Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
J Gen Virol ; 105(6)2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38922678

RESUMO

Highly pathogenic avian influenza (HPAI) H5N1 viruses are responsible for disease outbreaks in wild birds and poultry, resulting in devastating losses to the poultry sector. Since 2020, an increasing number of outbreaks of HPAI H5N1 was seen in wild birds. Infections in mammals have become more common, in most cases in carnivores after direct contact with infected birds. Although ruminants were previously not considered a host species for HPAI viruses, in March 2024 multiple outbreaks of HPAI H5N1 were detected in goats and cattle in the United States. Here, we have used primary bronchus-derived well-differentiated bovine airway epithelial cells (WD-AECs) cultured at air-liquid interface to assess the susceptibility and permissiveness of bovine epithelial cells to infection with European H5N1 virus isolates. We inoculated bovine WD-AECs with three low-passage HPAI clade 2.3.4.4b H5N1 virus isolates and detected rapid increases in viral genome loads and infectious virus during the first 24 h post-inoculation, without substantial cytopathogenic effects. Three days post-inoculation infected cells were still detectable by immunofluorescent staining. These data indicate that multiple lineages of HPAI H5N1 may have the propensity to infect the respiratory tract of cattle and support extension of avian influenza surveillance efforts to ruminants. Furthermore, this study underscores the benefit of WD-AEC cultures for pandemic preparedness by providing a rapid and animal-free assessment of the host range of an emerging pathogen.


Assuntos
Células Epiteliais , Virus da Influenza A Subtipo H5N1 , Replicação Viral , Animais , Bovinos , Células Epiteliais/virologia , Virus da Influenza A Subtipo H5N1/genética , Virus da Influenza A Subtipo H5N1/fisiologia , Virus da Influenza A Subtipo H5N1/isolamento & purificação , Células Cultivadas
2.
Microb Pathog ; 141: 103984, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31972269

RESUMO

In this study, we assessed the pathogenicity of two H5N1 viruses isolated from crows in mice. Eighteen 6-8 weeks BALB/c mice each were intranasally inoculated with 106 EID50/ml of H5N1 viruses A/crow/India/03CA04/2015 (H9N2-PB2 reassortant H5N1) and A/crow/India/02CA01/2012 (Non-reassortant H5N1). The infected mice showed dullness, weight loss and ruffled fur coat. Histopathological examination of lungs showed severe congestion, haemorrhage, thrombus, fibrinous exudate in perivascular area, interstitial septal thickening, bronchiolitis and alveolitis leading to severe pneumonic changes and these lesions were less pronounced in reassortant virus infected mice. Viral replication was demonstrated in nasal mucosa, lungs, trachea and brain in both the groups. Brain, lung, nasal mucosa and trachea showed significantly higher viral RNA copies and presence of antigen in immunohistochemistry in both the groups. This study concludes that both the crow viruses caused morbidity and mortality in mice and the viruses were phenotypically highly virulent in mice. The H5N1 viruses isolated from synanthropes pose a serious public health concern and should be monitored continuously for their human spill-over.


Assuntos
Virus da Influenza A Subtipo H5N1/patogenicidade , Infecções por Orthomyxoviridae/virologia , Animais , Biópsia , Corvos , Suscetibilidade a Doenças , Histocitoquímica , Virus da Influenza A Subtipo H5N1/classificação , Virus da Influenza A Subtipo H5N1/isolamento & purificação , Influenza Aviária , Camundongos , Camundongos Endogâmicos BALB C , Infecções por Orthomyxoviridae/patologia , RNA Viral , Vírus Reordenados/genética , Carga Viral , Replicação Viral
3.
Transbound Emerg Dis ; 67(2): 947-955, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31769586

RESUMO

Since 2004, there have been multiple outbreaks of H5 highly pathogenic avian influenza (HPAI) viruses in Laos. Here, we isolated H5N1 HPAI viruses from poultry outbreaks in Laos during 2015-2018 and investigated their genetic characteristics and pathogenicity in chickens. Phylogenetic analysis revealed that the isolates belonged to clade 2.3.2.1c and that they differed from previous Laos viruses with respect to genetic composition. In particular, the isolates were divided into two genotypes, each of which had a different NS segments. The results of possible migration analysis suggested a high likelihood that the Laos isolates were introduced from neighbouring countries, particularly Vietnam. The recent Laos isolate, A/Duck/Laos/NL-1504599/2018, had an intravenous pathogenicity index score of 3.0 and showed a 50% chicken lethal dose of 102.5 EID50 /0.1 ml, indicating high pathogenicity. The isolated viruses exhibited no critical substitution in the markers associated with mammalian adaptation, but possess markers related to neuraminidase inhibitor resistance. These results emphasize the need for ongoing surveillance of circulating influenza virus in South-East Asia, including Laos, to better prepare for and mitigate global spread of H5 HPAI.


Assuntos
Galinhas/virologia , Surtos de Doenças/veterinária , Virus da Influenza A Subtipo H5N1/genética , Influenza Aviária/epidemiologia , Doenças das Aves Domésticas/epidemiologia , Animais , Genótipo , Virus da Influenza A Subtipo H5N1/isolamento & purificação , Virus da Influenza A Subtipo H5N1/patogenicidade , Influenza Aviária/virologia , Laos/epidemiologia , Filogenia , Aves Domésticas , Doenças das Aves Domésticas/virologia , Organismos Livres de Patógenos Específicos
4.
Epidemiol Infect ; 147: e213, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-31364549

RESUMO

The epidemiology of H5N1 and H7N9 avian viruses of humans infected in China differs despite both viruses being avian reassortants that have inherited six internal genes from a common ancestor, H9N2. The median age of infected populations is substantially younger for H5N1 virus (26 years) compared with H7N9 virus (63 years). Population susceptibility to infection with seasonal influenza is understood to be influenced by cross-reactive CD8+ T cells directed towards immunogenic peptides derived from internal viral proteins which may provide some level of protection against further influenza infection. Prior exposure to seasonal influenza peptides may influence the age-related infection patterns observed for H5N1 and H7N9 viruses. A comparison of relatedness of immunogenic peptides between historical human strains and the two avian emerged viruses was undertaken for a possible explanation in the differences in age incidence observed. There appeared to be some relationship between past exposure to related peptides and the lower number of H5N1 virus cases in older populations, however the relationship between prior exposure and older populations among H7N9 virus patients was less clear.


Assuntos
Distribuição por Idade , Antígenos Virais/imunologia , Exposição Ambiental , Virus da Influenza A Subtipo H5N1/isolamento & purificação , Subtipo H7N9 do Vírus da Influenza A/isolamento & purificação , Influenza Humana/epidemiologia , Influenza Humana/virologia , China/epidemiologia , Suscetibilidade a Doenças , Feminino , Humanos , Incidência , Virus da Influenza A Subtipo H5N1/imunologia , Subtipo H7N9 do Vírus da Influenza A/imunologia , Influenza Humana/imunologia , Masculino , Peptídeos/imunologia
5.
Sci Rep ; 8(1): 13066, 2018 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-30166556

RESUMO

The role of the influenza virus polymerase complex in host range restriction has been well-studied and several host range determinants, such as the polymerase PB2-E627K and PB2-D701N mutations, have been identified. However, there may be additional, currently unknown, human adaptation polymerase mutations. Here, we used a database search of influenza virus H5N1 clade 1.1, clade 2.3.2.1 and clade 2.3.4 strains isolated from 2008-2012 in Southern China, Vietnam and Cambodia to identify polymerase adaptation mutations that had been selected in infected patients. Several of these mutations acted either alone or together to increase viral polymerase activity in human airway cells to levels similar to the PB2-D701N and PB2-E627K single mutations and to increase progeny virus yields in infected mouse lungs to levels similar to the PB2-D701N single mutation. In particular, specific mutations acted synergistically with the PB2-D701N mutation and showed synergistic effects on viral replication both in human airway cells and mice compared with the corresponding single mutations. Thus, H5N1 viruses in infected patients were able to acquire multiple polymerase mutations that acted cooperatively for human adaptation. Our findings give new insight into the human adaptation of AI viruses and help in avian influenza virus risk assessment.


Assuntos
Adaptação Fisiológica/genética , RNA Polimerases Dirigidas por DNA/genética , Virus da Influenza A Subtipo H5N1/genética , Virus da Influenza A Subtipo H5N1/isolamento & purificação , Mutação/genética , Células A549 , Animais , Ásia , Aves/virologia , RNA Polimerases Dirigidas por DNA/química , Células Epiteliais/virologia , Células HEK293 , Humanos , Virus da Influenza A Subtipo H5N1/crescimento & desenvolvimento , Pulmão/patologia , Camundongos , Modelos Moleculares , Replicação Viral
6.
Biochem Biophys Res Commun ; 490(4): 1301-1306, 2017 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-28688767

RESUMO

The interplay between highly pathogenic avian influenza (HPAI) H5N1 virus and immune cells has been extensively studied for years, as host immune components are thought to play significant roles in promoting the systemic spread of the virus and responsible for cytokine storm. Previous studies suggested that the interaction of B cells and monocytes could promote HPAI H5N1 infection by enhancing avian influenza virus receptor expression. In this study, we further investigate the relationship between the HPAI H5N1 virus, activated B cells, and DC-SIGN expression. DC-SIGN has been described as an important factor for mediating various types of viral infection. Here, we first demonstrate that HPAI H5N1 infection could induce an activation of B cells, which was associated with DC-SIGN expression. Using CD40L and recombinant IL-4 for B cell stimulation, we determined that DC-SIGN expressed on activated B cells was able to enhance its susceptibility to HPAI H5N1 infection. Our findings uncover the interplay between this H5N1 virus and B cells and provide important information in understanding how the virus overcomes our immune system, contributing to its unusual immunopathogenesis.


Assuntos
Linfócitos B/virologia , Moléculas de Adesão Celular/imunologia , Interações Hospedeiro-Patógeno , Virus da Influenza A Subtipo H5N1/fisiologia , Lectinas Tipo C/imunologia , Receptores de Superfície Celular/imunologia , Animais , Linfócitos B/imunologia , Antígeno B7-2/genética , Antígeno B7-2/imunologia , Aves/virologia , Ligante de CD40/farmacologia , Moléculas de Adesão Celular/genética , Suscetibilidade a Doenças , Regulação da Expressão Gênica , Humanos , Virus da Influenza A Subtipo H5N1/isolamento & purificação , Interleucina-4/farmacologia , Lectinas Tipo C/genética , Ativação Linfocitária/efeitos dos fármacos , Cultura Primária de Células , Receptores de Superfície Celular/genética , Proteínas Recombinantes/farmacologia , Transdução de Sinais
7.
J Infect Dis ; 214(4): 516-24, 2016 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-27448390

RESUMO

BACKGROUND: Influenza A viruses can replicate in the olfactory mucosa and subsequently use the olfactory nerve to enter the central nervous system (CNS). It is currently unknown whether intervention strategies are able to reduce or prevent influenza virus replication within the olfactory mucosa and subsequent spread to the CNS. Therefore, we tested the efficacy of homologous vaccination and prophylactic oseltamivir to prevent H5N1 virus CNS invasion via the olfactory nerve in our ferret model. METHODS: Ferrets were vaccinated intramuscularly or received oseltamivir (5 mg/kg twice daily) prophylactically before intranasal inoculation of highly pathogenic H5N1 virus (A/Indonesia/05/2005) and were examined using virology and pathology. RESULTS: Homologous vaccination reduced H5N1 virus replication in the olfactory mucosa and prevented subsequent virus spread to the CNS. However, prophylactic oseltamivir did not prevent H5N1 virus replication in the olfactory mucosa sufficiently, resulting in CNS invasion via the olfactory nerve causing a severe meningoencephalitis. CONCLUSIONS: Within our ferret model, vaccination is more effective than prophylactic oseltamivir in preventing CNS invasion by H5N1 virus via the olfactory nerve. This study highlights the importance of including the olfactory mucosa, olfactory nerve, and CNS tissues in future vaccine and antiviral studies, especially for viruses with a known neurotropic potential.


Assuntos
Antivirais/administração & dosagem , Virus da Influenza A Subtipo H5N1/isolamento & purificação , Vacinas contra Influenza/administração & dosagem , Meningoencefalite/prevenção & controle , Infecções por Orthomyxoviridae/complicações , Oseltamivir/administração & dosagem , Animais , Quimioprevenção/métodos , Modelos Animais de Doenças , Feminino , Furões , Virus da Influenza A Subtipo H5N1/efeitos dos fármacos , Virus da Influenza A Subtipo H5N1/imunologia , Injeções Intramusculares , Nervo Olfatório/virologia , Infecções por Orthomyxoviridae/patologia , Infecções por Orthomyxoviridae/virologia , Resultado do Tratamento
8.
mBio ; 6(2)2015 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-25852160

RESUMO

UNLABELLED: A change in viral hemagglutinin (HA) receptor binding specificity from α2,3- to α2,6-linked sialic acid is necessary for highly pathogenic avian influenza (AI) virus subtype H5N1 to become pandemic. However, details of the human-adaptive change in the H5N1 virus remain unknown. Our database search of H5N1 clade 2.2.1 viruses circulating in Egypt identified multiple HA mutations that had been selected in infected patients. Using reverse genetics, we found that increases in both human receptor specificity and the HA pH threshold for membrane fusion were necessary to facilitate replication of the virus variants in human airway epithelia. Furthermore, variants with enhanced replication in human cells had decreased HA stability, apparently to compensate for the changes in viral receptor specificity and membrane fusion activity. Our findings showed that H5N1 viruses could rapidly adapt to growth in the human airway microenvironment by altering their HA properties in infected patients and provided new insights into the human-adaptive mechanisms of AI viruses. IMPORTANCE: Circulation between bird and human hosts may allow H5N1 viruses to acquire amino acid changes that increase fitness for human infections. However, human-adaptive changes in H5N1 viruses have not been adequately investigated. In this study, we found that multiple HA mutations were actually selected in H5N1-infected patients and that H5N1 variants with some of these HA mutations had increased human-type receptor specificity and increased HA membrane fusion activity, both of which are advantageous for viral replication in human airway epithelia. Furthermore, HA mutants selected during viral replication in patients were likely to have less HA stability, apparently as a compensatory mechanism. These results begin to clarify the picture of the H5N1 human-adaptive mechanism.


Assuntos
Variação Genética , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Virus da Influenza A Subtipo H5N1/classificação , Virus da Influenza A Subtipo H5N1/isolamento & purificação , Influenza Humana/virologia , Mutação de Sentido Incorreto , Adaptação Biológica , Animais , Células Cultivadas , Modelos Animais de Doenças , Células Epiteliais/virologia , Humanos , Virus da Influenza A Subtipo H5N1/genética , Virus da Influenza A Subtipo H5N1/crescimento & desenvolvimento , Camundongos Endogâmicos BALB C , Infecções por Orthomyxoviridae/virologia , Análise de Sobrevida
9.
J Virol ; 89(11): 5835-46, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25787281

RESUMO

UNLABELLED: Influenza A viruses (IAVs) express the PB1-F2 protein from an alternate reading frame within the PB1 gene segment. The roles of PB1-F2 are not well understood but appear to involve modulation of host cell responses. As shown in previous studies, we find that PB1-F2 proteins of mammalian IAVs frequently have premature stop codons that are expected to cause truncations of the protein, whereas avian IAVs usually express a full-length 90-amino-acid PB1-F2. However, in contrast to other avian IAVs, recent isolates of highly pathogenic H5N1 influenza viruses had a high proportion of PB1-F2 truncations (15% since 2010; 61% of isolates in 2013) due to several independent mutations that have persisted and expanded in circulating viruses. One natural H5N1 IAV containing a mutated PB1-F2 start codon (i.e., lacking ATG) was 1,000-fold more virulent for BALB/c mice than a closely related H5N1 containing intact PB1-F2. In vitro, we detected expression of an in-frame protein (C-terminal PB1-F2) from downstream ATGs in PB1-F2 plasmids lacking the well-conserved ATG start codon. Transient expression of full-length PB1-F2, truncated (24-amino-acid) PB1-F2, and PB1-F2 lacking the initiating ATG in mammalian and avian cells had no effect on cell apoptosis or interferon expression in human lung epithelial cells. Full-length and C-terminal PB1-F2 mutants colocalized with mitochondria in A549 cells. Close monitoring of alterations of PB1-F2 and their frequency in contemporary avian H5N1 viruses should continue, as such changes may be markers for mammalian virulence. IMPORTANCE: Although most avian influenza viruses are harmless for humans, some (such as highly pathogenic H5N1 avian influenza viruses) are capable of infecting humans and causing severe disease with a high mortality rate. A number of risk factors potentially associated with adaptation to mammalian infection have been noted. Here we demonstrate that the protein PB1-F2 is frequently truncated in recent isolates of highly pathogenic H5N1 viruses. Truncation of PB1-F2 has been proposed to act as an adaptation to mammalian infection. We show that some forms of truncation of PB1-F2 may be associated with increased virulence in mammals. Our data support the assessment of PB1-F2 truncations for genomic surveillance of influenza viruses.


Assuntos
Virus da Influenza A Subtipo H5N1/isolamento & purificação , Virus da Influenza A Subtipo H5N1/fisiologia , Infecções por Orthomyxoviridae/virologia , Proteínas Virais/genética , Proteínas Virais/metabolismo , Fatores de Virulência/genética , Fatores de Virulência/metabolismo , Animais , Apoptose , Linhagem Celular , Códon sem Sentido , Modelos Animais de Doenças , Células Epiteliais/fisiologia , Células Epiteliais/virologia , Feminino , Humanos , Virus da Influenza A Subtipo H5N1/genética , Interferons/biossíntese , Camundongos Endogâmicos BALB C , Infecções por Orthomyxoviridae/patologia , Virulência
10.
Biosens Bioelectron ; 67: 546-52, 2015 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-25263315

RESUMO

Early screening of suspected cases is the key to control the spread of avian influenza (AI) H5N1. In our previous studies, an impedance biosensor with an interdigitated array microelectrode based biochip was developed and validated with pure AI H5 virus, but had limitations in cost and reliability of the biochip, specificity of the antibody against Asian in-field H5N1 virus and detection of H5N1 virus in real samples. The purpose of this study is to develop a low-cost impedance immunosensor for rapid detection of Asian in-field AI H5N1 virus in chicken swabs within 1h and validate it with the H5N1 virus. Specific monoclonal antibodies against AI H5N1 virus were developed by fusion of mouse myeloma cells with spleen cells isolated from an H5N1-virus-immunized mouse. Dot-ELISA analysis demonstrated that the developed antibodies had good affinity and specificity with the H5N1 virus. The microelectrodes were redesigned with compact size, fabricated using an improved wet-etching micro-fabrication process with a higher qualified production rate of 70-80%, and modified with the antibodies by the Protein A method. Equivalent circuit analysis indicated that electron transfer resistor was effective with the increase in impedance after capturing of the H5N1 viruses. Linear relationship between impedance change and logarithmic value of H5N1 virus at the concentrations from 2(-1) to 2(4) HAU/50 µl was found and the lower limit of detection was 2(-1) HAU/50 µl. No obvious interferences from non-target viruses such as H6N2, H9N2, Newcastle disease virus, and infectious bronchitis virus were found. Chicken swab tests showed that the impedance immunosensor had a comparable accuracy with real-time RT-PCR compared to viral isolation.


Assuntos
Anticorpos Monoclonais/química , Técnicas Biossensoriais , Virus da Influenza A Subtipo H5N1/isolamento & purificação , Influenza Aviária/virologia , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Antivirais/química , Anticorpos Antivirais/imunologia , Galinhas/imunologia , Impedância Elétrica , Ensaio de Imunoadsorção Enzimática , Virus da Influenza A Subtipo H5N1/patogenicidade , Influenza Aviária/diagnóstico , Camundongos , Microeletrodos
11.
BMC Infect Dis ; 14: 362, 2014 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-24992826

RESUMO

BACKGROUND: Sporadic emergence of the highly pathogenic avian influenza (HPAI) H5N1 virus infection in humans is a serious concern because of the potential for a pandemic. Conventional or quantitative RT-PCR is the standard laboratory test to detect viral influenza infections. However, this technology requires well-equipped laboratories and highly trained personnel. A rapid, sensitive, and specific alternative screening method is needed. METHODS: By a luminescence-linked enzyme immunoassay, we have developed a H5N1 HPAI virus detection kit using anti-H5 hemagglutinin monoclonal antibodies in combination with the detection of a universal NP antigen of the type A influenza virus. The process takes 15 minutes by use of the fully automated luminescence analyzer, POCube. RESUTLS: We tested this H5/A kit using 19 clinical specimens from 13 patients stored in Vietnam who were infected with clade 1.1 or clade 2.3.4 H5N1 HPAI virus. Approximately 80% of clinical specimens were H5-positive using the POCube system, whereas only 10% of the H5-positive samples were detected as influenza A-positive by an immunochromatography-based rapid diagnostic kit. CONCLUSIONS: This novel H5/A kit using POCube is served as a rapid and sensitive screening test for H5N1 HPAI virus infection in humans.


Assuntos
Virus da Influenza A Subtipo H5N1/isolamento & purificação , Influenza Humana/virologia , Humanos , Técnicas Imunoenzimáticas , Faringe/virologia , Kit de Reagentes para Diagnóstico , Sensibilidade e Especificidade , Vietnã
12.
PLoS Pathog ; 10(6): e1004192, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24945244

RESUMO

Highly pathogenic avian influenza (HPAI) viruses of the H5N1 subtype often cause severe pneumonia and multiple organ failure in humans, with reported case fatality rates of more than 60%. To develop a clinical antibody therapy, we generated a human-mouse chimeric monoclonal antibody (MAb) ch61 that showed strong neutralizing activity against H5N1 HPAI viruses isolated from humans and evaluated its protective potential in mouse and nonhuman primate models of H5N1 HPAI virus infections. Passive immunization with MAb ch61 one day before or after challenge with a lethal dose of the virus completely protected mice, and partial protection was achieved when mice were treated 3 days after the challenge. In a cynomolgus macaque model, reduced viral loads and partial protection against lethal infection were observed in macaques treated with MAb ch61 intravenously one and three days after challenge. Protective effects were also noted in macaques under immunosuppression. Though mutant viruses escaping from neutralization by MAb ch61 were recovered from macaques treated with this MAb alone, combined treatment with MAb ch61 and peramivir reduced the emergence of escape mutants. Our results indicate that antibody therapy might be beneficial in reducing viral loads and delaying disease progression during H5N1 HPAI virus infection in clinical cases and combined treatment with other antiviral compounds should improve the protective effects of antibody therapy against H5N1 HPAI virus infection.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Anticorpos Monoclonais Murinos/uso terapêutico , Imunização Passiva/métodos , Virus da Influenza A Subtipo H5N1/imunologia , Infecções por Orthomyxoviridae/terapia , Ácidos Carbocíclicos , Animais , Anticorpos Monoclonais Humanizados/imunologia , Anticorpos Monoclonais Murinos/imunologia , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Antivirais/uso terapêutico , Linhagem Celular , Ciclopentanos/uso terapêutico , Cães , Quimioterapia Combinada , Feminino , Guanidinas/uso terapêutico , Hospedeiro Imunocomprometido/imunologia , Virus da Influenza A Subtipo H5N1/isolamento & purificação , Interleucina-6/sangue , Pulmão/patologia , Pulmão/virologia , Macaca fascicularis , Células Madin Darby de Rim Canino , Camundongos , Camundongos Endogâmicos BALB C , Modelos Animais , Neuraminidase/antagonistas & inibidores , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/patologia , Carga Viral/imunologia
14.
Biosens Bioelectron ; 51: 170-6, 2014 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-23958581

RESUMO

In this work, we report on oligonucleotide probes bearing metallacarborane [3-iron bis(dicarbollide)] redox label, deposited on gold electrode for electrochemical determination of DNA sequence derived from Avian Influenza Virus (AIV), type H5N1. The oligonucleotide probes containing 5'-terminal NH2 group were covalently attached to the electrode, via NHS/EDC coupling to 3-mercaptopropionic acid SAM, previously deposited on the surface of gold. The changes in redox activity of Fe(III) centre of the metallacarborane complex before and after hybridization process was used as analytical signal. The signals generated upon hybridization with targets such as complementary or non-complementary 20-mer ssDNA or various PCR products consisting of 180-190 bp (dsDNA) were recorded by Osteryoung square-wave voltammetry (OSWV). The developed system was very sensitive towards targets containing sequence complementary to the probe with the detection limit estimated as 0.03 fM (S/N=3.0) and 0.08 fM (S/N=3.0) for 20-mer ssDNA and for dsDNA (PCR product), respectively. The non-complementary targets generated very weak responses. Furthermore, the proposed genosensor was suitable for discrimination of PCR products with different location of the complementarity region.


Assuntos
Sondas de DNA/química , DNA Viral/análise , Compostos Férricos/química , Virus da Influenza A Subtipo H5N1/genética , Influenza Aviária/virologia , Hibridização de Ácido Nucleico/métodos , Animais , Sequência de Bases , Técnicas Biossensoriais/métodos , Aves/virologia , Técnicas Eletroquímicas/métodos , Virus da Influenza A Subtipo H5N1/isolamento & purificação , Influenza Aviária/diagnóstico , Limite de Detecção , Modelos Moleculares , Oxirredução
15.
Virol J ; 10: 243, 2013 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-23886034

RESUMO

BACKGROUND: The genetic basis for avian to mammalian host switching in influenza A virus is largely unknown. The human A/HK/156/1997 (H5N1) virus that transmitted from poultry possesses NS1 gene mutations F103L + M106I that are virulence determinants in the mouse model of pneumonia; however their individual roles have not been determined. The emergent A/Shanghai/patient1/2013(H7N9)-like viruses also possess these mutations which may contribute to their virulence and ability to switch species. METHODS: NS1 mutant viruses were constructed by reverse genetics and site directed mutagenesis on human and mouse-adapted backbones. Mouse infections assessed virulence, virus yield, tissue infection, and IFN induction. NS1 protein properties were assessed for subcellular distribution, IFN antagonism (mouse and human), CPSF30 and RIG-I domain binding, host transcription (microarray); and the natural prevalence of 103L and 106I mutants was assessed. RESULTS: Each of the F103L and M106I mutations contributes additively to virulence to reduce the lethal dose by >800 and >3,200 fold respectively by mediating alveolar tissue infection with >100 fold increased infectious yields. The 106I NS1 mutant lost CPSF binding but the 103L mutant maintained binding that correlated with an increased general decrease in host gene expression in human but not mouse cells. Each mutation positively modulated the inhibition of IFN induction in mouse cells and activation of the IFN-ß promoter in human cells but not in combination in human cells indicating negative epistasis. Each of the F103L and M106I mutations restored a defect in cytoplasmic localization of H5N1 NS1 in mouse cells. Human H1N1 and H3N2 NS1 proteins bound to the CARD, helicase and RD RIG-I domains, whereas the H5N1 NS1 with the same consensus 103F and 106M mutations did not bind these domains, which was totally or partially restored by the M106I or F103L mutations respectively. CONCLUSIONS: The F103L and M106I mutations in the H5N1 NS1 protein each increased IFN antagonism and mediated interstitial pneumonia in mice that was associated with increased cytoplasmic localization and altered host factor binding. These mutations may contribute to the ability of previous HPAI H5N1 and recent LPAI H7N9 and H6N1 (NS1-103L+106M) viruses to switch hosts and cause disease in humans.


Assuntos
Fator de Especificidade de Clivagem e Poliadenilação/metabolismo , RNA Helicases DEAD-box/metabolismo , Virus da Influenza A Subtipo H5N1/imunologia , Virus da Influenza A Subtipo H5N1/patogenicidade , Interferons/antagonistas & inibidores , Mutação de Sentido Incorreto , Proteínas não Estruturais Virais/metabolismo , Substituição de Aminoácidos , Animais , Proteína DEAD-box 58 , Feminino , Interações Hospedeiro-Patógeno , Humanos , Virus da Influenza A Subtipo H5N1/isolamento & purificação , Pulmão/patologia , Pulmão/virologia , Doenças Pulmonares Intersticiais/patologia , Doenças Pulmonares Intersticiais/virologia , Camundongos , Mutagênese Sítio-Dirigida , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Genética Reversa , Proteínas não Estruturais Virais/genética , Virulência , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
16.
BMC Biotechnol ; 13: 51, 2013 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-23777281

RESUMO

BACKGROUND: A promising way in diagnostic and therapeutic applications is the development of peptide amphiphiles (PAs). Peptides with a palmitic acid alkylchain were designed and characterized to study the effect of the structure modifications on self-assembling capabilities and the multiple binding capacity to hemagglutinin (HA), the surface protein of influenza virus type A. The peptide amphiphiles consists of a hydrophilic headgroup with a biological functionality of the peptide sequence and a chemically conjugated hydrophobic tail. In solution they self-assemble easily to micelles with a hydrophobic core surrounded by a closely packed peptide-shell. RESULTS: In this study the effect of a multiple peptide binding partner to the receptor binding site of HA could be determined with surface plasmon resonance measurements. The applied modification of the peptides causes signal amplification in relationship to the unmodified peptide wherein the high constant specificity persists. The molecular assembly of the peptides was characterized by the determination of critical micelle concentration (CMC) with concentration of 10⁻5 M and the colloidal size distribution. CONCLUSION: The modification of the physico-chemical parameters by producing peptide amphiphiles form monomeric structures which enhances the binding affinity and allows a better examination of the interaction with the virus surface protein hemagglutinin.


Assuntos
Hemaglutininas/química , Virus da Influenza A Subtipo H5N1/química , Micelas , Peptídeos/química , Proteínas Virais/química , Sequência de Aminoácidos , Hemaglutininas/metabolismo , Virus da Influenza A Subtipo H5N1/enzimologia , Virus da Influenza A Subtipo H5N1/isolamento & purificação , Dados de Sequência Molecular , Peptídeos/metabolismo , Ligação Proteica , Ressonância de Plasmônio de Superfície , Propriedades de Superfície , Tensoativos/química , Proteínas Virais/metabolismo
17.
Hong Kong Med J ; 19 Suppl 4: 29-35, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23775184

RESUMO

1. Cyclooxygenase-2 (COX-2), along with TNF-α and other proinflammatory cytokines, was hyperinduced in H5N1- infected macrophages in vitro and in epithelial cells of autopsied lung tissues of infected patients. 2. The COX-2 mediated amplification of the proinflammatory response is rapid, and the effects elicited by the H5N1-triggered proinflammatory cascade are broader than those arising from direct viral infection. 3. Selective COX-2 inhibitors suppress the H5N1- hyperinduced cytokines in the proinflammatory cascade.


Assuntos
Antivirais/farmacologia , Inibidores de Ciclo-Oxigenase 2/farmacologia , Virus da Influenza A Subtipo H5N1/enzimologia , Influenza Humana/enzimologia , Linhagem Celular Tumoral , Ciclo-Oxigenase 2/efeitos dos fármacos , Citocinas/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/virologia , Humanos , Inflamação/tratamento farmacológico , Inflamação/virologia , Virus da Influenza A Subtipo H5N1/efeitos dos fármacos , Virus da Influenza A Subtipo H5N1/isolamento & purificação , Influenza Humana/tratamento farmacológico , Influenza Humana/virologia , Pulmão/citologia , Pulmão/virologia , Macrófagos/metabolismo , Macrófagos/virologia , Fator de Necrose Tumoral alfa/metabolismo
18.
Virol J ; 10: 45, 2013 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-23374292

RESUMO

BACKGROUND: Wild ducks are the natural hosts of influenza A viruses. Duck influenza, therefore, has been believed inapparent infection with influenza A viruses, including highly pathogenic avian influenza viruses (HPAIVs) in chickens. In fact, ducks experimentally infected with an HPAIV strain, A/Hong Kong/483/1997 (H5N1) (HK483), did not show any clinical signs. Another HPAIV strain, A/whooper swan/Mongolia/3/2005 (H5N1) (MON3) isolated from a dead swan, however, caused neurological dysfunction and death in ducks. METHOD: To understand the mechanism whereby MON3 shows high pathogenicity in ducks, HK483, MON3, and twenty-four reassortants generated between these two H5N1 viruses were compared for their pathogenicity in domestic ducks. RESULTS: None of the ducks infected with MON3-based single-gene reassortants bearing the PB2, NP, or NS gene segment of HK483 died, and HK483-based single-gene reassortants bearing PB2, NP, or NS genes of MON3 were not pathogenic in ducks, suggesting that multiple gene segments contribute to the pathogenicity of MON3 in ducks. All the ducks infected with the reassortant bearing PB2, PA, HA, NP, and NS gene segments of MON3 died within five days post-inoculation, as did those infected with MON3. Each of the viruses was assessed for replication in ducks three days post-inoculation. MON3 and multi-gene reassortants pathogenic in ducks were recovered from all of the tissues examined and replicated with high titers in the brains and lungs. CONCLUSION: The present results indicate that multigenic factors are responsible for efficient replication of MON3 in ducks. In particular, virus growth in the brain might correlate with neurological dysfunction and the disease severity.


Assuntos
Glicoproteínas de Hemaglutininação de Vírus da Influenza/metabolismo , Virus da Influenza A Subtipo H5N1/patogenicidade , Influenza Aviária/virologia , Doenças das Aves Domésticas/virologia , Proteínas de Ligação a RNA/metabolismo , RNA Polimerase Dependente de RNA/metabolismo , Proteínas do Core Viral/metabolismo , Proteínas não Estruturais Virais/metabolismo , Proteínas Virais/metabolismo , Animais , Patos , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Virus da Influenza A Subtipo H5N1/genética , Virus da Influenza A Subtipo H5N1/isolamento & purificação , Virus da Influenza A Subtipo H5N1/fisiologia , Proteínas do Nucleocapsídeo , Proteínas de Ligação a RNA/genética , RNA Polimerase Dependente de RNA/genética , Vírus Reordenados/genética , Vírus Reordenados/isolamento & purificação , Vírus Reordenados/patogenicidade , Vírus Reordenados/fisiologia , Proteínas do Core Viral/genética , Proteínas não Estruturais Virais/genética , Proteínas Virais/genética , Virulência , Replicação Viral
19.
Biosens Bioelectron ; 43: 412-8, 2013 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-23356997

RESUMO

A Rat Basophilic Leukemia (RBL) cell sensor is developed for the detection and identification of pathogenic viruses. Recombinant sdAb-Fc antibodies were constructed by linking virus-specific single domain antibody to mouse IgE-Fc fragment. The sdAb-Fc can bind to FcεRI receptors on RBL cells and can be cross-linked by target viruses leading to cell activation and Ca(2+) influx reflected by the increase of intracellular fluorescence. The responses of RBL cells to viruses in real time could be observed using fluorescence microscopy. 10(3) TCID50 of H5N1 viruses and 10 LD50 of rabies viruses could be detected in less than three minutes. An excess quantity of non-relevant viruses did not interfere with the recognition of target viruses.


Assuntos
Bioensaio/instrumentação , Técnicas Biossensoriais/instrumentação , Virus da Influenza A Subtipo H5N1/isolamento & purificação , Leucemia Basofílica Aguda/virologia , Vírus da Raiva/isolamento & purificação , Carga Viral/instrumentação , Animais , Linhagem Celular Tumoral , Desenho de Equipamento , Análise de Falha de Equipamento , Ratos
20.
Mod Pathol ; 26(3): 357-69, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23174938

RESUMO

Highly pathogenic avian H5N1 influenza virus (H5N1) infection in humans causes acute respiratory distress syndrome, leading to multiple organ failure. Five fatal cases of H5N1 infection in Vietnam were analyzed pathologically to reveal virus distribution, and local proinflammatory cytokine and chemokine expression profiles in formalin-fixed, paraffin-embedded lung tissues. Our main histopathological findings showed diffuse alveolar damage in the lungs. The infiltration of myeloperoxidase-positive and/or CD68 (clone KP-1)-positive neutrophils and monocytes/macrophages was remarkable in the alveolar septa and alveolar spaces. Immunohistochemistry revealed that H5N1 mainly infected alveolar epithelial cells and monocytes/macrophages in lungs. H5N1 replication was confirmed by detecting H5N1 mRNA in epithelial cells using in situ hybridization. Quantitation of H5N1 RNA using quantitative reverse transcription PCR assays revealed that the level of H5N1 RNA was increased in cases during early phases of the disease. We quantified the expression of tumor necrosis factor-alpha (TNF-α), interleukin (IL)-6, IL-8, regulated on activation normal T-cell expressed and secreted (commonly known as RANTES), and interferon-gamma-inducible protein of 10 kDa (IP-10) in formalin-fixed, paraffin-embedded lung sections. Their expression levels correlated with H5N1 RNA copy numbers detected in the same lung region. Double immunofluorescence staining revealed that TNF-α, IL-6, IL-8 and IP-10 were expressed in epithelial cells and/or monocytes/macrophages. In particular, IL-6 was also expressed in endothelial cells. The dissemination of H5N1 beyond respiratory organs was not confirmed in two cases examined in this study.


Assuntos
Virus da Influenza A Subtipo H5N1/isolamento & purificação , Influenza Humana/patologia , Pulmão/patologia , Células Epiteliais Alveolares/imunologia , Células Epiteliais Alveolares/patologia , Células Epiteliais Alveolares/virologia , Antígenos CD/análise , Antígenos de Diferenciação Mielomonocítica/análise , Biomarcadores/análise , Quimiocinas/análise , Quimiocinas/genética , Criança , Pré-Escolar , Citocinas/análise , Citocinas/genética , Evolução Fatal , Feminino , Fixadores , Imunofluorescência , Formaldeído , Humanos , Imuno-Histoquímica , Hibridização In Situ , Mediadores da Inflamação/análise , Virus da Influenza A Subtipo H5N1/genética , Virus da Influenza A Subtipo H5N1/patogenicidade , Influenza Humana/genética , Influenza Humana/imunologia , Influenza Humana/virologia , Pulmão/imunologia , Pulmão/virologia , Macrófagos/imunologia , Macrófagos/patologia , Macrófagos/virologia , Masculino , Neutrófilos/imunologia , Neutrófilos/patologia , Neutrófilos/virologia , Inclusão em Parafina , Peroxidase/análise , RNA Mensageiro/análise , RNA Viral/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fixação de Tecidos/métodos , Vietnã , Carga Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA