Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Nat Commun ; 13(1): 5103, 2022 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-36042245

RESUMEN

Intestinal barrier derangement allows intestinal bacteria and their products to translocate to the systemic circulation. Pseudomonas aeruginosa (PA) superimposed infection in critically ill patients increases gut permeability and leads to gut-driven sepsis. PA infections are challenging due to multi-drug resistance (MDR), biofilms, and/or antibiotic tolerance. Inhibition of the quorum-sensing transcriptional regulator MvfR(PqsR) is a desirable anti-PA anti-virulence strategy as MvfR controls multiple acute and chronic virulence functions. Here we show that MvfR promotes intestinal permeability and report potent anti-MvfR compounds, the N-Aryl Malonamides (NAMs), resulting from extensive structure-activity-relationship studies and thorough assessment of the inhibition of MvfR-controlled virulence functions. This class of anti-virulence non-native ligand-based agents has a half-maximal inhibitory concentration in the nanomolar range and strong target engagement. Using a NAM lead in monotherapy protects murine intestinal barrier function, abolishes MvfR-regulated small molecules, ameliorates bacterial dissemination, and lowers inflammatory cytokines. This study demonstrates the importance of MvfR in PA-driven intestinal permeability. It underscores the utility of anti-MvfR agents in maintaining gut mucosal integrity, which should be part of any successful strategy to prevent/treat PA infections and associated gut-derived sepsis in critical illness settings. NAMs provide for the development of crucial preventive/therapeutic monotherapy options against untreatable MDR PA infections.


Asunto(s)
Infecciones por Pseudomonas , Sepsis , Animales , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Proteínas Bacterianas/farmacología , Biopelículas , Enfermedad Crítica , Humanos , Ratones , Infecciones por Pseudomonas/tratamiento farmacológico , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/fisiología , Percepción de Quorum , Sepsis/tratamiento farmacológico , Virulencia
2.
Sci Rep ; 11(1): 14162, 2021 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-34238943

RESUMEN

The prevalence of atopic diseases has been steadily increasing since the mid twentieth century, a rise that has been linked to modern hygienic lifestyles that limit exposure to microbes and immune system maturation. Overactive type 2 CD4+ helper T (Th2) cells are known to be closely associated with atopy and represent a key target for treatment. In this study, we present an initial characterization of ammonia oxidizing bacteria (AOB) Nitrosomonas eutropha D23, an environmental microbe that is not associated with human pathology, and show AOB effectively suppress the polarization of Th2 cells and production of Th2-associated cytokines (IL-5, IL-13, and IL-4) by human peripheral blood mononuclear cells (PBMC). We show that AOB inhibit Th2 cell polarization not through Th1-mediated suppression, but rather through mechanisms involving the anti-inflammatory cytokine IL-10 and the potential inhibition of dendritic cells, as evidenced by a reduction in Major Histocompatibility Complex Class II (MHC II) and CD86 expression following AOB treatment. This is the first report of immunomodulatory properties of AOB, and provides initial support for the development of AOB as a potential therapeutic for atopic diseases.


Asunto(s)
Amoníaco/metabolismo , Antiinflamatorios/metabolismo , Polaridad Celular , Interleucina-10/metabolismo , Nitrosomonas/metabolismo , Células Th2/citología , Células Th2/microbiología , Células Dendríticas/metabolismo , Humanos , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/microbiología , Metaboloma , Oxidación-Reducción , Transducción de Señal , Células TH1/inmunología
4.
Methods Mol Biol ; 1673: 227-241, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29130177

RESUMEN

Quorum sensing (QS) systems play global regulatory roles in bacterial virulence. They synchronize the expression of multiple virulence factors and they control and modulate bacterial antibiotic tolerance systems and host defense mechanisms. Therefore, it is important to obtain knowledge about QS modes of action and to test putative therapeutics that may interrupt QS actions in the context of infections. This chapter describes methods to study bacterial pathogenesis in murine acute and persistent/relapsing infection models, using the Gram-negative bacterial pathogen Pseudomonas aeruginosa as an example. These infection models can be used to probe bacterial virulence functions and in mechanistic studies, as well as for the assessment of the therapeutic potential of antibacterials, including anti-virulence agents.


Asunto(s)
Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/metabolismo , Percepción de Quorum , Animales , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Quemaduras/tratamiento farmacológico , Quemaduras/microbiología , Quemaduras/patología , Modelos Animales de Enfermedad , Enfermedades Pulmonares/tratamiento farmacológico , Enfermedades Pulmonares/microbiología , Enfermedades Pulmonares/patología , Masculino , Ratones , Infecciones por Pseudomonas/tratamiento farmacológico , Infecciones por Pseudomonas/patología , Pseudomonas aeruginosa/efectos de los fármacos , Percepción de Quorum/efectos de los fármacos
5.
Artículo en Inglés | MEDLINE | ID: mdl-28923875

RESUMEN

Pseudomonas aeruginosa biofilms contribute to its survival on biotic and abiotic surfaces and represent a major clinical threat due to their high tolerance to antibiotics. Therefore, the discovery of antibiofilm agents may hold great promise. We show that pharmacological inhibition of the P. aeruginosa quorum-sensing regulator MvfR (PqsR) using a benzamide-benzimidazole compound interferes with biofilm formation and potentiates biofilm sensitivity to antibiotics. Such a strategy could have great potential against P. aeruginosa persistence in diverse environments.


Asunto(s)
Antibacterianos/farmacología , Proteínas Bacterianas/antagonistas & inhibidores , Biopelículas/efectos de los fármacos , Regulación Bacteriana de la Expresión Génica , Pseudomonas aeruginosa/efectos de los fármacos , Percepción de Quorum/efectos de los fármacos , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Benzamidas/síntesis química , Benzamidas/farmacología , Bencimidazoles/síntesis química , Bencimidazoles/farmacología , Biopelículas/crecimiento & desarrollo , Recuento de Colonia Microbiana , Medios de Cultivo/química , Sinergismo Farmacológico , Tolerancia a Medicamentos/genética , Meropenem , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/crecimiento & desarrollo , Tienamicinas/farmacología , Tobramicina/farmacología , Transcripción Genética
6.
Front Microbiol ; 8: 924, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28596760

RESUMEN

Pseudomonas aeruginosa is a Gram-negative bacterium, which causes opportunistic infections in immuno-compromised individuals. Due to its multiple resistances toward antibiotics, the development of new drugs is required. Interfering with Quorum Sensing (QS), a cell-to-cell communication system, has shown to be highly efficient in reducing P. aeruginosa pathogenicity. One of its QS systems employs Pseudomonas Quinolone Signal (PQS) and 4-hydroxy-2-heptylquinoline (HHQ) as signal molecules. Both activate the transcriptional regulator MvfR (Multiple Virulence Factor Regulator), also called PqsR, driving the production of QS molecules as well as toxins and biofilm formation. The aim of this work was to elucidate the effects of QS inhibitors (QSIs), such as MvfR antagonists and PqsBC inhibitors, on the biosynthesis of the MvfR-regulated small molecules 2'-aminoacetophenone (2-AA), dihydroxyquinoline (DHQ), HHQ, PQS, and 4-hydroxy-2-heptylquinoline-N-oxide (HQNO). The employed synthetic MvfR antagonist fully inhibited pqs small molecule formation showing expected sigmoidal dose-response curves for 2-AA, HQNO, HHQ and PQS. Surprisingly, DHQ levels were enhanced at lower antagonist concentrations followed by a full suppression at higher QSI amounts. This particular bi-phasic profile hinted at the accumulation of a biosynthetic intermediate resulting in the observed overproduction of the shunt product DHQ. Additionally, investigations on PqsBC inhibitors showed a reduction of MvfR natural ligands, while increased 2-AA, DHQ and HQNO levels compared to the untreated cells were detected. Moreover, PqsBC inhibitors did not show any significant effect in PA14 pqsC mutant demonstrating their target selectivity. As 2-AA is important for antibacterial tolerance, the QSIs were evaluated in their capability to attenuate persistence. Indeed, persister cells were reduced along with 2-AA inhibition resulting from MvfR antagonism, but not from PqsBC inhibition. In conclusion, antagonizing MvfR using a dosage capable of fully suppressing this QS system will lead to a favorable therapeutic outcome as DHQ overproduction is avoided and bacterial persistence is reduced.

7.
ACS Chem Biol ; 12(5): 1435-1443, 2017 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-28379691

RESUMEN

Pseudomonas aeruginosa is an important nosocomial pathogen that is frequently recalcitrant to available antibiotics, underlining the urgent need for alternative therapeutic options against this pathogen. Targeting virulence functions is a promising alternative strategy as it is expected to generate less-selective resistance to treatment compared to antibiotics. Capitalizing on our nonligand-based benzamide-benzimidazole (BB) core structure compounds reported to efficiently block the activity of the P. aeruginosa multiple virulence factor regulator MvfR, here we report the first class of inhibitors shown to interfere with PqsBC enzyme activity, responsible for the synthesis of the MvfR activating ligands HHQ and PQS, and the first to target simultaneously MvfR and PqsBC activity. The use of these compounds reveals that inhibiting PqsBC is sufficient to block P. aeruginosa's acute virulence functions, as the synthesis of MvfR ligands is inhibited. Our results show that MvfR remains the best target of this QS pathway, as we show that antagonists of this target block both acute and persistence-related functions. The structural properties of the compounds reported in this study provide several insights that are instrumental for the design of improved MvfR regulon inhibitors against both acute and persistent P. aeruginosa infections. Moreover, the data presented offer the possibility of a polypharmacology approach of simultaneous silencing two targets in the same pathway. Such a combined antivirulence strategy holds promise in increasing therapeutic efficacy and providing alternatives in the event of a single target's resistance development.


Asunto(s)
Polifarmacología , Pseudomonas aeruginosa/genética , Regulón/efectos de los fármacos , Tolerancia a Medicamentos , Inhibidores Enzimáticos/farmacología , Terapia Molecular Dirigida/métodos , Infecciones por Pseudomonas/tratamiento farmacológico , Pseudomonas aeruginosa/enzimología , Virulencia/efectos de los fármacos , Factores de Virulencia
8.
J Crohns Colitis ; 11(7): 840-847, 2017 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-28130329

RESUMEN

BACKGROUND AND AIMS: Adherent invasive Escherichia coli [AIEC] are abnormally predominant on the ileal mucosa of Crohn's disease [CD] patients. They bind to the CEACAM6 receptor expressed on the surface of epithelial cells. We aimed to assess the potential of bacteriophages, viruses infecting bacteria, to decrease the levels of AIEC bacteria associated with the intestinal mucosa. METHODS: We combined ex vivo and in vivo experiments with murine and human intestinal samples to quantify the ability of virulent bacteriophages to target the prototype AIEC strain LF82. RESULTS: We found that three virulent bacteriophages were able to replicate in ileal, caecal and colonic sections and faeces homogenates from murine gut samples colonised with the prototype AIEC strain LF82. A single day of per os treatment with the three bacteriophages cocktail given to LF82-colonised CEABAC10 transgenic mice, expressing the human CEACAM6 receptor for AIEC, decreased significantly the number of AIEC in faeces and in the adherent flora of intestinal sections. In addition, a single dose of the cocktail reduced dextran sodium sulphate-induced colitis symptoms on conventional mice colonised with the strain LF82 over a 2-week period. The cocktail targeted also LF82 bacteria in homogenates of ileal biopsies taken from CD patients. CONCLUSIONS: These findings demonstrate that bacteriophages are a new treatment option for targeting AIEC in CD patients and represent a strong basis for a clinical trial evaluation.


Asunto(s)
Bacteriófagos , Colitis/terapia , Enfermedad de Crohn/microbiología , Escherichia coli/virología , Mucosa Intestinal/microbiología , Terapia de Fagos , Animales , Antígenos CD/metabolismo , Bacteriófagos/crecimiento & desarrollo , Ciego/microbiología , Moléculas de Adhesión Celular/metabolismo , Colitis/inducido químicamente , Colitis/microbiología , Colon/microbiología , Enfermedad de Crohn/terapia , Células Epiteliales/metabolismo , Heces/microbiología , Femenino , Proteínas Ligadas a GPI/metabolismo , Humanos , Íleon/microbiología , Mucosa Intestinal/citología , Ratones , Ratones Transgénicos
9.
Nat Microbiol ; 1: 16174, 2016 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-27694949

RESUMEN

The mechanisms by which pathogens evade elimination without affecting host fitness are not well understood. For the pathogen Pseudomonas aeruginosa, this evasion appears to be triggered by excretion of the quorum-sensing molecule 2-aminoacetophenone, which dampens host immune responses and modulates host metabolism, thereby enabling the bacteria to persist at a high burden level. Here, we examined how 2-aminoacetophenone trains host tissues to become tolerant to a high bacterial burden, without compromising host fitness. We found that 2-aminoacetophenone regulates histone deacetylase 1 expression and activity, resulting in hypo-acetylation of lysine 18 of histone H3 at pro-inflammatory cytokine loci. Specifically, 2-aminoacetophenone induced reprogramming of immune cells occurs via alterations in histone acetylation of immune cytokines in vivo and in vitro. This host epigenetic reprograming, which was maintained for up to 7 days, dampened host responses to subsequent exposure to 2-aminoacetophenone or other unrelated pathogen-associated molecules. The process was found to involve a distinct molecular mechanism of host chromatin regulation. Inhibition of histone deacetylase 1 prevented the immunomodulatory effects of 2-aminoacetophenone. These observations provide the first mechanistic example of a quorum-sensing molecule regulating a host epigenome to enable tolerance of infection. These insights have enormous potential for developing preventive treatments against bacterial infections.


Asunto(s)
Acetofenonas/metabolismo , Citocinas/biosíntesis , Epigénesis Genética/efectos de los fármacos , Histona Desacetilasa 1/metabolismo , Interacciones Huésped-Patógeno , Pseudomonas aeruginosa/inmunología , Pseudomonas aeruginosa/metabolismo , Animales , Humanos , Evasión Inmune , Tolerancia Inmunológica , Ratones , Células RAW 264.7 , Células THP-1
10.
Sci Rep ; 6: 34083, 2016 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-27678057

RESUMEN

Pseudomonas aeruginosa defies eradication by antibiotics and is responsible for acute and chronic human infections due to a wide variety of virulence factors. Currently, it is believed that MvfR (PqsR) controls the expression of many of these factors indirectly via the pqs and phnAB operons. Here we provide strong evidence that MvfR may also bind and directly regulate the expression of additional 35 loci across the P. aeruginosa genome, including major regulators and virulence factors, such as the quorum sensing (QS) regulators lasR and rhlR, and genes involved in protein secretion, translation, and response to oxidative stress. We show that these anti-oxidant systems, AhpC-F, AhpB-TrxB2 and Dps, are critical for P. aeruginosa survival to reactive oxygen species and antibiotic tolerance. Considering that MvfR regulated compounds generate reactive oxygen species, this indicates a tightly regulated QS self-defense anti-poisoning system. These findings also challenge the current hierarchical regulation model of P. aeruginosa QS systems by revealing new interconnections between them that suggest a circular model. Moreover, they uncover a novel role for MvfR in self-defense that favors antibiotic tolerance and cell survival, further demonstrating MvfR as a highly desirable anti-virulence target.

11.
ACS Chem Biol ; 11(11): 3061-3067, 2016 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-27658001

RESUMEN

The Gram-negative bacterial pathogen Pseudomonas aeruginosa uses three interconnected intercellular signaling systems regulated by the transcription factors LasR, RhlR, and MvfR (PqsR), which mediate bacterial cell-cell communication via small-molecule natural products and control the production of a variety of virulence factors. The MvfR system is activated by and controls the biosynthesis of the quinolone quorum sensing factors HHQ and PQS. A key step in the biosynthesis of these quinolones is catalyzed by the anthranilyl-CoA synthetase PqsA. To develop inhibitors of PqsA as novel potential antivirulence antibiotics, we report herein the design and synthesis of sulfonyladeonsine-based mimics of the anthranilyl-AMP reaction intermediate that is bound tightly by PqsA. Biochemical, microbiological, and pharmacological studies identified two potent PqsA inhibitors, anthranilyl-AMS (1) and anthranilyl-AMSN (2), that decreased HHQ and PQS production in P. aeruginosa strain PA14. However, these compounds did not inhibit production of the virulence factor pyocyanin. Moreover, they exhibited limited bacterial penetration in compound accumulation studies. This work provides the most potent PqsA inhibitors reported to date and sets the stage for future efforts to develop analogues with improved cellular activity to investigate further the complex relationships between quinolone biosynthesis and virulence factor production in P. aeruginosa and the therapeutic potential of targeting PqsA.


Asunto(s)
Coenzima A Ligasas/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Pseudomonas aeruginosa/efectos de los fármacos , Quinolonas/metabolismo , Bibliotecas de Moléculas Pequeñas , Inhibidores Enzimáticos/química , Pseudomonas aeruginosa/enzimología , Pseudomonas aeruginosa/metabolismo
12.
Curr Opin Microbiol ; 33: 41-46, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27318551

RESUMEN

As antibiotic resistance remains a major public health threat, anti-virulence therapy research is gaining interest. Hundreds of potential anti-virulence compounds have been examined, but very few have made it to clinical trials and none have been approved. This review surveys the current anti-virulence research field with a focus on the highly resistant and deadly ESKAPE pathogens, especially Pseudomonas aeruginosa. We discuss timely considerations and caveats in anti-virulence drug development, including target identification, administration, preclinical development, and metrics for success in clinical trials. Development of a defined pipeline for anti-virulence agents, which differs in important ways from conventional antibiotics, is imperative for the future success of these critically needed drugs.


Asunto(s)
Antibacterianos/farmacología , Biopelículas/efectos de los fármacos , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/patogenicidad , Percepción de Quorum/efectos de los fármacos , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/patogenicidad , Biopelículas/crecimiento & desarrollo , Farmacorresistencia Bacteriana Múltiple/genética , Humanos , Virulencia/efectos de los fármacos
13.
Environ Microbiol ; 18(7): 2237-45, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26971586

RESUMEN

Uropathogenic Escherichia coli (UPEC) is the leading cause of urinary tract infections (UTIs) worldwide, causing over 150 million clinical cases annually. There is currently no specific treatment addressing the asymptomatic carriage in the gut of UPEC before they initiate UTIs. This study investigates the efficacy of virulent bacteriophages to decrease carriage of gut pathogens. Three virulent bacteriophages infecting an antibiotic-resistant UPEC strain were isolated and characterized both in vitro and in vivo. A new experimental murine model of gut carriage of E. coli was elaborated and the impact of virulent bacteriophages on colonization levels and microbiota diversity was assessed. A single dose of a cocktail of the three bacteriophages led to a sharp decrease in E. coli levels throughout the gut. We also observed that microbiota diversity was much less affected by bacteriophages than by antibiotics. Therefore, virulent bacteriophages can efficiently target UPEC strains residing in the gut, with potentially profound public health and economic impacts. These results open a new area with the possibility to manipulate specifically the microbiota using virulent bacteriophages, which could have broad applications in many gut-related disorders/diseases and beyond.


Asunto(s)
Antibacterianos/farmacología , Bacteriófagos/fisiología , Infecciones por Escherichia coli/microbiología , Escherichia coli/virología , Microbioma Gastrointestinal , Animales , Bacteriófagos/genética , Escherichia coli/efectos de los fármacos , Escherichia coli/fisiología , Infecciones por Escherichia coli/tratamiento farmacológico , Infecciones por Escherichia coli/virología , Femenino , Tracto Gastrointestinal/microbiología , Humanos , Ratones , Ratones Endogámicos BALB C , Infecciones Urinarias/tratamiento farmacológico , Infecciones Urinarias/microbiología , Infecciones Urinarias/virología
14.
Curr Biol ; 26(2): 195-206, 2016 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-26776731

RESUMEN

Bacterial programmed cell death and quorum sensing are direct examples of prokaryote group behaviors, wherein cells coordinate their actions to function cooperatively like one organism for the benefit of the whole culture. We demonstrate here that 2-n-heptyl-4-hydroxyquinoline-N-oxide (HQNO), a Pseudomonas aeruginosa quorum-sensing-regulated low-molecular-weight excreted molecule, triggers autolysis by self-perturbing the electron transfer reactions of the cytochrome bc1 complex. HQNO induces specific self-poisoning by disrupting the flow of electrons through the respiratory chain at the cytochrome bc1 complex, causing a leak of reducing equivalents to O2 whereby electrons that would normally be passed to cytochrome c are donated directly to O2. The subsequent mass production of reactive oxygen species (ROS) reduces membrane potential and disrupts membrane integrity, causing bacterial cell autolysis and DNA release. DNA subsequently promotes biofilm formation and increases antibiotic tolerance to beta-lactams, suggesting that HQNO-dependent cell autolysis is advantageous to the bacterial populations. These data identify both a new programmed cell death system and a novel role for HQNO as a critical inducer of biofilm formation and antibiotic tolerance. This newly identified pathway suggests intriguing mechanistic similarities with the initial mitochondrial-mediated steps of eukaryotic apoptosis.


Asunto(s)
Antibacterianos/farmacología , Biopelículas , Transporte de Electrón/efectos de los fármacos , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Pseudomonas aeruginosa/fisiología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Hidroxiquinolinas/farmacología , Membranas Mitocondriales/metabolismo , Pseudomonas aeruginosa/efectos de los fármacos , Percepción de Quorum/efectos de los fármacos , Percepción de Quorum/fisiología
15.
PLoS Pathog ; 10(8): e1004321, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25144274

RESUMEN

Etiological agents of acute, persistent, or relapsing clinical infections are often refractory to antibiotics due to multidrug resistance and/or antibiotic tolerance. Pseudomonas aeruginosa is an opportunistic Gram-negative bacterial pathogen that causes recalcitrant and severe acute chronic and persistent human infections. Here, we target the MvfR-regulated P. aeruginosa quorum sensing (QS) virulence pathway to isolate robust molecules that specifically inhibit infection without affecting bacterial growth or viability to mitigate selective resistance. Using a whole-cell high-throughput screen (HTS) and structure-activity relationship (SAR) analysis, we identify compounds that block the synthesis of both pro-persistence and pro-acute MvfR-dependent signaling molecules. These compounds, which share a benzamide-benzimidazole backbone and are unrelated to previous MvfR-regulon inhibitors, bind the global virulence QS transcriptional regulator, MvfR (PqsR); inhibit the MvfR regulon in multi-drug resistant isolates; are active against P. aeruginosa acute and persistent murine infections; and do not perturb bacterial growth. In addition, they are the first compounds identified to reduce the formation of antibiotic-tolerant persister cells. As such, these molecules provide for the development of next-generation clinical therapeutics to more effectively treat refractory and deleterious bacterial-human infections.


Asunto(s)
Antibacterianos/farmacología , Descubrimiento de Drogas , Farmacorresistencia Bacteriana/efectos de los fármacos , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/efectos de los fármacos , Percepción de Quorum/fisiología , Animales , Inmunoprecipitación de Cromatina , Modelos Animales de Enfermedad , Resistencia a Múltiples Medicamentos/efectos de los fármacos , Ratones , Virulencia/efectos de los fármacos
16.
Methods Mol Biol ; 1149: 699-707, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24818944

RESUMEN

Bacterial persistence, which is observed in a broad range of microbial species, is the capacity of a bacterial cell subpopulation called "persisters" to tolerate exposure to normally lethal concentrations of bactericidal antibiotics. This ability, which is not due to antibiotic-resistant mutants, has been implicated in antibiotic treatment failures and may account for latent, chronic, and relapsing infections. Antibiotic tolerant/Persister (AT/P) cells have been notoriously difficult to study due to their low frequency and transient nature. This chapter describes the main methods used to isolate and study Pseudomonas aeruginosa AT/P cells and discusses new technologies that may ease research of P. aeruginosa persisters in the near future.


Asunto(s)
Antibacterianos/farmacología , Farmacorresistencia Bacteriana/efectos de los fármacos , Pruebas de Sensibilidad Microbiana/métodos , Pseudomonas aeruginosa/citología , Pseudomonas aeruginosa/efectos de los fármacos , Recuento de Colonia Microbiana , Viabilidad Microbiana , Mutación/genética , Pseudomonas aeruginosa/crecimiento & desarrollo , Reproducibilidad de los Resultados
17.
PLoS One ; 8(12): e80140, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24367477

RESUMEN

Bacteria can be refractory to antibiotics due to a sub-population of dormant cells, called persisters that are highly tolerant to antibiotic exposure. The low frequency and transience of the antibiotic tolerant "persister" trait has complicated elucidation of the mechanism that controls antibiotic tolerance. In this study, we show that 2' Amino-acetophenone (2-AA), a poorly studied but diagnostically important small, volatile molecule produced by the recalcitrant gram-negative human pathogen Pseudomonas aeruginosa, promotes antibiotic tolerance in response to quorum-sensing (QS) signaling. Our results show that 2-AA mediated persister cell accumulation occurs via alteration of the expression of genes involved in the translational capacity of the cell, including almost all ribosomal protein genes and other translation-related factors. That 2-AA promotes persisters formation also in other emerging multi-drug resistant pathogens, including the non 2-AA producer Acinetobacter baumannii implies that 2-AA may play an important role in the ability of gram-negative bacteria to tolerate antibiotic treatments in polymicrobial infections. Given that the synthesis, excretion and uptake of QS small molecules is a common hallmark of prokaryotes, together with the fact that the translational machinery is highly conserved, we posit that modulation of the translational capacity of the cell via QS molecules, may be a general, widely distributed mechanism that promotes antibiotic tolerance among prokaryotes.


Asunto(s)
Antibacterianos/farmacología , Acinetobacter baumannii/efectos de los fármacos , Acinetobacter baumannii/metabolismo , Farmacorresistencia Bacteriana Múltiple , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/metabolismo , Percepción de Quorum/efectos de los fármacos
18.
Biol Aujourdhui ; 207(3): 181-90, 2013.
Artículo en Francés | MEDLINE | ID: mdl-24330971

RESUMEN

Bacteriophages were discovered in the early 20th century and rapidly used to treat bacterial infections in humans. As the first specific antibacterial agents, they were used worldwide until antibiotics ramped up. Thereafter, rapidly forgotten, they became the favorite toolbox for researchers that used them to elucidate some of the most fundamental aspects of the cellular life at the molecular level. Today, facing the threat of antibiotic resistant bacteria, bacteriophages are being reconsidered for their use in medicine. During the past century, knowledge on bacteriophages has improved considerably, nevertheless phage therapy is still in its infancy. Taking two examples of recently published experimental phage therapy results, this article summarizes the hopes but also the challenges that surround the future development of human phage therapy.


Asunto(s)
Infecciones Bacterianas/terapia , Bacteriófagos/fisiología , Enfermedades Pulmonares/microbiología , Animales , Modelos Animales de Enfermedad , Farmacorresistencia Bacteriana , Infecciones por Escherichia coli/terapia , Humanos , Enfermedades Intestinales/microbiología , Infecciones por Pseudomonas/terapia , Pseudomonas aeruginosa
19.
BMC Microbiol ; 12: 259, 2012 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-23148795

RESUMEN

BACKGROUND: There are several methods for quantitating bacterial cells, each with advantages and disadvantages. The most common method is bacterial plating, which has the advantage of allowing live cell assessment through colony forming unit (CFU) counts but is not well suited for high throughput screening (HTS). On the other hand, spectrophotometry is adaptable to HTS applications but does not differentiate between dead and living bacteria and has low sensitivity. RESULTS: Here, we report a bacterial cell counting method termed Start Growth Time (SGT) that allows rapid and serial quantification of the absolute or relative number of live cells in a bacterial culture in a high throughput manner. We combined the methodology of quantitative polymerase chain reaction (qPCR) calculations with a previously described qualitative method of bacterial growth determination to develop an improved quantitative method. We show that SGT detects only live bacteria and is sensitive enough to differentiate between 40 and 400 cells/mL. SGT is based on the re-growth time required by a growing cell culture to reach a threshold, and the notion that this time is proportional to the number of cells in the initial inoculum. We show several applications of SGT, including assessment of antibiotic effects on cell viability and determination of an antibiotic tolerant subpopulation fraction within a cell population. SGT results do not differ significantly from results obtained by CFU counts. CONCLUSION: SGT is a relatively quick, highly sensitive, reproducible and non-laborious method that can be used in HTS settings to longitudinally assess live cells in bacterial cell cultures.


Asunto(s)
Bacterias/crecimiento & desarrollo , Bacterias/aislamiento & purificación , Carga Bacteriana/métodos , Ensayos Analíticos de Alto Rendimiento/métodos , Viabilidad Microbiana , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Factores de Tiempo
20.
Antimicrob Agents Chemother ; 56(12): 6235-42, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23006754

RESUMEN

In vivo bacteriophage targeting of enteroaggregative Escherichia coli (EAEC) was assessed using a mouse intestinal model of colonization with the O104:H4 55989Str strain and a cocktail of three virulent bacteriophages. The colonization model was shown to mimic asymptomatic intestinal carriage found in humans. The addition of the cocktail to drinking water for 24 h strongly decreased ileal and weakly decreased fecal 55989Str concentrations in a dose-dependent manner. These decreases in ileal and fecal bacterial concentrations were only transient, since 55989Str concentrations returned to their original levels 3 days later. These transient decreases were independent of the mouse microbiota, as similar results were obtained with axenic mice. We studied the infectivity of each bacteriophage in the ileal and fecal environments and found that 55989Str bacteria in the mouse ileum were permissive to all three bacteriophages, whereas those in the feces were permissive to only one bacteriophage. Our results provide the first demonstration that bacterial permissivity to infection with virulent bacteriophages is not uniform throughout the gut; this highlights the need for a detailed characterization of the interactions between bacteria and bacteriophages in vivo for the further development of phage therapy targeting intestinal pathogens found in the gut of asymptomatic human carriers.


Asunto(s)
Bacteriófagos/fisiología , Infecciones por Escherichia coli/tratamiento farmacológico , Escherichia coli/virología , Enfermedades Intestinales/microbiología , Animales , Carga Bacteriana , Bacteriófagos/efectos de los fármacos , Colon/microbiología , Colon/patología , Infecciones por Escherichia coli/microbiología , Heces/microbiología , Femenino , Íleon/microbiología , Íleon/patología , Inmunohistoquímica , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Reacción en Cadena de la Polimerasa , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...