51
|
Sharma S, Mayank AK, Nailwal H, Tripathi S, Patel JR, Bowzard JB, Gaur P, Donis RO, Katz JM, Cox NJ, Lal RB, Farooqi H, Sambhara S, Lal SK. Influenza A viral nucleoprotein interacts with cytoskeleton scaffolding protein α-actinin-4 for viral replication. FEBS J 2014; 281:2899-914. [PMID: 24802111 PMCID: PMC7164065 DOI: 10.1111/febs.12828] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Revised: 04/03/2014] [Accepted: 04/30/2014] [Indexed: 02/03/2023]
Abstract
Influenza A virus (IAV), similar to other viruses, exploits the machinery of human host cells for its survival and replication. We identified α‐actinin‐4, a host cytoskeletal protein, as an interacting partner of IAV nucleoprotein (NP). We confirmed this interaction using co‐immunoprecipitation studies, first in a coupled in vitro transcription‐translation assay and then in cells either transiently co‐expressing the two proteins or infected with whole IAV. Importantly, the NP–actinin‐4 interaction was observed in several IAV subtypes, including the 2009 H1N1 pandemic virus. Moreover, immunofluorescence studies revealed that both NP and actinin‐4 co‐localized largely around the nucleus and also in the cytoplasmic region of virus‐infected A549 cells. Silencing of actinin‐4 expression resulted in not only a significant decrease in NP, M2 and NS1 viral protein expression, but also a reduction of both NP mRNA and viral RNA levels, as well as viral titers, 24 h post‐infection with IAV, suggesting that actinin‐4 was critical for viral replication. Furthermore, actinin‐4 depletion reduced the amount of NP localized in the nucleus. Treatment of infected cells with wortmannin, a known inhibitor of actinin‐4, led to a decrease in NP mRNA levels and also caused the nuclear retention of NP, further strengthening our previous observations. Taken together, the results of the present study indicate that actinin‐4, a novel interacting partner of IAV NP, plays a crucial role in viral replication and this interaction may participate in nuclear localization of NP and/or viral ribonucleoproteins. Structured digital abstract •http://www.uniprot.org/uniprot/P03466 http://www.ebi.ac.uk/ontology-lookup/?termId=MI:0915 with http://www.uniprot.org/uniprot/O43707 by http://www.ebi.ac.uk/ontology-lookup/?termId=MI:0006 (http://www.ebi.ac.uk/intact/interaction/EBI-9512541, http://www.ebi.ac.uk/intact/interaction/EBI-9512553)•http://www.uniprot.org/uniprot/Q8JR21 and http://www.uniprot.org/uniprot/O43707 http://www.ebi.ac.uk/ontology-lookup/?termId=MI:0403 by http://www.ebi.ac.uk/ontology-lookup/?termId=MI:0416 (http://www.ebi.ac.uk/intact/interaction/EBI-9514040)•http://www.uniprot.org/uniprot/Q91U50 http://www.ebi.ac.uk/ontology-lookup/?termId=MI:0915 with http://www.uniprot.org/uniprot/O43707 by http://www.ebi.ac.uk/ontology-lookup/?termId=MI:0006 (http://www.ebi.ac.uk/intact/interaction/EBI-9514006)•http://www.uniprot.org/uniprot/Q5L4H4 http://www.ebi.ac.uk/ontology-lookup/?termId=MI:0407 to http://www.uniprot.org/uniprot/O43707 by http://www.ebi.ac.uk/ontology-lookup/?termId=MI:0007 (http://www.ebi.ac.uk/intact/interaction/EBI-9512166, http://www.ebi.ac.uk/intact/interaction/EBI-9512219)•http://www.uniprot.org/uniprot/C3W6D7 http://www.ebi.ac.uk/ontology-lookup/?termId=MI:0915 with http://www.uniprot.org/uniprot/O43707 by http://www.ebi.ac.uk/ontology-lookup/?termId=MI:0006 (http://www.ebi.ac.uk/intact/interaction/EBI-9513951)•http://www.uniprot.org/uniprot/Q5L4H4 http://www.ebi.ac.uk/ontology-lookup/?termId=MI:0915 with http://www.uniprot.org/uniprot/O43707 by http://www.ebi.ac.uk/ontology-lookup/?termId=MI:0007 (http://www.ebi.ac.uk/intact/interaction/EBI-9512237)•http://www.uniprot.org/uniprot/Q6DPG0 http://www.ebi.ac.uk/ontology-lookup/?termId=MI:0915 with http://www.uniprot.org/uniprot/O43707 by http://www.ebi.ac.uk/ontology-lookup/?termId=MI:0006 (http://www.ebi.ac.uk/intact/interaction/EBI-9513984) •http://www.uniprot.org/uniprot/B2BU63 http://www.ebi.ac.uk/ontology-lookup/?termId=MI:0915 with http://www.uniprot.org/uniprot/O43707 by http://www.ebi.ac.uk/ontology-lookup/?termId=MI:0006 (http://www.ebi.ac.uk/intact/interaction/EBI-9513930) •http://www.uniprot.org/uniprot/Q5L4H4 http://www.ebi.ac.uk/ontology-lookup/?termId=MI:0915 with http://www.uniprot.org/uniprot/O43707 by http://www.ebi.ac.uk/ontology-lookup/?termId=MI:0018 (http://www.ebi.ac.uk/intact/interaction/EBI-9512145, http://www.ebi.ac.uk/intact/interaction/EBI-9512095) •http://www.uniprot.org/uniprot/C9S3S8 http://www.ebi.ac.uk/ontology-lookup/?termId=MI:0915 with http://www.uniprot.org/uniprot/O43707 by http://www.ebi.ac.uk/ontology-lookup/?termId=MI:0006 (http://www.ebi.ac.uk/intact/interaction/EBI-9513909)
Collapse
Affiliation(s)
- Shipra Sharma
- Virology Group, International Centre for Genetic Engineering & Biotechnology, New Delhi, India
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
52
|
Gerber M, Isel C, Moules V, Marquet R. Selective packaging of the influenza A genome and consequences for genetic reassortment. Trends Microbiol 2014; 22:446-55. [PMID: 24798745 DOI: 10.1016/j.tim.2014.04.001] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 03/25/2014] [Accepted: 04/02/2014] [Indexed: 10/25/2022]
Abstract
Influenza A viruses package their segmented RNA genome in a selective manner. Electron tomography, biochemical assays, and replication assays of viruses produced by reverse genetics recently unveiled molecular details of this mechanism, whereby different influenza viral strains form different and unique networks of direct intermolecular RNA-RNA interactions. Together with detailed views of the three-dimensional structure of the viral ribonucleoparticles, these recent advances help us understand the rules that govern genome packaging. They also have deep implications for the genetic reassortment processes, which are responsible for devastating pandemics.
Collapse
Affiliation(s)
- Marie Gerber
- Architecture et Réactivité de l'ARN, Université de Strasbourg, Centre National de Recherche Scientifique (CNRS), Institut de Biologie Moléculaire et Cellulaire (IBMC), 15 rue René Descartes, 67084 Strasbourg, France
| | - Catherine Isel
- Architecture et Réactivité de l'ARN, Université de Strasbourg, Centre National de Recherche Scientifique (CNRS), Institut de Biologie Moléculaire et Cellulaire (IBMC), 15 rue René Descartes, 67084 Strasbourg, France
| | - Vincent Moules
- Virologie et Pathologie Humaine, Université Lyon 1, EA4610, Faculté de Médecine RTH Laennec, 69008 Lyon, France; VirNext, Université Lyon 1, EA4610, Faculté de Médecine RTH Laennec, 69008 Lyon, France
| | - Roland Marquet
- Architecture et Réactivité de l'ARN, Université de Strasbourg, Centre National de Recherche Scientifique (CNRS), Institut de Biologie Moléculaire et Cellulaire (IBMC), 15 rue René Descartes, 67084 Strasbourg, France.
| |
Collapse
|
53
|
Isakova-Sivak I, Chen LM, Bourgeois M, Matsuoka Y, Voeten JTM, Heldens JGM, van den Bosch H, Klimov A, Rudenko L, Cox NJ, Donis RO. Characterization of reverse genetics-derived cold-adapted master donor virus A/Leningrad/134/17/57 (H2N2) and reassortants with H5N1 surface genes in a mouse model. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2014; 21:722-31. [PMID: 24648485 PMCID: PMC4018889 DOI: 10.1128/cvi.00819-13] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2013] [Accepted: 03/11/2014] [Indexed: 01/28/2023]
Abstract
Live attenuated influenza vaccines (LAIV) offer significant advantages over subunit or split inactivated vaccines to mitigate an eventual influenza pandemic, including simpler manufacturing processes and more cross-protective immune responses. Using an established reverse genetics (rg) system for wild-type (wt) A/Leningrad/134/1957 and cold-adapted (ca) A/Leningrad/134/17/1957 (Len17) master donor virus (MDV), we produced and characterized three rg H5N1 reassortant viruses carrying modified HA and intact NA genes from either A/Vietnam/1203/2004 (H5N1, VN1203, clade 1) or A/Egypt/321/2007 (H5N1, EG321, clade 2) virus. A mouse model of infection was used to determine the infectivity and tissue tropism of the parental wt viruses compared to the ca master donor viruses as well as the H5N1 reassortants. All ca viruses showed reduced replication in lungs and enhanced replication in nasal epithelium. In addition, the H5N1 HA and NA enhanced replication in lungs unless it was restricted by the internal genes of the ca MDV. Mice inoculated twice 4 weeks apart with the H5N1 reassortant LAIV candidate viruses developed serum hemagglutination inhibition HI and IgA antibody titers to the homologous and heterologous viruses consistent with protective immunity. These animals remained healthy after challenge inoculation with a lethal dose with homologous or heterologous wt H5N1 highly pathogenic avian influenza (HPAI) viruses. The profiles of viral replication in respiratory tissues and the immunogenicity and protective efficacy characteristics of the two ca H5N1 candidate LAIV viruses warrant further development into a vaccine for human use.
Collapse
MESH Headings
- Animal Structures/virology
- Animals
- Antibodies, Viral/blood
- Disease Models, Animal
- Female
- Hemagglutinin Glycoproteins, Influenza Virus/genetics
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Immunoglobulin A/blood
- Influenza A Virus, H2N2 Subtype/genetics
- Influenza A Virus, H2N2 Subtype/immunology
- Influenza A Virus, H5N1 Subtype/genetics
- Influenza A Virus, H5N1 Subtype/immunology
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/genetics
- Influenza Vaccines/immunology
- Mice, Inbred BALB C
- Neuraminidase/genetics
- Neuraminidase/immunology
- Orthomyxoviridae Infections/prevention & control
- Reassortant Viruses/genetics
- Reassortant Viruses/immunology
- Reverse Genetics
- Survival Analysis
- Vaccination/methods
- Vaccines, Attenuated/administration & dosage
- Vaccines, Attenuated/genetics
- Vaccines, Attenuated/immunology
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
- Viral Proteins/genetics
- Viral Proteins/immunology
- Virulence
Collapse
Affiliation(s)
- Irina Isakova-Sivak
- Influenza Division, Centers for Disease Control and Prevention, Department of Health and Human Services, Atlanta, Georgia, USA
- Institute of Experimental Medicine, Russian Academy of Medical Sciences, St. Petersburg, Russia
| | - Li-Mei Chen
- Influenza Division, Centers for Disease Control and Prevention, Department of Health and Human Services, Atlanta, Georgia, USA
| | - Melissa Bourgeois
- Influenza Division, Centers for Disease Control and Prevention, Department of Health and Human Services, Atlanta, Georgia, USA
| | - Yumiko Matsuoka
- Influenza Division, Centers for Disease Control and Prevention, Department of Health and Human Services, Atlanta, Georgia, USA
| | | | | | | | - Alexander Klimov
- Influenza Division, Centers for Disease Control and Prevention, Department of Health and Human Services, Atlanta, Georgia, USA
| | - Larisa Rudenko
- Institute of Experimental Medicine, Russian Academy of Medical Sciences, St. Petersburg, Russia
| | - Nancy J. Cox
- Influenza Division, Centers for Disease Control and Prevention, Department of Health and Human Services, Atlanta, Georgia, USA
| | - Ruben O. Donis
- Influenza Division, Centers for Disease Control and Prevention, Department of Health and Human Services, Atlanta, Georgia, USA
| |
Collapse
|
54
|
Deyde VM, Sampath R, Gubareva LV. RT-PCR/electrospray ionization mass spectrometry approach in detection and characterization of influenza viruses. Expert Rev Mol Diagn 2014; 11:41-52. [DOI: 10.1586/erm.10.107] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
55
|
Neverov AD, Lezhnina KV, Kondrashov AS, Bazykin GA. Intrasubtype reassortments cause adaptive amino acid replacements in H3N2 influenza genes. PLoS Genet 2014; 10:e1004037. [PMID: 24415946 PMCID: PMC3886890 DOI: 10.1371/journal.pgen.1004037] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Accepted: 11/01/2013] [Indexed: 01/15/2023] Open
Abstract
Reassortments and point mutations are two major contributors to diversity of Influenza A virus; however, the link between these two processes is unclear. It has been suggested that reassortments provoke a temporary increase in the rate of amino acid changes as the viral proteins adapt to new genetic environment, but this phenomenon has not been studied systematically. Here, we use a phylogenetic approach to infer the reassortment events between the 8 segments of influenza A H3N2 virus since its emergence in humans in 1968. We then study the amino acid replacements that occurred in genes encoded in each segment subsequent to reassortments. In five out of eight genes (NA, M1, HA, PB1 and NS1), the reassortment events led to a transient increase in the rate of amino acid replacements on the descendant phylogenetic branches. In NA and HA, the replacements following reassortments were enriched with parallel and/or reversing replacements; in contrast, the replacements at sites responsible for differences between antigenic clusters (in HA) and at sites under positive selection (in NA) were underrepresented among them. Post-reassortment adaptive walks contribute to adaptive evolution in Influenza A: in NA, an average reassortment event causes at least 2.1 amino acid replacements in a reassorted gene, with, on average, 0.43 amino acid replacements per evolving post-reassortment lineage; and at least ~9% of all amino acid replacements are provoked by reassortments.
Collapse
Affiliation(s)
- Alexey D. Neverov
- Federal Budget Institution of Science “Central Research Institute for Epidemiology”, Moscow, Russia
- Department of Bioengineering and Bioinformatics, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Ksenia V. Lezhnina
- Department of Bioengineering and Bioinformatics, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Alexey S. Kondrashov
- Department of Bioengineering and Bioinformatics, M.V. Lomonosov Moscow State University, Moscow, Russia
- Life Sciences Institute and Department of Ecology and Evolutionary Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Georgii A. Bazykin
- Department of Bioengineering and Bioinformatics, M.V. Lomonosov Moscow State University, Moscow, Russia
- Institute for Information Transmission Problems of the Russian Academy of Sciences (Kharkevich Institute), Moscow, Russia
- * E-mail:
| |
Collapse
|
56
|
Abstract
Reassortment is the process by which influenza viruses swap gene segments. This genetic exchange is possible due to the segmented nature of the viral genome and occurs when two differing influenza viruses co-infect a cell. The viral diversity generated through reassortment is vast and plays an important role in the evolution of influenza viruses. Herein we review recent insights into the contribution of reassortment to the natural history and epidemiology of influenza A viruses, gained through population scale phylogenic analyses. We describe methods currently used to study reassortment in the laboratory, and we summarize recent progress made using these experimental approaches to further our understanding of influenza virus reassortment and the contexts in which it occurs.
Collapse
Affiliation(s)
- John Steel
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | | |
Collapse
|
57
|
Li C, Chen H. Enhancement of influenza virus transmission by gene reassortment. Curr Top Microbiol Immunol 2014; 385:185-204. [PMID: 25048543 DOI: 10.1007/82_2014_389] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Influenza A virus is characterized by a genome composed of eight single-stranded, negative sense RNA segments, which allow for reassortment between different strains when they co-infect the same host cell. Reassortment is an important driving force for the evolution of influenza viruses. The ability of reassortment allows influenza virus to endlessly reinvent itself and pose a constant threat to the health of humans and other animals. Of the four human influenza pandemics since the beginning of the last century, three of them were caused by reassortant viruses bearing genes of avian, human or swine influenza virus origin. In the past decade, great efforts have been made to understand the transmissibility of influenza viruses. The use of reverse genetics technology has made it substantially easier to generate reassortant viruses and evaluate the contribution of individual virus gene on virus transmissibility in animal models such as ferrets and guinea pigs. H5, H7, and H9 avian influenza viruses represent the top three subtypes that are candidates to cause the next human influenza pandemic. Many studies have been conducted to determine whether the transmission of these avian influenza viruses could be enhanced by acquisition of gene segments from human influenza viruses. Moreover, the 2009 pdmH1N1 viruses and the triple reassortant swine influenza viruses were extensively studied to identify the gene segments that contribute to their transmissibility. These studies have greatly deepened our understanding of the transmissibility of reassortant influenza viruses, which, in turn, has improved our ability to be prepared for reassortant influenza virus with enhanced transmissibility and pandemic potential.
Collapse
Affiliation(s)
- Chengjun Li
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 427 Maduan Street, Harbin, 150001, Nangang, China,
| | | |
Collapse
|
58
|
Cong Y, Sun Y, Wang W, Meng Q, Ran W, Zhu L, Yang G, Yang W, Yang L, Wang C, Ding Z. Comparative analysis of receptor-binding specificity and pathogenicity in natural reassortant and non-reassortant H3N2 swine influenza virus. Vet Microbiol 2014; 168:105-15. [DOI: 10.1016/j.vetmic.2013.11.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Revised: 11/01/2013] [Accepted: 11/04/2013] [Indexed: 10/26/2022]
|
59
|
Abstract
Influenza has been recognized as a respiratory disease in swine since its first appearance concurrent with the 1918 "Spanish flu" human pandemic. All influenza viruses of significance in swine are type A, subtype H1N1, H1N2, or H3N2 viruses. Influenza viruses infect epithelial cells lining the surface of the respiratory tract, inducing prominent necrotizing bronchitis and bronchiolitis and variable interstitial pneumonia. Cell death is due to direct virus infection and to insult directed by leukocytes and cytokines of the innate immune system. The most virulent viruses consistently express the following characteristics of infection: (1) higher or more prolonged virus replication, (2) excessive cytokine induction, and (3) replication in the lower respiratory tract. Nearly all the viral proteins contribute to virulence. Pigs are susceptible to infection with both human and avian viruses, which often results in gene reassortment between these viruses and endemic swine viruses. The receptors on the epithelial cells lining the respiratory tract are major determinants of infection by influenza viruses from other hosts. The polymerases, especially PB2, also influence cross-species infection. Methods of diagnosis and characterization of influenza viruses that infect swine have improved over the years, driven both by the availability of new technologies and by the necessity of keeping up with changes in the virus. Testing of oral fluids from pigs for virus and antibody is a recent development that allows efficient sampling of large numbers of animals.
Collapse
Affiliation(s)
- B H Janke
- DVM, PhD, Veterinary Diagnostic Laboratory, Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA.
| |
Collapse
|
60
|
DBA/2 mouse as an animal model for anti-influenza drug efficacy evaluation. J Microbiol 2013; 51:866-71. [PMID: 24385366 DOI: 10.1007/s12275-013-3428-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Accepted: 09/03/2013] [Indexed: 01/01/2023]
Abstract
Influenza viruses are seasonally recurring human pathogens. Vaccines and antiviral drugs are available for influenza. However, the viruses, which often change themselves via antigenic drift and shift, demand constant efforts to update vaccine antigens every year and develop new agents with broad-spectrum antiviral efficacy. An animal model is critical for such efforts. While most human influenza viruses are unable to kill BALB/c mice, some strains have been shown to kill DBA/2 mice without prior adaptation. Therefore, in this study, we explored the feasibility of employing DBA/2 mice as a model in the development of anti-influenza drugs. Unlike the BALB/c strain, DBA/2 mice were highly susceptible and could be killed with a relatively low titer (50% DBA/2 lethal dose = 10(2.83) plaque-forming units) of the A/Korea/01/2009 virus (2009 pandemic H1N1 virus). When treated with a neuraminidase inhibitor, oseltamivir phosphate, infected DBA/2 mice survived until 14 days post-infection. The reduced morbidity of the infected DBA/2 mice was also consistent with the oseltamivir treatment. Taking these data into consideration, we propose that the DBA/2 mouse is an excellent animal model to evaluate antiviral efficacy against influenza infection and can be further utilized for combination therapies or bioactivity models of existing and newly developed anti-influenza drugs.
Collapse
|
61
|
Kreibich A, Stech O, Hundt J, Ziller M, Mettenleiter TC, Stech J. Avian influenza virus h3 hemagglutinin may enable high fitness of novel human virus reassortants. PLoS One 2013; 8:e79165. [PMID: 24265752 PMCID: PMC3827155 DOI: 10.1371/journal.pone.0079165] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Accepted: 09/20/2013] [Indexed: 11/18/2022] Open
Abstract
Reassortment of influenza A virus genes enables antigenic shift resulting in the emergence of pandemic viruses with novel hemagglutinins (HA) acquired from avian strains. Here, we investigated whether historic and contemporary avian strains with different replication capacity in human cells can donate their hemagglutinin to a pandemic human virus. We performed double-infections with two avian H3 strains as HA donors and a human acceptor strain, and determined gene compositions and replication of HA reassortants in mammalian cells. To enforce selection for the avian virus HA, we generated a strictly elastase-dependent HA cleavage site mutant from A/Hong Kong/1/68 (H3N2) (Hk68-Ela). This mutant was used for co-infections of human cells with A/Duck/Ukraine/1/63 (H3N8) (DkUkr63) or the more recent A/Mallard/Germany/Wv64-67/05 (H3N2) (MallGer05) in the absence of elastase but presence of trypsin. Among 21 plaques analyzed from each assay, we found 12 HA reassortants with DkUkr63 (4 genotypes) and 14 with MallGer05 (10 genotypes) that replicated in human cells comparable to the parental human virus. Although DkUkr63 replicated in mammalian cells at a reduced level compared to MallGer05 and Hk68, it transmitted its HA to the human virus, indicating that lower replication efficiency of an avian virus in a mammalian host may not constrain the emergence of viable HA reassortants. The finding that HA and HA/NA reassortants replicated efficiently like the human virus suggests that further HA adaptation remains a relevant barrier for emergence of novel HA reassortants.
Collapse
Affiliation(s)
- Anne Kreibich
- Friedrich-Loeffler-Institut, Institute of Molecular Biology, Greifswald-Insel Riems, Germany
| | - Olga Stech
- Friedrich-Loeffler-Institut, Institute of Molecular Biology, Greifswald-Insel Riems, Germany
| | - Jana Hundt
- Friedrich-Loeffler-Institut, Institute of Molecular Biology, Greifswald-Insel Riems, Germany
| | - Mario Ziller
- Friedrich-Loeffler-Institut, Biomathematics Working Group, Greifswald-Insel Riems, Germany
| | - Thomas C. Mettenleiter
- Friedrich-Loeffler-Institut, Institute of Molecular Biology, Greifswald-Insel Riems, Germany
| | - Juergen Stech
- Friedrich-Loeffler-Institut, Institute of Molecular Biology, Greifswald-Insel Riems, Germany
- * E-mail:
| |
Collapse
|
62
|
Tong S, Zhu X, Li Y, Shi M, Zhang J, Bourgeois M, Yang H, Chen X, Recuenco S, Gomez J, Chen LM, Johnson A, Tao Y, Dreyfus C, Yu W, McBride R, Carney PJ, Gilbert AT, Chang J, Guo Z, Davis CT, Paulson JC, Stevens J, Rupprecht CE, Holmes EC, Wilson IA, Donis RO. New world bats harbor diverse influenza A viruses. PLoS Pathog 2013; 9:e1003657. [PMID: 24130481 PMCID: PMC3794996 DOI: 10.1371/journal.ppat.1003657] [Citation(s) in RCA: 901] [Impact Index Per Article: 81.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2013] [Accepted: 08/09/2013] [Indexed: 12/30/2022] Open
Abstract
Aquatic birds harbor diverse influenza A viruses and are a major viral reservoir in nature. The recent discovery of influenza viruses of a new H17N10 subtype in Central American fruit bats suggests that other New World species may similarly carry divergent influenza viruses. Using consensus degenerate RT-PCR, we identified a novel influenza A virus, designated as H18N11, in a flat-faced fruit bat (Artibeus planirostris) from Peru. Serologic studies with the recombinant H18 protein indicated that several Peruvian bat species were infected by this virus. Phylogenetic analyses demonstrate that, in some gene segments, New World bats harbor more influenza virus genetic diversity than all other mammalian and avian species combined, indicative of a long-standing host-virus association. Structural and functional analyses of the hemagglutinin and neuraminidase indicate that sialic acid is not a ligand for virus attachment nor a substrate for release, suggesting a unique mode of influenza A virus attachment and activation of membrane fusion for entry into host cells. Taken together, these findings indicate that bats constitute a potentially important and likely ancient reservoir for a diverse pool of influenza viruses. Previous studies indicated that a novel influenza A virus (H17N10) was circulating in fruit bats from Guatemala (Central America). Herein, we investigated whether similar viruses are present in bat species from South America. Analysis of rectal swabs from bats sampled in the Amazon rainforest region of Peru identified another new influenza A virus from bats that is phylogenetically distinct from the one identified in Guatemala. The genes that encode the surface proteins of the new virus from the flat-faced fruit bat were designated as new subtype H18N11. Serologic testing of blood samples from several species of Peruvian bats indicated a high prevalence of antibodies to the surface proteins. Phylogenetic analyses demonstrate that bat populations from Central and South America maintain as much influenza virus genetic diversity in some gene segments as all other mammalian and avian species combined. The crystal structures of the hemagglutinin and neuraminidase proteins indicate that sialic acid is not a receptor for virus attachment nor a substrate for release, suggesting a novel mechanism of influenza A virus attachment and activation of membrane fusion for entry into host cells. In summary, our findings indicate that bats constitute a potentially important reservoir for influenza viruses.
Collapse
Affiliation(s)
- Suxiang Tong
- Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
- * E-mail: (ST); (JS); (CER); (ECH); (IAW); (ROD)
| | - Xueyong Zhu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Yan Li
- Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Mang Shi
- Sydney Emerging Infections and Biosecurity Institute, School of Biological Sciences and Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Jing Zhang
- Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Melissa Bourgeois
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Hua Yang
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Xianfeng Chen
- Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Sergio Recuenco
- Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Jorge Gomez
- Direccion General de Epidemiologıa, Ministerio de Salud - MINSA, Lima, Peru
| | - Li-Mei Chen
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Adam Johnson
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Ying Tao
- Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Cyrille Dreyfus
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Wenli Yu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Ryan McBride
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Paul J. Carney
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Amy T. Gilbert
- Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Jessie Chang
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Zhu Guo
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Charles T. Davis
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - James C. Paulson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California, United States of America
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California, United States of America
| | - James Stevens
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
- * E-mail: (ST); (JS); (CER); (ECH); (IAW); (ROD)
| | - Charles E. Rupprecht
- Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
- Global Alliance for Rabies Control, The Scripps Research Institute, La Jolla, California, United States of America
- * E-mail: (ST); (JS); (CER); (ECH); (IAW); (ROD)
| | - Edward C. Holmes
- Sydney Emerging Infections and Biosecurity Institute, School of Biological Sciences and Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
- Fogarty International Center, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail: (ST); (JS); (CER); (ECH); (IAW); (ROD)
| | - Ian A. Wilson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California, United States of America
- Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, California, United States of America
- * E-mail: (ST); (JS); (CER); (ECH); (IAW); (ROD)
| | - Ruben O. Donis
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
- * E-mail: (ST); (JS); (CER); (ECH); (IAW); (ROD)
| |
Collapse
|
63
|
Abdelwhab EM, Veits J, Mettenleiter TC. Avian influenza virus NS1: A small protein with diverse and versatile functions. Virulence 2013; 4:583-8. [PMID: 24051601 PMCID: PMC3906290 DOI: 10.4161/viru.26360] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Affiliation(s)
- E M Abdelwhab
- Friedrich-Loeffler-Institut; Federal Research Institute for Animal Health; Institute of Molecular Biology; Insel Riems, Germany
| | | | | |
Collapse
|
64
|
Hara K, Nakazono Y, Kashiwagi T, Hamada N, Watanabe H. Co-incorporation of the PB2 and PA polymerase subunits from human H3N2 influenza virus is a critical determinant of the replication of reassortant ribonucleoprotein complexes. J Gen Virol 2013; 94:2406-2416. [PMID: 23939981 DOI: 10.1099/vir.0.053959-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The influenza virus RNA polymerase, composed of the PB1, PB2 and PA subunits, has a potential role in influencing genetic reassortment. Recent studies on the reassortment of human H3N2 strains suggest that the co-incorporation of PB2 and PA from the same H3N2 strain appears to be important for efficient virus replication; however, the underlying mechanism remains unclear. Here, we reconstituted reassortant ribonucleoprotein (RNP) complexes and demonstrated that the RNP activity was severely impaired when the PA subunit of H3N2 strain A/NT/60/1968 (NT PA) was introduced into H1N1 or H5N1 polymerase. The NT PA did not affect the correct assembly of the polymerase trimeric complex, but it significantly reduced replication-initiation activity when provided with a vRNA promoter and severely impaired the accumulation of RNP, which led to the loss of RNP activity. Mutational analysis demonstrated that PA residues 184N and 383N were the major determinants of the inhibitory effect of NT PA and 184N/383N sequences were unique to human H3N2 strains. Significantly, NT PB2 specifically relieved the inhibitory effect of NT PA, and the PB2 residue 627K played a key role. Our results suggest that PB2 from the same H3N2 strain might be required for overcoming the inhibitory effect of H3N2 PA in the genetic reassortment of influenza virus.
Collapse
Affiliation(s)
- Koyu Hara
- Division of Infectious Diseases, Department of Infectious Medicine, Kurume University School of Medicine, Fukuoka, Japan
| | - Yoko Nakazono
- Division of Infectious Diseases, Department of Infectious Medicine, Kurume University School of Medicine, Fukuoka, Japan
| | - Takahito Kashiwagi
- Division of Infectious Diseases, Department of Infectious Medicine, Kurume University School of Medicine, Fukuoka, Japan
| | - Nobuyuki Hamada
- Division of Infectious Diseases, Department of Infectious Medicine, Kurume University School of Medicine, Fukuoka, Japan
| | - Hiroshi Watanabe
- Division of Infectious Diseases, Department of Infectious Medicine, Kurume University School of Medicine, Fukuoka, Japan
| |
Collapse
|
65
|
Huang CH, Chen CJ, Yen CT, Yu CP, Huang PN, Kuo RL, Lin SJ, Chang CK, Shih SR. Caspase-1 deficient mice are more susceptible to influenza A virus infection with PA variation. J Infect Dis 2013; 208:1898-905. [PMID: 23901080 DOI: 10.1093/infdis/jit381] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Reassortment within polymerase genes causes changes in the pathogenicity of influenza A viruses. We previously reported that the 2009 pH1N1 PA enhanced the pathogenicity of seasonal H1N1. We examined the effects of the PA gene from the HPAI H5N1 following its introduction into currently circulating seasonal influenza viruses. METHODS To evaluate the role of H5N1 PA in altering the virulence of seasonal influenza viruses, we generated a recombinant seasonal H3N2 (3446) that expressed the H5N1 PA protein (VPA) and evaluated the RNP activity, growth kinetics, and pathogenicity of the reassortant virus in mice. RESULTS Compared with the wild-type 3446 virus, the substitution of the H5N1 PA gene into the 3446 virus (VPA/3446) resulted in increased RNP activity and an increased replication rate in A549 cells. The recombinant VPA/3446 virus also caused more severe pneumonia in Casp 1(-/-) mice than in IL1β(-/-) and wild-type B6 mice. CONCLUSIONS Although the PA from H5N1 is incidentally compatible with a seasonal H3N2 backbone, the H5N1 PA affected the virulence of seasonal H3N2, particularly in inflammasome-related innate immunity deficient mice. These findings highlight the importance of monitoring PA reassortment in seasonal flu, and confirm the role of the Caspase-1 gene in influenza pathogenesis.
Collapse
|
66
|
He L, Zhao G, Zhong L, Liu Q, Duan Z, Gu M, Wang X, Liu X, Liu X. Isolation and characterization of two H5N1 influenza viruses from swine in Jiangsu Province of China. Arch Virol 2013; 158:2531-41. [PMID: 23836394 DOI: 10.1007/s00705-013-1771-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Accepted: 05/11/2013] [Indexed: 12/01/2022]
Abstract
Pigs are susceptible to infection with both human and avian influenza A viruses and are considered intermediate hosts that facilitate virus reassortment. Although H5N1 virus has spread to a wide range of avian and mammalian species, data about swine H5N1 isolates are scarce. To determine whether Asian H5N1 influenza viruses had been transmitted to pigs, a total of 1,107 nasal swab samples from healthy swine were collected from 2008 to 2009 in Jiangsu province of eastern China. In this survey, two H5N1 viruses A/swine/Jiangsu/1/2008 (JS/08) and A/swine/Jiangsu/2/2009 (JS/09) were isolated and identified. Phylogenetic analysis showed that JS/08 and JS/09 belonged to clade 7 and clade 2.3.4, respectively, and shared over 99.0 % sequence identity with poultry H5N1 isolates of the same clade in China. Receptor specificity analysis also showed that both of the swine H5N1 isolates bound preferentially to avian-type receptors. However, experiments in mammals indicated that JS/09 was moderately pathogenic to mice without prior adaption, whereas JS/08 had limited ability to replicate. Our findings suggest that pigs are naturally infected with avian H5N1 virus and highlight the potential threat to public health due to adaption or reassortment of H5N1 virus in this species.
Collapse
Affiliation(s)
- Liang He
- College of Veterinary Medicine, Yangzhou University, 12 East Wenhui Road, Yangzhou, Jiangsu, 225009, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
67
|
Ngai KLK, Chan MCW, Chan PKS. Replication and transcription activities of ribonucleoprotein complexes reconstituted from avian H5N1, H1N1pdm09 and H3N2 influenza A viruses. PLoS One 2013; 8:e65038. [PMID: 23750226 PMCID: PMC3672204 DOI: 10.1371/journal.pone.0065038] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2013] [Accepted: 04/22/2013] [Indexed: 01/14/2023] Open
Abstract
Avian influenza viruses pose a serious pandemic threat to humans. Better knowledge on cross-species adaptation is important. This study examined the replication and transcription efficiency of ribonucleoprotein complexes reconstituted by plasmid co-transfection between H5N1, H1N1pdm09 and H3N2 influenza A viruses, and to identify mutations in the RNA polymerase subunit that affect human adaptation. Viral RNA polymerase subunits PB1, PB2, PA and NP derived from influenza viruses were co-expressed with pPolI-vNP-Luc in human cells, and with its function evaluated by luciferase reporter assay. A quantitative RT-PCR was used to measure vRNA, cRNA, and mRNA levels for assessing the replication and transcription efficiency. Mutations in polymerase subunit were created to identify signature of increased human adaptability. H5N1 ribonucleoprotein complexes incorporated with PB2 derived from H1N1pdm09 and H3N2 viruses increased the polymerase activity in human cells. Furthermore, single amino acid substitutions at PB2 of H5N1 could affect polymerase activity in a temperature-dependent manner. By using a highly sensitive quantitative reverse transcription-polymerase chain reaction, an obvious enhancement in replication and transcription activities of ribonucleoproteins was observed by the introduction of lysine at residue 627 in the H5N1 PB2 subunit. Although less strongly in polymerase activity, E158G mutation appeared to alter the accumulation of H5N1 RNA levels in a temperature-dependent manner, suggesting a temperature-dependent mechanism in regulating transcription and replication exists. H5N1 viruses can adapt to humans either by acquisition of PB2 from circulating human-adapted viruses through reassortment, or by mutations at critical sites in PB2. This information may help to predict the pandemic potential of newly emerged influenza strains, and provide a scientific basis for stepping up surveillance measures and vaccine production.
Collapse
MESH Headings
- Animals
- HEK293 Cells
- Humans
- Influenza A Virus, H1N1 Subtype/enzymology
- Influenza A Virus, H1N1 Subtype/genetics
- Influenza A Virus, H1N1 Subtype/physiology
- Influenza A Virus, H3N2 Subtype/enzymology
- Influenza A Virus, H3N2 Subtype/genetics
- Influenza A Virus, H3N2 Subtype/physiology
- Influenza A Virus, H5N1 Subtype/enzymology
- Influenza A Virus, H5N1 Subtype/genetics
- Influenza A Virus, H5N1 Subtype/physiology
- Influenza A virus/enzymology
- Influenza A virus/genetics
- Influenza A virus/physiology
- Mutation
- Pandemics
- RNA-Dependent RNA Polymerase/genetics
- Ribonucleoproteins/genetics
- Temperature
- Transcription, Genetic
- Viral Proteins/genetics
- Virus Replication
Collapse
Affiliation(s)
- Karry L. K. Ngai
- Department of Microbiology, Faculty of Medicine, The Chinese University of Hong Kong, New Territories, Hong Kong Special Administration Region, People’s Republic of China
| | - Martin C. W. Chan
- Department of Microbiology, Faculty of Medicine, The Chinese University of Hong Kong, New Territories, Hong Kong Special Administration Region, People’s Republic of China
| | - Paul K. S. Chan
- Department of Microbiology and Stanley Ho Centre for Emerging Infectious Diseases, Faculty of Medicine, The Chinese University of Hong Kong, New Territories, Hong Kong Special Administration Region, People’s Republic of China
- * E-mail:
| |
Collapse
|
68
|
Zhang Y, Zhang Q, Kong H, Jiang Y, Gao Y, Deng G, Shi J, Tian G, Liu L, Liu J, Guan Y, Bu Z, Chen H. H5N1 hybrid viruses bearing 2009/H1N1 virus genes transmit in guinea pigs by respiratory droplet. Science 2013; 340:1459-63. [PMID: 23641061 DOI: 10.1126/science.1229455] [Citation(s) in RCA: 177] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
In the past, avian influenza viruses have crossed species barriers to trigger human pandemics by reassorting with mammal-infective viruses in intermediate livestock hosts. H5N1 viruses are able to infect pigs, and some of them have affinity for the mammalian type α-2,6-linked sialic acid airway receptor. Using reverse genetics, we systematically created 127 reassortant viruses between a duck isolate of H5N1, specifically retaining its hemagglutinin (HA) gene throughout, and a highly transmissible, human-infective H1N1 virus. We tested the virulence of the reassortants in mice as a correlate for virulence in humans and tested transmissibility in guinea pigs, which have both avian and mammalian types of airway receptor. Transmission studies showed that the H1N1 virus genes encoding acidic polymerase and nonstructural protein made the H5N1 virus transmissible by respiratory droplet between guinea pigs without killing them. Further experiments implicated other H1N1 genes in the enhancement of mammal-to-mammal transmission, including those that encode nucleoprotein, neuraminidase, and matrix, as well as mutations in H5 HA that improve affinity for humanlike airway receptors. Hence, avian H5N1 subtype viruses do have the potential to acquire mammalian transmissibility by reassortment in current agricultural scenarios.
Collapse
Affiliation(s)
- Ying Zhang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
69
|
Reassortment between Avian H5N1 and human influenza viruses is mainly restricted to the matrix and neuraminidase gene segments. PLoS One 2013; 8:e59889. [PMID: 23527283 PMCID: PMC3604002 DOI: 10.1371/journal.pone.0059889] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Accepted: 02/19/2013] [Indexed: 12/20/2022] Open
Abstract
Highly pathogenic avian influenza H5N1 viruses have devastated the poultry industry in many countries of the eastern hemisphere. Occasionally H5N1 viruses cross the species barrier and infect humans, sometimes with a severe clinical outcome. When this happens, there is a chance of reassortment between H5N1 and human influenza viruses. To assess the potential of H5N1 viruses to reassort with contemporary human influenza viruses (H1N1, H3N2 and pandemic H1N1), we used an in vitro selection method to generate reassortant viruses, that contained the H5 hemagglutinin gene, and that have a replication advantage in vitro. We found that the neuraminidase and matrix gene segments of human influenza viruses were preferentially selected by H5 viruses. However, these H5 reassortant viruses did not show a marked increase in replication in MDCK cells and human bronchial epithelial cells. In ferrets, inoculation with a mixture of H5N1-pandemic H1N1 reassortant viruses resulted in outgrowth of reassortant H5 viruses that had incorporated the neuraminidase and matrix gene segment of pandemic 2009 H1N1. This virus was not transmitted via aerosols or respiratory droplets to naïve recipient ferrets. Altogether, these data emphasize the potential of avian H5N1 viruses to reassort with contemporary human influenza viruses. The neuraminidase and matrix gene segments of human influenza viruses showed the highest genetic compatibility with HPAI H5N1 virus.
Collapse
|
70
|
Belser JA, Tumpey TM. H5N1 pathogenesis studies in mammalian models. Virus Res 2013; 178:168-85. [PMID: 23458998 DOI: 10.1016/j.virusres.2013.02.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Revised: 12/14/2012] [Accepted: 02/06/2013] [Indexed: 12/21/2022]
Abstract
H5N1 influenza viruses are capable of causing severe disease and death in humans, and represent a potential pandemic subtype should they acquire a transmissible phenotype. Due to the expanding host and geographic range of this virus subtype, there is an urgent need to better understand the contribution of both virus and host responses following H5N1 virus infection to prevent and control human disease. The use of mammalian models, notably the mouse and ferret, has enabled the detailed study of both complex virus-host interactions as well as the contribution of individual viral proteins and point mutations which influence virulence. In this review, we describe the behavior of H5N1 viruses which exhibit high and low virulence in numerous mammalian species, and highlight the contribution of inoculation route to virus pathogenicity. The involvement of host responses as studied in both inbred and outbred mammalian models is discussed. The roles of individual viral gene products and molecular determinants which modulate the severity of H5N1 disease in vivo are presented. This research contributes not only to our understanding of influenza virus pathogenesis, but also identifies novel preventative and therapeutic targets to mitigate the disease burden caused by avian influenza viruses.
Collapse
Affiliation(s)
- Jessica A Belser
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30333, United States
| | | |
Collapse
|
71
|
Abstract
Although the ferret model has been extensively used to study pathogenesis and transmission of influenza viruses, little has been done to determine whether ferrets are a good surrogate animal model to study influenza virus reassortment. It has been previously shown that the pandemic 2009 H1N1 (H1N1pdm) virus was able to transmit efficiently in ferrets. In coinfection studies with either seasonal H1N1 or H3N2 strains (H1N1s or H3N2s, respectively), the H1N1pdm virus was able to outcompete these strains and become the dominant transmissible virus. However, lack of reassortment could have been the result of differences in the cell or tissue tropism of these viruses in the ferret. To address this issue, we performed coinfection studies with recombinant influenza viruses carrying the surface genes of a seasonal H3N2 strain in the background of an H1N1pdm strain and vice versa. After serial passages in ferrets, a dominant H1N2 virus population was obtained with a constellation of gene segments, most of which, except for the neuraminidase (NA) and PB1 segments, were from the H1N1pdm strain. Our studies suggest that ferrets recapitulate influenza virus reassortment events. The H1N2 virus generated through this process resembles similar viruses that are emerging in nature, particularly in pigs.
Collapse
|
72
|
Ngunjiri JM, Buchek GM, Mohni KN, Sekellick MJ, Marcus PI. Influenza virus subpopulations: exchange of lethal H5N1 virus NS for H1N1 virus NS triggers de novo generation of defective-interfering particles and enhances interferon-inducing particle efficiency. J Interferon Cytokine Res 2012; 33:99-107. [PMID: 23215782 DOI: 10.1089/jir.2012.0070] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Reassortment of influenza A viruses is known to affect viability, replication efficiency, antigenicity, host range, and virulence, and can generate pandemic strains. In this study, we demonstrated that the specific exchange of the NS gene segment from highly pathogenic A/HK/156/97 (H5N1) [E92 or E92D NS1] virus for the cognate NS gene segment of A/PR/834(H1N1) [D92 NS1] virus did not cause a significant change in the sizes of infectious particle subpopulations. However, it resulted in 2 new phenotypic changes: (1) de novo generation of large subpopulations of defective-interfering particles (DIPs); and (2) enhancement of interferon (IFN)-inducing particle efficiency leading to an order of magnitude or higher quantum (peak) yield of IFN in both avian and mammalian cells. These changes were attributed to loss of function of the H5N1-NS gene products. Most notably, the NS exchange obliterated the usual IFN-induction-suppressing capacity associated with expression of full-size NS1 proteins, and hence functionally mimicked deletions in the NS1 gene. The loss of NS1-mediated suppression of IFN induction, de novo generation of DIPs, and the concomitant enhancement of IFN-inducing particle efficiency suggest that in an attenuated background, the H5N1-NS could be used to formulate a self-adjuvanting live attenuated influenza vaccine similar to viruses with deletions in the NS1 gene.
Collapse
Affiliation(s)
- John M Ngunjiri
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, CT 06269, USA
| | | | | | | | | |
Collapse
|
73
|
Co-circulation of influenza A virus strains and emergence of pandemic via reassortment: the role of cross-immunity. Epidemics 2012; 5:20-33. [PMID: 23438428 DOI: 10.1016/j.epidem.2012.10.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Revised: 10/09/2012] [Accepted: 10/29/2012] [Indexed: 12/24/2022] Open
Abstract
Reassortment is an important evolutionary route for influenza A viruses to generate pandemic strains. The pre-requisite for reassortment to occur is co-infection of different influenza virus strains in the same host population. Empirical evidence suggests that co-circulation of influenza A virus strains is common and co-infection in patients has been reported. Whether a novel virus can successfully spread among a host population is determined by its life-history (infectivity and infectious period). It is also well known that different influenza A strains interfere through the immune response of human body cells. The reassortant virus strain generated from co-infections deviates dramatically in antigenic and genetic properties from its parental strains such that human populations have limited immunity against it. We consider a mathematical model which includes two strains of influenza virus within a standard SIR model and integrate life history and cross-immunity into the evolutionary dynamics of influenza virus. We assume that, following primary infection by one strain and recovery, individuals are susceptible to secondary infection by the other strain only but with reduced probability due to cross-immunity. Co-infection is included to examine how life-history and cross-immunity interplay to regulate the co-circulation and co-infection of different influenza A virus strains in human populations. Further, we introduce novel strains via reassortment and investigate how the opportunities of a reassortant strain developing into a pandemic are constrained by its life-history and the residual immunity within human populations. We find that though the probability of pandemic emergence via reassortment increases with transmissibility of reassortant strains and the rate of reassortment, the existence of cross-immunity acquired through previous infections or vaccination can greatly constrain pandemic emergence.
Collapse
|
74
|
Fournier E, Moules V, Essere B, Paillart JC, Sirbat JD, Cavalier A, Rolland JP, Thomas D, Lina B, Isel C, Marquet R. Interaction network linking the human H3N2 influenza A virus genomic RNA segments. Vaccine 2012; 30:7359-67. [PMID: 23063835 DOI: 10.1016/j.vaccine.2012.09.079] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2011] [Revised: 04/24/2012] [Accepted: 09/28/2012] [Indexed: 12/14/2022]
Abstract
The genome of influenza A viruses is comprised of eight negative-sense viral RNAs (vRNAs) that form viral ribonucleoproteins (vRNPs). In order to be infectious, an influenza A viral particle must encapsidate at least one copy of each of the vRNAs. Thus, even though genome segmentation is evolutionary advantageous, it undeniably complicates viral assembly, which is believed to occur through a selective mechanism that still remains to be understood. Using electron tomography 3D-reconstructions, we show that the eight vRNPs of an influenza A Moscow/10/99 (H3N2) virus are interconnected within a star-like structure as they emerge from a unique "transition zone" at the budding tip of the virions. Notably, this "transition zone" is thick enough to accommodate all described packaging signals. We also report that, in vitro, each vRNA segment is involved in a direct contact with at least one other vRNA partner, in a single network of intermolecular interactions. We show that in several cases, the regions involved in vRNA/vRNA interactions overlap with previously identified packaging signals. Our results thus provide support for the involvement of RNA/RNA interactions in the selection and specific packaging of influenza A genomic RNAs, which appear embedded into an organised supramolecular complex likely held together by direct base-pairings between packaging signals.
Collapse
Affiliation(s)
- Emilie Fournier
- Architecture et Réactivité de l'ARN, Université de Strasbourg, CNRS, IBMC, 15 rue René Descartes, 67084 Strasbourg, France.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
75
|
Nelson MI, Detmer SE, Wentworth DE, Tan Y, Schwartzbard A, Halpin RA, Stockwell TB, Lin X, Vincent AL, Gramer MR, Holmes EC. Genomic reassortment of influenza A virus in North American swine, 1998-2011. J Gen Virol 2012; 93:2584-2589. [PMID: 22993190 DOI: 10.1099/vir.0.045930-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Revealing the frequency and determinants of reassortment among RNA genome segments is fundamental to understanding basic aspects of the biology and evolution of the influenza virus. To estimate the extent of genomic reassortment in influenza viruses circulating in North American swine, we performed a phylogenetic analysis of 139 whole-genome viral sequences sampled during 1998-2011 and representing seven antigenically distinct viral lineages. The highest amounts of reassortment were detected between the H3 and the internal gene segments (PB2, PB1, PA, NP, M and NS), while the lowest reassortment frequencies were observed among the H1γ, H1pdm and neuraminidase segments, particularly N1. Less reassortment was observed among specific haemagglutinin-neuraminidase combinations that were more prevalent in swine, suggesting that some genome constellations may be evolutionarily more stable.
Collapse
Affiliation(s)
- Martha I Nelson
- Fogarty International Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Susan E Detmer
- Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | | | - Yi Tan
- Fogarty International Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Aaron Schwartzbard
- Fogarty International Center, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | - Xudong Lin
- J. Craig Venter Institute, Rockville, MD 20850, USA
| | - Amy L Vincent
- Virus and Prion Research Unit, National Animal Center, USDA-ARS, Ames, IA 50010, USA
| | - Marie R Gramer
- University of Minnesota Veterinary Diagnostic Laboratory, St Paul, MN 55108, USA
| | - Edward C Holmes
- Center for Infectious Disease Dynamics, Department of Biology, The Pennsylvania State University, University Park, PA 16802, USA.,Fogarty International Center, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
76
|
Louz D, Bergmans HE, Loos BP, Hoeben RC. Animal models in virus research: their utility and limitations. Crit Rev Microbiol 2012; 39:325-61. [PMID: 22978742 DOI: 10.3109/1040841x.2012.711740] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Viral diseases are important threats to public health worldwide. With the number of emerging viral diseases increasing the last decades, there is a growing need for appropriate animal models for virus studies. The relevance of animal models can be limited in terms of mimicking human pathophysiology. In this review, we discuss the utility of animal models for studies of influenza A viruses, HIV and SARS-CoV in light of viral emergence, assessment of infection and transmission risks, and regulatory decision making. We address their relevance and limitations. The susceptibility, immune responses, pathogenesis, and pharmacokinetics may differ between the various animal models. These complexities may thwart translating results from animal experiments to the humans. Within these constraints, animal models are very informative for studying virus immunopathology and transmission modes and for translation of virus research into clinical benefit. Insight in the limitations of the various models may facilitate further improvements of the models.
Collapse
Affiliation(s)
- Derrick Louz
- National Institute for Public Health and the Environment (RIVM), GMO Office , Bilthoven , The Netherlands
| | | | | | | |
Collapse
|
77
|
Bodewes R, Nieuwkoop NJ, Verburgh RJ, Fouchier RAM, Osterhaus ADME, Rimmelzwaan GF. Use of influenza A viruses expressing reporter genes to assess the frequency of double infections in vitro. J Gen Virol 2012; 93:1645-1648. [PMID: 22535774 DOI: 10.1099/vir.0.042671-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Exchange of gene segments between mammalian and avian influenza A viruses may lead to the emergence of potential pandemic influenza viruses. Since co-infection of single cells with two viruses is a prerequisite for reassortment to take place, we assessed frequencies of double-infection in vitro using influenza A/H5N1 and A/H1N1 viruses expressing the reporter genes eGFP or mCherry. Double-infected A549 and Madin-Darby canine kidney cells were detected by confocal microscopy and flow cytometry.
Collapse
Affiliation(s)
- R Bodewes
- Erasmus Medical Centre, Dr. Molewaterplein 50, 3015GE Rotterdam, The Netherlands
| | - N J Nieuwkoop
- Erasmus Medical Centre, Dr. Molewaterplein 50, 3015GE Rotterdam, The Netherlands
| | - R J Verburgh
- Erasmus Medical Centre, Dr. Molewaterplein 50, 3015GE Rotterdam, The Netherlands
| | - R A M Fouchier
- Erasmus Medical Centre, Dr. Molewaterplein 50, 3015GE Rotterdam, The Netherlands
| | - A D M E Osterhaus
- ViroClinics Biosciences, Dr. Molewaterplein 50, 3015GE Rotterdam, The Netherlands
- Erasmus Medical Centre, Dr. Molewaterplein 50, 3015GE Rotterdam, The Netherlands
| | - G F Rimmelzwaan
- ViroClinics Biosciences, Dr. Molewaterplein 50, 3015GE Rotterdam, The Netherlands
- Erasmus Medical Centre, Dr. Molewaterplein 50, 3015GE Rotterdam, The Netherlands
| |
Collapse
|
78
|
Jang YH, Seong BL. Principles underlying rational design of live attenuated influenza vaccines. Clin Exp Vaccine Res 2012; 1:35-49. [PMID: 23596576 PMCID: PMC3623510 DOI: 10.7774/cevr.2012.1.1.35] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2012] [Revised: 05/23/2012] [Accepted: 06/10/2012] [Indexed: 12/18/2022] Open
Abstract
Despite recent innovative advances in molecular virology and the developments of vaccines, influenza virus remains a serious burden for human health. Vaccination has been considered a primary countermeasure for prevention of influenza infection. Live attenuated influenza vaccines (LAIVs) are particularly attracting attention as an effective strategy due to several advantages over inactivated vaccines. Cold-adaptation, as a classical means for attenuating viral virulence, has been successfully used for generating safe and effective donor strains of LAIVs against seasonal epidemics and occasional pandemics. Recently, the advent of reverse genetics technique expedited a variety of rational strategies to broaden the pool of LAIVs. Considering the breadth of antigenic diversity of influenza virus, the pool of LAIVs is likely to equip us with better options for controlling influenza pandemics. With a brief reflection on classical attenuating strategies used at the initial stage of development of LAIVs, especially on the principles underlying the development of cold-adapted LAIVs, we further discuss and outline other attenuation strategies especially with respect to the rationales for attenuation, and their practicality for mass production. Finally, we propose important considerations for a rational vaccine design, which will provide us with practical guidelines for improving the safety and effectiveness of LAIVs.
Collapse
Affiliation(s)
- Yo Han Jang
- Laboratory of Molecular Medicine, Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | | |
Collapse
|
79
|
Cheng X, Zengel JR, Xu Q, Jin H. Surface glycoproteins of influenza A H3N2 virus modulate virus replication in the respiratory tract of ferrets. Virology 2012; 432:91-8. [PMID: 22743127 DOI: 10.1016/j.virol.2012.06.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2012] [Revised: 05/08/2012] [Accepted: 06/01/2012] [Indexed: 11/16/2022]
Abstract
The hemagglutinin (HA) genes of the influenza A H3N2 subtype viruses isolated from 1968 to 2010 have evolved substantially but their neuraminidase (NA) genes have been relatively less divergent. The H3N2 viruses isolated since 1995 were found to replicate in the lower respiratory tract of ferrets less efficiently than the earlier isolates. To evaluate whether the HA or/and NA or the internal protein gene segments of the H3N2 virus affected viral replication in the respiratory tract of ferrets, recombinant A/California/07/2004 (CA04) (H3N2) virus and its reassortants that contained the same CA04 internal protein gene segments and the HA and/or NA of A/Udorn/309/1972 (UD72) or A/Wuhan/359/1995 (WH95) H3N2 viruses were generated and evaluated for their replication in the respiratory tract of ferrets. All the reassortant viruses replicated efficiently in the upper respiratory tract of ferrets, but their replication in the lower respiratory tract of ferrets varied. In contrast to the UD72-HA reassortant virus that replicated efficiently in the lungs of ferrets, the virus with the WH95-HA or the CA04-HA either replicated modestly or did not replicate in the lungs of ferrets. The reassortants with the WH95-HA and UD72-NA or CA04-NA had the tendency to lose a N-linked glycosylation site at residue 246 in the HA, resulting in viral lung titer of 100-fold higher than the virus with the HA and NA from WH95. The UD72-NA had the highest neuraminidase activity and increased viral replication by up to 100-fold in tissue culture cells during early infection. Thus, our data indicate that both the HA and NA glycoproteins play important roles in viral replication of the H3N2 influenza virus in ferrets.
Collapse
|
80
|
Thontiravong A, Kitikoon P, Wannaratana S, Tantilertcharoen R, Tuanudom R, Pakpinyo S, Sasipreeyajan J, Oraveerakul K, Amonsin A. Quail as a potential mixing vessel for the generation of new reassortant influenza A viruses. Vet Microbiol 2012; 160:305-13. [PMID: 22763173 DOI: 10.1016/j.vetmic.2012.05.043] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2012] [Revised: 05/28/2012] [Accepted: 05/31/2012] [Indexed: 10/28/2022]
Abstract
Quail has been proposed as one of the intermediate hosts supporting the generation of newly reassortant influenza A viruses (IAVs) with the potential to infect humans. To evaluate the role of quail as an intermediate host of IAVs, co-infections of quail with swine-origin pandemic H1N1 2009 (pH1N1) and low pathogenic avian influenza (LPAI) duck H3N2 (dkH3N2) viruses (n=10) or endemic Thai swine H1N1 (swH1N1) and dkH3N2 viruses (n=10) were conducted. Three additional groups of five quail were each inoculated with pH1N1, swH1N1 and dkH3N2 as control groups to verify that each virus can infect quail. Our result showed that co-infected quail shed higher viral titers from the respiratory tract than single virus infected quail. This study confirmed that reassortant viruses could be readily generated in the respiratory tract of quail from both the pH1N1/dkH3N2 co-infected group (100% of quail generating reassortant viruses) and the swH1N1/dkH3N2 (33% of quail generating reassortant viruses) co-infected group without discernible clinical signs. The reassortment efficacy between the two combination of viruses was different in that the frequency of reassortant viruses was significantly higher in pH1N1/dkH3N2 co-infected quail (21.4%) compared to swH1N1/dkH3N2 co-infected quail (0.8%), indicating that gene combinations in pH1N1 have a higher potential to reassort with dkH3N2 compared to swH1N1. In summary, our result confirmed that quail could be an intermediate host of IAVs for generating new reassortant viruses. Our finding highlights the importance of monitoring IAVs especially pH1N1 in quail.
Collapse
Affiliation(s)
- Aunyaratana Thontiravong
- Interdepartment of Biomedical Sciences, Faculty of Graduate School, Chulalongkorn University, Bangkok 10330, Thailand
| | | | | | | | | | | | | | | | | |
Collapse
|
81
|
Wang Q, Zhang S, Jiang H, Wang J, Weng L, Mao Y, Sekiguchi S, Yasui F, Kohara M, Buchy P, Deubel V, Xu K, Sun B, Toyoda T. PA from an H5N1 highly pathogenic avian influenza virus activates viral transcription and replication and induces apoptosis and interferon expression at an early stage of infection. Virol J 2012; 9:106. [PMID: 22681768 PMCID: PMC3507744 DOI: 10.1186/1743-422x-9-106] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2011] [Accepted: 05/21/2012] [Indexed: 12/28/2022] Open
Abstract
Background Although gene exchange is not likely to occur freely, reassortment between the H5N1 highly pathogenic avian influenza virus (HPAIV) and currently circulating human viruses is a serious concern. The PA polymerase subunit of H5N1 HPAIV was recently reported to activate the influenza replicon activity. Methods The replicon activities of PR8 and WSN strains (H1N1) of influenza containing PA from HPAIV A/Cambodia/P0322095/2005 (H5N1) and the activity of the chimeric RNA polymerase were analyzed. A reassortant WSN virus containing the H5N1 Cambodia PA (C-PA) was then reconstituted and its growth in cells and pathogenicity in mice examined. The interferon promoter, TUNEL, and caspase 3, 8, and 9 activities of C-PA-infected cells were compared with those of WSN-infected cells. Results The activity of the chimeric RNA polymerase was slightly higher than that of WSN, and C-PA replicated better than WSN in cells. However, the multi-step growth of C-PA and its pathogenicity in mice were lower than those of WSN. The interferon promoter, TUNEL, and caspase 3, 8, and 9 activities were strongly induced in early infection in C-PA-infected cells but not in WSN-infected cells. Conclusions Apoptosis and interferon were strongly induced early in C-PA infection, which protected the uninfected cells from expansion of viral infection. In this case, these classical host-virus interactions contributed to the attenuation of this strongly replicating virus.
Collapse
Affiliation(s)
- Qiang Wang
- Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, 411 Hefei Road, 200025, Shanghai, P. R. China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Driskell EA, Pickens JA, Humberd-Smith J, Gordy JT, Bradley KC, Steinhauer DA, Berghaus RD, Stallknecht DE, Howerth EW, Tompkins SM. Low pathogenic avian influenza isolates from wild birds replicate and transmit via contact in ferrets without prior adaptation. PLoS One 2012; 7:e38067. [PMID: 22675507 PMCID: PMC3365887 DOI: 10.1371/journal.pone.0038067] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2011] [Accepted: 04/30/2012] [Indexed: 12/02/2022] Open
Abstract
Direct transmission of avian influenza viruses to mammals has become an increasingly investigated topic during the past decade; however, isolates that have been primarily investigated are typically ones originating from human or poultry outbreaks. Currently there is minimal comparative information on the behavior of the innumerable viruses that exist in the natural wild bird host. We have previously demonstrated the capacity of numerous North American avian influenza viruses isolated from wild birds to infect and induce lesions in the respiratory tract of mice. In this study, two isolates from shorebirds that were previously examined in mice (H1N9 and H6N1 subtypes) are further examined through experimental inoculations in the ferret with analysis of viral shedding, histopathology, and antigen localization via immunohistochemistry to elucidate pathogenicity and transmission of these viruses. Using sequence analysis and glycan binding analysis, we show that these avian viruses have the typical avian influenza binding pattern, with affinity for cell glycoproteins/glycolipids having terminal sialic acid (SA) residues with α 2,3 linkage [Neu5Ac(α2,3)Gal]. Despite the lack of α2,6 linked SA binding, these AIVs productively infected both the upper and lower respiratory tract of ferrets, resulting in nasal viral shedding and pulmonary lesions with minimal morbidity. Moreover, we show that one of the viruses is able to transmit to ferrets via direct contact, despite its binding affinity for α 2,3 linked SA residues. These results demonstrate that avian influenza viruses, which are endemic in aquatic birds, can potentially infect humans and other mammals without adaptation. Finally this work highlights the need for additional study of the wild bird subset of influenza viruses in regard to surveillance, transmission, and potential for reassortment, as they have zoonotic potential.
Collapse
Affiliation(s)
- Elizabeth A. Driskell
- Department of Pathology, University of Georgia, Athens, Georgia, United States of America
| | - Jennifer A. Pickens
- Department of Infectious Diseases, University of Georgia, Athens, Georgia, United States of America
| | - Jennifer Humberd-Smith
- Department of Infectious Diseases, University of Georgia, Athens, Georgia, United States of America
| | - James T. Gordy
- Department of Infectious Diseases, University of Georgia, Athens, Georgia, United States of America
| | - Konrad C. Bradley
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - David A. Steinhauer
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Roy D. Berghaus
- Department of Population Health, University of Georgia, Athens, Georgia, United States of America
| | - David E. Stallknecht
- Department of Population Health, University of Georgia, Athens, Georgia, United States of America
| | - Elizabeth W. Howerth
- Department of Pathology, University of Georgia, Athens, Georgia, United States of America
| | - Stephen Mark Tompkins
- Department of Infectious Diseases, University of Georgia, Athens, Georgia, United States of America
- * E-mail:
| |
Collapse
|
83
|
Zhou F, Zhou J, Ma L, Song S, Zhang X, Li W, Jiang S, Wang Y, Liao G. High-yield production of a stable Vero cell-based vaccine candidate against the highly pathogenic avian influenza virus H5N1. Biochem Biophys Res Commun 2012; 421:850-4. [PMID: 22554519 DOI: 10.1016/j.bbrc.2012.04.101] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Accepted: 04/19/2012] [Indexed: 10/28/2022]
Abstract
Highly pathogenic avian influenza (HPAI) viruses pose a global pandemic threat, for which rapid large-scale vaccine production technology is critical for prevention and control. Because chickens are highly susceptible to HPAI viruses, the supply of chicken embryos for vaccine production might be depleted during a virus outbreak. Therefore, developing HPAI virus vaccines using other technologies is critical. Meeting vaccine demand using the Vero cell-based fermentation process has been hindered by low stability and yield. In this study, a Vero cell-based HPAI H5N1 vaccine candidate (H5N1/YNVa) with stable high yield was achieved by reassortment of the Vero-adapted (Va) high growth A/Yunnan/1/2005(H3N2) (YNVa) virus with the A/Anhui/1/2005(H5N1) attenuated influenza vaccine strain (H5N1delta) using the 6/2 method. The reassorted H5N1/YNVa vaccine maintained a high hemagglutination (HA) titer of 1024. Furthermore, H5N1/YNVa displayed low pathogenicity and uniform immunogenicity compared to that of the parent virus.
Collapse
Affiliation(s)
- Fangye Zhou
- No. 5, Department of Bioproducts, Institute of Medical Biology, Chinese Academy of Medical Science and Pecking Union Medical College, Jiaoling Avenue 935, Kunming, Yunnan Province 650102, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
84
|
Cold-adapted pandemic 2009 H1N1 influenza virus live vaccine elicits cross-reactive immune responses against seasonal and H5 influenza A viruses. J Virol 2012; 86:5953-8. [PMID: 22438541 DOI: 10.1128/jvi.07149-11] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The rapid transmission of the pandemic 2009 H1N1 influenza virus (pH1N1) among humans has raised the concern of a potential emergence of reassortment between pH1N1 and highly pathogenic influenza strains, especially the avian H5N1 influenza virus. Here, we report that the cold-adapted pH1N1 live attenuated vaccine (CApH1N1) elicits cross-reactive immunity to seasonal and H5 influenza A viruses in the mouse model. Immunization with CApH1N1 induced both systemic and mucosal antibodies with broad reactivity to seasonal and H5 strains, including HAPI H5N1 and the avian H5N2 virus, providing complete protection against heterologous and heterosubtypic lethal challenges. Our results not only accentuate the merit of using live attenuated influenza virus vaccines in view of cross-reactivity but also represent the potential of CApH1N1 live vaccine for mitigating the clinical severity of infections that arise from reassortments between pH1N1 and highly pathogenic H5 subtype viruses.
Collapse
|
85
|
Nakazono Y, Hara K, Kashiwagi T, Hamada N, Watanabe H. The RNA polymerase PB2 subunit of influenza A/HongKong/156/1997 (H5N1) restricts the replication of reassortant ribonucleoprotein complexes [corrected]. PLoS One 2012; 7:e32634. [PMID: 22389716 PMCID: PMC3289661 DOI: 10.1371/journal.pone.0032634] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2011] [Accepted: 01/28/2012] [Indexed: 11/18/2022] Open
Abstract
Background Genetic reassortment plays a critical role in the generation of pandemic strains of influenza virus. The influenza virus RNA polymerase, composed of PB1, PB2 and PA subunits, has been suggested to influence the efficiency of genetic reassortment. However, the role of the RNA polymerase in the genetic reassortment is not well understood. Methodology/Principal Findings Here, we reconstituted reassortant ribonucleoprotein (RNP) complexes, and demonstrated that the PB2 subunit of A/HongKong/156/1997 (H5N1) [HK PB2] dramatically reduced the synthesis of mRNA, cRNA and vRNA when introduced into the polymerase of other influenza strains of H1N1 or H3N2. The HK PB2 had no significant effect on the assembly of the polymerase trimeric complex, or on promoter binding activity or replication initiation activity in vitro. However, the HK PB2 was found to remarkably impair the accumulation of RNP. This impaired accumulation and activity of RNP was fully restored when four amino acids at position 108, 508, 524 and 627 of the HK PB2 were mutated. Conclusions/Significance Overall, we suggest that the PB2 subunit of influenza polymerase might play an important role for the replication of reassortant ribonucleoprotein complexes.
Collapse
Affiliation(s)
- Yoko Nakazono
- Division of Infectious Diseases, Department of Infectious Medicine, Kurume University School of Medicine, Fukuoka, Japan
| | - Koyu Hara
- Division of Infectious Diseases, Department of Infectious Medicine, Kurume University School of Medicine, Fukuoka, Japan
- * E-mail:
| | - Takahito Kashiwagi
- Division of Infectious Diseases, Department of Infectious Medicine, Kurume University School of Medicine, Fukuoka, Japan
| | - Nobuyuki Hamada
- Division of Infectious Diseases, Department of Infectious Medicine, Kurume University School of Medicine, Fukuoka, Japan
| | - Hiroshi Watanabe
- Division of Infectious Diseases, Department of Infectious Medicine, Kurume University School of Medicine, Fukuoka, Japan
| |
Collapse
|
86
|
Abstract
Influenza A virus reservoirs in animals have provided novel genetic elements leading to the emergence of global pandemics in humans. Most influenza A viruses circulate in waterfowl, but those that infect mammalian hosts are thought to pose the greatest risk for zoonotic spread to humans and the generation of pandemic or panzootic viruses. We have identified an influenza A virus from little yellow-shouldered bats captured at two locations in Guatemala. It is significantly divergent from known influenza A viruses. The HA of the bat virus was estimated to have diverged at roughly the same time as the known subtypes of HA and was designated as H17. The neuraminidase (NA) gene is highly divergent from all known influenza NAs, and the internal genes from the bat virus diverged from those of known influenza A viruses before the estimated divergence of the known influenza A internal gene lineages. Attempts to propagate this virus in cell cultures and chicken embryos were unsuccessful, suggesting distinct requirements compared with known influenza viruses. Despite its divergence from known influenza A viruses, the bat virus is compatible for genetic exchange with human influenza viruses in human cells, suggesting the potential capability for reassortment and contributions to new pandemic or panzootic influenza A viruses.
Collapse
|
87
|
Giles BM, Bissel SJ, DeAlmeida DR, Wiley CA, Ross TM. Antibody breadth and protective efficacy are increased by vaccination with computationally optimized hemagglutinin but not with polyvalent hemagglutinin-based H5N1 virus-like particle vaccines. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2012; 19:128-39. [PMID: 22190399 PMCID: PMC3272934 DOI: 10.1128/cvi.05533-11] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2011] [Accepted: 12/06/2011] [Indexed: 12/25/2022]
Abstract
One of the challenges for developing an H5N1 influenza vaccine is the diversity of antigenically distinct isolates within this subtype. Previously, our group described a novel hemagglutinin (HA) derived from a methodology termed computationally optimized broadly reactive antigen (COBRA). This COBRA HA, when used as an immunogen, elicits a broad antibody response against H5N1 isolates from different clades. In this report, the immune responses elicited by the COBRA HA virus-like particle (VLP) vaccine were compared to responses elicited by a mixture of VLPs expressing representative HA molecules from clade 2.1, 2.2, and 2.3 primary H5N1 isolates (polyvalent). The COBRA HA VLP vaccine elicited higher-titer antibodies to a panel of H5N1 HA proteins than did the other VLPs. Both COBRA and polyvalent vaccines protected vaccinated mice and ferrets from experimental infection with highly lethal H5N1 influenza viruses, but COBRA-vaccinated animals had decreased viral replication, less inflammation in the lungs of mice, and reduced virus recovery in ferret nasal washes. Both vaccines had similar cellular responses postchallenge, indicating that higher-titer serum antibodies likely restrict the duration of viral replication. Furthermore, passively transferred immune serum from the COBRA HA VLP-vaccinated mice protected recipient animals more efficiently than immune serum from polyvalent-vaccinated mice. This is the first report comparing these two vaccine strategies. The single COBRA HA antigen elicited a broader antibody response and reduced morbidity and viral titers more effectively than a polyvalent mixture of primary H5N1 HA antigens.
Collapse
Affiliation(s)
- Brendan M. Giles
- Center for Vaccine Research
- Graduate Program in Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Stephanie J. Bissel
- Division of Neuropathology, Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | - Clayton A. Wiley
- Division of Neuropathology, Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ted M. Ross
- Center for Vaccine Research
- Department of Microbiology and Molecular Genetics
- Graduate Program in Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
88
|
Gíria MT, Rebelo de Andrade H, Santos LA, Correia VM, Pedro SV, Santos MA. Genomic signatures and antiviral drug susceptibility profile of A(H1N1)pdm09. J Clin Virol 2011; 53:140-4. [PMID: 22177272 DOI: 10.1016/j.jcv.2011.11.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2011] [Revised: 09/20/2011] [Accepted: 11/08/2011] [Indexed: 11/28/2022]
Abstract
BACKGROUND Genetic changes in influenza surface and internal genes can alter viral fitness and virulence. Mutation trend analysis and antiviral drug susceptibility profiling of A(H1N1)pdm09 viruses is essential for risk assessment of emergent strains and disease management. OBJECTIVE To profile genomic signatures and antiviral drug resistance of A(H1N1)pdm09 viruses and to discuss the potential role of mutated residues in human host adaptation and virulence. STUDY DESIGN A(H1N1)pdm09 viruses circulating in Portugal during pandemic and post-pandemic periods and 2009/2010 season. Viruses were isolated in MDCK-SIAT1 cell culture and subjected to mutation analysis of surface and internal proteins, and to antiviral drug susceptibility profiling. RESULTS The A(H1N1)pdm09 strains circulating during the epidemic period in Portugal were resistant to amantadine. The majority of the strains were found to be susceptible to oseltamivir and zanamivir, with five outliers to neuraminidase inhibitors (NAIs) identified. Specific mutation patterns were detected within the functional domains of internal proteins PB2, PB1, PA, NP, NS1, M1 and NS2/NEP, which were common to all isolates and also some cluster-specific. DISCUSSION Modification of viral genome transcription, replication and apoptosis kinetics, changes in antigenicity and antiviral drug susceptibility are known determinants of virulence. We report several point mutations with putative roles in viral fitness and virulence, and discuss their potential to result in more virulent phenotypes. Monitoring of specific mutations and genetic patterns in influenza viral genes is essential for risk assessing emergent strains, disease epidemiology and public health implications.
Collapse
Affiliation(s)
- Marta Tiago Gíria
- CPM-URIA, Faculdade de Farmácia da Universidade de Lisboa, Lisboa, Portugal
| | | | | | | | | | | |
Collapse
|
89
|
Reassortment and mutation of the avian influenza virus polymerase PA subunit overcome species barriers. J Virol 2011; 86:1750-7. [PMID: 22090127 DOI: 10.1128/jvi.06203-11] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The emergence of new pandemic influenza A viruses requires overcoming barriers to cross-species transmission as viruses move from animal reservoirs into humans. This complicated process is driven by both individual gene mutations and genome reassortments. The viral polymerase complex, composed of the proteins PB1, PB2, and PA, is a major factor controlling host adaptation, and reassortment events involving polymerase gene segments occurred with past pandemic viruses. Here we investigate the ability of polymerase reassortment to restore the activity of an avian influenza virus polymerase that is normally impaired in human cells. Our data show that the substitution of human-origin PA subunits into an avian influenza virus polymerase alleviates restriction in human cells and increases polymerase activity in vitro. Reassortants with 2009 pandemic H1N1 PA proteins were the most active. Mutational analyses demonstrated that the majority of the enhancing activity in human PA results from a threonine-to-serine change at residue 552. Reassortant viruses with avian polymerases and human PA subunits, or simply the T552S mutation, displayed faster replication kinetics in culture and increased pathogenicity in mice compared to those containing a wholly avian polymerase complex. Thus, the acquisition of a human PA subunit, or the signature T552S mutation, is a potential mechanism to overcome the species-specific restriction of avian polymerases and increase virus replication. Our data suggest that the human, avian, swine, and 2009 H1N1-like viruses that are currently cocirculating in pig populations set the stage for PA reassortments with the potential to generate novel viruses that could possess expanded tropism and enhanced pathogenicity.
Collapse
|
90
|
Xu C, Hu WB, Xu K, He YX, Wang TY, Chen Z, Li TX, Liu JH, Buchy P, Sun B. Amino acids 473V and 598P of PB1 from an avian-origin influenza A virus contribute to polymerase activity, especially in mammalian cells. J Gen Virol 2011; 93:531-540. [PMID: 22090209 DOI: 10.1099/vir.0.036434-0] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
It has been reported that the avian-origin influenza A virus PB1 protein (avian PB1) enhances influenza A virus polymerase activity in mammalian cells when it replaces the human-origin PB1 protein (human PB1). Characterization of the amino acid residues that contribute to this enhancement is needed. In this study, it was found that PB1 from an avian-origin influenza A virus [A/Cambodia/P0322095/2005, H5N1 (Cam)] could enhance the polymerase activity of an attenuated human isolated virus, A/WSN/33, carrying the PB2 K627E mutation (WSN627E) in vitro. Furthermore, 473V and 598P in the Cam PB1 were identified as the residues responsible for this enhanced activity. The results from recombinant virus experiments demonstrated the contribution of PB1 amino acids 473V and 598P to polymerase activity in mammalian cells and in mice. Interestingly, 473V is conserved in pH1N1 viruses from the 2009 pandemic. Substitution of 473V by leucine in pH1N1 PB1 led to a decreased viral polymerase activity and a lower growth rate in mammalian cells, suggesting that the PB1 473V also plays a role in maintaining efficient virus replication of the pH1N1 virus. Thus, it was concluded that two amino acids in avian-origin PB1, 473V and 598P, contribute to the polymerase activity of the H5N1 virus, especially in mammalian cells, and that 473V in PB1 also contributes to efficient replication of the pH1N1 strain.
Collapse
Affiliation(s)
- Chen Xu
- Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200025, PR China
| | - Wei-Bin Hu
- Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200025, PR China
| | - Ke Xu
- Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200025, PR China
| | - Yun-Xia He
- Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200025, PR China
| | - Tong-Yan Wang
- Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200025, PR China
| | - Ze Chen
- Shanghai Institute of Biological Products, Shanghai 200052, PR China
| | - Tian-Xian Li
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, Hubei, PR China
| | - Jin-Hua Liu
- College of Veterinary Medicine, China Agricultural University, Beijing 100094, PR China
| | - Philippe Buchy
- Institut Pasteur in Cambodia, 5 Monivong Blvd, PO Box 983, Phnom Penh, Cambodia
| | - Bing Sun
- Laboratory of Molecular Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes of Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, PR China.,Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200025, PR China
| |
Collapse
|
91
|
Increased pathogenicity of a reassortant 2009 pandemic H1N1 influenza virus containing an H5N1 hemagglutinin. J Virol 2011; 85:12262-70. [PMID: 21917948 DOI: 10.1128/jvi.05582-11] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
A novel H1N1 influenza virus emerged in 2009 (pH1N1) to become the first influenza pandemic of the 21st century. This virus is now cocirculating with highly pathogenic H5N1 avian influenza viruses in many parts of the world, raising concerns that a reassortment event may lead to highly pathogenic influenza strains with the capacity to infect humans more readily and cause severe disease. To investigate the virulence of pH1N1-H5N1 reassortant viruses, we created pH1N1 (A/California/04/2009) viruses expressing individual genes from an avian H5N1 influenza strain (A/Hong Kong/483/1997). Using several in vitro models of virus replication, we observed increased replication for a reassortant CA/09 virus expressing the hemagglutinin (HA) gene of HK/483 (CA/09-483HA) relative to that of either parental CA/09 virus or reassortant CA/09 expressing other HK/483 genes. This increased replication correlated with enhanced pathogenicity in infected mice similar to that of the parental HK/483 strain. The serial passage of the CA/09 parental virus and the CA/09-483HA virus through primary human lung epithelial cells resulted in increased pathogenicity, suggesting that these viruses easily adapt to humans and become more virulent. In contrast, serial passage attenuated the parental HK/483 virus in vitro and resulted in slightly reduced morbidity in vivo, suggesting that sustained replication in humans attenuates H5N1 avian influenza viruses. Taken together, these data suggest that reassortment between cocirculating human pH1N1 and avian H5N1 influenza strains will result in a virus with the potential for increased pathogenicity in mammals.
Collapse
|
92
|
H5N1 influenza virus pathogenesis in genetically diverse mice is mediated at the level of viral load. mBio 2011; 2:mBio.00171-11. [PMID: 21896679 PMCID: PMC3171982 DOI: 10.1128/mbio.00171-11] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
UNLABELLED The genotype of the host is one of several factors involved in the pathogenesis of an infectious disease and may be a key parameter in the epidemiology of highly pathogenic H5N1 influenza virus infection in humans. Gene polymorphisms may affect the viral replication rate or alter the host's immune response to the virus. In humans, it is unclear which aspect dictates the severity of H5N1 virus disease. To identify the mechanism underlying differential responses to H5N1 virus infection in a genetically diverse population, we assessed the host responses and lung viral loads in 21 inbred mouse strains upon intranasal inoculation with A/Hong Kong/213/03 (H5N1). Resistant mouse strains survived large inocula while susceptible strains succumbed to infection with 1,000- to 10,000-fold-lower doses. Quantitative analysis of the viral load after inoculation with an intermediate dose found significant associations with lethality as early as 2 days postinoculation, earlier than any other disease indicator. The increased viral titers in the highly susceptible strains mediated a hyperinflamed environment, indicated by the distinct expression profiles and increased production of inflammatory mediators on day 3. Supporting the hypothesis that viral load rather than an inappropriate response to the virus was the key severity-determining factor, we performed quantitative real-time PCR measuring the cytokine/viral RNA ratio. No significant differences between susceptible and resistant mouse strains were detected, confirming that it is the host genetic component controlling viral load, and therefore replication dynamics, that is primarily responsible for a host's susceptibility to a given H5N1 virus. IMPORTANCE Highly pathogenic H5N1 influenza virus has circulated in Southeast Asia since 2003 but has been confirmed in relatively few individuals. It has been postulated that host genetic polymorphisms increase the susceptibility to infection and severe disease. The mechanisms and host proteins affected during severe disease are unknown. Inbred mouse strains vary considerably in their ability to resist H5N1 virus and were used to identify the primary mechanism determining disease severity. After inoculation with H5N1, resistant mouse strains had reduced amounts of virus in their lungs, which subsequently resulted in lower production of proinflammatory mediators and less pathology. We therefore conclude that the host genetic component controlling disease severity is primarily influencing viral replication. This is an important concept, as it emphasizes the need to limit virus replication through antiviral therapies and it shows that the hyperinflammatory environment is simply a reflection of more viral genetic material inducing a response.
Collapse
|
93
|
Christman MC, Kedwaii A, Xu J, Donis RO, Lu G. Pandemic (H1N1) 2009 virus revisited: an evolutionary retrospective. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2011; 11:803-11. [PMID: 21382522 PMCID: PMC3141221 DOI: 10.1016/j.meegid.2011.02.021] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2010] [Revised: 02/23/2011] [Accepted: 02/24/2011] [Indexed: 11/24/2022]
Abstract
The pandemic (H1N1) 2009 virus is unique in many aspects, especially in its genetics and evolution. In this paper, we examine the molecular mechanisms underlying the evolution of this novel virus through a comprehensive bioinformatics analysis, and present results in the context of a review of the literature. The pandemic virus was found to arise from a reassortment of two swine viruses, each of which ultimately arose from interspecies transmission. It experienced fast evolutionary rates and strong selection pressures, diverging into two different clusters at the early pandemic stage. Cluster I became extinct at the end of 2009 whereas Cluster II continued to circulate at much lower rates in 2010. Therefore, on August 10 of 2010 the WHO declared the end of the pandemic. Important mutations associated with host specificity, virulence, and drug resistance were detected in the pandemic virus, indicating effective transmission and increased severity in humans. Much has been learned about the evolutionary dynamics of this pandemic virus; however, it is still impossible to predict when the next pandemic will occur and which virus will be responsible. Improved surveillance at different levels (both national and international) and in different hosts (especially in swine) appears to be crucial for early detection and prevention of future influenza pandemics.
Collapse
Affiliation(s)
- MC Christman
- Department of Biology, University of Nebraska at Omaha, Omaha, NE 68182, USA
| | - A Kedwaii
- Department of Biology, University of Nebraska at Omaha, Omaha, NE 68182, USA
| | - J Xu
- Department of Biology, University of Nebraska at Omaha, Omaha, NE 68182, USA
| | - RO Donis
- Influenza Division, Molecular Virology and Vaccines Branch, Centers for Disease Control and Prevention, Atlanta, GA
| | - G Lu
- Department of Biology, University of Nebraska at Omaha, Omaha, NE 68182, USA
| |
Collapse
|
94
|
Zhang J, Mamlouk AM, Martinetz T, Chang S, Wang J, Hilgenfeld R. PhyloMap: an algorithm for visualizing relationships of large sequence data sets and its application to the influenza A virus genome. BMC Bioinformatics 2011; 12:248. [PMID: 21689434 PMCID: PMC3142226 DOI: 10.1186/1471-2105-12-248] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2011] [Accepted: 06/20/2011] [Indexed: 11/10/2022] Open
Abstract
Background Results of phylogenetic analysis are often visualized as phylogenetic trees. Such a tree can typically only include up to a few hundred sequences. When more than a few thousand sequences are to be included, analyzing the phylogenetic relationships among them becomes a challenging task. The recent frequent outbreaks of influenza A viruses have resulted in the rapid accumulation of corresponding genome sequences. Currently, there are more than 7500 influenza A virus genomes in the database. There are no efficient ways of representing this huge data set as a whole, thus preventing a further understanding of the diversity of the influenza A virus genome. Results Here we present a new algorithm, "PhyloMap", which combines ordination, vector quantization, and phylogenetic tree construction to give an elegant representation of a large sequence data set. The use of PhyloMap on influenza A virus genome sequences reveals the phylogenetic relationships of the internal genes that cannot be seen when only a subset of sequences are analyzed. Conclusions The application of PhyloMap to influenza A virus genome data shows that it is a robust algorithm for analyzing large sequence data sets. It utilizes the entire data set, minimizes bias, and provides intuitive visualization. PhyloMap is implemented in JAVA, and the source code is freely available at http://www.biochem.uni-luebeck.de/public/software/phylomap.html
Collapse
Affiliation(s)
- Jiajie Zhang
- Institute of Biochemistry, Center for Structural and Cell Biology in Medicine, University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany
| | | | | | | | | | | |
Collapse
|
95
|
Reassortment between seasonal H1N1 and pandemic (H1N1) 2009 influenza viruses is restricted by limited compatibility among polymerase subunits. J Virol 2011; 85:8449-52. [PMID: 21680507 DOI: 10.1128/jvi.05054-11] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Reassortment is important for influenza virus evolution and the generation of novel viruses with pandemic potential; however, the factors influencing reassortment are still poorly understood. Here, using reverse genetics and a replicon assay, we demonstrated that a mixed polymerase complex containing a pandemic (H1N1) 2009 influenza virus PB2 on a seasonal H1N1 virus background has reduced polymerase activity, leading to impaired virus viability. Adaptation of viruses containing the mixed polymerase complex resulted in compensatory mutations in PB1. Taken together, our results identify the cooperation between PB2 and PB1 as an important restricting factor for reassortment of influenza viruses.
Collapse
|
96
|
Virulence and genetic compatibility of polymerase reassortant viruses derived from the pandemic (H1N1) 2009 influenza virus and circulating influenza A viruses. J Virol 2011; 85:6275-86. [PMID: 21507962 DOI: 10.1128/jvi.02125-10] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Gene mutations and reassortment are key mechanisms by which influenza A virus acquires virulence factors. To evaluate the role of the viral polymerase replication machinery in producing virulent pandemic (H1N1) 2009 influenza viruses, we generated various polymerase point mutants (PB2, 627K/701N; PB1, expression of PB1-F2 protein; and PA, 97I) and reassortant viruses with various sources of influenza viruses by reverse genetics. Although the point mutations produced no significant change in pathogenicity, reassortment between the pandemic A/California/04/09 (CA04, H1N1) and current human and animal influenza viruses produced variants possessing a broad spectrum of pathogenicity in the mouse model. Although most polymerase reassortants had attenuated pathogenicity (including those containing seasonal human H3N2 and high-pathogenicity H5N1 virus segments) compared to that of the parental CA04 (H1N1) virus, some recombinants had significantly enhanced virulence. Unexpectedly, one of the five highly virulent reassortants contained a A/Swine/Korea/JNS06/04(H3N2)-like PB2 gene with no known virulence factors; the other four had mammalian-passaged avian-like genes encoding PB2 featuring 627K, PA featuring 97I, or both. Overall, the reassorted polymerase complexes were only moderately compatible for virus rescue, probably because of disrupted molecular interactions involving viral or host proteins. Although we observed close cooperation between PB2 and PB1 from similar virus origins, we found that PA appears to be crucial in maintaining viral gene functions in the context of the CA04 (H1N1) virus. These observations provide helpful insights into the pathogenic potential of reassortant influenza viruses composed of the pandemic (H1N1) 2009 influenza virus and prevailing human or animal influenza viruses that could emerge in the future.
Collapse
|
97
|
Kahn RE, Clouser DF, Richt JA. Emerging infections: a tribute to the one medicine, one health concept. Zoonoses Public Health 2011; 56:407-28. [PMID: 19486315 DOI: 10.1111/j.1863-2378.2009.01255.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Events in the last decade have taught us that we are now, more than ever, vulnerable to fatal zoonotic diseases such as those caused by haemorrhagic fever viruses, influenza, rabies and BSE/vCJD. Future research activities should focus on solutions to these problems arising at the interface between animals and humans. A 4-fold classification of emerging zoonoses was proposed: Type 1: from wild animals to humans (Hanta); Type 1 plus: from wild animals to humans with further human-to-human transmission (AIDS); Type 2: from wild animals to domestic animals to humans (Avian flu) and Type 2 plus: from wild animals to domestic animals to humans, with further human-to-human transmission (Severe Acute Respiratory Syndrome, SARS). The resulting holistic approach to emerging infections links microbiology, veterinary medicine, human medicine, ecology, public health and epidemiology. As emerging 'new' respiratory viruses are identified in many wild and domestic animals, issues of interspecies transmission have become of increasing concern. The development of safe and effective human and veterinary vaccines is a priority. For example, the spread of different influenza viruses has stimulated influenza vaccine development, just as the spread of Ebola and Marburg viruses has led to new approaches to filovirus vaccines. Interdisciplinary collaboration has become essential because of the convergence of human disease, animal disease and a common approach to biosecurity. High containment pathogens pose a significant threat to public health systems, as well as a major research challenge, because of limited experience in case management, lack of appropriate resources in affected areas and a limited number of animal research facilities in developed countries. Animal models that mimic certain diseases are key elements for understanding the underlying mechanisms of disease pathogenesis, as well as for the development and efficacy testing of therapeutics and vaccines. An updated veterinary curriculum is essential to empower future graduates to work in an international environment, applying international standards for disease surveillance, veterinary public health, food safety and animal welfare.
Collapse
Affiliation(s)
- R E Kahn
- Avian Flu Action, Warrington, Cheshire, UK
| | | | | |
Collapse
|
98
|
Abstract
Equine influenza viruses (EIVs) of the H3N8 and H7N7 subtypes are the causative agents of an important disease of horses. While EIV H7N7 apparently is extinct, H3N8 viruses have circulated for more than 50 years. Like human influenza viruses, EIV H3N8 caused a transcontinental pandemic followed by further outbreaks and epidemics, even in populations with high vaccination coverage. Recently, EIV H3N8 jumped the species barrier to infect dogs. Despite its importance as an agent of infectious disease, the mechanisms that underpin the evolutionary and epidemiological dynamics of EIV are poorly understood, particularly at a genomic scale. To determine the evolutionary history and phylodynamics of EIV H3N8, we conducted an extensive analysis of 82 complete viral genomes sampled during a 45-year span. We show that both intra- and intersubtype reassortment have played a major role in the evolution of EIV, and we suggest that intrasubtype reassortment resulted in enhanced virulence while heterosubtypic reassortment contributed to the extinction of EIV H7N7. We also show that EIV evolves at a slower rate than other influenza viruses, even though it seems to be subject to similar immune selection pressures. However, a relatively high rate of amino acid replacement is observed in the polymerase acidic (PA) segment, with some evidence for adaptive evolution. Most notably, an analysis of viral population dynamics provided evidence for a major population bottleneck of EIV H3N8 during the 1980s, which we suggest resulted from changes in herd immunity due to an increase in vaccination coverage.
Collapse
|
99
|
Maines TR, Chen LM, Belser JA, Van Hoeven N, Smith E, Donis RO, Tumpey TM, Katz JM. Multiple genes contribute to the virulent phenotype observed in ferrets of an H5N1 influenza virus isolated from Thailand in 2004. Virology 2011; 413:226-30. [PMID: 21388650 DOI: 10.1016/j.virol.2011.02.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2010] [Revised: 01/25/2011] [Accepted: 02/02/2011] [Indexed: 11/29/2022]
Abstract
Human infections with highly pathogenic H5N1 avian influenza viruses continue to occur in many parts of the world and pose a considerable public health threat. With the use of animal models, the identification of virulence determinants has been instrumental in improving our understanding of how these viruses cause severe disease in humans. Two genetically similar H5N1 viruses (A/Thailand/16/2004 and A/Thailand/SP83/2004) exhibit high or low virulence phenotypes, respectively, in multiple animal models. Reassortant viruses were generated from this virus pair and evaluated in ferrets. Each of the polymerase genes of A/Thailand/16/2004 virus individually conferred increased virulence to A/Thailand/SP83/2004 virus while the neuraminidase of the low virulence virus reduced virulence and replication efficiency of the virulent virus in ferrets unless the homologous HA was present. Our results demonstrate that H5N1 virus virulence determinants are polygenic and that there is an important correlation between polymerase adaptation, efficient replication in the host, and virulence.
Collapse
Affiliation(s)
- Taronna R Maines
- Influenza Division, National Center for Immunization and Respiratory Disease, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA
| | | | | | | | | | | | | | | |
Collapse
|
100
|
High genetic compatibility and increased pathogenicity of reassortants derived from avian H9N2 and pandemic H1N1/2009 influenza viruses. Proc Natl Acad Sci U S A 2011; 108:4164-9. [PMID: 21368167 DOI: 10.1073/pnas.1019109108] [Citation(s) in RCA: 144] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
H9N2 influenza viruses have been circulating worldwide in multiple avian species and repeatedly infecting mammals, including pigs and humans, posing a significant threat to public health. The coexistence of H9N2 and pandemic influenza H1N1/2009 viruses in pigs and humans provides an opportunity for these viruses to reassort. To evaluate the potential public risk of the reassortant viruses derived from these viruses, we used reverse genetics to generate 127 H9 reassortants derived from an avian H9N2 and a pandemic H1N1 virus, and evaluated their compatibility, replication ability, and virulence in mice. These hybrid viruses showed high genetic compatibility and more than half replicated to a high titer in vitro. In vivo studies of 73 of 127 reassortants revealed that all viruses were able to infect mice without prior adaptation and 8 reassortants exhibited higher pathogenicity than both parental viruses. All reassortants with higher virulence than parental viruses contained the PA gene from the 2009 pandemic virus, revealing the important role of the PA gene from the H1N1/2009 virus in generating a reassortant virus with high public health risk. Analyses of the polymerase activity of the 16 ribonucleoprotein combinations in vitro suggested that the PA of H1N1/2009 origin also enhanced polymerase activity. Our results indicate that some avian H9-pandemic reassortants could emerge with a potentially higher threat for humans and also highlight the importance of monitoring the H9-pandemic reassortant viruses that may arise, especially those that possess the PA gene of H1N1/2009 origin.
Collapse
|