Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Mol Pharm ; 20(12): 6197-6212, 2023 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-37955627

RESUMEN

Compound X is a weak basic drug targeting the early stages of Parkinson's disease, for which a theoretical risk assessment has indicated that elevated gastric pH conditions could potentially result in reduced plasma concentrations. Different in vitro dissolution methodologies varying in level of complexity and a physiologically based pharmacokinetic (PBPK) absorption model demonstrated that the dissolution, solubility, and intestinal absorption of compound X was indeed reduced under elevated gastric pH conditions. These observations were confirmed in a crossover pharmacokinetic study in Beagle dogs. As a result, the development of a formulation resulting in robust performance that is not sensitive to the exposed gastric pH levels is of crucial importance. The dynamic intestinal absorption MODel (Diamod), an advanced in vitro gastrointestinal transfer tool that allows to study the gastrointestinal dissolution and interconnected permeation of drugs, was selected as an in vitro tool for the formulation optimization activities given its promising predictive capacity and its capability to generate insights into the mechanisms driving formulation performance. Different pH-modifiers were screened for their potential to mitigate the pH-effect by decreasing the microenvironmental pH at the dissolution surface. Finally, an optimized formulation containing a clinically relevant dose of the drug and a functional amount of the selected pH-modifier was evaluated for its performance in the Diamod. This monolayer tablet formulation resulted in rapid gastric dissolution and supersaturation, inducing adequate intestinal supersaturation and permeation of compound X, irrespective of the gastric acidity level in the stomach. In conclusion, this study describes the holistic biopharmaceutics approach driving the development of a patient-centric formulation of compound X.


Asunto(s)
Absorción Intestinal , Atención Dirigida al Paciente , Humanos , Animales , Perros , Composición de Medicamentos , Administración Oral , Absorción Intestinal/fisiología , Solubilidad
2.
Appl Microbiol Biotechnol ; 100(9): 4097-107, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26861055

RESUMEN

Inulin-type fructans (ITF) are known to cause a health-promoting bifidogenic effect, although the ITF degradation capacity of bifidobacteria in different intestinal regions remains unclear. The present study aims at offering new insights into this link, making use of a collection of 190 bifidobacterial strains, encompassing strains from gut biopsies (terminal ileum and proximal colon; mucosa-associated strains) and the simulator of the human intestinal microbial ecosystem (SHIME®; proximal and distal colon vessels; lumen-associated strains). A multivariate data analysis of all fermentation data revealed four clusters corresponding with different types of ITF degradation fingerprints, which were not correlated with the region in the intestine, suggesting that the degradation of ITF is uniform along the human intestine. Strains from cluster 1 consumed fructose, while strains from cluster 2 consumed more oligofructose than fructose. Higher fructose and oligofructose consumption was characteristic for clusters 3 and 4 strains, which degraded inulin too. In general, the mucosa-associated strains from biopsy origin seemed to be more specialized in the consumption of fructose and oligofructose, while the lumen-associated strains from SHIME origin displayed a higher degradation degree of inulin. Further, intra-species variability in ITF degradation was found, indicating strain-specific variations. The coexistence of different bifidobacterial strains with different ITF degradation fingerprints within the same intestinal region suggests cooperation for the degradation of ITF, with opportunities for cross-feeding on strain and/or species level.


Asunto(s)
Bifidobacterium/metabolismo , Fructanos/metabolismo , Intestinos/microbiología , Fermentación , Humanos
3.
Appl Environ Microbiol ; 80(6): 1848-57, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24413595

RESUMEN

Four cocoa-specific acetic acid bacterium (AAB) strains, namely, Acetobacter pasteurianus 386B, Acetobacter ghanensis LMG 23848(T), Acetobacter fabarum LMG 24244(T), and Acetobacter senegalensis 108B, were analyzed kinetically and metabolically during monoculture laboratory fermentations. A cocoa pulp simulation medium (CPSM) for AAB, containing ethanol, lactic acid, and mannitol, was used. All AAB strains differed in their ethanol and lactic acid oxidation kinetics, whereby only A. pasteurianus 386B performed a fast oxidation of ethanol and lactic acid into acetic acid and acetoin, respectively. Only A. pasteurianus 386B and A. ghanensis LMG 23848(T) oxidized mannitol into fructose. Coculture fermentations with A. pasteurianus 386B or A. ghanensis LMG 23848(T) and Lactobacillus fermentum 222 in CPSM for lactic acid bacteria (LAB) containing glucose, fructose, and citric acid revealed oxidation of lactic acid produced by the LAB strain into acetic acid and acetoin that was faster in the case of A. pasteurianus 386B. A triculture fermentation with Saccharomyces cerevisiae H5S5K23, L. fermentum 222, and A. pasteurianus 386B, using CPSM for LAB, showed oxidation of ethanol and lactic acid produced by the yeast and LAB strain, respectively, into acetic acid and acetoin. Hence, acetic acid and acetoin are the major end metabolites of cocoa bean fermentation. All data highlight that A. pasteurianus 386B displayed beneficial functional roles to be used as a starter culture, namely, a fast oxidation of ethanol and lactic acid, and that these metabolites play a key role as substrates for A. pasteurianus in its indispensable cross-feeding interactions with yeast and LAB during cocoa bean fermentation.


Asunto(s)
Acetobacter/fisiología , Cacao/metabolismo , Microbiología de Alimentos , Interacciones Microbianas , Acetobacter/crecimiento & desarrollo , Acetobacter/metabolismo , Técnicas de Cocultivo , Medios de Cultivo/química , Etanol/metabolismo , Fermentación , Ácido Láctico/metabolismo , Limosilactobacillus fermentum/crecimiento & desarrollo , Limosilactobacillus fermentum/metabolismo , Manitol/metabolismo , Oxidación-Reducción , Saccharomyces cerevisiae/crecimiento & desarrollo , Saccharomyces cerevisiae/metabolismo
4.
Appl Environ Microbiol ; 80(1): 204-17, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24141124

RESUMEN

Arabinoxylan oligosaccharides (AXOS) are prebiotic carbohydrates with promising health-promoting properties that stimulate the activity of specific colon bacteria, in particular bifidobacteria. However, the mechanisms by which bifidobacterial strains break down these compounds in the colon is still unknown. This study investigates AXOS consumption of a large number of bifidobacterial strains (36), belonging to 11 different species, systematically. To determine their degradation mechanisms, all strains were grown on a mixture of arabinose and xylose, xylo-oligosaccharides, and complex AXOS molecules as the sole added energy sources. Based on principal component and cluster analyses of their different arabinose substituent and/or xylose backbone consumption patterns, five clusters that were species independent could be distinguished among the bifidobacterial strains tested. In parallel, the strains were screened for the presence of genes encoding several putative AXOS-degrading enzymes, but no clear-cut correlation could be made with the different degradation mechanisms. The intra- and interspecies differences in the consumption patterns of AXOS indicate that bifidobacterial strains could avoid competition among each other or even could cooperate jointly to degrade these complex prebiotics. The knowledge gained on the AXOS degradation mechanisms in bifidobacteria can be of importance in the rational design of prebiotics with tailor-made composition and thus increased specificity in the colon.


Asunto(s)
Bifidobacterium/metabolismo , Oligosacáridos/metabolismo , Xilanos/metabolismo , Bifidobacterium/enzimología , ADN Bacteriano/química , ADN Bacteriano/genética , Metabolismo Energético , Datos de Secuencia Molecular , Análisis de Secuencia de ADN , Xilosidasas/genética , Xilosidasas/metabolismo
5.
FEMS Microbiol Ecol ; 100(9)2024 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-39169462

RESUMEN

Candida species primarily exist as harmless commensals in the gastrointestinal tract of warm-blooded animals. However, they can also cause life-threatening infections, which are often associated with gut microbial dysbiosis. Identifying the microbial actors that restrict Candida to commensalism remains a significant challenge. In vitro models could enable a mechanistic study of the interactions between Candida and simulated colon microbiomes. Therefore, this study aimed to elucidate the spatial and temporal colonization kinetics of specific Candida, including C. albicans, C. tropicalis, and C. parapsilosis, and their relative Nakaseomyces glabratus, by using an adapted SHIME® model, simulating the ileum, and proximal and distal colons. We monitored fungal and bacterial colonization kinetics under conditions of eubiosis (commensal lifestyle) and antibiotic-induced dysbiosis (pathogenic lifestyle). Our findings highlighted the variability in the colonization potential of Candida species across different intestinal regions. The ileum compartment proved to be the most favourable environment for C. albicans and C. parapsilosis under conditions of eubiosis. Antibiotic-induced dysbiosis resulted in resurgence of opportunistic Candida species, especially C. tropicalis and C. albicans. Future research should focus on identifying specific bacterial species influencing Candida colonization resistance and explore the long-term effects of antibiotics on the mycobiome and bacteriome.


Asunto(s)
Candida , Disbiosis , Microbioma Gastrointestinal , Íleon , Humanos , Candida/efectos de los fármacos , Candida/crecimiento & desarrollo , Íleon/microbiología , Íleon/efectos de los fármacos , Disbiosis/microbiología , Microbioma Gastrointestinal/efectos de los fármacos , Tracto Gastrointestinal/microbiología , Antibacterianos/farmacología , Antibacterianos/efectos adversos , Modelos Biológicos , Mucosa Intestinal/microbiología
6.
J Control Release ; 369: 630-641, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38599548

RESUMEN

Successful treatment of ulcerative colitis (UC) is highly dependent on several parameters, including dosing regimen and the ability to deliver drugs to the disease site. In this study two strategies for delivering mesalazine (5-aminosalicylic acid, 5-ASA) to the colon were compared in an advanced in vitro model of the human gastrointestinal (GI) tract, the SHIME® system. Herein, a prodrug strategy employing bacteria-mediated drug release (sulfasalazine, Azulfidine®) was evaluated alongside a formulation strategy that utilised pH and bacteria-mediated release (5-ASA, Octasa® 1600 mg). SHIME® experiments were performed simulating both the GI physiology and colonic microbiota under healthy and inflammatory bowel disease (IBD) conditions, to study the impact of the disease state and ileal pH variability on colonic 5-ASA delivery. In addition, the effects of the products on the colonic microbiome were investigated by monitoring bacterial growth and metabolites. Results demonstrated that both the prodrug and formulation approaches resulted in a similar percentage of 5-ASA recovery under healthy conditions. On the contrary, during experiments simulating the GI physiology and microbiome of IBD patients (the target population) the formulation strategy resulted in a higher proportion of 5-ASA delivery to the colonic region as compared to the prodrug approach (P < 0.0001). Interestingly, the two products had distinct effects on the synthesis of key bacterial metabolites, such as lactate and short chain fatty acids, which varied according to disease state and ileal pH variability. Further, both 5-ASA and sulfasalazine significantly reduced the growth of the faecal microbiota sourced from six healthy humans. The findings support that the approach selected for colonic drug delivery could significantly influence the effectiveness of UC treatment, and highlight that drugs licensed for UC may differentially impact the growth and functioning of the colonic microbiota.


Asunto(s)
Antiinflamatorios no Esteroideos , Colon , Microbioma Gastrointestinal , Mesalamina , Sulfasalazina , Mesalamina/administración & dosificación , Mesalamina/farmacología , Humanos , Colon/microbiología , Colon/metabolismo , Colon/efectos de los fármacos , Microbioma Gastrointestinal/efectos de los fármacos , Antiinflamatorios no Esteroideos/administración & dosificación , Antiinflamatorios no Esteroideos/farmacología , Sulfasalazina/administración & dosificación , Profármacos/administración & dosificación , Sistemas de Liberación de Medicamentos , Colitis Ulcerosa/tratamiento farmacológico , Colitis Ulcerosa/microbiología , Concentración de Iones de Hidrógeno , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Enfermedades Inflamatorias del Intestino/microbiología , Liberación de Fármacos
7.
J Control Release ; 369: 163-178, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38521168

RESUMEN

The production of short chain fatty acids (SCFAs) by the colonic microbiome has numerous benefits for human health, including maintenance of epithelial barrier function, suppression of colitis, and protection against carcinogenesis. Despite the therapeutic potential, there is currently no optimal approach for elevating the colonic microbiome's synthesis of SCFAs. In this study, poly(D,l-lactide-co-glycolide) (PLGA) was investigated for this application, as it was hypothesised that the colonic microbiota would metabolise PLGA to its lactate monomers, which would promote the resident microbiota's synthesis of SCFAs. Two grades of spray dried PLGA, alongside a lactate bolus control, were screened in an advanced model of the human colon, known as the M-SHIME® system. Whilst the high molecular weight (Mw) grade of PLGA was stable in the presence of the microbiota sourced from three healthy humans, the low Mw PLGA (PLGA 2) was found to be metabolised. This microbial degradation led to sustained release of lactate over 48 h and increased concentrations of the SCFAs propionate and butyrate. Further, microbial synthesis of harmful ammonium was significantly reduced compared to untreated controls. Interestingly, both types of PLGA were found to influence the composition of the luminal and mucosal microbiota in a donor-specific manner. An in vitro model of an inflamed colonic epithelium also showed the polymer to affect the expression of pro- and anti-inflammatory markers, such as interleukins 8 and 10. The findings of this study reveal PLGA's sensitivity to enzymatic metabolism in the gut, which could be harnessed for therapeutic elevation of colonic SCFAs.


Asunto(s)
Ácidos Grasos Volátiles , Microbioma Gastrointestinal , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Humanos , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Microbioma Gastrointestinal/efectos de los fármacos , Ácidos Grasos Volátiles/metabolismo , Colon/metabolismo , Colon/microbiología , Ácido Láctico/metabolismo , Masculino , Adulto , Femenino
8.
Anal Chem ; 85(10): 4982-90, 2013 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-23541153

RESUMEN

Arabinoxylan-oligosaccharides (AXOS) are a new class of prebiotics with promising health-promoting characteristics. However, the mechanism by which bacteria break down these compounds in the colon is still uncharacterized, due to their structural complexity. A new analytical method that offers structural information was developed to characterize AXOS degradation during fermentation. The method was based on the simultaneous determination of arabinose, xylose, xylo-oligosaccharides (XOS), and AXOS by applying high-performance anion-exchange chromatography with pulsed amperometric detection. To study the structural features of AXOS in solution without the use of spectroscopic techniques or standards, enzymatic-based reference degradation chromatograms were generated based on enzymes with known specificity. The new method for fingerprinting showed to be a powerful and fast tool to study AXOS degradation with high repeatability with respect to peak area, peak width at half height, and retention time (respective relative standard deviations of ≤3.1%, 2.8%, and 0.8%). This method was successfully applied to study the degradation kinetics of AXOS in a complex fermentation medium by Bifidobacterium longum LMG 11047. The results showed that this strain could use both the arabinose side chains and xylose backbones up to xylotetraose. The characterization of the degradation abilities of AXOS by colon bacteria will allow a better understanding of the beneficial effects of these prebiotics. Furthermore, if the appropriate enzymes are available to design the reference degradation chromatograms, this new method for the qualitative fingerprinting of AXOS breakdown can also be applied for the breakdown of other complex oligosaccharides and polysaccharides.


Asunto(s)
Cromatografía por Intercambio Iónico/métodos , Medios de Cultivo , Fermentación , Oligosacáridos/aislamiento & purificación , Oligosacáridos/metabolismo , Prebióticos/microbiología , Xilanos/química , Bifidobacterium/crecimiento & desarrollo , Bifidobacterium/metabolismo , Electroquímica , Oligosacáridos/química , Reproducibilidad de los Resultados , Factores de Tiempo
9.
Br J Nutr ; 110(6): 998-1011, 2013 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-23507010

RESUMEN

In vitro experiments have shown that isolated human gut bacteria are able to metabolise PUFA into conjugated PUFA like conjugated linoleic acids (CLA). The hypothesis of the present paper was that high-fat (HF) diet feeding and supplementation with fermentable carbohydrates that have prebiotic properties modulate the in vivo production of CLA by the mouse gut microbiota. Mice were treated for 4 weeks as follows: control (CT) groups were fed a standard diet; HF groups were fed a HF diet rich in linoleic acid (18 : 2n-6); the third groups were fed with the HF diet supplemented with either inulin-type fructans (HF-ITF) or arabinoxylans (HF-Ax). HF diet feeding increased rumenic acid (cis-9,trans-11-18 : 2 CLA) content both in the caecal and liver tissues compared with the CT groups. ITF supplementation had no major effect compared with the HF diet whereas Ax supplementation increased further rumenic acid (cis-9,trans-11-18 : 2 CLA) in the caecal tissue. These differences between both prebiotics may be linked to the high fat-binding capacity of Ax that provides more substrates for bacterial metabolism and to differential modulation of the gut microbiota (specific increase in Roseburia spp. in HF-Ax v. HF). In conclusion, these experiments supply the proof of concept that the mouse gut microbiota produces CLA in vivo, with consequences on the level of CLA in the caecal and liver tissues. We postulate that the CLA-producing bacteria could be a mediator to consider in the metabolic effects of both HF diet feeding and prebiotic supplementation.


Asunto(s)
Bacterias/efectos de los fármacos , Carbohidratos/química , Grasas de la Dieta/farmacología , Intestinos/microbiología , Ácidos Linoleicos Conjugados/metabolismo , Animales , Metabolismo de los Hidratos de Carbono , Grasas de la Dieta/administración & dosificación , Ácidos Grasos Omega-6/metabolismo , Fermentación , Regulación Enzimológica de la Expresión Génica , Intestinos/enzimología , Hígado/enzimología , Masculino , Ratones , Ratones Endogámicos C57BL , Prebióticos , Estearoil-CoA Desaturasa/genética , Estearoil-CoA Desaturasa/metabolismo
10.
J Fungi (Basel) ; 9(9)2023 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-37754985

RESUMEN

From the estimated 2.2 to 3.8 million fungal species existing on Earth, only a minor fraction actively colonizes the human gastrointestinal tract. In fact, these fungi only represent 0.1% of the gastrointestinal biosphere. Despite their low abundance, fungi play dual roles in human health-both beneficial and detrimental. Fungal infections are often associated with bacterial dysbiosis following antibiotic use, yet our understanding of gut fungi-bacteria interactions remains limited. Here, we used the SHIME® gut model to explore the colonization of human fecal-derived fungi across gastrointestinal compartments. We accounted for the high inter-individual microbial diversity by using fecal samples from healthy adults, healthy babies, and Crohn's disease patients. Using quantitative Polymerase Chain Reaction and targeted next-generation sequencing, we demonstrated that SHIME®-colonized mycobiomes change upon loss of transient colonizers. In addition, SHIME® reactors from Crohn's disease patients contained comparable bacterial levels as healthy adults but higher fungal concentrations, indicating unpredictable correlations between fungal levels and total bacterial counts. Our findings rather link higher bacterial α-diversity to limited fungal growth, tied to colonization resistance. Hence, while healthy individuals had fewer fungi engrafting the colonic reactors, low α-diversity in impaired (Crohn's disease patients) or immature (babies) microbiota was associated with greater fungal abundance. To validate, antibiotic-treated healthy colonic microbiomes demonstrated increased fungal colonization susceptibility, and bacterial taxa that were negatively correlated with fungal expansion were identified. In summary, fungal colonization varied individually and transiently, and bacterial resistance to fungal overgrowth was more related with specific bacterial genera than total bacterial load. This study sheds light on fungal-bacterial dynamics in the human gut.

11.
Int J Pharm X ; 5: 100177, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37396621

RESUMEN

This study aimed at developing the Diamod® as a dynamic gastrointestinal transfer model with physically interconnected permeation. The Diamod® was validated by studying the impact of the intraluminal dilution of a cyclodextrin-based itraconazole solution and the negative food effect for indinavir sulfate for which clinical data are available demonstrating that the systemic exposure was strongly mediated by interconnected solubility, precipitation, and permeation processes. The Diamod® accurately simulated the impact of water intake on the gastrointestinal behavior of a Sporanox® solution. Water intake significantly decreased the duodenal solute concentrations of itraconazole as compared to no intake of water. Despite this duodenal behavior the amount of permeated itraconazole was not affected by water intake as observed in vivo. Next to this, the Diamod® accurately simulated the negative food effect for indinavir sulfate. Different fasted and fed state experiments demonstrated that this negative food effect was mediated by an increased stomach pH, entrapment of indinavir in colloidal structures and the slower gastric emptying of indinavir under fed state conditions. Therefore, it can be concluded that the Diamod® is a useful in vitro model to mechanistically study the gastrointestinal performance of drugs.

12.
Microbiol Spectr ; : e0393322, 2023 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-37750706

RESUMEN

Clostridioides difficile infection (CDI) is a major health concern and one of the leading causes of hospital-acquired diarrhea in many countries. C. difficile infection is challenging to treat as C. difficile is resistant to multiple antibiotics. Alternative solutions are needed as conventional treatment with broad-spectrum antibiotics often leads to recurrent CDI. Recent studies have shown that specific microbiota-based therapeutics such as bile acids (BAs) are promising approaches to treat CDI. Clostridium scindens encodes the bile acid-induced (bai) operon that carries out 7-alpha-dehydroxylation of liver-derived primary BAs to secondary BAs. This biotransformation is thought to increase the antibacterial effects of BAs on C. difficile. Here, we used an automated multistage fermentor to study the antibacterial actions of C. scindens and BAs on C. difficile in the presence/absence of a gut microbial community derived from healthy human donor fecal microbiota. We observed that C. scindens inhibited C. difficile growth when the medium was supplemented with primary BAs. Transcriptomic analysis indicated upregulation of C. scindens bai operon and suppressed expression of C. difficile exotoxins that mediate CDI. We also observed BA-independent antibacterial activity of the secretome from C. scindens cultured overnight in a medium without supplementary primary BAs, which suppressed growth and exotoxin expression in C. difficile mono-culture. Further investigation of the molecular basis of our observation could lead to a more specific treatment for CDI than current approaches. IMPORTANCE There is an urgent need for new approaches to replace the available treatment options against Clostridioides difficile infection (CDI). Our novel work reports a bile acid-independent reduction of C. difficile growth and virulence gene expression by the secretome of Clostridium scindens. This potential treatment combined with other antimicrobial strategies could facilitate the development of alternative therapies in anticipation of CDI and in turn reduce the risk of antimicrobial resistance.

13.
Front Microbiol ; 13: 1054061, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-37008301

RESUMEN

The human gastrointestinal tract consists of different regions, each characterized by a distinct physiology, anatomy, and microbial community. While the colonic microbiota has received a lot of attention in recent research projects, little is known about the small intestinal microbiota and its interactions with ingested compounds, primarily due to the inaccessibility of this region in vivo. This study therefore aimed to develop and validate a dynamic, long-term simulation of the ileal microbiota using the SHIME®-technology. Essential parameters were identified and optimized from a screening experiment testing different inoculation strategies, nutritional media, and environmental parameters over an 18-day period. Subjecting a synthetic bacterial consortium to the selected conditions resulted in a stable microbiota that was representative in terms of abundance [8.81 ± 0.12 log (cells/ml)], composition and function. Indeed, the observed community mainly consisted of the genera Streptococcus, Veillonella, Enterococcus, Lactobacillus, and Clostridium (qPCR and 16S rRNA gene targeted Illumina sequencing), while nutrient administration boosted lactate production followed by cross-feeding interactions towards acetate and propionate. Furthermore, similarly as in vivo, bile salts were only partially deconjugated and only marginally converted into secondary bile salts. After confirming reproducibility of the small intestinal microbiota model, it was integrated into the established M-SHIME® where it further increased the compositional relevance of the colonic community. This long-term in vitro model provides a representative simulation of the ileal bacterial community, facilitating research of the ileum microbiota dynamics and activity when, for example, supplemented with microbial or diet components. Furthermore, integration of this present in vitro simulation increases the biological relevance of the current M-SHIME® technology.

14.
Appl Environ Microbiol ; 77(18): 6694-8, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21803901

RESUMEN

Among various lactic acid bacterial strains tested, cocoa-specific strains of Lactobacillus fermentum were best adapted to the cocoa pulp ecosystem. They fermented glucose to lactic acid and acetic acid, reduced fructose to mannitol, and converted citric acid into lactic acid and 2,3-butanediol.


Asunto(s)
Cacao/metabolismo , Fabaceae/metabolismo , Fermentación , Manipulación de Alimentos , Ácido Láctico/metabolismo , Lactobacillus/metabolismo , Ácido Acético/metabolismo , Ácido Cítrico/metabolismo , Fructosa/metabolismo , Glucosa/metabolismo , Leuconostoc/metabolismo , Manitol/metabolismo , Oxidación-Reducción , Weissella/metabolismo
15.
Pathogens ; 10(8)2021 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-34451391

RESUMEN

Clostridioides difficile (formerly Clostridium difficile) infection (CDI) is one of the most common hospital-acquired infections, which is often triggered by a dysbiosed indigenous gut microbiota (e.g., upon antibiotic therapy). Symptoms can be as severe as life-threatening colitis. The current study assessed the antipathogenic potential of human milk oligosaccharides (HMOs), i.e., 2'-O-fucosyllactose (2'FL), lacto-N-neotetraose (LNnT), and a combination thereof (MIX), against C. difficile ATCC 9689 using in vitro gut models that allowed the evaluation of both direct and, upon microbiota modulation, indirect effects. During a first 48 h fecal batch study, dysbiosis and CDI were induced by dilution of the fecal inoculum. For each of the three donors tested, C. difficile levels strongly decreased (with >4 log CFU/mL) upon treatment with 2'FL, LNnT and MIX versus untreated blanks, coinciding with increased acetate/Bifidobacteriaceae levels. Interindividual differences among donors at an intermediate time point suggested that the antimicrobial effect was microbiota-mediated rather than being a direct effect of the HMOs. During a subsequent 11 week study with the PathogutTM model (specific application of the Simulator of the Human Intestinal Microbial Ecosystem (SHIME®)), dysbiosis and CDI were induced by clindamycin (CLI) treatment. Vancomycin (VNC) treatment cured CDI, but the further dysbiosis of the indigenous microbiota likely contributed to CDI recurrence. Upon co-supplementation with VNC, both 2'FL and MIX boosted microbial activity (acetate and to lesser extent propionate/butyrate). Moreover, 2'FL avoided CDI recurrence, potentially because of increased secondary bile acid production. Overall, while not elucidating the exact antipathogenic mechanisms-of-action, the current study highlights the potential of HMOs to combat CDI recurrence, help the gut microbial community recover after antibiotic treatment, and hence counteract the adverse effects of antibiotic therapies.

16.
Int J Pharm X ; 3: 100087, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34977556

RESUMEN

We report here the potential role of a 4-strain probiotic suspension for use with patients with Parkinson's disease (PD). Stool samples from a group of three patients with diagnosed PD were used to create microbiotas in an in-vitro gut model. The effects of dosing with an oral probiotic suspension (Symprove) on bacterial composition and metabolic activity in the microbiotas was evaluated over 48 h and compared with healthy controls. Additionally, the effect of probiotic dosing on epithelial tight-junction integrity, production of inflammatory markers and wound healing were evaluated in cell culture models. In general, the relative proportions of the main bacterial phyla in the microbiotas of PD patients differed from those of healthy subjects, with levels of Firmicutes raised and levels of Bacteroidetes reduced. Dosing with probiotic resulted in a change in bacterial composition in the microbiotas over a 48 h period. Several other indicators of gut health changed upon dosing with the probiotic; production of short chain fatty acids (SCFAs) and lactate was stimulated, levels of anti-inflammatory cytokines (IL-6, IL-10) increased and levels of pro-inflammatory cytokines and chemokines (MCP-1 and IL-8) decreased. Tight junction integrity was seen to improve with probiotic dosing and wound healing was seen to occur faster than a control. The data suggest that if development and/or progression of PD is influenced by gut microbiota dysbiosis then supplementation of the diet with a properly formulated probiotic may be a useful adjunct to standard treatment in clinic.

17.
Int J Pharm ; 587: 119648, 2020 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-32679260

RESUMEN

Symprove, a multi-strain probiotic, has been shown to exert a mild anti-inflammatory effect in patients with ulcerative colitis (UC). We examined stool samples from 3 patients with UC in order to create microbiotas in an in-vitro gut model. The effects of Symprove on bacterial diversity and metabolic activity in the microbiotas was evaluated over 48 h. In addition, the influence of probiotic dosing on epithelial tight-junction integrity, production of inflammatory markers and wound healing were evaluated in cell culture models. The relative proportions of the main bacterial phyla in UC patients differed from those of healthy subjects studied previously; levels of Firmicutes were lowered and levels of Bacteroidetes were raised. Addition of Symprove changed the bacterial composition in the microbiotas over a 48 h period. Several other factors generally implicated in good gut health changed after dosing with probiotic; production of short chain fatty acids (SCFAs) and lactate was stimulated, levels of anti-inflammatory cytokines (IL-6, IL-10) increased, levels of pro-inflammatory cytokines and chemokines (MCP-1 and IL-8) decreased, epithelial tight junction integrity improved and wound healing occurred faster than a control. The results imply it is not the simple addition of probiotic bacteria that improves gut health. Rather, the probiotic bacteria generate lactate, which then stimulates growth of commensal gut bacteria, raising SCFA levels (particularly butyrate). The increased butyrate concentration positively influences inflammation response and time of wound healing.


Asunto(s)
Colitis Ulcerosa , Microbioma Gastrointestinal , Probióticos , Colitis Ulcerosa/tratamiento farmacológico , Ácidos Grasos Volátiles , Humanos
18.
J Agric Food Chem ; 68(46): 13102-13110, 2020 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-31909618

RESUMEN

The current study evaluated the effect of five yeast-derived formulations (T1-T5) on microbial metabolism and composition of the canine and feline gut microbiota using a novel in vitro colonic incubation approach. This novel in vitro model allowed for growth of the entire spectrum of dog- and cat-derived bacteria from the inoculum, thus offering an excellent platform to evaluate effects of nutritional interventions on the gut microbiota. Further, yeast-derived ingredients differentially increased production of acetate, propionate, butyrate, ammonium, and branched short-chain fatty acids, with T5 and T1 consistently stimulating propionate and butyrate, respectively. 16S-targeted Illumina sequencing coupled with flow cytometry provided unprecedented high-resolution quantitative insights in canine and feline microbiota modulation by yeast-derived ingredients, revealing that effects on propionate production were related to Prevotellaceae, Tannerellaceae, Bacteroidaceae, and Veillonellaceae members, while effects on butyrate production were related to Erysipelotrichaceae, Lachnospiraceae, Ruminococcaceae, and Fusobacteriaceae. Overall, these findings strengthen the health-promoting potential of yeast-derived ingredients.


Asunto(s)
Bacterias/metabolismo , Colon/microbiología , Ácidos Grasos Volátiles/metabolismo , Microbioma Gastrointestinal , Levaduras/química , Animales , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Gatos , Suplementos Dietéticos/análisis , Perros , Fermentación , Levaduras/metabolismo
19.
Biomimetics (Basel) ; 4(3)2019 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-31547365

RESUMEN

This article presents some application of the morphing technology for aerodynamic performance improvement of turboprop regional aircraft. It summarizes the results obtained in the framework of the Clean Sky 2 AIRGREEN2 program for the development and application of dedicated morphing devices for take-off and landing, and their uses in off design conditions. The wing of the reference aircraft configuration considers Natural Laminar Flow (NLF) characteristics. A deformable leading edge morphing device ("droop nose") and a multi-functional segmented flap system have been considered. For the droop nose, the use of the deformable compliant structure was considered, as it allows a "clean" leading edge when not used, which is mandatory to keep natural laminar flow (NLF) properties at cruise. The use of a segmented flap makes it possible to avoid external flap track fairings, which will lead to performance improvement at cruise. An integrated tracking mechanism is used to set the flap at its take-off optimum setting, and, then, morphing is applied in order to obtain a high-performance level for landing. Lastly, some performance improvements can be obtained in climb conditions by using the last segment of the flap system to modify the load distribution on the wing in order to recover some extended laminar flow on the wing upper surface.

20.
Int J Pharm ; 555: 1-10, 2019 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-30445175

RESUMEN

Poorly formulated probiotic supplements intended for oral administration often fail to protect bacteria from the challenges of human digestion, meaning bacteria do not reach the small intestine in a viable state. As a result, the ability of probiotics to influence the human gut microbiota has not been proven. Here we show how (i) considered formulation of an aqueous probiotic suspension can facilitate delivery of viable probiotic bacteria to the gut and (ii) quantitate the effect of colonisation and proliferation of specific probiotic species on the human gut microbiota, using an in-vitro gut model. Our data revealed immediate colonisation and growth of three probiotic species in the luminal and mucosal compartments of the proximal and distal colon, and growth of a fourth species in the luminal proximal colon, leading to higher proximal and distal colonic lactate concentrations. The lactate stimulated growth of lactate-consuming bacteria, altering the bacterial diversity of the microbiota and resulting in increased short-chain fatty acid production, especially butyrate. Additionally, an immunomodulatory effect of the probiotics was seen; production of anti-inflammatory cytokines (IL-6 and IL-10) was increased and production of inflammatory chemokines (MCP-1, CXCL 10 and IL-8.) was reduced. The results indicate that the probiotic species alone do not result in a clinical effect; rather, they facilitate modulation of the gut microbiota composition and metabolic activity thereby influencing the immune response.


Asunto(s)
Butiratos/metabolismo , Colon/metabolismo , Microbioma Gastrointestinal/inmunología , Probióticos/administración & dosificación , Adulto , Bacterias/metabolismo , Células CACO-2 , Quimiocinas/inmunología , Colon/inmunología , Colon/microbiología , Ácidos Grasos Volátiles/metabolismo , Humanos , Interleucina-10/inmunología , Interleucina-6/inmunología , Ácido Láctico/metabolismo , Probióticos/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA