Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Elife ; 112022 04 27.
Article in English | MEDLINE | ID: mdl-35475759

ABSTRACT

Host proteins sense viral products and induce defence mechanisms, particularly in immune cells. Using cell-free assays and quantitative mass spectrometry, we determined the interactome of capsid-host protein complexes of herpes simplex virus and identified the large dynamin-like GTPase myxovirus resistance protein B (MxB) as an interferon-inducible protein interacting with capsids. Electron microscopy analyses showed that cytosols containing MxB had the remarkable capability to disassemble the icosahedral capsids of herpes simplex viruses and varicella zoster virus into flat sheets of connected triangular faces. In contrast, capsids remained intact in cytosols with MxB mutants unable to hydrolyse GTP or to dimerize. Our data suggest that MxB senses herpesviral capsids, mediates their disassembly, and thereby restricts the efficiency of nuclear targeting of incoming capsids and/or the assembly of progeny capsids. The resulting premature release of viral genomes from capsids may enhance the activation of DNA sensors, and thereby amplify the innate immune responses.


Subject(s)
Capsid , Herpesviridae , Capsid/metabolism , Capsid Proteins/metabolism , GTP Phosphohydrolases/metabolism , Interferons/metabolism , Simplexvirus
2.
J Exp Med ; 217(7)2020 07 06.
Article in English | MEDLINE | ID: mdl-32383759

ABSTRACT

Herpes simplex virus (HSV) is the main cause of viral encephalitis in the Western world, and the type I interferon (IFN) system is important for antiviral control in the brain. Here, we have compared Ifnb induction in mixed murine brain cell cultures by a panel of HSV1 mutants, each devoid of one mechanism to counteract the IFN-stimulating cGAS-STING pathway. We found that a mutant lacking the deubiquitinase (DUB) activity of the VP1-2 protein induced particularly strong expression of Ifnb and IFN-stimulated genes. HSV1 ΔDUB also induced elevated IFN expression in murine and human microglia and exhibited reduced viral replication in the brain. This was associated with increased ubiquitination of STING and elevated phosphorylation of STING, TBK1, and IRF3. VP1-2 associated directly with STING, leading to its deubiquitination. Recruitment of VP1-2 to STING was dependent on K150 of STING, which was ubiquitinated by TRIM32. Thus, the DUB activity of HSV1 VP1-2 is a major viral immune-evasion mechanism in the brain.


Subject(s)
Brain/virology , Deubiquitinating Enzymes/metabolism , Herpesvirus 1, Human/metabolism , Interferon Type I/metabolism , Membrane Proteins/metabolism , Viral Proteins/metabolism , Animals , Brain/pathology , Cells, Cultured , Cytoplasm/metabolism , DNA, Viral/metabolism , HEK293 Cells , Humans , Lysine/metabolism , Mice, Inbred C57BL , Microglia/metabolism , Mutation/genetics , Nucleotidyltransferases/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Ubiquitin/metabolism , Ubiquitination , Virus Replication/physiology
3.
PLoS Pathog ; 13(12): e1006813, 2017 12.
Article in English | MEDLINE | ID: mdl-29284065

ABSTRACT

Upon reactivation from latency and during lytic infections in neurons, alphaherpesviruses assemble cytosolic capsids, capsids associated with enveloping membranes, and transport vesicles harboring fully enveloped capsids. It is debated whether capsid envelopment of herpes simplex virus (HSV) is completed in the soma prior to axonal targeting or later, and whether the mechanisms are the same in neurons derived from embryos or from adult hosts. We used HSV mutants impaired in capsid envelopment to test whether the inner tegument proteins pUL36 or pUL37 necessary for microtubule-mediated capsid transport were sufficient for axonal capsid targeting in neurons derived from the dorsal root ganglia of adult mice. Such neurons were infected with HSV1-ΔUL20 whose capsids recruited pUL36 and pUL37, with HSV1-ΔUL37 whose capsids associate only with pUL36, or with HSV1-ΔUL36 that assembles capsids lacking both proteins. While capsids of HSV1-ΔUL20 were actively transported along microtubules in epithelial cells and in the somata of neurons, those of HSV1-ΔUL36 and -ΔUL37 could only diffuse in the cytoplasm. Employing a novel image analysis algorithm to quantify capsid targeting to axons, we show that only a few capsids of HSV1-ΔUL20 entered axons, while vesicles transporting gD utilized axonal transport efficiently and independently of pUL36, pUL37, or pUL20. Our data indicate that capsid motility in the somata of neurons mediated by pUL36 and pUL37 does not suffice for targeting capsids to axons, and suggest that capsid envelopment needs to be completed in the soma prior to targeting of herpes simplex virus to the axons, and to spreading from neurons to neighboring cells.


Subject(s)
Herpesvirus 1, Human/physiology , Herpesvirus 1, Human/pathogenicity , Neurons/virology , Animals , Axonal Transport , Axons/ultrastructure , Axons/virology , Capsid/physiology , Capsid/ultrastructure , Cells, Cultured , Chlorocebus aethiops , Ganglia, Spinal/virology , Herpes Simplex/virology , Herpesvirus 1, Human/genetics , Host-Pathogen Interactions , Humans , Mice , Microscopy, Electron, Transmission , Movement/physiology , Mutation , Neurons/ultrastructure , Vero Cells , Viral Proteins/genetics , Viral Proteins/physiology , Viral Structural Proteins/genetics , Viral Structural Proteins/physiology
4.
Eur J Immunol ; 47(10): 1819-1834, 2017 10.
Article in English | MEDLINE | ID: mdl-28771693

ABSTRACT

The MHC class I presentation is responsible for the presentation of viral proteins to CD8+ T lymphocytes and mainly depends on the classical antigen processing pathway. Recently, a second pathway involving autophagy has been implicated in this process. Here, we show an increase in the capacity of murine dendritic cells (DCs) to present viral antigens on MHC class I after infection with a mutant herpes simplex virus 1 (HSV-1-Δ34.5), lacking infected cell protein 34.5 (ICP34.5), when compared to its parental HSV-1 strain. The ICP34.5 protein counteracts host cell translational arrest and suppresses macroautophagy, and the lack of this protein resulted in a low viral protein abundance, which was processed and presented in an efficient way. Our study demonstrates an important role of autophagy in processing endogenous viral proteins in HSV-1-infected DCs.


Subject(s)
Autophagy/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Herpesvirus 1, Human/immunology , Herpesvirus 1, Human/physiology , Lymphocyte Activation , Animals , Antigen Presentation , Antigens, Viral/immunology , CD8-Positive T-Lymphocytes/physiology , Dendritic Cells/metabolism , Dendritic Cells/pathology , Dendritic Cells/virology , Herpesvirus 1, Human/genetics , Histocompatibility Antigens Class I/immunology , Mice , Viral Proteins/genetics , Viral Proteins/metabolism
5.
Sci Rep ; 6: 29903, 2016 07 18.
Article in English | MEDLINE | ID: mdl-27426251

ABSTRACT

Here, we describe a novel reliable method to assess the significance of individual mutations within the thymidine kinase (TK) gene of herpes simplex virus type 1 (HSV-1) to nucleoside analogue resistance. Eleven defined single nucleotide polymorphisms that occur in the TK gene of clinical HSV-1 isolates and a fluorescence reporter were introduced into the HSV-1 strain 17(+) that had been cloned into a bacterial artificial chromosome. The susceptibility of these different strains to aciclovir, penciclovir, brivudin, and foscarnet was determined with a modified cytopathic effect reduction assay. The strains were also tested for their aciclovir susceptibility by measuring the relative fluorescence intensity as an indicator for HSV-1 replication and by quantifying the virus yield. Our data indicate that the amino acid substitutions R41H, R106H, A118V, L139V, K219T, S276R, L298R, S345P, and V348I represent natural polymorphisms of the TK protein, whereas G61A and P84L mediate broad cross-resistance against aciclovir, penciclovir, brivudin, and susceptibility to foscarnet. This method allows the definition of the resistance genotype of otherwise unclear mutations in the TK gene of HSV-1. Thus, it provides a scientific basis for antiviral testing in clinical isolates of patients suffering from serious diseases and will facilitate testing of new antivirals against HSV-1.


Subject(s)
Acyclovir/pharmacology , Drug Resistance, Viral/genetics , Herpesvirus 1, Human/drug effects , Herpesvirus 1, Human/genetics , Mutation/genetics , Recombination, Genetic/genetics , Animals , Antiviral Agents/pharmacology , Chlorocebus aethiops , Drug Resistance, Viral/drug effects , Herpesvirus 1, Human/enzymology , Kinetics , Polymerase Chain Reaction , Thymidine Kinase/genetics , Thymidine Kinase/metabolism , Transfection , Vero Cells , Viral Load/genetics , Virus Replication/drug effects
6.
J Virol ; 90(11): 5368-5383, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27009950

ABSTRACT

UNLABELLED: Herpes simplex virus (HSV) replicates in the skin and mucous membranes, and initiates lytic or latent infections in sensory neurons. Assembly of progeny virions depends on the essential large tegument protein pUL36 of 3,164 amino acid residues that links the capsids to the tegument proteins pUL37 and VP16. Of the 32 tryptophans of HSV-1-pUL36, the tryptophan-acidic motifs (1766)WD(1767) and (1862)WE(1863) are conserved in all HSV-1 and HSV-2 isolates. Here, we characterized the role of these motifs in the HSV life cycle since the rare tryptophans often have unique roles in protein function due to their large hydrophobic surface. The infectivity of the mutants HSV-1(17(+))Lox-pUL36-WD/AA-WE/AA and HSV-1(17(+))Lox-CheVP26-pUL36-WD/AA-WE/AA, in which the capsid has been tagged with the fluorescent protein Cherry, was significantly reduced. Quantitative electron microscopy shows that there were a larger number of cytosolic capsids and fewer enveloped virions compared to their respective parental strains, indicating a severe impairment in secondary capsid envelopment. The capsids of the mutant viruses accumulated in the perinuclear region around the microtubule-organizing center and were not dispersed to the cell periphery but still acquired the inner tegument proteins pUL36 and pUL37. Furthermore, cytoplasmic capsids colocalized with tegument protein VP16 and, to some extent, with tegument protein VP22 but not with the envelope glycoprotein gD. These results indicate that the unique conserved tryptophan-acidic motifs in the central region of pUL36 are required for efficient targeting of progeny capsids to the membranes of secondary capsid envelopment and for efficient virion assembly. IMPORTANCE: Herpesvirus infections give rise to severe animal and human diseases, especially in young, immunocompromised, and elderly individuals. The structural hallmark of herpesvirus virions is the tegument, which contains evolutionarily conserved proteins that are essential for several stages of the herpesvirus life cycle. Here we characterized two conserved tryptophan-acidic motifs in the central region of the large tegument protein pUL36 of herpes simplex virus. When we mutated these motifs, secondary envelopment of cytosolic capsids and the production of infectious particles were severely impaired. Our data suggest that pUL36 and its homologs in other herpesviruses, and in particular such tryptophan-acidic motifs, could provide attractive targets for the development of novel drugs to prevent herpesvirus assembly and spread.


Subject(s)
Capsid/metabolism , Herpesvirus 1, Human/physiology , Tryptophan/chemistry , Viral Structural Proteins/chemistry , Viral Structural Proteins/metabolism , Virus Assembly , Amino Acid Motifs , Capsid/ultrastructure , Capsid Proteins/chemistry , Capsid Proteins/metabolism , Cell Line , Cytoplasm/virology , Herpes Simplex Virus Protein Vmw65/metabolism , Herpesvirus 1, Human/chemistry , Herpesvirus 1, Human/genetics , Humans , Life Cycle Stages , Microscopy, Electron , Mutation , Protein Binding , Protein Domains , Tryptophan/metabolism , Viral Structural Proteins/genetics
7.
Methods Mol Biol ; 1144: 43-62, 2014.
Article in English | MEDLINE | ID: mdl-24671676

ABSTRACT

Bacterial artificial chromosomes (BACs) are suitable vectors not only to maintain the large genomes of herpesviruses in Escherichia coli but also to enable the traceless introduction of any mutation using modern tools of bacterial genetics. To clone a herpes simplex virus genome, a BAC replication origin is first introduced into the viral genome by homologous recombination in eukaryotic host cells. As part of their nuclear replication cycle, genomes of herpesviruses circularize and these replication intermediates are then used to transform bacteria. After cloning, the integrity of the recombinant viral genomes is confirmed by restriction length polymorphism analysis and sequencing. The BACs may then be used to design virus mutants. Upon transfection into eukaryotic cells new herpesvirus strains harboring the desired mutations can be recovered and used for experiments in cultured cells as well as in animal infection models.


Subject(s)
Chromosomes, Artificial, Bacterial/genetics , Genome, Viral , Molecular Biology/methods , Simplexvirus/genetics , Cells, Cultured , Cloning, Molecular , DNA Replication , Escherichia coli/genetics , Eukaryotic Cells , Humans
8.
Cell Microbiol ; 15(2): 248-69, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23186167

ABSTRACT

As the inner tegument proteins pUL36 and pUL37 of alphaherpesviruses may contribute to efficient intracellular transport of viral particles, we investigated their role in cytosolic capsid motility during assembly of herpes simplex virus type 1 (HSV1). As reported previously for pUL36, untagged pUL37 and UL37GFP bound to cytosolic capsids before these acquired outer tegument and envelope proteins. Capsids tagged with CheVP26 analysed by live cell imaging were capable of directed long-distance cytoplasmic transport during the assembly of wild-type virions, while capsids of the HSV1-ΔUL37 or HSV1-ΔUL36 deletion mutants showed only random, undirected motion. The HSV1-ΔUL37 phenotype was restored when UL37GFP had been overexpressed prior to infection. Quantitative immunoelectron microscopy revealed that capsids of HSV1-ΔUL37 still recruited pUL36, whereas pUL37 did not colocalize with capsids of HSV1-ΔUL36. Nevertheless, the cytosolic capsids of neither mutant could undergo secondary envelopment. Our data suggest that pUL36 and pUL37 are important prior to their functions in linking the inner to the outer tegument. Efficient capsid transport to the organelle of secondary envelopment requires recruitment ofpUL37 onto capsids, most likely via its interaction with pUL36, while capsid-associated pUL36 alone is insufficient.


Subject(s)
Capsid/metabolism , Cytosol/virology , Herpesvirus 1, Human/metabolism , Viral Proteins/metabolism , Viral Structural Proteins/metabolism , Virion/metabolism , Animals , Biological Transport, Active , Capsid/chemistry , Capsid/ultrastructure , Cell Line , Chlorocebus aethiops , Cytosol/metabolism , Cytosol/ultrastructure , Gene Expression , Herpesvirus 1, Human/chemistry , Herpesvirus 1, Human/ultrastructure , Microscopy, Immunoelectron , Molecular Imaging , Mutation , Protein Binding , Vero Cells , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Structural Proteins/chemistry , Viral Structural Proteins/genetics , Virion/chemistry , Virion/ultrastructure
9.
J Virol ; 86(7): 3682-700, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22258258

ABSTRACT

The largest tegument protein of herpes simplex virus type 1 (HSV1), pUL36, is a multivalent cross-linker between the viral capsids and the tegument and associated membrane proteins during assembly that upon subsequent cell entry releases the incoming capsids from the outer tegument and viral envelope. Here we show that pUL36 was recruited to cytosolic progeny capsids that later colocalized with membrane proteins of herpes simplex virus type 1 (HSV1) and the trans-Golgi network. During cell entry, pUL36 dissociated from viral membrane proteins but remained associated with cytosolic capsids until arrival at the nucleus. HSV1 UL36 mutants lacking C-terminal portions of increasing size expressed truncated pUL36 but could not form plaques. Cytosolic capsids of mutants lacking the C-terminal 735 of the 3,164 amino acid residues accumulated in the cytosol but did not recruit pUL36 or associate with membranes. In contrast, pUL36 lacking only the 167 C-terminal residues bound to cytosolic capsids and subsequently colocalized with viral and host membrane proteins. Progeny virions fused with neighboring cells, but incoming capsids did not retain pUL36, nor could they target the nucleus or initiate HSV1 gene expression. Our data suggest that residues 2430 to 2893 of HSV1 pUL36, containing one binding site for the capsid protein pUL25, are sufficient to recruit pUL36 onto cytosolic capsids during assembly for secondary envelopment, whereas the 167 residues of the very C terminus with the second pUL25 binding site are crucial to maintain pUL36 on incoming capsids during cell entry. Capsids lacking pUL36 are targeted neither to membranes for virus assembly nor to nuclear pores for genome uncoating.


Subject(s)
Capsid Proteins/metabolism , Herpes Simplex/virology , Herpesvirus 1, Human/physiology , Viral Proteins/chemistry , Viral Proteins/metabolism , Virus Assembly , Virus Internalization , Amino Acid Motifs , Animals , Binding Sites , Capsid Proteins/chemistry , Capsid Proteins/genetics , Cell Line , Herpesvirus 1, Human/chemistry , Herpesvirus 1, Human/genetics , Humans , Protein Binding , Viral Proteins/genetics , trans-Golgi Network/virology
SELECTION OF CITATIONS
SEARCH DETAIL
...