Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros

Banco de datos
Tipo de estudio
Tipo del documento
Intervalo de año de publicación
1.
Arch Microbiol ; 206(7): 315, 2024 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-38904672

RESUMEN

Exploring probiotics for their crosstalk with the host microbiome through the fermentation of non-digestible dietary fibers (prebiotics) for their potential metabolic end-products, particularly short-chain fatty acids (SCFAs), is important for understanding the endogenous host-gut microbe interaction. This study was aimed at a systematic comparison of commercially available probiotics to understand their synergistic role with specific prebiotics in SCFAs production both in vitro and in the ex vivo gut microcosm model. Probiotic strains isolated from pharmacy products including Lactobacillus sporogenes (strain not labeled), Lactobacillus rhamnosus GG (ATCC53103), Streptococcus faecalis (T-110 JPC), Bacillus mesentericus (TO-AJPC), Bacillus clausii (SIN) and Saccharomyces boulardii (CNCM I-745) were assessed for their probiotic traits including survival, antibiotic susceptibility, and antibacterial activity against pathogenic strains. Our results showed that the microorganisms under study had strain-specific abilities to persist in human gastrointestinal conditions and varied anti-infective efficacy and antibiotic susceptibility. The probiotic strains displayed variation in the utilization of six different prebiotic substrates for their growth under aerobic and anaerobic conditions. Their prebiotic scores (PS) revealed which were the most suitable prebiotic carbohydrates for the growth of each strain and suggested xylooligosaccharide (XOS) was the poorest utilized among all. HPLC analysis revealed a versatile pattern of SCFAs produced as end-products of prebiotic fermentation by the strains which was influenced by growth conditions. Selected synbiotic (prebiotic and probiotic) combinations showing high PS and high total SCFAs production were tested in an ex vivo human gut microcosm model. Interestingly, significantly higher butyrate and propionate production was found only when synbiotics were applied as against when individual probiotic or prebiotics were applied alone. qRT-PCR analysis with specific primers showed that there was a significant increase in the abundance of lactobacilli and bifidobacteria with synbiotic blends compared to pre-, or probiotics alone. In conclusion, this work presents findings to suggest prebiotic combinations with different well-established probiotic strains that may be useful for developing effective synbiotic blends.


Asunto(s)
Ácidos Grasos Volátiles , Microbioma Gastrointestinal , Prebióticos , Probióticos , Simbióticos , Humanos , Probióticos/administración & dosificación , Ácidos Grasos Volátiles/metabolismo , Antibacterianos/farmacología , Fermentación , Tracto Gastrointestinal/microbiología , Lactobacillus/metabolismo , Bacterias/clasificación , Bacterias/metabolismo , Bacterias/genética , Bacterias/aislamiento & purificación , Saccharomyces boulardii/metabolismo
2.
Appl Microbiol Biotechnol ; 108(1): 153, 2024 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-38240846

RESUMEN

Evolutionary engineering experiments, in combination with omics technologies, revealed genetic markers underpinning the molecular mechanisms behind acetic acid stress tolerance in the probiotic yeast Saccharomyces cerevisiae var. boulardii. Here, compared to the ancestral Ent strain, evolved yeast strains could quickly adapt to high acetic acid levels (7 g/L) and displayed a shorter lag phase of growth. Bioinformatic-aided whole-genome sequencing identified genetic changes associated with enhanced strain robustness to acetic acid: a duplicated sequence in the essential endocytotic PAN1 gene, mutations in a cell wall mannoprotein (dan4Thr192del), a lipid and fatty acid transcription factor (oaf1Ser57Pro) and a thiamine biosynthetic enzyme (thi13Thr332Ala). Induction of PAN1 and its associated endocytic complex SLA1 and END3 genes was observed following acetic acid treatment in the evolved-resistant strain when compared to the ancestral strain. Genome-wide transcriptomic analysis of the evolved Ent acid-resistant strain (Ent ev16) also revealed a dramatic rewiring of gene expression among genes associated with cellular transport, metabolism, oxidative stress response, biosynthesis/organization of the cell wall, and cell membrane. Some evolved strains also displayed better growth at high acetic acid concentrations and exhibited adaptive metabolic profiles with altered levels of secreted ethanol (4.0-6.4% decrease), glycerol (31.4-78.5% increase), and acetic acid (53.0-60.3% increase) when compared to the ancestral strain. Overall, duplication/mutations and transcriptional alterations are key mechanisms driving improved acetic acid tolerance in probiotic strains. We successfully used adaptive evolutionary engineering to rapidly and effectively elucidate the molecular mechanisms behind important industrial traits to obtain robust probiotic yeast strains for myriad biotechnological applications. KEY POINTS: •Acetic acid adaptation of evolutionary engineered robust probiotic yeast S. boulardii •Enterol ev16 with altered genetic and transcriptomic profiles survives in up to 7 g/L acetic acid •Improved acetic acid tolerance of S. boulardii ev16 with mutated PAN1, DAN4, OAF1, and THI13 genes.


Asunto(s)
Probióticos , Saccharomyces boulardii , Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae/metabolismo , Ácido Acético/metabolismo , Saccharomyces boulardii/genética , Saccharomyces boulardii/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Probióticos/metabolismo , Biomarcadores/metabolismo , Proteínas de Unión al ADN/metabolismo , Factores de Transcripción/metabolismo
3.
Crit Rev Food Sci Nutr ; 63(4): 457-485, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-34254862

RESUMEN

Saccharomyces cerevisiae var. boulardii (S. boulardii) has been isolated from lychee (Litchi chinensis), mangosteen fruit, kombucha, and dairy products like kefir. Dairy products containing S. boulardii have been revealed to possess potential probiotic activities owing to their ability to produce organic acids, essential enzymes, vitamins, and other important metabolites such as vanillic acid, phenyl ethyl alcohol, and erythromycin. S. boulardii has a wide spectrum of anti-carcinogenic, antibacterial antiviral, and antioxidant activity, and is known to reduce serum cholesterol levels. However, this yeast has mainly been prescribed for prophylaxis treatment of gastrointestinal infectious diseases, and stimulating the immune system in a number of commercially available products. The present comprehensive review article reviews the properties of S. boulardii related to their use in fermented dairy foods as a probiotic microorganism or starter culture. Technical aspects regarding the integration of this yeast into the dairy foods matrix its health advantages, therapeutic functions, microencapsulation, and viability in harsh conditions, and safety aspects are highlighted.


Asunto(s)
Probióticos , Saccharomyces boulardii , Saccharomyces cerevisiae/metabolismo , Saccharomyces boulardii/metabolismo , Probióticos/metabolismo , Antioxidantes/metabolismo , Productos Lácteos
4.
Microb Cell Fact ; 22(1): 109, 2023 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-37287064

RESUMEN

The probiotic yeast Saccharomyces boulardii (Sb) is a promising chassis to deliver therapeutic proteins to the gut due to Sb's innate therapeutic properties, resistance to phage and antibiotics, and high protein secretion capacity. To maintain therapeutic efficacy in the context of challenges such as washout, low rates of diffusion, weak target binding, and/or high rates of proteolysis, it is desirable to engineer Sb strains with enhanced levels of protein secretion. In this work, we explored genetic modifications in both cis- (i.e. to the expression cassette of the secreted protein) and trans- (i.e. to the Sb genome) that enhance Sb's ability to secrete proteins, taking a Clostridioides difficile Toxin A neutralizing peptide (NPA) as our model therapeutic. First, by modulating the copy number of the NPA expression cassette, we found NPA concentrations in the supernatant could be varied by sixfold (76-458 mg/L) in microbioreactor fermentations. In the context of high NPA copy number, we found a previously-developed collection of native and synthetic secretion signals could further tune NPA secretion between 121 and 463 mg/L. Then, guided by prior knowledge of S. cerevisiae's secretion mechanisms, we generated a library of homozygous single gene deletion strains, the most productive of which achieved 2297 mg/L secretory production of NPA. We then expanded on this library by performing combinatorial gene deletions, supplemented by proteomics experiments. We ultimately constructed a quadruple protease-deficient Sb strain that produces 5045 mg/L secretory NPA, an improvement of > tenfold over wild-type Sb. Overall, this work systematically explores a broad collection of engineering strategies to improve protein secretion in Sb and highlights the ability of proteomics to highlight under-explored mediators of this process. In doing so, we created a set of probiotic strains that are capable of delivering a wide range of protein titers and therefore furthers the ability of Sb to deliver therapeutics to the gut and other settings to which it is adapted.


Asunto(s)
Probióticos , Saccharomyces boulardii , Saccharomyces , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Saccharomyces boulardii/genética , Saccharomyces boulardii/metabolismo , Saccharomyces/genética , Saccharomyces/metabolismo , Probióticos/metabolismo , Endopeptidasas/metabolismo
5.
Genome Res ; 29(9): 1478-1494, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31467028

RESUMEN

The yeast Saccharomyces boulardii has been used worldwide as a popular, commercial probiotic, but the basis of its probiotic action remains obscure. It is considered conspecific with budding yeast Saccharomyces cerevisiae, which is generally used in classical food applications. They have an almost identical genome sequence, making the genetic basis of probiotic potency in S. boulardii puzzling. We now show that S. boulardii produces at 37°C unusually high levels of acetic acid, which is strongly inhibitory to bacterial growth in agar-well diffusion assays and could be vital for its unique application as a probiotic among yeasts. Using pooled-segregant whole-genome sequence analysis with S. boulardii and S. cerevisiae parent strains, we succeeded in mapping the underlying QTLs and identified mutant alleles of SDH1 and WHI2 as the causative alleles. Both genes contain a SNP unique to S. boulardii (sdh1 F317Y and whi2 S287*) and are fully responsible for its high acetic acid production. S. boulardii strains show different levels of acetic acid production, depending on the copy number of the whi2 S287* allele. Our results offer the first molecular explanation as to why S. boulardii could exert probiotic action as opposed to S. cerevisiae They reveal for the first time the molecular-genetic basis of a probiotic action-related trait in S. boulardii and show that antibacterial potency of a probiotic microorganism can be due to strain-specific mutations within the same species. We suggest that acquisition of antibacterial activity through medium acidification offered a selective advantage to S. boulardii in its ecological niche and for its application as a probiotic.


Asunto(s)
Ácido Acético/metabolismo , Sitios de Carácter Cuantitativo , Saccharomyces boulardii/crecimiento & desarrollo , Secuenciación Completa del Genoma/métodos , Antibacterianos/metabolismo , Variaciones en el Número de Copia de ADN , Calor , Polimorfismo de Nucleótido Simple , Probióticos/metabolismo , Saccharomyces boulardii/genética , Saccharomyces boulardii/metabolismo , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Succinato Deshidrogenasa/genética
6.
Pharmacol Res ; 181: 106291, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35690329

RESUMEN

Saccharomyces boulardii (Sb) is a widely used fungal probiotic in treating various digestive diseases, including irritable bowel syndrome (IBS). However, the specific mechanisms of Sb relieving IBS remain unclear. The abnormal serotonin transporter (SERT) / 5-hydroxytryptamine (5-HT) system could cause disordered gastrointestinal sensation and motility, which closely related to IBS pathogenesis. The aim of this study was to explore the effects and mechanisms of Sb on regulating gut motility. Sb supernatant (SbS) was administered to intestinal epithelial cells and mice. SbS upregulated SERT expression via enhancing heparin-binding epidermal growth factor (HB-EGF) release to activate epidermal growth factor receptor (EGFR). EGFR kinase inhibitor treatment or HB-EGF siRNA transfection in cells blocked SbS upregulating SERT. Consistently, SbS-treated mice presented inhibited gut motility, and EGFR activation and SERT upregulation were found. Moreover, 16 S rDNA sequence presented an evident decrease in Firmicutes / Bacteroidetes ratio in SbS group. In genus level, SbS reduced Escherichia_Shigella, Alistipes, Clostridium XlVa, and Saccharibacteria_genera_incertae_sedis, meanwhile, increased Parasutterella. The abundance of Saccharibacteria_genera_incertae_sedis positively correlated with defecation parameters and intestinal 5-HT content. Fecal microbiota transplantation showed that SbS could modulate gut microbiota to influence gut motility. Interestingly, elimination of gut microbiota with antibiotic cocktail did not entirely block SbS regulating gut motility. Furthermore, SbS administration to IBS-D mice significantly upregulated SERT and inhibited gut motility. In conclusion, SbS could upregulate SERT by EGFR activation, and modulate gut microbiota to inhibit gut motility. This finding would provide more evidence for the application of this yeast probiotic in IBS and other diarrheal disorders.


Asunto(s)
Microbioma Gastrointestinal , Síndrome del Colon Irritable , Probióticos , Saccharomyces boulardii , Animales , Bacterias/metabolismo , Receptores ErbB/metabolismo , Factor de Crecimiento Similar a EGF de Unión a Heparina/metabolismo , Ratones , Probióticos/farmacología , Saccharomyces boulardii/metabolismo , Serotonina/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo
7.
Microb Cell Fact ; 21(1): 204, 2022 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-36207743

RESUMEN

BACKGROUND: Saccharomyces boulardii is a probiotic yeast that exhibits antimicrobial and anti-toxin activities. Although S. boulardii has been clinically used for decades to treat gastrointestinal disorders, several studies have reported weak or no beneficial effects of S. boulardii administration in some cases. These conflicting results of S. boulardii efficacity may be due to nutrient deficiencies in the intestine that make it difficult for S. boulardii to maintain its metabolic activity. RESULTS: To enable S. boulardii to overcome any nutritional deficiencies in the intestine, we constructed a S. boulardii strain that could metabolize L-fucose, a major component of mucin in the gut epithelium. The fucU, fucI, fucK, and fucA from Escherichia coli and HXT4 from S. cerevisiae were overexpressed in S. boulardii. The engineered S. boulardii metabolized L-fucose and produced 1,2-propanediol under aerobic and anaerobic conditions. It also produced large amounts of 1,2-propanediol under strict anaerobic conditions. An in silico genome-scale metabolic model analysis was performed to simulate the growth of S. boulardii on L-fucose, and elementary flux modes were calculated to identify critical metabolic reactions for assimilating L-fucose. As a result, we found that the engineered S. boulardii consumes L-fucose via (S)-lactaldehyde-(S)-lactate-pyruvate pathway, which is highly oxygen dependent. CONCLUSION: To the best of our knowledge, this is the first study in which S. cerevisiae and S. boulardii strains capable of metabolizing L-fucose have been constructed. This strategy could be used to enhance the metabolic activity of S. boulardii and other probiotic microorganisms in the gut.


Asunto(s)
Probióticos , Saccharomyces boulardii , Animales , Escherichia coli , Fucosa/metabolismo , Lactatos/metabolismo , Mamíferos , Análisis de Flujos Metabólicos , Mucinas/metabolismo , Oxígeno/metabolismo , Probióticos/metabolismo , Propilenglicol/metabolismo , Piruvatos/metabolismo , Saccharomyces boulardii/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo
8.
Microb Cell Fact ; 20(1): 160, 2021 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-34407819

RESUMEN

BACKGROUND: Saccharomyces cerevisiae var. boulardii is a representative probiotic yeast that has been widely used in the food and pharmaceutical industries. However, S. boulardii has not been studied as a microbial cell factory for producing useful substances. Agarose, a major component of red macroalgae, can be depolymerized into neoagarooligosaccharides (NAOSs) by an endo-type ß-agarase. NAOSs, including neoagarotetraose (NeoDP4), are known to be health-benefiting substances owing to their prebiotic effect. Thus, NAOS production in the gut is required. In this study, the probiotic yeast S. boulardii was engineered to produce NAOSs by expressing an endo-type ß-agarase, BpGH16A, derived from a human gut bacterium Bacteroides plebeius. RESULTS: In total, four different signal peptides were compared in S. boulardii for protein (BpGH16A) secretion for the first time. The SED1 signal peptide derived from Saccharomyces cerevisiae was selected as optimal for extracellular production of NeoDP4 from agarose. Expression of BpGH16A was performed in two ways using the plasmid vector system and the clustered regularly interspaced short palindromic repeat (CRISPR)-Cas9 system. The production of NeoDP4 by engineered S. boulardii was verified and quantified. NeoDP4 was produced by S. boulardii engineered using the plasmid vector system and CRISPR-Cas9 at 1.86 and 0.80 g/L in a 72-h fermentation, respectively. CONCLUSIONS: This is the first report on NAOS production using the probiotic yeast S. boulardii. Our results suggest that S. boulardii can be considered a microbial cell factory to produce health-beneficial substances in the human gut.


Asunto(s)
Ingeniería Metabólica/métodos , Oligosacáridos/biosíntesis , Probióticos/metabolismo , Saccharomyces boulardii/metabolismo , Bacteroides/genética , Fermentación , Glicósido Hidrolasas/química , Glicósido Hidrolasas/genética , Humanos , Oligosacáridos/química , Oligosacáridos/genética , Saccharomyces boulardii/genética , Saccharomyces cerevisiae/clasificación , Sefarosa/metabolismo
9.
J Sci Food Agric ; 101(13): 5487-5497, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-33682152

RESUMEN

BACKGROUND: Non-diary beverages with probiotic properties are of great interest nowadays. In this research, we evaluated the suitability of carob kibbles in the manufacture of kvass. Kvass is a low-alcohol drink popular in Central and Eastern Europe and indicated as a potential non-diary beverage with probiotic properties. Therefore, the viability of probiotic strains of Lactobacillus plantarum and Saccharomyces boulardii during 4 weeks' storage in manufactured beverages was tested. RESULTS: Carob kibbles introduced significant amounts of phenolic compounds into kvasses, especially gallic acid (up to 117.45 ± 10.56 mg L-1 ), and improved antiradical activity up to 78% after fermentation. Moreover, fermentation efficiently reduced furfural and hydroxymethylfurfural content in samples up to 12.9% and 29.9%, respectively. Kvasses with rye malt extract possessed coffee-like, chocolate-like, roasted and caramel-like odours and a more bitter taste. Whereas kvass with carob kibbles was characterized by fruit-like odour and sweeter taste. Fermentation contributed to a creation as well as degradation of volatiles. L. plantarum exhibited higher general mortality during storage, whereas, in the case of S. boulardii, the viability was significantly higher regardless of the sample composition. CONCLUSION: This is the first study reporting the use of carob kibbles for kvass production. The obtained results showed that carob kibbles can replace rye malt extract, at least partially, in the production of kvass, giving to the product added health benefits. Moreover, S. boulardii is a better choice for production of kvass with probiotic properties. © 2021 Society of Chemical Industry.


Asunto(s)
Bebidas Alcohólicas/análisis , Fabaceae/microbiología , Lactobacillus plantarum/metabolismo , Probióticos/análisis , Saccharomyces boulardii/metabolismo , Adulto , Bebidas Alcohólicas/microbiología , Fabaceae/química , Fabaceae/metabolismo , Femenino , Fermentación , Microbiología de Alimentos , Galactanos/metabolismo , Humanos , Lactobacillus plantarum/crecimiento & desarrollo , Masculino , Mananos/metabolismo , Persona de Mediana Edad , Gomas de Plantas/metabolismo , Probióticos/metabolismo , Saccharomyces boulardii/crecimiento & desarrollo , Gusto
10.
Fungal Genet Biol ; 137: 103333, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31923554

RESUMEN

The presence of commensal fungal species in the human gut indicates that organisms from this kingdom have the potential to benefit the host as well. Saccharomyces boulardii, a yeast strain isolated about a hundred years ago, is the most well-characterized probiotic yeast. Though for the most part it genetically resembles Saccharomyces cerevisiae, specific phenotypic differences make it better suited for the gut microenvironment such as better acid and heat tolerance. Several studies using animal hosts suggest that S. boulardii can be used as a biotherapeutic in humans. Clinical trials indicate that it can alleviate symptoms from gastrointestinal (GI) tract infections to some extent, but further trials are needed to understand the full therapeutic potential of S. boulardii. Improvement on probiotic function using engineered yeast is an attractive future direction, though genome modification tools for use in S. boulardii have been limited until recently. However, some tools available for S. cerevisiae should be applicable for S. boulardii as well. In this review, we summarize the observed probiotic effect of this yeast and the state of the art for genome engineering tools that could help enhance its probiotic properties.


Asunto(s)
Probióticos/metabolismo , Probióticos/uso terapéutico , Saccharomyces boulardii/metabolismo , Animales , Humanos , Saccharomyces/genética , Saccharomyces/metabolismo , Saccharomyces boulardii/genética , Levaduras/genética , Levaduras/metabolismo
11.
Cytokine ; 125: 154791, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31401369

RESUMEN

BACKGROUND AND AIM: Chemotherapy drugs that act via Toll-like receptors (TLRs) can exacerbate mucosal injury through the production of cytokines. Intestinal mucositis can activate TLR2 and TLR4, resulting in the activation of NF-κB. Intestinal mucositis characterized by intense inflammation is the main side effect associated with 5-fluorouracil (5-FU) treatment. Saccharomyces boulardii CNCM I-745 (S.b) is a probiotic yeast used in the treatment of gastrointestinal disorders. The main objective of the study was to evaluate the effect of S.b treatment on the Toll-like/MyD88/NF-κB/MAPK pathway activated during intestinal mucositis and in Caco-2 cells treated with 5-FU. METHODS: The mice were divided into three groups: saline (control), saline + 5-FU, and 5-FU + S.b (1.6 × 1010 colony forming units/kg). After 3 days of S.b administration by gavage, the mice were euthanized and the jejunum and ileum were removed. In vitro, Caco2 cells were treated with 5-FU (1 mM) alone or in the presence of lipopolysaccharide (1 ng/ml). When indicated, cells were exposed to S.b. The jejunum/ileum samples and Caco2 cells were examined for the expression or concentration of the inflammatory components. RESULTS: Treatment with S.b modulated the expressions of TLR2, TLR4, MyD88, NF-κB, ERK1/2, phospho-p38, phospho-JNK, TNF-α, IL-1ß, and CXCL-1 in the jejunum/ileum and Caco2 cells following treatment with 5-FU. CONCLUSION: Toll-like/MyD88/NF-κB/MAPK pathway are activated during intestinal mucositis and their modulation by S.b suggests a novel and valuable therapeutic strategy for intestinal inflammation.


Asunto(s)
Citocinas/metabolismo , Fluorouracilo/farmacología , Mucositis/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , FN-kappa B/metabolismo , Probióticos/farmacología , Saccharomyces boulardii/metabolismo , Receptores Toll-Like/metabolismo , Animales , Células CACO-2 , Quimiocina CXCL1/genética , Quimiocina CXCL1/metabolismo , Citocinas/genética , Fluorouracilo/efectos adversos , Humanos , Íleon/metabolismo , Inmunohistoquímica , Inflamación/metabolismo , Interleucina-1beta/genética , Quinasas Janus/metabolismo , Yeyuno/metabolismo , Lipopolisacáridos/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/genética , Masculino , Ratones , Mucositis/tratamiento farmacológico , Fosforilación , Probióticos/administración & dosificación , Receptor Toll-Like 2/genética , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , Receptores Toll-Like/genética , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
12.
J Am Coll Nutr ; 39(1): 72-81, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31082302

RESUMEN

Objective:Boza is a fermented cereal beverage which is produced by co-culture fermentation of lactic acid bacteria and yeasts. In addition to the nutritional properties of cereals used in the production, it is also suitable to be gaining functional properties by fermenting with probiotic microorganisms.Methods: In this study, protein content of probiotic boza was increased by the addition of gluten, zein and chickpea flour and the volatile compounds formed during co-culture fermentation of the cereal medium with Lactobacillus acidophilus, Bifidobacterium bifidum and Saccharomyces boulardii were determined.Results: It was determined that chickpea added boza provided the highest cell counts of Lactobacillus acidophilus (7.92 logs CFU/g), Bifidobacterium bifidum (7.32 log CFU/g) and Saccharomyces boulardii (3.26 log CFU/g) during storage. With the addition of gluten, the protein content of the sample was enriched four times more when compared with control boza. During fermentation and storage, a total of 36 different compounds were identified with the major compounds as 9,12-octadecadienoic acid, 9-octadecenoic acid, hexadecanoic acid and hexadecanoic acid, 2-hydroxy-1-(hydroxymethyl)ethyl ester. The concentration of volatile compounds generally decreased during storage of samples. According to Principle Cluster Analysis results, enriched protein samples had similar projections due to their fatty acid contents and the main difference was shown in the control sample.Conclusions: The results of this study indicate that chickpea, single or mixture with cereals, can be a good substrate for probiotic microorganism production for acceptance as probiotic foods.


Asunto(s)
Proteínas en la Dieta/administración & dosificación , Grano Comestible/química , Alimentos Fermentados/microbiología , Alimentos Fortificados/análisis , Probióticos/química , Bifidobacterium bifidum/metabolismo , Cicer/metabolismo , Técnicas de Cocultivo , Recuento de Colonia Microbiana , Fermentación , Harina , Microbiología de Alimentos , Glútenes/metabolismo , Humanos , Lactobacillus acidophilus/metabolismo , Saccharomyces boulardii/metabolismo , Zeína/metabolismo
13.
Appl Environ Microbiol ; 84(10)2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29523547

RESUMEN

The probiotic yeast Saccharomyces boulardii has been extensively studied for the prevention and treatment of diarrheal diseases, and it is now commercially available in some countries. S. boulardii displays notable phenotypic characteristics, such as a high optimal growth temperature, high tolerance against acidic conditions, and the inability to form ascospores, which differentiate S. boulardii from Saccharomyces cerevisiae The majority of prior studies stated that S. boulardii exhibits sluggish or halted galactose utilization. Nonetheless, the molecular mechanisms underlying inefficient galactose uptake have yet to be elucidated. When the galactose utilization of a widely used S. boulardii strain, ATCC MYA-796, was examined under various culture conditions, the S. boulardii strain could consume galactose, but at a much lower rate than that of S. cerevisiae While all GAL genes were present in the S. boulardii genome, according to analysis of genomic sequencing data in a previous study, a point mutation (G1278A) in PGM2, which codes for phosphoglucomutase, was identified in the genome of the S. boulardii strain. As the point mutation resulted in the truncation of the Pgm2 protein, which is known to play a pivotal role in galactose utilization, we hypothesized that the truncated Pgm2 might be associated with inefficient galactose metabolism. Indeed, complementation of S. cerevisiaePGM2 in S. boulardii restored galactose utilization. After reverting the point mutation to a full-length PGM2 in S. boulardii by Cas9-based genome editing, the growth rates of wild-type (with a truncated PGM2 gene) and mutant (with a full-length PGM2) strains with glucose or galactose as the carbon source were examined. As expected, the mutant (with a full-length PGM2) was able to ferment galactose faster than the wild-type strain. Interestingly, the mutant showed a lower growth rate than that of the wild-type strain on glucose at 37°C. Also, the wild-type strain was enriched in the mixed culture of wild-type and mutant strains on glucose at 37°C, suggesting that the truncated PGM2 might offer better growth on glucose at a higher temperature in return for inefficient galactose utilization. Our results suggest that the point mutation in PGM2 might be involved in multiple phenotypes with different effects.IMPORTANCESaccharomyces boulardii is a probiotic yeast strain capable of preventing and treating diarrheal diseases. However, the genetics and metabolism of this yeast are largely unexplored. In particular, molecular mechanisms underlying the inefficient galactose metabolism of S. boulardii remain unknown. Our study reports that a point mutation in PGM2, which codes for phosphoglucomutase, is responsible for inferior galactose utilization by S. boulardii After correction of the mutated PGM2 via genome editing, the resulting strain was able to use galactose faster than a parental strain. While the PGM2 mutation made the yeast use galactose slowly, investigation of the genomic sequencing data of other S. boulardii strains revealed that the PGM2 mutation is evolutionarily conserved. Interestingly, the PGM2 mutation was beneficial for growth at a higher temperature on glucose. We speculate that the PGM2 mutation was enriched due to selection of S. boulardii in the natural habitat (sugar-rich fruits in tropical areas).


Asunto(s)
Proteínas Fúngicas/genética , Galactosa/metabolismo , Fosfoglucomutasa/genética , Probióticos/metabolismo , Saccharomyces boulardii/metabolismo , Proteínas Fúngicas/metabolismo , Mutación , Fosfoglucomutasa/metabolismo , Saccharomyces boulardii/enzimología , Saccharomyces boulardii/genética , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Esporas Fúngicas
14.
J Appl Microbiol ; 122(6): 1663-1671, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28339160

RESUMEN

AIMS: Lactococcus lactis N8 and Saccharomyces boulardii SAA655 were investigated for their ability to synthesize B-vitamins (riboflavin and folate) and their functional role as microbial starters in idli fermentation. METHODS AND RESULTS: In this study, ultra-high performance liquid chromatography and microbiological assay were used to determine the total riboflavin and folate content respectively. Increased levels of folate were evident in both L. lactis N8 and S. boulardii SAA655 cultivated medium. Enhanced riboflavin levels were found only in S. boulardii SAA655 grown medium, whereas decreased riboflavin level was found in L. lactis N8 cultivated medium. To evaluate the functional role of microbial starter strains, L. lactis N8 and S. boulardii SAA655 were incorporated individually and in combination into idli batter, composed of wet grounded rice and black gram. For the experiments, naturally fermented idli batter was considered as control. The results indicated that natural idli fermentation did not enhance the riboflavin level and depleted folate levels by half. In comparison with control, L. lactis N8 and S. boulardii SAA655 incorporated idli batter (individually and in combination) increased riboflavin and folate levels by 40-90%. Apart from compensating the folate loss caused by natural fermentation, S. boulardii SAA655 fermented idli batter individually and in combination with L. lactis N8 also showed the highest leavening character. Moreover, the microbial starter incorporation did not significantly influence the pH of idli batter. CONCLUSION: Incorporation of L. lactis N8 and S. boulardii SAA655 can evidently enhance the functional and technological characteristics of idli batter. SIGNIFICANCE AND IMPACT OF THE STUDY: UN General Assembly declared 2016 the International Year of pulses emphasizing the importance of legumes as staple food. Furthermore, this is the first experimental report of in situ biofortifcation of riboflavin and folate using microbes in pulse based fermented staple food. The current study suggests possible avenues for research towards an economical strategy to reduce B-vitamin deficiency among the consuming population.


Asunto(s)
Biofortificación/métodos , Ácido Fólico/metabolismo , Lactococcus lactis/metabolismo , Riboflavina/metabolismo , Saccharomyces boulardii/metabolismo , Grano Comestible/metabolismo , Fermentación , Oryza , Vitaminas/metabolismo
15.
J Agric Food Chem ; 72(8): 4257-4266, 2024 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-38354318

RESUMEN

Selenium nanoparticles (SeNPs) are important and safe food and feed additives that can be used for dietary supplementation. In this study, a mutagenic strain of Saccharomyces boulardii was employed to obtain biologically synthesized SeNPs (BioSeNPs) with the desired particle size by controlling the dosage and duration of sodium selenite addition, and the average particle size achieved was 55.8 nm with protease A encapsulation. Transcriptomic analysis revealed that increased expression of superoxide dismutase 1 (SOD1) in the mutant strain effectively promoted the synthesis of BioSeNPs and the formation of smaller nanoparticles. Under sodium selenite stress, the mutant strain exhibited significantly increased expression of glutathione peroxidase 2 (GPx2), which was significantly greater in the mutant strain than in the wild type, facilitating the synthesis of glutathione selenol and providing abundant substrates for the production of BioSeNPs. Furthermore, based on the experimental results and transcriptomic analysis of relevant genes such as sod1, gpx2, the thioredoxin reductase 1 gene (trr1) and the thioredoxin reductase 2 gene (trr2), a yeast model for the size-controlled synthesis of BioSeNPs was constructed. This study provides an important theoretical and practical foundation for the green synthesis of controllable-sized BioSeNPs or other metal nanoparticles with potential applications in the fields of food, feed, and biomedicine.


Asunto(s)
Nanopartículas del Metal , Nanopartículas , Saccharomyces boulardii , Selenio , Catálisis , Saccharomyces boulardii/metabolismo , Selenio/metabolismo , Selenito de Sodio , Superóxido Dismutasa/genética , Superóxido Dismutasa-1
16.
J Trace Elem Med Biol ; 83: 127402, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38310829

RESUMEN

BACKGROUND AND OBJECTIVE: Yeasts have the remarkable capability to transform and integrate inorganic selenium into their cellular structures, thereby enhancing its bioavailability and reducing its toxicity. In recent years, yeasts have attracted attention as potential alternative sources of protein. METHODS: This study explores the selenium accumulation potential of two less explored yeast strains, namely the probiotic Saccharomyces boulardii CCDM 2020 and Pichia fermentas CCDM 2012, in comparison to the extensively studied Saccharomyces cerevisiae CCDM 272. Our investigation encompassed diverse stress conditions. Subsequently, the selenized yeasts were subjected to an INFOGEST gastrointestinal model. The adherence and hydrophobicity were determined with undigested cells RESULTS: Stress conditions had an important role in influencing the quantity and size of selenium nanoparticles (SeNPs) generated by the tested yeasts. Remarkably, SeMet synthesis was limited to Pichia fermentas CCDM 2012 and S. boulardii CCDM 2020, with S. cerevisiae CCDM 272 not displaying SeMet production at all. Throughout the simulated gastrointestinal digestion, the most substantial release of SeCys2, SeMet, and SeNPs from the selenized yeasts occurred during the intestinal phase. Notably, exception was found in strain CCDM 272, where the majority of particles were released during the oral phase. CONCLUSION: The utilization of both traditional and non-traditional selenized yeast types, harnessed for their noted functional attributes, holds potential for expanding the range of products available while enhancing their nutritional value and health benefits.


Asunto(s)
Probióticos , Saccharomyces boulardii , Selenio , Saccharomyces cerevisiae/química , Saccharomyces boulardii/metabolismo , Pichia , Selenio/metabolismo , Probióticos/metabolismo , Digestión
17.
ACS Synth Biol ; 13(6): 1851-1865, 2024 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-38787439

RESUMEN

Saccharomyces boulardii (Sb) is an emerging probiotic chassis for delivering biomolecules to the mammalian gut, offering unique advantages as the only eukaryotic probiotic. However, precise control over gene expression and gut residence time in Sb have remained challenging. To address this, we developed five ligand-responsive gene expression systems and repaired galactose metabolism in Sb, enabling inducible gene expression in this strain. Engineering these systems allowed us to construct AND logic gates, control the surface display of proteins, and turn on protein production in the mouse gut in response to dietary sugar. Additionally, repairing galactose metabolism expanded Sb's habitat within the intestines and resulted in galactose-responsive control over gut residence time. This work opens new avenues for precise dosing of therapeutics by Sb via control over its in vivo gene expression levels and localization within the gastrointestinal tract.


Asunto(s)
Galactosa , Probióticos , Saccharomyces boulardii , Animales , Ratones , Galactosa/metabolismo , Saccharomyces boulardii/genética , Saccharomyces boulardii/metabolismo , Tracto Gastrointestinal/microbiología , Tracto Gastrointestinal/metabolismo , Dieta
18.
Food Chem ; 454: 139839, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-38810444

RESUMEN

Probiotic lactic acid bacteria have been widely studied, but much less was focused on probiotic yeasts in food systems. In this study, probiotic Saccharomyces cerevisiae var. boulardii CNCM I-745 was employed to prepare ice cream added with and without inulin (1%, w/v). Metabolomics analysis on the effect of inulin showed 84 and 147 differentially expressed metabolites identified in the ice cream samples from day 1 and day 30 of storage (-18 °C), respectively. Various potential functional metabolites were found, including citric acid, ornithine, D-glucuronic acid, sennoside A, stachyose, maltotetraose, maltopentaose, maltohexaose, maltoheptaose, cis-aconitic acid, gamma-aminobutyric acid, L-threonine, L-glutamic acid, tryptophan, benzoic acid, and trehalose. Higher expression of these metabolites suggested their possible roles through relevant metabolic pathways in improving survivability of the probiotic yeast and functionality of ice cream. This study provides further understanding on the metabolic characteristics of probiotic yeast that potentially affect the functionality of ice cream.


Asunto(s)
Helados , Inulina , Metabolómica , Prebióticos , Probióticos , Saccharomyces cerevisiae , Simbióticos , Inulina/metabolismo , Probióticos/metabolismo , Simbióticos/análisis , Prebióticos/análisis , Saccharomyces cerevisiae/metabolismo , Helados/análisis , Helados/microbiología , Saccharomyces boulardii/metabolismo , Saccharomyces boulardii/química
19.
Microbiol Res ; 285: 127741, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38761487

RESUMEN

Fructooligosaccharides (FOS) are a common prebiotic widely used in functional foods. Meanwhile, Saccharomyces boulardii is a fungal probiotic frequenly used in the clinical treatment of diarrhea. Compared with single use, the combination of prebiotics and probiotics as symbiotics may be more effective in regulating gut microbiota as recently reported in the literature. The present study aimed to investigate the effects of FOS, S. boulardii and their combination on the structure and metabolism of the gut microbiota in healthy primary and secondary school students using an in vitro fermentation model. The results indicated that S. boulardii alone could not effectively regulate the community structure and metabolism of the microbiota. However, both FOS and the combination of FOS and S. boulardii could effectively regulate the microbiota, significantly inhibiting the growth of Escherichia-Shigella and Bacteroides, and controlling the production of the gases including H2S and NH3. In addition, both FOS and the combination could significantly promote the growth of Bifidobacteria and Lactobacillus, lower environmental pH, and enhance several physiological functions related to synthesis and metabolism. Nevertheless, the combination had more unique benefits as it promoted the growth of Lactobacillus, significantly increased CO2 production and enhanced the functional pathways of carbon metabolism and pyruvic acid metabolism. These findings provide guidance for clinical application and a theoretical basis for the development of synbiotic preparations.


Asunto(s)
Fermentación , Microbioma Gastrointestinal , Oligosacáridos , Prebióticos , Probióticos , Saccharomyces boulardii , Estudiantes , Oligosacáridos/metabolismo , Oligosacáridos/farmacología , Microbioma Gastrointestinal/efectos de los fármacos , Saccharomyces boulardii/metabolismo , Humanos , Probióticos/metabolismo , Niño , Masculino , Adolescente , Femenino , Lactobacillus/metabolismo , Lactobacillus/crecimiento & desarrollo , Bacterias/metabolismo , Bacterias/clasificación , Heces/microbiología , Bifidobacterium/metabolismo , Bifidobacterium/crecimiento & desarrollo
20.
Probiotics Antimicrob Proteins ; 15(4): 967-982, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-35608794

RESUMEN

Saccharomyces boulardii (S. boulardii) is a probiotic and is widely used to improve the nutritional and functional value of food. This study aimed to compare the probiotic properties of S. boulardii and Saccharomyces cerevisiae. A series of in vitro probiotic experiments was performed, including simulated gastrointestinal digestion, bile salt tolerance, hydrophobicity, self-aggregation, and antioxidant and antibacterial properties. Self-aggregation and hydrophobic properties of S. boulardii were relatively poor, but they showed high tolerance, antioxidant properties, and broad antibacterial properties. In addition, non-targeted metabolomics was used to comprehensively analyze the active metabolites of S. boulardii and the metabolic differences between S. boulardii and S. cerevisiae were compared. Saccharomyces boulardii produced many bioactive metabolites, which generally showed antioxidant, antibacterial, antitumor, anti-inflammatory, and other properties. In contrast to S. cerevisiae, S. boulardii produced phenyllactic acid and 2-hydroxyisocaproic acid. There were also significant differences in their metabolic pathways. These results may be of great significance in the medical and food industries and provide a basis for understanding the metabolism of S. boulardii. It also shows that metabolomics is an effective and novel method for screening microbial functional metabolites and identifying functional differences between similar microorganisms.


Asunto(s)
Probióticos , Saccharomyces boulardii , Saccharomyces cerevisiae/metabolismo , Saccharomyces boulardii/metabolismo , Antioxidantes/metabolismo , Antibacterianos/metabolismo , Probióticos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA