Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 398
Filtrar
Más filtros

Intervalo de año de publicación
1.
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
2.
Helicobacter ; 29(4): e13108, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39021274

RESUMEN

BACKGROUND: Helicobacter pylori infection-associated gastric adenocarcinoma is influenced by various factors, including the digestive microbiota. Lactic acid bacteria role in digestive carcinogenesis has been discussed, and some Lactobacillaceae family species have been shown to act against H. pylori-induced inflammation and colonization. However, their effects on H. pylori-related carcinogenesis have not yet been studied. Lactobacillaceae family effects on the epithelial-to-mesenchymal transition (EMT), emergence of cells with cancer stem cell (CSC) properties and the pro-inflammatory response of gastric epithelial cells to H. pylori infection were investigated. MATERIALS AND METHODS: A co-culture model of AGS gastric epithelial cells infected with a carcinogenic strain of H. pylori associated with 18 different probiotic strains candidates were used. Different EMT indicators and CSC properties were studied, including quantification of the mesenchymal phenotype, tumorsphere formation, EMT marker expression, and tight junction evaluation with immunofluorescence microscopy. The effect of the strains on the pro-inflammatory response to H. pylori was also evaluated by quantifying interleukin-8 (IL-8) production using ELISA. RESULTS: Among the strains tested, Lactobacillus gasseri BIO6369 and Lacticaseibacillus rhamnosus BIO5326 induced a 30.6% and 38.4% reduction in the mesenchymal phenotype, respectively, caused a significant decrease in Snail and Zeb1 EMT marker expression and prevented the loss of tight junctions induced by H. pylori infection. A separate co-culture with a Boyden chamber maintained the effects induced by the two strains. H. pylori-induced IL-8 production was also significantly reduced in the presence of L. gasseri BIO6369 and L. rhamnosus BIO5326. CONCLUSION: Lactobacillus gasseri BIO6369 and L. rhamnosus BIO5326 strains decreased epithelial-to-mesenchymal transition and inflammation induced by H. pylori infection, suggesting that these species may have a protective effect against H. pylori-induced gastric carcinogenesis.


Asunto(s)
Células Epiteliales , Transición Epitelial-Mesenquimal , Infecciones por Helicobacter , Helicobacter pylori , Lacticaseibacillus rhamnosus , Lactobacillus gasseri , Probióticos , Neoplasias Gástricas , Humanos , Infecciones por Helicobacter/microbiología , Infecciones por Helicobacter/patología , Helicobacter pylori/fisiología , Helicobacter pylori/patogenicidad , Neoplasias Gástricas/microbiología , Neoplasias Gástricas/patología , Lacticaseibacillus rhamnosus/fisiología , Células Epiteliales/microbiología , Técnicas de Cocultivo , Carcinogénesis
3.
J Appl Microbiol ; 135(6)2024 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-38857885

RESUMEN

AIMS: Klebsiella pneumoniae, an important opportunistic pathogen of nosocomial inflection, is known for its ability to form biofilm. The purpose of the current study is to assess how co- or mono-cultured probiotics affect K. pneumoniae's ability to produce biofilms and investigate the potential mechanisms by using a polyester nonwoven chemostat and a Caco-2 cell line. METHODS AND RESULTS: Compared with pure cultures of Lactobacillus rhamnosus and Lactobacillus sake, the formation of K. pneumoniae biofilm was remarkably inhibited by the mixture of L. rhamnosus, L. sake, and Bacillus subtilis at a ratio of 5:5:1 by means of qPCR and FISH assays. In addition, Lactobacillus in combination with B. subtilis could considerably reduce the adherence of K. pneumoniae to Caco-2 cells by using inhibition, competition, and displacement assays. According to the RT-PCR assay, the adsorption of K. pneumoniae to Caco-2 cells was effectively inhibited by the co-cultured probiotics, leading to significant reduction in the expression of proinflammatory cytokines induced by K. pneumoniae. Furthermore, the HPLC and RT-PCR analyses showed that the co-cultured probiotics were able to successfully prevent the expression of the biofilm-related genes of K. pneumoniae by secreting plenty of organic acids as well as the second signal molecule (c-di-GMP), resulting in inhibition on biofilm formation. CONCLUSION: Co-culture of L. sake, L. rhamnosus, and B. subtilis at a ratio of 5:5:1 could exert an antagonistic effect on the colonization of pathogenic K. pneumoniae by down-regulating the expression of biofilm-related genes. At the same time, the co-cultured probiotics could effectively inhibit the adhesion of K. pneumoniae to Caco-2 cells and block the expression of proinflammatory cytokines induced by K. pneumoniae.


Asunto(s)
Biopelículas , Técnicas de Cocultivo , Klebsiella pneumoniae , Probióticos , Biopelículas/crecimiento & desarrollo , Klebsiella pneumoniae/fisiología , Humanos , Probióticos/farmacología , Células CACO-2 , Bacillus subtilis/fisiología , Bacillus subtilis/genética , Lacticaseibacillus rhamnosus/fisiología , Adhesión Bacteriana , Lactobacillus/fisiología , Citocinas/metabolismo
4.
Int J Mol Sci ; 25(13)2024 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-39000222

RESUMEN

Persisters are antibiotic-tolerant bacteria, playing a role in the recalcitrance and relapse of many bacterial infections, including P. aeruginosa pulmonary infections in Cystic Fibrosis (CF) patients. Among novel antimicrobial strategies, the use of probiotics and their products is emerging as a particularly promising approach. The aim of this study was to evaluate the anti-persisters activity of culture filtrate supernatants of Lacticaseibacillus rhamnosus (LRM-CFS) against P. aeruginosa in artificial sputum medium (ASM), which resembles the CF lung environment. Planktonic persisters of two clinical strains of P. aeruginosa (PaCF1 and PaCF4) were obtained following two different procedures: (i) exposing stationary-phase cultures to cyanide m-chlorophenylhydrazone (CCCP) in LB medium; (ii) incubating stationary-phase cultures with high doses of tobramycin (128-fold MIC) in ASM. In addition, persisters from biofilm were obtained by exposing 48 h old biofilm of P. aeruginosa to 128 x MIC of ciprofloxacin. LRM-CFS at dilutions of 1:6 and 1:4 resulted in being bactericidal in ASM against both PaCF1 and PaCF4 persisters obtained after CCCP or tobramycin treatment. Moreover, LRM-CFS at dilution 1:4 caused a reduction of antibiotic-tolerant bacteria in the biofilm of both P. aeruginosa strains. Overall, LRM-CFS represents a promising adjuvant therapeutic strategy against P. aeruginosa recalcitrant infections in CF patients.


Asunto(s)
Antibacterianos , Biopelículas , Lacticaseibacillus rhamnosus , Pruebas de Sensibilidad Microbiana , Pseudomonas aeruginosa , Esputo , Pseudomonas aeruginosa/efectos de los fármacos , Esputo/microbiología , Biopelículas/efectos de los fármacos , Biopelículas/crecimiento & desarrollo , Humanos , Lacticaseibacillus rhamnosus/fisiología , Antibacterianos/farmacología , Fibrosis Quística/microbiología , Medios de Cultivo/farmacología , Medios de Cultivo/química , Medios de Cultivo Condicionados/farmacología , Infecciones por Pseudomonas/tratamiento farmacológico , Infecciones por Pseudomonas/microbiología , Tobramicina/farmacología
5.
Br J Cancer ; 126(10): 1421-1431, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35091695

RESUMEN

BACKGROUND: Gut microbiota dysbiosis is closely related to the progression of colorectal cancer. Our previous study revealed that early life colonisation with Lactobacillus rhamnosus GG (LGG) had long-term positive effects on health. We sought to investigate whether early life LGG colonisation could inhibit intestinal tumour formation in offspring. METHODS: Adult C57BL/6 female mice were mated with Apcmin/+ male mice. Pregnant mice with the same conception date received 108 cfu live or fixed LGG from day 18 of pregnancy until natural delivery. After genotyping, offspring mice received 107 cfu of live or fixed LGG for 0-5 days after birth. RESULTS: Early life LGG colonisation significantly promoted intestinal development, inhibited low-grade intestinal inflammation and altered the gut microbiota composition of offspring in the weaning period (3 week old). Notably, early life LGG colonisation reduced the multiplicity of intestinal tumours in adulthood (12 week old), possibly due to inhibition of Wnt signalling and promotion of tumour cell apoptosis. Importantly, at the genus level, Bifidobacterium and Anaeroplasma with potential anti-tumour effects were increased in adulthood, while Peptostreptococcus, which potentially contributes to tumour formation, was decreased. CONCLUSIONS: Early life LGG colonisation inhibited the intestinal tumour formation of offspring in adulthood.


Asunto(s)
Microbioma Gastrointestinal , Neoplasias Intestinales , Lacticaseibacillus rhamnosus , Probióticos , Adulto , Animales , Femenino , Humanos , Neoplasias Intestinales/prevención & control , Lacticaseibacillus rhamnosus/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Embarazo , Probióticos/farmacología
6.
Appl Environ Microbiol ; 88(3): e0116521, 2022 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-34818099

RESUMEN

Single-cell isolation and cultivation play an important role in studying physiology, gene expression, and functions of microorganisms. A series of single-cell isolation technologies have been developed, among which single-cell ejection technology is one of the most promising. Single-cell ejection technology has applied laser-induced forward transfer (LIFT) techniques to isolate bacteria, but the viability (or recovery rate) of cells after sorting has not been clarified in current research. In this work, to keep the cells alive as long as possible, we propose a three-layer LIFT system (top layer, 25-nm aluminum film; second layer, 3 µm agar media; third layer, liquid containing bacteria) for the isolation and cultivation of single Gram-negative (Escherichia coli), Gram-positive (Lactobacillus rhamnosus GG [LGG]), and eukaryotic (Saccharomyces cerevisiae) microorganisms. The experiment results showed that the average survival rates for ejected pure single cells were 63% for Saccharomyces cerevisiae, 22% for E. coli DH5α, and 74% for LGG. In addition, we successfully isolated and cultured the green fluorescent protein (GFP)-expressing E. coli JM109 from a mixture containing complex communities of soil bacteria by fluorescence signal. The average survival rate of E. coli JM109 was demonstrated to be 25.3%. In this study, the isolated and cultured single colonies were further confirmed by colony PCR and sequencing. Such precise sorting and cultivation techniques of live single microbial cells could be coupled with other microscopic approaches to isolate single microorganisms with specific functions, revealing their roles in the natural community. IMPORTANCE We developed a laser-induced forward transfer (LIFT) technology to accurately isolate single live microbial cells. The cultivation recovery rates of the ejected single cells were 63% for Saccharomyces cerevisiae, 22% for E. coli DH5α, and 74% for Lactobacillus rhamnosus GG (LGG). With coupled LIFT with a fluorescence microscope, we demonstrated that single cells of GFP-expressing E. coli JM109 were sorted according to fluorescence signal from a complex community of soil bacteria and subsequently cultured with 25% cultivation recovery rate. This single-cell live sorting technology could isolate single microbes with specific functions, revealing their roles in the natural community.


Asunto(s)
Escherichia coli , Lacticaseibacillus rhamnosus , Bacterias/genética , Escherichia coli/genética , Lacticaseibacillus rhamnosus/fisiología , Rayos Láser , Tecnología
7.
Toxicol Appl Pharmacol ; 439: 115923, 2022 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-35176292

RESUMEN

BACKGROUND: Intestinal epithelial barrier disruption and bacterial translocation exacerbates the progression of alcoholic liver disease. Lactobacillus rhamnosus GG (LGG), a probiotic, has been shown benefits in chronic liver disease and in regulating gut dysbiosis. Previous studies showed the protective roles of LGG in ethanol-disrupted gut barrier functions and liver injury. Inosine, a metabolite produced by intestinal bacteria, has the anti-inflammatory and immunregulatory functions. In this study, the synergistic effect of LGG and inosine was investigated in a mouse model of alcohol-induced liver disease (ALD). METHODS: Male C57BL/6 mice were fed with a Lieber-DeCarli diet containing 5% alcohol for four weeks to establish a model of alcohol-induced liver injury. LGG or a combination of LGG and inosine were administrated orally to explore a new therapeutic method for alcohol-induced liver disease and to investigate the underlying mechanisms. Liver damage was evaluated by transaminases and pathological changes. Tight junction proteins, composition of the gut microbiome, cytokines, lipopolysaccharides (LPS), glutathione (GSH), superoxide dismutase (SOD), malondialdehyde (MDA), F4/80+ macrophages, as well as p38, Jun N-terminal kinase (JNK), were determined by qRT-PCR, RNAseq, ELISA, IHC and western blot. Regulatory T (Treg) cells were characterized by positive staining of CD4, CD25 and Foxp3 using flow cytometry. IFN-γ-producing CD4+ T (Th1) cells were examined by intracellular cytokine staining. RESULTS: Alcohol consumption induced elevated liver enzymes, steatosis and inflammation, while LGG combined with inosine treatment was more significant to ameliorate these symptoms compared with LGG alone. When LGG combined with inosine were administered to ALD mice, intestinal microecology significantly improved reflected by intestinal villi and tight junction proteins recovery and the restoration of intestinal flora. Combined therapy inhibited phosphorylation of p38 and JNK to alleviate hepatic inflammation. Moreover, flow cytometry analysis showed that long-term excessive alcohol consumption reduced Tregs population while increased Th1 population, which was restored by a combination of LGG and inosine treatment. CONCLUSIONS: The findings from the study indicate that the combined LGG and inosine treatment ameliorates ALD by improving the gut ecosystem, intestinal barrier function, immune homeostasis and liver injury.


Asunto(s)
Enfermedad Hepática Crónica Inducida por Sustancias y Drogas , Lacticaseibacillus rhamnosus , Hepatopatías Alcohólicas , Animales , Ecosistema , Etanol/toxicidad , Inflamación , Inosina , Lacticaseibacillus rhamnosus/fisiología , Hepatopatías Alcohólicas/patología , Hepatopatías Alcohólicas/prevención & control , Masculino , Ratones , Ratones Endogámicos C57BL , Linfocitos T Reguladores , Células TH1 , Proteínas de Uniones Estrechas
8.
FASEB J ; 35(12): e21992, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34719821

RESUMEN

The colonic epithelial barrier is vital to preserve gut and host health by maintaining the immune homeostasis between host and microbes. The mechanisms underlying beneficial or harmful host-microbe interactions are poorly understood and impossible to study in vivo given the limited accessibility and ethical constraints. Moreover, existing in vitro models lack the required cellular complexity for the routine, yet profound, analysis of the intricate interplay between different types of host and microbial cells. We developed and characterized a broadly applicable, easy-to-handle in vitro triple coculture model that combines chemically-induced macrophage-like, goblet and epithelial cells covered by a mucus layer, which can be coincubated with complex human-derived gut microbiota samples for 16 h. Comparison with a standard epithelial monolayer model revealed that triple cocultures produce thicker mucus layers, morphologically organize in a network and upon exposure to human-derived gut microbiota samples, respond via pro-inflammatory cytokine production. Both model systems, however, were not suffering from cytotoxic stress or different microbial loads, indicating that the obtained endpoints were caused by the imposed conditions. Addition of the probiotic Lactobacillus rhamnosus GG to assess its immunomodulating capacity in the triple coculture slightly suppressed pro-inflammatory cytokine responses, based on transcriptomic microarray analyses. TNF conditioning of the models prior to microbial exposure did not cause shifts in cytokines, suggesting a strong epithelial barrier in which TNF did not reach the basolateral side. To conclude, the triple coculture model is tolerable towards manipulations and allows to address mechanistic host-microbe research questions in a stable in vitro environment.


Asunto(s)
Técnicas de Cocultivo/métodos , Colon/inmunología , Células Epiteliales/inmunología , Microbioma Gastrointestinal , Interacciones Microbiota-Huesped , Mucosa Intestinal/inmunología , Macrófagos/inmunología , Colon/citología , Colon/metabolismo , Colon/microbiología , Células Epiteliales/citología , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Humanos , Mucosa Intestinal/citología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Lacticaseibacillus rhamnosus/fisiología , Macrófagos/citología , Macrófagos/metabolismo , Macrófagos/microbiología , Transcriptoma
9.
Nutr Cancer ; 74(7): 2622-2631, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34918611

RESUMEN

The microbiome of the intestinal system is well-known as a modulatory factor. Having a balanced status of microbiota could help to prevent diseases, especially cancers related to the gastrointestinal system. We investigated the effects of Lactobacillus rhamnosus (Lr) and capecitabine on tumor size and physiologic features, such as bodyweight, liver enzymes, and blood profile, in a subcutaneously induced cancer model using CT-26 murine colon carcinoma cells. We divided 48 male Balb/c inbred mice into six groups. Lr had been orally pre-inoculated to the mice for 14 day consecutively. CT-26 cells were implanted subcutaneously into the mice's flank. Following the injection of cancer cells, Lr was inoculated to the mice three times per week for four weeks. Capecitabine was inoculated in the third week after the induction of cancer. The tumor size was significantly decreased in treated groups in comparison to the cancer group (1174.5 ± 63.8, 1119.2 ± 86.3, and 985.6 ± 48 mm3 vs. 1674.2 ± 66 mm3, P < 0.0001). Data showed that Lr and capecitabine enhanced Bax/Bcl-2 ratio and caspase-3 level compared to cancer group (p < 0.0001). White blood cells (WBCs) were significantly decreased in the capecitabine group compared to probiotic group (P < 0.05). Measurement of bodyweight, liver enzymes, and interleukin-6 (IL-6) level showed that Lr, in addition to preventive and therapeutic effects, might have protective effects against chemotherapy side effects. Preventing WBCs' reduction, protecting mice from losing weight, induction of apoptosis, and enhancing the serum level of IL-6 indicated that Lr might be associated with better management of colorectal cancer and chemotherapy side effects.


Asunto(s)
Neoplasias del Colon , Lacticaseibacillus rhamnosus , Probióticos , Animales , Capecitabina/farmacología , Neoplasias del Colon/patología , Interleucina-6 , Lacticaseibacillus rhamnosus/fisiología , Masculino , Ratones , Ratones Endogámicos BALB C , Probióticos/farmacología
10.
Lett Appl Microbiol ; 75(5): 1151-1159, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35775865

RESUMEN

Ionizing radiation is widely applied in food production as preservation technology and for correction of the gut microbiome of cancer patients, rescuers, astronauts etc. Lactic acid bacteria (LAB) can be used for the same reason. The main goal of this study was to investigate the effect of irradiation on some activities of Lactobacillus rhamnosus MDC 9661 and its effect on the survival of irradiated rats. The results indicate that both ultraviolet (during 45 min) and X-ray irradiations (with 2 Sv) decreased the CFU and the antibacterial activity of the strain. Higher than 700 Sv dose of X-ray irradiation resulted in the total inhibition of antibacterial activity with the total reduction of colony forming units less than 10 cells ml-1 , while irradiated with 1000 Sv dose L. rhamnosus MDC 9661 did not lose its proteolytic activity. It was also shown that L. rhamnosus MDC 9661 was not immunogenic in the organism of the rats and cannot lead to the development of autoimmune responses. L. rhamnosus MDC 9661 demonstrated the necessary properties for probiotics and can be effectively used for the correction of the gut microbiome of all target groups. The co-aggregation of the cells is one of the mechanisms for resistance of LAB to irradiation.


Asunto(s)
Lacticaseibacillus rhamnosus , Lactobacillales , Probióticos , Ratas , Animales , Lacticaseibacillus rhamnosus/fisiología , Rayos X , Probióticos/farmacología , Antibacterianos/farmacología
11.
Int J Mol Sci ; 23(5)2022 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-35269654

RESUMEN

A chronic kidney disease (CKD) causes uremic toxin accumulation and gut dysbiosis, which further induces gut leakage and worsening CKD. Lipopolysaccharide (LPS) of Gram-negative bacteria and (1➔3)-ß-D-glucan (BG) of fungi are the two most abundant gut microbial molecules. Due to limited data on the impact of intestinal fungi in CKD mouse models, the influences of gut fungi and Lacticaseibacillus rhamnosus L34 (L34) on CKD were investigated using oral C. albicans-administered 5/6 nephrectomy (5/6Nx) mice. At 16 weeks post-5/6Nx, Candida-5/6Nx mice demonstrated an increase in proteinuria, serum BG, serum cytokines (tumor necrotic factor-α; TNF-α and interleukin-6), alanine transaminase (ALT), and level of fecal dysbiosis (Proteobacteria on fecal microbiome) when compared to non-Candida-5/6Nx. However, serum creatinine, renal fibrosis, or gut barrier defect (FITC-dextran assay and endotoxemia) remained comparable between Candida- versus non-Candida-5/6Nx. The probiotics L34 attenuated several parameters in Candida-5/6Nx mice, including fecal dysbiosis (Proteobacteria and Bacteroides), gut leakage (fluorescein isothiocyanate (FITC)-dextran), gut-derived uremic toxin (trimethylamine-N-oxide; TMAO) and indoxyl sulfate; IS), cytokines, and ALT. In vitro, IS combined with LPS with or without BG enhanced the injury on Caco-2 enterocytes (transepithelial electrical resistance and FITC-dextran permeability) and bone marrow-derived macrophages (supernatant cytokines (TNF-α and interleukin-1 ß; IL-1ß) and inflammatory genes (TNF-α, IL-1ß, aryl hydrocarbon receptor, and nuclear factor-κB)), compared with non-IS activation. These injuries were attenuated by the probiotics condition media. In conclusion, Candida administration worsens kidney damage in 5/6Nx mice through systemic inflammation, partly from gut dysbiosis-induced uremic toxins, which were attenuated by the probiotics. The additive effects on cell injury from uremic toxin (IS) and microbial molecules (LPS and BG) on enterocytes and macrophages might be an important underlying mechanism.


Asunto(s)
Lacticaseibacillus rhamnosus , Insuficiencia Renal Crónica , Uremia , Animales , Células CACO-2 , Candida , Citocinas , Disbiosis/microbiología , Glucanos , Humanos , Lacticaseibacillus rhamnosus/fisiología , Lipopolisacáridos/toxicidad , Ratones , Factor de Necrosis Tumoral alfa/efectos adversos , Tóxinas Urémicas
12.
Int J Mol Sci ; 23(3)2022 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-35163782

RESUMEN

Dental caries is caused by the formation of cariogenic biofilm, leading to localized areas of enamel demineralization. Streptococcus mutans, a cariogenic pathogen, has long been considered as a microbial etiology of dental caries. We hypothesized that an antagonistic approach using a prebiotic collagen peptide in combination with probiotic Lactobacillus rhamnosus would modulate the virulence of this cariogenic biofilm. In vitro S. mutans biofilms were formed on saliva-coated hydroxyapatite discs, and the inhibitory effect of a combination of L. rhamnosus and collagen peptide on S. mutans biofilms were evaluated using microbiological, biochemical, confocal imaging, and transcriptomic analyses. The combination of L. rhamnosus with collagen peptide altered acid production by S. mutans, significantly increasing culture pH at an early stage of biofilm formation. Moreover, the 3D architecture of the S. mutans biofilm was greatly compromised when it was in the presence of L. rhamnosus with collagen peptide, resulting in a significant reduction in exopolysaccharide with unstructured and mixed bacterial organization. The presence of L. rhamnosus with collagen peptide modulated the virulence potential of S. mutans via down-regulation of eno, ldh, and atpD corresponding to acid production and proton transportation, whereas aguD associated with alkali production was up-regulated. Gly-Pro-Hyp, a common tripeptide unit of collagen, consistently modulated the cariogenic potential of S. mutans by inhibiting acid production, similar to the bioactivity of a collagen peptide. It also enhanced the relative abundance of commensal streptococci (S. oralis) in a mixed-species biofilm by inhibiting S. mutans colonization and dome-like microcolony formation. This work demonstrates that food-derived synbiotics may offer a useful means of disrupting cariogenic communities and maintaining microbial homeostasis.


Asunto(s)
Proteínas Bacterianas/genética , Biopelículas/efectos de los fármacos , Colágeno/química , Lacticaseibacillus rhamnosus/fisiología , Péptidos/farmacología , Streptococcus mutans/fisiología , Ácidos/metabolismo , Terapia Combinada , Medios de Cultivo/química , Caries Dental/microbiología , Caries Dental/prevención & control , Perfilación de la Expresión Génica , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Humanos , Concentración de Iones de Hidrógeno , Microscopía Confocal , Polisacáridos Bacterianos/metabolismo , Probióticos , Streptococcus mutans/efectos de los fármacos , Streptococcus mutans/metabolismo
13.
BMC Microbiol ; 21(1): 249, 2021 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-34536996

RESUMEN

BACKGROUND: Probiotics are widely used in intestinal microbiota imbalance caused by sepsis, however, the protective mechanism is still unclear. This study aimed to explore protective effect of Lacticaseibacillus rhamnosus TR08 on intestinal injury in septic mice. RESULTS: The levels of serum inflammatory factors were reduced significantly in septic mice treated with L. rhamnosus TR08. The levels of sIgA in terminal ileum were significantly higher in probiotic treatment group than sepsis group. Intestinal pathological damage in septic mice improved and the expression of tight junction proteins increased after probiotic treatment. Sequencing of fecal microbiota showed that the abundance and diversity of probiotic treatment group were significantly better than those of sepsis group, and beneficial bacteria increased while some bacteria decreased in the phylum level. CONCLUSION: L. rhamnosus TR08 could improve the integrity of intestinal barrier, enhance the intestinal mucosal immunity in septic mice, and rebalance the intestinal microecosystem.


Asunto(s)
Disbiosis/prevención & control , Enfermedades Intestinales/prevención & control , Lacticaseibacillus rhamnosus/fisiología , Probióticos/uso terapéutico , Sepsis/complicaciones , Animales , Bacterias/clasificación , Bacterias/genética , Disbiosis/microbiología , Microbioma Gastrointestinal/genética , Microbioma Gastrointestinal/inmunología , Inmunoglobulina A/análisis , Inmunoglobulina A/inmunología , Inflamación/sangre , Inflamación/prevención & control , Enfermedades Intestinales/etiología , Enfermedades Intestinales/microbiología , Intestinos/inmunología , Intestinos/patología , Masculino , Ratones , Probióticos/administración & dosificación , Sepsis/terapia
14.
BMC Microbiol ; 21(1): 146, 2021 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-33985438

RESUMEN

BACKGROUND: Gut microbiota closely communicate in the immune system to maintain a balanced immune homeostasis in the gastrointestinal tract of the host. Oral administration of probiotics modulates gut microbiota composition. In the present study, we isolated Lactobacillus rhamnosus HDB1258, which induced tumor necrosis factor (TNF)-α and interleukin (IL)-10 expression in macrophages, from the feces of breastfeeding infants and examined how HDB1258 could regulate the homeostatic immune response in mice with or without lipopolysaccharide (LPS)-induced systemic inflammation. RESULTS: Oral administration of HDB1258 significantly increased splenic NK cell cytotoxicity, peritoneal macrophage phagocytosis, splenic and colonic TNF-α expression, TNF-α to IL-10 expression ratio, and fecal IgA level in control mice, while Th1 and Treg cell differentiation was not affected in the spleen. However, HDB1258 treatment significantly suppressed peritoneal macrophage phagocytosis and blood prostaglandin E2 level in mice with LPS-induced systemic inflammation. Its treatment increased LPS-suppressed ratios of Treg to Th1 cell population, Foxp3 to T-bet expression, and IL-10 to TNF-α expression. Oral administration of HDB1258 significantly decreased LPS-induced colon shortening, myeloperoxidase activity and NF-κB+/CD11c+ cell population in the colon, while the ratio of IL-10 to TNF-α expression increased. Moreover, HDB1258 treatment shifted gut microbiota composition in mice with and without LPS-induced systemic inflammation: it increased the Cyanobacteria and PAC000664_g (belonging to Bacteroidetes) populations and reduced Deferribacteres and EU622763_s group (belonging to Bacteroidetes) populations. In particular, PAC001066_g and PAC001072_s populations were negatively correlated with the ratio of IL-10 to TNF-α expression in the colon, while the PAC001070_s group population was positively correlated. CONCLUSIONS: Oral administered HDB1258 may enhance the immune response by activating innate immunity including to macrophage phagocytosis and NK cell cytotoxicity in the healthy host and suppress systemic inflammation in the host with inflammation by the modulation of gut microbiota and IL-10 to TNF-α expression ratio in immune cells.


Asunto(s)
Microbioma Gastrointestinal/efectos de los fármacos , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Lacticaseibacillus rhamnosus/fisiología , Lipopolisacáridos/efectos adversos , Probióticos/administración & dosificación , Animales , Bacterias/clasificación , Bacterias/genética , Bacterias/inmunología , Bacterias/aislamiento & purificación , Humanos , Inmunidad , Inflamación/inducido químicamente , Inflamación/microbiología , Interleucina-10/genética , Interleucina-10/inmunología , Lacticaseibacillus rhamnosus/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Linfocitos T Reguladores/inmunología , Células TH1/inmunología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
15.
Arch Microbiol ; 203(3): 1221-1229, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33620523

RESUMEN

Salicylic acid, widely distributed in the whole plant kingdom, is a benzoic acid derivative acting as a signal substance in plants, but could be related to differences in cancer incidence, as many herbs and spices contain high amounts. Lactobacillus rhamnosus GG (LGG) is one of the best-known lactic acid bacteria that has been studied for over 30 years. Probiotic and/or commensal bacteria of the human microbiota are known to respond to diet constituents. Therefore, the present study aims at investigating the possible effects of salicylic acid on the probiotic properties of LGG, and in vitro cytotoxic effects of combination of salicylic acid and LGG on human colon and prostate cancer cells. Salicylic acid significantly (p < 0.05) increased co-aggregation of LGG with E. coli (~ twofold) and anti-oxidant properties. Furthermore, it also induced the cytotoxic effects of LGG against human colon cancer cells. These results suggest that interaction of LGG with salicylic acid can exert more probiotic properties.


Asunto(s)
Lacticaseibacillus rhamnosus/fisiología , Ácido Salicílico/farmacología , Simbióticos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Colon/citología , Colon/microbiología , Escherichia coli/efectos de los fármacos , Escherichia coli/fisiología , Humanos , Masculino , Microbiota/fisiología , Probióticos/farmacología , Neoplasias de la Próstata/microbiología
16.
Helicobacter ; 26(3): e12806, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33843101

RESUMEN

BACKGROUND: Eradication of Helicobacter pylori infection is the most direct and effective way for preventing gastric cancer. Lactic acid bacteria are considered as alternative therapeutic agents against H. pylori infection. METHODS: Effects of Lactobacillus rhamnosus JB3 (LR-JB3) on the virulence gene expression of H. pylori and infection-induced cellular responses of AGS cells were investigated by co-cultivating infected AGS cells with different multiplicity of infections (MOIs) of LR-JB3. RESULTS: LR-JB3, specifically at a MOI of 25, suppressed the association ability of H. pylori and its induced IL-8 levels, as well as the mRNA levels of vacA, sabA, and fucT of H. pylori, infection-induced Lewis (Le)x antigen and Toll-like receptor 4 (TLR4) expressions in AGS cells. However, the apoptosis mediated by infection was inhibited by LR-JB3 in a dose-dependent manner. In addition, autoinducer (AI)-2 was observed to have increased the association ability and fucT expression of H. pylori, and Lex antigen and TLR4 expression of AGS cells. Interestingly, an unknown bioactive cue was hypothesized to have been secreted from LR-JB3 at a MOI of 25 to act as an antagonist of AI-2. CONCLUSIONS: LR-JB3 possesses various means to interfere with H. pylori pathogenesis and infection-induced cellular responses of AGS cells to fight against infection.


Asunto(s)
Antibiosis , Helicobacter pylori , Lacticaseibacillus rhamnosus , Apoptosis , Línea Celular Tumoral , Células Epiteliales , Mucosa Gástrica , Infecciones por Helicobacter , Helicobacter pylori/patogenicidad , Humanos , Lacticaseibacillus rhamnosus/fisiología
17.
J Pediatr Hematol Oncol ; 43(4): e457-e461, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33031162

RESUMEN

INTRODUCTION: During the administration of antineoplastic drugs, acute complications because of toxicity occur, determining their hospital readmission, visits to the emergency department, use of antimicrobials, and possibilities of presenting systemic infections, impacting on their life quality. METHODS: Through a prospective cohort, 60 children with acute lymphoblastic leukemia were followed-up for 30 days after the hospital discharge because of chemotherapy administration, those patients were previously included in a single-blinded study in which 30 (group 1) received Lactobacillus rhamnosus GG probiotic during the administration of chemotherapy. The remaining 30 patients did not receive probiotics (group 2). There were evaluated gastrointestinal symptoms, such as diarrhea, dyspepsia, abdominal distension, meteorism, constipation, nausea, and vomit, development of infections, antibiotic use, number of emergency department visits, number of hospitalizations, and sepsis diagnosis. STATISTICAL ANALYSIS: To assess the impact of the use of probiotics, the difference in proportions between both study groups was evaluated. RESULTS: Gastrointestinal manifestations (nausea, vomiting, diarrhea, constipation) occurred in 30% of patients in group 1 versus 63% of group 2 (P=0.009). Nine of 30 patients (30.0%) in group 1 went to the emergency room, versus 33.3% of group 2 (P=0.7). Antimicrobials were used in 8 subjects (26.6%) in group 1 versus 6 subjects (53.3%) in group 2 (P=0.03) suspected of an infectious disease. Four (13.3%) group 1 patients were hospitalized versus 30% of group 2 (P=0.1). Two subjects (6.6%) in group 1 had sepsis versus 7 (23.3%) in group 2 (P=0.07).Conclusions:The results indicate that the use of probiotics can be a great alternative in the improvement of gastrointestinal symptoms and the adverse effects associated with chemotherapy.


Asunto(s)
Antineoplásicos/efectos adversos , Enfermedades Gastrointestinales/inducido químicamente , Enfermedades Gastrointestinales/prevención & control , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Probióticos/uso terapéutico , Antineoplásicos/uso terapéutico , Niño , Diarrea/inducido químicamente , Diarrea/prevención & control , Femenino , Humanos , Lacticaseibacillus rhamnosus/fisiología , Masculino , Náusea/inducido químicamente , Náusea/prevención & control , Estudios Prospectivos , Vómitos/inducido químicamente , Vómitos/prevención & control
18.
J Korean Med Sci ; 36(8): e58, 2021 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-33650336

RESUMEN

BACKGROUND: Human breast milk (HBM) contains optimal nutrients for infant growth. Probiotics are used to prevent disease and, when taken by the mother, they may affect infant microbiome as well as HBM. However, few studies have specifically investigated the effect of probiotic intake by the mother on HBM and infant microbiota at genus/species level. Therefore, we present a comprehensive analysis of paired HBM and infant feces (IF) microbiome samples before and after probiotic intake by HBM-producing mothers. METHODS: Lactating mothers were administered with Lactobacillus rhamnosus (n = 9) or Saccharomyces boulardii capsules (n = 9), for 2 months; or no probiotic (n = 7). Paired HBM and IF samples were collected before and after treatment and analyzed by next-generation sequencing. RESULTS: Forty-three HBM and 49 IF samples were collected and sequenced. Overall, in 43 HBM samples, 1,190 microbial species belonging to 684 genera, 245 families, 117 orders, and 56 classes were detected. In 49 IF samples, 372 microbial species belonging to 195 genera, 79 families, 42 orders, and 18 classes were identified. Eight of 20 most abundant genera in both HBM and IF samples overlapped: Streptococcus (14.42%), Lactobacillus, Staphylococcus, and Veillonella, which were highly abundant in the HBM samples; and Bifidobacterium (27.397%), Bacteroides, and Faecalibacterium, which were highly abundant in the IF samples. Several major bacterial genera and species were detected in the HBM and IF samples after probiotic treatment, illustrating complex changes in the microbiomes upon treatment. CONCLUSION: This is the first Korean microbiome study in which the effect of different probiotic intake by the mother on the microbiota in HBM and IF samples was investigated. This study provides a cornerstone to further the understanding of the effect of probiotics on the mother and infant microbiomes.


Asunto(s)
Heces/microbiología , Microbioma Gastrointestinal , Leche Humana/microbiología , Probióticos/administración & dosificación , Adulto , Bacterias/genética , Bacterias/aislamiento & purificación , Lactancia Materna , Análisis por Conglomerados , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Lactante , Lacticaseibacillus rhamnosus/fisiología , Persona de Mediana Edad , Madres , ARN Ribosómico 16S/química , ARN Ribosómico 16S/genética , ARN Ribosómico 16S/metabolismo , Saccharomyces boulardii/fisiología , Análisis de Secuencia de ADN , Adulto Joven
19.
Molecules ; 26(16)2021 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-34443691

RESUMEN

BACKGROUND: Milk is considered an important source of bioactive peptides, which can be produced by endogenous or starter bacteria, such as lactic acid bacteria, that are considered effective and safe producers of food-grade bioactive peptides. Among the various types of milk, donkey milk has been gaining more and more attention for its nutraceutical properties. METHODS: Lactobacillus rhamnosus 17D10 and Lactococcus lactis subsp. cremoris 40FEL3 were selected for their ability to produce peptides from donkey milk. The endogenous peptides and those obtained after bacterial fermentation were assayed for their antioxidant, antibacterial, and antiviral activities. The peptide mixtures were characterized by means of LC-MS/MS and then analyzed in silico using the Milk Bioactive Peptide DataBase. RESULTS: The peptides produced by the two selected bacteria enhanced the antioxidant activity and reduced E. coli growth. Only the peptides produced by L. rhamnosus 17D10 were able to reduce S. aureus growth. All the peptide mixtures were able to inhibit the replication of HSV-1 by more than 50%. Seventeen peptides were found to have 60% sequence similarity with already known bioactive peptides. CONCLUSIONS: A lactic acid bacterium fermentation process is able to enhance the value of donkey milk through bioactivities that are important for human health.


Asunto(s)
Antibacterianos/farmacología , Antivirales/farmacología , Fermentación , Lacticaseibacillus rhamnosus/fisiología , Lactococcus/fisiología , Leche/microbiología , Secuencia de Aminoácidos , Animales , Antioxidantes/farmacología , Quelantes/farmacología , Equidae , Proteínas de la Leche/análisis , Péptidos/química , Péptidos/farmacología
20.
J Nutr ; 150(5): 1313-1323, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32027752

RESUMEN

BACKGROUND: Probiotic Lactobacillius rhamnosus GG (LGG) shows beneficial immunomodulation on cultured cell lines in vitro and in mouse models. OBJECTIVE: The aim was to investigate the effects of LGG on intestinal injury and the underlying mechanisms by elucidating inflammatory signaling pathways and metabolomic response to LPS stimulation in the piglet intestine. METHODS: Piglets (Duroc × Landrace × Large White, including males and female; 8.6 ± 1.1 kg) aged 28 d were assigned to 3 groups (n = 6/group): oral inoculation with PBS for 2 wk before intraperitoneal injection of physiological saline [control (CON)] or LPS (25 µg/kg body weight; LPS) or oral inoculation with LGG for 2 wk before intraperitoneal injection of LPS (LGG+LPS). Piglets were killed 4 h after LPS injection. Systemic inflammation, intestinal integrity, inflammation signals, and metabolomic characteristics in the intestine were determined. RESULTS: Compared with CON, LPS stimulation significantly decreased ileal zonula occludens 1 (ZO-1; 44%), claudin-3 (44%), and occludin (41%) expression; increased serum diamineoxidase (73%), D-xylose (19%), TNF-α (43%), and IL-6 (55%) concentrations; induced p38 mitogen-activated protein kinase (p38 MAPK; 85%), extracellular signal-regulated kinase (ERK; 96%), and NF-κB p65 phosphorylation (37%) (P < 0.05). Compared with LPS stimulation alone, LGG pretreatment significantly enhanced the intestinal barrier by upregulating expressions of tight junction proteins (ZO-1, 73%; claudin-3, 55%; occludin, 67%), thereby decreasing serum diamineoxidase (26%) and D-xylose (28%) concentrations, and also reduced serum TNF-α expression (16%) and ileal p38 MAPK (79%), ERK (43%) and NF-κB p65 (37%) phosphorylation levels (P < 0.05). Metabolomic analysis showed clear separation between each group. The concentrations of caprylic acid [fold-change (FC) = 2.39], 1-mono-olein (FC = 2.68), erythritol (FC = 4.62), and ethanolamine (FC = 4.47) significantly increased in the intestine of LGG + LPS piglets compared with the LPS group (P < 0.05). CONCLUSIONS: These data suggest that LGG alleviates gut inflammation, improves intestinal barrier function, and modulates the metabolite profile of piglets challenged with LPS. This trial was registered at the Zhejiang University (http://www.lac.zju.edu.cn) as ZJU20170529.


Asunto(s)
Gastroenteritis/prevención & control , Enfermedades Gastrointestinales/prevención & control , Lacticaseibacillus rhamnosus/fisiología , Lipopolisacáridos/farmacología , Metaboloma/fisiología , Sus scrofa , Animales , Femenino , Gastroenteritis/inducido químicamente , Enfermedades Gastrointestinales/inducido químicamente , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Probióticos/administración & dosificación , Transducción de Señal/fisiología , Proteínas de Uniones Estrechas/genética , Factor de Transcripción ReIA/metabolismo , Regulación hacia Arriba/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA