Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 382
Filtrar
1.
Int J Food Microbiol ; 425: 110895, 2024 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-39222566

RESUMEN

Global concerns over folate deficiency, the risks of excessive synthetic folic acid consumption, and food loss implications for environmental sustainability and food security drive needs of innovative approaches that align food by-product valorisation with folate bio-enrichment. This study explored the use of three fruit by-products extracts (grape, passion fruit, and pitaya) and whey to develop a folate bio-enriched fermented whey-based beverage. Three strains (Lacticaseibacillus rhamnosus LGG, Bifidobacterium infantis BB-02, and Streptococcus thermophilus TH-4) were tested for folate production in different fermentation conditions in modified MRS medium and in a whey-based matrix prepared with water extracts of these fruit by-products. B. infantis BB-02 and S. thermophilus TH-4, alone and in co-culture, were the best folate producers. The selection of cultivation conditions, including the presence of different substrates and pH, with grape by-product water extract demonstrating the most substantial effect on folate production among the tested extracts, was crucial for successfully producing a biofortified fermented whey-based beverage (FWBB). The resulting FWBB provided 40.7 µg of folate per 100 mL after 24 h of fermentation at 37 °C, effectively leveraging food by-products. Moreover, the beverage showed no cytotoxicity in mouse fibroblast cells tests. This study highlights the potential for valorising fruit by-products and whey for the design of novel bioenriched foods, promoting health benefits and contributing to reduced environmental impact from improper disposal.


Asunto(s)
Fermentación , Ácido Fólico , Frutas , Suero Lácteo , Animales , Frutas/química , Ratones , Humanos , Suero Lácteo/química , Bebidas/microbiología , Streptococcus thermophilus/metabolismo , Streptococcus thermophilus/crecimiento & desarrollo , Lacticaseibacillus rhamnosus/metabolismo , Lacticaseibacillus rhamnosus/crecimiento & desarrollo , Bifidobacterium/metabolismo , Bifidobacterium/crecimiento & desarrollo , Vitis/química
2.
Food Res Int ; 195: 114976, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39277213

RESUMEN

Changes in dietary patterns and living habits have led to an increasing number of individuals with elevated cholesterol levels. Excessive consumption of high-cholesterol foods can disrupt the body's lipid metabolism. Numerous studies have firmly established the cholesterol-lowering effects of probiotics and prebiotics, with evidence showing that the synergistic use of synbiotics is functionally more potent than using probiotics or prebiotics alone. Currently, the screening strategy involves screening prebiotics for synbiotic development with probiotics as the core. However, in comparison to probiotics, there are fewer types of prebiotics available, leading to limited resources. Consequently, the combinations of synbiotics obtained are restricted, and probiotics and prebiotics are only relatively suitable. Therefore, in this study, a novel synbiotic screening strategy with prebiotics as the core was developed. The synbiotic combination of Lactobacillus rhamnosus S_82 and xylo-oligosaccharides was screened from the intestinal tract of young people through five generations of xylo-oligosaccharides. Subsequently, the cholesterol-lowering ability of the medium was simulated, and the two carbon sources of glucose and xylo-oligosaccharides were screened out. The results showed that synbiotics may participate in cholesterol-lowering regulation by down-regulating the expression of NPC1L1 gene, down-regulating ACAT2 and increasing the expression of ABCG8 gene in vitro through cell adsorption and cell absorption in vitro, and regulating the intestinal microbiota. Synbiotics hold promise as potential candidates for the prevention of hypercholesterolemia in humans and animals, and this study providing a theoretical foundation for the development of new synbiotic products.


Asunto(s)
Lacticaseibacillus rhamnosus , Oligosacáridos , Prebióticos , Simbióticos , Lacticaseibacillus rhamnosus/metabolismo , Oligosacáridos/farmacología , Humanos , Hipolipemiantes/farmacología , Colesterol/metabolismo , Colesterol/sangre , Microbioma Gastrointestinal/efectos de los fármacos , Probióticos , Glucuronatos
3.
Artículo en Inglés | MEDLINE | ID: mdl-39227166

RESUMEN

Spent coffee grounds (SCG) are commercial waste that are still rich in numerous valuable ingredients and can be further processed into useful products such as coffee oil, antioxidant extract, lactic acid, and lignin. The challenge and innovation is to develop the SCG processing technology, maximizing the use of raw material and minimizing the use of other resources within the sequential process. The presented research is focused on the aspect of biotechnological production of lactic acid from SCG by using the Lacticaseibacillus rhamnosus strain isolated from the environment. Thanks to the optimization of the processes of acid hydrolysis, neutralization, enzymatic hydrolysis of SCG, and fermentation, the obtained concentration of lactic acid was increased after 72 hr of culture from the initial 4.60 g/l to 48.6 g/l. In addition, the whole process has been improved, taking into account the dependence on other processes within the complete SCG biorefinery, economy, energy, and waste aspects. Costly enzymatic hydrolysis was completely eliminated, and it was proven that supplementation of SCG hydrolysate with expensive yeast extract can be replaced by cheap waste from the agri-food industry. ONE-SENTENCE SUMMARY: A process for efficient lactic acid production from spent coffee grounds using the Lacticaseibacillus rhamnosus strain was developed and optimized, including nutrient solution preparation, supplementation and fermentation.


Asunto(s)
Café , Fermentación , Ácido Láctico , Lacticaseibacillus rhamnosus , Lacticaseibacillus rhamnosus/metabolismo , Lacticaseibacillus rhamnosus/crecimiento & desarrollo , Ácido Láctico/metabolismo , Ácido Láctico/biosíntesis , Café/química , Hidrólisis , Residuos
4.
J Appl Microbiol ; 135(8)2024 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-39152088

RESUMEN

AIMS: Carbon source is a necessary nutrient for bacterial strain growth. In industrial production, the cost of using different carbon sources varies greatly. Moreover, the complex environment in space may cause metabolic a series of changes in the strain, and this method has been successfully applied in some basic research. To date, space mutagenesis is still limited number of studies, particularly in carbon metabolism of probiotics. METHODS AND RESULTS: HG-R7970-41 was isolated from bacterium suspension (Probio-M9) after space flight, which can produce capsular polysaccharide after space mutagenesis. Phenotype Microarray (PM) was used to evaluated the metabolism of HG-R7970-41 in 190 single carbon sources. RNA sequencing and total protein identification of two strains revealed their different carbon metabolism mechanisms. PM results demonstrated the metabolism of 10 carbon sources were different between Probio-M9 and HG-R7970-41. Transcriptomic and proteomic analyses revealed that this change in carbon metabolism of HG-R7970-41 mainly related to changes in phosphorylation and the glycolysis pathway. Based on the metabolic mechanism of different carbon sources and related gene cluster analysis, we found that the final metabolic activities of HG-R7970-41 and Probio-M9 were mainly regulated by PTS-specific membrane embedded permease, carbohydrate kinase and two rate-limiting enzymes (phosphofructokinase and pyruvate kinase) in the glycolysis pathway. The expanded culture test also confirmed that HG-R7970-41 had different metabolic characteristics from original strain. CONCLUSIONS: These results suggested that space environment could change carbon metabolism of Probio-M9. The new isolate (HG-R7970-41) showed a different carbon metabolism pattern from the original strain mainly by the regulation of two rate-limiting enzymes.


Asunto(s)
Carbono , Lacticaseibacillus rhamnosus , Carbono/metabolismo , Lacticaseibacillus rhamnosus/genética , Lacticaseibacillus rhamnosus/metabolismo , Lacticaseibacillus rhamnosus/aislamiento & purificación , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Mutación , Mutagénesis , Proteómica , Probióticos/metabolismo , Transcriptoma
5.
Food Funct ; 15(18): 9471-9487, 2024 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-39193624

RESUMEN

Equol is a highly active product of soy isoflavones produced by specific bacteria in the human or animal colon. However, equol production is influenced by differences in the gut flora carried by the body. Our previous research has shown that a synbiotic preparation comprising the probiotic Lactobacillus rhamnosus ATCC 7469 and the prebiotic lactulose can enhance equol production by modulating the intestinal flora. Nevertheless, the harsh environment of the gastrointestinal tract limits this capability by diminishing the number of probiotics reaching the colon. Microencapsulation of probiotics is an effective strategy to enhance their viability. In this study, probiotic gel microspheres (SA-S-CS) were prepared using an extrusion method, with sodium alginate (SA) and chitosan (CS) serving as the encapsulating materials. Scanning electron microscopy (SEM) was employed to observe the surface morphology and the internal distribution of bacteria within the microcapsules. The structural characteristics of the microcapsules were investigated using Fourier-transform infrared spectroscopy (FTIR) and X-ray diffraction (XRD). Furthermore, the thermal stability, storage stability, probiotic viability post-simulated gastrointestinal fluid treatment, and colon release rate were examined. Finally, the impact of probiotic microencapsulation on promoting equol production by the synbiotic preparation was assessed. The results indicated that the microcapsules exhibited a spherical structure with bacteria evenly distributed on the inner surface. Studies on thermal and storage stability showed that the number of viable cells in the probiotic microcapsule group significantly increased compared to the free probiotic group. Gastrointestinal tolerance studies revealed that after in vitro simulated gastrointestinal digestion, the amount of viable cells in the microcapsules was 7 log10 CFU g-1, demonstrating good gastrointestinal tolerance. Moreover, after incubation in simulated colonic fluid for 150 min, the release rate of probiotics reached 93.13%. This suggests that chitosan-coated sodium alginate microcapsules can shield Lactobacillus rhamnosus ATCC 7469 from the gastrointestinal environment, offering a novel model for synbiotic preparation to enhance equol production.


Asunto(s)
Equol , Lacticaseibacillus rhamnosus , Lactulosa , Probióticos , Simbióticos , Lacticaseibacillus rhamnosus/metabolismo , Lactulosa/metabolismo , Humanos , Alginatos/química , Composición de Medicamentos/métodos , Quitosano/química , Colon/microbiología , Colon/metabolismo
6.
PLoS One ; 19(8): e0304692, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39110654

RESUMEN

During the last years, growing interest in the use of mare's milk in food production is observed. The subject of the study was to evaluate the feasibility of mare's milk for the production of yogurt ice cream and synbiotic ice cream. Four variants of mare's milk ice cream were developed: ice cream with yogurt bacteria without inulin (YO) and with 2% of inulin (YO+I), synbiotic ice cream with 2% inulin and Lacticaseibacillus rhamnosus (LCR+I) and with Lactiplantibacillus plantarum (LP+I). Ice creams were enriched with inulin in order to evaluate its influence on the viability of LAB and on the product quality. Physicochemical, textural and sensory analyses were performed. Count of viable bacteria cells was also evaluated. Obtained ice creams did not differ in terms of protein, fat and total solids content (1.85-1.91%, 7.33-7.58% and 24.66-26.96% respectively), but differed in acidity. Ice cream YO, the only one without inulin, had the highest acidity, what suggests that inulin decrease this parameter. Regardless the type of LAB starter culture and inulin addition, samples had the same range of overrun (35.20-44.03%) and melting rate (73.49-79.87%). However the variant of ice cream influenced textural properties and colour parameters. All obtained mare's milk ice creams had high overall sensory quality. It was noticed, that ice cream with inulin had higher count of LAB (>7logCFU/g), than sample without inulin (>6logCFU/g). In conclusion, mare's milk may be considered as feasible raw material for yogurt ice cream and synbiotic ice cream production.


Asunto(s)
Helados , Leche , Simbióticos , Yogur , Helados/análisis , Helados/microbiología , Yogur/análisis , Yogur/microbiología , Animales , Simbióticos/análisis , Leche/química , Caballos , Femenino , Inulina , Lacticaseibacillus rhamnosus/metabolismo , Humanos , Microbiología de Alimentos
7.
Curr Microbiol ; 81(9): 271, 2024 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-39012492

RESUMEN

Probiotics and prebiotics have been considered as alternative approaches for promoting health. This study aimed to investigate the anticandidal potential of various probiotic Lactobacillus strains and their cell-free supernatants (CFSs). The study assessed the impact of inulin and some fruits as prebiotics on the growth of selected probiotic strains in relation to their anticandidal activity, production of short-chain fatty acids, total phenolic content, and antioxidant activity. Results revealed variations in anticandidal activity based on the specific strains and forms of probiotics used. Non-adjusted CFSs were the most effective against Candida strains, followed by probiotic cells and adjusted CFSs (pH 7). Lacticaseibacillus rhamnosus SD4, L. rhamnosus SD11 and L. rhamnosus GG displayed the strongest anticandidal activity. Non-adjusted CFSs from L. rhamnosus SD11, L. rhamnosus SD4 and L. paracasei SD1 exhibited notable anticandidal effects. The adjusted CFSs of L. rhamnosus SD11 showed the highest anticandidal activity against all non-albicans Candida (NAC) strains, whereas the others were ineffective. Supplementation of L. rhamnosus SD11 with prebiotics, particularly 2% (w/v) mangosteen, exhibited positive results in promoting probiotic growth, short-chain fatty acids production, total phenolic contents, and antioxidant activity, and the subsequent enhancing anticandidal activity against both C. albicans and NAC strains compared to conditions without prebiotics. In conclusion, both live cells and CFSs of tested strains, particularly L. rhamnosus SD11, exhibited the best anticandidal activity. Prebiotics supplementation, especially mangosteen, enhanced probiotic growth and beneficial metabolites against Candida growth. These finding suggested that probiotics and prebiotic supplementation may be an effective alternative treatment for Candida infections.


Asunto(s)
Lactobacillus , Prebióticos , Probióticos , Probióticos/farmacología , Lactobacillus/metabolismo , Candida/efectos de los fármacos , Candida/crecimiento & desarrollo , Antioxidantes/farmacología , Inulina/farmacología , Antifúngicos/farmacología , Ácidos Grasos Volátiles/metabolismo , Lacticaseibacillus rhamnosus/metabolismo , Fenoles/farmacología
8.
Med Microbiol Immunol ; 213(1): 14, 2024 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-38977511

RESUMEN

Mycobacterium tuberculosis, a lethal pathogen in human history, causes millions of deaths annually, which demands the development of new concepts of drugs. Considering this fact, earlier research has explored the anti-tuberculosis potential of a probiotic strain, Lactocaseibacillus rhamnosus PMC203, leading to a subsequent focus on the molecular mechanism involved in its effect, particularly on autophagy. In this current study, immunoblotting-based assay exhibited a remarkable expression of autophagy marker LC3-II in the PMC203 treated group compared to an untreated group. A remarkable degradation of p62 was also noticed within treated cells compared to control. Furthermore, the immunofluorescence-based assay showed significant fold change in fluorescence intensity for alexa-647-LC3 and alexa-488-LC3, whereas p62 was degraded noticeably. Moreover, lysosomal biogenesis generation was elevated significantly in terms of LAMP1 and acidic vesicular organelles. As a result, PMC203-induced autophagy played a vital role in reducing M. tuberculosis burden within the macrophages in treated groups compared to untreated group. A colony -forming unit assay also revealed a significant reduction in M. tuberculosis in the treated cells over time. Additionally, the candidate strain significantly upregulated the expression of autophagy induction and lysosomal biogenesis genes. Together, these results could enrich our current knowledge of probiotics-mediated autophagy in tuberculosis and suggest its implications for innovatively managing tuberculosis.


Asunto(s)
Autofagia , Lacticaseibacillus rhamnosus , Macrófagos , Mycobacterium tuberculosis , Probióticos , Mycobacterium tuberculosis/genética , Lacticaseibacillus rhamnosus/fisiología , Lacticaseibacillus rhamnosus/metabolismo , Macrófagos/microbiología , Humanos , Lisosomas/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Asociadas a Microtúbulos/genética , Carga Bacteriana , Tuberculosis/microbiología
9.
Adv Mater ; 36(35): e2402530, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38924628

RESUMEN

The compact three-dimensional (3D) structure of extracellular polymeric substances (EPS) within biofilms significantly hinders the penetration of antimicrobial agents, making biofilm eradication challenging and resulting in persistent biofilm-associated infections. To address this challenge, a solution is proposed: a probiotic bio-heterojunction (P-bioHJ) combining Lactobacillus rhamnosus with MXene (Ti3C2) quantum dots (MQDs)/FeS heterojunction. This innovation aims to break down the saccharides in EPS, enabling effective combat against biofilm-associated infections. Initially, the P-bioHJ targets saccharides through metabolic processes, causing the collapse of EPS and allowing infiltration into bacterial colonies. Simultaneously, upon exposure to near-infrared (NIR) irradiation, the P-bioHJ produces reactive oxygen species (ROS) and thermal energy, deploying physical mechanisms to combat bacterial biofilms effectively. Following antibiofilm treatment, the P-bioHJ adjusts the oxidative environment, reduces wound inflammation by scavenging ROS, boosts antioxidant enzyme activity, and mitigates the NF-κB inflammatory pathway, thereby accelerating wound healing. In vitro and in vivo experiments confirm the exceptional antibiofilm, antioxidant/anti-inflammatory, and wound-regeneration properties of P-bioHJ. In conclusion, this study provides a promising approach for treating biofilm-related infections.


Asunto(s)
Biopelículas , Matriz Extracelular de Sustancias Poliméricas , Lacticaseibacillus rhamnosus , Probióticos , Especies Reactivas de Oxígeno , Cicatrización de Heridas , Biopelículas/efectos de los fármacos , Probióticos/farmacología , Cicatrización de Heridas/efectos de los fármacos , Lacticaseibacillus rhamnosus/metabolismo , Animales , Especies Reactivas de Oxígeno/metabolismo , Matriz Extracelular de Sustancias Poliméricas/metabolismo , Matriz Extracelular de Sustancias Poliméricas/química , Antibacterianos/farmacología , Antibacterianos/química , Puntos Cuánticos/química , Ratones , Antioxidantes/farmacología , Antioxidantes/química
10.
PLoS One ; 19(6): e0306273, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38941324

RESUMEN

A growing increase in the number of serious infections caused by multidrug resistant bacteria (MDR) is challenging our society. Despite efforts to discover novel therapeutic options, few antibiotics targeting MDR have been approved by the Food and Drug Administration (FDA). Lactic acid bacteria have emerged as a promising therapeutic alternative due to their demonstrated ability to combat MDR pathogens in vitro. Our previous co-culture studies showed Lacticaseibacillus rhamnosus CRL 2244 as having a potent killing effect against carbapenem-resistant Acinetobacter baumannii (CRAB) strains. Here we report that cell-free conditioned media (CFCM) samples obtained from Lcb. rhamnosus CRL 2244 cultures incubated at different times display antimicrobial activity against 43 different pathogens, including CRAB, methicillin-resistant Staphylococcus aureus (MRSA) and carbapenemase Klebsiella pneumoniae (KPC)-positive strains. Furthermore, transwell and ultrafiltration analyses together with physical and chemical/biochemical tests showed that Lcb. rhamnosus CRL 2244 secretes a <3 kDa metabolite(s) whose antimicrobial activity is not significantly impaired by mild changes in pH, temperature and various enzymatic treatments. Furthermore, sensitivity and time-kill assays showed that the bactericidal activity of the Lcb. rhamnosus CRL 2244 metabolite(s) enhances the activity of some current FDA approved antibiotics. We hypothesize that this observation could be due to the effects of Lcb. rhamnosus CRL 2244 metabolite(s) on cell morphology and the enhanced transcriptional expression of genes coding for the phenylacetate (PAA) and histidine catabolic Hut pathways, metal acquisition and biofilm formation, all of which are associated with bacterial virulence. Interestingly, the extracellular presence of Lcb. rhamnosus CRL 2244 induced the transcription of the gene coding for the CidA/LgrA protein, which is involved in programmed cell death in some bacteria. Overall, the findings presented in this report underscore the promising potential of the compound(s) released by Lcb. rhamnosus CRL2244 as an alternative and/or complementary option to treat infections caused by A. baumannii as well as other MDR bacterial pathogens.


Asunto(s)
Antibacterianos , Farmacorresistencia Bacteriana Múltiple , Lacticaseibacillus rhamnosus , Lacticaseibacillus rhamnosus/metabolismo , Lacticaseibacillus rhamnosus/genética , Antibacterianos/farmacología , Farmacorresistencia Bacteriana Múltiple/efectos de los fármacos , Pruebas de Sensibilidad Microbiana , Acinetobacter baumannii/efectos de los fármacos , Sinergismo Farmacológico , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Medios de Cultivo Condicionados/farmacología , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética
11.
Microbiol Spectr ; 12(8): e0349723, 2024 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-38916329

RESUMEN

Bacteria are involved in numerous interactions during infection and among host-associated microbial populations. Salmonella enterica serovar Typhimurium is a foodborne pathogen of great importance as well as a model organism to study interactions within a microbial community. In this study, we found that S. Typhimurium becomes tolerant to azithromycin when co-cultured with lactobacilli strains. Similarly, acidified media, from cell-free supernatant of lactobacilli cultures for instance, also induced the tolerance of S. Typhimurium to azithromycin. The addition of membrane disruptors restored the normal sensitivity to azithromycin in acidified media, but not when lactobacilli were present. These results suggested that the acidification of the media led to modification in envelope homeostasis, but that a different mechanism promoted the tolerance to azithromycin in the presence of lactobacilli strains. To further understand how lactobacilli strains modify the sensitivity of S. Typhimurium to azithromycin, a high-throughput assay was performed using the single-gene deletion collection of the S. Typhimurium (1) in co-culture with Lacticaseibacillus rhamnosus and (2) in sterile acidic conditions (pH 5.5 media only). As expected, both screens identified genes involved in envelope homeostasis and membrane permeability. Our results also suggest that changes in the metabolism of S. Typhimurium induce the tolerance observed in the presence of L. rhamnosus. Our results thus highlight two different mechanisms by which lactobacilli induce the tolerance of S. Typhimurium to azithromycin.IMPORTANCEThis study provides valuable insights into the intricate interactions between bacteria during infections and within host-associated microbial communities. Specifically, it sheds light on the significant role of lactobacilli in inducing antibiotic tolerance in Salmonella enterica serovar Typhimurium, a critical foodborne pathogen and model organism for microbial community studies. The findings not only uncover the mechanisms underlying this antibiotic tolerance but also reveal two distinct pathways through which strains of lactobacilli might influence Salmonella's response to antibiotics. Understanding these mechanisms has the potential to enhance our knowledge of bacterial infections and may have implications for the development of strategies to combat antibiotic resistance in pathogens, such as Salmonella. Furthermore, our results underscore the necessity to explore beyond the direct antimicrobial effects of antibiotics, emphasizing the broader microbial community context.


Asunto(s)
Antibacterianos , Azitromicina , Lactobacillus , Salmonella typhimurium , Azitromicina/farmacología , Salmonella typhimurium/efectos de los fármacos , Salmonella typhimurium/genética , Salmonella typhimurium/crecimiento & desarrollo , Antibacterianos/farmacología , Lactobacillus/genética , Lactobacillus/efectos de los fármacos , Lactobacillus/metabolismo , Pruebas de Sensibilidad Microbiana , Farmacorresistencia Bacteriana/genética , Concentración de Iones de Hidrógeno , Infecciones por Salmonella/microbiología , Humanos , Lacticaseibacillus rhamnosus/genética , Lacticaseibacillus rhamnosus/efectos de los fármacos , Lacticaseibacillus rhamnosus/fisiología , Lacticaseibacillus rhamnosus/metabolismo
12.
Cell Mol Gastroenterol Hepatol ; 18(2): 101346, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38641207

RESUMEN

BACKGROUND & AIMS: Lacticaseibacillus rhamnosus GG (LGG) is the world's most consumed probiotic but its mechanism of action on intestinal permeability and differentiation along with its interactions with an essential source of signaling metabolites, dietary tryptophan (trp), are unclear. METHODS: Untargeted metabolomic and transcriptomic analyses were performed in LGG monocolonized germ-free mice fed trp-free or -sufficient diets. LGG-derived metabolites were profiled in vitro under anaerobic and aerobic conditions. Multiomic correlations using a newly developed algorithm discovered novel metabolites tightly linked to tight junction and cell differentiation genes whose abundances were regulated by LGG and dietary trp. Barrier-modulation by these metabolites were functionally tested in Caco2 cells, mouse enteroids, and dextran sulfate sodium experimental colitis. The contribution of these metabolites to barrier protection is delineated at specific tight junction proteins and enterocyte-promoting factors with gain and loss of function approaches. RESULTS: LGG, strictly with dietary trp, promotes the enterocyte program and expression of tight junction genes, particularly Ocln. Functional evaluations of fecal and serum metabolites synergistically stimulated by LGG and trp revealed a novel vitamin B3 metabolism pathway, with methylnicotinamide (MNA) unexpectedly being the most robust barrier-protective metabolite in vitro and in vivo. Reduced serum MNA is significantly associated with increased disease activity in patients with inflammatory bowel disease. Exogenous MNA enhances gut barrier in homeostasis and robustly promotes colonic healing in dextran sulfate sodium colitis. MNA is sufficient to promote intestinal epithelial Ocln and RNF43, a master inhibitor of Wnt. Blocking trp or vitamin B3 absorption abolishes barrier recovery in vivo. CONCLUSIONS: Our study uncovers a novel LGG-regulated dietary trp-dependent production of MNA that protects the gut barrier against colitis.


Asunto(s)
Colitis , Lacticaseibacillus rhamnosus , Probióticos , Triptófano , Animales , Lacticaseibacillus rhamnosus/metabolismo , Lacticaseibacillus rhamnosus/fisiología , Triptófano/metabolismo , Ratones , Humanos , Células CACO-2 , Probióticos/administración & dosificación , Colitis/metabolismo , Colitis/patología , Mucosa Intestinal/metabolismo , Enterocitos/metabolismo , Sulfato de Dextran , Niacinamida/farmacología , Niacinamida/metabolismo , Uniones Estrechas/metabolismo , Masculino , Modelos Animales de Enfermedad , Proteínas de Uniones Estrechas/metabolismo
13.
Braz J Microbiol ; 55(2): 1279-1286, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38652443

RESUMEN

Probiotic-containing foods are among the most appreciated functional foods; however, probiotic-based dairy products cannot be consumed by people who are lactose intolerant, allergic to milk, or vegetarian or vegan individuals. Thus, new non-dairy matrices have been tested for probiotics delivery. This study evaluated the growth and viability of Limosilactobacillus fermentum ATCC 23271 and Lacticaseibacillus rhamnosus ATCC 9595 in Pitanga juice (Eugenia uniflora L.). The effects of the fermentation on the antioxidant and anti-infective properties of the juice were also analyzed. The E. uniflora juice allowed lactobacilli growth without supplementation, reaching rates around 8.4 Log CFU/mL and producing organic acids (pH values < 4) after 72 h of fermentation. The strain remained viable after 35 days of refrigerated storage. Fermentation by these bacteria increases the antioxidant capacity of the juice. The central composite rotational design was employed to evaluate the effects of bacterial inoculum and pulp concentration on growth and organic acids production by L. fermentum ATCC 23271. The strain was viable and produced organic acids in all tested combinations. L. fermentum-fermented juice and its cell-free supernatant significantly increased the survival of Tenebrio molitor larvae infected by enteroaggregative Escherichia coli 042. The results obtained in this study provide more insights into the potential of Pitanga juice to develop a functional non-dairy probiotic beverage with antioxidant and anti-infective properties.


Asunto(s)
Antioxidantes , Eugenia , Fermentación , Limosilactobacillus fermentum , Probióticos , Antioxidantes/farmacología , Antioxidantes/metabolismo , Limosilactobacillus fermentum/metabolismo , Limosilactobacillus fermentum/crecimiento & desarrollo , Limosilactobacillus fermentum/fisiología , Limosilactobacillus fermentum/efectos de los fármacos , Probióticos/farmacología , Eugenia/química , Antiinfecciosos/farmacología , Jugos de Frutas y Vegetales/microbiología , Jugos de Frutas y Vegetales/análisis , Lacticaseibacillus rhamnosus/metabolismo , Lacticaseibacillus rhamnosus/crecimiento & desarrollo , Lacticaseibacillus rhamnosus/efectos de los fármacos , Viabilidad Microbiana/efectos de los fármacos
14.
Microb Cell Fact ; 23(1): 118, 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38659044

RESUMEN

BACKGROUND: Excessive alcohol consumption has been consistently linked to serious adverse health effects, particularly affecting the liver. One natural defense against the detrimental impacts of alcohol is provided by alcohol dehydrogenase (ADH) and acetaldehyde dehydrogenase (ALDH), which detoxify harmful alcohol metabolites. Recent studies have shown that certain probiotic strains, notably Lactobacillus spp., possess alcohol resistance and can produce these critical enzymes. Incorporating these probiotics into alcoholic beverages represents a pioneering approach that can potentially mitigate the negative health effects of alcohol while meeting evolving consumer preferences for functional and health-centric products. RESULTS: Five lactic acid bacteria (LAB) isolates were identified: Lactobacillus paracasei Alc1, Lacticaseibacillus rhamnosus AA, Pediococcus acidilactici Alc3, Lactobacillus paracasei Alc4, and Pediococcus acidilactici Alc5. Assessment of their alcohol tolerance, safety, adhesion ability, and immunomodulatory effects identified L. rhamnosus AA as the most promising alcohol-tolerant probiotic strain. This strain also showed high production of ADH and ALDH. Whole genome sequencing analysis revealed that the L. rhamnosus AA genome contained both the adh (encoding for ADH) and the adhE (encoding for ALDH) genes. CONCLUSIONS: L. rhamnosus AA, a novel probiotic candidate, showed notable alcohol resistance and the capability to produce enzymes essential for alcohol metabolism. This strain is a highly promising candidate for integration into commercial alcoholic beverages upon completion of comprehensive safety and functionality evaluations.


Asunto(s)
Alcohol Deshidrogenasa , Etanol , Probióticos , Humanos , Alcohol Deshidrogenasa/metabolismo , Alcohol Deshidrogenasa/genética , Etanol/metabolismo , Lactobacillus/metabolismo , Lactobacillus/genética , Lactobacillales/genética , Lactobacillales/metabolismo , Lacticaseibacillus rhamnosus/genética , Lacticaseibacillus rhamnosus/metabolismo , Aldehído Oxidorreductasas/metabolismo , Aldehído Oxidorreductasas/genética , Pediococcus acidilactici/metabolismo
15.
Microb Pathog ; 190: 106614, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38492825

RESUMEN

Lactic acid bacteria (LAB) have been recognized as safe microorganism that improve micro-flora disturbances and enhance immune response. A well-know traditional herbal medicine, Acanthopanax senticosus (As) was extensively utilized in aquaculture to improve growth performance and disease resistance. Particularly, the septicemia, skin wound and gastroenteritis caused by Aeromonas hydrophila threaten the health of aquatic animals and human. However, the effects of probiotic fermented with A. senticosus product on the immune regulation and pathogen prevention in fish remain unclear. Here, the aim of the present study was to elucidate whether the A. senticosus fermentation by Lactobacillus rhamnosus improve immune barrier function. The crucian carp were fed with basal diet supplemented with L. rhamnosus fermented A. senticosus cultures at 2 %, 4 %, 6 % and 8 % bacterial inoculum for 8 weeks. After trials, the weight gain rate (WGR), specific growth rate (SGR) were significantly increased, especially in LGG-6 group. The results confirmed that the level of the CAT, GSH-PX, SOD, lysozyme, and MDA was enhanced in fish received with probiotic fermented product. Moreover, the L. rhamnosus fermented A. senticosus cultures could trigger innate and adaptive immunity, including the up-regulation of the C3, C4, and IgM concentration. The results of qRT-PCR revealed that stronger mRNA transcription of IL-1ß, IL-10, IFN-γ, TNF-α, and MyD88 genes in the liver, spleen, kidney, intestine and gills tissues of fish treated with probiotic fermented with A. senticosus product. After infected with A. hydrophila, the survival rate of the LGG-2 (40 %), LGG-4 (50 %), LGG-6 (60 %), LGG-8 (50 %) groups was higher than the control group. Meanwhile, the pathological damage of the liver, spleen, head-kidney, and intestine tissues of probiotic fermentation-fed fish could be alleviated after pathogen infection. Therefore, the present work indicated that L. rhamnosus fermented A. senticosus could be regard as a potential intestine-target therapy strategy to protecting fish from pathogenic bacteria infection.


Asunto(s)
Aeromonas hydrophila , Antioxidantes , Carpas , Eleutherococcus , Fermentación , Enfermedades de los Peces , Lacticaseibacillus rhamnosus , Probióticos , Animales , Lacticaseibacillus rhamnosus/metabolismo , Carpas/microbiología , Probióticos/farmacología , Probióticos/administración & dosificación , Antioxidantes/metabolismo , Enfermedades de los Peces/prevención & control , Enfermedades de los Peces/microbiología , Enfermedades de los Peces/inmunología , Infecciones por Bacterias Gramnegativas/veterinaria , Infecciones por Bacterias Gramnegativas/prevención & control , Infecciones por Bacterias Gramnegativas/inmunología , Alimentación Animal , Inflamación/prevención & control , Citocinas/metabolismo , Acuicultura
16.
Small ; 20(26): e2306974, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38247174

RESUMEN

Adaptive laboratory evolution (ALE) can be used to make bacteria less susceptible to oxidative stress. An alternative to large batch scale ALE cultures is to use microfluidic platforms, which are often more economical and more efficient. Microfluidic ALE platforms have shown promise, but many have suffered from subpar cell passaging mechanisms and poor spatial definition. A new approach is presented using a microfluidic Evolution on a Chip (EVoc) design which progressively drives microbial cells from areas of lower H2O2 concentration to areas of higher concentration. Prolonged exposure, up to 72 h, revealed the survival of adaptive strains of Lacticaseibacillus rhamnosus GG, a beneficial probiotic often included in food products. After performing ALE on this microfluidic platform, the bacteria persisted under high H2O2 concentrations in repeated trials. After two progressive exposures, the ability of L. rhamnosus to grow in the presence of H2O2 increased from 1 mm H2O2 after a lag time of 31 h to 1 mm after 21 h, 2 mm after 28 h, and 3 mm after 42 h. The adaptive strains have different morphology, and gene expression compared to wild type, and genome sequencing revealed a potentially meaningful single nucleotide mutation in the protein omega-amidase.


Asunto(s)
Peróxido de Hidrógeno , Lacticaseibacillus rhamnosus , Microfluídica , Estrés Oxidativo , Probióticos , Estrés Oxidativo/efectos de los fármacos , Peróxido de Hidrógeno/farmacología , Peróxido de Hidrógeno/química , Peróxido de Hidrógeno/metabolismo , Lacticaseibacillus rhamnosus/metabolismo , Microfluídica/métodos , Evolución Molecular Dirigida/métodos
17.
Food Funct ; 13(23): 12144-12155, 2022 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-36326009

RESUMEN

Lactobacillus rhamnosus GG (LGG) is a well-known probiotic widely used in foods and drugs. It has been reported that LGG can improve bowel dysfunction in gastrointestinal motility disorders, such as constipation; however, the specific mechanisms remain unclear. The colonic mucus layer is mainly composed of mucin secreted by goblet cells, which plays important roles in lubricating colonic contents and maintaining normal defecation function. It has been reported that increased mucin production is beneficial for relieving constipation symptoms. In this study, we aimed to investigate the role of LGG in regulating intestinal mucin production and the associated mechanisms. Six-week-old C57BL/6J mice were randomized into 3 groups, and were treated with De-Man Rogosa and Sharpe broth (MRS group), tegaserod maleate (tegaserod group) and LGG supernatant (LGGs group) by gavage, respectively. After treatments, defecation parameters, intestinal mucin-2 (MUC2) and serotonin 4 receptor (5-HT4R), goblet cells, and microbiota composition of the mice in each group were assessed. In comparison with the MRS group, higher fecal water content and increased fecal pellet number were found in the tegaserod group and LGGs group. Moreover, LGGs increased the number of goblet cells and upregulated the expression of 5-HT4R and MUC2 in the mouse colon. In addition, Alcian Blue Periodic acid Schiff staining showed that activated 5-HT4R enhanced intestinal MUC2 secretion. Further exploration of the mechanism discovered that LGGs upregulated intestinal S100A10, which was found to be involved in regulating 5-HT4R expression. Furthermore, gut microbiota analysis showed the higher abundance of Alistipes, Allobaculum, Desulfovibrio, and Clostridium XlVa in the LGGs group, which have been reported to be involved in regulating gut motility and the intestinal barrier, and alleviating bowel dysfunction. Interestingly, gut dysbiosis was present in the tegaserod group. It is noteworthy that the fecal microbiota transplanted from LGGs-treated mice significantly improved the gut dysmotility in a constipation mouse model. Our results suggested that LGGs could upregulate 5-HT4R to promote MUC2 production, as well as modulate the gut microbiota, thus improving the defecation function in mice. This finding might provide evidence for the application of diet supplementary LGG in relieving gastrointestinal motility disorders.


Asunto(s)
Microbioma Gastrointestinal , Enfermedades Intestinales , Lacticaseibacillus rhamnosus , Probióticos , Ratones , Animales , Lacticaseibacillus rhamnosus/metabolismo , Mucinas/metabolismo , Mucosa Intestinal/metabolismo , Ratones Endogámicos C57BL , Mucina 2/genética , Mucina 2/metabolismo , Probióticos/uso terapéutico , Enfermedades Intestinales/metabolismo , Estreñimiento
18.
Nutrients ; 14(22)2022 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-36432537

RESUMEN

To further explore and improve the mechanism of probiotics to alleviate the disorder of lipid metabolism, transcriptomic and metabolomic with bioinformatic analysis were combined. In the present study, we successfully established a rat model of lipid metabolism disorder using a high-fat diet. Intervention with Lactobacillus rhamnosus hsryfm 1301 fermented milk resulted in a significant reduction in body weight, serum free fatty acid and blood lipid levels (p < 0.05), which predicted that the lipid metabolism disorder was alleviated in rats. Metabolomics and transcriptomics identified a total of 33 significantly different metabolites and 183 significantly different genes screened in the intervention group compared to the model group. Comparative analysis of Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway annotations identified a total of 61 pathways in which differential metabolites and genes were jointly involved, with linoleic acid metabolism, glycine, serine and threonine metabolism and glutamatergic synapse in both transcriptome and metabolome being found to be significantly altered (p < 0.05). Lactobacillus rhamnosus hsryfm 1301 fermented milk was able to directly regulate lipid metabolism disorders by regulating the metabolic pathways of linoleic acid metabolism, glycerophospholipid metabolism, fatty acid biosynthesis, alpha-linolenic acid metabolism, fatty acid degradation, glycerolipid metabolism and arachidonic acid metabolism. In addition, we found that Lactobacillus rhamnosus hsryfm 1301 fermented milk indirectly regulates lipid metabolism through regulating amino acid metabolism, the nervous system, the endocrine system and other pathways. Lactobacillus rhamnosus hsryfm 1301 fermented milk could alleviate the disorders of lipid metabolism caused by high-fat diet through multi-target synergy.


Asunto(s)
Lacticaseibacillus rhamnosus , Trastornos del Metabolismo de los Lípidos , Ratas , Animales , Dieta Alta en Grasa/efectos adversos , Lacticaseibacillus rhamnosus/metabolismo , Metabolismo de los Lípidos , Leche/metabolismo , Ácido Linoleico/metabolismo , Trastornos del Metabolismo de los Lípidos/metabolismo
19.
Int J Mol Sci ; 23(19)2022 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-36232464

RESUMEN

The intestinal barrier is vital for preventing inflammatory bowel disease (IBD). The objectives of this study were to assess whether the Lactobacillus rhamnosus CY12 could alleviate oxidative stress, inflammation, and the disruption of tight junction (TJ) barrier functions induced by lipopolysaccharide (LPS), and therefore to explore the potential underlying molecular mechanisms. Our results showed that LPS-induced Cancer coli-2 (Caco-2) cells significantly increased the levels of reactive oxygen species (ROS), lactate dehydrogenase, inflammatory cytokines interleukin-1ß, interleukin-6, interleukin-8, and tumor necrosis factor-α (IL-1ß, IL-6, IL-8, and TNF-α), and the cell apoptosis rate while decreasing the levels of TJ proteins occludin, zonula occludens-1 (ZO-1), and claudin and antioxidant enzymes, such as catalase, superoxide dismutase, and glutathione peroxidase(CAT, SOD, and GSH-Px) (p < 0.05). However, Lactobacillus rhamnosus CY12 could relieve cytotoxicity, apoptosis, oxidative stress, and pro-inflammatory cytokine expressions, and also inhibit the Toll-like receptor 4/nuclear factor kappa-B(TLR4/NF-κB) signaling pathway. Furthermore, the gene expression of antioxidant enzymes, as well as the mRNA and protein expressions of TJ proteins, was improved. Particularly, the concentration of 108 cfu/mL significantly prevented the inflammatory injury induced by LPS in Caco-2 cells (p < 0.05). These findings support a potential application of Lactobacillus rhamnosus CY12 as a probiotic to prevent LPS-induced intestinal injury and treat intestinal barrier dysfunction.


Asunto(s)
Lacticaseibacillus rhamnosus , Proteínas de Uniones Estrechas , Antioxidantes/metabolismo , Antioxidantes/farmacología , Células CACO-2 , Catalasa/metabolismo , Claudinas/metabolismo , Glutatión Peroxidasa/metabolismo , Humanos , Inflamación , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Lactato Deshidrogenasas/metabolismo , Lacticaseibacillus rhamnosus/metabolismo , Lipopolisacáridos/farmacología , FN-kappa B/metabolismo , Ocludina/genética , Ocludina/metabolismo , Estrés Oxidativo , ARN Mensajero/metabolismo , Especies Reactivas de Oxígeno , Superóxido Dismutasa/metabolismo , Proteínas de Uniones Estrechas/metabolismo , Receptor Toll-Like 4/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
20.
Microb Pathog ; 173(Pt A): 105831, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36280164

RESUMEN

The Th1 cytokines production associated to signal transducer and activator of transcription 4 (STAT4) signaling amplifies the pro-inflammatory response in acute respiratory distress syndrome (ARDS). The anti-inflammatory action of commensal bacteria has been described as a secondary effect dependent on IL-10- secreting Treg cells that can act in organs far from the gut, including the lung. Despite it, no data is showing whether the previous reported anti-inflammatory action of probiotics is associated with its immunomodulatory effect dependent on Treg cells in a murine model of ARDS. Therefore, herein we focused on the short-term pretreatment effect with Lacticaseibacillus rhamnosus (Lr) in STAT4-associated Th1 cytokines as well as in population of IL-10- secreting Treg cells in a murine model of ARDS. Assays were performed in experimental groups divided into control, LPS, and Lr + LPS. Total and differential cells from bronchoalveolar lavage fluid (BALF) were counted through microscopy and the IL-10, IL-12, IL-17, IL-18, IL-22, IL-23, IL-27, IFN-γ, MMP-9, and TIMP were measured by ELISA. The peribronchial neutrophils were assessed using morphometry and for pulmonary edema was measured by Evans blue dye extravasation. The gene expression for STAT4, T-bet, STAT3, RORÉ£t, STAT5, and Foxp3 were measured by Real-Time PCR. Population of IL-10-secreting Treg cells was performed by flow cytometer. Data showed that pretreatment with Lr attenuated the number of inflammatory cells, secretion of both Th1 and Th17 cytokines, expression of STAT4/T-bet and STAT3/RORÉ£t in lung as well as alterations in lung morphometry. Otherwise, Lr was not efficient to restore mRNA expression for STAT5 and Foxp3 expression and population of IL-10-secreting Treg cells. Thus, beneficial effect of short-term pretreatment with Lr in murine model of ARDS is not dependent on an increased immunomodulatory action of IL-10-secreting Treg cells, however the anti-inflammatory effect of Lr has as target the Th1 and Th17 cytokines as well as signaling involving the STAT4/T-bet and STAT3/RORÉ£t.


Asunto(s)
Lacticaseibacillus rhamnosus , Neumonía , Síndrome de Dificultad Respiratoria , Ratones , Animales , Citocinas/metabolismo , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Interleucina-10/metabolismo , Factor de Transcripción STAT5/metabolismo , Factor de Transcripción STAT5/farmacología , Lipopolisacáridos/farmacología , Modelos Animales de Enfermedad , Células Th17 , Linfocitos T Reguladores , Lacticaseibacillus rhamnosus/metabolismo , Factores de Transcripción Forkhead/metabolismo , Antiinflamatorios/farmacología , Factor de Transcripción STAT4
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA