Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 270
Filtrar
1.
J Virol ; 94(21)2020 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-32796072

RESUMO

Guanylate binding protein 5 (GBP5) belongs to the GTPase subfamily, which is mainly induced by interferon gamma (IFN-γ) and is involved in many important cellular processes, including inflammasome activation and innate immunity against a wide variety of microbial pathogens. However, it is unknown whether GBP5 inhibits respiratory syncytial virus (RSV) infection. In this study, we identified GBP5 as an effector of the anti-RSV activity of IFN-γ and found that in children, the weaker immune response, especially the weaker IFN-γ response and the decreased GBP5 expression, leads to RSV susceptibility. Furthermore, we revealed that GBP5 reduced the cell-associated levels of the RSV small hydrophobic (SH) protein, which was identified as a viroporin. In contrast, overexpression of the SH protein rescued RSV replication in the presence of GBP5. The GBP5-induced decrease in intracellular SH protein levels is because GBP5 promotes the release of the SH protein into the cell culture. Moreover, the GBP5 C583A mutants with changes at the C terminus or the GBP5 ΔC mutant lacking the C-terminal region, which impairs GBP5 localization in the Golgi, could not inhibit RSV infection, whereas the GTPase-defective GBP5 maintained RSV inhibition, suggesting that Golgi localization but not the GTPase activity of GBP5 is required for RSV inhibition. Interestingly, we found that RSV infection or RSV G protein downregulates GBP5 expression by upregulating DZIP3, an E3 ligase, which induces GBP5 degradation through the K48 ubiquitination and proteasomal pathways. Thus, this study reveals a complicated interplay between host restrictive factor GBP5 and RSV infection and provides important information for understanding the pathogenesis of RSV.IMPORTANCE RSV is a highly contagious virus that causes multiple infections in infants within their first year of life. It can also easily cause infection in elderly or immunocompromised individuals, suggesting that individual differences in immunity play an important role in RSV infection. Therefore, exploring the pathogenic mechanisms of RSV and identifying essential genes which inhibit RSV infection are necessary to develop an effective strategy to control RSV infection. Here, we report that the IFN-inducible gene GBP5 potently inhibits RSV replication by reducing the cell-associated levels of the RSV small hydrophobic (SH) protein, which is a viroporin. In contrast, the RSV G protein was shown to upregulate the expression of the DZIP3 protein, an E3 ligase that degrades GBP5 through the proteasomal pathway. Our study provides important information for the understanding of the pathogenic mechanisms of RSV and host immunity as well as the complicated interplay between the virus and host.


Assuntos
Proteínas de Ligação ao GTP/genética , Interações Hospedeiro-Patógeno/genética , Interferon gama/genética , Infecções por Vírus Respiratório Sincicial/genética , Vírus Sincicial Respiratório Humano/genética , Proteínas Oncogênicas de Retroviridae/genética , Adulto , Criança , Células Epiteliais/imunologia , Células Epiteliais/virologia , Feminino , Proteínas de Ligação ao GTP/imunologia , Regulação da Expressão Gênica , Complexo de Golgi/imunologia , Complexo de Golgi/virologia , Interações Hospedeiro-Patógeno/imunologia , Humanos , Imunidade Inata , Inflamassomos/genética , Inflamassomos/imunologia , Interferon gama/imunologia , Masculino , Mutação , Complexo de Endopeptidases do Proteassoma/imunologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/imunologia , Infecções por Vírus Respiratório Sincicial/imunologia , Infecções por Vírus Respiratório Sincicial/patologia , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sincicial Respiratório Humano/crescimento & desenvolvimento , Vírus Sincicial Respiratório Humano/imunologia , Proteínas Oncogênicas de Retroviridae/imunologia , Transdução de Sinais , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/imunologia , Ubiquitinação , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia
2.
Eur J Immunol ; 50(10): 1591-1597, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32470143

RESUMO

Mice bearing CT26 tumors can be cured by administration of L19-mIL12 or F8-mTNF, two antibody fusion proteins which selectively deliver their cytokine payload to the tumor. In both settings, cancer cures crucially depended on CD8+ T cells and the AH1 peptide (derived from the gp70 protein of the murine leukemia virus) acted as the main tumor-rejection antigen, with ∼50% of CD8+ T cells in the neoplastic mass being AH1-specific after therapy. In order to characterize the clonality of the T cell response, its phenotype, and activation status, we isolated CD8+ T cells from tumors and secondary lymphoid organs and submitted them to T cell receptor (TCR) and total mRNA sequencing. We found an extremely diverse repertoire of more than 40 000 unique TCR sequences, but the ten most abundant TCRs accounted for >60% of CD8+ T-cell clones in the tumor. AH1-specific TCRs were consistently found among the most abundant sequences. AH1-specific T cells in the tumor had a tissue-resident memory phenotype. Treatment with L19-mIL12 led to overexpression of IL-12 receptor and of markers of cell activation and proliferation. These data suggest that the antitumor response driven by antibody-cytokine fusions proceeds through an oligoclonal expansion and activation of tumor-infiltrating CD8+ T cells.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Colo/patologia , Neoplasias do Colo/terapia , Imunoterapia/métodos , Vírus da Leucemia Murina/genética , Animais , Antígenos de Neoplasias/imunologia , Linhagem Celular Tumoral , Neoplasias do Colo/imunologia , Modelos Animais de Doenças , Feminino , Humanos , Memória Imunológica , Interleucina-12/uso terapêutico , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C , Peptídeos/genética , Peptídeos/imunologia , Receptores de Antígenos de Linfócitos T/genética , Proteínas Oncogênicas de Retroviridae/genética , Proteínas Oncogênicas de Retroviridae/imunologia , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia
3.
PLoS Pathog ; 15(4): e1007689, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30964929

RESUMO

NOD-like receptor protein 3 (NLRP3) inflammasome activation triggers caspase-1 activation-induced maturation of interleukin (IL)-1ß and IL-18 and therefore is important for the development of the host defense against various RNA viral diseases. However, the implication of this protein complex in human metapneumovirus (HMPV) disease has not been fully studied. Herein, we report that NLRP3 inflammasome plays a detrimental role during HMPV infection because NLRP3 inflammasome inhibition protected mice from mortality and reduced weight loss and inflammation without impacting viral replication. We also demonstrate that NLRP3 inflammasome exerts its deleterious effect via IL-1ß production since we observed reduced mortality, weight loss and inflammation in IL-1ß-deficient (IL-1ß-/-) mice, as compared to wild-type animals during HMPV infection. Moreover, the effect on these evaluated parameters was not different in IL-1ß-/- and wild-type mice treated with an NLRP3 inflammasome inhibitor. The production of IL-1ß was also abrogated in bone marrow derived macrophages deficient for NLRP3. Finally, we show that small hydrophobic protein-deleted recombinant HMPV (HMPV ΔSH) failed to activate caspase-1, which is responsible for IL-1ß cleavage and maturation. Furthermore, HMPV ΔSH-infected mice had less weight loss, showed no mortality and reduced inflammation, as compared to wild-type HMPV-infected mice. Thus, NLRP3 inflammasome activation seems to be triggered by HMPV SH protein in HMPV disease. In summary, once activated by the HMPV SH protein, NLRP3 inflammasome promotes the maturation of IL-1ß, which exacerbates HMPV-induced inflammation. Therefore, the blockade of IL-1ß production by using NLRP3 inflammasome inhibitors might be a novel potential strategy for the therapy and prevention of HMPV infection.


Assuntos
Inflamassomos/imunologia , Inflamação/imunologia , Interleucina-1beta/fisiologia , Metapneumovirus/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/fisiologia , Infecções por Paramyxoviridae/imunologia , Proteínas Oncogênicas de Retroviridae/metabolismo , Animais , Feminino , Humanos , Inflamassomos/metabolismo , Inflamação/virologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Infecções por Paramyxoviridae/virologia , Proteínas Recombinantes/metabolismo , Proteínas Oncogênicas de Retroviridae/imunologia , Transdução de Sinais , Replicação Viral
4.
Aust Vet J ; 97(3): 47-55, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30809813

RESUMO

With the commercial release in Australia in 2004 of a vaccine against feline immunodeficiency virus (FIV; Fel-O-Vax FIV®), the landscape for FIV diagnostics shifted substantially. Point-of-care (PoC) antibody detection kits, which had been the mainstay for diagnosing FIV infection since the early 1990s, were no longer considered accurate to use in FIV-vaccinated cats, because of the production of vaccine-induced antibodies that were considered indistinguishable from those produced in natural FIV infections. Consequently, attention shifted to alternative diagnostic methods such as nucleic acid detection. However, over the past 5 years we have published a series of studies emphasising that FIV PoC test kits vary in their methodology, resulting in differing accuracy in FIV-vaccinated cats. Importantly, we demonstrated that two commercially available FIV antibody test kits (Witness™ and Anigen Rapid™) were able to accurately distinguish between FIV-vaccinated and FIV-infected cats, concluding that testing with either kit offers an alternative to PCR testing. This review summarises pertinent findings from our work published in a variety of peer-reviewed research journals to inform veterinarians (particularly veterinarians in Australia, New Zealand and Japan, where the FIV vaccine is currently commercially available) about how the approach to the diagnosis of FIV infection has shifted. Included in this review is our work investigating the performance of three commercially available FIV PoC test kits in FIV-vaccinated cats and our recommendations for the diagnosis of FIV infection; the effect of primary FIV vaccination (three FIV vaccines, 4 weeks apart) on PoC test kit performance; our recommendations regarding annual testing of FIV-vaccinated cats to detect 'vaccine breakthroughs'; and the potential off-label use of saliva for the diagnosis of FIV infection using some FIV PoC test kits. We also investigated the accuracy of the same three brands of test kits for feline leukaemia virus (FeLV) diagnosis, using both blood and saliva as diagnostic specimens. Based on these results, we discuss our recommendations for confirmatory testing when veterinarians are presented with a positive FeLV PoC test kit result. Finally, we conclude with our results from the largest and most recent FIV and FeLV seroprevalence study conducted in Australia to date.


Assuntos
Síndrome de Imunodeficiência Adquirida Felina/diagnóstico , Vírus da Imunodeficiência Felina/isolamento & purificação , Vírus da Leucemia Felina/isolamento & purificação , Leucemia Felina/diagnóstico , Animais , Anticorpos Antivirais/análise , Anticorpos Antivirais/sangue , Austrália/epidemiologia , Gatos , Síndrome de Imunodeficiência Adquirida Felina/epidemiologia , Síndrome de Imunodeficiência Adquirida Felina/prevenção & controle , Vírus da Imunodeficiência Felina/imunologia , Vírus da Leucemia Felina/imunologia , Leucemia Felina/epidemiologia , Leucemia Felina/prevenção & controle , Sistemas Automatizados de Assistência Junto ao Leito , Reação em Cadeia da Polimerase , Proteínas Oncogênicas de Retroviridae/imunologia , Saliva/virologia , Sensibilidade e Especificidade , Vacinas Virais/imunologia
5.
Viruses ; 10(6)2018 05 23.
Artigo em Inglês | MEDLINE | ID: mdl-29789500

RESUMO

Human metapneumovirus (hMPV), a leading cause of respiratory tract infections in infants, encodes a small hydrophobic (SH) protein of unknown function. Here we show that infection of plasmacytoid dendritic cells (pDCs) with a recombinant virus lacking SH expression (rhMPV-ΔSH) enhanced the secretion of type I interferons (IFNs), which required TLR7 and MyD88 expression. HMPV SH protein inhibited TLR7/MyD88/TRAF6 signaling leading to IFN gene transcription, identifying a novel mechanism by which paramyxovirus SH proteins modulate innate immune responses.


Assuntos
Células Dendríticas/virologia , Interferon Tipo I/metabolismo , Metapneumovirus/genética , Proteínas Oncogênicas de Retroviridae/imunologia , Células Dendríticas/imunologia , Células HEK293 , Humanos , Imunidade Inata , Metapneumovirus/fisiologia , Fator 88 de Diferenciação Mieloide/imunologia , Transdução de Sinais , Receptor 7 Toll-Like/imunologia
6.
Iran J Allergy Asthma Immunol ; 17(2): 144-150, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29757587

RESUMO

We aimed to develope a peptide-based indirect ELISA to detect antibodies against Human T-lymphotropic virus type I (HTLV-I). Two chimeric peptides (CP-1 and CP-2) were designed using linear immunodominant epitopes of gp-46-I, and gp21-I proteins, according to the sequence from Uniprot database. These peptides were studied initially in the ELISA using infected sera. The most promising peptideCP-1, was used to develop a peptide ELISA for detection of HTLV-I infected sera. The optimal conditions for CP-1ELISA were: the optimum coating buffer was 100mM NaHCO3, pH 9.6; coating peptide concentration was 10 µg/mL; the optimal blocking buffer was5% fetal bovine serum (FBS); the secondary antibody concentration was 1:2000; and serum dilution was 1:20. 20serum samples from HTLV-I infected patients were evaluated by ELISA developed. CP-1 showed high antigenicity while lacking any cross-reactivity with normal human sera. The results of evaluations indicated that in comparison with commercial ELISA, CP-1 ELISA showed good sensitivity and specificity. With further validation, CP-1as described in the present study could be introduced as novel reliable and cost-effective candidates for the high-specific screening of HTLV-I/-II infections in endemic regions.


Assuntos
Ensaio de Imunoadsorção Enzimática/métodos , Produtos do Gene env/química , Anticorpos Anti-HTLV-I/sangue , Infecções por HTLV-I/diagnóstico , Vírus Linfotrópico T Tipo 1 Humano/imunologia , Peptídeos/imunologia , Proteínas Oncogênicas de Retroviridae/química , Produtos do Gene env do Vírus da Imunodeficiência Humana/química , Sequência de Aminoácidos , Produtos do Gene env/imunologia , Antígenos HTLV-I/química , Antígenos HTLV-I/imunologia , Infecções por HTLV-I/sangue , Vírus Linfotrópico T Tipo 1 Humano/isolamento & purificação , Humanos , Epitopos Imunodominantes/química , Epitopos Imunodominantes/imunologia , Peptídeos/síntese química , Peptídeos/química , Proteínas Oncogênicas de Retroviridae/imunologia , Sensibilidade e Especificidade , Produtos do Gene env do Vírus da Imunodeficiência Humana/imunologia
7.
J Infect Dis ; 218(3): 378-387, 2018 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-29617814

RESUMO

Background: Respiratory syncytial virus infection can cause lower respiratory tract infection in older adults comparable to influenza, but no vaccines are available. Methods: This was a randomized, observer-blinded, first-in-humans study of a novel synthetic RSV antigen based on the ectodomain of the small hydrophobic glycoprotein (SHe) of RSV subgroup A, formulated with either the lipid and oil-based vaccine platform DepoVax (DPX-RSV[A]) or alum (RSV[A]-Alum), in healthy, 50-64-year-old individuals. Two dose levels (10 or 25 µg) of SHe with each formulation were compared to placebo. A booster dose was administered on day 56. Results: There was no indication that the vaccine was unsafe. Mild pain, drowsiness, and muscles aches were the most common solicited adverse events (AEs), and the frequencies of the AEs did not increase after dose 2. Robust anti-SHe-specific immune responses were demonstrated in the DPX-RSV(A) 10-µg and 25-µg groups (geometric mean titer, approximately 10-fold and 100-fold greater than that of placebo at days 56 and 236, respectively), and responses were sustained in the DPX-RSV(A) 25-µg group at day 421. Responses to the RSV(A)-Alum vaccines were very low. Conclusions: A novel antigen from the SH protein of RSV, formulated in a lipid and oil-based vaccine platform, was highly immunogenic, with sustained antigen-specific antibody responses, and had an acceptable safety profile.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Anticorpos Antivirais/sangue , Lipídeos/administração & dosagem , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vacinas contra Vírus Sincicial Respiratório/imunologia , Vírus Sincicial Respiratório Humano/imunologia , Proteínas Oncogênicas de Retroviridae/imunologia , Compostos de Alúmen/administração & dosagem , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/epidemiologia , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/patologia , Feminino , Voluntários Saudáveis , Humanos , Imunidade Humoral , Esquemas de Imunização , Masculino , Pessoa de Meia-Idade , Placebos/administração & dosagem , Vacinas contra Vírus Sincicial Respiratório/administração & dosagem , Vacinas contra Vírus Sincicial Respiratório/efeitos adversos , Método Simples-Cego , Vacinas de Subunidades/administração & dosagem , Vacinas de Subunidades/efeitos adversos , Vacinas de Subunidades/imunologia
8.
J Gen Virol ; 98(7): 1587-1599, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28714847

RESUMO

Bovine respiratory syncytial virus (BRSV) is an important cause of respiratory disease in young cattle and is closely related to human RSV (HRSV), which causes severe respiratory disease in infants and the elderly. The RSV genome encodes a small hydrophobic (SH) protein with viroporin activity. Previous studies have shown that recombinant BRSV lacking the SH gene (rBRSVΔSH) is attenuated in the lungs, but not in the upper respiratory tract, of calves and mucosal vaccination with rBRSVΔSH induced long-lasting protective immunity. Attenuation of rBRSVΔSH may be due to the ability of this virus to induce an early innate response as rBRSVΔSH induces higher levels of pro-inflammatory cytokines than wild-type (wt) rBRSV. In this study, we investigated the effects of the BRSV SH protein on NF-κB p65 phosphorylation, a master step in the regulation of pro-inflammatory cytokines. Expression of SH resulted in the inhibition of NF-κB p65 phosphorylation in response to BRSV infection and extracellular lipopolysaccharide, and a reduction in the production of pro-inflammatory cytokines. In contrast, rBRSVΔSH does not inhibit NF-κB p65 phosphorylation in bovine antigen-presenting cells, including monocytes, macrophages and dendritic cells, resulting in increased expression of pro-inflammatory cytokines and increased activation of T cells compared to cells infected with wt BRSV. These findings highlight an important role for the BRSV SH protein in immune modulation.


Assuntos
Citocinas/metabolismo , Células Dendríticas/imunologia , Macrófagos/imunologia , Monócitos/imunologia , Vírus Sincicial Respiratório Bovino/metabolismo , Proteínas Oncogênicas de Retroviridae/imunologia , Fator de Transcrição RelA/metabolismo , Animais , Bovinos , Doenças dos Bovinos/imunologia , Doenças dos Bovinos/virologia , Linhagem Celular , Células Dendríticas/metabolismo , Células Dendríticas/virologia , Humanos , Lipopolissacarídeos/metabolismo , Ativação Linfocitária/imunologia , Macrófagos/metabolismo , Macrófagos/virologia , Camundongos , Monócitos/metabolismo , Monócitos/virologia , Inibidor de NF-kappaB alfa/metabolismo , Fosforilação , Células RAW 264.7 , Vírus Sincicial Respiratório Bovino/genética , Vírus Sincicial Respiratório Bovino/imunologia , Proteínas Oncogênicas de Retroviridae/genética , Proteínas Oncogênicas de Retroviridae/metabolismo , Linfócitos T/imunologia
9.
AIDS ; 30(16): 2427-2438, 2016 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-27428745

RESUMO

Vaccination with SIVmac239Δnef provides robust protection against subsequent challenge with wild-type simian immunodeficiency virus (SIV), but safety issues have precluded designing an HIV-1 vaccine based on a live-attenuated virus concept. Safe immunogens and adjuvants that could reproduce identified immune correlates of SIVmac239Δnef protection therefore offer an alternative path for development of an HIV vaccine. Here we describe SIV envelope trimeric gp41 (gp41t) immunogens based on a protective correlate of antibodies to gp41t concentrated on the path of virus entry by the neonatal Fc receptor (FcRn) in cervical vaginal epithelium. We developed a gp41t immunogen-monophosphoryl lipid A adjuvant liposomal nanoparticle for intramuscular (i.m.) immunization and a gp41t-Fc immunogen for intranasal immunization for pilot studies in mice, rabbits, and rhesus macaques. Repeated immunizations to mimic persistent antigen exposure in infection elicited gp41t antibodies in rhesus macaques that were detectable in FcRn+ cervical vaginal epithelium, thus recapitulating one key feature of SIVmac239Δnef vaccinated and protected animals. Although this strategy did not reproduce the system of local production of antibody in SIVmac239Δnef-vaccinated animals, passive immunization experiments supported the concept that sufficiently high levels of antibody can be concentrated by the FcRn at mucosal frontlines, thus setting the stage for assessing protection against vaginal challenge by gp41t immunization.


Assuntos
Anticorpos Antivirais/imunologia , Produtos do Gene env/imunologia , Proteínas Oncogênicas de Retroviridae/imunologia , Vacinas contra a SAIDS/imunologia , Vírus da Imunodeficiência Símia/imunologia , Proteínas Virais de Fusão/imunologia , Adjuvantes Imunológicos/administração & dosagem , Administração Intranasal , Animais , Epitélio/imunologia , Produtos do Gene env/genética , Antígenos de Histocompatibilidade Classe I/imunologia , Imunidade nas Mucosas , Injeções Intramusculares , Lipídeo A/administração & dosagem , Macaca mulatta , Camundongos Endogâmicos BALB C , Coelhos , Receptores Fc/imunologia , Proteínas Oncogênicas de Retroviridae/genética , Vacinas contra a SAIDS/administração & dosagem , Vacinas contra a SAIDS/genética , Vírus da Imunodeficiência Símia/genética , Proteínas Virais de Fusão/genética
10.
Microb Pathog ; 97: 38-44, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27235335

RESUMO

To prevent the spread of HTLV-I (Human T-lymphotropic virus type 1), a safe and effective vaccine is required. To increase immune responses against the peptide antigens can be potentiated with polymer-based nanoparticles, like chitosan (CHT) and trimethylchitosan (TMC), as delivery system/adjuvant. CHT and TMC nanoparticles loaded with recombinant proteins (env23 & env13) of gp46 were prepared by direct coating of antigens with positively charged polymers. The size of CHT and TMC nanoparticles (NPs) loaded with each antigen was about 400 nm. The physical stability of NPs was followed for 4 weeks. Both formulations showed to be stable for about 15 days. The immunogenicity of NPs loaded with antigens was studied after nasal and subcutaneous immunization in mice. Three immunizations (7.5 µg antigen) were performed with 2 weeks intervals. Two weeks after the last booster dose, sera IgG subtypes were measured. After subcutaneous administration, for both nanoparticulate antigens, serum IgG1 and IgGtotal levels were higher than antigen solution (P < 0.001). After nasal administration, for env23, IgG2a levels and IgG2a/IgG1 ratio was significantly higher than groups with subcutaneous administration (P < 0.001). Both nanoparticles showed good immunoadjuvant potential. Env23 antigen was a better candidate for vaccination against HTLV-I, as it induced higher cellular immune responses, compared with env13.


Assuntos
Antígenos Virais/imunologia , Quitosana/administração & dosagem , Produtos do Gene env/imunologia , Vírus Linfotrópico T Tipo 1 Humano/imunologia , Nanopartículas/administração & dosagem , Proteínas Oncogênicas de Retroviridae/imunologia , Vacinas Virais/imunologia , Adjuvantes Imunológicos/administração & dosagem , Administração Intranasal , Animais , Anticorpos Antivirais/sangue , Antígenos Virais/genética , Portadores de Fármacos/administração & dosagem , Estabilidade de Medicamentos , Produtos do Gene env/genética , Vírus Linfotrópico T Tipo 1 Humano/genética , Imunoglobulina G/sangue , Injeções Subcutâneas , Masculino , Camundongos Endogâmicos BALB C , Proteínas Oncogênicas de Retroviridae/genética , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Vacinas Virais/administração & dosagem , Vacinas Virais/genética
11.
Viruses ; 8(2)2016 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-26848684

RESUMO

Although the number of human T-cell leukemia virus type-I (HTLV-I)-infected individuals in the world has been estimated at over 10 million, no prophylaxis vaccines against HTLV-I infection are available. In this study, we took a new approach for establishing the basis of protective vaccines against HTLV-I. We show here the potential of a passively administered HTLV-I neutralizing monoclonal antibody of rat origin (LAT-27) that recognizes epitopes consisting of the HTLV-I gp46 amino acids 191-196. LAT-27 completely blocked HTLV-I infection in vitro at a minimum concentration of 5 µg/mL. Neonatal rats born to mother rats pre-infused with LAT-27 were shown to have acquired a large quantity of LAT-27, and these newborns showed complete resistance against intraperitoneal infection with HTLV-I. On the other hand, when humanized immunodeficient mice were pre-infused intravenously with humanized LAT-27 (hu-LAT-27), all the mice completely resisted HTLV-I infection. These results indicate that hu-LAT-27 may have a potential for passive immunization against both horizontal and mother-to-child vertical infection with HTLV-I.


Assuntos
Anticorpos Monoclonais/imunologia , Transmissão de Doença Infecciosa/prevenção & controle , Produtos do Gene env/imunologia , Infecções por HTLV-I/prevenção & controle , Infecções por HTLV-I/transmissão , Vírus Linfotrópico T Tipo 1 Humano/fisiologia , Transmissão Vertical de Doenças Infecciosas/prevenção & controle , Proteínas Oncogênicas de Retroviridae/imunologia , Adulto , Animais , Anticorpos Monoclonais/administração & dosagem , Feminino , Produtos do Gene env/genética , Infecções por HTLV-I/imunologia , Infecções por HTLV-I/virologia , Vírus Linfotrópico T Tipo 1 Humano/genética , Vírus Linfotrópico T Tipo 1 Humano/imunologia , Humanos , Imunização Passiva , Lactente , Masculino , Camundongos , Ratos , Ratos Endogâmicos F344 , Ratos Sprague-Dawley , Proteínas Oncogênicas de Retroviridae/genética
12.
Artigo em Inglês | MEDLINE | ID: mdl-26459979

RESUMO

This study challenges the commonly held view that the feline immunodeficiency virus (FIV) infection status of FIV-vaccinated cats cannot be determined using point-of-care antibody test kits due to indistinguishable antibody production in FIV-vaccinated and naturally FIV-infected cats. The performance of three commercially available point-of-care antibody test kits was compared in a mixed population of FIV-vaccinated (n=119) and FIV-unvaccinated (n=239) cats in Australia. FIV infection status was assigned by considering the results of all antibody kits in concert with results from a commercially available PCR assay (FIV RealPCR™). Two lateral flow immunochromatography test kits (Witness FeLV/FIV; Anigen Rapid FIV/FeLV) had excellent overall sensitivity (100%; 100%) and specificity (98%; 100%) and could discern the true FIV infection status of cats, irrespective of FIV vaccination history. The lateral flow ELISA test kit (SNAP FIV/FeLV Combo) could not determine if antibodies detected were due to previous FIV vaccination, natural FIV infection, or both. The sensitivity and specificity of FIV RealPCR™ for detection of viral and proviral nucleic acid was 92% and 99%, respectively. These results will potentially change the way veterinary practitioners screen for FIV in jurisdictions where FIV vaccination is practiced, especially in shelter scenarios where the feasibility of mass screening is impacted by the cost of testing.


Assuntos
Anticorpos Antivirais/sangue , Síndrome de Imunodeficiência Adquirida Felina/diagnóstico , Síndrome de Imunodeficiência Adquirida Felina/imunologia , Vírus da Imunodeficiência Felina/imunologia , Proteínas Oncogênicas de Retroviridae/imunologia , Testes Sorológicos/veterinária , Vacinas Virais/imunologia , Animais , Austrália , Gatos , Cromatografia de Afinidade/veterinária , Ensaio de Imunoadsorção Enzimática/veterinária , Síndrome de Imunodeficiência Adquirida Felina/sangue , Sistemas Automatizados de Assistência Junto ao Leito , Reação em Cadeia da Polimerase/veterinária , Kit de Reagentes para Diagnóstico/veterinária , Sensibilidade e Especificidade , Vacinação/veterinária
14.
EMBO Mol Med ; 6(11): 1436-54, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25298406

RESUMO

Infections with human respiratory syncytial virus (HRSV) occur globally in all age groups and can have devastating consequences in young infants. We demonstrate that a vaccine based on the extracellular domain (SHe) of the small hydrophobic (SH) protein of HRSV, reduced viral replication in challenged laboratory mice and in cotton rats. We show that this suppression of viral replication can be transferred by serum and depends on a functional IgG receptor compartment with a major contribution of FcγRI and FcγRIII. Using a conditional cell depletion method, we provide evidence that alveolar macrophages are involved in the protection by SHe-specific antibodies. HRSV-infected cells abundantly express SH on the cell surface and are likely the prime target of the humoral immune response elicited by SHe-based vaccination. Finally, natural infection of humans and experimental infection of mice or cotton rats does not induce a strong immune response against HRSV SHe. Using SHe as a vaccine antigen induces immune protection against HRSV by a mechanism that differs from the natural immune response and from other HRSV vaccination strategies explored to date. Hence, HRSV vaccine candidates that aim at inducing protective neutralizing antibodies or T-cell responses could be complemented with a SHe-based antigen to further improve immune protection.


Assuntos
Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vacinas contra Vírus Sincicial Respiratório/imunologia , Vírus Sincicial Respiratório Humano/imunologia , Proteínas Oncogênicas de Retroviridae/imunologia , Transferência Adotiva , Adulto , Idoso , Animais , Anticorpos Antivirais/sangue , Feminino , Humanos , Lactente , Procedimentos de Redução de Leucócitos , Macrófagos Alveolares/imunologia , Masculino , Camundongos Endogâmicos BALB C , Infecções por Vírus Respiratório Sincicial/imunologia , Vacinas contra Vírus Sincicial Respiratório/administração & dosagem , Vacinas contra Vírus Sincicial Respiratório/isolamento & purificação , Sigmodontinae
15.
Retrovirology ; 11: 74, 2014 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-25163482

RESUMO

BACKGROUND: Human T-cell leukemia virus type 1 (HTLV-1) causes both neoplastic and inflammatory diseases, including adult T-cell leukemia and HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP). Because these life-threatening and disabling diseases are not yet curable, it is important to prevent new HTLV-1 infections. FINDINGS: In this study, we have established a simple humanized mouse model of HTLV-1 infection for evaluating prophylactic and therapeutic interventions. In this model, HTLV-1-negative normal human peripheral blood mononuclear cells (PBMCs) are transplanted directly into the spleens of severely immunodeficient NOD-SCID/γcnull (NOG) mice, together with mitomycin-treated HTLV-1-producing T cells. Using this model, we tested the efficacy of monoclonal antibodies (mAbs) specific to HTLV-1 as well as human IgG isolated from HAM/TSP patients (HAM-IgG) in preventing HTLV-1-infection. One hour before and 24 h after transplantation of the human cells, each antibody sample was inoculated intraperitoneally. On day 14, human PBMCs isolated from the mouse spleens were tested for HTLV-1 infection. Whereas fresh CD4-positive and CD8-positive T cells isolated from untreated mice or mice treated with isotype control mAb, HTLV-1 non-neutralizing mAbs to envelope gp46, gag p19, and normal human IgG were all infected with HTLV-1; the mice treated with either HTLV-1 neutralizing anti-gp46 mAb or HAM-IgG did not become infected. CONCLUSIONS: Our data indicate that the neutralizing function of the antibody, but not the antigen specificity, is essential for preventing the in vivo transmission of HTLV-1. The present animal model will also be useful for the in vivo evaluation of the efficacy of candidate molecules to be used as prophylactic and therapeutic intervention against HTLV-1 infection.


Assuntos
Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Infecções por HTLV-I/prevenção & controle , Vírus Linfotrópico T Tipo 1 Humano/imunologia , Linfócitos T/virologia , Animais , Produtos do Gene env/imunologia , Infecções por HTLV-I/transmissão , Vírus Linfotrópico T Tipo 1 Humano/fisiologia , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Paraparesia Espástica Tropical/imunologia , Proteínas Oncogênicas de Retroviridae/imunologia
16.
J Gen Virol ; 95(Pt 6): 1244-1254, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24700100

RESUMO

Bovine respiratory syncytial virus (BRSV) causes inflammation and obstruction of the small airways, leading to severe respiratory disease in young calves. The virus is closely related to human (H)RSV, a major cause of bronchiolitis and pneumonia in young children. The ability to manipulate the genome of RSV has provided opportunities for the development of stable, live attenuated RSV vaccines. The role of the SH protein in the pathogenesis of BRSV was evaluated in vitro and in vivo using a recombinant (r)BRSV in which the SH gene had been deleted. Infection of bovine epithelial cells and monocytes with rBRSVΔSH, in vitro, resulted in an increase in apoptosis, and higher levels of TNF-α and IL-1ß compared with cells infected with parental, wild-type (WT) rBRSV. Although replication of rBRSVΔSH and WT rBRSV, in vitro, were similar, the replication of rBRSVΔSH was moderately reduced in the lower, but not the upper, respiratory tract of experimentally infected calves. Despite the greater ability of rBRSVΔSH to induce pro-inflammatory cytokines, in vitro, the pulmonary inflammatory response in rBRSVΔSH-infected calves was significantly reduced compared with that in calves inoculated with WT rBRSV, 6 days previously. Virus lacking SH appeared to be as immunogenic and effective in inducing resistance to virulent virus challenge, 6 months later, as the parental rBRSV. These findings suggest that rBRSVΔSH may be an ideal live attenuated virus vaccine candidate, combining safety with a high level of immunogenicity.


Assuntos
Citocinas/biossíntese , Genes Virais , Vírus Sincicial Respiratório Bovino/genética , Vírus Sincicial Respiratório Bovino/imunologia , Animais , Apoptose , Bovinos , Doenças dos Bovinos/imunologia , Doenças dos Bovinos/prevenção & controle , Doenças dos Bovinos/virologia , Deleção de Genes , Humanos , Imunidade nas Mucosas , Mediadores da Inflamação/metabolismo , Interleucina-1beta/biossíntese , Infecções por Vírus Respiratório Sincicial/imunologia , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Infecções por Vírus Respiratório Sincicial/veterinária , Vacinas contra Vírus Sincicial Respiratório/genética , Vacinas contra Vírus Sincicial Respiratório/imunologia , Vírus Sincicial Respiratório Bovino/patogenicidade , Vírus Sincicial Respiratório Humano/imunologia , Vírus Sincicial Respiratório Humano/patogenicidade , Sistema Respiratório/virologia , Proteínas Oncogênicas de Retroviridae/genética , Proteínas Oncogênicas de Retroviridae/imunologia , Fator de Necrose Tumoral alfa/biossíntese , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Virulência/genética , Virulência/imunologia
17.
J Virol ; 88(11): 6453-69, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24672038

RESUMO

UNLABELLED: Human metapneumovirus (HMPV) is a major etiologic agent of respiratory disease worldwide. HMPV reinfections are common in healthy adults and children, suggesting that the protective immune response to HMPV is incomplete and short-lived. We used gene-deletion viruses to evaluate the role of the attachment G and small hydrophobic SH glycoproteins on virus uptake by primary human monocyte-derived dendritic cells (MDDC) in vitro and on subsequent MDDC maturation and activation of autologous T cells. HMPV with deletion of G and SH (ΔSHG) exhibited increased infectivity but had little effect on MDDC maturation. However, MDDC stimulated with ΔSHG induced increased proliferation of autologous Th1-polarized CD4(+) T cells. This effect was independent of virus replication. Increased T cell proliferation was strictly dependent on contact between virus-stimulated MDDC and CD4(+) T cells. Confocal microscopy revealed that deletion of SH and G was associated with an increased number of immunological synapses between memory CD4(+) T cells and virus-stimulated MDDC. Uptake of HMPV by MDDC was found to be primarily by macropinocytosis. Uptake of wild-type (WT) virus was reduced compared to that of ΔSHG, indicative of inhibition by the SH and G glycoproteins. In addition, DC-SIGN-mediated endocytosis provided a minor alternative pathway that depended on SH and/or G and thus operated only for WT. Altogether, our results show that SH and G glycoproteins reduce the ability of HMPV to be internalized by MDDC, resulting in a reduced ability of the HMPV-stimulated MDDC to activate CD4(+) T cells. This study describes a previously unknown mechanism of virus immune evasion. IMPORTANCE: Human metapneumovirus (HMPV) is a major etiologic agent of respiratory disease worldwide. HMPV reinfections are common in healthy adults and children, suggesting that the protective immune response to HMPV is incomplete and short-lived. We found that HMPV attachment G and small hydrophobic SH glycoproteins reduce the ability of HMPV to be internalized by macropinocytosis into human dendritic cells (DC). This results in a reduced ability of the HMPV-stimulated DC to activate Th1-polarized CD4(+) T cells. These results contribute to a better understanding of the nature of incomplete protection against this important human respiratory virus, provide new information on the entry of HMPV into human cells, and describe a new mechanism of virus immune evasion.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Células Dendríticas/virologia , Glicoproteínas/imunologia , Evasão da Resposta Imune/imunologia , Metapneumovirus/imunologia , Pinocitose/imunologia , Proteínas Oncogênicas de Retroviridae/imunologia , Proteínas Virais/imunologia , Análise de Variância , Moléculas de Adesão Celular/imunologia , Células Dendríticas/imunologia , Citometria de Fluxo , Humanos , Sinapses Imunológicas/imunologia , Lectinas Tipo C/imunologia , Ativação Linfocitária/imunologia , Metapneumovirus/genética , Microscopia Confocal , Receptores de Superfície Celular/imunologia , Internalização do Vírus
18.
Monoclon Antib Immunodiagn Immunother ; 33(1): 28-33, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24555933

RESUMO

Phage display has a fundamental role in protein isolation and engineering. Isolated proteins produced with this method can be modified for specific binding and affinity. P24 is the most produced protein during human immune deficiency virus (HIV) replication; especially in the early steps of HIV-1 infection, its evaluation may have diagnostic values. To test the HIV-1 infection, p24 antigen assay appears to be a very promising alternative to RNA assays. In this study, we have generated a recombinant mouse single chain antibody fragment against p24 of the HIV-1 with the use of phage display technology. After isolation of antibody variable-region (V) gene of B cells extracted from the spleen of an immunized mouse, a library of single chain Fv fragments (scFv) was constructed. The library was used in a series of bio-panning processes against recombinant p24 protein expressed from Escherichia coli. The isolated scFv antibody specifically recognizes the HIV-1 capsid protein p24. The affinity constant of the isolated scFv antibody (MF85) was found to be 2×10(-9) M. Our studies showed that the MF85 scFV antibody has similar properties as that of monoclonal antibodies produced by the hybridoma technology.


Assuntos
Técnicas de Visualização da Superfície Celular/métodos , Proteínas Oncogênicas de Retroviridae/imunologia , Anticorpos de Cadeia Única/imunologia , Animais , Especificidade de Anticorpos , Primers do DNA/genética , Infecções por HIV/diagnóstico , Infecções por HIV/patologia , HIV-1/imunologia , Humanos , Camundongos , Biblioteca de Peptídeos , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Anticorpos de Cadeia Única/biossíntese
19.
J Med Virol ; 86(4): 547-57, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24009128

RESUMO

The SH glycoprotein of human metapneumovirus (HMPV) is twice the size of that of human respiratory syncytial virus and possesses a large, hydrophilic luminal domain. The glycoprotein is located on the surface of the virion and of virus infected cells and, if immunogenic, might be expected to play a role in anti-viral immunity. Initial attempts to study anti-SH antibody immunogenicity were thwarted by the instability of the SH gene on passage both in human bronchial epithelial cells and in mice. Repeated passage of virus isolates in human bronchial epithelial cells in culture resulted in the appearance and eventual predominance of HMPV mutants lacking all or most of the luminal domain of SH coincidental with the loss of productive infection in mouse lungs. Where infection was established in mice with an early cell culture passage, the virus recovered from mouse lung differed markedly from the inoculum, carrying 19 coding mutations in the SH luminal domain. Immunization of mice with a mutant virus variant expressing only 14 amino acids of the luminal domain of SH induced a cross-reactive antibody response to both the F glycoprotein and the SH glycoprotein but a largely sub-group specific response to the G glycoprotein. Similar patterns of response were achieved by immunization with individual HMPV glycoproteins expressed from recombinant vaccinia viruses. Recombinant truncated SH glycoprotein induced sub-group cross-reactive antibodies capable of neutralizing wild-type virus. Recombinant F glycoprotein also induced cross-reactive neutralizing antibodies whilst recombinant G glycoprotein induced largely strain-specific, non-neutralizing antibodies.


Assuntos
Anticorpos Antivirais/imunologia , Glicoproteínas/imunologia , Metapneumovirus/imunologia , Infecções por Paramyxoviridae/imunologia , Proteínas Virais/imunologia , Animais , Anticorpos Neutralizantes/imunologia , Sequência de Bases , Linhagem Celular , Variação Genética , Glicoproteínas/genética , Células HeLa , Humanos , Imunização , Camundongos , Testes de Neutralização , Proteínas Oncogênicas de Retroviridae/genética , Proteínas Oncogênicas de Retroviridae/imunologia , Análise de Sequência de RNA , Proteínas Virais/genética , Vacinas Virais/imunologia
20.
J Virol ; 88(2): 992-1001, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24198407

RESUMO

We previously delineated a highly conserved immunosuppressive (IS) domain within murine and primate retroviral envelope proteins that is critical for virus propagation in vivo. The envelope-mediated immunosuppression was assessed by the ability of the proteins, when expressed by allogeneic tumor cells normally rejected by engrafted mice, to allow these cells to escape, at least transiently, immune rejection. Using this approach, we identified key residues whose mutation (i) specifically abolishes immunosuppressive activity without affecting the "mechanical" function of the envelope protein and (ii) significantly enhances humoral and cellular immune responses elicited against the virus. The objective of this work was to study the immunosuppressive activity of the envelope protein (p15E) of feline leukemia virus (FeLV) and evaluate the effect of its abolition on the efficacy of a vaccine against FeLV. Here we demonstrate that the FeLV envelope protein is immunosuppressive in vivo and that this immunosuppressive activity can be "switched off" by targeted mutation of a specific amino acid. As a result of the introduction of the mutated envelope sequence into a previously well characterized canarypox virus-vectored vaccine (ALVAC-FeLV), the frequency of vaccine-induced FeLV-specific gamma interferon (IFN-γ)-producing cells was increased, whereas conversely, the frequency of vaccine-induced FeLV-specific interleukin-10 (IL-10)-producing cells was reduced. This shift in the IFN-γ/IL-10 response was associated with a higher efficacy of ALVAC-FeLV against FeLV infection. This study demonstrates that FeLV p15E is immunosuppressive in vivo, that the immunosuppressive domain of p15E can modulate the FeLV-specific immune response, and that the efficacy of FeLV vaccines can be enhanced by inhibiting the immunosuppressive activity of the IS domain through an appropriate mutation.


Assuntos
Vírus da Varíola dos Canários/genética , Produtos do Gene env/química , Produtos do Gene env/imunologia , Imunossupressores/química , Vírus da Leucemia Felina/genética , Leucemia Felina/imunologia , Mutação de Sentido Incorreto , Proteínas Oncogênicas de Retroviridae/genética , Vacinas Virais/genética , Animais , Vírus da Varíola dos Canários/metabolismo , Gatos , Feminino , Produtos do Gene env/administração & dosagem , Produtos do Gene env/genética , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Imunossupressores/administração & dosagem , Imunossupressores/imunologia , Interferons/genética , Interferons/imunologia , Interleucina-10/genética , Interleucina-10/imunologia , Vírus da Leucemia Felina/química , Vírus da Leucemia Felina/imunologia , Leucemia Felina/prevenção & controle , Leucemia Felina/virologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Estrutura Terciária de Proteína , Proteínas Oncogênicas de Retroviridae/administração & dosagem , Proteínas Oncogênicas de Retroviridae/química , Proteínas Oncogênicas de Retroviridae/imunologia , Vacinas Virais/administração & dosagem , Vacinas Virais/química , Vacinas Virais/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...