scholarly journals The substitution of T271A in PB2 protein could enhance the infectivity and pathogenicity of Eurasian avian-like H1N1 swine influenza viruses in mice

2020 ◽  
Author(s):  
Zhaomin Feng ◽  
Wenfei Zhu ◽  
Lijuan Zhou ◽  
Yongkun Chen ◽  
Xiyan Li ◽  
...  

Abstract BackgroundCurrently, Eurasian avian-like H1N1 (EA H1N1) swine influenza viruses (SIVs) are widely prevalent in pigs in China, with sporadic human cases reported as well. As one of the key molecular makers detected in avian H5N1 and H 7N9 viruses and pandemic H1N1 2009 virus, contributions of T271A in PB2 protein to the EA H1N1 viruses are still unknown. In this study, we investigated the effects of residue 271 in PB2 protein on the viral properties of EA H1N1 viruses.MethodsInfectivity, replication, virulence and pathogenicity of the recombinant viruses containing A or T in position 271 in PB2 protein were studied in cells and mice.ResultsThe results showed that the substitution PB2-T271A increased the viral replication in mammalian and avian cell lines. In addition, the mutation enhanced the viral infectivity, virulence and pathogenicity in mice. The viral titers of lung tissue in the rgHuN271A virus were higher than that of the rgHuN271T at 1, 4, and 7 dpi. The MID50 of the rgHuN271A and rgHuN271T virus were 101.1 TCID50 and 101.9 TCID50, respectively. Besides, the substitution of PB2-T271A enhanced the viral polymerase activity in mammalian cells.ConclusionsThe pathogenicity and replication of EA H1N1 virus containing 271A in PB2 protein were higher than the EA H1N1 virus containing 271T in PB2 protein in vivo and in vitro. Therefore, the PB2-T271A mutation should be continually monitored in influenza viruses circulating in pigs and humans.

2015 ◽  
Vol 89 (10) ◽  
pp. 5651-5667 ◽  
Author(s):  
Daniel Dlugolenski ◽  
Les Jones ◽  
Elizabeth Howerth ◽  
David Wentworth ◽  
S. Mark Tompkins ◽  
...  

ABSTRACTSwine are susceptible to infection by both avian and human influenza viruses, and this feature is thought to contribute to novel reassortant influenza viruses. In this study, the influenza virus reassortment rate in swine and human cells was determined. Coinfection of swine cells with 2009 pandemic H1N1 virus (huH1N1) and an endemic swine H1N2 (A/swine/Illinois/02860/09) virus (swH1N2) resulted in a 23% reassortment rate that was independent of α2,3- or α2,6-sialic acid distribution on the cells. The reassortants had altered pathogenic phenotypes linked to introduction of the swine virus PA and neuraminidase (NA) into huH1N1. In mice, the huH1N1 PA and NA mediated increased MIP-2 expression early postinfection, resulting in substantial pulmonary neutrophilia with enhanced lung pathology and disease. The findings support the notion that swine are a mixing vessel for influenza virus reassortants independent of sialic acid distribution. These results show the potential for continued reassortment of the 2009 pandemic H1N1 virus with endemic swine viruses and for reassortants to have increased pathogenicity linked to the swine virus NA and PA genes which are associated with increased pulmonary neutrophil trafficking that is related to MIP-2 expression.IMPORTANCEInfluenza A viruses can change rapidly via reassortment to create a novel virus, and reassortment can result in possible pandemics. Reassortments among subtypes from avian and human viruses led to the 1957 (H2N2 subtype) and 1968 (H3N2 subtype) human influenza pandemics. Recent analyses of circulating isolates have shown that multiple genes can be recombined from human, avian, and swine influenza viruses, leading to triple reassortants. Understanding the factors that can affect influenza A virus reassortment is needed for the establishment of disease intervention strategies that may reduce or preclude pandemics. The findings from this study show that swine cells provide a mixing vessel for influenza virus reassortment independent of differential sialic acid distribution. The findings also establish that circulating neuraminidase (NA) and PA genes could alter the pathogenic phenotype of the pandemic H1N1 virus, resulting in enhanced disease. The identification of such factors provides a framework for pandemic modeling and surveillance.


2012 ◽  
Vol 93 (6) ◽  
pp. 1261-1268 ◽  
Author(s):  
Wenjun Ma ◽  
Qinfang Liu ◽  
Bhupinder Bawa ◽  
Chuanling Qiao ◽  
Wenbao Qi ◽  
...  

The 2009 pandemic H1N1 virus (pH1N1) contains neuraminidase (NA) and matrix (M) genes from Eurasian avian-like swine influenza viruses (SIVs), with the remaining six genes from North American triple-reassortant SIVs. To characterize the role of the pH1N1 NA and M genes in pathogenesis and transmission, their impact was evaluated in the background of an H1N1 triple-reassortant (tr1930) SIV in which the HA (H3) and NA (N2) of influenza A/swine/Texas/4199-2/98 virus were replaced with those from the classical H1N1 A/swine/Iowa/15/30 (1930) virus. The laboratory-adapted 1930 virus did not shed nor transmit in pigs, but tr1930 was able to shed in infected pigs. The NA, M or both genes of the tr1930 virus were then substituted by those of pH1N1. The resulting virus with both NA and M from pH1N1 grew to significantly higher titre in cell cultures than the viruses with single NA or M from pH1N1. In a pig model, only the virus containing both NA and M from pH1N1 was transmitted to and infected sentinels, whereas the viruses with single NA or M from pH1N1 did not. These results demonstrate that the right combination of NA and M genes is critical for the replication and transmissibility of influenza viruses in pigs.


2016 ◽  
Vol 90 (9) ◽  
pp. 4796-4806 ◽  
Author(s):  
Graham D. Williams ◽  
Amelia K. Pinto ◽  
Brittany Doll ◽  
Adrianus C. M. Boon

ABSTRACTReassortment between H5 or H9 subtype avian and mammalian influenza A viruses (IAV) can generate a novel virus that causes disease and transmits between mammals. Such information is currently not available for H7 subtype viruses. We evaluated the ability of a low-pathogenicity North American avian H7N3 virus (A/shorebird/Delaware/22/2006) to reassort with mammalian or avian viruses using a plasmid-based competition assay. In addition to genome segments derived from an avian H7N9 virus, the H7N3 virus reassorted efficiently with the PB2, NA, and M segments from the 2009 pandemic H1N1 (PH1N1) virus.In vitroandin vivoevaluation of the H7N3:PH1N1 (7 + 1) reassortant viruses revealed that the PB2, NA, or M segments fromPH1N1 largely do not attenuate the H7N3 virus, whereas the PB1, PA, NP, or NS genome segments fromPH1N1 do. Additionally, we assessed the functionality of the H7N3:PH1N1 7 + 1 reassortant viruses by measuring the inflammatory responsein vivo. We found that infection with wild-type H7N3 resulted in increased inflammatory cytokine production relative to that seen with thePH1N1 strain and that the increase was further exacerbated by substitution ofPH1N1 PB2 but not NA or M. Finally, we assessed if any adaptations occurred in the individually substituted segments afterin vivoinoculation and found no mutations, suggesting thatPH1N1 PB2, NA, and M are genetically stable in the background of this H7N3 virus. Taking the data together, we demonstrate that a North American avian H7N3 IAV is genetically and functionally compatible with multiple gene segments from the 2009 pandemic influenza virus strain without prior adaptation.IMPORTANCEThe 2009 pandemic H1N1 virus continues to circulate and reassort with other influenza viruses, creating novel viruses with increased replication and transmission potential in humans. Previous studies have found that this virus can also reassort with H5N1 and H9N2 avian influenza viruses. We now show that several genome segments of the 2009 H1N1 virus are also highly compatible with a low-pathogenicity avian H7N3 virus and that these reassortant viruses are stable and not attenuated in an animal model. These results highlight the potential for reassortment of H1N1 viruses with avian influenza virus and emphasize the need for continued surveillance of influenza viruses in areas of cocirculation between avian, human, and swine viruses.


mBio ◽  
2014 ◽  
Vol 5 (2) ◽  
Author(s):  
Wenjun Ma ◽  
Qinfang Liu ◽  
Chuanling Qiao ◽  
Gustavo del Real ◽  
Adolfo García-Sastre ◽  
...  

ABSTRACTThe 2009 pandemic H1N1 virus (pH1N1) was derived through reassortment of North American triple reassortant and Eurasian avian-like swine influenza viruses (SIVs). To date, when, how and where the pH1N1 arose is not understood. To investigate viral reassortment, we coinfected cell cultures and a group of pigs with or without preexisting immunity with a Eurasian H1N1 virus, A/Swine/Spain/53207/2004 (SP04), and a North American triple reassortant H1N1 virus, A/Swine/Kansas/77778/2007 (KS07). The infected pigs were cohoused with one or two groups of contact animals to investigate viral transmission. In coinfected MDCK or PK15 continuous cell lines with KS07 and SP04 viruses, more than 20 different reassortant viruses were found. In pigs without or with preexisting immunity (immunized with commercial inactivated swine influenza vaccines) and coinfected with both viruses, six or seven reassortant viruses, as well as the parental viruses, were identified in bronchoalveolar lavage fluid samples from the lungs. Interestingly, only one or two viruses transmitted to and were detected in contact animals. No reassortant containing a gene constellation similar to that of pH1N1 virus was found in either coinfected cells or pigs, indicating that the reassortment event that resulted in the generation of this virus is a rare event that likely involved specific viral strains and/or a favorable, not-yet-understood environment.IMPORTANCEThe 2009 pandemic-like H1N1 virus could not be reproduced either in cell cultures or in pigs coinfected with North American triple reassortant H1N1 and Eurasian H1N1 swine influenza viruses. This finding suggests that the generation of the 2009 pandemic H1N1 virus by reassortment was a rare event that likely involved specific viral strains and unknown factors. Different reassortant viruses were detected in coinfected pigs with and without preexisting immunity, indicating that host immunity plays a relevant role in driving viral reassortment of influenza A virus.


2010 ◽  
Vol 86 (1) ◽  
pp. A26
Author(s):  
Dale Barnard ◽  
Yousuke Furuta ◽  
Klaus Klummp ◽  
Donald Smee ◽  
John Morrey ◽  
...  

2019 ◽  
Vol 93 (23) ◽  
Author(s):  
Shuman Gao ◽  
Wenyu Zhang ◽  
Congyu Lu ◽  
Mengmeng Cao ◽  
Shan Cen ◽  
...  

ABSTRACT Type A and type B influenza viruses (FluA and FluB viruses) are two major human pathogens that share common structural and functional features. FluA and FluB viruses can reassort within each type but never between the types. Here, we bioinformatically analyzed all promoter sequences of FluA and FluB viruses and confirmed the presence of the type-specific promoter elements. We then studied the promoter elements with cell-based in vivo assays and an in vitro replication initiation assay. Our results identified, for the first time, a type-specific promoter element—the nucleotide at position 5 in the 3′ end of the viral RNA (vRNA)—that plays a key role(s) in modulating polymerase activity in a type-specific manner. Interestingly, swapping the promoter element between FluA and FluB recombinant viruses showed different tolerances: the replacement of FluA virus-specific U5 with FluB virus-specific C5 in influenza virus A/WSN/33 (H1N1) could be reverted to U5 after 2 to 3 passages, while the replacement of FluB virus-specific C5 with FluA virus-specific U5 in influenza virus B/Yamagata/88 could be maintained, but with significantly reduced replication efficiency. Therefore, our findings indicate that the nucleotide variation at position 5 in the 3′ end of the vRNA promoter between FluA and FluB viruses contributes to their RNP incompatibility, which may shed new light on the mechanisms of intertypic exclusion of reassortment between FluA and FluB viruses. IMPORTANCE Genetic reassortment of influenza virus plays a key role in virus evolution and the emergence of pandemic strains. The reassortment occurs extensively within either FluA or FluB viruses but never between them. Here, we bioinformatically compared available promoter sequences of FluA and FluB viruses and confirmed the presence of the type-specific promoter elements. Our in vivo and in vitro mutagenesis studies showed that a type-specific promoter element—the nucleotide at position 5 in the 3′ end of vRNA promoters—plays key roles in modulating polymerase activity. Interestingly, FluA and FluB viruses showed different tolerances upon key promoter element swapping in the context of virus infections. We concluded that the nucleotide at position 5 in the 3′ end of the vRNA promoters of FluA and FluB viruses is a critical type-specific determinant. This work has implications for further elucidating the mechanisms of the intertypic exclusion of reassortment between FluA and FluB viruses.


2012 ◽  
Vol 86 (17) ◽  
pp. 9361-9368 ◽  
Author(s):  
T. Watanabe ◽  
M. Imai ◽  
S. Watanabe ◽  
K. Shinya ◽  
M. Hatta ◽  
...  

Viruses ◽  
2020 ◽  
Vol 12 (1) ◽  
pp. 52 ◽  
Author(s):  
Mengkai Cai ◽  
Ruting Zhong ◽  
Chenxiao Qin ◽  
Zhiqing Yu ◽  
Xiaoyan Wen ◽  
...  

The Eurasian avian-like swine (EA) H1N1 virus has affected the Chinese swine industry, and human infection cases have been reported occasionally. However, little is known about the pathogenic mechanism of EA H1N1 virus. In this study, we compared the mouse pathogenicity of A/swine/Guangdong/YJ4/2014 (YJ4) and A/swine/Guangdong/MS285/2017 (MS285) viruses, which had similar genotype to A/Hunan/42443/2015 (HuN-like). None of the mice inoculated with 106 TCID50 of YJ4 survived at 7 days post infection, while the survival rate of the MS285 group was 100%. Therefore, a series of single fragment reassortants in MS285 background and two rescued wild-type viruses were generated by using the reverse genetics method, and the pathogenicity analysis revealed that the PB2 gene contributed to the high virulence of YJ4 virus. Furthermore, there were 11 amino acid differences in PB2 between MS285 and YJ4 identified by sequence alignment, and 11 single amino acid mutant viruses were generated in the MS285 background. We found that the R251K mutation significantly increased the virulence of MS285 in mice, contributed to high polymerase activity and enhanced viral genome transcription and replication. These results indicate that PB2-R251K contributes to the virulence of the EA H1N1 virus and provide new insight into future molecular epidemiological surveillance strategies.


Sign in / Sign up

Export Citation Format

Share Document