Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Angew Chem Int Ed Engl ; 57(34): 11020-11024, 2018 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-29943519

RESUMO

The macrocyclic antibiotic mangrolide A has been described to exhibit potent activity against a number of clinically important Gram-negative pathogens. Reported is the first enantioselective total synthesis of mangrolide A and derivatives. Salient features of this synthesis include a highly convergent macrocycle preparation, stereoselective synthesis of the disaccharide moiety, and two ß-selective glycosylations. The synthesis of mangrolide A and its analogues enabled the re-examination of its activity against bacterial pathogens, and only minimal activity was observed.


Assuntos
Antibacterianos/síntese química , Antibacterianos/farmacologia , Bactérias Gram-Negativas/efeitos dos fármacos , Bactérias Gram-Positivas/efeitos dos fármacos , Compostos Macrocíclicos/síntese química , Compostos Macrocíclicos/farmacologia , Antibacterianos/química , Sequência de Carboidratos , Farmacorresistência Bacteriana , Glicosilação , Compostos Macrocíclicos/química , Testes de Sensibilidade Microbiana , Estereoisomerismo
2.
Artigo em Inglês | MEDLINE | ID: mdl-28584149

RESUMO

Cadazolid (CDZ) is a new antibiotic currently in clinical development for the treatment of Clostridium difficile infections. CDZ interferes with the bacterial protein synthesis machinery. The aim of the present study was to identify resistance mechanisms for CDZ and compare the results to those obtained for linezolid (LZD) in C. difficile by whole-genome sequencing (WGS) of strains generated by in vitro passages and to those obtained for LZD-resistant clinical isolates. Clones of C. difficile 630 selected with CDZ during 46 passages had a maximally 4-fold increase in CDZ MIC, while the LZD MIC for clones selected with LZD increased up to 16-fold. CDZ cross-resistance with LZD was maximally 4-fold, and no cross-resistance with other antibiotics tested was observed. Our data suggest that there are different resistance mechanisms for CDZ and LZD in C. difficile Mutations after passages with CDZ were found in rplD (ribosomal protein L4) as well as in tra and rmt, whereas similar experiments with LZD showed mutations in rplC (ribosomal protein L3), reg, and tpr, indicating different resistance mechanisms. Although high degrees of variation between the sequenced genomes of the clinical isolates were observed, the same mutation in rplC was found in two clinical isolates with high LZD MICs. No mutations were found in the 23S rRNA genes, and attempts to isolate the cfr gene from resistant clinical isolates were unsuccessful. Analysis of 50% inhibitory concentrations (IC50s) determined in in vitro transcription/translation assays performed with C. difficile cell extracts from passaged clones correlated well with the MIC values for all antibiotics tested, indicating that the ribosomal mutations are causing the resistant phenotype.


Assuntos
Antibacterianos/farmacologia , Clostridioides difficile/efeitos dos fármacos , Clostridioides difficile/genética , Farmacorresistência Bacteriana/genética , Linezolida/farmacologia , Oxazolidinonas/farmacologia , Inibidores da Síntese de Proteínas/farmacologia , Proteínas Ribossômicas/genética , Sequência de Aminoácidos , Clostridioides difficile/isolamento & purificação , Farmacorresistência Bacteriana/fisiologia , Genoma Bacteriano/genética , Humanos , Testes de Sensibilidade Microbiana , RNA Ribossômico 23S/genética , Proteína Ribossômica L3 , Análise de Sequência de DNA
3.
Antimicrob Agents Chemother ; 60(1): 628-31, 2016 01.
Artigo em Inglês | MEDLINE | ID: mdl-26503650

RESUMO

The promotion of colonization with vancomycin-resistant enterococci (VRE) is one potential side effect during treatment of Clostridium difficile-associated diarrhea (CDAD), resulting from disturbances in gut microbiota. Cadazolid (CDZ) is an investigational antibiotic with potent in vitro activity against C. difficile and against VRE and is currently in clinical development for the treatment of CDAD. We report that CDZ treatment did not lead to intestinal VRE overgrowth in mice.


Assuntos
Antibacterianos/efeitos adversos , Clostridioides difficile/efeitos dos fármacos , Diarreia/tratamento farmacológico , Enterocolite Pseudomembranosa/tratamento farmacológico , Oxazolidinonas/farmacologia , Vancomicina/efeitos adversos , Aminoglicosídeos/farmacologia , Animais , Antibacterianos/administração & dosagem , Clostridioides difficile/crescimento & desenvolvimento , Clostridioides difficile/patogenicidade , Contagem de Colônia Microbiana , Diarreia/etiologia , Diarreia/microbiologia , Diarreia/patologia , Enterocolite Pseudomembranosa/etiologia , Enterocolite Pseudomembranosa/microbiologia , Enterocolite Pseudomembranosa/patologia , Fidaxomicina , Intestinos/efeitos dos fármacos , Intestinos/microbiologia , Intestinos/patologia , Metronidazol/farmacologia , Camundongos , Testes de Sensibilidade Microbiana , Infecções Estreptocócicas/tratamento farmacológico , Infecções Estreptocócicas/microbiologia , Infecções Estreptocócicas/patologia , Resultado do Tratamento , Vancomicina/administração & dosagem , Resistência a Vancomicina , Enterococos Resistentes à Vancomicina/efeitos dos fármacos , Enterococos Resistentes à Vancomicina/crescimento & desenvolvimento , Enterococos Resistentes à Vancomicina/patogenicidade
4.
J Virol ; 89(11): 5876-82, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25787289

RESUMO

UNLABELLED: The hepatitis C virus (HCV; genus Hepacivirus) is a highly relevant human pathogen. Unique hepaciviruses (HV) were discovered recently in animal hosts. The direct ancestor of HCV has not been found, but the genetically most closely related animal HVs exist in horses. To investigate whether other peridomestic animals also carry HVs, we analyzed sera from Ghanaian cattle for HVs by reverse transcription-PCR (RT-PCR). Nine of 106 specimens from different sampling sites contained HV RNA (8.5%) at median viral loads of 1.6 × 10(5) copies/ml. Infection seemed unrelated to cattle age and gender. Near-full-genome sequencing of five representative viruses confirmed taxonomic classifications. Cattle HVs formed two distinct phylogenetic lineages that differed by up to 17.7% on the nucleotide level in the polyprotein-encoding region, suggesting cocirculation of different virus subtypes. A conserved microRNA122-binding site in the 5' internal ribosomal entry site suggested liver tropism of cattle HVs. Phylogenetic analyses suggested the circulation of HVs in cattle for several centuries. Cattle HVs were genetically highly divergent from all other HVs, including HCV. HVs from genetically related equine and bovine hosts were not monophyletic, corroborating host shifts during the evolution of the genus Hepacivirus. Similar to equine HVs, the genetic diversity of cattle HVs was low compared to that of HCV genotypes. This suggests an influence of the human-modified ecology of peridomestic animals on virus diversity. Further studies should investigate the occurrence of cattle HVs in other geographic areas and breeds, virus pathogenicity in cattle, and the potential exposure of human risk groups, such as farmers, butchers, and abattoir workers. IMPORTANCE: HCV (genus Hepacivirus) is a major human pathogen, causing liver failure and cancer. Unique hepaciviruses (HVs) were discovered over the last few years in animals, but the direct ancestor of HCV has not been found. The animal HV most closely related to HCV so far originated from horses, suggesting that other livestock animals also harbor HVs. Therefore, we investigated African cattle and discovered previously unknown HVs at high prevalence and viral loads. Because of the agricultural importance of cattle, it may be relevant to investigate HV pathogenicity. The frequent exposure of humans to cattle also may warrant investigations of the zoonotic potential of these viruses. Evolutionary analyses suggested that cattle HVs have existed for centuries. Despite the genetic relatedness of their animal hosts, HVs from cattle and horses were not phylogenetically related, corroborating frequent host shifts during the evolution of the genus Hepacivirus.


Assuntos
Doenças dos Bovinos/virologia , Hepacivirus/classificação , Hepacivirus/isolamento & purificação , Hepatite C/veterinária , Animais , Bovinos , Análise por Conglomerados , Variação Genética , Genoma Viral , Genótipo , Gana , Hepacivirus/genética , Hepatite C/virologia , Dados de Sequência Molecular , Filogenia , RNA Viral/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de DNA , Homologia de Sequência , Soro/virologia , Carga Viral
5.
J Infect Dis ; 211(6): 889-97, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25057042

RESUMO

Middle East respiratory syndrome coronavirus (MERS-CoV) infection is associated with a high case-fatality rate, and the potential pandemic spread of the virus is a public health concern. The spike protein of MERS-CoV (MERS-S) facilitates viral entry into host cells, which depends on activation of MERS-S by cellular proteases. Proteolytic activation of MERS-S during viral uptake into target cells has been demonstrated. However, it is unclear whether MERS-S is also cleaved during S protein synthesis in infected cells and whether cleavage is required for MERS-CoV infectivity. Here, we show that MERS-S is processed by proprotein convertases in MERS-S-transfected and MERS-CoV-infected cells and that several RXXR motifs located at the border between the surface and transmembrane subunit of MERS-S are required for efficient proteolysis. However, blockade of proprotein convertases did not impact MERS-S-dependent transduction of target cells expressing high amounts of the viral receptor, DPP4, and did not modulate MERS-CoV infectivity. These results show that MERS-S is a substrate for proprotein convertases and demonstrate that processing by these enzymes is dispensable for S protein activation. Efforts to inhibit MERS-CoV infection by targeting host cell proteases should therefore focus on enzymes that process MERS-S during viral uptake into target cells.


Assuntos
Coronavirus/fisiologia , Pró-Proteína Convertases/antagonistas & inibidores , Glicoproteína da Espícula de Coronavírus/metabolismo , Sequência de Aminoácidos , Antivirais/farmacologia , Avaliação Pré-Clínica de Medicamentos , Células HEK293 , Humanos , Dados de Sequência Molecular , Inibidores de Proteases/farmacologia , Processamento de Proteína Pós-Traducional , Proteólise , Glicoproteína da Espícula de Coronavírus/química , Internalização do Vírus
6.
PLoS Pathog ; 9(6): e1003438, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23818848

RESUMO

Hepatitis C virus (HCV) is among the most relevant causes of liver cirrhosis and hepatocellular carcinoma. Research is complicated by a lack of accessible small animal models. The systematic investigation of viruses of small mammals could guide efforts to establish such models, while providing insight into viral evolutionary biology. We have assembled the so-far largest collection of small-mammal samples from around the world, qualified to be screened for bloodborne viruses, including sera and organs from 4,770 rodents (41 species); and sera from 2,939 bats (51 species). Three highly divergent rodent hepacivirus clades were detected in 27 (1.8%) of 1,465 European bank voles (Myodes glareolus) and 10 (1.9%) of 518 South African four-striped mice (Rhabdomys pumilio). Bats showed anti-HCV immunoblot reactivities but no virus detection, although the genetic relatedness suggested by the serologic results should have enabled RNA detection using the broadly reactive PCR assays developed for this study. 210 horses and 858 cats and dogs were tested, yielding further horse-associated hepaciviruses but none in dogs or cats. The rodent viruses were equidistant to HCV, exceeding by far the diversity of HCV and the canine/equine hepaciviruses taken together. Five full genomes were sequenced, representing all viral lineages. Salient genome features and distance criteria supported classification of all viruses as hepaciviruses. Quantitative RT-PCR, RNA in-situ hybridisation, and histopathology suggested hepatic tropism with liver inflammation resembling hepatitis C. Recombinant serology for two distinct hepacivirus lineages in 97 bank voles identified seroprevalence rates of 8.3 and 12.4%, respectively. Antibodies in bank vole sera neither cross-reacted with HCV, nor the heterologous bank vole hepacivirus. Co-occurrence of RNA and antibodies was found in 3 of 57 PCR-positive bank vole sera (5.3%). Our data enable new hypotheses regarding HCV evolution and encourage efforts to develop rodent surrogate models for HCV.


Assuntos
Evolução Molecular , Genoma Viral , Hepacivirus , Anticorpos Anti-Hepatite C/sangue , Hepatite C , Hepatite Animal , RNA Viral , Roedores , Animais , Sequência de Bases , Gatos , Cães , Hepacivirus/genética , Hepacivirus/metabolismo , Hepatite C/sangue , Hepatite C/genética , Hepatite C/virologia , Hepatite Animal/sangue , Hepatite Animal/genética , Hepatite Animal/virologia , Cavalos , Dados de Sequência Molecular , RNA Viral/sangue , RNA Viral/genética , Roedores/sangue , Roedores/virologia
7.
Emerg Infect Dis ; 20(4): 552-9, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24655412

RESUMO

Middle East respiratory syndrome coronavirus (MERS-CoV) has caused an ongoing outbreak of severe acute respiratory tract infection in humans in the Arabian Peninsula since 2012. Dromedary camels have been implicated as possible viral reservoirs. We used serologic assays to analyze 651 dromedary camel serum samples from the United Arab Emirates; 151 of 651 samples were obtained in 2003, well before onset of the current epidemic, and 500 serum samples were obtained in 2013. Recombinant spike protein-specific immunofluorescence and virus neutralization tests enabled clear discrimination between MERS-CoV and bovine CoV infections. Most (632/651, 97.1%) camels had antibodies against MERS-CoV. This result included all 151 serum samples obtained in 2003. Most (389/651, 59.8%) serum samples had MERS-CoV-neutralizing antibody titers >1,280. Dromedary camels from the United Arab Emirates were infected at high rates with MERS-CoV or a closely related, probably conspecific, virus long before the first human MERS cases.


Assuntos
Anticorpos Neutralizantes/imunologia , Camelus/imunologia , Camelus/virologia , Infecções por Coronavirus/imunologia , Coronavirus/imunologia , Infecções Respiratórias/imunologia , Animais , Anticorpos Antivirais/imunologia , Infecções por Coronavirus/epidemiologia , Testes de Neutralização/métodos , Infecções Respiratórias/epidemiologia , Infecções Respiratórias/virologia , Síndrome , Emirados Árabes Unidos/epidemiologia
8.
Antimicrob Agents Chemother ; 58(2): 901-8, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24277035

RESUMO

Cadazolid is a new oxazolidinone-type antibiotic currently in clinical development for the treatment of Clostridium difficile-associated diarrhea. Here, we report investigations on the mode of action and the propensity for spontaneous resistance development in C. difficile strains. Macromolecular labeling experiments indicated that cadazolid acts as a potent inhibitor of protein synthesis, while inhibition of DNA synthesis was also observed, albeit only at substantially higher concentrations of the drug. Strong inhibition of protein synthesis was also obtained in strains resistant to linezolid, in agreement with low MICs against such strains. Inhibition of protein synthesis was confirmed in coupled transcription/translation assays using extracts from different C. difficile strains, including strains resistant to linezolid, while inhibitory effects in DNA topoisomerase assays were weak or not detectable under the assay conditions. Spontaneous resistance frequencies of cadazolid were low in all strains tested (generally <10(-10) at 2× to 4× the MIC), and in multiple-passage experiments (up to 13 passages) MICs did not significantly increase. Furthermore, no cross-resistance was observed, as cadazolid retained potent activity against strains resistant or nonsusceptible to linezolid, fluoroquinolones, and the new antibiotic fidaxomicin. In conclusion, the data presented here indicate that cadazolid acts primarily by inhibition of protein synthesis, with weak inhibition of DNA synthesis as a potential second mode of action, and suggest a low potential for spontaneous resistance development.


Assuntos
Antibacterianos/farmacologia , Clostridioides difficile/efeitos dos fármacos , Farmacorresistência Bacteriana/genética , Biossíntese de Proteínas/efeitos dos fármacos , Acetamidas/farmacologia , Aminoglicosídeos/farmacologia , Clostridioides difficile/genética , Clostridioides difficile/crescimento & desenvolvimento , Clostridioides difficile/metabolismo , DNA Girase/genética , DNA Girase/metabolismo , Farmacorresistência Bacteriana/efeitos dos fármacos , Fidaxomicina , Fluoroquinolonas/farmacologia , Linezolida , Testes de Sensibilidade Microbiana , Oxazolidinonas/farmacologia , Biossíntese de Proteínas/genética , RNA/antagonistas & inibidores , RNA/biossíntese , Proteínas Recombinantes , Frações Subcelulares/química , Frações Subcelulares/metabolismo , Transcrição Gênica/efeitos dos fármacos , Vancomicina/farmacologia
9.
Antimicrob Agents Chemother ; 58(2): 892-900, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24277020

RESUMO

Clostridium difficile is a leading cause of health care-associated diarrhea with significant morbidity and mortality, and new options for the treatment of C. difficile-associated diarrhea (CDAD) are needed. Cadazolid is a new oxazolidinone-type antibiotic that is currently in clinical development for treatment of CDAD. Here, we report the in vitro and in vivo antibacterial evaluation of cadazolid against C. difficile. Cadazolid showed potent in vitro activity against C. difficile with a MIC range of 0.125 to 0.5 µg/ml, including strains resistant to linezolid and fluoroquinolones. In time-kill kinetics experiments, cadazolid showed a bactericidal effect against C. difficile isolates, with >99.9% killing in 24 h, and was more bactericidal than vancomycin. In contrast to metronidazole and vancomycin, cadazolid strongly inhibited de novo toxin A and B formation in stationary-phase cultures of toxigenic C. difficile. Cadazolid also inhibited C. difficile spore formation substantially at growth-inhibitory concentrations. In the hamster and mouse models for CDAD, cadazolid was active, conferring full protection from diarrhea and death with a potency similar to that of vancomycin. These findings support further investigations of cadazolid for the treatment of CDAD.


Assuntos
Antibacterianos/farmacologia , Clostridioides difficile/efeitos dos fármacos , Infecções por Clostridium/tratamento farmacológico , Enterocolite Pseudomembranosa/tratamento farmacológico , Oxazolidinonas/farmacologia , Esporos Bacterianos/efeitos dos fármacos , Acetamidas/farmacologia , Animais , Proteínas de Bactérias/antagonistas & inibidores , Proteínas de Bactérias/biossíntese , Toxinas Bacterianas/antagonistas & inibidores , Toxinas Bacterianas/biossíntese , Clostridioides difficile/crescimento & desenvolvimento , Clostridioides difficile/metabolismo , Infecções por Clostridium/microbiologia , Infecções por Clostridium/mortalidade , Cricetinae , Enterocolite Pseudomembranosa/microbiologia , Enterocolite Pseudomembranosa/mortalidade , Enterotoxinas/antagonistas & inibidores , Enterotoxinas/biossíntese , Feminino , Fluoroquinolonas/farmacologia , Humanos , Linezolida , Masculino , Metronidazol/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Testes de Sensibilidade Microbiana , Esporos Bacterianos/crescimento & desenvolvimento , Análise de Sobrevida , Vancomicina/farmacologia
10.
RSC Med Chem ; 15(6): 2146-2159, 2024 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-38911172

RESUMO

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused a global pandemic of coronavirus disease (COVID-19) since its emergence in December 2019. As of January 2024, there has been over 774 million reported cases and 7 million deaths worldwide. While vaccination efforts have been successful in reducing the severity of the disease and decreasing the transmission rate, the development of effective therapeutics against SARS-CoV-2 remains a critical need. The main protease (Mpro) of SARS-CoV-2 is an essential enzyme required for viral replication and has been identified as a promising target for drug development. In this study, we report the identification of novel Mpro inhibitors, using a combination of deep reinforcement learning for de novo drug design with 3D pharmacophore/shape-based alignment and privileged fragment match count scoring components followed by hit expansions and molecular docking approaches. Our experimentally validated results show that 3 novel series exhibit potent inhibitory activity against SARS-CoV-2 Mpro, with IC50 values ranging from 1.3 µM to 2.3 µM and a high degree of selectivity. These findings represent promising starting points for the development of new antiviral therapies against COVID-19.

11.
J Med Chem ; 67(11): 9465-9484, 2024 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-38753983

RESUMO

Clostridioides difficile (C. difficile) is one of the leading causes of healthcare-associated infections worldwide. The increasing incidence of strains resistant to currently available therapies highlights the need for alternative treatment options with a novel mode of action. Oxazolidinones that are connected to a quinolone moiety with a pyrrolidine linker, such as compound 1, are reported to exhibit potent broadspectrum antibacterial activity. In an effort to optimize this class of compounds for the treatment of C. difficile infection (CDI), we have identified cadazolid (9), a first-in-class quinoxolidinone antibiotic, which is a potent inhibitor of C. difficile protein synthesis. In order to achieve narrow-spectrum coverage of clinically most relevant strains without affecting the gut microbiota, an emphasis was placed on abolishing activity against commensals of the intestinal microbiome while retaining good coverage of pathogenic C. difficile, including hypervirulent and epidemic strains.


Assuntos
Antibacterianos , Clostridioides difficile , Infecções por Clostridium , Testes de Sensibilidade Microbiana , Relação Estrutura-Atividade , Antibacterianos/farmacologia , Antibacterianos/química , Antibacterianos/uso terapêutico , Antibacterianos/síntese química , Clostridioides difficile/efeitos dos fármacos , Infecções por Clostridium/tratamento farmacológico , Animais , Humanos , Descoberta de Drogas , Microbioma Gastrointestinal/efeitos dos fármacos , Camundongos , Oxazolidinonas
12.
Acta Crystallogr D Struct Biol ; 80(Pt 2): 123-136, 2024 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-38289714

RESUMO

To identify starting points for therapeutics targeting SARS-CoV-2, the Paul Scherrer Institute and Idorsia decided to collaboratively perform an X-ray crystallographic fragment screen against its main protease. Fragment-based screening was carried out using crystals with a pronounced open conformation of the substrate-binding pocket. Of 631 soaked fragments, a total of 29 hits bound either in the active site (24 hits), a remote binding pocket (three hits) or at crystal-packing interfaces (two hits). Notably, two fragments with a pose that was sterically incompatible with a more occluded crystal form were identified. Two isatin-based electrophilic fragments bound covalently to the catalytic cysteine residue. The structures also revealed a surprisingly strong influence of the crystal form on the binding pose of three published fragments used as positive controls, with implications for fragment screening by crystallography.


Assuntos
COVID-19 , SARS-CoV-2 , Humanos , Domínio Catalítico , Proteases 3C de Coronavírus , Cristalografia por Raios X
13.
J Bacteriol ; 195(18): 4067-73, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23836867

RESUMO

Bactericidal antibiotics kill by different mechanisms as a result of a specific interaction with their cellular targets. Over the past few years, alternative explanations for cidality have been proposed based on a postulated common pathway, depending on the intracellular production of reactive oxygen species. Detection of hydroxyl radicals relies on staining with specific fluorescent dyes that can penetrate the cell and are detected using flow cytometry. Flow cytometry has become an important tool in microbiology to study characteristics of individual cells within large heterogeneous cellular populations. We show here that Escherichia coli treated with different bactericidal antibiotics exhibits increased autofluorescence when analyzed by flow cytometry. We present evidence suggesting that this change in autofluorescence is caused by altered cell morphology upon antibiotic treatment. Consistent with this view, mutant cells that fail to elongate upon norfloxacin treatment show no increased auto-fluorescence response. Finally, we present data demonstrating that changes in autofluorescence can impact the results with fluorescent probes when using flow cytometry and confound the findings obtained with specific dyes. In summary, recent findings that correlate the exposure to cidal antibiotics with the production of reactive oxygen species need to be reconsidered in light of such changes in autofluorescence. Conclusive evidence for an increase of hydroxyl radicals after treatment with such drugs is still missing.


Assuntos
Antibacterianos/farmacologia , Escherichia coli/efeitos dos fármacos , Escherichia coli/fisiologia , Citometria de Fluxo/métodos , Corantes Fluorescentes/farmacologia , Radical Hidroxila/metabolismo , Ampicilina/metabolismo , Ampicilina/farmacologia , Antibacterianos/metabolismo , Escherichia coli/genética , Escherichia coli/ultraestrutura , Fluorescência , Corantes Fluorescentes/metabolismo , Testes de Sensibilidade Microbiana , Mutação , Norfloxacino/metabolismo , Norfloxacino/farmacologia , Espécies Reativas de Oxigênio/metabolismo
14.
J Gen Virol ; 93(Pt 12): 2601-2605, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22956738

RESUMO

STAT1-deficient mice are more susceptible to infection with severe acute respiratory syndrome coronavirus (SARS-CoV) than type I interferon (IFN) receptor-deficient mice. We used mice lacking functional receptors for both type I and type III IFN (double knockout, dKO) to evaluate the possibility that type III IFN plays a decisive role in SARS-CoV protection. We found that viral peak titres in lungs of dKO and STAT1-deficient mice were similar, but significantly higher than in wild-type mice. The kinetics of viral clearance from the lung were also comparable in dKO and STAT1-deficient mice. Surprisingly, however, infected dKO mice remained healthy, whereas infected STAT1-deficient mice developed liver pathology and eventually succumbed to neurological disease. Our data suggest that the failure of STAT1-deficient mice to control initial SARS-CoV replication efficiently in the lung is due to impaired type I and type III IFN signalling, whereas the failure to control subsequent systemic viral spread is due to unrelated defects in STAT1-deficient mice.


Assuntos
Interferon Tipo I/imunologia , Interferons/imunologia , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/patogenicidade , Animais , Pulmão/imunologia , Pulmão/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor de Interferon alfa e beta/deficiência , Receptor de Interferon alfa e beta/genética , Receptor de Interferon alfa e beta/imunologia , Receptores de Interferon/deficiência , Receptores de Interferon/genética , Receptores de Interferon/imunologia , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/imunologia , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/fisiologia , Fator de Transcrição STAT1/deficiência , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/imunologia , Replicação Viral/imunologia
15.
PLoS Pathog ; 6(3): e1000804, 2010 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-20300602

RESUMO

During long-term cystic fibrosis lung infections, Pseudomonas aeruginosa undergoes genetic adaptation resulting in progressively increased persistence and the generation of adaptive colony morphotypes. This includes small colony variants (SCVs), auto-aggregative, hyper-adherent cells whose appearance correlates with poor lung function and persistence of infection. The SCV morphotype is strongly linked to elevated levels of cyclic-di-GMP, a ubiquitous bacterial second messenger that regulates the transition between motile and sessile, cooperative lifestyles. A genetic screen in PA01 for SCV-related loci identified the yfiBNR operon, encoding a tripartite signaling module that regulates c-di-GMP levels in P. aeruginosa. Subsequent analysis determined that YfiN is a membrane-integral diguanylate cyclase whose activity is tightly controlled by YfiR, a small periplasmic protein, and the OmpA/Pal-like outer-membrane lipoprotein YfiB. Exopolysaccharide synthesis was identified as the principal downstream target for YfiBNR, with increased production of Pel and Psl exopolysaccharides responsible for many characteristic SCV behaviors. An yfi-dependent SCV was isolated from the sputum of a CF patient. Consequently, the effect of the SCV morphology on persistence of infection was analyzed in vitro and in vivo using the YfiN-mediated SCV as a representative strain. The SCV strain exhibited strong, exopolysaccharide-dependent resistance to nematode scavenging and macrophage phagocytosis. Furthermore, the SCV strain effectively persisted over many weeks in mouse infection models, despite exhibiting a marked fitness disadvantage in vitro. Exposure to sub-inhibitory concentrations of antibiotics significantly decreased both the number of suppressors arising, and the relative fitness disadvantage of the SCV mutant in vitro, suggesting that the SCV persistence phenotype may play a more important role during antimicrobial chemotherapy. This study establishes YfiBNR as an important player in P. aeruginosa persistence, and implicates a central role for c-di-GMP, and by extension the SCV phenotype in chronic infections.


Assuntos
Proteínas da Membrana Bacteriana Externa/genética , GMP Cíclico/análogos & derivados , Proteínas Periplásmicas/genética , Fósforo-Oxigênio Liases/genética , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/genética , Animais , Proteínas da Membrana Bacteriana Externa/metabolismo , Caenorhabditis elegans , Células Cultivadas , GMP Cíclico/metabolismo , Elementos de DNA Transponíveis/genética , Proteínas de Escherichia coli , Macrófagos/citologia , Macrófagos/imunologia , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Mutagênese/fisiologia , Óperon/genética , Proteínas Periplásmicas/metabolismo , Fagocitose/fisiologia , Fenótipo , Fósforo-Oxigênio Liases/metabolismo , Pneumonia Bacteriana/enzimologia , Pneumonia Bacteriana/imunologia , Pneumonia Bacteriana/microbiologia , Infecções por Pseudomonas/enzimologia , Infecções por Pseudomonas/imunologia , Pseudomonas aeruginosa/crescimento & desenvolvimento , Pseudomonas aeruginosa/metabolismo , Sistemas do Segundo Mensageiro/fisiologia
16.
Bioorg Med Chem Lett ; 22(21): 6705-11, 2012 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-23006603

RESUMO

A series of 2-amino-[1,8]-naphthyridine-3-carboxamides (ANCs) with potent inhibition of bacterial NAD(+)-dependent DNA ligases (LigAs) evolved from a 2,4-diaminopteridine derivative discovered by HTS. The design was guided by several highly resolved X-ray structures of our inhibitors in complex with either Streptococcus pneumoniae or Escherichia coli LigA. The structure-activity-relationship based on the ANC scaffold is discussed. The in-depth characterization of 2-amino-6-bromo-7-(trifluoromethyl)-[1,8]-naphthyridine-3-carboxamide, which displayed promising in vitro (MIC Staphylococcus aureus 1 mg/L) and in vivo anti-staphylococcal activity, is presented.


Assuntos
Antibacterianos/síntese química , Antibacterianos/farmacologia , DNA Ligases/antagonistas & inibidores , Desenho de Fármacos , Staphylococcus/efeitos dos fármacos , Animais , Antibacterianos/química , Antibacterianos/uso terapêutico , Cristalografia por Raios X , DNA Bacteriano/antagonistas & inibidores , Concentração Inibidora 50 , Camundongos , Testes de Sensibilidade Microbiana , Estrutura Molecular , Ratos , Infecções Estafilocócicas/tratamento farmacológico , Relação Estrutura-Atividade
17.
PLoS One ; 17(3): e0264450, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35259191

RESUMO

Eidolon helvum bats are reservoir hosts for highly pathogenic lyssaviruses often showing limited disease upon natural infection. An enhanced antiviral interferon (IFN) response combined with reduced inflammation might be linked to the apparent virus tolerance in bats. Lyssavirus phosphoproteins inhibit the IFN response with virus strain-specific efficiency. To date, little is known regarding the lyssavirus P-dependent anti-IFN countermeasures in bats, mainly due to a lack of in vitro tools. By using E. helvum bat cell cultures in a newly established bat-specific IFN-promoter activation assay, we analyzed the IFN-ß inhibitory activity of multiple lyssavirus P in E. helvum compared to human cells. Initial virus infection studies with a recently isolated E. helvum-borne Lagos bat virus street strain from Ghana showed enhanced LBV propagation in an E. helvum lung cell line compared to human A549 lung cells at later time points suggesting effective viral countermeasures against cellular defense mechanisms. A direct comparison of the IFN-ß inhibitory activity of the LBV-GH P protein with other lyssavirus P proteins showed that LBV-GH P and RVP both strongly inhibited the bat IFN-ß promotor activation (range 75-90%) in EidLu/20.2 and an E. helvum kidney cell line. Conversely, LBV-GH P blocked the activation of the human IFN-ß promoter less efficiently compared to a prototypic Rabies virus P protein (range LBV P 52-68% vs RVP 71-95%) in two different human cell lines (HEK-293T, A549). The same pattern was seen for two prototypic LBV P variants suggesting an overall reduced LBV P IFN-ß inhibitory activity in human cells as compared to E. helvum bat cells. Increased IFN-ß inhibition by lyssavirus P in reservoir host cells might be a result of host-specific adaptation processes towards an enhanced IFN response in bat cells.


Assuntos
Quirópteros , Lyssavirus , Infecções por Rhabdoviridae , Animais , Anticorpos Antivirais , Humanos , Interferon beta , Nigéria , Fosfoproteínas
18.
ChemMedChem ; 16(5): 891-897, 2021 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-33236408

RESUMO

Ribosomal protein synthesis is an important target in antibacterial drug discovery. Numerous natural products have served as starting points for the development of antibiotics. We report here the total synthesis of xenocoumacin 1, a natural product that binds to 16S ribosomal RNA at a highly conserved region, as well as analogues thereof. Preliminary structure-activity relationship studies were aimed at understanding and modulating the selectivity between eukaryotic and prokaryotic ribosomes. Modifications were mainly tolerated in the aromatic region. Whole-cell activity against Gram-negative bacteria is limited by efflux and penetration, as demonstrated in genetically modified strains of E. coli. Analogues with high selectivity for eukaryotic ribosomes were identified, but it was not possible to obtain inhibitors selective for bacterial protein synthesis. Achieving high selectivity (albeit not the desired one) was thus possible despite the high homology between eukaryotic and prokaryotic ribosomes in the binding region.


Assuntos
Antibacterianos/farmacologia , Benzopiranos/farmacologia , Escherichia coli/efeitos dos fármacos , Proteínas Ribossômicas/antagonistas & inibidores , Antibacterianos/química , Benzopiranos/química , Cristalografia por Raios X , Relação Dose-Resposta a Droga , Escherichia coli/metabolismo , Testes de Sensibilidade Microbiana , Modelos Moleculares , Estrutura Molecular , Proteínas Ribossômicas/biossíntese , Relação Estrutura-Atividade
19.
Mol Microbiol ; 72(6): 1500-16, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19460094

RESUMO

Biofilms are communities of surface-attached, matrix-embedded microbial cells that can resist antimicrobial chemotherapy and contribute to persistent infections. Using an Escherichia coli biofilm model we found that exposure of bacteria to subinhibitory concentrations of ribosome-targeting antibiotics leads to strong biofilm induction. We present evidence that this effect is elicited by the ribosome in response to translational stress. Biofilm induction involves upregulation of the polysaccharide adhesin poly-beta-1,6-N-acetyl-glucosamine (poly-GlcNAc) and two components of the poly-GlcNAc biosynthesis machinery, PgaA and PgaD. Poly-GlcNAc control depends on the bacterial signalling molecules guanosine-bis 3', 5'(diphosphate) (ppGpp) and bis-(3'-5')-cyclic di-GMP (c-di-GMP). Treatment with translation inhibitors causes a ppGpp hydrolase (SpoT)-mediated reduction of ppGpp levels, resulting in specific derepression of PgaA. Maximal induction of PgaD and poly-GlcNAc synthesis requires the production of c-di-GMP by the dedicated diguanylate cyclase YdeH. Our results identify a novel regulatory mechanism that relies on ppGpp signalling to relay information about ribosomal performance to the Pga machinery, thereby inducing adhesin production and biofilm formation. Based on the important synergistic roles of ppGpp and c-di-GMP in this process, we suggest that interference with bacterial second messenger signalling might represent an effective means for biofilm control during chronic infections.


Assuntos
Biofilmes/crescimento & desenvolvimento , GMP Cíclico/análogos & derivados , Escherichia coli/fisiologia , Guanosina Tetrafosfato/metabolismo , Ribossomos/efeitos dos fármacos , Sistemas do Segundo Mensageiro , Adesinas Bacterianas/metabolismo , Antibacterianos/farmacologia , GMP Cíclico/metabolismo , Escherichia coli/efeitos dos fármacos , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Regulação Bacteriana da Expressão Gênica , Fósforo-Oxigênio Liases/genética , Fósforo-Oxigênio Liases/metabolismo , Biossíntese de Proteínas/efeitos dos fármacos , Pirofosfatases/genética , Pirofosfatases/metabolismo , Processamento Pós-Transcricional do RNA , beta-Glucanas/metabolismo
20.
Chemotherapy ; 56(4): 318-24, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20714150

RESUMO

OBJECTIVES: In this study, the in vitro antimicrobial activity and spectrum of new dimeric compounds derived from the cyanobacterial alkaloid nostocarboline were investigated. The mechanism of action and selectivity to bacteria were studied and compared to the cationic antiseptic chlorhexidine. METHODS: Minimal inhibitory concentrations were determined against clinical isolates and against a panel of microbial reference strains using the CLSI microdilution method. Bacterial membrane damage was addressed by measuring ATP leakage and the mode of action was investigated in Escherichia coli reporter strains. Selectivity was tested by a cytotoxicity assay using MTS. RESULTS: The antimicrobial potency of dimers varied with length of the hydrophobic linker. The most potent compounds, NCD9 and NCD10, had a C10 and C12 linker, respectively, and showed strong activity against Gram-positive bacteria, notably methicillin-resistant Staphylococcus aureus strains. Similar to chlorhexidine, these compounds showed a rapid concentration-dependent bactericidal effect, which correlated with membrane damage as indicated by ATP leakage. NCD9, in contrast to NCD10 and chlorhexidine, lacked activity against yeast strains and showed low cytotoxicity in CHO cells indicating a high degree of selectivity. In E. coli reporter strains, NCD9 induced the DegP response pathway as well as the SOS response, suggesting interaction with both the cell envelope and DNA metabolism. CONCLUSIONS: The results presented in this report indicate the potential of this new class of cationic antimicrobial compounds for the design of potent and selective antibacterials with low cytotoxicity.


Assuntos
Alcaloides/farmacologia , Anti-Infecciosos Locais/farmacologia , Carbolinas/farmacologia , Clorexidina/farmacologia , Bactérias Gram-Negativas/efeitos dos fármacos , Bactérias Gram-Positivas/efeitos dos fármacos , Animais , Antibacterianos/farmacologia , Células CHO , Carbolinas/química , Ciprofloxacina/farmacologia , Ciprofloxacina/uso terapêutico , Cricetinae , Cricetulus , Dimerização , Escherichia coli/efeitos dos fármacos , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Testes de Sensibilidade Microbiana
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA