Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 282
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 118(43)2021 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-34686605

RESUMO

Self-amplifying RNA replicons are promising platforms for vaccine generation. Their defects in one or more essential functions for viral replication, particle assembly, or dissemination make them highly safe as vaccines. We previously showed that the deletion of the envelope (E) gene from the Middle East respiratory syndrome coronavirus (MERS-CoV) produces a replication-competent propagation-defective RNA replicon (MERS-CoV-ΔE). Evaluation of this replicon in mice expressing human dipeptidyl peptidase 4, the virus receptor, showed that the single deletion of the E gene generated an attenuated mutant. The combined deletion of the E gene with accessory open reading frames (ORFs) 3, 4a, 4b, and 5 resulted in a highly attenuated propagation-defective RNA replicon (MERS-CoV-Δ[3,4a,4b,5,E]). This RNA replicon induced sterilizing immunity in mice after challenge with a lethal dose of a virulent MERS-CoV, as no histopathological damage or infectious virus was detected in the lungs of challenged mice. The four mutants lacking the E gene were genetically stable, did not recombine with the E gene provided in trans during their passage in cell culture, and showed a propagation-defective phenotype in vivo. In addition, immunization with MERS-CoV-Δ[3,4a,4b,5,E] induced significant levels of neutralizing antibodies, indicating that MERS-CoV RNA replicons are highly safe and promising vaccine candidates.


Assuntos
Infecções por Coronavirus/prevenção & controle , Coronavírus da Síndrome Respiratória do Oriente Médio/genética , Coronavírus da Síndrome Respiratória do Oriente Médio/imunologia , RNA Viral/administração & dosagem , Replicon , Vacinas Virais/administração & dosagem , Animais , Anticorpos Neutralizantes/biossíntese , Anticorpos Antivirais/biossíntese , Infecções por Coronavirus/genética , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/virologia , Vírus Defeituosos/genética , Vírus Defeituosos/imunologia , Feminino , Deleção de Genes , Genes env , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Coronavírus da Síndrome Respiratória do Oriente Médio/patogenicidade , RNA Viral/genética , RNA Viral/imunologia , Vacinas de DNA , Vacinas de Partículas Semelhantes a Vírus/administração & dosagem , Vacinas de Partículas Semelhantes a Vírus/genética , Vacinas de Partículas Semelhantes a Vírus/imunologia , Vacinas Virais/genética , Vacinas Virais/imunologia , Virulência/genética , Virulência/imunologia
2.
J Virol ; 93(21)2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31413132

RESUMO

Vaccines aimed at inducing T cell responses to protect against human immunodeficiency virus (HIV) infection have been under development for more than 15 years. Replication-defective adenovirus (rAd) vaccine vectors are at the forefront of this work and have been tested extensively in the simian immunodeficiency virus (SIV) challenge macaque model. Vaccination with rAd vectors coding for SIV Gag or other nonenvelope proteins induces T cell responses that control virus load but disappointingly is unsuccessful so far in preventing infection, and attention has turned to inducing antibodies to the envelope. However, here we report that Mauritian cynomolgus macaques (MCM), Macaca fascicularis, vaccinated with unmodified SIV gag alone in a DNA prime followed by an rAd boost exhibit increased protection from infection by repeated intrarectal challenge with low-dose SIVmac251. There was no evidence of infection followed by eradication. A significant correlation was observed between cytokine expression by CD4 T cells and delayed infection. Vaccination with gag fused to the ubiquitin gene or fragmented, designed to increase CD8 magnitude and breadth, did not confer resistance to challenge or enhance immunity. On infection, a significant reduction in peak virus load was observed in all vaccinated animals, including those vaccinated with modified gag These findings suggest that a nonpersistent viral vector vaccine coding for internal virus proteins may be able to protect against HIV type 1 (HIV-1) infection. The mechanisms are probably distinct from those of antibody-mediated virus neutralization or cytotoxic CD8 cell killing of virus-infected cells and may be mediated in part by CD4 T cells.IMPORTANCE The simian immunodeficiency virus (SIV) macaque model represents the best animal model for testing new human immunodeficiency virus type 1 (HIV-1) vaccines. Previous studies employing replication-defective adenovirus (rAd) vectors that transiently express SIV internal proteins induced T cell responses that controlled virus load but did not protect against virus challenge. However, we show for the first time that SIV gag delivered in a DNA prime followed by a boost with an rAd vector confers resistance to SIV intrarectal challenge. Other partially successful SIV/HIV-1 protective vaccines induce antibody to the envelope and neutralize the virus or mediate antibody-dependent cytotoxicity. Induction of CD8 T cells which do not prevent initial infection but eradicate infected cells before infection becomes established has also shown some success. In contrast, the vaccine described here mediates resistance by a different mechanism from that described above, which may reflect CD4 T cell activity. This could indicate an alternative approach for HIV-1 vaccine development.


Assuntos
Produtos do Gene gag/imunologia , Vacinas contra a SAIDS/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/prevenção & controle , Vírus da Imunodeficiência Símia/imunologia , Adenoviridae/genética , Adenoviridae/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Vírus Defeituosos/genética , Vírus Defeituosos/imunologia , Produtos do Gene gag/genética , Vetores Genéticos/genética , Vetores Genéticos/imunologia , Macaca fascicularis , Masculino , Vacinas contra a SAIDS/administração & dosagem , Vacinas contra a SAIDS/genética , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/genética , Vacinação , Vacinas de DNA/administração & dosagem , Vacinas de DNA/genética , Vacinas de DNA/imunologia , Carga Viral
3.
J Virol ; 92(5)2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29237838

RESUMO

One of the first defenses against infecting pathogens is the innate immune system activated by cellular recognition of pathogen-associated molecular patterns (PAMPs). Although virus-derived RNA species, especially copyback (cb)-type defective interfering (DI) genomes, have been shown to serve as real PAMPs, which strongly induce interferon-beta (IFN-ß) during mononegavirus infection, the mechanisms underlying DI generation remain unclear. Here, for the first time, we identified a single amino acid substitution causing production of cbDI genomes by successful isolation of two distinct types of viral clones with cbDI-producing and cbDI-nonproducing phenotypes from the stock Sendai virus (SeV) strain Cantell, which has been widely used in a number of studies on antiviral innate immunity as a representative IFN-ß-inducing virus. IFN-ß induction was totally dependent on the presence of a significant amount of cbDI genome-containing viral particles (DI particles) in the viral stock, but not on deficiency of the IFN-antagonistic viral accessory proteins C and V. Comparison of the isolates indicated that a single amino acid substitution found within the N protein of the cbDI-producing clone was enough to cause the emergence of DI genomes. The mutated N protein of the cbDI-producing clone resulted in a lower density of nucleocapsids than that of the DI-nonproducing clone, probably causing both production of the DI genomes and their formation of a stem-loop structure, which serves as an ideal ligand for RIG-I. These results suggested that the integrity of mononegaviral nucleocapsids might be a critical factor in avoiding the undesirable recognition of infection by host cells.IMPORTANCE The type I interferon (IFN) system is a pivotal defense against infecting RNA viruses that is activated by sensing viral RNA species. RIG-I is a major sensor for infection with most mononegaviruses, and copyback (cb)-type defective interfering (DI) genomes have been shown to serve as strong RIG-I ligands in real infections. However, the mechanism underlying production of cbDI genomes remains unclear, although DI genomes emerge as the result of an error during viral replication with high doses of viruses. Sendai virus has been extensively studied and is unique in that its interaction with innate immunity reveals opposing characteristics, such as high-level IFN-ß induction and strong inhibition of type I IFN pathways. Our findings provide novel insights into the mechanism of production of mononegaviral cbDI genomes, as well as virus-host interactions during innate immunity.


Assuntos
Substituição de Aminoácidos/imunologia , Vírus Defeituosos/genética , Interferon beta/metabolismo , Nucleoproteínas/imunologia , Paramyxovirinae/genética , Paramyxovirinae/imunologia , Vírus Sendai/genética , Substituição de Aminoácidos/genética , Animais , Linhagem Celular , Proteína DEAD-box 58 , Vírus Defeituosos/imunologia , Feminino , Regulação da Expressão Gênica , Genoma Viral , Células HeLa , Interações Hospedeiro-Parasita/genética , Interações Hospedeiro-Parasita/imunologia , Humanos , Imunidade Inata , Fator Regulador 3 de Interferon/análise , Interferon Tipo I/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Nucleocapsídeo/metabolismo , Nucleoproteínas/genética , Infecções por Paramyxoviridae/imunologia , Infecções por Paramyxoviridae/virologia , Moléculas com Motivos Associados a Patógenos/imunologia , Moléculas com Motivos Associados a Patógenos/metabolismo , RNA Viral/genética , Receptores Imunológicos , Vírus Sendai/imunologia , Replicação Viral
4.
Biologicals ; 62: 57-64, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31588012

RESUMO

The present investigation deals with the characterization of defective interfering (DI) particles of Peste-des-petits ruminants (PPR) vaccine Sungri/96 strain generated as a result of high MOI in Vero cells. During the serial 10 passages, infectivity titres drastically reduced from 6.5 to 2.25 log10TCID50/ml at high MOI. Further, attenuation of CPE with high MOI indicated generation of DI particles that resulted in no/slow progression of CPE during the late passages. Monoclonal antibody based cell ELISA indicated normal protein (N & H) packaging in samples with DI activity. At genomic level, inconsistency in amplicon intensity of H gene was observed in RT-PCR, indicating a possible defect of H gene. Further analysis of copy number of PPRV by RT-qPCR indicated intermittent fluctuations of viral genomic RNA copies. The significant decline of viral RNA copies with MOI 3 (314 copies) compared to low MOI (512804 copies), proved that higher DI multiplicities cause more interference with the replication process of the standard virus. Therefore, MOI is critical for manufacturing of vaccines. These investigations will help in upscaling of PPR vaccines in view of ongoing National and Global PPR control and eradication programme.


Assuntos
Vírus Defeituosos , Genoma Viral , Vírus da Peste dos Pequenos Ruminantes , RNA Viral , Vacinas Virais , Animais , Anticorpos Monoclonais Murinos/imunologia , Anticorpos Antivirais/imunologia , Chlorocebus aethiops , Vírus Defeituosos/genética , Vírus Defeituosos/imunologia , Vírus da Peste dos Pequenos Ruminantes/genética , Vírus da Peste dos Pequenos Ruminantes/crescimento & desenvolvimento , RNA Viral/genética , RNA Viral/imunologia , Células Vero , Vacinas Virais/genética , Vacinas Virais/imunologia
5.
J Virol ; 90(16): 7405-7414, 2016 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-27279609

RESUMO

UNLABELLED: African horse sickness virus (AHSV), an orbivirus in the Reoviridae family with nine different serotypes, causes devastating disease in equids. The virion particle is composed of seven proteins organized in three concentric layers, an outer layer made of VP2 and VP5, a middle layer made of VP7, and inner layer made of VP3 that encloses a replicase complex of VP1, VP4, and VP6 and a genome of 10 double-stranded RNA segments. In this study, we sought to develop highly efficacious candidate vaccines against all AHSV serotypes, taking into account not only immunogenic and safety properties but also virus productivity and stability parameters, which are essential criteria for vaccine candidates. To achieve this goal, we first established a highly efficient reverse genetics (RG) system for AHSV serotype 1 (AHSV1) and, subsequently, a VP6-defective AHSV1 strain in combination with in trans complementation of VP6. This was then used to generate defective particles of all nine serotypes, which required the exchange of two to five RNA segments to achieve equivalent titers of particles. All reassortant-defective viruses could be amplified and propagated to high titers in cells complemented with VP6 but were totally incompetent in any other cells. Furthermore, these replication-incompetent AHSV particles were demonstrated to be highly protective against homologous virulent virus challenges in type I interferon receptor (IFNAR)-knockout mice. Thus, these defective viruses have the potential to be used for the development of safe and stable vaccine candidates. The RG system also provides a powerful tool for the study of the role of individual AHSV proteins in virus assembly, morphogenesis, and pathogenesis. IMPORTANCE: African horse sickness virus is transmitted by biting midges and causes African horse sickness in equids, with mortality reaching up to 95% in naive horses. Therefore, the development of efficient vaccines is extremely important due to major economic losses in the equine industry. Through the establishment of a highly efficient RG system, replication-deficient viruses of all nine AHSV serotypes were generated. These defective viruses achieved high titers in a cell line complemented with VP6 but failed to propagate in wild-type mammalian or insect cells. Importantly, these candidate vaccine strains showed strong protective efficacy against AHSV infection in an IFNAR(-/-) mouse model.


Assuntos
Vírus da Doença Equina Africana/imunologia , Doença Equina Africana/prevenção & controle , Vírus Defeituosos/imunologia , Vacinas Virais/metabolismo , Vírion/metabolismo , Montagem de Vírus , Replicação Viral , Vírus da Doença Equina Africana/genética , Vírus da Doença Equina Africana/fisiologia , Animais , Vírus Defeituosos/genética , Vírus Defeituosos/fisiologia , Modelos Animais de Doenças , Deleção de Genes , Camundongos , Camundongos Knockout , Genética Reversa , Sorogrupo , Vacinas Virais/administração & dosagem , Vacinas Virais/imunologia
6.
J Virol ; 90(3): 1557-68, 2016 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-26608320

RESUMO

UNLABELLED: The live attenuated measles virus vaccine is highly immunostimulatory. Identification and characterization of its components that activate the innate immune response might provide new strategies and agents for the rational design and development of chemically defined adjuvants. In this study, we report on the activation of type I interferon (IFN) production by a defective interfering (DI) RNA isolated from the Hu-191 vaccine strain of measles virus. We found that the Hu-191 virus induced IFN-ß much more potently than the Edmonston strain. In the search for IFN-inducing species in Hu-191, we identified a DI RNA specifically expressed by this strain. This DI RNA, which was of the copy-back type, was predicted to fold into a hairpin structure with a long double-stranded stem region of 206 bp, and it potently induced the expression of IFN-ß. Its IFN-ß-inducing activity was further enhanced when both cytoplasmic RNA sensor RIG-I and its partner, PACT, were overexpressed. On the contrary, this activity was abrogated in cells deficient in PACT or RIG-I. The DI RNA was found to be associated with PACT in infected cells. In addition, both the 5'-di/triphosphate end and the double-stranded stem region on the DI RNA were essential for its activation of PACT and RIG-I. Taken together, our findings support a model in which a viral DI RNA is sensed by PACT and RIG-I to initiate an innate antiviral response. Our work might also provide a foundation for identifying physiological PACT ligands and developing novel adjuvants or antivirals. IMPORTANCE: The live attenuated measles virus vaccine is one of the most successful human vaccines and has largely contained the devastating impact of a highly contagious virus. Identifying the components in this vaccine that stimulate the host immune response and understanding their mechanism of action might help to design and develop better adjuvants, vaccines, antivirals, and immunotherapeutic agents. We identified and characterized a defective interfering RNA from the Hu-191 vaccine strain of measles virus which has safely been used in millions of people for many years. We further demonstrated that this RNA potently induces an antiviral immune response through cellular sensors of viral RNA known as PACT and RIG-I. Similar types of viral RNA that bind with and activate PACT and RIG-I might retain the immunostimulatory property of measles virus vaccines but would not induce adaptive immunity. They are potentially useful as chemically defined vaccine adjuvants, antivirals, and immunostimulatory agents.


Assuntos
RNA Helicases DEAD-box/metabolismo , Vírus Defeituosos/imunologia , Interferon beta/biossíntese , Vacina contra Sarampo/imunologia , Vírus do Sarampo/imunologia , RNA Viral/genética , Proteínas de Ligação a RNA/metabolismo , Linhagem Celular , Proteína DEAD-box 58 , Vírus Defeituosos/genética , Humanos , Vacina contra Sarampo/genética , Vírus do Sarampo/genética , Modelos Moleculares , Conformação de Ácido Nucleico , RNA Viral/química , Receptores Imunológicos
7.
Virol J ; 14(1): 138, 2017 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-28738877

RESUMO

BACKGROUND: Defective interfering (DI) viruses are natural antivirals made by nearly all viruses. They have a highly deleted genome (thus being non-infectious) and interfere with the replication of genetically related infectious viruses. We have produced the first potential therapeutic DI virus for the clinic by cloning an influenza A DI RNA (1/244) which was derived naturally from genome segment 1. This is highly effective in vivo, and has unexpectedly broad-spectrum activity with two different modes of action: inhibiting influenza A viruses through RNA interference, and all other (interferon-sensitive) respiratory viruses through stimulating interferon type I. RESULTS: We have investigated the RNA inhibitory mechanism(s) of DI 1/244 RNA. Ablation of initiation codons does not diminish interference showing that no protein product is required for protection. Further analysis indicated that 1/244 DI RNA interferes by replacing the cognate full-length segment 1 RNA in progeny virions, while interfering with the expression of genome segment 1, its cognate RNA, and genome RNAs 2 and 3, but not genome RNA 6, a representative of the non-polymerase genes. CONCLUSIONS: Our data contradict the dogma that a DI RNA only interferes with expression from its cognate full-length segment. There is reciprocity as cloned segment 2 and 3 DI RNAs inhibited expression of RNAs from a segment 1 target. These data demonstrate an unexpected complexity in the mechanism of interference by this cloned therapeutic DI RNA.


Assuntos
Vírus Defeituosos/genética , Vírus Defeituosos/isolamento & purificação , Vírus da Influenza A/crescimento & desenvolvimento , Vírus da Influenza A/genética , RNA Viral/genética , RNA Viral/metabolismo , Vírus Defeituosos/imunologia , Células HEK293 , Humanos , Interferon Tipo I/metabolismo , Interferência de RNA
8.
J Virol ; 88(5): 2658-69, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24352454

RESUMO

UNLABELLED: Cells that undergo apoptosis in response to chemical or physical stimuli repress inflammatory reactions, but cells that undergo nonapoptotic death in response to such stimuli lack this activity. Whether cells dying from viral infection exhibit a cell death-type modulatory effect on inflammatory reactions is unknown. We compared the effects on macrophage inflammatory responses of cells dying an apoptotic or a nonapoptotic death as a result of adenoviral infection. The results were exactly opposite to the predictions from the conventional paradigm. Cells dying by apoptosis induced by infection with an adenovirus type 5 (Ad5) E1B 19-kilodalton (E1B 19K) gene deletion mutant did not repress macrophage NF-κB activation or cytokine responses to proinflammatory stimuli, whereas cells dying a nonapoptotic death from infection with E1B 19K-competent, wild-type Ad5 repressed these macrophage inflammatory responses as well as cells undergoing classical apoptosis in response to chemical injury. The immunorepressive, E1B 19K-related cell death activity depended upon direct contact of the virally infected corpses with responder macrophages. Replacement of the viral E1B 19K gene with the mammalian Bcl-2 gene in cis restored the nonapoptotic, immunorepressive cell death activity of virally infected cells. These results define a novel function of the antiapoptotic, adenoviral E1B 19K protein that may limit local host innate immune inflammation during accumulation of virally infected cells at sites of infection and suggest that E1B 19K-deleted, replicating adenoviral vectors might induce greater inflammatory responses to virally infected cells than E1B 19K-positive vectors, because of the net effect of their loss-of-function mutation. IMPORTANCE: We observed that cells dying a nonapoptotic cell death induced by adenovirus infection repressed macrophage proinflammatory responses while cells dying by apoptosis induced by infection with an E1B 19K deletion mutant virus did not repress macrophage proinflammatory responses and enhanced some cytokine responses. Our results define a new function of the antiapoptotic, adenoviral protein E1B 19K, which we have termed "apoptotic mimicry." Our studies suggest the possibility that the presence or absence of this E1B 19K function could alter the immunological outcome of both natural and therapeutic adenoviral infections. For example, emerging, highly immunopathogenic adenovirus serotypes might induce increased host inflammatory responses as a result of altered E1B 19K function or expression. It is also possible that engineered variations in E1B 19K expression/function could be created during adenovirus vector design that would increase the therapeutic efficacy of replicating adenovirus vectors for vaccines or oncolytic viral targeting of neoplastic cells.


Assuntos
Infecções por Adenoviridae/imunologia , Adenoviridae/imunologia , Proteínas E1B de Adenovirus/imunologia , Apoptose/imunologia , Imunidade Inata , Adenoviridae/genética , Infecções por Adenoviridae/genética , Infecções por Adenoviridae/metabolismo , Proteínas E1B de Adenovirus/genética , Proteínas E1B de Adenovirus/metabolismo , Animais , Comunicação Celular/imunologia , Morte Celular/imunologia , Linhagem Celular , Efeito Citopatogênico Viral , Vírus Defeituosos/genética , Vírus Defeituosos/imunologia , Ativação Enzimática , Expressão Gênica , Humanos , Mediadores da Inflamação/metabolismo , Pulmão/imunologia , Pulmão/virologia , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , NF-kappa B/metabolismo , Fenótipo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Deleção de Sequência , Ativação Transcricional
9.
J Virol ; 87(4): 2011-22, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23236073

RESUMO

Virus-like particles (VLPs) from hepatitis B and human papillomaviruses have been successfully used as preventative vaccines against these infectious agents. These VLPs consist of a self-associating capsid polymer formed from a single structure protein and are devoid of viral DNA. Since virions from herpesviruses consist of a large number of molecules of viral and cellular origin, generating VLPs from a subset of these would be a particularly arduous task. Therefore, we have adopted an alternative strategy that consists of producing DNA-free defective virus particles in a cell line infected by a herpesvirus mutant incapable of packaging DNA. We previously reported that an Epstein-Barr virus (EBV) mutant devoid of the terminal repeats (ΔTR) that act as packaging signals in herpesviruses produces substantial amounts of VLPs and of light particles (LPs). However, ΔTR virions retained some infectious genomes, and although these mutants had lost their transforming abilities, this poses potential concerns for clinical applications. Therefore, we have constructed a series of mutants that lack proteins involved in maturation and assessed their ability to produce viral DNA-free VLP/LPs. Some of the introduced mutations were deleterious for capsid maturation and virus production. However, deletion of BFLF1/BFRF1A or of BBRF1 resulted in the production of DNA-free VLPs/LPs. The ΔBFLF1/BFRF1A viruses elicited a potent CD4(+) T-cell response that was indistinguishable from the one obtained with wild-type controls. In summary, the defective particles produced by the ΔBFLF1/BFRF1A mutant fulfill the criteria of efficacy and safety expected from a preventative vaccine.


Assuntos
DNA Viral/genética , Vírus Defeituosos/genética , Vírus Defeituosos/imunologia , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/imunologia , Vacinas de Partículas Semelhantes a Vírus/imunologia , Linfócitos T CD4-Positivos/imunologia , Células Cultivadas , Deleção de Genes , Herpesvirus Humano 4/fisiologia , Humanos , Proteínas de Membrana/deficiência , Vacinas de Partículas Semelhantes a Vírus/genética , Proteínas Virais , Montagem de Vírus
10.
Mol Ther ; 21(9): 1787-95, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23774793

RESUMO

The induction of a robust neutralizing antibody (nAb) response is likely to be as essential as specific cell-mediated immunity (CMI) against multiple antigens for the development of effective preventive and therapeutic vaccines against hepatitis C virus (HCV) infection in humans. To date, no data on the immunogenicity of the replication-defective vaccinia virus (derived from the Tiantan strain) (rNTV)-based HCV vaccine in primates have been reported. This study describes in detail the immunogenicity of various vaccine candidates in rhesus macaques, including rNTV-based and replication-defective recombinant adenoviral (rAd)-based HCV vaccines, as well as HCV pseudotyped virus-like particles (HCVpp). Our data showed that rAd-HCV vaccine boosting induced robust CMI, while priming or boosting with HCVpp enhanced the antigen-specific nAb response after rAd-HCV vaccination; however, CMI was not enhanced. Vaccination includes rNTV-HCV priming induced robust antigen-specific antibody, particularly nAbs, and CMI responses. Furthermore, more robust and longer-lasting CMI and higher cytokine levels (both Th1 and Th2 types, especially IFN-γ) resulted from boosting with rAd-HCV. We conclude that the rNTV-based HCV vaccine induces robust nAbs and CMI when combined with a heterogeneous primer-booster strategy, which shows promise for development of a human HCV vaccine.


Assuntos
Citocinas/sangue , Hepacivirus/imunologia , Anticorpos Anti-Hepatite C/sangue , Linfócitos T/imunologia , Vaccinia virus/imunologia , Vacinas contra Hepatite Viral/imunologia , Adenoviridae , Animais , Anticorpos Neutralizantes/sangue , Vírus Defeituosos/genética , Vírus Defeituosos/imunologia , Feminino , Vetores Genéticos , Imunidade Celular , Imunização Secundária , Macaca/imunologia , Masculino , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Vacinas de Partículas Semelhantes a Vírus/imunologia , Vaccinia virus/genética , Vaccinia virus/fisiologia , Vacinas contra Hepatite Viral/genética , Replicação Viral
11.
Zhonghua Zhong Liu Za Zhi ; 36(3): 177-82, 2014 Mar.
Artigo em Zh | MEDLINE | ID: mdl-24785276

RESUMO

OBJECTIVE: To explore the anti-tumor effect and its mechanism of Sendai virus Tianjin strain defective interfering particles (DIP) on mouse models of colon carcinoma. METHODS: CT26 cells (5×10(6)/0.1 ml) were subcutaneously injected into the back of Bal B/c mice to establish murine colon carcinoma model. After the tumors reached 5 mm in diameter, the mice were randomly divided into Tianjin strain DIP group and saline control group. The former was intratumorally injected with Tianjin strain DIP (0.1 ml) once a day on day 4, 7, 10 and 13 after CT26 cell inoculation. The latter was intratumorally injected with the same volume of saline. Tumor volume and survival rate of the mice were calculated to confirm the anti-tumor effect of DIP. Flow cytometry and ELISA were used to examine the maturation and release of cytokines IL-6, IFN-α and TNF-α from murine myeloid dendritic cells (DCs) induced by Tianjin strain DIP. Moreover, real-time RT-PCR and immunohistochemistry were performed to identify whether the Tianjin strain DIP could induce infiltration of CD11c(+) DCs, CD4(+) and CD8(+) T cells in the tumors. RESULTS: On day 22 after CT26 cell inoculation, the average tumor volume of the Tianjin strain DIP group was (33.2 ± 2.0) mm(3), significantly smaller than that of the control group [(2 376.0 ± 130.8)mm(3), P < 0.01]. On day 50 after CT26 cell inoculation, the survival rate of mice was 90.0% in the Tianjin strain DIP group, much higher than that of the control group (30.0%, P < 0.01). Flow cytometry analysis showed that the expression of markers of DCs maturation, including CD40, CD80 and CD86, was dose-dependently increased by DIP or intact virus. No statistically significant difference was found betweent the DIP and intact virus groups. ELISA results showed that DIP could stimulate the secretion of IL-6, IFN-α and TNF-α from mouse DCs. The secretion of all of the cytokines was dose-dependently increased by DIP or intact virus. Real-time RT-PCR revealed that the expression of CD4, CD8 and CD11c mRNAs was increased in tumors treated with DIP compared with that of the saline group at all time points. Moreover, the expression level of all of them remained maximal at 120 h after the last treatment. Immunohistochemical staining revealed that the ratios of CD4(+), CD8(+) T cells or CD11c(+) DCs to total cells were (21.60 ± 1.49)%, (22.12 ± 2.84)% and (23.05 ± 2.91)%, respectively, in the DIP-treated tumors. In the tumors treated by saline, the ratios were (2.62 ± 0.60)%, (4.05 ± 0.12)% and (3.10 ± 0.09)%, respectively. The difference between experimental group and control group had statistical significance. CONCLUSIONS: Tianjin strain DIP may exert anti-tumor effect on tumor-bearing mice. The mechanism is related with the antitumor immunity induced by DCs and T cells.


Assuntos
Neoplasias do Colo/patologia , Vírus Defeituosos/imunologia , Células Dendríticas/metabolismo , Vírus Sendai/imunologia , Animais , Linhagem Celular Tumoral , Neoplasias do Colo/metabolismo , Citocinas/metabolismo , Feminino , Interferon-alfa/metabolismo , Interleucina-6/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Transplante de Neoplasias , Distribuição Aleatória , Linfócitos T/metabolismo , Carga Tumoral , Fator de Necrose Tumoral alfa/metabolismo
12.
Curr Top Microbiol Immunol ; 354: 53-73, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-21744306

RESUMO

Virus-like particles are a new type of vaccine platform that presents an attractive alternative to more traditional live-attenuated or inactivated/subunit vaccines. Virus-like particles (VLP) are composed of viral structural proteins that assemble spontaneously in cells, mimicking the live virus without the possibility of replication. They are readily recognized by the immune system, inducing both humoral and cellular immune responses. Here we review the development of VLP as vaccine delivery systems at mucosal surfaces. We first summarize the current status of VLP vaccines in general, and then discuss their use in mucosal vaccination approaches for several viruses that enter the host via the urogenital, respiratory or gastrointestinal tract.


Assuntos
Antígenos/administração & dosagem , Vírus Defeituosos/fisiologia , Sistemas de Liberação de Medicamentos/instrumentação , Mucosa/imunologia , Vacinação/instrumentação , Animais , Antígenos/imunologia , Vírus Defeituosos/imunologia , Sistemas de Liberação de Medicamentos/métodos , Humanos , Vacinação/métodos
13.
Nat Genet ; 7(3): 362-9, 1994 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-7522742

RESUMO

Although first generation recombinant adenoviruses, deleted of sequences spanning E1a and E1b, have been useful for in vivo applications of gene therapy, expression of the recombinant gene has been transient and often associated with the development of inflammation. We show that with first generation adenovirus-mediated gene transfer to the mouse lung, viral proteins are expressed leading to destructive cellular immune responses and repopulation of the lung with nontransgene containing cells. Second generation E1 deleted viruses further crippled by a temperature sensitive mutation in the E2a gene were associated with substantially longer recombinant gene expression and less inflammation. Stable expression of human CF transmembrane conductance regulator has been achieved in lungs of CF mice instilled with a second generation virus.


Assuntos
Proteínas E2 de Adenovirus/genética , Adenovírus Humanos/genética , Fibrose Cística/terapia , Vírus Defeituosos/genética , Terapia Genética , Vetores Genéticos , Infecções por Adenoviridae/virologia , Proteínas E1B de Adenovirus/deficiência , Proteínas E1B de Adenovirus/genética , Proteínas E2 de Adenovirus/deficiência , Adenovírus Humanos/imunologia , Adenovírus Humanos/patogenicidade , Animais , Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística , Vírus Defeituosos/imunologia , Vírus Defeituosos/patogenicidade , Regulação Viral da Expressão Gênica , Humanos , Imunidade Celular , Inflamação , Pulmão/imunologia , Pulmão/patologia , Pulmão/virologia , Proteínas de Membrana/biossíntese , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos CBA , Camundongos Mutantes , Camundongos Nus , Pneumonia Viral/etiologia , Pneumonia Viral/prevenção & controle , Pneumonia Viral/virologia , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Deleção de Sequência , Temperatura , Fatores de Tempo
14.
J Gen Virol ; 93(Pt 2): 299-307, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22049094

RESUMO

Conflicting reports exist regarding the requirement for virus replication in interferon (IFN) induction by paramyxoviruses. Our previous work has demonstrated that pathogen-associated molecular patterns capable of activating the IFN-induction cascade are not normally generated during virus replication, but are associated instead with the presence of defective interfering (DI) viruses. We demonstrate here that DIs of paramyxoviruses, including parainfluenza virus 5, mumps virus and Sendai virus, can activate the IFN-induction cascade and the IFN-ß promoter in the absence of virus protein synthesis. As virus protein synthesis is an absolute requirement for paramyxovirus genome replication, our results indicate that these DI viruses do not require replication to activate the IFN-induction cascade.


Assuntos
Interferon beta/biossíntese , Interferon beta/genética , Paramyxoviridae/imunologia , Paramyxoviridae/fisiologia , Regiões Promotoras Genéticas , Ativação Transcricional , Replicação Viral , Animais , Linhagem Celular , Vírus Defeituosos/genética , Vírus Defeituosos/imunologia , Humanos , Paramyxoviridae/genética , Rubulavirus , Proteínas Virais/biossíntese
15.
J Virol ; 85(19): 10213-21, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21795358

RESUMO

The reverse genetics technology for bluetongue virus (BTV) has been used in combination with complementing cell lines to recover defective BTV-1 mutants. To generate a potential disabled infectious single cycle (DISC) vaccine strain, we used a reverse genetics system to rescue defective virus strains with large deletions in an essential BTV gene that encodes the VP6 protein (segment S9) of the internal core. Four VP6-deficient BTV-1 mutants were generated by using a complementing cell line that provided the VP6 protein in trans. Characterization of the growth properties of mutant viruses showed that each mutant has the necessary characteristics for a potential vaccine strain: (i) viral protein expression in noncomplementing mammalian cells, (ii) no infectious virus generated in noncomplementing cells, and (iii) efficient replication in the complementing VP6 cell line. Further, a defective BTV-8 strain was made by reassorting the two RNA segments that encode the two outer capsid proteins (VP2 and VP5) of a highly pathogenic BTV-8 with the remaining eight RNA segments of one of the BTV-1 DISC viruses. The protective capabilities of BTV-1 and BTV-8 DISC viruses were assessed in sheep by challenge with specific virulent strains using several assay systems. The data obtained from these studies demonstrated that the DISC viruses are highly protective and could offer a promising alternative to the currently available attenuated and killed virus vaccines and are also compliant as DIVA (differentiating infected from vaccinated animals) vaccines.


Assuntos
Vírus Bluetongue/imunologia , Bluetongue/prevenção & controle , Vírus Defeituosos/imunologia , Vacinas Virais/imunologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Vírus Bluetongue/genética , Vírus Bluetongue/isolamento & purificação , Técnicas de Cultura de Células , Vírus Defeituosos/genética , Vírus Defeituosos/isolamento & purificação , Feminino , Masculino , Vírus Reordenados/genética , Vírus Reordenados/imunologia , Vírus Reordenados/isolamento & purificação , Ovinos , Vacinas Virais/genética , Vacinas Virais/isolamento & purificação , Viremia/prevenção & controle
16.
J Virol ; 85(5): 2004-11, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21177820

RESUMO

Replication-defective vaccine vectors based on vesicular stomatitis virus (VSV) lacking its envelope glycoprotein gene (G) are highly effective in animal models. However, such ΔG vectors are difficult to grow because they require complementation with the VSV G protein. In addition, the complementing G protein induces neutralizing antibodies in animals and thus limits multiple vector applications. In the process of generating an experimental Nipah virus (a paramyxovirus) vaccine, we generated two defective VSVΔG vectors, each expressing one of the two Nipah virus (NiV) glycoproteins (G and F) that are both required for virus entry to host cells. These replication-defective VSV vectors were effective at generating NiV neutralizing antibody in mice. Most interestingly, we found that these two defective viruses could be grown together and passaged in tissue culture cells in the absence of VSV G complementation. This mixture of complementing defective viruses was also highly effective at generating NiV neutralizing antibody in animals. This novel approach to growing and producing a vaccine from two defective viruses could be generally applicable to vaccine production for other paramyxoviruses or for other viruses where the expression of at least two different proteins is required for viral entry. Such an approach minimizes biosafety concerns that could apply to single, replication-competent VSV recombinants expressing all proteins required for infection.


Assuntos
Vírus Defeituosos/genética , Expressão Gênica , Vetores Genéticos/genética , Vírus Nipah/imunologia , Vesiculovirus/imunologia , Proteínas do Envelope Viral/imunologia , Vacinas Virais/imunologia , Animais , Linhagem Celular , Vírus Defeituosos/imunologia , Vírus Defeituosos/fisiologia , Feminino , Teste de Complementação Genética , Vetores Genéticos/imunologia , Infecções por Henipavirus/imunologia , Infecções por Henipavirus/prevenção & controle , Infecções por Henipavirus/virologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Vírus Nipah/genética , Vesiculovirus/genética , Vesiculovirus/fisiologia , Proteínas do Envelope Viral/genética , Vacinas Virais/genética , Replicação Viral
17.
J Immunol ; 185(3): 1513-21, 2010 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-20610651

RESUMO

Replication-defective adenovirus serotype 5 (rAd5) is the most potent recombinant vector for eliciting CD8 T cell responses in humans. In this study, the innate mechanisms that influence T cell responses following rAd5 immunization were assessed in mice. Using rAd5 expressing enhanced GFP (eGFP-rAd5), we show that rAd5 transfects CD11c(+) dendritic cells (DCs) in draining lymph nodes in vivo following s.c. or i.m. immunization. Among distinct DC subsets, eGFP expression was highest in CD11c(+)CD8(-)B220(-) with a lower frequency detected in CD11c(+)CD8(+)B220(-) and CD11c(+)B220(+) plasmacytoid DCs. CD11c(+) DCs but not CD11c(-) cells from mice immunized with rAd5 encoding the SIINFEKL peptide induced proliferation of naive OT-I CD8 T cells. Furthermore, CD11c(+)CD8(+)B220(-) was the most potent DC subset for eliciting naive OT-I CD8 T cell proliferation. Of note, mice with pre-existing immunity to rAd5 had a substantial decrease in eGFP expression in DCs, which was associated with approximately 2-fold decrease in Th1 and complete inhibition of CD8 responses. Thus, pre-existing rAd5 immunity has a greater influence on CD8 compared with CD4 T cell responses. In terms of how innate cytokines and signaling pathways influenced T cell immunity following rAd5 immunization, we show that the magnitude and quality of CD8 T cell responses are partially dependent on MyD88 but independent of IL-12, type I IFN, apoptosis-associated speck-like protein, nucleotide-binding oligomerization domain-like receptor protein 3, and IL-1. Taken together, these data demonstrate a critical role for CD11c(+) DCs for CD8 responses but striking redundancy for innate cytokines and signaling by TLR and nucleotide-binding oligomerization domain-like receptor pathways.


Assuntos
Adenovírus Humanos/imunologia , Antígeno CD11c/biossíntese , Linfócitos T CD8-Positivos/imunologia , Vírus Defeituosos/imunologia , Células Dendríticas/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Oligodesoxirribonucleotídeos/metabolismo , Receptores Toll-Like/fisiologia , Adenovírus Humanos/genética , Animais , Apresentação de Antígeno/imunologia , Antígeno CD11c/genética , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/virologia , Células Dendríticas/metabolismo , Células Dendríticas/virologia , Imunidade Inata , Imunofenotipagem , Interferon Tipo I/fisiologia , Interleucina-12/fisiologia , Linfonodos/imunologia , Linfonodos/patologia , Linfonodos/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Receptores Toll-Like/genética , Vacinas Virais/administração & dosagem , Vacinas Virais/genética , Vacinas Virais/imunologia , Vírion/imunologia , Vírion/patogenicidade
18.
Appl Microbiol Biotechnol ; 94(1): 39-46, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22382168

RESUMO

Virus-like particles (VLPs) are shell-like viruses that lack virus-specific genetic materials. Many viral-structured proteins can assemble into VLPs, which mimic the overall structure of virus particles and can elicit strong immune responses in a host. Dengue viruses (DENVs), from the genus Flavivirus, are transmitted to humans through the bites of an infected Aedes mosquito. DENVs cause several diseases that prevailed mainly in tropical and subtropical areas. However, effective treatment measures and preventive strategies for dengue diseases are still lacking. The present minireview summarized the assembly and maturation of DENVs, the strategies and effective factors for dengue VLP construction, and the application of DENV VLPs.


Assuntos
Vírus Defeituosos/fisiologia , Vírus da Dengue/fisiologia , Dengue/virologia , Animais , Vírus Defeituosos/genética , Vírus Defeituosos/imunologia , Dengue/imunologia , Vírus da Dengue/genética , Vírus da Dengue/imunologia , Humanos
19.
Mol Ther ; 19(3): 612-9, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21206484

RESUMO

Based on the concept that anticocaine antibodies could prevent inhaled cocaine from reaching its target receptors in the brain, an effective anticocaine vaccine could help reverse cocaine addiction. Leveraging the knowledge that E1(-)E3(-) adenovirus (Ad) gene transfer vectors are potent immunogens, we have developed a novel vaccine platform for addictive drugs by covalently linking a cocaine analog to the capsid proteins of noninfectious, disrupted Ad vector. The Ad-based anticocaine vaccine evokes high-titer anticocaine antibodies in mice sufficient to completely reverse, on a persistent basis, the hyperlocomotor activity induced by intravenous administration of cocaine.


Assuntos
Cocaína/análogos & derivados , Cocaína/imunologia , Transtornos Relacionados ao Uso de Substâncias , Vacinas , Adenoviridae/genética , Animais , Anticorpos/sangue , Cocaína/metabolismo , Vírus Defeituosos/genética , Vírus Defeituosos/imunologia , Vírus Defeituosos/metabolismo , Feminino , Técnicas de Transferência de Genes , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Locomoção/efeitos dos fármacos , Locomoção/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Atividade Motora/efeitos dos fármacos , Atividade Motora/imunologia , Transtornos Relacionados ao Uso de Substâncias/imunologia , Transtornos Relacionados ao Uso de Substâncias/prevenção & controle , Vacinas/administração & dosagem , Vacinas/imunologia
20.
J Gen Virol ; 92(Pt 9): 2122-2132, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21632569

RESUMO

Influenza A and B viruses are major human respiratory pathogens that contribute to the burden of seasonal influenza. They are both members of the family Orthomyxoviridae but do not interact genetically and are classified in different genera. Defective interfering (DI) influenza viruses have a major deletion of one or more of their eight genome segments, which renders them both non-infectious and able to interfere in cell culture with the production of infectious progeny by a genetically compatible, homologous virus. It has been shown previously that intranasal administration of a cloned DI influenza A virus, 244/PR8, protects mice from various homologous influenza A virus subtypes and that it also protects mice from respiratory disease caused by a heterologous virus belonging to the family Paramyxoviridae. The mechanisms of action in vivo differ, with homologous and heterologous protection being mediated by probable genome competition and type I interferon (IFN), respectively. In the current study, it was shown that 244/PR8 also protects against disease caused by a heterologous influenza B virus (B/Lee/40). Protection from B/Lee/40 challenge was partially eliminated in mice that did not express a functional type I IFN receptor, suggesting that innate immunity, and type I IFN in particular, are important in mediating protection against this virus. It was concluded that 244/PR8 has the ability to protect in vivo against heterologous IFN-sensitive respiratory viruses, in addition to homologous influenza A viruses, and that it acts by fundamentally different mechanisms.


Assuntos
Proteção Cruzada , Vírus Defeituosos/imunologia , Vírus da Influenza A/imunologia , Vírus da Influenza B/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Animais , Modelos Animais de Doenças , Feminino , Imunidade Inata , Interferon Tipo I/imunologia , Masculino , Camundongos , Doenças dos Roedores/prevenção & controle
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA