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










Base de dados
Intervalo de ano de publicação
1.
ACS Chem Biol ; 18(4): 794-802, 2023 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-37005433

RESUMO

Pseudouridimycin is a microbial C-nucleoside natural product that specifically inhibits bacterial RNA polymerases by binding to the active site and competing with uridine triphosphate for the nucleoside triphosphate (NTP) addition site. Pseudouridimycin consists of 5'-aminopseudouridine and formamidinylated, N-hydroxylated Gly-Gln dipeptide moieties to allow Watson-Crick base pairing and to mimic protein-ligand interactions of the triphosphates of NTP, respectively. The metabolic pathway of pseudouridimycin has been studied in Streptomyces species, but no biosynthetic steps have been characterized biochemically. Here, we show that the flavin-dependent oxidase SapB functions as a gate-keeper enzyme selecting pseudouridine (KM = 34 µM) over uridine (KM = 901 µM) in the formation of pseudouridine aldehyde. The pyridoxal phosphate (PLP)-dependent SapH catalyzes transamination, resulting in 5'-aminopseudouridine with a preference for arginine, methionine, or phenylalanine as cosubstrates as amino group donors. The binary structure of SapH in complex with pyridoxamine-5'-phosphate and site-directed mutagenesis identified Lys289 and Trp32 as key residues for catalysis and substrate binding, respectively. The related C-nucleoside oxazinomycin was accepted as a substrate by SapB with moderate affinity (KM = 181 µM) and was further converted by SapH, which opens possibilities for metabolic engineering to generate hybrid C-nucleoside pseudouridimycin analogues in Streptomyces.


Assuntos
Nucleosídeos , Pseudouridina , Vias Biossintéticas , RNA Polimerases Dirigidas por DNA/metabolismo , Nucleosídeos/metabolismo , Pseudouridina/biossíntese , Pseudouridina/metabolismo , Fosfato de Piridoxal/química , Streptomyces/química , Streptomyces/metabolismo
2.
J Mol Biol ; 434(12): 167588, 2022 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-35439479

RESUMO

The fidelity of initiator tRNA (i-tRNA) selection in the ribosomal P-site is a key step in translation initiation. The highly conserved three consecutive G:C base pairs (3GC pairs) in the i-tRNA anticodon stem play a crucial role in its selective binding in the P-site. Mutations in the 3GC pairs (3GC mutant) render the i-tRNA inactive in initiation. Here, we show that a mutation (E265K) in the unique C-terminal tail domain of RluD, a large ribosomal subunit pseudouridine synthase, results in compromised fidelity of initiation and allows initiation with the 3GC mutant i-tRNA. RluD modifies the uridine residues in H69 to pseudouridines. However, the role of its C-terminal tail domain remained unknown. The E265K mutation does not diminish the pseudouridine synthase activity of RluD, or the growth phenotype of Escherichia coli, or cause any detectable defects in the ribosomal assembly in our assays. However, in our in vivo analyses, we observed that the E265K mutation resulted in increased retention of the ribosome binding factor A (RbfA) on 30S suggesting a new role of RluD in contributing to RbfA release, a function which may be attributed to its (RluD) C-terminal tail domain. The studies also reveal that deficiency of RbfA release from 30S compromises the fidelity of i-tRNA selection in the ribosomal P-site.


Assuntos
Proteínas de Escherichia coli , Escherichia coli , Iniciação Traducional da Cadeia Peptídica , Proteínas Ribossômicas , Anticódon/genética , Anticódon/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Hidroliases/química , Mutação , Pseudouridina/biossíntese , RNA de Transferência de Metionina/genética , RNA de Transferência de Metionina/metabolismo , Proteínas Ribossômicas/genética , Proteínas Ribossômicas/metabolismo
3.
Urol Oncol ; 39(1): 63-71, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32712138

RESUMO

Epitranscriptomic analysis has recently led to the profiling of modified nucleosides in cancer cell biological matrices, helping to elucidate their functional roles in cancer and reigniting interest in exploring their use as potential markers of cancer development and progression. Pseudouridine, one of the most well-known and the most abundant of the RNA nucleotide modifications, is the C5-glycoside isomer of uridine and its distinctive physiochemical properties allows it to perform many essential functions. Pseudouridine functionally (a) confers rigidity to local RNA structure by enhancing RNA stacking, engaging in a cooperative effect on neighboring nucleosides that overall contributes to RNA stabilization (b) refines the structure of tRNAs, which influences their decoding activity (c) facilitates the accuracy of decoding and proofreading during translation and efficiency of peptide bond formation, thus collectively improving the fidelity of protein biosynthesis and (e) dynamically regulates mRNA coding and translation. Biochemical synthesis of pseudouridine is carried out by pseudouridine synthases. In this review we discuss the evidence supporting an association between elevated pseudouridine levels with the incidence and progression of human prostate cancer and the translational significance of the value of this modified nucleotide as a novel biomarker in prostate cancer progression to advanced disease.


Assuntos
Biomarcadores Tumorais/análise , Neoplasias da Próstata/química , Neoplasias da Próstata/metabolismo , Pseudouridina/análise , Previsões , Humanos , Masculino , Pseudouridina/biossíntese , Pseudouridina/química , Pseudouridina/fisiologia
4.
RNA ; 25(3): 336-351, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30530625

RESUMO

The nearly conserved U54 of tRNA is mostly converted to a version of ribothymidine (T) in Bacteria and eukaryotes and to a version of pseudouridine (Ψ) in Archaea. Conserved U55 is nearly always modified to Ψ55 in all organisms. Orthologs of TrmA and TruB that produce T54 and Ψ55, respectively, in Bacteria and eukaryotes are absent in Archaea. Pus10 produces both Ψ54 and Ψ55 in Archaea. Pus10 orthologs are found in nearly all sequenced archaeal and most eukaryal genomes, but not in yeast and bacteria. This coincides with the presence of Ψ54 in most archaeal tRNAs and some animal tRNAs, but its absence from yeast and bacteria. Moreover, Ψ54 is found in several tRNAs that function as primers for retroviral DNA synthesis. Previously, no eukaryotic tRNA Ψ54 synthase had been identified. We show here that human Pus10 can produce Ψ54 in select tRNAs, including tRNALys3, the primer for HIV reverse transcriptase. This synthase activity of Pus10 is restricted to the cytoplasm and is distinct from nuclear Pus10, which is known to be involved in apoptosis. The sequence GUUCAm1AAUC (m1A is 1-methyladenosine) at position 53-61 of tRNA along with a stable acceptor stem results in maximum Ψ54 synthase activity. This recognition sequence is unique for a Ψ synthase in that it contains another modification. In addition to Ψ54, SF9 cells-derived recombinant human Pus10 can also generate Ψ55, even in tRNAs that do not contain the Ψ54 synthase recognition sequence. This activity may be redundant with that of TruB.


Assuntos
Proteínas Arqueais/genética , Proteínas Arqueais/metabolismo , Hidroliases/genética , Hidroliases/metabolismo , Pseudouridina/biossíntese , RNA de Transferência/genética , Animais , Archaea/genética , Archaea/metabolismo , Sequência de Bases , Citoplasma , Ativação Enzimática , Humanos , Mamíferos/genética , Proteólise , Processamento Pós-Transcricional do RNA , RNA Arqueal , Proteínas Recombinantes , Elementos de Resposta , Seleção Genética
5.
Biochimie ; 113: 134-42, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25896443

RESUMO

The box H/ACA small ribonucleoprotein particles (H/ACA sRNPs) are RNP enzymes that isomerize uridines (U) into pseudouridines (Ψ) in archaeal RNAs. The RNA component acts as a guide by forming base-pair interactions with the substrate RNA to specify the target nucleotide of the modification to the catalytic subunit Cbf5. Here, we have analyzed association of an H/ACA sRNP enzyme from the hyperthermophilic archaeon Pyrococcus abyssi with synthetic substrate RNAs of different length and with target nucleotide variants, and estimated their turnover at high temperature. In these conditions, we found that a short substrate, which length is restricted to the interaction with RNA guide sequence, has higher turnover rate. However, the longer substrate with additional 5' and 3' sequences non-complementary to the guide RNA is better discriminated by the U to Ψ conversion allowing the RNP enzyme to distinguish the modified product from the substrate. In addition, we identified that the conserved residue Y179 in the catalytic center of Cbf5 is crucial for substrate selectivity.


Assuntos
Proteínas Arqueais/metabolismo , Pseudouridina/biossíntese , Pyrococcus abyssi/metabolismo , RNA Arqueal/metabolismo , Ribonucleoproteínas/metabolismo , Proteínas Arqueais/química , Proteínas Arqueais/genética , Pyrococcus abyssi/química , Pyrococcus abyssi/genética , RNA Arqueal/química , RNA Arqueal/genética , Ribonucleoproteínas/química , Ribonucleoproteínas/genética , Especificidade por Substrato/fisiologia
6.
J Mol Biol ; 425(20): 3863-74, 2013 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-23743107

RESUMO

Pus10 is the most recently identified pseudouridine synthase found in archaea and higher eukaryotes. It modifies uridine 55 in the TΨC arm of tRNAs. Here, we report the first quantitative biochemical analysis of tRNA binding and pseudouridine formation by Pyrococcus furiosus Pus10. The affinity of Pus10 for both substrate and product tRNA is high (Kd of 30nM), and product formation occurs with a Km of 400nM and a kcat of 0.9s(-1). Site-directed mutagenesis was used to demonstrate that the thumb loop in the catalytic domain is important for efficient catalysis; we propose that the thumb loop positions the tRNA within the active site. Furthermore, a new catalytic arginine residue was identified (arginine 208), which is likely responsible for triggering flipping of the target uridine into the active site of Pus10. Lastly, our data support the proposal that the THUMP-containing domain, found in the N-terminus of Pus10, contributes to binding of tRNA. Together, our findings are consistent with the hypothesis that tRNA binding by Pus10 occurs through an induced-fit mechanism, which is a prerequisite for efficient pseudouridine formation.


Assuntos
Hidroliases/metabolismo , RNA de Transferência/genética , RNA de Transferência/metabolismo , Catálise , Dicroísmo Circular , Hidroliases/química , Hidroliases/genética , Cinética , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Pseudouridina/biossíntese , Pyrococcus furiosus/genética , Pyrococcus furiosus/metabolismo , RNA de Transferência/química
7.
J Mol Biol ; 425(20): 3875-87, 2013 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-23707380

RESUMO

Human pseudouridine (Ψ) synthase Pus1 (hPus1) modifies specific uridine residues in several non-coding RNAs: tRNA, U2 spliceosomal RNA, and steroid receptor activator RNA. We report three structures of the catalytic core domain of hPus1 from two crystal forms, at 1.8Å resolution. The structures are the first of a mammalian Ψ synthase from the set of five Ψ synthase families common to all kingdoms of life. hPus1 adopts a fold similar to bacterial Ψ synthases, with a central antiparallel ß-sheet flanked by helices and loops. A flexible hinge at the base of the sheet allows the enzyme to open and close around an electropositive active-site cleft. In one crystal form, a molecule of Mes [2-(N-morpholino)ethane sulfonic acid] mimics the target uridine of an RNA substrate. A positively charged electrostatic surface extends from the active site towards the N-terminus of the catalytic domain, suggesting an extensive binding site specific for target RNAs. Two α-helices C-terminal to the core domain, but unique to hPus1, extend along the back and top of the central ß-sheet and form the walls of the RNA binding surface. Docking of tRNA to hPus1 in a productive orientation requires only minor conformational changes to enzyme and tRNA. The docked tRNA is bound by the electropositive surface of the protein employing a completely different binding mode than that seen for the tRNA complex of the Escherichia coli homologue TruA.


Assuntos
Hidroliases/metabolismo , RNA de Transferência/metabolismo , Bactérias/genética , Bactérias/metabolismo , Domínio Catalítico , Humanos , Hidroliases/química , Modelos Moleculares , Simulação de Acoplamento Molecular , Ligação Proteica , Conformação Proteica , Dobramento de Proteína , Domínios e Motivos de Interação entre Proteínas , Pseudouridina/biossíntese , RNA de Transferência/química
8.
Sci Rep ; 2: 663, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22993689

RESUMO

Cbf5 is a pseudouridine synthase that usually acts in a guide RNA-dependent manner as part of H/ACA small ribonucleoproteins; however archaeal Cbf5 can also act independently of guide RNA in modifying uridine 55 in tRNA. This guide-independent activity of Cbf5 is enhanced by proteins Nop10 and Gar1 which are also found in H/ACA small ribonucleoproteins. Here, we analyzed the specific contribution of Nop10 and Gar1 for Cbf5-catalyzed pseudouridylation of tRNA. Interestingly, both Nop10 and Gar1 not only increase Cbf5's affinity for tRNA, but they also directly enhance Cbf5's catalytic activity by increasing the k(cat) of the reaction. In contrast to the guide RNA-dependent reaction, Gar1 is not involved in product release after tRNA modification. These results in conjunction with structural information suggest that Nop10 and Gar1 stabilize Cbf5 in its active conformation; we hypothesize that this might also be true for guide-RNA dependent pseudouridine formation by Cbf5.


Assuntos
Proteínas Arqueais/química , Transferases Intramoleculares/química , Pyrococcus furiosus/enzimologia , RNA de Transferência/química , Ribonucleoproteínas Nucleolares Pequenas/química , Estabilidade Enzimática , Cinética , Ligação Proteica , Pseudouridina/biossíntese , Pequeno RNA não Traduzido
9.
RNA ; 16(3): 610-20, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20106954

RESUMO

Pseudouridine is the most abundant of more than 100 chemically distinct natural ribonucleotide modifications. Its synthesis consists of an isomerization reaction of a uridine residue in the RNA chain and is catalyzed by pseudouridine synthases. The unusual reaction mechanism has become the object of renewed research effort, frequently involving replacement of the substrate uridines with 5-fluorouracil (f(5)U). f(5)U is known to be a potent inhibitor of pseudouridine synthase activity, but the effect varies among the target pseudouridine synthases. Derivatives of f(5)U have previously been detected, which are thought to be either hydrolysis products of covalent enzyme-RNA adducts, or isomerization intermediates. Here we describe the interaction of pseudouridine synthase 1 (Pus1p) with f(5)U-containing tRNA. The interaction described is specific to Pus1p and position 27 in the tRNA anticodon stem, but the enzyme neither forms a covalent adduct nor stalls at a previously identified reaction intermediate of f(5)U. The f(5)U27 residue, as analyzed by a DNAzyme-based assay using TLC and mass spectrometry, displayed physicochemical properties unaltered by the reversible interaction with Pus1p. Thus, Pus1p binds an f(5)U-containing substrate, but, in contrast to other pseudouridine synthases, leaves the chemical structure of f(5)U unchanged. The specific, but nonproductive, interaction demonstrated here thus constitutes an intermediate of Pus turnover, stalled by the presence of f(5)U in an early state of catalysis. Observation of the interaction of Pus1p with fluorescence-labeled tRNA by a real-time readout of fluorescence anisotropy and FRET revealed significant structural distortion of f(5)U-tRNA structure in the stalled intermediate state of pseudouridine catalysis.


Assuntos
Fluoruracila/química , Hidroliases/química , Pseudouridina/biossíntese , Animais , Transferência Ressonante de Energia de Fluorescência , Humanos , Camundongos , Processamento Pós-Transcricional do RNA , RNA de Transferência de Leucina/química , Saccharomyces cerevisiae/enzimologia
10.
Anal Chim Acta ; 623(2): 117-25, 2008 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-18620915

RESUMO

Pseudouridine, the so-called fifth nucleoside due to its ubiquitous presence in ribonucleic acids (RNAs), remains among the most challenging modified nucleosides to characterize. As an isomer of the major nucleoside uridine, pseudouridine cannot be detected by standard reverse-transcriptase-based DNA sequencing or RNase mapping approaches. Thus, over the past 15 years, investigators have focused on the unique structural properties of pseudouridine to develop selective derivatization or fragmentation strategies for its determination. While the N-cyclohexyl-N'-beta-(4-methylmorpholinium)ethylcarbodiimide p-tosylate (CMCT)-reverse transcriptase assay remains both a popular and powerful approach to screen for pseudouridine in larger RNAs, mass-spectrometry-based approaches are poised to play an increasingly important role in either confirming the findings of the CMCT-reverse transcriptase assay or in characterizing pseudouridine sequence placement and abundance in smaller RNAs. This review includes a brief discussion of pseudouridine including a summary of its biosynthesis and known importance within various RNAs. The review then focuses on chemical derivatization approaches that can be used to selectively modify pseudouridine to improve its detection, and the development of mass-spectrometry-based assays for the identification and sequencing of pseudouridine in various RNAs.


Assuntos
Espectrometria de Massas/métodos , Pseudouridina/análise , Pseudouridina/química , RNA/química , Pseudouridina/biossíntese , Sensibilidade e Especificidade
11.
RNA Biol ; 4(2): 101-9, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17993784

RESUMO

RNA-guided pseudouridine (Psi) synthesis in Archaea and Eukarya requires a four-protein one-RNA containing box H/ACA ribonucleoprotein (RNP) complex. The proteins in the archaeal RNP are aCbf5, aNop10, aGar1 and L7Ae. Pyrococcus aCbf5-aNop10 is suggested to be the minimal catalytic core in this synthesis and the activity is enhanced by L7Ae and aGar1. The protein aCbf5 is homologous to eukaryal Cbf5 (dyskerin, NAP57) as well as to bacterial TruB and eukaryal Pus4; the last two produce YPsi55 in tRNAs in a guide RNA-independent manner. Here, using recombinant Methanocaldococcus jannaschii proteins, we report that aCbf5 and aGar1 together can function as a tRNA Psi55 synthase in a guide RNA-independent manner. This activity is enhanced by aNop10, but not by L7Ae. The aCbf5 alone can also produce Psi55 in tRNAs that contain the canonical 3'-CCA sequence and this activity is stimulated by aGar1. These results suggest that the roles of accessory proteins are different in guide RNA-dependent and independent Psi synthesis by aCbf5. The presence of conserved C (or U) and A at tRNA positions 56 and 58, respectively, which are required for TruB/Pus4 activity, is not essential for aCbf5-mediated Psi55 formation. Conserved A58 in tRNA normally forms a tertiary reverse Hoogstein base pair with an equally conserved U54. This base pair is recognized by TruB. Apparently aCbf5 does not require this base pair to recognize U55 for conversion to Psi55.


Assuntos
Proteínas Arqueais/química , Proteínas Arqueais/metabolismo , Mathanococcus/metabolismo , Pseudouridina/biossíntese , Liases Intramoleculares/metabolismo , Transferases Intramoleculares/metabolismo , Cinética , Conformação de Ácido Nucleico , RNA Arqueal/metabolismo , RNA de Transferência/química , Proteínas Recombinantes/metabolismo , Ribonucleoproteínas , Pequeno RNA não Traduzido
12.
FEBS J ; 274(21): 5759-66, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17937767

RESUMO

Pseudouridine synthase RluD converts uridines at positions 1911, 1915, and 1917 of 23S rRNA to pseudouridines. These nucleotides are located in the functionally important helix-loop 69 of 23S rRNA. RluD is the only pseudouridine synthase that is required for normal growth in Escherichia coli. We have analyzed substrate specificity of RluD in vivo. Mutational analyses have revealed: (a) RluD isomerizes uridine in vivo only at positions 1911, 1915, and 1917, regardless of the presence of uridine at other positions in the loop of helix 69 of 23S rRNA variants; (b) substitution of one U by C has no effect on the conversion of others (i.e. formation of pseudouridines at positions 1911, 1915, and 1917 are independent of each other); (c) A1916 is the only position in the loop of helix 69, where mutations affect the RluD specific pseudouridine formation. Pseudouridines were determined in the ribosomal particles from a ribosomal large subunit defective strain (RNA helicase DeaD(-)). An absence of pseudouridines in the assembly precursor particles suggests that RluD directed isomerization of uridines occurs as a late step during the assembly of the large ribosomal subunit.


Assuntos
Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Escherichia coli/enzimologia , Hidroliases/química , Hidroliases/metabolismo , Pseudouridina/biossíntese , Sequência de Bases , Dados de Sequência Molecular , Conformação de Ácido Nucleico , Mutação Puntual , RNA Ribossômico 23S/genética , RNA Ribossômico 23S/metabolismo , Especificidade por Substrato
13.
Curr Opin Struct Biol ; 17(3): 287-92, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17574834

RESUMO

H/ACA guide RNAs direct site-specific pseudouridylation of substrate RNAs by forming ribonucleoprotein (RNP) complexes with pseudouridine synthase Cbf5 and three accessory proteins. Recently determined crystal structures of H/ACA protein complexes and a fully assembled H/ACA RNP complex have provided significant insights into the architecture, assembly and mechanism of action of RNA-guided pseudouridine synthase. The binding of guide RNA is directed by its conserved secondary structure and sequence motifs, which enables guide RNA with different sequences to be incorporated into the same protein complex. Accessory proteins and peripheral domains crucially coordinate the position of guide RNA, and possibly regulate the reaction process.


Assuntos
Ribonucleoproteínas/fisiologia , Animais , Humanos , Pseudouridina/biossíntese , Ribonucleoproteínas/química , Pequeno RNA não Traduzido
14.
RNA ; 11(6): 928-38, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15923376

RESUMO

Conversion of uridines into pseudouridines (Psis) is the most frequent base modification in ribosomal RNAs (rRNAs). In eukaryotes, the pseudouridylation sites are specified by base-pairing with specific target sequences within H/ACA small nucleolar RNAs (snoRNAs). The yeast rRNAs harbor 44 Psis, but, when this work began, 15 Psis had completely unknown guide snoRNAs. This suggested that many snoRNAs remained to be discovered. To address this problem and further complete the snoRNA assignment to Psi sites, we identified the complete set of RNAs associated with the H/ACA snoRNP specific proteins Gar1p and Nhp2p by coupling TAP-tag purifications with genomic DNA microarrays experiments. Surprisingly, while we identified all the previously known H/ACA snoRNAs, we selected only three new snoRNAs. This suggested that most of the missing Psi guides were present in previously known snoRNAs but had been overlooked. We confirmed this hypothesis by systematically investigating the role of previously known, as well as of the newly identified snoRNAs, in specifying rRNA Psi sites and found all but one missing guide RNAs. During the completion of this work, another study, based on bioinformatic predictions, also reported the identification of most missing guide RNAs. Altogether, all Psi guides are now identified and we can tell that, in budding yeast, the 44 Psis are guided by 28 snoRNAs. Finally, aside from snR30, an atypical small RNA of heterogeneous length and at least one mRNA, all Gar1p and Nhp2p associated RNAs characterized by our work turned out to be snoRNAs involved in rRNA Psi specification.


Assuntos
Proteínas Nucleares/metabolismo , Pseudouridina/biossíntese , RNA Fúngico/metabolismo , RNA Ribossômico/metabolismo , RNA Nucleolar Pequeno/análise , Ribonucleoproteínas Nucleares Pequenas/metabolismo , Ribonucleoproteínas Nucleolares Pequenas/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/genética , Sequência de Bases , Genoma Fúngico , Dados de Sequência Molecular , Mutação , Proteínas Nucleares/genética , Conformação de Ácido Nucleico , Análise de Sequência com Séries de Oligonucleotídeos , RNA Fúngico/análise , RNA Ribossômico/genética , RNA Nucleolar Pequeno/genética , Ribonucleoproteínas Nucleares Pequenas/genética , Ribonucleoproteínas Nucleolares Pequenas/genética , Proteínas de Saccharomyces cerevisiae/genética
15.
J Am Chem Soc ; 126(40): 12758-9, 2004 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-15469254

RESUMO

RNA containing 5-fluorouridine, [f 5U]RNA, has been used as a mechanistic probe for the pseudouridine synthases, which convert uridine in RNA to its C-glycoside isomer, pseudouridine. Hydrated products of f 5U were attributed to ester hydrolysis of a covalent complex between an essential aspartic acid residue and f 5U, and the results were construed as strong support for a mechanism involving Michael addition by the aspartic acid residue. Labeling studies with [18O]water are now reported that rule out such ester hydrolysis in one pseudouridine synthase, TruB. The aspartic acid residue does not become labeled, and the hydroxyl group in the hydrated product of f 5U derives directly from solvent. The hydrated product, therefore, cannot be construed to support Michael addition during the conversion of uridine to pseudouridine, but the results do not rule out such a mechanism. A hypothesis is offered for the seemingly disparate behavior of different pseudouridine synthases toward [f 5U]RNA.


Assuntos
Hidroliases/metabolismo , Pseudouridina/biossíntese , Uridina/análogos & derivados , Escherichia coli/enzimologia , Escherichia coli/genética , Hidroliases/genética , Hidrólise , RNA/metabolismo , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Uridina/metabolismo
16.
Mol Cell Biol ; 24(13): 5797-807, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15199136

RESUMO

Pseudouridine, the most abundant modified nucleoside in RNA, is synthesized by posttranscriptional isomerization of uridines. In eukaryotic RNAs, site-specific synthesis of pseudouridines is directed primarily by box H/ACA guide RNAs. In this study, we have identified 61 novel putative pseudouridylation guide RNAs by construction and characterization of a cDNA library of human box H/ACA RNAs. The majority of the new box H/ACA RNAs are predicted to direct pseudouridine synthesis in rRNAs and spliceosomal small nuclear RNAs. We can attribute RNA-directed modification to 79 of the 97 pseudouridylation sites present in the human 18S, 5.8S, and 28S rRNAs and to 11 of the 21 pseudouridines reported for the U1, U2, U4, U5, and U6 spliceosomal RNAs. We have also identified 12 novel box H/ACA RNAs which lack apparent target pseudouridines in rRNAs and small nuclear RNAs. These putative guide RNAs likely function in the pseudouridylation of some other types of cellular RNAs, suggesting that RNA-guided pseudouridylation is more general than assumed before. The genomic organization of the new box H/ACA RNA genes indicates that in human cells, all box H/ACA pseudouridylation guide RNAs are processed from introns of pre-mRNA transcripts which either encode a protein product or lack protein-coding capacity.


Assuntos
Pseudouridina/biossíntese , Processamento Pós-Transcricional do RNA , Biblioteca Gênica , Genes de RNAr , Genoma Humano , Humanos , Íntrons , Substâncias Macromoleculares , Precursores de RNA/metabolismo , RNA Ribossômico/biossíntese , Pequeno RNA não Traduzido
17.
RNA ; 9(6): 711-21, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12756329

RESUMO

Putative pseudouridine synthase genes are members of a class consisting of four subgroups that possess characteristic amino acid sequence motifs. These genes have been found in all organisms sequenced to date. In Escherichia coli, 10 such genes have been identified, and the 10 synthase gene products have been shown to function in making all of the pseudouridines found in tRNA and ribosomal RNA except for tRNA(Glu) pseudouridine13. In this work, a protein able to make this pseudouridine was purified by standard biochemical procedures. Amino-terminal sequencing of the isolated protein identified the synthase as YgbO. Deletion of the ygbO gene caused the loss of tRNA(Glu) pseudouridine13 and plasmid-borne restoration of the structural gene restored pseudouridine13. Reaction of the overexpressed gene product, renamed TruD, with a tRNA(Glu) transcript made in vitro also yielded only pseudouridine13. A search of the database detected 58 homologs of TruD spanning all three phylogenetic domains, including ancient organisms. Thus, we have identified a new wide-spread class of pseudouridine synthase with no sequence homology to the previously known four subgroups. The only completely conserved sequence motif in all 59 organisms that contained aspartate was GXKD, in motif II. This aspartate was essential for in vitro activity.


Assuntos
Archaea/enzimologia , Bactérias/enzimologia , Proteínas de Escherichia coli/classificação , Células Eucarióticas/enzimologia , Hidroliases/classificação , Motivos de Aminoácidos , Sequência de Aminoácidos , Ácido Aspártico/fisiologia , Sequência de Bases , Escherichia coli/enzimologia , Escherichia coli/genética , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/fisiologia , Deleção de Genes , Genes Bacterianos , Genoma , Hidroliases/química , Hidroliases/genética , Hidroliases/fisiologia , Dados de Sequência Molecular , Pseudouridina/análise , Pseudouridina/biossíntese , RNA de Transferência de Ácido Glutâmico/biossíntese , RNA de Transferência de Ácido Glutâmico/química , Análise de Sequência de RNA , Homologia de Sequência de Aminoácidos , Especificidade por Substrato
18.
Biochim Biophys Acta ; 1574(2): 137-44, 2002 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-11955622

RESUMO

In order to establish the structure and sequence requirements for pseudouridine (Psi(35)) biosynthesis in Arabidopsis thaliana tRNA(Tyr) five mutants of nuclear pre-tRNA(Tyr) have been prepared and analyzed: DeltaI-tRNA(Tyr) transcript depleted of an intron, and 5UI, 7UI, 9UI and 12UI transcripts containing tracts of five, seven, nine and 12 U residues, respectively, instead of the wild type tRNA(Tyr) intron. The in vitro transcripts were incubated in a lupin seed extract containing Psi(35) synthase activity, and those containing an artificial intron composed of 12 or nine U residues turned out to be good substrates for Psi(35) synthase. The transcript with an intron composed of seven uridine residues was pseudouridylated up to 40%, whereas the remaining two were not pseudouridylated at all. The secondary structures of all transcripts were determined using enzymatic and chemical probes: S(1), V(1), T(1), A, P(1) and Pb(2+). All mutant pre-tRNAs show similar structural features: their anticodon arm contains a five base pair stem and a large loop which consists of five natural tRNA(Tyr) AC loop nucleotides to which five, seven, nine and 12 U residues are added. As the structure of the wild type pre-tRNA(Tyr) is different we propose that the role of its intron in the process of U(35) pseudouridylation is simply to expand the anticodon region to the required critical length.


Assuntos
Arabidopsis/metabolismo , Transferases Intramoleculares/metabolismo , Pseudouridina/genética , Precursores de RNA/metabolismo , RNA de Transferência de Tirosina/metabolismo , Anticódon , Arabidopsis/genética , Estrutura Molecular , Mutação , Pseudouridina/biossíntese , Pseudouridina/química , Precursores de RNA/genética , RNA de Transferência de Tirosina/genética
19.
FEBS Lett ; 514(1): 17-25, 2002 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-11904174

RESUMO

Pseudouridines are found in virtually all ribosomal RNAs but their function is unknown. There are four to eight times more pseudouridines in eukaryotes than in eubacteria. Mapping 19 Haloarcula marismortui pseudouridines on the three-dimensional 50S subunit does not show clustering. In bacteria, specific enzymes choose the site of pseudouridine formation. In eukaryotes, and probably also in archaea, selection and modification is done by a guide RNA-protein complex. No unique specific role for ribosomal pseudouridines has been identified. We propose that pseudouridine's function is as a molecular glue to stabilize required RNA conformations that would otherwise be too flexible.


Assuntos
Pseudouridina/biossíntese , RNA Ribossômico/análise , Animais , Archaea/metabolismo , Eubacterium/metabolismo , Células Eucarióticas , Humanos , Modelos Moleculares , Conformação de Ácido Nucleico , Pseudouridina/metabolismo , RNA Ribossômico/química , Pequeno RNA não Traduzido
20.
RNA ; 7(11): 1603-15, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11720289

RESUMO

There are 10 known putative pseudouridine synthase genes in Escherichia coli. The products of six have been previously assigned, one to formation of the single pseudouridine in 16S RNA, three to the formation of seven pseudouridines in 23S RNA, and three to the formation of three pseudouridines in tRNA (one synthase makes pseudouridine in 23S RNA and tRNA). Here we show that the remaining four putative synthase genes make bona fide pseudouridine synthases and identify which pseudouridines they make. RluB (formerly YciL) and RluE (formerly YmfC) make pseudouridine2605 and pseudouridine2457, respectively, in 23S RNA. RluF (formerly YjbC) makes the newly discovered pseudouridine2604 in 23S RNA, and TruC (formerly YqcB) makes pseudouridine65 in tRNA(Ile1) and tRNA(Asp). Deletion of each of these synthase genes individually had no effect on exponential growth in rich media at 25 degrees C, 37 degrees C, or 42 degrees C. A strain lacking RluB and RluF also showed no growth defect under these conditions. Mutation of a conserved aspartate in a common sequence motif, previously shown to be essential for the other six E. coli pseudouridine synthases and several yeast pseudouridine synthases, also caused a loss of in vivo activity in all four of the synthases studied in this work.


Assuntos
Escherichia coli/enzimologia , Hidroliases/genética , Pseudouridina/biossíntese , Sequência de Bases , Sítios de Ligação , Escherichia coli/genética , Escherichia coli/crescimento & desenvolvimento , Hidroliases/química , Hidroliases/fisiologia , Dados de Sequência Molecular , Conformação de Ácido Nucleico , RNA/biossíntese , RNA Bacteriano/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...