Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
PLoS Pathog ; 17(12): e1010174, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34919598

RESUMO

The mechanisms and consequences of genome evolution on viral fitness following host shifts are poorly understood. In addition, viral fitness -the ability of an organism to reproduce and survive- is multifactorial and thus difficult to quantify. Influenza A viruses (IAVs) circulate broadly among wild birds and have jumped into and become endemic in multiple mammalian hosts, including humans, pigs, dogs, seals, and horses. H3N8 equine influenza virus (EIV) is an endemic virus of horses that originated in birds and has been circulating uninterruptedly in equine populations since the early 1960s. Here, we used EIV to quantify changes in infection phenotype associated to viral fitness due to genome-wide changes acquired during long-term adaptation. We performed experimental infections of two mammalian cell lines and equine tracheal explants using the earliest H3N8 EIV isolated (A/equine/Uruguay/63 [EIV/63]), and A/equine/Ohio/2003 (EIV/2003), a monophyletic descendant of EIV/63 isolated 40 years after the emergence of H3N8 EIV. We show that EIV/2003 exhibits increased resistance to interferon, enhanced viral replication, and a more efficient cell-to-cell spread in cells and tissues. Transcriptomics analyses revealed virus-specific responses to each virus, mainly affecting host immunity and inflammation. Image analyses of infected equine respiratory explants showed that despite replicating at higher levels and spreading over larger areas of the respiratory epithelium, EIV/2003 induced milder lesions compared to EIV/63, suggesting that adaptation led to reduced tissue pathogenicity. Our results reveal previously unknown links between virus genotype and the host response to infection, providing new insights on the relationship between virus evolution and fitness.


Assuntos
Adaptação Fisiológica/fisiologia , Interações Hospedeiro-Patógeno/fisiologia , Vírus da Influenza A Subtipo H3N8/fisiologia , Vírus da Influenza A Subtipo H3N8/patogenicidade , Infecções por Orthomyxoviridae/virologia , Animais , Aptidão Genética/fisiologia , Cavalos
2.
Viruses ; 13(8)2021 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-34452416

RESUMO

Vaccination is an effective method for the prevention of influenza virus infection. Many manufacturers use embryonated chicken eggs (ECE) for the propagation of vaccine strains. However, the adaptation of viral strains during subsequent passages can lead to additional virus evolution and lower effectiveness of the resulting vaccines. In our study, we analyzed the distribution of single nucleotide variants (SNVs) of equine influenza virus (EIV) during passaging in ECE. Viral RNA from passage 0 (nasal swabs), passage 2 and 5 was sequenced using next generation technology. In total, 50 SNVs with an occurrence frequency above 2% were observed, 29 of which resulted in amino acid changes. The highest variability was found in passage 2, with the most variable segment being IV encoding hemagglutinin (HA). Three variants, HA (W222G), PB2 (A377E) and PA (R531K), had clearly increased frequency with the subsequent passages, becoming dominant. None of the five nonsynonymous HA variants directly affected the major antigenic sites; however, S227P was previously reported to influence the antigenicity of EIV. Our results suggest that although host-specific adaptation was observed in low passages of EIV in ECE, it should not pose a significant risk to influenza vaccine efficacy.


Assuntos
Ovos/virologia , Vírus da Influenza A Subtipo H3N8/genética , Vírus da Influenza A Subtipo H3N8/fisiologia , Polimorfismo de Nucleotídeo Único , Quase-Espécies/genética , RNA Viral/genética , Adaptação Fisiológica/genética , Animais , Galinhas/imunologia , Cavalos/virologia , Filogenia , Análise de Sequência de DNA , Inoculações Seriadas
3.
Viruses ; 13(8)2021 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-34452521

RESUMO

Equine influenza virus (EIV) is a constantly evolving viral pathogen that is responsible for yearly outbreaks of respiratory disease in horses termed equine influenza (EI). There is currently no evidence of circulation of the original H7N7 strain of EIV worldwide; however, the EIV H3N8 strain, which was first isolated in the early 1960s, remains a major threat to most of the world's horse populations. It can also infect dogs. The ability of EIV to constantly accumulate mutations in its antibody-binding sites enables it to evade host protective immunity, making it a successful viral pathogen. Clinical and virological protection against EIV is achieved by stimulation of strong cellular and humoral immunity in vaccinated horses. However, despite EI vaccine updates over the years, EIV remains relevant, because the protective effects of vaccines decay and permit subclinical infections that facilitate transmission into susceptible populations. In this review, we describe how the evolution of EIV drives repeated EI outbreaks even in horse populations with supposedly high vaccination coverage. Next, we discuss the approaches employed to develop efficacious EI vaccines for commercial use and the existing system for recommendations on updating vaccines based on available clinical and virological data to improve protective immunity in vaccinated horse populations. Understanding how EIV biology can be better harnessed to improve EI vaccines is central to controlling EI.


Assuntos
Doenças dos Cavalos/prevenção & controle , Vírus da Influenza A Subtipo H3N8/imunologia , Vírus da Influenza A Subtipo H7N7/imunologia , Vacinas contra Influenza/imunologia , Infecções por Orthomyxoviridae/veterinária , Animais , Anticorpos Antivirais/imunologia , Doenças dos Cavalos/imunologia , Doenças dos Cavalos/virologia , Cavalos , Vírus da Influenza A Subtipo H3N8/genética , Vírus da Influenza A Subtipo H3N8/fisiologia , Vírus da Influenza A Subtipo H7N7/genética , Vírus da Influenza A Subtipo H7N7/fisiologia , Vacinas contra Influenza/administração & dosagem , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Infecções por Orthomyxoviridae/virologia
4.
Viruses ; 13(3)2021 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-33808674

RESUMO

Reassortment among co-infecting influenza A viruses (IAVs) is an important source of viral diversity and can facilitate expansion into novel host species. Indeed, reassortment played a key role in the evolution of the last three pandemic IAVs. Observed patterns of reassortment within a coinfected host are likely to be shaped by several factors, including viral load, the extent of viral mixing within the host and the stringency of selection. These factors in turn are expected to vary among the diverse host species that IAV infects. To investigate host differences in IAV reassortment, here we examined reassortment of two distinct avian IAVs within their natural host (mallards) and a mammalian model system (guinea pigs). Animals were co-inoculated with A/wildbird/California/187718-36/2008 (H3N8) and A/mallard/Colorado/P66F1-5/2008 (H4N6) viruses. Longitudinal samples were collected from the cloaca of mallards or the nasal tract of guinea pigs and viral genetic exchange was monitored by genotyping clonal isolates from these samples. Relative to those in guinea pigs, viral populations in mallards showed higher frequencies of reassortant genotypes and were characterized by higher genotype richness and diversity. In line with these observations, analysis of pairwise segment combinations revealed lower linkage disequilibrium in mallards as compared to guinea pigs. No clear longitudinal patterns in richness, diversity or linkage disequilibrium were present in either host. Our results reveal mallards to be a highly permissive host for IAV reassortment and suggest that reduced viral mixing limits avian IAV reassortment in a mammalian host.


Assuntos
Vírus da Influenza A Subtipo H3N8/fisiologia , Influenza Aviária , Infecções por Orthomyxoviridae , Animais , Cães , Patos , Feminino , Cobaias , Influenza Aviária/epidemiologia , Influenza Aviária/virologia , Estudos Longitudinais , Células Madin Darby de Rim Canino , Infecções por Orthomyxoviridae/epidemiologia , Infecções por Orthomyxoviridae/veterinária , Infecções por Orthomyxoviridae/virologia , Vírus Reordenados
5.
J Virol ; 94(15)2020 07 16.
Artigo em Inglês | MEDLINE | ID: mdl-32461313

RESUMO

Equine-origin H3N8 and avian-origin H3N2 canine influenza viruses (CIVs) prevalent in dogs are thought to pose a public health threat arising from intimate contact between dogs and humans. However, our understanding of CIV virulence is still limited. Influenza A virus PA-X is a fusion protein encoded in part by a +1 frameshifted open reading frame (X-ORF) in segment 3. The X-ORF can be translated in full-length (61-amino-acid) or truncated (41-amino-acid) form. Genetic analysis indicated that the X-ORFs of equine H3N8 and avian H3N2 influenza viruses encoded 61 amino acids but were truncated after introduction into dogs. To determine the effect of PA-X truncation on the biological characteristics of CIVs, we constructed four recombinant viruses on H3N8 and H3N2 CIV backgrounds bearing truncated or full-length PA-Xs. We observed that truncation of PA-X increased growth of both H3N8 and H3N2 CIVs in MDCK cells and suppressed expression from cotransfected plasmids in MDCK cells. Furthermore, truncation of PA-X enhanced viral pathogenicity in dogs, as shown by aggravated clinical symptoms and histopathological changes, increased viral replication in the respiratory system, and prolonged virus shedding. Additionally, CIVs with truncated PA-Xs were transmitted more efficiently in dogs. Global gene expression profiling of the lungs of infected dogs revealed that differentially expressed genes were mainly associated with inflammatory responses, which might contribute to the pathogenicity of PA-X-truncated CIVs. Our findings revealed that truncation of PA-X might be important for the adaptation of influenza viruses to dogs.IMPORTANCE Epidemics of equine-origin H3N8 and avian-origin H3N2 influenza viruses in canine populations are examples of successful cross-species transmission of influenza A viruses. Genetic analysis showed that the PA-X genes of equine H3N8 or avian H3N2 influenza viruses were full-length, with X-ORFs encoding 61 amino acids; however, those of equine-origin H3N8 or avian-origin H3N2 CIVs were truncated, suggesting that PA-X truncation occurred after transmission to dogs. In this study, we extended the PA-X genes of H3N8 and H3N2 CIVs and compared the biological characteristics of CIVs bearing different lengths of PA-X. We demonstrated that for both H3N8 and H3N2 viruses, truncation of PA-X increased virus yields in MDCK cells and enhanced viral replication, pathogenicity, and transmission in dogs. These results might reflect enhanced suppression of host gene expression and upregulation of genes related to inflammatory responses. Collectively, our data partially explain the conservation of truncated PA-X in CIVs.


Assuntos
Vírus da Influenza A Subtipo H3N2 , Vírus da Influenza A Subtipo H3N8 , Infecções por Orthomyxoviridae , Proteínas Repressoras/metabolismo , Proteínas não Estruturais Virais/metabolismo , Replicação Viral , Eliminação de Partículas Virais , Animais , Cães , Células HEK293 , Humanos , Vírus da Influenza A Subtipo H3N2/patogenicidade , Vírus da Influenza A Subtipo H3N2/fisiologia , Vírus da Influenza A Subtipo H3N8/patogenicidade , Vírus da Influenza A Subtipo H3N8/fisiologia , Células Madin Darby de Rim Canino , Infecções por Orthomyxoviridae/metabolismo , Infecções por Orthomyxoviridae/transmissão
6.
Methods Mol Biol ; 2123: 355-360, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32170701

RESUMO

Equine influenza virus (EIV) is a common respiratory pathogen of horses and other equids in most parts of the world. EIV are Type A influenza viruses and two subtypes are known: H3N8 and H7N7. Both are believed to have evolved from avian influenza virus ancestors. The H3N8 subtype circulates widely, but the H7N7 subtype is thought to be extinct. The clinical disease in horses, caused by either subtype, is an upper respiratory infection of varying severity depending upon the immune status of the individual animal. It is not normally life-threatening in itself except in very young foals; however it predisposes infected equids to secondary infections capable of producing life-threatening pneumonias. Vaccines are available and widely used in some horse populations, but their effectiveness is limited by antigenic drift and other factors, and vaccinated animals with subclinical infections have been responsible for introduction of EIV into susceptible populations. EIV has spread into canines.


Assuntos
Doenças dos Cavalos/virologia , Cavalos/virologia , Vírus da Influenza A Subtipo H3N8/fisiologia , Vírus da Influenza A Subtipo H7N7/fisiologia , Infecções por Orthomyxoviridae/veterinária , Infecções por Orthomyxoviridae/virologia , Animais , Doenças dos Cavalos/epidemiologia , Doenças dos Cavalos/imunologia , Doenças dos Cavalos/transmissão , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/transmissão , Vigilância da População , Vacinação/veterinária
7.
Methods Mol Biol ; 2123: 401-412, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32170706

RESUMO

Serologic tests for equine influenza virus (EIV) antibodies are used for many purposes, including retrospective diagnosis, subtyping of virus isolates, antigenic comparison of different virus strains, and measurement of immune responses to EIV vaccines. The hemagglutination inhibition (HI) assay, single radial hemolysis (SRH), and serum micro-neutralization tests are the most widely used for these purposes and are described here. The presence of inhibitors of hemagglutination in equine serum complicates interpretation of HI assay results, and there are alternative protocols (receptor-destroying enzyme, periodate, trypsin-periodate) for their removal. With the EIV H3N8 strains in particular, equine antibody titers may be magnified by pre-treating the HI test antigen with Tween-80 and ether. The SRH assay offers stronger correlations between serum antibody titers and protection from disease. Other tests are sometimes used for specialized purposes such as the neuraminidase-inhibition assay for subtyping, or ELISA for measuring different specific antibody isotypes, and are not described here.


Assuntos
Cavalos/sangue , Cavalos/virologia , Vírus da Influenza A Subtipo H3N8/fisiologia , Testes Sorológicos/métodos , Animais , Cães , Testes de Inibição da Hemaglutinação , Hemólise , Doenças dos Cavalos/sangue , Doenças dos Cavalos/virologia , Células Madin Darby de Rim Canino , Testes de Neutralização , Infecções por Orthomyxoviridae/sangue , Infecções por Orthomyxoviridae/veterinária , Infecções por Orthomyxoviridae/virologia , Soro/metabolismo
8.
Vet Res ; 50(1): 87, 2019 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-31666126

RESUMO

Prior to the emergence of H3N8 canine influenza virus (CIV) and the latest avian-origin H3N2 CIV, there was no evidence of a circulating canine-specific influenza virus. Molecular and epidemiological evidence suggest that H3N8 CIV emerged from H3N8 equine influenza virus (EIV). This host-range shift of EIV from equine to canine hosts and its subsequent establishment as an enzootic CIV is unique because this host-range shift was from one mammalian host to another. To further understand this host-range shift, we conducted a comprehensive phylodynamic analysis using all the available whole-genome sequences of H3N8 CIV. We found that (1) the emergence of H3N8 CIV from H3N8 EIV occurred in approximately 2002; (2) this interspecies transmission was by a reassortant virus of the circulating Florida-1 clade H3N8 EIV; (3) once in the canine species, H3N8 CIV spread efficiently and remained an enzootic virus; (4) H3N8 CIV evolved and diverged into multiple clades or sublineages, with intra and inter-lineage reassortment. Our results provide a framework to understand the molecular basis of host-range shifts of influenza viruses and that dogs are potential "mixing vessels" for the establishment of novel influenza viruses.


Assuntos
Evolução Biológica , Doenças do Cão/virologia , Doenças dos Cavalos/virologia , Especificidade de Hospedeiro/fisiologia , Vírus da Influenza A Subtipo H3N8/fisiologia , Infecções por Orthomyxoviridae/veterinária , Adaptação Biológica , Animais , Cães , Cavalos , Infecções por Orthomyxoviridae/virologia , Filogenia
9.
Acta Virol ; 62(3): 266-276, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30160142

RESUMO

Avian influenza A viruses (IAVs) are able to overcome the interspecies barrier and adapt to the new non-avian host. The process of adaptation requires the adaptive changes of IAV genome resulting in amino acid substitutions. The aim of this work was the description of amino acid substitutions in avian influenza A viruses (IAVs) occurring during their adaptation to equine host. Today, viruses of the equine influenza H3N8 subtype, first isolated in 1963, represent a single genetic lineage of IAV causing a respiratory disease in horses. We compared the amino acid sequences of the conserved proteins PB2, PB1, PA, NP, M1, M2, NS1 and NEP of equine influenza H3N8 subtype IAV with sequences of avian viruses, both available in the NCBI's Influenza Virus Resource Database. The amino acid substitutions persisting in equine IAV isolates and occurring in avian IAV at f both hosts.


Assuntos
Doenças dos Cavalos/virologia , Vírus da Influenza A Subtipo H3N8/genética , Infecções por Orthomyxoviridae/veterinária , Adaptação Fisiológica , Substituição de Aminoácidos , Animais , Marcadores Genéticos , Cavalos , Humanos , Vírus da Influenza A Subtipo H3N8/fisiologia , Influenza Humana/virologia , Infecções por Orthomyxoviridae/virologia , Especificidade da Espécie , Proteínas Virais/genética
10.
Viruses ; 10(5)2018 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-29772683

RESUMO

Tetherin/BST-2/CD317 is an interferon-induced host restriction factor that can block the budding of enveloped viruses by tethering them to the cell surface. Many viruses use certain proteins to counteract restriction by tetherin from their natural hosts, but not from other species. The influenza A virus (FLUAV) has a wide range of subtypes with different host tropisms. Human tetherin (huTHN) has been reported to restrict only specific FLUAV strains and the viral hemagglutinin (HA) and neuraminidase (NA) genes determine the sensitivity to huTHN. Whether tetherins from other hosts can block human FLUAV is still unknown. Here, we evaluate the impact of equine tetherin (eqTHN) and huTHN on the replication of A/Sichuan/1/2009 (H1N1) and A/equine/Xinjiang/1/2007 (H3N8) strains. Our results show that eqTHN had higher restriction activity towards both viruses, and its shorter cytoplasmic tail contributed to that activity. We further demonstrated that HA and NA of A/Hamburg/4/2009 (H1N1) could counteract eqTHN. Notably, our results indicate that four amino acids, 13T and 49L of HA and 32T and 80V of NA, were involved in blocking the restriction activity of eqTHN. These findings reveal interspecies restriction by eqTHN towards FLUAV, and the role of the HA and NA proteins in overcoming this restriction.


Assuntos
Antígeno 2 do Estroma da Médula Óssea/metabolismo , Glicoproteínas de Hemaglutininação de Vírus da Influenza/metabolismo , Interações Hospedeiro-Patógeno , Vírus da Influenza A Subtipo H1N1/metabolismo , Neuraminidase/metabolismo , Animais , Antígeno 2 do Estroma da Médula Óssea/química , Antígeno 2 do Estroma da Médula Óssea/genética , Cães , Glicosilfosfatidilinositóis/fisiologia , Células HEK293 , Glicoproteínas de Hemaglutininação de Vírus da Influenza/química , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Cavalos , Humanos , Vírus da Influenza A Subtipo H1N1/genética , Vírus da Influenza A Subtipo H3N8/genética , Vírus da Influenza A Subtipo H3N8/fisiologia , Células Madin Darby de Rim Canino , Modelos Moleculares , Mutação , Neuraminidase/genética , Relação Estrutura-Atividade , Liberação de Vírus
11.
Arch Virol ; 163(7): 1955-1960, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29556821

RESUMO

Previous studies have shown that dogs are susceptible to influenza A viruses, and the close contact between dogs and humans poses a threat to public health. In 2015, a novel H3N8 influenza virus was isolated from a dog in eastern China. This strain was characterized by whole-genome sequencing with subsequent phylogenetic analysis and genetic comparison and found to be most closely related to avian influenza viruses co-circulating in China. It was able to replicate in mice without prior adaptation. The continued circulation of this novel H3N8 influenza virus in dogs could endanger other mammalian species.


Assuntos
Doenças do Cão/virologia , Cães/virologia , Vírus da Influenza A Subtipo H3N8/genética , Vírus da Influenza A Subtipo H3N8/isolamento & purificação , Infecções por Orthomyxoviridae/veterinária , Animais , Aves/virologia , China/epidemiologia , Doenças do Cão/epidemiologia , Genoma Viral , Humanos , Vírus da Influenza A Subtipo H3N8/classificação , Vírus da Influenza A Subtipo H3N8/fisiologia , Influenza Humana/epidemiologia , Influenza Humana/transmissão , Influenza Humana/virologia , Camundongos , Infecções por Orthomyxoviridae/epidemiologia , Infecções por Orthomyxoviridae/transmissão , Infecções por Orthomyxoviridae/virologia , Filogenia , Replicação Viral , Sequenciamento Completo do Genoma
12.
Prev Vet Med ; 151: 52-56, 2018 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-29496107

RESUMO

Equine influenza is a highly contagious respiratory disease considered the most important respiratory disease in equids. Although influenza A virus (IAV) has caused outbreaks in equids worldwide, surveillance in these species in Spain has not been conducted. A cross-sectional study was carried out to determine the individual and herd prevalence of antibodies against H3N8 and H7N7 IAV in equids in Andalusia (southern Spain). Antibodies againsts IAV were measured by the single radial haemolysis assay. A spatial scan statistical analysis was carried out using a Bernoulli model. Risk factors associated with IAV infection were assessed by multivariate analysis. Antibodies to H3N8 IAV were detected in 241 out of 464 unvaccinated equids (51.9%; 95% CI: 47.4-56.5). Seropositivity against the H7N7 subtype IAV was not found in any of the analysed animals. Significantly higher seropositivity was found in geriatric (OR = 6.1, P = 0.008, 95% CI = 1.6-23.1) and adult (OR = 4.8, P < 0.001, 95% CI = 2.5-9.0) equids compared to young animals. Specific antibodies against A/equine/Shropshire/2010 (H3N8) or A/equine/Newmarket/5/2003 (H3N8) only were confirmed in 11 and 45 of the animals, respectively. The spatial analysis showed a statistically significant cluster centred in the west part of Andalusia. The results confirmed widespread H3N8 subtype IAV exposure in equine species in Andalusia. Conversely, the absence of seropositivity against H7N7 IAV obtained in the present study suggests that this subtype has not circulated in southern Spain in recent years. Because of the animal health and economic consequences of IAV in equids, further surveillance and molecular studies are required to monitor and characterize the most prevalent IAV circulating in these species in Spain.


Assuntos
Doenças dos Cavalos/epidemiologia , Infecções por Orthomyxoviridae/veterinária , Animais , Estudos Transversais , Doenças dos Cavalos/virologia , Cavalos , Vírus da Influenza A Subtipo H3N8/fisiologia , Vírus da Influenza A Subtipo H7N7/fisiologia , Infecções por Orthomyxoviridae/epidemiologia , Infecções por Orthomyxoviridae/virologia , Prevalência , Fatores de Risco , Estudos Soroepidemiológicos , Espanha/epidemiologia
13.
Vaccine ; 36(22): 3101-3111, 2018 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-28571695

RESUMO

The influenza vaccine manufacturing industry is looking for production cell lines that are easily scalable, highly permissive to multiple viruses, and more effective in term of viral productivity. One critical characteristic of such cell lines is their ability to grow in suspension, in serum free conditions and at high cell densities. Influenza virus causing severe epidemics both in human and animals is an important threat to world healthcare. The repetitive apparition of influenza pandemic outbreaks in the last 20years explains that manufacturing sector is still looking for more effective production processes to replace/supplement embryonated egg-based process. Cell-based production strategy, with a focus on avian cell lines, is one of the promising solutions. Three avian cell lines, namely duck EB66®cells (Valneva), duck AGE.CR® cells (Probiogen) and quail QOR/2E11 cells (Baxter), are now competing with traditional mammalian cell platforms (Vero and MDCK cells) used for influenza vaccine productions and are currently at advance stage of commercial development for the manufacture of influenza vaccines. The DuckCelt®-T17 cell line presented in this work is a novel avian cell line developed by Transgene. This cell line was generated from primary embryo duck cells with the constitutive expression of the duck telomerase reverse transcriptase (dTERT). The DuckCelt®-T17 cells were able to grow in batch suspension cultures and serum-free conditions up to 6.5×106cell/ml and were easily scaled from 10ml up to 3l bioreactor. In the present study, DuckCelt®-T17 cell line was tested for its abilities to produce various human, avian and porcine influenza strains. Most of the viral strains were produced at significant infectious titers (>5.8 log TCID50/ml) with optimization of the infection conditions. Human strains H1N1 and H3N2, as well as all the avian strains tested (H5N2, H7N1, H3N8, H11N9, H12N5) were the most efficiently produced with highest titre reached of 9.05 log TCID50/ml for A/Panama/2007/99 influenza H3N2. Porcine strains were also greatly rescued with titres from 4 to 7 log TCID50/ml depending of the subtypes. Interestingly, viral kinetics showed maximal titers reached at 24h post-infection for most of the strains, allowing early harvest time (Time Of Harvest: TOH). The B strains present specific production kinetics with a delay of 24h before reaching the maximal viral particle release. Process optimization on H1N1 2009 human pandemic strain allowed identifying best operating conditions for production (MOI, trypsin concentration, cell density at infection) allowing improving the production level by 2 log. Our results suggest that the DuckCelt®-T17 cell line is a very promising platform for industrial production of influenza viruses and particularly for avian viral strains.


Assuntos
Técnicas de Cultura de Células/métodos , Linhagem Celular , Orthomyxoviridae/crescimento & desenvolvimento , Cultura de Vírus/métodos , Replicação Viral , Animais , Reatores Biológicos , Patos , Vírus da Influenza A Subtipo H1N1/crescimento & desenvolvimento , Vírus da Influenza A Subtipo H1N1/fisiologia , Vírus da Influenza A Subtipo H3N2/crescimento & desenvolvimento , Vírus da Influenza A Subtipo H3N2/fisiologia , Vírus da Influenza A Subtipo H3N8/crescimento & desenvolvimento , Vírus da Influenza A Subtipo H3N8/fisiologia , Vírus da Influenza A Subtipo H5N2/crescimento & desenvolvimento , Vírus da Influenza A Subtipo H5N2/fisiologia , Vírus da Influenza A Subtipo H7N1/crescimento & desenvolvimento , Vírus da Influenza A Subtipo H7N1/fisiologia , Vacinas contra Influenza , Orthomyxoviridae/fisiologia
14.
J Virol ; 91(22)2017 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-28835506

RESUMO

Canine influenza viruses (CIVs) are the causative agents of canine influenza, a contagious respiratory disease in dogs, and include the equine-origin H3N8 and the avian-origin H3N2 viruses. Influenza A virus (IAV) nonstructural protein 1 (NS1) is a virulence factor essential for counteracting the innate immune response. Here, we evaluated the ability of H3N8 CIV NS1 to inhibit host innate immune responses. We found that H3N8 CIV NS1 was able to efficiently counteract interferon (IFN) responses but was unable to block general gene expression in human or canine cells. Such ability was restored by a single amino acid substitution in position 186 (K186E) that resulted in NS1 binding to the 30-kDa subunit of the cleavage and polyadenylation specificity factor (CPSF30), a cellular protein involved in pre-mRNA processing. We also examined the frequency distribution of K186 and E186 among H3N8 CIVs and equine influenza viruses (EIVs), the ancestors of H3N8 CIV, and experimentally determined the impact of amino acid 186 in the ability of different CIV and EIV NS1s to inhibit general gene expression. In all cases, the presence of E186 was responsible for the control of host gene expression. In contrast, the NS1 protein of H3N2 CIV harbors E186 and blocks general gene expression in canine cells. Altogether, our results confirm previous studies on the strain-dependent ability of NS1 to block general gene expression. Moreover, the observed polymorphism on amino acid 186 between H3N8 and H3N2 CIVs might be the result of adaptive changes acquired during long-term circulation of avian-origin IAVs in mammals.IMPORTANCE Canine influenza is a respiratory disease of dogs caused by two CIV subtypes, the H3N8 and H3N2 viruses, of equine and avian origins, respectively. Influenza NS1 is the main viral factor responsible for the control of host innate immune responses, and changes in NS1 can play an important role in host adaptation. Here we assessed the ability of H3N8 CIV NS1 to inhibit host innate immune responses and gene expression. The H3N8 CIV NS1 did not block host gene expression, but this activity was restored by a single amino acid substitution (K186E), which was responsible for NS1 binding to the host factor CPSF30. In contrast, the H3N2 CIV NS1, which contains E186, blocks general gene expression. Our results suggest that the ability to block host gene expression is not required for influenza virus replication in mammals but might be important in the long-term adaptation of avian-origin influenza viruses to mammals.


Assuntos
Substituição de Aminoácidos , Regulação da Expressão Gênica/imunologia , Interações Hospedeiro-Patógeno/imunologia , Vírus da Influenza A Subtipo H3N8/fisiologia , Mutação de Sentido Incorreto , Infecções por Orthomyxoviridae/imunologia , Proteínas não Estruturais Virais/imunologia , Animais , Cães , Células HEK293 , Interações Hospedeiro-Patógeno/genética , Humanos , Células Madin Darby de Rim Canino , Infecções por Orthomyxoviridae/genética , Proteínas não Estruturais Virais/genética
15.
Biophys Chem ; 231: 116-124, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28410940

RESUMO

BACKGROUND: Avian influenza A viruses can cross naturally into mammals and cause severe diseases, as observed for H5N1. The high lethality of human infections causes major concerns about the real risk of a possible pandemic of severe diseases to which human susceptibility may be high and universal. High hydrostatic pressure (HHP) is a valuable tool for studies regarding the folding of proteins and the assembly of macromolecular structures such as viruses; furthermore, HHP has already been demonstrated to promote viral inactivation. METHODS: Here, we investigated the structural stability of avian and human influenza viruses using spectroscopic and light-scattering techniques. We found that both particles have similar structural stabilities and that HHP promotes structural changes. RESULTS: HHP induced slight structural changes to both human and avian influenza viruses, and these changes were largely reversible when the pressure returned to its initial level. The spectroscopic data showed that H3N2 was more pressure-sensitive than H3N8. Structural changes did not predict changes in protein function, as H3N2 fusion activity was not affected, while H3N8 fusion activity drastically decreased. The fusion activity of H1N1 was also strongly affected by HHP. In all cases, HHP caused inactivation of the different influenza viruses. CONCLUSIONS: HHP may be a useful tool for vaccine development, as it induces minor and reversible structural changes that may be associated with partial preservation of viral biological activities and may potentiate their immunogenic response while abolishing their infectivity. We also confirmed that, although pressure does not promote drastic changes in viral particle structure, it can distinctly affect viral fusion activity.


Assuntos
Vírus da Influenza A/química , Animais , Guanidina/química , Humanos , Pressão Hidrostática , Vírus da Influenza A Subtipo H1N1/química , Vírus da Influenza A Subtipo H1N1/fisiologia , Vírus da Influenza A Subtipo H3N2/química , Vírus da Influenza A Subtipo H3N2/fisiologia , Vírus da Influenza A Subtipo H3N8/química , Vírus da Influenza A Subtipo H3N8/fisiologia , Vírus da Influenza A/fisiologia , Temperatura , Ureia/química , Vacinas/imunologia , Inativação de Vírus
16.
Arch Virol ; 162(1): 13-21, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27400993

RESUMO

The non-structural protein of influenza A virus (NS1A protein) is a multifunctional protein that antagonizes host antiviral responses and contributes to efficient viral replication during infection. However, most of its functions have been elucidated by generating recombinant viruses expressing mutated NS1 proteins that do not exist in nature. Recently, the novel H3N8 A/Equine/Kyonggi/SA1/2011 (KG11) influenza virus was isolated in Korea from horses showing respiratory disease symptoms. KG11 virus contains a naturally truncated NS gene segment with the truncation in the NS1A coding region, resulting in truncation of the effector domain of the NS1A protein. Using this KG11 virus, we investigated the role of truncated NS1A protein in the virus life cycle and its effect on host immune responses were compared to the A/Equine/Miami/1/1963 H3N8 (MA63) virus, which encodes a full-length NS1A protein. The replication of KG11 virus was attenuated by 2 logs in multiple-cycle growth, and its plaque size was significantly smaller than that of the MA63 virus. To understand the attenuation of KG11 virus, we evaluated the level of activation in Akt and interferon regulatory factor 3 (IRF-3) pathways and measured the induction of downstream genes. Our results showed that the activation of Akt was reduced, whereas phosphorylation of IRF-3 was increased in cells infected with KG11 virus when compared to MA63-virus-infected cells. We also determined that the expression of antiviral and pro-inflammatory genes was significantly increased. Taken together, these results revealed that the KG11 virus expressing the naturally truncated NS1A protein impairs the inhibition of host antiviral responses, thereby resulting in the attenuation of viral replication.


Assuntos
Vírus da Influenza A Subtipo H3N8/crescimento & desenvolvimento , Vírus da Influenza A Subtipo H3N8/imunologia , Interferons/antagonistas & inibidores , Deleção de Sequência , Proteínas não Estruturais Virais/metabolismo , Fatores de Virulência/metabolismo , Animais , Doenças dos Cavalos/virologia , Cavalos , Interações Hospedeiro-Patógeno , Humanos , Vírus da Influenza A Subtipo H3N8/isolamento & purificação , Vírus da Influenza A Subtipo H3N8/fisiologia , Infecções por Orthomyxoviridae/virologia , Proteínas não Estruturais Virais/genética , Ensaio de Placa Viral , Virulência , Fatores de Virulência/genética , Replicação Viral
17.
Influenza Other Respir Viruses ; 10(5): 438-42, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27102577

RESUMO

Influenza viruses are a continuous threat to humans because of their ability to cross species barriers and adapt to new hosts. Data from murine studies, along with limited human data, suggest that CD8(+) cytotoxic T lymphocytes (CTL) that recognize conserved epitopes of structural influenza proteins are the main mediators of influenza virus clearance. Additionally, the fact that many CTLs recognize epitopes shared between different influenza strains offers the potential for broad cross-strain immunity. However, the mechanisms of cellular immunity against influenza viruses are poorly defined in humans, where the CTL response has been hard to measure and interpret. We developed a novel CTL assay that utilizes fully differentiated nasal human epithelial cells taken from volunteers as permissive targets for autologous peripheral blood-derived influenza virus-specific cytotoxic T lymphocytes. This in vitro system of human lymphocyte-epithelial cell co-cultures can be considered as the closest approximation to events in vivo and can be employed for studying the interactions between the pathogen and human host. Modeling of the natural interaction process between the primary cell type that supports the productive replication of influenza and immune cells may allow us to put in perspective CTLs as a correlate of immunity to influenza in humans.


Assuntos
Citotoxicidade Imunológica , Células Epiteliais/virologia , Interações Hospedeiro-Patógeno , Vírus da Influenza A Subtipo H1N1/imunologia , Vírus da Influenza A Subtipo H3N8/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Técnicas de Cocultura , Testes Imunológicos de Citotoxicidade , Cães , Epitopos , Humanos , Vírus da Influenza A Subtipo H1N1/fisiologia , Vírus da Influenza A Subtipo H3N8/fisiologia , Células Madin Darby de Rim Canino , Mucosa Nasal/citologia , Replicação Viral
18.
Sci Rep ; 6: 21428, 2016 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-26888262

RESUMO

An influenza H3N8 virus, carrying mammalian adaptation mutations, was isolated from New England harbor seals in 2011. We sought to assess the risk of its human transmissibility using two complementary approaches. First, we tested the binding of recombinant hemagglutinin (HA) proteins of seal H3N8 and human-adapted H3N2 viruses to respiratory tissues of humans and ferrets. For human tissues, we observed strong tendency of the seal H3 to bind to lung alveoli, which was in direct contrast to the human-adapted H3 that bound mainly to the trachea. This staining pattern was also consistent in ferrets, the primary animal model for human influenza pathogenesis. Second, we compared the binding of the recombinant HAs to a library of 610 glycans. In contrast to the human H3, which bound almost exclusively to α-2,6 sialylated glycans, the seal H3 bound preferentially to α-2,3 sialylated glycans. Additionally, the seal H3N8 virus replicated in human lung carcinoma cells. Our data suggest that the seal H3N8 virus has retained its avian-like receptor binding specificity, but could potentially establish infection in human lungs.


Assuntos
Glicoproteínas de Hemaglutininação de Vírus da Influenza/metabolismo , Vírus da Influenza A Subtipo H3N8/fisiologia , Tropismo Viral/fisiologia , Replicação Viral/fisiologia , Animais , Linhagem Celular Tumoral , Embrião de Galinha , Cães , Humanos , Células Madin Darby de Rim Canino , Especificidade da Espécie
19.
J Virol ; 89(13): 6860-73, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25903329

RESUMO

UNLABELLED: The A/H3N8 canine influenza virus (CIV) emerged from A/H3N8 equine influenza virus (EIV) around the year 2000 through the transfer of a single virus from horses to dogs. We defined and compared the biological properties of EIV and CIV by examining their genetic variation, infection, and growth in different cell cultures, receptor specificity, hemagglutinin (HA) cleavage, and infection and growth in horse and dog tracheal explant cultures. Comparison of sequences of viruses from horses and dogs revealed mutations that may be linked to host adaptation and tropism. We prepared infectious clones of representative EIV and CIV strains that were similar to the consensus sequences of viruses from each host. The rescued viruses, including HA and neuraminidase (NA) double reassortants, exhibited similar degrees of long-term growth in MDCK cells. Different host cells showed various levels of susceptibility to infection, but no differences in infectivity were seen when comparing viruses. All viruses preferred α2-3- over α2-6-linked sialic acids for infections, and glycan microarray analysis showed that EIV and CIV HA-Fc fusion proteins bound only to α2-3-linked sialic acids. Cleavage assays showed that EIV and CIV HA proteins required trypsin for efficient cleavage, and no differences in cleavage efficiency were seen. Inoculation of the viruses into tracheal explants revealed similar levels of infection and replication by each virus in dog trachea, although EIV was more infectious in horse trachea than CIV. IMPORTANCE: Influenza A viruses can cross species barriers and cause severe disease in their new hosts. Infections with highly pathogenic avian H5N1 virus and, more recently, avian H7N9 virus have resulted in high rates of lethality in humans. Unfortunately, our current understanding of how influenza viruses jump species barriers is limited. Our aim was to provide an overview and biological characterization of H3N8 equine and canine influenza viruses using various experimental approaches, since the canine virus emerged from horses approximately 15 years ago. We showed that although there were numerous genetic differences between the equine and canine viruses, this variation did not result in dramatic biological differences between the viruses from the two hosts, and the viruses appeared phenotypically equivalent in most assays we conducted. These findings suggest that the cross-species transmission and adaptation of influenza viruses may be mediated by subtle changes in virus biology.


Assuntos
Variação Genética , Vírus da Influenza A Subtipo H3N8/genética , Vírus da Influenza A Subtipo H3N8/fisiologia , Traqueia/virologia , Adaptação Biológica , Animais , Linhagem Celular , Cães , Glicoproteínas de Hemaglutininação de Vírus da Influenza/metabolismo , Cavalos , Vírus da Influenza A Subtipo H3N8/crescimento & desenvolvimento , Vírus da Influenza A Subtipo H3N8/isolamento & purificação , Mutação , Filogenia , Ligação Proteica , Receptores Virais/metabolismo , Ácidos Siálicos/metabolismo , Tropismo Viral , Ligação Viral
20.
J Gen Virol ; 96(Pt 5): 969-974, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25593159

RESUMO

Influenza A viruses circulate in a wide range of animals. H3N8 equine influenza virus (EIV) is an avian-origin virus that has established in dogs as canine influenza virus (CIV) and has also been isolated from camels and pigs. Previous work suggests that mutations acquired during EIV evolution might have played a role in CIV emergence. Given the potential role of pigs as a source of human infections, we determined the ability of H3N8 EIVs to replicate in pig cell lines and in respiratory explants. We show that phylogenetically distinct EIVs display different infection phenotypes along the pig respiratory tract, but not in cell lines. Our results suggest that EIV displays a dynamic host range along its evolutionary history, supporting the view that evolutionary processes play important roles in host range and tropism and also underscoring the utility of using explant cultures to study influenza pathogenesis.


Assuntos
Vírus da Influenza A Subtipo H3N8/fisiologia , Sistema Respiratório/virologia , Tropismo Viral , Animais , Linhagem Celular , Especificidade de Hospedeiro , Vírus da Influenza A Subtipo H3N8/isolamento & purificação , Suínos , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA