Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
PLoS Genet ; 19(11): e1011013, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37917668

RESUMO

Exposure of Escherichia coli to sub-inhibitory antibiotics stimulates biofilm formation through poorly characterized mechanisms. Using a high-throughput Congo Red binding assay to report on biofilm matrix production, we screened ~4000 E. coli K12 deletion mutants for deficiencies in this biofilm stimulation response. We screened using three different antibiotics to identify core components of the biofilm stimulation response. Mutants lacking acnA, nuoE, or lpdA failed to respond to sub-MIC cefixime and novobiocin, implicating central metabolism and aerobic respiration in biofilm stimulation. These genes are members of the ArcA/B regulon-controlled by a respiration-sensitive two-component system. Mutants of arcA and arcB had a 'pre-activated' phenotype, where biofilm formation was already high relative to wild type in vehicle control conditions, and failed to increase further with the addition of sub-MIC cefixime. Using a tetrazolium dye and an in vivo NADH sensor, we showed spatial co-localization of increased metabolic activity with sub-lethal concentrations of the bactericidal antibiotics cefixime and novobiocin. Supporting a role for respiratory stress, the biofilm stimulation response to cefixime and novobiocin was inhibited when nitrate was provided as an alternative electron acceptor. Deletion of a gene encoding part of the machinery for respiring nitrate abolished its ameliorating effects, and nitrate respiration increased during growth with sub-MIC cefixime. Finally, in probing the generalizability of biofilm stimulation, we found that the stimulation response to translation inhibitors, unlike other antibiotic classes, was minimally affected by nitrate supplementation, suggesting that targeting the ribosome stimulates biofilm formation in distinct ways. By characterizing the biofilm stimulation response to sub-MIC antibiotics at a systems level, we identified multiple avenues for design of therapeutics that impair bacterial stress management.


Assuntos
Antibacterianos , Escherichia coli , Antibacterianos/farmacologia , Escherichia coli/genética , Cefixima/farmacologia , Novobiocina/farmacologia , Nitratos , Biofilmes , Testes de Sensibilidade Microbiana
2.
Trends Microbiol ; 31(6): 571-585, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36709096

RESUMO

Novel approaches are required to address the looming threat of pan-resistant Gram-negative pathogens and forestall the rise of untreatable infections. Unconventional targets that are uniquely important during infection and tractable to high-throughput drug discovery methods hold high potential for innovation in antibiotic discovery programs. In this context, inhibitors of bacterial nutrient stress are particularly exciting candidates for future antibiotic development. Amino acid, nucleotide, and vitamin biosynthesis pathways are critical for bacterial growth in nutrient-limiting conditions in the laboratory and the host. Although historically dismissed as dispensable for pathogens, a wealth of transposon mutagenesis and single-mutant studies have emerged which demonstrate that several such pathways are critical for infection. Indeed, high-throughput screens of diverse synthetic compounds and natural products have uncovered inhibitors of nutrient biosynthesis. Herein, we review bacterial nutrient biosynthesis and its role during host infection. Further, we explore screening platforms developed to search for inhibitors of these targets and highlight successes among these. Finally, we feature important and sometimes surprising connections between bacterial nutrient biosynthesis, antibiotic activity, and antibiotic resistance.


Assuntos
Antibacterianos , Descoberta de Drogas , Antibacterianos/uso terapêutico , Bactérias/genética
3.
ACS Infect Dis ; 8(10): 2187-2197, 2022 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-36098580

RESUMO

Gram-negative bacteria are intrinsically resistant to a plethora of antibiotics that effectively inhibit the growth of Gram-positive bacteria. The intrinsic resistance of Gram-negative bacteria to classes of antibiotics, including rifamycins, aminocoumarins, macrolides, glycopeptides, and oxazolidinones, has largely been attributed to their lack of accumulation within cells due to poor permeability across the outer membrane, susceptibility to efflux pumps, or a combination of these factors. Due to the difficulty in discovering antibiotics that can bypass these barriers, finding targets and compounds that increase the activity of these ineffective antibiotics against Gram-negative bacteria has the potential to expand the antibiotic spectrum. In this study, we investigated the genetic determinants for resistance to rifampicin, novobiocin, erythromycin, vancomycin, and linezolid to determine potential targets of antibiotic-potentiating compounds. We subsequently performed a high-throughput screen of ∼50,000 diverse, synthetic compounds to uncover molecules that potentiate the activity of at least one of the five Gram-positive-targeting antibiotics. This led to the discovery of two membrane active compounds capable of potentiating linezolid and an inhibitor of lipid A biosynthesis capable of potentiating rifampicin and vancomycin. Furthermore, we characterized the ability of known inhibitors of lipid A biosynthesis to potentiate the activity of rifampicin against Gram-negative pathogens.


Assuntos
Antibacterianos , Oxazolidinonas , Antibacterianos/química , Antibacterianos/farmacologia , Eritromicina/farmacologia , Bactérias Gram-Negativas/genética , Linezolida , Lipídeo A , Novobiocina/farmacologia , Oxazolidinonas/farmacologia , Rifampina/farmacologia , Vancomicina/farmacologia
4.
ACS Infect Dis ; 6(3): 338-346, 2020 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-32017534

RESUMO

The spread of antimicrobial resistance continues to be a priority health concern worldwide, necessitating the exploration of alternative therapies. Cannabis sativa has long been known to contain antibacterial cannabinoids, but their potential to address antibiotic resistance has only been superficially investigated. Here, we show that cannabinoids exhibit antibacterial activity against methicillin-resistant Staphylococcus aureus (MRSA), inhibit its ability to form biofilms, and eradicate preformed biofilms and stationary phase cells persistent to antibiotics. We show that the mechanism of action of cannabigerol is through targeting the cytoplasmic membrane of Gram-positive bacteria and demonstrate in vivo efficacy of cannabigerol in a murine systemic infection model caused by MRSA. We also show that cannabinoids are effective against Gram-negative organisms whose outer membrane is permeabilized, where cannabigerol acts on the inner membrane. Finally, we demonstrate that cannabinoids work in combination with polymyxin B against multidrug resistant Gram-negative pathogens, revealing the broad-spectrum therapeutic potential for cannabinoids.


Assuntos
Antibacterianos/farmacologia , Biofilmes/efeitos dos fármacos , Canabinoides/farmacologia , Cannabis/química , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Animais , Antibacterianos/química , Membrana Celular/efeitos dos fármacos , Feminino , Bactérias Gram-Negativas/efeitos dos fármacos , Camundongos , Testes de Sensibilidade Microbiana , Polimixina B/farmacologia , Infecções Estafilocócicas/sangue , Infecções Estafilocócicas/tratamento farmacológico
5.
Nat Commun ; 10(1): 197, 2019 01 14.
Artigo em Inglês | MEDLINE | ID: mdl-30643129

RESUMO

Salmonella Typhimurium (S. Tm) establishes systemic infection in susceptible hosts by evading the innate immune response and replicating within host phagocytes. Here, we sought to identify inhibitors of intracellular S. Tm replication by conducting parallel chemical screens against S. Tm growing in macrophage-mimicking media and within macrophages. We identify several compounds that inhibit Salmonella growth in the intracellular environment and in acidic, ion-limited media. We report on the antimicrobial activity of the psychoactive drug metergoline, which is specific against intracellular S. Tm. Screening an S. Tm deletion library in the presence of metergoline reveals hypersensitization of outer membrane mutants to metergoline activity. Metergoline disrupts the proton motive force at the bacterial cytoplasmic membrane and extends animal survival during a systemic S. Tm infection. This work highlights the predictive nature of intracellular screens for in vivo efficacy, and identifies metergoline as a novel antimicrobial active against Salmonella.


Assuntos
Antibacterianos/farmacologia , Macrófagos/microbiologia , Metergolina/farmacologia , Infecções por Salmonella/tratamento farmacológico , Salmonella typhimurium/efeitos dos fármacos , Animais , Antibacterianos/uso terapêutico , Membrana Celular/efeitos dos fármacos , Membrana Celular/genética , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos/métodos , Feminino , Deleção de Genes , Ensaios de Triagem em Larga Escala/métodos , Humanos , Macrófagos/imunologia , Macrófagos/ultraestrutura , Metergolina/uso terapêutico , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Testes de Sensibilidade Microbiana , Microscopia de Força Atômica , Células RAW 264.7 , Infecções por Salmonella/imunologia , Infecções por Salmonella/microbiologia , Infecções por Salmonella/mortalidade , Salmonella typhimurium/genética , Salmonella typhimurium/patogenicidade , Resultado do Tratamento
6.
Methods Mol Biol ; 1787: 1-18, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29736706

RESUMO

Clinically approved antibiotics target a narrow spectrum of cellular processes, namely cell wall synthesis, DNA replication, and protein synthesis. Numerous screens have been designed to identify inhibitors that target one of these cellular processes. Indeed, this narrow range of drug mechanisms and a reliance on chemical classes discovered many decades ago are thought to be principally responsible for the current crisis of antibiotic drug resistance. Seeking to expand the target base of antibacterial drug discovery, we developed a nutrient stress screening platform that identifies inhibitors of the growth of in Escherichia coli under nutrient limitation. Under nutrient stress, bacteria require an expanded biosynthetic capacity that includes the synthesis of amino acids, vitamins, and nucleobases. Growing evidence suggests that these processes may be indispensable to certain pathogens and at particular sites of infection. Indeed, more than 100 biosynthetic enzymes become indispensable to E. coli grown under nutrient stress in vitro. The screening platform described here puts a focus on these novel targets for new antibiotics and prioritizes growth inhibitory compounds that can be suppressed by individual nutrients and pools thereof.


Assuntos
Avaliação Pré-Clínica de Medicamentos , Escherichia coli/efeitos dos fármacos , Escherichia coli/fisiologia , Fenômenos Fisiológicos da Nutrição/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas , Estresse Fisiológico/efeitos dos fármacos , Antibacterianos/farmacologia , Suplementos Nutricionais , Metabolismo Energético/efeitos dos fármacos
7.
ACS Infect Dis ; 3(12): 955-965, 2017 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-29069544

RESUMO

Actinomycete secondary metabolites are a renowned source of antibacterial chemical scaffolds. Herein, we present a target-specific approach that increases the detection of antimetabolites from natural sources by screening actinomycete-derived extracts against nutrient transporter deletion strains. On the basis of the growth rescue patterns of a collection of 22 Escherichia coli (E. coli) auxotrophic deletion strains representative of the major nutrient biosynthetic pathways, we demonstrate that antimetabolite detection from actinomycete-derived extracts prepared using traditional extraction platforms is masked by nutrient supplementation. In particular, we find poor sensitivity for the detection of antimetabolites targeting vitamin biosynthesis. To circumvent this and as a proof of principle, we exploit the differential activity of actinomycete extracts against E. coli ΔyigM, a biotin transporter deletion strain versus wildtype E. coli. We achieve more than a 100-fold increase in antimetabolite sensitivity using this method and demonstrate a successful bioassay-guided purification of the known biotin antimetabolite, amiclenomycin. Our findings provide a unique solution to uncover the full potential of naturally derived antibiotics.


Assuntos
Actinobacteria/metabolismo , Antimetabólitos/isolamento & purificação , Produtos Biológicos/isolamento & purificação , Descoberta de Drogas , Actinobacteria/crescimento & desenvolvimento , Antimetabólitos/farmacologia , Biotina/metabolismo , Escherichia coli/efeitos dos fármacos , Escherichia coli/metabolismo , Testes de Sensibilidade Microbiana
8.
Nat Microbiol ; 2: 17028, 2017 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-28263303

RESUMO

The increasing use of polymyxins1 in addition to the dissemination of plasmid-borne colistin resistance threatens to cause a serious breach in our last line of defence against multidrug-resistant Gram-negative pathogens, and heralds the emergence of truly pan-resistant infections. Colistin resistance often arises through covalent modification of lipid A with cationic residues such as phosphoethanolamine-as is mediated by Mcr-1 (ref. 2)-which reduce the affinity of polymyxins for lipopolysaccharide3. Thus, new strategies are needed to address the rapidly diminishing number of treatment options for Gram-negative infections4. The difficulty in eradicating Gram-negative bacteria is largely due to their highly impermeable outer membrane, which serves as a barrier to many otherwise effective antibiotics5. Here, we describe an unconventional screening platform designed to enrich for non-lethal, outer-membrane-active compounds with potential as adjuvants for conventional antibiotics. This approach identified the antiprotozoal drug pentamidine6 as an effective perturbant of the Gram-negative outer membrane through its interaction with lipopolysaccharide. Pentamidine displayed synergy with antibiotics typically restricted to Gram-positive bacteria, yielding effective drug combinations with activity against a wide range of Gram-negative pathogens in vitro, and against systemic Acinetobacter baumannii infections in mice. Notably, the adjuvant activity of pentamidine persisted in polymyxin-resistant bacteria in vitro and in vivo. Overall, pentamidine and its structural analogues represent unexploited molecules for the treatment of Gram-negative infections, particularly those having acquired polymyxin resistance determinants.


Assuntos
Antibacterianos/metabolismo , Colistina/metabolismo , Farmacorresistência Bacteriana , Sinergismo Farmacológico , Bactérias Gram-Negativas/efeitos dos fármacos , Pentamidina/metabolismo , Infecções por Acinetobacter/microbiologia , Acinetobacter baumannii/efeitos dos fármacos , Animais , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos/métodos
9.
ACS Infect Dis ; 2(7): 489-99, 2016 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-27626101

RESUMO

The bacterial cell wall has long been a celebrated target for antibacterial drug discovery due to its critical nature in bacteria and absence in mammalian systems. At the heart of the cell wall biosynthetic pathway lies undecaprenyl phosphate (Und-P), the lipid-linked carrier upon which the bacterial cell wall is built. This study exploits recent insights into the link between late-stage wall teichoic acid inhibition and Und-P production, in Gram-positive organisms, to develop a cell-based small-molecule screening platform that enriches for inhibitors of undecaprenyl diphosphate synthase (UppS). Screening a chemical collection of 142,000 small molecules resulted in the identification of 6 new inhibitors of UppS. To date, inhibitors of UppS have generally shown off-target effects on membrane potential due to their physical-chemical characteristics. We demonstrate that MAC-0547630, one of the six inhibitors identified, exhibits selective, nanomolar inhibition against UppS without off-target effects on membrane potential. Such characteristics make it a unique chemical probe for exploring the inhibition of UppS in bacterial cell systems.


Assuntos
Alquil e Aril Transferases/antagonistas & inibidores , Antibacterianos/farmacologia , Bactérias/efeitos dos fármacos , Proteínas de Bactérias/antagonistas & inibidores , Bibliotecas de Moléculas Pequenas/farmacologia , Alquil e Aril Transferases/genética , Alquil e Aril Transferases/metabolismo , Antibacterianos/química , Bactérias/enzimologia , Bactérias/genética , Bactérias/crescimento & desenvolvimento , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Parede Celular/efeitos dos fármacos , Parede Celular/enzimologia , Parede Celular/genética , Avaliação Pré-Clínica de Medicamentos , Testes de Sensibilidade Microbiana , Bibliotecas de Moléculas Pequenas/química , Relação Estrutura-Atividade
10.
PLoS One ; 10(6): e0128587, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26053039

RESUMO

Infections with the bacteria Burkholderia cepacia complex (Bcc) are very difficult to eradicate in cystic fibrosis patients due the intrinsic resistance of Bcc to most available antibiotics and the emergence of multiple antibiotic resistant strains during antibiotic treatment. In this work, we used a whole-cell based assay to screen a diverse collection of small molecules for growth inhibitors of a relevant strain of Bcc, B. cenocepacia K56-2. The primary screen used bacterial growth in 96-well plate format and identified 206 primary actives among 30,259 compounds. From 100 compounds with no previous record of antibacterial activity secondary screening and data mining selected a total of Bce bioactives that were further analyzed. An experimental pipeline, evaluating in vitro antibacterial and antibiofilm activity, toxicity and in vivo antibacterial activity using C. elegans was used for prioritizing compounds with better chances to be further investigated as potential Bcc antibacterial drugs. This high throughput screen, along with the in vitro and in vivo analysis highlights the utility of this experimental method to quickly identify bioactives as a starting point of antibacterial drug discovery.


Assuntos
Burkholderia cenocepacia/efeitos dos fármacos , Burkholderia cenocepacia/crescimento & desenvolvimento , Avaliação Pré-Clínica de Medicamentos/métodos , Bibliotecas de Moléculas Pequenas/farmacologia , Animais , Antibacterianos/farmacologia , Caenorhabditis elegans/efeitos dos fármacos , Comportamento Alimentar/efeitos dos fármacos , Hemólise/efeitos dos fármacos , Ensaios de Triagem em Larga Escala , Testes de Sensibilidade Microbiana , Viabilidade Microbiana/efeitos dos fármacos , Ovinos , Bibliotecas de Moléculas Pequenas/toxicidade
11.
ACS Infect Dis ; 1(11): 533-43, 2015 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-27623408

RESUMO

The widespread emergence of antibiotic drug resistance has resulted in a worldwide healthcare crisis. In particular, the extensive use of ß-lactams, a highly effective class of antibiotics, has been a driver for pervasive ß-lactam resistance. Among the most important resistance determinants are the metallo-ß-lactamases (MBL), which are zinc-requiring enzymes that inactivate nearly all classes of ß-lactams, including the last-resort carbapenem antibiotics. The urgent need for new compounds targeting MBL resistance mechanisms has been widely acknowledged; however, the development of certain types of compounds-namely metal chelators-is actively avoided due to host toxicity concerns. The work herein reports the identification of a series of zinc-selective spiro-indoline-thiadiazole analogues that, in vitro, potentiate ß-lactam antibiotics against an MBL-carrying pathogen by withholding zinc availability. This study demonstrates the ability of one such analogue to inhibit NDM-1 in vitro and, using a mouse model of infection, shows that combination treatment of the respective analogue with meropenem results in a significant decrease in bacterial burden in contrast to animals that received antibiotic treatment alone. These results support the therapeutic potential of these chelators in overcoming antibiotic resistance.

12.
Chem Biol ; 21(1): 136-45, 2014 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-24361049

RESUMO

The dwindling supply of antibiotics that remain effective against drug-resistant bacterial pathogens has precipitated efforts to identify new compounds that inhibit bacterial growth using untapped mechanisms of action. Here, we report both (1) a high-throughput screening methodology designed to discover chemical perturbants of the essential, yet unexploited, process of bacterial iron homeostasis, and (2) our findings from a small-molecule screen of more than 30,000 diverse small molecules that led to the identification and characterization of two spiro-indoline-thiadiazoles that disrupt iron homeostasis in bacteria. We show that these compounds are intracellular chelators with the capacity to exist in two isomeric states. Notably, these spiroheterocyles undergo a transition to an open merocyanine chelating form with antibacterial activity that is specifically induced in the presence of its transition-metal target.


Assuntos
Escherichia coli/efeitos dos fármacos , Homeostase/efeitos dos fármacos , Quelantes de Ferro/química , Quelantes de Ferro/farmacologia , Ferro/metabolismo , Compostos Organometálicos/farmacologia , Elementos de Transição/farmacologia , Antibacterianos/síntese química , Antibacterianos/química , Antibacterianos/farmacologia , Proteínas de Bactérias/antagonistas & inibidores , Benzopiranos/síntese química , Benzopiranos/química , Benzopiranos/farmacologia , Cristalografia por Raios X , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Escherichia coli/metabolismo , Ensaios de Triagem em Larga Escala , Indóis/síntese química , Indóis/química , Indóis/farmacologia , Quelantes de Ferro/síntese química , Compostos Organometálicos/síntese química , Compostos Organometálicos/química , Bibliotecas de Moléculas Pequenas , Compostos de Espiro/química , Estereoisomerismo , Relação Estrutura-Atividade , Tiadiazóis/química , Fatores de Transcrição/antagonistas & inibidores , Elementos de Transição/química
13.
J Biomol Screen ; 18(7): 830-6, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23606650

RESUMO

The synthesis of ribosomes is an essential process, which is aided by a variety of trans-acting factors in bacteria. Among these is a group of GTPases essential for bacterial viability and emerging as promising targets for new antibacterial agents. Herein, we describe a robust high-throughput screening process for inhibitors of one such GTPase, the Escherichia coli EngA protein. The primary screen employed an assay of phosphate production in a 384-well density. Reaction conditions were chosen to maximize sensitivity for the discovery of competitive inhibitors while maintaining a strong signal amplitude and low noise. In a pilot screen of 31,800 chemical compounds, 44 active compounds were identified. Furthermore, we describe the elimination of nonspecific inhibitors that were detergent sensitive or reactive as well as those that interfered with the high-throughput phosphate assay. Four inhibitors survived these common counterscreens for nonspecificity, but these chemicals were also inhibitors of the unrelated enzyme dihydrofolate reductase, suggesting that they too were promiscuously active. The high-throughput screen of the EngA protein described here provides a meticulous pilot study in the search for specific inhibitors of GTPases involved in ribosome biogenesis.


Assuntos
Antibacterianos/química , Proteínas de Escherichia coli/antagonistas & inibidores , Proteínas de Ligação ao GTP/antagonistas & inibidores , Avaliação Pré-Clínica de Medicamentos , Ensaios Enzimáticos , Escherichia coli/enzimologia , Proteínas de Escherichia coli/química , Proteínas de Ligação ao GTP/química , Ensaios de Triagem em Larga Escala , Reprodutibilidade dos Testes , Subunidades Ribossômicas Maiores de Bactérias/metabolismo , Subunidades Ribossômicas Menores de Bactérias/metabolismo , Razão Sinal-Ruído
14.
Nat Chem Biol ; 7(6): 348-50, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21516114

RESUMO

Combinations of antibiotics are commonly used in medicine to broaden antimicrobial spectrum and generate synergistic effects. Alternatively, combination of nonantibiotic drugs with antibiotics offers an opportunity to sample a previously untapped expanse of bioactive chemical space. We screened a collection of drugs to identify compounds that augment the activity of the antibiotic minocycline. Unexpected synergistic drug combinations exhibited in vitro and in vivo activity against bacterial pathogens, including multidrug-resistant isolates.


Assuntos
Antibacterianos/farmacologia , Quimioterapia Combinada/métodos , Antibacterianos/uso terapêutico , Avaliação Pré-Clínica de Medicamentos , Farmacorresistência Bacteriana Múltipla/efeitos dos fármacos , Sinergismo Farmacológico , Minociclina/farmacologia , Minociclina/uso terapêutico
15.
Chem Biol ; 17(8): 852-62, 2010 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-20797614

RESUMO

While cell-based screens have considerable power in identifying new chemical probes of biological systems and leads for new drugs, a major challenge to the utility of such compounds is in connecting phenotype with a cellular target. Here, we present a systematic study to elucidate the mechanism of action of uncharacterized inhibitors of the growth of Escherichia coli through careful analyses of interactions with compounds of known biological activity. We studied growth inhibition with a collection of 200 antibacterial compounds when systematically combined with a panel of 14 known antibiotics of diverse mechanism and chemical class. Our work revealed a high frequency of synergistic chemical-chemical interactions where the interaction profiles were unique to the various compound pairs. Thus, the work revealed that chemical-chemical interaction data provides a fingerprint of biological activity and testable hypotheses regarding the mechanism of action of the novel bioactive molecules. In the study reported here, we determined the mode of action of an inhibitor of folate biosynthesis and a DNA gyrase inhibitor. Moreover, we identified eight membrane-active compounds, found to be promiscuously synergistic with known bioactives.


Assuntos
Anti-Infecciosos/química , Anti-Infecciosos/farmacologia , Avaliação Pré-Clínica de Medicamentos/métodos , Escherichia coli/efeitos dos fármacos , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Análise por Conglomerados , Sinergismo Farmacológico , Escherichia coli/citologia , Escherichia coli/enzimologia , Escherichia coli/metabolismo , Ácido Fólico/biossíntese , Norfloxacino/farmacologia , Sulfametoxazol/farmacologia , Tetra-Hidrofolato Desidrogenase/metabolismo , Inibidores da Topoisomerase II
16.
Methods Mol Biol ; 486: 13-27, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19347613

RESUMO

Small-molecule screening campaigns of model bacteria have been conducted extensively in biotechnology and pharmaceutical companies to search for novel compounds with antibacterial activity. Recently, there has been increasing interest in running such high-throughput screens within academic settings to answer questions in biology. In this respect, whole-cell screening has the particular advantage of identifying compounds with physical and chemical properties compatible with microbial cell permeation, thereby providing probes with which to study diverse aspects of microbial cell physiology and biochemistry. The focus of this chapter is to describe a general method of running a high-throughput screen against a model bacterium to identify small molecules with growth inhibitory activity. Once the primary bioactives have been identified, the determination of their dose-response relationships with the target microbe further characterizes their growth inhibitory effect.


Assuntos
Antibacterianos/farmacologia , Avaliação Pré-Clínica de Medicamentos/métodos , Escherichia coli/efeitos dos fármacos , Testes de Sensibilidade Microbiana/métodos , Bibliotecas de Moléculas Pequenas , Sobrevivência Celular/efeitos dos fármacos , Escherichia coli/crescimento & desenvolvimento
18.
Chembiochem ; 9(16): 2650-62, 2008 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-18972510

RESUMO

Point mutations in beta-glucocerebrosidase (GCase) can result in a deficiency of both GCase activity and protein in lysosomes thereby causing Gaucher Disease (GD). Enzyme inhibitors such as isofagomine, acting as pharmacological chaperones (PCs), increase these levels by binding and stabilizing the native form of the enzyme in the endoplasmic reticulum (ER), and allow increased lysosomal transport of the enzyme. A high-throughput screen of the 50,000-compound Maybridge library identified two, non-carbohydrate-based inhibitory molecules, a 2,4-diamino-5-substituted quinazoline (IC(50) 5 microM) and a 5-substituted pyridinyl-2-furamide (IC(50) 8 microM). They raised the levels of functional GCase 1.5-2.5-fold in N370S or F213I GD fibroblasts. Immunofluorescence confirmed that treated GD fibroblasts had decreased levels of GCase in their ER and increased levels in lysosomes. Changes in protein dynamics, monitored by hydrogen/deuterium-exchange mass spectrometry, identified a domain III active-site loop (residues 243-249) as being significantly stabilized upon binding of isofagomine or either of these two new compounds; this suggests a common mechanism for PC enhancement of intracellular transport.


Assuntos
Avaliação Pré-Clínica de Medicamentos/métodos , Inibidores Enzimáticos/farmacologia , Doença de Gaucher/enzimologia , Glucosilceramidase/antagonistas & inibidores , Animais , Bovinos , Linhagem Celular , Medição da Troca de Deutério , Inibidores Enzimáticos/metabolismo , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/enzimologia , Doença de Gaucher/patologia , Glucosilceramidase/química , Glucosilceramidase/genética , Glucosilceramidase/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Lisossomos/efeitos dos fármacos , Lisossomos/enzimologia , Espectrometria de Massas , Mutação , Conformação Proteica/efeitos dos fármacos , Estabilidade Proteica/efeitos dos fármacos , Especificidade por Substrato
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA