HIV-1 tat promotes integrin-mediated HIV transmission to dendritic cells by binding Env spikes and competes neutralization by anti-HIV antibodies

Paolo Monini, Aurelio Cafaro, Indresh K Srivastava, Sonia Moretti, Victoria A Sharma, Claudia Andreini, Chiara Chiozzini, Flavia Ferrantelli, Maria R Pavone Cossut, Antonella Tripiciano, Filomena Nappi, Olimpia Longo, Stefania Bellino, Orietta Picconi, Emanuele Fanales-Belasio, Alessandra Borsetti, Elena Toschi, Ilaria Schiavoni, Ilaria Bacigalupo, Elaine Kan, Leonardo Sernicola, Maria T Maggiorella, Katy Montin, Marco Porcu, Patrizia Leone, Pasqualina Leone, Barbara Collacchi, Clelia Palladino, Barbara Ridolfi, Mario Falchi, Iole Macchia, Jeffrey B Ulmer, Stefano Buttò, Cecilia Sgadari, Mauro Magnani, Maurizio P M Federico, Fausto Titti, Lucia Banci, Franco Dallocchio, Rino Rappuoli, Fabrizio Ensoli, Susan W Barnett, Enrico Garaci, Barbara Ensoli, Paolo Monini, Aurelio Cafaro, Indresh K Srivastava, Sonia Moretti, Victoria A Sharma, Claudia Andreini, Chiara Chiozzini, Flavia Ferrantelli, Maria R Pavone Cossut, Antonella Tripiciano, Filomena Nappi, Olimpia Longo, Stefania Bellino, Orietta Picconi, Emanuele Fanales-Belasio, Alessandra Borsetti, Elena Toschi, Ilaria Schiavoni, Ilaria Bacigalupo, Elaine Kan, Leonardo Sernicola, Maria T Maggiorella, Katy Montin, Marco Porcu, Patrizia Leone, Pasqualina Leone, Barbara Collacchi, Clelia Palladino, Barbara Ridolfi, Mario Falchi, Iole Macchia, Jeffrey B Ulmer, Stefano Buttò, Cecilia Sgadari, Mauro Magnani, Maurizio P M Federico, Fausto Titti, Lucia Banci, Franco Dallocchio, Rino Rappuoli, Fabrizio Ensoli, Susan W Barnett, Enrico Garaci, Barbara Ensoli

Abstract

Use of Env in HIV vaccine development has been disappointing. Here we show that, in the presence of a biologically active Tat subunit vaccine, a trimeric Env protein prevents in monkeys virus spread from the portal of entry to regional lymph nodes. This appears to be due to specific interactions between Tat and Env spikes that form a novel virus entry complex favoring R5 or X4 virus entry and productive infection of dendritic cells (DCs) via an integrin-mediated pathway. These Tat effects do not require Tat-transactivation activity and are blocked by anti-integrin antibodies (Abs). Productive DC infection promoted by Tat is associated with a highly efficient virus transmission to T cells. In the Tat/Env complex the cysteine-rich region of Tat engages the Env V3 loop, whereas the Tat RGD sequence remains free and directs the virus to integrins present on DCs. V2 loop deletion, which unshields the CCR5 binding region of Env, increases Tat/Env complex stability. Of note, binding of Tat to Env abolishes neutralization of Env entry or infection of DCs by anti-HIV sera lacking anti-Tat Abs, which are seldom present in natural infection. This is reversed, and neutralization further enhanced, by HIV sera containing anti-Tat Abs such as those from asymptomatic or Tat-vaccinated patients, or by sera from the Tat/Env vaccinated monkeys. Thus, both anti-Tat and anti-Env Abs are required for efficient HIV neutralization. These data suggest that the Tat/Env interaction increases HIV acquisition and spreading, as a mechanism evolved by the virus to escape anti-Env neutralizing Abs. This may explain the low effectiveness of Env-based vaccines, which are also unlikely to elicit Abs against new Env epitopes exposed by the Tat/Env interaction. As Tat also binds Envs from different clades, new vaccine strategies should exploit the Tat/Env interaction for both preventative and therapeutic interventions.

Conflict of interest statement

Competing Interests: IKS, VAS, EK, JBU, RR and SWB are employees of Novartis Vaccines & Diagnostics, Inc. Novartis provided the recombinant HIV-1 Env proteins as a partner in several collaborative studies (the European projects AVIP and MUVAPRED, the “Joint Program ISS/Novartis (Chiron) for the development of a combined vaccine against HIV/AIDS.” There is one patent to declare: Novel Tat complexes, and vaccines comprising them (Inventor: Ensoli B.; Applicant: ISS (100%); priority date March 11, 2004; N. PCT/EP2005/003043, Publication N. WO2005/090391). There are no products in development or marketed products to declare. This does not alter the authors’ adherence to all the PLOS ONE policies on sharing data and materials, as detailed online in the guide for authors.

Figures

Figure 1. Virological outcome in Tat/Env-vaccinated or…
Figure 1. Virological outcome in Tat/Env-vaccinated or control monkeys after intrarectal challenge with the SHIVSF162P4cy (70 MID50).
Box plots of (A) Viral RNA, (B) proviral DNA in blood at 2, 3, 4 and 5 weeks after challenge, respectively; and (C) proviral DNA at week 4 after challenge in rectal tissue (RT) and inguinal lymph nodes (LN). Statistical analysis was performed by the one-sided Wilcoxon rank sum test. Red: monkeys vaccinated with Tat/Env (n = 6); blue: control animals (n = 6).
Figure 2. Enhancement by soluble Tat of…
Figure 2. Enhancement by soluble Tat of HIV-1 infection in MDDCs.
Peak infection fold increase reached 3–8 days after infection with the R5 pSF162LUC in MDDCs from 10 different donors. The virus was pre-incubated with 1 µM cys22 Tat or with PBS-0.1% BSA (control buffer). Fold increase were calculated as the ratio between the RLU from MDDCs infected with virus pre-incubated with cys22 Tat and RLU from MDDCs infected with virus pre-incubated with control buffer (baseline RLU: 298.95) (both values had been previously subtracted of the uninfected MDDCs RLU background).
Figure 3. Tat-mediated entry of VLPs expressing…
Figure 3. Tat-mediated entry of VLPs expressing R5 or X4 Env into MDDCs.
(A) and (B): entry in immature MDDCs of null-VLPs (control) or (A) VLPs expressing R5-Env (HIV-1 BaL) (VLP-R5Env) or (B) VLPs expressing X4-Env (HIV-1 HXBc2) (VLP-X4Env), in the absence or presence of increasing concentrations of cys22 Tat after 4 h of incubation, evaluated by flow cytometry. Results are shown as either dot-plot (A) or histograms (B). SSC: side scatter. Numbers in the insets represent the percentage of GFP positive cells. (C) Confocal microscopy analysis of VLP-R5Env entry into MDDCs in the presence of cys22 Tat (60 nM) after 4 h of incubation (0.2 µm optical sections form the apical to the basal cell side).
Figure 4. Tat released by producing T…
Figure 4. Tat released by producing T cells increases entry and infection of a Tat-independent replication-incompetent SF162 pseudovirus.
CEMss cells were either infected (CEMss-Tat) or not infected (CEMss) with VSV-G/HIV. After 24 h cells were co-cultured for 4 days with TZM-bl cells in the presence (TZM-bl/VP-SF162) or absence (TZM-bl) of a Tat-independent GFP-expressing single-cycle HIV-1 SF162 virus (VP/SF162), and then GFP expression evaluated by flow cytometry. Results are expressed as MFI fold increase with respect to the TZM-bl + CEMss co-culture (baseline MFI: 1.9).
Figure 5. Adherent Tat binds HIV particles…
Figure 5. Adherent Tat binds HIV particles and increases MDDC productive infection and transmission to T cells.
(A) Immuno-scanning electron microscopic analysis of HIV bound to Tat-coated latex beads. Images obtained with secondary electrons (left panels) or with backscattered electron (right panels) are shown. Latex beads were incubated with control buffer and HIV (A, B); with Tat and medium (C, D); or with Tat and HIV (E, F). Bar represents 5 µm. (B) MDDC infection with HIV SF162 on adherent wt Tat at different time points after infection. Infection of MDDCs from 2 different donors was carried out on plates coated with wt Tat (0.01 µM for donor 1, 0.1 µM for donor 2) or control buffer. Infection fold increase was calculated as the ratio between the content of p24 antigen detected in the supernatants of MDDCs seeded on adherent Tat and that detected in the supernatants of MDDCs seeded on control buffer (baseline, for donor 1, p24 was 154 and 119 pg/mL at day 7 and 10, respectively; for donor 2, p24 was 38 and 159 pg/mL at day 7 and 10, respectively). (C) Infection of PHA-activated CD4 T cell blasts with supernatants from MDDCs infected with HIV SF162 on plates coated with Tat wt (0.1 µM) or with control buffer (BSA). Infection was evaluated at the indicated time points by measuring p24 antigen in the culture supernatants in duplicate wells.
Figure 6. Anti-integrin antibodies block the enhancement…
Figure 6. Anti-integrin antibodies block the enhancement of MDDC infection induced by soluble or adherent Tat.
(A) pSF162LUC infection of MDDCs for 8 days in the presence of soluble cys22 Tat (0.1 or 1 µM) and a combination of mAbs directed against the α5β1, αvβ3 and αvβ5 integrins (10 µg/mL each) or the control isotype mAb (30 µg/mL). For each Tat concentration (Tat 0.1 µM, blu bar; Tat 1 µM, red bar) infection fold changes were calculated as the ratio between RLU from MDDCs pre-incubated with the control isotype mAb in the presence and in the absence of Tat (baseline RLU: 97) or as the ratio between RLU from MDDCs pre-incubated with anti-integrin mAbs in the presence and in the absence of Tat (baseline RLU: 112) (all values had been previously subtracted of the uninfected MDDC RLU background). Experiments were performed in duplicate. (B) Infection of MDDCs at different time points in the presence of coated wt Tat (0.01 µM for donor 1, 0.1 µM for donor 2) and of mAbs directed against the α5β1, αvβ3 and αvβ5 integrins (10 µg/mL each) or the control isotype mAb (30 µg/mL). Fold reduction of infection was calculated as the ratio between the content of p24 antigen detected in the supernatants of MDDCs seeded on adherent Tat in the presence of anti-integrin mAbs and that detected in the supernatants of MDDCs seeded on adherent Tat in the presence of the control isotype mAb (baseline, for donor 1, p24 was 186 and 479 pg/mL at day 7 and 10, respectively; for donor 2, p24 was 135 and 538 pg/mL at day 7 and 10, respectively). Experiments were performed in duplicate.
Figure 7. Entry of Tat or R5…
Figure 7. Entry of Tat or R5 Env-VLPs in MDDCs and block by anti-integrin antibodies.
(A) Entry of wt Tat into MDDCs from 3–10 different donors (depending on the anti-integrin mAbs used), in the presence of mAbs against the indicated integrins alone or combined, a control isotype mAb, or nil (buffer). (B) Entry of cys22 Tat into MDDCs from 3 different donors and block by the combined anti-integrin mAbs versus an Ig control isotype mAb. The percentages of Tat positive cells +/− standard deviations are shown. (C) Entry of VLP-R5Env (BaL) in MDDCs in the presence of Tat and block by anti-integrin mAbs or an Ig control isotype mAb. The percentages of fluorescent cells are shown. A representative experiment out of 4 performed is shown.
Figure 8. Tat-mediated entry of Env molecules…
Figure 8. Tat-mediated entry of Env molecules from different clades in MDDCs and block by anti-integrins antibodies.
(A) Clade B trimeric wt Env (twt Env), trimeric ΔV2 Env (tΔV2 Env), monomeric wt Env (mwt Env), or monomeric ΔV2 Env (mΔV2 Env) and (B) clade A and C trimeric wt Env (twt Env) molecules were incubated with control buffer or increasing concentrations of Tat, and then added to MDDCs at 1∶100 final dilution. Cells were then stained for intracellular Env. Open circles: control isotype mAb; filled circles: anti-integrin mAbs directed against the α5β1, αvβ3 and αvβ5 integrins (10 µg/mL each). Data from the same donor out of 3–8 donors tested are shown. Results are expressed as the percentage of Env-positive cells.
Figure 9. Tat/Env binding kinetics and thermodynamics.
Figure 9. Tat/Env binding kinetics and thermodynamics.
(A) Binding kinetics of trimeric wt Env (tw Env), trimeric ΔV2 Env (tΔV2 Env), monomeric wt Env (mwt Env) or monomeric ΔV2 Env (mΔV2 Env) to Tat bound to the Biacore chip. The range of concentrations of Env molecules is indicated. (B) Tat/Env binding thermodynamics by ITC. Tat was titrated with trimeric wt Env. The top panel shows calorimetric data versus time; the lower panel shows integrated areas normalized to the number of moles of Env subunits injected at each injection step.
Figure 10. Tat/Env complex and ternary Tat/Env/αvβ3…
Figure 10. Tat/Env complex and ternary Tat/Env/αvβ3 complex by modeling-docking calculations.
(A) ribbon representation of the Tat/Env complex showing that the Env CD4 binding site and the RGD domain of Tat are both exposed. Color code: ΔV1-2 Env: blue; Tat: red; Tat-RGD: yellow. (B) Surface representation of the ternary Tat/Env/αvβ3 complex. Color code: ΔV1-2 Env: green; Tat: purple; αvβ3 integrin: grey. See experimental procedures for details.
Figure 11. Tat/V3 loop interaction by ELISA…
Figure 11. Tat/V3 loop interaction by ELISA and ITC
(A). Tat interaction with cyclic (open circles) or linear (filled circles) V3 peptides by ELISA. Data are expressed as O.D. corrected for wells coated with the Tat buffer. (B) Tat interaction with cyclic (left panels) or linear (right panels) V3 peptides by ITC. Upper panels: calorimetry data versus time; lower panels: integrated areas normalized to the number of moles of V3 loop injected at each injection step.
Figure 12. Uptake of trimeric ΔV3 Env…
Figure 12. Uptake of trimeric ΔV3 Env by MDDCs and block by anti-integrins mAbs.
Uptake of ΔV2 Env or ΔV3 Env pre-incubated with buffer or increasing concentrations of Tat, in the presence or absence of anti-integrin mAbs (10 µg/mL each) or a control isotype mAb (30 µg/mL). Trimeric ΔV2 Env: circles; trimeric ΔV3 Env: squares. Control isotype Ab: open symbols; anti-integrin blocking mAbs combined: filled symbols.
Figure 13. Neutralization of Tat/Env complex entry…
Figure 13. Neutralization of Tat/Env complex entry in MDDCs by sera from HIV-infected individuals.
(A) Neutralization of trimeric ΔV2 Env entry in MDDCs by sera from HIV-infected subjects in the presence or absence of Tat in anti-Tat Ab negative (n = 8) and anti-Tat Ab positive (n = 8) subjects. The bars represent the percentage of entry of Env alone incubated in buffer (in blue) or with Tat (in red). The percentage of Env-positive cells is shown. Data are expressed as the mean with standard deviation of experiments performed in duplicate. The codes of the anti-Tat Ab negative or positive sera are indicated at the bottom of the bars. (B) Geometric mean (GM) of the ratio, with 95% confidence interval (CI) of the percentage of MDDCs internalizing Env in the absence (blue bar) or in the presence (red bar) of Tat in anti-Tat negative (n = 8) and anti-Tat positive (n = 8) subjects. Statistical analysis was performed by the two-tailed Student’s t-test.
Figure 14. Neutralization of Tat/Env complex entry…
Figure 14. Neutralization of Tat/Env complex entry or infection in MDDCs by sera from Tat-vaccinated HIV-infected individuals.
(A) Percentage of MDDCs internalizing Env in the presence of sera from trial subjects (n = 7) at baseline (week 0, anti-Tat Ab negative, in blue) and after Tat vaccination (week 48, anti-Tat Ab positive, in red) in the absence (empty bars) or in the presence (filled bars) of Tat. The codes of the sera are indicated at the bottom of the bars. (B) Geometric mean (GM) of the ratio (week 48 vs baseline), with 95% confidence interval (CI) of the percentage of MDDCs internalizing Env in the presence or in the absence of Tat in trial subjects (n = 7). Statistical analysis was performed by the two-tailed Student’s t-test. (C) MDDC infection performed in duplicate for 48 h with pSF162LUC in the presence of coated cys22 Tat (0.01 µM) or BSA (control) with sera from a representative trial subject before (baseline, anti-Tat Ab negative) and after Tat vaccination (anti-Tat positive). Sera prior to or after vaccination contained the same anti-Env Ab titers. Data are expressed as RLU.
Figure 15. Neutralization of Tat/Env complex entry…
Figure 15. Neutralization of Tat/Env complex entry in MDDCs by sera from Tat/Env-vaccinated monkeys.
(A) Percentage of MDDCs internalizing Env in the presence of plasma from Tat/Env vaccinated or control monkeys (6 animals/group) before (week 0, anti-Tat Ab negative, in blue) or after vaccination (week 38, anti-Tat Ab positive, in red) in the absence (empty bars) or in the presence (filled bars) of Tat. The codes of the plasma are indicated at the bottom of the bars: MU1-6, vaccinated monkeys; MU7-12, control monkeys. (B) Geometric mean (GM) of the ratio (week 38 vs baseline), with 95% confidence interval (CI) of the percentage of MDDCs internalizing Env in the presence or in the absence of Tat in vaccinated monkeys (n = 6). Statistical analysis was performed by the two-tailed Student’s t-test.
Figure 16. Outcome of DC infection in…
Figure 16. Outcome of DC infection in the absence or presence of Tat, anti-Env and/or anti-Tat antibodies.
Tat redirects HIV to RGD-binding integrins evading neutralization by anti-Env Abs and both anti-Env and anti-Tat Abs are required to block infection. Extracellular Tat released by infected neighbor cells binds to trimeric Env on HIV, decreasing recognition of C-type lectin receptors and promoting engagement of RGD-binding integrins, which are expressed by inflammatory DCs, macrophages and endothelial cells (ECs) present at the site of infection. As a result, virions escape neutralization by anti-Env Abs directed against high mannose determinants and enters target cells upon binding to RGD-binding integrins. Anti-Tat Abs neutralize this binding, preventing virus entry through RGD-binding integrins. DC-SIGN: Dendritic Cell-Specific Intercellular adhesion molecule-3-Grabbing Non-integrin; SIGN-R: homologue of DC-SIGN present on ECs; MR: mannose receptor.

References

    1. Rerks-Ngarm S, Pitisuttithum P, Nitayaphan S, Kaewkungwal J, Chiu J, et al. (2009) Vaccination with ALVAC and AIDSVAX to prevent HIV-1 infection in Thailand. N Engl J Med. 36: 2209–2220.
    1. Haynes BF, Gilbert PB, McElrath MJ, Zolla-Pazner S, Tomaras GD, et al. (2012) Immune-correlates analysis of an HIV-1 vaccine efficacy trial. N Engl J Med. 366: 1275–1286.
    1. Cafaro A, Macchia I, Maggiorella MT, Titti F, Ensoli B (2009). Innovative approaches to develop prophylactic and therapeutic vaccines against HIV/AIDS. In: Pharmaceutical Biotechnology (Guzman C.A. and Fuerstein G. Eds.), Chapter No. 14, 189–242.
    1. Cafaro A, Caputo A, Fracasso C, Maggiorella MT, Goletti D, et al. (1999) Control of SHIV-89.6P-infection of cynomolgus monkeys by HIV-1 Tat protein vaccine. Nat Med 5: 643–650.
    1. Cafaro A, Caputo A, Maggiorella MT, Baroncelli S, Fracasso C, et al. (2000) SHIV89.6P pathogenicity in cynomolgus monkeys and control of viral replication and disease onset by human immunodeficiency virus type 1 Tat vaccine. J Med Primatol 29: 193–208.
    1. Cafaro A, Titti F, Fracasso C, Maggiorella MT, Baroncelli S, et al. (2001) Vaccination with DNA containing tat coding sequences and unmethylated CpG motifs protects cynomolgus monkeys upon infection with simian/human immunodeficiency virus (SHIV89.6P). Vaccine 19: 2862–2877.
    1. Cafaro A, Bellino S, Titti F, Maggiorella MT, Sernicola L, et al. (2010) Impact of viral dose and major histocompatibility complex class IB haplotype on viral outcome in mauritian cynomolgus monkeys vaccinated with Tat upon challenge with simian/human immunodeficiency virus SHIV89.6P. J Virol 84: 8953–8958.
    1. Maggiorella MT, Baroncelli S, Michelini Z, Fanales-Belasio E, Moretti S, et al. (2004) Long-term protection against SHIV89.6P replication in HIV-1 Tat vaccinated cynomolgus monkeys. Vaccine 22: 3258–3269.
    1. Borsetti A, Baroncelli S, Maggiorella MT, Moretti S, Fanales-Belasio E, et al. (2009) Containment of infection in tat vaccinated monkeys after rechallenge with a higher dose of SHIV89.6P(cy243). Viral Immunol 22: 117–124.
    1. Ensoli B, Fiorelli V, Ensoli F, Lazzarin A, Visintini R, et al. (2008) Thetherapeutic phase I trial of the recombinant native HIV-1 Tat protein. AIDS 22: 2207–2209.
    1. Ensoli B, Fiorelli V, Ensoli F, Lazzarin A, Visintini R, et al. (2009) The preventive phase I trialwith the HIV-1 Tat-based vaccine. Vaccine 28: 371–378.
    1. Longo O, Tripiciano A, Fiorelli V, Bellino S, Scoglio A, et al. (2009) Phase I therapeutic trial of the HIV-1 Tat protein and long term follow-up. Vaccine 27: 3306–3312.
    1. Ensoli B, Bellino S, Tripiciano A, Longo O, Francavilla A, et al. (2010) Tat Reduces Immune Activation and Loss of Regulatory T-Cells and Improves Immune Function in Subjects on HAART. PLoS ONE 5: e13540.
    1. Ensoli B, Barillari G, Salahuddin SZ, Gallo RC, Wong-Staal F (1990) Tat protein of HIV-1 stimulates growth of cells derived from Kaposi’s sarcoma lesions of AIDS patients. Nature 345: 84–86.
    1. Ensoli B, Buonaguro L, Barillari G, Fiorelli V, Gendelman R, et al. (1993) Release, uptake, and effects of extracellular Human Immunodeficiency Virus type 1 Tat protein on cell growth and viral transactivation. J Virol 67: 277–287.
    1. Chang HC, Samaniego F, Nair BC, Buonaguro L, Ensoli B (1997) HIV-1 Tat protein exits from cells via a leaderless secretory pathway and binds to extracellular matrix-associated heparan sulfate proteoglycans through its basic region. AIDS 11: 1421–1431.
    1. Ensoli B, Gendelman R, Markham P, Fiorelli V, Colombini S, et al. (1994) Synergy between basic fibroblast growth factor and HIV-1 Tat protein in induction of Kaposi’s sarcoma. Nature 371: 674–680.
    1. Westendorp MO, Frank R, Ochsenbauer C, Stricker K, Dhein J, et al. (1995) Sensitization of T cells to CD95-mediated apoptosis by HIV-1 Tat and gp120. Nature 375: 497–500.
    1. Xiao H, Neuveut C, Tiffany HL, Benkirane M, Rich EA, et al. (2000) Selective CXCR4 antagonism by Tat: implications for in vivo expansion of coreceptor use by HIV-1. Proc Natl Acad Sci USA 97: 11466–11471.
    1. Marchio S, Alfano M, Primo L, Gramaglia D, Butini L, et al. (2005) Cell surface-associated Tat modulates HIV-1 infection and spreading through a specific interaction with gp120 viral envelope protein. Blood 105: 2802–2811.
    1. Rayne F, Debaisieux S, Yezid H, Lin YL, Mettling C, et al. (2010) Phosphatidylinositol-(4,5)-bisphosphate enables efficient secretion of HIV-1 Tat by infected T-cells. EMBO J. 29: 1348–1362.
    1. Barillari G, Gendelman R, Gallo RC, Ensoli B (1993) The Tat protein of human immunodeficiency virus type 1, a growth factor for AIDS Kaposi sarcoma and cytokine-activated vascular cells, induces adhesion of the same cell types by using integrin receptors recognizing the RGD amino acid sequence. Proc Natl Acad Sci U S A 90: 7941–7945.
    1. Barillari G, Sgadari C, Fiorelli V, Samaniego F, Colombini S, et al. (1999) The Tat protein of human immunodeficiency virus type-1 promotes vascular cell growth and locomotion by engaging the alpha5beta1 and alphavbeta3 integrins and by mobilizing sequestered basic fibroblast growth factor. Blood 94: 663–672.
    1. Barillari G, Sgadari C, Palladino C, Gendelman R, Caputo A, et al. (1999) Inflammatory cytokines synergize with the HIV-1 Tat protein to promote angiogenesis and Kaposi’s sarcoma via induction of basic fibroblast growth factor and the alpha v beta 3 integrin. J Immunol 163: 1929–1935.
    1. Davis LS, Sackler M, Brezinschek RI, Lightfoot E, Bailey JL, et al. (2002) Inflammation, immune reactivity, and angiogenesis in a severe combined immunodeficiency model of rheumatoid arthritis. Am J Pathol 160: 357–367.
    1. Koning GA, Schiffelers RM, Wauben MH, Kok RJ, Mastrobattista E, et al. (2006) Targeting of angiogenic endothelial cells at sites of inflammation by dexamethasone phosphate-containing RGD peptide liposomes inhibits experimental arthritis. Arthritis Rheum 54: 1198–1208.
    1. Stupack DG, Cheresh DA (2004) Integrins and angiogenesis. Curr Top Dev Biol 64: 207–238.
    1. Vogel BE, Lee SJ, Hildebrand A, Craig W, Pierschbacher MD, et al. (1993) A novel integrin specificity exemplified by binding of the alpha v beta 5 integrin to the basic domain of the HIV Tat protein and vitronectin. J Cell Biol 121: 461–468.
    1. Fanales-Belasio E, Moretti S, Nappi F, Barillari G, Micheletti F, et al. (2002) Native HIV-1 Tat protein targets monocyte-derived dendritic cells and enhances their maturation, function, and antigen-specific T cell responses. J Immunol 168: 197–206.
    1. Fanales-Belasio E, Moretti S, Fiorelli V, Tripiciano A, Pavone Cossut MR, et al. (2009) HIV-1 Tat addresses dendritic cells to induce a predominant Th1-type adaptive immune response that appears prevalent in the asymptomatic stage of infection. J Immunol 182: 2888–2897.
    1. Gavioli R, Gallerani E, Fortini C, Fabris M, Bottoni A, et al. (2004) HIV-1 tat protein modulates the generation of cytotoxic T cell epitopes by modifying proteasome composition and enzymatic activity. J Immunol 173: 3838–3843.
    1. Gavioli R, Cellini S, Castaldello A, Voltan R, Gallerani E, et al. (2008) The Tat protein broadens T cell responses directed to the HIV-1 antigens Gag and Env: implications for the design of new vaccination strategies against AIDS. Vaccine 26: 727–737.
    1. Nappi F, Chiozzini C, Bordignon V, Borsetti A, Bellino S, et al. (2009) Immobilized HIV-1 Tat protein promotes gene transfer via a transactivation-independent mechanism which requires binding of Tat to viral particles. J Gene Med 11: 955–965.
    1. Re MC, Vignoli M, Furlini G, Gibellini D, Colangeli V, et al. (2001) Antibodies against full-length Tat protein and some low-molecular-weight Tat-peptides correlate with low or undetectable viral load in HIV-1 seropositive patients. J Clin Virol 21: 81–89.
    1. Reiss P, de Wolf F, Kuiken CL, de Ronde A, Dekker J, et al. (1991) Contribution of antibody response to recombinant HIV-1 gene-encoded proucts nef, rev, tat, and protease in predicting development of AIDS in HIV-1-infected individuals. J Acquir Immune Defic Syndr 4: 165–172.
    1. Zagury JF, Sill A, Blattner W, Lachgar A, Le Buanec H, et al. (1998) Antibodies to the HIV-1 Tat protein correlated with non progression to AIDS: a rationale for the use of Tat toxoid as an HIV-1 vaccine. J Hum Virol 1: 282–292.
    1. Richardson MW, Mirchandani J, Duong J, Grimaldo S, Kocieda V, et al. (2003) Antibodies to Tat and Vpr in the GRIV cohort: differential association with maintenance of long-term non progression status in HIV-1 infection. Biomed Pharmacother 57: 4–14.
    1. Rezza G, Fiorelli V, Dorrucci M, Ciccozzi M, Tripiciano A, et al. (2005) The presence of anti-Tat antibodies is predictive of long-term non progression to AIDS or severe immunodeficiency: findings in a cohort of HIV-1 seroconverters. J Infect Dis 191: 1321–1324.
    1. Ensoli B, Cafaro A, Caputo A, Fiorelli V, Ensoli F, et al. (2005) Vaccines based on the native HIV Tat protein and on the combination of Tat and the structural HIV protein variant DeltaV2 Env. Microbes Infect 7: 1392–1399.
    1. Demberg T, Florese RH, Heath MJ, Larsen K, Kalisz I, et al. (2007) A replication-competent adenovirus-human immunodeficiency virus (Ad-HIV) tat and Ad-HIV env priming/Tat and envelope protein boosting regimen elicits enhanced protective efficacy against simian/human immunodeficiency virus SHIV89.6P challenge in rhesus macaques. J Virol 81: 3414–3427.
    1. Ferrantelli F, Maggiorella MT, Schiavoni I, Sernicola L, Olivieri E, et al. (2011) A combination HIV vaccine based on Tat and Env proteins was immunogenic and protected macaques from mucosal SHIV challenge in a pilot study. Vaccine 29: 2918–2932.
    1. Ferrantelli F, Cafaro A, Ensoli B (2004) Nonstructural HIV proteins as targets for prophylactic or therapeutic vaccines. Curr Opin Biotechnol 15: 543–556.
    1. Rossi C, Balboni PG, Betti M, Marconi PC, Bozzini R, et al. (1997) Inhibition of HIV-1 replication by a Tat transdominant negative mutant in human peripheral blood lymphocytes from healthy donors and HIV-1-infected patients. Gene Ther 4: 1261–1269.
    1. Srivastava IK, Stamatatos L, Kan E, Vajdy M, Lian Y, et al. (2003) Purification, characterization, and immunogenicity of a soluble trimeric envelope protein containing a partial deletion of the V2 loop derived from SF162, an R5-tropic human immunodeficiency virus type 1 isolate. J Virol 77: 11244–11259.
    1. Saunders CJ, McCaffrey RA, Zharkikh I, Kraft Z, Malenbaum SE, et al. (2005) The V1, V2, and V3 regions of the human immunodeficiency virus type 1 envelope differentially affect the viral phenotype in an isolate-dependent manner. J Virol 79: 9069–9080.
    1. Pizza M, Giuliani MM, Fontana MR, Monaci E, Douce G, et al. (2001) Mucosal vaccines: non toxic derivatives of LT and CT as mucosal adjuvants. Vaccine 19: 2534–2541.
    1. Goletti D, Macchia I, Leone P, Pace M, Sernicola L, et al. (2006) Innate anti-viral immunity is associated with the protection elicited by the simian immunodeficiency virus (SIV) live attenuated virus vaccine in cynomolgus monkeys. Med Sci Monit 12: BR330–340.
    1. Negri RM, Buffa V, Leone P, Bona R, Borghi M, et al. (2004) Use of retroviral vectors for the analysis of SIV/HIV-specific CD8 T cell responses. J Immunol Methods 291: 153–163.
    1. Yu X, Zhan X, D’Costa J, Tanavde VM, Ye Z, et al. (2003) Lentiviral vectors with two independent internal promoters transfer high-level expression of multiple transgenes to human hematopoietic stem-progenitor cells. Mol Ther 7: 827–838.
    1. Mochizuki H, Schwartz JP, Tanaka K, Brady RO, Reiser J (1998) High-titer human immunodeficiency virus type 1-based vector systems for gene delivery into nondividing cells. J Virol 72: 8873–8883.
    1. Rossi GB, Verani P, Macchi B, Federico M, Orecchia A, et al. (1987) Recovery of HIV-related retroviruses from Italian patients with AIDS or AIDS-related complex and from asymptomatic at-risk individuals. Ann N Y Acad Sci 511: 390–400.
    1. Huang L, Bosch I, Hofmann W, Sodroski J, Pardee AB (1998) Tat protein induces human immunodeficiency virus type 1 (HIV-1) coreceptors and promotes infection with both macrophage-tropic and T-lymphotropic HIV-1 strains. J Virol 72: 8952–8960.
    1. Muratori C, D’Aloja P, Superti F, Tinari A, Sol-Foulon N, et al. (2006) Generation and characterization of a stable cell population releasing fluorescent HIV-1-based Virus Like Particles in an inducible way. BMC. Biotechnol 6: 52.
    1. Sali A, Blundell TL (1993) Comparative protein modelling by satisfaction of spatial restraints. J Mol Biol 234: 779–815.
    1. Case DA, Cheatham TE 3rd, Darden T, Gohlke H, Luo R, et al (2005) The Amber biomolecular simulation programs. J Comput Chem 26: 1668–1688.
    1. Péloponèse JM Jr, Grégoire C, Opi S, Esquieu D, Sturgis J, et al. (2000) 1H-13C nuclear magnetic resonance assignment and structural characterization of HIV-1 Tat protein. C R Acad Sci III 323: 883–94.
    1. Grégoire C, Péloponèse JM Jr, Esquieu D, Opi S, Campbell G, et al. (2001) Homonuclear (1)H-NMR assignment and structural characterization of human immunodeficiency virus type 1 Tat Mal protein. Biopolymers 62: 324–35.
    1. Chen B, Vogan EM, Gong H, Skehel JJ, Wiley DC, et al. (2005) Structure of an unliganded simian immunodeficiency virus gp120 core. Nature 433: 834–841.
    1. Huang CC, Tang M, Zhang MY, Majeed S, Montabana E, et al. (2005) Structure of a V3-containing HIV-1 gp120 core. Science 310: 1025–1028.
    1. Palma PN, Krippahl L, Wampler JE, Moura JJ (2000) BiGGER: a new (soft) docking algorithm for predicting protein interactions. Proteins 39: 372–384.
    1. Comeau SR, Gatchell DW, Vajda S, Camacho CJ (2004) ClusPro: a fully automated algorithm for protein-protein docking. Nucleic Acids Res. 32: W96–W99.
    1. Dominguez C, Boelens R, Bonvin AM (2003) HADDOCK: a protein-protein docking approach based on biochemical or biophysical information. J Am Chem Soc 125: 1731–1737.
    1. Xiong JP, Stehle T, Zhang R, Joachimiak A, Frech M, et al. (2002) Crystal structure of the extracellular segment of integrin alpha Vbeta3 in complex with an Arg-Gly-Asp ligand. Science 296: 151–155.
    1. Dull T, Zufferey R, Kelly M, Mandel RJ, Nguyen M, et al. (1998) A third-generation lentivirus vector with a conditional packaging system. J Virol 72(11): 8463–71.
    1. Brown KA, Bedford P, Macey M, McCarthy DA, Leroy F, et al. (1997) Human blood dendritic cells: binding to vascular endothelium and expression of adhesion molecules. Clin Exp Immunol 107: 601–607.
    1. Benelli R, Mortarini R, Anichini A, Giunciuglio D, Noonan DM, et al. (1998) Monocyte-derived dendritic cells and monocytes migrate to HIV-Tat RGD and basic peptides. AIDS 12: 261–268.
    1. Ammon C, Meyer SP, Schwarzfischer L, Krause SW, Andreesen R, et al. (2000) Comparative analysis of integrin expression on monocyte-derived macrophages and monocyte-derived dendritic cells. Immunology 100: 364–369.
    1. Puig-Kröger A, Sanz-Rodríguez F, Longo N, Sánchez-Mateos P, Botella L, et al. (2000) Maturation-dependent expression and function of the CD49d integrin on monocyte-derived human dendritic cells. J Immunol 165: 4338–4345.
    1. Sada-Mikami R, Heike Y, Kanai S, Azuma M, Shirakawa K, et al. (2001) Efficient gene transduction by RGD-fiber modified recombinant adenovirus into dendritic cells. Jpn J Cancer Res 92: 321–327.
    1. Barczyk M, Carracedo S, Gullberg D (2010) Integrins. Cell Tissue Res 339: 269–280.
    1. Acharya AP (2010) The modulation of dendritic cell integrin binding and activation by RGD-peptide density gradient substrates. Biomaterials 31: 7444–7454.
    1. Luster AD, Alon R, von Andrian UH (2005) Immune cell migration in inflammation: present and future therapeutic targets. Nat Immunol 6: 1182–1190.
    1. Haase AT (2010) Targeting early infection to prevent HIV-1 mucosal transmission. Nature 11: 217–223.
    1. Marchiò S, Alfano M, Primo L, Gramaglia D, Butini L, et al. (2005) Cell surface-associated Tat modulates HIV-1 infection and spreading through a specific interaction with gp120 viral envelope protein. Blood 105: 2802–2811.
    1. Chohan B, Lang D, Sagar M, Korber B, Lavreys L, et al. (2005) Selection for human immunodeficiency virus type 1 envelope glycosylation variants with shorter V1-V2 loop sequences occurs during transmission of certain genetic subtypes and may impact viral RNA levels. J Virol 79: 6528–6531.
    1. Derdeyn CA, Decker JM, Bibollet-Ruche F, Mokili JL, Muldoon M, et al. (2004) Envelope-constrained neutralization-sensitive HIV-1 after heterosexual transmission. Science 303: 2019–2022.
    1. Rong R, Bibollet-Ruche F, Mulenga J, Allen S, Blackwell JL, et al. (2007) Role of V1V2 and other human immunodeficiency virus type 1 envelope domains in resistance to autologous neutralization during clade C infection. J Virol 81: 1350–1359.
    1. Sagar M, Wu X, Lee S, Overbaugh J (2006) Human immunodeficiency virus type 1 V1-V2 envelope loop sequences expand and add glycosylation sites over the course of infection, and these modifications affect antibody neutralization sensitivity. J Virol 80: 9586–9598.
    1. Barnett SW, Lu S, Srivastava I, Cherpelis S, Gettie A, et al. (2001) The ability of an oligomeric human immunodeficiency virus type 1 (HIV-1) envelope antigen to elicit neutralizing antibodies against primary HIV-1 isolates is improved following partial deletion of the second hypervariable region. J Virol 75: 5526–5540.
    1. Cao J, Sullivan N, Desjardin E, Parolin C, Robinson J, et al. (1997) Replication and neutralization of human immunodeficiency virus type 1 lacking the V1 and V2 variable loops of the gp120 envelope glycoprotein. J Virol 71: 9808–9812.
    1. Stamatatos L, Wiskerchen M, Cheng-Mayer C (1998) Effect of major deletions in the V1 and V2 loops of a macrophage-tropic HIV type 1 isolate on viral envelope structure, cell entry, and replication. AIDS Res Hum Retroviruses 14: 1129–1139.
    1. Wyatt R, Moore J, Accola M, Desjardin E, Robinson J, et al. (1995) Involvement of the V1/V2 variable loop structure in the exposure of human immunodeficiency virus type 1 gp120 epitopes induced by receptor binding. J Virol 69: 5723–5733.
    1. Ensoli B, Fiorelli V, Ensoli F, Cafaro A, Titti F, et al. (2006) Candidate HIV-1 Tat vaccine development: from basic science to clinical trials. AIDS 20: 2245–2261.

Source: PubMed

3
Předplatit