Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
RNA ; 16(10): 1902-14, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20688868

RESUMO

To study positioning of the polypeptide release factor eRF1 toward a stop signal in the ribosomal decoding site, we applied photoactivatable mRNA analogs, derivatives of oligoribonucleotides. The human eRF1 peptides cross-linked to these short mRNAs were identified. Cross-linkers on the guanines at the second, third, and fourth stop signal positions modified fragment 31-33, and to lesser extent amino acids within region 121-131 (the "YxCxxxF loop") in the N domain. Hence, both regions are involved in the recognition of the purines. A cross-linker at the first uridine of the stop codon modifies Val66 near the NIKS loop (positions 61-64), and this region is important for recognition of the first uridine of stop codons. Since the N domain distinct regions of eRF1 are involved in a stop-codon decoding, the eRF1 decoding site is discontinuous and is not of "protein anticodon" type. By molecular modeling, the eRF1 molecule can be fitted to the A site proximal to the P-site-bound tRNA and to a stop codon in mRNA via a large conformational change to one of its three domains. In the simulated eRF1 conformation, the YxCxxxF motif and positions 31-33 are very close to a stop codon, which becomes also proximal to several parts of the C domain. Thus, in the A-site-bound state, the eRF1 conformation significantly differs from those in crystals and solution. The model suggested for eRF1 conformation in the ribosomal A site and cross-linking data are compatible.


Assuntos
Códon de Terminação/genética , Códon de Terminação/metabolismo , Fatores de Terminação de Peptídeos/metabolismo , Sequência de Bases , Reagentes de Ligações Cruzadas , Humanos , Técnicas In Vitro , Modelos Moleculares , Mutagênese Sítio-Dirigida , Terminação Traducional da Cadeia Peptídica , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Mapeamento de Peptídeos , Fatores de Terminação de Peptídeos/química , Fatores de Terminação de Peptídeos/genética , Conformação Proteica , Estrutura Terciária de Proteína , RNA Mensageiro/química , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA de Transferência/química , RNA de Transferência/genética , RNA de Transferência/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Ribossomos/genética , Ribossomos/metabolismo
2.
Nucleic Acids Res ; 38(3): e15, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19910372

RESUMO

Inhibition of primer extension by ribosome-mRNA complexes (toeprinting) is a proven and powerful technique for studying mechanisms of mRNA translation. Here we have assayed an advanced toeprinting approach that employs fluorescently labeled DNA primers, followed by capillary electrophoresis utilizing standard instruments for sequencing and fragment analysis. We demonstrate that this improved technique is not merely fast and cost-effective, but also brings the primer extension inhibition method up to the next level. The electrophoretic pattern of the primer extension reaction can be characterized with a precision unattainable by the common toeprint analysis utilizing radioactive isotopes. This method allows us to detect and quantify stable ribosomal complexes at all stages of translation, including initiation, elongation and termination, generated during the complete translation process in both the in vitro reconstituted translation system and the cell lysate. We also point out the unique advantages of this new methodology, including the ability to assay sites of the ribosomal complex assembly on several mRNA species in the same reaction mixture.


Assuntos
Eletroforese Capilar , Biossíntese de Proteínas , RNA Mensageiro/análise , Ribossomos/metabolismo , Primers do DNA , Corantes Fluorescentes , RNA Mensageiro/metabolismo , Transcrição Reversa
3.
FEBS Lett ; 583(21): 3455-60, 2009 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-19796638

RESUMO

Although some data link archaeal and eukaryotic translation, the overall mechanism of protein synthesis in archaea remains largely obscure. Both archaeal (aRF1) and eukaryotic (eRF1) single release factors recognize all three stop codons. The archaeal genus Methanosarcinaceae contains two aRF1 homologs, and also uses the UAG stop to encode the 22nd amino acid, pyrrolysine. Here we provide an analysis of the last stage of archaeal translation in pyrrolysine-utilizing species. We demonstrated that only one of two Methanosarcina barkeri aRF1 homologs possesses activity and recognizes all three stop codons. The second aRF1 homolog may have another unknown function. The mechanism of pyrrolysine incorporation in the Methanosarcinaceae is discussed.


Assuntos
Lisina/análogos & derivados , Methanosarcinaceae/genética , Methanosarcinaceae/metabolismo , Biossíntese de Proteínas , Sequência de Aminoácidos , Proteínas Arqueais/química , Proteínas Arqueais/metabolismo , Códon de Terminação/genética , Códon de Terminação/metabolismo , Genoma Arqueal , Humanos , Lisina/metabolismo , Dados de Sequência Molecular , Fatores de Terminação de Peptídeos/química , Fatores de Terminação de Peptídeos/metabolismo , Filogenia
4.
PLoS One ; 4(5): e5231, 2009 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-19478941

RESUMO

BACKGROUND: Many different genetic alterations are observed in cancer cells. Individual cancer genes display point mutations such as base changes, insertions and deletions that initiate and promote cancer growth and spread. Somatic hypermutation is a powerful mechanism for generation of different mutations. It was shown previously that somatic hypermutability of proto-oncogenes can induce development of lymphomas. METHODOLOGY/PRINCIPAL FINDINGS: We found an exceptionally high incidence of single-base mutations in the tumor suppressor genes RASSF1 and RBSP3 (CTDSPL) both located in 3p21.3 regions, LUCA and AP20 respectively. These regions contain clusters of tumor suppressor genes involved in multiple cancer types such as lung, kidney, breast, cervical, head and neck, nasopharyngeal, prostate and other carcinomas. Altogether in 144 sequenced RASSF1A clones (exons 1-2), 129 mutations were detected (mutation frequency, MF = 0.23 per 100 bp) and in 98 clones of exons 3-5 we found 146 mutations (MF = 0.29). In 85 sequenced RBSP3 clones, 89 mutations were found (MF = 0.10). The mutations were not cytidine-specific, as would be expected from alterations generated by AID/APOBEC family enzymes, and appeared de novo during cell proliferation. They diminished the ability of corresponding transgenes to suppress cell and tumor growth implying a loss of function. These high levels of somatic mutations were found both in cancer biopsies and cancer cell lines. CONCLUSIONS/SIGNIFICANCE: This is the first report of high frequencies of somatic mutations in RASSF1 and RBSP3 in different cancers suggesting it may underlay the mutator phenotype of cancer. Somatic hypermutations in tumor suppressor genes involved in major human malignancies offer a novel insight in cancer development, progression and spread.


Assuntos
Mutação/genética , Neoplasias/genética , Proteínas Supressoras de Tumor/genética , Desaminase APOBEC-1 , Animais , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/patologia , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células , Células Clonais , Biologia Computacional , Citidina Desaminase/metabolismo , DNA Bacteriano/genética , DNA Complementar/genética , Proteínas de Escherichia coli/genética , Etiquetas de Sequências Expressas , Efeito Fundador , Genoma/genética , Hematopoese/genética , Humanos , Neoplasias Renais/genética , Neoplasias Renais/patologia , Camundongos , Camundongos SCID , Reação em Cadeia da Polimerase
5.
Biotechniques ; 44(1): 77-83, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18254383

RESUMO

To analyze RNA interactions with RNA binding molecules an RNA microchip containing immobilized oligoribonucleotides with protective groups [t-butyldimethylsilyl (tBDMS)] at 2'-O- positions was developed. The oligonucleotides were immobilized within three-dimensional (3-D) hydrogel pads fixed on a glass support. The protective groups preserved the oligoribonucleodes from degradation and were suitable to be removed directly on the microchip when needed, right before its use. These immobilized, deprotected oligoribonucleotides were tested for their interaction with afluorescently labeled oligodeoxyribonucleotide and analyzed for their availability to be cleaved enzymatically by the RNase binase. Stability of tBDMS-protected immobilized oligoribonucleotides after 2.5 years of storage as well as after direct RNase action was also tested. Melting curves of short RNA/DNA hybrids that had formed into gel pads of the microchip were found to exhibit clearly defined S-like shapes, with the melting temperatures in full accordance with those theoretically predicted for the same ionic strength. This approach, based on keeping the protective groups attached to oligoribonucleotides, can be applied for manufacturing any RNA microchips containing immobilized oligoribonucleotides, including microchips with two-dimensional (2-D) features. These RNA microchips can be used to measure thermodynamic parameters of RNA/RNA or RNA/DNA duplexes as well as to study ligand- or protein-RNA interactions.


Assuntos
Análise de Sequência com Séries de Oligonucleotídeos/métodos , Oligorribonucleotídeos/metabolismo , RNA/metabolismo , Sequência de Bases , DNA/metabolismo , Endorribonucleases/metabolismo , Corantes Fluorescentes/metabolismo , Hidrogel de Polietilenoglicol-Dimetacrilato , Cinética , Dados de Sequência Molecular , Ácidos Nucleicos Heteroduplexes , Oligorribonucleotídeos/genética , Termodinâmica
6.
Proteins ; 70(2): 388-93, 2008 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-17680691

RESUMO

Eukaryotic translational termination is triggered by polypeptide release factors eRF1, eRF3, and one of the three stop codons at the ribosomal A-site. Isothermal titration calorimetry shows that (i) the separated MC, M, and C domains of human eRF1 bind to eRF3; (ii) GTP binding to eRF3 requires complex formation with either the MC or M + C domains; (iii) the M domain interacts with the N and C domains; (iv) the MC domain and Mg2+ induce GTPase activity of eRF3 in the ribosome. We suggest that GDP binding site of eRF3 acquires an ability to bind gamma-phosphate of GTP if altered by cooperative action of the M and C domains of eRF1. Thus, the stop-codon decoding is associated with the N domain of eRF1 while the GTPase activity of eRF3 is controlled by the MC domain of eRF1 demonstrating a substantial structural uncoupling of these two activities though functionally they are interrelated.


Assuntos
Guanosina Trifosfato/metabolismo , Fatores de Terminação de Peptídeos/química , Fatores de Terminação de Peptídeos/metabolismo , Calorimetria , Códon de Terminação , GTP Fosfo-Hidrolases/metabolismo , Humanos , Termodinâmica
7.
FEBS J ; 274(16): 4223-37, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17651434

RESUMO

The eukaryotic class 1 polypeptide chain release factor is a three-domain protein involved in the termination of translation, the final stage of polypeptide biosynthesis. In attempts to understand the roles of the middle domain of the eukaryotic class 1 polypeptide chain release factor in the transduction of the termination signal from the small to the large ribosomal subunit and in peptidyl-tRNA hydrolysis, its high-resolution NMR structure has been obtained. The overall fold and the structure of the beta-strand core of the protein in solution are similar to those found in the crystal. However, the orientation of the functionally critical GGQ loop and neighboring alpha-helices has genuine and noticeable differences in solution and in the crystal. Backbone amide protons of most of the residues in the GGQ loop undergo fast exchange with water. However, in the AGQ mutant, where functional activity is abolished, a significant reduction in the exchange rate of the amide protons has been observed without a noticeable change in the loop conformation, providing evidence for the GGQ loop interaction with water molecule(s) that may serve as a substrate for the hydrolytic cleavage of the peptidyl-tRNA in the ribosome. The protein backbone dynamics, studied using 15N relaxation experiments, showed that the GGQ loop is the most flexible part of the middle domain. The conformational flexibility of the GGQ and 215-223 loops, which are situated at opposite ends of the longest alpha-helix, could be a determinant of the functional activity of the eukaryotic class 1 polypeptide chain release factor, with that helix acting as the trigger to transmit the signals from one loop to the other.


Assuntos
Fatores de Terminação de Peptídeos/química , Aminoacil-RNA de Transferência/química , Ribossomos/química , Motivos de Aminoácidos/genética , Sequência de Aminoácidos , Sítios de Ligação/genética , Células Eucarióticas/metabolismo , Hidrólise , Espectroscopia de Ressonância Magnética/métodos , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Terminação Traducional da Cadeia Peptídica , Fatores de Terminação de Peptídeos/genética , Fatores de Terminação de Peptídeos/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Aminoacil-RNA de Transferência/metabolismo , Ribossomos/metabolismo , Homologia de Sequência de Aminoácidos
8.
Proc Natl Acad Sci U S A ; 104(26): 10824-9, 2007 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-17573528

RESUMO

In universal-code eukaryotes, a single-translation termination factor, eukaryote class-1 polypeptide release factor (eRF1), decodes the three stop codons: UAA, UAG, and UGA. In some ciliates, like Stylonychia and Paramecium, eRF1s exhibit UGA-only decoding specificity, whereas UAG and UAA are reassigned as sense codons. Because variant-code ciliates may have evolved from universal-code ancestor(s), structural features should exist in ciliate eRF1s that restrict their stop codon recognition. In omnipotent eRF1s, stop codon recognition is associated with the N-terminal domain of the protein. Using both in vitro and in vivo assays, we show here that chimeric molecules composed of the N-terminal domain of Stylonychia eRF1 fused to the core domain (MC domain) of human eRF1 retained specificity toward UGA; this unambiguously associates eRF1 stop codon specificity to the nature of its N-terminal domain. Functional analysis of eRF1 chimeras constructed by swapping ciliate N-terminal domain sequences with the matching ones from the human protein highlighted the crucial role of the tripeptide QFM in restricting Stylonychia eRF1 specificity toward UGA. Using the site-directed mutagenesis, we show that Paramecium eRF1 specificity toward UGA resides within the NIKS (amino acids 61-64) and YxCxxxF (amino acids 124-131) motifs. Thus, we establish that eRF1 from two different ciliates relies on different molecular mechanisms to achieve specificity toward the UGA stop codon. This finding suggests that eRF1 restriction of specificity to only UGA might have been an early event occurring in independent instances in ciliate evolutionary history, possibly facilitating the reassignment of UAG and UAA to sense codons.


Assuntos
Cilióforos/genética , Códon de Terminação , Fatores de Terminação de Peptídeos/genética , Biossíntese de Proteínas/genética , Proteínas de Protozoários/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Humanos , Paramecium/genética , Fatores de Terminação de Peptídeos/fisiologia , Proteínas de Protozoários/fisiologia , Proteínas Recombinantes de Fusão , Especificidade por Substrato
9.
Biomol NMR Assign ; 1(2): 183-5, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19636860

RESUMO

We report NMR assignments of the protein backbone of the C-terminal domain (163 a.a.) of human class 1 translation termination factor eRF1. It was found that several protein loop residues exist in two slowly interconverting conformational states.


Assuntos
Espectroscopia de Ressonância Magnética/métodos , Fatores de Terminação de Peptídeos/química , Sequência de Aminoácidos , Humanos , Estrutura Terciária de Proteína
10.
Nucleic Acids Res ; 34(14): 3947-54, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16914449

RESUMO

GTP hydrolysis catalyzed in the ribosome by a complex of two polypeptide release factors, eRF1 and eRF3, is required for fast and efficient termination of translation in eukaryotes. Here, isothermal titration calorimetry is used for the quantitative thermodynamic characterization of eRF3 interactions with guanine nucleotides, eRF1 and Mg2+. We show that (i) eRF3 binds GDP (K(d) = 1.9 microM) and this interaction depends only minimally on the Mg(2+) concentration; (ii) GTP binds to eRF3 (K(d) = 0.5 microM) only in the presence of eRF1 and this interaction depends on the Mg2+ concentration; (iii) GTP displaces GDP from the eRF1*eRF3*GDP complex, and vice versa; (iv) eRF3 in the GDP-bound form improves its ability to bind eRF1; (v) the eRF1*eRF3 complex binds GDP as efficiently as free eRF3; (vi) the eRF1*eRF3 complex is efficiently formed in the absence of GDP/GTP but requires the presence of the C-terminus of eRF1 for complex formation. Our results show that eRF1 mediates GDP/GTP displacement on eRF3. We suggest that after formation of eRF1*eRF3*GTP*Mg2+, this quaternary complex binds to the ribosomal pretermination complex containing P-site-bound peptidyl-tRNA and the A-site-bound stop codon. The guanine nucleotide binding properties of eRF3 and of the eRF3*eRF1 complex profoundly differ from those of prokaryotic RF3.


Assuntos
Guanosina Trifosfato/metabolismo , Magnésio/metabolismo , Terminação Traducional da Cadeia Peptídica , Fatores de Terminação de Peptídeos/metabolismo , Fatores de Terminação de Peptídeos/fisiologia , Proteínas de Bactérias/fisiologia , Calorimetria , Guanosina Difosfato/metabolismo , Humanos , Modelos Biológicos
11.
Biochimie ; 88(7): 747-57, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16797113

RESUMO

In eukaryotes, termination of mRNA translation is triggered by the essential polypeptide chain release factors eRF1, recognizing all three stop codons, and eRF3, a member of the GTPase superfamily with a role that has remained opaque. We have studied the kinetic and thermodynamic parameters of the interactions between eRF3 and GTP, GDP and the non-hydrolysable GTP analogue GDPNP in the presence (K(D)(GDP)=1.3+/-0.2 muM, K(D)(GTP) approximately 200 muM and K(D)(GDPNP)>160 muM) as well as absence (K(D)(GDP)=1.9+/-0.3 muM, K(D)(GTP) 0.7+/-0.2 muM and K(D)(GDPNP) approximately 200 muM) of eRF1. From the present data we propose that (i) free eRF3 has a strong preference to bind GDP compared to GTP (ii) eRF3 in complex with eRF1 has much stronger affinity to GTP than free eRF3 (iii) eRF3 in complex with PABP has weak affinity to GTP (iv) eRF3 in complex with eRF1 does not have strong affinity to GDPNP, implying that GDPNP is a poor analogue of GTP for eRF3 binding.


Assuntos
Guanosina Trifosfato/metabolismo , Fatores de Terminação de Peptídeos/metabolismo , GTP Fosfo-Hidrolases/metabolismo , Guanosina Difosfato/metabolismo , Guanosina Trifosfato/análogos & derivados , Humanos , Cinética , Modelos Biológicos , Terminação Traducional da Cadeia Peptídica/fisiologia , Ligação Proteica/fisiologia , Ribossomos/metabolismo , Saccharomyces cerevisiae/química
12.
Cell ; 125(6): 1125-36, 2006 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-16777602

RESUMO

Eukaryotic translation termination is triggered by peptide release factors eRF1 and eRF3. Whereas eRF1 recognizes all three termination codons and induces hydrolysis of peptidyl tRNA, eRF3's function remains obscure. Here, we reconstituted all steps of eukaryotic translation in vitro using purified ribosomal subunits; initiation, elongation, and termination factors; and aminoacyl tRNAs. This allowed us to investigate termination using pretermination complexes assembled on mRNA encoding a tetrapeptide and to propose a model for translation termination that accounts for the cooperative action of eRF1 and eRF3 in ensuring fast release of nascent polypeptide. In this model, binding of eRF1, eRF3, and GTP to pretermination complexes first induces a structural rearrangement that is manifested as a 2 nucleotide forward shift of the toeprint attributed to pretermination complexes that leads to GTP hydrolysis followed by rapid hydrolysis of peptidyl tRNA. Cooperativity between eRF1 and eRF3 required the eRF3 binding C-terminal domain of eRF1.


Assuntos
Fatores de Terminação de Peptídeos/química , Biossíntese de Proteínas , Animais , Códon de Terminação , Guanosina Trifosfato/fisiologia , Hidrólise , Modelos Biológicos , Terminação Traducional da Cadeia Peptídica , Fatores de Terminação de Peptídeos/fisiologia , Ligação Proteica , Subunidades Proteicas/química , Aminoacil-RNA de Transferência/química , Aminoacil-RNA de Transferência/fisiologia , Coelhos , Ribossomos/química , Ribossomos/fisiologia
14.
Nucleic Acids Res ; 33(19): 6418-25, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16282590

RESUMO

In eukaryotic ribosome, the N domain of polypeptide release factor eRF1 is involved in decoding stop signals in mRNAs. However, structure of the decoding site remains obscure. Here, we specifically altered the stop codon recognition pattern of human eRF1 by point mutagenesis of the invariant Glu55 and Tyr125 residues in the N domain. The 3D structure of generated eRF1 mutants was not destabilized as demonstrated by calorimetric measurements and calculated free energy perturbations. In mutants, the UAG response was most profoundly and selectively affected. Surprisingly, Glu55Arg mutant completely retained its release activity. Substitution of the aromatic ring in position 125 reduced response toward all stop codons. This result demonstrates the critical importance of Tyr125 for maintenance of the intact structure of the eRF1 decoding site. The results also suggest that Tyr125 is implicated in recognition of the 3d stop codon position and probably forms an H-bond with Glu55. The data point to a pivotal role played by the YxCxxxF motif (positions 125-131) in purine discrimination of the stop codons. We speculate that eRF1 decoding site is formed by a 3D network of amino acids side chains.


Assuntos
Ácido Glutâmico/química , Fatores de Terminação de Peptídeos/química , Tirosina/química , Sequência de Aminoácidos , Códon de Terminação , Ácido Glutâmico/genética , Humanos , Ligação de Hidrogênio , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Fatores de Terminação de Peptídeos/genética , Fatores de Terminação de Peptídeos/metabolismo , Desnaturação Proteica , Tirosina/genética
15.
Nucleic Acids Res ; 33(16): 5226-34, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16162810

RESUMO

Termination of protein synthesis is promoted in ribosomes by proper stop codon discrimination by class 1 polypeptide release factors (RFs). A large set of prokaryotic RFs differing in stop codon specificity, RF1 for UAG and UAA, and RF2 for UGA and UAA, was analyzed by means of a recently developed computational method allowing identification of the specificity-determining positions (SDPs) in families composed of proteins with similar but not identical function. Fifteen SDPs were identified within the RF1/2 superdomain II/IV known to be implicated in stop codon decoding. Three of these SDPs had particularly high scores. Five residues invariant for RF1 and RF2 [invariant amino acid residues (IRs)] were spatially clustered with the highest-scoring SDPs that in turn were located in two zones within the SDP/IR area. Zone 1 (domain II) included PxT and SPF motifs identified earlier by others as 'discriminator tripeptides'. We suggest that IRs in this zone take part in the recognition of U, the first base of all stop codons. Zone 2 (domain IV) possessed two SDPs with the highest scores not identified earlier. Presumably, they also take part in stop codon binding and discrimination. Elucidation of potential functional role(s) of the newly identified SDP/IR zones requires further experiments.


Assuntos
Fatores de Terminação de Peptídeos/química , Algoritmos , Sequência de Aminoácidos , Aminoácidos/química , Códon de Terminação , Biologia Computacional , Modelos Moleculares , Dados de Sequência Molecular , Terminação Traducional da Cadeia Peptídica , Fatores de Terminação de Peptídeos/classificação , Fatores de Terminação de Peptídeos/metabolismo , Alinhamento de Sequência , Análise de Sequência de Proteína
16.
J Biol Chem ; 280(4): 2439-45, 2005 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-15509572

RESUMO

Polypeptide release factors from eubacteria and eukaryotes, although similar in function, belong to different protein families. They share one sequence motif, a GGQ tripeptide that is vital to release factor (RF) activity in both kingdoms. In bacteria, the Gln residue of the motif in RF1 and RF2 is modified to N(5)-methyl-Gln by the S-adenosyl l-methionine-dependent methyltransferase PrmC and the absence of Gln methylation decreases the release activity of Escherichia coli RF2 in vitro severalfold. We show here that the same modification is made to the GGQ motif of Saccharomyces cerevisiae release factor eRF1, the first time that N(5)-methyl-Gln has been found outside the bacterial kingdom. The product of the YDR140w gene is required for the methylation of eRF1 in vivo and for optimal yeast cell growth. YDR140w protein has significant homology to PrmC but lacks the N-terminal domain thought to be involved in the recognition of the bacterial release factors. Overproduced in S. cerevisiae, YDR140w can methylate eRF1 from yeast or man in vitro using S-adenosyl l-methionine as methyl donor provided that eRF3 and GTP are also present, suggesting that the natural substrate of the methyltransferase YDR140w is the ternary complex eRF1.eRF3.GTP.


Assuntos
Glutamina/química , Metiltransferases/fisiologia , Fatores de Terminação de Peptídeos/fisiologia , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/fisiologia , Saccharomyces cerevisiae/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Eletroforese em Gel de Poliacrilamida , Escherichia coli/metabolismo , Guanosina Trifosfato/metabolismo , Histidina/química , Espectrometria de Massas , Metilação , Metiltransferases/química , Modelos Moleculares , Dados de Sequência Molecular , Fatores de Terminação de Peptídeos/química , Plasmídeos/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
17.
Oncogene ; 23(34): 5719-28, 2004 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-15208675

RESUMO

We searched for chromosome 3p homo- and hemizygous losses in 23 lung cancer cell lines, 53 renal cell and 22 breast carcinoma biopsies using 31 microsatellite markers located in frequently deleted 3p regions. In addition, two sequence-tagged site markers (NLJ-003 and NL3-001) located in the Alu-PCR clone 20 region (AP20) and lung cancer (LUCA) regions, respectively, were used for quantitative real-time PCR (QPCR). We found frequent (10-18%) homozygous deletions (HDs) in both 3p21.3 regions in the biopsies and lung cancer cell lines. In addition, we discovered that amplification of 3p is a very common (15-42.5%) event in these cancers and probably in other epithelial malignancies. QPCR showed that aberrations of either NLJ-003 or NL3-001 were detected in more than 90% of all studied cases. HDs were frequently detected simultaneously both in NLJ-003 or NL3-001 loci in the same tumour (P<3-10(-7)). This observation suggests that tumour suppressor genes (TSG) in these regions could have a synergistic effect. The exceptionally high frequency of chromosome aberrations in NLJ-003 and NL3-001 loci suggests that multiple TSG(s) involved in different malignancies are located very near to these markers. Precise mapping of 15 independent HDs in the LUCA region allowed us to establish the smallest HD region in 3p21.3C located between D3S1568 (CACNA2D2 gene) and D3S4604 (SEMA3F gene). This region contains 17 genes. Mapping of 19 HDs in the AP20 region resulted in the localization of the minimal region to the interval flanked by D3S1298 and D3S3623 markers. Only four genes were discovered in this interval, namely, APRG1, ITGA9, HYA22 and VILL.


Assuntos
Neoplasias da Mama/genética , Carcinoma/genética , Neoplasias Renais/genética , Neoplasias Pulmonares/genética , Deleção de Sequência , Canais de Cálcio/genética , Carcinoma de Células Renais/genética , Carcinoma de Células Pequenas/genética , Aberrações Cromossômicas , Cromossomos Humanos Par 3 , Feminino , Rearranjo Gênico , Marcadores Genéticos , Homozigoto , Humanos , Perda de Heterozigosidade , Glicoproteínas de Membrana/genética , Proteínas de Membrana/genética , Proteínas do Tecido Nervoso/genética , Reação em Cadeia da Polimerase/métodos , Semaforinas , Células Tumorais Cultivadas
18.
Proc Natl Acad Sci U S A ; 101(14): 4906-11, 2004 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-15051889

RESUMO

Chromosome 3p21.3 region is frequently (>90%) deleted in lung and other major human carcinomas. We subdivided 3p21.3 into LUCA and AP20 subregions and discovered frequent homozygous deletions (10-18%) in both subregions. This finding strongly implies that they harbor multiple tumor suppressor genes involved in the origin and/or development of major epithelial cancers. In this study, we performed an initial analysis of RBSP3/HYA22, a candidate tumor suppressor genes located in the AP20 region. Two sequence splice variants of RBSP3/HYA22 (A and B) were identified, and we provide evidence for their tumor suppressor function. By sequence analysis RBSP3/HYA22 belongs to a gene family of small C-terminal domain phosphatases that may control the RNA polymerase II transcription machinery. Expression of the gene was drastically (>20-fold) decreased in 11 of 12 analyzed carcinoma cell lines and in three of eight tumor biopsies. We report missense and nonsense mutations in tumors where RBSP3/HYA22 was expressed, growth suppression with regulated transgenes in culture, suppression of tumor formation in severe combined immunodeficient mice, and dephosphorylation of ppRB by RBSP3/HYA22, presumably leading to a block of the cell cycle at the G1/S boundary.


Assuntos
Genes Supressores de Tumor , Proteínas Supressoras de Tumor/genética , Sequência de Aminoácidos , Sequência de Bases , Divisão Celular/genética , Linhagem Celular Tumoral , Metilação de DNA , Primers do DNA , Sondas de DNA , Deleção de Genes , Humanos , Repetições de Microssatélites , Dados de Sequência Molecular , Fosforilação , Reação em Cadeia da Polimerase , Splicing de RNA , Homologia de Sequência de Aminoácidos , Sitios de Sequências Rotuladas , Proteínas Supressoras de Tumor/química , Proteínas Supressoras de Tumor/fisiologia
19.
J Mol Biol ; 331(4): 745-58, 2003 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-12909007

RESUMO

To investigate the codon dependence of human eRF1 binding to the mRNA-ribosome complex, we examined the formation of photocrosslinks between ribosomal components and mRNAs bearing a photoactivable 4-thiouridine probe in the first position of the codon located in the A site. Addition of eRF1 to the phased mRNA-ribosome complexes triggers a codon-dependent quenching of crosslink formation. The concentration of eRF1 triggering half quenching ranges from low for the three stop codons, to intermediate for s4UGG and high for other near-cognate triplets. A theoretical analysis of the photochemical processes occurring in a two-state bimolecular model raises a number of stringent conditions, fulfilled by the system studied here, and shows that in any case sound KD values can be extracted if the ratio mT/KD<<1 (mT is total concentration of mRNA added). Considering the KD values obtained for the stop, s4UGG and sense codons (approximately 0.06 microM, 0.45 microM and 2.3 microM, respectively) and our previous finding that only the stop and s4UGG codons are able to promote formation of an eRF1-mRNA crosslink, implying a role for the NIKS loop at the tip of the N domain, we propose a two-step model for eRF1 binding to the A site: a codon-independent bimolecular step is followed by an isomerisation step observed solely with stop and s4UGG codons. Full recognition of the stop codons by the N domain of eRF1 triggers a rearrangement of bound eRF1 from an open to a closed conformation, allowing the universally conserved GGQ loop at the tip of the M domain to come into close proximity of the peptidyl transferase center of the ribosome. UGG is expected to behave as a cryptic stop codon, which, owing to imperfect eRF1-codon recognition, does not allow full reorientation of the M domain of eRF1. As far as the physical steps of eRF1 binding to the ribosome are considered, they appear to closely mimic the behaviour of the tRNA/EF-Tu/GTP complex, but clearly eRF1 is endowed with a greater conformational flexibility than tRNA.


Assuntos
Códon de Terminação/genética , Códon de Terminação/metabolismo , Fatores de Terminação de Peptídeos/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ribossomos/química , Ribossomos/metabolismo , Sequência de Bases , Sítios de Ligação , Reagentes de Ligações Cruzadas , Humanos , Substâncias Macromoleculares , Modelos Biológicos , Peso Molecular , Oligonucleotídeos/genética , Oligonucleotídeos/metabolismo , Ligação Proteica , Biossíntese de Proteínas
20.
FEBS Lett ; 548(1-3): 97-102, 2003 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-12885414

RESUMO

Positioning of the mRNA codon towards the 18S ribosomal RNA in the A site of human 80S ribosomes has been studied applying short mRNA analogs containing either the stop codon UAA or the sense codon UCA with a perfluoroaryl azide group at the uridine residue. Bound to the ribosomal A site, a modified codon crosslinks exclusively to the 40S subunits under mild UV irradiation. This result is inconsistent with the hypothesis [Ivanov et al. (2001) RNA 7, 1683-1692] which requires direct contact between the large rRNA and the stop codon of the mRNA as recognition step at translation termination. Both sense and stop codons crosslink to the same A1823/A1824 invariant dinucleotide in helix 44 of 18S rRNA. The data point to the resemblance between the ternary complexes formed at elongation (sense codon.aminoacyl-tRNA.AA dinucleotide of 18S rRNA) and termination (stop codon.eRF1.AA dinucleotide of 18S rRNA) steps of protein synthesis and support the view that eRF1 may be considered as a functional mimic of aminoacyl-tRNA.


Assuntos
Códon de Terminação , Códon , RNA Ribossômico 18S/metabolismo , Azidas , Sequência de Bases , Sítios de Ligação , Reagentes de Ligações Cruzadas , Fosfatos de Dinucleosídeos , Oligorribonucleotídeos/metabolismo , Biossíntese de Proteínas/genética , RNA Bacteriano/metabolismo , RNA Mensageiro/metabolismo , RNA Ribossômico 18S/química , Aminoacil-RNA de Transferência , RNA de Transferência de Fenilalanina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...