Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 85
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Gen Virol ; 85(Pt 1): 185-196, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14718634

RESUMO

We have examined the subcellular localization properties of human adenovirus 2 (HAdV-2) preMu and mature Mu (pX) proteins as fusions with enhanced green fluorescence protein (EGFP). We determined that preMu is exclusively a nucleolar protein with a single nucleolar accumulation signal within the Mu sequence. In addition, we noted that both preMu-EGFP and Mu-EGFP are excluded from adenovirus DNA-binding protein (DBP)-rich replication centres in adenovirus-infected cells. Surprisingly, we observed that cells in which preMu-EGFP (but not Mu-EGFP) is transiently expressed prior to or shortly after infection with Ad2 did not express late adenovirus genes. Further investigation suggested this might be due to a failure to express pre-terminal protein (preTP) from the E2 region, despite expression of another E2 protein, DBP. Deletion mutagenesis identified a highly conserved region in the C terminus of preMu responsible for these observations. Thus our data suggest that preMu may play a role in modulating accumulation of proteins from the E2 region.


Assuntos
Proteínas E2 de Adenovirus/metabolismo , Nucléolo Celular/metabolismo , Proteínas de Ligação a DNA/metabolismo , Regulação Viral da Expressão Gênica , Peptídeos/metabolismo , Precursores de Proteínas/metabolismo , Proteínas do Core Viral/metabolismo , Adenovírus Humanos/patogenicidade , Sequência de Aminoácidos , Deleção de Genes , Proteínas de Fluorescência Verde , Células HeLa , Humanos , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Dados de Sequência Molecular , Proteínas Recombinantes de Fusão/metabolismo
2.
Curr Top Microbiol Immunol ; 272: 253-86, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12747553

RESUMO

Adenovirus makes extensive use of RNA splicing to produce a complex set of spliced mRNAs during virus replication. All transcription units, except pIX and IVa2, encode multiple alternatively spliced mRNAs. The accumulation of viral mRNAs is subjected to a temporal regulation, a mechanism that ensures that proteins that are needed at certain stages of the viral life cycle are produced. The complex interaction between host cell RNA splicing factors and viral regulatory elements has been studied intensely during the last decade. Such studies have begun to produce a picture of how adenovirus remodels the host cell RNA splicing machinery to orchestrate the shift from the early to the late profile of viral mRNA accumulation. Recent progress has to a large extent focused on the mechanisms regulating E1A and L1 alternative splicing. Here we will review the current knowledge of cis-acting sequence element, trans-acting factors and mechanisms controlling E1A and L1 alternative splicing.


Assuntos
Infecções por Adenoviridae/metabolismo , Processamento Alternativo , Splicing de RNA/fisiologia , Spliceossomos/metabolismo , Adenovírus Humanos/genética , Adenovírus Humanos/fisiologia , Animais , DNA Viral/biossíntese , Éxons , Regulação Viral da Expressão Gênica , Humanos , Íntrons , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
3.
J Virol ; 75(20): 9579-84, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11559789

RESUMO

To construct recombinant adenoviruses expressing biologically active proteins may be impossible, or result in a significant reduction in virus yield, if the protein expressed has an inhibitory effect on virus replication or cellular growth. To overcome this problem, we previously designed adenovirus vectors expressing foreign proteins from inducible promoters. However, during our work with a replication-deficient virus expressing the ASF/SF2 splicing factor from a progesterone antagonist-inducible gene cassette, we discovered that ASF/SF2 was expressed at a significant level in the 293 producer cell line, even in the absence of inducer. 293 cells code for adenovirus E1A and E1B proteins and thus support the growth of E1-deficient adenoviruses. Here we show that this background ASF/SF2 expression results from a low level of E1A-mediated transactivation of the basal promoter driving transgene expression. To overcome the problem of leaky expression, we reconstructed a novel gene cassette that combines an inducible promoter and a Lac repressor protein-based block to reduce transcriptional elongation. We show that this novel vector system dramatically reduced background transgene expression and therefore should be useful for the rescue and propagation of high-titer stocks of recombinant adenoviruses expressing toxic proteins.


Assuntos
Adenoviridae/genética , Proteínas E1A de Adenovirus/metabolismo , Vetores Genéticos , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Fatores de Transcrição/metabolismo , Adenoviridae/fisiologia , Linhagem Celular , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-ets , Splicing de RNA , Proteínas de Ligação a RNA , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Fatores de Processamento de Serina-Arginina , Transativadores , Transcrição Gênica , Replicação Viral
4.
Mol Cell Biol ; 21(11): 3684-91, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11340162

RESUMO

In a panel of four human melanoma cell lines, equitoxic doses of cisplatin induced the proapoptotic conformation of the Bcl-2 family protein Bak prior to the execution phase of apoptosis. Because cisplatin-induced modulation of the related Bax protein was seen in only one cell line, a degree of specificity in the signal to Bak is indicated. Little is known about upstream regulation of Bak activity. In this study, we examined whether the apoptosis-specific pathway mediated by a kinase fragment of MEKK1 (DeltaMEKK1) is involved in the observed Bak modulation. We report that expression of a kinase-inactive fragment of MEKK1 (dominant negative MEKK [dnMEKK]) efficiently blocked cisplatin-induced modulation of Bak and cytochrome c release and consequently also reduced DEVDase activation and nuclear fragmentation. Accordingly, expression of a kinase-active MEKK1 fragment (dominant positive MEKK) was sufficient to induce modulation of Bak in three cell lines and to induce apoptosis in two of these. dnMEKK did not block cisplatin-induced c-Jun N-terminal kinase (JNK) activation, in agreement with a specifically proapoptotic role for the DeltaMEKK1 pathway. Finally, we show that reduction of Bak expression by antisense Bak reduced cisplatin-induced loss of mitochondrial integrity and caspase cleavage activity in breast cancer cell lines. In summary, we have identified Bak as a cisplatin-regulated component downstream in a proapoptotic, JNK-independent DeltaMEKK1 pathway.


Assuntos
Antineoplásicos/metabolismo , Apoptose , Cisplatino/metabolismo , MAP Quinase Quinase Quinase 1 , Proteínas de Membrana/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Antineoplásicos/farmacologia , Cisplatino/farmacologia , Grupo dos Citocromos c/metabolismo , Ativação Enzimática , Humanos , Proteína Quinase 8 Ativada por Mitógeno , Proteína Quinase 9 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Células Tumorais Cultivadas , Proteína Killer-Antagonista Homóloga a bcl-2 , Proteína X Associada a bcl-2
5.
EMBO J ; 20(4): 864-71, 2001 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-11179230

RESUMO

SR proteins purified from uninfected HeLa cells inhibit adenovirus IIIa pre-mRNA splicing by binding to the intronic IIIa repressor element (3RE). In contrast, SR proteins purified from late adenovirus-infected cells are functionally inactivated as splicing repressor proteins by a virus-induced dephosphorylation. We have shown that the adenovirus E4-ORF4 protein, which binds the cellular protein phos phatase 2A (PP2A) and activates IIIa splicing in vitro and in vivo, induces SR protein dephosphorylation. Here we show that E4-ORF4 interacts with only a subset of SR proteins present in HeLa cells. Thus, E4-ORF4 interacts efficiently with SF2/ASF and SRp30c, but not with other SR proteins. Interestingly, E4-ORF4 interacts with SF2/ASF through the latter's RNA recognition motifs. Furthermore, E4-ORF4 interacts preferentially with the hyperphosphorylated form of SR proteins found in uninfected HeLa cells. E4-ORF4 mutant proteins that fail to bind strongly to PP2A or SF2/ASF do not relieve the repressive effect of HeLa SR proteins on IIIa pre-mRNA splicing in transient transfection experiments, suggesting that an interaction between all three proteins is required for E4-ORF4-induced SR protein dephosphorylation.


Assuntos
Proteínas E4 de Adenovirus/metabolismo , Proteínas de Neoplasias/metabolismo , Splicing de RNA , Células HeLa , Humanos , Fases de Leitura Aberta , Fosforilação , Ligação Proteica
6.
J Virol ; 74(19): 9002-9, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10982344

RESUMO

Expression of cytoplasmic mRNA from most adenovirus transcription units is subjected to a temporal regulation at the level of alternative pre-mRNA splicing. The general tendency is that splice site selection changes from proximal to distal late after infection. Interestingly, ASF/SF2, which is a prototypical member of the SR family of splicing factors, has the opposite effect on splice site selection, inducing an increase in proximal splice site usage. We have previously shown that SR proteins late during an adenovirus infection become partially inactivated as splicing regulatory proteins. A prediction from these results is that overexpression of an SR protein, such as ASF/SF2, during virus growth will interfere with virus replication by disturbing the balance of functional and nonfunctional ASF/SF2 in the infected cell. To test this hypothesis, we reconstructed a recombinant adenovirus expressing ASF/SF2 under the transcriptional control of a regulated promoter. The results show that, as predicted, induction of ASF/SF2 during lytic virus growth prevents the early to late shift in mRNA expression from both early (E1A and E1B) and late (L1) transcription units. Furthermore, ASF/SF2 overexpression blocks viral DNA replication and reduces selectively cytoplasmic accumulation of major late mRNA, resulting in a lower virus yield. Collectively, our results provide additional support for the hypothesis that viral control of SR protein function is important for the proper expression of viral proteins during lytic virus growth.


Assuntos
Adenoviridae/genética , Regulação Viral da Expressão Gênica , Proteínas Nucleares/genética , Precursores de RNA/genética , RNA Viral/genética , Células HeLa , Humanos , Splicing de RNA , Proteínas de Ligação a RNA , Fatores de Processamento de Serina-Arginina
7.
Cell Growth Differ ; 11(7): 373-84, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10939591

RESUMO

The genes encoding the cyclin-dependent kinase inhibitors p16INK4A (CDKN2A) and p15INK4B (CDKN2B) are frequently homozygously deleted in a variety of tumor cell lines and primary tumors, including glioblastomas in which 40-50% of primary tumors display homozygous deletions of these two loci. Although the role of p16 as a tumor suppressor has been well documented, it has remained less well studied whether p15 plays a similar growth-suppressing role. Here, we have used replication-defective recombinant adenoviruses to compare the effects of expressing wild-type p16 and p15 in glioma cell lines. After infection, high levels of p16 and p15 were observed in two human glioma cell lines (U251 MG and U373 MG). Both inhibitors were found in complex with CDK4 and CDK6. Expression of p16 and p15 had indistinguishable effects on U251 MG, which has homozygous deletion of CDKN2A and CDKN2B, but a wild-type retinoblastoma (RB) gene. Cells were growth-arrested, showed no increased apoptosis, and displayed a markedly altered cellular morphology and repression of telomerase activity. Transduced cells became enlarged and flattened and expressed senescence-associated beta-galactosidase, thus fulfilling criteria for replicative senescence. In contrast, the growth and morphology of U373 MG, which expresses p16 and p15 endogenously, but undetectable levels of RB protein, were not affected by exogenous overexpression of either inhibitor. Thus, we conclude that overexpression of p15 has a similar ability to inhibit cell proliferation, to cause replicative senescence, and to inhibit telomerase activity as p16 in glioma cells with an intact RB protein pathway.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular , Divisão Celular , Senescência Celular , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Quinases Ciclina-Dependentes/antagonistas & inibidores , Proteínas Proto-Oncogênicas , Telomerase/antagonistas & inibidores , Proteínas Supressoras de Tumor , Adenoviridae/genética , Adenoviridae/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/imunologia , Linhagem Celular , Tamanho Celular , Ciclina D1/genética , Ciclina D1/metabolismo , Quinase 4 Dependente de Ciclina , Quinase 6 Dependente de Ciclina , Inibidor de Quinase Dependente de Ciclina p15 , Inibidor p16 de Quinase Dependente de Ciclina/genética , Inibidor p16 de Quinase Dependente de Ciclina/imunologia , Quinases Ciclina-Dependentes/genética , Quinases Ciclina-Dependentes/metabolismo , Indução Enzimática , Glioma , Humanos , Immunoblotting , Mutação , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Recombinantes/metabolismo , Proteína do Retinoblastoma/genética , Proteína do Retinoblastoma/metabolismo , Telomerase/metabolismo , Transfecção , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
8.
FEBS Lett ; 476(3): 248-52, 2000 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-10913622

RESUMO

The adenovirus E1B-55K protein is a multifunctional phosphoprotein that regulates nuclear to cytoplasmic export of host cell and viral mRNAs during lytic viral growth. E1B-55K also blocks apoptosis by binding and functionally inactivating the human tumor suppressor protein p53. Here, we show that E1B-55K interacts with histone deacetylase 1 (HDAC1) and the transcriptional corepressor protein mSin3A, both in the adenovirus-transformed 293 cell line and during a lytic adenovirus infection. Furthermore, we show that the central amino acids 156-261 in E1B-55K are necessary for efficient HDAC1 interaction. Importantly, the E1B-55K/mSin3A/HDAC1 complex is also enzymatically active, catalyzing deacetylation of a histone substrate peptide. Collectively, our results suggest that E1B-55K interaction with mSin3A/HDAC1 containing complexes may be significant for one or several of the multiple activities ascribed to this protein.


Assuntos
Proteínas E1B de Adenovirus/metabolismo , Adenovírus Humanos/metabolismo , Histona Desacetilases/metabolismo , Proteínas Repressoras/metabolismo , Proteínas E1B de Adenovirus/química , Proteínas E1B de Adenovirus/genética , Adenovírus Humanos/genética , Animais , Células COS , Histona Desacetilase 1 , Histona Desacetilases/química , Histona Desacetilases/genética , Humanos , Técnicas In Vitro , Substâncias Macromoleculares , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Repressoras/química , Proteínas Repressoras/genética , Complexo Correpressor Histona Desacetilase e Sin3 , Transfecção
9.
Mol Cell Biol ; 20(7): 2317-25, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10713155

RESUMO

Splicing of the adenovirus IIIa pre-mRNA is subjected to a temporal regulation, such that efficient IIIa 3' splice site usage is confined to the late phase of the infectious cycle. Here we show that IIIa pre-mRNA splicing is activated more than 200-fold in nuclear extracts prepared from late adenovirus-infected cells (Ad-NE) compared to uninfected HeLa cell nuclear extracts (HeLa-NE). In contrast, splicing of the beta-globin pre-mRNA is repressed in Ad-NE. We constructed hybrid pre-mRNAs between IIIa and beta-globin in order to identify the minimal IIIa sequence element conferring enhanced splicing in Ad-NE. Using this approach, we show that the IIIa branch site/pyrimidine tract functions as a Janus element: it blocks splicing in HeLa-NE and functions as a splicing enhancer in Ad-NE. Therefore, we named this sequence the IIIa virus infection-dependent splicing enhancer (3VDE). This element is essential for regulated IIIa pre-mRNA splicing in Ad-NE and sufficient to confer an enhanced splicing phenotype to the beta-globin pre-mRNA in Ad-NE. We further show that the increase in IIIa splicing observed in Ad-NE is not accompanied by a similar increase in U2AF binding to the IIIa pyrimidine tract. This finding suggests that splicing activation by the 3VDE may operate without efficient U2AF interaction with the pre-mRNA. Importantly, this report represents the first description of a splicing enhancer that has evolved to function selectively in the context of a virus infection, a finding that adds a new level at which viruses may subvert the host cell RNA biosynthetic machinery to facilitate their own replication.


Assuntos
Adenoviridae/genética , Regulação Viral da Expressão Gênica/genética , Precursores de RNA/metabolismo , Splicing de RNA , Globinas/genética , Células HeLa , Humanos , Mutação , Proteínas Nucleares , RNA Viral/metabolismo , Proteínas de Ligação a RNA/genética , Ribonuclease H/metabolismo , Ribonucleoproteína Nuclear Pequena U2/metabolismo , Ribonucleoproteínas/genética , Fator de Processamento U2AF
10.
Nucleic Acids Res ; 28(5): E14, 2000 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-10666475

RESUMO

Mammalian proteins expressed in Escherichia coli are used in a variety of applications. A major drawback in producing eukaryotic proteins in E.coli is that the bacteria lack most eukaryotic post-translational modification systems, including serine/threonine protein kinase(s). Here we show that a eukaryotic protein can be phosphorylated in E.coli by simultaneous expression of a mammalian protein kinase and its substrate. We show that in bacteria expressing SRPK1, ASF/SF2 becomes phosphorylated to a degree resembling native ASF/SF2 present in interphase HeLa cell nuclei. The E.coli phosphorylated ASF/SF2 is functional in splicing and, contrary to the unphosphorylated protein, soluble under native conditions.


Assuntos
Proteínas Nucleares/genética , Proteínas Serina-Treonina Quinases/genética , Splicing de RNA , Clonagem Molecular , Escherichia coli , Células HeLa , Humanos , Proteínas Nucleares/isolamento & purificação , Proteínas Nucleares/metabolismo , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas de Ligação a RNA , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Fatores de Processamento de Serina-Arginina , Solubilidade
11.
Apoptosis ; 5(4): 355-67, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11227217

RESUMO

Mutationally activated Ras is involved in tumor progression and likely also in drug resistance. Using survival, viability and apoptosis assays, we have here compared the cisplatin sensitivities of FR3T3 rat fibroblasts and a 12V-H-ras transformed subline (Ras2:3). Around 24 h after cisplatin treatment Ras2:3 cells showed higher apoptosis levels and lower viability than FR3T3. This increased sensitivity correlated with weaker cisplatin-induced activation of Jun N-terminal kinase (JNK). In contrast to apoptosis assays, colony formation assays showed that Ras2:3 were more resistant to cisplatin than were FR3T3. This was partly due to the increased cisplatin sensitivity of FR3T3 seeded at low densities, as required in colony formation assays. In addition, Ras2:3 cisplatin survivors had a higher relative proliferative capacity. Cell cycle analyses showed that FR3T3 cells initially responded with a dose-dependent G2 arrest, while Ras2:3 accumulated in S-phase. Experiments with an anti-apoptotic mutant of MEKK1 suggested that the apoptotic response of Ras2:3 cells is not specific to the S-phase fraction. In summary, the cisplatin response of ras-transformed fibroblasts is distinct from that of parental cells, in that they show increased apoptosis, a different cell cycle response and increased post-treatment proliferative capacity. The results illustrate the need to carefully consider methods and protocols for in vitro studies on chemotherapy sensitivity.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Transformação Celular Neoplásica , Cisplatino/farmacologia , Genes ras , MAP Quinase Quinase Quinase 1 , Animais , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Transformada , Núcleo Celular/metabolismo , Relação Dose-Resposta a Droga , Ativação Enzimática , Fibroblastos , Citometria de Fluxo , Humanos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Ratos , Ensaio Tumoral de Célula-Tronco
12.
FEBS Lett ; 451(1): 10-4, 1999 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-10356974

RESUMO

Splicing enhancers have previously been shown to promote processing of introns containing weak splicing signals. Here, we extend these studies by showing that also 'strong' constitutively active introns are absolutely dependent on a downstream splicing enhancer for activity in vitro. SR protein binding to exonic enhancer elements or U1 snRNP binding to a downstream 5' splice site serve redundant functions as activators of splicing. We further show that a 5' splice site is most effective as an enhancer of splicing. Thus, a 5' splice site is functional in S100 extracts, under conditions where a SR enhancer is nonfunctional. Also, splice site pairing occurs efficiently in the absence of exonic SR enhancers, emphasizing the significance of a downstream 5' splice site as the enhancer element in vertebrate splicing.


Assuntos
Adenovírus Humanos/genética , Elementos Facilitadores Genéticos , Proteínas de Homeodomínio/genética , Precursores de RNA , Splicing de RNA , Animais , Drosophila/genética , Proteínas de Drosophila , Fatores de Transcrição Fushi Tarazu , Genes de Insetos , Genes Virais , Células HeLa , Humanos , Íntrons , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Proteínas de Ligação a RNA , Ribonucleoproteína Nuclear Pequena U1/metabolismo , Fatores de Processamento de Serina-Arginina
13.
Virology ; 256(2): 313-21, 1999 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-10191196

RESUMO

Previous studies have shown that the cell cycle-regulated E2F transcription factor is subjected to both positive and negative control by phosphorylation. Here we show that in transient transfection experiments, adenovirus E1A activation of the viral E2 promoter is abrogated by coexpression of the viral E4 open reading frame 4 (E4-ORF4) protein. This effect does not to require the retinoblastoma protein that previously has been shown to regulate E2F activity. The inhibitory activity of E4-ORF4 appears to be specific because E4-ORF4 had little effect on, for example, E4-ORF6/7 transactivation of the E2 promoter. We further show that the repressive effect of E4-ORF4 on E2 transcription works mainly through the E2F DNA-binding sites in the E2 promoter. In agreement with this, we find that E4-ORF4 inhibits E2F-1/DP-1-mediated transactivation. We also show that E4-ORF4 inhibits E2 mRNA expression during virus growth. E4-ORF4 has previously been shown to bind to and activate the cellular protein phosphatase 2A. The inhibitory effect of E4-ORF4 was relieved by okadaic acid, which inhibits protein phosphatase 2A activity, suggesting that E4-ORF4 represses E2 transcription by inducing transcription factor dephosphorylation. Interestingly, E4-ORF4 did not inhibit the transactivation capacity of a Gal4-E2F hybrid protein. Instead, E4-ORF4 expression appears to result in reduced stability of E2F/DNA complexes.


Assuntos
Proteínas E1A de Adenovirus/genética , Proteínas E2 de Adenovirus/genética , Proteínas E4 de Adenovirus/metabolismo , Proteínas de Transporte , Proteínas de Ciclo Celular , Proteínas de Drosophila , Regulação Viral da Expressão Gênica , Fases de Leitura Aberta , Regiões Promotoras Genéticas , Proteína do Retinoblastoma/metabolismo , Fatores de Transcrição/metabolismo , Proteínas E4 de Adenovirus/genética , Sítios de Ligação , Proteínas de Ligação a DNA/genética , Dimerização , Fatores de Transcrição E2F , Fator de Transcrição E2F1 , Células HeLa , Humanos , Fosforilação , RNA Mensageiro , Proteína 1 de Ligação ao Retinoblastoma , Transativadores/genética , Fator de Transcrição DP1 , Ativação Transcricional , Células Tumorais Cultivadas
14.
EMBO J ; 18(4): 1014-24, 1999 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-10022843

RESUMO

The cellular protein p32 was isolated originally as a protein tightly associated with the essential splicing factor ASF/SF2 during its purification from HeLa cells. ASF/SF2 is a member of the SR family of splicing factors, which stimulate constitutive splicing and regulate alternative RNA splicing in a positive or negative fashion, depending on where on the pre-mRNA they bind. Here we present evidence that p32 interacts with ASF/SF2 and SRp30c, another member of the SR protein family. We further show that p32 inhibits ASF/SF2 function as both a splicing enhancer and splicing repressor protein by preventing stable ASF/SF2 interaction with RNA, but p32 does not block SRp30c function. ASF/SF2 is highly phosphorylated in vivo, a modification required for stable RNA binding and protein-protein interaction during spliceosome formation, and this phosphorylation, either through HeLa nuclear extracts or through specific SR protein kinases, is inhibited by p32. Our results suggest that p32 functions as an ASF/SF2 inhibitory factor, regulating ASF/SF2 RNA binding and phosphorylation. These findings place p32 into a new group of proteins that control RNA splicing by sequestering an essential RNA splicing factor into an inhibitory complex.


Assuntos
Receptores de Hialuronatos , Proteínas Nucleares/genética , Proteínas/genética , Splicing de RNA/genética , Proteínas de Ligação a RNA/genética , Proteínas de Transporte , Citoplasma/genética , Regulação da Expressão Gênica/genética , Globinas/genética , Células HeLa , Humanos , Proteínas Mitocondriais , Fosfoproteínas/genética , Fosforilação , Proteínas Recombinantes/genética , Fatores de Processamento de Serina-Arginina , Transfecção/genética
15.
J Virol ; 72(10): 8358-61, 1998 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9733884

RESUMO

Two new adenovirus vector systems based on the tetracycline-regulated Tet-ON- (Gossen, M., et al., Science 268:1766-1769, 1995) and the RU 486-regulated progesterone antagonist (Wang, Y., et al., Proc. Natl. Acad. Sci. USA 91:8180-8184, 1994)-induced gene expression systems are described. We show that both systems permit a tight control of chloramphenicol acetyltransferase reporter gene expression in a variety of cell types, with induction levels of approximately 1,800-fold (Tet-ON system) and 600-fold (RU 486-regulated system), respectively. A significant advantage of our vector systems is that reporter protein expression can be adjusted over a wide range by varying the amount of inducer. The Tet-ON system is also shown to permit an efficient control of reporter gene expression in mice.


Assuntos
Adenoviridae/genética , Regulação Viral da Expressão Gênica/efeitos dos fármacos , Técnicas de Transferência de Genes , Vetores Genéticos , Células 3T3 , Animais , Células COS , Cloranfenicol O-Acetiltransferase/genética , Células HeLa , Antagonistas de Hormônios/farmacologia , Humanos , Camundongos , Mifepristona/farmacologia , Tetraciclina/farmacologia
16.
J Virol ; 72(7): 5978-83, 1998 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9621060

RESUMO

We have previously shown that the nonconserved carboxy-terminal exon of the adenovirus type 2 E1A-289R protein contains two interchangeable sequence elements, auxiliary region (AR) 1 and AR2, that are required for efficient CR3-mediated transcriptional activation of the viral E4 promoter (M. Bondesson, C. Svensson, S. Linder, and G. Akusjärvi, EMBO J. 11:3347-3354, 1992). Here we show that CR3-mediated transactivation of all adenovirus early promoters and the HSP70 promoter requires the AR1 element. We further show that AR2 can substitute for AR1 only when artificially juxtaposed to CR3. AR1 consists of six tandem glutamic acid-proline (EP) repeats and is positioned immediately downstream of CR3. Genetic dissection of AR1 showed that the number of EP repeats in AR1 is critical for CR3 function. Thus, reducing or increasing the number of EP repeats reduces the CR3 transactivation capacity. Furthermore, the introduction of amino acid substitutions into AR1 suggested that the net negative charge in AR1 is of critical importance for its function as an enhancer of CR3-mediated transcriptional activation. Using an in vitro binding approach, we showed that the AR1 element is not part of the CR3 promoter localization signal mediating contact with the Sp1, ATF-2, or c-Jun upstream-binding transcription factors. Previous studies have suggested that the 49-amino-acid sequence constituting CR3 represents the minimal domain required for E1A-induced activation of viral early promoters. Since AR1 was required for efficient CR3-mediated transcriptional activation of all tested promoters, we suggest that the carboxy-terminal boundary for the CR3 transactivation domain should be extended to include the AR1 element.


Assuntos
Proteínas E1A de Adenovirus/química , Ativação Transcricional , Adenoviridae/genética , Proteínas E1A de Adenovirus/fisiologia , Sequência de Aminoácidos , Proteínas de Choque Térmico HSP70/genética , Células HeLa , Humanos , Dados de Sequência Molecular , Regiões Promotoras Genéticas
17.
Nature ; 393(6681): 185-7, 1998 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-9603524

RESUMO

SR proteins are a family of essential splicing factors required for early recognition of splice sites during spliceosome assembly. They also function as alternative RNA splicing factors when overexpressed in vivo or added in excess to extracts in vitro. SR proteins are highly phosphorylated in vivo, a modification that is required for their function in spliceosome assembly and splicing catalysis. Here we show that SR proteins purified from late adenovirus-infected cells are inactivated as splicing enhancer or splicing repressor proteins by virus-induced dephosphorylation. We further show that the virus-encoded protein E4-ORF4 activates dephosphorylation by protein phosphatase 2A of HeLa SR proteins and converts their splicing properties into that of SR proteins purified from late adenovirus-infected cells. Taken together, our results suggest that E4-ORF4 is an important factor controlling the temporal shift in adenovirus alternative RNA splicing. We conclude that alternative pre-mRNA splicing, like many other biological processes, is regulated by reversible protein phosphorylation.


Assuntos
Adenoviridae/genética , Proteínas E4 de Adenovirus/genética , Splicing de RNA , Proteínas de Ligação a RNA/metabolismo , Proteínas Virais/metabolismo , Escherichia coli , Células HeLa , Humanos , Fases de Leitura Aberta , Fosforilação , Transfecção , Proteínas Virais/genética
18.
FEBS Lett ; 414(1): 111-6, 1997 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-9305742

RESUMO

p300 and CBP are two related transcriptional co-activator proteins required by many cellular transcription factors for activity. The adenovirus E1A protein binds p300 and CBP through its amino-terminus and conserved region (CR) 1. Fusing CR1 to a heterologous DNA-binding domain creates a potent transcriptional activator, suggesting that CR1 might activate transcription by recruiting p300/CBP to the promoter. We show that both p300 and CBP enhances CR1-dependent transactivation. However, this enhancement occurs independently of a direct interaction with E1A and does not correlate with the CR1 activator function.


Assuntos
Proteínas E1A de Adenovirus/metabolismo , Proteínas Nucleares/metabolismo , Transativadores , Fatores de Transcrição/metabolismo , Ativação Transcricional/fisiologia , Proteína de Ligação a CREB , Cloranfenicol O-Acetiltransferase/genética , Cloranfenicol O-Acetiltransferase/metabolismo , Escherichia coli/genética , Expressão Gênica , Genes Reporter , Células HeLa , Humanos , Mutação , Proteínas Nucleares/química , Proteínas Nucleares/genética , Ligação Proteica , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/metabolismo , Fatores de Transcrição/química , Fatores de Transcrição/genética , Transcrição Gênica , Transfecção
19.
Nature ; 381(6582): 535-8, 1996 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-8632829

RESUMO

The adenovirus L1 unit represents an example of an alternatively spliced precursor messenger (pre-mRNA) where on 5' splice can be jointed to one of two alternative 3' splice sites, producing the 52,55K or the IIIa mRNAs (Fig. 1a). Efficient usage of the distal IIIa 3' splice site requires late viral protein synthesis and is therefore confined to the late phase of virus infection. Here we show that, in extracts from uninfected cells, the classical SR proteins, which are essential splicing factors, inhibit IIIa pre-mRNA splicing by binding to an intronic repressor element and preventing recruitment of the U2 small nuclear ribonucleoprotein particle to the spliceosome. We further show that the viral repressor element has splicing-enhancer activity when appropriately placed in the pre-mRNA. Together, our results demonstrate that SR proteins function as activators or repressors of splicing depending on where on the pre-mRNA they bind.


Assuntos
Adenoviridae/genética , Precursores de RNA/metabolismo , Splicing de RNA , RNA Viral/metabolismo , Proteínas de Ligação a RNA/metabolismo , Sequências Reguladoras de Ácido Nucleico , Processamento Alternativo , Animais , Sequência de Bases , Bovinos , DNA/genética , Células HeLa , Humanos , Dados de Sequência Molecular , Splicing de RNA/genética , Coelhos , Spliceossomos/metabolismo
20.
J Virol ; 70(6): 3844-51, 1996 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-8648720

RESUMO

Here we show that the adenovirus early region 4 (E4) open reading frame 4 (ORF4) protein autoregulates its own transcription by inhibiting adenovirus E1A-induced activation of E4 transcription both in transient transfection experiments and during lytic virus growth. The inhibitory activity of E4-ORF4 was selective for E1A-CR3-dependent transactivation and had no effect on CR1 transactivation. The inhibitory activity of E4-ORF4 was relieved by okadaic acid treatment, which inhibits the cellular protein phosphatase 2A (PP2A), suggesting that E4-ORF4 controls the phosphorylated status of transcription factors important for E4 promoter activity. This conclusion agrees with previous demonstrations that E4-ORF4 associates with PP2A and causes a partial dephosphorylation of certain transcription factors, including E1A (U. Müller, T. Kleinberger, and T. Shenk, J. Virol. 66:5869-5878, 1992; T. Kleinberger and T. Shenk, J. Virol. 67:7556-7560, 1993). However, our results indicate that dephosphorylation of E1A itself might not be the primary target for E4-ORF4. Instead, the E4-ORF4-PP2A complex appears to work by dephosphorylation of multiple cellular transcription factors that are involved in E1A transactivation of the E4 promoter.


Assuntos
Adenoviridae/genética , Proteínas E1A de Adenovirus/fisiologia , Regulação Viral da Expressão Gênica , Fases de Leitura Aberta , Regiões Promotoras Genéticas , Ativação Transcricional , Sequência de Bases , Éteres Cíclicos/farmacologia , Homeostase , Dados de Sequência Molecular , Ácido Okadáico , Fosforilação , RNA Mensageiro/análise , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA