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

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Antimicrob Agents Chemother ; 68(4): e0172823, 2024 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-38470133

RESUMEN

Left ventricular assist devices (LVAD) are increasingly used for management of heart failure; infection remains a frequent complication. Phage therapy has been successful in a variety of antibiotic refractory infections and is of interest in treating LVAD infections. We performed a retrospective review of four patients that underwent five separate courses of intravenous (IV) phage therapy with concomitant antibiotic for treatment of endovascular Pseudomonas aeruginosa LVAD infection. We assessed phage susceptibility, bacterial strain sequencing, serum neutralization, biofilm activity, and shelf-life of phage preparations. Five treatments of one to four wild-type virulent phage(s) were administered for 14-51 days after informed consent and regulatory approval. There was no successful outcome. Breakthrough bacteremia occurred in four of five treatments. Two patients died from the underlying infection. We noted a variable decline in phage susceptibility following three of five treatments, four of four tested developed serum neutralization, and prophage presence was confirmed in isolates of two tested patients. Two phage preparations showed an initial titer drop. Phage biofilm activity was confirmed in two. Phage susceptibility alone was not predictive of clinical efficacy in P. aeruginosa endovascular LVAD infection. IV phage was associated with serum neutralization in most cases though lack of clinical effect may be multifactorial including presence of multiple bacterial isolates with varying phage susceptibility, presence of prophages, decline in phage titers, and possible lack of biofilm activity. Breakthrough bacteremia occurred frequently (while the organism remained susceptible to administered phage) and is an important safety consideration.


Asunto(s)
Bacteriemia , Bacteriófagos , Corazón Auxiliar , Terapia de Fagos , Infecciones por Pseudomonas , Humanos , Pseudomonas aeruginosa , Corazón Auxiliar/efectos adversos , Infecciones por Pseudomonas/terapia , Infecciones por Pseudomonas/microbiología , Antibacterianos/uso terapéutico , Profagos , Bacteriemia/tratamiento farmacológico
2.
Clin Infect Dis ; 77(Suppl 5): S337-S351, 2023 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-37932122

RESUMEN

Using phages as salvage therapy for nonhealing infections is gaining recognition as a viable solution for patients with such infections. The escalating issue of antibiotic resistance further emphasizes the significance of using phages in treating bacterial infections, encompassing compassionate-use scenarios and clinical trials. Given the high specificity of phages, selecting the suitable phage(s) targeting the causative bacteria becomes critical for achieving treatment success. However, in contrast to conventional antibiotics, where susceptibility-testing procedures were well established for phage therapy, there is a lack of standard frameworks for matching phages from a panel to target bacterial strains and assessing their interactions with antibiotics or other agents. This review discusses and compares published methods for clinical phage microbiology, also known as phage susceptibility testing, and proposes guidelines for establishing a standard pipeline based on our findings over the past 5 years of phage therapy at the Israeli Phage Therapy Center.


Asunto(s)
Bacteriófagos , Terapia de Fagos , Humanos , Israel , Bacterias , Antibacterianos , Estándares de Referencia
3.
Int J Mol Sci ; 25(1)2023 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-38203644

RESUMEN

Dysbiosis of oral microbiota is associated with the initiation and progression of periodontitis. The cause-and-effect relationship between genetics, periodontitis, and oral microbiome dysbiosis is poorly understood. Here, we demonstrate the power of the collaborative cross (CC) mice model to assess the effect of the genetic background on microbiome diversity shifts during periodontal infection and host suitability status. We examined the bacterial composition in plaque samples from seven different CC lines using 16s rRNA sequencing before and during periodontal infection. The susceptibility/resistance of the CC lines to alveolar bone loss was determined using the micro-CT technique. A total of 53 samples (7 lines) were collected before and after oral infection using oral swaps followed by DNA extraction and 16 s rRNA sequencing analysis. CC lines showed a significant variation in response to the co-infection (p < 0.05). Microbiome compositions were significantly different before and after infection and between resistant and susceptible lines to periodontitis (p < 0.05). Gram-positive taxa were significantly higher at the resistant lines compared to susceptible lines (p < 0.05). Gram-positive bacteria were reduced after infection, and gram-negative bacteria, specifically anaerobic groups, increased after infection. Our results demonstrate the utility of the CC mice in exploring the interrelationship between genetic background, microbiome composition, and periodontitis.


Asunto(s)
Pérdida de Hueso Alveolar , Periodontitis , Animales , Ratones , Pérdida de Hueso Alveolar/genética , Disbiosis/genética , ARN Ribosómico 16S/genética , Cognición , Periodontitis/genética
4.
PLoS Biol ; 17(3): e3000182, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30925180

RESUMEN

In experimental evolution, scientists evolve organisms in the lab, typically by challenging them to new environmental conditions. How best to evolve a desired trait? Should the challenge be applied abruptly, gradually, periodically, sporadically? Should one apply chemical mutagenesis, and do strains with high innate mutation rate evolve faster? What are ideal population sizes of evolving populations? There are endless strategies, beyond those that can be exposed by individual labs. We therefore arranged a community challenge, Evolthon, in which students and scientists from different labs were asked to evolve Escherichia coli or Saccharomyces cerevisiae for an abiotic stress-low temperature. About 30 participants from around the world explored diverse environmental and genetic regimes of evolution. After a period of evolution in each lab, all strains of each species were competed with one another. In yeast, the most successful strategies were those that used mating, underscoring the importance of sex in evolution. In bacteria, the fittest strain used a strategy based on exploration of different mutation rates. Different strategies displayed variable levels of performance and stability across additional challenges and conditions. This study therefore uncovers principles of effective experimental evolutionary regimens and might prove useful also for biotechnological developments of new strains and for understanding natural strategies in evolutionary arms races between species. Evolthon constitutes a model for community-based scientific exploration that encourages creativity and cooperation.


Asunto(s)
Evolución Biológica , Escherichia coli/metabolismo , Humanos , Modelos Genéticos , Mutación/genética , Saccharomyces cerevisiae/metabolismo , Temperatura
5.
Acta Derm Venereol ; 100(17): adv00295, 2020 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-33021324

RESUMEN

Antibiotic-resistant Cutibacterium acnes has been reported worldwide, but data from Israeli patients with acne is currently lacking. This study evaluated the antibiotic susceptibility of C. acnes, isolated from 50 Israeli patients with acne to commonly prescribed antibiotics, using the Epsilometer test (E-test). Matrix-assisted laser desorption ionization-time of flight (MALDI-TOF) analysis, 16S rRNA sequencing and single locus sequence typing (SLST) molecular typing were used to identify and characterize C. acnes. Among 36 strains isolated, phylotype IA1 was most common. Resistance to at least one antibiotic was found in 30.6% of tested strains. Resistance rates were highest for erythromycin (25.0%), followed by doxycycline (19.4%), clindamycin (16.7%), minocycline (11.1%) and tetracycline (8.3%). Significant correlation was found between resistance to multiple antibiotics, with 5.6% of isolates resistant to all antibiotics tested. When reviewing resistances rate worldwide antibiotic resistance was found to be prevalent in Israel. Measures to limit the emergence of antibiotic-resistant strains of Cutibacterium acnes should be taken and alternative treatments should be sought.


Asunto(s)
Acné Vulgar , Propionibacterium acnes , Acné Vulgar/diagnóstico , Acné Vulgar/tratamiento farmacológico , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Humanos , Israel/epidemiología , Pruebas de Sensibilidad Microbiana , Propionibacterium acnes/genética , ARN Ribosómico 16S/genética
7.
Appl Environ Microbiol ; 81(8): 2696-705, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25662974

RESUMEN

Phage therapy has been proven to be more effective, in some cases, than conventional antibiotics, especially regarding multidrug-resistant biofilm infections. The objective here was to isolate an anti-Enterococcus faecalis bacteriophage and to evaluate its efficacy against planktonic and biofilm cultures. E. faecalis is an important pathogen found in many infections, including endocarditis and persistent infections associated with root canal treatment failure. The difficulty in E. faecalis treatment has been attributed to the lack of anti-infective strategies to eradicate its biofilm and to the frequent emergence of multidrug-resistant strains. To this end, an anti-E. faecalis and E. faecium phage, termed EFDG1, was isolated from sewage effluents. The phage was visualized by electron microscopy. EFDG1 coding sequences and phylogeny were determined by whole genome sequencing (GenBank accession number KP339049), revealing it belongs to the Spounavirinae subfamily of the Myoviridae phages, which includes promising candidates for therapy against Gram-positive pathogens. This analysis also showed that the EFDG1 genome does not contain apparent harmful genes. EFDG1 antibacterial efficacy was evaluated in vitro against planktonic and biofilm cultures, showing effective lytic activity against various E. faecalis and E. faecium isolates, regardless of their antibiotic resistance profile. In addition, EFDG1 efficiently prevented ex vivo E. faecalis root canal infection. These findings suggest that phage therapy using EFDG1 might be efficacious to prevent E. faecalis infection after root canal treatment.


Asunto(s)
Biopelículas , Cavidad Pulpar/microbiología , Enfermedades de la Pulpa Dental/prevención & control , Enterococcus faecalis/fisiología , Genoma Viral , Infecciones por Bacterias Grampositivas/prevención & control , Myoviridae/fisiología , Enfermedades de la Pulpa Dental/microbiología , Enterococcus faecalis/virología , Infecciones por Bacterias Grampositivas/microbiología , Datos de Secuencia Molecular , Myoviridae/genética , Plancton/fisiología , Plancton/virología , Análisis de Secuencia de ADN , Aguas del Alcantarillado/virología
8.
STAR Protoc ; 5(2): 102949, 2024 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-38691464

RESUMEN

Phage therapy has re-emerged as a promising treatment for non-resolving infections. Given the lack of approved phage treatments, there is a need to establish a compassionate use pipeline. Here, we present a protocol for phage matching, treatment, and monitoring for compassionate bacteriophage use in non-resolving infections. We describe steps for consultation and request implementation, evaluating and comparing different aspects of phage activity, and phage production. We then detail procedures for multidisciplinary meetings, ethics approvals, phage therapy, and follow-up. For complete details on the use and execution of this protocol, please refer to Onallah et al.1,2.


Asunto(s)
Bacteriófagos , Ensayos de Uso Compasivo , Terapia de Fagos , Humanos , Bacteriófagos/fisiología , Terapia de Fagos/métodos , Infecciones Bacterianas/terapia
9.
Vet Q ; 44(1): 1-9, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38726795

RESUMEN

Antibiotic-resistant pathogens are a growing global issue, leading to untreatable infectious diseases in both humans and animals. Personalized bacteriophage (phage) therapy, the use of specific anti-bacterial viruses, is currently a leading approach to combat antibiotic-resistant infections. The implementation of phage therapy has primarily been focused on humans, almost neglecting the impact of such infections on the health and welfare of companion animals. Pets also have the potential to spread resistant infections to their owners or the veterinary staff through zoonotic transmission. Here, we showcase personalized phage-antibiotic treatment of a cat with a multidrug-resistant Pseudomonas aeruginosa implant-associated infection post-arthrodesis surgery. The treatment encompassed a tailored combination of an anti-P. aeruginosa phage and ceftazidime, precisely matched to the pathogen. The phage was topically applied to the surgical wound while the antibiotic was administered intramuscularly. After two treatment courses spanning 7 and 3 weeks, the surgical wound, which had previously remained open for five months, fully closed. To the best of our knowledge, this is the first case of personalized phage therapy application in felines, which provides further evidence of the effectiveness of this approach. The successful outcome paves the way for personalized phage-antibiotic treatments against persistent infections therapy in veterinary practice.


Asunto(s)
Antibacterianos , Enfermedades de los Gatos , Terapia de Fagos , Infecciones por Pseudomonas , Pseudomonas aeruginosa , Animales , Gatos , Terapia de Fagos/veterinaria , Infecciones por Pseudomonas/veterinaria , Infecciones por Pseudomonas/tratamiento farmacológico , Infecciones por Pseudomonas/terapia , Enfermedades de los Gatos/terapia , Enfermedades de los Gatos/tratamiento farmacológico , Enfermedades de los Gatos/microbiología , Pseudomonas aeruginosa/efectos de los fármacos , Antibacterianos/uso terapéutico , Ceftazidima/uso terapéutico , Farmacorresistencia Bacteriana Múltiple , Bacteriófagos
10.
PLoS Pathog ; 7(8): e1002192, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21829370

RESUMEN

A significant number of environmental microorganisms can cause serious, even fatal, acute and chronic infections in humans. The severity and outcome of each type of infection depends on the expression of specific bacterial phenotypes controlled by complex regulatory networks that sense and respond to the host environment. Although bacterial signals that contribute to a successful acute infection have been identified in a number of pathogens, the signals that mediate the onset and establishment of chronic infections have yet to be discovered. We identified a volatile, low molecular weight molecule, 2-amino acetophenone (2-AA), produced by the opportunistic human pathogen Pseudomonas aeruginosa that reduces bacterial virulence in vivo in flies and in an acute mouse infection model. 2-AA modulates the activity of the virulence regulator MvfR (multiple virulence factor regulator) via a negative feedback loop and it promotes the emergence of P. aeruginosa phenotypes that likely promote chronic lung infections, including accumulation of lasR mutants, long-term survival at stationary phase, and persistence in a Drosophila infection model. We report for the first time the existence of a quorum sensing (QS) regulated volatile molecule that induces bistability phenotype by stochastically silencing acute virulence functions in P. aeruginosa. We propose that 2-AA mediates changes in a subpopulation of cells that facilitate the exploitation of dynamic host environments and promote gene expression changes that favor chronic infections.


Asunto(s)
Acetofenonas/metabolismo , Proteínas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica/fisiología , Neumonía Bacteriana/metabolismo , Infecciones por Pseudomonas/metabolismo , Pseudomonas aeruginosa/metabolismo , Pseudomonas aeruginosa/patogenicidad , Percepción de Quorum/fisiología , Transactivadores/metabolismo , Enfermedad Aguda , Animales , Proteínas Bacterianas/genética , Enfermedad Crónica , Modelos Animales de Enfermedad , Drosophila melanogaster , Humanos , Ratones , Mutación , Neumonía Bacteriana/genética , Infecciones por Pseudomonas/genética , Pseudomonas aeruginosa/genética , Transactivadores/genética
11.
Open Forum Infect Dis ; 10(5): ofad221, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-37234511

RESUMEN

The use of bacteriophages (phages) is reemerging as a potential treatment option for antibiotic-resistant or nonresolving bacterial infections. Phages are bacteria-specific viruses that may serve as a personalized therapeutic option with minimal collateral damage to the patient or the microbiome. In 2018 we established the Israeli Phage Therapy Center (IPTC) as a shared initiative of the Hadassah Medical Center and the Hebrew University of Jerusalem, aiming to conduct all of the steps required for phage-based solutions, from phage isolation and characterization to treatments, for nonresolving bacterial infections. So far, a total of 159 requests for phage therapy arrived to the IPTC; 145 of them were from Israel and the rest from other countries. This number of registered requests is growing annually. Multidrug-resistant bacteria accounted for 38% of all phage requests. Respiratory and bone infections were the most prevalent among clinical indications and accounted for 51% of the requests. To date, 20 phage therapy courses were given to 18 patients by the IPTC. In 77.7% (n = 14) of the cases, a favorable clinical outcome of infection remission or recovery was seen. Clearly, establishing an Israeli phage center has led to an increased demand for compassionate use of phages with favorable outcomes for many previously failed infections. As clinical trials are still lacking, publishing patient data from cohort studies is pertinent to establish clinical indications, protocols, and success and failure rates. Last, workflow processes and bottlenecks should be shared to enable faster availability and authorization of phages for clinical use.

12.
ACS Appl Mater Interfaces ; 15(43): 50330-50343, 2023 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-37861446

RESUMEN

Multifunctional drug-loaded polymer-metal nanocapsules have attracted increasing attention in drug delivery due to their multifunctional potential endowed by drug activity and response to physicochemical stimuli. Current chemical synthesis methods of polymer/metal capsules require specific optimization of the different components to produce particles with precise properties, being particularly complex for Janus structures combining polymers and ferromagnetic and highly reactive metals. With the aim to generate tunable synergistic nanotherapeutic actuation with enhanced drug effects, here we demonstrate a versatile hybrid chemical/physical fabrication strategy to incorporate different functional metals with tailored magnetic, optical, or chemical properties on solid drug-loaded polymer nanoparticles. As archetypical examples, we present poly(lactic-co-glycolic acid) (PLGA) nanoparticles (diameters 100-150 nm) loaded with paclitaxel, indocyanine green, or erythromycin that are half-capped by either Fe, Au, or Cu layers, respectively, with application in three biomedical models. The Fe coating on paclitaxel-loaded nanocapsules permitted efficient magnetic enhancement of the cancer spheroid assembly, with 40% reduction of the cross-section area after 24 h, as well as a higher paclitaxel effect. In addition, the Fe-PLGA nanocapsules enabled external contactless manipulation of multicellular cancer spheroids with a speed of 150 µm/s. The Au-coated and indocyanine green-loaded nanocapsules demonstrated theranostic potential and enhanced anticancer activity in vitro and in vivo due to noninvasive fluorescence imaging with long penetration near-infrared (NIR) light and simultaneous photothermal-photodynamic actuation, showing a 3.5-fold reduction in the tumor volume growth with only 5 min of NIR illumination. Finally, the Cu-coated erythromycin-loaded nanocapsules exhibited enhanced antibacterial activity with a 2.5-fold reduction in the MIC50 concentration with respect to the free or encapsulated drug. Altogether, this technology can extend a nearly unlimited combination of metals, polymers, and drugs, thus enabling the integration of magnetic, optical, and electrochemical properties in drug-loaded nanoparticles to externally control and improve a wide range of biomedical applications.


Asunto(s)
Nanocápsulas , Nanocápsulas/química , Verde de Indocianina/farmacología , Verde de Indocianina/química , Línea Celular Tumoral , Paclitaxel/farmacología , Polímeros/química , Eritromicina/farmacología
13.
Nat Commun ; 14(1): 1005, 2023 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-36813793

RESUMEN

Acne vulgaris is a common neutrophil-driven inflammatory skin disorder in which Cutibacterium acnes (C. acnes) is known to play a key role. For decades, antibiotics have been widely employed to treat acne vulgaris, inevitably resulting in increased bacterial antibiotic resistance. Phage therapy is a promising strategy to combat the growing challenge of antibiotic-resistant bacteria, utilizing viruses that specifically lyse bacteria. Herein, we explore the feasibility of phage therapy against C. acnes. Eight novel phages, isolated in our laboratory, and commonly used antibiotics eradicate 100% of clinically isolated C. acnes strains. Topical phage therapy in a C. acnes-induced acne-like lesions mouse model affords significantly superior clinical and histological scores. Moreover, the decrease in inflammatory response was reflected by the reduced expression of chemokine CXCL2, neutrophil infiltration, and other inflammatory cytokines when compared with the infected-untreated group. Overall, these findings indicate the potential of phage therapy for acne vulgaris as an additional tool to conventional antibiotics.


Asunto(s)
Acné Vulgar , Terapia de Fagos , Animales , Ratones , Antibacterianos/farmacología , Piel/microbiología , Farmacorresistencia Bacteriana , Propionibacterium acnes
14.
Med ; 4(9): 600-611.e4, 2023 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-37562400

RESUMEN

BACKGROUND: A growing number of compassionate phage therapy cases were reported in the last decade, with a limited number of clinical trials conducted and few unsuccessful clinical trials reported. There is only a little evidence on the role of phages in refractory infections. Our objective here was to present the largest compassionate-use single-organism/phage case series in 16 patients with non-resolving Pseudomonas aeruginosa infections. METHODS: We summarized clinical phage microbiology susceptibility data, administration protocol, clinical data, and outcomes of all cases treated with PASA16 phage. In all intravenous phage administrations, PASA16 phage was manufactured and provided pro bono by Adaptive Phage Therapeutics. PASA16 was administered intravenously, locally to infection site, or by topical use to 16 patients, with data available for 15 patients, mainly with osteoarticular and foreign-device-associated infections. FINDINGS: A few minor side effects were noted, including elevated liver function enzymes and a transient reduction in white blood cell count. Good clinical outcome was documented in 13 out of 15 patients (86.6%). Two clinical failures were reported. The minimum therapy duration was 8 days with a once- to twice-daily regimen. CONCLUSIONS: PASA16 with antibiotics was found to be relatively successful in patients for whom traditional treatment approaches have failed previously. Such pre-phase-1 cohorts can outline potential clinical protocols and facilitate the design of future trials. FUNDING: The study was funded in part by The Israeli Science Foundation IPMP (ISF_1349/20), Rosetrees Trust (A2232), United States-Israel Binational Science Foundation (2017123), and the Milgrom Family Support Program.


Asunto(s)
Bacteriófagos , Infecciones por Pseudomonas , Fagos Pseudomonas , Humanos , Infecciones por Pseudomonas/tratamiento farmacológico , Infecciones por Pseudomonas/microbiología , Ensayos de Uso Compasivo , Antibacterianos/uso terapéutico
15.
PLoS Pathog ; 6(3): e1000810, 2010 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-20300606

RESUMEN

Pathogenic bacteria use interconnected multi-layered regulatory networks, such as quorum sensing (QS) networks to sense and respond to environmental cues and external and internal bacterial cell signals, and thereby adapt to and exploit target hosts. Despite the many advances that have been made in understanding QS regulation, little is known regarding how these inputs are integrated and processed in the context of multi-layered QS regulatory networks. Here we report the examination of the Pseudomonas aeruginosa QS 4-hydroxy-2-alkylquinolines (HAQs) MvfR regulatory network and determination of its interaction with the QS acyl-homoserine-lactone (AHL) RhlR network. The aim of this work was to elucidate paradigmatically the complex relationships between multi-layered regulatory QS circuitries, their signaling molecules, and the environmental cues to which they respond. Our findings revealed positive and negative homeostatic regulatory loops that fine-tune the MvfR regulon via a multi-layered dependent homeostatic regulation of the cell-cell signaling molecules PQS and HHQ, and interplay between these molecules and iron. We discovered that the MvfR regulon component PqsE is a key mediator in orchestrating this homeostatic regulation, and in establishing a connection to the QS rhlR system in cooperation with RhlR. Our results show that P. aeruginosa modulates the intensity of its virulence response, at least in part, through this multi-layered interplay. Our findings underscore the importance of the homeostatic interplay that balances competition within and between QS systems via cell-cell signaling molecules and environmental cues in the control of virulence gene expression. Elucidation of the fine-tuning of this complex relationship offers novel insights into the regulation of these systems and may inform strategies designed to limit infections caused by P. aeruginosa and related human pathogens.


Asunto(s)
Homeostasis/fisiología , Hierro/metabolismo , Pseudomonas aeruginosa , Regulón/fisiología , Transducción de Señal/fisiología , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Dípteros , Retroalimentación Fisiológica/fisiología , Femenino , Regulación Bacteriana de la Expresión Génica , Genes Reporteros , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/metabolismo , Pseudomonas aeruginosa/patogenicidad , Percepción de Quorum/fisiología , Virulencia
16.
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
17.
Dig Dis Sci ; 57(8): 2130-6, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22447433

RESUMEN

BACKGROUND: NOTES via the gastrointestinal tract raises the specter of intra-peritoneal infection. Various anti-microbial techniques have been employed in animal and human survival studies, including saline lavage, intravenous and topical antibiotics, and povidone-iodine, although there is a paucity of data regarding their general effectiveness. AIM: To assess the effectiveness of existing sterilization techniques for NOTES by quantifying and speciating colony-forming units (CFUs) before and after treatment. DESIGN: Ex vivo animal studies; bacteriological study. METHODS: Stomachs and distal colons were harvested en bloc from ten fasted adult white pigs following euthanasia. Half received cefazolin 1 g intravenously prior to killing. Multiple tissue samples were obtained from each resected organ. Each tissue sample was then assigned to one of five treatment arms: (1) normal saline, (2) Betadine, (3) cefazolin/metronidazole suspension, (4) chlorhexidine, (5) no treatment. Fifteen samples were used per arm. After treatment, the mucosal surface of each sample was swabbed and inoculated in normal saline, followed by serial dilutions, which were then plated onto sheep's blood agar plates and incubated under aerobic and anaerobic conditions. CFUs were quantified and speciated. RESULTS: Median bacterial density was estimated to be 8.0 × 10(5) CFUs/ml (stomach) and 1.9 × 10(6) CFUs/ml (colon). The predominant organisms were Escherichia coli (stomach) and both E. coli and Enterococcus sp. (colon). Saline and antibiotic suspension lavages caused a 1-log reduction in stomach and colon. Betadine/chlorhexidine lavage resulted in a 4-log reduction. Intravenous antibiotics alone resulted in a 4-log reduction. Combining intravenous antibiotics and either Betadine or chlorhexidine decreased counts to the 10(1) level. By Kruskal-Wallis method, differences were statistically significant (p = 0.001). CONCLUSIONS: The use of intravenous antibiotics in addition to topical Betadine or chlorhexidine effectively reduced microbial burden in both gastric and colonic mucosa in this porcine model to the 10(1) level.


Asunto(s)
Antibacterianos/administración & dosificación , Antiinfecciosos Locales/uso terapéutico , Cefazolina/administración & dosificación , Infecciones Intraabdominales/microbiología , Cirugía Endoscópica por Orificios Naturales/efectos adversos , Animales , Colon/microbiología , Femenino , Lavado Gástrico , Mucosa Gástrica/microbiología , Inyecciones Intravenosas , Mucosa Intestinal/microbiología , Infecciones Intraabdominales/prevención & control , Porcinos
18.
Phage (New Rochelle) ; 3(2): 85-94, 2022 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-36157284

RESUMEN

The fascinating scientific history of phage therapy has been documented in numerous publications. In this study, however, we focus on an angle of the story that hitherto has remained relatively neglected, namely, phage therapy treatments, and the protagonists that conducted these in Mandatory-Palestine and subsequently the state of Israel, as part of a global trend. We complete the story by describing efforts in the new era of phage therapy in present-day Israel.

19.
Microbiol Resour Announc ; 11(4): e0009222, 2022 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-35258339

RESUMEN

PASA16 is a Pseudomonas aeruginosa phage isolated from a soil sample and used to treat several patients suffering from persistent infections in various countries. PASA16's genome was sequenced, analyzed, and deposited in GenBank.

20.
iScience ; 24(7): 102745, 2021 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-34258566

RESUMEN

Numerous observations demonstrate that microorganisms can survive very long periods of nutrient deprivation and starvation. Moreover, it is evident that prolonged periods of starvation are a feature of many habitats, and many cells in all kingdoms of life are found in prolonged starvation conditions. Bacteria exhibit a range of responses to long-term starvation. These include genetic adaptations such as the long-term stationary phase and the growth advantage in stationary phase phenotypes characterized by mutations in stress-signaling genes and elevated mutation rates. Here, we suggest using the term "endurance of prolonged nutrient prevention" (EPNP phase), to describe this phase, which was also recently described in eukaryotes. Here, we review this literature and describe the current knowledge about the adaptations to very long-term starvation conditions in bacteria and eukaryotes, its conceptual and structural conservation across all kingdoms of life, and point out possible directions that merit further research.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA