Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Hum Vaccin Immunother ; 20(1): 2330770, 2024 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-38602539

RESUMO

The immunogenicity and safety of the concomitant administration of recombinant COVID-19 vaccine and quadrivalent inactivated influenza vaccine (Split Virion) (QIIV) in Chinese adults are unclear. In this open-label, randomized controlled trial, participants aged ≥ 18 years were recruited. Eligible healthy adults were randomly assigned (1:1) to receive QIIV at the same time as the first dose of COVID-19 vaccine (simultaneous-group) or 14 days after the second dose of COVID-19 vaccine (non-simultaneous-group). The primary outcome was to compare the difference in immunogenicity of QIIV (H1N1, H3N2, Yamagata, and Victoria) between the two groups. A total of 299 participants were enrolled, 149 in the simultaneous-group and 150 in the non-simultaneous-group. There were no significant differences in geometric mean titer (GMT) [H1N1: 386.4 (95%CI: 299.2-499.0) vs. 497.4 (95%CI: 377.5-655.3); H3N2: 66.9 (95%CI: 56.1-79.8) vs. 81.4 (95%CI: 67.9-97.5); Yamagata: 95.6 (95%CI: 79.0-115.8) vs. 74.3 (95%CI: 58.6-94.0); and Victoria: 48.5 (95%CI: 37.6-62.6) vs. 65.8 (95%CI: 49.0-88.4)] and seroconversion rate (H1N1: 87.5% vs. 90.1%; H3N2: 58.1% vs. 62.0%; Yamagata: 75.0% vs. 64.5%; and Victoria: 55.1% vs. 62.8%) of QIIV antibodies between the simultaneous and non-simultaneous groups. For the seroprotection rate of QIIV antibodies, a higher seroprotection rate of Yamagata antibody was observed only in the simultaneous-group than in the non-simultaneous-group [86.0% vs. 76.0%, p = .040]. In addition, no significant difference in adverse events was observed between the two groups (14.2% vs. 23.5%, p = .053). In conclusion, no immune interference or safety concerns were found for concomitant administration of COVID-19 vaccine with QIIV in adults aged ≥ 18 years.


Assuntos
COVID-19 , Vírus da Influenza A Subtipo H1N1 , Vacinas contra Influenza , Adulto , Humanos , Vacinas contra COVID-19/efeitos adversos , COVID-19/prevenção & controle , Vírus da Influenza A Subtipo H3N2 , Vacinas contra Influenza/efeitos adversos , Anticorpos , China
2.
Mol Pharm ; 17(7): 2546-2554, 2020 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-32426985

RESUMO

Naturally occurring pentacyclic triterpenes, such as betulinic acid (BA) and its derivatives, exhibit various pharmaceutical activities and have been the subject of great interest, in particular for their antiviral properties. Here, we found a new anti-influenza virus conjugate, hexakis 6-deoxy-6-[4-N-(3ß-hydroxy-lup-20(29)-en-28-oate)aminomethyl-1H-1,2,3-triazol-1-yl]-2,3-di-O-acetyl-α-cyclodextrin (CYY1-11, 1), in a mini library of pentacyclic triterpene-cyclodextrin conjugates by performing a cell-based screening assay and then exploring the underlying mechanisms. Our results showed that conjugate 1 possessed a high-level activity against the influenza virus A/WSN/33 with an IC50 value of 5.20 µM (SI > 38.4). The study of the mechanism of action indicated that conjugate 1 inhibited viral replication by directly targeting the influenza hemagglutinin protein (KD = 1.50 µM), thus efficiently preventing the attachment of the virion to its receptors on host cells and subsequent infection. This study suggests that multivalent BA derivatives have possible use as a new class of influenza virus entry inhibitors.


Assuntos
Antivirais/química , Antivirais/farmacologia , Hemaglutininas/metabolismo , Vírus da Influenza A Subtipo H1N1/efeitos dos fármacos , Influenza Humana/tratamento farmacológico , Triterpenos Pentacíclicos/química , Triazóis/farmacologia , Internalização do Vírus/efeitos dos fármacos , Células A549 , Animais , Antivirais/síntese química , Antivirais/uso terapêutico , Ciclodextrinas/química , Cães , Hemaglutininas/química , Humanos , Vírus da Influenza A Subtipo H1N1/metabolismo , Vírus da Influenza A Subtipo H1N1/patogenicidade , Concentração Inibidora 50 , Células Madin Darby de Rim Canino , Espectrometria de Massas , Ligação Proteica , Triazóis/síntese química , Triazóis/química , Triazóis/uso terapêutico , Ácido Betulínico
3.
Front Microbiol ; 8: 1086, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28659900

RESUMO

H9N2 avian influenza virus is a zoonotic agent with a broad host range that can contribute genetic information to H5 or H7N9 subtype viruses, which are significant threats to both humans and birds. Thus, there is a great need for a vaccine to control H9N2 avian influenza. Three mutant viruses of an H9N2 virus A/chicken/Taixing/10/2010 (rTX-NS1-73, rTX-NS1-100, and rTX-NS1-128) were constructed with different NS1 gene truncations and confirmed by western blot analysis. The genetic stability, pathogenicity, transmissibility, and host immune responses toward these mutants were evaluated. The mutant virus rTX-NS1-128 exhibited the most attenuated phenotype and lost transmissibility. The expression levels of interleukin 12 in the nasal and tracheal tissues from chickens immunized with rTX-NS1-128 were significantly upregulated on day 3 post-immunization and the IgA and IgG antibody levels were significantly increased on days 7, 14, and 21 post-immunization when compared to chickens that received an inactivated vaccine. rTX-NS1-128 also protected chickens from challenge by homologous and heterologous H9N2 avian influenza viruses. The results indicate that rTX-NS1-128 can be used as a potential live-attenuated vaccine against H9N2 avian influenza.

4.
Vaccine ; 34(3): 350-7, 2016 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-26638027

RESUMO

BACKGROUND: H5N1 highly pathogenic avian influenza (HPAI) has raised global concern for causing huge economic losses in poultry industry, and an effective vaccine against HPAI is highly desirable. Live attenuated influenza vaccine with trunctated NS1 protein as a potential strategy will be extremely useful for improving immune efficacy. METHODS: A series of H5N1 avian influenza virus reassortants harboring amino-terminal 48, 70, 73, and 99 aa in NS1 proteins, along with a modified low pathogenic HA protein was generated, and named as S-HALo/NS48, S-HALo/NS70, S-HALo/NS73, and S-HALo/NS99, respectively. In addition, their biological and immunological characteristics were further analyzed. RESULTS: The viruses S-HALo/NS70, S-HALo/NS73, and S-HALo/NS99, but not S-HALo/NS48, had a comparable growth property with the full-length NS1 virus, S-HALo/NSFu. Mice and chickens studies demonstrated that the viruses with truncated NS1 protein were further attenuated when compared to the virus S-HALo/NSFu. Vaccination with the virus S-HALo/NS73 in chickens induced significant cross-protection against homologous clade 2.3.4 H5 virus and heterologous clade 7.2, 2.3.2.1, and 2.3.4.4 H5 viruses. CONCLUSION: A 70-aa amino-terminal fragment of NS1 protein may be long enough for viral replication. The recombinant virus S-HALo/NS73 is a broad-spectrum live attenuated H5N1 avian influenza vaccine candidate in chickens.


Assuntos
Proteção Cruzada , Virus da Influenza A Subtipo H5N1/imunologia , Vacinas contra Influenza/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Deleção de Sequência , Proteínas não Estruturais Virais/genética , Animais , Galinhas , Feminino , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Humanos , Virus da Influenza A Subtipo H5N1/genética , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/genética , Camundongos Endogâmicos BALB C , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Fatores de Virulência/genética
5.
Vet Microbiol ; 178(1-2): 144-9, 2015 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-25934533

RESUMO

H9N2 avian influenza virus (AIV) evolves rapidly in both genovariation and antigenicity. It is essential to monitor the change of antigenicity, in particular in the hemagglutinin (HA) protein. Here we reported the selection of antigenic variants from A/Chicken/Shanghai/F/98 (H9N2) and A/chicken/Taixing/10/2010 (H9N2) viruses using HA-specific monoclonal antibodies (MAbs). Based on the reactivity of these variant and wild-type strains with the MAbs, we identified 6 critical amino acid positions (92, 145, 166, 167, 168, and 197) in the H9 antigenic sites, including the position 92 that has never been reported. Among AIVs originated from chicken in mainland China, the rates of Gly and Arg at position 92 within BJ/94-like (A/chicken/Beijing/1/1994) lineage viruses were 62.2% (28/45) and 37.8% (17/45), respectively; whereas the rates of Gly and Arg at position 92 within Y280-like (A/duck/Hong Kong/Y280/97) lineage viruses were 0.3% (2/670) and 99.1% (673/679), respectively. Our study suggests that G92R mutation together with other identified antigenic sites may serve as molecular markers for H9N2 virus evolution, and may aid improving AIV vaccine effectiveness.


Assuntos
Variação Antigênica/genética , Antígenos Virais/genética , Epitopos/genética , Evolução Molecular , Hemaglutininas/genética , Vírus da Influenza A Subtipo H9N2/genética , Sequência de Aminoácidos , Animais , Galinhas , China , Patos , Epitopos/imunologia
6.
Arch Virol ; 160(4): 917-27, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25616845

RESUMO

The pathogenicity and transmissibility of H9N2 influenza viruses has been widely investigated; however, few studies comparing the biological characteristics of H9N2 viruses isolated from different hosts have been performed. In this study, eight H9N2 viruses, isolated from chickens (Ck/F98, Ck/AH and Ck/TX), pigeons (Pg/XZ), quail/(Ql/A39), ducks (Dk/Y33) and swine (Sw/YZ and Sw/TZ) were selected, and their biological characteristics were determined. The results showed that all H9N2 viruses maintained a preference for both the avian- and human-type receptors, except for Sw/TZ, which had exclusive preference for the human-type receptor. The viruses replicated well in DF-1 and MDCK cells, whereas only three isolates, Ck/F98, Ck/TX and Sw/TZ, could replicate in A549 cells and also replicated in mouse lungs, resulting in body weight loss in mice. All H9N2 viruses were nonpathogenic to chickens and were detected in the trachea and lung tissues. The viruses were shed primarily by the oropharynx and were transmitted efficiently to naïve contact chickens. Our findings suggest that all H9N2 viruses from different hosts exhibit efficient replication and contact-transmission among chickens, and chickens serve as a good reservoir for the persistence and interspecies transmission of H9N2 influenza viruses.


Assuntos
Vírus da Influenza A Subtipo H9N2/isolamento & purificação , Vírus da Influenza A Subtipo H9N2/fisiologia , Influenza Aviária/virologia , Infecções por Orthomyxoviridae/veterinária , Doenças dos Suínos/virologia , Animais , Linhagem Celular , Galinhas , Columbidae , Patos , Feminino , Especificidade de Hospedeiro , Humanos , Vírus da Influenza A Subtipo H9N2/classificação , Vírus da Influenza A Subtipo H9N2/genética , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Infecções por Orthomyxoviridae/virologia , Filogenia , Aves Domésticas , Doenças das Aves Domésticas/virologia , Codorniz , Suínos
7.
Vet Microbiol ; 175(2-4): 244-56, 2015 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-25544041

RESUMO

The location and number of glycosylation in HA proteins exhibit large variations among H5 subtype avian influenza viruses (AIVs). To investigate the effect of glycosylation in the globular head of HA on the pathogenicity and antigenicity of H5N1 AIVs, seven rescued AIVs differing in their glycosylation patterns (144N, 158N and 169N) within the HA globular head of A/Mallard/Huadong/S/2005 were generated using site directed mutagenesis. Results showed that loss of glycosylation 158N was the prerequisite for H5 AIV binding to the α2,6-linked receptor. Only in conjunction with the removal of the 158N glycosylation, the H5 AIVs harboring both 144N and 169N glycosylations obtained an optimal binding preference to the α2,6-linked receptor. Compared with the wild-type virus, growth of viruses lacking glycosylation at either 158N or 169N was significantly reduced both in MDCK and A549 cells, while replication of viruses with additional glycosylation 144N was significantly promoted. Mutant viruses with loss of 158N or 169N glycosylation sites showed increased pathogenicity, systemic spread and pulmonary inflammation in mice compared to the wild-type H5N1 virus. In addition, chicken studies demonstrated that inactivated de-glycosylation 169N mutant induced cross-reaction HI and neutralization antibody against various clades of H5N1 AIVs. Moreover, this type of glycan pattern vaccine virus provided better cross-protection in chickens compared to wild-type vaccine virus. Thus, the glycosylation alteration of HA should be considered in the global surveillance and vaccine design of H5 subtype AIVs.


Assuntos
Glicoproteínas de Hemaglutininação de Vírus da Influenza/metabolismo , Virus da Influenza A Subtipo H5N1/patogenicidade , Influenza Aviária/virologia , Animais , Anticorpos Neutralizantes , Galinhas/imunologia , Reações Cruzadas , Regulação Viral da Expressão Gênica/fisiologia , Glicosilação , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Virus da Influenza A Subtipo H5N1/genética , Camundongos , Mutagênese Sítio-Dirigida , Infecções por Orthomyxoviridae/virologia , Doenças das Aves Domésticas/virologia , Virulência
8.
PLoS One ; 9(4): e95539, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24743258

RESUMO

Since 2003, H5N1-subtype avian influenza viruses (AIVs) with both a deletion of 20 amino acids in the stalk of the neuraminidase (NA) glycoprotein (A-) and a deletion of five amino acids at positions 80 to 84 in the non-structural protein NS1 (S-) have become predominant. To understand the influence of these double deletions in the NA and NS1 proteins on the pathogenicity of H5N1-subtype AIVs, we selected A/mallard/Huadong/S/2005 as a parental strain to generate rescued wild-type A-S- and three variants (A-S+ with a five-amino-acid insertion in the NS1 protein, A+S- with a 20-amino-acid insertion in the NA stalk, and A+S+ with insertions in both NA and NS1 proteins) and evaluated their biological characteristics and virulence. The titers of the AIVs with A- and/or S- replicated in DEF cells were higher than that of A+S+, and the A-S- virus exhibited a replication predominance when co-infected with the other variants in DEF cells. In addition, A-S- induced a more significant increase in the expression of immune-related genes in peripheral blood mononuclear cells of mallard ducks in vitro compared with the other variants. Furthermore, an insertion in the NA and/or NS1 proteins of AIVs resulted in a notable decrease in virulence in ducks, as determined by intravenous pathogenicity index, and the two insertions exerted a synergistic effect on the attenuation of pathogenicity in ducks. In addition, compared with A+S+ and A+S-, the A-S+ and A-S- viruses that were introduced via the intranasal inoculation route exhibited a faster replication ability in the lungs of ducks. These data indicate that both the deletions in the NA stalk and the NS1 protein contribute to the high pathogenicity of H5N1 AIVs in ducks.


Assuntos
Virus da Influenza A Subtipo H5N1/genética , Influenza Aviária/virologia , Neuraminidase/genética , Proteínas não Estruturais Virais/genética , Animais , Patos , Virus da Influenza A Subtipo H5N1/fisiologia , Neuraminidase/fisiologia , Proteínas não Estruturais Virais/fisiologia
9.
Vet Microbiol ; 162(2-4): 614-622, 2013 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-23265247

RESUMO

H5N1 avian influenza virus (AIV) undergoes rapid evolution, and its antigenicity needs to be constantly evaluated in order to update the vaccine strain. In this report, a clade 7.2 AIV isolate named A/Chicken/Huadong/4/2008 (DT) is identified. Antigenic analysis revealed that DT had a significant low cross-reactive HI titre with antiserum against a clade 7 representative AIV, A/Chicken/Shanxi/2/2006 (Re-4). Animal experiments demonstrated that the Re-4+Re-5 vaccine provided 80% protection against DT challenge in chickens. Antisera cross-reactivity showed that a mutant with a change of residues 129, 139, 140 in site A in the HA protein had reduced reactivity with DT antiserum and increased reactivity with Re-4 antiserum. Furthermore, residue Leu129 in site A of the HA protein was confirmed to be critical for maintenance of the reactivity with the DT antiserum, and Asn140, possessing a new glycosylation site, was confirmed to be critical for reducing reactivity with the Re-4 antiserum. These results imply that there is an antigenic drift within clade 7 viruses, and insertion and glycosylation of amino acid residues in site A of the HA protein may contribute to the antigenic variation.


Assuntos
Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Virus da Influenza A Subtipo H5N1/genética , Virus da Influenza A Subtipo H5N1/imunologia , Influenza Aviária/virologia , Sequência de Aminoácidos , Animais , Células COS , Embrião de Galinha , Galinhas , Chlorocebus aethiops , Reações Cruzadas , Cães , Patos , Mapeamento de Epitopos , Epitopos/genética , Epitopos/imunologia , Glicoproteínas de Hemaglutininação de Vírus da Influenza/química , Virus da Influenza A Subtipo H5N1/isolamento & purificação , Virus da Influenza A Subtipo H5N1/patogenicidade , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/imunologia , Influenza Aviária/imunologia , Influenza Aviária/prevenção & controle , Células Madin Darby de Rim Canino , Camundongos , Camundongos Endogâmicos BALB C , Modelos Moleculares , Dados de Sequência Molecular , Filogenia , Virulência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...