Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Acta Biochim Biophys Sin (Shanghai) ; 49(1): 74-82, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27974288

RESUMO

Respiratory syncytial virus (RSV) is a leading cause of lower respiratory infection in infants and children, but there is still no licensed vaccine available. In this report, we developed virus-like particle (VLP) vaccines based on the Bac-to-Bac baculovirus expression system, consisting of an influenza virus matrix (M1) protein and the RSV fusion protein (F) or glycoprotein (G). These RSV VLPs were identified by western blot analysis and electron microscopy. Female BALB/c mice immunized intranasally (i.n.) with RSV-F VLPs, RSV-G VLPs, or both showed viral-specific antibody responses against RSV. Total IgG, IgG1, IgG2a, and mucosal IgA were detected in mice with RSV-F plus RSV-G VLPs, revealing potent cellular and mucosal immune responses. Moreover, we found that these mixed RSV VLPs conferred enhanced protection against live RSV challenges, showing significant decreases in lung viral replication and obvious attenuation of histopathological changes associated with viral infections. These results demonstrate that RSV-F plus RSV-G VLPs by intranasal vaccination is a promising vaccine candidate that warrants further evaluation using cotton rat and primate models.


Assuntos
Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vacinas de Partículas Semelhantes a Vírus/administração & dosagem , Administração Intranasal , Animais , Feminino , Imunidade Celular , Imunidade nas Mucosas , Camundongos , Camundongos Endogâmicos BALB C , Infecções por Vírus Respiratório Sincicial/imunologia , Vacinas de Partículas Semelhantes a Vírus/imunologia
2.
J Gen Virol ; 95(Pt 9): 1886-1891, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24914066

RESUMO

Respiratory syncytial virus (RSV) is the most common cause of respiratory infection in infants and the elderly, and no vaccine against this virus has yet been licensed. Here, we report a recombinant PR8 influenza virus with the RSV fusion (F) protein epitopes of the subgroup A gene inserted into the influenza virus non-structural (NS) gene (rFlu/RSV/F) that was generated as an RSV vaccine candidate. The rescued viruses were assessed by microscopy and Western blotting. The proper expression of NS1, the NS gene product, and the nuclear export protein (NEP) of rFlu/RSV/F was also investigated using an immunofluorescent assay. The rescued virus replicated well in the MDCK kidney cell line, A549 lung adenocarcinoma cell line and CNE-2Z nasopharyngeal carcinoma cell line. BALB/c mice immunized intranasally with rFlu/RSV/F had specific haemagglutination inhibition antibody responses against the PR8 influenza virus and RSV neutralization test proteins. Furthermore, intranasal immunization with rFlu/RSV/F elicited T helper type 1-dominant cytokine profiles against the RSV strain A2 virus. Taken together, our findings suggested that rFlu/RSV/F was immunogenic in vivo and warrants further development as a promising candidate vaccine.


Assuntos
Vírus da Influenza A/genética , Vírus Sinciciais Respiratórios/genética , Vírus Sinciciais Respiratórios/imunologia , Proteínas Virais de Fusão/imunologia , Administração Intranasal , Animais , Anticorpos Antivirais/imunologia , Células COS , Linhagem Celular Tumoral , Embrião de Galinha , Chlorocebus aethiops , Cães , Feminino , Técnicas de Transferência de Genes , Vetores Genéticos , Testes de Inibição da Hemaglutinação , Humanos , Imunização , Vírus da Influenza A/imunologia , Células Madin Darby de Rim Canino , Camundongos , Camundongos Endogâmicos BALB C , Testes de Neutralização , Infecções por Vírus Respiratório Sincicial/imunologia , Proteínas Virais de Fusão/biossíntese , Proteínas Virais de Fusão/genética , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/imunologia
3.
Clin Lab ; 60(5): 815-9, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24839825

RESUMO

BACKGROUND: Type B influenza virus is a major epidemic strain responsible for considerable mortality and morbidity. METHODS: A colloidal gold immunochromatographic strip for the rapid detection of human influenza B virus was developed. This test is based on membrane chromatography and uses colloidal gold conjugated with influenza B virus anti-NP monoclonal antibody as the tracer. The assembled test strip was housed in a plastic case. RESULTS: The colloid gold strip (CGS) specifically detected all influenza B viruses tested and did not react with other respiratory viruses. Compared with SYBR Green real-time PCR, the sensitivity and specificity of the CGS test was 89.76% and 99.56%, respectively, and the consistency ratio between CGS and real-time PCR was 96.06% in detecting influenza B virus in 710 nasopharyngeal swabs from patients with influenza-like illness in the hospital. CONCLUSIONS: The CGS array developed in this study enabled typing of influenza B viruses in human clinical specimens. Thus, together with the advantages of rapid detection and easy operation without requiring specialized personnel and equipment, this technique is a convenient and relatively inexpensive diagnostic tool for large-scale screening of clinical samples.


Assuntos
Cromatografia de Afinidade , Vírus da Influenza B/isolamento & purificação , Influenza Humana/diagnóstico , Influenza Humana/virologia , Adenovírus Humanos/isolamento & purificação , Adolescente , Adulto , Idoso , Criança , Cromatografia de Afinidade/métodos , Humanos , Vírus da Influenza A Subtipo H3N2/isolamento & purificação , Vírus da Influenza A/isolamento & purificação , Vírus da Influenza B/patogenicidade , Pessoa de Meia-Idade , Vírus Sinciciais Respiratórios/isolamento & purificação , Adulto Jovem
4.
Cell Death Differ ; 28(10): 2900-2915, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-33990776

RESUMO

Accumulating evidence has shown that long noncoding RNAs (lncRNAs) are involved in several biological processes, including immune responses. However, the role of lncRNAs in antiviral innate immune responses remains largely elusive. Here, we identify an uncharacterized human lncRNA AVAN from influenza A virus (IAV) infected patients, that is significantly upregulated following RNA virus infection. During IAV infection, AVAN play an indispensable role in antiviral immune responses. In vivo, we enforced the expression of AVAN in transgenic mice or adeno-associated virus encoding AVAN delivery system and found that AVAN significantly alleviated IAV virulence and virus replication. Mechanistically, nuclear AVAN positively regulates the transcription of forkhead box O3A (FOXO3a) by associating with its promoter and inducing chromatin remodeling to promote neutrophil chemotaxis. Meanwhile, cytoplasmic AVAN binds directly to the E3 ligase TRIM25 and enhances TRIM25-mediated K63-linked ubiquitination of RIG-I, thereby promoting TRIM25- and RIG-I-mediated antiviral innate immune responses, including the induction of type I interferon and ISGs. Moreover, AVAN binds to the B Box/CCD domain of TRIM25 and 1-200nt of AVAN were the functional moieties. Collectively, our findings highlight the potential clinical implications of human lncRNA AVAN as a key positive regulator of the antiviral innate immune response and a promising target for developing broad antiviral therapeutics.


Assuntos
Proteínas de Ligação a DNA/imunologia , Proteína Forkhead Box O3/imunologia , RNA Longo não Codificante/imunologia , Fatores de Transcrição/imunologia , Proteínas com Motivo Tripartido/imunologia , Ubiquitina-Proteína Ligases/imunologia , Células A549 , Animais , Feminino , Proteína Forkhead Box O3/genética , Humanos , Imunidade Inata , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , RNA Longo não Codificante/genética , Transdução de Sinais/imunologia , Transcrição Gênica , Regulação para Cima
5.
Biochem Biophys Res Commun ; 396(4): 973-7, 2010 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-20497905

RESUMO

The continued spread of a highly pathogenic avian influenza (HPAI) H5N1 virus among poultry and wild birds has posed a potential threat to human public health. An influenza pandemic happens, when a new subtype that has not previously circulated in humans emerges. Almost all of the influenza pandemics in history have originated from avian influenza viruses (AIV). Birds are significant reservoirs of influenza viruses. In the present study, we performed a survey of avian influenza virus in ostriches and H5N1 virus (A/Ostrich/SuZhou/097/03, China097) was isolated. This H5N1 virus is highly pathogenic to both chickens and mice. It is also able to replicate in the lungs of, and to cause death in, BALB/c mice following intranasal administration. It forms plaques in chicken embryo fibroblast (CEF) cells in the absence of trypsin. The hemagglutinin (HA) gene of the virus is genetically similar to A/Goose/Guangdong/1/96(H5N1) and belongs to clade 0. The HA sequence contains multiple basic amino acids adjacent to the cleavage site, a motif associated with HPAI viruses. More importantly, the existence of H5N1 isolates in ostriches highlights the potential threat of wild bird infections to veterinary and public health.


Assuntos
Virus da Influenza A Subtipo H5N1/patogenicidade , Influenza Aviária/epidemiologia , Influenza Aviária/virologia , Struthioniformes/virologia , Animais , Embrião de Galinha , Galinhas/virologia , China/epidemiologia , Humanos , Virus da Influenza A Subtipo H5N1/classificação , Virus da Influenza A Subtipo H5N1/isolamento & purificação , Influenza Humana/epidemiologia , Influenza Humana/virologia , Pulmão/virologia , Camundongos , Camundongos Endogâmicos BALB C , Filogenia , Replicação Viral
6.
Vaccine ; 37(29): 3810-3819, 2019 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-31147275

RESUMO

Staphylococcal enterotoxin B (SEB) produced by the Staphylococcus aureus bacteriumis most commonly associated with food poisoning and is known to also cause toxic shock syndrome. Currently, no approved vaccine or specific drug is available to treat SEB intoxication. In this study, we fabricated dissolving microneedles (MNs) loaded with recombinant SEB (rSEB) protein, and evaluated its characteristics, including dissolution profile, protein particle size, insertion depth, antigen retention time in vivo, and skin irritation. Our results showed that rSEB protein-loaded dissolving MNs made of chondroitin sulfate (2%) and trehalose (0.8%) could easily penetrate into the mouse skin within 5 min. The rSEB particle size was unchanged before and after MN fabrication. The skin penetration depth of the MNs was 260 µm. Moreover, the MNs also significantly extended the antigen retention time in vivo. rSEB protein-loaded dissolving MNs also triggered slight erythema at the beginning of administration, but this erythema disappeared within a few hours. More importantly, we investigated the immunogenicity and protective efficacy of rSEB protein-loaded dissolving MNs. Challenge studies in mice revealed that mice in full-dose MN group had a high level of SEB specific antibody response thatprovided100% protection against a lethal SEB toxin challenge. However, there was only 60% protection observed in mice that were in the half-dose MN (dose sparing) group. We also determined the pathological alterations in the tissues of the immunized mice. Taken together, these dissolving MNs may present a promising transcutaneous immunization strategy for treating SEB intoxication.


Assuntos
Enterotoxinas/administração & dosagem , Imunização/métodos , Microinjeções/instrumentação , Agulhas , Infecções Estafilocócicas/prevenção & controle , Administração Cutânea , Animais , Anticorpos Antibacterianos/sangue , Enterotoxinas/genética , Enterotoxinas/imunologia , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/imunologia , Pele/efeitos dos fármacos , Pele/patologia , Infecções Estafilocócicas/imunologia , Staphylococcus aureus , Suínos
7.
Int Immunopharmacol ; 55: 142-150, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29253820

RESUMO

Outbreaks of hand, foot and mouth disease (HFMD), which is caused by Enterovirus 71 (EV71), have erupted in recent years. Andrographolide sulfonate (Trade name: Xiyanping injection) has been recommended to treat severe HFMD in China because of its conventional antithermic and antitoxic activities, but its actual mechanism has not been revealed clearly until now. To explore its therapeutic efficacy and mechanism, a Xiyanping injection treatment mouse model was established. Based on the therapeutic model, routine clinical parameters and histopathologic changes were investigated, in the same time, viral loads, immune cells, inflammatory molecules and cell signaling pathways were determined. Xiyanping injection treatment protected mice from lethal EV71 challenge in a therapeutic regimen-dependent manner, which may mostly depend on its direct immunomodulatory activities on neutrophil and T lymphocyte. Reduced inflammatory molecular production of neutrophil and elevated T lymphocyte activity may result from its marked inhibition of some signaling pathways. Taken together, Xiyanping injection was an effective treatment for severe HFMD by improving hosts' immunity.


Assuntos
Antivirais/uso terapêutico , Diterpenos/uso terapêutico , Enterovirus Humano A/imunologia , Infecções por Enterovirus/tratamento farmacológico , Doença de Mão, Pé e Boca/tratamento farmacológico , Animais , China , Modelos Animais de Doenças , Humanos , Imunomodulação , Medicina Tradicional Chinesa , Camundongos , Camundongos Endogâmicos ICR , Modelos Biológicos , Mortalidade
8.
J Mol Cell Biol ; 10(6): 573-585, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-29121325

RESUMO

Influenza virus (IAV) infection is a major cause of severe respiratory illness that affects almost every country in the world. IAV infections result in respiratory illness and even acute lung injury and death, but the underlying mechanisms responsible for IAV pathogenesis have not yet been fully elucidated. In this study, the basic fibroblast growth factor 2 (FGF2) level was markedly increased in H1N1 virus-infected humans and mice. FGF2, which is predominately derived from epithelial cells, recruits and activates neutrophils via the FGFR2-PI3K-AKT-NFκB signaling pathway. FGF2 depletion or knockout exacerbated influenza-associated disease by impairing neutrophil recruitment and activation. More importantly, administration of the recombinant FGF2 protein significantly alleviated the severity of IAV-induced lung injury and promoted the survival of IAV-infected mice. Based on the results from experiments in which neutrophils were depleted and adoptively transferred, FGF2 protected mice against IAV infection by recruiting neutrophils. Thus, FGF2 plays a critical role in preventing IAV-induced lung injury, and FGF2 is a promising potential therapeutic target during IAV infection.


Assuntos
Lesão Pulmonar Aguda/virologia , Fator 2 de Crescimento de Fibroblastos/imunologia , Vírus da Influenza A Subtipo H1N1/imunologia , Influenza Humana/imunologia , Infiltração de Neutrófilos , Infecções por Orthomyxoviridae/imunologia , Lesão Pulmonar Aguda/etiologia , Lesão Pulmonar Aguda/imunologia , Animais , Feminino , Fator 2 de Crescimento de Fibroblastos/uso terapêutico , Humanos , Influenza Humana/complicações , Influenza Humana/terapia , Camundongos , Camundongos Endogâmicos C57BL , Infecções por Orthomyxoviridae/complicações , Infecções por Orthomyxoviridae/terapia , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/uso terapêutico
9.
Vaccine ; 35(30): 3709-3717, 2017 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-28576572

RESUMO

Enterovirus 71 (EV71) and Coxsackievirus A16 (CVA16) are the two major causative agents of hand, foot and mouth disease (HFMD), which erupts in the Asia-Pacific regions. A bivalent vaccine against both EV71 and CVA16 is highly desirable. In the present study, on the bases that an experimental bivalent vaccine comprising of inactivated EV71 and CVA16 induces a balanced protective immunity against both EV71 and CVA16, we compare the immunogenicity and reactogenicity of one fourth of a full dose of an intradermal vaccine administered by needle-free liquid jet injector with a full dose of an intramuscular vaccine administered by needle-syringe in monkeys. The results suggest that intradermal injection of a fractional dose of an inactivated HFMD vaccine elicits similar immunogenicity and reactogenicity to intramuscular inoculation of a full dose of an Al(OH)3-adjuvanted vaccine, regardless of whether monovalent or bivalent vaccines were used. Our results support the use of an intradermal bivalent vaccine strategy for HFMD vaccination in order to satisfy the requirements and reduce the costs.


Assuntos
Adjuvantes Imunológicos , Hidróxido de Alumínio/imunologia , Enterovirus Humano A/imunologia , Doença de Mão, Pé e Boca/prevenção & controle , Imunogenicidade da Vacina , Injeções Intramusculares/métodos , Vacinas Virais/imunologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Relação Dose-Resposta Imunológica , Enterovirus Humano A/genética , Doença de Mão, Pé e Boca/imunologia , Humanos , Injeções Intradérmicas , Macaca mulatta , Camundongos , Vacinação , Vacinas Virais/administração & dosagem
10.
Antiviral Res ; 145: 168-174, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28778831

RESUMO

Hepatitis C virus (HCV) has a devastating impact on human health, and infections can progress into liver fibrosis, cirrhosis, and hepatocellular carcinoma. There is no effective HCV vaccine. In this study, we rescued a recombinant PR8 influenza viral vector, called rgFLU-HCVCE1E2, carrying the core and envelope glycoprotein (C/E1/E2) epitopes of HCV inserted into the influenza nonstructural protein 1 gene. The morphological characteristics of rgFLU-HCVCE1E2 and the expression of the C/E1/E2 epitopes of HCV were examined. rgFLU-HCVCE1E2 replicated in various cell lines, including MDCK, A549, and Huh7.5 cells. More importantly, in BALB/c mice immunized intranasally twice at a 21-day interval with 104, 105, or 106 TCID50 rgFLU-HCVCE1E2, the viral vector induced a robust antibody response to influenza and HCV and potent IFN-γ and IL-4 secretion in response to HCV antigens in a dose-dependent manner. The rgFLU-HCVCE1E2 virus also stimulated IFN-γ production by virus-specific peripheral blood mononuclear cells in patients with chronic HCV infection. The study demonstrated that rgFLU-HCVCE1E2 carrying HCV antigens is immunogenic in vivo and has potential for the development of a HCV vaccine.


Assuntos
Epitopos/imunologia , Hepacivirus/imunologia , Imunogenicidade da Vacina , Vacinas contra Hepatite Viral/imunologia , Células A549 , Administração Intranasal , Animais , Anticorpos Antivirais/biossíntese , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Cães , Vetores Genéticos , Hepacivirus/genética , Humanos , Imunização , Interferon gama/imunologia , Interferon gama/metabolismo , Interleucina-4/imunologia , Interleucina-4/metabolismo , Células Madin Darby de Rim Canino , Camundongos , Camundongos Endogâmicos BALB C , Vacinas Sintéticas/imunologia , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia , Proteínas do Envelope Viral/metabolismo , Vacinas contra Hepatite Viral/administração & dosagem , Vacinas contra Hepatite Viral/química , Vacinas contra Hepatite Viral/genética
11.
Sci Rep ; 6: 19840, 2016 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-26813885

RESUMO

Respiratory syncytial virus (RSV) infection is a major cause of severe lower respiratory illness in infants and young children, but the underlying mechanisms responsible for viral pathogenesis have not been fully elucidated. To date, no drugs or vaccines have been employed to improve clinical outcomes for RSV-infected patients. In this paper, we report that angiotensin-converting enzyme-2 (ACE2) protected against severe lung injury induced by RSV infection in an experimental mouse model and in pediatric patients. Moreover, ACE2 deficiency aggravated RSV-associated disease pathogenesis, mainly by its action on the angiotensin II type 1 receptor (AT1R). Furthermore, administration of a recombinant ACE2 protein alleviated the severity of RSV-induced lung injury. These findings demonstrate that ACE2 plays a critical role in preventing RSV-induced lung injury, and suggest that ACE2 is a promising potential therapeutic target in the management of RSV-induced lung disease.


Assuntos
Lesão Pulmonar/metabolismo , Peptidil Dipeptidase A/metabolismo , Infecções por Vírus Respiratório Sincicial/metabolismo , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Enzima de Conversão de Angiotensina 2 , Animais , Modelos Animais de Doenças , Lesão Pulmonar/genética , Lesão Pulmonar/patologia , Lesão Pulmonar/virologia , Camundongos , Camundongos Knockout , Peptidil Dipeptidase A/genética , Peptidil Dipeptidase A/farmacologia , Receptor Tipo 1 de Angiotensina/metabolismo , Receptor Tipo 2 de Angiotensina/metabolismo , Sistema Renina-Angiotensina , Infecções por Vírus Respiratório Sincicial/genética , Infecções por Vírus Respiratório Sincicial/patologia , Infecções por Vírus Respiratório Sincicial/virologia , Índice de Gravidade de Doença
12.
PLoS One ; 10(4): e0123659, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25884801

RESUMO

The emergence of severe cases of human influenza A (H7N9) viral infection in China in the spring of 2003 resulted in a global effort to rapidly develop an effective candidate vaccine. In this study, a cold-adapted (ca), live attenuated monovalent reassortant influenza H7N9 virus (Ah01/AA ca) was generated using reverse genetics that contained hemagglutinin (HA) and neuraminidase (NA) genes from a 2013 pandemic A H7N9 isolate, A/Anhui/01/2013 virus (Ah01/H7N9); the remaining six backbone genes derived from the cold-adapted influenza H2N2 A/Ann Arbor/6/60 virus (AA virus). Ah01/AA ca virus exhibited temperature sensitivity (ts), ca, and attenuation (att) phenotypes. Intranasal immunization of female BALB/c mice with Ah01/AA ca twice at a 2-week interval induced robust humoral, mucosal, and cell-mediated immune responses in a dose-dependent manner. Furthermore, the candidate Ah01/AA ca virus was immunogenic and offered partial or complete protection of mice against a lethal challenge by the live 2013 influenza A H7N9 (A/Anhui/01/2013). Protection was demonstrated by the inhibition of viral replication and the attenuation of histopathological changes in the challenged mouse lung. Taken together, these data support the further evaluation of this Ah01/AA ca candidate vaccine in primates.


Assuntos
Imunização , Subtipo H7N9 do Vírus da Influenza A/imunologia , Vacinas contra Influenza/imunologia , Influenza Humana/prevenção & controle , Infecções por Orthomyxoviridae/prevenção & controle , Animais , Feminino , Humanos , Influenza Humana/virologia , Camundongos , Camundongos Endogâmicos BALB C , Infecções por Orthomyxoviridae/virologia
13.
Hum Vaccin Immunother ; 11(4): 1057-64, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25933187

RESUMO

Respiratory syncytial virus (RSV) is a major pathogen in infants and the elderly, causing pneumonia and bronchiolitis. Despite decades of research, to date there is still no approved RSV vaccine available. In this study, we developed RSV virus-like particle (VLP) vaccines containing an RSV fusion (F) and/or attachment (G) protein with Newcastle disease virus (NDV) as the platform. The VLPs were expressed in a baculovirus system and purified by sucrose gradient centrifugation. BALB/c mice immunized intranasally (i.n.) with rNDV/RSV/F plus rNDV/RSV/G developed robust humoral, mucosal RSV-specific antibodies and cellular immune responses. Furthermore, rNDV/RSV/F plus rNDV/RSV/G provided better protection than did rNDV/RSV/F or rNDV/RSV/G alone, as shown by an obvious decrease in viral replication together with alleviation of histopathological changes in the lungs of the challenged mice. Our data demonstrate that the intranasal vaccination of combined RSV virus-like particle vaccine candidates has great potential for protection against RSV infection.


Assuntos
Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vacinas contra Vírus Sincicial Respiratório/administração & dosagem , Vacinas contra Vírus Sincicial Respiratório/imunologia , Vacinas de Partículas Semelhantes a Vírus/imunologia , Administração Intranasal , Animais , Anticorpos Neutralizantes/imunologia , Linhagem Celular , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Infecções por Vírus Respiratório Sincicial/imunologia , Vacinas contra Vírus Sincicial Respiratório/uso terapêutico
14.
Int Immunopharmacol ; 23(1): 134-8, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25192652

RESUMO

Influenza A H7N9 virus has demonstrated considerable pandemic potential in China ever since early spring 2013. Until now, there have been no specific medicines to treat influenza A H7N9 virus infected patients. Development of a safe and effective H7N9 therapeutic preparation is urgently needed. To this end, we prepared and evaluated the pepsin-digested F(ab')2 fragments of serum IgGs from the horses inoculated with a inactivated influenza A H7N9 whole virus antigens. The protective effects of the F(ab')2 fragments against H7N9 virus infection were determined in cultured MDCK cells by cytopathic effect (CPE) and evaluated in a BALB/c mouse model by observing death, weight loss and viral load. The in vitro results showed that the F(ab')2 fragments had an HI titer of 1:2048 and a neutralization titer of 1: 31,623. The in vivo assays suggested that 600 U of the preparations could efficiently protect BALB/c mice from a lethal dose of A/Anhui/01/2013 (H7N9) infection even when administered two days post infection. Thus, this highly purified preparation should be a potential candidate for treating severe patients suffering from influenza A H7N9.


Assuntos
Anticorpos Neutralizantes/administração & dosagem , Fragmentos Fab das Imunoglobulinas/administração & dosagem , Imunoterapia/métodos , Subtipo H7N9 do Vírus da Influenza A/imunologia , Influenza Humana/terapia , Infecções por Orthomyxoviridae/terapia , Animais , Modelos Animais de Doenças , Cães , Cavalos , Humanos , Influenza Humana/imunologia , Células Madin Darby de Rim Canino , Camundongos , Camundongos Endogâmicos BALB C , Infecções por Orthomyxoviridae/imunologia , Carga Viral/efeitos dos fármacos
15.
Antiviral Res ; 111: 1-7, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25173575

RESUMO

A novel influenza virus of H7N9 subtype circulated throughout China in 2013. The high fatality rate, appearance of several family clusters, and transmission in animal models observed during this outbreak accelerated efforts to identify effective strategies to prevent the spread of this influenza subtype. In this study, the recombinant protein NP-M1-HSP60, a fusion of the nucleoprotein and M1 matrix protein of the A/PR/8/34 (H1N1) influenza virus strain and HSP60, was effectively expressed in Escherichia coli and purified as a candidate component for an influenza vaccine. Intranasal immunization of female BALB/c mice with NP-M1-HSP60 in combination with an oil-in-water adjuvant twice at a 2-week interval induced robust humoral, mucosal, and cell-mediated immune responses. Moreover, this immunization strategy completely protected mice from lethal influenza H7N9 virus challenge and significantly inhibited viral replication in the challenged mouse lung. These data suggest that this vaccine construct has great potential for the basic development of an influenza H7N9 vaccine.


Assuntos
Chaperonina 60/administração & dosagem , Subtipo H7N9 do Vírus da Influenza A/imunologia , Vacinas contra Influenza/administração & dosagem , Influenza Humana/prevenção & controle , Proteínas de Ligação a RNA/administração & dosagem , Proteínas do Core Viral/administração & dosagem , Proteínas da Matriz Viral/administração & dosagem , Animais , Anticorpos Antivirais/imunologia , Chaperonina 60/genética , Chaperonina 60/imunologia , Feminino , Humanos , Subtipo H7N9 do Vírus da Influenza A/genética , Vacinas contra Influenza/genética , Vacinas contra Influenza/imunologia , Influenza Humana/imunologia , Influenza Humana/virologia , Camundongos , Camundongos Endogâmicos BALB C , Proteínas do Nucleocapsídeo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/imunologia , Vacinação , Proteínas do Core Viral/genética , Proteínas do Core Viral/imunologia , Proteínas da Matriz Viral/genética , Proteínas da Matriz Viral/imunologia
16.
Antiviral Res ; 104: 110-7, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24509239

RESUMO

Respiratory syncytial virus (RSV) is an important viral pathogen that causes life-threatening respiratory infections in both infants and the elderly; no vaccines are at present available. In this report, we examined the use of influenza virus as a vehicle for production of an experimental RSV vaccine. We used reverse genetics to generate a recombinant influenza A virus with epitopes from the RSV fusion (F) and attachment (G) proteins (rFlu/RSV/F+G) in the influenza virus nonstructural (NS1) protein gene. Expression of RSV F+G epitope proteins was confirmed by Western blotting, and no changes in viral morphology were evident following examination by electron microscopy. BALB/c mice immunized intranasally with rFlu/RSV/F+G showed viral-specific antibody responses against both influenza and RSV. Total IgG, IgG1, IgG2a and IgA were measured in mice immunized with rFlu/RSV/F+G, revealing robust cellular and mucosal immune responses. Furthermore, we found that rFlu/RSV/F+G conferred protection against subsequent influenza and RSV challenges, showing significant decreases in viral replication and obvious attenuation of histopathological changes associated with viral infections. These findings suggest that rFlu/RSV/F+G is a promising vaccine candidate, which should be further assessed using cotton rat and primate models.


Assuntos
Anticorpos Antivirais/imunologia , Epitopos/imunologia , Vacinas contra Influenza/imunologia , Infecções por Vírus Respiratório Sincicial/imunologia , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vírus Sinciciais Respiratórios/imunologia , Proteínas Virais de Fusão/imunologia , Animais , Linhagem Celular , Chlorocebus aethiops , Modelos Animais de Doenças , Cães , Epitopos/química , Feminino , Ordem dos Genes , Vetores Genéticos/genética , Imunidade nas Mucosas , Imunização , Vacinas contra Influenza/genética , Fígado/imunologia , Fígado/virologia , Pulmão/imunologia , Pulmão/patologia , Pulmão/virologia , Camundongos , Camundongos Endogâmicos BALB C , Vírus Sinciciais Respiratórios/genética , Células Th1/imunologia , Proteínas Virais de Fusão/genética
17.
PLoS One ; 9(6): e99322, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24937303

RESUMO

In early spring 2013, the emergence of the influenza A (H7N9) virus in humans in Eastern China raised concerns of a new influenza pandemic. Development of a safe and effective H7N9 influenza vaccine is urgently needed. To this end, we first synthesized the hemagglutinin (HA) and neuraminidase (NA) genes of the influenza A (H7N9) virus A/AnHui/1/2013. Using reverse genetics, we rescued a reassortant virus (H7N9/PR8) that contained the HA and NA genes from wild-type H7N9 and six genes encoding internal proteins from the A/Puerto Rico/8/34 (PR8) virus. Next, the pathogenicity of the reassortant virus was evaluated both in vivo and in vitro. We found that the virus was non-pathogenic in mice and was stable after serial passaging in eggs. Furthermore, we found that a monovalent influenza A (H7N9) split vaccine prepared from the virus was immunogenic in mice and ferrets. When given intramuscularly, the vaccine (two doses of at least 15-µg) completely protected mice from normally lethal wild-type H7N9 virus challenge. In summary, our H7N9 vaccine, developed over a short time, is a potential candidate for further clinical evaluation and human use.


Assuntos
Subtipo H7N9 do Vírus da Influenza A/imunologia , Vacinas contra Influenza/imunologia , Influenza Humana/prevenção & controle , Animais , Capsídeo/ultraestrutura , Galinhas , Chlorocebus aethiops , Feminino , Furões , Humanos , Subtipo H7N9 do Vírus da Influenza A/genética , Subtipo H7N9 do Vírus da Influenza A/ultraestrutura , Influenza Humana/imunologia , Influenza Humana/virologia , Camundongos Endogâmicos BALB C , Genética Reversa , Vacinação , Vacinas Atenuadas/imunologia , Células Vero , Replicação Viral
18.
Sci Rep ; 4: 7027, 2014 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-25391767

RESUMO

Since March 2013, the emergence of an avian-origin influenza A (H7N9) virus has raised concern in China. Although most infections resulted in respiratory illness, some severe cases resulted in acute respiratory distress syndrome (ARDS), which is a severe form of acute lung injury (ALI) that further contributes to morbidity. To date, no effective drugs that improve the clinical outcome of influenza A (H7N9) virus-infected patients have been identified. Angiotensin-converting enzyme (ACE) and ACE2 are involved in several pathologies such as cardiovascular functions, renal disease, and acute lung injury. In the current study, we report that ACE2 could mediate the severe acute lung injury induced by influenza A (H7N9) virus infection in an experimental mouse model. Moreover, ACE2 deficiency worsened the disease pathogenesis markedly, mainly by targeting the angiotensin II type 1 receptor (AT1). The current findings demonstrate that ACE2 plays a critical role in influenza A (H7N9) virus-induced acute lung injury, and suggest that might be a useful potential therapeutic target for future influenza A (H7N9) outbreaks.


Assuntos
Lesão Pulmonar Aguda/genética , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Antivirais/farmacologia , Losartan/farmacologia , Infecções por Orthomyxoviridae/genética , Peptidil Dipeptidase A/genética , Lesão Pulmonar Aguda/tratamento farmacológico , Lesão Pulmonar Aguda/mortalidade , Lesão Pulmonar Aguda/virologia , Bloqueadores do Receptor Tipo 2 de Angiotensina II/farmacologia , Enzima de Conversão de Angiotensina 2 , Animais , Expressão Gênica , Humanos , Imidazóis/farmacologia , Subtipo H7N9 do Vírus da Influenza A/efeitos dos fármacos , Subtipo H7N9 do Vírus da Influenza A/patogenicidade , Subtipo H7N9 do Vírus da Influenza A/fisiologia , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/patologia , Pulmão/virologia , Camundongos , Camundongos Endogâmicos C57BL , Infecções por Orthomyxoviridae/tratamento farmacológico , Infecções por Orthomyxoviridae/mortalidade , Infecções por Orthomyxoviridae/virologia , Peptidil Dipeptidase A/deficiência , Piridinas/farmacologia , Receptor Tipo 1 de Angiotensina/genética , Receptor Tipo 1 de Angiotensina/metabolismo , Receptor Tipo 2 de Angiotensina/genética , Receptor Tipo 2 de Angiotensina/metabolismo , Transdução de Sinais , Análise de Sobrevida , Carga Viral/efeitos dos fármacos
19.
Antiviral Res ; 98(1): 4-11, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23416215

RESUMO

The matrix protein 2ectodomain (M2e) of the influenza A virus is a rational target antigen candidate for the development of a universal influenza virus-like particle (VLP) vaccine. In this study, a recombinant M2 protein with three tandem copies of M2e (3M2e), nucleoprotein (NP) epitopes and hepatitis B virus core (HBc), were expressed in Escherichia coli and purified by column chromatography. Mice immunized with 3M2e-NP-HBc in combination with an oil-in-water SP01 adjuvant produced robust M2e specific antibodies and cellular immune responses. Most importantly, the 3M2e-NP-HBc VLP vaccine provided enhanced protection against a lethal challenge with pandemic 2009 H1N1 and HPAI H5N1 virus through increased survival rates, a significant decrease in viral replication, and obvious alleviation of histopathological lung changes in challenged mice. Our results imply that a cellular immune response to NP is a plausible mechanism mediating this enhanced protection. These findings suggest that 3M2e-NP-HBc VLP has great potential as the basis development of a broadly protective influenza vaccine.


Assuntos
Vacinas contra Influenza/imunologia , Influenza Humana/imunologia , Influenza Humana/prevenção & controle , Nucleoproteínas/imunologia , Vacinas de Partículas Semelhantes a Vírus/imunologia , Proteínas da Matriz Viral/imunologia , Animais , Anticorpos Antivirais/imunologia , Epitopos/genética , Epitopos/imunologia , Feminino , Humanos , Imunização , Vírus da Influenza A Subtipo H1N1/genética , Vírus da Influenza A Subtipo H1N1/imunologia , Virus da Influenza A Subtipo H5N1/genética , Virus da Influenza A Subtipo H5N1/imunologia , Vacinas contra Influenza/genética , Influenza Humana/virologia , Camundongos , Camundongos Endogâmicos BALB C , Nucleoproteínas/genética , Vacinas de Partículas Semelhantes a Vírus/genética , Proteínas da Matriz Viral/genética
20.
PLoS One ; 7(1): e30252, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22279575

RESUMO

BACKGROUND: The increase in recent outbreaks and unpredictable changes of highly pathogenic avian influenza (HPAI) H5N1 in birds and humans highlights the urgent need to develop a cross-protective H5N1 vaccine. We here report our development of a multiple-clade H5N1 influenza vaccine tested for immunogenicity and efficacy to confer cross-protection in an animal model. METHODOLOGY/PRINCIPAL FINDINGS: Mice received two doses of influenza split vaccine with oil-in-water emulsion adjuvant SP01 by intranasal administration separated by two weeks. Single vaccines (3 µg HA per dose) included rg-A/Vietnam/1203/2004(Clade 1), rg-A/Indonesia/05/2005(Clade 2.1), and rg-A/Anhui/1/2005(Clade 2.3.4). The trivalent vaccine contained 1 µg HA per dose of each single vaccine. Importantly, complete cross-protection was observed in mice immunized using trivalent vaccine with oil-in-water emulsion adjuvant SP01 that was subsequently challenged with the lethal A/OT/SZ/097/03 influenza strain (Clade 0), whereas only the survival rate was up to 60% in single A/Anhui/1/2005 vaccine group. CONCLUSION/SIGNIFICANCE: Our findings demonstrated that the multiple-clade H5N1 influenza vaccine was able to elicit a cross-protective immune response to heterologous HPAI H5N1 virus, thus giving rise to a broadly cross-reactive vaccine to potential prevention use ahead of the strain-specific pandemic influenza vaccine in the event of an HPAI H5N1 influenza outbreak. Also, the multiple-clade adjuvanted vaccine could be useful in allowing timely initiation of vaccination against unknown pandemic virus.


Assuntos
Proteção Cruzada/imunologia , Virus da Influenza A Subtipo H5N1/imunologia , Vacinas contra Influenza/imunologia , Infecções por Orthomyxoviridae/imunologia , Administração Intranasal , Animais , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Reações Cruzadas/imunologia , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Imunoglobulina A/sangue , Imunoglobulina A/imunologia , Virus da Influenza A Subtipo H5N1/classificação , Vacinas contra Influenza/administração & dosagem , Interferon gama/imunologia , Interferon gama/metabolismo , Interleucina-4/imunologia , Interleucina-4/metabolismo , Linfócitos/efeitos dos fármacos , Linfócitos/imunologia , Linfócitos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Infecções por Orthomyxoviridae/prevenção & controle , Infecções por Orthomyxoviridae/virologia , Especificidade da Espécie , Vacinação/métodos , Replicação Viral/efeitos dos fármacos , Replicação Viral/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA