Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Artigo em Inglês | MEDLINE | ID: mdl-17381320

RESUMO

Some gammaherpesviruses encode nuclear noncoding RNAs (ncRNAs) that assemble with host proteins. Their conservation and abundance implies that they serve important functions for the virus. This paper focuses on our studies of three classes of nuclear noncoding herpesvirus RNAs. (1) EBERs 1 and 2 are expressed by Epstein-Barr virus in latent infection of human B lymphocytes. Recent studies revealed three sites on EBER1 that associate with ribosomal protein L22. In addition, heterokaryon assays have definitively shown that both EBERs are confined to the nucleus, arguing that their contribution to viral latency is purely nuclear. (2) HSURs 1-7 are U RNAs encoded by Herpesvirus saimiri, which causes aggressive T-cell leukemias and lymphomas. Comparison of monkey T cells transformed with wild-type or mutant virus lacking HSURs 1 and 2 revealed significant changes in host mRNAs implicated in T-cell signaling. (3) PAN is a 1-kb polyadenylated RNA that accumulates in the nucleus of Kaposi's sarcoma-associated herpesvirus lytically infected cells. A novel element, the ENE, is essential for its high accumulation. Recent results indicate that the ENE functions to counteract poly(A)-dependent RNA degradation, which we propose contributes to nuclear surveillance of mRNA transcripts in mammalian cells. Continuing studies of these viral RNAs will provide insights into both cellular and viral gene expression.


Assuntos
RNA Viral/genética , RNA Viral/metabolismo , Ribonucleoproteínas Nucleares Pequenas/genética , Ribonucleoproteínas Nucleares Pequenas/metabolismo , Animais , Linfócitos B/virologia , Sequência de Bases , Herpesvirus Saimiriíneo 2/genética , Herpesvirus Saimiriíneo 2/metabolismo , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/metabolismo , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/metabolismo , Humanos , Dados de Sequência Molecular , Conformação de Ácido Nucleico , RNA Viral/química , Ribonucleoproteínas Nucleares Pequenas/química
2.
Science ; 294(5548): 1895-901, 2001 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-11729309

RESUMO

The transport of messenger RNAs (mRNAs) from the nucleus to the cytoplasm involves adapter proteins that bind the mRNA as well as receptor proteins that interact with the nuclear pore complex. We demonstrate the utility of cell-permeable peptides designed to interfere with interactions between potential adapter and receptor proteins to define the pathways accessed by particular mRNAs. We show that HuR, a protein implicated in the stabilization of short-lived mRNAs containing AU-rich elements (AREs), serves as an adapter for c-fos mRNA export through two pathways. One involves the HuR shuttling domain, HNS, which exhibits a heat shock-sensitive interaction with transportin 2 (Trn2); the other involves two protein ligands of HuR-pp32 and APRIL-which contain leucine-rich nuclear export signals (NES) recognized by the export receptor CRM1. Heterokaryon and in situ hybridization experiments reveal that the peptides selectively block the nucleocytoplasmic shuttling of their respective adapter proteins without perturbing the overall cellular distribution of polyadenylated mRNAs.


Assuntos
Antígenos de Superfície , Permeabilidade da Membrana Celular , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Genes fos/genética , Fragmentos de Peptídeos/metabolismo , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/metabolismo , Receptores Citoplasmáticos e Nucleares , Fatores de Transcrição , Sequência de Aminoácidos , Animais , Proteína do Homeodomínio de Antennapedia , Transporte Biológico/efeitos dos fármacos , Linhagem Celular , Núcleo Celular/efeitos dos fármacos , Citoplasma/efeitos dos fármacos , Proteínas ELAV , Proteína Semelhante a ELAV 1 , Resposta ao Choque Térmico , Proteínas de Homeodomínio/química , Proteínas de Homeodomínio/metabolismo , Humanos , Carioferinas/metabolismo , Dados de Sequência Molecular , Neuropeptídeos/química , Neuropeptídeos/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Fragmentos de Peptídeos/farmacologia , Fosfoproteínas/metabolismo , Ligação Proteica/efeitos dos fármacos , Estrutura Terciária de Proteína , Estabilidade de RNA , RNA Mensageiro/genética , Proteínas de Ligação a RNA/antagonistas & inibidores , Proteínas de Ligação a RNA/química , Sequências Reguladoras de Ácido Nucleico/genética , Reprodutibilidade dos Testes , Tetra-Hidrofolato Desidrogenase/genética , Proteína Exportina 1
3.
RNA ; 7(9): 1348-61, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11565755

RESUMO

AU-rich elements (AREs) located in the 3' UTRs of the messenger RNAs (mRNAs) of many mammalian early response genes promote rapid mRNA turnover. HuR, an RRM-containing RNA-binding protein, specifically interacts with AREs, stabilizing these mRNAs. HuR is primarily nucleoplasmic, but shuttles between the nucleus and the cytoplasm via a domain called HNS located between RRM2 and RRM3. We recently showed that HuR interacts with two protein ligands, pp32 and APRIL, which are also shuttling proteins, but rely on NES domains recognized by CRM1 for export. Here we show that heat shock induces increased association of HuR with pp32 and APRIL through protein-protein interactions and that these ligands partially colocalize with HuR in cytoplasmic foci. HuR associations with the hnRNP complex also increase, but through RNA links. CRM1 coimmunoprecipitates with HuR only after heat shock, and nuclear export of HuR becomes sensitive to leptomycin B, an inhibitor of CRM1. Export after heat shock requires the same domains of HuR (HNS and RRM3) that are essential for binding pp32 and APRIL. In situ hybridization and coimmunoprecipitation experiments show that LMB treatment blocks both hsp70 mRNA nuclear export and its cytoplasmic interaction with HuR after heat shock. Together, our results argue that upon heat shock, HuR switches its export pathway to that of its ligands pp32 and APRIL, which involves the nuclear export factor CRM1. HuR and its ligands may be instrumental in the nuclear export of heat-shock mRNAs.


Assuntos
Antígenos de Superfície , Proteínas de Transporte/metabolismo , Resposta ao Choque Térmico , Carioferinas , Proteínas de Ligação a RNA/metabolismo , Receptores Citoplasmáticos e Nucleares , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Citoplasma/metabolismo , Proteínas ELAV , Proteína Semelhante a ELAV 1 , Ácidos Graxos Insaturados/farmacologia , Células HeLa , Humanos , Ligantes , Neuropeptídeos/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , RNA Mensageiro/metabolismo , Proteína Exportina 1
4.
Cell Mol Life Sci ; 58(2): 266-77, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11289308

RESUMO

An important mechanism of posttranscriptional gene regulation in mammalian cells is the rapid degradation of messenger RNAs (mRNAs) signaled by AU-rich elements (AREs) in their 3' untranslated regions. HuR, a ubiquitously expressed member of the Hu family of RNA-binding proteins related to Drosophila ELAV, selectively binds AREs and stabilizes ARE-containing mRNAs when overexpressed in cultured cells. This review discusses mRNA decay as a general form of gene regulation, decay signaled by AREs, and the role of HuR and its Hu-family relatives in antagonizing this mRNA degradation pathway. The influence of newly identified protein ligands to HuR on HuR function in both normal and stressed cells may explain how ARE-mediated mRNA decay is regulated in response to environmental change.


Assuntos
Antígenos de Superfície , Estabilidade de RNA , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Transporte Biológico Ativo , Proteínas de Transporte/metabolismo , Diferenciação Celular , Citoplasma/metabolismo , Proteínas ELAV , Proteína Semelhante a ELAV 1 , Humanos , Modelos Biológicos , Dados de Sequência Molecular , RNA Mensageiro/genética , Proteínas de Ligação a RNA/genética , Homologia de Sequência de Aminoácidos
5.
Genes Dev ; 15(6): 774-88, 2001 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-11274061

RESUMO

An in vitro system that recapitulates the in vivo effect of AU-rich elements (AREs) on mRNA deadenylation has been developed from Xenopus activated egg extracts. ARE-mediated deadenylation is uncoupled from mRNA body decay, and the rate of deadenylation increases with the number of tandem AUUUAs. A novel ARE-binding protein called ePAB (for embryonic poly(A)-binding protein) has been purified from this extract by ARE affinity selection. ePAB exhibits 72% identity to mammalian and Xenopus PABP1 and is the predominant poly(A)-binding protein expressed in the stage VI oocyte and during Xenopus early development. Immunodepletion of ePAB increases the rate of both ARE-mediated and default deadenylation in vitro. In contrast, addition of even a small excess of ePAB inhibits deadenylation, demonstrating that the ePAB concentration is critical for determining the rate of ARE-mediated deadenylation. These data argue that ePAB is the poly(A)-binding protein responsible for stabilization of poly(A) tails and is thus a potential regulator of mRNA deadenylation and translation during early development.


Assuntos
Poli A/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/fisiologia , Proteínas de Xenopus , Xenopus/genética , Monofosfato de Adenosina/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Western Blotting , Metilação de DNA , Regulação da Expressão Gênica no Desenvolvimento , Cinética , Dados de Sequência Molecular , Oócitos/metabolismo , Plasmídeos/metabolismo , Proteínas de Ligação a Poli(A) , Testes de Precipitina , Ligação Proteica , Proteínas de Ligação a RNA/metabolismo , Proteínas Recombinantes/metabolismo , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos , Fatores de Tempo , Ativação Transcricional , Raios Ultravioleta
6.
J Cell Biol ; 152(6): 1279-88, 2001 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-11257127

RESUMO

U2 small nuclear (sn)RNA contains a large number of posttranscriptionally modified nucleotides, including a 5' trimethylated guanosine cap, 13 pseudouridines, and 10 2'-O-methylated residues. Using Xenopus oocytes, we demonstrated previously that at least some of these modified nucleotides are essential for biogenesis of a functional snRNP. Here we address the subcellular site of U2 internal modification. Upon injection into the cytoplasm of oocytes, G-capped U2 that is transported to the nucleus becomes modified, whereas A-capped U2 that remains in the cytoplasm is not modified. Furthermore, by injecting U2 RNA into isolated nuclei or enucleated oocytes, we observe that U2 internal modifications occur exclusively in the nucleus. Analysis of the intranuclear localization of fluorescently labeled RNAs shows that injected wild-type U2 becomes localized to nucleoli and Cajal bodies. Both internal modification and nucleolar localization of U2 are dependent on the Sm binding site. An Sm-mutant U2 is targeted only to Cajal bodies. The Sm binding site can be replaced by a nucleolar localization signal derived from small nucleolar RNAs (the box C/D motif), resulting in rescue of internal modification as well as nucleolar localization. Analysis of additional chimeric U2 RNAs reveals a correlation between internal modification and nucleolar localization. Together, our results suggest that U2 internal modification occurs within the nucleolus.


Assuntos
Autoantígenos/metabolismo , Núcleo Celular/metabolismo , Oócitos/fisiologia , Processamento Pós-Transcricional do RNA , RNA Nuclear Pequeno/metabolismo , Ribonucleoproteínas Nucleares Pequenas , Transporte Ativo do Núcleo Celular , Animais , Autoantígenos/genética , Autorradiografia , Microinjeções , Conformação de Ácido Nucleico , Oócitos/citologia , Ligação Proteica , RNA Nuclear Pequeno/química , RNA Nuclear Pequeno/genética , Xenopus laevis , Proteínas Centrais de snRNP
7.
J Cell Biol ; 151(1): 1-14, 2000 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-11018049

RESUMO

AU-rich elements (AREs) present in the 3' untranslated regions of many protooncogene, cytokine, and lymphokine messages target them for rapid degradation. HuR, a ubiquitously expressed member of the ELAV (embryonic lethal abnormal vision) family of RNA binding proteins, selectively binds AREs and stabilizes ARE-containing mRNAs in transiently transfected cells. Here, we identify four mammalian proteins that bind regions of HuR known to be essential for its ability to shuttle between the nucleus and the cytoplasm and to stabilize mRNA: SETalpha, SETbeta, pp32, and acidic protein rich in leucine (APRIL). Three have been reported to be protein phosphatase 2A inhibitors. All four ligands contain long, acidic COOH-terminal tails, while pp32 and APRIL share a second motif: rev-like leucine-rich repeats in their NH(2)-terminal regions. We show that pp32 and APRIL are nucleocytoplasmic shuttling proteins that interact with the nuclear export factor CRM1 (chromosomal region maintenance protein 1). The inhibition of CRM1 by leptomycin B leads to the nuclear retention of pp32 and APRIL, their increased association with HuR, and an increase in HuR's association with nuclear poly(A)+ RNA. Furthermore, transcripts from the ARE-containing c-fos gene are selectively retained in the nucleus, while the cytoplasmic distribution of total poly(A)+ RNA is not altered. These data provide evidence that interaction of its ligands with HuR modulate HuR's ability to bind its target mRNAs in vivo and suggest that CRM1 is instrumental in the export of at least some cellular mRNAs under certain conditions. We discuss the possible role of these ligands upstream of HuR in pathways that govern the stability of ARE-containing mRNAs.


Assuntos
Antígenos de Superfície , Carioferinas , Estabilidade de RNA , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/metabolismo , Receptores Citoplasmáticos e Nucleares , Transporte Ativo do Núcleo Celular , Sequência de Aminoácidos , Sítios de Ligação , Proteínas de Transporte/metabolismo , Cromatografia de Afinidade , Citoplasma/metabolismo , Proteínas ELAV , Proteína Semelhante a ELAV 1 , Ácidos Graxos Insaturados/farmacologia , Células HeLa , Humanos , Ligantes , Dados de Sequência Molecular , Neuropeptídeos/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas Fosfatases/antagonistas & inibidores , Fosfoproteínas/metabolismo , Ligação Proteica , Proteína Fosfatase 2 , Transporte Proteico , Análise de Sequência de Proteína , Proteína Exportina 1
8.
Proc Natl Acad Sci U S A ; 97(7): 3073-8, 2000 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-10737787

RESUMO

AU-rich elements (AREs) located in the 3' untranslated region target the mRNAs encoding many protooncoproteins, cytokines, and lymphokines for rapid degradation. HuR, a ubiquitously expressed member of the embryonic lethal abnormal vision (ELAV) family of RNA-binding proteins, binds ARE sequences and selectively stabilizes ARE-containing reporter mRNAs when overexpressed in transiently transfected cells. HuR appears predominantly nucleoplasmic but has been shown to shuttle between the nucleus and cytoplasm via a novel shuttling sequence HNS. We report generation of a mouse monoclonal antibody 3A2 that both immunoblots and immunoprecipitates HuR protein; it recognizes an epitope located in the first of HuR's three RNA recognition motifs. This antibody was used to probe HuR interactions with mRNA before and after heat shock, a condition that has been reported to stabilize ARE-containing mRNAs. At 37 degrees C, approximately one-third of the cytoplasmic HuR appears polysome associated, and in vivo UV crosslinking reveals that HuR interactions with poly(A)(+) RNA are predominantly cytoplasmic rather than nuclear. This comprises evidence that HuR directly interacts with mRNA in vivo. After heat shock, 12-15% of HuR accumulates in discrete foci in the cytoplasm, but surprisingly the majority of HuR crosslinks instead to nuclear poly(A)(+) RNA, whose levels are dramatically increased in the stressed cells. This behavior of HuR differs from that of another ARE-binding protein, hnRNP D, which has been implicated as an effector of mRNA decay rather than mRNA stabilization and of the general pre-RNA-binding protein hnRNP A1. We interpret these differences to mean that the temporal association of HuR with ARE-containing mRNAs is different from that of these other two proteins.


Assuntos
Antígenos de Superfície , Temperatura Alta , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/metabolismo , Anticorpos Monoclonais/imunologia , Especificidade de Anticorpos , Sequência de Bases , Citoplasma/metabolismo , Primers do DNA , Proteínas ELAV , Proteína Semelhante a ELAV 1 , Células HeLa , Humanos , Ligação Proteica , Proteínas de Ligação a RNA/imunologia
9.
Genes Dev ; 13(7): 851-63, 1999 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-10197985

RESUMO

We have investigated the formation of prespliceosomal complex A in HeLa nuclear extracts on a splicing substrate containing an AT-AC (U12-type) intron from the P120 gene. Using an RNase H protection assay and specific blocking oligonucleotides, we find that recognition of the 5' splice-site (5'ss) and branchpoint sequence (BPS) elements by U11 and U12 snRNPs, respectively, displays strong cooperativity, requiring both sites in the pre-mRNA substrate for efficient complex formation. Deletion analysis indicates that beside the 5'ss and BPS, no additional elements in the pre-mRNA are necessary for A-complex formation, although 5' exon sequences provide stimulation. Cross-linking studies with pre-mRNAs containing the 5'ss or BPS alone indicate that recognition of the BPS by the U12 snRNP is stimulated at least 20- to 30-fold by the binding of the U11 snRNP to the 5'ss in the same pre-mRNA molecule, whereas recognition of the 5'ss by U11 is stimulated approximately fivefold by the U12/BPS interaction. These results argue that intron recognition in the U12-dependent splicing pathway is carried out by a single U11/U12 di-snRNP complex, suggesting greater rigidity in the intron recognition process than in the major spliceosome.


Assuntos
Splicing de RNA/fisiologia , Ribonucleoproteínas Nucleares Pequenas/metabolismo , Adenoviridae/genética , Northern Blotting , Relação Dose-Resposta a Droga , Evolução Molecular , Ficusina/metabolismo , Células HeLa , Ribonucleoproteínas Nucleares Heterogêneas , Humanos , Íntrons , Modelos Genéticos , Oligonucleotídeos/metabolismo , Ribonuclease H/metabolismo , Ribonucleoproteína Nuclear Pequena U4-U6/metabolismo , Ribonucleoproteínas/metabolismo , Fatores de Tempo
10.
Proc Natl Acad Sci U S A ; 95(26): 15293-8, 1998 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-9860962

RESUMO

Proteins are transported into and out of the cell nucleus via specific signals. The two best-studied nuclear transport processes are mediated either by classical nuclear localization signals or nuclear export signals. There also are shuttling sequences that direct the bidirectional transport of RNA-binding proteins. Two examples are the M9 sequence in heterogeneous nuclear ribonucleoprotein A1 and the heterogeneous nuclear ribonucleoprotein K shuttling domain (KNS) sequence in heterogeneous nuclear ribonucleoprotein K, both of which appear to contribute importantly to the export of mRNA to the cytoplasm. HuR is an RNA-binding protein that can stabilize labile mRNAs containing AU-rich elements in their 3' untranslated regions and has been shown to shuttle between the nucleus and cytoplasm (18, 19). We have identified in HuR a shuttling sequence that also possess transcription-dependent nuclear localization signal activity. We propose that HuR first may bind AU-rich element-containing mRNAs in the nucleus and then escort them through the nuclear pore, providing protection during and after export to the cytoplasmic compartment.


Assuntos
Antígenos de Superfície , Proteínas de Ligação a RNA/química , Proteínas de Ligação a RNA/metabolismo , Células 3T3 , Sequência de Aminoácidos , Animais , Núcleo Celular/metabolismo , Sequência Conservada , Citoplasma/metabolismo , Proteínas ELAV , Proteína Semelhante a ELAV 1 , Células HeLa , Ribonucleoproteínas Nucleares Heterogêneas Grupo K , Humanos , Camundongos , Dados de Sequência Molecular , RNA Mensageiro/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Ribonucleoproteínas/química , Ribonucleoproteínas/metabolismo , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Transfecção , Xenopus
11.
Mol Cell Biol ; 18(12): 6897-909, 1998 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9819378

RESUMO

We have identified gas5 (growth arrest-specific transcript 5) as a non-protein-coding multiple small nucleolar RNA (snoRNA) host gene similar to UHG (U22 host gene). Encoded within the 11 introns of the mouse gas5 gene are nine (10 in human) box C/D snoRNAs predicted to function in the 2'-O-methylation of rRNA. The only regions of conservation between mouse and human gas5 genes are their snoRNAs and 5'-end sequences. Mapping the 5' end of the mouse gas5 transcript demonstrates that it possesses an oligopyrimidine tract characteristic of the 5'-terminal oligopyrimidine (5'TOP) class of genes. Arrest of cell growth or inhibition of translation by cycloheximide, pactamycin, or rapamycin-which specifically inhibits the translation of 5'TOP mRNAs-results in accumulation of the gas5 spliced RNA. Classification of gas5 as a 5'TOP gene provides an explanation for why it is a growth arrest specific transcript: while the spliced gas5 RNA is normally associated with ribosomes and rapidly degraded, during arrested cell growth it accumulates in mRNP particles, as has been reported for other 5'TOP messages. Strikingly, inspection of the 5'-end sequences of currently known snoRNA host gene transcripts reveals that they all exhibit features of the 5'TOP gene family.


Assuntos
Nucléolo Celular/genética , Proteínas de Membrana/genética , Família Multigênica/genética , RNA Nuclear Pequeno/genética , RNA Nucleolar Pequeno , Células 3T3 , Animais , Sequência de Bases , Divisão Celular/genética , Clonagem Molecular , Humanos , Camundongos , Dados de Sequência Molecular , Biossíntese de Proteínas/genética , Splicing de RNA/genética , RNA Antissenso/genética , RNA Mensageiro/genética , Ribonucleoproteínas/genética , Ribossomos/genética , Transcrição Gênica/genética
12.
Mol Cell Biol ; 18(12): 7537-45, 1998 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9819439

RESUMO

To study the regulation of AUUUA-mediated RNA deadenylation and destabilization during Xenopus early development, we microinjected chimeric mRNAs containing Xenopus or mammalian 3' untranslated region (3'-UTR) sequences into Xenopus oocytes, mature eggs, or fertilized embryos. We found that the AU-rich elements (ARE) of Xenopus c-myc II and the human granulocyte-macrophage colony-stimulating factor gene (GMCSF) both direct deadenylation of chimeric mRNAs in an AUUUA-dependent manner. In the case of the Xenopus c-myc II ARE, mutation of a single AUUUA within an absolutely conserved 11-nucleotide region in c-myc 3'-UTRs prevents ARE-mediated deadenylation. AUUUA-specific deadenylation appears to be developmentally regulated: low deadenylation activity is observed in the oocyte, whereas rapid deadenylation occurs following egg activation or fertilization. Deadenylation results in the accumulation of stable deadenylated RNAs that become degraded only following mid-blastula transition. We conclude that ARE-mediated mRNA deadenylation can be uncoupled from ARE-mediated mRNA decay and that AUUUAs directly signal deadenylation during Xenopus early development.


Assuntos
Processamento Pós-Transcricional do RNA/genética , RNA Mensageiro/genética , Xenopus/crescimento & desenvolvimento , Regiões 3' não Traduzidas/genética , Animais , Sequência Conservada/genética , Fertilização/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Microinjeções , Oligodesoxirribonucleotídeos/genética , Oócitos/metabolismo , Poli A/genética , Xenopus/embriologia
13.
EMBO J ; 17(12): 3448-60, 1998 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-9628880

RESUMO

The messenger RNAs of many proto-oncogenes, cytokines and lymphokines are targeted for rapid degradation through AU-rich elements (AREs) located in their 3' untranslated regions (UTRs). HuR, a ubiquitously expressed member of the Elav family of RNA binding proteins, exhibits specific affinities for ARE-containing RNA sequences in vitro which correlate with their in vivo decay rates, thereby implicating HuR in the ARE-mediated degradation pathway. We have transiently transfected HuR into mouse L929 cells and observed that overexpression of HuR enhances the stability of beta-globin reporter mRNAs containing either class I or class II AREs. The increase in mRNA stability parallels the level of HuR overexpression, establishing an in vivo role for HuR in mRNA decay. Furthermore, overexpression of HuR deletion mutants lacking RNA recognition motif 3 (RRM 3) does not exert a stabilizing effect, indicating that RRM 3 is important for HuR function. We have also developed polyclonal anti-HuR antibodies. Immunofluorescent staining of HeLa and L929 cells using affinity-purified anti-HuR antibody shows that both endogenous and overexpressed HuR proteins are localized in the nucleus. By forming HeLa-L929 cell heterokaryons, we demonstrate that HuR shuttles between the nucleus and cytoplasm. Thus, HuR may initially bind to ARE-containing mRNAs in the nucleus and provide protection during and after their export to the cytoplasmic compartment.


Assuntos
Antígenos de Superfície , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/metabolismo , Animais , Núcleo Celular/metabolismo , Células Cultivadas , Citoplasma/metabolismo , Proteínas ELAV , Proteína Semelhante a ELAV 1 , Células HeLa , Humanos , Camundongos
14.
RNA ; 3(11): 1301-12, 1997 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9409621

RESUMO

Four peptides are shown to block mammalian spliceosome assembly and pre-mRNA splicing in vitro. Previously, these peptides have been shown to inhibit Ca2+-dependent calmodulin kinase II (CaMK II) via distinct mechanisms. One is a competitive inhibitor of the kinase, two interfere with autophosphorylation events, and one competes for binding to calmodulin, a CaMK II-activating protein. However, because EGTA does not inhibit splicing, the involvement of CaMK II itself in splicing is unlikely; rather, a protein similar to CaMK II may be involved in spliceosome assembly and splicing. Two of the inhibitory peptides, the calmodulin binding domain (CBD) and glycogen synthase (GS) fragment, block assembly of spliceosomal complex C. These peptides inhibited splicing if they were added to reactions any time within the first 10 min of splicing assays. No inhibition of spliceosome assembly or splicing occurred in the presence of randomized versions of the CBD or GS peptide. Additionally, the GS peptide inhibited splicing when added to assays at later time points, despite the fact that spliceosomal complex C had formed. Cumulatively, these analyses suggest that the peptides inhibit at least two distinct events in the spliceosomal cycle. The first event occurs early during in vitro splicing. For this event, prolonged incubations of splicing reactions do not result in a recovery of splicing activity. The second event occurs later and represents a slowing of an essential step, because splicing activity can be recovered in prolonged incubations. Peptides known to inhibit protein kinase A and protein kinase C had no effect on pre-mRNA splicing, underscoring the specificity of the observed inhibitory effects.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Peptídeos/farmacologia , Precursores de RNA/metabolismo , Splicing de RNA/efeitos dos fármacos , Spliceossomos/efeitos dos fármacos , Sequência de Aminoácidos , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Inibidores Enzimáticos/metabolismo , Células HeLa , Humanos , Dados de Sequência Molecular , Peptídeos/química , Peptídeos/metabolismo , Especificidade por Substrato
15.
Genes Dev ; 11(19): 2557-68, 1997 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-9334320

RESUMO

AU-rich elements (AREs, usually containing repeated copies of AUUUA), when present in the 3'-untranslated regions (UTRs) of many mammalian mRNAs, confer instability on their host RNA molecules. The viral small nuclear RNA (snRNA) Herpesvirus saimiri U RNA 1 (HSUR 1) also contains an AUUUA-rich sequence. Here, we report that this ARE induces rapid degradation of HSUR 1 itself and of other snRNAs including HSUR 2 and cellular U1. Mutational analyses of the viral ARE establish that sequence requirements for mRNA and snRNA decay are the same, suggesting a similar mechanism. Moreover, the in vivo degradation activity of mutant AREs correlates with their in vitro binding to the HuR protein, implicated previously as a component of the mRNA degradation machinery. Our results suggest that ARE-mediated instability can be uncoupled from both ongoing translation and deadenylation of the target RNA.


Assuntos
Antígenos de Superfície , Herpesvirus Saimiriíneo 2/química , RNA Mensageiro/metabolismo , RNA Nuclear Pequeno/metabolismo , RNA Viral/metabolismo , Sequência de Bases , Proteínas ELAV , Proteína Semelhante a ELAV 1 , Regulação da Expressão Gênica/genética , Genes Reporter , Globinas/genética , Herpesvirus Saimiriíneo 2/genética , Dados de Sequência Molecular , Mutação , RNA Mensageiro/genética , RNA Nuclear Pequeno/genética , RNA Viral/genética , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Sequências Repetitivas de Ácido Nucleico , Ribonucleases/metabolismo , Transcrição Gênica , Transfecção
16.
EMBO J ; 16(8): 2130-9, 1997 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-9155038

RESUMO

Expression of many proto-oncogenes, cytokines and lymphokines is regulated by targeting their messenger RNAs for rapid degradation. Essential signals for this control are AU-rich elements (AREs) in the 3' untranslated region (UTR) of these messages. The ARE is loosely defined as the five-nucleotide sequence AUUUA embedded in a uracil-rich region. A transacting factor, presumably a protein, binds the ARE and initiates recognition by the destabilization machinery. Numerous candidate ARE-binding proteins have been proposed. We show that a 32 kDa protein in HeLa nuclear extracts characterized previously has RNA-binding specificity that correlates with the activity of an ARE in directing mRNA decay. Purification and subsequent analyses demonstrate that this 32 kDa protein is identical to a recently identified member of the Elav-like gene family (ELG) called HuR. The in vitro binding selectivity of HuR is indicative of an ARE sequence's ability to destabilize a mRNA in vivo, suggesting a critical role for HuR in the regulation of mRNA degradation.


Assuntos
Antígenos de Superfície , Regulação da Expressão Gênica/fisiologia , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/metabolismo , Células 3T3 , Sequência de Aminoácidos , Animais , Composição de Bases , Extratos Celulares , Reagentes de Ligações Cruzadas , Proteínas ELAV , Proteína Semelhante a ELAV 1 , Células HeLa , Humanos , Camundongos , Dados de Sequência Molecular , Peso Molecular , Proteínas de Ligação a RNA/química , Proteínas de Ligação a RNA/isolamento & purificação , Sequências Reguladoras de Ácido Nucleico/genética , Raios Ultravioleta
17.
RNA ; 1(6): 644-56, 1995 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-7489523

RESUMO

A minor class of introns with noncanonical splice sites has been identified in both vertebrate and invertebrate genomes. The divergent consensus sequences within these introns suggest that splicing might be via a mechanism distinct from that used by the major class of introns. The low abundance U12 snRNA has been proposed to base pair with the predicted branch site sequence of these minor class introns, probably bulging out an adenosine to act as the nucleophile in the first step of splicing. We have identified homologues of the previously characterized human U12 snRNA in both mouse and chicken, where the minor class of introns has also been found. The U12 sequences that potentially base pair with the putative branch site are invariant. Additional conserved sequences at the 5' end of U12 snRNA could dynamically base pair with U6 snRNA sequences flanking the hexanucleotide ACAGAG to form structures analogous to those of three U2-U6 interactions genetically defined as important in the major class of spliceosome. We have also isolated two human U12 snRNA genes. One gene is functional for transcription of U12 snRNA, whereas the other appears to be a pseudogene. Sequences of the 3' box in both U12 snRNA genes are strikingly similar and bear high resemblance to those of U1 and U2 genes. Upstream elements, including the PSE and the DSE, have been identified and characterized in the functional gene. These features indicate that transcription of U12 snRNA is driven by RNA polymerase II.


Assuntos
Genoma Humano , RNA Nuclear Pequeno/análise , RNA Nuclear Pequeno/genética , Ribonucleoproteínas Nucleares Pequenas/genética , Animais , Sequência de Bases , Evolução Biológica , Galinhas , Sequência Consenso , Sequência Conservada , Primers do DNA , Humanos , Íntrons , Camundongos , Dados de Sequência Molecular , Conformação de Ácido Nucleico , Nucleotídeos/análise , Precursores de RNA , Splicing de RNA , RNA Mensageiro
18.
Proc Natl Acad Sci U S A ; 92(7): 2504-8, 1995 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-7708674

RESUMO

Ser/Arg-rich proteins (SR proteins) are essential splicing factors that commit pre-messenger RNAs to splicing and also modulate 5' splice site choice in the presence or absence of functional U1 small nuclear ribonucleoproteins (snRNPs). Here, we perturbed the U1 snRNP in HeLa cell nuclear extract by detaching the U1-specific A protein using a 2'-O-methyl oligonucleotide (L2) complementary to its binding site in U1 RNA. In this extract, the standard adenovirus substrate is spliced normally, but excess amounts of SR proteins do not exclusively switch splicing from the normal 5' splice site to a proximal site (site 125 within the adenovirus intron), suggesting that modulation of 5' splice site choice exerted by SR proteins requires integrity of the U1 snRNP. The observation that splicing does not necessarily follow U1 binding indicates that interactions between the U1 snRNP and components assembled on the 3' splice site via SR proteins may also be critical for 5' splice site selection. Accordingly, we found that SR proteins promote the binding of the U2 snRNP to the branch site and stabilize the complex formed on a 3'-half substrate in the presence or absence of functional U1 snRNPs. A novel U2/U6/3'-half substrate crosslink was also detected and promoted by SR proteins. Our results suggest that SR proteins in collaboration with the U1 snRNP function in two distinct steps to modulate 5' splice site selection.


Assuntos
Precursores de RNA/metabolismo , Splicing de RNA , Ribonucleoproteína Nuclear Pequena U1/metabolismo , Western Blotting , Núcleo Celular/metabolismo , Reagentes de Ligações Cruzadas , Furocumarinas , Células HeLa , Humanos , Oligodesoxirribonucleotídeos , Ligação Proteica , Precursores de RNA/isolamento & purificação , Ribonucleoproteína Nuclear Pequena U1/isolamento & purificação
19.
Genes Dev ; 8(22): 2704-17, 1994 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-7958927

RESUMO

SR proteins are essential splicing factors that also influence 5' splice site choice. We show that addition of excess mixed SR proteins to a HeLa in vitro splicing system stimulates utilization of a novel 5' splice site (site 125) within the intron of the standard adenovirus pre-mRNA substrate. When U1 snRNPs are debilitated by sequestering the 5' end of U1 snRNA with a 2'-O-methyl oligoribonucleotide, excess SR proteins not only rescue splicing at the normal site and site 125 but also activate yet another 5' splice site (site 47) in the adenovirus intron. One SR protein, SC35, is sufficient to exhibit the above activities. The possibility that excess SR proteins recruit residual unblocked U1 snRNPs to participate in 5' splice site recognition has been ruled out by psoralen cross-linking studies, which demonstrate that the 2'-O-methyl oligoribonucleotide effectively blocks 5' splice site/U1 interaction. Native gel analysis reveals a nearly normal splicing complex profile in the 2'-O-methyl oligoribonucleotide pretreated, SR protein-supplemented extract. These results indicate that SR proteins can replace some functions of the U1 snRNP but underscore the contribution of U1 to the fidelity of 5' splice site selection.


Assuntos
Proteínas Nucleares/fisiologia , Fosfoproteínas/fisiologia , Splicing de RNA , Ribonucleoproteína Nuclear Pequena U1/fisiologia , Ribonucleoproteína Nuclear Pequena U4-U6/fisiologia , Spliceossomos/fisiologia , Adenoviridae/genética , Processamento Alternativo , Sequência de Bases , Ficusina/farmacologia , Células HeLa , Humanos , Íntrons , Dados de Sequência Molecular , Proteínas de Ligação a RNA , Fatores de Processamento de Serina-Arginina
20.
Proc Natl Acad Sci U S A ; 91(8): 3463-7, 1994 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-8159770

RESUMO

Epstein-Barr virus (EBV), an oncogenic herpesvirus, encodes two small RNAs (EBERs) that are expressed at high levels during latent transformation of human B lymphocytes. Here we report that a 15-kDa cellular protein called EAP (for EBER associated protein), previously shown to bind EBER1, is in fact the ribosomal protein L22. Approximately half of the L22 in EBV-positive cells is contained within the EBER1 ribonucleoprotein (RNP) particle, whereas the other half residues in monoribosomes and polysomes. Immunofluorescence with anti-L22 antibodies demonstrates that L22 is localized in the cytoplasm and the nucleoli of uninfected human cells, as expected, whereas EBV-positive lymphocytes also show strong nucleoplasmic staining. In situ hybridization indicates that the EBER RNPs are predominantly nucleoplasmic, suggesting that L22 relocalization correlates with binding to EBER1 in vivo. Since incubation of uninfected cell extracts with excess EBER1 RNA does not remove L22 from preexisting ribosomes, in vivo binding of L22 by EBER1 may precede ribosome assembly. The gene encoding L22 has recently been identified as the target of a chromosomal translocation in certain patients with leukemia, suggesting that L22 levels may be a determinant in cell transformation.


Assuntos
Infecções por Herpesviridae/metabolismo , Herpesvirus Humano 4/genética , RNA Viral/metabolismo , Proteínas de Ligação a RNA/metabolismo , Proteínas Ribossômicas , Infecções Tumorais por Vírus/metabolismo , Linfócitos B/microbiologia , Sequência de Bases , Compartimento Celular , Linhagem Celular , Primers do DNA/química , Humanos , Dados de Sequência Molecular , Ribonucleoproteínas/química , Ribossomos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA