Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Mol Cell ; 83(14): 2464-2477.e5, 2023 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-37369200

RESUMO

Co-transcriptional capping of the nascent pre-mRNA 5' end prevents degradation of RNA polymerase (Pol) II transcripts and suppresses the innate immune response. Here, we provide mechanistic insights into the three major steps of human co-transcriptional pre-mRNA capping based on six different cryoelectron microscopy (cryo-EM) structures. The human mRNA capping enzyme, RNGTT, first docks to the Pol II stalk to position its triphosphatase domain near the RNA exit site. The capping enzyme then moves onto the Pol II surface, and its guanylyltransferase receives the pre-mRNA 5'-diphosphate end. Addition of a GMP moiety can occur when the RNA is ∼22 nt long, sufficient to reach the active site of the guanylyltransferase. For subsequent cap(1) methylation, the methyltransferase CMTR1 binds the Pol II stalk and can receive RNA after it is grown to ∼29 nt in length. The observed rearrangements of capping factors on the Pol II surface may be triggered by the completion of catalytic reaction steps and are accommodated by domain movements in the elongation factor DRB sensitivity-inducing factor (DSIF).


Assuntos
Processamento Pós-Transcricional do RNA , RNA Mensageiro , Humanos , RNA Mensageiro/química , RNA Mensageiro/metabolismo , RNA Mensageiro/ultraestrutura , Microscopia Crioeletrônica , RNA Polimerase II/química , RNA Polimerase II/metabolismo , RNA Polimerase II/ultraestrutura , Transcrição Gênica , Metiltransferases/química , Metiltransferases/metabolismo , Metiltransferases/ultraestrutura , Modelos Químicos
2.
Nature ; 613(7943): 383-390, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36599982

RESUMO

Specific, regulated modification of RNAs is important for proper gene expression1,2. tRNAs are rich with various chemical modifications that affect their stability and function3,4. 7-Methylguanosine (m7G) at tRNA position 46 is a conserved modification that modulates steady-state tRNA levels to affect cell growth5,6. The METTL1-WDR4 complex generates m7G46 in humans, and dysregulation of METTL1-WDR4 has been linked to brain malformation and multiple cancers7-22. Here we show how METTL1 and WDR4 cooperate to recognize RNA substrates and catalyse methylation. A crystal structure of METTL1-WDR4 and cryo-electron microscopy structures of METTL1-WDR4-tRNA show that the composite protein surface recognizes the tRNA elbow through shape complementarity. The cryo-electron microscopy structures of METTL1-WDR4-tRNA with S-adenosylmethionine or S-adenosylhomocysteine along with METTL1 crystal structures provide additional insights into the catalytic mechanism by revealing the active site in multiple states. The METTL1 N terminus couples cofactor binding with conformational changes in the tRNA, the catalytic loop and the WDR4 C terminus, acting as the switch to activate m7G methylation. Thus, our structural models explain how post-translational modifications of the METTL1 N terminus can regulate methylation. Together, our work elucidates the core and regulatory mechanisms underlying m7G modification by METTL1, providing the framework to understand its contribution to biology and disease.


Assuntos
Microscopia Crioeletrônica , Proteínas de Ligação ao GTP , Metilação , Metiltransferases , Processamento Pós-Transcricional do RNA , RNA de Transferência , Humanos , Domínio Catalítico , Cristalografia por Raios X , Proteínas de Ligação ao GTP/química , Proteínas de Ligação ao GTP/metabolismo , Proteínas de Ligação ao GTP/ultraestrutura , Metiltransferases/química , Metiltransferases/metabolismo , Metiltransferases/ultraestrutura , RNA de Transferência/química , RNA de Transferência/metabolismo , RNA de Transferência/ultraestrutura , S-Adenosil-Homocisteína/metabolismo , S-Adenosilmetionina/metabolismo , Especificidade por Substrato , Biocatálise
3.
Nat Chem Biol ; 17(4): 485-491, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33462497

RESUMO

Tryptophan 2C methyltransferase (TsrM) methylates C2 of the indole ring of L-tryptophan during biosynthesis of the quinaldic acid moiety of thiostrepton. TsrM is annotated as a cobalamin-dependent radical S-adenosylmethionine (SAM) methylase; however, TsrM does not reductively cleave SAM to the universal 5'-deoxyadenosyl 5'-radical intermediate, a hallmark of radical SAM (RS) enzymes. Herein, we report structures of TsrM from Kitasatospora setae, which are the first structures of a cobalamin-dependent radical SAM methylase. Unexpectedly, the structures show an essential arginine residue that resides in the proximal coordination sphere of the cobalamin cofactor, and a [4Fe-4S] cluster that is ligated by a glutamyl residue and three cysteines in a canonical CXXXCXXC RS motif. Structures in the presence of substrates suggest a substrate-assisted mechanism of catalysis, wherein the carboxylate group of SAM serves as a general base to deprotonate N1 of the tryptophan substrate, facilitating the formation of a C2 carbanion.


Assuntos
Metiltransferases/metabolismo , Metiltransferases/ultraestrutura , Arginina/química , Catálise , Coenzimas , Proteínas Ferro-Enxofre/metabolismo , Metilação , S-Adenosilmetionina , Streptomycetaceae/genética , Streptomycetaceae/metabolismo , Tioestreptona/biossíntese , Triptofano/metabolismo , Vitamina B 12/química , Difração de Raios X/métodos
4.
Life Sci ; 259: 118169, 2020 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-32738360

RESUMO

AIMS: The recent outbreak of pandemic severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has led the world towards a global health emergency. Currently, no proper medicine or effective treatment strategies are available; therefore, repurposing of FDA approved drugs may play an important role in overcoming the situation. MATERIALS AND METHODS: The SARS-CoV-2 genome encodes for 2-O-methyltransferase (2'OMTase), which plays a key role in methylation of viral RNA for evading host immune system. In the present study, the protein sequence of 2'OMTase of SARS-CoV-2 was analyzed, and its structure was modeled by a comparative modeling approach and validated. The library of 3000 drugs was screened against the active site of 2'OMTase followed by re-docking analysis. The apo and ligand-bound 2'OMTase were further validated and analyzed by using molecular dynamics simulation. KEY FINDINGS: The modeled structure displayed the conserved characteristic fold of class I MTase family. The quality assessment analysis by SAVES server reveals that the modeled structure follows protein folding rules and of excellent quality. The docking analysis displayed that the active site of 2'OMTase accommodates an array of drugs, which includes alkaloids, antivirals, cardiac glycosides, anticancer, steroids, and other drugs. The redocking and MD simulation analysis of the best 5 FDA approved drugs reveals that these drugs form a stable conformation with the 2'OMTase. SIGNIFICANCE: The results suggested that these drugs may be used as potential inhibitors for 2'OMTase for combating the SARS-CoV-2 infection.


Assuntos
Betacoronavirus/efeitos dos fármacos , Betacoronavirus/enzimologia , Infecções por Coronavirus/tratamento farmacológico , Metiltransferases/antagonistas & inibidores , Pneumonia Viral/tratamento farmacológico , Antivirais/química , Antivirais/farmacologia , COVID-19 , Biologia Computacional/métodos , Infecções por Coronavirus/virologia , Reposicionamento de Medicamentos/métodos , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Humanos , Metilação/efeitos dos fármacos , Metiltransferases/química , Metiltransferases/metabolismo , Metiltransferases/ultraestrutura , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Terapia de Alvo Molecular , Pandemias , Pneumonia Viral/virologia , SARS-CoV-2 , Homologia de Sequência de Aminoácidos
5.
Nucleic Acids Res ; 48(3): 1572-1582, 2020 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-31919512

RESUMO

BCDIN3 domain containing RNA methyltransferase, BCDIN3D, monomethylates the 5'-monophosphate of cytoplasmic tRNAHis with a G-1:A73 mispair at the top of an eight-nucleotide-long acceptor helix, using S-adenosyl-l-methionine (SAM) as a methyl group donor. In humans, BCDIN3D overexpression is associated with the tumorigenic phenotype and poor prognosis in breast cancer. Here, we present the crystal structure of human BCDIN3D complexed with S-adenosyl-l-homocysteine. BCDIN3D adopts a classical Rossmann-fold methyltransferase structure. A comparison of the structure with that of the closely related methylphosphate capping enzyme, MePCE, which monomethylates the 5'-γ-phosphate of 7SK RNA, revealed the important residues for monomethyl transfer from SAM onto the 5'-monophosphate of tRNAHis and for tRNAHis recognition by BCDIN3D. A structural model of tRNAHis docking onto BCDIN3D suggested the molecular mechanism underlying the different activities between BCDIN3D and MePCE. A loop in BCDIN3D is shorter, as compared to the corresponding region that forms an α-helix to recognize the 5'-end of RNA in MePCE, and the G-1:A73 mispair in tRNAHis allows the N-terminal α-helix of BCDIN3D to wedge the G-1:A73 mispair of tRNAHis. As a result, the 5'-monophosphate of G-1 of tRNAHis is deep in the catalytic pocket for 5'-phosphate methylation. Thus, BCDIN3D is a tRNAHis-specific 5'-monomethylphosphate capping enzyme that discriminates tRNAHis from other tRNA species, and the structural information presented in this study also provides the molecular basis for the development of drugs against breast cancers.


Assuntos
Metiltransferases/ultraestrutura , RNA de Transferência de Histidina/ultraestrutura , RNA de Transferência/genética , S-Adenosil-Homocisteína/química , Antineoplásicos/química , Antineoplásicos/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Cristalografia por Raios X , Citoplasma/química , Citoplasma/genética , Feminino , Regulação Enzimológica da Expressão Gênica/genética , Humanos , Metilação , Metiltransferases/química , Metiltransferases/genética , Conformação Proteica em alfa-Hélice , Dobramento de Proteína , RNA de Transferência/química , RNA de Transferência de Histidina/química , RNA de Transferência de Histidina/genética
6.
J Biosci ; 452020.
Artigo em Inglês | MEDLINE | ID: mdl-31965988

RESUMO

S-adenosyl-L-methionine (AdoMet)-dependent methyltransferases (MTases) are involved in diverse cellular functions. These enzymes show little sequence conservation but have a conserved structural fold. The DNA MTases have characteristic motifs that are involved in AdoMet binding, DNA target recognition and catalysis. Motif III of these MTases have a highly conserved acidic residue, often an aspartate, whose functional significance is not clear. Here, we report a mutational study of the residue in the ß family MTase of the Type III restriction-modification enzyme EcoP15I. Replacement of this residue by alanine affects its methylation activity. We propose that this residue contributes to the affinity of the enzyme for AdoMet. Analysis of the structures of DNA, RNA and protein MTases reveal that the acidic residue is conserved in all of them, and interacts with N6 of the adenine moiety of AdoMet. Interestingly, in the SET-domain protein lysine MTases, which have a fold different from other AdoMet-dependent MTases, N6 of the adenine moiety is hydrogen bonded to the main chain carbonyl group of the histidine residue of the highly conserved motif III. Our study reveals the evolutionary conservation of a carbonyl group in DNA, RNA and protein AdoMet-dependent MTases for specific interaction by hydrogen bond with AdoMet, despite the lack of overall sequence conservation.


Assuntos
DNA/genética , Metiltransferases/genética , Proteína-Arginina N-Metiltransferases/ultraestrutura , Proteínas Repressoras/ultraestrutura , DNA Metiltransferases Sítio Específica (Adenina-Específica)/ultraestrutura , Sequência de Aminoácidos/genética , Sequência Conservada/genética , DNA/ultraestrutura , Metilação de DNA/genética , Enzimas de Restrição-Modificação do DNA/genética , Enzimas de Restrição-Modificação do DNA/ultraestrutura , Humanos , Ligação de Hidrogênio , Metiltransferases/ultraestrutura , Mutagênese Sítio-Dirigida , Conformação de Ácido Nucleico , Conformação Proteica , Conformação Proteica em Folha beta/genética , Dobramento de Proteína , Proteína-Arginina N-Metiltransferases/genética , RNA/genética , RNA/ultraestrutura , Proteínas Repressoras/genética , S-Adenosilmetionina/metabolismo , DNA Metiltransferases Sítio Específica (Adenina-Específica)/genética
7.
J Biol Chem ; 294(40): 14482-14498, 2019 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-31395658

RESUMO

Benzylisoquinoline alkaloids (BIAs) are a structurally diverse class of plant-specialized metabolites that have been particularly well-studied in the order Ranunculales. The N-methyltransferases (NMTs) in BIA biosynthesis can be divided into three groups according to substrate specificity and amino acid sequence. Here, we report the first crystal structures of enzyme complexes from the tetrahydroprotoberberine NMT (TNMT) subclass, specifically for GfTNMT from the yellow horned poppy (Glaucium flavum). GfTNMT was co-crystallized with the cofactor S-adenosyl-l-methionine (dmin = 1.6 Å), the product S-adenosyl-l-homocysteine (dmin = 1.8 Å), or in complex with S-adenosyl-l-homocysteine and (S)-cis-N-methylstylopine (dmin = 1.8 Å). These structures reveal for the first time how a mostly hydrophobic L-shaped substrate recognition pocket selects for the (S)-cis configuration of the two central six-membered rings in protoberberine BIA compounds. Mutagenesis studies confirm and functionally define the roles of several highly-conserved residues within and near the GfTNMT-active site. The substrate specificity of TNMT enzymes appears to arise from the arrangement of subgroup-specific stereospecific recognition elements relative to catalytic elements that are more widely-conserved among all BIA NMTs. The binding mode of protoberberine compounds to GfTNMT appears to be similar to coclaurine NMT, with the isoquinoline rings buried deepest in the binding pocket. This binding mode differs from that of pavine NMT, in which the benzyl ring is bound more deeply than the isoquinoline rings. The insights into substrate recognition and catalysis provided here form a sound basis for the rational engineering of NMT enzymes for chemoenzymatic synthesis and metabolic engineering.


Assuntos
Alcaloides de Berberina/química , Metiltransferases/ultraestrutura , Conformação Proteica , Relação Estrutura-Atividade , Alcaloides/química , Alcaloides/metabolismo , Benzilisoquinolinas/química , Benzilisoquinolinas/metabolismo , Alcaloides de Berberina/metabolismo , Sítios de Ligação/genética , Cristalografia por Raios X , Metiltransferases/química , Metiltransferases/metabolismo , Mutagênese , Ligação Proteica/genética , Ranunculales/enzimologia , S-Adenosilmetionina/química , S-Adenosilmetionina/metabolismo
8.
Nat Commun ; 10(1): 3795, 2019 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-31439846

RESUMO

Histone H3 lysine 36 methylation (H3K36me) is a conserved histone modification deposited by the Set2 methyltransferases. Recent findings show that over-expression or mutation of Set2 enzymes promotes cancer progression, however, mechanisms of H3K36me are poorly understood. Set2 enzymes show spurious activity on histones and histone tails, and it is unknown how they obtain specificity to methylate H3K36 on the nucleosome. In this study, we present 3.8 Å cryo-EM structure of Set2 bound to the mimic of H2B ubiquitinated nucleosome. Our structure shows that Set2 makes extensive interactions with the H3 αN, the H3 tail, the H2A C-terminal tail and stabilizes DNA in the unwrapped conformation, which positions Set2 to specifically methylate H3K36. Moreover, we show that ubiquitin contributes to Set2 positioning on the nucleosome and stimulates the methyltransferase activity. Notably, our structure uncovers interfaces that can be targeted by small molecules for development of future cancer therapies.


Assuntos
Proteínas Fúngicas/metabolismo , Histonas/metabolismo , Metiltransferases/metabolismo , Nucleossomos/metabolismo , Ubiquitina/metabolismo , Chaetomium , Microscopia Crioeletrônica , Metilação de DNA , Proteínas Fúngicas/isolamento & purificação , Proteínas Fúngicas/ultraestrutura , Código das Histonas , Histonas/isolamento & purificação , Histonas/ultraestrutura , Metiltransferases/isolamento & purificação , Metiltransferases/ultraestrutura , Modelos Moleculares , Nucleossomos/ultraestrutura , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestrutura , Ubiquitina/ultraestrutura
9.
Nat Chem Biol ; 15(2): 132-140, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30559425

RESUMO

Among RNA 5'-cap structures, γ-phosphate monomethylation is unique to a small subset of noncoding RNAs, 7SK and U6 in humans. 7SK is capped by methylphosphate capping enzyme (MePCE), which has a second nonenzymatic role as a core component of the 7SK ribonuclear protein (RNP), an essential regulator of RNA transcription. We report 2.0- and 2.1-ŠX-ray crystal structures of the human MePCE methyltransferase domain bound to S-adenosylhomocysteine (SAH) and uncapped or capped 7SK substrates, respectively. 7SK recognition is achieved by protein contacts to a 5'-hairpin-single-stranded RNA region, thus explaining MePCE's specificity for 7SK and U6. The structures reveal SAH and product RNA in a near-transition-state geometry. Unexpectedly, binding experiments showed that MePCE has higher affinity for capped versus uncapped 7SK, and kinetic data support a model of slow product release. This work reveals the molecular mechanism of methyl transfer and 7SK retention by MePCE for subsequent assembly of 7SK RNP.


Assuntos
Metiltransferases/metabolismo , Metiltransferases/ultraestrutura , Células HeLa , Humanos , Metilação , Organofosfatos/metabolismo , Fosfatos , Capuzes de RNA , RNA Longo não Codificante/metabolismo , RNA Nuclear Pequeno/metabolismo , RNA não Traduzido , S-Adenosil-Homocisteína/metabolismo
10.
Mol Cell ; 71(6): 1001-1011.e4, 2018 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-30197297

RESUMO

S-adenosylmethionine (SAM) is an essential metabolite that acts as a cofactor for most methylation events in the cell. The N6-methyladenosine (m6A) methyltransferase METTL16 controls SAM homeostasis by regulating the abundance of SAM synthetase MAT2A mRNA in response to changing intracellular SAM levels. Here we present crystal structures of METTL16 in complex with MAT2A RNA hairpins to uncover critical molecular mechanisms underlying the regulated activity of METTL16. The METTL16-RNA complex structures reveal atomic details of RNA substrates that drive productive methylation by METTL16. In addition, we identify a polypeptide loop in METTL16 near the SAM binding site with an autoregulatory role. We show that mutations that enhance or repress METTL16 activity in vitro correlate with changes in MAT2A mRNA levels in cells. Thus, we demonstrate the structural basis for the specific activity of METTL16 and further suggest the molecular mechanisms by which METTL16 efficiency is tuned to regulate SAM homeostasis.


Assuntos
Metiltransferases/metabolismo , Metiltransferases/ultraestrutura , Regiões 3' não Traduzidas , Adenosina/análogos & derivados , Sítios de Ligação , Células HEK293 , Homeostase , Humanos , Metionina Adenosiltransferase/metabolismo , Metilação , Metiltransferases/fisiologia , RNA , RNA Mensageiro , RNA Nuclear Pequeno/metabolismo , S-Adenosilmetionina/metabolismo
11.
Mol Cell ; 71(6): 986-1000.e11, 2018 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-30197299

RESUMO

Internal modification of RNAs with N6-methyladenosine (m6A) is a highly conserved means of gene expression control. While the METTL3/METTL14 heterodimer adds this mark on thousands of transcripts in a single-stranded context, the substrate requirements and physiological roles of the second m6A writer METTL16 remain unknown. Here we describe the crystal structure of human METTL16 to reveal a methyltransferase domain furnished with an extra N-terminal module, which together form a deep-cut groove that is essential for RNA binding. When presented with a random pool of RNAs, METTL16 selects for methylation-structured RNAs where the critical adenosine is present in a bulge. Mouse 16-cell embryos lacking Mettl16 display reduced mRNA levels of its methylation target, the SAM synthetase Mat2a. The consequence is massive transcriptome dysregulation in ∼64-cell blastocysts that are unfit for further development. This highlights the role of an m6A RNA methyltransferase in facilitating early development via regulation of SAM availability.


Assuntos
Adenosina/análogos & derivados , Metiltransferases/metabolismo , Metiltransferases/ultraestrutura , Adenosina/metabolismo , Animais , Desmetilação , Desenvolvimento Embrionário/genética , Desenvolvimento Embrionário/fisiologia , Expressão Gênica/genética , Células HEK293 , Humanos , Metionina Adenosiltransferase , Metilação , Metiltransferases/fisiologia , Camundongos/embriologia , Camundongos Knockout , RNA , Processamento Pós-Transcricional do RNA/fisiologia , RNA Mensageiro/metabolismo , RNA Nuclear Pequeno/metabolismo
12.
Sci Rep ; 8(1): 5311, 2018 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-29593291

RESUMO

N6-methyladenosine (m6A) is an abundant modification in messenger RNA and noncoding RNAs that affects RNA metabolism. Methyltransferase-like protein 16 (METTL16) is a recently confirmed m6A RNA methyltransferase that methylates U6 spliceosomal RNA and interacts with the 3'-terminal RNA triple helix of MALAT1 (metastasis-associated lung adenocarcinoma transcript 1). Here, we present two X-ray crystal structures of the N-terminal methyltransferase domain (residues 1-291) of human METTL16 (METTL16_291): an apo structure at 1.9 Å resolution and a post-catalytic S-adenosylhomocysteine-bound complex at 2.1 Å resolution. The structures revealed a highly conserved Rossmann fold that is characteristic of Class I S-adenosylmethionine-dependent methyltransferases and a large, positively charged groove. This groove likely represents the RNA-binding site and it includes structural elements unique to METTL16. In-depth analysis of the active site led to a model of the methyl transfer reaction catalyzed by METTL16. In contrast to the major m6A methyltransferase heterodimer METTL3/METTL14, full-length METTL16 forms a homodimer and METTL16_291 exists as a monomer based on size-exclusion chromatography. A native gel-shift assay shows that METTL16 binds to the MALAT1 RNA triple helix, but monomeric METTL16_291 does not. Our results provide insights into the molecular structure of METTL16, which is distinct from METTL3/METTL14.


Assuntos
Metiltransferases/ultraestrutura , Adenosina/metabolismo , Sítios de Ligação , Cristalografia por Raios X , Humanos , Metiltransferases/química , Metiltransferases/metabolismo , RNA/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , RNA Mensageiro/genética , RNA Nuclear Pequeno/metabolismo , S-Adenosilmetionina/metabolismo
13.
Biochem Biophys Res Commun ; 493(1): 240-245, 2017 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-28911863

RESUMO

The wyosine hypermodification found exclusively at G37 of tRNAPhe in eukaryotes and archaea is a very complicated process involving multiple steps and enzymes, and the derivatives are essential for the maintenance of the reading frame during translation. In the archaea Pyrococcus abyssi, two key enzymes from the Trm5 family, named PaTrm5a and PaTrm5b respectively, start the process by forming N1-methylated guanosine (m1G37). In addition, PaTrm5a catalyzes the further methylation of C7 on 4-demethylwyosine (imG-14) to produce isowyosine (imG2) at the same position. The structural basis of the distinct methylation capacities and possible conformational changes during catalysis displayed by the Trm5 enzymes are poorly studied. Here we report the 3.3 Å crystal structure of the mono-functional PaTrm5b, which shares 32% sequence identity with PaTrm5a. Interestingly, structural superposition reveals that the PaTrm5b protein exhibits an extended conformation similar to that of tRNA-bound Trm5b from Methanococcus jannaschii (MjTrm5b), but quite different from the open conformation of apo-PaTrm5a or well folded apo-MjTrm5b reported previously. Truncation of the N-terminal D1 domain leads to reduced tRNA binding as well as the methyltransfer activity of PaTrm5b. The differential positioning of the D1 domains from three reported Trm5 structures were rationalized, which could be attributable to the dissimilar inter-domain interactions and crystal packing patterns. This study expands our understanding on the methylation mechanism of the Trm5 enzymes and wyosine hypermodification.


Assuntos
Proteínas Arqueais/química , Proteínas Arqueais/ultraestrutura , Mathanococcus/enzimologia , Metiltransferases/química , Metiltransferases/ultraestrutura , Pyrococcus abyssi/enzimologia , Sítios de Ligação , Simulação por Computador , Ativação Enzimática , Guanosina/análogos & derivados , Guanosina/química , Modelos Químicos , Modelos Moleculares , Ligação Proteica , Conformação Proteica , RNA de Transferência/química , RNA de Transferência/ultraestrutura , Especificidade da Espécie , Relação Estrutura-Atividade
14.
Science ; 352(6283): 309-12, 2016 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-27081063

RESUMO

RlmN is a dual-specificity RNA methylase that modifies C2 of adenosine 2503 (A2503) in 23S rRNA and C2 of adenosine 37 (A37) in several Escherichia coli transfer RNAs (tRNAs). A related methylase, Cfr, modifies C8 of A2503 via a similar mechanism, conferring resistance to multiple classes of antibiotics. Here, we report the x-ray structure of a key intermediate in the RlmN reaction, in which a Cys(118)→Ala variant of the protein is cross-linked to a tRNA(Glu)substrate through the terminal methylene carbon of a formerly methylcysteinyl residue and C2 of A37. RlmN contacts the entire length of tRNA(Glu), accessing A37 by using an induced-fit strategy that completely unfolds the tRNA anticodon stem-loop, which is likely critical for recognition of both tRNA and ribosomal RNA substrates.


Assuntos
Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/ultraestrutura , Metiltransferases/química , Metiltransferases/ultraestrutura , RNA Bacteriano/química , RNA de Transferência de Ácido Glutâmico/química , RNA de Transferência de Ácido Glutâmico/ultraestrutura , Adenosina/química , Alanina/química , Alanina/genética , Substituição de Aminoácidos , Anticódon/química , Domínio Catalítico , Cristalografia por Raios X , Cisteína/química , Cisteína/genética , Proteínas de Escherichia coli/genética , Metilação , Metiltransferases/genética , Conformação de Ácido Nucleico , Estrutura Terciária de Proteína , S-Adenosilmetionina/química
15.
Biochem Biophys Res Commun ; 474(1): 226-231, 2016 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-27109476

RESUMO

Methylesterase family member 16 (MES16) is an integral component of chlorophyll breakdown. It catalyzes the demethylation of fluorescent chlorophyll catabolite (FCC) and pheophorbide in vitro, and specifically demethylates FCC in vivo. Here we report the crystal structure of MES16 from Arabidopsis thaliana at 2.8 Å resolution. The structure confirm that MES16 is a member of the α/ß-hydrolase superfamily with Ser-87, His-239, and Asp-211 as the catalytic triad. Our biochemical studies reveal that MES16 has esterase activity with methyl-indole acetic acid as the substrate, and the catalytically essential role of Ser-87 has been demonstrated.


Assuntos
Arabidopsis/enzimologia , Ácidos Indolacéticos/química , Metiltransferases/química , Metiltransferases/ultraestrutura , Sítios de Ligação , Ativação Enzimática , Ligação Proteica , Conformação Proteica
16.
Arch Biochem Biophys ; 590: 125-137, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26657067

RESUMO

Two families of methionine synthases, distinct in catalytic and structural features, have been encountered: MetH, the cobalamin-dependent enzyme and MetE, the cobalamin-independent form. The MetE family is of mechanistic interest due to the chemically challenging nature of the reaction and is a potential target for antifungal therapeutics since the human genome encodes only MetH. Here we report the identification, purification, and crystal structure of MetE from the filamentous fungus Neurospora crassa (ncMetE). ncMetE was highly thermostable and crystallized readily, making it ideal for study. Crystal structures of native ncMetE in complex with either Zn(2+)or Cd(2+) were solved at resolution limits of 2.10 Å and 1.88 Å, respectively. The monomeric protein contains two domains, each containing a (ßα)8 barrel core, and a long α-helical segment spans the length of the protein, connecting the domains. Zn(2+) bound in the C-terminal domain exhibits tetrahedral coordination with the side chains of His 652, Cys 654, Glu 676 and Cys 737. A Cd(2+) replete structure revealed a supermetalated enzyme and demonstrated the inate flexibility of the metal binding site. An extensive analysis of sequence conservation within the MetE family identified 57 highly conserved residues and 60 additional residues that were conserved in all fungal sequences examined.


Assuntos
Metais/química , Metiltransferases/química , Metiltransferases/ultraestrutura , Modelos Químicos , Neurospora crassa/enzimologia , Zinco/química , Sequência de Aminoácidos , Sítios de Ligação , Ativação Enzimática , Modelos Moleculares , Dados de Sequência Molecular , Ligação Proteica , Conformação Proteica , Especificidade por Substrato
17.
J Mol Biol ; 427(4): 901-909, 2015 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-25545590

RESUMO

The cobalamin-independent methionine synthase enzyme catalyzes a challenging reaction: the direct transfer of a methyl from 5-methyl-tetrahydrofolate-glutamate3 to the l-homocysteine thiol. The enzyme has a dual (ßα)8 TIM barrel structure that binds, activates and brings the reactants into reaction proximity by conformational movements. In the previously observed open structures, the substrates bind too far apart to react, but we have captured a ternary complex with both substrates bound in a closed form of the enzyme. The closing is described in terms of a hinge between the N- and C-terminal TIM barrels and a rearrangement of key loops within the C domain. The substrate specificity can now be rationalized and the structure reveals His707 as the acid that protonates the THF leaving group through a water molecule trapped in the closed active site. The substrates are correctly oriented for an in-line attack by l-homocysteine on the N(5)-methyl.


Assuntos
Homocisteína/química , Metiltransferases/química , Tetra-Hidrofolatos/química , Substituição de Aminoácidos , Domínio Catalítico , Metionina/biossíntese , Metiltransferases/ultraestrutura , Ligação Proteica , Especificidade por Substrato
18.
Arch Biochem Biophys ; 477(2): 313-23, 2008 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-18555004

RESUMO

The role of Try-81 in the reaction catalyzed by Saccharomyces cerevisiae sterol 24-C-methyltransferase (Erg6p) was investigated kinetically and for product differences against a panel of position-81 mutants in which Tyr was substituted with Trp, Phe, Ile, Leu, Val and Ala. The residue chosen for mutation is one that was reported previously to accept fecosterol and yield a set 24-ethyl (idene) sterol products typical of plants, showing the amino acid residue is located close to the transient C25 carbocation intermediate in the active site. One group of mutants (aromatic) tested with the natural substrate zymosterol accelerated the C-methylation reaction (k(cat)/K(m)) whereas the other group of mutants (aliphatics) decreased catalytic competence as the amino acid side chain was downsized. Mutating to aromatic and assaying with the substrate analog designed as a suicide substrate 26,27-dehydrozymosterol favored C26-monol formation, whereas mutating to the aliphatic of smaller size favored C26-diol formation (a measure of enzyme alkylation). In no case was zymosterol converted to an intermediate that formed a C25-diol. Thermodynamic analysis (determination of E(a), DeltaG(double dagger), DeltaH(double dagger) and TDeltaS(double dagger)) for the C-methylation reaction performed by these enzymes assayed with the substrate and its analog or zymosterol paired with the "charged' high energy intermediate (HEI) analogs 24(R,S)25,epiminolanosterol and 25-azalanosterol or "neutral" membrane insert ergosterol showed that mutation to aromatics can reduce inhibitor potency (measured as K(m)/K(i)), yet catalysis can improve in Trp81 by the introduction of a gain in free energy associated with stabilization of the transition state of a rate-controlling step directed toward turnover. Alternatively, mutation to the smaller aliphatic amino acid side chains led to a destabilization in the active site structure which was accompanied by increases in the partition ratios associated with abortive complex formation. The results are explained by consideration of the functional differences attributed to Tyr81 substitution to aromatics and aliphatics of different size involved with cation-pi or hydrogen bonding interactions and in the activation barriers required of differing side chain conformations to orient the reactants in the direction of turnover versus enzyme inactivation.


Assuntos
Metiltransferases/química , Metiltransferases/ultraestrutura , Modelos Químicos , Modelos Moleculares , Saccharomyces cerevisiae/enzimologia , Tirosina/química , Catálise , Simulação por Computador , Sequência Conservada , Ativação Enzimática , Humanos , Mutagênese Sítio-Dirigida , Mutação , Conformação Proteica , Relação Estrutura-Atividade
19.
Arch Biochem Biophys ; 449(1-2): 57-63, 2006 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-16620761

RESUMO

The methyltransferase KsgA modifies two adjacent adenosines in 16S rRNA by adding two methyl groups to the N(6) position of each nucleotide. Unlike nearly all other rRNA modifications, these modifications and the responsible enzyme are highly conserved phylogenetically, suggesting that the modification system has an important role in ribosome biogenesis. It has been known for some time that KsgA recognizes a complex pre-30S substrate in vitro, but there is disagreement in the literature as to what that substrate can be. That disagreement is resolved in this report; KsgA is unable to methylate 30S subunits in the translationally active conformation, but rather can modify 30S when in an experimentally well established translationally inactive conformation. Recent 30S crystal structures provide some basis for explaining why it is impossible for KsgA to methylate 30S in the translationally active conformation. Previous work identified one set of ribosomal proteins important for efficient methylation by KsgA and another set refractory methylation. With the exception of S21 the recent crystal structures of 30S also instructs that the proteins important for KsgA activity all exert their influence indirectly. Unfortunately, S21, which is inhibitory to KsgA activity, has not had its position determined by X-ray crystallography. A reevaluation of published biophysical data on the location also suggests that the refractory nature of S21 is also indirect. Therefore, it appears that KsgA solely senses the conformation 16S rRNA when carrying out its enzymatic activity.


Assuntos
Metiltransferases/química , Metiltransferases/ultraestrutura , Modelos Químicos , Modelos Moleculares , Proteínas Ribossômicas/química , Proteínas Ribossômicas/ultraestrutura , Sítios de Ligação , Simulação por Computador , Ativação Enzimática , Ligação Proteica , Conformação Proteica , Relação Estrutura-Atividade
20.
Int J Biochem Cell Biol ; 30(1): 13-26, 1998 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9597750

RESUMO

Glycine N-methyltransferase (EC 2.1.1.20) catalyzes the transfer of the methyl group of S-adenosylmethionine (AdoMet) to glycine to form S-adenosylhomocysteine and sarcosine. Unlike most AdoMet-dependent methyltransferases, glycine N-methyltransferase is a tetramer of identical subunits. Crystallography of recombinant rat glycine N-methyltransferase indicates that four nearly spherical subunits are arranged to form a flat, square tetramer with a large hole in the centre. The enzyme occurs abundantly in the livers of rat, rabbit and mouse. Glycine N-methyltransferases from rat, rabbit, human and pig livers are shown to have similar amino acid sequences and, with the enzymes from rat and rabbit livers, it is demonstrated that the N-terminal valine is acetylated. Glycine N-methyltransferases from livers exhibit sigmoidal rate behaviour with respect to AdoMet and hyperbolic behaviour with respect to glycine at all pH tested. However, recombinant rat glycine N-methyltransferase which lacks the N-terminal acetyl group shows no cooperativity toward AdoMet at neutral pH, suggesting that elimination of the positive charge at the N-terminus is required for cooperative behaviour. Glycine N-methyltransferase binds 5-methyltetrahydropteroylpentaglutamate tightly, resulting in inhibition of the catalytic activity. The nature of these unique functional features is discussed in the light of the three-dimensional structure of the enzyme. The tissue and subcellular localization of the enzyme and its possible role in methionine metabolism are reviewed.


Assuntos
Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Metiltransferases/química , Metiltransferases/metabolismo , Sequência de Aminoácidos , Animais , Proteínas de Transporte/ultraestrutura , Catálise , Cristalografia por Raios X , Escherichia coli/enzimologia , Regulação Enzimológica da Expressão Gênica/genética , Glicina/química , Glicina/metabolismo , Glicina N-Metiltransferase , Humanos , Fígado/enzimologia , Metiltransferases/ultraestrutura , Camundongos , Dados de Sequência Molecular , Ácidos Pteroilpoliglutâmicos/química , Ácidos Pteroilpoliglutâmicos/metabolismo , Coelhos , Ratos , S-Adenosilmetionina/química , S-Adenosilmetionina/metabolismo , Homologia de Sequência de Aminoácidos , Especificidade da Espécie
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA