Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters











Publication year range
1.
J Virol ; 96(2): e0090921, 2022 01 26.
Article in English | MEDLINE | ID: mdl-34730389

ABSTRACT

Human metapneumovirus (HMPV) causes severe respiratory diseases in young children. The HMPV RNA genome is encapsidated by the viral nucleoprotein (N), forming an RNA-N complex (NNuc), which serves as the template for genome replication and mRNA transcription by the RNA-dependent RNA polymerase (RdRp). The RdRp is formed by the association of the large polymerase subunit (L), which has RNA polymerase, capping, and methyltransferase activities, and the tetrameric phosphoprotein (P). P plays a central role in the RdRp complex by binding to NNuc and L, allowing the attachment of the L polymerase to the NNuc template. During infection these proteins concentrate in cytoplasmic inclusion bodies (IBs) where viral RNA synthesis occurs. By analogy to the closely related pneumovirus respiratory syncytial virus (RSV), it is likely that the formation of IBs depends on the interaction between HMPV P and NNuc, which has not been demonstrated yet. Here, we finely characterized the binding P-NNuc interaction domains by using recombinant proteins, combined with a functional assay for the polymerase complex activity, and the study of the recruitment of these proteins to IBs by immunofluorescence. We show that the last 6 C-terminal residues of HMPV P are necessary and sufficient for binding to NNuc and that P binds to the N-terminal domain of N (NNTD), and we identified conserved N residues critical for the interaction. Our results allowed us to propose a structural model for the HMPV P-NNuc interaction. IMPORTANCE Human metapneumovirus (HMPV) is a leading cause of severe respiratory infections in children but also affects human populations of all ages worldwide. Currently, no vaccine or efficient antiviral treatments are available for this pneumovirus. A better understanding of the molecular mechanisms involved in viral replication could help the design or discovery of specific antiviral compounds. In this work, we have investigated the interaction between two major viral proteins involved in HMPV RNA synthesis, the N and P proteins. We finely characterized their domains of interaction and identified a pocket on the surface of the N protein, a potential target of choice for the design of compounds interfering with N-P complexes and inhibiting viral replication.


Subject(s)
Metapneumovirus/chemistry , Nucleocapsid Proteins/chemistry , Phosphoproteins/chemistry , Animals , Binding Sites , Cell Line , Cricetinae , Inclusion Bodies/metabolism , Metapneumovirus/physiology , Models, Molecular , Mutation , Nucleocapsid Proteins/genetics , Nucleocapsid Proteins/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein Binding , Protein Interaction Domains and Motifs , RNA, Viral/metabolism , RNA-Dependent RNA Polymerase/metabolism , Virus Replication
2.
Virology ; 531: 248-254, 2019 05.
Article in English | MEDLINE | ID: mdl-30946995

ABSTRACT

The human metapneumovirus (HMPV) fusion protein (F) mediates fusion of the viral envelope and cellular membranes to establish infection. HMPV F from some, but not all, viral strains promotes fusion only after exposure to low pH. Previous studies have identified several key residues involved in low pH triggering, including H435 and a proposed requirement for glycine at position 294. We analyzed the different levels of fusion activity, protein expression and cleavage of three HMPV F proteins not previously examined. Interestingly, low pH-triggered fusion in the absence of G294 was identified in one F protein, while a novel histidine residue (H434) was identified that enhanced low pH promoted fusion in another. The third F protein failed to promote cell-to-cell fusion, suggesting other requirements for F protein triggering. Our results demonstrate HMPV F triggering is more complex than previously described and suggest a more intricate mechanism for fusion protein function and activation.


Subject(s)
Metapneumovirus/metabolism , Paramyxoviridae Infections/virology , Viral Fusion Proteins/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Chlorocebus aethiops , Humans , Metapneumovirus/chemistry , Metapneumovirus/genetics , Protein Stability , Sequence Alignment , Vero Cells , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/genetics
3.
Sci Rep ; 7(1): 14865, 2017 11 01.
Article in English | MEDLINE | ID: mdl-29093501

ABSTRACT

The phosphoprotein (P) is the main and essential cofactor of the RNA polymerase (L) of non-segmented, negative-strand RNA viruses. P positions the viral polymerase onto its nucleoprotein-RNA template and acts as a chaperone of the nucleoprotein (N), thereby preventing nonspecific encapsidation of cellular RNAs. The phosphoprotein of human metapneumovirus (HMPV) forms homotetramers composed of a stable oligomerization domain (Pcore) flanked by large intrinsically disordered regions (IDRs). Here we combined x-ray crystallography of Pcore with small angle x-ray scattering (SAXS)-based ensemble modeling of the full-length P protein and several of its fragments to provide a structural description of P that captures its dynamic character, and highlights the presence of varyingly stable structural elements within the IDRs. We discuss the implications of the structural properties of HMPV P for the assembly and functioning of the viral transcription/replication machinery.


Subject(s)
Metapneumovirus/chemistry , Phosphoproteins/chemistry , DNA-Directed RNA Polymerases/metabolism , Humans , Nucleoproteins/metabolism , Phosphoproteins/metabolism , Protein Stability , Scattering, Small Angle , Viral Proteins/chemistry , Viral Proteins/metabolism , Virus Replication , X-Ray Diffraction
4.
Virology ; 499: 361-368, 2016 12.
Article in English | MEDLINE | ID: mdl-27743962

ABSTRACT

Human metapneumovirus (hMPV) is a major cause of lower respiratory infection in young children. Repeated infections occur throughout life, but its immune evasion mechanisms are largely unknown. We recently found that hMPV M2-2 protein elicits immune evasion by targeting mitochondrial antiviral-signaling protein (MAVS), an antiviral signaling molecule. However, the molecular mechanisms underlying such inhibition are not known. Our mutagenesis studies revealed that PDZ-binding motifs, 29-DEMI-32 and 39-KEALSDGI-46, located in an immune inhibitory region of M2-2, are responsible for M2-2-mediated immune evasion. We also found both motifs prevent TRAF5 and TRAF6, the MAVS downstream adaptors, to be recruited to MAVS, while the motif 39-KEALSDGI-46 also blocks TRAF3 migrating to MAVS. In parallel, these TRAFs are important in activating transcription factors NF-kB and/or IRF-3 by hMPV. Our findings collectively demonstrate that M2-2 uses its PDZ motifs to launch the hMPV immune evasion through blocking the interaction of MAVS and its downstream TRAFs.


Subject(s)
Immune Evasion , Immunity, Innate , Metapneumovirus/immunology , Paramyxoviridae Infections/immunology , Viral Proteins/chemistry , Viral Proteins/immunology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Amino Acid Motifs , Cell Line , Humans , Metapneumovirus/chemistry , Metapneumovirus/genetics , Paramyxoviridae Infections/virology , TNF Receptor-Associated Factor 6/genetics , TNF Receptor-Associated Factor 6/immunology , Viral Proteins/genetics
5.
Clin Vaccine Immunol ; 23(6): 451-9, 2016 06.
Article in English | MEDLINE | ID: mdl-27030590

ABSTRACT

Respiratory syncytial virus (RSV) is the leading cause of severe respiratory disease in infants and children and represents an important global health burden for the elderly and the immunocompromised. Despite decades of research efforts, no licensed vaccine for RSV is available. We have developed virus-like particle (VLP)-based RSV vaccines assembled with the human metapneumovirus (hMPV) matrix protein (M) as the structural scaffold and the RSV fusion glycoprotein (F) in either the postfusion or prefusion conformation as its prime surface immunogen. Vaccines were composed of postfusion F, prefusion F, or a combination of the two conformations and formulated with a squalene-based oil emulsion as adjuvant. Immunization with these VLP vaccines afforded full protection against RSV infection and prevented detectable viral replication in the mouse lung after challenge. Analyses of lung cytokines and chemokines showed that VLP vaccination mostly induced the production of gamma interferon (IFN-γ), a marker of the Th1-mediated immune response, which is predominantly required for viral protection. Conversely, immunization with a formalin-inactivated RSV (FI-RSV) vaccine induced high levels of inflammatory chemokines and cytokines of the Th2- and Th17-mediated types of immune responses, as well as severe lung inflammation and histopathology. The VLP vaccines showed restricted production of these immune mediators and did not induce severe bronchiolitis or perivascular infiltration as seen with the FI-RSV vaccine. Remarkably, analysis of the serum from immunized mice showed that the VLP vaccine formulated using a combination of postfusion and prefusion F elicited the highest level of neutralizing antibody and enhanced the Th1-mediated immune response.


Subject(s)
Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/chemistry , Respiratory Syncytial Viruses/immunology , Vaccines, Virus-Like Particle/chemistry , Viral Fusion Proteins/chemistry , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Cytokines/immunology , Humans , Immunization , Interferon-gamma/immunology , Lung/immunology , Lung/virology , Metapneumovirus/chemistry , Mice , Mice, Inbred BALB C , Protein Conformation , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Vaccines/administration & dosage , Respiratory Syncytial Virus Vaccines/genetics , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Viruses/chemistry , Respiratory Syncytial Viruses/genetics , Th17 Cells/immunology , Th2 Cells/immunology , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/chemistry , Vaccines, Inactivated/genetics , Vaccines, Inactivated/immunology , Vaccines, Virus-Like Particle/administration & dosage , Vaccines, Virus-Like Particle/genetics , Vaccines, Virus-Like Particle/immunology , Viral Fusion Proteins/adverse effects , Viral Fusion Proteins/genetics , Viral Fusion Proteins/immunology , Viral Load , Viral Matrix Proteins/immunology
6.
Bing Du Xue Bao ; 32(6): 758-67, 2016 11.
Article in Chinese | MEDLINE | ID: mdl-30004649

ABSTRACT

The human metapneumovirus (HMPV) is an important pathogen in respiratory-tract infections in children. We undertook genomic sequence analyses and described the genetic characteristics of an uncommon sub-genotype, the HMPV A1 strain, and provide useful data for further studies. The HMPV A1(BJ-1610)strain was identified from a nasopharyngeal aspirate collected from a 3-month-old female with bronchopneumonia. Gene fragments of BJ-1610 were amplified by reverse transcription-polymerase chain reaction(RT-PCR)and assembled by DNAStar software. Sequence alignment for BJ-1610 and other HMPV reference strains with four known genotypes available in the GenBank database was conducted by DNAStar. Phylogenetic trees were created using MEGA 6.06 software. The whole genome of BJ-1610 was 13406nt in length (GenBank accession number:KU821121).Compared with HMPV reference strains,BJ-1610 shared the highest similarities with HMPV/AUS/150229278/2003/A(KC562226)from Australia, which was classified into sub-genotype A1.The nucleotide identity of the full genome between BJ-1610 and KC562226was 98.4%.N,P,F,M2-2and L genes had great similarity with KC562226 compared with other reference strains, whereas SH and G genes shared higher similarities with other strains of sub-genotype A1.Phylogenetic analyses of the whole genome showed that BJ-1610 was clustered into sub-genotype A1 and was close to KC562226.The N,P,M,F,M2-1,M2-2and L genes of BJ-1610 showed the same genetic features as the whole genome, whereas the variable genes SH and G were closest to KC403980.The F protein of BJ-1610 showed high genetic conservation. The length of the SH protein of BJ-1610 changed from 552 bp to 567 bp due to mutations in the stop codon. The amino-acid mutations on protein G led to a decrease in the number of N-glycosylation sites. As an infrequently circulating genotype, sequence analyses of the whole genome of a HMPV A1strain(BJ-1610)will promote further studies on its epidemiology and pathogenicity, and aid the development of vaccines and antiviral drugs.


Subject(s)
Metapneumovirus/genetics , Metapneumovirus/isolation & purification , Paramyxoviridae Infections/virology , Pneumonia/virology , Amino Acid Sequence , China , Female , Genome, Viral , Genotype , Humans , Infant , Male , Metapneumovirus/chemistry , Metapneumovirus/classification , Molecular Sequence Data , Phylogeny , Sequence Alignment , Viral Proteins/chemistry , Viral Proteins/genetics , Whole Genome Sequencing
7.
Anal Chem ; 87(20): 10247-54, 2015 Oct 20.
Article in English | MEDLINE | ID: mdl-26376123

ABSTRACT

The rapid, sensitive, and specific identification of infectious pathogens from clinical isolates is a critical need in the hospital setting. Mass spectrometry (MS) has been widely adopted for identification of bacterial pathogens, although polymerase chain reaction remains the mainstay for the identification of viral pathogens. Here, we explored the capability of MS for the detection of human metapneumovirus (HMPV), a common cause of respiratory tract infections in children. Liquid chromatography-tandem mass spectrometry (LC-MS/MS) sequencing of a single HMPV reference strain (CAN97-83) was used to develop a multiple reaction monitoring (MRM) assay that employed stable isotope-labeled peptide internal standards for quantitation of HMPV. Using this assay, we confirmed the presence of HMPV in viral cultures from 10 infected patients and further assigned genetic lineage based on the presence/absence of variant peptides belonging to the viral matrix and nucleoproteins. Similar results were achieved for primary clinical samples (nasopharyngeal aspirates) from the same individuals. As validation, virus lineages, and variant coding sequences, were confirmed by next-generation sequencing of viral RNA obtained from the culture samples. Finally, separate dilution series of HMPV A and B lineages were used to further refine and assess the robustness of the assay and to determine limits of detection in nasopharyngeal aspirates. Our results demonstrate the applicability of MRM for identification of HMPV, and assignment of genetic lineage, from both viral cultures and clinical samples. More generally, this approach should prove tractable as an alternative to nucleic-acid based sequencing for the multiplexed identification of respiratory virus infections.


Subject(s)
Metapneumovirus/chemistry , Metapneumovirus/growth & development , Paramyxoviridae Infections/virology , Proteome/analysis , Proteomics , Viral Proteins/analysis , Cells, Cultured , Chromatography, Liquid , Humans , Metapneumovirus/genetics , Metapneumovirus/isolation & purification , RNA, Viral/analysis , RNA, Viral/genetics , Tandem Mass Spectrometry
8.
Arch Virol ; 160(10): 2445-53, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26175070

ABSTRACT

The entry of enveloped viruses into host cells requires the fusion of viral and cell membranes. These membrane fusion reactions are mediated by virus-encoded glycoproteins. In the case of avian metapneumovirus (aMPV), the fusion (F) protein alone can mediate virus entry and induce syncytium formation in vitro. To investigate the fusogenic activity of the aMPV F protein, we compared the fusogenic activities of three subtypes of aMPV F proteins using a TCSD50 assay developed in this study. Interestingly, we found that the F protein of aMPV subtype B (aMPV/B) strain VCO3/60616 (aMPV/vB) was hyperfusogenic when compared with F proteins of aMPV/B strain aMPV/f (aMPV/fB), aMPV subtype A (aMPV/A), and aMPV subtype C (aMPV/C). We then further demonstrated that the amino acid (aa) residue 149F contributed to the hyperfusogenic activity of the aMPV/vB F protein. Moreover, we revealed that residue 149F had no effect on the fusogenic activities of aMPV/A, aMPV/C, and human metapneumovirus (hMPV) F proteins. Collectively, we provide the first evidence that the amino acid at position 149 affects the fusogenic activity of the aMPV/B F protein, and our findings will provide new insights into the fusogenic mechanism of this protein.


Subject(s)
Genetic Variation , Metapneumovirus/genetics , Paramyxoviridae Infections/veterinary , Poultry Diseases/virology , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Chlorocebus aethiops , Humans , Metapneumovirus/chemistry , Metapneumovirus/classification , Metapneumovirus/metabolism , Molecular Sequence Data , Paramyxoviridae Infections/virology , Sequence Alignment , Turkeys/virology , Vero Cells , Viral Fusion Proteins/genetics
9.
J Virol ; 88(19): 11611-6, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25031352

ABSTRACT

Human metapneumovirus is a major cause of respiratory tract infections worldwide. Previous reports have shown that the viral attachment glycoprotein (G) modulates innate and adaptive immune responses, leading to incomplete immunity and promoting reinfection. Using bioinformatics analyses, static light scattering, and small-angle X-ray scattering, we show that the extracellular region of G behaves as a heavily glycosylated, intrinsically disordered polymer. We discuss potential implications of these findings for the modulation of immune responses by G.


Subject(s)
Glycoproteins/chemistry , Metapneumovirus/chemistry , Viral Proteins/chemistry , Glycoproteins/immunology , Glycoproteins/metabolism , Glycosylation , Humans , Immunity, Innate , Metapneumovirus/immunology , Metapneumovirus/metabolism , Models, Molecular , Protein Structure, Tertiary , Viral Proteins/immunology , Viral Proteins/metabolism
10.
Structure ; 22(1): 5-7, 2014 Jan 07.
Article in English | MEDLINE | ID: mdl-24411575

ABSTRACT

In this issue of Structure, Leyrat and colleagues provide the first structural analysis of the human metapneumovirus (HMPV) matrix protein, a key regulator of viral assembly. Though structurally similar to other matrix proteins, two calcium binding sites suggest intriguing differences in regulation.


Subject(s)
Calcium-Binding Proteins/chemistry , Calcium/chemistry , Metapneumovirus/chemistry , Viral Matrix Proteins/chemistry , Virion/chemistry , Humans
11.
Structure ; 22(1): 136-48, 2014 Jan 07.
Article in English | MEDLINE | ID: mdl-24316400

ABSTRACT

The matrix protein (M) of paramyxoviruses plays a key role in determining virion morphology by directing viral assembly and budding. Here, we report the crystal structure of the human metapneumovirus M at 2.8 Å resolution in its native dimeric state. The structure reveals the presence of a high-affinity Ca²âº binding site. Molecular dynamics simulations (MDS) predict a secondary lower-affinity site that correlates well with data from fluorescence-based thermal shift assays. By combining small-angle X-ray scattering with MDS and ensemble analysis, we captured the structure and dynamics of M in solution. Our analysis reveals a large positively charged patch on the protein surface that is involved in membrane interaction. Structural analysis of DOPC-induced polymerization of M into helical filaments using electron microscopy leads to a model of M self-assembly. The conservation of the Ca²âº binding sites suggests a role for calcium in the replication and morphogenesis of pneumoviruses.


Subject(s)
Calcium-Binding Proteins/chemistry , Calcium/chemistry , Metapneumovirus/chemistry , Viral Matrix Proteins/chemistry , Virion/chemistry , Calcium/metabolism , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Humans , Metapneumovirus/physiology , Metapneumovirus/ultrastructure , Microscopy, Electron , Molecular Dynamics Simulation , Phosphatidylcholines/chemistry , Protein Multimerization , Protein Structure, Secondary , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Scattering, Small Angle , Thermodynamics , Viral Matrix Proteins/genetics , Viral Matrix Proteins/metabolism , Virion/physiology , Virion/ultrastructure , Virus Assembly , Virus Replication , X-Ray Diffraction
12.
Nat Struct Mol Biol ; 19(4): 461-3, 2012 Mar 04.
Article in English | MEDLINE | ID: mdl-22388735

ABSTRACT

Human metapneumovirus and respiratory syncytial virus cause lower respiratory tract infections. The virus fusion (F) glycoprotein promotes membrane fusion by refolding from a metastable pre-fusion to a stable post-fusion conformation. F is also a major target of the neutralizing antibody response. Here we show that a potently neutralizing anti-human metapneumovirus antibody (DS7) binds a structurally invariant domain of F, revealing a new epitope that could be targeted in vaccine development.


Subject(s)
Antibodies, Neutralizing/chemistry , Epitopes/chemistry , Metapneumovirus/chemistry , Viral Fusion Proteins/chemistry , Antibodies, Neutralizing/immunology , Epitopes/immunology , Humans , Metapneumovirus/immunology , Models, Molecular , Paramyxoviridae Infections/immunology , Paramyxoviridae Infections/virology , Protein Structure, Tertiary , Viral Fusion Proteins/immunology
13.
J Virol ; 86(6): 3230-43, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22238303

ABSTRACT

Human metapneumovirus (HMPV) is a major worldwide respiratory pathogen that causes acute upper and lower respiratory tract disease. The mechanism by which this virus recognizes and gains access to its target cell is still largely unknown. In this study, we addressed the initial steps in virus binding and infection and found that the first binding partner for HMPV is heparan sulfate (HS). While wild-type CHO-K1 cells are permissive to HMPV infection, mutant cell lines lacking the ability to synthesize glycosaminoglycans (GAGs), specifically, heparan sulfate proteoglycans (HSPGs), were resistant to binding and infection by HMPV. The permissiveness to HMPV infection was also abolished when CHO-K1 cells were treated with heparinases. Importantly, using recombinant HMPV lacking both the G and small hydrophobic (SH) proteins, we report that this first virus-cell binding interaction is driven primarily by the fusion protein (HMPV F) and that this interaction is needed to establish a productive infection. Finally, HMPV binding to cells did not require ß1 integrin expression, and RGD-mediated interactions were not essential in promoting HMPV F-mediated cell-to-cell membrane fusion. Cells lacking ß1 integrin, however, were less permissive to HMPV infection, indicating that while ß1 integrins play an important role in promoting HMPV infection, the interaction between integrins and HMPV occurs after the initial binding of HMPV F to heparan sulfate proteoglycans.


Subject(s)
Heparitin Sulfate/metabolism , Metapneumovirus/metabolism , Paramyxoviridae Infections/metabolism , Receptors, Virus/metabolism , Viral Fusion Proteins/metabolism , Amino Acid Motifs , Animals , Cell Line , Humans , Metapneumovirus/chemistry , Metapneumovirus/genetics , Paramyxoviridae Infections/virology , Protein Binding , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/genetics
14.
Virus Res ; 161(2): 131-9, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21827798

ABSTRACT

Human metapneumovirus (hMPV) is a member of the Pneumovirinea subfamily within the Paramyxoviridea family. Since its discovery in 2001, hMPV has been isolated in several continents, which suggests its prevalence worldwide. hMPV resembles human respiratory syncytial virus with regard to disease symptoms and its ability to infect and cause disease in young infants as well as individuals of all ages. The aim of the current study was to construct an efficient high-level yeast expression system for the generation of hMPV nucleocapsid (N) protein and to develop monoclonal antibodies (MAbs) suitable for hMPV detection. The genome of hMPV was isolated from oral fluid of an infected patient by using specific primers and reverse transcriptase polymerase chain reaction (RT-PCR). DNA sequence corresponding to the N protein gene was inserted into yeast expression vector under inducible GAL7 promoter. SDS-PAGE analysis of crude lysates of yeast Saccharomyces cerevisiae harbouring recombinant plasmid revealed the presence of a protein band of approximately 43 kDa corresponding to the molecular weight of hMPV N protein. Electron microscopy analysis of purified N protein revealed nucleocapsid-like structures with typical herring-bone morphology: rods of 20 nm diameter with repeated serration along the edges and central core of 5 nm. Recombinant hMPV N protein was reactive with human serum specimens collected from patients with confirmed hMPV infection. After immunization of mice with recombinant hMPV N protein, a panel of MAbs was generated. The specificity of newly generated MAbs was proven by immunofluorescence analysis of hMPV-infected cells. Epitope mapping using truncated variants of hMPV N revealed localization of linear MAb epitopes at the N-terminus of hMPV N protein, between amino acid residues 1 and 90. The MAbs directed against conformational epitopes did not recognize hMPV N protein variants containing either N- or C-terminal truncations. The reactivity of recombinant hMPV N protein with hMPV-positive serum specimens and the ability of MAbs to recognize virus-infected cells confirms the antigenic similarity between yeast-expressed hMPV N protein and native viral nucleocapsids. In conclusion, recombinant hMPV N protein and hMPV-specific MAbs provide new diagnostic reagents for hMPV infection.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Metapneumovirus/immunology , Nucleocapsid Proteins/immunology , Paramyxoviridae Infections/immunology , Amino Acid Motifs , Animals , Cell Line , Child , Epitope Mapping , Female , Humans , Male , Metapneumovirus/chemistry , Metapneumovirus/genetics , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Nucleocapsid Proteins/chemistry , Nucleocapsid Proteins/genetics , Paramyxoviridae Infections/virology , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology
15.
Virus Res ; 160(1-2): 102-7, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21683102

ABSTRACT

The small hydrophobic protein (SH) is a type II integral membrane protein that is packaged into virions and is only present in certain paramyxoviruses including metapneumovirus. In addition to a highly divergent primary sequence, SH proteins vary significantly in size amongst the different viruses. Human respiratory syncytial virus (HRSV) encodes the smallest SH protein consisting of only 64 amino acids, while metapneumoviruses have the longest SH protein ranging from 174 to 179 amino acids in length. Little is currently known about the cellular localization and topology of the metapneumovirus SH protein. Here we characterize for the first time metapneumovirus SH protein with respect to topology, subcellular localization, and transport using avian metapneumovirus subgroup C (AMPV-C) as a model system. We show that AMPV-C SH is an integral membrane protein with N(in)C(out) orientation located in both the plasma membrane as well as within intracellular compartments, which is similar to what has been described previously for SH proteins of other paramyxoviruses. Furthermore, we demonstrate that AMPV-C SH protein localizes in the endoplasmic reticulum (ER), Golgi, and cell surface, and is transported through ER-Golgi secretory pathway.


Subject(s)
Metapneumovirus/chemistry , Metapneumovirus/metabolism , Retroviridae Proteins, Oncogenic/chemistry , Retroviridae Proteins, Oncogenic/metabolism , Cell Membrane/chemistry , Cytoplasm/chemistry , Endoplasmic Reticulum/chemistry , Golgi Apparatus/chemistry , Protein Transport
16.
J Gen Virol ; 92(Pt 7): 1666-1675, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21450943

ABSTRACT

The fusion (F) protein is an important membrane glycoprotein necessary for cellular entry and replication of human metapneumovirus (hMPV). Selective prevention of N-linked glycosylation may compromise the catalytic and fusion functions of the F protein. By using site-directed mutagenesis and reverse genetics, recombinant mutant viruses lacking one or two N-linked glycosylation sites in the F protein were constructed. M1, which lacked glycosylation at position 57 of the F protein, had slightly compromised replication, whereas M2 and M4, which lacked glycosylation at position(s) 172 or 57 and 172, respectively, showed profound impairment of replication when compared with wild-type (WT) NL/1/00-GFP virus in both Vero E6 cells and mouse lungs. M2 was less fit than WT virus in vitro and in immunocompromised mouse lungs. The F proteins of WT and mutant viruses were similarly expressed on the infected cell membrane, while the activated fusion protein subunits, F1 of M2 and M4, were produced in lower quantities compared with those of WT and M1 virus. The mutated viruses lacking N-linked glycosylation at position 353, either individually or together with other sites, could not be recovered. Thus, N-linked glycosylation may be involved in the catalysis of the fusion protein from F0 to F1 and F2, which is critical for fusion function. Strategies targeting N-linked glycosylation may be helpful for developing attenuated live vaccines or antiviral drugs for hMPV.


Subject(s)
Lung/virology , Metapneumovirus/physiology , Paramyxoviridae Infections/virology , Viral Fusion Proteins/metabolism , Virus Replication , Amino Acid Motifs , Animals , Cell Line , Female , Glycosylation , Humans , Male , Metapneumovirus/chemistry , Metapneumovirus/genetics , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/genetics
17.
Virus Genes ; 41(3): 389-95, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20676749

ABSTRACT

Complete nucleotide sequences were determined for subtype B avian metapneumovirus (aMPV), the attenuated vaccine strain VCO3/50 and its parental pathogenic strain VCO3/60616. The genomes of both strains comprised 13,508 nucleotides (nt), with a 42-nt leader at the 3'-end and a 46-nt trailer at the 5'-end. The genome contains eight genes in the order 3'-N-P-M-F-M2-SH-G-L-5', which is the same order shown in the other metapneumoviruses. The genes are flanked on either side by conserved transcriptional start and stop signals and have intergenic sequences varying in length from 1 to 88 nt. Comparison of nt and predicted amino acid (aa) sequences of VCO3/60616 with those of other metapneumoviruses revealed higher homology with aMPV subtype A virus than with other metapneumoviruses. A total of 18 nt and 10 deduced aa differences were seen between the strains, and one or a combination of several differences could be associated with attenuation of VCO3/50.


Subject(s)
Genome, Viral , Metapneumovirus/genetics , Paramyxoviridae Infections/veterinary , Poultry Diseases/virology , Amino Acid Sequence , Animals , Base Sequence , Chlorocebus aethiops , Metapneumovirus/chemistry , Metapneumovirus/classification , Metapneumovirus/isolation & purification , Molecular Sequence Data , Paramyxoviridae Infections/virology , Sequence Alignment , Turkey , Vero Cells , Viral Proteins/chemistry , Viral Proteins/genetics
18.
Virol J ; 6: 138, 2009 Sep 09.
Article in English | MEDLINE | ID: mdl-19740442

ABSTRACT

BACKGROUND: Human metapneumovirus (HMPV) is an important cause of acute respiratory illness in children. We examined the diversity and molecular evolution of HMPV using 85 full-length F (fusion) gene sequences collected over a 20-year period. RESULTS: The F gene sequences fell into two major groups, each with two subgroups, which exhibited a mean of 96% identity by predicted amino acid sequences. Amino acid identity within and between subgroups was higher than nucleotide identity, suggesting structural or functional constraints on F protein diversity. There was minimal progressive drift over time, and the genetic lineages were stable over the 20-year period. Several canonical amino acid differences discriminated between major subgroups, and polymorphic variations tended to cluster in discrete regions. The estimated rate of mutation was 7.12 x 10(-4) substitutions/site/year and the estimated time to most recent common HMPV ancestor was 97 years (95% likelihood range 66-194 years). Analysis suggested that HMPV diverged from avian metapneumovirus type C (AMPV-C) 269 years ago (95% likelihood range 106-382 years). CONCLUSION: HMPV F protein remains conserved over decades. HMPV appears to have diverged from AMPV-C fairly recently.


Subject(s)
Evolution, Molecular , Genetic Variation , Metapneumovirus/genetics , Viral Fusion Proteins/genetics , Humans , Metapneumovirus/chemistry , Metapneumovirus/classification , Molecular Sequence Data , Phylogeny , Viral Fusion Proteins/chemistry
19.
J Virol ; 83(3): 1511-22, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19036821

ABSTRACT

Human metapneumovirus (HMPV) is a significant respiratory pathogen classified in the Pneumovirinae subfamily of the paramyxovirus family. Recently, we demonstrated that HMPV F protein-promoted cell-cell fusion is stimulated by exposure to low pH, in contrast to what is observed for other paramyxovirus F proteins. In the present study, we examined the potential role of histidine protonation in HMPV F fusion and investigated the role of low pH in HMPV viral entry. Mutagenesis of the three ectodomain histidine residues of the HMPV F protein demonstrated that the mutation of a histidine in the heptad repeat B linker domain (H435) ablated fusion activity without altering cell surface expression or proteolytic processing significantly. Modeling of the HMPV F protein revealed several basic residues surrounding this histidine residue, and the mutation of these residues also reduced fusion activity. These results suggest that electrostatic repulsion in the heptad repeat B linker region may contribute to the triggering of HMPV F. In addition, we examined the effect of inhibitors of endosomal acidification or endocytosis on the entry of a recombinant green fluorescent protein-expressing HMPV. Interestingly, chemicals that raise the pH of endocytic vesicles resulted in a 30 to 50% decrease in HMPV infection, while the inhibitors of endocytosis reduced infection by as much as 90%. These data suggest that HMPV utilizes an endocytic entry mechanism, in contrast to what has been hypothesized for most paramyxoviruses. In addition, our results indicate that HMPV uses the low pH of the endocytic pathway to enhance infectivity, though the role of low pH likely differs from classically described mechanisms.


Subject(s)
Hydrogen-Ion Concentration , Membrane Fusion , Metapneumovirus/physiology , Animals , Chlorocebus aethiops , Genes, Reporter , Humans , Metapneumovirus/chemistry , Vero Cells
20.
J Virol ; 80(12): 5798-806, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16731919

ABSTRACT

The contribution of cleavage activation of the fusion F protein of human metapneumovirus (HMPV) to replication and pathogenicity in rodents and nonhuman primates was investigated. Recombinant HMPVs were generated in which the naturally occurring trypsin-dependent cleavage sequence (R-Q-S-R downward arrow) was replaced by each of three sequences whose cleavage in vitro does not depend upon added trypsin. Two of these were multibasic sequences derived from avian metapneumovirus type A (R-R-R-R) or type C (R-K-A-R), with the former containing the consensus furin protease cleavage motif (R-X-R/K-R downward arrow). The third one (R-Q-P-R) was derived from a recently described trypsin independent HMPV isolate (J. H. Schickli, J. Kaur, N. Ulbrandt, R. R. Spaete, and R. S. Tang, J. Virol. 79:10678-10689, 2005). To preclude the possibility of conferring even greater virulence to this significant human pathogen, the modifications were done in an HMPV variant that was attenuated by the deletion of two of the three envelope glycoproteins, SH and G. Each of the introduced cleavage sequences conferred trypsin independent F cleavage and growth to HMPV in vitro. However, they differed in the efficiency of trypsin independent growth and plaque formation in vitro: R-R-R-R > R-K-A-R > R-Q-P-R > R-Q-S-R. The R-R-R-R mutant was the only one whose growth in vitro was not augmented by added trypsin, indicative of highly efficient trypsin independent cleavage. When inoculated intranasally into hamsters, there was essentially no difference in the magnitude of replication in the upper or lower respiratory tract between the mutants, and virus was not detected in organs outside of the respiratory tract. Evaluation of the most cleavage-efficient mutant, R-R-R-R, in African green monkeys showed that there was no detectable change in the magnitude of replication in the upper and lower respiratory tract or in immunogenicity and protective efficacy against HMPV challenge. These results suggest that cleavage activation is not a major determinant of HMPV virulence.


Subject(s)
Metapneumovirus/chemistry , Trypsin/metabolism , Viral Fusion Proteins/physiology , Amino Acid Sequence , Animals , Binding Sites , Chlorocebus aethiops , Cricetinae , Disease Transmission, Infectious , Immunization , Metapneumovirus/pathogenicity , Metapneumovirus/physiology , Mutation , Viral Fusion Proteins/administration & dosage , Viral Fusion Proteins/pharmacology , Virus Replication/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL