Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Virol ; 90(1): 611-5, 2016 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-26468526

RESUMO

We report the crystal structure of the M2 ectodomain (M2e) in complex with a monoclonal antibody that binds the amino terminus of M2. M2e extends into the antibody binding site to form an N-terminal ß-turn near the bottom of the paratope. This M2e folding differs significantly from that of M2e in complex with an antibody that binds another part of M2e. This suggests that M2e can adopt at least two conformations that can elicit protective antibodies.


Assuntos
Anticorpos Monoclonais/química , Anticorpos Antivirais/química , Proteínas da Matriz Viral/química , Animais , Anticorpos Monoclonais/isolamento & purificação , Anticorpos Monoclonais/metabolismo , Anticorpos Antivirais/isolamento & purificação , Anticorpos Antivirais/metabolismo , Linhagem Celular , Cristalografia por Raios X , Humanos , Camundongos Endogâmicos BALB C , Ligação Proteica , Conformação Proteica , Proteínas da Matriz Viral/metabolismo
2.
J Virol ; 89(21): 10879-90, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26292322

RESUMO

UNLABELLED: The interferon-induced Mx1 gene is an important part of the mammalian defense against influenza viruses. Mus musculus Mx1 inhibits influenza A virus replication and transcription by suppressing the polymerase activity of viral ribonucleoproteins (vRNPs). Here, we compared the anti-influenza virus activity of Mx1 from Mus musculus A2G with that of its ortholog from Mus spretus. We found that the antiviral activity of M. spretus Mx1 was less potent than that of M. musculus Mx1. Comparison of the M. musculus Mx1 sequence with the M. spretus Mx1 sequence revealed 25 amino acid differences, over half of which were present in the GTPase domain and 2 of which were present in loop L4. However, the in vitro GTPase activity of Mx1 from the two mouse species was similar. Replacement of one of the residues in loop L4 in M. spretus Mx1 by the corresponding residue of A2G Mx1 increased its antiviral activity. We also show that deletion of loop L4 prevented the binding of Mx1 to influenza A virus nucleoprotein and, hence, abolished the antiviral activity of mouse Mx1. These results indicate that loop L4 of mouse Mx1 is a determinant of antiviral activity. Our findings suggest that Mx proteins from different mammals use a common mechanism to inhibit influenza A viruses. IMPORTANCE: Mx proteins are evolutionarily conserved in vertebrates and inhibit a wide range of viruses. Still, the exact details of their antiviral mechanisms remain largely unknown. Functional comparison of the Mx genes from two species that diverged relatively recently in evolution can provide novel insights into these mechanisms. We show that both Mus musculus A2G Mx1 and Mus spretus Mx1 target the influenza virus nucleoprotein. We also found that loop L4 in mouse Mx1 is crucial for its antiviral activity, as was recently reported for primate MxA. This indicates that human and mouse Mx proteins, which have diverged by 75 million years of evolution, recognize and inhibit influenza A viruses by a common mechanism.


Assuntos
Antivirais/imunologia , Vírus da Influenza A/efeitos dos fármacos , Proteínas de Resistência a Myxovirus/genética , Proteínas de Resistência a Myxovirus/imunologia , Sequência de Aminoácidos , Animais , Antivirais/farmacologia , Sequência de Bases , Citometria de Fluxo , Vetores Genéticos/genética , Células HEK293 , Humanos , Imunoprecipitação , Camundongos , Microscopia de Fluorescência , Dados de Sequência Molecular , Proteínas de Resistência a Myxovirus/farmacologia , Nucleoproteínas/metabolismo , Ligação Proteica , Conformação Proteica , Análise de Regressão , Análise de Sequência de DNA , Especificidade da Espécie
3.
J Virol ; 89(7): 3700-11, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25609808

RESUMO

UNLABELLED: The extracellular domain of influenza A virus matrix protein 2 (M2e) is conserved and is being evaluated as a quasiuniversal influenza A vaccine candidate. We describe the crystal structure at 1.6 Å resolution of M2e in complex with the Fab fragment of an M2e-specific monoclonal antibody that protects against influenza A virus challenge. This antibody binds M2 expressed on the surfaces of cells infected with influenza A virus. Five out of six complementary determining regions interact with M2e, and three highly conserved M2e residues are critical for this interaction. In this complex, M2e adopts a compact U-shaped conformation stabilized in the center by the highly conserved tryptophan residue in M2e. This is the first description of the three-dimensional structure of M2e. IMPORTANCE: M2e of influenza A is under investigation as a universal influenza A vaccine, but its three-dimensional structure is unknown. We describe the structure of M2e stabilized with an M2e-specific monoclonal antibody that recognizes natural M2. We found that the conserved tryptophan is positioned in the center of the U-shaped structure of M2e and stabilizes its conformation. The structure also explains why previously reported in vivo escape viruses, selected with a similar monoclonal antibody, carried proline residue substitutions at position 10 in M2.


Assuntos
Proteínas da Matriz Viral/química , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais/isolamento & purificação , Anticorpos Monoclonais/metabolismo , Anticorpos Antivirais/química , Anticorpos Antivirais/isolamento & purificação , Anticorpos Antivirais/metabolismo , Cristalografia por Raios X , Fragmentos Fab das Imunoglobulinas/química , Fragmentos Fab das Imunoglobulinas/isolamento & purificação , Fragmentos Fab das Imunoglobulinas/metabolismo , Camundongos Endogâmicos BALB C , Ligação Proteica , Conformação Proteica
4.
J Virol ; 88(15): 8278-96, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24829341

RESUMO

UNLABELLED: Influenza virus neuraminidase (NA) is an interesting target of small-molecule antiviral drugs. We isolated a set of H5N1 NA-specific single-domain antibodies (N1-VHHm) and evaluated their in vitro and in vivo antiviral potential. Two of them inhibited the NA activity and in vitro replication of clade 1 and 2 H5N1 viruses. We then generated bivalent derivatives of N1-VHHm by two methods. First, we made N1-VHHb by genetically joining two N1-VHHm moieties with a flexible linker. Second, bivalent N1-VHH-Fc proteins were obtained by genetic fusion of the N1-VHHm moiety with the crystallizable region of mouse IgG2a (Fc). The in vitro antiviral potency against H5N1 of both bivalent N1-VHHb formats was 30- to 240-fold higher than that of their monovalent counterparts, with 50% inhibitory concentrations in the low nanomolar range. Moreover, single-dose prophylactic treatment with bivalent N1-VHHb or N1-VHH-Fc protected BALB/c mice against a lethal challenge with H5N1 virus, including an oseltamivir-resistant H5N1 variant. Surprisingly, an N1-VHH-Fc fusion without in vitro NA-inhibitory or antiviral activity also protected mice against an H5N1 challenge. Virus escape selection experiments indicated that one amino acid residue close to the catalytic site is required for N1-VHHm binding. We conclude that single-domain antibodies directed against influenza virus NA protect against H5N1 virus infection, and when engineered with a conventional Fc domain, they can do so in the absence of detectable NA-inhibitory activity. IMPORTANCE: Highly pathogenic H5N1 viruses are a zoonotic threat. Outbreaks of avian influenza caused by these viruses occur in many parts of the world and are associated with tremendous economic loss, and these viruses can cause very severe disease in humans. In such cases, small-molecule inhibitors of the viral NA are among the few treatment options for patients. However, treatment with such drugs often results in the emergence of resistant viruses. Here we show that single-domain antibody fragments that are specific for NA can bind and inhibit H5N1 viruses in vitro and can protect laboratory mice against a challenge with an H5N1 virus, including an oseltamivir-resistant virus. In addition, plant-produced VHH fused to a conventional Fc domain can protect in vivo even in the absence of NA-inhibitory activity. Thus, NA of influenza virus can be effectively targeted by single-domain antibody fragments, which are amenable to further engineering.


Assuntos
Antivirais/uso terapêutico , Virus da Influenza A Subtipo H5N1/efeitos dos fármacos , Neuraminidase/antagonistas & inibidores , Infecções por Orthomyxoviridae/prevenção & controle , Anticorpos de Domínio Único/uso terapêutico , Animais , Antivirais/imunologia , Modelos Animais de Doenças , Feminino , Virus da Influenza A Subtipo H5N1/imunologia , Concentração Inibidora 50 , Camundongos , Camundongos Endogâmicos BALB C , Testes de Sensibilidade Microbiana , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/virologia , Anticorpos de Domínio Único/imunologia , Resultado do Tratamento
5.
J Virol ; 87(6): 3314-23, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23302879

RESUMO

Respiratory syncytial virus (RSV) is the leading cause of lower respiratory tract infections in infants worldwide. Despite decades of research, there is still no registered vaccine available for this major pathogen. We investigated the protective efficacy of a recombinant influenza virus, PR8/NA-F(85-93), that carries the RSV CD8(+) T cell epitope F(85-93) in its neuraminidase stalk. F(85-93)-specific cytotoxic T lymphocytes (CTLs) were induced in mice after a single intranasal immunization with PR8/NA-F(85-93) virus, and these CTLs provided a significant reduction in the lung viral load upon a subsequent challenge with RSV. To avoid influenza-induced morbidity, we treated mice with matrix protein 2 (M2e)-specific monoclonal antibodies before PR8/NA-F(85-93) virus infection. Treatment with anti-M2e antibodies reduced the infiltration of immune cells in the lungs upon PR8/NA-F(85-93) infection, whereas the formation of inducible bronchus-associated lymphoid tissue was not affected. Moreover, this treatment prevented body weight loss yet still permitted the induction of RSV F-specific T cell responses and significantly reduced RSV replication upon challenge. These results demonstrate that it is possible to take advantage of the infection-permissive protection of M2e-specific antibodies against influenza A virus to induce heterologous CD8(+) T cell-mediated immunity by an influenza A virus vector expressing the RSV F(85-93) epitope.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Epitopos de Linfócito T/imunologia , Vírus da Influenza A/genética , Vírus Sinciciais Respiratórios/genética , Vírus Sinciciais Respiratórios/imunologia , Proteínas Virais de Fusão/imunologia , Replicação Viral , Animais , Peso Corporal , Epitopos de Linfócito T/genética , Feminino , Pulmão/virologia , Camundongos , Camundongos Endogâmicos BALB C , Neuraminidase/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Recombinação Genética , Infecções por Vírus Respiratório Sincicial/patologia , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sinciciais Respiratórios/fisiologia , Proteínas Virais de Fusão/genética , Proteínas Virais/genética
6.
J Virol ; 86(24): 13445-55, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23015724

RESUMO

Mx1 is a GTPase that is part of the antiviral response induced by type I and type III interferons in the infected host. It inhibits influenza virus infection by blocking viral transcription and replication, but the molecular mechanism is not known. Polymerase basic protein 2 (PB2) and nucleoprotein (NP) were suggested to be the possible target of Mx1, but a direct interaction between Mx1 and any of the viral proteins has not been reported. We investigated the interplay between Mx1, NP, and PB2 to identify the mechanism of Mx1's antiviral activity. We found that Mx1 inhibits the PB2-NP interaction, and the strength of this inhibition correlated with a decrease in viral polymerase activity. Inhibition of the PB2-NP interaction is an active process requiring enzymatically active Mx1. We also demonstrate that Mx1 interacts with the viral proteins NP and PB2, which indicates that Mx1 protein has a direct effect on the viral ribonucleoprotein complex. In a minireplicon system, avian-like NP from swine virus isolates was more sensitive to inhibition by murine Mx1 than NP from human influenza A virus isolates. Likewise, murine Mx1 displaced avian NP from the viral ribonucleoprotein complex more easily than human NP. The stronger resistance of the A/H1N1 pandemic 2009 virus against Mx1 also correlated with reduced inhibition of the PB2-NP interaction. Our findings support a model in which Mx1 interacts with the influenza ribonucleoprotein complex and interferes with its assembly by disturbing the PB2-NP interaction.


Assuntos
Proteínas de Ligação ao GTP/fisiologia , Vírus da Influenza A Subtipo H1N1/fisiologia , Ribonucleoproteínas/fisiologia , Proteínas Virais/fisiologia , Montagem de Vírus/fisiologia , Sequência de Aminoácidos , Proteínas de Ligação ao GTP/química , Células HEK293 , Humanos , Dados de Sequência Molecular , Proteínas de Resistência a Myxovirus , Homologia de Sequência de Aminoácidos
7.
J Immunol ; 186(2): 1022-31, 2011 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-21169548

RESUMO

The ectodomain of matrix protein 2 (M2e) of influenza A virus is an attractive target for a universal influenza A vaccine: the M2e sequence is highly conserved across influenza virus subtypes, and induced humoral anti-M2e immunity protects against a lethal influenza virus challenge in animal models. Clinical phase I studies with M2e vaccine candidates have been completed. However, the in vivo mechanism of immune protection induced by M2e-carrier vaccination is unclear. Using passive immunization experiments in wild-type, FcRγ(-/-), FcγRI(-/-), FcγRIII(-/-), and (FcγRI, FcγRIII)(-/-) mice, we report in this study that Fc receptors are essential for anti-M2e IgG-mediated immune protection. M2e-specific IgG1 isotype Abs are shown to require functional FcγRIII for in vivo immune protection but other anti-M2e IgG isotypes can rescue FcγRIII(-/-) mice from a lethal challenge. Using a conditional cell depletion protocol, we also demonstrate that alveolar macrophages (AM) play a crucial role in humoral M2e-specific immune protection. Additionally, we show that adoptive transfer of wild-type AM into (FcγRI, FcγRIII)(-/-) mice restores protection by passively transferred anti-M2e IgG. We conclude that AM and Fc receptor-dependent elimination of influenza A virus-infected cells are essential for protection by anti-M2e IgG.


Assuntos
Imunoglobulina G/metabolismo , Vírus da Influenza A/imunologia , Vacinas contra Influenza/imunologia , Macrófagos Alveolares/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Domínios e Motivos de Interação entre Proteínas/imunologia , Receptores Fc/fisiologia , Proteínas da Matriz Viral/imunologia , Animais , Morte Celular/genética , Morte Celular/imunologia , Citotoxicidade Imunológica , Modelos Animais de Doenças , Feminino , Imunização Passiva , Imunoglobulina G/toxicidade , Vírus da Influenza A/genética , Vacinas contra Influenza/genética , Vacinas contra Influenza/uso terapêutico , Depleção Linfocítica/métodos , Macrófagos Alveolares/patologia , Macrófagos Alveolares/virologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/patologia , Domínios e Motivos de Interação entre Proteínas/genética , Receptores Fc/deficiência , Receptores Fc/uso terapêutico , Receptores de IgG/deficiência , Receptores de IgG/metabolismo , Receptores de IgG/fisiologia , Proteínas da Matriz Viral/genética , Proteínas da Matriz Viral/uso terapêutico
8.
J Gen Virol ; 92(Pt 2): 301-6, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20965983

RESUMO

The recent emergence and rapid spread of the pandemic H1N1 swine influenza virus reminded us once again of the need for a universal influenza vaccine that can elicit heterosubtypic protection. Here, we show the superior immunogenicity and immunoprotective capacity of the full-length matrix protein 2 ectodomain (M2e) peptide coupled to keyhole limpet haemocyanin (KLH) compared with the N-terminal 9 aa residues of M2e (SP1). Immunization with M2e-KLH protected mice against a lethal challenge with influenza A virus and significantly reduced weight loss and lung virus titres. In addition, passive transfer of serum raised in rabbits against M2e-KLH protected mice against a lethal influenza virus challenge, whereas serum from rabbits immunized with SP1-KLH did not. Nevertheless, immunofluorescence staining revealed that rabbit serum raised against SP1-KLH bound specifically to infected Madin-Darby canine kidney cells. We conclude that the peptide SP1 contains an immunogenic epitope that is not sufficient for immunoprotection.


Assuntos
Soros Imunes/imunologia , Vírus da Influenza A/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Infecções por Orthomyxoviridae/virologia , Proteínas da Matriz Viral/classificação , Proteínas da Matriz Viral/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Antivirais/imunologia , Linhagem Celular , Cães , Regulação Viral da Expressão Gênica , Hemocianinas , Vírus da Influenza A/genética , Vírus da Influenza A/metabolismo , Vacinas contra Influenza/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Coelhos , Proteínas da Matriz Viral/química , Proteínas da Matriz Viral/genética
9.
J Biomed Biotechnol ; 2010: 863985, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20508820

RESUMO

Influenza is a vaccine preventable disease that causes severe illness and excess mortality in humans. Licensed influenza vaccines induce humoral immunity and protect against strains that antigenically match the major antigenic components of the vaccine, but much less against antigenically diverse influenza strains. A vaccine that protects against different influenza viruses belonging to the same subtype or even against viruses belonging to more than one subtype would be a major advance in our battle against influenza. Heterosubtypic immunity could be obtained by cytotoxic T-cell (CTL) responses against conserved influenza virus epitopes. The molecular mechanisms involved in inducing protective CTL responses are discussed here. We also focus on CTL vaccine design and point to the importance of immune-related databases and immunoinformatics tools in the quest for new vaccine candidates. Some techniques for analysis of T-cell responses are also highlighted, as they allow estimation of cellular immune responses induced by vaccine preparations and can provide correlates of protection.

10.
Drug News Perspect ; 22(2): 80-92, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19330167

RESUMO

The possible emergence of a new influenza pandemic is considered a major threat for human health worldwide. Pandemics start by the introduction or reintroduction and spread in the human population of an influenza virus subtype against which almost nobody has protective immunity. Currently used influenza vaccines provide good protection only against antigenically matching influenza strains. However, neither the timing nor the subtype of the next pandemic virus is known. Therefore, different approaches are being pursued in anticipation of this pandemic threat. In the present article, we review approaches that aim to induce heterosubtypic immunity, that is, protection against challenge with influenza A viruses belonging to two or more subtypes. Experimental and epidemiological studies indicate that natural infection can provide some heterosubtypic immunity, possibly involving cellular immune responses directed against matrix and/or nucleoprotein as well as humoral responses against neuraminidase. Other approaches have focused on the use of conserved epitopes of the viral proteins, including matrix protein 2 ectodomain (M2e) and nucleoprotein (NP). Proof-of-concept of protection by these novel vaccines has been obtained in animal models, and promising results from several clinical trials have recently been reported. Demonstrating the efficacy of these new vaccines against a potential pandemic influenza endowed with human transmissibility remains a major challenge.


Assuntos
Surtos de Doenças/prevenção & controle , Hemaglutininas Virais/imunologia , Vacinas contra Influenza/imunologia , Vacinas contra Influenza/uso terapêutico , Influenza Humana/prevenção & controle , Proteínas da Matriz Viral/imunologia , Sequência de Aminoácidos , Animais , Defesa Civil/métodos , Ensaios Clínicos como Assunto , Epitopos , Humanos , Vírus da Influenza A/imunologia , Vacinas contra Influenza/classificação , Dados de Sequência Molecular , Linfócitos T Citotóxicos/imunologia
11.
Expert Rev Vaccines ; 16(2): 123-136, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27653543

RESUMO

INTRODUCTION: Current influenza vaccines can prevent disease caused by influenza viruses but require annual administration and almost yearly reformulation. An attractive alternative approach would be to use a vaccine that provides broad and, ideally, lifelong protection against all influenza A and B virus strains. The extracellular domain of matrix protein 2 (M2e) of influenza A viruses is conserved and thus fits well in such a broadly protective vaccine. Areas covered: Recent advances in M2e vaccine design, the mode of action of M2e-based immunity and clinical progress of M2-based influenza vaccines. Expert commentary: Many M2e vaccine have been successfully tested for efficacy against a panel of divergent influenza viruses in animal models. More recently, clinical studies have been conducted with M2e vaccine candidates, which demonstrated their safety and immunogenicity in humans. Efficacy studies in humans are still needed to provide evidence that an M2e-based vaccine can protect against human influenza.


Assuntos
Vacinas contra Influenza/imunologia , Proteínas da Matriz Viral/imunologia , Animais , Ensaios Clínicos como Assunto , Modelos Animais de Doenças , Descoberta de Drogas/tendências , Avaliação Pré-Clínica de Medicamentos , Humanos , Vacinas contra Influenza/efeitos adversos , Vacinas contra Influenza/genética , Vacinas Sintéticas/efeitos adversos , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Proteínas da Matriz Viral/genética
12.
J Control Release ; 264: 55-65, 2017 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-28842314

RESUMO

We describe a novel live oral vaccine type. Conceptually, this vaccine is based on a non-lytic, recombinant filamentous bacteriophage that displays an antigen of interest. To provide proof of concept we used the amino-terminal part of a conserved influenza A virus epitope, i.e. matrix protein 2 ectodomain (M2e) residues 2 to 16, as the antigen of interest. Rather than using the phages as purified virus-like particles as a vaccine, these phages were delivered to intestinal Peyer's patches as a live bacterium-phage combination that comprises Escherichia coli cells that conditionally express invasin derived from Yersinia pseudotuberculosis. Invasin-expressing E. coli cells were internalized by mammalian Hep-2 cells in vitro and adhered to mouse intestinal microfold (M) cells ex vivo. Invasin-expressing E. coli cells were permissive for recombinant filamentous bacteriophage f88 that displays M2e and became persistently infected. Oral administration of the live engineered E. coli-invasin-phage combination to mice induced M2e-specific serum IgG antibodies. Mice that had been immunized with invasin-expressing E. coli cells that carried M2e2-16 displaying fd phages seroconverted to M2e and showed partial protection against challenge with influenza A virus. Oral delivery of a live vaccine comprising a bacterial host that is targeted to Peyer's patches and is persistently infected with an antigen-displaying phage, can thus be exploited as an oral vaccine.


Assuntos
Antígenos/imunologia , Bacteriófagos/imunologia , Escherichia coli/virologia , Vírus da Influenza A/imunologia , Vacinas contra Influenza , Proteínas da Matriz Viral/imunologia , Adesinas Bacterianas/imunologia , Administração Oral , Animais , Linhagem Celular Tumoral , Escherichia coli/imunologia , Feminino , Humanos , Imunoglobulina G/sangue , Camundongos Endogâmicos BALB C , Nódulos Linfáticos Agregados/microbiologia , Domínios Proteicos/imunologia
13.
Antiviral Res ; 141: 155-164, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28257797

RESUMO

The ectodomain of the influenza A matrix protein 2 (M2e) is highly conserved amongst all influenza virus A subtypes. M2e is present on the surface of influenza A virus-infected cells, and therefore a suitable target for broadly protective therapies. We designed bispecific T cell engaging (BiTE®) antibody constructs specific for M2e by genetically fusing a single chain variable fragment (scFv) derived from an M2e-specific murine monoclonal antibody with a CD3ɛ-specific scFv. These so-called FLU BiTE® antibody constructs selectively mediate T cell dependent lysis of M2-expressing and influenza A virus infected cells and protect BALB/c mice against challenge with different influenza A virus subtypes. By humanizing the M2e-binding scFv, we generated human-like FLU BiTE® antibody constructs, with increased in vitro cytotoxic activity and in vivo protective capacity against influenza A virus infection. FLU BiTE® antibody constructs represent a promising new curative and prophylactic treatment option for influenza disease.


Assuntos
Anticorpos Biespecíficos/imunologia , Vírus da Influenza A/química , Vírus da Influenza A/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Linfócitos T/imunologia , Proteínas da Matriz Viral/imunologia , Animais , Anticorpos Biespecíficos/administração & dosagem , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais Humanizados/administração & dosagem , Anticorpos Monoclonais Humanizados/imunologia , Anticorpos Antivirais/sangue , Testes Imunológicos de Citotoxicidade , Memória Imunológica , Vacinas contra Influenza/administração & dosagem , Camundongos
14.
Sci Rep ; 6: 24402, 2016 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-27072615

RESUMO

There is mounting evidence that in the absence of neutralizing antibodies cross-reactive T cells provide protection against pandemic influenza viruses. Here, we compared protection and CD8+ T cell responses following challenge with H1N1 2009 pandemic and H3N2 viruses of mice that had been immunized with hemagglutinin (HA), neuraminidase (NA) and the extracellular domain of matrix protein 2 (M2e) fused to a virus-like particle (VLP). Mice were challenged a first time with a sublethal dose of H1N1 2009 pandemic virus and, four weeks later, challenged again with an H3N2 virus. Mice that had been vaccinated with HA, NA, NA + M2e-VLP and HA + NA + M2e-VLP were protected against homologous H1N1 virus challenge. Challenged NA and NA + M2e-VLP vaccinated mice mounted CD8+ T cell responses that correlated with protection against secondary H3N2 challenge. HA-vaccinated mice were fully protected against challenge with homologous H1N1 2009 virus, failed to mount cross-reactive CD8+ T cells and succumbed to the second challenge with heterologous H3N2 virus. In summary, NA- and M2e-based immunity can protect against challenge with (homologous) virus without compromising the induction of robust cross-reactive CD8+ T cell responses upon exposure to virus.


Assuntos
Vírus da Influenza A/imunologia , Influenza Humana/prevenção & controle , Neuraminidase/imunologia , Proteínas da Matriz Viral/imunologia , Animais , Anticorpos Neutralizantes/imunologia , Linfócitos T CD8-Positivos/imunologia , Reações Cruzadas , Feminino , Humanos , Vírus da Influenza A/fisiologia , Vacinas contra Influenza/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Replicação Viral
15.
Vaccines (Basel) ; 3(1): 105-36, 2015 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-26344949

RESUMO

The successful isolation of a human influenza virus in 1933 was soon followed by the first attempts to develop an influenza vaccine. Nowadays, vaccination is still the most effective method to prevent human influenza disease. However, licensed influenza vaccines offer protection against antigenically matching viruses, and the composition of these vaccines needs to be updated nearly every year. Vaccines that target conserved epitopes of influenza viruses would in principle not require such updating and would probably have a considerable positive impact on global human health in case of a pandemic outbreak. The extracellular domain of Matrix 2 (M2e) protein is an evolutionarily conserved region in influenza A viruses and a promising epitope for designing a universal influenza vaccine. Here we review the seminal and recent studies that focused on M2e as a vaccine antigen. We address the mechanism of action and the clinical development of M2e-vaccines. Finally, we try to foresee how M2e-based vaccines could be implemented clinically in the future.

16.
PLoS One ; 10(3): e0121491, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25816132

RESUMO

The severity of influenza-related illness is mediated by many factors, including in vivo cell tropism, timing and magnitude of the immune response, and presence of pre-existing immunity. A direct way to study cell tropism and virus spread in vivo is with an influenza virus expressing a reporter gene. However, reporter gene-expressing influenza viruses are often attenuated in vivo and may be genetically unstable. Here, we describe the generation of an influenza A virus expressing GFP from a tri-cistronic NS segment. To reduce the size of this engineered gene segment, we used a truncated NS1 protein of 73 amino acids combined with a heterologous dimerization domain to increase protein stability. GFP and nuclear export protein coding information were fused in frame with the truncated NS1 open reading frame and separated from each other by 2A self-processing sites. The resulting PR8-NS1(1-73)GFP virus was successfully rescued and replicated as efficiently as the parental PR8 virus in vitro and was slightly attenuated in vivo. Flow cytometry-based monitoring of cells isolated from PR8-NS1(1-73)GFP virus infected BALB/c mice revealed that GFP expression peaked on day two in all cell types tested. In particular respiratory epithelial cells and myeloid cells known to be involved in antigen presentation, including dendritic cells (CD11c+) and inflammatory monocytes (CD11b+ GR1+), became GFP positive following infection. Prophylactic treatment with anti-M2e monoclonal antibody or oseltamivir reduced GFP expression in all cell types studied, demonstrating the usefulness of this reporter virus to analyze the efficacy of antiviral treatments in vivo. Finally, deep sequencing analysis, serial in vitro passages and ex vivo analysis of PR8-NS1(1-73)GFP virus, indicate that this virus is genetically and phenotypically stable.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Proteínas de Fluorescência Verde/metabolismo , Vírus da Influenza A/fisiologia , Infecções por Orthomyxoviridae/prevenção & controle , Proteínas não Estruturais Virais/metabolismo , Tropismo Viral/efeitos dos fármacos , Animais , Anticorpos Monoclonais/uso terapêutico , Antivirais/administração & dosagem , Antivirais/farmacologia , Células Cultivadas , Células Dendríticas/metabolismo , Células Dendríticas/virologia , Cães , Proteínas de Fluorescência Verde/genética , Vírus da Influenza A/efeitos dos fármacos , Vírus da Influenza A/genética , Células Madin Darby de Rim Canino , Camundongos , Monócitos/metabolismo , Monócitos/virologia , Infecções por Orthomyxoviridae/metabolismo , Infecções por Orthomyxoviridae/virologia , Oseltamivir/administração & dosagem , Oseltamivir/farmacologia , Estabilidade Proteica , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas da Matriz Viral/antagonistas & inibidores , Proteínas não Estruturais Virais/genética , Replicação Viral/efeitos dos fármacos
17.
PLoS One ; 10(5): e0126650, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25973787

RESUMO

Human influenza viruses are responsible for annual epidemics and occasional pandemics that cause severe illness and mortality in all age groups worldwide. Matrix protein 2 (M2) of influenza A virus is a tetrameric type III membrane protein that functions as a proton-selective channel. The extracellular domain of M2 (M2e) is conserved in human and avian influenza A viruses and is being pursued as a component for a universal influenza A vaccine. To develop a M2e vaccine that is economical and easy to purify, we genetically fused M2e amino acids 2-16 to the N-terminus of pVIII, the major coat protein of filamentous bacteriophage f88. We show that the resulting recombinant f88-M2e2-16 phages are replication competent and display the introduced part of M2e on the phage surface. Immunization of mice with purified f88-M2e2-16 phages in the presence of incomplete Freund's adjuvant, induced robust M2e-specific serum IgG and protected BALB/c mice against challenge with human and avian influenza A viruses. Thus, replication competent filamentous bacteriophages can be used as efficient and economical carriers to display conserved B cell epitopes of influenza A.


Assuntos
Proteínas do Capsídeo/genética , Colífagos/imunologia , Vírus da Influenza A Subtipo H1N1/imunologia , Proteínas da Matriz Viral/genética , Sequência de Aminoácidos , Animais , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Antígenos Virais/biossíntese , Antígenos Virais/imunologia , Proteínas do Capsídeo/química , Colífagos/isolamento & purificação , Colífagos/fisiologia , Ensaio de Imunoadsorção Enzimática , Feminino , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Vírus da Influenza A Subtipo H1N1/metabolismo , Vacinas contra Influenza/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Infecções por Orthomyxoviridae/prevenção & controle , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Proteínas da Matriz Viral/química , Replicação Viral
18.
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
19.
PLoS One ; 8(3): e59198, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23555631

RESUMO

BACKGROUND: Intranasal delivery of vaccines directed against respiratory pathogens is an attractive alternative to parenteral administration. However, using this delivery route for inactivated vaccines usually requires the use of potent mucosal adjuvants, and no such adjuvant has yet been approved for human use. METHODOLOGY/PRINCIPAL FINDINGS: We have developed a live attenuated Bordetella pertussis vaccine, called BPZE1, and show here that it can be used to present the universal influenza virus epitope M2e to the mouse respiratory tract to prime for protective immunity against viral challenge. Three copies of M2e were genetically fused to the N-terminal domain of filamentous hemagglutinin (FHA) and produced in recombinant BPZE1 derivatives in the presence or absence of endogenous full-length FHA. Only in the absence of FHA intranasal administration of the recombinant BPZE1 derivative induced antibody responses to M2e and effectively primed BALB/c mice for protection against influenza virus-induced mortality and reduced the viral load after challenge. Strong M2e-specific antibody responses and protection were observed after a single nasal administration with the recombinant BPZE1 derivative, followed by a single administration of M2e linked to a virus-like particle without adjuvant, whereas priming alone with the vaccine strain did not protect. CONCLUSIONS/SIGNIFICANCE: Using recombinant FHA-3M2e-producing BPZE1 derivatives for priming and the universal influenza M2e peptide linked to virus-like particles for boosting may constitute a promising approach for needle-free and adjuvant-free nasal vaccination against influenza.


Assuntos
Adesinas Bacterianas/imunologia , Anticorpos Antivirais/sangue , Bordetella pertussis/imunologia , Vacinas contra Influenza/imunologia , Influenza Humana/prevenção & controle , Proteínas Recombinantes de Fusão/imunologia , Proteínas da Matriz Viral/imunologia , Fatores de Virulência de Bordetella/imunologia , Adesinas Bacterianas/genética , Administração Intranasal , Animais , Bordetella pertussis/genética , Humanos , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/genética , Influenza Humana/imunologia , Influenza Humana/virologia , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/genética , Sistema Respiratório/efeitos dos fármacos , Sistema Respiratório/imunologia , Sistema Respiratório/virologia , Análise de Sobrevida , Vacinação , Vacinas Sintéticas , Proteínas da Matriz Viral/genética , Fatores de Virulência de Bordetella/genética
20.
PLoS One ; 8(3): e59081, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23527091

RESUMO

The ectodomain of influenza A matrix protein 2 (M2e) is a candidate for a universal influenza A vaccine. We used recombinant Hepatitis B core antigen to produce virus-like particles presenting M2e (M2e-VLPs). We produced the VLPs with and without entrapped nucleic acids and compared their immunogenicity and protective efficacy. Immunization of BALB/c mice with M2e-VLPs containing nucleic acids induced a stronger, Th1-biased antibody response compared to particles lacking nucleic acids. The former also induced a stronger M2e-specific CD4(+) T cell response, as determined by ELISPOT. Mice vaccinated with alum-adjuvanted M2e-VLPs containing the nucleic acid-binding domain were better protected against influenza A virus challenge than mice vaccinated with similar particles lacking this domain, as deduced from the loss in body weight following challenge with X47 (H3N2) or PR/8 virus. Challenge of mice that had been immunized with M2e-VLPs with or without nucleic acids displayed significantly lower mortality, morbidity and lung virus titers than control-immunized groups. We conclude that nucleic acids present in M2e-VLPs correlate with improved immune protection.


Assuntos
Imunidade Adaptativa , Vírus da Influenza A Subtipo H3N2/imunologia , Vacinas contra Influenza/imunologia , RNA/metabolismo , Células Th1/imunologia , Vacinas de Partículas Semelhantes a Vírus/imunologia , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Feminino , Humanos , Imunidade Celular , Vacinas contra Influenza/metabolismo , Influenza Humana/imunologia , Influenza Humana/prevenção & controle , Pulmão/imunologia , Pulmão/patologia , Pulmão/virologia , Camundongos , Fator 88 de Diferenciação Mieloide/metabolismo , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Transdução de Sinais , Vacinação , Vacinas de Partículas Semelhantes a Vírus/metabolismo , Carga Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA