Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Int J Mol Sci ; 24(14)2023 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-37511205

RESUMO

Intranasal vaccination using influenza vectors is a promising approach to developing vaccines against respiratory pathogens due to the activation of the mucosa-associated immune response. However, there is no clear evidence of a vector design that could be considered preferable. To find the optimal structure of an influenza vector with a modified NS genomic segment, we constructed four vector expressing identical transgene sequences inherited from the F protein of the respiratory syncytial virus (RSV). Two vectors were designed aiming at transgene accumulation in the cytosol. Another two were supplemented with an IgGκ signal peptide prior to the transgene for its extracellular delivery. Surprisingly, adding the IgGκ substantially enhanced the T-cell immune response to the CD8 epitope of the transgene. Moreover, this strategy allowed us to obtain a better protection of mice from the RSV challenge after a single intranasal immunization. Protection was achieved without antibodies, mediated by a balanced T-cell immune response including the formation of the RSV specific effector CD8+ IFNγ+/IL10+-producing cells and the accumulation of Treg cells preventing immunopathology in the lungs of infected mice. In addition to the presented method for optimizing the influenza vector, our results highlight the possibility of achieving protection against RSV through a respiratory-associated T-cell immune response alone.


Assuntos
Vacinas contra Influenza , Influenza Humana , Infecções por Vírus Respiratório Sincicial , Vacinas contra Vírus Sincicial Respiratório , Vírus Sincicial Respiratório Humano , Animais , Camundongos , Humanos , Anticorpos Antivirais , Vírus Sincicial Respiratório Humano/genética , Camundongos Endogâmicos BALB C
2.
Arch Virol ; 160(10): 2525-34, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26215439

RESUMO

The existence of multiple antigenically distinct types and subtypes of influenza viruses allows the construction of a multivalent vector system for the mucosal delivery of foreign sequences. Influenza A viruses have been exploited successfully for the expression of extraneous antigens as well as immunostimulatory molecules. In this study, we describe the development of an influenza B virus vector whose functional part of the interferon antagonist NS1 was replaced by human interleukin 2 (IL2) as a genetic adjuvant. We demonstrate that IL2 expressed by this viral vector displays immune adjuvant activity in immunized mice. Animals vaccinated with the IL2 viral vector showed an increased hemagglutination inhibition antibody response and higher protective efficacy after challenge with a wild-type influenza B virus when compared to mice vaccinated with a control virus. Our results demonstrate that it is feasible to construct influenza B vaccine strains expressing immune-potentiating foreign sequences from the NS genomic segment. Based on these data, it is now hypothetically possible to create a trivalent (or quadrivalent) live attenuated influenza vaccine in which each component expresses a selected genetic adjuvant with tailored expression levels.


Assuntos
Vetores Genéticos/imunologia , Vírus da Influenza B/imunologia , Influenza Humana/prevenção & controle , Adjuvantes Imunológicos/administração & dosagem , Animais , Anticorpos Antivirais/imunologia , Feminino , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Humanos , Imunização , Vírus da Influenza B/genética , Vírus da Influenza B/fisiologia , Influenza Humana/imunologia , Influenza Humana/virologia , Interleucina-2/genética , Interleucina-2/imunologia , Camundongos
3.
J Gen Virol ; 95(Pt 2): 337-349, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24222196

RESUMO

The development of influenza virus vectors with long insertions of foreign sequences remains difficult due to the small size and instable nature of the virus. Here, we used the influenza virus inherent property of self-optimization to generate a vector stably expressing long transgenes from the NS1 protein ORF. This was achieved by continuous selection of bright fluorescent plaques of a GFP-expressing vector during multiple passages in mouse B16f1 cells. The newly generated vector acquired stability in IFN-competent cell lines and in vivo in murine lungs. Although improved vector fitness was associated with the appearance of four coding mutations in the polymerase (PB2), haemagglutinin and non-structural (NS) segments, the stability of the transgene expression was dependent primarily on the single mutation Q20R in the nuclear export protein (NEP). Importantly, a longer insert, such as a cassette of 1299 nt encoding two Mycobacterium tuberculosis Esat6 and Ag85A proteins, could substitute for the GFP transgene. Thus, the inherent property of the influenza virus to adapt can also be used to adjust a vector backbone to give stable expression of long transgenes.


Assuntos
Transporte Ativo do Núcleo Celular , Expressão Gênica , Vírus da Influenza A/genética , Vírus da Influenza A/fisiologia , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo , Replicação Viral , Animais , Linhagem Celular , Genes Reporter , Vetores Genéticos , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Camundongos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Carga Viral
4.
Virol J ; 11: 69, 2014 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-24716528

RESUMO

BACKGROUND: We generated novel, effective candidate vaccine against Brucella abortus based on recombinant influenza viruses expressing the Brucella ribosomal protein L7/L12 or outer membrane protein (Omp)-16 from the NS1 open reading frame. The main purpose of this work was to evaluate the safety, immunogenicity and protectiveness of vaccine candidate in laboratory animals. METHODS AND RESULTS: Four recombinant influenza A viral constructs of the subtypes Н5N1 or H1N1 expressing the Brucella proteins L7/L12 or Omp16 were obtained by a reverse genetics method: Flu-NS1-124-L7/L12-H5N1, Flu-NS1-124-Omp16-H5N1, Flu-NS1-124-L7/L12-H1N1 and Flu-NS1-124-Omp16-H1N1. Despite of substantial modification of NS1 gene, all constructs replicated well and were retain their Brucella inserts over five passages in embryonated chicken eggs (CE). Administration of the mono- or bivalent vaccine formulation via prime-boost intranasal (i.n.), conjunctival (c.) or subcutaneous (s.c.) immunization was safe in mice; no deaths, body weight loss or pathomorphological changes were observed over 56 days. Moreover, guinea pigs vaccinated i.n. with vaccine vectors did not shed the vaccine viruses through their upper respiratory tract after the prime and booster vaccination. These findings confirmed the replication-deficient phenotype of viral vectors. The highest antibody response to Brucella antigen was obtained with constructs expressing L7/L12 (ELISA, GMT 242.5-735.0); whereas the highest T-cell immune response- with construct expressing Omp16 (ELISPOT, 337 ± 52-651 ± 45 spots/4×105cells), which was comparable (P > 0.05) to the response induced by the commercial vaccine B. abortus 19. Interestingly, c. immunization appeared to be optimal for eliciting T-cell immune response. In guinea pigs, the highest protective efficacy after challenge with B. abortus 544 was achieved with Omp16 expressing constructs in both monovalent or bivalent vaccine formulations; protective efficacy was comparable to those induced by a commercial live B. abortus 19 vaccine. CONCLUSION: Thus, influenza vectors expressing Brucella protective antigens can be developed as novel influenza vectored vaccine against B. abortus infection.


Assuntos
Antígenos de Bactérias/imunologia , Vacina contra Brucelose/imunologia , Brucella abortus/imunologia , Brucelose/prevenção & controle , Portadores de Fármacos , Vírus da Influenza A Subtipo H1N1/genética , Virus da Influenza A Subtipo H5N1/genética , Animais , Anticorpos Antibacterianos/sangue , Antígenos de Bactérias/genética , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/imunologia , Vacina contra Brucelose/administração & dosagem , Vacina contra Brucelose/genética , Brucella abortus/genética , Brucelose/imunologia , Modelos Animais de Doenças , Vetores Genéticos , Instabilidade Genômica , Cobaias , Camundongos , Proteínas Ribossômicas/genética , Proteínas Ribossômicas/imunologia , Análise de Sobrevida , Linfócitos T/imunologia , Vacinação/métodos , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Replicação Viral
5.
J Virol ; 85(5): 2469-73, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21177819

RESUMO

Segment 8 of the influenza A virus codes for two proteins (NS1 and NS2/NEP) via splicing. Here, we developed a viral vector expressing a cytokine or chemokine instead of the interferon antagonist NS1. To achieve both the desired genetic stability and high transgene expression levels, NS2/NEP mRNA splicing efficacy had to be fine-tuned by modification of splicing elements. Expression levels of secreted foreign proteins could be further enhanced by fusing the N-terminal 13 amino acids of NS1 with an IgK-derived secretion signal peptide. Thus, the first start codon was used for translation initiation of both NS2/NEP and the foreign protein.


Assuntos
Vírus Defeituosos/genética , Expressão Gênica , Vetores Genéticos/genética , Vírus da Influenza A/genética , Splicing de RNA , Vírus Defeituosos/fisiologia , Vetores Genéticos/fisiologia , Vírus da Influenza A/fisiologia , Replicação Viral
6.
J Virol ; 85(21): 11139-45, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21849438

RESUMO

In general, antibiotics are not rated as substances that inhibit or support influenza virus replication. We describe here the enhancing effect of the polyene antibiotic amphotericin B (AmB) on influenza virus growth in Vero cells. We show that isolation rates of influenza A and B viruses from clinical samples can be dramatically enhanced by adding AmB to the culture medium. We demonstrate that AmB promotes the viral uptake and endocytic processing of the virus particles. This effect is specific for Vero and human nasal epithelial cells and was not observed in Madin-Darby canine kidney cells. The effect of AmB was subtype specific and more prominent for human seasonal influenza strains but absent for H5N1 human viruses. The AmB-enhancing effect seemed to be solely due to the viral hemagglutinin function. Our results indicate that the use of AmB may facilitate influenza virus isolation and production in Vero cells.


Assuntos
Anfotericina B/metabolismo , Antifúngicos/metabolismo , Vírus da Influenza A/efeitos dos fármacos , Vírus da Influenza A/crescimento & desenvolvimento , Vírus da Influenza B/efeitos dos fármacos , Vírus da Influenza B/crescimento & desenvolvimento , Replicação Viral/efeitos dos fármacos , Animais , Linhagem Celular , Chlorocebus aethiops , Cães , Endocitose/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/virologia , Humanos , Vírus da Influenza A/isolamento & purificação , Vírus da Influenza B/isolamento & purificação , Influenza Humana/virologia
7.
J Infect Dis ; 201(3): 354-62, 2010 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-20039806

RESUMO

BACKGROUND. The nonstructural protein NS1 of influenza virus counteracts the interferon-mediated immune response of the host. By deleting the open reading frame of NS1, we have generated a novel type of influenza vaccine. We studied the safety and immunogenicity of an influenza strain lacking the NS1 gene (DeltaNS1-H1N1) in healthy volunteers. METHODS. Healthy seronegative adult volunteers were randomized to receive either a single intranasal dose of the DeltaNS1-H1N1 A/New Caledonia vaccine at 1 of 5 dose levels (6.4, 6.7, 7.0, 7.4, and 7.7 log(10) median tissue culture infective dose) (n = 36 recipients) or placebo (n = 12 recipients). RESULTS. Intranasal vaccination with the replication-deficient DeltaNS1-H1N1 vaccine was well tolerated. Rhinitis-like symptoms and headache were the most common adverse events identified during the 28-day observation period. Adverse events were similarly distributed between the treatment and placebo groups. Vaccine-specific local and serum antibodies were induced in a dose-dependent manner. In the highest dose group, vaccine-specific antibodies were detected in 10 of 12 volunteers. Importantly, the vaccine also induced neutralizing antibodies against heterologous drift variants. CONCLUSIONS. We show that vaccination with an influenza virus strain lacking the viral interferon antagonist NS1 induces statistically significant levels of strain-specific and cross-neutralizing antibodies despite the highly attenuated replication-deficient phenotype. Further studies are warranted to determine whether these results translate into protection from influenza virus infection. TRIAL REGISTRATION. ClinicalTrials.gov identifier: NCT00724997 .


Assuntos
Vírus da Influenza A Subtipo H1N1/imunologia , Vacinas contra Influenza/imunologia , Influenza Humana/prevenção & controle , Vacinas Atenuadas/imunologia , Proteínas não Estruturais Virais/genética , Adulto , Anticorpos Antivirais/sangue , Anticorpos Antivirais/isolamento & purificação , Relação Dose-Resposta Imunológica , Método Duplo-Cego , Deleção de Genes , Humanos , Vírus da Influenza A Subtipo H1N1/genética , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/efeitos adversos , Líquido da Lavagem Nasal/imunologia , Líquido da Lavagem Nasal/virologia , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/efeitos adversos , Eliminação de Partículas Virais
8.
Microorganisms ; 9(4)2021 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-33810549

RESUMO

Influenza viruses with an impaired NS1 protein are unable to antagonize the innate immune system and, therefore, are highly immunogenic because of the self-adjuvating effect. Hence, NS1-mutated viruses are considered promising candidates for the development of live-attenuated influenza vaccines and viral vectors for intranasal administration. We investigated whether the immunogenic advantage of the virus expressing only the N-terminal half of the NS1 protein (124 a.a.) can be translated into the induction of protective immunity against a heterologous influenza virus in mice. We found that immunization with either the wild-type A/PR/8/34 (H1N1) influenza strain (A/PR8/NSfull) or its NS1-shortened counterpart (A/PR8/NS124) did not prevent the viral replication in the lungs after the challenge with the A/Aichi/2/68 (H3N2) virus. However, mice immunized with the NS1-shortened virus were better protected from lethality after the challenge with the heterologous virus. Besides showing the enhanced influenza-specific CD8+ T-cellular response in the lungs, immunization with the A/PR8/NS124 virus resulted in reduced concentrations of proinflammatory cytokines and the lower extent of leukocyte infiltration in the lungs after the challenge compared to A/PR8/NSfull or the control group. The data show that intranasal immunization with the NS1-truncated virus may better induce not only effector T-cells but also certain immunoregulatory mechanisms, reducing the severity of the innate immune response after the heterologous challenge.

9.
Med Microbiol Immunol ; 199(2): 93-101, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20012989

RESUMO

Tumor resistance to lysis by resting natural killer (NK) cells may be overcome by priming of NK cells with cytokines or by binding of NK activating receptors to ligands expressed on target cells. In this study, major histocompatibility complex class I (MHC-I)-negative LNCaP and MHC-I-positive DU145 cells were infected with genetically modified influenza A virus lacking the non-structural gene 1 (NS1 IAV). The cells were used to investigate the influence of NS1 IAV infection on NK cell lysis of tumor cells as well as to prime NK cells for lysis of LNCaP and DU145 cells. While LNCaP cells infected with DeltaNS1 IAV showed enhanced lysis when compared with mock-infected cells (93% +/- 1.47 vs. 52% +/- 0.74), both mock-infected and DeltaNS1 IAV-infected DU145 cells were resistant to NK cell lysis. Moreover, NK cells primed with DeltaNS1 IAV-infected LNCaP/DU145 cells effectively lysed resistant DU145 and sensitive LNCaP cells to a greater extent than NK cells primed with mock-infected LNCaP/DU145 or non-primed NK cells. Also, NK cell priming with DeltaNS1 IAV-infected tumor cells enhanced extracellular signal-regulated kinase phosphorylation and increased granule release in NK cells. The increased granule release was specifically mediated by NKp46, which eventually potentiated NK cells primed with DeltaNS1 IAV-infected tumor cells to overcome the inhibitory effects posed by MHC-I expression on DU145 cells. These findings show that in addition to direct lytic activity of NK cells, DeltaNS1 IAV may influence anti-tumoral responses by priming NK cells.


Assuntos
Vírus da Influenza A/imunologia , Células Matadoras Naturais/imunologia , Vírus Oncolíticos/imunologia , Vacinas Anticâncer , Degranulação Celular , Linhagem Celular Tumoral , Sobrevivência Celular , Antígenos de Histocompatibilidade Classe I/imunologia , Humanos , Masculino , Receptor 1 Desencadeador da Citotoxicidade Natural/imunologia , Neoplasias da Próstata/imunologia , Neoplasias da Próstata/virologia
10.
Vaccines (Basel) ; 7(4)2019 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-31744208

RESUMO

: Influenza virus infections pre-dispose an individual to secondary pneumococcal infections, which represent a serious public health concern. Matching influenza vaccination was demonstrated helpful in preventing postinfluenza bacterial infections and associated illnesses in humans. Yet, the impact of influenza hemagglutinin (HA)-specific immunity alone in this dual-infection scenario remains elusive. In the present study, we assessed the protective effect of neutralizing and non-neutralizing anti-hemagglutinin immunity in a BALB/c influenza-pneumococcus superinfection model. Our immunogens were insect cell-expressed hemagglutinin-Gag virus-like particles that had been differentially-treated for the inactivation of bioprocess-related baculovirus impurities. We evaluated the potential of several formulations to restrain the primary infection with vaccine-matched or -mismatched influenza strains and secondary bacterial replication. In addition, we investigated the effect of anti-HA immunity on the interferon status in mouse lungs prior to bacterial challenge. In our experimental setup, neutralizing anti-HA immunity provided significant but incomplete protection from postinfluenza bacterial superinfection, despite effective control of viral replication. In view of this, it was surprising to observe a survival advantage with non-neutralizing adaptive immunity when using a heterologous viral challenge strain. Our findings suggest that both neutralizing and non-neutralizing anti-HA immunity can reduce disease and mortality caused by postinfluenza pneumococcal infections.

11.
Surgery ; 161(3): 735-746, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27776794

RESUMO

BACKGROUND: Interleukin-15 has become a promising molecule in the context of eliciting an effective, antitumor immune response because it is able to stimulate cells of the innate and adaptive immune system. METHODS: We generated an interleukin-15-expressing oncolytic influenza A virus for the treatment of an established murine tumor model. RESULTS: Our oncolytic influenza A virus produced large amounts of interleukin-15 and induced proliferation and activation of human T cells in vitro. Intraperitoneal administration increased the amount of mouse natural killer cells and effector memory T cells, as well as T cell reactivity in vivo. Moreover, intratumoral injection induced a profound decrease in growth of established tumors in mice and increased the amount of tumor-infiltrating T cells and natural killer cells. CONCLUSION: We established a stable, IL-15-producing oncolytic influenza A virus with promising immunostimulatory and antitumor attributes.


Assuntos
Vírus da Influenza A , Interleucina-15/fisiologia , Melanoma Experimental/patologia , Terapia Viral Oncolítica , Vírus Oncolíticos , Neoplasias Cutâneas/patologia , Animais , Técnicas de Cultura de Células , Proliferação de Células/efeitos dos fármacos , Feminino , Humanos , Células Matadoras Naturais/efeitos dos fármacos , Melanoma Experimental/terapia , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias Cutâneas/terapia , Linfócitos T/efeitos dos fármacos
12.
Mol Ther Oncolytics ; 7: 37-44, 2017 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-29034314

RESUMO

Oncolytic viruses are currently established as a novel type of immunotherapy. The challenge is to safely target oncolytic viruses to tumors. Previously, we have generated influenza A viruses (IAVs) containing deletions in the viral interferon antagonist. Those deletions have attenuated the virus in normal tissue but allowed replication in tumor cells. IAV entry is mediated by hemagglutinin (HA), which needs to be activated by a serine protease, for example, through trypsin. To further target the IAV to tumors, we have changed the trypsin cleavage site to an elastase cleavage site. We chose this cleavage site because elastase is expressed in the tumor microenvironment. Moreover, the exchange of the cleavage site previously has been shown to attenuate viral growth in lungs. Newly generated elastase-activated influenza viruses (AE viruses) grew to similar titers in tumor cells as the trypsin-activated counterparts (AT viruses). Intratumoral injection of AE viruses into syngeneic B16f1 melanoma-derived tumors in mice reduced tumor growth similar to AT viruses and had a better therapeutic effect in heterologous human PANC-1-derived tumors. Therefore, the introduction of the attenuation marker "elastase cleavage site" in viral HA allows for safe, effective oncolytic virus therapy.

13.
Antiviral Res ; 123: 93-104, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26367324

RESUMO

New anti-viral agents and strategies are urgently needed to fight rapidly mutating viruses, as vaccine programs cannot react fast enough to prevent pandemics. Recently, we have shown that interleukin-24 (IL-24) sensitizes tumor cells to toll-like receptor 3 (TLR3) mediated apoptosis. As influenza A virus stimulates the TLR3 receptor, we hypothesized that IL-24 might also exert an anti-viral effect. This study demonstrates that IL-24 reduces the titer of different influenza A virus subtypes independently of type I interferon in an apoptosis dependent manner. The anti-viral effect of IL-24 correlated with caspase-3 activation and could be blocked by a pan-caspase inhibitor and by small interfering RNA (siRNA) directed towards TLR3. Surprisingly, caspase-3 activation in influenza A virus/IL-24-stimulated cells correlated with the down-regulation of the B-cell lymphoma 2 (Bcl-2) family member myeloid cell leukemia 1 (Mcl-1). Correspondingly, knockdown of Mcl-1 by siRNA enhanced caspase activation in influenza A virus infected cells and was furthermore linked to a reduction of viral titers. We conclude that IL-24 exerts an anti-viral role selectively purging virally infected cells by leading to a down-regulation of Mcl-1. Our findings might therefore represent the first step towards a new rational concept in the development of anti-viral strategies based on the induction of apoptosis.


Assuntos
Apoptose , Vírus da Influenza A/imunologia , Vírus da Influenza A/fisiologia , Interleucinas/metabolismo , Receptor 3 Toll-Like/metabolismo , Replicação Viral , Animais , Linhagem Celular , Humanos , Camundongos Endogâmicos BALB C
14.
Virus Res ; 103(1-2): 187-93, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15163508

RESUMO

The African green monkey kidney (Vero) cell line was used as a substrate for the development of a live cold-adapted (ca) reassortant influenza vaccine. For that purpose, a new master strain was generated by an adaptation of the wild type (wt) A/Singapore/1/57 virus to growth at 25 degrees C in a Vero cell line. The resulting cold-adapted (ca) muster strain A/Singapore/1/57ca showed temperature sensitive (ts) phenotype and was attenuated in animal models and protective in the challenge experiments in ferrets. Two vaccine candidates of influenza A(H1N1) and A(H3N2) subtypes (6/2 reassortants) inheriting six genes coding internal proteins from the new master strain and the surface antigens hemagglutinin (HA) and neuraminidase (NA) from the epidemic viruses were obtained by a standard method of genetic reassortment. All steps of the vaccine preparation were done exclusively in Vero cells, including the isolation of the epidemic viruses. Both vaccine strains were used for immunization of young adult volunteers in a limited clinical trial and appeared to be safe, well tolerated and immunogenic after intranasal administration.


Assuntos
Adaptação Fisiológica , Temperatura Baixa , Vírus da Influenza A/fisiologia , Vacinas contra Influenza , Adulto , Animais , Chlorocebus aethiops , Furões , Humanos , Vírus da Influenza A/genética , Vírus da Influenza A/patogenicidade , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/efeitos adversos , Vacinas contra Influenza/imunologia , Influenza Humana/prevenção & controle , Camundongos , Camundongos Endogâmicos BALB C , Vacinação , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/efeitos adversos , Vacinas Atenuadas/imunologia , Células Vero/virologia , Replicação Viral
15.
J Virol Methods ; 110(1): 111-4, 2003 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-12757928

RESUMO

A recombinant baculovirus vector containing mammalian cell-active promoters and transcription terminators was used to deliver a mutated influenza NS gene into Vero cells. In addition to the influenza NS gene, the baculovirus contained a reporter gene expression cassette (Green fluorescent protein, GFP), allowing to monitor the Vero cell transduction efficiency. More than 90% of Vero cells were expressing GFP 24-48 h post transduction. After infecting baculovirus transduced cells with influenza helper virus, progeny of attenuated influenza virus carrying the recombinant NS gene could be selected. Baculovirus delivery was highly reproducible and efficient in Vero cells. This new method for influenza gene delivery could contribute to influenza virus research and vaccine development.


Assuntos
Baculoviridae/genética , Vetores Genéticos , Vírus da Influenza A/genética , Transdução Genética , Proteínas não Estruturais Virais/genética , Animais , Chlorocebus aethiops , Proteínas de Fluorescência Verde , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Recombinação Genética , Células Vero , Proteínas não Estruturais Virais/metabolismo
16.
PLoS One ; 7(5): e36506, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22563505

RESUMO

Oncolytic influenza A viruses with deleted NS1 gene (delNS1) replicate selectively in tumour cells with defective interferon response and/or activated Ras/Raf/MEK/ERK signalling pathway. To develop a delNS1 virus with specific immunostimulatory properties, we used an optimised technology to insert the interleukin-15 (IL-15) coding sequence into the viral NS gene segment (delNS1-IL-15). DelNS1 and delNS1-IL-15 exerted similar oncolytic effects. Both viruses replicated and caused caspase-dependent apoptosis in interferon-defective melanoma cells. Virus replication was required for their oncolytic activity. Cisplatin enhanced the oncolytic activity of delNS1 viruses. The cytotoxic drug increased delNS1 replication and delNS1-induced caspase-dependent apoptosis. Interference with MEK/ERK signalling by RNAi-mediated depletion or the MEK inhibitor U0126 did not affect the oncolytic effects of the delNS1 viruses. In oncolysis sensitive melanoma cells, delNS1-IL-15 (but not delNS1) infection resulted in the production of IL-15 levels ranging from 70 to 1140 pg/mL in the cell culture supernatants. The supernatants of delNS1-IL-15-infected (but not of delNS1-infected) melanoma cells induced primary human natural killer cell-mediated lysis of non-infected tumour cells. In conclusion, we constructed a novel oncolytic influenza virus that combines the oncolytic activity of delNS1 viruses with immunostimulatory properties through production of functional IL-15. Moreover, we showed that the oncolytic activity of delNS1 viruses can be enhanced in combination with cytotoxic anti-cancer drugs.


Assuntos
Vírus da Influenza A Subtipo H1N1/genética , Interleucina-15/genética , Vírus Oncolíticos/genética , Proteínas não Estruturais Virais/genética , Clorometilcetonas de Aminoácidos/farmacologia , Animais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Western Blotting , Inibidores de Caspase , Caspases/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Chlorocebus aethiops , Cisplatino/farmacologia , Inibidores de Cisteína Proteinase/farmacologia , Deleção de Genes , Humanos , Vírus da Influenza A Subtipo H1N1/crescimento & desenvolvimento , Interleucina-15/metabolismo , Melanoma/genética , Melanoma/patologia , Melanoma/virologia , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Vírus Oncolíticos/crescimento & desenvolvimento , Interferência de RNA , Células Vero , Replicação Viral/efeitos dos fármacos
17.
PLoS One ; 7(6): e39921, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22761928

RESUMO

The nonstructural protein 1 (NS1) of influenza A virus (IAV) enables the virus to disarm the host cell type 1 IFN defense system. Mutation or deletion of the NS1 gene leads to attenuation of the virus and enhances host antiviral response making such live-attenuated influenza viruses attractive vaccine candidates. Sublingual (SL) immunization with live influenza virus has been found to be safe and effective for inducing protective immune responses in mucosal and systemic compartments. Here we demonstrate that SL immunization with NS1 deleted IAV (DeltaNS1 H1N1 or DeltaNS1 H5N1) induced protection against challenge with homologous as well as heterosubtypic influenza viruses. Protection was comparable with that induced by intranasal (IN) immunization and was associated with high levels of virus-specific antibodies (Abs). SL immunization with DeltaNS1 virus induced broad Ab responses in mucosal and systemic compartments and stimulated immune cells in mucosa-associated and systemic lymphoid organs. Thus, SL immunization with DeltaNS1 offers a novel potential vaccination strategy for the control of influenza outbreaks including pandemics.


Assuntos
Vírus da Influenza A/imunologia , Vacinas contra Influenza/administração & dosagem , Proteínas não Estruturais Virais/imunologia , Administração Sublingual , Animais , Anticorpos Antivirais/biossíntese , Ensaio de Imunoadsorção Enzimática , Feminino , Imunidade nas Mucosas , Tecido Linfoide/imunologia , Camundongos , Camundongos Endogâmicos BALB C
18.
Vaccine ; 29(19): 3517-24, 2011 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-21406268

RESUMO

The isolation and cultivation of human influenza viruses in embryonated hen eggs or cell lines often leads to amino acid substitutions in the haemagglutinin (HA) molecule. We found that the propagation of influenza A H3N2 viruses on Vero cells may trigger the appearance of HA destabilising mutations, affecting viral resistance to low pH or high temperature treatment. Two ΔNS1 reassortants, containing the HA sequences identical to the original human H3N2 influenza virus isolates were constructed. Passages of these viruses on Vero cells led to the appearance of single mutations in the HA(1) L194P or HA(2) G75R subunits that impaired virus stability. The original HA sequences and the stable phenotypes of the primary isolates were preserved if reassortants were passaged by infection at pH 5.6 and cultivation in medium at pH 6.5. Corresponding ΔNS1 reassortants were compared for their immunogenicity in ferrets upon intranasal immunisation. Vaccine candidates containing HA mutations demonstrated significantly lower immunogenicity compared to those without mutations. Thus, the retaining of the original HA sequences of human viruses during vaccine production might be crucial for the efficacy of live attenuated influenza vaccines.


Assuntos
Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Vírus da Influenza A Subtipo H3N2/genética , Administração Intranasal , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Linhagem Celular , Chlorocebus aethiops , Feminino , Furões , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Concentração de Íons de Hidrogênio , Imunidade Humoral , Imunização , Vírus da Influenza A Subtipo H3N2/imunologia , Vacinas contra Influenza/genética , Vacinas contra Influenza/imunologia , Masculino , Mutação , Vírus Reordenados/genética , Vírus Reordenados/imunologia , Células Vero
19.
PLoS One ; 6(4): e18577, 2011 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-21490925

RESUMO

BACKGROUND: H5N1 influenza vaccines, including live intranasal, appear to be relatively less immunogenic compared to seasonal analogs. The main influenza virus surface glycoprotein hemagglutinin (HA) of highly pathogenic avian influenza viruses (HPAIV) was shown to be more susceptible to acidic pH treatment than that of human or low pathogenic avian influenza viruses. The acidification machinery of the human nasal passageway in response to different irritation factors starts to release protons acidifying the mucosal surface (down to pH of 5.2). We hypothesized that the sensitivity of H5 HA to the acidic environment might be the reason for the low infectivity and immunogenicity of intranasal H5N1 vaccines for mammals. METHODOLOGY/PRINCIPAL FINDINGS: We demonstrate that original human influenza viruses infect primary human nasal epithelial cells at acidic pH (down to 5.4), whereas H5N1 HPAIVs lose infectivity at pH ≤ 5.6. The HA of A/Vietnam/1203/04 was modified by introducing the single substitution HA2 58K→I, decreasing the pH of the HA conformational change. The H5N1 reassortants containing the indicated mutation displayed an increased resistance to acidic pH and high temperature treatment compared to those lacking modification. The mutation ensured a higher viral uptake as shown by immunohistochemistry in the respiratory tract of mice and 25 times lower mouse infectious dose50. Moreover, the reassortants keeping 58K→I mutation designed as a live attenuated vaccine candidate lacking an NS1 gene induced superior systemic and local antibody response after the intranasal immunization of mice. CONCLUSION/SIGNIFICANCE: Our finding suggests that an efficient intranasal vaccination with a live attenuated H5N1 virus may require a certain level of pH and temperature stability of HA in order to achieve an optimal virus uptake by the nasal epithelial cells and induce a sufficient immune response. The pH of the activation of the H5 HA protein may play a substantial role in the infectivity of HPAIVs for mammals.


Assuntos
Virus da Influenza A Subtipo H5N1/genética , Virus da Influenza A Subtipo H5N1/imunologia , Vacinas contra Influenza/genética , Vacinas contra Influenza/imunologia , Infecções por Orthomyxoviridae/genética , Infecções por Orthomyxoviridae/imunologia , Administração Intranasal , Animais , Chlorocebus aethiops , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Imunoglobulina A/sangue , Imunoglobulina G/sangue , Virus da Influenza A Subtipo H5N1/patogenicidade , Vacinas contra Influenza/administração & dosagem , Camundongos , Mutação , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Células Vero
20.
Cancer Biol Ther ; 10(6): 592-9, 2010 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-20647775

RESUMO

Previously we have developed a prototype for conditionally replicating oncolytic influenza A virus which is based on deletions in the non-structural (NS1) protein. Multi-cycle replication of influenza A virus in malignant tissue is critically dependent on a protease which cleaves the viral entry protein. Here we demonstrate that the malignant colon cancer cell lines Caco-2, HT-29 and SW-620 can endogenously provide a virus-activating protease, which allows lytic multi-cycle replication of NS1 deletion viruses in those cancer cells in vitro. The oncolytic potency of an influenza NS1 deletion virus (NS1-80) was further tested in SCID mice bearing HT-29 derived tumors. The intra-tumoral injection of live, but not of heat inactivated NS1-80 virus significantly inhibited progression of established tumors. We conclude that a selected set of human cancer expressing virus activating- proteases will be a preferred target for oncolytic tumor therapy using influenza A virus mutants.


Assuntos
Neoplasias do Colo/enzimologia , Vírus da Influenza A/metabolismo , Vírus Oncolíticos/metabolismo , Peptídeo Hidrolases/metabolismo , Proteínas não Estruturais Virais/metabolismo , Animais , Células CACO-2 , Linhagem Celular Tumoral , Chlorocebus aethiops , Neoplasias do Colo/patologia , Neoplasias do Colo/virologia , Feminino , Células HCT116 , Células HT29 , Interações Hospedeiro-Patógeno , Humanos , Vírus da Influenza A/genética , Vírus da Influenza A/fisiologia , Camundongos , Camundongos SCID , Mutação , Neoplasias Experimentais/enzimologia , Neoplasias Experimentais/patologia , Neoplasias Experimentais/virologia , Vírus Oncolíticos/genética , Vírus Oncolíticos/fisiologia , Infecções por Orthomyxoviridae/enzimologia , Infecções por Orthomyxoviridae/patologia , Infecções por Orthomyxoviridae/virologia , Peptídeo Hidrolases/genética , Transplante Heterólogo , Células Vero , Proteínas não Estruturais Virais/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa