Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Apoptosis ; 16(3): 256-71, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21107701

RESUMO

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.


Assuntos
Apoptose/efeitos dos fármacos , Caspase 8/metabolismo , Proteína Ligante Fas/farmacologia , Herpesvirus Humano 1/enzimologia , Herpesvirus Humano 2/enzimologia , Ribonucleotídeo Redutases/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Caspase 8/química , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Citoproteção/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteína de Domínio de Morte Associada a Fas/metabolismo , Deleção de Genes , Herpes Simples/metabolismo , Herpes Simples/virologia , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Ligação Proteica/efeitos dos fármacos , Estrutura Terciária de Proteína , Subunidades Proteicas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos
2.
Exp Cell Res ; 314(8): 1652-66, 2008 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-18367172

RESUMO

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.


Assuntos
Expansão das Repetições de DNA , Peptídeos/genética , Proteína II de Ligação a Poli(A)/genética , Animais , Células COS , Estruturas do Núcleo Celular/química , Sobrevivência Celular , Chlorocebus aethiops , Células HeLa , Humanos , Membrana Nuclear/química , Proteínas Nucleares/análise , Peptídeos/química , Proteína II de Ligação a Poli(A)/análise , Proteína II de Ligação a Poli(A)/metabolismo , RNA Mensageiro/análise , Ribonucleoproteínas/análise , Deleção de Sequência , Fatores de Processamento de Serina-Arginina
3.
J Gen Virol ; 88(Pt 2): 384-394, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17251554

RESUMO

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.


Assuntos
Apoptose , Herpesvirus Humano 2/enzimologia , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/metabolismo , Ribonucleotídeo Redutases/química , Ribonucleotídeo Redutases/metabolismo , Linhagem Celular , Células HeLa , Herpesvirus Humano 2/genética , Herpesvirus Humano 2/patogenicidade , Humanos , Mutação , Proteínas Serina-Treonina Quinases/genética , Estrutura Terciária de Proteína , Ribonucleotídeo Redutases/genética , Transfecção , Fator de Necrose Tumoral alfa/metabolismo
4.
J Immunol ; 174(9): 5740-9, 2005 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-15843576

RESUMO

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.


Assuntos
Apoptose/imunologia , Endotélio Vascular/patologia , Endotélio Vascular/fisiopatologia , Fibroblastos/metabolismo , Fibroblastos/patologia , Escleroderma Sistêmico/patologia , Escleroderma Sistêmico/fisiopatologia , Transdução de Sinais/imunologia , Adulto , Sequência de Aminoácidos , Diferenciação Celular/imunologia , Linhagem Celular , Sulfatos de Condroitina/farmacologia , Meios de Cultivo Condicionados , Endotélio Vascular/enzimologia , Endotélio Vascular/imunologia , Fibroblastos/imunologia , Proteoglicanas de Heparan Sulfato/fisiologia , Humanos , Imunidade Inata , Mediadores da Inflamação/metabolismo , Mediadores da Inflamação/fisiologia , Dados de Sequência Molecular , Fragmentos de Peptídeos/síntese química , Fragmentos de Peptídeos/farmacologia , Fosfatidilinositol 3-Quinases/fisiologia , Estrutura Terciária de Proteína , Escleroderma Sistêmico/imunologia
5.
Neurobiol Dis ; 18(3): 551-67, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15755682

RESUMO

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.


Assuntos
Corpos de Inclusão Intranuclear/metabolismo , Distrofia Muscular Oculofaríngea/genética , Distrofia Muscular Oculofaríngea/metabolismo , Proteínas Nucleares/biossíntese , Proteínas Nucleares/genética , Proteína I de Ligação a Poli(A)/biossíntese , Proteína I de Ligação a Poli(A)/genética , Regulação para Cima/fisiologia , Animais , Bovinos , Linhagem Celular , Regulação da Expressão Gênica/fisiologia , Humanos , Corpos de Inclusão Intranuclear/genética
6.
FEBS Lett ; 545(2-3): 213-8, 2003 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-12804778

RESUMO

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.


Assuntos
Proteínas de Choque Térmico/metabolismo , Herpesvirus Humano 1/enzimologia , Ribonucleotídeo Redutases/química , Ribonucleotídeo Redutases/metabolismo , Sequência de Aminoácidos , Apoptose , Citrato (si)-Sintase/metabolismo , Sequência Consenso , Células HeLa , Herpesvirus Humano 1/genética , Temperatura Alta , Humanos , Cinética , Dados de Sequência Molecular , Peso Molecular , Mutação , Desnaturação Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Homologia de Sequência de Aminoácidos , Células Tumorais Cultivadas
7.
J Gen Virol ; 83(Pt 11): 2779-2789, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12388814

RESUMO

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.


Assuntos
Apoptose , Caspases/metabolismo , Herpesvirus Humano 2/enzimologia , Proteínas Serina-Treonina Quinases/metabolismo , Ribonucleotídeo Redutases/metabolismo , Proteínas E1A de Adenovirus/metabolismo , Animais , Caspase 8 , Caspase 9 , Linhagem Celular , Cricetinae , Ativação Enzimática , Proteína Ligante Fas , Células HeLa , Humanos , Glicoproteínas de Membrana/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
8.
J Gen Virol ; 83(Pt 10): 2411-2416, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12237422

RESUMO

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.


Assuntos
Coronavirus/genética , Genoma Viral , Sequência de Aminoácidos , Animais , Bovinos , Coronavirus/isolamento & purificação , Infecções por Coronavirus/virologia , Humanos , Dados de Sequência Molecular , Fases de Leitura Aberta , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos , Suínos , Proteínas não Estruturais Virais/genética , Proteínas Estruturais Virais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...