150 results on '"El-Kadi AO"'
Search Results
2. Pharmacokinetics of metformin in the rat: assessment of the effect of hyperlipidemia and evidence for its metabolism to guanylurea.
- Author
-
Gabr RQ, El-Sherbeni AA, Ben-Eltriki M, El-Kadi AO, and Brocks DR
- Subjects
- Animals, Antiporters metabolism, Catecholamine Plasma Membrane Transport Proteins metabolism, Hepatocytes drug effects, Hepatocytes metabolism, Kidney drug effects, Kidney metabolism, Male, Metformin metabolism, Metformin pharmacology, Organic Cation Transport Proteins metabolism, Organic Cation Transporter 2 metabolism, Rats, Rats, Sprague-Dawley, Urea metabolism, Guanidines metabolism, Hyperlipidemias metabolism, Metformin pharmacokinetics, Urea analogs & derivatives
- Abstract
Metformin pharmacokinetics are highly dependent upon organic cationic transporters. There is evidence of a change in its renal clearance in hyperlipidemic obese patients, and no information on its metabolic fate. To study some of these aspects, the influence of poloxamer 407 (P407)-induced hyperlipidemia on metformin pharmacokinetics was assessed. Control and P407-treated adult male rats were administered 30 mg/kg metformin intravenously (i.v.). The pharmacokinetic assessments were performed at 2 time points, 36 and 108 h, following the intraperitoneal dose of P407 (1 g/kg). mRNA and protein expressions of cationic drug transporters were also measured. There was no evidence of a change in metformin pharmacokinetics after i.v. doses as a consequence of short-term hyperlipidemia, and a change in transporter mRNA but not protein expression was observed in the P407- treated rats 108 h after P407 injection. Urinary recovery of unchanged drug was high (>90%) but incomplete. Presumed metabolite peaks were detected in chromatograms of hepatocytes and microsomal protein spiked with metformin. Comparative chromatographic elution times and mass spectra suggested that one of the predominant metabolites was guanylurea. Hyperlipidemia by itself did not affect the pharmacokinetics of metformin. Guanylurea is a putative metabolite of metformin in rats.
- Published
- 2017
- Full Text
- View/download PDF
3. The role of cytochrome P450 1B1 and its associated mid-chain hydroxyeicosatetraenoic acid metabolites in the development of cardiac hypertrophy induced by isoproterenol.
- Author
-
Maayah ZH, Althurwi HN, El-Sherbeni AA, Abdelhamid G, Siraki AG, and El-Kadi AO
- Subjects
- Animals, Cardiomegaly chemically induced, Cardiomegaly genetics, Cell Line, Cytochrome P-450 CYP1B1 genetics, Gene Expression Regulation, Enzymologic drug effects, Humans, Male, Myocytes, Cardiac cytology, Myocytes, Cardiac drug effects, Myocytes, Cardiac metabolism, Rats, Rats, Sprague-Dawley, Cardiomegaly metabolism, Cytochrome P-450 CYP1B1 metabolism, Hydroxyeicosatetraenoic Acids metabolism, Isoproterenol adverse effects
- Abstract
Numerous experimental studies have demonstrated the role of cytochrome P450 1B1 (CYP1B1) and its associated mid-chain hydroxyeicosatetraenoic acids (mid-chain HETEs) metabolite in the pathogenesis of cardiac hypertrophy. However, the ability of isoproterenol (ISO) to induce cardiac hypertrophy through mid-chain HETEs has not been investigated yet. Therefore, we hypothesized that ISO induces cardiac hypertrophy through the induction of CYP1B1 and its associated mid-chain HETE metabolites. To test our hypothesis, Sprague-Dawley rats were treated with ISO (5 mg/kg i.p.) for 12 and 72 h whereas, human ventricular cardiomyocytes RL-14 cells were exposed to 100 μM ISO in the presence and absence of 0.5 μM tetramethoxystilbene (TMS) a selective CYP1B1 inhibitor, or 25 nM CYP1B1-siRNA. Moreover, RL-14 cells were transiently transfected with the CRISPR-CYP1B1 plasmid. Thereafter, real-time PCR, western blot analysis, and liquid chromatography-electrospray ionization mass spectroscopy were used to determine the level of gene expression, protein expression, and mid-chain HETEs, respectively. Our results showed that ISO induced CYP1B1 protein expression and the level of cardiac mid-chain HETEs in vivo at pre-hypertrophic and hypertrophic stage. In vitro, inhibition of CYP1B1 using TMS or CYP1B1-siRNA significantly attenuates ISO-induced hypertrophy. Furthermore, overexpression of CYP1B1 significantly induced cellular hypertrophy and mid-chain HETEs metabolite. Mechanistically, the protective effect of TMS against cardiac hypertrophy was mediated through the modulation of superoxide anion, mitogen-activated protein kinases (MAPKs), and nuclear factor-κB (NF-κB). In conclusion, our study provides the first evidence that CYP1B1 and its associated mid-chain HETE metabolites are directly involved in the ISO-induced cardiac hypertrophy.
- Published
- 2017
- Full Text
- View/download PDF
4. Down-regulation of cytochrome P450 1A1 by monomethylarsonous acid in human HepG2 cells.
- Author
-
Elshenawy OH, Abdelhamid G, Soshilov AA, Denison MS, and El-Kadi AO
- Subjects
- Arsenites toxicity, Basic Helix-Loop-Helix Transcription Factors genetics, Basic Helix-Loop-Helix Transcription Factors metabolism, Cell Survival drug effects, Cytochrome P-450 CYP1A1 genetics, Heme Oxygenase-1 genetics, Heme Oxygenase-1 metabolism, Hep G2 Cells, Humans, Oxidative Stress drug effects, Polychlorinated Dibenzodioxins toxicity, Protein Processing, Post-Translational, Protein Stability drug effects, RNA, Messenger genetics, RNA, Messenger metabolism, Reactive Oxygen Species metabolism, Receptors, Aryl Hydrocarbon genetics, Receptors, Aryl Hydrocarbon metabolism, Signal Transduction, Sodium Compounds toxicity, Cytochrome P-450 CYP1A1 metabolism, Down-Regulation, Gene Expression Regulation, Enzymologic, Organometallic Compounds toxicity
- Abstract
Inorganic arsenic is a human toxicant and carcinogen that has been extensively studied over decades; however, no definitive understanding of the underlying mechanisms has been established yet. Arsenic is capable of modulating the expression of aryl hydrocarbon receptor (AhR)-regulated genes, nevertheless, whether its trivalent organic metabolites have similar effects or not need to be investigated. Therefore, in this study we examined the effects of monomethylarsonous acid (MMA(III)) as compared to its parent compound sodium arsenite (As(III)) on the expression of CYP1A1 in HepG2 cells. HepG2 cells were treated with MMA(III) (5μM) or its parents compound, As(III) (5μM), in the absence and presence of the prototypical AhR ligand, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD; 1nM). Experiments were conducted at 6h for gene expression; 24h for XRE-driven luciferase activity, protein expression, and EROD activity. Our results showed that both MMA(III) and As(III) decreased CYP1A1 mRNA, protein, and catalytic activity levels; and inhibit the TCDD-mediated induction of CYP1A1 mRNA, protein, and catalytic activity levels. MMA(III) and As(III) significantly inhibited XRE-driven luciferase activity and it inhibited the TCDD-mediated induction of XRE-driven luciferase reporter gene expression. Although MMA(III) and As(III) were not shown to be AhR ligands, both compounds showed inhibition of nuclear accumulation of AhR transcription factor as evidenced by immunocytochemical analysis. MMA(III) and As(III) had no effect on CYP1A1 mRNA stability; however MMA(III), but not As(III), decreased the protein stability of CYP1A1. As(III), but not MMA(III), induced HO-1 mRNA levels. Both MMA(III) and As(III) increased ROS production. Our results demonstrate for the first time that, MMA(III) down-regulates CYP1A1 mainly through transcriptional and post-translational mechanisms. This modulation of CYP1A1 proves that trivalent metabolites of arsenic are highly reactive and could participate in arsenic toxicity., (Copyright © 2017 Elsevier B.V. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
5. Clinical Implications of 20-Hydroxyeicosatetraenoic Acid in the Kidney, Liver, Lung and Brain: An Emerging Therapeutic Target.
- Author
-
Elshenawy OH, Shoieb SM, Mohamed A, and El-Kadi AO
- Abstract
Cytochrome P450-mediated metabolism of arachidonic acid (AA) is an important pathway for the formation of eicosanoids. The ω-hydroxylation of AA generates significant levels of 20-hydroxyeicosatetraenoic acid (20-HETE) in various tissues. In the current review, we discussed the role of 20-HETE in the kidney, liver, lung, and brain during physiological and pathophysiological states. Moreover, we discussed the role of 20-HETE in tumor formation, metabolic syndrome and diabetes. In the kidney, 20-HETE is involved in modulation of preglomerular vascular tone and tubular ion transport. Furthermore, 20-HETE is involved in renal ischemia/reperfusion (I/R) injury and polycystic kidney diseases. The role of 20-HETE in the liver is not clearly understood although it represents 50%-75% of liver CYP-dependent AA metabolism, and it is associated with liver cirrhotic ascites. In the respiratory system, 20-HETE plays a role in pulmonary cell survival, pulmonary vascular tone and tone of the airways. As for the brain, 20-HETE is involved in cerebral I/R injury. Moreover, 20-HETE has angiogenic and mitogenic properties and thus helps in tumor promotion. Several inhibitors and inducers of the synthesis of 20-HETE as well as 20-HETE analogues and antagonists are recently available and could be promising therapeutic options for the treatment of many disease states in the future.
- Published
- 2017
- Full Text
- View/download PDF
6. Microsomal cytochrome P450 as a target for drug discovery and repurposing.
- Author
-
El-Sherbeni AA and El-Kadi AO
- Subjects
- Arachidonic Acid metabolism, Arachidonic Acid physiology, Cytochrome P-450 Enzyme Inducers pharmacology, Cytochrome P-450 Enzyme Inhibitors pharmacology, Cytochrome P-450 Enzyme System metabolism, Cytochrome P-450 Enzyme System physiology, Drug Interactions, Epoxide Hydrolases antagonists & inhibitors, Humans, Cytochrome P-450 Enzyme System drug effects, Drug Discovery, Drug Repositioning, Microsomes enzymology
- Abstract
Cytochrome P450 (P450) enzymes are ancient electron-transfer-chain system of remarkable biological importance. Microsomal P450 enzymes are the P450 attached to endoplasmic reticulum, which, in humans, are critical for body's defenses against xenobiotics by mediating their metabolism, and cell signaling by mediating arachidonic acid (AA) transformation to several potent bioactive molecules. Only recently, modulating P450-mediated AA metabolism has risen as a promising new drug target. This review presents the therapeutic potential of finding effective, selective and safe treatments targeting P450-mediated AA metabolism, and the several approaches that have been used to find these treatments; among which, our focus was on modulators of P450 activities. We detailed the efforts done to develop new molecular entities designed to modulate P450, and the more recent efforts tried to employ our previous knowledge on drug metabolism to repurpose old drugs with the capacity to alter P450-mediated drug metabolism to target AA metabolism. Because of the long recognition of P450 role in xenobiotic metabolism, several clinically approved agents were identified to alter P450 activity. Repurposing old drugs as P450 modulators can facilitate bringing treatments targeting P450-mediated AA metabolism to clinical trials. However, the capacity of the modulation of P450-derived AA metabolites of clinically approved drugs has to be systematically investigated and validated for their new use in humans.
- Published
- 2017
- Full Text
- View/download PDF
7. Repurposing Resveratrol and Fluconazole To Modulate Human Cytochrome P450-Mediated Arachidonic Acid Metabolism.
- Author
-
El-Sherbeni AA and El-Kadi AO
- Subjects
- Chromatography, Liquid, Drug Repositioning methods, Humans, Microsomes, Liver metabolism, Resveratrol, Spectrometry, Mass, Electrospray Ionization, Arachidonic Acid chemistry, Arachidonic Acid metabolism, Cytochrome P-450 Enzyme System metabolism, Fluconazole chemistry, Fluconazole metabolism, Stilbenes chemistry, Stilbenes metabolism
- Abstract
Cytochrome P450 (P450) enzymes metabolize arachidonic acid (AA) to several biologically active epoxyeicosatrienoic acids (EETs) and hydroxyeicosatetraenoic acids (HETEs). Repurposing clinically-approved drugs could provide safe and readily available means to control EETs and HETEs levels in humans. Our aim was to determine how to significantly and selectively modulate P450-AA metabolism in humans by clinically-approved drugs. Liquid chromatography-mass spectrometry was used to determine the formation of 15 AA metabolites by human recombinant P450 enzymes, as well as human liver and kidney microsomes. CYP2C19 showed the highest EET-forming activity, while CYP1B1 and CYP2C8 showed the highest midchain HETE-forming activities. CYP1A1 and CYP4 showed the highest subterminal- and 20-HETE-forming activity, respectively. Resveratrol and fluconazole produced the most selective and significant modulation of hepatic P450-AA metabolism, comparable to investigational agents. Monte Carlo simulations showed that 90% of human population would experience a decrease by 6-22%, 16-39%, and 16-35% in 16-, 18-, and 20-HETE formation, respectively, after 2.5 g daily of resveratrol, and by 22-31% and 14-23% in 8,9- and 14,15-EET formation after 50 mg of fluconazole. In conclusion, clinically-approved drugs can provide selective and effective means to modulate P450-AA metabolism, comparable to investigational drugs. Resveratrol and fluconazole are good candidates to be repurposed as new P450-based treatments.
- Published
- 2016
- Full Text
- View/download PDF
8. CYP1B1 inhibition attenuates doxorubicin-induced cardiotoxicity through a mid-chain HETEs-dependent mechanism.
- Author
-
Maayah ZH, Althurwi HN, Abdelhamid G, Lesyk G, Jurasz P, and El-Kadi AO
- Subjects
- Animals, Cardiotoxicity metabolism, Cardiotoxicity pathology, Cell Line, Cytochrome P-450 CYP1B1 metabolism, Humans, Male, Myocardium metabolism, Myocardium pathology, Rats, Sprague-Dawley, Antibiotics, Antineoplastic toxicity, Cardiotoxicity drug therapy, Cytochrome P-450 CYP1B1 antagonists & inhibitors, Doxorubicin toxicity, Enzyme Inhibitors therapeutic use, Hydroxyeicosatetraenoic Acids metabolism, Stilbenes therapeutic use
- Abstract
Doxorubicin (DOX) has been reported to be a very potent and effective anticancer agent. However, clinical treatment with DOX has been greatly limited due to its cardiotoxicity. Furthermore, several studies have suggested a role for cytochrome P450 1B1 (CYP1B1) and mid-chain hydroxyeicosatetraenoic acids (mid-chain HETEs) in DOX-induced cardiac toxicity. Therefore, we hypothesized that DOX induced cardiotoxicity is mediated through the induction of CYP1B1 and its associated mid-chain HETEs metabolite. To test our hypothesis, Sprague-Dawley rats and RL-14 cells were treated with DOX in the presence and absence of 2,3',4,5'-tetramethoxystilbene (TMS), a selective CYP1B1 inhibitor. Thereafter, cardiotoxicity parameters were determined using echocardiography, histopathology, and gene expression. Further, the level of mid-chain HETEs was quantified using liquid chromatography-electron spray ionization-mass spectrometry. Our results showed that DOX induced cardiotoxicity in vivo and in vitro as evidenced by deleterious changes in echocardiography, histopathology, and hypertrophic markers. Importantly, the TMS significantly reversed these changes. Moreover, the DOX-induced cardiotoxicity was associated with a proportional increase in the formation of cardiac mid-chain HETEs both in vivo and in our cell culture model. Interestingly, the inhibition of cardiotoxicity by TMS was associated with a dramatic decrease in the formation of cardiac mid-chain HETEs suggesting a mid-chain HETEs-dependent mechanism. Mechanistically, the protective effect of TMS against DOX-induced cardiotoxicity was mediated through the inhibition of mitogen activated protein kinases (MAPKs) and nuclear factor-κB (NF-κB). In conclusion, our study provides the first evidence that the inhibition of CYP1B1 and mid-chain HETE formation attenuate DOX-induced cardiotoxicity., (Copyright © 2016 Elsevier Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
9. Ketoconazole Stereoisomers Differentially Induce Cytochrome P450 1A1 Between Human Hepatoma HepG2 and Mouse Hepatoma Hepa1c1c7 Cells.
- Author
-
Anwar-Mohamed A, El-Sherbeni AA, Hamdy DA, Korashy HM, Brocks DR, and El-Kadi AO
- Subjects
- Animals, Cell Line, Tumor, Hep G2 Cells, Humans, Liver Neoplasms drug therapy, Liver Neoplasms metabolism, Mice, RNA, Messenger metabolism, Stereoisomerism, Antifungal Agents pharmacology, Carcinoma, Hepatocellular drug therapy, Carcinoma, Hepatocellular metabolism, Cytochrome P-450 CYP1A1 metabolism, Ketoconazole pharmacology
- Abstract
Ketoconazole (KTZ) has 2 chiral centers with the therapeutically active form being a racemic mixture of 2 cis-enantiomers, namely, (2R,4S)-(+)-KTZ and (2S,4R)-(-)-KTZ. The aims of the present study were to examine the effects of (+)-KTZ, (-)-KTZ, and (±)-KTZ on aryl hydrocarbon receptor activation and subsequently CYP1A1 induction in both human HepG2 and murine Hepa1c1c7 hepatoma cells, and to further test their inhibitory effect using recombinant human and mouse CYP1A1 enzymes. Our results demonstrated that (+)-KTZ induced human CYP1A1 more than (-)-KTZ, whereas on the other hand (-)-KTZ induced murine Cyp1a1 more than (+)-KTZ at the mRNA, and activity levels. Human CYP1A1 showed higher affinity to 7ER compared with murine Cyp1a1 (Km values 13.29 nM for human vs. 168.1 nM for murine). The intrinsic clearance values for human and murine CYP1A1 were 194.1 and 87.6 μL/pmol P450/min, respectively, whereas, Vmax values were 2.58 and 14.73 pmol/pmol P450/min, respectively. (+)-KTZ and (-)-KTZ directly inhibited CYP1A1 activity by noncompetitive mechanism. The affinity of (-)-KTZ to interact with human CYP1A1 and murine Cyp1a1 was significantly different from (+)-KTZ, as the Ki values for human CYP1A1 and murine Cyp1a1 were 199.4 and 413.7 nM, respectively, for (+)-KTZ, and 269.3 and 230.8 nM, respectively, for (-)-KTZ., (Copyright © 2016 American Pharmacists Association®. Published by Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
10. 5-, 12- and 15-Hydroxyeicosatetraenoic acids induce cellular hypertrophy in the human ventricular cardiomyocyte, RL-14 cell line, through MAPK- and NF-κB-dependent mechanism.
- Author
-
Maayah ZH and El-Kadi AO
- Subjects
- Arachidonic Acid metabolism, Atrial Natriuretic Factor genetics, Cardiomegaly pathology, Cell Line, Cell Size drug effects, Cell Survival drug effects, Dose-Response Relationship, Drug, Heart Ventricles cytology, Humans, Mitogen-Activated Protein Kinases metabolism, Myocytes, Cardiac metabolism, NF-kappa B metabolism, Natriuretic Peptide, Brain genetics, 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid adverse effects, Cardiomegaly chemically induced, Hydroxyeicosatetraenoic Acids adverse effects, Myocytes, Cardiac drug effects, Myocytes, Cardiac pathology
- Abstract
Recent studies have established the role of mid-chain hydroxyeicosatetraenoic acids (HETEs) in the development of cardiovascular disease. Mid-chain HETEs have been reported to have vasoconstrictive and pro-inflammatory effects. However, whether mid-chain HETEs can induce cardiac hypertrophy remains unclear. Therefore, the overall objective of the present study was to elucidate the potential hypertrophic effect of mid-chain HETEs in the human ventricular cardiomyocytes, RL-14 cells, and to explore the mechanisms involved. For this purpose, RL-14 cells were treated with increasing concentrations of mid-chain HETEs (2.5, 5, 10 and 20 µM). Thereafter, the cardiac hypertrophy markers and cell size were determined using real-time polymerase chain reaction and phase contrast imaging, respectively. Phosphorylated mitogen-activated protein kinase (MAPK) level and nuclear factor kappa B (NF-κB) binding activity were determined. Our results showed that mid-chain HETEs induced cellular hypertrophy in RL-14 cells as evidenced by the induction of cardiac hypertrophy markers, α- and β-myocin heavy chain and atrial and brain natriuretic peptide as well as the increase in cell size. Mechanistically, all mid-chain HETEs were able to induce the binding activity of NF-κB to its responsive element in a HETE-dependent manner, and they significantly induced the phosphorylation of ERK 1/2. The induction of cellular hypertrophy was associated with proportional increase in the formation of dihydroxyeicosatrienoic acids parallel to the increase of soluble epoxide hydrolase enzyme activity. In conclusion, our study provides the first evidence that mid-chain HETEs induce cellular hypertrophy in RL-14 cells through MAPK- and NF-κB-dependent mechanism.
- Published
- 2016
- Full Text
- View/download PDF
11. The role of mid-chain hydroxyeicosatetraenoic acids in the pathogenesis of hypertension and cardiac hypertrophy.
- Author
-
Maayah ZH and El-Kadi AO
- Subjects
- Animals, Cardiomegaly drug therapy, Cardiomegaly epidemiology, Cardiomegaly physiopathology, Cardiovascular Agents therapeutic use, Cardiovascular System drug effects, Cardiovascular System physiopathology, Cytochrome P-450 CYP1B1 antagonists & inhibitors, Cytochrome P-450 CYP1B1 metabolism, Cytochrome P-450 Enzyme Inhibitors therapeutic use, Drug Design, Humans, Hypertension drug therapy, Hypertension epidemiology, Hypertension physiopathology, Lipoxygenase metabolism, Lipoxygenase Inhibitors therapeutic use, Molecular Targeted Therapy, Cardiomegaly metabolism, Cardiovascular System metabolism, Hydroxyeicosatetraenoic Acids metabolism, Hypertension metabolism, Signal Transduction drug effects
- Abstract
The incidence, prevalence, and hospitalization rates associated with cardiovascular diseases (CVDs) are projected to increase substantially in the world. Understanding of the biological and pathophysiological mechanisms of survival can help the researchers to develop new management modalities. Numerous experimental studies have demonstrated that mid-chain HETEs are strongly involved in the pathogenesis of the CVDs. Mid-chain HETEs are biologically active eicosanoids that result from the metabolism of arachidonic acid (AA) by both lipoxygenase and CYP1B1 (lipoxygenase-like reaction). Therefore, identifying the localizations and expressions of the lipoxygenase and CYP1B1 and their associated AA metabolites in the cardiovascular system is of major importance in understanding their pathological roles. Generally, the expression of these enzymes is shown to be induced during several CVDs, including hypertension and cardiac hypertrophy. The induction of these enzymes is associated with the generation of mid-chain HETEs and subsequently causation of cardiovascular events. Of interest, inhibiting the formation of mid-chain HETEs has been reported to confer a protection against different cardiac hypertrophy and hypertension models such as angiotensin II, Goldblatt, spontaneously hypertensive rat and deoxycorticosterone acetate (DOCA)-salt-induced models. Although the exact mechanisms of mid-chain HETEs-mediated cardiovascular dysfunction are not fully understood, the present review proposes several mechanisms which include activating G-protein-coupled receptor, protein kinase C, mitogen-activated protein kinases, and nuclear factor kappa B. This review provides a clear understanding of the role of mid-chain HETEs in the pathogenesis of cardiovascular diseases and their importance as novel targets in the treatment for hypertension and cardiac hypertrophy.
- Published
- 2016
- Full Text
- View/download PDF
12. 19-Hydroxyeicosatetraenoic acid and isoniazid protect against angiotensin II-induced cardiac hypertrophy.
- Author
-
Elkhatali S, El-Sherbeni AA, Elshenawy OH, Abdelhamid G, and El-Kadi AO
- Subjects
- Animals, Arachidonic Acid metabolism, Cardiomegaly metabolism, Cells, Cultured, Cytochrome P-450 Enzyme System metabolism, Humans, Male, Myocytes, Cardiac drug effects, Myocytes, Cardiac metabolism, Rats, Rats, Sprague-Dawley, Angiotensin II pharmacology, Cardiomegaly chemically induced, Cardiomegaly drug therapy, Hydroxyeicosatetraenoic Acids pharmacology, Isoniazid pharmacology, Protective Agents pharmacology
- Abstract
We have recently demonstrated that 19-hydroxyeicosatetraenoic acid (19-HETE) is the major subterminal-HETE formed in the heart tissue, and its formation was decreased during cardiac hypertrophy. In the current study, we examined whether 19-HETE confers cardioprotection against angiotensin II (Ang II)-induced cardiac hypertrophy. The effect of Ang II, with and without 19-HETE (20 μM), on the development of cellular hypertrophy in cardiomyocyte RL-14 cells was assessed by real-time PCR. Also, cardiac hypertrophy was induced in Sprague-Dawley rats by Ang II, and the effect of increasing 19-HETE by isoniazid (INH; 200mg/kg/day) was assessed by heart weight and echocardiography. Also, alterations in cardiac cytochrome P450 (CYP) and their associated arachidonic acid (AA) metabolites were determined by real-time PCR, Western blotting and liquid-chromatography-mass-spectrometry. Our results demonstrated that 19-HETE conferred a cardioprotective effect against Ang II-induced cellular hypertrophy in vitro, as indicated by the significant reduction in β/α-myosin heavy chain ratio. In vivo, INH improved heart dimensions, and reversed the increase in heart weight to tibia length ratio caused by Ang II. We found a significant increase in cardiac 19-HETE, as well as a significant reduction in AA and its metabolite, 20-HETE. In conclusion, 19-HETE, incubated with cardiomyocytes in vitro or induced in the heart by INH in vivo, provides cardioprotection against Ang II-induced hypertrophy. This further confirms the role of CYP, and their associated AA metabolites in the development of cardiac hypertrophy., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
13. Design and synthesis of resveratrol-salicylate hybrid derivatives as CYP1A1 inhibitors.
- Author
-
Aldawsari FS, Elshenawy OH, El Gendy MA, Aguayo-Ortiz R, Baksh S, El-Kadi AO, and Velázquez-Martínez CA
- Subjects
- Cytochrome P-450 CYP1A1 chemistry, Cytochrome P-450 CYP1A1 metabolism, Dose-Response Relationship, Drug, Enzyme Inhibitors chemistry, Humans, Models, Molecular, Molecular Structure, RNA, Messenger antagonists & inhibitors, RNA, Messenger genetics, RNA, Messenger metabolism, Resveratrol, Salicylates chemistry, Stilbenes chemistry, Structure-Activity Relationship, Cytochrome P-450 CYP1A1 antagonists & inhibitors, Drug Design, Enzyme Inhibitors chemical synthesis, Enzyme Inhibitors pharmacology, Salicylates pharmacology, Stilbenes pharmacology
- Abstract
Resveratrol and aspirin are known to exert potential chemopreventive effects through modulation of numerous targets. Considering that the CYP450 system is responsible for the activation of environmental procarcinogens, the aim of this study was to design a new class of hybrid resveratrol-aspirin derivatives possessing the stilbene and the salicylate scaffolds. Using HepG2 cells, we evaluated (a) the inhibition of TCDD-mediated induction of CYP1A1 exerted by resveratrol-aspirin derivatives using the EROD assay, and (b) CYP1A1 mRNA in vitro. We observed significant inhibition (84%) of CYP1A1 activity and a substantial decrease in CYP1A1 mRNA with compound 3, compared to control. Resveratrol did not exert inhibition under the same experimental conditions. This inhibitory profile was supported by docking studies using the crystal structure of human CYP1A1. The potential effect exerted by compound 3 (the most active), provide preliminary evidence supporting the design of hybrid molecules combining the chemical features of resveratrol and aspirin.
- Published
- 2015
- Full Text
- View/download PDF
14. Development of cellular hypertrophy by 8-hydroxyeicosatetraenoic acid in the human ventricular cardiomyocyte, RL-14 cell line, is implicated by MAPK and NF-κB.
- Author
-
Maayah ZH, Abdelhamid G, and El-Kadi AO
- Subjects
- Adult, Animals, Cardiomegaly enzymology, Cardiomegaly pathology, Cell Line, Gene Expression Regulation, Humans, MAP Kinase Signaling System, Myocytes, Cardiac enzymology, Myocytes, Cardiac metabolism, Myocytes, Cardiac pathology, Phosphorylation, Rats, Cardiomegaly chemically induced, Cardiomegaly metabolism, Hydroxyeicosatetraenoic Acids toxicity, Mitogen-Activated Protein Kinases metabolism, Myocytes, Cardiac drug effects, NF-kappa B metabolism
- Abstract
Recent studies have established the role of mid-chain hydroxyeicosatetraenoic acids (mid-chain HETEs) in the development of cardiovascular disease. Among these mid-chains, 8-HETE has been reported to have a proliferator and proinflammatory action. However, whether 8-HETE can induce cardiac hypertrophy has never been investigated before. Therefore, the overall objectives of the present study are to elucidate the potential hypertrophic effect of 8-HETE in the human ventricular cardiomyocytes, RL-14 cells, and to explore the mechanism(s) involved. Our results showed that 8-HETE induced cellular hypertrophy in RL-14 cells as evidenced by the induction of cardiac hypertrophy markers ANP, BNP, α-MHC, and β-MHC in a concentration- and time-dependent manner as well as the increase in cell surface area. Mechanistically, 8-HETE was able to induce the NF-κB activity as well as it significantly induced the phosphorylation of ERK1/2. The induction of cellular hypertrophy was associated with a proportional increase in the formation of dihydroxyeicosatrienoic acids (DHETs) parallel to the increase of soluble epoxide hydrolase (sEH) enzyme activity. Blocking the induction of NF-κB, ERK1/2, and sEH signaling pathways significantly inhibited 8-HETE-induced cellular hypertrophy. Our study provides the first evidence that 8-HETE induces cellular hypertrophy in RL-14 cells through MAPK- and NF-κB-dependent mechanism
- Published
- 2015
- Full Text
- View/download PDF
15. Modulation of aryl hydrocarbon receptor-regulated enzymes by trimethylarsine oxide in C57BL/6 mice: In vivo and in vitro studies.
- Author
-
Elshenawy OH and El-Kadi AO
- Subjects
- Animals, Arsenic Poisoning enzymology, Cell Culture Techniques, Cell Survival drug effects, Cells, Cultured, Cytochrome P-450 Enzyme System biosynthesis, Cytochrome P-450 Enzyme System genetics, Cytosol drug effects, Cytosol enzymology, Enzyme Induction, Glutathione Transferase biosynthesis, Glutathione Transferase genetics, Heme Oxygenase-1 biosynthesis, Heme Oxygenase-1 genetics, Hepatocytes drug effects, Hepatocytes enzymology, Hepatocytes pathology, Isoenzymes biosynthesis, Isoenzymes genetics, Ligands, Liver enzymology, Liver pathology, Male, Membrane Proteins biosynthesis, Membrane Proteins genetics, Mice, Inbred C57BL, Microsomes, Liver drug effects, Microsomes, Liver enzymology, NAD(P)H Dehydrogenase (Quinone) biosynthesis, NAD(P)H Dehydrogenase (Quinone) genetics, Polychlorinated Dibenzodioxins toxicity, Arsenic Poisoning metabolism, Arsenicals, Gene Expression Regulation, Enzymologic drug effects, Liver drug effects, Receptors, Aryl Hydrocarbon metabolism
- Abstract
Arsenic is a worldwide environmental pollutant that is associated with skin and several types of internal cancers. Recent reports revealed that arsenic biomethylation could activate the toxic and carcinogenic potential of arsenic. Therefore, we investigated the effect of trimethylarsine oxide (TMAO) on the activation of AhR-regulated genes in vivo and in vitro. In vivo, C57BL/6 mice received TMAO (13mg/kg i.p.) with or without the prototypical AhR ligand, TCDD (15μg/kg), then the livers were harvested at 6 and 24h post-treatment. In vitro, isolated hepatocytes from C57BL/6 mice were treated with TMAO (5μM) in the absence and presence of TCDD (1nM) for 6 and 24h. Our in vivo results demonstrated that, TMAO alone increased Cyp1a1, Cyp1a2, Cyp1b1, Nqo1, Gsta1, and Ho-1 at mRNA level. Upon co-exposure to TMAO and TCDD, TMAO potentiated the TCDD-mediated induction of Cyp1a1, Cyp1b1, and Nqo1 mRNA levels. Western blotting revealed that, TMAO alone increased Cyp1a1, Cyp1a2, Nqo1, Gsta1/2, and Ho-1 protein levels, and potentiated the TCDD-mediated induction of Cyp1a1 and Cyp1b1 protein level. In addition, TMAO alone significantly increased Cyp1a1, Cyp1a2, Nqo1, Gst, and Ho-1 activities and significantly potentiated the TCDD-mediated induction of Cyp1a1 activity. At the in vitro level, TMAO induced Cyp1a1 and potentiated the TCDD-mediated induction of Cyp1a1 at mRNA, protein and activity levels. In addition, TMAO increased the nuclear localization of AhR and AhR-dependent XRE-driven luciferase activity. Our results demonstrate that the TMAO, modulates AhR-regulated genes which could potentially participate, at least in part, in arsenic induced toxicity and carcinogenicity., (Copyright © 2015 Elsevier Ireland Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
16. Early Changes in Cytochrome P450s and Their Associated Arachidonic Acid Metabolites Play a Crucial Role in the Initiation of Cardiac Hypertrophy Induced by Isoproterenol.
- Author
-
Althurwi HN, Maayah ZH, Elshenawy OH, and El-Kadi AO
- Subjects
- Animals, Cardiomegaly diagnostic imaging, Cell Line, Heart drug effects, Heart Function Tests, Humans, Hydroxyeicosatetraenoic Acids metabolism, Male, Microsomes enzymology, Myocardium metabolism, Myocardium pathology, Myocytes, Cardiac drug effects, Myocytes, Cardiac pathology, Myocytes, Cardiac ultrastructure, NF-kappa B drug effects, NF-kappa B metabolism, RNA, Messenger biosynthesis, RNA, Messenger genetics, Rats, Rats, Sprague-Dawley, Ultrasonography, Adrenergic beta-Antagonists adverse effects, Arachidonic Acids metabolism, Cardiomegaly chemically induced, Cardiomegaly pathology, Cytochrome P-450 Enzyme System metabolism, Isoproterenol adverse effects
- Abstract
Cytochrome P450 enzymes (P450s), along with their cardioprotective metabolites the epoxyeicosatrienoic acids (EETs) and cardiotoxic metabolite 20-hydroxyeicosatetraeonic acid (20-HETE), were found to be altered in cardiac hypertrophy; however, it is unclear whether these changes are causal or epiphenomenon. Therefore, we hypothesized that P450s and their metabolites play a crucial role in the initiation of cardiac hypertrophy. To test our hypothesis, rats and RL-14 cells were treated with the hypertrophic agonist isoproterenol for different time periods. Thereafter, in vivo heart function and wall thickness were assessed using echocardiography. Moreover, the role of P450 epoxygenases, ω-hydroxylases, and soluble epoxide hydrolase (sEH) were determined at mRNA, protein, and activity levels using real-time polymerase chain reaction, Western blot, and liquid chromatography-mass spectrometry, respectively. Our results show that in vivo and in vitro hypertrophy was initiated after 72 hours and 6 hours of isoproterenol treatment, respectively. Studies performed at the prehypertrophy phase showed a significant decrease in P450 epoxygenases along with a significant induction of sEH activity. Consequently, lower EET and higher dihydroxyeicosatrienoic acid levels were observed during this phase. However, significant increases in P450 ω-hydroxylase along with its associated metabolite, 20-HETE, were detected only in vivo. Interestingly, increasing EET levels by P450 epoxygenase induction, sEH inhibition, or exogenous administration of EET prevented the initiation of cardiac hypertrophy through a nuclear factor-κB-mediated mechanism. Taken together, these findings reveal a crucial role of P450 epoxygenases and EETs in the development of cardiac hypertrophy, which could uncover novel targets for prevention of heart failure at early stages., (Copyright © 2015 by The American Society for Pharmacology and Experimental Therapeutics.)
- Published
- 2015
- Full Text
- View/download PDF
17. Correction: the tumor suppressor gene, RASSF1A, is essential for protection against inflammation -induced injury.
- Author
-
Gordon M, El-Kalla M, Zhao Y, Fiteih Y, Law J, Volodko N, Mohamed A, El-Kadi AO, Liu L, Odenbach J, Thiesen A, Onyskiw C, Ghazaleh HA, Park J, Lee SB, Yu VC, Fernandez-Patron C, Alexander RT, Wine E, and Baksh S
- Published
- 2015
- Full Text
- View/download PDF
18. Buthionine sulfoximine, an inhibitor of glutathione biosynthesis, induces expression of soluble epoxide hydrolase and markers of cellular hypertrophy in a rat cardiomyoblast cell line: roles of the NF-κB and MAPK signaling pathways.
- Author
-
Abdelhamid G and El-Kadi AO
- Subjects
- Animals, Antioxidants pharmacology, Atrial Natriuretic Factor biosynthesis, Butadienes pharmacology, Cell Line, Cell Survival, Enzyme Activation, Epoxide Hydrolases genetics, Extracellular Signal-Regulated MAP Kinases metabolism, Flavonoids pharmacology, Gene Expression Regulation physiology, Glutathione biosynthesis, Heart Failure pathology, I-kappa B Proteins biosynthesis, I-kappa B Proteins metabolism, JNK Mitogen-Activated Protein Kinases metabolism, Myoblasts, Cardiac metabolism, NF-KappaB Inhibitor alpha, NF-kappa B p50 Subunit antagonists & inhibitors, NF-kappa B p50 Subunit biosynthesis, Natriuretic Peptide, Brain biosynthesis, Nitriles pharmacology, Oxidative Stress, Phosphorylation, Proline analogs & derivatives, Proline pharmacology, RNA, Messenger biosynthesis, Rats, Thiocarbamates pharmacology, Transcription Factor RelA metabolism, Up-Regulation, p38 Mitogen-Activated Protein Kinases metabolism, Buthionine Sulfoximine pharmacology, Cardiomegaly pathology, Epoxide Hydrolases biosynthesis, MAP Kinase Signaling System physiology, NF-kappa B p50 Subunit metabolism
- Abstract
Evidence suggests that upregulation of soluble epoxide hydrolase (sEH) is associated with the development of myocardial infarction, dilated cardiomyopathy, cardiac hypertrophy, and heart failure. However, the upregulation mechanism is still unknown. In this study, we treated H9C2 cells with buthionine sulfoximine (BSO) to explore whether oxidative stress upregulates sEH gene expression and to identify the molecular and cellular mechanisms behind this upregulatory response. Real-time PCR and Western blot analyses were used to measure mRNA and protein expression, respectively. We demonstrated that BSO significantly upregulated sEH at mRNA levels in a concentration- and time-dependent manner, leading to a significant increase in the cellular hypertrophic markers, atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP). Furthermore, BSO significantly increased the cytosolic phosphorylated IκB-α and translocation of NF-κB p50 subunits, as measured by Western blot analysis. This level of translocation was paralleled by an increase in the DNA-binding activity of NF-κB P50 subunits. Moreover, our results demonstrated that pretreatment with the NF-κB inhibitor PDTC significantly inhibited BSO-mediated induction of sEH and cellular hypertrophic marker gene expression in a dose-dependent manner. Additionally, mitogen-activated protein kinases (MAPKs) were transiently phosphorylated by BSO treatment. To understand further the role of MAPKs pathway in BSO-mediated induction of sEH mRNA, we examined the role of extracellular signal-regulated kinase (ERK), c-JunN-terminal kinase (JNK), and p38 MAPK. Indeed, treatment with the MEK/ERK signal transduction inhibitor, PD98059, partially blocked the activation of IκB-α and translocation of NF-κB p50 subunits induced by BSO. Moreover, pretreatment with MEK/ERK signal transduction inhibitors, PD98059 and U0126, significantly inhibited BSO-mediated induction of sEH and cellular hypertrophic marker gene expression. These results clearly demonstrated that the NF-κB signaling pathway is involved in BSO-mediated induction of sEH gene expression, and appears to be associated with the activation of the MAPK pathway. Furthermore, our findings provide a strong link between sEH-induced cardiac dysfunction and involvement of NF-κB in the development of cellular hypertrophy., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
19. Metformin inhibits 7,12-dimethylbenz[a]anthracene-induced breast carcinogenesis and adduct formation in human breast cells by inhibiting the cytochrome P4501A1/aryl hydrocarbon receptor signaling pathway.
- Author
-
Maayah ZH, Ghebeh H, Alhaider AA, El-Kadi AO, Soshilov AA, Denison MS, Ansari MA, and Korashy HM
- Subjects
- 9,10-Dimethyl-1,2-benzanthracene administration & dosage, 9,10-Dimethyl-1,2-benzanthracene metabolism, Animals, Breast Neoplasms chemically induced, Breast Neoplasms genetics, Breast Neoplasms metabolism, Carcinogenesis drug effects, Carcinogens administration & dosage, Carcinogens metabolism, Cell Line, Tumor, Cytochrome P-450 CYP1A1 metabolism, Female, Guanine analogs & derivatives, Guanine metabolism, Humans, Mice, NAD(P)H Dehydrogenase (Quinone) metabolism, RNA, Messenger metabolism, Receptors, Aryl Hydrocarbon metabolism, Signal Transduction drug effects, 9,10-Dimethyl-1,2-benzanthracene analogs & derivatives, Anticarcinogenic Agents pharmacology, Breast Neoplasms prevention & control, Cytochrome P-450 CYP1A1 antagonists & inhibitors, DNA Adducts biosynthesis, Metformin pharmacology, Receptors, Aryl Hydrocarbon antagonists & inhibitors
- Abstract
Recent studies have established that metformin (MET), an oral anti-diabetic drug, possesses antioxidant activity and is effective against different types of cancer in several carcinogen-induced animal models and cell lines. However, whether MET can protect against breast cancer has not been reported before. Therefore, the overall objectives of the present study are to elucidate the potential chemopreventive effect of MET in non-cancerous human breast MCF10A cells and explore the underlying mechanism involved, specifically the role of cytochrome P4501A1 (CYP1A1)/aryl hydrocarbon receptor (AhR) pathway. Transformation of the MCF10A cells into initiated breast cancer cells with DNA adduct formation was conducted using 7,12-dimethylbenz[a]anthracene (DMBA), an AhR ligand. The chemopreventive effect of MET against DMBA-induced breast carcinogenesis was evidenced by the capability of MET to restore the induction of the mRNA levels of basic excision repair genes, 8-oxoguanine DNA glycosylase (OGG1) and apurinic/apyrimidinic endonuclease1 (APE1), and the level of 8-hydroxy-2-deoxyguanosine (8-OHdG). Interestingly, the inhibition of DMBA-induced DNA adduct formation was associated with proportional decrease in CYP1A1 and in, Nad(p)h: quinone oxidoreductase 1 (NQO1) gene expression. Mechanistically, the involvements of AhR and nuclear factor erythroid 2-related factor-2 (Nrf2) in the MET-mediated inhibition of DMBA-induced CYP1A1 and NQO1 gene expression were evidenced by the ability of MET to inhibit DMBA-induced xenobiotic responsive element and antioxidant responsive element luciferase reporter gene expression which suggests an AhR- and Nrf2-dependent transcriptional control. However, the inability of MET to bind to AhR suggests that MET is not an AhR ligand. In conclusion, the present work shows a strong evidence that MET inhibits the DMBA-mediated carcinogenicity and adduct formation by inhibiting the expression of CYP1A1 through an AhR ligand-independent mechanism., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
20. Human fetal ventricular cardiomyocyte, RL-14 cell line, is a promising model to study drug metabolizing enzymes and their associated arachidonic acid metabolites.
- Author
-
Maayah ZH, Elshenawy OH, Althurwi HN, Abdelhamid G, and El-Kadi AO
- Subjects
- Cells, Cultured, Cytochrome P-450 Enzyme System genetics, Humans, Isoenzymes genetics, Isoenzymes metabolism, RNA, Messenger genetics, RNA, Messenger metabolism, Arachidonic Acid metabolism, Cytochrome P-450 Enzyme System metabolism, Fenofibrate pharmacology, Models, Biological, Polychlorinated Dibenzodioxins pharmacology
- Abstract
Introduction: RL-14 cells, human fetal ventricular cardiomyocytes, are a commercially available cell line that has been established from non-proliferating primary cultures derived from human fetal heart tissue. However, the expression of different drug metabolizing enzymes (DMEs) in RL-14 cells has not been elucidated yet. Therefore, the main objectives of the current work were to investigate the capacity of RL-14 cells to express different cytochrome P450 (CYP) isoenzymes and correlate this expression to primary cardiomyocytes., Methods: The expression of CYP isoenzymes was determined at mRNA, protein and catalytic activity levels using real time-PCR, Western blot analysis and liquid chromatography-electron spray ionization-mass spectrometry (LC-ESI-MS), respectively., Results: Our results showed that RL-14 cells constitutively express CYP ω-hydroxylases, CYP1A, 1B, 4A and 4F; CYP epoxygenases, CYP2B, 2C and 2J; in addition to soluble epoxide hydrolayse (EPHX2) at mRNA and protein levels. The basal expression of CYP ω-hydroxylases, epoxygenases and EPHX2 was supported by the ability of RL-14 cells to convert arachidonic acid to its biologically active metabolites, 20-hydroxyeicosatetraenoic acids (20-HETEs), 14,15-epoxyeicosatrienoic acids (14,15-EET), 11,12-EET, 8,9-EET, 5,6-EET, 14,15-dihydroxyeicosatrienoic acid (14,15-DHET), 11,12-DHET, 8,9-DHET and 5,6-DHET. Furthermore, RL-14 cells express CYP epoxygenases and ω-hydroxylase at comparable levels to those expressed in adult and fetal human primary cardiomyocytes cells implying the importance of RL-14 cells as a model for studying DMEs in vitro. Lastly, different CYP families were induced in RL-14 cells using 2,3,7,8-tetrachlorodibenzo-p-dioxin and fenofibrate at mRNA and protein levels., Discussion: The current study provides the first evidence that RL-14 cells express CYP isoenzymes at comparable levels to those expressed in the primary cells and thus offers a unique in vitro model to study DMEs in the heart., (Copyright © 2014 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
21. Modulation of aryl hydrocarbon receptor regulated genes by acute administration of trimethylarsine oxide in the lung, kidney and heart of C57BL/6 mice.
- Author
-
Elshenawy OH and El-Kadi AO
- Subjects
- Animals, Arsenicals pharmacology, Cytochrome P-450 CYP1A1 genetics, Cytochrome P-450 CYP1A1 metabolism, Cytochrome P-450 CYP1A2 genetics, Cytochrome P-450 CYP1A2 metabolism, Cytochrome P-450 CYP1B1 genetics, Cytochrome P-450 CYP1B1 metabolism, Enzymes metabolism, Glutathione Transferase genetics, Glutathione Transferase metabolism, Heart drug effects, Heme Oxygenase-1 genetics, Heme Oxygenase-1 metabolism, Isoenzymes genetics, Isoenzymes metabolism, Kidney drug effects, Lung drug effects, Male, Membrane Proteins genetics, Membrane Proteins metabolism, Mice, Inbred C57BL, NAD(P)H Dehydrogenase (Quinone) genetics, NAD(P)H Dehydrogenase (Quinone) metabolism, Polychlorinated Dibenzodioxins toxicity, Receptors, Aryl Hydrocarbon, Arsenicals pharmacokinetics, Enzymes genetics, Gene Expression Regulation drug effects
- Abstract
1. Arsenite alters the expression of aryl hydrocarbon receptor (AhR)-regulated genes in extrahepatic tissues; yet, the effect of organic arsenicals still unknown. Therefore, C57BL/6 mice received trimethylarsine oxide (TMAO; 13 mg/kg i.p.) with or without 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD; 15 μg/kg), and euthanized at 6 or 24 h. 2. Our results demonstrated that TMAO increased Cyp1a1 and Cyp1b1 mRNA, protein and activity in the lung. TMAO potentiated the TCDD-mediated induction of Cyp1a1 and Cyp1a2 mRNA, protein and activity in the lung. In the kidney, TMAO increased Cyp1b1 mRNA and protein. TMAO potentiated the TCDD-mediated induction of Cyp1a1 and Cyp1b1 mRNA, protein and activity. In the heart, TMAO potentiated the TCDD-mediated induction of Cyp1a1 and Cyp1b1 mRNA. 3. Moreover, TMAO induced Nqo1 mRNA in the lung, kidney and heart, with subsequent increase in Nqo1 protein and activity in the lung. TMAO increased Gsta mRNA in the heart; and increased Gsta protein and activity in the lung and kidney. TMAO increased Nqo1 mRNA as compared to TCDD in the kidney and heart, and potentiated the TCDD-mediated induction of Gsta protein and activity in the kidney. 4. In conclusion, TMAO modulates AhR-regulated genes in a tissue- and enzyme-specific manner.
- Published
- 2015
- Full Text
- View/download PDF
22. The role of epoxide hydrolases in health and disease.
- Author
-
El-Sherbeni AA and El-Kadi AO
- Subjects
- Animals, Epoxide Hydrolases genetics, Humans, Microsomes metabolism, Polymorphism, Genetic, Xenobiotics toxicity, Epoxide Hydrolases metabolism, Epoxy Compounds metabolism, Xenobiotics metabolism
- Abstract
Epoxide hydrolases (EH) are ubiquitously expressed in all living organisms and in almost all organs and tissues. They are mainly subdivided into microsomal and soluble EH and catalyze the hydration of epoxides, three-membered-cyclic ethers, to their corresponding dihydrodiols. Owning to the high chemical reactivity of xenobiotic epoxides, microsomal EH is considered protective enzyme against mutagenic and carcinogenic initiation. Nevertheless, several endogenously produced epoxides of fatty acids function as important regulatory mediators. By mediating the formation of cytotoxic dihydrodiol fatty acids on the expense of cytoprotective epoxides of fatty acids, soluble EH is considered to have cytotoxic activity. Indeed, the attenuation of microsomal EH, achieved by chemical inhibitors or preexists due to specific genetic polymorphisms, is linked to the aggravation of the toxicity of xenobiotics, as well as the risk of cancer and inflammatory diseases, whereas soluble EH inhibition has been emerged as a promising intervention against several diseases, most importantly cardiovascular, lung and metabolic diseases. However, there is reportedly a significant overlap in substrate selectivity between microsomal and soluble EH. In addition, microsomal and soluble EH were found to have the same catalytic triad and identical molecular mechanism. Consequently, the physiological functions of microsomal and soluble EH are also overlapped. Thus, studying the biological effects of microsomal or soluble EH alterations needs to include the effects on both the metabolism of reactive metabolites, as well as epoxides of fatty acids. This review focuses on the multifaceted role of EH in the metabolism of xenobiotic and endogenous epoxides and the impact of EH modulations.
- Published
- 2014
- Full Text
- View/download PDF
23. Characterization of arachidonic acid metabolism by rat cytochrome P450 enzymes: the involvement of CYP1As.
- Author
-
El-Sherbeni AA and El-Kadi AO
- Subjects
- Animals, Benzoflavones metabolism, Kinetics, Male, Microsomes, Liver metabolism, Rats, Rats, Sprague-Dawley, Arachidonic Acid metabolism, Cytochrome P-450 Enzyme System metabolism
- Abstract
Cytochrome P450 (P450) enzymes mediate arachidonic acid (AA) oxidation to several biologically active metabolites. Our aims in this study were to characterize AA metabolism by different recombinant rat P450 enzymes and to identify new targets for modulating P450-AA metabolism in vivo. A liquid chromatography-mass spectrometry method was developed and validated for the simultaneous measurements of AA and 15 of its P450 metabolites. CYP1A1, CYP1A2, CYP2B1, CYP2C6, and CYP2C11 were found to metabolize AA with high catalytic activity, and CYP2A1, CYP2C13, CYP2D1, CYP2E1, and CYP3A1 had lower activity. CYP1A1 and CYP1A2 produced ω-1→4 hydroxyeicosatetraenoic acids (HETEs) as 88.7 and 62.7%, respectively, of the total metabolites formed. CYP2C11 produced epoxyeicosatrienoic acids (EETs) as 61.3%, and CYP2C6 produced midchain HETEs and EETs as 48.3 and 29.4%, respectively, of the total metabolites formed. The formation of CYP1A1, CYP1A2, CYP2C6, and CYP2C11 major metabolites followed an atypical kinetic profile of substrate inhibition. CYP1As inhibition by α-naphthoflavone or anti-CYP1As antibodies significantly reduced ω-1→4 HETE formation in the lungs and liver, whereas CYP1As induction by 3-methylcholanthrene resulted in a significant increase in ω-1→4 HETEs formation in the heart, lungs, kidney, and livers by 370, 646, 532, and 848%, respectively. In conclusion, our results suggest that CYP1As and CYP2Cs are major players in the metabolism of AA. The significant contribution of CYP1As to AA metabolism and their strong inducibility suggest their possible use as targets for the prevention and treatment of several diseases., (Copyright © 2014 by The American Society for Pharmacology and Experimental Therapeutics.)
- Published
- 2014
- Full Text
- View/download PDF
24. Acute arsenic treatment alters arachidonic acid and its associated metabolite levels in the brain of C57Bl/6 mice.
- Author
-
Anwar-Mohamed A, Elshenawy OH, El-Sherbeni AA, Abdelrady M, and El-Kadi AO
- Subjects
- Animals, Brain metabolism, Cytochrome P-450 Enzyme System metabolism, Eicosanoids metabolism, Epoxide Hydrolases metabolism, Lipoxygenases metabolism, Male, Mice, Inbred C57BL, Prostaglandin-Endoperoxide Synthases metabolism, Arachidonic Acid metabolism, Arsenites toxicity, Brain drug effects, Environmental Pollutants toxicity, Sodium Compounds toxicity
- Abstract
The toxic effects of arsenic on the whole brain, as well as the discrete regions, has been previously reported for mice. We investigated the effects of acute arsenite (As(III)) on brain levels of arachidonic acid (AA) and its associated metabolites generated through cytochrome P450 (CYP), cyclooxygenase (COX), and lipoxygenase (LOX) pathways. Our results demonstrated that acute As(III) treatment (12.5 mg·(kg body mass)(-1)) decreases cytosolic phospholipase A2 (cPLA2) with a subsequent decrease in its catalytic activity and brain AA levels. In addition, As(III) differentially altered CYP epoxygenases and CYP ω-hydroxylases, but it did not affect brain Ephx2 mRNA or sEH catalytic activity levels. As(III)-mediated effects on Cyps caused an increase in brain 5,6-epoxyeicosatrienoic acid (5,6-EET) and 16/17-hydroxyeicosatetreinoic acid (16/17-HETE) levels, and a decrease in 18- and 20-HETE levels. Furthermore, As(III) increased cyclooxygenase-2 (COX-2) mRNA while decreasing prostaglandins F2α (PGF2α) and PGJ2. As(III) also increased brain 5-lipoxygenase (5-LOX) and 15-LOX mRNA, but decreased 12-LOX mRNA. These changes in LOX mRNA were associated with a decrease in 8/12-HETE levels only. In conclusion, this is the first demonstration that As(III) decreases AA levels coinciding with alterations to EET, HETE, and PG levels, which affects brain development and neurochemistry.
- Published
- 2014
- Full Text
- View/download PDF
25. A rapid and sensitive HPLC assay of some concomitant anti-migraine drugs.
- Author
-
Rezk MR, Michael AM, Lotfy HM, El-Kadi AO, and Shehata MA
- Subjects
- Analgesics chemistry, Linear Models, Naproxen chemistry, Oxazolidinones chemistry, Propranolol chemistry, Reproducibility of Results, Sensitivity and Specificity, Tryptamines chemistry, Analgesics analysis, Chromatography, High Pressure Liquid methods, Naproxen analysis, Oxazolidinones analysis, Propranolol analysis, Tryptamines analysis
- Abstract
This work describes a simple and sensitive method for simultaneous determination of zolmitriptan, naproxen and propranolol in their dosage forms using HPLC. The drugs were separated isocratically on a Zorbax C8 (4.6 × 250 mm with 5 µm particle size) column using a mobile phase composed of 20 mM phosphate citrate buffer [0.1% TEA (pH 3.1)]:methanol:THF (5:3:2, by volumes). The detection was accomplished fluorometrically setting the excitation wavelength at 280 nm and emission wavelength at 360 nm. The method was validated over a linearity range of 100-900 ng/mL for zolmitriptan, 50-300 ng/mL for naproxen and 100-800 ng/mL for propranolol. The assay was successfully applied to the determination of the studied drugs in pharmaceutical dosage forms without interference from tablet excipients with high specificity. The method can be applied successfully in the future for the pharmacokinetic study of these drugs in the human plasma with high accuracy especially that LOQs of zolmitriptan and propranolol in the proposed method cover their Cmax., (© Crown copyright 2013.)
- Published
- 2014
- Full Text
- View/download PDF
26. Acute mercury toxicity modulates cytochrome P450, soluble epoxide hydrolase and their associated arachidonic acid metabolites in C57Bl/6 mouse heart.
- Author
-
Amara IE, Elshenawy OH, Abdelrady M, and El-Kadi AO
- Subjects
- 8,11,14-Eicosatrienoic Acid analogs & derivatives, 8,11,14-Eicosatrienoic Acid metabolism, Animals, Atrial Natriuretic Factor metabolism, Biomarkers metabolism, Cardiomegaly enzymology, Cardiomegaly genetics, Cytochrome P-450 Enzyme System genetics, Gene Expression Regulation, Enzymologic drug effects, Injections, Intraperitoneal, Isoenzymes, Male, Mercuric Chloride administration & dosage, Mice, Mice, Inbred C57BL, Natriuretic Peptide, Brain metabolism, RNA, Messenger metabolism, Arachidonic Acid metabolism, Cardiomegaly chemically induced, Cytochrome P-450 Enzyme System metabolism, Epoxide Hydrolases metabolism, Heart drug effects, Mercuric Chloride toxicity, Myocardium enzymology
- Abstract
Mercury exposure is associated with increased risk of cardiovascular disease and profound cardiotoxicity. However, the correlation between Hg(2+)-mediated toxicity and alteration in cardiac cytochrome P450s (Cyp) and their dependent arachidonic acid metabolites has never been investigated. Therefore, we investigated the effect of acute mercury toxicity on the expression of Cyp-epoxygenases and Cyp-ω-hydroxylases and their associated arachidonic acid metabolites in mice hearts. In addition, we examined the expression and activity of soluble epoxide hydrolase (sEH) as a key player in arachidonic acid metabolism pathway. Mercury toxicity was induced by a single intraperitoneal injection (IP) of 2.5 mg/kg of mercuric chloride (HgCl₂). Our results showed that mercury treatment caused a significant induction of the cardiac hypertrophy markers, atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP); in addition to Cyp1a1, Cyp1b1, Cyp2b9, Cyp2b10, Cyp2b19, Cyp2c29, Cyp2c38, Cyp4a10, Cyp4a12, Cyp4a14, Cyp4f13, Cyp4f15, Cyp4f16 and Cyp4f18 gene expression. Moreover, Hg(2+) significantly increased sEH protein expression and activity levels in hearts of mercury-treated mice, with a consequent decrease in 14,15-, and 11,12-epoxyeicosatrienoic acids (EETs) levels. Whereas the formation of 14,15-, 11,12-, 8,9-dihydroxyeicosatrienoic acids (DHETs) was significantly increased. In conclusion, acute Hg(2+) toxicity modulates the expression of several Cyp and sEH enzymes with a consequent decrease in the cardioprotective EETs which could represent a novel mechanism by which mercury causes progressive cardiotoxicity. Furthermore, inhibiting sEH might represent a novel therapeutic approach to prevent Hg(2+)-induced hypertrophy., (Copyright © 2014 Elsevier Ireland Ltd. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
27. Acute and long-term effects of arsenite in HepG2 cells: modulation of insulin signaling.
- Author
-
Hamann I, Petroll K, Hou X, Anwar-Mohamed A, El-Kadi AO, and Klotz LO
- Subjects
- Androstadienes pharmacology, Hep G2 Cells, Humans, P-Selectin metabolism, Phosphatidylinositol 3-Kinases metabolism, Phosphoinositide-3 Kinase Inhibitors, Phosphorylation drug effects, Proto-Oncogene Proteins c-akt metabolism, Wortmannin, Arsenites administration & dosage, Arsenites pharmacology, Insulin metabolism, Signal Transduction drug effects
- Abstract
Epidemiological studies have indicated a relationship between the prevalence of diabetes and exposure to arsenic. Mechanisms by which arsenic may cause this diabetogenic effect are largely unknown. The phosphoinositide 3'-kinase (PI3K)/Akt signaling pathway plays an important role in insulin signaling by controlling glucose metabolism, in part through regulating the activity of FoxO transcription factors. The present study aimed at investigating the effect of short and long-term exposure to arsenite on insulin signaling in HepG2 human hepatoma cells, the role of PI3K/Akt signaling therein and the modulation of target genes controlled by insulin. Exposure of cells to arsenite for 24 h rendered cells less responsive toward stimulation of Akt by insulin. At the same time, short-term exposure to arsenite induced a concentration-dependent increase in phosphorylation of Akt at Ser-473, followed by phosphorylation of FoxO proteins at sites known to be phosphorylated by Akt. Phosphorylation of FoxOs was prevented by wortmannin, pointing to the involvement of PI3K. Arsenite exposure resulted in attenuation of FoxO DNA binding and in nuclear exclusion of FoxO1a-EGFP. A 24-h exposure of HepG2 cells to submicromolar concentrations of arsenite resulted in downregulation of glucose 6-phosphatase (G6Pase) and selenoprotein P (SelP) mRNA levels. Curiously, arsenite had a dual effect on SelP protein levels, inducing a small increase in the nanomolar and a distinct decrease in the micromolar concentration range. Interestingly, arsenite-induced long-term effects on G6Pase and SelP mRNA or SelP protein levels were not blocked by the PI3K inhibitor, wortmannin. In conclusion, arsenite perturbs cellular signaling pathways involved in fuel metabolism: it impairs cellular responsiveness toward insulin, while at the same time stimulating insulin-like signaling to attenuate the expression of genes involved in glucose metabolism and the release of the hepatokine SelP, which is known to modulate peripheral insulin sensitivity.
- Published
- 2014
- Full Text
- View/download PDF
28. Effects of serum lipoproteins on cyclosporine A cellular uptake and renal toxicity in vitro.
- Author
-
Brocks DR, Chaudhary HR, Ben-Eltriki M, Elsherbiny ME, and El-Kadi AO
- Subjects
- ATP Binding Cassette Transporter, Subfamily B, Member 1 metabolism, Animals, Atorvastatin, Cells, Cultured, Cyclosporine toxicity, Hepatocytes drug effects, Hepatocytes metabolism, Heptanoic Acids pharmacology, Immunosuppressive Agents toxicity, Kidney metabolism, Pyrroles pharmacology, Rats, Rats, Sprague-Dawley, Cyclosporine metabolism, Hypolipidemic Agents pharmacology, Immunosuppressive Agents metabolism, Kidney drug effects, Lipoproteins blood
- Abstract
In-vitro studies were performed to shed light on previous findings that showed increased uptake of cyclosporine A in the kidneys and liver of hyperlipidemic rats, and increased signs of kidney toxicity. Hepatocytes were obtained from rats, cultured, and exposed to a diluted serum from hyperlipidemic rats. Some cells were also exposed to lipid-lowering drugs. After washing out the rat serum or lipid-lowering drugs, cells were exposed to cyclosporine A embedded in serum lipoproteins. Pretreatment with hyperlipidemic serum and lipid-lowering drugs was associated with an increased uptake of cyclosporine A. As expected, atorvastatin caused an increase in low density lipoprotein receptor and a decrease in MDR1A mRNA in the hepatocytes. A decrease in NRK-52E rat renal tubular cellular viability caused by cyclosporine A was noted when cells were preincubated with diluted hyperlipidemic serum. This was matched with evidence of hyperlipidemic-serum-associated increases in the NRK-52E cellular uptake of cyclosporine A and rhodamine-123. The findings of these experiments suggested that in hyperlipidemia the expression and (or) the functional activity of P-glycoprotein was diminished, leading to greater hepatic and renal uptake of cyclosporine A, and renal cellular toxicity.
- Published
- 2014
- Full Text
- View/download PDF
29. Alterations in cytochrome P450-derived arachidonic acid metabolism during pressure overload-induced cardiac hypertrophy.
- Author
-
El-Sherbeni AA and El-Kadi AO
- Subjects
- Animals, Cardiomegaly metabolism, Cytochrome P-450 Enzyme System genetics, Gene Expression Regulation, Enzymologic physiology, Rats, Rats, Sprague-Dawley, Arachidonic Acid metabolism, Cardiomegaly pathology, Cytochrome P-450 Enzyme System metabolism
- Abstract
Cardiac hypertrophy is a major risk factor for many serious heart diseases. Recent data demonstrated the role of cytochrome P450 (CYP)-derived arachidonic acid (AA) metabolites in cardiovascular pathophysiology. In the current study our aim was to determine the aberrations in CYP-mediated AA metabolism in the heart during cardiac hypertrophy. Pressure overload cardiac hypertrophy was induced in Sprague Dawley rats using the descending aortic constriction procedure. Five weeks post-surgery, the cardiac levels of AA metabolites were determined in hypertrophied and normal hearts. In addition, the formation rate of AA metabolites, as well as, CYP expression in cardiac microsomal fraction was also determined. AA metabolites were measured by liquid chromatography-electrospray ionization-mass spectroscopy, whereas, the expression of CYPs was determined by Western blot analysis. Non-parametric analysis was performed to examine the association between metabolites formation and CYP expressions. Our results showed that 5,6-, 8,9-, 11,12-, and 14,15-epoxyeicosatrienoic acids (EETs), and 5-, 12-, 15-, and 20-hydroxyeicosatetraenoic acids (HETEs) levels were increased, whereas, 19-HETE formation was decreased in hypertrophied hearts. The increase in EETs was linked to CYP2B2. On the other hand, CYP1B1 and CYP2J3 were involved in mid-chain HETE metabolism, whereas, CYP4A2/3 inhibition was involved in the decrease in 19-HETE formation in hypertrophied hearts. In conclusion, CYP1B1 played cardiotoxic role, whereas, CYP2B2, CYP2J3 and CYP4A2/3 played cardioprotective roles during pressure overload-induced cardiac hypertrophy. These CYP can be valid targets for the development of drugs to treat and prevent cardiac hypertrophy and heart failure., (Copyright © 2013 Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
30. Fenofibrate modulates cytochrome P450 and arachidonic acid metabolism in the heart and protects against isoproterenol-induced cardiac hypertrophy.
- Author
-
Althurwi HN, Elshenawy OH, and El-Kadi AO
- Subjects
- 8,11,14-Eicosatrienoic Acid analogs & derivatives, 8,11,14-Eicosatrienoic Acid metabolism, Animals, Cytochrome P-450 CYP2J2, Cytochrome P-450 Enzyme System genetics, Cytochrome P-450 Enzyme System metabolism, Gene Expression Regulation, Enzymologic drug effects, Hydroxyeicosatetraenoic Acids metabolism, Hypolipidemic Agents pharmacology, Isoproterenol toxicity, Male, Rats, Rats, Sprague-Dawley, Arachidonic Acid metabolism, Cardiomegaly prevention & control, Cytochrome P-450 Enzyme System drug effects, Fenofibrate pharmacology
- Abstract
It has been previously shown that the cytochrome P450 (P450) modulator, fenofibrate, protects against cardiovascular diseases. P450 and their metabolites, epoxyeicosatrienoic acids (EETs) and 20-hydroxyeicosatetraenoic acid (20-HETE) were found to play an important role in cardiovascular diseases. Therefore, it is important to examine whether fenofibrate would modulate the cardiac P450 and its associated arachidonic acid metabolites and whether this modulation protects against isoproterenol-induced cardiac hypertrophy. For this purpose, male Sprague-Dawley rats were treated with fenofibrate (30 mg·kg·d), isoproterenol (4.2 mg·kg·d), or the combination of both. The expression of hypertrophic markers and different P450s along with their metabolites was determined. Our results showed that fenofibrate significantly induced the cardiac P450 epoxygenases, such as CYP2B1, CYP2B2, CYP2C11, and CYP2C23, whereas it decreased the cardiac ω-hydroxylase, CYP4A3. Moreover, fenofibrate significantly increased the formation of 14,15-EET, 11,12-EET, and 8,9-EET, whereas it decreased the formation of 20-HETE in the heart. Furthermore, fenofibrate significantly decreased the hypertrophic markers and the increase in heart-to-body weight ratio induced by isoproterenol. This study demonstrates that fenofibrate alters the expression of cardiac P450s and their metabolites and partially protects against isoproterenol-induced cardiac hypertrophy, which further confirms the role of P450s, EETs, and 20-HETE in the development of cardiac hypertrophy.
- Published
- 2014
- Full Text
- View/download PDF
31. Methylated pentavalent arsenic metabolites are bifunctional inducers, as they induce cytochrome P450 1A1 and NAD(P)H:quinone oxidoreductase through AhR- and Nrf2-dependent mechanisms.
- Author
-
Anwar-Mohamed A, Elshenawy OH, Soshilov AA, Denison MS, Chris Le X, Klotz LO, and El-Kadi AO
- Subjects
- Antioxidant Response Elements, Cytochrome P-450 CYP1A1 metabolism, Gene Expression Regulation, Genes, Reporter, HSP90 Heat-Shock Proteins genetics, HSP90 Heat-Shock Proteins metabolism, Hep G2 Cells, Humans, Isothiocyanates pharmacology, Luciferases genetics, Luciferases metabolism, Methylation, NAD(P)H Dehydrogenase (Quinone) metabolism, NF-E2-Related Factor 2 metabolism, Polychlorinated Dibenzodioxins pharmacology, Receptors, Aryl Hydrocarbon metabolism, Sulfoxides, Arsenicals pharmacology, Cytochrome P-450 CYP1A1 genetics, NAD(P)H Dehydrogenase (Quinone) genetics, NF-E2-Related Factor 2 genetics, Receptors, Aryl Hydrocarbon genetics, Signal Transduction drug effects
- Abstract
Activation of the aryl hydrocarbon receptor (AhR) ultimately leads to the induction of the carcinogen-activating enzyme cytochrome P450 1A1 (CYP1A1), and activation of the nuclear factor-erythroid 2 p45-related factor 2 (Nrf2) in addition to the AhR pathway induces the expression of the NADP(H):quinone oxidoreductase (NQO1). Therefore, the aim of this study was to examine the effect of As(III) pentavalent metabolites, MMA(V), DMA(V), and TMA(V), on AhR and Nrf2 activation and on the expression of their prototypical downstream targets CYP1A1 and NQO1, respectively. Our results showed that treatment of HepG2 cells with MMA(V), DMA(V), or TMA(V) in the absence and presence of 2,3,7,8-tetrachlorodibenzo-p-dioxin or sulforaphane significantly induced both CYP1A1 and NQO1 at the mRNA, protein, and catalytic activity levels. Furthermore, these metabolites increased the AhR-dependent XRE-driven and the Nrf2-dependent ARE-driven luciferase reporter activities, which coincided with increased nuclear accumulation of both transcription factors. However, none of these metabolites were shown to be AhR ligands. The induction of CYP1A1 by these metabolites seems to be ligand-independent, possibly through a decrease in HSP90 protein expression levels. The metabolites also increased ROS production, which was significantly higher than that produced by As(III). Upon knockdown of AhR and Nrf2 the MMA(V)-, DMA(V)-, and TMA(V)-mediated induction of both CYP1A1 and NQO1 proteins was significantly decreased. In conclusion, this study demonstrates for the first time that methylated pentavalent arsenic metabolites are bifunctional inducers, as they increase CYP1A1 by activating the AhR/XRE signaling pathway and they increase NQO1 by activating the Nrf2/ARE signaling pathway in addition to the AhR/XRE pathway., (© 2013 Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
32. Modulation of aryl hydrocarbon receptor-regulated genes by acute administration of ammonium metavanadate in kidney, lung and heart of C57BL/6 mice.
- Author
-
Abdelhamid G, Amara IE, Anwar-Mohamed A, and El-Kadi AO
- Subjects
- Animals, Blotting, Western, Cytochrome P-450 CYP1A1 genetics, Cytochrome P-450 CYP1A2 genetics, Cytochrome P-450 CYP1B1 genetics, Drug Interactions, Glutathione Transferase genetics, Isoenzymes genetics, Kidney metabolism, Lung metabolism, Male, Mice, Inbred C57BL, Microsomes drug effects, Microsomes metabolism, Myocardium metabolism, NAD(P)H Dehydrogenase (Quinone) genetics, Real-Time Polymerase Chain Reaction, Gene Expression Regulation drug effects, Heart drug effects, Kidney drug effects, Lung drug effects, Polychlorinated Dibenzodioxins toxicity, Receptors, Aryl Hydrocarbon genetics, Vanadates pharmacology
- Abstract
We recently reported that vanadium (V(5+) ) was able to decrease the 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-mediated induction of Cyp1a1 and Nqo1 at mRNA, protein and catalytic activity levels in mouse hepatoma Hepa 1c1c7 and human hepatoma HepG2 cells. However, little is known regarding the in vivo effects. Thus, the objective of this study was to investigate whether similar effects would occur at the in vivo level. Therefore, we examined the effect of exposure to V(5+) (5 mg kg(-1) ) with or without TCDD (15 µg kg(-1) ) on the AhR-regulated genes in kidney, lung and heart of C57BL/6 J mice. Our results demonstrated that V(5+) alone significantly decreased Cyp1b1 protein and catalytic activity levels in kidney at 24 h. Moreover, it significantly potentiated Nqo1 and Gsta1 gene expression in the heart, and only Gsta1 gene expression in the lung. Upon co-exposure, we found that V(5+) significantly inhibited the TCDD-mediated induction of Cyp1a1, Cyp1a2 and Cyp1b1 mRNA, protein and catalytic activity levels in the kidney at 24 h. On the other hand, V(5+) significantly potentiated the TCDD-mediated induction of Nqo1 and Gsta1 protein and activity levels in the kidney. Cyp1a1, Cyp1b1, Nqo1 mRNA, protein and catalytic activity levels in the lung were significantly potentiated at 6 h. Interestingly, all tested genes in the heart were significantly decreased at 6 h with the exception of Gsta1 mRNA. The present study demonstrates that V(5+) modulates TCDD-induced AhR-regulated genes. Furthermore, the effect on one of these enzymes could not be generalized to other enzymes even if it was in the same organ., (Copyright © 2012 John Wiley & Sons, Ltd.)
- Published
- 2013
- Full Text
- View/download PDF
33. The tumor suppressor gene, RASSF1A, is essential for protection against inflammation -induced injury.
- Author
-
Gordon M, El-Kalla M, Zhao Y, Fiteih Y, Law J, Volodko N, Anwar-Mohamed A, El-Kadi AO, Liu L, Odenbach J, Thiesen A, Onyskiw C, Ghazaleh HA, Park J, Lee SB, Yu VC, Fernandez-Patron C, Alexander RT, Wine E, and Baksh S
- Subjects
- Adaptor Proteins, Signal Transducing genetics, Adaptor Proteins, Signal Transducing metabolism, Animals, Apoptosis drug effects, Benzamides pharmacology, Cell Cycle Proteins, Cell Proliferation drug effects, Colitis, Ulcerative chemically induced, Colitis, Ulcerative drug therapy, Colitis, Ulcerative pathology, Colon drug effects, Colon pathology, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Dextran Sulfate, Epithelial Cells drug effects, Epithelial Cells pathology, Gene Expression Regulation, Imatinib Mesylate, Inflammation chemically induced, Inflammation drug therapy, Inflammation genetics, Inflammation pathology, Intestinal Mucosa drug effects, Intestinal Mucosa pathology, Mice, Mice, Knockout, NF-kappa B metabolism, Nuclear Proteins genetics, Nuclear Proteins metabolism, Phosphoproteins genetics, Phosphoproteins metabolism, Piperazines pharmacology, Proto-Oncogene Proteins c-abl pharmacology, Pyrimidines pharmacology, Signal Transduction, Toll-Like Receptors metabolism, Tumor Protein p73, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 metabolism, Tumor Suppressor Proteins deficiency, Tumor Suppressor Proteins metabolism, YAP-Signaling Proteins, bcl-2-Associated X Protein genetics, bcl-2-Associated X Protein metabolism, Colitis, Ulcerative genetics, Colon metabolism, Epithelial Cells metabolism, Intestinal Mucosa metabolism, NF-kappa B genetics, Toll-Like Receptors genetics, Tumor Suppressor Proteins genetics
- Abstract
Ras association domain family protein 1A (RASSF1A) is a tumor suppressor gene silenced in cancer. Here we report that RASSF1A is a novel regulator of intestinal inflammation as Rassf1a(+/-) , Rassf1a(-/-) and an intestinal epithelial cell specific knockout mouse (Rassf1a (IEC-KO) ) rapidly became sick following dextran sulphate sodium (DSS) administration, a chemical inducer of colitis. Rassf1a knockout mice displayed clinical symptoms of inflammatory bowel disease including: increased intestinal permeability, enhanced cytokine/chemokine production, elevated nuclear factor of kappa light polypeptide gene enhancer in B-cells (NFκB) activity, elevated colonic cell death and epithelial cell injury. Furthermore, epithelial restitution/repair was inhibited in DSS-treated Rassf1a(-/-) mice with reduction of several makers of proliferation including Yes associated protein (YAP)-driven proliferation. Surprisingly, tyrosine phosphorylation of YAP was detected which coincided with increased nuclear p73 association, Bax-driven epithelial cell death and p53 accumulation resulting in enhanced apoptosis and poor survival of DSS-treated Rassf1a knockout mice. We can inhibit these events and promote the survival of DSS-treated Rassf1a knockout mice with intraperitoneal injection of the c-Abl and c-Abl related protein tyrosine kinase inhibitor, imatinib/gleevec. However, p53 accumulation was not inhibited by imatinib/gleevec in the Rassf1a(-/-) background which revealed the importance of p53-dependent cell death during intestinal inflammation. These observations suggest that tyrosine phosphorylation of YAP (to drive p73 association and up-regulation of pro-apoptotic genes such as Bax) and accumulation of p53 are consequences of inflammation-induced injury in DSS-treated Rassf1a(-/-) mice. Mechanistically, we can detect robust associations of RASSF1A with membrane proximal Toll-like receptor (TLR) components to suggest that RASSF1A may function to interfere and restrict TLR-driven activation of NFκB. Failure to restrict NFκB resulted in the inflammation-induced DNA damage driven tyrosine phosphorylation of YAP, subsequent p53 accumulation and loss of intestinal epithelial homeostasis.
- Published
- 2013
- Full Text
- View/download PDF
34. Effects of growth hormone and ultrasound on mandibular growth in rats: MicroCT and toxicity analyses.
- Author
-
Khan I, El-Kadi AO, and El-Bialy T
- Subjects
- Analysis of Variance, Animals, Bone Density, Gene Expression, Growth Hormone toxicity, Mandible diagnostic imaging, Mandible growth & development, RNA, Messenger, Rats, Rats, Sprague-Dawley, Real-Time Polymerase Chain Reaction, Temporomandibular Joint, Tomography, X-Ray Computed, Ultrasonic Therapy, Ultrasonography, X-Ray Microtomography, Genes, jun genetics, Growth Hormone administration & dosage, Liver metabolism, Mandible abnormalities
- Abstract
It has been shown by previous studies that mandibular growth can be enhanced by the systemic administration of recombinant growth hormone (rGH) and/or local application of therapeutic low intensity pulsed ultrasound (LIPUS). The purpose of this study was to determine if local injection of rGH and application of LIPUS to the temporomandibular joint (TMJ) would synergistically enhance mandibular growth. In an animal study, the effect of rGH, LIPUS, and combination of rGH and LIPUS on male Sprague-Dawley rats was observed. Mandibular growth was evaluated by measuring total hemimandibular and condylar bone volume and bone surface area as well as condylar bone mineral density (BMD) after 21 days on dissected rats' mandibles using micro-computed tomography (MicroCT). The expression of c-jun mRNA extracted from the liver of each of these rats was also quantified by real-time polymerase chain reaction to evaluate possible systemic effect of local rGH administration. Significant growth stimulation was observed in the mandibular and condylar bone of the animals treated with rGH, LIPUS, and rGH/LIPUS combined when compared with the control group. Bone volume, surface area, condylar bone mineral density, and c-jun expression were also compared between the treatment groups and the control in the liver. The results suggest that mandibular growth may be enhanced by injection of rGH or LIPUS application. The current study although showed synergetic effect of rGH and LIPUS application in increasing mandibular condylar head length, there was no significant changes in mandibular bone volume using both treatments together when compared to the two individual treatments. Moreover, combined rGH and LIPUS decreased condylar bone mineral density than each treatment separately. Future research could be directed to investigate the effects of different rGH doses and/or different LIPUS exposures parameters on lower jaw growth., (Copyright © 2013 Elsevier Ltd. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
35. Acute arsenic treatment alters cytochrome P450 expression and arachidonic acid metabolism in lung, liver and kidney of C57Bl/6 mice.
- Author
-
Anwar-Mohamed A, El-Sherbeni A, Kim SH, Elshenawy OH, Althurwi HN, Zordoky BN, and El-Kadi AO
- Subjects
- Animals, Cytochrome P-450 Enzyme System genetics, Epoxide Hydrolases genetics, Epoxide Hydrolases metabolism, Gene Expression Regulation, Enzymologic drug effects, Humans, Hydroxyeicosatetraenoic Acids metabolism, Kidney drug effects, Liver drug effects, Lung drug effects, Male, Mice, Mice, Inbred C57BL, Arachidonic Acid metabolism, Arsenic toxicity, Cytochrome P-450 Enzyme System metabolism, Kidney enzymology, Liver enzymology, Lung enzymology
- Abstract
1. Arsenic (As(III)) toxicity has received increasing attention as human exposure to arsenic is associated with pulmonary, hepatic and renal toxicities. Therefore, in the present study, we investigated the effect of acute As(III) treatment on pulmonary, hepatic and renal cytochrome (CYP) P450-mediated arachidonic acid metabolism. 2. Our results demonstrated that acute As(III) treatment (12.5 mg/kg) altered CYP epoxygenases, CYP ω-hydroxylases and EPHX2 mRNA levels that were isozyme and tissue specific. 3. Furthermore, As(III) increased the formation of epoxyeicosatrienoic acids (EETs) in the kidney without affecting their levels in the lung or liver. In addition, acute As(III) treatment increased dihydroxyeicosatrienoic acid (DHETs) formation in the lung, while it did not affect liver DHETs formation and decreased kidney DHETs formation. 4. As(III) also increased total epoxygenases activity in the lung while it decreased its levels in the kidney and had no effect on the liver. Furthermore, As(III) increased 20-hydroxyeicosatetraenoic acid formation in the liver while it decreased its formation in the kidney. 5. Lastly, As(III) increased soluble epoxide hydrolase activity in the lung, while it decreased its levels in the kidney and had no effect on the liver. In conclusion, this is the first demonstration that As(III) alters arachidonic acid metabolism in a tissue specific manner.
- Published
- 2013
- Full Text
- View/download PDF
36. Modulation of cytochrome P450 1 (Cyp1) by vanadium in hepatic tissue and isolated hepatocyte of C57BL/6 mice.
- Author
-
Abdelhamid G, Amara IE, Anwar-Mohamed A, and El-Kadi AO
- Subjects
- Animals, Aryl Hydrocarbon Hydroxylases genetics, Aryl Hydrocarbon Hydroxylases metabolism, Cell Survival drug effects, Cells, Cultured, Cytochrome P-450 CYP1A1 genetics, Cytochrome P-450 CYP1A2 genetics, Cytochrome P-450 CYP1B1, Dose-Response Relationship, Drug, Gene Expression Regulation, Enzymologic, Heme Oxygenase-1 genetics, Hemoglobins metabolism, Hepatocytes enzymology, Hepatocytes metabolism, Liver enzymology, Liver metabolism, Male, Membrane Proteins genetics, Mice, Mice, Inbred C57BL, Receptors, Aryl Hydrocarbon metabolism, Cytochrome P-450 CYP1A1 metabolism, Cytochrome P-450 CYP1A2 metabolism, Hepatocytes drug effects, Liver drug effects, Polychlorinated Dibenzodioxins pharmacology, Vanadium pharmacology
- Abstract
The objective of the current study was to investigate the effect of vanadium (V(5+)) on Cyp1 expression and activity in C57BL/6 mice liver and isolated hepatocytes. For this purpose, C57BL6 mice were injected intraperitoneally with V(5+) (5 mg/kg) in the absence and presence of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) (15 μg/kg) for 6 and 24 h. Furthermore, isolated hepatocytes from C57BL6 mice were treated with V(5+) (5, 10, and 20 μM) in the absence and presence of TCDD (1 nM) for 3, 6, 12, and 24 h. In vivo, V(5+) alone did not significantly alter Cyp1a1, Cyp1a2, or Cyp1b1 mRNA, protein, or catalytic activity levels. Upon co-exposure to V(5+) and TCDD, V(5+) significantly potentiated the TCDD-mediated induction of the Cyp1a1, Cyp1a2, and Cyp1b1 mRNA, protein, and catalytic activity levels at 24 h. In vitro, V(5+) decreased the TCDD-mediated induction of Cyp1a1 mRNA, protein, and catalytic activity levels. Furthermore, V(5+) significantly inhibited the TCDD-induced AhR-dependent luciferase activity. V(5+) also increased serum hemoglobin (Hb) levels in animals treated for 24 h. Upon treatment of isolated hepatocytes with Hb alone or in the presence of TCDD, there was an increase in the AhR-dependent luciferase activity. When isolated hepatocytes were treated for 2 h with V(5+) in the presence of TCDD, followed by replacement of the medium with new medium containing Hb, there was further potentiation to the TCDD-mediated effect. The present study demonstrates that there is a differential modulation of Cyp1a1 by V(5+) in C57BL/6 mice livers and isolated hepatocytes and demonstrates Hb as an in vivo specific modulator.
- Published
- 2013
- Full Text
- View/download PDF
37. 20-Hydroxyeicosatetraenoic acid is a potential therapeutic target in cardiovascular diseases.
- Author
-
Elshenawy OH, Anwar-Mohamed A, and El-Kadi AO
- Subjects
- Animals, Arachidonic Acid metabolism, Humans, Hydroxylation, Cardiovascular Diseases metabolism, Hydroxyeicosatetraenoic Acids metabolism
- Abstract
Arachidonic acid (AA) is metabolized by enzymes of the cytochrome P450 (CYP) 4A and CYP4F subfamilies to 20- hydroxyeicosatetraeonic acid (20-HETE), which plays an important role in the cardiovascular system. In the current work, we reviewed the formation of 20-HETE in different species by different CYPs; 20-HETE metabolism by cyclooxygenases (COXs) and different isomerases; and the current available inducers and inhibitors of 20-HETE formation in addition to its agonists and antagonists. Moreover we reviewed the negative role of 20-HETE in cardiac hypertrophy, cardiotoxicity, diabetic cardiomyopathy, and in ischemia/reperfusion (I/R) injury. Lastly, we reviewed the role of 20-HETE in different hypertension models such as the renin/angiotensin II model, Goldblatt model, spontaneously hypertensive rat model, androgen-induced model, slat- and deoxycorticosterone acetate (DOCA)-salt-induced models, and high fat diet model. 20-HETE can affect pro- and anti-hypertensive mechanisms dependent upon where, when, and by which isoform it has been produced. In contrast to hypertension we also reviewed the role of 20-HETE in endotoxin-induced hypotension and the natriuretic effects of 20-HETE. Based on the recent studies, 20-HETE production and/or action might be a therapeutic target to protect against the initiation and progression of cardiovascular diseases.
- Published
- 2013
- Full Text
- View/download PDF
38. Differential effects of soluble epoxide hydrolase inhibition and CYP2J2 overexpression on postischemic cardiac function in aged mice.
- Author
-
Chaudhary KR, Zordoky BN, Edin ML, Alsaleh N, El-Kadi AO, Zeldin DC, and Seubert JM
- Subjects
- 8,11,14-Eicosatrienoic Acid metabolism, 8,11,14-Eicosatrienoic Acid pharmacology, Age Factors, Animals, Cells, Cultured, Cytochrome P-450 CYP2J2, Cytochrome P-450 Enzyme System genetics, Epoxide Hydrolases deficiency, Female, Gene Expression Regulation, Heart physiopathology, Male, Mice, Mice, Knockout, Myocardial Reperfusion Injury enzymology, Myocardial Reperfusion Injury physiopathology, Myocytes, Cardiac drug effects, Myocytes, Cardiac enzymology, Myocytes, Cardiac pathology, Organ Culture Techniques, Oxidative Stress, Protein Phosphatase 2 genetics, Protein Phosphatase 2 metabolism, Stearic Acids metabolism, Urea pharmacology, 8,11,14-Eicosatrienoic Acid analogs & derivatives, Benzoates pharmacology, Cardiotonic Agents pharmacology, Cytochrome P-450 Enzyme System metabolism, Epoxide Hydrolases antagonists & inhibitors, Heart drug effects, Myocardial Reperfusion Injury prevention & control, Urea analogs & derivatives
- Abstract
Cardioprotective effects of epoxyeicosatrienoic acids (EETs) have been demonstrated in models of young mice with either the cardiomyocyte specific over-expression of cytochrome P450 2J2 (CYP2J2 Tr) or deletion of soluble epoxide hydrolase (sEH null). In this study we examined differences in EET-induced cardioprotection in young (2 months) and aged (12 months) CYP2J2 Tr and sEHnull mice using Langendorff isolated perfused heart model. Improved postischemic functional recovery was observed in both young and aged sEH null mice compared to age matched WT. Conversely, the cardioprotective effect observed in young CYP2J2 Tr was lost in aged CYP2J2 Tr mice. The loss of cardioprotection in aged CYP2J2 Tr was regained following perfusion with the sEH inhibitor t-AUCB. Data demonstrated increased levels of leukotoxin diol (DiHOME) and oxidative stress as well decreased protein phosphatase 2A (PP2A) activation in aged CYP2J2 Tr. In conclusion, inhibition of sEH and EET-induced cardioprotection is maintained in aged mice. However, the loss of protective effects observed in aged CYP2J2 Tr might be attributed to increased levels of DiHOME, oxidative stress and/or decreased PP2A activity., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
39. Posttranslational mechanisms modulating the expression of the cytochrome P450 1A1 gene by methylmercury in HepG2 cells: a role of heme oxygenase-1.
- Author
-
Amara IE, Anwar-Mohamed A, and El-Kadi AO
- Subjects
- Blotting, Western, Cytochrome P-450 CYP1A1 antagonists & inhibitors, Cytochrome P-450 CYP1A1 biosynthesis, Gene Expression Regulation drug effects, Gene Knockdown Techniques, Heme Oxygenase-1 physiology, Hep G2 Cells drug effects, Hep G2 Cells metabolism, Humans, Polychlorinated Dibenzodioxins pharmacology, Protein Processing, Post-Translational drug effects, Real-Time Polymerase Chain Reaction, Cytochrome P-450 CYP1A1 genetics, Heme Oxygenase-1 metabolism, Methylmercury Compounds pharmacology
- Abstract
Recently we demonstrated the ability of mercuric chloride (Hg(2+)) in human hepatoma HepG2 cells to significantly decrease the TCDD-mediated induction of Cytochrome P450 1A1 (CYP1A1) mRNA, protein, and catalytic activity levels. In this study we investigated the effect of methylmercury (MeHg) on CYP1A1 in HepG2 cells. For this purpose, cells were co-exposed to MeHg and TCDD and the expression of CYP1A1 mRNA, protein, and catalytic activity levels were determined. Our results showed that MeHg did not alter the TCDD-mediated induction of CYP1A1 mRNA, or protein levels; however it was able to significantly decrease CYP1A1 catalytic activity levels in a concentration-dependent manner. Importantly, this inhibition was specific to CYP1A1and was not radiated to other aryl hydrocarbon receptor (AhR)-regulated genes, as MeHg induced NAD(P)H:quinone oxidoreductase 1 mRNA and protein levels. Mechanistically, the inhibitory effect of MeHg on the induction of CYP1A1 coincided with an increase in heme oxygenase-1 (HO-1) mRNA levels. Furthermore, the inhibition of HO-1 activity, by tin mesoporphyrin, caused a complete restoration of MeHg-mediated inhibition of CYP1A1 activity, induced by TCDD. In addition, transfection of HepG2 cells with siRNA targeting the human HO-1 gene reversed the MeHg-mediated inhibition of TCDD-induced CYP1A1. In conclusion, this study demonstrated that MeHg inhibited the TCDD-mediated induction of CYP1A1 through a posttranslational mechanism and confirms the role of HO-1 in a MeHg-mediated effect., (Copyright © 2013 Elsevier Ireland Ltd. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
40. Sunitinib, a tyrosine kinase inhibitor, induces cytochrome P450 1A1 gene in human breast cancer MCF7 cells through ligand-independent aryl hydrocarbon receptor activation.
- Author
-
Maayah ZH, El Gendy MA, El-Kadi AO, and Korashy HM
- Subjects
- Breast Neoplasms enzymology, Cytochrome P-450 CYP1A1 biosynthesis, Dactinomycin pharmacology, Dose-Response Relationship, Drug, Enzyme Induction drug effects, Female, Gene Expression Regulation, Enzymologic drug effects, Humans, Ligands, MCF-7 Cells, RNA, Messenger metabolism, Receptors, Aryl Hydrocarbon drug effects, Receptors, Aryl Hydrocarbon metabolism, Resveratrol, Stilbenes pharmacology, Sunitinib, Transcriptional Activation drug effects, Antineoplastic Agents pharmacology, Breast Neoplasms drug therapy, Cytochrome P-450 CYP1A1 genetics, Enzyme Inhibitors pharmacology, Indoles pharmacology, Protein-Tyrosine Kinases antagonists & inhibitors, Pyrroles pharmacology
- Abstract
Sunitinib (SUN) is a new multi-targeted oral tyrosine kinase inhibitor that has both anti-angiogenic and anti-tumor activities. However, information reported in the literature on the effects of SUN on the constitutive expression of cytochrome P450 1A1 (CYP1A1) gene in cells from mammalian species remains unclear. Therefore, the main objectives of the current work were to investigate the potentiality of SUN to induce CYP1A1 gene expression in human breast cancer MCF7 cells and to explore the molecular mechanisms involved. Our results showed that SUN induced the CYP1A1 mRNA, protein, and activity levels in a concentration-dependent manner in MCF7 cells. The increase in CYP1A1 mRNA by SUN was completely blocked by the transcriptional inhibitor, actinomycin D; implying that SUN increased de novo RNA synthesis. Furthermore, the ability of SUN to increase luciferase reporter gene expression suggests an aryl hydrocarbon receptor (AhR)-dependent transcriptional control and excludes the possibility of any posttranscriptional mechanisms. In addition, blocking of AhR activation by resveratrol, a well-known AhR antagonist, prevented the SUN-induced CYP1A1 gene expression, further confirms the involvement of AhR. Interestingly, this was associated with the inability of SUN to directly bind to and induce transformation of cytosolic AhR to its DNA-binding form in vitro, suggesting that the effect of SUN does not involve direct binding to AhR. The current manuscript provides the first evidence for the ability of SUN to induce CYP1A1 gene expression in MCF7 cells through AhR ligand-independent mechanisms.
- Published
- 2013
- Full Text
- View/download PDF
41. Differential modulation of cytochrome P450 1a1 by arsenite in vivo and in vitro in C57BL/6 mice.
- Author
-
Anwar-Mohamed A, Abdelhamid G, Amara IE, and El-Kadi AO
- Subjects
- Animals, Cells, Cultured, Free Radicals metabolism, Gene Expression Regulation drug effects, Heme Oxygenase-1 metabolism, Hepatocytes drug effects, Hepatocytes metabolism, Liver drug effects, Liver enzymology, Membrane Proteins metabolism, Metals, Heavy pharmacology, Mice, RNA, Messenger drug effects, Receptors, Aryl Hydrocarbon metabolism, Arsenites pharmacology, Cytochrome P-450 CYP1A1 metabolism, Hemoglobins metabolism, Polychlorinated Dibenzodioxins pharmacology
- Abstract
Heavy metals, typified by arsenite (As(III)), have been implicated in altering the carcinogenicity of aryl hydrocarbon receptor (AhR) ligands, typified by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), by modulating the induction of the Cyp1a1 enzyme, but the mechanism remains unresolved. In this study, the effects of As(III) on Cyp1a1 expression and activity were investigated in C57BL/6 mouse livers and isolated hepatocytes. For this purpose, C57BL/6 mice were injected intraperitoneally with As(III) (12.5 mg/kg) in the absence and presence of TCDD (15 μg/kg) for 6 and 24 h. Furthermore, isolated hepatocytes from C57BL/6 mice were treated with As(III) (1, 5, and 10 μM) in the absence and presence of TCDD (1 nM) for 3, 6, 12, and 24 h. At the in vivo level, As(III) decreased the TCDD-mediated induction of Cyp1a1 mRNA at 6h while potentiating its mRNA, protein, and catalytic activity levels at 24 h. At the in vitro level, As(III) decreased the TCDD-mediated induction of Cyp1a1 mRNA in a concentration- and time-dependent manner. Moreover, As(III) decreased the TCDD-mediated induction of Cyp1a1 protein and catalytic activity levels at 24 h. Interestingly, As(III) increased the serum hemoglobin (Hb) levels in animals treated for 24 h. Upon treatment of isolated hepatocytes with Hb alone, there was an increase in the nuclear accumulation of AhR and AhR-dependent luciferase activity. Furthermore, Hb potentiated the TCDD-induced AhR-dependent luciferase activity. Importantly, when isolated hepatocytes were treated for 5h with As(III) in the presence of TCDD and the medium was then replaced with new medium containing Hb, there was potentiation of the TCDD-mediated effect. Taken together, these results demonstrate for the first time that there is a differential modulation of the TCDD-mediated induction of Cyp1a1 by As(III) in C57BL/6 mouse livers and isolated hepatocytes. Thus, this study implicates Hb as an in vivo-specific modulator., (Copyright © 2013 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
42. Cytochrome P450 epoxygenase metabolite, 14,15-EET, protects against isoproterenol-induced cellular hypertrophy in H9c2 rat cell line.
- Author
-
Tse MM, Aboutabl ME, Althurwi HN, Elshenawy OH, Abdelhamid G, and El-Kadi AO
- Subjects
- 8,11,14-Eicosatrienoic Acid pharmacology, Animals, Cardiomegaly chemically induced, Cell Line, Cytochrome P-450 Enzyme System genetics, Epoxide Hydrolases genetics, Gene Expression Regulation drug effects, Hydroxyeicosatetraenoic Acids toxicity, Myocytes, Cardiac pathology, RNA, Messenger metabolism, Rats, Real-Time Polymerase Chain Reaction, Time Factors, 8,11,14-Eicosatrienoic Acid analogs & derivatives, Cardiomegaly prevention & control, Isoproterenol toxicity, Myocytes, Cardiac drug effects
- Abstract
We have previously shown that isoproterenol-induced cardiac hypertrophy causes significant changes to cytochromes P450 (CYPs) and soluble epoxide hydrolase (sEH) gene expression. Therefore, in this study, we examined the effect of isoproterenol in H9c2 cells, and the protective effects of 14,15-EET against isoproterenol-induced cellular hypertrophy. Isoproterenol was incubated with H9c2 cells for 24 and 48 h. To determine the protective effects of 14,15-EET, H9c2 cells were incubated with isoproterenol in the absence and presence of 14,15-EET. Thereafter, the expression of hypertrophic markers and different CYP genes were determined by real time-PCR. Our results demonstrated that isoproterenol significantly increased the expression of hypertrophic marker, atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP), parallel to a significant increase in cell surface area. Also, isoproterenol increased the mRNA expression of CYP1A1, CYP1B1, CYP2J3, CYP4F4 and CYP4F5, as well as the gene encoding sEH, EPHX2. On other hand, 14,15-EET significantly attenuated the isoproterenol-mediated induction of ANP, BNP, CYP1A1, CYP2J3, CYP4F4, CYP4F5 and EPHX2. Moreover 14,15-EET prevented the isoproterenol-mediated increase in cell surface area. Interestingly, 20-hydroxyeicosatetraenoic acid (20-HETE) treatment caused similar effects to that of isoproterenol treatment and induced cellular hypertrophy in H9c2 cells. In conclusion, isoproterenol induces cellular hypertrophy and modulates the expression of CYPs and EPHX2 in H9c2 cells. Furthermore, 14,15-EET exerts a protective effect against isoproterenol-induced cellular hypertrophy whereas, 20-HETE induced cellular hypertrophy in H9c2 cells., (Copyright © 2013 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
43. Role of cytochrome P450-mediated arachidonic acid metabolites in the pathogenesis of cardiac hypertrophy.
- Author
-
Alsaad AM, Zordoky BN, Tse MM, and El-Kadi AO
- Subjects
- Animals, Cardiomegaly pathology, Humans, Hydroxyeicosatetraenoic Acids metabolism, Signal Transduction, Arachidonic Acid metabolism, Cardiomegaly metabolism, Cytochrome P-450 Enzyme System metabolism
- Abstract
A plethora of studies have demonstrated the expression of cytochrome P450 (CYP) and soluble epoxide hydrolase (sEH) enzymes in the heart and other cardiovascular tissues. In addition, the expression of these enzymes is altered during several cardiovascular diseases (CVDs), including cardiac hypertrophy (CH). The alteration in CYP and sEH expression results in derailed CYP-mediated arachidonic acid (AA) metabolism. In animal models of CH, it has been reported that there is an increase in 20-hydroxyeicosatetraenoic acid (20-HETE) and a decrease in epoxyeicosatrienoic acids (EETs). Further, inhibiting 20-HETE production by CYP ω-hydroxylase inhibitors and increasing EET stability by sEH inhibitors have been proven to protect against CH as well as other CVDs. Therefore, CYP-mediated AA metabolites 20-HETE and EETs are potential key players in the pathogenesis of CH. Some studies have investigated the molecular mechanisms by which these metabolites mediate their effects on cardiomyocytes and vasculature leading to pathological CH. Activation of several intracellular signaling cascades, such as nuclear factor of activated T cells, nuclear factor kappa B, mitogen-activated protein kinases, Rho-kinases, Gp130/signal transducer and activator of transcription, extracellular matrix degradation, apoptotic cascades, inflammatory cytokines, and oxidative stress, has been linked to the pathogenesis of CH. In this review, we discuss how 20-HETE and EETs can affect these signaling pathways to result in, or protect from, CH, respectively. However, further understanding of these metabolites and their effects on intracellular cascades will be required to assess their potential translation to therapeutic approaches for the prevention and/or treatment of CH and heart failure.
- Published
- 2013
- Full Text
- View/download PDF
44. The role of aryl hydrocarbon receptor signaling pathway in cardiotoxicity of acute lead intoxication in vivo and in vitro rat model.
- Author
-
Ansari MA, Maayah ZH, Bakheet SA, El-Kadi AO, and Korashy HM
- Subjects
- Animals, Blotting, Western, Creatine Kinase metabolism, Cytochrome P-450 CYP1A1 metabolism, Heart Diseases enzymology, Heart Diseases metabolism, Histocytochemistry, L-Lactate Dehydrogenase metabolism, Male, Myocardium enzymology, Myocytes, Cardiac, Myosin Heavy Chains genetics, Myosin Heavy Chains metabolism, Natriuretic Peptide, Brain genetics, Natriuretic Peptide, Brain metabolism, RNA, Messenger chemistry, RNA, Messenger genetics, Random Allocation, Rats, Rats, Wistar, Real-Time Polymerase Chain Reaction, Receptors, Aryl Hydrocarbon antagonists & inhibitors, Resveratrol, Signal Transduction drug effects, Stilbenes pharmacology, Heart drug effects, Heart Diseases chemically induced, Lead toxicity, Myocardium metabolism, Receptors, Aryl Hydrocarbon metabolism
- Abstract
Lead (Pb(2+)) is a naturally occurring systemic toxicant heavy metal that affects several organs in the body including the kidneys, liver, and central nervous system. However, Pb(2+)-induced cardiotoxicity has never been investigated yet and the exact mechanism of Pb(2+) associated cardiotoxicity has not been studied. The current study was designed to investigate the potential effect of Pb(2+) to induce cardiotoxicity in vivo and in vitro rat model and to explore the molecular mechanisms and the role of aryl hydrocarbon receptor (AhR) and regulated gene, cytochrome P4501A1 (CYP1A1), in Pb(2+)-mediated cardiotoxicity. For these purposes, Wistar albino rats were treated with Pb(2+) (25, 50 and 100mg/kg, i.p.) for three days and the effects on physiological and histopathological parameters of cardiotoxicity were determined. At the in vitro level, rat cardiomyocyte H9c2 cell lines were incubated with increasing concentration of Pb(2+) (25, 50, and 100 μM) and the expression of hypertrophic genes, α- and β-myosin heavy chain (α-MHC and β-MHC), brain Natriuretic Peptide (BNP), and CYP1A1 were determined at the mRNA and protein levels using real-time PCR and Western blot analysis, respectively. The results showed that Pb(2+) significantly induced cardiotoxicity and heart failure as evidenced by increase cardiac enzymes, lactate dehydrogenase and creatine kinase and changes in histopathology in vivo. In addition, Pb(2+) treatment induced β-MHC and BNP whereas inhibited α-MHC mRNA and protein levels in vivo in a dose-dependent manner. In contrast, at the in vitro level, Pb(2+) treatment induced both β-MHC and α-MHC mRNA levels in time- and dose-dependent manner. Importantly, these changes were accompanied with a proportional increase in the expression of CYP1A1 mRNA and protein expression levels, suggesting a role for the CYP1A1 in cardiotoxicity. The direct evidence for the involvement of CYP1A1 in the induction of cardiotoxicity by Pb(2+) was evidenced by the ability of AhR antagonist, resveratrol, to significantly inhibit the Pb(2+)-modulated effect on β-MHC and α-MHC mRNAs. It was concluded that acute lead exposure induced cardiotoxicity through AhR/CYP1A1-mediated mechanism., (Copyright © 2013 Elsevier Ireland Ltd. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
45. Soluble epoxide hydrolase inhibitor, TUPS, protects against isoprenaline-induced cardiac hypertrophy.
- Author
-
Althurwi HN, Tse MM, Abdelhamid G, Zordoky BN, Hammock BD, and El-Kadi AO
- Subjects
- Animals, Atrial Natriuretic Factor metabolism, Body Weight drug effects, Cardiomegaly chemically induced, Cell Line, Cytochrome P-450 Enzyme System genetics, Drug Antagonism, Epoxide Hydrolases genetics, Epoxide Hydrolases metabolism, Gene Expression Regulation, Heart drug effects, Humans, Kidney metabolism, Liver metabolism, Natriuretic Peptide, Brain metabolism, Rats, Rats, Sprague-Dawley, Real-Time Polymerase Chain Reaction, Cardiomegaly prevention & control, Cardiotonic Agents administration & dosage, Cytochrome P-450 Enzyme System metabolism, Epoxide Hydrolases antagonists & inhibitors, Isoproterenol adverse effects, Phenylurea Compounds administration & dosage, Piperidines administration & dosage
- Abstract
Background and Purpose: We have previously shown that isoprenaline-induced cardiac hypertrophy causes significant changes in the expression of cytochromes P450 (CYP) and soluble epoxide hydrolase (sEH) genes. Therefore, it is important to examine whether the inhibition of sEH by 1-(1-methanesulfonyl-piperidin-4-yl)-3-(4-trifluoromethoxy-phenyl)-urea (TUPS) will protect against isoprenaline-induced cardiac hypertrophy., Experimental Approach: Male Sprague-Dawley rats were treated with TUPS (0.65 mg kg(-1) day(-1), p.o.), isoprenaline (5 mg kg(-1) day(-1), i.p.) or the combination of both. In vitro H9c2 cells were treated with isoprenaline (100 μM) in the presence and absence of either TUPS (1 μM) or 11,12 EET (1 μM). The expression of hypertrophic, fibrotic markers and different CYP genes were determined by real-time PCR., Key Results: Isoprenaline significantly induced the hypertrophic, fibrotic markers as well as the heart to body weight ratio, which was significantly reversed by TUPS. Isoprenaline also caused an induction of CYP1A1, CYP1B1, CYP2B1, CYP2B2, CYP4A3 and CYP4F4 gene expression and TUPS significantly inhibited this isoprenaline-mediated effect. Moreover, isoprenaline significantly reduced 5,6-, 8,9-, 11,12- and 14,15-EET and increased their corresponding 8,9-, 11,12- and 14,15-dihydroxyeicosatrienoic acid (DHET) and the 20-HETE metabolites. TUPS abolished these isoprenaline-mediated changes in arachidonic acid (AA) metabolites. In H9c2 cells, isoprenaline caused a significant induction of ANP, BNP and EPHX2 mRNA levels. Both TUPS and 11,12-EET significantly decreased this isoprenaline-mediated induction of ANP, BNP and EPHX2., Conclusions and Implications: TUPS partially protects against isoprenaline-induced cardiac hypertrophy, which confirms the role of sEH and CYP enzymes in the development of cardiac hypertrophy., (© 2012 The Authors. British Journal of Pharmacology © 2012 The British Pharmacological Society.)
- Published
- 2013
- Full Text
- View/download PDF
46. Murine atrial HL-1 cell line is a reliable model to study drug metabolizing enzymes in the heart.
- Author
-
Elshenawy OH, Anwar-Mohamed A, Abdelhamid G, and El-Kadi AO
- Subjects
- Animals, Arachidonic Acid metabolism, Cell Line, Cytochrome P-450 CYP2J2, Enzyme Induction drug effects, Enzymes metabolism, Heart Atria cytology, Male, Mice, Mice, Inbred C57BL, Models, Biological, RNA, Messenger metabolism, Receptors, Cytoplasmic and Nuclear metabolism, Reproducibility of Results, Cytochrome P-450 Enzyme System metabolism, Gene Expression Regulation, Enzymologic, Heart Atria enzymology, Myocardium enzymology
- Abstract
HL-1 cells are currently the only cells that spontaneously contract while maintaining a differentiated cardiac phenotype. Thus, our objective was to examine murine HL-1 cells as a new in vitro model to study drug metabolizing enzymes. We examined the expression of cytochrome P450s (Cyps), phase II enzymes, and nuclear receptors and compared their levels to mice hearts. Our results demonstrated that except for Cyp4a12 and Cyp4a14 all Cyps, phase II enzymes: glutathione-S-transferases (Gsts), heme oxygenase-1 (HO-1), and NAD(P)H: quinone oxidoreductase (Nqo1), nuclear receptors: aryl hydrocarbon receptor (AhR), constitutive androstane receptor (CAR), pregnane X receptor (PXR), and peroxisome proliferator activated receptor (PPAR-alpha) were all constitutively expressed in HL-1 cells. Cyp2b19, Cyp2c29, Cyp2c38, Cyp2c40, and Cyp4f16 mRNA levels were higher in HL-1 cells compared to mice hearts. Cyp2b9, Cyp2c44, Cyp2j9, Cyp2j11, Cyp2j13, Cyp4f13, Cyp4f15 mRNA levels were expressed to the same extent to that of mice hearts. Cyp1a1, Cyp1a2, Cyp1b1, Cyp2b10, Cyp2d10, Cyp2d22, Cyp2e1, Cyp2j5, Cyp2j6, Cyp3a11, Cyp4a10, and Cyp4f18 mRNA levels were lower in HL-1 cells compared to mice hearts. Moreover, 3-methylcholanthrene induced Cyp1a1 while fenofibrate induced Cyp2j9 and Cyp4f13 mRNA levels in HL-1 cells. Examining the metabolism of arachidonic acid (AA) by HL-1 cells, our results demonstrated that HL-1 cells metabolize AA to epoxyeicosatrienoic acids, dihydroxyeicosatrienoic acids, and 20-hydroxyeicosatetraenoic acids. In conclusion, HL-1 cells provide a valuable in vitro model to study the role of Cyps and their associated AA metabolites in addition to phase II enzymes in cardiovascular disease states., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
47. Mercury modulates the cytochrome P450 1a1, 1a2 and 1b1 in C57BL/6J mice: in vivo and in vitro studies.
- Author
-
Amara IE, Anwar-Mohamed A, Abdelhamid G, and El-Kadi AO
- Subjects
- Animals, Blotting, Western, Heme Oxygenase-1 antagonists & inhibitors, Heme Oxygenase-1 genetics, Hemoglobins metabolism, Hepatocytes drug effects, Hepatocytes enzymology, Hepatocytes metabolism, Liver enzymology, Liver metabolism, Male, Metalloporphyrins toxicity, Mice, Mice, Inbred C57BL, RNA chemistry, RNA genetics, Real-Time Polymerase Chain Reaction, Cytochrome P-450 CYP1A1 metabolism, Heme Oxygenase-1 metabolism, Liver drug effects, Mercury toxicity, Polychlorinated Dibenzodioxins toxicity
- Abstract
In the current study C57BL/6J mice were injected intraperitoneally with Hg(2+) in the absence and presence of TCDD. After 6 and 24h the liver was harvested and the expression of Cyps was determined. In vitro, isolated hepatocytes were incubated with TCDD in the presence and absence of Hg(2+). At the in vivo level, Hg(2+) significantly decreased the TCDD-mediated induction of Cyps at 6h while potentiating their levels at 24h. In vitro, Hg(2+) significantly inhibited the TCDD-mediated induction of Cyp1a1 in a concentration- and time-dependent manner. Interestingly, Hg(2+) increased the serum hemoglobin (Hb) levels in mice treated for 24h. Upon treatment of isolated hepatocytes with Hb alone, there was an increase in the AhR-dependent luciferase activity with a subsequent increase in Cyp1a1 protein and catalytic activity levels. Importantly, when hepatocytes were treated for 2h with Hg(2+) in the presence of TCDD, then the medium was replaced with new medium containing Hb, there was potentiation of the TCDD-mediated effect. In addition, Hg(2+) increased heme oxygenase-1 (HO-1) mRNA, which coincided with a decrease in the Cyp1a1 activity level. When the competitive HO-1 inhibitor, tin mesoporphyrin was applied to the hepatocytes there was a partial restoration of Hg(2+)-mediated inhibition of Cyp1a1 activity. In conclusion, we demonstrate for the first time that there is a differential modulation of the TCDD-mediated induction of Cyp1a1 by Hg(2+) in C57BL/6J mice livers and isolated hepatocytes. Moreover, this study implicates Hb as an in vivo specific modulator of Cyp1 family., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
48. Determination of the dominant arachidonic acid cytochrome p450 monooxygenases in rat heart, lung, kidney, and liver: protein expression and metabolite kinetics.
- Author
-
El-Sherbeni AA, Aboutabl ME, Zordoky BN, Anwar-Mohamed A, and El-Kadi AO
- Subjects
- Animals, Cytochrome P-450 CYP2J2, Hydroxylation, Kinetics, Male, Rats, Rats, Sprague-Dawley, Arachidonic Acid metabolism, Cytochrome P-450 Enzyme System metabolism, Kidney enzymology, Liver enzymology, Lung enzymology, Myocardium enzymology
- Abstract
Cytochrome P450 (P450)-derived arachidonic acid (AA) metabolites serve pivotal physiological roles. Therefore, it is important to determine the dominant P450 AA monooxygenases in different organs. We investigated the P450 AA monooxygenases protein expression as well as regioselectivity, immunoinhibition, and kinetic profile of AA epoxygenation and hydroxylation in rat heart, lung, kidney, and liver. Thereafter, the predominant P450 epoxygenases and P450 hydroxylases in these organs were characterized. Microsomes from heart, lung, kidney, and liver were incubated with AA. The protein expression of CYP2B1/2, CYP2C11, CYP2C23, CYP2J3, CYP4A1/2/3, and CYP4Fs in the heart, lung, kidney, and liver were determined by Western blot analysis. The levels of AA metabolites were determined by liquid chromatography-electrospray ionization mass spectroscopy. This was followed by determination of regioselectivity, immunoinhibition effect, and the kinetic profile of AA metabolism. AA was metabolized to epoxyeicosatrienoic acids and 19- and 20-hydroxyeicosatetraenoic acid in the heart, lung, kidney, and liver but with varying metabolic activities and regioselectivity. Anti-P450 antibodies were found to differentially inhibit AA epoxygenation and hydroxylation in these organs. Our data suggest that the predominant epoxygenases are CYP2C11, CYP2B1, CYP2C23, and CYP2C11/CYP2C23 for the heart, lung, kidney, and liver, respectively. On the other hand, CYP4A1 is the major ω-hydroxylase in the heart and kidney; whereas CYP4A2 and/or CYP4F1/4 are probably the major hydroxlases in the lung and liver. These results provide important insights into the activities of P450 epoxygenases and P450 hydroxylases-mediated AA metabolism in different organs and their associated P450 protein levels.
- Published
- 2013
- Full Text
- View/download PDF
49. Effect of rat serum lipoproteins on mRNA levels and amiodarone metabolism by cultured primary rat hepatocytes.
- Author
-
Brocks DR, Hamdy DA, Ben-Eltriki M, Patel JP, and El-Kadi AO
- Subjects
- ATP Binding Cassette Transporter, Subfamily B genetics, ATP Binding Cassette Transporter, Subfamily B metabolism, Alcohol Oxidoreductases genetics, Alcohol Oxidoreductases metabolism, Animals, Cells, Cultured, Cytochrome P-450 CYP1A1 genetics, Cytochrome P-450 CYP1A1 metabolism, Cytochrome P-450 CYP3A genetics, Cytochrome P-450 CYP3A metabolism, Cytochrome P450 Family 2, Disease Models, Animal, Half-Life, Hyperlipidemias genetics, Male, Membrane Proteins genetics, Membrane Proteins metabolism, Metabolic Clearance Rate, Organic Anion Transporters genetics, Organic Anion Transporters metabolism, Primary Cell Culture, Rats, Rats, Sprague-Dawley, Receptors, LDL genetics, Receptors, LDL metabolism, Time Factors, Amiodarone metabolism, Hepatocytes metabolism, Hyperlipidemias blood, Lipoproteins blood, RNA, Messenger metabolism
- Abstract
Hyperlipidemia can significantly increase amiodarone (AM) in vivo liver uptake and decrease its velocity of microsomal metabolism. Here, hepatocytes isolated from normolipidemic (NL) and hyperlipidemic rats were incubated with AM in the presence or absence of diluted NL or hyperlipidemic serum. The serum was added either as preincubation before drug, or concurrently with drug; incubations without rat serum were used as controls. The hepatocyte levels of mRNA for several proteins and enzymes were also measured. Disappearance of AM was seen up to 72 h. There was little difference between hepatocytes from NL or hyperlipidemic animals in intrinsic clearance (CL(int) ) of AM. The effect of hyperlipidemic rat serum, either before or with AM, was profound, causing a significant reduction in the CL(int) . Reductions were seen in mRNA for cytochrome P450 1A1, 3A2, and 2D1, some transporters, and low-density lipoprotein receptors after exposure of hepatocytes to lipoprotein-rich sera. In conclusion, exposure of isolated hepatocytes to hyperlipidemic serum caused decreases in AM CL(int) and lower mRNA levels for some proteins involved in the uptake and metabolism of AM. When coincubated with serum, an additional effect of increased binding to lipoproteins seemed to further contribute to a reduced CL of AM., (Copyright © 2012 Wiley Periodicals, Inc.)
- Published
- 2013
- Full Text
- View/download PDF
50. Harmine and harmaline downregulate TCDD-induced Cyp1a1 in the livers and lungs of C57BL/6 mice.
- Author
-
El Gendy MA and El-Kadi AO
- Subjects
- Animals, Environmental Pollutants, Gene Expression Regulation, Enzymologic, Liver metabolism, Lung metabolism, Male, Mice, Mice, Inbred C57BL, Microsomes drug effects, Cytochrome P-450 CYP1A1 metabolism, Harmaline pharmacology, Harmine pharmacology, Liver drug effects, Lung drug effects, Polychlorinated Dibenzodioxins pharmacology
- Abstract
We previously demonstrated that Peganum harmala L. extract and its main active constituents, harmine and harmaline inhibit the 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-mediated induction of the carcinogen-activating enzyme, Cyp1a1, in vitro. However, the effect of both alkaloids on Cyp1a1 in vivo has not been investigated. Therefore, the aim of this study is to examine the effect of harmine and harmaline on TCDD-mediated induction of Cyp1a1 in mice livers and lungs. C57BL/6 male mice were distributed into four groups (n = 6). First group received vehicle, while the second group received TCDD (i.p.). The third and fourth groups received either harmine or harmaline (i.p.) × 3 times along with TCDD one time with the mid dose of harmine and harmaline. All mice were sacrificed after 14 h from TCDD injection, and livers and lungs were isolated. The effect of harmine and harmaline on TCDD-mediated induction of Cyp1a1 mRNA, protein, and activity levels was determined using real-time PCR, Western blot analysis, and 7-ethoxyresurofin as a substrate, respectively. Our results showed that harmine and harmaline significantly decreased the TCDD-mediated induction of Cyp1a1 in both the livers and lungs. We concluded that harmine and harmaline are promising candidate to inhibit TCDD-mediated induction of Cyp1a1 in mice hepatic and extrahepatic tissues.
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.