1
|
Symmonds J, Gaufin T, Xu C, Raehtz KD, Ribeiro RM, Pandrea I, Apetrei C. Making a Monkey out of Human Immunodeficiency Virus/Simian Immunodeficiency Virus Pathogenesis: Immune Cell Depletion Experiments as a Tool to Understand the Immune Correlates of Protection and Pathogenicity in HIV Infection. Viruses 2024; 16:972. [PMID: 38932264 PMCID: PMC11209256 DOI: 10.3390/v16060972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 05/31/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
Understanding the underlying mechanisms of HIV pathogenesis is critical for designing successful HIV vaccines and cure strategies. However, achieving this goal is complicated by the virus's direct interactions with immune cells, the induction of persistent reservoirs in the immune system cells, and multiple strategies developed by the virus for immune evasion. Meanwhile, HIV and SIV infections induce a pandysfunction of the immune cell populations, making it difficult to untangle the various concurrent mechanisms of HIV pathogenesis. Over the years, one of the most successful approaches for dissecting the immune correlates of protection in HIV/SIV infection has been the in vivo depletion of various immune cell populations and assessment of the impact of these depletions on the outcome of infection in non-human primate models. Here, we present a detailed analysis of the strategies and results of manipulating SIV pathogenesis through in vivo depletions of key immune cells populations. Although each of these methods has its limitations, they have all contributed to our understanding of key pathogenic pathways in HIV/SIV infection.
Collapse
Affiliation(s)
- Jen Symmonds
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA; (J.S.); (C.X.); (K.D.R.); (I.P.)
- Department of Infectious Diseases and Microbiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Thaidra Gaufin
- Tulane National Primate Research Center, Tulane University, Covington, LA 70433, USA;
| | - Cuiling Xu
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA; (J.S.); (C.X.); (K.D.R.); (I.P.)
- Division of Infectious Diseases, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Kevin D. Raehtz
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA; (J.S.); (C.X.); (K.D.R.); (I.P.)
- Division of Infectious Diseases, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Ruy M. Ribeiro
- Theoretical Biology and Biophysics Group, Los Alamos National Laboratory, Los Alamos, NM 87545, USA
| | - Ivona Pandrea
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA; (J.S.); (C.X.); (K.D.R.); (I.P.)
- Department of Infectious Diseases and Microbiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Cristian Apetrei
- Department of Infectious Diseases and Microbiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Division of Infectious Diseases, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|
2
|
Laliberté A, Prelli Bozzo C, Stahl-Hennig C, Hunszinger V, Joas S, Sauermann U, Roshani B, Klippert A, Daskalaki M, Mätz-Rensing K, Stolte-Leeb N, Tharp GK, Fuchs D, Gupta PM, Silvestri G, Nelson SA, Parodi L, Giavedoni L, Bosinger SE, Sparrer KM, Kirchhoff F. Vpr attenuates antiviral immune responses and is critical for full pathogenicity of SIV mac239 in rhesus macaques. iScience 2023; 26:108351. [PMID: 38025783 PMCID: PMC10679897 DOI: 10.1016/j.isci.2023.108351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 09/05/2023] [Accepted: 10/24/2023] [Indexed: 12/01/2023] Open
Abstract
The accessory viral protein R (Vpr) is encoded by all primate lentiviruses. Vpr counteracts DNA repair pathways, modulates viral immune sensing, and induces cell-cycle arrest in cell culture. However, its impact in vivo is controversial. Here, we show that deletion of vpr is associated with delayed viral replication kinetics, rapid innate immune activation, development and maintenance of strong B and T cell responses, and increased neutralizing activity against SIVmac239 in rhesus macaques. All wild-type SIVmac239-infected animals maintained high viral loads, and five of six developed fatal immunodeficiency during ∼80 weeks of follow-up. Lack of Vpr was associated with better preservation of CD4+ T cells, lower viral loads, and an attenuated clinical course of infection in most animals. Our results show that Vpr contributes to efficient viral immune evasion and the full pathogenic potential of SIVmacin vivo. Inhibition of Vpr may improve humoral immune control of viral replication.
Collapse
Affiliation(s)
- Alexandre Laliberté
- Institute of Molecular Virology – Ulm University Medical Center, Meyerhofstraße 1, 89081 Ulm, Germany
| | - Caterina Prelli Bozzo
- Institute of Molecular Virology – Ulm University Medical Center, Meyerhofstraße 1, 89081 Ulm, Germany
| | | | - Victoria Hunszinger
- Institute of Molecular Virology – Ulm University Medical Center, Meyerhofstraße 1, 89081 Ulm, Germany
| | - Simone Joas
- Institute of Molecular Virology – Ulm University Medical Center, Meyerhofstraße 1, 89081 Ulm, Germany
| | | | - Berit Roshani
- German Primate Center, Kellnerweg 4, 37077 Göttingen, Germany
| | | | - Maria Daskalaki
- German Primate Center, Kellnerweg 4, 37077 Göttingen, Germany
| | | | | | - Gregory K. Tharp
- Emory National Primate Research Center, Emory Vaccine Center and Department of Pathology & Laboratory Medicine, Emory University, Atlanta, GA, USA
| | - Dietmar Fuchs
- German Primate Center, Kellnerweg 4, 37077 Göttingen, Germany
| | - Prachi Mehrotra Gupta
- Emory National Primate Research Center, Emory Vaccine Center and Department of Pathology & Laboratory Medicine, Emory University, Atlanta, GA, USA
| | - Guido Silvestri
- Emory National Primate Research Center, Emory Vaccine Center and Department of Pathology & Laboratory Medicine, Emory University, Atlanta, GA, USA
| | - Sydney A. Nelson
- Emory National Primate Research Center, Emory Vaccine Center and Department of Pathology & Laboratory Medicine, Emory University, Atlanta, GA, USA
| | - Laura Parodi
- Host-Pathogen Interactions Program, Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Luis Giavedoni
- Host-Pathogen Interactions Program, Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Steven E. Bosinger
- Emory National Primate Research Center, Emory Vaccine Center and Department of Pathology & Laboratory Medicine, Emory University, Atlanta, GA, USA
| | - Konstantin M.J. Sparrer
- Institute of Molecular Virology – Ulm University Medical Center, Meyerhofstraße 1, 89081 Ulm, Germany
| | - Frank Kirchhoff
- Institute of Molecular Virology – Ulm University Medical Center, Meyerhofstraße 1, 89081 Ulm, Germany
| |
Collapse
|
3
|
Zhao J, Song E, Huang Y, Yu A, Mechref Y. Variability in the Glycosylation Patterns of gp120 Proteins from Different Human Immunodeficiency Virus Type 1 Isolates Expressed in Different Host Cells. J Proteome Res 2021; 20:4862-4874. [PMID: 34448591 DOI: 10.1021/acs.jproteome.1c00587] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The mature HIV-1 envelope (Env) glycoprotein is composed of gp120, the exterior subunit, and gp41, the transmembrane subunit assembled as trimer by noncovalent interaction. There is a great body of literature to prove that gp120 binds to CD4 first, then to the coreceptor. Binding experiments and functional assays have demonstrated that CD4 binding induces conformational changes in gp120 that enable or enhance its interaction with a coreceptor. Previous studies provided different glycomic maps for the HIV-1 gp120. Here, we build on previous work to report that the use of LC-MS/MS, in conjunction with hydrophilic interaction liquid chromatography (HILIC) enrichment to glycosylation sites, associated with the assorted neutralizing or binding events of glycosylation targeted antibodies from different clades or strains. In this study, the microheterogeneity of the glycosylation from 4 different clades of gp120s is deeply investigated. Aberrant glycosylation patterns were detected on gp120 that originated from different clades, viral sequences, and host cells. The results of this study may help provide a better understanding of the mechanism of how the glycans participate in the antibody neutralizing process that targets glycosylation sites.
Collapse
Affiliation(s)
- Jingfu Zhao
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, Texas 79409, United States
| | - Ehwang Song
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, Texas 79409, United States
| | - Yifan Huang
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, Texas 79409, United States
| | - Aiying Yu
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, Texas 79409, United States
| | - Yehia Mechref
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, Texas 79409, United States
| |
Collapse
|
4
|
Swanstrom AE, Del Prete GQ, Deleage C, Elser SE, Lackner AA, Hoxie JA. The SIV Envelope Glycoprotein, Viral Tropism, and Pathogenesis: Novel Insights from Nonhuman Primate Models of AIDS. Curr HIV Res 2019; 16:29-40. [PMID: 29173176 DOI: 10.2174/1570162x15666171124123116] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 10/12/2017] [Accepted: 10/13/2017] [Indexed: 12/14/2022]
Abstract
BACKGROUND Cellular tropism of human immunodeficiency virus (HIV-1) is closely linked to interactions between the viral envelope glycoprotein (Env) with CD4 and chemokine receptor family members, CCR5 and CXCR4. This interaction plays a key role in determining anatomic sites that are infected in vivo and the cascade of early and late events that result in chronic immune activation, immunosuppression and ultimately, AIDS. CD4+ T cells are critical to adaptive immune responses, and their early and rapid infection in gut lamina propria and secondary lymphoid tissues in susceptible hosts likely contributes to viral persistence and progression to disease. CD4+ macrophages are also infected, although their role in HIV-1 pathogenesis is more controversial. METHODS Pathogenic infection by simian immunodeficiency viruses (SIV) in Asian macaques as models of HIV-1 infection has enabled the impact of cellular tropism on pathogenesis to be directly probed. This review will highlight examples in which experimental interventions during SIV infection or the introduction of viral mutations have altered cellular tropism and, subsequently, pathogenesis. RESULTS Alterations to the interaction of Env and its cellular receptors has been shown to result in changes to CD4 dependence, coreceptor specificity, and viral tropism for gut CD4+ T cells and macrophages. CONCLUSION Collectively, these findings have yielded novel insights into the critical role of the viral Env and tropism as a driver of pathogenesis and host control and have helped to identify new areas for targeted interventions in therapy and prevention of HIV-1 infection.
Collapse
Affiliation(s)
- Adrienne E Swanstrom
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc., Frederick, MD, United States
| | - Gregory Q Del Prete
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc., Frederick, MD, United States
| | - Claire Deleage
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc., Frederick, MD, United States
| | - Samra E Elser
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Andrew A Lackner
- Tulane National Primate Research Center, Covington, LA, United States
| | - James A Hoxie
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| |
Collapse
|
5
|
RAJKHOWA TK, VANLALRUATI C, SINGH YD, RAVINDRAN R, ARYA RS. Genetic variation of highly pathogenic Indian porcine reproductive and respiratory syndrome viruses after introduction in 2013. THE INDIAN JOURNAL OF ANIMAL SCIENCES 2018. [DOI: 10.56093/ijans.v88i10.84071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
To study its possible link to pathogenicity, the genomic variation in full ORF5 and ORF7 genes, and their encoded proteins in 26 field HP-PRRSV isolates from three major HP-PRRS outbreaks occurred in India, since 2013 was analysed. Sequence analysis and phylogenetic tree revealed involvement of genetically different strain in each outbreak of India rather persistence of a single strain. Analysis and comparison of N protein amino acid sequences of HP-PRRSV with VR2332 revealed consistent mutation at position 15D to N or K and 46 K to R in all the HP-PRRSV. GP5 protein showed consistent mutations at 29 positions from that of VR2332. The potential Nglycosylation sites in GP5 was found variable from 4–5 with one additional N-glycan moiety around the neutralizing epitope B. However, the ‘decoy’ epitope A was found highly conserved in all the HP-PRRSV.
Collapse
|
6
|
Martins MA, Tully DC, Pedreño-Lopez N, von Bredow B, Pauthner MG, Shin YC, Yuan M, Lima NS, Bean DJ, Gonzalez-Nieto L, Domingues A, Gutman MJ, Maxwell HS, Magnani DM, Ricciardi MJ, Bailey VK, Altman JD, Burton DR, Ejima K, Allison DB, Evans DT, Rakasz EG, Parks CL, Bonaldo MC, Capuano S, Lifson JD, Desrosiers RC, Allen TM, Watkins DI. Mamu-B*17+ Rhesus Macaques Vaccinated with env, vif, and nef Manifest Early Control of SIVmac239 Replication. J Virol 2018; 92:e00690-18. [PMID: 29875239 PMCID: PMC6069176 DOI: 10.1128/jvi.00690-18] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 05/28/2018] [Indexed: 12/22/2022] Open
Abstract
Certain major histocompatibility complex class I (MHC-I) alleles are associated with spontaneous control of viral replication in human immunodeficiency virus (HIV)-infected people and simian immunodeficiency virus (SIV)-infected rhesus macaques (RMs). These cases of "elite" control of HIV/SIV replication are often immune-mediated, thereby providing a framework for studying anti-lentiviral immunity. In this study, we examined how vaccination impacts SIV replication in RMs expressing the MHC-I allele Mamu-B*17 Approximately 21% of Mamu-B*17+ and 50% of Mamu-B*08+ RMs control chronic-phase viremia after SIVmac239 infection. Because CD8+ T cells targeting Mamu-B*08-restricted SIV epitopes have been implicated in virologic suppression in Mamu-B*08+ RMs, we investigated whether this might also be true for Mamu-B*17+ RMs. Two groups of Mamu-B*17+ RMs were vaccinated with genes encoding Mamu-B*17-restricted epitopes in Vif and Nef. These genes were delivered by themselves (group 1) or together with env (group 2). Group 3 included MHC-I-matched RMs and served as the control group. Surprisingly, the group 1 vaccine regimen had little effect on viral replication compared to group 3, suggesting that unlike Mamu-B*08+ RMs, preexisting SIV-specific CD8+ T cells alone do not facilitate long-term virologic suppression in Mamu-B*17+ RMs. Remarkably, however, 5/8 group 2 vaccinees controlled viremia to <15 viral RNA copies/ml soon after infection. No serological neutralizing activity against SIVmac239 was detected in group 2, although vaccine-elicited gp140-binding antibodies correlated inversely with nadir viral loads. Collectively, these data shed new light on the unique mechanism of elite control in Mamu-B*17+ RMs and implicate vaccine-induced, nonneutralizing anti-Env antibodies in the containment of immunodeficiency virus infection.IMPORTANCE A better understanding of the immune correlates of protection against HIV might facilitate the development of a prophylactic vaccine. Therefore, we investigated simian immunodeficiency virus (SIV) infection outcomes in rhesus macaques expressing the major histocompatibility complex class I allele Mamu-B*17 Approximately 21% of Mamu-B*17+ macaques spontaneously controlled chronic phase viremia after SIV infection, an effect that may involve CD8+ T cells targeting Mamu-B*17-restricted SIV epitopes. We vaccinated Mamu-B*17+ macaques with genes encoding immunodominant epitopes in Vif and Nef alone (group 1) or together with env (group 2). Although neither vaccine regimen prevented SIV infection, 5/8 group 2 vaccinees controlled viremia to below detection limits shortly after infection. This outcome, which was not observed in group 1, was associated with vaccine-induced, nonneutralizing Env-binding antibodies. Together, these findings suggest a limited contribution of Vif- and Nef-specific CD8+ T cells for virologic control in Mamu-B*17+ macaques and implicate anti-Env antibodies in containment of SIV infection.
Collapse
Affiliation(s)
| | - Damien C Tully
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA
| | | | - Benjamin von Bredow
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, Wisconsin, USA
| | - Matthias G Pauthner
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, California, USA
- Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery (CHAVI-ID), The Scripps Research Institute, La Jolla, California, USA
| | - Young C Shin
- Department of Pathology, University of Miami, Miami, Florida, USA
| | - Maoli Yuan
- International AIDS Vaccine Initiative, AIDS Vaccine Design and Development Laboratory, Brooklyn, New York, USA
| | - Noemia S Lima
- Laboratório de Biologia Molecular de Flavivirus, Instituto Oswaldo Cruz-FIOCRUZ, Rio de Janeiro, Brazil
| | - David J Bean
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA
| | | | - Aline Domingues
- Department of Pathology, University of Miami, Miami, Florida, USA
| | - Martin J Gutman
- Department of Pathology, University of Miami, Miami, Florida, USA
| | - Helen S Maxwell
- Department of Pathology, University of Miami, Miami, Florida, USA
| | - Diogo M Magnani
- Department of Pathology, University of Miami, Miami, Florida, USA
| | | | - Varian K Bailey
- Department of Pathology, University of Miami, Miami, Florida, USA
| | - John D Altman
- Department of Microbiology and Immunology, Emory University, Atlanta, Georgia, USA
| | - Dennis R Burton
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, California, USA
- Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery (CHAVI-ID), The Scripps Research Institute, La Jolla, California, USA
| | - Keisuke Ejima
- School of Public Health, Indiana University Bloomington, Bloomington, Indiana, USA
| | - David B Allison
- School of Public Health, Indiana University Bloomington, Bloomington, Indiana, USA
| | - David T Evans
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, Wisconsin, USA
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, Wisconsin, USA
| | - Eva G Rakasz
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, Wisconsin, USA
| | - Christopher L Parks
- International AIDS Vaccine Initiative, AIDS Vaccine Design and Development Laboratory, Brooklyn, New York, USA
| | - Myrna C Bonaldo
- Laboratório de Biologia Molecular de Flavivirus, Instituto Oswaldo Cruz-FIOCRUZ, Rio de Janeiro, Brazil
| | - Saverio Capuano
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, Wisconsin, USA
| | - Jeffrey D Lifson
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | | | - Todd M Allen
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA
| | - David I Watkins
- Department of Pathology, University of Miami, Miami, Florida, USA
| |
Collapse
|
7
|
High-Resolution Sequencing of Viral Populations during Early Simian Immunodeficiency Virus Infection Reveals Evolutionary Strategies for Rapid Escape from Emerging Env-Specific Antibody Responses. J Virol 2018; 92:JVI.01574-17. [PMID: 29343575 DOI: 10.1128/jvi.01574-17] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 01/08/2018] [Indexed: 01/01/2023] Open
Abstract
Primate lentiviruses, including the human and simian immunodeficiency viruses (HIV and SIV), produce infections marked by persistent, ongoing viral replication. This occurs despite the presence of virus-specific adaptive immune responses, including antibodies targeting the viral envelope glycoprotein (Env), and evolution of antibody-escape variants is a well-documented feature of lentiviral infection. Here, we examined the evolutionary dynamics of the SIV env gene during early infection (≤29 weeks postinfection) in a cohort of four SIVmac251-infected rhesus macaques. We tracked env evolution during acute and early infection using frequent sampling and ultradeep sequencing of viral populations, capturing a transmission bottleneck and the subsequent reestablishment of Env diversity. A majority of changes in the gp120 subunit mapped to two short clusters, one in the first variable region (V1) and one in V4, while most changes in the gp41 subunit appeared in the cytoplasmic domain. Variation in V1 was dominated by short duplications and deletions of repetitive sequence, while variation in V4 was marked by short in-frame deletions and closely overlapping substitutions. The most common substitutions in both patches did not alter viral replicative fitness when tested using a highly sensitive, deep-sequencing-based competition assay. Our results, together with the observation that very similar or identical patterns of sequence evolution also occur in different macaque species infected with related but divergent strains of SIV, suggest that resistance to early, strain-specific anti-Env antibodies is the result of temporally and mutationally predictable pathways of escape that occur during the early stages of infection.IMPORTANCE The envelope glycoprotein (Env) of primate lentiviruses mediates entry by binding to host cell receptors followed by fusion of the viral membrane with the cell membrane. The exposure of Env complexes on the surface of the virion results in targeting by antibodies, leading to selection for virus escape mutations. We used the SIV/rhesus macaque model to track in vivo evolution of variation in Env during acute/early infection in animals with and without antibody responses to Env, uncovering remarkable variation in animals with antibody responses within weeks of infection. Using a deep-sequencing-based fitness assay, we found substitutions associated with antibody escape had little to no effect on inherent replicative capacity. The ability to readily propagate advantageous changes that incur little to no replicative fitness costs may be a mechanism to maintain continuous replication under constant immune selection, allowing the virus to persist for months to years in the infected host.
Collapse
|
8
|
Han SY, Antoine A, Howard D, Chang B, Chang WS, Slein M, Deikus G, Kossida S, Duroux P, Lefranc MP, Sebra RP, Smith ML, Fofana IBF. Coupling of Single Molecule, Long Read Sequencing with IMGT/HighV-QUEST Analysis Expedites Identification of SIV gp140-Specific Antibodies from scFv Phage Display Libraries. Front Immunol 2018; 9:329. [PMID: 29545792 PMCID: PMC5837965 DOI: 10.3389/fimmu.2018.00329] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 02/06/2018] [Indexed: 12/20/2022] Open
Abstract
The simian immunodeficiency virus (SIV)/macaque model of human immunodeficiency virus (HIV)/acquired immunodeficiency syndrome pathogenesis is critical for furthering our understanding of the role of antibody responses in the prevention of HIV infection, and will only increase in importance as macaque immunoglobulin (IG) gene databases are expanded. We have previously reported the construction of a phage display library from a SIV-infected rhesus macaque (Macaca mulatta) using oligonucleotide primers based on human IG gene sequences. Our previous screening relied on Sanger sequencing, which was inefficient and generated only a few dozen sequences. Here, we re-analyzed this library using single molecule, real-time (SMRT) sequencing on the Pacific Biosciences (PacBio) platform to generate thousands of highly accurate circular consensus sequencing (CCS) reads corresponding to full length single chain fragment variable. CCS data were then analyzed through the international ImMunoGeneTics information system® (IMGT®)/HighV-QUEST (www.imgt.org) to identify variable genes and perform statistical analyses. Overall the library was very diverse, with 2,569 different IMGT clonotypes called for the 5,238 IGHV sequences assigned to an IMGT clonotype. Within the library, SIV-specific antibodies represented a relatively limited number of clones, with only 135 different IMGT clonotypes called from 4,594 IGHV-assigned sequences. Our data did confirm that the IGHV4 and IGHV3 gene usage was the most abundant within the rhesus antibodies screened, and that these genes were even more enriched among SIV gp140-specific antibodies. Although a broad range of VH CDR3 amino acid (AA) lengths was observed in the unpanned library, the vast majority of SIV gp140-specific antibodies demonstrated a more uniform VH CDR3 length (20 AA). This uniformity was far less apparent when VH CDR3 were classified according to their clonotype (range: 9–25 AA), which we believe is more relevant for specific antibody identification. Only 174 IGKV and 588 IGLV clonotypes were identified within the VL sequences associated with SIV gp140-specific VH. Together, these data strongly suggest that the combination of SMRT sequencing with the IMGT/HighV-QUEST querying tool will facilitate and expedite our understanding of polyclonal antibody responses during SIV infection and may serve to rapidly expand the known scope of macaque V genes utilized during these responses.
Collapse
Affiliation(s)
- Seung Yub Han
- Biology Department, Boston College, Chestnut Hill, MA, United States
| | - Alesia Antoine
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, Icahn Institute of Genomics and Multiscale Biology, New York, NY, United States
| | - David Howard
- Biology Department, Boston College, Chestnut Hill, MA, United States
| | - Bryant Chang
- Biology Department, Boston College, Chestnut Hill, MA, United States
| | - Woo Sung Chang
- Biology Department, Boston College, Chestnut Hill, MA, United States
| | - Matthew Slein
- Biology Department, Boston College, Chestnut Hill, MA, United States
| | - Gintaras Deikus
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, Icahn Institute of Genomics and Multiscale Biology, New York, NY, United States
| | - Sofia Kossida
- The international ImMunoGeneTics information system® (IMGT®), Laboratoire d'ImmunoGénétique Moléculaire (LIGM), Institut de Génétique Humaine (IGH), UMR CNRS, Montpellier University, Montpellier, France
| | - Patrice Duroux
- The international ImMunoGeneTics information system® (IMGT®), Laboratoire d'ImmunoGénétique Moléculaire (LIGM), Institut de Génétique Humaine (IGH), UMR CNRS, Montpellier University, Montpellier, France
| | - Marie-Paule Lefranc
- The international ImMunoGeneTics information system® (IMGT®), Laboratoire d'ImmunoGénétique Moléculaire (LIGM), Institut de Génétique Humaine (IGH), UMR CNRS, Montpellier University, Montpellier, France
| | - Robert P Sebra
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, Icahn Institute of Genomics and Multiscale Biology, New York, NY, United States
| | - Melissa L Smith
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, Icahn Institute of Genomics and Multiscale Biology, New York, NY, United States
| | | |
Collapse
|
9
|
Ita S, Agostinho MR, Sullivan K, Yub Han S, Akleh R, Johnson WE, Fofana IBF. Analysis of SIVmac Envelope-Specific Antibodies Selected Through Phage Display. AIDS Res Hum Retroviruses 2017; 33:869-879. [PMID: 28075174 DOI: 10.1089/aid.2016.0247] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
We have constructed a single chain fragment variable (scFv) phage display library from a simian immunodeficiency virus (SIV)-infected rhesus macaque that developed unusually high-titer neutralizing antibody responses against tier-3, neutralization-resistant SIVmac239. The library was screened using trimeric (gp140) and monomeric (gp120) forms of the SIVmac239 envelope (Env) glycoprotein. We also cloned variable-heavy and variable-light (VH-VL) antibody fragments from seven previously described rhesus macaque B-cell lines (BLCLs) that produce SIV gp120-specific monoclonal antibodies (mAbs). Thirty-two gp140-specific mAbs were selected along with 20 gp120-specific ones. gp120-specific mAbs were only from the VH4 family, while gp41-specific mAbs were primarily from VH1, followed by VH4 and VH3. Rhesus macaque BLCL-derived mAbs belonged primarily to the VH4 family of antibodies followed by VH3 and a smaller number of VH1s. A preferential VH combination with Vλ light chain was observed with phage display-selected SIV Env-specific mAbs (gp120 and gp140), but not with BLCL-derived antibodies or the unpanned library. None of the tested antibodies had detectable neutralizing activity against tier-3 SIVmac239. The majority of gp120-specifc mAbs potently neutralized tier-1 SIVmac316 with 50% inhibitory concentration (IC50) values below 1 μg/ml. For gp140-specific antibodies, which were all specific for the gp41-subunit, 2 out of 11 tested neutralized SIVmac316 (IC50 of 7 and 5 μg/ml, respectively). These data suggest an order of preferential VH segment usage for SIV-specific antibodies in rhesus macaques. These antibodies will be useful in assessing the contribution of non-neutralizing antibodies to inhibition of SIV infection in vitro and in vivo.
Collapse
Affiliation(s)
- Sergio Ita
- Biology Department, Boston College, Chestnut Hill, Massachusetts
- Virology Program, Harvard Medical School, Boston, Massachusetts
| | - Mayara R. Agostinho
- Biology Department, Boston College, Chestnut Hill, Massachusetts
- Brazil Scientific Mobility Program, College of Nursing, University of New Mexico, Albuquerque, New Mexico
| | | | - Seung Yub Han
- Biology Department, Boston College, Chestnut Hill, Massachusetts
| | - Rana Akleh
- Biology Department, Boston College, Chestnut Hill, Massachusetts
| | | | | |
Collapse
|
10
|
Martins MA, Shin YC, Gonzalez-Nieto L, Domingues A, Gutman MJ, Maxwell HS, Castro I, Magnani DM, Ricciardi M, Pedreño-Lopez N, Bailey V, Betancourt D, Altman JD, Pauthner M, Burton DR, von Bredow B, Evans DT, Yuan M, Parks CL, Ejima K, Allison DB, Rakasz E, Barber GN, Capuano S, Lifson JD, Desrosiers RC, Watkins DI. Vaccine-induced immune responses against both Gag and Env improve control of simian immunodeficiency virus replication in rectally challenged rhesus macaques. PLoS Pathog 2017; 13:e1006529. [PMID: 28732035 PMCID: PMC5540612 DOI: 10.1371/journal.ppat.1006529] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 08/02/2017] [Accepted: 07/13/2017] [Indexed: 01/28/2023] Open
Abstract
The ability to control lentivirus replication may be determined, in part, by the extent to which individual viral proteins are targeted by the immune system. Consequently, defining the antigens that elicit the most protective immune responses may facilitate the design of effective HIV-1 vaccines. Here we vaccinated four groups of rhesus macaques with a heterologous vector prime/boost/boost/boost (PBBB) regimen expressing the following simian immunodeficiency virus (SIV) genes: env, gag, vif, rev, tat, and nef (Group 1); env, vif, rev, tat, and nef (Group 2); gag, vif, rev, tat, and nef (Group 3); or vif, rev, tat, and nef (Group 4). Following repeated intrarectal challenges with a marginal dose of the neutralization-resistant SIVmac239 clone, vaccinees in Groups 1-3 became infected at similar rates compared to control animals. Unexpectedly, vaccinees in Group 4 became infected at a slower pace than the other animals, although this difference was not statistically significant. Group 1 exhibited the best post-acquisition virologic control of SIV infection, with significant reductions in both peak and chronic phase viremia. Indeed, 5/8 Group 1 vaccinees had viral loads of less than 2,000 vRNA copies/mL of plasma in the chronic phase. Vaccine regimens that did not contain gag (Group 2), env (Group 3), or both of these inserts (Group 4) were largely ineffective at decreasing viremia. Thus, vaccine-induced immune responses against both Gag and Env appeared to maximize control of immunodeficiency virus replication. Collectively, these findings are relevant for HIV-1 vaccine design as they provide additional insights into which of the lentiviral proteins might serve as the best vaccine immunogens.
Collapse
Affiliation(s)
- Mauricio A. Martins
- Department of Pathology, University of Miami, Miami, Florida, United States of America
| | - Young C. Shin
- Department of Pathology, University of Miami, Miami, Florida, United States of America
| | - Lucas Gonzalez-Nieto
- Department of Pathology, University of Miami, Miami, Florida, United States of America
| | - Aline Domingues
- Department of Pathology, University of Miami, Miami, Florida, United States of America
| | - Martin J. Gutman
- Department of Pathology, University of Miami, Miami, Florida, United States of America
| | - Helen S. Maxwell
- Department of Pathology, University of Miami, Miami, Florida, United States of America
| | - Iris Castro
- Department of Pathology, University of Miami, Miami, Florida, United States of America
| | - Diogo M. Magnani
- Department of Pathology, University of Miami, Miami, Florida, United States of America
| | - Michael Ricciardi
- Department of Pathology, University of Miami, Miami, Florida, United States of America
| | - Nuria Pedreño-Lopez
- Department of Pathology, University of Miami, Miami, Florida, United States of America
| | - Varian Bailey
- Department of Pathology, University of Miami, Miami, Florida, United States of America
| | - Dillon Betancourt
- Department of Microbiology and Immunology, University of Miami, Miami, Florida, United States of America
| | - John D. Altman
- Department of Microbiology and Immunology, Emory University, Atlanta, Georgia, United States of America
| | - Matthias Pauthner
- Department of Immunology and Microbiology, IAVI Neutralizing Antibody Center, Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery (CHAVI-ID), The Scripps Research Institute, La Jolla, California, United States of America
| | - Dennis R. Burton
- Department of Immunology and Microbiology, IAVI Neutralizing Antibody Center, Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery (CHAVI-ID), The Scripps Research Institute, La Jolla, California, United States of America
| | - Benjamin von Bredow
- Department of Pathology and Laboratory Medicine, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - David T. Evans
- Department of Pathology and Laboratory Medicine, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
- Wisconsin National Primate Research Center, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - Maoli Yuan
- International AIDS Vaccine Initiative, AIDS Vaccine Design and Development Laboratory, Brooklyn, New York, United States of America
| | - Christopher L. Parks
- International AIDS Vaccine Initiative, AIDS Vaccine Design and Development Laboratory, Brooklyn, New York, United States of America
| | - Keisuke Ejima
- Section on Statistical Genetics, Department of Biostatistics, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - David B. Allison
- Section on Statistical Genetics, Department of Biostatistics, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Eva Rakasz
- Wisconsin National Primate Research Center, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - Glen N. Barber
- Department of Cell Biology, University of Miami, Miami, Florida, United States of America
| | - Saverio Capuano
- Wisconsin National Primate Research Center, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - Jeffrey D. Lifson
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Ronald C. Desrosiers
- Department of Pathology, University of Miami, Miami, Florida, United States of America
| | - David I. Watkins
- Department of Pathology, University of Miami, Miami, Florida, United States of America
| |
Collapse
|
11
|
Keele BF, Li W, Borducchi EN, Nkolola JP, Abbink P, Chen B, Seaman MS, Barouch DH. Adenovirus prime, Env protein boost vaccine protects against neutralization-resistant SIVsmE660 variants in rhesus monkeys. Nat Commun 2017; 8:15740. [PMID: 28580942 PMCID: PMC5465370 DOI: 10.1038/ncomms15740] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 04/24/2017] [Indexed: 02/02/2023] Open
Abstract
Previous studies have shown that DNA prime, Ad5 boost vaccines protect against neutralization-sensitive but not neutralization-resistant virus variants within the SIVsmE660 swarm. Here we show that Ad prime, Env protein boost vaccines protect against neutralization-resistant SIVsmE660 variants. We perform two studies in rhesus monkeys with Ad35/Ad26 vectors expressing SIVmac239 Gag/Pol/Env with or without an AS01B-adjuvanted SIVmac32H gp140 protein boost. In a repetitive, low-dose challenge study, we observe robust protection against acquisition of infection by both Ad Alone and Ad/Env vaccines. In a single, high-dose challenge study, only the Ad/Env vaccine affords significant protection against acquisition of infection. Analysis of transmitted/founder (T/F) viruses from this study demonstrates that the Ad/Env vaccine blocks both neutralization-sensitive and neutralization-resistant SIVsmE660 variants in rhesus monkeys with restrictive TRIM5α alleles. These data demonstrate that the adjuvanted Env protein boost is critical for protecting against high-dose SIVsmE660 challenge and for blocking neutralization-resistant viruses within the SIVsmE660 swarm.
Collapse
Affiliation(s)
- Brandon F. Keele
- AIDS and Cancer Virus Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland 21702, USA
| | - Wenjun Li
- Division of Preventive and Behavioral Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01655, USA
| | - Erica N. Borducchi
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, USA
| | - Joseph P. Nkolola
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, USA
| | - Peter Abbink
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, USA
| | - Bing Chen
- Children's Hospital, Boston, Massachusetts 02115, USA
| | - Michael S. Seaman
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, USA
| | - Dan H. Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, USA
- Ragon Institute of MGH, MIT and Harvard, Boston, Massachusetts 02139, USA
| |
Collapse
|
12
|
Fujita M. Commentary: MARCH8 Inhibits HIV-1 Infection by Reducing Virion Incorporation of Envelope Glycoproteins. Front Microbiol 2016; 7:254. [PMID: 26941737 PMCID: PMC4764732 DOI: 10.3389/fmicb.2016.00254] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Accepted: 02/15/2016] [Indexed: 11/13/2022] Open
Affiliation(s)
- Mikako Fujita
- Research Institute for Drug Discovery, School of Pharmacy, Kumamoto University Kumamoto, Japan
| |
Collapse
|
13
|
Characterization and Implementation of a Diverse Simian Immunodeficiency Virus SIVsm Envelope Panel in the Assessment of Neutralizing Antibody Breadth Elicited in Rhesus Macaques by Multimodal Vaccines Expressing the SIVmac239 Envelope. J Virol 2015; 89:8130-51. [PMID: 26018167 DOI: 10.1128/jvi.01221-14] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 09/03/2014] [Indexed: 02/02/2023] Open
Abstract
UNLABELLED Antibodies that can neutralize diverse viral strains are likely to be an important component of a protective human immunodeficiency virus type 1 (HIV-1) vaccine. To this end, preclinical simian immunodeficiency virus (SIV)-based nonhuman primate immunization regimens have been designed to evaluate and enhance antibody-mediated protection. However, these trials often rely on a limited selection of SIV strains with extreme neutralization phenotypes to assess vaccine-elicited antibody activity. To mirror the viral panels used to assess HIV-1 antibody breadth, we created and characterized a novel panel of 14 genetically and phenotypically diverse SIVsm envelope (Env) glycoproteins. To assess the utility of this panel, we characterized the neutralizing activity elicited by four SIVmac239 envelope-expressing DNA/modified vaccinia virus Ankara vector- and protein-based vaccination regimens that included the immunomodulatory adjuvants granulocyte-macrophage colony-stimulating factor, Toll-like receptor (TLR) ligands, and CD40 ligand. The SIVsm Env panel exhibited a spectrum of neutralization sensitivity to SIV-infected plasma pools and monoclonal antibodies, allowing categorization into three tiers. Pooled sera from 91 rhesus macaques immunized in the four trials consistently neutralized only the highly sensitive tier 1a SIVsm Envs, regardless of the immunization regimen. The inability of vaccine-mediated antibodies to neutralize the moderately resistant tier 1b and tier 2 SIVsm Envs defined here suggests that those antibodies were directed toward epitopes that are not accessible on most SIVsm Envs. To achieve a broader and more effective neutralization profile in preclinical vaccine studies that is relevant to known features of HIV-1 neutralization, more emphasis should be placed on optimizing the Env immunogen, as the neutralization profile achieved by the addition of adjuvants does not appear to supersede the neutralizing antibody profile determined by the immunogen. IMPORTANCE Many in the HIV/AIDS vaccine field believe that the ability to elicit broadly neutralizing antibodies capable of blocking genetically diverse HIV-1 variants is a critical component of a protective vaccine. Various SIV-based nonhuman primate vaccine studies have investigated ways to improve antibody-mediated protection against a heterologous SIV challenge, including administering adjuvants that might stimulate a greater neutralization breadth. Using a novel SIV neutralization panel and samples from four rhesus macaque vaccine trials designed for cross comparison, we show that different regimens expressing the same SIV envelope immunogen consistently elicit antibodies that neutralize only the very sensitive tier 1a SIV variants. The results argue that the neutralizing antibody profile elicited by a vaccine is primarily determined by the envelope immunogen and is not substantially broadened by including adjuvants, resulting in the conclusion that the envelope immunogen itself should be the primary consideration in efforts to elicit antibodies with greater neutralization breadth.
Collapse
|
14
|
Yen PJ, Mefford ME, Hoxie JA, Williams KC, Desrosiers RC, Gabuzda D. Identification and characterization of a macrophage-tropic SIV envelope glycoprotein variant in blood from early infection in SIVmac251-infected macaques. Virology 2014; 458-459:53-68. [PMID: 24928039 DOI: 10.1016/j.virol.2014.03.024] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2014] [Revised: 02/23/2014] [Accepted: 03/22/2014] [Indexed: 12/01/2022]
Abstract
Macrophages play an important role in HIV/SIV pathogenesis by serving as a reservoir for viral persistence in brain and other tissues. Infected macrophages have been detected in brain early after infection, but macrophage-tropic viruses are rarely isolated until late-stage infection. Little is known about early variants that establish persistent infection in brain. Here, we characterize a unique macrophage-tropic SIV envelope glycoprotein (Env) variant from two weeks post-infection in blood of an SIVmac251-infected macaque that is closely related to sequences in brain from animals with neurological disease. SIVmac251 clones expressing this Env are highly fusogenic, and replicate efficiently in T cells and macrophages. N173 and N481 were identified as novel determinants of macrophage tropism and neutralization sensitivity. These results imply that macrophage-tropic SIV capable of establishing viral reservoirs in brain can be present in blood during early infection. Furthermore, these SIVmac251 clones will be useful for studies on pathogenesis, eradication, and vaccines.
Collapse
Affiliation(s)
- Po-Jen Yen
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, MA, USA; Division of Medical Sciences Program in Virology, Harvard Medical School, Boston, MA, USA
| | - Megan E Mefford
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, MA, USA; Division of Medical Sciences Program in Virology, Harvard Medical School, Boston, MA, USA
| | - James A Hoxie
- Department of Medicine, Hematology-Oncology Division, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Ronald C Desrosiers
- New England Primate Research Center, Department of Microbiology and Immunobiology, Harvard Medical School, Southborough, MA, USA
| | - Dana Gabuzda
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Neurology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
15
|
Heterogeneity in neutralization sensitivities of viruses comprising the simian immunodeficiency virus SIVsmE660 isolate and vaccine challenge stock. J Virol 2013; 87:5477-92. [PMID: 23468494 DOI: 10.1128/jvi.03419-12] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The sooty mangabey-derived simian immunodeficiency virus (SIV) strain E660 (SIVsmE660) is a genetically heterogeneous, pathogenic isolate that is commonly used as a vaccine challenge strain in the nonhuman primate (NHP) model of human immunodeficiency virus type 1 (HIV-1) infection. Though it is often employed to assess antibody-based vaccine strategies, its sensitivity to antibody-mediated neutralization has not been well characterized. Here, we utilize single-genome sequencing and infectivity assays to analyze the neutralization sensitivity of the uncloned SIVsmE660 isolate, individual viruses comprising the isolate, and transmitted/founder (T/F) viruses arising from low-dose mucosal inoculation of macaques with the isolate. We found that the SIVsmE660 isolate overall was highly sensitive to neutralization by SIV-infected macaque plasma samples (50% inhibitory concentration [IC50] < 10(-5)) and monoclonal antibodies targeting V3 (IC50 < 0.01 μg/ml), CD4-induced (IC50 < 0.1 μg/ml), CD4 binding site (IC50 ~ 1 μg/ml), and V4 (IC50, ~5 μg/ml) epitopes. In comparison, SIVmac251 and SIVmac239 were highly resistant to neutralization by these same antibodies. Differences in neutralization sensitivity between SIVsmE660 and SIVmac251/239 were not dependent on the cell type in which virus was produced or tested. These findings indicate that in comparison to SIVmac251/239 and primary HIV-1 viruses, SIVsmE660 generally exhibits substantially less masking of antigenically conserved Env epitopes. Interestingly, we identified a minor population of viruses (~10%) in both the SIVsmE660 isolate and T/F viruses arising from it that were substantially more resistant (>1,000-fold) to antibody neutralization and another fraction (~20%) that was intermediate in neutralization resistance. These findings may explain the variable natural history and variable protection afforded by heterologous Env-based vaccines in rhesus macaques challenged by high-dose versus low-dose SIVsmE660 inoculation regimens.
Collapse
|
16
|
Johnson WE. Rapid adversarial co-evolution of viruses and cellular restriction factors. Curr Top Microbiol Immunol 2013; 371:123-51. [PMID: 23686234 DOI: 10.1007/978-3-642-37765-5_5] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Since the discovery of viruses over a century ago, virologists have recognized that host genetics plays a major role in viral tropism and the distribution of viruses in nature. Traditionally, studies of tropism have centered on identification of cellular factors required for viral replication, such as cell-surface entry receptors. However, over the past 20 years, there has been a steady increase in the identification and characterization of restriction factors (RFs), here defined as dominant cellular factors that have evolved specifically to interfere with viral replication. Genetic studies suggest that restriction factors impose significant barriers to interspecies movement of viruses and are therefore critical determinants of viral tropism. Furthermore, the scope of the ever-expanding list of restriction factors, and the variety of antiviral mechanisms they represent, testifies to the extraordinary impact viruses have had on organismal evolution-an impact hitherto underappreciated by evolutionary biologists and virologists alike. Recent studies of RF-encoding genes that combine molecular evolutionary analysis with functional assays illustrate the potential for asking questions about virus-host interactions as they play out in natural populations and across evolutionary timescales. Most notably, it has become common to apply tests of positive selection to RF genes and couple these analyses with virological assays, to reveal evidence for antagonistic virus-host co-evolution. Herein, I summarize recent work on the evolutionary genetics of mammalian RFs, particularly those of humans, non-human primates, and model organisms, and how RFs can reveal the influence of virus-host interactions on organismal evolution. Because intensive investigation of RF evolution is fairly new (and because there is still much to learn), the discussion is organized around five broad, outstanding questions that will need to be answered before we can fully appreciate the evolutionary biology of restriction.
Collapse
|
17
|
Bilello JP, Manrique JM, Shin YC, Lauer W, Li W, Lifson JD, Mansfield KG, Johnson RP, Desrosiers RC. Vaccine protection against simian immunodeficiency virus in monkeys using recombinant gamma-2 herpesvirus. J Virol 2011; 85:12708-20. [PMID: 21900170 PMCID: PMC3209374 DOI: 10.1128/jvi.00865-11] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2011] [Accepted: 08/27/2011] [Indexed: 12/21/2022] Open
Abstract
Recombinant strains of replication-competent rhesus monkey rhadinovirus (RRV) were constructed in which strong promoter/enhancer elements were used to drive expression of simian immunodeficiency virus (SIV) Env or Gag or a Rev-Tat-Nef fusion protein. Cultured rhesus monkey fibroblasts infected with each recombinant strain were shown to express the expected protein. Three RRV-negative and two RRV-positive rhesus monkeys were inoculated intravenously with a mixture of these three recombinant RRVs. Expression of SIV Gag was readily detected in lymph node biopsy specimens taken at 3 weeks postimmunization. Impressive anti-SIV cellular immune responses were elicited on the basis of major histocompatibility complex (MHC) tetramer staining and gamma interferon enzyme-linked immunospot (ELISPOT) assays. Responses were much greater in magnitude in the monkeys that were initially RRV negative but were still readily detected in the two monkeys that were naturally infected with RRV at the time of immunization. By 3 weeks postimmunization, responses measured by MHC tetramer staining in the two Mamu-A*01(+) RRV-negative monkeys reached 9.3% and 13.1% of all CD8(+) T cells in peripheral blood to the Gag CM9 epitope and 2.3% and 7.3% of all CD8(+) T cells in peripheral blood to the Tat SL8 epitope. Virus-specific CD8(+) T cell responses persisted at high levels up to the time of challenge at 18 weeks postimmunization, and responding cells maintained an effector memory phenotype. Despite the ability of the RRVenv recombinant to express high levels of Env in cultured cells, and despite the appearance of strong anti-RRV antibody responses in immunized monkeys, anti-Env antibody responses were below our ability to detect them. Immunized monkeys, together with three unimmunized controls, were challenged intravenously with 10 monkey infectious doses of SIVmac239. All five immunized monkeys and all three controls became infected with SIV, but peak viral loads were 1.2 to 3.0 log(10) units lower and chronic-phase viral loads were 1.0 to 3.0 log(10) units lower in immunized animals than the geometric mean of unimmunized controls. These differences were statistically significant. Anti-Env antibody responses following challenge indicated an anamnestic response in the vaccinated monkeys. These findings further demonstrate the potential of recombinant herpesviruses as preventive vaccines for AIDS. We hypothesize that this live, replication-competent, persistent herpesvirus vector could match, or come close to matching, live attenuated strains of SIV in the degree of protection if the difficulty with elicitation of anti-Env antibody responses can be overcome.
Collapse
MESH Headings
- Animals
- Antibodies, Viral/immunology
- Blotting, Western
- Enzyme-Linked Immunosorbent Assay
- Flow Cytometry
- Gammaherpesvirinae/genetics
- Gammaherpesvirinae/immunology
- Gene Products, env/administration & dosage
- Gene Products, env/genetics
- Gene Products, env/immunology
- Gene Products, gag/administration & dosage
- Gene Products, gag/genetics
- Gene Products, gag/immunology
- Gene Products, nef/genetics
- Gene Products, nef/immunology
- Genetic Vectors
- Herpesviridae Infections/genetics
- Herpesviridae Infections/metabolism
- Herpesviridae Infections/virology
- Humans
- Immunity, Cellular
- Immunoenzyme Techniques
- Kidney/cytology
- Kidney/metabolism
- Kidney/virology
- Macaca mulatta/genetics
- Macaca mulatta/immunology
- Macaca mulatta/virology
- Neutralization Tests
- Plasmids
- Recombination, Genetic
- SAIDS Vaccines/administration & dosage
- SAIDS Vaccines/genetics
- SAIDS Vaccines/immunology
- Simian Acquired Immunodeficiency Syndrome/immunology
- Simian Acquired Immunodeficiency Syndrome/prevention & control
- Simian Acquired Immunodeficiency Syndrome/virology
- Simian Immunodeficiency Virus/immunology
- Vaccination
- Viral Load
- Virus Replication
Collapse
Affiliation(s)
- John P. Bilello
- New England Primate Research Center, Harvard Medical School, Southborough, Massachusetts 01772-9102
| | - Julieta M. Manrique
- New England Primate Research Center, Harvard Medical School, Southborough, Massachusetts 01772-9102
| | - Young C. Shin
- New England Primate Research Center, Harvard Medical School, Southborough, Massachusetts 01772-9102
| | - William Lauer
- New England Primate Research Center, Harvard Medical School, Southborough, Massachusetts 01772-9102
| | - Wenjun Li
- University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, Massachusetts 01655
| | - Jeffrey D. Lifson
- AIDS and Cancer Virus Program, SAIC Frederick Inc., National Cancer Institute, NCI Frederick, Frederick, Maryland 21702
| | - Keith G. Mansfield
- New England Primate Research Center, Harvard Medical School, Southborough, Massachusetts 01772-9102
| | - R. Paul Johnson
- New England Primate Research Center, Harvard Medical School, Southborough, Massachusetts 01772-9102
| | - Ronald C. Desrosiers
- New England Primate Research Center, Harvard Medical School, Southborough, Massachusetts 01772-9102
| |
Collapse
|
18
|
Han X, Zou J, Wang X, Guo W, Huo G, Shen R, Xiang W. Amino acid mutations in the env gp90 protein that modify N-linked glycosylation of the Chinese EIAV vaccine strain enhance resistance to neutralizing antibodies. Viral Immunol 2010; 23:531-9. [PMID: 20883167 DOI: 10.1089/vim.2009.0006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The Chinese EIAV vaccine is an attenuated live-virus vaccine obtained by serial passage of a virulent horse isolate (EIAV(L)) in donkeys (EIAV(D)), and subsequently in donkey cells in vitro. In this study, we compare the env gene of the original horse virulent virus (EIAV(L)) with attenuated strains serially passaged in donkey MDM (EIAV(DLV)), and donkey dermal cells (EIAV(FDDV)). Genetic comparisons among parental and attenuated strains found that vaccine strains contained amino acid substitutions/deletions in gp90 that resulted in a loss of three potential N-linked glycosylation sites, designated g5, g9, and g10. To investigate the biological significance of these changes, reverse-mutated viruses were constructed in the backbone of the EIAV(FDDV) infectious molecular clone (pLGFD3). The resulting virus stocks were characterized for replication efficiency in donkey dermal cells and donkey MDM, and were tested for sensitivity to neutralization using sera from two ponies experimentally infected with EIAV(FDDV). The results clearly show that these mutations generated by site-directed mutagenesis resulted in cloned viruses with enhanced resistance to serum-neutralizing antibodies that were also able to recognize parental viruses. The results of this study indicate that these mutations play an important role in the attenuation of the EIAV vaccine strains.
Collapse
Affiliation(s)
- Xiue Han
- Heilongjiang Dairy Industry Technical Development Center, Northeast Agricultural University, Harbin, China.
| | | | | | | | | | | | | |
Collapse
|
19
|
Bixby JG, Laur O, Johnson WE, Desrosiers RC. Diversity of envelope genes from an uncloned stock of SIVmac251. AIDS Res Hum Retroviruses 2010; 26:1115-31. [PMID: 20836705 DOI: 10.1089/aid.2010.0029] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
AIDS vaccine and pathogenesis research will benefit from a more diverse array of cloned SIV challenge stocks from which to choose. Toward this end, 20 envelope genes were cloned from an extensively used, primary stock of uncloned SIVmac251. Each of the 20 clones had a unique sequence. Their translated sequences differed by as many as 26 amino acids from one another and by as many as 45 amino acids from the commonly used clone SIVmac239. Envelope sequences up to and including the membrane-spanning domain were exchanged into the infectious pathogenic SIVmac239 clone and virus stocks were produced by HEK293T cell transfection. Seventeen of the 20 recombinants were replication competent. The infectivities per ng p27 of the 17 new replication-competent recombinants in C8166-SEAP cells and in TZM-bl cells ranged from minus 32-fold to plus 7.6-fold relative to SIVmac239. A range of sensitivities to neutralization by sCD4 and by sera from SIV-infected macaques was observed but none was as sensitive to these neutralizing agents as SIVmac316, the highly macrophage-competent derivative of SIVmac239. Four strains that were most sensitive to sCD4 inhibition were also among the most sensitive to antibody-mediated neutralization. None of the new recombinant viruses replicated as well as SIVmac316 in primary alveolar macrophage cultures from rhesus monkeys but three of the strains did exhibit significant levels of delayed replication in these primary macrophages, reaching peak levels of virus production of ≥50 ng/ml p27 compared to 600-800 ng/ml p27 with SIVmac316. These new SIV clones are being contributed to the NIH AIDS Reagent Repository and are available to the scientific community.
Collapse
Affiliation(s)
- Jacqueline G. Bixby
- New England Primate Research Center, Harvard Medical School, Southborough, Massachusetts
| | - Olga Laur
- New England Primate Research Center, Harvard Medical School, Southborough, Massachusetts
| | - Welkin E. Johnson
- New England Primate Research Center, Harvard Medical School, Southborough, Massachusetts
| | - Ronald C. Desrosiers
- New England Primate Research Center, Harvard Medical School, Southborough, Massachusetts
| |
Collapse
|
20
|
Yamamoto H, Matano T. Neutralizing antibodies in SIV control: co-impact with T cells. Vaccine 2010; 28 Suppl 2:B13-7. [PMID: 20510737 DOI: 10.1016/j.vaccine.2009.09.080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2009] [Revised: 08/19/2009] [Accepted: 09/18/2009] [Indexed: 11/25/2022]
Abstract
Human immunodeficiency virus type 1 (HIV-1) and pathogenic simian immunodeficiency virus (SIV)-infected naïve hosts experience a characteristic absence of early and potent virus-specific neutralizing antibody (NAb) responses preceding establishment of persistent infection. Yet conversely, we have recently shown that NAbs passively immunized in rhesus macaques at early post-SIV challenge are capable of playing a critical role in non-sterile viremia control with implications of antibody-enhanced antigen presentation. In a current follow-up study we have further reported that NAbs mediate rapid elicitation of polyfunctional virus-specific CD4+ T-cells in vivo. The NAb-immunized macaques mounting these responses exhibited sustained viremia control for over 1 year, accompanied with robust anti-SIV cellular immunity. Perspectives obtained from the results are discussed.
Collapse
Affiliation(s)
- Hiroyuki Yamamoto
- International Research Center for Infectious Diseases, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | | |
Collapse
|
21
|
The role of porcine reproductive and respiratory syndrome (PRRS) virus structural and non-structural proteins in virus pathogenesis. Anim Health Res Rev 2010; 11:135-63. [DOI: 10.1017/s1466252310000034] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
AbstractPorcine reproductive and respiratory syndrome (PRRS) is an economically devastating viral disease affecting the swine industry worldwide. The etiological agent, PRRS virus (PRRSV), possesses a RNA viral genome with nine open reading frames (ORFs). The ORF1a and ORF1b replicase-associated genes encode the polyproteins pp1a and pp1ab, respectively. The pp1a is processed in nine non-structural proteins (nsps): nsp1α, nsp1β, and nsp2 to nsp8. Proteolytic cleavage of pp1ab generates products nsp9 to nsp12. The proteolytic pp1a cleavage products process and cleave pp1a and pp1ab into nsp products. The nsp9 to nsp12 are involved in virus genome transcription and replication. The 3′ end of the viral genome encodes four minor and three major structural proteins. The GP2a, GP3and GP4(encoded by ORF2a, 3 and 4), are glycosylated membrane associated minor structural proteins. The fourth minor structural protein, the E protein (encoded by ORF2b), is an unglycosylated membrane associated protein. The viral envelope contains two major structural proteins: a glycosylated major envelope protein GP5(encoded by ORF5) and an unglycosylated membrane M protein (encoded by ORF6). The third major structural protein is the nucleocapsid N protein (encoded by ORF7). All PRRSV non-structural and structural proteins are essential for virus replication, and PRRSV infectivity is relatively intolerant to subtle changes within the structural proteins. PRRSV virulence is multigenic and resides in both the non-structural and structural viral proteins. This review discusses the molecular characteristics, biological and immunological functions of the PRRSV structural and nsps and their involvement in the virus pathogenesis.
Collapse
|
22
|
[Theory for prophylactic AIDS vaccine development]. Uirusu 2010; 59:267-75. [PMID: 20218335 DOI: 10.2222/jsv.59.267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
In the natural courses of human immunodeficiency virus (HIV) infections, host immune responses fail to contain the virus and allow persistent HIV replication, leading to AIDS progression. For development of an effective vaccine against those viral infections which do not show spontaneous remission, it is important to elucidate which immune responses to be induced for viral control. This review focuses on antibodies and cytotoxic T lymphocytes, key adaptive immune effectors, and discusses possible mechanisms for HIV control by vaccine-induced antibody, memory B lymphocyte, and (effector and central) memory T lymphocyte responses. Finally, we mention the ongoing international project for a clinical trial of our Sendai virus vector-based AIDS vaccine.
Collapse
|
23
|
Tonsillar application of AT-2 SIV affords partial protection against rectal challenge with SIVmac239. J Acquir Immune Defic Syndr 2009; 52:433-42. [PMID: 19779309 DOI: 10.1097/qai.0b013e3181b880f3] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Although mucosal responses are important for preventing infections with HIV, the optimal strategies for inducing them remain unclear. To evaluate vaccine strategies targeting the oral mucosal lymphoid tissue inductive sites as an approach to provide immunity at distal sites, we vaccinated healthy macaques via the palatine/lingual tonsils with aldrithiol 2 (AT-2) inactivated Simian immunodeficiency virus (SIV)mac239, combined with CpG-C immunostimulatory oligonucleotide (CpG-C ISS-ODN, C274) as the adjuvant. METHODS Macaques received 5 doses of C274 or control ODN C661 and AT-2 SIV on the tonsillar tissues every 6 weeks before being challenged rectally with SIVmac239, 8 weeks after the last immunization. RESULTS Although no T-cell or B-cell responses were detected in the blood before challenge, antibody (Ab) responses were detected in the rectum. Immunization with AT-2 SIV significantly reduced the frequency of infection compared with nonimmunized controls, irrespective of adjuvant. In the vaccinated animals that became infected, peak viremias were somewhat reduced. SIV-specific responses were detected in the blood once animals became infected with no detectable differences between the differently immunized groups and the controls. CONCLUSION This work provides evidence that vaccine immunogens applied to the oral mucosal associated lymphoid tissues can provide benefit against rectal challenge, a finding with important implications for mucosal vaccination strategies.
Collapse
|
24
|
Yamamoto H, Matano T. Anti-HIV adaptive immunity: determinants for viral persistence. Rev Med Virol 2008; 18:293-303. [PMID: 18416450 DOI: 10.1002/rmv.577] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The immense difficulty in primary control of human immunodeficiency virus (HIV) and simian immunodeficiency virus (SIV) infection by adaptive immune responses has been a topic of exceptional importance. CD8+ cytotoxic T lymphocytes (CTLs) do play a central role in primary resolution of viremia, but their potency in viral control is generally constrained in the natural courses of HIV/SIV infections. The overall repertoire of CTLs is dependent on both the host and the virus genetic polymorphisms, and the potency of each individual CTL is affected by immunological and virological determinants. HIV/SIV infections lack early appearance of neutralising antibodies (NAbs), and our recent finding has suggested a possibility of their absence contributing to diminished virus-specific CD4+ T-cell responses leading to failure in primary viral control. Extrapolations from studies in macaque models of SIV infection and analyses of the cohorts of HIV control in humans have to date delineated the numerous requirements for attainment of viral control. Understanding of the individual components of adaptive immune responses and their optimal concert required for HIV/SIV control would contribute to development of an effective AIDS vaccine. Here, we discuss current insights into CTLs and NAbs, and speculate their possible protective mechanism against establishment of persistent HIV/SIV infection.
Collapse
Affiliation(s)
- Hiroyuki Yamamoto
- International Research Center for Infectious Diseases, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | | |
Collapse
|
25
|
Glycosylation of gp41 of simian immunodeficiency virus shields epitopes that can be targets for neutralizing antibodies. J Virol 2008; 82:12472-86. [PMID: 18829751 DOI: 10.1128/jvi.01382-08] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Human immunodeficiency virus type 1 and simian immunodeficiency virus possess three closely spaced, highly conserved sites for N-linked carbohydrate attachment in the extracellular domain of the transmembrane protein gp41. We infected rhesus monkeys with a variant of cloned SIVmac239 lacking the second and third sites or with a variant strain lacking all three of SIVmac239's glycosylation sites in gp41. For each mutation, asparagine (N) in the canonical N-X-S/T recognition sequence for carbohydrate attachment was changed to the structurally similar glutamine such that two nucleotide changes would be required for a reversion of the mutated codon. By 16 weeks, experimentally infected monkeys made antibodies that neutralized the mutant viruses to high titers. Such antibodies were not observed in monkeys infected with the parental virus. Thus, new specificities were revealed as a result of the carbohydrate attachment mutations, and antibodies of these specificities had neutralizing activity. Unlike monkeys infected with the parental virus, monkeys infected with the mutant viruses made antibodies that reacted with peptides corresponding to the sequences in this region. Furthermore, there was strong selective pressure for the emergence of variant sequences in this region during the course of infection. By analyzing the neutralization profiles of sequence variants, we were able to define three mutations (Q625R, K631N, and Q634H) in the region of the glycosylation site mutations that conferred resistance to neutralization by plasma from the monkeys infected with mutant virus. Based on the reactivity of antibodies to peptides in this region and the colocalization of neutralization escape mutations, we conclude that N-linked carbohydrates in the ectodomain of the transmembrane protein shield underlying epitopes that would otherwise be the direct targets of neutralizing antibodies.
Collapse
|
26
|
Potent antibody-mediated neutralization and evolution of antigenic escape variants of simian immunodeficiency virus strain SIVmac239 in vivo. J Virol 2008; 82:9739-52. [PMID: 18667507 DOI: 10.1128/jvi.00871-08] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Here, we describe the evolution of antigenic escape variants in a rhesus macaque that developed unusually high neutralizing antibody titers to SIVmac239. By 42 weeks postinfection, 50% neutralization of SIVmac239 was achieved with plasma dilutions of 1:1,000. Testing of purified immunoglobulin confirmed that the neutralizing activity was antibody mediated. Despite the potency of the neutralizing antibody response, the animal displayed a typical viral load profile and progressed to terminal AIDS with a normal time course. Viral envelope sequences from week 16 and week 42 plasma contained an excess of nonsynonymous substitutions, predominantly in V1 and V4, including individual sites with ratios of nonsynonymous to synonymous substitution rates (dN/dS) highly suggestive of strong positive selection. Recombinant viruses encoding envelope sequences isolated from these time points remained resistant to neutralization by all longitudinal plasma samples, revealing the failure of the animal to mount secondary responses to the escaped variants. Substitutions at two sites with significant dN/dS values, one in V1 and one in V4, were independently sufficient to confer nearly complete resistance to neutralization. Substitutions at three additional sites, one in V4 and two in gp41, conferred moderate to high levels of resistance when tested individually. All the amino acid changes leading to escape resulted from single nucleotide substitutions. The observation that antigenic escape resulted from individual, single amino acid replacements at sites well separated in current structural models of Env indicates that the virus can utilize multiple independent pathways to rapidly achieve similar levels of resistance.
Collapse
|
27
|
Vaccine protection by live, attenuated simian immunodeficiency virus in the absence of high-titer antibody responses and high-frequency cellular immune responses measurable in the periphery. J Virol 2008; 82:4135-48. [PMID: 18272584 DOI: 10.1128/jvi.00015-08] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
An attenuated derivative of simian immunodeficiency virus strain 239 deleted of V1-V2 sequences in the envelope gene (SIV239DeltaV1-V2) was used for vaccine/challenge experiments in rhesus monkeys. Peak levels of viral RNA in plasma of 10(4) to 10(6.5) copies/ml in the weeks immediately following inoculation of SIV239DeltaV1-V2 were 10- to 1,000-fold lower than those observed with parental SIV239 ( approximately 10(7.3) copies/ml). Viral loads consistently remained below 200 copies/ml after 8 weeks of infection by the attenuated SIV239DeltaV1-V2 strain. Viral localization experiments revealed large numbers of infected cells within organized lymphoid nodules of the colonic gut-associated lymphoid tissue at 14 days; double-labeling experiments indicated that 93.5% of the virally infected cells at this site were positive for the macrophage marker CD68. Cellular and humoral immune responses measured principally by gamma interferon enzyme-linked immunospot and neutralization assays were variable in the five vaccinated monkeys. One monkey had responses in these assays comparable to or only slightly less than those observed in monkeys infected with parental, wild-type SIV239. Four of the vaccinated monkeys, however, had low, marginal, or undetectable responses in these same assays. These five vaccinated monkeys and three naïve control monkeys were subsequently challenged intravenously with wild-type SIV239. Three of the five vaccinated monkeys, including the one with strong anti-SIV immune responses, were strongly protected against the challenge on the basis of viral load measurements. Surprisingly, two of the vaccinated monkeys were strongly protected against SIV239 challenge despite the presence of cellular anti-SIV responses of low-frequency and low-titer anti-SIV antibody responses. These results indicate that high-titer anti-SIV antibody responses and high-frequency anti-SIV cellular immune responses measurable by standard assays from the peripheral blood are not needed to achieve strong vaccine protection, even against a difficult, neutralization-resistant strain such as SIV239.
Collapse
|
28
|
Craigo JK, Zhang B, Barnes S, Tagmyer TL, Cook SJ, Issel CJ, Montelaro RC. Envelope variation as a primary determinant of lentiviral vaccine efficacy. Proc Natl Acad Sci U S A 2007; 104:15105-10. [PMID: 17846425 PMCID: PMC1986620 DOI: 10.1073/pnas.0706449104] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Lentiviral envelope antigenic variation and associated immune evasion are believed to present major obstacles to effective vaccine development. Although this perception is widely assumed by the scientific community, there is, to date, no rigorous experimental data assessing the effect of increasing levels of lentiviral Env variation on vaccine efficacy. It is our working hypothesis that Env is, in fact, a primary determinant of vaccine effectiveness. We previously reported that a successful experimental attenuated equine infectious anemia virus vaccine, derived by mutation of the viral S2 accessory gene, provided 100% protection from disease after virulent virus challenge. Here, we sought to comprehensively test our hypothesis by challenging vaccinated animals with proviral strains of defined, increasing Env variation, using variant envelope SU genes that arose naturally during experimental infection of ponies with equine infectious anemia virus. The reference attenuated vaccine combined with these variant Env challenge strains facilitated evaluation of the protection conferred by ancestral immunogens, because the Env of the attenuated vaccine is a direct ancestor to the variant proviral strain Envs. The results demonstrated that ancestral Env proteins did not impart broad levels of protection against challenge. Furthermore, the results displayed a significant inverse linear correlation of Env divergence and protection from disease. This study demonstrates potential obstacles to the use of single isolate ancestral Env immunogens. Finally, these findings reveal that relatively minor Env variation can pose a substantial challenge to lentiviral vaccine immunity, even when attenuated vaccines are used that, to date, achieve the highest levels of vaccine protection.
Collapse
Affiliation(s)
- Jodi K. Craigo
- *Center for Vaccine Research
- Department of Molecular Genetics and Biochemistry, University of Pittsburgh, Pittsburgh, PA 15261; and
| | - Baoshan Zhang
- *Center for Vaccine Research
- Department of Molecular Genetics and Biochemistry, University of Pittsburgh, Pittsburgh, PA 15261; and
| | - Shannon Barnes
- *Center for Vaccine Research
- Department of Molecular Genetics and Biochemistry, University of Pittsburgh, Pittsburgh, PA 15261; and
| | - Tara L. Tagmyer
- *Center for Vaccine Research
- Department of Molecular Genetics and Biochemistry, University of Pittsburgh, Pittsburgh, PA 15261; and
| | - Sheila J. Cook
- Department of Veterinary Science, Gluck Equine Research Center, University of Kentucky, Lexington, KY 40516
| | - Charles J. Issel
- Department of Veterinary Science, Gluck Equine Research Center, University of Kentucky, Lexington, KY 40516
| | - Ronald C. Montelaro
- *Center for Vaccine Research
- Department of Molecular Genetics and Biochemistry, University of Pittsburgh, Pittsburgh, PA 15261; and
- To whom correspondence should be addressed at:
Department of Molecular Genetics and Biochemistry, W1144 Biomedical Science Tower, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261. E-mail:
| |
Collapse
|
29
|
Laird ME, Desrosiers RC. Infectivity and neutralization of simian immunodeficiency virus with FLAG epitope insertion in gp120 variable loops. J Virol 2007; 81:10838-48. [PMID: 17686865 PMCID: PMC2045528 DOI: 10.1128/jvi.00831-07] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
A FLAG epitope tag was substituted within variable loop 1 (V1), 2 (V2), or 4 (V4) of the gp120 envelope glycoprotein of simian immunodeficiency virus strain 239 (SIV239) to evaluate the extent to which each variable loop may serve as a target for antibody-mediated neutralization. Two sites within each variable loop of SIV239 were chosen for individual epitope tag insertions. FLAG epitope substitutions were also made in the V1, V2, and V4 loops of a neutralization-sensitive derivative of SIV239, SIV316. Of the 10 FLAG-tagged recombinant viruses analyzed, three (SIV239FV1b, SIV239FV2b, and SIV239FV4a) replicated with kinetics similar to those of the parental strain, SIV239, in both CEMx174 cells and the immortalized rhesus monkey T-cell line 221. The SIV316FV1b and SIV316FV4a FLAG variants replicated with a substantial lag, and the five remaining recombinants did not replicate detectably. Both gp160 and gp120 from replication-competent FLAG variants could be immunoprecipitated from transfected 293T cells by the anti-gp120 rhesus monoclonal antibody (RhMAb) 3.11H, the anti-FLAG MAb M2, and CD4-immunoglobulin, whereas only unprocessed gp160 was detected in 293T cells transfected with replication-defective variants. Furthermore, gp120 was detectably incorporated only into virions that were infectious. SIV239FV1b was sensitive to neutralization by MAb M2, with a 50% inhibitory concentration of 1 mug/ml. Neither SIV239FV2b nor SIV239FV4a was sensitive to M2 neutralization. The ability of the M2 antibody to neutralize SIV239FV1b infectivity was associated with an increased ability of the M2 antibody to detect native, oligomeric SIV239FV1b envelope protein on the surfaces of cells relative to that for the other SIV FLAG variants. Furthermore, SIV239FV1b was globally more sensitive to antibody-mediated neutralization than was parental SIV239 when these strains were screened with a panel of anti-SIV MAbs of various specificities. These results indicate that the V1 loop can serve as an effective target for neutralization on SIV239FV1b. However, antibody-mediated neutralization of this variant, similar to that of other SIV239 variants that have been studied previously, was associated with a global increase in neutralization sensitivity. These results suggest that the variable loops on the neutralization-resistant SIV239 strain are difficult for antibodies to access effectively and that mutations that allow neutralization have global effects on the trimeric envelope glycoprotein structure and accessibility.
Collapse
Affiliation(s)
- Melissa E Laird
- New England Primate Research Center, One Pine Hill Drive, Box 9102, Southborough, MA 01772-9102, USA
| | | |
Collapse
|
30
|
Igarashi T, Donau OK, Imamichi H, Nishimura Y, Theodore TS, Iyengar R, Erb C, Buckler-White A, Buckler CE, Martin MA. Although macrophage-tropic simian/human immunodeficiency viruses can exhibit a range of pathogenic phenotypes, a majority of isolates induce no clinical disease in immunocompetent macaques. J Virol 2007; 81:10669-79. [PMID: 17626082 PMCID: PMC2045473 DOI: 10.1128/jvi.00517-07] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Unlike prototypical lentiviruses like visna and caprine arthritis-encephalitis viruses, which are mainly macrophage tropic (M-tropic), primate lentiviruses primarily target CD4+ T lymphocytes. We previously reported that during the late phase of highly pathogenic chimeric simian/human immunodeficiency virus (SHIV) infections of rhesus macaques, when CD4+ T cells have been systemically eliminated, high levels of viremia are maintained from productively infected macrophages. The availability of several different M-tropic SHIVs from such late-stage immunocompromised animals provided the opportunity to assess whether they might contribute to the immune deficiency induced by their T-cell-tropic parental viruses or possibly cause a distinct disease based on their capacity to infect macrophages. Pairs of rhesus monkeys were therefore inoculated intravenously with six different M-tropic SHIV preparations, and their plasma viral RNA loads, circulating lymphocyte subset numbers, and eventual disease outcomes were monitored. Only one of these six M-tropic SHIVs induced any disease; the disease phenotype observed was the typical rapid, complete, and irreversible depletion of CD4+ T cells induced by pathogenic SHIVs. An analysis of two asymptomatic monkeys, previously inoculated with an M-tropic SHIV recovered directly from alveolar macrophages, revealed that this inoculum targeted alveolar macrophages in vivo, compared to a T-cell-tropic virus, yet no clinical disease occurred. Although one isolate did, in fact, induce the prototypical rapid, irreversible, and complete loss of CD4+ T cells, indicating that M-tropism and pathogenicity may not be inversely related, the majority of M-tropic SHIVs induced no clinical disease in immunocompetent macaques.
Collapse
Affiliation(s)
- Tatsuhiko Igarashi
- Laboratory of Molecular Microbiology, Building 4, Room 315, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
This is a brief summary of the animal models session held during the 12th Annual Meeting of the Society on NeuroImmune Pharmacology, Santa Fe, NM, USA. This session provided important information for participants on availability and utility of animal models for the studies of HIV-1 central nervous system infection and drug abuse. It highlighted animal model relevance to human disease/condition, its utility for the studies of pathogenesis, potential importance for the development of therapeutics, and demonstrated limitations/pitfalls.
Collapse
Affiliation(s)
- Yuri Persidsky
- Department of Pharmacology/Experimental Neuroscience, University of Nebraska Medical Center, 985215 Nebraska Medical Center, Omaha, NE 68198-5215, USA.
| | | |
Collapse
|
32
|
Abstract
Animal models for human immunodeficiency virus (HIV) infection play a key role in understanding the pathogenesis of AIDS and the development of therapeutic agents and vaccines. As the only lentivirus that causes an immunodeficiency resembling that of HIV infection, in its natural host, feline immunodeficiency virus (FIV) has been a unique and powerful model for AIDS research. FIV was first described in 1987 by Niels Pedersen and co-workers as the causative agent for a fatal immunodeficiency syndrome observed in cats housed in a cattery in Petaluma, California. Since this landmark observation, multiple studies have shown that natural and experimental infection of cats with biological isolates of FIV produces an AIDS syndrome very similar in pathogenesis to that observed for human AIDS. FIV infection induces an acute viremia associated with Tcell alterations including depressed CD4 :CD8 T-cell ratios and CD4 T-cell depletion, peripheral lymphadenopathy, and neutropenia. In later stages of FIV infection, the host suffers from chronic persistent infections that are typically self-limiting in an immunocompetent host, as well as opportunistic infections, chronic diarrhea and wasting, blood dyscracias, significant CD4 T-cell depletion, neurologic disorders, and B-cell lymphomas. Importantly, chronic FIV infection induces a progressive lymphoid and CD4 T-cell depletion in the infected cat. The primary mode of natural FIV transmission appears to be blood-borne facilitated by fighting and biting. However, experimental infection through transmucosal routes (rectal and vaginal mucosa and perinatal) have been well documented for specific FIV isolates. Accordingly, FIV disease pathogenesis exhibits striking similarities to that described for HIV-1 infection.
Collapse
|
33
|
Humbert M, Antoni S, Brill B, Landersz M, Rodes B, Soriano V, Wintergerst U, Knechten H, Staszewski S, von Laer D, Dittmar MT, Dietrich U. Mimotopes selected with antibodies from HIV-1-neutralizing long-term non-progressor plasma. Eur J Immunol 2007; 37:501-15. [PMID: 17236253 DOI: 10.1002/eji.200636560] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
A promising approach to identify HIV-1 vaccine candidates is to dissect the natural immune response against the virus in persons controlling the infection over decades without any antiviral therapy. Here we focus on a group of such persons, eight long-term non-progressors (LTNP), in which we proved the presence of broadly neutralizing antibodies against HIV-1 in the plasma as very likely cause for their LTNP status. The aim of this study was to identify the epitopes for these neutralizing antibodies, as these should represent immunogens potentially able to elicit neutralizing antibodies upon vaccination. We screened random peptide phage libraries with plasma antibodies from eight LTNP. After several rounds of positive and negative selection, about 700 HIV-specific mimotopes were sequenced. The mimotope sequences were analyzed for homology to HIV-1 Env, in particular for their capacity to represent conformational epitopes on the surface of the gp120 structure using our software 3DEX. Related phage groups were analyzed for crossreactivity with the LTNP plasma by ELISA as well as for their capacity to induce HIV-1-neutralizing antibodies in mice. Based on this study interesting mimotopes can now be selected for further immunization studies.
Collapse
Affiliation(s)
- Michael Humbert
- Georg-Speyer-Haus, Institute for Biomedical Research, Frankfurt am Main, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Prabakaran P, Dimitrov AS, Fouts TR, Dimitrov DS. Structure and function of the HIV envelope glycoprotein as entry mediator, vaccine immunogen, and target for inhibitors. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2007; 55:33-97. [PMID: 17586312 PMCID: PMC7111665 DOI: 10.1016/s1054-3589(07)55002-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This chapter discusses the advances of the envelope glycoprotein (Env) structure as related to the interactions of conserved Env structures with receptor molecules and antibodies with implications for the design of vaccine immunogens and inhibitors. The human immunodeficiency virus (HIV) Env binds to cell surface–associated receptor (CD4) and coreceptor (CCR5 or CXCR4) by one of its two non-covalently associated subunits, gp120. The induced conformational changes activate the other subunit (gp41), which causes the fusion of the viral with the plasma cell membranes resulting in the delivery of the viral genome into the cell and the initiation of the infection cycle. As the only HIV protein exposed to the environment, the Env is also a major immunogen to which neutralizing antibodies are directed and a target that is relatively easy to access by inhibitors. A fundamental problem in the development of effective vaccines and inhibitors against HIV is the rapid generation of alterations at high levels of expression during long chronic infection and the resulting significant heterogeneity of the Env. The preservation of the Env function as an entry mediator and limitations on size and expression impose restrictions on its variability and lead to the existence of conserved structures.
Collapse
Affiliation(s)
- Ponraj Prabakaran
- Protein Interactions Group, CCRNP, CCR, NCI-Frederick, NIH Frederick, MD 21702, USA
| | | | | | | |
Collapse
|
35
|
Kaur A, Sanford HB, Garry D, Lang S, Klumpp SA, Watanabe D, Bronson RT, Lifson JD, Rosati M, Pavlakis GN, Felber BK, Knipe DM, Desrosiers RC. Ability of herpes simplex virus vectors to boost immune responses to DNA vectors and to protect against challenge by simian immunodeficiency virus. Virology 2006; 357:199-214. [PMID: 16962628 PMCID: PMC1819472 DOI: 10.1016/j.virol.2006.08.007] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2006] [Revised: 06/14/2006] [Accepted: 08/02/2006] [Indexed: 10/24/2022]
Abstract
The immunogenicity and protective capacity of replication-defective herpes simplex virus (HSV) vector-based vaccines were examined in rhesus macaques. Three macaques were inoculated with recombinant HSV vectors expressing Gag, Env, and a Tat-Rev-Nef fusion protein of simian immunodeficiency virus (SIV). Three other macaques were primed with recombinant DNA vectors expressing Gag, Env, and a Pol-Tat-Nef-Vif fusion protein prior to boosting with the HSV vectors. Robust anti-Gag and anti-Env cellular responses were detected in all six macaques. Following intravenous challenge with wild-type, cloned SIV239, peak and 12-week plasma viremia levels were significantly lower in vaccinated compared to control macaques. Plasma SIV RNA in vaccinated macaques was inversely correlated with anti-Rev ELISPOT responses on the day of challenge (P value<0.05), anti-Tat ELISPOT responses at 2 weeks post challenge (P value <0.05) and peak neutralizing antibody titers pre-challenge (P value 0.06). These findings support continued study of recombinant herpesviruses as a vaccine approach for AIDS.
Collapse
Affiliation(s)
- Amitinder Kaur
- New England Primate Research Center, Harvard Medical School, One Pine Hill Drive, P.O. Box 9102, Southborough, MA 01772-9102, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Davis D, Donners H, Willems B, Ntemgwa M, Vermoesen T, van der Groen G, Janssens W. Neutralization kinetics of sensitive and resistant subtype B primary human immunodeficiency virus type 1 isolates. J Med Virol 2006; 78:864-76. [PMID: 16721864 DOI: 10.1002/jmv.20635] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The aim of the study was to determine if sensitive and resistant human immunodeficiency virus type 1 (HIV-1) subtype B primary isolates have different neutralization kinetics. Neutralization assays were undertaken where either the time allowed for virus to react with antibodies or the subsequent period of this mixture's exposure to target cells were varied. The relative neutralization sensitivity/resistance is a reproducible property of the isolates. In a minority of combinations, the titre falls exponentially for as long as the free virions are exposed to antibody. In the remainder, neutralization kinetics shows deviations which may be attributed to events occurring after the virus-antibody mixture is added to the target cells: significant neutralization with minimal exposure of the free virions to antibody; a plot where reduction in virus titre is parallel to the duration of the incubation phase of the assay. Neutralization rate constants are similar for primary HIV-1 SF33, HIV-1 SF162, and HIV-1 89.6, reaching 5 x 10(5)-1 x 10(6)/M sec for the monoclonal antibody IgG1 b12. However, although increased antibody levels produced greater reductions in virus titre the rate of neutralization was not proportional to the antibody concentration. Neutralization of either the free virion or cell-associated virus does not correlate with the resistance/sensitivity of primary subtype B isolates. The target cells play an active role, so that in designing neutralization assays with primary isolates of HIV-1, events following the virus-antibody complex binding to the cell surface have to be taken into consideration.
Collapse
Affiliation(s)
- David Davis
- Department of Microbiology, Virology Unit, Institute of Tropical Medicine, Nationalestraat, Antwerp, Belgium.
| | | | | | | | | | | | | |
Collapse
|
37
|
Yuste E, Sanford HB, Carmody J, Bixby J, Little S, Zwick MB, Greenough T, Burton DR, Richman DD, Desrosiers RC, Johnson WE. Simian immunodeficiency virus engrafted with human immunodeficiency virus type 1 (HIV-1)-specific epitopes: replication, neutralization, and survey of HIV-1-positive plasma. J Virol 2006; 80:3030-41. [PMID: 16501112 PMCID: PMC1395451 DOI: 10.1128/jvi.80.6.3030-3041.2006] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
To date, only a small number of anti-human immunodeficiency virus type 1 (HIV-1) monoclonal antibodies (MAbs) with relatively broad neutralizing activity have been isolated from infected individuals. Adequate techniques for defining how frequently antibodies of these specificities arise in HIV-infected people have been lacking, although it is generally assumed that such antibodies are rare. In order to create an epitope-specific neutralization assay, we introduced well-characterized HIV-1 epitopes into the heterologous context of simian immunodeficiency virus (SIV). Specifically, epitope recognition sequences for the 2F5, 4E10, and 447-52D anti-HIV-1 neutralizing monoclonal antibodies were introduced into the corresponding regions of SIVmac239 by site-directed mutagenesis. Variants with 2F5 or 4E10 recognition sequences in gp41 retained replication competence and were used for neutralization assays. The parental SIVmac239 and the neutralization-sensitive SIVmac316 were not neutralized by the 2F5 and 4E10 MAbs, nor were they neutralized significantly by any of the 96 HIV-1-positive human plasma samples that were tested. The SIV239-2F5 and SIV239-4E10 variants were specifically neutralized by the 2F5 and 4E10 MAbs, respectively, at concentrations within the range of what has been reported previously for HIV-1 primary isolates (J. M. Binley et al., J. Virol. 78:13232-13252, 2004). The SIV239-2F5 and SIV239-4E10 epitope-engrafted variants were used as biological screens for the presence of neutralizing activity of these specificities. None of the 92 HIV-1-positive human plasma samples that were tested exhibited significant neutralization of SIV239-2F5. One plasma sample exhibited >90% neutralization of SIV239-4E10, but this activity was not competed by a 4E10 target peptide and was not present in concentrated immunoglobulin G (IgG) or IgA fractions. We thus confirm by direct analysis that neutralizing activities of the 2F5 and 4E10 specificities are either rare among HIV-1-positive individuals or, if present, represent only a very small fraction of the total neutralizing activity in any given plasma sample. We further conclude that the structures of gp41 from SIVmac239 and HIV-1 are sufficiently similar such that epitopes engrafted into SIVmac239 can be readily recognized by the cognate anti-HIV-1 monoclonal antibodies.
Collapse
Affiliation(s)
- Eloisa Yuste
- New England Primate Research Center, Department of Microbiology and Molecular Genetics, Harvard Medical Scool, One Pine Hill Drive, Box 9102, Southborough, Massachusetts 01772-9102, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Laurén A, Thorstensson R, Fenyö EM. Comparative studies on mucosal and intravenous transmission of simian immunodeficiency virus (SIVsm): the kinetics of evolution to neutralization resistance are related to progression rate of disease. J Gen Virol 2006; 87:595-606. [PMID: 16476980 DOI: 10.1099/vir.0.81409-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The kinetics of appearance of autologous neutralizing antibodies were studied in cynomolgus macaques infected with simian immunodeficiency virus (SIVsm) by the intravenous (IV) route (six monkeys) or the intrarectal (IR) route (ten monkeys). The SIVsm inoculum virus and reisolates obtained at 2 weeks, 3 or 4 months and later than 1 year were tested in a GHOST(3) cell line-based plaque-reduction assay with autologous sera collected at the same sampling times. All monkeys developed a neutralizing-antibody response to the inoculum virus, those infected by the IV route earlier than monkeys infected by the IR route. Animals were divided into progressor (P), slow-progressor (SP) and long-term non-progressor (LTNP) monkeys, based on progression rate. In P monkeys, neutralization escape could be demonstrated by 3 months post-infection. Neutralization-resistant variants also emerged in SP and LTNP monkeys, but were much delayed compared with P monkeys. Evolution of neutralization resistance was also demonstrated by a positive-control serum in the heterologous reaction. Pooled sera from four LTNP monkeys showed a broad neutralizing capacity, including neutralization of escape variants. These results from a large group of infected monkeys showed that SIV evolves to neutralization resistance in the infected host and that the kinetics of this evolution are related to the route of transmission and the progression rate of SIV disease. The results suggest an important role for neutralizing antibodies in controlling viraemia. Although this control is transient in the infected host, neutralization resistance is relative and variant viruses may be neutralized by a broadly cross-neutralizing serum pool.
Collapse
Affiliation(s)
- Anna Laurén
- Division of Medical Microbiology, Department of Laboratory Medicine, Lund University, Sölvegatan 23, 223 62 Lund, Sweden
| | | | - Eva Maria Fenyö
- Division of Medical Microbiology, Department of Laboratory Medicine, Lund University, Sölvegatan 23, 223 62 Lund, Sweden
| |
Collapse
|
39
|
Xu R, Srivastava IK, Kuller L, Zarkikh I, Kraft Z, Fagrouch Z, Letvin NL, Heeney JL, Barnett SW, Stamatatos L. Immunization with HIV-1 SF162-derived Envelope gp140 proteins does not protect macaques from heterologous simian-human immunodeficiency virus SHIV89.6P infection. Virology 2006; 349:276-89. [PMID: 16527321 DOI: 10.1016/j.virol.2006.01.043] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2005] [Revised: 01/19/2006] [Accepted: 01/26/2006] [Indexed: 11/21/2022]
Abstract
Immunization by the SF162gp140 or the DeltaV2gp140 HIV-1 envelope proteins results in the generation of strong homologous neutralizing antibodies (NAbs) that offer similar degree of protection from disease-development to macaques challenged with homologous virus. These two immunogens elicit weak cross-reactive NAbs and their effectiveness against heterologous challenge is currently unknown. To examine this issue, we immunized macaques with SIVGag p55 and either the SF162gp140 or the DeltaV2gp140 and challenged them intravenously with SHIV-89.6P. All animals became infected but previous immunization with SF162gp140 accelerated the development of anti-SHIV89.6P neutralizing antibody responses following infection. DeltaV2gp140 is derived from SF162gp140 following the deletion of 30 amino acids and one N-linked glycosylation site from the V2 loop. Our results suggest that even small differences in HIV Envelope immunogen structure can affect the neutralizing antibody responses generated following infection.
Collapse
Affiliation(s)
- Rong Xu
- Seattle Biomedical Research Institute, Seattle, WA 98109, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Mao H, Lafont BAP, Igarashi T, Nishimura Y, Brown C, Hirsch V, Buckler-White A, Sadjadpour R, Martin MA. CD8+ and CD20+ lymphocytes cooperate to control acute simian immunodeficiency virus/human immunodeficiency virus chimeric virus infections in rhesus monkeys: modulation by major histocompatibility complex genotype. J Virol 2006; 79:14887-98. [PMID: 16282488 PMCID: PMC1287589 DOI: 10.1128/jvi.79.23.14887-14898.2005] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
We have previously described two isogenic molecularly cloned simian immunodeficiency virus/human immunodeficiency virus chimeric viruses (SHIVs) that differ from one another by 9 amino acids and direct distinct clinical outcomes in inoculated rhesus monkeys. SHIV(DH12R-Clone 7), like other highly pathogenic CXCR4-tropic SHIVs, induces rapid and complete depletions of CD4+ T lymphocytes and immunodeficiency in infected animals. In contrast, macaques inoculated with SHIV(DH12R-Clone 8) experience only partial and transient losses of CD4+ T cells, show prompt control of their viremia, and remain healthy for periods of time extending for up to 4 years. The contributions of CD8+ and CD20+ lymphocytes in suppressing the replication of the attenuated SHIV(DH12R-Clone 8) and maintaining a prolonged asymptomatic clinical course was assessed by treating animals with monoclonal antibodies that deplete each lymphocyte subset at the time of virus inoculation. The absence of either CD8+ or CD20+ cells during the SHIV(DH12R-Clone 8) acute infection resulted in the rapid, complete, and irreversible loss of CD4+ T cells; sustained high levels of postpeak plasma viremia; and symptomatic disease in Mamu-A*01-negative Indian rhesus monkeys. In Mamu-A*01-positive animals, however, the aggressive, highly pathogenic phenotype was observed only in macaques depleted of CD8+ cells; SHIV(DH12R-Clone 8) was effectively controlled in Mamu-A*01-positive monkeys in the absence of B lymphocytes. Taken together, these results indicate that both CD8+ and CD20+ B cells contribute to the control of primate lentiviral infection in Mamu-A*01-negative macaques. Furthermore, the major histocompatibility complex genotype of an infected animal, as exemplified by the Mamu-A*01 allele in this study, has the additional capacity to shift the balance of the composite immune response.
Collapse
Affiliation(s)
- Hanwen Mao
- Laboratory of Molecular Microbiology, NIAID, NIH, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Pikora CA, Wittish C, Desrosiers RC. p6gag of human and simian immunodeficiency viruses is tolerant to small in-frame deletions downstream of the late domain. Virology 2005; 346:479-89. [PMID: 16332383 DOI: 10.1016/j.virol.2005.10.040] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2005] [Accepted: 10/25/2005] [Indexed: 10/25/2022]
Abstract
Short, unique, in-frame deletions were consistently detected within p6gag sequences obtained over time from three of eight HIV-1-infected long-term nonprogressors (Alexander, L., Weiskopf, E., Greenough, T.C., Gaddis, N.C., Auerbach, M.R., Malim, M.H., O'Brien, S.J., Walker, B.D., Sullivan, J.L., Desrosiers, R.C., 2000. Unusual polymorphisms in Human Immunodeficiency Virus Type 1 associated with nonprogressive infection. J. Virol. 74, 4361-4376). Using PCR mutagenesis, we created 11 mutant forms of SIV239 and 8 mutant forms of HIV-1 NL4-3 with serial 2 amino acid deletions within p6gag downstream of the PTAP late domain. Nine of the 11 SIV239 mutants assembled and released virion particles similar to wild-type, displayed wild-type infectivity, and replicated similar to wild-type SIV239 in cultured cells. Two of the 11 SIV239 mutants, both involving D at position 21, were grossly defective for intracellular gag accumulation and did not replicate detectably in cultured cells. Similar to the 9 SIV239 mutants, 7 of the 8 HIV-1 mutants replicated well in cultured cells. Only the mutant deleted of ES at positions 19 and 20, immediately adjacent to the PTAP sequence, was markedly impaired in its replicative capacity. These results demonstrate an overall high tolerance of SIV and HIV to two amino acid deletions within p6gag downstream of the late domain.
Collapse
Affiliation(s)
- Cheryl A Pikora
- New England Primate Research Center, Harvard Medical School, One Pine Hill Drive, Southborough, MA 01772, USA
| | | | | |
Collapse
|
42
|
Mori K, Sugimoto C, Ohgimoto S, Nakayama EE, Shioda T, Kusagawa S, Takebe Y, Kano M, Matano T, Yuasa T, Kitaguchi D, Miyazawa M, Takahashi Y, Yasunami M, Kimura A, Yamamoto N, Suzuki Y, Nagai Y. Influence of glycosylation on the efficacy of an Env-based vaccine against simian immunodeficiency virus SIVmac239 in a macaque AIDS model. J Virol 2005; 79:10386-96. [PMID: 16051831 PMCID: PMC1182680 DOI: 10.1128/jvi.79.16.10386-10396.2005] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The envelope glycoprotein (Env) of human immunodeficiency viruses (HIVs) and simian immunodeficiency viruses (SIVs) is heavily glycosylated, and this feature has been speculated to be a reason for the insufficient immune control of these viruses by their hosts. In a macaque AIDS model, we demonstrated that quintuple deglycosylation in Env altered a pathogenic virus, SIVmac239, into a novel attenuated mutant virus (delta5G). In delta5G-infected animals, strong protective immunity against SIVmac239 was elicited. These HIV and SIV studies suggested that an understanding of the role of glycosylation is critical in defining not only the virological properties but also the immunogenicity of Env, suggesting that glycosylation in Env could be modified for the development of effective vaccines. To examine the effect of deglycosylation, we constructed prime-boost vaccines consisting of Env from SIVmac239 and delta5G and compared their immunogenicities and vaccine efficacies by challenge infection with SIVmac239. Vaccination-induced immune responses differed between the two vaccine groups. Both Env-specific cellular and humoral responses were higher in wild-type (wt)-Env-immunized animals than in delta5G Env-immunized animals. Following the challenge, viral loads in SIVmac239 Env (wt-Env)-immunized animals were significantly lower than in vector controls, with controlled viral replication in the chronic phase. Unexpectedly, viral loads in delta5G Env-immunized animals were indistinguishable from those in vector controls. This study demonstrated that the prime-boost Env vaccine was effective against homologous SIVmac239 challenge. Changes in glycosylation affected both cell-mediated and humoral immune responses and vaccine efficacy.
Collapse
Affiliation(s)
- Kazuyasu Mori
- AIDS Research Center, National Institute of Infectious Diseases, Tokyo, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Saunders CJ, McCaffrey RA, Zharkikh I, Kraft Z, Malenbaum SE, Burke B, Cheng-Mayer C, Stamatatos L. The V1, V2, and V3 regions of the human immunodeficiency virus type 1 envelope differentially affect the viral phenotype in an isolate-dependent manner. J Virol 2005; 79:9069-80. [PMID: 15994801 PMCID: PMC1168758 DOI: 10.1128/jvi.79.14.9069-9080.2005] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
It is well documented that removal of the V1V2 region or of the V2 loop alone from the envelope glycoprotein of human immunodeficiency virus type 1 (HIV-1) or simian immunodeficiency virus (SIV) increases the susceptibility of these viruses to neutralization by antibodies. The specific role of the V1 loop in defining the neutralization susceptibility of HIV is, however, not well documented. Our current studies indicate that although the V1V2 region is a global modulator of the HIV-1 neutralization susceptibility, the individual roles the V1 and V2 loops have in defining the neutralization susceptibility profile of HIV-1 differ and in some cases are opposite. While deletion of the V2 loop renders the virus more susceptible to neutralization by antibodies that recognize diverse epitopes, in particular certain ones located in the CD4 binding site and the V3 loop, deletion of the V1 loop renders the virus refractory to neutralization, especially by antibodies that recognize CD4-induced epitopes and certain CD4-site binding antibodies. Our current studies also indicate that the relative involvement of the V2 loop of the HIV-1 envelope during virus-cell entry appears to be envelope background dependent. As a result, although deletion of the V2 loop from the clade B, R5-tropic SF162 HIV-1 virus resulted in a virus that was replication competent, the same modification introduced on the background of two other R5-tropic isolates, SF128A (clade B) or SF170 (clade A), abrogated the ability of these envelopes to mediate virus-cell entry.
Collapse
Affiliation(s)
- Cheryl J Saunders
- Seattle Biomedical Research Institute, 307 Westlake Avenue North, Seattle, WA 98109, USA
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Borda JT, Alvarez X, Kondova I, Aye P, Simon MA, Desrosiers RC, Lackner AA. Cell tropism of simian immunodeficiency virus in culture is not predictive of in vivo tropism or pathogenesis. THE AMERICAN JOURNAL OF PATHOLOGY 2005; 165:2111-22. [PMID: 15579453 PMCID: PMC1618703 DOI: 10.1016/s0002-9440(10)63261-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
SIVmac239/316 is a molecular clone derived from SIVmac239 that differs from the parental virus by nine amino acids in env. This virus, unlike the parental SIVmac239, is able to replicate well in alveolar macrophages in culture. We have not however, observed macrophage-associated inflammatory disease in any animal infected with SIVmac239/316. Therefore, we sought to examine the cell tropism of this virus in vivo in multiple tissues using in situ hybridization combined with immunohistochemistry and multilabel confocal microscopy for viral nucleic acid and multiple cell-type-specific markers for macrophages and T lymphocytes. Tissues examined included brain, heart, lung, lymph nodes, spleen, thymus, and small and large intestine. Matched tissues from macaques infected with the parental SIVmac239 and uninfected macaques were also examined. Many infected cells were detected in the tissues of animals infected with SIVmac239 and SIVmac239/316 although there appeared to be fewer positive cells in animals infected with SIVmac239/316. Surprisingly, in light of the cell culture observations, nearly every simian immunodeficiency virus-infected cell in animals inoculated with SIVmac239/316 was a T lymphocyte rather than a macrophage. This was true both during early infection (first 2 months) and in terminal disease. In contrast, as previously described, SIVmac239 was found in both T cells and macrophages in tissues as early as 21 days after infection. These studies indicate that during both acute and chronic SIVmac239/316 infection T lymphocytes rather than macrophages are the principal targets in vivo. These data combined with the absence of macrophage-associated lesions in SIVmac239/316-infected animals indicate that in vitro cell tropism is not predictive of in vivo tropism or disease pathogenesis.
Collapse
Affiliation(s)
- Juan T Borda
- Tulane National Primate Research Center, Tulane University, Covington, LA 70433, USA
| | | | | | | | | | | | | |
Collapse
|
45
|
Swigut T, Alexander L, Morgan J, Lifson J, Mansfield KG, Lang S, Johnson RP, Skowronski J, Desrosiers R. Impact of Nef-mediated downregulation of major histocompatibility complex class I on immune response to simian immunodeficiency virus. J Virol 2004; 78:13335-44. [PMID: 15542684 PMCID: PMC525019 DOI: 10.1128/jvi.78.23.13335-13344.2004] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Functional activities that have been ascribed to the nef gene product of simian immunodeficiency virus (SIV) and human immunodeficiency virus (HIV) include CD4 downregulation, major histocompatibility complex (MHC) class I downregulation, downregulation of other plasma membrane proteins, and lymphocyte activation. Monkeys were infected experimentally with SIV containing difficult-to-revert mutations in nef that selectively eliminated MHC downregulation but not these other activities. Monkeys infected with these mutant forms of SIV exhibited higher levels of CD8(+) T-cell responses 4 to 16 weeks postinfection than seen in monkeys infected with the parental wild-type virus. Furthermore, unusual compensatory mutations appeared by 16 to 32 weeks postinfection which restored some or all of the MHC-downregulating activity. These results indicate that nef does serve to limit the virus-specific CD8 cellular response of the host and that the ability to downregulate MHC class I contributes importantly to the totality of nef function.
Collapse
Affiliation(s)
- Tomek Swigut
- New England Primate Research Center, One Pine Hill Drive, Box 9102, Southborough, MA 01772-9102, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Yuste E, Reeves JD, Doms RW, Desrosiers RC. Modulation of Env content in virions of simian immunodeficiency virus: correlation with cell surface expression and virion infectivity. J Virol 2004; 78:6775-85. [PMID: 15194752 PMCID: PMC421677 DOI: 10.1128/jvi.78.13.6775-6785.2004] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Specific mutations were created in the cytoplasmic domain of the gp41 transmembrane protein of simian immunodeficiency virus strain 239 (SIV239). The resultant strains included a mutant in which Env residue 767 was changed to a stop codon, a double mutant in which positions 738 and 739 were changed to stop codons, another mutant in which a prominent endocytosis motif was changed from YRPV to GRPV by the substitution of tyrosine 721, and a final combination mutant bearing Q738stop, Q739stop, and Y721G mutations. The effects of these mutations on cell surface expression, on Env incorporation into virions, and on viral infectivity were examined. The molar ratio of Gag to gp120 of 54:1 that we report here for SIV239 virions agrees very well with the ratio of 60:1 reported previously by Chertova et al. (E. Chertova, J. W. Bess, Jr., B. J. Crise, R. C. Sowder II, T. M. Schaden, J. M. Hilburn, J. A. Hoxie, R. E. Benveniste, J. D. Lifson, L. E. Henderson, and L. O. Arthur, J. Virol. 76:5315-5325, 2002), although they were determined by very different methodologies. Assuming 1,200 to 2,500 Gag molecules per virion, this corresponds to 7 to 16 Env trimers per SIV239 virion particle. Although all of the mutations increased Env levels in virions, E767stop had the most dramatic effect, increasing the Env content per virion 25- to 50-fold. Increased levels of Env content in virions correlated strictly with higher levels of Env expression on the cell surface. The increased Env content with the E767stop mutation also correlated with an increased infectivity, but the degree of change was not proportional: the 25- to 50-fold increase in Env content only increased infectivity 2- to 3-fold. All of the mutants replicated efficiently in the CEMx174 and Rh221-89 cell lines. Although some of these findings have been reported previously, our findings show that the effects of the cytoplasmic domain of gp41 on the Env content in virions can be dramatic, that the Env content in virions correlates strictly with the levels of cell surface expression, and that the Env content in virions can determine infectivity; furthermore, our results define a particular change with the most dramatic effects.
Collapse
Affiliation(s)
- Eloísa Yuste
- New England Primate Research Center, Harvard Medical School, Southborough, MA 01772-9102, USA
| | | | | | | |
Collapse
|
47
|
Pugach P, Kuhmann SE, Taylor J, Marozsan AJ, Snyder A, Ketas T, Wolinsky SM, Korber BT, Moore JP. The prolonged culture of human immunodeficiency virus type 1 in primary lymphocytes increases its sensitivity to neutralization by soluble CD4. Virology 2004; 321:8-22. [PMID: 15033560 DOI: 10.1016/j.virol.2003.12.012] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2003] [Revised: 12/15/2003] [Accepted: 12/15/2003] [Indexed: 10/26/2022]
Abstract
Primary strains of human immunodeficiency virus type 1 (HIV-1) are known to adapt to replication in cell lines in vitro by becoming sensitive to soluble CD4 (sCD4) and neutralizing antibodies (NAb). T-cell lines favor isolation of variants that use CXCR4 as a co-receptor, while primary isolates predominantly use CCR5. We have now studied how a primary R5 isolate, CC1/85, adapts to prolonged replication in primary human peripheral blood mononuclear cells (PBMC). After 19 passages, a variant virus, CCcon.19, had increased sensitivity to both sCD4 and NAb b12 that binds to a CD4-binding site (CD4BS)-associated epitope, but decreased sensitivity to anti-CD4 antibodies. CCcon.19 retains the R5 phenotype, its resistance to other NAbs was unaltered, its sensitivity to various entry inhibitors was unchanged, and its ability to replicate in macrophages was modestly increased. We define CCcon.19 as a primary T-cell adapted (PTCA) variant. Genetic sequence analysis combined with mutagenesis studies on clonal, chimeric viruses derived from CC1/85 and the PTCA variant showed that the most important changes were in the V1/V2 loop structure, one of them involving the loss of an N-linked glycosylation site. Monomeric gp120 proteins expressed from CC1/85 and the PTCA variant did not differ in their affinities for sCD4, suggesting that the structural consequences of the sequence changes were manifested at the level of the native, trimeric Env complex. Overall, the adaptation process probably involves selection for variants with higher CD4 affinity and hence greater fusion efficiency, but this also involves the loss of some resistance to neutralization by agents directed at or near to the CD4BS. The loss of neutralization resistance is of no relevance under in vitro conditions, but NAbs would presumably be a counter-selection pressure against such adaptive changes in vivo, at least when the humoral immune response is intact.
Collapse
Affiliation(s)
- Pavel Pugach
- Department of Microbiology and Immunology, Weill Medical College of Cornell University, New York, NY 10021, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Zhang ZQ, Schleif WA, Casimiro DR, Handt L, Chen M, Davies ME, Liang X, Fu TM, Tang A, Wilson KA, McElhaugh M, Carella A, Tan C, Connolly B, Hill S, Klein H, Emini EA, Shiver JW. The impact of early immune destruction on the kinetics of postacute viral replication in rhesus monkey infected with the simian-human immunodeficiency virus 89.6P. Virology 2004; 320:75-84. [PMID: 15003864 DOI: 10.1016/j.virol.2003.11.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2003] [Revised: 10/31/2003] [Accepted: 11/03/2003] [Indexed: 10/26/2022]
Abstract
Set-point viral load is positively correlated with the extent of initial viral replication in pathogenic simian-human immunodeficiency virus (SHIV) infection. To elucidate the mechanisms underlying the correlation, we conducted a systematic investigation in rhesus monkeys infected with the highly pathogenic SHIV 89.6P. This model is widely used in the preclinical evaluation of AIDS vaccine candidates and a thorough understanding of the model's biology is important to the proper interpretation of these evaluations. We found that the levels of peak viremia were positively correlated not only with the levels of set-point viremia but, importantly, with the extent of initial overall immune destruction as indicated by the degree of CD4+ T cell depletion and lymph node germinal center (GC) formation. The extent of initial overall immune destruction was inversely correlated with subsequent development and maintenance of virus-specific cellular and humoral immune responses. Thus, these data suggest that the extent of early immune damage determines the development and durability of virus-specific immunity, thereby playing a critical role in establishing the levels of set-point viral replication in SHIV infection. Vaccines that limit both the initial viral replication and the extent of early immune damage will therefore mediate long-term virus replication control and mitigation of long-term immune destruction in this model of immunodeficiency virus infection.
Collapse
Affiliation(s)
- Zhi-Qiang Zhang
- Department of Viral Vaccine Research, Merck Research Laboratories, West Point, PA 19486, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Buckner C, Gines LG, Saunders CJ, Vojtech L, Srivastava I, Gettie A, Bohm R, Blanchard J, Barnett SW, Safrit JT, Stamatatos L. Priming B cell-mediated anti-HIV envelope responses by vaccination allows for the long-term control of infection in macaques exposed to a R5-tropic SHIV. Virology 2004; 320:167-80. [PMID: 15003872 DOI: 10.1016/j.virol.2003.12.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2003] [Revised: 12/02/2003] [Accepted: 12/03/2003] [Indexed: 11/18/2022]
Abstract
The potential of vaccine-elicited anti-HIV envelope antibodies to control HIV-infection was evaluated by immunizing macaques with the HIV envelope protein and transiently depleting them of their CD8+ cells before intravenous challenge with the pathogenic CCR5-tropic SIV/HIV chimeric virus, SHIV(SF162P4). Although sterilizing immunity was not achieved, all vaccinated animals effectively controlled infection and remained free of disease for the duration of observation (over 3 years). In contrast, during the same period, the control animals progressed to disease. Both the vaccinees and the controls developed robust cell-mediated antiviral and neutralizing antibody responses following infection. A comparative analysis of these responses suggests that the more effective long-term control of infection by the vaccinated animals is due to the more rapid development of anti-HIV envelope antibodies. These studies suggest that priming by vaccination of B cell anti-HIV envelope responses maybe crucial for the long-term control of HIV infection.
Collapse
Affiliation(s)
- Clarisa Buckner
- Aaron Diamond AIDS Research Center, The Rockefeller University, New York, NY 10016, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Negri DRM, Baroncelli S, Catone S, Comini A, Michelini Z, Maggiorella MT, Sernicola L, Crostarosa F, Belli R, Mancini MG, Farcomeni S, Fagrouch Z, Ciccozzi M, Boros S, Liljestrom P, Norley S, Heeney J, Titti F. Protective efficacy of a multicomponent vector vaccine in cynomolgus monkeys after intrarectal simian immunodeficiency virus challenge. J Gen Virol 2004; 85:1191-1201. [PMID: 15105535 DOI: 10.1099/vir.0.79794-0] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
We investigated the protective efficacy of a systemic triple vector (DNA/rSFV/rMVA)-based vaccine against mucosal challenge with pathogenic simian immunodeficiency virus (SIV) in cynomolgus monkeys. Animals were immunized at week 0 with DNA (intradermally), at weeks 8 and 16 with recombinant Semliki Forest virus (rSFV, subcutaneously) and finally, at week 24, with recombinant modified vaccinia virus Ankara strain (rMVA, intramuscularly). Both DNA and recombinant viral vectors expressed a wide range of SIV proteins (Gag, Pol, Tat, Rev, Env and Nef). This immunization strategy elicited cell-mediated rather than humoral responses that were especially increased following the last boost. Upon intrarectal challenge with pathogenic SIVmac251, three of the four vaccinated monkeys dramatically abrogated virus load to undetectable levels up to 41 weeks after challenge. A major contribution to this vaccine effect appeared to be the T-cell-mediated immune response to vaccine antigens (Gag, Rev, Tat, Nef) seen in the early phase of infection in three of the four vaccinated monkeys. Indeed, the frequency of T-cells producing antigen-induced IFN-γ mirrored virus clearance in the vaccinated and protected monkeys. These results, reminiscent of the efficacy of live attenuated virus vaccines, suggest that vaccination with a combination of many viral antigens can induce a robust and stable vaccine-induced immunity able to abrogate virus replication.
Collapse
Affiliation(s)
- Donatella R M Negri
- Laboratory of Virology, Istituto Superiore di Sanità, Viale R. Elena 299, 00161 Rome, Italy
| | - Silvia Baroncelli
- Laboratory of Virology, Istituto Superiore di Sanità, Viale R. Elena 299, 00161 Rome, Italy
| | - Stefania Catone
- Laboratory of Virology, Istituto Superiore di Sanità, Viale R. Elena 299, 00161 Rome, Italy
| | - Antonella Comini
- Laboratory of Virology, Istituto Superiore di Sanità, Viale R. Elena 299, 00161 Rome, Italy
| | - Zuleika Michelini
- Laboratory of Virology, Istituto Superiore di Sanità, Viale R. Elena 299, 00161 Rome, Italy
| | - Maria T Maggiorella
- Laboratory of Virology, Istituto Superiore di Sanità, Viale R. Elena 299, 00161 Rome, Italy
| | - Leonardo Sernicola
- Laboratory of Virology, Istituto Superiore di Sanità, Viale R. Elena 299, 00161 Rome, Italy
| | - Federica Crostarosa
- Laboratory of Virology, Istituto Superiore di Sanità, Viale R. Elena 299, 00161 Rome, Italy
| | - Roberto Belli
- Laboratory of Virology, Istituto Superiore di Sanità, Viale R. Elena 299, 00161 Rome, Italy
| | - Maria G Mancini
- Laboratory of Virology, Istituto Superiore di Sanità, Viale R. Elena 299, 00161 Rome, Italy
| | - Stefania Farcomeni
- Laboratory of Virology, Istituto Superiore di Sanità, Viale R. Elena 299, 00161 Rome, Italy
| | - Zahra Fagrouch
- Department of Virology, Biomedical Primate Research Center, PO Box 3306, 2280 GH Rijswijk, The Netherlands
| | - Massimo Ciccozzi
- Laboratory of Epidemiology and Biostatistics, Istituto Superiore di Sanità, Viale R. Elena 299, 00161 Rome, Italy
| | - Stefano Boros
- Laboratory of Epidemiology and Biostatistics, Istituto Superiore di Sanità, Viale R. Elena 299, 00161 Rome, Italy
| | - Peter Liljestrom
- Microbiology and Tumor-Biology Center, Karolinska Institute, Box 280, S-17177 Stockholm, Sweden
| | - Stephen Norley
- AIDS Immunopathogenesis and Vaccine Development, Robert Koch Institute, 13353 Berlin, Germany
| | - Jonathan Heeney
- Department of Virology, Biomedical Primate Research Center, PO Box 3306, 2280 GH Rijswijk, The Netherlands
| | - Fausto Titti
- Laboratory of Virology, Istituto Superiore di Sanità, Viale R. Elena 299, 00161 Rome, Italy
| |
Collapse
|