Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Viruses ; 15(5)2023 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-37243234

RESUMO

Herpes simplex virus type 2 (HSV-2) is a leading cause of genital ulcer disease and a major risk factor for acquisition and transmission of HIV. Frequent recurrent genital lesions and concerns about transmitting infection to intimate partners affect the quality of life of infected individuals. Therapeutic vaccines are urgently needed to reduce the frequency of genital lesions and transmission. S-540956 is a novel vaccine adjuvant that contains CpG oligonucleotide ODN2006 annealed to its complementary sequence and conjugated to a lipid that targets the adjuvant to lymph nodes. Our primary goal was to compare S-540956 administered with HSV-2 glycoprotein D (gD2) with no treatment in a guinea pig model of recurrent genital herpes (studies 1 and 2). Our secondary goals were to compare S-540956 with oligonucleotide ODN2006 (study1) or glucopyranosyl lipid A in a stable oil-in-water nano-emulsion (GLA-SE) (study 2). gD2/S-540956 reduced the number of days with recurrent genital lesions by 56%, vaginal shedding of HSV-2 DNA by 49%, and both combined by 54% compared to PBS, and was more efficacious than the two other adjuvants. Our results indicate that S-540956 has great potential as an adjuvant for a therapeutic vaccine for genital herpes, and merits further evaluation with the addition of potent T cell immunogens.


Assuntos
Herpes Genital , Vacinas , Feminino , Cobaias , Animais , Herpes Genital/prevenção & controle , Herpesvirus Humano 2/genética , Anticorpos Neutralizantes , Anticorpos Antivirais , Qualidade de Vida , Proteínas do Envelope Viral , Adjuvantes Imunológicos , Genitália , Linfonodos , DNA
2.
J Clin Invest ; 131(23)2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34618692

RESUMO

Nucleoside-modified mRNA vaccines have gained global attention because of COVID-19. We evaluated a similar vaccine approach for preventing a chronic, latent genital infection rather than an acute respiratory infection. We used animal models to compare an HSV-2 trivalent nucleoside-modified mRNA vaccine with the same antigens prepared as proteins, with an emphasis on antigen-specific memory B cell responses and immune correlates of protection. In guinea pigs, serum neutralizing-antibody titers were higher at 1 month and declined far less by 8 months in mRNA- compared with protein-immunized animals. Both vaccines protected against death and genital lesions when infected 1 month after immunization; however, protection was more durable in the mRNA group compared with the protein group when infected after 8 months, an interval representing greater than 15% of the animal's lifespan. Serum and vaginal neutralizing-antibody titers correlated with protection against infection, as measured by genital lesions and vaginal virus titers 2 days after infection. In mice, the mRNA vaccine generated more antigen-specific memory B cells than the protein vaccine at early times after immunization that persisted for up to 1 year. High neutralizing titers and robust B cell immune memory likely explain the more durable protection by the HSV-2 mRNA vaccine.


Assuntos
Herpes Genital , Herpesvirus Humano 2/imunologia , Memória Imunológica , Células B de Memória/imunologia , RNA Viral/imunologia , Vacinas Sintéticas/imunologia , Vacinas Virais/imunologia , Animais , COVID-19/imunologia , COVID-19/prevenção & controle , Modelos Animais de Doenças , Feminino , Cobaias , Herpes Genital/imunologia , Herpes Genital/prevenção & controle , SARS-CoV-2/imunologia , Vacinas de mRNA
3.
J Virol ; 88(4): 2000-10, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24284325

RESUMO

A prophylactic vaccine for genital herpes disease remains an elusive goal. We report the results of two studies performed collaboratively in different laboratories that assessed immunogenicity and vaccine efficacy in herpes simplex virus 1 (HSV-1)-seropositive guinea pigs immunized and subsequently challenged intravaginally with HSV-2. In study 1, HSV-2 glycoproteins C (gC2) and D (gD2) were produced in baculovirus and administered intramuscularly as monovalent or bivalent vaccines with CpG and alum. In study 2, gD2 was produced in CHO cells and given intramuscularly with monophosphoryl lipid A (MPL) and alum, or gC2 and gD2 were produced in glycoengineered Pichia pastoris and administered intramuscularly as a bivalent vaccine with Iscomatrix and alum to HSV-1-naive or -seropositive guinea pigs. In both studies, immunization boosted neutralizing antibody responses to HSV-1 and HSV-2. In study 1, immunization with gC2, gD2, or both immunogens significantly reduced the frequency of genital lesions, with the bivalent vaccine showing the greatest protection. In study 2, both vaccines were highly protective against genital disease in naive and HSV-1-seropositive animals. Comparisons between gD2 and gC2/gD2 in study 2 must be interpreted cautiously, because different adjuvants, gD2 doses, and antigen production methods were used; however, significant differences invariably favored the bivalent vaccine. Immunization of naive animals with gC2/gD2 significantly reduced the number of days of vaginal shedding of HSV-2 DNA compared with that for mock-immunized animals. Surprisingly, in both studies, immunization of HSV-1-seropositive animals had little effect on recurrent vaginal shedding of HSV-2 DNA, despite significantly reducing genital disease.


Assuntos
Herpes Genital/prevenção & controle , Herpesvirus Humano 1/imunologia , Proteínas do Envelope Viral/imunologia , Vacinas Virais/farmacologia , Análise de Variância , Animais , Anticorpos Neutralizantes/imunologia , Baculoviridae , Células CHO , Cricetinae , Cricetulus , Ensaio de Imunoadsorção Enzimática , Feminino , Cobaias , Injeções Intramusculares , Lipídeo A/análogos & derivados , Pichia , Reação em Cadeia da Polimerase em Tempo Real , Vacinas Virais/administração & dosagem
4.
J Virol ; 86(8): 4586-98, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22318147

RESUMO

A herpes simplex virus 2 (HSV-2) glycoprotein E deletion mutant (gE2-del virus) was evaluated as a replication-competent, attenuated live virus vaccine candidate. The gE2-del virus is defective in epithelial cell-to-axon spread and in anterograde transport from the neuron cell body to the axon terminus. In BALB/c and SCID mice, the gE2-del virus caused no death or disease after vaginal, intravascular, or intramuscular inoculation and was 5 orders of magnitude less virulent than wild-type virus when inoculated directly into the brain. No infectious gE2-del virus was recovered from dorsal root ganglia (DRG) after multiple routes of inoculation; however, gE2-del DNA was detected by PCR in lumbosacral DRG at a low copy number in some mice. Importantly, no recurrent vaginal shedding of gE2-del DNA was detected in immunized guinea pigs. Intramuscular immunization outperformed subcutaneous immunization in all parameters evaluated, although individual differences were not significant, and two intramuscular immunizations were more protective than one. Immunized animals had reduced vaginal disease, vaginal titers, DRG infection, recurrent genital lesions, and recurrent vaginal shedding of HSV-2 DNA; however, protection was incomplete. A combined modality immunization using live virus and HSV-2 glycoprotein C and D subunit antigens in guinea pigs did not totally eliminate recurrent lesions or recurrent vaginal shedding of HSV-2 DNA. The gE2-del virus used as an immunotherapeutic vaccine in previously HSV-2-infected guinea pigs greatly reduced the frequency of recurrent genital lesions. Therefore, the gE2-del virus is safe, other than when injected at high titer into the brain, and is efficacious as a prophylactic and immunotherapeutic vaccine.


Assuntos
Deleção de Genes , Vacinas contra o Vírus do Herpes Simples/imunologia , Herpesvirus Humano 2/genética , Herpesvirus Humano 2/imunologia , Neurônios/virologia , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Linhagem Celular , Chlorocebus aethiops , DNA Viral , Feminino , Gânglios Espinais/virologia , Cobaias , Herpes Genital/mortalidade , Herpes Genital/prevenção & controle , Herpes Genital/terapia , Herpes Simples/mortalidade , Herpes Simples/prevenção & controle , Herpes Simples/terapia , Vacinas contra o Vírus do Herpes Simples/administração & dosagem , Vacinas contra o Vírus do Herpes Simples/efeitos adversos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Medula Espinal/virologia , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/efeitos adversos , Vacinas Atenuadas/imunologia
5.
J Virol ; 85(20): 10472-86, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21813597

RESUMO

Attempts to develop a vaccine to prevent genital herpes simplex virus 2 (HSV-2) disease have been only marginally successful, suggesting that novel strategies are needed. Immunization with HSV-2 glycoprotein C (gC-2) and gD-2 was evaluated in mice and guinea pigs to determine whether adding gC-2 to a gD-2 subunit vaccine would improve protection by producing antibodies that block gC-2 immune evasion from complement. Antibodies produced by gC-2 immunization blocked the interaction between gC-2 and complement C3b, and passive transfer of gC-2 antibody protected complement-intact mice but not C3 knockout mice against HSV-2 challenge, indicating that gC-2 antibody is effective, at least in part, because it prevents HSV-2 evasion from complement. Immunization with gC-2 also produced neutralizing antibodies that were active in the absence of complement; however, the neutralizing titers were higher when complement was present, with the highest titers in animals immunized with both antigens. Animals immunized with the gC-2-plus-gD-2 combination had robust CD4+ T-cell responses to each immunogen. Multiple disease parameters were evaluated in mice and guinea pigs immunized with gC-2 alone, gD-2 alone, or both antigens. In general, gD-2 outperformed gC-2; however, the gC-2-plus-gD-2 combination outperformed gD-2 alone, particularly in protecting dorsal root ganglia in mice and reducing recurrent vaginal shedding of HSV-2 DNA in guinea pigs. Therefore, the gC-2 subunit antigen enhances a gD-2 subunit vaccine by stimulating a CD4+ T-cell response, by producing neutralizing antibodies that are effective in the absence and presence of complement, and by blocking immune evasion domains that inhibit complement activation.


Assuntos
Gânglios Espinais/imunologia , Herpes Genital/prevenção & controle , Vacina contra Herpes Zoster/imunologia , Herpes Zoster/prevenção & controle , Herpesvirus Humano 2/imunologia , Proteínas do Envelope Viral/imunologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Linfócitos T CD4-Positivos/imunologia , Feminino , Cobaias , Herpes Genital/imunologia , Herpes Zoster/imunologia , Vacina contra Herpes Zoster/administração & dosagem , Imunização/métodos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Prevenção Secundária , Vagina/virologia , Eliminação de Partículas Virais
6.
J Virol ; 85(7): 3239-49, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21228231

RESUMO

Herpes simplex virus 1 (HSV-1) glycoprotein E (gE) mediates cell-to-cell spread and functions as an IgG Fc receptor (FcγR) that blocks the Fc domain of antibody targeting the virus or infected cell. Efforts to assess the functions of the HSV-1 FcγR in vivo have been hampered by difficulties in preparing an FcγR-negative strain that is relatively intact for spread. Here we report the FcγR and spread phenotypes of NS-gE264, which is a mutant strain that has four amino acids inserted after gE residue 264. The virus is defective in IgG Fc binding yet causes zosteriform disease in the mouse flank model that is only minimally reduced compared with wild-type and the rescue strains. The presence of zosteriform disease suggests that NS-gE264 spread functions are well maintained. The HSV-1 FcγR binds the Fc domain of human, but not murine IgG; therefore, to assess FcγR functions in vivo, mice were passively immunized with human IgG antibody to HSV. When antibody was inoculated intraperitoneally 20 h prior to infection or shortly after virus reached the dorsal root ganglia, disease severity was significantly reduced in mice infected with NS-gE264, but not in mice infected with wild-type or rescue virus. Studies of C3 knockout mice and natural killer cell-depleted mice demonstrated that the HSV-1 FcγR blocked both IgG Fc-mediated complement activation and antibody-dependent cellular cytotoxicity. Therefore, the HSV-1 FcγR promotes immune evasion from IgG Fc-mediated activities and likely contributes to virulence at times when antibody is present, such as during recurrent infections.


Assuntos
Anticorpos Antivirais/imunologia , Citotoxicidade Celular Dependente de Anticorpos , Ativação do Complemento , Herpesvirus Humano 1/imunologia , Herpesvirus Humano 1/patogenicidade , Evasão da Resposta Imune , Proteínas do Envelope Viral/imunologia , Animais , Complemento C3/deficiência , Complemento C3/imunologia , Modelos Animais de Doenças , Herpes Zoster/patologia , Herpes Zoster/virologia , Humanos , Células Matadoras Naturais/imunologia , Procedimentos de Redução de Leucócitos , Camundongos , Camundongos Endogâmicos BALB C , Índice de Gravidade de Doença , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Fatores de Virulência/genética , Fatores de Virulência/imunologia , Fatores de Virulência/metabolismo
7.
Vaccine ; 27(49): 6845-53, 2009 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-19761834

RESUMO

Herpes simplex virus type 1 (HSV-1) glycoprotein C (gC-1) binds complement component C3b and inhibits complement-mediated immunity. HSV-1 glycoprotein D (gD-1) is a potent immunogen and a candidate antigen for a subunit vaccine. We evaluated whether combined immunization with gD-1 and gC-1 provides better protection against challenge than gD-1 alone based on antibodies to gC-1 preventing HSV-1-mediated immune evasion. IgG purified from mice immunized with gC-1 blocked C3b binding to gC-1 and greatly increased neutralization by gD-1 IgG in the presence of complement. Passive transfer of gC-1 IgG protected complement intact mice against HSV-1 challenge but not C3 knockout mice, indicating that gC-1 antibody activity in vivo is complement-dependent. Immunizing mice with gD-1 and gC-1 provided better protection than gD-1 alone in preventing zosteriform disease and infection of dorsal root ganglia. Therefore, gC-1 immunization prevents HSV-1 evasion from complement and enhances the protection provided by gD-1 immunization.


Assuntos
Complemento C3b/imunologia , Vacinas contra o Vírus do Herpes Simples/imunologia , Herpes Simples/prevenção & controle , Evasão da Resposta Imune , Proteínas do Envelope Viral/imunologia , Animais , Anticorpos Antivirais/imunologia , Chlorocebus aethiops , Relação Dose-Resposta Imunológica , Feminino , Gânglios Espinais/imunologia , Herpes Simples/imunologia , Herpesvirus Humano 1/imunologia , Imunização Passiva , Imunoglobulina G/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Testes de Neutralização , Vacinas de Subunidades Antigênicas/imunologia , Células Vero
8.
Arch Virol ; 154(4): 609-18, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19291362

RESUMO

Galectin-3 binds beta-galactoside-containing sugars and is a chemoattractant for monocytes, macrophages, and neutrophils. Galectin-3 was identified by mass spectrometry from an anti-gI affinity column; however, we determined that galectin-3 did not bind gI, but rather that HSV-1 infection increased galectin-3 binding to carbohydrate residues on IgG. Our conclusions are based on the following observations: (1) galectin-3 from cells infected with a gI-deleted HSV-1 mutant virus bound anti-gI IgG; (2) galectin-3 from wild-type HSV-1 infected cells bound nonimmune IgG; (3) more galectin-3 from infected than uninfected cells bound IgG; and (4) binding to IgG was blocked by lactose, a competitive inhibitor of galectin-3 carbohydrate binding. HSV-1 infection did not increase galectin-3 expression, but did increase its secretion. We propose that increased carbohydrate binding and secretion of galectin-3 contribute to an early pro-inflammatory innate immune response to HSV-1 infection.


Assuntos
Metabolismo dos Carboidratos , Galectina 3/metabolismo , Herpes Simples/imunologia , Herpes Simples/virologia , Herpesvirus Humano 1/imunologia , Animais , Linhagem Celular , Galectina 3/imunologia , Humanos , Imunoglobulina G/metabolismo , Ligação Proteica
9.
J Virol ; 82(17): 8431-41, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18562543

RESUMO

Herpes simplex virus type 1 (HSV-1) produces oral lesions, encephalitis, keratitis, and severe infections in the immunocompromised host. HSV-1 is almost as common as HSV-2 in causing first episodes of genital herpes, a disease that is associated with an increased risk of human immunodeficiency virus acquisition and transmission. No approved vaccines are currently available to protect against HSV-1 or HSV-2 infection. We developed a novel HSV vaccine strategy that uses a replication-competent strain of HSV-1, NS-gEnull, which has a defect in anterograde and retrograde directional spread and cell-to-cell spread. Following scratch inoculation on the mouse flank, NS-gEnull replicated at the site of inoculation without causing disease. Importantly, the vaccine strain was not isolated from dorsal root ganglia (DRG). We used the flank model to challenge vaccinated mice and demonstrated that NS-gEnull was highly protective against wild-type HSV-1. The challenge virus replicated to low titers at the site of inoculation; therefore, the vaccine strain did not provide sterilizing immunity. Nevertheless, challenge by HSV-1 or HSV-2 resulted in less-severe disease at the inoculation site, and vaccinated mice were totally protected against zosteriform disease and death. After HSV-1 challenge, latent virus was recovered by DRG explant cocultures from <10% of vaccinated mice compared with 100% of mock-vaccinated mice. The vaccine provided protection against disease and death after intravaginal challenge and markedly lowered the titers of the challenge virus in the vagina. Therefore, the HSV-1 gEnull strain is an excellent candidate for further vaccine development.


Assuntos
Vírus Defeituosos/imunologia , Vacinas contra o Vírus do Herpes Simples/imunologia , Herpesvirus Humano 1/imunologia , Herpesvirus Humano 1/fisiologia , Replicação Viral/fisiologia , Adolescente , Animais , Criança , Feminino , Vacinas contra o Vírus do Herpes Simples/efeitos adversos , Herpesvirus Humano 1/isolamento & purificação , Humanos , Imunização Secundária/métodos , Camundongos , Camundongos Endogâmicos BALB C , Vacinas Atenuadas/imunologia
10.
Vaccine ; 26(9): 1195-203, 2008 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-18243431

RESUMO

HSV-1 glycoprotein D (gD) interacts with HVEM and nectin-1 cell receptors to initiate virus entry. We prepared an HSV-1 strain with mutations in the gD gene at amino acid residues 3 and 38 by changing alanine to cysteine and tyrosine to cysteine, respectively (A3C/Y38C). These mutations were constructed with the intent of evaluating infection in vivo when virus enters by HVEM but not nectin-1 receptors and were based on prior reports demonstrating that purified gDA3C/Y38C protein binds to HVEM but not to nectin-1. While preparing a high-titered purified virus pool, the cysteine mutation at position 38 reverted to tyrosine, which occurred on two separate occasions. The resultant HSV-1 strain, KOS-gDA3C, had a single amino acid mutation at residue 3 and exhibited reduced entry into both HVEM and nectin-1 expressing cells. When tested in the murine flank model, the mutant virus was markedly attenuated for virulence and caused only mild disease, while the parental and rescued viruses produced much more severe disease. Thirty days after KOS-gDA3C infection, mice were challenged with a lethal dose of HSV-1 and were highly resistant to disease. The KOS-gDA3C mutation was stable during 30 passages in vitro and was present in each of 3 isolates obtained from infected mice. Therefore, this gD mutant virus impaired in entry may represent a novel candidate for an attenuated live HSV-1 vaccine.


Assuntos
Vacinas contra o Vírus do Herpes Simples , Herpes Simples/prevenção & controle , Herpesvirus Humano 1/patogenicidade , Mutação , Vacinas Atenuadas , Proteínas do Envelope Viral/genética , Animais , Moléculas de Adesão Celular/metabolismo , Linhagem Celular Tumoral , Chlorocebus aethiops , Feminino , Herpes Simples/imunologia , Herpes Simples/virologia , Vacinas contra o Vírus do Herpes Simples/administração & dosagem , Vacinas contra o Vírus do Herpes Simples/genética , Vacinas contra o Vírus do Herpes Simples/imunologia , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/imunologia , Herpesvirus Humano 1/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Nectinas , Membro 14 de Receptores do Fator de Necrose Tumoral/metabolismo , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Células Vero , Proteínas do Envelope Viral/administração & dosagem , Proteínas do Envelope Viral/imunologia
11.
J Virol ; 80(8): 4038-46, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16571820

RESUMO

Glycoprotein C (gC) of herpes simplex virus type 1 (HSV-1) and type 2 (HSV-2) binds complement component C3b and protects virus from complement-mediated neutralization. Differences in complement interacting domains exist between gC of HSV-1 (gC1) and HSV-2 (gC2), since the amino terminus of gC1 blocks complement C5 from binding to C3b, while gC2 fails to interfere with this activity. We previously reported that neutralization of HSV-1 gC-null virus by HSV antibody-negative human serum requires activation of C5 but not of downstream components of the classical complement pathway. In this report, we evaluated whether activation of C5 is sufficient to neutralize HSV-2 gC-null virus, or whether formation of the membrane attack complex by C6 to C9 is required for neutralization. We found that activation of the classical complement pathway up to C5 was sufficient to neutralize HSV-2 gC-null virus by HSV antibody-negative human serum. We evaluated the mechanisms by which complement activation occurred in seronegative human serum. Interestingly, natural immunoglobulin M antibodies bound to virus, which triggered activation of C1q and the classical complement pathway. HSV antibody-negative sera obtained from four individuals differed over an approximately 10-fold range in their potency for complement-mediated virus neutralization. These findings indicate that humans differ in the ability of their innate immune systems to neutralize HSV-1 or HSV-2 gC-null virus and that a critical function of gC1 and gC2 is to prevent C5 activation.


Assuntos
Anticorpos Antivirais/imunologia , Proteínas do Sistema Complemento/fisiologia , Imunoglobulina M/imunologia , Proteínas do Envelope Viral/fisiologia , Animais , Chlorocebus aethiops , Complemento C1q/fisiologia , Complemento C3/fisiologia , Complemento C5/fisiologia , Complemento C6/fisiologia , Via Alternativa do Complemento , Lectina de Ligação a Manose da Via do Complemento , Testes de Neutralização , Células Vero
12.
Vaccine ; 23(38): 4658-65, 2005 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-15936852

RESUMO

Herpes simplex virus type I (HSV-1) glycoprotein gC (gC-1) is an immune evasion molecule that inhibits complement activation by binding C3b. Three assays were used to assess whether IgG antibodies produced by HSV-1 infection in humans block the interaction between C3b and gC-1. In two assays human IgG had no effect, while in one assay IgG partially inhibited C3b binding, which occurred at IgG concentrations approaching the upper limits of those found in human serum. Mice infected with HSV-1 produced antibodies that partially blocked C3b binding at lower IgG concentrations than human IgG. Importantly, gC-1 immunization in mice produced higher titers of gC-1 antibodies than infection. We previously reported that gC-1 immunization in mice totally blocks C3b binding and reduces disease severity. Therefore, gC-1 immunization in humans may also induce blocking antibodies that modify disease, despite the rather limited ability of infection to produce these antibodies.


Assuntos
Antígenos Virais/imunologia , Infecções por Herpesviridae/prevenção & controle , Simplexvirus/imunologia , Proteínas do Envelope Viral/imunologia , Vacinas Virais/administração & dosagem , Animais , Anticorpos Antivirais/biossíntese , Infecções por Herpesviridae/imunologia , Camundongos , Camundongos Endogâmicos BALB C
13.
J Virol ; 78(5): 2562-71, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14963159

RESUMO

Herpes simplex virus type 1 (HSV-1) glycoprotein gE functions as an immunoglobulin G (IgG) Fc receptor (FcgammaR) that promotes immune evasion. When an IgG antibody binds by the F(ab')(2) domain to an HSV antigen, the Fc domain of some of the same antibody molecules binds to the FcgammaR, which blocks Fc-mediated functions. gE is a type 1 membrane glycoprotein with a large ectodomain that is expressed on the virion envelope and infected-cell surface. Our goal was to determine if immunizing with gE protein fragments could produce antibodies that bind by the F(ab')(2) domain to gE and block the FcgammaR, as measured by competitively inhibiting nonimmune human IgG binding to the FcgammaR. Three gE peptides were constructed in baculovirus spanning almost the entire ectodomain and used to immunize mice and rabbits. Two fragments were highly effective at producing antibodies that bind by the F(ab')(2) domain and block the FcgammaR. The most potent of these two antibodies was far more effective at blocking the FcgammaR than antibodies that are only capable of binding by the Fc domains to the FcgammaR, including anti-gC, anti-gD, and nonimmune IgG. These results suggest that immunizing with gE fragments has potential for preventing immune evasion by blocking activities mediated by the HSV-1 FcgammaR.


Assuntos
Anticorpos Antivirais/imunologia , Herpesvirus Humano 1/imunologia , Receptores de IgG/imunologia , Proteínas do Envelope Viral/imunologia , Animais , Anticorpos Antivirais/farmacologia , Baculoviridae/genética , Ligação Competitiva , Células COS , Citometria de Fluxo , Vacinas contra o Vírus do Herpes Simples/genética , Vacinas contra o Vírus do Herpes Simples/imunologia , Herpesvirus Humano 1/efeitos dos fármacos , Herpesvirus Humano 1/genética , Humanos , Imunização , Imunoglobulina G/imunologia , Camundongos , Coelhos , Receptores de IgG/antagonistas & inibidores , Receptores de IgG/genética , Proteínas do Envelope Viral/antagonistas & inibidores , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/farmacologia
14.
J Virol ; 77(23): 12639-45, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14610186

RESUMO

Many microorganisms encode immune evasion molecules to escape host defenses. Herpes simplex virus type 1 glycoprotein gC is an immunoevasin that inhibits complement activation by binding complement C3b. gC is expressed on the virus envelope and infected cell surface, which makes gC potentially accessible to blocking antibodies. Mice passively immunized with gC monoclonal antibodies prior to infection were protected against herpes simplex virus challenge only if the gC antibodies blocked C3b binding. Mice treated 1 or 2 days postinfection with gC monoclonal antibodies that block C3b binding had less severe disease than control mice treated with nonimmune immunoglobulin G (IgG). Mice immunized with gC protein produced antibodies that blocked C3b binding to gC. Immunized mice were significantly protected against challenge by wild-type virus, but not against a gC mutant virus lacking the C3b binding domain, suggesting that protection was mediated by antibodies that target the gC immune evasion domain. IgG and complement from subjects immunized with an experimental herpes simplex virus glycoprotein gD vaccine neutralized far more mutant virus defective in immune evasion than wild-type virus, supporting the importance of immune evasion molecules in reducing vaccine potency. These results suggest that it is possible to block immune evasion domains on herpes simplex virus and that this approach has therapeutic potential and may enhance vaccine efficacy.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Infecções por Herpesviridae/prevenção & controle , Infecções por Herpesviridae/terapia , Animais , Chlorocebus aethiops , Complemento C3b/imunologia , Complemento C3b/metabolismo , Infecções por Herpesviridae/imunologia , Imunização Passiva , Camundongos , Camundongos Endogâmicos BALB C , Testes de Neutralização , Vacinação , Células Vero
15.
J Virol ; 76(18): 9232-41, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12186907

RESUMO

Herpes simplex virus type 1 (HSV-1) encodes a complement-interacting glycoprotein, gC, and an immunoglobulin G (IgG) Fc binding glycoprotein, gE, that mediate immune evasion by affecting multiple aspects of innate and acquired immunity, including interfering with complement components C1q, C3, C5, and properdin and blocking antibody-dependent cellular cytotoxicity. Previous studies evaluated the individual contributions of gC and gE to immune evasion. Experiments in a murine model that examines the combined effects of gC and gE immune evasion on pathogenesis are now reported. Virulence of wild-type HSV-1 is compared with mutant viruses defective in gC-mediated C3 binding, gE-mediated IgG Fc binding, or both immune evasion activities. Eliminating both activities greatly increased susceptibility of HSV-1 to antibody and complement neutralization in vitro and markedly reduced virulence in vivo as measured by disease scores, virus titers, and mortality. Studies with C3 knockout mice indicated that other activities attributed to these glycoproteins, such as gC-mediated virus attachment to heparan sulfate or gE-mediated cell-to-cell spread, do not account for the reduced virulence of mutant viruses. The results support the importance of gC and gE immune evasion in vivo and suggest potential new targets for prevention and treatment of HSV disease.


Assuntos
Anticorpos Antivirais/imunologia , Proteínas do Sistema Complemento/imunologia , Herpesvirus Humano 1/imunologia , Herpesvirus Humano 1/patogenicidade , Proteínas do Envelope Viral/metabolismo , Animais , Anticorpos Antivirais/metabolismo , Proteínas do Sistema Complemento/metabolismo , Feminino , Herpes Simples/imunologia , Herpes Simples/mortalidade , Herpes Simples/fisiopatologia , Herpes Simples/virologia , Herpesvirus Humano 1/genética , Fragmentos Fc das Imunoglobulinas/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Mutação , Testes de Neutralização , Receptores de IgG/metabolismo , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia , Virulência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA