451
|
Ray K, Potdar VA, Cherian SS, Pawar SD, Jadhav SM, Waregaonkar SR, Joshi AA, Mishra AC. Characterization of the complete genome of influenza A (H5N1) virus isolated during the 2006 outbreak in poultry in India. Virus Genes 2008; 36:345-53. [PMID: 18214665 DOI: 10.1007/s11262-007-0195-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2007] [Accepted: 12/27/2007] [Indexed: 11/26/2022]
Abstract
An outbreak of highly pathogenic avian influenza A (H5N1) virus in poultry was reported from Nandurbar and Jalgaon districts of Maharashtra and adjoining areas of Uchhal in Gujarat and Burhanpur in Madhya Pradesh in India from January to April, 2006. In the present study, the full genome of two previously uncharacterized strains of H5N1 viruses isolated at the National Institute of Virology (NIV), Pune, from post-mortem tissues of chicken collected from Navapur, Nandurbar district during the outbreak, has been presented. All the genes belong to clade 2.2 of the Z genotype and are close to the 2006 isolates from Iran, Afghanistan, Mongolia, Italy, and Krasnodar. In a study reported earlier, based on the partial gene sequences of HA, the authors (Pattnaik et al.) hypothesized that the viruses in Jalgaon and Navapur, causing outbreaks 12 days apart, were introduced at different times from different sources. However, our Navapur isolates are closer to the isolate reported from Jalgaon than that from Navapur. Molecular markers suggest that the isolates are sensitive to both drugs Oseltamivir and Amantadine. Amino acid residues responsible for pathogenesis, glycosylation, and receptor binding have also been discussed. The relationship between the Indian viruses and those in the East Africa/West-Asia flyway of migratory birds and the position of Nandurbar in this route suggests that the viruses in India may have been introduced through migratory birds although the role of trade as a possible route of introduction of the virus cannot be ruled out.
Collapse
Affiliation(s)
- Koninika Ray
- Microbial Containment Complex, National Institute of Virology, Sus Road, Pashan, Pune 411021, India.
| | | | | | | | | | | | | | | |
Collapse
|
452
|
Uiprasertkul M, Kitphati R, Puthavathana P, Kriwong R, Kongchanagul A, Ungchusak K, Angkasekwinai S, Chokephaibulkit K, Srisook K, Vanprapar N, Auewarakul P. Apoptosis and pathogenesis of avian influenza A (H5N1) virus in humans. Emerg Infect Dis 2008; 13:708-12. [PMID: 17553248 PMCID: PMC2738443 DOI: 10.3201/eid1305.060572] [Citation(s) in RCA: 126] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Apoptosis may play a crucial role in the pathogenesis of pneumonia and lymphopenia caused by this virus in humans. The pathogenesis of avian influenza A (H5N1) virus in humans has not been clearly elucidated. Apoptosis may also play an important role. We studied autopsy specimens from 2 patients who died of infection with this virus. Apoptosis was observed in alveolar epithelial cells, which is the major target cell type for the viral replication. Numerous apoptotic leukocytes were observed in the lung of a patient who died on day 6 of illness. Our data suggest that apoptosis may play a major role in the pathogenesis of influenza (H5N1) virus in humans by destroying alveolar epithelial cells. This pathogenesis causes pneumonia and destroys leukocytes, leading to leukopenia, which is a prominent clinical feature of influenza (H5N1) virus in humans. Whether observed apoptotic cells were a direct result of the viral replication or a consequence of an overactivation of the immune system requires further studies.
Collapse
|
453
|
|
454
|
|
455
|
H5-type influenza virus hemagglutinin is functionally recognized by the natural killer-activating receptor NKp44. J Virol 2007; 82:2028-32. [PMID: 18077718 DOI: 10.1128/jvi.02065-07] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Antiviral immune defenses involve natural killer (NK) cells. We previously showed that the NK-activating receptor NKp44 is involved in the functional recognition of H1-type influenza virus strains by NK cells. In the present study, we investigated the interaction of NKp44 and the hemagglutinin of a primary influenza virus H5N1 isolate. Here we show that recombinant NKp44 recognizes H5-expressing cells and specifically interacts with soluble H5 hemagglutinin. H5-pseudotyped lentiviral particles bind to NK cells expressing NKp44. Following interaction with target cells expressing H5, pseudotyped lentiviral particles, or membrane-associated H5, NK cells show NKp44-mediated induced activity. These findings indicate that NKp44-H5 interactions induce functional NK activation.
Collapse
|
456
|
Abstract
INTRODUCTION Acute respiratory infections are the most common infections in the human population. Among them, virus infections, especially those caused by influenza viruses, have an important place. TYPE A INFLUENZA Type A influeza virus caused three epidemics during the last century. A high percetage of deceased in pandemics of 1918, and 1919 were young, healthy persons, with many of the deaths due to an unusually severe, hemorrhagic pneumonia. At the end of 2003, and the beginning of 2004, an epidemic emerged in South East Asia of poultry influenza caused by animal (avian) virus. Later it spread to the human populaton, with a high death rate of 73% and with a possibility of interhuman transmission. This review article provides an overview of the clinical manifestations, laboratory findings and chest radiographs. Apart from the symptomatic and supportive therapy, there are antiviral drugs and corticosteriods. CONCLUSION The use of vaccine containing subtypes of virus hemagglutinins and neuraminidase from an influenza virus currently infecting the population has a great importance.
Collapse
|
457
|
Rimmelzwaan GF, Nieuwkoop NJ, de Mutsert G, Boon ACM, Kuiken T, Fouchier RAM, Osterhaus ADME. Attachment of infectious influenza A viruses of various subtypes to live mammalian and avian cells as measured by flow cytometry. Virus Res 2007; 129:175-81. [PMID: 17714820 DOI: 10.1016/j.virusres.2007.07.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2007] [Revised: 07/10/2007] [Accepted: 07/12/2007] [Indexed: 11/19/2022]
Abstract
At present there is much interest in the cell tropism and host range of influenza viruses, especially those of the H5N1 subtype. We wished to develop a method that would enable investigation of attachment of infectious virus through the interaction of the hemagglutinin molecule and live mammalian and avian cells and the subsequent infection of these cells. To this end, influenza viruses of various HA subtypes were constructed that either carry the green fluorescent protein (GFP) instead of the neuraminidase protein, or that express GFP in the cytoplasm of infected cells. The HA genes were derived from influenza viruses A/PR/8/34 (H1N1), A/Netherlands/178/95 (H3N2) and A/Vietnam/1194/04 (H5N1). Using these pairs of viruses, attachment and post-attachment events in the virus replication cycle can be distinguished. In general, the expression of NeuAc(alpha2-3)Gal or NeuAc(alpha2-6)Gal receptors on the cells tested corresponded with the attachment of the viruses that were studied with respect to predicted receptor specificity. Virus attachment was not always predictive for efficient infection of the cells.
Collapse
MESH Headings
- Animals
- Birds/virology
- Cell Line
- Flow Cytometry
- Hemagglutinin Glycoproteins, Influenza Virus/genetics
- Hemagglutinin Glycoproteins, Influenza Virus/metabolism
- Humans
- Influenza A Virus, H1N1 Subtype/genetics
- Influenza A Virus, H1N1 Subtype/isolation & purification
- Influenza A Virus, H1N1 Subtype/physiology
- Influenza A Virus, H3N2 Subtype/genetics
- Influenza A Virus, H3N2 Subtype/isolation & purification
- Influenza A Virus, H3N2 Subtype/physiology
- Influenza A Virus, H5N1 Subtype/genetics
- Influenza A Virus, H5N1 Subtype/isolation & purification
- Influenza A Virus, H5N1 Subtype/physiology
- Influenza A virus/genetics
- Influenza A virus/isolation & purification
- Influenza A virus/physiology
- Orthomyxoviridae Infections/virology
- Receptors, Virus/metabolism
- Virus Attachment
- Virus Replication
Collapse
Affiliation(s)
- Guus F Rimmelzwaan
- Department of Virology, Postgraduate School Molecular Medicine, Erasmus Medical Center, Rotterdam, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
458
|
Landolt GA, Olsen CW. Up to new tricks - a review of cross-species transmission of influenza A viruses. Anim Health Res Rev 2007; 8:1-21. [PMID: 17692139 DOI: 10.1017/s1466252307001272] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Influenza is a highly contagious disease that has burdened both humans and animals since ancient times. In humans, the most dramatic consequences of influenza are associated with periodically occurring pandemics. Pandemics require the emergence of an antigenically novel virus to which the majority of the population lacks protective immunity. Historically, influenza A viruses from animals have contributed to the generation of human pandemic viruses and they may do so again in the future. It is, therefore, critical to understand the epidemiological and molecular mechanisms that allow influenza A viruses to cross species barriers. This review summarizes the current knowledge of influenza ecology, and the viral factors that are thought to determine influenza A virus species specificity.
Collapse
Affiliation(s)
- Gabriele A Landolt
- Department of Clinical Sciences, Colorado State University, 300 West Drake Road, Fort Collins, CO 80523, USA.
| | | |
Collapse
|
459
|
Nicholls JM, Bourne AJ, Chen H, Guan Y, Peiris JSM. Sialic acid receptor detection in the human respiratory tract: evidence for widespread distribution of potential binding sites for human and avian influenza viruses. Respir Res 2007; 8:73. [PMID: 17961210 PMCID: PMC2169242 DOI: 10.1186/1465-9921-8-73] [Citation(s) in RCA: 227] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2007] [Accepted: 10/25/2007] [Indexed: 12/02/2022] Open
Abstract
Background Influenza virus binds to cell receptors via sialic acid (SA) linked glycoproteins. They recognize SA on host cells through their haemagglutinins (H). The distribution of SA on cell surfaces is one determinant of host tropism and understanding its expression on human cells and tissues is important for understanding influenza pathogenesis. The objective of this study therefore was to optimize the detection of α2,3-linked and α2,6-linked SA by lectin histochemistry by investigating the binding of Sambucus nigra agglutinin (SNA) for SAα2,6Gal and Maackia amurensis agglutinin (MAA) for SAα2,3Gal in the respiratory tract of normal adults and children. Methods We used fluorescent and biotinylated SNA and MAA from different suppliers on archived and prospectively collected biopsy and autopsy specimens from the nasopharynx, trachea, bronchus and lungs of fetuses, infants and adults. We compared different methods of unmasking for tissue sections to determine if these would affect lectin binding. Using serial sections we then compared the lectin binding of MAA from different suppliers. Results We found that unmasking using microwave treatment in citrate buffer produced increased lectin binding to the ciliated and glandular epithelium of the respiratory tract. In addition we found that there were differences in tissue distribution of the α2,3 linked SA when 2 different isoforms of MAA (MAA1 and MAA2) lectin were used. MAA1 had widespread binding throughout the upper and lower respiratory tract and showed more binding to the respiratory epithelium of children than in adults. By comparison, MAA2 binding was mainly restricted to the alveolar epithelial cells of the lung with weak binding to goblet cells. SNA binding was detected in bronchial and alveolar epithelial cells and binding of this lectin was stronger to the paediatric epithelium compared to adult epithelium. Furthermore, the MAA lectins from 2 suppliers (Roche and EY Labs) tended to only bind in a pattern similar to MAA1 (Vector Labs) and produced a different binding pattern to MAA2 from Vector Labs. Conclusion The lectin binding pattern of MAA may vary depending on the supplier and the different isoforms of MAA show a different tissue distribution in the respiratory tract. This finding is important if conclusions about the potential binding sites of SAα2,3 binding viruses, such as influenza or human parainfluenza are to be made.
Collapse
Affiliation(s)
- John M Nicholls
- Pathology Department, The University of Hong Kong, Pok Fu Lam, Hong Kong, Hong Kong SAR.
| | | | | | | | | |
Collapse
|
460
|
Yao L, Korteweg C, Hsueh W, Gu J. Avian influenza receptor expression in H5N1-infected and noninfected human tissues. FASEB J 2007; 22:733-40. [PMID: 17925493 DOI: 10.1096/fj.06-7880com] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Avian and human influenza viruses preferentially bind to alpha-2,3-linked and alpha-2,6-linked sialic acids, respectively. Until today, the distributions of these two receptor types had never been investigated in H5N1-infected human tissue samples. Here, the expression of avian (AIV-Rs) and human influenza receptors (HuIV-Rs) is studied in various organs (upper and lower respiratory tracts, brain, placenta, liver, kidney, heart, intestines, and spleen) of two H5N1 cases and 14 control cases. Histochemical stains using biotinylated Maackia amurensis lectin II and Sambucus nigra agglutinin were performed to localize AIV-Rs and HuIV-Rs, respectively. Immunohistochemical stainings were performed to identify the receptor-bearing cells. AIV-Rs were detected on type II pneumocytes; a limited number of epithelial cells of the upper respiratory tract; and the bronchi, bronchioli, and trachea; as well as on Kupffer cells, glomerular cells, splenic T cells, and neurons in the brain and intestines. HuIV-Rs were abundantly present in the respiratory tract and lungs. They were also detected on Hofbauer cells, glomerular cells, splenic B cells, and in the liver. Moreover, endothelial cells of all organs examined expressed both receptor types. In conclusion, the distribution pattern of AIV-Rs is partially inconsistent with the pattern of infected cells as detected in previous studies, which suggests there may be other receptors or mechanisms involved in H5N1 infection. In addition, the diffuse presence of receptors on endothelial cells may account for the multiple organ involvement in H5N1 influenza. Finally, the relative lack of AIV-Rs in the upper airway may be a one of the factors preventing efficient human-to-human transmission of H5N1 influenza.
Collapse
Affiliation(s)
- Lu Yao
- Department of Pathology, School of Basic Medical Sciences, Peking (Beijing) University, 38 Xueyuan Rd., 100083 Beijing, China
| | | | | | | |
Collapse
|
461
|
Gu J, Xie Z, Gao Z, Liu J, Korteweg C, Ye J, Lau LT, Lu J, Gao Z, Zhang B, McNutt MA, Lu M, Anderson VM, Gong E, Yu ACH, Lipkin WI. H5N1 infection of the respiratory tract and beyond: a molecular pathology study. Lancet 2007; 370:1137-45. [PMID: 17905166 PMCID: PMC7159293 DOI: 10.1016/s0140-6736(07)61515-3] [Citation(s) in RCA: 285] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
BACKGROUND Human infection with avian influenza H5N1 is an emerging infectious disease characterised by respiratory symptoms and a high fatality rate. Previous studies have shown that the human infection with avian influenza H5N1 could also target organs apart from the lungs. METHODS We studied post-mortem tissues of two adults (one man and one pregnant woman) infected with H5N1 influenza virus, and a fetus carried by the woman. In-situ hybridisation (with sense and antisense probes to haemagglutinin and nucleoprotein) and immunohistochemistry (with monoclonal antibodies to haemagglutinin and nucleoprotein) were done on selected tissues. Reverse-transcriptase (RT) PCR, real-time RT-PCR, strand-specific RT-PCR, and nucleic acid sequence-based amplification (NASBA) detection assays were also undertaken to detect viral RNA in organ tissue samples. FINDINGS We detected viral genomic sequences and antigens in type II epithelial cells of the lungs, ciliated and non-ciliated epithelial cells of the trachea, T cells of the lymph node, neurons of the brain, and Hofbauer cells and cytotrophoblasts of the placenta. Viral genomic sequences (but no viral antigens) were detected in the intestinal mucosa. In the fetus, we found viral sequences and antigens in the lungs, circulating mononuclear cells, and macrophages of the liver. The presence of viral sequences in the organs and the fetus was also confirmed by RT-PCR, strand-specific RT-PCR, real-time RT-PCR, and NASBA. INTERPRETATION In addition to the lungs, H5N1 influenza virus infects the trachea and disseminates to other organs including the brain. The virus could also be transmitted from mother to fetus across the placenta.
Collapse
Affiliation(s)
- Jiang Gu
- Infectious Disease Centre, Peking University, Beijing, China.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
462
|
Zeng H, Goldsmith C, Thawatsupha P, Chittaganpitch M, Waicharoen S, Zaki S, Tumpey TM, Katz JM. Highly pathogenic avian influenza H5N1 viruses elicit an attenuated type i interferon response in polarized human bronchial epithelial cells. J Virol 2007; 81:12439-49. [PMID: 17855549 PMCID: PMC2169033 DOI: 10.1128/jvi.01134-07] [Citation(s) in RCA: 138] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The unparalleled spread of highly pathogenic avian influenza A (HPAI) H5N1 viruses has resulted in devastating outbreaks in domestic poultry and sporadic human infections with a high fatality rate. To better understand the mechanism(s) of H5N1 virus pathogenesis and host responses in humans, we utilized a polarized human bronchial epithelial cell model that expresses both avian alpha-2,3- and human alpha-2,6-linked sialic acid receptors on the apical surface and supports productive replication of both H5N1 and H3N2 viruses. Using this model, we compared the abilities of selected 2004 HPAI H5N1 viruses isolated from humans and a recent human H3N2 virus to trigger the type I interferon (IFN) response. H5N1 viruses elicited significantly less IFN regulatory factor 3 (IRF3) nuclear translocation, as well as delayed and reduced production of IFN-beta compared with the H3N2 virus. Furthermore, phosphorylation of Stat2 and induction of IFN-stimulated genes (ISGs), such as MX1, ISG15, IRF7, and retinoic acid-inducible gene I, were substantially delayed and reduced in cells infected with H5N1 viruses. We also observed that the highly virulent H5N1 virus replicated more efficiently and induced a weaker IFN response than the H5N1 virus that exhibited low virulence in mammals in an earlier study. Our data suggest that the H5N1 viruses tested, especially the virus with the high-pathogenicity phenotype, possess greater capability to attenuate the type I IFN response than the human H3N2 virus. The attenuation of this critical host innate immune defense may contribute to the virulence of H5N1 viruses observed in humans.
Collapse
Affiliation(s)
- Hui Zeng
- Immunology and Pathogenesis Branch, Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA
| | | | | | | | | | | | | | | |
Collapse
|
463
|
Mansfield KG. Viral tropism and the pathogenesis of influenza in the Mammalian host. THE AMERICAN JOURNAL OF PATHOLOGY 2007; 171:1089-92. [PMID: 17717138 PMCID: PMC1988860 DOI: 10.2353/ajpath.2007.070695] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Keith G Mansfield
- Harvard Medical School, 1 Pine Hill Dr., Southborough, MA 01772, USA.
| |
Collapse
|
464
|
Sun X, Whittaker GR. Role of the actin cytoskeleton during influenza virus internalization into polarized epithelial cells. Cell Microbiol 2007; 9:1672-82. [PMID: 17578407 DOI: 10.1111/j.1462-5822.2007.00900.x] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The in vivo site of influenza virus infection is a polarized epithelium, and it is well established that the virus preferentially enters from the apical surface of polarized epithelial cells; however, many of the molecular events involved during the endocytosis of the virus into polarized epithelia remain unclear. Here we examined the role of actin microfilaments and the myosin VI motor protein during influenza entry into a panel of polarized and non-polarized cells. By treatment of cells with cytochalasin D and jasplakinolide, we show that influenza virus entry into all the polarized epithelial cells tested requires actin dynamics, with a specific role for the actin cytoskeleton in the process of virus internalization from the plasma membrane. In contrast, influenza could still could efficiently enter and infect all non-polarized cells tested after disruption or stabilization of the actin cytoskeleton. To examine the role of the actin motor protein, myosin VI, we expressed a dominant-negative construct in both polarized and non-polarized cells. Influenza virus infectivity in myosin VI tail mutant-transfected cells was significantly decreased in polarized epithelial cells, but not in non-polarized cells. As a whole, our data suggest indispensable roles of a dynamic actin cytoskeleton for influenza virus entry into polarized epithelial cells, a feature not shared with non-polarized cells.
Collapse
Affiliation(s)
- Xiangjie Sun
- Department of Microbiology & Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | | |
Collapse
|
465
|
Owen RE, Yamada E, Thompson CI, Phillipson LJ, Thompson C, Taylor E, Zambon M, Osborn HMI, Barclay WS, Borrow P. Alterations in receptor binding properties of recent human influenza H3N2 viruses are associated with reduced natural killer cell lysis of infected cells. J Virol 2007; 81:11170-8. [PMID: 17670834 PMCID: PMC2045558 DOI: 10.1128/jvi.01217-07] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Natural killer (NK) cell recognition of influenza virus-infected cells involves hemagglutinin (HA) binding to sialic acid (SA) on activating NK receptors. SA also acts as a receptor for the binding of influenza virus to its target host cells. The SA binding properties of H3N2 influenza viruses have been observed to change during circulation in humans: recent isolates are unable to agglutinate chicken red blood cells and show reduced affinity for synthetic glycopolymers representing SA-alpha-2,3-lactose (3'SL-PAA) and SA-alpha-2,6-N-acetyl lactosamine (6'SLN-PAA) carbohydrates. Here, NK lysis of cells infected with human H3N2 influenza viruses isolated between 1969 and 2003 was analyzed. Cells infected with recent isolates (1999 to 2003) were found to be lysed less effectively than cells infected with older isolates (1969 to 1996). This change occurred concurrently with the acquisition of two new potential glycosylation site motifs in HA. Deletion of the potential glycosylation site motif at 133 to 135 in HA1 from a recent isolate partially restored the agglutination phenotype to a recombinant virus, indicating that the HA-SA interaction is inhibited by the glycosylation modification. Deletion of either of the recently acquired potential glycosylation sites from HA led to increased NK lysis of cells infected with recombinant viruses carrying modified HA. These results indicate that alterations in HA glycosylation may affect NK cell recognition of influenza virus-infected cells in addition to virus binding to host cells.
Collapse
Affiliation(s)
- Rachel E Owen
- Imperial College London, Department of Virology, Division of Investigative Science, Faculty of Medicine, St. Mary's Campus, Norfolk Place, London W2 1PG, United Kingdom.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
466
|
Hidari KIPJ, Shimada S, Suzuki Y, Suzuki T. Binding kinetics of influenza viruses to sialic acid-containing carbohydrates. Glycoconj J 2007; 24:583-90. [PMID: 17624609 DOI: 10.1007/s10719-007-9055-y] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2007] [Revised: 05/31/2007] [Accepted: 06/05/2007] [Indexed: 11/26/2022]
Abstract
To elucidate the molecular mechanisms of transmission of influenza viruses between different host species, such as human and birds, binding properties of sialic acid-containing carbohydrates that are recognized by human and/or avian influenza viruses were characterized by the surface plasmon resonance (SPR) method. Differences in the binding of influenza viruses to three gangliosides were monitored in real-time and correlated with receptor specificity between avian and human viruses. SPR analysis with ganglioside-containing lipid bilayers demonstrated the recognition profile of influenza viruses to not only sialic acid linkages, but also core carbohydrate structures on the basis of equilibrated rate constants. Kinetic analysis showed different binding preferences to gangliosides between avian and human strains. An avian strain bound to Neu5Acalpha2-3nLc4Cer with much slower dissociation rate than its sialyl-linkage analog, Neu5Acalpha2-6nLc4Cer, on the lipid bilayer. In contrast, a human strain bound equally to both gangliosides. An avian strain, but not a human strain, also interacted with GM3 carrying a shorter carbohydrate chain. Our findings demonstrated the remarkable distinction in the binding kinetics of sialic acid-containing carbohydrates between avian and human influenza viruses on the lipid bilayer.
Collapse
Affiliation(s)
- Kazuya I P J Hidari
- Department of Biochemistry, School of Pharmaceutical Sciences, Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Corporation, University of Shizuoka, Shizuoka-shi, Shizuoka, Japan.
| | | | | | | |
Collapse
|
467
|
Cong YL, Pu J, Liu QF, Wang S, Zhang GZ, Zhang XL, Fan WX, Brown EG, Liu JH. Antigenic and genetic characterization of H9N2 swine influenza viruses in China. J Gen Virol 2007; 88:2035-2041. [PMID: 17554038 DOI: 10.1099/vir.0.82783-0] [Citation(s) in RCA: 123] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
As pigs are susceptible to infection with both avian and human influenza A viruses, they have been proposed to be an intermediate host for the generation of pandemic virus through reassortment. Antigenic and genetic characterization was performed for five swine H9N2 influenza viruses isolated from diseased pigs from different farms. The haemagglutinin (HA) antigenicity of swine H9N2 viruses was different from that of chicken H9N2 viruses prevalent in northern China. Genetic analysis revealed that all five isolates had an RLSR motif at the cleavage site of HA, which was different from those of A/duck/Hong Kong/Y280/97 (Dk/HK/Y280/97)-like viruses established in chickens in China. Phylogenetic analyses indicated that the five swine H9N2 viruses formed novel HA and neuraminidase sublineages that were related closely to those of earlier chicken H9 viruses and were also consistent with the extent of the observed antigenic variation. The six internal genes of the isolates possessed H5N1-like sequences, indicating that they were reassortants of H9 and H5 viruses. The present results indicate that avian to porcine interspecies transmission of H9N2 viruses might have resulted in the generation of viruses with novel antigenic and genetic characteristics; therefore, surveillance of swine influenza should be given a high priority.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antigens, Viral/genetics
- Chickens/virology
- China
- Hemagglutinin Glycoproteins, Influenza Virus/genetics
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Humans
- Influenza A Virus, H9N2 Subtype/classification
- Influenza A Virus, H9N2 Subtype/genetics
- Influenza A Virus, H9N2 Subtype/immunology
- Influenza A Virus, H9N2 Subtype/isolation & purification
- Influenza in Birds/transmission
- Influenza in Birds/virology
- Influenza, Human/transmission
- Influenza, Human/virology
- Molecular Sequence Data
- Orthomyxoviridae Infections/transmission
- Orthomyxoviridae Infections/veterinary
- Orthomyxoviridae Infections/virology
- Phylogeny
- Reassortant Viruses/classification
- Reassortant Viruses/genetics
- Reassortant Viruses/immunology
- Reassortant Viruses/isolation & purification
- Sequence Homology, Amino Acid
- Sus scrofa/virology
- Swine
- Swine Diseases/transmission
- Swine Diseases/virology
- Zoonoses/transmission
- Zoonoses/virology
Collapse
Affiliation(s)
- Yan L Cong
- Laboratory of Infectious Diseases, College of Veterinary Medicine, State Key Laboratory for Agrobiotechnology, China Agricultural University, Beijing 100094, PR China
| | - Juan Pu
- Laboratory of Infectious Diseases, College of Veterinary Medicine, State Key Laboratory for Agrobiotechnology, China Agricultural University, Beijing 100094, PR China
| | - Qin F Liu
- Laboratory of Infectious Diseases, College of Veterinary Medicine, State Key Laboratory for Agrobiotechnology, China Agricultural University, Beijing 100094, PR China
| | - Shuai Wang
- Laboratory of Infectious Diseases, College of Veterinary Medicine, State Key Laboratory for Agrobiotechnology, China Agricultural University, Beijing 100094, PR China
| | - Guo Z Zhang
- Laboratory of Infectious Diseases, College of Veterinary Medicine, State Key Laboratory for Agrobiotechnology, China Agricultural University, Beijing 100094, PR China
| | - Xing L Zhang
- Laboratory of Infectious Diseases, College of Veterinary Medicine, State Key Laboratory for Agrobiotechnology, China Agricultural University, Beijing 100094, PR China
| | - Wei X Fan
- China Animal Health and Epidemiology Center, Qingdao 266032, PR China
| | - Earl G Brown
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, ON, Canada
| | - Jin H Liu
- Laboratory of Infectious Diseases, College of Veterinary Medicine, State Key Laboratory for Agrobiotechnology, China Agricultural University, Beijing 100094, PR China
| |
Collapse
|
468
|
Newby CM, Sabin L, Pekosz A. The RNA binding domain of influenza A virus NS1 protein affects secretion of tumor necrosis factor alpha, interleukin-6, and interferon in primary murine tracheal epithelial cells. J Virol 2007; 81:9469-80. [PMID: 17596305 PMCID: PMC1951395 DOI: 10.1128/jvi.00989-07] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Primary differentiated respiratory epithelial cell cultures closely model the in vivo environment and allow for studies of innate immune responses generated specifically by epithelial cells, the primary cell type infected by human influenza A virus strains. We used primary murine tracheal epithelial cell (mTEC) cultures to investigate antiviral and cytokine responses to influenza A virus infection, focusing on the contribution of the RNA binding domain of the NS1 protein. rWSN NS1 R38A replication is attenuated in mTEC cultures; however, viral antigen is detected predominantly in ciliated cells, similar to wild-type virus. NS1 and NS1 R38A proteins display a primarily cytoplasmic localization in infected mTEC cultures. Increased production of tumor necrosis factor alpha, interleukin-6, and beta interferon is observed during rWSN NS1 R38A infection, and cytokines are secreted in a directional manner. Cytokine pretreatment of mTEC cultures and Vero cells suggest that rWSN NS1 R38A is more sensitive to the presence of antiviral/inflammatory cytokines than wild-type virus. Our results demonstrate that the RNA binding domain is a critical regulator of both cytokine production and cytokine sensitivity during influenza A virus infection of primary tracheal epithelial cells.
Collapse
Affiliation(s)
- Celeste M Newby
- Department of Molecular Microbiology, Washington University School of Medicine, 660 S. Euclid Avenue, Campus Box 8230, St. Louis, Missouri 63110, USA
| | | | | |
Collapse
|
469
|
Amonsen M, Smith DF, Cummings RD, Air GM. Human parainfluenza viruses hPIV1 and hPIV3 bind oligosaccharides with alpha2-3-linked sialic acids that are distinct from those bound by H5 avian influenza virus hemagglutinin. J Virol 2007; 81:8341-5. [PMID: 17522226 PMCID: PMC1951310 DOI: 10.1128/jvi.00718-07] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We investigated the binding of human parainfluenza virus types 1 and 3 (hPIV1 and hPIV3, respectively) to the glycan array of the Consortium for Functional Glycomics and binding and their release from erythrocytes under conditions where neuraminidase is inactive or active. hPIV1 and hPIV3 bind modifications of Neu5Acalpha2-3Galbeta1-4GlcNAc, including the sialyl-Lewis(x) motif and structures containing 6-sulfogalactose. hPIV1 and hPIV3 thus bind typical N-linked glycans, in contrast to avian influenza virus H5 hemagglutinin (J. Stevens, O. Blixt, T. M. Tumpey, J. K. Taubenberger, J. C. Paulson, and I. A. Wilson, Science 312:404-410, 2006), which binds less-common motifs. While the receptor is not the sole determinant of tropism, hPIV or H5 influenza virus infection of specific cells that express receptors may contribute to their different pathologies.
Collapse
Affiliation(s)
- Mary Amonsen
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, P.O. Box 26901, Oklahoma City, OK 73190, USA
| | | | | | | |
Collapse
|
470
|
Bragstad K, Jørgensen PH, Handberg K, Hammer AS, Kabell S, Fomsgaard A. First introduction of highly pathogenic H5N1 avian influenza A viruses in wild and domestic birds in Denmark, Northern Europe. Virol J 2007; 4:43. [PMID: 17498292 PMCID: PMC1876802 DOI: 10.1186/1743-422x-4-43] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2007] [Accepted: 05/11/2007] [Indexed: 11/11/2022] Open
Abstract
Background Since 2005 highly pathogenic (HP) avian influenza A H5N1 viruses have spread from Asia to Africa and Europe infecting poultry, humans and wild birds. HP H5N1 virus was isolated in Denmark for the first time in March 2006. A total of 44 wild birds were found positive for the HP H5N1 infection. In addition, one case was reported in a backyard poultry flock. Results Full-genome characterisation of nine isolates revealed that the Danish H5N1 viruses were highly similar to German H5N1 isolates in all genes from the same time period. The haemagglutinin gene grouped phylogenetically in H5 clade 2 subclade 2 and closest relatives besides the German isolates were isolates from Croatia in 2005, Nigeria and Niger in 2006 and isolates from Astrakhan in Russia 2006. The German and Danish isolates shared unique substitutions in the NA, PB1 and NS2 proteins. Conclusion The first case of HP H5N1 infection of wild and domestic birds in Denmark was experienced in March 2006. This is the first full genome characterisation of HP H5N1 avian influenza A virus in the Nordic countries. The Danish viruses from this time period have their origin from the wild bird strains from Qinghai in 2005. These viruses may have been introduced to the Northern Europe through unusual migration due to the cold weather in Eastern Europe at that time.
Collapse
Affiliation(s)
- Karoline Bragstad
- Laboratory for Virus Research and Development, Statens Serum Institut, Artillerivej 5, DK-2300 Copenhagen S, Denmark
| | - Poul H Jørgensen
- National Veterinary Institute, Technical University of Denmark, Hangøvej 2, DK-8200 Aarhus N, Denmark
| | - Kurt Handberg
- National Veterinary Institute, Technical University of Denmark, Hangøvej 2, DK-8200 Aarhus N, Denmark
| | - Anne S Hammer
- National Veterinary Institute, Technical University of Denmark, Hangøvej 2, DK-8200 Aarhus N, Denmark
| | - Susanne Kabell
- National Veterinary Institute, Technical University of Denmark, Hangøvej 2, DK-8200 Aarhus N, Denmark
| | - Anders Fomsgaard
- Laboratory for Virus Research and Development, Statens Serum Institut, Artillerivej 5, DK-2300 Copenhagen S, Denmark
| |
Collapse
|
471
|
Wan H, Perez DR. Amino acid 226 in the hemagglutinin of H9N2 influenza viruses determines cell tropism and replication in human airway epithelial cells. J Virol 2007; 81:5181-91. [PMID: 17344280 PMCID: PMC1900221 DOI: 10.1128/jvi.02827-06] [Citation(s) in RCA: 229] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2006] [Accepted: 03/01/2007] [Indexed: 11/20/2022] Open
Abstract
Influenza A viruses of the H9N2 subtype are endemic in poultry in many Eurasian countries and have occasionally caused clinical respiratory diseases in humans. While some avian H9N2 viruses have glutamine (Q) at amino acid position 226 of the hemagglutinin (HA) receptor-binding site, an increasing number of isolates have leucine (L) at this position, which has been associated with the establishment of stable lineages of the H2 and H3 subtypes of viruses in humans. Little is known about the importance of this molecular trait in the infection of H9N2 viruses in humans. We show here that during the course of a single cycle of infection in human airway epithelial (HAE) cells cultured in vitro, the L-226-containing H9N2 viruses displayed human virus-like cell tropisms (preferentially infecting nonciliated cells) different from the tropisms showed by Q-226-containing H9N2 isolates (which infect both ciliated and nonciliated cells at ratios of 1:1 to 3:2) or other waterfowl viruses (which preferentially infect ciliated cells). During multiple cycles of replication in HAE cultures, L-226-containing H9N2 isolates grew consistently more efficiently and reached approximately 100-fold-higher peak titers than those containing Q-226, although peak titers were significantly lower than those induced by human H3N2 viruses. Our results suggest that the variation in residue 226 in the HA affects both cell tropism and replication of H9N2 viruses in HAE cells and may have implications for the abilities of these viruses to infect humans.
Collapse
Affiliation(s)
- Hongquan Wan
- Department of Veterinary Medicine, University of Maryland, College Park, 8075 Greenmead Drive, College Park, MD 20742, USA
| | | |
Collapse
|
472
|
Yen HL, Lipatov AS, Ilyushina NA, Govorkova EA, Franks J, Yilmaz N, Douglas A, Hay A, Krauss S, Rehg JE, Hoffmann E, Webster RG. Inefficient transmission of H5N1 influenza viruses in a ferret contact model. J Virol 2007; 81:6890-8. [PMID: 17459930 PMCID: PMC1933302 DOI: 10.1128/jvi.00170-07] [Citation(s) in RCA: 124] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The abilities to infect and transmit efficiently among humans are essential for a novel influenza A virus to cause a pandemic. To evaluate the pandemic potential of widely disseminated H5N1 influenza viruses, a ferret contact model using experimental groups comprised of one inoculated ferret and two contact ferrets was used to study the transmissibility of four human H5N1 viruses isolated from 2003 to 2006. The effects of viral pathogenicity and receptor binding specificity (affinity to synthetic sialosaccharides with alpha2,3 or alpha2,6 linkages) on transmissibility were assessed. A/Vietnam/1203/04 and A/Vietnam/JP36-2/05 viruses, which possess "avian-like" alpha2,3-linked sialic acid (SA) receptor specificity, caused neurological symptoms and death in ferrets inoculated with 10(3) 50% tissue culture infectious doses. A/Hong Kong/213/03 and A/Turkey/65-596/06 viruses, which show binding affinity for "human-like" alpha2,6-linked SA receptors in addition to their affinity for alpha2,3-linked SA receptors, caused mild clinical symptoms and were not lethal to the ferrets. No transmission of A/Vietnam/1203/04 or A/Turkey/65-596/06 virus was detected. One contact ferret developed neutralizing antibodies to A/Hong Kong/213/03 but did not exhibit any clinical signs or detectable virus shedding. In two groups, one of two naïve contact ferrets had detectable virus after 6 to 8 days when housed together with the A/Vietnam/JP36-2/05 virus-inoculated ferrets. Infected contact ferrets showed severe clinical signs, although little or no virus was detected in nasal washes. This limited virus shedding explained the absence of secondary transmission from the infected contact ferret to the other naïve ferret that were housed together. Our results suggest that despite their receptor binding affinity, circulating H5N1 viruses retain molecular determinants that restrict their spread among mammalian species.
Collapse
Affiliation(s)
- Hui-Ling Yen
- Department of Infectious Diseases, St. Jude Children's Research Hospital, 332 N. Lauderdale, Memphis, TN 38105, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
473
|
Sims AC, Burkett SE, Yount B, Pickles RJ. SARS-CoV replication and pathogenesis in an in vitro model of the human conducting airway epithelium. Virus Res 2007; 133:33-44. [PMID: 17451829 PMCID: PMC2384224 DOI: 10.1016/j.virusres.2007.03.013] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2006] [Revised: 03/15/2007] [Accepted: 03/15/2007] [Indexed: 01/12/2023]
Abstract
SARS coronavirus (SARS-CoV) emerged in 2002 as an important cause of severe lower respiratory tract infection in humans and in vitro models of the lung are needed to elucidate cellular targets and the consequences of viral infection. The severe and sudden onset of symptoms, resulting in an atypical pneumonia with dry cough and persistent high fever in cases of severe acute respiratory virus brought to light the importance of coronaviruses as potentially lethal human pathogens and the identification of several zoonotic reservoirs has made the reemergence of new strains and future epidemics all the more possible. In this chapter, we describe the pathology of SARS-CoV infection in humans and explore the use of two models of the human conducting airway to develop a better understanding of the replication and pathogenesis of SARS-CoV in relevant in vitro systems. The first culture model is a human bronchial epithelial cell line Calu-3 that can be inoculated by viruses either as a non-polarized monolayer of cells or polarized cells with tight junctions and microvilli. The second model system, derived from primary cells isolated from human airway epithelium and grown on Transwells, form a pseudostratified mucociliary epithelium that recapitulates the morphological and physiological features of the human conducting airway in vivo. Experimental results using these lung epithelial cell models demonstrate that in contrast to the pathology reported in late stage cases SARS-CoV replicates to high titers in epithelial cells of the conducting airway. The SARS-CoV receptor, human angiotensin 1 converting enzyme 2 (hACE2), was detected exclusively on the apical surface of cells in polarized Calu-3 cells and human airway epithelial cultures (HAE), indicating that hACE2 was accessible by SARS-CoV after lumenal airway delivery. Furthermore, in HAE, hACE2 was exclusively localized to ciliated airway epithelial cells. In support of the hACE2 localization data, the most productive route of inoculation and progeny virion egress in both polarized Calu-3 and ciliated cells of HAE was the apical surface suggesting mechanisms to release large quantities of virus into the lumen of the human lung. Preincubation of the apical surface of cultures with antisera directed against hACE2 reduced viral titers by two logs while antisera against DC-SIGN/DC-SIGNR did not reduce viral replication levels suggesting that hACE2 is the primary receptor for entry of SARS-CoV into the ciliated cells of HAE cultures. To assess infectivity in ciliated airway cultures derived from susceptible animal species we generated a recombinant SARS-CoV by deletion of open reading frame 7a/7b (ORF 7a/7b) and insertion of the green fluorescent protein (GFP) resulting in SARS-CoV GFP. SARS-CoV GFP replicated to similar titers as wild type viruses in Vero E6, MA104, and CaCo2 cells. In addition, SARS-CoV replication in airway epithelial cultures generated from Golden Syrian hamster tracheas reached similar titers to the human cultures by 72 h post-infection. Efficient SARS-CoV infection of ciliated cell-types in HAE provides a useful in vitro model of human lung origin to study characteristics of SARS-CoV replication and pathogenesis.
Collapse
Affiliation(s)
- Amy C Sims
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States.
| | | | | | | |
Collapse
|
474
|
Zhirnov OP, Vorobjeva IV, Saphonova OA, Malyshev NA, Ovcharenko AV, Klenk HD. Specific biochemical features of replication of clinical influenza viruses in human intestinal cell culture. BIOCHEMISTRY (MOSCOW) 2007; 72:398-408. [PMID: 17511604 DOI: 10.1134/s0006297907040062] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Influenza A viruses isolated from the respiratory tract of patients with influenza were cultured in human intestinal epithelium cells (CACO-2 line). The CACO-2 cells were found to be 100-fold more susceptible to the clinical viruses than MDCK cells and chicken embryos. On passaging in CACO-2 cells, clinical isolates of the subtype H3N2 retained the original "human" phenotype and agglutinated human but not chicken erythrocytes, whereas on passaging in MDCK cells the virus phenotype changed to the "avian" one. On comparison with laboratory strains (grown in chicken embryos or MDCK cells), the clinical viruses were characterized by higher stability of the anti-interferon protein NS1 but had a reduced synthesis of the matrix protein M1, and this could facilitate the virus adaptation and escape of the infected cells from immune attack in the human body. The increased tropism to the human CACO-2 cells correlated with higher adsorption of the clinical viruses on cellular receptors. However, in the CACO-2 and MDCK cells the ratio of sialyl-containing glycoreceptors of the 2-3 and 2-6 type was similar. These observations indicated that not only sialic acid residues were involved in the adsorption and penetration of the clinical viruses into human cells, but also the protein moiety of the cellular receptor itself and/or an additional cellular coreceptor. Thus, clinical influenza viruses are shown to possess a specific mechanism of sorption and entry into human epithelial cells, which is responsible for their higher tropism to human cells and is unlike such a mechanism in canine cells.
Collapse
Affiliation(s)
- O P Zhirnov
- Ivanovsky Institute of Virology, Russian Academy of Medical Sciences, ul. Gamalei 16, 123098 Moscow, Russia.
| | | | | | | | | | | |
Collapse
|
475
|
Guo CT, Takahashi N, Yagi H, Kato K, Takahashi T, Yi SQ, Chen Y, Ito T, Otsuki K, Kida H, Kawaoka Y, Hidari KIPJ, Miyamoto D, Suzuki T, Suzuki Y. The quail and chicken intestine have sialyl-galactose sugar chains responsible for the binding of influenza A viruses to human type receptors. Glycobiology 2007; 17:713-24. [PMID: 17389652 DOI: 10.1093/glycob/cwm038] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The receptor specificity of influenza viruses is one factor that allows avian influenza viruses to cross the species barrier. The recent transmissions of avian H5N1 and H9N2 influenza viruses from chickens and/or quails to humans indicate that avian influenza viruses can directly infect humans without an intermediate host, such as pigs. In this study, we used two strains of influenza A virus (A/PR/8/34, which preferentially binds to an avian-type receptor, and A/Memphis/1/71, which preferentially binds to a human-type receptor) to probe the receptor specificities in host cells. Epithelial cells of both quail and chicken intestines (colons) could bind both avian- and human-type viruses. Infected cultured quail colon cells expressed viral protein and allowed replication of the virus strain A/PR/8/34 or A/Memphis/1/71. To understand the molecular basis of these phenomena, we further investigated the abundance of sialic acid (Sia) linked to galactose (Gal) by the alpha2-3 linkage (Siaalpha2-3Gal) and Siaalpha2-6Gal in host cells. In glycoprotein and glycolipid fractions from quail and chicken colon epithelial cells, there were some bound components of Sia-Gal linkage-specific lectins, Maackia amurensis agglutinin (specific for Siaalpha2-3 Gal) and Sambucus nigra agglutinin (specific for Siaalpha2-6Gal), indicating that both Siaalpha2-3Gal and Siaalpha2-6Gal exist in quail and chicken colon cells. Furthermore, we demonstrated by fluorescence high-performance liquid chromatography (HPLC) analysis that 5-N-acetylneuraminic acid was the main molecular species of Sia, and we demonstrated by multi-dimensional HPLC mapping and matrix-assisted laser desorption/ionization time-of-flight mass spectrometry analysis that bi-antennary complex-type glycans alpha2-6 sialylated at the terminal Gal residue(s) are major (more than 79%) sialyl N-glycans expressed by intestinal epithelial tissues in both the chicken and quail. Taken together, these results indicate that quails and chickens have molecular characterization as potential intermediate hosts for avian influenza virus transmission to humans and could generate new influenza viruses with pandemic potential.
Collapse
Affiliation(s)
- Chao-Tan Guo
- Institute of Bioengineering, Zhejiang Academy of Medical Sciences, 182 Tianmushan Road, Hangzhou 310016, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
476
|
Weber S, Harder T, Starick E, Beer M, Werner O, Hoffmann B, Mettenleiter TC, Mundt E. Molecular analysis of highly pathogenic avian influenza virus of subtype H5N1 isolated from wild birds and mammals in northern Germany. J Gen Virol 2007; 88:554-558. [PMID: 17251574 DOI: 10.1099/vir.0.82300-0] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Analysis of the full-length sequences of all eight segments of the German wild-bird H5N1 highly pathogenic avian influenza virus index isolate, A/Cygnus cygnus/Germany/R65/2006, and an H5N1 isolate from a cat (A/cat/Germany/R606/2006) obtained during an outbreak in February 2006 revealed a very high similarity between these two sequences. One amino acid substitution in the PA gene, encoding a protein involved in virus RNA replication, and one amino acid substitution in the haemagglutinin (HA) protein were observed. Phylogenetic analyses of the HA and neuraminidase nucleotide sequences showed that avian influenza H5N1 isolates from the Astrakhan region located in southern Russia were the closest relatives. Reassortment events could be excluded in comparison with other 'Qinghai-like' H5N1 viruses. In addition, an H5N1 isolate originating from a single outbreak in poultry in Germany was found to be related closely to the H5N1 viruses circulating at that time in the wild-bird population.
Collapse
Affiliation(s)
- Siegfried Weber
- Institute of Molecular Biology, Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany
| | - Timm Harder
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany
| | - Elke Starick
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany
| | - Martin Beer
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany
| | - Ortrud Werner
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany
| | - Bernd Hoffmann
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany
| | - Thomas C Mettenleiter
- Institute of Molecular Biology, Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany
| | - Egbert Mundt
- Institute of Molecular Biology, Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany
| |
Collapse
|
477
|
Stuart AD, Brown TDK. Alpha2,6-linked sialic acid acts as a receptor for Feline calicivirus. J Gen Virol 2007; 88:177-186. [PMID: 17170450 DOI: 10.1099/vir.0.82158-0] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Feline calicivirus (FCV) is a major causative agent of respiratory disease in cats. It is also one of the few cultivatable members of the family Caliciviridae. It has recently been reported that FCV binding is in part due to interaction with junction adhesion molecule-A. This report describes the characterization of additional receptor components for FCV. Chemical treatment of cells with sodium periodate showed that FCV recognized carbohydrate moieties on the surface of permissive cells. Enzymic treatment with Vibrio cholerae neuraminidase demonstrated that sialic acid was a major determinant of virus binding. Further characterization using linkage-specific lectins from Maackia amurensis and Sambucus nigra revealed that FCV recognized sialic acid with an alpha2,6 linkage. Using various proteases and metabolic inhibitors, it was shown that alpha2,6-linked sialic acid recognized by FCV is present on an N-linked glycoprotein.
Collapse
Affiliation(s)
- Amanda D Stuart
- Division of Virology, Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK
| | - T David K Brown
- Division of Virology, Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK
| |
Collapse
|
478
|
Glaser L, Conenello G, Paulson J, Palese P. Effective replication of human influenza viruses in mice lacking a major alpha2,6 sialyltransferase. Virus Res 2007; 126:9-18. [PMID: 17313986 DOI: 10.1016/j.virusres.2007.01.011] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2006] [Revised: 01/11/2007] [Accepted: 01/13/2007] [Indexed: 12/22/2022]
Abstract
The hemagglutinins of influenza viruses isolated from humans typically prefer binding to sialic acid in an alpha2,6 linkage. Presumably, the virus uses the presence of these receptors on the respiratory tract to gain entrance into the host cell. The ST6Gal I sialyltransferase knock-out mouse lacks the main enzyme necessary for the attachment of alpha2,6 sialic acid to N-linked glycoproteins on the cell surface. Yet even in the absence of detectable alpha2,6 sialic acid in the mouse respiratory tract, human influenza viruses can still infect these mice and grow to similar titers in the lung and trachea as compared to wild-type animals. This work demonstrates that the presence of a major alpha2,6 sialic acid on N-linked glycoproteins is not essential for human influenza virus infection in mice.
Collapse
Affiliation(s)
- Laurel Glaser
- Department of Microbiology, Mount Sinai School of Medicine, New York, NY 10029, USA.
| | | | | | | |
Collapse
|
479
|
Lee DCW, Lau ASY. Avian influenza virus signaling: implications for the disease severity of H5N1 infection. ACTA ACUST UNITED AC 2007. [DOI: 10.1002/sita.200600117] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
480
|
Matrosovich M, Matrosovich T, Uhlendorff J, Garten W, Klenk HD. Avian-virus-like receptor specificity of the hemagglutinin impedes influenza virus replication in cultures of human airway epithelium. Virology 2007; 361:384-90. [PMID: 17207830 DOI: 10.1016/j.virol.2006.11.030] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2006] [Revised: 10/04/2006] [Accepted: 11/27/2006] [Indexed: 11/26/2022]
Abstract
A non-optimal receptor-binding specificity of avian influenza viruses is believed to hamper their replication in humans; however, the magnitude of this restriction remains undefined. Here we generated recombinant viruses, R1 and R2, that differed solely by two amino acids in the receptor-binding site of their hemagglutinin (HA). R1 harbored the original HA of the pandemic human virus A/Hong Kong/1/68 (H3N2), whereas R2 was the L226Q/S228G HA mutant with avian-virus-like receptor specificity. In differentiated cultures of human tracheo-bronchial epithelial cells, R1 preferentially infected non-ciliated cells, whereas R2 predominantly infected ciliated cells indicating that cell tropism was determined by the viral receptor specificity. In the course of multi-cycle replication in these cultures, R2 spread less efficiently and grew to 2-10-fold lower titers than did R1. These results for the first time estimate the level of receptor-dependent restriction of avian influenza viruses in human airway epithelium. They support a theory that alteration of the receptor specificity of an avian virus could facilitate its human-to-human transmission.
Collapse
|
481
|
Nicholls JM, Chan MCW, Chan WY, Wong HK, Cheung CY, Kwong DLW, Wong MP, Chui WH, Poon LLM, Tsao SW, Guan Y, Peiris JSM. Tropism of avian influenza A (H5N1) in the upper and lower respiratory tract. Nat Med 2007; 13:147-9. [PMID: 17206149 DOI: 10.1038/nm1529] [Citation(s) in RCA: 254] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2006] [Accepted: 12/04/2006] [Indexed: 02/04/2023]
Abstract
Poor human-to-human transmission of influenza A H5N1 virus has been attributed to the paucity of putative sialic acid alpha2-3 virus receptors in the epithelium of the human upper respiratory tract, and thus to the presumed inability of the virus to replicate efficiently at this site. We now demonstrate that ex vivo cultures of human nasopharyngeal, adenoid and tonsillar tissues can be infected with H5N1 viruses in spite of an apparent lack of these receptors.
Collapse
Affiliation(s)
- J M Nicholls
- Department of Pathology, University of Hong Kong, Pok Fu Lam Road, Hong Kong SAR, China.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
482
|
Update on Avian Influenza for Critical Care Physicians. Intensive Care Med 2007. [PMCID: PMC7122579 DOI: 10.1007/978-0-387-49518-7_90] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
Human influenza pandemics over the last 100 years have been caused by H1, H2, and H3 subtypes of influenza A viruses. More recently, avian influenza viruses have been found to directly infect humans from their avian hosts. The recent emergence, host expansion, and spread of a highly pathogenic avian influenza (HPAI) H5N1 subtype in Asia has heightened concerns globally, both in regards to mortality of HPAI H5N1 in humans and the potential of a new pandemic. In response, many agencies and organizations have been working collaboratively to develop early detection systems, preparedness plans, and objectives for further research. As a result, there has been a large influx of published information regarding potential risk, surveillance, prevention and control of highly pathogenic avian influenza, particularly in regards to animal to human and subsequent human to human transmission. This chapter will review the current human infections with avian influenza and its public health and medical implications.
Collapse
|
483
|
Bermingham A, Henrickson K, Hayden F, Zambon M. VII International Symposium on Respiratory Viral Infections. Antivir Ther 2007. [DOI: 10.1177/135965350701200s09.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The VII International Symposium on Respiratory Viral Infections was a multidisciplinary forum for the presentation of recent advances in respiratory virus research with special emphasis on antiviral therapies and vaccine strategies. Topics covered in invited lectures included detection of novel respiratory viral pathogens and viral evolution, characterization of the 1918 pandemic virus, human metapneumovirus infections, human respiratory epithelial cultures for studying viral pathogenesis, the role of respiratory viruses in the pathogenesis of asthma, influenza-bacterial interactions, advances in generating vaccine candidates against global respiratory threats like avian influenza and SARS, antiviral resistance surveillance in influenza viruses, and a mini-symposium on advances in viral diagnostics. Other talks covered the live, attenuated intranasal influenza vaccine, monoclonals for respiratory syncytial virus (RSV), mechanisms of antiviral resistance in influenza B, and novel inhibitors for influenza, RSV and rhinovirus infections.
Collapse
Affiliation(s)
| | | | - Frederick Hayden
- University of Virginia School of Medicine, Charlottesville, VA, USA and Global Influenza Program, World Health Organization, Geneva, Switzerland
| | | |
Collapse
|
484
|
Developments in Avian Influenza Virus Vaccines. J Poult Sci 2007. [DOI: 10.2141/jpsa.44.238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
485
|
Abstract
Continuing occurrences of human infections with avian influenza A (H5N1) viruses have ignited increasing fears that the next influenza pandemic is imminent. Fortunately, options for antiviral prophylaxis and treatment have been improved dramatically since the previous pandemics by the availability of neuraminidase inhibitors such as zanamivir and oseltamivir. However, although the prophylactic and therapeutic efficacy of these drugs is well established for uncomplicated seasonal human influenza, clinical effectiveness seems limited for human H5N1 infections despite in vitro susceptibility and efficacy in animal studies. Factors which might contribute to this apparently limited efficacy include suboptimal dosing or routes of administration, suboptimal timing of treatment and the inability of antiviral drugs to interfere with immunopathology, and the development of drug resistance. Efforts to optimize the use of neuraminidase inhibitor treatment in H5N1 disease are urgently needed and might eventually aid in the judicious use of stockpiled neuraminidase inhibitors in the event of a pandemic.
Collapse
Affiliation(s)
- Martin Crusat
- Oxford University Clinical Research Unit, Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
| | - Menno D De Jong
- Oxford University Clinical Research Unit, Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
| |
Collapse
|
486
|
Abstract
Highly pathogenic H5N1 influenza viruses have become endemic in poultry populations throughout Southeast Asia and continue to infect humans with a greater than 50% case fatality rate. So far, human-to-human transmission of these viruses has been limited. Here, we discuss the molecular features of H5N1 influenza viruses that might affect their pathogenicity, and explain the current lack of efficient human-to-human transmission. Such knowledge is critical in evaluating the pandemic risk these viruses pose.
Collapse
Affiliation(s)
- Gabriele Neumann
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Kyoko Shinya
- The Avian Zoonosis Research Centre, Tottori University, Tottori, Japan
| | - Yoshihiro Kawaoka
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
- Division of Virology, Department of Microbiology and Immunology and International Research Center for Infectious Diseases, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| |
Collapse
|
487
|
Update on Avian Influenza for Critical Care Physicians. YEARBOOK OF INTENSIVE CARE AND EMERGENCY MEDICINE 2007. [PMCID: PMC7123627 DOI: 10.1007/978-3-540-49433-1_90] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Human influenza pandemics over the last 100 years have been caused by H1, H2, and H3 subtypes of influenza A viruses. More recently, avian influenza viruses have been found to directly infect humans from their avian hosts. The recent emergence, host expansion, and spread of a highly pathogenic avian influenza (HPAI) H5N1 subtype in Asia has heightened concerns globally, both in regards to mortality of HPAI H5N1 in humans and the potential of a new pandemic. In response, many agencies and organizations have been working collaboratively to develop early detection systems, preparedness plans, and objectives for further research. As a result, there has been a large influx of published information regarding potential risk, surveillance, prevention and control of highly pathogenic avian influenza, particularly in regards to animal to human and subsequent human to human transmission. This chapter will review the current human infections with avian influenza and its public health and medical implications.
Collapse
|
488
|
Kogure T, Suzuki T, Takahashi T, Miyamoto D, Hidari KIPJ, Guo CT, Ito T, Kawaoka Y, Suzuki Y. Human trachea primary epithelial cells express both sialyl(alpha2-3)Gal receptor for human parainfluenza virus type 1 and avian influenza viruses, and sialyl(alpha2-6)Gal receptor for human influenza viruses. Glycoconj J 2006; 23:101-6. [PMID: 16575527 DOI: 10.1007/s10719-006-5442-z] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
We reported previously that the dominant receptors of influenza A and B viruses, and human and murine respiroviruses, were sialylglycoproteins and gangliosides containing monosialo-lactosamine type I-and II-residues, such as sialic acid-alpha2-3(6)-Galbeta1-3(4)-GlcNAcbeta1-. In addition, the Siaalpha2-3Gal linkage was predominantly recognized by avian and horse influenza viruses, and human parainfluenza virus type 1 (hPIV-1), whereas the Siaalpha2-6Gal linkage was mainly recognized by human influenza viruses (Paulson JC in "The Receptors'' [Conn M Ed] 2, 131-219 (1985); Suzuki Y, Prog Lipid Res 33, 429-57 (1994); Ito T, J Virol 73, 6743-51 (2000); Suzuki Y, J Virol 74, 11825-31 (2000); Suzuki T, J. Virol 75, 4604-4613 (2001); Suzuki Y, Biol. Pharm. Bull. 28, 399-408 (2005)). To clarify the distribution of influenza virus receptors on the human bronchial epithelium cell surface, we investigated a primary culture of normal human bronchial epithelial (NHBE) cells using two types of lectin (MAA and SNA), which recognize sialyl linkages (alpha2-3 and alpha2-6), using fluorescence-activated cell-sorting analysis. The results showed that both alpha2-3- and alpha2-6-linked Sias were expressed on the surface of primary human bronchial epithelial cells. The cells infected by hPIV-1 bound to MAA, confirming that cells targeted by hPIV-1 have alpha2-3-linked oligosaccharides. We also compared the ability of hPIV-1 and human influenza A virus to infect primary human bronchial epithelial cells pre-treated with Siaalpha2-3Gal-specific sialidase from Salmonella typhimurium. No difference was observed in the number of sialidase pre-treated and non-treated cells infected with human influenza A virus, which binds to Siaalpha2-6Gal-linked oligosaccharides. By contrast, the number of cells infected with hPIV-1 decreased significantly upon sialidase treatment. Thus, cultured NHBE cells showed both alpha2-3-linked Sias recognized by hPIV-1 and avian influenza virus receptors, and alpha2-6-linked Sias recognized by human influenza virus receptors.
Collapse
Affiliation(s)
- Toshihiro Kogure
- Department of Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka 422-8526, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
489
|
Banet-Noach C, Panshin A, Golender N, Simanov L, Rozenblut E, Pokamunski S, Pirak M, Tendler Y, García M, Gelman B, Pasternak R, Perk S. Genetic analysis of nonstructural genes (NS1 and NS2) of H9N2 and H5N1 viruses recently isolated in Israel. Virus Genes 2006; 34:157-68. [PMID: 17171546 DOI: 10.1007/s11262-006-0057-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2006] [Accepted: 11/10/2006] [Indexed: 10/23/2022]
Abstract
The avian influenza virus subtype H9N2 affects wild birds, domestic poultry, swine, and humans; it has circulated amongst domestic poultry in Israel during the last 6 years. The H5N1 virus was recorded in Israel for the first time in March 2006. Nonstructural (NS) genes and NS proteins are important in the life cycle of the avian influenza viruses. In the present study, NS genes of 21 examples of H9N2 and of two examples of H5N1 avian influenza viruses, isolated in Israel during 2000-2006, were completely sequenced and phylogenetically analyzed. All the H9N2 isolates fell into a single group that, in turn, was subdivided into three subgroups in accordance with the time of isolation; their NS1 and NS2 proteins possessed 230 and 121 amino acids, respectively. The NS1 protein of the H5N1 isolates had five amino acid deletions, which was typical of highly pathogenic H5N1 viruses isolated in various countries during 2005-2006. Comparative analysis showed that the NS proteins of the H9N2 Israeli isolates contained few amino acid sequences associated with high pathogenicity or human host specificity.
Collapse
Affiliation(s)
- Caroline Banet-Noach
- Division of Avian and Aquatic Diseases, Kimron Veterinary Institute, P.O.B. 12, Beit Dagan, ZC, 50250, Israel
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
490
|
Opinion of the Scientific Panel on Animal Health and Welfare (AHAW) on a request from the Commission related with animal health and welfare risks associated with the import of wild birds other than poultry into the European Union. EFSA J 2006; 4:410. [PMID: 32313576 PMCID: PMC7163484 DOI: 10.2903/j.efsa.2006.410] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
|
491
|
Böttcher E, Matrosovich T, Beyerle M, Klenk HD, Garten W, Matrosovich M. Proteolytic activation of influenza viruses by serine proteases TMPRSS2 and HAT from human airway epithelium. J Virol 2006; 80:9896-8. [PMID: 16973594 PMCID: PMC1617224 DOI: 10.1128/jvi.01118-06] [Citation(s) in RCA: 372] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Host cell proteases that cleave the hemagglutinin (HA) of influenza viruses in the human respiratory tract are still not identified. Here we cloned two human type II transmembrane serine proteases with known airway localization, TMPRSS2 and HAT, into mammalian expression vector. Cotransfection of mammalian cells with plasmids encoding HA and either protease resulted in HA cleavage in situ. Transient expression of either protease in MDCK cells enabled multicycle replication of influenza viruses in these cells in the absence of exogenous trypsin. These data suggest that TMPRSS2 and HAT are candidates for proteolytic activation of influenza viruses in vivo.
Collapse
Affiliation(s)
- Eva Böttcher
- Institute of Virology, Philipps University, Hans-Meerwein str. 3, 35043 Marburg, Germany
| | | | | | | | | | | |
Collapse
|
492
|
Wu Z, Miller E, Agbandje-McKenna M, Samulski RJ. Alpha2,3 and alpha2,6 N-linked sialic acids facilitate efficient binding and transduction by adeno-associated virus types 1 and 6. J Virol 2006; 80:9093-103. [PMID: 16940521 PMCID: PMC1563919 DOI: 10.1128/jvi.00895-06] [Citation(s) in RCA: 222] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Recombinant adeno-associated viruses (AAVs) are promising vectors in the field of gene therapy. Different AAV serotypes display distinct tissue tropism, believed to be related to the distribution of their receptors on target cells. Of the 11 well-characterized AAV serotypes, heparan sulfate proteoglycan and sialic acid have been suggested to be the attachment receptors for AAV type 2 and types 4 and 5, respectively. In this report, we identify the receptor for the two closely related serotypes, AAV1 and AAV6. First, we demonstrate using coinfection experiments and luciferase reporter analysis that AAV1 and AAV6 compete for similar receptors. Unlike heparin sulfate, enzymatic or genetic removal of sialic acid markedly reduced AAV1 and AAV6 binding and transduction. Further analysis using lectin staining and lectin competition assays identified that AAV1 and AAV6 use either alpha2,3-linked or alpha2,6-linked sialic acid when transducing numerous cell types (HepG2, Pro-5, and Cos-7). Treatment of cells with proteinase K but not glycolipid inhibitor reduced AAV1 and AAV6 infection, supporting the hypothesis that the sialic acid that facilitates infection is associated with glycoproteins rather than glycolipids. In addition, we determined by inhibitor (N-benzyl GalNAc)- and cell line-specific (Lec-1) studies that AAV1 and AAV6 require N-linked and not O-linked sialic acid. Furthermore, a resialylation experiment on a deficient Lec-2 cell line confirmed a 2,3 and 2,6 N-linked sialic acid requirement, while studies of mucin with O-linked sialic acid showed no inhibition effect for AAV1 and AAV6 transduction on Cos-7 cells. Finally, using a glycan array binding assay we determined that AAV1 efficiently binds to NeuAcalpha2-3GalNAcbeta1-4GlcNAc, as well as two glycoproteins with alpha2,3 and alpha2,6 N-linked sialic acids. Taken together, competition, genetic, inhibitor, enzymatic reconstitution, and glycan array experiments support alpha2,3 and alpha2,6 sialic acids that are present on N-linked glycoproteins as primary receptors for efficient AAV1 and AAV6 viral infection.
Collapse
Affiliation(s)
- Zhijian Wu
- Gene Therapy Center, CB # 7352, 7119 Thurston Building, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7352, USA
| | | | | | | |
Collapse
|
493
|
Stevens J, Blixt O, Paulson JC, Wilson IA. Glycan microarray technologies: tools to survey host specificity of influenza viruses. Nat Rev Microbiol 2006; 4:857-64. [PMID: 17013397 PMCID: PMC7097745 DOI: 10.1038/nrmicro1530] [Citation(s) in RCA: 263] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
New technologies are urgently required for rapid surveillance of the current H5N1 avian influenza A outbreaks to gauge the potential for adaptation of the virus to the human population, a crucial step in the emergence of pandemic influenza virus strains. Owing to the species-specific nature of the interaction between the virus and host glycans, attention has recently focused on novel glycan array technologies that can rapidly assess virus receptor specificity and the potential emergence of human-adapted H5N1 viruses.
Collapse
Affiliation(s)
- James Stevens
- Department of Molecular Biology,
- Glycan Array Synthesis Core-D, Consortium for Functional Glycomics,The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, 92037 California USA
| | - Ola Blixt
- Department of Molecular Biology,
- Glycan Array Synthesis Core-D, Consortium for Functional Glycomics,The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, 92037 California USA
| | - James C. Paulson
- Department of Molecular Biology,
- Glycan Array Synthesis Core-D, Consortium for Functional Glycomics,The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, 92037 California USA
| | - Ian A. Wilson
- Department of Molecular Biology,
- Glycan Array Synthesis Core-D, Consortium for Functional Glycomics,The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, 92037 California USA
| |
Collapse
|
494
|
Li G, Tao S, Wang X. Sequence and epitope analysis of surface proteins of avian influenza H5N1 viruses from Asian patients. ACTA ACUST UNITED AC 2006. [DOI: 10.1007/s11434-006-2140-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
495
|
Gillim-Ross L, Subbarao K. Emerging respiratory viruses: challenges and vaccine strategies. Clin Microbiol Rev 2006; 19:614-36. [PMID: 17041137 PMCID: PMC1592697 DOI: 10.1128/cmr.00005-06] [Citation(s) in RCA: 118] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
The current threat of avian influenza to the human population, the potential for the reemergence of severe acute respiratory syndrome (SARS)-associated coronavirus, and the identification of multiple novel respiratory viruses underline the necessity for the development of therapeutic and preventive strategies to combat viral infection. Vaccine development is a key component in the prevention of widespread viral infection and in the reduction of morbidity and mortality associated with many viral infections. In this review we describe the different approaches currently being evaluated in the development of vaccines against SARS-associated coronavirus and avian influenza viruses and also highlight the many obstacles encountered in the development of these vaccines. Lessons learned from current vaccine studies, coupled with our increasing knowledge of the host and viral factors involved in viral pathogenesis, will help to increase the speed with which efficacious vaccines targeting newly emerging viral pathogens can be developed.
Collapse
Affiliation(s)
- Laura Gillim-Ross
- Laboratory of Infectious Diseases, National Insitute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | | |
Collapse
|
496
|
Horimoto T, Kawaoka Y. Strategies for developing vaccines against H5N1 influenza A viruses. Trends Mol Med 2006; 12:506-14. [PMID: 17011235 DOI: 10.1016/j.molmed.2006.09.003] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2006] [Revised: 08/16/2006] [Accepted: 09/15/2006] [Indexed: 11/21/2022]
Abstract
Recent outbreaks of highly pathogenic avian influenza A virus (H5N1 subtype) infections in poultry and humans (through direct contact with infected birds) have raised concerns that a new influenza pandemic might occur in the near future. Effective vaccines against H5N1 virus are, therefore, urgently needed. Reverse-genetics-based inactivated vaccines have been prepared according to World Health Organization (WHO) recommendations and are now undergoing clinical evaluation in several countries. Here, we review the current strategies for the development of H5N1 influenza vaccines, and future directions for vaccine development.
Collapse
Affiliation(s)
- Taisuke Horimoto
- Division of Virology, Department of Microbiology and Immunology, International Research Center for Infectious Diseases, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | | |
Collapse
|
497
|
Asahi-Ozaki Y, Itamura S, Ichinohe T, Strong P, Tamura SI, Takahashi H, Sawa H, Moriyama M, Tashiro M, Sata T, Kurata T, Hasegawa H. Intranasal administration of adjuvant-combined recombinant influenza virus HA vaccine protects mice from the lethal H5N1 virus infection. Microbes Infect 2006; 8:2706-14. [PMID: 16968669 DOI: 10.1016/j.micinf.2006.07.018] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2006] [Revised: 07/25/2006] [Accepted: 07/30/2006] [Indexed: 11/24/2022]
Abstract
Attenuated recombinant H5N1 influenza virus was constructed to develop a safe H5N1 influenza vaccine. The immunogenicity and protective effect of the vaccine prepared from haemagglutinin-modified recombinant H5N1 influenza virus was evaluated in mice intranasally co-administered with cholera toxin B subunit containing a trace amount of holotoxin (CTB*), synthetic double-stranded RNA, poly (I:C) or chitin microparticles (CMP) as adjuvants. Intranasal administration of recombinant H5 HA split vaccine with CTB* or poly(I:C) and/or CMP elicited an immunological response with both anti-H5 HA IgA in the nasal wash and anti-H5 HA IgG antibody in the serum, and showed a protective against lethal H5N1 A/Hong Kong/483/97 (HK483) infection. We also demonstrated that intranasal co-administration of antigen with both poly (I:C) and CMP enhanced the expression of Toll-like receptor (TLR) 3, TLR7 in the spleen. These results indicate that poly (I:C) and CMP are highly effective as mucosal adjuvants for use with the nasal H5N1 vaccine.
Collapse
MESH Headings
- Adjuvants, Immunologic
- Administration, Intranasal
- Animals
- Antibodies, Viral/analysis
- Antibodies, Viral/blood
- Chitin/immunology
- Cholera Toxin/immunology
- Disease Models, Animal
- Female
- Hemagglutinin Glycoproteins, Influenza Virus/genetics
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Immunity, Mucosal
- Immunoglobulin A/analysis
- Immunoglobulin G/blood
- Influenza A Virus, H5N1 Subtype/immunology
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/immunology
- Lymphoid Tissue/immunology
- Mice
- Mice, Inbred BALB C
- Microspheres
- Nasal Mucosa/immunology
- Orthomyxoviridae Infections/prevention & control
- Orthomyxoviridae Infections/virology
- Poly I-C/immunology
- RNA, Double-Stranded/immunology
- Spleen/immunology
- Toll-Like Receptors/biosynthesis
- Vaccines, Subunit/genetics
- Vaccines, Subunit/immunology
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
Collapse
Affiliation(s)
- Yasuko Asahi-Ozaki
- Department of Pathology, National Institute of Infectious Diseases, 4-7-1 Gakuen, Musashimurayama-shi, Tokyo 208-0011, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
498
|
Ibricevic A, Pekosz A, Walter MJ, Newby C, Battaile JT, Brown EG, Holtzman MJ, Brody SL. Influenza virus receptor specificity and cell tropism in mouse and human airway epithelial cells. J Virol 2006; 80:7469-80. [PMID: 16840327 PMCID: PMC1563738 DOI: 10.1128/jvi.02677-05] [Citation(s) in RCA: 299] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Recent human infections caused by the highly pathogenic avian influenza virus H5N1 strains emphasize an urgent need for assessment of factors that allow viral transmission, replication, and intra-airway spread. Important determinants for virus infection are epithelial cell receptors identified as glycans terminated by an alpha2,3-linked sialic acid (SA) that preferentially bind avian strains and glycans terminated by an alpha2,6-linked SA that bind human strains. The mouse is often used as a model for study of influenza viruses, including recent avian strains; however, the selectivity for infection of specific respiratory cell populations is not well described, and any relationship between receptors in the mouse and human lungs is incompletely understood. Here, using in vitro human and mouse airway epithelial cell models and in vivo mouse infection, we found that the alpha2,3-linked SA receptor was expressed in ciliated airway and type II alveolar epithelial cells and was targeted for cell-specific infection in both species. The alpha2,6-linked SA receptor was not expressed in the mouse, a factor that may contribute to the inability of some human strains to efficiently infect the mouse lung. In human airway epithelial cells, alpha2,6-linked SA was expressed and functional in both ciliated and goblet cells, providing expanded cellular tropism. Differences in receptor and cell-specific expression in these species suggest that differentiated human airway epithelial cell cultures may be superior for evaluation of some human strains, while the mouse can provide a model for studying avian strains that preferentially bind only the alpha2,3-linked SA receptor.
Collapse
Affiliation(s)
- Aida Ibricevic
- Department of Internal Medicine, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | | | | | | | | | | | | | | |
Collapse
|
499
|
Baccam P, Beauchemin C, Macken CA, Hayden FG, Perelson AS. Kinetics of influenza A virus infection in humans. J Virol 2006; 80:7590-9. [PMID: 16840338 PMCID: PMC1563736 DOI: 10.1128/jvi.01623-05] [Citation(s) in RCA: 495] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Currently, little is known about the viral kinetics of influenza A during infection within an individual. We utilize a series of mathematical models of increasing complexity, which incorporate target cell limitation and the innate interferon response, to examine influenza A virus kinetics in the upper respiratory tracts of experimentally infected adults. The models were fit to data from an experimental H1N1 influenza A/Hong Kong/123/77 infection and suggest that it is important to include the eclipse phase of the viral life cycle in viral dynamic models. Doing so, we estimate that after a delay of approximately 6 h, infected cells begin producing influenza virus and continue to do so for approximately 5 h. The average lifetime of infected cells is approximately 11 h, and the half-life of free infectious virus is approximately 3 h. We calculated the basic reproductive number, R(0), which indicated that a single infected cell could produce approximately 22 new productive infections. This suggests that antiviral treatments have a large hurdle to overcome in moderating symptoms and limiting infectiousness and that treatment has to be initiated as early as possible. For about 50% of patients, the curve of viral titer versus time has two peaks. This bimodal behavior can be explained by incorporating the antiviral effects of interferon into the model. Our model also compared well to an additional data set on viral titer after experimental infection and treatment with the neuraminidase inhibitor zanamivir, which suggests that such models may prove useful in estimating the efficacies of different antiviral therapies for influenza A infection.
Collapse
Affiliation(s)
- Prasith Baccam
- Theoretical Biology and Biophysics Group, Los Alamos National Laboratory, Los Alamos, NM 87545, USA
| | | | | | | | | |
Collapse
|
500
|
McKay T, Patel M, Pickles RJ, Johnson LG, Olsen JC. Influenza M2 envelope protein augments avian influenza hemagglutinin pseudotyping of lentiviral vectors. Gene Ther 2006; 13:715-24. [PMID: 16397505 DOI: 10.1038/sj.gt.3302715] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Lentivirus-based gene transfer has the potential to efficiently deliver DNA-based therapies into non-dividing epithelial cells of the airway for the treatment of lung diseases such as cystic fibrosis. However, significant barriers both to lung-specific gene transfer and to production of lentivirus vectors must be overcome before these vectors can be routinely used for applications to the lung. In this study, we investigated whether the ability to produce lentiviral vectors pseudotyped with fowl plague virus hemagglutinin (HA) could be improved by co-expression of influenza virus M2 in vector-producing cells. We found that M2 expression led to a 10-30-fold increase in production of HA-pseudotyped lentivirus vectors based upon equine infectious anemia virus (EIAV) or human immunodeficiency virus type 1 (HIV-1). Experiments using the M2 inhibitor amantadine and a drug-resistant mutant of M2 established that the ion channel activity of M2 was important for M2-dependent augmentation of vector production. Furthermore, the neuraminidase activity necessary for particle release from producer cells could also be incorporated into producer cells by co-expression of influenza NA cDNA. Lentiviral vectors pseudotyped with influenza envelope proteins were able to efficiently transduce via the apical membrane of polarized mouse tracheal cultures in vitro as well as mouse tracheal epithelia in vivo.
Collapse
Affiliation(s)
- T McKay
- Department of Medicine, Cystic Fibrosis/Pulmonary Research and Treatment Center, University of North Carolina at Chapel Hill, 27599, USA
| | | | | | | | | |
Collapse
|