Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 142
1.
Plant Foods Hum Nutr ; 78(4): 698-703, 2023 Dec.
Article En | MEDLINE | ID: mdl-37919537

Dietary prebiotic fibers play an important role in modulating gut microbiota by enhancing the abundance of beneficial microorganisms and their bioactive metabolites. However, dietary fibers are a structurally heterogeneous class of polysaccharides, varying in molar mass, branching patterns, and monosaccharide composition, which could influence their utilization by various gut microorganisms. The present study aimed to investigate the effects of molar mass and chemical structure of wheat arabinoxylan fiber (AX) on the growth and metabolism of two key gut resident bacteria (Faecalibacterium prausnitzii and Lacticaseibacillus rhamnosus LGG), which are linked to human health. For this purpose, low, medium, and high molar masses of AX (LAX, MAX, and HAX, respectively) were modified with specific α-arabinofuranosidases to leave only singly substituted, only doubly substituted, or unsubstituted xylose units. Almost all the modified AX samples showed a better prebiotic score than unmodified AX for different molar masses. The modified LAX exhibited a better prebiotic effect than HAX and MAX. In addition, LAX, with doubly substituted xylose units, exhibited the highest prebiotic potential and SCFA production by both microorganisms. Furthermore, AX, either singly or doubly substituted, had a consistent impact on L. rhamnosus growth, whereas AX, with all arabinose residues removed, had a greater impact on F. prausnitzii. These findings support the potential of bioengineered AX as next-generation prebiotics targeting health-related gut microbes.


Gastrointestinal Microbiome , Prebiotics , Humans , Prebiotics/microbiology , Triticum/chemistry , Xylose , Dietary Fiber/analysis , Xylans/chemistry
2.
Int J Food Sci Nutr ; 74(7): 760-780, 2023 Nov.
Article En | MEDLINE | ID: mdl-37771001

Studies have shown that gut dysbiosis is associated with the steatotic liver disease associated with metabolic dysfunction (MALSD) and its severity. This study evaluated the effects of two commercially available prebiotics fructooligosaccharides (FOS) and galactooligosaccharides(GOS) on hepatic adipogenesis, inflammation, and gut microbiota in high-fat diet-induced MALSD. The results indicated that FOS and GOS effectively reduced insulin resistance, hyperglycaemia, triglyceridemia, cholesterolaemia, and IL-1ß serum levels. Moreover, FOS and GOS modulated the lipogenic (SREBP-1c, ACC, and FAS) and lipolytic (ATGL) signalling pathways, and reduced inflammatory markers such as p-NFκB-65, IL-6, iNOS, COX-2, TNF-α, IL-1ß, and nitrotyrosine. FOS and GOS also enhanced the abundance of acetate producers' bacteria Bacteroides acidifaciens and Bacteroides dorei. FOS and GOS also induced positive POMC/GPR43 neurons at the arcuate nucleus, indicating hypothalamic signalling modulation. Our results suggest that FOS and GOS attenuated MALSD by reducing the hepatic lipogenic pathways and intestinal permeability through the gut microbiota-brain axis.


Fatty Liver , Gastrointestinal Microbiome , Microbiota , Humans , Oligosaccharides/pharmacology , Oligosaccharides/metabolism , Prebiotics/microbiology , Brain/metabolism
3.
Rev Med Suisse ; 19(830): 1149-1153, 2023 Jun 07.
Article Fr | MEDLINE | ID: mdl-37283386

The microbiota represents all the microorganisms including viruses, bacteria, fungi, and parasites, that have a symbiotic relationship with their host and that are present in a particular system (or niche) of the human body such as the skin, the respiratory tract, the urogenital tract or the digestive tract. This paper is a narrative review of all talks given at the 8th edition of the « Feeding the Microbiota ¼ symposium organized at the Geneva University Hospitals. The symposium gathered 346 participants, both onsite and online, from 23 countries all-around the world. The main thematic of this edition focused on the composition of the gut microbiota as affected by prebiotics and postbiotics and their effects on various diseases.


Le microbiote représente l'ensemble des micro-organismes (virus, bactéries, champignons et parasites) qui ont une relation symbiotique avec leur hôte et qui sont présents dans un système particulier du corps humain comme la peau, les voies respiratoires et/ou uro-génitales ou encore le tube digestif. Cet article est une revue narrative des différentes thématiques exposées lors du 8e symposium « Feeding the Microbiota ¼ organisé aux HUG le 9 février 2023. L'événement a réuni 346 participants en présentiel et en ligne venant de 23 pays différents. La thématique de cette édition s'est focalisée sur les effets des prébiotiques et des probiotiques sur la composition du microbiote et dans le contexte de certaines maladies.


Gastrointestinal Microbiome , Microbiota , Probiotics , Humans , Prebiotics/microbiology , Probiotics/therapeutic use , Gastrointestinal Tract/microbiology
4.
Int J Mol Sci ; 23(3)2022 Jan 26.
Article En | MEDLINE | ID: mdl-35163307

The study of endoxylanases as catalysts to valorize hemicellulosic residues and to obtain glycosides with improved properties is a topic of great industrial interest. In this work, a GH10 ß-1,4-endoxylanase (XynSOS), from the ascomycetous fungus Talaromyces amestolkiae, has been heterologously produced in Pichia pastoris, purified, and characterized. rXynSOS is a highly glycosylated monomeric enzyme of 53 kDa that contains a functional CBM1 domain and shows its optimal activity on azurine cross-linked (AZCL)-beechwood xylan at 70 °C and pH 5. Substrate specificity and kinetic studies confirmed its versatility and high affinity for beechwood xylan and wheat arabinoxylan. Moreover, rXynSOS was capable of transglycosylating phenolic compounds, although with low efficiencies. For expanding its synthetic capacity, a glycosynthase variant of rXynSOS was developed by directed mutagenesis, replacing its nucleophile catalytic residue E236 by a glycine (rXynSOS-E236G). This novel glycosynthase was able to synthesize ß-1,4-xylooligosaccharides (XOS) of different lengths (four, six, eight, and ten xylose units), which are known to be emerging prebiotics. rXynSOS-E236G was also much more active than the native enzyme in the glycosylation of a broad range of phenolic compounds with antioxidant properties. The interesting capabilities of rXynSOS and its glycosynthase variant make them promising tools for biotechnological applications.


Glucuronates/metabolism , Glycosides/metabolism , Oligosaccharides/metabolism , Phenols/metabolism , Talaromyces/metabolism , Endo-1,4-beta Xylanases/metabolism , Kinetics , Pichia/metabolism , Prebiotics/microbiology , Substrate Specificity , Xylans/metabolism , Xylose/metabolism
5.
Food Funct ; 13(3): 1256-1267, 2022 Feb 07.
Article En | MEDLINE | ID: mdl-35023534

The aim of this study was to investigate the prebiotic activities of dextran (LM742) produced by Leuconostoc mesenteroides SPCL742 in the aspect of the human gut microbial ecosystem focusing on microbiome and metabolome changes in in vitro colonic fermentation. LM742 dextran had a medium-chain structure with the molecular weight of 1394.87 kDa (DP = 7759.22) and α-1,6 and α-1,3 linkages with a 26.11 : 1 ratio. The LM742 dextran was resistent to digestive enzymes in the human gastrointestinal conditions. The individual cultivation of 30 intestinal bacteria with LM742 dextran showed the growth of Bacteroides spp., whereas in vitro human fecal fermentation with LM742 exhibited the symbiotic growth of Bacteroides spp. and beneficial bacteria such as Bifidobacterium spp. Further co-cultivation of Bacteroides xylanisolvens and several probiotics indicated that B. xylanisolvens provides a cross-feeding of dextran to probiotics. In fecal fermentation, LM742 dextran resulted in increased concentrations of short-chain fatty acids, valerate and pantothenate, but it rarely affected the conversion of betaine to trimethylamine. Lastly, LM742 dextran inhibited the adhesion of pathogenic E. coli to human epithelial cells. Taken together, these results demonstrate the prebiotic potential of LM742 dextran as a health-beneficial polysaccharide in the human intestine.


Dextrans/metabolism , Gastrointestinal Microbiome , Leuconostoc mesenteroides/metabolism , Prebiotics/microbiology , Humans
6.
Int J Biol Macromol ; 194: 412-421, 2022 Jan 01.
Article En | MEDLINE | ID: mdl-34813784

Pectins have proven to be advantageous for human health as they regulate beneficial microbial communities and enhance immunity. The fruit of Clausena lansium (Lour.) Skeels (Wampee), also referred to as "treasure in fruit", is rich in pectin polysaccharides. In this study, a homogalacturonan-type pectin (CCP2) with a molecular weight of 8.9 × 104 Da and degree of esterification of 42.86% was isolated from Wampee fruit. The gut microbiota regulation and phagocytosis-enhancing properties of CCP2 were examined in vivo and in vitro, respectively. Oral administration of CCP2 dramatically decreased the abundance of Bacteroidetes and increased the abundance of Firmicutes in intestinal bacteria in mice. The content of short-chain fatty acids in the feces also significantly improved. Moreover, CCP2 exhibited excellent phagocytosis-enhancing activities on RAW 264.7 macrophages. These results suggested that CCP2 could be a potential gut microbiota regulator and phagocytosis-enhancer, which could be used in food products to promote health through beneficial manipulation of gut microbiota.


Clausena/metabolism , Fruit/metabolism , Gastrointestinal Microbiome/drug effects , Pectins/therapeutic use , Prebiotics/microbiology , Animals , Animals, Outbred Strains , Mice , RAW 264.7 Cells
7.
Nutrients ; 13(12)2021 Nov 25.
Article En | MEDLINE | ID: mdl-34959781

Chickpea-based foods are known for their low allergenicity and rich nutritional package. As an essential dietary legume, chickpea is often processed into milk or hummus or as an industrial source of protein and starch. The current study explores the feasibility of using the chickpea-derived prebiotic substances as a scaffold for growing Bacillus subtilis (a prospective probiotic bacterium) to develop synbiotic chickpea-based functional food. We report that the chickpea-derived fibers enhance the formation of the B. subtilis biofilms and the production of the antimicrobial pigment pulcherrimin. Furthermore, electron micrograph imaging confirms the bacterial embedding onto the chickpea fibers, which may provide a survival tactic to shield and protect the bacterial population from environmental insults. Overall, it is believed that chickpea-derived prebiotic substances provide a staple basis for developing functional probiotics and synbiotic food.


Bacillus subtilis/growth & development , Biofilms/growth & development , Cicer/microbiology , Functional Food/microbiology , Prebiotics/microbiology , Amino Acids, Sulfur/biosynthesis , Feasibility Studies , Humans , Piperidines , Probiotics/analysis , Synbiotics/analysis , Tissue Scaffolds
8.
Int J Mol Sci ; 22(24)2021 Dec 15.
Article En | MEDLINE | ID: mdl-34948270

In recent years, commensal bacteria colonizing the human body have been recognized as important determinants of health and multiple pathologic conditions. Among the most extensively studied commensal bacteria are the gut microbiota, which perform a plethora of functions, including the synthesis of bioactive products, metabolism of dietary compounds, and immunomodulation, both through attenuation and immunostimulation. An imbalance in the microbiota population, i.e., dysbiosis, has been linked to many human pathologies, including various cancer types and neurodegenerative diseases. Targeting gut microbiota and microbiome-host interactions resulting from probiotics, prebiotics, and postbiotics is a growing opportunity for the effective treatment of various diseases. As more research is being conducted, the microbiome field is shifting from simple descriptive analysis of commensal compositions to more molecular, cellular, and functional studies. Insight into these mechanisms is of paramount importance for understanding and modulating the effects that microbiota, probiotics, and their derivatives exert on host health.


Gastrointestinal Microbiome/physiology , Host Microbial Interactions/drug effects , Bacteria , Diet , Dysbiosis/microbiology , Humans , Microbiota/physiology , Prebiotics/microbiology , Probiotics/metabolism , Probiotics/pharmacology , Symbiosis/physiology
9.
Sci Rep ; 11(1): 20567, 2021 10 18.
Article En | MEDLINE | ID: mdl-34663866

The objective of this present study was to investigate the potentiality of prebiotics (mannan oligosaccharides-MOS and fructo-oligosaccharides-FOS) in replacement of antibiotic growth promoter and their relationship with physico-chemical indices, antioxidant and oxidative stability and carcass traits of broiler chickens meat. Accordingly, 240 day-old broiler chicks of uniform body weight divided in 6 treatment groups with 5 replicate each (5 × 6 = 30) having 8 birds in each replicate. Six corn based dietary treatments were formulated viz. T1 (control diet), T2 (T1 + Bacitracin methylene di-salicylate @ 0.002%), T3 (T1 + 0.1% MOS), T4 (T1 + 0.2% MOS), T5 (T1 + 0.1% FOS), and T6 (T1 + 0.2% FOS). Significant (p < 0.05) increase in cut up part yields (%) and reduction in cholesterol and fat content in T4 (0.2% MOS) group. The water holding capacity (WHC) and extract release volume (ERV) were increase (p < 0.05) in 0.1 or 0.2% MOS supplemented group. DPPH (1, 1-diphenyl-2-picrylhydrazy) was higher (p < 0.05) and lipid oxidation (free fatty acid and thio-barbituric acid reactive substances) was lower (p < 0.05) in T4 group. The standard plate count (SPC), staphylococcus and coliform counts were decreased (p < 0.05) in T3 or T4 group. Thus, it can be concluded that mannan oligosaccharides (MOS) may be incorporated at 0.2% level in diet for improved physico-chemical indices, antioxidant and oxidative stability and carcass characteristics of broiler chickens meat and it may be suitable replacer of antibiotic growth promoter.


Chickens/metabolism , Dietary Supplements/analysis , Mannans/pharmacology , Meat , Animal Feed/analysis , Animal Husbandry/methods , Animal Nutritional Physiological Phenomena , Animals , Antioxidants , Chickens/growth & development , Diet/veterinary , Lipid Metabolism , Oligosaccharides/pharmacology , Oxidation-Reduction , Oxidative Stress , Prebiotics/microbiology
10.
Rev. neurol. (Ed. impr.) ; 73(8): 282-295, Oct 16, 2021. ilus, tab
Article Es | IBECS | ID: ibc-229589

Introducción: El trastorno del espectro autista es un trastorno del neurodesarrollo con heterogeneidad fenotípica y curso sintomático variable de etiología parcialmente desconocida. La prevalencia de trastornos gastrointestinales en este perfil de pacientes invita a investigar el papel que la microbiota intestinal puede tener como factor causal y a plantear intervenciones terapéuticas específicas. El papel de la microbiota en el desarrollo y la función cerebral, demostrado en modelos animales, justifica su investigación en este trastorno neuropsiquiátrico. Objetivo: Investigar la relación entre la alteración en la composición de la microbiota y el trastorno del espectro autista, y evaluar el papel terapéutico de prebióticos, probióticos y trasplante fecal en este trastorno del neurodesarrollo. Desarrollo: Se realizó una revisión bibliográfica en PubMed, Cochrane Library y Google Scholar con el fin de seleccionar los artículos relevantes relacionados con el tema que se publicaron entre enero de 2012 y abril de 2020. Se seleccionaron 35 artículos relevantes. En 23 de ellos se encontraron diferencias significativas en la composición y la diversidad de la microbiota en niños con TEA, así como en biomoléculas involucradas en determinadas rutas metabólicas. Las otras 12 investigaciones describieron mejorías gastrointestinales y comportamentales tras la intervención terapéutica. Conclusiones:Resulta razonable afirmar que existe evidencia suficiente para apoyar la existencia de una relación entre la microbiota intestinal y los trastornos del espectro autista. Esta vinculación ha de ser explorada en profundidad para perfilar el peso etiopatogénico de la disbiosis y las posibles herramientas terapéuticas.(AU)


Introduction: Autism spectrum disorder is a neurodevelopmental disorder with phenotypic heterogeneity and variable symptomatic course of partly unknown etiology. The prevalence of gastrointestinal disorders in autism leads to investigate the role that intestinal microbiota may have as a causal factor and to propose specific therapeutic interventions. The role of microbiota in brain development and function, demonstrated in animal models, justifies its investigation in this neuropsychiatric disorder. Objective: The aim was to investigate the relationship between altered microbiota composition and autism spectrum disorder, and to assess the therapeutic role of prebiotics, probiotics and fecal transplantation in this neurodevelopmental disorder. Development: A literature review was conducted in PubMed, Cochrane Library and Google Scholar to select relevant articles related to the topic that were published between January 2012 and April 2020. Thirty-five relevant articles were selected. In 23 of them, significant differences were found in the composition and diversity of the microbiota in children with ASD, as well as in the biomolecules involved in certain metabolic pathways. The other 12 investigations reported gastrointestinal and behavioral improvements after therapeutic intervention. Conclusions: It is reasonable to state that there is enough evidence to support the existence of a relationship between intestinal microbiota and autism spectrum disorders. This fact should be explored in depth to assess the etiopathogenic burden of dysbiosis and the possible therapeutic tools.(AU)


Humans , Male , Female , Gastrointestinal Microbiome/drug effects , Gastrointestinal Diseases/drug therapy , Autism Spectrum Disorder/drug therapy , Neurodevelopmental Disorders/genetics , Prebiotics/microbiology , Probiotics , Neurology , Nervous System Diseases , Fecal Microbiota Transplantation
11.
Nutrients ; 13(9)2021 Aug 27.
Article En | MEDLINE | ID: mdl-34578853

Human milk serves as a model for infant formula providing nutritional solutions for infants not able to receive enough mother's milk. Infant formulas aim to mimic the composition and functionality of human milk by providing ingredients reflecting those of the latest human milk insights, such as prebiotics, probiotics and postbiotics. The aim of this study was to examine the effects of the supplementation with a postbiotic (LactofidusTM) and its combination with the prebiotics short-chain galactooligosaccharides (scGOS) and long-chain fructooligosaccharides (lcFOS) in a preclinical model of healthy suckling rats. Pups were supplemented daily with LactofidusTM (POST group) and/or scGOS/lcFOS (P+P and PRE groups, respectively). Body weight and fecal consistency were analyzed. At the end of the study, immunoglobulin (Ig) profile, intestinal gene expression, microbiota composition and short chain fatty acid (SCFA) proportion were quantified. The supplementation with all nutritional interventions modulated the Ig profile, but the prebiotic mixture and the postbiotic induced differential effects: whereas scGOS/lcFOS induced softer feces and modulated microbiota composition and SCFA profile, Lactofidus™ upregulated Toll-like receptors gene expression. The use of the combination of scGOS/lcFOS and Lactofidus™ showed the effects observed for the oligosaccharides separately, as well as showing a synergistic impact on animal growth. Thus, the combined use of both products seems to be a good strategy to modulate immune and microbial features in early life.


Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/immunology , Immunity/drug effects , Immunity/immunology , Prebiotics/administration & dosage , Probiotics/administration & dosage , Animals , Animals, Newborn , Models, Animal , Prebiotics/microbiology , Rats
12.
Biochem Biophys Res Commun ; 571: 125-130, 2021 09 24.
Article En | MEDLINE | ID: mdl-34325127

This study investigated the properties of Latilactobacillus curvatus MS2 isolated from Korean traditional fermented seafood as probiotics and the effect of reducing cholesterol as a synbiotic with isomalto-oligosaccharide (IMO) in BALB/c mice. The isolated strain showed high resistance to acids and bile acids and exhibited a high DPPH scavenging capacity of 72.27 ± 0.38 %. In the intestinal adhesion test using HT-29 cells, the adhesion rate of MS2 was 17.10 ± 1.78 %, which was higher than the adhesion rate of the other investigated probiotics. MS2 showed good antimicrobial activity against food-borne pathogens, especially Staphylococcus aureus, S. epidermidis, Escherichia coli, and Vibrio vulnificus. This strain had high availability for IMO among the prebiotics of fructo-oligosaccharide, inulin and IMO. Oral administration of MS2 and IMO to BALB/c mice for 5 weeks resulted in a significant reduction in blood cholesterol levels by regulating liver lipid metabolism. These results suggest that the combination of MS2 and IMO has potential for application in functional foods.


Cholesterol/metabolism , Fermentation , Lactobacillaceae/isolation & purification , Oligosaccharides/metabolism , Prebiotics/microbiology , Seafood/microbiology , Animals , Male , Mice , Mice, Inbred BALB C , Republic of Korea , Synbiotics
13.
Int J Mol Sci ; 22(14)2021 Jul 18.
Article En | MEDLINE | ID: mdl-34299291

Schizophrenia is a chronic, heterogeneous neurodevelopmental disorder that has complex symptoms and uncertain etiology. Mounting evidence indicates the involvement of genetics and epigenetic disturbances, alteration in gut microbiome, immune system abnormalities, and environmental influence in the disease, but a single root cause and mechanism involved has yet to be conclusively determined. Consequently, the identification of diagnostic markers and the development of psychotic drugs for the treatment of schizophrenia faces a high failure rate. This article surveys the etiology of schizophrenia with a particular focus on gut microbiota regulation and the microbial signaling system that correlates with the brain through the vagus nerve, enteric nervous system, immune system, and production of postbiotics. Gut microbially produced molecules may lay the groundwork for further investigations into the role of gut microbiota dysbiosis and the pathophysiology of schizophrenia. Current treatment of schizophrenia is limited to psychotherapy and antipsychotic drugs that have significant side effects. Therefore, alternative therapeutic options merit exploration. The use of psychobiotics alone or in combination with antipsychotics may promote the development of novel therapeutic strategies. In view of the individual gut microbiome structure and personalized response to antipsychotic drugs, a tailored and targeted manipulation of gut microbial diversity naturally by novel prebiotics (non-digestible fiber) may be a successful alternative therapeutic for the treatment of schizophrenia patients.


Antipsychotic Agents/therapeutic use , Gastrointestinal Microbiome/drug effects , Probiotics/therapeutic use , Schizophrenia/microbiology , Schizophrenia/therapy , Brain/microbiology , Dysbiosis/immunology , Dysbiosis/metabolism , Dysbiosis/microbiology , Gastrointestinal Microbiome/physiology , Humans , Immune System , Prebiotics/microbiology
14.
Genes (Basel) ; 12(5)2021 05 01.
Article En | MEDLINE | ID: mdl-34062867

Epigenetic regulation of gene expression is a form of interaction of the external environment on reading and transcription of genetic information encoded in nucleic acids. We provided evidence that early stimulation of the chicken microbiota with in ovo delivered synbiotics influenced gene expression and DNA methylation in the liver. Therefore, we hypothesize that the stimulation of microbiota by administering bioactive substances in ovo also affects the activity of miRNA in liver. For the analysis of miRNA activity, RNA was isolated from liver of adult broiler chicken and native chicken breed. The animals received a prebiotic, probiotic and synbiotic in ovo on day 12 of egg incubation. The analysis of miRNA expression was performed using the LNA method on a miRNA panel selected on the basis of previous microarray experiments. We have found increased miRNA expression activity after probiotic and synbiotic administration, especially in native chicken breed. Our results suggest that prebiotics reduce or do not affect miRNA activity. We have also shown that miRNA activity is regulated by the substance and genotype of the chicken. We can conclude that miRNAs constitute an important component of the molecular mechanism of host-probiotic interaction in liver.


Gastrointestinal Microbiome , Liver/metabolism , MicroRNAs/genetics , Synbiotics/administration & dosage , Animals , Chickens , MicroRNAs/metabolism , Prebiotics/administration & dosage , Prebiotics/microbiology , Probiotics/administration & dosage , Transcriptome
15.
Nutrients ; 13(6)2021 May 21.
Article En | MEDLINE | ID: mdl-34064139

The therapeutic potential of Sargassum siliquosum grown in Australian tropical waters was tested in a rat model of metabolic syndrome. Forty-eight male Wistar rats were divided into four groups of 12 rats and each group was fed a different diet for 16 weeks: corn starch diet (C); high-carbohydrate, high-fat diet (H) containing fructose, sucrose, saturated and trans fats; and C or H diets with 5% S. siliquosum mixed into the food from weeks 9 to 16 (CS and HS). Obesity, hypertension, dyslipidaemia, impaired glucose tolerance, fatty liver and left ventricular fibrosis developed in H rats. In HS rats, S. siliquosum decreased body weight (H, 547 ± 14; HS, 490 ± 16 g), fat mass (H, 248 ± 27; HS, 193 ± 19 g), abdominal fat deposition and liver fat vacuole size but did not reverse cardiovascular and liver effects. H rats showed marked changes in gut microbiota compared to C rats, while S. siliquosum supplementation increased gut microbiota belonging to the family Muribaculaceae. This selective increase in gut microbiota likely complements the prebiotic actions of the alginates. Thus, S. siliquosum may be a useful dietary additive to decrease abdominal and liver fat deposition.


Dietary Supplements , Metabolic Syndrome/therapy , Obesity/therapy , Sargassum , Seaweed/microbiology , Abdominal Fat/microbiology , Animals , Body Weight/physiology , Diet/adverse effects , Disease Models, Animal , Gastrointestinal Microbiome/physiology , Liver/microbiology , Male , Metabolic Syndrome/etiology , Metabolic Syndrome/microbiology , Obesity/etiology , Obesity/microbiology , Prebiotics/microbiology , Rats , Rats, Wistar
16.
Int J Biol Macromol ; 181: 877-889, 2021 Jun 30.
Article En | MEDLINE | ID: mdl-33864864

ß-glucans are polysaccharides which can be obtained from different sources, and which have been described as potential prebiotics. The beneficial effects associated with ß-glucan intake are that they reduce energy intake, lower cholesterol levels and support the immune system. Nevertheless, the mechanism(s) of action underpinning these health effects related to ß-glucans are still unclear, and the precise impact of ß-glucans on the gut microbiota has been subject to debate and revision. In this review, we summarize the most recent advances involving structurally different types of ß-glucans as fermentable substrates for Bacteroidetes (mainly Bacteroides) and Bifidobacterium species as glycan degraders. Bacteroides is one of the most abundant bacterial components of the human gut microbiota, while bifidobacteria are widely employed as a probiotic ingredient. Both are generalist glycan degraders capable of using a wide range of substrates: Bacteroides spp. are specialized as primary degraders in the metabolism of complex carbohydrates, whereas Bifidobacterium spp. more commonly metabolize smaller glycans, in particular oligosaccharides, sometimes through syntrophic interactions with Bacteroides spp., in which they act as secondary degraders.


Bacteroides/metabolism , Bifidobacterium/metabolism , Gastrointestinal Microbiome/genetics , beta-Glucans/metabolism , Carbohydrate Metabolism/genetics , Humans , Prebiotics/microbiology
17.
Molecules ; 26(6)2021 Mar 11.
Article En | MEDLINE | ID: mdl-33799580

Postbiotics are health-promoting microbial metabolites delivered as a functional food or a food supplement. They either directly influence signaling pathways of the body or indirectly manipulate metabolism and the composition of intestinal microflora. Cancer is the second leading cause of death worldwide and even though the prognosis of patients is improving, it is still poor in the substantial part of the cases. The preventable nature of cancer and the importance of a complex multi-level approach in anticancer therapy motivate the search for novel avenues of establishing the anticancer environment in the human body. This review summarizes the principal findings demonstrating the usefulness of both natural and synthetic sources of postbotics in the prevention and therapy of cancer. Specifically, the effects of crude cell-free supernatants, the short-chain fatty acid butyrate, lactic acid, hydrogen sulfide, and ß-glucans are described. Contradictory roles of postbiotics in healthy and tumor tissues are highlighted. In conclusion, the application of postbiotics is an efficient complementary strategy to combat cancer.


Gastrointestinal Microbiome/drug effects , Neoplasms/diet therapy , Probiotics/pharmacology , Butyrates/pharmacology , Dietary Supplements/microbiology , Fatty Acids, Volatile/metabolism , Fatty Acids, Volatile/pharmacology , Humans , Hydrogen Sulfide/pharmacology , Lactic Acid/pharmacology , Metabolome , Neoplasms/metabolism , Prebiotics/microbiology , Probiotics/metabolism , beta-Glucans/pharmacology
18.
Gut Microbes ; 13(1): 1-20, 2021.
Article En | MEDLINE | ID: mdl-33439065

Butyrate produced by gut microbiota has multiple beneficial effects on host health, and oligosaccharides derived from host diets and glycans originating from host mucus are major sources of its production. A significant reduction of butyrate-producing bacteria has been reported in patients with inflammatory bowel diseases and colorectal cancers. Although gut butyrate levels are important for host health, oligosaccharide metabolic properties in butyrate producers are poorly characterized. We studied the metabolic properties of fructooligosaccharides (FOSs) and other prebiotic oligosaccharides (i.e. raffinose and xylooligosaccharides; XOSs) in gut butyrate producers. 1-Kestose (kestose) and nystose, FOSs with degrees of polymerization of 3 and 4, respectively, were also included. Fourteen species of butyrate producers were divided into four groups based on their oligosaccharide metabolic properties, which are group A (two species) metabolizing all oligosaccharides tested, group F (four species) metabolizing FOSs but not raffinose and XOSs, group XR (four species) metabolizing XOSs and/or raffinose but not FOSs, and group N (four species) metabolizing none of the oligosaccharides tested. Species assigned to groups A and XR are rich glycoside hydrolase (GH) holders, whereas those in groups F and N are the opposite. In total, 17 enzymes assigned to GH32 were observed in nine of the 14 butyrate producers tested, and species that metabolized FOSs had at least one active GH32 enzyme. The GH32 enzymes were divided into four clusters by phylogenetic analysis. Heterologous gene expression analysis revealed that the GH32 enzymes in each cluster had similar FOS degradation properties within clusters, which may be linked to the conservation/substitution of amino acids to bind with substrates in GH32 enzymes. This study provides important knowledge to understand the impact of FOS supplementation on the activation of gut butyrate producers. Abbreviations: SCFA, short chain fatty acid; FOS, fructooligosaccharide; XOS, xylooligosaccharide; CAZy, Carbohydrate Active Enzymes; CBM, carbohydrate-binding module; PUL, polysaccharide utilization locus; S6PH sucrose-6-phosphate hydrolase.


Bacteria/metabolism , Butyrates/metabolism , Gastrointestinal Microbiome , Oligosaccharides/metabolism , Bacteria/classification , Bacteria/enzymology , Bacteria/genetics , Genome, Bacterial/genetics , Glycoside Hydrolases/genetics , Glycoside Hydrolases/metabolism , Humans , Phylogeny , Prebiotics/microbiology
19.
J Agric Food Chem ; 69(1): 170-182, 2021 Jan 13.
Article En | MEDLINE | ID: mdl-33382612

Prebiotic human milk oligosaccharides (HMOs) are found in human milk, which are not digested by infants but are metabolized by beneficial gut bacteria. We determined the ability of 57 bacterial strains within the Family Lactobacillaceae and genera Bifidobacterium and Bacteroides and potentially pathogenic bacteria to ferment the HMOs 2'-fucosyllactose, 3-fucosyllactose, and difucosyllactose. In addition, prebiotic galacto-oligosaccharides (GOS), lactose, fucose, and glucose were evaluated as carbon sources for these bacterial strains. Bacterial growth was monitored using the automatic Bioscreen C system. Only certain bifidobacteria, such as Bifidobacterium longum subsp. infantis and Bifidobacterium bifidum, as well as Bacteroides fragilis, Bacteroides vulgatus, and Bacteroides thetaiotaomicron utilized the studied HMOs as their sole carbon source, whereas almost all studied bacterial strains were able to utilize GOS, lactose, and glucose. The selectivity in utilization of HMOs by only certain bacteria can be advantageous by promoting beneficial microbes but not supporting the harmful pathogens in contrast to other less selective prebiotics.


Bacteroides/metabolism , Bifidobacterium/metabolism , Lactobacillaceae/metabolism , Milk, Human/metabolism , Oligosaccharides/metabolism , Prebiotics/microbiology , Probiotics/metabolism , Trisaccharides/metabolism , Humans , Milk, Human/microbiology , Prebiotics/analysis
20.
Compr Rev Food Sci Food Saf ; 19(4): 1908-1933, 2020 07.
Article En | MEDLINE | ID: mdl-33337097

The bioactive ingredients in commonly consumed foods include, but are not limited to, prebiotics, prebiotic-like components, probiotics, and postbiotics. The bioactive ingredients in functional foods have also been associated with beneficial effects on human health. For example, they aid in shaping of gut microflora and promotion of immunity. These functional components also contribute in preventing serious diseases such as cardiovascular malfunction and tumorigenesis. However, the specific mechanisms of these positive influences on human health are still under investigation. In this review, we aim to emphasize the major contents of probiotics, prebiotics, and prebiotic-like components commonly found in consumable functional foods, and we present an overview of direct and indirect benefits they provide on human health. The major contributors are certain families of metabolites, specifically short-chain fatty acids and polyunsaturated fatty acids produced by probiotics, and prebiotics, or prebiotic-like components such as flavonoids, polyphenols, and vitamins that are found in functional foods. These functional ingredients in foods influence the gut microbiota by stimulating the growth of beneficial microbes and the production of beneficial metabolites that, in turn, have direct benefits to the host, while also providing protection from pathogens and maintaining a balanced gut ecosystem. The complex interactions that arise among functional food ingredients, human physiology, the gut microbiota, and their respective metabolic pathways have been found to minimize several factors that contribute to the incidence of chronic disease, such as inflammation oxidative stress.


Functional Food , Prebiotics/microbiology , Probiotics/chemistry , Fatty Acids , Gastrointestinal Microbiome/physiology , Humans , Probiotics/pharmacology
...