Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
J Phys Chem B ; 123(34): 7302-7312, 2019 08 29.
Article in English | MEDLINE | ID: mdl-31298856

ABSTRACT

The liver performs critical physiological functions, including metabolizing and removing substances, such as toxins and drugs, from the bloodstream. Hepatotoxicity itself is intimately linked to abnormal hepatic transport, and hepatotoxicity remains the primary reason drugs in development fail and approved drugs are withdrawn from the market. For this reason, we propose to analyze, across liver compartments, the transport kinetics of fluorescein-a fluorescent marker used as a proxy for drug molecules-using intravital microscopy data. To resolve the transport kinetics quantitatively from fluorescence data, we account for the effect that different liver compartments (with different chemical properties) have on fluorescein's emission rate. To do so, we develop ordinary differential equation transport models from the data where the kinetics is related to the observable fluorescence levels by "measurement parameters" that vary across different liver compartments. On account of the steep non-linearities in the kinetics and stochasticity inherent to the model, we infer kinetic and measurement parameters by generalizing the method of parameter cascades. For this application, the method of parameter cascades ensures fast and precise parameter estimates from noisy time traces.


Subject(s)
Intravital Microscopy , Liver/metabolism , Animals , Biological Transport , Intravital Microscopy/methods , Kinetics , Liver/drug effects , Liver/ultrastructure , Models, Biological , Rats , Renal Insufficiency, Chronic/chemically induced , Renal Insufficiency, Chronic/metabolism , Taurolithocholic Acid/metabolism , Taurolithocholic Acid/pharmacokinetics , Taurolithocholic Acid/toxicity
2.
Pancreatology ; 17(5): 689-697, 2017.
Article in English | MEDLINE | ID: mdl-28648518

ABSTRACT

OBJECTIVES: To evaluate the therapeutic potential of I-BET-762, an inhibitor of the bromodomain and extra-terminal (BET) protein family, in experimental acute pancreatitis (AP). METHODS: AP was induced by retrograde infusion of taurolithocholic acid sulphate into the biliopancreatic duct (TLCS-AP) or 2 intraperitoneal (i.p.) injections of ethanol and palmitoleic acid 1 h apart (FAEE-AP) or 12 hourly i.p. injections of caerulein (CER-AP). In all treatment groups, I-BET-762 (30 mg/kg, i.p.) was administered at the time of disease induction and again 12 h later. AP severity was assessed at 24 h by serum biochemistry, multiple cytokines and histopathology. RESULTS: TLCS-AP, FAEE-AP and CER-AP resulted in characteristic elevations in serum amylase and cytokine levels, increased pancreatic trypsin and myeloperoxidase activity, typical pancreatic histopathological changes and lung injury. Treatment with I-BET-762 significantly reduced biochemical, cytokine and histopathological responses in TLCS-AP and FAEE-AP, but not CER-AP. CONCLUSIONS: These results suggest that in different forms of AP there are significant differences in the epigenetic control of gene transcription contributing to the severity of disease responses. There is therapeutic potential in targeting bromodomains for the treatment of gallstone- and alcohol-related pancreatitis.


Subject(s)
Benzodiazepines/pharmacology , Bile Acids and Salts/toxicity , Ceruletide/toxicity , Nerve Tissue Proteins/antagonists & inhibitors , Pancreatitis/chemically induced , Receptors, Cell Surface/antagonists & inhibitors , Taurolithocholic Acid/analogs & derivatives , Acute Disease , Amylases/blood , Amylases/metabolism , Animals , Cytokines/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Inflammation/prevention & control , Lung/enzymology , Male , Mice , Pancreas/enzymology , Pancreas/pathology , Pancreatitis/therapy , Peroxidase/genetics , Peroxidase/metabolism , Taurolithocholic Acid/toxicity , Trypsin/metabolism
3.
J Physiol ; 594(21): 6147-6164, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27406326

ABSTRACT

KEY POINTS: Acute biliary pancreatitis is a sudden and severe condition initiated by bile reflux into the pancreas. Bile acids are known to induce Ca2+ signals and necrosis in isolated pancreatic acinar cells but the effects of bile acids on stellate cells are unexplored. Here we show that cholate and taurocholate elicit more dramatic Ca2+ signals and necrosis in stellate cells compared to the adjacent acinar cells in pancreatic lobules; whereas taurolithocholic acid 3-sulfate primarily affects acinar cells. Ca2+ signals and necrosis are strongly dependent on extracellular Ca2+ as well as Na+ ; and Na+ -dependent transport plays an important role in the overall bile acid uptake in pancreatic stellate cells. Bile acid-mediated pancreatic damage can be further escalated by bradykinin-induced signals in stellate cells and thus killing of stellate cells by bile acids might have important implications in acute biliary pancreatitis. ABSTRACT: Acute biliary pancreatitis, caused by bile reflux into the pancreas, is a serious condition characterised by premature activation of digestive enzymes within acinar cells, followed by necrosis and inflammation. Bile acids are known to induce pathological Ca2+ signals and necrosis in acinar cells. However, bile acid-elicited signalling events in stellate cells remain unexplored. This is the first study to demonstrate the pathophysiological effects of bile acids on stellate cells in two experimental models: ex vivo (mouse pancreatic lobules) and in vitro (human cells). Sodium cholate and taurocholate induced cytosolic Ca2+ elevations in stellate cells, larger than those elicited simultaneously in the neighbouring acinar cells. In contrast, taurolithocholic acid 3-sulfate (TLC-S), known to induce Ca2+ oscillations in acinar cells, had only minor effects on stellate cells in lobules. The dependence of the Ca2+ signals on extracellular Na+ and the presence of sodium-taurocholate cotransporting polypeptide (NTCP) indicate a Na+ -dependent bile acid uptake mechanism in stellate cells. Bile acid treatment caused necrosis predominantly in stellate cells, which was abolished by removal of extracellular Ca2+ and significantly reduced in the absence of Na+ , showing that bile-dependent cell death was a downstream event of Ca2+ signals. Finally, combined application of TLC-S and the inflammatory mediator bradykinin caused more extensive necrosis in both stellate and acinar cells than TLC-S alone. Our findings shed new light on the mechanism by which bile acids promote pancreatic pathology. This involves not only signalling in acinar cells but also in stellate cells.


Subject(s)
Bile/metabolism , Calcium Signaling , Pancreatic Stellate Cells/metabolism , Pancreatitis, Acute Necrotizing/metabolism , Sodium/metabolism , Acinar Cells/drug effects , Acinar Cells/metabolism , Acinar Cells/pathology , Animals , Bradykinin/pharmacology , Cells, Cultured , Humans , Male , Mice , Mice, Inbred C57BL , Pancreatic Stellate Cells/drug effects , Pancreatic Stellate Cells/pathology , Pancreatitis, Acute Necrotizing/etiology , Taurolithocholic Acid/analogs & derivatives , Taurolithocholic Acid/toxicity
4.
J Huazhong Univ Sci Technolog Med Sci ; 36(2): 221-225, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27072966

ABSTRACT

The expression of microRNA-19b (miR-19b) in acute necrotizing pancreatitis (ANP) and its functional role in acinar cell necrosis of SD rats were investigated. Twelve SD rats were divided into two groups randomly, including control group and ANP group. The rat ANP models were established by intraperitoneal injection of L-arginine (2400 mg/kg body weight), and equal volume of 0.9% NaCl was injected in the control group. MiRNA chip assay was performed to examine the expression of miRNAs in the pancreas in two different groups. Besides, to further explore the role of miR-19b in ANP in vitro, taurolithocholic acid 3-sulfate disodium salt (TLC-S) (200 µmol/L) was administrated to treat the rat pancreatic acinar cell line, AR42J, for establishing the ANP cells model. The quantitative real-time PCR (qRT-PCR) was adopted to measure the miR-19b expression. Moreover, the mimic miRNA, miRNA antisense oligonucleotide (AMO) and control vector were used to transfect AR42J cells, the expression of miR-19b was confirmed by qRT-PCR and the necrotizing rate of AR42J cells was detected with AO/EB method. The expression of miR-19b was significantly higher in ANP group than in control group as displayed by the miRNA chip assay. Furthermore, after inducing necrosis of AR42J cells in vitro, the expression of miR-19b was significantly increased by 2.51±0.14 times in comparison with the control group. As revealed by qRT-PCR assay, the expression of miR-19b was 5.94±0.95 times higher in the mimic miRNA group than in the control vector group, companied with an obviously increased acinar cell necrotizing rate (50.3%±1.5% vs. 39.6%±2.3%, P<0.05). Moreover, the expression of miR-19b in the miRNA AMO group was 0.38±0.15 times lower than in the control vector group, and the cell necrosis rate was much lower accordingly (23.1%±3.3% vs. 39.6%±2.3%, P<0.05). Besides, there was no significant difference between the control vector cells and the cells without treatment (P>0.05). The expression of miR-19b was significantly induced in ANP. In addition, up-regulation of miR-19b could promote the necrosis of pancreatic acinar cells and miR-19b deficiency could decrease the rate of pancreatic acinar cell necrosis.


Subject(s)
Acinar Cells/metabolism , MicroRNAs/genetics , Pancreatitis, Acute Necrotizing/metabolism , Acinar Cells/pathology , Animals , Arginine/toxicity , Cell Line , MicroRNAs/metabolism , Necrosis , Pancreatitis, Acute Necrotizing/etiology , Rats , Rats, Sprague-Dawley , Taurolithocholic Acid/analogs & derivatives , Taurolithocholic Acid/toxicity , Up-Regulation
5.
J Cell Biochem ; 110(5): 1219-25, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20564217

ABSTRACT

Cholestasis, encountered in a variety of clinical disorders, is characterized by intracellular accumulation of toxic bile acids in the liver. Furthermore, oxidative stress plays an important role in the pathogenesis of bile acids. Taurolithocholic acid (TLC) was revealed in previous studies as the most pro-oxidative bile acid. Melatonin, a well-known antioxidant, is a safe and widely used therapeutic agent. Herein, we investigated the hepatoprotective role of melatonin on lipid and protein oxidation induced by TLC alone and in combination with FeCl(3) and ascorbic acid in rat liver homogenates and hepatic membranes. The lipid peroxidation products, malondialdehyde and 4-hydroxyalkenals (MDA + 4-HDA), and carbonyl levels were quantified as indices of oxidative damage to hepatic lipids and proteins, respectively. In the current study, the rise in MDA + 4-HDA levels induced by TLC was inhibited by melatonin in a concentration-dependent manner in both liver homogenates and in hepatic membranes. Melatonin also had protective effects against structural damage to proteins induced by TLC in membranes. These results suggest that the indoleamine melatonin may potentially act as a protective agent in the therapy of those diseases that involve bile acid toxicity.


Subject(s)
Liver/drug effects , Melatonin/pharmacology , Oxidative Stress/drug effects , Taurolithocholic Acid/toxicity , Aldehydes/metabolism , Animals , Antioxidants/pharmacology , Ascorbic Acid/pharmacology , Chlorides/pharmacology , Cholagogues and Choleretics/toxicity , Dose-Response Relationship, Drug , Ferric Compounds/pharmacology , Lipid Peroxidation/drug effects , Liver/metabolism , Malondialdehyde/metabolism , Oxidation-Reduction/drug effects , Protein Carbonylation/drug effects , Rats , Rats, Sprague-Dawley
6.
Nat Protoc ; 5(2): 335-41, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20134432

ABSTRACT

Mechanistic studies of acute pancreatitis require animal models because clinical material is generally not available during the early phases of the disease. Here we describe a protocol to induce biliary pancreatitis by retrogradely infusing bile acids into the pancreatic duct of anesthetized mice. The resulting model replicates events believed to be responsible for the onset of clinical biliary (i.e., gallstone) pancreatitis and creates highly reproducible pancreatitis with a severity that depends on the concentration of infused bile acid. Pancreatitis reaches its maximal level of severity within 24 h of induction, and it resolves over the subsequent week. This protocol enables the investigator to use genetically modified strains of mice, and it requires only relatively simple and easily learned techniques of small animal surgery. With practice and gentle technique, the surgery (from induction of anesthesia to completion of the infusion) can be completed within 25 min per animal.


Subject(s)
Bile Acids and Salts/toxicity , Pancreatic Ducts/pathology , Pancreatitis/chemically induced , Animals , Bile Acids and Salts/administration & dosage , Disease Models, Animal , Infusions, Parenteral , Mice , Pancreatic Ducts/drug effects , Pancreatitis/pathology , Pancreatitis/surgery , Reference Values , Taurocholic Acid/toxicity , Taurolithocholic Acid/analogs & derivatives , Taurolithocholic Acid/toxicity
7.
Biochem Pharmacol ; 69(7): 1113-20, 2005 Apr 01.
Article in English | MEDLINE | ID: mdl-15763547

ABSTRACT

Estradiol-17beta-d-glucuronide (E(2)17G) and taurolithocholate (TLC) induce acute cholestasis-associated with retrieval of the bile salt export pump (Bsep), which parallels with alteration in transport activity. cAMP stimulates the apically directed vesicular trafficking of transporters, partially preventing these alterations. The hepatoprotector, silymarin, which inhibits cAMP-phosphodiesterase, prevents the cholestasis induced in vivo by both estrogens and TLC. We aimed to assess the ability of silibinin (Sil), the silymarin active component, to prevent the retrieval of Bsep induced by TLC and E(2)17G, and the associated alteration in its transport function. The possible involvement of cAMP as a second messenger and the intracellular signalling pathways implicated were also evaluated. Functional studies were performed analysing the proportion of isolated rat hepatocyte couplets (IRHC) accumulating the fluorescent bile salt analogue, cholyl-lysylfluorescein (CLF), into their sealed canalicular vacuoles. Cellular localisation of Bsep was assessed by immunofluorescent staining. Intracellular levels of cAMP were measured by ELISA. Sil (2.5microM) elevated by 40+/-3% intracellular cAMP, and mimicked the ability of dibutyryl-cAMP (10microM) to prevent internalisation of Bsep and the TLC (2.5microM)- and E(2)17G (50microM)-induced impairment in the capacity of IRHC to accumulate CLF apically. Preventive effects of Sil and dibutyryl-cAMP were not abolished by the specific protein kinase A inhibitors, KT5720 and H89. Contrarily, the intracellular Ca(2+) chelator, BAPTA/AM, significantly blocked the protective effect of both compounds. We conclude that Sil prevented TLC- and E(2)17G-induced bile salt secretory failure, at least in part, by avoiding redistribution of Bsep, by a mechanism probably involving cAMP-induced cytosolic Ca(2+) elevations.


Subject(s)
ATP-Binding Cassette Transporters/physiology , Cholestasis/physiopathology , Cyclic AMP/physiology , Estradiol/analogs & derivatives , Hepatocytes/physiology , Silymarin/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 11 , ATP-Binding Cassette Transporters/drug effects , Animals , Bucladesine/pharmacology , Cell Culture Techniques , Cholestasis/prevention & control , Estradiol/toxicity , Hepatocytes/drug effects , Male , Silybum marianum , Rats , Rats, Wistar , Silybin , Taurolithocholic Acid/toxicity
8.
Toxicol Lett ; 116(1-2): 69-77, 2000 Jul 27.
Article in English | MEDLINE | ID: mdl-10906424

ABSTRACT

The magnitude of cholestasis induced by taurolithocholic acid (TLCA) and its relationship with phase I metabolism were analyzed in rats treated with bromobenzene (BZ), a chemical that causes selective necrosis of perivenous (zone 3) hepatocytes. Forty-eight hours after BZ administration (600 mg/Kg bw), a single dose of 20 micromol/Kg bw of TLCA was injected. Bile was collected during 180 min and bile flow and total bile acid excretion rate were determined. Biliary bile acid composition was analyzed by gas-liquid chromatography-mass spectrometry. BZ administration did not affect the development of TLCA-induced cholestasis, but exacerbated the bile acid-induced decrease in bile flow during the period of recovery from cholestasis. Biliary excretion of total bile acids after TLCA injection relative to basal value was not effected by BZ. The analysis of bile acid composition in bile revealed that TLCA was partially converted to hyodeoxycholic and muricholic acids. The cumulative excretion of all exogenous bile acids and their contribution to the composition of the biliary bile acid pool were not substantially affected by zone 3 necrosis, suggesting that synthesis and secretion of hydroxylated derivatives of TLCA were maintained by zone 1 and 2 hepatocytes. The relative content of endogenous bile acids was not affected by BZ during TLCA-induced cholestasis. Thus, it seems unlikely that the exacerbation of the cholestasis in BZ-treated rats is due to different choleretic properties and/or toxicity of the bile acid pool.


Subject(s)
Cholestasis/chemically induced , Liver/cytology , Taurolithocholic Acid/toxicity , Animals , Bile/drug effects , Bile Acids and Salts/analysis , Bromobenzenes/toxicity , Male , Rats , Rats, Sprague-Dawley
9.
Hepatology ; 29(2): 471-6, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9918924

ABSTRACT

The monohydroxy bile acid, taurolithocholate (TLC), causes cholestasis in vivo and in isolated perfused livers. It is also cholestatic in vitro and, in this study using isolated rat hepatocyte couplets, causes a reduction of the accumulation of (fluorescent) bile acid in the canalicular vacuoles (cVA) of this polarized cell preparation. The hepatoprotective bile acid, tauroursodeoxycholate (TUDCA), partially protects against the action of TLC when added at the same time. It also partially reverses the cholestatic effect if added after the cells have been exposed to TLC. A second hepatoprotective compound, S-adenosyl-L-methionine (SAMe) also not only partially protects against the action of TLC when added at the same time, but it too is able to partially reverse the cholestatic effect. Neither hepatoprotective agent is fully effective alone, but their effects are additive. In combination, a full restoration of cVA is observed in moderate cholestasis, but not in severe cholestasis. We discuss briefly some possible mechanisms involved in the additive mode of action of both hepatoprotective compounds. In summary, we show for the first time that SAMe and TUDCA can exert an additive effect in the amelioration of TLC-induced cholestasis in isolated rat hepatocyte couplets. This finding may be of possible clinical relevance.


Subject(s)
Cholestasis/chemically induced , S-Adenosylmethionine/pharmacology , Taurochenodeoxycholic Acid/pharmacology , Taurolithocholic Acid/toxicity , Animals , Bile Acids and Salts/metabolism , Bile Canaliculi/ultrastructure , Cholestasis/prevention & control , Fluorescent Dyes , Male , Rats , Rats, Wistar , Vacuoles/drug effects , Vacuoles/metabolism
10.
J Toxicol Environ Health ; 50(1): 41-52, 1997 Jan.
Article in English | MEDLINE | ID: mdl-9015131

ABSTRACT

Occupational exposure to methyl isobutyl ketone (MiBK) or methyl n-butyl ketone (MnBK) normally occurs by inhalation. The present study reports that exposure to both ketones can potentiate cholestasis experimentally induced by taurolithocholic acid (TLC, 30 mumol/kg) or by a combination of manganese (Mn, 4.5 mg/kg) and bilirubin (BR, 25 mg/kg). Male Sprague-Dawley rats were exposed for 3 d, 4 h/d, to MiBK or MnBK vapors using 0.5, 1, 1.5, or 2 times the minimal effective concentration (MEC). The estimated MiBK or MnBK MEC for potentiating TLC- or Mn-BR-induced cholestasis were 400 and 150 ppm, respectively. Eighteen hours after ketone exposure, rats were injected i.v. with TLC or Mn-BR. Bile flow was measured from 15 to 150 min after the cholestatic regimen. Rats exposed to MiBK or MnBK exhibited an enhanced diminution in bile flow compared to controls that was dose-dependent with the inhaled ketone dose. The dose-effect characteristics of the potentiation phenomenon were established. Results indicate that MiBK or MnBK inhalation potentiated both TLC and Mn-BR cholestasis in a dose-related fashion. Quantitative differences, however, exist between both ketones with respect to their ability to potentiate both models. Comparison between the two isomers was established, and MnBK was found to be more potent than MiBK.


Subject(s)
Air Pollutants, Occupational/toxicity , Cholestasis, Intrahepatic/chemically induced , Methyl n-Butyl Ketone/toxicity , Administration, Inhalation , Air Pollutants, Occupational/administration & dosage , Animals , Bilirubin/toxicity , Cholagogues and Choleretics/toxicity , Dose-Response Relationship, Drug , Drug Synergism , Isomerism , Male , Manganese Poisoning , Methyl n-Butyl Ketone/administration & dosage , Occupational Exposure , Rats , Rats, Sprague-Dawley , Regression Analysis , Taurolithocholic Acid/toxicity
11.
Dakar Med ; 39(1): 99-107, 1994.
Article in French | MEDLINE | ID: mdl-7493531

ABSTRACT

The aim of this study is: 1. to isolate intrahepatic biliary epithelial cells and, 2. to study the toxicity of cholestatic biliary acids on these cells. Cells were isolated from rats with proliferated bile duct-cells, that were induced by a 21 days bile duct ligation. They were characterized by glutamyltranspeptidase and cytokeratins 7 and 19 using histochemical or immuno cytochemical methods. Isolated cells were incubated with different concentrations (250, 500, 1000 and 2000 microM) of cholestatic biliary acids, lithocholate sulfate (LCS) and taurolithocholate sulfate (TLCS. Cells viability is estimated by three methods: Trypan blue, LDH release and MTT test. We obtained purified fractions of non parenchymal liver cells enriched in biliary epithelial cells (> 95%). On these cells, we showed toxicity of LCS and TLCS and determined CI 10 and CI 50 of these acids which were respectively 800 microM and 2 mM for LCS; 1.4 and 2 mM for TLCS. These results indicate that cholestatic biliary acids (LCS and TLCS) are toxic for biliary cells. This cytotoxicity can be probably a possible mechanism of cholestasis.


Subject(s)
Bile Acids and Salts/toxicity , Bile Ducts, Intrahepatic/drug effects , Cholestasis/metabolism , Animals , Bile Ducts, Intrahepatic/cytology , Cell Death/drug effects , Keratins/analysis , Ligation , Lithocholic Acid/toxicity , Rats , Rats, Sprague-Dawley , Taurolithocholic Acid/toxicity , gamma-Glutamyltransferase/analysis
12.
Toxicol Lett ; 63(3): 243-52, 1992 Dec.
Article in English | MEDLINE | ID: mdl-1283232

ABSTRACT

In rats, pretreatment with certain ketones results in enhanced taurolithocholic acid (TLCA)-induced reduction in bile flow, whereas pretreatment with inhibitors of protein synthesis diminishes the effect on bile flow of cholestatic regimens. In the present study, the possible role of cytochrome P-450 in the ketone potentiation phenomenon was investigated. Male rats were pretreated with inducers or inhibitors of hepatic cytochrome P-450 and the impact of these pretreatments on TLCA-induced cholestasis assessed. Phenobarbital, 3-methylcholanthrene, chlordecone or mirex were used as inducers, and SKF 525-A, piperonyl butoxide, or cobaltous chloride as inhibitors of monooxygenase activity. Phenobarbital and 3-methylcholanthrene pretreatment enhanced TLCA-induced reduction of bile flow, while mirex and chlordecone were without effect. The three inhibitors of monooxygenase activity did not diminish TLCA-induced cholestasis. Instead, piperonyl butoxide and cobaltous chloride appeared to enhance the action of TLCA. Consequently, an increase in cytochrome P-450 (or specific isozymes) as a common denominator in the potentiation phenomenon appears unlikely. While hepatic proteins may play an important role in the potentiation of TLCA-induced cholestasis following pretreatment with ketones, the pattern of potentiation after pretreatment of rats with different inducers or inhibitors of cytochrome P-450 does not appear to implicate this family of proteins.


Subject(s)
Cholestasis/chemically induced , Cytochrome P-450 Enzyme System/metabolism , Ketones/toxicity , Liver/drug effects , Taurolithocholic Acid/toxicity , Animals , Bile/drug effects , Bile/physiology , Body Weight/drug effects , Chlordecone/toxicity , Cobalt/toxicity , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/biosynthesis , Drug Synergism , Enzyme Induction , Male , Methylcholanthrene/toxicity , Mirex/toxicity , Organ Size/drug effects , Phenobarbital/toxicity , Piperonyl Butoxide/toxicity , Proadifen/toxicity , Rats , Rats, Sprague-Dawley
13.
J Hepatol ; 10(3): 280-3, 1990 May.
Article in English | MEDLINE | ID: mdl-2365980

ABSTRACT

Ursodeoxycholate has been advocated for the treatment of cholestatic liver diseases. The coinfusion of tauroursodeoxycholate with taurolithocholate in the perfused rat liver completely prevented the decrease of bile flow and the increase of oxygen uptake found with taurolithocholate only. Bile flow and bile salt secretion were increased with the coinfusion of both bile acids as compared with the infusion of tauroursodeoxycholate only (+4.30 microliters/g liver per 30 min) with 16 and 32 mumol/l tauroursodeoxycholate (+1.55 microliters/g liver per 30 min with 80 and 160 mumol/l). Morphological examination revealed a 50% decrease of the number of necrotic cells in the periportal area. Tauroursodeoxycholate did not inhibit the uptake of taurolithocholate, but increased its transcellular passage and biotransformation. Thus, tauroursodeoxycholate prevents taurolithocholate-induced cholestasis and liver cell toxicity probably by an intracellular mechanism.


Subject(s)
Chenodeoxycholic Acid/analogs & derivatives , Cholestasis/prevention & control , Lithocholic Acid/analogs & derivatives , Liver/drug effects , Taurochenodeoxycholic Acid/therapeutic use , Taurolithocholic Acid/adverse effects , Animals , Cholestasis/chemically induced , Liver/pathology , Rats , Taurolithocholic Acid/toxicity
14.
Res Exp Med (Berl) ; 190(2): 121-9, 1990.
Article in English | MEDLINE | ID: mdl-2349395

ABSTRACT

The infusion of taurolithocholate (TLC) in vivo or in the isolated perfused liver of the rat causes cholestasis and cellular necrosis. In order to analyze the protective effect of bile salts differing in number and steric position of their hydroxy groups against TLC-induced cholestasis, isolated rat livers were perfused with taurocholate (TC), taurohyocholate (THC), tauroursocholate (TUC), taurodehydrocholate (TDHC), and tauroursodeoxycholate (TUDC) (16 and 32 mumol/l) with or without TLC (8 and 16 mumol/l). Bile flow, bile salt secretion, and the hydroxylation pattern of the bile salts secreted were analyzed. TLC caused complete cholestasis after 15 min of perfusion. All bile salts studied had a protective effect. THC, TC, and TUDC completely abolished the cholestasis induced by TLC while TUC did so only for the first 10 min. TDHC was protective only as long as it was biotransformed into hydroxyoxo bile salts. Coinfusion of bile salts did not influence uptake of TLC (greater than or equal to 93% of dose). Differences were found regarding the amount of TLC biotransformed (% of uptake): TC 50%; THC 32%; TUDC 36%; TUC 20%. Light microscopy revealed cellular necrosis, and dilated canaliculi were found in livers perfused with TLC only or in combination with TUC or TDHC, while the other bile salts prevented these changes. We conclude that bile salts with low micelle-forming capacity have little protective effect against TLC-induced cholestasis. These bile salts induce less biotransformation of TLC than TC, THC, and TUDC. The protective effect is not dependent on the hydrocholeretic effect of the added bile salt and is not due to an uptake inhibition.


Subject(s)
Bile Acids and Salts/therapeutic use , Cholestasis/prevention & control , Lithocholic Acid/analogs & derivatives , Taurolithocholic Acid/toxicity , Animals , Bile Acids and Salts/pharmacology , Biotransformation , Cholestasis/chemically induced , Cholestasis/pathology , In Vitro Techniques , Male , Rats , Taurolithocholic Acid/pharmacokinetics
15.
Toxicol Appl Pharmacol ; 80(2): 228-34, 1985 Sep 15.
Article in English | MEDLINE | ID: mdl-4024113

ABSTRACT

Haloalkane-induced hepatonecrogenesis can be potentiated by the prior administration of methyl isobutyl ketone (MIBK) and methyl n-butyl ketone (MBK). We investigated the possibility that these ketones could potentiate the cholestasis induced by taurolithocholate (TLC) in rats. Daily ketone pretreatment for 3 or 7 days resulted in an enhancement of the diminution in bile flow observed after TLC challenge. When the ketones were administered without TLC challenge, cholestasis was not observed; in fact, slight increases in bile flow did occur. The data suggest that MIBK may be more effective than MBK as a potentiator. Preliminary experiments with 2,5-hexanedione (HD), a metabolite of MBK and a potent potentiator of haloalkane hepatonecrosis, were included in the study. HD appeared to be a less potent potentiator of TLC-induced cholestasis. Although some ketones can potentiate cholestatic as well as hepatonecrogenic reactions, different mechanisms of action appear to be involved in these two phenomena.


Subject(s)
Bile/metabolism , Cholestasis/chemically induced , Ketones/pharmacology , Lithocholic Acid/analogs & derivatives , Methyl n-Butyl Ketone/pharmacology , Taurolithocholic Acid/toxicity , Animals , Drug Synergism , Hexanones/pharmacology , Injections, Intravenous , Male , Rats , Rats, Inbred Strains
16.
Hepatology ; 4(4): 661-6, 1984.
Article in English | MEDLINE | ID: mdl-6745854

ABSTRACT

To characterize the relative toxicity of different bile salts, isolated hepatocytes were incubated with different concentrations of one bile salt or with identical concentrations of different bile salts and their conjugates. Incubation lasted for 1 hr; samples were taken at intervals and studied for enzyme release, urea synthesis and stimulation by glucagon, and by electron microscopy. While the trihydroxylated bile salt, taurocholate, did not produce alterations at concentrations up to 1,500 microM, the dihydroxylated salts, chenodeoxy- and deoxycholate, caused enzyme release and membrane lysis, and inhibited urea synthesis at concentrations above 500 microM. In contrast, ursodeoxycholate was ineffective at concentrations up to 1,500 microM. Conjugation of these bile salts did not result in significant differences with the exception of deoxycholate conjugates which induced enzyme leakage more rapidly. Studies of lipid membrane vesicles revealed corresponding alterations. The monohydroxylated salt, taurolithocholate, caused cellular damage as indicated by enzyme loss and impairment of hormonal sensitivity of cells at low concentrations (30 to 100 microM). Dihydroxylated salts produced a different time course of membrane leakage, ultrastructural changes and release of volume marker and lipid in liposomes, suggesting a possible different mechanism of damage induced by this bile salt. Both systems can readily be used to study bile salt membrane interactions.


Subject(s)
Bile Acids and Salts/metabolism , Liver/drug effects , Membrane Lipids/metabolism , Animals , Bile Acids and Salts/toxicity , Cell Membrane/drug effects , Cell Membrane/ultrastructure , Cell Separation , Chenodeoxycholic Acid/toxicity , Deoxycholic Acid/toxicity , Female , Hydroxylation , Liposomes/analysis , Liver/cytology , Liver/enzymology , Rats , Rats, Inbred Strains , Taurolithocholic Acid/toxicity , Ursodeoxycholic Acid/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL