Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Apoptosis ; 16(3): 256-71, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21107701

ABSTRACT

We previously reported that HSV-2 R1, the R1 subunit (ICP10; UL39) of herpes simplex virus type-2 ribonucleotide reductase, protects cells against apoptosis induced by the death receptor (DR) ligands tumor necrosis factor-alpha- (TNFα) and Fas ligand (FasL) by interrupting DR-mediated signaling at, or upstream of, caspase-8 activation. Further investigation of the molecular mechanism underlying HSV-2 R1 protection showed that extracellular-regulated kinase 1/2 (ERK1/2), phosphatidylinositol 3-kinase (PI3-K)/Akt, NF-κB and JNK survival pathways do not play a major role in this antiapoptotic function. Interaction studies revealed that HSV-2 R1 interacted constitutively with caspase-8. The HSV-2 R1 deletion mutant R1(1-834)-GFP and Epstein-Barr virus (EBV) R1, which did not protect against apoptosis induced by DR ligands, did not interact with caspase-8, indicating that interaction is required for protection. HSV-2 R1 impaired caspase-8 activation induced by caspase-8 over-expression, suggesting that interaction between the two proteins prevents caspase-8 dimerization/activation. HSV-2 R1 bound to caspase-8 directly through its prodomain but did not interact with either its caspase domain or Fas-associated death domain protein (FADD). Interaction between HSV-2 R1 and caspase-8 disrupted FADD-caspase-8 binding. We further demonstrated that individually expressed HSV-1 R1 (ICP6) shares, with HSV-2 R1, the ability to bind caspase-8 and to protect cells against DR-induced apoptosis. Finally, as the long-lived Fas protein remained stable during the early period of infection, experiments with the HSV-1 UL39 deletion mutant ICP6∆ showed that HSV-1 R1 could be essential for the protection of HSV-1-infected cells against FasL.


Subject(s)
Apoptosis/drug effects , Caspase 8/metabolism , Fas Ligand Protein/pharmacology , Herpesvirus 1, Human/enzymology , Herpesvirus 2, Human/enzymology , Ribonucleotide Reductases/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Caspase 8/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Cytoprotection/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Fas-Associated Death Domain Protein/metabolism , Gene Deletion , Herpes Simplex/metabolism , Herpes Simplex/virology , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Protein Binding/drug effects , Protein Structure, Tertiary , Protein Subunits/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects
2.
Exp Cell Res ; 314(8): 1652-66, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18367172

ABSTRACT

Expansions of a (GCN)10/polyalanine tract in the Poly(A) Binding Protein Nuclear 1 (PABPN1) cause autosomal dominant oculopharyngeal muscular dystrophy (OPMD). In OPMD muscles, as in models, PABPN1 accumulates in intranuclear inclusions (INIs) whereas in other diseases caused by similar polyalanine expansions, the mutated proteins have been shown to abnormally accumulate in the cytoplasm. This study presents the impact on the subcellular localization of PABPN1 produced by large expansions or deletion of its polyalanine tract. Large tracts of more than 24 alanines result in the nuclear accumulation of PABPN1 in SFRS2-positive functional speckles and a significant decline in cell survival. These large expansions do not cause INIs formation nor do they lead to cytoplasmic accumulation. Deletion of the polyalanine tract induces the formation of aggregates that are located on either side and cross the nuclear membrane, highlighting the possible role of the N-terminal polyalanine tract in PABPN1 nucleo-cytoplasmic transport. We also show that even though five other proteins with polyalanine tracts tend to aggregate when over-expressed they do not co-aggregate with PABPN1 INIs. This study presents the first experimental evidence that there may be a relative loss of function in OPMD by decreasing the availability of PABPN1 through an INI-independent mechanism.


Subject(s)
DNA Repeat Expansion , Peptides/genetics , Poly(A)-Binding Protein II/genetics , Animals , COS Cells , Cell Nucleus Structures/chemistry , Cell Survival , Chlorocebus aethiops , HeLa Cells , Humans , Nuclear Envelope/chemistry , Nuclear Proteins/analysis , Peptides/chemistry , Poly(A)-Binding Protein II/analysis , Poly(A)-Binding Protein II/metabolism , RNA, Messenger/analysis , Ribonucleoproteins/analysis , Sequence Deletion , Serine-Arginine Splicing Factors
3.
J Gen Virol ; 88(Pt 2): 384-394, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17251554

ABSTRACT

The R1 subunit (ICP10) of herpes simplex virus type 2 (HSV-2) ribonucleotide reductase (RR), which in addition to its C-terminal reductase domain possesses a unique N-terminal domain of about 400 aa, protects cells against apoptosis. As the NH(2) domain on its own is not antiapoptotic, it has been postulated that both domains of R1 or part(s) of them could be necessary for this function. Here, N- and C-terminal deletions were introduced in HSV-2 R1 to map the domain(s) involved in its antiapoptotic potential. The results showed that, whereas most of the NH(2) domain including part of the recently described putative alpha-crystallin domain is dispensable for antiapoptotic activity, it is the integrity of the structured RR domain that is required for protection. As the alpha-crystallin domain appears to play an important role in protein folding and oligomerization, the N-terminal boundary of the antiapoptotic domain could not be defined precisely. In addition, this study provided evidence that overexpression of HSV-2 R2 at levels up to 30-fold more than HSV-2 R1 did not decrease protection from tumour necrosis factor alpha, indicating that the R1 surface where R2 binds is not involved in antiapoptotic activity. Importantly, this result suggests that the co-expression of both RR subunits during the lytic cycle should not affect protection from this cytokine.


Subject(s)
Apoptosis , Herpesvirus 2, Human/enzymology , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Ribonucleotide Reductases/chemistry , Ribonucleotide Reductases/metabolism , Cell Line , HeLa Cells , Herpesvirus 2, Human/genetics , Herpesvirus 2, Human/pathogenicity , Humans , Mutation , Protein Serine-Threonine Kinases/genetics , Protein Structure, Tertiary , Ribonucleotide Reductases/genetics , Transfection , Tumor Necrosis Factor-alpha/metabolism
4.
J Immunol ; 174(9): 5740-9, 2005 May 01.
Article in English | MEDLINE | ID: mdl-15843576

ABSTRACT

Apoptosis of endothelial cells (EC) is appreciated as a primary pathogenic event in systemic sclerosis. Yet, how apoptosis of EC leads to fibrosis remains to be determined. We report that apoptosis of EC triggers the release of novel fibrogenic mediators. Medium conditioned by apoptotic EC (SSC) was found to inhibit apoptosis of fibroblasts, whereas medium conditioned by EC in which apoptosis was blocked (with either pan-caspase inhibition or Bcl-x(L) overexpression) did not. PI3K was activated in fibroblasts exposed to SSC. This was associated with downstream repression of Bim-EL and long-term up-regulation of Bcl-x(L) protein levels. RNA interference for Bim-EL in fibroblasts blocked apoptosis. SSC also induced PI3K-dependent myofibroblast differentiation with expression of alpha-smooth muscle actin, formation of stress fibers, and production of collagen I. A C-terminal fragment of the domain V of perlecan was identified as one of the fibrogenic mediators present in SSC. A synthetic peptide containing an EGF motif present on the perlecan fragment and chondroitin 4-sulfate, a glycosaminoglycan anchored on the domain V of perlecan, induced PI3K-dependent resistance to apoptosis in fibroblasts and myofibroblast differentiation. Human fibroblasts derived from sclerodermic skin lesions were more sensitive to the antiapoptotic activities of the synthetic peptide and chondroitin 4-sulfate than fibroblasts derived from normal controls. Hence, we propose that a chronic increase in endothelial apoptosis and/or increased sensitivity of fibroblasts to mediators produced by apoptotic EC could form the basis of a fibrotic response characterized by sustained induction of an antiapoptotic phenotype in fibroblasts and persistent myofibroblast differentiation.


Subject(s)
Apoptosis/immunology , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Fibroblasts/metabolism , Fibroblasts/pathology , Scleroderma, Systemic/pathology , Scleroderma, Systemic/physiopathology , Signal Transduction/immunology , Adult , Amino Acid Sequence , Cell Differentiation/immunology , Cell Line , Chondroitin Sulfates/pharmacology , Culture Media, Conditioned , Endothelium, Vascular/enzymology , Endothelium, Vascular/immunology , Fibroblasts/immunology , Heparan Sulfate Proteoglycans/physiology , Humans , Immunity, Innate , Inflammation Mediators/metabolism , Inflammation Mediators/physiology , Molecular Sequence Data , Peptide Fragments/chemical synthesis , Peptide Fragments/pharmacology , Phosphatidylinositol 3-Kinases/physiology , Protein Structure, Tertiary , Scleroderma, Systemic/immunology
5.
Neurobiol Dis ; 18(3): 551-67, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15755682

ABSTRACT

Oculopharyngeal muscular dystrophy (OPMD) is an adult-onset disease caused by expanded (GCN)12-17 stretches encoding the N-terminal polyalanine domain of the poly(A) binding protein nuclear 1 (PABPN1). OPMD is characterized by intranuclear inclusions (INIs) in skeletal muscle fibers, which contain PABPN1, molecular chaperones, ubiquitin, proteasome subunits, and poly(A)-mRNA. We describe an adenoviral model of PABPN1 expression that produces INIs in most cells. Microarray analysis revealed that PABPN1 overexpression reproducibly changed the expression of 202 genes. Sixty percent of upregulated genes encode nuclear proteins, including many RNA and DNA binding proteins. Immunofluorescence microscopy revealed that all tested nuclear proteins encoded by eight upregulated genes colocalize with PABPN1 within the INIs: CUGBP1, SFRS3, FKBP1A, HMG2, HNRPA1, PRC1, S100P, and HSP70. In addition, CUGBP1, SFRS3, and FKBP1A were also found in OPMD muscle INIs. This study demonstrates that a large number of nuclear proteins are sequestered in OPMD INIs, which may compromise cellular function.


Subject(s)
Intranuclear Inclusion Bodies/metabolism , Muscular Dystrophy, Oculopharyngeal/genetics , Muscular Dystrophy, Oculopharyngeal/metabolism , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , Poly(A)-Binding Protein I/biosynthesis , Poly(A)-Binding Protein I/genetics , Up-Regulation/physiology , Animals , Cattle , Cell Line , Gene Expression Regulation/physiology , Humans , Intranuclear Inclusion Bodies/genetics
6.
FEBS Lett ; 545(2-3): 213-8, 2003 Jun 19.
Article in English | MEDLINE | ID: mdl-12804778

ABSTRACT

HSV-2 R1, the R1 subunit of herpes simplex virus (HSV) ribonucleotide reductase, protects cells against apoptosis. Here, we report the presence in HSV-2 R1 of a stretch exhibiting similarity to the alpha-crystallin domain of the small heat shock proteins, a domain known to be important for oligomerization and cytoprotective activities of these proteins. Also, the HSV-2 R1 protein, which forms multimeric structures in the absence of nucleotide, displayed chaperone ability as good as Hsp27 in a thermal denaturation assay using citrate synthase. In contrast, mammalian R1, which does not contain an alpha-crystallin domain, has neither chaperone nor anti-apoptotic activity. Thus, we propose that the chaperone activity of HSV-2 R1 could play an important role in viral pathogenesis.


Subject(s)
Heat-Shock Proteins/metabolism , Herpesvirus 1, Human/enzymology , Ribonucleotide Reductases/chemistry , Ribonucleotide Reductases/metabolism , Amino Acid Sequence , Apoptosis , Citrate (si)-Synthase/metabolism , Consensus Sequence , HeLa Cells , Herpesvirus 1, Human/genetics , Hot Temperature , Humans , Kinetics , Molecular Sequence Data , Molecular Weight , Mutation , Protein Denaturation , Protein Structure, Secondary , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Tumor Cells, Cultured
7.
J Gen Virol ; 83(Pt 11): 2779-2789, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12388814

ABSTRACT

The R1 subunit of herpes simplex virus (HSV) ribonucleotide reductase, which in addition to its C-terminal reductase domain possesses a unique N-terminal domain of about 400 amino acids, is thought to have an additional, as yet unknown, function. Here, we report that the full-length HSV-2 R1 has an anti-apoptotic function able to protect cells against death triggered by expression of R1(Delta2-357), an HSV-2 R1 subunit with its first 357 amino acids deleted. We further substantiate the R1 anti-apoptotic activity by showing that its accumulation at low level could completely block apoptosis induced by TNF-receptor family triggering. Activation of caspase-8 induced either by TNF or by Fas ligand expression was prevented by the R1 protein. As HSV R1 did not inhibit cell death mediated by several agents acting via the mitochondrial pathway (Bax overexpression, etoposide, staurosporine and menadione), it is proposed that it functions to interrupt specifically death receptor-mediated signalling at, or upstream of, caspase-8 activation. The N-terminal domain on its own did not exhibit anti-apoptotic activity, suggesting that both domains of R1 or part(s) of them are necessary for this new function. Evidence for the importance of HSV R1 in protecting HSV-infected cells against cytokine-induced apoptosis was obtained with the HSV-1 R1 deletion mutants ICP6Delta and hrR3. These results show that, in addition to its ribonucleotide reductase function, which is essential for virus reactivation, HSV R1 could contribute to virus propagation by preventing apoptosis induced by the immune system.


Subject(s)
Apoptosis , Caspases/metabolism , Herpesvirus 2, Human/enzymology , Protein Serine-Threonine Kinases/metabolism , Ribonucleotide Reductases/metabolism , Adenovirus E1A Proteins/metabolism , Animals , Caspase 8 , Caspase 9 , Cell Line , Cricetinae , Enzyme Activation , Fas Ligand Protein , HeLa Cells , Humans , Membrane Glycoproteins/metabolism , Tumor Necrosis Factor-alpha/pharmacology
8.
J Gen Virol ; 83(Pt 10): 2411-2416, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12237422

ABSTRACT

A cytopathogenic coronavirus, serologically identified as porcine haemagglutinating encephalomyelitis virus (HEV), has recently been associated with acute outbreaks of wasting and encephalitis in nursing piglets from pig farms in southern Québec and Ontario, Canada. The 3'-terminal end of the genome of the prototype HEV-67N strain and that of the recent Québec IAF-404 field isolate, both propagated in HRT-18 cells, were sequenced. Overall, sequencing data indicated that HEV has remained antigenically and genetically stable since its first isolation in North America in 1962. Compared with the prototype strain of bovine enteropathogenic coronavirus (BCoV), HEV, as well as the human respiratory coronavirus (HCoV-OC43) showed a major deletion in their ORF4 gene. Deduced amino acid sequences for both HEV strains revealed 89/88, 80, 93/92 and 95/94% identities with the structural proteins HE, S, M and N of BCoV and HCoV-OC43, respectively. Major variations were observed in the S1 portion of the S gene of both HEV strains, with only 73/71% amino acid identities compared with those of the two other haemagglutinating coronaviruses.


Subject(s)
Coronavirus/genetics , Genome, Viral , Amino Acid Sequence , Animals , Cattle , Coronavirus/isolation & purification , Coronavirus Infections/virology , Humans , Molecular Sequence Data , Open Reading Frames , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Swine , Viral Nonstructural Proteins/genetics , Viral Structural Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...