451
|
Planque S, Salas M, Mitsuda Y, Sienczyk M, Escobar MA, Mooney JP, Morris MK, Nishiyama Y, Ghosh D, Kumar A, Gao F, Hanson CV, Paul S. Neutralization of genetically diverse HIV-1 strains by IgA antibodies to the gp120-CD4-binding site from long-term survivors of HIV infection. AIDS 2010; 24:875-84. [PMID: 20186035 PMCID: PMC2881561 DOI: 10.1097/qad.0b013e3283376e88] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To identify an HIV epitope suitable for vaccine development. DESIGN Diverse HIV-1 strains express few structurally constant regions on their surface vulnerable to neutralizing antibodies. The mostly conserved CD4-binding site (CD4BS) of gp120 is essential for host cell binding and infection by the virus. Antibodies that recognize the CD4BS are rare, and one component of the CD4BS, the 421-433 peptide region, expresses B-cell superantigenic character, a property predicted to impair the anti-CD4BS adaptive immune response. METHODS IgA samples purified from the plasma of patients with HIV infection were analyzed for the ability to bind synthetic mimetics containing the 416-433 gp120 region and full-length gp120. Infection of peripheral blood mononuclear cells by clinical HIV isolates was measured by p24 ELISA. RESULTS IgA preparations from three patients with subtype B infection for 19-21 years neutralized heterologous, coreceptor CCR5-dependent subtype A, B, C, D, and AE strains with exceptional potency. The IgAs displayed specific binding of a synthetic 416-433 peptide mimetic dependent on recognition of the CD4-binding residues located in this region. Immunoadsorption, affinity chromatography, and mutation procedures indicated that HIV neutralization occurred by IgA recognition of the CD4BS. CONCLUSION These observations identify the 421-433 peptide region as a vulnerable HIV site to which survivors of infection can produce powerful neutralizing antibodies. This indicates that the human immune system can bypass restrictions on the adaptive B cell response to the CD4BS, opening the route to targeting the 421-433 region for attaining control of HIV infection.
Collapse
Affiliation(s)
- Stephanie Planque
- Chemical Immunology Research Center and Gulf States Hemophilia and Thrombophilia Center, Departments of Pathology and Pediatrics, University of Texas–Houston Medical School, Houston, Texas 77030, U.S.A
| | - Maria Salas
- Viral and Rickettsial Disease Laboratory, California Department of Public Health, Richmond, California 94804, U.S.A
| | - Yukie Mitsuda
- Chemical Immunology Research Center and Gulf States Hemophilia and Thrombophilia Center, Departments of Pathology and Pediatrics, University of Texas–Houston Medical School, Houston, Texas 77030, U.S.A
| | - Marcin Sienczyk
- Chemical Immunology Research Center and Gulf States Hemophilia and Thrombophilia Center, Departments of Pathology and Pediatrics, University of Texas–Houston Medical School, Houston, Texas 77030, U.S.A
| | - Miguel A. Escobar
- Chemical Immunology Research Center and Gulf States Hemophilia and Thrombophilia Center, Departments of Pathology and Pediatrics, University of Texas–Houston Medical School, Houston, Texas 77030, U.S.A
| | - Jason P. Mooney
- Viral and Rickettsial Disease Laboratory, California Department of Public Health, Richmond, California 94804, U.S.A
| | - Mary-Kate Morris
- Viral and Rickettsial Disease Laboratory, California Department of Public Health, Richmond, California 94804, U.S.A
| | - Yasuhiro Nishiyama
- Chemical Immunology Research Center and Gulf States Hemophilia and Thrombophilia Center, Departments of Pathology and Pediatrics, University of Texas–Houston Medical School, Houston, Texas 77030, U.S.A
| | - Dipanjan Ghosh
- Chemical Immunology Research Center and Gulf States Hemophilia and Thrombophilia Center, Departments of Pathology and Pediatrics, University of Texas–Houston Medical School, Houston, Texas 77030, U.S.A
| | - Amit Kumar
- Department of Medicine, Duke University, Durham, North Carolina 27710, U.S.A
| | - Feng Gao
- Department of Medicine, Duke University, Durham, North Carolina 27710, U.S.A
| | - Carl V. Hanson
- Viral and Rickettsial Disease Laboratory, California Department of Public Health, Richmond, California 94804, U.S.A
| | - Sudhir Paul
- Chemical Immunology Research Center and Gulf States Hemophilia and Thrombophilia Center, Departments of Pathology and Pediatrics, University of Texas–Houston Medical School, Houston, Texas 77030, U.S.A
| |
Collapse
|
452
|
Brower ET, Schön A, Freire E. Naturally occurring variability in the envelope glycoprotein of HIV-1 and development of cell entry inhibitors. Biochemistry 2010; 49:2359-67. [PMID: 20166763 PMCID: PMC2840190 DOI: 10.1021/bi1000933] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Naturally occurring genetic variability across HIV-1 subtypes causes amino acid polymorphisms in encoded HIV-1 proteins including the envelope glycoproteins associated with viral entry. The effects of amino acid polymorphisms on the mechanism of HIV-1 entry into cells, a process initiated by the binding of the viral envelope glycoprotein gp120 to the cellular CD4 receptor, are largely unknown. In this study, we demonstrate that amino acid polymorphisms affect the structural stability and domain cooperativity of gp120 and that those differences are reflected in the binding mechanism of the viral envelope glycoprotein to the cell surface receptor and coreceptor. Moreover, subtype differences also affect the binding behavior of experimental HIV cell entry inhibitors. While gp120-A has a slightly lower denaturation temperature than gp120-B, the most notable stability difference is that for gp120-B the van't Hoff to calorimetric enthalpy ratio (DeltaH(vH)/DeltaH) is 0.95 whereas for gp120-A is 0.6, indicative of more cooperative domain/domain interactions in gp120-B, as this protein more closely approaches a two-state transition. Isothermal titration calorimetry demonstrates that CD4 and 17b (a surrogate antibody for the chemokine coreceptor) exhibit 7- and 3-fold weaker binding affinities for gp120-A. The binding of these proteins as well as that of the experimental entry inhibitor NBD-556 induces smaller conformational changes in gp120-A as evidenced by significantly smaller binding enthalpies and binding entropies. Together, these results describe the effects of gp120 polymorphisms on binding to host cell receptors and emphasize that guidelines for developing future entry inhibitors must recognize and deal with genomic differences between HIV strains.
Collapse
Affiliation(s)
- Evan T. Brower
- Department of Biology, The Johns Hopkins University, Baltimore, Maryland 21218
| | - Arne Schön
- Department of Biology, The Johns Hopkins University, Baltimore, Maryland 21218
| | - Ernesto Freire
- Department of Biology, The Johns Hopkins University, Baltimore, Maryland 21218
| |
Collapse
|
453
|
Finzi A, Xiang SH, Pacheco B, Wang L, Haight J, Kassa A, Danek B, Pancera M, Kwong PD, Sodroski J. Topological layers in the HIV-1 gp120 inner domain regulate gp41 interaction and CD4-triggered conformational transitions. Mol Cell 2010; 37:656-67. [PMID: 20227370 PMCID: PMC2854584 DOI: 10.1016/j.molcel.2010.02.012] [Citation(s) in RCA: 183] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2009] [Revised: 01/14/2010] [Accepted: 02/13/2010] [Indexed: 11/18/2022]
Abstract
The entry of human immunodeficiency virus (HIV-1) into cells is initiated by binding of the gp120 exterior envelope glycoprotein to the receptor, CD4. How does CD4 binding trigger conformational changes in gp120 that allow the gp41 transmembrane envelope glycoprotein to mediate viral-cell membrane fusion? The transition from the unliganded to the CD4-bound state is regulated by two potentially flexible topological layers (layers 1 and 2) in the gp120 inner domain. Both layers apparently contribute to the noncovalent association of unliganded gp120 with gp41. After CD4 makes initial contact with the gp120 outer domain, layer 1-layer 2 interactions strengthen gp120-CD4 binding by reducing the off rate. Layer 1-layer 2 interactions also destabilize the activated state induced on HIV-1 by treatment with soluble CD4. Thus, despite lack of contact with CD4, the gp120 inner-domain layers govern CD4 triggering by participating in conformational transitions within gp120 and regulating the interaction with gp41.
Collapse
Affiliation(s)
- Andrés Finzi
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Department of Pathology, Division of AIDS, Harvard Medical School, Boston, MA 02115, U.S.A
| | - Shi-Hua Xiang
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Department of Pathology, Division of AIDS, Harvard Medical School, Boston, MA 02115, U.S.A
| | - Beatriz Pacheco
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Department of Pathology, Division of AIDS, Harvard Medical School, Boston, MA 02115, U.S.A
| | - Liping Wang
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Department of Pathology, Division of AIDS, Harvard Medical School, Boston, MA 02115, U.S.A
| | - Jessica Haight
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Department of Pathology, Division of AIDS, Harvard Medical School, Boston, MA 02115, U.S.A
| | - Aemro Kassa
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Department of Pathology, Division of AIDS, Harvard Medical School, Boston, MA 02115, U.S.A
| | - Brenda Danek
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Department of Pathology, Division of AIDS, Harvard Medical School, Boston, MA 02115, U.S.A
| | - Marie Pancera
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, U.S.A
| | - Peter D. Kwong
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, U.S.A
| | - Joseph Sodroski
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Department of Pathology, Division of AIDS, Harvard Medical School, Boston, MA 02115, U.S.A
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA 02115, U.S.A
| |
Collapse
|
454
|
[Theory for prophylactic AIDS vaccine development]. Uirusu 2010; 59:267-75. [PMID: 20218335 DOI: 10.2222/jsv.59.267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
In the natural courses of human immunodeficiency virus (HIV) infections, host immune responses fail to contain the virus and allow persistent HIV replication, leading to AIDS progression. For development of an effective vaccine against those viral infections which do not show spontaneous remission, it is important to elucidate which immune responses to be induced for viral control. This review focuses on antibodies and cytotoxic T lymphocytes, key adaptive immune effectors, and discusses possible mechanisms for HIV control by vaccine-induced antibody, memory B lymphocyte, and (effector and central) memory T lymphocyte responses. Finally, we mention the ongoing international project for a clinical trial of our Sendai virus vector-based AIDS vaccine.
Collapse
|
455
|
Neutralizing antibodies against HIV-1: can we elicit them with vaccines and how much do we need? Curr Opin HIV AIDS 2010; 4:347-51. [PMID: 20048696 DOI: 10.1097/coh.0b013e32832f4a4d] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
PURPOSE OF REVIEW This review describes some of the major obstacles that have impeded progress in the development of an effective neutralizing antibody-based HIV-1 vaccine and explains why it may be possible to overcome these obstacles. A renewed interest in the B-cell response in HIV-1-infected individuals is emphasized. RECENT FINDINGS New assay technologies and access to large numbers of clinical specimens have permitted a detailed assessment of the neutralizing antibody response in HIV-1-infected individuals. Recent studies have demonstrated that B cells can be stimulated to generate high titers of broadly cross-reactive neutralizing antibodies against multiple genetic subtypes of the virus. Preliminary evidence suggests that some of these antibodies are directed against epitopes in the CD4 binding site on monomeric gp120, whereas many others are directed against epitopes that remain to be identified. SUMMARY The rationale for pursuing an effective neutralizing antibody-based HIV-1 vaccine is strengthened by the recent demonstration of potent neutralizing antibody responses in a subset of HIV-1-infected individuals. Information on how this response develops and what epitopes are targeted could provide the insights that are needed to design improved vaccine strategies.
Collapse
|
456
|
Abstract
PURPOSE OF REVIEW This review summarizes current and novel virologic reagents employed for the development and application of in-vitro assays that assess neutralizing activity of antibodies against HIV-1. Characteristics of several virologic approaches are placed in context with various cellular targets and assay read-outs intended to determine potency and breadth of neutralization in patient cohorts and clinical vaccine trials. RECENT FINDINGS New molecular virologic reagents developed for in-vitro primary cell-based assays promise to facilitate rigorous and standardized assessment of anti-HIV-1-neutralizing antibody responses elicited by vaccine immunogens. SUMMARY Comprehensive assessment of anti-HIV-1 antibody potency and breadth is essential for evaluating vaccine immunogens, the advancement of vaccine candidates into clinical trials, and ultimately the development of effective vaccine strategies. Env-pseudovirion and recombinant reporter cell line neutralization assays are important tools for rapid and standardized measurement of neutralizing antibody activity. However, recent studies indicate that reporter cell lines fail to detect neutralization activity of certain antibodies observed when analyzed in peripheral blood mononuclear cells and may yield results on neutralizing antibody breadth that are discordant with peripheral blood mononuclear cell assays. Importantly, it remains unknown whether current in-vitro assays may be predictive of a protective neutralizing antibody response elicited by vaccine immunogens. This situation underscores the significance of standardizing existing, complementary methods as well as developing new assay concepts that assess neutralization in primary cells. Thus, this chapter focuses on new virologic reagents that promise to facilitate reaching this goal.
Collapse
|
457
|
Large-scale conformational sampling of proteins using temperature-accelerated molecular dynamics. Proc Natl Acad Sci U S A 2010; 107:4961-6. [PMID: 20194785 DOI: 10.1073/pnas.0914540107] [Citation(s) in RCA: 133] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
We show how to apply the method of temperature-accelerated molecular dynamics (TAMD) in collective variables [Maragliano L, Vanden-Eijnden E (2006) Chem Phys Lett 426:168-175] to sample the conformational space of multidomain proteins in all-atom, explicitly solvated molecular dynamics simulations. The method allows the system to hyperthermally explore the free-energy surface in a set of collective variables computed at the physical temperature. As collective variables, we pick Cartesian coordinates of centers of contiguous subdomains. The method is applied to the GroEL subunit, a 55-kDa, three-domain protein, and HIV-1 gp120. For GroEL, the method induces in about 40 ns conformational changes that recapitulate the t --> r('') transition and are not observed in unaccelerated molecular dynamics: The apical domain is displaced by 30 A, with a twist of 90 degrees relative to the equatorial domain, and the root-mean-squared deviation relative to the r('') conformer is reduced from 13 to 5 A, representing fairly high predictive capability. For gp120, the method predicts both counterrotation of inner and outer domains and disruption of the so-called bridging sheet. In particular, TAMD on gp120 initially in the CD4-bound conformation visits conformations that deviate by 3.6 A from the gp120 conformer in complex with antibody F105, again reflecting good predictive capability. TAMD generates plausible all-atom models of the so-far structurally uncharacterized unliganded conformation of HIV-1 gp120, which may prove useful in the development of inhibitors and immunogens. The fictitious temperature employed also gives a rough estimate of 10 kcal/mol for the free-energy barrier between conformers in both cases.
Collapse
|
458
|
Lagenaur LA, Villarroel VA, Bundoc V, Dey B, Berger EA. sCD4-17b bifunctional protein: extremely broad and potent neutralization of HIV-1 Env pseudotyped viruses from genetically diverse primary isolates. Retrovirology 2010; 7:11. [PMID: 20158904 PMCID: PMC2843639 DOI: 10.1186/1742-4690-7-11] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2009] [Accepted: 02/16/2010] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND We previously described a potent recombinant HIV-1 neutralizing protein, sCD4-17b, composed of soluble CD4 attached via a flexible polypeptide linker to an SCFv of the 17b human monoclonal antibody directed against the highly conserved CD4-induced bridging sheet of gp120 involved in coreceptor binding. The sCD4 moiety of the bifunctional protein binds to gp120 on free virions, thereby enabling the 17b SCFv moiety to bind and block the gp120/coreceptor interaction required for entry. The previous studies using the MAGI-CCR5 assay system indicated that sCD4-17b (in concentrated cell culture medium, or partially purified) potently neutralized several genetically diverse HIIV-1 primary isolates; however, at the concentrations tested it was ineffective against several other strains despite the conservation of binding sites for both CD4 and 17b. To address this puzzle, we designed variants of sCD4-17b with different linker lengths, and tested the neutralizing activities of the immunoaffinity purified proteins over a broader concentration range against a large number of genetically diverse HIV-1 primary isolates, using the TZM-bl Env pseudotype assay system. We also examined the sCD4-17b sensitivities of isogenic viruses generated from different producer cell types. RESULTS We observed that immunoaffinity purified sCD4-17b effectively neutralized HIV-1 pseudotypes, including those from HIV-1 isolates previously found to be relatively insensitive in the MAGI-CCR5 assay. The potencies were equivalent for the original construct and a variant with a longer linker, as observed with both pseudotype particles and infectious virions; by contrast, a construct with a linker too short to enable simultaneous binding of the sCD4 and 17b SCFv moieties was much less effective. sCD4-17b displayed potent neutralizing activity against 100% of nearly 4 dozen HIV-1 primary isolates from diverse genetic subtypes (clades A, B, C, D, F, and circulating recombinant forms AE and AG). The neutralization breadth and potency were superior to what have been reported for the broadly neutralizing monoclonal antibodies IgG b12, 2G12, 2F5, and 4E10. The activity of sCD4-17b was found to be similar against isogenic virus particles from infectious molecular clones derived either directly from the transfected producer cell line or after a single passage through PBMCs; this contrasted with the monoclonal antibodies, which were less potent against the PMBC-passaged viruses. CONCLUSIONS The results highlight the extremely potent and broad neutralizing activity of sCD4-17b against genetically diverse HIV-1 primary isolates. The bifunctional protein has potential applications for antiviral approaches to combat HIV infection.
Collapse
Affiliation(s)
- Laurel A Lagenaur
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Vadim A Villarroel
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Virgilio Bundoc
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Barna Dey
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Edward A Berger
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
459
|
Berry JD, Hay K, Rini JM, Yu M, Wang L, Plummer FA, Corbett CR, Andonov A. Neutralizing epitopes of the SARS-CoV S-protein cluster independent of repertoire, antigen structure or mAb technology. MAbs 2010; 2:53-66. [PMID: 20168090 DOI: 10.4161/mabs.2.1.10788] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Neutralizing antibody responses to the surface glycoproteins of enveloped viruses play an important role in immunity. Many of these glycoproteins, including the severe acute respiratory syndrome-coronavirus (SARS-CoV) spike (S) protein form trimeric units in the membrane of the native virion. There is substantial experimental and pre-clinical evidence showing that the S protein is a promising lead for vaccines and therapeutics. Previously we generated a panel of monoclonal antibodies (mAbs) to whole inactivated SARS-CoV which neutralize the virus in vitro. Here, we define their specificity and affinity, map several of their epitopes and lastly characterise chimeric versions of them. Our data show that the neutralizing mAbs bind to the angiotensin-converting enzyme 2 (ACE2) receptor-binding domain (RBD) of the SARS S protein. Three of the chimeric mAbs retain their binding specificity while one conformational mAb, F26G19, lost its ability to bind the S protein despite high level expression. The affinity for recombinant S is maintained in all of the functional chimeric versions of the parental mAbs. Both parental mAb F26G18 and the chimeric version neutralize the TO R2 strain of SARS-CoV with essentially identical titres (2.07 and 2.47 nM, respectively). Lastly, a comparison with other neutralizing mAbs to SARS-CoV clearly shows that the dominance of a 33 amino acid residue loop of the SARS-CoV RBD is independent of repertoire, species, quaternary structure, and importantly, the technology used to derive the mAbs. In cases like this, the dominance of a compact RBD antigenic domain and the central role of the S protein in pathogenesis may inherently create immunoselection pressure on viruses to evolve more complex evasion strategies or die out of a host species. The apparent simplicity of the mechanism of SARS-CoV neutralization is in stark contrast to the complexity shown by other enveloped viruses.
Collapse
Affiliation(s)
- Jody D Berry
- Department of Medical Microbiology, University of Manitoba, Winnipeg, Canada.
| | | | | | | | | | | | | | | |
Collapse
|
460
|
Evaluation of CD4-CD4i antibody architectures yields potent, broadly cross-reactive anti-human immunodeficiency virus reagents. J Virol 2010; 84:261-9. [PMID: 19864392 DOI: 10.1128/jvi.01528-09] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The envelope glycoprotein of human immunodeficiency virus type 1 (HIV-1) has several adaptations that allow the virus to evade antibody neutralization. Nevertheless, a few broadly cross-reactive neutralizing antibodies as well as reagents containing portions of CD4, the HIV receptor, have demonstrated partial efficacy in suppressing viral replication. One type of reagent designed for improved HIV neutralization fuses the CD4 D1-D2 domains to the variable regions of an antibody recognizing the CD4-induced (CD4i) coreceptor binding site on the gp120 portion of the HIV envelope spike. We designed, expressed, purified, and tested the neutralization potencies of CD4-CD4i antibody reagents with different architectures, antibody combining sites, and linkers. We found that fusing CD4 to the heavy chain of the CD4i antibody E51 yields a bivalent reagent including an antibody Fc region that expresses well, is expected to have a long serum half-life, and has comparable or greater neutralization activity than well-known broadly neutralizing anti-HIV antibodies. A CD4 fusion with the anti-HIV carbohydrate antibody 2G12 also results in a potent neutralizing reagent with more broadly neutralizing activity than 2G12 alone.
Collapse
|
461
|
A V3 loop-dependent gp120 element disrupted by CD4 binding stabilizes the human immunodeficiency virus envelope glycoprotein trimer. J Virol 2010; 84:3147-61. [PMID: 20089638 DOI: 10.1128/jvi.02587-09] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Human immunodeficiency virus (HIV-1) entry into cells is mediated by a trimeric complex consisting of noncovalently associated gp120 (exterior) and gp41 (transmembrane) envelope glycoproteins. The binding of gp120 to receptors on the target cell alters the gp120-gp41 relationship and activates the membrane-fusing capacity of gp41. Interaction of gp120 with the primary receptor, CD4, results in the exposure of the gp120 third variable (V3) loop, which contributes to binding the CCR5 or CXCR4 chemokine receptors. We show here that insertions in the V3 stem or polar substitutions in a conserved hydrophobic patch near the V3 tip result in decreased gp120-gp41 association (in the unliganded state) and decreased chemokine receptor binding (in the CD4-bound state). Subunit association and syncytium-forming ability of the envelope glycoproteins from primary HIV-1 isolates were disrupted more by V3 changes than those of laboratory-adapted HIV-1 envelope glycoproteins. Changes in the gp120 beta2, beta19, beta20, and beta21 strands, which evidence suggests are proximal to the V3 loop in unliganded gp120, also resulted in decreased gp120-gp41 association. Thus, a gp120 element composed of the V3 loop and adjacent beta strands contributes to quaternary interactions that stabilize the unliganded trimer. CD4 binding dismantles this element, altering the gp120-gp41 relationship and rendering the hydrophobic patch in the V3 tip available for chemokine receptor binding.
Collapse
|
462
|
Kelker HC, Itri VR, Valentine FT. A strategy for eliciting antibodies against cryptic, conserved, conformationally dependent epitopes of HIV envelope glycoprotein. PLoS One 2010; 5:e8555. [PMID: 20052405 PMCID: PMC2797330 DOI: 10.1371/journal.pone.0008555] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2009] [Accepted: 12/10/2009] [Indexed: 11/18/2022] Open
Abstract
Background Novel strategies are needed for the elicitation of broadly neutralizing antibodies to the HIV envelope glycoprotein, gp120. Experimental evidence suggests that combinations of antibodies that are broadly neutralizing in vitro may protect against challenge with HIV in nonhuman primates, and a small number of these antibodies have been selected by repertoire sampling of B cells and by the fractionation of antiserum from some patients with prolonged disease. Yet no additional strategies for identifying conserved epitopes, eliciting antibodies to these epitopes, and determining whether these epitopes are accessible to antibodies have been successful to date. The defining of additional conserved, accessible epitopes against which one can elicit antibodies will increase the probability that some may be the targets of broadly neutralizing antibodies. Methodology/Principal Findings We postulate that additional cryptic epitopes of gp120 are present, against which neutralizing antibodies might be elicited even though these antibodies are not elicited by gp120, and that many of these epitopes may be accessible to antibodies should they be formed. We demonstrate a strategy for eliciting antibodies in mice against selected cryptic, conformationally dependent conserved epitopes of gp120 by immunizing with multiple identical copies of covalently linked peptides (MCPs). This has been achieved with MCPs representing 3 different domains of gp120. We show that some cryptic epitopes on gp120 are accessible to the elicited antibodies, and some epitopes in the CD4 binding region are not accessible. The antibodies bind to gp120 with relatively high affinity, and bind to oligomeric gp120 on the surface of infected cells. Conclusions/Significance Immunization with MCPs comprised of selected peptides of HIV gp120 is able to elicit antibodies against conserved, conformationally dependent epitopes of gp120 that are not immunogenic when presented as gp120. Some of these cryptic epitopes are accessible to the elicited antibodies.
Collapse
Affiliation(s)
- Hanna C. Kelker
- Department of Medicine, New York University School of Medicine, New York, New York, United States of America
| | - Vincenza R. Itri
- Department of Medicine, New York University School of Medicine, New York, New York, United States of America
| | - Fred T. Valentine
- Department of Medicine, New York University School of Medicine, New York, New York, United States of America
- Department of Microbiology, New York University School of Medicine, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
463
|
Elder JH, Lin YC, Fink E, Grant CK. Feline immunodeficiency virus (FIV) as a model for study of lentivirus infections: parallels with HIV. Curr HIV Res 2010; 8:73-80. [PMID: 20210782 PMCID: PMC2853889 DOI: 10.2174/157016210790416389] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2009] [Accepted: 11/02/2009] [Indexed: 12/22/2022]
Abstract
FIV is a significant pathogen in the cat and is, in addition, the smallest available natural model for the study of lentivirus infections. Although divergent at the amino acid level, the cat lentivirus has an abundance of structural and pathophysiological commonalities with HIV and thus serves well as a model for development of intervention strategies relevant to infection in both cats and man. The following review highlights both the strengths and shortcomings of the FIV/cat model, particular as regards development of antiviral drugs.
Collapse
Affiliation(s)
- John H Elder
- Department of Immunology and Microbial Sciences, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | | | | | | |
Collapse
|
464
|
Structural Biology and the Design of Effective Vaccines for HIV-1 and Other Viruses. NATIONAL INSTITUTE OF ALLERGY AND INFECTIOUS DISEASES, NIH 2010. [PMCID: PMC7176257 DOI: 10.1007/978-1-60761-512-5_39] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Structural biology provides a wealth of information about the three-dimensional organization and chemical makeup of proteins. An understanding of atomic-level structure offers enormous potential to design rationally proteins that stimulate specific immune responses. Yet current vaccine development efforts makes little use of structural information. At the Vaccine Research Center, a major goal is to apply structural techniques to vaccine design for challenging pathogens, that include human immunodeficiency virus type 1 (HIV-1) and other enveloped viruses such as influenza, Ebola, and respiratory syncytial viruses. Our three-part strategy involves 1.) the definition of the functional viral spike at the atomic level 2.) achieving a structural understanding of how neutralizing antibodies recognize the spike, and 3.) rational development of proteins that can elicit a specific antibody response. Overall, our strategy aims to incorporate information about viral spike-antibody interactions, to assimilate immunogenic feedback, and to leverage recent advances in immunofocusing and computational biology.
Collapse
|
465
|
Cranage MP, Fraser CA, Stevens Z, Huting J, Chang M, Jeffs SA, Seaman MS, Cope A, Cole T, Shattock RJ. Repeated vaginal administration of trimeric HIV-1 clade C gp140 induces serum and mucosal antibody responses. Mucosal Immunol 2010; 3:57-68. [PMID: 19741600 PMCID: PMC3761690 DOI: 10.1038/mi.2009.110] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Vaccine-mediated prevention of primary infection with human immunodeficiency virus (HIV) may require the sustained production of antibody at mucosal portals of entry. Here, we describe a novel approach of repeated mucosal immunization by delivering an HIV-1 envelope glycoprotein (gp) in a gel formulated for intravaginal delivery. Rabbits were immunized over one to three 19-day cycles of intravaginal dosing with soluble recombinant trimeric HIV-1 clade C gp140 administered in Carbopol gel. The formulation was well tolerated. A single immunization cycle induced immunoglobulin G (IgG) antibody detected in the serum and female genital tract, and titers were boosted on further immunization. Vaccine-induced serum antibodies neutralized the infectivity of a pseudovirus carrying a heterologous clade C envelope. Our data prove the concept that repeated exposure of the female genital tract to HIV envelope can induce mucosally detectable antibody.
Collapse
Affiliation(s)
- M P Cranage
- Centre for Infection, Division of Cellular and Molecular Medicine, St George's University of London, London, UK.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
466
|
Structure of HIV-1 gp120 with gp41-interactive region reveals layered envelope architecture and basis of conformational mobility. Proc Natl Acad Sci U S A 2009; 107:1166-71. [PMID: 20080564 DOI: 10.1073/pnas.0911004107] [Citation(s) in RCA: 271] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The viral spike of HIV-1 is composed of three gp120 envelope glycoproteins attached noncovalently to three gp41 transmembrane molecules. Viral entry is initiated by binding to the CD4 receptor on the cell surface, which induces large conformational changes in gp120. These changes not only provide a model for receptor-triggered entry, but affect spike sensitivity to drug- and antibody-mediated neutralization. Although some of the details of the CD4-induced conformational change have been visualized by crystal structures and cryoelectron tomograms, the critical gp41-interactive region of gp120 was missing from previous atomic-level characterizations. Here we determine the crystal structure of an HIV-1 gp120 core with intact gp41-interactive region in its CD4-bound state, compare this structure to unliganded and antibody-bound forms to identify structurally invariant and plastic components, and use ligand-oriented cryoelectron tomograms to define component mobility in the viral spike context. Newly defined gp120 elements proximal to the gp41 interface complete a 7-stranded beta-sandwich, which appeared invariant in conformation. Loop excursions emanating from the sandwich form three topologically separate--and structurally plastic--layers, topped off by the highly glycosylated gp120 outer domain. Crystal structures, cryoelectron tomograms, and interlayer chemistry were consistent with a mechanism in which the layers act as a shape-changing spacer, facilitating movement between outer domain and gp41-associated beta-sandwich and providing for conformational diversity used in immune evasion. A "layered" gp120 architecture thus allows movement among alternative glycoprotein conformations required for virus entry and immune evasion, whereas a beta-sandwich clamp maintains gp120-gp41 interaction and regulates gp41 transitions.
Collapse
|
467
|
Hatada M, Yoshimura K, Harada S, Kawanami Y, Shibata J, Matsushita S. Human immunodeficiency virus type 1 evasion of a neutralizing anti-V3 antibody involves acquisition of a potential glycosylation site in V2. J Gen Virol 2009; 91:1335-45. [PMID: 20032207 DOI: 10.1099/vir.0.017426-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
It has been reported that the addition of a potential N-linked glycosylation site (PNGS) to the gp120 human immunodeficiency virus type 1 (HIV-1) envelope glycoprotein provides protection against neutralizing antibodies (NAbs) by acting as a 'glycan shield'. In this study, we induced insertion of a PNGS into the V2 region of HIV-1(BaL) with the KD-247 anti-V3 neutralizing monoclonal antibody. In the presence of KD-247 (200 microg ml(-1)) at passage five, viruses with 3 aa mutations in the C2 (T240S and I283T) and V3 (T319A) regions expanded from pre-existing variants. After six passages with KD-247 (>300 microg ml(-1)), a PNGS emerged in the V2 region in addition to C2 (T240S) and V3 mutations (R315K and F317L). A variant with a PNGS insertion in V2, but no V3 mutations was sensitive to KD-247, whereas a clone with a V2 PNGS insertion and mutations in V3 demonstrated a high level of resistance to KD-247. Replication kinetic analysis revealed that the F317L mutation in V3 played a compensatory role for fitness-loss caused by the PNGS insertion in V2. The evading HIV-1 variant did not revert back to the wild-type virus after 14 passages without KD-247. These findings demonstrate that the virus with fitness-loss mutations can replicate equally as well as the wild-type virus to acquire some key mutations in the V3 stem and the C2 region, and the compensated variants containing PNGS do not revert back to the ancestral virus even in the absence of NAb.
Collapse
Affiliation(s)
- Makiko Hatada
- Division of Clinical Retrovirology and Infectious Diseases, Center for AIDS Research, Kumamoto University, Kumamoto 860-0811, Japan
| | | | | | | | | | | |
Collapse
|
468
|
Chen L, Kwon YD, Zhou T, Wu X, O'Dell S, Cavacini L, Hessell AJ, Pancera M, Tang M, Xu L, Yang ZY, Zhang MY, Arthos J, Burton DR, Dimitrov DS, Nabel GJ, Posner MR, Sodroski J, Wyatt R, Mascola JR, Kwong PD. Structural basis of immune evasion at the site of CD4 attachment on HIV-1 gp120. Science 2009; 326:1123-7. [PMID: 19965434 DOI: 10.1126/science.1175868] [Citation(s) in RCA: 254] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The site on HIV-1 gp120 that binds to the CD4 receptor is vulnerable to antibodies. However, most antibodies that interact with this site cannot neutralize HIV-1. To understand the basis of this resistance, we determined co-crystal structures for two poorly neutralizing, CD4-binding site (CD4BS) antibodies, F105 and b13, in complexes with gp120. Both antibodies exhibited approach angles to gp120 similar to those of CD4 and a rare, broadly neutralizing CD4BS antibody, b12. Slight differences in recognition, however, resulted in substantial differences in F105- and b13-bound conformations relative to b12-bound gp120. Modeling and binding experiments revealed these conformations to be poorly compatible with the viral spike. This incompatibility, the consequence of slight differences in CD4BS recognition, renders HIV-1 resistant to all but the most accurately targeted antibodies.
Collapse
Affiliation(s)
- Lei Chen
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
469
|
Caputo A, Gavioli R, Bellino S, Longo O, Tripiciano A, Francavilla V, Sgadari C, Paniccia G, Titti F, Cafaro A, Ferrantelli F, Monini P, Ensoli F, Ensoli B. HIV-1 Tat-based vaccines: an overview and perspectives in the field of HIV/AIDS vaccine development. Int Rev Immunol 2009; 28:285-334. [PMID: 19811313 DOI: 10.1080/08830180903013026] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
The HIV epidemic continues to represent one of the major problems worldwide, particularly in the Asia and Sub-Saharan regions of the world, with social and economical devastating effects. Although antiretroviral drugs have had a dramatically beneficial impact on HIV-infected individuals that have access to treatment, it has had a negligible impact on the global epidemic. Hence, the inexorable spreading of the HIV pandemic and the increasing deaths from AIDS, especially in developing countries, underscore the urgency for an effective vaccine against HIV/AIDS. However, the generation of such a vaccine has turned out to be extremely challenging. Here we provide an overview on the rationale for the use of non-structural HIV proteins, such as the Tat protein, alone or in combination with other HIV early and late structural HIV antigens, as novel, promising preventative and therapeutic HIV/AIDS vaccine strategies.
Collapse
Affiliation(s)
- Antonella Caputo
- Department of Histology, Microbiology and Medical Biotechnology, University of Padova, Padova, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
470
|
Abdel-Motal UM, Wang S, Awad A, Lu S, Wigglesworth K, Galili U. Increased immunogenicity of HIV-1 p24 and gp120 following immunization with gp120/p24 fusion protein vaccine expressing alpha-gal epitopes. Vaccine 2009; 28:1758-65. [PMID: 20034607 DOI: 10.1016/j.vaccine.2009.12.015] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2009] [Revised: 11/23/2009] [Accepted: 12/09/2009] [Indexed: 01/18/2023]
Abstract
Developing an effective HIV-1 vaccine will require strategies to enhance antigen presentation to the immune system. In a previous study we demonstrated a marked increase in immunogenicity of the highly glycosylated HIV-1 gp120 protein following enzymatic addition of alpha-gal epitopes to the carbohydrate chains. In the present study we determined whether gp120(alphagal) can also serve as an effective platform for targeting other HIV-1 proteins to APC and thus increase immunogenicity of both proteins. For this purpose we produced a recombinant fusion protein between gp120 and the HIV-1 matrix p24 protein (gp120/p24). Multiple alpha-gal epitopes were synthesized enzymatically on the gp120 portion of the fusion protein to generate a gp120(alphagal)/p24 vaccine. Immune responses to gp120(alphagal)/p24 compared to gp120/p24 vaccine lacking alpha-gal epitopes were evaluated in alpha1,3galactosyltransferase knockout (KO) mice. These mice lack alpha-gal epitopes and, therefore, are capable of producing the anti-Gal antibody. T cell responses to p24, as assessed by ELISPOT and by CD8+ T cells intracellular staining assays for IFNgamma, was on average 12- and 10-fold higher, respectively, in gp120(alphagal)/p24 immunized mice than in mice immunized with gp120/p24. In addition, cellular and humoral immune responses against gp120 were higher by 10-30-fold in mice immunized with gp120(alphagal)/p24 than in gp120/p24 immunized mice. Our data suggest that the alpha-gal epitopes on the gp120 portion of the fusion protein can significantly augment the immunogenicity of gp120, as well as that of the fused viral protein which lacks alpha-gal epitopes. This strategy of anti-Gal mediated targeting to APC may be used for production of effective HIV-1 vaccines comprised of various viral proteins fused to gp120.
Collapse
Affiliation(s)
- Ussama M Abdel-Motal
- Department of Surgery, University of Massachusetts Medical School, Worcester, MA 01655, USA.
| | | | | | | | | | | |
Collapse
|
471
|
Abstract
PURPOSE OF REVIEW The only unequivocal correlate of protection against primate immunodeficiency virus infection is the presence of neutralizing antibody at sufficient titre. This correlate has been determined experimentally using animal models, and the data are reproducible and robust. Recent advances have added further depth to this knowledge by moving us closer to understanding how antibodies neutralize HIV-1, and what effects they may have in vivo with regard to protection from infection and disease. RECENT FINDINGS This review will cover recent advances in our understanding of the structural basis of HIV-1 neutralization by antibody and how this understanding may relate to vaccine design, and incorporate this into the broader context of how antibodies may influence viral transmission, replication and disease. SUMMARY The sum of these findings provides a strong rationale for designing an HIV-1 vaccine on the principle of induction of neutralizing antibodies, although other effector functions of antibodies such as complement and antibody-mediated cellular immunity should also be borne in mind, as should CD4 and CD8 T cell responses.
Collapse
|
472
|
Abstract
The early immune response to HIV-1 infection is likely to be an important factor in determining the clinical course of disease. Recent data indicate that the HIV-1 quasispecies that arise following a mucosal infection are usually derived from a single transmitted virus. Moreover, the finding that the first effective immune responses drive the selection of virus escape mutations provides insight into the earliest immune responses against the transmitted virus and their contributions to the control of acute viraemia. Strong innate and adaptive immune responses occur subsequently but they are too late to eliminate the infection. In this Review, we discuss recent studies on the kinetics and quality of early immune responses to HIV-1 and their implications for developing a successful preventive HIV-1 vaccine.
Collapse
|
473
|
Tian Y, Zhang H. Glycoproteomics and clinical applications. Proteomics Clin Appl 2009; 4:124-32. [PMID: 21137038 DOI: 10.1002/prca.200900161] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2009] [Revised: 10/27/2009] [Accepted: 11/05/2009] [Indexed: 11/11/2022]
Abstract
Glycosylation is the most structurally complicated and diverse type of protein modifications. Protein glycosylation has long been recognized to play fundamental roles in many biological processes, as well as in disease genesis and progression. Glycoproteomics focuses on characterization of proteins modified by carbohydrates. Glycoproteomic studies normally include strategies to enrich glycoproteins containing particular carbohydrate structures from protein mixtures followed by quantitative proteomic analysis. These glycoproteomic studies determine which proteins are glycosylated, the glycosylation sites, the carbohydrate structures, as well as the abundance and function of the glycoproteins in different biological and pathological processes. Here we review the recent development in methods used in glycoproteomic analysis. These techniques are essential in elucidation of the relationships between protein glycosylation and disease states. We also review the clinical applications of different glycoproteomic methods.
Collapse
Affiliation(s)
- Yuan Tian
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | | |
Collapse
|
474
|
Ex vivo comparison of microbicide efficacies for preventing HIV-1 genomic integration in intraepithelial vaginal cells. Antimicrob Agents Chemother 2009; 54:763-72. [PMID: 19949052 DOI: 10.1128/aac.00891-09] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Vaginally applied microbicides hold promise as a strategy to prevent sexual HIV transmission. Several nonspecific microbicides, including the polyanion cellulose sulfate, have been evaluated in large-scale clinical trials but have failed to show significant efficacy. These findings have prompted a renewed search for preclinical testing systems that can predict negative outcomes of microbicide trials. Moreover, the pipeline of potential topical microbicides has been expanded to include antiretroviral agents, such as reverse transcriptase, fusion, and integrase inhibitors. Using a novel ex vivo model of vaginal HIV-1 infection, we compared the prophylactic potentials of two forms of the fusion inhibitor T-20, the CCR5 antagonist TAK-778, the integrase inhibitor 118-D-24, and cellulose sulfate (Ushercell). The T-20 peptide with free N- and C-terminal amino acids was the most efficacious compound, causing significantly greater inhibition of viral genomic integration in intraepithelial vaginal leukocytes, measured by an optimized real-time PCR assay, than the more water-soluble N-acetylated T-20 peptide (Fuzeon) (50% inhibitory concentration [IC50], 0.153 microM versus 51.2 microM [0.687 ng/ml versus 230 ng/ml]; P<0.0001). In contrast, no significant difference in IC50s was noted in peripheral blood cells (IC50, 13.58 microM versus 7.57 microM [61 ng/ml versus 34 ng/ml]; P=0.0614). Cellulose sulfate was the least effective of all the compounds tested (IC50, 1.8 microg/ml). These results highlight the merit of our model for screening the mucosal efficacies of novel microbicides and their formulations and potentially rank ordering candidates for clinical evaluation.
Collapse
|
475
|
Tiered categorization of a diverse panel of HIV-1 Env pseudoviruses for assessment of neutralizing antibodies. J Virol 2009; 84:1439-52. [PMID: 19939925 DOI: 10.1128/jvi.02108-09] [Citation(s) in RCA: 547] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The restricted neutralization breadth of vaccine-elicited antibodies is a major limitation of current human immunodeficiency virus-1 (HIV-1) candidate vaccines. In order to permit the efficient identification of vaccines with enhanced capacity for eliciting cross-reactive neutralizing antibodies (NAbs) and to assess the overall breadth and potency of vaccine-elicited NAb reactivity, we assembled a panel of 109 molecularly cloned HIV-1 Env pseudoviruses representing a broad range of genetic and geographic diversity. Viral isolates from all major circulating genetic subtypes were included, as were viruses derived shortly after transmission and during the early and chronic stages of infection. We assembled a panel of genetically diverse HIV-1-positive (HIV-1(+)) plasma pools to assess the neutralization sensitivities of the entire virus panel. When the viruses were rank ordered according to the average sensitivity to neutralization by the HIV-1(+) plasmas, a continuum of average sensitivity was observed. Clustering analysis of the patterns of sensitivity defined four subgroups of viruses: those having very high (tier 1A), above-average (tier 1B), moderate (tier 2), or low (tier 3) sensitivity to antibody-mediated neutralization. We also investigated potential associations between characteristics of the viral isolates (clade, stage of infection, and source of virus) and sensitivity to NAb. In particular, higher levels of NAb activity were observed when the virus and plasma pool were matched in clade. These data provide the first systematic assessment of the overall neutralization sensitivities of a genetically and geographically diverse panel of circulating HIV-1 strains. These reference viruses can facilitate the systematic characterization of NAb responses elicited by candidate vaccine immunogens.
Collapse
|
476
|
Role of HIV membrane in neutralization by two broadly neutralizing antibodies. Proc Natl Acad Sci U S A 2009; 106:20234-9. [PMID: 19906992 DOI: 10.1073/pnas.0908713106] [Citation(s) in RCA: 212] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Induction of effective antibody responses against HIV-1 infection remains an elusive goal for vaccine development. Progress may require in-depth understanding of the molecular mechanisms of neutralization by monoclonal antibodies. We have analyzed the molecular actions of two rare, broadly neutralizing, human monoclonal antibodies, 4E10 and 2F5, which target the transiently exposed epitopes in the membrane proximal external region (MPER) of HIV-1 gp41 envelope during viral entry. Both have long CDR H3 loops with a hydrophobic surface facing away from the peptide epitope. We find that the hydrophobic residues of 4E10 mediate a reversible attachment to the viral membrane and that they are essential for neutralization, but not for interaction with gp41. We propose that these antibodies associate with the viral membrane in a required first step and are thereby poised to capture the transient gp41 fusion intermediate. These results bear directly on strategies for rational design of HIV-1 envelope immunogens.
Collapse
|
477
|
Envelope vaccination shapes viral envelope evolution following simian immunodeficiency virus infection in rhesus monkeys. J Virol 2009; 84:953-63. [PMID: 19906933 DOI: 10.1128/jvi.01679-09] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The evolution of envelope mutations by replicating primate immunodeficiency viruses allows these viruses to escape from the immune pressure mediated by neutralizing antibodies. Vaccine-induced anti-envelope antibody responses may accelerate and/or alter the specificity of the antibodies, thus shaping the evolution of envelope mutations in the replicating virus. To explore this possibility, we studied the neutralizing antibody response and the envelope sequences in rhesus monkeys vaccinated with either gag-pol-nef immunogens or gag-pol-nef immunogens in combination with env and then infected with simian immunodeficiency virus (SIV). Using a pseudovirion neutralization assay, we demonstrate that envelope vaccination primed for an accelerated neutralizing antibody response following virus challenge. To monitor viral envelope evolution in these two cohorts of monkeys, full-length envelopes from plasma virus isolated at weeks 37 and 62 postchallenge were sequenced by single genome amplification to identify sites of envelope mutations. We show that env vaccination was associated with a change in the pattern of envelope mutations. Prevalent mutations in sequences from gag-pol-nef vaccinees included deletions in both variable regions 1 and 4 (V1 and V4), whereas deletions in the env vaccinees occurred only in V1. These data show that env vaccination altered the focus of the antibody-mediated selection pressure on the evolution of envelope following SIV challenge.
Collapse
|
478
|
Hioe CE, Visciano ML, Kumar R, Liu J, Mack EA, Simon RE, Levy DN, Tuen M. The use of immune complex vaccines to enhance antibody responses against neutralizing epitopes on HIV-1 envelope gp120. Vaccine 2009; 28:352-60. [PMID: 19879224 DOI: 10.1016/j.vaccine.2009.10.040] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2009] [Revised: 10/03/2009] [Accepted: 10/12/2009] [Indexed: 11/27/2022]
Abstract
The capacity of immune complexes to augment antibody (Ab) responses is well established. The enhancing effects of immune complexes have been attributed mainly to Fc-mediated adjuvant activity, while the ability of Abs to induce antigenic alterations of specific epitopes as a result of immune complex formation has been less well studied. Previously we have shown that the interaction of anti-CD4-binding site (CD4bs) Abs with HIV-1 gp120 induces conformation changes that lead to enhanced antigenicity and immunogenicity of neutralizing epitopes in the V3 loop. The present study shows that significant increases in the antigenicity of the V3 and C1 regions of gp120 were attained for several subtype B gp120s and a subtype C gp120 upon immune complex formation with the anti-CD4bs monoclonal Ab (mAb) 654-D. Such enhancement was observed with immune complexes made with other anti-CD4bs mAbs and anti-V2 mAbs, but not with anti-C2 mAbs, indicating this activity is determined by antigen specificity of the mAb that formed the immune complex. When immune complexes of gp120(LAI)/654-D and gp120(JRFL)/654-D were tested as immunogens in mice, serum Abs to gp120 and V3 were generated at significantly higher titers than those induced by the respective uncomplexed gp120s. Notably, the anti-V3 Ab responses had distinct fine specificities; gp120(JRFL)/654-D stimulated more cross-reactive anti-V3 Abs than gp120(LAI)/654-D. Neutralizing activities against viruses with heterologous envelope were also detected in sera of mice immunized with gp120(JRFL)/654-D, although the neutralization breadth was still limited. Overall this study shows the potential use of gp120/Ab complexes to augment the immunogenicity of HIV-1 envelope gp120, but further improvements are needed to elicit virus-neutralizing Ab responses with higher potency and breadth.
Collapse
Affiliation(s)
- Catarina E Hioe
- Department of Pathology, New York University School of Medicine, New York, NY 10010, United States.
| | | | | | | | | | | | | | | |
Collapse
|
479
|
Neutralizing inter-clade cross-reactivity of HIV-1 V1/V2-specific secretory immunoglobulin A in Colombian and French cohorts. AIDS 2009; 23:2219-22. [PMID: 19830891 DOI: 10.1097/qad.0b013e328329d134] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Neutralizing activity of secretory immunoglobulin A (S-IgA) directed against the V1/V2 domain of HIV-1 was studied in parotid saliva of HIV-1- infected patients in Colombian and French cohorts. Purified V1/V2-specific S-IgA antibodies were found to neutralize clades A, B and C primary isolates in five out 76 and 82 patients from each cohort, respectively. These results suggest that neutralizing S-IgA antibodies targeting the V1/V2 domain may provide protection against HIV-1 infection in vivo and may be beneficial in mucosal vaccines.
Collapse
|
480
|
Latinovic O, Kuruppu J, Davis C, Le N, Heredia A. Pharmacotherapy of HIV-1 Infection: Focus on CCR5 Antagonist Maraviroc. ACTA ACUST UNITED AC 2009; 1:1497-1510. [PMID: 19920876 DOI: 10.4137/cmt.s2365] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Sustained inhibition of HIV-1, the goal of antiretroviral therapy, is often impeded by the emergence of viral drug resistance. For patients infected with HIV-1 resistant to conventional drugs from the viral reverse transcriptase and protease inhibitor classes, the recently approved entry and integration inhibitors effectively suppress HIV-1 and offer additional therapeutic options. Entry inhibitors are particularly attractive because, unlike conventional antiretrovirals, they target HIV-1 extracellularly, thereby sparing cells from both viral- and drug-induced toxicities. The fusion inhibitor enfuvirtide and the CCR5 antagonist maraviroc are the first entry inhibitors licensed for patients with drug-resistant HIV-1, with maraviroc restricted to those infected with CCR5-tropic HIV-1 (R5 HIV-1) only. Vicriviroc (another CCR5 antagonist) is in Phase III clinical trials, whereas the CCR5 antibodies PRO 140 and HGS 004 are in early stages of clinical development. Potent antiviral synergy between maraviroc and CCR5 antibodies, coupled with distinct patterns of resistance, suggest their combinations might be particularly effective in patients. In addition, given that oral administration of maraviroc achieves high drug levels in cervicovaginal fluid, combinations of maraviroc and other CCR5 inhibitors could be effective in preventing HIV-1 transmission. Moreover, since CCR5 antagonists prevent rejection of transplanted organs, maraviroc could both suppress HIV-1 and prolong organ survival for the growing number of HIV-1 patients with kidney or liver failure necessitating organ transplantation. Thus, maraviroc offers an important treatment option for patients with drug-resistant R5 HIV-1, who presently account for >50% of drug-resistance cases.
Collapse
Affiliation(s)
- Olga Latinovic
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | | | | | | | | |
Collapse
|
481
|
Da LT, Quan JM, Wu YD. Understanding of the Bridging Sheet Formation of HIV-1 Glycoprotein gp120. J Phys Chem B 2009; 113:14536-43. [DOI: 10.1021/jp9081239] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Lin-Tai Da
- Laboratory of Chemical Genomics, Shenzhen Graduate School of Peking University, Shenzhen 518055, China, and Department of Chemistry, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Jun-Min Quan
- Laboratory of Chemical Genomics, Shenzhen Graduate School of Peking University, Shenzhen 518055, China, and Department of Chemistry, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Yun-Dong Wu
- Laboratory of Chemical Genomics, Shenzhen Graduate School of Peking University, Shenzhen 518055, China, and Department of Chemistry, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| |
Collapse
|
482
|
Verkoczy L, Moody MA, Holl TM, Bouton-Verville H, Scearce RM, Hutchinson J, Alam SM, Kelsoe G, Haynes BF. Functional, non-clonal IgMa-restricted B cell receptor interactions with the HIV-1 envelope gp41 membrane proximal external region. PLoS One 2009; 4:e7215. [PMID: 19806186 PMCID: PMC2751816 DOI: 10.1371/journal.pone.0007215] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2009] [Accepted: 08/24/2009] [Indexed: 11/24/2022] Open
Abstract
The membrane proximal external region (MPER) of HIV-1 gp41 has several features that make it an attractive antibody-based vaccine target, but eliciting an effective gp41 MPER-specific protective antibody response remains elusive. One fundamental issue is whether the failure to make gp41 MPER-specific broadly neutralizing antibodies like 2F5 and 4E10 is due to structural constraints with the gp41 MPER, or alternatively, if gp41 MPER epitope-specific B cells are lost to immunological tolerance. An equally important question is how B cells interact with, and respond to, the gp41 MPER epitope, including whether they engage this epitope in a non-canonical manner i.e., by non-paratopic recognition via B cell receptors (BCR). To begin understanding how B cells engage the gp41 MPER, we characterized B cell-gp41 MPER interactions in BALB/c and C57BL/6 mice. Surprisingly, we found that a significant (∼7%) fraction of splenic B cells from BALB/c, but not C57BL/6 mice, bound the gp41 MPER via their BCRs. This strain-specific binding was concentrated in IgMhi subsets, including marginal zone and peritoneal B1 B cells, and correlated with enriched fractions (∼15%) of gp41 MPER-specific IgM secreted by in vitro-activated splenic B cells. Analysis of Igha (BALB/c) and Ighb (C57BL/6) congenic mice demonstrated that gp41 MPER binding was controlled by determinants of the Igha locus. Mapping of MPER gp41 interactions with IgMa identified MPER residues distinct from those to which mAb 2F5 binds and demonstrated the requirement of Fc CH regions. Importantly, gp41 MPER ligation produced detectable BCR-proximal signaling events, suggesting that interactions between gp41 MPER and IgMa determinants may elicit partial B cell activation. These data suggest that low avidity, non-paratopic interactions between the gp41 MPER and membrane Ig on naïve B cells may interfere with or divert bnAb responses.
Collapse
Affiliation(s)
- Laurent Verkoczy
- Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, United States of America.
| | | | | | | | | | | | | | | | | |
Collapse
|
483
|
Rong R, Li B, Lynch RM, Haaland RE, Murphy MK, Mulenga J, Allen SA, Pinter A, Shaw GM, Hunter E, Robinson JE, Gnanakaran S, Derdeyn CA. Escape from autologous neutralizing antibodies in acute/early subtype C HIV-1 infection requires multiple pathways. PLoS Pathog 2009; 5:e1000594. [PMID: 19763269 PMCID: PMC2741593 DOI: 10.1371/journal.ppat.1000594] [Citation(s) in RCA: 148] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2009] [Accepted: 08/27/2009] [Indexed: 12/15/2022] Open
Abstract
One aim for an HIV vaccine is to elicit neutralizing antibodies (Nab) that can limit replication of genetically diverse viruses and prevent establishment of a new infection. Thus, identifying the strengths and weaknesses of Nab during the early stages of natural infection could prove useful in achieving this goal. Here we demonstrate that viral escape readily occurred despite the development of high titer autologous Nab in two subjects with acute/early subtype C infection. To provide a detailed portrayal of the escape pathways, Nab resistant variants identified at multiple time points were used to create a series of envelope (Env) glycoprotein chimeras and mutants within the background of a corresponding newly transmitted Env. In one subject, Nab escape was driven predominantly by changes in the region of gp120 that extends from the beginning of the V3 domain to the end of the V5 domain (V3V5). However, Nab escape pathways in this subject oscillated and at times required cooperation between V1V2 and the gp41 ectodomain. In the second subject, escape was driven by changes in V1V2. This V1V2-dependent escape pathway was retained over time, and its utility was reflected in the virus's ability to escape from two distinct monoclonal antibodies (Mabs) derived from this same patient via introduction of a single potential N-linked glycosylation site in V2. Spatial representation of the sequence changes in gp120 suggested that selective pressure acted upon the same regions of Env in these two subjects, even though the Env domains that drove escape were different. Together the findings argue that a single mutational pathway is not sufficient to confer escape in early subtype C HIV-1 infection, and support a model in which multiple strategies, including potential glycan shifts, direct alteration of an epitope sequence, and cooperative Env domain conformational masking, are used to evade neutralization.
Collapse
Affiliation(s)
- Rong Rong
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, United States of America
- Emory Vaccine Center at Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Bing Li
- Emory Vaccine Center at Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Rebecca M. Lynch
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, United States of America
- Emory Vaccine Center at Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Richard E. Haaland
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, United States of America
- Emory Vaccine Center at Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Megan K. Murphy
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, United States of America
- Emory Vaccine Center at Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Joseph Mulenga
- Zambia Emory HIV Research Project, ZEHRP, Lusaka, Zambia
- Zambia Blood Transfusion Service, Lusaka, Zambia
| | - Susan A. Allen
- Zambia Emory HIV Research Project, ZEHRP, Lusaka, Zambia
- Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, Georgia, United States of America
| | - Abraham Pinter
- Public Health Research Institute, Newark, New Jersey, United States of America
- New Jersey School of Medicine, University of Medicine and Dentistry, Newark, New Jersey, United States of America
| | - George M. Shaw
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Eric Hunter
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, United States of America
- Emory Vaccine Center at Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - James E. Robinson
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - S. Gnanakaran
- Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
| | - Cynthia A. Derdeyn
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, United States of America
- Emory Vaccine Center at Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
484
|
Adaptive immunity to hepatitis C virus. Viruses 2009; 1:276-97. [PMID: 21994550 PMCID: PMC3185498 DOI: 10.3390/v1020276] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2009] [Revised: 08/14/2009] [Accepted: 08/25/2009] [Indexed: 12/23/2022] Open
Abstract
The precise role of adaptive immune responses in the clinical outcome of HCV infection is still only partially defined. Recent studies suggest that viral-host cell interactions during the acute phase of infection are essential for viral clearance or progression into chronic HCV infection. This review focuses on different aspects of the adaptive immune responses as determinants of the different outcomes of HCV infection, clearance or persistent infection, and outlines current concepts of HCV evasion strategies. Unravelling these important mechanisms of virus-host interaction will contribute to the development of novel strategies to prevent and control HCV infection.
Collapse
|
485
|
Gray ES, Taylor N, Wycuff D, Moore PL, Tomaras GD, Wibmer CK, Puren A, DeCamp A, Gilbert PB, Wood B, Montefiori DC, Binley JM, Shaw GM, Haynes BF, Mascola JR, Morris L. Antibody specificities associated with neutralization breadth in plasma from human immunodeficiency virus type 1 subtype C-infected blood donors. J Virol 2009; 83:8925-37. [PMID: 19553335 PMCID: PMC2738176 DOI: 10.1128/jvi.00758-09] [Citation(s) in RCA: 151] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2009] [Accepted: 06/13/2009] [Indexed: 02/03/2023] Open
Abstract
Defining the specificities of the anti-human immunodeficiency virus type 1 (HIV-1) envelope antibodies able to mediate broad heterologous neutralization will assist in identifying targets for an HIV-1 vaccine. We screened 70 plasmas from chronically HIV-1-infected individuals for neutralization breadth. Of these, 16 (23%) were found to neutralize 80% or more of the viruses tested. Anti-CD4 binding site (CD4bs) antibodies were found in almost all plasmas independent of their neutralization breadth, but they mainly mediated neutralization of the laboratory strain HxB2 with little effect on the primary virus, Du151. Adsorption with Du151 monomeric gp120 reduced neutralizing activity to some extent in most plasma samples when tested against the matched virus, although these antibodies did not always confer cross-neutralization. For one plasma, this activity was mapped to a site overlapping the CD4-induced (CD4i) epitope and CD4bs. Anti-membrane-proximal external region (MPER) (r = 0.69; P < 0.001) and anti-CD4i (r = 0.49; P < 0.001) antibody titers were found to be correlated with the neutralization breadth. These anti-MPER antibodies were not 4E10- or 2F5-like but spanned the 4E10 epitope. Furthermore, we found that anti-cardiolipin antibodies were correlated with the neutralization breadth (r = 0.67; P < 0.001) and anti-MPER antibodies (r = 0.6; P < 0.001). Our study suggests that more than one epitope on the envelope glycoprotein is involved in the cross-reactive neutralization elicited during natural HIV-1 infection, many of which are yet to be determined, and that polyreactive antibodies are possibly involved in this phenomenon.
Collapse
Affiliation(s)
- Elin S Gray
- AIDS Virus Research Unit, National Institute for Communicable Diseases, Johannesburg, Private Bag X4, Sandringham 2131, Johannesburg, South Africa
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
486
|
Go EP, Chang Q, Liao HX, Sutherland LL, Alam SM, Haynes BF, Desaire H. Glycosylation site-specific analysis of clade C HIV-1 envelope proteins. J Proteome Res 2009; 8:4231-42. [PMID: 19610667 PMCID: PMC2756219 DOI: 10.1021/pr9002728] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The extensive glycosylation of HIV-1 envelope proteins (Envs), gp120/gp41, is known to play an important role in evasion of host immune response by masking key neutralization epitopes and presenting the Env glycosylation as "self" to the host immune system. The Env glycosylation is mostly conserved but continues to evolve to modulate viral infectivity. Thus, profiling Env glycosylation and distinguishing interclade and intraclade glycosylation variations are necessary components in unraveling the effects of glycosylation on Env's immunogenicity. Here, we describe a mass spectrometry-based approach to characterize the glycosylation profiles of two rVV-expressed clade C Envs by identifying the glycan motifs on each glycosylation site and determining the degree of glycosylation site occupancy. One Env is a wild-type Env, while the other is a synthetic "consensus" Env (C.CON). The observed differences in the glycosylation profiles between the two clade C Envs show that C.CON has more unutilized sites and high levels of high mannose glycans; these features mimic the glycosylation profile of a Group M consensus immunogen, CON-S. Our results also reveal a clade-specific glycosylation pattern. Discerning interclade and intraclade glycosylation variations could provide valuable information in understanding the molecular differences among the different HIV-1 clades and in designing new Env-based immunogens.
Collapse
Affiliation(s)
- Eden P. Go
- Department of Chemistry, University of Kansas, Lawrence, KS, 66045
| | - Qing Chang
- Department of Chemistry, University of Kansas, Lawrence, KS, 66045
| | - Hua-Xin Liao
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, 22270
| | - Laura L. Sutherland
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, 22270
| | - S. Munir Alam
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, 22270
| | - Barton F. Haynes
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, 22270
| | - Heather Desaire
- Department of Chemistry, University of Kansas, Lawrence, KS, 66045
| |
Collapse
|
487
|
Zhao C, Crews CJ, Derdeyn CA, Blackwell JL. Lac-regulated system for generating adenovirus 5 vaccine vectors expressing cytolytic human immunodeficiency virus 1 genes. J Virol Methods 2009; 160:101-10. [PMID: 19409930 PMCID: PMC2704014 DOI: 10.1016/j.jviromet.2009.04.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2009] [Accepted: 04/23/2009] [Indexed: 01/14/2023]
Abstract
Adenovirus (Ad) vectors have been developed as human immunodeficiency-1 (HIV-1) vaccine vectors because they consistently induce immune responses in preclinical animal models and human trials. Strong promoters and codon-optimization are often used to enhance vaccine-induced HIV-1 gene expression and immunogenicity. However, if the transgene is inherently cytotoxic in the cell line used to produce the vector, and is expressed at high levels, it is difficult to rescue a stable Ad HIV-1 vaccine vector. Therefore we hypothesized that generation of Ad vaccine vectors expressing cytotoxic genes, such as HIV-1 env, would be more efficient if expression of the transgene was down-regulated during Ad rescue. To test this hypothesis, a Lac repressor-operator system was applied to regulate expression of reporter luciferase and HIV-1 env transgenes during Ad rescue. The results demonstrate that during Ad rescue, constitutive expression of the Lac repressor in 293 cells reduced transgene expression levels to approximately 5% of that observed in the absence of regulation. Furthermore, Lac-regulation translated into more efficient Ad rescue compared to traditional 293 cells. Importantly, Ad vectors rescued with this system showed high levels of transgene expression when transduced into cells that lack the Lac repressor protein. The Lac-regulated system also facilitated the rescue of modified Ad vectors that have non-native receptor tropism. These tropism-modified Ad vectors infect a broader range of cell types than the unmodified Ad, which could increase their effectiveness as a vaccine vector. Overall, the Lac-regulated system described here (i) is backwards compatible with Ad vector methods that employ bacterial-mediated homologous recombination, (ii) is adaptable for the engineering of tropism-modified Ad vectors, and (iii) does not require co-expression of regulatory genes from the vector or the addition of exogenous chemicals to induce or repress transgene expression. This system therefore could facilitate the development of Ad-based vaccine candidates that otherwise would not be feasible to generate.
Collapse
Affiliation(s)
- Chunxia Zhao
- Emory Vaccine Center, Emory University, Atlanta, GA 30329, United States
| | | | | | | |
Collapse
|
488
|
Mester B, Manor R, Mor A, Arshava B, Rosen O, Ding FX, Naider F, Anglister J. HIV-1 Peptide Vaccine Candidates: Selecting Constrained V3 Peptides with Highest Affinity to Antibody 447-52D. Biochemistry 2009; 48:7867-77. [DOI: 10.1021/bi900146g] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Brenda Mester
- Department of Structural Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Revital Manor
- Department of Structural Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Amit Mor
- Department of Structural Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Boris Arshava
- Department of Chemistry, College of Staten Island of the City University of New York, Staten Island, New York 10314
| | - Osnat Rosen
- Department of Structural Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Fa-Xiang Ding
- Department of Chemistry, College of Staten Island of the City University of New York, Staten Island, New York 10314
| | - Fred Naider
- Department of Chemistry, College of Staten Island of the City University of New York, Staten Island, New York 10314
| | - Jacob Anglister
- Department of Structural Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
489
|
Darwinian selection for sites of Asn-linked glycosylation in phylogenetically disparate eukaryotes and viruses. Proc Natl Acad Sci U S A 2009; 106:13421-6. [PMID: 19666543 DOI: 10.1073/pnas.0905818106] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Numerous protists and rare fungi have truncated Asn-linked glycan precursors and lack N-glycan-dependent quality control (QC) systems for glycoprotein folding in the endoplasmic reticulum. Here, we show that the abundance of sequons (NXT or NXS), which are sites for N-glycosylation of secreted and membrane proteins, varies by more than a factor of 4 among phylogenetically diverse eukaryotes, based on a few variables. There is positive correlation between the density of sequons and the AT content of coding regions, although no causality can be inferred. In contrast, there appears to be Darwinian selection for sequons containing Thr, but not Ser, in eukaryotes that have N-glycan-dependent QC systems. Selection for sequons with Thr, which nearly doubles the sequon density in human secreted and membrane proteins, occurs by an increased conditional probability that Asn and Thr are present in sequons rather than elsewhere. Increasing sequon densities of the hemagglutinin (HA) of influenza viruses A/H3N2 and A/H1N1 during the past few decades of human infection also result from an increased conditional probability that Asn, Thr, and Ser are present in sequons rather than elsewhere. In contrast, there is no selection on sequons by this mechanism in HA of A/H5N1 or 2009 A/H1N1 (Swine flu). Very strong selection for sequons with both Thr and Ser in glycoprotein of M(r) 120,000 (gp120) of HIV and related retroviruses results from this same mechanism, as well as amino acid composition bias and increases in AT content. We conclude that there is Darwinian selection for sequons in phylogenetically disparate eukaryotes and viruses.
Collapse
|
490
|
Wan Y, Liu L, Wu L, Huang X, Ma L, Xu J. Deglycosylation or partial removal of HIV-1 CN54 gp140 V1/V2 domain enhances env-specific T cells. AIDS Res Hum Retroviruses 2009; 25:607-17. [PMID: 19500018 DOI: 10.1089/aid.2008.0289] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
It remains a great challenge to develop an effective HIV vaccine against the most prevalent HIV-1 clade, B'/C recombinant, in China. Our objective was to test the influence of a new modification of the V1/V2 loops of HIV-1(CN54) gp140 on the immunogenicity of Env. HIV-1(CN54) gp140 was deglycosylated by replacing all six N residues in V1/V2 loops with six Q residues (gp140dG) or partially deleted on V1/V2 loops (gp140dV). gp140, gp140dG, and gp140dV were transferred into plasmid vector and recombinant Tiantan vaccinia (rTTV) vector to generate three DNA vaccines and three rTTV vaccines for vaccination of female BALB/c mice in a prime-boost regimen. An Elispot assay was used to read out the T cell immunity and ELISA and a poly-l-leucine (PLL) ELISA was employed to assess humoral immune responses. Surprisingly, gp140dV (1570 +/- 1569 SFCs/10(6) splenocytes) and gp140dG (731 +/- 471 SFCs/10(6) splenocytes) could elicit significantly higher Env-specific T cells than gp140 (224 +/- 140 SFCs/10(6) splenocytes). Three T cell epitopes were newly identified in BALB/c mice at the N terminus of C1, C terminus of C4, and N terminus of HR, respectively. Env-specific binding antibodies and linear antibodies elicited by gp140 tended to be higher than that stimulated by gp140dG and gp140dV but did not reach statistical difference. Our data demonstrated that the deglycosylation and partial deletion of V1/V2 loops of B'/C recombinant gp140 could lead to improvement of specific T cell immune responses.
Collapse
Affiliation(s)
- Yanmin Wan
- State Key Laboratory for Infectious Disease Prevention and Control, National Center for AIDS/STD Control and Prevention (NCAIDS), Chinese Center for Disease Control and Prevention (CDC), Beijing, China
- Scientific Research Center, Shanghai Public Health Clinical Center, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Lianxing Liu
- State Key Laboratory for Infectious Disease Prevention and Control, National Center for AIDS/STD Control and Prevention (NCAIDS), Chinese Center for Disease Control and Prevention (CDC), Beijing, China
| | - Lan Wu
- State Key Laboratory for Infectious Disease Prevention and Control, National Center for AIDS/STD Control and Prevention (NCAIDS), Chinese Center for Disease Control and Prevention (CDC), Beijing, China
- Present address: Vaccine Research Center, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, Maryland
| | - Xianggang Huang
- State Key Laboratory for Infectious Disease Prevention and Control, National Center for AIDS/STD Control and Prevention (NCAIDS), Chinese Center for Disease Control and Prevention (CDC), Beijing, China
| | - Liying Ma
- State Key Laboratory for Infectious Disease Prevention and Control, National Center for AIDS/STD Control and Prevention (NCAIDS), Chinese Center for Disease Control and Prevention (CDC), Beijing, China
| | - Jianqing Xu
- State Key Laboratory for Infectious Disease Prevention and Control, National Center for AIDS/STD Control and Prevention (NCAIDS), Chinese Center for Disease Control and Prevention (CDC), Beijing, China
- Scientific Research Center, Shanghai Public Health Clinical Center, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
491
|
Zeisel MB, Baumert TF. HCV entry and neutralizing antibodies: lessons from viral variants. Future Microbiol 2009; 4:511-7. [PMID: 19492962 PMCID: PMC2898794 DOI: 10.2217/fmb.09.34] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Evaluation of: Grove J, Nielsen S, Zhong J et al.: Identification of a residue in hepatitis C virus E2 glycoprotein that determines scavenger receptor BI and CD81 receptor dependency and sensitivity to neutralizing antibodies. J. Virol. 82 (24), 12020-12029 (2008). Recent data suggest that a strong, early, broad neutralizing antibody response may contribute to the control of HCV in the acute phase of infection. However, the majority of individuals fail to clear HCV during the first months following infection and develop chronic infection despite the presence of anti-HCV antibodies. A prerequisite of the understanding behind the mechanisms of viral escape from antibody-mediated neutralization is the identification of various host-entry factors mediating the first steps of viral infection - binding and entry of HCV is believed to be a multistep process involving HCV envelope glycoproteins E1 and E2 as well as several host-cell surface molecules such as CD81, scavenger receptor class B type I, members of the claudin family and occludin. In this article, Grove et al. describe a single mutation in the HCV envelope glycoprotein E2 that alters glycoprotein structure thereby modulating viral interaction with scavenger receptor class B type I and CD81, and increasing sensitivity to neutralizing antibodies. The results of this study highlight the importance of the characterization of the interplay between HCV particles and host-cell factors for the understanding of virus neutralization by host-immune responses and pathogenesis of HCV infection.
Collapse
|
492
|
Abstract
New modes of humoral recognition have been identified by studies of antibodies that neutralize human immunodeficiency virus type 1 and influenza A viruses. Understanding how such modes of antibody-antigen recognition can occur in the context of sophisticated mechanisms of humoral evasion has implications for the development of effective vaccines. Here we describe eight modes of antibody recognition first observed with human immunodeficiency virus type 1. Similarities to four of these modes have been identified with antibodies to a conserved 'stem' epitope on influenza A viruses. We outline how each of these different modes of antibody recognition is particularly suited to overcoming a specific viral evasion tactic and assess potential routes of re-elicitation in vaccine settings.
Collapse
Affiliation(s)
- Peter D Kwong
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA.
| | | |
Collapse
|
493
|
Structure-based stabilization of HIV-1 gp120 enhances humoral immune responses to the induced co-receptor binding site. PLoS Pathog 2009; 5:e1000445. [PMID: 19478876 PMCID: PMC2680979 DOI: 10.1371/journal.ppat.1000445] [Citation(s) in RCA: 107] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2009] [Accepted: 04/25/2009] [Indexed: 11/23/2022] Open
Abstract
The human immunodeficiency virus type 1 (HIV-1) exterior envelope glycoprotein, gp120, possesses conserved binding sites for interaction with the primary virus receptor, CD4, and also for the co-receptor, generally CCR5. Although gp120 is a major target for virus-specific neutralizing antibodies, the gp120 variable elements and its malleable nature contribute to evasion of effective host-neutralizing antibodies. To understand the conformational character and immunogenicity of the gp120 receptor binding sites as potential vaccine targets, we introduced structure-based modifications to stabilize gp120 core proteins (deleted of the gp120 major variable regions) into the conformation recognized by both receptors. Thermodynamic analysis of the re-engineered core with selected ligands revealed significant stabilization of the receptor-binding regions. Stabilization of the co-receptor-binding region was associated with a marked increase in on-rate of ligand binding to this site as determined by surface plasmon resonance. Rabbit immunization studies showed that the conformational stabilization of core proteins, along with increased ligand affinity, was associated with strikingly enhanced humoral immune responses against the co-receptor-binding site. These results demonstrate that structure-based approaches can be exploited to stabilize a conformational site in a large functional protein to enhance immunogenic responses specific for that region. Vaccination is an effective means to control worldwide human diseases caused by viruses and other pathogens. Most viral vaccines work by inducing the immune system to generate neutralizing antibodies. The human immunodeficiency virus (HIV) continues to cause huge tolls in terms of human death and disease. The generation of neutralizing antibodies against HIV remains a key but elusive goal for the development of an effective vaccine. Here, we describe a novel approach that uses atomic-level structures of the HIV surface protein, gp120, together with extensive biophysical analysis of this protein, to design modified vaccine candidates. Immunization with these modified gp120 proteins revealed a new relationship between structure-guided protein stability and the efficient elicitation of antibodies against the highly conserved co-receptor binding site of HIV. These data demonstrate the potential for using the design principles established here to develop improved antibody-generating HIV vaccines and for vaccines against other pathogens.
Collapse
|
494
|
Human immunodeficiency virus type 1 elite neutralizers: individuals with broad and potent neutralizing activity identified by using a high-throughput neutralization assay together with an analytical selection algorithm. J Virol 2009; 83:7337-48. [PMID: 19439467 DOI: 10.1128/jvi.00110-09] [Citation(s) in RCA: 481] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The development of a rapid and efficient system to identify human immunodeficiency virus type 1 (HIV-1)-infected individuals with broad and potent HIV-1-specific neutralizing antibody responses is an important step toward the discovery of critical neutralization targets for rational AIDS vaccine design. In this study, samples from HIV-1-infected volunteers from diverse epidemiological regions were screened for neutralization responses using pseudovirus panels composed of clades A, B, C, and D and circulating recombinant forms (CRFs). Initially, 463 serum and plasma samples from Australia, Rwanda, Uganda, the United Kingdom, and Zambia were screened to explore neutralization patterns and selection ranking algorithms. Samples were identified that neutralized representative isolates from at least four clade/CRF groups with titers above prespecified thresholds and ranked based on a weighted average of their log-transformed neutralization titers. Linear regression methods selected a five-pseudovirus subset, representing clades A, B, and C and one CRF01_AE, that could identify top-ranking samples with 50% inhibitory concentration (IC(50)) neutralization titers of >or=100 to multiple isolates within at least four clade groups. This reduced panel was then used to screen 1,234 new samples from the Ivory Coast, Kenya, South Africa, Thailand, and the United States, and 1% were identified as elite neutralizers. Elite activity is defined as the ability to neutralize, on average, more than one pseudovirus at an IC(50) titer of 300 within a clade group and across at least four clade groups. These elite neutralizers provide promising starting material for the isolation of broadly neutralizing monoclonal antibodies to assist in HIV-1 vaccine design.
Collapse
|
495
|
Granados-González V, Piedrahita LD, Martínez M, Genin C, Riffard S, Urcuqui-Inchima S. [Role of the HIV-1 gp120 V1/V2 domains in the induction of neutralizing antibodies]. Enferm Infecc Microbiol Clin 2009; 27:523-30. [PMID: 19409660 DOI: 10.1016/j.eimc.2008.02.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2007] [Accepted: 02/11/2008] [Indexed: 11/25/2022]
Abstract
The development of a preventive vaccine against human immunodeficiency virus type-1 (HIV-1) provides hope for control of the pandemic over the coming years. Nevertheless, it is clear that one of the greatest difficulties in achieving this vaccine is the high mutation rate of the virus, which enables it to evade the host's immune response. The production of neutralizing antibodies (NAb) against the HIV-1 envelope proteins is believed to play an important role in controlling the infection and in providing effective protection following vaccination. Several studies have shown that the V1/V2 domain of the HIV-1 gp120 envelope protein is involved in viral tropism during infection, in masking conserved neutralizing epitopes, in the conformational changes occurring after coreceptor binding, and in NAb induction. Nonetheless, this domain has been poorly investigated. However, because the V1/V2 domain is highly glycosylated, numerous studies have determined the influence of carbohydrates on NAb production. The present review focuses on the importance of NAb directed against epitopes of the variable regions, mainly V1/V2, their importance in protecting against HIV-1 infection, and the role these regions play in evading the immune response. Lastly, we will discuss the importance of NAb in the search for an effective vaccine against HIV-1.
Collapse
Affiliation(s)
- Viviana Granados-González
- Groupe Immunité des Muqueuses et Agents Pathogènes, University of Saint Etienne, Saint Etienne, Francia.
| | | | | | | | | | | |
Collapse
|
496
|
Examination of the contributions of size and avidity to the neutralization mechanisms of the anti-HIV antibodies b12 and 4E10. Proc Natl Acad Sci U S A 2009; 106:7385-90. [PMID: 19372381 DOI: 10.1073/pnas.0811427106] [Citation(s) in RCA: 127] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Monoclonal antibodies b12 and 4E10 are broadly neutralizing against a variety of strains of the human immunodeficiency virus type 1 (HIV-1). The epitope for b12 maps to the CD4-binding site in the gp120 subunit of HIV-1's trimeric gp120-gp41 envelope spike, whereas 4E10 recognizes the membrane-proximal external region (MPER) of gp41. Here, we constructed and compared a series of architectures for the b12 and 4E10 combining sites that differed in size, valency, and flexibility. In a comparative analysis of the ability of the b12 and 4E10 constructs to neutralize a panel of clade B HIV-1 strains, we observed that the ability of bivalent constructs to cross-link envelope spikes on the virion surface made a greater contribution to neutralization by b12 than by 4E10. Increased distance and flexibility between antibody combining sites correlated with enhanced neutralization for both antibodies, suggesting restricted mobility for the trimeric spikes embedded in the virion surface. The size of a construct did not appear to be correlated with neutralization potency for b12, but larger 4E10 constructs exhibited a steric occlusion effect, which we interpret as evidence for restricted access to its gp41 epitope. The combination of limited avidity and steric occlusion suggests a mechanism for evading neutralization by antibodies that target epitopes in the highly conserved MPER of gp41.
Collapse
|
497
|
Stoll-Keller F, Barth H, Fafi-Kremer S, Zeisel MB, Baumert TF. Development of hepatitis C virus vaccines: challenges and progress. Expert Rev Vaccines 2009; 8:333-45. [PMID: 19249975 DOI: 10.1586/14760584.8.3.333] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Development of an effective vaccine against the hepatitis C virus (HCV) has long been defined as a difficult challenge due to the considerable variability of this RNA virus and the observation that convalescent humans and chimpanzees could be re-infected after re-exposure. On the other hand, progress in the understanding of antiviral immune responses in patients with viral clearance has elucidated key mechanisms playing a role in the control of viral infection. Studies investigating prophylactic vaccine approaches in chimpanzees have confirmed that the induction and maintenance of strong helper and cytotoxic T-cell immune responses against multiple viral epitopes is necessary for protection against viral clearance and chronic infection. A multispecific B-cell response, resulting in rapid induction of cross-neutralizing antibodies may assist cellular responses. Therapeutic vaccine formulations currently being evaluated in clinical trials are facing the fact that the immune system of chronic carriers is impaired and needs the restoration of T-cell functions to enhance their efficacy.
Collapse
Affiliation(s)
- Françoise Stoll-Keller
- Inserm, U748 et Laboratoire de Virologie des Hôpitaux Universitaires de Strasbourg, 3 rue Koeberlé 67000 Strasbourg, France.
| | | | | | | | | |
Collapse
|
498
|
Haim H, Si Z, Madani N, Wang L, Courter JR, Princiotto A, Kassa A, DeGrace M, McGee-Estrada K, Mefford M, Gabuzda D, Smith AB, Sodroski J. Soluble CD4 and CD4-mimetic compounds inhibit HIV-1 infection by induction of a short-lived activated state. PLoS Pathog 2009; 5:e1000360. [PMID: 19343205 PMCID: PMC2655723 DOI: 10.1371/journal.ppat.1000360] [Citation(s) in RCA: 145] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2008] [Accepted: 03/02/2009] [Indexed: 11/19/2022] Open
Abstract
Binding to the CD4 receptor induces conformational changes in the human immunodeficiency virus (HIV-1) gp120 exterior envelope glycoprotein. These changes allow gp120 to bind the coreceptor, either CCR5 or CXCR4, and prime the gp41 transmembrane envelope glycoprotein to mediate virus-cell membrane fusion and virus entry. Soluble forms of CD4 (sCD4) and small-molecule CD4 mimics (here exemplified by JRC-II-191) also induce these conformational changes in the HIV-1 envelope glycoproteins, but typically inhibit HIV-1 entry into CD4-expressing cells. To investigate the mechanism of inhibition, we monitored at high temporal resolution inhibitor-induced changes in the conformation and functional competence of the HIV-1 envelope glycoproteins that immediately follow engagement of the soluble CD4 mimics. Both sCD4 and JRC-II-191 efficiently activated the envelope glycoproteins to mediate infection of cells lacking CD4, in a manner dependent on coreceptor affinity and density. This activated state, however, was transient and was followed by spontaneous and apparently irreversible changes of conformation and by loss of functional competence. The longevity of the activated intermediate depended on temperature and the particular HIV-1 strain, but was indistinguishable for sCD4 and JRC-II-191; by contrast, the activated intermediate induced by cell-surface CD4 was relatively long-lived. The inactivating effects of these activation-based inhibitors predominantly affected cell-free virus, whereas virus that was prebound to the target cell surface was mainly activated, infecting the cells even at high concentrations of the CD4 analogue. These results demonstrate the ability of soluble CD4 mimics to inactivate HIV-1 by prematurely triggering active but transient intermediate states of the envelope glycoproteins. This novel strategy for inhibition may be generally applicable to high-potential-energy viral entry machines that are normally activated by receptor binding.
Collapse
Affiliation(s)
- Hillel Haim
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Division of AIDS, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Zhihai Si
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Division of AIDS, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Navid Madani
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Division of AIDS, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Liping Wang
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Division of AIDS, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Joel R. Courter
- Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Amy Princiotto
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Division of AIDS, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Aemro Kassa
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Division of AIDS, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Marciella DeGrace
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Division of AIDS, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Kathleen McGee-Estrada
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Division of AIDS, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Megan Mefford
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Division of AIDS, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Dana Gabuzda
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Division of AIDS, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Amos B. Smith
- Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Joseph Sodroski
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Division of AIDS, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, Massachusetts, United States of America
| |
Collapse
|
499
|
Kamiyama H, Yoshii H, Tanaka Y, Sato H, Yamamoto N, Kubo Y. Raft localization of CXCR4 is primarily required for X4-tropic human immunodeficiency virus type 1 infection. Virology 2009; 386:23-31. [PMID: 19178925 DOI: 10.1016/j.virol.2008.12.033] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2008] [Revised: 11/10/2008] [Accepted: 12/17/2008] [Indexed: 11/16/2022]
Abstract
Human immunodeficiency virus type 1 (HIV-1) infection is initiated by successive interactions of viral envelope glycoprotein gp120 with two cellular surface proteins, CD4 and chemokine receptor. The two most common chemokine receptors that allow HIV-1 entry are the CCR5 and CXCR4. The CD4 and CCR5 are mainly localized to the particular plasma membrane microdomains, termed raft, which is rich in glycolipids and cholesterol. However, the CXCR4 is localized only partially to the raft region. Although the raft domain is suggested to participate in HIV-1 infection, its role in entry of CXCR4-tropic (X4-tropic) virus is still unclear. Here, we used a combination of CD4-independent infection system and cholesterol-depletion-inducing reagent, methyl-beta-cyclodextrin (MbetaCD), to address the requirement of raft domain in the X4-tropic virus infection. Treatment of CD4-negative, CXCR4-positive human cells with MbetaCD inhibited CD4-independent infection of the X4-tropic strains. This inhibitory effect of the cholesterol depletion was observed even when the CXCR4 was over-expressed on the target cells. Soluble CD4-induced infection was also inhibited by MbetaCD. The MbetaCD had no effect on the levels of cell surface expression of CXCR4. In contrast to these infections, MbetaCD treatment did not inhibit CD4-dependent HIV-1 infection in the wild type CD4-expressing cells. This study and previous reports showing that CD4 mutants localized to non-raft domains function as HIV-1 receptor indicate that CXCR4 clustering in the raft microdomains, rather than CD4, is the key step for the HIV-1 entry.
Collapse
Affiliation(s)
- Haruka Kamiyama
- Department of AIDS Research, Institute of Tropical Medicine, Nagasaki University, 1-12-4 Sakamoto, Nagasaki, Nagasaki 852-8523, Japan
| | | | | | | | | | | |
Collapse
|
500
|
[Vaccines for the future]. ANNALES PHARMACEUTIQUES FRANÇAISES 2009; 67:203-12. [PMID: 19446671 DOI: 10.1016/j.pharma.2009.02.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2008] [Revised: 12/19/2008] [Accepted: 02/06/2009] [Indexed: 12/27/2022]
Abstract
The field of vaccines and vaccinology has seen remarkable progress during the past 20 years. Many vaccines, however, still need to be improved, either because the protection they provide is relatively short-lived and would greatly benefit from the development of booster formulations (as is the case for tuberculosis), or because they only cover part of the many serotypes of the pathogen that causes the disease (rotaviruses, papillomaviruses, or Streptococcus pneumoniae). In addition, still many diseases lack a proper preventive vaccine, such as AIDS, hepatitis C, malaria, viral pneumonias, croup and bronchiolitis, dengue fever, leishmaniasis, Staphylococcus aureus, groups A and B Streptococcus, Shigellas and enterotoxigenic Escherichia coli, to only name a few. These are the current targets of vaccines under development, a great many of which will hopefully reach the market within the coming 10 years. The development of preventive vaccines against chronic diseases such as AIDS and hepatitis C will probably require more time, due to basic science complexities to be overcome first. It is likely that the future will also see an emphasis on therapeutic vaccines targeted against noninfectious diseases such as cancers (lung, skin, prostate, etc) and metabolic or neurologic diseases (atherosclerosis, Alzheimer's disease). This review will focus on examples of preventive vaccines under development that target infectious diseases with a heavy global burden on public health.
Collapse
|