Comprehensive Physiology Wiley Online Library

Bile Acid Metabolism and Signaling

Full Article on Wiley Online Library



Abstract

Bile acids are important physiological agents for intestinal nutrient absorption and biliary secretion of lipids, toxic metabolites, and xenobiotics. Bile acids also are signaling molecules and metabolic regulators that activate nuclear receptors and G protein‐coupled receptor (GPCR) signaling to regulate hepatic lipid, glucose, and energy homeostasis and maintain metabolic homeostasis. Conversion of cholesterol to bile acids is critical for maintaining cholesterol homeostasis and preventing accumulation of cholesterol, triglycerides, and toxic metabolites, and injury in the liver and other organs. Enterohepatic circulation of bile acids from the liver to intestine and back to the liver plays a central role in nutrient absorption and distribution, and metabolic regulation and homeostasis. This physiological process is regulated by a complex membrane transport system in the liver and intestine regulated by nuclear receptors. Toxic bile acids may cause inflammation, apoptosis, and cell death. On the other hand, bile acid‐activated nuclear and GPCR signaling protects against inflammation in liver, intestine, and macrophages. Disorders in bile acid metabolism cause cholestatic liver diseases, dyslipidemia, fatty liver diseases, cardiovascular diseases, and diabetes. Bile acids, bile acid derivatives, and bile acid sequestrants are therapeutic agents for treating chronic liver diseases, obesity, and diabetes in humans. © 2013 American Physiological Society. Compr Physiol 3:1191‐1212, 2013.

Comprehensive Physiology offers downloadable PowerPoint presentations of figures for non-profit, educational use, provided the content is not modified and full credit is given to the author and publication.

Download a PowerPoint presentation of all images


Figure 1. Figure 1.

Bile acid biosynthetic pathways. Two major bile acid biosynthetic pathways are shown. The neutral (or classic) pathway is initiated by cholesterol 7α‐hydroxylase (CYP7A1) located in the endoplasmic reticulum of the liver, whereas the acidic (or alternative) pathway is initiated by mitochondrial sterol 27‐hydroxylase (CYP27A1). There are three sterol hydroxylases that convert cholesterol to oxysterols: CYP27A1 in macrophages and other tissues, microsomal sterol 25‐hydroxylase in liver microsomes, and sterol 24‐hydroxylase (CYP46A1) in the brain. Oxysterol 7α‐hydroxylase (CYP7B1) is nonspecific and catalyzes hydroxylation of 27‐ and 25‐hydroxycholesterol to 3β, 7α‐dihydroxy‐5‐cholestenoic acid and 5‐cholesten‐3β, 7α, 25‐triol, respectively. A brain‐specific oxysterol 7α‐hydroxylase (CYP39A1) catalyzes hydroxylation of 24‐hydroxycholesterol to 5‐cholesten‐3β, 7α, 24(S)‐triol. These oxysterols could be converted to CDCA if transported to the liver. In the liver, 3β‐hydoxysteroid dehydrogenase (3βHSD, HSD3B7) convert 7α‐hydroxycholesterol to 7α‐hydroxy‐4‐cholesten‐3‐one (C4), which is converted to 7α, 12α‐dihydroxy‐4‐cholesten‐3‐one by a sterol 12α‐hydroxylase (CYP8B1), leading to synthesis of cholic acid (CA). Without 12α‐hydroxylation, the pathway produces CDCA. Aldos‐keto reductase 1D1 (AKR1D1) and AKR1C1 catalyze isomerization and saturation of the steroid ring. Then CYP27A1 catalyzes steroid side‐chain oxidation to form cholestanoic acids, THCA, and DHCA. Bile acid‐Co‐A synthase (BACS) or very long‐chain Co‐A synthase (VLCS) in the endoplasmic reticulum (ER) ligates Co‐A to the carboxyl groups. Bile acid thioesters are transported into peroxisomes, where an α‐methylacyl‐CoA racemase (AMACR) converts the methyl group from 25(R) to 25(S) conformation, and three peroxisomal β‐oxidation enzymes, branched‐chain acyl‐CoA oxidase, D‐bifunctional enzyme, and thiolase (or sterol carrier protein x) catalyze oxidative cleavage of a propionyl group from the steroid side‐chain to form cholyl‐CoA and chenodeoxycholyl‐CoA. Cytosolic or peroxisomal bile acid: amino‐acid transferase (BAAT) catalyzes conjugation of amino acids, glycine or taurine to the carboxyl group of cholyl‐CoA and chenodeoxycholyl‐CoA to form tauro‐ or glycol‐conjugated CA or CDCA. ER: endoplasmic reticulum.

Figure 2. Figure 2.

Enterohepatic circulation of bile acids. An average man produces ∼0.5g bile acid per day by synthesis in the liver, and secretes ∼0.5g/day. This daily turnover of bile acids accounts for about 5% of total bile acid pool. The remaining 95% of bile acids in the pool are recycled 4 to 12 times a day. Most bile acids are reabsorbed in the ileum by active transport, while a small amount is reabsorbed by passive diffusion in the upper intestine to portal blood for circulation to the liver. Small amounts of bile acids spilled over into the systemic circulation are recovered in kidney.

Figure 3. Figure 3.

Bile acid synthesis regulates cholesterol homeostasis in hepatocytes. Cholesterol homeostasis is maintained by dietary uptake of cholesterol, de novo cholesterol synthesis from acetyl‐CoA, and conversion of cholesterol to bile acids. Oxysterols are derived from cholesterol and bile acids. When intracellular cholesterol/oxysterol levels are high, steroid response element binding protein 2 (SREBP‐2) precursor (125 kDa) interacts with insulin induced gene 1/2 (Insig1/2) and is retained in endoplasmic reticulum (ER) membrane. When intracellular oxysterol levels are low, SREBP cleavage and activating protein (SCAP) escorts SREBP‐2 precursor to the Golgi apparatus, where sterol sensitive proteases S1P and S2P are activated to cleave a N‐terminal fragment (65 kDa), which is translocated to the nucleus to bind to the steroid response elements in the gene promoters of all cholesterogenic genes and stimulates de novo cholesterol synthesis. Oxysterols activate LXRα, which induces CYP7A1 gene transcription to stimulate bile acid synthesis in mice, but not humans. Bile acids (CDCA) activate farnesoid X receptor (FXR) to inhibit CYP7A1 gene transcription and bile acid synthesis. This may lead to increased cholesterol levels and inhibited de novo cholesterol synthesis and absorption of dietary cholesterol.

Figure 4. Figure 4.

Nuclear receptors. The general structure of nuclear receptors is shown on the top. The NR1 family of genes involved in metabolic regulation, and their respective endogenous ligands are shown. The putative nuclear receptor response element binding sequence, arranged in direct repeat (DR), everted repeat (ER), and inverted repeat (IR), is shown. Ligand‐activated receptors recruit coactivators to replace corepressors and results in transactivation of target gene expression. AF‐1‐2, activation function‐1 and ‐2; CAR, constitutive androstane receptor; FXR, farnesoid X receptor; LXR, liver orphan receptor; PPAR, peroxisome proliferator‐activated receptor; PXR, pregnane X receptor; NLS, nuclear localization sequence.

Figure 5. Figure 5.

Farnesoid X receptor (FXR) regulates enterohepatic circulation of bile acids. Major bile acid transporters in human hepatocytes and enterocytes are shown. Enzymes and transporters regulated by FXR are indicated. In hepatocytes, bile acids activate FXR to inhibit CYP7A1 gene transcription by two pathways: (i) FXR induces small heterodimer partner (SHP), which inhibits CYP7A1 by inhibiting nuclear receptors liver related homologue‐1 (LRH‐1) or hepatocyte nuclear factor (HNF4), which bind to the CYP7A1 promoter. (ii) In enterocytes, FXR induces intestinal hormone fibroblast growth factor 19 (FGF19), which is circulated to hepatocytes to activate FGF receptor 4 (FGFR4) signaling to inhibit CYP7A1 via activation of the extracellular stress‐activated receptor kinase 1/2 (ERK1/2) pathway. FXR induces bile salt expert pump (BSEP) to efflux bile acids into bile; multidrug resistance protein 2/3 (MDR2/3) to efflux phosphatidylcholine (PC) to bile; and MDR related protein 2 (MRP2) to efflux organic anions including glucuronidated‐ and sulfated‐bile acids, organic anions, and drugs into bile. Bile acids also facilitate efflux of cholesterol to bile by ATP binding casette G5/G8(ABCG5/G8). In the bile, bile acids, PC, and cholesterol form mixed micelles, which are stored in the gallbladder. In the brush border membrane of the ileum, bile acids are reabsorbed by the apical sodium bile salt transporter (ASBT). In enterocytes, bile acids activate FXR, which induces ileum bile acid binding protein (IBABP) to bind bile salts and may facilitate intracellular transport of bile acids to organic solute transporter α/β (OSTα/β) located in the basolateral membrane for efflux of bile acids into portal circulation. Bile acids in portal blood are reabsorbed into hepatocytes by Na+‐dependent taurocholate cotransport peptide (NTCP). FXR inhibits NTCP transcription as a feedback inhibition of bile acid uptake to prevent liver injury. In the sinusoidal membrane of enterocytes and hepatocytes, FXR also induces MRP3/4 to efflux bile acids as an adaptive response to cholestasis. In hepatocytes, FXR also induces OSTα/β to efflux bile acids into sinusoidal blood to prevent bile acid accumulation in hepatocytes. MRP3 may be induced by FXR as an adaptive response to cholestasis. Bile acids returned to hepatic sinusoid are also taken up by Na+‐independent organic anion transport proteins (OATP2). Many of these membrane transporters (ASBT, OSTα/β, and MRP2/3) also are present in cholangiocytes for reabsorption of bile acids, and in renal proximal tubule cells for reabsorption of bile acids from blood circulation and excretion of hydrophilic bile acids.

Figure 6. Figure 6.

Bile acid‐activated TGR5 signaling. TGR5 is expressed in brown adipocytes, macrophages/monocytes and hepatic Kupffer cells, gallbladder epithelium, and intestine, with especially high levels in the colon. TGR5 is the first G protein‐coupled receptor (GPCR) identified as a bile acid‐activated membrane receptor. TGR5 is a Gαs GPCR activated by secondary bile acids, lithocholic acid (LCA) and TLCA to induce cAMP signaling through activation of adenylyl cyclase (AC). TGR5 signaling may increase insulin sensitivity through two mechanisms. (i) In brown adipose tissue, cAMP induces type 2 deiodinase (DIO2), which converts and activates thyroid hormone T4 to T3 to stimulate energy metabolism in mitochondria by activating oxidative phosphorylation (OXphor) and uncoupling protein‐1 (UCP‐1). (ii) In the intestine, cAMP stimulates glucagon like peptide‐1 (GLP‐1) in L cells, which stimulates insulin secretion in the pancreas. TGR5 also has anti‐inflammatory functions by antagonizing of TNFα and NF‐κB‐dependent induction of proinflammatory cytokines in intestine and macrophages, thus, protecting against colitis, inflammatory bowel disease and Crohn's disease, and also atherosclerosis.

Figure 7. Figure 7.

Bile acid‐activated S1P2 signaling. Tauro‐conjugated bile acids stimulates sphingosine‐1‐phosphate receptor 2 (S1P2), a Gαi protein‐coupled receptor, which activates the extracellular signal‐regulated kinase 1/2 (ERK1/2) and AKT pathways. S1P2 signaling activates the insulin receptor/AKT pathway through activation of Src and epidermal growth factor receptors (EGFRs) leading to activation of the insulin receptor (IR), which phosphorylates and activates insulin receptor substrate‐1 (IRS‐1). IRS‐1 phosphorylates phosphatidylinositol 3‐kinase (PI3K), which phosphorylates phosphatidylinositol 4′, 5′ bis‐phosphate to phosphatidylinositol 3′, 4′, 5′‐trisphosphate (PIP3). PIP3 then ohosphorylates pyruvate dehydrogenase kinase‐1 (PDK‐1) to phosphorylate and activate AKT (also know as PKB). Phosphorylated AKT phosphorylates and inactivates glycogen synthase kinase 3β (GSK3β) and resulting in dephosphorylation and activation of glycogen synthase (GS), a key enzyme in glycogenesis. AKT also phosphorylates and inactivates a transcription factor, FoxO1 and resulted in inhibiting phosphoenolpyruvate carboxykinase (PEPCK) and glucose‐6‐phosphatase (G6Pase) in gluconeogenesis. S1P2 signaling also may activate FXR/SHP pathway to inhibit peroxisome proliferator‐activated receptor αγ (PPARα/γ)/PGC‐1α‐mediated fatty acid oxidation and steroid response element binding protein 1c (SREBP‐1c)‐mediated fatty acid synthesis. Therefore, SiP2 signaling may reduce serum glucose and triglycerides, and improve insulin sensitivity by stimulating glycogensis and inhibiting gluconeogenesis and lipogenesis.

Figure 8. Figure 8.

Farnesoid X receptor (FXR) regulation of hepatic glucose and lipid metabolism in liver, adipocytes, and intestine. Glucose and insulin stimulate glycolysis to form acetyl‐CoA, which is a precursor of cholesterol and fatty acids. The FXR/small heterodimer partner (SHP) pathway may inhibit steroid response element binding protein 1c (SREBP‐1c), which induces all genes involved in lipogenesis, acetyl CoA carboxylase (ACC), fatty acid synthase (FAS), and stearoyl CoA desaturase (SCD). The FXR/SHP pathway also inhibits SREBP‐2, which induces all genes in de novo cholesterol synthesis. FXR activates mitochondria fatty acid β‐oxidation by inducing peroxisome proliferator‐activated receptor α (PPARα). FXR inhibits PCSK9, which is an inhibitor of LDL receptor. Thus FXR induces LDL‐R and also syndecan‐1 involved in cholesterol uptake. FXR inhibits mitochondria triglyceride transport protein (MTP), which is required for assembly of VLDL particles with ApoB100. FXR also induces ApoE, which is a high affinity ligand of LDL receptor and scavenger receptor B1 (SR‐B1), and a component of VLDL and chylomicron remnants, which are taken up by ApoE receptors. FXR also induces phospholipid transport protein (PLTP) involved in reverse cholesterol transport of cholesterol from peripheral tissues to liver by HDL/SR‐B1 receptor‐mediated mechanisms. On the other hand, FXR inhibits ApoA1, a component of HDL, and ANGPTL3, which is involved in hydrolysis of triglycerides in liver and adipocytes. FXR induces FGF19 synthesis in the intestine, which activates FGFR4 receptor in hepatocytes to activate ERK1/2 signaling to inhibit CYP7A1 and bile acid synthesis. In colon, bile acids activate TGR5 signaling to stimulate GLP‐1 release. GLP‐1 increases insulin sensitivity. TGR5 in brown adipocytes stimulates cAMP production, which induce deiodinase 2 (DIO2) to convert T4 to T3, which stimulates mitochondrial energy metabolism via activation of PGC‐1α.

Figure 9. Figure 9.

FXR regulation of hepatic glucose metabolism. FXR signaling phosphorylates and inhibits glycogen synthase kinase 3β (GSK3β), which is an inhibitor of glycogen synthase activity. This results in stimulating glycogenesis. The FXR/SHP pathway inhibits PEPCK and glucose 6‐phosphatase (G6Pase) to inhibit gluconeogenesis. This results in increasing glucose tolerance and insulin sensitivity.

Figure 10. Figure 10.

Mechanism of nonalcoholic fatty liver disease (NAFLD). Metabolic syndrome is a constellation of five clinical symptoms, hypertension, hyperglycemia, hypertriglyceridemia, insulin resistance, and obesity. Metabolic syndrome is linked to cardiovascular disease, type II diabetes, and NAFLD. NAFLD is progressed to nonalcoholic steatohepatitis (NASH) by many factors. The first hit is high fat diet (HFD)‐induced hepatic steatosis, followed by inflammation involving reactive oxidizing species, drugs, and endoplasmid reticulum stress to NASH. NASH patients have prevalence for liver fibrosis and cirrhosis, while NAFLD is linked to obesity, steatosis, and insulin resistance. HFD, high fat diet; ROS, reactive oxidizing species.



Figure 1.

Bile acid biosynthetic pathways. Two major bile acid biosynthetic pathways are shown. The neutral (or classic) pathway is initiated by cholesterol 7α‐hydroxylase (CYP7A1) located in the endoplasmic reticulum of the liver, whereas the acidic (or alternative) pathway is initiated by mitochondrial sterol 27‐hydroxylase (CYP27A1). There are three sterol hydroxylases that convert cholesterol to oxysterols: CYP27A1 in macrophages and other tissues, microsomal sterol 25‐hydroxylase in liver microsomes, and sterol 24‐hydroxylase (CYP46A1) in the brain. Oxysterol 7α‐hydroxylase (CYP7B1) is nonspecific and catalyzes hydroxylation of 27‐ and 25‐hydroxycholesterol to 3β, 7α‐dihydroxy‐5‐cholestenoic acid and 5‐cholesten‐3β, 7α, 25‐triol, respectively. A brain‐specific oxysterol 7α‐hydroxylase (CYP39A1) catalyzes hydroxylation of 24‐hydroxycholesterol to 5‐cholesten‐3β, 7α, 24(S)‐triol. These oxysterols could be converted to CDCA if transported to the liver. In the liver, 3β‐hydoxysteroid dehydrogenase (3βHSD, HSD3B7) convert 7α‐hydroxycholesterol to 7α‐hydroxy‐4‐cholesten‐3‐one (C4), which is converted to 7α, 12α‐dihydroxy‐4‐cholesten‐3‐one by a sterol 12α‐hydroxylase (CYP8B1), leading to synthesis of cholic acid (CA). Without 12α‐hydroxylation, the pathway produces CDCA. Aldos‐keto reductase 1D1 (AKR1D1) and AKR1C1 catalyze isomerization and saturation of the steroid ring. Then CYP27A1 catalyzes steroid side‐chain oxidation to form cholestanoic acids, THCA, and DHCA. Bile acid‐Co‐A synthase (BACS) or very long‐chain Co‐A synthase (VLCS) in the endoplasmic reticulum (ER) ligates Co‐A to the carboxyl groups. Bile acid thioesters are transported into peroxisomes, where an α‐methylacyl‐CoA racemase (AMACR) converts the methyl group from 25(R) to 25(S) conformation, and three peroxisomal β‐oxidation enzymes, branched‐chain acyl‐CoA oxidase, D‐bifunctional enzyme, and thiolase (or sterol carrier protein x) catalyze oxidative cleavage of a propionyl group from the steroid side‐chain to form cholyl‐CoA and chenodeoxycholyl‐CoA. Cytosolic or peroxisomal bile acid: amino‐acid transferase (BAAT) catalyzes conjugation of amino acids, glycine or taurine to the carboxyl group of cholyl‐CoA and chenodeoxycholyl‐CoA to form tauro‐ or glycol‐conjugated CA or CDCA. ER: endoplasmic reticulum.



Figure 2.

Enterohepatic circulation of bile acids. An average man produces ∼0.5g bile acid per day by synthesis in the liver, and secretes ∼0.5g/day. This daily turnover of bile acids accounts for about 5% of total bile acid pool. The remaining 95% of bile acids in the pool are recycled 4 to 12 times a day. Most bile acids are reabsorbed in the ileum by active transport, while a small amount is reabsorbed by passive diffusion in the upper intestine to portal blood for circulation to the liver. Small amounts of bile acids spilled over into the systemic circulation are recovered in kidney.



Figure 3.

Bile acid synthesis regulates cholesterol homeostasis in hepatocytes. Cholesterol homeostasis is maintained by dietary uptake of cholesterol, de novo cholesterol synthesis from acetyl‐CoA, and conversion of cholesterol to bile acids. Oxysterols are derived from cholesterol and bile acids. When intracellular cholesterol/oxysterol levels are high, steroid response element binding protein 2 (SREBP‐2) precursor (125 kDa) interacts with insulin induced gene 1/2 (Insig1/2) and is retained in endoplasmic reticulum (ER) membrane. When intracellular oxysterol levels are low, SREBP cleavage and activating protein (SCAP) escorts SREBP‐2 precursor to the Golgi apparatus, where sterol sensitive proteases S1P and S2P are activated to cleave a N‐terminal fragment (65 kDa), which is translocated to the nucleus to bind to the steroid response elements in the gene promoters of all cholesterogenic genes and stimulates de novo cholesterol synthesis. Oxysterols activate LXRα, which induces CYP7A1 gene transcription to stimulate bile acid synthesis in mice, but not humans. Bile acids (CDCA) activate farnesoid X receptor (FXR) to inhibit CYP7A1 gene transcription and bile acid synthesis. This may lead to increased cholesterol levels and inhibited de novo cholesterol synthesis and absorption of dietary cholesterol.



Figure 4.

Nuclear receptors. The general structure of nuclear receptors is shown on the top. The NR1 family of genes involved in metabolic regulation, and their respective endogenous ligands are shown. The putative nuclear receptor response element binding sequence, arranged in direct repeat (DR), everted repeat (ER), and inverted repeat (IR), is shown. Ligand‐activated receptors recruit coactivators to replace corepressors and results in transactivation of target gene expression. AF‐1‐2, activation function‐1 and ‐2; CAR, constitutive androstane receptor; FXR, farnesoid X receptor; LXR, liver orphan receptor; PPAR, peroxisome proliferator‐activated receptor; PXR, pregnane X receptor; NLS, nuclear localization sequence.



Figure 5.

Farnesoid X receptor (FXR) regulates enterohepatic circulation of bile acids. Major bile acid transporters in human hepatocytes and enterocytes are shown. Enzymes and transporters regulated by FXR are indicated. In hepatocytes, bile acids activate FXR to inhibit CYP7A1 gene transcription by two pathways: (i) FXR induces small heterodimer partner (SHP), which inhibits CYP7A1 by inhibiting nuclear receptors liver related homologue‐1 (LRH‐1) or hepatocyte nuclear factor (HNF4), which bind to the CYP7A1 promoter. (ii) In enterocytes, FXR induces intestinal hormone fibroblast growth factor 19 (FGF19), which is circulated to hepatocytes to activate FGF receptor 4 (FGFR4) signaling to inhibit CYP7A1 via activation of the extracellular stress‐activated receptor kinase 1/2 (ERK1/2) pathway. FXR induces bile salt expert pump (BSEP) to efflux bile acids into bile; multidrug resistance protein 2/3 (MDR2/3) to efflux phosphatidylcholine (PC) to bile; and MDR related protein 2 (MRP2) to efflux organic anions including glucuronidated‐ and sulfated‐bile acids, organic anions, and drugs into bile. Bile acids also facilitate efflux of cholesterol to bile by ATP binding casette G5/G8(ABCG5/G8). In the bile, bile acids, PC, and cholesterol form mixed micelles, which are stored in the gallbladder. In the brush border membrane of the ileum, bile acids are reabsorbed by the apical sodium bile salt transporter (ASBT). In enterocytes, bile acids activate FXR, which induces ileum bile acid binding protein (IBABP) to bind bile salts and may facilitate intracellular transport of bile acids to organic solute transporter α/β (OSTα/β) located in the basolateral membrane for efflux of bile acids into portal circulation. Bile acids in portal blood are reabsorbed into hepatocytes by Na+‐dependent taurocholate cotransport peptide (NTCP). FXR inhibits NTCP transcription as a feedback inhibition of bile acid uptake to prevent liver injury. In the sinusoidal membrane of enterocytes and hepatocytes, FXR also induces MRP3/4 to efflux bile acids as an adaptive response to cholestasis. In hepatocytes, FXR also induces OSTα/β to efflux bile acids into sinusoidal blood to prevent bile acid accumulation in hepatocytes. MRP3 may be induced by FXR as an adaptive response to cholestasis. Bile acids returned to hepatic sinusoid are also taken up by Na+‐independent organic anion transport proteins (OATP2). Many of these membrane transporters (ASBT, OSTα/β, and MRP2/3) also are present in cholangiocytes for reabsorption of bile acids, and in renal proximal tubule cells for reabsorption of bile acids from blood circulation and excretion of hydrophilic bile acids.



Figure 6.

Bile acid‐activated TGR5 signaling. TGR5 is expressed in brown adipocytes, macrophages/monocytes and hepatic Kupffer cells, gallbladder epithelium, and intestine, with especially high levels in the colon. TGR5 is the first G protein‐coupled receptor (GPCR) identified as a bile acid‐activated membrane receptor. TGR5 is a Gαs GPCR activated by secondary bile acids, lithocholic acid (LCA) and TLCA to induce cAMP signaling through activation of adenylyl cyclase (AC). TGR5 signaling may increase insulin sensitivity through two mechanisms. (i) In brown adipose tissue, cAMP induces type 2 deiodinase (DIO2), which converts and activates thyroid hormone T4 to T3 to stimulate energy metabolism in mitochondria by activating oxidative phosphorylation (OXphor) and uncoupling protein‐1 (UCP‐1). (ii) In the intestine, cAMP stimulates glucagon like peptide‐1 (GLP‐1) in L cells, which stimulates insulin secretion in the pancreas. TGR5 also has anti‐inflammatory functions by antagonizing of TNFα and NF‐κB‐dependent induction of proinflammatory cytokines in intestine and macrophages, thus, protecting against colitis, inflammatory bowel disease and Crohn's disease, and also atherosclerosis.



Figure 7.

Bile acid‐activated S1P2 signaling. Tauro‐conjugated bile acids stimulates sphingosine‐1‐phosphate receptor 2 (S1P2), a Gαi protein‐coupled receptor, which activates the extracellular signal‐regulated kinase 1/2 (ERK1/2) and AKT pathways. S1P2 signaling activates the insulin receptor/AKT pathway through activation of Src and epidermal growth factor receptors (EGFRs) leading to activation of the insulin receptor (IR), which phosphorylates and activates insulin receptor substrate‐1 (IRS‐1). IRS‐1 phosphorylates phosphatidylinositol 3‐kinase (PI3K), which phosphorylates phosphatidylinositol 4′, 5′ bis‐phosphate to phosphatidylinositol 3′, 4′, 5′‐trisphosphate (PIP3). PIP3 then ohosphorylates pyruvate dehydrogenase kinase‐1 (PDK‐1) to phosphorylate and activate AKT (also know as PKB). Phosphorylated AKT phosphorylates and inactivates glycogen synthase kinase 3β (GSK3β) and resulting in dephosphorylation and activation of glycogen synthase (GS), a key enzyme in glycogenesis. AKT also phosphorylates and inactivates a transcription factor, FoxO1 and resulted in inhibiting phosphoenolpyruvate carboxykinase (PEPCK) and glucose‐6‐phosphatase (G6Pase) in gluconeogenesis. S1P2 signaling also may activate FXR/SHP pathway to inhibit peroxisome proliferator‐activated receptor αγ (PPARα/γ)/PGC‐1α‐mediated fatty acid oxidation and steroid response element binding protein 1c (SREBP‐1c)‐mediated fatty acid synthesis. Therefore, SiP2 signaling may reduce serum glucose and triglycerides, and improve insulin sensitivity by stimulating glycogensis and inhibiting gluconeogenesis and lipogenesis.



Figure 8.

Farnesoid X receptor (FXR) regulation of hepatic glucose and lipid metabolism in liver, adipocytes, and intestine. Glucose and insulin stimulate glycolysis to form acetyl‐CoA, which is a precursor of cholesterol and fatty acids. The FXR/small heterodimer partner (SHP) pathway may inhibit steroid response element binding protein 1c (SREBP‐1c), which induces all genes involved in lipogenesis, acetyl CoA carboxylase (ACC), fatty acid synthase (FAS), and stearoyl CoA desaturase (SCD). The FXR/SHP pathway also inhibits SREBP‐2, which induces all genes in de novo cholesterol synthesis. FXR activates mitochondria fatty acid β‐oxidation by inducing peroxisome proliferator‐activated receptor α (PPARα). FXR inhibits PCSK9, which is an inhibitor of LDL receptor. Thus FXR induces LDL‐R and also syndecan‐1 involved in cholesterol uptake. FXR inhibits mitochondria triglyceride transport protein (MTP), which is required for assembly of VLDL particles with ApoB100. FXR also induces ApoE, which is a high affinity ligand of LDL receptor and scavenger receptor B1 (SR‐B1), and a component of VLDL and chylomicron remnants, which are taken up by ApoE receptors. FXR also induces phospholipid transport protein (PLTP) involved in reverse cholesterol transport of cholesterol from peripheral tissues to liver by HDL/SR‐B1 receptor‐mediated mechanisms. On the other hand, FXR inhibits ApoA1, a component of HDL, and ANGPTL3, which is involved in hydrolysis of triglycerides in liver and adipocytes. FXR induces FGF19 synthesis in the intestine, which activates FGFR4 receptor in hepatocytes to activate ERK1/2 signaling to inhibit CYP7A1 and bile acid synthesis. In colon, bile acids activate TGR5 signaling to stimulate GLP‐1 release. GLP‐1 increases insulin sensitivity. TGR5 in brown adipocytes stimulates cAMP production, which induce deiodinase 2 (DIO2) to convert T4 to T3, which stimulates mitochondrial energy metabolism via activation of PGC‐1α.



Figure 9.

FXR regulation of hepatic glucose metabolism. FXR signaling phosphorylates and inhibits glycogen synthase kinase 3β (GSK3β), which is an inhibitor of glycogen synthase activity. This results in stimulating glycogenesis. The FXR/SHP pathway inhibits PEPCK and glucose 6‐phosphatase (G6Pase) to inhibit gluconeogenesis. This results in increasing glucose tolerance and insulin sensitivity.



Figure 10.

Mechanism of nonalcoholic fatty liver disease (NAFLD). Metabolic syndrome is a constellation of five clinical symptoms, hypertension, hyperglycemia, hypertriglyceridemia, insulin resistance, and obesity. Metabolic syndrome is linked to cardiovascular disease, type II diabetes, and NAFLD. NAFLD is progressed to nonalcoholic steatohepatitis (NASH) by many factors. The first hit is high fat diet (HFD)‐induced hepatic steatosis, followed by inflammation involving reactive oxidizing species, drugs, and endoplasmid reticulum stress to NASH. NASH patients have prevalence for liver fibrosis and cirrhosis, while NAFLD is linked to obesity, steatosis, and insulin resistance. HFD, high fat diet; ROS, reactive oxidizing species.

References
 1. Adorini L, Pruzanski M, Shapiro D. Farnesoid X receptor targeting to treat nonalcoholic steatohepatitis. Drug Discov Today 17: 988‐997, 2012.
 2. Agellon LB, Cheema SK. The 3′‐untranslated region of the mouse cholesterol 7α‐hydroxylase mRNA contains elements responsive to post‐transcriptional regulation by bile acids. Biochem J 328: 393‐399, 1997.
 3. Aldridge MA, Ito MK. Colesevelam hydrochloride: A novel bile acid‐binding resin. Ann Pharmacother 35: 898‐907, 2001.
 4. Alvarez L, Jara P, Sanchez‐Sabate E, Hierro L, Larrauri J, Diaz MC, Camarena C, De La Vega A, Frauca E, Lopez‐Collazo E, Lapunzina P. Reduced hepatic expression of Farnesoid X Receptor in hereditary cholestasis associated to mutation in ATP8B1. Hum Mol Genet 13: 2451‐2460, 2004.
 5. Amaral JD, Viana RJ, Ramalho RM, Steer CJ, Rodrigues CM. Bile acids: Regulation of apoptosis by ursodeoxycholic ccid. J Lipid Res 50: 1721‐1734, 2009.
 6. Angelin B, Einarsson K, Hellstrom K, Leijd B. Bile acid kinetics in relation to endogenous triglyceride metabolism in various types of hyperlipoproteinemia. J Lipid Res 19: 1004‐1016, 1978.
 7. Angelin B, Einarsson K, Hellstrom K, Leijd B. Effects of cholestyramine and chenodeoxycholic acid on the metabolism of endogenous triglyceride in hyperlipoproteinemia. J Lipid Res 19: 1017‐1024, 1978.
 8. Baker DM, Wang SL, Bell DJ, Drevon CA, Davis RA. One or more labile proteins regulate the stability of chimeric mRNAs containing the 3′‐untranslated region of cholesterol‐7alpha ‐ hydroxylase mRNA. J Biol Chem 275: 19985‐19991, 2000.
 9. Ballatori N, Christian WV, Lee JY, Dawson PA, Soroka CJ, Boyer JL, Madejczyk MS, Li N. OSTalpha‐OSTbeta: A major basolateral bile acid and steroid transporter in human intestinal, renal, and biliary epithelia. Hepatology 42: 1270‐1279, 2005.
 10. Ballatori N, Fang F, Christian WV, Li N, Hammond CL. Ostalpha‐Ostbeta is required for bile acid and conjugated steroid disposition in the intestine, kidney, and liver. Am J Physiol Gastrointest Liver Physiol 295: G179‐G186, 2008.
 11. Bays HE, Goldberg RB, Truitt KE, Jones MR. Colesevelam hydrochloride therapy in patients with type 2 diabetes mellitus treated with metformin: Glucose and lipid effects. Arch Intern Med 168: 1975‐1983, 2008.
 12. Bennion LJ, Grundy SM. Effects of diabetes mellitus on cholesterol metabolism in man. N Engl J Med 296: 1365‐1371, 1977.
 13. Bertolotti M, Carulli L, Concari M, Martella P, Loria P, Tagliafico E, Ferrari S, Del Puppo M, Amati B, De Fabiani E, Crestani M, Amorotti C, Manenti A, Carubbi F, Pinetti A, Carulli N. Suppression of bile acid synthesis, but not of hepatic cholesterol 7alpha‐hydroxylase expression, by obstructive cholestasis in humans. Hepatology 34: 234‐242., 2001.
 14. Beysen C, Murphy EJ, Deines K, Chan M, Tsang E, Glass A, Turner SM, Protasio J, Riiff T, Hellerstein MK. Effect of bile acid sequestrants on glucose metabolism, hepatic de novo lipogenesis, and cholesterol and bile acid kinetics in type 2 diabetes: A randomised controlled study. Diabetologia 55: 432‐442, 2012.
 15. Bhalla S, Ozalp C, Fang S, Xiang L, Kemper JK. Ligand‐activated PXR interferes with HNF‐4 signaling by targeting a common coactivator PGC‐1alpha : Functional implications in hepatic cholesterol and glucose metabolism. J Biol Chem 279: 45139‐45147, 2004.
 16. Bhatnagar S, Damron HA, Hillgartner FB. Fibroblast growth factor‐19, a novel factor that inhibits hepatic fatty acid synthesis. J Biol Chem 284: 10023‐10033, 2009.
 17. Bjorkhem I. Inborn errors of metabolism with consequences for bile acid biosynthesis. A minireview. Scand J Gastroenterol Suppl 204: 68‐72, 1994.
 18. Bjorkhem I, Leitersdorf I. Sterol 27‐hydroxylase deficiency: A rare cause of xanthomas in normocholesterolemic humans. Trends Endocrinol Metab 11: 180‐183, 2000.
 19. Bove KE, Daugherty CC, Tyson W, Mierau G, Heubi JE, Balistreri WF, Setchell KD. Bile acid synthetic defects and liver disease. Pediatr Dev Pathol 3: 1‐16, 2000.
 20. Brown MS, Goldstein JL. The SREBP pathway: Regulation of cholesterol metabolism by proteolysis of a membrane‐bound transcription factor. Cell 89: 331‐340, 1997.
 21. Brufau G, Groen AK, Kuipers F. Reverse cholesterol transport revisited: Contribution of biliary versus intestinal cholesterol excretion. Arterioscler Thromb Vasc Biol 31: 1726‐1733, 2011.
 22. Brufau G, Stellaard F, Prado K, Bloks VW, Jonkers E, Boverhof R, Kuipers F, Murphy EJ. Improved glycemic control with colesevelam treatment in patients with type 2 diabetes is not directly associated with changes in bile acid metabolism. Hepatology 52: 1455‐1464, 2010.
 23. Cai SY, Gautam S, Nguyen T, Soroka CJ, Rahner C, Boyer JL. ATP8B1 deficiency disrupts the bile canalicular membrane bilayer structure in hepatocytes, but FXR expression and activity are maintained. Gastroenterology 136: 1060‐1069, 2009.
 24. Calkin AC, Tontonoz P. Transcriptional integration of metabolism by the nuclear sterol‐activated receptors LXR and FXR. Nat Rev Mol Cell Biol 13: 213‐224, 2012.
 25. Cariou B, Bouchaert E, Abdelkarim M, Dumont J, Caron S, Fruchart JC, Burcelin R, Kuipers F, Staels B. FXR‐deficiency confers increased susceptibility to torpor. FEBS Lett 581: 5191‐5198, 2007.
 26. Cariou B, van Harmelen K, Duran‐Sandoval D, van Dijk TH, Grefhorst A, Abdelkarim M, Caron S, Torpier G, Fruchart JC, Gonzalez FJ, Kuipers F, Staels B. The farnesoid X receptor modulates adiposity and peripheral insulin sensitivity in mice. J Biol Chem 281: 11039‐11049, 2006.
 27. Carlton VE, Harris BZ, Puffenberger EG, Batta AK, Knisely AS, Robinson DL, Strauss KA, Shneider BL, Lim WA, Salen G, Morton DH, Bull LN. Complex inheritance of familial hypercholanemia with associated mutations in TJP2 and BAAT. Nat Genet 34: 91‐96, 2003.
 28. Chawla A, Repa JJ, Evans RM, Mangelsdorf DJ. Nuclear receptors and lipid physiology: Opening the X‐files. Science 294: 1866‐1870., 2001.
 29. Chen F, Ananthanarayanan M, Emre S, Neimark E, Bull LN, Knisely AS, Strautnieks SS, Thompson RJ, Magid MS, Gordon R, Balasubramanian N, Suchy FJ, Shneider BL. Progressive familial intrahepatic cholestasis, type 1, is associated with decreased farnesoid X receptor activity. Gastroenterology 126: 756‐764, 2004.
 30. Chen G, Liang G, Ou J, Goldstein JL, Brown MS. Central role for liver X receptor in insulin‐mediated activation of Srebp‐1c transcription and stimulation of fatty acid synthesis in liver. Proc Natl Acad Sci U S A 101: 11245‐11250, 2004.
 31. Chiang JY. Bile Acid regulation of gene expression: Roles of nuclear hormone receptors. Endocr Rev 23: 443‐463, 2002.
 32. Chiang JY. Regulation of bile acid synthesis: Pathways, nuclear receptors, and mechanisms. J Hepatol 40: 539‐551, 2004.
 33. Chiang JY. Bile acids: Regulation of synthesis. J Lipid Res 50: 1955‐1966, 2009.
 34. Chiang JYL. Regulation of bile acid synthesis. Front Biosci 3: D176‐D193, 1998.
 35. Chong CP, Mills PB, McClean P, Gissen P, Bruce C, Stahlschmidt J, Knisely AS, Clayton PT. Bile acid‐CoA ligase deficiency–a new inborn error of bile acid metabolism. J Inherit Metab Dis 35: 521‐530, 2012.
 36. Cipriani S, Mencarelli A, Chini MG, Distrutti E, Renga B, Bifulco G, Baldelli F, Donini A, Fiorucci S. The bile acid receptor GPBAR‐1 (TGR5) modulates integrity of intestinal barrier and immune response to experimental colitis. PLoS One 6: e25637, 2011.
 37. Cipriani S, Mencarelli A, Palladino G, Fiorucci S. FXR activation reverses insulin resistance and lipid abnormalities and protects against liver steatosis in Zucker (fa/fa) obese rats. J Lipid Res 51: 771‐784, 2010.
 38. Claudel T, Staels B, Kuipers F. The Farnesoid X receptor: A molecular link between bile acid and lipid and glucose metabolism. Arterioscler Thromb Vasc Biol 25: 2020‐2030, 2005.
 39. Clayton PT. Disorders of bile acid synthesis. J Inherit Metab Dis 34: 593‐604, 2011.
 40. Clayton PT, Leonard JV, Lawson AM, Setchell KD, Andersson S, Egestad B, Sjovall J. Familial giant cell hepatitis associated with synthesis of 3 beta, 7 alpha‐dihydroxy‐and 3 beta,7 alpha, 12 alpha‐trihydroxy‐5‐cholenoic acids. J Clin Invest 79: 1031‐1038, 1987.
 41. Cohen JC, Horton JD, Hobbs HH. Human fatty liver disease: Old questions and new insights. Science 332: 1519‐1523, 2011.
 42. Cordenonsi M, Montagner M, Adorno M, Zacchigna L, Martello G, Mamidi A, Soligo S, Dupont S, Piccolo S. Integration of TGF‐beta and Ras/MAPK signaling through p53 phosphorylation. Science 315: 840‐843, 2007.
 43. Davidson MH, Dillon MA, Gordon B, Jones P, Samuels J, Weiss S, Isaacsohn J, Toth P, Burke SK. Colesevelam hydrochloride (cholestagel): A new, potent bile acid sequestrant associated with a low incidence of gastrointestinal side effects. Arch Intern Med 159: 1893‐1900, 1999.
 44. Dawson PA, Hubbert M, Haywood J, Craddock AL, Zerangue N, Christian WV, Ballatori N. The heteromeric organic solute transporter alpha‐beta, Ostalpha‐Ostbeta, is an ileal basolateral bile acid transporter. J Biol Chem 280: 6960‐6968, 2005.
 45. Day CP, James OF. Steatohepatitis: A tale of two “hits”? Gastroenterology 114: 842‐845, 1998.
 46. De Fabiani E, Mitro N, Gilardi F, Caruso D, Galli G, Crestani M. Coordinated control of cholesterol catabolism to bile acids and of gluconeogenesis via a novel mechanism of transcription regulation linked to the fasted‐to‐fed cycle. J Biol Chem 278: 39124‐39132, 2003.
 47. DeBose‐Boyd RA, Ou J, Goldstein JL, Brown MS. Expression of sterol regulatory element‐binding protein 1c (SREBP‐1c) mRNA in rat hepatoma cells requires endogenous LXR ligands. Proc Natl Acad Sci U S A 98: 1477‐1482., 2001.
 48. Del Castillo‐Olivares A, Campos JA, Pandak WM, Gil G. Role of FTF/LRH‐1 on bile acid biosynthesis. A known nuclear receptor activator that can Act as a suppressor of bile acid biosynthesis. J Biol Chem 279: 16813‐16821, 2004.
 49. Denson LA, Sturm E, Echevarria W, Zimmerman TL, Makishima M, Mangelsdorf DJ, Karpen SJ. The orphan nuclear receptor, shp, mediates bile acid‐induced inhibition of the rat bile acid transporter, ntcp. Gastroenterology 121: 140‐147, 2001.
 50. Dent P, Fang Y, Gupta S, Studer E, Mitchell C, Spiegel S, Hylemon PB. Conjugated bile acids promote ERK1/2 and AKT activation via a pertussis toxin‐sensitive mechanism in murine and human hepatocytes. Hepatology 42: 1291‐1299, 2005.
 51. Drury JE, Mindnich R, Penning TM. Characterization of disease‐related 5beta‐reductase (AKR1D1) mutations reveals their potential to cause bile acid deficiency. J Biol Chem 285: 24529‐24537, 2010.
 52. Duane WC, Javitt NB. 27‐hydroxycholesterol. Production rates in normal human subjects. J Lipid Res 40: 1194‐1199, 1999.
 53. Duran‐Sandoval D, Mautino G, Martin G, Percevault F, Barbier O, Fruchart JC, Kuipers F, Staels B. Glucose regulates the expression of the farnesoid X receptor in liver. Diabetes 53: 890‐898, 2004.
 54. Edwards PA, Tabor D, Kast HR, Venkateswaran A. Regulation of gene expression by SREBP and SCAP. Biochim Biophys Acta 1529: 103‐113, 2000.
 55. Eloranta JJ, Kullak‐Ublick GA. The role of FXR in disorders of bile acid homeostasis. Physiology (Bethesda) 23: 286‐295, 2008.
 56. Engelking LJ, Kuriyama H, Hammer RE, Horton JD, Brown MS, Goldstein JL, Liang G. Overexpression of Insig‐1 in the livers of transgenic mice inhibits SREBP processing and reduces insulin‐stimulated lipogenesis. J Clin Invest 113: 1168‐1175, 2004.
 57. Fang Y, Han SI, Mitchell C, Gupta S, Studer E, Grant S, Hylemon PB, Dent P. Bile acids induce mitochondrial ROS, which promote activation of receptor tyrosine kinases and signaling pathways in rat hepatocytes. Hepatology 40: 961‐971, 2004.
 58. Fang Y, Studer E, Mitchell C, Grant S, Pandak WM, Hylemon PB, Dent P. Conjugated bile acids regulate hepatocyte glycogen synthase activity in vitro and in vivo via Galphai signaling. Mol Pharmacol 71: 1122‐1128, 2007.
 59. Farrell GC, Larter CZ. Nonalcoholic fatty liver disease: From steatosis to cirrhosis. Hepatology 43: S99‐S112, 2006.
 60. Ferdinandusse S, Denis S, Faust PL, Wanders RJ. Bile acids: Role of peroxisomes. J Lipid Res 50: 2139‐2147, 2009.
 61. Ferdinandusse S, Overmars H, Denis S, Waterham HR, Wanders RJ, Vreken P. Plasma analysis of di‐ and trihydroxycholestanoic acid diastereoisomers in peroxisomal alpha‐methylacyl‐CoA racemase deficiency. J Lipid Res 42: 137‐141, 2001.
 62. Fiorucci S, Baldelli F. Farnesoid X receptor agonists in biliary tract disease. Curr Opin Gastroenterol 25: 252‐259, 2009.
 63. Fiorucci S, Cipriani S, Mencarelli A, Renga B, Distrutti E, Baldelli F. Counter‐regulatory role of bile acid activated receptors in immunity and inflammation. Curr Mol Med 10: 579‐595, 2010.
 64. Foretz M, Pacot C, Dugail I, Lemarchand P, Guichard C, Le Liepvre X, Berthelier‐Lubrano C, Spiegelman B, Kim JB, Ferre P, Foufelle F. ADD1/SREBP‐1c is required in the activation of hepatic lipogenic gene expression by glucose. Mol Cell Biol 19: 3760‐3768, 1999.
 65. Francis GA, Fayard E, Picard F, Auwerx J. Nuclear receptors and the control of metabolism. Annu Rev Physiol 65: 261‐311, 2003.
 66. Frankenberg T, Rao A, Chen F, Haywood J, Shneider BL, Dawson PA. Regulation of the mouse organic solute transporter alpha‐beta, Ostalpha‐Ostbeta, by bile acids. Am J Physiol Gastrointest Liver Physiol 290: G912‐G922, 2006.
 67. Fu L, John LM, Adams SH, Yu XX, Tomlinson E, Renz M, Williams PM, Soriano R, Corpuz R, Moffat B, Vandlen R, Simmons L, Foster J, Stephan JP, Tsai SP, Stewart TA. Fibroblast growth factor 19 increases metabolic rate and reverses dietary and leptin‐deficient diabetes. Endocrinology 145: 2594‐2603, 2004.
 68. Gadaleta RM, Oldenburg B, Willemsen EC, Spit M, Murzilli S, Salvatore L, Klomp LW, Siersema PD, van Erpecum KJ, van Mil SW. Activation of bile salt nuclear receptor FXR is repressed by pro‐inflammatory cytokines activating NF‐kappa B signaling in the intestine. Biochim Biophys Acta 1812: 851‐858, 2011.
 69. Gadaleta RM, van Erpecum KJ, Oldenburg B, Willemsen EC, Renooij W, Murzilli S, Klomp LW, Siersema PD, Schipper ME, Danese S, Penna G, Laverny G, Adorini L, Moschetta A, van Mil SW. Farnesoid X receptor activation inhibits inflammation and preserves the intestinal barrier in inflammatory bowel disease. Gut 60: 463‐472, 2011.
 70. Ginsberg HN, Zhang YL, Hernandez‐Ono A. Metabolic syndrome: Focus on dyslipidemia. Obesity (Silver Spring) 14(Suppl 1): 41S‐49S, 2006.
 71. Goldberg RB, Fonseca VA, Truitt KE, Jones MR. Efficacy and safety of colesevelam in patients with type 2 diabetes mellitus and inadequate glycemic control receiving insulin‐based therapy. Arch Intern Med 168: 1531‐1540, 2008.
 72. Goldfisher S, Moore CL, Johnson AB, Spiro AI, Valsamis MP, Wisniewski HK, Ritch RH, Norton WT, Rapin I, Gartner M. Peroxisomal and mitochondrial defects in the cerebro‐hepato‐renal syndrome. Science 182: 62‐64, 1997.
 73. Gonzales E, Cresteil D, Baussan C, Dabadie A, Gerhardt MF, Jacquemin E. SRD5B1 (AKR1D1) gene analysis in delta(4)‐3‐oxosteroid 5beta‐reductase deficiency: Evidence for primary genetic defect. J Hepatol 40: 716‐718, 2004.
 74. Goodwin B, Jones SA, Price RR, Watson MA, McKee DD, Moore LB, Galardi C, Wilson JG, Lewis MC, Roth ME, Maloney PR, Willson TM, Kliewer SA. A regulatory cascade of the nuclear receptors FXR, SHP‐1, and LRH‐1 represses bile acid biosynthesis. Mol Cell 6: 517‐526, 2000.
 75. Groen AK, Oude Elferink RP, Verkade HJ, Kuipers F. The ins and outs of reverse cholesterol transport. Ann Med 36: 135‐145, 2004.
 76. Guo GL, Lambert G, Negishi M, Ward JM, Brewer HB, Jr., Kliewer SA, Gonzalez FJ, Sinal CJ. Complementary roles of farnesoid X receptor, pregnane X receptor, and constitutive androstane receptor in protection against bile acid toxicity. J Biol Chem 278: 45062‐45071, 2003.
 77. Hageman J, Herrema H, Groen AK, Kuipers F. A role of the bile salt receptor FXR in atherosclerosis. Arterioscler Thromb Vasc Biol 30: 1519‐1528, 2010.
 78. Han S, Chiang JY. Mechanism of vitamin D receptor inhibition of cholesterol 7alpha‐hydroxylase gene transcription in human hepatocytes. Drug Metab Dispos 37: 469‐478, 2009.
 79. Hansen M, Sonne DP, Mikkelsen KH, Gluud LL, Vilsboll T, Knop FK. Effect of bile acid sequestrants on glycaemic control: Protocol for a systematic review with meta‐analysis of randomised controlled trials. BMJ open 2: 2012.
 80. Hanson RF, Szczepanik‐Van Leeuwen P, Williams GC, Grabowski G, Sharp HL. Defects of bile acid synthesis in Zellweger's syndrome. Science 203: 1107‐1108, 1979.
 81. He J, Nishida S, Xu M, Makishima M, Xie W. PXR prevents cholesterol gallstone disease by regulating biosynthesis and transport of bile salts. Gastroenterology 140: 2095‐2106, 2011.
 82. Hofmann AF. Detoxification of lithocholic acid, a toxic bile acid: Relevance to drug hepatotoxicity. Drug Metab Rev 36: 703‐722, 2004.
 83. Holt JA, Luo G, Billin AN, Bisi J, McNeill YY, Kozarsky KF, Donahee M, Wang da Y, Mansfield TA, Kliewer SA, Goodwin B, Jones SA. Definition of a novel growth factor‐dependent signal cascade for the suppression of bile acid biosynthesis. Genes Dev 17: 1581‐1591, 2003.
 84. Hong C, Bradley MN, Rong X, Wang X, Wagner A, Grijalva V, Castellani LW, Salazar J, Realegeno S, Boyadjian R, Fogelman AM, Van Lenten BJ, Reddy ST, Lusis AJ, Tangirala RK, Tontonoz P. LXRalpha is uniquely required for maximal reverse cholesterol transport and atheroprotection in ApoE‐deficient mice. J Lipid Res 53: 1126‐1133, 2012.
 85. Horton JD, Goldstein JL, Brown MS. SREBPs: Activators of the complete program of cholesterol and fatty acid synthesis in the liver. J Clin Invest 109: 1125‐1131, 2002.
 86. Houten SM, Watanabe M, Auwerx J. Endocrine functions of bile acids. Embo J 25: 1419‐1425, 2006.
 87. Hylemon PB, Zhou H, Pandak WM, Ren S, Gil G, Dent P. Bile acids as regulatory molecules. J Lipid Res 50: 1509‐1520, 2009.
 88. Inagaki T, Choi M, Moschetta A, Peng L, Cummins CL, McDonald JG, Luo G, Jones SA, Goodwin B, Richardson JA, Gerard RD, Repa JJ, Mangelsdorf DJ, Kliewer SA. Fibroblast growth factor 15 functions as an enterohepatic signal to regulate bile acid homeostasis. Cell Metab 2: 217‐225, 2005.
 89. Inagaki T, Moschetta A, Lee YK, Peng L, Zhao G, Downes M, Yu RT, Shelton JM, Richardson JA, Repa JJ, Mangelsdorf DJ, Kliewer SA. Regulation of antibacterial defense in the small intestine by the nuclear bile acid receptor. Proc Natl Acad Sci U S A 103: 3920‐3925, 2006.
 90. Ito S, Fujimori T, Furuya A, Satoh J, Nabeshima Y. Impaired negative feedback suppression of bile acid synthesis in mice lacking betaKlotho. J Clin Invest 115: 2202‐2208, 2005.
 91. Jacquemin E. Role of multidrug resistance 3 deficiency in pediatric and adult liver disease: one gene for three diseases. Semin Liver Dis 21: 551‐562, 2001.
 92. Jahan A, Chiang JY. Cytokine regulation of human sterol 12{alpha}‐hydroxylase (CYP8B1) gene. Am J Physiol Gastrointest Liver Physiol 288: G685‐G695, 2005.
 93. Jansen PL, Sturm E. Genetic cholestasis, causes and consequences for hepatobiliary transport. Liver Int 23: 315‐322, 2003.
 94. Kast HR, Nguyen CM, Sinal CJ, Jones SA, Laffitte BA, Reue K, Gonzalez FJ, Willson TM, Edwards PA. Farnesoid x‐activated receptor induces apolipoprotein c‐ii transcription: A molecular mechanism linking plasma triglyceride levels to bile acids. Mol Endocrinol 15: 1720‐1728., 2001.
 95. Katsuma S, Hirasawa A, Tsujimoto G. Bile acids promote glucagon‐like peptide‐1 secretion through TGR5 in a murine enteroendocrine cell line STC‐1. Biochem Biophys Res Commun 329: 386‐390, 2005.
 96. Kawamata Y, Fujii R, Hosoya M, Harada M, Yoshida H, Miwa M, Fukusumi S, Habata Y, Itoh T, Shintani Y, Hinuma S, Fujisawa Y, Fujino M. A G protein‐coupled receptor responsive to bile acids. J Biol Chem 278: 9435‐9440, 2003.
 97. Keitel V, Cupisti K, Ullmer C, Knoefel WT, Kubitz R, Haussinger D. The membrane‐bound bile acid receptor TGR5 is localized in the epithelium of human gallbladders. Hepatology 50: 861‐870, 2009.
 98. Keitel V, Donner M, Winandy S, Kubitz R, Haussinger D. Expression and function of the bile acid receptor TGR5 in Kupffer cells. Biochem Biophys Res Commun 372: 78‐84, 2008.
 99. Keitel V, Nies AT, Brom M, Hummel‐Eisenbeiss J, Spring H, Keppler D. A common Dubin‐Johnson syndrome mutation impairs protein maturation and transport activity of MRP2 (ABCC2). Am J Physiol Gastrointest Liver Physiol 284: G165‐G174, 2003.
 100. Keitel V, Reinehr R, Gatsios P, Rupprecht C, Gorg B, Selbach O, Haussinger D, Kubitz R. The G‐protein coupled bile salt receptor TGR5 is expressed in liver sinusoidal endothelial cells. Hepatology 45: 695‐704, 2007.
 101. Kerr TA, Saeki S, Schneider M, Schaefer K, Berdy S, Redder T, Shan B, Russell DW, Schwarz M. Loss of nbuclear receptor SHP impairs but does not eliminate negative feedback regulation of bile acid synthesis. Dev Cell 2: 713‐720, 2002.
 102. Kim I, Ahn SH, Inagaki T, Choi M, Ito S, Guo GL, Kliewer SA, Gonzalez FJ. Differential regulation of bile acid homeostasis by the farnesoid X receptor in liver and intestine. J Lipid Res 48: 2664‐2672, 2007.
 103. Kullak‐Ublick GA, Meier PJ. Mechanisms of cholestasis. Clin Liver Dis 4: 357‐385, 2000.
 104. Lang C, Meier Y, Stieger B, Beuers U, Lang T, Kerb R, Kullak‐Ublick GA, Meier PJ, Pauli‐Magnus C. Mutations and polymorphisms in the bile salt export pump and the multidrug resistance protein 3 associated with drug‐induced liver injury. Pharmacogenet Genomics 17: 47‐60, 2007.
 105. Lang T, Haberl M, Jung D, Drescher A, Schlagenhaufer R, Keil A, Mornhinweg E, Stieger B, Kullak‐Ublick GA, Kerb R. Genetic variability, haplotype structures, and ethnic diversity of hepatic transporters MDR3 (ABCB4) and bile salt export pump (ABCB11). Drug Metab Dispos 34: 1582‐1599, 2006.
 106. Lefebvre P, Cariou B, Lien F, Kuipers F, Staels B. Role of bile acids and bile acid receptors in metabolic regulation. Physiol Rev 89: 147‐191, 2009.
 107. Lemonde HA, Custard EJ, Bouquet J, Duran M, Overmars H, Scambler PJ, Clayton PT. Mutations in SRD5B1 (AKR1D1), the gene encoding delta(4)‐3‐oxosteroid 5beta‐reductase, in hepatitis and liver failure in infancy. Gut 52: 1494‐1499, 2003.
 108. Lewis GF, Rader DJ. New insights into the regulation of HDL metabolism and reverse cholesterol transport. Circ Res 96: 1221‐1232, 2005.
 109. Li T, Chanda D, Zhang Y, Choi HS, Chiang JYL. Glucose stimulates cholesterol 7alpha‐hydroxylase gene transcription in human hepatocytes. J Lipid Res 51: 832‐842, 2010.
 110. Li T, Chiang JY. Mechanism of rifampicin and pregnane X receptor (PXR) inhibition of human cholesterol 7{alpha}‐hydroxylase gene (CYP7A1) transcription. Am J Physiol Gastrointest Liver Physiol 288: G74‐G84, 2005.
 111. Li T, Chiang JY. A novel role of transforming growth factor beta1 in transcriptional repression of human cholesterol 7alpha‐hydroxylase gene. Gastroenterology 133: 1660‐1669, 2007.
 112. Li T, Francl JM, Boehme S, Ochoa A, Zhang Y, Klaassen CD, Erickson SK, Chiang JY. Glucose and insulin induction of bile acid synthesis: Mechanisms and implication in diabetes and obesity. J Biol Chem 287: 1861‐1873, 2012.
 113. Li T, Holmstrom SR, Kir S, Umetani M, Schmidt DR, Kliewer SA, Mangelsdorf DJ. The G protein‐coupled bile acid receptor, TGR5, stimulates gallbladder filling. Mol Endocrinol 25: 1066‐1071, 2011.
 114. Li T, Jahan A, Chiang JY. Bile acids and cytokines inhibit the human cholesterol 7alpha‐hydroxylase gene via the JNK/c‐jun pathway in human liver cells. Hepatology 43: 1202‐1210, 2006.
 115. Li T, Kong X, Owsley E, Ellis E, Strom S, Chiang JY. Insulin regulation of cholesterol 7{alpha}‐hydroxylase expression in human hepatocytes: Roles of forkhead box o1 and sterol regulatory element‐binding protein 1c. J Biol Chem 281: 28745‐28754, 2006.
 116. Li T, Matozel M, Boehme S, Kong B, Nilsson LM, Guo G, Ellis E, Chiang JY. Overexpression of cholesterol 7alpha‐hydroxylase promotes hepatic bile acid synthesis and secretion and maintains cholesterol homeostasis. Hepatology 53: 996‐1006, 2011.
 117. Li T, Owsley E, Matozel M, Hsu P, Novak CM, Chiang JY. Transgenic expression of cholesterol 7alpha‐hydroxylase in the liver prevents high‐fat diet‐induced obesity and insulin resistance in mice. Hepatology 52: 678‐690, 2010.
 118. Li YC, Wang DP, Chiang JY. Regulation of cholesterol 7 alpha‐hydroxylase in the liver. Cloning, sequencing, and regulation of cholesterol 7 alpha‐hydroxylase mRNA. J Biol Chem 265: 12012‐12019, 1990.
 119. Li YT, Swales KE, Thomas GJ, Warner TD, Bishop‐Bailey D. Farnesoid x receptor ligands inhibit vascular smooth muscle cell inflammation and migration. Arterioscler Thromb Vasc Biol 27: 2606‐2611, 2007.
 120. Lin BC, Wang M, Blackmore C, Desnoyers LR. Liver specific activities of FGF19 require KLOTHO beta. J Biol Chem 282: 27277‐27284, 2007.
 121. Lu TT, Makishima M, Repa JJ, Schoonjans K, Kerr TA, Auwerx J, Mangelsdorf DJ. Molecular basis for feedback regulation of bile acid synthesis by nuclear receptors. Mol Cell 6: 507‐515, 2000.
 122. Lundasen T, Galman C, Angelin B, Rudling M. Circulating intestinal fibroblast growth factor 19 has a pronounced diurnal variation and modulates hepatic bile acid synthesis in man. J Intern Med 260: 530‐536, 2006.
 123. Lundasen T, Liao W, Angelin B, Rudling M. Leptin induces the hepatic high density lipoprotein receptor scavenger receptor B type I (SR‐BI) but not cholesterol 7alpha‐hydroxylase (Cyp7a1) in leptin‐deficient (ob/ob) mice. J Biol Chem 278: 43224‐43228, 2003.
 124. Ma K, Saha PK, Chan L, Moore DD. Farnesoid X receptor is essential for normal glucose homeostasis. J Clin Invest 116: 1102‐1109, 2006.
 125. Maeda Y, Seidel SD, Wei G, Liu X, Sladek FM. Repression of hepatocyte nuclear factor 4alpha tumor suppressor p53: Involvement of the ligand‐binding domain and histone deacetylase activity. Mol Endocrinol 16: 402‐410, 2002.
 126. Makishima M, Lu TT, Xie W, Whitfield GK, Domoto H, Evans RM, Haussler MR, Mangelsdorf DJ. Vitamin D receptor as an intestinal bile acid sensor. Science 296: 1313‐1316., 2002.
 127. Makishima M, Okamoto AY, Repa JJ, Tu H, Learned RM, Luk A, Hull MV, Lustig KD, Mangelsdorf DJ, Shan B. Identification of a nuclear receptor for bile acids. Science 284: 1362‐1365, 1999.
 128. Maruyama T, Miyamoto Y, Nakamura T, Tamai Y, Okada H, Sugiyama E, Itadani H, Tanaka K. Identification of membrane‐type receptor for bile acids (M‐BAR). Biochem Biophys Res Commun 298: 714‐719, 2002.
 129. Maruyama T, Tanaka K, Suzuki J, Miyoshi H, Harada N, Nakamura T, Miyamoto Y, Kanatani A, Tamai Y. Targeted disruption of G protein‐coupled bile acid receptor 1 (Gpbar1/M‐Bar) in mice. J Endocrinol 191: 197‐205, 2006.
 130. Miao J, Xiao Z, Kanamaluru D, Min G, Yau PM, Veenstra TD, Ellis E, Strom S, Suino‐Powell K, Xu HE, Kemper JK. Bile acid signaling pathways increase stability of small heterodimer partner (SHP) by inhibiting ubiquitin‐proteasomal degradation. Genes Dev 23: 986‐996, 2009.
 131. Miyake JH, Duong‐Polk XT, Taylor JM, Du EZ, Castellani LW, Lusis AJ, Davis RA. Transgenic expression of cholesterol‐7‐alpha‐hydroxylase prevents atherosclerosis in C57BL/6J mice. Arterioscler Thromb Vasc Biol 22: 121‐126, 2002.
 132. Miyake JH, Wang SL, Davis RA. Bile acid induction of cytokine expression by macrophages correlates with repression of hepatic cholesterol 7alpha‐hydroxylase. J Biol Chem 275: 21805‐21808, 2000.
 133. Modica S, Gadaleta RM, Moschetta A. Deciphering the nuclear bile acid receptor FXR paradigm. Nuclear receptor signaling 8: e005, 2010.
 134. Moustafa T, Fickert P, Magnes C, Guelly C, Thueringer A, Frank S, Kratky D, Sattler W, Reicher H, Sinner F, Gumhold J, Silbert D, Fauler G, Hofler G, Lass A, Zechner R, Trauner M. Alterations in lipid metabolism mediate inflammation, fibrosis, and proliferation in a mouse model of chronic cholestatic liver injury. Gastroenterology 142: 140‐151 e112, 2012.
 135. Mullenbach R, Bennett A, Tetlow N, Patel N, Hamilton G, Cheng F, Chambers J, Howard R, Taylor‐Robinson SD, Williamson C. ATP8B1 mutations in British cases with intrahepatic cholestasis of pregnancy. Gut 54: 829‐834, 2005.
 136. Myant NB, Mitropoulos KA. Cholesterol 7a‐hydroxylase. J Lipid Res 18: 135‐153, 1977.
 137. Noe J, Kullak‐Ublick GA, Jochum W, Stieger B, Kerb R, Haberl M, Mullhaupt B, Meier PJ, Pauli‐Magnus C. Impaired expression and function of the bile salt export pump due to three novel ABCB11 mutations in intrahepatic cholestasis. J Hepatol 43: 536‐543, 2005.
 138. Painter JN, Savander M, Ropponen A, Nupponen N, Riikonen S, Ylikorkala O, Lehesjoki AE, Aittomaki K. Sequence variation in the ATP8B1 gene and intrahepatic cholestasis of pregnancy. Eur J Hum Genet 13: 435‐439, 2005.
 139. Pandak WM, Stravitz RT, Lucas V, Heuman DM, Chiang JY. Hep G2 cells: A model for studies on regulation of human cholesterol 7alpha‐hydroxylase at the molecular level. Am J Physiol 270: G401‐G410, 1996.
 140. Parks DJ, Blanchard SG, Bledsoe RK, Chandra G, Consler TG, Kliewer SA, Stimmel JB, Willson TM, Zavacki AM, Moore DD, Lehmann JM. Bile acids: Natural ligands for an orphan nuclear receptor. Science 284: 1365‐1368, 1999.
 141. Patti ME, Houten SM, Bianco AC, Bernier R, Larsen PR, Holst JJ, Badman MK, Maratos‐Flier E, Mun EC, Pihlajamaki J, Auwerx J, Goldfine AB. Serum bile acids are higher in humans with prior gastric bypass: Potential contribution to improved glucose and lipid metabolism. Obesity (Silver Spring) 17: 1671‐1677, 2009.
 142. Pauli‐Magnus C, Lang T, Meier Y, Zodan‐Marin T, Jung D, Breymann C, Zimmermann R, Kenngott S, Beuers U, Reichel C, Kerb R, Penger A, Meier PJ, Kullak‐Ublick GA. Sequence analysis of bile salt export pump (ABCB11) and multidrug resistance p‐glycoprotein 3 (ABCB4, MDR3) in patients with intrahepatic cholestasis of pregnancy. Pharmacogenetics 14: 91‐102, 2004.
 143. Pellicciari R, Costantino G, Camaioni E, Sadeghpour BM, Entrena A, Willson TM, Fiorucci S, Clerici C, Gioiello A. Bile acid derivatives as ligands of the farnesoid X receptor. Synthesis, evaluation, and structure‐activity relationship of a series of body and side chain modified analogues of chenodeoxycholic acid. J Med Chem 47: 4559‐4569, 2004.
 144. Pellicciari R, Gioiello A, Macchiarulo A, Thomas C, Rosatelli E, Natalini B, Sardella R, Pruzanski M, Roda A, Pastorini E, Schoonjans K, Auwerx J. Discovery of 6alpha‐ethyl‐23(S)‐methylcholic acid (S‐EMCA, INT‐777) as a potent and selective agonist for the TGR5 receptor, a novel target for diabesity. J Med Chem 52: 7958‐7961, 2009.
 145. Pellicciari R, Sato H, Gioiello A, Costantino G, Macchiarulo A, Sadeghpour BM, Giorgi G, Schoonjans K, Auwerx J. Nongenomic actions of bile acids. Synthesis and preliminary characterization of 23‐ and 6,23‐alkyl‐substituted bile acid derivatives as selective modulators for the G‐protein coupled receptor TGR5. J Med Chem 50: 4265‐4268, 2007.
 146. Pols TW, Nomura M, Harach T, Lo Sasso G, Oosterveer MH, Thomas C, Rizzo G, Gioiello A, Adorini L, Pellicciari R, Auwerx J, Schoonjans K. TGR5 activation inhibits atherosclerosis by reducing macrophage inflammation and lipid loading. Cell Metab 14: 747‐757, 2011.
 147. Pols TW, Noriega LG, Nomura M, Auwerx J, Schoonjans K. The bile acid membrane receptor TGR5 as an emerging target in metabolism and inflammation. J Hepatol 54: 1263‐1272, 2011.
 148. Porez G, Prawitt J, Gross B, Staels B. Bile acid receptors as targets for the treatment of dyslipidemia and cardiovascular disease: Thematic review series: New lipid and lipoprotein targets for the treatment of cardiometabolic diseases. J Lipid Res 53: 1723‐1737, 2012.
 149. Pournaras DJ, Glicksman C, Vincent RP, Kuganolipava S, Alaghband‐Zadeh J, Mahon D, Bekker JH, Ghatei MA, Bloom SR, Walters JR, Welbourn R, le Roux CW. The Role of Bile After Roux‐en‐Y Gastric Bypass in Promoting Weight Loss and Improving Glycaemic Control. Endocrinology 153: 3613‐3619, 2012.
 150. Prawitt J, Abdelkarim M, Stroeve JH, Popescu I, Duez H, Velagapudi VR, Dumont J, Bouchaert E, van Dijk TH, Lucas A, Dorchies E, Daoudi M, Lestavel S, Gonzalez FJ, Oresic M, Cariou B, Kuipers F, Caron S, Staels B. Farnesoid X receptor deficiency improves glucose homeostasis in mouse models of obesity. Diabetes 60: 1861‐1871, 2011.
 151. Puigserver P, Rhee J, Donovan J, Walkey CJ, Yoon JC, Oriente F, Kitamura Y, Altomonte J, Dong H, Accili D, Spiegelman BM. Insulin‐regulated hepatic gluconeogenesis through FOXO1‐PGC‐1alpha interaction. Nature 423: 550‐555, 2003.
 152. Pullinger CR, Eng C, Salen G, Shefer S, Batta AK, Erickson SK, Verhagen A, Rivera CR, Mulvihill SJ, Malloy MJ, Kane JP. Human cholesterol 7alpha‐hydroxylase (CYP7A1) deficiency has a hypercholesterolemic phenotype. J Clin Invest 110: 109‐117., 2002.
 153. Qiao L, McKinstry R, Gupta S, Gilfor D, Windle JJ, Hylemon PB, Grant S, Fisher PB, Dent P. Cyclin kinase inhibitor p21 potentiates bile acid‐induced apoptosis in hepatocytes that is dependent on p53. Hepatology 36: 39‐48, 2002.
 154. Ratliff EP, Gutierrez A, Davis RA. Transgenic expression of CYP7A1 in LDL receptor‐deficient mice blocks diet‐induced hypercholesterolemia. J Lipid Res 47: 1513‐1520, 2006.
 155. Reaven G, Abbasi F, McLaughlin T. Obesity, insulin resistance, and cardiovascular disease. Recent Prog Horm Res 59: 207‐223, 2004.
 156. Reaven GM. Banting lecture 1988. Role of insulin resistance in human disease. Diabetes 37: 1595‐1607, 1988.
 157. Reinehr R, Becker S, Keitel V, Eberle A, Grether‐Beck S, Haussinger D. Bile salt‐induced apoptosis involves NADPH oxidase isoform activation. Gastroenterology 129: 2009‐2031, 2005.
 158. Renga B, Mencarelli A, Vavassori P, Brancaleone V, Fiorucci S. The bile acid sensor FXR regulates insulin transcription and secretion. Biochim Biophys Acta 1802: 363‐372, 2010.
 159. Russell DW. The enzymes, regulation, and genetics of bile acid synthesis. Annu Rev Biochem 72: 1370174, 2003.
 160. Schaap FG, van der Gaag NA, Gouma DJ, Jansen PL. High expression of the bile salt‐homeostatic hormone fibroblast growth factor 19 in the liver of patients with extrahepatic cholestasis. Hepatology 49: 1228‐1235, 2009.
 161. Schwarz M, Wright AC, Davis DL, Nazer H, Bjorkhem I, Russell DW. The bile acid synthetic gene 3beta‐hydroxy‐Delta(5)‐C(27)‐steroid oxidoreductase is mutated in progressive intrahepatic cholestasis. J Clin Invest 106: 1175‐1184., 2000.
 162. Setchell KD, Heubi JE, Bove KE, O'Connell NC, Brewsaugh T, Steinberg SJ, Moser A, Squires RH, Jr. Liver disease caused by failure to racemize trihydroxycholestanoic acid: Gene mutation and effect of bile acid therapy. Gastroenterology 124: 217‐232, 2003.
 163. Setchell KD, Street JM. Inborn errors of bile acid synthesis. Semin Liver Dis 7: 85‐99, 1987.
 164. Setchell KD, Suchy FJ, Welsh MB, Zimmer‐Nechemias L, Heubi J, Balistreri WF. Delta 4‐3‐oxosteroid 5 beta‐reductase deficiency described in identical twins with neonatal hepatitis. A new inborn error in bile acid synthesis. J Clin Invest 82: 2148‐2157, 1988.
 165. Setchell KDR, Schwarz M, O'Connell NC, Lund EG, Davis DL, Lathe R, Thompson HR, Weslie Tyson R, Sokol RJ, Russell DW. Identification of a new inborn error in bile acid synthesis: Mutation of the oxysterol 7α‐hydroxylase gene causes severe neonatal liver disease. J Clin Invest 102: 1690‐1703, 1998.
 166. Shaham O, Wei R, Wang TJ, Ricciardi C, Lewis GD, Vasan RS, Carr SA, Thadhani R, Gerszten RE, Mootha VK. Metabolic profiling of the human response to a glucose challenge reveals distinct axes of insulin sensitivity. Mol Syst Biol 4: 214, 2008.
 167. Shang Q, Saumoy M, Holst JJ, Salen G, Xu G. Colesevelam improves insulin resistance in a diet‐induced obesity (F‐DIO) rat model by increasing the release of GLP‐1. Am J Physiol Gastrointest Liver Physiol 298: G419‐424, 2010.
 168. Shimomura I, Bashmakov Y, Horton JD. Increased levels of nuclear SREBP‐1c associated with fatty livers in two mouse models of diabetes mellitus. J Biol Chem 274: 30028‐30032, 1999.
 169. Shimomura I, Bashmakov Y, Ikemoto S, Horton JD, Brown MS, Goldstein JL. Insulin selectively increases SREBP‐1c mRNA in the livers of rats with streptozotocin‐induced diabetes. Proc Natl Acad Sci U S A 96: 13656‐13661., 1999.
 170. Shin DJ, Campos JA, Gil G, Osborne TF. PGC‐1a activates CYP7A1 and bile acid biosynthesis. J Biol Chem 278: 50047‐50052, 2003.
 171. Shneider BL, Setchell KD, Whitington PF, Neilson KA, Suchy FJ. Delta 4‐3‐oxosteroid 5 beta‐reductase deficiency causing neonatal liver failure and hemochromatosis [see comments]. J Pediatr 124: 234‐238, 1994.
 172. Sinal CJ, Tohkin M, Miyata M, Ward JM, Lambert G, Gonzalez FJ. Targeted disruption of the nuclear receptor FXR/BAR impairs bile acid and lipid homeostasis. Cell 102: 731‐744, 2000.
 173. Sola S, Amaral JD, Aranha MM, Steer CJ, Rodrigues CM. Modulation of hepatocyte apoptosis: cross‐talk between bile acids and nuclear steroid receptors. Curr Med Chem 13: 3039‐3051, 2006.
 174. Song KH, Chiang JY. Glucagon and cAMP inhibit cholesterol 7alpha‐hydroxylase (CYP7a1) gene expression in human hepatocytes: Discordant regulation of bile acid synthesis and gluconeogenesis. Hepatology 43: 117‐125, 2006.
 175. Song KH, Ellis E, Strom S, Chiang JY. Hepatocyte growth factor signaling pathway inhibits cholesterol 7alpha‐hydroxylase and bile acid synthesis in human hepatocytes. Hepatology 46: 1993‐2002, 2007.
 176. Song KH, Li T, Owsley E, Strom S, Chiang JY. Bile acids activate fibroblast growth factor 19 signaling in human hepatocytes to inhibit cholesterol 7alpha‐hydroxylase gene expression. Hepatology 49: 297‐305, 2009.
 177. Song MB, Yu XJ, Zhu GX, Chen JF, Zhao G, Huang L. Transfection of HGF gene enhances endothelial progenitor cell (EPC) function and improves EPC transplant efficiency for balloon‐induced arterial injury in hypercholesterolemic rats. Vascul Pharmacol 51: 205‐213, 2009.
 178. Sonoda J, Xie W, Rosenfeld JM, Barwick JL, Guzelian PS, Evans RM. Regulation of a xenobiotic sulfonation cascade by nuclear pregnane X receptor (PXR). Proc Natl Acad Sci U S A 99: 13801‐13806, 2002.
 179. Staels B. A review of bile acid sequestrants: Potential mechanism(s) for glucose‐lowering effects in type 2 diabetes mellitus. Postgrad Med 121: 25‐30, 2009.
 180. Staels B, Kuipers F. Bile acid sequestrants and the treatment of type 2 diabetes mellitus. Drugs 67: 1383‐1392, 2007.
 181. Stayrook KR, Bramlett KS, Savkur RS, Ficorilli J, Cook T, Christe ME, Michael LF, Burris TP. Regulation of carbohydrate metabolism by the farnesoid x receptor. Endocrinology 146: 984‐991, 2005.
 182. Strautnieks SS, Bull LN, Knisely AS, Kocoshis SA, Dahl N, Arnell H, Sokal E, Dahan K, Childs S, Ling V, Tanner MS, Kagalwalla AF, Nemeth A, Pawlowska J, Baker A, Mieli‐Vergani G, Freimer NB, Gardiner RM, Thompson RJ. A gene encoding a liver‐specific ABC transporter is mutated in progressive familial intrahepatic cholestasis. Nat Genet 20: 233‐238, 1998.
 183. Studer E, Zhou X, Zhao R, Wang Y, Takabe K, Nagahashi M, Pandak WM, Dent P, Spiegel S, Shi R, Xu W, Liu X, Bohdan P, Zhang L, Zhou H, Hylemon PB. Conjugated bile acids activate the sphingosine‐1‐phosphate receptor 2 in primary rodent hepatocytes. Hepatology 55: 267‐276, 2012.
 184. Subbiah MTR, Yunker RL. Cholesterol 7a‐hydroxylase of rat liver: an insulin sensitive enzyme. Biochem Biophys Res Commun 124: 896‐902, 1984.
 185. Subramaniam P, Clayton PT, Portmann BC, Mieli‐Vergani G, Hadzic N. Variable clinical spectrum of the most common inborn error of bile acid metabolism–3beta‐hydroxy‐Delta 5‐C27‐steroid dehydrogenase deficiency. J Pediatr Gastroenterol Nutr 50: 61‐66, 2010.
 186. Temel RE, Brown JM. Biliary and nonbiliary contributions to reverse cholesterol transport. Curr Opin Lipidol 23: 85‐90, 2012.
 187. Thomas C, Auwerx J, Schoonjans K. Bile acids and the membrane bile acid receptor TGR5–connecting nutrition and metabolism. Thyroid 18: 167‐174, 2008.
 188. Thomas C, Gioiello A, Noriega L, Strehle A, Oury J, Rizzo G, Macchiarulo A, Yamamoto H, Mataki C, Pruzanski M, Pellicciari R, Auwerx J, Schoonjans K. TGR5‐mediated bile acid sensing controls glucose homeostasis. Cell Metab 10: 167‐177, 2009.
 189. Thomas C, Pellicciari R, Pruzanski M, Auwerx J, Schoonjans K. Targeting bile‐acid signalling for metabolic diseases. Nat Rev Drug Discov 7: 678‐693, 2008.
 190. Tiniakos DG, Vos MB, Brunt EM. Nonalcoholic fatty liver disease: Pathology and pathogenesis. Annu Rev Pathol 5: 145‐171, 2010.
 191. Tomlinson E, Fu L, John L, Hultgren B, Huang X, Renz M, Stephan JP, Tsai SP, Powell‐Braxton L, French D, Stewart TA. Transgenic mice expressing human fibroblast growth factor‐19 display increased metabolic rate and decreased adiposity. Endocrinology 143: 1741‐1747, 2002.
 192. Trauner M, Meier PJ, Boyer JL. Molecular pathogenesis of cholestasis. N Engl J Med 339: 1217‐1227, 1998.
 193. Tu H, Okamoto AY, Shan B. FXR, a bile acid receptor and biological sensor. Trends Cardiovasc Med 10: 30‐35, 2000.
 194. Uppal H, Toma D, Saini SP, Ren S, Jones TJ, Xie W. Combined loss of orphan receptors PXR and CAR heightens sensitivity to toxic bile acids in mice. Hepatology 41: 168‐176, 2005.
 195. Van Mil SW, Milona A, Dixon PH, Mullenbach R, Geenes VL, Chambers J, Shevchuk V, Moore GE, Lammert F, Glantz AG, Mattsson LA, Whittaker J, Parker MG, White R, Williamson C. Functional variants of the central bile acid sensor FXR identified in intrahepatic cholestasis of pregnancy. Gastroenterology 133: 507‐516, 2007.
 196. Vassileva G, Golovko A, Markowitz L, Abbondanzo SJ, Zeng M, Yang S, Hoos L, Tetzloff G, Levitan D, Murgolo NJ, Keane K, Davis HR, Jr., Hedrick J, Gustafson EL. Targeted deletion of Gpbar1 protects mice from cholesterol gallstone formation. Biochem J 398: 423‐430, 2006.
 197. Vavassori P, Mencarelli A, Renga B, Distrutti E, Fiorucci S. The bile acid receptor FXR is a modulator of intestinal innate immunity. J. Immunol 183: 6251‐6261, 2009.
 198. Wagner M, Zollner G, Trauner M. New molecular insights into the mechanisms of cholestasis. J Hepatol 51: 565‐580, 2009.
 199. Wang H, Chen J, Hollister K, Sowers LC, Forman BM. Endogenous bile acids are ligands for the nuclear receptor FXR/BAR. Mol Cell 3: 543‐553, 1999.
 200. Wang J, Gafvels M, Rudling M, Murphy C, Bjorkhem I, Einarsson C, Eggertsen G. Critical role of cholic acid for development of hypercholesterolemia and gallstones in diabetic mice. Biochem Biophys Res Commun 342: 1382‐1388, 2006.
 201. Wang L, Lee YK, Bundman D, Han Y, Thevananther S, Kim CS, Chua SS, Wei P, Heyman RA, Karin M, Moore DD. Redundant pathways for negative feedback regulation of bile Acid production. Dev Cell 2: 721‐731., 2002.
 202. Wang L, Liu J, Saha P, Huang J, Chan L, Spiegelman B, Moore DD. The orphan nuclear receptor SHP regulates PGC‐1alpha expression and energy production in brown adipocytes. Cell Metab 2: 227‐238, 2005.
 203. Wang YD, Chen WD, Wang M, Yu D, Forman BM, Huang W. Farnesoid X receptor antagonizes nuclear factor kappaB in hepatic inflammatory response. Hepatology 48: 1632‐1643, 2008.
 204. Wang YD, Chen WD, Yu D, Forman BM, Huang W. The G‐Protein‐coupled bile acid receptor, Gpbar1 (TGR5), negatively regulates hepatic inflammatory response through antagonizing nuclear factor kappa light‐chain enhancer of activated B cells (NF‐kappaB) in mice. Hepatology 54: 1421‐1432, 2011.
 205. Wasmuth HE, Glantz A, Keppeler H, Simon E, Bartz C, Rath W, Mattsson LA, Marschall HU, Lammert F. Intrahepatic cholestasis of pregnancy: The severe form is associated with common variants of the hepatobiliary phospholipid transporter ABCB4 gene. Gut 56: 265‐270, 2007.
 206. Watanabe M, Horai Y, Houten SM, Morimoto K, Sugizaki T, Arita E, Mataki C, Sato H, Tanigawara Y, Schoonjans K, Itoh H, Auwerx J. Lowering bile acid pool size with a synthetic farnesoid X receptor (FXR) agonist induces obesity and diabetes through reduced energy expenditure. J Biol Chem 286: 26913‐26920, 2011.
 207. Watanabe M, Houten SM, Mataki C, Christoffolete MA, Kim BW, Sato H, Messaddeq N, Harney JW, Ezaki O, Kodama T, Schoonjans K, Bianco AC, Auwerx J. Bile acids induce energy expenditure by promoting intracellular thyroid hormone activation. Nature 439: 484‐489, 2006.
 208. Watanabe M, Houten SM, Wang L, Moschetta A, Mangelsdorf DJ, Heyman RA, Moore DD, Auwerx J. Bile acids lower triglyceride levels via a pathway involving FXR, SHP, and SREBP‐1c. J Clin Invest 113: 1408‐1418, 2004.
 209. Weingartner O, Laufs U, Bohm M, Lutjohann D. An alternative pathway of reverse cholesterol transport: The oxysterol 27‐hydroxycholesterol. Atherosclerosis 209: 39‐41, 2010.
 210. Wilkinson DS, Ogden SK, Stratton SA, Piechan JL, Nguyen TT, Smulian GA, Barton MC. A direct intersection between p53 and transforming growth factor beta pathways targets chromatin modification and transcription repression of the alpha‐fetoprotein gene. Mol Cell Biol 25: 1200‐1212, 2005.
 211. Wittenburg H, Lyons MA, Li R, Churchill GA, Carey MC, Paigen B. FXR and ABCG5/ABCG8 as determinants of cholesterol gallstone formation from quantitative trait locus mapping in mice. Gastroenterology 125: 868‐881, 2003.
 212. Xie W, Radominska‐Pandya A, Shi Y, Simon CM, Nelson MC, Ong ES, Waxman DJ, Evans RM. An essential role for nuclear receptors SXR/PXR in detoxification of cholestatic bile acids. Proc Natl Acad Sci U S A 98: 3375‐3380., 2001.
 213. Xie Y, Blanc V, Kerr TA, Kennedy S, Luo J, Newberry EP, Davidson NO. Decreased expression of cholesterol 7{alpha}‐hydroxylase and altered bile acid metabolism in apobec‐1‐/‐ mice lead to increased gallstone susceptibility. J Biol Chem 284: 16860‐16871, 2009.
 214. Yabe D, Brown MS, Goldstein JL. Insig‐2, a second endoplasmic reticulum protein that binds SCAP and blocks export of sterol regulatory element‐binding proteins. Proc Natl Acad Sci U S A 99: 12753‐12758, 2002.
 215. Yamagata K, Daitoku H, Shimamoto Y, Matsuzaki H, Hirota K, Ishida J, Fukamizu A. Bile acids regulate gluconeogenic gene expression via small heterodimer partner‐mediated repression of hepatocyte nuclear factor 4 and Foxo1. J Biol Chem 279: 23158‐23165, 2004.
 216. Yeh MM, Brunt EM. Pathology of nonalcoholic fatty liver disease. Am J Clin Pathol 128: 837‐847, 2007.
 217. Yu C, Wang F, Jin C, Huang X, McKeehan WL. Independent repression of bile acid synthesis and activation of c‐Jun N‐terminal kinase (JNK) by activated hepatocyte fibroblast growth factor receptor 4 (FGFR4) and bile acids. J Biol Chem 280: 17707‐17714, 2005.
 218. Yu C, Wang F, Kan M, Jin C, Jones RB, Weinstein M, Deng CX, McKeehan WL. Elevated cholesterol metabolism and bile acid synthesis in mice lacking membrane tyrosine kinase receptor FGFR4. J Biol Chem 275: 15482‐15489, 2000.
 219. Zhang M, Chiang JY. Transcriptional regulation of the human sterol 12α‐hydroxylase gene (CYP8B1): Roles of hepatocyte nuclear factor 4α (HNF4α) in mediating bile acid repression. J Biol Chem 276: 41690‐41699, 2001.
 220. Zhang Y, Breevoort SR, Angdisen J, Fu M, Schmidt DR, Holmstrom SR, Kliewer SA, Mangelsdorf DJ, Schulman IG. Liver LXRalpha expression is crucial for whole body cholesterol homeostasis and reverse cholesterol transport in mice. J Clin Invest 122: 1688‐1699, 2012.
 221. Zhang Y, Edwards PA. FXR signaling in metabolic disease. FEBS Lett 582: 10‐18, 2008.
 222. Zhang Y, Lee FY, Barrera G, Lee H, Vales C, Gonzalez FJ, Willson TM, Edwards PA. Activation of the nuclear receptor FXR improves hyperglycemia and hyperlipidemia in diabetic mice. Proc Natl Acad Sci U S A 103: 1006‐1011, 2006.
 223. Zollner G, Marschall HU, Wagner M, Trauner M. Role of nuclear receptors in the adaptive response to bile acids and cholestasis: Pathogenetic and therapeutic considerations. Mol Pharm 3: 231‐251, 2006.
 224. Zollner G, Trauner M. Nuclear receptors as therapeutic targets in cholestatic liver diseases. Br J Pharmacol 156: 7‐27, 2009.

Related Articles:

Diabetes and Obesity
Top cited articles of 2018

Contact Editor

Submit a note to the editor about this article by filling in the form below.

* Required Field

How to Cite

John Y. L. Chiang. Bile Acid Metabolism and Signaling. Compr Physiol 2013, 3: 1191-1212. doi: 10.1002/cphy.c120023