Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Curr Res Food Sci ; 8: 100736, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38681527

RESUMO

In this study, an in vitro co-culture model using an electric cell-substrate impedance sensing system (ECIS) for testing the impact of real-time fermentation of non-digestible carbohydrates (NDCs) by the intestinal microbiota on gut barrier function was established. We applied Lactobacillus plantarum WCFS1 as a model intestinal bacterium and alginate-pectin as immobilization polymers as well as a source of NDCs to determine the impact of pectin fermentation on the barrier function of T84 gut epithelial cells. In the first design, L. plantarum WCFS1 was encapsulated in an alginate capsule followed by embedding in an agar layer to mimic a firm mucus layer that might be present in the colon. In this experimental design, the presence of the agar layer interfered with the transepithelial electrical resistance (TEER) measurement of T84 cells. Subsequently, we removed the agar layer and used encapsulated bacteria in an alginate gel and found that the TEER measurement was adequate. The encapsulation of the L. plantarum WCFS1 does avoid direct contact with cells. Also, the encapsulation system allows higher amounts of packing densities of L. plantarum WCFS1 in a limited space which can limit the oxygen concentration within the capsule and therefore create anaerobic conditions. To test this design, T84 cells were co-incubated with L. plantarum alginate-capsules supplemented with graded loads of fermentable pectin (0, 4, and 8 mg/ml per capsule) to investigate the effect of pectin fermentation on gut barrier function. We observed that as the pectin content in the L. plantarum capsules increased, pectin showed a gradually stronger protective effect on the TEER of the gut epithelium. This could partly be explained by enhanced SCFA production as both lactate and acetate were enhanced in L. plantarum containing alginate capsules with 8 mg/ml pectin. Overall, this newly designed in vitro co-culture model allows for studying the impact of bacteria-derived fermentation products but also for studying the direct effects of NDCs on gut barrier function in a relatively high-throughput way.

3.
Food Funct ; 15(2): 569-579, 2024 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-38170495

RESUMO

This study investigates the anti-inflammatory effects of pectins with different degrees of methyl esterification (DM) on intestinal epithelial cells (IECs) expressing low and high levels of TLR2. It also studies the influence of soluble TLR2 (sTLR2) which may be enhanced in patients with inflammatory bowel syndrome on the inflammation-attenuating effects of pectins. Also, it examines the impact of pectins on tight junction gene expression in IECs. Lemon pectins with DM18 and DM88 were characterized, and their effects on TLR2-1-induced IL8 gene expression and secretion were investigated in low-TLR2 expressing Caco-2 and high-TLR2 expressing DLD-1 cells. The results demonstrate that both DM18 and DM88 pectins can counteract TLR2-1-induced IL-8 expression and secretion, with more pronounced effects observed in DLD-1 cells expressing high levels of TLR2. Furthermore, the presence of sTLR2 does not interfere with the attenuating effects of low DM18 pectin and may even support its anti-inflammatory effects in Caco-2 cells. The impact of pectins and sTLR2 on tight junction gene expression also demonstrates cell-type-dependent effects. Overall, these findings suggest that low DM pectins possess potent anti-inflammatory properties and may influence tight junction gene expression in IECs, thereby contributing to the maintenance of gut homeostasis.


Assuntos
Interleucina-8 , Receptor 2 Toll-Like , Humanos , Receptor 2 Toll-Like/genética , Receptor 2 Toll-Like/metabolismo , Interleucina-8/genética , Interleucina-8/metabolismo , Células CACO-2 , Junções Íntimas/metabolismo , Esterificação , Expressão Gênica , Pectinas/farmacologia , Pectinas/metabolismo , Anti-Inflamatórios/metabolismo
4.
Front Immunol ; 13: 1028953, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36466902

RESUMO

Inflammatory Bowel Disease (IBD) is characterized by a loss of intestinal barrier function caused by an aberrant interaction between the immune response and the gut microbiota. In IBD, imbalance in cholesterol homeostasis and mitochondrial bioenergetics have been identified as essential events for activating the inflammasome-mediated response. Mitochondrial alterations, such as reduced respiratory complex activities and reduced production of tricarboxylic acid (TCA) cycle intermediates (e.g., citric acid, fumarate, isocitric acid, malate, pyruvate, and succinate) have been described in in vitro and clinical studies. Under inflammatory conditions, mitochondrial architecture in intestinal epithelial cells is dysmorphic, with cristae destruction and high dynamin-related protein 1 (DRP1)-dependent fission. Likewise, these alterations in mitochondrial morphology and bioenergetics promote metabolic shifts towards glycolysis and down-regulation of antioxidant Nuclear erythroid 2-related factor 2 (Nrf2)/Peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α) signaling. Although the mechanisms underlying the mitochondrial dysfunction during mucosal inflammation are not fully understood at present, metabolic intermediates and cholesterol may act as signals activating the NLRP3 inflammasome in IBD. Notably, dietary phytochemicals exhibit protective effects against cholesterol imbalance and mitochondrial function alterations to maintain gastrointestinal mucosal renewal in vitro and in vivo conditions. Here, we discuss the role of cholesterol and mitochondrial metabolism in IBD, highlighting the therapeutic potential of dietary phytochemicals, restoring intestinal metabolism and function.


Assuntos
Inflamassomos , Doenças Inflamatórias Intestinais , Humanos , Mitocôndrias , Colesterol , Doença Crônica , Glicólise , Ácido Pirúvico
5.
mSystems ; 7(5): e0064622, 2022 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-36005398

RESUMO

The gut microbiota is constituted by thousands of microbial interactions, some of which correspond to the exchange of metabolic by-products or cross-feeding. Inulin and xylan are two major dietary polysaccharides that are fermented by members of the human gut microbiota, resulting in different metabolic profiles. Here, we integrated community modeling and bidirectional culturing assays to study the metabolic interactions between two gut microbes, Phocaeicola dorei and Lachnoclostridium symbiosum, growing in inulin or xylan, and how they provide a protective effect in cultured cells. P. dorei (previously belonging to the Bacteroides genus) was able to consume inulin and xylan, while L. symposium only used certain inulin fractions to produce butyrate as a major end product. Constrained-based flux simulations of refined genome-scale metabolic models of both microbes predicted high lactate and succinate cross-feeding fluxes between P. dorei and L. symbiosum when growing in each fiber. Bidirectional culture assays in both substrates revealed that L. symbiosum growth increased in the presence of P. dorei. Carbohydrate consumption analyses showed a faster carbohydrate consumption in cocultures compared to monocultures. Lactate and succinate concentrations in bidirectional cocultures were lower than in monocultures, pointing to cross-feeding as initially suggested by the model. Butyrate concentrations were similar across all conditions. Finally, supernatants from both bacteria cultured in xylan in bioreactors significantly reduced tumor necrosis factor-α-induced inflammation in HT-29 cells and exerted a protective effect against the TcdB toxin in Caco-2 epithelial cells. Surprisingly, this effect was not observed in inulin cocultures. Overall, these results highlight the predictive value of metabolic models integrated with microbial culture assays for probing microbial interactions in the gut microbiota. They also provide an example of how metabolic exchange could lead to potential beneficial effects in the host. IMPORTANCE Microbial interactions represent the inner connections in the gut microbiome. By integrating mathematical modeling tools and microbial bidirectional culturing, we determined how two gut commensals engage in the exchange of cross-feeding metabolites, lactate and succinate, for increased growth in two fibers. These interactions underpinned butyrate production in cocultures, resulting in a significant reduction in cellular inflammation and protection against microbial toxins when applied to cellular models.


Assuntos
Toxinas Bacterianas , Clostridioides difficile , Microbioma Gastrointestinal , Humanos , Fibras na Dieta/farmacologia , Inulina/farmacologia , Xilanos , Toxinas Bacterianas/metabolismo , Células CACO-2 , Fermentação , Clostridioides difficile/metabolismo , Butiratos/análise , Inflamação , Lactatos , Succinatos
6.
Amino Acids ; 54(3): 311-324, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-34235577

RESUMO

Each day, varying amounts of undigested or partially digested proteins reach the colon where they are metabolized by the microbiota, resulting in the formation of compounds such as ammonia, p-cresol, skatole, phenol, indole, and hydrogen sulfide (H2S). In farm animals, the excessive production of these metabolites can affect the quality of meat and milk and is a source of contaminating emissions from animal manure. In humans, their accumulation is potentially harmful, and it has been proposed that they could be involved in the development of pathologies such as colorectal cancer and ulcerative colitis, among others. This review assesses the evidence supporting the use of dietary polyphenols to reduce the production of these metabolites. Most studies have used condensed (proanthocyanidins) or hydrolyzable (ellagitannins and gallotannins) tannins, and have been carried out in farm animals. Several show that the administration of tannins in pigs, chicken, and ruminants decreases the levels of ammonia, p-cresol, skatole, and/or H2S, improving meat/milk quality and reducing manure odor. Direct application of tannins to manure also decreases ammonia emissions. Few studies were carried out in rats and humans and their results confirm, to a lesser extent, those reported in farm animals. These effects would be due to the capacity of tannins to trap ammonia and H2S, and to modify the composition of the microbiota, reducing the bacterial populations producing metabolites. In addition, PACs prevent p-cresol and H2S-induced alterations on intestinal cells in vitro. Tannins, therefore, appear as an interesting tool for improving the quality of animal products, human health, and the harmful emissions associated with breeding.


Assuntos
Microbiota , Polifenóis , Aminoácidos , Animais , Colo , Polifenóis/farmacologia , Ratos , Suínos , Taninos
7.
Front Immunol ; 12: 658354, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34122415

RESUMO

The diverse and dynamic microbial community of the human gastrointestinal tract plays a vital role in health, with gut microbiota supporting the development and function of the gut immune barrier. Crosstalk between microbiota-gut epithelium and the gut immune system determine the individual health status, and any crosstalk disturbance may lead to chronic intestinal conditions, such as inflammatory bowel diseases (IBD) and celiac disease. Microbiota-derived metabolites are crucial mediators of host-microbial interactions. Some beneficially affect host physiology such as short-chain fatty acids (SCFAs) and secondary bile acids. Also, tryptophan catabolites determine immune responses, such as through binding to the aryl hydrocarbon receptor (AhR). AhR is abundantly present at mucosal surfaces and when activated enhances intestinal epithelial barrier function as well as regulatory immune responses. Exogenous diet-derived indoles (tryptophan) are a major source of endogenous AhR ligand precursors and together with SCFAs and secondary bile acids regulate inflammation by lowering stress in epithelium and gut immunity, and in IBD, AhR expression is downregulated together with tryptophan metabolites. Here, we present an overview of host microbiota-epithelium- gut immunity crosstalk and review how microbial-derived metabolites contribute to host immune homeostasis. Also, we discuss the therapeutic potential of bacterial catabolites for IBD and celiac disease and how essential dietary components such as dietary fibers and bacterial tryptophan catabolites may contribute to intestinal and systemic homeostasis.


Assuntos
Bactérias/metabolismo , Microbioma Gastrointestinal , Interações entre Hospedeiro e Microrganismos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Animais , Ácidos e Sais Biliares/metabolismo , Fibras na Dieta , Suscetibilidade a Doenças , Microbioma Gastrointestinal/imunologia , Homeostase , Interações entre Hospedeiro e Microrganismos/imunologia , Humanos , Doenças Inflamatórias Intestinais/etiologia , Doenças Inflamatórias Intestinais/metabolismo , Ligantes , Receptores de Hidrocarboneto Arílico/metabolismo , Triptofano/metabolismo
8.
Int J Mol Sci ; 22(6)2021 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-33802759

RESUMO

This review describes current evidence supporting butyrate impact in the homeostatic regulation of the digestive ecosystem in health and inflammatory bowel diseases (IBDs). Butyrate is mainly produced by bacteria from the Firmicutes phylum. It stimulates mature colonocytes and inhibits undifferentiated malignant and stem cells. Butyrate oxidation in mature colonocytes (1) produces 70-80% of their energetic requirements, (2) prevents stem cell inhibition by limiting butyrate access to crypts, and (3) consumes oxygen, generating hypoxia and maintaining luminal anaerobiosis favorable to the microbiota. Butyrate stimulates the aryl hydrocarbon receptor (AhR), the GPR41 and GPR109A receptors, and inhibits HDAC in different cell types, thus stabilizing the gut barrier function and decreasing inflammatory processes. However, some studies indicate contrary effects according to butyrate concentrations. IBD patients exhibit a lower abundance of butyrate-producing bacteria and butyrate content. Additionally, colonocyte butyrate oxidation is depressed in these subjects, lowering luminal anaerobiosis and facilitating the expansion of Enterobacteriaceae that contribute to inflammation. Accordingly, gut dysbiosis and decreased barrier function in IBD seems to be secondary to the impaired mitochondrial disturbance in colonic epithelial cells.


Assuntos
Butiratos/farmacologia , Colo/patologia , Homeostase , Doenças Inflamatórias Intestinais/patologia , Animais , Colo/efeitos dos fármacos , Epigênese Genética/efeitos dos fármacos , Microbioma Gastrointestinal/efeitos dos fármacos , Microbioma Gastrointestinal/genética , Homeostase/efeitos dos fármacos , Homeostase/genética , Humanos
9.
Rev Endocr Metab Disord ; 22(2): 367-388, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33387285

RESUMO

Obesity is a global public health problem that results in chronic pathologies such as diabetes, cardiovascular diseases, and cancer. The treatment approach based on energy restriction and promotion of physical activity is ineffective in the long term. Due to the high prevalence of this pathology, complementary treatments such as brown adipose tissue activation (BAT) and white adipose tissue browning (WAT) have been proposed. Dietary polyphenols are plant secondary metabolites that can stimulate browning and thermogenesis of adipose tissue. They have also been shown to prevent body weight gain, and decrease systemic inflammation produced by high-fat diets. Ingested dietary polyphenols that reach the colon are metabolized by the gut microbiota (GM), regulating its composition and generating a great array of metabolites. GM is involved in the production of short chain fatty acids and secondary bile salts that regulate energetic metabolism. The alteration in the composition of GM observed in metabolic diseases such as obesity and type 2 diabetes can be attenuated by polyphenols. Recent studies support the hypothesis that GM would mediate WAT browning and BAT thermogenesis activation induced by polyphenol administration. Together, these results indicate that GM in the presence of polyphenols plays a fundamental role in the control of obesity possible through BAT activation.


Assuntos
Diabetes Mellitus Tipo 2 , Microbioma Gastrointestinal , Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Branco/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Metabolismo Energético , Humanos , Obesidade/metabolismo , Obesidade/prevenção & controle , Polifenóis/metabolismo , Polifenóis/farmacologia , Termogênese
10.
Front Nutr ; 7: 163, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33072794

RESUMO

Macroalgae stand out for their high content of dietary fiber (30-75%) that include soluble, sulfated (fucoidan, agaran, carrageenan, and ulvan) and non-sulfated (laminaran and alginate) polysaccharides. Many studies indicate that these compounds exert varied biological activities and health-promoting effects and for this reason, there is a growing interest for using them in food products. The aim of this review was to critically evaluate prebiotic properties of algal polysaccharides, i.e., their ability to exert biological activities by modulating the composition and/or diversity of gut microbiota (GM). Pre-clinical studies show that the non-sulfated alginate and laminaran are well-fermented by GM, promoting the formation of short chain fatty acids (SCFAs) including butyrate, and preventing that of harmful putrefactive compounds (NH3, phenol, p-cresol, indole and H2S). Alginate increases Bacteroides, Bifidobacterium, and Lactobacillus species while laminaran mostly stimulates Bacteroides sp. Results with sulfated polysaccharides are more questionable. Agarans are poorly fermentable but agarose-oligosaccharides exhibit an interesting prebiotic potential, increasing butyrate-producing bacteria and SCFAs. Though carrageenan-oligosaccharides are also fermented, their use is currently limited due to safety concerns. Regarding fucoidan, only one study reports SCFAs production, suggesting that it is poorly fermented. Its effect on GM does not indicate a clear pattern, making difficult to conclude whether it is beneficial or not. Notably, fucoidan impact on H2S production has not been evaluated, though some studies report it increases sulfate-reducing bacteria. Ulvan is badly fermented by GM and some studies show that part of its sulfate is dissimilated to H2S, which could affect colonic mitochondrial function. Accordingly, these results support the use of laminaran, alginate and agaro-oligosaccharides as prebiotics while more studies are necessary regarding that of fucoidan, carrageenan and ulvan. However, the realization of clinical trials is necessary to confirm such prebiotic properties in humans.

11.
Rev. chil. nutr ; 47(2): 317-327, abr. 2020. tab, graf
Artigo em Espanhol | LILACS | ID: biblio-1115503

RESUMO

RESUMEN La microbiota intestinal (MI) es considerada como un nuevo blanco para la prevención y manejo nutricional de las alteraciones inflamatorias y metabólicas asociadas a las enfermedades crónicas no-transmisibles. Los prebióticos son principalmente fibras solubles cuyo consumo favorece el crecimiento de poblaciones bacterianas beneficiosas de la MI e impacta favorablemente la salud del consumidor. Esta revisión presenta a los fitoquímicos dietarios, que incluyen a más de 8.000 compuestos, como una nueva clase de prebióticos debido a su capacidad de estimular poblaciones de Lactobacillus, Bifidobacterium, Akkermansia y de bacterias productoras de butirato en el colon, a expensa de bacterias potencialmente dañinas como C. histolyticum. Además, los fitoquímicos son transformados por la MI en múltiples metabolitos que ejercen actividades biológicas a veces más potentes que la molécula inicial de la cual provienen. Individuos con distintos metabotipos han sido descritos de acuerdo a su capacidad de responder al consumo de isoflavonas, lignanos o elagitaninos, dependiendo de la presencia en su MI de bacterias capaces de transformar dichos polifenoles en equol, enterolactona/enterodiol y urolitinas, respectivamente, los cuales exhiben actividades biológicas. Valerolactonas y ácidos aromáticos también son producidos por la MI a través del metabolismo de las proantocianidinas. El efecto prebiótico de los fitoquímicos contribuiría a explicar los efectos saludables del consumo de frutas y verduras ricos en fitoquímicos.


ABSTRACT Intestinal microbiota (IM) is considered as a new target for the prevention and nutritional management of inflammatory and metabolic alterations associated with non-transmissible chronic diseases. Prebiotics are mainly soluble fibers whose consumption favors the growth of beneficial bacterial populations of the IM and positively impacts health. This review discusses dietary phytochemicals, which include more than 8,000 compounds, as a new class of prebiotics due to its ability to stimulate populations of Lactobacillus, Bifidobacterium, Akkermansia and butyrate producing bacteria in the colon at the expense of potentially harmful bacteria, such as C. histolyticum. In addition, phytochemicals are transformed by IM into a great array of metabolites exerting biological activities and are sometimes more potent than the initial molecule from which they are derived. Individuals with different metabotypes have been described according to their ability to respond to the consumption of isoflavones, lignans or ellagitannins, depending on the presence in their IM of bacteria capable of transforming these polyphenols into equol, enterolactone/enterodiol and urolithins, respectively, which exhibit biological activities. Valerolactones and aromatic acids are also produced by the IM through proanthocyanidin metabolism. The prebiotic effect of phytochemicals would help to explain the healthy effects associated with the consumption of fruits and vegetables rich in phytochemicals.


Assuntos
Humanos , Prebióticos , Compostos Fitoquímicos/metabolismo , Compostos Fitoquímicos/química , Produtos Biológicos , Dieta , Polifenóis/classificação , Polifenóis/metabolismo , Polifenóis/química , Disbiose , Microbioma Gastrointestinal
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...