Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
RNA ; 16(10): 1902-14, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20688868

RESUMEN

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.


Asunto(s)
Codón de Terminación/genética , Codón de Terminación/metabolismo , Factores de Terminación de Péptidos/metabolismo , Secuencia de Bases , Reactivos de Enlaces Cruzados , Humanos , Técnicas In Vitro , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Terminación de la Cadena Péptídica Traduccional , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Mapeo Peptídico , Factores de Terminación de Péptidos/química , Factores de Terminación de Péptidos/genética , Conformación Proteica , Estructura Terciaria de Proteína , ARN Mensajero/química , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN de Transferencia/química , ARN de Transferencia/genética , ARN de Transferencia/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Ribosomas/genética , Ribosomas/metabolismo
2.
Nucleic Acids Res ; 38(3): e15, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19910372

RESUMEN

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.


Asunto(s)
Electroforesis Capilar , Biosíntesis de Proteínas , ARN Mensajero/análisis , Ribosomas/metabolismo , Cartilla de ADN , Colorantes Fluorescentes , ARN Mensajero/metabolismo , Transcripción Reversa
3.
Proc Natl Acad Sci U S A ; 104(26): 10824-9, 2007 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-17573528

RESUMEN

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.


Asunto(s)
Cilióforos/genética , Codón de Terminación , Factores de Terminación de Péptidos/genética , Biosíntesis de Proteínas/genética , Proteínas Protozoarias/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Humanos , Paramecium/genética , Factores de Terminación de Péptidos/fisiología , Proteínas Protozoarias/fisiología , Proteínas Recombinantes de Fusión , Especificidad por Sustrato
4.
Proteins ; 70(2): 388-93, 2008 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-17680691

RESUMEN

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.


Asunto(s)
Guanosina Trifosfato/metabolismo , Factores de Terminación de Péptidos/química , Factores de Terminación de Péptidos/metabolismo , Calorimetría , Codón de Terminación , GTP Fosfohidrolasas/metabolismo , Humanos , Termodinámica
5.
Biotechniques ; 44(1): 77-83, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18254383

RESUMEN

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.


Asunto(s)
Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Oligorribonucleótidos/metabolismo , ARN/metabolismo , Secuencia de Bases , ADN/metabolismo , Endorribonucleasas/metabolismo , Colorantes Fluorescentes/metabolismo , Hidrogel de Polietilenoglicol-Dimetacrilato , Cinética , Datos de Secuencia Molecular , Ácidos Nucleicos Heterodúplex , Oligorribonucleótidos/genética , Termodinámica
6.
Nucleic Acids Res ; 34(14): 3947-54, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16914449

RESUMEN

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.


Asunto(s)
Guanosina Trifosfato/metabolismo , Magnesio/metabolismo , Terminación de la Cadena Péptídica Traduccional , Factores de Terminación de Péptidos/metabolismo , Factores de Terminación de Péptidos/fisiología , Proteínas Bacterianas/fisiología , Calorimetría , Guanosina Difosfato/metabolismo , Humanos , Modelos Biológicos
7.
FEBS J ; 274(16): 4223-37, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17651434

RESUMEN

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.


Asunto(s)
Factores de Terminación de Péptidos/química , Aminoacil-ARN de Transferencia/química , Ribosomas/química , Secuencias de Aminoácidos/genética , Secuencia de Aminoácidos , Sitios de Unión/genética , Células Eucariotas/metabolismo , Hidrólisis , Espectroscopía de Resonancia Magnética/métodos , Modelos Moleculares , Datos de Secuencia Molecular , Mutación , Terminación de la Cadena Péptídica Traduccional , Factores de Terminación de Péptidos/genética , Factores de Terminación de Péptidos/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Aminoacil-ARN de Transferencia/metabolismo , Ribosomas/metabolismo , Homología de Secuencia de Aminoácido
8.
Nucleic Acids Res ; 33(16): 5226-34, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16162810

RESUMEN

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.


Asunto(s)
Factores de Terminación de Péptidos/química , Algoritmos , Secuencia de Aminoácidos , Aminoácidos/química , Codón de Terminación , Biología Computacional , Modelos Moleculares , Datos de Secuencia Molecular , Terminación de la Cadena Péptídica Traduccional , Factores de Terminación de Péptidos/clasificación , Factores de Terminación de Péptidos/metabolismo , Alineación de Secuencia , Análisis de Secuencia de Proteína
9.
Nucleic Acids Res ; 33(19): 6418-25, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16282590

RESUMEN

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.


Asunto(s)
Ácido Glutámico/química , Factores de Terminación de Péptidos/química , Tirosina/química , Secuencia de Aminoácidos , Codón de Terminación , Ácido Glutámico/genética , Humanos , Enlace de Hidrógeno , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Factores de Terminación de Péptidos/genética , Factores de Terminación de Péptidos/metabolismo , Desnaturalización Proteica , Tirosina/genética
10.
Biochimie ; 88(7): 747-57, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16797113

RESUMEN

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.


Asunto(s)
Guanosina Trifosfato/metabolismo , Factores de Terminación de Péptidos/metabolismo , GTP Fosfohidrolasas/metabolismo , Guanosina Difosfato/metabolismo , Guanosina Trifosfato/análogos & derivados , Humanos , Cinética , Modelos Biológicos , Terminación de la Cadena Péptídica Traduccional/fisiología , Unión Proteica/fisiología , Ribosomas/metabolismo , Saccharomyces cerevisiae/química
11.
Nucleic Acids Res ; 30(14): 3163-70, 2002 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-12136098

RESUMEN

A set of 22 551 unique human NotI flanking sequences (16.2 Mb) was generated. More than 40% of the set had regions with significant similarity to known proteins and expressed sequences. The data demonstrate that regions flanking NotI sites are less likely to form nucleosomes efficiently and resemble promoter regions. The draft human genome sequence contained 55.7% of the NotI flanking sequences, Celera's database contained matches to 57.2% of the clones and all public databases (including non-human and previously sequenced NotI flanks) matched 89.2% of the NotI flanking sequences (identity > or =90% over at least 50 bp, data from December 2001). The data suggest that the shotgun sequencing approach used to generate the draft human genome sequence resulted in a bias against cloning and sequencing of NotI flanks. A rough estimation (based primarily on chromosomes 21 and 22) is that the human genome contains 15 000-20 000 NotI sites, of which 6000-9000 are unmethylated in any particular cell. The results of the study suggest that the existing tools for computational determination of CpG islands fail to identify a significant fraction of functional CpG islands, and unmethylated DNA stretches with a high frequency of CpG dinucleotides can be found even in regions with low CG content.


Asunto(s)
ADN/metabolismo , Desoxirribonucleasas de Localización Especificada Tipo II/metabolismo , Análisis de Secuencia de ADN/métodos , Línea Celular Transformada , Cromosomas Humanos Par 21/genética , Cromosomas Humanos Par 22/genética , Islas de CpG/genética , ADN/química , ADN/genética , Bases de Datos de Ácidos Nucleicos , Genes/genética , Genoma Humano , Humanos , Datos de Secuencia Molecular , Secuencias Repetitivas de Ácidos Nucleicos/genética
12.
Structure ; 10(1): 8-9, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11796105

RESUMEN

Although the eukaryotic (eRF1) and prokaryotic (RF2) polypeptide release (translation termination) factors are functionally similar, they turn out to be very different in overall shape and architecture and in the location of key functional elements.


Asunto(s)
Factores de Terminación de Péptidos/química , Humanos , Modelos Moleculares , Factores de Terminación de Péptidos/metabolismo , Estructura Terciaria de Proteína , Ribosomas/metabolismo
13.
Oncogene ; 23(34): 5719-28, 2004 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-15208675

RESUMEN

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.


Asunto(s)
Neoplasias de la Mama/genética , Carcinoma/genética , Neoplasias Renales/genética , Neoplasias Pulmonares/genética , Eliminación de Secuencia , Canales de Calcio/genética , Carcinoma de Células Renales/genética , Carcinoma de Células Pequeñas/genética , Aberraciones Cromosómicas , Cromosomas Humanos Par 3 , Femenino , Reordenamiento Génico , Marcadores Genéticos , Homocigoto , Humanos , Pérdida de Heterocigocidad , Glicoproteínas de Membrana/genética , Proteínas de la Membrana/genética , Proteínas del Tejido Nervioso/genética , Reacción en Cadena de la Polimerasa/métodos , Semaforinas , Células Tumorales Cultivadas
14.
J Mol Biol ; 331(4): 745-58, 2003 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-12909007

RESUMEN

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.


Asunto(s)
Codón de Terminación/genética , Codón de Terminación/metabolismo , Factores de Terminación de Péptidos/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ribosomas/química , Ribosomas/metabolismo , Secuencia de Bases , Sitios de Unión , Reactivos de Enlaces Cruzados , Humanos , Sustancias Macromoleculares , Modelos Biológicos , Peso Molecular , Oligonucleótidos/genética , Oligonucleótidos/metabolismo , Unión Proteica , Biosíntesis de Proteínas
15.
Gene ; 283(1-2): 209-17, 2002 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-11867227

RESUMEN

We have identified a novel human gene UNC93B1 encoding a protein related to unc-93 of Caenorhabditis elegans. The combined sequence derived from several cDNA clones is 2282 bp and comparison with genomic sequence shows that the gene contains 11 exons. The longest open reading frame encodes a deduced sequence of 597 amino acids. Homology analysis shows that the hUNC93B1 gene is highly conserved and related to sequences in Arabidopsis thaliana, C. elegans, Drosophila melanogaster, chicken and mouse. Structural analysis of the deduced amino acid sequence of hUNC93B1 points to possible existence of multiple membrane-spanning domains. hUNC93B1 protein also displays some similarities to the bacterial ABC-2 type transporter signature and to ion transporters of Deinococcus radiodurans and Helicobacter pylori. As revealed by Northern analysis, the level of expression varies significantly between tissues, with the highest level detected in the heart. The gene was mapped to chromosomal band 11q13 by fluorescence in situ hybridization. We suggest that this gene is a member of a novel hUNC93B-related gene family.


Asunto(s)
Proteínas de Caenorhabditis elegans , Genes/genética , Proteínas de Transporte de Membrana/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Caenorhabditis elegans/genética , Mapeo Cromosómico , Cromosomas Humanos Par 11/genética , ADN/genética , ADN Complementario/química , ADN Complementario/genética , Femenino , Proteínas del Helminto/genética , Humanos , Hibridación Fluorescente in Situ , Proteínas de la Membrana/genética , Datos de Secuencia Molecular , Alineación de Secuencia , Análisis de Secuencia de ADN
16.
FEBS Lett ; 548(1-3): 97-102, 2003 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-12885414

RESUMEN

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.


Asunto(s)
Codón de Terminación , Codón , ARN Ribosómico 18S/metabolismo , Azidas , Secuencia de Bases , Sitios de Unión , Reactivos de Enlaces Cruzados , Fosfatos de Dinucleósidos , Oligorribonucleótidos/metabolismo , Biosíntesis de Proteínas/genética , ARN Bacteriano/metabolismo , ARN Mensajero/metabolismo , ARN Ribosómico 18S/química , Aminoacil-ARN de Transferencia , ARN de Transferencia de Fenilalanina
17.
FEBS Lett ; 514(1): 96-101, 2002 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-11904189

RESUMEN

To study positioning of the mRNA stop signal with respect to polypeptide chain release factors (RFs) and ribosomal components within human 80S ribosomes, photoreactive mRNA analogs were applied. Derivatives of the UUCUAAA heptaribonucleotide containing the UUC codon for Phe and the stop signal UAAA, which bore a perfluoroaryl azido group at either the fourth nucleotide or the 3'-terminal phosphate, were synthesized. The UUC codon was directed to the ribosomal P site by the cognate tRNA(Phe), targeting the UAA stop codon to the A site. Mild UV irradiation of the ternary complexes consisting of the 80S ribosome, the mRNA analog and tRNA resulted in tRNA-dependent crosslinking of the mRNA analogs to the 40S ribosomal proteins and the 18S rRNA. mRNA analogs with the photoreactive group at the fourth uridine (the first base of the stop codon) crosslinked mainly to protein S15 (and much less to S2). For the 3'-modified mRNA analog, the major crosslinking target was protein S2, while protein S15 was much less crosslinked. Crosslinking of eukaryotic (e) RF1 was entirely dependent on the presence of a stop signal in the mRNA analog. eRF3 in the presence of eRF1 did not crosslink, but decreased the yield of eRF1 crosslinking. We conclude that (i) proteins S15 and S2 of the 40S ribosomal subunit are located near the A site-bound codon; (ii) eRF1 can induce spatial rearrangement of the 80S ribosome leading to movement of protein L4 of the 60S ribosomal subunit closer to the codon located at the A site; (iii) within the 80S ribosome, eRF3 in the presence of eRF1 does not contact the stop codon at the A site and is probably located mostly (if not entirely) on the 60S subunit.


Asunto(s)
Codón de Terminación/metabolismo , Factores de Terminación de Péptidos/metabolismo , Proteínas Ribosómicas/metabolismo , Ribosomas/metabolismo , Factor 1 Eucariótico de Iniciación/química , Factor 1 Eucariótico de Iniciación/metabolismo , Humanos , ARN Mensajero/síntesis química , ARN Mensajero/química , ARN Mensajero/metabolismo
18.
FEBS Lett ; 583(21): 3455-60, 2009 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-19796638

RESUMEN

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.


Asunto(s)
Lisina/análogos & derivados , Methanosarcinaceae/genética , Methanosarcinaceae/metabolismo , Biosíntesis de Proteínas , Secuencia de Aminoácidos , Proteínas Arqueales/química , Proteínas Arqueales/metabolismo , Codón de Terminación/genética , Codón de Terminación/metabolismo , Genoma Arqueal , Humanos , Lisina/metabolismo , Datos de Secuencia Molecular , Factores de Terminación de Péptidos/química , Factores de Terminación de Péptidos/metabolismo , Filogenia
19.
PLoS One ; 4(5): e5231, 2009 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-19478941

RESUMEN

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.


Asunto(s)
Mutación/genética , Neoplasias/genética , Proteínas Supresoras de Tumor/genética , Desaminasas APOBEC-1 , Animales , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Línea Celular , Línea Celular Tumoral , Proliferación Celular , Células Clonales , Biología Computacional , Citidina Desaminasa/metabolismo , ADN Bacteriano/genética , ADN Complementario/genética , Proteínas de Escherichia coli/genética , Etiquetas de Secuencia Expresada , Efecto Fundador , Genoma/genética , Hematopoyesis/genética , Humanos , Neoplasias Renales/genética , Neoplasias Renales/patología , Ratones , Ratones SCID , Reacción en Cadena de la Polimerasa
20.
Biomol NMR Assign ; 1(2): 183-5, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19636860

RESUMEN

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.


Asunto(s)
Espectroscopía de Resonancia Magnética/métodos , Factores de Terminación de Péptidos/química , Secuencia de Aminoácidos , Humanos , Estructura Terciaria de Proteína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA