Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Angew Chem Int Ed Engl ; 62(51): e202311924, 2023 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-37909922

RESUMO

5-Methylcytosine (m5 C) is an RNA modification prevalent on tRNAs, where it can protect tRNAs from endonucleolytic cleavage to maintain protein synthesis. The NSUN family (NSUN1-7 in humans) of RNA methyltransferases are capable of installing the methyl group onto the C5 position of cytosines in RNA. NSUNs are implicated in a wide range of (patho)physiological processes, but selective and cell-active inhibitors of these enzymes are lacking. Here, we use cysteine-directed activity-based protein profiling (ABPP) to discover azetidine acrylamides that act as stereoselective covalent inhibitors of human NSUN2. Despite targeting a conserved catalytic cysteine in the NSUN family, the NSUN2 inhibitors show negligible cross-reactivity with other human NSUNs and exhibit good proteome-wide selectivity. We verify that the azetidine acrylamides inhibit the catalytic activity of recombinant NSUN2, but not NSUN6, and demonstrate that these compounds stereoselectively disrupt NSUN2-tRNA interactions in cancer cells, leading to a global reduction in tRNA m5 C content. Our findings thus highlight the potential to create isotype-selective and cell-active inhibitors of NSUN2 with covalent chemistry targeting a conserved catalytic cysteine.


Assuntos
Azetidinas , Inibidores Enzimáticos , Metiltransferases , tRNA Metiltransferases , Humanos , Acrilamidas , Cisteína/metabolismo , Metilação , Metiltransferases/antagonistas & inibidores , Proteômica , RNA de Transferência/química , tRNA Metiltransferases/antagonistas & inibidores , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia
2.
Neoplasia ; 22(11): 617-629, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33045527

RESUMO

Protein arginine methyltransferase 5 (PRMT5) is an important type II arginine methyltransferase that can play roles in cancers in a highly tissue-specific manner, but its role in the carcinogenesis and metastasis of head and neck squamous cell carcinoma (HNSCC) remains unclear. Here, we detected PRMT5 expression in HNSCC tissues and performed series of in vivo and in vitro assays to investigate the function and mechanism of PRMT5 in HNSCC. We found that PRMT5 was overexpressed in dysplastic and cancer tissues, and associated with lymph node metastasis and worse patient survival. PRMT5 knockdown repressed the malignant phenotype of HNSCC cells in vitro and in vivo. PRMT5 specific inhibitor blocked the formation of precancerous lesion and HNSCC in 4NQO-induced tongue carcinogenesis model, prevented lymph node metastasis in tongue orthotopic xenograft model and inhibited cancer development in subcutaneous xenograft model and Patient-Derived tumor Xenograft (PDX) model. Mechanistically, PRMT5-catalyzed H3R2me2s promotes the enrichment of H3K4me3 in the Twist1 promoter region by recruiting WDR5, and subsequently activates the transcription of Twist1. The rescue experiments indicated that overexpressed Twist1 abrogated the inhibition of cell invasion induced by PRMT5 inhibitor. In summary, this study elucidates that PRMT5 inhibition could reduce H3K4me3-mediated Twist1 transcription and retard the carcinogenesis and metastasis of HNSCC.


Assuntos
Transformação Celular Neoplásica/efeitos dos fármacos , Transformação Celular Neoplásica/metabolismo , Epigênese Genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Proteínas Nucleares/genética , Proteína 1 Relacionada a Twist/genética , tRNA Metiltransferases/antagonistas & inibidores , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Humanos , Imuno-Histoquímica , Camundongos , Modelos Biológicos , Prognóstico , Carcinoma de Células Escamosas de Cabeça e Pescoço/genética , Carcinoma de Células Escamosas de Cabeça e Pescoço/metabolismo , Carcinoma de Células Escamosas de Cabeça e Pescoço/mortalidade , Carcinoma de Células Escamosas de Cabeça e Pescoço/patologia , Transcrição Gênica , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Nucleic Acids Res ; 48(14): 8099-8112, 2020 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-32602532

RESUMO

Translational frameshift errors are often deleterious to the synthesis of functional proteins and could therefore be promoted therapeutically to kill bacteria. TrmD (tRNA-(N(1)G37) methyltransferase) is an essential tRNA modification enzyme in bacteria that prevents +1 errors in the reading frame during protein translation and represents an attractive potential target for the development of new antibiotics. Here, we describe the application of a structure-guided fragment-based drug discovery approach to the design of a new class of inhibitors against TrmD in Mycobacterium abscessus. Fragment library screening, followed by structure-guided chemical elaboration of hits, led to the rapid development of drug-like molecules with potent in vitro TrmD inhibitory activity. Several of these compounds exhibit activity against planktonic M. abscessus and M. tuberculosis as well as against intracellular M. abscessus and M. leprae, indicating their potential as the basis for a novel class of broad-spectrum mycobacterial drugs.


Assuntos
Antibacterianos/farmacologia , Proteínas de Bactérias/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , RNA de Transferência/metabolismo , tRNA Metiltransferases/antagonistas & inibidores , Antibacterianos/química , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Descoberta de Drogas/métodos , Inibidores Enzimáticos/química , Simulação de Acoplamento Molecular , Mycobacterium abscessus/efeitos dos fármacos , Mycobacterium abscessus/enzimologia , Mycobacterium leprae/efeitos dos fármacos , Mycobacterium leprae/enzimologia , Ligação Proteica , tRNA Metiltransferases/química , tRNA Metiltransferases/metabolismo
5.
Trends Cancer ; 6(5): 365-368, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32348733

RESUMO

The major molecular mode of action of the cytotoxic drug 5-fluorouracil (5-FU) is generally considered to result from thymidylate synthase inhibition. Recent findings relating to the function of the human uracil-5 methyltransferase (U5MT), TRMT2A, and its interaction with 5-FU metabolites incorporated within tRNAs, lead to an additional hypothesis that is proposed here.


Assuntos
Fluoruracila/farmacologia , Neoplasias/tratamento farmacológico , Reparo de DNA por Recombinação/efeitos dos fármacos , tRNA Metiltransferases/antagonistas & inibidores , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/genética , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Dano ao DNA , Fluoruracila/uso terapêutico , Humanos , Metilação/efeitos dos fármacos , Neoplasias/genética , RNA de Transferência/metabolismo , Uridina Trifosfato/análogos & derivados , Uridina Trifosfato/metabolismo , tRNA Metiltransferases/metabolismo
6.
J Med Chem ; 62(17): 7788-7805, 2019 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-31442049

RESUMO

Among the >120 modified ribonucleosides in the prokaryotic epitranscriptome, many tRNA modifications are critical to bacterial survival, which makes their synthetic enzymes ideal targets for antibiotic development. Here we performed a structure-based design of inhibitors of tRNA-(N1G37) methyltransferase, TrmD, which is an essential enzyme in many bacterial pathogens. On the basis of crystal structures of TrmDs from Pseudomonas aeruginosa and Mycobacterium tuberculosis, we synthesized a series of thienopyrimidinone derivatives with nanomolar potency against TrmD in vitro and discovered a novel active site conformational change triggered by inhibitor binding. This tyrosine-flipping mechanism is uniquely found in P. aeruginosa TrmD and renders the enzyme inaccessible to the cofactor S-adenosyl-l-methionine (SAM) and probably to the substrate tRNA. Biophysical and biochemical structure-activity relationship studies provided insights into the mechanisms underlying the potency of thienopyrimidinones as TrmD inhibitors, with several derivatives found to be active against Gram-positive and mycobacterial pathogens. These results lay a foundation for further development of TrmD inhibitors as antimicrobial agents.


Assuntos
Inibidores Enzimáticos/farmacologia , Pirimidinas/farmacologia , Tirosina/farmacologia , tRNA Metiltransferases/antagonistas & inibidores , Sítios de Ligação/efeitos dos fármacos , Relação Dose-Resposta a Droga , Desenho de Fármacos , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Modelos Moleculares , Estrutura Molecular , Pseudomonas aeruginosa/enzimologia , Pirimidinas/síntese química , Pirimidinas/química , Relação Estrutura-Atividade , Tirosina/química , tRNA Metiltransferases/metabolismo
7.
J Med Chem ; 62(15): 7210-7232, 2019 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-31282680

RESUMO

Mycobacterium abscessus (Mab) is a rapidly growing species of multidrug-resistant nontuberculous mycobacteria that has emerged as a growing threat to individuals with cystic fibrosis and other pre-existing chronic lung diseases. Mab pulmonary infections are difficult, or sometimes impossible, to treat and result in accelerated lung function decline and premature death. There is therefore an urgent need to develop novel antibiotics with improved efficacy. tRNA (m1G37) methyltransferase (TrmD) is a promising target for novel antibiotics. It is essential in Mab and other mycobacteria, improving reading frame maintenance on the ribosome to prevent frameshift errors. In this work, a fragment-based approach was employed with the merging of two fragments bound to the active site, followed by structure-guided elaboration to design potent nanomolar inhibitors against Mab TrmD. Several of these compounds exhibit promising activity against mycobacterial species, including Mycobacterium tuberculosis and Mycobacterium leprae in addition to Mab, supporting the use of TrmD as a target for the development of antimycobacterial compounds.


Assuntos
Antibacterianos/química , Desenvolvimento de Medicamentos/métodos , Proteínas de Escherichia coli/antagonistas & inibidores , Proteínas de Escherichia coli/metabolismo , Mycobacterium abscessus/efeitos dos fármacos , Mycobacterium abscessus/enzimologia , tRNA Metiltransferases/antagonistas & inibidores , tRNA Metiltransferases/metabolismo , Antibacterianos/farmacologia , Cristalografia por Raios X/métodos , Humanos , Estrutura Secundária de Proteína
8.
J Med Chem ; 56(18): 7278-88, 2013 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-23981144

RESUMO

The tRNA-(N(1)G37) methyltransferase (TrmD) is essential for growth and highly conserved in both Gram-positive and Gram-negative bacterial pathogens. Additionally, TrmD is very distinct from its human orthologue TRM5 and thus is a suitable target for the design of novel antibacterials. Screening of a collection of compound fragments using Haemophilus influenzae TrmD identified inhibitory, fused thieno-pyrimidones that were competitive with S-adenosylmethionine (SAM), the physiological methyl donor substrate. Guided by X-ray cocrystal structures, fragment 1 was elaborated into a nanomolar inhibitor of a broad range of Gram-negative TrmD isozymes. These compounds demonstrated no activity against representative human SAM utilizing enzymes, PRMT1 and SET7/9. This is the first report of selective, nanomolar inhibitors of TrmD with demonstrated ability to order the TrmD lid in the absence of tRNA.


Assuntos
Antibacterianos/farmacologia , Inibidores Enzimáticos/farmacologia , Haemophilus influenzae/enzimologia , tRNA Metiltransferases/antagonistas & inibidores , Adenosina/metabolismo , Aminas/síntese química , Aminas/química , Aminas/metabolismo , Aminas/farmacologia , Antibacterianos/síntese química , Antibacterianos/química , Antibacterianos/metabolismo , Avaliação Pré-Clínica de Medicamentos , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Haemophilus influenzae/efeitos dos fármacos , Humanos , Metionina/metabolismo , Testes de Sensibilidade Microbiana , Modelos Moleculares , Estrutura Terciária de Proteína , RNA de Transferência/química , RNA de Transferência/metabolismo , Relação Estrutura-Atividade , Especificidade por Substrato , tRNA Metiltransferases/química , tRNA Metiltransferases/metabolismo
9.
PLoS One ; 8(6): e65609, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23755257

RESUMO

The amyloid-ß peptide (Aß) is suggested to cause mitochondrial dysfunction in Alzheimer's disease. The mitochondrial dehydrogenase SDR5C1 (also known as ABAD) was shown to bind Aß and was proposed to thereby mediate mitochondrial toxicity, but the molecular mechanism has not been clarified. We recently identified SDR5C1 as an essential component of human mitochondrial RNase P and its associated tRNA:m¹R9 methyltransferase, the enzymes responsible for tRNA 5'-end processing and methylation of purines at tRNA position 9, respectively. With this work we investigated whether SDR5C1's role as a subunit of these two tRNA-maturation activities represents the mechanistic link between Aß and mitochondrial dysfunction. Using recombinant enzyme components, we tested RNase P and methyltransferase activity upon titration of Aß. Micromolar concentrations of monomeric or oligomerized Aß were required to inhibit tRNA 5'-end processing and position 9 methylation catalyzed by the SDR5C1-containing enzymes, yet similar concentrations of Aß also inhibited related RNase P and methyltransferase activities, which do not contain an SDR5C1 homolog. In conclusion, the proposed deleterious effect of Aß on mitochondrial function cannot be explained by a specific inhibition of mitochondrial RNase P or its tRNA:m¹R9 methyltransferase subcomplex, and the molecular mechanism of SDR5C1-mediated Aß toxicity remains unclear.


Assuntos
3-Hidroxiacil-CoA Desidrogenases/química , Peptídeos beta-Amiloides/química , Fragmentos de Peptídeos/química , Ribonuclease P/química , Humanos , Metilação , Metiltransferases/antagonistas & inibidores , Metiltransferases/química , Mitocôndrias/enzimologia , RNA de Transferência de Histidina/química , Proteínas de Saccharomyces cerevisiae/antagonistas & inibidores , Proteínas de Saccharomyces cerevisiae/química , tRNA Metiltransferases/antagonistas & inibidores , tRNA Metiltransferases/química
10.
Mol Carcinog ; 51(10): 796-806, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22965424

RESUMO

Tropomyosins (TMs) are a family of cytoskeletal proteins that bind to and stabilize actin microfilaments. Non-muscle cells express multiple isoforms of TMs including three high molecular weight (HMW) isoforms: TM1, TM2, and TM3. While reports have indicated downregulation of TMs in transformed cells and several human cancers, nevertheless, little is known about the underlying mechanism of TMs suppression. In present study the expression of HMW TMs was investigated in squamous cell carcinoma of esophagus (SCCE), relative to primary cell cultures of normal esophagus by western blotting and real-time RT-PCR. Our results showed that TM1, TM2, and TM3 were significantly downregulated in cell line of SCCE. Moreover, mRNA level of TPM1 and TPM2 were markedly decreased by 93% and 96%, in tumor cell line relative to esophagus normal epithelial cells. Therefore, downregulation of TMs could play an important role in tumorigenesis of esophageal cancer. To asses the mechanism of TM downregulation in esophageal cancer, the role of Ras dependent signaling and promoter hypermethylation were investigated. We found that inhibition of two Ras effectory downstream pathways; MEK/ERK and PI3K/Akt leads to significant increased expression of TM1 protein and both TPM1 and TPM2 mRNAs. In addition, methyltransferase inhibition significantly upregulated TM1, suggesting the prominent contribution of promoter hypermethylation in TM1 downregulation in esophageal cancer. These data indicate that downregulation of HMW TMs occurs basically in SCCE and the activation of MEK/ERK and PI3K/Akt pathways as well as the epigenetic mechanism of promoter hypermethylation play important role in TM1 suppression in SCCE.


Assuntos
Carcinoma de Células Escamosas/metabolismo , Neoplasias Esofágicas/metabolismo , Tropomiosina/metabolismo , Proteínas ras/metabolismo , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patologia , Movimento Celular , Cromonas/farmacologia , Regulação para Baixo , Inibidores Enzimáticos/farmacologia , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patologia , Esôfago/citologia , Regulação Neoplásica da Expressão Gênica , Humanos , MAP Quinase Quinase Quinases/genética , MAP Quinase Quinase Quinases/metabolismo , Metilação , Morfolinas/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Cultura Primária de Células , Isoformas de Proteínas/metabolismo , Transdução de Sinais , Tropomiosina/genética , Quinases raf/genética , Quinases raf/metabolismo , Proteínas ras/genética , tRNA Metiltransferases/antagonistas & inibidores , tRNA Metiltransferases/metabolismo
11.
Bioorg Chem ; 43: 37-43, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22172597

RESUMO

Uracil methylation is essential for survival of organisms and passage of information from generation to generation with high fidelity. Two alternative uridyl methylation enzymes, flavin-dependent thymidylate synthase and folate/FAD-dependent RNA methyltransferase, have joined the long-known classical enzymes, thymidylate synthase and SAM-dependent RNA methyltransferase. These alternative enzymes differ significantly from their classical counterparts in structure, cofactor requirements and chemical mechanism. This review covers the available structural and mechanistic knowledge of the classical and alternative enzymes in biological uracil methylation, and offers a possibility of using inhibitors specifically aiming at microbial thymidylate production as antimicrobial drugs.


Assuntos
Timidilato Sintase/metabolismo , Uracila/metabolismo , tRNA Metiltransferases/metabolismo , Anti-Infecciosos/metabolismo , Biocatálise , Escherichia coli/enzimologia , Flavinas/metabolismo , Metilação , Timidilato Sintase/antagonistas & inibidores , tRNA Metiltransferases/antagonistas & inibidores
12.
RNA ; 17(7): 1236-46, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21602303

RESUMO

Bacterial TrmD and eukaryotic-archaeal Trm5 form a pair of analogous tRNA methyltransferase that catalyze methyl transfer from S-adenosyl methionine (AdoMet) to N(1) of G37, using catalytic motifs that share no sequence or structural homology. Here we show that natural and synthetic analogs of AdoMet are unable to distinguish TrmD from Trm5. Instead, fragments of AdoMet, adenosine and methionine, are selectively inhibitory of TrmD rather than Trm5. Detailed structural information of the two enzymes in complex with adenosine reveals how Trm5 escapes targeting by adopting an altered structure, whereas TrmD is trapped by targeting due to its rigid structure that stably accommodates the fragment. Free energy analysis exposes energetic disparities between the two enzymes in how they approach the binding of AdoMet versus fragments and provides insights into the design of inhibitors selective for TrmD.


Assuntos
Metano/análogos & derivados , Fragmentos de Peptídeos/farmacologia , tRNA Metiltransferases/química , tRNA Metiltransferases/classificação , tRNA Metiltransferases/metabolismo , Adenosina/metabolismo , Adenosina/farmacologia , Desenho de Fármacos , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Metano/química , Metano/metabolismo , Metionina/metabolismo , Metionina/farmacologia , Modelos Biológicos , Modelos Moleculares , Fragmentos de Peptídeos/química , Ligação Proteica , S-Adenosilmetionina/metabolismo , S-Adenosilmetionina/farmacologia , Homologia de Sequência , Especificidade por Substrato , tRNA Metiltransferases/antagonistas & inibidores
13.
EMBO J ; 24(9): 1696-705, 2005 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-15861136

RESUMO

A substrate for protein kinase B (PKB)alpha in HeLa cell extracts was identified as methyltransferase-like protein-1 (METTL1), the orthologue of trm8, which catalyses the 7-methylguanosine modification of tRNA in Saccharomyces cerevisiae. PKB and ribosomal S6 kinase (RSK) both phosphorylated METTL1 at Ser27 in vitro. Ser27 became phosphorylated when HEK293 cells were stimulated with insulin-like growth factor-1 (IGF-1) and this was prevented by inhibition of phosphatidyinositol 3-kinase. The IGF-1-induced Ser27 phosphorylation did not occur in 3-phosphoinositide-dependent protein kinase-1 (PDK1)-deficient embryonic stem cells, but occurred normally in PDK1[L155E] cells, indicating that the effect of IGF-1 is mediated by PKB. METTL1 also became phosphorylated at Ser27 in response to phorbol-12-myristate 13-acetate and this was prevented by PD 184352 or pharmacological inhibition of RSK. Phosphorylation of METTL1 by PKB or RSK inactivated METTL1 in vitro, as did mutation of Ser27 to Asp or Glu. Expression of METTL1[S27D] or METTL1[S27E] did not rescue the growth phenotype of yeast lacking trm8. In contrast, expression of METTL1 or METTL1[S27A] partially rescued growth. These results demonstrate that METTL1 is inactivated by PKB and RSK in cells, and the potential implications of this finding are discussed.


Assuntos
Metiltransferases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , RNA de Transferência/metabolismo , Proteínas Quinases S6 Ribossômicas/metabolismo , Sequência de Aminoácidos , Animais , Proteínas de Ligação ao GTP , Células HeLa , Humanos , Fator de Crescimento Insulin-Like I/farmacologia , Metiltransferases/antagonistas & inibidores , Camundongos , Fosforilação , Proteínas Proto-Oncogênicas c-akt , Serina/metabolismo , Acetato de Tetradecanoilforbol/farmacologia , tRNA Metiltransferases/antagonistas & inibidores , tRNA Metiltransferases/metabolismo
14.
Nucleic Acids Res ; 24(6): 1059-64, 1996 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-8604339

RESUMO

tRNA in which uracil is completely replaced by 5-nitro-uracil was prepared by substituting 5-nitro-UTP for UTP in an in vitro transcription reaction. The rationale was that the 5-nitro substituent activates the 6-carbon of the Ura heterocycle towards nucleophiles, and hence could provide mechanism-based inhibitors of enzymes which utilize this feature in their catalytic mechanism. When assayed shortly after mixing, the tRNA analog, NO2Ura-tRNA, is a potent competitive inhibitor of tRNA-Ura methyl transferase (RUMT). Upon incubation, the analog causes a time-dependent inactivation of RUMT which could be reversed by dilution into a large excess of tRNA substrate. Covalent RUMT-NO2Ura-tRNA complexes could be isolated on nitrocellulose filters or by SDS-PAGE. The interaction of RUMT and NO2Ura-tRNA was deduced to involve formation of a reversible complex, followed by formation of a reversible covalent complex in which Cys 324 of RUMT is linked to the 6-position of NO2Ura 54 in NO2Ura-tRNA.


Assuntos
RNA de Transferência/metabolismo , Uracila/análogos & derivados , tRNA Metiltransferases/metabolismo , Sítios de Ligação , Ligação Competitiva , Colódio , Escherichia coli/metabolismo , Cinética , Substâncias Macromoleculares , RNA Bacteriano/química , RNA Bacteriano/metabolismo , RNA Bacteriano/farmacologia , RNA de Transferência/química , RNA de Transferência/farmacologia , Especificidade por Substrato , Uracila/química , Uracila/metabolismo , Uracila/farmacologia , tRNA Metiltransferases/antagonistas & inibidores
15.
J Antibiot (Tokyo) ; 44(10): 1141-7, 1991 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-1720117

RESUMO

Sinefungin is an antibiotic structurally related to S-adenosylmethionine. It has been described as an inhibitor of RNA transmethylation reactions in viruses and eukaryotic organisms, but not in bacteria. We show here that sinefungin strongly inhibits RNA methyltransferase activity, but not the biosynthesis of these enzymes in Streptomyces. All the methylated bases found in Streptomyces RNA (1-methyladenine, N6-methyladenine, N6,N6-dimethyladenine and 7-methylguanine) are inhibited by this antibiotic. Experiments with sinefungin analogues show that specific changes in the ornithine radical of the molecule still preserve its inhibitory capability. The substitution of the adenine radical by uridine causes the loss of the inhibitory effect. These results and our former studies on Streptomyces DNA methylation, suggest that nucleic acid modification is the main target of sinefungin in Streptomyces.


Assuntos
Adenosina/análogos & derivados , Metilases de Modificação do DNA/efeitos dos fármacos , Streptomyces/enzimologia , tRNA Metiltransferases/efeitos dos fármacos , Adenosina/farmacologia , Cromatografia em Camada Fina , DNA/metabolismo , Metilação , RNA Bacteriano/metabolismo , RNA Ribossômico/metabolismo , RNA de Transferência/metabolismo , Streptomyces/efeitos dos fármacos , Streptomyces/crescimento & desenvolvimento , tRNA Metiltransferases/antagonistas & inibidores , tRNA Metiltransferases/biossíntese
17.
Biochemistry ; 26(26): 8599-606, 1987 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-3327525

RESUMO

tRNA (Ura-5-)methyltransferase catalyzes the transfer of a methyl group from S-adenosylmethionine (AdoMet) to the 5-carbon of a specific Urd residue in tRNA. This results in stoichiometric release of tritium from [5-3H]Urd-labeled substrate tRNA isolated from methyltransferase-deficient Escherichia coli. The enzyme also catalyzes an AdoMet-independent exchange reaction between [5-3H]-Urd-labeled substrate tRNA and protons of water at a rate that is about 1% that of the normal methylation reaction, but with identical stoichiometry. S-Adenosylhomocysteine inhibits the rate of the exchange reaction by 2-3-fold, whereas an analogue having the sulfur of AdoMet replaced by nitrogen accelerates the exchange reaction 9-fold. In the presence (but not absence) of AdoMet, 5-fluorouracil-substituted tRNA (FUra-tRNA) leads to the first-order inactivation of the enzyme. This is accompanied by the formation of a stable covalent complex containing the enzyme, FUra-tRNA, and the methyl group of AdoMet. A mechanism for catalysis is proposed that explains both the 5-H exchange reaction and the inhibition by FUra-tRNA: the enzyme forms a covalent Michael adduct with substrate or inhibitor tRNA by attack of a nucleophilic group of the enzyme at carbon 6 of the pyrimidine residue to be modified. As a result, an anion equivalent is generated at carbon 5 that is sufficiently reactive to be methylated by AdoMet. Preliminary experiments and precedents suggest that the nucleophilic catalyst of the enzyme is a thiol group of cysteine. The potent irreversible inhibition by FUra-tRNA suggests that a mechanism for the "RNA" effects of FUra may also involve irreversible inhibition of RNA-modifying enzymes.


Assuntos
tRNA Metiltransferases/antagonistas & inibidores , Aminobutiratos/farmacologia , Escherichia coli/metabolismo , Fluoruracila/farmacologia , Cinética , Metilação , RNA de Transferência/metabolismo , S-Adenosil-Homocisteína/farmacologia , S-Adenosilmetionina/farmacologia , Trítio
18.
J Virol ; 45(3): 1207-10, 1983 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-6187942

RESUMO

We have previously shown that the inhibition of methylation reactions by the treatment of B77 avian sarcoma virus-infected cells with medium containing cycloleucine results in an inhibition in the intracellular accumulation of the spliced subgenomic mRNA for the virion envelope protein precursor, whereas the genome-size RNA accumulates in larger than normal amounts (C. M. Stoltzfus and R. W. Dane, J. Virol. 42:918-931, 1982). To measure the production of virus particles, we have now determined the reverse transcriptase activity in the culture fluid from infected cells treated with various concentrations of cycloleucine. The activity was somewhat greater in the fluid from the cycloleucine-treated cells than it was in the fluid from the control cells, suggesting an enhancement of particle production in the presence of cycloleucine. In contrast, the production of infectious virions, as determined by the focus assay, decreased when the cycloleucine concentration of the medium increased. We determined the polypeptide compositions of purified particles produced from infected cells treated with or without cycloleucine and labeled with [(3)H]leucine. The relative amounts of radioactivity associated with p19 and p27 were approximately the same in all of the preparations. In contrast, significant decreases were observed in the relative amounts of [(3)H]leucine radioactivity associated with the virion glycoproteins gp85 and gp37. The extent of the decrease in the ratio of gp85 to p27 was a function of the cycloleucine concentration and correlated well with the decrease in the infectivity of the virus particles. Therefore, it is probable that the observed reduction of specific infectivity results from the reduced amounts of envelope glycoproteins in the particles budding from cycloleucine-treated cells.


Assuntos
Aminoácidos/farmacologia , Vírus do Sarcoma Aviário/efeitos dos fármacos , Cicloleucina/farmacologia , Glicoproteínas/deficiência , RNA Mensageiro/metabolismo , RNA Viral/metabolismo , Proteínas Virais/deficiência , Animais , Vírus do Sarcoma Aviário/enzimologia , Vírus do Sarcoma Aviário/patogenicidade , Embrião de Galinha , Fibroblastos , Metilação , DNA Polimerase Dirigida por RNA/análise , Proteínas do Envelope Viral , Virulência , tRNA Metiltransferases/antagonistas & inibidores
20.
Biochem J ; 202(1): 153-62, 1982 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-7082304

RESUMO

Inhibitors of polyamine synthesis (alpha-methylornithine and 1,3-diaminopropan-2-ol) were used to study the relationship between polyamine synthesis and specific methylations of tRNA in Dictyostelium discoideum during vegetative growth. Polyamine concentrations were found to be 10 mM for putrescine, 1.6 mM for spermidine and 7 mM for 1,3-diaminopropane throughout the growth stage. On treatment of growing amoebae with alpha-methylornithine or with 1,3-diaminopropan-2-ol (each at 5 mM), the syntheses of putrescine, spermidine and 1,3-diaminopropane were arrested within 4h. After polyamine synthesis had ceased, the incorporation of methyl groups into tRNA was considerably decreased under conditions that had no effect on the incorporation of uridine into tRNA, or on net syntheses of protein and of DNA. The following nucleosides in tRNA were concerned: 1 methyladenosine, 5-methylcytidine, 7-methylguanosine, 2-methylguanosine, N2N2-dimethylguanosine and 5-methyluridine (ribosylthymine). The corresponding tRNA methyltransferases, determined in Mg2+-free enzyme extracts, proved to be inactive unless polyamines were added. Putrescine and/or spermidine at concentrations of 10 mM or 1-2 mM respectively stimulate the transmethylation reaction in vitro to a maximal rate and to an optimal extent at exactly the same concentrations as found in vegetative cells. In contrast, 1,3-diaminopropane, which is formed from spermidine, does not affect the methylation of tRNA in vitro at physiological concentrations. Putrescine and/or spermidine stabilize the tRNA methyltransferases in crude extracts in the presence but not in the absence of the substrate tRNA. The results support the view that S-adenosylmethionine-dependent transmethylation reactions can be regulated by alterations of polyamine concentrations in vivo.


Assuntos
Ornitina/análogos & derivados , Propanolaminas/farmacologia , Putrescina/biossíntese , RNA de Transferência/metabolismo , Espermidina/biossíntese , tRNA Metiltransferases/metabolismo , Diaminas/biossíntese , Dictyostelium/efeitos dos fármacos , Dictyostelium/metabolismo , Metilação , Nucleosídeos/metabolismo , Ornitina/farmacologia , tRNA Metiltransferases/antagonistas & inibidores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...