451
|
Julien JP, Cupo A, Sok D, Stanfield RL, Lyumkis D, Deller MC, Klasse PJ, Burton DR, Sanders RW, Moore JP, Ward AB, Wilson IA. Crystal structure of a soluble cleaved HIV-1 envelope trimer. Science 2013; 342:1477-83. [PMID: 24179159 DOI: 10.1126/science.1245625] [Citation(s) in RCA: 706] [Impact Index Per Article: 58.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
HIV-1 entry into CD4(+) target cells is mediated by cleaved envelope glycoprotein (Env) trimers that have been challenging to characterize structurally. Here, we describe the crystal structure at 4.7 angstroms of a soluble, cleaved Env trimer that is stabilized and antigenically near-native (termed the BG505 SOSIP.664 gp140 trimer) in complex with a potent broadly neutralizing antibody, PGT122. The structure shows a prefusion state of gp41, the interaction between the component gp120 and gp41 subunits, and how a close association between the gp120 V1/V2/V3 loops stabilizes the trimer apex around the threefold axis. The complete epitope of PGT122 on the trimer involves gp120 V1, V3, and several surrounding glycans. This trimer structure advances our understanding of how Env functions and is presented to the immune system, and provides a blueprint for structure-based vaccine design.
Collapse
Affiliation(s)
- Jean-Philippe Julien
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
452
|
Sattentau QJ. Envelope Glycoprotein Trimers as HIV-1 Vaccine Immunogens. Vaccines (Basel) 2013; 1:497-512. [PMID: 26344344 PMCID: PMC4494206 DOI: 10.3390/vaccines1040497] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Revised: 10/11/2013] [Accepted: 10/12/2013] [Indexed: 12/15/2022] Open
Abstract
The HIV-1 envelope glycoprotein spike is the target of neutralizing antibody attack, and hence represents the only relevant viral antigen for antibody-based vaccine design. Various approaches have been attempted to recapitulate Env in membrane-anchored and soluble forms, and these will be discussed here in the context of recent successes and challenges still to be overcome.
Collapse
Affiliation(s)
- Quentin J Sattentau
- The Sir William Dunn School of Pathology, The University of Oxford, South Parks Road, Oxford OX13RE, UK.
| |
Collapse
|
453
|
The potential of the human immune system to develop broadly neutralizing HIV-1 antibodies: implications for vaccine development. AIDS 2013; 27:2529-39. [PMID: 24100711 PMCID: PMC3815086 DOI: 10.1097/qad.0000000000000015] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVES AND DESIGN Developing an effective HIV-1 vaccine that elicits broadly neutralizing HIV-1 human antibodies (bnAbs) remains a challenging goal. Extensive studies on HIV-1 have revealed various strategies employed by the virus to escape host immune surveillance. Here, we investigated the human antibody gene repertoires of uninfected and HIV-1-infected individuals at genomic DNA (gDNA) and cDNA levels by deep sequencing followed by high-throughput sequence analysis to determine the frequencies of putative germline antibody genes of known HIV-1 monoclonal bnAbs (bnmAbs). METHODS Combinatorial gDNA and cDNA antibody libraries were constructed using the gDNAs and mRNAs isolated from uninfected and HIV-1-infected human peripheral blood mononuclear cells (PBMCs). All libraries were deep sequenced and sequences analysed using IMGT/HighV-QUEST software (http://imgt.org/HighV-QUEST/index.action). The frequencies of putative germline antibodies of known bnmAbs in the gDNA and cDNA libraries were determined. RESULTS AND CONCLUSION The human gDNA antibody libraries were more diverse in heavy and light chain V-gene lineage usage than the cDNA libraries, indicating that the human gDNA antibody gene repertoires may have more potential than the cDNA repertoires to develop HIV-1 bnAbs. The frequencies of the heavy and kappa and lambda light chain variable regions with identical V(D)J recombinations to known HIV-1 bnmAbs were extremely low in human antibody gene repertoires. However, we found relatively high frequencies of the heavy and kappa and lambda light chain variable regions that used the same V-genes and had the same CDR3 lengths as known HIV-1 bnmAbs regardless of (D)J-gene usage. B-cells bearing B-cell receptors of such heavy and kappa and lambda light chain variable regions may be stimulated to induce HIV-1 bnAbs.
Collapse
|
454
|
Schiffner T, Sattentau QJ, Duncan CJA. Cell-to-cell spread of HIV-1 and evasion of neutralizing antibodies. Vaccine 2013; 31:5789-97. [PMID: 24140477 DOI: 10.1016/j.vaccine.2013.10.020] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Revised: 09/29/2013] [Accepted: 10/04/2013] [Indexed: 01/13/2023]
Abstract
Cell-to-cell spread of human immunodeficiency virus (HIV-1) between immune cells was first observed over 20 years ago. During this time, the question of whether this infection route favours viral evasion of neutralizing antibodies (NAbs) targeting the virus envelope glycoprotein (Env) has been repeatedly investigated, but with conflicting results. A clearer picture has formed in the last few years as more broadly neutralizing antibodies have been isolated and we gain further insight into the mechanisms of HIV-1 transmission at virological and infectious synapses. Nevertheless consensus is still lacking, a situation which may be at least partly explained by variability in the experimental approaches used to study the activity of NAbs in the cell-to-cell context. In this review we focus on the most critical question concerning the activity of NAbs against cell-to-cell transmission: is NAb inhibition of cell-to-cell HIV-1 quantitatively or qualitatively different from cell-free infection? Overall, data consistently show that NAbs are capable of blocking HIV-1 infection at synapses, supporting the concept that cell-to-cell infection occurs through directed transfer of virions accessible to the external environment. However, more recent findings suggest that higher concentrations of certain NAbs might be needed to inhibit synaptic infection, with important potential implications for prophylactic vaccine development. We discuss several mechanistic explanations for this relative and selective loss of activity, and highlight gaps in knowledge that are still to be explored.
Collapse
Affiliation(s)
- Torben Schiffner
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, United Kingdom
| | | | | |
Collapse
|
455
|
Shivatare SS, Chang SH, Tsai TI, Ren CT, Chuang HY, Hsu L, Lin CW, Li ST, Wu CY, Wong CH. Efficient convergent synthesis of bi-, tri-, and tetra-antennary complex type N-glycans and their HIV-1 antigenicity. J Am Chem Soc 2013; 135:15382-91. [PMID: 24032650 DOI: 10.1021/ja409097c] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The structural diversity of glycoproteins often comes from post-translational glycosylation with heterogeneous N-glycans. Understanding the complexity of glycans related to various biochemical processes demands a well-defined synthetic sugar library. We report herein a unified convergent strategy for the rapid production of bi-, tri-, and tetra-antennary complex type N-glycans with and without terminal N-acetylneuraminic acid residues connected via the α-2,6 or α-2,3 linkages. Moreover, using sialyltransferases to install sialic acid can minimize synthetic steps through the use of shared intermediates to simplify the complicated procedures associated with conventional sialic acid chemistry. Furthermore, these synthetic complex oligosaccharides were compiled to create a glycan array for the profiling of HIV-1 broadly neutralizing antibodies PG9 and PG16 that were isolated from HIV infected donors. From the study of antibody PG16, we identified potential natural and unnatural glycan ligands, which may facilitate the design of carbohydrate-based immunogens and hasten the HIV vaccine development.
Collapse
Affiliation(s)
- Sachin S Shivatare
- Genomics Research Center, Academia Sinica , 128 Academia Road, Section 2, Nankang, Taipei 115, Taiwan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
456
|
Bewley CA, Shahzad-ul-Hussan S. Characterizing carbohydrate-protein interactions by nuclear magnetic resonance spectroscopy. Biopolymers 2013; 99:796-806. [PMID: 23784792 PMCID: PMC3820370 DOI: 10.1002/bip.22329] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Accepted: 06/13/2013] [Indexed: 11/07/2022]
Abstract
Interactions between proteins and soluble carbohydrates and/or surface displayed glycans are central to countless recognition, attachment and signaling events in biology. The physical chemical features associated with these binding events vary considerably, depending on the biological system of interest. For example, carbohydrate-protein interactions can be stoichiometric or multivalent, the protein receptors can be monomeric or oligomeric, and the specificity of recognition can be highly stringent or rather promiscuous. Equilibrium dissociation constants for carbohydrate binding are known to vary from micromolar to millimolar, with weak interactions being far more prevalent; and individual carbohydrate-binding sites can be truly symmetrical or merely homologous, and hence, the affinities of individual sites within a single protein can vary, as can the order of binding. Several factors, including the weak affinities with which glycans bind their protein receptors, the dynamic nature of the glycans themselves, and the nonequivalent interactions among oligomeric carbohydrate receptors, have made nuclear magnetic resonance (NMR) an especially powerful tool for studying and defining carbohydrate-protein interactions. Here, we describe those NMR approaches that have proven to be the most robust in characterizing these systems, and explain what type of information can (or cannot) be obtained from each. Our goal is to provide the reader the information necessary for selecting the correct experiment or sets of experiments to characterize their carbohydrate-protein interaction of interest.
Collapse
Affiliation(s)
- Carole A Bewley
- Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD
| | | |
Collapse
|
457
|
van den Kerkhof TLGM, Feenstra KA, Euler Z, van Gils MJ, Rijsdijk LWE, Boeser-Nunnink BD, Heringa J, Schuitemaker H, Sanders RW. HIV-1 envelope glycoprotein signatures that correlate with the development of cross-reactive neutralizing activity. Retrovirology 2013; 10:102. [PMID: 24059682 PMCID: PMC3849187 DOI: 10.1186/1742-4690-10-102] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Accepted: 09/12/2013] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Current HIV-1 envelope glycoprotein (Env) vaccines are unable to induce cross-reactive neutralizing antibodies. However, such antibodies are elicited in 10-30% of HIV-1 infected individuals, but it is unknown why these antibodies are induced in some individuals and not in others. We hypothesized that the Envs of early HIV-1 variants in individuals who develop cross-reactive neutralizing activity (CrNA) might have unique characteristics that support the induction of CrNA. RESULTS We retrospectively generated and analyzed env sequences of early HIV-1 clonal variants from 31 individuals with diverse levels of CrNA 2-4 years post-seroconversion. These sequences revealed a number of Env signatures that coincided with CrNA development. These included a statistically shorter variable region 1 and a lower probability of glycosylation as implied by a high ratio of NXS versus NXT glycosylation motifs. Furthermore, lower probability of glycosylation at position 332, which is involved in the epitopes of many broadly reactive neutralizing antibodies, was associated with the induction of CrNA. Finally, Sequence Harmony identified a number of amino acid changes associated with the development of CrNA. These residues mapped to various Env subdomains, but in particular to the first and fourth variable region as well as the underlying α2 helix of the third constant region. CONCLUSIONS These findings imply that the development of CrNA might depend on specific characteristics of early Env. Env signatures that correlate with the induction of CrNA might be relevant for the design of effective HIV-1 vaccines.
Collapse
Affiliation(s)
- Tom L G M van den Kerkhof
- Department of Experimental Immunology and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
| | - K Anton Feenstra
- Center for Integrative Bioinformatics VU (IBIVU) and Amsterdam Institute for Molecules, Medicine and Systems (AIMMS), VU University Amsterdam, 1081 HV Amsterdam, the Netherlands
- Netherlands Bioinformatics Center (NBIC), 6525 GA Nijmegen, the Netherlands
| | - Zelda Euler
- Department of Experimental Immunology and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
| | - Marit J van Gils
- Department of Experimental Immunology and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
- Department of Medical Microbiology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
| | - Linda W E Rijsdijk
- Center for Integrative Bioinformatics VU (IBIVU) and Amsterdam Institute for Molecules, Medicine and Systems (AIMMS), VU University Amsterdam, 1081 HV Amsterdam, the Netherlands
| | - Brigitte D Boeser-Nunnink
- Department of Experimental Immunology and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
| | - Jaap Heringa
- Center for Integrative Bioinformatics VU (IBIVU) and Amsterdam Institute for Molecules, Medicine and Systems (AIMMS), VU University Amsterdam, 1081 HV Amsterdam, the Netherlands
- Netherlands Bioinformatics Center (NBIC), 6525 GA Nijmegen, the Netherlands
- Department of Medical Microbiology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
| | - Hanneke Schuitemaker
- Department of Experimental Immunology and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
- Crucell Holland BV, 2333 CN Leiden, the Netherlands
| | - Rogier W Sanders
- Department of Medical Microbiology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
- Department of Microbiology and Immunology, Weill Medical College, Cornell University, New York, NY 10065 USA
| |
Collapse
|
458
|
Klein F, Mouquet H, Dosenovic P, Scheid JF, Scharf L, Nussenzweig MC. Antibodies in HIV-1 vaccine development and therapy. Science 2013; 341:1199-204. [PMID: 24031012 DOI: 10.1126/science.1241144] [Citation(s) in RCA: 371] [Impact Index Per Article: 30.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Despite 30 years of study, there is no HIV-1 vaccine and, until recently, there was little hope for a protective immunization. Renewed optimism in this area of research comes in part from the results of a recent vaccine trial and the use of single-cell antibody-cloning techniques that uncovered naturally arising, broad and potent HIV-1-neutralizing antibodies (bNAbs). These antibodies can protect against infection and suppress established HIV-1 infection in animal models. The finding that these antibodies develop in a fraction of infected individuals supports the idea that new approaches to vaccination might be developed by adapting the natural immune strategies or by structure-based immunogen design. Moreover, the success of passive immunotherapy in small-animal models suggests that bNAbs may become a valuable addition to the armamentarium of drugs that work against HIV-1.
Collapse
Affiliation(s)
- Florian Klein
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA.
| | | | | | | | | | | |
Collapse
|
459
|
A next-generation cleaved, soluble HIV-1 Env trimer, BG505 SOSIP.664 gp140, expresses multiple epitopes for broadly neutralizing but not non-neutralizing antibodies. PLoS Pathog 2013; 9:e1003618. [PMID: 24068931 PMCID: PMC3777863 DOI: 10.1371/journal.ppat.1003618] [Citation(s) in RCA: 773] [Impact Index Per Article: 64.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Accepted: 07/30/2013] [Indexed: 01/17/2023] Open
Abstract
A desirable but as yet unachieved property of a human immunodeficiency virus type 1 (HIV-1) vaccine candidate is the ability to induce broadly neutralizing antibodies (bNAbs). One approach to the problem is to create trimeric mimics of the native envelope glycoprotein (Env) spike that expose as many bNAb epitopes as possible, while occluding those for non-neutralizing antibodies (non-NAbs). Here, we describe the design and properties of soluble, cleaved SOSIP.664 gp140 trimers based on the subtype A transmitted/founder strain, BG505. These trimers are highly stable, more so even than the corresponding gp120 monomer, as judged by differential scanning calorimetry. They are also homogenous and closely resemble native virus spikes when visualized by negative stain electron microscopy (EM). We used several techniques, including ELISA and surface plasmon resonance (SPR), to determine the relationship between the ability of monoclonal antibodies (MAbs) to bind the soluble trimers and neutralize the corresponding virus. In general, the concordance was excellent, in that virtually all bNAbs against multiple neutralizing epitopes on HIV-1 Env were highly reactive with the BG505 SOSIP.664 gp140 trimers, including quaternary epitopes (CH01, PG9, PG16 and PGT145). Conversely, non-NAbs to the CD4-binding site, CD4-induced epitopes or gp41ECTO did not react with the trimers, even when their epitopes were present on simpler forms of Env (e.g. gp120 monomers or dissociated gp41 subunits). Three non-neutralizing MAbs to V3 epitopes did, however, react strongly with the trimers but only by ELISA, and not at all by SPR and to only a limited extent by EM. These new soluble trimers are useful for structural studies and are being assessed for their performance as immunogens.
Collapse
|
460
|
Kumar R, Tuen M, Liu J, Nàdas A, Pan R, Kong X, Hioe CE. Elicitation of broadly reactive antibodies against glycan-modulated neutralizing V3 epitopes of HIV-1 by immune complex vaccines. Vaccine 2013; 31:5413-21. [PMID: 24051158 DOI: 10.1016/j.vaccine.2013.09.010] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Revised: 07/11/2013] [Accepted: 09/06/2013] [Indexed: 01/25/2023]
Abstract
HIV-1 envelope gp120 is the target for neutralizing antibodies (NAbs) against the virus. Various approaches have been explored to improve immunogenicity of broadly neutralizing epitopes on this antigen with limited success. We previously demonstrated that immunogenicity of gp120 and especially its V3 epitopes was enhanced when gp120 was co-administered as immune-complex vaccines with monoclonal antibodies (mAb) to the CD4-binding site (CD4bs). To define the mechanisms by which immune complexes influence V3 immunogenicity, we compared gp120 complexed with mAbs specific for the C2 region (1006-30), the V2 loop (2158), or the CD4bs (654), and found that the gp120/654 and gp120/2158 complexes elicited anti-V3 NAbs, but the gp120/654 complex was the most effective. gp120 complexed with 654 F(ab')2 was as potent, indicating that V3 immunogenicity is determined by the specificity of the mAb's Fab fragment used to form the complexes. Importantly, the gp120/654 complex not only induced anti-gp120 antibodies (Abs) to higher titers, but also of greater avidity. The Abs were cross-reactive with V3 peptides from most subtype B and some subtype C isolates. Neutralization was detected only against Tier-1 HIV-1 pseudoviruses, while Tier-2 viruses, including the homologous JRFL strain, were not neutralized. However, JRFL produced in the presence of a mannosidase inhibitor was sensitive to anti-V3 NAbs in the immune sera. These results demonstrate that the gp120/654 complex is a potent immunogen for eliciting cross-reactive functional NAbs against V3 epitopes, of which exposure is determined by the specific compositions of glycans shrouding the HIV-1 envelope glycoproteins.
Collapse
Affiliation(s)
- Rajnish Kumar
- VA New York Harbor Healthcare System, Manhattan Campus and New York University School of Medicine, Department of Pathology, New York, NY 10010, United States
| | | | | | | | | | | | | |
Collapse
|
461
|
Isolation of HIV-1-reactive antibodies using cell surface-expressed gp160Δc(BaL.). J Immunol Methods 2013; 397:47-54. [PMID: 24041474 DOI: 10.1016/j.jim.2013.09.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Revised: 09/04/2013] [Accepted: 09/04/2013] [Indexed: 10/26/2022]
Abstract
Significant efforts have been made to identify HIV-1 neutralizing antibodies because they are considered to be critical to the design of an effective HIV-1 vaccine. Although soluble HIV-1 envelope proteins can be used for this purpose, these reagents differ from membrane-anchored HIV-1 envelope spike in a number of important ways and display only a subset of its native epitopes. Consistent with this, some broadly neutralizing antibodies preferentially bind cell surface-expressed HIV-1 envelope, but not the soluble protein. Here we report the details of a new method for isolating anti-HIV-1 specific B cells based on capturing cells that produce antibodies to cell surface-expressed gp160Δc(BaL). While this method is far less efficient than sorting with soluble envelope proteins, it isolated broadly neutralizing anti-HIV-1 antibodies that bind cell surface-expressed gp160Δc(BaL) but not soluble envelope proteins.
Collapse
|
462
|
HIV-1 envelope glycoprotein resistance to monoclonal antibody 2G12 is subject-specific and context-dependent in macaques and humans. PLoS One 2013; 8:e75277. [PMID: 24040404 PMCID: PMC3767832 DOI: 10.1371/journal.pone.0075277] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Accepted: 08/15/2013] [Indexed: 11/19/2022] Open
Abstract
HIV-1 Envelope (Env) protein is the sole target of neutralizing antibodies (NAbs) that arise during infection to neutralize autologous variants. Under this immune pressure, HIV escape variants are continuously selected and over the course of infection Env becomes more neutralization resistant. Many common alterations are known to affect sensitivity to NAbs, including residues encoding potential N-linked glycosylation sites (PNGS). Knowledge of Env motifs associated with neutralization resistance is valuable for the design of an effective Env-based vaccine so we characterized Envs isolated longitudinally from a SHIV(SF162P4) infected macaque for sensitivity to neutralizing monoclonal antibodies (MAbs) B12, 2G12, 4E10 and 2F5. The early Env, isolated from plasma at day 56 after infection, was the most sensitive and the late Env, from day 670, was the most resistant to MAbs. We identified four PNGS in these Envs that accumulated over time at positions 130, 139, 160 and 397. We determined that removal of these PNGS significantly increased neutralization sensitivity to 2G12, and conversely, we identified mutations by in silico analyses that contributed resistance to 2G12 neutralization. In order to expand our understanding of these PNGS, we analyzed Envs from clade B HIV-infected human subjects and identified additional glycan and amino acid changes that could affect neutralization by 2G12 in a context-dependent manner. Taken together, these in vitro and in silico analyses of clade B Envs revealed that 2G12 resistance is achieved by previously unrecognized PNGS substitutions in a context-dependent manner and by subject-specific pathways.
Collapse
|
463
|
Rabbit anti-HIV-1 monoclonal antibodies raised by immunization can mimic the antigen-binding modes of antibodies derived from HIV-1-infected humans. J Virol 2013; 87:10221-31. [PMID: 23864637 PMCID: PMC3754018 DOI: 10.1128/jvi.00843-13] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The rabbit is a commonly used animal model in studying antibody responses in HIV/AIDS vaccine development. However, no rabbit monoclonal antibodies (MAbs) have been developed previously to study the epitope-specific antibody responses against HIV-1 envelope (Env) glycoproteins, and little is known about how the rabbit immune system can mimic the human immune system in eliciting such antibodies. Here we present structural analyses of two rabbit MAbs, R56 and R20, against the third variable region (V3) of HIV-1 gp120. R56 recognizes the well-studied immunogenic region in the V3 crown, while R20 targets a less-studied region at the C terminus of V3. By comparison of the Fab/epitope complex structures of these two antibodies raised by immunization with that of the corresponding human antibodies derived from patients chronically infected with HIV-1, we found that rabbit antibodies can recognize immunogenic regions of gp120 and mimic the binding modes of human antibodies. This result can provide new insight into the use of the rabbit as an animal model in AIDS vaccine development.
Collapse
|
464
|
Chen Y, Vaine M, Wallace A, Han D, Wan S, Seaman MS, Montefiori D, Wang S, Lu S. A novel rabbit monoclonal antibody platform to dissect the diverse repertoire of antibody epitopes for HIV-1 Env immunogen design. J Virol 2013; 87:10232-43. [PMID: 23864612 PMCID: PMC3754024 DOI: 10.1128/jvi.00837-13] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Accepted: 07/07/2013] [Indexed: 01/13/2023] Open
Abstract
The majority of available monoclonal antibodies (MAbs) in the current HIV vaccine field are generated from HIV-1-infected people. In contrast, preclinical immunogenicity studies have mainly focused on polyclonal antibody responses in experimental animals. Although rabbits have been widely used for antibody studies, there has been no report of using rabbit MAbs to dissect the specificity of antibody responses for AIDS vaccine development. Here we report on the production of a panel of 12 MAbs from a New Zealand White (NZW) rabbit that was immunized with an HIV-1 JR-FL gp120 DNA prime and protein boost vaccination regimen. These rabbit MAbs recognized a diverse repertoire of envelope (Env) epitopes ranging from the highly immunogenic V3 region to several previously underappreciated epitopes in the C1, C4, and C5 regions. Nine MAbs showed cross-reactivity to gp120s of clades other than clade B. Increased somatic mutation and extended CDR3 were observed with Ig genes of several molecularly cloned rabbit MAbs. Phylogenic tree analysis showed that the heavy chains of MAbs recognizing the same region on gp120 tend to segregate into an independent subtree. At least three rabbit MAbs showed neutralizing activities with various degrees of breadth and potency. The establishment of this rabbit MAb platform will significantly enhance our ability to test optimal designs of Env immunogens to gain a better understanding of the structural specificity and evolution process of Env-specific antibody responses elicited by candidate AIDS vaccines.
Collapse
Affiliation(s)
- Yuxin Chen
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Michael Vaine
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Aaron Wallace
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Dong Han
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Shengqin Wan
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Michael S. Seaman
- Department of Medicine, Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - David Montefiori
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Shixia Wang
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Shan Lu
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
465
|
Schiffner T, Kong L, Duncan CJA, Back JW, Benschop JJ, Shen X, Huang PS, Stewart-Jones GB, DeStefano J, Seaman MS, Tomaras GD, Montefiori DC, Schief WR, Sattentau QJ. Immune focusing and enhanced neutralization induced by HIV-1 gp140 chemical cross-linking. J Virol 2013; 87:10163-72. [PMID: 23843636 PMCID: PMC3754013 DOI: 10.1128/jvi.01161-13] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 07/04/2013] [Indexed: 11/20/2022] Open
Abstract
Experimental vaccine antigens based upon the HIV-1 envelope glycoproteins (Env) have failed to induce neutralizing antibodies (NAbs) against the majority of circulating viral strains as a result of antibody evasion mechanisms, including amino acid variability and conformational instability. A potential vaccine design strategy is to stabilize Env, thereby focusing antibody responses on constitutively exposed, conserved surfaces, such as the CD4 binding site (CD4bs). Here, we show that a largely trimeric form of soluble Env can be stably cross-linked with glutaraldehyde (GLA) without global modification of antigenicity. Cross-linking largely conserved binding of all potent broadly neutralizing antibodies (bNAbs) tested, including CD4bs-specific VRC01 and HJ16, but reduced binding of several non- or weakly neutralizing antibodies and soluble CD4 (sCD4). Adjuvanted administration of cross-linked or unmodified gp140 to rabbits generated indistinguishable total gp140-specific serum IgG binding titers. However, sera from animals receiving cross-linked gp140 showed significantly increased CD4bs-specific antibody binding compared to animals receiving unmodified gp140. Moreover, peptide mapping of sera from animals receiving cross-linked gp140 revealed increased binding to gp120 C1 and V1V2 regions. Finally, neutralization titers were significantly elevated in sera from animals receiving cross-linked gp140 rather than unmodified gp140. We conclude that cross-linking favors antigen stability, imparts antigenic modifications that selectively refocus antibody specificity and improves induction of NAbs, and might be a useful strategy for future vaccine design.
Collapse
Affiliation(s)
- T. Schiffner
- The Sir William Dunn School of Pathology, Oxford, United Kingdom
| | - L. Kong
- The Sir William Dunn School of Pathology, Oxford, United Kingdom
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California, USA
| | - C. J. A. Duncan
- The Sir William Dunn School of Pathology, Oxford, United Kingdom
| | - J. W. Back
- Pepscan Therapeutics, Lelystad, The Netherlands
| | | | - X. Shen
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - P. S. Huang
- Department of Biochemistry, University of Washington, Seattle, Washington, USA
| | - G. B. Stewart-Jones
- The Weatherall Institute of Molecular Medicine, The John Radcliffe Hospital, Oxford, United Kingdom
| | - J. DeStefano
- International AIDS Vaccine Initiative, Brooklyn, New York, USA
| | - M. S. Seaman
- Division of Viral Pathogenesis, Department of Medicine, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - G. D. Tomaras
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - D. C. Montefiori
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - W. R. Schief
- Department of Biochemistry, University of Washington, Seattle, Washington, USA
- IAVI Neutralizing Antibody Center and Department of Immunology and Microbial Sciences, The Scripps Research Institute, La Jolla, California, USA
| | - Q. J. Sattentau
- The Sir William Dunn School of Pathology, Oxford, United Kingdom
| |
Collapse
|
466
|
Abstract
Antiviral vaccines have been the most successful biomedical intervention for preventing epidemic viral disease. Vaccination for smallpox in humans and rinderpest in cattle was the basis for disease eradication, and recent progress in polio eradication is promising. Although early vaccines were developed empirically by passage in live animals or eggs, more recent vaccines have been developed because of the advent of new technologies, particularly cell culture and molecular biology. Recent technological advances in gene delivery and expression, nanoparticles, protein manufacturing, and adjuvants have created the potential for new vaccine platforms that may provide solutions for vaccines against viral pathogens for which no interventions currently exist. In addition, the technological convergence of human monoclonal antibody isolation, structural biology, and high-throughput sequencing is providing new opportunities for atomic-level immunogen design. Selection of human monoclonal antibodies can identify immunodominant antigenic sites associated with neutralization and provide reagents for stabilizing and solving the structure of viral surface proteins. Understanding the structural basis for neutralization can guide selection of vaccine targets. Deep sequencing of the antibody repertoire and defining the ontogeny of the desired antibody responses can reveal the junctional recombination and somatic mutation requirements for B-cell recognition and affinity maturation. Collectively, this information will provide new strategic approaches for selecting vaccine antigens, formulations, and regimens. Moreover, it creates the potential for rational vaccine design and establishing a catalogue of vaccine technology platforms that would be effective against any given family or class of viral pathogens and improve our readiness to address new emerging viral threats.
Collapse
Affiliation(s)
- Barney S Graham
- NIAID, NIH, Vaccine Research Center, Bethesda, MD 20892-3017, USA.
| |
Collapse
|
467
|
Gach JS, Quendler H, Tong T, Narayan KM, Du SX, Whalen RG, Binley JM, Forthal DN, Poignard P, Zwick MB. A human antibody to the CD4 binding site of gp120 capable of highly potent but sporadic cross clade neutralization of primary HIV-1. PLoS One 2013; 8:e72054. [PMID: 23991039 PMCID: PMC3753353 DOI: 10.1371/journal.pone.0072054] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Accepted: 07/06/2013] [Indexed: 01/21/2023] Open
Abstract
Primary isolates of HIV-1 resist neutralization by most antibodies to the CD4 binding site (CD4bs) on gp120 due to occlusion of this site on the trimeric spike. We describe 1F7, a human CD4bs monoclonal antibody that was found to be exceptionally potent against the HIV-1 primary isolate JR-FL. However, 1F7 failed to neutralize a patient-matched primary isolate, JR-CSF even though the two isolates differ by <10% in gp120 at the protein level. In an HIV-1 cross clade panel (n = 157), 1F7 exhibited moderate breadth, but occasionally achieved considerable potency. In binding experiments using monomeric gp120s of select resistant isolates and domain-swap chimeras between JR-FL and JR-CSF, recognition by 1F7 was limited by sequence polymorphisms involving at least the C2 region of Env. Putative N-linked glycosylation site (PNGS) mutations, notably at position 197, allowed 1F7 to neutralize JR-CSF potently without improving binding to the cognate, monomeric gp120. In contrast, flow cytometry experiments using the same PNGS mutants revealed that 1F7 binding is enhanced on cognate trimeric Env. BN-PAGE mobility shift experiments revealed that 1F7 is sensitive to the diagnostic mutation D368R in the CD4 binding loop of gp120. Our data on 1F7 reinforce how exquisitely targeted CD4bs antibodies must be to achieve cross neutralization of two closely related primary isolates. High-resolution analyses of trimeric Env that show the orientation of glycans and polymorphic elements of the CD4bs that affect binding to antibodies like 1F7 are desirable to understand how to promote immunogenicity of more conserved elements of the CD4bs.
Collapse
Affiliation(s)
- Johannes S. Gach
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California, United States of America
- Division of Infectious Diseases, University of California Irvine, Irvine, California, United States of America
- * E-mail: (JSG); (MBZ)
| | - Heribert Quendler
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California, United States of America
| | - Tommy Tong
- Torrey Pines Institute for Molecular Studies, San Diego, California, United States of America
| | | | - Sean X. Du
- Altravax, Inc., Sunnyvale, California, United States of America
| | | | - James M. Binley
- Torrey Pines Institute for Molecular Studies, San Diego, California, United States of America
| | - Donald N. Forthal
- Division of Infectious Diseases, University of California Irvine, Irvine, California, United States of America
| | - Pascal Poignard
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California, United States of America
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, California, United States of America
| | - Michael B. Zwick
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California, United States of America
- * E-mail: (JSG); (MBZ)
| |
Collapse
|
468
|
Bertok T, Klukova L, Sediva A, Kasak P, Semak V, Micusik M, Omastova M, Chovanová L, Vlček M, Imrich R, Vikartovska A, Tkac J. Ultrasensitive impedimetric lectin biosensors with efficient antifouling properties applied in glycoprofiling of human serum samples. Anal Chem 2013; 85:7324-32. [PMID: 23808876 PMCID: PMC4881809 DOI: 10.1021/ac401281t] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Ultrasensitive impedimetric lectin biosensors recognizing different glycan entities on serum glycoproteins were constructed. Lectins were immobilized on a novel mixed self-assembled monolayer containing 11-mercaptoundecanoic acid for covalent immobilization of lectins and betaine terminated thiol to resist nonspecific interactions. Construction of biosensors based on Concanavalin A (Con A), Sambucus nigra agglutinin type I (SNA), and Ricinus communis agglutinin (RCA) on polycrystalline gold electrodes was optimized and characterized with a battery of tools including electrochemical impedance spectroscopy, various electrochemical techniques, quartz crystal microbalance (QCM), Fourier transform infrared (FT-IR) spectroscopy, atomic force microscopy (AFM), and X-ray photoelectron spectroscopy (XPS) and compared with a protein/lectin microarray. The lectin biosensors were able to detect glycoproteins from 1 fM (Con A), 10 fM (Ricinus communis agglutinin (RCA), or 100 fM (SNA) with a linear range spanning 6 (SNA), 7 (RCA), or 8 (Con A) orders of magnitude. Furthermore, a detection limit for the Con A biosensor down to 1 aM was achieved in a sandwich configuration. A nonspecific binding of proteins for the Con A biosensor was only 6.1% (probed with an oxidized invertase) of the signal toward its analyte invertase and a negligible nonspecific interaction of the Con A biosensor was observed in diluted human sera (1000×), as well. The performance of the lectin biosensors was finally tested by glycoprofiling of human serum samples from healthy individuals and those having rheumatoid arthritis, which resulted in a distinct glycan pattern between these two groups.
Collapse
Affiliation(s)
- Tomas Bertok
- Department of Glycobiotechnology, Institute of Chemistry, Slovak Academy of Sciences, Dúbravská cesta 9, 845 38, Bratislava, Slovak Republic
| | - Ludmila Klukova
- Department of Glycobiotechnology, Institute of Chemistry, Slovak Academy of Sciences, Dúbravská cesta 9, 845 38, Bratislava, Slovak Republic
| | - Alena Sediva
- Department of Glycobiotechnology, Institute of Chemistry, Slovak Academy of Sciences, Dúbravská cesta 9, 845 38, Bratislava, Slovak Republic
| | - Peter Kasak
- Center for Advanced Materials, Qatar University, P.O.Box 2713, Doha, Qatar
| | - Vladislav Semak
- Department of Composite Materials, Polymer Institute, Slovak Academy of Sciences, Dúbravská cesta 9, 845 41, Bratislava, Slovak Republic
| | - Matej Micusik
- Department of Composite Materials, Polymer Institute, Slovak Academy of Sciences, Dúbravská cesta 9, 845 41, Bratislava, Slovak Republic
| | - Maria Omastova
- Department of Composite Materials, Polymer Institute, Slovak Academy of Sciences, Dúbravská cesta 9, 845 41, Bratislava, Slovak Republic
| | - Lucia Chovanová
- Laboratory of Human Endocrinology, Institute of Experimental Endocrinology, Slovak Academy of Sciences, Vlárska 3, 833 06, Bratislava, Slovak Republic
| | - Miroslav Vlček
- Laboratory of Human Endocrinology, Institute of Experimental Endocrinology, Slovak Academy of Sciences, Vlárska 3, 833 06, Bratislava, Slovak Republic
| | - Richard Imrich
- Laboratory of Human Endocrinology, Institute of Experimental Endocrinology, Slovak Academy of Sciences, Vlárska 3, 833 06, Bratislava, Slovak Republic
| | - Alica Vikartovska
- Department of Glycobiotechnology, Institute of Chemistry, Slovak Academy of Sciences, Dúbravská cesta 9, 845 38, Bratislava, Slovak Republic
| | - Jan Tkac
- Department of Glycobiotechnology, Institute of Chemistry, Slovak Academy of Sciences, Dúbravská cesta 9, 845 38, Bratislava, Slovak Republic
| |
Collapse
|
469
|
Yuan T, Li J, Zhang MY. HIV-1 envelope glycoprotein variable loops are indispensable for envelope structural integrity and virus entry. PLoS One 2013; 8:e69789. [PMID: 23936354 PMCID: PMC3731308 DOI: 10.1371/journal.pone.0069789] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Accepted: 06/17/2013] [Indexed: 11/19/2022] Open
Abstract
HIV-1 envelope (Env) glycoprotein is a trimer of heterodimer of gp120 and gp41, and derives from a trimeric glycoprotein precursor, gp160. Gp120 contains five conserved regions that are interspersed with 5 variable loop regions (V1-V5). Env variations in variable loop length and amino acid composition may associate with virus pathogenesis, virus sensitivity to neutralizing antibodies (nAbs) and disease progression. To investigate the role of each variable loop in Env function, we generated a panel of JRFL gp160 loop deletion mutants and examined the effects of each loop deletion on Env expression, Env cell surface display and Env-mediated virus entry into permissive cells. We found that deletion of V1 and V2 (ΔV1V2), or loop D (ΔlpD) abolished virus entry, the same effect as deletion of V3 (ΔV3), while deletion of V3 crown (ΔV3C) significantly enhanced virus assembly and entry. We further found that deletion of V4 (ΔV4) or V5 (ΔV5), or replacement of V4 or V5 with flexible linkers of the same lengths knocked out the receptor and coreceptor binding sites in gp120, but significantly enhanced the exposure of the N-trimer structure and the membrane proximal external region (MPER) in gp41. Although deletion of V4 or V5 did not affect Env expression, they negatively affected Env cell surface display, leading to the failure in virus assembly and subsequent entry. Taken together, we found that Env variable loops were indispensable for Env structural integrity and virus entry. Our findings may have implications for development of HIV-1 vaccine immunogens and therapeutics.
Collapse
Affiliation(s)
- Tingting Yuan
- AIDS Institute, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Jingjing Li
- AIDS Institute, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Mei-Yun Zhang
- AIDS Institute, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- * E-mail:
| |
Collapse
|
470
|
Adamo R, Nilo A, Castagner B, Boutureira O, Berti F, Bernardes GJL. Synthetically defined glycoprotein vaccines: current status and future directions. Chem Sci 2013; 4:2995-3008. [PMID: 25893089 PMCID: PMC4396375 DOI: 10.1039/c3sc50862e] [Citation(s) in RCA: 122] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2013] [Accepted: 05/03/2013] [Indexed: 12/19/2022] Open
Abstract
Primary examples in vaccine design have shown good levels of carbohydrate-specific antibody generation when raised using extracted or fully synthetic capsular polysaccharide glycans covalently coupled to a protein carrier. Herein, we cover recent clinical developments of carbohydrate-based vaccines and describe how novel cutting-edge methodology for the total synthesis of oligosaccharides and for the precise placement of carbohydrates at pre-determined sites within a protein may be used to further improve the safety and efficacy of glycovaccines.
Collapse
Affiliation(s)
- Roberto Adamo
- Research Center , Novartis Vaccines and Diagnostics , Via Fiorentina 1 , 53100 Siena , Italy .
| | - Alberto Nilo
- Research Center , Novartis Vaccines and Diagnostics , Via Fiorentina 1 , 53100 Siena , Italy .
| | - Bastien Castagner
- Department of Chemistry and Applied Biosciences , ETH Zürich , Wolfgang-Pauli-Str. 10 , 8093 Zürich , Switzerland
| | - Omar Boutureira
- Departament de Química Analítica i Química Orgànica , Universitat Rovira i Virgili , C/Marcel·lí Domingo s/n , 43007 Tarragona , Spain
| | - Francesco Berti
- Research Center , Novartis Vaccines and Diagnostics , Via Fiorentina 1 , 53100 Siena , Italy .
| | - Gonçalo J L Bernardes
- Department of Chemistry , University of Cambridge , Lensfield Road , Cambridge , CB2 1EW , UK . ; Instituto de Medicina Molecular , Faculdade de Medicina da Universidade de Lisboa , Av. Prof. Egas Moniz , 1649-028 Lisboa , Portugal .
| |
Collapse
|
471
|
Ho YS, Saksena NK. Glycosylation in HIV-1 envelope glycoprotein and its biological implications. Future Virol 2013. [DOI: 10.2217/fvl.13.64] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Glycosylation of HIV-1 envelope proteins (Env gp120/gp41) plays a vital role in viral evasion from the host immune response, which occurs through the masking of key neutralization epitopes and the presentation of the Env glycosylation as ‘self’ to the host immune system. Env glycosylation is generally conserved, yet its continual evolution plays an important role in modulating viral infectivity and Env immunogenicity. Thus, it is believed that Env glycosylation, which is a vital part of the HIV-1 architecture, also controls intra- and inter-clade genetic variations. Discerning intra- and inter-clade glycosylation variations could therefore yield important information for understanding the molecular and biological differences between HIV clades and may assist in effectively designing Env-based immunogens and in clearly understanding HIV vaccines. This review provides an in-depth perspective of various aspects of Env glycosylation in the context of HIV-1 pathogenesis.
Collapse
Affiliation(s)
- Yung Shwen Ho
- Computational Bioscience Research Center, Biological & Environmental Sciences & Engineering Division, King Abdullah University of Science & Technology, Thuwal 23955, Kingdom of Saudi Arabia
| | - Nitin K Saksena
- Retroviral Genetics Division, Center for Virus Research, Westmead Millennium Instiute & Westmead Hospital, University of Sydney, Westmead, Sydney, NSW 2145, Australia
| |
Collapse
|
472
|
Host-pathogen interaction in HIV infection. Curr Opin Immunol 2013; 25:463-9. [PMID: 23890585 DOI: 10.1016/j.coi.2013.07.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Revised: 06/26/2013] [Accepted: 07/04/2013] [Indexed: 12/24/2022]
Abstract
The host-pathogen interaction is strikingly complex during HIV infection. While several immune effector mechanisms (i.e. cytotoxic T cells, neutralizing antibodies, NK cells, among others) can play a strong antiviral role in vivo, the virus is remarkably able to evade these responses. In addition, the virus preferentially infects and kills activated memory CD4+ T cells, thus exploiting the host antiviral immune response as a source of new cellular targets for infection. Recent advances in understanding (i) how HIV perturbs the host immune system, (ii) how the immune system fights HIV; and (iii) how HIV disease persists when virus replication is suppressed by antiretroviral drugs may hopefully lead to better prevention and treatment strategies for this deadly viral infection.
Collapse
|
473
|
Bispecific antibodies directed to CD4 domain 2 and HIV envelope exhibit exceptional breadth and picomolar potency against HIV-1. Proc Natl Acad Sci U S A 2013; 110:13540-5. [PMID: 23878231 DOI: 10.1073/pnas.1304985110] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
In the absence of an effective HIV-1 vaccine, passive immunization using broadly neutralizing Abs or Ab-like molecules could provide an alternative to the daily administration of oral antiretroviral agents that has recently shown promise as preexposure prophylaxis. Currently, no single broadly neutralizing Ab (bNAb) or combination of bNAbs neutralizes all HIV-1 strains at practically achievable concentrations in vivo. To address this problem, we created bispecific Abs that combine the HIV-1 inhibitory activity of ibalizumab (iMab), a humanized mAb directed to domain 2 of human CD4, with that of anti-gp120 bNAbs. These bispecific bNAbs (BibNAbs) exploit iMab's potent anti-HIV-1 activity and demonstrated clinical efficacy and safety to anchor and thereby concentrate a second broadly neutralizing agent at the site of viral entry. Two BibNabs, PG9-iMab and PG16-iMab, exhibit exceptional breadth and potency, neutralizing 100% of the 118 viruses tested at low picomolar concentrations, including viruses resistant to both parental mAbs. The enhanced potency of these BibNAbs was entirely dependent on CD4 anchoring, not on membrane anchoring per se, and required optimal Ab geometry and linker length. We propose that iMab-based BibNAbs, such as PG9-iMab and PG16-iMab, are promising candidates for passive immunization to prevent HIV-1 infection.
Collapse
|
474
|
Schiffner T, Sattentau QJ, Dorrell L. Development of prophylactic vaccines against HIV-1. Retrovirology 2013; 10:72. [PMID: 23866844 PMCID: PMC3722125 DOI: 10.1186/1742-4690-10-72] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Accepted: 07/11/2013] [Indexed: 01/12/2023] Open
Abstract
The focus of most current HIV-1 vaccine development is on antibody-based approaches. This is because certain antibody responses correlated with protection from HIV-1 acquisition in the RV144 phase III trial, and because a series of potent and broad spectrum neutralizing antibodies have been isolated from infected individuals. Taken together, these two findings suggest ways forward to develop a neutralizing antibody-based vaccine. However, understanding of the correlates of protection from disease in HIV-1 and other infections strongly suggests that we should not ignore CTL-based research. Here we review recent progress in the field and highlight the challenges implicit in HIV-1 vaccine design and some potential solutions.
Collapse
Affiliation(s)
- Torben Schiffner
- The Sir William Dunn School of Pathology, The University of Oxford, South Parks Road, Oxford OX1 3RE, UK
| | | | | |
Collapse
|
475
|
Heavy chain-only IgG2b llama antibody effects near-pan HIV-1 neutralization by recognizing a CD4-induced epitope that includes elements of coreceptor- and CD4-binding sites. J Virol 2013; 87:10173-81. [PMID: 23843638 DOI: 10.1128/jvi.01332-13] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The conserved HIV-1 site of coreceptor binding is protected from antibody-directed neutralization by conformational and steric restrictions. While inaccessible to most human antibodies, the coreceptor site has been shown to be accessed by antibody fragments. In this study, we used X-ray crystallography, surface plasmon resonance, and pseudovirus neutralization to characterize the gp120-envelope glycoprotein recognition and HIV-1 neutralization of a heavy chain-only llama antibody, named JM4. We describe full-length IgG2b and IgG3 versions of JM4 that target the coreceptor-binding site and potently neutralize over 95% of circulating HIV-1 isolates. Contrary to established trends that show improved access to the coreceptor-binding region by smaller antibody fragments, the single-domain (VHH) version of JM4 neutralized less well than the full-length IgG2b version of JM4. The crystal structure at 2.1-Å resolution of VHH JM4 bound to HIV-1 YU2 gp120 stabilized in the CD4-bound state by the CD4-mimetic miniprotein, M48U1, revealed a JM4 epitope that combined regions of coreceptor recognition (including the gp120 bridging sheet, V3 loop, and β19 strand) with gp120 structural elements involved in recognition of CD4 such as the CD4-binding loop. The structure of JM4 with gp120 thus defines a novel CD4-induced site of vulnerability involving elements of both coreceptor- and CD4-binding sites. The potently neutralizing JM4 IgG2b antibody that targets this newly defined site of vulnerability adds to the expanding repertoire of broadly neutralizing antibodies that effectively neutralize HIV-1 and thereby potentially provides a new template for vaccine development and target for HIV-1 therapy.
Collapse
|
476
|
Francesconi O, Nativi C, Gabrielli G, Gentili M, Palchetti M, Bonora B, Roelens S. Pyrrolic Tripodal Receptors for the Molecular Recognition of Carbohydrates: Ditopic Receptors for Dimannosides. Chemistry 2013; 19:11742-52. [DOI: 10.1002/chem.201204298] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Indexed: 11/08/2022]
|
477
|
Feizi T. Carbohydrate recognition in the immune system: contributions of neoglycolipid-based microarrays to carbohydrate ligand discovery. Ann N Y Acad Sci 2013; 1292:33-44. [PMID: 23834439 PMCID: PMC4260124 DOI: 10.1111/nyas.12210] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Oligosaccharide sequences in glycomes of eukaryotes and prokaryotes are enormously diverse. The reasons are not fully understood, but there is an increasing number of examples of the involvement of specific oligosaccharide sequences as ligands in protein-carbohydrate interactions in health and, directly or indirectly, in every major disease, be it infectious or noninfectious. The pinpointing and characterizing of oligosaccharide ligands within glycomes has been one of the most challenging aspects of molecular cell biology, as oligosaccharides cannot be cloned and are generally available in limited amounts. This overview recounts the background to the development of a microarray system that is poised for surveying proteomes for carbohydrate-binding activities and glycomes for assigning the oligosaccharide ligands. Examples are selected by way of illustrating the potential of "designer" microarrays for ligand discovery at the interface of infection, immunity, and glycobiology. Particularly highlighted are sulfo-oligosaccharide and gluco-oligosaccharide recognition systems elucidated using microarrays.
Collapse
Affiliation(s)
- Ten Feizi
- The Glycosciences Laboratory, Department of Medicine, Imperial College London, London, United Kingdom.
| |
Collapse
|
478
|
Bouvin-Pley M, Morgand M, Moreau A, Jestin P, Simonnet C, Tran L, Goujard C, Meyer L, Barin F, Braibant M. Evidence for a continuous drift of the HIV-1 species towards higher resistance to neutralizing antibodies over the course of the epidemic. PLoS Pathog 2013; 9:e1003477. [PMID: 23853594 PMCID: PMC3701719 DOI: 10.1371/journal.ppat.1003477] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Accepted: 05/21/2013] [Indexed: 11/24/2022] Open
Abstract
We compared the neutralization sensitivity of early/transmitted HIV-1 variants from patients infected by subtype B viruses at 3 periods of the epidemic (1987–1991, 1996–2000, 2006–2010). Infectious pseudotyped viruses expressing envelope glycoproteins representative of the viral quasi-species infecting each patient were tested for sensitivity to neutralization by pools of sera from HIV-1 chronically infected patients and by an updated panel of 13 human monoclonal neutralizing antibodies (HuMoNAbs). A progressive significantly enhanced resistance to neutralization was observed over calendar time, by both human sera and most of the HuMoNAbs tested (b12, VRC01, VRC03, NIH45-46G54W, PG9, PG16, PGT121, PGT128, PGT145). Despite this evolution, a combination of two HuMoNAbs (NIH45-46G54W and PGT128) still would efficiently neutralize the most contemporary transmitted variants. In addition, we observed a significant reduction of the heterologous neutralizing activity of sera from individuals infected most recently (2003–2007) compared to patients infected earlier (1987–1991), suggesting that the increasing resistance of the HIV species to neutralization over time coincided with a decreased immunogenicity. These data provide evidence for an ongoing adaptation of the HIV-1 species to the humoral immunity of the human population, which may add an additional obstacle to the design of an efficient HIV-1 vaccine. Most of the patients develop autologous neutralizing antibodies (NAbs) during HIV-1 infection. These NAbs drive the viral evolution and lead to the selection of escape variants at the individual level. The aim of our study was to check if, subsequently to the selective pressure exerted by the individual NAbs responses, the HIV-1 species has evolved at the population level towards an enhanced resistance to antibody neutralization. By comparing HIV-1 subtype B variants collected at three periods spanning more than 2 decades, we found a significantly progressive enhanced resistance to neutralization of the HIV-1 species over time. In addition, the enhanced resistance of the HIV species to neutralization coincided with a decreased capability of the virus to induce NAbs in infected patients. Despite this evolution, one combination of two human monoclonal broadly NAbs still were able to neutralize the most recent HIV-1 variants, suggesting that this combination should be preferentially included in future human immunoprophylaxis trials.
Collapse
Affiliation(s)
| | - Marion Morgand
- Université François Rabelais, Inserm U966, Tours, France
| | - Alain Moreau
- Université François Rabelais, Inserm U966, Tours, France
| | - Pauline Jestin
- Université Paris Sud, CESP Inserm U1018, Paris, France
- AP-HP Hôpital de Bicêtre, Le Kremlin-Bicêtre, France
| | | | - Laurent Tran
- Université Paris Sud, CESP Inserm U1018, Paris, France
- AP-HP Hôpital de Bicêtre, Le Kremlin-Bicêtre, France
- for the ANRS SEROCO and PRIMO study groups
| | - Cécile Goujard
- Université Paris Sud, CESP Inserm U1018, Paris, France
- AP-HP Hôpital de Bicêtre, Le Kremlin-Bicêtre, France
- for the ANRS SEROCO and PRIMO study groups
| | - Laurence Meyer
- Université Paris Sud, CESP Inserm U1018, Paris, France
- AP-HP Hôpital de Bicêtre, Le Kremlin-Bicêtre, France
- for the ANRS SEROCO and PRIMO study groups
| | - Francis Barin
- Université François Rabelais, Inserm U966, Tours, France
- Centre National de Référence VIH, Laboratoire de Bactériologie-Virologie, CHU Bretonneau, Tours, France
| | - Martine Braibant
- Université François Rabelais, Inserm U966, Tours, France
- * E-mail:
| |
Collapse
|
479
|
Influences on trimerization and aggregation of soluble, cleaved HIV-1 SOSIP envelope glycoprotein. J Virol 2013; 87:9873-85. [PMID: 23824824 DOI: 10.1128/jvi.01226-13] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We describe methods to improve the properties of soluble, cleaved gp140 trimers of the human immunodeficiency virus type 1 (HIV-1) envelope glycoproteins (Env) for use in structural studies and as immunogens. In the absence of nonionic detergents, gp140 of the KNH1144 genotype, terminating at residue 681 in gp41 (SOSIP.681), has a tendency to form higher-order complexes or aggregates, which is particularly undesirable for structure-based research. We found that this aggregation in the absence of detergent does not involve the V1, V2, or V3 variable regions of gp120. Moreover, we observed that detergent forms micelles around the membrane-proximal external region (MPER) of the SOSIP.681 gp140 trimers, whereas deletion of most of the MPER residues by terminating the gp140 at residue 664 (SOSIP.664) prevented the aggregation that otherwise occurs in SOSIP.681 in the absence of detergent. Although the MPER can contribute to trimer formation, truncation of most of it only modestly reduced trimerization and lacked global adverse effects on antigenicity. Thus, the MPER deletion minimally influenced the kinetics of the binding of soluble CD4 and a CD4-binding site antibody to immobilized trimers, as detected by surface plasmon resonance. Furthermore, the MPER deletion did not alter the overall three-dimensional structure of the trimers, as viewed by negative-stain electron microscopy. Homogeneous and aggregate-free MPER-truncated SOSIP Env trimers are therefore useful for immunogenicity and structural studies.
Collapse
|
480
|
Crispin M, Bowden TA. Antibodies expose multiple weaknesses in the glycan shield of HIV. Nat Struct Mol Biol 2013; 20:771-2. [DOI: 10.1038/nsmb.2627] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
481
|
Structural characterization of cleaved, soluble HIV-1 envelope glycoprotein trimers. J Virol 2013; 87:9865-72. [PMID: 23824817 DOI: 10.1128/jvi.01222-13] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Human immunodeficiency virus type 1 (HIV-1) infection is a significant global public health problem for which development of an effective prophylactic vaccine remains a high scientific priority. Many concepts for a vaccine are focused on induction of appropriate titers of broadly neutralizing antibodies (bNAbs) against the viral envelope (Env) glycoproteins gp120 and gp41, but no immunogen has yet accomplished this goal in animals or humans. One approach to induction of bNAbs is to design soluble, trimeric mimics of the native viral Env trimer. Here, we describe structural studies by negative-stain electron microscopy of several variants of soluble Env trimers based on the KNH1144 subtype A sequence. These Env trimers are fully cleaved between the gp120 and gp41 components and stabilized by specific amino acid substitutions. We also illustrate the structural consequences of deletion of the V1/V2 and V3 variable loops from gp120 and the membrane-proximal external region (MPER) from gp41. All of these variants adopt a trimeric configuration that appropriately mimics native Env spikes, including the CD4 receptor-binding site and the epitope for the VRC PG04 bNAb. These cleaved, soluble trimer designs can be adapted for use with multiple different env genes for both vaccine and structural studies.
Collapse
|
482
|
Abstract
The development of an effective vaccine has been hindered by the enormous diversity of human immunodeficiency virus-1 (HIV-1) and its ability to escape a myriad of host immune responses. In addition, conserved vulnerable regions on the HIV-1 envelope glycoprotein are often poorly immunogenic and elicit broadly neutralizing antibody responses (BNAbs) in a minority of HIV-1-infected individuals and only after several years of infection. All of the known BNAbs demonstrate high levels of somatic mutations and often display other unusual traits, such as a long heavy chain complementarity determining region 3 (CDRH3) and autoreactivity that can be limited by host tolerance controls. Nonetheless, the demonstration that HIV-1-infected individuals can make potent BNAbs is encouraging, and recent progress in isolating such antibodies and mapping their immune pathways of development is providing new strategies for vaccination.
Collapse
Affiliation(s)
- John R Mascola
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA.
| | | |
Collapse
|
483
|
Abstract
PURPOSE OF REVIEW In this review, examples of recent progress in HIV-1 vaccine research are discussed. RECENT FINDINGS New insights from the immune correlates analyses of the RV144 efficacy trial have accelerated vaccine development with leads to follow in nonhuman primate studies and improved vaccine designs. Several new vaccine vector approaches offer promise in the exquisite control of acute infection and in improving the breadth of T-cell responses. New targets of broadly neutralizing antibodies (BnAbs) have been elucidated, and improved understanding of how the human host controls BnAb development have emerged from BnAb knock-in mice and from analyses of BnAb maturation and virus evolution in individuals followed from the time of HIV-1 transmission to BnAb induction. SUMMARY Based on these observations, it is clear that the development of a successful HIV-1 vaccine will require new vaccine approaches and iterative testing of immunogens in well designed animal and human trials.
Collapse
Affiliation(s)
- Barton F Haynes
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC 27710, USA.
| | | |
Collapse
|
484
|
Ping LH, Joseph SB, Anderson JA, Abrahams MR, Salazar-Gonzalez JF, Kincer LP, Treurnicht FK, Arney L, Ojeda S, Zhang M, Keys J, Potter EL, Chu H, Moore P, Salazar MG, Iyer S, Jabara C, Kirchherr J, Mapanje C, Ngandu N, Seoighe C, Hoffman I, Gao F, Tang Y, Labranche C, Lee B, Saville A, Vermeulen M, Fiscus S, Morris L, Karim SA, Haynes BF, Shaw GM, Korber BT, Hahn BH, Cohen MS, Montefiori D, Williamson C, Swanstrom R. Comparison of viral Env proteins from acute and chronic infections with subtype C human immunodeficiency virus type 1 identifies differences in glycosylation and CCR5 utilization and suggests a new strategy for immunogen design. J Virol 2013; 87:7218-33. [PMID: 23616655 PMCID: PMC3700278 DOI: 10.1128/jvi.03577-12] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Accepted: 04/15/2013] [Indexed: 12/18/2022] Open
Abstract
Understanding human immunodeficiency virus type 1 (HIV-1) transmission is central to developing effective prevention strategies, including a vaccine. We compared phenotypic and genetic variation in HIV-1 env genes from subjects in acute/early infection and subjects with chronic infections in the context of subtype C heterosexual transmission. We found that the transmitted viruses all used CCR5 and required high levels of CD4 to infect target cells, suggesting selection for replication in T cells and not macrophages after transmission. In addition, the transmitted viruses were more likely to use a maraviroc-sensitive conformation of CCR5, perhaps identifying a feature of the target T cell. We confirmed an earlier observation that the transmitted viruses were, on average, modestly underglycosylated relative to the viruses from chronically infected subjects. This difference was most pronounced in comparing the viruses in acutely infected men to those in chronically infected women. These features of the transmitted virus point to selective pressures during the transmission event. We did not observe a consistent difference either in heterologous neutralization sensitivity or in sensitivity to soluble CD4 between the two groups, suggesting similar conformations between viruses from acute and chronic infection. However, the presence or absence of glycosylation sites had differential effects on neutralization sensitivity for different antibodies. We suggest that the occasional absence of glycosylation sites encoded in the conserved regions of env, further reduced in transmitted viruses, could expose specific surface structures on the protein as antibody targets.
Collapse
Affiliation(s)
- Li-Hua Ping
- UNC Center for AIDS Research, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Sarah B. Joseph
- UNC Center for AIDS Research, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Jeffrey A. Anderson
- UNC Center for AIDS Research, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Division of Infectious Diseases, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Melissa-Rose Abrahams
- Institute of Infectious Diseases and Molecular Medicine, Division of Medical Virology, University of Cape Town and National Health Laboratory Services, Cape Town, South Africa
| | | | - Laura P. Kincer
- UNC Center for AIDS Research, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Florette K. Treurnicht
- Institute of Infectious Diseases and Molecular Medicine, Division of Medical Virology, University of Cape Town and National Health Laboratory Services, Cape Town, South Africa
| | - Leslie Arney
- UNC Center for AIDS Research, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Suany Ojeda
- UNC Center for AIDS Research, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Ming Zhang
- Theoretical Biology, Los Alamos National Laboratory, Los Alamos, New Mexico, USA
- Department of Epidemiology and Biostatistics, University of Georgia, Athens, Georgia, USA
| | - Jessica Keys
- UNC Center for AIDS Research, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - E. Lake Potter
- UNC Center for AIDS Research, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Haitao Chu
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Penny Moore
- Centre for HIV and STIs, National Institute for Communicable Diseases, Johannesburg, South Africa
| | - Maria G. Salazar
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Shilpa Iyer
- Departments of Medicine and Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Cassandra Jabara
- UNC Center for AIDS Research, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Jennifer Kirchherr
- Duke Human Vaccine Institute, Department of Medicine, Duke University, Durham, North Carolina, USA
| | | | - Nobubelo Ngandu
- Institute of Infectious Diseases and Molecular Medicine, Division of Medical Virology, University of Cape Town and National Health Laboratory Services, Cape Town, South Africa
| | | | - Irving Hoffman
- Division of Infectious Diseases, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Feng Gao
- Duke Human Vaccine Institute, Department of Medicine, Duke University, Durham, North Carolina, USA
| | - Yuyang Tang
- UNC Center for AIDS Research, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Celia Labranche
- Department of Surgery, Duke University, Durham, North Carolina, USA
| | - Benhur Lee
- Department of Microbiology, Immunology and Molecular Genetics, University of California at Los Angeles, Los Angeles, California, USA
| | - Andrew Saville
- South African National Blood Service, Weltevreden Park, South Africa
| | - Marion Vermeulen
- South African National Blood Service, Weltevreden Park, South Africa
| | - Susan Fiscus
- UNC Center for AIDS Research, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Lynn Morris
- Centre for HIV and STIs, National Institute for Communicable Diseases, Johannesburg, South Africa
| | - Salim Abdool Karim
- Center for AIDS Program Research in South Africa, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Barton F. Haynes
- Duke Human Vaccine Institute, Department of Medicine, Duke University, Durham, North Carolina, USA
| | - George M. Shaw
- Departments of Medicine and Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Bette T. Korber
- Los Alamos National Laboratory, Los Alamos, New Mexico, USA
- Santa Fe Institute, Santa Fe, New Mexico, USA
| | - Beatrice H. Hahn
- Departments of Medicine and Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Myron S. Cohen
- UNC Center for AIDS Research, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Division of Infectious Diseases, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - David Montefiori
- Department of Surgery, Duke University, Durham, North Carolina, USA
| | - Carolyn Williamson
- Institute of Infectious Diseases and Molecular Medicine, Division of Medical Virology, University of Cape Town and National Health Laboratory Services, Cape Town, South Africa
| | - Ronald Swanstrom
- UNC Center for AIDS Research, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
485
|
Pancera M, Shahzad-Ul-Hussan S, Doria-Rose NA, McLellan JS, Bailer RT, Dai K, Loesgen S, Louder MK, Staupe RP, Yang Y, Zhang B, Parks R, Eudailey J, Lloyd KE, Blinn J, Alam SM, Haynes BF, Amin MN, Wang LX, Burton DR, Koff WC, Nabel GJ, Mascola JR, Bewley CA, Kwong PD. Structural basis for diverse N-glycan recognition by HIV-1-neutralizing V1-V2-directed antibody PG16. Nat Struct Mol Biol 2013; 20:804-13. [PMID: 23708607 PMCID: PMC4046252 DOI: 10.1038/nsmb.2600] [Citation(s) in RCA: 230] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Accepted: 05/02/2013] [Indexed: 11/08/2022]
Abstract
HIV-1 uses a diverse N-linked-glycan shield to evade recognition by antibody. Select human antibodies, such as the clonally related PG9 and PG16, recognize glycopeptide epitopes in the HIV-1 V1-V2 region and penetrate this shield, but their ability to accommodate diverse glycans is unclear. Here we report the structure of antibody PG16 bound to a scaffolded V1-V2, showing an epitope comprising both high mannose-type and complex-type N-linked glycans. We combined structure, NMR and mutagenesis analyses to characterize glycan recognition by PG9 and PG16. Three PG16-specific residues, arginine, serine and histidine (RSH), were critical for binding sialic acid on complex-type glycans, and introduction of these residues into PG9 produced a chimeric antibody with enhanced HIV-1 neutralization. Although HIV-1-glycan diversity facilitates evasion, antibody somatic diversity can overcome this and can provide clues to guide the design of modified antibodies with enhanced neutralization.
Collapse
Affiliation(s)
- Marie Pancera
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, US National Institutes of Health, Bethesda, Maryland, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
486
|
Synthetic glycopeptides reveal the glycan specificity of HIV-neutralizing antibodies. Nat Chem Biol 2013; 9:521-6. [PMID: 23831758 PMCID: PMC3730851 DOI: 10.1038/nchembio.1288] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2012] [Accepted: 05/30/2013] [Indexed: 12/31/2022]
Abstract
A new class of glycan-reactive HIV-neutralizing antibodies, including PG9 and PG16, has been recently discovered that appear to recognize novel glycopeptide epitopes on HIV-1 gp120. However, further characterization and reconstitution of the precise neutralizing epitopes are complicated by the heterogeneity of glycosylation. We report here the design, synthesis, and antigenic evaluation of novel cyclic V1V2 glycopeptides carrying defined N-linked glycans at the conserved glycosylation sites (N160 and N156/N173) derived from gp120 of two HIV-1 isolates. Antibody binding studies confirmed the necessity of a Man5GlcNAc2 glycan at N160 for recognition by PG9 and PG16, and further revealed a critical role of a sialylated N-glycan at the secondary site (N156/N173) in the context of glycopeptides for antibody binding. In addition to defining the glycan specificities of PG9 and PG16, the identified synthetic glycopeptides provide a valuable template for HIV-1 vaccine design.
Collapse
|
487
|
Detection and isolation of auto-reactive human antibodies from primary B cells. Methods 2013; 64:153-9. [PMID: 23811296 DOI: 10.1016/j.ymeth.2013.06.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Revised: 06/13/2013] [Accepted: 06/18/2013] [Indexed: 12/20/2022] Open
Abstract
The isolation of human monoclonal antibodies (hmAb) has emerged as a versatile platform in a wide variety of contexts ranging from vaccinology to therapeutics. In particular, the presence of high titers of circulating auto-antibodies is implicated in the pathology and outcome of autoimmune diseases. Therefore, the molecular characterization of these hmAb provides an avenue to understanding the pathogenesis of autoimmune diseases. Additionally, the phenotype of the auto-reactive B cells may have direct relevance for therapeutic intervention. In this report, we describe a high-throughput single-cell assay, microengraving, for the screening, characterization and isolation of anti-citrullinated protein antibodies (ACPA) from peripheral blood mononuclear cells (PBMC) of rheumatoid arthritis (RA) patients. Stimulated B cells are profiled at the single-cell level in a large array of sub-nanoliter nanowells (∼10(5)), assessing both the phenotype of the cells and their ability to secrete cyclic-citrullinated peptide (CCP)-specific antibodies. Single B cells secreting ACPA are retrieved by automated micromanipulation, and amplification of the immunoglobulin (Ig) heavy and light chains is performed prior to recombinant expression. The methodology offers a simple, rapid and low-cost platform for isolation of auto-reactive antibodies from low numbers of input cells and can be easily adapted for isolation and characterization of auto-reactive antibodies in other autoimmune diseases.
Collapse
|
488
|
Bontjer I, Melchers M, Tong T, van Montfort T, Eggink D, Montefiori D, Olson WC, Moore JP, Binley JM, Berkhout B, Sanders RW. Comparative Immunogenicity of Evolved V1V2-Deleted HIV-1 Envelope Glycoprotein Trimers. PLoS One 2013; 8:e67484. [PMID: 23840716 PMCID: PMC3694020 DOI: 10.1371/journal.pone.0067484] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Accepted: 05/16/2013] [Indexed: 12/16/2022] Open
Abstract
Despite almost 30 years of research, no effective vaccine has yet been developed against HIV-1. Probably such a vaccine would need to induce both an effective T cell and antibody response. Any vaccine component focused on inducing humoral immunity requires the HIV-1 envelope (Env) glycoprotein complex as it is the only viral protein exposed on the virion surface. HIV-1 has evolved several mechanisms to evade broadly reactive neutralizing antibodies. One such a mechanism involves variable loop domains, which are highly flexible structures that shield the underlying conserved epitopes. We hypothesized that removal of such loops would increase the exposure and immunogenicity of these conserved regions. Env variable loop deletion however often leads to protein misfolding and aggregation because hydrophobic patches becoming solvent accessible. We have therefore previously used virus evolution to acquire functional Env proteins lacking the V1V2 loop. We then expressed them in soluble (uncleaved) gp140 forms. Three mutants were found to perform optimally in terms of protein expression, stability, trimerization and folding. In this study, we characterized the immune responses to these antigens in rabbits. The V1V2 deletion mutant ΔV1V2.9.VK induced a prominent response directed to epitopes that are not fully available on the other Env proteins tested but that effectively bound and neutralized the ΔV1V2 Env virus. This Env variant also induced more efficient neutralization of the tier 1 virus SF162. The immune refocusing effect was lost after booster immunization with a full-length gp140 protein with intact V1V2 loops. Collectively, this result suggests that deletion of variable domains could alter the specificity of the humoral immune response, but did not result in broad neutralization of neutralization-resistant virus isolates.
Collapse
Affiliation(s)
- Ilja Bontjer
- Department of Medical Microbiology, Laboratory of Experimental Virology, Center for Infection and Immunity Amsterdam (CINIMA), Academic Medical Center, Amsterdam, The Netherlands
| | - Mark Melchers
- Department of Medical Microbiology, Laboratory of Experimental Virology, Center for Infection and Immunity Amsterdam (CINIMA), Academic Medical Center, Amsterdam, The Netherlands
| | - Tommy Tong
- Torrey Pines Institute for Molecular Studies, San Diego, California, United States of America
| | - Thijs van Montfort
- Department of Medical Microbiology, Laboratory of Experimental Virology, Center for Infection and Immunity Amsterdam (CINIMA), Academic Medical Center, Amsterdam, The Netherlands
| | - Dirk Eggink
- Department of Medical Microbiology, Laboratory of Experimental Virology, Center for Infection and Immunity Amsterdam (CINIMA), Academic Medical Center, Amsterdam, The Netherlands
| | - David Montefiori
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, United States of America
| | - William C. Olson
- Progenics Pharmaceuticals, Tarrytown, New York, United States of America
| | - John P. Moore
- Department of Microbiology and Immunology, Weill Medical College of Cornell University, New York, New York, United States of America
| | - James M. Binley
- Torrey Pines Institute for Molecular Studies, San Diego, California, United States of America
| | - Ben Berkhout
- Department of Medical Microbiology, Laboratory of Experimental Virology, Center for Infection and Immunity Amsterdam (CINIMA), Academic Medical Center, Amsterdam, The Netherlands
| | - Rogier W. Sanders
- Department of Medical Microbiology, Laboratory of Experimental Virology, Center for Infection and Immunity Amsterdam (CINIMA), Academic Medical Center, Amsterdam, The Netherlands
- Department of Microbiology and Immunology, Weill Medical College of Cornell University, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
489
|
Kulp DW, Schief WR. Advances in structure-based vaccine design. Curr Opin Virol 2013; 3:322-31. [PMID: 23806515 DOI: 10.1016/j.coviro.2013.05.010] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Revised: 05/13/2013] [Accepted: 05/13/2013] [Indexed: 01/02/2023]
Abstract
Despite the tremendous successes of current vaccines, infectious diseases still take a heavy toll on the global population, and that provides strong rationale for broadening our vaccine development repertoire. Structural vaccinology, in which protein structure information is utilized to design immunogens, has promise to provide new vaccines against traditionally difficult targets. Crystal structures of antigens containing one or more protection epitopes, especially when in complex with a protective antibody, are the launching point for immunogen design. Integrating structure and sequence information for families of broadly neutralizing antibodies (bNAbs) has recently enabled the creation of germline-targeting immunogens that bind and activate germline B-cells in order to initiate the elicitation of such antibodies. The contacts between antigen and neutralizing antibody define a structural epitope, and methods have been developed to transplant epitopes to scaffold proteins for structural stabilization, and to design minimized antigens that retain one or more key epitopes while eliminating other potentially distracting or unnecessary features. To develop vaccines that protect against antigenically variable pathogens, pioneering structure-based work demonstrated that multiple strain-specific epitopes could be engineered onto a single immunogen. We review these recent structural vaccinology efforts to engineer germline-targeting, epitope-specific, and/or broad coverage immunogens.
Collapse
Affiliation(s)
- Daniel W Kulp
- IAVI Neutralizing Antibody Center and Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | |
Collapse
|
490
|
Isik G, Chung NPY, van Montfort T, Menis S, Matthews K, Schief WR, Moore JP, Sanders RW. An HIV-1 envelope glycoprotein trimer with an embedded IL-21 domain activates human B cells. PLoS One 2013; 8:e67309. [PMID: 23826263 PMCID: PMC3691133 DOI: 10.1371/journal.pone.0067309] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Accepted: 05/16/2013] [Indexed: 12/17/2022] Open
Abstract
Broadly neutralizing antibodies (bNAbs) that target the HIV-1 envelope glycoproteins (Env) can prevent virus acquisition, but several Env properties limit its ability to induce an antibody response that is of sufficient quantity and quality. The immunogenicity of Env can be increased by fusion to co-stimulatory molecules and here we describe novel soluble Env trimers with embedded interleukin-4 (IL-4) or interleukin-21 (IL-21) domains, designed to activate B cells that recognize Env. In particular, the chimeric EnvIL-21 molecule activated B cells efficiently and induced the differentiation of antibody secreting plasmablast-like cells. We studied whether we could increase the activity of the embedded IL-21 by designing a chimeric IL-21/IL-4 (ChimIL-21/4) molecule and by introducing amino acid substitutions in the receptor binding domain of IL-21 that were predicted to enhance its binding. In addition, we incorporated IL-21 into a cleavable Env trimer and found that insertion of IL-21 did not impair Env cleavage, while Env cleavage did not impair IL-21 activity. These studies should guide the further design of chimeric proteins and EnvIL-21 may prove useful in improving antibody responses against HIV-1.
Collapse
Affiliation(s)
- Gözde Isik
- Laboratory of Experimental Virology, Department of Medical Microbiology Center for Infection and Immunity Amsterdam, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Nancy P. Y. Chung
- Department of Microbiology and Immunology, Weill Medical College of Cornell University, New York, New York, United States of America
| | - Thijs van Montfort
- Laboratory of Experimental Virology, Department of Medical Microbiology Center for Infection and Immunity Amsterdam, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Sergey Menis
- Department of Biochemistry, University of Washington, Seattle, Washington, United States of America
- IAVI Neutralizing Antibody Center and Department of Immunology and Microbial Sciences, The Scripps Research Institute, San Diego, California, United States of America
| | - Katie Matthews
- Department of Microbiology and Immunology, Weill Medical College of Cornell University, New York, New York, United States of America
| | - William R. Schief
- Department of Immunology and Microbial Science, The Scripps Research Institute, San Diego, California, United States of America
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, San Diego, California, United States of America
- Scripps Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute, San Diego, California, United States of America
| | - John P. Moore
- Department of Microbiology and Immunology, Weill Medical College of Cornell University, New York, New York, United States of America
| | - Rogier W. Sanders
- Laboratory of Experimental Virology, Department of Medical Microbiology Center for Infection and Immunity Amsterdam, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Department of Microbiology and Immunology, Weill Medical College of Cornell University, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
491
|
Jennifer M, Leslie G, Maxwel MO, Ruth N, Julie O. HIV-1 maternal and infant variants show similar sensitivity to broadly neutralizing antibodies, but sensitivity varies by subtype. AIDS 2013; 27:1535-44. [PMID: 23856624 PMCID: PMC4080909 DOI: 10.1097/qad.0b013e32835faba5] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
RATIONALE To protect against HIV infection, passively transferred and/or vaccine-elicited neutralizing antibodies (NAbs) need to effectively target diverse subtypes that are transmitted globally. These variants are a limited subset of those present during chronic infection and display some unique features. In the case of mother-to-child transmission (MTCT), transmitted variants tend to be resistant to neutralization by maternal autologous NAbs. METHOD To investigate whether variants transmitted during MTCT are generally resistant to HIV-1-specific NAbs, 107 maternal or infant variants representing the dominant HIV-1 subtypes were tested against six recently identified HIV-1-specific broadly neutralizing monoclonal antibodies (bNAbs), NIH45-46W, VRC01, PGT128, PGT121, PG9 and PGT145. RESULTS Infant and maternal variants did not differ in their neutralization sensitivity to individual bNAbs, nor did viruses from transmitting versus nontransmitting mothers, although there was a trend for viruses from transmitting mothers to be less sensitive overall. No single bNAb neutralized all viruses, but a combination of bNAbs that target distinct epitopes covered 100% of the variants tested. Compared with heterosexually transmitted variants, vertically transmitted variants were significantly more sensitive to neutralization by PGT128 and PGT121 (P=0.03 in both cases), but there were no differences for the other bNAbs. Overall, subtype A variants were significantly more sensitive to NIH45-46 (P=0.04), VRC01 (P=0.002) and PGT145 (P=0.03) compared with the nonsubtype A and less sensitive to PGT121 than subtype Cs (P=0.0001). CONCLUSION A combination of bNAbs against distinct epitopes may be needed to provide maximum coverage against viruses in different modes of transmission and diverse subtypes.
Collapse
Affiliation(s)
- Mabuka Jennifer
- Division of Human Biology and Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, US 98109
- Program of Pathobiology, Department of Global Health, University of Washington, Seattle, WA
| | - Goo Leslie
- Division of Human Biology and Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, US 98109
- Program of Pathobiology, Department of Global Health, University of Washington, Seattle, WA
| | - Majiwa O. Maxwel
- Division of Human Biology and Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, US 98109
- Program of Pathobiology, Department of Global Health, University of Washington, Seattle, WA
| | - Nduati Ruth
- Department of Pediatrics, University of Nairobi, Nairobi, Kenya
| | - Overbaugh Julie
- Division of Human Biology and Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, US 98109
| |
Collapse
|
492
|
Sampath S, Carrico C, Janes J, Gurumoorthy S, Gibson C, Melcher M, Chitnis CE, Wang R, Schief WR, Smith JD. Glycan masking of Plasmodium vivax Duffy Binding Protein for probing protein binding function and vaccine development. PLoS Pathog 2013; 9:e1003420. [PMID: 23853575 PMCID: PMC3681752 DOI: 10.1371/journal.ppat.1003420] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Accepted: 04/30/2013] [Indexed: 11/19/2022] Open
Abstract
Glycan masking is an emerging vaccine design strategy to focus antibody responses to specific epitopes, but it has mostly been evaluated on the already heavily glycosylated HIV gp120 envelope glycoprotein. Here this approach was used to investigate the binding interaction of Plasmodium vivax Duffy Binding Protein (PvDBP) and the Duffy Antigen Receptor for Chemokines (DARC) and to evaluate if glycan-masked PvDBPII immunogens would focus the antibody response on key interaction surfaces. Four variants of PVDBPII were generated and probed for function and immunogenicity. Whereas two PvDBPII glycosylation variants with increased glycan surface coverage distant from predicted interaction sites had equivalent binding activity to wild-type protein, one of them elicited slightly better DARC-binding-inhibitory activity than wild-type immunogen. Conversely, the addition of an N-glycosylation site adjacent to a predicted PvDBP interaction site both abolished its interaction with DARC and resulted in weaker inhibitory antibody responses. PvDBP is composed of three subdomains and is thought to function as a dimer; a meta-analysis of published PvDBP mutants and the new DBPII glycosylation variants indicates that critical DARC binding residues are concentrated at the dimer interface and along a relatively flat surface spanning portions of two subdomains. Our findings suggest that DARC-binding-inhibitory antibody epitope(s) lie close to the predicted DARC interaction site, and that addition of N-glycan sites distant from this site may augment inhibitory antibodies. Thus, glycan resurfacing is an attractive and feasible tool to investigate protein structure-function, and glycan-masked PvDBPII immunogens might contribute to P. vivax vaccine development. An important goal of many vaccine efforts is to inhibit pathogen invasion of host cells, but few approaches exist to target vaccine antibodies on invasion blocking epitopes. Glycan masking is a vaccine design strategy to hide protein surfaces with carbohydrates and focus antibodies on exposed surfaces. This approach has mostly been evaluated on the heavily glycosylated HIV envelope glycoprotein, but it has never been tested on eukaryotic pathogens, such as Plasmodium, which have limited N-glycosylation machinery and therefore may provide a better platform to explore this strategy. Here, we used glycan masking to investigate the binding interaction between Plasmodium vivax Duffy binding protein (PvDBP) and the Duffy Antigen Receptor for Chemokines (DARC). This study showed that addition of an N-glycan site in a predicted host interaction surface abolished binding and potentially covered up an inhibitory antibody epitope. In contrast, addition of multiple N-glycan sites distant from predicted interaction surfaces did not inhibit binding but did slightly enhance elicitation of inhibitory antibodies. This analysis shows that glycan resurfacing offers an integrated approach to characterize protein function and immunogenicity and that glycan resurfacing of PvDBPII immunogens may have utility in P. vivax-malaria vaccine development.
Collapse
Affiliation(s)
- Sowmya Sampath
- Seattle Biomedical Research Institute, Seattle, Washington, United States of America
| | - Chris Carrico
- Department of Biochemistry, University of Washington, Seattle, Washington, United States of America
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Joel Janes
- Seattle Biomedical Research Institute, Seattle, Washington, United States of America
| | - Sairam Gurumoorthy
- Seattle Biomedical Research Institute, Seattle, Washington, United States of America
| | - Claire Gibson
- Seattle Biomedical Research Institute, Seattle, Washington, United States of America
| | - Martin Melcher
- Seattle Biomedical Research Institute, Seattle, Washington, United States of America
| | - Chetan E. Chitnis
- International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Ruobing Wang
- Seattle Biomedical Research Institute, Seattle, Washington, United States of America
| | - William R. Schief
- Department of Biochemistry, University of Washington, Seattle, Washington, United States of America
- Department of Immunology and Microbial Science and IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, California, United States of America
- Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute, La Jolla, California, United States of America
- * E-mail: (WRS); (JDS)
| | - Joseph D. Smith
- Seattle Biomedical Research Institute, Seattle, Washington, United States of America
- Department of Pathobiology, University of Washington, Seattle, Washington, United States of America
- * E-mail: (WRS); (JDS)
| |
Collapse
|
493
|
The griffithsin dimer is required for high-potency inhibition of HIV-1: evidence for manipulation of the structure of gp120 as part of the griffithsin dimer mechanism. Antimicrob Agents Chemother 2013; 57:3976-89. [PMID: 23752505 DOI: 10.1128/aac.00332-13] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Griffithsin (Grft) is a protein lectin derived from red algae that tightly binds the HIV envelope protein gp120 and effectively inhibits virus infection. This inhibition is due to the binding by Grft of high-mannose saccharides on the surface of gp120. Grft has been shown to be a tight dimer, but the role of the dimer in Grft's anti-HIV function has not been fully explored. To investigate the role of the Grft dimer in anti-HIV function, an obligate dimer of Grft was designed by expressing the protein with a peptide linker between the two subunits. This "Grft-linker-Grft" is a folded protein dimer, apparently nearly identical in structural properties to the wild-type protein. A "one-armed" obligate dimer was also designed (Grft-linker-Grft OneArm), with each of the three carbohydrate binding sites of one subunit mutated while the other subunit remained intact. While both constructed dimers retained the ability to bind gp120 and the viral surface, Grft-linker-Grft OneArm was 84- to 1,010-fold less able to inhibit HIV than wild-type Grft, while Grft-linker-Grft had near-wild-type antiviral potency. Furthermore, while the wild-type protein demonstrated the ability to alter the structure of gp120 by exposing the CD4 binding site, Grft-linker-Grft OneArm largely lost this ability. In experiments to investigate gp120 shedding, it was found that Grft has different effects on gp120 shedding for strains from subtype B and subtype C, and this might correlate with Grft function. Evidence is provided that the dimer form of Grft is critical to the function of this protein in HIV inhibition.
Collapse
|
494
|
Bernardi A, Jiménez-Barbero J, Casnati A, De Castro C, Darbre T, Fieschi F, Finne J, Funken H, Jaeger KE, Lahmann M, Lindhorst TK, Marradi M, Messner P, Molinaro A, Murphy PV, Nativi C, Oscarson S, Penadés S, Peri F, Pieters RJ, Renaudet O, Reymond JL, Richichi B, Rojo J, Sansone F, Schäffer C, Turnbull WB, Velasco-Torrijos T, Vidal S, Vincent S, Wennekes T, Zuilhof H, Imberty A. Multivalent glycoconjugates as anti-pathogenic agents. Chem Soc Rev 2013; 42:4709-27. [PMID: 23254759 PMCID: PMC4399576 DOI: 10.1039/c2cs35408j] [Citation(s) in RCA: 429] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Multivalency plays a major role in biological processes and particularly in the relationship between pathogenic microorganisms and their host that involves protein-glycan recognition. These interactions occur during the first steps of infection, for specific recognition between host and bacteria, but also at different stages of the immune response. The search for high-affinity ligands for studying such interactions involves the combination of carbohydrate head groups with different scaffolds and linkers generating multivalent glycocompounds with controlled spatial and topology parameters. By interfering with pathogen adhesion, such glycocompounds including glycopolymers, glycoclusters, glycodendrimers and glyconanoparticles have the potential to improve or replace antibiotic treatments that are now subverted by resistance. Multivalent glycoconjugates have also been used for stimulating the innate and adaptive immune systems, for example with carbohydrate-based vaccines. Bacteria present on their surfaces natural multivalent glycoconjugates such as lipopolysaccharides and S-layers that can also be exploited or targeted in anti-infectious strategies.
Collapse
Affiliation(s)
- Anna Bernardi
- Università di Milano, Dipartimento di Chimica Organica e Industriale and Centro di Eccellenza CISI, via Venezian 21, 20133 Milano, Italy
| | | | - Alessandro Casnati
- Università degli Studi di Parma, Dipartimento di Chimica, Parco Area delle Scienze 17/a, 43100 Parma, Italy
| | - Cristina De Castro
- Department of Chemical Sciences, Università di Napoli Federico II, Complesso Universitario Monte Santangelo, Via Cintia 4, I-80126 Napoli, Italy
| | - Tamis Darbre
- Department of Chemistry and Biochemistry, University of Berne, Freiestrasse 3, CH-3012, Berne, Switzerland
| | - Franck Fieschi
- Institut de Biologie Structurale, 41 rue Jules Horowitz, 38027 Grenoble Cedex 1, France
| | - Jukka Finne
- Department of Biosciences, University of Helsinki, P. O. Box 56, FI-00014 Helsinki, Finland
| | - Horst Funken
- Institute of Molecular Enzyme Technology, Heinrich-Heine-University Düsseldorf, Forschungszentrum Jülich, D-42425 Jülich, Germany
| | - Karl-Erich Jaeger
- Institute of Molecular Enzyme Technology, Heinrich-Heine-University Düsseldorf, Forschungszentrum Jülich, D-42425 Jülich, Germany
| | - Martina Lahmann
- School of Chemistry, Bangor University, Deiniol Road Bangor, Gwynedd LL57 2UW, UK
| | - Thisbe K. Lindhorst
- Otto Diels Institute of Organic Chemistry, Christiana Albertina University of Kiel, Otto-Hahn-Platz 3-4, D-24098 Kiel, Germany
| | - Marco Marradi
- Laboratory of GlycoNanotechnology, CIC biomaGUNE and CIBER-BBN, P1 de Miramón 182, 20009 San Sebastián, Spain
| | - Paul Messner
- Department of NanoBiotechnology, NanoGlycobiology Unit, University of Natural Resources and Life Sciences, Muthgasse 11, A-1190 Vienna, Austria
| | - Antonio Molinaro
- Department of Chemical Sciences, Università di Napoli Federico II, Complesso Universitario Monte Santangelo, Via Cintia 4, I-80126 Napoli, Italy
| | - Paul V. Murphy
- School of Chemistry, National University of Ireland, Galway, University Road, Galway, Ireland
| | - Cristina Nativi
- Dipartimento di Chimica, Universitá degli Studi di Firenze, Via della Lastruccia, 13, I-50019 Sesto Fiorentino – Firenze, Italy
| | - Stefan Oscarson
- Centre for Synthesis and Chemical Biology, UCD School of Chemistry and Chemical Biology, University College Dublin, Belfield, Dublin 4, Ireland
| | - Soledad Penadés
- Laboratory of GlycoNanotechnology, CIC biomaGUNE and CIBER-BBN, P1 de Miramón 182, 20009 San Sebastián, Spain
| | - Francesco Peri
- Organic and Medicinal Chemistry, University of Milano-Bicocca, Piazza della Scienza, 2, 20126 Milano, Italy
| | - Roland J. Pieters
- Department of Medicinal Chemistry and Chemical Biology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, P.O. Box 80082, 3508 TB Utrecht, The Netherlands
| | - Olivier Renaudet
- Département de Chimie Moléculaire, UMR-CNRS 5250 & ICMG FR 2607, Université Joseph Fourier, BP53, 38041 Grenoble Cedex 9, France
| | - Jean-Louis Reymond
- Department of Chemistry and Biochemistry, University of Berne, Freiestrasse 3, CH-3012, Berne, Switzerland
| | - Barbara Richichi
- Dipartimento di Chimica, Universitá degli Studi di Firenze, Via della Lastruccia, 13, I-50019 Sesto Fiorentino – Firenze, Italy
| | - Javier Rojo
- Glycosystems Laboratory, Instituto de Investigaciones Químicas, CSIC – Universidad de Sevilla, Av. Américo Vespucio, 49, Seville 41092, Spain
| | - Francesco Sansone
- Università degli Studi di Parma, Dipartimento di Chimica, Parco Area delle Scienze 17/a, 43100 Parma, Italy
| | - Christina Schäffer
- Department of NanoBiotechnology, NanoGlycobiology Unit, University of Natural Resources and Life Sciences, Muthgasse 11, A-1190 Vienna, Austria
| | - W. Bruce Turnbull
- School of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | | | - Sébastien Vidal
- Institut de Chimie et Biochimie Moléculaires et Supramoléculaires UMR 5246, CNRS, Université Claude Bernard Lyon 1, 43 Boulevard du 11 Novembre 1918, F-69622 Villeurbanne, France
| | - Stéphane Vincent
- University of Namur (FUNDP), Département de Chimie, Laboratoire de Chimie Bio-Organique, rue de Bruxelles 61, B-5000 Namur, Belgium
| | - Tom Wennekes
- Laboratory of Organic Chemistry, Wageningen University, Dreijenplein 8, 6703 HB Wageningen, The Netherlands
| | - Han Zuilhof
- Laboratory of Organic Chemistry, Wageningen University, Dreijenplein 8, 6703 HB Wageningen, The Netherlands
- Department of Chemical and Materials Engineering, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Anne Imberty
- Centre de Recherche sur les Macromolécules Végétales (CERMAV – CNRS), affiliated with Grenoble-Université and ICMG, F-38041 Grenoble, France
| |
Collapse
|
495
|
Emergence of gp120 V3 variants confers neutralization resistance in an R5 simian-human immunodeficiency virus-infected macaque elite neutralizer that targets the N332 glycan of the human immunodeficiency virus type 1 envelope glycoprotein. J Virol 2013; 87:8798-804. [PMID: 23720719 DOI: 10.1128/jvi.00878-13] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Neutralization-resistant simian-human immunodeficiency virus AD8 (SHIVAD8) variants that emerged in an infected macaque elite neutralizer targeting the human immunodeficiency virus type 1 (HIV-1) gp120 N332 glycan acquired substitutions of critical amino acids in the V3 region rather than losing the N332 glycosylation site. One of these resistant variants, carrying the full complement of gp120 V3 changes, was also resistant to the potent anti-HIV-1 monoclonal neutralizing antibodies PGT121 and 10-1074, both of which are also dependent on the presence of the gp120 N332 glycan.
Collapse
|
496
|
Kong L, Lee JH, Doores KJ, Murin CD, Julien JP, McBride R, Liu Y, Marozsan A, Cupo A, Klasse PJ, Hoffenberg S, Caulfield M, King CR, Hua Y, Le KM, Khayat R, Deller MC, Clayton T, Tien H, Feizi T, Sanders RW, Paulson JC, Moore JP, Stanfield RL, Burton DR, Ward AB, Wilson IA. Supersite of immune vulnerability on the glycosylated face of HIV-1 envelope glycoprotein gp120. Nat Struct Mol Biol 2013; 20:796-803. [PMID: 23708606 PMCID: PMC3823233 DOI: 10.1038/nsmb.2594] [Citation(s) in RCA: 279] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Accepted: 04/05/2013] [Indexed: 12/17/2022]
Abstract
A substantial proportion of the broadly neutralizing antibodies (bnAbs) identified in certain HIV-infected donors recognize glycan-dependent epitopes on HIV-1 gp120. Here we elucidate how the bnAb PGT 135 binds its Asn332 glycan-dependent epitope from its 3.1-Å crystal structure with gp120, CD4 and Fab 17b. PGT 135 interacts with glycans at Asn332, Asn392 and Asn386, using long CDR loops H1 and H3 to penetrate the glycan shield and access the gp120 protein surface. EM reveals that PGT 135 can accommodate the conformational and chemical diversity of gp120 glycans by altering its angle of engagement. Combined structural studies of PGT 135, PGT 128 and 2G12 show that this Asn332-dependent antigenic region is highly accessible and much more extensive than initially appreciated, which allows for multiple binding modes and varied angles of approach; thereby it represents a supersite of vulnerability for antibody neutralization.
Collapse
Affiliation(s)
- Leopold Kong
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
497
|
A brief history of the global effort to develop a preventive HIV vaccine. Vaccine 2013; 31:3502-18. [PMID: 23707164 DOI: 10.1016/j.vaccine.2013.05.018] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2013] [Revised: 05/01/2013] [Accepted: 05/07/2013] [Indexed: 01/09/2023]
Abstract
Soon after HIV was discovered as the cause of AIDS in 1983-1984, there was an expectation that a preventive vaccine would be rapidly developed. In trying to achieve that goal, three successive scientific paradigms have been explored: induction of neutralizing antibodies, induction of cell mediated immunity, and exploration of combination approaches and novel concepts. Although major progress has been made in understanding the scientific basis for HIV vaccine development, efficacy trials have been critical in moving the field forward. In 2009, the field was reinvigorated with the modest results obtained from the RV144 trial conducted in Thailand. Here, we review those vaccine development efforts, with an emphasis on events that occurred during the earlier years. The goal is to provide younger generations of scientists with information and inspiration to continue the search for an HIV vaccine.
Collapse
|
498
|
A mechanistic understanding of allosteric immune escape pathways in the HIV-1 envelope glycoprotein. PLoS Comput Biol 2013; 9:e1003046. [PMID: 23696718 PMCID: PMC3656115 DOI: 10.1371/journal.pcbi.1003046] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Accepted: 03/15/2013] [Indexed: 11/19/2022] Open
Abstract
The HIV-1 envelope (Env) spike, which consists of a compact, heterodimeric trimer of the glycoproteins gp120 and gp41, is the target of neutralizing antibodies. However, the high mutation rate of HIV-1 and plasticity of Env facilitates viral evasion from neutralizing antibodies through various mechanisms. Mutations that are distant from the antibody binding site can lead to escape, probably by changing the conformation or dynamics of Env; however, these changes are difficult to identify and define mechanistically. Here we describe a network analysis-based approach to identify potential allosteric immune evasion mechanisms using three known HIV-1 Env gp120 protein structures from two different clades, B and C. First, correlation and principal component analyses of molecular dynamics (MD) simulations identified a high degree of long-distance coupled motions that exist between functionally distant regions within the intrinsic dynamics of the gp120 core, supporting the presence of long-distance communication in the protein. Then, by integrating MD simulations with network theory, we identified the optimal and suboptimal communication pathways and modules within the gp120 core. The results unveil both strain-dependent and -independent characteristics of the communication pathways in gp120. We show that within the context of three structurally homologous gp120 cores, the optimal pathway for communication is sequence sensitive, i.e. a suboptimal pathway in one strain becomes the optimal pathway in another strain. Yet the identification of conserved elements within these communication pathways, termed inter-modular hotspots, could present a new opportunity for immunogen design, as this could be an additional mechanism that HIV-1 uses to shield vulnerable antibody targets in Env that induce neutralizing antibody breadth.
Collapse
|
499
|
Sato S, Inokuma T, Otsubo N, Burton DR, Barbas CF. Chemically Programmed Antibodies AS HIV-1 Attachment Inhibitors. ACS Med Chem Lett 2013; 4:460-465. [PMID: 23750312 DOI: 10.1021/ml400097z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Herein we describe the design and application of two small-molecule anti-HIV compounds for the creation of chemically programmed antibodies. N-acyl-β-lactam derivatives of two previously described molecules BMS-378806 and BMS-488043 that inhibit the interaction between HIV-1 gp120 and T-cells were synthesized and used to program the binding activity of aldolase antibody 38C2. Discovery of a successful linkage site to BMS-488043 allowed for the synthesis of chemically programmed antibodies with affinity for HIV-1 gp120 and potent HIV-1 neutralization activity. Derivation of a successful conjugation strategy for this family of HIV-1 entry inhibitors enables its application in chemically programmed antibodies and vaccines and may facilitate the development of novel bispecific antibodies and topical microbicides.
Collapse
Affiliation(s)
- Shinichi Sato
- Department
of Molecular Biology and Chemistry and the
Skaggs Institute for Chemical Biology and Department of Immunology
and Microbial Science, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United
States
| | - Tsubasa Inokuma
- Department
of Molecular Biology and Chemistry and the
Skaggs Institute for Chemical Biology and Department of Immunology
and Microbial Science, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United
States
| | - Nobumasa Otsubo
- Department
of Molecular Biology and Chemistry and the
Skaggs Institute for Chemical Biology and Department of Immunology
and Microbial Science, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United
States
| | - Dennis R. Burton
- Department
of Molecular Biology and Chemistry and the
Skaggs Institute for Chemical Biology and Department of Immunology
and Microbial Science, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United
States
| | - Carlos F. Barbas
- Department
of Molecular Biology and Chemistry and the
Skaggs Institute for Chemical Biology and Department of Immunology
and Microbial Science, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United
States
| |
Collapse
|
500
|
Georgiev IS, Doria-Rose NA, Zhou T, Do Kwon Y, Staupe RP, Moquin S, Chuang GY, Louder MK, Schmidt SD, Altae-Tran HR, Bailer RT, McKee K, Nason M, O'Dell S, Ofek G, Pancera M, Srivatsan S, Shapiro L, Connors M, Migueles SA, Morris L, Nishimura Y, Martin MA, Mascola JR, Kwong PD. Delineating Antibody Recognition in Polyclonal Sera from Patterns of HIV-1 Isolate Neutralization. Science 2013; 340:751-6. [DOI: 10.1126/science.1233989] [Citation(s) in RCA: 181] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|