Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Virol ; 95(20): e0090621, 2021 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-34319155

RESUMEN

Cleavage of the influenza A virus (IAV) hemagglutinin (HA) by host proteases is indispensable for virus replication. Most IAVs possess a monobasic HA cleavage site cleaved by trypsin-like proteases. Previously, the transmembrane protease TMPRSS2 was shown to be essential for proteolytic activation of IAV HA subtypes H1, H2, H7, and H10 in mice. In contrast, additional proteases are involved in activation of certain H3 IAVs, indicating that HAs with monobasic cleavage sites can differ in their sensitivity to host proteases. Here, we investigated the role of TMPRSS2 in proteolytic activation of avian HA subtypes H1 to H11 and H14 to H16 in human and mouse airway cell cultures. Using reassortant viruses carrying representative HAs, we analyzed HA cleavage and multicycle replication in (i) lung cells of TMPRSS2-deficient mice and (ii) Calu-3 cells and primary human bronchial cells subjected to morpholino oligomer-mediated knockdown of TMPRSS2 activity. TMPRSS2 was found to be crucial for activation of H1 to H11, H14, and H15 in airway cells of human and mouse. Only H9 with an R-S-S-R cleavage site and H16 were proteolytically activated in the absence of TMPRSS2 activity, albeit with reduced efficiency. Moreover, a TMPRSS2-orthologous protease from duck supported activation of H1 to H11, H15, and H16 in MDCK cells. Together, our data demonstrate that in human and murine respiratory cells, TMPRSS2 is the major activating protease of almost all IAV HA subtypes with monobasic cleavage sites. Furthermore, our results suggest that TMPRSS2 supports activation of IAV with a monobasic cleavage site in ducks. IMPORTANCE Human infections with avian influenza A viruses upon exposure to infected birds are frequently reported and have received attention as a potential pandemic threat. Cleavage of the envelope glycoprotein hemagglutinin (HA) by host proteases is a prerequisite for membrane fusion and essential for virus infectivity. In this study, we identify the transmembrane protease TMPRSS2 as the major activating protease of avian influenza virus HAs of subtypes H1 to H11, H14 and H15 in human and murine airway cells. Our data demonstrate that inhibition of TMPRSS2 activity may provide a useful approach for the treatment of human infections with avian influenza viruses that should be considered for pandemic preparedness as well. Additionally, we show that a TMPRSS2-orthologous protease from duck can activate avian influenza virus HAs with a monobasic cleavage site and, thus, represents a potential virus-activating protease in waterfowl, the primary reservoir for influenza A viruses.


Asunto(s)
Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Virus de la Influenza A/metabolismo , Serina Endopeptidasas/metabolismo , Animales , Bronquios/citología , Línea Celular , Perros , Femenino , Células HEK293 , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Hemaglutininas Virales/genética , Hemaglutininas Virales/metabolismo , Interacciones Huésped-Patógeno , Humanos , Subtipo H1N1 del Virus de la Influenza A/fisiología , Subtipo H3N2 del Virus de la Influenza A/fisiología , Virus de la Influenza A/inmunología , Virus de la Influenza A/patogenicidad , Pulmón/virología , Células de Riñón Canino Madin Darby , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Péptido Hidrolasas/metabolismo , Proteolisis , Mucosa Respiratoria/metabolismo , Serina Endopeptidasas/fisiología , Replicación Viral
2.
PLoS Pathog ; 17(4): e1009490, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33891662

RESUMEN

Repeated outbreaks due to H3N1 low pathogenicity avian influenza viruses (LPAIV) in Belgium were associated with unusually high mortality in chicken in 2019. Those events caused considerable economic losses and prompted restriction measures normally implemented for eradicating high pathogenicity avian influenza viruses (HPAIV). Initial pathology investigations and infection studies suggested this virus to be able to replicate systemically, being very atypical for H3 LPAIV. Here, we investigate the pathogenesis of this H3N1 virus and propose a mechanism explaining its unusual systemic replication capability. By intravenous and intracerebral inoculation in chicken, we demonstrate systemic spread of this virus, extending to the central nervous system. Endoproteolytic viral hemagglutinin (HA) protein activation by either tissue-restricted serine peptidases or ubiquitous subtilisin-like proteases is the functional hallmark distinguishing (H5 or H7) LPAIV from HPAIV. However, luciferase reporter assays show that HA cleavage in case of the H3N1 strain in contrast to the HPAIV is not processed by intracellular proteases. Yet the H3N1 virus replicates efficiently in cell culture without trypsin, unlike LPAIVs. Moreover, this trypsin-independent virus replication is inhibited by 6-aminohexanoic acid, a plasmin inhibitor. Correspondingly, in silico analysis indicates that plasminogen is recruitable by the viral neuraminidase for proteolytic activation due to the loss of a strongly conserved N-glycosylation site at position 130. This mutation was shown responsible for plasminogen recruitment and neurovirulence of the mouse brain-passaged laboratory strain A/WSN/33 (H1N1). In conclusion, our findings provide good evidence in natural chicken strains for N1 neuraminidase-operated recruitment of plasminogen, enabling systemic replication leading to an unusual high pathogenicity phenotype. Such a gain of function in naturally occurring AIVs representing an established human influenza HA-subtype raises concerns over potential zoonotic threats.


Asunto(s)
Brotes de Enfermedades/veterinaria , Virus de la Influenza A/patogenicidad , Gripe Aviar/virología , Neuraminidasa/metabolismo , Plasminógeno/metabolismo , Enfermedades de las Aves de Corral/virología , Animales , Pollos , Glicosilación , Virus de la Influenza A/enzimología , Virus de la Influenza A/fisiología , Neuraminidasa/genética , Replicación Viral
3.
Emerg Microbes Infect ; 8(1): 1324-1336, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31503518

RESUMEN

Avian influenza A viruses (AIV) of the H7 subtype continue to evolve posing a pandemic threat. However, molecular markers of H7N7 AIV pathogenicity and transmission in mammals remain poorly understood. In this study, we performed a systematic in vitro and in vivo analysis by comparing an H7N7 highly pathogenic AIV and its ferret adapted variant. Passaging an H7N7 AIV in ferrets led to six mutations in genes encoding the viral polymerase complex and the viral surface proteins. Here, we show that mutations in the H7 hemagglutinin gene cause increased pathogenicity in mice. Contact transmission between guinea pigs required additional mutations in the gene encoding the polymerase subunit PB1. Thus, particular vigilance is required with respect to HA and PB1 mutations as predictive molecular markers to assess the pandemic risk posed by emerging H7 avian influenza viruses.


Asunto(s)
Transmisión de Enfermedad Infecciosa , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Subtipo H7N7 del Virus de la Influenza A/patogenicidad , Proteínas Mutantes/genética , Infecciones por Orthomyxoviridae/transmisión , Infecciones por Orthomyxoviridae/virología , Proteínas Virales/genética , Animales , Modelos Animales de Enfermedad , Hurones , Cobayas , Subtipo H7N7 del Virus de la Influenza A/genética , Infecciones por Orthomyxoviridae/patología , Pase Seriado , Factores de Virulencia/genética
4.
Vet Microbiol ; 231: 160-168, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30955804

RESUMEN

Influenza A viruses (IAV) have caused seasonal epidemics and severe pandemics in humans. Novel pandemic strains as in 2009 may emerge from pigs, serving as perpetual virus reservoir. However, reliably effective vaccination has remained a key issue for humans and swine. Here, we generated a novel double-attenuated influenza live vaccine by reverse genetics and subjected immunized mice and pigs to infection with the homologous wild-type, another homosubtypic H1N1, or a heterosubtypic H3N2 virus to address realistic challenge constellations. This attenuated mutant contains an artificial, strictly elastase-dependent hemagglutinin cleavage site and a C-terminally truncated NS1 protein from the IAV A/Bayern/74/2009 (H1N1pdm09). Prior to challenge, we immunized mice once and pigs twice intranasally. In vitro, the double-attenuated mutant replicated strictly elastase-dependently. Immunized mice and pigs developed neither clinical symptoms nor detectable virus replication after homologous challenge. In pigs, we observed considerably reduced clinical signs and no nasal virus shedding after homosubtypic and reduced viral loads in respiratory tracts after heterosubtypic infection. Protection against homosubtypic challenge suggests that an optimized backbone strain may require less frequent updates with recent HA and NA genes and still induce robust protection in relevant IAV hosts against drifted viruses.


Asunto(s)
Protección Cruzada , Vacunas contra la Influenza/inmunología , Infecciones por Orthomyxoviridae/veterinaria , Enfermedades de los Porcinos/prevención & control , Animales , Anticuerpos Antivirales , Femenino , Subtipo H1N1 del Virus de la Influenza A , Subtipo H3N2 del Virus de la Influenza A , Vacunas contra la Influenza/administración & dosificación , Ratones , Ratones Endogámicos BALB C , Mutación , Infecciones por Orthomyxoviridae/prevención & control , Genética Inversa , Serogrupo , Porcinos , Enfermedades de los Porcinos/inmunología , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/inmunología , Esparcimiento de Virus
5.
Genome Med ; 10(1): 58, 2018 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-30081931

RESUMEN

BACKGROUND: The 2014-2016 Ebola virus (EBOV) outbreak in West Africa highlighted the need for improved therapeutic options against this virus. Approaches targeting host factors/pathways essential for the virus are advantageous because they can potentially target a wide range of viruses, including newly emerging ones and because the development of resistance is less likely than when targeting the virus directly. However, systematic approaches for screening host factors important for EBOV have been hampered by the necessity to work with this virus at biosafety level 4 (BSL4). METHODS: In order to identify host factors involved in the EBOV life cycle, we performed a genome-wide siRNA screen comprising 64,755 individual siRNAs against 21,566 human genes to assess their activity in EBOV genome replication and transcription. As a screening platform, we used reverse genetics-based life cycle modelling systems that recapitulate these processes without the need for a BSL4 laboratory. RESULTS: Among others, we identified the de novo pyrimidine synthesis pathway as an essential host pathway for EBOV genome replication and transcription, and confirmed this using infectious EBOV under BSL4 conditions. An FDA-approved drug targeting this pathway showed antiviral activity against infectious EBOV, as well as other non-segmented negative-sense RNA viruses. CONCLUSIONS: This study provides a minable data set for every human gene regarding its role in EBOV genome replication and transcription, shows that an FDA-approved drug targeting one of the identified pathways is highly efficacious in vitro, and demonstrates the power of life cycle modelling systems for conducting genome-wide host factor screens for BSL4 viruses.


Asunto(s)
Antivirales/farmacología , Ebolavirus/fisiología , Genoma Humano , Replicación Viral , Animales , Línea Celular Tumoral , Chlorocebus aethiops , Clonación Molecular , Ebolavirus/efectos de los fármacos , Ebolavirus/patogenicidad , Técnicas de Silenciamiento del Gen , Células HEK293 , Interacciones Huésped-Patógeno/genética , Humanos , Células Vero
6.
J Virol ; 92(17)2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29899102

RESUMEN

Zoonotic highly pathogenic avian influenza viruses (HPAIV) have raised serious public health concerns of a novel pandemic. These strains emerge from low-pathogenic precursors by the acquisition of a polybasic hemagglutinin (HA) cleavage site, the prime virulence determinant. However, required coadaptations of the HA early in HPAIV evolution remained uncertain. To address this question, we generated several HA1/HA2 chimeras and point mutants of an H5N1 clade 2.2.2 HPAIV and an H5N1 low-pathogenic strain. Initial surveys of 3,385 HPAIV H5 HA sequences revealed frequencies of 0.5% for the single amino acids 123R and 124I but a frequency of 97.5% for the dual combination. This highly conserved dual motif is still retained in contemporary H5 HPAIV, including the novel H5NX reassortants carrying neuraminidases of different subtypes, like the H5N8 and the zoonotic H5N6 strains. Remarkably, the earliest Asian H5N1 HPAIV, the Goose/Guangdong strains from 1996/1997, carried 123R only, whereas 124I appeared later in 1997. Experimental reversion in the HPAIV HA to the two residues 123S and124T, characteristic of low-pathogenic strains, prevented virus rescue, while the single substitutions attenuated the virus in both chicken and mice considerably, accompanied by a decreased HA fusion pH. This increased pH sensitivity of H5 HPAIV enables HA-mediated membrane fusion at a higher endosomal pH. Therefore, this HA adaptation may permit infection of cells with less-acidic endosomes, e.g., within the respiratory tract, resulting in an extended organ tropism. Taken together, HA coadaptation to increased acid sensitivity promoted the early evolution of H5 Goose/Guangdong-like HPAIV strains and is still required for their zoonotic potential.IMPORTANCE Zoonotic highly pathogenic avian influenza viruses (HPAIV) have raised serious public health concerns of a novel pandemic. Their prime virulence determinant is the polybasic hemagglutinin (HA) cleavage site. However, required coadaptations in the HA (and other genes) remained uncertain. Here, we identified the dual motif 123R/124I in the HA head that increases the activation pH of HA-mediated membrane fusion, essential for virus genome release into the cytoplasm. This motif is extremely predominant in H5 HPAIV and emerged already in the earliest 1997 H5N1 HPAIV. Reversion to 123S or 124T, characteristic of low-pathogenic strains, attenuated the virus in chicken and mice, accompanied by a decreased HA activation pH. This increased pH sensitivity of H5 HPAIV extends the viral tropism to cells with less-acidic endosomes, e.g., within the respiratory tract. Therefore, early HA adaptation to increased acid sensitivity promoted the emergence of H5 Goose/Guangdong-like HPAIV strains and is required for their zoonotic potential.


Asunto(s)
Secuencia Conservada , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Subtipo H5N1 del Virus de la Influenza A/genética , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Internalización del Virus , Animales , Análisis por Conglomerados , Evolución Molecular , Gansos , Concentración de Iones de Hidrógeno , Filogenia , Análisis de Secuencia de ADN , Virulencia , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
7.
Vet Microbiol ; 211: 129-134, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29102108

RESUMEN

We analyzed the adaptation of influenza viruses to growth in differentiated airway epithelial cells of a new host by passaging an avian H9N2 virus three times in porcine precision-cut lung slices (PCLS). Sequence analysis revealed four mutations: one each in the PB2 and NS1 proteins, and two in the HA protein. In this study, we characterized the PB2 mutation G685R by generating recombinant H9N2 viruses containing the PB2 single mutation alone or in combination with one of the HA mutations (A190V or T212I). When analyzed in porcine cells - a tracheal cell line (NPTr) or PCLS - the PB2-685 mutant did not provide a growth advantage and had no effect on the ciliary activity which is a virulence marker of swine influenza viruses. Pathogenicity for mice was also not increased by the single PB2 mutation. However, both double mutants (HA-190+PB2-685 and HA-212+PB2-685) showed significantly increased virulence in mice. Therefore, the mutations in the HA and PB2 proteins may confer early adaptation of an avian H9N2 virus to a mammalian host. In conclusion, we expect that a broader ensemble of mutations will be required to render an H9N2 virus virulent for pigs.


Asunto(s)
Subtipo H9N2 del Virus de la Influenza A/patogenicidad , Gripe Aviar/virología , Infecciones por Orthomyxoviridae/veterinaria , Animales , Aves , Línea Celular , Células Epiteliales/virología , Subtipo H9N2 del Virus de la Influenza A/genética , Pulmón/virología , Ratones , Mutación , Infecciones por Orthomyxoviridae/virología , Recombinación Genética , Sistema Respiratorio/virología , Porcinos , Virulencia
8.
J Gen Virol ; 97(12): 3193-3204, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27902339

RESUMEN

Highly pathogenic H5N1 avian influenza virus (A/H5N1) devastated the poultry industry and continues to pose a pandemic threat. Studying the progressive genetic changes in A/H5N1 after long-term circulation in poultry may help us to better understand A/H5N1 biology in birds. A/H5N1 clade 2.2.1.1 antigenic drift viruses have been isolated from vaccinated commercial poultry in Egypt. They exhibit a peculiar stepwise accumulation of glycosylation sites (GS) in the haemagglutinin (HA) with viruses carrying, beyond the conserved 5 GS, additional GS at amino acid residues 72, 154, 236 and 273 resulting in 6, 7, 8 or 9 GS in the HA. Available information about the impact of glycosylation on virus fitness and pathobiology is mostly derived from mammalian models. Here, we generated recombinant viruses imitating the progressive acquisition of GS in HA and investigated their biological relevance in vitro and in vivo. Our in vitro results indicated that the accumulation of GS correlated with increased glycosylation, increased virus replication, neuraminidase activity, cell-to-cell spread and thermostability, however, strikingly, without significant impact on virus escape from neutralizing antibodies. In vivo, glycosylation modulated virus virulence, tissue tropism, replication and chicken-to-chicken transmission. Predominance in the field was towards viruses with hyperglycosylated HA. Together, progressive glycosylation of the HA may foster persistence of A/H5N1 by increasing replication, stability and bird-to-bird transmission without significant impact on antigenic drift.


Asunto(s)
Variación Antigénica , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Subtipo H5N1 del Virus de la Influenza A/fisiología , Gripe Aviar/transmisión , Enfermedades de las Aves de Corral/virología , Replicación Viral , Secuencias de Aminoácidos , Animales , Pollos , Egipto , Glicosilación , Glicoproteínas Hemaglutininas del Virus de la Influenza/química , Subtipo H5N1 del Virus de la Influenza A/clasificación , Subtipo H5N1 del Virus de la Influenza A/genética , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Gripe Aviar/virología , Filogenia , Virulencia
9.
Virulence ; 7(5): 546-57, 2016 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-26981790

RESUMEN

Highly pathogenic (HP) avian influenza viruses (AIV) evolve from low pathogenic (LP) precursors after circulation in poultry by reassortment and/or single mutations in different gene segments including that encoding NS1. The carboxyl terminal end (CTE) of NS1 exhibits deletions between amino acid 202 and 230 with still unknown impact on virulence of AIV in chickens. In this study, NS1 protein sequences of all AIV subtypes in birds from 1902 to 2015 were analyzed to study the prevalence and distribution of CTE truncation (ΔCTE). Thirteen different ΔCTE forms were observed in NS1 proteins from 11 HA and 8 NA subtypes with high prevalences in H9, H7, H6 and H10 and N9, N2, N6 and N1 subtypes particularly in chickens and minor poultry species. With 88% NS217 lacking amino acids 218-230 was the most common ΔCTE form followed by NS224 (3.6%). NS217 was found in 10 and 8 different HA and NA subtypes, respectively, whereas NS224 was detected exclusively in the Italian HPAIV H7N1 suggesting relevance for virulence. To test this assumption, 3 recombinant HPAIV H7N1 were constructed carrying wild-type HP NS1 (Hp-NS224), NS1 with extended CTE (Hp-NS230) or NS1 from LPAIV H7N1 (Hp-NSLp), and tested in-vitro and in-vivo. Extension of CTE in Hp NS1 significantly decreased virus replication in chicken embryo kidney cells. Truncation in the NS1 decreased the tropism of Hp-NS224 to the endothelium, central nervous system and respiratory tract epithelium without significant difference in virulence in chickens. This study described the variable forms of ΔCTE in NS1 and indicated that CTE is not an essential virulence determinant particularly for the Italian HPAIV H7N1 but may be a host-adaptation marker required for efficient virus replication.


Asunto(s)
Subtipo H7N1 del Virus de la Influenza A/genética , Subtipo H7N1 del Virus de la Influenza A/patogenicidad , Virus de la Influenza A/genética , Virus de la Influenza A/patogenicidad , Gripe Aviar/virología , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/genética , Adaptación Biológica , Animales , Sistema Nervioso Central/virología , Pollos , Subtipo H7N1 del Virus de la Influenza A/fisiología , Virus de la Influenza A/fisiología , Prevalencia , Virus Reordenados/genética , Mucosa Respiratoria/virología , Análisis de Secuencia de Proteína , Tropismo Viral , Factores de Virulencia/genética , Replicación Viral
10.
J Virol ; 90(1): 400-11, 2016 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-26491158

RESUMEN

UNLABELLED: In 1999, after circulation for a few months in poultry in Italy, low-pathogenic (LP) avian influenza (AI) H7N1 virus mutated into a highly pathogenic (HP) form by acquisition of a unique multibasic cleavage site (mCS), PEIPKGSRVRR*GLF (asterisk indicates the cleavage site), in the hemagglutinin (HA) and additional alterations with hitherto unknown biological function. To elucidate these virulence-determining alterations, recombinant H7N1 viruses carrying specific mutations in the HA of LPAI A/chicken/Italy/473/1999 virus (Lp) and HPAI A/chicken/Italy/445/1999 virus (Hp) were generated. Hp with a monobasic CS or carrying the HA of Lp induced only mild or no disease in chickens, thus resembling Lp. Conversely, Lp with the HA of Hp was as virulent and transmissible as Hp. While Lp with a multibasic cleavage site (Lp_CS445) was less virulent than Hp, full virulence was exhibited when HA2 was replaced by that of Hp. In HA2, three amino acid differences consistently detected between LP and HP H7N1 viruses were successively introduced into Lp_CS445. Q450L in the HA2 stem domain increased virulence and transmission but was detrimental to replication in cell culture, probably due to low-pH activation of HA. A436T and/or K536R restored viral replication in vitro and in vivo. Viruses possessing A436T and K536R were observed early in the HPAI outbreak but were later superseded by viruses carrying all three mutations. Together, besides the mCS, stepwise mutations in HA2 increased the fitness of the Italian H7N1 virus in vivo. The shift toward higher virulence in the field was most likely gradual with rapid optimization. IMPORTANCE: In 1999, after 9 months of circulation of low-pathogenic (LP) avian influenza virus (AIV), a devastating highly pathogenic (HP) H7N1 AIV emerged in poultry, marking the largest epidemic of AIV reported in a Western country. The HPAIV possessed a unique multibasic cleavage site (mCS) complying with the minimum motif for HPAIV. The main finding in this report is the identification of three mutations in the HA2 domain that are required for replication and stability, as well as for virulence, transmission, and tropism of H7N1 in chickens. In addition to the mCS, Q450L was required for full virulence and transmissibility of the virus. Nonetheless, it was detrimental to virus replication and required A436T and/or K536R to restore replication, systemic spread, and stability. These results are important for better understanding of the evolution of highly pathogenic avian influenza viruses from low-pathogenic precursors.


Asunto(s)
Hemaglutininas Virales/genética , Hemaglutininas Virales/metabolismo , Subtipo H7N1 del Virus de la Influenza A/crecimiento & desarrollo , Subtipo H7N1 del Virus de la Influenza A/genética , Gripe Aviar/patología , Gripe Aviar/virología , Mutación Missense , Animales , Pollos , Italia , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Recombinación Genética , Genética Inversa , Virulencia
11.
Vaccine ; 33(49): 6997-7001, 2015 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-26469725

RESUMEN

We evaluated the virulence of replication-competent NA-negative (no functional neuraminidase) avian influenza viruses in chicken of different ages. Interestingly, the virulence of a previously described NA-negative H5-virus (Kalthoff et al., J Virol 2013;87:13556) is very much age-dependent. Day-old-chicks succumb to a systemic infection, while chicken one week of age do not show any clinical signs at all. Therefore, the day-old-chick organism is most likely not able to restrict replication of the virus as older chicken do. In addition to virulence dependency on host age, the exchange of the H5 HA by an H7 HA for an H7NA-deleted HA reassortant remarkably led to a highly pathogenic phenotype even in adult chicken. This provides evidence, that the virulence of the H7-subtype is less dependent on the NA-protein than that of the H5-virus. These striking observations suggest that the HA/NA interplay might be governed by other mechanisms in HPAIV with the different serotypes H5 or H7.


Asunto(s)
Eliminación de Gen , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Subtipo H7N1 del Virus de la Influenza A/patogenicidad , Gripe Aviar/virología , Neuraminidasa/genética , Animales , Pollos , Gripe Aviar/mortalidad , Gripe Aviar/patología , Carga Viral , Virulencia
12.
J Virol Methods ; 225: 87-9, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26404948

RESUMEN

Reverse genetics of influenza A viruses facilitates both basic research and vaccine development. However, efficient cloning of virus gene segments was cumbersome in established systems due to the necessary cleavage of amplicons with outside cutter restriction enzymes followed by ligation. Occasionally, virus genes may contain cleavage sites for those enzymes. To circumvent that problem, we previously established target-primed plasmid amplification using the negative selection marker ccdB cloned into the plasmid pHW2000, flanked by the highly conserved gene segment termini. Here, we further introduced the LacZα fragment downstream of the ccdB region for additional ad-hoc selection of transformed bacteria by blue/white pre-screening. For comparison, we cloned three gene segments (PA, HA, and NS) from the influenza strain A/Swine/Belgium/1/1979 (H1N1) (SwBelg79) into plasmid vectors pHWSccdB and pHWSccdB-LacZα and observed same cloning efficiency. Furthermore, the plasmid pHWSccdB-LacZα allows easy elimination of bacterial colonies containing empty plasmid clones. Using this improved plasmid, we obtained the complete genomic set of eight functional plasmids for SwBelg79.


Asunto(s)
Clonación Molecular/métodos , Virus de la Influenza A/genética , Genética Inversa/métodos , beta-Galactosidasa/análisis , Pruebas Genéticas , Vectores Genéticos , Plásmidos , Selección Genética , beta-Galactosidasa/genética
13.
Sci Rep ; 5: 13493, 2015 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-26306544

RESUMEN

Highly pathogenic avian influenza viruses (HPAIV) cause devastating losses in gallinaceous poultry world-wide and raised concerns of a novel pandemic. HPAIV develop from low-pathogenic precursors by acquisition of a polybasic HA cleavage site (HACS), the prime virulence determinant. Beside that HACS, other adaptive changes accumulate in those precursors prior to transformation into an HPAIV. Here, we aimed to unravel such virulence determinants in addition to the HA gene. Stepwise reduction of HPAIV genes revealed that the HPAIV HA and NA form a minimum set of virulence determinants, sufficient for a lethal phenotype in chicken. Abolishing the NA stalk deletion considerably reduced lethality and prevented transmission. Conversely, the analogous stalk deletion reconstructed in the NA of an LPAIV reassortant carrying only the HPAIV HA resulted in 100% lethality both after primary and contact infection. Remarkably, the unmodified LPAIV NA with its long stalk, when exclusively introduced into the H5N1 HPAIV, still enabled high virulence and efficient transmission. Therefore, irrespective of an NA stalk deletion, minor virulence determinants in addition to the essential polybasic HACS contribute to high virulence, whereas the NA stalk deletion alone may serve as major virulence determinant.


Asunto(s)
Subtipo H5N1 del Virus de la Influenza A/enzimología , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Gripe Aviar/virología , Neuraminidasa/genética , Neuraminidasa/metabolismo , Factores de Virulencia/metabolismo , Animales , Pollos , Subtipo H5N1 del Virus de la Influenza A/genética , Neuraminidasa/química , Relación Estructura-Actividad , Factores de Virulencia/química , Factores de Virulencia/genética
14.
PLoS One ; 9(2): e89529, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24586849

RESUMEN

Binding of influenza viruses to target cells is mediated by the viral surface protein hemagglutinin. To determine the presence of binding sites for influenza A viruses on cells and tissues, soluble hemagglutinins of the H7 and H9 subtype were generated by connecting the hemagglutinin ectodomain to the Fc portion of human immunoglobulin G (H7Fc and H9Fc). Both chimeric proteins bound to different cells and tissues in a sialic acid-dependent manner. Pronounced differences were observed between H7Fc and H9Fc, in the binding both to different mammalian and avian cultured cells and to cryosections of the respiratory epithelium of different virus host species (turkey, chicken and pig). Binding of the soluble hemagglutinins was similar to the binding of virus particles, but showed differences in the binding pattern when compared to two sialic acid-specific plant lectins. These findings were substantiated by a comparative glycan array analysis revealing a very narrow recognition of sialoglycoconjugates by the plant lectins that does not reflect the glycan structures preferentially recognized by H7Fc and H9Fc. Thus, soluble hemagglutinins may serve as sialic acid-specific lectins and are a more reliable indicator of the presence of binding sites for influenza virus HA than the commonly used plant lectins.


Asunto(s)
Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Virus de la Influenza A/fisiología , Ácido N-Acetilneuramínico/metabolismo , Animales , Sitios de Unión , Conformación de Carbohidratos , Secuencia de Carbohidratos , Línea Celular Tumoral , Pollos , Epitelio/virología , Glucolípidos/metabolismo , Glicoproteínas Hemaglutininas del Virus de la Influenza/química , Humanos , Glicoproteínas de Membrana/metabolismo , Datos de Secuencia Molecular , Ácido N-Acetilneuramínico/química , Neuraminidasa/química , Unión Proteica , Mucosa Respiratoria/virología , Sus scrofa , Pavos , Acoplamiento Viral
15.
J Virol ; 87(24): 13556-68, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24109212

RESUMEN

Influenza viruses are highly genetically variable and escape from immunogenic pressure by antigenic changes in their surface proteins, referred to as "antigenic drift" and "antigenic shift." To assess the potential genetic plasticity under strong selection pressure, highly pathogenic avian influenza virus (HPAIV) of subtype H5N1 was passaged 50 times in embryonated chicken eggs in the presence of a neutralizing, polyclonal chicken serum. The resulting mutant acquired major alterations in the neuraminidase (NA)-encoding segment. Extensive deletions and rearrangements were detected, in contrast to only 12 amino acid substitutions within all other segments. Interestingly, this new neuraminidase segment resulted from complex sequence shuffling and insertion of a short fragment originating from the PA segment. Characterization of that novel variant revealed a loss of the neuraminidase protein and enzymatic activity, but its replication efficiency remained comparable to that of the wild type. Using reverse genetics, a recombinant virus consisting of the wild-type backbone and the shortened NA segment could be generated; however, generation of this recombinant virus required the polybasic hemagglutinin cleavage site. Two independent repetitions starting with egg passage 30 in the presence of alternative chicken-derived immune sera selected mutants with similar but different large deletions within the NA segment without any neuraminidase activity, indicating a general mechanism. In chicken, these virus variants were avirulent, even though the HPAIV polybasic hemagglutinin cleavage site was still present. Overall, the variants reported here are the first HPAIV H5N1 strains without a functional neuraminidase shown to grow efficiently without any helper factor. These novel HPAIV variants may facilitate future studies shedding light on the role of neuraminidase in virus replication and pathogenicity.


Asunto(s)
Subtipo H5N1 del Virus de la Influenza A/enzimología , Subtipo H5N1 del Virus de la Influenza A/fisiología , Gripe Aviar/virología , Neuraminidasa/genética , Enfermedades de las Aves de Corral/virología , Eliminación de Secuencia , Proteínas Virales/genética , Replicación Viral , Sustitución de Aminoácidos , Animales , Pollos , Subtipo H5N1 del Virus de la Influenza A/genética , Mutagénesis Insercional , Neuraminidasa/química , Neuraminidasa/deficiencia , Pase Seriado , Proteínas Virales/química , Proteínas Virales/metabolismo
16.
Virus Genes ; 45(1): 14-23, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22669540

RESUMEN

An evolutionary analysis was conducted of 354 hemagglutinin (HA) and 208 neuraminidase (NA) genes, including newly generated sequences of 5 HA and 30 NA, of Egyptian H5N1 clade 2.2.1 viruses isolated from poultry and humans. Five distinct phylogenetically distinguishable clusters arose from a monophyletic origin since 2006. Only two clusters remained in circulation after 2009: (i) A cluster of viruses arose in 2007 in industrial-vaccinated chickens and carried multiple mutations in or adjacent to the immunogenic epitopes of the HA. Viruses within this cluster evolved with significantly elevated mutation rates indicating persisting selective pressures, e.g. to escape host immunity and (ii) The second group arose in 2008 and harboured strains from recent human infections featuring a conspicuous deletion in the HA receptor-binding domain and substitutions close to the highly conserved active site of the NA. In both sublineages, a number of positively selected amino acids, different glycosylation patterns and variations in the polybasic proteolytic cleavage site were observed. Continuous monitoring of the evolving H5N1 virus in Egypt is essential to develop new control campaigns in poultry and human population.


Asunto(s)
Evolución Molecular , Subtipo H5N1 del Virus de la Influenza A/genética , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Gripe Aviar/epidemiología , Gripe Humana/epidemiología , Enfermedades de las Aves de Corral/epidemiología , Animales , Pollos/virología , Patos/virología , Egipto/epidemiología , Enfermedades Endémicas , Gansos/virología , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Humanos , Gripe Aviar/virología , Gripe Humana/virología , Mutación , Neuraminidasa/genética , Filogenia , Enfermedades de las Aves de Corral/virología , Análisis de Secuencia de ADN , Pavos/virología
18.
Virology ; 427(1): 1-9, 2012 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-22365327

RESUMEN

The 2009 influenza A virus (IAV) pandemic resulted from reassortment of avian, human and swine strains probably in pigs. To elucidate the role of viral genes in host adaptation regarding innate immune responses, we focussed on the effect of genes from an avian H5N1 and a porcine H1N1 IAV on infectivity and activation of porcine GM-CSF-induced dendritic cells (DC). The highest interferon type I responses were achieved by the porcine virus reassortant containing the avian polymerase gene PB2. This finding was not due to differential tropism since all viruses infected DC equally. All viruses equally induced MHC class II, but porcine H1N1 expressing the avian viral PB2 induced more prominent nuclear NF-κB translocation compared to its parent IAV. The enhanced activation of DC may be detrimental or beneficial. An over-stimulation of innate responses could result in either pronounced tissue damage or increased resistance against IAV reassortants carrying avian PB2.


Asunto(s)
Células Dendríticas/virología , Interacciones Huésped-Patógeno , Subtipo H1N1 del Virus de la Influenza A/inmunología , Subtipo H5N1 del Virus de la Influenza A/inmunología , Interferón Tipo I/metabolismo , Infecciones por Orthomyxoviridae/inmunología , ARN Polimerasa Dependiente del ARN/inmunología , Virus Reordenados/inmunología , Enfermedades de los Porcinos/virología , Proteínas Virales/inmunología , Animales , Aves , Línea Celular , Embrión de Pollo , Células Dendríticas/inmunología , Perros , Genes Virales , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Humanos , Subtipo H1N1 del Virus de la Influenza A/genética , Subtipo H5N1 del Virus de la Influenza A/genética , Complejo Mayor de Histocompatibilidad , Ratones , FN-kappa B/metabolismo , Infecciones por Orthomyxoviridae/veterinaria , Infecciones por Orthomyxoviridae/virología , Pandemias , Regiones Promotoras Genéticas/genética , Virus Reordenados/genética , Porcinos , Enfermedades de los Porcinos/inmunología , Replicación Viral/genética
19.
Proc Natl Acad Sci U S A ; 109(7): 2579-84, 2012 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-22308331

RESUMEN

High-pathogenic avian influenza viruses (HPAIVs) evolve from low-pathogenic precursors specifying the HA serotypes H5 or H7 by acquisition of a polybasic HA cleavage site. As the reason for this serotype restriction has remained unclear, we aimed to distinguish between compatibility of a polybasic cleavage site with H5/H7 HA only and unique predisposition of these two serotypes for insertion mutations. To this end, we introduced a polybasic cleavage site into the HA of several low-pathogenic avian strains with serotypes H1, H2, H3, H4, H6, H8, H10, H11, H14, or H15, and rescued HA reassortants after cotransfection with the genes from either a low-pathogenic H9N2 or high-pathogenic H5N1 strain. Oculonasal inoculation with those reassortants resulted in varying pathogenicity in chicken. Recombinants containing the engineered H2, H4, H8, or H14 in the HPAIV background were lethal and exhibited i.v. pathogenicity indices of 2.79, 2.37, 2.85, and 2.61, respectively, equivalent to naturally occurring H5 or H7 HPAIV. Moreover, the H2, H4, and H8 reassortants were transmitted to some contact chickens. The H2 reassortant gained two mutations in the M2 proton channel gate region, which is affected in some HPAIVs of various origins. Taken together, in the presence of a polybasic HA cleavage site, non-H5/H7 HA can support a highly pathogenic phenotype in the appropriate viral background, indicating requirement for further adaptation. Therefore, the restriction of natural HPAIV to serotypes H5 and H7 is likely a result of their unique predisposition for acquisition of a polybasic HA cleavage site.


Asunto(s)
Glicoproteínas Hemaglutininas del Virus de la Influenza/fisiología , Virus de la Influenza A/aislamiento & purificación , Gripe Aviar/virología , Animales , Aves , Virus de la Influenza A/patogenicidad
20.
J Infect Dis ; 204(10): 1483-90, 2011 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-21917878

RESUMEN

BACKGROUND: Both pandemic and interpandemic influenza is associated with high morbidity and mortality worldwide. Seasonal epidemics are caused by both influenza A and B virus strains that cocirculate with varying predominance and may give rise to severe illness equally. According to World Health Organization recommendations, current annual vaccines are composed of 2 type A and 1 type B virus-specific component. METHODS: As a novel attenuated live vaccine against influenza B virus, we generated a hemagglutinin cleavage site mutant of strain B/Lee/40 by replacing the common monobasic cleavage site recognized by trypsinlike proteases with an elastase-sensitive site, and we investigated the in vitro properties, attenuation, humoral responses, and efficacy in mice. RESULTS: This mutant virus replicated in cell culture equally well as the wild type but in a strictly elastase-dependent manner. In contrast to the mouse-pathogenic parental virus, the cleavage site mutant was fully attenuated in mice and not detectable in their lungs. After 1 intranasal immunization, the animals survived lethal challenge with wild-type virus without weight loss or any other signs of disease. Furthermore, no challenge virus could be reisolated from the lungs of vaccinated mice. CONCLUSIONS: These findings demonstrate that proteolytic activation mutants can serve as live vaccine against influenza B virus.


Asunto(s)
Virus de la Influenza B/genética , Vacunas contra la Influenza/inmunología , Mutación , Animales , Anticuerpos Antivirales/sangre , Femenino , Hemaglutinación por Virus/genética , Virus de la Influenza B/química , Virus de la Influenza B/inmunología , Vacunas contra la Influenza/genética , Ratones , Ratones Endogámicos BALB C , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...