Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Mol Pharmacol ; 83(1): 245-55, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23093495

RESUMO

Bortezomib is a highly selective inhibitor of the 26S proteasome and has been approved for clinical use in the treatment of relapsing and refractory multiple myeloma and mantle cell lymphoma. Clinical trials are also underway to assess the role of bortezomib in several other human malignancies, including leukemia. However, the mechanism(s) by which bortezomib acts remain to be fully understood. Here, we studied the molecular requirements of bortezomib-induced apoptosis using the human T-cell leukemic Jurkat cells stably transfected with or without shRNA against apoptotic protease-activating factor-1 (Apaf-1). The Apaf-1-deficient Jurkat T cells were resistant to bortezomib-induced apoptosis, as assessed by caspase-3 activity, poly(ADP-ribose) polymerase cleavage, phosphatidylserine externalization, and hypodiploid DNA content. In contrast, Apaf-1-deficient cells were sensitive to Fas-induced apoptosis. Bortezomib induced an upregulation of the pro-apoptotic protein Noxa, loss of mitochondrial transmembrane potential, and release of cytochrome c in cells expressing or not expressing Apaf-1. Transient silencing of Apaf-1 expression in RPMI 8402 T-cell leukemic cells also diminished bortezomib-induced apoptosis. Fas-associated death domain (FADD)-deficient Jurkat cells were resistant to Fas-mediated apoptosis yet remained sensitive to bortezomib. Our results show that bortezomib induces apoptosis by regulating pathways that are mechanistically different from those activated upon death receptor ligation. Furthermore, in silico analyses of public transcriptomics databases indicated elevated Apaf-1 expression in several hematologic malignancies, including acute lymphoblastic and myeloid leukemia. We also noted variable Apaf-1 expression in a panel of samples from patients with acute lymphoblastic leukemia. Our results suggest that the expression of Apaf-1 may be predictive of the response to proteasome inhibition.


Assuntos
Antineoplásicos/farmacologia , Apoptose , Fator Apoptótico 1 Ativador de Proteases/metabolismo , Ácidos Borônicos/farmacologia , Pirazinas/farmacologia , Receptor fas/fisiologia , Adolescente , Fator Apoptótico 1 Ativador de Proteases/genética , Bortezomib , Caspase 3/metabolismo , Criança , Pré-Escolar , Citocromos c/metabolismo , Proteína de Domínio de Morte Associada a Fas/genética , Proteína de Domínio de Morte Associada a Fas/fisiologia , Inativação Gênica , Humanos , Células Jurkat , Leucemia Mieloide/metabolismo , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/patologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , RNA Interferente Pequeno/genética , Transcriptoma
2.
Mol Biol Cell ; 16(8): 3821-31, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15917298

RESUMO

Here, we identified caspase-2, protein kinase C (PKC)delta, and c-Jun NH2-terminal kinase (JNK) as key components of the doxorubicin-induced apoptotic cascade. Using cells stably transfected with an antisense construct for caspase-2 (AS2) as well as a chemical caspase-2 inhibitor, we demonstrate that caspase-2 is required in doxorubicin-induced apoptosis. We also identified PKCdelta as a novel caspase-2 substrate. PKCdelta was cleaved/activated in a caspase-2-dependent manner after doxorubicin treatment both in cells and in vitro. PKCdelta is furthermore required for efficient doxorubicin-induced apoptosis because its chemical inhibition as well as adenoviral expression of a kinase dead (KD) mutant of PKCdelta severely attenuated doxorubicin-induced apoptosis. Furthermore, PKCdelta and JNK inhibition show that PKCdelta lies upstream of JNK in doxorubicin-induced death. Jnk-deficient mouse embryo fibroblasts (MEFs) were highly resistant to doxorubicin compared with wild type (WT), as were WT Jurkat cells treated with SP600125, further supporting the importance of JNK in doxorubicin-induced apoptosis. Chemical inhibitors for PKCdelta and JNK do not synergize and do not function in doxorubicin-treated AS2 cells. Caspase-2, PKCdelta, and JNK were furthermore implicated in doxorubicin-induced apoptosis of primary acute lymphoblastic leukemia blasts. The data thus support a sequential model involving caspase-2, PKCdelta, and JNK signaling in response to doxorubicin, leading to the activation of Bak and execution of apoptosis.


Assuntos
Apoptose/efeitos dos fármacos , Caspases/metabolismo , Doxorrubicina/farmacologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Caspase 2 , Inibidores de Caspase , Linhagem Celular , Citocromos c/metabolismo , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Mutação/genética
3.
Haematologica ; 92(11): 1460-9, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18024393

RESUMO

BACKGROUND AND OBJECTIVES: The mechanism of glucocorticoid -induced apoptosis is not fully understood and early predictive assays based on apoptotic markers for clinical outcome in acute lymphoblastic leukemia (ALL) are scarce. The aim of this study was to characterize the involvement of Bcl-2 family members and caspase activation in dexamethasone(Dex)-induced apoptosis in ALL. DESIGN AND METHODS: Primary childhood ALL samples, the pre-B ALL cell line RS(4;11), and the T-ALL cell line CCRF-CEM were used. The involvement of Bcl-2 family members was evaluated by flow cytometry, immunocytochemistry, and western and northern blotting. Apoptosis was analyzed by annexin V and TMRE staining. Caspase activity was evaluated by a fluorometric assay. RESULTS: Dex induced significant down-regulation of the anti-apoptotic Bcl-2 family members Bcl-2 and Bcl-xL, differential activation of the pro-apoptotic Bak and Bax, loss of Delta psi m and cytochrome c release. Dex-induced apoptosis also involved early activation of caspases 2 and -3. Inhibition of caspase activity did not, however, protect against Dex-induced Bak/Bax activation, loss of Delta psi m or cell death. In 12 primary ALL samples Dex-induced apoptosis was associated with activation of Bax (p=0.045) and down-regulation of Bcl-2 (p=0.016) and/or Bcl-xL (p=0.004). Furthermore, ex vivo Dex-sensitivity was associated with an early treatment response to polychemotherapy (p=0.026). INTERPRETATION AND CONCLUSIONS: The differential regulation of pro- and anti-apoptotic Bcl-2 family members appears to be a key event in the execution of Dex-induced apoptosis in ALL cell lines, and also indicates a role for these proteins in primary ALL cells.


Assuntos
Apoptose/efeitos dos fármacos , Dexametasona/farmacologia , Regulação Neoplásica da Expressão Gênica , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Reguladoras de Apoptose/genética , Caspases/metabolismo , Linhagem Celular Tumoral , Dexametasona/uso terapêutico , Citometria de Fluxo , Humanos , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/patologia , Células Tumorais Cultivadas
4.
Oncogene ; 22(29): 4543-56, 2003 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-12881711

RESUMO

We have recently reported that the cytokine interferon-alpha (IFNalpha), commonly used in the treatment of cancer, induced a caspase-dependent apoptosis in tumor cell lines. The signaling mechanisms involved have not been defined. Here, we show that both proapoptotic Bcl-2 family members Bak and Bax were activated by IFNalpha, strictly in correlation with the induction of apoptosis. Using double stainings, we demonstrated that Bak was activated prior to cytochrome c (cyt c) release and caspase-3 activation, whereas activated Bax was only found in cells with released cyt c, mitochondrial depolarization, as well as activated caspase-3. Furthermore, IFNalpha-induced activation of Bak, and to a large extent also of Bax, was dependent on caspase activity. With the use of a panel of specific caspase inhibitors we found, however, that none of caspases-1 to -10 were responsible for this activation. Neither was the Ca(2+)-dependent protease calpain nor the stress-activated p38 SAPK pathway significantly involved. Overexpression of Bcl-2 blocked apoptosis induced by IFNalpha totally abolished Bak activation, as well as decreased the amount of activated Bax. We conclude that IFNalpha induces Bak and Bax activation via distinct mechanisms involving an unknown protease, and that their activation is regulated by Bcl-2.


Assuntos
Apoptose/efeitos dos fármacos , Linfoma de Burkitt/metabolismo , Interferon-alfa/farmacologia , Proteínas de Membrana/metabolismo , Mieloma Múltiplo/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Apoptose/fisiologia , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3 , Linfoma de Burkitt/tratamento farmacológico , Linfoma de Burkitt/patologia , Calpaína/antagonistas & inibidores , Calpaína/metabolismo , Proteínas de Transporte/efeitos dos fármacos , Proteínas de Transporte/metabolismo , Inibidores de Caspase , Caspases/metabolismo , Inibidores de Cisteína Proteinase/farmacologia , Grupo dos Citocromos c/efeitos dos fármacos , Grupo dos Citocromos c/metabolismo , Inibidores Enzimáticos/farmacologia , Humanos , Potenciais da Membrana/efeitos dos fármacos , Proteínas de Membrana/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/patologia , Proteínas Proto-Oncogênicas/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/genética , Células Tumorais Cultivadas , Proteína Killer-Antagonista Homóloga a bcl-2 , Proteína X Associada a bcl-2 , Proteínas Quinases p38 Ativadas por Mitógeno
5.
Oncogene ; 21(8): 1251-62, 2002 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-11850845

RESUMO

Interferon alpha (IFNalpha) has been used in the treatment of several types of cancer for almost 30 years, yet the mechanism(s) responsible for its anti-tumoral action remains unknown. A variety of cellular responses, including inhibition of cell growth and induction of apoptosis are induced by IFNs, and apoptotic induction by this cytokine has been proposed to be of importance for both its anti-tumoral in addition to its anti-viral responses. The aim of the present study was to delineate the pathways activated during IFNalpha-induced apoptosis in malignant cell lines. We found that apoptosis induced by IFNalpha was associated with activation of caspases-1, -2, -3, -8 and -9 and that this activation was a critical event. Caspase-3 activation was dependent on activity of caspases-8 and -9, moreover, activation of caspase-8 seems to be the upstream event in IFNalpha-induced caspase cascade. We also found loss of mitochondrial membrane potential as well as release of cytochrome c post IFN-treatment, clearly implicating the involvement of mitochondria in IFN-mediated apoptosis. Furthermore, IFNalpha-induced apoptosis was found to be independent on interactions between the Fas-receptor and its ligand. These studies form the basis for further investigations aiming to improve IFN therapy and the development of future strategies to overcome the IFN resistance observed in some malignancies.


Assuntos
Apoptose/efeitos dos fármacos , Interferon-alfa/farmacologia , Metástase Neoplásica/patologia , Neoplasias/patologia , Clorometilcetonas de Aminoácidos/farmacologia , Anexina A5/metabolismo , Western Blotting , Inibidores de Caspase , Caspases/metabolismo , Ciclo Celular/efeitos dos fármacos , Grupo dos Citocromos c/metabolismo , Ativação Enzimática/efeitos dos fármacos , Citometria de Fluxo , Humanos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Transdução de Sinais/efeitos dos fármacos , Células Tumorais Cultivadas , Receptor fas/metabolismo
6.
J Interferon Cytokine Res ; 25(12): 799-810, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16375608

RESUMO

Type I interferons (IFNs) are pleiotropic cytokines that exert multiple effects on normal and tumor cells. These effects are supposedly mediated through the stimulation of several signal transduction pathways by type I IFNs. These include the well-studied canonical Jak-Stat pathway, largely responsible for the antiviral and growth-inhibitory activities of IFNs, as well as the phosphatidylinositol 3-kinase (PI3K) and mitogen-activated protein kinase (MAPK) pathways, whose importance in IFN-induced biologic outcomes has not been precisely established. One of the effects of type I IFNs on tumor cells is the induction of programmed cell death, apoptosis, which has been studied extensively over the last decade and has been suggested to be an important effector mechanism for IFN's antitumor effects in the treatment of cancer. The aim of this review is to summarize and discuss the recent data in the field of type I IFN-induced apoptosis, with special emphasis on the molecular mechanisms of apoptosis and on the role of alternative noncanonical signaling pathways stimulated by type I IFNs in this process.


Assuntos
Apoptose/fisiologia , Interferon Tipo I/fisiologia , Transdução de Sinais/fisiologia , Animais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Humanos , Interferon Tipo I/farmacologia , Sistema de Sinalização das MAP Quinases , Mitocôndrias/fisiologia , Modelos Biológicos , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Neoplasias/fisiopatologia , Proteínas Proto-Oncogênicas c-bcl-2/fisiologia , Receptores do Fator de Necrose Tumoral/fisiologia , Proteínas Recombinantes , Estresse Fisiológico/fisiopatologia
7.
J Interferon Cytokine Res ; 25(2): 63-72, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15695927

RESUMO

Interferons (IFNs) exert antitumor effects in several human malignancies, but their mechanism of action is unclear. There is a great variability in sensitivity to IFN treatment depending on both tumor type and the individual patient. The reason for this variable sensitivity is not known. The fact that several IFN-induced anticellular effects are exerted through modulation of proto-oncogenes and tumor suppressor genes may indicate that the malignant genotype may be decisive in the cell's sensitivity to IFN. To determine if a deregulated oncogene could alter the cellular response to IFN, a mouse lymphoma cell line (J3D) was stably transfected with the viral human papillomavirus-16 (HPV-16) E7 oncogene. The E7-transfected cells and their respective mock-transfected sister clones were treated with IFN-alpha and examined for possible IFN-induced anticellular effects. We found that the E7-transfected clones were greatly sensitized to IFN-alpha-induced apoptosis compared with their mock-transfected counterparts. Induction of apoptosis in the transfected cells correlated with the ability of IFN to activate parts of the proapoptotic machinery specifically in these cells, including activation of caspases and the proapoptotic protein Bak. In summary, our data suggest that transfection of malignant cells with the E7 oncogene can sensitize them to IFN-alpha-induced apoptosis. This demonstrates that an oncogenic event may alter the cellular sensitivity to IFN and might also have implications for treatment of HPV-related diseases with IFN.


Assuntos
Apoptose/efeitos dos fármacos , Interferon-alfa/farmacologia , Proteínas Oncogênicas Virais/metabolismo , Papillomaviridae , Animais , Caspases/metabolismo , Linhagem Celular Transformada , Linhagem Celular Tumoral , Transformação Celular Viral , Ativação Enzimática/efeitos dos fármacos , Citometria de Fluxo , Proteínas de Membrana/metabolismo , Camundongos , Proteínas Oncogênicas Virais/genética , Proteínas E7 de Papillomavirus , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Retroviridae/genética , Transfecção , Proteína Killer-Antagonista Homóloga a bcl-2
8.
Nat Commun ; 3: 976, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22864569

RESUMO

Fbw7 is a ubiquitin-ligase that targets several oncoproteins for proteolysis, but the full range of Fbw7 substrates is not known. Here we show that by performing quantitative proteomics combined with degron motif searches, we effectively screened for a more complete set of Fbw7 targets. We identify 89 putative Fbw7 substrates, including several disease-associated proteins. The transcription factor NF-κB2 (p100/p52) is one of the candidate Fbw7 substrates. We show that Fbw7 interacts with p100 via a conserved degron and that it promotes degradation of p100 in a GSK3ß phosphorylation-dependent manner. Fbw7 inactivation increases p100 levels, which in the presence of NF-κB pathway stimuli, leads to increased p52 levels and activity. Accordingly, the apoptotic threshold can be increased by loss of Fbw7 in a p100-dependent manner. In conclusion, Fbw7-mediated destruction of p100 is a regulatory component restricting the response to NF-κB2 pathway stimulation.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Proteínas F-Box/metabolismo , Subunidade p52 de NF-kappa B/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Proteínas de Ciclo Celular/genética , Linhagem Celular , Linhagem Celular Tumoral , Biologia Computacional , Proteínas F-Box/genética , Proteína 7 com Repetições F-Box-WD , Quinase 3 da Glicogênio Sintase/genética , Quinase 3 da Glicogênio Sintase/metabolismo , Humanos , Immunoblotting , Imunoprecipitação , Subunidade p52 de NF-kappa B/genética , Fosforilação/genética , Fosforilação/fisiologia , Proteômica , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Espectrometria de Massas em Tandem , Ubiquitina-Proteína Ligases/genética
9.
Autophagy ; 5(8): 1198-200, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19855186

RESUMO

Glucocorticoids are fundamental drugs used in the treatment of lymphoid malignancies with apoptotic cell death as the hitherto proposed mechanism of action. We have recently shown that dexamethasone induces autophagy in lymphoid leukemia cells and in this particular setting this cell death modality is a prerequisite for the efficient killing of the leukemic cells by dexamethasone. Hence, inhibition of autophagy by siRNA-mediated silencing of Beclin 1, as well as chemical inhibition of type III PtdIns3K, inhibits apoptosis, demonstrating an important role of autophagy in dexamethasone-induced cell death. In this brief report, we review these findings and introduce the multiple myeloma cells as a novel system to study autophagy in response to dexamethasone.


Assuntos
Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Dexametasona/farmacologia , Glucocorticoides/farmacologia , Neoplasias Hematológicas/patologia , Anexina A5/metabolismo , Humanos , Propídio/metabolismo , Coloração e Rotulagem
10.
Exp Cell Res ; 313(2): 404-14, 2007 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-17141757

RESUMO

IFNalpha activates JAK-STAT signaling, followed by up-regulation of a cohort of genes. Also the PI3K pathway is activated by IFNalpha, but the significance of this activation for IFN-induced gene expression and biological functions remains unclear. We used a cDNA microarray to identify IFNalpha target genes whose expression is dependent on PI3K signaling. cDNAs from U266-1984 cells, untreated and IFNalpha-treated with or without PI3K inhibitor, Ly294002, was used in hybridization to a microarray representing 7000 genes. Among the 260 genes stimulated by IFNalpha, the expression of 95.4% was not affected by the presence of Ly294002. Luciferase reporter assays using consensus IFN-stimulated sequences confirmed that general regulation of transcription by IFNalpha is not altered by Ly294002. Up-regulation of 10 genes (3.8%) was affected in the presence of Ly294002. Bioinformatic analysis revealed the presence of consensus sequences of both STAT-specific and the PI3K pathway-regulated transcription factors, further suggesting that these genes are regulated by both pathways. We have recently shown that IFNalpha-induced apoptosis in the myeloma cell line U266-1984 was efficiently blocked by inhibition of PI3K. Therefore we suggest that the genes that are regulated by both the STAT and the PI3K pathways by IFNalpha in these cells may be specifically involved in the induction of apoptosis.


Assuntos
Regulação da Expressão Gênica , Interferon-alfa/farmacologia , Fosfatidilinositol 3-Quinases/fisiologia , Apoptose/genética , Sequência de Bases , Linhagem Celular Tumoral , Cromonas/farmacologia , Biologia Computacional , Sequência Consenso , Inibidores Enzimáticos/farmacologia , Expressão Gênica/efeitos dos fármacos , Genes Reporter/efeitos dos fármacos , Humanos , Interferon-alfa/fisiologia , Luciferases/análise , Luciferases/genética , Morfolinas/farmacologia , Análise de Sequência com Séries de Oligonucleotídeos , Inibidores de Fosfoinositídeo-3 Quinase , Regiões Promotoras Genéticas/efeitos dos fármacos , Fatores de Transcrição STAT/fisiologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina/genética
11.
J Biol Chem ; 277(46): 44317-26, 2002 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-12193597

RESUMO

The anthracyclin doxorubicin (DXR) is a major antitumor agent known to cause cellular damage via a number of mechanisms including free radical formation and inhibition of topoisomerase II. It is not clear, however, how the subsequent lesions may lead to the apoptotic death of the cell. We have here examined the effects of DXR on activation of pro-apoptotic members of the Bcl-2 family, all of which are connected to the mitochondrial events of apoptosis. In two human cell lines (lymphoma and myeloma), clinically relevant concentrations of DXR were found to induce apoptosis, first observed after 24 h of treatment. Apoptosis correlated with modulation of Bak and Bax to their active conformations. bax- as well as bak-deficient mouse embryo fibroblasts were resistant to DXR compared with wild-type mouse embryo fibroblasts further supporting a role for these proteins as main DXR-induced apoptosis regulators. Furthermore, using immunocytochemistry as well as chemical blocking of putative apical pathways we could demonstrate that Bak is activated prior to Bax. In the human cell lines, DXR was furthermore found to induce high protein levels of Bik, another BH3-only protein. DXR-induced apoptosis was completely blocked in Bcl-2-overexpressing U266 cells. Interestingly, in Bcl-2-transfected cells Bak activation was also blocked, while Bax was still partially active in agreement with differential regulation of these two proteins. Furthermore, co-incubation of the phosphatidylinositol 3-kinase (PI3K)-inhibitor LY294002 potentiated the apoptotic response to DXR. This enhanced apoptosis was preceded by enhanced Bak and Bax activation, and both responses as well as apoptosis were blocked in transfectants overexpressing Bcl-2. In summary, several pieces of evidence suggest that DXR induces apoptosis through a sequential and differential activation of Bak and Bax.


Assuntos
Apoptose , Doxorrubicina/farmacologia , Proteínas de Membrana/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2 , Proteínas Proto-Oncogênicas/metabolismo , Animais , Antineoplásicos/farmacologia , Western Blotting , Linhagem Celular , Cromonas/farmacologia , Inibidores Enzimáticos/farmacologia , Fibroblastos/metabolismo , Citometria de Fluxo , Camundongos , Microscopia de Fluorescência , Modelos Biológicos , Morfolinas/farmacologia , Inibidores de Fosfoinositídeo-3 Quinase , Fatores de Tempo , Transfecção , Células Tumorais Cultivadas , Proteína Killer-Antagonista Homóloga a bcl-2 , Proteína X Associada a bcl-2
12.
J Gen Virol ; 82(Pt 2): 345-358, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11161273

RESUMO

We have previously shown that Epstein-Barr virus (EBV)-encoded EBNA-5 is localized to PML bodies (PODs) in EBV-immortalized lymphoblastoid cell lines (LCLs). Here we have extended our study of the subnuclear localization of EBNA-5 and found a strict co-localization with PML in LCLs and in BL lines with an immunoblastic, LCL-like phenotype. Moreover, GFP-EBNA-5 accumulated in PML bodies upon transfection into LCLs. In contrast, transfection of cell lines of non-immunoblastic origin with an EBNA-5 expression construct showed preferential localization of the protein to the nucleoplasm. Since PML is involved in proteasome-dependent protein degradation, we investigated the total levels and sub-cellular localization of EBNA-5 upon inhibition of proteasome activity. We found that a proteasome inhibitor, MG132, induced the translocation of both endogenous and transfected EBNA-5 to the nucleoli in every cell line tested. The total EBNA-5 protein levels were not affected by the proteasomal block. EBNA-5 forms complexes with heat shock protein Hsp70. The proteasome inhibitor induced a rise in total levels of Hsp70 and dramatically changed its homogeneous nuclear and cytoplasmic distribution into nucleolar and cytoplasmic. This effect was EBNA-5-independent. The nucleolar localization of Hsp70 was enhanced by the presence of EBNA-5, however. EBNA-5 also enhanced the nucleolar translocation of a mutant p53 in a colon cancer line, SW480, treated with MG132. The coordinated changes in EBNA-5 and Hsp70 localization and the effect of EBNA-5 on mutant p53 distribution upon MG132 treatment might reflect the involvement of EBNA-5 in the regulation of intracellular protein trafficking associated with the proteasome-mediated degradation.


Assuntos
Nucléolo Celular/metabolismo , Antígenos Nucleares do Vírus Epstein-Barr/metabolismo , Herpesvirus Humano 4/fisiologia , Leupeptinas/farmacologia , Complexos Multienzimáticos/antagonistas & inibidores , Linfócitos B/efeitos dos fármacos , Linfócitos B/metabolismo , Nucléolo Celular/química , Nucléolo Celular/efeitos dos fármacos , Cisteína Endopeptidases/metabolismo , Citoplasma/efeitos dos fármacos , Citoplasma/metabolismo , Antígenos Nucleares do Vírus Epstein-Barr/genética , Proteínas de Choque Térmico HSP70/metabolismo , Humanos , Complexos Multienzimáticos/metabolismo , Mutação/genética , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , Matriz Nuclear/química , Matriz Nuclear/efeitos dos fármacos , Matriz Nuclear/metabolismo , Complexo de Endopeptidases do Proteassoma , Ligação Proteica , Transporte Proteico/efeitos dos fármacos , Transfecção , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
13.
Int J Cancer ; 105(5): 644-53, 2003 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-12740913

RESUMO

Epstein-Barr virus (EBV) carrying lymphoblastoid cells of normal origin express the full program of all 9 virus-encoded, growth transformation associated proteins. They have an intact p53 pathway as a rule. This raises the question of whether any of the viral proteins impair the pathway functionally. Using a yeast 2-hybrid system, we have shown that EBNA-5 but not the other EBNAs interacts with the p14ARF protein, a regulator of the p53 pathway. The interaction was confirmed in vitro using a GST pull-down assay. Moreover, expression of EBNA-5 increased the survival of p14ARF-transfected cells. EBV infection of resting B cells induced the expression of p14ARF mRNA without increased level of the protein. A fraction of the p14ARF localized to the nucleoli but the bulk of the protein accumulated in nuclear but extranucleolar inclusions. Formation of the extranucleolar inclusions led to complete relocalization of EBNA-5 from nucleoplasm to these structures. The inclusions also contained p53 and HDM2, and were surrounded by PML bodies and proteasomes, which suggests that these inclusions could be targets for proteasome dependent protein degradation.


Assuntos
Linfócitos B/virologia , Núcleo Celular/metabolismo , Antígenos Nucleares do Vírus Epstein-Barr/fisiologia , Herpesvirus Humano 4/fisiologia , Proteínas Nucleares , Proteína Supressora de Tumor p14ARF/metabolismo , Células 3T3/metabolismo , Células 3T3/virologia , Transporte Ativo do Núcleo Celular , Adenocarcinoma/patologia , Animais , Linfócitos B/metabolismo , Linfócitos B/ultraestrutura , Neoplasias Ósseas/patologia , Nucléolo Celular/metabolismo , Sobrevivência Celular , Transformação Celular Viral/fisiologia , Neoplasias do Colo/patologia , Cisteína Endopeptidases/metabolismo , Regulação Viral da Expressão Gênica , Células HeLa/metabolismo , Células HeLa/virologia , Humanos , Camundongos , Complexos Multienzimáticos/metabolismo , Osteossarcoma/patologia , Complexo de Endopeptidases do Proteassoma , Ligação Proteica , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-mdm2 , Proteínas Recombinantes de Fusão/metabolismo , Transfecção , Células Tumorais Cultivadas/metabolismo , Células Tumorais Cultivadas/virologia , Proteína Supressora de Tumor p14ARF/genética , Proteína Supressora de Tumor p53/metabolismo , Técnicas do Sistema de Duplo-Híbrido
14.
J Gen Virol ; 80 ( Pt 11): 2889-2900, 1999 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-10580050

RESUMO

Subnuclear distribution of the human herpesvirus-8 (HHV-8)- encoded nuclear protein LNA-1 was analysed at high resolution in body cavity (BC) lymphoma-derived cell lines, in cell hybrids between BC cells and various human and mouse cells and in freshly infected K562 and ECV cell lines. Three-dimensional reconstruction of nuclei from optical sections and quantitative analysis of the distribution of LNA-1 fluorescence in relation to chromatin showed that LNA-1 associates preferentially with the border of heterochromatin in the interphase nuclei. This was further confirmed in the following systems: in endo- and exonuclease-digested nuclei, in human-mouse (BC-1-Sp2- 0) hybrids and on chromatin spreads. LNA-1 was found to bind to mitotic chromosomes at random. Epstein-Barr virus (EBV), but not HHV-8, was rapidly lost from mouse-human hybrid cells in parallel with the loss of human chromosomes. HHV-8 could persist on the residual mouse background for more than 8 months. In early human-mouse hybrids that contain a single fused nucleus, LNA-1 preferentially associates with human chromatin. After the gradual loss of the human chromosomes, LNA-1 becomes associated with the murine pericentromeric heterochromatin. In human-human hybrids derived from the fusion of the HHV-8-carrying BCBL-1 cells and the EBV-immortalized lymphoblastoid cell line IB4, LNA-1 did not co-localize with EBNA-1, EBNA-2, EBNA-5 or EBNA-6. LNA-1 was not associated with PML containing ND10 bodies either. DNase but not RNase or detergent treatment of isolated nuclei destroys LNA-1 bodies. In advanced apoptotic cells LNA- 1 bodies remain intact but are not included in the apoptotic bodies themselves.


Assuntos
Antígenos Virais/análise , Núcleo Celular/química , Herpesvirus Humano 8/química , Heterocromatina/química , Proteínas Nucleares/análise , Fosfoproteínas , Animais , Apoptose , Linhagem Celular , Cromossomos/química , Antígenos Nucleares do Vírus Epstein-Barr/análise , Humanos , Células Híbridas , Metáfase , Camundongos
15.
Int J Cancer ; 99(5): 705-12, 2002 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-12115504

RESUMO

Epstein-Barr virus (EBV) drives the proliferation of human B cells in vitro and during primary infection in vivo. The transformed immunoblasts express nuclear proteins EBNA1-6, transcribed from the Cp/Wp promoter, and the membrane proteins LMP-1, -2A and -2B (lymphoblastoid type of latency). EBV persists through life in resting memory B cells with a restricted type of latency in the absence of the Cp/Wp promoter activity. Since CD40 crosslinking can reportedly inhibit the growth of EBV-transformed lymphoblastoid cell lines (LCLs), we have examined the effect of CD40 ligation on the expression of EBNAs and LMP-1 and on Cp EBV promoter activity together with several phenotypic markers. CD40 crosslinking led to a partial downregulation of EBNA-2, EBNA3-6 and LMP-1 in LCLs, paralleled by downregulation of Cp promoter activity. It also induced upregulation of the germinal center marker CD77 on the LCL cells. Our findings suggest that the encounter of proliferating EBV-transformed immunoblasts with CD40L, as would occur when normal B cells generate memory cells in germinal centers, may switch the viral transcription program from the full lymphoblastoid to a more restricted latency program in a proportion of cells. This would permit virus persistence in the B-cell memory compartment.


Assuntos
Linfócitos B/metabolismo , Linfócitos B/virologia , Antígenos CD40/metabolismo , Proteínas de Transporte/genética , Antígenos Nucleares do Vírus Epstein-Barr/genética , Proteínas Adaptadoras de Transdução de Sinal , Antígenos CD/análise , Linfócitos B/imunologia , Western Blotting , Ligante de CD40/metabolismo , Ligante de CD40/farmacologia , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Transformação Celular Viral , Proteínas do Citoesqueleto , Citometria de Fluxo , Imunofluorescência , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas com Domínio LIM , RNA Mensageiro/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas Virais
16.
J Biol Chem ; 279(23): 24152-62, 2004 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-15056668

RESUMO

Interferon (IFN) alpha induces a caspase-dependent apoptosis that is associated with activation of the proapoptotic Bak and Bax, loss of mitochondrial membrane potential, and release of cytochrome c. In addition to the onset of the classical Jak-STAT pathway, IFNalpha also induced phosphoinositide 3-kinase (PI3K) activity. Pharmacological inhibition of PI3K activity by Ly294002 disrupted IFN-induced apoptosis upstream of mitochondria. Inhibition of mTOR by rapamycin or by overexpression of a kinase dead mutant of mTOR, efficiently blocked IFNalpha-induced apoptosis. A PI3K and mTOR-dependent phosphorylation of p70S6 kinase and 4E-BP1 repressor was induced by IFNalpha treatment of cells and was strongly inhibited by Ly294002 or rapamycin. The activation of Jak-STAT signaling upon IFNalpha stimulation was not affected by abrogating PI3K/mTOR pathway. Neither was the expression of several IFNalpha target genes affected, nor the ability of IFNalpha to protect against virus-induced cell death affected by inhibition of the PI3K/mTOR pathway. These data demonstrate that an intact PI3K/mTOR pathway is necessary for the ability of IFNalpha to induce apoptosis, whereas activation of the Jak-STAT pathway alone appears to be insufficient for this specific IFNalpha-induced effect.


Assuntos
Apoptose , Interferon-alfa/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Quinases/metabolismo , Transdução de Sinais , Proteínas Adaptadoras de Transdução de Sinal , Anexina A5/farmacologia , Antibióticos Antineoplásicos/farmacologia , Western Blotting , Proteínas de Transporte/metabolismo , Caspases/metabolismo , Proteínas de Ciclo Celular , Morte Celular , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Cromonas/farmacologia , Corantes/farmacologia , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Proteínas de Fluorescência Verde , Humanos , Imuno-Histoquímica , Interferon-alfa/metabolismo , Queratinócitos/metabolismo , Proteínas Luminescentes/metabolismo , Potenciais da Membrana , Mitocôndrias/metabolismo , Morfolinas/farmacologia , Fosfoproteínas/metabolismo , Fosforilação , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Sirolimo/farmacologia , Serina-Treonina Quinases TOR , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA