Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
J Comput Chem ; 44(26): 2096-2102, 2023 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-37381904

RESUMO

The ongoing COVID-19 pandemic, and constant demand for new therapies in unmet clinical needs, necessitates strategies to identify drug candidates for rapid clinical deployment. Over the years, fragment-based drug design (FBDD) has emerged as a mainstream lead discovery strategy in academia, biotechnology start-ups, and large pharma. Chemical building block libraries are the fundamental component of virtually any FBDD campaign. Current trends focus on smaller and smarter libraries that offer synthetically amenable starting points for rational lead generation. Therefore, there remains an ever-increasing need for new methods to generate fragment libraries to seed early-stage drug discovery programs. Here, we present FRAGMENTISE-a new user-friendly, cross-platform tool for user-tunable retrosynthetic small-molecule fragmentation. FRAGMENTISE allows for visualization, similarity search, annotation, and in-depth analysis of the fragment databases in the medicinal chemistry context. FRAGMENTISE is available as standalone software for Linux, Windows, and macOS users, with a graphical interface or command-line version.


Assuntos
COVID-19 , Bibliotecas de Moléculas Pequenas , Humanos , Bibliotecas de Moléculas Pequenas/química , Pandemias , Descoberta de Drogas , Desenho de Fármacos
2.
BMC Bioinformatics ; 16: 336, 2015 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-26493560

RESUMO

BACKGROUND: GmrSD is a modification-dependent restriction endonuclease that specifically targets and cleaves glucosylated hydroxymethylcytosine (glc-HMC) modified DNA. It is encoded either as two separate single-domain GmrS and GmrD proteins or as a single protein carrying both domains. Previous studies suggested that GmrS acts as endonuclease and NTPase whereas GmrD binds DNA. METHODS: In this work we applied homology detection, sequence conservation analysis, fold recognition and homology modeling methods to study sequence-structure-function relationships in the GmrSD restriction endonucleases family. We also analyzed the phylogeny and genomic context of the family members. RESULTS: Results of our comparative genomics study show that GmrS exhibits similarity to proteins from the ParB/Srx fold which can have both NTPase and nuclease activity. In contrast to the previous studies though, we attribute the nuclease activity also to GmrD as we found it to contain the HNH endonuclease motif. We revealed residues potentially important for structure and function in both domains. Moreover, we found that GmrSD systems exist predominantly as a fused, double-domain form rather than as a heterodimer and that their homologs are often encoded in regions enriched in defense and gene mobility-related elements. Finally, phylogenetic reconstructions of GmrS and GmrD domains revealed that they coevolved and only few GmrSD systems appear to be assembled from distantly related GmrS and GmrD components. CONCLUSIONS: Our study provides insight into sequence-structure-function relationships in the yet poorly characterized family of Type IV restriction enzymes. Comparative genomics allowed to propose possible role of GmrD domain in the function of the GmrSD enzyme and possible active sites of both GmrS and GmrD domains. Presented results can guide further experimental characterization of these enzymes.


Assuntos
Enzimas de Restrição do DNA/genética , DNA/genética , Genômica/métodos , Domínio Catalítico , Filogenia , Conformação Proteica , Estrutura Terciária de Proteína , Relação Estrutura-Atividade
3.
Nat Commun ; 5: 3004, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24402442

RESUMO

The 5' cap of human messenger RNA contains 2'-O-methylation of the first and often second transcribed nucleotide that is important for its processing, translation and stability. Human enzymes that methylate these nucleotides, termed CMTr1 and CMTr2, respectively, have recently been identified. However, the structures of these enzymes and their mechanisms of action remain unknown. In the present study, we solve the crystal structures of the active CMTr1 catalytic domain in complex with a methyl group donor and a capped oligoribonucleotide, thereby revealing the mechanism of specific recognition of capped RNA. This mechanism differs significantly from viral enzymes, thus providing a framework for their specific targeting. Based on the crystal structure of CMTr1, a comparative model of the CMTr2 catalytic domain is generated. This model, together with mutational analysis, leads to the identification of residues involved in RNA and methyl group donor binding.


Assuntos
Metiltransferases/metabolismo , Capuzes de RNA/metabolismo , RNA Mensageiro/metabolismo , Cristalografia por Raios X , Humanos , Modelos Moleculares , Estrutura Terciária de Proteína , Processamento Pós-Transcricional do RNA
4.
Mol Syst Biol ; 9: 662, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23632384

RESUMO

In pharmacology, it is crucial to understand the complex biological responses that drugs elicit in the human organism and how well they can be inferred from model organisms. We therefore identified a large set of drug-induced transcriptional modules from genome-wide microarray data of drug-treated human cell lines and rat liver, and first characterized their conservation. Over 70% of these modules were common for multiple cell lines and 15% were conserved between the human in vitro and the rat in vivo system. We then illustrate the utility of conserved and cell-type-specific drug-induced modules by predicting and experimentally validating (i) gene functions, e.g., 10 novel regulators of cellular cholesterol homeostasis and (ii) new mechanisms of action for existing drugs, thereby providing a starting point for drug repositioning, e.g., novel cell cycle inhibitors and new modulators of α-adrenergic receptor, peroxisome proliferator-activated receptor and estrogen receptor. Taken together, the identified modules reveal the conservation of transcriptional responses towards drugs across cell types and organisms, and improve our understanding of both the molecular basis of drug action and human biology.


Assuntos
Reposicionamento de Medicamentos , Redes Reguladoras de Genes/efeitos dos fármacos , Genoma , Fígado/efeitos dos fármacos , Farmacogenética , Transcrição Gênica/efeitos dos fármacos , Animais , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/genética , Linhagem Celular Tumoral , Colesterol/genética , Colesterol/metabolismo , Bases de Dados Genéticas , Perfilação da Expressão Gênica , Humanos , Fígado/citologia , Fígado/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/genética , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Ratos , Receptores Adrenérgicos alfa/genética , Receptores Adrenérgicos alfa/metabolismo , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo , Especificidade da Espécie , Relação Estrutura-Atividade
5.
Nucleic Acids Res ; 40(16): 8163-74, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22718974

RESUMO

Exonuclease VII (ExoVII) is a bacterial nuclease involved in DNA repair and recombination that hydrolyses single-stranded DNA. ExoVII is composed of two subunits: large XseA and small XseB. Thus far, little was known about the molecular structure of ExoVII, the interactions between XseA and XseB, the architecture of the nuclease active site or its mechanism of action. We used bioinformatics methods to predict the structure of XseA, which revealed four domains: an N-terminal OB-fold domain, a middle putatively catalytic domain, a coiled-coil domain and a short C-terminal segment. By series of deletion and site-directed mutagenesis experiments on XseA from Escherichia coli, we determined that the OB-fold domain is responsible for DNA binding, the coiled-coil domain is involved in binding multiple copies of the XseB subunit and residues D155, R205, H238 and D241 of the middle domain are important for the catalytic activity but not for DNA binding. Altogether, we propose a model of sequence-structure-function relationships in ExoVII.


Assuntos
Enzimas Reparadoras do DNA/química , Proteínas de Escherichia coli/química , Exodesoxirribonucleases/química , Sequência de Aminoácidos , Sequência de Bases , Enzimas Reparadoras do DNA/classificação , Enzimas Reparadoras do DNA/metabolismo , Proteínas de Escherichia coli/classificação , Proteínas de Escherichia coli/metabolismo , Exodesoxirribonucleases/classificação , Exodesoxirribonucleases/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Filogenia , Estrutura Terciária de Proteína , Subunidades Proteicas/química , Alinhamento de Sequência , Relação Estrutura-Atividade
6.
Nucleic Acids Res ; 39(11): 4756-68, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21310715

RESUMO

The 5' cap of human messenger RNA consists of an inverted 7-methylguanosine linked to the first transcribed nucleotide by a unique 5'-5' triphosphate bond followed by 2'-O-ribose methylation of the first and often the second transcribed nucleotides, likely serving to modify efficiency of transcript processing, translation and stability. We report the validation of a human enzyme that methylates the ribose of the second transcribed nucleotide encoded by FTSJD1, henceforth renamed HMTR2 to reflect function. Purified recombinant hMTr2 protein transfers a methyl group from S-adenosylmethionine to the 2'-O-ribose of the second nucleotide of messenger RNA and small nuclear RNA. Neither N(7) methylation of the guanosine cap nor 2'-O-ribose methylation of the first transcribed nucleotide are required for hMTr2, but the presence of cap1 methylation increases hMTr2 activity. The hMTr2 protein is distributed throughout the nucleus and cytosol, in contrast to the nuclear hMTr1. The details of how and why specific transcripts undergo modification with these ribose methylations remains to be elucidated. The 2'-O-ribose RNA cap methyltransferases are present in varying combinations in most eukaryotic and many viral genomes. With the capping enzymes in hand their biological purpose can be ascertained.


Assuntos
Metiltransferases/metabolismo , Capuzes de RNA/metabolismo , Evolução Molecular , Humanos , Metilação , Metiltransferases/química , Metiltransferases/genética , Família Multigênica , Proteínas Nucleares/análise , Estrutura Terciária de Proteína , Capuzes de RNA/química , RNA Nuclear Pequeno/metabolismo , Proteínas Recombinantes/metabolismo
7.
Nucleic Acids Res ; 38(12): 4120-32, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20194115

RESUMO

Sgm (Sisomicin-gentamicin methyltransferase) from antibiotic-producing bacterium Micromonospora zionensis is an enzyme that confers resistance to aminoglycosides like gentamicin and sisomicin by specifically methylating G1405 in bacterial 16S rRNA. Sgm belongs to the aminoglycoside resistance methyltransferase (Arm) family of enzymes that have been recently found to spread by horizontal gene transfer among disease-causing bacteria. Structural characterization of Arm enzymes is the key to understand their mechanism of action and to develop inhibitors that would block their activity. Here we report the structure of Sgm in complex with cofactors S-adenosylmethionine (AdoMet) and S-adenosylhomocysteine (AdoHcy) at 2.0 and 2.1 A resolution, respectively, and results of mutagenesis and rRNA footprinting, and protein-substrate docking. We propose the mechanism of methylation of G1405 by Sgm and compare it with other m(7)G methyltransferases, revealing a surprising diversity of active sites and binding modes for the same basic reaction of RNA modification. This analysis can serve as a stepping stone towards developing drugs that would specifically block the activity of Arm methyltransferases and thereby re-sensitize pathogenic bacteria to aminoglycoside antibiotics.


Assuntos
Proteínas de Bactérias/química , Metiltransferases/química , RNA Ribossômico 16S/química , Sequência de Aminoácidos , Aminoglicosídeos/farmacologia , Antibacterianos/biossíntese , Antibacterianos/farmacologia , Sequência de Bases , Calorimetria , Domínio Catalítico , Sequência Conservada , Farmacorresistência Bacteriana , Metilação , Micromonospora/enzimologia , Modelos Moleculares , Dados de Sequência Molecular , RNA Ribossômico 16S/metabolismo , Subunidades Ribossômicas Menores de Bactérias/química , S-Adenosil-Homocisteína/química , S-Adenosilmetionina/química , Homologia de Sequência de Aminoácidos
8.
J Biol Chem ; 285(19): 14701-10, 2010 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-20167602

RESUMO

The YTH (YT521-B homology) domain was identified by sequence comparison and is found in 174 different proteins expressed in eukaryotes. It is characterized by 14 invariant residues within an alpha-helix/beta-sheet structure. Here we show that the YTH domain is a novel RNA binding domain that binds to a short, degenerated, single-stranded RNA sequence motif. The presence of the binding motif in alternative exons is necessary for YT521-B to directly influence splice site selection in vivo. Array analyses demonstrate that YT521-B predominantly regulates vertebrate-specific exons. An NMR titration experiment identified the binding surface for single-stranded RNA on the YTH domain. Structural analyses indicate that the YTH domain is related to the pseudouridine synthase and archaeosine transglycosylase (PUA) domain. Our data show that the YTH domain conveys RNA binding ability to a new class of proteins that are found in all eukaryotic organisms.


Assuntos
Proteínas do Tecido Nervoso/genética , Splicing de RNA/fisiologia , Proteínas de Ligação a RNA/genética , RNA/metabolismo , Sítios de Ligação , Biomarcadores/metabolismo , Perfilação da Expressão Gênica , Humanos , Espectroscopia de Ressonância Magnética , Modelos Moleculares , Análise de Sequência com Séries de Oligonucleotídeos , Conformação Proteica , Dobramento de Proteína , Estrutura Terciária de Proteína , RNA/genética , Fatores de Processamento de RNA
9.
Nucleic Acids Res ; 38(9): 3019-30, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20071747

RESUMO

Genome comparison and genome context analysis were used to find a putative mobile element in the genome of Photorhabdus luminescens, an entomopathogenic bacterium. The element is composed of 16-bp direct repeats in the terminal regions, which are identical to a part of insertion sequences (ISs), a DNA methyltransferase gene homolog, two genes of unknown functions and an open reading frame (ORF) (plu0599) encoding a protein with no detectable sequence similarity to any known protein. The ORF (plu0599) product showed DNA endonuclease activity, when expressed in a cell-free expression system. Subsequently, the protein, named R.PluTI, was expressed in vivo, purified and found to be a novel type IIF restriction enzyme that recognizes 5'-GGCGC/C-3' (/ indicates position of cleavage). R.PluTI cleaves a two-site supercoiled substrate at both the sites faster than a one-site supercoiled substrate. The modification enzyme homolog encoded by plu0600, named M.PluTI, was expressed in Escherichia coli and shown to protect DNA from R.PluTI cleavage in vitro, and to suppress the lethal effects of R.PluTI expression in vivo. These results suggested that they constitute a restriction-modification system, present on the putative mobile element. Our approach thus allowed detection of a previously uncharacterized family of DNA-interacting proteins.


Assuntos
Metilases de Modificação do DNA/genética , Desoxirribonucleases de Sítio Específico do Tipo II/genética , Sequências Repetitivas Dispersas , Photorhabdus/genética , Sequência de Aminoácidos , Clivagem do DNA , Metilases de Modificação do DNA/metabolismo , Desoxirribonucleases de Sítio Específico do Tipo II/química , Desoxirribonucleases de Sítio Específico do Tipo II/metabolismo , Genoma Bacteriano , Cinética , Dados de Sequência Molecular , Photorhabdus/enzimologia , Sequências Repetitivas de Ácido Nucleico , Homologia de Sequência de Aminoácidos
10.
Nucleic Acids Res ; 38(5): 1652-63, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20007606

RESUMO

The Cfr methyltransferase confers combined resistance to five classes of antibiotics that bind to the peptidyl tranferase center of bacterial ribosomes by catalyzing methylation of the C-8 position of 23S rRNA nucleotide A2503. The same nucleotide is targeted by the housekeeping methyltransferase RlmN that methylates the C-2 position. Database searches with the Cfr sequence have revealed a large group of closely related sequences from all domains of life that contain the conserved CX(3)CX(2)C motif characteristic of radical S-adenosyl-l-methionine (SAM) enzymes. Phylogenetic analysis of the Cfr/RlmN family suggests that the RlmN subfamily is likely the ancestral form, whereas the Cfr subfamily arose via duplication and horizontal gene transfer. A structural model of Cfr has been calculated and used as a guide for alanine mutagenesis studies that corroborate the model-based predictions of a 4Fe-4S cluster, a SAM molecule coordinated to the iron-sulfur cluster (SAM1) and a SAM molecule that is the putative methyl group donor (SAM2). All mutations at predicted functional sites affect Cfr activity significantly as assayed by antibiotic susceptibility testing and primer extension analysis. The investigation has identified essential amino acids and Cfr variants with altered reaction mechanisms and represents a first step towards understanding the structural basis of Cfr activity.


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/classificação , Metiltransferases/química , Metiltransferases/classificação , RNA Ribossômico 23S/metabolismo , S-Adenosilmetionina/química , Sequência de Aminoácidos , Proteínas de Bactérias/metabolismo , Farmacorresistência Bacteriana , Evolução Molecular , Ligantes , Metilação , Metiltransferases/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese , Filogenia , S-Adenosilmetionina/metabolismo , Homologia de Sequência de Aminoácidos
11.
Genes Dev ; 23(14): 1650-64, 2009 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-19605687

RESUMO

Spliceosomal small nuclear ribonucleoproteins (snRNPs) in trypanosomes contain either the canonical heptameric Sm ring (U1, U5, spliced leader snRNPs), or variant Sm cores with snRNA-specific Sm subunits (U2, U4 snRNPs). Searching for specificity factors, we identified SMN and Gemin2 proteins that are highly divergent from known orthologs. SMN is splicing-essential in trypanosomes and nuclear-localized, suggesting that Sm core assembly in trypanosomes is nuclear. We demonstrate in vitro that SMN is sufficient to confer specificity of canonical Sm core assembly and to discriminate against binding to nonspecific RNA and to U2 and U4 snRNAs. SMN interacts transiently with the SmD3B subcomplex, contacting specifically SmB. SMN remains associated throughout the assembly of the Sm heteroheptamer and dissociates only when a functional Sm site is incorporated. These data establish a novel role of SMN, mediating snRNP specificity in Sm core assembly, and yield new biochemical insight into the mechanism of SMN activity.


Assuntos
Proteínas do Complexo SMN/metabolismo , Trypanosoma brucei brucei/metabolismo , Proteínas Centrais de snRNP/metabolismo , Sequência de Aminoácidos , Animais , Chaperonas Moleculares/metabolismo , Dados de Sequência Molecular , Proteínas Nucleares/metabolismo , Ribonucleoproteínas Nucleares Pequenas/química , Ribonucleoproteínas Nucleares Pequenas/metabolismo , Proteínas do Complexo SMN/química , Alinhamento de Sequência , Spliceossomos/metabolismo , Proteínas Centrais de snRNP/química
12.
Nucleic Acids Res ; 37(Database issue): D118-21, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18854352

RESUMO

MODOMICS, a database devoted to the systems biology of RNA modification, has been subjected to substantial improvements. It provides comprehensive information on the chemical structure of modified nucleosides, pathways of their biosynthesis, sequences of RNAs containing these modifications and RNA-modifying enzymes. MODOMICS also provides cross-references to other databases and to literature. In addition to the previously available manually curated tRNA sequences from a few model organisms, we have now included additional tRNAs and rRNAs, and all RNAs with 3D structures in the Nucleic Acid Database, in which modified nucleosides are present. In total, 3460 modified bases in RNA sequences of different organisms have been annotated. New RNA-modifying enzymes have been also added. The current collection of enzymes includes mainly proteins for the model organisms Escherichia coli and Saccharomyces cerevisiae, and is currently being expanded to include proteins from other organisms, in particular Archaea and Homo sapiens. For enzymes with known structures, links are provided to the corresponding Protein Data Bank entries, while for many others homology models have been created. Many new options for database searching and querying have been included. MODOMICS can be accessed at http://genesilico.pl/modomics.


Assuntos
Bases de Dados de Ácidos Nucleicos , RNA/química , RNA/metabolismo , Sequência de Bases , Escherichia coli/enzimologia , RNA/biossíntese , RNA Ribossômico/química , RNA Ribossômico/metabolismo , Saccharomyces cerevisiae/enzimologia
13.
Genome Biol ; 9(11): R163, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19025584

RESUMO

BACKGROUND: Alteration in epigenetic methylation can affect gene expression and other processes. In Prokaryota, DNA methyltransferase genes frequently move between genomes and present a potential threat. A methyl-specific deoxyribonuclease, McrBC, of Escherichia coli cuts invading methylated DNAs. Here we examined whether McrBC competes with genome methylation systems through host killing by chromosome cleavage. RESULTS: McrBC inhibited the establishment of a plasmid carrying a PvuII methyltransferase gene but lacking its recognition sites, likely through the lethal cleavage of chromosomes that became methylated. Indeed, its phage-mediated transfer caused McrBC-dependent chromosome cleavage. Its induction led to cell death accompanied by chromosome methylation, cleavage and degradation. RecA/RecBCD functions affect chromosome processing and, together with the SOS response, reduce lethality. Our evolutionary/genomic analyses of McrBC homologs revealed: a wide distribution in Prokaryota; frequent distant horizontal transfer and linkage with mobility-related genes; and diversification in the DNA binding domain. In these features, McrBCs resemble type II restriction-modification systems, which behave as selfish mobile elements, maintaining their frequency by host killing. McrBCs are frequently found linked with a methyltransferase homolog, which suggests a functional association. CONCLUSIONS: Our experiments indicate McrBC can respond to genome methylation systems by host killing. Combined with our evolutionary/genomic analyses, they support our hypothesis that McrBCs have evolved as mobile elements competing with specific genome methylation systems through host killing. To our knowledge, this represents the first report of a defense system against epigenetic systems through cell death.


Assuntos
Enzimas de Restrição do DNA/metabolismo , Escherichia coli/genética , Evolução Molecular , Genoma Bacteriano , Metilação de DNA , Enzimas de Restrição do DNA/genética , Escherichia coli/fisiologia , Escherichia coli/virologia , Sequências Repetitivas Dispersas , Viabilidade Microbiana
14.
BMC Struct Biol ; 8: 48, 2008 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-19014591

RESUMO

BACKGROUND: Catalytic domains of Type II restriction endonucleases (REases) belong to a few unrelated three-dimensional folds. While the PD-(D/E)XK fold is most common among these enzymes, crystal structures have been also determined for single representatives of two other folds: PLD (R.BfiI) and half-pipe (R.PabI). Bioinformatics analyses supported by mutagenesis experiments suggested that some REases belong to the HNH fold (e.g. R.KpnI), and that a small group represented by R.Eco29kI belongs to the GIY-YIG fold. However, for a large fraction of REases with known sequences, the three-dimensional fold and the architecture of the active site remain unknown, mostly due to extreme sequence divergence that hampers detection of homology to enzymes with known folds. RESULTS: R.Hpy188I is a Type II REase with unknown structure. PSI-BLAST searches of the non-redundant protein sequence database reveal only 1 homolog (R.HpyF17I, with nearly identical amino acid sequence and the same DNA sequence specificity). Standard application of state-of-the-art protein fold-recognition methods failed to predict the relationship of R.Hpy188I to proteins with known structure or to other protein families. In order to increase the amount of evolutionary information in the multiple sequence alignment, we have expanded our sequence database searches to include sequences from metagenomics projects. This search resulted in identification of 23 further members of R.Hpy188I family, both from metagenomics and the non-redundant database. Moreover, fold-recognition analysis of the extended R.Hpy188I family revealed its relationship to the GIY-YIG domain and allowed for computational modeling of the R.Hpy188I structure. Analysis of the R.Hpy188I model in the light of sequence conservation among its homologs revealed an unusual variant of the active site, in which the typical Tyr residue of the YIG half-motif had been substituted by a Lys residue. Moreover, some of its homologs have the otherwise invariant Arg residue in a non-homologous position in sequence that nonetheless allows for spatial conservation of the guanidino group potentially involved in phosphate binding. CONCLUSION: The present study eliminates a significant "white spot" on the structural map of REases. It also provides important insight into sequence-structure-function relationships in the GIY-YIG nuclease superfamily. Our results reveal that in the case of proteins with no or few detectable homologs in the standard "non-redundant" database, it is useful to expand this database by adding the metagenomic sequences, which may provide evolutionary linkage to detect more remote homologs.


Assuntos
Domínio Catalítico , Desoxirribonucleases de Sítio Específico do Tipo II/química , Sequência de Aminoácidos , Biologia Computacional/métodos , DNA/química , DNA/metabolismo , Bases de Dados de Proteínas , Desoxirribonucleases de Sítio Específico do Tipo II/metabolismo , Helicobacter pylori/enzimologia , Modelos Moleculares , Dados de Sequência Molecular , Filogenia , Dobramento de Proteína , Alinhamento de Sequência , Especificidade por Substrato
15.
RNA ; 14(10): 2234-44, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18755835

RESUMO

Pseudouridines in the stable RNAs of Bacteria are seldom subjected to further modification. There are 11 pseudouridine (Psi) sites in Escherichia coli rRNA, and further modification is found only at Psi1915 in 23S rRNA, where the N-3 position of the base becomes methylated. Here, we report the identity of the E. coli methyltransferase that specifically catalyzes methyl group addition to form m(3)Psi1915. Analyses of E. coli rRNAs using MALDI mass spectrometry showed that inactivation of the ybeA gene leads to loss of methylation at nucleotide Psi1915. Methylation is restored by complementing the knockout strain with a plasmid-encoded copy of ybeA. Homologs of the ybeA gene, and thus presumably the ensuing methylation at nucleotide m(3)Psi1915, are present in most bacterial lineages but are essentially absent in the Archaea and Eukaryota. Loss of ybeA function in E. coli causes a slight slowing of the growth rate. Phylogenetically, ybeA and its homologs are grouped with other putative S-adenosylmethionine-dependent, SPOUT methyltransferase genes in the Cluster of Orthologous Genes COG1576; ybeA is the first member to be functionally characterized. The YbeA methyltransferase is active as a homodimer and docks comfortably into the ribosomal A site without encroaching into the P site. YbeA makes extensive interface contacts with both the 30S and 50S subunits to align its active site cofactor adjacent to nucleotide Psi1915. Methylation by YbeA (redesignated RlmH for rRNA large subunit methyltransferase H) possibly functions as a stamp of approval signifying that the 50S subunit has engaged in translational initiation.


Assuntos
Proteínas de Escherichia coli/metabolismo , Escherichia coli/enzimologia , Metiltransferases/metabolismo , Pseudouridina/metabolismo , RNA Ribossômico 23S/metabolismo , Subunidades Ribossômicas Maiores de Bactérias/metabolismo , Sequência de Aminoácidos , Sequência Conservada , Dimerização , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Metilação , Metiltransferases/genética , Dados de Sequência Molecular , Conformação Proteica
16.
Nucleic Acids Res ; 36(10): 3252-62, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18420655

RESUMO

N(1)-methylation of adenosine to m(1)A occurs in several different positions in tRNAs from various organisms. A methyl group at position N(1) prevents Watson-Crick-type base pairing by adenosine and is therefore important for regulation of structure and stability of tRNA molecules. Thus far, only one family of genes encoding enzymes responsible for m(1)A methylation at position 58 has been identified, while other m(1)A methyltransferases (MTases) remain elusive. Here, we show that Bacillus subtilis open reading frame yqfN is necessary and sufficient for N(1)-adenosine methylation at position 22 of bacterial tRNA. Thus, we propose to rename YqfN as TrmK, according to the traditional nomenclature for bacterial tRNA MTases, or TrMet(m(1)A22) according to the nomenclature from the MODOMICS database of RNA modification enzymes. tRNAs purified from a DeltatrmK strain are a good substrate in vitro for the recombinant TrmK protein, which is sufficient for m(1)A methylation at position 22 as are tRNAs from Escherichia coli, which natively lacks m(1)A22. TrmK is conserved in Gram-positive bacteria and present in some Gram-negative bacteria, but its orthologs are apparently absent from archaea and eukaryota. Protein structure prediction indicates that the active site of TrmK does not resemble the active site of the m(1)A58 MTase TrmI, suggesting that these two enzymatic activities evolved independently.


Assuntos
Bacillus subtilis/enzimologia , Proteínas de Bactérias/química , tRNA Metiltransferases/química , Sequência de Aminoácidos , Bacillus subtilis/crescimento & desenvolvimento , Proteínas de Bactérias/classificação , Proteínas de Bactérias/genética , Sequência de Bases , Modelos Moleculares , Dados de Sequência Molecular , Filogenia , RNA de Transferência/química , RNA de Transferência/metabolismo , Homologia de Sequência de Aminoácidos , tRNA Metiltransferases/classificação , tRNA Metiltransferases/genética
17.
Proteins ; 71(4): 2076-85, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18186482

RESUMO

MnmC catalyses the last two steps in the biosynthesis of 5-methylaminomethyl-2-thiouridine (mnm(5)s(2)U) in tRNA. Previously, we reported that this bifunctional enzyme is encoded by the yfcK open reading frame in the Escherichia coli K12 genome. However, the mechanism of its activity, in particular the potential structural and functional dependence of the domains responsible for catalyzing the two modification reactions, remains unknown. With the aid of the protein fold-recognition method, we constructed a structural model of MnmC in complex with the ligands and target nucleosides and studied the role of individual amino acids and entire domains by site-directed and deletion mutagenesis, respectively. We found out that the N-terminal domain contains residues responsible for binding of the S-adenosylmethionine cofactor and catalyzing the methylation of nm(5)s(2)U to form mnm(5)s(2)U, while the C-terminal domain contains residues responsible for binding of the FAD cofactor. Further, point mutants with compromised activity of either domain can complement each other to restore a fully functional enzyme. Thus, in the conserved fusion protein MnmC, the individual domains retain independence as enzymes. Interestingly, the N-terminal domain is capable of independent folding, while the isolated C-terminal domain is incapable of folding on its own, a situation similar to the one reported recently for the rRNA modification enzyme RsmC.


Assuntos
Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Complexos Multienzimáticos/química , Complexos Multienzimáticos/metabolismo , RNA de Transferência/biossíntese , Tiouridina/análogos & derivados , Sequência de Aminoácidos , Substituição de Aminoácidos , Sítios de Ligação , Catálise , Biologia Computacional/métodos , Sequência Conservada , Proteínas de Escherichia coli/análise , Proteínas de Escherichia coli/genética , Flavina-Adenina Dinucleotídeo/metabolismo , Teste de Complementação Genética , Ligantes , Metilação , Modelos Moleculares , Dados de Sequência Molecular , Complexos Multienzimáticos/análise , Complexos Multienzimáticos/genética , Ligação Proteica , Dobramento de Proteína , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , RNA Bacteriano/biossíntese , Análise de Sequência , Deleção de Sequência , Homologia de Sequência de Aminoácidos , Eletricidade Estática , Relação Estrutura-Atividade , Tiouridina/metabolismo
19.
Proteins ; 70(1): 1-18, 2008 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-17910062

RESUMO

TRNAs from all organisms contain posttranscriptionally modified nucleosides, which are derived from the four canonical nucleosides. In most tRNAs that read codons beginning with U, adenosine in the position 37 adjacent to the 3' position of the anticodon is modified to N(6)-(Delta(2)-isopentenyl) adenosine (i(6)A). In many bacteria, such as Escherichia coli, this residue is typically hypermodified to N(6)-isopentenyl-2-thiomethyladenosine (ms(2)i(6)A). In a few bacteria, such as Salmonella typhimurium, ms(2)i(6)A can be further hydroxylated to N(6)-(cis-4-hydroxyisopentenyl)-2-thiomethyladenosine (ms(2)io(6)A). Although the enzymes that introduce the respective modifications (prenyltransferase MiaA, methylthiotransferase MiaB, and hydroxylase MiaE) have been identified, their structures remain unknown and sequence-function relationships remain obscure. We carried out sequence analysis and structure prediction of MiaA, MiaB, and MiaE, using the protein fold-recognition approach. Three-dimensional models of all three proteins were then built using a new modeling protocol designed to overcome uncertainties in the alignments and divergence between the templates. For MiaA and MiaB, the catalytic core was built based on the templates from the P-loop NTPase and Radical-SAM superfamilies, respectively. For MiaB, we have also modeled the C-terminal TRAM domain and the newly predicted N-terminal flavodoxin-fold domain. For MiaE, we confidently predict that it shares the three-dimensional fold with the ferritin-like four-helix bundle proteins and that it has a similar active site and mechanism of action to diiron carboxylate enzymes, in particular, methane monooxygenase (E.C.1.14.13.25) that catalyses the biological hydroxylation of alkanes. Our models provide the first structural platform for enzymes involved in the biosynthesis of i(6)A, ms(2)i(6)A, and ms(2)io(6)A, explain the data available from the literature and will help to design further experiments and interpret their results.


Assuntos
Biologia Computacional , Enzimas/química , RNA de Transferência/biossíntese , Sequência de Aminoácidos , Domínio Catalítico , Enzimas/metabolismo , Evolução Molecular , Dados de Sequência Molecular , Filogenia , Conformação Proteica , Dobramento de Proteína , Homologia de Sequência de Aminoácidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA