Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
EMBO J ; 20(19): 5491-502, 2001 Oct 01.
Article in English | MEDLINE | ID: mdl-11574481

ABSTRACT

mRNA silencing and storage play an important role in gene expression under diverse circumstances, such as throughout early metazoan development and in response to many types of environmental stress. Here we demonstrate that the major mRNA-associated protein YB-1, also termed p50, is a potent cap-dependent mRNA stabilizer. YB-1 addition or overexpression dramatically increases mRNA stability in vitro and in vivo, whereas YB-1 depletion results in accelerated mRNA decay. The cold shock domain of YB-1 is responsible for the mRNA stabilizing activity, and a blocked mRNA 5' end is required for YB-1-mediated stabilization. Significantly, exogenously added YB-1 destabilizes the interaction of the cap binding protein, eIF4E, with the mRNA cap structure. Conversely, sequestration of eIF4E from the cap increases the association of endogenous YB-1 with mRNA at or near the cap, and significantly enhances mRNA stability. These data support a model whereby down-regulation of eIF4E activity or increasing the YB-1 mRNA binding activity or concentration in cells activates a general default pathway for mRNA stabilization.


Subject(s)
RNA Caps/metabolism , RNA Stability , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Repressor Proteins/metabolism , Animals , Cell-Free System , Eukaryotic Initiation Factor-4E , HeLa Cells , Humans , Models, Genetic , Peptide Initiation Factors/metabolism , Protein Biosynthesis , Protein Structure, Tertiary , Rabbits , Reticulocytes
2.
Mol Cell Biol ; 21(19): 6440-9, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11533233

ABSTRACT

Ceruloplasmin (Cp) is a glycoprotein secreted by the liver and monocytic cells and probably plays roles in inflammation and iron metabolism. We showed previously that gamma interferon (IFN-gamma) induced Cp synthesis by human U937 monocytic cells but that the synthesis was subsequently halted by a transcript-specific translational silencing mechanism involving the binding of a cytosolic factor(s) to the Cp mRNA 3' untranslated region (UTR). To investigate how protein interactions at the Cp 3'-UTR inhibit translation initiation at the distant 5' end, we considered the "closed-loop" model of mRNA translation. In this model, the transcript termini are brought together by interactions of poly(A)-binding protein (PABP) with both the poly(A) tail and initiation factor eIF4G. The effect of these elements on Cp translational control was tested using chimeric reporter transcripts in rabbit reticulocyte lysates. The requirement for poly(A) was shown since the cytosolic inhibitor from IFN-gamma-treated cells minimally inhibited the translation of a luciferase reporter upstream of the Cp 3'-UTR but almost completely blocked the translation of a transcript containing a poly(A) tail. Likewise, a requirement for poly(A) was shown for silencing of endogenous Cp mRNA. We considered the possibility that the cytosolic inhibitor blocked the interaction of PABP with the poly(A) tail or with eIF4G. We found that neither of these interactions were inhibited, as shown by immunoprecipitation of PABP followed by quantitation of the poly(A) tail by reverse transcription-PCR and of eIF4G by immunoblot analysis. We considered the alternate possibility that these interactions were required for translational silencing. When PABP was depleted from the reticulocyte lysate with anti-human PABP antibody, the cytosolic factor did not inhibit translation of the chimeric reporter, thus showing the requirement for PABP. Similarly, in lysates treated with anti-human eIF4G antibody, the cytosolic extract did not inhibit the translation of the chimeric reporter, thereby showing a requirement for eIF4G. These data show that translational silencing of Cp requires interactions of three essential elements of mRNA circularization, poly(A), PABP, and eIF4G. We suggest that Cp mRNA circularization brings the cytosolic Cp 3'-UTR-binding factor into the proximity of the translation initiation site, where it silences translation by an undetermined mechanism. These results suggest that in addition to its important function in increasing the efficiency of translation, transcript circularization may serve as an essential structural determinant for transcript-specific translational control.


Subject(s)
Ceruloplasmin/genetics , Gene Silencing , Peptide Initiation Factors/physiology , Protein Biosynthesis , RNA, Messenger/chemistry , RNA-Binding Proteins/physiology , 3' Untranslated Regions , Animals , Ceruloplasmin/biosynthesis , Eukaryotic Initiation Factor-4G , Humans , Interferon-gamma/pharmacology , Models, Genetic , Poly(A)-Binding Proteins , RNA/chemistry , RNA/metabolism , RNA, Circular , RNA, Messenger/metabolism , Rabbits , U937 Cells
3.
Mol Cell Biol ; 21(15): 5200-13, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11438674

ABSTRACT

The cap structure and the poly(A) tail of eukaryotic mRNAs act synergistically to enhance translation. This effect is mediated by a direct interaction of eukaryotic initiation factor 4G and poly(A) binding protein (PABP), which brings about circularization of the mRNA. Of the two recently identified PABP-interacting proteins, one, Paip1, stimulates translation, and the other, Paip2, which competes with Paip1 for binding to PABP, represses translation. Here we studied the Paip2-PABP interaction. Biacore data and far-Western analysis revealed that Paip2 contains two binding sites for PABP, one encompassing a 16-amino-acid stretch located in the C terminus and a second encompassing a larger central region. PABP also contains two binding regions for Paip2, one located in the RNA recognition motif (RRM) region and the other in the carboxy-terminal region. A two-to-one stoichiometry for binding of Paip2 to PABP with two independent K(d)s of 0.66 and 74 nM was determined. Thus, our data demonstrate that PABP and Paip2 could form a trimeric complex containing one PABP molecule and two Paip2 molecules. Significantly, only the central Paip2 fragment, which binds with high affinity to the PABP RRM region, inhibits PABP binding to poly(A) RNA and translation.


Subject(s)
Carrier Proteins/metabolism , Protein Biosynthesis , RNA-Binding Proteins/metabolism , Amino Acid Sequence , Binding Sites , Binding, Competitive , Blotting, Western , Carrier Proteins/chemistry , Genetic Vectors , Humans , Kinetics , Models, Theoretical , Molecular Sequence Data , Mutation , Peptide Initiation Factors/metabolism , Poly(A)-Binding Proteins , Protein Binding , Protein Structure, Tertiary , RNA, Messenger/metabolism , Recombinant Proteins/metabolism , Time Factors
4.
RNA ; 7(12): 1743-52, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11780631

ABSTRACT

The eukaryotic mRNA 3' poly(A) tail and the 5' cap cooperate to synergistically enhance translation. This interaction is mediated, at least in part, by elF4G, which bridges the mRNA termini by simultaneous binding the poly(A)-binding protein (PABP) and the cap-binding protein, elF4E. The poly(A) tail also stimulates translation from the internal ribosome binding sites (IRES) of a number of picornaviruses. elF4G is likely to mediate this translational stimulation through its direct interaction with the IRES. Here, we support this hypothesis by cleaving elF4G to separate the PABP-binding site from the portion that promotes internal initiation. elF4G cleavage abrogates the stimulatory effect of poly(A) tail on translation. In addition, translation in extracts in which elF4G is cleaved is resistant to inhibition by the PABP-binding protein 2 (Paip2). The elF4G cleavage-induced loss of the stimulatory effect of poly(A) on translation was mimicked by the addition of the C-terminal portion of elF4G. Thus, PABP stimulates picornavirus translation through its interaction with elF4G.


Subject(s)
Peptide Initiation Factors/metabolism , Picornaviridae/genetics , Protein Biosynthesis , RNA-Binding Proteins/metabolism , Encephalomyocarditis virus/genetics , Eukaryotic Initiation Factor-4G , Peptide Fragments/metabolism , Poliovirus/genetics , Poly(A)-Binding Proteins , RNA Caps , RNA, Messenger/metabolism , Ribosomes/metabolism
5.
Curr Biol ; 10(18): 1147-50, 2000 Sep 21.
Article in English | MEDLINE | ID: mdl-10996799

ABSTRACT

The poly(A)-binding protein Pab1p interacts directly with the eukaryotic translation initiation factor 4G (eIF4G) to facilitate translation initiation of polyadenylated mRNAs in yeast [1,2]. Although the eIF4G-PABP interaction has also been demonstrated in a mammalian system [3,4], its biological significance in vertebrates is unknown. In Xenopus oocytes, cytoplasmic polyadenylation of several mRNAs coincides with their translational activation and is critical for maturation [5-7]. Because the amount of PABP is very low in oocytes [8], it has been argued that the eIF4G-PABP interaction does not play a major role in translational activation during oocyte maturation. Also, overexpression of PABP in Xenopus oocytes has only a modest stimulatory effect on translation of polyadenylated mRNA and does not alter either the efficiency or the kinetics of progesterone-induced maturation [9]. Here, we report that the expression of an eIF4GI mutant defective in PABP binding in Xenopus oocytes reduces translation of polyadenylated mRNA and dramatically inhibits progesterone-induced maturation. Our results show that the eIF4G-PABP interaction is critical for translational control of maternal mRNAs during Xenopus development.


Subject(s)
Oocytes/growth & development , Peptide Initiation Factors/metabolism , Protein Biosynthesis/genetics , RNA-Binding Proteins/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Blotting, Western , Eukaryotic Initiation Factor-4G , Genes, mos/genetics , Humans , Luciferases/metabolism , Microinjections , Molecular Sequence Data , Oocytes/drug effects , Peptide Initiation Factors/genetics , Poly(A)-Binding Proteins , Progesterone/pharmacology , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , Xenopus/embryology
6.
EMBO J ; 19(3): 434-44, 2000 Feb 01.
Article in English | MEDLINE | ID: mdl-10654941

ABSTRACT

The eukaryotic translation initiation factor 4G (eIF4G) proteins play a critical role in the recruitment of the translational machinery to mRNA. The eIF4Gs are phosphoproteins. However, the location of the phosphorylation sites, how phosphorylation of these proteins is modulated and the identity of the intracellular signaling pathways regulating eIF4G phosphorylation have not been established. In this report, two-dimensional phosphopeptide mapping demonstrates that the phosphorylation state of specific eIF4GI residues is altered by serum and mitogens. Phosphopeptides resolved by this method were mapped to the C-terminal one-third of the protein. Mass spectrometry and mutational analyses identified the serum-stimulated phosphorylation sites in this region as serines 1108, 1148 and 1192. Phosphoinositide-3-kinase (PI3K) inhibitors and rapamycin, an inhibitor of the kinase FRAP/mTOR (FKBP12-rapamycin-associated protein/mammalian target of rapamycin), prevent the serum-induced phosphorylation of these residues. Finally, the phosphorylation state of N-terminally truncated eIF4GI proteins acquires resistance to kinase inhibitor treatment. These data suggest that the kinases phosphorylating serines 1108, 1148 and 1192 are not directly downstream of PI3K and FRAP/mTOR, but that the accessibility of the C-terminus to kinases is modulated by this pathway(s).


Subject(s)
Peptide Initiation Factors/chemistry , Protein Kinases , Sirolimus/pharmacology , Amino Acid Sequence , Binding Sites , DNA Mutational Analysis , Electrophoresis, Gel, Two-Dimensional , Enzyme Inhibitors/pharmacology , Eukaryotic Initiation Factor-4G , Humans , Mass Spectrometry , Molecular Sequence Data , Peptide Initiation Factors/genetics , Peptide Mapping , Phosphoinositide-3 Kinase Inhibitors , Phosphopeptides/chemistry , Phosphorylation , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Biosynthesis , RNA, Messenger/metabolism , Sequence Alignment , Signal Transduction , TOR Serine-Threonine Kinases
7.
Mol Cell Biol ; 20(2): 468-77, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10611225

ABSTRACT

The mammalian eukaryotic initiation factor 4GI (eIF4GI) may be divided into three roughly equal regions; an amino-terminal one-third (amino acids [aa] 1 to 634), which contains the poly(A) binding protein (PABP) and eIF4E binding sites; a middle third (aa 635 to 1039), which binds eIF4A and eIF3; and a carboxy-terminal third (aa 1040 to 1560), which harbors a second eIF4A binding site and a docking sequence for the Ser/Thr kinase Mnk1. Previous reports demonstrated that the middle one-third of eIF4GI is sufficient for cap-independent translation. To delineate the eIF4GI core sequence required for cap-dependent translation, various truncated versions of eIF4GI were examined in an in vitro ribosome binding assay with beta-globin mRNA. A sequence of 540 aa encompassing aa 550 to 1090, which contains the eIF4E binding site and the middle region of eIF4GI, is the minimal sequence required for cap-dependent translation. In agreement with this, a point mutation in eIF4GI which abolished eIF4A binding in the middle region completely inhibited ribosomal binding. However, the eIF4GI C-terminal third region, which does not have a counterpart in yeast, modulates the activity of the core sequence. When the eIF4A binding site in the C-terminal region of eIF4GI was mutated, ribosome binding was decreased three- to fourfold. These data indicate that the interaction of eIF4A with the middle region of eIF4GI is necessary for translation, whereas the interaction of eIF4A with the C-terminal region plays a modulatory role.


Subject(s)
Peptide Fragments/chemistry , Peptide Fragments/metabolism , Peptide Initiation Factors/chemistry , Peptide Initiation Factors/metabolism , Protein Biosynthesis/genetics , RNA Caps/genetics , Ribosomes/metabolism , Amino Acid Sequence , Binding Sites , Eukaryotic Initiation Factor-4A , Eukaryotic Initiation Factor-4E , Eukaryotic Initiation Factor-4G , Globins/genetics , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins , Models, Biological , Molecular Sequence Data , Peptide Fragments/genetics , Peptide Initiation Factors/genetics , Point Mutation/genetics , Precipitin Tests , Protein Binding , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Ribosomes/genetics , Sequence Alignment , Sequence Deletion/genetics , Templates, Genetic
8.
Mol Cell Biol ; 19(11): 7336-46, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10523622

ABSTRACT

Eukaryotic initiation factor 4A (eIF4A) is an RNA-dependent ATPase and ATP-dependent RNA helicase that is thought to melt the 5' proximal secondary structure of eukaryotic mRNAs to facilitate attachment of the 40S ribosomal subunit. eIF4A functions in a complex termed eIF4F with two other initiation factors (eIF4E and eIF4G). Two isoforms of eIF4A, eIF4AI and eIF4AII, which are encoded by two different genes, are functionally indistinguishable. A third member of the eIF4A family, eIF4AIII, whose human homolog exhibits 65% amino acid identity to human eIF4AI, has also been cloned from Xenopus and tobacco, but its function in translation has not been characterized. In this study, human eIF4AIII was characterized biochemically. While eIF4AIII, like eIF4AI, exhibits RNA-dependent ATPase activity and ATP-dependent RNA helicase activity, it fails to substitute for eIF4AI in an in vitro-reconstituted 40S ribosome binding assay. Instead, eIF4AIII inhibits translation in a reticulocyte lysate system. In addition, whereas eIF4AI binds independently to the middle and carboxy-terminal fragments of eIF4G, eIF4AIII binds to the middle fragment only. These functional differences between eIF4AI and eIF4AIII suggest that eIF4AIII might play an inhibitory role in translation under physiological conditions.


Subject(s)
Peptide Initiation Factors/genetics , Peptide Initiation Factors/metabolism , Protein Biosynthesis , Adenosine Triphosphatases/metabolism , Amino Acid Sequence , Conserved Sequence , DNA Helicases/metabolism , Eukaryotic Cells , Eukaryotic Initiation Factor-4A , Eukaryotic Initiation Factor-4G , Gene Expression , Gene Library , Models, Theoretical , Molecular Sequence Data , Multigene Family , Protein Binding , Protein Isoforms , Ribosomes/metabolism , Sequence Analysis, DNA , Sequence Homology, Amino Acid
9.
Endocrinology ; 140(6): 2517-25, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10342837

ABSTRACT

The present study examined the trans-activation potential of basic transcription element-binding protein (BTEB), a recently identified member of the Sp family of GC box-binding transcription factors, on the expression of the gene encoding the pregnancy-associated, epithelial-specific, and progesterone (P)-induced porcine uterine endometrial secretory protein, uteroferrin (UF). Endometrial expression of BTEB, P receptor (PR), and UF genes was analyzed by RT-PCR as a function of pregnancy stage and cell type and was correlated with the levels of endometrial BTEB that were quantified by Western blot and/or electrophoretic mobility shift assay. PR, BTEB, and UF messenger RNAs (mRNAs) were present in early (day 12) and mid(day 60) pregnancy pig endometrium, although expression levels varied for each mRNA (UF, day 12 << day 60; PR and BTEB, day 12 = day 60). Within the endometrium, glandular epithelial (GE) cells manifested higher amounts of UF mRNA than stromal fibroblastic cells, whereas both cell types had comparable amounts of BTEB and PR mRNAs. Expression of BTEB, however, was limited to endometrial GE cells. A BTEB expression vector (pcDNA-3BTEB) was used to examine the effect of increased BTEB protein on UF gene expression and promoter activity in primary cultures of pig endometrial GE cells. Cells transiently transfected with pcDNA-3BTEB had 2-fold higher UF mRNA levels than those transfected with the empty expression vector (pcDNA-3). Further, cells cotransfected with a UF promoter-luciferase (-1935UF-Luc) reporter gene and the BTEB expression vector had 2-fold higher Luc activity than those cotransfected with reporter gene and pcDNA-3. This effect of BTEB was not observed in transfected endometrial stromal fibroblastic cells, but was apparent in the human endometrial epithelial carcinoma cell lines ECC-1 and Hec-1-A, which exhibit low levels of BTEB protein and low or undetectable PR mRNA levels, respectively. The respective contributions of BTEB and PR to the modulation of UF promoter activity were examined by cotransfection of Hec-1-A and ECC-1 cells with expression plasmids for BTEB and PR and one of two UF promoter constructs (-831UF-Luc or -1935UF-Luc) in the absence or presence of P. The increase in UF promoter activity with BTEB was mimicked by PR in a P-dependent manner in both cell lines. The combined effect of PR/P and BTEB appeared additive in Hec-1-A cells and was synergistic in ECC-1 cells. These results highlight the cell context dependence of the trans-activation potential of BTEB and suggest its unique role, in concert with PR, in directing the temporal expression of endometrial epithelial genes of pregnancy.


Subject(s)
Endometrium/metabolism , Metalloproteins/genetics , Receptors, Progesterone/physiology , Trans-Activators/physiology , Zinc Fingers , Acid Phosphatase , Animals , Cells, Cultured , Epithelial Cells/metabolism , Female , Humans , Isoenzymes , Pregnancy , Promoter Regions, Genetic , RNA, Messenger/analysis , Receptors, Progesterone/genetics , Swine , Tartrate-Resistant Acid Phosphatase , Trans-Activators/genetics
10.
J Virol ; 73(4): 3467-72, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10074204

ABSTRACT

For many members of the Picornaviridae family, infection of cells results in a shutoff of host protein synthesis. For rhinoviruses and enteroviruses, the shutoff has been explained in part by the cleavage of eukaryotic initiation factor 4GI (eIF4GI), a component of the cap-binding protein complex eIF4F. The cleavage of eIF4GI is mediated by the virus-specific proteinase 2Apro and results in inhibition of cap-dependent, but not cap-independent, translation. The inhibition of host protein synthesis after infection with human rhinovirus 14 (HRV-14) lags behind the cleavage of eIF4GI. Recently, we discovered a functional homolog of eIF4GI, termed eIF4GII, and showed that cleavage of eIF4GII coincides with the shutoff of host cell protein synthesis after poliovirus infection (Gradi et al., Proc. Natl. Acad. Sci. USA 95:11089-11094, 1998). We wished to determine whether eIF4GII cleavage kinetics could also explain the lack of correlation between the kinetics of eIF4GI cleavage and the shutoff of host protein synthesis after rhinovirus infection. In this study, we examined the correlation between human rhinovirus-induced shutoff of host protein synthesis and cleavage of eIF4GI and eIF4GII. In HRV-14-infected HeLa cells, almost no intact eIF4GI could be detected by 4 h postinfection, while only 4% of eIF4GII was cleaved at this time. By 6 h, however, 67% of eIF4GII was cleaved, and this cleavage coincided with a significant (60%) decline of host translation. These results suggest that cleavage of both eIF4GI and eIF4GII is required for HRV-mediated inhibition of host cell protein synthesis and that the cleavage of eIF4GII is the rate-limiting step in the shutoff of host cell protein synthesis after rhinovirus infection.


Subject(s)
Eukaryotic Initiation Factor-4G , Peptide Fragments/metabolism , Peptide Initiation Factors/metabolism , Picornaviridae Infections/metabolism , Rhinovirus , Cell Line , Gene Expression Regulation, Viral , Humans , Peptide Fragments/genetics , Peptide Initiation Factors/genetics , Picornaviridae Infections/genetics , Protein Biosynthesis
11.
EMBO J ; 18(1): 270-9, 1999 Jan 04.
Article in English | MEDLINE | ID: mdl-9878069

ABSTRACT

Human eukaryotic translation initiation factor 4E (eIF4E) binds to the mRNA cap structure and interacts with eIF4G, which serves as a scaffold protein for the assembly of eIF4E and eIF4A to form the eIF4F complex. eIF4E is an important modulator of cell growth and proliferation. It is the least abundant component of the translation initiation machinery and its activity is modulated by phosphorylation and interaction with eIF4E-binding proteins (4E-BPs). One strong candidate for the eIF4E kinase is the recently cloned MAPK-activated protein kinase, Mnk1, which phosphorylates eIF4E on its physiological site Ser209 in vitro. Here we report that Mnk1 is associated with the eIF4F complex via its interaction with the C-terminal region of eIF4G. Moreover, the phosphorylation of an eIF4E mutant lacking eIF4G-binding capability is severely impaired in cells. We propose a model whereby, in addition to its role in eIF4F assembly, eIF4G provides a docking site for Mnk1 to phosphorylate eIF4E. We also show that Mnk1 interacts with the C-terminal region of the translational inhibitor p97, an eIF4G-related protein that does not bind eIF4E, raising the possibility that p97 can block phosphorylation of eIF4E by sequestering Mnk1.


Subject(s)
Peptide Initiation Factors/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Binding Sites , Cell Line , Eukaryotic Initiation Factor-4E , Eukaryotic Initiation Factor-4G , Humans , In Vitro Techniques , Intracellular Signaling Peptides and Proteins , Mice , Models, Biological , Peptide Initiation Factors/genetics , Phosphorylation , Point Mutation , Protein Binding , Transfection
12.
EMBO J ; 17(24): 7480-9, 1998 Dec 15.
Article in English | MEDLINE | ID: mdl-9857202

ABSTRACT

Most eukaryotic mRNAs possess a 5' cap and a 3' poly(A) tail, both of which are required for efficient translation. In yeast and plants, binding of eIF4G to poly(A)-binding protein (PABP) was implicated in poly(A)-dependent translation. In mammals, however, there has been no evidence that eIF4G binds PABP. Using 5' rapid amplification of cDNA, we have extended the known human eIF4GI open reading frame from the N-terminus by 156 amino acids. Co-immunoprecipitation experiments showed that the extended eIF4GI binds PABP, while the N-terminally truncated original eIF4GI cannot. Deletion analysis identified a 29 amino acid sequence in the new N-terminal region as the PABP-binding site. The 29 amino acid stretch is almost identical in eIF4GI and eIF4GII, and the full-length eIF4GII also binds PABP. As previously shown for yeast, human eIF4G binds to a fragment composed of RRM1 and RRM2 of PABP. In an in vitro translation system, an N-terminal fragment which includes the PABP-binding site inhibits poly(A)-dependent translation, but has no effect on translation of a deadenylated mRNA. These results indicate that, in addition to a recently identified mammalian PABP-binding protein, PAIP-1, eIF4G binds PABP and probably functions in poly(A)-dependent translation in mammalian cells.


Subject(s)
Peptide Initiation Factors/metabolism , Protein Biosynthesis , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Amino Acid Sequence , Binding Sites , Eukaryotic Initiation Factor-4G , Humans , Molecular Sequence Data , Peptide Fragments/genetics , Peptide Fragments/metabolism , Peptide Initiation Factors/genetics , Poly(A)-Binding Proteins , Precipitin Tests , Protein Binding , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid
13.
Proc Natl Acad Sci U S A ; 95(19): 11089-94, 1998 Sep 15.
Article in English | MEDLINE | ID: mdl-9736694

ABSTRACT

Eukaryotic initiation factor (eIF) 4GI is a component of the cap-binding protein complex eIF4F, which is required for cap-dependent translation. Infection of cells by poliovirus results in a precipitous decline of host cell protein synthesis, which is preceded by the cleavage of eIF4GI. Cleavage of eIF4GI results in the inhibition of cap-dependent translation. Poliovirus translation is not affected by eIF4GI cleavage, however, because poliovirus mRNA is translated by a cap-independent mechanism. Cleavage of eIF4GI alone cannot explain the shutoff of host protein synthesis, because after infection in the presence of inhibitors of virus replication, eIF4GI is cleaved, yet host protein synthesis is only partially inhibited. Here we show that eIF4GII, a recently discovered functional homolog of eIF4GI, is more resistant to poliovirus-mediated cleavage than eIF4GI, and that its proteolysis is concomitant with the shutoff of host cell protein synthesis. Moreover, infection with poliovirus in the presence of inhibitors of virus replication resulted in efficient cleavage of eIF4GI, but only partial proteolysis of eIF4GII. Thus, cleavage of both eIF4GI and eIF4GII appears to be required for the shutoff of host protein synthesis after poliovirus infection. These results explain several earlier reports documenting the lack of correlation between eIF4GI cleavage and inhibition of cellular mRNA translation after poliovirus infection.


Subject(s)
Eukaryotic Initiation Factor-4G , Peptide Fragments/metabolism , Peptide Initiation Factors/metabolism , Poliovirus/metabolism , Protein Biosynthesis/genetics , Guanidine/pharmacology , HeLa Cells/virology , Humans , Kinetics , Methionine/metabolism , Monensin/pharmacology , Protein Synthesis Inhibitors/metabolism , Proteins/analysis , RNA, Messenger/metabolism , Viral Proteins/metabolism
14.
Mol Cell Biol ; 18(1): 334-42, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9418880

ABSTRACT

Mammalian eukaryotic translation initiation factor 4F (eIF4F) is a cap-binding protein complex consisting of three subunits: eIF4E, eIF4A, and eIF4G. In yeast and plants, two related eIF4G species are encoded by two different genes. To date, however, only one functional eIF4G polypeptide, referred to here as eIF4GI, has been identified in mammals. Here we describe the discovery and functional characterization of a closely related homolog, referred to as eIF4GII. eIF4GI and eIF4GII share 46% identity at the amino acid level and possess an overall similarity of 56%. The homology is particularly high in certain regions of the central and carboxy portions, while the amino-terminal regions are more divergent. Far-Western analysis and coimmunoprecipitation experiments were used to demonstrate that eIF4GII directly interacts with eIF4E, eIF4A, and eIF3. eIF4GII, like eIF4GI, is also cleaved upon picornavirus infection. eIF4GII restores cap-dependent translation in a reticulocyte lysate which had been pretreated with rhinovirus 2A to cleave endogenous eIF4G. Finally, eIF4GII exists as a complex with eIF4E in HeLa cells, because eIF4GII and eIF4E can be purified together by cap affinity chromatography. Taken together, our findings indicate that eIF4GII is a functional homolog of eIF4GI. These results may have important implications for the understanding of the mechanism of shutoff of host protein synthesis following picornavirus infection.


Subject(s)
Peptide Initiation Factors/genetics , Amino Acid Sequence , Cloning, Molecular , Eukaryotic Initiation Factor-4F , Eukaryotic Initiation Factor-4G , Humans , Molecular Sequence Data , Peptide Initiation Factors/isolation & purification , Protein Biosynthesis , Sequence Alignment
15.
Mol Cell Biol ; 17(12): 6940-7, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9372926

ABSTRACT

Mammalian translation initiation factor 4F (eIF4F) consists of three subunits, eIF4A, eIF4E, and eIF4G. eIF4G interacts directly with both eIF4A and eIF4E. The binding site for eIF4E is contained in the amino-terminal third of eIF4G, while the binding site for eIF4A was mapped to the carboxy-terminal third of the molecule. Here we show that human eIF4G possesses two separate eIF4A binding domains in the middle third (amino acids [aa] 478 to 883) and carboxy-terminal third (aa 884 to 1404) of the molecule. The amino acid sequence of the middle portion of eIF4G is well conserved between yeasts and humans. We show that mutations of conserved amino acid stretches in the middle domain abolish or reduce eIF4A binding as well as eIF3 binding. In addition, a separate and nonoverlapping eIF4A binding domain exists in the carboxy-terminal third (aa 1045 to 1404) of eIF4G, which is not present in yeast. The C-terminal two-thirds region (aa 457 to 1404) of eIF4G, containing both eIF4A binding sites, is required for stimulating translation. Neither one of the eIF4A binding domains alone activates translation. In contrast to eIF4G, human p97, a translation inhibitor with homology to eIF4G, binds eIF4A only through the amino-terminal proximal region, which is homologous to the middle domain of eIF4G.


Subject(s)
Peptide Initiation Factors/chemistry , Peptide Initiation Factors/metabolism , Amino Acid Sequence , Binding Sites/genetics , Conserved Sequence , Eukaryotic Initiation Factor-4A , Eukaryotic Initiation Factor-4G , Humans , In Vitro Techniques , Models, Biological , Molecular Sequence Data , Mutagenesis, Site-Directed , Peptide Initiation Factors/genetics , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Sequence Deletion , Sequence Homology, Amino Acid , Species Specificity
16.
EMBO J ; 16(4): 817-25, 1997 Feb 17.
Article in English | MEDLINE | ID: mdl-9049310

ABSTRACT

Translation initiation in eukaryotes is facilitated by the cap structure, m7GpppN (where N is any nucleotide). Eukaryotic translation initiation factor 4F (eIF4F) is a cap binding protein complex that consists of three subunits: eIF4A, eIF4E and eIF4G. eIF4G interacts directly with eIF4E and eIF4A. The binding site of eIF4E resides in the N-terminal third of eIF4G, while eIF4A and eIF3 binding sites are present in the C-terminal two-thirds. Here, we describe a new eukaryotic translational regulator (hereafter called p97) which exhibits 28% identity to the C-terminal two-thirds of eIF4G. p97 mRNA has no initiator AUG and translation starts exclusively at a GUG codon. The GUG-initiated open reading frame (907 amino acids) has no canonical eIF4E binding site. p97 binds to eIF4A and eIF3, but not to eIF4E. Transient transfection experiments show that p97 suppresses both cap-dependent and independent translation, while eIF4G supports both translation pathways. Furthermore, inducible expression of p97 reduces overall protein synthesis. These results suggest that p97 functions as a general repressor of translation by forming translationally inactive complexes that include eIF4A and eIF3, but exclude eIF4E.


Subject(s)
Peptide Initiation Factors/genetics , Sequence Homology, Amino Acid , Amino Acid Sequence , Animals , Base Sequence , COS Cells , Cloning, Molecular , Codon, Initiator/genetics , DNA, Complementary/genetics , Encephalomyocarditis virus/genetics , Eukaryotic Initiation Factor-4E , Eukaryotic Initiation Factor-4G , Gene Expression , HeLa Cells , Humans , Molecular Sequence Data , Open Reading Frames , Peptide Chain Initiation, Translational/genetics , Peptide Initiation Factors/metabolism , Placenta , Protein Binding , RNA, Messenger/analysis , Sequence Analysis, DNA
17.
J Biochem ; 117(1): 91-5, 1995 Jan.
Article in English | MEDLINE | ID: mdl-7775404

ABSTRACT

BTEB is a small GC box-binding protein containing three contiguous zinc finger structures in the C-terminal region of the molecule, and activates or represses the transcription of genes with the GC box sequence in the promoter, depending on the repetitiveness of the GC box sequence [Imataka et al. (1992) EMBO J. 11, 3663-3671]. We have analyzed functional domains of BTEB in a transient expression system using Y-1 cells (a mouse adrenal cortex cell line). BTEB contained two regions responsible for the transcriptional activation. These regions showed a sequence similarity to each other and appeared to enhance the transcription independently. The two sequences were rich in hydrophobic amino acids and showed no similarity to the sequences of transactivation domains so far elucidated. The DNA-binding properties of BTEB were also analyzed by means of the gel mobility shift assay. Three contiguous zinc finger motifs were needed for the binding activity. Furthermore, it was revealed that a short basic region immediately N-terminal to the zinc finger motifs was required for the DNA binding.


Subject(s)
DNA-Binding Proteins/chemistry , Promoter Regions, Genetic , Protein Structure, Tertiary , Transcription Factors/chemistry , Transcriptional Activation , Zinc Fingers , Amino Acid Sequence , Amino Acids/chemistry , Animals , Base Sequence , Cell Line , Kruppel-Like Transcription Factors , Mice , Molecular Sequence Data , Solubility , Water/chemistry
18.
J Biol Chem ; 269(32): 20668-73, 1994 Aug 12.
Article in English | MEDLINE | ID: mdl-8051167

ABSTRACT

BTEB is a GC-box binding transcription factor that can activate human immunodeficiency virus type 1 long terminal repeat and cellular gene promoters containing multiple GC boxes. The present studies showed that although BTEB mRNA was expressed in various tissues of mammals and cell lines, the expression of BTEB protein was confined to the brain and a neuroblastoma Neuro2A (N2A), suggesting that the BTEB expression was translationally regulated in a cell-specific or tissue-specific manner. The BTEB mRNA was characterized by a long (1.26 kilobases( 5'-untranslated region (5'-UTR) containing 10 upstream AUGs (uAUGs) and a GC-rich tract. To examine whether the 5'-UTR controlled the translation in a cell-specific manner, a fusion plasmid composed of the BTEB 5'-UTR and the chloramphenicol acetyltransferase gene was transfected into HeLa and N2A cells. Translational efficiency of the transcribed mRNA was estimated from the chloramphenicol acetyltransferase activity normalized on the basis of the amount of the mRNA. The 5'-UTR was found to decrease the translational efficiency by 7-fold in HeLa cells; that in N2A was not affected. When one of the uAUGs in the 5'-UTR was mutated to AAG, the inhibition of the translation by the 5'-UTR in HeLa cells was reversed; no effect of the mutation was observed in N2A cells. These results suggest that an uAUG in the 5'-UTR of the BTEB mRNA is, at least in part, responsible for the cell-specific translational control of the BTEB expression.


Subject(s)
DNA-Binding Proteins/genetics , Protein Biosynthesis , Transcription Factors/genetics , Animals , Base Sequence , Cell Line , Chloramphenicol O-Acetyltransferase/genetics , DNA, Complementary , Humans , Kruppel-Like Transcription Factors , Male , Molecular Sequence Data , RNA, Messenger/genetics , Rats , Rats, Wistar
19.
J Biochem ; 114(4): 605-9, 1993 Oct.
Article in English | MEDLINE | ID: mdl-8276776

ABSTRACT

We have expressed truncated forms of BTEB and Sp1 in Escherichia coli and investigated the DNA-binding properties of the two proteins. The two proteins as well as their chimeric proteins protected the same DNA region in the BTE sequence (a GC box in the P-4501A1 gene) as examined by ortho-phenanthroline-Cu footprinting. The region overlapped nearly perfectly with the GC box consensus sequence. Methylation interference footprinting revealed that all the guanines within the region and two other guanines in the close vicinity interacted with the proteins. Competitive gel mobility shift assay using various synthetic oligonucleotides of the GC box sequences as the competitors demonstrated that BTEB and Sp1 have similar sequence specificities for DNA binding. We have purified the bacterially expressed BTEB and measured the dissociation constant of the BTEB-BTE complex using gel mobility shift assay. The dissociation constant was (3.0 +/- 1.0) x 10(-10) M and was comparable to that of Sp1 binding to a GC box. Taken together, these findings indicate that the binding modes of BTEB and Sp1 to the GC box are similar to each other.


Subject(s)
DNA-Binding Proteins/metabolism , Sp1 Transcription Factor/metabolism , Transcription Factors/metabolism , Base Sequence , Binding Sites , Binding, Competitive , Consensus Sequence , Copper , DNA/metabolism , DNA-Binding Proteins/genetics , Escherichia coli/genetics , Gene Expression , Kruppel-Like Transcription Factors , Methylation , Molecular Sequence Data , Mutagenesis, Site-Directed , Phenanthrolines , Plasmids , Promoter Regions, Genetic , Recombinant Fusion Proteins/metabolism , Sp1 Transcription Factor/genetics , Transcription Factors/genetics , Transformation, Bacterial
20.
AIDS Res Hum Retroviruses ; 9(9): 825-31, 1993 Sep.
Article in English | MEDLINE | ID: mdl-8257632

ABSTRACT

BTEB, a GC box-binding transcription factor, was tested for its ability to activate the human immunodeficiency virus type 1 long terminal repeat (HIV-1 LTR). An electrophoretic mobility shift assay demonstrated specific binding of BTEB to GC boxes of the HIV-1 LTR. When a BTEB expression vector was cotransfected into A3.01 cells with a fusion gene of HIV-1 LTR and chloramphenicol acetyltransferase (CAT) structural gene, the CAT activity was increased. This increase was accompanied by an increase in the content of CAT mRNA. Transcriptional activity of the HIV-1 LTR, stimulated by Tat, was further enhanced by the expression of BTEB. BTEB also activated the LTR activity in cooperation with phorbol 12-myristate 13-acetate. Northern blot analysis showed that various T cell and macrophage/monocyte cell lines expressed the BTEB mRNA to a level comparable with that of Sp1, another GC box-binding transcription factor. These results suggest that BTEB, like Sp1, is involved in transcriptional activation of the HIV-1 LTR.


Subject(s)
DNA-Binding Proteins/pharmacology , HIV Long Terminal Repeat/drug effects , HIV-1/growth & development , Transcription Factors/pharmacology , Virus Activation/drug effects , Base Sequence , Binding Sites , Gene Products, tat/physiology , HIV-1/drug effects , Humans , Kruppel-Like Transcription Factors , Molecular Sequence Data , NF-kappa B/metabolism , T-Lymphocytes/microbiology , Tetradecanoylphorbol Acetate/pharmacology , Transcription, Genetic , Tumor Cells, Cultured , Zinc Fingers , tat Gene Products, Human Immunodeficiency Virus
SELECTION OF CITATIONS
SEARCH DETAIL
...