Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Cell Chem Biol ; 27(12): 1532-1543.e6, 2020 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-33186541

RESUMO

Pioneering microbial genomic surveys have revealed numerous untapped biosynthetic gene clusters, unveiling the great potential of new natural products. Here, using a combination of genome mining, mutasynthesis, and activity screening in an infection model comprising Caenorhabditis elegans and Pseudomonas aeruginosa, we identified candidate virulence-blocking amychelin siderophore compounds from actinomycetes. Subsequently, we developed unreported analogs of these virulence-blocking siderophores with improved potency by exploiting an Amycolatopsis methanolica strain 239T chorismate to salicylate a biosynthetic subpathway for mutasynthesis. This allowed us to generate the fluorinated amychelin, fluoroamychelin I, which rescued C. elegans from P. aeruginosa-mediated killing with an EC50 value of 1.4 µM, outperforming traditional antibiotics including ceftazidime and meropenem. In general, this paper describes an efficient platform for the identification and production of classes of anti-microbial compounds with potential unique modes of action.


Assuntos
Mineração de Dados , Genômica , Halogenação , Pseudomonas aeruginosa/efeitos dos fármacos , Pseudomonas aeruginosa/genética , Sideróforos/química , Sideróforos/farmacologia , Animais , Antibacterianos/química , Antibacterianos/farmacologia , Caenorhabditis elegans/genética , Ceftazidima/farmacologia , Avaliação Pré-Clínica de Medicamentos , Meropeném/farmacologia
2.
Nature ; 556(7699): 103-107, 2018 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-29590091

RESUMO

A challenge in the treatment of Staphylococcus aureus infections is the high prevalence of methicillin-resistant S. aureus (MRSA) strains and the formation of non-growing, dormant 'persister' subpopulations that exhibit high levels of tolerance to antibiotics and have a role in chronic or recurrent infections. As conventional antibiotics are not effective in the treatment of infections caused by such bacteria, novel antibacterial therapeutics are urgently required. Here we used a Caenorhabditis elegans-MRSA infection screen to identify two synthetic retinoids, CD437 and CD1530, which kill both growing and persister MRSA cells by disrupting lipid bilayers. CD437 and CD1530 exhibit high killing rates, synergism with gentamicin, and a low probability of resistance selection. All-atom molecular dynamics simulations demonstrated that the ability of retinoids to penetrate and embed in lipid bilayers correlates with their bactericidal ability. An analogue of CD437 was found to retain anti-persister activity and show an improved cytotoxicity profile. Both CD437 and this analogue, alone or in combination with gentamicin, exhibit considerable efficacy in a mouse model of chronic MRSA infection. With further development and optimization, synthetic retinoids have the potential to become a new class of antimicrobials for the treatment of Gram-positive bacterial infections that are currently difficult to cure.


Assuntos
Antibacterianos/classificação , Antibacterianos/farmacologia , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Retinoides/farmacologia , Infecções Estafilocócicas/tratamento farmacológico , Infecções Estafilocócicas/microbiologia , Animais , Antibacterianos/efeitos adversos , Antibacterianos/uso terapêutico , Benzoatos/química , Benzoatos/farmacologia , Benzoatos/uso terapêutico , Benzoatos/toxicidade , Caenorhabditis elegans/efeitos dos fármacos , Caenorhabditis elegans/microbiologia , Morte Celular/efeitos dos fármacos , Linhagem Celular , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Sinergismo Farmacológico , Gentamicinas/farmacologia , Gentamicinas/uso terapêutico , Humanos , Bicamadas Lipídicas/química , Staphylococcus aureus Resistente à Meticilina/citologia , Staphylococcus aureus Resistente à Meticilina/genética , Staphylococcus aureus Resistente à Meticilina/crescimento & desenvolvimento , Camundongos , Testes de Sensibilidade Microbiana , Simulação de Dinâmica Molecular , Mutação , Naftóis/química , Naftóis/farmacologia , Naftóis/uso terapêutico , Naftóis/toxicidade , Retinoides/química , Retinoides/uso terapêutico , Retinoides/toxicidade
3.
Artigo em Inglês | MEDLINE | ID: mdl-28923873

RESUMO

Bacterial persisters are a quasidormant subpopulation of cells that are tolerant to antibiotic treatment. The combination of the aminoglycoside tobramycin with fumarate as an antibacterial potentiator utilizes an antipersister strategy that is aimed at reducing recurrent Pseudomonas aeruginosa infections by enhancing the killing of P. aeruginosa persisters. Stationary-phase cultures of P. aeruginosa were used to generate persister cells. A range of tobramycin concentrations was tested with a range of metabolite concentrations to determine the potentiation effect of the metabolite under a variety of conditions, including a range of pH values and in the presence of azithromycin or cystic fibrosis (CF) patient sputum. In addition, 96-well dish biofilm and colony biofilm assays were performed, and the cytotoxicity of the tobramycin-fumarate combination was determined utilizing a lactate dehydrogenase (LDH) assay. Enhanced killing of up to 6 orders of magnitude of P. aeruginosa persisters over a range of CF isolates, including mucoid and nonmucoid strains, was observed for the tobramycin-fumarate combination compared to killing with tobramycin alone. Furthermore, significant fumarate-mediated potentiation was seen in the presence of azithromycin or CF patient sputum. Fumarate also reduced the cytotoxicity of tobramycin-treated P. aeruginosa to human epithelial airway cells. Finally, in mucoid and nonmucoid CF isolates, complete eradication of P. aeruginosa biofilm was observed in the colony biofilm assay due to fumarate potentiation. These data suggest that a combination of tobramycin with fumarate as an antibacterial potentiator may be an attractive therapeutic for eliminating recurrent P. aeruginosa infections in CF patients through the eradication of bacterial persisters.


Assuntos
Antibacterianos/farmacologia , Fumaratos/farmacologia , Infecções por Pseudomonas/tratamento farmacológico , Pseudomonas aeruginosa/efeitos dos fármacos , Tobramicina/farmacologia , Azitromicina/farmacologia , Biofilmes/crescimento & desenvolvimento , Fibrose Cística , Farmacorresistência Bacteriana , Quimioterapia Combinada , Humanos , Testes de Sensibilidade Microbiana , Infecções por Pseudomonas/microbiologia , Escarro/química , Escarro/microbiologia
4.
Artigo em Inglês | MEDLINE | ID: mdl-28652232

RESUMO

Francisella tularensis is a highly infectious Gram-negative intracellular pathogen that causes tularemia. Because of its potential as a bioterrorism agent, there is a need for new therapeutic agents. We therefore developed a whole-animal Caenorhabditis elegans-F. tularensis pathosystem for high-throughput screening to identify and characterize potential therapeutic compounds. We found that the C. elegans p38 mitogen-activate protein (MAP) kinase cascade is involved in the immune response to F. tularensis, and we developed a robust F. tularensis-mediated C. elegans killing assay with a Z' factor consistently of >0.5, which was then utilized to screen a library of FDA-approved compounds that included 1,760 small molecules. In addition to clinically used antibiotics, five FDA-approved drugs were also identified as potential hits, including the anti-inflammatory drug diflunisal that showed anti-F. tularensis activity in vitro Moreover, the nonsteroidal anti-inflammatory drug (NSAID) diflunisal, at 4× MIC, blocked the replication of an F. tularensis live vaccine strain (LVS) in primary human macrophages and nonphagocytic cells. Diflunisal was nontoxic to human erythrocytes and HepG2 human liver cells at concentrations of ≥32 µg/ml. Finally, diflunisal exhibited synergetic activity with the antibiotic ciprofloxacin in both a checkerboard assay and a macrophage infection assay. In conclusion, the liquid C. elegans-F. tularensis LVS assay described here allows screening for anti-F. tularensis compounds and suggests that diflunisal could potentially be repurposed for the management of tularemia.


Assuntos
Antibacterianos/farmacologia , Anti-Inflamatórios/farmacologia , Caenorhabditis elegans/efeitos dos fármacos , Francisella tularensis/efeitos dos fármacos , Animais , Vacinas Bacterianas/imunologia , Caenorhabditis elegans/imunologia , Linhagem Celular Tumoral , Ciprofloxacina/farmacologia , Eritrócitos/microbiologia , Francisella tularensis/imunologia , Células Hep G2 , Humanos , Fígado/microbiologia , Macrófagos/microbiologia , Vacinas Atenuadas/imunologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
5.
PLoS One ; 10(6): e0127640, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26039584

RESUMO

Persisters are a subpopulation of normal bacterial cells that show tolerance to conventional antibiotics. Persister cells are responsible for recalcitrant chronic infections and new antibiotics effective against persisters would be a major development in the treatment of these infections. Using the reporter dye SYTOX Green that only stains cells with permeabilized membranes, we developed a fluorescence-based screening assay in a 384-well format for identifying compounds that can kill methicillin-resistant Staphylococcus aureus (MRSA) persisters. The assay proved robust and suitable for high throughput screening (Z`-factor: >0.7). In screening a library of hits from a previous screen, which identified compounds that had the ability to block killing of the nematode Caenorhabditis by MRSA, we discovered that the low molecular weight compound NH125, a bacterial histidine kinase inhibitor, kills MRSA persisters by causing cell membrane permeabilization, and that 5 µg/mL of the compound can kill all cells to the limit of detection in a 108 CFU/mL culture of MRSA persisters within 3h. Furthermore, NH125 disrupts 50% of established MRSA biofilms at 20 µg/mL and completely eradicates biofilms at 160 µg/mL. Our results suggest that the SYTOX Green screening assay is suitable for large-scale projects to identify small molecules effective against MRSA persisters and should be easily adaptable to a broad range of pathogens that form persisters. Since NH125 has strong bactericidal properties against MRSA persisters and high selectivity to bacteria, we believe NH125 is a good anti-MRSA candidate drug that should be further evaluated.


Assuntos
Antibacterianos/farmacocinética , Biofilmes/efeitos dos fármacos , Caenorhabditis elegans/microbiologia , Fluorescência , Staphylococcus aureus Resistente à Meticilina/fisiologia , Infecções Estafilocócicas/tratamento farmacológico , Animais , Antibacterianos/química , Avaliação Pré-Clínica de Medicamentos , Compostos Orgânicos/química
6.
PLoS One ; 9(2): e89189, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24586584

RESUMO

Staphylococcus aureus, the leading cause of hospital-acquired infections in the United States, is also pathogenic to the model nematode Caenorhabditis elegans. The C. elegans-S. aureus infection model was previously carried out on solid agar plates where the bacteriovorous C. elegans feeds on a lawn of S. aureus. However, agar-based assays are not amenable to large scale screens for antibacterial compounds. We have developed a high throughput liquid screening assay that uses robotic instrumentation to dispense a precise amount of methicillin resistant S. aureus (MRSA) and worms in 384-well assay plates, followed by automated microscopy and image analysis. In validation of the liquid assay, an MRSA cell wall defective mutant, MW2ΔtarO, which is attenuated for killing in the agar-based assay, was found to be less virulent in the liquid assay. This robust assay with a Z'-factor consistently greater than 0.5 was utilized to screen the Biomol 4 compound library consisting of 640 small molecules with well characterized bioactivities. As proof of principle, 27 of the 30 clinically used antibiotics present in the library conferred increased C. elegans survival and were identified as hits in the screen. Surprisingly, the antihelminthic drug closantel was also identified as a hit in the screen. In further studies, we confirmed the anti-staphylococcal activity of closantel against vancomycin-resistant S. aureus isolates and other Gram-positive bacteria. The liquid C. elegans-S. aureus assay described here allows screening for anti-staphylococcal compounds that are not toxic to the host.


Assuntos
Antibacterianos/farmacologia , Caenorhabditis elegans/efeitos dos fármacos , Caenorhabditis elegans/microbiologia , Avaliação Pré-Clínica de Medicamentos/métodos , Staphylococcus aureus Resistente à Meticilina/genética , Animais , Ensaios de Triagem em Larga Escala/métodos , Salicilanilidas , Bibliotecas de Moléculas Pequenas/química
7.
PLoS One ; 4(9): e7025, 2009 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-19750012

RESUMO

Candida albicans, the most common human pathogenic fungus, can establish a persistent lethal infection in the intestine of the microscopic nematode Caenorhabditis elegans. The C. elegans-C. albicans infection model was previously adapted to screen for antifungal compounds. Modifications to this screen have been made to facilitate a high-throughput assay including co-inoculation of nematodes with C. albicans and instrumentation allowing precise dispensing of worms into assay wells, eliminating two labor-intensive steps. This high-throughput method was utilized to screen a library of 3,228 compounds represented by 1,948 bioactive compounds and 1,280 small molecules derived via diversity-oriented synthesis. Nineteen compounds were identified that conferred an increase in C. elegans survival, including most known antifungal compounds within the chemical library. In addition to seven clinically used antifungal compounds, twelve compounds were identified which are not primarily used as antifungal agents, including three immunosuppressive drugs. This assay also allowed the assessment of the relative minimal inhibitory concentration, the effective concentration in vivo, and the toxicity of the compound in a single assay.


Assuntos
Antifúngicos/farmacologia , Candida albicans/efeitos dos fármacos , Técnicas de Química Combinatória , Testes de Sensibilidade Microbiana , Animais , Caenorhabditis elegans , Química Farmacêutica/métodos , Desenho de Fármacos , Descoberta de Drogas , Avaliação Pré-Clínica de Medicamentos , Imunossupressores/uso terapêutico , Mutação
8.
ACS Chem Biol ; 4(7): 527-33, 2009 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-19572548

RESUMO

The nematode Caenorhabditis elegans is a unique whole animal model system for identifying small molecules with in vivo anti-infective properties. C. elegans can be infected with a broad range of human pathogens, including Enterococcus faecalis, an important human nosocomial pathogen. Here, we describe an automated, high-throughput screen of 37,200 compounds and natural product extracts for those that enhance survival of C. elegans infected with E. faecalis. Using a robot to dispense live, infected animals into 384-well plates and automated microscopy and image analysis, we identified 28 compounds and extracts not previously reported to have antimicrobial properties, including six structural classes that cure infected C. elegans animals but do not affect the growth of the pathogen in vitro, thus acting by a mechanism of action distinct from antibiotics currently in clinical use.


Assuntos
Antibacterianos/farmacologia , Caenorhabditis elegans/efeitos dos fármacos , Caenorhabditis elegans/microbiologia , Modelos Animais de Doenças , Enterococcus faecalis/efeitos dos fármacos , Animais , Antibacterianos/química , Técnicas de Química Combinatória , Avaliação Pré-Clínica de Medicamentos , Enterococcus faecalis/crescimento & desenvolvimento , Infecções por Bactérias Gram-Positivas/tratamento farmacológico , Humanos , Estrutura Molecular
9.
J Med Microbiol ; 57(Pt 7): 809-813, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18566137

RESUMO

Expression of a myriad of virulence factors and innate antibiotic resistance enables the opportunistic human pathogen Pseudomonas aeruginosa to create intractable infections. Using a nematode model, we screened for novel inhibitors of this pathogen. Aqueous extracts of three plants, Conocarpus erectus, Callistemon viminalis and Bucida buceras, were examined for their effects on P. aeruginosa killing of the nematode Caenorhabditis elegans. The results were evaluated in toxin-based and infection-based assays using P. aeruginosa strains PAO1 and PA14. The tested plant extracts prevented mortality via gut infection in approximately 60 % of the worms and caused a 50-90 % reduction in death from toxin production. All extracts inhibited nematode death by P. aeruginosa without host toxicity, indicating their potential for further development as anti-infectives.


Assuntos
Caenorhabditis elegans/microbiologia , Myrtaceae/química , Extratos Vegetais/farmacologia , Plantas Medicinais/química , Pseudomonas aeruginosa/efeitos dos fármacos , Pseudomonas aeruginosa/patogenicidade , Animais , Caenorhabditis elegans/crescimento & desenvolvimento , Modelos Animais de Doenças , Humanos , Plantas Medicinais/classificação , Infecções por Pseudomonas/microbiologia , Virulência
11.
ACS Chem Biol ; 1(9): 594-600, 2006 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-17168555

RESUMO

In bacteria, multidrug-resistance pumps (MDRs) confer resistance to chemically unrelated amphipathic toxins. A major challenge in developing efficacious antibiotics is identifying antimicrobial compounds that are not rapidly pumped out of bacterial cells. The plant antimicrobial berberine, the active component of the medicinal plants echinacea and golden seal, is a cation that is readily extruded by bacterial MDRs, thereby rendering it relatively ineffective as a therapeutic agent. However, inhibition of MDR efflux causes a substantial increase in berberine antimicrobial activity, suggesting that berberine and potentially many other compounds could be more efficacious if an effective MDR pump inhibitor could be identified. Here we show that covalently linking berberine to INF 55 , an inhibitor of Major Facilitator MDRs, results in a highly effective antimicrobial that readily accumulates in bacteria. The hybrid molecule showed good efficacy in a Caenorhabditis elegans model of enterococcal infection, curing worms of the pathogen.


Assuntos
Anti-Infecciosos/farmacologia , Infecções Bacterianas/prevenção & controle , Berberina/química , Farmacorresistência Bacteriana Múltipla , Animais , Infecções Bacterianas/tratamento farmacológico , Fenômenos Fisiológicos Bacterianos , Berberina/farmacologia , Caenorhabditis elegans , Relação Dose-Resposta a Droga , Desenho de Fármacos , Echinacea/metabolismo , Testes de Sensibilidade Microbiana , Modelos Químicos , Extratos Vegetais/metabolismo , Staphylococcus aureus/metabolismo
12.
Proc Natl Acad Sci U S A ; 103(27): 10414-10419, 2006 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-16801562

RESUMO

The alarming increase of antibiotic-resistant bacterial pathogens points to the need for novel therapeutic approaches to combat infection. To discover novel antimicrobials, we devised a screen to identify compounds that promoted the survival of the model laboratory nematode Caenorhabditis elegans infected with the human opportunistic pathogen Enterococcus faecalis. E. faecalis colonizes the nematode intestinal tract, forming a persistent lethal infection. Infected nematodes were rescued by antibiotic treatment in a dose-dependent manner, and antibiotic treatment markedly reduced the number of bacteria colonizing the nematode intestine. To facilitate high throughput screening of compound libraries, we adapted a previously developed agar-based C. elegans-E. faecalis infection assay so that it could be carried out in liquid medium in standard 96-well microtiter plates. We used this simple infection system to screen 6,000 synthetic compounds and 1,136 natural product extracts. We identified 16 compounds and 9 extracts that promoted nematode survival. Some of the compounds and extracts inhibited E. faecalis growth in vitro, but, in contrast to traditional antibiotics, the in vivo effective dose of many of these compounds was significantly lower than the minimum inhibitory concentration needed to prevent the growth of E. faecalis in vitro. Moreover, many of the compounds and extracts had little or no affect on in vitro bacterial growth. Our findings indicate that the whole-animal C. elegans screen identifies not only traditional antibiotics, but also compounds that target bacterial virulence or stimulate host defense.


Assuntos
Antibacterianos/farmacologia , Caenorhabditis elegans/efeitos dos fármacos , Caenorhabditis elegans/microbiologia , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos/métodos , Enterococcus faecalis/efeitos dos fármacos , Infecções por Bactérias Gram-Positivas/tratamento farmacológico , Animais , Antibacterianos/análise , Antibacterianos/química , Antibacterianos/uso terapêutico , Caenorhabditis elegans/genética , Caenorhabditis elegans/imunologia , Meios de Cultura , Enterococcus faecalis/fisiologia , Infecções por Bactérias Gram-Positivas/genética , Infecções por Bactérias Gram-Positivas/microbiologia , Infecções por Bactérias Gram-Positivas/patologia , Estrutura Molecular , Mutação/genética , Taxa de Sobrevida
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA