Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Viruses ; 12(2)2020 01 31.
Article in English | MEDLINE | ID: mdl-32023860

ABSTRACT

Non-human primates (NHP) are the only animal model suitable to evaluate the protection efficacy of HIV-1 vaccines. It is important to understand how and when neutralizing antibodies (nAbs) with specificities similar to those of human broadly neutralizing antibodies (bnAbs) develop in NHPs. To address these questions, we determined plasma neutralization specificities in two macaques which developed neutralization breadth after long-term simian/human immunodeficiency virus (SHIV) infection and identified neutralization escape mutations by analyzing the env sequences from longitudinal plasma samples. Neutralization activities targeting V2, CD4bs, V3 and gp120-gp41 interface only became detectable in week 350 plasma from macaques G1015R and G1020R using 25710 env mutants. When mapped with CAP45 env mutants, only V2 specificity was detected at week 217 and persisted until week 350 in G1015R. Neutralization escape mutations were found in CD4bs and V2 regions. However, all of them were different from those resistant mutations identified for human bnAbs. These results show that nAbs with specificities similar to human bnAbs are only detectable after long-term SHIV infection and that neutralization escape mutations in macaques are different from those found in HIV-1-infected individuals. These findings can have important implications in the best utilization of the NHP model to evaluate HIV-1 vaccines.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , HIV Antibodies/immunology , HIV-1/immunology , Simian Immunodeficiency Virus/immunology , Animals , Antibodies, Neutralizing/biosynthesis , Genes, env/genetics , Genes, env/immunology , HIV Antibodies/biosynthesis , HIV Seropositivity , Macaca mulatta/immunology , Mutation , Neutralization Tests , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/genetics , Time Factors
2.
J Virol ; 93(12)2019 06 15.
Article in English | MEDLINE | ID: mdl-30944182

ABSTRACT

Most broadly neutralizing antibodies and many entry inhibitors target the pretriggered (state 1) conformation of the human immunodeficiency virus type 1 (HIV-1) envelope glycoprotein (Env). Here we examine two previously reported Env mutants designed to be stabilized in this conformation by the introduction of artificial disulfide bonds: A501C/T605C (called SOS) and I201C/A433C (called DS). SOS Env supported virus entry and cell-cell fusion only after exposure to a reducing agent, dithiothreitol (DTT). Deletion of the Env cytoplasmic tail improved the efficiency with which the SOS Env supported virus infection in a reducing environment. The antigenicity of the SOS Env was similar to that of the unmodified Env, except for greater sensitivity to some state 1-preferring ligands. In contrast, viruses with the DS Env were not infectious, even after DTT treatment. The proteolytic maturation of the DS Env on both cell surfaces and virions was severely compromised compared with that of the unmodified Env. The DS Env exhibited detectable cell-fusing activity when DTT was present. However, the profiles of cell-surface Env recognition and cell-cell fusion inhibition by antibodies differed for the DS Env and the unmodified Env. Thus, the DS Env appears to be stabilized in an off-pathway conformation that is nonfunctional on the virus. The SOS change exerted more subtle, context-dependent effects on Env conformation and function.IMPORTANCE The human immunodeficiency virus type 1 (HIV-1) envelope proteins (Envs) bind receptors on the host cell and change shape to allow the virus to enter the cell. Most virus-inhibiting antibodies and drugs recognize a particular shape of Env called state 1. Disulfide bonds formed by cysteine residues have been introduced into soluble forms of the flexible envelope proteins in an attempt to lock them into state 1 for use in vaccines and as research tools. We evaluated the effect of these cysteine substitutions on the ability of the membrane Env to support virus entry and on susceptibility to inhibition by antibodies and small molecules. We found that the conformation of the envelope proteins with the cysteine substitutions differed from that of the unmodified membrane envelope proteins. Awareness of these effects can assist efforts to create stable HIV-1 Env complexes that more closely resemble the state 1 conformation.


Subject(s)
HIV-1/immunology , env Gene Products, Human Immunodeficiency Virus/genetics , env Gene Products, Human Immunodeficiency Virus/immunology , Antibodies, Neutralizing/immunology , Disulfides/immunology , Genes, env/genetics , Genes, env/immunology , HEK293 Cells , HIV Antibodies/immunology , HIV Envelope Protein gp120/immunology , HIV Envelope Protein gp120/metabolism , HIV Envelope Protein gp41/immunology , HIV Envelope Protein gp41/metabolism , HIV-1/metabolism , Humans , Membrane Glycoproteins/immunology , Protein Conformation , Structure-Activity Relationship , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Virus Internalization , env Gene Products, Human Immunodeficiency Virus/metabolism
3.
J Virol ; 92(5)2018 03 01.
Article in English | MEDLINE | ID: mdl-29237828

ABSTRACT

The subtype C HIV-1 isolate MW965.26 is a highly neutralization-sensitive tier 1a primary isolate that is widely used in vaccine studies, but the basis for the sensitive neutralization phenotype of this isolate is not known. Substituting the MW965.26 V1/V2 domain into a neutralization-sensitive SF162 Env clone resulted in high resistance to standard anti-V3 monoclonal antibodies, demonstrating that this region possesses strong masking activity in a standard Env backbone and indicating that determinants elsewhere in MW965.26 Env are responsible for its unusual neutralization sensitivity. Key determinants for this phenotype were mapped by generating chimeric Envs between MW965.26 Env and a typical resistant Env clone, the consensus C (ConC) clone, and localized to two residues, Cys384 in the C3 domain and Asn502 in the C5 domain. Substituting the sensitizing mutations Y384C and K502N at these positions into several resistant primary Envs resulted in conversion to neutralization-sensitive phenotypes, demonstrating the generalizability of this effect. In contrast to the sensitizing effects of these substitutions on normally masked epitopes, these mutations reduced the sensitivity of VRC01-like epitopes overlapping the CD4-binding domain, while they had no effect on several other classes of broadly neutralizing epitopes, including members of several lineages of V2-dependent quaternary epitopes and representatives of N332 glycan-dependent epitopes (PGT121) and quaternary, cleavage-dependent epitopes centered at the gp41-gp120 interface on intact HIV-1 Env trimers (PGT151). These results identify novel substitutions in gp120 that regulate the expression of alternative conformations of Env and differentially affect the exposure of different classes of epitopes, thereby influencing the neutralization phenotype of primary HIV-1 isolates.IMPORTANCE A better understanding of the mechanisms that determine the wide range of neutralization sensitivity of circulating primary HIV-1 isolates would provide important information about the natural structural and conformational diversity of HIV-1 Env and how this affects the neutralization phenotype. A useful way of studying this is to determine the molecular basis for the unusually high neutralization sensitivities of the limited number of available tier 1a viruses. This study localized the neutralization sensitivity of MW965.26, an extremely sensitive subtype C-derived primary isolate, to two rare substitutions in the C3 and C5 domains and demonstrated that the sequences at these positions differentially affect the presentation of epitopes recognized by different classes of standard and conformation-dependent broadly neutralizing antibodies. These results provide novel insight into how these regions regulate the neutralization phenotype and provide tools for controlling the Env conformation that could have applications both for structural studies and in vaccine design.


Subject(s)
Antibodies, Neutralizing/immunology , Genes, env/immunology , HIV Antibodies/immunology , HIV-1/immunology , env Gene Products, Human Immunodeficiency Virus/immunology , Amino Acid Sequence , Drug Substitution , Epitopes/genetics , Epitopes/immunology , Genes, env/genetics , HEK293 Cells , HIV Envelope Protein gp120/chemistry , HIV Envelope Protein gp120/genetics , HIV Envelope Protein gp120/immunology , HIV Envelope Protein gp160/immunology , HIV Envelope Protein gp41/immunology , HIV Infections/immunology , HIV Infections/virology , HIV-1/chemistry , HIV-1/genetics , HIV-1/isolation & purification , Humans , In Vitro Techniques , Mutation , Neutralization Tests , Phenotype , Protein Conformation , env Gene Products, Human Immunodeficiency Virus/chemistry , env Gene Products, Human Immunodeficiency Virus/genetics
4.
J Virol ; 90(20): 9471-82, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27512064

ABSTRACT

UNLABELLED: Soluble forms of trimeric HIV-1 envelope glycoprotein (Env) have long been sought as immunogens and as reagents for analysis of Env structure and function. Isolation of trimers that mimic native Env, derived from diverse viruses, however, represents a major challenge. Thus far, the most promising native-like (NL) structures have been obtained by engineering trimer-stabilizing mutations, termed SOSIP, into truncated Env sequences. However, the abundances of NL trimeric conformers vary among Envs, necessitating purification by monoclonal antibodies (MAbs) like PGT145, which target specific epitopes. To surmount this inherent limitation, we developed an approach that uses lectin affinity chromatography, ion-exchange chromatography, hydrophobic-interaction chromatography (HIC), and size exclusion chromatography (SEC) to isolate NL trimers from nonnative Env species. We validated this method with SOSIP trimers from HIV-1 clades A and B. Analyses by SEC, blue native PAGE, SDS-PAGE, and dynamic light scattering indicated that the resulting material was homogeneous (>95% pure), fully cleaved, and of the appropriate molecular weight and size for SOSIP trimers. Negative-stain electron microscopy further demonstrated that our preparations were composed of NL trimeric structures. By hydrogen/deuterium-exchange mass spectrometry, these HIC-pure trimers exhibited structural organization consistent with NL trimers and inconsistent with profiles seen in nonnative Envs. Screened for antigenicity, some Envs, like BS208.b1 and KNH1144 T162A, did not present the glycan/quaternary structure-dependent epitope for PGT145 binding, suggesting that these SOSIPs would be challenging to isolate by existing MAb affinity methods. By selecting based on biochemical rather than antigenic properties, our method offers an epitope-independent alternative to MAbs for isolation of NL Env trimers. IMPORTANCE: The production and purification of diverse soluble Env trimers that maintain native-like (NL) structure present technical challenges that must be overcome in order to advance vaccine development and provide reagents for HIV research. Low levels of NL trimer expression amid heterogeneous Env conformers, even with the addition of stabilizing mutations, have presented a major challenge. In addition, it has been difficult to separate the NL trimers from these heterogeneous mixtures. While MAbs with specificity for quaternary NL trimer epitopes have provided one approach to purifying the desirable species, such methods are dependent on the Env displaying the proper epitope. In addition, MAb affinity chromatography can be expensive, the necessary MAb may be in limited supply, and large-scale purification may not be feasible. Our method based on biochemical separation techniques offers an epitope-independent approach to purification of NL trimers with general application to diverse Envs.


Subject(s)
Antigens, Viral/immunology , Epitopes/chemistry , HIV-1/chemistry , env Gene Products, Human Immunodeficiency Virus/chemistry , AIDS Vaccines/immunology , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Cell Line , Chromatography, Affinity/methods , Chromatography, Ion Exchange/methods , Epitopes/immunology , Genes, env/immunology , HIV Antibodies/immunology , HIV-1/immunology , Humans , Protein Multimerization/immunology , env Gene Products, Human Immunodeficiency Virus/immunology
5.
Retrovirology ; 11: 49, 2014 Jun 24.
Article in English | MEDLINE | ID: mdl-24961156

ABSTRACT

BACKGROUND: Tat is a key HIV-1 virulence factor, which plays pivotal roles in virus gene expression, replication, transmission and disease progression. After release, extracellular Tat accumulates in tissues and exerts effects on both the virus and the immune system, promoting immune activation and virus spreading while disabling the host immune defense. In particular, Tat binds Env spikes on virus particles forming a virus entry complex, which favors infection of dendritic cells and efficient transmission to T cells via RGD-binding integrins. Tat also shields the CCR5-binding sites of Env rendering ineffective virus neutralization by anti-Env antibodies (Abs). This is reversed by the anti-Tat Abs present in natural infection or induced by vaccination. FINDINGS: Here we present the results of a cohort study, showing that the presence of anti-Tat Abs in asymptomatic and treatment-naïve HIV-infected subjects is associated with containment of CD4+ T-cell loss and viral load and with a delay of disease progression. In fact, no subjects with high anti-Tat Ab titers initiated antiretroviral therapy during the three years of follow-up. In contrast, no significant effects were seen for anti-Env and anti-Gag Abs. The increase of anti-Env Ab titers was associated with a reduced risk of starting therapy only in the presence of anti-Tat Abs, suggesting an effect of combined anti-Tat and anti-Env Abs on the Tat/Env virus entry complex and on virus neutralization. CONCLUSIONS: Anti-Tat immunity may help delay HIV disease progression, thus, targeting Tat may offer a novel therapeutic intervention to postpone antiretroviral treatment or to increase its efficacy.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , CD4-Positive T-Lymphocytes/immunology , HIV Infections/immunology , HIV-1/immunology , tat Gene Products, Human Immunodeficiency Virus/immunology , AIDS Vaccines/administration & dosage , AIDS Vaccines/immunology , Adult , Cohort Studies , Disease Progression , Female , Gene Products, env/immunology , Genes, env/immunology , HIV Infections/virology , Humans , Male , Viral Load
6.
AIDS ; 27(7): 1081-90, 2013 Apr 24.
Article in English | MEDLINE | ID: mdl-23324659

ABSTRACT

OBJECTIVES: To produce new fusion inhibitor peptides for HIV-1 and HIV-2 based on ancestral envelope sequences. METHODS: HIV-2/simian immunodeficiency virus (SIV) ancestral transmembrane protein sequences were reconstructed and ancestral peptides were derived from the helical region 2 (HR2). The activity of one ancestral peptide (named P3) was examined against a panel of HIV-1 and HIV-2 primary isolates in TZM-bl cells and peripheral blood mononuclear cells and compared to T-20. Peptide secondary structure was analyzed by circular dichroism. Resistant viruses were selected and resistance mutations were identified by sequencing the env gene. RESULTS: P3 has 34 residues and overlaps the N-terminal pocket-binding region and heptad repeat core of HR2. In contrast to T-20, P3 forms a typical α-helical structure in solution, binds strongly to the transmembrane protein, and potently inhibits both HIV-2 (mean IC50, 63.8 nmol/l) and HIV-1 (11 nmol/l) infection, including T-20-resistant isolates. The N43K mutation in the HR1 region of HIV-1 leads to 120-fold resistance to P3 indicating that the HR1 region in transmembrane glycoprotein is the target of P3. No HIV-2-resistant mutations could be selected by P3 suggesting that the genetic barrier to resistance is higher in HIV-2 than in HIV-1. HIV-1-infected patients presented significantly lower P3-specific antibody reactivity compared to T-20. CONCLUSION: P3 is an HIV-2/SIV ancestral peptide with low antigenicity, high stability, and potent activity against both HIV-1, including variants resistant to T-20, and HIV-2. Similar evolutionary biology strategies should be explored to enhance the production of antiviral peptide drugs, microbicides, and vaccines.


Subject(s)
Carrier Proteins/pharmacology , HIV Fusion Inhibitors/pharmacology , HIV Infections/immunology , HIV-1/immunology , HIV-2/immunology , Peptides/pharmacology , Simian Immunodeficiency Virus/immunology , Adult , Animals , Drug Design , Drug Resistance, Viral , Female , Genes, env/immunology , HIV Fusion Inhibitors/therapeutic use , HIV Infections/drug therapy , HIV Infections/genetics , HIV-1/isolation & purification , HIV-2/isolation & purification , Humans , Intercellular Signaling Peptides and Proteins , Male , Simian Immunodeficiency Virus/isolation & purification
7.
Nano Lett ; 12(4): 2003-12, 2012 Apr 11.
Article in English | MEDLINE | ID: mdl-22372996

ABSTRACT

With the intense international response to the AIDS pandemic, HIV vaccines have been extensively investigated but have failed due to issues of safety or efficacy in humans. Adjuvants for HIV/AIDS vaccines are under intense research but a rational design approach is still lacking. Nanomaterials represent an obvious opportunity in this field due to their unique physicochemical properties. Gold nanostructures are being actively studied as a promising and versatile platform for biomedical application. Herein, we report novel surface-engineered gold nanorods (NRs) used as promising DNA vaccine adjuvant for HIV treatment. We have exploited the effects of surface chemistry on the adjuvant activity of the gold nanorod by placing three kinds of molecules, that is, cetyltrimethylammonium bromide (CTAB), poly(diallydimethylammonium chloride) (PDDAC), and polyethyleneimine (PEI) on the surface of the nanorod. These PDDAC- or PEI-modified Au NRs can significantly promote cellular and humoral immunity as well as T cell proliferation through activating antigen-presenting cells if compared to naked HIV-1 Env plasmid DNA treatment in vivo. These findings have shed light on the rational design of low-toxic nanomaterials as a versatile platform for vaccine nanoadjuvants/delivery systems.


Subject(s)
Genes, env/immunology , Gold/chemistry , HIV-1/immunology , Metal Nanoparticles/chemistry , Vaccines, DNA/immunology , Cetrimonium , Cetrimonium Compounds/chemistry , HIV-1/genetics , Humans , Polyethyleneimine/chemistry , Polyethylenes/chemistry , Quaternary Ammonium Compounds/chemistry , Surface Properties , Vaccines, DNA/genetics
8.
PLoS One ; 6(5): e19644, 2011.
Article in English | MEDLINE | ID: mdl-21625608

ABSTRACT

Currently, MVA virus vectors carrying HIV-1 genes are being developed as HIV-1/AIDS prophylactic/therapeutic vaccines. Nevertheless, little is known about the impact of these vectors on human dendritic cells (DC) and their capacity to present HIV-1 antigens to human HIV-specific T cells. This study aimed to characterize the interaction of MVA and MVA expressing the HIV-1 genes Env-Gag-Pol-Nef of clade B (referred to as MVA-B) in human monocyte-derived dendritic cells (MDDC) and the subsequent processes of HIV-1 antigen presentation and activation of memory HIV-1-specific T lymphocytes. For these purposes, we performed ex vivo assays with MDDC and autologous lymphocytes from asymptomatic HIV-infected patients. Infection of MDDC with MVA-B or MVA, at the optimal dose of 0.3 PFU/MDDC, induced by itself a moderate degree of maturation of MDDC, involving secretion of cytokines and chemokines (IL1-ra, IL-7, TNF-α, IL-6, IL-12, IL-15, IL-8, MCP-1, MIP-1α, MIP-1ß, RANTES, IP-10, MIG, and IFN-α). MDDC infected with MVA or MVA-B and following a period of 48 h or 72 h of maturation were able to migrate toward CCL19 or CCL21 chemokine gradients. MVA-B infection induced apoptosis of the infected cells and the resulting apoptotic bodies were engulfed by the uninfected MDDC, which cross-presented HIV-1 antigens to autologous CD8(+) T lymphocytes. MVA-B-infected MDDC co-cultured with autologous T lymphocytes induced a highly functional HIV-specific CD8(+) T cell response including proliferation, secretion of IFN-γ, IL-2, TNF-α, MIP-1ß, MIP-1α, RANTES and IL-6, and strong cytotoxic activity against autologous HIV-1-infected CD4(+) T lymphocytes. These results evidence the adjuvant role of the vector itself (MVA) and support the clinical development of prophylactic and therapeutic anti-HIV vaccines based on MVA-B.


Subject(s)
AIDS Vaccines/immunology , CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Genetic Vectors/administration & dosage , HIV Infections/immunology , Poxviridae/genetics , Poxviridae/immunology , AIDS Vaccines/genetics , Apoptosis/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Chemokines/metabolism , Cytokines/metabolism , Dendritic Cells/metabolism , Flow Cytometry , Genes, env/immunology , HIV Antigens/genetics , HIV Antigens/immunology , HIV Infections/genetics , HIV Infections/metabolism , HIV-1/genetics , HIV-1/immunology , Humans , Monocytes/immunology , Monocytes/metabolism
9.
AIDS ; 25(2): 159-64, 2011 Jan 14.
Article in English | MEDLINE | ID: mdl-21124203

ABSTRACT

BACKGROUND: administration of recombinant human interleukin (IL)-7 leads to CD4 and CD8 T-cell expansions in HIV-infected individuals, demonstrating promising capacity for immune reconstitution. However, a proportion of patients treated with recombinant human IL-7 experience transient increases in plasma HIV-RNA ('blips'), possibly reflecting 'purging' of a quiescent reservoir that provides a barrier to viral eradication. OBJECTIVE: to identify the sources of HIV detected during transient viremic episodes following IL-7 administration, viral quasispecies were analyzed in a total of 281 primary sequences derived from seven patients who experienced the episodic blips following IL-7 therapy. METHOD: the C2-V3 regions of the HIV-1 env gene were sequenced from HIV-1 RNA in plasma and HIV DNA from peripheral blood mononuclear cells (PBMCs) obtained at baseline (day 0 of recombinant human IL-7 therapy), during the episode of viral blips (day 4), and at a time when levels of plasma HIV-RNA had returned to less than 50 copies/ml (day 28). RESULTS: the HIV sequences detected during transient viremia following IL-7 administration were closely related to those of the plasma viruses present before and after cytokine administration. All virus quasispecies detected during blips were also present in proviral sequences in PBMCs. CONCLUSION: the low level viremia induced by IL-7 likely reflects predominantly transient induction or release of virus from a preexisting pool rather than activation of silent quasispecies.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Genes, env/immunology , HIV Infections/immunology , Interleukin-7/therapeutic use , Lymphocyte Activation/immunology , Antiretroviral Therapy, Highly Active , Female , Genes, env/drug effects , Genes, env/genetics , HIV Infections/drug therapy , HIV Infections/genetics , HIV-1/physiology , Humans , Lymphocyte Activation/drug effects , Lymphocyte Activation/genetics , Male , Phylogeny , RNA, Viral , Viral Load , Viremia , Virus Latency/genetics
10.
J Immunol ; 185(6): 3348-58, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20702730

ABSTRACT

Indian rhesus macaques infected with the Rev-independent live-attenuated SIVmac239 strains control viremia to undetectable levels, have persistent but low cellular and humoral anti-SIV responses, and show no signs of immune deficiency. To analyze the immune mechanisms responsible for viral control, five macaques infected at day 1 after birth were subjected to CD8(+) cell depletion at 6.7 y postinfection. This resulted in viremia increases to 3.7-5.5 log(10) RNA copies, supporting a role of CD8-mediated responses in the control of viral replication. The rebounding viremia was rapidly controlled to levels below the threshold of detection, and occurred in the absence of SIV-specific CD8(+) T cells and significant CD8(+) T cell recovery in four of the five animals, suggesting that other mechanisms are involved in the immunological control of viremia. Monitoring immune responses at the time of viral control demonstrated a burst of circulating SIV-specific CD4(+) T cells characterized as CD45RA(-)CD28(+)CD95(+)CCR7(-) and also granzyme B(+), suggesting cytotoxic ability. Control of viremia was also concomitant with increases in humoral responses to Gag and Env, including a transient increase in neutralizing Abs against the neutralization-resistant SIVmac239 in four of five animals. These data demonstrate that a combination of cellular responses mediated by CD4(+) T cells and humoral responses was associated with the rapid control of the rebounding viremia in macaques infected by the Rev-independent live-attenuated SIV, even in the absence of measurable SIV-specific CD8(+) T cells in the blood, emphasizing the importance of different components of the immune response for full control of SIV infection.


Subject(s)
Antibodies, Viral/biosynthesis , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Genes, env/immunology , Lymphocyte Depletion , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Immunodeficiency Virus/immunology , Up-Regulation/immunology , Viremia/prevention & control , Animals , Antibodies, Neutralizing/biosynthesis , Antibodies, Neutralizing/physiology , Antibodies, Viral/physiology , CD4-Positive T-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/virology , Cytotoxicity, Immunologic , Epitopes, T-Lymphocyte/physiology , Macaca mulatta , SAIDS Vaccines/administration & dosage , SAIDS Vaccines/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/pathology , Simian Immunodeficiency Virus/genetics , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology , Viremia/immunology , Viremia/pathology , Virus Replication/immunology
11.
Article in Chinese | MEDLINE | ID: mdl-21604562

ABSTRACT

OBJECTIVE: To construct DNA and recombinant adenovirus vector vaccines containing an env gene from the prevalent subtype B strain in China and try to use them for therapeutic and prophylactic vaccines. METHODS: The candidate plasmid DNA vaccine pVR-gp160 and recombinant adenovirus vaccine rAdV-gp160 were constructed separately. BALB/c mice were immunized with these two vaccines in different administration schemes. HIV-1 Gp120-specific cellular responses and antibody levels were detected by ELISPOT and ELISA respectively. RESULTS: DNA vaccine alone and combined vaccines in a DNA prime/rAdV-gp160 boost vaccination regimen induced high level of Gp120-specific cellular responses. While low level of Gp120-specific antibodies were elicited in all groups. CONCLUSION: DNA and rAdV vaccines could efficiently express Gp160 protein and activate specific cellular responses.


Subject(s)
AIDS Vaccines/immunology , Adenoviridae/immunology , Genes, env/immunology , Genetic Vectors/immunology , HIV-1/immunology , Plasmids/immunology , Vaccines, DNA/immunology , AIDS Vaccines/genetics , Adenoviridae/genetics , Animals , China , Genetic Vectors/genetics , HIV Antibodies/genetics , HIV Antibodies/immunology , HIV Envelope Protein gp120/genetics , HIV Envelope Protein gp120/immunology , HIV Envelope Protein gp160/genetics , HIV Envelope Protein gp160/immunology , HIV-1/genetics , Mice , Mice, Inbred BALB C , Plasmids/genetics , Vaccines, DNA/genetics , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
12.
AIDS Res Hum Retroviruses ; 25(4): 425-31, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19320568

ABSTRACT

Dendritic cells (DCs) are fundamental for the initiation of immune responses and are important players in AIDS immunopathogenesis. Impairment of DC function may result from bystander effects of HIV-1 envelope proteins independently of direct HIV-1 infection. HIV-2 envelope proteins are thought to interact with a broader range of receptors than those of HIV-1, and have been shown to have T cell immunosuppressive properties mediated by monocytes. The effects of HIV-2 envelope on DC differentiation and maturation were investigated. The modulatory properties of the HIV-2 envelope on DC generated from monocytes were assessed using both recombinant proteins (HIV-2(ROD) and HIV-2(ALI)) and whole chemically inactivated virus (aldrithiol-2-treated HIV-2(ROD)). DC phenotype was assessed by flow cytometry and DC function by their ability to stimulate allogeneic T cells and to produce cytokines. We demonstrate that HIV-2 Env had no effects upon DC differentiation and maturation despite its broad receptor usage and ability to modulate monocyte function. It is plausible to speculate that a reduced ability of the HIV-2 Env to impair myeloid DC function could represent a contributory factor to the relatively benign course of HIV-2 disease.


Subject(s)
Cell Differentiation , Dendritic Cells/cytology , Dendritic Cells/immunology , HIV-2/immunology , env Gene Products, Human Immunodeficiency Virus/immunology , Cell Differentiation/immunology , Cells, Cultured , Genes, env/immunology , Humans , Monocytes/cytology , Monocytes/immunology , Recombinant Proteins/genetics , Recombinant Proteins/immunology , env Gene Products, Human Immunodeficiency Virus/genetics
13.
Vaccine ; 27(2): 260-9, 2009 Jan 07.
Article in English | MEDLINE | ID: mdl-18984025

ABSTRACT

To determine whether systemic immunization with plasmid DNA and virus vector against visna/maedi virus (VMV) would induce protective immune responses, sheep were immunized with VMV gag and/or env sequences using particle-mediated epidermal bombardment and injection of recombinant modified vaccinia Ankara. The results showed that immunization induced both humoral and cell-mediated responses prior to and after virus challenge. The vaccination protocol did not prevent infection, but immunization with the gag gene or a combination of gag and env genes resulted in significantly reduced provirus loads in blood and mediastinal lymph node, respectively. Provirus loads in lung and draining lymph node were unaffected, but p25 expression was undetectable in lungs of animals immunized with a combination of gag and env genes. Analysis of target tissues for lesions at post-mortem showed that immunization with the env gene caused a significant increase in lesion score, while the gag gene or a combination of gag and env genes had no effect. Inclusion of the ovine interferon-gamma gene in the initial priming mixture had minimal effect on immune responses, provirus load, or lesion development, although it resulted in a decreased p25 expression in the lung. The results thus show that systemic immunization with gag or a combination of gag and env genes reduces provirus load in blood and lymphoid tissue, respectively whereas env immunization has no effect on provirus load but increased lesion development.


Subject(s)
Biolistics , Genes, env/genetics , Genes, gag/genetics , Pneumonia, Progressive Interstitial, of Sheep/prevention & control , Vaccines, DNA , Vaccinia virus/genetics , Animals , Antibodies, Viral/blood , Epidermis/virology , Female , Genes, env/immunology , Genes, gag/immunology , Immunization , Male , Pneumonia, Progressive Interstitial, of Sheep/virology , Proviruses/isolation & purification , Sheep , T-Lymphocytes/immunology , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics , Vaccines, DNA/immunology , Vaccinia virus/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/genetics , Viral Vaccines/immunology , Virion/genetics , Virion/immunology , Visna-maedi virus
14.
Vaccine ; 26(40): 5128-34, 2008 Sep 19.
Article in English | MEDLINE | ID: mdl-18471943

ABSTRACT

Chikungunya virus (CHIKV) is an emerging arbovirus and is an important human pathogen. Infection of humans by CHIKV can cause a syndrome characterized by fever, headache, rash, nausea, vomiting, myalgia, arthralgia and occasionally neurological manifestations such as acute limb weakness. It is also associated with a fatal haemorrhagic condition. CHIKV is geographically distributed from Africa through Southeast Asia and South America, and its transmission to humans is mainly through the Aedes aegypti species mosquitoes. The frequency of recent epidemics in the Indian Ocean and La Reunion islands suggests that a new vector perhaps is carrying the virus, as A. aegypti are not found there. In fact, a relative the Asian tiger mosquito, Aedes albopictus, may be the culprit which has raised concerns in the world health community regarding the potential for a CHIK virus pandemic. Accordingly steps should be taken to develop methods for the control of CHIKV. Unfortunately, currently there is no specific treatment for Chikungunya virus and there is no vaccine currently available. Here we present data of a novel consensus-based approach to vaccine design for CHIKV, employing a DNA vaccine strategy. The vaccine cassette was designed based on CHIKV capsid- and envelope-specific consensus sequences with several modifications, including codon optimization, RNA optimization, the addition of a Kozak sequence, and a substituted immunoglobulin E leader sequence. The expression of capsid, envelope E1 and E1 was evaluated using T7-coupled transcription/translation and immunoblot analysis. A recently developed, adaptive constant-current electroporation technique was used to immunize C57BL/6 mice with an intramuscular injection of plasmid coding for the CHIK-Capsid, E1 and E2. Analysis of cellular immune responses, including epitope mapping, demonstrates that electroporation of these constructs induces both potent and broad cellular immunity. In addition, antibody ELISAs demonstrate that these synthetic immunogens are capable of inducing high titer antibodies capable of recognizing native antigen. Taken together, these data support further study of the use of consensus CHIK antigens in a potential vaccine cocktail.


Subject(s)
Alphavirus Infections/immunology , Capsid Proteins/immunology , Chikungunya virus/immunology , Genes, env/immunology , Vaccines, DNA/immunology , Viral Vaccines/immunology , Alphavirus Infections/prevention & control , Alphavirus Infections/virology , Animals , Antibodies, Viral/blood , Capsid/immunology , Capsid Proteins/genetics , Chikungunya virus/genetics , Cricetinae , Electroporation , Female , Mice , Mice, Inbred C57BL , T-Lymphocytes, Cytotoxic/immunology , Vaccination , Vaccines, DNA/administration & dosage , Viral Vaccines/administration & dosage , Viral Vaccines/genetics
15.
Curr HIV Res ; 5(6): 572-7, 2007 Nov.
Article in English | MEDLINE | ID: mdl-18045113

ABSTRACT

Centralized HIV-1 genes (consensus, most recent common ancestor and center of the tree) have recently been explored for induction of broadly reactive immune responses to overcome the extraordinary genetic diversity among HIV-1 strains. Although all of these strategies are based on artificial sequences predicted by computer programs, they retain biological function, and use the CCR5 co-receptor for entry into target cells as transmitted HIV-1 Envs. Results from laboratory animals indicate that centralized immunogens are superior to many wild-type immunogens for inducing cross-subtype T and B cell immune responses. Structural modifications have improved the ability of consensus Envs to elicit antibody responses that neutralize a spectrum of HIV-1 Env pseudoviruses. However, the more difficult to neutralize tier 2 Env pseudoviruses are generally not neutralized well by anti-consensus Env antibodies, indicating the need for further modifications, new formulations, or additional strategies to generate antibodies that neutralize a full spectrum of transmitted HIV-1 strains.


Subject(s)
AIDS Vaccines/immunology , Consensus Sequence , env Gene Products, Human Immunodeficiency Virus/immunology , Consensus Sequence/genetics , Consensus Sequence/immunology , Genes, env/genetics , Genes, env/immunology , HIV-1/classification , HIV-1/genetics , HIV-1/immunology , Humans , Receptors, CCR5 , Vaccines, Synthetic
16.
Curr HIV Res ; 5(2): 199-209, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17346134

ABSTRACT

Immunization with more than one immunogen (co-immunization) is an efficient regimen to induce immunity to multiple antigens. However, immune interference has been reported using multi-plasmid DNA immunizations. HIV-1 envelope (Env) and Gag gene products are the predominant immunogens used in current AIDS vaccines, although, few studies have evaluated possible immune interference when these two antigens are co-administered. Therefore, in this study, immune interference during co-inoculation was examined using DNA vaccines expressing lentiviral Envs and Gag from gene sequences optimized for efficient expression in mammalian cells (codon-optimized). BALB/c mice vaccinated in separate hind legs with each plasmid individually elicited high titer immune responses, however, when HIV-1 Env(gp120) and HIV-1 Gag(p55) DNA plasmids were co-inoculated, there was a reduction in the immune responses elicited to HIV-1 Gag(p55). To determine if the anti-HIV-1 Gag(p55) immune interference was specific to HIV-1 Env(gp120), mice were co-immunized with plasmids expressing the surface envelope protein from two additional lentiviruses, Env(gp130)-SIV or Env(gp90)-EIAV, or a soluble form of hemagglutinin (sHA) from influenza virus and HIV-1 Gag(p55)- or SIV Gag(p55)-DNA. Interestingly, there was no reduction in anti-HIV-1 Gag(p55) immune responses using other lentiviral envelopes or the influenza sHA. Also, none of the lentiviral envelopes reduced anti-SIV Gag(p55) immune responses during co-immunization. Therefore, anti-HIV-1 Gag immune interference appears specific to co-immunizations with HIV-1 Env(gp120) and may involve a yet undefined immunological mechanism(s).


Subject(s)
Genes, env/immunology , Genes, gag/immunology , HIV-1/immunology , Immunization/methods , Vaccines, DNA/antagonists & inhibitors , Vaccines, DNA/immunology , Animals , Dose-Response Relationship, Immunologic , Drug Antagonism , Female , HIV Envelope Protein gp120/genetics , HIV-1/genetics , Mice , Mice, Inbred BALB C , Models, Animal , Plasmids
17.
Mol Ther ; 15(2): 411-21, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17235321

ABSTRACT

An important goal for human immunodeficiency virus (HIV) vaccines is to develop immunogens that induce broader and more potent cellular immune responses. In this study of DNA vaccine potency, we constructed a novel subtype B env gene (EY2E1-B) with the goal of increasing vaccine antigen immune potency. The vaccine cassette was designed based on subtype B-specific consensus sequence with several modifications, including codon optimization, RNA optimization, the addition of a Kozak sequence, and a substituted immunoglobulin E leader sequence. The V1 and V2 loops were shortened and the cytoplasmic tail was truncated to prevent envelope recycling. Three different strains of mice (BALB/c, C57BL/6, and HLA-A2 transgenic mice) were immunized three times with pEY2E1-B or the primary DNA immunogen pEK2P-B alone. The analysis of specific antibody responses suggested that EY2E1-B could induce a moderate subtype B-specific antibody response. Moreover, this construct was up to four times more potent at driving cellular immune responses. Epitope mapping results indicated that there is an increase in the breadth and magnitude of cross-reactive cellular responses induced by the EY2E1-B immunogen. These properties suggest that such a synthetic immunogen deserves further examination for its potential to serve as a component antigen in an HIV vaccine cocktail.


Subject(s)
AIDS Vaccines/immunology , Genes, env/immunology , HIV-1/immunology , Immunity, Cellular/immunology , AIDS Vaccines/genetics , Amino Acid Sequence , Animals , Female , Fluorescent Antibody Technique, Indirect , Genes, env/genetics , HIV Antigens/genetics , HIV Antigens/immunology , HIV-1/genetics , HLA-A2 Antigen/genetics , HLA-A2 Antigen/immunology , Immunization/methods , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Sequence Alignment
18.
AIDS Res Hum Retroviruses ; 22(1): 109-13, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16438653

ABSTRACT

The aim of this study was to investigate the genetic diversity of HIV-1 strains circulating in Slovenia. Proviral DNA isolated from peripheral blood mononuclear cells (PBMCs) of 20 randomly selected HIV-1-infected individuals was classified into subtypes by sequence-based phylogenetic analysis of the env (C2V3) and gag (p24) regions of the viral genome. The phylogenetic tree based on env C2V3 sequences showed that 15 of the 20 samples were subtype B, two A1, one F1, one CRF01_AE, and one CRF02_AG. The phylogenetic analysis of the gag gene yielded identical results expect for one sample that had a discordant subtype; it was identified as subtype A1 in the env and AE in the gag region. Our study confirmed that although subtype B predominates, other subtypes and circulating recombinant forms (CRFs) are also present in Slovenia. The high intrasubtype genetic diversity of subtype B sequences suggests a multiple introduction of subtype B strains into Slovenia.


Subject(s)
Gene Products, gag/genetics , Genes, env/genetics , Genetic Variation , HIV Infections/epidemiology , HIV-1/genetics , Amino Acid Sequence , Gene Products, gag/immunology , Genes, env/immunology , HIV Infections/immunology , HIV-1/classification , Molecular Sequence Data , Phylogeny , Slovenia/epidemiology
19.
J Gen Virol ; 87(Pt 2): 399-410, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16432028

ABSTRACT

In this study, the design and preclinical development of a multigene human immunodeficiency virus type 1 (HIV-1) subtype C DNA vaccine are described, developed as part of the South African AIDS Vaccine Initiative (SAAVI). Genetic variation remains a major obstacle in the development of an HIV-1 vaccine and recent strategies have focused on constructing vaccines based on the subtypes dominant in the developing world, where the epidemic is most severe. The vaccine, SAAVI DNA-C, contains an equimolar mixture of two plasmids, pTHr.grttnC and pTHr.gp150CT, which express a polyprotein derived from Gag, reverse transcriptase (RT), Tat and Nef, and a truncated Env, respectively. Genes included in the vaccine were obtained from individuals within 3 months of infection and selection was based on closeness to a South African subtype C consensus sequence. All genes were codon-optimized for increased expression in humans. The genes have been modified for safety, stability and immunogenicity. Tat was inactivated through shuffling of gene fragments, whilst maintaining all potential epitopes; the active site of RT was mutated; 124 aa were removed from the cytoplasmic tail of gp160; and Nef and Gag myristylation sites were inactivated. Following vaccination of BALB/c mice, high levels of cytotoxic T lymphocytes were induced against multiple epitopes and the vaccine stimulated strong CD8+ gamma interferon responses. In addition, high titres of antibodies to gp120 were induced in guinea pigs. This vaccine is the first component of a prime-boost regimen that is scheduled for clinical trials in humans in the USA and South Africa.


Subject(s)
AIDS Vaccines/immunology , HIV-1/immunology , AIDS Vaccines/administration & dosage , Animals , Drug Design , Drug Evaluation, Preclinical , Genes, env/immunology , Genes, gag/immunology , Genes, tat/immunology , HIV-1/drug effects , Humans , Mice , Vaccines, DNA/administration & dosage , Vaccines, DNA/immunology
20.
Asian Pac J Allergy Immunol ; 23(2-3): 143-52, 2005.
Article in English | MEDLINE | ID: mdl-16252845

ABSTRACT

Two HIV-1 subtypes have accounted for virtually all infections in Thailand: subtype B', found mainly in injection drug users (IDUs), and CRF01_AE (initially subtype E), found in over 90% of sexually infected persons and increasingly in IDUs in recent years. During 1997-1998, 227 blood samples were collected from HIV-1 infected individuals consisting of 92 mothers, 35 children and 100 IDUs. The blood samples were subtyped by heteroduplex mobility assay (HMA) and peptide enzyme-linked immunosorbent assay (PEIA). Using gag and env HMA, CRF01_AE and subtype B' accounted for 96-97% and 3-4% of both the mothers and the children, respectively. In the IDU group, 10% of the plasma samples could only be performed by gag HMA and gave the result as CRF01_AE. CRF01_AE and subtype B' using PEIA accounted for 67% and 33% of the IDUs. There was 100% concordance of the results between gag HMA and env HMA. Ninety-five percentages of concordant results were observed between HMA and PEIA. Of the 6/134 (5%) subjects with discordant results, nucleotide sequencing, used as a gold standard, confirmed the HMA result. In this study, HIV-1 was successfully genotyped by HMA and PEIA. However, a comparison of the subtyping results between HMA and PEIA revealed that HMA was slightly more accurate than PEIA.


Subject(s)
Electrophoretic Mobility Shift Assay , Enzyme-Linked Immunosorbent Assay , Genes, env/immunology , Genes, gag/immunology , HIV-1/classification , Heteroduplex Analysis , Immunophenotyping , DNA, Viral/genetics , DNA, Viral/immunology , DNA, Viral/isolation & purification , Enzyme-Linked Immunosorbent Assay/methods , Female , Genes, env/genetics , Genes, gag/genetics , HIV Infections/genetics , HIV Infections/immunology , HIV-1/genetics , HIV-1/immunology , HIV-1/isolation & purification , Heteroduplex Analysis/methods , Humans , Infant , Male , Peptides/immunology , Polymerase Chain Reaction , Recombination, Genetic , Sequence Analysis, DNA , Thailand/epidemiology
SELECTION OF CITATIONS
SEARCH DETAIL
...