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










Publication year range
1.
NPJ Vaccines ; 9(1): 97, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38821988

ABSTRACT

Current Influenza virus vaccines primarily induce antibody responses against variable epitopes in hemagglutinin (HA), necessitating frequent updates. However, antibodies against neuraminidase (NA) can also confer protection against influenza, making NA an attractive target for the development of novel vaccines. In this study, we aimed to enhance the immunogenicity of recombinant NA antigens by presenting them multivalently on a nanoparticle carrier. Soluble tetrameric NA antigens of the N1 and N2 subtypes, confirmed to be correctly folded by cryo-electron microscopy structural analysis, were conjugated to Mi3 self-assembling protein nanoparticles using the SpyTag-SpyCatcher system. Immunization of mice with NA-Mi3 nanoparticles induced higher titers of NA-binding and -inhibiting antibodies and improved protection against a lethal challenge compared to unconjugated NA. Additionally, we explored the co-presentation of N1 and N2 antigens on the same Mi3 particles to create a mosaic vaccine candidate. These mosaic nanoparticles elicited antibody titers that were similar or superior to the homotypic nanoparticles and effectively protected against H1N1 and H3N2 challenge viruses. The NA-Mi3 nanoparticles represent a promising vaccine candidate that could complement HA-directed approaches for enhanced potency and broadened protection against influenza A virus.

2.
Cells ; 13(2)2024 01 10.
Article in English | MEDLINE | ID: mdl-38247817

ABSTRACT

The membrane (M) glycoprotein of coronaviruses (CoVs) serves as the nidus for virion assembly. Using a yeast two-hybrid screen, we identified the interaction of the cytosolic tail of Murine Hepatitis Virus (MHV-CoV) M protein with Myosin Vb (MYO5B), specifically with the alternative splice variant of cellular MYO5B including exon D (MYO5B+D), which mediates interaction with Rab10. When co-expressed in human lung epithelial A549 and canine kidney epithelial MDCK cells, MYO5B+D co-localized with the MHV-CoV M protein, as well as with the M proteins from Porcine Epidemic Diarrhea Virus (PEDV-CoV), Middle East Respiratory Syndrome (MERS-CoV) and Severe Acute Respiratory Syndrome 2 (SARS-CoV-2). Co-expressed M proteins and MYO5B+D co-localized with endogenous Rab10 and Rab11a. We identified point mutations in MHV-CoV M that blocked the interaction with MYO5B+D in yeast 2-hybrid assays. One of these point mutations (E121K) was previously shown to block MHV-CoV virion assembly and its interaction with MYO5B+D. The E to K mutation at homologous positions in PEDV-CoV, MERS-CoV and SARS-CoV-2 M proteins also blocked colocalization with MYO5B+D. The knockdown of Rab10 blocked the co-localization of M proteins with MYO5B+D and was rescued by re-expression of CFP-Rab10. Our results suggest that CoV M proteins traffic through Rab10-containing systems, in association with MYO5B+D.


Subject(s)
Coronavirus M Proteins , Animals , Dogs , Humans , Madin Darby Canine Kidney Cells/metabolism , Madin Darby Canine Kidney Cells/virology , Middle East Respiratory Syndrome Coronavirus , Myosins , rab GTP-Binding Proteins/genetics , Saccharomyces cerevisiae , Swine , Viral Matrix Proteins , SARS-CoV-2/metabolism , Murine hepatitis virus/metabolism , A549 Cells/metabolism , A549 Cells/virology , Porcine epidemic diarrhea virus/metabolism
3.
J Virol ; 92(4)2018 02 15.
Article in English | MEDLINE | ID: mdl-29167342

ABSTRACT

There is increasing evidence to suggest that antibodies directed toward influenza A virus (IAV) neuraminidase (NA) are an important correlate of protection against influenza in humans. Moreover, the potential of NA-specific antibodies to provide broader protection than conventional hemagglutinin (HA) antibodies has been recognized. Here, we describe the isolation of two monoclonal antibodies, N1-7D3 and N1-C4, directed toward the N1 NA. N1-7D3 binds to a conserved linear epitope in the membrane-distal, carboxy-terminal part of the NA and reacted with the NA of seasonal H1N1 isolates ranging from 1977 to 2007 and the 2009 H1N1pdm virus, as well as A/Vietnam/1194/04 (H5N1). However, N1-7D3 lacked NA inhibition (NI) activity and the ability to protect BALB/c mice against a lethal challenge with a range of H1N1 viruses. Conversely, N1-C4 bound to a conformational epitope that is conserved between two influenza virus subtypes, 2009 H1N1pdm and H5N1 IAV, and displayed potent in vitro antiviral activity mediating both NI and plaque size reduction. Moreover, N1-C4 could provide heterosubtypic protection in BALB/c mice against a lethal challenge with 2009 H1N1pdm or H5N1 virus. Glutamic acid residue 311 in the NA was found to be critical for the NA binding and antiviral activity of monoclonal antibody N1-C4. Our data provide further evidence for cross-protective epitopes within the N1 subtype and highlight the potential of NA as an important target for vaccine and therapeutic approaches.IMPORTANCE Influenza remains a worldwide burden on public health. As such, the development of novel vaccines and therapeutics against influenza virus is crucial. Human challenge studies have recently highlighted the importance of antibodies directed toward the viral neuraminidase (NA) as an important correlate of reduced influenza-associated disease severity. Furthermore, there is evidence that anti-NA antibodies can provide broader protection than antibodies toward the viral hemagglutinin. Here, we describe the isolation and detailed characterization of two N1 NA-specific monoclonal antibodies. One of these monoclonal antibodies broadly binds N1-type NAs, and the second displays NA inhibition and in vitro and in vivo antiviral activity against 2009 H1N1pdm and H5N1 influenza viruses. These two new anti-NA antibodies contribute to our understanding of the antigenic properties and protective potential of the influenza virus NA antigen.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibodies, Viral/therapeutic use , Neuraminidase/immunology , Orthomyxoviridae Infections/prevention & control , Viral Proteins/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Cross Protection , Disease Models, Animal , Female , Immunization, Passive , Influenza A Virus, H1N1 Subtype , Influenza A Virus, H5N1 Subtype , Mice , Mice, Inbred BALB C
4.
Adv Virus Res ; 96: 29-57, 2016.
Article in English | MEDLINE | ID: mdl-27712627

ABSTRACT

Coronaviruses (CoVs) have a remarkable potential to change tropism. This is particularly illustrated over the last 15 years by the emergence of two zoonotic CoVs, the severe acute respiratory syndrome (SARS)- and Middle East respiratory syndrome (MERS)-CoV. Due to their inherent genetic variability, it is inevitable that new cross-species transmission events of these enveloped, positive-stranded RNA viruses will occur. Research into these medical and veterinary important pathogens-sparked by the SARS and MERS outbreaks-revealed important principles of inter- and intraspecies tropism changes. The primary determinant of CoV tropism is the viral spike (S) entry protein. Trimers of the S glycoproteins on the virion surface accommodate binding to a cell surface receptor and fusion of the viral and cellular membrane. Recently, high-resolution structures of two CoV S proteins have been elucidated by single-particle cryo-electron microscopy. Using this new structural insight, we review the changes in the S protein that relate to changes in virus tropism. Different concepts underlie these tropism changes at the cellular, tissue, and host species level, including the promiscuity or adaptability of S proteins to orthologous receptors, alterations in the proteolytic cleavage activation as well as changes in the S protein metastability. A thorough understanding of the key role of the S protein in CoV entry is critical to further our understanding of virus cross-species transmission and pathogenesis and for development of intervention strategies.


Subject(s)
Middle East Respiratory Syndrome Coronavirus/metabolism , Protein Subunits/chemistry , Receptors, Virus/chemistry , Severe acute respiratory syndrome-related coronavirus/metabolism , Spike Glycoprotein, Coronavirus/chemistry , Viral Tropism , Animals , Gene Expression , Genetic Variation , Humans , Middle East Respiratory Syndrome Coronavirus/genetics , Middle East Respiratory Syndrome Coronavirus/ultrastructure , Models, Molecular , Protein Conformation , Protein Domains , Protein Subunits/genetics , Proteolysis , Receptors, Virus/genetics , Receptors, Virus/ultrastructure , Severe acute respiratory syndrome-related coronavirus/genetics , Severe acute respiratory syndrome-related coronavirus/ultrastructure , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/ultrastructure , Virion/genetics , Virion/metabolism , Virion/ultrastructure , Virus Internalization
5.
Vaccine ; 32(41): 5323-9, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25066737

ABSTRACT

Virus replicon particles are capable of infection, genome replication and gene expression, but are unable to produce progeny virions, rendering their use inherently safe. By virtue of this unique combination of features, replicon particles hold great promise for vaccine applications. We previously developed replicon particles of Rift Valley fever virus (RVFV) and demonstrated their high efficacy as a RVFV vaccine in the natural target species. We have now investigated the feasibility of using this nonspreading RVFV (NSR) as a vaccine vector using influenza virus hemagglutinin as a model antigen. NSR particles were designed to express either the full-length hemagglutinin of influenza A virus H1N1 (NSR-HA) or the respective soluble ectodomain (NSR-sHA). The efficacies of the two NSR vector vaccines, applied via either the intramuscular or the intranasal route, were evaluated. A single vaccination with NSR-HA protected all mice from a lethal challenge dose, while vaccination with NSR-sHA was not protective. Interestingly, whereas intramuscular vaccination elicited superior systemic immune responses, intranasal vaccination provided optimal clinical protection.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza A Virus, H1N1 Subtype , Influenza Vaccines/immunology , Orthomyxoviridae Infections/prevention & control , Rift Valley fever virus/immunology , Administration, Intranasal , Animals , Antibodies, Viral/blood , Antigens, Viral/immunology , Cytokines/immunology , Female , Immunoglobulin G/blood , Injections, Intramuscular , Mice, Inbred BALB C , Replicon/immunology , Th1 Cells/immunology , Vaccination/methods
6.
Sci Rep ; 3: 3058, 2013 Oct 28.
Article in English | MEDLINE | ID: mdl-24162312

ABSTRACT

The emergence of the novel H7N9 influenza A virus (IAV) has caused global concerns about the ability of this virus to spread between humans. Analysis of the receptor-binding properties of this virus using a recombinant protein approach in combination with fetuin-binding, glycan array and human tissue-binding assays demonstrates increased binding of H7 to both α2-6 and α2-8 sialosides as well as reduced binding to α2-3-linked SIAs compared to a closely related avian H7N9 virus from 2008. These differences could be attributed to substitutions Q226L and G186V. Analysis of the enzymatic activity of the neuraminidase N9 protein indicated a reduced sialidase activity, consistent with the reduced binding of H7 to α2-3 sialosides. However, the novel H7N9 virus still preferred binding to α2-3- over α2-6-linked SIAs and was not able to efficiently bind to epithelial cells of human trachea in contrast to seasonal IAV, consistent with its limited human-to-human transmission.


Subject(s)
Fetuins/metabolism , Hemagglutinins/metabolism , Influenza A Virus, H7N9 Subtype/metabolism , Neuraminidase/metabolism , Epithelial Cells/metabolism , Fetuins/chemistry , HEK293 Cells , Hemagglutinins/genetics , Humans , Lung/metabolism , Lung/pathology , Lung/virology , Mutation , Neuraminidase/genetics , Polysaccharides/metabolism , Protein Binding , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Trachea/metabolism , Trachea/pathology , Trachea/virology
7.
J Virol ; 86(24): 13767-71, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23015725

ABSTRACT

Rift Valley fever virus (RVFV), an emerging arthropod-borne pathogen, has a broad host and cell tropism. Here we report that the glycosaminoglycan heparan sulfate, abundantly present on the surface of most animal cells, is required for efficient entry of RVFV. Entry was significantly reduced by preincubating the virus inoculum with highly sulfated heparin, by enzymatic removal of heparan sulfate from cells and in cells genetically deficient in heparan sulfate synthesis.


Subject(s)
Heparitin Sulfate/physiology , Membrane Fusion/physiology , Rift Valley fever virus/physiology , Animals , CHO Cells , Cricetinae , Cricetulus , Viral Tropism
8.
J Virol ; 85(17): 8903-12, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21697468

ABSTRACT

The binding of viruses to host cells is the first step in determining tropism and pathogenicity. While avian infectious bronchitis coronavirus (IBV) infection and avian influenza A virus (IAV) infection both depend on α2,3-linked sialic acids, the host tropism of IBV is restricted compared to that of IAV. Here we investigated whether the interaction between the viral attachment proteins and the host could explain these differences by using recombinant spike domains (S1) of IBV strains with different pathogenicities, as well as the hemagglutinin (HA) protein of IAV H5N1. Protein histochemistry showed that S1 of IBV strain M41 and HA of IAV subtype H5N1 displayed sialic acid-dependent binding to chicken respiratory tract tissue. However, while HA bound with high avidity to a broad range of α2,3-linked sialylated glycans, M41 S1 recognized only one particular α2,3-linked disialoside in a glycan array. When comparing the binding of recombinant IBV S1 proteins derived from IBV strains with known differences in tissue tropism and pathogenicity, we observed that while M41 S1 displayed binding to cilia and goblet cells of the chicken respiratory tract, S1 derived from the vaccine strain H120 or the nonvirulent Beaudette strain had reduced or no binding to chicken tissues, respectively, in agreement with the reduced abilities of these viruses to replicate in vivo. While the S1 protein derived from the nephropathogenic IBV strain B1648 also hardly displayed binding to respiratory tract cells, distinct binding to kidney cells was observed, but only after the removal of sialic acid from S1. In conclusion, our data demonstrate that the attachment patterns of the IBV S proteins correlate with the tropisms and pathogenicities of the corresponding viruses.


Subject(s)
Coronavirus/pathogenicity , Host-Pathogen Interactions , Membrane Glycoproteins/metabolism , Receptors, Virus/metabolism , Viral Envelope Proteins/metabolism , Viral Tropism , Animals , Chickens , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Influenza A Virus, H5N1 Subtype/pathogenicity , Protein Binding , Respiratory Mucosa/virology , Spike Glycoprotein, Coronavirus
9.
Vaccine ; 29(8): 1545-50, 2011 Feb 11.
Article in English | MEDLINE | ID: mdl-21219983

ABSTRACT

In 2009 a new influenza A/H1N1 virus strain ("pandemic (H1N1) 2009", H1N1v) emerged that rapidly spread around the world. The virus is suspected to have originated in swine through reassortment and to have subsequently crossed the species-barrier towards humans. Several cases of reintroduction into pigs have since been reported, which could possibly create a reservoir for human exposure or ultimately become endemic in the pig population with similar clinical disease problems as current swine influenza strains. A soluble trimer of hemagglutinin (HA), derived from the H1N1v, was used as a vaccine in pigs to investigate the extent to which this vaccine would be able to protect pigs against infection with the H1N1v influenza strain, especially with respect to reducing virus replication and excretion. In a group of unvaccinated control pigs, no clinical symptoms were observed, but (histo)pathological changes consistent with an influenza infection were found on days 1 and 3 after inoculation. Live virus was isolated from the upper and lower respiratory tract, with titres up to 10(6) TCID(50) per gram of tissue. Furthermore, live virus was detected in brain samples. Control pigs were shedding live virus for up to 6 days after infection, with titres of up to 10(5) TCID(50) per nasal or oropharyngeal swab. The soluble H1N1v HA trimer diminished virus replication and excretion after a double vaccination and subsequent challenge. Live virus could not be detected in any of the samples taken from the vaccinated pigs. Vaccines based on soluble HA trimers provide an attractive alternative to the current inactivated vaccines.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/prevention & control , Animals , Antibodies, Viral/blood , Hemagglutination Inhibition Tests , Neutralization Tests , Orthomyxoviridae Infections/immunology , Recombinant Proteins/immunology , Swine , Vaccines, Synthetic/immunology , Virus Shedding
10.
J Virol ; 82(12): 6078-83, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18400867

ABSTRACT

A longstanding enigmatic feature of the group 1 coronaviruses is the uncleaved phenotype of their spike protein, an exceptional property among class I fusion proteins. Here, however, we show that some group 1 coronavirus spike proteins carry a furin enzyme recognition motif and can actually be cleaved, as demonstrated for a feline coronavirus. Interestingly, this feature can be lost during cell culture adaptation by a single mutation in the cleavage motif; this, however, preserves a heparan sulfate binding motif and renders infection by the virus heparan sulfate dependent. We identified a similar cell culture adaptation for the human coronavirus OC43.


Subject(s)
Adaptation, Physiological , Furin/physiology , Heparitin Sulfate/metabolism , Membrane Glycoproteins/metabolism , Viral Envelope Proteins/metabolism , Amino Acid Sequence , Animals , Cats , Cell Line , Humans , Membrane Glycoproteins/chemistry , Molecular Sequence Data , Spike Glycoprotein, Coronavirus , Viral Envelope Proteins/chemistry
11.
J Virol ; 81(22): 12323-36, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17855519

ABSTRACT

The coronavirus nonstructural proteins (nsp's) derived from the replicase polyproteins collectively constitute the viral replication complexes, which are anchored to double-membrane vesicles. Little is known about the biogenesis of these complexes, the membrane anchoring of which is probably mediated by nsp3, nsp4, and nsp6, as they contain several putative transmembrane domains. As a first step to getting more insight into the formation of the coronavirus replication complex, the membrane topology, processing, and subcellular localization of nsp4 of the mouse hepatitis virus (MHV) and severe acute respiratory syndrome-associated coronavirus (SARS-CoV) were elucidated in this study. Both nsp4 proteins became N glycosylated, while their amino and carboxy termini were localized to the cytoplasm. These observations imply nsp4 to assemble in the membrane as a tetraspanning transmembrane protein with a Nendo/Cendo topology. The amino terminus of SARS-CoV nsp4, but not that of MHV nsp4, was shown to be (partially) processed by signal peptidase. nsp4 localized to the endoplasmic reticulum (ER) when expressed alone but was recruited to the replication complexes in infected cells. nsp4 present in these complexes did not colocalize with markers of the ER or Golgi apparatus, while the susceptibility of its sugars to endoglycosidase H indicated that the protein had also not traveled trough the latter compartment. The important role of the early secretory pathway in formation of the replication complexes was also demonstrated by the inhibition of coronaviral replication when the ER export machinery was blocked by use of the kinase inhibitor H89 or by expression of a mutant, Sar1[H79G].


Subject(s)
Cell Membrane/enzymology , RNA-Dependent RNA Polymerase/analysis , RNA-Dependent RNA Polymerase/metabolism , Severe acute respiratory syndrome-related coronavirus/physiology , Viral Nonstructural Proteins/analysis , Viral Nonstructural Proteins/metabolism , Virus Replication , Animals , Cats , Cell Line , Cell Membrane/virology , Computational Biology , Endoplasmic Reticulum/enzymology , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Mice , Murine hepatitis virus/genetics , Murine hepatitis virus/metabolism , RNA-Dependent RNA Polymerase/genetics , Recombinant Proteins/analysis , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Severe acute respiratory syndrome-related coronavirus/enzymology , Viral Nonstructural Proteins/genetics
12.
J Antimicrob Chemother ; 60(4): 741-9, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17704516

ABSTRACT

OBJECTIVES: Many enveloped viruses carry carbohydrate-containing proteins on their surface. These glycoproteins are key to the infection process as they are mediators of the receptor binding and membrane fusion of the virion with the host cell. Therefore, they are attractive therapeutic targets for the development of novel antiviral therapies. Recently, carbohydrate-binding agents (CBA) were shown to possess antiviral activity towards coronaviruses. The current study further elucidates the inhibitory mode of action of CBA. METHODS: Different strains of two coronaviruses, mouse hepatitis virus and feline infectious peritonitis virus, were exposed to CBA: the plant lectins Galanthus nivalis agglutinin, Hippeastrum hybrid agglutinin and Urtica dioica agglutinin (UDA) and the non-peptidic mannose-binding antibiotic pradimicin A. RESULTS AND CONCLUSIONS: Our results indicate that CBA target the two glycosylated envelope glycoproteins, the spike (S) and membrane (M) protein, of mouse hepatitis virus and feline infectious peritonitis virus. Furthermore, CBA did not inhibit virus-cell attachment, but rather affected virus entry at a post-binding stage. The sensitivity of coronaviruses towards CBA was shown to be dependent on the processing of the N-linked carbohydrates. Inhibition of mannosidases in host cells rendered the progeny viruses more sensitive to the mannose-binding agents and even to the N-acetylglucosamine-binding UDA. In addition, inhibition of coronaviruses was shown to be dependent on the cell-type used to grow the virus stocks. All together, these results show that CBA exhibit promising capabilities to inhibit coronavirus infections.


Subject(s)
Anthracyclines/metabolism , Antiviral Agents/metabolism , Coronavirus, Feline/drug effects , Membrane Glycoproteins/metabolism , Murine hepatitis virus/drug effects , Plant Lectins/metabolism , Viral Envelope Proteins/metabolism , Viral Matrix Proteins/metabolism , Animals , Anthracyclines/pharmacology , Antiviral Agents/pharmacology , Cats , Cell Line , Coronavirus M Proteins , Mice , Plant Lectins/pharmacology , Spike Glycoprotein, Coronavirus , Virus Attachment/drug effects , Virus Internalization/drug effects
13.
Antiviral Res ; 76(1): 21-9, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17560666

ABSTRACT

Coronaviruses are important human and animal pathogens, the relevance of which increased due to the emergence of new human coronaviruses like SARS-CoV, HKU1 and NL63. Together with toroviruses, arteriviruses, and roniviruses the coronaviruses belong to the order Nidovirales. So far antivirals are hardly available to combat infections with viruses of this order. Therefore, various antiviral strategies to counter nidoviral infections are under evaluation. Lectins, which bind to N-linked oligosaccharide elements of enveloped viruses, can be considered as a conceptionally new class of virus inhibitors. These agents were recently evaluated for their antiviral activity towards a variety of enveloped viruses and were shown in most cases to inhibit virus infection at low concentrations. However, limited knowledge is available for their efficacy towards nidoviruses. In this article the application of the plant lectins Hippeastrum hybrid agglutinin (HHA), Galanthus nivalis agglutinin (GNA), Cymbidium sp. agglutinin (CA) and Urtica dioica agglutinin (UDA) as well as non-plant derived pradimicin-A (PRM-A) and cyanovirin-N (CV-N) as potential antiviral agents was evaluated. Three antiviral tests were compared based on different evaluation principles: cell viability (MTT-based colorimetric assay), number of infected cells (immunoperoxidase assay) and amount of viral protein expression (luciferase-based assay). The presence of carbohydrate-binding agents strongly inhibited coronaviruses (transmissible gastroenteritis virus, infectious bronchitis virus, feline coronaviruses serotypes I and II, mouse hepatitis virus), arteriviruses (equine arteritis virus and porcine respiratory and reproductive syndrome virus) and torovirus (equine Berne virus). Remarkably, serotype II feline coronaviruses and arteriviruses were not inhibited by PRM-A, in contrast to the other viruses tested.


Subject(s)
Nidovirales/drug effects , Plant Lectins/pharmacology , Animals , Anthracyclines/pharmacology , Antiviral Agents/pharmacology , Bacterial Proteins/pharmacology , Carrier Proteins/pharmacology , Cats , Cell Line , Chlorocebus aethiops , Colorimetry/methods , Female , Galanthus/chemistry , Immunohistochemistry , Liliaceae/chemistry , Luciferases/genetics , Magnoliopsida/chemistry , Mice , Microbial Sensitivity Tests , Nidovirales/genetics , Plant Lectins/isolation & purification , RNA Virus Infections/virology , Swine , Tetrazolium Salts , Thiazoles , Urtica dioica/chemistry
15.
J Virol ; 80(5): 2326-36, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16474139

ABSTRACT

The severe acute respiratory syndrome coronavirus (SARS-CoV) open reading frame 3a protein has recently been shown to be a structural protein. The protein is encoded by one of the so-called group-specific genes and has no sequence homology with any of the known structural or group-specific proteins of coronaviruses. It does, however, have several similarities to the coronavirus M proteins; (i) they are triple membrane spanning with the same topology, (ii) they have similar intracellular localizations (predominantly Golgi), (iii) both are viral structural proteins, and (iv) they appear to interact with the E and S proteins, as well as with each other. The M protein plays a crucial role in coronavirus assembly and is glycosylated in all coronaviruses, either by N-linked or by O-linked oligosaccharides. The conserved glycosylation of the coronavirus M proteins and the resemblance of the 3a protein to them led us to investigate the glycosylation of these two SARS-CoV membrane proteins. The proteins were expressed separately using the vaccinia virus T7 expression system, followed by metabolic labeling. Pulse-chase analysis showed that both proteins were modified, although in different ways. While the M protein acquired cotranslationally oligosaccharides that could be removed by PNGaseF, the 3a protein acquired its modifications posttranslationally, and they were not sensitive to the N-glycosidase enzyme. The SARS-CoV 3a protein, however, was demonstrated to contain sialic acids, indicating the presence of oligosaccharides. O-glycosylation of the 3a protein was indeed confirmed using an in situ O-glycosylation assay of endoplasmic reticulum-retained mutants. In addition, we showed that substitution of serine and threonine residues in the ectodomain of the 3a protein abolished the addition of the O-linked sugars. Thus, the SARS-CoV 3a protein is an O-glycosylated glycoprotein, like the group 2 coronavirus M proteins but unlike the SARS-CoV M protein, which is N glycosylated.


Subject(s)
Protein Modification, Translational , Protein Processing, Post-Translational , Severe acute respiratory syndrome-related coronavirus/chemistry , Viral Matrix Proteins/metabolism , Viral Proteins/metabolism , Amino Acid Sequence , Coronavirus M Proteins , Gene Expression , Glycosylation , Molecular Sequence Data , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/metabolism , Radioisotopes , Sialic Acids/analysis , Staining and Labeling , Viral Envelope Proteins , Viral Matrix Proteins/chemistry , Viral Proteins/chemistry , Viroporin Proteins
16.
J Virol ; 80(3): 1250-60, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16415002

ABSTRACT

Murine hepatitis coronavirus (MHV)-A59 infection depends on the interaction of its spike (S) protein with the cellular receptor mCEACAM1a present on murine cells. Human cells lack this receptor and are therefore not susceptible to MHV. Specific alleviation of the tropism barrier by redirecting MHV to a tumor-specific receptor could lead to a virus with appealing properties for tumor therapy. To demonstrate that MHV can be retargeted to a nonnative receptor on human cells, we produced bispecific adapter proteins composed of the N-terminal D1 domain of mCEACAM1a linked to a short targeting peptide, the six-amino-acid His tag. Preincubation of MHV with the adapter proteins and subsequent inoculation of human cells expressing an artificial His receptor resulted in infection of these otherwise nonsusceptible cells and led to subsequent production of progeny virus. To generate a self-targeted virus able to establish multiround infection of the target cells, we subsequently incorporated the gene encoding the bispecific adapter protein as an additional expression cassette into the MHV genome through targeted RNA recombination. When inoculated onto murine LR7 cells, the resulting recombinant virus indeed expressed the adapter protein. Furthermore, inoculation of human target cells with the virus resulted in a His receptor-specific infection that was multiround. Extensive cell-cell fusion and rapid cell killing of infected target cells was observed. Our results show that MHV can be genetically redirected via adapters composed of the S protein binding part of mCEACAM1a and a targeting peptide recognizing a nonnative receptor expressed on human cells, consequently leading to rapid cell death. The results provide interesting leads for further investigations of the use of coronaviruses as antitumor agents.


Subject(s)
Murine hepatitis virus/physiology , Murine hepatitis virus/pathogenicity , Receptors, Virus/physiology , Animals , Base Sequence , Binding Sites , Carcinoembryonic Antigen/genetics , Carcinoembryonic Antigen/physiology , Cats , Cell Line , DNA, Recombinant/genetics , Gene Products, vif/genetics , Gene Products, vif/physiology , Humans , Membrane Fusion , Membrane Glycoproteins/genetics , Membrane Glycoproteins/physiology , Mice , Murine hepatitis virus/genetics , Receptors, Virus/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Spike Glycoprotein, Coronavirus , Viral Envelope Proteins/genetics , Viral Envelope Proteins/physiology , Virus Replication
17.
J Virol ; 79(24): 15314-22, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16306602

ABSTRACT

The mouse hepatitis coronavirus (MHV) infects murine cells by binding of its spike (S) protein to murine CEACAM1a. The N-terminal part of this cellular receptor (soR) is sufficient for S binding and for subsequent induction of the conformational changes required for virus-cell membrane fusion. Here we analyzed whether these characteristics can be used to redirect MHV to human cancer cells. To this end, the soR domain was coupled to single-chain monoclonal antibody 425, which is directed against the human epidermal growth factor receptor (EGFR), resulting in a bispecific adapter protein (soR-425). The soR and soR-425 proteins, both produced with the vaccinia virus system, were able to neutralize MHV infection of murine LR7 cells. However, only soR-425 was able to target MHV to human EGFR-expressing cancer cells. Interestingly, the targeted infections induced syncytium formation. Furthermore, the soR-425-mediated infections were blocked by heptad repeat-mimicking peptides, indicating that virus entry requires the regular S protein fusion process. We conclude that the specific spike-binding property of the CEACAM1a N-terminal fragment can be exploited to direct the virus to selected cells by linking it to a moiety able to bind a receptor on those cells. This approach might be useful in the development of tumor-targeted coronaviruses.


Subject(s)
Coronavirus Infections/metabolism , ErbB Receptors/metabolism , Membrane Glycoproteins/physiology , Murine hepatitis virus/physiology , Viral Envelope Proteins/physiology , Animals , Coronavirus Infections/immunology , Humans , Membrane Glycoproteins/chemistry , Mice , Spike Glycoprotein, Coronavirus , Viral Envelope Proteins/chemistry
18.
Gene Ther ; 12(18): 1394-404, 2005 Sep.
Article in English | MEDLINE | ID: mdl-15843808

ABSTRACT

To explore the potential of using non-human coronaviruses for cancer therapy, we first established their ability to kill human tumor cells. We found that the feline infectious peritonitis virus (FIPV) and a felinized murine hepatitis virus (fMHV), both normally incapable of infecting human cells, could rapidly and effectively kill human cancer cells artificially expressing the feline coronavirus receptor aminopeptidase N. Also 3-D multilayer tumor spheroids established from such cells were effectively eradicated. Next, we investigated whether FIPV and fMHV could be targeted to human cancer cells by constructing a bispecific single-chain antibody directed on the one hand against the feline coronavirus spike protein--responsible for receptor binding and subsequent cell entry through virus-cell membrane fusion--and on the other hand against the human epidermal growth factor receptor (EGFR). The targeting antibody mediated specific infection of EGFR-expressing human cancer cells by both coronaviruses. Furthermore, in the presence of the targeting antibody, infected cancer cells formed syncytia typical of productive coronavirus infection. By their potent cytotoxicity, the selective targeting of non-human coronaviruses to human cancer cells provides a rationale for further investigations into the use of these viruses as anticancer agents.


Subject(s)
Antibodies, Bispecific/administration & dosage , Gene Targeting/methods , Neoplasms/therapy , Oncolytic Virotherapy/methods , Oncolytic Viruses/genetics , Animals , Antibodies, Bispecific/genetics , CD13 Antigens/genetics , Cats , Coronavirus Infections/immunology , Coronavirus Infections/metabolism , Coronavirus, Feline/genetics , Cytotoxicity, Immunologic , ErbB Receptors/immunology , Feline Infectious Peritonitis/metabolism , Humans , Membrane Glycoproteins/immunology , Mice , Murine hepatitis virus/genetics , Neoplasms/immunology , Neoplasms/virology , Protein Transport , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Spike Glycoprotein, Coronavirus , Viral Envelope Proteins/immunology
20.
J Virol ; 74(11): 4967-78, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10799570

ABSTRACT

The viral membrane proteins M and E are the minimal requirements for the budding of coronavirus particles. Since the E protein occurs in particles only in trace amounts, the lateral interactions between the M proteins apparently generate the major driving force for envelope formation. By using coimmunoprecipitation and envelope incorporation assays, we provide extensive evidence for the existence of such M-M interactions. In addition, we determined which domains of the M protein are involved in this homotypic association, using a mutagenetic approach. Mutant M proteins which were not able to assemble into viruslike particles (VLPs) by themselves (C. A. M. de Haan, L. Kuo, P. S. Masters, H. Vennema, and P. J. M. Rottier, J. Virol. 72:6838-6850, 1998) were tested for the ability to associate with other M proteins and to be rescued into VLPs formed by assembly-competent M proteins. We found that M proteins lacking parts of the transmembrane cluster, of the amphipathic domain, or of the hydrophilic carboxy-terminal tail, or M proteins that had their luminal domain replaced by heterologous ectodomains, were still able to associate with assembly-competent M proteins, resulting in their coincorporation into VLPs. Only a mutant M protein in which all three transmembrane domains had been replaced lost this ability. The results indicate that M protein molecules interact with each other through multiple contact sites, particularly at the transmembrane level. Finally, we tested the stringency with which membrane proteins are selected for incorporation into the coronavirus envelope by probing the coassembly of some foreign proteins. The observed efficient exclusion from budding of the vesicular stomatitis virus G protein and the equine arteritis virus M protein indicates that envelope assembly is indeed a highly selective sorting process. The low but detectable incorporation of CD8 molecules, however, demonstrated that this process is not perfect.


Subject(s)
Murine hepatitis virus/metabolism , Viral Envelope Proteins/biosynthesis , Viral Matrix Proteins/metabolism , Virus Assembly/physiology , Animals , Binding Sites , Cell Compartmentation , Chromosome Mapping , Coronavirus M Proteins , Golgi Apparatus , Humans , Mice , Murine hepatitis virus/genetics , Murine hepatitis virus/physiology , Rabbits , Viral Matrix Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...