Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 128
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 120(47): e2307773120, 2023 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-37963246

RESUMEN

The expansion and intensification of livestock production is predicted to promote the emergence of pathogens. As pathogens sometimes jump between species, this can affect the health of humans as well as livestock. Here, we investigate how livestock microbiota can act as a source of these emerging pathogens through analysis of Streptococcus suis, a ubiquitous component of the respiratory microbiota of pigs that is also a major cause of disease on pig farms and an important zoonotic pathogen. Combining molecular dating, phylogeography, and comparative genomic analyses of a large collection of isolates, we find that several pathogenic lineages of S. suis emerged in the 19th and 20th centuries, during an early period of growth in pig farming. These lineages have since spread between countries and continents, mirroring trade in live pigs. They are distinguished by the presence of three genomic islands with putative roles in metabolism and cell adhesion, and an ongoing reduction in genome size, which may reflect their recent shift to a more pathogenic ecology. Reconstructions of the evolutionary histories of these islands reveal constraints on pathogen emergence that could inform control strategies, with pathogenic lineages consistently emerging from one subpopulation of S. suis and acquiring genes through horizontal transfer from other pathogenic lineages. These results shed light on the capacity of the microbiota to rapidly evolve to exploit changes in their host population and suggest that the impact of changes in farming on the pathogenicity and zoonotic potential of S. suis is yet to be fully realized.


Asunto(s)
Infecciones Estreptocócicas , Streptococcus suis , Enfermedades de los Porcinos , Animales , Humanos , Porcinos , Infecciones Estreptocócicas/veterinaria , Granjas , Enfermedades de los Porcinos/epidemiología , Virulencia/genética , Streptococcus suis/genética , Ganado
2.
PLoS Pathog ; 19(12): e1011345, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38060591

RESUMEN

The quorum sensing two-component system (TCS) QseBC has been linked to virulence, motility and metabolism regulation in multiple Gram-negative pathogens, including Enterohaemorrhagic Escherichia coli (EHEC), Uropathogenic E. coli (UPEC) and Salmonella enterica. In EHEC, the sensor histidine kinase (HK) QseC detects the quorum sensing signalling molecule AI-3 and also acts as an adrenergic sensor binding host epinephrine and norepinephrine. Downstream changes in gene expression are mediated by phosphorylation of its cognate response regulator (RR) QseB, and 'cross-talks' with non-cognate regulators KdpE and QseF to activate motility and virulence. In UPEC, cross-talk between QseBC and TCS PmrAB is crucial in the regulation and phosphorylation of QseB RR that acts as a repressor of multiple pathways, including motility. Here, we investigated QseBC regulation of motility in the atypical Enteropathogenic E. coli (EPEC) strain O125ac:H6, causative agent of persistent diarrhoea in children, and its possible cross-talk with the KdpDE and PmrAB TCS. We showed that in EPEC QseB acts as a repressor of genes involved in motility, virulence and stress response, and in absence of QseC HK, QseB is likely activated by the non-cognate PmrB HK, similarly to UPEC. We show that in absence of QseC, phosphorylated QseB activates its own expression, and is responsible for the low motility phenotypes seen in a QseC deletion mutant. Furthermore, we showed that KdpD HK regulates motility in an independent manner to QseBC and through a third unidentified party different to its own response regulator KdpE. We showed that PmrAB has a role in iron adaptation independent to QseBC. Finally, we showed that QseB is the responsible for activation of colistin and polymyxin B resistance genes while PmrA RR acts by preventing QseB activation of these resistance genes.


Asunto(s)
Escherichia coli Enteropatógena , Proteínas de Escherichia coli , Niño , Humanos , Escherichia coli Enteropatógena/genética , Escherichia coli Enteropatógena/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Colistina , Transducción de Señal , Fosforilación , Regulación Bacteriana de la Expresión Génica , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Proteínas de Unión al ADN/metabolismo
3.
Proc Natl Acad Sci U S A ; 119(30): e2118262119, 2022 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-35858453

RESUMEN

Human infections with methicillin-resistant Staphylococcus aureus (MRSA) are commonly treated with vancomycin, and strains with decreased susceptibility, designated as vancomycin-intermediate S. aureus (VISA), are associated with treatment failure. Here, we profiled the phenotypic, mutational, and transcriptional landscape of 10 VISA strains adapted by laboratory evolution from one common MRSA ancestor, the USA300 strain JE2. Using functional and independent component analysis, we found that: 1) despite the common genetic background and environmental conditions, the mutational landscape diverged between evolved strains and included mutations previously associated with vancomycin resistance (in vraT, graS, vraFG, walKR, and rpoBCD) as well as novel adaptive mutations (SAUSA300_RS04225, ssaA, pitAR, and sagB); 2) the first wave of mutations affected transcriptional regulators and the second affected genes involved in membrane biosynthesis; 3) expression profiles were predominantly strain-specific except for sceD and lukG, which were the only two genes significantly differentially expressed in all clones; 4) three independent virulence systems (φSa3, SaeR, and T7SS) featured as the most transcriptionally perturbed gene sets across clones; 5) there was a striking variation in oxacillin susceptibility across the evolved lineages (from a 10-fold increase to a 63-fold decrease) that also arose in clinical MRSA isolates exposed to vancomycin and correlated with susceptibility to teichoic acid inhibitors; and 6) constitutive expression of the VraR regulon explained cross-susceptibility, while mutations in walK were associated with cross-resistance. Our results show that adaptation to vancomycin involves a surprising breadth of mutational and transcriptional pathways that affect antibiotic susceptibility and possibly the clinical outcome of infections.


Asunto(s)
Antibacterianos , Staphylococcus aureus Resistente a Meticilina , Infecciones Estafilocócicas , Staphylococcus aureus , Resistencia a la Vancomicina , Vancomicina , Antibacterianos/química , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Evolución Molecular , Humanos , Staphylococcus aureus Resistente a Meticilina/metabolismo , Pruebas de Sensibilidad Microbiana , Oxacilina/química , Oxacilina/farmacología , Infecciones Estafilocócicas/tratamiento farmacológico , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/genética , Staphylococcus aureus/patogenicidad , Vancomicina/química , Vancomicina/farmacología , Vancomicina/uso terapéutico , Resistencia a la Vancomicina/genética , Virulencia/genética
4.
BMC Genomics ; 25(1): 173, 2024 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-38350904

RESUMEN

BACKGROUND: There is increasing interest in using intestinal organoids to study complex traits like feed efficiency (FE) and host-microbe interactions. The aim of this study was to investigate differences in the molecular phenotype of organoids derived from pigs divergent for FE as well as their responses to challenge with adherent and invasive Escherichia coli (E. coli). RESULTS: Colon and ileum tissue from low and high FE pigs was used to generate 3D organoids and two dimensional (2D) monolayers of organoid cells for E. coli challenge. Genome-wide gene expression was used to investigate molecular differences between pigs that were phenotypically divergent for FE and to study the difference in gene expression after challenge with E. coli. We showed, (1) minor differences in gene expression of colon organoids from pigs with low and high FE phenotypes, (2) that an E. coli challenge results in a strong innate immune gene response in both colon and ileum organoids, (3) that the immune response seems to be less pronounced in the colon organoids of high FE pigs and (4) a slightly stronger immune response was observed in ileum than in colon organoids. CONCLUSIONS: These findings demonstrate the potential for using organoids to gain insights into complex biological mechanisms such as FE.


Asunto(s)
Escherichia coli , Intestinos , Animales , Porcinos , Escherichia coli/genética , Inmunidad Innata , Perfilación de la Expresión Génica , Organoides
5.
Am J Physiol Gastrointest Liver Physiol ; 326(6): G687-G696, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38591144

RESUMEN

Tryptophan is an essential amino acid transformed by host and gut microbial enzymes into metabolites that regulate mucosal homeostasis through aryl hydrocarbon receptor (AhR) activation. Alteration of tryptophan metabolism has been associated with chronic inflammation; however, whether tryptophan supplementation affects the metabolite repertoire and AhR activation under physiological conditions in humans is unknown. We performed a randomized, double blind, placebo-controlled, crossover study in 20 healthy volunteers. Subjects on a low tryptophan background diet were randomly assigned to a 3-wk l-tryptophan supplementation (3 g/day) or placebo, and after a 2-wk washout switched to opposite interventions. We assessed gastrointestinal and psychological symptoms by validated questionnaires, AhR activation by cell reporter assay, tryptophan metabolites by liquid chromatography and high-resolution mass spectrometry, cytokine production in isolated monocytes by ELISA, and microbiota profile by 16S rRNA Illumina technique. Oral tryptophan supplementation was well tolerated, with no changes in gastrointestinal or psychological scores. Compared with placebo, tryptophan increased AhR activation capacity by duodenal contents, but not by feces. This was paralleled by higher urinary and plasma kynurenine metabolites and indoles. Tryptophan had a modest impact on fecal microbiome profiles and no significant effect on cytokine production. At the doses used in this study, oral tryptophan supplementation in humans induces microbial indole and host kynurenine metabolic pathways in the small intestine, known to be immunomodulatory. The results should prompt tryptophan intervention strategies in inflammatory conditions of the small intestine where the AhR pathway is impaired.NEW & NOTEWORTHY We demonstrate that in healthy subjects, orally administered tryptophan activates microbial indole and host kynurenine pathways in the small intestine, the primary metabolic site for dietary components, and the richest source of immune cells along the gut. This study provides novel insights in how to optimally activate immunomodulatory AhR pathways and indole metabolism in the small intestine, serving as basis for future therapeutic trials using l-tryptophan supplementation in chronic inflammatory conditions affecting the small intestine.


Asunto(s)
Estudios Cruzados , Duodeno , Voluntarios Sanos , Receptores de Hidrocarburo de Aril , Triptófano , Humanos , Triptófano/metabolismo , Triptófano/administración & dosificación , Receptores de Hidrocarburo de Aril/metabolismo , Masculino , Adulto , Femenino , Duodeno/metabolismo , Duodeno/efectos de los fármacos , Método Doble Ciego , Suplementos Dietéticos , Microbioma Gastrointestinal/efectos de los fármacos , Adulto Joven , Administración Oral , Quinurenina/metabolismo , Citocinas/metabolismo , Heces/microbiología , Heces/química , Indoles/farmacología , Indoles/administración & dosificación , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico
6.
Crit Rev Food Sci Nutr ; : 1-15, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38950607

RESUMEN

In recent years, the role of microbial tryptophan (Trp) catabolism in host-microbiota crosstalk has become a major area of scientific interest. Microbiota-derived Trp catabolites positively contribute to intestinal and systemic homeostasis by acting as ligands of aryl hydrocarbon receptor and pregnane X receptor, and as signaling molecules in microbial communities. Accumulating evidence suggests that microbial Trp catabolism could be therapeutic targets in treating human diseases. A number of bacteria and metabolic pathways have been identified to be responsible for the conversion of Trp in the intestine. Interestingly, many Trp-degrading bacteria can benefit from the supplementation of specific dietary fibers and polyphenols, which in turn increase the microbial production of beneficial Trp catabolites. Thus, this review aims to highlight the emerging role of diets and food components, i.e., food matrix, fiber, and polyphenol, in modulating the microbial catabolism of Trp and discuss the opportunities for potential therapeutic interventions via specifically designed diets targeting the Trp-microbiome axis.

7.
Annu Rev Nutr ; 42: 165-200, 2022 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-35697048

RESUMEN

The intestinal barrier is essential in early life to prevent infection, inflammation, and food allergies. It consists of microbiota, a mucus layer, an epithelial layer, and the immune system. Microbial metabolites, the mucus, antimicrobial peptides, and secretory immunoglobulin A (sIgA) protect the intestinal mucosa against infection. The complex interplay between these functionalities of the intestinal barrier is crucial in early life by supporting homeostasis, development of the intestinal immune system, and long-term gut health. Exclusive breastfeeding is highly recommended during the first 6 months. When breastfeeding is not possible, milk-based infant formulas are a safe alternative. Breast milk contains many bioactive components that help to establish the intestinal microbiota and influence the development of the intestinal epithelium and the immune system. Importantly, breastfeeding lowers the risk for intestinal and respiratory tract infections. Here we review all aspects of intestinal barrier function and the nutritional components that impact its functionality in early life, such asmicronutrients, bioactive milk proteins, milk lipids, and human milk oligosaccharides. These components are present in breast milk and can be added to milk-based infant formulas to support gut health and immunity.


Asunto(s)
Microbioma Gastrointestinal , Leche Humana , Lactancia Materna , Femenino , Tracto Gastrointestinal , Humanos , Lactante , Mucosa Intestinal/metabolismo
8.
BMC Microbiol ; 22(1): 224, 2022 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-36163011

RESUMEN

BACKGROUND: The palatine tonsils are part of the mucosal immune system and stimulate immune responses through M cell uptake sampling of antigens and bacteria in the tonsillar crypts. Little is known about the development of the tonsillar microbiota and the factors determining the establishment and proliferation of disease-associated bacteria such as Streptococcus suis. In this study, we assessed tonsillar microbiota development in piglets during the first 5 weeks of life and identified the relative importance of maternal and environmental farm parameters influencing the tonsillar microbiota at different ages. Additionally, we studied the effect sow vaccination with a bacterin against S. suis on microbiota development and S. suis colonisation in their offspring. RESULTS: Amplicon sequencing of the 16S rRNA gene V3-V4 region revealed that a diverse tonsillar microbiota is established shortly after birth, which then gradually changes during the first 5 weeks of life without a large impact of weaning on composition or diversity. We found a strong litter effect, with siblings sharing a more similar microbiota compared to non-sibling piglets. Co-housing in rooms, within which litters were housed in separate pens, also had a large impact on microbiota composition. Sow parity and prepartum S. suis bacterin vaccination of sows had weaker but significant associations with microbiota composition, impacting on the abundance of Streptococcus species before and after weaning. Sex and birthweight had limited impact on the tonsillar microbiota, and none of the measured factors had consistent associations with microbiota diversity. CONCLUSIONS: The piglet tonsillar microbiota is established shortly after birth. While microbiota development is associated with both environmental and maternal parameters, weaning has limited impact on microbiota composition. Intramuscular vaccination of sows pre-partum had a significant effect on the tonsillar microbiota composition of their piglets. These findings provide new insights into the mechanisms shaping the tonsillar microbiota.


Asunto(s)
Microbiota , Tonsila Palatina , Animales , Animales Recién Nacidos , Bacterias/genética , Vacunas Bacterianas , Femenino , Lactancia , Tonsila Palatina/microbiología , Embarazo , ARN Ribosómico 16S/genética , Porcinos , Destete
9.
Vet Res ; 52(1): 43, 2021 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-33691792

RESUMEN

Organoids are self-organizing, self-renewing three-dimensional cellular structures that resemble organs in structure and function. They can be derived from adult stem cells, embryonic stem cells, or induced pluripotent stem cells. They contain most of the relevant cell types with a topology and cell-to-cell interactions resembling that of the in vivo tissue. The widespread and increasing adoption of organoid-based technologies in human biomedical research is testament to their enormous potential in basic, translational- and applied-research. In a similar fashion there appear to be ample possibilities for research applications of organoids from livestock and companion animals. Furthermore, organoids as in vitro models offer a great possibility to reduce the use of experimental animals. Here, we provide an overview of studies on organoids in livestock and companion animal species, with focus on the methods developed for organoids from a variety of tissues/organs from various animal species and on the applications in veterinary research. Current limitations, and ongoing research to address these limitations, are discussed. Further, we elaborate on a number of fields of research in animal nutrition, host-microbe interactions, animal breeding and genomics, and animal biotechnology, in which organoids may have great potential as an in vitro research tool.


Asunto(s)
Crianza de Animales Domésticos/métodos , Técnicas In Vitro/veterinaria , Ganado , Organoides/fisiología , Mascotas , Aves de Corral , Medicina Veterinaria/métodos , Fenómenos Fisiológicos Nutricionales de los Animales , Animales , Biotecnología/métodos , Cruzamiento/métodos , Genómica/métodos , Interacciones Microbiota-Huesped , Técnicas In Vitro/métodos
10.
BMC Genomics ; 20(1): 1028, 2019 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-31888466

RESUMEN

BACKGROUND: The mammalian intestine is a complex biological system that exhibits functional plasticity in its response to diverse stimuli to maintain homeostasis. To improve our understanding of this plasticity, we performed a high-level data integration of 14 whole-genome transcriptomics datasets from samples of intestinal mouse mucosa. We used the tool Centrality based Pathway Analysis (CePa), along with information from the Reactome database. RESULTS: The results show an integrated response of the mouse intestinal mucosa to challenges with agents introduced orally that were expected to perturb homeostasis. We observed that a common set of pathways respond to different stimuli, of which the most reactive was the Regulation of Complement Cascade pathway. Altered expression of the Regulation of Complement Cascade pathway was verified in mouse organoids challenged with different stimuli in vitro. CONCLUSIONS: Results of the integrated transcriptomics analysis and data driven experiment suggest an important role of epithelial production of complement and host complement defence factors in the maintenance of homeostasis.


Asunto(s)
Proteínas del Sistema Complemento/inmunología , Homeostasis , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Transcriptoma , Animales , Activación de Complemento , Biología Computacional/métodos , Perfilación de la Expresión Génica , Ratones , Modelos Biológicos , Anotación de Secuencia Molecular , Transducción de Señal
11.
J Clin Microbiol ; 57(7)2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30944194

RESUMEN

Streptococcus suis is one of the most important zoonotic bacterial pathogens of pigs, causing significant economic losses to the global swine industry. S. suis is also a very successful colonizer of mucosal surfaces, and commensal strains can be found in almost all pig populations worldwide, making detection of the S. suis species in asymptomatic carrier herds of little practical value in predicting the likelihood of future clinical relevance. The value of future molecular tools for surveillance and preventative health management lies in the detection of strains that genetically have increased potential to cause disease in presently healthy animals. Here we describe the use of genome-wide association studies to identify genetic markers associated with the observed clinical phenotypes (i) invasive disease and (ii) asymptomatic carriage on the palatine tonsils of pigs on UK farms. Subsequently, we designed a multiplex PCR to target three genetic markers that differentiated 115 S. suis isolates into disease-associated and non-disease-associated groups, that performed with a sensitivity of 0.91, a specificity of 0.79, a negative predictive value of 0.91, and a positive predictive value of 0.79 in comparison to observed clinical phenotypes. We describe evaluation of our pathotyping tool, using an out-of-sample collection of 50 previously uncharacterized S. suis isolates, in comparison to existing methods used to characterize and subtype S. suis isolates. In doing so, we show our pathotyping approach to be a competitive method to characterize S. suis isolates recovered from pigs on UK farms and one that can easily be updated to incorporate global strain collections.


Asunto(s)
Portador Sano/veterinaria , Infecciones Estreptocócicas/veterinaria , Streptococcus suis/aislamiento & purificación , Streptococcus suis/patogenicidad , Enfermedades de los Porcinos/microbiología , Animales , Portador Sano/microbiología , Inglaterra , Marcadores Genéticos/genética , Genoma Bacteriano/genética , Técnicas de Diagnóstico Molecular , Reacción en Cadena de la Polimerasa Multiplex , Tonsila Palatina/microbiología , Infecciones Estreptocócicas/microbiología , Streptococcus suis/genética , Porcinos , Virulencia/genética , Gales
12.
BMC Microbiol ; 19(1): 33, 2019 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-30736731

RESUMEN

BACKGROUND: Lactobacillus mucosae DPC 6426 has previously demonstrated potentially cardio-protective properties, in the form of dyslipidaemia and hypercholesterolemia correction in an apolipoprotein-E deficient mouse model. This study aims to characterise the manner in which this microbe may modulate host bile pool composition and immune response, in the context of cardiovascular disease. Lactobacillus mucosae DPC 6426 was assessed for bile salt hydrolase activity and specificity. The microbe was compared against several other enteric strains of the same species, as well as a confirmed bile salt hydrolase-active strain, Lactobacillus reuteri APC 2587. RESULTS: Quantitative bile salt hydrolase assays revealed that enzymatic extracts from Lactobacillus reuteri APC 2587 and Lactobacillus mucosae DPC 6426 demonstrate the greatest activity in vitro. Bile acid profiling of porcine and murine bile following incubation with Lactobacillus mucosae DPC 6426 confirmed a preference for hydrolysis of glyco-conjugated bile acids. In addition, the purified exopolysaccharide and secretome of Lactobacillus mucosae DPC 6426 were investigated for immunomodulatory capabilities using RAW264.7 macrophages. Gene expression data revealed that both fractions stimulated increases in interleukin-6 and interleukin-10 gene transcription in the murine macrophages, while the entire secretome was necessary to increase CD206 transcription. Moreover, the exopolysaccharide elicited a dose-dependent increase in nitric oxide and interleukin-10 production from RAW264.7 macrophages, concurrent with increased tumour necrosis factor-α secretion at all doses. CONCLUSIONS: This study indicates that Lactobacillus mucosae DPC 6426 modulates both bile pool composition and immune system tone in a manner which may contribute significantly to the previously identified cardio-protective phenotype.


Asunto(s)
Amidohidrolasas/biosíntesis , Bilis/metabolismo , Inmunomodulación , Lactobacillus/enzimología , Lactobacillus/inmunología , Macrófagos/inmunología , Animales , Enfermedades Cardiovasculares/inmunología , Enfermedades Cardiovasculares/microbiología , Glicosiltransferasas/metabolismo , Hidrólisis , Interleucina-10/metabolismo , Interleucina-6/metabolismo , Limosilactobacillus reuteri/enzimología , Lectinas Tipo C/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/microbiología , Receptor de Manosa , Lectinas de Unión a Manosa/metabolismo , Ratones , Óxido Nítrico/metabolismo , Polisacáridos Bacterianos/farmacología , Células RAW 264.7 , Receptores de Superficie Celular/metabolismo , Porcinos , Factor de Necrosis Tumoral alfa/metabolismo
13.
J Immunol ; 199(4): 1418-1428, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28710255

RESUMEN

Activation of the immune system needs to be tightly regulated to provide protection against infections and, at the same time, to prevent excessive inflammation to limit collateral damage to the host. This tight regulation includes regulating the activation of TLRs, which are key players in the recognition of invading microbes. A group of short cationic antimicrobial peptides, called cathelicidins, have previously been shown to modulate TLR activation by synthetic or purified TLR ligands and may play an important role in the regulation of inflammation during infections. However, little is known about how these cathelicidins affect TLR activation in the context of complete and viable bacteria. In this article, we show that chicken cathelicidin-2 kills Escherichia coli in an immunogenically silent fashion. Our results show that chicken cathelicidin-2 kills E. coli by permeabilizing the bacterial inner membrane and subsequently binds the outer membrane-derived lipoproteins and LPS to inhibit TLR2 and TLR4 activation, respectively. In addition, other cathelicidins, including human, mouse, pig, and dog cathelicidins, which lack antimicrobial activity under cell culture conditions, only inhibit macrophage activation by nonviable E. coli In total, this study shows that cathelicidins do not affect immune activation by viable bacteria and only inhibit inflammation when bacterial viability is lost. Therefore, cathelicidins provide a novel mechanism by which the immune system can discriminate between viable and nonviable Gram-negative bacteria to tune the immune response, thereby limiting collateral damage to the host and the risk for sepsis.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/fisiología , Proteínas Sanguíneas/fisiología , Escherichia coli/inmunología , Bacterias Gramnegativas/inmunología , Activación de Macrófagos , Viabilidad Microbiana , Precursores de Proteínas/fisiología , Receptor Toll-Like 2/inmunología , Receptor Toll-Like 4/inmunología , Animales , Proteínas Sanguíneas/aislamiento & purificación , Proteínas Sanguíneas/metabolismo , Catelicidinas/fisiología , Pollos/inmunología , Perros , Bacterias Gramnegativas/fisiología , Humanos , Inflamación/inmunología , Ratones , Precursores de Proteínas/aislamiento & purificación , Precursores de Proteínas/metabolismo , Porcinos/inmunología
14.
Anaerobe ; 56: 98-101, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30794874

RESUMEN

The toll-like receptors involved in recognition of the exopolysaccharide produced by two isogenic, ropy and non-ropy, Bifidobacterium animalis subsp. lactis strains were investigated. Both strains interact with human embryonic kidney (HEK)-293 cells via TLR2, whereas purified EPSs specifically stimulate TLR4 regardless their molar mass.


Asunto(s)
Bifidobacterium animalis/metabolismo , Células Epiteliales/metabolismo , Polisacáridos Bacterianos/metabolismo , Receptor Toll-Like 4/metabolismo , Línea Celular , Humanos , Unión Proteica
15.
Am J Physiol Gastrointest Liver Physiol ; 312(3): G171-G193, 2017 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-27908847

RESUMEN

The gut barrier plays a crucial role by spatially compartmentalizing bacteria to the lumen through the production of secreted mucus and is fortified by the production of secretory IgA (sIgA) and antimicrobial peptides and proteins. With the exception of sIgA, expression of these protective barrier factors is largely controlled by innate immune recognition of microbial molecular ligands. Several specialized adaptations and checkpoints are operating in the mucosa to scale the immune response according to the threat and prevent overreaction to the trillions of symbionts inhabiting the human intestine. A healthy microbiota plays a key role influencing epithelial barrier functions through the production of short-chain fatty acids (SCFAs) and interactions with innate pattern recognition receptors in the mucosa, driving the steady-state expression of mucus and antimicrobial factors. However, perturbation of gut barrier homeostasis can lead to increased inflammatory signaling, increased epithelial permeability, and dysbiosis of the microbiota, which are recognized to play a role in the pathophysiology of a variety of gastrointestinal disorders. Additionally, gut-brain signaling may be affected by prolonged mucosal immune activation, leading to increased afferent sensory signaling and abdominal symptoms. In turn, neuronal mechanisms can affect the intestinal barrier partly by activation of the hypothalamus-pituitary-adrenal axis and both mast cell-dependent and mast cell-independent mechanisms. The modulation of gut barrier function through nutritional interventions, including strategies to manipulate the microbiota, is considered a relevant target for novel therapeutic and preventive treatments against a range of diseases. Several biomarkers have been used to measure gut permeability and loss of barrier integrity in intestinal diseases, but there remains a need to explore their use in assessing the effect of nutritional factors on gut barrier function. Future studies should aim to establish normal ranges of available biomarkers and their predictive value for gut health in human cohorts.


Asunto(s)
Enfermedades Gastrointestinales/fisiopatología , Tracto Gastrointestinal/fisiología , Homeostasis/fisiología , Microbiota/fisiología , Animales , Enfermedades Gastrointestinales/microbiología , Tracto Gastrointestinal/microbiología , Humanos
16.
Br J Nutr ; 117(1): 93-107, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-28102115

RESUMEN

Intestinal barrier integrity is a prerequisite for homeostasis of mucosal function, which is balanced to maximise absorptive capacity, while maintaining efficient defensive reactions against chemical and microbial challenges. Evidence is mounting that disruption of epithelial barrier integrity is one of the major aetiological factors associated with several gastrointestinal diseases, including infection by pathogens, obesity and diabetes, necrotising enterocolitis, irritable bowel syndrome and inflammatory bowel disease. The notion that specific probiotic bacterial strains can affect barrier integrity fuelled research in which in vitro cell lines, animal models and clinical trials are used to assess whether probiotics can revert the diseased state back to homeostasis and health. This review catalogues and categorises the lines of evidence available in literature for the role of probiotics in epithelial integrity and, consequently, their beneficial effect for the reduction of gastrointestinal disease symptoms.


Asunto(s)
Enfermedades Intestinales/prevención & control , Intestinos/efectos de los fármacos , Intestinos/fisiología , Probióticos/farmacología , Animales , Humanos
17.
BMC Microbiol ; 16(1): 291, 2016 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-27978817

RESUMEN

BACKGROUND: Streptococcus suis is an encapsulated Gram-positive bacterium and the leading cause of sepsis and meningitis in young pigs, resulting in considerable economic losses in the porcine industry. S. suis is considered an emerging zoonotic agent with increasing numbers of human cases over the last years. In the environment, both avirulent and virulent strains occur in pigs, with no evidence for consistent adapatation of virulent strains to the human host. Currently, there is an urgent need for a convenient, reliable and standardised animal model to rapidly assess S. suis virulence. Wax moth (Galleria mellonella) larvae have successfully been used in human and animal infectious disease studies. Here, we developed G. mellonella larvae as a model to assess virulence of S. suis strains. RESULTS: Fourteen isolates of S. suis belonging to different serotypes killed G. mellonella larvae in a dose-dependent manner. Larvae infected with the virulent serotype 2 strain, S. suis S3881/S10, were rescued by antibiotic therapy. Crucially, the observed virulence of the different serotypes and mutants was in agreement with virulence observed in piglets (Sus scrofa) and the zebrafish larval infection model. Infection with heat-inactivated bacteria or bacteria-free culture supernatants showed that in most cases live bacteria are needed to cause mortality in G. mellonella. CONCLUSIONS: The G. mellonella model is simple, cost-efficient, and raises less ethical issues than experiments on vertebrates and reduces infrastructure requirements. Furthermore, it allows experiments to be performed at the host temperature (37 °C). The results reported here, indicate that the G. mellonella model may aid our understanding of veterinary microbial pathogens such as the emerging zoonotic pathogen S. suis and generate hypotheses for testing in the target animal host. Ultimately, this might lead to the timely introduction of new effective remedies for infectious diseases. Last but not least, use of the G. mellonella infection model to study S. suis virulence adheres to the principles of replacement, reduction and refinement (3Rs) and can potentially reduce the number of vertebrates used for experimental infection studies.


Asunto(s)
Mariposas Nocturnas/microbiología , Infecciones Estreptocócicas/microbiología , Streptococcus suis/patogenicidad , Animales , Antibacterianos/farmacología , Modelos Animales de Enfermedad , Larva/microbiología , Mutación , Infecciones Estreptocócicas/tratamiento farmacológico , Streptococcus suis/genética , Sus scrofa , Virulencia , Pez Cebra
18.
Trends Immunol ; 34(5): 208-15, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23485516

RESUMEN

The gut microbiota provide important stimuli to the human innate and adaptive immune system and co-mediate metabolic and immune homeostasis. Probiotic bacteria can be regarded as part of the natural human microbiota, and have been associated with improving homeostasis, albeit with different levels of success. Composition of microbiota, probiotic strain identity, and host genetic differences may account for differential modulation of immune responses by probiotics. Here, we review the mechanisms of immunomodulating capacities of specific probiotic strains, the responses they can induce in the host, and how microbiota and genetic differences between individuals may co-influence host responses and immune homeostasis.


Asunto(s)
Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/microbiología , Intestinos/inmunología , Lactobacillus/inmunología , Microbiota/inmunología , Probióticos , Animales , Interacción Gen-Ambiente , Homeostasis/inmunología , Interacciones Huésped-Patógeno , Humanos , Inmunidad Mucosa , Inmunomodulación , Enfermedades Inflamatorias del Intestino/terapia , Intestinos/microbiología
19.
Cell Microbiol ; 17(2): 226-40, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25224879

RESUMEN

Faecalibacterium prausnitzii, an abundant member of the human commensal microbiota, has been proposed to have a protective role in the intestine. However, it is an obligate anaerobe, difficult to co-culture in viable form with oxygen-requiring intestinal cells. To overcome this limitation, a unique apical anaerobic model of the intestinal barrier, which enabled co-culture of live obligate anaerobes with the human intestinal cell line Caco-2, was developed. Caco-2 cells remained viable and maintained an intact barrier for at least 12 h, consistent with gene expression data, which suggested Caco-2 cells had adapted to survive in an oxygen-reduced atmosphere. Live F. prausnitzii cells, but not ultraviolet (UV)-killed F. prausnitzii, increased the permeability of mannitol across the epithelial barrier. Gene expression analysis showed inflammatory mediators to be expressed at lower amounts in Caco-2 cells exposed to live F. prausnitzii than UV-killed F. prausnitzii, This, consistent with previous reports, implies that live F. prausnitzii produces an anti-inflammatory compound in the culture supernatant, demonstrating the value of a physiologically relevant co-culture system that allows obligate anaerobic bacteria to remain viable.


Asunto(s)
Clostridium/crecimiento & desarrollo , Células Epiteliales/microbiología , Células Epiteliales/fisiología , Mucosa Intestinal/microbiología , Mucosa Intestinal/fisiología , Células CACO-2 , Supervivencia Celular , Técnicas de Cocultivo , Perfilación de la Expresión Génica , Humanos , Mediadores de Inflamación/metabolismo , Manitol/metabolismo , Modelos Teóricos , Permeabilidad
20.
BMC Microbiol ; 14: 264, 2014 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-25384512

RESUMEN

BACKGROUND: Streptococcus suis is a major problem in the swine industry causing meningitis, arthritis and pericarditis in piglets. Pathogenesis of S. suis is poorly understood. We previously showed that introduction of a 3 kb genomic fragment from virulent serotype 2 strain 10 into a weakly virulent serotype 2 strain S735, generated a hypervirulent isolate. The 3 kb genomic fragment contained two complete open reading frames (ORF) in an operon-structure of which one ORF showed similarity to folylpolyglutamate synthetase, whereas the function of the second ORF could not be predicted based on database searches for protein similarity. RESULTS: In this study we demonstrate that introduction of orf2 from strain 10 into strain S735 is sufficient to dramatically increase the virulence of S735 in pigs. This increase in virulence could not be associated with changes in pro-inflammatory responses of porcine blood mononucleated cells in response to S. suis in vitro. Sequence analysis of the orf2-folC-operon of S. suis isolates 10 and S735 revealed an SNP in the -35 region of the putative promoter sequence of the operon, as well as several SNPs resulting in amino acid substitutions in the ORF2 protein. Transcript levels of orf2 and folC were significantly higher in the virulent strain 10 than in the weakly virulent strain S735 and in vitro mutagenesis of the orf2 promoter confirmed that this was due to a SNP in the predicted -35 region upstream of the orf2 promoter. In this study, we demonstrated that the stronger promoter was present in all virulent and highly virulent S. suis isolates included in our study. This highlights a correlation between high orf2 expression and virulence. Conversely, the weaker promoter was present in isolates known to be weakly pathogenic or non-pathogenic. CONCLUSION: In summary, we demonstrate the importance of orf2 in the virulence of S. suis.


Asunto(s)
Polimorfismo de Nucleótido Simple , Regiones Promotoras Genéticas , Infecciones Estreptocócicas/microbiología , Infecciones Estreptocócicas/patología , Streptococcus suis/crecimiento & desarrollo , Streptococcus suis/genética , Factores de Virulencia/genética , Animales , Perfilación de la Expresión Génica , Operón/genética , Análisis de Secuencia de ADN , Porcinos , Virulencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA