Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 120(30): e2302697120, 2023 07 25.
Article in English | MEDLINE | ID: mdl-37467267

ABSTRACT

Autoreactive encephalitogenic T cells exist in the healthy immune repertoire but need a trigger to induce CNS inflammation. The underlying mechanisms remain elusive, whereby microbiota were shown to be involved in the manifestation of CNS autoimmunity. Here, we used intravital imaging to explore how microbiota affect the T cells as trigger of CNS inflammation. Encephalitogenic CD4+ T cells transduced with the calcium-sensing protein Twitch-2B showed calcium signaling with higher frequency than polyclonal T cells in the small intestinal lamina propria (LP) but not in Peyer's patches. Interestingly, nonencephalitogenic T cells specific for OVA and LCMV also showed calcium signaling in the LP, indicating a general stimulating effect of microbiota. The observed calcium signaling was microbiota and MHC class II dependent as it was significantly reduced in germfree animals and after administration of anti-MHC class II antibody, respectively. As a consequence of T cell stimulation in the small intestine, the encephalitogenic T cells start expressing Th17-axis genes. Finally, we show the migration of CD4+ T cells from the small intestine into the CNS. In summary, our direct in vivo visualization revealed that microbiota induced T cell activation in the LP, which directed T cells to adopt a Th17-like phenotype as a trigger of CNS inflammation.


Subject(s)
Intestinal Mucosa , Intestine, Small , Animals , Duodenum , Inflammation , Ileum
2.
Probiotics Antimicrob Proteins ; 15(3): 451-459, 2023 06.
Article in English | MEDLINE | ID: mdl-34647241

ABSTRACT

The functional characteristics of Lactobacillus johnsonii BFE6154, first isolated from Maasai traditional fermented milk, were previously identified in vitro, but its cholesterol-lowering properties have not been verified yet. In this study, we investigated the effect of L. johnsonii BFE6154 on cholesterol regulation and the mode of action. Stimulation of Caco-2 intestinal epithelial cells with L. johnsonii BFE6154 downregulated the gene expression of Niemann-Pick C1-like 1 (NPC1L1) through the activation of liver X receptor (LXR). Also, stimulation of HepG2 cells with the metabolites produced by L. johnsonii BFE6154 revealed an increase in the gene expression of low-density lipoprotein receptor (LDLR). Oral administration of L. johnsonii BFE6154 in mice receiving a high-fat and high-cholesterol diet (HFHCD), reduced total cholesterol and low-density lipoprotein-cholesterol (LDL) and increased high-density lipoprotein-cholesterol (HDL) in the blood, compared to the control. Diet-induced hypercholesterolemic mice receiving L. johnsonii BFE6154 showed a suppression of cholesterol absorption under the control of NPC1L1 in the intestine. Furthermore, L. johnsonii BFE6154 consumption ameliorated the hepatic cholesterol level and LDLR expression, which was reduced by HFHCD. These molecular modulations led to the increase of cholesterol excretion and the decrease of cholesterol levels in the feces and liver, respectively. Taken together, these results suggest that L. johnsonii BFE6154 may protect against diet-induced hypercholesterolemia through the regulation of cholesterol metabolism in the intestine and liver.


Subject(s)
Hypercholesterolemia , Lactobacillus johnsonii , Humans , Mice , Animals , Hypercholesterolemia/etiology , Hypercholesterolemia/therapy , Caco-2 Cells , Membrane Transport Proteins/metabolism , Cholesterol , Diet , Cholesterol, LDL/metabolism
3.
J Appl Microbiol ; 132(1): 562-570, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34133840

ABSTRACT

AIMS: Two putative probiotic strains, Lacticaseibacillus (Lc.) rhamnosus BFE5264 and Lactiplantibacillus (Lp.) plantarum NR74, have been shown to suppress cholesterol uptake and promote cholesterol efflux in Caco-2 cells. However, an in vivo beneficial effect of these strains on plasma cholesterol levels has not been verified yet; neither have the underlying mechanisms of regulating cholesterol metabolism clarified thus far. This study has focused on these two aspects. METHODS AND RESULTS: A murine model has been used, and the animals receiving a high-fat/high-cholesterol diet showed elevated plasma cholesterol levels. However, supplementation of Lc. rhamnosus BFE5264 and Lp. plantarum NR74 resulted in the down regulation of Niemann-Pick C1-like 1 (NPC1L1) in the intestine in addition to counteracting the diet-induced suppression of low-density lipoprotein receptor expression in the liver. ATP Binding Cassette Subfamily A Member 1 (ABCA1) was only significantly increased upon administration of Lc. rhamnosus BFE5264. CONCLUSIONS: The present findings demonstrate that supplementation with Lc. rhamnosus BFE5264 and Lp. plantarum NR74 may improve diet-induced hypercholesterolemia by suppression of cholesterol absorption in the small intestine and by supporting the regulation of cholesterol metabolism in the liver. SIGNIFICANCE AND IMPACT OF THE STUDY: This work contributes to understanding the beneficial effects of probiotics on host cholesterol metabolism and underlying mechanisms related to hypercholesterolemia.


Subject(s)
Hypercholesterolemia , Probiotics , Animals , Caco-2 Cells , Cholesterol/metabolism , Diet , Humans , Hypercholesterolemia/metabolism , Intestinal Absorption , Intestines , Liver/metabolism , Membrane Transport Proteins/metabolism , Mice
4.
Biomolecules ; 9(4)2019 03 28.
Article in English | MEDLINE | ID: mdl-30925749

ABSTRACT

Short chain fatty acids (SCFAs) are generated by the degradation and fermentation of complex carbohydrates, (i.e., dietary fiber) by the gut microbiota relevant for microbe⁻host communication. Here, we present a method for the quantification of SCFAs in fecal samples by liquid chromatography tandem mass spectrometry (LC-MS/MS) upon derivatization to 3-nitrophenylhydrazones (3NPH). The method includes acetate, propionate, butyrate, and isobutyrate with a run time of 4 min. The reproducible (coefficients of variation (CV) below 10%) quantification of SCFAs in human fecal samples was achieved by the application of stable isotope labelled internal standards. The specificity was demonstrated by the introduction of a quantifier and qualifier ions. The method was applied to investigate the pre-analytic stability of SCFAs in human feces. Concentrations of SCFA may change substantially within hours; the degree and kinetics of these changes revealed huge differences between the donors. The fecal SCFA level could be preserved by the addition of organic solvents like isopropanol. An analysis of the colon content of mice either treated with antibiotics or fed with a diet containing a non-degradable and -fermentable fiber source showed decreased SCFA concentrations. In summary, this fast and reproducible method for the quantification of SCFA in fecal samples provides a valuable tool for both basic research and large-scale studies.


Subject(s)
Fatty Acids, Volatile/analysis , Animals , Chromatography, Liquid , Humans , Mice , Mice, Inbred C57BL , Quality Control , Tandem Mass Spectrometry
5.
Nat Commun ; 9(1): 3760, 2018 09 14.
Article in English | MEDLINE | ID: mdl-30218046

ABSTRACT

Interactions between the gut microbial ecosystem and host lipid homeostasis are highly relevant to host physiology and metabolic diseases. We present a comprehensive multi-omics view of the effect of intestinal microbial colonization on hepatic lipid metabolism, integrating transcriptomic, proteomic, phosphoproteomic, and lipidomic analyses of liver and plasma samples from germfree and specific pathogen-free mice. Microbes induce monounsaturated fatty acid generation by stearoyl-CoA desaturase 1 and polyunsaturated fatty acid elongation by fatty acid elongase 5, leading to significant alterations in glycerophospholipid acyl-chain profiles. A composite classification score calculated from the observed alterations in fatty acid profiles in germfree mice clearly differentiates antibiotic-treated mice from untreated controls with high sensitivity. Mechanistic investigations reveal that acetate originating from gut microbial degradation of dietary fiber serves as precursor for hepatic synthesis of C16 and C18 fatty acids and their related glycerophospholipid species that are also released into the circulation.


Subject(s)
Acetates/metabolism , Acetyltransferases/metabolism , Dietary Fiber/metabolism , Fatty Acids/metabolism , Gastrointestinal Microbiome/physiology , Liver/metabolism , Stearoyl-CoA Desaturase/metabolism , Animals , Fatty Acid Elongases , Fatty Acids, Monounsaturated/metabolism , Fatty Acids, Unsaturated/metabolism , Gene Expression Profiling , Germ-Free Life , Lipid Metabolism , Mice , Proteomics
6.
Cell Mol Gastroenterol Hepatol ; 6(3): 370-388.e3, 2018.
Article in English | MEDLINE | ID: mdl-30182050

ABSTRACT

Background & Aims: Antibiotic (ABx) therapy is associated with increased risk for Crohn's disease but underlying mechanisms are unknown. We observed high fecal serine protease activity (PA) to be a frequent side effect of ABx therapy. The aim of the present study was to unravel whether this rise in large intestinal PA may promote colitis development via detrimental effects on the large intestinal barrier. Methods: Transwell experiments were used to assess the impact of high PA in ABx-treated patients or vancomycin/metronidazole-treated mice on the epithelial barrier. Serine protease profiling was performed using liquid chromatography-mass spectrometry/mass spectrometry analysis. The impact of high large intestinal PA on the intestinal barrier in wild-type and interleukin (IL)10-/- mice and on colitis development in IL10-/- mice was investigated using vancomycin/metronidazole with or without oral serine protease inhibitor (AEBSF) treatment. Results: The ABx-induced, high large intestinal PA was caused by significantly increased levels of pancreatic proteases and impaired epithelial barrier integrity. In wild-type mice, the rise in PA caused a transient increase in intestinal permeability but did not affect susceptibility to chemically induced acute colitis. In IL10-/- mice, increased PA caused a consistent impairment of the intestinal barrier associated with inflammatory activation in the large intestinal tissue. In the long term, the vancomycin/metronidazole-induced lasting increase in PA aggravated colitis development in IL10-/- mice. Conclusions: High large intestinal PA is a frequent adverse effect of ABx therapy, which is detrimental to the large intestinal barrier and may contribute to the development of chronic intestinal inflammation in susceptible individuals.


Subject(s)
Anti-Bacterial Agents/adverse effects , Colitis/metabolism , Intestine, Large/enzymology , Serine Proteases/metabolism , Animals , Colitis/chemically induced , Dextran Sulfate/pharmacology , Disease Models, Animal , Feces/enzymology , Feces/microbiology , Humans , Intestine, Large/microbiology , Metronidazole/adverse effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Risk Factors , Sulfones/pharmacology , Vancomycin/adverse effects
7.
J Sci Food Agric ; 93(4): 781-7, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-22806829

ABSTRACT

BACKGROUND: The balance between the rate of cholesterol uptake/accumulation and the rate of cholesterol efflux is reflected in the amount of lipid accumulation in macrophages. Based upon the fact that liver X receptors (LXRs) play a role in cholesterol efflux, we studied the effects of probiotics on cholesterol efflux and anti-inflammatory action in macrophages. We confirmed changes in LXR expression by treatment of LXR-transfected CHO-K1 cells with lactic acid bacteria (LAB), and co-cultured THP-1 cells with LAB to investigate changes in cholesterol efflux and inflammation. RESULTS: The experiment with CHO-K1 cells showed upregulation of LXR-ß by LAB. Treatment of THP-1 cells with LAB promoted LXR expression in THP-1, which eventually led to significant upregulation of ABCA1 and ABCG1 expression. The treatment with live LAB also significantly promoted cholesterol efflux. LAB suppressed expression of interleukin (IL)-1ß and tumor necrosis factor (TNF)-α, which resulted from activation of LXR. CONCLUSION: Our study shows that Lactobacillus rhamnosus BFE5264 and Lactobacillus plantarum NR74 activated LXR and induced cholesterol efflux by promoting expression of ABCA1 and ABCG1. Both strains also suppressed proinflammatory cytokines including IL-1ß and TNF-α. This study could account for the observation that LAB may block foam cell formation by cholesterol efflux and immune modulation.


Subject(s)
Cholesterol/metabolism , Inflammation/prevention & control , Lacticaseibacillus rhamnosus , Lactobacillus plantarum , Macrophages/metabolism , Orphan Nuclear Receptors/metabolism , Probiotics/therapeutic use , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/metabolism , Biological Transport , Cell Line , Foam Cells/metabolism , Humans , Inflammation/metabolism , Interleukin-1beta/metabolism , Liver X Receptors , Neoplasm Proteins/metabolism , Species Specificity , Transfection , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation
8.
Int J Food Sci Nutr ; 64(1): 44-52, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22816655

ABSTRACT

Hypercholesterolaemia is a major risk factor related to atherosclerosis, and it may be influenced by our diet. This study addresses the impact of Lactobacillus rhamnosus BFE5264 (isolated from Maasai fermented milk) and Lactobacillus plantarum NR74 (from Korean kimchi) on the control of cholesterol absorption through down-regulation of Niemann-Pick C1-like 1 (NPC1L1) expression. Caco-2 enterocytes were treated with the live, heat-killed (HK) bacteria, bacterial cell wall extracts and metabolites; mRNA level and protein expression were measured. Caco-2 cells showed lower NPC1L1 expression in the presence of the live test strains than the control, elucidating down-regulation of cholesterol uptake, and were compared well with the positive control, L. rhamnosus GG. This effect was also observed with HK bacteria and cell wall fractions but not with their metabolites. The potential of some Lactobacillus strains associated with traditional fermented foods to suppress cholesterol uptake and promote its efflux in enterocytes has been suggested from these data.


Subject(s)
Cholesterol/metabolism , Diet , Hypercholesterolemia/metabolism , Lacticaseibacillus rhamnosus , Lactobacillus plantarum , Membrane Proteins/metabolism , Probiotics , Anticholesteremic Agents/therapeutic use , Caco-2 Cells , Cell Wall , Down-Regulation , Enterocytes/metabolism , Fermentation , Food Microbiology , Hot Temperature , Humans , Hypercholesterolemia/prevention & control , Intestinal Absorption , Lactobacillus plantarum/isolation & purification , Lactobacillus plantarum/metabolism , Lacticaseibacillus rhamnosus/isolation & purification , Lacticaseibacillus rhamnosus/metabolism , Membrane Transport Proteins , Probiotics/therapeutic use , RNA, Messenger/metabolism
9.
J Med Food ; 14(10): 1097-106, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21861722

ABSTRACT

The purpose of the current study was to determine the anti-obesity and anti-inflammatory effects of an extract of purple sweet potatoes (PSPs) on 3T3-L1 adipocytes. For this purpose, differentiated 3T3-L1 adipocytes were treated with a PSP extract at concentrations of 1,000, 2,000, and 3,000 µg/mL for 24 hours. Then, we measured the changes in the sizes of the adipocytes, the secretion of leptin, and the mRNA/protein expression of lipogenic, inflammatory, and lipolytic factors after the treatment with the PSP extract. The PSP extract diminished leptin secretion, indicating that growth of fat droplets was suppressed. The extract also suppressed the expression of mRNAs of lipogenic and inflammatory factors and promoted lipolytic action. The antioxidative activity of the PSP extract was also measured using three different in vitro methods: 1,1-diphenyl-2-picrylhydrazyl free radical scavenging activity, ferric reducing ability potential assay, and chelating activity of transition metal ions. Taken together, our study shows that PSP extract has antilipogenic, anti-inflammatory, and lipolytic effects on adipocytes and has radical scavenging and reducing activity.


Subject(s)
Adipocytes/drug effects , Anti-Obesity Agents/pharmacology , Antioxidants/pharmacology , Ipomoea batatas/chemistry , Plant Extracts/pharmacology , 3T3-L1 Cells , Adipogenesis/drug effects , Animals , Anthocyanins/isolation & purification , Anthocyanins/pharmacology , Anti-Inflammatory Agents/pharmacology , Biphenyl Compounds/metabolism , Blotting, Western , Cell Differentiation/drug effects , Inflammation/drug therapy , Leptin/metabolism , Lipolysis , Mice , Picrates/metabolism , Real-Time Polymerase Chain Reaction
10.
Int J Food Microbiol ; 145(1): 155-61, 2011 Jan 31.
Article in English | MEDLINE | ID: mdl-21215484

ABSTRACT

The objective of this study was to evaluate the functional properties of lactic acid bacteria (LAB) from kimchi, a traditional Korean fermented vegetable product generally consumed raw as a side-dish with practically every meal. Twelve mild acid producing facultatively heterofermentative Lactobacillus strains were selected for their potential as starter cultures for fermentation of kimchi, and evaluated for their functional properties. Eleven strains were identified as Lactobacillus sakei and one as Lactobacillus plantarum. The strains identified as L. sakei differed in some physiological features; of particular interest was the fact that 9 of these strains produced L(+) lactic acid from glucose in presence of acetate. All strains were able to survive gastrointestinal conditions simulating stomach and duodenum passage. In addition, they showed higher adherence to HT-29 cells than Lactobacillus rhamnosus GG, a commercial probiotic strain used worldwide. These strains also showed antimicrobial activity against a number of food-borne pathogens. Their ability to lower cholesterol was demonstrated by BSH (bile salt hydrolytic) activity, and cholesterol assimilation tests in vitro. The results suggest the probiotic potential of these strains for use in kimchi fermentation.


Subject(s)
Fermentation , Food Microbiology , Lactobacillus/isolation & purification , Probiotics , Acetic Acid/metabolism , Antibiosis , Bacterial Adhesion , Bile Acids and Salts/metabolism , Carbohydrate Metabolism , Cholesterol/metabolism , Duodenum/microbiology , Glucose/metabolism , HT29 Cells , Humans , Hydrogen-Ion Concentration , Hydrophobic and Hydrophilic Interactions , Lactic Acid/biosynthesis , Lactobacillus/metabolism , Lactobacillus/physiology , Stomach/microbiology , Vegetables/microbiology
11.
Probiotics Antimicrob Proteins ; 3(3-4): 194-203, 2011 Dec.
Article in English | MEDLINE | ID: mdl-26781680

ABSTRACT

The effect of two putative probiotic strains, Lactobacillus rhamnosus BFE5264 and Lactobacillus plantarum NR74, on the control of cholesterol efflux in enterocytes was assessed by focusing on the promotion of ATP-binding cassette sub-family G members 5 and 8 (ABCG5 and ABCG8). Differentiated Caco-2 enterocytes were treated with live bacteria, heat-killed bacteria, a bacterial cell wall fraction, and metabolites and were subjected to cholesterol uptake assay, mRNA analysis, and protein analyses. Following LXR-transfection by incubation with CHO-K1 cells in DNA-lipofectin added media, the luciferase assay was conducted for LXR analysis. Treatment of Caco-2 cells with L. rhamnosus BFE5264 (isolated from traditional fermented Maasai milk) and L. plantarum NR74 (isolated from Korean kimchi) resulted in the up-regulation of LXR, concomitantly with the elevated expression of ABCG5 and ABCG8. This was associated with the promotion of cholesterol efflux at significantly higher levels compared to the positive control strain L. rhamnosus GG (LGG). The experiment with CHO-K1 cells confirmed up-regulation of LXR-beta by the test strains, and treatment with the live L. rhamnosus BFE5264 and L. plantarum NR74 strains significantly increased cholesterol efflux. Heat-killed cells and cell wall fractions of both LAB strains induced the upregulation of ABCG5/8 through LXR activation. By contrast, LAB metabolites did not show any effect on ABCG5/8 and LXR expression. Data from this study suggest that LAB strains, such as L. rhamnosus BFE5264 and L. plantarum NR74, may promote cholesterol efflux in enterocytes, and thus potentially contribute to the prevention of hypercholesterolemia and atherosclerosis.

SELECTION OF CITATIONS
SEARCH DETAIL
...