Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
Cell ; 186(19): 4074-4084.e11, 2023 09 14.
Article in English | MEDLINE | ID: mdl-37669665

ABSTRACT

H3N8 avian influenza viruses (AIVs) in China caused two confirmed human infections in 2022, followed by a fatal case reported in 2023. H3N8 viruses are widespread in chicken flocks; however, the zoonotic features of H3N8 viruses are poorly understood. Here, we demonstrate that H3N8 viruses were able to infect and replicate efficiently in organotypic normal human bronchial epithelial (NHBE) cells and lung epithelial (Calu-3) cells. Human isolates of H3N8 virus were more virulent and caused severe pathology in mice and ferrets, relative to chicken isolates. Importantly, H3N8 virus isolated from a patient with severe pneumonia was transmissible between ferrets through respiratory droplets; it had acquired human-receptor-binding preference and amino acid substitution PB2-E627K necessary for airborne transmission. Human populations, even when vaccinated against human H3N2 virus, appear immunologically naive to emerging mammalian-adapted H3N8 AIVs and could be vulnerable to infection at epidemic or pandemic proportion.


Subject(s)
Influenza A Virus, H3N8 Subtype , Influenza, Human , Animals , Humans , Mice , Chickens , Ferrets , Influenza A Virus, H3N2 Subtype , Respiratory Aerosols and Droplets
2.
Adv Sci (Weinh) ; 9(34): e2203088, 2022 12.
Article in English | MEDLINE | ID: mdl-36257906

ABSTRACT

Splicing of influenza A virus (IAV) RNA is an essential process in the viral life cycle that involves the co-opting of host factors. Here, it is demonstrated that induction of host serine and arginine-rich splicing factor 5 (SRSF5) by IAV facilitated viral replication by enhancing viral M mRNA splicing. Mechanistically, SRSF5 with its RRM2 domain directly bounds M mRNA at conserved sites (M mRNA position 163, 709, and 712), and interacts with U1 small nuclear ribonucleoprotein (snRNP) to promote M mRNA splicing and M2 production. Mutations introduced to the three binding sites, without changing amino acid code, significantly attenuates virus replication and pathogenesis in vivo. Likewise, SRSF5 conditional knockout in the lung protects mice against lethal IAV challenge. Furthermore, anidulafungin, an approved antifungal drug, is identified as an inhibitor of SRSF5 that effectively blocks IAV replication in vitro and in vivo. In conclusion, SRSF5 as an activator of M mRNA splicing promotes IAV replication and is a host-derived antiviral target.


Subject(s)
Influenza A virus , Orthomyxoviridae Infections , Animals , Mice , Alternative Splicing , RNA, Messenger , Virus Replication
3.
PLoS Pathog ; 18(7): e1010645, 2022 07.
Article in English | MEDLINE | ID: mdl-35793327

ABSTRACT

Avian influenza virus (AIV) can evolve multiple strategies to combat host antiviral defenses and establish efficient infectivity in mammals, including humans. H9N2 AIV and its reassortants (such as H5N6 and H7N9 viruses) pose an increasing threat to human health; however, the mechanisms involved in their increased virulence remain poorly understood. We previously reported that the M1 mutation T37A has become predominant among chicken H9N2 isolates in China. Here, we report that, since 2010, this mutation has also been found in the majority of human isolates of H9N2 AIV and its emerging reassortants. The T37A mutation of M1 protein enhances the replication of H9N2 AIVs in mice and in human cells. Interestingly, having A37 instead of T37 increases the M1 protein stability and resistance to proteasomal degradation. Moreover, T37 of the H9N2 M1 protein is phosphorylated by protein kinase G (PKG), and this phosphorylation induces the rapid degradation of M1 and reduces viral replication. Similar effects are also observed in the novel H5N6 virus. Additionally, ubiquitination at K187 contributes to M1-37T degradation and decreased replication of the virus harboring T37 in the M1 protein. The prevailing AIVs thereby evolve a phospho-resistant mutation in the M1 protein to avoid viral protein degradation by host factors, which is advantageous in terms of replication in mammalian hosts.


Subject(s)
Influenza A Virus, H7N9 Subtype , Influenza A Virus, H9N2 Subtype , Influenza in Birds , Orthomyxoviridae Infections , Animals , Influenza A Virus, H7N9 Subtype/genetics , Influenza in Birds/genetics , Mammals , Mice , Mutation
4.
PNAS Nexus ; 1(3): pgac085, 2022 Jul.
Article in English | MEDLINE | ID: mdl-36741455

ABSTRACT

Clade 2.3.4.4 avian H5Ny viruses, namely H5N2, H5N6, and H5N8, have exhibited unprecedented intercontinental spread in poultry. Among them, only H5N6 viruses are frequently reported to infect mammals and cause serious human infections. In this study, the genetic and biological characteristics of surface hemagglutinin (HA) from clade 2.3.4.4 H5Ny avian influenza viruses (AIVs) were examined for adaptation in mammalian infection. Phylogenetic analysis identified an amino acid (AA) deletion at position 131 of HA as a distinctive feature of H5N6 virus isolated from human patients. This single AA deletion was found to enhance H5N6 virus replication and pathogenicity in vitro and in mammalian hosts (mice and ferrets) through HA protein acid and thermal stabilization that resulted in reduced pH threshold from pH 5.7 to 5.5 for viral-endosomal membrane fusion. Mass spectrometry and crystal structure revealed that the AA deletion in HA at position 131 introduced an N-linked glycosylation site at 129, which increases compactness between HA monomers, thus stabilizes the trimeric structure. Our findings provide a molecular understanding of how HA protein stabilization promotes cross-species avian H5N6 virus infection to mammalian hosts.

5.
PLoS Pathog ; 17(12): e1010098, 2021 12.
Article in English | MEDLINE | ID: mdl-34860863

ABSTRACT

H5N6 highly pathogenic avian influenza virus (HPAIV) clade 2.3.4.4 not only exhibits unprecedented intercontinental spread in poultry, but can also cause serious infection in humans, posing a public health threat. Phylogenetic analyses show that 40% (8/20) of H5N6 viruses that infected humans carried H9N2 virus-derived internal genes. However, the precise contribution of H9N2 virus-derived internal genes to H5N6 virus infection in humans is unclear. Here, we report on the functional contribution of the H9N2 virus-derived matrix protein 1 (M1) to enhanced H5N6 virus replication capacity in mammalian cells. Unlike H5N1 virus-derived M1 protein, H9N2 virus-derived M1 protein showed high binding affinity for H5N6 hemagglutinin (HA) protein and increased viral progeny particle release in different mammalian cell lines. Human host factor, G protein subunit beta 1 (GNB1), exhibited strong binding to H9N2 virus-derived M1 protein to facilitate M1 transport to budding sites at the cell membrane. GNB1 knockdown inhibited the interaction between H9N2 virus-derived M1 and HA protein, and reduced influenza virus-like particles (VLPs) release. Our findings indicate that H9N2 virus-derived M1 protein promotes avian H5N6 influenza virus release from mammalian, in particular human cells, which could be a major viral factor for H5N6 virus cross-species infection.


Subject(s)
Influenza A Virus, H9N2 Subtype/genetics , Influenza in Birds/virology , Influenza, Human/virology , Reassortant Viruses/genetics , Viral Matrix Proteins/metabolism , Viral Zoonoses/virology , Animals , Chickens/virology , Humans , Influenza A virus/genetics , Virus Release
6.
Virulence ; 12(1): 2946-2956, 2021 12.
Article in English | MEDLINE | ID: mdl-34793280

ABSTRACT

The struggle to control the COVID-19 pandemic is made challenging by the emergence of virulent SARS-CoV-2 variants. To gain insight into their replication dynamics, emergent Alpha (A), Beta (B) and Delta (D) SARS-CoV-2 variants were assessed for their infection performance in single variant- and co-infections. The effectiveness of thapsigargin (TG), a recently discovered broad-spectrum antiviral, against these variants was also examined. Of the 3 viruses, the D variant exhibited the highest replication rate and was most able to spread to in-contact cells; its replication rate at 24 h post-infection (hpi) based on progeny viral RNA production was over 4 times that of variant A and 9 times more than the B variant. In co-infections, the D variant boosted the replication of its co-infected partners at the expense of its own initial performance. Furthermore, co-infection with AD or AB combination conferred replication synergy where total progeny (RNA) output was greater than the sum of corresponding single-variant infections. All variants were highly sensitive to TG inhibition. A single pre-infection priming dose of TG effectively blocked all single-variant infections and every combination (AB, AD, BD variants) of co-infection at greater than 95% (relative to controls) at 72 hpi. Likewise, TG was effective in inhibiting each variant in active preexisting infection. In conclusion, against the current backdrop of the dominant D variant that could be further complicated by co-infection synergy with new variants, the growing list of viruses susceptible to TG, a promising host-centric antiviral, now includes a spectrum of contemporary SARS-CoV-2 viruses.


Subject(s)
COVID-19 Drug Treatment , Coinfection , SARS-CoV-2 , Thapsigargin , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Humans , Pandemics , SARS-CoV-2/drug effects , Thapsigargin/pharmacology , Thapsigargin/therapeutic use
7.
Nat Microbiol ; 6(7): 932-945, 2021 07.
Article in English | MEDLINE | ID: mdl-33986530

ABSTRACT

The retinoic acid-inducible gene I (RIG-I) receptor senses cytoplasmic viral RNA and activates type I interferons (IFN-I) and downstream antiviral immune responses. How RIG-I binds to viral RNA and how its activation is regulated remains unclear. Here, using IFI16 knockout cells and p204-deficient mice, we demonstrate that the DNA sensor IFI16 enhances IFN-I production to inhibit influenza A virus (IAV) replication. IFI16 positively upregulates RIG-I transcription through direct binding to and recruitment of RNA polymerase II to the RIG-I promoter. IFI16 also binds to influenza viral RNA via its HINa domain and to RIG-I protein with its PYRIN domain, thus promoting IAV-induced K63-linked polyubiquitination and RIG-I activation. Our work demonstrates that IFI16 is a positive regulator of RIG-I signalling during influenza virus infection, highlighting its role in the RIG-I-like-receptor-mediated innate immune response to IAV and other RNA viruses, and suggesting its possible exploitation to modulate the antiviral response.


Subject(s)
DEAD Box Protein 58/genetics , Influenza A virus/physiology , Nuclear Proteins/metabolism , Orthomyxoviridae Infections/virology , Phosphoproteins/metabolism , RNA, Viral/metabolism , Receptors, Immunologic/genetics , Animals , DEAD Box Protein 58/metabolism , Host-Pathogen Interactions , Humans , Immunity, Innate , Interferon Type I/metabolism , Mice , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/metabolism , Phosphoproteins/chemistry , Phosphoproteins/genetics , Promoter Regions, Genetic , Protein Binding , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Receptors, Immunologic/metabolism , Signal Transduction , Up-Regulation/genetics
8.
Emerg Microbes Infect ; 10(1): 472-480, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33657971

ABSTRACT

Pandemic influenza, typically caused by the reassortment of human and avian influenza viruses, can result in severe or fatal infections in humans. Timely identification of potential pandemic viruses must be a priority in influenza virus surveillance. However, the range of host species responsible for the generation of novel pandemic influenza viruses remains unclear. In this study, we conducted serological surveys for avian and human influenza virus infections in farmed mink and determined the susceptibility of mink to prevailing avian and human virus subtypes. The results showed that farmed mink were commonly infected with human (H3N2 and H1N1/pdm) and avian (H7N9, H5N6, and H9N2) influenza A viruses. Correlational analysis indicated that transmission of human influenza viruses occurred from humans to mink, and that feed source was a probable route of avian influenza virus transmission to farmed mink. Animal experiments showed that mink were susceptible and permissive to circulating avian and human influenza viruses, and that human influenza viruses (H3N2 and H1N1/pdm), but not avian viruses, were capable of aerosol transmission among mink. These results indicate that farmed mink could be highly permissive "mixing vessels" for the reassortment of circulating human and avian influenza viruses. Therefore, to reduce the risk of emergence of novel pandemic viruses, feeding mink with raw poultry by-products should not be permitted, and epidemiological surveillance of influenza viruses in mink farms should be urgently implemented.


Subject(s)
Influenza A virus/pathogenicity , Mink/virology , Orthomyxoviridae Infections/transmission , Animals , Disease Models, Animal , Humans , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza A Virus, H3N2 Subtype/immunology , Influenza A Virus, H3N2 Subtype/pathogenicity , Influenza A Virus, H7N9 Subtype/immunology , Influenza A Virus, H7N9 Subtype/pathogenicity , Influenza A Virus, H9N2 Subtype/immunology , Influenza A Virus, H9N2 Subtype/pathogenicity , Influenza A virus/immunology , Mink/immunology , Neutralization Tests , Orthomyxoviridae Infections/immunology , Reassortant Viruses/immunology , Reassortant Viruses/pathogenicity
9.
J Virol ; 95(11)2021 05 10.
Article in English | MEDLINE | ID: mdl-33731452

ABSTRACT

H9N2 Avian influenza virus (AIV) is regarded as a principal donor of viral genes through reassortment to co-circulating influenza viruses that can result in zoonotic reassortants. Whether H9N2 virus can maintain sustained evolutionary impact on such reassortants is unclear. Since 2013, avian H7N9 virus had caused five sequential human epidemics in China; the fifth wave in 2016-2017 was by far the largest but the mechanistic explanation behind the scale of infection is not clear. Here, we found that, just prior to the fifth H7N9 virus epidemic, H9N2 viruses had phylogenetically mutated into new sub-clades, changed antigenicity and increased its prevalence in chickens vaccinated with existing H9N2 vaccines. In turn, the new H9N2 virus sub-clades of PB2 and PA genes, housing mammalian adaptive mutations, were reassorted into co-circulating H7N9 virus to create a novel dominant H7N9 virus genotype that was responsible for the fifth H7N9 virus epidemic. H9N2-derived PB2 and PA genes in H7N9 virus conferred enhanced polymerase activity in human cells at 33°C and 37°C, and increased viral replication in the upper and lower respiratory tracts of infected mice which could account for the sharp increase in human cases of H7N9 virus infection in the 2016-2017 epidemic. The role of H9N2 virus in the continual mutation of H7N9 virus highlights the public health significance of H9N2 virus in the generation of variant reassortants of increasing zoonotic potential.IMPORTANCEAvian H9N2 influenza virus, although primarily restricted to chicken populations, is a major threat to human public health by acting as a donor of variant viral genes through reassortment to co-circulating influenza viruses. We established that the high prevalence of evolving H9N2 virus in vaccinated flocks played a key role, as donor of new sub-clade PB2 and PA genes in the generation of a dominant H7N9 virus genotype (G72) with enhanced infectivity in humans during the 2016-2017 N7N9 virus epidemic. Our findings emphasize that the ongoing evolution of prevalent H9N2 virus in chickens is an important source, via reassortment, of mammalian adaptive genes for other influenza virus subtypes. Thus, close monitoring of prevalence and variants of H9N2 virus in chicken flocks is necessary in the detection of zoonotic mutations.

10.
Viruses ; 13(2)2021 02 03.
Article in English | MEDLINE | ID: mdl-33546185

ABSTRACT

The long-term control strategy of SARS-CoV-2 and other major respiratory viruses needs to include antivirals to treat acute infections, in addition to the judicious use of effective vaccines. Whilst COVID-19 vaccines are being rolled out for mass vaccination, the modest number of antivirals in use or development for any disease bears testament to the challenges of antiviral development. We recently showed that non-cytotoxic levels of thapsigargin (TG), an inhibitor of the sarcoplasmic/endoplasmic reticulum (ER) Ca2+ ATPase pump, induces a potent host innate immune antiviral response that blocks influenza A virus replication. Here we show that TG is also highly effective in blocking the replication of respiratory syncytial virus (RSV), common cold coronavirus OC43, SARS-CoV-2 and influenza A virus in immortalized or primary human cells. TG's antiviral performance was significantly better than remdesivir and ribavirin in their respective inhibition of OC43 and RSV. Notably, TG was just as inhibitory to coronaviruses (OC43 and SARS-CoV-2) and influenza viruses (USSR H1N1 and pdm 2009 H1N1) in separate infections as in co-infections. Post-infection oral gavage of acid-stable TG protected mice against a lethal influenza virus challenge. Together with its ability to inhibit the different viruses before or during active infection, and with an antiviral duration of at least 48 h post-TG exposure, we propose that TG (or its derivatives) is a promising broad-spectrum inhibitor against SARS-CoV-2, OC43, RSV and influenza virus.


Subject(s)
Antiviral Agents/pharmacology , Betacoronavirus/drug effects , Coronavirus OC43, Human/drug effects , Influenza A Virus, H1N1 Subtype/drug effects , Respiratory Syncytial Virus, Human/drug effects , SARS-CoV-2/drug effects , Thapsigargin/pharmacology , Animals , Antiviral Agents/therapeutic use , Betacoronavirus/physiology , Cell Line , Cell Line, Tumor , Cells, Cultured , Coronavirus OC43, Human/physiology , Endoplasmic Reticulum Stress , Humans , Influenza A Virus, H1N1 Subtype/physiology , Mice , Microbial Sensitivity Tests , Orthomyxoviridae Infections/drug therapy , Orthomyxoviridae Infections/virology , Respiratory Syncytial Virus, Human/physiology , Ribavirin/pharmacology , SARS-CoV-2/physiology , Thapsigargin/therapeutic use , Virus Replication/drug effects
11.
J Virol ; 95(7)2021 03 10.
Article in English | MEDLINE | ID: mdl-33408179

ABSTRACT

Avian influenza viruses (AIVs) are zoonotic viruses that exhibit a range infectivity and severity in the human host. Severe human cases of AIVs infection are often accompanied by neurological symptoms, however, the factors involved in the infection of the central nervous system (CNS) are not well known. In this study, we discovered that avian-like sialic acid (SA)-α2, 3 Gal receptor is highly presented in mammalian (human and mouse) brains. In the generation of a mouse-adapted neurotropic H9N2 AIV (SD16-MA virus) in BALB/c mice, we identified key adaptive mutations in its hemagglutinin (HA) and polymerase basic protein 2 (PB2) genes that conferred viral replication ability in mice brain. The SD16-MA virus showed binding affinity for avian-like SA-α2, 3 Gal receptor, enhanced viral RNP polymerase activity, increased viral protein production and transport that culminated in elevated progeny virus production and severe pathogenicity. We further established that host Fragile X Mental Retardation Protein (FMRP), a highly expressed protein in the brain that physically associated with viral nucleocapsid protein (NP) to facilitate RNP assembly and export, was an essential host factor for the neuronal replication of neurotropic AIVs (H9N2, H5N1 and H10N7 viruses). Our study identified a mechanistic process for AIVs to acquire neurovirulence in mice.IMPORTANCE Infection of the CNS is a serious complication of human cases of AIVs infection. The viral and host factors associated with neurovirulence of AIVs infection are not well understood. We identified and functionally characterized specific changes in the viral HA and PB2 genes of a mouse-adapted neurotropic avian H9N2 virus responsible for enhanced virus replication in neuronal cells and pathogenicity in mice. Importantly, we showed that host FMRP was a crucial host factor that was necessary for neurotropic AIVs (H9N2, H5N1 and H10N7 viruses) to replicate in neuronal cells. Our findings have provided insights into the pathogenesis of neurovirulence of AIV infection.

12.
Viruses ; 12(10)2020 09 27.
Article in English | MEDLINE | ID: mdl-32992478

ABSTRACT

Influenza A virus is a major global pathogen of humans, and there is an unmet need for effective antivirals. Current antivirals against influenza A virus directly target the virus and are vulnerable to mutational resistance. Harnessing an effective host antiviral response is an attractive alternative. We show that brief exposure to low, non-toxic doses of thapsigargin (TG), an inhibitor of the sarcoplasmic/endoplasmic reticulum (ER) Ca2+ ATPase pump, promptly elicits an extended antiviral state that dramatically blocks influenza A virus production. Crucially, oral administration of TG protected mice against lethal virus infection and reduced virus titres in the lungs of treated mice. TG-induced ER stress unfolded protein response appears as a key driver responsible for activating a spectrum of host antiviral defences that include an enhanced type I/III interferon response. Our findings suggest that TG is potentially a viable host-centric antiviral for the treatment of influenza A virus infection without the inherent problem of drug resistance.


Subject(s)
Antiviral Agents/pharmacology , Influenza A Virus, H1N1 Subtype/growth & development , Influenza A Virus, H3N8 Subtype/growth & development , Thapsigargin/pharmacology , Virus Replication/drug effects , Animals , Cell Line , Chick Embryo , Chlorocebus aethiops , Dogs , Endoplasmic Reticulum Stress/drug effects , Female , Host-Pathogen Interactions/drug effects , Humans , Immunity, Innate/drug effects , Immunity, Innate/immunology , Influenza, Human/drug therapy , Interferon Type I/drug effects , Interferon Type I/immunology , Interferons/drug effects , Interferons/immunology , Mice , Mice, Inbred BALB C , Sarcoplasmic Reticulum Calcium-Transporting ATPases/antagonists & inhibitors , Swine , Unfolded Protein Response/drug effects , Vero Cells , Interferon Lambda
13.
Proc Natl Acad Sci U S A ; 117(29): 17204-17210, 2020 07 21.
Article in English | MEDLINE | ID: mdl-32601207

ABSTRACT

Pigs are considered as important hosts or "mixing vessels" for the generation of pandemic influenza viruses. Systematic surveillance of influenza viruses in pigs is essential for early warning and preparedness for the next potential pandemic. Here, we report on an influenza virus surveillance of pigs from 2011 to 2018 in China, and identify a recently emerged genotype 4 (G4) reassortant Eurasian avian-like (EA) H1N1 virus, which bears 2009 pandemic (pdm/09) and triple-reassortant (TR)-derived internal genes and has been predominant in swine populations since 2016. Similar to pdm/09 virus, G4 viruses bind to human-type receptors, produce much higher progeny virus in human airway epithelial cells, and show efficient infectivity and aerosol transmission in ferrets. Moreover, low antigenic cross-reactivity of human influenza vaccine strains with G4 reassortant EA H1N1 virus indicates that preexisting population immunity does not provide protection against G4 viruses. Further serological surveillance among occupational exposure population showed that 10.4% (35/338) of swine workers were positive for G4 EA H1N1 virus, especially for participants 18 y to 35 y old, who had 20.5% (9/44) seropositive rates, indicating that the predominant G4 EA H1N1 virus has acquired increased human infectivity. Such infectivity greatly enhances the opportunity for virus adaptation in humans and raises concerns for the possible generation of pandemic viruses.


Subject(s)
Genes, Viral , Influenza A Virus, H1N1 Subtype/genetics , Influenza, Human/epidemiology , Influenza, Human/virology , Orthomyxoviridae Infections/epidemiology , Orthomyxoviridae Infections/virology , Swine Diseases/epidemiology , Swine Diseases/virology , Animals , China , Cross Reactions , Epithelial Cells/virology , Genetic Variation , Genotype , Humans , Influenza A Virus, H1N1 Subtype/classification , Influenza, Human/immunology , Influenza, Human/transmission , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/transmission , Pandemics , Phylogeny , Prevalence , Reassortant Viruses/genetics , Seroepidemiologic Studies , Swine
14.
J Gen Virol ; 100(9): 1273-1281, 2019 09.
Article in English | MEDLINE | ID: mdl-31305236

ABSTRACT

Adaptation of PB2 protein is important for the establishment of avian influenza viruses in mammalian hosts. Here, we identify I292V as the prevalent mutation in PB2 of circulating avian H9N2 and pandemic H1N1 viruses. The same dominant PB2 mutation is also found in most human isolates of emergent avian H7N9 and H10N8 viruses. In human cells, PB2-292V in H9N2 virus has the combined ability of conferring higher viral polymerase activity and stronger attenuation of IFN-ß induction than that of its predecessor PB2-292I. IFN-ß attenuation is accompanied by higher binding affinity of PB2-292V for host mitochondrial antiviral signalling protein, an important intermediary protein in the induction of IFN-ß. In the mouse in vivo model, PB2-292V mutation increases H9N2 virus replication with ensuing increase in disease severity. Collectively, PB2-292V is a new mammalian adaptive marker that promotes H9N2 virus replication in mammalian hosts with the potential to improve transmission from birds to humans.


Subject(s)
DNA-Directed DNA Polymerase/metabolism , Influenza A Virus, H9N2 Subtype/physiology , Influenza in Birds/virology , Interferon-beta/metabolism , Viral Proteins/metabolism , Adaptation, Physiological/genetics , Animals , Chickens , DNA-Directed DNA Polymerase/genetics , Female , Gene Expression Regulation, Enzymologic , HEK293 Cells , Humans , Influenza A Virus, H1N1 Subtype , Influenza, Human/virology , Interferon-beta/genetics , Mice , Mice, Inbred BALB C , Mutation , Species Specificity , Viral Proteins/genetics
15.
Virol J ; 15(1): 68, 2018 04 10.
Article in English | MEDLINE | ID: mdl-29636078

ABSTRACT

BACKGROUND: With the recent discovery of novel H17N10 and H18N11 influenza viral RNA in bats and report on high frequency of avian H9 seroconversion in a species of free ranging bats, an important issue to address is the extent bats are susceptible to conventional avian and human influenza A viruses. METHOD: To this end, three bat species (Eidolon helvum, Carollia perspicillata and Tadarida brasiliensis) of lung epithelial cells were separately infected with two avian and two human influenza viruses to determine their relative host innate immune resistance to infection. RESULTS: All three species of bat cells were more resistant than positive control Madin-Darby canine kidney (MDCK) cells to all four influenza viruses. TB1-Lu cells lacked sialic acid α2,6-Gal receptors and were most resistant among the three bat species. Interestingly, avian viruses were relatively more replication permissive in all three bat species of cells than with the use of human viruses which suggest that bats could potentially play a role in the ecology of avian influenza viruses. Chemical inhibition of the JAK-STAT pathway in bat cells had no effect on virus production suggesting that type I interferon signalling is not a major factor in resisting influenza virus infection. CONCLUSION: Although all three species of bat cells are relatively more resistant to influenza virus infection than control MDCK cells, they are more permissive to avian than human viruses which suggest that bats could have a contributory role in the ecology of avian influenza viruses.


Subject(s)
Chiroptera , Epithelial Cells/virology , Host Specificity , Influenza A virus/immunology , Orthomyxoviridae Infections/virology , Animals , Cell Survival , Cells, Cultured , Dogs , Gene Expression , Influenza A virus/classification , Lung/cytology , Lung/immunology , Madin Darby Canine Kidney Cells , Neuraminidase/pharmacology , Receptors, Virus/metabolism , Viral Proteins/genetics , Virus Release/drug effects , Virus Replication
16.
J Gen Virol ; 98(6): 1185-1195, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28597818

ABSTRACT

The H10 subtype of avian influenza viruses (AIVs) circulates globally in wild birds and poultry, and this subtype has been shown to be increasingly prevalent in China. Among the various H10 viruses, H10N7 AIVs have caused repeated mammal and human infections. To investigate their genetic adaptation in mammals, we generated a mouse-adapted avian H10N7 variant (A/mallard/Beijing/27/2011-MA; BJ27-MA) which exhibited increased virulence in mice compared to wild-type virus and acquired neurotropism. Sequencing showed the absence of the widely recognized mammalian adaptation markers of E627K and D701N in PB2 in the mouse-adapted strain; instead, five amino acid mutations were identified: E158G and M631L in PB2; G218E in haemagglutinin (H3 numbering); and K110E and S453I in neuraminidase (NA). The neurovirulence of the BJ27-MA virus necessitated the combined presence of the PB2 and NA mutations. Mutations M631L and E158G of PB2 and K110E of NA were required to mediate increased virus replication and severity of infection in mice and mammalian cells. PB2-M631L was functionally the most dominant mutation in that it strongly upregulated viral polymerase activity and played a critical role in the enhancement of virus replication and disease severity in mice. K110E mutation in NA, on the other hand, significantly promoted NA enzymatic activity. These results indicate that the novel mutations in PB2 and NA genes are critical for the adaptation of H10N7 AIV in mice, and they could serve as molecular signatures of virus transmission to mammalian hosts, including humans.


Subject(s)
Adaptation, Biological , Influenza A virus/growth & development , Influenza A virus/pathogenicity , Mutation, Missense , Neuraminidase/genetics , RNA-Dependent RNA Polymerase/genetics , Viral Proteins/genetics , Virulence Factors/genetics , Animals , Central Nervous System/virology , Female , Mice, Inbred BALB C , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology , Serial Passage , Viral Tropism , Virulence
17.
J Virol ; 91(8)2017 04 15.
Article in English | MEDLINE | ID: mdl-28148803

ABSTRACT

Segment reassortment and base mutagenesis of influenza A viruses are the primary routes to the rapid evolution of high-fitness virus genotypes. We recently described a predominant G57 genotype of avian H9N2 viruses that caused countrywide outbreaks in chickens in China during 2010 to 2013, which led to the zoonotic emergence of H7N9 viruses. One of the key features of the G57 genotype is the replacement of the earlier A/chicken/Beijing/1/1994 (BJ/94)-like M gene with the A/quail/Hong Kong/G1/1997 (G1)-like M gene of quail origin. We report here the functional significance of the G1-like M gene in H9N2 viruses in conferring increased infection severity and infectivity in primary chicken embryonic fibroblasts and chickens. H9N2 virus housing the G1-like M gene, in place of the BJ/94-like M gene, showed an early surge in viral mRNA and viral RNA (vRNA) transcription that was associated with enhanced viral protein production and with an early elevated release of progeny virus comprising largely spherical rather than filamentous virions. Importantly, H9N2 virus with the G1-like M gene conferred extrapulmonary virus spread in chickens. Five highly represented signature amino acid residues (37A, 95K, 224N, and 242N in the M1 protein and 21G in the M2 protein) encoded by the prevalent G1-like M gene were demonstrated to be prime contributors to enhanced infectivity. Therefore, the genetic evolution of the M gene in H9N2 virus increases reproductive virus fitness, indicating its contribution to the rising virus prevalence in chickens in China.IMPORTANCE We recently described the circulation of a dominant genotype (genotype G57) of H9N2 viruses in countrywide outbreaks in chickens in China, which was responsible, through reassortment, for the emergence of H7N9 viruses that cause severe human infections. A key feature of the genotype G57 H9N2 virus is the presence of the quail-origin G1-like M gene, which had replaced the earlier BJ/94-like M gene. We found that H9N2 virus with the G1-like M gene, but not the BJ/94-like M gene, showed an early surge in progeny virus production and more severe pathology and extrapulmonary virus spread in chickens. Five highly represented amino acid residues in the M1 and M2 proteins derived from the G1-like M gene were shown to mediate enhanced virus infectivity. These observations enhance what we currently know about the roles of reassortment and mutations in virus fitness and have implications for assessing the potential of variant influenza viruses that can cause a rising prevalence in chickens.


Subject(s)
Fibroblasts/virology , Influenza A Virus, H9N2 Subtype/physiology , Influenza in Birds/pathology , Reassortant Viruses/physiology , Viral Matrix Proteins/genetics , Virulence Factors/genetics , Virus Replication , Animals , Chickens , DNA Mutational Analysis , Influenza A Virus, H9N2 Subtype/genetics , Influenza A Virus, H9N2 Subtype/pathogenicity , Influenza A Virus, H9N2 Subtype/ultrastructure , Influenza in Birds/virology , Reassortant Viruses/genetics , Virion/ultrastructure , Virulence
18.
J Virol ; 90(18): 8105-14, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27384648

ABSTRACT

UNLABELLED: Adaptation of the viral polymerase complex comprising PB1, PB2, and PA is necessary for efficient influenza A virus replication in new host species. We found that PA mutation K356R (PA-K356R) has become predominant since 2014 in avian H9N2 viruses in China as with seasonal human H1N1 viruses. The same mutation is also found in most human isolates of emergent avian H7N9 and H10N8 viruses whose six internal gene segments are derived from the H9N2 virus. We further demonstrated the mammalian adaptive functionality of the PA-K356R mutation. Avian H9N2 virus with the PA-K356R mutation in human A549 cells showed increased nuclear accumulation of PA and increased viral polymerase activity that resulted in elevated levels of viral transcription and virus output. The same mutant virus in mice also enhanced virus replication and caused lethal infection. In addition, combined mutation of PA-K356R and PB2-E627K, a well-known mammalian adaptive marker, in the H9N2 virus showed further cooperative increases in virus production and severity of infection in vitro and in vivo In summary, PA-K356R behaves as a novel mammalian tropism mutation, which, along with other mutations such as PB2-E627K, might render avian H9N2 viruses adapted for human infection. IMPORTANCE: Mutations of the polymerase complex (PB1, PB2, and PA) of influenza A virus are necessary for viral adaptation to new hosts. This study reports a novel and predominant mammalian adaptive mutation, PA-K356R, in avian H9N2 viruses and human isolates of emergent H7N9 and H10N8 viruses. We found that PA-356R in H9N2 viruses causes significant increases in virus replication and severity of infection in human cells and mice and that PA-K356R cooperates with the PB2-E627K mutation, a well-characterized human adaptive marker, to exacerbate mammalian infection in vitro and in vivo Therefore, the PA-K356R mutation is a significant adaptation in H9N2 viruses and related H7N9 and H10N8 reassortants toward human infectivity.


Subject(s)
Influenza A Virus, H9N2 Subtype/physiology , Influenza A Virus, H9N2 Subtype/pathogenicity , Mutant Proteins/metabolism , Mutation, Missense , RNA-Dependent RNA Polymerase/metabolism , Viral Proteins/metabolism , Virus Replication , Animals , Cell Line , Disease Models, Animal , Epithelial Cells/virology , Humans , Mice , Mutant Proteins/genetics , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology , RNA-Dependent RNA Polymerase/genetics , Survival Analysis , Viral Proteins/genetics , Viral Tropism
19.
Sci Rep ; 6: 27067, 2016 06 02.
Article in English | MEDLINE | ID: mdl-27252023

ABSTRACT

Given the present extensive co-circulation in pigs of Eurasian avian-like (EA) swine H1N1 and 2009 pandemic (pdm/09) H1N1 viruses, reassortment between them is highly plausible but largely uncharacterized. Here, experimentally co-infected pigs with a representative EA virus and a pdm/09 virus yielded 55 novel reassortant viruses that could be categorized into 17 genotypes from Gt1 to Gt17 based on segment segregation. Majority of novel reassortants were isolated from the lower respiratory tract. Most of reassortant viruses were more pathogenic and contagious than the parental EA viruses in mice and guinea pigs. The most transmissible reassortant genotypes demonstrated in guinea pigs (Gt2, Gt3, Gt7, Gt10 and Gt13) were also the most lethal in mice. Notably, nearly all these highly virulent reassortants (all except Gt13) were characterized with possession of EA H1 and full complement of pdm/09 ribonucleoprotein genes. Compositionally, we demonstrated that EA H1-222G contributed to virulence by its ability to bind avian-type sialic acid receptors, and that pdm/09 RNP conferred the most robust polymerase activity to reassortants. The present study revealed high reassortment compatibility between EA and pdm/09 viruses in pigs, which could give rise to progeny reassortant viruses with enhanced virulence and transmissibility in mice and guinea pig models.


Subject(s)
Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza, Human/transmission , Animals , Birds , Coinfection/virology , Dogs , Female , Genes, Viral , Genotype , Guinea Pigs , HEK293 Cells , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Humans , Hybridization, Genetic , Influenza Vaccines/immunology , Influenza in Birds/virology , Influenza, Human/prevention & control , Influenza, Human/virology , Madin Darby Canine Kidney Cells , Mice, Inbred BALB C , Pandemics , Protein Binding , Receptors, Cell Surface/chemistry , Respiratory System/virology , Sus scrofa , Virulence , Virus Replication
20.
J Virol ; 90(14): 6235-6243, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27122581

ABSTRACT

UNLABELLED: Since May 2014, highly pathogenic avian influenza H5N6 virus has been reported to cause six severe human infections three of which were fatal. The biological properties of this subtype, in particular its relative pathogenicity and transmissibility in mammals, are not known. We characterized the virus receptor-binding affinity, pathogenicity, and transmissibility in mice and ferrets of four H5N6 isolates derived from waterfowl in China from 2013-2014. All four H5N6 viruses have acquired a binding affinity for human-like SAα2,6Gal-linked receptor to be able to attach to human tracheal epithelial and alveolar cells. The emergent H5N6 viruses, which share high sequence similarity with the human isolate A/Guangzhou/39715/2014 (H5N6), were fully infective and highly transmissible by direct contact in ferrets but showed less-severe pathogenicity than the parental H5N1 virus. The present results highlight the threat of emergent H5N6 viruses to poultry and human health and the need to closely track their continual adaptation in humans. IMPORTANCE: Extended epizootics and panzootics of H5N1 viruses have led to the emergence of the novel 2.3.4.4 clade of H5 virus subtypes, including H5N2, H5N6, and H5N8 reassortants. Avian H5N6 viruses from this clade have caused three fatalities out of six severe human infections in China since the first case in 2014. However, the biological properties of this subtype, especially the pathogenicity and transmission in mammals, are not known. Here, we found that natural avian H5N6 viruses have acquired a high affinity for human-type virus receptor. Compared to the parental clade 2.3.4 H5N1 virus, emergent H5N6 isolates showed less severe pathogenicity in mice and ferrets but acquired efficient in-contact transmission in ferrets. These findings suggest that the threat of avian H5N6 viruses to humans should not be ignored.


Subject(s)
Influenza A virus/pathogenicity , Influenza, Human/transmission , Orthomyxoviridae Infections/transmission , Reassortant Viruses/pathogenicity , Receptors, Cell Surface/metabolism , Virus Attachment , Animals , China , Disease Models, Animal , Epithelial Cells/metabolism , Epithelial Cells/virology , Female , Ferrets , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Humans , Influenza A virus/classification , Influenza, Human/pathology , Influenza, Human/virology , Male , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology , Phylogeny , Respiratory Mucosa/metabolism , Respiratory Mucosa/virology , Virulence
SELECTION OF CITATIONS
SEARCH DETAIL
...