Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 423
Filter
1.
Sci Rep ; 14(1): 9478, 2024 04 25.
Article in English | MEDLINE | ID: mdl-38658619

ABSTRACT

Irritable bowel syndrome (IBS) is frequently linked with coexisting mental illnesses. Our previous study discovered that 32.1% of IBS patients had subthreshold depression (SD), placing them at higher risk of developing major depression. Gut microbiota modulation through psychobiotics was found to influence depression via the gut-brain axis. However, the efficacy of lessening depression among IBS patients remains ambiguous. The study's aim was to investigate the roles of cultured milk drinks containing 109 cfu Lactobacillus acidophilus LA-5 and Lactobacillus paracasei L. CASEI-01 on depression and related variables among IBS participants with SD. A total of 110 IBS participants with normal mood (NM) and SD, were randomly assigned to one of four intervention groups: IBS-NM with placebo, IBS-NM with probiotic, IBS-SD with placebo, and IBS-SD with probiotic. Each participant was required to consume two bottles of cultured milk every day for a duration of 12 weeks. The following outcomes were assessed: depression risk, quality of life, the severity of IBS, and hormonal changes. The depression scores were significantly reduced in IBS-SD with probiotic and placebo from baseline (p < 0.001). Only IBS-SD with probiotic showed a significant rise in serotonin serum levels (p < 0.05). A significantly higher life quality measures were seen in IBS-SD with probiotic, IBS-SD with placebo, and IBS-NM with placebo (p < 0.05). All groups, both placebo and probiotic, reported significant improvement in IBS severity post-intervention with a higher prevalence of remission and mild IBS (p < 0.05). Dual strains lactobacillus-containing cultured milk drink via its regulation of relevant biomarkers, is a potential anti-depressive prophylactic agent for IBS patients at risk.


Subject(s)
Depression , Irritable Bowel Syndrome , Probiotics , Humans , Irritable Bowel Syndrome/microbiology , Irritable Bowel Syndrome/therapy , Irritable Bowel Syndrome/psychology , Female , Male , Adult , Probiotics/therapeutic use , Probiotics/administration & dosage , Double-Blind Method , Depression/therapy , Depression/microbiology , Middle Aged , Cultured Milk Products/microbiology , Quality of Life , Animals , Milk , Lactobacillus acidophilus/physiology , Lactobacillus , Treatment Outcome , Lacticaseibacillus paracasei
2.
PLoS One ; 19(3): e0299792, 2024.
Article in English | MEDLINE | ID: mdl-38536854

ABSTRACT

Two probiotic candidates, Lactobacillus reuteri C1 (C1) and Lactobacillus acidophilus C5 (C5), which were previously isolated from canines, were evaluated in the present study. L. reuteri and L. acidophilus have anti-oxidant, anti-inflammatory, immune-enhancing, and anti-cancer properties and exhibit various probiotic effects in humans and animals. The strains C1 and C5 demonstrated good tolerance to acid and bile salt exposure, exhibited effective adhesion to HT-29 cell monolayer, and displayed sensitivity to antibiotics, thus affirming their probiotic characteristics. Moreover, C1 and C5 exhibited the ability to downregulate the expression of inducible NO synthase (iNOS), an immunomodulatory factor, leading to a reduction in NO production in lipopolysaccharide (LPS)-stimulated RAW 264.7 cells. These strains also demonstrated potent anti-inflammatory effects in LPS-stimulated RAW 264.7 cells, achieved through the augmentation of anti-inflammatory cytokine IL-10 expression and the inhibition of pro-inflammatory cytokine IL-1ß expression. These anti-inflammatory effects of C1 and C5 were closely associated with the mitogen-activated protein kinase (MAPK) signaling pathway. The results of the present study suggest that the C1 and C5 probiotic candidates attenuate LPS-induced inflammation via the MAPK signaling pathway and the strains can be used as probiotics considering their anti-inflammatory potential.


Subject(s)
Limosilactobacillus reuteri , Probiotics , Humans , Animals , Dogs , Lactobacillus , Lipopolysaccharides/pharmacology , Lipopolysaccharides/metabolism , Anti-Inflammatory Agents/pharmacology , MAP Kinase Signaling System , Cytokines/metabolism , Feces , Lactobacillus acidophilus/physiology , Probiotics/pharmacology , NF-kappa B/metabolism
3.
Probiotics Antimicrob Proteins ; 16(2): 474-489, 2024 Apr.
Article in English | MEDLINE | ID: mdl-36976517

ABSTRACT

The present investigation was carried out with the aim to establish the comparative efficacy of a canine-sourced probiotic meant for canine feeding and a conventional dairy-sourced probiotic. For this purpose, canine-origin Lactobacillus johnsonii CPN23 and dairy-origin Lactobacillus acidophilus NCDC15 were evaluated for potential probiotics health benefits in the rat model. Forty-eight weaned Wistar rats enrolled in this experiment of 8 weeks were fed a basal diet and divided into three dietary treatments. Rats of group I enrolled as control (CON) were given MRS placebo at 1 mL/head/day, while rats of group II (LAJ) and III (LAC) were administered with overnight MRS broth grown-culture of L. johnsonii CPN23 and L. acidophilus NCDC15, respectively, at 1 mL/head/day (108 cfu/mL). The average daily gain and net gain in body weight were significantly higher (p < 0.05) in LAJ and LAC than in CON. Fecal and digesta biochemical attributes altered (p < 0.05) positively in response to both probiotics. Total fecal and pooled digesta SCFAs were higher (p < 0.05) in both LAJ and LAC than in CON. The microbial population in cecal and colonic digesta responded (p < 0.05) positively to both probiotics. The diameter of intestinal segments was higher (p < 005) in LAJ as compared to CON. The number and height of villi in jejunum tended to be higher in LAJ as compared to CON. The humoral immune response to sheep erythrocytes as well as chicken egg-white lysozyme was higher in LAJ as compared to CON. Overall, the results of the study have demonstrated the effectiveness of the canine-sourced L. johnsonii CPN23 as a potential probiotic, with a comparatively better response than the dairy-sourced L. acidophilus NCDC15. It could thus be recommended for use in feeding dogs to help augment their health.


Subject(s)
Lactobacillus acidophilus , Lactobacillus johnsonii , Probiotics , Animals , Dogs , Rats , Feces , Lactobacillus acidophilus/physiology , Rats, Wistar , Sheep , Chickens
4.
Nutrients ; 15(24)2023 Dec 12.
Article in English | MEDLINE | ID: mdl-38140334

ABSTRACT

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


Subject(s)
Ammonium Compounds , Gastrointestinal Microbiome , Probiotics , Humans , Probiotics/pharmacology , Dietary Supplements , Lactobacillus acidophilus/physiology , Fatty Acids, Volatile
5.
Wei Sheng Yan Jiu ; 52(5): 749-755, 2023 Sep.
Article in Chinese | MEDLINE | ID: mdl-37802897

ABSTRACT

OBJECTIVE: To investigate the repairing effect of Lactobacillus acidophilus LA11-Only on ceftriaxone induced intestinal micronbiota imbalance in mice and its alleviating effect on lipid metabolism. METHODS: A total of 30 SPF BALB/c male mice were randomly divided into control group(10) and model group(20). The model group was given ceftriaxone sodium solution by gavage for a week to create a mouse model of antibiotic flora disorder. The natural recovery group(10) was given 10% skimmed milk solution by gavage, and the LA11-Onlly group was given Lactobacillus acidophilus skimmed milk solution(1.0×10~9CFU/mL). Two weeks later, the feces were collected aseptically.16S rRNA sequencing technology and ultra-high-performance liquid chromatography combined with mass spectrometry(UPLC-MS) were used to analyze the repair effect of LA11-Onlly on antibiotic induced bacterial flora disorder and lipid metabolism in mice. RESULTS: After ceftriaxone treatment, the diversity of intestinal microbiota in mice decreased, and there was a significant difference in the composition of flora compared with the control group; After treatment with LA11-Onlly, the Alpha diversity increased, the abundance of beneficial bacteria such as Lactobacillus and Butyricicoccus increased, and the composition of flora was closer to the control group; At the same time, the concentration of short chain fatty acids(SCFAs) increased in varying degrees. Compared with the natural recovery group, the levels of butyric acid and valeric acid in LA11-Onlly group increased significantly, close to the same level as the control group. CONCLUSION: Lactobacillus acidophilus LA11-Onlly probiotics can inhibit the growth of harmful bacteria by regulating the diversity and community composition of intestinal microbiota, promote the increase of the concentration of SCFAs, and alleviate the damage of antibiotics to the body.


Subject(s)
Lactobacillus acidophilus , Probiotics , Mice , Male , Animals , Lactobacillus acidophilus/physiology , Ceftriaxone/pharmacology , Dysbiosis , RNA, Ribosomal, 16S , Chromatography, Liquid , Tandem Mass Spectrometry , Probiotics/pharmacology , Butyric Acid , Anti-Bacterial Agents/adverse effects , Mice, Inbred C57BL
6.
Eur J Pharm Biopharm ; 190: 81-93, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37479065

ABSTRACT

The emergence of probiotics as an alternative and adjunct to antibiotic treatment for microbiological disturbances of the female genitourinary system requires innovative delivery platforms for vaginal applications. This study developed a new, rapid-dissolving form using electrospun polyethylene oxide (PEO) fibers for delivery of antibiotic metronidazole or probiotic Lactobacillus acidophilus, and performed evaluation in vitro and in vivo. Fibers did not generate overt pathophysiology or encourage Gardnerella growth in a mouse vaginal colonization model, inducing no alterations in vaginal mucosa at 24 hr post-administration. PEO-fibers incorporating metronidazole (100 µg MET/mg polymer) effectively prevented and treated Gardnerella infections (∼3- and 2.5-log reduction, respectively, 24 hr post treatment) when administered vaginally. Incorporation of live Lactobacillus acidophilus (107 CFU/mL) demonstrated viable probiotic delivery in vitro by PEO and polyvinyl alcohol (PVA) fibers to inhibit Gardnerella (108 CFU/mL) in bacterial co-cultures (9.9- and 7.0-log reduction, respectively, 24 hr post-inoculation), and in the presence of vaginal epithelial cells (6.9- and 8.0-log reduction, respectively, 16 hr post-inoculation). Administration of Lactobacillus acidophilus in PEO-fibers achieved vaginal colonization in mice similar to colonization observed with free Lactobacillus. acidophilus. These experiments provide proof-of-concept for rapid-dissolving electrospun fibers as a successful platform for intra-vaginal antibiotic or probiotic delivery.


Subject(s)
Nanofibers , Probiotics , Female , Animals , Mice , Anti-Bacterial Agents/therapeutic use , Metronidazole , Treatment Outcome , Lactobacillus acidophilus/physiology
7.
Mol Med Rep ; 27(6)2023 Jun.
Article in English | MEDLINE | ID: mdl-37144488

ABSTRACT

The gut microbiota plays a key role in maintaining health and regulating the host's immune response. The use of probiotics and concomitant vitamins can increase mucus secretion by improving the intestinal microbial population and prevent the breakdown of tight junction proteins by reducing lipopolysaccharide concentration. Changes in the intestinal microbiome mass affect multiple metabolic and physiological functions. Studies on how this microbiome mass and the regulation in the gastrointestinal tract are affected by probiotic supplements and vitamin combinations have attracted attention. The current study evaluated vitamins K and E and probiotic combinations effects on Escherichia coli and Staphylococcus aureus. Minimal inhibition concentrations of vitamins and probiotics were determined. In addition, inhibition zone diameters, antioxidant activities and immunohistochemical evaluation of the cell for DNA damage were performed to evaluate the effects of vitamins and probiotics. At the specified dose intervals, L. acidophilus and vitamin combinations inhibit the growth of Escherichia coli and Staphylococcus aureus. It could thus contribute positively to biological functions by exerting immune system­strengthening activities.


Subject(s)
Probiotics , Staphylococcal Infections , Humans , Lactobacillus acidophilus/physiology , Escherichia coli , Staphylococcus aureus , Vitamin K 3/pharmacology , Vitamin E/pharmacology , Probiotics/pharmacology , Vitamins/pharmacology , Vitamin K
8.
Probiotics Antimicrob Proteins ; 15(6): 1529-1538, 2023 Dec.
Article in English | MEDLINE | ID: mdl-36376613

ABSTRACT

Based on the adhesion and surface properties of Lactobacillus acidophilus NCFM, five common polyphenols in fruits and vegetables, including resveratrol, epicatechin, quercetin, hesperidin, and caffeic acid, were screened, and the reasons for resveratrol promoting adhesion were systematically explained. The results showed that resveratrol could significantly enhance NCFM adhesion to mucin (1.73 fold), followed by epicatechin (1.47 fold), caffeic acid (1.30 fold), and hesperidin (0.99 fold), while quercetin had a certain degree of inhibition (0.84 fold). The effects of these polyphenols on surface hydrophobicity and auto-aggregation of NCFM were consistent with adhesion results. Then, how resveratrol promotes NCFM adhesion was further explored. The results of the proteomic analysis showed that resveratrol changed the surface layer proteins of NCFM, involving 4 up-regulated proteins and 12 down-regulated proteins. In addition, resveratrol promoted the expression of mucin genes and the glycosylation of mucins on the HT-29 cell surface. Our results indicate that resveratrol changes the surface layer proteins of NCFM to modify surface properties and adhere to mucins. Meanwhile, resveratrol promotes expression and glycosylation of mucins in HT-29 cells. Our findings provide theoretical support for an in-depth explanation of the interaction among resveratrol, NCFM, and the HT-29 cells.


Subject(s)
Catechin , Hesperidin , Humans , Resveratrol/pharmacology , Resveratrol/metabolism , Lactobacillus acidophilus/physiology , Bacterial Adhesion , Catechin/pharmacology , Hesperidin/metabolism , Hesperidin/pharmacology , Proteomics , Quercetin/pharmacology , Quercetin/metabolism , Polyphenols/metabolism , Polyphenols/pharmacology , Mucins/metabolism
9.
J Anim Physiol Anim Nutr (Berl) ; 107(1): 165-172, 2023 Jan.
Article in English | MEDLINE | ID: mdl-34726311

ABSTRACT

This study investigates the use of nanofiber microcapsules produced by electrostatic spinning as a carrier for the delivery of lactic acid bacteria (LAB) to the intestine of ruminants. We hypothesized that the LAB encapsulated into nanofiber microcapsules can be delivered to a ruminant's intestinal tract with little effect on the rumen fermentation and related bacteria. The in vitro experiment included three treatments: control group; 0.01g Lactobacillus acidophilus NCFM (L. acidophilus NCFM) encapsulated in nanofiber microcapsules by electrostatic spinning group (ELAN, 2.0 × 1011  CFU/g); and 0.01g L. acidophilus NCFM powder group (LANP, 2.0 × 1011  CFU/g), each incubated with 30 ml of buffer rumen fluid for 48h to determine the effect on rumen fermentation, then the abundance of L. acidophilus NCFM in the intestine was estimated using the modified in vitro three-step procedure. Treatment responses were statistically analysed using one-way ANOVA. The results showed that compared to the control, the ELAN group had a significant increase in pH (p < 0.05), while the LANP group had a non-significant decrease in pH (p > 0.05). LANP and ELAN groups had no significant influence on total volatile fatty acid and individual volatile fatty acids (p > 0.05), apart from isobutyric acid of both groups, which reduced (p < 0.05). ELAN group had a decreasing trend of gas production and dry matter digestion, while the LANP group increased them significantly (p < 0.05). During the 16h and 48h rumen incubation, compared with control, there was no significant change in all bacteria in the ELAN group (p > 0.05), while the LANP group increased the relative abundance levels of S. bovis, S. ruminantium, M. elsdenii, F. succinogenes, B. fibrisolvens, Lactobacillus, L. acidophilus NCFM (p < 0.05). In the intestinal part, compared with control, the relative abundance of L. acidophilus NCFM in the ELAN group increased significantly (p < 0.05), while the result was not observed in the LANP group. We concluded based on our findings that L. acidophilus NCFM could be protected by nanofiber microcapsules and delivered to the intestinal site with little influence on the rumen fermentation and bacterial community, suggesting nanofiber microcapsules prepared by electrospinning technology could be used as a carrier for rumen-protected study.


Subject(s)
Lactobacillales , Nanofibers , Animals , Capsules/metabolism , Intestines , Lactobacillus acidophilus/physiology , Fatty Acids, Volatile/metabolism , Ruminants , Rumen/metabolism , Fermentation
10.
Appl Neuropsychol Adult ; 30(5): 552-560, 2023.
Article in English | MEDLINE | ID: mdl-34493130

ABSTRACT

A growing body of research studies the relationship between probiotic bacteria in the gut and the host organism, including the impact on cognitive functioning. Data from human studies are scarce; however, recent studies point toward the beneficial role of probiotics for cognitive functioning. One of the mechanisms involved in this relationship is the probiotic's ability to influence inflammation and immune response. The aim of this initial study was to investigate the effects of probiotic supplementation with Bifidobacterium Lactis BS01 and Lactobacillus Acidophilus LA02 on cognitive functioning in healthy, young adult females. A total of 53 participants aged 19-31 were enrolled, and 38 completed the trial. A 6-week probiotic or placebo treatment was conducted. Five measures of cognitive functioning were applied pre- and post-treatment. Both groups showed general improvement at the second assessment. Contrary to our hypothesis, the placebo group improved slightly, but significantly, in four out of five measures of cognitive functioning, with the exception of the Wisconsin Card Sorting Test (WCST). The supplementation group improved significantly in two measures of the WCST, compared to the placebo group. Similar results have been previously reported. Probiotic supplementation, while not harmful, might not be beneficial for cognition in the healthy population, or at least not universally.


Subject(s)
Bifidobacterium animalis , Probiotics , Female , Humans , Young Adult , Bifidobacterium/physiology , Cognition , Double-Blind Method , Lactobacillus acidophilus/physiology , Probiotics/pharmacology , Probiotics/therapeutic use
11.
J Microbiol Biotechnol ; 32(10): 1226-1233, 2022 Oct 28.
Article in English | MEDLINE | ID: mdl-36196014

ABSTRACT

Probiotics are live microorganisms that can be consumed by humans in amounts sufficient to offer health-promoting effects. Owing to their various biological functions, probiotics are widely used in biological engineering, industry and agriculture, food safety, and the life and health fields. Lactobacillus acidophilus (L. acidophilus), an important human intestinal probiotic, was originally isolated from the human gastrointestinal tract and its functions have been widely studied ever since it was named in 1900. L. acidophilus has been found to play important roles in many aspects of human health. Due to its good resistance against acid and bile salts, it has broad application prospects in functional, edible probiotic preparations. In this review, we explore the basic characteristics and biological functions of L. acidophilus based on the research progress made thus far worldwide. Various problems to be solved regarding the applications of probiotic products and their future development are also discussed.


Subject(s)
Lactobacillus acidophilus , Probiotics , Humans , Lactobacillus acidophilus/physiology , Intestines , Gastrointestinal Tract , Bile Acids and Salts/pharmacology
12.
Nutrients ; 14(5)2022 Feb 25.
Article in English | MEDLINE | ID: mdl-35267950

ABSTRACT

Age-related alterations in the gut microbiome composition and its impacts on the host's health have been well-described; however, detailed analyses of the gut microbial structure defining ecological microbe-microbe interactions are limited. One of the ways to determine these interactions is by understanding microbial co-occurrence patterns. We previously showed promising abilities of Lactobacillus acidophilus DDS-1 on the aging gut microbiome and immune system. However, the potential of the DDS-1 strain to modulate microbial co-occurrence patterns is unknown. Hence, we aimed to investigate the ability of L. acidophilus DDS-1 to modulate the fecal-, mucosal-, and cecal-related microbial co-occurrence networks in young and aging C57BL/6J mice. Our Kendall's tau correlation measures of co-occurrence revealed age-related changes in the gut microbiome, which were characterized by a reduced number of nodes and associations across sample types when compared to younger mice. After four-week supplementation, L. acidophilus DDS-1 differentially modulated the overall microbial community structure in fecal and mucosal samples as compared to cecal samples. Beneficial bacteria such as Lactobacillus, Oscillospira, and Akkermansia acted as connectors in aging networks in response to L. acidophilus DDS-1 supplementation. Our findings provided the first evidence of the DDS-1-induced gut microbial ecological interactions, revealing the complex structure of microbial ecosystems with age.


Subject(s)
Gastrointestinal Microbiome , Microbiota , Aging , Animals , Lactobacillus acidophilus/physiology , Mice , Mice, Inbred C57BL
13.
Microbiol Spectr ; 10(1): e0181521, 2022 02 23.
Article in English | MEDLINE | ID: mdl-35019699

ABSTRACT

Evidence for the concept of the "gut-brain axis" (GBA) has risen. Many types of research demonstrated the mechanism of the GBA and the effect of probiotic intake. Although many studies have been reported, most were focused on neurodegenerative disease and, it is still not clear what type of bacterial strains have positive effects. We designed an experiment to discover a strain that positively affects brain function, which can be recognized through changes in cognitive processes using healthy mice. The experimental group consisted of a control group and three probiotic consumption groups, namely, Lactobacillus acidophilus, Lacticaseibacillus paracasei, and Lacticaseibacillus rhamnosus. Three experimental groups fed probiotics showed an improved cognitive ability by cognitive-behavioral tests, and the group fed on L. acidophilus showed the highest score. To provide an understanding of the altered microbial composition effect on the brain, we performed full 16S-23S rRNA sequencing using Nanopore, and operational taxonomic units (OTUs) were identified at species level. In the group fed on L. acidophilus, the intestinal bacterial ratio of Firmicutes and Proteobacteria phyla increased, and the bacterial proportions of 16 species were significantly different from those of the control group. We estimated that the positive results on the cognitive behavioral tests were due to the increased proportion of the L. acidophilus EG004 strain in the subjects' intestines since the strain can produce butyrate and therefore modulate neurotransmitters and neurotrophic factors. We expect that this strain expands the industrial field of L. acidophilus and helps understand the mechanism of the gut-brain axis. IMPORTANCE Recently, the concept of the "gut-brain axis" has risen and suggested that microbes in the GI tract affect the brain by modulating signal molecules. Although many pieces of research were reported in a short period, a signaling mechanism and the effects of a specific bacterial strain are still unclear. Besides, since most of the research was focused on neurodegenerative disease, the study with a healthy animal model is still insufficient. In this study, we show using a healthy animal model that a bacterial strain (Lactobacillus acidophilus EG004) has a positive effect on mouse cognitive ability. We experimentally verified an improved cognitive ability by cognitive behavioral tests. We performed full 16S-23S rRNA sequencing using a Nanopore MinION instrument and provided the gut microbiome composition at the species level. This microbiome composition consisted of candidate microbial groups as a biomarker that shows positive effects on cognitive ability. Therefore, our study suggests a new perspective for probiotic strain use applicable for various industrialization processes.


Subject(s)
Cognition , Feces/microbiology , Gastrointestinal Microbiome/genetics , Gastrointestinal Microbiome/physiology , Lactobacillus acidophilus/genetics , Lactobacillus acidophilus/physiology , Metagenome , RNA, Ribosomal, 23S/genetics , Animals , Biodiversity , Brain-Gut Axis , Disease Models, Animal , Lactobacillus/genetics , Lactobacillus/physiology , Male , Mice , Mice, Inbred C57BL , Neurodegenerative Diseases , Probiotics/pharmacology , Probiotics/therapeutic use
14.
Cytokine ; 149: 155743, 2022 01.
Article in English | MEDLINE | ID: mdl-34662821

ABSTRACT

Immunotherapy has been applied in cancer treatments for many years as an alternative treatment method to radiotherapy, chemotherapy. It is well known that immunotherapy could suppress tumor formation by modulating the immune system of the host. The aim of the study is to investigate supportive therapy potential of acidophilus milk (AS) and propolis extract (PE) in the mouse xenograft breast cancer model. For this purpose, firstly cytotoxic effect of PE was determined by MTT assay against 4 T1 mouse breast cancer cells. Apoptotic effect of PE analyzed by flow cytometry. The antibacterial activity of PE was determined by the 96-well microplate broth-dilution method on Lactobacillus acidophilus LA-5. Then, Balb/c mice were injected subcutaneously with 4 T1 cells (2x105 cells/mouse) and also mice were given daily oral gavage with PE (66 mg/kg/day) and/or acidophilus milk (108 CFU/mL/mouse/day) for 14 days. The Balb/c mice were weighed throughout the study, and the tumor sizes were measured by caliper at the 14th day. The proliferation of splenocytes which collected spleen from mice was measured by MTT. CD8 + T cell response was analyzed by flow cytometry and results were evaluated in comparison with control and tumor control groups. The IC50 value for PE on 4 T1 cells was determined as 129.25 ± 1.90 µg/mL. The apoptotic effect of PE at IC50 concentration was determined as 3.3% of cells to late-apoptosis, 4.3% of cells to pro-apoptosis and 2.5% of cells to necrosis. The MIC and MBC values for PE on L. acidophilus LA-5 were 5000 ppm. The treatment of PE, AS and the combination of PE and AS were inhibited the tumor volumes by 59.16%, 28.29% and 63.39%, respectively. Acidophilus milk and PE combination significantly enhanced the ConA-, LPS- and PHA-induced splenocyte proliferation (P < 0.05). The acidophilus milk and PE combination were also found to stimulate IFN- γ production. In conclusion, the best anti-tumor effect was obtained by the combination of acidophilus milk and propolis.


Subject(s)
Breast Neoplasms/drug therapy , Lactobacillus acidophilus/physiology , Milk/microbiology , Propolis/pharmacology , Administration, Oral , Animals , Anti-Bacterial Agents/pharmacology , Breast Neoplasms/metabolism , Cell Line, Tumor , Female , Immunologic Factors/metabolism , Mice , Mice, Inbred BALB C , Probiotics/pharmacology , Spleen/drug effects , Spleen/metabolism
15.
Int. j. morphol ; 39(5): 1493-1501, oct. 2021. ilus, tab
Article in English | LILACS | ID: biblio-1385512

ABSTRACT

SUMMARY: This study evaluates the effect of probiotics Saccharomyces cerevisiae, Lactobacillus acidophilus and Bacillus subtilis on production parameters and intestinal histomorphology of broilers of 45 days of age. Eleven 45-day-old Ross 500 broilers were used and classified as control group (CG) (n = 5) or supplemented with probiotics group (n = 8). Histopathological evaluation of duodenum, ileum, and jejunum was performed. The area of the villi height, base and apex were evaluated as well as the size and number of crypts. In addition, mucus production was quantified in different portions of the small intestine. The villi present duodenum of broilers supplemented with probiotics had a greater area (p = 0.0127), a greater basal width (p = 0.0049) and a greater apical width (p = 0.0024), as well as a greater crypt area (p = 0.0189). Significantly higher levels of mucus were noted in the duodenum (p = 0.0480) and jejunum (p = 0.0480) of broilers supplemented with probiotics. We suggest that probiotic supplementation improve the intestinal nutrients absorption.


RESUMEN: Este estudio evalúa el efecto del uso de probióticos como: Saccharomyces cerevisiae, Lactobacillus acidophilus, Bacillus subtilis en parámetros productivos e histomorfologia intestinal de pollos de engorde de 45 días de edad. Fueron usados 11, los cuales fueron clasificados en grupo control (CG) (n = 5) y grupo suplementado con probióticos (PG) (n = 8). Fue realizado análisis histopatológico de secciones de duodeno, íleon y yeyuno. Fue evaluado ancho, altura y área del ápice de la vellosidad, área y número de criptas. Además, fue estimada la producción de moco en los diferentes segmentos del intestino delgado. Fue observada mayor área de la vellosidad en duodeno, PG (p = 0.0127), ancho basal mayor en PG (p = 0.0049) ancho apical mayor en PG (p = 0.0024), así como mayor área de criptas en PG (p = 0.0189). No fueron encontradas diferencias significativas respecto a los segmentos de yeyuno e íleon. PG presentó mayor producción de moco en duodeno (p = 0.0480) y en yeyuno (p = 0.0480). Concluimos que la suplementación con probióticos en pollos de engorde genera cambios en la histomorfologia intestinal, evidenciables en áreas apicales y basales de las vellosidades intestinales. Soporte financiero: Dirección General de Investigaciones - Universidad de los Llanos.


Subject(s)
Animals , Poultry , Probiotics/administration & dosage , Intestines/anatomy & histology , Saccharomyces cerevisiae/physiology , Bacillus subtilis/physiology , Chickens , Dietary Supplements , Intestines/microbiology , Lactobacillus acidophilus/physiology
16.
Braz J Microbiol ; 52(4): 2319-2334, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34487329

ABSTRACT

In this study, different dairy products such as ice cream, yoghurt, white pickled cheese, and fermented acidophilus milk were manufactured by using either Lactobacillus acidophilus DSM 20,079 or Lactobacillus acidophilus NCFM. The counts of L. acidophilus in the samples on days 1, 15, and 30 of the storage were determined. Additionally, the samples contained L. acidophilus were passed through a dynamic gastrointestinal model designed in laboratory conditions to compare the protective effect of different dairy products on viability of L. acidophilus against stress factors of the gastrointestinal model. The counts of L. acidophilus NCFM and L. acidophilus DSM 20,079 in the samples decreased by between 0.04 and 0.37 log units and by between 0.11 and 0.27 log units, respectively, within 30 days of storage. During the passage through the gastrointestinal model, the highest percentage reduction in the counts of L. acidophilus was determined in yoghurt followed by fermented acidophilus milk, white pickled cheese, and ice cream, respectively. The reduction in the counts of L. acidophilus in the samples during the passage through the model increased with extension of storage time. The results of this study showed that the reduction in the counts L. acidophilus in the samples during the passage through the model was influenced significantly by the matrix of the dairy product and storage period.


Subject(s)
Dairy Products/microbiology , Gastrointestinal Tract/microbiology , Lactobacillus acidophilus , Probiotics , Animals , Cheese/microbiology , Lactobacillus acidophilus/physiology , Milk/microbiology , Models, Biological , Time Factors , Yogurt/microbiology
17.
Benef Microbes ; 12(1): 85-93, 2021 Feb 24.
Article in English | MEDLINE | ID: mdl-33550937

ABSTRACT

Several studies have demonstrated a decrease in upper respiratory infection (URI) frequency and severity in subjects taking probiotic supplements. We hypothesised beneficial effects of probiotics on viral URI in children are due to modulation of inflammatory innate immune responses. We tested this hypothesis, providing children with a probiotic combination of Lactobacillus acidophilus/Bidfidobacterium animalis ssp. lactis Bi-07 (NCFM/Bi-07) and measuring levels of cytokines in response to stimulation of peripheral blood mononuclear cells (PBMCs) to toll-like receptor (TLR) 7/8 agonist resiquimod (R848). In this open label study, 21 (2 dropouts) children received probiotic containing 5×109 cfu each of NCFM/(Bi-07) daily for 30 days. Whole blood was taken from each subject at study entry and 30 days for culture of PBMCs. PBMCs stimulated with resiquimod (R848) or unstimulated were incubated and a panel of immune markers was measured. There was a significant decrease in the net (stimulated-null) level of myeloid progenitor inhibitory factor 1 (MPIF-1) (mean decrease 0.1 ng/ml, 95% confidence interval 0.01-0.24, P=0.032) following probiotic supplementation. The change in immune marker levels after supplementation, when analysed together with respect to expected inflammatory/anti-inflammatory effects, was increased for interleukin (IL)-10 and decreased for MPIF-1, IL-8, interferon gamma induced protein 10, macrophage inflammatory protein 3 alpha (MIP-3α) and E-selectin (P=0.01). Adverse events were mild. In conclusion, supplementation with this probiotic combination was safe and resulted in significant modulation of PBMC limited immune response to TLR7/8 agonist R848 and in levels of MPIF-1 and MIP-3α. The anti-inflammatory effect may be one mechanism by which probiotics modulate the immune system however further study is needed.


Subject(s)
Bifidobacterium animalis/physiology , Imidazoles/administration & dosage , Lactobacillus acidophilus/physiology , Leukocytes, Mononuclear/drug effects , Probiotics/administration & dosage , Respiratory Tract Infections/drug therapy , Child, Preschool , Female , Humans , Immunity, Innate , Infant , Interleukin-10/genetics , Interleukin-10/immunology , Leukocytes, Mononuclear/immunology , Male , Respiratory Tract Infections/genetics , Respiratory Tract Infections/immunology , Toll-Like Receptor 7/genetics , Toll-Like Receptor 7/immunology , Toll-Like Receptor 8/genetics , Toll-Like Receptor 8/immunology
18.
Future Microbiol ; 16: 241-255, 2021 03.
Article in English | MEDLINE | ID: mdl-33625248

ABSTRACT

Aim: The inhibitory and antibiofilm effects of Thymus vulgaris (EOTv) and Hyptis spicigera essential oils (EOHs) on cariogenic microorganisms were evaluated. Materials & methods: The chemical characterization of EOTv was performed by gas chromatography/mass spectrometry. Streptococcus mutans, Streptococcus gordonii, Streptococcus sanguinis, Streptococcus mitis, Streptococcus sobrinus, Lactobacillus acidophilus and Actinomyces naeslundii were used for agar diffusion assays and determination of minimal inhibitory and minimal bactericide concentrations. In addition, 20 streptococci and lactobacilli clinical isolates were also tested. The effects of essential oil on microbial initial biofilm formation and on preformed microcosm biofilm formed from human saliva were studied. Results & conclusion: Both essential oils had inhibitory effects on the cariogenic species and reduced the bacterial adherence to dental enamel. Essential oils were able to disrupt preformed microcosm biofilms. Thymus vulgaris and Hyptis spicigera essential oils have potential to be used in the development of formulations to the control of cariogenic biofilms.


Subject(s)
Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Dental Caries/microbiology , Hyptis/chemistry , Oils, Volatile/pharmacology , Plant Oils/pharmacology , Thymus Plant/chemistry , Actinomyces/drug effects , Actinomyces/physiology , Anti-Bacterial Agents/chemistry , Humans , Lactobacillus acidophilus/drug effects , Lactobacillus acidophilus/physiology , Oils, Volatile/chemistry , Plant Oils/chemistry , Saliva/microbiology , Streptococcus/drug effects , Streptococcus/physiology
19.
Am J Pathol ; 191(5): 872-884, 2021 05.
Article in English | MEDLINE | ID: mdl-33607043

ABSTRACT

Defective intestinal tight junction (TJ) barrier is an important pathogenic factor of inflammatory bowel disease. To date, no effective therapies that specifically target the intestinal TJ barrier are available. The purpose of this study was to identify probiotic bacterial species or strains that induce a rapid and sustained enhancement of intestinal TJ barrier and protect against the development of intestinal inflammation by targeting the TJ barrier. After high-throughput screening of >20 Lactobacillus and other probiotic bacterial species or strains, a specific strain of Lactobacillus acidophilus, referred to as LA1, uniquely produced a marked enhancement of the intestinal TJ barrier. LA1 attached to the apical membrane surface of intestinal epithelial cells in a Toll-like receptor (TLR)-2-dependent manner and caused a rapid increase in enterocyte TLR-2 membrane expression and TLR-2/TLR-1 and TLR-2/TLR-6 hetero-complex-dependent enhancement in intestinal TJ barrier function. Oral administration of LA1 caused a rapid enhancement in mouse intestinal TJ barrier, protected against a dextran sodium sulfate (DSS) increase in intestinal permeability, and prevented the DSS-induced colitis in a TLR-2- and intestinal TJ barrier-dependent manner. In conclusion, we report for the first time that a specific strain of LA causes a strain-specific enhancement of intestinal TJ barrier through a novel mechanism that involves the TLR-2 receptor complex and protects against the DSS-induced colitis by targeting the intestinal TJ barrier.


Subject(s)
Colitis/prevention & control , Inflammation/prevention & control , Lactobacillus acidophilus/physiology , Probiotics , Toll-Like Receptor 2/metabolism , Animals , Colitis/chemically induced , Colitis/microbiology , Colitis/pathology , Dextran Sulfate/adverse effects , Epithelial Cells/pathology , Intestines/drug effects , Intestines/pathology , Mice , Mice, Inbred C57BL , Permeability/drug effects , Tight Junctions/drug effects , Tight Junctions/pathology , Toll-Like Receptor 2/genetics
20.
Food Chem Toxicol ; 149: 112049, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33561518

ABSTRACT

The effect of sesamin on intestinal flora was studied by in vitro animal fecal anaerobic fermentation system, and were analyzed by 16S rDNA sequencing. Results showed that sesamin modulated the composition of intestinal microorganisms and reshaped gut microbiome. The abundance of probiotics Lactobacillaceae and Bifidobacteriaceae increased, while the abundance of Enterobacteriaceae decreased. The properties of probiotics (Bifidobacterium bifidum and Lactophilus acidophilus) adhesion to epithelial colon cells (NCM460) were assessed by gram staining and plate counting methods. Results showed that sesamin increased the adhesive index of probiotics. Analysis of RT-qPCR, Western blot and immunofluorescence staining indicated that sesamin up-regulated the expression of the adhesive protein (ß-cadherin and E-cadherin) of NCM460 cells. In conclusion, sesamin could promote the proliferation and adhesion of intestinal probiotics leading to modulating gut microbiota, which provided basis for sesamin as a food-borne functional factor for improving intestinal health.


Subject(s)
Bifidobacterium bifidum/drug effects , Dioxoles/pharmacology , Lactobacillus acidophilus/drug effects , Lignans/pharmacology , Animals , Bacterial Adhesion/drug effects , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bifidobacterium bifidum/physiology , Cell Line , Dioxoles/administration & dosage , Dose-Response Relationship, Drug , Epithelial Cells/drug effects , Feces/microbiology , Gastrointestinal Microbiome/drug effects , Gene Expression Regulation, Bacterial , Humans , Intestinal Mucosa/cytology , Lactobacillus acidophilus/physiology , Lignans/administration & dosage , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...