Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Viruses ; 15(2)2023 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-36851708

RESUMEN

The French Phage Network (Phages.fr) has continuously grown since its foundation, eight years ago. The annual conference, held at the Institut Pasteur in Paris, attracted 164 participants from the 11th to the 13th of October 2022. Researchers from academic laboratories, hospitals and private companies shared their ongoing projects and breakthroughs in the very institute where Felix d'Hérelle developed phage therapy over a century ago. The conference was divided into four thematic sessions, each opened by a keynote lecture: "Interaction between phages, mobile genetic elements and bacterial immune system," "Ecology and evolution of phage-bacteria interactions," "Molecular interplay between phages and their hosts" and "Therapeutic and biotechnological applications of phages." A total of 32 talks and 33 posters were presented during the conference.


Asunto(s)
Bacteriófagos , Humanos , Bacteriófagos/genética , Biotecnología , Ecología , Hospitales , Laboratorios
2.
J Extracell Biol ; 2(2): e75, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38938523

RESUMEN

Research on extracellular vesicles (EVs) and bacteriophages (phages) has been steadily expanding over the past decades as many of their roles in medicine, biology, and ecosystems have been unveiled. Such interest has brought about the need for new tools to quantify and determine the sizes of these biological nanoparticles. A new device based on interferometric light microscopy (ILM), the Videodrop, was recently developed for this purpose. Here, we compared this new device to two nanoparticle tracking analysis (NTA) devices, the NanoSight and the ZetaView, for the analysis of EVs and phages. We used EVs isolated from bacteria, fecal samples, bovine milk and human cells, and phages of various sizes and shape, ranging from 30 to 120 nm of diameter. While NTA instruments correctly enumerated most phages, the Videodrop detected only the largest one, indicating a lower sensitivity threshold compared to the NTA devices. Nevertheless, the performance of the Videodrop compared favourably to that of the NTA devices for the determination of the concentration of eukaryotic EV samples. The NanoSight instrument provided the most precise size distributions but the Videodrop was by far the most time-saving device, making it worthy of consideration for studies conducted on a large number of samples composed of nanoparticles larger than 90 nm.

3.
Sci Rep ; 12(1): 9440, 2022 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-35676403

RESUMEN

In the gut ecosystem, microorganisms regulate group behaviour and interplay with the host via a molecular system called quorum sensing (QS). The QS molecule 3-oxo-C12:2-HSL, first identified in human gut microbiota, exerts anti-inflammatory effects and could play a role in inflammatory bowel diseases where dysbiosis has been described. Our aim was to identify which signalling pathways are involved in this effect. We observed that 3-oxo-C12:2-HSL decreases expression of pro-inflammatory cytokines such as Interleukine-1ß (- 35%) and Tumor Necrosis Factor-α (TNFα) (- 40%) by stimulated immune RAW264.7 cells and decreased TNF secretion by stimulated PBMC in a dose-dependent manner, between 25 to 100 µM. Transcriptomic analysis of RAW264.7 cells exposed to 3-oxo-C12:2-HSL, in a pro-inflammatory context, highlighted JAK-STAT, NF-κB and TFN signalling pathways and we confirmed that 3-oxo-C12:2-HSL inhibited JAK1 and STAT1 phosphorylation. We also showed through a screening assay that 3-oxo-C12:2-HSL interacted with several human bitter taste receptors. Its anti-inflammatory effect involved TAS2R38 as shown by pharmacologic inhibition and led to an increase in intracellular calcium levels. We thus unravelled the involvement of several cellular pathways in the anti-inflammatory effects exerted by the QS molecule 3-oxo-C12:2-HSL.


Asunto(s)
Microbioma Gastrointestinal , Percepción de Quorum , 4-Butirolactona/metabolismo , Antiinflamatorios/metabolismo , Ecosistema , Homoserina/metabolismo , Humanos , Leucocitos Mononucleares/metabolismo , Pseudomonas aeruginosa/fisiología , Gusto
4.
Cell Host Microbe ; 30(4): 431-434, 2022 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-35421337

RESUMEN

Five years ago, my first study on the mechanisms that govern the coexistence of intestinal bacteria and bacteriophages was published in Cell Host & Microbe. In this commentary, I use the following evolutionary steps of my career to discuss the larger frame of bacteriophage biology in gut health and disease.


Asunto(s)
Bacteriófagos , Microbioma Gastrointestinal , Bacterias , Bacteriófagos/genética
5.
Cell Host Microbe ; 30(4): 556-569.e5, 2022 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-35421351

RESUMEN

Abundance and diversity of bacteria and their viral predators, bacteriophages (phages), in the digestive tract are associated with human health. Particularly intriguing is the long-term coexistence of these two antagonistic populations. We performed genome-wide RNA sequencing on a human enteroaggregative Escherichia coli isolate to identify genes differentially expressed between in vitro conditions and in murine intestines. We experimentally demonstrated that four of these differentially expressed genes modified the interactions between E. coli and three virulent phages by either increasing or decreasing its susceptibility/resistance pattern and also by interfering with biofilm formation. Therefore, the regulation of bacterial genes expression during the colonization of the digestive tract influences the coexistence of phages and bacteria, highlighting the intricacy of tripartite relationships between phages, bacteria, and the animal host in intestinal homeostasis.


Asunto(s)
Bacteriófagos , Animales , Bacterias/genética , Bacteriófagos/fisiología , Escherichia coli/genética , Expresión Génica , Genes Bacterianos , Ratones
6.
Curr Opin Virol ; 51: 141-148, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34700287

RESUMEN

Mucosal surfaces in contact with the environment host specific microbiota. The intestinal tract harbours the most abundant and diverse bacterial and viral populations interacting with each other as well as with the host. Viruses of the microbiota are important components of this ecosystem, as shown by viral alterations associated with various pathologies. However, practical and ethical constraints limit functional studies of the virome in humans, making animal models invaluable experimental tools to understand its impact on intestinal physiology. In this review, we present the recent advances in the study of virome in animal models. We focus on the strategies used to characterise viral changes in disease models and approaches to modulate the microbiota using viruses. In reviewing the interplay between viruses, bacteria, and the animal host, we highlight the potential and limitations of these models in elucidating the role of the virome in determining human health and disease.


Asunto(s)
Interacciones Microbiota-Huesped , Intestinos/virología , Modelos Animales , Viroma , Animales , Modelos Animales de Enfermedad , Humanos
7.
Science ; 369(6506): 936-942, 2020 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-32820119

RESUMEN

Intestinal microbiota have been proposed to induce commensal-specific memory T cells that cross-react with tumor-associated antigens. We identified major histocompatibility complex (MHC) class I-binding epitopes in the tail length tape measure protein (TMP) of a prophage found in the genome of the bacteriophage Enterococcus hirae Mice bearing E. hirae harboring this prophage mounted a TMP-specific H-2Kb-restricted CD8+ T lymphocyte response upon immunotherapy with cyclophosphamide or anti-PD-1 antibodies. Administration of bacterial strains engineered to express the TMP epitope improved immunotherapy in mice. In renal and lung cancer patients, the presence of the enterococcal prophage in stools and expression of a TMP-cross-reactive antigen by tumors correlated with long-term benefit of PD-1 blockade therapy. In melanoma patients, T cell clones recognizing naturally processed cancer antigens that are cross-reactive with microbial peptides were detected.


Asunto(s)
Antígenos de Neoplasias/inmunología , Bacteriófagos/inmunología , Enterococcus hirae/virología , Microbioma Gastrointestinal/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Inmunoterapia/métodos , Neoplasias/terapia , Proteínas de la Cola de los Virus/inmunología , Animales , Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos Alquilantes/uso terapéutico , Linfocitos T CD8-positivos/inmunología , Reacciones Cruzadas , Ciclofosfamida/uso terapéutico , Epítopos/inmunología , Heces/virología , Antígenos H-2/inmunología , Humanos , Ratones , Neoplasias/dietoterapia , Neoplasias/inmunología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/inmunología , Proteínas de la Cola de los Virus/uso terapéutico
8.
Cell Host Microbe ; 28(3): 390-401.e5, 2020 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-32615090

RESUMEN

The ecological dynamics underlying the coexistence between antagonistic populations of bacteria and their viruses, bacteriophages (phages), in the mammalian gut microbiota remain poorly understood. We challenged a murine synthetic bacterial community with phages to study the factors allowing phages-bacteria coexistence. Coexistence was not dependent on the development of phage-resistant clones nor on the ability of phages to extend their host range. Instead, our data suggest that phage-inaccessible sites in the mucosa serve as a spatial refuge for bacteria. From there, bacteria disseminate in the gut lumen where they are predated by luminal phages fostering the presence of intestinal phage populations. The heterogeneous biogeography of microbes contributes to the long-term coexistence of phages with phage-susceptible bacteria. This observation could explain the persistence of intestinal phages in humans as well as the low efficiency of oral phage therapy against enteric pathogens in animal models and clinical trials.


Asunto(s)
Bacterias/crecimiento & desarrollo , Bacterias/virología , Bacteriófagos/fisiología , Escherichia coli/crecimiento & desarrollo , Escherichia coli/virología , Tracto Gastrointestinal/microbiología , Membrana Mucosa/microbiología , Animales , Ecosistema , Heces/microbiología , Femenino , Microbioma Gastrointestinal , Vida Libre de Gérmenes , Masculino , Ratones , Ratones Endogámicos C57BL , Interacciones Microbianas , Modelos Animales
9.
Viruses ; 11(5)2019 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-31058805

RESUMEN

Klebsiella pneumoniae is a bacterial pathogen of high public health importance. Its polysaccharide capsule is highly variable but only a few capsular types are associated with emerging pathogenic sublineages. The aim of this work is to isolate and characterize new lytic bacteriophages and assess their potential to control infections by the ST23 and ST258 K. pneumoniae sublineages using a Galleria mellonella larvae model. Three selected bacteriophages, targeting lineages ST258 (bacteriophages vB_KpnP_KL106-ULIP47 and vB_KpnP_KL106-ULIP54) and ST23 (bacteriophage vB_KpnP_K1-ULIP33), display specificity for capsular types KL106 and K1, respectively. These podoviruses belong to the Autographivirinae subfamily and their genomes are devoid of lysogeny or toxin-associated genes. In a G. mellonella larvae model, a mortality rate of 70% was observed upon infection by K. pneumoniae ST258 and ST23. This number was reduced to 20% upon treatment with bacteriophages at a multiplicity of infection of 10. This work increases the number of characterized bacteriophages infecting K. pneumoniae and provides information regarding genome sequence and efficacy during preclinical phage therapy against two prominent sublineages of this bacterial species.


Asunto(s)
Bacteriófagos/fisiología , Infecciones por Klebsiella/microbiología , Klebsiella pneumoniae/virología , Animales , Modelos Animales de Enfermedad , Genoma Viral , Genómica/métodos , Infecciones por Klebsiella/mortalidad , Infecciones por Klebsiella/terapia , Larva , Mariposas Nocturnas/microbiología , Terapia de Fagos
10.
Cell Host Microbe ; 25(2): 210-218, 2019 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-30763535

RESUMEN

The intestinal microbiota is intimately linked to human health. Decoding the mechanisms underlying its stability in healthy subjects should uncover causes of microbiota-associated diseases and pave the way for treatment. Bacteria and bacteriophages (phages) are the most abundant biological entities in the gastrointestinal tract, where their coexistence is dynamic and affixed. Phages drive and maintain bacterial diversity by perpetuating the coevolutionary interactions with their microbial prey. This review brings together recent in silico, in vitro, and in vivo work dissecting the complexity of phage-bacteria interactions in the intestinal microbiota, including coevolution perspectives. We define the types of dynamics encountered in the gastrointestinal tract and the parameters that affect their outcome. The impact of intestinal physiology on phage-bacterial coevolution is analyzed in the light of its potential contribution to the relationship between the microbiota and human health.


Asunto(s)
Bacterias/virología , Bacteriófagos/fisiología , Tracto Gastrointestinal/microbiología , Interacciones Microbianas , Animales , Coevolución Biológica , Microbioma Gastrointestinal/fisiología , Humanos , Ratones , Organismos Libres de Patógenos Específicos
11.
Gut Microbes ; 10(1): 92-99, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-29913091

RESUMEN

Viruses that infect bacteria, or bacteriophages, are among the most abundant entities in the gut microbiome. However, their role and the mechanisms by which they infect bacteria in the intestinal tract remain poorly understood. We recently reported that intestinal bacteria are an evolutionary force, driving the expansion of the bacteriophage host range by boosting the genetic variability of these viruses. Here, we expand these observations by studying antagonistic bacteriophage-bacteria coevolution dynamics and revealing that bacterial genetic variability is also increased under the pressure of bacteriophage predation. We propose a model showing how the expansion of bacteriophage-bacteria infection networks is relative to the opportunities for coevolution encountered in the intestinal tract. Our data suggest that predator-prey dynamics are perpetuated and differentiated in parallel, to generate and maintain intestinal microbial diversity and equilibrium.


Asunto(s)
Bacterias/virología , Bacteriófagos/fisiología , Coevolución Biológica , Microbioma Gastrointestinal/fisiología , Adaptación Fisiológica , Animales , Antibiosis/genética , Bacterias/genética , Bacteriófagos/genética , Biodiversidad , Microbioma Gastrointestinal/genética , Variación Genética , Humanos , Intestinos/microbiología , Intestinos/virología , Modelos Biológicos
12.
Viruses ; 10(6)2018 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-29914064

RESUMEN

Phage therapy is based on a simple concept: the use of a virus (bacteriophage) that is capable of killing specific pathogenic bacteria to treat bacterial infections. Since the pioneering work of Félix d’Herelle, bacteriophages (phages) isolated in vitro have been shown to be of therapeutic value. Over decades of study, a large number of rather complex mechanisms that are used by phages to hijack bacterial resources and to produce their progeny have been deciphered. While these mechanisms have been identified and have been studied under optimal conditions in vitro, much less is known about the requirements for successful viral infections in relevant natural conditions. This is particularly true in the context of phage therapy. Here, we highlight the parameters affecting phage replication in both in vitro and in vivo environments, focusing, in particular, on the mammalian digestive tract. We propose avenues for increasing the knowledge-guided implementation of phages as therapeutic tools.


Asunto(s)
Bacterias/virología , Bacteriófagos/crecimiento & desarrollo , Interacciones Huésped-Parásitos , Animales , Tracto Gastrointestinal/microbiología , Tracto Gastrointestinal/virología , Humanos , Mamíferos
13.
Cell Host Microbe ; 22(6): 801-808.e3, 2017 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-29174401

RESUMEN

The intestinal microbiota and human health are intimately linked, but interactions between bacteria and bacteriophages in the context of the mammalian intestine remain largely unexplored. We used comparative population genomics to study a tripartite network consisting of a virulent bacteriophage, its bacterial host, and a phage-insensitive bacterial strain both in vitro and within the murine gut. The bacteriophage adapted to infect the insensitive strain when the three partners co-existed in the gut of conventional mice, but not in dixenic mice or in planktonic cultures. The molecular changes associated with modifications in the bacteriophage host spectrum included single amino acid substitutions and an unusual homologous intragenomic recombination event within the genome of the bacteriophage. An intermediate bacterial host isolated from the murine microbiota mediated bacteriophage adaptation. Our data indicate that by offering access to new hosts, the microbiota shifts the genetic diversity of bacteriophages, thereby promoting long-term persistence of bacteriophage populations.


Asunto(s)
Adaptación Biológica , Bacteriófagos/crecimiento & desarrollo , Bacteriófagos/genética , Microbioma Gastrointestinal , Especificidad del Huésped , Animales , Bacteriófagos/clasificación , Ratones
14.
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
15.
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
16.
PLoS One ; 10(8): e0135039, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26313448

RESUMEN

BACKGROUND: Clostridium difficile is the leading cause of antibiotic-associated diarrhoea and pseudo membranous colitis in the developed world. The aim of this study was to explore whether Photodynamic Antimicrobial Chemotherapy (PACT) could be used as a novel approach to treating C. difficile infections. METHODS: PACT utilises the ability of light-activated photosensitisers (PS) to produce reactive oxygen species (ROS) such as free radical species and singlet oxygen, which are lethal to cells. We screened thirteen PS against C. difficile planktonic cells, biofilm and germinating spores in vitro, and cytotoxicity of effective compounds was tested on the colorectal adenocarcinoma cell-line HT-29. RESULTS: Three PS were able to kill 99.9% of bacteria in both aerobic and anaerobic conditions, both in the planktonic state and in a biofilm, after exposure to red laser light (0.2 J/cm2) without harming model colon cells. The applicability of PACT to eradicate C. difficile germinative spores indirectly was also shown, by first inducing germination with the bile salt taurocholate, followed by PACT. CONCLUSION: This innovative and simple approach offers the prospect of a new antimicrobial therapy using light to treat C. difficile infection of the colon.


Asunto(s)
Clostridioides difficile/efectos de los fármacos , Fotoquimioterapia , Fármacos Fotosensibilizantes/farmacología , Biopelículas/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Clorofilidas , Clostridioides difficile/efectos de la radiación , Células HT29 , Humanos , Azul de Metileno/farmacología , Pruebas de Sensibilidad Microbiana , Porfirinas/farmacología
17.
PLoS One ; 7(8): e42894, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22880130

RESUMEN

Hsp12p is considered to be a small heat shock protein and conserved among fungal species. To investigate the expression of this heat shock protein in the fungal pathogen Candida albicans we developed an anti-CaHsp12p antibody. We show that this protein is induced during stationary phase growth and under stress conditions including heat shock, osmotic, oxidative and heavy metal stress. Furthermore, we find that CaHsp12p expression is influenced by the quorum sensing molecule farnesol, the change of CO(2) concentration and pH. Notably we show that the key transcription factor Efg1p acts as a positive regulator of CaHsp12p in response to heat shock and oxidative stress and demonstrate that CaHsp12p expression is additionally modulated by Hog1p and the cAMP-PKA signaling pathway. To study the function of Hsp12p in C. albicans we generated a null mutant, in which all four CaHSP12 genes have been deleted. Phenotypic analysis of the strain shows that CaHSP12 is not essential for stress resistance, morphogenesis or virulence when tested in a Drosophila model of infection. However, when overexpressed, CaHSP12 significantly enhanced cell-cell adhesion, germ tube formation and susceptibility to azole antifungal agents whilst desensitizing C. albicans to the quorum sensing molecule farnesol.


Asunto(s)
Candida albicans/metabolismo , Proteínas Fúngicas/metabolismo , Proteínas de Choque Térmico Pequeñas/metabolismo , Antifúngicos/farmacología , Azoles/farmacología , Candida albicans/efectos de los fármacos , Candida albicans/genética , Candida albicans/crecimiento & desarrollo , Codón Iniciador/genética , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Farnesol/farmacología , Proteínas Fúngicas/genética , Regulación Fúngica de la Expresión Génica/efectos de los fármacos , Genes Fúngicos/genética , Proteínas de Choque Térmico Pequeñas/genética , Respuesta al Choque Térmico/efectos de los fármacos , Respuesta al Choque Térmico/genética , Mutación/genética , Presión Osmótica/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/genética , Polienos/farmacología , Percepción de Quorum/efectos de los fármacos , Percepción de Quorum/genética , Saccharomyces cerevisiae/efectos de los fármacos , Saccharomyces cerevisiae/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Especificidad de la Especie , Estrés Fisiológico/efectos de los fármacos , Estrés Fisiológico/genética , Virulencia/efectos de los fármacos , Virulencia/genética
18.
Eukaryot Cell ; 10(8): 1034-42, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21666074

RESUMEN

Living as a commensal, Candida albicans must adapt and respond to environmental cues generated by the mammalian host and by microbes comprising the natural flora. These signals have opposing effects on C. albicans, with host cues promoting the yeast-to-hyphal transition and bacteria-derived quorum-sensing molecules inhibiting hyphal development. Hyphal development is regulated through modulation of the cyclic AMP (cAMP)/protein kinase A (PKA) signaling pathway, and it has been postulated that quorum-sensing molecules can affect filamentation by inhibiting the cAMP pathway. Here, we show that both farnesol and 3-oxo-C(12)-homoserine lactone, a quorum-sensing molecule secreted by Pseudomonas aeruginosa, block hyphal development by affecting cAMP signaling; they both directly inhibited the activity of the Candida adenylyl cyclase, Cyr1p. In contrast, the 12-carbon alcohol dodecanol appeared to modulate hyphal development and the cAMP signaling pathway without directly affecting the activity of Cyr1p. Instead, we show that dodecanol exerted its effects through a mechanism involving the C. albicans hyphal repressor, Sfl1p. Deletion of SFL1 did not affect the response to farnesol but did interfere with the response to dodecanol. Therefore, quorum sensing in C. albicans is mediated via multiple mechanisms of action. Interestingly, our experiments raise the possibility that the Burkholderia cenocepacia diffusible signal factor, BDSF, also mediates its effects via Sfl1p, suggesting that dodecanol's mode of action, but not farnesol or 3-oxo-C(12)-homoserine lactone, may be used by other quorum-sensing molecules.


Asunto(s)
4-Butirolactona/análogos & derivados , Candida albicans/fisiología , Dodecanol/farmacología , Farnesol/farmacología , Percepción de Quorum , 4-Butirolactona/farmacología , Inhibidores de Adenilato Ciclasa , Adenilil Ciclasas/metabolismo , Candida albicans/efectos de los fármacos , Candida albicans/crecimiento & desarrollo , Núcleo Celular/metabolismo , AMP Cíclico/farmacología , AMP Cíclico/fisiología , Pruebas de Enzimas , Proteínas Fúngicas/antagonistas & inhibidores , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Expresión Génica/efectos de los fármacos , Viabilidad Microbiana/efectos de los fármacos , Estrés Oxidativo , Transducción de Señal , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
19.
PLoS Pathog ; 6(11): e1001193, 2010 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-21124988

RESUMEN

When colonising host-niches or non-animated medical devices, individual cells of the fungal pathogen Candida albicans expand into significant biomasses. Here we show that within such biomasses, fungal metabolically generated CO(2) acts as a communication molecule promoting the switch from yeast to filamentous growth essential for C. albicans pathology. We find that CO(2)-mediated intra-colony signalling involves the adenylyl cyclase protein (Cyr1p), a multi-sensor recently found to coordinate fungal responses to serum and bacterial peptidoglycan. We further identify Lys 1373 as essential for CO(2)/bicarbonate regulation of Cyr1p. Disruption of the CO(2)/bicarbonate receptor-site interferes selectively with C. albicans filamentation within fungal biomasses. Comparisons between the Drosophila melanogaster infection model and the mouse model of disseminated candidiasis, suggest that metabolic CO(2) sensing may be important for initial colonisation and epithelial invasion. Our results reveal the existence of a gaseous Candida signalling pathway and its molecular mechanism and provide insights into an evolutionary conserved CO(2)-signalling system.


Asunto(s)
Adenilil Ciclasas/metabolismo , Candida albicans/patogenicidad , Candidiasis/metabolismo , Dióxido de Carbono/metabolismo , Comunicación Celular/fisiología , Saccharomyces cerevisiae/patogenicidad , Animales , Bicarbonatos/metabolismo , Biomasa , Southern Blotting , Western Blotting , Candidiasis/microbiología , Modelos Animales de Enfermedad , Drosophila melanogaster/fisiología , Femenino , Ratones , Ratones Endogámicos BALB C , Mutagénesis Sitio-Dirigida , Peptidoglicano/farmacología , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Tasa de Supervivencia
20.
FEMS Yeast Res ; 9(7): 990-9, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19845041

RESUMEN

Microorganisms have evolved a complex signature of communication termed quorum sensing (QS), which is based on the exchange and sensing of low molecular- weight signal compounds. The ability to communicate within the microbial population gives the advantage to coordinate a groups behaviour leading to a higher fitness in the environment. The polymorphic fungus Candida albicans is an opportunistic human pathogen able to regulate virulence traits through the production of at least two QS signal molecules: farnesol and tyrosol. The ability to adopt multiple morphotypes and form biofilms on infected surfaces are the most important pathogenic characteristics regulated by QS and are of clinical relevance. In fact, traditional antimicrobial approaches are often ineffective towards these characteristics. Moreover, the intimate association between C. albicans and other pathogens, such as Pseudomonas aeruginosa, increases the complexity of the infection system. This review outlines the current knowledge on fungal QS and fungal-bacterial interactions emphasizing on C. albicans. Further investigations need to concentrate on the molecular mechanisms and the genetic regulation of these phenomena in order to identify putative novel therapeutic options.


Asunto(s)
Candida albicans/fisiología , Pseudomonas aeruginosa/fisiología , Percepción de Quorum , Animales , Humanos , Virulencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...