Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Oncogene ; 34(23): 3011-22, 2015 Jun 04.
Article in English | MEDLINE | ID: mdl-25220416

ABSTRACT

c-Myc transcription factor is a key protein involved in cellular growth, proliferation and metabolism. c-Myc is one of the most frequently activated oncogenes, highlighting the need to identify intracellular molecules that interact directly with c-Myc to suppress its function. Here we show that Hhex is able to interact with the basic region/helix-loop-helix/leucine zipper of c-Myc. Knockdown of Hhex increases proliferation rate in hepatocellular carcinoma cells, whereas Hhex expression cell-autonomously reduces cell proliferation rate in multiple cell lines by increasing G1 phase length through a c-Myc-dependent mechanism. Global transcriptomic analysis shows that Hhex counter-regulates multiple c-Myc targets involved in cell proliferation and metabolism. Concomitantly, Hhex expression leads to reduced cell size, lower levels of cellular RNA, downregulation of metabolism-related genes, decreased sensitivity to methotrexate and severe reduction in the ability to form tumours in nude mouse xenografts, all indicative of decreased c-Myc activity. Our data suggest that Hhex is a novel regulator of c-Myc function that limits c-Myc activity in transformed cells.


Subject(s)
Carcinoma, Hepatocellular/pathology , Homeodomain Proteins/metabolism , Liver Neoplasms/pathology , Proto-Oncogene Proteins c-myc/metabolism , Transcription Factors/metabolism , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cell Proliferation , Cell Size , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Hep G2 Cells , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Mice , Mice, Nude , Molecular Sequence Data , NIH 3T3 Cells , Neoplasm Transplantation , Proto-Oncogene Proteins c-myc/genetics
2.
Mol Pharmacol ; 63(5): 1180-9, 2003 May.
Article in English | MEDLINE | ID: mdl-12695546

ABSTRACT

Cytochrome P450 3A4 (CYP3A4) is involved in the metabolism of more than 50% of currently used therapeutic drugs, yet the mechanisms that control CYP3A4 basal expression in liver are poorly understood. Several putative binding sites for CCAAT/enhancer-binding protein (C/EBP) and hepatic nuclear factor 3 (HNF-3) were found by computer analysis in CYP3A4 promoter. The use of reporter gene assays, electrophoretic mobility shift assays, and site-directed mutagenesis revealed that one proximal and two distal C/EBP alpha binding sites are essential sites for the trans-activation of CYP3A4 promoter. No trans-activation was found in similar reporter gene experiments with a HNF-3 gamma expression vector. The relevance of these findings was further explored in the more complex DNA/chromatin structure within endogenous CYP3A4 gene. Using appropriate adenoviral expression vectors, we found that both hepatic and nonhepatic cells overexpressing C/EBP alpha had increased CYP3A4 mRNA levels, but no effect was observed when HNF-3 gamma was overexpressed. In contrast, overexpression of HNF-3 gamma simultaneously with C/EBP alpha resulted in a greater activation of the CYP3A4 gene. This cooperative effect was hepatic-specific and also occurred in CYP3A5 and CYP3A7 genes. To investigate the mechanism for HNF-3 gamma action, we studied its binding to CYP3A4 promoter and the effect of the deacetylase inhibitor trichostatin A. HNF-3 gamma was able to bind CYP3A4 promoter at a distal position, near the most distal C/EBP alpha binding site. Trichostatin A increased C/EBP alpha effect but abolished HNF-3 gamma cooperative action. These findings revealed that C/EBP alpha and HNF-3 gamma cooperatively regulate CYP3A4 expression in hepatic cells by a mechanism that probably involves chromatin remodeling.


Subject(s)
CCAAT-Enhancer-Binding Protein-alpha/physiology , Cytochrome P-450 Enzyme System/genetics , DNA-Binding Proteins/physiology , Gene Expression Regulation, Enzymologic , Nuclear Proteins/physiology , Transcription Factors , Transcription, Genetic , Adenoviridae/genetics , Cells, Cultured , Cytochrome P-450 CYP3A , Enzyme Inhibitors/pharmacology , Genetic Vectors , Hepatocyte Nuclear Factor 3-gamma , Hepatocytes/drug effects , Hepatocytes/enzymology , Hepatocytes/physiology , Histone Deacetylase Inhibitors , Humans , Promoter Regions, Genetic/physiology , Transcriptional Activation/drug effects , Transfection
3.
Fundam Clin Pharmacol ; 15(1): 47-54, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11468013

ABSTRACT

The pharmacological effects of glucocorticoids are greatly influenced by their pharmacokinetic properties. In the present report, the in vitro biotransformation of the 22R and 22S epimers of the topical steroid budesonide was studied in the S-9 fraction of human liver, bronchus, skin and colonic mucosa. The disappearance of unchanged epimers of budesonide was measured during 90 min of incubation by high performance liquid chromatography. The rate of disappearance was high in human liver while little biotransformation occurred in bronchial tissue and colonic mucosa, and none was detected in the skin. A marked decay of the initial concentration of unchanged budesonide epimers was noticed after 2 h incubation in cultured human hepatocytes, while only a small decrease was observed after 24 h incubation in cultured human airway smooth muscle cells and BEAS-2B cells. The 22R epimer of budesonide suffered greater in vitro biotransformation than the 22S epimer in human hepatic, bronchial and colonic tissues. These findings extend those of other studies, and confirm that the high therapeutic ratio of budesonide is due to negligible local biotransformation combined with high level of liver metabolism for locally absorbed budesonide.


Subject(s)
Budesonide/pharmacokinetics , Liver/metabolism , Skin/metabolism , Administration, Topical , Anti-Inflammatory Agents/pharmacokinetics , Biotransformation , Bronchi/metabolism , Budesonide/analogs & derivatives , Cell Line , Cells, Cultured , Colon/metabolism , Culture Techniques , Glucocorticoids , Hepatocytes/metabolism , Humans , Intestinal Mucosa/metabolism , Stereoisomerism
4.
Altern Lab Anim ; 29(3): 225-31, 2001.
Article in English | MEDLINE | ID: mdl-11387019

ABSTRACT

Hepatotoxins can be classified as intrinsic when they exert their effects on all individuals in a dose-dependent manner, and as idiosyncratic when their effects are the consequence of an abnormal metabolism of the drug by susceptible individuals (metabolic idiosyncrasy) or of an immune-mediated injury to hepatocytes (allergic hepatitis). Some xenobiotics are electrophilic, and others are biotransformed by the liver into highly reactive metabolites that are usually more toxic than the parent compound. This activation process is the key to many hepatotoxic phenomena. Mitochondria are a frequent target of hepatotoxic drugs, and the alteration of their function has immediate effects on the energy balance of cells (depletion of ATP). Lipid peroxidation, oxidative stress, alteration of Ca(2+) homeostasis, and covalent binding to cell macromolecules are the molecular mechanisms that are frequently involved in the toxicity of xenobiotics. Against these potential hazards, cells have their own defence mechanisms (for example, glutathione, DNA repair, suicide inactivation). Ultimately, toxicity is the balance between bioactivation and detoxification, which determines whether a reactive metabolite elicits a toxic effect. The ultimate goal of in vitro experiments is to generate the type of scientific information needed to identify compounds that are potentially toxic to man. For this purpose, both the design of the experiments and the interpretation of the results are critical.]


Subject(s)
Hepatocytes/drug effects , Hepatocytes/metabolism , Toxicity Tests/methods , Xenobiotics/toxicity , Adenosine Triphosphate/analysis , Adenosine Triphosphate/biosynthesis , Animals , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Anti-Inflammatory Agents, Non-Steroidal/toxicity , Biotransformation , Diclofenac/metabolism , Diclofenac/pharmacokinetics , Diclofenac/toxicity , Formazans/chemistry , Glutathione/analysis , Glutathione/biosynthesis , Humans , In Vitro Techniques , Lipid Peroxides/analysis , Lipid Peroxides/biosynthesis , Rats , Tetrazolium Salts/chemistry , Xenobiotics/metabolism , Xenobiotics/pharmacokinetics
5.
Hepatology ; 33(3): 668-75, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11230748

ABSTRACT

Hepatocyte nuclear factor 4 (HNF4) is a member of the nuclear receptor super-family that has shown activating effects on particular cytochrome P450 (CYP) promoters from several species. However, its role in the regulation of human CYPs in the liver is still poorly understood, as no comprehensive studies in human-relevant models have been performed. In the present study, we have investigated whether HNF4 plays a general role in the expression of 7 major CYP genes in primary cultured human hepatocytes. To this end, we developed an adenoviral vector for efficient expression of HNF4 antisense RNA. Transduction of human hepatocytes with the recombinant adenovirus resulted in a time-dependent increase in the antisense transcript, followed by a concomitant decrease in apolipoprotein C III mRNA (a target gene of HNF4). Specificity was confirmed by showing that increasing levels of HNF4 antisense RNA resulted in the reduction of HNF4 protein, whereas retinoic X receptoralpha-(RXRalpha), the closest homologous member of the nuclear receptor super-family, was unaffected. Analysis of CYP gene expression in human hepatocytes transfected with HNF4 antisense RNA revealed singular behaviors: (1) CYP3A4, CYP3A5, and CYP2A6 showed an important, dose-dependent down-regulation on blockage of HNF4 translation; (2) a moderate inhibition of CYP2B6, CYP2C9, and CYP2D6 expression was observed (40%-45% reduction); (3) the levels of CYP2E1 were not affected even in the absence of this transcription factor. In conclusion, using an original strategy (efficient antisense RNA expression vector), our study shows that HNF4 is a general regulator supporting the expression of major drug-metabolizing CYPs in human hepatocytes.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , DNA-Binding Proteins , Hepatocytes/metabolism , Phosphoproteins/physiology , Transcription Factors/physiology , Adenoviridae/genetics , Adult , Aged , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Cells, Cultured , Cytochrome P-450 Enzyme System/genetics , Female , Gene Expression/physiology , Gene Targeting , Hepatocyte Nuclear Factor 4 , Humans , Liver/physiology , Male , Middle Aged , Oligonucleotides, Antisense/genetics , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/genetics , RNA/genetics , RNA, Messenger/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcription Factors/metabolism
6.
Hepatology ; 32(1): 49-55, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10869288

ABSTRACT

Expression of cytochromes P450 (CYP) is markedly reduced during inflammatory processes. In vitro studies with hepatocytes have shown that cytokines generated during these processes down-regulate CYP. However, it is not clear to what extent each individual cytokine contributes to the overall reduced expression of the various CYP isoenzymes in vivo. Interleukin 6 (IL-6), a major player during inflammatory processes, is recognized as the most important cytokine modulating the hepatic expression of acute-phase protein (APP) genes. For this reason, we selected the IL-6(-/-) mouse as a model to investigate the role of IL-6 in the down-regulation of hepatic CYP during experimental inflammation. Our results show that the reduction in messenger RNA (mRNA) levels of CYP1A2, CYP2A5, and CYP3A11 during turpentine-induced inflammation was abrogated in IL-6-deficient mice, confirming that IL-6 is an indispensable player for the down-regulation of hepatic CYP during aseptic inflammation. Moreover, the different CYP isoenzymes showed a variable grade of dependence on IL-6, CYP2A5 being the most sensitive one. In the case of CYP2E1, differences between IL-6(-/-) and wild-type mice were no longer maintained after 24 hours, suggesting a delayed, rather than abrogated, CYP down-regulation in the absence of IL-6. As opposed to that, hepatic CYP repression took place in IL-6-deficient mice during lipopolysaccharide (LPS)-mediated inflammation. This contrasting behavior observed for CYP is surprisingly similar to the one seen for extracellular (serum amyloid A, beta-fibrinogen) and intracellular (metallothionein-1) APPs and points to the fact that, in the model of bacterial inflammation (LPS), the effects of IL-6 on CYP down-regulation are likely to be substituted by other cytokines or mediators.


Subject(s)
Cytochrome P-450 Enzyme System/genetics , Gene Expression Regulation, Enzymologic , Inflammation/enzymology , Interleukin-6/physiology , Liver/enzymology , Acute-Phase Proteins/biosynthesis , Animals , Down-Regulation , Female , Isoenzymes/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/analysis
7.
Biochem Pharmacol ; 58(10): 1579-85, 1999 Nov 15.
Article in English | MEDLINE | ID: mdl-10535748

ABSTRACT

Increased toxicity of cocaine to human hepatocytes is observed when cells are simultaneously incubated with ethanol. Ethanol might exacerbate cocaine hepatocyte toxicity by three different pathways: a) by increasing the oxidative metabolism of cocaine and hence the oxidative damage; b) by the formation of a more toxic metabolite, namely cocaethylene; or c) by decreasing the defence mechanisms of the cell (i.e. GSH). In the present study, experiments were conducted to investigate the feasibility of these hypotheses. In hepatocytes preincubated for 48 hr with ethanol, neither significant changes in cocaine metabolism nor cytotoxicity were found despite differences in hepatocyte p-nitrophenol hydroxylase (largely CYP2E1 activity). Cocaethylene, the transesterification product of cocaine and ethanol, was found to be more toxic than cocaine for human hepatocytes (3x). However, the small amount formed when human hepatocytes were incubated with cocaine and ethanol would hardly explain the increased toxicity observed. On the other hand, the simultaneous presence of cocaine and ethanol caused a sustained decline in the intracellular GSH content that was larger than that observed in cocaine- or ethanol-treated cultures. Parallel to this phenomenon, a significant increase in lipid peroxidation was observed, as compared to cells treated with equimolar amounts of cocaine, ethanol, or cocaethylene. Finally, depletion of hepatocyte GSH with diethylmaleate down to levels similar to those found in ethanol-treated cells made hepatocytes more susceptible to cocaine. Taken together, the results of this research suggest that by decreasing GSH levels, ethanol makes human hepatocytes more sensitive to cocaine-induced oxidative damage.


Subject(s)
Cocaine/toxicity , Ethanol/pharmacology , Glutathione Transferase/metabolism , Liver/drug effects , Aged , Cell Survival/drug effects , Cells, Cultured , Cocaine/analogs & derivatives , Cocaine/metabolism , Cocaine/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Drug Synergism , Female , Humans , Liver/cytology , Liver/enzymology , Male , Middle Aged , Oxidative Stress
8.
Biochem Pharmacol ; 58(5): 787-96, 1999 Sep 01.
Article in English | MEDLINE | ID: mdl-10449188

ABSTRACT

The aim of this study was to re-examine the human hepatic metabolism of diclofenac, with special focus on the generation of minor hydroxylated metabolites implicated in the idiosyncratic hepatotoxicity of the drug. Different experimental approaches were used: human hepatocytes, human microsomes, and engineered cells expressing single human CYP (cytochromes P450). Human hepatocytes formed 3'-hydroxy-, 4'-hydroxy-, 5-hydroxy- 4',5-dihydroxy-, and N,5-dihydroxydiclofenac, as well as several lactams. Formation of 4'- and 5-hydroxydiclofenac by human liver microsomes followed a Michaelis-Menten kinetics (Km 9 +/- 1 microM; Vmax 432 +/- 15 pmol/min/mg and Km 43 +/- 5 microM; and Vmax 15.4 +/- 0.6 pmol/min/mg, respectively). Secondary metabolites were detected after incubation of 5-hydroxydiclofenac with human liver microsomes, yielding 4',5-dihydroxydiclofenac (Km 15 +/- 1 microM; Vmax 96 +/- 3 pmol/min/mg) and small amounts of N,5-dihydroxydiclofenac (non-Michaelis-Menten kinetics). Based on microsome studies and the incubations with human hepatocytes and engineered cells, we estimated that in vivo CYP2C9 would be exclusively responsible for the 4' hydroxylation of diclofenac (>99.5%) as well as 5-hydroxydiclofenac (>97%). CYP2C9 was exclusively responsible for the formation of 3'-hydroxydiclofenac. Multiple regression analysis evidenced that the rate of production of 5-hydroxydiclofenac in human microsomes followed the algorithm: 0.040 x S-mephenytoin 4'-hydroxylation + 0.083 x tolbutamide methylhydroxylation, (multiple correlation coefficient = 0.969). However, the incubation of diclofenac with cell lines expressing different human CYP suggested that 7 isoforms could be involved. Comparison of data obtained with CYP-expressing cells and human hepatocytes suggests that CYP2C8 > CYP2C19 approximately CYP2C18 >> CYP2B6 are the isoforms implicated in the 5-hydroxylation of diclofenac in vivo.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/metabolism , Aryl Hydrocarbon Hydroxylases , Cytochrome P-450 Enzyme System/metabolism , Diclofenac/metabolism , Microsomes, Liver/drug effects , Steroid 16-alpha-Hydroxylase , Steroid Hydroxylases/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Biotransformation , Cell Line , Cytochrome P-450 CYP2C8 , Cytochrome P-450 CYP2C9 , Diclofenac/pharmacology , Humans , In Vitro Techniques , Microsomes, Liver/enzymology , Oxidation-Reduction
9.
J Pharmacol Exp Ther ; 288(1): 65-72, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9862754

ABSTRACT

Diclofenac, a 2-arylacetic acid, nonsteroidal anti-inflammatory drug, has been reported to cause adverse hepatic effects in certain individuals. To discriminate among possible mechanisms of hepatotoxicity, we examined the effects of diclofenac on human and rat hepatocytes and hepatic cell lines (HepG2, FaO), investigated the major biochemical events in the course of diclofenac cytotoxicity (calcium homeostasis, lipid peroxidation, and mitochondrial dysfunction), and investigated whether cytotoxicity could be related to drug metabolism by cytochrome P-450. Acute diclofenac-induced toxicity in hepatocytes was preluded by a decrease in ATP levels, whereas no significant oxidative stress (decrease in glutathione and lipid peroxidation) or increase in intracellular calcium concentration could be observed at early incubation stages. Diclofenac was more cytotoxic to drug metabolizing cells (rat and human primary cultured hepatocytes) than to nonmetabolizing cell lines (HepG2, FaO). Despite the fact that diclofenac itself was effective in impairing ATP synthesis by mitochondria, we found evidence that toxicity was also related to drug metabolism and was reduced by the addition of cytochrome P-450 inhibitors (proadifen and ketoconazole) to culture medium. The in vitro cytotoxicity correlated well with the formation by hepatocytes of 5-hydroxydiclofenac and, in particular, N,5-dihydroxydiclofenac, a minor metabolite first characterized in this article. Hepatic microsomes showed the ability to both oxidize 5-hydroxydiclofenac to N,5-dihydroxydiclofenac and back reduce the latter to 5-hydroxydiclofenac with the consumption of NADPH. The experimental results suggest that the toxic effect of diclofenac on hepatocytes may be caused by drug-induced mitochondrial impairment, together with a futile consumption of NADPH.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/toxicity , Diclofenac/toxicity , Liver/drug effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Cells, Cultured , Diclofenac/metabolism , Liver/cytology , Male , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Oxidation-Reduction/drug effects , Rats , Rats, Sprague-Dawley
10.
Toxicol In Vitro ; 13(4-5): 633-8, 1999.
Article in English | MEDLINE | ID: mdl-20654527

ABSTRACT

Cell lines stably expressing high levels of single isozymes of human CYP2C genes (CYP2C8, CYP2C9, CYP2C18 and CYP2C19) have been successfully generated by transfecting liver epithelial human cells (THLE) with an appropriate expression vector. To this aim, cDNAs encoding for each CYP2C gene were inserted by blunt-ended cloning into the unique insertion site of the singular expression vector pCMVneo. The recombinant pCMV2C8, pCMV2C9, pCMV2C18 and pCMV2C19 vectors were liposome-mediated transfected into THLE cells. The resulting transgenic cells, designated as T5-2C8, T5-2C9, T5-2C18 and T5-2C19, were cloned and the expression of the ectopic gene, mRNA and protein, was investigated by RT-PCR and Western blot analysis. The functionality of each expressed CYP2C was assessed by determining specific catalytic activities in these cells, that is, taxol-6-hydroxylation for CYP2C8; diclofenac-4'-hydroxylation for CYP2C9; S-mephenytoin-4'-hydroxylation for CYP2C18; S-mephenytoin-4'-hydroxylation for CYP2C19. As a result of the combined strategies used here, the transfected cells showed activities four to seven times higher than those of 24-hour cultured hepatocytes.

11.
FEBS Lett ; 431(2): 227-30, 1998 Jul 17.
Article in English | MEDLINE | ID: mdl-9708908

ABSTRACT

Cytochrome P450 (CYP) activity is very low or even absent in human hepatomas, a phenomenon that is accompanied by low levels of some liver transcription factors, notably C/EBP alpha. To investigate a possible link between this transcription factor and hepatic CYP expression, we have stably transfected HepG2 cells with a C/EBP alpha vector containing a Zn-inducible metallothionein promoter. Expression of functional C/EBP alpha up to liver levels concomitantly increased the mRNAs of several members of the CYP2 family (2B6, 2C9 and 2D6), suggesting that this transcription factor may play a relevant role in controlling the hepatic expression of CYP enzymes.


Subject(s)
Aryl Hydrocarbon Hydroxylases , Cytochrome P-450 CYP2D6/biosynthesis , Cytochrome P-450 Enzyme System/biosynthesis , Cytochrome P-450 Enzyme System/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Enzymologic , Nuclear Proteins/metabolism , Oxidoreductases, N-Demethylating/biosynthesis , Steroid 16-alpha-Hydroxylase , Steroid Hydroxylases/biosynthesis , Transcription Factors/metabolism , CCAAT-Enhancer-Binding Proteins , Carcinoma, Hepatocellular , Cells, Cultured , Cytochrome P-450 CYP2B6 , Cytochrome P-450 CYP2C9 , Cytochrome P-450 CYP2D6/genetics , Cytochrome P-450 Enzyme System/genetics , DNA-Binding Proteins/genetics , Enzyme Induction , Humans , Nuclear Proteins/genetics , Oxidoreductases, N-Demethylating/genetics , RNA, Messenger/metabolism , Steroid Hydroxylases/genetics , Transcription Factors/genetics , Transfection , Tumor Cells, Cultured
12.
J Cell Physiol ; 177(4): 553-62, 1998 Dec.
Article in English | MEDLINE | ID: mdl-10092208

ABSTRACT

Hepatocytes entrapped in collagen gel and cultured in serum-free conditions survived longer than cells cultured on plastic (5 days vs. 3 weeks), showed fewer signs of early cell senescence (no increase in c-fos oncoprotein expression), and maintained the expression of differentiated hepatic metabolic functions over a longer period of time. Cells cultured in collagen gels retained their ability to respond to hormones. The insulin-stimulated glycogen synthesis rate remained fairly constant during 18 days in culture (between 5.4 +/- 0.37 and 9 +/- 2.7 nmol glucose/h/microg DNA). Collagen-cultured hepatocytes recovered glycogen stores to levels similar to those found in liver, or in hepatocytes isolated from fed rats. Urea synthesis from ammonia remained stable for more than 2 weeks (average value, 23 +/- 4 nmol urea/h/microg DNA). The rate of albumin synthesis in collagen-entrapped cells was maintained above the day-1 level during 18 days in culture. Cells showed high levels of glutathione (GSH) (1,278 +/- 152 pmol/microg DNA). Biotransformation activities CYP4501A1, CYP4502A2, CYP4502B1, and CYP4503A1 remained fairly stable in collagen-cultured hepatocytes. CYP4502E1 and CYP4502C11 decreased but were still measurable after 18 days. After 4 days in culture, GST activity returned to levels observed in isolated hepatocytes. In contrast with plastic cultures, cells responded to CYP450 inducers (methylcholanthrene for CYP4501A1, CYP4501A2, and glutathione-transferase, and ethanol for CYP4502E1) for more than 2 weeks. CYP4501A1, CYP4501A2, and glutathione-transferase A2 (GST A2) induction was preceded by an increase in specific mRNA, while the effects on CYP4502E1 seemed to be at a posttranslational level. Analysis of the expression of relevant hepatic genes by reverse Northern and semiquantitative reverse transcriptase-polymerase chain reaction (RT-PCR) revealed that culturing hepatocytes in collagen gels results in a sustained higher expression of key liver transcription factor genes DBP, C/EBP-alpha and -beta, and HNF-1 and -4, as well as specific liver enzyme genes (phosphoenol pyryvate carboxykinase, and carbamoylphosphate-synthetase I).


Subject(s)
Cell Culture Techniques/methods , Liver/cytology , Animals , Biotransformation , Cell Differentiation , Cells, Cultured , Collagen , Culture Media, Serum-Free , Cytochrome P-450 Enzyme System/biosynthesis , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/physiology , Enzyme Induction/drug effects , Extracellular Matrix , Gene Expression Regulation , Genes, fos , Insulin/pharmacology , Isoenzymes/biosynthesis , Isoenzymes/genetics , Isoenzymes/physiology , Liver/metabolism , Liver Glycogen/metabolism , Male , Microsomes, Liver/enzymology , Pharmaceutical Preparations/metabolism , Proto-Oncogene Proteins c-fos/biosynthesis , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/biosynthesis , Transcription Factors/genetics , Urea/metabolism
13.
Cell Biol Toxicol ; 13(4-5): 331-8, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9298253

ABSTRACT

In the course of biotransformation reactions catalyzed both by cytochrome P450 and by conjugating enzymes, drug-derived reactive metabolites and active oxygen species can appear that may escape the detoxification process, initiating radical chain reactions (e.g., lipid peroxidation), covalently binding to macromolecules (proteins, DNA), or impairing the energetic balance of cells. This is usually followed by alterations of ion homeostasis that precede irreversible biochemical changes and cell death. There are, however, cellular mechanisms of defense that prevent, or repair, the damage caused by these reactive intermediates. Ultimately it is the balance between bioactivation, detoxification, and defense mechanisms that determines whether a compound will or will not elicit a toxic effect. Cultures of hepatocytes, including those of human origin, can be used to elucidate the mechanisms of drug toxicity. This is illustrated in the study of the mechanism of hepatotoxicity by diclofenac. Much less cytotoxicity is observed in nonmetabolizing hepatomas than in hepatocytes. The observed cell dysfunction parallels the biotransformation of the drug, and particularly the formation of the minor metabolite N,5-dihydroxydiclofenac by hepatocytes. This compound is able to inhibit mitochondrial ATP synthesis in hepatocytes.


Subject(s)
Cells, Cultured , Liver/cytology , Toxicity Tests , Animals , Anti-Inflammatory Agents, Non-Steroidal/toxicity , Diclofenac/toxicity , Humans , Liver/drug effects , Liver/metabolism , Tumor Cells, Cultured
15.
Drug Metab Dispos ; 24(9): 969-75, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8886606

ABSTRACT

Aceclofenac ([2-(2',6'-dichlorophenylamino)phenyl]acetoxyacetic acid) is a novel nonsteroidal antiinflammatory drug, the pharmacokinetics and drug metabolism of which show species differences. After oral administration to the rat, circulating aceclofenac rapidly disappears yielding [2-(2',6'-dichlorophenylamino)phenyl]acetic acid (diclofenac), which is then further oxidized to [2-(2',6'-dichloro-4'-hydroxyphenylamino)phenyl[acetic acid (4'-hydroxydiclofenac) and [2-(2',6'-dichloro-4'-hydroxyphenylamino)phenyl]acetic acid (4'-hydroxydiclofenac) and [2-(2',6'-dichlorophenylamino)-5-hydroxyphenyl]acetic acid (5-hydroxydiclofenac). This is a minor route in humans, wherein aceclofenac is hydroxylated to [2-(2',6'-dichloro-4'-hydroxyphenylamino)phenyl]acetoxyacetic acid (4'-hydroxyaceclofenac), which becomes the major metabolite. In the monkey, the conversion of aceclofenac to diclofenac takes place, but to a much lesser extent than in the rat, and the 4'-hydroxylated metabolites from both compounds are found in monkeys' urine. The mechanistic basis for this species-dependent variations seems to be the different stability of the drug toward liver esterases. In the rat, the most efficient aceclofenac-hydrolyzing activity is found in hepatic microsomes (Vmax = 2113 +/- 177 pmol/min/mg protein and KM = 191 +/- 40 microM) and cytosol (Vmax = 479 +/- 37 pmol/min/mg protein and KM = 75 +/- 22 microM). Consequently, incubation of aceclofenac with cultured rat hepatocytes or in the rat in vivo results in a rapid hydrolysis of the drug, followed by oxidative metabolism of the resulting diclofenac, yielding 4'- and 5-hydroxylated derivatives as the major metabolites. In contrast, the aceclofenac ester bond is much more stable toward human hepatic microsomal (Vmax = 27 +/- 10 pmol/min/mg protein and KM = 792 +/- 498 microM) and cytosolic (Vmax = 87 +/- 5 pmol/min/mg protein and KM 218 +/- 30 microM) esterases, and 4'-hydroxyaceclofenac becomes the major metabolite in cultured human hepatocytes, as well as in human urine. The research presented herein also illustrates the suitability of cultured human hepatocytes for predicting aceclofenac metabolism in humans.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/metabolism , Diclofenac/analogs & derivatives , Liver/enzymology , Animals , Cells, Cultured , Cytosol/enzymology , Diclofenac/metabolism , Diclofenac/pharmacokinetics , Diclofenac/pharmacology , Esterases/metabolism , Female , Half-Life , Humans , In Vitro Techniques , Kinetics , Macaca fascicularis , Male , Microsomes, Liver/enzymology , Rats , Species Specificity
16.
Drug Metab Dispos ; 24(8): 834-41, 1996 Aug.
Article in English | MEDLINE | ID: mdl-8869816

ABSTRACT

Metabolism of the new nonsteroidal antiinflammatory drug aceclofenac ([2-(2',6'-dichlorophenylamino)phenyl]acetoxyacetic acid) was investigated both in the in vitro hepatic human models and in vivo. Aceclofenac is metabolized in human hepatocytes and human microsomes to form [2-(2',6'-dichloro-4'-hydroxy- phenylamino)phenyl]acetoxyacetic acid as the major metabolite, which is then further conjugated. Minor metabolites were [2-(2',6'-dichlorophenylamino)-5-hydroxyphenyl]acetoxyacetic acid and [2-(2',6'-dichlorophenylamino)phenyl]acetic acid, as well as the hydroxylated derivatives [2-(2',6'-dichloro-4'- hydroxyphenylamino)phenyl]acetic acid and [2-(2',6'-dichlorophenylamino)- 5-hydroxyphenyl]acetic acid. After oral administration to human volunteers (100 mg, single dose), aceclofenac reached a Cmax value of 7.6 +/- 1.3 micrograms/ml and a tmax of 2.6 +/- 1.8. The same metabolites as those detected in cell culture or microsome incubations were found in 12-hr urine after an oral administration of 100 mg aceclofenac to human volunteers. Cytochrome 2C9 is the enzyme responsible for the hydroxylation at position 4'. This could be demonstrated by: 1) selective inhibition by sulfaphenazole; 2) correlation between the formation of the hydroxylated metabolite and tolbutamide hydroxylase activity; and 3) formation of this metabolite only when incubated with microsomes obtained from cells expressing human cytochrome 2C9. However, no conclusive information could be obtained concerning the cytochrome catalyzing the hydroxylation at position 5. The comparison between human microsomes and human hepatocytes metabolism on one hand, and human in vivo metabolism on the other, supports human hepatocytes in primary culture as the model that best anticipated the metabolism of the drug in vivo.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/metabolism , Diclofenac/analogs & derivatives , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Cells, Cultured , Chromatography, High Pressure Liquid , Cytochrome P-450 Enzyme System/metabolism , Diclofenac/metabolism , Diclofenac/pharmacokinetics , Humans , Hydroxylation , Isoenzymes/metabolism , Liver/cytology , Liver/metabolism , Male , Microsomes, Liver/enzymology , Microsomes, Liver/metabolism , Reference Values
17.
Toxicol In Vitro ; 9(4): 439-44, 1995 Aug.
Article in English | MEDLINE | ID: mdl-20650110

ABSTRACT

A certain number of case reports of adverse hepatic reactions to diclofenac are known, suggesting that diclofenac-associated hepatitis may be more common than previously recognized. In order to discriminate among possible molecular mechanisms of toxicity, the following were investigated: (a) cytotoxicity of diclofenac on metabolizing (rat hepatocytes) and non-metabolizing hepatic cells (HepG2, FaO); (b) changes in calcium homoeostasis, glutathione (GSH), lipid peroxidation and ATP levels, and (c) diclofenac metabolism in relation to cytotoxicity. The results indicate that toxicity is associated with the oxidative metabolism of the drug, and correlated with the formation of a minor oxidation metabolite. Inhibitors of diclofenac metabolism concomitantly reduced the toxicity of the drug. Hepatocyte injury was preceded by a decrease in ATP levels. No oxidative stress (no changes in GSH, no lipid peroxidation) could be demonstrated at this early stage. Cytotoxicity was prevented when cells were incubated with fructose, suggesting that the inability of mitochondria to produce ATP is the probable cause of diclofenac hepatotoxicity.

18.
Healthc Financ Manage ; 48(1): 78, 80-2, 84, 1994 Jan.
Article in English | MEDLINE | ID: mdl-10145937

ABSTRACT

Additional paperwork, escalating costs, and an outdated accounts payable system at St. Joseph Health System forced management staff to alter the way the accounts payable department operates. This article describes the process the health system used to automate one of its accounts payable departments by using electronic data interchange/electronic funds transfer (EDI/EFT) technology.


Subject(s)
Accounts Payable and Receivable , Computer Communication Networks/economics , Financial Management, Hospital/trends , Management Information Systems , Multi-Institutional Systems/economics , California , Computer Security , Evaluation Studies as Topic
19.
Am J Physiol ; 262(4 Pt 1): E483-7, 1992 Apr.
Article in English | MEDLINE | ID: mdl-1314496

ABSTRACT

Certain parathyroid hormone (PTH) analogues have been shown to selectively impair some but not all physiological actions of PTH. In this study, transaminated rat (r) PTH [TA-rPTH-(1-34)], a PTH analogue that differs from the rPTH-(1-34) fragment in that the NH2-terminal alanine is converted to pyruvate, was infused into mice to determine its properties in vivo and specifically to determine whether stimulation of 25-hydroxyvitamin D-1 alpha-hydroxylase (1 alpha-hydroxylase) activity was more dependent on concomitant renal handling of phosphate or on generation of adenosine 3',5'-cyclic monophosphate (cAMP). High-performance liquid chromatography-purified TA-rPTH-(1-34) was infused into C57BL mice at 10 or 30 pmol/h for 24 h. At 30 pmol/h, TA-rPTH-(1-34) was comparable with rPTH-(1-34) in its hypophosphatemic and phosphaturic effects but was less potent than rPTH-(1-34) in raising serum calcium. TA-rPTH-(1-34) was markedly less effective in stimulating renal 1 alpha-hydroxylase than rPTH-(1-34). Stimulation of urinary cAMP excretion occurred after infusion with TA-rPTH-(1-34), but this effect was significantly less than that seen with rPTH-(1-34). These findings indicate that PTH-induced hypophosphatemia and phosphaturia can be uncoupled from PTH stimulation of 1 alpha-hydroxylase. Furthermore, cAMP-related signal transduction appears to be more significant in regulation of 1 alpha-hydroxylase than mechanisms that mediate PTH-sensitive phosphate transport, independent of cAMP.


Subject(s)
Parathyroid Hormone/pharmacology , Phosphates/urine , Steroid Hydroxylases/metabolism , Animals , Calcium/blood , Calcium/urine , Cholestanetriol 26-Monooxygenase , Cyclic AMP/urine , Mice , Mice, Inbred C57BL , Phosphorus/blood , Phosphorus/urine , Steroid Hydroxylases/chemistry
20.
Endocrinology ; 123(5): 2491-8, 1988 Nov.
Article in English | MEDLINE | ID: mdl-3049051

ABSTRACT

Monoclonal antibodies (MAbs) have been produced which recognize specific epitopes on bovine renal mitochondrial vitamin D3 1 alpha- and 24-hydroxylases. Renal mitochondria cytochrome P-450s were partially purified to 0.5-2 nmol/mg by Emulgen 911 and cholate solubilization, followed by chromatography on a 2-(4,6-dichloro-O-biphenyloxy)ethylamine HBR affinity column. Reduced carbon monoxide difference spectra determined that this preparation contained 0.5-2 nmol P-450/mg protein. This preparation contained both 1 alpha- and 24-hydroxylase activities, and Eadie-Hofstee plots of product formation as a function of substrate concentrations have maximum velocities of 1.4 and 4 pmol product/30 min.mg protein and Km values of 690 and 1300 nM, respectively. Bovine renal hydroxylases were isolated by immunoprecipitation from this partially purified P-450 preparation with a polyclonal antibody specific for rat liver microsomal cytochrome P-450 RLM5. This polyclonal antibody immunoprecipitated both 1 alpha- and 24-hydroxylase activities as well as renal mitochondrial cytochrome P-450, as determined by reduced CO spectra. Bovine renal mitochondrial components were immunoisolated and used to immunize BALB/c mouse spleen cells in vitro. MAbs then produced were screened for 1) immunoisolation of renal mitochondrial hydroxylase activity from a partially purified preparation, 2) immunohistochemical detection of antigen in renal proximal tubule cells, and 3) immunoquantitation of renal hydroxylases in a solid phase sandwich (enzyme-linked immunosorbent assay) and by 4) Western blot analysis. MAbs were isolated with specifically immunoprecipitated 1 alpha-hydroxylase activity, 24-hydroxylase activity, or both. In 10 micron sections of bovine kidney, antibodies detected antigen only in proximal tubule cells on the basal surface, which is rich in mitochondria. No antigen was detected in sections of pancreas or liver. In the solid phase sandwich enzyme-linked immunosorbent assay, MAbs detected 1 alpha and 24-hydroxylases only in renal mitochondria and not in liver microsomes or adrenal gland mitochondria. In a Western blot, MAbs specific for epitopes expressed on both hydroxylases detected a single band(s) at 52,000-53,000 daltons. Apparently it is not possible to discriminate between hydroxylases by sodium dodecyl sulfate-polyacrylamide gel electrophoresis Western blots. By these criteria, MAbs have been generated which are specific to epitopes expressed on bovine renal mitochondrial 1 alpha- and 24-hydroxylases.


Subject(s)
25-Hydroxyvitamin D3 1-alpha-Hydroxylase/immunology , Antibodies, Monoclonal/immunology , Kidney/enzymology , Steroid Hydroxylases/immunology , 25-Hydroxyvitamin D3 1-alpha-Hydroxylase/analysis , Animals , Antibody Specificity , Blotting, Western , Cattle , Cytochrome P-450 Enzyme System/immunology , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Immunosorbent Techniques , Kinetics , Mice , Mice, Inbred BALB C , Mitochondria/enzymology , Steroid Hydroxylases/analysis , Tissue Distribution , Vitamin D3 24-Hydroxylase
SELECTION OF CITATIONS
SEARCH DETAIL
...