Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 152
Filtrar
1.
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
2.
Sci Rep ; 14(1): 14426, 2024 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-38910172

RESUMEN

Vaginitis, a prevalent gynecological condition in women, is mainly caused by an imbalance in the vaginal micro-ecology. The two most common types of vaginitis are vaginal bacteriosis and vulvovaginal candidiasis, triggered by the virulent Gardnerella vaginalis and Candida albicans, respectively. In this study, a strain capable of inhibiting G. vaginalis and C. albicans was screened from vaginal secretions and identified as Lactobacillus gasseri based on 16S rRNA sequences. The strain, named L. gasseri VHProbi E09, could inhibit the growth of G. vaginalis and C. albicans under co-culture conditions by 99.07% ± 0.26% and 99.95% ± 0.01%, respectively. In addition, it could significantly inhibit the adhesion of these pathogens to vaginal epithelial cells. The strain further showed the ability to inhibit the enteropathogenic bacteria Escherichia coli and Salmonella enteritidis, to tolerate artificial gastric and intestinal fluids and to adhere to intestinal Caco-2 cells. These results suggest that L. gasseri VHProbi E09 holds promise for clinical trials and animal studies whether administered orally or directly into the vagina. Whole-genome analysis also revealed a genome consisting of 1752 genes for L. gasseri VHProbi E09, with subsequent analyses identifying seven genes related to adhesion and three genes related to bacteriocins. These adhesion- and bacteriocin-related genes provide a theoretical basis for understanding the mechanism of bacterial inhibition of the strain. The research conducted in this study suggests that L. gasseri VHProbi E09 may be considered as a potential probiotic, and further research can delve deeper into its efficacy as an agent which can restore a healthy vaginal ecosystem.


Asunto(s)
Candida albicans , Gardnerella vaginalis , Lactobacillus gasseri , Probióticos , Vagina , Femenino , Humanos , Lactobacillus gasseri/genética , Células CACO-2 , Gardnerella vaginalis/genética , Vagina/microbiología , Adhesión Bacteriana , Vaginitis/microbiología , ARN Ribosómico 16S/genética
3.
Nutrients ; 16(8)2024 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-38674829

RESUMEN

The intestinal epithelial barrier can prevent the invasion of pathogenic microorganisms and food antigens to maintain a consistent intestinal homeostasis. However, an imbalance in this barrier can result in various diseases, such as inflammatory bowel disease, malnutrition, and metabolic disease. Thus, the aim of this study was to select probiotic strains with epithelial barrier-enhancing ability in cell-based model and further investigate them for their improving effects on colitis mouse and weaned piglet models. The results showed that selected specific cell-free fermentation supernatants (CFSs) from Ligilactobacillus salivarius P1, Lactobacillus gasseri P12, and Limosilactobacillus reuteri G7 promoted intestinal epithelial cell growth and proliferation, strengthening the intestinal barrier in an intestinal epithelial cell line Caco-2 model. Further, the administration of CFSs of L. salivarius P1, L. gasseri P12, and L. reuteri G7 were found to ameliorate DSS-induced colitis in mice. Additionally, spray-dried powders of CFS from the three strains were examined in a weaned piglet model, only CFS powder of L. reuteri G7 could ameliorate the feed/gain ratio and serum levels of D-lactate and endotoxin. In conclusion, a new potential probiotic strain, L. reuteri G7, was selected and showed ameliorating effects in both colitis mouse and weaned piglet models.


Asunto(s)
Colitis , Modelos Animales de Enfermedad , Fermentación , Mucosa Intestinal , Limosilactobacillus reuteri , Probióticos , Destete , Animales , Probióticos/farmacología , Colitis/inducido químicamente , Colitis/metabolismo , Colitis/microbiología , Humanos , Ratones , Porcinos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Células CACO-2 , Ligilactobacillus salivarius , Lactobacillus gasseri , Sulfato de Dextran , Masculino , Proliferación Celular/efectos de los fármacos
4.
Benef Microbes ; 15(3): 293-310, 2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38677716

RESUMEN

Oral administration of probiotics has been proposed as a promising biotherapy to prevent and treat different diseases related to gastrointestinal disorders, such as irritable bowel syndrome (IBS). Due to the increasing research area on the characterisation of new probiotic bacterial strains, it is necessary to perform suitable in vitro experiments, using pertinent cellular models, in order to establish appropriate readout profiles based on IBS symptoms and subtypes. In this work, a collection of 30 candidate strains, belonging mainly to the Lactobacillus and Bifidobacterium genera, were screened using three different sets of in vitro experiments with different readouts to identify promising probiotic strains with: (1) the ability to inhibit the synthesis of IL-8 production by TNF-α stimulated HT-29 cells, (2) immunomodulatory properties quantified as increased IL-10 levels in peripheral blood mononuclear cell (PBMCs), and (3) the ability to maintain epithelial barrier integrity by increasing the trans-epithelial/endothelial electrical resistance (TEER) values in Caco-2 cells. Based on these criteria, three strains were selected: Lactobacillus gasseri PI41, Lacticaseibacillus rhamnosus PI48 and Bifidobacterium animalis subsp. lactis PI50, and tested in a murine model of low-grade inflammation induced by dinitrobenzene sulfonic acid (DNBS), which mimics some of the symptoms of IBS. Among the three strains, L. gasseri PI41 improved overall host well-being by preventing body weight loss in DNBS-treated mice and restored gut homeostasis by normalising the intestinal permeability and reducing pro-inflammatory markers. Therefore, the potential of this strain was confirmed in a second murine model known to reproduce IBS symptoms: the neonatal maternal separation (NMS) model. The PI41 strain was effective in preventing intestinal permeability and reducing colonic hypersensitivity. In conclusion, the set of in vitro experiments combined with in vivo assessments allowed us to identify a promising probiotic candidate strain, L. gasseri PI41, in the context of IBS.


Asunto(s)
Síndrome del Colon Irritable , Probióticos , Probióticos/administración & dosificación , Probióticos/farmacología , Síndrome del Colon Irritable/terapia , Síndrome del Colon Irritable/microbiología , Humanos , Animales , Ratones , Células CACO-2 , Células HT29 , Modelos Animales de Enfermedad , Leucocitos Mononucleares/inmunología , Lactobacillus/fisiología , Interleucina-8/metabolismo , Bifidobacterium/fisiología , Interleucina-10 , Lactobacillus gasseri , Lacticaseibacillus rhamnosus/fisiología , Masculino , Bifidobacterium animalis/fisiología
5.
J Food Sci ; 89(5): 3078-3093, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38605580

RESUMEN

Human milk contains a variety of microorganisms that exert benefit for human health. In the current study, we isolated a novel Lactobacillus gasseri strain named Lactobacillus gasseri (L. gasseri) SHMB 0001 from human milk and aimed to evaluate the probiotic characteristics and protective effects on murine colitis of the strain. The results showed that L. gasseri SHMB 0001 possessed promising potential probiotic characteristics, including good tolerance against artificial gastric and intestinal fluids, adhesion to Caco-2 cells, susceptibility to antibiotic, no hemolytic activity, and without signs of toxicity or infection in mice. Administration of L. gasseri SHMB 0001 (1 × 108 CFU per gram of mouse weight per day) reduced weight loss, the disease activity index, and colon shortening in mice during murine colitis conditions. Histopathological analysis revealed that L. gasseri SHMB 0001 treatment attenuated epithelial damage and inflammatory infiltration in the colon. L. gasseri SHMB 0001 treatment increased the expression of colonic occludin and claudin-1 while decreasing the expression of pro-inflammatory cytokine genes. L. gasseri SHMB 0001 modified the composition and structure of the gut microbiota community and partially recovered the Clusters of Orthologous Groups (COG) and Kyoto Encyclopedia of Genes and Genomes (KEGG) metabolic pathways altered by dextran sulfate sodium (DSS). Overall, our results indicated that the human breast milk-derived L. gasseri SHMB 0001 exhibited promising probiotic properties and ameliorative effect on DSS-induced colitis in mice. L. gasseri SHMB 0001 may be applied as a promising probiotic against intestinal inflammation in the future. PRACTICAL APPLICATION: L. gasseri SHMB 0001 isolated from human breast milk showed good tolerance to gastrointestinal environment, safety, and protective effect against DSS-induced mice colitis via enforcing gut barrier, downregulating pro-inflammatory cytokines, and modulating gut microbiota. L. gasseri SHMB 0001 may be a promising probiotic candidate for the treatment of intestinal inflammation.


Asunto(s)
Colitis , Sulfato de Dextran , Microbioma Gastrointestinal , Lactobacillus gasseri , Leche Humana , Probióticos , Probióticos/farmacología , Animales , Humanos , Ratones , Colitis/inducido químicamente , Colitis/terapia , Colitis/microbiología , Sulfato de Dextran/efectos adversos , Microbioma Gastrointestinal/efectos de los fármacos , Células CACO-2 , Femenino , Colon/microbiología , Colon/patología , Colon/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad
6.
J Med Food ; 27(5): 385-395, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38574296

RESUMEN

This study aimed to investigate the effects and mechanism of Lactobacillus gasseri BNR17, a probiotic strain isolated from human breast milk, on dexamethasone-induced muscle loss in mice and cultured myotubes. BALB/c mice were intraperitoneally injected with dexamethasone, and orally administered L. gasseri BNR17 for 21 days. L. gasseri BNR17 treatment ameliorated dexamethasone-induced decline in muscle function, as evidenced by an increase in forelimb grip strength, treadmill running time, and rotarod retention time in both female and male mice. In addition, L. gasseri BNR17 treatment significantly increased the mass of the gastrocnemius and quadriceps muscles. Dual-energy X-ray absorptiometry showed a significant increase in lean body mass and a decrease in fat mass in both whole body and hind limb after treatment with L. gasseri BNR17. It was found that L. gasseri BNR17 treatment downregulated serum myostatin level and the protein degradation pathway composed of muscle-specific ubiquitin E3 ligases, MuRF1 and MAFbx, and their transcription factor FoxO3. In contrast, L. gasseri BNR17 treatment upregulated serum insulin-like growth factor-1 level and Akt-mTOR-p70S6K signaling pathway involved in protein synthesis in muscle. As a result, L. gasseri BNR17 treatment significantly increased the levels of major muscular proteins such as myosin heavy chain and myoblast determination protein 1. Consistent with in vivo results, L. gasseri BNR17 culture supernatant significantly ameliorated dexamethasone-induced C2C12 myotube atrophy in vitro. In conclusion, L. gasseri BNR17 ameliorates muscle loss by downregulating the protein degradation pathway and upregulating the protein synthesis pathway.


Asunto(s)
Dexametasona , Lactobacillus gasseri , Ratones Endogámicos BALB C , Fibras Musculares Esqueléticas , Proteínas Musculares , Músculo Esquelético , Atrofia Muscular , Probióticos , Ubiquitina-Proteína Ligasas , Animales , Dexametasona/efectos adversos , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/efectos de los fármacos , Ratones , Femenino , Masculino , Proteínas Musculares/metabolismo , Atrofia Muscular/inducido químicamente , Atrofia Muscular/metabolismo , Atrofia Muscular/tratamiento farmacológico , Lactobacillus gasseri/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/efectos de los fármacos , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina-Proteína Ligasas/genética , Proteínas Ligasas SKP Cullina F-box/metabolismo , Proteínas Ligasas SKP Cullina F-box/genética , Proteínas de Motivos Tripartitos/metabolismo , Proteínas de Motivos Tripartitos/genética , Proteína Forkhead Box O3/metabolismo , Proteína Forkhead Box O3/genética , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
7.
Sci Rep ; 14(1): 5711, 2024 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-38459201

RESUMEN

This study aimed to investigate the biological activities of Lactobacillus gasseri SM 05 (L. gasseri) and Lacticaseibacillus casei subsp. casei PTCC 1608 (L. casei) in the black raspberry (Rubus dolichocarpus) juice (BRJ) environment, and also the anti-adhesion activity against Salmonella typhimurium (S. typhimurium) in fermented black raspberry juice (FBRJ). Results showed significant anti-adhesion activity in Caco-2 epithelial cells. In the anti-adhesion process, lactic acid bacteria (LAB) improve intestinal health by preventing the adhesion of pathogens. Adding LAB to BRJ produces metabolites with bacteriocin properties. Major findings of this research include improved intestinal health, improved antidiabetic properties, inhibition of degradation of amino acids, and increase in the nutritional value of foods that have been subjected to heat processing by preventing Maillard inhibition, and inhibition of oxidation of foodstuff by increased antioxidant activity of BRJ. Both species of Lactobacillus effectively controlled the growth of S. typhimurium during BRJ fermentation. Moreover, in all tests, as well as Maillard's and α-amylase inhibition, L. gasseri was more effective than L. casei. The phenolic and flavonoid compounds increased significantly after fermentation by both LAB (p < 0.05). Adding Stevia extract to FBRJ and performing the HHP process showed convenient protection of phenolic compounds compared to heat processing.


Asunto(s)
Lacticaseibacillus casei , Lactobacillus gasseri , Probióticos , Rubus , Stevia , Humanos , Fermentación , Células CACO-2 , Extractos Vegetales/farmacología
8.
ACS Synth Biol ; 13(3): 951-957, 2024 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-38335132

RESUMEN

Lactic acid bacteria (LAB) are important for many biotechnological applications such as bioproduction and engineered probiotics for therapy. Inducible promoters are key gene expression control elements, yet those available in LAB are mainly based on bacteriocin systems and have many drawbacks, including large gene clusters, costly inducer peptides, and little portability to in vivo settings. Using Lactobacillus gasseri, a model commensal bacteria from the human gut, we report the engineering of synthetic LactoSpanks promoters (Pls), a collection of variable strength inducible promoters controlled by the LacI repressor from E. coli and induced by isopropyl ß-d-1-thiogalactopyranoside (IPTG). We first show that the Phyper-spank promoter from Bacillus subtilis is functional in L. gasseri, albeit with substantial leakage. We then construct and screen a semirational library of Phyper-spank variants to select a set of four IPTG-inducible promoters that span a range of expression levels and exhibit reduced leakages and operational dynamic ranges (from ca. 9 to 28 fold-change). With their low genetic footprint and simplicity of use, LactoSpanks will support many applications in L. gasseri, and potentially other lactic acid and Gram-positive bacteria.


Asunto(s)
Lactobacillales , Lactobacillus gasseri , Humanos , Lactobacillus gasseri/genética , Isopropil Tiogalactósido/farmacología , Lactobacillales/genética , Escherichia coli/metabolismo , Regiones Promotoras Genéticas/genética
9.
Microb Pathog ; 188: 106559, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38272328

RESUMEN

Helicobacter pylori has been recognized as a true pathogen, which is associated with various gastroduodenal diseases, and gastric adenocarcinoma. The crosstalk between H. pylori virulence factors and host autophagy remains challenging. H. pylori can produce extracellular vesicles (EVs) that contribute to gastric inflammation and malignancy. Some probiotic strains have been documented to modulate cell autophagy process. This study was aimed to investigate the modulatory effect of cell-free supernatant (CFS) obtained from Lactobacillus gasseri ATCC 33323 on autophagy induced by H. pylori-derived EVs. EVs were isolated from two clinical H. pylori strains (BY-1 and OC824), and characterized using transmission electron microscopy (TEM) and dynamic light scattering (DLS). The viability of AGS cells was assessed after exposure to different concentrations of H. pylori EVs, and L. gasseri CFS. Based on MTT assay and Annexin V-FITC/PI staining, 50 µg/ml of H. pylori EVs and 10 % v/v of L. gasseri CFS were used for further cell treatment experiments. Autophagy was examined using acridin orange (AO) staining, RT-qPCR analysis for autophagy mediators (LC3B, ATG5, ATG12, ATG16L1, BECN1, MTOR, and NOD1), and western blotting for LC3B expression. H. pylori EVs were detected to range in size from 50 to 200 nm. EVs of both H. pylori strains and L. gasseri CFS showed no significant effect on cell viability as compared to untreated cells. H. pylori EVs promoted the development of acidic vesicular organelles and the expression of autophagy-related genes (LC3B, ATG5, ATG12, ATG16L1, BECN1, and NOD1), and decreased the expression of MTOR in AGS cells at 12 and 24 h time periods. In addition, the production of LC3B was increased following 12 h of treatment in AGS cells. In contrast, L. gasseri CFS effectively inhibited EVs-induced autophagy, as evidenced by reduced acidic vesicular organelle formation and modulation of autophagy markers. Our study indicated that L. gasseri CFS can effectively suppress H. pylori EV-induced autophagy in AGS cells. Further investigations are required to decipher the mechanism of action L. gasseri CFS and its metabolites on autophagy inhibition induced by H. pylori.


Asunto(s)
Vesículas Extracelulares , Infecciones por Helicobacter , Helicobacter pylori , Lactobacillus gasseri , Humanos , Helicobacter pylori/genética , Células Epiteliales , Autofagia , Serina-Treonina Quinasas TOR , Infecciones por Helicobacter/terapia
10.
J Dairy Sci ; 107(4): 1857-1876, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37923200

RESUMEN

Constipation is directly related to the intestinal microenvironment, in which the promotion of gastrointestinal (GI) motility and improvement of gut microbiota distribution are important for alleviating symptoms. Herein, after the intervention of probiotic fermented milk (FMMIX) containing Lacticaseibacillus paracasei JY062 and Lactobacillus gasseri JM1 for 14 d in Kunming mice with loperamide-induced constipation, the results indicated that FMMIX significantly increased the secretion of serum motilin, gastrin and 5-hydroxytryptamine, as well as decreased the secretion of peptide YY, vasoactive intestinal peptide, and nitric oxide in mice. As determined by immunohistochemical analysis, FMMIX promoted an augmentation in the quantity of Cajal interstitial cells. In addition, the mRNA and protein expression of c-kit and stem cell factor (SCF) were upregulated to facilitate intestinal motility. High-throughput sequencing and gas chromatography techniques revealed that FMMIX led to an increase in the relative abundance of beneficial bacteria (Lactobacillus, Oscillospira, Ruminococcus, Coprococcus, and Akkermansia), reduced the presence of harmful bacteria (Prevotella), and resulted in elevated levels of short-chain fatty acids (SCFA) with a superior improvement compared with unfermented milk. Untargeted metabolomics revealed significant upregulation of functional metabolites such as l-pipecolinic acid, dl-phenylalanine, and naringenin in FMMIX, presumably playing a potential role in constipation relief. Overall, our results showed that FMMIX had the potential to alleviate constipation symptoms in mice by improving the secretion of serum GI regulatory peptides and neurotransmitters, increasing the expression of c-kit and SCF proteins, and modulating the gut microbiota structure and SCFA levels, and may be associated with an increase in these functional metabolites. This suggested that FMMIX could be a promising adjunctive strategy for managing constipation symptoms and could contribute to the development of functional foods aimed at improving gut health.


Asunto(s)
Microbioma Gastrointestinal , Lacticaseibacillus paracasei , Lactobacillus gasseri , Probióticos , Ratones , Animales , Leche , Estreñimiento/inducido químicamente , Estreñimiento/terapia , Estreñimiento/veterinaria , Motilidad Gastrointestinal , Probióticos/uso terapéutico , Probióticos/farmacología
11.
Nutrients ; 15(20)2023 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-37892560

RESUMEN

Menopause is a significant phase in a woman's life. Menopausal symptoms can affect overall well-being and quality of life. Conventionally, hormone replacement therapy (HRT) is used to alleviate menopausal symptoms; however, depending on the conditions, HRT may lead to side effects, necessitating the exploration of alternative therapies with fewer side effects. In this study, we investigated the effects of a combination of soybean germ extract (S30) containing 30% (w/w) isoflavone and a probiotic, Lactobacillus gasseri (LGA1), on menopausal conditions in an ovariectomized (OVX) rat model. We evaluated the impact of S30+LGA on body weight, estrogen markers, uterine and bone health, vascular markers, and neurotransmitter levels. The results revealed that treatment with S30+LGA1 significantly improved body weight and uterine and bone health. Moreover, S30+LGA1 demonstrated promising effects on lipid profile, liver function, and vascular markers and positively impacted serotonin and norepinephrine levels, indicating potential mood-enhancing effects. In conclusion, S30+LGA1, possessing anti-menopausal effects in vitro and in vivo, can be recommended as a soy-based diet, which offers various health benefits, especially for menopausal women.


Asunto(s)
Glycine max , Lactobacillus gasseri , Ratas , Animales , Femenino , Humanos , Calidad de Vida , Menopausia , Extractos Vegetales/farmacología , Peso Corporal
12.
Food Funct ; 14(18): 8504-8520, 2023 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-37655696

RESUMEN

Type 2 diabetes mellitus (T2DM) is a chronic metabolic disorder characterized by dysregulation of lipid metabolism, insulin resistance, and gut microbiota disorder. Compared to drug interventions, probiotic interventions may have a more enduring effect without producing any side effects. Thus, the potential of probiotics as a therapeutic approach for diabetes and other metabolic disorders has gained increasing attention in recent years. In this study, we evaluated the therapeutic efficacy of Lactobacillus gasseri CKCC1913, a potential probiotic strain, in high-fat diet-induced insulin-resistant diabetes using the C57BL/6J mouse animal model. From the results, L. gasseri CKCC1913 has been shown to increase glucose tolerance, reduce fasting blood glucose levels in diabetic mice, and reduce the expression of pro-inflammatory cytokines, such as TNF-α and IL-6. Besides, L. gasseri CKCC1913 intervention effectively alleviated oxidative stress damage by increasing SOD activity, decreasing MDA levels, reducing insulin resistance, and improving dyslipidemia caused by diabetes. The potential mechanism of L. gasseri CKCC1913 in improving metabolic health and alleviating diabetes involves an increased abundance of beneficial bacteria, such as Parabacteroides merdae, which directly produce short-chain fatty acids that help regulate immune cells and reduce inflammation. SCFAs also enter the bloodstream and promote antioxidant enzyme activity in the liver, protecting against oxidative damage. Additionally, L. gasseri CKCC1913 influences local bacterial metabolism pathways, such as the superpathway of unsaturated fatty acid biosynthesis, leading to an increase in unsaturated fatty acids, increasing high-density lipoprotein cholesterol (HDL-C) levels and improving lipid metabolism and glucose control in diabetic mice. In summary, in this study, L. gasseri CKCC1913 and its potential impact on metabolic health highlight the promising potential of probiotics as a therapeutic approach for diabetes. Future research should focus on identifying the optimal dose and duration, investigating the long-term effects and mechanisms of action, and exploring the potential use of probiotics as an adjunct to other therapies or in preventing metabolic disorders.


Asunto(s)
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2 , Resistencia a la Insulina , Lactobacillus gasseri , Animales , Ratones , Ratones Endogámicos C57BL , Diabetes Mellitus Tipo 2/terapia , Diabetes Mellitus Experimental/terapia , Hígado , Antioxidantes
13.
Front Immunol ; 14: 1125239, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37575226

RESUMEN

Bacterial vaginosis (BV) is a common infection of the lower genital tract with a vaginal microbiome dysbiosis caused by decreasing of lactobacilli. Previous studies suggested that supplementation with live Lactobacillus may benefit the recovery of BV, however, the outcomes vary in people from different regions. Herein, we aim to evaluate the effectiveness of oral Chinese-origin Lactobacillus with adjuvant metronidazole (MET) on treating Chinese BV patients. In total, 67 Chinese women with BV were enrolled in this parallel controlled trial and randomly assigned to two study groups: a control group treated with MET vaginal suppositories for 7 days and a probiotic group treated with oral Lactobacillus gasseri TM13 and Lactobacillus crispatus LG55 as an adjuvant to MET for 30 days. By comparing the participants with Nugent Scores ≥ 7 and < 7 on days 14, 30, and 90, we found that oral administration of probiotics did not improve BV cure rates (72.73% and 84.00% at day 14, 57.14% and 60.00% at day 30, 32.14% and 48.39% at day 90 for probiotic and control group respectively). However, the probiotics were effective in restoring vaginal health after cure by showing higher proportion of participants with Nugent Scores < 4 in the probiotic group compared to the control group (87.50% and 71.43% on day 14, 93.75% and 88.89% on day 30, and 77.78% and 66.67% on day 90). The relative abundance of the probiotic strains was significantly increased in the intestinal microbiome of the probiotic group compared to the control group at day 14, but no significance was detected after 30 and 90 days. Also, the probiotics were not detected in vaginal microbiome, suggesting that L. gasseri TM13 and L. crispatus LG55 mainly acted through the intestine. A higher abundance of Prevotella timonensis at baseline was significantly associated with long-term cure failure of BV and greatly contributed to the enrichment of the lipid IVA synthesis pathway, which could aggravate inflammation response. To sum up, L. gasseri TM13 and L. crispatus LG55 can restore the vaginal health of patients recovering from BV, and individualized intervention mode should be developed to restore the vaginal health of patients recovering from BV. Clinical trial registration: https://classic.clinicaltrials.gov/ct2/show/, identifier NCT04771728.


Asunto(s)
Lactobacillus crispatus , Lactobacillus gasseri , Vaginosis Bacteriana , Femenino , Humanos , Adyuvantes Inmunológicos/uso terapéutico , Adyuvantes Farmacéuticos , Lactobacillus/fisiología , Metronidazol/uso terapéutico , Resultado del Tratamiento , Vagina/microbiología , Vaginosis Bacteriana/microbiología
14.
Sci Rep ; 13(1): 12861, 2023 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-37553432

RESUMEN

Periodontitis is an infectious inflammation in the gums characterized by loss of periodontal ligaments and alveolar bone. Its persistent inflammation could result in tooth loss and other health issues. Ixeris dentata (IXD) and Lactobacillus gasseri media (LGM) demonstrated strong antioxidant activity, which may prevent oxidative and inflammatory periodontitis. Here, IXD and LGM extracts were investigated for antioxidative activity against oral discomfort and evaluated for their synergistic effect against oxidative and inflammatory periodontitis in a mouse model. IXD/LGM suppressed pro-inflammatory cytokines like interleukin (IL)-1ß, IL-6, and TNF-α. Additionally, it reduced pro-inflammatory mediators, nitric oxide, iNOS (inducible nitric oxide synthase), and COX-2 (cyclooxygenase-2) and enhanced AKT, Nrf2, and HO-1 activation. Similarly, IXD/LGM treatment elevated osteogenic proteins and mRNAs; alkaline phosphatase, collagen type 1 (COL1), osteopontin (OPN), and runt-related transcription factor 2 (RUNX2). Hematoxylin and Eosin (H&E) staining and micro-CT analysis confirm the positive impact of IXD/LGM on the periodontal structure and its associated inflammation. These findings demonstrate that IXD/LGM inhibits oxidative stress, periodontal inflammation, and its resultant alveolar bone loss in which Akt (also known as protein kinase B)-nuclear factor-erythroid 2-related factor 2 (Nrf2)-hemoxygenase-1 (HO-1) signaling is involved. Thus, IXD/LGM is a potential candidate against oxidative/inflammatory stress-associated periodontitis.


Asunto(s)
Asteraceae , Lactobacillus gasseri , Periodontitis , Ratones , Animales , Proteínas Proto-Oncogénicas c-akt , Factor 2 Relacionado con NF-E2/metabolismo , Periodontitis/prevención & control , Inflamación , Antioxidantes , Asteraceae/metabolismo , Hemo-Oxigenasa 1
15.
J Zhejiang Univ Sci B ; 24(8): 734-748, 2023 Aug 15.
Artículo en Inglés, Chino | MEDLINE | ID: mdl-37551559

RESUMEN

A growing body of evidence has linked the gut microbiota to liver metabolism. The manipulation of intestinal microflora has been considered as a promising avenue to promote liver health. However, the effects of Lactobacillus gasseri LA39, a potential probiotic, on liver metabolism remain unclear. Accumulating studies have investigated the proteomic profile for mining the host biological events affected by microbes, and used the germ-free (GF) mouse model to evaluate host-microbe interaction. Here, we explored the effects of L. gasseri LA39 gavage on the protein expression profiles of the liver of GF mice. Our results showed that a total of 128 proteins were upregulated, whereas a total of 123 proteins were downregulated by treatment with L. gasseri LA39. Further bioinformatics analyses suggested that the primary bile acid (BA) biosynthesis pathway in the liver was activated by L. gasseri LA39. Three differentially expressed proteins (cytochrome P450 family 27 subfamily A member 1 (CYP27A1), cytochrome P450 family 7 subfamily B member 1 (CYP7B1), and cytochrome P450 family 8 subfamily B member 1 (CYP8B1)) involved in the primary BA biosynthesis pathway were further validated by western blot assay. In addition, targeted metabolomic analyses demonstrated that serum and fecal ß|-muricholic acid (a primary BA), dehydrolithocholic acid (a secondary BA), and glycolithocholic acid-3-sulfate (a secondary BA) were significantly increased by L. gasseri LA39. Thus, our data revealed that L. gasseri LA39 activates the hepatic primary BA biosynthesis and promotes the intestinal secondary BA biotransformation. Based on these findings, we suggest that L. gasseri LA39 confers an important function in the gut‒liver axis through regulating BA metabolism.


Asunto(s)
Ácidos y Sales Biliares , Lactobacillus gasseri , Ratones , Animales , Ácidos y Sales Biliares/metabolismo , Proteómica , Hígado/metabolismo , Biotransformación
16.
Anaerobe ; 82: 102761, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37467948

RESUMEN

Membrane vesicles (MVs) are bioactive, nano-sized entities produced by all organisms. MVs of L. gasseri ATCC 19992 were isolated and their effect on the biofilms of vaginal pathogens, G. vaginalis and S. aureus was studied. The L. gasseri MVs resulted in significant disruption of biofilms of the vaginal pathogens.


Asunto(s)
Lactobacillus gasseri , Femenino , Humanos , Staphylococcus aureus , Vagina , Biopelículas
17.
Braz J Microbiol ; 54(3): 2047-2062, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37430135

RESUMEN

This study aimed to compare the effects of the probiotic bacteria, L. gasseri (52b), L. plantarum (M11), L. acidophilus (AC2), and L. fermentum (19SH), isolated from human source and traditional food products on the modulation of the immune system and inflammatory response on BALB/c mouse model bearing CT26 tumor. Five groups of female inbred BALB/c mice were orally administered with the probiotics and their mixes (MIX, at a 1:1 ratio) at varying dosages (1.5 × 108 cfu/ml and 1.2 × 109 cfu/ml) before and after the injection of a subcutaneous CT26 tumor over the course of 38 days via gavage. Finally, their effects on the tumor apoptosis and the cytokine levels in spleen cell cultures were analyzed and compared. M11, MIX, and 52b groups had the greatest levels of interleukin-12 (IL-12) and interferon gamma (IFN-γ) production. The highest production level of granzyme B (GrB) was related to the MIX and 52b groups. Moreover, these groups showed the lowest production level of (IL-4) and transforming growth factor beta (TGF-ß). Furthermore, the groups of MIX and 52b demonstrated the greatest amount of lymphocyte proliferation of spleen cells in response to the tumor antigen. The delayed-type hypersensitivity (DTH) response significantly increased in the groups of MIX and 52b compared with the control (p < 0.05). The findings demonstrated that the oral treatment of the human strain (52b) and the combination of these bacteria generated strong T helper type 1 (Th1) immune responses in the tumor tissue of the tumor-bearing mice, which led to the suppression of the tumor development.


Asunto(s)
Lactobacillus gasseri , Lactobacillus plantarum , Limosilactobacillus fermentum , Neoplasias , Probióticos , Humanos , Ratones , Femenino , Animales , Lactobacillus acidophilus , Inmunidad , Probióticos/farmacología
18.
Sci Rep ; 13(1): 12212, 2023 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-37500806

RESUMEN

Probiotics are defined as live organisms in the host that contribute to health benefits. Lactobacillus gasseri LM1065, isolated from human breast milk, was investigated for its probiotic properties based on its genome. Draft genome map and de novo assembly were performed using the PacBio RS II system and hierarchical genome assembly process (HGAP). Probiotic properties were determined by the resistance to gastric conditions, adherence ability, enzyme production, safety assessment and mobile genetic elements. The fungistatic effect and inhibition of hyphae transition were studied using the cell-free supernatant (CFS). L. gasseri LM1065 showed high gastric pepsin tolerance and mild tolerance to bile salts. Auto-aggregation and hydrophobicity were measured to be 61.21% and 61.55%, respectively. The adherence to the human intestinal epithelial cells was measured to be 2.02%. Antibiotic-resistance genes and putative virulence genes were not predicted in the genomic analysis, and antibiotic susceptibility was satisfied by the criteria of the European Food Safety Authority. CFS showed a fungistatic effect and suppressed the tricarboxylic acid cycle in Candida albicans (29.02%). CFS also inhibited the transition to true hyphae and damaged the blastoconidia. This study demonstrates the essential properties of this novel probiotic, L. gasseri LM1065, and potential to inhibit vaginal C. albicans infection.


Asunto(s)
Lactobacillus gasseri , Probióticos , Femenino , Humanos , Lactobacillus gasseri/fisiología , Intestinos , Antibacterianos/farmacología , Industria de Alimentos , Probióticos/farmacología
19.
Clin Nutr ; 42(8): 1314-1321, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37413809

RESUMEN

BACKGROUND & AIM: The gut-brain axis is one of the proposed interactions between the brain and peripheral intestinal functions; of particular interest is the influence of food components on the gut-brain axis mediated via the gut microbiome. Probiotics and paraprobiotics have been proposed to interact with the intestinal environment and provide health benefits such as improving sleep quality. The aim of this research was to undertake a systematic literature review and meta-analysis to evaluate the current evidence regarding the effects of Lactobacillus gasseri CP2305 on sleep quality for the general population. METHODS: A systematic literature search was conducted of peer-reviewed articles published up to 04 November 2022. Randomised controlled trials were identified that investigated the effects of Lactobacillus gasseri CP2305 on sleep parameters in adults. Meta-analysis of the change in the Pittsburgh Sleep Quality Index (PSQI) global score was conducted. Quality assessments of individual studies were conducted using the Cochrane Risk of Bias and Health Canada tools. RESULTS: Seven studies were included in the systematic literature review; six studies included data for meta-analysis to quantify the effect of L. gasseri CP2305 on sleep quality. The ingestion of L. gasseri CP2305 resulted in significant improvement in the PSQI global score compared to control (-0.77, 95% CI: -1.37 to -0.16, P = 0.01). In the two studies that included electroencephalogram (EEG) data, output was significantly improved for at least half of the measured EEG outcomes after consumption of L. gasseri CP2305. No serious concerns were found in the potential biases of included studies, indirectness of the included evidence, and other methodological issues. CONCLUSION: The present systematic review and meta-analysis indicates significant improvement in sleep quality of adults with mild to moderate stress as an effect of daily consumption of L. gasseri CP2305. Based on existing evidence, the relationship between L. gasseri CP2305 and enhanced sleep quality is plausible, however further investigations are required to confirm the mechanisms of actions for this effect.


Asunto(s)
Microbioma Gastrointestinal , Lactobacillus gasseri , Probióticos , Humanos , Adulto , Calidad del Sueño , Sueño
20.
Enzyme Microb Technol ; 169: 110276, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37321015

RESUMEN

Lactobacillus contribute to maintain the human healthy and use for nutritional additives as probiotics. In this study, a cholesterol-lowering bacterium, Lactobacillus gasseri TF08-1, was isolated from the feces of a healthy adolescent, and its probiotic potentials were evaluated through genomic mining and in vitro test. The assembled draft genome comprised of 1,974,590 bp and was predicted total of 1,940 CDSs. The annotation of the genome revealed that L. gasseri TF08-1 harbored abundant categories of functional genes in metabolic and information processing. Moreover, strain TF08-1 has capacity to utilize D-Glucose, Sucrose, D-Maltose, Salicin, D-Xylose, D-Cellobiose, D-Mannose, and D-Trehalose, as the carbon source. The safety assessment showed strain TF08-1 contained few antibiotic resistance genes and virulence factors and was only resistant to 2 antibiotics detected by antimicrobial susceptibility test. A high bile salt hydrolase activity was found and a cholesterol-reducing effect was determined in vitro, which the result showed a remarkable cholesterol removal capability of L. gasseri TF08-1 with an efficiency of 84.40 %. This study demonstrated that the strain showed great capability of exopolysaccharide production, and tolerance to acid and bile salt. Therefore, these results indicate that L. gasseri TF08-1 can be considered as a safe candidate for probiotic, especially its potential in biotherapeutic for metabolic diseases.


Asunto(s)
Lactobacillus gasseri , Probióticos , Adolescente , Humanos , Lactobacillus gasseri/genética , Lactobacillus/genética , Heces/microbiología , Colesterol , Antibacterianos , Probióticos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA