151
|
Vani J, Kaveri SV, Bayry J. Myeloid dendritic cell dysfunction during primary HIV-1 infection is independent of interaction with gp120. J Infect Dis 2012; 205:1893-5. [PMID: 22492849 DOI: 10.1093/infdis/jis292] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
152
|
Zabihollahi R, Sadat SM, Vahabpour R, Salehi M, Azadmanesh K, Siadat SD, Azizi Saraji AR, Pouriavali MH, Momen SB, Aghasadeghi MR. Introducing a frameshift mutation to the Pol sequence of HIV-1 provirus and evaluation of the immunogenic characteristics of the mutated virions (RINNL4-3). Mol Biol 2012. [DOI: 10.1134/s0026893312030107] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
153
|
Nakamura GR, Fonseca DPAJ, O'Rourke SM, Vollrath AL, Berman PW. Monoclonal antibodies to the V2 domain of MN-rgp120: fine mapping of epitopes and inhibition of α4β7 binding. PLoS One 2012; 7:e39045. [PMID: 22720026 PMCID: PMC3374778 DOI: 10.1371/journal.pone.0039045] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2011] [Accepted: 05/17/2012] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Recombinant gp120 (MN-rgp120) was a major component of the AIDSVAX B/E vaccine used in the RV144 trial. This was the first clinical trial to show that vaccination could prevent HIV infection in humans. A recent RV144 correlates of protection study found that protection correlated with the presence of antibodies to the V2 domain. It has been proposed that antibodies to the α4β7 binding site in the V2 domain might prevent HIV-1 infection by blocking the ability of virions to recognize α4β7 on activated T-cells. In this study we investigated the specificity of monoclonal antibodies (MAbs) to the V2 domain of MN-rgp120 and examined the possibility that these antibodies could inhibit the binding of MN-rgp120 to the α4β7 integrin. METHODOLOGY/PRINCIPAL FINDINGS Nine MAbs to the V2 domain were isolated from mice immunized with recombinant envelope proteins. The ability of these MAbs to inhibit HIV infection, block the binding of gp120 to CD4, and block the binding of MN-rgp120 to the α4β7 integrin was measured. Mutational analysis showed that eight of the MAbs recognized two immunodominant clusters of amino acids (166-168 and 178-183) located at either end of the C strand within the four-strand anti-parallel sheet structure comprising the V1/V2 domain. CONCLUSIONS/SIGNIFICANCE These studies showed that the antigenic structure of the V2 domain is exceedingly complex and that MAbs isolated from mice immunized with MN-rgp120 exhibited a high level of strain specificity compared to MAbs to the V2 domain isolated from HIV-infected humans. We found that immunization with MN-rgp120 readily elicits antibodies to the V2 domain and some of these were able to block the binding of MN-rgp120 to the α4β7 integrin.
Collapse
Affiliation(s)
- Gerald R. Nakamura
- Antibody Engineering Department, Genentech, Incorporated, South San Francisco, California, United States of America
| | - Dora P. A. J. Fonseca
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, California, United States of America
| | - Sara M. O'Rourke
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, California, United States of America
| | - Aaron L. Vollrath
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, California, United States of America
| | - Phillip W. Berman
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, California, United States of America
| |
Collapse
|
154
|
Depetris RS, Julien JP, Khayat R, Lee JH, Pejchal R, Katpally U, Cocco N, Kachare M, Massi E, David KB, Cupo A, Marozsan AJ, Olson WC, Ward AB, Wilson IA, Sanders RW, Moore JP. Partial enzymatic deglycosylation preserves the structure of cleaved recombinant HIV-1 envelope glycoprotein trimers. J Biol Chem 2012; 287:24239-54. [PMID: 22645128 DOI: 10.1074/jbc.m112.371898] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The trimeric envelope glycoprotein complex (Env) is the focus of vaccine development programs aimed at generating protective humoral responses to human immunodeficiency virus type 1 (HIV-1). N-Linked glycans, which constitute almost half of the molecular mass of the external Env domains, produce considerable structural heterogeneity and are a major impediment to crystallization studies. Moreover, by shielding the peptide backbone, glycans can block attempts to generate neutralizing antibodies against a substantial subset of potential epitopes when Env proteins are used as immunogens. Here, we describe the partial deglycosylation of soluble, cleaved recombinant Env trimers by inhibition of the synthesis of complex N-glycans during Env production, followed by treatment with glycosidases under conditions that preserve Env trimer integrity. The partially deglycosylated trimers are stable, and neither abnormally sensitive to proteolytic digestion nor prone to aggregation. Moreover, the deglycosylated trimers retain or increase their ability to bind CD4 and antibodies that are directed to conformational epitopes, including the CD4-binding site and the V3 region. However, as expected, they do not react with glycan-dependent antibodies 2G12 and PGT123, or the C-type lectin receptor DC-SIGN. Electron microscopic analysis shows that partially deglycosylated trimers have a structure similar to fully glycosylated trimers, indicating that removal of glycans does not substantially perturb the structural integrity of the trimer. The glycan-depleted Env trimers should be useful for structural and immunogenicity studies.
Collapse
Affiliation(s)
- Rafael S Depetris
- Department of Microbiology and Immunology, Weill Medical College of Cornell University, New York, New York 10065, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
155
|
McCoy LE, Quigley AF, Strokappe NM, Bulmer-Thomas B, Seaman MS, Mortier D, Rutten L, Chander N, Edwards CJ, Ketteler R, Davis D, Verrips T, Weiss RA. Potent and broad neutralization of HIV-1 by a llama antibody elicited by immunization. ACTA ACUST UNITED AC 2012; 209:1091-103. [PMID: 22641382 PMCID: PMC3371729 DOI: 10.1084/jem.20112655] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
A heavy chain–only antibody isolated from a llama repeatedly immunized with trimeric HIV-1 Env neutralizes 96% of tested HIV-1 strains. Llamas (Lama glama) naturally produce heavy chain–only antibodies (Abs) in addition to conventional Abs. The variable regions (VHH) in these heavy chain–only Abs demonstrate comparable affinity and specificity for antigens to conventional immunoglobulins despite their much smaller size. To date, immunizations in humans and animal models have yielded only Abs with limited ability to neutralize HIV-1. In this study, a VHH phagemid library generated from a llama that was multiply immunized with recombinant trimeric HIV-1 envelope proteins (Envs) was screened directly for HIV-1 neutralization. One VHH, L8CJ3 (J3), neutralized 96 of 100 tested HIV-1 strains, encompassing subtypes A, B, C, D, BC, AE, AG, AC, ACD, CD, and G. J3 also potently neutralized chimeric simian-HIV strains with HIV subtypes B and C Env. The sequence of J3 is highly divergent from previous anti–HIV-1 VHH and its own germline sequence. J3 achieves broad and potent neutralization of HIV-1 via interaction with the CD4-binding site of HIV-1 Env. This study may represent a new benchmark for immunogens to be included in B cell–based vaccines and supports the development of VHH as anti–HIV-1 microbicides.
Collapse
Affiliation(s)
- Laura E McCoy
- Wohl Virion Centre, University College London, London WC1E 6BT, England, UK
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
156
|
Ringe R, Bhattacharya J. Association of enhanced HIV-1 neutralization by a single Y681H substitution in gp41 with increased gp120-CD4 interaction and macrophage infectivity. PLoS One 2012; 7:e37157. [PMID: 22606344 PMCID: PMC3351407 DOI: 10.1371/journal.pone.0037157] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2011] [Accepted: 04/14/2012] [Indexed: 11/18/2022] Open
Abstract
HIV-1 variants that show unusual sensitivity to autologous antibodies due to presence of critical neutralization signatures would likely contribute towards rational envelope based HIV-1 vaccine design. In the present study, we found that presence of a naturally occurring H681 in gp41 membrane proximal external region (MPER) of a clade C envelope (Env) obtained from a recently infected Indian patient conferred increased sensitivity to autologous and heterologous plasma antibodies. Furthermore, Env-pseudotyped viruses expressing H681 showed increased sensitivity to soluble CD4, b12 and 4E10 monoclonal antibodies both in related and unrelated Envs and was corroborated with increased Env susceptibility and binding to cellular CD4 as well as with prolonged exposure of MPER epitopes. The increased gp120-CD4 interaction was further associated with relative exposure of CD4-induced epitopes and macrophage infectivity. In summary, our data indicate that Y681H substitution exposes neutralizing epitopes in CD4bs and MPER towards comprehensive interference in HIV-1 entry.
Collapse
Affiliation(s)
| | - Jayanta Bhattacharya
- Department of Molecular Virology, National AIDS Research Institute, Indian Council of Medical Research, Bhosari, Pune, India
- * E-mail:
| |
Collapse
|
157
|
Thippeshappa R, Ruan H, Kimata JT. Breaking Barriers to an AIDS Model with Macaque-Tropic HIV-1 Derivatives. BIOLOGY 2012; 1:134-64. [PMID: 23336082 PMCID: PMC3546514 DOI: 10.3390/biology1020134] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Revised: 06/14/2012] [Accepted: 06/18/2012] [Indexed: 12/15/2022]
Abstract
The development of an animal model of human immunodeficiency virus type 1 (HIV-1)/AIDS that is suitable for preclinical testing of antiretroviral therapy, vaccines, curative strategies, and studies of pathogenesis has been hampered by the human-specific tropism of HIV-1. Although simian immunodeficiency virus (SIV) or HIV-1/SIV chimeric viruses (SHIVs)-rhesus macaque models are excellent surrogates for AIDS research, the genetic differences between SIV or SHIV and HIV-1 limit their utility as model systems. The identification of innate retro viral restriction factors has increased our understanding about blockades to HIV-1 replication in macaques and provided a guide for the construction of macaque-tropic HIV-1 clones. However, while these viruses replicate in macaque cells in vitro, they are easily controlled and have not caused AIDS in host animals, indicating that we may not fully understand the restrictive barriers of innate immunity. In this review, we discuss recent findings regarding HIV-1 restriction factors, particularly as they apply to cross-species transmission of primate lentiviruses and the development of a macaque model of HIV-1/AIDS.
Collapse
Affiliation(s)
| | | | - Jason T. Kimata
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA; (R.T.); (H.R.)
| |
Collapse
|
158
|
Kinetic mechanism for HIV-1 neutralization by antibody 2G12 entails reversible glycan binding that slows cell entry. Proc Natl Acad Sci U S A 2012; 109:7829-34. [PMID: 22547820 DOI: 10.1073/pnas.1109728109] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Despite structural knowledge of broadly neutralizing monoclonal antibodies (NMAbs) complexed to HIV-1 gp120 and gp41 envelope glycoproteins, virus inactivation mechanisms have been difficult to prove, in part because neutralization assays are complex and were previously not understood. Concordant with recent evidence that HIV-1 titers are determined by a race between entry of cell-attached virions and competing inactivation processes, we show that NMAb 2G12, which binds to gp120 N-glycans with α (1, 2)-linked mannose termini and inhibits replication after passive transfer into patients, neutralizes by slowing entry of adsorbed virions. Accordingly, apparent neutralization is attenuated when a kinetically competing virus inactivation pathway is blocked. Moreover, removing 2G12 from media causes its dissociation from virions coupled to accelerated entry and restored infectivity, demonstrating the reversibility of neutralization. A difference between 2G12 dissociation and infectivity recovery rates implies that the inhibited complexes at virus-cell junctions contain several 2G12's that must dissociate before entry commences. Quantitative microscopy of 2G12 binding and dissociation from single virions and studies using a split CCR5 coreceptor suggest that 2G12 competitively inhibits interactions between gp120's V3 loop and the tyrosine sulfate-containing CCR5 amino terminus, thereby reducing assembly of complexes that catalyze entry. These results reveal a unique reversible kinetic mechanism for neutralization by an antibody that binds near a critical V3 region in the glycan shield of gp120.
Collapse
|
159
|
Sato S, Ouellet M, St-Pierre C, Tremblay MJ. Glycans, galectins, and HIV-1 infection. Ann N Y Acad Sci 2012; 1253:133-48. [DOI: 10.1111/j.1749-6632.2012.06475.x] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
160
|
Kong L, Sattentau QJ. Antigenicity and Immunogenicity in HIV-1 Antibody-Based Vaccine Design. JOURNAL OF AIDS & CLINICAL RESEARCH 2012; S8:3. [PMID: 23227445 PMCID: PMC3515071 DOI: 10.4172/2155-6113] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Neutralizing antibodies can protect from infection by immunodeficiency viruses. However, the induction by active vaccination of antibodies that can potently neutralize a broad range of circulating virus strains is a goal not yet achieved, despite more than 2 decades of research. Here we review progress made in the field, from early empirical studies to today's rational structure-based vaccine antigen design. We discuss the existence of broadly neutralizing antibodies, their implications for epitope discovery and recent progress made in antigen design. Finally, we consider the relationship between antigenicity and immunogenicity for B cell recognition and antibody production, a major hurdle for rational vaccine design to overcome.
Collapse
Affiliation(s)
- Leopold Kong
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
- The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Quentin J Sattentau
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| |
Collapse
|
161
|
Rubio-Infante N, Govea-Alonso DO, Alpuche-Solís ÁG, García-Hernández AL, Soria-Guerra RE, Paz-Maldonado LMT, Ilhuicatzi-Alvarado D, Varona-Santos JT, Verdín-Terán L, Korban SS, Moreno-Fierros L, Rosales-Mendoza S. A chloroplast-derived C4V3 polypeptide from the human immunodeficiency virus (HIV) is orally immunogenic in mice. PLANT MOLECULAR BIOLOGY 2012; 78:337-49. [PMID: 22228408 DOI: 10.1007/s11103-011-9870-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Accepted: 12/10/2011] [Indexed: 05/23/2023]
Abstract
Although the human immunodeficiency virus (HIV) causes one of the most important infectious diseases worldwide, attempts to develop an effective vaccine remain elusive. Designing recombinant proteins capable of eliciting significant and protective mammalian immune responses remain a priority. Moreover, large-scale production of proteins of interest at affordable cost remains a challenge for modern biotechnology. In this study, a synthetic gene encoding a C4V3 recombinant protein, known to induce systemic and mucosal immune responses in mammalian systems, has been introduced into tobacco chloroplasts to yield high levels of expression. Integration of the transgene into the tobacco plastome has been verified by Southern blot hybridization. The recombinant C4V3 protein is also detected in tobacco chloroplasts by confocal microscopy. Reactivity of the heterologous protein with both an anti-C4V3 rabbit serum as well as sera from HIV positive patients have been assayed using Western blots. When administered by the oral route in a four-weekly dose immunization scheme, the plant-derived C4V3 has elicited both systemic and mucosal antibody responses in BALB/c mice, as well as CD4+ T cell proliferation responses. These findings support the viability of using plant chloroplasts as biofactories for HIV candidate vaccines, and could serve as important vehicles for the development of a plant-based candidate vaccine against HIV.
Collapse
Affiliation(s)
- Néstor Rubio-Infante
- Laboratorio de Biofarmacéuticos Recombinantes, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, 78210 San Luis Potosí, Mexico
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
162
|
Rosales-Mendoza S, Rubio-Infante N, Govea-Alonso DO, Moreno-Fierros L. Current status and perspectives of plant-based candidate vaccines against the human immunodeficiency virus (HIV). PLANT CELL REPORTS 2012; 31:495-511. [PMID: 22159962 DOI: 10.1007/s00299-011-1194-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Revised: 11/03/2011] [Accepted: 11/18/2011] [Indexed: 05/31/2023]
Abstract
Genetically engineered plants are economical platforms for the large-scale production of recombinant proteins and have been used over the last 21 years as models for oral vaccines against a wide variety of human infectious and autoimmune diseases with promising results. The main inherent advantages of this approach consist in the absence of purification needs and easy production and administration. One relevant infectious agent is the human immunodeficiency virus (HIV), since AIDS evolved as an alarming public health problem implicating very high costs for government agencies in most African and developing countries. The design of an effective and inexpensive vaccine able to limit viral spread and neutralizing the viral entry is urgently needed. Due to the limited efficacy of the vaccines assessed in clinical trials, new HIV vaccines able to generate broad immune profiles are a priority in the field. This review discusses the current advances on the topic of using plants as alternative expression systems to produce functional vaccine components against HIV, including antigens from Env, Gag and early proteins such as Tat and Nef. Ongoing projects of our group based on the expression of chimeric proteins comprising C4 and V3 domains from gp120, as an approach to elicit broadly neutralizing antibodies are mentioned. The perspectives of the revised approaches, such as the great need of assessing the oral immunogenicity and a detailed immunological characterization of the elicited immune responses, are also discussed.
Collapse
Affiliation(s)
- Sergio Rosales-Mendoza
- Laboratorio de Biofarmacéuticos recombinantes, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, San Luis Potosí 78210, Mexico.
| | | | | | | |
Collapse
|
163
|
Subtle alteration of residues including N-linked glycans in V2 loop modulate HIV-1 neutralization by PG9 and PG16 monoclonal antibodies. Virology 2012; 426:34-41. [PMID: 22314018 DOI: 10.1016/j.virol.2012.01.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2011] [Revised: 12/27/2011] [Accepted: 01/13/2012] [Indexed: 11/23/2022]
Abstract
Recent discovery of several potent and broadly neutralizing monoclonal antibodies (MAbs) (such as PG9 and PG16) to HIV-1 provided clues on newer vaccine targets. In the present study, we found an env clone obtained from a slow progressor showing significant resistance to PG9 and PG16 MAbs in sharp contrast to other contemporaneous autologous env clones. By constructing chimeric envelopes and specific substitutions we found that both loop length and spatial orientation of glycan residues in addition to the net charge of the β sheet C region that directly binds to PG9 CDRH3 within V2 loop significantly modulated HIV-1 sensitivity to PG9 and PG16 MAbs. Similar observation were made with several other Envs which varied in length, glycan content and net charge in PG9 contacting complementary region in V2 loop. Our data indicated that subtle change within V2 loop alone modulates exposition of quaternary epitopes that are targets of PG9/PG16 MAbs.
Collapse
|
164
|
Huang X, Jin W, Hu K, Luo S, Du T, Griffin GE, Shattock RJ, Hu Q. Highly conserved HIV-1 gp120 glycans proximal to CD4-binding region affect viral infectivity and neutralizing antibody induction. Virology 2012; 423:97-106. [DOI: 10.1016/j.virol.2011.11.023] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2011] [Revised: 11/15/2011] [Accepted: 11/28/2011] [Indexed: 10/14/2022]
|
165
|
Baschieri S. Virus Glycoproteins Tagged with the Human Fc Domain as Second Generation Vaccine Candidates. INNOVATION IN VACCINOLOGY 2012. [PMCID: PMC7122206 DOI: 10.1007/978-94-007-4543-8_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Traditional vaccines such as inactivated or live attenuated vaccines, are gradually giving way to more biochemically defined vaccines that are most often based on a recombinant antigen known to possess neutralizing epitopes. Such vaccines can offer improvements in speed, safety and manufacturing process but an inevitable consequence of their high degree of purification is that immunogenicity is reduced through the lack of the innate triggering molecules present in more complex preparations. Targeting recombinant vaccines to antigen presenting cells (APCs) such as dendritic cells however can improve immunogenicity by ensuring that antigen processing is as efficient as possible. Immune complexes, one of a number of routes of APC targeting, are mimicked by a recombinant approach, crystallizable fragment (Fc) fusion proteins, in which the target immunogen is linked directly to an antibody effector domain capable of interaction with receptors, FcR, on the APC cell surface. A number of virus Fc fusion proteins have been expressed in insect cells using the baculovirus expression system and shown to be efficiently produced and purified. Their use for immunization next to non-Fc tagged equivalents shows that they are powerfully immunogenic in the absence of added adjuvant and that immune stimulation is the result of the Fc-FcR interaction.
Collapse
Affiliation(s)
- Selene Baschieri
- Italian National Agency for New Technolo, Energy and Sustainable Economic Developm, ENEA, Via Anguillarese 301, Rome, 00123 Italy
| |
Collapse
|
166
|
Klasse PJ, Sanders RW, Cerutti A, Moore JP. How can HIV-type-1-Env immunogenicity be improved to facilitate antibody-based vaccine development? AIDS Res Hum Retroviruses 2012; 28:1-15. [PMID: 21495876 DOI: 10.1089/aid.2011.0053] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
No vaccine candidate has induced antibodies (Abs) that efficiently neutralize multiple primary isolates of HIV-1. Preexisting high titers of neutralizing antibodies (NAbs) are essential, because the virus establishes infection before anamnestic responses could take effect. HIV-1 infection elicits Abs against Env, Gag, and other viral proteins, but of these only a subset of the anti-Env Abs can neutralize the virus. Whereas the corresponding proteins from other viruses form the basis of successful vaccines, multiple large doses of HIV-1 Env elicit low, transient titers of Abs that are not protective in humans. The inaccessibility of neutralization epitopes hinders NAb induction, but Env may also subvert the immune response by interacting with receptors on T cells, B cells, monocytes, macrophages, and dendritic cells. Here, we discuss evidence from immunizations of different species with various modified Env constructs. We also suggest how the divergent Ab responses to Gag and Env during infection may reflect differences in B cell regulation. Drawing on these analyses, we outline strategies for improving Env as a component of a vaccine aimed at inducing strong and sustained NAb responses.
Collapse
Affiliation(s)
- Per Johan Klasse
- Department of Microbiology and Immunology, Cornell University, Weill Cornell Medical College, New York, New York
| | - Rogier W. Sanders
- Department of Microbiology and Immunology, Cornell University, Weill Cornell Medical College, New York, New York
- Laboratory of Experimental Virology, Department of Medical Microbiology, Center for Infection and Immunity Amsterdam, Academic Medical Center of the University of Amsterdam, Amsterdam, The Netherlands
| | - Andrea Cerutti
- Immunology Institute, Department of Medicine, Mount Sinai School of Medicine, New York, New York
- Catalan Institute for Research and Advanced Studies, Barcelona Biomedical Research Park, IMIM-Hospital del Mar, Barcelona, Spain
| | - John P. Moore
- Department of Microbiology and Immunology, Cornell University, Weill Cornell Medical College, New York, New York
| |
Collapse
|
167
|
Targeting HIV-1 envelope glycoprotein trimers to B cells by using APRIL improves antibody responses. J Virol 2011; 86:2488-500. [PMID: 22205734 DOI: 10.1128/jvi.06259-11] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
An HIV-1 vaccine remains elusive, in part because various factors limit the quantity and quality of the antibodies raised against the viral envelope glycoprotein complex (Env). We hypothesized that targeting Env vaccines directly to B cells, by fusing them to molecules that bind and activate these cells, would improve Env-specific antibody responses. Therefore, we fused trimeric Env gp140 to A PRoliferation-Inducing Ligand (APRIL), B-cell Activating Factor (BAFF), and CD40 Ligand (CD40L). The Env-APRIL, Env-BAFF, and Env-CD40L gp140 trimers all enhanced the expression of activation-induced cytidine deaminase (AID), the enzyme responsible for inducing somatic hypermutation, antibody affinity maturation, and antibody class switching. They also triggered IgM, IgG, and IgA secretion from human B cells in vitro. The Env-APRIL trimers induced higher anti-Env antibody responses in rabbits, including neutralizing antibodies against tier 1 viruses. The enhanced Env-specific responses were not associated with a general increase in total plasma antibody concentrations, indicating that the effect of APRIL was specific for Env. All the rabbit sera raised against gp140 trimers, irrespective of the presence of CD40L, BAFF, or APRIL, recognized trimeric Env efficiently, whereas sera raised against gp120 monomers did not. The levels of trimer-binding and virus-neutralizing antibodies were strongly correlated, suggesting that gp140 trimers are superior to gp120 monomers as immunogens. Targeting and activating B cells with a trimeric HIV-1 Env-APRIL fusion protein may therefore improve the induction of humoral immunity against HIV-1.
Collapse
|
168
|
Abstract
The genetic diversity of pathogens presents a challenge to the development of broadly effective vaccines. In this issue, Scarselli et al. combine atomic-level structural information with genomics and classical vaccinology to design a single immunogen that elicits protective immunity against more than 300 natural variants of the bacterial pathogen meningococcus B. This accomplishment provides a glimpse of the power of structure-based vaccine design to create immunogens capable of eliciting protective responses against genetically diverse pathogens.
Collapse
Affiliation(s)
- Peter D Kwong
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| | | |
Collapse
|
169
|
Nabel GJ, Kwong PD, Mascola JR. Progress in the rational design of an AIDS vaccine. Philos Trans R Soc Lond B Biol Sci 2011; 366:2759-65. [PMID: 21893538 DOI: 10.1098/rstb.2011.0096] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Human immunodeficiency virus-1 (HIV-1) has a high degree of genetic and antigenic diversity that has impeded the development of an effective vaccine using traditional methods. We are attempting to develop an AIDS vaccine by employing strategies that include structural biology and computational modelling, in an effort to develop immunogens capable of eliciting neutralizing antibodies of the requisite breadth and potency against circulating strains of HIV-1.
Collapse
Affiliation(s)
- Gary J Nabel
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 40 Convent Drive, MSC 3005, Bethesda, MD 20892, USA.
| | | | | |
Collapse
|
170
|
Feng Y, McKee K, Tran K, O'Dell S, Schmidt SD, Phogat A, Forsell MN, Karlsson Hedestam GB, Mascola JR, Wyatt RT. Biochemically defined HIV-1 envelope glycoprotein variant immunogens display differential binding and neutralizing specificities to the CD4-binding site. J Biol Chem 2011; 287:5673-86. [PMID: 22167180 DOI: 10.1074/jbc.m111.317776] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
HIV-1 gp120 binds the primary receptor CD4. Recently, a plethora of broadly neutralizing antibodies to the gp120 CD4-binding site (CD4bs) validated this region as a target for immunogen design. Here, we asked if modified HIV-1 envelope glycoproteins (Env) designed to increase CD4 recognition might improve recognition by CD4bs neutralizing antibodies and more efficiently elicit such reactivities. We also asked if CD4bs stabilization, coupled with altering the Env format (monomer to trimer or cross-clade), might better elicit neutralizing antibodies by focusing the immune response on the functionally conserved CD4bs. We produced monomeric and trimeric Envs stabilized by mutations within the gp120 CD4bs cavity (pocket-filling; PF2) or by appending heterologous trimerization motifs to soluble Env ectodomains (gp120/gp140). Recombinant glycoproteins were purified to relative homogeneity, and ligand binding properties were analyzed by ELISA, surface plasmon resonance, and isothermal titration microcalorimetry. In some formats, the PF2 substitutions increased CD4 affinity, and importantly, PF2-containing proteins were better recognized by the broadly neutralizing CD4bs mAbs, VRC01 and VRC-PG04. Based on this analysis, we immunized selected Env variants into rabbits using heterologous or homologous regimens. Analysis of the sera revealed that homologous inoculation of the PF2-containing, variable region-deleted YU2 gp120 trimers (ΔV123/PF2-GCN4) more rapidly elicited CD4bs-directed neutralizing antibodies compared with other regimens, whereas homologous trimers elicited increased neutralization potency, mapping predominantly to the gp120 third major variable region (V3). These results suggest that some engineered Env proteins may more efficiently direct responses toward the conserved CD4bs and be valuable to elicit antibodies of greater neutralizing capacity.
Collapse
Affiliation(s)
- Yu Feng
- IAVI Neutralizing Antibody Center, Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California 92037, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
171
|
Moir S, Malaspina A, Fauci AS. Prospects for an HIV vaccine: leading B cells down the right path. Nat Struct Mol Biol 2011; 18:1317-21. [PMID: 22139037 DOI: 10.1038/nsmb.2194] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Susan Moir
- Laboratory of Immunoregulation, US National Institutes of Health, Bethesda, Maryland, USA.
| | | | | |
Collapse
|
172
|
Boonchawalit S, Jullaksorn D, Uttiyoung J, Yowang A, Krathong N, Chautrakul S, Yamashita A, Ikuta K, Roobsoong A, Kanitvittaya S, Sawanpanyalert P, Kameoka M. Molecular evolution of HIV-1 CRF01_AE Env in Thai patients. PLoS One 2011; 6:e27098. [PMID: 22073263 PMCID: PMC3206936 DOI: 10.1371/journal.pone.0027098] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2011] [Accepted: 10/10/2011] [Indexed: 12/03/2022] Open
Abstract
Background The envelope glycoproteins (Env), gp120 and gp41, are the most variable proteins of human immunodeficiency virus type 1 (HIV-1), and are the major targets of humoral immune responses against HIV-1. A circulating recombinant form of HIV-1, CRF01_AE, is prevalent throughout Southeast Asia; however, only limited information regarding the immunological characteristics of CRF01_AE Env is currently available. In this study, we attempted to examine the evolutionary pattern of CRF01_AE Env under the selection pressure of host immune responses. Methodology/Principal Findings Peripheral blood samples were collected periodically over 3 years from 15 HIV-1-infected individuals residing in northern Thailand, and amplified env genes from the samples were subjected to computational analysis. The V5 region of gp120 showed highest variability in several samples over 3 years, whereas the V1/V2 and/or V4 regions of gp120 also showed high variability in many samples. In addition, the N-terminal part of the C3 region of gp120 showed highest amino acid diversity among the conserved regions of gp120. Chronological changes in the numbers of amino acid residues in gp120 variable regions and potential N-linked glycosylation (PNLG) sites are involved in increasing the variability of Env gp120. Furthermore, the C3 region contained several amino acid residues potentially under positive selection, and APOBEC3 family protein-mediated G to A mutations were frequently detected in such residues. Conclusions/Significance Several factors, including amino acid substitutions particularly in gp120 C3 and V5 regions as well as changes in the number of PNLG sites and in the length of gp120 variable regions, were revealed to be involved in the molecular evolution of CRF01_AE Env. In addition, a similar tendency was observed between CRF01_AE and subtype C Env with regard to the amino acid variation of gp120 V3 and C3 regions. These results may provide important information for understanding the immunological characteristics of CRF01_AE Env.
Collapse
Affiliation(s)
- Samatchaya Boonchawalit
- Thailand-Japan Research Collaboration Center on Emerging and Re-emerging Infections (RCC-ERI), Nonthaburi, Thailand
| | - Duangrat Jullaksorn
- National Institute of Health, Department of Medical Sciences (DMSc), Ministry of Public Health (MOPH), Nonthaburi, Thailand
| | - Jiraporn Uttiyoung
- Regional Medical Science Center Chiangrai, DMSc, MOPH, Chiangrai, Thailand
| | - Amara Yowang
- Regional Medical Science Center Chiangrai, DMSc, MOPH, Chiangrai, Thailand
| | | | | | | | - Kazuyoshi Ikuta
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | | | | | - Pathom Sawanpanyalert
- National Institute of Health, Department of Medical Sciences (DMSc), Ministry of Public Health (MOPH), Nonthaburi, Thailand
| | - Masanori Kameoka
- Thailand-Japan Research Collaboration Center on Emerging and Re-emerging Infections (RCC-ERI), Nonthaburi, Thailand
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- * E-mail:
| |
Collapse
|
173
|
Machiels B, Lété C, Guillaume A, Mast J, Stevenson PG, Vanderplasschen A, Gillet L. Antibody evasion by a gammaherpesvirus O-glycan shield. PLoS Pathog 2011; 7:e1002387. [PMID: 22114560 PMCID: PMC3219721 DOI: 10.1371/journal.ppat.1002387] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2011] [Accepted: 10/04/2011] [Indexed: 12/02/2022] Open
Abstract
All gammaherpesviruses encode a major glycoprotein homologous to the Epstein-Barr virus gp350. These glycoproteins are often involved in cell binding, and some provide neutralization targets. However, the capacity of gammaherpesviruses for long-term transmission from immune hosts implies that in vivo neutralization is incomplete. In this study, we used Bovine Herpesvirus 4 (BoHV-4) to determine how its gp350 homolog--gp180--contributes to virus replication and neutralization. A lack of gp180 had no impact on the establishment and maintenance of BoHV-4 latency, but markedly sensitized virions to neutralization by immune sera. Antibody had greater access to gB, gH and gL on gp180-deficient virions, including neutralization epitopes. Gp180 appears to be highly O-glycosylated, and removing O-linked glycans from virions also sensitized them to neutralization. It therefore appeared that gp180 provides part of a glycan shield for otherwise vulnerable viral epitopes. Interestingly, this O-glycan shield could be exploited for neutralization by lectins and carbohydrate-specific antibody. The conservation of O-glycosylation sites in all gp350 homologs suggests that this is a general evasion mechanism that may also provide a therapeutic target.
Collapse
Affiliation(s)
- Bénédicte Machiels
- Immunology-Vaccinology (B43b), Department of Infectious and Parasitic Diseases (B43b), Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Céline Lété
- Immunology-Vaccinology (B43b), Department of Infectious and Parasitic Diseases (B43b), Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Antoine Guillaume
- Immunology-Vaccinology (B43b), Department of Infectious and Parasitic Diseases (B43b), Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Jan Mast
- Department Biocontrole, Research Unit Electron Microscopy, Veterinary and Agrochemical Research Centre, CODA-CERVA, Groeselenberg, Ukkel, Belgium
| | - Philip G. Stevenson
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Alain Vanderplasschen
- Immunology-Vaccinology (B43b), Department of Infectious and Parasitic Diseases (B43b), Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Laurent Gillet
- Immunology-Vaccinology (B43b), Department of Infectious and Parasitic Diseases (B43b), Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| |
Collapse
|
174
|
Broad and potent neutralizing antibody responses elicited in natural HIV-2 infection. J Virol 2011; 86:947-60. [PMID: 22031948 DOI: 10.1128/jvi.06155-11] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Compared with human immunodeficiency virus type 1 (HIV-1), little is known about the susceptibility of HIV-2 to antibody neutralization. We characterized the potency and breadth of neutralizing antibody (NAb) responses in 64 subjects chronically infected with HIV-2 against three primary HIV-2 strains: HIV-2(7312A), HIV-2(ST), and HIV-2(UC1). Surprisingly, we observed in a single-cycle JC53bl-13/TZM-bl virus entry assay median reciprocal 50% inhibitory concentration (IC(50)) NAb titers of 1.7 × 10(5), 2.8 × 10(4), and 3.3 × 10(4), respectively. A subset of 5 patient plasma samples tested against a larger panel of 17 HIV-2 strains where the extracellular gp160 domain was substituted into the HIV-2(7312A) proviral backbone showed potent neutralization of all but 4 viruses. The specificity of antibody neutralization was confirmed using IgG purified from patient plasma, HIV-2 Envs cloned by single-genome amplification, viruses grown in human CD4(+) T cells and tested for neutralization sensitivity on human CD4(+) T target cells, and, as negative controls, env-minus viruses pseudotyped with HIV-1, vesicular stomatitis virus, or murine leukemia virus Env glycoproteins. Human monoclonal antibodies (MAbs) specific for HIV-2 V3 (6.10F), V4 (1.7A), CD4 binding site (CD4bs; 6.10B), CD4 induced (CD4i; 1.4H), and membrane-proximal external region (MPER; 4E10) epitopes potently neutralized the majority of 32 HIV-2 strains bearing Envs from 13 subjects. Patient antibodies competed with V3, V4, and CD4bs MAbs for binding to monomeric HIV-2 gp120 at titers that correlated significantly with NAb titers. HIV-2 MPER antibodies did not contribute to neutralization breadth or potency. These findings indicate that HIV-2 Env is highly immunogenic in natural infection, that high-titer broadly neutralizing antibodies are commonly elicited, and that unlike HIV-1, native HIV-2 Env trimers expose multiple broadly cross-reactive epitopes readily accessible to NAbs.
Collapse
|
175
|
Ketas TJ, Holuigue S, Matthews K, Moore JP, Klasse PJ. Env-glycoprotein heterogeneity as a source of apparent synergy and enhanced cooperativity in inhibition of HIV-1 infection by neutralizing antibodies and entry inhibitors. Virology 2011; 422:22-36. [PMID: 22018634 DOI: 10.1016/j.virol.2011.09.019] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2011] [Revised: 08/19/2011] [Accepted: 09/17/2011] [Indexed: 10/16/2022]
Abstract
We measured the inhibition of infectivity of HIV-1 isolates and derivative clones by combinations of neutralizing antibodies (NAbs) and other entry inhibitors in a single-cycle-replication assay. Synergy was analyzed both by the current linear and a new non-linear method. The new method reduced spurious indications of synergy and antagonism. Synergy between NAbs was overall weaker than between other entry inhibitors, and no stronger where one ligand is known to enhance the binding of another. However, synergy was stronger for a genetically heterogeneous HIV-1 R5 isolate than for its derivative clones. Enhanced cooperativity in inhibition by combinations, compared with individual inhibitors, correlated with increased synergy at higher levels of inhibition, while being less variable. Again, cooperativity enhancement was stronger for isolates than clones. We hypothesize that genetic, post-translational or conformational heterogeneity of the Env protein and of other targets for inhibitors can yield apparent synergy and increased cooperativity between inhibitors.
Collapse
Affiliation(s)
- Thomas J Ketas
- Department of Microbiology and Immunology, Weill Medical College of Cornell University, New York, NY 10065-4896, USA
| | | | | | | | | |
Collapse
|
176
|
Hosie MJ, Pajek D, Samman A, Willett BJ. Feline immunodeficiency virus (FIV) neutralization: a review. Viruses 2011; 3:1870-90. [PMID: 22069520 PMCID: PMC3205386 DOI: 10.3390/v3101870] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Revised: 09/28/2011] [Accepted: 09/29/2011] [Indexed: 11/16/2022] Open
Abstract
One of the major obstacles that must be overcome in the design of effective lentiviral vaccines is the ability of lentiviruses to evolve in order to escape from neutralizing antibodies. The primary target for neutralizing antibodies is the highly variable viral envelope glycoprotein (Env), a glycoprotein that is essential for viral entry and comprises both variable and conserved regions. As a result of the complex trimeric nature of Env, there is steric hindrance of conserved epitopes required for receptor binding so that these are not accessible to antibodies. Instead, the humoral response is targeted towards decoy immunodominant epitopes on variable domains such as the third hypervariable loop (V3) of Env. For feline immunodeficiency virus (FIV), as well as the related human immunodeficiency virus-1 (HIV-1), little is known about the factors that lead to the development of broadly neutralizing antibodies. In cats infected with FIV and patients infected with HIV-1, only rarely are plasma samples found that contain antibodies capable of neutralizing isolates from other clades. In this review we examine the neutralizing response to FIV, comparing and contrasting with the response to HIV. We ask whether broadly neutralizing antibodies are induced by FIV infection and discuss the comparative value of studies of neutralizing antibodies in FIV infection for the development of more effective vaccine strategies against lentiviral infections in general, including HIV-1.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Cat Diseases/immunology
- Cat Diseases/prevention & control
- Cat Diseases/virology
- Cats
- Gene Products, env/genetics
- Gene Products, env/immunology
- Gene Products, env/metabolism
- Humans
- Immune Evasion
- Immunity, Humoral
- Immunodeficiency Virus, Feline/genetics
- Immunodeficiency Virus, Feline/immunology
- Immunodominant Epitopes/immunology
- Lentivirus Infections/immunology
- Lentivirus Infections/prevention & control
- Lentivirus Infections/veterinary
- Lentivirus Infections/virology
- Molecular Sequence Data
Collapse
Affiliation(s)
- Margaret J Hosie
- Medical Research Council, University of Glasgow Centre for Virus Research, Henry Wellcome Building for Comparative Medical Sciences, 464 Bearsden Road, Glasgow G61 1QH, UK.
| | | | | | | |
Collapse
|
177
|
|
178
|
Wu X, Zhou T, Zhu J, Zhang B, Georgiev I, Wang C, Chen X, Longo NS, Louder M, McKee K, O’Dell S, Perfetto S, Schmidt SD, Shi W, Wu L, Yang Y, Yang ZY, Yang Z, Zhang Z, Bonsignori M, Crump JA, Kapiga SH, Sam NE, Haynes BF, Simek M, Burton DR, Koff WC, Doria-Rose NA, Connors M, Mullikin JC, Nabel GJ, Roederer M, Shapiro L, Kwong PD, Mascola JR. Focused evolution of HIV-1 neutralizing antibodies revealed by structures and deep sequencing. Science 2011; 333:1593-602. [PMID: 21835983 PMCID: PMC3516815 DOI: 10.1126/science.1207532] [Citation(s) in RCA: 696] [Impact Index Per Article: 49.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Antibody VRC01 is a human immunoglobulin that neutralizes about 90% of HIV-1 isolates. To understand how such broadly neutralizing antibodies develop, we used x-ray crystallography and 454 pyrosequencing to characterize additional VRC01-like antibodies from HIV-1-infected individuals. Crystal structures revealed a convergent mode of binding for diverse antibodies to the same CD4-binding-site epitope. A functional genomics analysis of expressed heavy and light chains revealed common pathways of antibody-heavy chain maturation, confined to the IGHV1-2*02 lineage, involving dozens of somatic changes, and capable of pairing with different light chains. Broadly neutralizing HIV-1 immunity associated with VRC01-like antibodies thus involves the evolution of antibodies to a highly affinity-matured state required to recognize an invariant viral structure, with lineages defined from thousands of sequences providing a genetic roadmap of their development.
Collapse
MESH Headings
- AIDS Vaccines
- Amino Acid Sequence
- Antibodies, Neutralizing/chemistry
- Antibodies, Neutralizing/genetics
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/isolation & purification
- Antibody Affinity
- Antibody Specificity
- Base Sequence
- Binding Sites
- Binding Sites, Antibody
- CD4 Antigens/metabolism
- Complementarity Determining Regions/genetics
- Crystallography, X-Ray
- Epitopes
- Evolution, Molecular
- Genes, Immunoglobulin Heavy Chain
- HIV Antibodies/chemistry
- HIV Antibodies/genetics
- HIV Antibodies/immunology
- HIV Antibodies/isolation & purification
- HIV Envelope Protein gp120/chemistry
- HIV Envelope Protein gp120/immunology
- HIV Envelope Protein gp120/metabolism
- HIV Infections/immunology
- HIV-1/chemistry
- HIV-1/immunology
- High-Throughput Nucleotide Sequencing
- Humans
- Immunoglobulin Fab Fragments/chemistry
- Immunoglobulin Fab Fragments/immunology
- Immunoglobulin Heavy Chains/chemistry
- Immunoglobulin Heavy Chains/immunology
- Immunoglobulin J-Chains/genetics
- Immunoglobulin Light Chains/chemistry
- Immunoglobulin Light Chains/immunology
- Models, Molecular
- Molecular Sequence Data
- Mutation
- Sequence Analysis, DNA
Collapse
Affiliation(s)
- Xueling Wu
- Vaccine Research Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Tongqing Zhou
- Vaccine Research Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jiang Zhu
- Vaccine Research Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Baoshan Zhang
- Vaccine Research Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ivelin Georgiev
- Vaccine Research Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Charlene Wang
- Vaccine Research Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Xuejun Chen
- Vaccine Research Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nancy S. Longo
- Vaccine Research Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mark Louder
- Vaccine Research Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Krisha McKee
- Vaccine Research Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sijy O’Dell
- Vaccine Research Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Stephen Perfetto
- Vaccine Research Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Stephen D. Schmidt
- Vaccine Research Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Wei Shi
- Vaccine Research Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Lan Wu
- Vaccine Research Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yongping Yang
- Vaccine Research Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Zhi-Yong Yang
- Vaccine Research Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Zhongjia Yang
- Vaccine Research Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Zhenhai Zhang
- Vaccine Research Center, National Institutes of Health, Bethesda, MD 20892, USA
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10032, USA
| | - Mattia Bonsignori
- Duke Human Vaccine Institute, Duke University School of Medicine, and Duke University Medical Center, Durham, NC 27710, USA
| | - John A. Crump
- Division of Infectious Diseases and International Health, Department of Medicine, and Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
- Kilimanjaro Christian Medical Centre and Kilimanjaro Christian Medical College, Tumaini University, Moshi, Tanzania
| | | | - Noel E. Sam
- Kilimanjaro Christian Medical Centre and Kilimanjaro Christian Medical College, Tumaini University, Moshi, Tanzania
- Kilimanjaro Reproductive Health Programme, Moshi, Tanzania
| | - Barton F. Haynes
- Duke Human Vaccine Institute, Duke University School of Medicine, and Duke University Medical Center, Durham, NC 27710, USA
| | - Melissa Simek
- International AIDS Vaccine Initiative (IAVI), New York, NY 10038, USA
| | - Dennis R. Burton
- Department of Immunology and Microbial Science and IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard, Cambridge, MA 02129, USA
| | - Wayne C. Koff
- International AIDS Vaccine Initiative (IAVI), New York, NY 10038, USA
| | - Nicole A. Doria-Rose
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mark Connors
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | - James C. Mullikin
- NIH Intramural Sequencing Center (NISC), National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Gary J. Nabel
- Vaccine Research Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mario Roederer
- Vaccine Research Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Lawrence Shapiro
- Vaccine Research Center, National Institutes of Health, Bethesda, MD 20892, USA
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10032, USA
| | - Peter D. Kwong
- Vaccine Research Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - John R. Mascola
- Vaccine Research Center, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
179
|
Tang H, Robinson JE, Gnanakaran S, Li M, Rosenberg ES, Perez LG, Haynes BF, Liao HX, LaBranche CC, Korber BT, Montefiori DC. epitopes immediately below the base of the V3 loop of gp120 as targets for the initial autologous neutralizing antibody response in two HIV-1 subtype B-infected individuals. J Virol 2011; 85:9286-99. [PMID: 21734041 PMCID: PMC3165744 DOI: 10.1128/jvi.02286-10] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2010] [Accepted: 06/15/2011] [Indexed: 01/01/2023] Open
Abstract
Epitopes that drive the initial autologous neutralizing antibody response in HIV-1-infected individuals could provide insights for vaccine design. Although highly strain specific, these epitopes are immunogenic, vulnerable to antibody attack on infectious virus, and could be involved in the ontogeny of broadly neutralizing antibody responses. To delineate such epitopes, we used site-directed mutagenesis, autologous plasma samples, and autologous monoclonal antibodies to map the amino acid changes that led to escape from the initial autologous neutralizing antibody response in two HIV-1 subtype B-infected individuals. Additional mapping of the epitopes was accomplished by using alanine scanning mutagenesis. Escape in the two individuals occurred by different pathways, but the responses in both cases appeared to be directed against the same region of gp120. In total, three amino acid positions were identified that were independently associated with autologous neutralization. Positions 295 and 332 are located immediately before and after the N- and C-terminal cysteines of the V3 loop, respectively, the latter of which affected an N-linked glycan that was critical to the neutralization epitope. Position 415 affected an N-linked glycan at position 413 in the C terminus of V4 that might mask epitopes near the base of V3. All three sites lie in close proximity on a four-stranded antiparallel sheet on the outer domain of gp120. We conclude that a region just below the base of the V3 loop, near the coreceptor binding domain of gp120, can be a target for autologous neutralization.
Collapse
Affiliation(s)
| | - James E. Robinson
- Department of Pediatrics, Tulane University Medical Center, New Orleans, Louisiana
| | - S. Gnanakaran
- Theoretical Biology and Biophysics Group, Los Alamos National Laboratory, Los Alamos, New Mexico
| | | | - Eric S. Rosenberg
- Pathology Service and Infectious Disease Division, Massachusetts General Hospital, and Harvard Medical School, Boston, Massachusetts
| | | | - Barton F. Haynes
- Medicine, Duke University Medical Center, Durham, North Carolina
| | - Hua-Xin Liao
- Medicine, Duke University Medical Center, Durham, North Carolina
| | | | - Bette T. Korber
- Theoretical Biology and Biophysics Group, Los Alamos National Laboratory, Los Alamos, New Mexico
| | | |
Collapse
|
180
|
Bonomelli C, Doores KJ, Dunlop DC, Thaney V, Dwek RA, Burton DR, Crispin M, Scanlan CN. The glycan shield of HIV is predominantly oligomannose independently of production system or viral clade. PLoS One 2011; 6:e23521. [PMID: 21858152 PMCID: PMC3156772 DOI: 10.1371/journal.pone.0023521] [Citation(s) in RCA: 178] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2011] [Accepted: 07/19/2011] [Indexed: 01/04/2023] Open
Abstract
The N-linked oligomannose glycans of HIV gp120 are a target for both microbicide and vaccine design. The extent of cross-clade conservation of HIV oligomannose glycans is therefore a critical consideration for the development of HIV prophylaxes. We measured the oligomannose content of virion-associated gp120 from primary virus from PBMCs for a range of viral isolates and showed cross-clade elevation (62–79%) of these glycans relative to recombinant, monomeric gp120 (∼30%). We also confirmed that pseudoviral production systems can give rise to notably elevated gp120 oligomannose levels (∼98%), compared to gp120 derived from a single-plasmid viral system using the HIVLAI backbone (56%). This study highlights differences in glycosylation between virion-associated and recombinant gp120.
Collapse
Affiliation(s)
- Camille Bonomelli
- Department of Biochemistry, Oxford Glycobiology Institute, University of Oxford, Oxford, United Kingdom
| | - Katie J. Doores
- Department of Immunology and Microbial Science and IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, California, United States of America
- Ragon Institute of MGH, MIT and Harvard, Boston, Massachusetts, United States of America
| | - D. Cameron Dunlop
- Department of Immunology and Microbial Science and IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, California, United States of America
| | - Victoria Thaney
- Department of Immunology and Microbial Science and IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, California, United States of America
| | - Raymond A. Dwek
- Department of Biochemistry, Oxford Glycobiology Institute, University of Oxford, Oxford, United Kingdom
| | - Dennis R. Burton
- Department of Immunology and Microbial Science and IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, California, United States of America
- Ragon Institute of MGH, MIT and Harvard, Boston, Massachusetts, United States of America
| | - Max Crispin
- Department of Biochemistry, Oxford Glycobiology Institute, University of Oxford, Oxford, United Kingdom
| | - Christopher N. Scanlan
- Department of Biochemistry, Oxford Glycobiology Institute, University of Oxford, Oxford, United Kingdom
- * E-mail:
| |
Collapse
|
181
|
Pruzina S, Williams GT, Kaneva G, Davies SL, Martin-Lopez A, Bruggemann M, Vieira SM, Jeffs SA, Sattentau QJ, Neuberger MS. Human monoclonal antibodies to HIV-1 gp140 from mice bearing YAC-based human immunoglobulin transloci. Protein Eng Des Sel 2011; 24:791-9. [DOI: 10.1093/protein/gzr038] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
182
|
Morelli L, Poletti L, Lay L. Carbohydrates and Immunology: Synthetic Oligosaccharide Antigens for Vaccine Formulation. European J Org Chem 2011. [DOI: 10.1002/ejoc.201100296] [Citation(s) in RCA: 117] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Laura Morelli
- Dipartimento di Chimica Organica e Industriale, CISI and ISTM‐CNR, Università degli Studi di Milano, Via Venezian 21, 20133 Milano, Italy
| | - Laura Poletti
- Dipartimento di Chimica Organica e Industriale, CISI and ISTM‐CNR, Università degli Studi di Milano, Via Venezian 21, 20133 Milano, Italy
| | - Luigi Lay
- Dipartimento di Chimica Organica e Industriale, CISI and ISTM‐CNR, Università degli Studi di Milano, Via Venezian 21, 20133 Milano, Italy
| |
Collapse
|
183
|
HIV microbicides: state-of-the-art and new perspectives on the development of entry inhibitors. Future Med Chem 2011; 2:1141-59. [PMID: 21426161 DOI: 10.4155/fmc.10.203] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Since the discovery of HIV at the beginning of the 1980s, numerous efforts have been devoted to the search of an efficient vaccine. There are at least 25 drugs available for HIV treatment, but no cure is available. The observation that therapy for HIV disease is life long and that these drugs are associated with a number of side effects underlines the need for approaches aimed at preventing rather than treating infection. Additionally, the economic burden of treatment for the HIV infection occupies an increasing share of healthcare expenditure, making current practices likely to become difficult to sustain in the long run. Unfortunately, no effective vaccine for this disease is foreseeable in the near future. Microbicides could be an alternate way to build preventative approaches to HIV infection. Strategies based on preventing the virus from reaching its target cells seem to have some room for development and application. In this review we explore the state-of-the-art of available microbicides, focusing on HIV entry inhibitors. In addition, we discuss new compounds that show anti-HIV activity, which could be effective candidates.
Collapse
|
184
|
Zhuang K, Finzi A, Tasca S, Shakirzyanova M, Knight H, Westmoreland S, Sodroski J, Cheng-Mayer C. Adoption of an "open" envelope conformation facilitating CD4 binding and structural remodeling precedes coreceptor switch in R5 SHIV-infected macaques. PLoS One 2011; 6:e21350. [PMID: 21760891 PMCID: PMC3132741 DOI: 10.1371/journal.pone.0021350] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2011] [Accepted: 05/25/2011] [Indexed: 11/18/2022] Open
Abstract
A change in coreceptor preference from CCR5 to CXCR4 towards the end stage disease in some HIV-1 infected individuals has been well documented, but the reasons and mechanisms for this tropism switch remain elusive. It has been suggested that envelope structural constraints in accommodating amino acid changes required for CXCR4 usage is an obstacle to tropism switch, limiting the rate and pathways available for HIV-1 coreceptor switching. The present study was initiated in two R5 SHIV(SF162P3N)-infected rapid progressor macaques with coreceptor switch to test the hypothesis that an early step in the evolution of tropism switch is the adoption of a less constrained and more "open" envelope conformation for better CD4 usage, allowing greater structural flexibility to accommodate further mutational changes that confer CXCR4 utilization. We show that, prior to the time of coreceptor switch, R5 viruses in both macaques evolved to become increasingly sCD4-sensitive, suggestive of enhanced exposure of the CD4 binding site and an "open" envelope conformation, and this correlated with better gp120 binding to CD4 and with more efficient infection of CD4(low) cells such as primary macrophages. Moreover, significant changes in neutralization sensitivity to agents and antibodies directed against functional domains of gp120 and gp41 were seen for R5 viruses close to the time of X4 emergence, consistent with global changes in envelope configuration and structural plasticity. These observations in a simian model of R5-to-X4 evolution provide a mechanistic basis for the HIV-1 coreceptor switch.
Collapse
Affiliation(s)
- Ke Zhuang
- Aaron Diamond AIDS Research Center, New York, New York, United States of America
| | - Andres Finzi
- Division of AIDS, Department of Cancer Immunology and AIDS, Department of Pathology, Dana-Faber Cancer Institute, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Silvana Tasca
- Aaron Diamond AIDS Research Center, New York, New York, United States of America
| | - Madina Shakirzyanova
- Aaron Diamond AIDS Research Center, New York, New York, United States of America
| | - Heather Knight
- Division of Comparative Pathology, New England Primate Research Center, Harvard Medical School, Southborough, Massachusetts, United States of America
| | - Susan Westmoreland
- Division of Comparative Pathology, New England Primate Research Center, Harvard Medical School, Southborough, Massachusetts, United States of America
| | - Joseph Sodroski
- Division of AIDS, Department of Cancer Immunology and AIDS, Department of Pathology, Dana-Faber Cancer Institute, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Cecilia Cheng-Mayer
- Aaron Diamond AIDS Research Center, New York, New York, United States of America
| |
Collapse
|
185
|
Relative reactivity of HIV-1 polyclonal plasma antibodies directed to V3 and MPER regions suggests immunodominance of V3 over MPER and dependence of high anti-V3 antibody titers on virus persistence. Arch Virol 2011; 156:1787-94. [PMID: 21735212 DOI: 10.1007/s00705-011-1053-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2011] [Accepted: 06/18/2011] [Indexed: 10/18/2022]
Abstract
Antibodies to two crucial regions, the third variable loop (V3) of gp120 and the membrane-proximal external region (MPER) of gp41 are important for HIV-1 neutralization. We here evaluated the relative binding of polyclonal plasma antibodies from 99 HIV-1-infected individuals from India to the consensus-C V3 and MPER peptides and observed immunodominance of V3 over MPER (p < 0.0001). We further examined the V3- and MPER-specific antibody correlates with clinical parameters. Our results revealed that anti-V3 antibody titers are significantly lower in patients on ART compared to drug-naive individuals (p < 0.0001), most likely due to a decrease in plasma viral load, irrespective of their CD4 counts and total IgG. No such association was observed for MPER, with a similar trend in four follow-up patients. These findings strongly suggest that high titers of V3-specific antibodies are dependent on persistence of virus in circulation, while antibodies to MPER are probably not.
Collapse
|
186
|
Mechanism of neutralization by the broadly neutralizing HIV-1 monoclonal antibody VRC01. J Virol 2011; 85:8954-67. [PMID: 21715490 DOI: 10.1128/jvi.00754-11] [Citation(s) in RCA: 190] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The structure of VRC01 in complex with the HIV-1 gp120 core reveals that this broadly neutralizing CD4 binding site (CD4bs) antibody partially mimics the interaction of the primary virus receptor, CD4, with gp120. Here, we extended the investigation of the VRC01-gp120 core interaction to the biologically relevant viral spike to better understand the mechanism of VRC01-mediated neutralization and to define viral elements associated with neutralization resistance. In contrast to the interaction of CD4 or the CD4bs monoclonal antibody (MAb) b12 with the HIV-1 envelope glycoprotein (Env), occlusion of the VRC01 epitope by quaternary constraints was not a major factor limiting neutralization. Mutagenesis studies indicated that VRC01 contacts within the gp120 loop D, the CD4 binding loop, and the V5 region were necessary for optimal VRC01 neutralization, as suggested by the crystal structure. In contrast to interactions with the soluble gp120 monomer, VRC01 interaction with the native viral spike did not occur in a CD4-like manner; VRC01 did not induce gp120 shedding from the Env spike or enhance gp41 membrane proximal external region (MPER)-directed antibody binding to the Env spike. Finally, VRC01 did not display significant reactivity with human antigens, boding well for potential in vivo applications. The data indicate that VRC01 interacts with gp120 in the context of the functional spike in a manner distinct from that of CD4. It achieves potent neutralization by precisely targeting the CD4bs without requiring alterations of Env spike configuration and by avoiding steric constraints imposed by the quaternary structure of the functional Env spike.
Collapse
|
187
|
Girard MP, Osmanov S, Assossou OM, Kieny MP. Human immunodeficiency virus (HIV) immunopathogenesis and vaccine development: a review. Vaccine 2011; 29:6191-218. [PMID: 21718747 DOI: 10.1016/j.vaccine.2011.06.085] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2011] [Revised: 06/20/2011] [Accepted: 06/22/2011] [Indexed: 02/08/2023]
Abstract
The development of a safe, effective and globally affordable HIV vaccine offers the best hope for the future control of the HIV-1 pandemic. Since 1987, scores of candidate HIV-1 vaccines have been developed which elicited varying degrees of protective responses in nonhuman primate models, including DNA vaccines, subunit vaccines, live vectored recombinant vaccines and various prime-boost combinations. Four of these candidate vaccines have been tested for efficacy in human volunteers, but, to the exception of the recent RV144 Phase III trial in Thailand, which elicited a modest but statistically significant level of protection against infection, none has shown efficacy in preventing HIV-1 infection or in controlling virus replication and delaying progression of disease in humans. Protection against infection was observed in the RV144 trial, but intensive research is needed to try to understand the protective immune mechanisms at stake. Building-up on the results of the RV144 trial and deciphering what possibly are the immune correlates of protection are the top research priorities of the moment, which will certainly accelerate the development of an highly effective vaccine that could be used in conjunction with other HIV prevention and treatment strategies. This article reviews the state of the art of HIV vaccine development and discusses the formidable scientific challenges met in this endeavor, in the context of a better understanding of the immunopathogenesis of the disease.
Collapse
Affiliation(s)
- Marc P Girard
- University Paris 7, French National Academy of Medicine, 39 rue Seignemartin, FR 69008 Lyon, France.
| | | | | | | |
Collapse
|
188
|
Trimeric HIV-1 glycoprotein gp140 immunogens and native HIV-1 envelope glycoproteins display the same closed and open quaternary molecular architectures. Proc Natl Acad Sci U S A 2011; 108:11440-5. [PMID: 21709254 DOI: 10.1073/pnas.1101414108] [Citation(s) in RCA: 136] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The initial step in HIV-1 infection occurs with the binding of cell surface CD4 to trimeric HIV-1 envelope glycoproteins (Env), a heterodimer of a transmembrane glycoprotein (gp41) and a surface glycoprotein (gp120). The design of soluble versions of trimeric Env that display structural and functional properties similar to those observed on intact viruses is highly desirable from the viewpoint of designing immunogens that could be effective as vaccines against HIV/AIDS. Using cryoelectron tomography combined with subvolume averaging, we have analyzed the structure of SOSIP gp140 trimers, which are cleaved, solubilized versions of the ectodomain of trimeric HIV-1 Env. We show that unliganded gp140 trimers adopt a quaternary arrangement similar to that displayed by native unliganded trimers on the surface of intact HIV-1 virions. When complexed with soluble CD4, Fab 17b, which binds to gp120 at its chemokine coreceptor binding site, or both soluble CD4 and 17b Fab, gp140 trimers display an open conformation in which there is an outward rotation and displacement of each gp120 protomer. We demonstrate that the molecular arrangements of gp120 trimers in the closed and open conformations of the soluble trimer are the same as those observed for the closed and open states, respectively, of trimeric gp120 on intact HIV-1 BaL virions, establishing that soluble gp140 trimers can be designed to mimic the quaternary structural transitions displayed by native trimeric Env.
Collapse
|
189
|
Dimitrov JD, Kazatchkine MD, Kaveri SV, Lacroix-Desmazes S. "Rational vaccine design" for HIV should take into account the adaptive potential of polyreactive antibodies. PLoS Pathog 2011; 7:e1002095. [PMID: 21698229 PMCID: PMC3116824 DOI: 10.1371/journal.ppat.1002095] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Affiliation(s)
- Jordan D. Dimitrov
- INSERM U872, Paris, France
- Centre de Recherche des Cordeliers, Paris, France
- Université Pierre et Marie Curie-Paris6, UMR S 872, Paris, France
- * E-mail: (JDD); (SLD)
| | - Michel D. Kazatchkine
- The Global Fund to Fight AIDS, Tuberculosis and Malaria, WHO, Vernier – Geneva, Switzerland
| | - Srinivas V. Kaveri
- INSERM U872, Paris, France
- Centre de Recherche des Cordeliers, Paris, France
- Université Pierre et Marie Curie-Paris6, UMR S 872, Paris, France
| | - Sebastien Lacroix-Desmazes
- INSERM U872, Paris, France
- Centre de Recherche des Cordeliers, Paris, France
- Université Pierre et Marie Curie-Paris6, UMR S 872, Paris, France
- * E-mail: (JDD); (SLD)
| |
Collapse
|
190
|
Vanderford TH, Bleckwehl C, Engram JC, Dunham RM, Klatt NR, Feinberg MB, Garber DA, Betts MR, Silvestri G. Viral CTL escape mutants are generated in lymph nodes and subsequently become fixed in plasma and rectal mucosa during acute SIV infection of macaques. PLoS Pathog 2011; 7:e1002048. [PMID: 21625590 PMCID: PMC3098234 DOI: 10.1371/journal.ppat.1002048] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2010] [Accepted: 03/11/2011] [Indexed: 11/18/2022] Open
Abstract
SIVmac239 infection of rhesus macaques (RMs) results in AIDS despite the generation of a strong antiviral cytotoxic T lymphocyte (CTL) response, possibly due to the emergence of viral escape mutants that prevent recognition of infected cells by CTLs. To determine the anatomic origin of these SIV mutants, we longitudinally assessed the presence of CTL escape variants in two MamuA*01-restricted immunodominant epitopes (Tat-SL8 and Gag-CM9) in the plasma, PBMCs, lymph nodes (LN), and rectal biopsies (RB) of fifteen SIVmac239-infected RMs. As expected, Gag-CM9 did not exhibit signs of escape before day 84 post infection. In contrast, Tat-SL8 escape mutants were apparent in all tissues by day 14 post infection. Interestingly LNs and plasma exhibited the highest level of escape at day 14 and day 28 post infection, respectively, with the rate of escape in the RB remaining lower throughout the acute infection. The possibility that CTL escape occurs in LNs before RBs is confirmed by the observation that the specific mutants found at high frequency in LNs at day 14 post infection became dominant at day 28 post infection in plasma, PBMC, and RB. Finally, the frequency of escape mutants in plasma at day 28 post infection correlated strongly with the level Tat-SL8-specific CD8 T cells in the LN and PBMC at day 14 post infection. These results indicate that LNs represent the primary source of CTL escape mutants during the acute phase of SIVmac239 infection, suggesting that LNs are the main anatomic sites of virus replication and/or the tissues in which CTL pressure is most effective in selecting SIV escape variants. Strong antiviral CD8+ T lymphocytes limit SIV replication by recognizing short pathogen-derived peptide epitopes. The cytotoxic CD8+ T cell responses specific for this highly mutable virus can select for viruses bearing mutations that prevent CD8+ T cell recognition of cells infected with these escape mutants. To determine the anatomic origin of these escape mutants, we tracked a particular escape mutant in multiple tissues (plasma virus, lymph nodes, rectal mucosa, and peripheral blood immune cells) during the early, acute phase of SIVmac239 infection of rhesus macaques. We found that escape mutants first reach high frequency in lymph nodes 2 weeks after infection, and the particular mutants generated in lymph nodes disseminate to other tissues by week 4. Furthermore, we found that epitope-specific CD8+ T lymphocyte responses in the lymph nodes and peripheral blood, but not the gut mucosa, are significantly correlated with the frequency of escape mutants in the plasma virus at week 4. This suggests that lymph nodes, and not the gut, are the primary site of anti-SIV CD8+ T cell responses and/or SIV replication during the acute phase of infection.
Collapse
Affiliation(s)
- Thomas H. Vanderford
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Chelsea Bleckwehl
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Jessica C. Engram
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Richard M. Dunham
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Nichole R. Klatt
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Mark B. Feinberg
- Merck Vaccines and Infectious Diseases, Merck and Co., Inc., West Point, Pennsylvania, United States of America
| | - David A. Garber
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Michael R. Betts
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Guido Silvestri
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
191
|
Uchtenhagen H, Friemann R, Raszewski G, Spetz AL, Nilsson L, Achour A. Crystal structure of the HIV-2 neutralizing Fab fragment 7C8 with high specificity to the V3 region of gp125. PLoS One 2011; 6:e18767. [PMID: 21541316 PMCID: PMC3082531 DOI: 10.1371/journal.pone.0018767] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2010] [Accepted: 03/18/2011] [Indexed: 02/05/2023] Open
Abstract
7C8 is a mouse monoclonal antibody specific for the third hypervariable region (V3) of the human immunodeficiency virus type 2 (HIV-2)-associated protein gp125. The three-dimensional crystal structure of the Fab fragment of 7C8, determined to 2.7 Å resolution, reveals a deep and narrow antigen-binding cleft with architecture appropriate for an elongated epitope. The highly hydrophobic cleft is bordered on one side by the negatively charged second complementarity determining region (CDR2) and the unusually long positively charged CDR3 of the heavy chain and, on the other side, by the CDR1 of the light chain. Analysis of 7C8 in complex with molecular models of monomeric and trimeric gp125 highlights the importance of a conserved stretch of residues FHSQ that is localized centrally on the V3 region of gp125. Furthermore, modeling also indicates that the Fab fragment neutralizes the virus by sterically impairing subsequent engagement of the gp125 trimer with the co-receptor on the target cell.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antibodies, Monoclonal/chemistry
- Antibodies, Monoclonal/immunology
- Antibodies, Neutralizing/chemistry
- Antibodies, Neutralizing/immunology
- Antibody Specificity/immunology
- Binding Sites
- Complementarity Determining Regions/chemistry
- Complementarity Determining Regions/immunology
- Computer Simulation
- Crystallography, X-Ray
- Epitopes/chemistry
- Epitopes/immunology
- HIV-2/immunology
- Humans
- Hydrophobic and Hydrophilic Interactions
- Immunoglobulin Fab Fragments/chemistry
- Immunoglobulin Fab Fragments/immunology
- Mice
- Models, Molecular
- Molecular Sequence Data
- Protein Multimerization
- Protein Structure, Secondary
- Protein Structure, Tertiary
- env Gene Products, Human Immunodeficiency Virus/chemistry
- env Gene Products, Human Immunodeficiency Virus/immunology
Collapse
Affiliation(s)
- Hannes Uchtenhagen
- F59 Department of Medicine, Center for Infectious Medicine (CIM), Karolinska University Hospital Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Rosmarie Friemann
- Department of Cell and Molecular Biology, Molecular Biophysics, Biomedical Center, Uppsala University, Uppsala, Sweden
| | - Grzegorz Raszewski
- Department of Biosciences and Nutrition, Center for Biosciences, Karolinska Institutet, Huddinge, Sweden
| | - Anna-Lena Spetz
- F59 Department of Medicine, Center for Infectious Medicine (CIM), Karolinska University Hospital Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Lennart Nilsson
- Department of Biosciences and Nutrition, Center for Biosciences, Karolinska Institutet, Huddinge, Sweden
| | - Adnane Achour
- F59 Department of Medicine, Center for Infectious Medicine (CIM), Karolinska University Hospital Huddinge, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
192
|
Schellinger JG, Danan-Leon LM, Hoch JA, Kassa A, Srivastava I, Davis D, Gervay-Hague J. Synthesis of a trimeric gp120 epitope mimic conjugated to a T-helper peptide to improve antigenicity. J Am Chem Soc 2011; 133:3230-3. [PMID: 21341746 DOI: 10.1021/ja1083915] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
A fully synthetic trivalent mimotope of gp120 conjugated to pan allelic HLA DR binding epitope was prepared using solid-phase peptide synthesis and optimized copper-catalyzed azide-alkyne cycloaddition. The methodology efficiently provides chemically uniform heteromultimeric peptide constructs with enhanced binding, avidity, and specificity toward an established HIV-neutralizing human antibody, MAb b12. The versatile synthetic strategy serves as a powerful platform for the development of synthetic peptides as potential HIV-1 vaccine candidates.
Collapse
Affiliation(s)
- Joan G Schellinger
- Chemistry Department, Campus Mass Spectrometry Facility, University of California at Davis, 1 Shields Avenue, Davis, California 95616, United States
| | | | | | | | | | | | | |
Collapse
|
193
|
Mattiacio J, Walter S, Brewer M, Domm W, Friedman AE, Dewhurst S. Dense display of HIV-1 envelope spikes on the lambda phage scaffold does not result in the generation of improved antibody responses to HIV-1 Env. Vaccine 2011; 29:2637-47. [PMID: 21310193 DOI: 10.1016/j.vaccine.2011.01.038] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2010] [Revised: 01/01/2011] [Accepted: 01/09/2011] [Indexed: 10/18/2022]
Abstract
The generation of strong, virus-neutralizing antibody responses to the HIV-1 envelope spike (Env) is a major goal in HIV-1 vaccine research. To try to enhance the Env-specific response, we displayed oligomeric gp140 on a virus-like scaffold provided by the lambda phage capsid. To do this, an in vitro complementation system was used to "decorate" phage particles with glycosylated, mammalian cell-derived envelope oligomers. We compared the immune response to lambda phage particles displaying HIV-1 Env to that elicited by soluble oligomeric gp140 in rabbits. Env-binding antibody titers were higher in animals that received oligomeric gp140 as compared to Env decorated phage particles, as were virus neutralizing antibody responses. The Env decorated phage particles were, however, able to efficiently boost a protein-primed humoral response to levels equivalent to those elicited by high-dose adjuvanted Env oligomers. These results show that display of HIV-1 envelope spikes on the bacteriophage lambda capsid does not result in an improved, Env-specific humoral immune response.
Collapse
Affiliation(s)
- Jonelle Mattiacio
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | | | | | | | | | | |
Collapse
|
194
|
Paradoxical suppression of poly-specific broadly neutralizing antibodies in the presence of strain-specific neutralizing antibodies following HIV infection. J Theor Biol 2011; 277:55-66. [PMID: 21315731 DOI: 10.1016/j.jtbi.2011.01.050] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2010] [Revised: 01/17/2011] [Accepted: 01/31/2011] [Indexed: 11/21/2022]
Abstract
One of the first immunologic responses against HIV infection is the presence of neutralizing antibodies that seem able to inactivate several HIV strains. Moreover, in vitro studies have shown the existence of monoclonal antibodies that exhibit broad crossclade neutralizing potential. Yet their number is low and slow to develop in vivo. In this paper, we investigate the potential benefits of inducing poly-specific neutralizing antibodies in vivo throughout immunization. We develop a mathematical model that considers the activation of families of B lymphocytes producing poly-specific and strain-specific antibodies and use it to demonstrate that, even if such families are successful in producing neutralizing antibodies, the competition between them may limit the poly-specific response allowing the virus to escape. We modify this model to account for viral evolution under the pressure of antibody responses in natural HIV infection. The model can reproduce viral escape under certain conditions of B lymphocyte competition. Using these models we provide explanations for the observed antibody failure in controlling natural infection and predict quantitative measures that need to be satisfied for long-term control of HIV infection.
Collapse
|
195
|
Cicala C, Arthos J, Fauci AS. HIV-1 envelope, integrins and co-receptor use in mucosal transmission of HIV. J Transl Med 2011; 9 Suppl 1:S2. [PMID: 21284901 PMCID: PMC3105502 DOI: 10.1186/1479-5876-9-s1-s2] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
It is well established that HIV-1 infection typically involves an interaction between the viral envelope protein gp120/41 and the CD4 molecule followed by a second interaction with a chemokine receptor, usually CCR5 or CXCR4. In the early stages of an HIV-1 infection CCR5 using viruses (R5 viruses) predominate. In some viral subtypes there is a propensity to switch to CXCR4 usage (X4 viruses). The receptor switch occurs in ~ 40% of the infected individuals and is associated with faster disease progression. This holds for subtypes B and D, but occurs less frequently in subtypes A and C. There are several hypotheses to explain the preferential transmission of R5 viruses and the mechanisms that lead to switching of co-receptor usage; however, there is no definitive explanation for either. One important consideration regarding transmission is that signaling by R5 gp120 may facilitate transmission of R5 viruses by inducing a permissive environment for HIV replication. In the case of sexual transmission, infection by HIV requires the virus to breach the mucosal barrier to gain access to the immune cell targets that it infects; however, the immediate events that follow HIV exposure at genital mucosal sites are not well understood. Upon transmission, the HIV quasispecies that is replicating in an infected donor contracts through a “genetic bottleneck”, and often infection results from a single infectious event. Many details surrounding this initial infection remain unresolved. In mucosal tissues, CD4+ T cells express high levels of CCR5, and a subset of these CD4+/CCR5high cells express the integrin α4β7, the gut homing receptor. CD4+/CCR5high/ α4β7high T cells are highly susceptible to infection by HIV-1 and are ideal targets for an efficient productive infection at the point of transmission. In this context we have demonstrated that the HIV-1 envelope protein gp120 binds to α4β7 on CD4+ T cells. On CD4+/CCR5high/ α4β7high T cells, α4β7 is closely associated with CD4 and CCR5. Furthermore, α4β7 is ~3 times the size of CD4 on the cell surface, that makes it a prominent receptor for an efficient virus capture. gp120-α4β7 interactions mediate the activation of the adhesion-associated integrin LFA-1. LFA-1 facilitates the formation of virological synapses and cell-to-cell spread of HIV-1. gp120 binding to α4β7 is mediated by a tripeptide located in the V1/V2 domain of gp120. Of note, the V1/V2 domain of gp120 has been linked to variations in transmission fitness among viral isolates raising the intriguing possibility that gp120-α4β7 interactions may be linked to transmission fitness. Although many details remain unresolved, we hypothesize that gp120-α4β7 interactions play an important role in the very early events following sexual transmission of HIV and may have important implication in the design of vaccine strategies for the prevention of acquisition of HIV infection
Collapse
Affiliation(s)
- Claudia Cicala
- Laboratory of Immunoregulation National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | |
Collapse
|
196
|
Abstract
PURPOSE OF REVIEW Recent clinical trial results have indicated that it may be possible for vaccines to induce protection against HIV. To build on this result, strategies should be designed to enhance duration, breadth, and magnitude of antibody production. Strategic formulation of agonists of the innate immune system and carriers that selectively present the target antigen yields a class of pharmaceuticals, named 'adjuvants', that greatly influence immunity resulting from vaccination. As researchers begin to focus not only on creating an immune response to an antigen, but also on the quality of that response, the role of adjuvants is becoming increasingly significant. This review is intended to give an overview of recent findings on how adjuvants model the immune response to antigens with a focus on the field of vaccines for HIV. RECENT FINDINGS It is clear that innate and adaptive immunity are linked by communication channels that allow innate signals to influence the quality of adaptive responses as well as adaptive signals that temper innate responses. Adjuvants take advantage of this bridge to shape the immune response to antigens. In this review, we will discuss the different classes of adjuvants currently available; recent findings on the relationship between adjuvants and the type of immune profile generated; and the breadth of neutralizing antibodies as influenced by adjuvants. SUMMARY Because adjuvants influence the breadth of antibodies generated and the type of cells that proliferate in response to a vaccine this review is relevant for scientists clinicians involved in creating a new HIV vaccine.
Collapse
|
197
|
Effect of B-cell depletion on coreceptor switching in R5 simian-human immunodeficiency virus infection of rhesus macaques. J Virol 2011; 85:3086-94. [PMID: 21248033 DOI: 10.1128/jvi.02150-10] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We recently described a coreceptor switch in rapid progressor (RP) R5 simian-human immunodeficiency virus SF162P3N (SHIV(SF162P3N))-infected rhesus macaques that had high virus replication and undetectable or weak and transient antiviral antibody response (S. H. Ho et al., J. Virol. 81:8621-8633, 2007; S. H. Ho, N. Trunova, A. Gettie, J. Blanchard, and C. Cheng-Mayer, J. Virol. 82:5653-5656, 2008; and W. Ren et al., J. Virol. 84:340-351, 2010). The lack of antibody selective pressure, together with the observation that the emerging X4 variants were neutralization sensitive, suggested that the absence or weakening of the virus-specific humoral immune response could be an environmental factor fostering coreceptor switching in vivo. To test this possibility, we treated four macaques with 50 mg/kg of body weight of the anti-CD20 antibody rituximab every 2 to 3 weeks starting from the week prior to intravenous infection with SHIV(SF162P3N) for a total of six infusions. Rituximab treatment successfully depleted peripheral and lymphoid CD20(+) cells for up to 25 weeks according to flow cytometry and immunohistochemical staining, with partial to full recovery in two of the four treated monkeys thereafter. Three of the four treated macaques failed to mount a detectable anti-SHIV antibody response, while the response was delayed in the remaining animal. The three seronegative macaques progressed to disease, but in none of them could the presence of X4 variants be demonstrated by V3 sequence and tropism analyses. Furthermore, viruses did not evolve early in these diseased macaques to be more soluble CD4 sensitive. These results demonstrate that the absence or diminution of humoral immune responses by itself is insufficient to drive the R5-to-X4 switch and the neutralization susceptibility of the evolving viruses.
Collapse
|
198
|
Abstract
We have used cryoelectron tomography of vitreous-ice-embedded HIV-1 virions to compare the envelope (Env) spikes of a wild-type strain with those of a mutant strain in which the V1/V2 loop has been deleted. Deletion of V1/V2 results in a spike with far more structural heterogeneity than is observed in the wild type, likely reflecting greatly enhanced gp120 protomer flexibility. A major difference between the two forms is a pronounced loss of mass from the "peak" of the native Env spike. The apparent loss of contact among three gp120 protomers likely accounts for the more open structure, heterogeneity in configuration, and previous observations that broadly neutralizing epitopes and reactive sites on other structural elements are more exposed in such constructs.
Collapse
|
199
|
Swetnam J, Shmelkov E, Zolla-Pazner S, Cardozo T. Comparative magnitude of cross-strain conservation of HIV variable loop neutralization epitopes. PLoS One 2010; 5:e15994. [PMID: 21209919 PMCID: PMC3012121 DOI: 10.1371/journal.pone.0015994] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2010] [Accepted: 12/02/2010] [Indexed: 01/17/2023] Open
Abstract
Although the sequence variable loops of the human immunodeficiency virus' (HIV-1) surface envelope glycoprotein (gp120) can exhibit good immunogenicity, characterizing conserved (invariant) cross-strain neutralization epitopes within these loops has proven difficult. We recently developed a method to derive sensitive and specific signature motifs for the three-dimensional (3D) shapes of the HIV-1 neutralization epitopes in the third variable (V3) loop of gp120 that are recognized by human monoclonal antibodies (mAbs). We used the signature motif method to estimate the conservation of these epitopes across circulating worldwide HIV-1 strains. The epitope targeted by the anti-V3 loop neutralizing mAb 3074 is present in 87% of circulating strains, distributed nearly evenly among all subtypes. The results for other anti-V3 Abs are: 3791, present in 63% of primarily non-B subtypes; 2219, present in 56% of strains across all subtypes; 2557, present in 52% across all subtypes; 447-52D, present in 11% of primarily subtype B strains; 537-10D, present in 9% of primarily subtype B strains; and 268-D, present in 5% of primarily subtype B strains. The estimates correlate with in vitro tests of these mAbs against diverse viral panels. The mAb 3074 thus targets an epitope that is nearly completely conserved among circulating HIV-1 strains, demonstrating the presence of an invariant structure hidden in the dynamic and sequence-variable V3 loop in gp120. Since some variable loop regions are naturally immunogenic, designing immunogens to mimic their conserved epitopes may be a promising vaccine discovery approach. Our results suggest one way to quantify and compare the magnitude of the conservation.
Collapse
Affiliation(s)
- James Swetnam
- Department of Pharmacology, New York University School of Medicine, New York, New York, United States of America
| | - Evgeny Shmelkov
- Department of Pharmacology, New York University School of Medicine, New York, New York, United States of America
| | - Susan Zolla-Pazner
- Department of Pathology, New York University School of Medicine, New York, New York, United States of America
- New York Veterans Affairs Medical Center, New York, New York, United States of America
| | - Timothy Cardozo
- Department of Pharmacology, New York University School of Medicine, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
200
|
Nara PL, Tobin GJ, Chaudhuri AR, Trujillo JD, Lin G, Cho MW, Levin SA, Ndifon W, Wingreen NS. How can vaccines against influenza and other viral diseases be made more effective? PLoS Biol 2010; 8:e1000571. [PMID: 21203586 PMCID: PMC3006352 DOI: 10.1371/journal.pbio.1000571] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Affiliation(s)
- Peter L. Nara
- Biological Mimetics, Inc., Frederick, Maryland, United States of America
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, Iowa, United States of America
| | - Gregory J. Tobin
- Biological Mimetics, Inc., Frederick, Maryland, United States of America
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, Iowa, United States of America
| | - A. Ray Chaudhuri
- Biological Mimetics, Inc., Frederick, Maryland, United States of America
| | - Jessie D. Trujillo
- Department of Veterinary Preventive Medicine and Microbiology, Iowa State University, Ames, Iowa, United States of America
| | - George Lin
- Biological Mimetics, Inc., Frederick, Maryland, United States of America
| | - Michael W. Cho
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, Iowa, United States of America
| | - Simon A. Levin
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, New Jersey, United States of America
- * E-mail:
| | - Wilfred Ndifon
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| | - Ned S. Wingreen
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
| |
Collapse
|