Structure and accessibility of HA trimers on intact 2009 H1N1 pandemic influenza virus to stem region-specific neutralizing antibodies

Rapid antigenic variation of HA, the major virion surface protein of influenza A virus, remains the principal challenge to the development of broader and more effective vaccines. Some regions of HA, such as the stem region proximal to the viral membrane, are nevertheless highly conserved across strains and among most subtypes. A fundamental question in vaccine design is the extent to which HA stem regions on the surface of the virus are accessible to broadly neutralizing antibodies. Here we report 3D structures derived from cryoelectron tomography of HA on intact 2009 H1N1 pandemic virions in the presence and absence of the antibody C179, which neutralizes viruses expressing a broad range of HA subtypes, including H1, H2, H5, H6, and H9. By fitting previously derived crystallographic structures of trimeric HA into the density maps, we deduced the locations of the molecular surfaces of HA involved in interaction with C179. Using computational methods to distinguish individual unliganded HA trimers from those that have bound C179 antibody, we demonstrate that ∼75% of HA trimers on the surface of the virus have C179 bound to the stem domain. Thus, despite their close packing on the viral membrane, the majority of HA trimers on intact virions are available to bind anti-stem antibodies that target conserved HA epitopes, establishing the feasibility of universal influenza vaccines that elicit such antibodies.

[1]  G. Sapiro,et al.  A collaborative framework for 3D alignment and classification of heterogeneous subvolumes in cryo-electron tomography. , 2013, Journal of structural biology.

[2]  N. S. Laursen,et al.  Highly Conserved Protective Epitopes on Influenza B Viruses , 2012, Science.

[3]  Guillermo Sapiro,et al.  Structural Mechanism of Trimeric HIV-1 Envelope Glycoprotein Activation , 2012, PLoS pathogens.

[4]  G. Sapiro,et al.  Computational separation of conformational heterogeneity using cryo-electron tomography and 3D sub-volume averaging. , 2012, Journal of structural biology.

[5]  K. Lindblade,et al.  A distinct lineage of influenza A virus from bats , 2012, Proceedings of the National Academy of Sciences.

[6]  Dennis C Winkler,et al.  Structural Changes in Influenza Virus at Low pH Characterized by Cryo-Electron Tomography , 2012, Journal of Virology.

[7]  Gabriele Neumann,et al.  Experimental adaptation of an influenza H5 haemagglutinin (HA) confers respiratory droplet transmission to a reassortant H5 HA/H1N1 virus in ferrets , 2012, Nature.

[8]  K. Subbarao,et al.  Eurasian-Origin Gene Segments Contribute to the Transmissibility, Aerosol Release, and Morphology of the 2009 Pandemic H1N1 Influenza Virus , 2011, PLoS pathogens.

[9]  F. Ennis,et al.  Complement-Dependent Lysis of Influenza A Virus-Infected Cells by Broadly Cross-Reactive Human Monoclonal Antibodies , 2011, Journal of Virology.

[10]  Martin H. Koldijk,et al.  A Highly Conserved Neutralizing Epitope on Group 2 Influenza A Viruses , 2011, Science.

[11]  J. Skehel,et al.  A Neutralizing Antibody Selected from Plasma Cells That Binds to Group 1 and Group 2 Influenza A Hemagglutinins , 2011, Science.

[12]  Surender Khurana,et al.  Broadly neutralizing human antibody that recognizes the receptor-binding pocket of influenza virus hemagglutinin , 2011, Proceedings of the National Academy of Sciences.

[13]  Ryo Takano,et al.  A cross-reactive neutralizing monoclonal antibody protects mice from H5N1 and pandemic (H1N1) 2009 virus infection. , 2010, Antiviral research.

[14]  A. Bartesaghi,et al.  Molecular Architectures of Trimeric SIV and HIV-1 Envelope Glycoproteins on Intact Viruses: Strain-Dependent Variation in Quaternary Structure , 2010, PLoS pathogens.

[15]  D. Ekiert,et al.  Vaccination with a synthetic peptide from the influenza virus hemagglutinin provides protection against distinct viral subtypes , 2010, Proceedings of the National Academy of Sciences.

[16]  Jianhua He,et al.  The 2009 pandemic H1N1 neuraminidase N1 lacks the 150-cavity in its active site , 2010, Nature Structural &Molecular Biology.

[17]  G. Nabel,et al.  Induction of Broadly Neutralizing H1N1 Influenza Antibodies by Vaccination , 2010, Science.

[18]  Steven J. Gamblin,et al.  Influenza Hemagglutinin and Neuraminidase Membrane Glycoproteins , 2010, The Journal of Biological Chemistry.

[19]  P. Rosenthal,et al.  Structural organization of a filamentous influenza A virus , 2010, Proceedings of the National Academy of Sciences.

[20]  James E. Crowe,et al.  Structural Basis of Preexisting Immunity to the 2009 H1N1 Pandemic Influenza Virus , 2010, Science.

[21]  Kelly K. Lee Architecture of a nascent viral fusion pore , 2010, The EMBO journal.

[22]  R. Hai,et al.  Broadly Protective Monoclonal Antibodies against H3 Influenza Viruses following Sequential Immunization with Different Hemagglutinins , 2010, PLoS pathogens.

[23]  Ron A M Fouchier,et al.  Antigenic and Genetic Characteristics of Swine-Origin 2009 A(H1N1) Influenza Viruses Circulating in Humans , 2009, Science.

[24]  Gira Bhabha,et al.  Antibody Recognition of a Highly Conserved Influenza Virus Epitope , 2009, Science.

[25]  Boguslaw Stec,et al.  Structural and functional bases for broad-spectrum neutralization of avian and human influenza A viruses , 2009, Nature Structural &Molecular Biology.

[26]  G Sapiro,et al.  Classification and 3D averaging with missing wedge correction in biological electron tomography. , 2008, Journal of structural biology.

[27]  Mark Horowitz,et al.  Markov random field based automatic image alignment for electron tomography. , 2007, Journal of structural biology.

[28]  Giovanni Cardone,et al.  Influenza virus pleiomorphy characterized by cryoelectron tomography , 2006, Proceedings of the National Academy of Sciences.

[29]  David J. Stevens,et al.  Haemagglutinin mutations responsible for the binding of H5N1 influenza A viruses to human-type receptors , 2006, Nature.

[30]  P. Wingfield,et al.  Epitope diversity of hepatitis B virus capsids: quasi-equivalent variations in spike epitopes and binding of different antibodies to the same epitope. , 2006, Journal of molecular biology.

[31]  J. Taubenberger,et al.  1918 Influenza: the Mother of All Pandemics , 2006, Emerging infectious diseases.

[32]  Conrad C. Huang,et al.  UCSF Chimera—A visualization system for exploratory research and analysis , 2004, J. Comput. Chem..

[33]  J. Skehel,et al.  H1 and H7 influenza haemagglutinin structures extend a structural classification of haemagglutinin subtypes. , 2004, Virology.

[34]  Robyn L Stanfield,et al.  Contrasting IgG structures reveal extreme asymmetry and flexibility. , 2002, Journal of molecular biology.

[35]  T. Baker,et al.  Adding the Third Dimension to Virus Life Cycles: Three-Dimensional Reconstruction of Icosahedral Viruses from Cryo-Electron Micrographs , 2000, Microbiology and Molecular Biology Reviews.

[36]  M. van Heel,et al.  Structure of influenza haemagglutinin at neutral and at fusogenic pH by electron cryo‐microscopy , 1999, FEBS letters.

[37]  A. Osterhaus,et al.  An epitope shared by the hemagglutinins of H1, H2, H5, and H6 subtypes of influenza A virus. , 1999, Acta virologica.

[38]  L J Harris,et al.  Crystallographic structure of an intact IgG1 monoclonal antibody. , 1998, Journal of molecular biology.

[39]  S. Durell,et al.  Dilation of the influenza hemagglutinin fusion pore revealed by the kinetics of individual cell-cell fusion events , 1996, The Journal of cell biology.

[40]  J R Kremer,et al.  Computer visualization of three-dimensional image data using IMOD. , 1996, Journal of structural biology.

[41]  Timothy S Baker,et al.  Nucleocapsid and glycoprotein organization in an enveloped virus , 1995, Cell.

[42]  J. Skehel,et al.  Structure of influenza haemagglutinin at the pH of membrane fusion , 1994, Nature.

[43]  Y. Isegawa,et al.  A common neutralizing epitope conserved between the hemagglutinins of influenza A virus H1 and H2 strains , 1993, Journal of virology.

[44]  W. Chiu,et al.  Localization of VP4 neutralization sites in rotavirus by three-dimensional cryo-electron microscopy , 1990, Nature.

[45]  I. Wilson,et al.  Changes in the conformation of influenza virus hemagglutinin at the pH optimum of virus-mediated membrane fusion. , 1982, Proceedings of the National Academy of Sciences of the United States of America.

[46]  I. Wilson,et al.  Structure of the haemagglutinin membrane glycoprotein of influenza virus at 3 Å resolution , 1981, Nature.

[47]  M. Gething,et al.  Cloning and DNA sequence of double-stranded copies of haemagglutinin genes from H2 and H3 strains elucidates antigenic shift and drift in human influenza virus , 1980, Nature.

[48]  G. Air,et al.  Antigenic drift in type A influenza virus: peptide mapping and antigenic analysis of A/PR/8/34 (HON1) variants selected with monoclonal antibodies. , 1979, Proceedings of the National Academy of Sciences of the United States of America.

[49]  P. Choppin,et al.  STUDIES OF TWO KINDS OF VIRUS PARTICLES WHICH COMPRISE INFLUENZA A2 VIRUS STRAINS , 1960, The Journal of experimental medicine.

[50]  R. Wyckoff,et al.  Electron Micrography of the Virus of Influenza , 1946, Nature.

[51]  J. Taubenberger,et al.  Influenza : the Mother of All Pandemics , 2022 .