Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
J Virol ; 90(4): 2135-41, 2016 02 15.
Article in English | MEDLINE | ID: mdl-26656695

ABSTRACT

Protein kinase R (PKR) and RNase L are host cell components that function to contain viral spread after infections. In this study, we analyzed the role of both proteins in the abortive infection of human HeLa cells with the poxvirus strain NYVAC, for which an inhibition of viral A27L and B5R gene expression is described. Specifically, the translation of these viral genes is independent of PKR activation, but their expression is dependent on the RNase L activity.


Subject(s)
Endoribonucleases/metabolism , Epithelial Cells/immunology , Host-Pathogen Interactions , Membrane Glycoproteins/antagonists & inhibitors , Vaccinia virus/immunology , Viral Envelope Proteins/antagonists & inhibitors , Viral Proteins/antagonists & inhibitors , eIF-2 Kinase/metabolism , HeLa Cells , Humans , Protein Biosynthesis
2.
Proc Natl Acad Sci U S A ; 112(11): E1333-42, 2015 Mar 17.
Article in English | MEDLINE | ID: mdl-25739961

ABSTRACT

Neutrophils are antigen-transporting cells that generate vaccinia virus (VACV)-specific T-cell responses, yet how VACV modulates neutrophil recruitment and its significance in the immune response are unknown. We generated an attenuated VACV strain that expresses HIV-1 clade C antigens but lacks three specific viral genes (A52R, K7R, and B15R). We found that these genes act together to inhibit the NFκB signaling pathway. Triple ablation in modified virus restored NFκB function in macrophages. After virus infection of mice, NFκB pathway activation led to expression of several cytokines/chemokines that increased the migration of neutrophil populations (Nα and Nß) to the infection site. Nß cells displayed features of antigen-presenting cells and activated virus-specific CD8 T cells. Enhanced neutrophil trafficking to the infection site correlated with an increased T-cell response to HIV vector-delivered antigens. These results identify a mechanism for poxvirus-induced immune response and alternatives for vaccine vector design.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , HIV-1/immunology , Immune System Diseases , Leukocyte Disorders , NF-kappa B/metabolism , Animals , Antigen-Presenting Cells/immunology , Cell Line , Gene Deletion , Genes, Viral , HIV Antigens/immunology , Humans , Lymphocyte Activation/immunology , Mice, Inbred BALB C , Mice, Inbred C57BL , Models, Biological , Neutrophil Infiltration , Species Specificity , Vaccinia/immunology , Vaccinia/virology , Vaccinia virus/genetics
3.
PLoS One ; 7(10): e48524, 2012.
Article in English | MEDLINE | ID: mdl-23119046

ABSTRACT

Vaccinia virus (VACV) encodes an anti-apoptotic Bcl-2-like protein F1 that acts as an inhibitor of caspase-9 and of the Bak/Bax checkpoint but the role of this gene in immune responses is not known. Because dendritic cells that have phagocytosed apoptotic infected cells cross-present viral antigens to cytotoxic T cells inducing an antigen-specific immunity, we hypothesized that deletion of the viral anti-apoptotic F1L gene might have a profound effect on the capacity of poxvirus vectors to activate specific immune responses to virus-expressed recombinant antigens. This has been tested in a mouse model with an F1L deletion mutant of the HIV/AIDS vaccine candidate MVA-C that expresses Env and Gag-Pol-Nef antigens (MVA-C-ΔF1L). The viral gene F1L is not required for virus replication in cultured cells and its deletion in MVA-C induces extensive apoptosis and expression of immunomodulatory genes in infected cells. Analysis of the immune responses induced in BALB/c mice after DNA prime/MVA boost revealed that, in comparison with parental MVA-C, the mutant MVA-C-ΔF1L improves the magnitude of the HIV-1-specific CD8 T cell adaptive immune responses and impacts on the CD8 T cell memory phase by enhancing the magnitude of the response, reducing the contraction phase and changing the memory differentiation pattern. These findings reveal the immunomodulatory role of F1L and that the loss of this gene is a valid strategy for the optimization of MVA as vaccine vector.


Subject(s)
AIDS Vaccines/immunology , HIV Antigens/immunology , HIV-1/immunology , Vaccinia virus/genetics , Vaccinia virus/immunology , Viral Proteins/genetics , Viral Proteins/immunology , Amino Acid Sequence , Animals , Apoptosis/genetics , Apoptosis/immunology , Base Sequence , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line , Chick Embryo , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Epitopes, T-Lymphocyte/immunology , Gene Deletion , Gene Expression , Gene Order , Genetic Vectors/genetics , Genetic Vectors/immunology , HIV Antigens/genetics , HIV Envelope Protein gp120/chemistry , HIV Envelope Protein gp120/genetics , HIV Envelope Protein gp120/immunology , HIV-1/genetics , Human Immunodeficiency Virus Proteins/chemistry , Human Immunodeficiency Virus Proteins/genetics , Human Immunodeficiency Virus Proteins/immunology , Humans , Immunologic Memory/immunology , Interferon Type I/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , Molecular Sequence Data , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
4.
J Virol ; 86(9): 5026-38, 2012 May.
Article in English | MEDLINE | ID: mdl-22419805

ABSTRACT

Poxviruses encode multiple inhibitors of the interferon (IFN) system, acting at different levels and blocking the induction of host defense mechanisms. Two viral gene products, B19 and B8, have been shown to act as decoy receptors of type I and type II IFNs, blocking the binding of IFN to its receptor. Since IFN plays a major role in innate immune responses, in this investigation we asked to what extent the viral inhibitors of the IFN system impact the capacity of poxvirus vectors to activate immune responses. This was tested in a mouse model with single and double deletion mutants of the vaccine candidate NYVAC-C, which expresses the HIV-1 Env, Gag, Pol, and Nef antigens. When deleted individually or in double, the type I (B19) and type II (B8) IFN binding proteins were not required for virus replication in cultured cells. Studies of immune responses in mice after DNA prime/NYVAC boost revealed that deletion of B8R and/or B19R genes improved the magnitude and quality of HIV-1-specific CD8(+) T cell adaptive immune responses and impacted their memory phase, changing the contraction, the memory differentiation, the effect magnitude, and the functionality profile. For B cell responses, deletion of the viral gene B8R and/or B19R had no effect on antibody levels to HIV-1 Env. These findings revealed that single or double deletion of viral factors (B8 and B19) targeting the IFN pathway is a useful approach in the design of improved poxvirus-based vaccines.


Subject(s)
AIDS Vaccines/immunology , Adaptive Immunity , Immunologic Memory , Interferon Type I/metabolism , Interferon-gamma/metabolism , AIDS Vaccines/genetics , Animals , CD8-Positive T-Lymphocytes/immunology , Chick Embryo , Gene Deletion , Genetic Vectors/immunology , HIV Envelope Protein gp120/genetics , HIV Envelope Protein gp120/immunology , HIV Infections/immunology , HIV Infections/prevention & control , HIV-1/genetics , HIV-1/immunology , Haplorhini , Mice , Mice, Inbred BALB C , Signal Transduction/immunology , T-Lymphocytes/immunology , Vaccinia virus/genetics , Vaccinia virus/immunology
5.
PLoS One ; 6(8): e24244, 2011.
Article in English | MEDLINE | ID: mdl-21909386

ABSTRACT

The vaccinia virus (VACV) C6 protein has sequence similarities with the poxvirus family Pox_A46, involved in regulation of host immune responses, but its role is unknown. Here, we have characterized the C6 protein and its effects in virus replication, innate immune sensing and immunogenicity in vivo. C6 is a 18.2 kDa protein, which is expressed early during virus infection and localizes to the cytoplasm of infected cells. Deletion of the C6L gene from the poxvirus vector MVA-B expressing HIV-1 Env, Gag, Pol and Nef antigens from clade B (MVA-B ΔC6L) had no effect on virus growth kinetics; therefore C6 protein is not essential for virus replication. The innate immune signals elicited by MVA-B ΔC6L in human macrophages and monocyte-derived dendritic cells (moDCs) are characterized by the up-regulation of the expression of IFN-ß and IFN-α/ß-inducible genes. In a DNA prime/MVA boost immunization protocol in mice, flow cytometry analysis revealed that MVA-B ΔC6L enhanced the magnitude and polyfunctionality of the HIV-1-specific CD4+ and CD8+ T-cell memory immune responses, with most of the HIV-1 responses mediated by the CD8+ T-cell compartment with an effector phenotype. Significantly, while MVA-B induced preferentially Env- and Gag-specific CD8+ T-cell responses, MVA-B ΔC6L induced more Gag-Pol-Nef-specific CD8+ T-cell responses. Furthermore, MVA-B ΔC6L enhanced the levels of antibodies against Env in comparison with MVA-B. These findings revealed that C6 can be considered as an immunomodulator and that deleting C6L gene in MVA-B confers an immunological benefit by enhancing IFN-ß-dependent responses and increasing the magnitude and quality of the T-cell memory immune responses to HIV-1 antigens. Our observations are relevant for the improvement of MVA vectors as HIV-1 vaccines.


Subject(s)
AIDS Vaccines/immunology , Genes, Viral/genetics , HIV-1/immunology , Immunologic Memory/immunology , T-Lymphocytes/immunology , Vaccinia virus/genetics , Animals , Antibodies, Viral/immunology , Chickens , Dendritic Cells/immunology , Dendritic Cells/virology , HIV Envelope Protein gp120/immunology , Humans , Immunity, Humoral/immunology , Immunization , Interferon-beta/metabolism , Macrophages/immunology , Macrophages/virology , Mice , Mice, Inbred C57BL , Mutation/genetics , Protein Transport , Species Specificity
6.
J Virol ; 85(21): 11468-78, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21865377

ABSTRACT

Attenuated poxvirus vectors expressing human immunodeficiency virus type 1 (HIV-1) antigens are considered promising HIV/AIDS vaccine candidates. Here, we describe the nature of T cell immune responses induced in healthy volunteers participating in a phase I clinical trial in Spain after intramuscular administration of three doses of the recombinant MVA-B-expressing monomeric gp120 and the fused Gag-Pol-Nef (GPN) polyprotein of clade B. The majority (92.3%) of the volunteers immunized had a positive specific T cell response at any time postvaccination as detected by gamma interferon (IFN-γ) intracellular cytokine staining (ICS) assay. The CD4(+) T cell responses were predominantly Env directed, whereas the CD8(+) T cell responses were similarly distributed against Env, Gag, and GPN. The proportion of responders after two doses of MVA-B was similar to that obtained after the third dose of MVA-B vaccination, and the responses were sustained (84.6% at week 48). Vaccine-induced CD8(+) T cells to HIV-1 antigens after 1 year were polyfunctional and distributed mainly within the effector memory (TEM) and terminally differentiated effector memory (TEMRA) T cell populations. Antivector T cell responses were mostly induced by CD8(+) T cells, highly polyfunctional, and of TEMRA phenotype. These findings demonstrate that the poxvirus MVA-B vaccine candidate given alone is highly immunogenic, inducing broad, polyfunctional, and long-lasting CD4 and CD8 T cell responses to HIV-1 antigens, with preference for TEM. Thus, on the basis of the immune profile of MVA-B in humans, this immunogen can be considered a promising HIV/AIDS vaccine candidate.


Subject(s)
AIDS Vaccines/administration & dosage , AIDS Vaccines/immunology , Acquired Immunodeficiency Syndrome/prevention & control , HIV Antigens/immunology , Immunologic Memory , T-Lymphocytes/immunology , AIDS Vaccines/genetics , Drug Carriers , Genetic Vectors , HIV Antigens/genetics , HIV-1/immunology , Humans , Immunization, Secondary/methods , Injections, Intramuscular , Interferon-gamma/biosynthesis , Spain , Time Factors , Vaccination/methods , Vaccinia virus/genetics , Vaccinia virus/immunology
7.
PLoS One ; 6(2): e16711, 2011 Feb 02.
Article in English | MEDLINE | ID: mdl-21311764

ABSTRACT

Most viruses express during infection products that prevent or neutralize the effect of the host dsRNA activated protein kinase (PKR). Translation of Sindbis virus (SINV) mRNA escapes to PKR activation and eIF2 phosphorylation in infected cells by a mechanism that requires a stem loop structure in viral 26S mRNA termed DLP to initiate translation in the absence of functional eIF2. Unlike the rest of viruses tested, we found that Alphavirus infection allowed a strong PKR activation and eIF2α phosphorylation in vitro and in infected animals so that the presence of DLP structure in mRNA was critical for translation and replication of SINV. Interestingly, infection of MEFs with some viruses that express PKR inhibitors prevented eIF2α phosphorylation after superinfection with SINV, suggesting that viral anti-PKR mechanisms could be exchangeable. Thus, translation of SINV mutant lacking the DLP structure (ΔDLP) in 26S mRNA was partially rescued in cells expressing vaccinia virus (VV) E3 protein, a known inhibitor of PKR. This case of heterotypic complementation among evolutionary distant viruses confirmed experimentally a remarkable case of convergent evolution in viral anti-PKR mechanisms. Our data reinforce the critical role of PKR in regulating virus-host interaction and reveal the versatility of viruses to find different solutions to solve the same conflict.


Subject(s)
Evolution, Molecular , Genetic Variation , Host-Pathogen Interactions/genetics , Viruses/genetics , eIF-2 Kinase/antagonists & inhibitors , Animals , Base Sequence , Cells, Cultured , Genetic Variation/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Molecular Sequence Data , Nucleic Acid Conformation , Signal Transduction/genetics , Sindbis Virus/genetics , Sindbis Virus/metabolism , Virus Replication/genetics , Viruses/metabolism , Viruses/pathogenicity , eIF-2 Kinase/genetics
8.
PLoS One ; 5(8): e12395, 2010 Aug 24.
Article in English | MEDLINE | ID: mdl-20811493

ABSTRACT

BACKGROUND: The immune parameters of HIV/AIDS vaccine candidates that might be relevant in protection against HIV-1 infection are still undefined. The highly attenuated poxvirus strain MVA is one of the most promising vectors to be use as HIV-1 vaccine. We have previously described a recombinant MVA expressing HIV-1 Env, Gag, Pol and Nef antigens from clade B (referred as MVA-B), that induced HIV-1-specific immune responses in different animal models and gene signatures in human dendritic cells (DCs) with immunoregulatory function. METHODOLOGY/PRINCIPAL FINDINGS: In an effort to characterize in more detail the immunogenic profile of MVA-B and to improve its immunogenicity we have generated a new vector lacking two genes (A41L and B16R), known to counteract host immune responses by blocking the action of CC-chemokines and of interleukin 1beta, respectively (referred as MVA-B DeltaA41L/DeltaB16R). A DNA prime/MVA boost immunization protocol was used to compare the adaptive and memory HIV-1 specific immune responses induced in mice by the parental MVA-B and by the double deletion mutant MVA-B DeltaA41L/DeltaB16R. Flow cytometry analysis revealed that both vectors triggered HIV-1-specific CD4(+) and CD8(+) T cells, with the CD8(+) T-cell compartment responsible for >91.9% of the total HIV-1 responses in both immunization groups. However, MVA-B DeltaA41L/DeltaB16R enhanced the magnitude and polyfunctionality of the HIV-1-specific CD4(+) and CD8(+) T-cell immune responses. HIV-1-specific CD4(+) T-cell responses were polyfunctional and preferentially Env-specific in both immunization groups. Significantly, while MVA-B induced preferentially Env-specific CD8(+) T-cell responses, MVA-B DeltaA41L/DeltaB16R induced more GPN-specific CD8(+) T-cell responses, with an enhanced polyfunctional pattern. Both vectors were capable of producing similar levels of antibodies against Env. CONCLUSIONS/SIGNIFICANCE: These findings revealed that MVA-B and MVA-B DeltaA41L/DeltaB16R induced in mice robust, polyfunctional and durable T-cell responses to HIV-1 antigens, but the double deletion mutant showed enhanced magnitude and quality of HIV-1 adaptive and memory responses. Our observations are relevant in the immune evaluation of MVA-B and on improvements of MVA vectors as HIV-1 vaccines.


Subject(s)
AIDS Vaccines/genetics , AIDS Vaccines/immunology , Gene Deletion , Genes, Viral/genetics , HIV Antigens/genetics , Poxviridae/genetics , Poxviridae/immunology , Animals , Antibodies, Viral/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Gene Expression , Genetic Vectors/genetics , Genetic Vectors/immunology , HIV Antigens/immunology , HIV Envelope Protein gp120/immunology , Immunologic Memory/immunology , Mice , Species Specificity
9.
PLoS One ; 5(6): e11406, 2010 Jun 30.
Article in English | MEDLINE | ID: mdl-20613977

ABSTRACT

BACKGROUND: The highly attenuated vaccinia virus strain NYVAC expressing HIV-1 components has been evaluated as a vaccine candidate in preclinical and clinical trials with encouraging results. We have previously described that the presence of C7L in the NYVAC genome prevents the induction of apoptosis and renders the vector capable of replication in human and murine cell lines while maintaining an attenuated phenotype in mice. METHODOLOGY/PRINCIPAL FINDINGS: In an effort to improve the immunogenicity of NYVAC, we have developed a novel poxvirus vector by inserting the VACV host-range C7L gene into the genome of NYVAC-B, a recombinant virus that expresses four HIV-1 antigens from clade B (Env, Gag, Pol and Nef) (referred as NYVAC-B-C7L). In the present study, we have compared the in vitro and in vivo behavior of NYVAC-B and NYVAC-B-C7L. In cultured cells, NYVAC-B-C7L expresses higher levels of heterologous antigen than NYVAC-B as determined by Western blot and fluorescent-activated cell sorting to score Gag expressing cells. In a DNA prime/poxvirus boost approach with BALB/c mice, both recombinants elicited robust, broad and multifunctional antigen-specific T-cell responses to the HIV-1 immunogens expressed from the vectors. However, the use of NYVAC-B-C7L as booster significantly enhanced the magnitude of the T cell responses, and induced a more balanced cellular immune response to the HIV-1 antigens in comparison to that elicited in animals boosted with NYVAC-B. CONCLUSIONS/SIGNIFICANCE: These findings demonstrate the possibility to enhance the immunogenicity of the highly attenuated NYVAC vector by the insertion of the host-range gene C7L and suggest the use of this modified vector as an improved vaccine candidate against HIV/AIDS.


Subject(s)
AIDS Vaccines/immunology , Genome, Viral , HIV Antigens/immunology , HIV-1/immunology , Vaccinia virus/genetics , Animals , Cells, Cultured , Genetic Vectors , Humans , Mice , Mice, Inbred BALB C , Vaccinia virus/physiology , Virus Replication
10.
J Gen Virol ; 88(Pt 9): 2473-2478, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17698656

ABSTRACT

Recombinant vaccinia viruses based on the attenuated NYVAC and MVA strains are promising vaccine candidates against a broad spectrum of diseases. Whilst these vectors are safe and immunogenic in animals and humans, little is known about their comparative behaviour in vivo. In this investigation, a head-to-head analysis was carried out of virus dissemination in mice inoculated by the mucosal or systemic route with replication-competent (WRluc) and attenuated recombinant (MVAluc and NYVACluc) viruses expressing the luciferase gene. Bioluminescence imaging showed that, in contrast to WRluc, the attenuated recombinants expressed the reporter gene transiently, with MVAluc expression limited to the first 24 h and NYVACluc giving a longer signal, up to 72 h post-infection, for most of the routes assayed. Moreover, luciferase levels in MVAluc-infected tissues peaked earlier than those in tissues infected by NYVACluc. These findings may be of immunological relevance when these vectors are used as recombinant vaccines.


Subject(s)
Poxviridae/immunology , Viral Vaccines/pharmacology , Animals , Genes, Reporter , Kinetics , Luciferases/genetics , Mice , Poxviridae/drug effects , Vaccines, Attenuated/pharmacology
11.
J Virol ; 81(16): 8707-21, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17537851

ABSTRACT

Although recombinants based on the attenuated poxvirus vectors MVA and NYVAC are currently in clinical trials, the nature of the genes triggered by these vectors in antigen-presenting cells is poorly characterized. Using microarray technology and various analysis conditions, we compared specific changes in gene expression profiling following MVA and NYVAC infection of immature human monocyte-derived dendritic cells (MDDC). Microarray analysis was performed at 6 h postinfection, since these viruses induced extensive cytopathic effects, rRNA breakdown, and apoptosis at late times postinfection. MVA- and NYVAC-infected MDDC shared upregulation of 195 genes compared to uninfected cells: MVA specifically upregulated 359 genes, and NYVAC upregulated 165 genes. Microarray comparison of NYVAC and MVA infection revealed 544 genes with distinct expression patterns after poxvirus infection and 283 genes specifically upregulated after MVA infection. Both vectors upregulated genes for cytokines, cytokine receptors, chemokines, chemokine receptors, and molecules involved in antigen uptake and processing, including major histocompatibility complex genes. mRNA levels for interleukin 12beta (IL-12beta), beta interferon, and tumor necrosis factor alpha were higher after MVA infection than after NYVAC infection. The expression profiles of transcription factors such as NF-kappaB/Rel and STAT were regulated similarly by both viruses; in contrast, OASL, MDA5, and IRIG-I expression increased only during MVA infection. Type I interferon, IL-6, and Toll-like receptor pathways were specifically induced after MVA infection. Following MVA or NYVAC infection in MDDC, we found similarities as well as differences between these virus strains in the expression of cellular genes with immunological function, which should have an impact when these vectors are used as recombinant vaccines.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/virology , Gene Expression Profiling , Genetic Vectors/immunology , Vaccinia virus/immunology , Apoptosis/genetics , Cells, Cultured , Cytokines/genetics , Gene Expression Regulation , Genetic Vectors/genetics , HeLa Cells , Humans , Monocytes/immunology , Oligonucleotide Array Sequence Analysis , Vaccines, Attenuated/immunology , Vaccinia virus/genetics
12.
Vaccine ; 25(11): 1969-92, 2007 Mar 01.
Article in English | MEDLINE | ID: mdl-17224219

ABSTRACT

Recombinants based on the attenuated vaccinia virus strains MVA and NYVAC are considered candidate vectors against different human diseases. In this study we have generated and characterized in BALB/c and in transgenic HHD mice the immunogenicity of two attenuated poxvirus vectors expressing in a single locus (TK) the codon optimized HIV-1 genes encoding gp120 and Gag-Pol-Nef (GPN) polyprotein of clade C (referred as MVA-C and NYVAC-C). In HHD mice primed with either MVA-C or NYVAC-C, or primed with DNA-C and boosted with the poxvirus vectors, the splenic T cell responses against clade C peptides spanning gp120/GPN was broad and mainly directed against Gag-1, Env-1 and Env-2 peptide pools. In BALB/c mice immunized with the homologous or the heterologous combination of poxvirus vectors or with Semliki forest virus (SFV) vectors expressing gp120/GPN, the immune response was also broad but the most immunogenic peptides were Env-1, GPN-1 and GPN-2. Differences in the magnitude of the cellular immune responses were observed between the poxvirus vectors depending on the protocol used. The specific cellular immune response triggered by the poxvirus vectors was Th1 type. The cellular response against the vectors was higher for NYVAC than for MVA in both HHD and BALB/c mice, but differences in viral antigen recognition between the vectors was observed in sera from the poxvirus-immunized animals. These results demonstrate the immunogenic potential of MVA-C and NYVAC-C as novel vaccine candidates against clade C of HIV-1.


Subject(s)
AIDS Vaccines/immunology , HIV Antigens/immunology , HIV-1/immunology , AIDS Vaccines/genetics , Animals , Base Sequence , Codon/genetics , Gene Products, gag/genetics , Gene Products, gag/immunology , Gene Products, nef/genetics , Gene Products, nef/immunology , Gene Products, pol/genetics , Gene Products, pol/immunology , Genetic Vectors , HIV Antigens/genetics , HIV Envelope Protein gp120/genetics , HIV Envelope Protein gp120/immunology , HIV-1/genetics , Humans , Immunization, Secondary , Interferon-gamma/biosynthesis , Interleukin-2/biosynthesis , Mice , Mice, Inbred BALB C , Mice, Transgenic , Models, Animal , Molecular Sequence Data , Semliki forest virus , Spleen/immunology , T-Lymphocytes/immunology , Vaccines, DNA/genetics , Vaccines, DNA/immunology , Vaccinia virus , Viral Vaccines , nef Gene Products, Human Immunodeficiency Virus
13.
Vaccine ; 25(15): 2863-85, 2007 Apr 12.
Article in English | MEDLINE | ID: mdl-17113200

ABSTRACT

In this investigation we have generated and defined the immunogenicity of two novel HIV/AIDS vaccine candidates based on the highly attenuated vaccinia virus strains, MVA and NYVAC, efficiently expressing in the same locus (TK) and under the same viral promoter the codon optimized HIV-1 genes encoding gp120 and Gag-Pol-Nef antigens of clade B (referred as MVA-B and NYVAC-B). In infected human HeLa cells, gp120 is released from cells and GPN is produced as a polyprotein; NYVAC-B induces severe apoptosis but not MVA-B. The two poxvirus vectors showed genetic stability of the inserts. In BALB/c and in transgenic HHD mice for human HLA-A2 class I, both vectors are efficient immunogens and induced broad cellular immune responses against peptides represented in the four HIV-1 antigens. Some differences were observed in the magnitude and breadth of the immune response in the mouse models. In DNA prime/poxvirus boost protocols, the strongest immune response, as measured by fresh IFN-gamma and IL-2 ELISPOT, was obtained in BALB/c mice boosted with NYVAC-B, while in HHD mice there were no differences between the poxvirus vectors. When the prime/boost was performed with homologous or with combination of poxvirus vectors, the protocols MVA-B/MVA-B and NYVAC-B/NYVAC-B, or the combination NYVAC-B/MVA-B gave the most consistent broader immune response in both mouse models, although the magnitude of the overall response was higher for the DNA-B/poxvirus-B regime. All of the immunization protocols induced some humoral response against the gp160 protein from HIV-1 clone LAV. Our findings indicate that MVA-B and NYVAC-B meet the criteria to be potentially useful vaccine candidates against HIV/AIDS.


Subject(s)
AIDS Vaccines/immunology , Antigens, Viral/immunology , HIV Envelope Protein gp120/immunology , Viral Vaccines/immunology , AIDS Vaccines/genetics , Animals , Antigens, Viral/biosynthesis , Antigens, Viral/genetics , Apoptosis/immunology , Base Sequence , Chick Embryo , Fusion Proteins, gag-pol/biosynthesis , Fusion Proteins, gag-pol/genetics , Fusion Proteins, gag-pol/immunology , Gene Products, nef/biosynthesis , Gene Products, nef/genetics , Gene Products, nef/immunology , Genomic Instability , HIV Envelope Protein gp120/biosynthesis , HIV Envelope Protein gp120/genetics , HLA-A2 Antigen/immunology , HeLa Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Transgenic , Molecular Sequence Data , Polymerase Chain Reaction/methods , Poxviridae/genetics , Poxviridae/immunology , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Viral Vaccines/genetics
14.
J Virol ; 80(12): 6033-47, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16731942

ABSTRACT

The poxvirus strains NYVAC and MVA are two candidate vectors for the development of vaccines against a broad spectrum of diseases. Although these attenuated virus strains have proven to be safe in animals and humans, little is known about their comparative behavior in vitro. In contrast with MVA, NYVAC infection triggers greater cytopathic effect in a range of permissive and nonpermissive cell lines. The yields of NYVAC cell-associated virus in permissive cells (BHK-21) were slightly reduced compared with those of MVA infection. During the course of infection in HeLa cells, there is a translational block induced by NYVAC late in infection, which correlated with a marked increase in phosphorylation levels of the initiation factor eIF-2alpha. In contrast to MVA, the synthesis of certain late viral proteins was only blocked in NYVAC-infected HeLa cells. Electron microscopy (EM) analysis revealed that morphogenesis of NYVAC in HeLa cells was blocked at the stage of formation of immature viral forms. Phase-contrast microscopy, EM, flow cytometry, and rRNA analyses demonstrated that contrary to MVA, NYVAC infection induces potent apoptosis, a phenomenon dependent on activation of caspases and RNase L. Apoptosis induced by NYVAC was prevented when the virus gene C7L was placed back into the NYVAC genome, recovering the ability of NYVAC to replicate in HeLa cells and maintaining the attenuated phenotype in mice. Overall, our findings demonstrate distinct behavior between NYVAC and MVA strains in cultured cells, as well as a new role for the C7L viral gene as an inhibitor of apoptosis in NYVAC infection.


Subject(s)
Genes, Viral/physiology , Poxviridae/pathogenicity , Viral Vaccines , Animals , Apoptosis , HeLa Cells , Humans , Mice , Poxviridae Infections , Vaccines, Attenuated , Virus Replication
15.
J Virol ; 80(2): 985-98, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16379000

ABSTRACT

NYVAC has been engineered as a safe, attenuated vaccinia virus (VV) vector for use in vaccination against a broad spectrum of pathogens and tumors. Due to the interest in NYVAC-based vectors as vaccines and current phase I/II clinical trials with this vector, there is a need to analyze the human host response to NYVAC infection. Using high-density cDNA microarrays, we found 368 differentially regulated genes after NYVAC infection of HeLa cells. Clustering of the regulated genes identified six discrete gene clusters with altered expression patterns. Clusters 1 to 3 represented 47.5% of the regulated genes, with three patterns of gene activation kinetics, whereas clusters 4 to 6 showed distinct repression kinetics. Quantitative real-time reverse transcription-PCR analysis of selected genes validated the array data. Upregulated transcripts correlated with genes implicated in immune responses, including those encoding interleukin-1 receptor 2 (IL-1R2), IL-6, ISG-15, CD-80, and TNFSF7. NYVAC upregulated several intermediates of apoptotic cascades, including caspase-9, correlating with its ability to induce apoptosis. NYVAC infection also stimulated the expression of NF-kappaB1 and NF-kappaB2 as well as that of NF-kappaB target genes. Expression of the VV host range K1L gene during NYVAC infection prevented NF-kappaB activation, but not the induction of apoptosis. This study is the first overall analysis of the transcriptional response of human cells to NYVAC infection and provides a framework for future functional studies to evaluate this vector and its derivatives as human vaccines.


Subject(s)
Apoptosis , NF-kappa B/metabolism , Vaccinia virus/physiology , Activating Transcription Factors/genetics , Activating Transcription Factors/metabolism , Apoptosis/genetics , Blood Proteins/genetics , Blood Proteins/metabolism , Caspase 9 , Caspases/genetics , Caspases/metabolism , Gene Expression Profiling , HeLa Cells/metabolism , Humans , NF-kappa B/genetics , Protein Array Analysis , Up-Regulation
16.
J Immunol ; 172(10): 6209-20, 2004 May 15.
Article in English | MEDLINE | ID: mdl-15128809

ABSTRACT

Vaccines intended to prevent mucosal transmission of HIV should be able to induce multiple immune effectors in the host including Abs and cell-mediated immune responses at mucosal sites. The aim of this study was to characterize and to enhance the immunogenicity of a recombinant modified vaccinia virus Ankara (MVA) expressing HIV-1 Env IIIB Ag (MVAenv) inoculated in BALB/c mice by mucosal routes. Intravaginal inoculation of MVAenv was not immunogenic, whereas intranasally it induced a significant immune response to the HIV Ag. Intranasal codelivery of MVAenv plus cholera toxin (CT) significantly enhanced the cellular and humoral immune response against Env in the spleen and genitorectal draining lymph nodes, respectively. Heterologous DNAenv prime-MVAenv boost by intranasal immunization, together with CT, produced a cellular immune response in the spleen 10-fold superior to that in the absence of CT. A key finding of these studies was that both MVAenv/MVAenv and DNAenv/MVAenv schemes, plus CT, induced a specific mucosal CD8(+) T cell response in genital tissue and draining lymph nodes. In addition, both immunizations also generated systemic Abs, and more importantly, mucosal IgA and IgG Abs in vaginal washings. Specific secretion of beta-chemokines was also generated by both immunizations, with a stronger response in mice immunized by the DNA-CT/MVA-CT regimen. Our findings are of relevance in the area of vaccine development and support the optimization of protocols of immunization based on MVA as vaccine vectors to induce mucosal immune responses against HIV.


Subject(s)
AIDS Vaccines/administration & dosage , Adjuvants, Immunologic/administration & dosage , HIV-1/immunology , Immunization Schedule , Immunization, Secondary , Urogenital System/immunology , Vaccines, DNA/administration & dosage , Vaccinia virus/genetics , AIDS Vaccines/genetics , AIDS Vaccines/immunology , Adjuvants, Immunologic/genetics , Administration, Intranasal , Administration, Intravaginal , Animals , Cholera Toxin/administration & dosage , Cholera Toxin/immunology , Female , Gene Products, env/biosynthesis , Gene Products, env/immunology , Genetic Vectors , HIV Antibodies/biosynthesis , HIV-1/genetics , Immunity, Cellular , Immunity, Mucosal/genetics , Immunization, Secondary/methods , Interferon-gamma/metabolism , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymph Nodes/virology , Mice , Mice, Inbred BALB C , Rectum/immunology , Rectum/pathology , Rectum/virology , Urogenital System/virology , Vaccines, DNA/genetics , Vaccines, DNA/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Vaccinia virus/immunology , Vaccinia virus/physiology , Virus Replication/immunology
17.
J Virol ; 77(12): 7048-57, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12768024

ABSTRACT

Vaccines that elicit systemic and mucosal immune responses should be the choice to control human immunodeficiency virus (HIV) infections. We have previously shown that prime-boost immunizations with influenza virus Env and vaccinia virus (VV) WR Env recombinants induced an enhanced systemic CD8(+) T-cell response against HIV-1 Env antigen. In this report, we analyzed in BALB/c mice after priming with influenza virus Env the ability of two VV recombinants expressing HIV-1 Env B (VV WR Env and the highly attenuated modified VV Ankara [MVA] Env) to boost cellular immune responses in the spleen and in the lymph nodes draining the genital and rectal tracts. Groups of mice were primed by the intranasal route with 10(4) PFU of influenza virus Env and boosted 14 days later by the intraperitoneal or intranasal route with 10(7) PFU of MVA Env or VV WR Env, while the control group received two immunizations with influenza virus Env. We found that the combined immunization (Flu/VV) increased more than 60 times the number of gamma interferon-specific CD8(+) T cells compared to the Flu/Flu scheme. Significantly, boosting with MVA Env by the intraperitoneal route induced a response 1.25 or 2.5 times (spleen or genital lymph nodes) higher with respect to that found after the boost with VV WR Env. Mice with an enhanced CD8(+) T-cell response also had an increased Th1/Th2 ratio, evaluated by the cytokine pattern secreted following in vitro restimulation with gp160 protein and by the specific immunoglobulin G2a (IgG2a)/IgG1 ratio in serum. By the intranasal route recombinant WR Env booster gave a more efficient immune response (10 and 1.3 times in spleen and genital lymph nodes, respectively) than recombinant MVA Env. However, the scheme influenza virus Env/MVA Env increased four times the response in the spleen, giving a low but significant response in the genital lymph nodes compared with a single intranasal immunization with MVA Env. These results demonstrate that the combination Flu/MVA in prime-booster immunization regimens is an effective vaccination approach to generate cellular immune responses to HIV antigens at sites critical for protective responses.


Subject(s)
AIDS Vaccines/administration & dosage , Gene Products, env/immunology , Genetic Vectors/administration & dosage , Orthomyxoviridae/immunology , Vaccinia virus/immunology , 3T3 Cells , AIDS Vaccines/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Line , Gene Products, env/genetics , Genetic Vectors/immunology , Genitalia/immunology , HIV Antibodies/blood , HIV Envelope Protein gp120/genetics , HIV Envelope Protein gp120/immunology , HIV Infections/prevention & control , Humans , Immunization Schedule , Immunization, Secondary , Lymph Nodes/immunology , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Orthomyxoviridae/genetics , Peptide Fragments/genetics , Peptide Fragments/immunology , Recombination, Genetic , Rectum/immunology , Vaccinia virus/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...