Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
Add more filters










Publication year range
1.
Conf Proc IEEE Eng Med Biol Soc ; 2006: 3198-201, 2006.
Article in English | MEDLINE | ID: mdl-17947014

ABSTRACT

Digital lock-in detection provides spectroscopic and imaging instruments a means of measuring physical quantities with improved signal to noise ratios compared to analogue detection schemes. We introduce a digital lock-in detection algorithm for measuring the amplitude and phase of multiple amplitude modulated signals simultaneously by using particular modulation and sampling constraints and averaging filters. The technique exhibits exceptional reduction in both noise and inter-source distortion. It is shown that the digital lock-in technique can be performed as a simple matrix multiplication in order to reduce computation time. The digital lock-in algorithm is described and analyzed under certain sampling and modulation conditions. Results are shown for experimental data.


Subject(s)
Algorithms , Diagnostic Imaging/statistics & numerical data , Spectrum Analysis/statistics & numerical data , Biomedical Engineering , Diagnostic Imaging/instrumentation , Signal Processing, Computer-Assisted , Spectrum Analysis/instrumentation
2.
J Am Chem Soc ; 123(36): 8780-4, 2001 Sep 12.
Article in English | MEDLINE | ID: mdl-11535083

ABSTRACT

The influence of two different di(1-pyrazolyl)alkane ligands on the rate constant of aqua ligand substitution of ruthenium(II) complexes with the formula [Ru(H2O)(L2)(tpmm)]2+ (L2 = di(1-pyrazolyl)methane (DPMet) or 2,2-di(1-pyrazolyl)propane (DPPro)) was investigated. A 9.4 x 10(5)-fold increase in the rate constant of ligand substitution at pH = 6.86 was observed when DPMet was replaced with DPPro. This remarkable increase was unexpected, considering that these bidentate ligands appear quite similar. To help lend insight into this dramatic spectator ligand effect, the activation parameters for the ligand substitution reactions were determined, and single-crystal X-ray data were collected on the structurally analogous (chloro)ruthenium(II) complexes, [Ru(Cl)(L2)(tpmm)]+. These results are discussed in the context of a heteroscorpionate effect exerted by the DPPro ligand.

3.
J Pharmacol Exp Ther ; 293(2): 677-85, 2000 May.
Article in English | MEDLINE | ID: mdl-10773044

ABSTRACT

The expression of glutathione (GSH)-dependent enzymes and cytochrome P450 (P450) proteins in freshly isolated proximal tubular cells from human kidney (hPT), and the effect of primary culture on these enzymes, were determined. Freshly isolated hPT cells had relatively high activities of gamma-glutamyltransferase, gamma-glutamylcysteine synthetase, glutathione S-transferase (GST), glutathione disulfide reductase, and GSH peroxidase. Cytochrome P450 4A11 was detected in freshly isolated hPT cells, whereas CYP2E1 was not. Freshly isolated hPT cells also expressed GSTA, GSTP, and GSTT but not GSTM. Primary cultures of hPT cells maintained their epithelial-like nature and diploid status, based on measurements of morphology, cytokeratin expression, and flow cytometric analysis. hPT cells retained GSH-dependent enzyme activities during primary culture, whereas cells that had undergone subsequent passage exhibited a loss of activities of most GSH-dependent enzymes and no longer expressed P450s or GSTs. CYP4A11 expression in primary cultures of hPT cells was significantly increased after treatment for 48 h with either ethanol (50 mM) or dexamethasone (7 nM). GSTA, GSTP, and GSTT contents, although still detectable, were decreased compared with those of freshly isolated hPT cells. Our data show that hPT cells express enzymes involved in xenobiotic disposition, and that they thus provide a model suitable for studies of human renal drug metabolism. Furthermore, primary cultures of hPT cells may afford the opportunity to study factors regulating P450 enzyme expression in human kidney.


Subject(s)
Cytochrome P-450 Enzyme System/biosynthesis , Enzymes/metabolism , Glutathione/physiology , Kidney Tubules, Proximal/enzymology , Kidney/enzymology , Adult , Aged , Blotting, Western , Cells, Cultured , Cytochrome P-450 Enzyme Inhibitors , Cytosol/drug effects , Cytosol/enzymology , Female , Flow Cytometry , Humans , Immunohistochemistry , Isoenzymes/antagonists & inhibitors , Isoenzymes/biosynthesis , Keratins/metabolism , Kidney/cytology , Kidney/drug effects , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/drug effects , Male , Microsomes/drug effects , Microsomes/enzymology , Middle Aged , Pharmaceutical Preparations/metabolism , Vimentin/metabolism
4.
Drug Metab Dispos ; 28(3): 354-9, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10681382

ABSTRACT

Tolbutamide is a sulfonylurea-type oral hypoglycemic agent whose action is terminated by hydroxylation of the tolylsulfonyl methyl moiety catalyzed by cytochrome P-450 (CYP) enzymes of the human CYP2C subfamily. Although most studies have implicated CYP2C9 as the exclusive catalyst of hepatic tolbutamide hydroxylation in humans, there is evidence that other CYP2C enzymes (e.g., CYP2C19) may also participate. To that end, we used an immunochemical approach to assess the role of individual CYP2Cs in microsomal tolbutamide metabolism. Polyclonal antibodies were raised to CYP2C9 purified from human liver, and were then back-adsorbed against recombinant CYP2C19 coupled to a solid-phase support. Western blotting revealed that the absorbed anti-human CYP2C9 preparation reacted with only recombinant CYP2C9 and the corresponding native protein in hepatic microsomes, and no longer recognized CYP2C19 and CYP2C8. Monospecific anti-CYP2C9 not only retained the ability to inhibit CYP2C9-catalyzed reactions, as evidenced by its marked (90%) inhibition of diclofenac 4'-hydroxylation by purified CYP2C9 and by human liver microsomes, but also exhibited metabolic specificity, as indicated by its negligible (<15%) inhibitory effect on S-mephenytoin 4'-hydroxylation by purified CYP2C19 or hepatic microsomes containing CYP2C19. Monospecific anti-CYP2C9 was also found to inhibit rates of tolbutamide hydroxylation by 93 +/- 4 and 78 +/- 6% in CYP2C19-deficient and CYP2C19-containing human liver microsomes, respectively. Taken together, our results indicate that both CYP2C9 and CYP2C19 are involved in tolbutamide hydroxylation by human liver microsomes, and that CYP2C19 underlies at least 14 to 22% of tolbutamide metabolism. Although expression of CYP2C19 in human liver is less than that of CYP2C9, it may play an important role in tolbutamide disposition in subjects expressing either high levels of CYP2C19 or a catalytically deficient CYP2C9 enzyme.


Subject(s)
Aryl Hydrocarbon Hydroxylases , Cytochrome P-450 Enzyme System/metabolism , Microsomes, Liver/metabolism , Mixed Function Oxygenases/metabolism , Steroid 16-alpha-Hydroxylase , Tolbutamide/metabolism , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/isolation & purification , Antibodies, Monoclonal/pharmacology , Cross Reactions , Cytochrome P-450 CYP2C19 , Cytochrome P-450 CYP2C8 , Cytochrome P-450 CYP2C9 , Cytochrome P-450 Enzyme System/immunology , Diclofenac/metabolism , Humans , Hydroxylation/drug effects , Male , Mephenytoin/metabolism , Microsomes, Liver/drug effects , Mixed Function Oxygenases/immunology , Rabbits , Steroid Hydroxylases/immunology , Steroid Hydroxylases/metabolism
5.
J Biol Chem ; 275(6): 4118-26, 2000 Feb 11.
Article in English | MEDLINE | ID: mdl-10660572

ABSTRACT

20-hydroxyeicosatetraenoic acid (20-HETE), an omega-hydroxylated arachidonic acid (AA) metabolite, elicits specific effects on kidney vascular and tubular function that, in turn, influence blood pressure control. The human kidney's capacity to convert AA to 20-HETE is unclear, however, as is the underlying P450 catalyst. Microsomes from human kidney cortex were found to convert AA to a single major product, namely 20-HETE, but failed to catalyze AA epoxygenation and midchain hydroxylation. Despite the monophasic nature of renal AA omega-hydroxylation kinetics, immunochemical studies revealed participation of two P450s, CYP4F2 and CYP4A11, since antibodies to these enzymes inhibited 20-HETE formation by 65. 9 +/- 17 and 32.5 +/- 14%, respectively. Western blotting confirmed abundant expression of these CYP4 proteins in human kidney and revealed that other AA-oxidizing P450s, including CYP2C8, CYP2C9, and CYP2E1, were not expressed. Immunocytochemistry showed CYP4F2 and CYP4A11 expression in only the S2 and S3 segments of proximal tubules in cortex and outer medulla. Our results demonstrate that CYP4F2 and CYP4A11 underlie conversion of AA to 20-HETE, a natriuretic and vasoactive eicosanoid, in human kidney. Considering their proximal tubular localization, these P450 enzymes may partake in pivotal renal functions, including the regulation of salt and water balance, and arterial blood pressure itself.


Subject(s)
Hydroxyeicosatetraenoic Acids/biosynthesis , Kidney/metabolism , Vasoconstrictor Agents/pharmacology , Antibodies/pharmacology , Arachidonic Acid/metabolism , Blood Pressure/drug effects , Cytochrome P-450 Enzyme System/immunology , Cytochrome P-450 Enzyme System/metabolism , Fatty Acids, Unsaturated/pharmacology , Humans , Hydroxylation , Immunohistochemistry , Kidney/enzymology , Kidney Tubules/metabolism , Kinetics , Liver/enzymology , Liver/metabolism , Microsomes/enzymology , Microsomes/metabolism
6.
Arch Biochem Biophys ; 359(1): 89-98, 1998 Nov 01.
Article in English | MEDLINE | ID: mdl-9799565

ABSTRACT

Leukotriene B4 (LTB4), an arachidonic acid derivative, is a potent proinflammatory agent whose actions are terminated by catabolism via a microsomal omega-hydroxylation pathway. Although the liver serves as the principal site for LTB4 clearance from the systemic circulation, the attributes of hepatic LTB4 metabolism are ill defined in humans. Thus, we examined metabolism of LTB4 to its omega-hydroxylated metabolite 20-hydroxyleukotriene B4 (20-OH LTB4) by human liver microsomes and also purified the hepatic P450 enzyme underlying this reaction. Liver microsomes from 10 different subjects converted LTB4 to 20-OH LTB4 at similar rates (1.06 +/- 0.3 nmol/min/nmol P450; 0.25 +/- 0.1 nmol/min/mg protein). Analysis of the microsomal LTB4 20-hydroxylation reaction revealed kinetic parameters (apparent Km of 74.8 microM with a VMAX of 2.42 nmol/min/nmol P450) consistent with catalysis by a single P450 enzyme. Conventional chromatography combined with immunochemical screening with rat CYP4A1 antibodies was then used to isolate a P450 enzyme from human liver microsomes with a molecular weight of 57,000 and an NH2-terminal amino acid sequence 94% homologous (12Trp --> 12Gly) over the first 17 residues with the human CYP4F2 cDNA-derived sequence. Upon reconstitution with P450 reductase and phospholipid, CYP4F2 converted LTB4 to 20-OH LTB4 at a turnover rate of 392 pmol/min/nmol P450, whereas the other human liver P450s tested, including CYP4A11, exhibited neglible LTB4 omega-hydroxylase activity. Polyclonal antibodies to CYP4F2 were found to markedly inhibit (91.9 +/- 5%; n = 5) LTB4 20-hydroxylation by human liver microsomes. Microsomal 20-OH LTB4 formation was also inhibited 30% by arachidonic acid, a known CYP4F2 substrate, and 50% by prostaglandin A1 but was unaffected by lauric acid, palmitic acid, and PGF2alpha. Finally, a strong correlation (r = 0.86; P < 0.002; n = 10) was observed between CYP4F2 content and LTB4 20-hydroxylase activity in the human liver samples. Our results indicate that CYP4F2 is the principle LTB4 omega-hydroxylating enzyme expressed in human liver and, as such, may play an important role in regulating circulating as well as hepatic levels of this powerful proinflammatory eicosanoid.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Leukotriene B4/metabolism , Microsomes, Liver/enzymology , Mixed Function Oxygenases/metabolism , Amino Acid Sequence , Cytochrome P-450 Enzyme System/isolation & purification , Cytochrome P-450 Enzyme System/physiology , Cytochrome P450 Family 4 , Humans , Hydroxylation , Immunochemistry , Inflammation/enzymology , Leukotriene B4/analogs & derivatives , Leukotriene B4/analysis , Leukotriene B4/biosynthesis , Leukotriene B4/physiology , Microsomes, Liver/metabolism , Mixed Function Oxygenases/isolation & purification , Mixed Function Oxygenases/physiology , Molecular Sequence Data
7.
Arch Biochem Biophys ; 353(1): 16-28, 1998 May 01.
Article in English | MEDLINE | ID: mdl-9578596

ABSTRACT

Individuals with drug metabolism polymorphisms involving CYP2C enzymes exhibit deficient oxidation of important therapeutic agents, including S-mephenytoin, omeprazole, warfarin, tolbutamide, and nonsteroidal anti-inflammatory drugs. While recombinant CYP2C19 and CYP2C9 proteins expressed in yeast or Escherichia coli have been shown to oxidize these agents, the capacity of the corresponding native P450s isolated from human liver to do so is ill defined. To that end, we purified CYP2C19, CYP2C9, and CYP2C8 from human liver samples using conventional chromatographic techniques and examined their capacity to oxidize S-mephenytoin, omeprazole, and tolbutamide. Upon reconstitution, CYP2C19 metabolized S-mephenytoin and omeprazole at rates that were 11- and 8-fold higher, respectively, than those of intact liver microsomes, whereas neither CYP2C9 nor CYP2C8 displayed appreciable metabolic activity with these substrates. CYP2C19 also proved an efficient catalyst of tolbutamide metabolism, exhibiting a turnover rate similar to CYP2C9 preparations (2.0-6.4 vs 2.4-4.3 nmol hydroxytolbutamide formed/min/nmol P450). The kinetic parameters of CYP2C19-mediated tolbutamide hydroxylation (Km = 650 microM, Vmax = 3.71 min-1) somewhat resembled those of the CYP2C9-catalyzed reaction (Km = 178-407 microM, Vmax = 2.95-7.08 min-1). Polyclonal CYP2C19 antibodies markedly decreased S-mephenytoin 4'-hydroxylation (98% inhibition) and omeprazole 5-hydroxylation (85% inhibition) by human liver microsomes. CYP2C19 antibodies also potently inhibited (>90%) microsomal tolbutamide hydroxylation, which was similar to the inhibition (>85%) observed with antibodies to CYP2C9. Moreover, excellent correlations were found between immunoreactive CYP2C19 content, S-mephenytoin 4'-hydroxylase activity (r = 0.912; P < 0. 001), and omeprazole 5-hydroxylase activity (r = 0.906; P < 0.001) in liver samples from 13-17 different subjects. A significant relationship was likewise observed between microsomal tolbutamide hydroxylation and CYP2C9 content (r = 0.664; P < 0.02) but not with CYP2C19 content (r = 0.393; P = 0.184). Finally, immunoquantitation revealed that in these human liver samples, expression of CYP2C9 (88. 5 +/- 36 nmol/mg) was 5-fold higher than that of CYP2C19 (17.8 +/- 14 nmol/mg) and nearly 8-fold higher than that of CYP2C8 (11.5 +/- 12 nmol/mg). Our results, like those obtained with recombinant CYP2C enzymes, indicate that CYP2C19 is a primary determinant of S-mephenytoin 4'-hydroxylation and low-Km omeprazole 5-hydroxylation in human liver. Despite its tolbutamide hydroxylase activity, the low levels of hepatic CYP2C19 expression (relative to CYP2C9) may preclude an important role for this enzyme in hepatic tolbutamide metabolism and any polymorphisms thereof.


Subject(s)
Aryl Hydrocarbon Hydroxylases , Cytochrome P-450 Enzyme System/metabolism , Mephenytoin/pharmacokinetics , Microsomes, Liver/enzymology , Mixed Function Oxygenases/metabolism , Omeprazole/pharmacokinetics , Steroid 16-alpha-Hydroxylase , Steroid Hydroxylases/metabolism , Tolbutamide/pharmacokinetics , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Chromatography, DEAE-Cellulose , Chromatography, Ion Exchange , Cytochrome P-450 CYP2C19 , Cytochrome P-450 CYP2C8 , Cytochrome P-450 CYP2C9 , Cytochrome P-450 Enzyme System/isolation & purification , Electrophoresis, Polyacrylamide Gel , Humans , Hydroxylation , Kinetics , Mixed Function Oxygenases/isolation & purification , Steroid Hydroxylases/isolation & purification , Substrate Specificity , Ultrafiltration
8.
J Pharmacol Exp Ther ; 285(3): 1327-36, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9618440

ABSTRACT

20-Hydroxy-5,8,11,14-eicosatetraenoic acid (20-HETE) is a principal arachidonic acid (AA) metabolite formed via P450-dependent oxidation in hepatic and renal microsomes. Although 20-HETE plays an important role in the regulation of cell and/or organ physiology, the P450 enzyme(s) catalyzing its formation in humans remain undefined. In this study, we have characterized AA omega-hydroxylation to 20-HETE by human hepatic microsomes and identified the underlying P450s. Analysis of microsomal AA omega-hydroxylation revealed biphasic kinetics (KM1 and VMAX1 = 23 microM and 5.5 min-1; KM2 and VMAX2 = 144 microM and 18.8 min-1) consistent with catalysis by at least two enzymes. Of the human P450s examined, CYP4A11 and CYP4F2 were both potent AA omega-hydroxylases, exhibiting rates of 15.6 and 6.8 nmol 20-HETE formed/min/nmol P450, respectively. Kinetic parameters of 20-HETE formation by CYP4F2 (KM = 24 microM; VMAX = 7.4 min-1) and CYP4A11 (KM = 228 microM; VMAX = 49.1 min-1) resembled the low and high KM components, respectively, found in liver microsomes. Antibodies to CYP4F2 markedly inhibited (93.4 +/- 6%; n = 5) formation of 20-HETE by hepatic microsomes, whereas antibodies to CYP4A11 were much less inhibitory (13.0 +/- 9%; n = 5). Moreover, a strong correlation (r = 0.78; P < .02) was found between microsomal CYP4F2 content and AA omega-hydroxylation among nine subjects. The correlation (r = 0.76; P < .02) also noted between CYP4A11 content and 20-HETE formation stemmed from the relationship (r = 0.83; P < . 02) between hepatic CYP4A11 and CYP4F2 levels in the subjects. Finally, immunoblot analysis revealed that in addition to liver, both P450s also were expressed in human kidney. Our results indicate that AA omega-hydroxylation in human liver is catalyzed by two enzymes of the CYP4 gene family, namely CYP4F2 and CYP4A11, and that CYP4F2 underlies most 20-HETE formation occurring at relevant AA concentrations.


Subject(s)
Arachidonic Acid/metabolism , Cytochrome P-450 Enzyme System/metabolism , Microsomes, Liver/enzymology , Arachidonic Acid/isolation & purification , Cytochrome P-450 CYP4A , Cytochrome P-450 Enzyme System/analysis , Humans , Hydroxyeicosatetraenoic Acids/isolation & purification , Hydroxyeicosatetraenoic Acids/metabolism , Kidney/enzymology , Kidney/metabolism , Microsomes, Liver/metabolism , Mixed Function Oxygenases/analysis , Mixed Function Oxygenases/metabolism
9.
Biochem Pharmacol ; 55(6): 757-66, 1998 Mar 15.
Article in English | MEDLINE | ID: mdl-9586947

ABSTRACT

Our laboratory has shown that human liver microsomes metabolize the anti-HIV drug 3'-azido-3'-deoxythymidine (AZT) via a P450-type reductive reaction to a toxic metabolite 3'-amino-3'-deoxythymidine (AMT). In the present study, we examined the role of specific human P450s and other microsomal enzymes in AZT reduction. Under anaerobic conditions in the presence of NADPH, human liver microsomes converted AZT to AMT with kinetics indicative of two enzymatic components, one with a low Km (58-74 microM) and Vmax (107-142 pmol AMT formed/min/mg protein) and the other with a high Km (4.33-5.88 mM) and Vmax (1804-2607 pmol AMT formed/min/mg). Involvement of a specific P450 enzyme in AZT reduction was not detected by using human P450 substrates and inhibitors. Antibodies to human CYP2E1, CYP3A4, CYP2C8, CYP2C9, CYP2C19, and CYP2A6 were also without effect on this reaction. NADH was as effective as NADPH in promoting microsomal AZT reduction, raising the possibility of cytochrome b5 (b5) involvement. Indeed, AZT reduction among six human liver samples correlated strongly with microsomal b5 content (r2 = 0.96) as well as with aggregate P450 content (r2 = 0.97). Upon reconstitution, human liver b5 plus NADH:b5 reductase and CYP2C9 plus NADPH:P450 reductase were both effective catalysts of AZT reduction, which was also supported when CYP2A6 or CYP2E1 was substituted for CYP2C9. Kinetic analysis revealed an AZT Km of 54 microM and Vmax of 301 pmol/min for b5 plus NADH:b5 reductase and an AZT Km of 103 microM and Vmax of 397 pmol/min for CYP2C9 plus NADPH:P450 reductase. Our results indicate that AZT reduction to AMT by human liver microsomes involves both b5 and P450 enzymes plus their corresponding reductases. The capacity of these proteins and b5 to reduce AZT may be a function of their heme prothestic groups.


Subject(s)
Cytochrome P-450 Enzyme System/physiology , Cytochromes b5/physiology , Dideoxynucleosides/metabolism , Microsomes, Liver/enzymology , Zidovudine/metabolism , Humans , Linear Models , Oxidation-Reduction
10.
Arch Biochem Biophys ; 335(1): 219-26, 1996 Nov 01.
Article in English | MEDLINE | ID: mdl-8914854

ABSTRACT

Human liver microsomes are capable of oxidizing lauric acid (laurate), a model medium-chain fatty acid, at both the omega- and omega-1 positions to form 12- and 11-hydroxylaurate, respectively. These laurate hydroxylation reactions are apparently catalyzed by distinct P450 enzymes. While the P450 responsible for microsomal laurate omega-1 hydroxylation in human liver has been identified as CYP2E1, the enzyme catalyzing omega-hydroxylation remains poorly defined. To that end, we employed conventional purification and immunochemical techniques to characterize the major hepatic laurate omega-hydroxylase in humans. Western blotting with rat CYP4A1 antibodies was used to monitor a cross-reactive P450 protein (M(r) = 52 kDa) during its isolation from human liver microsomes. The purified enzyme (7.4 nmol P450/mg protein) had an NH2-terminal amino acid sequence identical to that predicted from the human CYP4A11 cDNA over the first 20 residues found. Upon reconstitution with P450 reductase and cytochrome b5, CYP4A11 proved to be a potent laurate omega-hydroxylase, exhibiting a turnover rate of 45.7 nmol 12-hydroxylaurate formed/min/nmol P450 (12-fold greater than intact microsomes), while catalyzing the omega-1 hydroxylation reaction at much lower rates (5.4 nmol 11-hydroxylaurate formed/min/nmol P450). Analysis of the laurate omega-hydroxylation reaction in human liver microsomes revealed kinetic parameters (a lone Km of 48.9 microM with a VMAX of 3.72 nmol 12-hydroxylaurate formed/min/nmol P450) consistent with catalysis by CYP4A11. In fact, incubation of human liver microsomes with antibodies raised to CYP4A11 resulted in nearly 85% inhibition of laurate omega-hydroxylase activity while omega-1 hydroxylase activity remained unaffected. Furthermore, a strong correlation (r = 0.89; P < 0.001) was found between immunochemically determined CYP4A11 content and laurate omega-hydroxylase activity in liver samples from 11 different subjects. From the foregoing, it appears that CYP4A11 is the principle laurate omega-hydroxylating enzyme expressed in human liver.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Microsomes, Liver/enzymology , Mixed Function Oxygenases/metabolism , Animals , Antibodies , Blotting, Western , Cross Reactions , Cytochrome P-450 CYP4A , Cytochrome P-450 Enzyme System/isolation & purification , Humans , Lauric Acids/metabolism , Mixed Function Oxygenases/isolation & purification , Rats , Substrate Specificity
11.
Drug Metab Dispos ; 24(10): 1081-7, 1996 Oct.
Article in English | MEDLINE | ID: mdl-8894508

ABSTRACT

Omeprazole (OP) is a potent antiulcer drug that is metabolized by liver cytochrome P450 (P450) enzymes. However, the identities of the P450 isoforms responsible for its metabolism have been controversial. 5-Hydroxyomeprazole (5OH-OP) formation cosegregates with the polymorphism of (S)-mephenytoin 4'-hydroxylation in humans, which is now known to be mediated by CYP2C19. Previous in vitro studies have indicated that liver microsomal 50H-OP formation correlates with both (S)-mephenytoin 4'-hydroxylase and CYP3A content. Inhibitor and CYP2C antibody studies also suggested that both enzymes may be involved in the 5-hydroxylation of OP, whereas CYP3A appears to be the predominant enzyme involved in OP sulfone (OP-S) formation. The present studies assessed the contribution of various CYP2C and CYP3A4 enzymes to OP metabolism by using recombinant human enzymes. CYP2C19, CYP2C8, CYP2C18, and CYP2C9 formed a single metabolite with an HPLC retention time identical to that of 5OH-OP. The turnover number for CYP2C19 was 13.4 +/- 1.4 nmol/min/nmol of P450, whereas those for CYP2C8, CYP2C18, and CYP2C9 were 2.2 +/- 0.1, 1.5 +/- 0.1, and approximately equal to 0.5 nmol/min/nmol of P450, respectively. Recombinant human CYP3A4 formed 5OH-OP and OP-S with turnover numbers of 5.7 +/- 1.1 and 7.4 +/- 0.9 nmol/min/nmol of P450, respectively, and formed a minor unidentified metabolite. CYP2C19 had a substantially lower KM for 5OH-OP formation than did CYP3A4, CYP2C8, or CYP2C18. Antibody to CYP2C proteins inhibited approximately equal to 70% of OP 5-hydroxylation at low substrate concentrations, comparable to those that may be encountered at therapeutically relevant doses, whereas antibody to CYP3A4 inhibited approximately equal to 30% of the activity. At high substrate concentrations, the contributions of the two enzymes to OP hydroxylation were roughly comparable (40-50%). In contrast, OP-S formation was completely inhibited by antibody to CYP3A4 proteins. The present study provides the first direct confirmation, using human recombinant P450 enzymes and selective antibody inhibition, that CYP2C19 is a major high affinity OP 5-hydroxylase and CYP3A4 is a low affinity OP-hydroxylating enzyme. The current work also shows, for the first time, that other CYP2C enzymes (CYP2C8, CYP2C9, and CYP2C18) may contribute to OP hydroxylation at high substrate concentrations. In contrast, OP-S was formed principally by CYP3A4.


Subject(s)
Aryl Hydrocarbon Hydroxylases , Cytochrome P-450 Enzyme System/metabolism , Mixed Function Oxygenases/metabolism , Anti-Ulcer Agents/pharmacokinetics , Carbon Radioisotopes , Chromatography, High Pressure Liquid , Cytochrome P-450 CYP2C19 , Enzyme Inhibitors/pharmacokinetics , Humans , Microsomes, Liver/enzymology , Omeprazole/pharmacokinetics , Recombinant Proteins/metabolism , Substrate Specificity
12.
Drug Metab Dispos ; 24(9): 932-9, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8886601

ABSTRACT

The microsomal metabolism of fentanyl, a synthetic opioid commonly used in anesthesia, was investigated in human liver. Incubation of fentanyl with human hepatic microsomes fortified with NADPH resulted in the formation of a single major metabolite, namely norfentanyl, as determined by GC/MS. No evidence was obtained for the formation of either desproprionylfentanyl or N-phenylpropionamide, the latter arising via N-dealkylation of the fentanyl amide nitrogen. Kinetic analysis of microsomal fentanyl oxidation revealed a single K(m) of 117 microM and a Vmax of 3.86 nmol of norfentanyl formed/min/nmol of cytochrome P450 (P450). Studies using chemical inhibitors of human P450 enzymes revealed that only agents known to inhibit CYP3A4 (e.g. ketoconazole and erythromycin) were capable of strongly inhibiting (> or = 90%) microsomal fentanyl oxidation. Marked inhibition (> 90%) of norfentanyl formation by liver microsomes was also observed with polyclonal antibodies to CYP3A4, whereas antibodies to other human P450s were without effect. Furthermore, rates of norfentanyl production by 10 individual human liver samples were highly correlated (r2 = 0.876, F = 56.46 p < 0.001) with immunochemically determined levels of CYP3A4 present in the samples but not with levels of CYP2C8, CYP2C9, CYP2C19, or CYP2E1. Our results indicate that CYP3A4 is the major catalyst involved in fentanyl oxidation to norfentanyl in human liver. Alterations in CYP3A4 levels or activity, as well as the concomitant administration of other therapeutic agents metabolized by this P450 enzyme, could lead to marked perturbations in fentanyl disposition and, hence, analgesic response.


Subject(s)
Analgesics, Opioid/metabolism , Aryl Hydrocarbon Hydroxylases , Cytochrome P-450 Enzyme System/metabolism , Fentanyl/metabolism , Microsomes, Liver/enzymology , Mixed Function Oxygenases/metabolism , Steroid 16-alpha-Hydroxylase , Cytochrome P-450 CYP1A2/metabolism , Cytochrome P-450 CYP2C8 , Cytochrome P-450 CYP2C9 , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme Inhibitors , Fentanyl/analogs & derivatives , Fentanyl/pharmacology , Gas Chromatography-Mass Spectrometry , Humans , Ketoconazole/pharmacology , Kinetics , Mixed Function Oxygenases/antagonists & inhibitors , Steroid Hydroxylases/metabolism , Theophylline/analogs & derivatives , Theophylline/pharmacology , Tolbutamide/pharmacology
13.
Mol Pharmacol ; 49(5): 814-21, 1996 May.
Article in English | MEDLINE | ID: mdl-8622631

ABSTRACT

Ethanol can be oxidized to the 1-hydroxyethyl radical (HER) by rat and deer mice liver microsomal systems. Experiments were carried out to evaluate the ability of human liver microsomes to catalyze this reaction, compare the effectiveness of NADH with that of NADPH, and assess the possible role of cytochrome b5 in HER formation. HER was detected as the alpha-(4-pyridly-1 -oxide)-N-t-butylnitrone/HER adduct. Human liver microsomes catalyzed HER formation with either NADPH or NADH as cofactor; rates with NADH were approximately 50% those found with NADPH. Chelex-100 treatment of the reaction mixture produced marked inhibition of HER formation, suggesting that a transition metal, such as iron, was required to catalyze the reaction. The addition of ferric chloride restore HER formation. Catalase (2600 units/ml) and superoxide dismutases (500 units/ml) nearly completely inhibited the reaction with either NADPH or NADH. The NADH-dependent rates of superoxide production, detected as 5,5-dimethyl-1-pyrroline-N-oxide-O2H, were approximately 50% the NADPH-dependent rates, which is consistent with the rates of HER formation. Anti-cytochrome b5 IgG decreased NADPH- and NADH-dependent HER formation, and this was associated with inhibition of superoxide formation with both reductants. These results indicate that human liver microsomes can catalyze the oxidation of ethanol of HER with either NADPH or NADH as reductant. The effectiveness of NADH may be significant in view of the increased NADH/NAD+ redox ratio in the liver as a consequence of ethanol oxidation by alcohol dehydrogenase. HER formation by human liver microsomes seems to be catalyzed by an oxidant derived from the interaction of iron with superoxide or H2O2, and a close association exists between HER formation and superoxide production. Cytochrome b5 seems to play a role in HER formation, most likely due to its effect on superoxide production.


Subject(s)
Ethanol/metabolism , Microsomes, Liver/metabolism , Adult , Animals , Cell-Free System , Cytochromes b5/metabolism , Electron Spin Resonance Spectroscopy , Female , Free Radicals , Humans , Male , Middle Aged , NAD/metabolism , NADP/metabolism , Oxidation-Reduction , Rats , Rats, Sprague-Dawley , Superoxide Dismutase/metabolism , Superoxides/metabolism
14.
Mol Pharmacol ; 49(2): 260-8, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8632758

ABSTRACT

The mechanisms responsible for ethanol-mediated teratogenesis have not been resolved. However, possible etiologies include the local formation of the teratogen acetaldehyde or oxygen radicals by fetal ethanol-oxidizing enzymes. As alcohol dehydrogenases are expressed at very low concentrations in human embryonic tissues, the ethanol-inducible P450 enzyme, CYP2E1, could be the sole catalyst of fetal ethanol oxidation. With this in mind, we examined the expression of this P450 in liver samples from fetuses ranging in gestational age from 16 to 24 weeks. Immunoblot analysis of fetal liver microsomes revealed the presence of a protein immunoreactive with CYP2E1 antibodies that exhibited a slightly lower molecular weight than that found in adult liver samples. Embryonic CYP2E1 expression was further confirmed by the reverse transcriptase reaction with RNA from a 19-week gestational fetal liver used as template. Catalytic capabilities of human fetal microsomes were assessed by measurement of the rate of ethanol oxidation to acetaldehyde, which were 12-27% of those exhibited by adult liver microsomes. Immunoinhibition studies with CYP2E1 antibodies revealed that the corresponding antigen was the major catalyst of this reaction in both fetal and adult tissues. We then assessed whether embryonic CYP2E1 was, like the adult enzyme, inducible by xenobiotics. Treatment of primary fetal hepatocyte cultures with either ethanol or clofibrate demonstrated a 2-fold increase in CYP2E1 levels compared with untreated cells. Collectively, our results indicate that CYP2E1 is present in human fetal liver, that the enzyme is functionally similar to CYP2E1 from adults, and that fetal hepatocyte CYP2E1 is inducible in culture by xenobiotics, including ethanol. Because fetal CYP2E1 mediates ethanol metabolism, the enzyme may play a pivotal role in the local production of acetaldehyde and free radicals, both of which have potential deleterious effects on the developing fetus.


Subject(s)
Cytochrome P-450 Enzyme System/biosynthesis , Ethanol/pharmacology , Gene Expression Regulation, Developmental , Liver/embryology , Microsomes, Liver/enzymology , Oxidoreductases, N-Demethylating/biosynthesis , Adult , Cells, Cultured , Cytochrome P-450 CYP2E1 , Cytochrome P-450 Enzyme System/metabolism , Embryonic and Fetal Development , Enzyme Induction , Enzyme Stability , Ethanol/metabolism , Fetus , Gestational Age , Humans , Immunohistochemistry , Kinetics , Liver/drug effects , Liver/enzymology , Oxidoreductases, N-Demethylating/metabolism , Polymerase Chain Reaction , Templates, Genetic
15.
Hepatology ; 22(5): 1462-8, 1995 Nov.
Article in English | MEDLINE | ID: mdl-7590664

ABSTRACT

Carbohydrate-deficient transferrin (CDT) is now considered to be the most sensitive and specific biological marker of alcohol abuse. However, the mechanism by which chronic alcohol consumption causes an elevation of CDT levels in serum is still not understood. Therefore, we fed eight pairs of male rats a nutritionally adequate liquid diet containing either alcohol (36% of energy) or isocaloric dextrose (control) for 4 weeks, after which blood and liver samples were obtained. Serum CDT content in alcohol-treated rats increased by 45% (P < .05) in ethanol-fed animals compared with their corresponding controls. In contrast, in rats fed ethanol, the activities of sialyltransferase (ST), galactosyltransferase (GT), and N-acetylglucosamine transferase (N-AGT), which are glycosyltransferases involved in transferrin carbohydrate side chain synthesis, were diminished by 24% and 40% (P < .05), 23% and 51% (P < .05, .001), and 20% and 26% (P < .05) in total liver homogenates and Golgi fraction (GF) 1, respectively, when expressed as units/100 g body weight. These enzymes were also significantly less active in hepatic GFs 2 and 3. The depression of the transferase activities in ethanol-fed rats appeared to be due, at least in part, to enzyme inactivation by acetaldehyde, whereas ethanol itself was without effect. Similar results were obtained in humans: five alcohol abusers were found to exhibit a 23% decrease in hepatic sialyltransferase and a 41% increase in sialidase activities, respectively, when compared with three nondrinking subjects.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Alcoholism/metabolism , Liver/pathology , Transferrin/chemistry , Alcoholism/pathology , Animals , Carbohydrates/chemistry , Glycosyltransferases/biosynthesis , Humans , Liver/enzymology , Male , Neuraminidase/biosynthesis , Rats , Rats, Sprague-Dawley , Transferrin/analysis
16.
J Pharmacol Exp Ther ; 271(3): 1427-33, 1994 Dec.
Article in English | MEDLINE | ID: mdl-7996455

ABSTRACT

In the present study, the regio- and stereoselective epoxidation of arachidonic acid by cytochromes P450 2C8 and 2C9, two members of the CYP2C gene subfamily expressed in human liver, was determined. Purified P450 isozymes, reconstituted with NADPH:P450 oxidoreductase, cytochrome b5 and lipid, or microsomes isolated from human liver, were incubated with [1-14C]-arachidonic acid. For regioselective analysis, the epoxide metabolites formed, 14,15-, 11,12- and 8,9-epoxyeicosatrienoic acids (EETs), were resolved by reverse-phase high-performance liquid chromatography. P450 2C8 produces only the 14,15- and 11,12-EETs in a 1.25:1.00 ratio. The two epoxides represent 68% of the total metabolites. P450 2C9 produces 14,15-, 11,12- and 8,9-EETs in a 2.3:1.0:0.5 ratio. The three epoxides represent 69% of the total metabolites. Neither P450 isoform catalyzes the formation of 5,6-EET. For chiral analysis, the two major epoxide metabolites, 14,15- and 11,12-EETs, were derivatized to methyl and pentafluorbenzyl esters, respectively. Enantiomers of 14,15- and 11,12-EET esters were subsequently resolved on Chiralcel OB and OD columns (J.T. Baker, Phillipsburg, PA), respectively. Both P450 2C8 and 2C9 are stereoselective at the 14,15- position, preferentially producing 14(R), 15(S)-EET with 86.2% and 62.5% selectivity, respectively. Both enzymes are also stereoselective at the 11,12-position but have the opposite selectivity. P450 2C8 is 81.1% selective for 11(R), 12(S)-EET; P450 2C9 is 69.4% selective for the 11(S), 12(R)-EET. Immunoinhibition studies performed with anti-2C9 immunoglobulin G (which also reacts with P450 2C8) and hepatic microsomes indicate that these two P450s are important arachidonic acid epoxygenases in human liver.


Subject(s)
8,11,14-Eicosatrienoic Acid/analogs & derivatives , Arachidonic Acid/metabolism , Aryl Hydrocarbon Hydroxylases , Cytochrome P-450 Enzyme System/physiology , Steroid 16-alpha-Hydroxylase , Steroid Hydroxylases/physiology , 8,11,14-Eicosatrienoic Acid/metabolism , Amino Acid Sequence , Animals , Humans , In Vitro Techniques , Male , Microsomes, Liver/metabolism , Molecular Sequence Data , Rabbits , Stereoisomerism
17.
Biochemistry ; 33(7): 1743-52, 1994 Feb 22.
Article in English | MEDLINE | ID: mdl-8110777

ABSTRACT

The present study assesses the role of members of the human CYP2C subfamily in the 4'-hydroxylation of (S)-mephenytoin. When recombinant CYP2C proteins were expressed using a yeast cDNA expression system, 2C19 stereospecifically 4'-hydroxylated (S)-mephenytoin with a turnover number at least 10 times higher than that of human liver microsomes. 2C9 (both Ile359 and Leu359 alleles) and 2C18 (Thr385 and Met385 alleles) metabolized this substrate at a rate 100-fold lower than 2C19, and metabolism by these 2C proteins was not stereospecific for the S-enantiomer. 2C8 exhibited very little mephenytoin 4'-hydroxylase activity. In contrast, the Ile359 allele of 2C9 had a high turnover number for the hydroxylation of tolbutamide, while the Leu359 allele was less active toward this substrate. Immunoblot analysis of 16 human liver donor samples indicated that (S)-mephenytoin 4'-hydroxylase activity correlated with the hepatic CYP2C19 content, but it did not correlate with the hepatic content of CYP2C9. Moreover, direct sequencing of the polymerase chain reaction (PCR) products of 2C9 mRNA from six of these human livers through areas of known allelic variations indicated that the identity of the allele of 2C9 (Cys144 vs Arg, Tyr358 vs Cys, Ile359 vs Leu, or Gly417 vs Asp) did not appear to influence (S)-mephenytoin 4'-hydroxylase activity in these samples. These data indicate that 2C19 is the principal determinant of (S)-mephenytoin 4'-hydroxylase activity in human liver.


Subject(s)
Aryl Hydrocarbon Hydroxylases , Cytochrome P-450 Enzyme System/metabolism , Liver/enzymology , Mixed Function Oxygenases/metabolism , Base Sequence , Cytochrome P-450 CYP2C19 , Gene Expression , Humans , Hydroxylation , Immunoblotting , Mephenytoin/metabolism , Microsomes, Liver/enzymology , Molecular Sequence Data , Polymerase Chain Reaction , RNA, Messenger/chemistry , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Sequence Analysis, RNA , Stereoisomerism , Substrate Specificity , Tolbutamide/metabolism
18.
Arch Biochem Biophys ; 304(1): 209-18, 1993 Jul.
Article in English | MEDLINE | ID: mdl-8323286

ABSTRACT

P4502E1 (2E1), an ethanol-inducible P450 enzyme, plays an important role in the bioactivation of certain hepatotoxins and chemical carcinogens. Different mechanisms of 2E1 induction by ethanol and other agents (e.g., acetone) have been proposed, ranging from enhanced de novo enzyme synthesis caused by an increase in 2E1 mRNA and/or the efficiency with which it is translated to decreased enzyme degradation stemming from substrate stabilization. To evaluate these mechanisms, we first examined the time course of hepatic 2E1 protein induction in rats pair-fed liquid diets containing 36% of total calories as either ethanol or dextrin-maltose (controls) for 28 days. Western blot analysis with anti-2E1 immunoglobulins revealed that 2E1 reached a new steady-state level (eightfold greater than that found with controls) after ethanol feeding for 10 days and remained elevated for the duration of treatment. Microsomal p-nitrophenol hydroxylation, a 2E1-catalyzed reaction, exhibited a similar induction time course, with the maximal increase in enzyme activity also observed on Day 10 of ethanol administration. We then determined steady-state 2E1 protein turnover in ethanol-fed and control animals that were given [35S]methionine plus[3H]aminolevulinate to radiolabel 2E1 apoprotein and the prosthetic heme group, respectively. Monophasic exponential decay curves showed that hepatic 2E1 protein and heme half-lives (27-28 h and 17 h, respectively) did not differ between the treatment groups. However, rates of 2E1 synthesis, assessed by measuring initial rates of incorporation of [35S]methionine and [3H]aminolevulinate into 2E1 apoprotein and heme, were increased in animals fed ethanol. Our results indicate that the in vivo induction of hepatic 2E1 protein by ethanol involves increased enzyme synthesis rather than decreased enzyme degradation. This enhancement of de novo 2E1 synthesis most likely entails the ethanol-mediated increase of steady-state levels of 2E1 mRNA and/or the stimulation of its translational efficiency.


Subject(s)
Cytochrome P-450 Enzyme System/biosynthesis , Ethanol/pharmacology , Microsomes, Liver/enzymology , Oxidoreductases, N-Demethylating/biosynthesis , Animals , Body Weight/drug effects , Cytochrome P-450 CYP2E1 , Enzyme Induction/drug effects , Gene Expression/drug effects , In Situ Hybridization , Male , Precipitin Tests , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Streptozocin/pharmacology
19.
Biochem Pharmacol ; 45(7): 1483-92, 1993 Apr 06.
Article in English | MEDLINE | ID: mdl-8471070

ABSTRACT

The molecular mechanism(s) underlying induction of the hepatic microsomal cytochrome P4502E1 (2E1) by xenobiotics (e.g. ethanol and acetone) is controversial. Proposed mechanisms include increased rates of enzyme synthesis due to elevated 2E1 mRNA levels, enhanced translation of pre-existing mRNA, or stabilization of 2E1 protein. To further assess which, if any, of these events predominates during the initial stages of 2E1 protein induction, we investigated the effects of acetone treatment on 2E1 content in cultured rabbit hepatocytes, an in vitro system that allows for precise control of the cellular mileau. Hepatocytes harvested from female rabbits and plated on plastic dishes with serum-supplemented medium were 90-100% viable for at least 48 hr in culture. Analysis of immunoreactive 2E1 content and aniline hydroxylase activity in microsomes isolated from hepatocytes cultured for up to 24 hr revealed that 2E1 expression was equal to that of microsomes from unplated cells and by 48 hr of culture, 2E1 levels decreased by only 35%. Moreover, microsomes isolated from cells exposed to 17 mM acetone for 24 hr exhibited a 53 and 62% increase in aniline hydroxylase activity and 2E1 content, respectively, compared to untreated cells. To explain these increases, the rate of 2E1 protein synthesis was determined in untreated cells or in cells treated with 17 mM acetone by first exposing hepatocytes to medium supplemented with 35S-labeled methionine and cysteine ([35S]Met/Cys) and subsequently assessing radiolabel incorporation into 2E1 protein. While no difference was found between untreated and acetone-treated cells in the incorporation of [35S]Met/Cys into trichloracetic acid-precipitable microsomal proteins, immunoaffinity purification of 2E1 revealed that incorporation of 35S-labeled amino acids specifically into 2E1 was elevated by acetone to 200% of control values. Treatment of hepatocytes with the transcriptional inhibitor, alpha-amanitin, markedly inhibited this acetone-mediated increase in [35S]Met/Cys incorporation into 2E1. Analysis of hepatocyte RNA revealed that acetone increased 2E1 mRNA to 130 and 160% of control levels at 6 and 24 hr, respectively, and that these increases were prevented by pretreatment with alpha-amanitin. Our results indicate that acetone increases 2E1 protein levels in cultured rabbit hepatocytes by stimulating its rate of de novo synthesis. Since this increase in 2E1 synthesis stems, at least in part, from the acetone-mediated enhancement of hepatocyte 2E1 mRNA content and is inhibitable by alpha-amanitin, transcriptional activation of the rabbit CYP2E1 gene is apparently involved in the induction of 2E1 protein by acetone.


Subject(s)
Acetone/pharmacology , Cytochrome P-450 Enzyme System/biosynthesis , Liver/drug effects , Oxidoreductases, N-Demethylating/biosynthesis , RNA, Messenger/biosynthesis , Animals , Cells, Cultured , Cytochrome P-450 CYP2E1 , Cytochrome P-450 Enzyme System/genetics , Enzyme Induction/drug effects , Female , Liver/enzymology , Oxidoreductases, N-Demethylating/genetics , Protein Biosynthesis , Rabbits , Up-Regulation
20.
Hepatology ; 17(2): 236-45, 1993 Feb.
Article in English | MEDLINE | ID: mdl-8428720

ABSTRACT

The propensity of centrilobular liver damage to develop in alcohol abusers after exposure to various hepatotoxins, including ethanol itself, has been linked to the induction by ethanol of P-4502E1, a microsomal P-450 enzyme that bioactivates these agents to reactive metabolites. Whereas long-term ethanol consumption elicits a marked increase in hepatic P-4502E1 content, the molecular mechanism by which ethanol produces this effect is the subject of controversy in animals, and it has not been elucidated in human beings. Possible mechanisms include increased enzyme synthesis stemming from elevated 2E1 messenger RNA levels, enhanced translation of preexisting messenger RNA or stabilization of P-4502E1 protein. To determine which, if any, of these mechanisms underlies P-4502E1 induction in human beings, we examined the effects of ethanol intake on the hepatic intralobular distribution of P-4502E1 messenger RNA and the corresponding protein. Liver sections derived from needle biopsy specimens were obtained from five recently drinking alcoholics (last drink no more than 36 hr before) and eight control subjects (five abstaining alcoholics [last drink 96 hr or more before] and three nondrinkers). In situ hybridization of these liver sections with a human P-4502E1 complementary DNA probe was used to localize P-4502E1 messenger RNA transcripts. Quantitative image analysis of hybridized sections from control subjects revealed that P-4502E1 transcript content in perivenular (zone 3) hepatocytes was significantly higher (p < 0.05) than in midzonal (zone 2) and periportal (zone 1) cells (18.3 +/- 1, 9.5 +/- 2 and 3.1 +/- 2 arbitrary density units, respectively; mean +/- S.E.M.). In recent drinkers, acinar regions containing P-4502E1 transcripts were elevated 2.9-fold compared with those in controls (32.8% +/- 7% vs. 11.2% +/- 2%; p < 0.01), with this messenger RNA increase occurring mainly in perivenular cells (29.6 +/- 3 vs. 18.3 +/- 1 units; p < 0.01). P-4502E1 protein distribution, assessed by the immunohistochemical staining of liver sections with P-4502E1 antibodies, was found to be analogous to that of the messenger RNA in control subjects (the level in perivenular cells was greater than that in midzonal cells, which was greater than that in periportal cells), whereas recent drinkers exhibited marked elevations in enzyme content in both perivenular and midzonal hepatocytes. Moreover, cellular levels of P-4502E1 protein and messenger RNA were significantly correlated (rs = 0.79; p < 0.001) in all patients.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Ethanol/pharmacology , Liver/enzymology , Oxidoreductases, N-Demethylating/metabolism , RNA, Messenger/metabolism , Adult , Aged , Base Sequence , Cytochrome P-450 CYP2E1 , Cytochrome P-450 Enzyme System/genetics , Enzyme Induction/drug effects , Humans , Image Processing, Computer-Assisted , Immunohistochemistry , In Situ Hybridization , Liver/pathology , Male , Middle Aged , Molecular Probes/genetics , Molecular Sequence Data , Oxidoreductases, N-Demethylating/genetics , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...