Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nucleic Acids Res ; 46(8): 4114-4128, 2018 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-29538767

RESUMO

Imidazopyrazinones (IPYs) are a new class of compounds that target bacterial topoisomerases as a basis for their antibacterial activity. We have characterized the mechanism of these compounds through structural/mechanistic studies showing they bind and stabilize a cleavage complex between DNA gyrase and DNA ('poisoning') in an analogous fashion to fluoroquinolones, but without the requirement for the water-metal-ion bridge. Biochemical experiments and structural studies of cleavage complexes of IPYs compared with an uncleaved gyrase-DNA complex, reveal conformational transitions coupled to DNA cleavage at the DNA gate. These involve movement at the GyrA interface and tilting of the TOPRIM domains toward the scissile phosphate coupled to capture of the catalytic metal ion. Our experiments show that these structural transitions are involved generally in poisoning of gyrase by therapeutic compounds and resemble those undergone by the enzyme during its adenosine triphosphate-coupled strand-passage cycle. In addition to resistance mutations affecting residues that directly interact with the compounds, we characterized a mutant (D82N) that inhibits formation of the cleavage complex by the unpoisoned enzyme. The D82N mutant appears to act by stabilizing the binary conformation of DNA gyrase with uncleaved DNA without direct interaction with the compounds. This provides general insight into the resistance mechanisms to antibiotics targeting bacterial type II topoisomerases.


Assuntos
Antibacterianos/química , DNA Girase/química , Pirazinas/química , Inibidores da Topoisomerase II/química , Trifosfato de Adenosina/metabolismo , Antibacterianos/classificação , Ciprofloxacina/química , DNA/química , Clivagem do DNA , DNA Girase/metabolismo , Farmacorresistência Bacteriana , Fluoroquinolonas/química , Magnésio/química , Modelos Moleculares , Mutação
2.
Proc Natl Acad Sci U S A ; 114(22): E4492-E4500, 2017 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-28507124

RESUMO

A paucity of novel acting antibacterials is in development to treat the rising threat of antimicrobial resistance, particularly in Gram-negative hospital pathogens, which has led to renewed efforts in antibiotic drug discovery. Fluoroquinolones are broad-spectrum antibacterials that target DNA gyrase by stabilizing DNA-cleavage complexes, but their clinical utility has been compromised by resistance. We have identified a class of antibacterial thiophenes that target DNA gyrase with a unique mechanism of action and have activity against a range of bacterial pathogens, including strains resistant to fluoroquinolones. Although fluoroquinolones stabilize double-stranded DNA breaks, the antibacterial thiophenes stabilize gyrase-mediated DNA-cleavage complexes in either one DNA strand or both DNA strands. X-ray crystallography of DNA gyrase-DNA complexes shows the compounds binding to a protein pocket between the winged helix domain and topoisomerase-primase domain, remote from the DNA. Mutations of conserved residues around this pocket affect activity of the thiophene inhibitors, consistent with allosteric inhibition of DNA gyrase. This druggable pocket provides potentially complementary opportunities for targeting bacterial topoisomerases for antibiotic development.


Assuntos
Antibacterianos , Clivagem do DNA , DNA Girase , Tiofenos , Antibacterianos/química , Antibacterianos/metabolismo , Cristalografia por Raios X , DNA Girase/química , DNA Girase/metabolismo , Descoberta de Drogas , Modelos Moleculares , Tiofenos/química , Tiofenos/metabolismo
3.
Nucleic Acids Res ; 41(15): 7313-31, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23757188

RESUMO

Sperm chromatin incubated in Xenopus egg extracts undergoes origin licensing and nuclear assembly before DNA replication. We found that depletion of DNA topoisomerase IIα (topo IIα), the sole topo II isozyme of eggs and its inhibition by ICRF-193, which clamps topo IIα around DNA have opposite effects on these processes. ICRF-193 slowed down replication origin cluster activation and fork progression in a checkpoint-independent manner, without altering replicon size. In contrast, topo IIα depletion accelerated origin cluster activation, and topo IIα add-back negated overinitiation. Therefore, topo IIα is not required for DNA replication, but topo IIα clamps slow replication, probably by forming roadblocks. ICRF-193 had no effect on DNA synthesis when added after nuclear assembly, confirming that topo IIα activity is dispensable for replication and revealing that topo IIα clamps formed on replicating DNA do not block replication, presumably because topo IIα acts behind and not in front of forks. Topo IIα depletion increased, and topo IIα addition reduced, chromatin loading of MCM2-7 replicative helicase, whereas ICRF-193 did not affect MCM2-7 loading. Therefore, topo IIα restrains MCM2-7 loading in an ICRF-193-resistant manner during origin licensing, suggesting a model for establishing the sequential firing of origin clusters.


Assuntos
Replicação do DNA/efeitos dos fármacos , Proteínas de Ligação a DNA/antagonistas & inibidores , Óvulo/enzimologia , Origem de Replicação , Xenopus/genética , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Animais , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Sequência de Bases , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Pontos de Checagem do Ciclo Celular , Núcleo Celular/genética , Núcleo Celular/metabolismo , DNA Topoisomerases Tipo II/genética , DNA Topoisomerases Tipo II/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Dicetopiperazinas , Masculino , Componente 2 do Complexo de Manutenção de Minicromossomo , Componente 7 do Complexo de Manutenção de Minicromossomo , Óvulo/citologia , Piperazinas/farmacologia , Replicon , Fase S , Espermatozoides/citologia , Espermatozoides/metabolismo , Fatores de Tempo , Xenopus/metabolismo , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo
5.
Elife ; 122024 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-38856655

RESUMO

DNA gyrase, a ubiquitous bacterial enzyme, is a type IIA topoisomerase formed by heterotetramerisation of 2 GyrA subunits and 2 GyrB subunits, to form the active complex. DNA gyrase can loop DNA around the C-terminal domains (CTDs) of GyrA and pass one DNA duplex through a transient double-strand break (DSB) established in another duplex. This results in the conversion from a positive (+1) to a negative (-1) supercoil, thereby introducing negative supercoiling into the bacterial genome by steps of 2, an activity essential for DNA replication and transcription. The strong protein interface in the GyrA dimer must be broken to allow passage of the transported DNA segment and it is generally assumed that the interface is usually stable and only opens when DNA is transported, to prevent the introduction of deleterious DSBs in the genome. In this paper, we show that DNA gyrase can exchange its DNA-cleaving interfaces between two active heterotetramers. This so-called interface 'swapping' (IS) can occur within a few minutes in solution. We also show that bending of DNA by gyrase is essential for cleavage but not for DNA binding per se and favors IS. Interface swapping is also favored by DNA wrapping and an excess of GyrB. We suggest that proximity, promoted by GyrB oligomerization and binding and wrapping along a length of DNA, between two heterotetramers favors rapid interface swapping. This swapping does not require ATP, occurs in the presence of fluoroquinolones, and raises the possibility of non-homologous recombination solely through gyrase activity. The ability of gyrase to undergo interface swapping explains how gyrase heterodimers, containing a single active-site tyrosine, can carry out double-strand passage reactions and therefore suggests an alternative explanation to the recently proposed 'swivelling' mechanism for DNA gyrase (Gubaev et al., 2016).


Assuntos
DNA Girase , DNA Girase/metabolismo , DNA Girase/química , DNA Girase/genética , Multimerização Proteica , DNA Bacteriano/metabolismo , DNA Bacteriano/genética , Escherichia coli/genética , Escherichia coli/enzimologia , Escherichia coli/metabolismo , DNA/metabolismo , DNA/química
6.
EMBO J ; 28(18): 2803-11, 2009 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-19680223

RESUMO

The decatenation activity of topoisomerase II (Top2), which is widely conserved within the eukaryotic domain, is essential for chromosomal segregation in mitosis. It is less clear, however, whether Top2 performs the same function uniformly across the whole genome, and whether all its functions rely on decatenation. In the fission yeast, Schizosaccharomyces pombe, telomeres are bound by Taz1, which promotes smooth replication fork progression through the repetitive telomeric sequences. Hence, replication forks stall at taz1 Delta telomeres. This leads to telomeric entanglements at low temperatures (

Assuntos
DNA Topoisomerases Tipo II/química , Schizosaccharomyces/enzimologia , Telômero/ultraestrutura , Alelos , Catálise , Cromossomos/ultraestrutura , DNA/química , Dimerização , Escherichia coli/metabolismo , Íntrons , Mutagênese Sítio-Dirigida , Mutação , Mutação Puntual , Estrutura Terciária de Proteína , Schizosaccharomyces/genética , Temperatura
7.
RSC Med Chem ; 13(7): 831-839, 2022 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-35919336

RESUMO

By 2050, it is predicted that antimicrobial resistance will be responsible for 10 million global deaths annually, more deaths than cancer, costing the world economy $100 trillion. Clearly, strategies to address this problem are essential as bacterial evolution is rendering our current antibiotics ineffective. The discovery of an allosteric binding site on the established antibacterial target DNA gyrase offers a new medicinal chemistry strategy. As this site is distinct from the fluoroquinolone binding site, resistance is not yet documented. Using in silico molecular design methods, we have designed and synthesised a novel series of biphenyl-based inhibitors inspired by a published thiophene-based allosteric inhibitor. This series was evaluated in vitro against Escherichia coli DNA gyrase and E. coli topoisomerase IV with the most potent compounds exhibiting IC50 values towards the low micromolar range for DNA gyrase and only ∼2-fold less active against topoisomerase IV. The structure-activity relationships reported herein suggest insights to further exploit this allosteric site, offering a pathway to overcome developing fluoroquinolone resistance.

8.
Nat Commun ; 12(1): 150, 2021 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-33420011

RESUMO

Novel bacterial type II topoisomerase inhibitors (NBTIs) stabilize single-strand DNA cleavage breaks by DNA gyrase but their exact mechanism of action has remained hypothetical until now. We have designed a small library of NBTIs with an improved DNA gyrase-binding moiety resulting in low nanomolar inhibition and very potent antibacterial activity. They stabilize single-stranded cleavage complexes and, importantly, we have obtained the crystal structure where an NBTI binds gyrase-DNA in a single conformation lacking apparent static disorder. This directly proves the previously postulated NBTI mechanism of action and shows that they stabilize single-strand cleavage through asymmetric intercalation with a shift of the scissile phosphate. This crystal stucture shows that the chlorine forms a halogen bond with the backbone carbonyls of the two symmetry-related Ala68 residues. To the best of our knowledge, such a so-called symmetrical bifurcated halogen bond has not been identified in a biological system until now.


Assuntos
Antibacterianos/farmacologia , Cloro/metabolismo , DNA Girase/metabolismo , Inibidores da Topoisomerase II/farmacologia , Alanina/química , Alanina/metabolismo , Antibacterianos/química , Cristalografia por Raios X , DNA Girase/química , DNA Topoisomerases Tipo II , DNA de Cadeia Simples/metabolismo , Desenho de Fármacos , Canal de Potássio ERG1/metabolismo , Escherichia coli/efeitos dos fármacos , Escherichia coli/enzimologia , Células Hep G2 , Células Endoteliais da Veia Umbilical Humana , Humanos , Concentração Inibidora 50 , Testes de Sensibilidade Microbiana , Simulação de Acoplamento Molecular , Proteínas de Ligação a Poli-ADP-Ribose/antagonistas & inibidores , Quinolinas/química , Quinolinas/farmacologia , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/enzimologia , Inibidores da Topoisomerase II/química
9.
Nucleic Acids Res ; 36(17): 5623-34, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18765475

RESUMO

Replication origins in Xenopus egg extracts are located at apparently random sequences but are activated in clusters that fire at different times during S phase under the control of ATR/ATM kinases. We investigated whether chromosomal domains and single sequences replicate at distinct times during S phase in egg extracts. Replication foci were found to progressively appear during early S phase and foci labelled early in one S phase colocalized with those labelled early in the next S phase. However, the distribution of these two early labels did not coincide between single origins or origin clusters on single DNA fibres. The 4 Mb Xenopus rDNA repeat domain was found to replicate later than the rest of the genome and to have a more nuclease-resistant chromatin structure. Replication initiated more frequently in the transcription unit than in the intergenic spacer. These results suggest for the first time that in this embryonic system, where transcription does not occur, replication timing is deterministic at the scale of large chromatin domains (1-5 Mb) but stochastic at the scale of replicons (10 kb) and replicon clusters (50-100 kb).


Assuntos
Cromossomos/química , Replicação do DNA , Origem de Replicação , Replicon , Fase S/genética , Animais , Extratos Celulares , Núcleo Celular/metabolismo , Cromossomos/metabolismo , DNA Ribossômico/biossíntese , DNA Ribossômico/química , Cinética , Masculino , Óvulo/metabolismo , Espermatozoides/metabolismo , Processos Estocásticos , Xenopus laevis
10.
J Mol Biol ; 431(18): 3427-3449, 2019 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-31301408

RESUMO

Type II topoisomerases regulate DNA topology by making a double-stranded break in one DNA duplex, transporting another DNA segment through this break and then resealing it. Bacterial type IIA topoisomerase inhibitors, such as fluoroquinolones and novel bacterial topoisomerase inhibitors, can trap DNA cleavage complexes with double- or single-stranded cleaved DNA. To study the mode of action of such compounds, 21 crystal structures of a "gyraseCORE" fusion truncate of Staphyloccocus aureus DNA gyrase complexed with DNA and diverse inhibitors have been published, as well as 4 structures lacking inhibitors. These structures have the DNA in various cleavage states and appear to track trajectories along the catalytic paths of the DNA cleavage/religation steps. The various conformations sampled by these multiple "gyraseCORE" structures show rigid body movements of the catalytic GyrA WHD and GyrB TOPRIM domains across the dimer interface. Conformational changes common to all compound-bound structures suggest common mechanisms for DNA cleavage-stabilizing compounds. The structures suggest that S. aureus gyrase uses a single moving-metal ion for cleavage and that the central four base pairs need to be stretched between the two catalytic sites, in order for a scissile phosphate to attract a metal ion to the A-site to catalyze cleavage, after which it is "stored" in another coordination configuration (B-site) in the vicinity. We present a simplified model for the catalytic cycle in which capture of the transported DNA segment causes conformational changes in the ATPase domain that push the DNA gate open, resulting in stretching and cleaving the gate-DNA in two steps.


Assuntos
Antibacterianos/farmacologia , Clivagem do DNA/efeitos dos fármacos , DNA Topoisomerases Tipo I/efeitos dos fármacos , DNA/química , Inibidores da Topoisomerase/farmacologia , Domínio Catalítico , DNA/metabolismo , DNA Girase/química , DNA Girase/genética , DNA Topoisomerases Tipo I/química , DNA Topoisomerases Tipo II/química , DNA Topoisomerases Tipo II/efeitos dos fármacos , Fluoroquinolonas/farmacologia , Metais , Modelos Moleculares , Conformação Proteica , Quinolonas , Staphylococcus aureus/enzimologia , Inibidores da Topoisomerase II/farmacologia
11.
J Med Chem ; 61(8): 3565-3581, 2018 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-29596745

RESUMO

In our quest for new antibiotics able to address the growing threat of multidrug resistant infections caused by Gram-negative bacteria, we have investigated an unprecedented series of non-quinolone bacterial topoisomerase inhibitors from the Sanofi patrimony, named IPYs for imidazopyrazinones, as part of the Innovative Medicines Initiative (IMI) European Gram Negative Antibacterial Engine (ENABLE) organization. Hybridization of these historical compounds with the quinazolinediones, a known series of topoisomerase inhibitors, led us to a novel series of tricyclic IPYs that demonstrated potential for broad spectrum activity, in vivo efficacy, and a good developability profile, although later profiling revealed a genotoxicity risk. Resistance studies revealed partial cross-resistance with fluoroquinolones (FQs) suggesting that IPYs bind to the same region of bacterial topoisomerases as FQs and interact with at least some of the keys residues involved in FQ binding.


Assuntos
Antibacterianos/farmacologia , Proteínas de Bactérias/antagonistas & inibidores , Imidazóis/farmacologia , Pirazinas/farmacologia , Quinazolinonas/farmacologia , Inibidores da Topoisomerase/farmacologia , Animais , Antibacterianos/síntese química , Antibacterianos/farmacocinética , Antibacterianos/toxicidade , Farmacorresistência Bacteriana/efeitos dos fármacos , Bactérias Gram-Negativas/efeitos dos fármacos , Células Hep G2 , Humanos , Imidazóis/síntese química , Imidazóis/farmacocinética , Imidazóis/toxicidade , Masculino , Camundongos , Testes de Sensibilidade Microbiana , Pirazinas/síntese química , Pirazinas/farmacocinética , Pirazinas/toxicidade , Quinazolinonas/síntese química , Quinazolinonas/farmacocinética , Quinazolinonas/toxicidade , Inibidores da Topoisomerase/síntese química , Inibidores da Topoisomerase/farmacocinética , Inibidores da Topoisomerase/toxicidade
12.
EMBO Rep ; 6(8): 729-35, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16025133

RESUMO

DNA topoisomerase II (topo II) is involved in unlinking replicating DNA and organizing mitotic chromosomes. Topo II is the target of many antitumour drugs. Topo II inhibition results in extensive catenation of newly replicated DNA and may potentially perturb chromatin assembly. Here, we show that the topo II inhibitor ICRF-193 does not prevent the bulk of nucleosome deposition, but perturbs nucleosome spacing in Xenopus egg extracts. This is due to the trapping of topo II-closed clamps on the DNA rather than increased DNA catenation. Inhibition of replicative DNA decatenation has in itself little or no effect on nucleosome deposition and spacing, suggesting that DNA can easily accommodate the sharp bending constraints imposed by the co-habitation of nucleosomes and catenane nodes. Chromatin perturbation by topo II clamps may explain some dominant cellular effects of ICRF-193. Nucleosome-driven bending of precatenane nodes may facilitate their unlinking by topo II during unperturbed replication.


Assuntos
Antineoplásicos/farmacologia , Cromatina/metabolismo , DNA Topoisomerases Tipo II/química , DNA/química , Piperazinas/farmacologia , Animais , Ciclo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Cromatina/química , Estruturas Cromossômicas , DNA/efeitos dos fármacos , Replicação do DNA , DNA Topoisomerases Tipo I/química , Dicetopiperazinas , Inibidores Enzimáticos/farmacologia , Masculino , Modelos Biológicos , Nucleossomos/efeitos dos fármacos , Nucleossomos/metabolismo , Plasmídeos/metabolismo , Espermatozoides/metabolismo , Xenopus
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA