Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Molecules ; 26(11)2021 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-34200016

RESUMO

The increase in antibacterial resistance is a serious challenge for both the health and defence sectors and there is a need for both novel antibacterial targets and antibacterial strategies. RNA degradation and ribonucleases, such as the essential endoribonuclease RNase E, encoded by the rne gene, are emerging as potential antibacterial targets while antisense oligonucleotides may provide alternative antibacterial strategies. As rne mRNA has not been previously targeted using an antisense approach, we decided to explore using antisense oligonucleotides to target the translation initiation region of the Escherichia coli rne mRNA. Antisense oligonucleotides were rationally designed and were synthesised as locked nucleic acid (LNA) gapmers to enable inhibition of rne mRNA translation through two mechanisms. Either LNA gapmer binding could sterically block translation and/or LNA gapmer binding could facilitate RNase H-mediated cleavage of the rne mRNA. This may prove to be an advantage over the majority of previous antibacterial antisense oligonucleotide approaches which used oligonucleotide chemistries that restrict the mode-of-action of the antisense oligonucleotide to steric blocking of translation. Using an electrophoretic mobility shift assay, we demonstrate that the LNA gapmers bind to the translation initiation region of E. coli rne mRNA. We then use a cell-free transcription translation reporter assay to show that this binding is capable of inhibiting translation. Finally, in an in vitro RNase H cleavage assay, the LNA gapmers facilitate RNase H-mediated mRNA cleavage. Although the challenges of antisense oligonucleotide delivery remain to be addressed, overall, this work lays the foundations for the development of a novel antibacterial strategy targeting rne mRNA with antisense oligonucleotides.


Assuntos
Antibacterianos/farmacologia , Endorribonucleases/genética , Escherichia coli/enzimologia , Oligonucleotídeos/farmacologia , Sistema Livre de Células , Endorribonucleases/antagonistas & inibidores , Escherichia coli/efeitos dos fármacos , Proteínas de Escherichia coli/genética , Oligonucleotídeos/síntese química , Iniciação Traducional da Cadeia Peptídica/efeitos dos fármacos , RNA Mensageiro/antagonistas & inibidores
2.
PLoS One ; 16(3): e0248119, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33764972

RESUMO

Burkholderia pseudomallei is a soil-dwelling organism present throughout the tropics. It is the causative agent of melioidosis, a disease that is believed to kill 89,000 people per year. It is naturally resistant to many antibiotics, requiring at least two weeks of intravenous treatment with ceftazidime, imipenem or meropenem followed by 6 months of orally delivered co-trimoxazole. This places a large treatment burden on the predominantly middle-income nations where the majority of disease occurs. We have established a high-throughput assay for compounds that could be used as a co-therapy to potentiate the effect of ceftazidime, using the related non-pathogenic bacterium Burkholderia thailandensis as a surrogate. Optimization of the assay gave a Z' factor of 0.68. We screened a library of 61,250 compounds and identified 29 compounds with a pIC50 (-log10(IC50)) greater than five. Detailed investigation allowed us to down select to six "best in class" compounds, which included the licensed drug chloroxine. Co-treatment of B. thailandensis with ceftazidime and chloroxine reduced culturable cell numbers by two orders of magnitude over 48 hours, compared to treatment with ceftazidime alone. Hit expansion around chloroxine was performed using commercially available compounds. Minor modifications to the structure abolished activity, suggesting that chloroxine likely acts against a specific target. Finally, an initial study demonstrates the utility of chloroxine to act as a co-therapy to potentiate the effect of ceftazidime against B. pseudomallei. This approach successfully identified potential co-therapies for a recalcitrant Gram-negative bacterial species. Our assay could be used more widely to aid in chemotherapy to treat infections caused by these bacteria.


Assuntos
Antibacterianos/farmacologia , Infecções por Burkholderia/tratamento farmacológico , Burkholderia/efeitos dos fármacos , Ceftazidima/farmacologia , Cloroquinolinóis/farmacologia , Burkholderia pseudomallei/efeitos dos fármacos , Descoberta de Drogas , Sinergismo Farmacológico , Humanos , Melioidose/tratamento farmacológico , Testes de Sensibilidade Microbiana
3.
Biochem Biophys Rep ; 23: 100773, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32548313

RESUMO

Increasing resistance of bacteria to antibiotics is a serious global challenge and there is a need to unlock the potential of novel antibacterial targets. One such target is the essential prokaryotic endoribonuclease RNase E. Using a combination of in silico high-throughput screening and in vitro validation we have identified three novel small molecule inhibitors of RNase E that are active against RNase E from Escherichia coli, Francisella tularensis and Acinetobacter baumannii. Two of the inhibitors are non-natural small molecules that could be suitable as lead compounds for the development of broad-spectrum antibiotics targeting RNase E. The third small molecule inhibitor is glucosamine-6-phosphate, a precursor of bacterial cell envelope peptidoglycans and lipopolysaccharides, hinting at a novel metabolite-mediated mechanism of regulation of RNase E.

4.
Microb Genom ; 5(11)2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31580793

RESUMO

Yersinia pseudotuberculosis is a Gram-negative bacterium capable of causing gastrointestinal infection and is closely related to the highly virulent plague bacillus Yersinia pestis. Infections by both species are currently treatable with antibiotics such as ciprofloxacin, a quinolone-class drug of major clinical importance in the treatment of many other infections. Our current understanding of the mechanism of action of ciprofloxacin is that it inhibits DNA replication by targeting DNA gyrase, and that resistance is primarily due to mutation of this target site, along with generic efflux and detoxification strategies. We utilized transposon-directed insertion site sequencing (TraDIS or TnSeq) to identify the non-essential chromosomal genes in Y. pseudotuberculosis that are required to tolerate sub-lethal concentrations of ciprofloxacin in vitro. As well as highlighting recognized antibiotic resistance genes, we provide evidence that multiple genes involved in regulating DNA replication and repair are central in enabling Y. pseudotuberculosis to tolerate the antibiotic, including DksA (yptb0734), a regulator of RNA polymerase, and Hda (yptb2792), an inhibitor of DNA replication initiation. We furthermore demonstrate that even at sub-lethal concentrations, ciprofloxacin causes severe cell-wall stress, requiring lipopolysaccharide lipid A, O-antigen and core biosynthesis genes to resist the sub-lethal effects of the antibiotic. It is evident that coping with the consequence(s) of antibiotic-induced stress requires the contribution of scores of genes that are not exclusively engaged in drug resistance.


Assuntos
Ciprofloxacina/farmacologia , Resistência Microbiana a Medicamentos/genética , Yersinia pseudotuberculosis/genética , Anti-Infecciosos/farmacologia , Sequência de Bases/genética , Cromossomos/genética , Ciprofloxacina/metabolismo , Reparo do DNA/genética , Replicação do DNA/genética , Evolução Molecular , Genoma Bacteriano , Mutação , Virulência/genética , Fatores de Virulência/genética , Yersinia pestis/genética , Yersinia pestis/metabolismo , Yersinia pseudotuberculosis/metabolismo , Infecções por Yersinia pseudotuberculosis/microbiologia
5.
Front Microbiol ; 10: 904, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31118924

RESUMO

The efficacy of the novel fluoroquinolone finafloxacin was evaluated as a potential therapeutic in vitro and in vivo, following an intranasal infection of Francisella tularensis strain SchuS4 in BALB/c mice. We demonstrated that short treatment courses of finafloxacin provide high levels of protection, with a single dose resulting in a significant increase in time to death when compared to ciprofloxacin. In addition, following investigation into the window of opportunity for treatment, we have shown that finafloxacin can provided protection when administered up to 96 h post-challenge. This is particularly encouraging since mice displayed severe signs of disease at this time point. In summary, finafloxacin may be a promising therapy for use in the event of exposure to F. tularensis, perhaps enabling the treatment regimen to be shortened or if therapy is delayed. The efficacy of finafloxacin against other biological threat agents also warrants investigation.

6.
Sci Rep ; 9(1): 7952, 2019 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-31138855

RESUMO

Regulation of gene expression through processing and turnover of RNA is a key mechanism that allows bacteria to rapidly adapt to changing environmental conditions. Consequently, RNA degrading enzymes (ribonucleases; RNases) such as the endoribonuclease RNase E, frequently play critical roles in pathogenic bacterial virulence and are potential antibacterial targets. RNase E consists of a highly conserved catalytic domain and a variable non-catalytic domain that functions as the structural scaffold for the multienzyme degradosome complex. Despite conservation of the catalytic domain, a recent study identified differences in the response of RNase E homologues from different species to the same inhibitory compound(s). While RNase E from Escherichia coli has been well-characterised, far less is known about RNase E homologues from other bacterial species. In this study, we structurally and biochemically characterise the RNase E catalytic domains from four pathogenic bacteria: Yersinia pestis, Francisella tularensis, Burkholderia pseudomallei and Acinetobacter baumannii, with a view to exploiting RNase E as an antibacterial target. Bioinformatics, small-angle x-ray scattering and biochemical RNA cleavage assays reveal globally similar structural and catalytic properties. Surprisingly, subtle species-specific differences in both structure and substrate specificity were also identified that may be important for the development of effective antibacterial drugs targeting RNase E.


Assuntos
Acinetobacter baumannii/enzimologia , Proteínas de Bactérias/química , Burkholderia pseudomallei/enzimologia , Endorribonucleases/química , Francisella tularensis/enzimologia , Yersinia pestis/enzimologia , Acinetobacter baumannii/genética , Acinetobacter baumannii/patogenicidade , Sequência de Aminoácidos , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Burkholderia pseudomallei/genética , Burkholderia pseudomallei/patogenicidade , Domínio Catalítico , Clonagem Molecular , Endorribonucleases/genética , Endorribonucleases/metabolismo , Escherichia coli/enzimologia , Escherichia coli/genética , Escherichia coli/patogenicidade , Francisella tularensis/genética , Francisella tularensis/patogenicidade , Expressão Gênica , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Cinética , Modelos Moleculares , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , RNA/química , RNA/genética , RNA/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Alinhamento de Sequência , Homologia Estrutural de Proteína , Especificidade por Substrato , Virulência , Yersinia pestis/genética , Yersinia pestis/patogenicidade
7.
Int J Antimicrob Agents ; 54(1): 85-88, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31029736

RESUMO

Disulfiram (DSF) can help treat alcohol dependency by inhibiting aldehyde dehydrogenase (ALDH). Genomic analysis revealed that Francisella tularensis, the causative agent of tularemia, has lost all but one ALDH-like domain and that this domain retains the target of DSF. In this study, minimum inhibitory concentration (MIC) assays demonstrated that both DSF and its primary metabolite diethyldithiocarbamate (DDC) have strong antimicrobial activity against F. tularensis strain SCHU S4, with the MIC of DSF determined as 2 µg/mL in comparison with 8 µg/mL for DDC. The activity of DSF was further confirmed using an in vitro human macrophage infection assay. Francisella tularensis bacteria in DSF-treated cells were reduced in comparison with untreated and DDC-treated cells, comparable with that observed in doxycycline-treated cells. This suggests that DSF may be suitable for further investigation as an in vivo therapy for tularemia.


Assuntos
Inibidores de Acetaldeído Desidrogenases/farmacologia , Dissuasores de Álcool/farmacologia , Antibacterianos/farmacologia , Dissulfiram/farmacologia , Francisella tularensis/efeitos dos fármacos , Francisella tularensis/crescimento & desenvolvimento , Carga Bacteriana , Humanos , Testes de Sensibilidade Microbiana , Monócitos/efeitos dos fármacos , Monócitos/microbiologia , Células THP-1
8.
J Bacteriol ; 201(7)2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30642993

RESUMO

The highly virulent intracellular pathogen Francisella tularensis is a Gram-negative bacterium that has a wide host range, including humans, and is the causative agent of tularemia. To identify new therapeutic drug targets and vaccine candidates and investigate the genetic basis of Francisella virulence in the Fischer 344 rat, we have constructed an F. tularensis Schu S4 transposon library. This library consists of more than 300,000 unique transposon mutants and represents a transposon insertion for every 6 bp of the genome. A transposon-directed insertion site sequencing (TraDIS) approach was used to identify 453 genes essential for growth in vitro Many of these essential genes were mapped to key metabolic pathways, including glycolysis/gluconeogenesis, peptidoglycan synthesis, fatty acid biosynthesis, and the tricarboxylic acid (TCA) cycle. Additionally, 163 genes were identified as required for fitness during colonization of the Fischer 344 rat spleen. This in vivo selection screen was validated through the generation of marked deletion mutants that were individually assessed within a competitive index study against the wild-type F. tularensis Schu S4 strain.IMPORTANCE The intracellular bacterial pathogen Francisella tularensis causes a disease in humans characterized by the rapid onset of nonspecific symptoms such as swollen lymph glands, fever, and headaches. F. tularensis is one of the most infectious bacteria known and following pulmonary exposure can have a mortality rate exceeding 50% if left untreated. The low infectious dose of this organism and concerns surrounding its potential as a biological weapon have heightened the need for effective and safe therapies. To expand the repertoire of targets for therapeutic development, we initiated a genome-wide analysis. This study has identified genes that are important for F. tularensis under in vitro and in vivo conditions, providing candidates that can be evaluated for vaccine or antibacterial development.


Assuntos
Francisella tularensis/crescimento & desenvolvimento , Francisella tularensis/genética , Genes Bacterianos , Tularemia/microbiologia , Fatores de Virulência/genética , Animais , Análise Mutacional de DNA , Elementos de DNA Transponíveis , Modelos Animais de Doenças , Testes Genéticos , Mutagênese Insercional , Neocallimastigales , Ratos Endogâmicos F344
9.
BMC Microbiol ; 18(1): 46, 2018 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-29855259

RESUMO

BACKGROUND: Yersinia pseudotuberculosis is a zoonotic pathogen, causing mild gastrointestinal infection in humans. From this comparatively benign pathogenic species emerged the highly virulent plague bacillus, Yersinia pestis, which has experienced significant genetic divergence in a relatively short time span. Much of our knowledge of Yersinia spp. evolution stems from genomic comparison and gene expression studies. Here we apply transposon-directed insertion site sequencing (TraDIS) to describe the essential gene set of Y. pseudotuberculosis IP32953 in optimised in vitro growth conditions, and contrast these with the published essential genes of Y. pestis. RESULTS: The essential genes of an organism are the core genetic elements required for basic survival processes in a given growth condition, and are therefore attractive targets for antimicrobials. One such gene we identified is yptb3665, which encodes a peptide deformylase, and here we report for the first time, the sensitivity of Y. pseudotuberculosis to actinonin, a deformylase inhibitor. Comparison of the essential genes of Y. pseudotuberculosis with those of Y. pestis revealed the genes whose importance are shared by both species, as well as genes that were differentially required for growth. In particular, we find that the two species uniquely rely upon different iron acquisition and respiratory metabolic pathways under similar in vitro conditions. CONCLUSIONS: The discovery of uniquely essential genes between the closely related Yersinia spp. represent some of the fundamental, species-defining points of divergence that arose during the evolution of Y. pestis from its ancestor. Furthermore, the shared essential genes represent ideal candidates for the development of novel antimicrobials against both species.


Assuntos
Genes Essenciais , Mutagênese Insercional/métodos , Yersinia pestis/crescimento & desenvolvimento , Yersinia pseudotuberculosis/crescimento & desenvolvimento , Proteínas de Bactérias/genética , Elementos de DNA Transponíveis , Evolução Molecular , Especiação Genética , Humanos , Análise de Sequência de DNA , Yersinia pestis/genética , Yersinia pseudotuberculosis/genética
10.
BMC Microbiol ; 17(1): 163, 2017 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-28732479

RESUMO

BACKGROUND: The World Health Organization has categorized plague as a re-emerging disease and the potential for Yersinia pestis to also be used as a bioweapon makes the identification of new drug targets against this pathogen a priority. Environmental temperature is a key signal which regulates virulence of the bacterium. The bacterium normally grows outside the human host at 28 °C. Therefore, understanding the mechanisms that the bacterium used to adapt to a mammalian host at 37 °C is central to the development of vaccines or drugs for the prevention or treatment of human disease. RESULTS: Using a library of over 1 million Y. pestis CO92 random mutants and transposon-directed insertion site sequencing, we identified 530 essential genes when the bacteria were cultured at 28 °C. When the library of mutants was subsequently cultured at 37 °C we identified 19 genes that were essential at 37 °C but not at 28 °C, including genes which encode proteins that play a role in enabling functioning of the type III secretion and in DNA replication and maintenance. Using genome-scale metabolic network reconstruction we showed that growth conditions profoundly influence the physiology of the bacterium, and by combining computational and experimental approaches we were able to identify 54 genes that are essential under a broad range of conditions. CONCLUSIONS: Using an integrated computational-experimental approach we identify genes which are required for growth at 37 °C and under a broad range of environments may be the best targets for the development of new interventions to prevent or treat plague in humans.


Assuntos
Proteínas de Bactérias/genética , Biologia Computacional/métodos , Genes Essenciais , Peste/microbiologia , Yersinia pestis/genética , Proteínas de Bactérias/metabolismo , Regulação Bacteriana da Expressão Gênica , Humanos , Mutação , Yersinia pestis/crescimento & desenvolvimento , Yersinia pestis/metabolismo
11.
Artigo em Inglês | MEDLINE | ID: mdl-28438936

RESUMO

Burkholderia pseudomallei is the causative agent of melioidosis, a serious disease endemic in Southeast Asia and Northern Australia. Antibiotic treatment is lengthy and relapse often occurs. Finafloxacin is a novel fluoroquinolone with increased antibacterial activity in acidic conditions in contrast to other fluoroquinolones which demonstrate reduced activity at a lower pH. Therefore, finafloxacin may have improved efficacy against B. pseudomallei, which can survive within host cells where the local pH is acidic. In vitro analysis was performed using MICs, minimal bactericidal concentrations (MBCs), time-kill assays, persister cell assays, and macrophage assays. Finafloxacin showed increased bactericidal activity at pH 5 in comparison to pH 7 and ciprofloxacin at pH 5. In vivo studies in BALB/c mice included pharmacokinetic studies to inform an appropriate dosing regimen. Finafloxacin efficacy was evaluated in an inhalational murine model of melioidosis where antibiotic treatment was initiated at 6 or 24 h postchallenge and continued for 14 days, and mice were observed for 63 days. The survival of infected mice following 14 days of treatment was 80%, 60% or 0% for treatments initiated at 6 h and 60%, 30% or 0% for treatments initiated at 24 h for finafloxacin, co-trimoxazole, or ciprofloxacin, respectively. In summary, finafloxacin has increased bactericidal activity for B. pseudomallei under acidic conditions in vitro and improves survival in a murine model of melioidosis compared with those for ciprofloxacin. Furthermore, finafloxacin improves bacteriological clearance compared with that of co-trimoxazole, suggesting it may offer an effective postexposure prophylaxis against B. pseudomallei.


Assuntos
Antibacterianos/farmacologia , Burkholderia pseudomallei/efeitos dos fármacos , Fluoroquinolonas/farmacologia , Animais , Ciprofloxacina/farmacologia , Concentração de Íons de Hidrogênio , Camundongos , Testes de Sensibilidade Microbiana
12.
Front Microbiol ; 8: 91, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28220110

RESUMO

Inhalation of Yersinia pestis can lead to pneumonic plague, which without treatment is inevitably fatal. Two novel formulations of liposome-encapsulated ciprofloxacin, 'ciprofloxacin for inhalation' (CFI, Lipoquin®) and 'dual release ciprofloxacin for inhalation' (DRCFI, Pulmaquin®) containing CFI and ciprofloxacin solution, are in development. These were evaluated as potential therapies for infection with Y. pestis. In a murine model of pneumonic plague, human-like doses of aerosolized CFI, aerosolized DRCFI or intraperitoneal (i.p.) ciprofloxacin were administered at 24 h (representing prophylaxis) or 42 h (representing treatment) post-challenge. All three therapies provided a high level of protection when administered 24 h post-challenge. A single dose of CFI, but not DRCFI, significantly improved survival compared to a single dose of ciprofloxacin. Furthermore, single doses of CFI and DRCFI reduced bacterial burden in lungs and spleens to below the detectable limit at 60 h post-challenge. When therapy was delayed until 42 h post-challenge, a single dose of CFI or DRCFI offered minimal protection. However, single doses of CFI or DRCFI were able to significantly reduce the bacterial burden in the spleen compared to empty liposomes. A three-day treatment regimen of ciprofloxacin, CFI, or DRCFI resulted in high levels of protection (90-100% survival). This study suggests that CFI and DRCFI may be useful therapies for Y. pestis infection, both as prophylaxis and for the treatment of plague.

13.
Antimicrob Agents Chemother ; 60(12): 7206-7215, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27671061

RESUMO

Bioluminescence imaging (BLI) enables real-time, noninvasive tracking of infection in vivo and longitudinal infection studies. In this study, a bioluminescent Francisella tularensis strain, SCHU S4-lux, was used to develop an inhalational infection model in BALB/c mice. Mice were infected intranasally, and the progression of infection was monitored in real time using BLI. A bioluminescent signal was detectable from 3 days postinfection (3 dpi), initially in the spleen and then in the liver and lymph nodes, before finally becoming systemic. The level of bioluminescent signal correlated with bacterial numbers in vivo, enabling noninvasive quantification of bacterial burdens in tissues. Treatment with levofloxacin (commencing at 4 dpi) significantly reduced the BLI signal. Furthermore, BLI was able to distinguish noninvasively between different levofloxacin treatment regimens and to identify sites of relapse following treatment cessation. These data demonstrate that BLI and SCHU S4-lux are suitable for the study of F. tularensis pathogenesis and the evaluation of therapeutics for tularemia.


Assuntos
Antibacterianos/farmacologia , Francisella tularensis/efeitos dos fármacos , Francisella tularensis/crescimento & desenvolvimento , Tularemia/tratamento farmacológico , Tularemia/patologia , Animais , Modelos Animais de Doenças , Feminino , Francisella tularensis/metabolismo , Levofloxacino/farmacologia , Fígado/microbiologia , Medições Luminescentes , Linfonodos/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Baço/microbiologia , Tularemia/microbiologia
14.
PLoS One ; 11(7): e0158575, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27391310

RESUMO

Toll-like receptors (TLRs) recognise invading pathogens and mediate downstream immune signalling via Toll/IL-1 receptor (TIR) domains. TIR domain proteins (Tdps) have been identified in multiple pathogenic bacteria and have recently been implicated as negative regulators of host innate immune activation. A Tdp has been identified in Bacillus anthracis, the causative agent of anthrax. Here we present the first study of this protein, designated BaTdp. Recombinantly expressed and purified BaTdp TIR domain interacted with several human TIR domains, including that of the key TLR adaptor MyD88, although BaTdp expression in cultured HEK293 cells had no effect on TLR4- or TLR2- mediated immune activation. During expression in mammalian cells, BaTdp localised to microtubular networks and caused an increase in lipidated cytosolic microtubule-associated protein 1A/1B-light chain 3 (LC3), indicative of autophagosome formation. In vivo intra-nasal infection experiments in mice showed that a BaTdp knockout strain colonised host tissue faster with higher bacterial load within 4 days post-infection compared to the wild type B. anthracis. Taken together, these findings indicate that BaTdp does not play an immune suppressive role, but rather, its absence increases virulence. BaTdp present in wild type B. anthracis plausibly interact with the infected host cell, which undergoes autophagy in self-defence.


Assuntos
Autofagia/fisiologia , Bacillus anthracis/metabolismo , Proteínas de Bactérias/metabolismo , Microtúbulos/metabolismo , Animais , Antraz/microbiologia , Autofagia/genética , Bacillus anthracis/genética , Proteínas de Bactérias/genética , Linhagem Celular , Sobrevivência Celular/fisiologia , Feminino , Células HEK293 , Humanos , Camundongos , Microscopia Confocal , Filogenia , Ligação Proteica , Transdução de Sinais , Virulência/genética , Virulência/fisiologia
15.
Curr Opin Microbiol ; 29: 94-103, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26803404

RESUMO

The Burkholderia genus contains a group of soil-dwelling Gram-negative organisms that are prevalent in warm and humid climates. Two species in particular are able to cause disease in animals, B. mallei primarily infects Equus spp. and B. pseudomallei (BPS), that is able to cause potentially life-threatening disease in humans. BPS is naturally resistant to many antibiotics and there is no vaccine available. Although not a specialised human pathogen, BPS possesses a large genome and many virulence traits that allow it to adapt and survive very successfully in the human host. Key to this survival is the ability of BPS to replicate intracellularly. In this review we highlight recent advances in our understanding of the intracellular survival of BPS, including how it overcomes host immune defenses and other challenges to establish its niche and then spread the infection. Knowledge of these mechanisms increases our capacity for therapeutic interventions against a well-armed foe.


Assuntos
Burkholderia pseudomallei/crescimento & desenvolvimento , Burkholderia pseudomallei/imunologia , Citoplasma/microbiologia , Interações Hospedeiro-Patógeno , Melioidose/microbiologia , Actinas/metabolismo , Animais , Burkholderia pseudomallei/genética , Burkholderia pseudomallei/patogenicidade , Replicação do DNA , Células Gigantes/microbiologia , Humanos , Melioidose/terapia , Sistemas de Secreção Tipo VI/metabolismo , Virulência/genética , Fatores de Virulência/genética , Fatores de Virulência/fisiologia
16.
Res Microbiol ; 167(3): 159-67, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26654915

RESUMO

Kynurenine formamidase (KynB) forms part of the kynurenine pathway which metabolises tryptophan to anthranilate. This metabolite can be used for downstream production of 2-alkyl-4-quinolone (AQ) signalling molecules that control virulence in Pseudomonas aeruginosa. Here we investigate the role of kynB in the production of AQs and virulence-associated phenotypes of Burkholderia pseudomallei K96243, the causative agent of melioidosis. Deletion of kynB resulted in reduced AQ production, increased biofilm formation, decreased swarming and increased tolerance to ciprofloxacin. Addition of exogenous anthranilic acid restored the biofilm phenotype, but not the persister phenotype. This study suggests the kynurenine pathway is a critical source of anthranilate and signalling molecules that may regulate B. pseudomallei virulence.


Assuntos
Arilformamidase/metabolismo , Biofilmes/crescimento & desenvolvimento , Burkholderia pseudomallei/enzimologia , Burkholderia pseudomallei/fisiologia , Locomoção , Quinolonas/metabolismo , Transdução de Sinais , Burkholderia pseudomallei/genética , Deleção de Genes , Triptofano/metabolismo , Virulência , ortoaminobenzoatos/metabolismo
17.
Infect Immun ; 84(3): 701-10, 2015 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-26712202

RESUMO

Burkholderia pseudomallei, the causative agent of melioidosis, has complex and poorly understood extracellular and intracellular lifestyles. We used transposon-directed insertion site sequencing (TraDIS) to retrospectively analyze a transposon library that had previously been screened through a BALB/c mouse model to identify genes important for growth and survival in vivo. This allowed us to identify the insertion sites and phenotypes of negatively selected mutants that were previously overlooked due to technical constraints. All 23 unique genes identified in the original screen were confirmed by TraDIS, and an additional 105 mutants with various degrees of attenuation in vivo were identified. Five of the newly identified genes were chosen for further characterization, and clean, unmarked bpsl2248, tex, rpiR, bpsl1728, and bpss1528 deletion mutants were constructed from the wild-type strain K96243. Each of these mutants was tested in vitro and in vivo to confirm their attenuated phenotypes and investigate the nature of the attenuation. Our results confirm that we have identified new genes important to in vivo virulence with roles in different stages of B. pseudomallei pathogenesis, including extracellular and intracellular survival. Of particular interest, deletion of the transcription accessory protein Tex was shown to be highly attenuating, and the tex mutant was capable of providing protective immunity against challenge with wild-type B. pseudomallei, suggesting that the genes identified in our TraDIS screen have the potential to be investigated as live vaccine candidates.


Assuntos
Proteínas de Bactérias/metabolismo , Burkholderia pseudomallei/crescimento & desenvolvimento , Burkholderia pseudomallei/metabolismo , Melioidose/microbiologia , Fatores de Virulência/metabolismo , Animais , Proteínas de Bactérias/genética , Burkholderia pseudomallei/genética , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Viabilidade Microbiana , Fatores de Virulência/genética
18.
Artigo em Inglês | MEDLINE | ID: mdl-24995163

RESUMO

Liposome-encapsulation has been suggested as method to improve the efficacy of ciprofloxacin against the intracellular pathogen, Francisella tularensis. Early work with a prototype formulation, evaluated for use against the F. tularensis live vaccine strain, showed that a single dose of liposomal ciprofloxacin given by the intranasal or inhalational route could provide protection in a mouse model of pneumonic tularemia. Liposomal ciprofloxacin offered better protection than ciprofloxacin given by the same routes. Liposomal ciprofloxacin has been further developed by Aradigm Corporation for Pseudomonas aeruginosa infections in patients with cystic fibrosis and non-cystic fibrosis bronchiectasis. This advanced development formulation is safe, effective and well tolerated in human clinical trials. Further evaluation of the advanced liposomal ciprofloxacin formulation against the highly virulent F. tularensis Schu S4 strain has shown that aerosolized CFI (Ciprofloxacin encapsulated in liposomes for inhalation) provides significantly better protection than oral ciprofloxacin. Thus, liposomal ciprofloxacin is a promising treatment for tularemia and further research with the aim of enabling licensure under the animal rule is warranted.


Assuntos
Antibacterianos/administração & dosagem , Ciprofloxacina/administração & dosagem , Portadores de Fármacos/administração & dosagem , Lipossomos/administração & dosagem , Tularemia/tratamento farmacológico , Administração por Inalação , Administração Intranasal , Animais , Modelos Animais de Doenças , Resultado do Tratamento
19.
Antimicrob Agents Chemother ; 58(6): 3053-9, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24637682

RESUMO

Liposome-encapsulated ciprofloxacin for inhalation (CFI) was investigated as a putative postexposure therapeutic for two strains of Francisella tularensis. The efficacies of oral ciprofloxacin and intranasally instilled CFI could not be distinguished in a mouse model of infection with the F. tularensis live vaccine strain (LVS), where a single dose of either formulation offered full protection against a lethal challenge. However, mouse studies with the more virulent Schu S4 strain of F. tularensis demonstrated that a higher level of protection against a lethal aerosol infection is provided by CFI than by oral ciprofloxacin. In addition, using this infection model, it was possible to discriminate the efficacy of intranasally instilled CFI from that of aerosolized CFI, with aerosolized CFI providing full protection after just a single dose. The improved efficacy of CFI compared to oral ciprofloxacin is likely due to the high sustained concentrations of ciprofloxacin in the lung. In summary, CFI may be a promising therapy, perhaps enabling the prophylactic regimen to be shortened, for use in the event of a deliberate release of F. tularensis. The prophylactic efficacy of CFI against other biological warfare (BW) threat agents also warrants investigation.


Assuntos
Ciprofloxacina/administração & dosagem , Francisella tularensis/efeitos dos fármacos , Lipossomos , Tularemia/tratamento farmacológico , Vacinas Atenuadas/imunologia , Administração por Inalação , Administração Intranasal , Aerossóis , Animais , Vacinas Bacterianas/imunologia , Disponibilidade Biológica , Ciprofloxacina/farmacocinética , Modelos Animais de Doenças , Feminino , Francisella tularensis/imunologia , Francisella tularensis/patogenicidade , Pulmão/imunologia , Pulmão/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Análise de Sobrevida , Virulência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA