Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 134
Filter
Add more filters

Complementary Medicines
Country/Region as subject
Publication year range
1.
Int J Biol Macromol ; 242(Pt 2): 124650, 2023 Jul 01.
Article in English | MEDLINE | ID: mdl-37119914

ABSTRACT

Amuc_1100 (hereafter called Amuc) is a highly abundant pili-like protein on the outer membrane of Akkermansia muciniphila and has been found to be effective for in anti-obesity, which is probably through the activation of TLR2. However, the precise mechanisms underlying the contributions of TLR2 to obesity resistance remain unknown. Here, TLR2 knockout mice were used to decipher the anti-obesity mechanism of Amuc. Mice exposed to a high-fat diet (HFD) were treated with Amuc (60 µg) every other day for 8 weeks. The results showed that Amuc supplementation decreased mouse body weight and lipid deposition by regulating fatty acid metabolism and reducing bile acid synthesis by activating TGR5 and FXR and strengthening the intestinal barrier function. The ablation of TLR2 partially reversed the positive effect of Amuc on obesity. Furthermore, we revealed that Amuc altered the gut microbiota composition by increasing the relative abundance of Peptostreptococcaceae, Faecalibaculum, Butyricicoccus, and Mucispirillum_schaedleri_ASF457, and decreasing Desulfovibrionaceae, which may serve as a contributor for Amuc to reinforce the intestinal barrier in HFD-induced mice. Therefore, the anti-obesity effect of Amuc was accompanied by the mitigation of gut microbes. These findings provide support for the use of Amuc as a therapy targeting obesity-associated metabolic syndrome.


Subject(s)
Gastrointestinal Microbiome , Metabolic Syndrome , Mice , Animals , Diet, High-Fat/adverse effects , Toll-Like Receptor 2 , Verrucomicrobia , Obesity/etiology , Obesity/chemically induced , Fatty Acids/pharmacology , Bile Acids and Salts/pharmacology , Mice, Inbred C57BL
2.
Cell Biol Toxicol ; 39(4): 1215-1235, 2023 08.
Article in English | MEDLINE | ID: mdl-35802278

ABSTRACT

Epimedii folium (EF) is an effective herbal medicine in osteoporosis treatment, but the clinical utilization of EF has been limited due to potential hepatotoxicity. The previous studies identified that baohuoside I (BI), the main active component of EF, was relevant to EF-induced liver injury. However, the mechanisms of BI causing direct injury to hepatocytes remain unclear. Here, we reveal that BI inhibits FXR-mediated signaling pathway via targeting estrogen receptor α (ER α), leading to the accumulation of bile acids (BAs). Targeted bile acid analyses show BI alters the BA composition and distribution, resulting in impaired BA homeostasis. Mechanistically, BI induces FXR-dependent hepatotoxicity at transcriptional level. Additionally, ER α is predicted to bind to the FXR promoter region based on transcription factor binding sites databases and we further demonstrate that ER α positively regulates FXR promoter activity and affects the expression of target genes involved in BA metabolism. Importantly, we discover that ER α and its mediated FXR transcription regulation might be involved in BI-induced liver injury via ligand-dependent ER α degradation. Collectively, our findings indicate that FXR is a newly discovered target gene of ER α mediated BI-induced liver injury, and suggest BI may be responsible for EF-induced liver injury.


Subject(s)
Chemical and Drug Induced Liver Injury, Chronic , Receptors, Cytoplasmic and Nuclear , Humans , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Cytoplasmic and Nuclear/pharmacology , Bile Acids and Salts/metabolism , Bile Acids and Salts/pharmacology , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Chemical and Drug Induced Liver Injury, Chronic/metabolism , Liver , Homeostasis , Signal Transduction
3.
Front Immunol ; 13: 983501, 2022.
Article in English | MEDLINE | ID: mdl-36389720

ABSTRACT

With the rapid aging of the population, the control of age-related disease susceptibility and prognosis faces greater challenges. There is an urgent need for a strategy to maintain the vitality of elderly people. In this study, the effect of Renshen Guben (RSGB) oral liquid was investigated on an accelerated aging mice model of thyrotoxicosis by conventional detection methods combined with multiomics technology. The results showed that RSGB increased the number of neutrophils and lymphocytes, enhanced the function of lymphocytes, and increased the levels of complement and antimicrobial peptides, which indicated that RSGB improved the immunity of thyrotoxicosis mice at the cellular and molecular levels. RSGB corrected malnutrition in thyrotoxicosis mice by improving anemia, hypoalbuminemia, ion transporters, and vitamin-binding proteins. RSGB significantly reduced the lipotoxicity by reducing the level of fatty acids, triglyceride, sphingolipids, and glucocorticoids, thus increasing the level of docosapentaenoic acid (DPA) and bile acids, which contributed to improve immunosenescence. The intestinal defense ability of thyrotoxicosis mice was enhanced with the increase of bile acids and lactic acid bacteria by the RSGB treatment. The plant metabolomics analysis showed that there were various active components in RSGB oral liquid and medicated serum, including terpenoids, phenolic acids, flavonoids, tannin, alkaloids, organic acids, phenolamines, amino acids, and others. They have antioxidant, immune regulation, and anti-aging effects, which was the material basis of RSGB. Totally, RSGB protected the thyrotoxicosis mice against aging by improving immunosenescence, hypoproteinemia, lipotoxicity, and the intestinal flora. It will be beneficial for improving the disease susceptibility and prognosis of the elderly.


Subject(s)
Gastrointestinal Microbiome , Hypoproteinemia , Immunosenescence , Panax , Thyrotoxicosis , Mice , Animals , Disease Susceptibility , Aging , Bile Acids and Salts/pharmacology
4.
FEMS Microbiol Lett ; 369(1)2022 10 17.
Article in English | MEDLINE | ID: mdl-36073496

ABSTRACT

Although probiotics have been isolated from different sources, few were isolated from traditional Chinese medicine. The current study firstly isolates Pulsatilla Radix-utilising Pediococcus pentosaceus PR-1 from human faeces. Subsequently, the tolerance of PR-1 to low pH, bile salts, simulated gastric juice and succus entericus, antioxidant activity, antimicrobial activity, cholesterol assimilation and antibiotics susceptibility were investigated. After 2 h of incubation at pH 2.0, over 80% of PR-1 survived. The cell viability of PR-1 at 2 h under 0.1% bile salt condition was 99.2%. The survival rate of PR-1 in gastric juice and succus entericus was 64.48% and 81.86%, respectively. Cell-free supernatant of PR-1 culture also showed antimicrobial activity against Escherichia coli, Staphylococcus aureus and Salmonella typhimurium. Besides, antioxidant activity of PR-1 CFS was significantly greater than cell pellet. PR-1 was shown to be resistant to kanamycin, streptomycin, vancomycin and norfloxacin and was able to lower the cholesterol level to 72.5±1.5%. In addition, PR-1 displayed γ-haemolysis and was non-pathogenic.


Subject(s)
Anti-Infective Agents , Probiotics , Pulsatilla , Anti-Bacterial Agents/pharmacology , Antioxidants/pharmacology , Bile Acids and Salts/pharmacology , Feces/microbiology , Humans , Kanamycin , Norfloxacin , Pediococcus , Pediococcus pentosaceus , Streptomycin , Vancomycin
5.
Front Cell Infect Microbiol ; 12: 945263, 2022.
Article in English | MEDLINE | ID: mdl-36072221

ABSTRACT

Qiwei Baizhu Powder (QWBZP) is a traditional Chinese medicine formula for treating diarrhea induced by various causes. It elicits an anti-diarrheal effect by regulating the gut microbiota (diversity, structure, and abundance). However, the contribution of different components in the QWBZP decoction to this effect remains unclear. In this study, we used the QWBZP decoction as a reference standard to investigate the effects of total glycosides (TGs) extracted from QWBZP decoction on the gut microbiota and bile acid metabolism in mice with antibiotic-associated diarrhea (AAD). The results of 16S rRNA gene sequencing and liquid chromatography-mass spectrometry (LC-MS) analysis showed that the effect of total glycosides of Qiwei Baizhu Powder (QWBZP-TG) on specific intestinal bacteria and bile acids was similar to that of the QWBZP decoction, but the intensity of this effect was more significant in the case of QWBZP-TG. The QWBZP decoction and QWBZP-TG promoted the proliferation of Lactobacillus and inhibited the growth of Proteus, Clostridium, Eubacterium, Facklamia, and Escherichia in AAD mice. They also increased the levels of deoxycholic acid and beta-muricholic acid and decreased those of taurocholate acid, tauro-alpha-muricholic acid, and tauro-beta-muricholic acid in AAD mice. Lactobacillus was the key bacterial genus responding to QWBZP-TG. Thus, this study provides novel insights into the bioactive components of QWBZP and their contribution to its effects.


Subject(s)
Gastrointestinal Microbiome , Animals , Bile Acids and Salts/pharmacology , Diarrhea/drug therapy , Glycosides , Mice , Powders/pharmacology , RNA, Ribosomal, 16S/genetics
6.
Int J Biol Macromol ; 219: 964-979, 2022 Oct 31.
Article in English | MEDLINE | ID: mdl-35940431

ABSTRACT

Organic chromium is of great interest and has become an important chromium supplement resource in recent years because of its low toxicity and easy absorption. In our previous study, we synthesized a novel organic chromium [GLP-Cr] through the chelation of Ganoderma lucidum polysaccharide and chromium (III). The purpose of this study was to investigate the beneficial effects of GLP-Cr on the improvement of metabolic syndromes (MetS) in mice fed with a high-fat and high-fructose diet (HFHFD) and its mechanism of action. The results indicated that oral administration of GLP-Cr inhibited the excessive exaltation of body weight, glucose tolerance, fasting blood glucose and lipid levels, hepatic total cholesterol (TC), triglyceride (TG) levels caused by HFHFD. Besides, 16S rRNA amplicon sequencing showed that GLP-Cr intervention evidently ameliorated intestinal microbiota dysbiosis by changing the proportions of some intestinal microbial phylotypes. In addition, correlation network-based analysis indicated that the key intestinal microbial phylotypes were closely related to biochemical parameters associated with MetS under GLP-Cr intervention. Liver metabolomics analysis suggested that GLP-Cr intervention significantly regulated the levels of some biomarkers involved in alpha-linolenic acid metabolism, fatty acid biosynthesis, steroid hormone biosynthesis, glycerophospholipid metabolism, glycerolipid metabolism, steroid hormone biosynthesis, primary bile acid biosynthesis, and so on. Moreover, GLP-Cr intervention regulated liver mRNA levels of key genes associated with glucose and lipid metabolism. The mRNA level of glucose transporter type 4 (Glut4) was markedly increased by GLP-Cr intervention, and the mRNA levels of phosphoenolpyruvate carboxykinase (Pepck) and glucose-6-phosphatase (G6Pase) in the liver were significantly decreased. Meanwhile, GLP-Cr intervention significantly decreased hepatic mRNA levels of cluster of differentiation 36 (Cd36), acetyl-CoA carboxylase 1 (Acc1) and sterol regulatory element binding protein-1c (Srebp-1c), indicating that GLP-Cr intervention inhibited the excessive accumulation of free fatty acids in the liver. These findings suggest that the prevention of hyperglycemia and dyslipidemia by GLP-Cr may be closely related to the regulation of gut microbial composition and hepatic metabolic pathways, thus GLP-Cr can be serving as a functional component in the prevention of MetS.


Subject(s)
Gastrointestinal Microbiome , Metabolic Syndrome , Reishi , Acetyl-CoA Carboxylase/genetics , Acetyl-CoA Carboxylase/metabolism , Acetyl-CoA Carboxylase/pharmacology , Animals , Bile Acids and Salts/pharmacology , Biomarkers , Blood Glucose/metabolism , Cholesterol , Chromium/chemistry , Diet , Diet, High-Fat/adverse effects , Dysbiosis/drug therapy , Fatty Acids, Nonesterified , Fructose/adverse effects , Glucose/metabolism , Glucose Transporter Type 4 , Glucose-6-Phosphatase/metabolism , Glucose-6-Phosphatase/pharmacology , Glycerophospholipids , Hormones , Metabolic Syndrome/drug therapy , Metabolic Syndrome/etiology , Mice , Phosphoenolpyruvate/pharmacology , Polysaccharides/pharmacology , RNA, Messenger/metabolism , RNA, Ribosomal, 16S , Reishi/genetics , Steroids/pharmacology , Sterol Regulatory Element Binding Protein 1/metabolism , Triglycerides , alpha-Linolenic Acid/pharmacology
7.
J Med Food ; 25(6): 652-659, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35708629

ABSTRACT

Cholestatic liver disease, or cholestasis, is a condition characterized by liver inflammation and fibrosis following a bile duct obstruction and an intrahepatic accumulation of bile acids. Inhibiting inflammation is a promising therapeutic strategy for cholestatic liver diseases. Acer tegmentosum Maxim extract (ATE) is best known for its anti-inflammatory and antioxidative properties. In this study, we investigated the effects of ATE on liver injury and fibrosis in mice with bile duct ligation (BDL)-induced cholestasis through analysis of gene expression, cytokines, and histological examination. Oral administration of ATE (20 or 50 mg/kg) for 14 days significantly attenuated hepatocellular necrosis compared to vehicle-treated BDL mice, which was accompanied by the reduced level of serum bile acids and bilirubin. We determined that ATE treatment reduced liver inflammation, oxidative stress, and fibrosis. These beneficial effects of ATE were concurrent with the decreased expression of genes involved in the NF-κB pathway, suggesting that the anti-inflammatory effect of ATE could be a possible mechanism against cholestasis-associated liver injury. Our findings substantiate ATE's role as an alternative therapeutic agent for cholestasis-induced liver injury and fibrosis.


Subject(s)
Acer , Cholestasis , Hepatitis , Animals , Anti-Inflammatory Agents/metabolism , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Bile Acids and Salts/metabolism , Bile Acids and Salts/pharmacology , Bile Acids and Salts/therapeutic use , Bile Ducts/metabolism , Bile Ducts/surgery , Cholestasis/drug therapy , Cholestasis/metabolism , Cholestasis/pathology , Fibrosis , Hepatitis/complications , Hepatitis/drug therapy , Hepatitis/pathology , Inflammation/drug therapy , Liver/metabolism , Liver Cirrhosis/drug therapy , Liver Cirrhosis/etiology , Mice , Plant Extracts/pharmacology
8.
J Med Food ; 25(4): 435-442, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35438551

ABSTRACT

Although multiple studies have shown that Angelica keiskei of the Umbelliferae family has potent anti-inflammatory and antioxidative activities and that it reduces the serum bile acids in humans, whether A. keiskei has protective effects against cholestasis-induced liver injury remains unexplored until now. This study tests the hypothesis that Angelica keiskei root extract (AKE) alleviates liver injury, inflammation, and fibrosis in mouse models of acute cholestasis induced by bile duct ligation (BDL). Oral administration of AKE (200 or 500 mg/kg) attenuated hepatocellular necrosis and significantly reduced serum levels of bile acids and bilirubin in BDL mice. The critical enzyme of bile acid synthesis, CYP7A1, was repressed by AKE, suggesting that reduced bile acid production may contribute to liver protection. Moreover, we determined through gene expression and cytokine analysis and histological examination that AKE treatment decreased liver inflammation, oxidative stress, and fibrosis. AKE also suppressed the NF-κB pathway, suggesting this as a possible mediator of its anti-inflammatory effect. Our findings substantiate that AKE may be promising for treating cholestatic liver diseases in the future.


Subject(s)
Angelica , Chemical and Drug Induced Liver Injury, Chronic , Cholestasis , Animals , Anti-Inflammatory Agents/metabolism , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Bile Acids and Salts/metabolism , Bile Acids and Salts/pharmacology , Bile Acids and Salts/therapeutic use , Bile Ducts/metabolism , Bile Ducts/surgery , Cholestasis/drug therapy , Cholestasis/metabolism , Cholestasis/pathology , Fibrosis , Inflammation/metabolism , Liver/metabolism , Mice , Plant Extracts/therapeutic use
9.
Curr Microbiol ; 79(5): 147, 2022 Apr 09.
Article in English | MEDLINE | ID: mdl-35397017

ABSTRACT

Enterococcus faecalis (E. faecalis) belongs to lactic acid bacteria which can be used as a probiotic additive and feed, bringing practical value to the health of humans and animals. The prebiotic function of tea polyphenols lays a foundation for green tea polyphenols (GTP) to repair the adverse changes of E. faecalis under stress conditions. In this study, RNA-sequence analysis was used to explore the protective effect of GTP on E. faecalis under bile salt stress. A total of 50 genes were found to respond to GTP under bile salts stress, containing 18 up-regulated and 32 down-regulated genes. The results showed that multiple genes associated with cell wall and membrane, transmembrane transport, nucleotide transport and metabolism were significantly differentially expressed (P < 0.05), while GTP intervention can partly alleviate the detrimental effects of bile salt on amino acid metabolism and transport. The present study provides the whole genome transcriptomics of E. faecalis under bile salt stress and GTP intervention which help us understand the growth and mechanism of continuous adaptation of E. faecalis under stress conditions.


Subject(s)
Enterococcus faecalis , Polyphenols , Animals , Antioxidants/pharmacology , Bile , Bile Acids and Salts/metabolism , Bile Acids and Salts/pharmacology , Enterococcus faecalis/genetics , Guanosine Triphosphate/metabolism , Guanosine Triphosphate/pharmacology , Polyphenols/pharmacology , RNA-Seq , Salt Stress , Tea/chemistry , Transcriptome
10.
J Bioenerg Biomembr ; 54(1): 45-57, 2022 02.
Article in English | MEDLINE | ID: mdl-34718922

ABSTRACT

Gualou Xiebai Decoction (GXD), a classic prescription, is widely used to dealing with inflammatory diseases in China for thousands of years. Abnormal metabolic state of bile acids (BAs) is confirmed to cause intestinal epithelial barrier dysfunction. In preliminary work, we observed that GXD could decrease intestinal permeability in hyperlipidemia mice. The present study aimed to explore the protective effect of GXD on intestinal mucosa in vitro. Caco-2 cell monolayer permeability among different groups was determined by measuring the concentrations of FITC-dextran in the lower compartments and transepithelial electrical resistance (TEER). Meanwhile, mRNA and protein expressions of tight junctions (TJs) were investigated. Generation of intracellular reactive oxygen species (ROS) and the ratio of cell apoptosis induced by BAs were assessed by fluorescence probe and flow cytometry. GXD was shown to keep the cell monolayer in low permeable status, increase TEER and mRNA and protein expressions of occludin (Ocln) and zonula occluden 2 (ZO2) remarkably in cells challenged with cholic acid (CA), deoxycholic acid (DCA) and glycocholic acid (GCA). However, no significant effects were uncovered against the pathological effects of taurocholic acid (TCA). Meanwhile, generation of ROS and increased levels of apoptotic cells caused by CA, DCA and GCA were dramatically decreased by GXD, which were not observed on TCA. GXD could significantly attenuate intestinal barrier dysfunction induced by BAs via TJs regulation, oxidative stress suppression and cell apoptosis decrease, but such effects and behind mechanisms differed among different kinds of BAs.


Subject(s)
Bile Acids and Salts , Tight Junctions , Animals , Apoptosis , Bile Acids and Salts/metabolism , Bile Acids and Salts/pharmacology , Caco-2 Cells , Drugs, Chinese Herbal , Humans , Mice , Oxidative Stress , Permeability , Tight Junctions/metabolism
11.
J Antimicrob Chemother ; 76(10): 2569-2577, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34245280

ABSTRACT

BACKGROUND: Novel therapeutic strategies are urgently needed for Neisseria gonorrhoeae, given its increasing antimicrobial resistance. Treatment of oropharyngeal N. gonorrhoeae infections has proven particularly challenging, with most reported treatment failures of the first-line drug ceftriaxone occurring at this site and lower cure rates in recent trials of new antibiotics reported for oropharyngeal infections compared with other sites of infection. However, the accessibility of the oropharynx to topical therapeutics provides an opportunity for intervention. Local delivery of a therapeutic at a high concentration would enable the use of non-traditional antimicrobial candidates, including biological molecules that exploit underlying chemical sensitivities of N. gonorrhoeae but lack the potency or pharmacokinetic profiles required for effective systemic administration. METHODS: Two classes of molecules that are thought to limit gonococcal viability in vivo, bile acids and short- and medium-chain fatty acids, were examined for rapid bactericidal activity. RESULTS: The bile acids deoxycholic acid (DCA) and chenodeoxycholic acid (CDCA), but not other bile acid species, exerted extremely rapid bactericidal properties against N. gonorrhoeae, reducing viability more than 100 000-fold after 1 min. The short-chain fatty acids formic acid and hexanoic acid shared this rapid bactericidal activity. All four molecules are effective against a phylogenetically diverse panel of N. gonorrhoeae strains, including clinical isolates with upregulated efflux pumps and resistance alleles to the most widely used classes of existing antimicrobials. DCA and CDCA are both approved therapeutics for non-infectious indications and are well-tolerated by cultured epithelial cells. CONCLUSIONS: DCA and CDCA are attractive candidates for further development as anti-gonococcal agents.


Subject(s)
Gonorrhea , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Bile Acids and Salts/pharmacology , Drug Resistance, Bacterial , Fatty Acids/pharmacology , Gonorrhea/drug therapy , Humans , Microbial Sensitivity Tests , Neisseria gonorrhoeae
12.
Fish Shellfish Immunol ; 116: 52-60, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34216786

ABSTRACT

The aim of this study was to investigate the effects of dietary bile acids (BAs) on intestinal healthy status of tongue sole in terms of immunity, antioxidant status, digestive ability, mucosal barrier-related genes expression and microbiota. Three experimental diets were prepared with BA levels at 0 mg/kg (CT), 300 mg/kg (BA1) and 900 mg/kg (BA2) in a commercial basal diet. Each diet was fed to three replicates with 120 fish (10.87 ± 0.32 g) in each tank. After an 8-week feeding trial, growth parameters were significantly enhanced in both BAs supplementary groups (P < 0.05), and compared with CT group, survival rate in BA2 group was significantly improved (P < 0.05). Intestinal lysozyme activity and contents of immunoglobulin M and complement 3 were significantly increased in both BAs supplementary groups (P < 0.05), suggesting an enhancement effect on the non-specific immune response. BAs inclusion also significantly improved intestinal antioxidant capabilities by increasing antioxidase activities and decreasing malondialdehyde levels. In addition, compared with CT group, intestinal digestive ability was substantially enhanced as indicated by the significantly increased lipase activity in BA2 group (P < 0.05) and significantly increased amylase activity in BA1 and BA2 groups (P < 0.05). Coincidentally, BAs inclusion significantly upregulated the relative expression of intestinal mucosal barrier-related genes (P < 0.05). Further, dietary BAs distinctly remodeled intestinal microbiota by decreased the abundance of some potential pathogenic bacteria. In conclusion, dietary BAs supplementation is an effective way to improve the intestinal healthy status of tongue sole.


Subject(s)
Bile Acids and Salts/pharmacology , Dietary Supplements , Flatfishes , Gastrointestinal Microbiome/drug effects , Intestinal Mucosa/drug effects , Alkaline Phosphatase/immunology , Amylases/metabolism , Animals , Complement C3/immunology , Diet/veterinary , Fish Proteins/metabolism , Flatfishes/genetics , Flatfishes/immunology , Flatfishes/metabolism , Flatfishes/microbiology , Gene Expression Regulation/drug effects , Immunoglobulin M/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Lipase/metabolism , Muramidase/immunology , Oxidoreductases/metabolism , Peptide Hydrolases/metabolism , Tight Junction Proteins/genetics
13.
J Microbiol Biotechnol ; 31(6): 840-846, 2021 Jun 28.
Article in English | MEDLINE | ID: mdl-33958508

ABSTRACT

To improve the bile salt and acid tolerance of probiotics against gastrointestinal stresses, we investigated the effects of soybean lecithin and whey protein concentrate (WPC) 80 on the bile salt tolerance of Lacticaseibacillus paracasei L9 using a single-factor methodology, which was optimized using response surface methodology (RSM). The survival rate of L. paracasei L9 treated with 0.3% (w/v) bile salt for 2.5 h, and combined with soybean lecithin or WPC 80, was lower than 1%. After optimization, the survival rate of L. paracasei L9 incubated in 0.3% bile salt for 2.5 h reached 52.5% at a ratio of 0.74% soybean lecithin and 2.54% WPC 80. Moreover, this optimized method improved the survival rate of L. paracasei L9 in low pH condition and can be applied to other lactic acid bacteria (LAB) strains. Conclusively, the combination of soybean lecithin and WPC 80 significantly improved the bile salt and acid tolerance of LAB. Our study provides a novel approach for enhancing the gastrointestinal tolerance of LAB by combining food-derived components that have different properties.


Subject(s)
Adaptation, Physiological/drug effects , Bile Acids and Salts/metabolism , Lecithins/pharmacology , Probiotics/metabolism , Whey Proteins/pharmacology , Bile Acids and Salts/pharmacology , Lactobacillaceae/drug effects , Lactobacillaceae/metabolism , Models, Statistical , Glycine max/chemistry
14.
Front Immunol ; 12: 609644, 2021.
Article in English | MEDLINE | ID: mdl-34017324

ABSTRACT

Bacterial therapeutics are the emergent alternatives in treating autoimmune diseases such as Rheumatoid Arthritis [RA]. P. histicola MCI 001 is one such therapeutic bacterium that has been proven to treat autoimmune diseases such as RA and multiple sclerosis [MS] in animal models. The present study characterized P. histicola MCI 001 isolated from a human duodenal biopsy, and evaluated its impact on the gut microbial and metabolic profile in a longitudinal study using the collagen-induced arthritis model in HLA-DQ8.AEo transgenic mice. P. histicola MCI 001 though closely related to the type strain of P. histicola, DSM 19854, differed in utilizing glycerol. In culture, P. histicola MCI 001 produced vitamins such as biotin and folate, and was involved in digesting complex carbohydrates and production of acetate. Colonization study showed that duodenum was the predominant niche for the gavaged MCI 001. A longitudinal follow-up of gut microbial profile in arthritic mice treated with MCI 001 suggested that dysbiosis caused due to arthritis was partially restored to the profile of naïve mice after treatment. A taxon-level analysis suggested an expansion of intestinal genus Allobaculum in MCI001 treated arthritic mice. Eubiosis achieved post treatment with P. histicola MCI 001 was also reflected in the increased production of short-chain fatty acids [SCFAs]. Present study suggests that the treatment with P. histicola MCI 001 leads to an expansion of Allobaculum by increasing the availability of simple carbohydrates and acetate. Restoration of microbial profile and metabolites like butyrate induce immune and gut homeostasis.


Subject(s)
Biological Therapy/methods , Butyrates/metabolism , Prevotella/physiology , Symbiosis , Adaptation, Physiological , Animals , Arthritis, Rheumatoid/diagnosis , Arthritis, Rheumatoid/etiology , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/therapy , Bile Acids and Salts/pharmacology , Disease Models, Animal , Fatty Acids, Volatile/metabolism , Gastric Juice , Gastrointestinal Microbiome , Humans , Hydrogen-Ion Concentration , Mice , Mice, Transgenic , Prevotella/classification , Prevotella/drug effects , Prevotella/genetics
15.
J Nutr ; 151(1): 20-29, 2021 01 04.
Article in English | MEDLINE | ID: mdl-33245135

ABSTRACT

BACKGROUND: Whether dietary choline and bile acids affect lipid use via gut microbiota is unclear. OBJECTIVES: This study aimed to investigate the effect of choline and bile acids on growth performance, lipid use, intestinal immunology, gut microbiota, and bacterial metabolites in weaned piglets. METHODS: A total of 128 weaned piglets [Duroc × (Landrace × Yorkshire), 21-d-old, 8.21 ± 0.20 kg body weight (BW)] were randomly allocated to 4 treatments (8 replicate pens per treatment, each pen containing 2 males and 2 females; n = 32 per treatment) for 28 d. Piglets were fed a control diet (CON) or the CON diet supplemented with 597 mg choline/kg (C), 500 mg bile acids/kg (BA) or both (C + BA) in a 2 × 2 factorial design. Growth performance, intestinal function, gut microbiota, and metabolites were determined. RESULTS: Compared with diets without choline, choline supplementation increased BW gain (6.13%), average daily gain (9.45%), gain per feed (8.18%), jejunal lipase activity (60.2%), and duodenal IL10 gene expression (51%), and decreased the mRNA abundance of duodenal TNFA (TNFα) (40.7%) and jejunal toll-like receptor 4 (32.9%) (P < 0.05); additionally, choline increased colonic butyrate (29.1%) and the abundance of Lactobacillus (42.3%), while decreasing the bile acid profile (55.8% to 57.6%) and the abundance of Parabacteroides (75.8%), Bacteroides (80.7%), and unidentified-Ruminococcaceae (32.5%) (P ≤ 0.05). Compared with diets without BA, BA supplementation decreased the mRNA abundance of colonic TNFA (37.4%), NF-κB p65 (42.4%), and myeloid differentiation factor 88 (42.5%) (P ≤ 0.01); BA also increased colonic butyrate (20.9%) and the abundance of Lactobacillus (39.7%) and Faecalibacterium (71.6%) and decreased that of Parabacteroides (67.7%) (P < 0.05). CONCLUSIONS: Choline supplementation improved growth performance and prevented gut inflammation in weaned piglets by altering gut microbiota and lipid metabolism. BA supplementation suppressed intestinal inflammation with no effect on growth performance, which was associated with changed gut microbiota and metabolites.


Subject(s)
Choline/administration & dosage , Gastrointestinal Microbiome/drug effects , Inflammation/veterinary , Intestinal Diseases/veterinary , Lipid Metabolism/drug effects , Swine/growth & development , Animals , Bile Acids and Salts/administration & dosage , Bile Acids and Salts/pharmacology , Cytokines/genetics , Cytokines/metabolism , Dietary Supplements , Female , Gene Expression Regulation/drug effects , Intestinal Diseases/prevention & control , Male , Receptors, Cytoplasmic and Nuclear/metabolism , Swine Diseases/prevention & control , Transcriptome
16.
J Sep Sci ; 43(23): 4305-4313, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33001559

ABSTRACT

Qishen granules, derived from clinical experience formula, has been widely used to improve and treat myocardial ischemic chronic heart failure in China. However, the mechanism of action of Qishen granules in the treatment of chronic heart failure is unclear. This study aimed to discover potential biomarkers of isoproterenol-induced chronic heart failure rats and investigate the potential mechanism of Qishen granules treatment of chronic heart failure. The fecal metabolomics method based on ultra-high-performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry was used to analyze the therapeutic effect and metabolic changes of Qishen granules on chronic heart failure rats. Totally, 17 potential biomarkers were identified, involving bile acid metabolism, fatty acid metabolism, inflammatory response, and amino acid metabolism. For bile acid metabolism, we selected 12 bile acids (two of which were potential biomarkers in nontargeted metabolomics) for quantitative analysis. The quantitative results of bile acids showed that after Qishen granules treatment, the contents of bile acids such as ursodeoxycholic acid and glycodeoxycholic acid were similar to those of health group. This study helps to understand the pathogenesis of isoproterenol-induced chronic heart failure and the therapeutic mechanism of Qishen granules from the perspective of metabolic pathways.


Subject(s)
Bile Acids and Salts/pharmacology , Cardiotonic Agents/pharmacology , Drugs, Chinese Herbal/pharmacology , Heart Failure/drug therapy , Isoproterenol/antagonists & inhibitors , Metabolomics , Animals , Bile Acids and Salts/analysis , Bile Acids and Salts/metabolism , Cardiotonic Agents/analysis , Cardiotonic Agents/metabolism , Chromatography, High Pressure Liquid , Chronic Disease , Drugs, Chinese Herbal/analysis , Drugs, Chinese Herbal/metabolism , Gastrointestinal Microbiome , Heart Failure/chemically induced , Male , Mass Spectrometry , Rats , Rats, Sprague-Dawley
17.
PLoS One ; 15(1): e0227751, 2020.
Article in English | MEDLINE | ID: mdl-31971960

ABSTRACT

One of the most promising applications of human pluripotent stem cells is their utilization for human-based pharmacological models. Despite the fact that membrane transporters expressed in the liver play pivotal role in various hepatic functions, thus far only little attention was devoted to the membrane transporter composition of the stem cell-derived liver models. In the present work, we have differentiated HUES9, a human embryonic stem cell line, toward the hepatic lineage, and monitored the expression levels of numerous differentiation marker and liver transporter genes with special focus on ABC transporters. In addition, the effect of bile acid treatment and polarizing culturing conditions on hepatic maturation has been assessed. We found that most transporter genes crucial for hepatic functions are markedly induced during hepatic differentiation; however, as regards the transporter composition the end-stage cells still exhibited dual, hepatocyte and cholangiocyte character. Although the bile acid treatment and sandwich culturing only slightly influenced the gene expressions, the stimulated cell polarization resulted in formation of bile canaliculi and proper localization of transporters. Our results point to the importance of membrane transporters in human stem cell-derived hepatic models and demonstrate the relevance of cell polarization in generation of applicable cellular models with correctly localized transporters. On the basis of our observations we suggest that conventional criteria for the evaluation of the quality of stem cell-derived hepatocyte-like cells ought to be augmented with additional elements, such as polarized and functional expression of hepatic transporters.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Drug Evaluation, Preclinical/methods , Hepatocytes/metabolism , Human Embryonic Stem Cells/metabolism , ATP-Binding Cassette Transporters/genetics , Bile Acids and Salts/pharmacology , Cell Culture Techniques/methods , Cell Differentiation , Cell Line , Gene Expression/drug effects , Gene Expression Profiling , Humans , Oligonucleotide Array Sequence Analysis
18.
Clin Nutr ; 39(3): 928-934, 2020 03.
Article in English | MEDLINE | ID: mdl-31000340

ABSTRACT

BACKGROUND & AIMS: Short bowel syndrome patients (SBS) receiving parenteral nutrition (PN) often have dyslipidaemia and can develop intestinal failure-associated liver disease (IFALD). These patients demonstrate increased cholesterol synthesis and hepatic lipogenesis. These lipid disturbances may be due to a decreased concentration of the bile acid pool or malabsorption. The aim of this pilot study was to evaluate the effect of bile acid administration on lipid synthesis in patients with SBS. METHODS: The 24 h fractional synthesis rate (FSR) of cholesterol and triglycerides was measured by the isotopic method (deuterated water) before and after 4 months of ursodeoxycholic acid (UDCA) treatment (20 mg/kg/day). Five short bowel patients (age: 53.4 ± 19.2 years) who had normal liver function and lipid plasmatic profiles received 1920 ± 300 ml of PN for 151 ± 74 days (mean PN energy intake was 27.0 ± 6.0 kcal/kg body weight, composed with 3.87 ± 1.38 g/kg of carbohydrate, 0.72 ± 0.25 g/kg of fat and 1.10 ± 0.23 g/kg of amino acids). Plasma metabolites, liver enzymes, 7-α-OH-cholesterol and steatosis levels were also evaluated before and after treatment. Student's t-tests were performed, and the results were expressed in means (±SD). RESULTS: After treatment, decreases in the absolute values of cholesterol synthesis (0.31 ± 0.12 mmol L-1 to 0.24 ± 0.11 mmol L-1; p < 0.05), FSR of cholesterol (31.6 ± 4.7% to 26.4 ± 4.7%; p = 0.06) and FSR of triglycerides (12.8 ± 5.8% to 9.2 ± 5.5%; p < 0.01) were observed. Cholesterol and alanine aminotransferase concentrations also decreased (ALT) (p < 0.05). The absolute values of triglyceride synthesis and triglyceride concentrations remained unchanged. CONCLUSIONS: In SBS patients, UDCA decreases the hepatic synthesis of triglycerides and cholesterol. These results suggest that UDCA could prevent the onset of the IFALD.


Subject(s)
Bile Acids and Salts/pharmacology , Dietary Supplements , Lipogenesis/drug effects , Short Bowel Syndrome/metabolism , Adult , Aged , Bile Acids and Salts/metabolism , Cholesterol/metabolism , Female , Humans , Male , Middle Aged , Pilot Projects , Triglycerides/metabolism , Young Adult
19.
Am J Physiol Endocrinol Metab ; 317(3): E494-E502, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31237451

ABSTRACT

Bile acids, glucagon-like peptide-1 (GLP-1), and fibroblast growth factor 19 (FGF19) play an important role in postprandial metabolism. In this study, we investigated the postprandial bile acid response in plasma and its relation to insulin, GLP-1, and FGF19. First, we investigated the postprandial response to 40-h fast. Then we administered glycine-conjugated deoxycholic acid (gDCA) with the meal. We performed two separate observational randomized crossover studies on healthy, lean men. In experiment 1: we tested 4-h mixed meal after an overnight fast and a 40-h fast. In experiment 2, we tested a 4-h mixed meal test with and without gDCA supplementation. Both studies measured postprandial glucose, insulin, bile acids, GLP-1, and FGF19. In experiment 1, 40 h of fasting induced insulin resistance and increased postprandial GLP-1 and FGF19 concentrations. After an overnight fast, we observed strong correlations between postprandial insulin and gDCA levels at specific time points. In experiment 2, administration of gDCA increased GLP-1 levels and lowered late postprandial glucose without effect on FGF19. Energy expenditure was not affected by gDCA administration. Unexpectedly, 40 h of fasting increased both GLP-1 and FGF19, where the former appeared bile acid independent and the latter bile acid dependent. Second, a single dose of gDCA increased postprandial GLP-1. Therefore, our data add complexity to the physiological regulation of the enterokines GLP-1 and FGF19 by bile acids.


Subject(s)
Bile Acids and Salts/pharmacology , Fasting/physiology , Fibroblast Growth Factors/biosynthesis , Glucagon-Like Peptide 1/biosynthesis , Bile Acids and Salts/blood , Blood Glucose , Cross-Over Studies , Deoxycholic Acid/pharmacology , Dietary Supplements , Energy Metabolism , Fibroblast Growth Factors/blood , Glucagon-Like Peptide 1/blood , Humans , Insulin/blood , Insulin Resistance , Male , Postprandial Period , Young Adult
20.
Folia Microbiol (Praha) ; 64(6): 735-750, 2019 Nov.
Article in English | MEDLINE | ID: mdl-30739237

ABSTRACT

As potential probiotic traits of human milk-isolated bacteria have increasingly been recognized, this study aimed to evaluate the probiotic properties of bacteriocin-producing Enterococcus faecium strains isolated from human milk and colostrum. Among 118 human milk- and colostrum-isolated lactic cocci, only 29 were identified as Enterococcus. Of these, only four Enterococcus faecium isolates exhibited bacteriocigenic activity against several pathogenic Gram-positive bacteria, including Listeria monocytogenes. These isolates exhibited high acid (up to pH 3.0) and bile tolerance (0.5% oxgall) in simulated gastrointestinal conditions, demonstrating their ability to survive through the upper gastrointestinal tract. All of the E. faecium strains were shown to be sensitive to most of the antibiotics including vancomycin, tetracycline, rifampicin, and erythromycin, while they were resistant to kanamycin and chloramphenicol. None of the strains showed any virulence (gelE, agg2, clyA, clyB, clyM) and antibiotic resistance genes (vanA, vanB, ermB, tetM, and aac(6')-le-aph(2″)-la). In addition, all the strains were able to assimilate cholesterol, ranging between 25.2-64.1% and they exhibited variable adherence (19-36%) to Caco-2 cells. Based on the overall results of this in vitro study, four of the E. faecium strains isolated from human milk and colostrum can be considered as promising probiotic candidates; however, further in vivo evaluations are required.


Subject(s)
Bacteriocins/metabolism , Colostrum/microbiology , Enterococcus faecium/isolation & purification , Milk, Human/microbiology , Probiotics , Anti-Bacterial Agents/pharmacology , Antibiosis , Bacterial Adhesion , Bile Acids and Salts/pharmacology , Caco-2 Cells , Cholesterol/metabolism , Drug Resistance, Bacterial , Enterococcus faecium/drug effects , Enterococcus faecium/genetics , Enterococcus faecium/metabolism , Gastric Juice , Humans , Listeria monocytogenes/physiology , Microbial Sensitivity Tests
SELECTION OF CITATIONS
SEARCH DETAIL