Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Gut Microbes ; 16(1): 2335879, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38695302

RESUMEN

Dietary omega-3 polyunsaturated fatty acids (n-3 PUFAs) and the gut microbiome affect each other. We investigated the impact of supplementation with Buglossoides arvensis oil (BO), rich in stearidonic acid (SDA), on the human gut microbiome. Employing the Mucosal Simulator of the Human Intestinal Microbial Ecosystem (M-SHIME), we simulated the ileal and ascending colon microbiomes of four donors. Our results reveal two distinct microbiota clusters influenced by BO, exhibiting shared and contrasting shifts. Notably, Bacteroides and Clostridia abundance underwent similar changes in both clusters, accompanied by increased propionate production in the colon. However, in the ileum, cluster 2 displayed a higher metabolic activity in terms of BO-induced propionate levels. Accordingly, a triad of bacterial members involved in propionate production through the succinate pathway, namely Bacteroides, Parabacteroides, and Phascolarctobacterium, was identified particularly in this cluster, which also showed a surge of second-generation probiotics, such as Akkermansia, in the colon. Finally, we describe for the first time the capability of gut bacteria to produce N-acyl-ethanolamines, and particularly the SDA-derived N-stearidonoyl-ethanolamine, following BO supplementation, which also stimulated the production of another bioactive endocannabinoid-like molecule, commendamide, in both cases with variations across individuals. Spearman correlations enabled the identification of bacterial genera potentially involved in endocannabinoid-like molecule production, such as, in agreement with previous reports, Bacteroides in the case of commendamide. This study suggests that the potential health benefits on the human microbiome of certain dietary oils may be amenable to stratified nutrition strategies and extend beyond n-3 PUFAs to include microbiota-derived endocannabinoid-like mediators.


Asunto(s)
Bacterias , Endocannabinoides , Microbioma Gastrointestinal , Humanos , Microbioma Gastrointestinal/efectos de los fármacos , Bacterias/clasificación , Bacterias/metabolismo , Bacterias/aislamiento & purificación , Bacterias/genética , Endocannabinoides/metabolismo , Colon/microbiología , Colon/metabolismo , Íleon/microbiología , Íleon/metabolismo , Ácidos Grasos Omega-3/metabolismo , Aceites de Plantas/metabolismo , Aceites de Plantas/farmacología , Suplementos Dietéticos , Adulto , Masculino
2.
NPJ Biofilms Microbiomes ; 10(1): 18, 2024 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-38448452

RESUMEN

Cranberry is associated with multiple health benefits, which are mostly attributed to its high content of (poly)phenols, particularly flavan-3-ols. However, clinical trials attempting to demonstrate these positive effects have yielded heterogeneous results, partly due to the high inter-individual variability associated with gut microbiota interaction with these molecules. In fact, several studies have demonstrated the ability of these molecules to modulate the gut microbiota in animal and in vitro models, but there is a scarcity of information in human subjects. In addition, it has been recently reported that cranberry also contains high concentrations of oligosaccharides, which could contribute to its bioactivity. Hence, the aim of this study was to fully characterize the (poly)phenolic and oligosaccharidic contents of a commercially available cranberry extract and evaluate its capacity to positively modulate the gut microbiota of 28 human subjects. After only four days, the (poly)phenols and oligosaccharides-rich cranberry extract, induced a strong bifidogenic effect, along with an increase in the abundance of several butyrate-producing bacteria, such as Clostridium and Anaerobutyricum. Plasmatic and fecal short-chain fatty acids profiles were also altered by the cranberry extract with a decrease in acetate ratio and an increase in butyrate ratio. Finally, to characterize the inter-individual variability, we stratified the participants according to the alterations observed in the fecal microbiota following supplementation. Interestingly, individuals having a microbiota characterized by the presence of Prevotella benefited from an increase in Faecalibacterium with the cranberry extract supplementation.


Asunto(s)
Microbioma Gastrointestinal , Vaccinium macrocarpon , Animales , Humanos , Butiratos , Fenoles , Extractos Vegetales/farmacología , Oligosacáridos , Suplementos Dietéticos
3.
Mol Nutr Food Res ; 68(5): e2300641, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38350729

RESUMEN

Clinical trials investigating the health effects of flavan-3-ols yield heterogeneous results due to interindividual variability in the gut microbiota metabolism. In fact, different groups in the population have similar metabolic profiles following (-)-epicatechin and (+)-catechin gut microbial metabolism and can be regrouped into so-called metabotypes. In this study, the capacity of 34 donors to metabolize polymeric B-type flavan-3-ols from aronia and oligomeric A-type flavan-3-ols from cranberry is investigated by in vitro fecal batch fermentations. Less than 1% of the flavan-3-ols from both sources are converted into microbial metabolites, such as phenyl-γ-valerolactones (PVLs). To further confirm this result, gut microbial metabolites from flavan-3-ols are quantified in urine samples collected from participants, before and after a 4-day supplementation of cranberry extract providing 82.3 mg of flavan-3-ols per day. No significant difference is observed in the urinary excretion of flavan-3-ols microbial metabolites. Hence, it demonstrates by both in vitro and in vivo approaches that flavan-3-ols from aronia and cranberry are poorly degraded by the gut microbiota. The beneficial health impacts of these molecules likely stem from their capacity to affect gut microbiota and their interactions with the gut epithelium, rather than from their breakdown into smaller metabolites.


Asunto(s)
Catequina , Microbioma Gastrointestinal , Photinia , Vaccinium macrocarpon , Humanos , Flavonoides/farmacología , Catequina/metabolismo , Extractos Vegetales/farmacología
4.
FASEB J ; 38(2): e23398, 2024 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-38214938

RESUMEN

In vitro systems are widely employed to assess the impact of dietary compounds on the gut microbiota and their conversion into beneficial bacterial metabolites. However, the complex fluid dynamics and multi-segmented nature of these systems can complicate the comprehensive analysis of dietary compound fate, potentially confounding physical dilution or washout with microbial catabolism. In this study, we developed fluid dynamics models based on sets of ordinary differential equations to simulate the behavior of an inert compound within two commonly used in vitro systems: the continuous two-stage PolyFermS system and the semi-continuous multi-segmented SHIME® system as well as into various declinations of those systems. The models were validated by investigating the fate of blue dextran, demonstrating excellent agreement between experimental and modeling data (with r2 values ranging from 0.996 to 0.86 for different approaches). As a proof of concept for the utility of fluid dynamics models in in vitro system, we applied generated models to interpret metabolomic data of procyanidin A2 (ProA2) generated from the addition of proanthocyanidin (PAC)-rich cranberry extract to both the PolyFermS and SHIME® systems. The results suggested ProA2 degradation by the gut microbiota when compared to the modeling of an inert compound. Models of fluid dynamics developed in this study provide a foundation for comprehensive analysis of gut metabolic data in commonly utilized in vitro PolyFermS and SHIME® bioreactor systems and can enable a more accurate understanding of the contribution of bacterial metabolism to the variability in the concentration of target metabolites.


Asunto(s)
Microbioma Gastrointestinal , Hidrodinámica , Fermentación , Modelos Teóricos , Bacterias
5.
Commun Biol ; 6(1): 959, 2023 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-37735539

RESUMEN

N-oleoylglycine (OlGly), a lipid derived from the basic component of olive oil, oleic acid, and N-oleoylalanine (OlAla) are endocannabinoid-like mediators. We report that OlGly and OlAla, by activating the peroxisome proliferator-activated receptor alpha (PPARα), reduce the rewarding properties of a highly palatable food, dopamine neuron firing in the ventral tegmental area, and the obesogenic effect of a high-fat diet rich in lard (HFD-L). An isocaloric olive oil HFD (HFD-O) reduced body weight gain compared to the HFD-L, in a manner reversed by PPARα antagonism, and enhanced brain and intestinal OlGly levels and gut microbial diversity. OlGly or OlAla treatment of HFD-L mice resulted in gut microbiota taxonomic changes partly similar to those induced by HFD-O. We suggest that OlGly and OlAla control body weight by counteracting highly palatable food overconsumption, and possibly rebalancing the gut microbiota, and provide a potential new mechanism of action for the obeso-preventive effects of olive oil-rich diets.


Asunto(s)
Endocannabinoides , PPAR alfa , Animales , Ratones , Aceite de Oliva/farmacología , Obesidad/etiología , Obesidad/prevención & control , Peso Corporal
6.
J Agric Food Chem ; 71(37): 13814-13827, 2023 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-37683128

RESUMEN

Although the relationship between gut microbiota and flavan-3-ol metabolism differs greatly between individuals, the specific metabolic profiles, known as metabotypes, have not yet been clearly defined. In this study, fecal batch fermentations of 34 healthy donors inoculated with (-)-epicatechin were stratified into groups based on their conversion rate of (-)-epicatechin and their quali-quantitative metabolic profile. Fast and slow converters of (-)-epicatechin, high producers of 1-(3'-hydroxyphenyl)-3-(2″,4″,6″-trihydroxyphenyl)-propan-2-ol (3-HPP-2-ol) and 5-(3',4'-dihydroxyphenyl)-γ-valerolactone (3,4-DHPVL) were identified. Fecal microbiota analysis revealed that fast conversion of (-)-epicatechin was associated with short-chain fatty acid (SCFA)-producing bacteria, such as Faecalibacterium spp. and Bacteroides spp., and higher levels of acetate, propionate, butyrate, and valerate were observed for fast converters. Other bacteria were associated with the conversion of 1-(3',4'-dihydroxyphenyl)-3-(2″,4″,6″-trihydroxyphenyl)-propan-2-ol into 3-HPP-2-ol (Lachnospiraceae UCG-010 spp.) and 3,4-DHPVL (Adlercreutzia equolifaciens). Such stratification sheds light on the mechanisms of action underlying the high interindividual variability associated with the health benefits of flavan-3-ols.


Asunto(s)
Catequina , Humanos , 2-Propanol , Butiratos , Clostridiales , Heces
7.
NPJ Biofilms Microbiomes ; 8(1): 86, 2022 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-36266277

RESUMEN

The intestinal mucus layer has a dual role in human health constituting a well-known microbial niche that supports gut microbiota maintenance but also acting as a physical barrier against enteric pathogens. Enterotoxigenic Escherichia coli (ETEC), the major agent responsible for traveler's diarrhea, is able to bind and degrade intestinal mucins, representing an important but understudied virulent trait of the pathogen. Using a set of complementary in vitro approaches simulating the human digestive environment, this study aimed to describe how the mucus microenvironment could shape different aspects of the human ETEC strain H10407 pathophysiology, namely its survival, adhesion, virulence gene expression, interleukin-8 induction and interactions with human fecal microbiota. Using the TNO gastrointestinal model (TIM-1) simulating the physicochemical conditions of the human upper gastrointestinal (GI) tract, we reported that mucus secretion and physical surface sustained ETEC survival, probably by helping it to face GI stresses. When integrating the host part in Caco2/HT29-MTX co-culture model, we demonstrated that mucus secreting-cells favored ETEC adhesion and virulence gene expression, but did not impede ETEC Interleukin-8 (IL-8) induction. Furthermore, we proved that mucosal surface did not favor ETEC colonization in a complex gut microbial background simulated in batch fecal experiments. However, the mucus-specific microbiota was widely modified upon the ETEC challenge suggesting its role in the pathogen infectious cycle. Using multi-targeted in vitro approaches, this study supports the major role played by mucus in ETEC pathophysiology, opening avenues in the design of new treatment strategies.


Asunto(s)
Escherichia coli Enterotoxigénica , Infecciones por Escherichia coli , Microbiota , Humanos , Escherichia coli Enterotoxigénica/fisiología , Interleucina-8/genética , Virulencia , Diarrea , Células CACO-2 , Infecciones por Escherichia coli/microbiología , Viaje , Bacterias , Moco , Mucinas
8.
Gut Microbes ; 14(1): 2120344, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36109831

RESUMEN

Consumption of omega-3 polyunsaturated fatty acids (ω-3 PUFAs) eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) provides multifaceted health benefits. Recent studies suggest that ω-3 PUFAs modulate the gut microbiota by enhancing health-promoting bacteria, such as the mucin specialist Akkermansia muciniphila. However, these prebiotic properties have been poorly investigated and direct effects on the gut microbiome have never been explored dynamically across gut regions and niches (lumen vs. mucus-associated microbiota). Thus, we studied the effects of 1 week EPA- and DHA-enriched ω-3 fish-oil supplementation on the composition and functionality of the human microbiome in a Mucosal Simulator of the Human Intestinal Microbial Ecosystem (M-SHIME®). Gut microbial communities derived from one individual harvested in two different seasons were tested in duplicate. Luminal and outer mucus-associated microbiota of the ileum, ascending, transverse and descending colons were cultivated over 28 d from fecal inoculates and supplemented with ω-3 PUFAs for the last 7 d. We show that ω-3 PUFA supplementation modulates the microbiota in a gut region- and niche-dependent fashion. The outer mucus-associated microbiota displayed a higher resilience than the luminal mucin habitat to ω-3 PUFAs, with a remarkable blooming of Akkermansia muciniphila in opposition to a decrease of Firmicutes-mucolytic bacteria. The ω-3 PUFAs also induced a gradual and significant depletion of non-mucolytic Clostridia members in luminal habitats. Finally, increased concentrations of the short chain fatty acids (SCFA) propionate in colon regions at the end of the supplementation was associated positively with the bloom of Akkermansia muciniphila and members of the Desulfovibrionia class.


Asunto(s)
Ácidos Grasos Omega-3 , Microbioma Gastrointestinal , Microbiota , Akkermansia , Ácidos Docosahexaenoicos/farmacología , Ácido Eicosapentaenoico/farmacología , Expectorantes/farmacología , Ácidos Grasos Omega-3/farmacología , Ácidos Grasos Volátiles , Fermentación , Firmicutes , Humanos , Mucinas , Prebióticos , Propionatos/farmacología , Verrucomicrobia
9.
Microbiol Spectr ; 10(5): e0243221, 2022 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-35972287

RESUMEN

Uropathogenic Escherichia coli (UPEC) ecology-pathophysiology from the gut reservoir to its urothelium infection site is poorly understood, resulting in equivocal benefits in the use of cranberry as prophylaxis against urinary tract infections. To add further understanding from the previous findings on PAC antiadhesive properties against UPEC, we assessed in this study the effects of proanthocyanidins (PAC) rich cranberry extract microbial metabolites on UTI89 virulence and fitness in contrasting ecological UPEC's environments. For this purpose, we developed an original model combining a colonic fermentation system (SHIME) with a dialysis cassette device enclosing UPEC and a 3D tissue-engineered urothelium. Two healthy fecal donors inoculated the colons. Dialysis cassettes containing 7log10 CFU/mL UTI89 were immersed for 2h in the SHIME colons to assess the effect of untreated (7-day control diet)/treated (14-day PAC-rich extract) metabolomes on UPEC behavior. Engineered urothelium were then infected with dialysates containing UPEC for 6 h. This work demonstrated for the first time that in the control fecal microbiota condition without added PAC, the UPEC virulence genes were activated upstream the infection site, in the gut. However, PAC microbial-derived cranberry metabolites displayed a remarkable propensity to blunt activation of genes encoding toxin, adhesin/invasins in the gut and on the urothelium, in a donor-dependent manner. Variability in subjects' gut microbiota and ensuing contrasting cranberry PAC metabolism affects UPEC virulence and should be taken into consideration when designing cranberry efficacy clinical trials. IMPORTANCE Uropathogenic Escherichia coli (UPEC) are the primary cause of recurrent urinary tract infections (UTI). The poor understanding of UPEC ecology-pathophysiology from its reservoir-the gut, to its infection site-the urothelium, partly explains the inadequate and abusive use of antibiotics to treat UTI, which leads to a dramatic upsurge in antibiotic-resistance cases. In this context, we evaluated the effect of a cranberry proanthocyanidins (PAC)-rich extract on the UPEC survival and virulence in a bipartite model of a gut microbial environment and a 3D urothelium model. We demonstrated that PAC-rich cranberry extract microbial metabolites significantly blunt activation of UPEC virulence genes at an early stage in the gut reservoir. We also showed that altered virulence in the gut affects infectivity on the urothelium in a microbiota-dependent manner. Among the possible mechanisms, we surmise that specific microbial PAC metabolites may attenuate UPEC virulence, thereby explaining the preventative, yet contentious properties of cranberry against UTI.


Asunto(s)
Infecciones por Escherichia coli , Proteínas de Escherichia coli , Proantocianidinas , Infecciones Urinarias , Escherichia coli Uropatógena , Vaccinium macrocarpon , Humanos , Antibacterianos/farmacología , Infecciones por Escherichia coli/prevención & control , Infecciones por Escherichia coli/tratamiento farmacológico , Extractos Vegetales/farmacología , Extractos Vegetales/uso terapéutico , Proantocianidinas/farmacología , Proantocianidinas/uso terapéutico , Infecciones Urinarias/prevención & control , Infecciones Urinarias/tratamiento farmacológico , Urotelio , Virulencia
10.
Biotechnol Adv ; 54: 107796, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34252564

RESUMEN

The human digestion is a multi-step and multi-compartment process essential for human health, at the heart of many issues raised by academics, the medical world and industrials from the food, nutrition and pharma fields. In the first years of life, major dietary changes occur and are concomitant with an evolution of the whole child digestive tract anatomy and physiology, including colonization of gut microbiota. All these phenomena are influenced by child exposure to environmental compounds, such as drugs (especially antibiotics) and food pollutants, but also childhood infections. Due to obvious ethical, regulatory and technical limitations, in vivo approaches in animal and human are more and more restricted to favor complementary in vitro approaches. This review summarizes current knowledge on the evolution of child gut physiology from birth to 3 years old regarding physicochemical, mechanical and microbial parameters. Then, all the available in vitro models of the child digestive tract are described, ranging from the simplest static mono-compartmental systems to the most sophisticated dynamic and multi-compartmental models, and mimicking from the oral phase to the colon compartment. Lastly, we detail the main applications of child gut models in nutritional, pharmaceutical and microbiological studies and discuss the limitations and challenges facing this field of research.


Asunto(s)
Contaminantes Ambientales , Microbioma Gastrointestinal , Animales , Niño , Digestión , Tracto Gastrointestinal/microbiología , Tracto Gastrointestinal/fisiología , Humanos
11.
Gut Microbes ; 13(1): 1953246, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34432600

RESUMEN

Enterotoxigenic Escherichia coli (ETEC) is one of the most common causes of acute traveler's diarrhea. Adhesins and enterotoxins constitute the major ETEC virulence traits. With the dramatic increase in antibiotic resistance, probiotics are considered a wholesome alternative to prevent or treat ETEC infections. Here, we examined the antimicrobial properties of the probiotic Saccharomyces cerevisiae CNCM I-3856 against ETEC H10407 pathogenesis upon co-administration in the TNO gastrointestinal Model (TIM-1), simulating the physicochemical and enzymatic conditions of the human upper digestive tract and preventive treatment in the Mucosal Simulator of the Human Intestinal Microbial Ecosystem (M-SHIME), integrating microbial populations of the ileum and ascending colon. Interindividual variability was assessed by separate M-SHIME experiments with microbiota from six human individuals. The probiotic did not affect ETEC survival along the digestive tract. However, ETEC pathogenicity was significantly reduced: enterotoxin encoding virulence genes were repressed, especially in the TIM-1 system, and a lower enterotoxin production was noted. M-SHIME experiments revealed that 18-days probiotic treatment stimulate the growth of Bifidobacterium and Lactobacillus in different gut regions (mucosal and luminal, ileum and ascending colon) while a stronger metabolic activity was noted in terms of short-chain fatty acids (acetate, propionate, and butyrate) and ethanol production. Moreover, the probiotic pre-treated microbiota displayed a higher robustness in composition following ETEC challenge compared to the control condition. We thus demonstrated the multi-inhibitory properties of the probiotic S. cerevisiae CNCM I-3856 against ETEC in the overall simulated human digestive tract, regardless of the inherent variability across individuals in the M-SHIME.


Asunto(s)
Escherichia coli Enterotoxigénica/efectos de los fármacos , Infecciones por Escherichia coli/tratamiento farmacológico , Enfermedades Transmitidas por los Alimentos/tratamiento farmacológico , Microbioma Gastrointestinal/efectos de los fármacos , Probióticos/farmacología , Probióticos/uso terapéutico , Virulencia/efectos de los fármacos , Infecciones por Escherichia coli/fisiopatología , Humanos , Saccharomyces cerevisiae/química
12.
BMC Biol ; 18(1): 141, 2020 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-33054775

RESUMEN

BACKGROUND: Enterotoxigenic Escherichia coli (ETEC) substantially contributes to the burden of diarrheal illnesses in developing countries. With the use of complementary in vitro models of the human digestive environment, TNO gastrointestinal model (TIM-1), and Mucosal Simulator of the Human Intestinal Microbial Ecosystem (M-SHIME), we provided the first detailed report on the spatial-temporal modulation of ETEC H10407 survival, virulence, and its interplay with gut microbiota. These systems integrate the main physicochemical parameters of the human upper digestion (TIM-1) and simulate the ileum vs ascending colon microbial communities and luminal vs mucosal microenvironments, captured from six fecal donors (M-SHIME). RESULTS: A loss of ETEC viability was noticed upon gastric digestion, while a growth renewal was found at the end of jejunal and ileal digestion. The remarkable ETEC mucosal attachment helped to maintain luminal concentrations above 6 log10 mL-1 in the ileum and ascending colon up to 5 days post-infection. Seven ETEC virulence genes were monitored. Most of them were switched on in the stomach and switched off in the TIM-1 ileal effluents and in a late post-infectious stage in the M-SHIME ascending colon. No heat-labile enterotoxin production was measured in the stomach in contrast to the ileum and ascending colon. Using 16S rRNA gene-based amplicon sequencing, ETEC infection modulated the microbial community structure of the ileum mucus and ascending colon lumen. CONCLUSIONS: This study provides a better understanding of the interplay between ETEC and gastrointestinal cues and may serve to complete knowledge on ETEC pathogenesis and inspire novel prophylactic strategies for diarrheal diseases.


Asunto(s)
Escherichia coli Enterotoxigénica/fisiología , Escherichia coli Enterotoxigénica/patogenicidad , Infecciones por Escherichia coli/microbiología , Microbioma Gastrointestinal/fisiología , Colon Ascendente/microbiología , Humanos , Íleon/microbiología , Viabilidad Microbiana
13.
Future Microbiol ; 12: 73-93, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27983878

RESUMEN

Enterotoxigenic Escherichia coli (ETEC) are a major cause of traveler's diarrhea and infant mortality in developing countries. Given the rise of antibiotic resistance worldwide, there is an urgent need for the development of new preventive strategies. Among them, a promising approach is the use of probiotics. Although many studies, mostly performed under piglet digestive conditions, have shown the beneficial effects of probiotics on ETEC by interfering with their survival, virulence or adhesion to mucosa, underlying mechanisms remain unclear. This review describes ETEC pathogenesis, its modulation by human gastrointestinal cues as well as novel preventive strategies with a particular emphasis on probiotics. The potential of in vitro models simulating human digestion in elucidating probiotic mode of action will be discussed.


Asunto(s)
Escherichia coli Enterotoxigénica/aislamiento & purificación , Enfermedades Transmitidas por los Alimentos/prevención & control , Microbioma Gastrointestinal , Probióticos/administración & dosificación , Infecciones por Escherichia coli/microbiología , Infecciones por Escherichia coli/prevención & control , Enfermedades Transmitidas por los Alimentos/microbiología , Enfermedades Gastrointestinales/microbiología , Enfermedades Gastrointestinales/prevención & control , Tracto Gastrointestinal/microbiología , Humanos , Concentración de Iones de Hidrógeno
14.
Pediatr Res ; 80(5): 734-743, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27429202

RESUMEN

BACKGROUND: Enterohemorrhagic Escherichia coli (EHEC) are major foodborne pathogens that constitute a serious public health threat, mainly in young children. Shiga toxins (Stx) are the main virulence determinants of EHEC pathogenesis but adhesins like intimin (eae) and Long polar fimbriae (Lpf) also contribute to infection. The TNO GastroIntestinal Model (TIM) was used for a comparative study of EHEC O157:H7 survival and virulence under adult and child digestive conditions. METHODS: Survival kinetics in the in vitro digestive tract were determined by plating while bacterial viability was assessed by flow cytometry analysis. Expression of stx, eae, and lpf genes was followed by reverse transcriptase-quantitative PCR (RT-qPCR) and Stx production was measured by ELISA (enzyme-linked immunosorbent assay). RESULTS: Upon gastrointestinal passage, a higher amount of viable cells was found in the simulated ileal effluents of children compared to that of adults (with 34 and 6% of viable cells, respectively). Expression levels of virulence genes were up to 125-fold higher in children. Stx was detected only in child ileal effluents. CONCLUSION: Differences in digestive physicochemical parameters may partially explain why children are more susceptible to EHEC infection than adults. Such data are essential for a full understanding of EHEC pathogenesis and would help in designing novel therapeutic approaches.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Escherichia coli Enterohemorrágica/crecimiento & desarrollo , Escherichia coli Enterohemorrágica/genética , Infecciones por Escherichia coli/microbiología , Toxina Shiga/metabolismo , Factores de Virulencia/metabolismo , Adhesinas Bacterianas/genética , Adulto , Niño , Escherichia coli Enterohemorrágica/patogenicidad , Escherichia coli O157/genética , Escherichia coli O157/crecimiento & desarrollo , Escherichia coli O157/patogenicidad , Citometría de Flujo , Mucosa Gástrica/metabolismo , Humanos , Intestino Delgado/metabolismo , Cinética , Modelos Biológicos , Toxina Shiga/genética , Virulencia , Factores de Virulencia/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...