Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 34
1.
Nat Commun ; 13(1): 2060, 2022 04 19.
Article En | MEDLINE | ID: mdl-35440584

Calorie restriction (CR) and fasting are common approaches to weight reduction, but the maintenance is difficult after resuming food consumption. Meanwhile, the gut microbiome associated with energy harvest alters dramatically in response to nutrient deprivation. Here, we reported that CR and high-fat diet (HFD) both remodeled the gut microbiota with similar microbial composition, Parabacteroides distasonis was most significantly decreased after CR or HFD. CR altered microbiota and reprogramed metabolism, resulting in a distinct serum bile acid profile characterized by depleting the proportion of non-12α-hydroxylated bile acids, ursodeoxycholic acid and lithocholic acid. Downregulation of UCP1 expression in brown adipose tissue and decreased serum GLP-1 were observed in the weight-rebound mice. Moreover, treatment with Parabacteroides distasonis or non-12α-hydroxylated bile acids ameliorated weight regain via increased thermogenesis. Our results highlighted the gut microbiota-bile acid crosstalk in rebound weight gain and Parabacteroides distasonis as a potential probiotic to prevent rapid post-CR weight gain.


Gastrointestinal Microbiome , Animals , Bacteroidetes , Bile Acids and Salts , Caloric Restriction , Diet, High-Fat/adverse effects , Gastrointestinal Microbiome/physiology , Mice , Mice, Inbred C57BL , Weight Gain
2.
Adv Sci (Weinh) ; 9(16): e2200263, 2022 05.
Article En | MEDLINE | ID: mdl-35285172

Bile reflux gastritis (BRG) is associated with the development of gastric cancer (GC), but the specific mechanism remains elusive. Here, a comprehensive study is conducted to explore the roles of refluxed bile acids (BAs) and microbiome in gastric carcinogenesis. The results show that conjugated BAs, interleukin 6 (IL-6), lipopolysaccharide (LPS), and the relative abundance of LPS-producing bacteria are increased significantly in the gastric juice of both BRG and GC patients. A secondary BA, taurodeoxycholic acid (TDCA), is significantly and positively correlated with the LPS-producing bacteria in the gastric juice of these patients. TDCA promotes the proliferation of normal gastric epithelial cells (GES-1) through activation of the IL-6/JAK1/STAT3 pathway. These results are further verified in two mouse models, one by gavage of TDCA, LPS, and LPS-producing bacteria (Prevotella melaninogenica), respectively, and the other by bile reflux (BR) surgery, mimicking clinical bile refluxing. Moreover, the bile reflux induced gastric precancerous lesions observed in the post BR surgery mice can be prevented by treatment with cryptotanshinone, a plant-derived STAT3 inhibitor. These results reveal an important underlying mechanism by which bile reflux promotes gastric carcinogenesis and provide an alternative strategy for the prevention of GC associated with BRG.


Bile Reflux , Carcinogenesis , Gastritis , Gastrointestinal Microbiome , Stomach Neoplasms , Taurodeoxycholic Acid , Animals , Bile Reflux/complications , Bile Reflux/pathology , Carcinogenesis/metabolism , Gastritis/complications , Gastritis/pathology , Humans , Interleukin-6/metabolism , Lipopolysaccharides , Mice , Stomach Neoplasms/etiology , Stomach Neoplasms/metabolism , Taurodeoxycholic Acid/metabolism
3.
J Gastroenterol Hepatol ; 37(1): 15-23, 2022 Jan.
Article En | MEDLINE | ID: mdl-34850445

Metabolism-associated fatty liver disease (MAFLD) is defined as the presence of excess fat in the liver in the absence of excess alcohol consumption and metabolic dysfunction. It has also been described as the hepatic manifestation of metabolic syndrome. The incidence of MAFLD has been reported to be 43-60% in diabetics, ~90% in patients with hyperlipidemia, and 91% in morbidly obese patients. Risk factors that have been associated with the development of MAFLD include male gender, increasing age, obesity, insulin resistance, diabetes, and hyperlipidemia. All of these risk factors have been linked to alterations of the gut microbiota, that is, gut dysbiosis. MAFLD can progress to non-alcoholic steatohepatitis with the presence of inflammation and ballooning, which can deteriorate into cirrhosis, MAFLD-related hepatocellular carcinoma, and liver failure. In this review, we will be focused on the role of the gut microbial metabolome in the development, progression, and potential treatment of MAFLD.


Gastrointestinal Microbiome , Metabolic Diseases , Non-alcoholic Fatty Liver Disease , Gastrointestinal Microbiome/physiology , Humans , Metabolic Diseases/complications , Non-alcoholic Fatty Liver Disease/epidemiology
5.
EBioMedicine ; 66: 103290, 2021 Apr.
Article En | MEDLINE | ID: mdl-33752128

BACKGROUND: Significantly elevated serum and hepatic bile acid (BA) concentrations have been known to occur in patients with liver fibrosis. However, the roles of different BA species in liver fibrogenesis are not fully understood. METHODS: We quantitatively measured blood BA concentrations in nonalcoholic steatohepatitis (NASH) patients with liver fibrosis and healthy controls. We characterized BA composition in three mouse models induced by carbon tetrachloride (CCl4), streptozotocin-high fat diet (STZ-HFD), and long term HFD, respectively. The molecular mechanisms underlying the fibrosis-promoting effects of BAs were investigated in cell line models, a 3D co-culture system, and a Tgr5 (HSC-specific) KO mouse model. FINDINGS: We found that a group of conjugated 12α-hydroxylated (12α-OH) BAs, such as taurodeoxycholate (TDCA) and glycodeoxycholate (GDCA), significantly increased in NASH patients and liver fibrosis mouse models. 12α-OH BAs significantly increased HSC proliferation and protein expression of fibrosis-related markers. Administration of TDCA and GDCA directly activated HSCs and promoted liver fibrogenesis in mouse models. Blockade of BA binding to TGR5 or inhibition of ERK1/2 and p38 MAPK signaling both significantly attenuated the BA-induced fibrogenesis. Liver fibrosis was attenuated in mice with Tgr5 depletion. INTERPRETATION: Increased hepatic concentrations of conjugated 12α-OH BAs significantly contributed to liver fibrosis via TGR5 mediated p38MAPK and ERK1/2 signaling. Strategies to antagonize TGR5 or inhibit ERK1/2 and p38 MAPK signaling may effectively prevent or reverse liver fibrosis. FUNDINGS: This study was supported by the National Institutes of Health/National Cancer Institute Grant 1U01CA188387-01A1, the National Key Research and Development Program of China (2017YFC0906800); the State Key Program of National Natural Science Foundation (81430062); the National Natural Science Foundation of China (81974073, 81774196), China Postdoctoral Science Foundation funded project, China (2016T90381), and E-institutes of Shanghai Municipal Education Commission, China (E03008).


Bile Acids and Salts/metabolism , Disease Susceptibility , Liver Cirrhosis/etiology , Liver Cirrhosis/metabolism , Animals , Bile Acids and Salts/blood , Biomarkers , Carbon Tetrachloride/adverse effects , Case-Control Studies , Cell Line , Diet, High-Fat/adverse effects , Disease Models, Animal , Hepatic Stellate Cells/metabolism , Humans , Hydroxylation , Liver Cirrhosis/pathology , Mice , Mice, Transgenic , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Signal Transduction , Streptozocin/adverse effects
6.
Nat Commun ; 12(1): 1487, 2021 03 05.
Article En | MEDLINE | ID: mdl-33674561

Hyocholic acid (HCA) is a major bile acid (BA) species in the BA pool of pigs, a species known for its exceptional resistance to spontaneous development of diabetic phenotypes. HCA and its derivatives are also present in human blood and urine. We investigate whether human HCA profiles can predict the development of metabolic disorders. We find in the first cohort (n = 1107) that both obesity and diabetes are associated with lower serum concentrations of HCA species. A separate cohort study (n = 91) validates this finding and further reveals that individuals with pre-diabetes are associated with lower levels of HCA species in feces. Serum HCA levels increase in the patients after gastric bypass surgery (n = 38) and can predict the remission of diabetes two years after surgery. The results are replicated in two independent, prospective cohorts (n = 132 and n = 207), where serum HCA species are found to be strong predictors for metabolic disorders in 5 and 10 years, respectively. These findings underscore the association of HCA species with diabetes, and demonstrate the feasibility of using HCA profiles to assess the future risk of developing metabolic abnormalities.


Biomarkers/blood , Cholic Acids/blood , Cholic Acids/urine , Metabolic Diseases/diagnosis , Adult , Cohort Studies , Cross-Sectional Studies , Diabetes Mellitus, Type 2/metabolism , Feces/chemistry , Female , Humans , Longitudinal Studies , Male , Middle Aged , Obesity/metabolism , Overweight/metabolism , Prediabetic State/diagnosis , Prospective Studies
7.
Cancers (Basel) ; 13(4)2021 Feb 10.
Article En | MEDLINE | ID: mdl-33578739

Three of the most lethal cancers in the world are the gastrointestinal cancers-gastric (GC), esophageal (EC) and colorectal cancer (CRC)-which are ranked as third, sixth and fourth in cancer deaths globally. Early detection of these cancers is difficult, and a quest is currently on to find non-invasive screening tests to detect these cancers. The reprogramming of energy metabolism is a hallmark of cancer, notably, an increased dependence on aerobic glycolysis which is often referred to as the Warburg effect. This metabolic change results in a unique metabolic profile that distinguishes cancer cells from normal cells. Serum metabolomics analyses allow one to measure the end products of both host and microbiota metabolism present at the time of sample collection. It is a non-invasive procedure requiring only blood collection which encourages greater patient compliance to have more frequent screenings for cancer. In the following review we will examine some of the most current serum metabolomics studies in order to compare their results and test a hypothesis that different tumors, notably, from EC, GC and CRC, have distinguishing serum metabolite profiles.

8.
Cell Metab ; 33(4): 791-803.e7, 2021 04 06.
Article En | MEDLINE | ID: mdl-33338411

Hyocholic acid (HCA) and its derivatives are found in trace amounts in human blood but constitute approximately 76% of the bile acid (BA) pool in pigs, a species known for its exceptional resistance to type 2 diabetes. Here, we show that BA depletion in pigs suppressed secretion of glucagon-like peptide-1 (GLP-1) and increased blood glucose levels. HCA administration in diabetic mouse models improved serum fasting GLP-1 secretion and glucose homeostasis to a greater extent than tauroursodeoxycholic acid. HCA upregulated GLP-1 production and secretion in enteroendocrine cells via simultaneously activating G-protein-coupled BA receptor, TGR5, and inhibiting farnesoid X receptor (FXR), a unique mechanism that is not found in other BA species. We verified the findings in TGR5 knockout, intestinal FXR activation, and GLP-1 receptor inhibition mouse models. Finally, we confirmed in a clinical cohort, that lower serum concentrations of HCA species were associated with diabetes and closely related to glycemic markers.


Cholic Acids/therapeutic use , Diabetes Mellitus, Experimental/drug therapy , Glucose/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Blood Glucose/analysis , Cell Line , Cholic Acids/blood , Cholic Acids/chemistry , Cholic Acids/pharmacology , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor/antagonists & inhibitors , Glucagon-Like Peptide-1 Receptor/metabolism , Humans , Isoxazoles/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/genetics , Signal Transduction/drug effects , Swine
9.
Protein Cell ; 12(5): 411-425, 2021 05.
Article En | MEDLINE | ID: mdl-33252713

The gut microbiota is profoundly involved in glucose and lipid metabolism, in part by regulating bile acid (BA) metabolism and affecting multiple BA-receptor signaling pathways. BAs are synthesized in the liver by multi-step reactions catalyzed via two distinct routes, the classical pathway (producing the 12α-hydroxylated primary BA, cholic acid), and the alternative pathway (producing the non-12α-hydroxylated primary BA, chenodeoxycholic acid). BA synthesis and excretion is a major pathway of cholesterol and lipid catabolism, and thus, is implicated in a variety of metabolic diseases including obesity, insulin resistance, and nonalcoholic fatty liver disease. Additionally, both oxysterols and BAs function as signaling molecules that activate multiple nuclear and membrane receptor-mediated signaling pathways in various tissues, regulating glucose, lipid homeostasis, inflammation, and energy expenditure. Modulating BA synthesis and composition to regulate BA signaling is an interesting and novel direction for developing therapies for metabolic disease. In this review, we summarize the most recent findings on the role of BA synthetic pathways, with a focus on the role of the alternative pathway, which has been under-investigated, in treating hyperglycemia and fatty liver disease. We also discuss future perspectives to develop promising pharmacological strategies targeting the alternative BA synthetic pathway for the treatment of metabolic diseases.


Bile Acids and Salts/metabolism , Gastrointestinal Microbiome , Lipid Metabolism , Metabolic Diseases , Non-alcoholic Fatty Liver Disease , Signal Transduction , Animals , Humans , Liver/metabolism , Metabolic Diseases/metabolism , Metabolic Diseases/microbiology , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/microbiology
10.
Protein Cell ; 12(5): 374-393, 2021 05.
Article En | MEDLINE | ID: mdl-32797354

Colorectal cancer (CRC) and hepatocellular carcinoma (HCC) are the second and third most common causes of death by cancer, respectively. The etiologies of the two cancers are either infectious insult or due to chronic use of alcohol, smoking, diet, obesity and diabetes. Pathological changes in the composition of the gut microbiota that lead to intestinal inflammation are a common factor for both HCC and CRC. However, the gut microbiota of the cancer patient evolves with disease pathogenesis in unique ways that are affected by etiologies and environmental factors. In this review, we examine the changes that occur in the composition of the gut microbiota across the stages of the HCC and CRC. Based on the idea that the gut microbiota are an additional "lifeline" and contribute to the tumor microenvironment, we can observe from previously published literature how the microbiota can cause a shift in the balance from normal → inflammation → diminished inflammation from early to later disease stages. This pattern leads to the hypothesis that tumor survival depends on a less pro-inflammatory tumor microenvironment. The differences observed in the gut microbiota composition between different disease etiologies as well as between HCC and CRC suggest that the tumor microenvironment is unique for each case.


Carcinoma, Hepatocellular/microbiology , Colorectal Neoplasms/microbiology , Gastrointestinal Microbiome , Liver Neoplasms/microbiology , Tumor Microenvironment , Humans , Risk Factors
11.
Metabolites ; 10(11)2020 Nov 23.
Article En | MEDLINE | ID: mdl-33238385

Theabrownin is one of the most bioactive compounds in Pu-erh tea. Our previous study revealed that the hypocholesterolemic effect of theabrownin was mediated by the modulation of bile salt hydrolase (BSH)-enriched gut microbiota and bile acid metabolism. In this study, we demonstrated that theabrownin ameliorated high-fat-diet (HFD)-induced obesity by modifying gut microbiota, especially those with 7α-dehydroxylation on the species level, and these changed microbes were positively correlated with secondary bile acid (BA) metabolism. Thus, altered intestinal BAs resulted in shifting bile acid biosynthesis from the classic to the alternative pathway. This shift changed the BA pool by increasing non-12α-hydroxylated-BAs (non-12OH-BAs) and decreasing 12α-hydroxylated BAs (12OH-BAs), which improved energy metabolism in white and brown adipose tissue. This study showed that theabrownin was a potential therapeutic modality for obesity and other metabolic disorders via gut microbiota-driven bile acid alternative synthesis.

12.
Sci Rep ; 10(1): 11601, 2020 07 14.
Article En | MEDLINE | ID: mdl-32665615

Bile acids (BAs) play essential roles in facilitating lipid digestion and absorption in the intestine. Gastric BAs were attributed to abnormal refluxing from duodenal compartments and correlated with the occurrence of gastric inflammation and carcinogenesis. However, the differences in gastric BAs between physiologically compromised and healthy individuals have not been fully investigated. In this study, gastric juice was collected from patients clinically diagnosed as gastritis with/without bile reflux and healthy subjects for BA profiles measurements. As a result, we found that the conjugated BAs became prominent components in bile reflux juice, whereas almost equal amounts of conjugated and unconjugated BAs existed in non-bile reflux and healthy juice. To investigate whether gastric BA changes were regulated by hepatic BA synthesis, C57BL/6J mice were intervened with GW4064/resin to decrease/increase hepatic BA synthesis. The results revealed that changes of gastric BAs were coordinated with hepatic BA changes. Additionally, gastric BAs were detected in several healthy mammals, in which there were no obvious differences between the conjugated and unconjugated BAs. Pigs were an exception. Thus, increased levels of conjugated BAs are associated with human bile reflux gastritis. Gastric conjugated BAs could become a panel of biomarkers to facilitate diagnosis of pathological bile reflux.


Bile Acids and Salts/metabolism , Bile Reflux/metabolism , Gastritis/metabolism , Liver/metabolism , Animals , Bile Acids and Salts/biosynthesis , Bile Reflux/genetics , Bile Reflux/pathology , Digestion/physiology , Disease Models, Animal , Gastric Juice/metabolism , Gastritis/pathology , Humans , Intestinal Mucosa/metabolism , Intestines/pathology , Isoxazoles/pharmacology , Lipids/chemistry , Mice
13.
BMC Med ; 18(1): 144, 2020 06 05.
Article En | MEDLINE | ID: mdl-32498677

BACKGROUND: Accurate and noninvasive diagnosis and staging of liver fibrosis are essential for effective clinical management of chronic liver disease (CLD). We aimed to identify serum metabolite markers that reliably predict the stage of fibrosis in CLD patients. METHODS: We quantitatively profiled serum metabolites of participants in 2 independent cohorts. Based on the metabolomics data from cohort 1 (504 HBV associated liver fibrosis patients and 502 normal controls, NC), we selected a panel of 4 predictive metabolite markers. Consequently, we constructed 3 machine learning models with the 4 metabolite markers using random forest (RF), to differentiate CLD patients from normal controls (NC), to differentiate cirrhosis patients from fibrosis patients, and to differentiate advanced fibrosis from early fibrosis, respectively. RESULTS: The panel of 4 metabolite markers consisted of taurocholate, tyrosine, valine, and linoelaidic acid. The RF models of the metabolite panel demonstrated the strongest stratification ability in cohort 1 to diagnose CLD patients from NC (area under the receiver operating characteristic curve (AUROC) = 0.997 and the precision-recall curve (AUPR) = 0.994), to differentiate fibrosis from cirrhosis (0.941, 0.870), and to stage liver fibrosis (0.918, 0.892). The diagnostic accuracy of the models was further validated in an independent cohort 2 consisting of 300 CLD patients with chronic HBV infection and 90 NC. The AUCs of the models were consistently higher than APRI, FIB-4, and AST/ALT ratio, with both greater sensitivity and specificity. CONCLUSIONS: Our study showed that this 4-metabolite panel has potential usefulness in clinical assessments of CLD progression in patients with chronic hepatitis B virus infection.


Biomarkers/blood , Hepatitis B, Chronic/complications , Liver Cirrhosis/diagnosis , Adult , China , Cohort Studies , Female , Hepatitis B, Chronic/blood , Humans , Liver Cirrhosis/blood , Male , Sensitivity and Specificity
14.
EBioMedicine ; 55: 102766, 2020 May.
Article En | MEDLINE | ID: mdl-32408110

BACKGROUND: The composition of the bile acid (BA) pool is closely associated with obesity and is modified by gut microbiota. Perturbations of gut microbiota shape the BA composition, which, in turn, may alter important BA signaling and affect host metabolism. METHODS: We investigated BA composition of high BMI subjects from a human cohort study and a high fat diet (HFD) obesity prone (HF-OP) / HFD obesity resistant (HF-OR) mice model. Gut microbiota was analysed by metagenomics sequencing. GLP-1 secretion and gene regulation studies involved ELISA, qPCR, Western blot, Immunohistochemistry, and Immunofluorescence staining. FINDINGS: We found that the proportion of non-12-OH BAs was significantly decreased in the unhealthy high BMI subjects. The HF-OR mice had an enhanced level of non-12-OH BAs. Non-12-OH BAs including ursodeoxycholate (UDCA), chenodeoxycholate (CDCA), and lithocholate (LCA) were decreased in the HF-OP mice and associated with altered gut microbiota. Clostridium scindens was decreased in HF-OP mice and had a positive correlation with UDCA and LCA. Gavage of Clostridium scindens in mice increased the levels of hepatic non-12-OH BAs, accompanied by elevated serum 7α-hydroxy-4-cholesten-3-one (C4) levels. In HF-OP mice, altered BA composition was associated with significantly downregulated expression of GLP-1 in ileum and PGC1α, UCP1 in brown adipose tissue. In addition, we identified that UDCA attenuated the high fat diet-induced obesity via enhancing levels of non-12-OH BAs. INTERPRETATION: Our study highlights that dysregulated BA signaling mediated by gut microbiota contributes to obesity susceptibility, suggesting modulation of BAs could be a promising strategy for obesity therapy.


Chenodeoxycholic Acid/metabolism , Gastrointestinal Microbiome , Ileum/microbiology , Lithocholic Acid/metabolism , Obesity/microbiology , Ursodeoxycholic Acid/metabolism , Adipose Tissue, Brown/metabolism , Animals , Body Mass Index , Cholestenones/metabolism , Clostridiales/metabolism , Clostridiales/pathogenicity , Cohort Studies , Diet, High-Fat/adverse effects , Disease Models, Animal , Disease Susceptibility , Gene Expression Regulation , Glucagon-Like Peptide 1/genetics , Glucagon-Like Peptide 1/metabolism , Humans , Ileum/metabolism , Male , Metagenomics/methods , Mice , Mice, Inbred C57BL , Obesity/etiology , Obesity/genetics , Obesity/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism
15.
Med Res Rev ; 40(4): 1496-1507, 2020 07.
Article En | MEDLINE | ID: mdl-31808182

Recent epidemiological and molecular studies have linked the disruption of cholesterol homeostasis to increased risk for developing Alzheimer's disease (AD). Emerging evidence also suggests that brain cholesterol accumulation contributes to the progression of hepatic encephalopathy (HE) via bile acid (BA)-mediated effects on the farnesoid X receptor. In this perspective paper, we reviewed several recently published studies that suggested a role for the gut microbiota transformation of BAs as a factor in AD and HE development/progression. We hypothesize that in addition to cholesterol elimination pathways, alteration of the gut microbiota and subsequent changes in both the serum and brain BA profiles are mechanistically involved in the development of both AD and HE, and thus, are a potential target for the prevention and treatment of the two diseases. Our understanding of the microbiome-BAs-brain axis in central nervous system disease is still evolving, and critical questions regarding the emerging links among central, peripheral, and intestinal metabolic failures contributing to brain health and disease during aging have yet to be addressed.


Alzheimer Disease/pathology , Bile Acids and Salts/metabolism , Brain/metabolism , Disease Progression , Gastrointestinal Microbiome , Hepatic Encephalopathy/pathology , Alzheimer Disease/microbiology , Hepatic Encephalopathy/microbiology , Humans , Models, Biological
16.
Nat Commun ; 10(1): 4971, 2019 10 31.
Article En | MEDLINE | ID: mdl-31672964

Pu-erh tea displays cholesterol-lowering properties, but the underlying mechanism has not been elucidated. Theabrownin is one of the most active and abundant pigments in Pu-erh tea. Here, we show that theabrownin alters the gut microbiota in mice and humans, predominantly suppressing microbes associated with bile-salt hydrolase (BSH) activity. Theabrownin increases the levels of ileal conjugated bile acids (BAs) which, in turn, inhibit the intestinal FXR-FGF15 signaling pathway, resulting in increased hepatic production and fecal excretion of BAs, reduced hepatic cholesterol, and decreased lipogenesis. The inhibition of intestinal FXR-FGF15 signaling is accompanied by increased gene expression of enzymes in the alternative BA synthetic pathway, production of hepatic chenodeoxycholic acid, activation of hepatic FXR, and hepatic lipolysis. Our results shed light into the mechanisms behind the cholesterol- and lipid-lowering effects of Pu-erh tea, and suggest that decreased intestinal BSH microbes and/or decreased FXR-FGF15 signaling may be potential anti-hypercholesterolemia and anti-hyperlipidemia therapies.


Bile Acids and Salts/metabolism , Catechin/analogs & derivatives , Fermented Foods , Gastrointestinal Microbiome/drug effects , Hypercholesterolemia/metabolism , Tea , Adult , Amidohydrolases/metabolism , Animals , Catechin/pharmacology , Chenodeoxycholic Acid/metabolism , Cholesterol/metabolism , Diet, High-Fat , Fecal Microbiota Transplantation , Fibroblast Growth Factors/drug effects , Fibroblast Growth Factors/metabolism , Gastrointestinal Microbiome/genetics , Gastrointestinal Microbiome/physiology , Humans , Ileum/drug effects , Ileum/metabolism , Lipogenesis/drug effects , Liver/drug effects , Liver/metabolism , Male , Metabolomics , Mice , Plant Extracts/pharmacology , RNA, Ribosomal, 16S , Receptors, Cytoplasmic and Nuclear/drug effects , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction , Young Adult
17.
EBioMedicine ; 37: 294-306, 2018 Nov.
Article En | MEDLINE | ID: mdl-30344125

BACKGROUND: Hepatic encephalopathy (HE), a severe neuropsychiatric complication, is associated with increased blood levels of ammonia and bile acids (BAs). We sought to determine (1) whether abnormally increased blood BAs in liver cirrhotic patients with HE is caused by elevation of apical sodium-dependent BA transporter (ASBT)-mediated BA reabsorption; and (2) whether increased BA reabsorption would exacerbate ammonia-induced brain injuries. METHODS: We quantitatively measured blood BA and ammonia levels in liver cirrhosis patients with or without HE and healthy controls. We characterized ASBT expression, BA profiles, and ammonia concentrations in a chronic liver disease (CLD) mouse model induced by streptozotocin-high fat diet (STZ-HFD) and an azoxymethane (AOM) - induced acute liver failure (ALF) mouse model. These two mouse models were treated with SC-435 (ASBT inhibitor) and budesonide (ASBT activator), respectively. FINDINGS: Blood concentrations of ammonia and conjugated BAs were substantially increased in cirrhotic patients with HE (n = 75) compared to cirrhotic patients without HE (n = 126). Pharmacological inhibition of the enterohepatic BA circulation using a luminal- restricted ASBT inhibitor, SC-435, in mice with AOM-induced ALF and STZ-HFD -induced CLD effectively reduced BA and ammonia concentrations in the blood and brain, and alleviated liver and brain damages. Budesonide treatment induced liver and brain damages in normal mice, and exacerbated these damages in AOM-treated mice. INTERPRETATION: ASBT mediated BA reabsorption increases intestinal luminal pH and facilitates conversion of intestinal ammonium to ammonia, leading to abnormally high levels of neurotoxic ammonia and cytotoxic BAs in the blood and brain. Inhibition of intestinal ASBT with SC-435 can effectively remove neurotoxic BAs and ammonia from the bloodstream and thus, mitigate liver and brain injuries resulting from liver failure.


Ammonia/blood , Bile Acids and Salts/blood , End Stage Liver Disease/blood , Hepatic Encephalopathy/blood , Liver Failure, Acute/blood , Animals , Cell Line , Disease Models, Animal , End Stage Liver Disease/chemically induced , End Stage Liver Disease/pathology , Female , Hepatic Encephalopathy/chemically induced , Hepatic Encephalopathy/pathology , Humans , Liver Failure, Acute/chemically induced , Liver Failure, Acute/pathology , Male , Mice , Middle Aged
18.
Front Med ; 12(6): 608-623, 2018 Dec.
Article En | MEDLINE | ID: mdl-30306382

Diabetes is a widespread, rapidly increasing metabolic disease that is driven by hyperglycemia. Early glycemic control is of primary importance to avoid vascular complications including development of retinal disorders leading to blindness, end-stage renal disease, and accelerated atherosclerosis with a higher risk of myocardial infarction, stroke and limb amputations. Even after hyperglycemia has been brought under control, "metabolic memory," a cluster of irreversible metabolic changes that allow diabetes to progress, may persist depending on the duration of hyperglycemia. Manipulation of bile acid (BA) receptors and the BA pool have been shown to be useful in establishing glycemic control in diabetes due to their ability to regulate energy metabolism by binding and activating nuclear transcription factors such as farnesoid X receptor (FXR) in liver and intestine as well as the G-protein coupled receptor, TGR5, in enteroendocrine cells and pancreatic ß-cells. The downstream targets of BA activated FXR, FGF15/21, are also important for glucose/insulin homeostasis. In this review we will discuss the effect of BAs on glucose and lipid metabolism and explore recent research on establishing glycemic control in diabetes through the manipulation of BAs and their receptors in the liver, intestine and pancreas, alteration of the enterohepatic circulation, bariatric surgery and alignment of circadian rhythms.


Bile Acids and Salts/metabolism , Blood Glucose/metabolism , Diabetes Mellitus/metabolism , Intestinal Mucosa/metabolism , Liver/metabolism , Animals , Bile Acids and Salts/blood , Blood Glucose/drug effects , Circadian Rhythm , Diabetes Mellitus/blood , Diabetes Mellitus/drug therapy , Energy Metabolism , Homeostasis , Humans , Hyperglycemia/metabolism , Hyperglycemia/physiopathology , Hypoglycemic Agents/therapeutic use , Intestines/drug effects , Lipid Metabolism , Liver/drug effects , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction
19.
EBioMedicine ; 35: 124-132, 2018 Sep.
Article En | MEDLINE | ID: mdl-30100397

Clinical prediction of advanced hepatic fibrosis (HF) and cirrhosis has long been challenging due to the gold standard, liver biopsy, being an invasive approach with certain limitations. Less invasive blood test tandem with a cutting-edge machine learning algorithm shows promising diagnostic potential. In this study, we constructed and compared machine learning methods with the FIB-4 score in a discovery dataset (n = 490) of hepatitis B virus (HBV) patients. Models were validated in an independent HBV dataset (n = 86). We further employed these models on two independent hepatitis C virus (HCV) datasets (n = 254 and 230) to examine their applicability. In the discovery data, gradient boosting (GB) stably outperformed other methods as well as FIB-4 scores (p < .001) in the prediction of advanced HF and cirrhosis. In the HBV validation dataset, for classification between early and advanced HF, the area under receiver operating characteristic curves (AUROC) of GB model was 0.918, while FIB-4 was 0.841; for classification between non-cirrhosis and cirrhosis, GB showed AUROC of 0.871, while FIB-4 was 0.830. Additionally, GB-based prediction demonstrated good classification capacity on two HCV datasets while higher cutoffs for both GB and FIB-4 scores were required to achieve comparable specificity and sensitivity. Using the same parameters as FIB-4, the GB-based prediction system demonstrated steady improvements relative to FIB-4 in HBV and HCV cohorts with different cutoff values required in different etiological groups. A user-friendly web tool, LiveBoost, makes our prediction models freely accessible for further clinical studies and applications.


Hepacivirus/physiology , Hepatitis B virus/physiology , Liver Cirrhosis/diagnosis , Liver Cirrhosis/virology , Machine Learning , Adult , Area Under Curve , Cohort Studies , Female , Humans , Internet , Male , Middle Aged , Models, Biological , ROC Curve , Reproducibility of Results
20.
Adv Exp Med Biol ; 1061: 95-110, 2018.
Article En | MEDLINE | ID: mdl-29956209

Non-alcoholic fatty liver disease (NAFLD) is defined as the presence of excess fat in the liver parenchyma in the absence of excess alcohol consumption and overt inflammation. It has also been described as the hepatic manifestation of metabolic syndrome (Than NN, Newsome PN, Atherosclerosis. 239:192-202, 2015). The incidence of NAFLD has been reported to be 43-60% in diabetics, ~90% in patients with hyperlipidemia and 91% in morbidly obese patients (Than NN, Newsome PN, Atherosclerosis. 239:192-202, 2015, Machado M, Marques-Vidal P, Cortez-Pinto H, J Hepatol, 45:600-606, 2006, Vernon G, Baranova A, Younossi ZM, Aliment Pharmacol Ther, 34:274-285, 2011). The risk factors that have been associated with the development of NAFLD include male gender, increasing age, obesity, insulin resistance, diabetes and hyperlipidemia (Attar BM, Van Thiel DH, Sci World J, 2013:481893, 2013, Gaggini M, Morelli M, Buzzigoli E, DeFronzo RA, Bugianesi E, Gastaldelli A, Forum Nutr, 5:1544-1460, 2013). All of these risk factors have been linked to alterations of the gut microbiota, ie., gut dysbiosis (He X, Ji G, Jia W, Li H, Int J Mol Sci, 17:300, 2016). However, it must be pointed out that the prevalence of NAFLD in normal weight individuals without metabolic risk factors is ~16% (Than NN, Newsome PN, Atherosclerosis. 239:192-202, 2015). This fact has led some investigators to hypothesize that the gut microbiota can impact lipid metabolism in the liver independently of obesity-related metabolic factors (Marchesi JR, Adams DH, Fava F, Hermes GD, Hirschfield GM, Hold g, et al., Gut, 65:330 339, 2016) (Le Roy T, Llopis M, Lepage P, Bruneau A, Rabot S, Bevilacqua C, et al., Gut, 62:1787-1794, 2013). In this chapter, we will explore the effect of the gut microbiota on hepatic lipid metabolism and how this affects the development of NAFLD.


Gastrointestinal Microbiome , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/microbiology , Animals , Humans , Non-alcoholic Fatty Liver Disease/pathology
...