Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Vaccine ; 32(12): 1398-406, 2014 Mar 10.
Article in English | MEDLINE | ID: mdl-24462481

ABSTRACT

The rational design and development of genetically attenuated HSV-2 mutant viruses represent an attractive approach for developing both prophylactic and therapeutic vaccines for genital herpes. Previously, HSV-2 UL24 was shown to be a virulence determinant in both murine and guinea pig vaginal infection models. An UL24-ßgluc insertion mutant produced syncytial plaques and replicated to nearly wild type levels in tissue culture, but induced little or no pathological effects in recipient mice or guinea pigs following vaginal infection. Here we report that immunization of mice or guinea pigs with high or low doses of UL24-ßgluc elicited a highly protective immune response. UL24-ßgluc immunization via the vaginal or intramuscular routes was demonstrated to protect mice from a lethal vaginal challenge with wild type HSV-2. Moreover, antigen re-stimulated splenic lymphocytes harvested from immunized mice exhibited both HSV-2 specific CTL activity and IFN-γ expression. Humoral anti-HSV-2 responses in serum were Th1-polarized (IgG2a>IgG1) and contained high-titer anti-HSV-2 neutralizing activity. Guinea pigs vaccinated subcutaneously with UL24-ßgluc or the more virulent parental strain (186) were challenged with a heterologous HSV-2 strain (MS). Acute disease scores were nearly indistinguishable in guinea pigs immunized with either virus. Recurrent disease scores were reduced in UL24-ßgluc immunized animals but not to the same extent as those immunized with strain 186. In addition, challenge virus was not detected in 75% of guinea pigs subcutaneously immunized with UL24-ßgluc. In conclusion, disruption of the UL24 gene is a prime target for the development of a genetically attenuated live HSV-2 vaccine.


Subject(s)
Herpes Genitalis/prevention & control , Herpes Simplex Virus Vaccines/immunology , Herpesvirus 2, Human/genetics , Vagina/virology , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Disease Models, Animal , Female , Guinea Pigs , Herpesvirus 2, Human/pathogenicity , Immunity, Cellular , Immunity, Humoral , Immunoglobulin G/blood , Interferon-gamma/immunology , Mice , Mice, Inbred BALB C , Mutagenesis, Insertional , T-Lymphocytes, Cytotoxic/immunology , Vaccines, Attenuated/immunology , Vagina/immunology , Viral Proteins/genetics , Virulence
2.
Vaccine ; 27(22): 2930-9, 2009 May 14.
Article in English | MEDLINE | ID: mdl-19428903

ABSTRACT

Recombinant vesicular stomatitis viruses (rVSVs) are being developed as potential HIV-1 vaccine candidates. To characterize the in vivo replication and dissemination of rVSV vectors in mice, high doses of a highly attenuated vector expressing HIV-1 Gag, rVSV(IN)-N4CT9-Gag1, and a prototypic reference virus, rVSV(IN)-HIVGag5, were delivered intramuscularly (IM), intranasally (IN), or intravenously (IV). We used quantitative, real-time RT-PCR (Q-PCR) and standard plaque assays to measure the temporal dissemination of these viruses to various tissues. Following IM inoculation, both viruses were detected primarily at the injection site as well as in draining lymph nodes; neither virus induced significant weight loss, pathologic signs, or evidence of neuroinvasion. In contrast, following IN inoculation, the prototypic virus was detected in all tissues tested and caused significant weight loss leading to death. IN administration of rVSV(IN)-N4CT9-Gag1 resulted in detection in numerous tissues (brain, lung, nasal turbinates, and lymph nodes) albeit in significantly reduced levels, which caused little or no weight loss nor any mortality. Following IV inoculation, both prototypic and attenuated viruses were detected by Q-PCR in all tissues tested. In contrast to the prototype, rVSV(IN)-N4CT9-Gag1 viral loads were significantly lower in all organs tested, and no infectious virus was detected in the brain following IV inoculation, despite the presence of viral RNA. These studies demonstrated significant differences in the biodistribution patterns of and the associated pathogenicity engendered by the prototypic and attenuated vectors in a highly susceptible host.


Subject(s)
AIDS Vaccines/administration & dosage , AIDS Vaccines/pharmacokinetics , Genetic Vectors , Vesiculovirus/growth & development , Vesiculovirus/genetics , gag Gene Products, Human Immunodeficiency Virus/genetics , AIDS Vaccines/adverse effects , Administration, Intranasal , Animals , Female , Injections, Intramuscular , Injections, Intravenous , Mice , Mice, Inbred BALB C , RNA, Viral/genetics , Reverse Transcriptase Polymerase Chain Reaction , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/adverse effects , Vaccines, Attenuated/pharmacokinetics , Viral Plaque Assay
3.
J Virol ; 82(1): 207-19, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17942549

ABSTRACT

Recombinant vesicular stomatitis virus (rVSV) has shown great potential as a new viral vector for vaccination. However, the prototypic rVSV vector described previously was found to be insufficiently attenuated for clinical evaluation when assessed for neurovirulence in nonhuman primates. Here, we describe the attenuation, neurovirulence, and immunogenicity of rVSV vectors expressing human immunodeficiency virus type 1 Gag. These rVSV vectors were attenuated by combinations of the following manipulations: N gene translocations (N4), G gene truncations (CT1 or CT9), noncytopathic M gene mutations (Mncp), and positioning of the gag gene into the first position of the viral genome (gag1). The resulting N4CT1-gag1, N4CT9-gag1, and MncpCT1-gag1 vectors demonstrated dramatically reduced neurovirulence in mice following direct intracranial inoculation. Surprisingly, in spite of a very high level of attenuation, the N4CT1-gag1 and N4CT9-gag1 vectors generated robust Gag-specific immune responses following intramuscular immunization that were equivalent to or greater than immune responses generated by the more virulent prototypic vectors. MncpCT1-gag1 also induced Gag-specific immune responses following intramuscular immunization that were equivalent to immune responses generated by the prototypic rVSV vector. Placement of the gag gene in the first position of the VSV genome was associated with increased in vitro expression of Gag protein, in vivo expression of Gag mRNA, and enhanced immunogenicity of the vector. These findings demonstrate that through directed manipulation of the rVSV genome, vectors that have reduced neurovirulence and enhanced immunogenicity can be made.


Subject(s)
AIDS Vaccines/immunology , Genetic Vectors , HIV-1/genetics , Vesiculovirus/genetics , Viral Vaccines/immunology , AIDS Vaccines/genetics , Animals , Cytokines/biosynthesis , HIV Antibodies/blood , Injections, Intramuscular , Lethal Dose 50 , Mice , Mice, Inbred BALB C , Point Mutation , Recombination, Genetic , Sequence Deletion , T-Lymphocytes, Cytotoxic/immunology , Translocation, Genetic , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Viral Vaccines/genetics , Virulence , gag Gene Products, Human Immunodeficiency Virus/genetics , gag Gene Products, Human Immunodeficiency Virus/immunology
4.
Vaccine ; 25(12): 2296-305, 2007 Mar 08.
Article in English | MEDLINE | ID: mdl-17239997

ABSTRACT

The safety of a propagation-defective Venezuelan equine encephalitis virus (VEEV) replicon particle vaccine was examined in mice. After intracranial inoculation we observed approximately 5% body weight loss, modest inflammatory changes in the brain, genome replication, and foreign gene expression. These changes were transient and significantly less severe than those caused by TC-83, a live-attenuated vaccinal strain of VEEV that has been safely used to immunize military personnel and laboratory workers. Replicon particles injected intramuscularly or intravenously were detected at limited sites 3 days post-administration, and were undetectable by day 22. There was no evidence of dissemination to spinal cord or brain after systemic administration. These results demonstrate that propagation-defective VEEV replicon particles are minimally neurovirulent and lack neuroinvasive potential.


Subject(s)
Encephalitis Virus, Venezuelan Equine/immunology , Encephalomyelitis, Venezuelan Equine/immunology , Replicon/genetics , Viral Envelope Proteins/genetics , Animals , Body Weight , Brain/metabolism , Brain/virology , Encephalitis Virus, Venezuelan Equine/genetics , Encephalomyelitis, Venezuelan Equine/prevention & control , Injections, Intramuscular , Injections, Intravenous , Mice , RNA, Viral/genetics , RNA, Viral/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Spinal Cord/metabolism , Spinal Cord/virology , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Virus Replication/genetics
5.
Cell Immunol ; 239(2): 113-20, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16762332

ABSTRACT

The glycoprotein D of HSV-2 (gD2) is currently a leading candidate vaccine target for genital herpes vaccines as both cellular and humoral responses can be generated against it. However, little is known about how vaccine composition will affect T cell epitope selection. A panel of 15-mer peptides (with 11 amino acid overlap) spanning full-length gD2 was used to investigate the fine specificity of T cell responses to gD2 as well as the role of vaccine composition on epitope selection. Spleen cells from BALB/c mice (H-2(d)) immunized with gD2, formulated with or without AlPO(4) and/or IL-12, were stimulated in vitro with overlapping gD2 peptides. Cellular responses (lymphoproliferation and IFN-gamma expression) were mapped to four epitopes within the gD2 molecule: gD2(49-63), gD2(105-119), gD2(245-259), and gD2(333-347). CTL analysis of these four epitopes indicated that not all of them could serve as a CTL epitope. Mice immunized with gD2 expressed from a viral vector mounted CTL responses primarily to one epitope located in the extracellular domain of gD2 (gD2(245-259)). More importantly, mice immunized with gD2 co-administered with IL-12 mounted CTL responses to an additional epitope located at the transmembrane-cytoplasmic junction of gD2 (gD2(333-347)). The location of this novel epitope emphasizes the benefit of using full-length versions of glycoproteins when designing vaccine components.


Subject(s)
CD4 Antigens/metabolism , Cell Membrane/immunology , Cytoplasm/immunology , Epitope Mapping , Epitopes, T-Lymphocyte/biosynthesis , Herpesvirus 2, Human/immunology , T-Lymphocytes, Cytotoxic/immunology , Viral Envelope Proteins/immunology , Amino Acid Sequence , Animals , CHO Cells , Chlorocebus aethiops , Cricetinae , Cricetulus , Epitopes, T-Lymphocyte/genetics , Female , Herpesvirus 2, Human/genetics , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Peptide Fragments/immunology , T-Lymphocytes, Cytotoxic/metabolism , Vero Cells , Viral Envelope Proteins/genetics
6.
J Virol ; 80(9): 4447-57, 2006 May.
Article in English | MEDLINE | ID: mdl-16611905

ABSTRACT

Recombinant vesicular stomatitis virus (rVSV) vectors offer an attractive approach for the induction of robust cellular and humoral immune responses directed against human pathogen target antigens. We evaluated rVSV vectors expressing full-length glycoprotein D (gD) from herpes simplex virus type 2 (HSV-2) in mice and guinea pigs for immunogenicity and protective efficacy against genital challenge with wild-type HSV-2. Robust Th1-polarized anti-gD immune responses were demonstrated in the murine model as measured by induction of gD-specific cytotoxic T lymphocytes and increased gamma interferon expression. The isotype makeup of the serum anti-gD immunoglobulin G (IgG) response was consistent with the presence of a Th1-CD4+ anti-gD response, characterized by a high IgG2a/IgG1 IgG subclass ratio. Functional anti-HSV-2 neutralizing serum antibody responses were readily demonstrated in both guinea pigs and mice that had been immunized with rVSV-gD vaccines. Furthermore, guinea pigs and mice were prophylactically protected from genital challenge with high doses of wild-type HSV-2. In addition, guinea pigs were highly protected against the establishment of latent infection as evidenced by low or absent HSV-2 genome copies in dorsal root ganglia after virus challenge. In summary, rVSV-gD vectors were successfully used to elicit potent anti-gD Th1-like cellular and humoral immune responses that were protective against HSV-2 disease in guinea pigs and mice.


Subject(s)
Herpes Simplex Virus Vaccines/immunology , Herpesvirus 2, Human/immunology , Th1 Cells/immunology , Vagina/immunology , Vagina/virology , Vesicular stomatitis Indiana virus/genetics , Viral Envelope Proteins/immunology , Animals , Antibody Formation/immunology , Female , Genetic Vectors/genetics , Glycoproteins/genetics , Glycoproteins/immunology , Glycoproteins/metabolism , Guinea Pigs , Herpes Simplex Virus Vaccines/genetics , Herpesvirus 2, Human/genetics , Herpesvirus 2, Human/metabolism , Mice , Models, Animal , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism
7.
J Virol ; 79(16): 10498-506, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16051842

ABSTRACT

A herpes simplex virus type 2 (HSV-2) UL24 beta-glucuronidase (UL24-betagluc) insertion mutant was derived from HSV-2 strain 186 via standard marker transfer techniques. Cell monolayers infected with UL24-betagluc yielded cytopathic effect with syncytium formation. UL24-betagluc replicated to wild-type viral titers in three different cell lines. UL24-betagluc was not virulent after intravaginal inoculation of BALB/c mice in that all inoculated animals survived doses up to 400 times the 50% lethal dose (LD50) of the parental virus. Furthermore, few UL24-betagluc-inoculated mice developed any vaginal lesions. Intravaginal inoculation of guinea pigs with UL24-betagluc at a dose equivalent to the LD50 of parental virus (approximately 5 x 10(3) PFU) was not lethal (10/10 animals survived). Although genital lesions developed in some UL24-betagluc-inoculated guinea pigs, both the overall number of lesions and the severity of disease were far less than that observed for animals infected with parental strain 186.


Subject(s)
Disease Models, Animal , Herpes Simplex/etiology , Herpesvirus 2, Human/genetics , Viral Proteins/genetics , Animals , Base Sequence , Female , Genome, Viral , Guinea Pigs , Herpesvirus 2, Human/pathogenicity , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Thymidine Kinase/physiology , Virulence , Virus Replication
8.
Vaccine ; 23(2): 236-46, 2004 Nov 25.
Article in English | MEDLINE | ID: mdl-15531043

ABSTRACT

The type of immune response elicited against HSV-2 infection may be a factor in the frequency and severity of recurrent disease, with non-recurrent status being associated with a Th1-like response. As administration of glycoprotein D subunit formulated with an aluminum-based adjuvant induces predominantly Th2-like immune responses, we sought to assess the ability of IL-12 to redirect anti-HSV immunity towards a Th1 response. Co-administration of gD with IL-12 resulted in gD-specific antibody subclass switching from predominantly IgG1 observed in mice immunized with either gD or gD/AlPO4 to a more balanced combination of IgG1 and IgG2a, and enhanced virus neutralizing activity. Spleen cells from mice immunized with gD and IL-12, and restimulated in vitro with HSV-2, developed into effector cells capable of secreting IFN-gamma and lysing HSV-2 infected targets, while those obtained from gD or gD/ALPO4 immunized mice did not express lytic activity. In vitro studies determined that these CTLs were CD4+ and that the cytotoxicity was primarily perforin dependent. Vaginal challenge with HSV-2 demonstrated that IL-12 co-administration with gD resulted in increased efficacy of this vaccine as compared to administration of gD antigen alone. This acquired protection persisted up to 1 year. Finally, adsorbing gD and IL-12 to AlPO4 decreased the optimal dose of IL-12 required to enhance gD immunogenicity and shift responses towards a Th1-like profile.


Subject(s)
Adjuvants, Immunologic , Aluminum Compounds/chemistry , Immunity/drug effects , Interleukin-12/pharmacology , Phosphates/chemistry , Animals , Antibodies/chemistry , Disease Models, Animal , Herpes Simplex/immunology , Herpes Simplex/prevention & control , Herpesvirus 2, Human/immunology , Humans , Interleukin-12/immunology , Mice , Mice, Inbred BALB C , Solubility/drug effects , Th1 Cells/drug effects , Th1 Cells/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...