Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 101
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Biol Chem ; 299(5): 104672, 2023 05.
Article in English | MEDLINE | ID: mdl-37019215

ABSTRACT

It is a great honor to be invited to write a reflections article on my scientific journey and lifelong bile acid research for the Journal of Biological Chemistry, in which I am proud to have published 24 articles. I have also published 21 articles in the Journal of Lipid Research, another journal of the American Society of Biochemistry and Molecular Biology. I begin my reflections from my early education in Taiwan, my coming to America for graduate study, and continue with my postdoctoral training in cytochrome P450 research, and my lifelong bile acid research career at Northeast Ohio Medical University. I have witnessed and helped in the transformation of this rural not so visible medical school to a well-funded leader in liver research. Writing this reflections article on my long and rewarding journey in bile acid research brings back many good memories. I am proud of my scientific contributions and attribute my academic success to hard work, perseverance, good mentoring, and networking. I hope these reflections of my academic career would help inspire young investigators to pursue an academic career in biochemistry and metabolic diseases.


Subject(s)
Bile Acids and Salts , Biochemistry , Biomedical Research , Liver , Humans , Bile Acids and Salts/metabolism , Biochemistry/history , Liver/enzymology , Liver/metabolism , Liver/pathology , Taiwan , Cytochrome P-450 Enzyme System , Ohio , Biomedical Research/history
2.
Curr Opin Gastroenterol ; 39(3): 249-255, 2023 05 01.
Article in English | MEDLINE | ID: mdl-36976875

ABSTRACT

PURPOSE OF REVIEW: This review aims to provide a concise update on recent advances in understanding of the bile acid metabolism and signaling in health and diseases. RECENT FINDINGS: CYP2C70 has been identified as the murine cytochrome p450 enzyme that mediates the synthesis of muricholic acids to account for the major different bile acid composition between human and mice. Several studies have linked nutrient sensing bile acid signaling to the regulation of hepatic autophagy-lysosome activity, an integral pathway of the cellular adaptive response to starvation. Distinct bile acid-mediated signaling mechanisms have been shown to contribute to the complex metabolic changes post bariatric surgery, suggesting that pharmacological manipulation of the enterohepatic bile acid signaling could be a potential nonsurgical alternative to weight loss surgery. SUMMARY: Basic and clinical studies have continued to discover novel roles of the enterohepatic bile acid signaling in regulation of key metabolic pathways. Such knowledge forms the molecular basis needed for developing safe and effective bile acid-based therapeutics for treating metabolic and inflammatory diseases.


Subject(s)
Bariatric Surgery , Bile Acids and Salts , Humans , Mice , Animals , Bile Acids and Salts/metabolism , Signal Transduction , Liver
3.
Int J Mol Sci ; 23(15)2022 07 28.
Article in English | MEDLINE | ID: mdl-35955473

ABSTRACT

The digestive tract is replete with complex and diverse microbial communities that are important for the regulation of multiple pathophysiological processes in humans and animals, particularly those involved in the maintenance of intestinal homeostasis, immunity, inflammation, and tumorigenesis. The diversity of bile acids is a result of the joint efforts of host and intestinal microflora. There is a bidirectional relationship between the microbial community of the intestinal tract and bile acids in that, while the microbial flora tightly modulates the metabolism and synthesis of bile acids, the bile acid pool and composition affect the diversity and the homeostasis of the intestinal flora. Homeostatic imbalances of bile acid and intestinal flora systems may lead to the development of a variety of diseases, such as inflammatory bowel disease (IBD), colorectal cancer (CRC), hepatocellular carcinoma (HCC), type 2 diabetes (T2DM), and polycystic ovary syndrome (PCOS). The interactions between bile acids and intestinal flora may be (in)directly involved in the pathogenesis of these diseases.


Subject(s)
Carcinoma, Hepatocellular , Diabetes Mellitus, Type 2 , Gastrointestinal Microbiome , Liver Neoplasms , Animals , Bile Acids and Salts , Humans
4.
Am J Physiol Gastrointest Liver Physiol ; 318(3): G554-G573, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31984784

ABSTRACT

Bile acid synthesis is the most significant pathway for catabolism of cholesterol and for maintenance of whole body cholesterol homeostasis. Bile acids are physiological detergents that absorb, distribute, metabolize, and excrete nutrients, drugs, and xenobiotics. Bile acids also are signal molecules and metabolic integrators that activate nuclear farnesoid X receptor (FXR) and membrane Takeda G protein-coupled receptor 5 (TGR5; i.e., G protein-coupled bile acid receptor 1) to regulate glucose, lipid, and energy metabolism. The gut-to-liver axis plays a critical role in the transformation of primary bile acids to secondary bile acids, in the regulation of bile acid synthesis to maintain composition within the bile acid pool, and in the regulation of metabolic homeostasis to prevent hyperglycemia, dyslipidemia, obesity, and diabetes. High-fat and high-calorie diets, dysbiosis, alcohol, drugs, and disruption of sleep and circadian rhythms cause metabolic diseases, including alcoholic and nonalcoholic fatty liver diseases, obesity, diabetes, and cardiovascular disease. Bile acid-based drugs that target bile acid receptors are being developed for the treatment of metabolic diseases of the liver.


Subject(s)
Bile Acids and Salts/metabolism , Fatty Liver/metabolism , Liver/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Fatty Liver/diagnosis , Fatty Liver/drug therapy , Gastrointestinal Agents/therapeutic use , Humans , Liver/drug effects , Liver/pathology , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, G-Protein-Coupled/agonists , Signal Transduction
5.
Annu Rev Nutr ; 39: 175-200, 2019 08 21.
Article in English | MEDLINE | ID: mdl-31018107

ABSTRACT

Bile acids facilitate nutrient absorption and are endogenous ligands for nuclear receptors that regulate lipid and energy metabolism. The brain-gut-liver axis plays an essential role in maintaining overall glucose, bile acid, and immune homeostasis. Fasting and feeding transitions alter nutrient content in the gut, which influences bile acid composition and pool size. In turn, bile acid signaling controls lipid and glucose use and protection against inflammation. Altered bile acid metabolism resulting from gene mutations, high-fat diets, alcohol, or circadian disruption can contribute to cholestatic and inflammatory diseases, diabetes, and obesity. Bile acids and their derivatives are valuable therapeutic agents for treating these inflammatory metabolic diseases.


Subject(s)
Bile Acids and Salts/metabolism , Animals , Gastrointestinal Microbiome , Glucose/metabolism , Homeostasis , Humans , Signal Transduction/physiology
6.
Hepatology ; 70(3): 955-970, 2019 09.
Article in English | MEDLINE | ID: mdl-30664797

ABSTRACT

Activation of the nuclear bile acid receptor farnesoid X receptor (FXR) protects against hepatic inflammation and injury, while Takeda G protein-coupled receptor 5 (TGR5) promotes adipose tissue browning and energy metabolism. Here, we examined the physiological and metabolic effects of the deficiency of these two bile acid receptors on hepatic metabolism and injury in mice. Fxr/Tgr5 double knockout mice (DKO) were generated for metabolic phenotyping. Male DKO mice fed a chow diet had reduced liver lipid levels but increased serum cholesterol levels. Liver cholesterol 7α-hydroxylase (Cyp7a1) activity and sterol 12α-hydroxylase mRNA levels were induced, while ileum FXR target genes were suppressed in DKO mice compared to wild-type (WT) mice. Bile acid pool size was increased in DKO mice, with increased taurocholic acid and decreased tauromuricholic acids. RNA sequencing analysis of the liver transcriptome revealed that bile acid synthesis and fibrosis gene expression levels are increased in chow-fed DKO mice compared to WT mice and that the top regulated pathways are involved in steroid/cholesterol biosynthesis, liver cirrhosis, and connective tissue disease. Cholestyramine treatment further induced Cyp7a1 mRNA and protein in DKO mice and increased bile acid pool size, while cholic acid also induced Cyp7a1 in DKO mice, suggesting impaired bile acid feedback regulation. A Western diet containing 0.2% cholesterol increased oxidative stress and markers of liver fibrosis but not hepatic steatosis in DKO mice. Conclusion: FXR and TGR5 play critical roles in protecting the liver from inflammation and fibrosis, and deficiency of both of these bile acid receptors in mice increased cholic acid synthesis and the bile acid pool, liver fibrosis, and inflammation; FXR and TGR5 DKO mice may be a model for liver fibrosis.


Subject(s)
Cholesterol 7-alpha-Hydroxylase/genetics , Liver Cirrhosis/genetics , Receptors, Cytoplasmic and Nuclear/deficiency , Receptors, G-Protein-Coupled/genetics , Animals , Bile Acids and Salts/metabolism , Biopsy, Needle , Cholic Acid/metabolism , Diet, Western , Disease Models, Animal , Disease Progression , Fatty Liver/metabolism , Fatty Liver/pathology , Gene Expression Regulation , Immunohistochemistry , Liver Cirrhosis/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidative Stress , Random Allocation , Receptors, Cytoplasmic and Nuclear/genetics , Signal Transduction/genetics
7.
Hepatology ; 68(4): 1574-1588, 2018 10.
Article in English | MEDLINE | ID: mdl-29486523

ABSTRACT

Bile acids activate farnesoid X receptor (FXR) and G protein-coupled bile acid receptor-1 (aka Takeda G protein-coupled receptor-5 [TGR5]) to regulate bile acid metabolism and glucose and insulin sensitivity. FXR and TGR5 are coexpressed in the enteroendocrine L cells, but their roles in integrated regulation of metabolism are not completely understood. We reported recently that activation of FXR induces TGR5 to stimulate glucagon-like peptide-1 (GLP-1) secretion to improve insulin sensitivity and hepatic metabolism. In this study, we used the intestine-restricted FXR agonist fexaramine (FEX) to study the effect of activation of intestinal FXR on the gut microbiome, bile acid metabolism, and FXR and TGR5 signaling. The current study revealed that FEX markedly increased taurolithocholic acid, increased secretion of fibroblast growth factors 15 and 21 and GLP-1, improved insulin and glucose tolerance, and promoted white adipose tissue browning in mice. Analysis of 16S ribosomal RNA sequences of the gut microbiome identified the FEX-induced and lithocholic acid-producing bacteria Acetatifactor and Bacteroides. Antibiotic treatment completely reversed the FEX-induced metabolic phenotypes and inhibited taurolithocholic acid synthesis, adipose tissue browning, and liver bile acid synthesis gene expression but further increased intestinal FXR target gene expression. FEX treatment effectively improved lipid profiles, increased GLP-1 secretion, improved glucose and insulin tolerance, and promoted adipose tissue browning, while antibiotic treatment reversed the beneficial metabolic effects of FEX in obese and diabetic mice. CONCLUSION: This study uncovered a mechanism in which activation of intestinal FXR shaped the gut microbiota to activate TGR5/GLP-1 signaling to improve hepatic glucose and insulin sensitivity and increase adipose tissue browning; the gut microbiota plays a critical role in bile acid metabolism and signaling to regulate metabolic homeostasis in health and disease. (Hepatology 2018).


Subject(s)
Bile Acids and Salts/metabolism , GTP-Binding Proteins/metabolism , Gastrointestinal Microbiome/drug effects , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, G-Protein-Coupled/metabolism , Animals , Disease Models, Animal , Glucagon-Like Peptide 1/metabolism , Lipid Metabolism , Male , Mice , Mice, Inbred C57BL , Random Allocation , Receptors, Cytoplasmic and Nuclear/pharmacology , Sensitivity and Specificity , Signal Transduction
8.
J Biol Chem ; 292(26): 11055-11069, 2017 06 30.
Article in English | MEDLINE | ID: mdl-28478385

ABSTRACT

The bile acid-activated receptors, nuclear farnesoid X receptor (FXR) and the membrane Takeda G-protein receptor 5 (TGR5), are known to improve glucose and insulin sensitivity in obese and diabetic mice. However, the metabolic roles of these two receptors and the underlying mechanisms are incompletely understood. Here, we studied the effects of the dual FXR and TGR5 agonist INT-767 on hepatic bile acid synthesis and intestinal secretion of glucagon-like peptide-1 (GLP-1) in wild-type, Fxr-/-, and Tgr5-/- mice. INT-767 efficaciously stimulated intracellular Ca2+ levels, cAMP activity, and GLP-1 secretion and improved glucose and lipid metabolism more than did the FXR-selective obeticholic acid and TGR5-selective INT-777 agonists. Interestingly, INT-767 reduced expression of the genes in the classic bile acid synthesis pathway but induced those in the alternative pathway, which is consistent with decreased taurocholic acid and increased tauromuricholic acids in bile. Furthermore, FXR activation induced expression of FXR target genes, including fibroblast growth factor 15, and unexpectedly Tgr5 and prohormone convertase 1/3 gene expression in the ileum. We identified an FXR-responsive element on the Tgr5 gene promoter. Fxr-/- and Tgr5-/- mice exhibited reduced GLP-1 secretion, which was stimulated by INT-767 in the Tgr5-/- mice but not in the Fxr-/- mice. Our findings uncovered a novel mechanism in which INT-767 activation of FXR induces Tgr5 gene expression and increases Ca2+ levels and cAMP activity to stimulate GLP-1 secretion and improve hepatic glucose and lipid metabolism in high-fat diet-induced obese mice. Activation of both FXR and TGR5 may therefore represent an effective therapy for managing hepatic steatosis, obesity, and diabetes.


Subject(s)
Bile Acids and Salts/biosynthesis , Gene Expression Regulation , Liver/metabolism , Obesity/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Bile Acids and Salts/genetics , Dietary Fats , Glucagon-Like Peptide 1/genetics , Glucagon-Like Peptide 1/metabolism , Glucose/metabolism , Lipid Metabolism , Mice , Mice, Knockout , Obesity/genetics , Obesity/pathology , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, G-Protein-Coupled/genetics
9.
Am J Pathol ; 187(8): 1658-1659, 2017 08.
Article in English | MEDLINE | ID: mdl-28728746

ABSTRACT

This commentary highlights the article by Jena et al that studied the complex interplay between diet, bile acids, sex, and dysbiosis in hepatic steatosis and inflammation.


Subject(s)
Diet, High-Fat , Non-alcoholic Fatty Liver Disease , Bile Acids and Salts , Gastrointestinal Microbiome , Humans , Sex Characteristics
10.
Hepatology ; 65(3): 813-827, 2017 03.
Article in English | MEDLINE | ID: mdl-27351453

ABSTRACT

Bile acids are signaling molecules that play a critical role in regulation of hepatic metabolic homeostasis by activating nuclear farnesoid X receptor (Fxr) and membrane G-protein-coupled receptor (Takeda G-protein-coupled receptor 5; Tgr5). The role of FXR in regulation of bile acid synthesis and hepatic metabolism has been studied extensively. However, the role of TGR5 in hepatic metabolism has not been explored. The liver plays a central role in lipid metabolism, and impaired response to fasting and feeding contributes to steatosis and nonalcoholic fatty liver and obesity. We have performed a detailed analysis of gallbladder bile acid and lipid metabolism in Tgr5-/- mice in both free-fed and fasted conditions. Lipid profiles of serum, liver and adipose tissues, bile acid composition, energy metabolism, and messenger RNA and protein expression of the genes involved in lipid metabolism were analyzed. Results showed that deficiency of the Tgr5 gene in mice alleviated fasting-induced hepatic lipid accumulation. Expression of liver oxysterol 7α-hydroxylase in the alternative bile acid synthesis pathway was reduced. Analysis of gallbladder bile acid composition showed marked increase of taurocholic acid and decrease of tauro-α and ß-muricholic acid in Tgr5-/- mice. Tgr5-/- mice had increased hepatic fatty acid oxidation rate and decreased hepatic fatty acid uptake. Interestingly, fasting induction of fibroblast growth factor 21 in liver was attenuated. In addition, fasted Tgr5-/- mice had increased activation of hepatic growth hormone-signal transducer and activator of transcription 5 (GH-Stat5) signaling compared to wild-type mice. CONCLUSION: TGR5 may play a role in determining bile acid composition and in fasting-induced hepatic steatosis through a novel mechanism involving activation of the GH-Stat5 signaling pathway. (Hepatology 2017;65:813-827).


Subject(s)
Bile Acids and Salts/metabolism , Fatty Liver/metabolism , Gene Expression Regulation , RNA-Binding Proteins/metabolism , Receptors, G-Protein-Coupled/genetics , Analysis of Variance , Animals , Disease Models, Animal , Energy Metabolism/physiology , Fasting , Fatty Liver/pathology , Homeostasis/genetics , Lipid Metabolism/genetics , Male , Mice , Mice, Inbred C57BL , Oxygen Consumption/physiology , Random Allocation , Signal Transduction
11.
Gene Expr ; 18(2): 71-87, 2018 05 18.
Article in English | MEDLINE | ID: mdl-29325602

ABSTRACT

Bile acids facilitate intestinal nutrient absorption and biliary cholesterol secretion to maintain bile acid homeostasis, which is essential for protecting liver and other tissues and cells from cholesterol and bile acid toxicity. Bile acid metabolism is tightly regulated by bile acid synthesis in the liver and bile acid biotransformation in the intestine. Bile acids are endogenous ligands that activate a complex network of nuclear receptor farnesoid X receptor and membrane G protein-coupled bile acid receptor-1 to regulate hepatic lipid and glucose metabolic homeostasis and energy metabolism. The gut-to-liver axis plays a critical role in the regulation of enterohepatic circulation of bile acids, bile acid pool size, and bile acid composition. Bile acids control gut bacteria overgrowth, and gut bacteria metabolize bile acids to regulate host metabolism. Alteration of bile acid metabolism by high-fat diets, sleep disruption, alcohol, and drugs reshapes gut microbiome and causes dysbiosis, obesity, and metabolic disorders. Gender differences in bile acid metabolism, FXR signaling, and gut microbiota have been linked to higher prevalence of fatty liver disease and hepatocellular carcinoma in males. Alteration of bile acid homeostasis contributes to cholestatic liver diseases, inflammatory diseases in the digestive system, obesity, and diabetes. Bile acid-activated receptors are potential therapeutic targets for developing drugs to treat metabolic disorders.


Subject(s)
Bile Acids and Salts/metabolism , Liver Diseases/etiology , Liver/metabolism , Animals , Circadian Rhythm , Gastrointestinal Microbiome , Humans , Liver/microbiology , Signal Transduction
12.
Dig Dis ; 35(3): 241-245, 2017.
Article in English | MEDLINE | ID: mdl-28249273

ABSTRACT

Bile acids play a critical role in the regulation of glucose, lipid and energy metabolisms by activating the nuclear bile acid receptor farnesoid X receptor (FXR) and membrane G protein-coupled bile acid receptor-1 (aka takeda G protein couple receptor 5, TGR5) signaling. Paradoxical roles of FXR in the regulation of glucose and lipid metabolism and metabolic disorder have been reported recently. The activation or inhibition of intestinal FXR signaling has been shown to improve insulin and glucose sensitivity and energy metabolism to prevent diabetes, obesity and non-alcoholic fatty liver disease (NAFLD). TGR5 has an anti-inflammatory function in the intestine and stimulates glucagon-like peptide-1 (GLP-1) secretion in the intestine to stimulate insulin secretion from the pancreas. The role of TGR5 in metabolism and metabolic regulation is not clear and warrants further study. FXR and TGR5 are co-expressed in the ileum and colon. These 2 bile acid-activated receptors may cooperate to stimulate GLP-1 secretion and improve hepatic metabolism. FXR and TGR5 dual agonists may have therapeutic potential for treating diabetes and NAFLD.


Subject(s)
Intestinal Mucosa/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Animals , Bile Acids and Salts/biosynthesis , Bile Acids and Salts/metabolism , Humans , Liver/metabolism
13.
Pharmacol Rev ; 66(4): 948-83, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25073467

ABSTRACT

Bile acids are the end products of cholesterol catabolism. Hepatic bile acid synthesis accounts for a major fraction of daily cholesterol turnover in humans. Biliary secretion of bile acids generates bile flow and facilitates hepatobiliary secretion of lipids, lipophilic metabolites, and xenobiotics. In the intestine, bile acids are essential for the absorption, transport, and metabolism of dietary fats and lipid-soluble vitamins. Extensive research in the last 2 decades has unveiled new functions of bile acids as signaling molecules and metabolic integrators. The bile acid-activated nuclear receptors farnesoid X receptor, pregnane X receptor, constitutive androstane receptor, vitamin D receptor, and G protein-coupled bile acid receptor play critical roles in the regulation of lipid, glucose, and energy metabolism, inflammation, and drug metabolism and detoxification. Bile acid synthesis exhibits a strong diurnal rhythm, which is entrained by fasting and refeeding as well as nutrient status and plays an important role for maintaining metabolic homeostasis. Recent research revealed an interaction of liver bile acids and gut microbiota in the regulation of liver metabolism. Circadian disturbance and altered gut microbiota contribute to the pathogenesis of liver diseases, inflammatory bowel diseases, nonalcoholic fatty liver disease, diabetes, and obesity. Bile acids and their derivatives are potential therapeutic agents for treating metabolic diseases of the liver.


Subject(s)
Bile Acids and Salts/metabolism , Metabolic Diseases/drug therapy , Metabolic Diseases/metabolism , Animals , Bile Acids and Salts/biosynthesis , Bile Acids and Salts/therapeutic use , Circadian Rhythm/physiology , Glucose/metabolism , Humans , Lipid Metabolism/physiology , Liver/metabolism , MicroRNAs/metabolism , Microbiota/physiology , Models, Biological , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/physiology
14.
J Lipid Res ; 57(7): 1144-54, 2016 07.
Article in English | MEDLINE | ID: mdl-27146480

ABSTRACT

Cholesterol 7α-hydroxylase (CYP7A1) is the first and rate-limiting enzyme in the conversion of cholesterol to bile acids in the liver. In addition to absorption and digestion of nutrients, bile acids play a critical role in the regulation of lipid, glucose, and energy homeostasis. We have backcrossed Cyp7a1(-/-) mice in a mixed B6/129Sv genetic background to C57BL/6J mice to generate Cyp7a1(-/-) mice in a near-pure C57BL/6J background. These mice survive well and have normal growth and a bile acid pool size ∼60% of WT mice. The expression of the genes in the alternative bile acid synthesis pathway are upregulated, resulting in a more hydrophilic bile acid composition with reduced cholic acid (CA). Surprisingly, Cyp7a1(-/-) mice have improved glucose sensitivity with reduced liver triglycerides and fecal bile acid excretion, but increased fecal fatty acid excretion and respiratory exchange ratio (RER) when fed a high-fat/high-cholesterol diet. Supplementing chow and Western diets with CA restored bile acid composition, reversed the glucose tolerant phenotype, and reduced the RER. Our current study points to a critical role of bile acid composition, rather than bile acid pool size, in regulation of glucose, lipid, and energy metabolism to improve glucose and insulin tolerance, maintain metabolic homeostasis, and prevent high-fat diet-induced metabolic disorders.


Subject(s)
Cholesterol 7-alpha-Hydroxylase/genetics , Cholesterol/metabolism , Metabolic Diseases/genetics , Animals , Bile Acids and Salts/genetics , Bile Acids and Salts/metabolism , Cholesterol 7-alpha-Hydroxylase/metabolism , Diet, High-Fat , Disease Models, Animal , Exhalation/genetics , Glucose/metabolism , Homeostasis , Humans , Lipid Metabolism/genetics , Liver/enzymology , Liver/pathology , Metabolic Diseases/metabolism , Mice
15.
Biochim Biophys Acta ; 1851(1): 19-29, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24796972

ABSTRACT

Bile acid synthesis is the major pathway for catabolism of cholesterol. Cholesterol 7α-hydroxylase (CYP7A1) is the rate-limiting enzyme in the bile acid biosynthetic pathway in the liver and plays an important role in regulating lipid, glucose and energy metabolism. Transgenic mice overexpressing CYP7A1 (CYP7A1-tg mice) were resistant to high-fat diet (HFD)-induced obesity, fatty liver, and diabetes. However the mechanism of resistance to HFD-induced obesity of CYP7A1-tg mice has not been determined. In this study, metabolomic and lipidomic profiles of CYP7A1-tg mice were analyzed to explore the metabolic alterations in CYP7A1-tg mice that govern the protection against obesity and insulin resistance by using ultra-performance liquid chromatography-coupled with electrospray ionization quadrupole time-of-flight mass spectrometry combined with multivariate analyses. Lipidomics analysis identified seven lipid markers including lysophosphatidylcholines, phosphatidylcholines, sphingomyelins and ceramides that were significantly decreased in serum of HFD-fed CYP7A1-tg mice. Metabolomics analysis identified 13 metabolites in bile acid synthesis including taurochenodeoxycholic acid, taurodeoxycholic acid, tauroursodeoxycholic acid, taurocholic acid, and tauro-ß-muricholic acid (T-ß-MCA) that differed between CYP7A1-tg and wild-type mice. Notably, T-ß-MCA, an antagonist of the farnesoid X receptor (FXR) was significantly increased in intestine of CYP7A1-tg mice. This study suggests that reducing 12α-hydroxylated bile acids and increasing intestinal T-ß-MCA may reduce high fat diet-induced increase of phospholipids, sphingomyelins and ceramides, and ameliorate diabetes and obesity. This article is part of a Special Issue entitled Linking transcription to physiology in lipodomics.


Subject(s)
Bile Acids and Salts/metabolism , Diabetes Mellitus/metabolism , Lipid Metabolism/physiology , Metabolome/genetics , Obesity/metabolism , Animals , Bile Acids and Salts/genetics , Cholesterol 7-alpha-Hydroxylase/genetics , Cholesterol 7-alpha-Hydroxylase/metabolism , Diabetes Mellitus/genetics , Diet, High-Fat/methods , Female , Glucose/genetics , Glucose/metabolism , Homeostasis , Insulin Resistance , Intestinal Mucosa/metabolism , Liver/metabolism , Male , Metabolomics/methods , Mice , Mice, Inbred C57BL , Mice, Transgenic , Obesity/genetics , Rats , Signal Transduction , Taurocholic Acid/analogs & derivatives , Taurocholic Acid/genetics , Taurocholic Acid/metabolism
16.
Biochem J ; 470(2): 181-93, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26348907

ABSTRACT

Bile acids are primarily synthesized from cholesterol in the liver and have important roles in dietary lipid absorption and cholesterol homoeostasis. Detailed roles of the orphan nuclear receptors regulating cholesterol 7α-hydroxylase (CYP7A1), the rate-limiting enzyme in bile acid synthesis, have not yet been fully elucidated. In the present study, we report that oestrogen-related receptor γ (ERRγ) is a novel transcriptional regulator of CYP7A1 expression. Activation of cannabinoid receptor type 1 (CB1 receptor) signalling induced ERRγ-mediated transcription of the CYP7A1 gene. Overexpression of ERRγ increased CYP7A1 expression in vitro and in vivo, whereas knockdown of ERRγ attenuated CYP7A1 expression. Deletion analysis of the CYP7A1 gene promoter and a ChIP assay revealed an ERRγ-binding site on the CYP7A1 gene promoter. Small heterodimer partner (SHP) inhibited the transcriptional activity of ERRγ and thus regulated CYP7A1 expression. Overexpression of ERRγ led to increased bile acid levels, whereas an inverse agonist of ERRγ, GSK5182, reduced CYP7A1 expression and bile acid synthesis. Finally, GSK5182 significantly reduced hepatic CB1 receptor-mediated induction of CYP7A1 expression and bile acid synthesis in alcohol-treated mice. These results provide the molecular mechanism linking ERRγ and bile acid metabolism.


Subject(s)
Cholesterol 7-alpha-Hydroxylase/biosynthesis , Liver/metabolism , Receptor, Cannabinoid, CB1/metabolism , Receptors, Estrogen/metabolism , Animals , Bile Acids and Salts/metabolism , Cells, Cultured , Cholesterol 7-alpha-Hydroxylase/genetics , Drug Inverse Agonism , Ethanol/pharmacology , Gene Expression , Glycerides/pharmacology , HEK293 Cells , Hepatocytes/metabolism , Humans , Mice, Inbred C57BL , Mice, Knockout , Promoter Regions, Genetic , Rats, Sprague-Dawley , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Estrogen/genetics , Transcription, Genetic
18.
Hepatology ; 59(5): 1750-60, 2014 May.
Article in English | MEDLINE | ID: mdl-24038081

ABSTRACT

UNLABELLED: Mice deficient in small heterodimer partner (SHP) are protected from diet-induced hepatic steatosis resulting from increased fatty acid oxidation and decreased lipogenesis. The decreased lipogenesis appears to be a direct consequence of very low expression of peroxisome proliferator-activated receptor gamma 2 (PPAR-γ2), a potent lipogenic transcription factor, in the SHP(-/-) liver. The current study focused on the identification of a SHP-dependent regulatory cascade that controls PPAR-γ2 gene expression, thereby regulating hepatic fat accumulation. Illumina BeadChip array (Illumina, Inc., San Diego, CA) and real-time polymerase chain reaction were used to identify genes responsible for the linkage between SHP and PPAR-γ2 using hepatic RNAs isolated from SHP(-/-) and SHP-overexpressing mice. The initial efforts identify that hairy and enhancer of split 6 (Hes6), a novel transcriptional repressor, is an important mediator of the regulation of PPAR-γ2 transcription by SHP. The Hes6 promoter is specifically activated by the retinoic acid receptor (RAR) in response to its natural agonist ligand, all-trans retinoic acid (atRA), and is repressed by SHP. Hes6 subsequently represses hepatocyte nuclear factor 4 alpha (HNF-4α)-activated PPAR-γ2 gene expression by direct inhibition of HNF-4α transcriptional activity. Furthermore, we provide evidences that atRA treatment or adenovirus-mediated RAR-α overexpression significantly reduced hepatic fat accumulation in obese mouse models, as observed in earlier studies, and the beneficial effect is achieved by the proposed transcriptional cascade. CONCLUSIONS: Our study describes a novel transcriptional regulatory cascade controlling hepatic lipid metabolism that identifies retinoic acid signaling as a new therapeutic approach to nonalcoholic fatty liver diseases.


Subject(s)
Fatty Liver/drug therapy , PPAR gamma/genetics , Receptors, Cytoplasmic and Nuclear/physiology , Tretinoin/therapeutic use , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Blood Glucose/analysis , Fatty Liver/metabolism , Gene Expression Regulation , Lipid Metabolism , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease , Receptors, Retinoic Acid/physiology , Repressor Proteins/genetics , Retinoic Acid Receptor alpha , Transcription, Genetic , Tretinoin/pharmacology
19.
Curr Opin Gastroenterol ; 31(2): 159-65, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25584736

ABSTRACT

PURPOSE OF REVIEW: This review focuses on the latest understanding of the molecular mechanisms underlying the complex interactions between intestine and liver bile acid signaling, gut microbiota, and their impact on whole-body lipid, glucose and energy metabolism. RECENT FINDINGS: Hepatic bile acid synthesis is tightly regulated by the bile acid negative feedback mechanisms. Modulating the enterohepatic bile acid signaling greatly impacts the whole-body metabolic homeostasis. Recently, a positive feedback mechanism through intestine farnesoid X receptor (FXR) antagonism has been proposed to link gut microbiota to the regulation of bile acid composition and pool size. Two studies identified intestine Diet1 and hepatic SHP-2 as novel regulators of CYP7A1 and bile acid synthesis through the gut-liver FXR-fibroblast growth factor 15/19-FGF receptor four signaling axis. New evidence suggests that enhancing bile acid signaling in the distal ileum and colon contributes to the metabolic benefits of bile acid sequestrants and bariatric surgery. SUMMARY: Small-molecule ligands that target TGR5 and FXR have shown promise in treating various metabolic and inflammation-related human diseases. New insights into the mechanisms underlying the bariatric surgery and bile acid sequestrant treatment suggest that targeting the enterohepatic circulation to modulate gut-liver bile acid signaling, incretin production and microbiota represents a new strategy to treat obesity and type 2 diabetes.


Subject(s)
Bile Acids and Salts/metabolism , Diabetes Mellitus, Type 2/metabolism , Inflammation/metabolism , Intestine, Small/metabolism , Liver/metabolism , Obesity/metabolism , Biological Transport , Cholesterol 7-alpha-Hydroxylase/metabolism , Energy Metabolism , Homeostasis , Humans , Intestine, Small/pathology , Liver/pathology , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction
20.
J Lipid Res ; 55(3): 455-65, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24343899

ABSTRACT

Vitamin D receptor (VDR) mediates vitamin D signaling involved in bone metabolism, cellular growth and differentiation, cardiovascular function, and bile acid regulation. Mice with an intestine-specific disruption of VDR (Vdr(ΔIEpC)) have abnormal body size, colon structure, and imbalance of bile acid metabolism. Lithocholic acid (LCA), a secondary bile acid that activates VDR, is among the most toxic of the bile acids that when overaccumulated in the liver causes hepatotoxicity. Because cytochrome P450 3A4 (CYP3A4) is a target gene of VDR-involved bile acid metabolism, the role of CYP3A4 in VDR biology and bile acid metabolism was investigated. The CYP3A4 gene was inserted into Vdr(ΔIEpC) mice to produce the Vdr(ΔIEpC)/3A4 line. LCA was administered to control, transgenic-CYP3A4, Vdr(ΔIEpC), and Vdr(ΔIEpC)/3A4 mice, and hepatic toxicity and bile acid levels in the liver, intestine, bile, and urine were measured. VDR deficiency in the intestine of the Vdr(ΔIEpC) mice exacerbates LCA-induced hepatotoxicity manifested by increased necrosis and inflammation, due in part to over-accumulation of hepatic bile acids including taurocholic acid and taurodeoxycholic acid. Intestinal expression of CYP3A4 in the Vdr(ΔIEpC)/3A4 mouse line reduces LCA-induced hepatotoxicity through elevation of LCA metabolism and detoxification, and suppression of bile acid transporter expression in the small intestine. This study reveals that intestinal CYP3A4 protects against LCA hepatotoxicity.


Subject(s)
Chemical and Drug Induced Liver Injury/metabolism , Cytochrome P-450 CYP3A/metabolism , Intestinal Mucosa/metabolism , Receptors, Calcitriol/deficiency , Animals , Bile/metabolism , Bile Acids and Salts/metabolism , Blotting, Western , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/genetics , Cholesterol/blood , Cholesterol/metabolism , Cytochrome P-450 CYP3A/genetics , Cytochrome P-450 CYP3A/urine , Gallbladder/metabolism , Lithocholic Acid , Liver/metabolism , Liver/pathology , Male , Metabolomics/methods , Mice , Mice, Knockout , Mice, Transgenic , Receptors, Calcitriol/genetics , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL