Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Retrovirology ; 12: 57, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26129669

ABSTRACT

BACKGROUND: Matrin 3 is a nuclear matrix protein involved in multiple nuclear processes. In HIV-1 infection, Matrin 3 serves as a Rev cofactor important for the cytoplasmic accumulation of HIV-1 transcripts. ZAP is a potent host restriction factor of multiple viruses including retroviruses HIV-1 and MoMuLV. In this study we sought to further characterize Matrin 3 functions in the regulation of HIV gene expression. RESULTS: Here we describe a function for Matrin 3 as a negative regulator of the ZAP-mediated restriction of retroviruses. Mass spectrometry analysis of Matrin 3-associated proteins uncovered interactions with proteins of the ZAP degradation complex, DDX17 and EXOSC3. Coimmunoprecipitation studies confirmed Matrin 3 associations with DDX17, EXOSC3 and ZAP, in a largely RNA-dependent manner, indicating that RNA is mediating the Matrin 3 interactions with these components of the ZAP degradation complex. Silencing Matrin 3 expression caused a remarkably enhanced ZAP-driven inhibition of HIV-1 and MoMuLV luciferase reporter viruses. This effect was shared with additional nuclear matrix proteins. ZAP targets multiply-spliced HIV-1 transcripts, but in the context of Matrin 3 suppression, this ZAP restriction was broadened to unspliced and multiply-spliced RNAs. CONCLUSIONS: Here we reveal an unprecedented role for a nuclear matrix protein, Matrin 3, in the regulation of ZAP's antiretroviral activity. Suppressing Matrin 3 powers a heightened and broader ZAP restriction of HIV-1 gene expression. This study suggests that this ZAP regulatory mechanism is shared with additional nuclear matrix proteins.


Subject(s)
Gene Expression Regulation, Viral , HIV-1/immunology , HIV-1/physiology , Host-Pathogen Interactions , Immunity, Innate , Nuclear Matrix-Associated Proteins/metabolism , RNA-Binding Proteins/metabolism , Cell Line , Humans , Virus Replication
2.
J Virol ; 85(1): 568-81, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20962082

ABSTRACT

The protein kinase found in the short region of alphaherpesviruses, termed US3 in herpes simplex virus type 1 (HSV-1) and pseudorabies virus (PRV) and ORF66 in varicella-zoster virus (VZV), affects several viral and host cell processes, and its specific targets remain an area of active investigation. Reports suggesting that HSV-1 US3 substrates overlap with those of cellular protein kinase A (PKA) prompted the use of an antibody specific for phosphorylated PKA substrates to identify US3/ORF66 targets. HSV-1, VZV, and PRV induced very different substrate profiles that were US3/ORF66 kinase dependent. The predominant VZV-phosphorylated 125-kDa species was identified as matrin 3, one of the major nuclear matrix proteins. Matrin 3 was also phosphorylated by HSV-1 and PRV in a US3 kinase-dependent manner and by VZV ORF66 kinase at a novel residue (KRRRT150EE). Since VZV-directed T150 phosphorylation was not blocked by PKA inhibitors and was not induced by PKA activation, and since PKA predominantly targeted matrin 3 S188, it was concluded that phosphorylation by VZV was PKA independent. However, purified VZV ORF66 kinase did not phosphorylate matrin 3 in vitro, suggesting that additional cellular factors were required. In VZV-infected cells in the absence of the ORF66 kinase, matrin 3 displayed intranuclear changes, while matrin 3 showed a pronounced cytoplasmic distribution in late-stage cells infected with US3-negative HSV-1 or PRV. This work identifies phosphorylation of the nuclear matrix protein matrin 3 as a new conserved target of this kinase group.


Subject(s)
Alphaherpesvirinae/enzymology , Gene Expression Regulation , Nuclear Matrix-Associated Proteins/metabolism , Open Reading Frames/physiology , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , RNA-Binding Proteins/metabolism , Viral Proteins/metabolism , Alphaherpesvirinae/classification , Alphaherpesvirinae/genetics , Alphaherpesvirinae/metabolism , Cell Line , Fibroblasts/virology , Herpesvirus 1, Human/enzymology , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/metabolism , Herpesvirus 1, Suid/enzymology , Herpesvirus 1, Suid/genetics , Herpesvirus 1, Suid/metabolism , Herpesvirus 3, Human/enzymology , Herpesvirus 3, Human/genetics , Herpesvirus 3, Human/metabolism , Humans , Kidney/cytology , Kidney/virology , Nuclear Matrix-Associated Proteins/genetics , Phosphorylation , Protein Kinases/genetics , RNA-Binding Proteins/genetics
3.
Curr Top Microbiol Immunol ; 342: 79-98, 2010.
Article in English | MEDLINE | ID: mdl-20186610

ABSTRACT

The varicella-zoster virus (VZV) open reading frame (ORF) 66 encodes a basophilic kinase orthologous to the US3 protein kinases found in all alphaherpesviruses. This review summarizes current information on the ORF66 kinase, and outlines apparent differences from other US3 kinases, as well as some of the conserved functions. One critical difference is the VZV ORF66 kinase targeting of the major regulatory VZV IE62 protein to control its nuclear import and assembly into the VZV virion, which is so far unprecedented in the alphaherpesviruses. However, ORF66 targets some cellular targets which are also targeted by US3 kinases of other herpesviruses, including the histone deacetylase-1 and 2 proteins, pathways that lead to changes in actin dynamics, and the targeting of substrates of protein kinase A, including the nuclear matrix protein matrin 3.


Subject(s)
Herpesvirus 3, Human/enzymology , Immediate-Early Proteins/physiology , Protein Kinases/physiology , Trans-Activators/physiology , Viral Envelope Proteins/physiology , Amino Acid Sequence , Herpesvirus 3, Human/genetics , Humans , Molecular Sequence Data , Open Reading Frames , Protein Kinases/genetics , Protein Kinases/metabolism
4.
J Virol ; 83(22): 11502-13, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19740981

ABSTRACT

ORF66p, a virion-associated varicella-zoster virus (VZV) protein, is a member of a conserved Alphaherpesvirinae kinase family with homology to herpes simplex virus US3 kinase. Expression of ORF66p in cells infected with VZV or an adenovirus expressing only ORF66p results in hyperphosphorylation of histone deacetylase 1 (HDAC1) and HDAC2. Mapping studies reveal that phosphorylation is at a unique conserved Ser residue in the C terminus of both HDACs. This modification requires an active kinase domain in ORF66p, as neither protein is phosphorylated in cells infected with VZV lacking kinase activity. However, hyperphosphorylation appears to occur indirectly, as within the context of in vitro kinase reactions, purified ORF66p phosphorylates a peptide derived from ORF62p, a known substrate, but does not phosphorylate HDAC. These results support a model where ORF66p is necessary but not sufficient to effect hyperphosphorylation of HDAC1 and HDAC2.


Subject(s)
Herpes Zoster/enzymology , Herpesvirus 3, Human/metabolism , Histone Deacetylase 1/metabolism , Histone Deacetylase 2/metabolism , Cell Line , Cell Line, Tumor , Genetic Vectors , Herpesvirus 3, Human/enzymology , Humans , Phosphorylation , Protein Kinases/metabolism , Viral Proteins/physiology
5.
J Virol ; 83(1): 47-57, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18945788

ABSTRACT

The herpes simplex virus protein UL25 attaches to the external vertices of herpes simplex virus type 1 capsids and is required for the stable packaging of viral DNA. To define regions of the protein important for viral replication and capsid attachment, the 580-amino-acid UL25 open reading frame was disrupted by transposon mutagenesis. The UL25 mutants were assayed for complementation of a UL25 deletion virus, and in vitro-synthesized protein was tested for binding to UL25-deficient capsids. Of the 11 mutants analyzed, 4 did not complement growth of the UL25 deletion mutant, and analysis of these and additional mutants in the capsid-binding assay demonstrated that UL25 amino acids 1 to 50 were sufficient for capsid binding. Several UL25 mutations were transferred into recombinant viruses to analyze the effect of the mutations on UL25 capsid binding and on DNA cleavage and packaging. Studies of these mutants demonstrated that amino acids 1 to 50 of UL25 are essential for its stable interaction with capsids and that the C terminus is essential for DNA packaging and the production of infectious virus through its interactions with other viral packaging or tegument proteins. Analysis of viral DNA cleavage demonstrated that in the absence of a functional UL25 protein, aberrant cleavage takes place at the unique short end of the viral genome, resulting in truncated viral genomes that are not retained in capsids. Based on these observations, we propose a model where UL25 is required for the formation of DNA-containing capsids by acting to stabilize capsids that contain full-length viral genomes.


Subject(s)
Herpesvirus 1, Human/physiology , Viral Proteins/metabolism , Virus Assembly , Animals , Binding Sites , Chlorocebus aethiops , DNA Transposable Elements , DNA, Viral/metabolism , Gene Deletion , Genetic Complementation Test , Mutagenesis, Insertional , Mutant Proteins/genetics , Mutant Proteins/metabolism , Protein Interaction Mapping , Vero Cells , Viral Proteins/genetics
6.
J Virol ; 82(15): 7653-65, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18495764

ABSTRACT

Varicella-zoster virus (VZV) open reading frame 66 (ORF66) encodes a serine/threonine protein kinase that is not required for VZV growth in most cell types but is needed for efficient growth in T cells. The ORF66 kinase affects nuclear import and virion packaging of IE62, the major regulatory protein, and is known to regulate apoptosis in T cells. Here, we further examined the importance of ORF66 using VZV recombinants expressing green fluorescent protein (GFP)-tagged functional and kinase-negative ORF66 proteins. VZV virions with truncated or kinase-inactivated ORF66 protein were marginally reduced for growth and progeny yields in MRC-5 fibroblasts but were severely growth and replication impaired in low-passage primary human corneal stromal fibroblasts (PCF). To determine if the growth impairment was due to ORF66 kinase regulation of IE62 nuclear import, recombinant VZVs that expressed IE62 with alanine residues at S686, the suspected target by which ORF66 kinase blocks IE62 nuclear import, were made. IE62 S686A expressed by the VZV recombinant remained nuclear throughout infection and was not packaged into virions. However, the mutant virus still replicated efficiently in PCF cells. We also show that inactivation of the ORF66 kinase resulted in only marginally increased levels of apoptosis in PCF cells, which could not fully account for the cell-specific growth requirement of ORF66 kinase. Thus, the unique short region VZV kinase has important cell-type-specific functions that are separate from those affecting IE62 and apoptosis.


Subject(s)
Fibroblasts/virology , Herpesvirus 3, Human/enzymology , Herpesvirus 3, Human/physiology , Protein Serine-Threonine Kinases/metabolism , Viral Nonstructural Proteins/metabolism , Virus Replication , Amino Acid Substitution/genetics , Cell Nucleus/chemistry , Cells, Cultured , Genes, Reporter , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Herpesvirus 3, Human/growth & development , Humans , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Mutagenesis, Site-Directed , Protein Serine-Threonine Kinases/genetics , Sequence Deletion , Staining and Labeling , Trans-Activators/genetics , Trans-Activators/metabolism , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Viral Nonstructural Proteins/genetics , Virion/chemistry
7.
J Virol ; 81(17): 9034-49, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17567702

ABSTRACT

We show here that the varicella-zoster virus (VZV) open reading frame 66 (ORF66) protein kinase is one mechanism employed to reduce class I major histocompatibility complex (MHC-I) surface expression in VZV-infected cells. Cells expressing enhanced green fluorescent protein-tagged functional and inactivated ORF66 (GFP-66 and GFP-66kd) from replication-defective adenovirus vectors revealed that ORF66 reduced MHC-I surface levels in a manner dependent on kinase activity. Cells infected with recombinant VZV expressing GFP-66 exhibited a significantly greater reduction in MHC-I surface expression than that observed in cells infected with VZV disrupted in GFP-66 expression. MHC-I maturation was delayed in its transport from the endoplasmic reticulum through the Golgi in both adenovirus-transduced cells expressing only GFP-66 and in VZV-infected cells expressing high levels of GFP-66, and this was predominantly kinase dependent. MHC-I levels were reduced in VZV-infected cells, and analyses of intracellular MHC-I revealed accumulation of folded MHC-I in the Golgi region, irrespective of ORF66 expression. Thus, the ORF66 kinase is important for VZV-mediated MHC-I downregulation, but additional mechanisms also may be involved. Analyses of the VZV ORF9a protein, the ortholog of the bovine herpesvirus 1 transporter associated with antigen processing inhibitor UL49.5 revealed no effects on MHC-I. These results establish a new role for viral protein kinases in immune evasion and suggest that VZV utilizes unique mechanisms to inhibit antigen presentation.


Subject(s)
Down-Regulation , Herpesvirus 3, Human/immunology , Histocompatibility Antigens Class I/biosynthesis , Protein Kinases/physiology , Viral Proteins/physiology , Cell Line , Golgi Apparatus/chemistry , Humans , Protein Kinases/genetics , Protein Processing, Post-Translational , Viral Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...