Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24.662
Filter
1.
Nat Commun ; 15(1): 6618, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39103350

ABSTRACT

A mechanistic understanding of host-microbe interactions in the gut microbiome is hindered by poorly annotated bacterial genomes. While functional genomics can generate large gene-to-phenotype datasets to accelerate functional discovery, their applications to study gut anaerobes have been limited. For instance, most gain-of-function screens of gut-derived genes have been performed in Escherichia coli and assayed in a small number of conditions. To address these challenges, we develop Barcoded Overexpression BActerial shotgun library sequencing (Boba-seq). We demonstrate the power of this approach by assaying genes from diverse gut Bacteroidales overexpressed in Bacteroides thetaiotaomicron. From hundreds of experiments, we identify new functions and phenotypes for 29 genes important for carbohydrate metabolism or tolerance to antibiotics or bile salts. Highlights include the discovery of a D-glucosamine kinase, a raffinose transporter, and several routes that increase tolerance to ceftriaxone and bile salts through lipid biosynthesis. This approach can be readily applied to develop screens in other strains and additional phenotypic assays.


Subject(s)
Bile Acids and Salts , Carbon , Gastrointestinal Microbiome , Carbon/metabolism , Gastrointestinal Microbiome/genetics , Bile Acids and Salts/metabolism , Anti-Bacterial Agents/pharmacology , Stress, Physiological/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacteroides thetaiotaomicron/genetics , Bacteroides thetaiotaomicron/metabolism , Gene Expression Regulation, Bacterial , Bacteroidetes/genetics , Bacteroidetes/metabolism , Carbohydrate Metabolism/genetics , Humans , Genes, Bacterial/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Genome, Bacterial
2.
Commun Biol ; 7(1): 947, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39103440

ABSTRACT

Clostridium septicum infections are highly predictive of certain malignancies in human patients. To initiate infections, C. septicum spores must first germinate and regain vegetative growth. Yet, what triggers the germination of C. septicum spores is still unknown. Here, we observe that C. septicum germinates in response to specific bile salts. Putative bile salt recognition genes are identified in C. septicum based on their similarity in sequence and organization to bile salt-responsive csp genes in Clostridioides difficile. Inactivating two of these csp orthologs (cspC-82 and cspC-1718) results in mutant spores that no longer germinate in the presence of their respective cognate bile salts. Additionally, inactivating the putative cspBA or sleC genes in C. septicum abrogates the germination response to all bile salt germinants, suggesting that both act at a convergent point downstream of cspC-82 and cspC-1718. Molecular dynamics simulations show that both CspC-82 and CspC-1718 bear a strong structural congruence with C. difficile's CspC. The existence of functional bile salt germination sensors in C. septicum may be relevant to the association between infection and malignancy.


Subject(s)
Bacterial Proteins , Bile Acids and Salts , Clostridioides difficile , Clostridium septicum , Spores, Bacterial , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bile Acids and Salts/metabolism , Spores, Bacterial/genetics , Clostridioides difficile/genetics , Clostridium septicum/genetics , Molecular Dynamics Simulation , Gene Expression Regulation, Bacterial , Clostridium Infections/microbiology , Carrier Proteins
3.
J Transl Med ; 22(1): 746, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39113144

ABSTRACT

Acute graft-versus-host disease (aGVHD) is primarily driven by allogeneic donor T cells associated with an altered composition of the host gut microbiome and its metabolites. The severity of aGVHD after allogeneic hematopoietic stem cell transplantation (allo-HSCT) is not solely determined by the host and donor characteristics; however, the underlying mechanisms remain unclear. Using single-cell RNA sequencing, we decoded the immune cell atlas of 12 patients who underwent allo-HSCT: six with aGVHD and six with non-aGVHD. We performed a fecal microbiota (16SrRNA sequencing) analysis to investigate the fecal bacterial composition of 82 patients: 30 with aGVHD and 52 with non-aGVHD. Fecal samples from these patients were analyzed for bile acid metabolism. Through multi-omic analysis, we identified a feedback loop involving "immune cell-gut microbes-bile acid metabolites" contributing to heightened immune responses in patients with aGVHD. The dysbiosis of the gut microbiota and disruption of bile acid metabolism contributed to an exaggerated interleukin-1 mediated immune response. Our findings suggest that resistin and defensins are crucial in mitigating against aGVHD. Therefore, a comprehensive multi-omic atlas incorporating immune cells, gut microbes, and bile acid metabolites was developed in this study and used to propose novel, non-immunosuppressive approaches to prevent aGVHD.


Subject(s)
Bile Acids and Salts , Feces , Gastrointestinal Microbiome , Graft vs Host Disease , Bile Acids and Salts/metabolism , Humans , Graft vs Host Disease/immunology , Graft vs Host Disease/microbiology , Gastrointestinal Microbiome/immunology , Female , Male , Feces/microbiology , Middle Aged , Acute Disease , Adult , Feedback, Physiological , Immunity , Metabolomics , Hematopoietic Stem Cell Transplantation , Multiomics
4.
Cells ; 13(15)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39120326

ABSTRACT

Cholestatic liver diseases, including primary biliary cholangitis (PBC) and primary sclerosing cholangitis (PSC), result from an impairment of bile flow that leads to the hepatic retention of bile acids, causing liver injury. Until recently, the only approved treatments for PBC were ursodeoxycholic acid (UDCA) and obeticholic acid (OCA). While these therapies slow the progression of PBC in the early stage of the disease, approximately 40% of patients respond incompletely to UDCA, and advanced cases do not respond. UDCA does not improve survival in patients with PSC, and patients often have dose-limiting pruritus reactions to OCA. Left untreated, these diseases can progress to fibrosis and cirrhosis, resulting in liver failure and the need for transplantation. These shortcomings emphasize the urgent need for alternative treatment strategies. Recently, nuclear hormone receptors have been explored as pharmacological targets for adjunct therapy because they regulate enzymes involved in bile acid metabolism and detoxification. In particular, the peroxisome proliferator-activated receptor (PPAR) has emerged as a therapeutic target for patients with PBC or PSC who experience an incomplete response to UDCA. PPARα is predominantly expressed in the liver, and it plays an essential role in the regulation of cytochrome P450 (CYP) and uridine 5'-diphospho-glucuronosyltransferase (UGT) enzymes, both of which are critical enzyme families involved in the regulation of bile acid metabolism and glucuronidation, respectively. Importantly, PPARα agonists, e.g., fenofibrate, have shown therapeutic benefits in reducing elevated markers of cholestasis in patients with PBC and PSC, and elafibranor, the first PPAR (dual α, ß/δ) agonist, has been FDA-approved for the second-line treatment of PBC. Additionally, newer PPAR agonists that target various PPAR isoforms (ß/δ, γ) are under development as an adjunct therapy for PBC or PSC, although their impact on glucuronidation pathways are less characterized. This review will focus on PPAR-mediated bile acid glucuronidation as a therapeutic pathway to improve outcomes for patients with PBC and PSC.


Subject(s)
Bile Acids and Salts , Humans , Bile Acids and Salts/metabolism , Peroxisome Proliferator-Activated Receptors/metabolism , Peroxisome Proliferator-Activated Receptors/agonists , Cholestasis/metabolism , Cholestasis/drug therapy , Animals , Liver Cirrhosis, Biliary/metabolism , Liver Cirrhosis, Biliary/drug therapy , Cholangitis, Sclerosing/drug therapy , Cholangitis, Sclerosing/metabolism
5.
BMC Microbiol ; 24(1): 286, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39090543

ABSTRACT

BACKGROUND: Bile acids (BAs) are steroid-derived molecules with important roles in digestion, the maintenance of host metabolism, and immunomodulation. Primary BAs are synthesized by the host, while secondary BAs are produced by the gut microbiome through transformation of the former. The regulation of microbial production of secondary BAs is not well understood, particularly the production of 7-dehydroxylated BAs, which are the most potent agonists for host BA receptors. The 7-dehydroxylation of cholic acid (CA) is well established and is linked to the expression of a bile acid-inducible (bai) operon responsible for this process. However, little to no 7-dehydroxylation has been reported for other host-derived BAs (e.g., chenodeoxycholic acid, CDCA or ursodeoxycholic acid, UDCA). RESULTS: Here, we demonstrate that the 7-dehydroxylation of CDCA and UDCA by the human isolate Clostridium scindens is induced when CA is present, suggesting that CA-dependent transcriptional regulation is required for substantial 7-dehydroxylation of these primary BAs. This is supported by the finding that UDCA alone does not promote expression of bai genes. CDCA upregulates expression of the bai genes but the expression is greater when CA is present. In contrast, the murine isolate Extibacter muris exhibits a distinct response; CA did not induce significant 7-dehydroxylation of primary BAs, whereas BA 7-dehydroxylation was promoted upon addition of germ-free mouse cecal content in vitro. However, E. muris was found to 7-dehydroxylate in vivo. CONCLUSIONS: The distinct expression responses amongst strains indicate that bai genes are regulated differently. CA promoted bai operon gene expression and the 7-dehydroxylating activity in C. scindens strains. Conversely, the in vitro activity of E. muris was promoted only after the addition of cecal content and the isolate did not alter bai gene expression in response to CA. The accessory gene baiJ was only upregulated in the C. scindens ATCC 35704 strain, implying mechanistic differences amongst isolates. Interestingly, the human-derived C. scindens strains were also capable of 7-dehydroxylating murine bile acids (muricholic acids) to a limited extent. This study shows novel 7-dehydroxylation activity in vitro resulting from the presence of CA and suggests distinct bai gene expression across bacterial species.


Subject(s)
Bile Acids and Salts , Cholic Acid , Cholic Acid/metabolism , Animals , Bile Acids and Salts/metabolism , Mice , Humans , Clostridium/metabolism , Clostridium/genetics , Gene Expression Regulation, Bacterial , Hydroxylation , Operon , Chenodeoxycholic Acid/metabolism , Ursodeoxycholic Acid/metabolism , Gastrointestinal Microbiome
6.
Cell Host Microbe ; 32(8): 1444-1454.e6, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39146801

ABSTRACT

Pregnant women undergoing a cesarean section (CS) typically receive antibiotics prior to skin incision to prevent infections. To investigate if the timing of antibiotics influences the infant gut microbiome, we conducted a randomized controlled trial (NCT06030713) in women delivering via a scheduled CS who received antibiotics either before skin incision or after umbilical cord clamping. We performed a longitudinal analysis on 172 samples from 28 infants at 8 post-birth time points and a cross-sectional analysis at 1 month in 79 infants from 3 cohorts. Although no significant associations with bacterial composition, metabolic pathways, short-chain fatty acids, and bile acids were found, we observed subtle differences between the groups at the bacterial strain level and in the load of antibiotic resistance genes. Rather, feeding mode was a predominant and defining factor impacting infant microbial composition. In conclusion, antibiotic administration during CS has only limited effects on the early-life gut microbiome.


Subject(s)
Anti-Bacterial Agents , Antibiotic Prophylaxis , Cesarean Section , Gastrointestinal Microbiome , Humans , Gastrointestinal Microbiome/drug effects , Female , Antibiotic Prophylaxis/methods , Pregnancy , Anti-Bacterial Agents/administration & dosage , Infant , Infant, Newborn , Adult , Cross-Sectional Studies , Bacteria/genetics , Bacteria/drug effects , Bacteria/classification , Longitudinal Studies , Bile Acids and Salts/metabolism , Feces/microbiology
7.
Clin Chim Acta ; 562: 119896, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39098629

ABSTRACT

BACKGROUND: Elevated maternal serum total bile acids (sTBA) level during pregnancy was associated with adverse fetal outcomes. Women with elevated sTBA could complicate with hepatic dysfunction or vascular disorders (hypertensive disorders of pregnancy, HDP), which aggravated adverse fetal outcomes. However, the relationships among sTBA level, hepatic dysfunction, HDP and adverse fetal outcomes were still illusive. OBJECTIVE: We aimed to explore whether hepatic dysfunction or vascular disorders (HDP) mediated the associations between elevated sTBA level and adverse fetal outcomes. METHODS: A large retrospective cohort study encompassing 117,789 Chinese pregnant women with singleton delivery between Jan 2014 and Dec 2022 was conducted. Causal mediation analysis was applied to assess the mediating role of hepatic dysfunction (alanine transaminase > 40 U/L) or HDP in explaining the relationship between high maternal sTBA level (≥10 µmol/L) and adverse fetal outcomes, including low birth weight (LBW), small for gestational age (SGA), and preterm birth (PTB). RESULTS: sTBA level were positively associated with LBW (adjusted odds ratio (aOR) = 1.40; [95 % confidence interval (CI): 1.24-1.59]), SGA (aOR=1.31; [95 % CI: 1.18-1.46]), and PTB (aOR=1.27; [95 % CI: 1.15-1.41]), respectively. The estimated proportions of the total associations mediated by HDP were 47 % [95 % CI: 31 %-63 %] for LBW, 24 % [95 % CI: 13 %-35 %] for SGA, and 34 % [95 % CI: 19 %-49 %] for PTB, excepting the direct effects of high sTBA level. The contribution of hepatic dysfunction as a mediator was weaker on the association between high sTBA level on fetal outcomes, as the proportions mediated and 95 % CI were 16 % [4 %-29 %], 4 % [-6%-14 %], 32 % [15 %-50 %] for LBW, SGA, and PTB, respectively. Moreover, the mediating effect of hepatic dysfunction was nearly eliminated after excluding cases of HDP in the sensitivity analysis. CONCLUSIONS: The substantial mediating effects through HDP highlighted its significant role in adverse fetal outcomes associated with elevated sTBA level. The findings also provoked new insights into understanding the mechanism and developing clinical management strategies (i.e. vascular protection) for adverse fetal outcomes associated with elevated sTBA level.


Subject(s)
Bile Acids and Salts , Hypertension, Pregnancy-Induced , Pregnancy Outcome , Humans , Pregnancy , Female , Bile Acids and Salts/blood , Adult , China/epidemiology , Retrospective Studies , Hypertension, Pregnancy-Induced/blood , Hypertension, Pregnancy-Induced/epidemiology , Cohort Studies , Infant, Newborn , Premature Birth/blood
8.
Front Cell Infect Microbiol ; 14: 1408581, 2024.
Article in English | MEDLINE | ID: mdl-39119290

ABSTRACT

Background: Statins, being the primary pharmacological intervention for hypercholesterolemia, exhibit a notable degree of interpatient variability in their effectiveness, which may be associated with gut microbiota. This study sought to identify the biomarkers for evaluating differences in statin efficacy. Methods: A quasi case-control study was conducted among participants with hypercholesterolemia and coronary heart disease taking rosuvastatin essential. According to the level of low density lipoprotein cholesterol (LDL-C), participants was divided into the "Up to standard" (US) group and the "Below standard" (BS) group. 16S rDNA sequencing and untargeted metabolomics were applied to detected the information of gut microbiota and related metabolites. Results: A total of 8 US and 8 BS group matched by age and sex were included in the final analysis. 16S rDNA sequencing results indicated that the characteristic strains of the US group were f-Eubacterium_coprostanoligenes and g-Papillibacter, while the characteristic flora of the BS group were o-C0119, g-Pseudolabrys, s-Dyella-Marensis and f-Xanthobacaceae. Metabolomic results suggested that the levels of chenodeoxycholic acid-3-ß-D-glucuronide, 1-methylnicotinamide and acetoacetate in stool samples of the US group were significantly higher than those of the BS group. By identifying the differentially abundant bacterial taxa, the gut microbiota could modulate the efficacy of statins through producing enzymes involved in cholesterol metabolism. Conclusions: The findings suggest that the difference in statin efficacy may be related to gut microbiota strains that can produce short-chain fatty acids and secondary bile acids and affect the efficacy of statins by regulating the activities of cholesterol metabolite-related proteins. Metabolites related to short-chain fatty acids and secondary bile acids in the gut are expected to be biomarkers indicating the efficacy of statins.


Subject(s)
Coronary Disease , Gastrointestinal Microbiome , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Hypercholesterolemia , Aged , Female , Humans , Male , Middle Aged , Bacteria/metabolism , Bile Acids and Salts/metabolism , Biomarkers/blood , Case-Control Studies , China , Cholesterol, LDL/blood , Cholesterol, LDL/metabolism , Coronary Disease/microbiology , Coronary Disease/drug therapy , Coronary Disease/metabolism , East Asian People , Feces/microbiology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Hypercholesterolemia/drug therapy , Metabolomics , RNA, Ribosomal, 16S/genetics , Rosuvastatin Calcium/therapeutic use , Treatment Outcome
9.
Gut Microbes ; 16(1): 2383353, 2024.
Article in English | MEDLINE | ID: mdl-39105259

ABSTRACT

The role of the gut microbiota in the occurrence and progression of primary biliary cholangitis (PBC) is not fully understood. First, the fecal microbiota of patients with PBC (n = 4) (PBC-FMT) or healthy individuals (n = 3) (HC-FMT) was transplanted into pseudo germ-free mice or 2OA-BSA-induced PBC models. The functions, histology and transcriptome of the liver, and microbiota and metabolome of the feces were analyzed. Second, the liver transcriptomes of PBC patients (n = 7) and normal individuals (n = 7) were analyzed. Third, the liver transcriptomes of patients with other liver diseases were collected from online databases and compared with our human and mouse data. Our results showed that PBC-FMT increased the serum ALP concentration, total bile acid content, liver injury and number of disease-related pathways enriched with upregulated liver genes in pseudo germ-free mice and increased the serum glycylproline dipeptidyl aminopeptidase level and liver damage in a 2OA-BSA-induced PBC model. The gut microbiota and metabolome differed between PBC-FMT and HC-FMT mice and reflected those of their donors. PBC-FMT tended to upregulate hepatic immune and signal transduction pathways but downregulate metabolic pathways, as in some PBC patients. The hematopoietic cell lineage, Toll-like receptor, and PPAR signaling pathway were not affected in patients with alcoholic hepatitis, HBV, HCV, HCV cirrhosis, or NASH, indicating their potential roles in the gut microbiota affecting PBC. In conclusion, the altered gut microbiota of PBC patients plays an important role in the occurrence and progression of PBC. The improvement of the gut microbiota is worthy of in-depth research and promotion as a critical aspect of PBC prevention and treatment.


Subject(s)
Disease Models, Animal , Feces , Gastrointestinal Microbiome , Liver Cirrhosis, Biliary , Liver , Animals , Humans , Mice , Liver Cirrhosis, Biliary/microbiology , Liver Cirrhosis, Biliary/pathology , Liver Cirrhosis, Biliary/metabolism , Liver/pathology , Liver/metabolism , Liver/microbiology , Feces/microbiology , Female , Bacteria/classification , Bacteria/isolation & purification , Bacteria/genetics , Fecal Microbiota Transplantation , Male , Bile Acids and Salts/metabolism , Transcriptome , Mice, Inbred C57BL
10.
Nutrients ; 16(15)2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39125351

ABSTRACT

Syrian hamsters are valuable models for studying lipid metabolism due to their sensitivity to dietary cholesterol, yet the precise impact of varying cholesterol levels has not been comprehensively assessed. This study examined the impact of varying dietary cholesterol levels on lipid metabolism in Syrian hamsters. Diets ranging from 0% to 1% cholesterol were administered to assess lipid profiles and oxidative stress markers. Key findings indicate specific cholesterol thresholds for inducing distinct lipid profiles: below 0.13% for normal lipids, 0.97% for elevated LDL-C, 0.43% for increased VLDL-C, and above 0.85% for heightened hepatic lipid accumulation. A cholesterol supplementation of 0.43% induced hypercholesterolemia without adverse liver effects or abnormal lipoprotein expression. Furthermore, cholesterol supplementation significantly increased liver weight, plasma total cholesterol, LDL-C, and VLDL-C levels while reducing the HDL-C/LDL-C ratio. Fecal cholesterol excretion increased, with stable bile acid levels. High cholesterol diets correlated with elevated plasma ALT activities, reduced hepatic lipid peroxidation, and altered leptin and CETP levels. These findings underscore Syrian hamsters as robust models for hyperlipidemia research, offering insights into experimental methodologies. The identified cholesterol thresholds facilitate precise lipid profile manipulation, enhancing the hamster's utility in lipid metabolism studies and potentially informing clinical approaches to managing lipid disorders.


Subject(s)
Cholesterol, Dietary , Lipid Metabolism , Liver , Mesocricetus , Animals , Cholesterol, Dietary/administration & dosage , Liver/metabolism , Male , Cricetinae , Feces/chemistry , Oxidative Stress , Hypercholesterolemia/metabolism , Hypercholesterolemia/blood , Cholesterol, LDL/blood , Lipid Peroxidation , Cholesterol/blood , Cholesterol/metabolism , Bile Acids and Salts/metabolism , Leptin/blood , Leptin/metabolism , Cholesterol Ester Transfer Proteins/metabolism
11.
Nutrients ; 16(15)2024 Aug 03.
Article in English | MEDLINE | ID: mdl-39125429

ABSTRACT

BACKGROUND: Cholelithiasis is one of the more common complications following bariatric surgery. This may be related to the rapid weight loss during this period, although the exact mechanism of gallstone formation after bariatric surgery has not been fully elucidated. METHODS: The present literature review focuses on risk factors, prevention options and the impact of the gut microbiota on the development of gallbladder stones after bariatric surgery. RESULTS: A potential risk factor for the development of cholelithiasis after bariatric surgery may be changes in the composition of the intestinal microbiota and bile acids. One of the bile acids-ursodeoxycholic acid-is considered to reduce the concentration of mucin proteins and thus contribute to reducing the formation of cholesterol crystals in patients with cholelithiasis. Additionally, it reduces the risk of both asymptomatic and symptomatic gallstones after bariatric surgery. Patients who developed gallstones after bariatric surgery had a higher abundance of Ruminococcus gnavus and those who did not develop cholelithiasis had a higher abundance of Lactobacillaceae and Enterobacteriaceae. CONCLUSION: The exact mechanism of gallstone formation after bariatric surgery has not yet been clarified. Research suggests that the intestinal microbiota and bile acids may have an important role in this.


Subject(s)
Bariatric Surgery , Bile Acids and Salts , Cholelithiasis , Gastrointestinal Microbiome , Humans , Bariatric Surgery/adverse effects , Cholelithiasis/prevention & control , Cholelithiasis/etiology , Bile Acids and Salts/metabolism , Risk Factors , Gallstones/prevention & control , Gallstones/etiology , Ursodeoxycholic Acid
13.
BMC Pregnancy Childbirth ; 24(1): 525, 2024 Aug 10.
Article in English | MEDLINE | ID: mdl-39127651

ABSTRACT

BACKGROUND: The pregnant women with intrahepatic cholestasis were at high risk of fetal distress, preterm birth and unexpected stillbirth. Intrahepatic cholestasis of pregnancy (ICP) was mainly caused by disorder of bile acid metabolism, whereas the specific mechanism was obscure. METHODS: We performed proteomics analysis of 10 ICP specimens and 10 placenta specimens from patients without ICP through data-independent acquisition (DIA) technique to disclose differentially expressed proteins. We executed metabolomic analysis of 30 ICP specimens and 30 placenta specimens from patients without ICP through UPLC-MS/MS to identify differentially expressed metabolites. Enrichment and correlation analysis was used to obtain the direct molecular insights of ICP development. The ICP rat models were constructed to validate pathological features. RESULTS: The heatmap of proteomics analysis showed the top 30 up-regulated and 30 down-regulated proteins. The metabolomic analysis revealed 20 richer and 4 less abundant metabolites in ICP samples compared with placenta specimens from patients without ICP, and enrichment pathways by these metabolites included primary bile acid biosynthesis, cholesterol metabolism, bile secretion, nicotinate and nicotinamide metabolism, purine metabolism and metabolic pathways. Combined analysis of multiple omics results demonstrated that bile acids such as Glycohyocholic acid, Glycine deoxycholic acid, beta-Muricholic acid, Noncholic acid, cholic acid, Gamma-Mercholic Acid, alpha-Muricholic acid and Glycochenodeoxycholic Aicd were significantly associated with the expression of GLRX3, MYL1, MYH7, PGGT1B, ACTG1, SP3, LACTB2, C2CD5, APBB2, IPO9, MYH2, PPP3CC, PIN1, BLOC1S1, DNAJC7, RASAL2 and ATCN3 etc. The core protein ACAT2 was involved in lipid metabolic process and animal model showed that ACAT2 was up-regulated in placenta and liver of pregnant rats and fetal rats. The neonates had low birth weight and Safranin O-Fast green FCF staining of animal models showed that poor osteogenic and chondrogenic differentiation of fetal rats. CONCLUSION: Multiple metabolites-alpha-Muricholic acid, beta-Muricholic acid, Glycine deoxycholic acid and Glycochenodeoxycholic Acid etc. were perfect biomarkers to predict occurrence of ICP. Bile acids were significantly associated with varieties of protein expression and these proteins were differentially expressed in ICP samples. Our study provided several biomarkers for ICP detection and potential therapeutic targets for ICP development.


Subject(s)
Bile Acids and Salts , Biomarkers , Cholestasis, Intrahepatic , Metabolomics , Placenta , Pregnancy Complications , Proteomics , Female , Cholestasis, Intrahepatic/metabolism , Cholestasis, Intrahepatic/diagnosis , Humans , Pregnancy , Pregnancy Complications/metabolism , Pregnancy Complications/diagnosis , Biomarkers/metabolism , Biomarkers/analysis , Proteomics/methods , Bile Acids and Salts/metabolism , Rats , Placenta/metabolism , Animals , Metabolomics/methods , Adult , Disease Models, Animal , Tandem Mass Spectrometry
14.
Sci Rep ; 14(1): 18705, 2024 08 12.
Article in English | MEDLINE | ID: mdl-39134638

ABSTRACT

The incidence of duodenal tumors (DTs) is increasing. However, the mechanisms underlying its development remain unclear. Environmental factors, including the microbiome and bile acids (BAs), are believed to influence tumor development. Therefore, we conducted a single-center, prospective, observational study to investigate the potential differences between patients with DTs and healthy controls (HCs) based on these factors. In addition, the BAs in the duodenal fluid were measured using liquid chromatography-tandem mass spectrometry. We recruited 41 patients and performed 16S rRNA-seq. There was no difference in the observed ASVs or PCoA plot of Bray-Curtis dissimilarity between the DTs and HCs. The lithocholic acid concentration was significantly lower in the DT group than in the control group. The ratio of CDCA to LCA was significantly higher in patients with DTs. No significant differences in microbiota were observed between DTs and HCs. In patients with DTs, the lithocholic acid concentration in duodenal was significantly lower than in HCs.


Subject(s)
Bile Acids and Salts , Duodenal Neoplasms , Duodenum , Gastrointestinal Microbiome , RNA, Ribosomal, 16S , Humans , Male , Bile Acids and Salts/metabolism , Female , Prospective Studies , Middle Aged , Duodenal Neoplasms/microbiology , Duodenal Neoplasms/metabolism , Duodenum/metabolism , Duodenum/microbiology , Aged , RNA, Ribosomal, 16S/genetics , Adult , Lithocholic Acid/metabolism , Microbiota , Case-Control Studies
15.
Elife ; 122024 Aug 13.
Article in English | MEDLINE | ID: mdl-39137024

ABSTRACT

Hepatic factors secreted by the liver promote homeostasis and are pivotal for maintaining the liver-gut axis. Bile acid metabolism is one such example wherein, bile acid synthesis occurs in the liver and its biotransformation happens in the intestine. Dysfunctional interactions between the liver and the intestine stimulate varied pathological outcomes through its bidirectional portal communication. Indeed, aberrant bile acid metabolism has been reported in inflammatory bowel disease (IBD). However, the molecular mechanisms underlying these crosstalks that perpetuate intestinal permeability and inflammation remain obscure. Here, we identify a novel hepatic gene program regulated by Rela and Stat3 that accentuates the inflammation in an acute experimental colitis model. Hepatocyte-specific ablation of Rela and Stat3 reduces the levels of primary bile acids in both the liver and the gut and shows a restricted colitogenic phenotype. On supplementation of chenodeoxycholic acid (CDCA), knock-out mice exhibit enhanced colitis-induced alterations. This study provides persuasive evidence for the development of multi-organ strategies for treating IBD and identifies a hepatocyte-specific Rela-Stat3 network as a promising therapeutic target.


Subject(s)
Bile Acids and Salts , Colitis , Disease Models, Animal , Hepatocytes , Mice, Knockout , STAT3 Transcription Factor , Transcription Factor RelA , Animals , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , Colitis/chemically induced , Colitis/metabolism , Colitis/genetics , Colitis/pathology , Hepatocytes/metabolism , Transcription Factor RelA/metabolism , Transcription Factor RelA/genetics , Mice , Bile Acids and Salts/metabolism , Gene Expression Regulation , Liver/metabolism , Liver/pathology , Mice, Inbred C57BL
16.
EBioMedicine ; 106: 105265, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39096744

ABSTRACT

BACKGROUND: Bariatric surgery is an effective treatment option for obesity and provides long-term weight loss and positive effects on metabolism, but the underlying mechanisms are poorly understood. Alterations in bile acid metabolism have been suggested as a potential contributing factor, but comprehensive studies in humans are lacking. METHODS: In this study, we analysed the postprandial responses of bile acids, C4 and FGF19 in plasma, and excretion of bile acids in faeces, before and after bariatric surgery in patients (n = 38; 74% females) with obesity with or without type 2 diabetes from the BARIA cohort. FINDINGS: We observed that total fasting plasma bile acid levels increased, and faecal excretion of bile acids decreased after surgery suggesting increased reabsorption of bile acids. Consistent with increased bile acid levels after surgery we observed increased postprandial levels of FGF19 and suppression of the bile acid synthesis marker C4, suggesting increased FXR activation in the gut. We also noted that a subset of bile acids had altered postprandial responses before and after surgery. Finally, fasting plasma levels of 6α-hydroxylated bile acids, which are TGR5 agonists and associated with improved glucose metabolism, were increased after surgery and one of them, HDCA, covaried with diabetes remission in an independent cohort. INTERPRETATION: Our findings provide new insights regarding bile acid kinetics and suggest that bariatric surgery in humans alters bile acid profiles leading to activation of FXR and TGR5, which may contribute to weight loss, improvements in glucose metabolism, and diabetes remission. FUNDING: Novo Nordisk Fonden, Leducq Foundation, Swedish Heart-Lung Foundation, Knut and Alice Wallenberg Foundation, the ALF-agreement, ZonMw.


Subject(s)
Bariatric Surgery , Bile Acids and Salts , Diabetes Mellitus, Type 2 , Fibroblast Growth Factors , Obesity , Humans , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/surgery , Diabetes Mellitus, Type 2/blood , Bile Acids and Salts/metabolism , Bile Acids and Salts/blood , Bariatric Surgery/methods , Female , Male , Obesity/surgery , Obesity/metabolism , Obesity/blood , Middle Aged , Adult , Fibroblast Growth Factors/blood , Fibroblast Growth Factors/metabolism , Postprandial Period , Biomarkers , Feces/chemistry , Kinetics , Fasting
17.
Biol Pharm Bull ; 47(8): 1429-1436, 2024.
Article in English | MEDLINE | ID: mdl-39135238

ABSTRACT

Farnesoid X receptor (FXR) is a nuclear receptor that regulates the synthesis and enterohepatic circulation of bile acids (BAs). It also regulates lipid and carbohydrate metabolism, making FXR ligands potential therapeutic agents for systemic and/or hepatic metabolic disorders. We previously synthesized a series of FXR antagonists and showed that oral administration of FLG249 reduced the expression of several FXR target genes in the mouse ileum. Here, we investigated the effects of FLG249 on lipid metabolism in mice fed a high-fat diet (HFD). When FLG249 was administered for 4 weeks to HFD-induced obese mice, it altered the expression of genes related to BA metabolism, ceramide synthesis and fatty acid ß-oxidation, improving lipid metabolism in the liver and ileum without decreasing body weight. These findings suggest that FLG249 has the potential to be a low toxicity pharmaceutical compound and likely acts as a nonsteroidal FXR antagonist to improve lipid metabolism disorders.


Subject(s)
Cholesterol , Diet, High-Fat , Liver , Mice, Inbred C57BL , Obesity , Receptors, Cytoplasmic and Nuclear , Triglycerides , Animals , Diet, High-Fat/adverse effects , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/metabolism , Male , Liver/metabolism , Liver/drug effects , Obesity/drug therapy , Obesity/metabolism , Obesity/blood , Cholesterol/blood , Triglycerides/blood , Lipid Metabolism/drug effects , Bile Acids and Salts/metabolism , Mice , Mice, Obese , Ileum/metabolism , Ileum/drug effects
18.
Front Cell Infect Microbiol ; 14: 1411843, 2024.
Article in English | MEDLINE | ID: mdl-39104854

ABSTRACT

Biliary atresia (BA) is a progressive fibroinflammatory disease affecting both the extrahepatic and intrahepatic bile ducts, potentially leading to chronic cholestasis and biliary cirrhosis. Despite its prevalence, the exact mechanisms behind BA development remain incompletely understood. Recent research suggests that the gut microbiota and its metabolites may play significant roles in BA development. This paper offers a comprehensive review of the changing characteristics of gut microbiota and their metabolites at different stages of BA in children. It discusses their influence on the host's inflammatory response, immune system, and bile acid metabolism. The review also explores the potential of gut microbiota and metabolites as a therapeutic target for BA, with interventions like butyrate and gut microbiota preparations showing promise in alleviating BA symptoms. While progress has been made, further research is necessary to untangle the complex interactions between gut microbiota and BA, paving the way for more effective prevention and treatment strategies for this challenging condition.


Subject(s)
Bile Acids and Salts , Biliary Atresia , Gastrointestinal Microbiome , Gastrointestinal Microbiome/physiology , Humans , Biliary Atresia/microbiology , Biliary Atresia/metabolism , Bile Acids and Salts/metabolism , Animals
19.
J Ethnopharmacol ; 334: 118526, 2024 Nov 15.
Article in English | MEDLINE | ID: mdl-38972531

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Lean nonalcoholic steatohepatitis (NASH) poses a serious threat to public health worldwide. Herbs of the genus Gentiana have been used for centuries to treat hepatic disease or have been consumed for hepatic protection efficiency. Gentiopicroside (GPS), the main bioactive component of Gentiana herbs, has been shown to be beneficial for protecting the liver, improving intestinal disorders, modulating bile acid profiles, ameliorating alcoholic hepatosteatosis, and so on. It is plausible to speculate that GPS may hold potential as a therapeutic strategy for lean NASH. However, no related studies have been conducted thus far. AIM OF THE STUDY: The present work aimed to investigate the benefit of GPS on NASH in a lean mouse model. MATERIALS AND METHODS: NASH in a lean mouse model was successfully established via a published method. GPS of 50 and 100 mg/kg were orally administered to verify the effect. Untargeted metabolomics, 16S rDNA sequencing and bile acid (BA) profiling, as well as qPCR and Western blotting analysis were employed to investigate the mechanism underlying the alleviating effect. RESULTS: GPS significantly reduced the increase in serum biochemicals and liver index, and attenuated the accumulation of fat in the livers of lean mice with NASH. Forty-two potential biomarkers were identified by metabolomics analysis, leading to abnormal metabolic pathways of primary bile acid biosynthesis and fatty acid biosynthesis, which were subsequently rebalanced by GPS. A decreased Firmicutes/Bacteroidetes (F/B) ratio and disturbed BA related GM profiles were revealed in lean mice with NASH but were partially recovered by GPS. Furthermore, serum profiling of 23 BAs confirmed that serum BA levels were elevated in the lean model but downregulated by GPS treatment. Pearson correlation analysis validated associations between BA profiles, serum biochemical indices and related GM. qPCR and Western blotting analysis further elucidated the regulation of genes associated with liver lipid synthesis and bile acid metabolism. CONCLUSIONS: GPS may ameliorate steatosis in lean mice with NASH, regulating the metabolomic profile, BA metabolism, fatty acid biosynthesis, and BA-related GM. All these factors may contribute to its beneficial effect.


Subject(s)
Bile Acids and Salts , Iridoid Glucosides , Metabolomics , Non-alcoholic Fatty Liver Disease , RNA, Ribosomal, 16S , Animals , Iridoid Glucosides/pharmacology , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/blood , Bile Acids and Salts/metabolism , Mice , Male , RNA, Ribosomal, 16S/genetics , Mice, Inbred C57BL , Liver/drug effects , Liver/metabolism , Disease Models, Animal
20.
Front Endocrinol (Lausanne) ; 15: 1430720, 2024.
Article in English | MEDLINE | ID: mdl-39076513

ABSTRACT

Background: Bile acids (BAs), products of gut microbiota metabolism, have long been implicated in atherosclerotic disease pathogenesis. Characterizing the serum bile acid profile and exploring its potential role in carotid atherosclerosis (CAS) development are crucial tasks. Methods: In this study, we recruited 73 patients with CAS as the disease group and 77 healthy individuals as the control group. We systematically measured the serum concentrations of 15 bile acids using ultrahigh-performance liquid chromatography-mass spectrometry (UPLC-MS/MS). Multivariate logistic regression and least absolute shrinkage and selection operator (LASSO) regression were applied to analyze the impact of bile acids on the disease and select the key BAs. The possible molecular mechanism was elucidated by network pharmacology. Results: (1) The BA profile of patients with CAS significantly differed. (2) Multifactorial logistic regression analysis identified elevated levels of GCDCA (OR: 1.01, P < 0.001), DCA (OR: 1.01, P = 0.005), and TDCA (OR: 1.05, P = 0.002) as independent risk factors for CAS development. Conversely, GCA (OR: 0.99, P = 0.020), LCA (OR: 0.83, P = 0.002), and GUDCA (OR: 0.99, P = 0.003) were associated with protective effects against the disease. GCA, DCA, LCA, and TDCA were identified as the four key BAs. (3) TNF, FXR, GPBAR1, ESR1 and ACE were predicted to be targets of BAs against AS. These four BAs potentially impact AS progression by triggering signaling pathways, including cAMP, PPAR, and PI3K-AKT pathways, via their targets. Conclusion: This study offers valuable insights into potential therapeutic strategies for atherosclerosis that target bile acids.


Subject(s)
Bile Acids and Salts , Carotid Artery Diseases , Metabolomics , Network Pharmacology , Humans , Bile Acids and Salts/metabolism , Bile Acids and Salts/blood , Male , Female , Carotid Artery Diseases/metabolism , Carotid Artery Diseases/drug therapy , Carotid Artery Diseases/blood , Middle Aged , Metabolomics/methods , Aged , Case-Control Studies , Biomarkers/blood , Receptors, G-Protein-Coupled/metabolism , Tandem Mass Spectrometry
SELECTION OF CITATIONS
SEARCH DETAIL