Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Mol Pharmacol ; 93(1): 14-24, 2018 01.
Article in English | MEDLINE | ID: mdl-29093019

ABSTRACT

The contributions of cytochrome P450 3A5 to the metabolic clearance of marketed drugs is unclear, but its probable role is to augment the metabolism of several drugs that are largely cleared by P450 3A4. Selective metabolism by 3A4 is often a concern in drug development owing to potential drug-drug interactions and the variability of 3A4 and 3A5 expression. The contribution of P450 3A5 to these clearance pathways varies between individuals owing to genetic differences and similarities and differences in the metabolic properties of 3A5 compared with 3A4. To better understand the structural differences between P450s 3A4 and 3A5, the structure of 3A5 complexed with ritonavir was determined by X-ray crystallography to a limiting resolution of 2.91 Å. The secondary and tertiary structures of 3A5 and 3A4 are similar, but the architectures of their active sites differ. The 3A5 active site is taller and narrower than that of 3A4. As a result, ritonavir adopts a distinctly different conformation to fit into the cavity of 3A5 than seen for 3A4. These structural changes reflect amino acid differences that alter the conformation of the helix F through helix G region in the upper portion of the cavity and ionic interactions between residues in the beta-sheet domain that reduce the width of the cavity. The structural differences exhibited by 3A4 and 3A5 suggest that the overlap of catalytic activities may reflect molecular flexibility that determines how alternative conformers fit into the different active site architectures of the two enzymes.


Subject(s)
Cytochrome P-450 CYP3A Inhibitors/chemistry , Cytochrome P-450 CYP3A/chemistry , Ritonavir/chemistry , Binding Sites/physiology , Crystallography, X-Ray/methods , Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 CYP3A Inhibitors/metabolism , Humans , Protein Structure, Secondary , Protein Structure, Tertiary , Ritonavir/metabolism
2.
J Biol Chem ; 292(27): 11230-11242, 2017 07 07.
Article in English | MEDLINE | ID: mdl-28533430

ABSTRACT

Cytochrome P450 (P450, CYP) 4A11 is a human fatty acid ω-hydroxylase that catalyzes the oxidation of arachidonic acid to the eicosanoid 20-hydroxyeicosatetraenoic acid (20-HETE), which plays important roles in regulating blood pressure regulation. Variants of P450 4A11 have been associated with high blood pressure and resistance to anti-hypertensive drugs, and 20-HETE has both pro- and antihypertensive properties relating to increased vasoconstriction and natriuresis, respectively. These physiological activities are likely influenced by the redox environment, but the mechanisms are unclear. Here, we found that reducing agents (e.g. dithiothreitol and tris(2-carboxyethyl)phosphine) strongly enhanced the catalytic activity of P450 4A11, but not of 10 other human P450s tested. Conversely, added H2O2 attenuated P450 4A11 catalytic activity. Catalytic roles of five of the potentially eight implicated Cys residues of P450 4A11 were eliminated by site-directed mutagenesis. Using an isotope-coded dimedone/iododimedone-labeling strategy and mass spectrometry of peptides, we demonstrated that the heme-thiolate cysteine (Cys-457) is selectively sulfenylated in an H2O2 concentration-dependent manner. This sulfenylation could be reversed by reducing agents, including dithiothreitol and dithionite. Of note, we observed heme ligand cysteine sulfenylation of P450 4A11 ex vivo in kidneys and livers derived from CYP4A11 transgenic mice. We also detected sulfenylation of murine P450 4a12 and 4b1 heme peptides in kidneys. To our knowledge, reversible oxidation of the heme thiolate has not previously been observed in P450s and may have relevance for 20-HETE-mediated functions.


Subject(s)
Cytochrome P-450 CYP4A/chemistry , Dithiothreitol/chemistry , Heme/chemistry , Hydrogen Peroxide/chemistry , Animals , Catalysis , Cytochrome P-450 CYP4A/genetics , Cytochrome P-450 CYP4A/metabolism , Dithiothreitol/metabolism , Heme/genetics , Heme/metabolism , Humans , Hydrogen Peroxide/metabolism , Hydroxyeicosatetraenoic Acids/biosynthesis , Hydroxyeicosatetraenoic Acids/chemistry , Hydroxyeicosatetraenoic Acids/genetics , Kidney/enzymology , Liver/enzymology , Mice , Mice, Transgenic , Oxidation-Reduction , Rats
3.
J Biol Chem ; 291(32): 16904-19, 2016 08 05.
Article in English | MEDLINE | ID: mdl-27298316

ABSTRACT

Male and female homozygous 129/Sv mice carrying four copies of the human cytochrome P450 4A11 gene (CYP4A11) under control of its native promoter (B-129/Sv-4A11(+/+)) develop hypertension (142 ± 8 versus 113 ± 7 mm Hg systolic blood pressure (BP)), and exhibit increased 20-hydroxyeicosatetraenoic acid (20-HETE) in kidney and urine. The hypertension is reversible by a low-sodium diet and by the CYP4A inhibitor HET0016. B-129/Sv-4A11(+/+) mice display an 18% increase of plasma potassium (p < 0.02), but plasma aldosterone, angiotensin II (ANGII), and renin activities are unchanged. This phenotype resembles human genetic disorders with elevated activity of the sodium chloride co-transporter (NCC) and, accordingly, NCC abundance is increased by 50% in transgenic mice, and NCC levels are normalized by HET0016. ANGII is known to increase NCC abundance, and renal mRNA levels of its precursor angiotensinogen are increased 2-fold in B-129/Sv-4A11(+/+), and blockade of the ANGII receptor type 1 with losartan normalizes BP. A pro-hypertensive role for 20-HETE was implicated by normalization of BP and reversal of renal angiotensin mRNA increases by administration of the 20-HETE antagonists 2-((6Z,15Z)-20-hydroxyicosa-6,15-dienamido)acetate or (S)-2-((6Z,15Z)-20-hydroxyicosa-6,15-dienamido)succinate. SGK1 expression is also increased in B-129/Sv-4A11(+/+) mice and paralleled increases seen for NCC. Losartan, HET0016, and 20-HETE antagonists each normalized SGK1 mRNA expression. These results point to a potential 20-HETE dependence of intrarenal angiotensinogen production and ANGII receptor type 1 activation that are associated with increases in NCC and SGK1 and identify elevated P450 4A11 activity and 20-HETE as potential risk factors for salt-sensitive human hypertension by perturbation of the renal renin-angiotensin axis.


Subject(s)
Blood Pressure , Cytochrome P-450 CYP4A/biosynthesis , Hydroxyeicosatetraenoic Acids/metabolism , Hypertension/enzymology , Renin-Angiotensin System , Angiotensins/genetics , Angiotensins/metabolism , Animals , Cytochrome P-450 CYP4A/genetics , Female , Humans , Hydroxyeicosatetraenoic Acids/genetics , Hypertension/genetics , Losartan/pharmacology , Male , Mice , Mice, Inbred BALB C , Mice, Transgenic , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 1/metabolism , Sodium Chloride, Dietary/pharmacology , Solute Carrier Family 12, Member 3/biosynthesis , Solute Carrier Family 12, Member 3/genetics
4.
J Biol Chem ; 287(14): 10834-43, 2012 Mar 30.
Article in English | MEDLINE | ID: mdl-22308038

ABSTRACT

Human cytochrome P450 2D6 contributes to the metabolism of >15% of drugs used in clinical practice. This study determined the structure of P450 2D6 complexed with a substrate and potent inhibitor, prinomastat, to 2.85 Å resolution by x-ray crystallography. Prinomastat binding is well defined by electron density maps with its pyridyl nitrogen bound to the heme iron. The structure of ligand-bound P450 2D6 differs significantly from the ligand-free structure reported for the P450 2D6 Met-374 variant (Protein Data Bank code 2F9Q). Superposition of the structures reveals significant differences for ß sheet 1, helices A, F, F', G", G, and H as well as the helix B-C loop. The structure of the ligand complex exhibits a closed active site cavity that conforms closely to the shape of prinomastat. The closure of the open cavity seen for the 2F9Q structure reflects a change in the direction and pitch of helix F and introduction of a turn at Gly-218, which is followed by a well defined helix F' that was not observed in the 2F9Q structure. These differences reflect considerable structural flexibility that is likely to contribute to the catalytic versatility of P450 2D6, and this new structure provides an alternative model for in silico studies of substrate interactions with P450 2D6.


Subject(s)
Cytochrome P-450 CYP2D6/chemistry , Cytochrome P-450 CYP2D6/metabolism , Enzyme Inhibitors/metabolism , Organic Chemicals/metabolism , Catalytic Domain , Crystallography, X-Ray , Cytochrome P-450 CYP2D6 Inhibitors , Humans , Ligands , Models, Molecular , Protein Binding
5.
J Pharmacol Exp Ther ; 337(1): 125-36, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21205922

ABSTRACT

Activators of AMP-activated protein kinase (AMPK) increase the expression of the human microsomal fatty acid ω-hydroxylase CYP4F2. A 24-h treatment of either primary human hepatocytes or the human hepatoma cell line HepG2 with 5-aminoimidazole-4-carboxamide-1-ß-D-ribofuranoside (AICAR), which is converted to 5-aminoimidazole-4-carboxamide-1-ß-D-ribofuranosyl 5'-monophosphate, an activator of AMPK, caused an average 2.5- or 7-fold increase, respectively, of CYP4F2 mRNA expression but not of CYP4A11 or CYP4F3, CYP4F11, and CYP4F12 mRNA. Activation of CYP4F2 expression by AICAR was significantly reduced in HepG2 cells by an AMPK inhibitor, 6-[4-(2-piperidin-1-yl-ethoxy)-phenyl)]-3-pyridin-4-yl-pyrrazolo[1,5-a]-pyrimidine (compound C) or by transfection with small interfering RNAs for AMPKα isoforms α1 and α2. A 2.5-fold increase in CYP4F2 mRNA expression was observed upon treatment of HepG2 cells with 6,7-dihydro-4-hydroxy-3-(2'-hydroxy[1,1'-biphenyl]-4-yl)-6-oxo-thieno[2,3-b]pyridine-5-carbonitrile (A-769662), a direct activator for AMPK. In addition, the indirect activators of AMPK, genistein and resveratrol increased CYP4F2 mRNA expression in HepG2 cells. Pretreatment with compound C or 1,2-dihydro-3H-naphtho[2,1-b]pyran-3-one (splitomicin), an inhibitor of the NAD(+) activated deacetylase SIRT1, only partially blocked activation of CYP4F2 expression by resveratrol, suggesting that a SIRT1/AMPK-independent pathway also contributes to increased CYP4F2 expression. Compound C greatly diminished genistein activation of CYP4F2 expression. 7H-benz[de]benzimidazo[2,1-a]isoquinoline-7-one-3-carboxylic acid acetate (STO-609), a calmodulin kinase kinase (CaMKK) inhibitor, reduced the level of expression of CYP4F2 elicited by genistein, suggesting that CaMKK activation contributed to AMPK activation by genistein. Transient transfection studies in HepG2 cells with reporter constructs containing the CYP4F2 proximal promoter demonstrated that AICAR, genistein, and resveratrol stimulated transcription of the reporter gene. These results suggest that activation of AMPK by cellular stress and endocrine or pharmacologic stimulation is likely to activate CYP4F2 gene expression.


Subject(s)
AMP-Activated Protein Kinases/biosynthesis , Aminoimidazole Carboxamide/analogs & derivatives , Cytochrome P-450 Enzyme System/biosynthesis , Gene Expression Regulation, Enzymologic , Genistein/pharmacology , Ribonucleosides/pharmacology , Stilbenes/pharmacology , Aged , Aminoimidazole Carboxamide/pharmacology , Cells, Cultured , Cytochrome P450 Family 4 , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Enzyme Activation/physiology , Enzyme Induction/drug effects , Enzyme Induction/physiology , Female , Hep G2 Cells , Hepatocytes/enzymology , Humans , Male , Middle Aged , Resveratrol , Signal Transduction/drug effects , Signal Transduction/physiology
6.
J Biol Chem ; 286(7): 5736-43, 2011 Feb 18.
Article in English | MEDLINE | ID: mdl-21147782

ABSTRACT

The atomic structure of human P450 1B1 was determined by x-ray crystallography to 2.7 Å resolution with α-naphthoflavone (ANF) bound in the active site cavity. Although the amino acid sequences of human P450s 1B1 and 1A2 have diverged significantly, both enzymes exhibit narrow active site cavities, which underlie similarities in their substrate profiles. Helix I residues adopt a relatively flat conformation in both enzymes, and a characteristic distortion of helix F places Phe(231) in 1B1 and Phe(226) in 1A2 in similar positions for π-π stacking with ANF. ANF binds in a distinctly different orientation in P450 1B1 from that observed for 1A2. This reflects, in part, divergent conformations of the helix B'-C loop that are stabilized by different hydrogen-bonding interactions in the two enzymes. Additionally, differences between the two enzymes for other amino acids that line the edges of the cavity contribute to distinct orientations of ANF in the two active sites. Thus, the narrow cavity is conserved in both P450 subfamily 1A and P450 subfamily 1B with sequence divergence around the edges of the cavity that modify substrate and inhibitor binding. The conservation of these P450 1B1 active site amino acid residues across vertebrate species suggests that these structural features are conserved.


Subject(s)
Aryl Hydrocarbon Hydroxylases/chemistry , Benzoflavones/chemistry , Cytochrome P-450 Enzyme System/chemistry , Aryl Hydrocarbon Hydroxylases/metabolism , Benzoflavones/metabolism , Catalytic Domain , Crystallography, X-Ray , Cytochrome P-450 CYP1B1 , Cytochrome P-450 Enzyme System/metabolism , Humans , Protein Binding , Protein Structure, Secondary
7.
J Biol Chem ; 284(24): 16541-16552, 2009 Jun 12.
Article in English | MEDLINE | ID: mdl-19366684

ABSTRACT

CYP4A11 transgenic mice (CYP4A11 Tg) were generated to examine in vivo regulation of the human CYP4A11 gene. Expression of CYP4A11 in mice yields liver and kidney P450 4A11 levels similar to those found in the corresponding human tissues and leads to an increased microsomal capacity for omega-hydroxylation of lauric acid. Fasted CYP4A11 Tg mice exhibit 2-3-fold increases in hepatic CYP4A11 mRNA and protein, and this response is absent in peroxisome proliferator-activated receptor alpha (PPARalpha) null mice. Dietary administration of either of the PPARalpha agonists, fenofibrate or clofibric acid, increases hepatic and renal CYP4A11 levels by 2-3-fold, and these responses were also abrogated in PPARalpha null mice. Basal liver CYP4A11 levels are reduced differentially in PPARalpha-/- females (>95%) and males (<50%) compared with PPARalpha-/+ mice. Quantitative and temporal differences in growth hormone secretion are known to alter hepatic lipid metabolism and to underlie sexually dimorphic gene expression, respectively. Continuous infusion of low levels of growth hormone reduced CYP4A11 expression by 50% in PPARalpha-proficient male and female transgenic mice. A larger decrease was observed for the expression of CYP4A11 in PPARalpha-/- CYP4A11 Tg male mice to levels similar to that of female PPARalpha-deficient mice. These results suggest that PPARalpha contributes to the maintenance of basal CYP4A11 expression and mediates CYP4A11 induction in response to fibrates or fasting. In contrast, increased exposure to growth hormone down-regulates CYP4A11 expression in liver.


Subject(s)
Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Growth Hormone/metabolism , Liver/enzymology , PPAR alpha/metabolism , Animals , Clofibric Acid/pharmacology , Cytochrome P-450 CYP4A , Fasting/physiology , Female , Fenofibrate/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/physiology , Growth Hormone/pharmacology , Humans , Hypolipidemic Agents/pharmacology , Kidney/enzymology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microsomes, Liver/enzymology , PPAR alpha/agonists , Pregnancy , RNA, Messenger/metabolism , Sex Characteristics
8.
Drug Metab Rev ; 39(2-3): 515-38, 2007.
Article in English | MEDLINE | ID: mdl-17786636

ABSTRACT

The microsomal cytochrome P450 (CYP) family 4 monooxygenases are the major fatty acid omega-hydroxylases. These enzymes remove excess free fatty acids to prevent lipotoxicity, catabolize leukotrienes and prostanoids, and also produce bioactive metabolites from arachidonic acid omega-hydroxylation. In addition to endogenous substrates, recent evidence indicates that CYP4 monooxygenases can also metabolize xenobiotics, including therapeutic drugs. This review focuses on human CYP4 enzymes and updates current knowledge concerning catalytic activity profiles, genetic variation and regulation of expression. Comparative differences between the human and rodent CYP4 enzymes regarding catalytic function and conditional expression are also discussed.


Subject(s)
Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/physiology , Gene Expression Regulation, Enzymologic/physiology , Animals , Cytochrome P-450 Enzyme System/biosynthesis , Genetic Variation , Humans , Isoenzymes/biosynthesis , Isoenzymes/genetics , Isoenzymes/physiology , Rodentia
9.
J Biol Chem ; 282(8): 5225-36, 2007 Feb 23.
Article in English | MEDLINE | ID: mdl-17142457

ABSTRACT

This report provides the first evidence that human P450 4F2 (CYP4F2) is induced by statins, which are widely used to treat hypercholesterolemia. Real time PCR and immunoblots indicate that lovastatin treatment increases expression of the endogenous CYP4F2 gene in human primary hepatocytes and HepG2 cells. The effects of lovastatin on gene expression are often mediated through sterol regulatory element-binding proteins (SREBPs). Immunoblots indicate that lovastatin-treated human hepatocytes display increased proteolytic processing of SREBP-2. In HepG2 cells, co-administration of a potent suppressor of SREBP-2 activation, 25-hydroxycholesterol, inhibits CYP4F2 mRNA induction by lovastatin. HepG2 cells transfected with an expression vector for the active nuclear form of SREBP-1a (nSREBP-1a) also display elevated endogenous CYP4F2 expression. Luciferase reporters containing the CYP4F2 proximal promoter are transactivated by nSREBPs (-1a, -1c, and -2) or a dominant positive form of the SREBP cleavage-activating protein (SCAP), which facilitates activation of endogenous SREBPs. Lovastatin-induced reporter expression is inhibited by overexpressed Insig-1, which prevents proteolytic activation of endogenous SREBPs. Electrophoretic mobility shift assays with in vitro translated nSREBP-1a identified two SREBP binding sites at -169/-152 and -109/-92, relative to the CYP4F2 transcription start site. Mutations in each site abolish SREBP binding. Chromatin immunoprecipitation experiments indicate that more SREBP-1 is associated with the CYP4F2 promoter after overexpression of nSREBP-1a. Transfection studies and mutagenesis indicate that the -109/-92 region is the primary site responsible for the effects of statins. Collectively, these results demonstrate that SREBPs transactivate CYP4F2 transcription and that CYP4F2 induction by statins is mediated by SREBP-2.


Subject(s)
Anticholesteremic Agents/pharmacology , Cytochrome P-450 Enzyme System/biosynthesis , Gene Expression Regulation, Enzymologic/drug effects , Hepatocytes/enzymology , Lovastatin/pharmacology , Sterol Regulatory Element Binding Protein 2/metabolism , Anticholesteremic Agents/therapeutic use , Cell Line, Tumor , Cytochrome P-450 Enzyme System/genetics , Cytochrome P450 Family 4 , Gene Expression Regulation, Enzymologic/genetics , Humans , Hydroxycholesterols/pharmacology , Hypercholesterolemia/drug therapy , Hypercholesterolemia/enzymology , Hypercholesterolemia/genetics , Intracellular Signaling Peptides and Proteins , Lovastatin/therapeutic use , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mutation , Response Elements/genetics , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , Transcriptional Activation/drug effects , Transcriptional Activation/genetics
10.
Arch Biochem Biophys ; 436(2): 377-85, 2005 Apr 15.
Article in English | MEDLINE | ID: mdl-15797250

ABSTRACT

Cytochrome P450 genes (CYPs) encoding two new subfamilies designated CYP4X1 and CYP4Z1 were identified in the human genome and the Expressed Sequence Tags database. Partial cDNAs encoding both P450s were isolated from human kidney and used to determine tissue distribution. CYP4X1 was predominantly expressed in trachea and aorta, whereas CYP4Z1 mRNA was preferentially expressed in mammary tissue. In T47-D cells, CYP4Z1 mRNA levels were induced by dexamethasone (14-fold) or by progesterone (10-fold). The induction by these compounds was suppressed by co-treatment with the progesterone and glucocorticoid receptor antagonist mifepristone (RU486). In the progesterone receptor negative MCF-7 cells, CYP4Z1 mRNA was induced by dexamethasone but not by progesterone treatment. CYP4Z1 mRNA levels were unaffected by 17beta-estradiol. In confluent cultures of human hepatoma HepG2 cells that stably express a mouse peroxisome proliferator activated receptor-alpha (PPARalpha) mutant, CYP4X1 mRNA was undetectable in vehicle-treated cells but was readily detectable following addition of the PPARalpha agonist Wy14643. This suggests that PPARalpha activation can affect human CYP4X1 gene transcription. These results demonstrate selective tissue expression and implicate PPARalpha in CYP4X1 regulation, and the glucocorticoid and progesterone receptors in CYP4Z1 gene activation.


Subject(s)
Cytochrome P-450 Enzyme System/chemistry , Gene Expression Regulation , Isoenzymes/chemistry , Animals , Aorta/metabolism , Breast/metabolism , COS Cells , Cell Line, Tumor , Cloning, Molecular , Cytochrome P450 Family 4 , DNA Restriction Enzymes/metabolism , DNA, Complementary/metabolism , Databases as Topic , Dexamethasone/pharmacology , Expressed Sequence Tags , Humans , Immunoblotting , Mifepristone/pharmacology , Polymerase Chain Reaction , Progesterone/metabolism , Pyrimidines/pharmacology , RNA/metabolism , RNA, Messenger/metabolism , Receptors, Glucocorticoid/metabolism , Receptors, Progesterone/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tissue Distribution , Trachea/metabolism , Transcription, Genetic , Transcriptional Activation
11.
Arch Biochem Biophys ; 409(1): 212-20, 2003 Jan 01.
Article in English | MEDLINE | ID: mdl-12464261

ABSTRACT

HepG2 cells that stably overexpress PPARalpha were used to examine the regulation of the two known human CYP4A genes by Wy14643. Specific PCR amplification across intron 5 and restriction endonuclease analysis indicated that HepG2 cells possess genes corresponding to both the CYP4A11 cDNA and a more recently characterized gene, CYP4A22, that exhibits 95% identity to CYP4A11 in the coding region. These are unlikely to represent alleles because both genes were present in DNA samples from 100 of 100 individuals. Quantitative real-time PCR determined that CYP4A22 mRNA is expressed at significantly lower levels than CYP4A11 mRNA in human liver samples. The PPARalpha agonist Wy14643 induced CYP4A11 mRNA in confluent cultures of HepG2 cells stably expressing the murine PPARalpha-E282G mutant. This mutant exhibits a significantly decreased ligand-independent trans-activation and can be activated by Wy14643 to a level similar to that of wild-type PPARalpha. Dexamethasone induced CYP4A11 mRNA in both control and PPARalpha- E282G-expressing HepG2 cells, indicating that the induction of CYP4A11 by dexamethasone is independent of elevated PPARalpha expression. Wy14643 or dexamethasone induction of CYP4A22 mRNA was not evident in either control or PPARalpha -E282G-expressing HepG2 cells. The results indicate that CYP4A11 expression can be induced by glucocorticoids and peroxisome proliferators.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Dexamethasone/pharmacology , Gene Expression Regulation, Enzymologic , Mixed Function Oxygenases/genetics , Mixed Function Oxygenases/metabolism , Animals , Base Sequence , Cell Line , Cytochrome P-450 CYP4A , DNA, Complementary/metabolism , Glucocorticoids/pharmacology , Humans , Introns , Mice , Models, Genetic , Molecular Sequence Data , Mutation , Peroxisome Proliferators/pharmacology , Pyrimidines/pharmacology , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction
12.
Toxicology ; 181-182: 203-6, 2002 Dec 27.
Article in English | MEDLINE | ID: mdl-12505311

ABSTRACT

The induction of P450 4A enzymes by peroxisome proliferators (PPs) and fatty acids is mediated by the peroxisome proliferator activated receptor alpha (PPAR alpha) that binds to response elements in target genes as a heterodimer with the retinoid X receptor (RXR). The consensus sequence recognized by PPAR/RXR heterodimers, contains an imperfect direct repeat of two nuclear receptor binding motifs separated by a single nucleotide. This repeat is preceded by a conserved A/T rich sequence that is required for function. In mice, chronic exposure to PPs results in PPAR alpha mediated liver hypertrophy, hyperplasia and carcinogenesis accompanied by a proliferation of peroxisomes. In contrast, humans exhibit a reduced sensitivity to PP pathogenesis. This could reflect >10-fold lower PPAR alpha levels relative to mice as well as differences in targeted genes. In order to identify PPAR responsive human genes, the human hepatoma cell line, HepG2, was engineered to express increased levels of PPAR alpha. Several genes encoding rate-limiting enzymes and branch points in ketone body formation are regulated by PPAR alpha in these cells. In contrast, significant induction by PP is not evident for peroxisomal fatty acid oxidation that is associated with peroxisome proliferation in mice. Human P450 4A11 is not expressed in dividing cultures of cells with enhanced PPAR alpha levels, but it is expressed in confluent cultures expressing elevated amounts of PPAR alpha.


Subject(s)
Cytochrome P-450 Enzyme System/biosynthesis , Gene Expression Regulation, Enzymologic/genetics , Mixed Function Oxygenases/biosynthesis , Peroxisome Proliferators/pharmacology , Receptors, Drug/drug effects , Animals , Cytochrome P-450 CYP4A , Cytochrome P-450 Enzyme System/genetics , Fatty Acids/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Humans , Mixed Function Oxygenases/genetics , Receptors, Cytoplasmic and Nuclear/drug effects , Receptors, Cytoplasmic and Nuclear/genetics , Species Specificity , Transcription Factors/drug effects , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...