Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Drug Metab Dispos ; 29(11): 1446-53, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11602520

ABSTRACT

Under certain culture conditions, exposure of the human colon adenocarcinoma cell line Caco-2 to 1,25-(OH)(2)-D(3) induces expression of CYP3A4 to levels comparable to that in human small intestinal epithelium. To determine whether 1,25-(OH)(2)-D(3) could be used to restore CYP3A expression in other culture models, we examined several cell lines derived from malignancies of human tissues known to express CYP3A enzymes: Hep G2 (liver), LS180 (colon), HPAC (pancreas), Hs746T (stomach). Primary cultures of human hepatocytes from two donors were also examined. 1,25-(OH)(2)-D(3) increased CYP3A catalytic activity in LS180 (15-fold), HPAC (6-fold), and hepatocytes (2- to 3-fold); this was accompanied by induction of CYP3A4 mRNA and CYP3A immunoreactive protein. However, 1,25-(OH)(2)-D(3) had no effect on CYP3A expression in Hs746T or Hep G2. Known ligands for pregnane X receptor (PXR) (rifampin, dexamethasone, and dexamethasone t-butyl acetate) markedly induced CYP3A4 expression in human hepatocytes. In contrast, these ligands had little or no effect on CYP3A4 expression in Caco-2 cells, even at concentrations 1 to 2 orders of magnitude greater than effective concentrations of 1,25-(OH)(2)-D(3) or two other vitamin D receptor (VDR) ligands (25-OH-D(3) and 1-OH-D(3)). The retinoic acid receptor ligand all-trans-retinoic acid augmented the 1,25-(OH)(2)-D(3)-mediated induction of CYP3A4 catalytic activity up to 2-fold in Caco-2 cells, while having no demonstrable effect on levels of CYP3A4 mRNA or protein. The retinoid X receptor ligand 9-cis-retinoic acid appeared to slightly reduce CYP3A4 catalytic activity. We conclude that 1,25-(OH)(2)-D(3) can be used to increase CYP3A4 expression in some, but not all, human cell lines derived from tissues known to express CYP3A enzymes. The mechanisms involved in this induction are unlikely to involve PXR and may involve VDR.


Subject(s)
Calcitriol/pharmacology , Calcium Channel Agonists/pharmacology , Cytochrome P-450 Enzyme System/biosynthesis , Mixed Function Oxygenases/biosynthesis , Receptors, Cytoplasmic and Nuclear/physiology , Receptors, Steroid/physiology , Caco-2 Cells/drug effects , Caco-2 Cells/enzymology , Calcitriol/analogs & derivatives , Cells, Cultured , Cytochrome P-450 CYP3A , Enzyme Induction/drug effects , Hepatocytes/drug effects , Hepatocytes/enzymology , Humans , Pregnane X Receptor
2.
J Pharmacol Exp Ther ; 289(2): 1134-42, 1999 May.
Article in English | MEDLINE | ID: mdl-10215697

ABSTRACT

Cytochrome P-450 (CYP) 3A4 accounts for approximately 50% of all P-450s found in the small intestine (Paine et al., 1997) and contributes to the extensive and variable first-pass extraction of drugs such as cyclosporine and saquinavir. We recently demonstrated that CYP3A4 expression in a differentiated Caco-2 subclone is increased when cell monolayers are treated with 1alpha,25-dihydroxy-vitamin-D3 (Schmiedlin-Ren et al., 1997). This improved metabolic capacity permits the in vitro modeling of first-pass intestinal metabolic kinetics. Midazolam (MDZ) 1'-hydroxylation was used as a specific probe for CYP3A-mediated metabolism in modified Caco-2 monolayers. Caco-2 cells were grown to confluence on laminin-coated culture inserts, and then for two additional weeks in the presence of 1alpha,25-dihydroxy vitamin-D3. Cell monolayers were subsequently exposed to MDZ for varying lengths of time and concentrations. The amount of MDZ in the monolayer increased rapidly after apical drug administration, reaching a pseudo steady state within 6 min. The cellular uptake rate was considerably slower after a basolateral dose. By either route of administration, the rate of 1'-hydroxymidazolam formation was stable and linear for 2 h. Under basolateral sink conditions and low apical MDZ dosing concentration (1-8 microM), the first-pass extraction ratio was found to be approximately 15%. Higher dosing concentrations led to saturation of the hydroxylation reaction and reduction in the extraction ratio. The modified Caco-2 cell monolayer is an excellent model for studying drug absorption and first-pass intestinal metabolic kinetic processes. In this system, the selective CYP3A probe MDZ was rapidly absorbed, yet extensively metabolized, as is observed in vivo.


Subject(s)
Calcitriol/pharmacology , Midazolam/metabolism , Biotransformation , Blood Proteins/metabolism , Blotting, Western , Caco-2 Cells , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/metabolism , Electric Conductivity , Extracellular Space/metabolism , Humans , Kinetics , Midazolam/analogs & derivatives , Midazolam/blood , Midazolam/pharmacokinetics , Mixed Function Oxygenases/metabolism , Permeability , Reproducibility of Results
3.
Drug Metab Dispos ; 27(3): 360-4, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10064566

ABSTRACT

Human cytochrome P-450 1A1 (CYP1A1) is located primarily in extrahepatic tissues. To begin the characterization of this enzyme in the small intestine, we screened a bank of 18 human small intestinal microsomal preparations for CYP1A1 catalytic [(7-ethoxyresorufin O-deethylase (EROD)] activity and protein content. Although EROD activity was below detectable limits in 12 of the preparations, 6 exhibited measurable activity (1.4-123.5 pmol/min/mg), some exceeding that for 2 human liver microsomal preparations (11.0 and 26.4 pmol/min/mg). This variation was not due to variable quality of the preparations because each sample displayed readily detectable CYP3A4 catalytic activity and immunoreactive protein. We inadvertently found that intestinal EROD activity was inhibitable by ketoconazole at a concentration commonly believed to selectively inhibit CYP3A4. The possibility that CYP3A4 metabolizes 7-ethoxyresorufin was excluded because there was no correlation between intestinal CYP3A4 catalytic and EROD activity, and cDNA-expressed human CYP3A4 exhibited no EROD activity. Moreover, CYP1A1 immunoreactive protein was most abundant in the three intestinal preparations with the highest EROD activities, and the mean apparent Ki of ketoconazole observed for these three preparations (40 nM) was essentially identical with that for cDNA-expressed human CYP1A1 (37 nM). In summary, there is large interindividual variation in CYP1A1 expression in human small bowel, and ketoconazole is not a selective CYP3A4 inhibitor in in vitro metabolism studies involving intestinal tissue obtained from some individuals. These observations raise the possibility that in vivo drug interactions involving ketoconazole could result from CYP1A1 inhibition in the intestine in some individuals.


Subject(s)
Cytochrome P-450 CYP1A1/antagonists & inhibitors , Cytochrome P-450 CYP1A1/biosynthesis , Enzyme Inhibitors/pharmacology , Intestine, Small/drug effects , Intestine, Small/enzymology , Ketoconazole/pharmacology , Blotting, Western , Caco-2 Cells/enzymology , Cytochrome P-450 CYP1A1/metabolism , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/metabolism , Humans , Microsomes/drug effects , Microsomes/enzymology , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Mixed Function Oxygenases/antagonists & inhibitors , Mixed Function Oxygenases/metabolism , Sensitivity and Specificity
4.
Drug Metab Dispos ; 25(11): 1228-33, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9351897

ABSTRACT

Grapefruit juice increases the oral availability of a variety of CYP3A4 substrates. It has been shown that recurrent grapefruit juice ingestion results in a loss of CYP3A4 from the small bowel epithelium. We now show that the reduction in intestinal CYP3A4 concentration is rapid; a 47% decrease occurred in a healthy volunteer within 4 hr after consuming grapefruit juice. To identify the specific components of the juice responsible for this effect, we used a recently developed Caco-2 cell culture model of human intestinal epithelium that expresses catalytically active CYP3A4. We found that grapefruit oil and two furanocoumarin constituents (6', 7'-dihydroxybergamottin and a closely related dimer) caused a dose-dependent fall in CYP3A4 catalytic activity and immunoreactive CYP3A4 concentration. The effect was selective in that concentrations of CYP1A1 and CYP2D6 did not fall, consistent with previous results obtained in vivo. Assays of various juices confirmed that 6',7'-dihydroxybergamottin is the major furanocoumarin present and, although its concentration varies significantly among types and brands of grapefruit juice, it is consistently present in concentrations exceeding the IC50 (1 microM) for loss of midazolam 1'-hydroxylase activity determined in the Caco-2 cells. Studies with recombinant CYP3A4 revealed that 6', 7'-dihydroxybergamottin is a mechanism-based inactivator, which supports the idea that loss of CYP3A4 results from accelerated degradation of the enzyme. We conclude that the effect of grapefruit juice on oral availability of CYP3A4 substrates can be largely accounted for by the presence of 6',7'-dihydroxybergamottin although other furanocoumarins probably also contribute.


Subject(s)
Citrus/chemistry , Cytochrome P-450 Enzyme Inhibitors , Enzyme Inhibitors/pharmacology , Furocoumarins/pharmacology , Mixed Function Oxygenases/antagonists & inhibitors , Biological Availability , Caco-2 Cells , Chromatography, High Pressure Liquid , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/biosynthesis , Cytochrome P-450 Enzyme System/metabolism , Enzyme Inhibitors/chemistry , Escherichia coli/metabolism , Furocoumarins/chemistry , Humans , Immunoblotting , Intestinal Mucosa/cytology , Intestinal Mucosa/drug effects , Intestinal Mucosa/enzymology , Microsomes/drug effects , Microsomes/enzymology , Mixed Function Oxygenases/biosynthesis , Mixed Function Oxygenases/metabolism
5.
Clin Pharmacol Ther ; 62(3): 248-60, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9333100

ABSTRACT

Interpatient differences in the oral clearance of cyclosporine (INN, ciclosporin) have been partially attributed to variation in the activity of a single liver enzyme termed CYP3A4. Recently it has been shown that small bowel also contains CYP3A4, as well as P-glycoprotein, a protein able to transport cyclosporine. To assess the importance of these intestinal proteins, the oral pharmacokinetics of cyclosporine were measured in 25 kidney transplant recipients who each had their liver CYP3A4 activity quantitated by the intravenous [14C-N-methyl]-erythromycin breath test and who underwent small bowel biopsy for measurement of CYP3A4 and P-glycoprotein. Forward multiple regression revealed that 56% (i.e., r2 = 0.56) and 17% of the variability in apparent oral clearance [log (dose/area under the curve)] were accounted for by variation in liver CYP3A4 activity (p < 0.0001) and intestinal P-glycoprotein concentration (p = 0.0059), respectively. For peak blood concentration, liver CYP3A4 activity accounted for 32% (p = 0.0002) and P-glycoprotein accounted for an additional 30% (p = 0.0024) of the variability. Intestinal levels of CYP3A4, which varied tenfold, did not appear to influence any cyclosporine pharmacokinetic parameter examined. We conclude that intestinal P-glycoprotein plays a significant role in the first-pass elimination of cyclosporine, presumably by being a rate-limiting step in absorption. Drug interactions with cyclosporine previously ascribed to intestinal CYP3A4 may instead be mediated by interactions with intestinal P-glycoprotein.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Cyclosporine/pharmacokinetics , Cytochrome P-450 Enzyme System/metabolism , Immunosuppressive Agents/pharmacokinetics , Mixed Function Oxygenases/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/blood , Administration, Oral , Adult , Aged , Area Under Curve , Biological Availability , Breath Tests , Cyclosporine/administration & dosage , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/blood , Duodenum/metabolism , Female , Humans , Immunoblotting , Immunosuppressive Agents/administration & dosage , Kidney Transplantation , Liver/metabolism , Male , Middle Aged , Mixed Function Oxygenases/blood , Regression Analysis
6.
Mol Pharmacol ; 51(5): 741-54, 1997 May.
Article in English | MEDLINE | ID: mdl-9145912

ABSTRACT

The human colon carcinoma cell line, Caco-2, is widely used as a model for oral absorption of xenobiotics. The usefulness of Caco-2 cells has been limited, however, because they do not express appreciable quantities of CYP3A4, the principle cytochrome P450 present in human small bowel epithelial cells. We report that treatment of Caco-2 cells with 1 alpha,25-dihydroxyvitamin D3, beginning at confluence, results in a dose- and duration-dependent increase in CYP3A4 mRNA and protein, with little apparent effect on the expression of CYP3A5 or CYP3A7. This treatment also results in increases in NADPH cytochrome P450 reductase and P-glycoprotein (the MDR1 gene product) but has no detectable effect on expression of CYP1A1, CYP2D6, cytochrome b5, liver or intestinal fatty acid binding proteins, or villin. Maximal expression of CYP3A4 requires an extracellular matrix on a permeable support and the presence of serum. In the treated cells, the intrinsic formation clearance of 1'-hydroxymidazolam (a reaction characteristically catalyzed by CYP3A enzymes) was estimated to be somewhat lower than that of human jejunal mucosa (1.14 and 3.67 ml/min/g of cells, respectively). The 1'-OH-midazolam/4-OH-midazolam product ratio produced by the cells (approximately 5.3) is comparable to, but somewhat lower than, that observed in human jejunal microsomes (7.4-15.4), which may reflect the presence of CYP3A7 in the Caco-2 cells. 25-Hydroxyvitamin D3 is less efficacious but reproduces the effects of the dihydroxy compound, whereas unhydroxylated vitamin D is without appreciable effect. These observations, together with the time course of response, suggest that the vitamin D receptor may be involved in CYP3A4 regulation. The culture model we describe should prove useful in defining the role of CYP3A4 in limiting the oral bioavailability of many xenobiotics.


Subject(s)
Calcitriol/pharmacology , Cytochrome P-450 Enzyme System/genetics , Extracellular Matrix/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Intestine, Small/enzymology , Mixed Function Oxygenases/genetics , Caco-2 Cells , Cell Division , Cytochrome P-450 CYP3A , Humans , Intestine, Small/cytology , Molecular Sequence Data , Polymerase Chain Reaction
7.
Pharmacogenetics ; 6(5): 379-85, 1996 Oct.
Article in English | MEDLINE | ID: mdl-8946469

ABSTRACT

There is a marked variation between people in the activity of CYP3A4 in liver and intestine. We reasoned that if CYP3A4 was expressed in peripheral blood cells, a simple blood based test of CYP3A4 phenotype might be feasible. We prepared peripheral blood smears from healthy volunteers and performed immunostaining with a rabbit polyclonal antibody that selectively reacts with enzymes within the CYP3A subfamily. Staining was observed only within the cytoplasm of neutrophils (PMNs). cDNA prepared from isolated PMNs and mononuclear cells was subjected the polymerase chain reaction using as primers synthetic oligonucleotides that selectively amplify fragments of each known CYP3A cDNAs (CYP3A3, CYP3A4, CYP3A5, and CYP3A7). Amplification was only obtained with the CYP3A5 specific oligonucleotides, predominantly in PMNs, and the identity of the amplified fragment was confirmed by sequencing. Next, whole white cell homogenate prepared from human blood was reacted on immunoblots with a monoclonal antibody that recognizes all CYP3A proteins or an absorbed polyclonal antibody that recognizes only CYP3A5. Both antibodies recognized a protein in the white cells that comigrated with purified CYP3A5. However, metabolism of midazolam, a substrate of CYP3A, could not be detected in homogenates of isolated granulocytes, in homogenates of the whole WBC fractions, or in incubations with unlysed WBC preparations. We conclude that CYP3A4 is not expressed in peripheral blood and hence a blood phenotyping test for this enzyme will not be feasible. Our discovery that CYP3A5 is present may be important since this enzyme is also present in the liver intestine and kidney of many people.


Subject(s)
Cytochrome P-450 Enzyme System/analysis , Cytochrome P-450 Enzyme System/blood , Mixed Function Oxygenases/analysis , Neutrophils/enzymology , Cytochrome P-450 CYP3A , Humans , Immunohistochemistry , Polymerase Chain Reaction , RNA, Messenger/isolation & purification , Tissue Distribution
8.
Pharmacogenetics ; 4(5): 247-59, 1994 Oct.
Article in English | MEDLINE | ID: mdl-7894497

ABSTRACT

CYP3A4, a major Phase I xenobiotic metabolizing enzyme present in liver, is also present in human small bowel epithelium where it appears to catalyse significant 'first pass' metabolism of some drugs. To determine whether CYP3A4 or the related enzymes CYP3A3, CYP3A5, and CYP3A7 are present in other regions of the digestive tract, we used CYP3A-specific antibodies to examine histological sections and epithelial microsomes obtained from a human organ donor. CYP3A-related proteins were detected in epithelia throughout the digestive tract and in gastric parietal cells, in pericentral hepatocytes, and in ductular cells of the pancreas. Immunoblot analysis suggested that the major CYP3A protein present in liver, jejunum, colon, and pancreas was CYP3A4 or CYP3A3, whereas CYP3A5 was the major protein present in stomach. Both CYP3A4 and CYP3A5 mRNA were detectable in all regions of the digestive tract using the polymerase chain reaction (PCR); however, only CYP3A4 could be detected by Northern blot analysis. CYP3A7 mRNA was consistently detected only in the liver by PCR and CYP3A3 mRNA was not detected in any of the tissues. We conclude that CYP3A4 and CYP3A5 are present throughout the human digestive tract and that differences in the expression of these enzymes may account for inter-organ differences in the metabolism of CYP3A substrates.


Subject(s)
Cytochrome P-450 Enzyme System/genetics , Gene Expression Regulation, Enzymologic , Intestinal Mucosa/enzymology , Intestine, Small/enzymology , Mixed Function Oxygenases/genetics , Adult , Base Sequence , Blotting, Western , Cytochrome P-450 CYP2E1 , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/metabolism , DNA, Complementary , Female , Humans , Immunohistochemistry , Mixed Function Oxygenases/metabolism , Molecular Sequence Data , RNA, Messenger/genetics , RNA, Messenger/metabolism
9.
Gastroenterology ; 106(2): 433-9, 1994 Feb.
Article in English | MEDLINE | ID: mdl-8299909

ABSTRACT

BACKGROUND/AIMS: Hepatic CYP3A enzymes have been implicated in the bioactivation of aflatoxin B1 (AFB1) to DNA binding metabolites. CYP3A enzymes are also abundant in the small bowel, and we therefore examined the ability of this tissue to form intracellular AFB1 adducts. METHODS: Immunohistochemistry using a antibody to the stable AFB1-DNA adduct was performed on small bowel sections obtained from rats orally gavaged with AFB1 and on human small bowel biopsy specimens maintained in explant culture. 3H-AFB1 was instilled into a loop of small bowel of untreated rats and rats pretreated with the CYP3A inducer dexamethasone during vivisection. DNA was isolated from the loop 2 hours later and assayed for specific activity. RESULTS: In both rats and humans, AFB1-adducts were detected exclusively in mature enterocytes in a pattern similar to the distribution of CYP3A enzymes. Induction of enterocyte CYP3A in rats resulted in an increase in enterocyte immunoreactive AFB1 adducts and in a 1.8-fold increase in 3H-AFB1-nucleic acid adducts (P = 0.01). CONCLUSIONS: Intracellular AFB1 adducts are formed in the small intestine, and this reflects, at least in part, the catalytic activity of CYP3A enzymes. Because these AFB1 adducts should ultimately pass in stool, enterocyte CYP3A may represent a regulatable barrier to dietary aflatoxins.


Subject(s)
Aflatoxin B1/metabolism , DNA Adducts , DNA/metabolism , Intestine, Small/metabolism , Animals , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/physiology , Female , Humans , Immunohistochemistry , Intestine, Small/cytology , Isoenzymes/physiology , Rats , Rats, Sprague-Dawley
10.
Biochem Pharmacol ; 46(5): 905-18, 1993 Sep 01.
Article in English | MEDLINE | ID: mdl-8373442

ABSTRACT

Enzymes within the CYP3A subfamily are major Phase I drug-metabolizing enzymes present in hepatocytes and small bowel enterocytes. These enzymes are highly inducible in the liver by many structurally diverse compounds, including a number of commonly used medications. Studies indicate that CYP3A enzymes present in small bowel enterocytes are also inducible. However, the regulation of CYP3A enzymes in this tissue has not been well characterized, in part because in vivo studies are difficult, especially in humans. Our goals was to develop an in vitro model to study the regulation of CYP3A in enterocytes. To this end, we defined culture conditions under which adult rat jejunal explants maintained viable appearing villi for 21 hr. When dexamethasone, the prototypical inducer of CYP3A1 in rat hepatocytes, was added to the culture medium, there was a time-dependent induction of CYP3A1 mRNA and CYP3A protein in explant enterocytes which was essentially indistinguishable from the time course of induction of CYP3A1 mRNA and protein in enterocytes in vivo. This effect of dexamethasone appeared to be specific since dexamethasone had no consistent effect on the explant concentration of another enterocyte specific mRNA, intestinal fatty acid binding protein. Using this explant culture model, we found that CYP3A1 mRNA was also inducible by clotrimazole but we were unable to detect induction by rifampicin or troleandomycin. Our observations suggest that jejunal explants may provide an appropriate model for the study of the regulation of CYP3A and other drug-metabolizing enzymes.


Subject(s)
Aryl Hydrocarbon Hydroxylases , Cytochrome P-450 Enzyme System/genetics , Isoenzymes/genetics , Jejunum/enzymology , Animals , Clotrimazole/pharmacology , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/biosynthesis , Dexamethasone/pharmacology , Enzyme Induction/drug effects , Female , Gene Expression Regulation , Jejunum/cytology , Jejunum/ultrastructure , Models, Biological , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Time Factors
11.
J Clin Invest ; 90(5): 1871-8, 1992 Nov.
Article in English | MEDLINE | ID: mdl-1430211

ABSTRACT

Enzymes within the P450IIIA (CYP3A) subfamily appear to account for significant "first pass" metabolism of some drugs in the intestine. To identify which of the known P450IIIA genes are expressed in intestine, enterocyte RNA was hybridized on Northern blots with synthetic oligonucleotides complementary to hypervariable regions of hepatic P450IIIA4, P450IIIA5, and P450IIIA7 cDNAs. Hybridization was detected only with the P450IIIA4-specific oligonucleotide. The identity of the hybridizing mRNA was confirmed to be P450IIIA4 by direct sequencing of a DNA fragment amplified from enterocyte cDNA by the polymerase chain reaction. To determine if enterocyte P450IIIA4 is inducible, biopsies of small bowel mucosa were obtained from five volunteers before and after they received 7d of treatment with rifampin, a known inducer of P450IIIA4 in liver. Rifampin treatment resulted in a five- or eightfold mean increase (P < 0.05) in the biopsy concentration of P450IIIA4 mRNA when normalized for content of sucrase isomaltase or intestinal fatty acid binding protein mRNAs, respectively. Rifampin also induced P450IIIA immunoreactive protein in enterocytes in each of the subjects, as judged by immunohistochemistry, and resulted in a 10-fold increase in P450IIIA4-specific catalytic activity (erythromycin N-demethylation) in the one patient studied. Our identification of inducible P450IIIA4 in enterocytes may in part account for drug interactions characteristic of P450IIIA4 substrates and suggests a strategy for controlling entry into the body of a major class of xenobiotics.


Subject(s)
Cytochrome P-450 Enzyme System/biosynthesis , Intestine, Small/enzymology , Rifampin/pharmacology , Adult , Base Sequence , Cyclosporine/pharmacology , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/genetics , Enzyme Induction , Female , Humans , Male , Molecular Sequence Data , RNA, Messenger/analysis
12.
Gastroenterology ; 102(4 Pt 1): 1186-98, 1992 Apr.
Article in English | MEDLINE | ID: mdl-1551527

ABSTRACT

The P450IIIA (CYP3A) cytochromes are a major family of enzymes that play an important role in the metabolism of many medications, including cyclosporine A, as well as some dietary xenobiotics, including aflatoxin B1. The purpose of the studies was to detect, localize, and characterize P450IIIA enzymes present throughout the digestive tract. To this end, P450IIIA-specific antibodies were used to examine gut epithelial microsomes and histological tissue sections obtained from the digestive tract of both male and female rats. P450IIIA-related proteins were detected in epithelia throughout the gut; however, the specific proteins expressed appeared to differ among digestive organs and between male and female rats. RNA obtained from the gut epithelia was also analyzed using P450IIIA-specific synthetic oligonucleotides as probes on Northern blots and as primers for the polymerase chain reaction. P450IIIA1, which is a dexamethasone inducible enzyme in liver, was also found to be induced by dexamethasone treatment in epithelia from stomach and jejunum, but not from colon or esophagus. It was concluded that P450IIIA enzymes are present in mature epithelia throughout the gastrointestinal tract. However, expression of the P450IIIA enzymes is influenced by anatomic location and gender.


Subject(s)
Cytochrome P-450 Enzyme System/analysis , Digestive System/enzymology , Animals , Base Sequence , Cytochrome P-450 Enzyme System/genetics , DNA/analysis , Dexamethasone/pharmacology , Epithelium/enzymology , Female , Immunoblotting , Immunohistochemistry , Male , Molecular Sequence Data , RNA, Messenger/analysis , Rats , Rats, Inbred Strains , Sex Factors
13.
Transplantation ; 53(3): 596-602, 1992 Mar.
Article in English | MEDLINE | ID: mdl-1549853

ABSTRACT

Cyclosporine is converted to its major metabolites (M-17, M-1, and M-21) in human liver by enzymes belonging to the P450IIIA subfamily. These enzymes are also present in rat and human enterocytes; however, the possibility that CsA is metabolized in enterocytes has not been previously investigated. We therefore directly compared metabolism of 3H-CsA in microsomes prepared from liver and jejunal enterocytes. M-17, M-1, and M-21 were the major CsA metabolites produced by enterocyte microsomes. This metabolism appeared to be catalyzed by P450IIIA, because pretreatment of rats with the P450IIIA inducer dexamethasone significantly increased the rate of CsA metabolism in enterocyte microsomes and preincubation of enterocyte microsomes with anti-P450IIIA IgG inhibited the production of CsA metabolites by greater than 95%. To determine if enterocyte P450IIIA metabolizes CsA in vivo, rats were pretreated with the P450IIIA inducer dexamethasone, the P450IIIA inhibitor erythromycin, or vehicle alone. At laparotomy, 2 mg/kg of 3H-CsA was injected into a sealed loop of jejunum, and after collection of the mesenteric venous blood draining this segment for 45 min, the production of M-17 and M-1 was measured. In the control group, a mean of 3.9% of the recovered radioactivity was found as M-1 and M-17. In the rats pretreated with dexamethasone, a mean of 8.4% of the radioactivity was found as M-1 and M-17 (P less than 0.05 relative to control) and this decreased to 2.3% in the group pretreated with erythromycin (P = 0.08 relative to control). We conclude that P450IIIA in jejunal enterocytes readily metabolizes CsA. Furthermore, the metabolism of CsA by enterocytes in vivo is substantial and likely contributes to "first pass metabolism" of orally administered CsA. Our observations provide novel hypotheses to explain some important drug interactions and interpatient differences in CsA dosing requirements.


Subject(s)
Cyclosporine/metabolism , Cytochrome P-450 Enzyme System/metabolism , Jejunum/cytology , Administration, Oral , Animals , Biological Availability , Cyclosporine/administration & dosage , Cyclosporine/pharmacokinetics , Dexamethasone/pharmacology , Female , Jejunum/enzymology , Jejunum/metabolism , Male , Microsomes/enzymology , Microsomes, Liver/enzymology , Rats , Rats, Inbred Strains
SELECTION OF CITATIONS
SEARCH DETAIL
...