Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
Toxicol Lett ; 331: 11-21, 2020 Oct 01.
Article in English | MEDLINE | ID: mdl-32439580

ABSTRACT

Cholestasis represents pathophysiologic syndromes defined as impaired bile flow from the liver. As an outcome, bile acids accumulate and promote hepatocyte injury, followed by liver cirrhosis and liver failure. Glycochenodeoxycholic acid (GCDCA) is relatively toxic and highly concentrated in bile and serum after cholestasis. However, the mechanism underlying GCDCA-induced hepatotoxicity remains unclear. In this study, we found that GCDCA inhibits autophagosome formation and impairs lysosomal function by inhibiting lysosomal proteolysis and increasing lysosomal pH, contributing to defects in autophagic clearance and subsequently leading to the death of L02 human hepatocyte cells. Notably, through tandem mass tag (TMT)-based quantitative proteomic analysis and database searches, 313 differentially expressed proteins were identified, of which 71 were increased and 242 were decreased in the GCDCA group compared with those in the control group. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis revealed that GCDCA suppressed the signaling pathway of transcription factor E3 (TFE3), which was the most closely associated with autophagic flux impairment. In contrast, GCDCA-inhibited lysosomal function and autophagic flux were efficiently attenuated by TFE3 overexpression. Specifically, the decreased expression of TFE3 was closely related to the disruption of reactive oxygen species (ROS) homeostasis, which could be prevented by inhibiting intracellular ROS with N-acetyl cysteine (NAC). In summary, our study is the first to demonstrate that manipulation of ROS/TFE3 signaling may be a therapeutic approach for antagonizing GCDCA-induced hepatotoxicity.


Subject(s)
Autophagy/drug effects , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Glycochenodeoxycholic Acid/toxicity , Hepatocytes/drug effects , Homeostasis/drug effects , Reactive Oxygen Species/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Bile Acids and Salts/metabolism , Cell Line , Gene Expression/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Lysosomes/drug effects , Proteomics
2.
J Cell Physiol ; 234(7): 10899-10906, 2019 07.
Article in English | MEDLINE | ID: mdl-30548625

ABSTRACT

OBJECTIVE: Glycochenodeoxycholate acid (GCDA) is a toxic component in bile salts. It plays an important role in the development and progression of liver cancer. In this study, we investigated the underlying mechanism of GCDA in hepatocarcinogenesis and chemotherapy resistance. MATERIALS AND METHODS: Cell proliferation was measured by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and clonality by Ki-67 and colony-formation assay. Apoptosis was examined by flow cytometry. Real-time polymerase chain reaction (PCR) and western blot analysis were used to measure messenger RNA and protein levels, respectively. Short hairpin RNA was used to silence signal transducer and activator of transcription 3 (Stat3) expression. RESULTS: Bile salts (GCDA) promoted the proliferation of hepatocellular carcinoma (HCC) cells (HepG2 and QGY-7703), and GCDA treatment reduced the chemosensitivity of 5-fluorouracil (5FU) in HepG2 and QGY-7703 cells. GCDA upregulated the expression of antiapoptosis proteins Mcl-1/Survivin/Bcl-2. GCDA had no discernible effect on basal protein level or subcellular localization of phosphorylated Stat3. 5FU increased the apoptosis of HepG2 cells with silenced Stat3 expression, but GCDA-induced chemoresistance was not reversed. CONCLUSIONS: GCDA-reduced HCC cell chemosensitivity may occur by upregulating antiapoptosis proteins Mcl-1/Survivin/Bcl-2. Stat3 may be a target for enhancing the chemosensitivity of hepatocellular carcinoma cells, but GCDA-induced chemoresistance is independent of Stat3.


Subject(s)
Carcinoma, Hepatocellular/pathology , Drug Resistance, Neoplasm/drug effects , Glycochenodeoxycholic Acid/toxicity , Liver Neoplasms/pathology , Antimetabolites, Antineoplastic/pharmacology , Apoptosis/drug effects , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Fluorouracil/pharmacology , Hep G2 Cells , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Survivin/genetics , Survivin/metabolism
3.
Tumour Biol ; 39(7): 1010428317712102, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28681695

ABSTRACT

In cholestasis, increases in bile acid levels result in the generation of reactive oxygen species and the induction of DNA damage and mutation. It is believed that bile acid accumulation is associated with liver tumorigenesis. However, the mechanism that underpins this phenomenon remains to be elucidated. Mcl-1, which is overexpressed in hepatic cells, is a pro-survival member of the Bcl-2 family. In this study, we observed that Mcl-1 potently suppresses the repair of bile acid-induced abasic (apurinic/apyrimidinic) sites in DNA lesions. Upon exposure of hepatic cells to glycochenodeoxycholate, one of the major conjugated human bile acids, we observed an increase in AP site accumulation along with induction of poly(ADP-ribose) polymerase and XRCC1 ( X-Ray Repair Cross Complementing 1). In addition, accumulation of Mcl-1 was observed in the nuclei of QGY-7703 cells in response to glycochenodeoxycholate stimulation. Knockdown of endogenous Mcl-1 by RNA interference significantly accelerated the repair of DNA lesions in glycochenodeoxycholate-treated cells. However, unlike XRCC1, poly(ADP-ribose) polymerase was induced following Mcl-1 knockdown. Conversely, poly(ADP-ribose) polymerase suppression was observed following glycochenodeoxycholate treatment of cells overexpressing Mcl-1. Moreover, AP-site counting analyses revealed that DNA repair activity was enhanced in cells overexpressing poly(ADP-ribose) polymerase under glycochenodeoxycholate stress conditions. It is well known that poly(ADP-ribose) polymerase plays a crucial role in the base excision repair pathway. Thus, our findings suggest that Mcl-1 suppresses base excision repair by inhibiting poly(ADP-ribose) polymerase induction following glycochenodeoxycholate-induced DNA damage. These results potentially explain how bile acid accumulation results in genetic instability and carcinogenesis.


Subject(s)
Cholestasis/genetics , DNA-Binding Proteins/genetics , Liver Neoplasms/genetics , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Poly(ADP-ribose) Polymerases/genetics , Apurinic Acid/genetics , Bile Acids and Salts/standards , Bile Acids and Salts/toxicity , Cholestasis/metabolism , Cholestasis/pathology , DNA Damage/drug effects , DNA Repair/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Glycochenodeoxycholic Acid/toxicity , Hep G2 Cells , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Reactive Oxygen Species/metabolism , X-ray Repair Cross Complementing Protein 1
4.
Am J Physiol Gastrointest Liver Physiol ; 312(6): G615-G622, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28336546

ABSTRACT

Gastroesophageal reflux disease (GERD) clinically predisposes to columnar Barrett's metaplasia (BM) in the distal esophagus. We demonstrate evidence supporting the cellular origin of BM from reprograming or transcommitment of resident normal esophageal squamous (NES) epithelial cells in response to acid and bile (A + B) exposure using an in vitro cell culture model. The hTERT-immortalized NES cell line NES-B10T was exposed 5 min/day to an A + B mixture for 30 wk. Morphological changes, mRNA, and protein expression levels for the inflammatory marker cyclooxygenase-2; the lineage-determining transcription factors TAp63 (squamous), CDX2, and SOX9 (both columnar); and the columnar lineage markers Villin, Muc-2, CK8, and mAb Das-1 (incomplete phenotype of intestinal metaplasia) were assessed every 10 wk. Markers of columnar lineage and inflammation increased progressively, while squamous lineage-determining transcriptional factors were significantly decreased both at the mRNA and/or protein level in the NES-B10T cells at/after A + B treatment for 30 wk. Distinct modifications in morphological features were only observed at/after 30 wk of A + B exposure. These changes acquired by the NES-B10T 30-wk cells were retained even after cessation of A + B exposure for at least 3 wk. This study provides evidence that chronic exposure to the physiological components of gastric refluxate leads to repression of the discernable squamous transcriptional factors and activation of latent columnar transcriptional factors. This reflects the alteration in lineage commitment of the precursor-like biphenotypic, NES-B10T cells in response to A + B exposure as the possible origin of BM from the resident NES cells.NEW & NOTEWORTHY This study provides evidence of the origins of Barrett's metaplasia from lineage transcommitment of resident esophageal cells after chronic exposure to gastroesophageal refluxate. The preterminal progenitor-like squamous cells alter their differentiation and develop biphenotypic characteristics, expressing markers of incomplete-type columnar metaplasia. Development of these biphenotypic precursors in vitro is a unique model to study pathogenesis of Barrett's metaplasia and esophageal adenocarcinoma.


Subject(s)
Barrett Esophagus/etiology , Cellular Reprogramming , Epithelial Cells/pathology , Esophageal Mucosa/pathology , Gastroesophageal Reflux/complications , Barrett Esophagus/metabolism , Barrett Esophagus/pathology , Cell Line, Transformed , Cell Lineage , Cell Shape , Cellular Reprogramming/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Esophageal Mucosa/drug effects , Esophageal Mucosa/metabolism , Gene Expression Regulation , Glycochenodeoxycholic Acid/toxicity , Humans , Hydrochloric Acid/toxicity , Metaplasia , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Telomerase/genetics , Telomerase/metabolism , Time Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Transfection
5.
PLoS One ; 11(8): e0160525, 2016.
Article in English | MEDLINE | ID: mdl-27490694

ABSTRACT

The harmful effects of bile acid accumulation occurring during cholestatic liver diseases have been associated with oxidative stress increase and endothelial nitric oxide synthase (NOS-3) expression decrease in liver cells. We have previously reported that glycochenodeoxycholic acid (GCDCA) down-regulates gene expression by increasing SP1 binding to the NOS-3 promoter in an oxidative stress dependent manner. In the present study, we aimed to investigate the role of transcription factor (TF) AP-1 on the NOS-3 deregulation during GCDCA-induced cholestasis. The cytotoxic response to GCDCA was characterized by 1) the increased expression and activation of TFs cJun and c-Fos; 2) a higher binding capability of these at position -666 of the NOS-3 promoter; 3) a decrease of the transcriptional activity of the promoter and the expression and activity of NOS-3; and 4) the expression increase of cyclin D1. Specific inhibition of AP-1 by the retinoid SR 11302 counteracted the cytotoxic effects induced by GCDCA while promoting NOS-3 expression recovery and cyclin D1 reduction. NOS activity inhibition by L-NAME inhibited the protective effect of SR 11302. Inducible NOS isoform was no detected in this experimental model of cholestasis. Our data provide direct evidence for the involvement of AP-1 in the NOS-3 expression regulation during cholestasis and define a critical role for NOS-3 in regulating the expression of cyclin D1 during the cell damage induced by bile acids. AP-1 appears as a potential therapeutic target in cholestatic liver diseases given its role as a transcriptional repressor of NOS-3.


Subject(s)
Apoptosis/drug effects , Down-Regulation/drug effects , Glycochenodeoxycholic Acid/toxicity , Nitric Oxide Synthase Type III/metabolism , Retinoids/pharmacology , Transcription Factor AP-1/metabolism , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cyclin D1/antagonists & inhibitors , Cyclin D1/metabolism , Genes, Reporter , Hep G2 Cells , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase Type III/antagonists & inhibitors , Oxidative Stress/drug effects , Promoter Regions, Genetic , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Retinoids/chemistry , Retinoids/metabolism , Transcription Factor AP-1/antagonists & inhibitors , Up-Regulation/drug effects
6.
Cell Physiol Biochem ; 38(4): 1500-9, 2016.
Article in English | MEDLINE | ID: mdl-27050423

ABSTRACT

BACKGROUND/AIMS: In nucleated cells, bile acids may activate cation channels subsequently leading to entry of Ca2+. In erythrocytes, increase of cytosolic Ca2+ activity triggers eryptosis, the suicidal death of erythrocytes characterized by phosphatidylserine exposure at the cell surface and cell shrinkage. Eryptosis is triggered by bile duct ligation, an effect partially attributed to conjugated bilirubin. The present study explored, whether bile acids may stimulate eryptosis. METHODS: Phosphatidylserine exposing erythrocytes have been identified utilizing annexin V binding, cell volume estimated from forward scatter, cytosolic Ca2+ activity determined using Fluo-3 fluorescence, and ceramide abundance at the erythrocyte surface utilizing specific antibodies. RESULTS: The exposure of human erythrocytes to glycochenodesoxycholic (GCDC) and taurochenodesoxycholic (TCDC) acid was followed by a significant decrease of forward scatter and significant increase of Fluo-3 fluorescence, ceramide abundance as well as annexin V binding. The effect on annexin V binding was significantly blunted, but not abolished by removal of extracellular Ca2+. CONCLUSION: Bile acids stimulate suicidal cell death, an effect paralleled by and in part due to Ca2+ entry and ceramide. The bile acid induced eryptosis may in turn lead to accelerated clearance of circulating erythrocytes and, thus, may contribute to anemia in cholestatic patients.


Subject(s)
Bile Acids and Salts/toxicity , Eryptosis/drug effects , Aniline Compounds/chemistry , Aniline Compounds/metabolism , Calcium/metabolism , Cells, Cultured , Ceramides/metabolism , Cholagogues and Choleretics/pharmacology , Detergents/pharmacology , Erythrocytes/cytology , Erythrocytes/drug effects , Erythrocytes/metabolism , Flow Cytometry , Glycochenodeoxycholic Acid/toxicity , Hemolysis/drug effects , Humans , Phosphatidylserines/metabolism , Taurochenodeoxycholic Acid/toxicity , Xanthenes/chemistry , Xanthenes/metabolism
7.
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
8.
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
9.
Free Radic Res ; 49(10): 1275-84, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26118716

ABSTRACT

Mitochondrial oxidative damage is hypothesized to contribute to the pathogenesis of chronic cholestatic liver diseases. Melatonin, an indolamine synthesized in the pineal gland, shows a wide range of physiological functions, and is under clinical investigation for expanded applications. Melatonin has demonstrated efficient protective effects against various types of oxidative damage in the liver system. This study investigates the protective effects of melatonin pretreatment on glycochenodeoxycholic acid (GCDCA)-induced hepatotoxicity and elucidates the potential mechanism of melatonin-mediated protection. Melatonin markedly decreased mitochondrial ROS (mROS) production in L02 cells treated with 100 µM GCDCA, and inhibited GCDCA-stimulated cytotoxicity. Notably, melatonin exerted its hepatoprotective effects by upregulating sirtuin 3 (SIRT3) activity and its expression level, thus regulating superoxide dismutase 2 (SOD2) acetylation and inhibiting the production of mROS induced by GCDCA. Moreover, siRNA targeting SIRT3 blocked the melatonin-mediated elevation in mitochondrial function by inhibiting SIRT3/SOD2 signaling. Importantly, melatonin-activated SIRT3 activity was completely abolished by AMP-activated, alpha 1 catalytic subunit (AMPK) siRNA transfection. Similar results were obtained in rat with bile duct ligation or BDL. In summary, our findings indicate that melatonin is a novel hepatoprotective small molecule that functions by elevating SIRT3, stimulating SOD2 activity, and suppressing mitochondrial oxidative stress at least through AMPK, and that SIRT3 may be of therapeutic value in liver cell protection for GCDCA-induced hepatotoxicity.


Subject(s)
AMP-Activated Protein Kinases/physiology , Glycochenodeoxycholic Acid/toxicity , Hepatocytes/drug effects , Melatonin/pharmacology , Mitochondria, Liver/drug effects , Oxidative Stress/drug effects , Signal Transduction/drug effects , Sirtuin 3/physiology , Superoxide Dismutase/physiology , Acetylation , Adenosine Triphosphate/metabolism , Animals , Hepatocytes/metabolism , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria, Liver/metabolism , Protein Processing, Post-Translational , RNA Interference , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Random Allocation , Rats , Sirtuin 3/biosynthesis , Sirtuin 3/genetics
10.
Cell Physiol Biochem ; 36(3): 866-83, 2015.
Article in English | MEDLINE | ID: mdl-26044599

ABSTRACT

BACKGROUND/AIMS: Ursodeoxycholic acid, which in vivo is rapidly converted into its taurine conjugate, is frequently used for the treatment of cholestatic liver disease. Apart from its choleretic effects, tauroursodeoxycholate (TUDC) can protect hepatocytes from bile acid-induced apoptosis, but the mechanisms underlying its anti-apoptotic effects are poorly understood. METHODS: These mechanisms were investigated in perfused rat liver and isolated rat hepatocytes. RESULTS: It was found that TUDC inhibited the glycochenodeoxycholate (GCDC)-induced activation of the CD95 death receptor at the level of association between CD95 and the epidermal growth factor receptor. This was due to a rapid TUDC-induced ß1-integrin-dependent cyclic AMP (cAMP) signal with induction of the dual specificity mitogen-activated protein (MAP) kinase phosphatase 1 (MKP-1), which prevented GCDC-induced phosphorylation of mitogen-activated protein kinase kinase 4 (MKK4) and c-jun-NH2-terminal kinase (JNK) activation. Furthermore, TUDC induced a protein kinase A (PKA)-mediated serine/threonine phosphorylation of the CD95, which was recently identified as an internalization signal for CD95. Furthermore, TUDC inhibited GCDC-induced CD95 targeting to the plasma membrane in a ß1-integrin-and PKA-dependent manner. In line with this, the ß1-integrin siRNA knockdown in sodium taurocholate cotransporting polypeptide (Ntcp)-transfected HepG2 cells abolished the protective effect of TUDC against GCDC-induced apoptosis. CONCLUSION: TUDC exerts its anti-apoptotic effect via a ß1-integrin-mediated formation of cAMP, which prevents CD95 activation by hydrophobic bile acids at the levels of JNK activation and CD95 serine/threonine phosphorylation.


Subject(s)
Apoptosis/drug effects , Cyclic AMP-Dependent Protein Kinases/genetics , Glycochenodeoxycholic Acid/antagonists & inhibitors , Hepatocytes/drug effects , Integrin beta1/genetics , Taurochenodeoxycholic Acid/pharmacology , Animals , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Dual Specificity Phosphatase 1/genetics , Dual Specificity Phosphatase 1/metabolism , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gene Expression Regulation , Glycochenodeoxycholic Acid/toxicity , Hepatocytes/cytology , Hepatocytes/metabolism , Integrin beta1/metabolism , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Liver/cytology , Liver/drug effects , Liver/metabolism , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/metabolism , Male , Organ Culture Techniques , Organic Anion Transporters, Sodium-Dependent/genetics , Organic Anion Transporters, Sodium-Dependent/metabolism , Phosphorylation , Primary Cell Culture , Pulsatile Flow , Rats , Rats, Wistar , Signal Transduction , Symporters/genetics , Symporters/metabolism , fas Receptor/genetics , fas Receptor/metabolism
11.
Toxicol Appl Pharmacol ; 283(3): 168-77, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25636263

ABSTRACT

Accumulation of bile acids is a major mediator of cholestatic liver injury. Recent studies indicate bile acid composition between humans and rodents is dramatically different, as humans have a higher percent of glycine conjugated bile acids and increased chenodeoxycholate content, which increases the hydrophobicity index of bile acids. This increase may lead to direct toxicity that kills hepatocytes, and promotes inflammation. To address this issue, this study assessed how pathophysiological concentrations of bile acids measured in cholestatic patients affected primary human hepatocytes. Individual bile acid levels were determined in serum and bile by UPLC/QTOFMS in patients with extrahepatic cholestasis with, or without, concurrent increases in serum transaminases. Bile acid levels increased in serum of patients with liver injury, while biliary levels decreased, implicating infarction of the biliary tracts. To assess bile acid-induced toxicity in man, primary human hepatocytes were treated with relevant concentrations, derived from patient data, of the model bile acid glycochenodeoxycholic acid (GCDC). Treatment with GCDC resulted in necrosis with no increase in apoptotic parameters. This was recapitulated by treatment with biliary bile acid concentrations, but not serum concentrations. Marked elevations in serum full-length cytokeratin-18, high mobility group box 1 protein (HMGB1), and acetylated HMGB1 confirmed inflammatory necrosis in injured patients; only modest elevations in caspase-cleaved cytokeratin-18 were observed. These data suggest human hepatocytes are more resistant to human-relevant bile acids than rodent hepatocytes, and die through necrosis when exposed to bile acids. These mechanisms of cholestasis in humans are fundamentally different to mechanisms observed in rodent models.


Subject(s)
Bile Acids and Salts/toxicity , Cholestasis, Extrahepatic/pathology , Glycochenodeoxycholic Acid/toxicity , Hepatocytes/drug effects , Jaundice, Obstructive/pathology , Acetylation , Animals , Bile Acids and Salts/blood , Biomarkers/blood , Cells, Cultured , Cholestasis, Extrahepatic/blood , Dose-Response Relationship, Drug , HMGB1 Protein/blood , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Jaundice, Obstructive/blood , Keratin-18/blood , Mice, Inbred C57BL , Necrosis , Primary Cell Culture , Species Specificity
12.
Biochim Biophys Acta ; 1828(9): 2121-33, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23685124

ABSTRACT

The cell-toxic bile salt glycochenodeoxycholic acid (GCDCA) and taurochenodeoxycholic acid (TCDCA) are responsible for hepatocyte demise in cholestatic liver diseases, while tauroursodeoxycholic acid (TUDCA) is regarded hepatoprotective. We demonstrate the direct mitochondrio-toxicity of bile salts which deplete the mitochondrial membrane potential and induce the mitochondrial permeability transition (MPT). The bile salt mediated mechanistic mode of destruction significantly differs from that of calcium, the prototype MPT inducer. Cell-toxic bile salts initially bind to the mitochondrial outer membrane. Subsequently, the structure of the inner boundary membrane disintegrates. And it is only thereafter that the MPT is induced. This progressive destruction occurs in a dose- and time-dependent way. We demonstrate that GCDCA and TCDCA, but not TUDCA, preferentially permeabilize liposomes containing the mitochondrial membrane protein ANT, a process resembling the MPT induction in whole mitochondria. This suggests that ANT is one decisive target for toxic bile salts. To our knowledge this is the first report unraveling the consecutive steps leading to mitochondrial destruction by cell-toxic bile salts.


Subject(s)
Glycochenodeoxycholic Acid/toxicity , Mitochondria, Liver/drug effects , Mitochondrial ADP, ATP Translocases/agonists , Taurochenodeoxycholic Acid/pharmacology , Animals , Cell Membrane Permeability/drug effects , Dose-Response Relationship, Drug , Liposomes/chemistry , Liver/chemistry , Membrane Potential, Mitochondrial/drug effects , Mitochondria, Heart/chemistry , Mitochondria, Liver/metabolism , Mitochondria, Liver/pathology , Mitochondrial ADP, ATP Translocases/isolation & purification , Mitochondrial Membrane Transport Proteins/agonists , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Membranes/chemistry , Mitochondrial Membranes/drug effects , Mitochondrial Permeability Transition Pore , Myocardium/chemistry , Rats , Taurochenodeoxycholic Acid/toxicity , Voltage-Dependent Anion Channels/chemistry , Voltage-Dependent Anion Channels/isolation & purification
13.
World J Gastroenterol ; 18(36): 4985-93, 2012 Sep 28.
Article in English | MEDLINE | ID: mdl-23049206

ABSTRACT

Cholestasis results in a buildup of bile acids in serum and in hepatocytes. Early studies into the mechanisms of cholestatic liver injury strongly implicated bile acid-induced apoptosis as the major cause of hepatocellular injury. Recent work has focused both on the role of bile acids in cell signaling as well as the role of sterile inflammation in the pathophysiology. Advances in modern analytical methodology have allowed for more accurate measuring of bile acid concentrations in serum, liver, and bile to very low levels of detection. Interestingly, toxic bile acid levels are seemingly far lower than previously hypothesized. The initial hypothesis has been based largely upon the exposure of µmol/L concentrations of toxic bile acids and bile salts to primary hepatocytes in cell culture, the possibility that in vivo bile acid concentrations may be far lower than the observed in vitro toxicity has far reaching implications in the mechanism of injury. This review will focus on both how different bile acids and different bile acid concentrations can affect hepatocytes during cholestasis, and additionally provide insight into how these data support recent hypotheses that cholestatic liver injury may not occur through direct bile acid-induced apoptosis, but may involve largely inflammatory cell-mediated liver cell necrosis.


Subject(s)
Bile Acids and Salts/toxicity , Cholestasis/pathology , Hepatocytes/pathology , Animals , Apoptosis , Bile Acids and Salts/analysis , Glycochenodeoxycholic Acid/toxicity , Humans , Ligation , Necrosis , Signal Transduction
14.
J Hepatobiliary Pancreat Sci ; 18(5): 740-50, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21526375

ABSTRACT

BACKGROUND/PURPOSE: Rifampicin has been used for the treatment of patients with jaundice and pruritus. This study evaluated the effect of rifampicin on the expression of different detoxification systems and bile acid transporters during in-vivo and in-vitro experimental models of cholestasis. METHODS: Rifampicin was administered to glycochenodeoxycholic acid (GCDCA)-treated human hepatocytes and bile duct-obstructed rats. Different parameters related to cell death, and the expression of phase I and II drug metabolizing enzymes (DME) and bile acid transporters were determined. RESULTS: The induction of hepatocellular injury induced by cholestasis was associated with a reduction in cytochrome P4503A4 (CYP3A4), CYP7A1, and UDP-glucuronosyltransferase 2B4 (UGT2B4) expression, as well as an increase in import (Na(+)-taurocholate co-transporting polypeptide, NTCP) system expression. The beneficial properties of rifampicin were associated with an increase in DME and export bile acid systems (multidrug resistance-associated protein 4, MRP4, and bile acid export pump to bile duct, BSEP) expression, as well as a reduction in NTCP expression. CONCLUSIONS: The beneficial effect of rifampicin in cholestasis is associated with an increase in DME expression involved in toxic, bile acid and cholesterol metabolism, as well as a reduction in the bile acid importing system in hepatocytes.


Subject(s)
Carrier Proteins/genetics , Cholestasis/drug therapy , Gene Expression Regulation , Membrane Glycoproteins/genetics , RNA, Messenger/genetics , Rifampin/pharmacology , Animals , Apoptosis , Bile Acids and Salts/metabolism , Carrier Proteins/biosynthesis , Carrier Proteins/drug effects , Cells, Cultured , Chemical and Drug Induced Liver Injury/drug therapy , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Cholestasis/genetics , Cholestasis/metabolism , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Female , Glycochenodeoxycholic Acid/toxicity , Hepatectomy , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Male , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/drug effects , Middle Aged , Polymerase Chain Reaction , RNA, Messenger/metabolism , Rats , Rats, Wistar
15.
J Hepatol ; 55(1): 133-44, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21145864

ABSTRACT

BACKGROUND & AIMS: Reactive oxygen species (ROS) and nitric oxide (NO) exert a relevant role during bile acid-induced hepatotoxicity. Whether α-Tocopherol regulates oxidative and nitrosative stress, bile acid transporter expression and their NO-dependent post-translational modifications, and cell death were assessed in vitro and in vivo. METHODS: α-Tocopherol and/or NO donors (DETA-NONOate or CSNO, and V-PYRRO/NO) were administered to glycochenodeoxycholic acid (GCDCA)-treated cultured human hepatocytes or to bile duct obstructed rats. Cell injury, superoxide anion (O⁻2) production, as well as inducible nitric oxide synthase (NOS-2), cytochrome P4507A1 (CYP7A1), heme oxygenase-1, (HO-1) and bile acid transporter expression were determined. Cysteine S-nitrosylation and tyrosine nitration of Na(+)-taurocholate co-transporting polypeptide (NTCP), as well as taurocholic acid (TC) uptake were also evaluated. RESULTS: GCDCA-induced cell death was associated with increased (O⁻2) production, NTCP and HO-1 expression, and with a reduction of CYP7A1 and NOS-2 expression. α-Tocopherol reduced cell death, (O⁻2) production, CYP7A1, NTCP, and HO-1 expression, as well as increased NOS-2 expression and NO production in GCDCA-treated hepatocytes. α-Tocopherol and NO donors increased NTCP cysteine S-nitrosylation and tyrosine nitration, and reduced TC uptake in hepatocytes. α-Tocopherol and V-PYRRO/NO reduced liver injury and NTCP expression in obstructed rats. CONCLUSIONS: The regulation of CYP7A1, NTCP, and HO-1 expression may be relevant for the cytoprotective properties of α-Tocopherol and NO against mitochondrial dysfunction, oxidative stress and cell death in GCDCA-treated hepatocytes. The regulation of NO-dependent post-translational modifications of NTCP by α-Tocopherol and NO donors reduces the uptake of toxic bile acids by hepatocytes.


Subject(s)
Hepatocytes/drug effects , Hepatocytes/metabolism , Nitric Oxide/metabolism , alpha-Tocopherol/pharmacology , Adult , Aged , Animals , Cell Death/drug effects , Cells, Cultured , Cholestasis/drug therapy , Cholestasis/metabolism , Cholestasis/pathology , Cholesterol 7-alpha-Hydroxylase/metabolism , Cytoprotection/drug effects , Disease Models, Animal , Female , Glycochenodeoxycholic Acid/toxicity , Heme Oxygenase-1/metabolism , Hepatocytes/cytology , Humans , In Vitro Techniques , Male , Membrane Potential, Mitochondrial/drug effects , Middle Aged , Nitric Oxide Donors/pharmacology , Organic Anion Transporters, Sodium-Dependent/genetics , Organic Anion Transporters, Sodium-Dependent/metabolism , Oxidative Stress/drug effects , Protein Processing, Post-Translational/drug effects , Rats , Symporters/genetics , Symporters/metabolism , Transcription, Genetic/drug effects
16.
Biochim Biophys Acta ; 1803(12): 1368-75, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20816704

ABSTRACT

UNLABELLED: Hepatocytes in primary culture undergo apoptosis upon exposure to glycochenodeoxycholate (GCDC). The signaling mechanisms of GCDC-induced apoptosis remain unclear. To investigate the role of antiapoptotic genes, we compared apoptotic response in primary hepatocytes following GCDC treatment. The hepatocytes from adult Sprague-Dawley rats were cultured in collagen-coated dishes and treated with GCDC in varying concentrations, or the same concentration at different time intervals. Apoptosis was detected by the terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL) assay, DNA fragmentation assay, and caspase assays. Expression of apoptosis-related genes and proteins was evaluated by RT-PCR, quantitative real-time PCR (qRT-PCR), and Western blotting, respectively. The DNA-binding property of a nuclear protein was assessed by electrophoretic mobility shift assay (EMSA) and chromatin immunoprecipitation (ChIP) assay. An interesting result was that GCDC caused hepatocyte apoptosis to display a biphasic phenomenon at a dosage of 50µM, whereas it was not found at higher dosages such as 200µM. GCDC stimulated the expression of antiapoptotic Survivin, which also presented a biphasic response. The activation of nuclear factor-kappaB (NF-κB) corresponded with the up-regulation of Survivin. The inhibitor of NF-κB, BAY 11-7082, suppressed the expression of Survivin and simultaneously eliminated the biphasic response. The expression of Survivin was transcriptionally mediated by the activation of NF-κB, as shown by EMSA and ChIP assay. CONCLUSIONS: These results demonstrated that a low dosage of GCDC induced the hepatocyte apoptosis to exhibit the biphasic response, which was regulated by the expression of Survivin through NF-κB signaling pathway.


Subject(s)
Apoptosis/drug effects , Glycochenodeoxycholic Acid/toxicity , Microtubule-Associated Proteins/metabolism , NF-kappa B/metabolism , Signal Transduction , Animals , Rats , Rats, Sprague-Dawley , Survivin
17.
J Hepatol ; 53(5): 918-26, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20675006

ABSTRACT

BACKGROUND & AIMS: Glycochenodeoxycholate (GCDC) and taurolithocholate (TLC) are hepatotoxic and cholestatic bile salts, whereas tauroursodeoxycholate (TUDC) is cytoprotective and anticholestatic. Yet they all act, in part, through phosphatidylinositol-3-kinase(PI3K)-dependent mechanisms ("PI3K-paradox"). Hepatocytes express three catalytic PI3K Class I isoforms (p110α/ß/γ), specific functions of which, in liver, are unclear. In other cell types, p110γ is associated with detrimental effects. To shed light on the PI3K enigma, we determined whether hydrophobic and hydrophilic bile salts differentially activate distinct p110 isoforms in hepatocytes, and whether p110γ mediates bile salt-induced hepatocyte cell death. METHODS: Isoform-specific PI3K activity assays were established to determine isoform activation by bile salts in rat hepatocytes. Activation of Akt and JNK was determined by immunoblotting. Following stimulation with hydrophobic bile salts, hepatocellular apoptosis was determined morphologically after Hoechst staining and by analysis of caspase-3/-7 activity or caspase-3 cleavage. Activity or expression of PI3K p110γ was inhibited pharmacologically (AS604850) or by knock-down using specific siRNA. RESULTS: All bile salts tested activated p110ß, while p110α was activated by TUDC and GCDC. Intriguingly, only hydrophobic bile salts activated p110γ. Inhibition of p110γ attenuated GCDC-induced Akt- and JNK-activation, but did not alter TUDC- or cAMP-induced Akt-signaling in rat hepatocytes. Inhibition or knock-down of p110γ markedly attenuated hydrophobic bile salt-induced apoptosis in rat hepatocytes and human hepatoma cell lines but did not alter Fas-, tumor necrosis factor α- and etoposide-induced apoptosis. Depletion of Ca(++) prevented GCDC-induced toxicity in rat hepatocytes but did not affect GCDC-induced Akt- and JNK-activation. CONCLUSIONS: PI3K p110γ is activated by hydrophobic, but not hydrophilic bile salts. Bile salt-induced hepatocyte apoptosis is partly mediated via a PI3K p110γ dependent signaling pathway, potentially involving JNK.


Subject(s)
Apoptosis/drug effects , Bile Acids and Salts/pharmacology , Carcinoma, Hepatocellular/pathology , Class Ib Phosphatidylinositol 3-Kinase/physiology , Hepatocytes/drug effects , Liver Neoplasms/pathology , Animals , Cells, Cultured , Dioxoles/pharmacology , Enzyme Activation/drug effects , Glycochenodeoxycholic Acid/toxicity , Hep G2 Cells , Hepatocytes/pathology , Humans , JNK Mitogen-Activated Protein Kinases/physiology , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Thiazolidinediones/pharmacology
18.
Chem Res Toxicol ; 22(12): 1984-91, 2009 Dec.
Article in English | MEDLINE | ID: mdl-20020783

ABSTRACT

Ca(2+) mobilization, nitric oxide (NO), and oxidative stress have been involved in cell death induced by hydrophobic bile acid in hepatocytes. The aim of the study was the elucidation of the effect of the antioxidant mitochondrial-driven ubiquinone (Mito Q) on the intracellular Ca(2+) concentration, NO production, and cell death in glycochenodeoxycholic acid (GCDCA)-treated HepG2 cells. The role of the regulation of the intracellular Ca(2+) concentration by Ca(2+) chelators (EGTA or BAPTA-AM), agonist of Ca(2+) entrance (A23187) or NO (L-NAME or NO donor), was assessed during Mito Q cytoprotection in GCDCA-treated HepG2 cells. Cell death, NO synthase (NOS)-1, -2, and -3 expression, Ca(2+) mobilization, and NO production were evaluated. GCDCA reduced the intracellular Ca(2+) concentration and NOS-3 expression and enhanced cell death in HepG2. NO donor prevented and L-NAME enhanced GCDCA-induced cell death. The reduction of Ca(2+) entry by EGTA, but not its release from intracellular stores by BAPTA-AM, reduced the expression of NOS-3 and enhanced cell death in control and GCDCA-treated cells. Mito Q prevented the reduction of intracellular Ca(2+) concentration, NOS-3 expression, NO production, and cell death in GCDCA-treated HepG2 cells. The conclusion is that the recovery of Ca(2+)-dependent NOS-3 expression by Mito Q may be considered an additional cytoprotective property of an antioxidant.


Subject(s)
Apoptosis/drug effects , Calcium/metabolism , Glycochenodeoxycholic Acid/chemistry , Hepatocytes/metabolism , Mitochondria/metabolism , Nitric Oxide/metabolism , Ubiquinone/metabolism , Calcimycin/pharmacology , Caspase 3/metabolism , Glycochenodeoxycholic Acid/toxicity , Hep G2 Cells , Humans , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism
19.
Biochem Biophys Res Commun ; 371(3): 441-5, 2008 Jul 04.
Article in English | MEDLINE | ID: mdl-18452708

ABSTRACT

Delivery of free fatty acids to the liver in nonalcoholic fatty liver disease (NAFLD) may render hepatocytes more vulnerable to glycochenodeoxycholic acid (GCDCA)-induced apoptosis. Fat overloading was induced in HepG2-Ntcp cells and primary rat hepatocytes by incubation with palmitic or oleic acid. Apoptosis was quantified by measuring caspase 3/7 activity and transcription of interleukin (IL) 8 and IL-22 by quantitative real-time PCR. Oleic acid (500 microM) alone did not induce apoptosis, while palmitic acid (500 microM) increased apoptosis 5-fold. GCDCA did not induce significant apoptosis at low micromolar concentrations (5-30 microM) in non-steatotic cells. However, at the same concentrations, GCDCA increased apoptosis 3-fold in oleic acid-pretreated HepG2-Ntcp cells and 3.5-fold in primary rat hepatocytes. Pretreatment with oleic acid increased GCDCA-induced gene transcription of the proinflammatory cytokines IL-8 and IL-22 5-fold and 19-fold, respectively. Thus, low levels of cholestasis normally not considered harmful could advance liver injury in patients with NAFLD.


Subject(s)
Apoptosis , Fats/metabolism , Fatty Acids, Nonesterified/metabolism , Glycochenodeoxycholic Acid/toxicity , Hepatocytes/metabolism , Animals , Cell Line, Tumor , Cells, Cultured , Fatty Acids, Nonesterified/pharmacology , Hepatocytes/drug effects , Humans , Oleic Acid/metabolism , Oleic Acid/pharmacology , Palmitic Acid/metabolism , Palmitic Acid/pharmacology , Rats , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...