Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros

Base de datos
Tipo del documento
Intervalo de año de publicación
1.
J Sci Food Agric ; 104(7): 4165-4175, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38299445

RESUMEN

BACKGROUND: Neonatal feces are one of the most important sources for probiotic isolation. The purpose of this study was the isolation and identification of Bifidobacterium spp. from neonatal feces and the evaluation of in vitro probiotic properties of strains including safety tests. RESULTS: A total of 40 isolates were obtained from 14 healthy newborns' feces in Erzurum province, Türkiye. By their rep-PCR patterns and 16S rRNA gene sequences, isolates were identified as 26 Bifidobacterium breve and 14 Bifidobacterium longum. Fifteen of the isolates tolerated bile salts and showed high resistance to simulated gastric juice. Isolates exhibited varying rates of auto-aggregation and hydrophobicity. In addition, most of the isolates displayed antibacterial activity against Escherichia coli O157:H7, Staphylococcus aureus ATCC 29213, Salmonella Typhimurium RSHMB 95091, and Pseudomonas aeruginosa ATCC 9027. However, only one strain showed bile salt hydrolase activity and two strains showed the ability to produce H2O2. Bifidobacterium strains were generally sensitive to the tested antibiotics and lacked kanamycin, gentamicin, and streptomycin resistance genes, and hemolytic and DNAse activities. On the other hand, it was determined that five strains had various virulence genes including gelE, esp, efaAfs, hyl, and ace. CONCLUSION: Results of the present study suggested that B. longum BH28, B. breve BH4 and B. breve BH5 strains have the potential as probiotic candidates for further studies. © 2024 The Authors. Journal of The Science of Food and Agriculture published by John Wiley & Sons Ltd on behalf of Society of Chemical Industry.


Asunto(s)
Bifidobacterium , Probióticos , Recién Nacido , Humanos , ARN Ribosómico 16S/genética , Peróxido de Hidrógeno , Turquía , Heces/microbiología , Antibacterianos/farmacología
2.
Int J Food Sci Nutr ; 75(1): 45-57, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37845639

RESUMEN

Olive oil, essential ingredient of the Mediterranean diet, is attracting a growing interest due to increasing evidence on its beneficial effects on human health. This study investigated whether extra virgin olive oil (EVOO) possess prebiotic properties. Twenty different monovarietal EVOO samples from 5 Marche region cultivars (Italy) were studied. The prebiotic activity of EVOOs was assessed monitoring the selective stimulation of gut bacterial species and the short chain fatty acids (SCFAs) production, using an in vitro fermentation system. All EVOOs selectively stimulated Lactobacillus spp., with a stronger activity than that observed in the inulin fermentation (positive control). Also, the bifidobacteria population increased; this bifidogenic stimulation was of EVOOs from Raggia cultivar. SCFAs appeared significantly higher after 24 h in all EVOO fermentations than in the control. Acetic and propionic acids production was particularly stimulated. Overall, most of the investigated EVOOs had a potential prebiotic activity, similar or stronger than inulin.


Asunto(s)
Antioxidantes , Inulina , Humanos , Aceite de Oliva , Italia , Proyectos de Investigación
3.
Adv Nutr ; 15(1): 100162, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38072119

RESUMEN

Overweight and obesity are associated with increased intestinal permeability, characterized by loss of gut epithelial integrity, resulting in unregulated passage of lipopolysaccharide (LPS) and other inflammatory triggers into circulation, i.e., metabolic endotoxemia. In obesity, shifts in the gut microbiome negatively impact intestinal permeability. Probiotics are an intervention that can target the gut microbiome by introducing beneficial microbial species, potentially restoring gut barrier integrity. Currently, the role of probiotic supplementation in ameliorating obesity- and overweight-associated increases in gut permeability has not been reviewed. This systematic review aimed to summarize findings from both animal and clinical studies that evaluated the effect of probiotic supplementation on obesity-induced impairment in intestinal permeability (International Prospective Register of Systematic Reviews, CRD42022363538). A literature search was conducted using PubMed (Medline), Web of Science, and CAB Direct from origin until August 2023 using keywords of intestinal permeability, overweight or obesity, and probiotic supplementation. Of 920 records, 26 eligible records were included, comprising 12 animal and 14 clinical studies. Clinical trials ranged from 3 to 26 wk and were mostly parallel-arm (n = 13) or crossover (n = 1) design. In both animal and clinical studies, plasma/serum LPS was the most common measure of intestinal permeability. Eleven of 12 animal studies reported a positive effect of probiotic supplementation in reducing intestinal permeability. However, results from clinical trials were inconsistent, with half reporting reductions in serum LPS and half reporting no differences after probiotic supplementation. Bifidobacterium, Lactobacillus, and Akkermansia emerged as the most common genera in probiotic formulations among the animal and clinical studies that yielded positive results, suggesting that specific bacteria may be more effective at reducing intestinal permeability and improving gut barrier function. However, better standardization of strain use, dosage, duration, and the delivery matrix is needed to fully understand the probiotic impact on intestinal permeability in individuals with overweight and obesity.


Asunto(s)
Sobrepeso , Probióticos , Animales , Humanos , Sobrepeso/terapia , Lipopolisacáridos , Funcion de la Barrera Intestinal , Ensayos Clínicos Controlados Aleatorios como Asunto , Revisiones Sistemáticas como Asunto , Probióticos/farmacología , Probióticos/uso terapéutico , Obesidad/terapia
4.
Lett Appl Microbiol ; 76(12)2023 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-38081214

RESUMEN

Several studies have shown that probiotics can prevent and reduce inflammation in inflammation-related diseases. However, few studies have focused on the interaction between host and probiotics in modulating the immune system through autophagy. Therefore, we aimed to investigate the preventive and/or therapeutic effects of native potential probiotic breast milk-isolated Bifidobacterium spp. (i.e. B. bifidum, B. longum, and B. infantis) on the inflammatory cascade by affecting autophagy gene expression 24 and 48 h after treatment. Autophagy genes involved in different stages of the autophagy process were selected by quantitative polymerase chain reaction (qPCR). Gene expression investigation was accomplished by exposing the human colorectal adenocarcinoma cell line (HT-29) to sonicated pathogens (1.5 × 108 bacterial CFU ml-1) and adding Bifidobacterium spp. (MOI10) before, after, and simultaneously with induction of inflammation. An equal volume of RPMI medium was used as a control. Generally, our native potential probiotic Bifidobacterium spp. can increase the autophagy gene expression in comparison with pathogen. Moreover, an increase in gene expression was observed with our probiotic strains' consumption in all stages of autophagy. Totally, our selected Bifidobacterium spp. can increase autophagy gene expression before, simultaneously, and after the inflammation induction, so they can prevent and reduce inflammation in an in vitro model of inflammation.


Asunto(s)
Bifidobacterium , Probióticos , Femenino , Humanos , Bifidobacterium/metabolismo , Leche Humana , Inflamación/prevención & control
5.
Nutrients ; 15(24)2023 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-38140334

RESUMEN

Scientific evidence has increasingly supported the beneficial effects of probiotic-based food supplements on human intestinal health. This ex vivo study investigated the effects on the composition and metabolic activity of the intestinal microbiota of three probiotic-based food supplements, containing, respectively, (1) Bifidobacterium longum ES1, (2) Lactobacillus acidophilus NCFM®, and (3) a combination of L. acidophilus NCFM®, Lactobacillus paracasei Lpc-37™, Bifidobacterium lactis Bi-07™, and Bifidobacterium lactis Bl-04™. This study employed fecal samples from six healthy donors, inoculated in a Colon-on-a-plate® system. After 48 h of exposure or non-exposure to the food supplements, the effects were measured on the overall microbial fermentation (pH), changes in microbial metabolic activity through the production of short-chain and branched-chain fatty acids (SCFAs and BCFAs), ammonium, lactate, and microbial composition. The strongest effect on the fermentation process was observed for the combined formulation probiotics, characterized by the significant stimulation of butyrate production, a significant reduction in BCFAs and ammonium in all donors, and a significant stimulatory effect on bifidobacteria and lactobacilli growth. Our findings suggest that the combined formulation probiotics significantly impact the intestinal microbiome of the healthy individuals, showing changes in metabolic activity and microbial abundance as the health benefit endpoint.


Asunto(s)
Compuestos de Amonio , Microbioma Gastrointestinal , Probióticos , Humanos , Probióticos/farmacología , Suplementos Dietéticos , Lactobacillus acidophilus/fisiología , Ácidos Grasos Volátiles
6.
Crit Rev Microbiol ; : 1-17, 2023 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-37551693

RESUMEN

Recently, more consumers are interested in purchasing probiotic food and beverage products that may improve their immune health. The market for functional foods and beverages that include Bifidobacterium is expanding because of their potential uses in both food and therapeutic applications. However, maintaining Bifidobacterium's viability during food processing and storage remains a challenge. Microencapsulation technique has been explored to improve the viability of Bifidobacterium. Despite the technical, microbiological, and economic challenges, the market potential for immune-supporting functional foods and beverages is significant. Additionally, there is a shift toward postbiotics as a solution for product innovation, a promising postbiotic product that can be incorporated into various food and beverage formats is also introduced in this review. As consumers become more health-conscious, future developments in the functional food and beverage market discussed in this review could serve as a reference for researchers and industrialist.

8.
Biomedicines ; 11(2)2023 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-36831029

RESUMEN

Crohn's disease (CD) and ulcerative colitis (UC) are chronic inflammatory diseases of the gastrointestinal tract affecting millions of patients worldwide. The gut microbiome partly determines the pathogenesis of both diseases. Even though probiotics have been widely used as a potential treatment, their efficacy in inducing and maintaining remission is still controversial. Our study aims to review the present-day literature about the possible role of probiotics in treating inflammatory bowel diseases in adults. This research was performed according to the Preferred Reporting Items for Systematic reviews and Meta-Analyses (PRISMA) guidelines. We included studies concerning adult patients who compared probiotics with placebo or non-probiotic intervention. We identified thirty-three studies, including 2713 patients from fourteen countries. The role of probiotics in Crohn's disease was examined in eleven studies. Only four studies presented statistically significant results in the remission of disease, primarily when used for three to six months. On the other hand, in twenty-one out of twenty-five studies, probiotics proved effective in achieving or maintaining remission in ulcerative colitis. Supplementation with Bifidobacterium sp. or a combination of probiotics is the most effective intervention, especially when compared with a placebo. There is strong evidence supporting the usage of probiotic supplementation in patients with ulcerative colitis, yet more research is needed to justify their efficacy in Crohn's disease.

9.
Adv Nutr ; 14(2): 238-255, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36775788

RESUMEN

Carotenoids have been related to a number of health benefits. Their dietary intake and circulating levels have been associated with a reduced incidence of obesity, diabetes, certain types of cancer, and even lower total mortality. Their potential interaction with the gut microbiota (GM) has been generally overlooked but may be of relevance, as carotenoids largely bypass absorption in the small intestine and are passed on to the colon, where they appear to be in part degraded into unknown metabolites. These may include apo-carotenoids that may have biological effects because of higher aqueous solubility and higher electrophilicity that could better target transcription factors, i.e., NF-κB, PPARγ, and RAR/RXRs. If absorbed in the colon, they could have both local and systemic effects. Certain microbes that may be supplemented were also reported to produce carotenoids in the colon. Although some bactericidal aspects of carotenoids have been shown in vitro, a few studies have also demonstrated a prebiotic-like effect, resulting in bacterial shifts with health-associated properties. Also, stimulation of IgA could play a role in this respect. Carotenoids may further contribute to mucosal and gut barrier health, such as stabilizing tight junctions. This review highlights potential gut-related health-beneficial effects of carotenoids and emphasizes the current research gaps regarding carotenoid-GM interactions.


Asunto(s)
Carotenoides , Microbioma Gastrointestinal , Humanos , Carotenoides/farmacología , Carotenoides/metabolismo , Colon/metabolismo , Prebióticos , Suplementos Dietéticos
10.
J Complement Integr Med ; 20(1): 223-232, 2023 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-34758244

RESUMEN

OBJECTIVES: Evidence for the contribution of the brain-gut-microbiota axis to the depression pathophysiology is increasing nowadays. Disturbed gut microbiota equilibrium along with bad dietary habits both lead to kynurenine pathway abnormalities contributing to the depression pathophysiology. In this respect, many studies are found but the interventional clinical trials are limited. The present interventional study aims to evaluate the impact of Bifidobacterium spp. supplementation together with improving dietary intake on depressive mood and well-being and their correlation with kynurenine blood level in adult Egyptian healthy volunteers. METHODS: A number of 98 healthy female volunteers with a mean age of 46.96 ± 1.82 years were selected and enrolled in this study. They were given yogurt enriched with Bifidobacterium spp. daily for eight weeks. Clinical examination as well as questionnaires for the evaluation of psychological well-being and depression were done at base line and after eight weeks of intervention. Fasting blood samples and stool samples were collected from all subjects at baseline and eight weeks after the intervention for the investigation of serum kynurenine concentration, blood hemoglobin, serum transaminases (ALT & AST) serum urea and creatinine as well as fecal Bifidobacterium count. RESULTS: Data revealed that both depression and well-being showed highly significant improvement combined with significant drop in kynurenine blood level after intervention. Also, a significant rise in fecal Bifidobacterium count and a significant improvement in hemoglobin level and activity of liver enzymes were recorded. After intervention, a significant negative correlation was recorded between depression and fecal Bifidobacterium count as well as between serum kynurenine level, and well-being. CONCLUSION: Bifidobacterium spp. supplementation combined with improvement in dietary intake resulted in improvement of depressive mood and well-being and reduced kynurenine blood level.


Asunto(s)
Bifidobacterium , Probióticos , Adulto , Humanos , Femenino , Persona de Mediana Edad , Probióticos/uso terapéutico , Depresión/terapia , Quinurenina , Afecto
11.
Crit Rev Food Sci Nutr ; 63(26): 8048-8065, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-35319324

RESUMEN

Members of Bifidobacterium are among the first microbes to colonize the human intestine naturally, their abundance and diversity in the colon are closely related to host health. Recently, the gut microbiota has been gradually proven to be crucial mediators of various metabolic processes between the external environment and the host. Therefore, the health-promoting benefits of Bifidobacterium spp. and their applications in food have gradually been widely concerned. The main purpose of this review is to comprehensively introduce general features, colonization methods, and safety of Bifidobacterium spp. in the human gut, highlighting its health benefits and industrial applications. On this basis, the existing limitations and scope for future research are also discussed. Bifidobacteria have beneficial effects on the host's digestive system, immune system, and nervous system. However, the first prerequisite for functioning is to have enough live bacteria before consumption and successfully colonize the colon after ingestion. At present, strain breeding, optimization (e.g., selecting acid and bile resistant strains, adaptive evolution, high cell density culture), and external protection technology (e.g., microencapsulation and protectants) are the main strategies to address these challenges in food application.


Asunto(s)
Microbioma Gastrointestinal , Probióticos , Humanos , Bifidobacterium/metabolismo , Alimentos Funcionales
12.
Int J Mol Sci ; 23(24)2022 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-36555802

RESUMEN

The human oral pathobionts Aggregatibacter actinomycetemcomitans, Streptococcus mitis and Streptococcus mutans, in dysbiosis-promoting conditions, lead to oral infections, which also represent a threat to human systemic health. This scenario may be worsened by antibiotic misuse, which favours multi-drug resistance, making the research on pathogen containment strategies more than crucial. Therefore, we aimed to in vitro select the most promising probiotic strains against oral pathogen growth, viability, biofilm formation, and co-aggregation capacity, employing both the viable probiotics and their cell-free supernatants (CFSs). Interestingly, we also assessed probiotic efficacy against the three-pathogen co-culture, mimicking an environment similar to that in vivo. Overall, the results showed that Lactobacillus CFSs performed better than the Bifidobacterium, highlighting Limosilactobacillus reuteri LRE11, Lacticaseibacillus rhamnosus LR04, Lacticaseibacillus casei LC04, and Limosilactobacillus fermentum LF26 as the most effective strains, opening the chance to deeper investigation of their action and CFS composition. Altogether, the methodologies presented in this study can be used for probiotic efficacy screenings, in order to better focus the research on a viable probiotic, or on its postbiotics, suitable in case of infections.


Asunto(s)
Lacticaseibacillus casei , Probióticos , Humanos , Lactobacillus , Bifidobacterium , Streptococcus mutans , Probióticos/uso terapéutico
13.
Int J Mol Sci ; 23(20)2022 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-36293436

RESUMEN

The accurate identification of microorganisms belonging to vaginal microflora is crucial for establishing which microorganisms are responsible for microbial shifting from beneficial symbiotic to pathogenic bacteria and understanding pathogenesis leading to vaginosis and vaginal infections. In this study, we involved the surface-enhanced Raman spectroscopy (SERS) technique to compile the spectral signatures of the most significant microorganisms being part of the natural vaginal microbiota and some vaginal pathogens. Obtained data will supply our still developing spectral SERS database of microorganisms. The SERS results were assisted by Partial Least Squares Regression (PLSR), which visually discloses some dependencies between spectral images and hence their biochemical compositions of the outer structure. In our work, we focused on the most common and typical of the reproductive system microorganisms (Lactobacillus spp. and Bifidobacterium spp.) and vaginal pathogens: bacteria (e.g., Gardnerella vaginalis, Prevotella bivia, Atopobium vaginae), fungi (e.g., Candida albicans, Candida glabrata), and protozoa (Trichomonas vaginalis). The obtained results proved that each microorganism has its unique spectral fingerprint that differentiates it from the rest. Moreover, the discrimination was obtained at a high level of explained information by subsequent factors, e.g., in the inter-species distinction of Candida spp. the first three factors explain 98% of the variance in block Y with 95% of data within the X matrix, while in differentiation between Lactobacillus spp. and Bifidobacterium spp. (natural flora) and pathogen (e.g., Candida glabrata) the information is explained at the level of 45% of the Y matrix with 94% of original data. PLSR gave us insight into discriminating variables based on which the marker bands representing specific compounds in the outer structure of microorganisms were found: for Lactobacillus spp. 1400 cm-1, for fungi 905 and 1209 cm-1, and for protozoa 805, 890, 1062, 1185, 1300, 1555, and 1610 cm-1. Then, they can be used as significant marker bands in the analysis of clinical subjects, e.g., vaginal swabs.


Asunto(s)
Microbiota , Vaginosis Bacteriana , Femenino , Humanos , Análisis de los Mínimos Cuadrados , Gardnerella vaginalis , Vagina/microbiología , Lactobacillus , Bacterias , Bifidobacterium
14.
Mol Neurobiol ; 59(11): 6896-6902, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36050597

RESUMEN

Children with autism spectrum disorder (ASD) are usually unable to express abdominal discomfort properly, and thus gastrointestinal symptoms (GIS) are sometimes shadowed by aggression, which is sometimes misunderstood as a behavioral characteristic of ASD. Several studies have reported interesting correlations between the severity of behavioral and gastrointestinal symptoms in ASD children. The present study aimed to investigate the potential effects of probiotics as an adjuvant therapy to modulate the clinical status of ASD children. This study included 40 children with ASD aged 2-5 years. The feeding product was prepared from whey powder (without casein) and some minced cooked yellow vegetables in adequate ratios fortified with the studied probiotic strains (Bifidobacterium spp. and Lactobacillus spp.). Bifidobacterium strains were assessed from stool samples of children with ASD. Bifidobacterium strains were analyzed in the stools of ASD children. Recruited ASD patients received 10 g of the nutritional supplement once a day for 3 months. Childhood Autism Rating Scale (CARS) and Autism Diagnostic Interview-Revised (ADIR) were reevaluated clinically. Questionnaire on Pediatric Gastrointestinal Symptoms-Rome III Version was used for all children with ASD before and after. There is a significant increase in the colony counts of both Bifidobacterium spp. and Lactobacillus spp., which present in the stool of ASD children after probiotic supplementation for 3 months. It was highly significant in the case of Bifidobacterium spp. (p value 0.000) and a significant increase in Lactobacillus spp. (p value 0.015). The present study showed reduced anxiety and observation of deep sleep for children with ASD (80%) after taking the supplementation. This indicates that probiotics may have a potential effect in reducing symptoms and severity of ASD and in correcting dysbiosis.


Asunto(s)
Trastorno del Espectro Autista , Enfermedades Gastrointestinales , Probióticos , Trastorno del Espectro Autista/tratamiento farmacológico , Bifidobacterium , Caseínas/uso terapéutico , Niño , Humanos , Polvos/uso terapéutico , Probióticos/uso terapéutico
15.
Microorganisms ; 10(8)2022 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-36013992

RESUMEN

The features of gut microbiota in metabolic syndrome (MS) and ways to correct it using autoprobiotics, based on indigenous bacteria obtained from fecal samples of the host, remain unexplored. The aim of the study was to investigate the effectiveness of an indigenous consortium (IC) of fecal bacteria in treatment of patients with MS. The study was carried out on 36 patients with MS, manifested with abdominal obesity, eating disorders, dyslipidemia, and hypertension. The control group was formed by 20 healthy volunteers. Samples of IC and gut microbiota content were examined by qPCR and metagenome (16S rRNA) analysis before and after therapy. The decrease in anthropometric parameters of obesity, liver enzyme level correction, reduction in C reactive protein and triglyceride concentrations were revealed after IC usage. The decrease in genera Bifidobacterium, Enterobacter, Paraprevotella, and Prevotella, as well as an increase in Bacteroides fragilis and Oscillospira spp. populations were shown after consumption of IC. A negative correlation between the quantity of B. fragilis and the anthropometric parameters of obesity (r = -0.48) and C reactive protein level (r = -0.36) in serum was established. Thus, IC can be considered as a potential functional personified product for the therapy of MS.

16.
Nutrients ; 14(16)2022 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-36014803

RESUMEN

Background: Numerous studies have investigated the effects of the supplementation of fructooligosaccharides (FOS) on the number of bacteria in the gut that are good for health, but the results have been inconsistent. Additionally, due to its high fermentability, supplementation of FOS may be associated with adverse gastrointestinal symptoms such as bloating and flatulence. Therefore, we assessed the effects of FOS interventions on the composition of gut microbiota and gastrointestinal symptoms in a systematic review and meta-analysis. Design: All randomized controlled trials published before 10 July 2022 that investigated the effects of FOS supplementation on the human gut microbiota composition and gastrointestinal symptoms and met the selection criteria were included in this study. Using fixed or random-effects models, the means and standard deviations of the differences between the two groups before and after the intervention were combined into weighted mean differences using 95% confidence intervals (CIs). Results: Eight studies containing 213 FOS supplements and 175 controls remained in this meta-analysis. Bifidobacterium spp. counts significantly increased during FOS ingestion (0.579, 95% CI: 0.444−0.714) in comparison with that of the control group. Subgroup analysis showed greater variation in Bifidobacterium spp. in adults (0.861, 95% CI: 0.614−1.108) than in infants (0.458, 95% CI: 0.297−0.619). The increase in Bifidobacterium spp. counts were greater in the group with an intervention duration greater than 4 weeks (0.841, 95% CI: 0.436−1.247) than an intervention time less than or equal to four weeks (0.532, 95% CI: 0.370−0.694), and in the group with intervention doses > 5 g (1.116, 95% CI: 0.685−1.546) the counts were higher than those with doses ≤ 5 g (0.521, 95% CI: 0.379−0.663). No differences in effect were found between FOS intervention and comparators in regard to the abundance of other prespecified bacteria or adverse gastrointestinal symptoms. Conclusions: This is the first meta-analysis to explore the effect of FOS on gut microbiota and to evaluate the adverse effects of FOS intake on the gastrointestinal tract. FOS supplementation could increase the number of colonic Bifidobacterium spp. while higher dose (7.5−15 g/d) and longer duration (>4 weeks) showed more distinct effects and was well tolerated.


Asunto(s)
Microbioma Gastrointestinal , Adulto , Bacterias , Bifidobacterium , Suplementos Dietéticos , Humanos , Lactante , Oligosacáridos/farmacología , Ensayos Clínicos Controlados Aleatorios como Asunto
17.
J Microbiol Biotechnol ; 32(9): 1186-1194, 2022 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-36039384

RESUMEN

The intake of probiotic lactic acid bacteria not only promotes digestion through the microbiome regulated host intestinal metabolism but also improves diseases such as irritable bowel syndrome and inflammatory bowel disease, and suppresses pathogenic harmful bacteria. This investigation aimed to evaluate the immunomodulatory effects in intestinal epithelial cells and to study the clinical efficacy of the selected the Bifidobacterium breve and Bifidobacterium longum groups. The physiological and biochemical properties were characterized, and immunomodulatory activity was measured against pathogenic bacteria. In order to find out the mechanism of inflammatory action of the eight viable and sonicated Bifidobacterium spp., we tried to confirm the changes in the pro-inflammatory cytokines (TNF-α, interleukin (IL)-6, IL-12) and anti-inflammatory cytokine (IL-10), and chemokines, (monocyte chemoattractant protein-1, IL-8) and inflammatory enzymatic mediator (nitric oxide) against Enterococcus faecalis ATCC 29212 infection in Caco-2 cells and RAW 264.7 cells. The clinical efficacy of the selected B. breve and B. longum group was studied as a probiotic adjuvant for acute diarrhea in children by oral administration. The results showed significant immunomodulatory effects on the expression levels of TNF-α, IL-6, IL-12, MCP-1, IL-8 and NO, in sonicated Bifidobacterium extracts and viable bifidobacteria. Moreover, each of the Bifidobacterium strains was found to react more specifically to different cytokines. However, treatment with sonicated Bifidobacterium extracts showed a more significant effect compared to treatment with the viable bacteria. We suggest that probiotics functions should be subdivided according to individual characteristics, and that personalized probiotics should be designed to address individual applications.


Asunto(s)
Bifidobacterium breve , Bifidobacterium longum , Probióticos , Bifidobacterium/metabolismo , Bifidobacterium breve/metabolismo , Bifidobacterium longum/metabolismo , Células CACO-2 , Quimiocina CCL2 , Quimiocinas , Niño , Citocinas/metabolismo , Diarrea/terapia , Humanos , Interleucina-10 , Interleucina-12 , Interleucina-6 , Interleucina-8 , Óxido Nítrico , Factor de Necrosis Tumoral alfa
18.
Lett Appl Microbiol ; 75(5): 1254-1263, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35876252

RESUMEN

Inflammatory bowel disease (IBD) is a chronic inflammatory disease with relapses and periods of remission. Forasmuch as, dysregulation of the immune system is one of the triggers of IBD, taking probiotics as one of the immunomodulators in the gut, could help to control inflammation and IBD via influencing signalling pathways. Here, we aimed to investigate the efficacy of five selected Bifidobacterium strains in modulating JAK/STAT and NF-kB inflammatory signalling pathways via using the in vitro assay. A quantitative real-time polymerase chain reaction assay was used to analyse the expression of JAK/STAT and inflammatory genes followed by potential probiotic treatments before, after and simultaneously with the inflammation induction (sonicated pathogen). The production of IL-6 and IL-1ß after probiotic treatment was evaluated. Probiotic treatment resulted in the downregulation of TIRAP, IRAK4, NEMO and RIP genes in the NF-kB pathway, as well as JAK genes compared to sonicate-treated cells. The expression of STAT genes was different after our selected Bifidobacterium strains treatment. The production of IL-6 and IL-1ß decreased after probiotic treatment. These strains of Bifidobacterium spp. showed anti-inflammatory effects on HT-29 cells via modulation of JAK/STAT and NF-kB signalling pathways. The use of Bifidobacterium spp. could be considered as a suitable preventive and complementary treatment for patients with inflammatory bowel disease.


Asunto(s)
Enfermedades Inflamatorias del Intestino , Probióticos , Humanos , Bifidobacterium , Quinasas Asociadas a Receptores de Interleucina-1 , Interleucina-6/genética , FN-kappa B/genética , Probióticos/farmacología , Inflamación/terapia , Enfermedades Inflamatorias del Intestino/prevención & control , Antiinflamatorios
19.
Food Sci Nutr ; 10(6): 1994-2008, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35702306

RESUMEN

Commercial vegetable and fruit juices with probiotics are new functional type of beverages; however, limitations including persistence and impact of probiotic bacteria on palatability and shelf life may prevent their industrial development. This study evaluated the effect of antioxidant compounds (ascorbic acid, astaxanthin, and ginseng) on viability and persistence of Bifidobacterium spp. in Jerusalem Artichoke (JA) juice; and determine the impact of these antioxidants on the sensory (color, texture, flavor, acidity) properties, free reducing sugar (inulin and fructose), and shelf life in the fortified JA juice. Overall, the JA juice fortified with ascorbic acid showed a significant impact on the rate of persistence of two targeted bifidobacterial strains from 1 to 28 days at 5°C. Both strains produced slight acidity in ascorbic acid fortified JA juice as compared to other tested samples. Similarly, the JA juice fortified with ascorbic acid showed a significantly high increase in the total number of bifidobacterial cells of both species, enhanced palatability, and shelf life as compared to astaxanthin and ginseng extract. The quadratic model indicated a strong association between ascorbic acid, ginseng extract, and astaxanthin with a bifidobacterial cell concentration in the fortified JA juices. The Box-Behnken design was considered a feasible analysis for describing fortified JA juice and the rate of viability and persistence of bifidobacteria during 28 days of storage at 5°C in all trials. In conclusion, JA juice fortified with ascorbic acid showed a significant impact on improving the cell viability and persistence of probiotic bacteria, enhanced palatability, and shelf life as compared to other compounds tested.

20.
Int J Mol Sci ; 23(2)2022 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-35054790

RESUMEN

Intestinal homeostasis is a dynamic balance involving the interaction between the host intestinal mucosa, immune barrier, intestinal microecology, nutrients, and metabolites. Once homeostasis is out of balance, it will increase the risk of intestinal diseases and is also closely associated with some systemic diseases. Probiotics (Escherichia coli Nissle 1917, Akkermansia muciniphila, Clostridium butyricum, lactic acid bacteria and Bifidobacterium spp.), maintaining the gut homeostasis through direct interaction with the intestine, can also exist as a specific agent to prevent, alleviate, or cure intestinal-related diseases. With genetic engineering technology advancing, probiotics can also show targeted therapeutic properties. The aims of this review are to summarize the roles of potential native and engineered probiotics in oncology, inflammatory bowel disease, and obesity, discussing the therapeutic applications of these probiotics.


Asunto(s)
Diabetes Mellitus/microbiología , Enfermedades Intestinales/microbiología , Mucosa Intestinal/fisiología , Obesidad/microbiología , Probióticos/uso terapéutico , Akkermansia , Animales , Bifidobacterium , Clostridium butyricum , Diabetes Mellitus/terapia , Escherichia coli , Homeostasis , Humanos , Enfermedades Inflamatorias del Intestino/microbiología , Enfermedades Inflamatorias del Intestino/terapia , Enfermedades Intestinales/terapia , Mucosa Intestinal/microbiología , Lactobacillales , Obesidad/terapia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA