Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Virus Res ; 259: 28-37, 2019 01 02.
Article in English | MEDLINE | ID: mdl-30296457

ABSTRACT

Morbilliviruses (e.g. measles virus [MeV] or canine distemper virus [CDV]) employ the attachment (H) and fusion (F) envelope glycoproteins for cell entry. H protein engagement to a cognate receptor eventually leads to F-triggering. Upon activation, F proteins transit from a prefusion to a postfusion conformation; a refolding process that is associated with membrane merging. Small-molecule morbilliviral fusion inhibitors such as the compound 3G (a chemical analog in the AS-48 class) were previously generated and mechanistic studies revealed a stabilizing effect on morbilliviral prefusion F trimers. Here, we aimed at designing 3G-resistant CDV F mutants by introducing single cysteine residues at hydrophobic core positions of the helical stalk region. Covalently-linked F dimers were generated, which highlighted substantial conformational flexibility within the stalk to achieve those irregular F conformations. Our findings demonstrate that "top-stalk" CDV F cysteine mutants (F-V571C and F-L575C) remained functional and gained resistance to 3G. Conversely, although not all "bottom-stalk" F cysteine variants preserved proper bioactivity, those that remained functional exhibited 3G-sensitivity. According to the recently determined prefusion MeV F trimer/AS-48 co-crystal structure, CDV residues F-V571 and F-L575 may directly interact with 3G. A combination of conformation-specific anti-F antibodies and low-resolution electron microscopy structural analyses confirmed that 3G lost its stabilizing effect on "top-stalk" F cysteine mutants thus suggesting a primary resistance mechanism. Overall, our data suggest that the fusion inhibitor 3G stabilizes prefusion CDV F trimers by docking at the top of the stalk domain.


Subject(s)
Antiviral Agents/pharmacology , Distemper Virus, Canine/drug effects , Distemper Virus, Canine/physiology , Drug Resistance, Viral , Viral Fusion Proteins/antagonists & inhibitors , Amino Acid Sequence , Animals , Cell Line , Chlorocebus aethiops , Distemper , Models, Molecular , Mutation , Protein Conformation , Vero Cells , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/genetics , Viral Fusion Proteins/metabolism
2.
PLoS Pathog ; 11(5): e1004880, 2015 May.
Article in English | MEDLINE | ID: mdl-25946112

ABSTRACT

Despite large vaccination campaigns, measles virus (MeV) and canine distemper virus (CDV) cause major morbidity and mortality in humans and animals, respectively. The MeV and CDV cell entry system relies on two interacting envelope glycoproteins: the attachment protein (H), consisting of stalk and head domains, co-operates with the fusion protein (F) to mediate membrane fusion. However, how receptor-binding by the H-protein leads to F-triggering is not fully understood. Here, we report that an anti-CDV-H monoclonal antibody (mAb-1347), which targets the linear H-stalk segment 126-133, potently inhibits membrane fusion without interfering with H receptor-binding or F-interaction. Rather, mAb-1347 blocked the F-triggering function of H-proteins regardless of the presence or absence of the head domains. Remarkably, mAb-1347 binding to headless CDV H, as well as standard and engineered bioactive stalk-elongated CDV H-constructs treated with cells expressing the SLAM receptor, was enhanced. Despite proper cell surface expression, fusion promotion by most H-stalk mutants harboring alanine substitutions in the 126-138 "spacer" section was substantially impaired, consistent with deficient receptor-induced mAb-1347 binding enhancement. However, a previously reported F-triggering defective H-I98A variant still exhibited the receptor-induced "head-stalk" rearrangement. Collectively, our data spotlight a distinct mechanism for morbillivirus membrane fusion activation: prior to receptor contact, at least one of the morbillivirus H-head domains interacts with the membrane-distal "spacer" domain in the H-stalk, leaving the F-binding site located further membrane-proximal in the stalk fully accessible. This "head-to-spacer" interaction conformationally stabilizes H in an auto-repressed state, which enables intracellular H-stalk/F engagement while preventing the inherent H-stalk's bioactivity that may prematurely activate F. Receptor-contact disrupts the "head-to-spacer" interaction, which subsequently "unlocks" the stalk, allowing it to rearrange and trigger F. Overall, our study reveals essential mechanistic requirements governing the activation of the morbillivirus membrane fusion cascade and spotlights the H-stalk "spacer" microdomain as a possible drug target for antiviral therapy.


Subject(s)
Antigens, CD/metabolism , Cell Adhesion Molecules/metabolism , Models, Molecular , Morbillivirus/physiology , Receptors, Cell Surface/metabolism , Viral Proteins/metabolism , Virus Internalization , Amino Acid Substitution , Animals , Antibodies, Monoclonal/pharmacology , Antigens, CD/chemistry , Antigens, CD/genetics , Cell Adhesion Molecules/chemistry , Cell Adhesion Molecules/genetics , Chlorocebus aethiops , Distemper Virus, Canine/metabolism , Dogs , HEK293 Cells , Humans , Membrane Fusion/drug effects , Morbillivirus/drug effects , Mutation , Protein Conformation , Protein Folding/drug effects , Protein Interaction Domains and Motifs , Protein Stability/drug effects , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Signaling Lymphocytic Activation Molecule Family Member 1 , Vero Cells , Viral Proteins/antagonists & inhibitors , Viral Proteins/chemistry , Viral Proteins/genetics , Virus Attachment/drug effects , Virus Internalization/drug effects
3.
J Virol ; 89(10): 5724-33, 2015 May.
Article in English | MEDLINE | ID: mdl-25787275

ABSTRACT

UNLABELLED: Measles and canine distemper viruses (MeV and CDV, respectively) first replicate in lymphatic and epithelial tissues by using SLAM and nectin-4 as entry receptors, respectively. The viruses may also invade the brain to establish persistent infections, triggering fatal complications, such as subacute sclerosis pan-encephalitis (SSPE) in MeV infection or chronic, multiple sclerosis-like, multifocal demyelinating lesions in the case of CDV infection. In both diseases, persistence is mediated by viral nucleocapsids that do not require packaging into particles for infectivity but are directly transmitted from cell to cell (neurons in SSPE or astrocytes in distemper encephalitis), presumably by relying on restricted microfusion events. Indeed, although morphological evidence of fusion remained undetectable, viral fusion machineries and, thus, a putative cellular receptor, were shown to contribute to persistent infections. Here, we first showed that nectin-4-dependent cell-cell fusion in Vero cells, triggered by a demyelinating CDV strain, remained extremely limited, thereby supporting a potential role of nectin-4 in mediating persistent infections in astrocytes. However, nectin-4 could not be detected in either primary cultured astrocytes or the white matter of tissue sections. In addition, a bioengineered "nectin-4-blind" recombinant CDV retained full cell-to-cell transmission efficacy in primary astrocytes. Combined with our previous report demonstrating the absence of SLAM expression in astrocytes, these findings are suggestive for the existence of a hitherto unrecognized third CDV receptor expressed by glial cells that contributes to the induction of noncytolytic cell-to-cell viral transmission in astrocytes. IMPORTANCE: While persistent measles virus (MeV) infection induces SSPE in humans, persistent canine distemper virus (CDV) infection causes chronic progressive or relapsing demyelination in carnivores. Common to both central nervous system (CNS) infections is that persistence is based on noncytolytic cell-to-cell spread, which, in the case of CDV, was demonstrated to rely on functional membrane fusion machinery complexes. This inferred a mechanism where nucleocapsids are transmitted through macroscopically invisible microfusion events between infected and target cells. Here, we provide evidence that CDV induces such microfusions in a SLAM- and nectin-4-independent manner, thereby strongly suggesting the existence of a third receptor expressed in glial cells (referred to as GliaR). We propose that GliaR governs intercellular transfer of nucleocapsids and hence contributes to viral persistence in the brain and ensuing demyelinating lesions.


Subject(s)
Antigens, CD/metabolism , Astrocytes/virology , Cell Adhesion Molecules/metabolism , Distemper Virus, Canine/physiology , Distemper Virus, Canine/pathogenicity , Receptors, Cell Surface/metabolism , Amino Acid Substitution , Animals , Antigens, CD/genetics , Brain/metabolism , Brain/virology , Cell Adhesion Molecules/genetics , Cells, Cultured , Chlorocebus aethiops , Distemper/metabolism , Distemper/transmission , Distemper/virology , Distemper Virus, Canine/genetics , Dogs , Genes, Viral , Host-Pathogen Interactions , Humans , Measles virus/pathogenicity , Nectins , Receptors, Cell Surface/genetics , Receptors, Virus/genetics , Receptors, Virus/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signaling Lymphocytic Activation Molecule Family Member 1 , Subacute Sclerosing Panencephalitis/etiology , Vero Cells , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Internalization
4.
J Virol ; 89(2): 1445-51, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25355896

ABSTRACT

Membrane fusion for morbillivirus cell entry relies on critical interactions between the viral fusion (F) and attachment (H) envelope glycoproteins. Through extensive mutagenesis of an F cavity recently proposed to contribute to F's interaction with the H protein, we identified two neighboring hydrophobic residues responsible for severe F-to-H binding and fusion-triggering deficiencies when they were mutated in combination. Since both residues reside on one side of the F cavity, the data suggest that H binds the F globular head domain sideways.


Subject(s)
Distemper Virus, Canine/physiology , Protein Multimerization , Viral Envelope Proteins/metabolism , Virus Internalization , Animals , DNA Mutational Analysis , Distemper Virus, Canine/genetics , Models, Molecular , Protein Binding , Protein Conformation , Viral Envelope Proteins/genetics
5.
J Virol ; 88(14): 8057-64, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24807725

ABSTRACT

The hemagglutinin (H) gene of canine distemper virus (CDV) encodes the receptor-binding protein. This protein, together with the fusion (F) protein, is pivotal for infectivity since it contributes to the fusion of the viral envelope with the host cell membrane. Of the two receptors currently known for CDV (nectin-4 and the signaling lymphocyte activation molecule [SLAM]), SLAM is considered the most relevant for host susceptibility. To investigate how evolution might have impacted the host-CDV interaction, we examined the functional properties of a series of missense single nucleotide polymorphisms (SNPs) naturally accumulating within the H-gene sequences during the transition between two distinct but related strains. The two strains, a wild-type strain and a consensus strain, were part of a single continental outbreak in European wildlife and occurred in distinct geographical areas 2 years apart. The deduced amino acid sequence of the two H genes differed at 5 residues. A panel of mutants carrying all the combinations of the SNPs was obtained by site-directed mutagenesis. The selected mutant, wild type, and consensus H proteins were functionally evaluated according to their surface expression, SLAM binding, fusion protein interaction, and cell fusion efficiencies. The results highlight that the most detrimental functional effects are associated with specific sets of SNPs. Strikingly, an efficient compensational system driven by additional SNPs appears to come into play, virtually neutralizing the negative functional effects. This system seems to contribute to the maintenance of the tightly regulated function of the H-gene-encoded attachment protein. Importance: To investigate how evolution might have impacted the host-canine distemper virus (CDV) interaction, we examined the functional properties of naturally occurring single nucleotide polymorphisms (SNPs) in the hemagglutinin gene of two related but distinct strains of CDV. The hemagglutinin gene encodes the attachment protein, which is pivotal for infection. Our results show that few SNPs have a relevant detrimental impact and they generally appear in specific combinations (molecular signatures). These drastic negative changes are neutralized by compensatory mutations, which contribute to maintenance of an overall constant bioactivity of the attachment protein. This compensational mechanism might reflect the reaction of the CDV machinery to the changes occurring in the virus following antigenic variations critical for virulence.


Subject(s)
Amino Acid Substitution , Distemper Virus, Canine/genetics , Distemper Virus, Canine/physiology , Hemagglutinins, Viral/genetics , Hemagglutinins, Viral/metabolism , Mutation, Missense , Virus Attachment , Animals , Animals, Wild , Antigens, CD/metabolism , DNA Mutational Analysis , Distemper/epidemiology , Distemper/virology , Distemper Virus, Canine/isolation & purification , Europe/epidemiology , Evolution, Molecular , Mutagenesis, Site-Directed , Mutant Proteins/genetics , Mutant Proteins/metabolism , Protein Binding , Receptors, Cell Surface/metabolism , Receptors, Virus/metabolism , Signaling Lymphocytic Activation Molecule Family Member 1 , Suppression, Genetic , Viral Fusion Proteins/metabolism
6.
J Virol ; 88(5): 2951-66, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24371057

ABSTRACT

UNLABELLED: The morbillivirus cell entry machinery consists of a fusion (F) protein trimer that refolds to mediate membrane fusion following receptor-induced conformational changes in its binding partner, the tetrameric attachment (H) protein. To identify molecular determinants that control F refolding, we generated F chimeras between measles virus (MeV) and canine distemper virus (CDV). We located a central pocket in the globular head domain of CDV F that regulates the stability of the metastable, prefusion conformational state of the F trimer. Most mutations introduced into this "pocket'" appeared to mediate a destabilizing effect, a phenotype associated with enhanced membrane fusion activity. Strikingly, under specific triggering conditions (i.e., variation of receptor type and H protein origin), some F mutants also exhibited resistance to a potent morbillivirus entry inhibitor, which is known to block F triggering by enhancing the stability of prefusion F trimers. Our data reveal that the molecular nature of the F stimulus and the intrinsic stability of metastable prefusion F both regulate the efficiency of F refolding and escape from small-molecule refolding blockers. IMPORTANCE: With the aim to better characterize the thermodynamic basis of morbillivirus membrane fusion for cell entry and spread, we report here that the activation energy barrier of prefusion F trimers together with the molecular nature of the triggering "stimulus" (attachment protein and receptor types) define a "triggering range," which governs the initiation of the membrane fusion process. A central "pocket" microdomain in the globular F head contributes substantially to the regulation of the conformational stability of the prefusion complexes. The triggering range also defines the mechanism of viral escape from entry inhibitors and describes how the cellular environment can affect membrane fusion efficiency.


Subject(s)
Distemper Virus, Canine/physiology , Membrane Fusion , Viral Fusion Proteins/metabolism , Amino Acid Substitution , Animals , CHO Cells , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Fusion , Chlorocebus aethiops , Cricetulus , Dogs , Models, Molecular , Mutation , Nectins , Protein Binding , Protein Conformation , Protein Multimerization , Protein Stability , Receptors, Virus/metabolism , Vero Cells , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/genetics , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Internalization
SELECTION OF CITATIONS
SEARCH DETAIL
...