Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 556(7699): 103-107, 2018 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-29590091

RESUMEN

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


Asunto(s)
Antibacterianos/clasificación , Antibacterianos/farmacología , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Retinoides/farmacología , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/microbiología , Animales , Antibacterianos/efectos adversos , Antibacterianos/uso terapéutico , Benzoatos/química , Benzoatos/farmacología , Benzoatos/uso terapéutico , Benzoatos/toxicidad , Caenorhabditis elegans/efectos de los fármacos , Caenorhabditis elegans/microbiología , Muerte Celular/efectos de los fármacos , Línea Celular , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Sinergismo Farmacológico , Gentamicinas/farmacología , Gentamicinas/uso terapéutico , Humanos , Membrana Dobles de Lípidos/química , Staphylococcus aureus Resistente a Meticilina/citología , Staphylococcus aureus Resistente a Meticilina/genética , Staphylococcus aureus Resistente a Meticilina/crecimiento & desarrollo , Ratones , Pruebas de Sensibilidad Microbiana , Simulación de Dinámica Molecular , Mutación , Naftoles/química , Naftoles/farmacología , Naftoles/uso terapéutico , Naftoles/toxicidad , Retinoides/química , Retinoides/uso terapéutico , Retinoides/toxicidad
2.
Infect Immun ; 91(1): e0037822, 2023 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-36602381

RESUMEN

Recent studies have found that the coexistence of fungi and bacteria in the airway may increase the risk of infection, contribute to the development of pneumonia, and increase the severity of disease. Interleukin 17A (IL-17A) plays important roles in host resistance to bacterial and fungal infections. The objective of this study was to determine the effects of IL-17A on Acinetobacter baumannii-infected rats with a previous Candida albicans airway inoculation. The incidence of A. baumannii pneumonia was higher in rats with C. albicans in the airway than in noninoculated rats, and it decreased when amphotericin B was used to clear C. albicans, which influenced IL-17A levels. IL-17A had a protective effect in A. baumannii pneumonia associated with C. albicans in the airway. Compared with A. baumannii-infected rats with C. albicans in the airway that did not receive IL-17A, recombinant IL-17A (rIL-17A) supplementation decreased the incidence of A. baumannii pneumonia (10/15 versus 5/17; P = 0.013) and the proportion of neutrophils in the lung (84 ± 3.5 versus 74 ± 4.3%; P = 0.033), reduced tissue destruction and inflammation, and decreased levels of myeloperoxidase (MPO) (1.267 ± 0.15 versus 0.233 ± 0.06 U/g; P = 0.0004), reactive oxygen species (ROS) (132,333 ± 7,505 versus 64,667 ± 10,115 AU; P = 0.0007) and lactate dehydrogenase (LDH) (2.736 ± 0.05 versus 2.1816 ± 0.29 U/g; P = 0.0313). In vitro experiments revealed that IL-17A had no significant effect on the direct migration ability and bactericidal capability of neutrophils. However, IL-17A restrained lysis cell death and increased apoptosis of neutrophils (2.9 ± 1.14 versus 7 ± 0.5%; P = 0.0048). Taken together, our results suggest that C. albicans can depress IL-17A levels, which when supplemented may have a regulatory function that limits the accumulation of neutrophils in inflammatory areas, providing inflammatory response homeostasis.


Asunto(s)
Infecciones por Acinetobacter , Acinetobacter baumannii , Neumonía Bacteriana , Neumonía , Ratas , Animales , Candida albicans/metabolismo , Interleucina-17/metabolismo , Acinetobacter baumannii/metabolismo , Pulmón/metabolismo , Neutrófilos/metabolismo , Bacterias/metabolismo
3.
Proc Natl Acad Sci U S A ; 116(33): 16529-16534, 2019 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-31358625

RESUMEN

Treatment of Staphylococcus aureus infections is complicated by the development of antibiotic tolerance, a consequence of the ability of S. aureus to enter into a nongrowing, dormant state in which the organisms are referred to as persisters. We report that the clinically approved anthelmintic agent bithionol kills methicillin-resistant S. aureus (MRSA) persister cells, which correlates with its ability to disrupt the integrity of Gram-positive bacterial membranes. Critically, bithionol exhibits significant selectivity for bacterial compared with mammalian cell membranes. All-atom molecular dynamics (MD) simulations demonstrate that the selectivity of bithionol for bacterial membranes correlates with its ability to penetrate and embed in bacterial-mimic lipid bilayers, but not in cholesterol-rich mammalian-mimic lipid bilayers. In addition to causing rapid membrane permeabilization, the insertion of bithionol increases membrane fluidity. By using bithionol and nTZDpa (another membrane-active antimicrobial agent), as well as analogs of these compounds, we show that the activity of membrane-active compounds against MRSA persisters positively correlates with their ability to increase membrane fluidity, thereby establishing an accurate biophysical indicator for estimating antipersister potency. Finally, we demonstrate that, in combination with gentamicin, bithionol effectively reduces bacterial burdens in a mouse model of chronic deep-seated MRSA infection. This work highlights the potential repurposing of bithionol as an antipersister therapeutic agent.


Asunto(s)
Antibacterianos/farmacología , Membrana Celular/efectos de los fármacos , Reposicionamiento de Medicamentos , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Animales , Bitionol/farmacología , Permeabilidad de la Membrana Celular/efectos de los fármacos , Colesterol/química , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Gentamicinas/farmacología , Membrana Dobles de Lípidos/química , Fluidez de la Membrana/efectos de los fármacos , Staphylococcus aureus Resistente a Meticilina/ultraestructura , Simulación de Dinámica Molecular , Fosfatidilcolinas/química , Relación Estructura-Actividad , Liposomas Unilamelares
4.
Cell Microbiol ; 22(10): e13234, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32543022

RESUMEN

Cutibacterium acnes is capable of inducing inflammation in acne and can lead to a chronic prostatic infection. The diverse pathogenicity among different strains of C. acnes has been presented, but simple appropriate animal models for the evaluation of this bacterium are lacking. In this study, the nematode Caenorhabditis elegans was used as an invertebrate infection model. We revealed that C. acnes type strain ATCC 6919 caused lethal infections to C. elegans in solid and liquid culture media (p < .0001). Compared with the strain ATCC 6919, the antibiotic-resistant strain HM-513 was more virulent, resulting in reduced survival (p < .0001). Four different C. acnes strains killed worms with a p value of less than .0001 when provided to C. elegans at 4.8 × 108 CFU/ml. The infection model was also employed to explore host defence responses. An increase in numerous immune effectors in response to C. acnes was detected. We focused on nine C-type lectins, including: clec-13, clec-17, clec-47, clec-52, clec-60, clec-61, clec-70, clec-71 and clec-227. The induced expression of these C-type lectin genes was down-regulated in mutant worms deficient in the p38 mitogen-activated protein kinase (MAPK) pathway. Meanwhile, PMK-1 (MAPK) was phosphorylated and activated at the onset of C. acnes infection. By monitoring the survival of mutant worms, we found that PMK-1, SEK-1 (MAPKK) and TIR-1 (MAPKKK) were critical in responding to C. acnes infection. C. elegans pmk-1 and tir-1 mutants exhibited higher mortality to C. acnes infection (p < .0001). In conclusion, C. elegans serves as a simple and valuable model to study C. acnes virulence and facilitates improvements in understanding of host innate immune responses.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/microbiología , Lectinas Tipo C/metabolismo , Sistema de Señalización de MAP Quinasas , Propionibacteriaceae/patogenicidad , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Caenorhabditis elegans/inmunología , Proteínas de Caenorhabditis elegans/genética , Regulación hacia Abajo , Inmunidad Innata , Lectinas Tipo C/genética , MAP Quinasa Quinasa 4/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosforilación , Receptores Acoplados a Proteínas G/metabolismo
5.
Artículo en Inglés | MEDLINE | ID: mdl-32253208

RESUMEN

Probiotics might provide an alternative approach for the control of oral candidiasis. However, studies on the antifungal activity of probiotics in the oral cavity are based on the consumption of yogurt or other dietary products, and it is necessary to use appropriate biomaterials and specific strains to obtain probiotic formulations targeted for local oral administration. In this study, we impregnated gellan gum, a natural biopolymer used as a food additive, with a probiotic and investigated its antifungal activity against Candida albicansLactobacillus paracasei 28.4, a strain recently isolated from the oral cavity of a caries-free individual, was incorporated in several concentrations of gellan gum (0.6% to 1% [wt/vol]). All tested concentrations could incorporate L. paracasei cells while maintaining bacterial viability. Probiotic-gellan gum formulations were stable for 7 days when stored at room temperature or 4°C. Long-term storage of bacterium-impregnated gellan gum was achieved when L. paracasei 28.4 was lyophilized. The probiotic-gellan gum formulations provided a release of L. paracasei cells over 24 h that was sufficient to inhibit the growth of C. albicans, with effects dependent on the cell concentrations incorporated into gellan gum. The probiotic-gellan gum formulations also had inhibitory activity against Candida sp. biofilms by reducing the number of Candida sp. cells (P < 0.0001), decreasing the total biomass (P = 0.0003), and impairing hyphae formation (P = 0.0002), compared to the control group which received no treatment. Interestingly, a probiotic formulation of 1% (wt/vol) gellan gum provided an oral colonization of L. paracasei in mice with approximately 6 log CFU/ml after 10 days. This formulation inhibited C. albicans growth (P < 0.0001), prevented the development of candidiasis lesions (P = 0.0013), and suppressed inflammation (P = 0.0006) compared to the mice not treated in the microscopic analysis of the tongue dorsum. These results indicate that gellan gum is a promising biomaterial and can be used as a carrier system to promote oral colonization for probiotics that prevent oral candidiasis.


Asunto(s)
Candidiasis Bucal , Lacticaseibacillus paracasei , Probióticos , Animales , Ratones , Polisacáridos Bacterianos
6.
Microb Pathog ; 117: 80-87, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29432910

RESUMEN

The objective of this study was to evaluate the influence of microbe-microbe interactions to identify a strain of Lactobacillus that could reduce the filamentation of Candida albicans ATCC 18804 using in vitro and in vivo models. Thus presenting a probiotic effect against the fungal pathogen. First, we analyzed the ability of 25 clinical isolates of Lactobacillus to reduce filamentation in C. albicans in vitro. We found that L. paracasei isolate 28.4 exhibited the greatest reduction of C. albicans hyphae (p = 0.0109). This reduction was confirmed by scanning electron microscopy analysis. The influence of C. albicans filamentation was found to be contributed through reduced gene expression of filament associated genes (TEC1 and UME6). In an in vivo study, prophylactic provisions with L. paracasei increased the survival of Caenorhabditis elegans worms infected with C. albicans (p = 0.0001) by 29%. Prolonged survival was accompanied by the prevention of cuticle rupture of 27% of the worms by filamentation of C. albicans, a phenotype that is characteristic of C. albicans killing of nematodes, compared to the control group. Lactobacillus paracasei isolate 28.4 reduced the filamentation of C. albicans in vitro by negatively regulating the TEC1 and UME6 genes that are essential for the production of hyphae. Prophylactic provision of Lactobacillus paracasei 28.4 protected C. elegans against candidiasis in vivo. L. paracasei 28.4 has the potential to be employed as an alternative method to control candidiasis.


Asunto(s)
Caenorhabditis elegans/microbiología , Candida albicans/crecimiento & desarrollo , Hifa/crecimiento & desarrollo , Lacticaseibacillus paracasei/fisiología , Modelos Teóricos , Animales , Antibiosis , Candida albicans/genética , Candidiasis/microbiología , Candidiasis/prevención & control , Candidiasis/terapia , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Proteínas Fúngicas/genética , Regulación Fúngica de la Expresión Génica , Genes Fúngicos/genética , Hifa/citología , Lacticaseibacillus paracasei/aislamiento & purificación , Interacciones Microbianas , Probióticos , Proteínas Represoras/genética , Factores de Transcripción/genética
7.
Biofouling ; 34(2): 212-225, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29380647

RESUMEN

This study isolated Lactobacillus strains from caries-free subjects and evaluated the inhibitory effects directly on three strains of C. albicans, two clinical strains and one reference strain. Thirty Lactobacillus strains were isolated and evaluated for antimicrobial activity against in vitro C. albicans biofilms. L. paracasei 28.4, L. rhamnosus 5.2 and L. fermentum 20.4 isolates exhibited the most significant inhibitory activity against C. albicans. Co-incubation between these microorganisms resulted in deterrence of biofilm development and retardation of hyphal formation. The hindrance of biofilm development was characterized by the downregulated expression of C. albicans biofilm-specific genes (ALS3, HWP1, EFG1 and CPH1). L. paracasei 28.4, L. rhamnosus 5.2 and L. fermentum 20.4 demonstrated the ability to exert antifungal activity through the inhibition of C. albicans biofilms.


Asunto(s)
Antibiosis , Antifúngicos/farmacología , Biopelículas/efectos de los fármacos , Candida albicans/efectos de los fármacos , Candidiasis Bucal/prevención & control , Lactobacillus/fisiología , Probióticos/farmacología , Biopelículas/crecimiento & desarrollo , Candida albicans/genética , Candida albicans/fisiología , Humanos , Hifa/efectos de los fármacos , Hifa/crecimiento & desarrollo
8.
Artículo en Inglés | MEDLINE | ID: mdl-28483966

RESUMEN

The increasing prevalence of antibiotic resistance has created an urgent need for alternative drugs with new mechanisms of action. Antimicrobial peptides (AMPs) are promising candidates that could address the spread of multidrug-resistant bacteria, either alone or in combination with conventional antibiotics. We studied the antimicrobial efficacy and bactericidal mechanism of cecropin A2, a 36-residue α-helical cationic peptide derived from Aedes aegypti cecropin A, focusing on the common pathogen Pseudomonas aeruginosa The peptide showed little hemolytic activity and toxicity toward mammalian cells, and the MICs against most clinical P. aeruginosa isolates were 32 to 64 µg/ml, and its MICs versus other Gram-negative bacteria were 2 to 32 µg/ml. Importantly, cecropin A2 demonstrated synergistic activity against P. aeruginosa when combined with tetracycline, reducing the MICs of both agents by 8-fold. The combination was also effective in vivo in the P. aeruginosa/Galleria mellonella model (P < 0.001). We found that cecropin A2 bound to P. aeruginosa lipopolysaccharides, permeabilized the membrane, and interacted with the bacterial genomic DNA, thus facilitating the translocation of tetracycline into the cytoplasm. In summary, the combination of cecropin A2 and tetracycline demonstrated synergistic antibacterial activity against P. aeruginosain vitro and in vivo, offering an alternative approach for the treatment of P. aeruginosa infections.


Asunto(s)
Antibacterianos/farmacología , Cecropinas/farmacología , Pseudomonas aeruginosa/efectos de los fármacos , Tetraciclina/farmacología , Aedes , Animales , Sinergismo Farmacológico , Eritrocitos/efectos de los fármacos , Eritrocitos/metabolismo , Bacterias Gramnegativas/efectos de los fármacos , Células Hep G2 , Humanos , Pruebas de Sensibilidad Microbiana , Pseudomonas aeruginosa/genética
9.
Artículo en Inglés | MEDLINE | ID: mdl-28533240

RESUMEN

Human cryptococcosis can occur as a primary or opportunistic infection and develops as an acute, subacute, or chronic systemic infection involving different organs of the host. Given the limited therapeutic options and the occasional resistance to fluconazole, there is a need to develop novel drugs for the treatment of cryptococcosis. In this report, we describe promising thiazole compounds 1, 2, 3, and 4 and explore their possible modes of action against Cryptococcus To this end, we show evidence of interference in the Cryptococcus antioxidant system. The tested compounds exhibited MICs ranging from 0.25 to 2 µg/ml against Cryptococcus neoformans strains H99 and KN99α. Interestingly, the knockout strains for Cu oxidase and sarcosine oxidase were resistant to thiazoles. MIC values of thiazole compounds 1, 2, and 4 against these mutants were higher than for the parental strain. After the treatment of C. neoformans ATCC 24067 (or C. deneoformans) and C. gattii strain L27/01 (or C. deuterogattii) with thiazoles, we verified an increase in intracellular reactive oxygen species (ROS). Also, we verified the synergistic interactions among thiazoles and menadione, which generates superoxides, with fractional inhibitory concentrations (FICs) equal to 0.1874, 0.3024, 0.25, and 0.25 for the thiazole compounds 1, 2, 3, and 4, respectively. In addition, thiazoles exhibited antagonistic interactions with parasulphonatephenyl porphyrinato ferrate III (FeTPPS). Thus, in this work, we showed that the action of these thiazoles is related to an interference with the antioxidant system. These findings suggest that oxidative stress may be primarily related to the accumulation of superoxide radicals.


Asunto(s)
Antifúngicos/farmacología , Criptococosis/tratamiento farmacológico , Cryptococcus gattii/efectos de los fármacos , Cryptococcus neoformans/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Tiazoles/farmacología , Farmacorresistencia Fúngica , Humanos , Pruebas de Sensibilidad Microbiana , Oxidorreductasas/genética , Sarcosina-Oxidasa/genética , Vitamina K 3/metabolismo
10.
Mycopathologia ; 181(1-2): 17-25, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26384671

RESUMEN

The echinocandin family of drugs is well characterized for antifungal function that inhibits ß-D-glucan synthesis. The aim of this work was to study whether micafungin, a member of the echinocandin family, elicits additional activities that prime the host's immune response. We found that in a Galleria mellonella model, prophylactic treatment with micafungin extended the life of Staphylococcus aureus-infected larvae (a pathogen to which the drug demonstrates no direct antimicrobial activity) compared to insects that did not receive micafungin (P < 0.05). The inhibition of pathogens in the G. mellonella infection model was characterized by a 2.43-fold increase in hemocyte density, compared to larvae inoculated with PBS. In a murine model where animals were provided micafungin prophylaxis 3 days prior to macrophage collection, macrophages were found associated with an average 0.9 more fungal cells per macrophage as compared to saline-treated animals. Interestingly, micafungin-stimulated macrophages killed 11.6 ± 6.2 % of fungal cells compared to 3.8 ± 2.4 % of macrophages from saline-treated animals. The prophylactic provision of micafungin prior to Candida albicans infection was characterized by an increase in the proinflammatory cytokines CXCL13 and SPP1 by 11- and 6.9-fold, respectively. In conclusion, micafungin demonstrated the ability to stimulate phagocytic cells and promote an immune response that can inhibit microbial infections.


Asunto(s)
Equinocandinas/administración & dosificación , Equinocandinas/farmacología , Factores Inmunológicos/administración & dosificación , Factores Inmunológicos/farmacología , Lipopéptidos/administración & dosificación , Lipopéptidos/farmacología , Animales , Candida albicans/inmunología , Candida albicans/fisiología , Modelos Animales de Enfermedad , Lepidópteros , Macrófagos/inmunología , Macrófagos/microbiología , Micafungina , Ratones , Viabilidad Microbiana , Infecciones Estafilocócicas/tratamiento farmacológico , Análisis de Supervivencia , Resultado del Tratamiento
11.
Clin Microbiol Rev ; 27(3): 490-526, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24982319

RESUMEN

Invasive fungal infections constitute a serious threat to an ever-growing population of immunocompromised individuals and other individuals at risk. Traditional diagnostic methods, such as histopathology and culture, which are still considered the gold standards, have low sensitivity, which underscores the need for the development of new means of detecting fungal infectious agents. Indeed, novel serologic and molecular techniques have been developed and are currently under clinical evaluation. Tests like the galactomannan antigen test for aspergillosis and the ß-glucan test for invasive Candida spp. and molds, as well as other antigen and antibody tests, for Cryptococcus spp., Pneumocystis spp., and dimorphic fungi, have already been established as important diagnostic approaches and are implemented in routine clinical practice. On the other hand, PCR and other molecular approaches, such as matrix-assisted laser desorption ionization (MALDI) and fluorescence in situ hybridization (FISH), have proved promising in clinical trials but still need to undergo standardization before their clinical use can become widespread. The purpose of this review is to highlight the different diagnostic approaches that are currently utilized or under development for invasive fungal infections and to identify their performance characteristics and the challenges associated with their use.


Asunto(s)
Hongos/fisiología , Técnicas Microbiológicas/métodos , Micosis/diagnóstico , Micosis/microbiología , Animales , Humanos , Técnicas Microbiológicas/normas , Reproducibilidad de los Resultados
12.
J Infect Dis ; 211(2): 298-305, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25114160

RESUMEN

A multi-host approach was followed to screen a library of 1201 signature-tagged deletion strains of Cryptococcus neoformans mutants to identify previously unknown virulence factors. The primary screen was performed using a Caenorhabditis elegans-C. neoformans infection assay. The hits among these strains were reconfirmed as less virulent than the wild type in the insect Galleria mellonella-C. neoformans infection assay. After this 2-stage screen, and to prioritize hits, we performed serial evaluations of the selected strains, using the C. elegans model. All hit strains identified through these studies were validated in a murine model of systemic cryptococcosis. Twelve strains were identified through a stepwise screening assay. Among them, 4 (CSN1201, SRE1, RDI1, and YLR243W) were previously discovered, providing proof of principle for this approach, while the role of the remaining 8 genes (CKS101, CNC5600, YOL003C, CND1850, MLH3, HAP502, MSL5, and CNA2580) were not previously described in cryptococcal virulence. The multi-host approach is an efficient method of studying the pathogenesis of C. neoformans. We used diverse model hosts, C. elegans, G. mellonella, and mice, with physiological differences and identified 12 genes associated with mammalian infection. Our approach may be suitable for large pathogenesis screens.


Asunto(s)
Caenorhabditis elegans/microbiología , Cryptococcus neoformans/patogenicidad , Mariposas Nocturnas/microbiología , Factores de Virulencia/análisis , Animales , Criptococosis/microbiología , Criptococosis/patología , Cryptococcus neoformans/genética , Modelos Animales de Enfermedad , Femenino , Eliminación de Gen , Pruebas Genéticas , Ratones , Factores de Virulencia/genética
13.
Antimicrob Agents Chemother ; 59(10): 6581-7, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26259795

RESUMEN

Candida parapsilosis is the main non-albicans Candida species isolated from patients in Latin America. Mutations in the ERG11 gene and overexpression of membrane transporter proteins have been linked to fluconazole resistance. The aim of this study was to evaluate the molecular mechanisms in fluconazole-resistant strains of C. parapsilosis isolated from critically ill patients. The identities of the nine collected C. parapsilosis isolates at the species level were confirmed through molecular identification with a TaqMan qPCR assay. The clonal origin of the strains was checked by microsatellite typing. The Galleria mellonella infection model was used to confirm in vitro resistance. We assessed the presence of ERG11 mutations, as well as the expression of ERG11 and two additional genes that contribute to antifungal resistance (CDR1 and MDR1), by using real-time quantitative PCR. All of the C. parapsilosis (sensu stricto) isolates tested exhibited fluconazole MICs between 8 and 16 µg/ml. The in vitro data were confirmed by the failure of fluconazole in the treatment of G. mellonella infected with fluconazole-resistant strains of C. parapsilosis. Sequencing of the ERG11 gene revealed a common mutation leading to a Y132F amino acid substitution in all of the isolates, a finding consistent with their clonal origin. After fluconazole exposure, overexpression was noted for ERG11, CDR1, and MDR1 in 9/9, 9/9, and 2/9 strains, respectively. We demonstrated that a combination of molecular mechanisms, including the presence of point mutations in the ERG11 gene, overexpression of ERG11, and genes encoding efflux pumps, are involved in fluconazole resistance in C. parapsilosis.


Asunto(s)
Antifúngicos/farmacología , Candida/efectos de los fármacos , Candida/patogenicidad , Fluconazol/farmacología , Larva/microbiología , Mariposas Nocturnas/microbiología , Animales , Farmacorresistencia Fúngica , Pruebas de Sensibilidad Microbiana
14.
Antimicrob Agents Chemother ; 59(3): 1728-37, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25583713

RESUMEN

The rise of multidrug-resistant Acinetobacter baumannii and a concomitant decrease in antibiotic treatment options warrants a search for new classes of antibacterial agents. We have found that A. baumannii is pathogenic and lethal to the model host organism Caenorhabditis elegans and have exploited this phenomenon to develop an automated, high-throughput, high-content screening assay in liquid culture that can be used to identify novel antibiotics effective against A. baumannii. The screening assay involves coincubating C. elegans with A. baumannii in 384-well plates containing potential antibacterial compounds. At the end of the incubation period, worms are stained with a dye that stains only dead animals, and images are acquired using automated microscopy and then analyzed using an automated image analysis program. This robust assay yields a Z' factor consistently greater than 0.7. In a pilot experiment to test the efficacy of the assay, we screened a small custom library of synthetic antimicrobial peptides (AMPs) that were synthesized using publicly available sequence data and/or transcriptomic data from immune-challenged insects. We identified cecropin A and 14 other cecropin or cecropin-like peptides that were able to enhance C. elegans survival in the presence of A. baumannii. Interestingly, one particular hit, BR003-cecropin A, a cationic peptide synthesized by the mosquito Aedes aegypti, showed antibiotic activity against a panel of Gram-negative bacteria and exhibited a low MIC (5 µg/ml) against A. baumannii. BR003-cecropin A causes membrane permeability in A. baumannii, which could be the underlying mechanism of its lethality.


Asunto(s)
Acinetobacter baumannii/efectos de los fármacos , Caenorhabditis elegans/microbiología , Cecropinas/farmacología , Insectos/metabolismo , Animales , Antibacterianos/farmacología , Modelos Animales de Enfermedad , Farmacorresistencia Bacteriana Múltiple/efectos de los fármacos
15.
Bioorg Med Chem Lett ; 25(22): 5203-7, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26459212

RESUMEN

The emergence of multidrug-resistant bacterial strains has heightened the need for new antimicrobial agents based on novel chemical scaffolds that are able to circumvent current modes of resistance. We recently developed a whole-animal drug-screening methodology in pursuit of this goal and now report the discovery of 3-(phenylsulfonyl)-2-pyrazinecarbonitrile (PSPC) as a novel antibacterial effective against resistant nosocomial pathogens. The minimum inhibitory concentrations (MIC) of PSPC against Staphylococcus aureus and Enterococcus faecium were 4 µg/mL and 8 µg/mL, respectively, whereas the MICs were higher against the Gram-negative bacteria Klebsiella pneumoniae (64 µg/mL), Acinetobacter baumannii (32 µg/mL), Pseudomonas aeruginosa (>64 µg/mL), and Enterobacter spp. (>64 µg/mL). However, co-treatment of PSPC with the efflux pump inhibitor phenylalanine arginyl ß-naphthylamide (PAßN) or with sub-inhibitory concentrations of the lipopeptide antibiotic polymyxin B reduced the MICs of PSPC against the Gram-negative strains by >4-fold. A sulfide analog of PSPC (PSPC-1S) showed no antibacterial activity, whereas the sulfoxide analog (PSPC-6S) showed identical activity as PSPC across all strains, confirming structure-dependent activity for PSPC and suggesting a target-based mechanism of action. PSPC displayed dose dependent toxicity to both Caenorhabditis elegans and HEK-293 mammalian cells, culminating with a survival rate of 16% (100 µg/mL) and 8.5% (64 µg/mL), respectively, at the maximum tested concentration. However, PSPC did not result in hemolysis of erythrocytes, even at a concentration of 64 µg/mL. Together these results support PSPC as a new chemotype suitable for further development of new antibiotics against Gram-positive and Gram-negative bacteria.


Asunto(s)
Antibacterianos/farmacología , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Pirazinas/farmacología , Animales , Caenorhabditis elegans , Dipéptidos/farmacología , Farmacorresistencia Bacteriana , Bacterias Gramnegativas/efectos de los fármacos , Bacterias Grampositivas/efectos de los fármacos , Células HEK293 , Ensayos Analíticos de Alto Rendimiento , Humanos , Pruebas de Sensibilidad Microbiana , Polimixina B/farmacología , Pirazinas/sangre , Ovinos , Vancomicina/farmacología
16.
Antimicrob Agents Chemother ; 57(1): 445-51, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23129051

RESUMEN

The objective of this study was to evaluate whether Candida albicans exhibits altered pathogenicity characteristics following sublethal antimicrobial photodynamic inactivation (APDI) and if such alterations are maintained in the daughter cells. C. albicans was exposed to sublethal APDI by using methylene blue (MB) as a photosensitizer (0.05 mM) combined with a GaAlAs diode laser (λ 660 nm, 75 mW/cm(2), 9 to 27 J/cm(2)). In vitro, we evaluated APDI effects on C. albicans growth, germ tube formation, sensitivity to oxidative and osmotic stress, cell wall integrity, and fluconazole susceptibility. In vivo, we evaluated C. albicans pathogenicity with a mouse model of systemic infection. Animal survival was evaluated daily. Sublethal MB-mediated APDI reduced the growth rate and the ability of C. albicans to form germ tubes compared to untreated cells (P < 0.05). Survival of mice systemically infected with C. albicans pretreated with APDI was significantly increased compared to mice infected with untreated yeast (P < 0.05). APDI increased C. albicans sensitivity to sodium dodecyl sulfate, caffeine, and hydrogen peroxide. The MIC for fluconazole for C. albicans was also reduced following sublethal MB-mediated APDI. However, none of those pathogenic parameters was altered in daughter cells of C. albicans submitted to APDI. These data suggest that APDI may inhibit virulence factors and reduce in vivo pathogenicity of C. albicans. The absence of alterations in daughter cells indicates that APDI effects are transitory. The MIC reduction for fluconazole following APDI suggests that this antifungal could be combined with APDI to treat C. albicans infections.


Asunto(s)
Candida albicans/efectos de los fármacos , Candidiasis/tratamiento farmacológico , Azul de Metileno/farmacología , Fármacos Fotosensibilizantes/farmacología , Factores de Virulencia/antagonistas & inhibidores , Animales , Antifúngicos/farmacología , Candida albicans/crecimiento & desarrollo , Candida albicans/patogenicidad , Candida albicans/efectos de la radiación , Candidiasis/microbiología , Candidiasis/mortalidad , Pared Celular/efectos de los fármacos , Pared Celular/efectos de la radiación , Femenino , Fluconazol/farmacología , Patrón de Herencia , Láseres de Semiconductores , Luz , Masculino , Ratones , Ratones Endogámicos BALB C , Presión Osmótica , Estrés Oxidativo , Análisis de Supervivencia , Factores de Virulencia/metabolismo
17.
Front Cell Infect Microbiol ; 13: 1135942, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37313344

RESUMEN

Microbe entry through catheter ports can lead to biofilm accumulation and complications from catheter-related bloodstream infection and ultimately require antimicrobial treatment and catheter replacement. Although strides have been made with microbial prevention by applying standardized antiseptic techniques during catheter implantation, both bacterial and fungal microbes can present health risks to already sick individuals. To reduce microbial adhesion, murine and human catheters were coated with polyurethane and auranofin using a dip coating method and compared to non-coated materials. Upon passage of fluid through the coated material in vitro, flow dynamics were not impacted. The unique antimicrobial properties of the coating material auranofin has shown inhibitory activity against bacteria such as Staphylococcus aureus and fungi such as Candida albicans. Auranofin coating on catheters at 10mg/mL reduced C. albicans accumulation in vitro from 2.0 x 108 to 7.8 x 105 CFU for mouse catheters and from 1.6 x 107 to 2.8 x 106 for human catheters, showing an impact to mature biofilms. Assessment of a dual microbe biofilm on auranofin-coated catheters resulted in a 2-log reduction in S. aureus and a 3-log reduction in C. albicans compared to uncoated catheters. In vivo assessment in a murine subcutaneous model demonstrated that catheters coated with 10 mg/mL auranofin reduced independent S. aureus and C. albicans accumulation by 4-log and 1-log, respectively, compared to non-coated catheters. In conclusion, the auranofin-coated catheters demonstrate proficiency at inhibiting multiple pathogens by decreasing S. aureus and C. albicans biofilm accumulation.


Asunto(s)
Auranofina , Staphylococcus aureus , Humanos , Animales , Ratones , Auranofina/farmacología , Bacterias , Biopelículas , Candida albicans , Catéteres
18.
Clin Infect Dis ; 54(9): 1322-31, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22362884

RESUMEN

Currently accepted fungal diagnostic techniques, such as culture, biopsy, and serology, lack rapidity and efficiency. Newer diagnostic methods, such as polymerase chain reaction (PCR)-based assays, have the potential to improve fungal diagnostics in a faster, more sensitive, and specific manner. Preliminary data indicate that, when PCR-based fungal diagnostic assays guide antifungal therapy, they may lower patient mortality and decrease unnecessary antifungal treatment, improving treatment-associated costs and avoiding toxicity. Moreover, newer PCR techniques can identify antifungal resistance DNA loci, but the clinical correlation between those loci and clinical failure has to be studied further. In addition, future studies need to focus on the implementation of PCR techniques in clinical decision making and on combining them with other diagnostic tests. A consensus on the standardization of PCR techniques, along with validation from large prospective studies, is necessary to allow widespread adoption of these assays.


Asunto(s)
Hongos/genética , Micosis/diagnóstico , Reacción en Cadena de la Polimerasa/métodos , Antifúngicos/uso terapéutico , ADN de Hongos/genética , Farmacorresistencia Fúngica/genética , Humanos , Micosis/tratamiento farmacológico , Reacción en Cadena de la Polimerasa/normas , Sensibilidad y Especificidad
19.
Med Mycol ; 50(5): 488-96, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22225407

RESUMEN

Fusariosis is an emerging infectious complication of immune deficiency, but models to study this infection are lacking. The use of the soil nematode Caenorhabditis elegans as a model host to study the pathogenesis of Fusarium spp. was investigated. We observed that Fusarium conidia consumed by C. elegans can cause a lethal infection and result in more than 90% killing of the host within 120 hours, and the nematode had a significantly longer survival when challenged with Fusarium proliferatum compared to other species. Interestingly, mycelium production appears to be a major contributor in nematode killing in this model system, and C. elegans mutant strains with the immune response genes, tir-1 (encoding a protein containing a TIR domain that functions upstream of PMK-1) and pmk-1 (the homolog of the mammalian p38 MAPK) lived significantly shorter when challenged with Fusarium compared to the wild type strain. Furthermore, we used the C. elegans model to assess the efficacy and toxicity of various compounds against Fusarium. We demonstrated that amphotericin B, voriconazole, mancozeb, and phenyl mercury acetate significantly prolonged the survival of Fusarium-infected C. elegans, although mancozeb was toxic at higher concentrations. In conclusion, we describe a new model system for the study of Fusarium pathogenesis and evolutionarily preserved host responses to this important fungal pathogen.


Asunto(s)
Caenorhabditis elegans/microbiología , Modelos Animales de Enfermedad , Fusariosis/inmunología , Fusarium/patogenicidad , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Micelio/metabolismo , Animales , Antifúngicos/farmacología , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Fusariosis/microbiología , Fusarium/clasificación , Fusarium/efectos de los fármacos , Interacciones Huésped-Patógeno , Humanos , Proteínas Quinasas Activadas por Mitógenos/genética , Transducción de Señal/inmunología
20.
Proc Natl Acad Sci U S A ; 105(38): 14585-90, 2008 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-18794525

RESUMEN

Prokaryote-eukaryote interactions are ubiquitous and have important medical and environmental significance. Despite this, a paucity of data exists on the mechanisms and pathogenic consequences of bacterial-fungal encounters within a living host. We used the nematode Caenorhabditis elegans as a substitute host to study the interactions between two ecologically related and clinically troublesome pathogens, the prokaryote, Acinetobacter baumannii, and the eukaryote, Candida albicans. After co-infecting C. elegans with these organisms, we observed that A. baumannii inhibits filamentation, a key virulence determinant of C. albicans. This antagonistic, cross-kingdom interaction led to attenuated virulence of C. albicans, as determined by improved nematode survival when infected with both pathogens. In vitro coinfection assays in planktonic and biofilm environments supported the inhibitory effects of A. baumannii toward C. albicans, further showing a predilection of A. baumannii for C. albicans filaments. Interestingly, we demonstrate a likely evolutionary defense by C. albicans against A. baumannii, whereby C. albicans inhibits A. baumannii growth once a quorum develops. This counteroffensive is at least partly mediated by the C. albicans quorum-sensing molecule farnesol. We used the C. elegans-A. baumannii-C. albicans coinfection model to screen an A. baumannii mutant library, leading to the identification of several mutants attenuated in their inhibitory activity toward C. albicans. These findings present an extension to the current paradigm of studying monomicrobial pathogenesis in C. elegans and by use of genetic manipulation, provides a whole-animal model system to investigate the complex dynamics of a polymicrobial infection.


Asunto(s)
Acinetobacter baumannii/fisiología , Caenorhabditis elegans/microbiología , Candida albicans/fisiología , Acinetobacter baumannii/efectos de los fármacos , Acinetobacter baumannii/genética , Acinetobacter baumannii/patogenicidad , Animales , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/farmacología , Biopelículas/crecimiento & desarrollo , Candida albicans/efectos de los fármacos , Candida albicans/patogenicidad , Farnesol/farmacología , Pruebas Genéticas , Mutación , Virulencia/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA