Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
Food Funct ; 13(20): 10665-10679, 2022 Oct 17.
Article in English | MEDLINE | ID: mdl-36172720

ABSTRACT

The improvement of lipid metabolism by capsaicin (CAP) has been extensively studied, mostly with respect to the vanilloid type 1 (TRPV1) ion channel and intestinal flora. In this study, a model was established in germ-free mice by using resiniferatoxin (RTX) to ablate TRPV1 ion channels. Bile acid composition, blood parameters, and colonic transcriptome analyses revealed that CAP could improve dyslipidemia caused by high-fat diet even in the absence of TRPV1 ion channels and intestinal flora. CAP fed to germ mice decreased the concentrations of low-density lipoprotein cholesterol (LDL-C), triglycerides (TG), fasting blood glucose and fasting insulin, increased the concentration of high-density lipoprotein (HDL-C), and decreased the levels of plasma endotoxin and pro-inflammatory factor interleukin 6 (IL-6). Furthermore, CAP could affect both classical and alternative pathways of cholesterol conversion by changing the composition of bile acids, reducing the concentrations of glycocholic acid (GCA), ursodeoxycholic acid (UDCA) and glycochenodeoxycholic acid (GCDCA). First, changing the composition of bile acids inhibited the expression of colon Fgf15. CAP promoted the expression of Cyp7a1 (Cytochrome p450, family 7, subfamily a, and polypeptide 1) in the liver, and thus reduced TC and TG levels. In addition, it could change the composition of bile acids and increase the expression of Cyp7b1 (Cytochrome p450, family 7, subfamily b, and polypeptide 1) in the colon, increase Cyp7b1 protein in the liver and thus inhibit fat accumulation. In conclusion, CAP could alter the composition of bile acids and promote the conversion of cholesterol to bile acids, thereby improving lipid metabolism abnormalities caused by a high-fat diet.


Subject(s)
Dyslipidemias , Insulins , Animals , Bile Acids and Salts/metabolism , Blood Glucose/metabolism , Capsaicin , Cholesterol/metabolism , Cholesterol 7-alpha-Hydroxylase/genetics , Cholesterol 7-alpha-Hydroxylase/metabolism , Cholesterol, LDL/metabolism , Diet, High-Fat/adverse effects , Dyslipidemias/drug therapy , Dyslipidemias/genetics , Dyslipidemias/metabolism , Endotoxins , Glycochenodeoxycholic Acid/metabolism , Insulins/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Lipoproteins, HDL , Liver/metabolism , Mice , Triglycerides/metabolism , Ursodeoxycholic Acid/metabolism
2.
Food Funct ; 12(10): 4315-4324, 2021 May 21.
Article in English | MEDLINE | ID: mdl-34031676

ABSTRACT

Bile salt hydrolase (BSH)-producing bacteria are negatively related to the body weight gain and energy storage of the host. We aimed to obtain a novel BSH-producing strain with excellent anti-obesity effect and explained its mechanism. Here, we selected a strain named Lactiplantibacillus plantarum H-87 (H-87) with excellent ability to hydrolyze glycochenodeoxycholic acid (GCDCA) and tauroursodeoxycholic acid (TUDCA) in vitro from 12 lactobacilli, and evaluated its anti-obesity effect in high-fat diet (HFD)-fed C57BL/6J mice. The results suggested that H-87 could inhibit HFD-induced body weight gain, fat accumulation, liver lipogenesis and injury, insulin resistance and dyslipidemia. In addition, H-87 could colonize in the ileum and hydrolyze GCDCA and TUDCA, reflected as changes in the concentrations of GCDCA, TUDCA, CDCA and UDCA in the ileum or liver. Furthermore, the study identified that H-87 reduced TUDCA and GCDCA levels in the ileum, which decreased the GLP-1 secretion by L cells to alleviate insulin resistance in HFD-fed mice. Furthermore, H-87 increased the CDCA level in the ileum and liver to activate FXR signaling pathways to inhibit liver lipogenesis in HFD-fed mice. In addition, the decrease of intestinal conjugated bile acids (TUDCA and GCDCA) also increased fecal lipid content and decreased intestinal lipid digestibility. In conclusion, H-87 could inhibit liver fat deposition, insulin resistance and lipid digestion by changing bile acid enterohepatic circulation, and eventually alleviate HFD-induced obesity.


Subject(s)
Diet, High-Fat/adverse effects , Lactobacillus plantarum/metabolism , Obesity/microbiology , Obesity/prevention & control , Animals , Bile Acids and Salts/metabolism , Dyslipidemias/microbiology , Dyslipidemias/prevention & control , Glycochenodeoxycholic Acid/metabolism , Insulin Resistance , Lipid Metabolism , Liver/metabolism , Liver Diseases/microbiology , Liver Diseases/prevention & control , Male , Mice , Mice, Inbred C57BL , Obesity/etiology , Taurochenodeoxycholic Acid/metabolism
3.
Int J Biol Macromol ; 177: 111-118, 2021 Apr 30.
Article in English | MEDLINE | ID: mdl-33592267

ABSTRACT

7α-Hydroxysteroid dehydrogenase (7α-HSDH) plays an important role in the efficient biotransformation of taurochenodeoxycholic acid (TCDCA) to tauroursodeoxycholic acid (TUDCA). In this paper, a novel NADP(H)-dependent 7α-HSDH (named J-1-1) was discovered, heterologously expressed in Escherichia coli and biochemically characterized. J-1-1 exhibited high enzymatic activities. The specific activities of J-1-1 toward TCDCA, glycochenodeoxycholic acid (GCDCA) and ethyl benzoylacetate (EBA) were 188.3 ± 0.2, 217.6 ± 0.4, and 20.0 ± 0.2 U·mg-1, respectively, in 50 mM Glycine-NaOH, pH 10.5. Simultaneously, J-1-1 showed high thermostability; 73% of its activity maintained after heat treatment at 40 °C for 100 h. Particularly noteworthy is that magnesium ion could stabilize the structure of J-1-1, resulting in the enhancement of its enzymatic activity and thermostability. The enzymatic activity of J-1-1 increased 40-fold in the presence of 50 mM Mg2+, and T0.5 increased by approximately 6 °C. Furthermore, after heat treatment at 40 °C for 20 min, the control group only retained 52% of the residual enzyme activity, while the residual enzyme activity of the experimental group was still 77% of the J-1-1 enzyme activity with Mg2+ and without heat treatment. These properties of 7α-HSDH would be expected to contribute to more extensive applications in the biotransformation of related substrates.


Subject(s)
Hydroxysteroid Dehydrogenases/genetics , Hydroxysteroid Dehydrogenases/metabolism , Ions/metabolism , Magnesium/metabolism , Amino Acid Sequence , Biotransformation/immunology , Escherichia coli/genetics , Escherichia coli/metabolism , Glycochenodeoxycholic Acid/genetics , Glycochenodeoxycholic Acid/metabolism , Sequence Alignment , Taurochenodeoxycholic Acid/genetics
4.
Clin Pharmacol Ther ; 109(3): 646-657, 2021 03.
Article in English | MEDLINE | ID: mdl-32961594

ABSTRACT

The aim of this study was to investigate the sensitivity and specificity of endogenous glycochenodeoxycholate and glycodeoxycholate 3-O-glucuronides (GCDCA-3G and GDCA-3G) as substrates for organic anion transporting polypeptide 1B1 (OATP1B1) in humans. We measured fasting levels of plasma GCDCA-3G and GDCA-3G using liquid chromatography-tandem mass spectrometry in 356 healthy volunteers. The mean plasma levels of both compounds were ~ 50% lower in women than in men (P = 2.25 × 10-18 and P = 4.73 × 10-9 ). In a microarray-based genome-wide association study, the SLCO1B1 rs4149056 (c.521T>C, p.Val174Ala) variation showed the strongest association with the plasma GCDCA-3G (P = 3.09 × 10-30 ) and GDCA-3G (P = 1.60 × 10-17 ) concentrations. The mean plasma concentration of GCDCA-3G was 9.2-fold (P = 8.77 × 10-31 ) and that of GDCA-3G was 6.4-fold (P = 2.45x10-13 ) higher in individuals with the SLCO1B1 c.521C/C genotype than in those with the c.521T/T genotype. No other variants showed independent genome-wide significant associations with GCDCA-3G or GDCA-3G. GCDCA-3G was highly efficacious in detecting the SLCO1B1 c.521C/C genotype with an area under the receiver operating characteristic curve of 0.996 (P < 0.0001). The sensitivity (98-99%) and specificity (100%) peaked at a cutoff value of 180 ng/mL for men and 90 ng/mL for women. In a haplotype-based analysis, SLCO1B1*5 and *15 were associated with reduced, and SLCO1B1*1B, *14, and *35 with increased OATP1B1 function. In vitro, both GCDCA-3G and GDCA-3G showed at least 6 times higher uptake by OATP1B1 than OATP1B3 or OATP2B1. These data indicate that the hepatic uptake of GCDCA-3G and GDCA-3G is predominantly mediated by OATP1B1. GCDCA-3G, in particular, is a highly sensitive and specific OATP1B1 biomarker in humans.


Subject(s)
Glucuronides/metabolism , Glycochenodeoxycholic Acid/metabolism , Liver-Specific Organic Anion Transporter 1/metabolism , Liver/metabolism , Adult , Biomarkers/metabolism , Chromatography, Liquid , Female , Genome-Wide Association Study , Genotype , Glucuronides/blood , Glycochenodeoxycholic Acid/blood , HEK293 Cells , Healthy Volunteers , Humans , Liver-Specific Organic Anion Transporter 1/deficiency , Liver-Specific Organic Anion Transporter 1/genetics , Male , Metabolic Detoxication, Phase II , Oligonucleotide Array Sequence Analysis , Pharmacogenomic Variants , Phenotype , Polymorphism, Single Nucleotide , Tandem Mass Spectrometry , Young Adult
5.
Cell Physiol Biochem ; 52(6): 1427-1445, 2019.
Article in English | MEDLINE | ID: mdl-31088037

ABSTRACT

BACKGROUND/AIMS: Hydrophobic bile salts, such as glycochenodeoxycholate (GCDC) can trigger hepatocyte apoptosis, which is prevented by tauroursodesoxycholate (TUDC), but the effects of GCDC and TUDC on sinusoidal bile salt uptake via the Na⁺-taurocholate transporting polypeptide (Ntcp) are unclear. METHODS: The effects of GCDC and TUDC on the plasma membrane localization of Ntcp were studied in perfused rat liver by means of immunofluorescence analysis and super-resolution microscopy. The underlying signaling events were investigated by Western blotting and inhibitor studies. RESULTS: GCDC (20 µmol/l) induced within 60 min a retrieval of Ntcp from the basolateral membrane into the cytosol, which was accompanied by an activating phosphorylation of the Src kinases Fyn and Yes. Both, Fyn activation and the GCDC-induced Ntcp retrieval from the plasma membrane were sensitive to the NADPH oxidase inhibitor apocynin, the antioxidant N-acetylcysteine and the Src family kinase inhibitors SU6656 and PP-2, whereas PP-2 did not inhibit GCDC-induced Yes activation. Internalization of Ntcp by GCDC was also prevented by the protein kinase C (PKC) inhibitor Gö6850. TUDC (20 µmol/l) reversed the GCDC-induced retrieval of Ntcp from the plasma membrane and prevented the activation of Fyn and Yes in GCDC-perfused rat livers. Reinsertion of Ntcp into the basolateral membrane in GCDC-perfused livers by TUDC was sensitive to the protein kinase A (PKA) inhibitor H89 and the integrin-inhibitory peptide GRGDSP, whereas the control peptide GRADSP was ineffective. Ex posure of cultured rat hepatocytes to GCDC (50 µmol/l, 15min) increased the fluorescence intensity of the reactive oxygen fluorescent indicator DCF to about 1.6-fold of untreated controls in a TUDC (50 µmol/l)-sensitive way. GCDC caused a TUDC-sensitive canalicular dilatation without evidence for Bsep retrieval from the canalicular membrane. CONCLUSION: The present study suggests that GCDC triggers the retrieval of Ntcp from the basolateral membrane into the cytosol through an oxidative stress-dependent activation of Fyn. TUDC prevents the GCDC-induced Fyn activation and Ntcp retrieval through integrin-dependent activation of PKA.


Subject(s)
Cell Membrane/metabolism , Glycochenodeoxycholic Acid , Liver/metabolism , Organic Anion Transporters, Sodium-Dependent/metabolism , Symporters/metabolism , Taurocholic Acid , Animals , Gene Expression Regulation/drug effects , Glycochenodeoxycholic Acid/metabolism , Glycochenodeoxycholic Acid/pharmacology , Male , Protein Transport/drug effects , Rats , Rats, Wistar , Signal Transduction/drug effects , Taurocholic Acid/metabolism , Taurocholic Acid/pharmacology
6.
Cancer Lett ; 454: 215-223, 2019 07 10.
Article in English | MEDLINE | ID: mdl-30980867

ABSTRACT

Metastasis and recurrence severely impact the treatment effect of hepatocellular carcinoma (HCC). HCC complicated with cholestasis is more prone to recurrence and metastasis. Previous studies have implicated pathogenesis of HCC by bile acid; however, the underlying mechanism is unknown yet. Glycochenodeoxycholate (GCDC) is one of most important component of bile acid (BA). In the present study, the role of GCDC in HCC cells invasion was detected by in vitro and in vivo assays. GCDC was found to significantly enhance the invasive potential of HCC cells; Further studies showed that GCDC could induce autophagy activation and higher invasive capability in HCC cells. Interestingly, inhibition of autophagy by chloroquine (CQ) reversed this phenomenon. Subsequently, the correlation between TBA expression level and clinicopathological characteristics was analyzed in HCC patients. Clinically, high TBA level in HCC tissue was found to be associated with more invasive and poor survival in HCC patients. Mechanistic study showed that bile acid induced autophagy by targeting the AMPK/mTOR pathway in HCC cells. Therefore, our results suggest that bile acid may promote HCC invasion via activation of autophagy and the level of bile acid may serve as a potential useful indicator for prognosis of HCC patients.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Carcinoma, Hepatocellular/metabolism , Glycochenodeoxycholic Acid/metabolism , Liver Neoplasms/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Autophagy/drug effects , Autophagy/physiology , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/physiology , Female , Glycochenodeoxycholic Acid/pharmacology , Humans , Liver Neoplasms/chemically induced , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Neoplasm Invasiveness
7.
J Pharm Biomed Anal ; 165: 112-118, 2019 Feb 20.
Article in English | MEDLINE | ID: mdl-30529824

ABSTRACT

Colesevelam hydrochloride is a bile acid sequestrant used as a low density lipoprotein (LDL) reducing agent in hyperlipidemia with an additional advantage to improve glycemic control in type 2 diabetes patients. The objective of the study was to develop and validate a liquid chromatography tandem mass spectroscopic method for the simultaneous in-vitro estimation of bile acid salts of Glycocholic acid (GC), Glycochenodeoxycholic acid (GCDC) and Taurodeoxycholic acid (TDC) and its application in performing in-vitro binding study with Colesevelam Hydrochloride tablets. The method was developed using C-18 (50 x 4.6 mm, 3 µm) column with detection on negative ion mode and acquisition time of 3.5 min. The calibration range was linear from 0.0002 mM to 0.0065 mM for GC, 0.0002 mM to 0.0065 mM for GCDC and 0.0001 mM to 0.0021 mM for TDC. The precision was less than 3.0% and accuracy was found well within the range of 85 to 115%. The validated method was further applied to conduct in-vitro equilibrium binding study. The data was subjected to Langmuir isotherm and affinity constant (k1) and capacity constant (k2) were calculated.


Subject(s)
Anticholesteremic Agents/metabolism , Chromatography, High Pressure Liquid/methods , Colesevelam Hydrochloride/metabolism , Tandem Mass Spectrometry/methods , Calibration , Glycochenodeoxycholic Acid/metabolism , Glycocholic Acid/metabolism , Reproducibility of Results , Tablets , Taurodeoxycholic Acid/metabolism
8.
Am J Physiol Gastrointest Liver Physiol ; 314(5): G597-G609, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29420067

ABSTRACT

The heteromeric steroid transporter organic solute transporter α/ß (OSTα/ß, SLC51A/B) was discovered over a decade ago, but its physiological significance in the liver remains uncertain. A major challenge has been the lack of suitable models expressing OSTα/ß. Based on observations first reported here that hepatic OSTα/ß is upregulated in nonalcoholic steatohepatitis, the aim of this research was to develop an in vitro model to evaluate OSTα/ß function and interaction with drugs and bile acids. OSTα/ß expression in human liver tissue was analyzed by quantitative RT-PCR, Western blotting, and immunofluorescence. Radiolabeled compounds were used to determine OSTα/ß-mediated transport in the established in vitro model. The effect of bile acids and drugs, including those associated with cholestatic drug-induced liver injury, on OSTα/ß-mediated transport was evaluated. Expression of OSTα/ß was elevated in the liver of patients with nonalcoholic steatohepatitis and primary biliary cholangitis, whereas hepatocyte expression of OSTα/ß was low in control liver tissue. Studies in the novel cell-based system showed rapid and linear OSTα/ß-mediated transport for all tested compounds: dehydroepiandrosterone sulfate, digoxin, estrone sulfate, and taurocholate. The interaction study with 26 compounds revealed novel OSTα/ß inhibitors: a biomarker for cholestasis, glycochenodeoxycholic acid; the major metabolite of troglitazone, troglitazone sulfate; and a macrocyclic antibiotic, fidaxomicin. Additionally, some drugs (e.g., digoxin) consistently stimulated taurocholate uptake in OSTα/ß-overexpressing cells. Our findings demonstrate that OSTα/ß is an important transporter in liver disease and imply a role for this transporter in bile acid-bile acid and drug-bile acid interactions, as well as cholestatic drug-induced liver injury. NEW & NOTEWORTHY The organic solute transporter OSTα/ß is highly expressed in hepatocytes of liver tissue obtained from patients with nonalcoholic steatohepatitis and primary biliary cholangitis. OSTα/ß substrates exhibit rapid, linear, and concentration-driven transport in an OSTα/ß-overexpressing cell line. Drugs associated with hepatotoxicity modulate OSTα/ß-mediated taurocholate transport. These data suggest that hepatic OSTα/ß plays an essential role in patients with cholestasis and may have important clinical implications for bile acid and drug disposition.


Subject(s)
Chemical and Drug Induced Liver Injury/metabolism , Hepatocytes/metabolism , Liver Cirrhosis, Biliary/metabolism , Membrane Transport Proteins/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Taurocholic Acid/metabolism , Bile Acids and Salts/metabolism , Biological Transport/physiology , Biomarkers/metabolism , Cell Line , Cholestasis/metabolism , Female , Glycochenodeoxycholic Acid/metabolism , Humans , Liver/metabolism , Male , Middle Aged
9.
Oncol Rep ; 38(3): 1742-1750, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28731137

ABSTRACT

Hepatocellular carcinoma (HCC) is a highly malignant tumor and can evolve rapidly to resistance to chemotherapies. Glycochenodeoxycholate (GCDA), which is toxic and hydrophobic, is the main ingredient in the bile and associated with carcinogenesis of gastrointenstinal tumors. Bcl-2 is the most important anti-apoptotic protein and overexpressed in various human tumors. In the present study, we found that GCDA can induce the chemoresistance of human liver cancer cells and specific depletion of Bcl-2 by RNA interference blocks GCDA-stimulated chemoresistance, which indicate the pivotal role of Bcl-2 in such process. Mechanistically, GCDA simultaneously stimulates phosphorylation of Bcl-2 at Ser70 site and activates extracellular signal-regulated kinase 1/2 (ERK1/2), and inhibition of ERK1/2 by PD98059 (MAPK/ERK1/2 inhibitor) or siRNA (targeting ERK1/2) suppresses GCDA-stimulated phosphorylation of Bcl-2 and significantly attenuates the survival and chemoresistance induced by GCDA in liver cancer cells. Thus, GCDA-induced survival and chemoresistance of liver cancer cells may occur through activation of Bcl-2 by phosphorylation at Ser70 site through MAPK/ERK1/2 pathway, which may contribute to the development of human liver cancer and chemoresistance.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Drug Resistance, Neoplasm/physiology , Glycochenodeoxycholic Acid/metabolism , Phosphorylation/physiology , Proto-Oncogene Proteins c-bcl-2/metabolism , Apoptosis/physiology , Apoptosis Regulatory Proteins/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/physiology , Hep G2 Cells , Humans , Liver Neoplasms , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinases/metabolism , RNA Interference/physiology , Signal Transduction/physiology
10.
Anal Sci ; 33(5): 573-578, 2017.
Article in English | MEDLINE | ID: mdl-28496060

ABSTRACT

A method was developed for analyzing broad spectrum small molecule metabolites in the serum of hepatocellular carcinoma (HCC) patients based on ultrafast liquid chromatography-ion trap-time of flight tandem mass spectrometry (UFLC-IT-TOF MS). Serum samples were collected from 80 HCC patients and healthy persons. After pretreatment process for protein precipitation, the supernatant was analyzed with the UFLC-IT-TOF MS to obtain information on the metabonomics of small molecules. The eight compounds of glycocholic acid, choline glycerophosphate, acetyl-L-phenylalanine, oleamide, tetradecanamide, acetylcarnitine, lysolecithin and glycochenodeoxycholic acid in the HCC group were identified with significant differences from those in the health group (P <0.01). By using multidimensional analysis of variation coefficient and principal component analysis for the repeatability and 48 h stability, the method was demonstrated to have good repeatability, excellent precision, and high stability, which can satisfy the metabonomics research requirement. The high throughput and practical usability of the method further shows perspective for metabonomic analysis of large-batch serum samples.


Subject(s)
Carcinoma, Hepatocellular/chemistry , Small Molecule Libraries/metabolism , Acetylcarnitine/analysis , Acetylcarnitine/metabolism , Amides/analysis , Amides/metabolism , Carcinoma, Hepatocellular/metabolism , Chromatography, High Pressure Liquid , Glycerylphosphorylcholine/analysis , Glycerylphosphorylcholine/metabolism , Glycochenodeoxycholic Acid/analysis , Glycochenodeoxycholic Acid/metabolism , Glycocholic Acid/analysis , Glycocholic Acid/metabolism , Humans , Lysophosphatidylcholines/analysis , Lysophosphatidylcholines/metabolism , Oleic Acids/analysis , Oleic Acids/metabolism , Phenylalanine/analysis , Phenylalanine/metabolism , Small Molecule Libraries/analysis , Tandem Mass Spectrometry
11.
Pharm Res ; 34(8): 1601-1614, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28550384

ABSTRACT

PURPOSE: To assess the use of glycochenodeoxycholate-3-sulfate (GCDCA-S) and chenodeoxycholate 3- or 24-glucuronide (CDCA-3G or -24G) as surrogate endogenous substrates in the investigation of drug interactions involving OATP1B1 and OATP1B3. METHODS: Uptake of GCDCA-S and CDCA-24G was examined in HEK293 cells transfected with cDNA for OATP1B1, OATP1B3, and NTCP and in cryopreserved human hepatocytes. Plasma concentrations of bile acids and their metabolites (GCDCA-S, CDCA-3G, and CDCA-24G) were determined by LC-MS/MS in eight healthy volunteers with or without administration of rifampicin (600 mg, po). RESULTS: GCDCA-S and CDCA-24G were substrates for OATP1B1, OATP1B3, and NTCP. The uptake of [3H]atorvastatin, GCDCA-S, and CDCA-24G by human hepatocytes was significantly inhibited by both rifampicin and pioglitazone, whereas that of taurocholate was inhibited only by pioglitazone. Rifampicin elevated plasma concentrations of GCDCA-S more than those of other bile acids. The area under the plasma concentration-time curve for GCDCA-S was 20.3 times higher in rifampicin-treated samples. CDCA-24G could be detected only in plasma from the rifampicin-treatment phase, and CDCA-3G was undetectable in both phases. CONCLUSIONS: We identified GCDCA-S and CDCA-24G as substrates of NTCP, OATP1B1, and OATP1B3. GCDCA-S is a surrogate endogenous probe for the assessment of drug interactions involving hepatic OATP1B1 and OATP1B3.


Subject(s)
Chenodeoxycholic Acid/metabolism , Glucuronides/metabolism , Glycochenodeoxycholic Acid/analogs & derivatives , Liver-Specific Organic Anion Transporter 1/metabolism , Solute Carrier Organic Anion Transporter Family Member 1B3/metabolism , Adult , Atorvastatin/metabolism , Bile Acids and Salts/blood , Drug Interactions , Glycochenodeoxycholic Acid/metabolism , HEK293 Cells , Hepatocytes/metabolism , Humans , Male , Organic Anion Transporters, Sodium-Dependent/metabolism , Pioglitazone , Rifampin/pharmacology , Symporters/metabolism , Taurocholic Acid/pharmacology , Thiazolidinediones/pharmacology , Young Adult
12.
Anal Chem ; 88(16): 8172-8, 2016 08 16.
Article in English | MEDLINE | ID: mdl-27458657

ABSTRACT

Evidence is presented that binding isotherms, simple or biphasic, can be extracted directly from noninterpreted, complex 2D NMR spectra using principal component analysis (PCA) to reveal the largest trend(s) across the series. This approach renders peak picking unnecessary for tracking population changes. In 1:1 binding, the first principal component captures the binding isotherm from NMR-detected titrations in fast, slow, and even intermediate and mixed exchange regimes, as illustrated for phospholigand associations with proteins. Although the sigmoidal shifts and line broadening of intermediate exchange distorts binding isotherms constructed conventionally, applying PCA directly to these spectra along with Pareto scaling overcomes the distortion. Applying PCA to time-domain NMR data also yields binding isotherms from titrations in fast or slow exchange. The algorithm readily extracts from magnetic resonance imaging movie time courses such as breathing and heart rate in chest imaging. Similarly, two-step binding processes detected by NMR are easily captured by principal components 1 and 2. PCA obviates the customary focus on specific peaks or regions of images. Applying it directly to a series of complex data will easily delineate binding isotherms, equilibrium shifts, and time courses of reactions or fluctuations.


Subject(s)
Nuclear Magnetic Resonance, Biomolecular , Proteins/metabolism , Algorithms , Animals , Brain/diagnostic imaging , Brain/metabolism , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Glycochenodeoxycholic Acid/chemistry , Glycochenodeoxycholic Acid/metabolism , Magnetic Resonance Imaging , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/metabolism , Principal Component Analysis , Protein Binding , Proteins/chemistry
13.
Am J Physiol Gastrointest Liver Physiol ; 310(10): G865-73, 2016 05 15.
Article in English | MEDLINE | ID: mdl-26999807

ABSTRACT

The hepatotoxic bile acid glycochenodeoxycholate (GCDC) modulates hepatocyte cell death through activation of JNK, Akt, and Erk. The nonhepatotoxic bile acid taurocholate activates Akt and Erk through the sphingosine-1-phosphate receptor 2 (S1PR2). The role of the S1PR2 in GCDC-mediated apoptosis and kinase activation is unknown. Studies were done in rat hepatocytes, HUH7 cells, and HUH7 cells stably transfected with rat Ntcp (HUH7-Ntcp). Cells were treated with GCDC and apoptosis was monitored morphologically by Hoechst staining and biochemically by immunoblotting for the active cleaved fragment of caspase 3. Kinase activation was determined by immunoblotting with phospho-specific antibodies. JTE-013, an inhibitor of S1PR2, significantly attenuated morphological evidence of GCDC-induced apoptosis and prevented caspase 3 cleavage in rat hepatocytes and HUH7-Ntcp cells. In hepatocytes, JTE-013 mildly suppressed, augmented, and had no effect on GCDC-induced JNK, Akt, and Erk phosphorylation, respectively. Similar results were seen in HUH7-Ntcp cells except for mild suppression of JNK and Erk phosphorylation. Knockdown of S1PR2 in HUH7-Ntcp augmented Akt, inhibited JNK, and had no effect on Erk phosphorylation. GCDC failed to induce apoptosis or kinase activation in HUH7 cells. In conclusion, SIPR2 inhibition attenuates GCDC-induced apoptosis and inhibits and augments GCDC-induced JNK and Akt phosphorylation, respectively. In addition, GCDC must enter hepatocytes to mediate cell death or activate kinases. These results suggest that SIPR2 activation is proapoptotic in GCDC-induced cell death but that this effect is not due to direct ligation of the S1PR2 by the bile acid.


Subject(s)
Apoptosis , Carcinoma, Hepatocellular/metabolism , Glycochenodeoxycholic Acid/metabolism , Hepatocytes/metabolism , Liver Neoplasms/metabolism , Receptors, Lysosphingolipid/metabolism , Animals , Cell Line, Tumor , Cells, Cultured , Glycochenodeoxycholic Acid/toxicity , Humans , MAP Kinase Kinase 4/metabolism , MAP Kinase Signaling System , Male , Proto-Oncogene Proteins c-akt/metabolism , Pyrazoles/pharmacology , Pyridines/pharmacology , Rats , Rats, Wistar , Receptors, Lysosphingolipid/antagonists & inhibitors , Sphingosine-1-Phosphate Receptors
14.
Basic Clin Pharmacol Toxicol ; 118(2): 160-7, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26176423

ABSTRACT

Cholestatic liver injury is a pathological component of numerous disease states. Much of the current literature on cholestatic liver injury is derived from in vitro studies using rodent hepatocytes or cell lines transfected with bile acid (BA) uptake transporters. While these studies demonstrate BA-driven apoptosis, it is debatable whether these models reflect the human pathophysiology, as primary human hepatocytes undergo primarily necrosis. HepaRG cells are a bipotential, human hepatoma line that express apical and basolateral BA transporters. Thus, we sought to determine whether HepaRG cells could replicate the response of primary human hepatocytes to BA exposure in vitro. HepG2 cells, primary murine hepatocytes (PMH) or HepaRG cells, were exposed to taurocholic acid (TCA), or glycochenodeoxycholate (GCDC) and lactate dehydrogenase release were measured to determine cell death. Cell death occurred dose-responsively in HepaRG cells when exposed to GCDC; however, HepG2 cells died acutely only at very high concentrations of GCDC. In HepaRG cells, pre-treatment with the caspase inhibitor z-VD-FMK had no effect on cell death, indicating a lack of apoptotic cell death, and while c-jun N-terminal kinase (JNK) protein was activated by GCDC treatment in HepaRG cells, the inhibition of JNK did not protect. Although previous data indicate that TCA stimulates pro-inflammatory gene induction in PMH, there was no change in gene expression after TCA stimulation in HepaRG cells, which mimicked previous data found in primary human hepatocytes. These data provide evidence for HepaRG cells as a new model for the study of the effect of BA on human hepatocytes.


Subject(s)
Amino Acid Chloromethyl Ketones/pharmacology , Apoptosis/drug effects , Cholestasis/metabolism , Glycochenodeoxycholic Acid , Hepatocytes , Taurocholic Acid , Animals , Bile Acids and Salts/metabolism , Bile Acids and Salts/toxicity , Glycochenodeoxycholic Acid/metabolism , Glycochenodeoxycholic Acid/toxicity , Hep G2 Cells , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Protective Agents/pharmacology , Taurocholic Acid/metabolism , Taurocholic Acid/toxicity
15.
Cell Commun Signal ; 13: 28, 2015 Jun 09.
Article in English | MEDLINE | ID: mdl-26050734

ABSTRACT

BACKGROUND: In many cells, bile acids (BAs) have a multitude of effects, some of which may be mediated by specific receptors such the TGR5 or FXR receptors. In pancreas systemic BAs, as well as intra-ductal BAs from bile reflux, can affect pancreatic secretion. Extracellular ATP and purinergic signalling are other important regulators of similar secretory mechanisms in pancreas. The aim of our study was to elucidate whether there is interplay between ATP and BA signalling. RESULTS: Here we show that CDCA (chenodeoxycholic acid) caused fast and concentration-dependent ATP release from acini (AR42J) and duct cells (Capan-1). Taurine and glycine conjugated forms of CDCA had smaller effects on ATP release in Capan-1 cells. In duct monolayers, CDCA stimulated ATP release mainly from the luminal membrane; the releasing mechanisms involved both vesicular and non-vesicular secretion pathways. Duct cells were not depleted of intracellular ATP with CDCA, but acinar cells lost some ATP, as detected by several methods including ATP sensor AT1.03(YEMK). In duct cells, CDCA caused reversible increase in the intracellular Ca(2+) concentration [Ca(2 +)]i, which could be significantly inhibited by antagonists of purinergic receptors. The TGR5 receptor, expressed on the luminal side of pancreatic ducts, was not involved in ATP release and Ca(2+) signals, but could stimulate Na(+)/Ca(2+) exchange in some conditions. CONCLUSIONS: CDCA evokes significant ATP release that can stimulate purinergic receptors, which in turn increase [Ca(2+)]i. The TGR5 receptor is not involved in these processes but can play a protective role at high intracellular Ca(2+) conditions. We propose that purinergic signalling could be taken into consideration in other cells/organs, and thereby potentially explain some of the multifaceted effects of BAs.


Subject(s)
Adenosine Triphosphate/metabolism , Bile Acids and Salts/metabolism , Pancreas/metabolism , Signal Transduction , Animals , Calcium/metabolism , Cell Line , Chenodeoxycholic Acid/metabolism , Glycochenodeoxycholic Acid/metabolism , Humans , Pancreas/cytology , Rats , Receptors, G-Protein-Coupled/metabolism , Receptors, Purinergic P2/metabolism , Taurochenodeoxycholic Acid/metabolism
16.
Exp Biol Med (Maywood) ; 240(2): 252-60, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25125499

ABSTRACT

An increase of toxic bile acids such as glycochenodeoxycholic acid occurs during warm ischemia reperfusion causing cholestasis and damage in hepatocytes and intrahepatic biliary epithelial cells. We aim to test antiapoptosis effects of ursodeoxycholyl lysophosphatidylethanolamide under cholestatic induction by glycochenodeoxycholic acid treatment of mouse hepatocytes and hypoxia induction by cobalt chloride treatment of intrahepatic biliary epithelial cancer Mz-ChA-1cell line. Such treatments caused marked increases in apoptosis as evidenced by activation of caspase 3, caspase 8 and poly (ADP-ribose) polymerase-1. Co-treatment with ursodeoxycholyl lysophosphatidylethanolamide significantly inhibited these increases. Interestingly, ursodeoxycholyl lysophosphatidylethanolamide was able to increase expression of antiapoptotic cellular FLICE-inhibitory protein in both cell types. Ursodeoxycholyl lysophosphatidylethanolamide also prevented the decreases of myeloid cell leukemia sequence-1 protein in both experimental systems, and this protection was due to ursodeoxycholyl lysophosphatidylethanolamide's ability to inhibit ubiquitination-mediated degradation of myeloid cell leukemia sequence-1, and to increase the phosphorylation of GSK-3ß. In addition, ursodeoxycholyl lysophosphatidylethanolamide was able to prevent the decreased expression of another antiapoptotic cellular inhibitor of apoptosis 2 in cobalt chloride-treated Mz-ChA-1 cells. Hence, ursodeoxycholyl lysophosphatidylethanolamide mediated cytoprotection against apoptosis during toxic bile-acid and ischemic stresses by a mechanism involving accumulation of cellular FLICE-inhibitory protein, myeloid cell leukemia sequence-1 and cellular inhibitor of apoptosis 2 proteins. Ursodeoxycholyl lysophosphatidylethanolamide may thus be used as an agent to prevent hepatic ischemia reperfusion.


Subject(s)
Apoptosis Regulatory Proteins/biosynthesis , Apoptosis/drug effects , Cholestasis/metabolism , Liver , Phosphatidylethanolamines/pharmacology , Up-Regulation/drug effects , Ursodeoxycholic Acid/pharmacology , Animals , Cell Line, Tumor , Cholestasis/pathology , Glycochenodeoxycholic Acid/metabolism , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Liver/blood supply , Liver/metabolism , Liver/pathology , Mice , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Phosphorylation/drug effects
17.
PLoS One ; 8(8): e71773, 2013.
Article in English | MEDLINE | ID: mdl-23951244

ABSTRACT

BACKGROUND: Metformin is used in the treatment of Diabetes Mellitus type II and improves liver function in patients with non-alcoholic fatty liver disease (NAFLD). Metformin activates AMP-activated protein kinase (AMPK), the cellular energy sensor that is sensitive to changes in the AMP/ATP-ratio. AMPK is an inhibitor of mammalian target of rapamycin (mTOR). Both AMPK and mTOR are able to modulate cell death. AIM: To evaluate the effects of metformin on hepatocyte cell death. METHODS: Apoptotic cell death was induced in primary rat hepatocytes using either the bile acid glycochenodeoxycholic acid (GCDCA) or TNFα in combination with actinomycin D (actD). AMPK, mTOR and phosphoinositide-3 kinase (PI3K)/Akt were inhibited using pharmacological inhibitors. Apoptosis and necrosis were quantified by caspase activation, acridine orange staining and Sytox green staining respectively. RESULTS: Metformin dose-dependently reduces GCDCA-induced apoptosis, even when added 2 hours after GCDCA, without increasing necrotic cell death. Metformin does not protect against TNFα/ActD-induced apoptosis. The protective effect of metformin is dependent on an intact PI3-kinase/Akt pathway, but does not require AMPK/mTOR-signaling. Metformin does not inhibit NF-κB activation. CONCLUSION: Metformin protects against bile acid-induced apoptosis and could be considered in the treatment of chronic liver diseases accompanied by inflammation.


Subject(s)
Apoptosis/drug effects , Bile Acids and Salts/pharmacology , Hepatocytes/drug effects , Hepatocytes/metabolism , Hypoglycemic Agents/pharmacology , Metformin/pharmacology , AMP-Activated Protein Kinases/metabolism , Animals , Bile Acids and Salts/metabolism , Caspase 3/metabolism , Cell Membrane/metabolism , Dose-Response Relationship, Drug , Glycochenodeoxycholic Acid/metabolism , Hepatocytes/pathology , Male , NF-kappa B/metabolism , Necrosis/drug therapy , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
18.
Arch Biochem Biophys ; 530(2): 101-7, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23357279

ABSTRACT

The filamentous bacterium Streptomyces coelicolor has a complex life cycle involving the formation of hair-like aerial mycelia on the colony surface, which differentiate into chains of spores. Genes required for the initiation of aerial mycelium formation have been termed 'bld' (bald), describing the smooth, undifferentiated colonies of mutant strains. We report the identification of a new bld gene designated as sco3099 and biochemical analysis of its encoded enzyme, cytochrome P450 (P450, or CYP) 107U1. Deletion of sco3099 resulted in a mutant defective in aerial hyphae sporulation and sensitive to heat shock, indicating that P450 107U1 plays a key role in growth and development of S. coelicolor. This is the first P450 reported to participate in a sporulation process in Streptomycetes. The substrate and catalytic properties of P450 107U1 were further investigated in mass spectrometry-based metabolomic studies. Glycocholic acid (from the medium) was identified as a substrate of P450 107U1 and was oxidized to glyco-7-oxo-deoxycholic acid. Although this reaction is apparently not relevant to the observed sporulation deficiency, it suggests that P450 107U1 might exert its physiological function by oxidizing other steroid-like molecules.


Subject(s)
Anti-Bacterial Agents/biosynthesis , Bacterial Proteins/metabolism , Cytochrome P-450 Enzyme System/metabolism , Streptomyces coelicolor/enzymology , Bacterial Proteins/genetics , Cytochrome P-450 Enzyme System/genetics , Glycochenodeoxycholic Acid/analogs & derivatives , Glycochenodeoxycholic Acid/metabolism , Mutation , Oxidation-Reduction , Streptomyces coelicolor/genetics
19.
Biochemistry ; 51(9): 1848-61, 2012 Mar 06.
Article in English | MEDLINE | ID: mdl-22329738

ABSTRACT

Human ileal bile acid binding protein (I-BABP), a member of the family of intracellular lipid binding proteins, is thought to play a role in the enterohepatic circulation of bile salts. Previously, we have shown by stopped-flow fluorescence analysis that positive binding cooperativity exhibited by I-BABP in its interactions with glycocholate (GCA) and glycochenodeoxycholate (GCDA), the two primary bile salts in humans, is related to a slow conformational change in the protein. In this study, we used backbone (15)N relaxation nuclear magnetic resonance (NMR) techniques to obtain residue-specific information about the internal dynamics of apo I-BABP and the doubly ligated I-BABP:GCA:GCDA complex on various time scales. According to our NMR data, bile salt binding is accompanied by a slight rigidification of the (15)N-(1)H bond vectors on the picosecond to nanosecond time scale, with most pronounced changes occurring in the C-D region. In contrast to the minor effects of ligation on fast motions, relaxation dispersion NMR experiments indicate a marked difference between the two protein states on the microsecond to millisecond time scale. In the apo form, an extensive network of conformational fluctuations is detected throughout segments of the EFGHIJ ß-strands and the C-D loop, which cease upon complexation. Our NMR data are in agreement with a conformational selection model we proposed earlier for I-BABP and support the hypothesis of an allosteric mechanism of ligand binding. According to the NMR measurements, the helical cap region may have a less crucial role in mediating ligand entry and release than what has been indicated for fatty acid binding proteins.


Subject(s)
Hydroxysteroid Dehydrogenases/chemistry , Ileum/metabolism , Binding Sites , Glycochenodeoxycholic Acid/chemistry , Glycochenodeoxycholic Acid/metabolism , Glycocholic Acid/chemistry , Glycocholic Acid/metabolism , Humans , Hydroxysteroid Dehydrogenases/metabolism , Ligands , Magnetic Resonance Spectroscopy , Protein Conformation
20.
J Biol Chem ; 286(45): 39307-17, 2011 Nov 11.
Article in English | MEDLINE | ID: mdl-21917914

ABSTRACT

Ileal bile acid-binding proteins (I-BABP), belonging to the family of intracellular lipid-binding proteins, control bile acid trafficking in enterocytes and participate in regulating the homeostasis of these cholesterol-derived metabolites. I-BABP orthologues share the same structural fold and are able to host up to two ligands in their large internal cavities. However variations in the primary sequences determine differences in binding properties such as the degree of binding cooperativity. To investigate the molecular requirements for cooperativity we adopted a gain-of-function approach, exploring the possibility to turn the noncooperative chicken I-BABP (cI-BABP) into a cooperative mutant protein. To this aim we first solved the solution structure of cI-BABP in complex with two molecules of the physiological ligand glycochenodeoxycholate. A comparative structural analysis with closely related members of the same protein family provided the basis to design a double mutant (H99Q/A101S cI-BABP) capable of establishing a cooperative binding mechanism. Molecular dynamics simulation studies of the wild type and mutant complexes and essential dynamics analysis of the trajectories supported the role of the identified amino acid residues as hot spot mediators of communication between binding sites. The emerging picture is consistent with a binding mechanism that can be described as an extended conformational selection model.


Subject(s)
Fatty Acid-Binding Proteins/chemistry , Gastrointestinal Hormones/chemistry , Glycochenodeoxycholic Acid/chemistry , Models, Molecular , Amino Acid Substitution , Animals , Chickens , Fatty Acid-Binding Proteins/metabolism , Gastrointestinal Hormones/metabolism , Glycochenodeoxycholic Acid/metabolism , Mutation, Missense , Protein Structure, Tertiary , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...