Epitope mapping of anti-CCR5 Abs. A, Abs eluted from peptide/beads (see Table I⇓) were assayed in the Mip1β binding inhibition assay. Note that reduction/alkylation of peptide 3 abolishes its binding activity. B, Neutralization of HIV-1 primary isolate (HIV36) by Abs derived from ESN55, ESN32, and USN sera and affinity-purified on peptide 3.

View this table:
  • View inline
  • View popup
Table I.

Peptides covering the sequence of the extramembrane region of CCR5

Affinity-purified anti-CCR5 Abs inhibit HIV-1 replication

Abs to peptide 3 were affinity-purified from ESN55 and ESN32 sera and tested in HIV neutralization assays. The neutralizing titers obtained with Igs eluted from peptide 3 (Fig. 7⇑B) were higher (IC50 of 1 μg/ml for ESN55 and 5 μg/ml for ESN32) than those of total serum Igs (IC50 of 13 μg/ml for ESN55 and 82.7 μg/ml for ESN32) (Fig. 4⇑A).

Discussion

Exposure to HIV does not inevitably lead to infection. Thus, individuals who do not seroconvert despite being repeatedly exposed to HIV have been described in different cohorts of at-risk individuals including: sexually exposed gay men, neonates of HIV-seropositive mothers, accidentally exposed health care workers, prostitutes, and heterosexual partners of HIV-seropositive individuals. ESN individuals are clearly a heterogeneous group because a number of different immunological features have been associated with the ESN status at variable frequencies (1, 2, 3, 4, 5, 6, 7, 12). That resistance to HIV-infection can also have a genetic basis was suggested by the observation that a mutant form of one of the most important coreceptors for HIV on the surface of CD4+ cells, CCR5, is present in a subset of such individuals. Mutations in CCR5 lead to a defective coreceptor function for those HIV-1 virus strains that use CCR5 as their coreceptor (R5 strains), thereby impeding the penetration of HIV into the target cell and the onset of disease (19, 21).

Given that 1) the specific ligand for CCR5 is Mip1β, 2) a functional CCR5 coreceptor is a requirement for infection by HIV-1 R5 strains (36, 37), 3) R5 strains are responsible for the establishment of most new infections (24), and 4) the occupancy of CCR5 receptor by its ligands (38, 39) or by Abs (31, 40) prevents infection by HIV, we investigated whether binding of Mip1β to CCR5 could be inhibited by sera of ESN. Sera from neither controls nor HIV-infected individuals could interfere with the binding of Mip1β to CCR5; in contrast, inhibitory activity was detected in sera of a number of ESN: binding of Mip1β to CCR5 was inhibited by sera of 6/48 (12.5%) ESN, but not by sera of either 45 USN or 40 HIV+ seropositive individuals (p < 0.001). Characterization of this inhibitory activity indicated that anti-CCR5 Abs 1) are present in the sera of some ESN, 2) recognize a conformational epitope in the first cysteine loop of CCR5, 3) inhibit Mip1β binding to CCR5, 4) down-modulate CCR5 expression on PBMC in vivo, 5) inhibit Mip1β-induced chemotaxis of control PBMC, and 6) block the HIV coreceptor function of CCR5, neutralizing the infectivity of R5 strains of HIV-1.

Although the potentially protective role of such Abs is evident, their origin is a matter of speculation: anti-CCR5 Abs 1) may be “naturally” present in some rare individuals (<1/85, which is the sum of the two control populations), 2) may be elicited by exposure to HIV or by particular and still unidentified factor(s) involved in HIV exposure, or 3) may be elicited by some HIV-unrelated antigenic stimulation. In this context, it is interesting that a recent report showed that autoantibodies to CCR5 could be induced in C57BL/6 mice by inoculation with a papilloma virus modified to express CCR5 peptides (41). Such Abs could inhibit binding of β-chemokines to CCR5, as well as block infection with HIV-1 (41). Interestingly, the CCR5 epitope used to immunize mice and the epitope recognized by the CCR5-specific Igs detected in ESN sera show >90% homology.

The autoantibodies produced during the course of viral infections have been attributed to virus-induced self-alterations that become autoimmunogenic (42). Therefore, the infrequent anti-CCR5 response might be due to a particular antigenic stimulation or to the particular reactivity of rare individuals. Lehner et al. (43) have shown that xenogenic immunization of macaques with SIV grown in human cells induces protective immunity and the generation of Abs reactive with simian CCR5 that are capable of neutralizing SIV in in vitro assays. Allogenic, rather than xenogenic, immunization might have produced the same result in our ESN individuals. The recent report that CCR5 can act as an alloantigen in CCR5-Δ32 homozygous individuals eliciting Abs that compete with RANTES and inhibit infection by R5, but not X4, primary isolates of HIV-1 supports this hypothesis (44). Along the same lines, therapeutic strategies aimed at preventing HIV-1 infection by means of Abs to CCR5 elicited via immunization with a modified CCR5 gene are currently being developed (45). In this context, it is interesting that, whereas interruption of at-risk sex in ESN is followed by a sharp drop in the titers of HIV-specific IgA and in the loss of HIV-specific cell-mediated immunity (5), no modifications were observed in the concentration of anti-CCR5 Abs. These observations indicate that, whereas HIV-specific immunity in ESN is dependent on a continuous exposure to the virus, anti-CCR5 Abs could be maintained independently of exposure to HIV. These results also suggest that 1) immune protection against HIV infection may be secondary to the activation of multiple mechanisms that, beside potent HIV-specific cell-mediated immunity in peripheral blood and mucosal immunity in the genital tract, include anti-CCR5-specific Abs; 2) anti-CCR5 Ab-mediated protection against HIV infection could be maintained even in the absence of HIV-specific immune responses in ESN; 3) vaccine strategies aimed at elicitation of anti-CCR5 Abs could be less dependent on the need for repeated vaccine boost, compared with vaccine strategies based on the induction of HIV-specific immune responses.

In conclusion, our results provide further information on the heterogeneous mechanisms underlying resistance to HIV infection in ESN individuals and reinforce the concept that immune protection can be mediated by multiple mechanisms, one of which could be anti-CCR5 Abs. Because other, still unclear mechanisms, including complement, chemokines, and other antiviral factors, are likely to be involved in the phenomenon of resistance to HIV infection, more research is necessary to shed further light on the immunologic correlates of this fortunate condition.

Acknowledgments

We thank P. Lusso for his support, helpful discussions, and for providing mAb 2D7. We also thank V. Nardese, D. Trabattoni, and S. Piconi for contributions to this work.

Footnotes

  • ↵1 This work was supported by grants from Istituto Superiore di Sanita’ “I Progetto AIDS 1997 and II Progetto AIDS 1998” and from the European Community “Concerted Action # BMH4-CT97-205.”

  • ↵2 Address correspondence and reprint requests to Dr. Lucia Lopalco, Centro San Luigi, San Raffaele Scientific Institute, via Stamira D’Ancona 20, 20127 Milano, Italy. E-mail address: lopalco.lucia{at}hsr.it

  • ↵3 Abbreviations used in this paper: ESN, exposed seronegative; USN, unexposed seronegative; Mip1β, macrophage inflammatory protein 1β.

  • Accepted January 5, 2000.
  • Copyright © 2000 by The American Association of Immunologists

References

  1. ↵
    Lopalco, L., C. Pastori, A. Cosma, S. E. Burastero, B. Capiluppi, E. Boeri, A. Beretta, A. Lazzarin, A. G. Siccardi. 2000. Anti cell antibodies in exposed seronegative individuals with HIV-1 neutralizing activity. AIDS Res. Hum. Retrovir. 16: 109
    OpenUrlCrossRefPubMed
  2. ↵
    Beretta, A. L., S. Furci, A. Burastero, M. E. Cosma, L. Dinelli, C. Lopalco, G. DeSantis, E. Tambussi, S. Sabbatani Carrow, et al 1996. HIV-1-specific immunity in persistently seronegative individuals at high risk for HIV infection. Immunol. Lett. 51: 39
    OpenUrlCrossRefPubMed
  3. ↵
    Burastero, S. E., D. Gaffi, L. Lopalco, G. Tambussi, B. Borgonovo, C. DeSantis, C. Abecasis, P. Robbioni, A. Gasparri, A. Lazzarin, et al 1996. Autoantibodies to CD4 in HIV type I-exposed seronegative individuals. AIDS Res. Hum. Retrovir. 12: 273
    OpenUrlCrossRefPubMed
  4. ↵
    Mazzoli, S., D. Trabattoni, S. Lo Caputo, S. Piconi, C. Ble, F. Meacci, S. Ruzzante, A. Salvi, F. Semplici, R. Longhi, et al 1997. HIV-specific mucosal and cellular immunity in HIV-seronegative partners of HIV-seropositive individuals. Nat. Med. 3: 1250
    OpenUrlCrossRefPubMed
  5. ↵
    Mazzoli, S., L. Lopalco, A. Salvi, D. Trabattoni, S. Lo Caputo, F. Semplici, M. Biasin, C. Ble, A. Cosma, C. Pastori, et al 1999. HIV-specific IgA and HIV neutralizing activity in the serum of exposed seronegative partners of HIV-seropositive individuals. J. Infect. Dis. 180: 871
    OpenUrlAbstract/FREE Full Text
  6. ↵
    Rowland-Jones, S. L., A. McMichael. 1995. Immune responses in HIV-exposed seronegatives: have they repelled the virus?. Curr. Opin. Immunol. 7: 448
    OpenUrlCrossRefPubMed
  7. ↵
    Shearer, G. M., M. Clerici. 1996. Protective immunity against HIV infection: has nature done the experiment for us?. Immunol. Today 17: 21
    OpenUrlCrossRefPubMed
  8. ↵
    Stranford, S. A., J. Skurnick, D. Louria, D. Osmond, S. Y. Chang, J. Sninsky, G. Ferrari, K. Weinhold, C. Lindquist, J. A. Levy. 1999. Lack of infection in HIV-exposed individuals is associated with a strong CD8+ cell noncytotoxic anti-HIV response. Proc. Natl. Acad. Sci. USA 96: 1030
    OpenUrlAbstract/FREE Full Text
  9. ↵
    Clerici, M., J. V. Giorgi, C. C. Chou, V. K. Gudeman, J. A. Zack, P. Gupta, H.-N. Ho, P. G. Nishanian, J. A. Berzofsky, G. M. Shearer. 1992. Cell-mediated immune response to human immunodeficiency virus (HIV) type-1 in seronegative homosexual men with recent sexual exposure to HIV-1. J. Infect. Dis. 165: 1012
    OpenUrlAbstract/FREE Full Text
  10. ↵
    Clerici, M., J. M. Levin, H. A. Kessler, A. Harris, J. A. Berzofsky, A. L. Landay, G. M. Shearer. 1994. HIV-specific T helper activity in seronegative health care workers exposed to contaminated blood. J. Am. Med. Assoc. 271: 42
    OpenUrlCrossRefPubMed
  11. ↵
    Pinto, L. A., J. Sullivan, J. A. Berzofsky, M. Clerici, H. A. Kessler, A. L. Landay, G. M. Shearer. 1995. ENV-specific cytotoxic T lymphocyte responses in HIV seronegative health care workers occupationally exposed to HIV-contaminated body fluids. J. Clin. Invest. 96: 867
    OpenUrlCrossRefPubMed
  12. ↵
    Furci, L., G. Scarlatti, S. Burastero, G. Tambussi, C. Colognesi, C. Quillent C, R. Longhi, P. Loverro, B. Borgonovo, D. Gaffi, et al 1997. Antigen-driven C-C chemokine-mediated HIV-1 suppression by CD4+ T cells from exposed uninfected individuals expressing the wild-type CCR-5 allele. J. Exp. Med. 186: 455
    OpenUrlAbstract/FREE Full Text
  13. ↵
    Cheynier, R. P., P. Langlade-Demoyen, M. R. Marescot, F. Plata. 1992. Cytotoxic T lymphocyte responses in the peripheral blood of children born to human immunodeficiency virus-1 infected mothers. Eur. J. Immunol. 22: 2211
    OpenUrlCrossRefPubMed
  14. ↵
    Langlade-Demoyen, P., N. Ngo-Giang-Huong, F. Ferchal, E. Oksenhendler. 1994. HIV-nef specific cytotoxic T lymphocytes in non-infected heterosexual contact of HIV-infected patients. J. Clin. Invest. 93: 1293
    OpenUrlCrossRefPubMed
  15. ↵
    Rowland-Jones, S. L.. 1994. HIV-specific cytotoxic T-cells in HIV-exposed but uninfected Gambian women. Nat. Med. 1: 59
    OpenUrl
  16. ↵
    Langlois, A. J., K. J. Weinhold, T. J. Mattheus, M. L. Greenberg, D. P. Bolognesi. 1992. The ability of certain SIV vaccines to provoke reactions against normal cells. Science 255: 292
    OpenUrlFREE Full Text
  17. ↵
    Stott, E. J.. 1991. Anti-cell antibody in macaques. Nature 353: 393
    OpenUrlCrossRefPubMed
  18. ↵
    Chan, W. L, A. Rodgers, C. Grief, N. Almond, S. Ellis, B. Flanagan, P. Silvera, J. Bootman, J. Stott, K. Kent. 1995. Immunization with class I human histocompatibility leukocyte antigen can protect macaques against challenge infection with SIVmac-32H. AIDS 9: 223
    OpenUrlCrossRefPubMed
  19. ↵
    Liu, R., W. A. Paxton, S. Choe, D. Ceradini, S. R. Martin, R. Horuk, M. E. MacDonald, H. Stuhlmann, R. A. Koup, N. R. Landau. 1996. Homozygous defect in HIV-1 coreceptor accounts for resistance of some multiply-exposed individuals to HIV-1 infection. Cell 86: 1
    OpenUrlCrossRefPubMed
  20. ↵
    Paxton, W. A., S. R. Martin, D. Tse, T. R. O’Brien, J. Skurnick, N. L. VanDevanter, N. Padian, J. F. Braun, D. P. Kotler, S. M. Wolinsky, R. A. Koup. 1996. Relative resistance to HIV-1 infection of CD4+ lymphocytes from persons who remain uninfected despite multiple high-risk sexual exposures. Nat. Med. 2: 412
    OpenUrlCrossRefPubMed
  21. ↵
    Quillent, C., E. Oberlin, J. Braun, D. Rousset, G. Gonzalez-Canali, P. Metais, L. Montagnier, J. L. Virelizier, F. Arenzana-Seisdedos, A. Beretta. 1998. HIV-1 resistance phenotype conferred by combination of two separate inherited mutations of CCR5 gene. Lancet 351: 14
    OpenUrlCrossRefPubMed
  22. ↵
    Kostrikis, L.. 1998. The 641 allele of CCR2 delays HIV-1 disease progression and is associated with a mutation within the CCR5 regulatory region. Nat. Med. 4: 350
    OpenUrlCrossRefPubMed
  23. ↵
    Berger, E. A., R. W. Doms, E. M. Fenyo, B. T. Korber, D. R. Littman, J. P. Moore, Q. J. Sattentau, H. Schuitemaker, J. Sodroski, R. A. Weiss. 1998. A new classification for HIV-1. Nature 391: 240
    OpenUrlCrossRefPubMed
  24. ↵
    Blaak, H., A. B. Van’t Wout, M. Brouwer, M. Cornelissen, N. A. Kootstra, N. Albrecht-van Lent, R. P. Keet, J. Goudsmit, R. A. Coutinho, H. Schuitemaker. 1998. Infectious cellular load in human immmunodeficiency virus type 1 (HIV-1)-infected individuals and susceptibility of peripheral blood mononuclear cells from their exposed partners to non-syncytium-inducing HIV-1 as major determinants for HIV-1 transmission in homosexual couples. J. Virol. 72: 218
    OpenUrlAbstract/FREE Full Text
  25. ↵
    Stewart, G.. 1998. Chemokine genes beating the odds. Nat. Med. 4: 275
    OpenUrlCrossRefPubMed
  26. ↵
    Simmons, G., R. R. Clapham, L. Picard, R. E. Offord, M. M. Rosenkilde, T. W. Schwartz, R. Buser, T. N. C. Wells, A. E. Proudfoot. 1997. Potent inhibition of HIV-1 infectivity in macrophages and lymphocytes by a novel CCR5 antagonist. Science 276: 276
    OpenUrlAbstract/FREE Full Text
  27. ↵
    Moore, J. P.. 1997. Coreceptors: implications for HIV pathogenesis and therapy. Science 276: 51
    OpenUrlFREE Full Text
  28. ↵
    Cohen, J.. 1997. Exploiting the HIV-chemokine nexus. Science 275: 1261
    OpenUrlFREE Full Text
  29. ↵
    Trkola, A., T. Ketas, V. N. Kewalramani, F. Endorf, J. M. Binley, H. Katinger, J. Robinson, D. R. Littman, J. P. Moore. 1996. CD4-dependent, antibody-sensitive interactions between HIV-1 and its co-receptor CCR5. Nature 384: 184
    OpenUrlCrossRefPubMed
  30. ↵
    Grassi, F., R. Meneveri, M. Gullberg, L. Lopalco, G.B. Rossi, P. Lanza, C. De Santis, G. Brattsand, S. Butto, E. Ginelli, A.G. Siccardi. 1991. Human immunodeficiency virus type 1 gp120 mimics a hidden monomorphic epitope borne by class I major histocompatibility complex heavy chains. J. Exp. Med. 174: 53
    OpenUrlAbstract/FREE Full Text
  31. ↵
    Wu, L., W. A. Paxton, N. Kassam, N. Ruffing, J. B. Rottman, N. Sullivan, H. Choe, J. Sodroski, W. Newman, R. A. Koup, C. R. Mackay. 1997. CCR5 levels and expression pattern correlate with infectability by macrophage-tropic HIV-1, in vitro. J. Exp. Med. 185: 1681
    OpenUrlAbstract/FREE Full Text
  32. ↵
    Moore, J. P., J. A. McKeating, Y. Huang, A. Ashkenazi, D. D. Ho. 1992. Virions of primary human immunodeficiency virus type 1 isolates resistant to soluble CD4 (sCD4) neutralization differ in sCD4 binding and glycopreotein gp120 retention from sCD4-sensitive isolates. J. Virol. 66: 235
    OpenUrlAbstract/FREE Full Text
  33. ↵
    Zolla Pazner, S., S. Sharpe. 1995. A resting cell assay for improved detection of antibody-mediated neutralization of HIV type 1 primary isolates. AIDS Res. Hum. Retovir. 11: 1449
    OpenUrlCrossRefPubMed
  34. ↵
    Fields, G. B., R. L. Noble. 1990. Solid phase peptide synthesis utilizing 9-fluorenylmethoxycarbonyl amino acids. Int. J. Pept. Prot. Res. 35: 161
    OpenUrl
  35. ↵
    Kings, D. S., C. G. Fields, G. B. Fields. 1990. A cleveage method which minimizes side chain reactions following Fmoc solid phase peptide synthesis. Int. J. Pept. Prot. Res. 36: 255
    OpenUrl
  36. ↵
    Alkhatib, G., C. Combadiere, C. C. Broder, Y. Feng, P. E. Kennedy, P. M. Murphy, E. A. Berger. 1996. CC CKR5: a RANTES, MIP-1α, MIP-1β, receptor as a fusion cofactor for macrophage-tropic HIV-1. Science 272: 1955
    OpenUrlAbstract/FREE Full Text
  37. ↵
    D’Souza, M. P., V. Harden. 1996. Chemokines and HIV-1 second receptors. Nat. Med. 2: 1293
    OpenUrlCrossRefPubMed
  38. ↵
    Cocchi, F., A. De Vico, A. Garzimo-Demo, S. K. Arya, R. C. Gallo, P. Lusso. 1995. Identification of RANTES, Mip1α and Mip1β as the major HIV-suppressive factors produced by CD8+ T cells. Science 270: 1811
    OpenUrlAbstract/FREE Full Text
  39. ↵
    Trkola, A., W. A. Paxton, S. P. Monard, J. A. Hoxie, M. A. Siani, D. A. Thompson, L. Wu, C. R. Mackay, R. Horuk, J. P. Moore. 1998. Genetic subtype-independent inhibition of human immunodeficiency virus type 1 replication by CC and CXC chemokines. J. Virol. 72: 396
    OpenUrlAbstract/FREE Full Text
  40. ↵
    Wu, L., G. LaRosa, N. Kassam, C. J. Gordon, H. Heath, N. Ruffing, H. Chen, J. Humblias, M. Samson, M. Parmentier, J. P. Moore, C. R. Mackay. 1997. Interaction of chemokine receptor CCR5 with its ligands: multiple domains for HIV-1 gp120 binding and a single domain for chemokine binding. J. Exp. Med. 186: 1373
    OpenUrlAbstract/FREE Full Text
  41. ↵
    Chackerian, B., D. R. Lowy, J. T. Schiller. 1999. Induction of autoantibodies to mouse CCR5 with recombinant papilloma virus. Proc. Nat. Acad. Sci. USA 96: 2373
    OpenUrlAbstract/FREE Full Text
  42. ↵
    Bachmann, M. F., R. M. Zinkernagel. 1997. Neutralizing antiviral B cell responses. Annu. Rev. Immunol. 15: 235
    OpenUrlCrossRefPubMed
  43. ↵
    Lehner, T., Y. Wang, C. Doyle, L. Tao, L. A. Bergmeier, E. Mitchell, W. M. J. Bogers, J. Heeney, C. G. Kelly. 1999. Induction of inhibitory antibodies to the CCR5 chemokine receptor and their complementary role in preventing SIV infection in macaques. Eur. J. Immunol. 28: 877
    OpenUrl
  44. ↵
    Ditzel, H. J., M. M. Rosenkilde, P. Garred, M. Wang, K. Koefoed, C. Pedersen, D. R. Burton, T. W. Schwartz. 1998. The CCR5 receptor acts as an alloantigen in CCR5Δ32 homozygous individuals: identification of chemokine-and HIV-1-blocking human antibodies. Proc. Nat. Acad. Sci. USA 95: 5241
    OpenUrlAbstract/FREE Full Text
  45. ↵
    Zuber, B., M. Levi, M. Karlsson, B. M. Sioberg, M. Brytting, and B. Wahren. 1998. Immune reaction against the HIV-1 coreceptor CCR5. Third European Conference on AIDS Research, Munich, Germany, p. 6.S1. (Abstr.).
PreviousNext
Back to top

In this issue

The Journal of Immunology: 164 (6)
The Journal of Immunology
Vol. 164, Issue 6
15 Mar 2000
  • Table of Contents
Print
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for your interest in spreading the word about The Journal of Immunology.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
CCR5-Reactive Antibodies in Seronegative Partners of HIV-Seropositive Individuals Down-Modulate Surface CCR5 In Vivo and Neutralize the Infectivity of R5 Strains of HIV-1 In Vitro
(Your Name) has forwarded a page to you from The Journal of Immunology
(Your Name) thought you would like to see this page from the The Journal of Immunology web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
CCR5-Reactive Antibodies in Seronegative Partners of HIV-Seropositive Individuals Down-Modulate Surface CCR5 In Vivo and Neutralize the Infectivity of R5 Strains of HIV-1 In Vitro
Lucia Lopalco, Claudia Barassi, Claudia Pastori, Renato Longhi, Samuele E. Burastero, Giuseppe Tambussi, Francesco Mazzotta, Adriano Lazzarin, Mario Clerici, Antonio G. Siccardi
The Journal of Immunology March 15, 2000, 164 (6) 3426-3433; DOI: 10.4049/jimmunol.164.6.3426

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
CCR5-Reactive Antibodies in Seronegative Partners of HIV-Seropositive Individuals Down-Modulate Surface CCR5 In Vivo and Neutralize the Infectivity of R5 Strains of HIV-1 In Vitro
Lucia Lopalco, Claudia Barassi, Claudia Pastori, Renato Longhi, Samuele E. Burastero, Giuseppe Tambussi, Francesco Mazzotta, Adriano Lazzarin, Mario Clerici, Antonio G. Siccardi
The Journal of Immunology March 15, 2000, 164 (6) 3426-3433; DOI: 10.4049/jimmunol.164.6.3426
del.icio.us logo Digg logo Reddit logo Twitter logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like

Jump to section

  • Article
    • Abstract
    • Materials and Methods
    • Results
    • Discussion
    • Acknowledgments
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF

Related Articles

Cited By...

More in this TOC Section

  • Differential Susceptibility to Staphylococcal Superantigen (SsAg)-Induced Apoptosis of CD4+ T Cells from Atopic Dermatitis Patients and Healthy Subjects: The Inhibitory Effect of IL-4 on SsAg-Induced Apoptosis
  • HIV-1 Vaccination Administered Intramuscularly Can Induce Both Systemic and Mucosal T Cell Immunity in HIV-1-Uninfected Individuals
  • Osteopontin (Eta-1) and Fibroblast Growth Factor-2 Cross-Talk in Angiogenesis
Show more CLINICAL IMMUNOLOGY

Similar Articles

Navigate

  • Home
  • Current Issue
  • Next in The JI
  • Archive
  • Brief Reviews
  • Pillars of Immunology
  • Translating Immunology

For Authors

  • Submit a Manuscript
  • Instructions for Authors
  • About the Journal
  • Journal Policies
  • Editors

General Information

  • Advertisers
  • Subscribers
  • Rights and Permissions
  • Accessibility Statement
  • FAR 889
  • Privacy Policy
  • Disclaimer

Journal Services

  • Email Alerts
  • RSS Feeds
  • ImmunoCasts
  • Twitter

Copyright © 2022 by The American Association of Immunologists, Inc.

Print ISSN 0022-1767        Online ISSN 1550-6606