Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Jerome M. Lasker is active.

Publication


Featured researches published by Jerome M. Lasker.


Archives of Biochemistry and Biophysics | 1989

Acetaminophen activation by human liver cytochromes P450IIE1 and P450IA2

Judy L. Raucy; Jerome M. Lasker; Charles S. Lieber; Martin Black

Acetaminophen (APAP), a widely used over-the-counter analgesic, is known to cause hepatotoxicity when ingested in large quantities in both animals and man, especially when administered after chronic ethanol consumption. Hepatotoxicity stems from APAP activation by microsomal P450 monooxygenases to a reactive metabolite that binds to tissue macromolecules, thereby initiating cellular necrosis. Alcohol consumption also causes the induction of P450IIE1, a liver microsomal enzyme that in reconstitution studies has proven to be an effective catalyst of APAP oxidation. Thus, elevated microsomal P450IIE1 levels could explain not only the known increase in APAP bioactivating activity of liver microsomes after prolonged ethanol ingestion but also the enhanced susceptibility to APAP toxicity. We therefore examined the role of P450IIE1 in human liver microsomal APAP activation. Liver microsomes from seven non-alcoholic subjects were found to convert 1 mM APAP to a reactive intermediate (detected as an APAP-cysteine conjugate by high-pressure liquid chromatography) at a rate of 0.25 +/- 0.1 nmol conjugate formed/min/nmol microsomal P450 (mean +/- SD), whereas at 10 mM, this rate increased to 0.73 +/- 0.2 nmol product/min/nmol P450. In a reconstituted system, purified human liver P450IIE1 catalyzed APAP activation at rates threefold higher than those obtained with microsomes whereas two other human P450s, P450IIC8 and P450IIC9, exhibited negligible APAP-oxidizing activity. Monospecific antibodies (IgG) directed against human P450IIE1 inhibited APAP activation in each of the human samples, with anti-P450IIE1 IgG-mediated inhibition averaging 52% (range = 30-78%) of the rates determined in the presence of control IgG. The ability of anti-P450IIE1 IgG to inhibit only one-half of the total APAP activation by microsomes suggests, however, that other P450 isozymes besides P450IIE1 contribute to bioactivation of this compound in human liver. Of the other purified P450 isozymes examined, a beta-naphthoflavone (BNF)-inducible hamster liver P450 promoted APAP activation at rates even higher than those obtained with human P450IIE1. The extensive APAP-oxidizing capacity of this hamster P450, designated P450IA2 based upon its similarity to rat P450d and rabbit form 4 in terms of NH2-terminal amino acid sequence, spectral characteristics, immunochemical properties, and inducibility by BNF, agrees with previous reports concerning the APAP substrate specificity of the rat and rabbit P450IA2 proteins.(ABSTRACT TRUNCATED AT 400 WORDS)


Critical Reviews in Toxicology | 1993

Bioactivation of Halogenated Hydrocarbons by Cytochrome P4502E1

Judy L. Raucy; James C. Kraner; Jerome M. Lasker

Numerous halogenated hydrocarbons of the alkane, alkene, and alkyne classes are metabolized by P450 enzymes to products that elicit cytotoxic and/or carcinogenic effects. Such halogenated hydrocarbons include anesthetics (e.g., halothane and enflurane) and industrial solvents (e.g., carbon tetrachloride, chloroform, and vinylidine chloride). Formation of reaction intermediates from these compounds occurs via P450-promoted dehalogenation, reduction, or reductive oxygenation, with certain hydrocarbons undergoing all three reaction types. Of the multiple forms of P450 present in liver microsomes, P4502E1 has been identified as the primary catalyst of hydrocarbon bioactivation in animals and, most likely, in humans as well. As hepatic concentrations of this P450 enzyme are highly inducible by ethanol and similar agents, prior exposure to 2E1-inducing compounds can play a pivotal role in halogenated hydrocarbon toxicity. Considering that metabolism governs the cytotoxicity and carcinogenicity of halogenated hydrocarbons, an understanding of the mechanism(s) underlying 2E1 induction in man becomes all the more important.


Journal of Biological Chemistry | 2000

Formation of 20-Hydroxyeicosatetraenoic Acid, a Vasoactive and Natriuretic Eicosanoid, in Human Kidney ROLE OF CYP4F2 AND CYP4A11

Jerome M. Lasker; W. Bill Chen; Imre Wolf; Barbara P. Bloswick; Patricia D. Wilson; Pnina K. Powell

20-Hydroxyeicosatetraenoic acid (20-HETE), an ω-hydroxylated arachidonic acid (AA) metabolite, elicits specific effects on kidney vascular and tubular function that, in turn, influence blood pressure control. The human kidneys capacity to convert AA to 20-HETE is unclear, however, as is the underlying P450 catalyst. Microsomes from human kidney cortex were found to convert AA to a single major product, namely 20-HETE, but failed to catalyze AA epoxygenation and midchain hydroxylation. Despite the monophasic nature of renal AA ω-hydroxylation kinetics, immunochemical studies revealed participation of two P450s, CYP4F2 and CYP4A11, since antibodies to these enzymes inhibited 20-HETE formation by 65.9 ± 17 and 32.5 ± 14%, respectively. Western blotting confirmed abundant expression of these CYP4 proteins in human kidney and revealed that other AA-oxidizing P450s, including CYP2C8, CYP2C9, and CYP2E1, were not expressed. Immunocytochemistry showed CYP4F2 and CYP4A11 expression in only the S2 and S3 segments of proximal tubules in cortex and outer medulla. Our results demonstrate that CYP4F2 and CYP4A11 underlie conversion of AA to 20-HETE, a natriuretic and vasoactive eicosanoid, in human kidney. Considering their proximal tubular localization, these P450 enzymes may partake in pivotal renal functions, including the regulation of salt and water balance, and arterial blood pressure itself.


Gastroenterology | 1990

Immunohistochemical localization of ethanol-inducible P450IIE1 in the rat alimentary tract

Masanori Shimizu; Jerome M. Lasker; Mikihiro Tsutsumi; Charles S. Lieber

To determine whether P450IIE1, a microsomal P450 enzyme inducible by ethanol in the liver, is also present and inducible in the alimentary tract, corresponding frozen tissue sections were prepared from rats pair-fed liquid diets containing 36% of total calories as either ethanol or carbohydrate (control) for 3 weeks. Immunohistochemical staining was performed using the peroxidase-antiperoxidase method after tissue sections were reacted with antibody against human P450IIE1. In control animals, immunoreactive P450IIE1 was detected only in duodenal and jejunal villous cells. After ethanol treatment, the content of P450IIE1 increased in duodenal and jejunal villi, and the enzyme was now also found in squamous epithelial cells of the cheek mucosa, tongue, esophagus, and forestomach, and in surface epithelium of the proximal colon. P450IIE1 was neither expressed nor induced by alcohol in the epithelium of stomach fundic and antral mucosa, ileum, distal colon, and rectum. When considered together with the xenobiotic activation properties of P450IIE1, these results may partly explain why alcohol abuse is a risk factor for cellular damage or cancer or both in those alimentary tract tissues in which P450IIE1 is inducible by chronic ethanol intake.


Methods in Enzymology | 1991

Isolation of P450 enzymes from human liver

Judy L. Raucy; Jerome M. Lasker

Publisher Summary This chapter discusses the isolation of P450 enzymes from human liver. Considerable progress has been made in purifying and characterizing P450 enzymes from experimental animals. Although the information gathered has provided a foundation for the study of human P450 enzymes, there are limitations when extrapolating from experimental animals to humans. First, orthologous animal and human P450s differ in their catalytic specificity. Second, sexual bimorphism among P450 enzymes occurs in animals but probably not in humans. Experimental animals also give a biased view of differences among the human population, in that animals are not completely outbred and they consume defined diets. The genetically determined variations that exist in the structure and function of human P450 enzymes are rarely encountered in experimental animals.


Drug Metabolism and Disposition | 2004

CONVERSION OF THE HIV PROTEASE INHIBITOR NELFINAVIR TO A BIOACTIVE METABOLITE BY HUMAN LIVER CYP2C19

Vandana Hirani; Judy L. Raucy; Jerome M. Lasker

Antiretroviral therapy for human immunodeficiency virus (HIV) infection includes treatment with both reverse transcriptase inhibitors and protease inhibitors, which markedly suppress viral replication and circulating HIV RNA levels. Cytochrome P450 (P450) enzymes in human liver, chiefly CYP3A4, play a pivotal role in protease inhibitor biotransformation, converting these agents to largely inactive metabolites. However, the protease inhibitor nelfinavir (Viracept) is metabolized mainly to nelfinavir hydroxy-t-butylamide (M8), which exhibits potent antiviral activity, and to other minor products (termed M1 and M3) that are inactive. Since indirect evidence suggests that CYP2C19 underlies M8 formation, we examined the role of this inducible, polymorphic P450 enzyme in nelfinavir t-butylamide hydroxylation by human liver. Rates of microsomal M8 formation were 50.6 ± 28.3 pmol of product formed/min/nmol P450 (n = 5 subjects), whereas kinetic analysis of the reaction revealed a KM of 21.6 μM and a Vmax of 24.6 pmol/min/nmol P450. In reconstituted systems, CYP2C19 catalyzed nelfinavir t-butylamide hydroxylation at a turnover rate of 2.2 min-1, whereas CYP2C9, CYP2C8, and CYP3A4 were inactive toward nelfinavir. Polyclonal anti-CYP2C9 (cross-reactive with CYP2C19) and monoclonal anti-CYP2C19 completely inhibited microsomal M8 production, whereas monoclonal CYP2C9 and polyclonal CYP3A4 antibodies were without effect. Similarly, the CYP2C19 substrate omeprazole strongly inhibited (75%) hepatic nelfinavir t-butylamide hydroxylation at a concentration of only 12.5 μM. Our study shows that CYP2C19 underlies formation in human liver of M8, a bioactive nelfinavir metabolite. The inducibility of CYP2C19 by agents (e.g., rifampicin) often taken concurrently with nelfinavir, together with this P450s known polymorphic nature, may thus be important determinants of nelfinavirs antiviral potency.


Biochemical Pharmacology | 1987

Characterization of the cytochrome P-450 monooxygenase system of hamster liver microsomes: Effects of prior treatment with ethanol and other xenobiotics

C.Murray Ardies; Jerome M. Lasker; Charles S. Lieber

The cytochrome P-450 monooxygenase system of hamster liver microsomes and its response to prior treatment with ethanol and other xenobiotics have been examined. Male Syrian golden hamsters were administered ethanol (ETOH), phenobarbital (PB), 5,6-benzoflavone (BF) or isoniazid (INH). Each treatment resulted in a moderate increase (20-60%) in the specific content of liver microsomal cytochrome P-450 along with a unique hemeprotein ferrous carbonyl Soret maximum. Sodium dodecyl sulfate-polyacrylamide gel electrophoretic analysis of liver microsomes revealed distinctive changes in protein banding patterns in the cytochrome P-450 (45-60 kDa) region with each treatment. NADPH: cytochrome c reductase activity was increased by both PB and INH, whereas cytochrome b5 content was increased by INH only. Microsomal oxidation of ETOH and aniline p-hydroxylation (expressed per nmol cytochrome P-450) were enhanced dramatically by ETOH and INH, whereas PB and BF had no effect on these enzymatic activities. Both ETOH and INH also increased zoxazolamine 6-hydroxylation but, in contrast to other rodent species, this drug-metabolizing activity was decreased in hamster liver microsomes after treatment with either PB or BF. Microsomal benzphetamine N-demethylation was decreased by ETOH, INH and BF administration and was only modestly enhanced after treatment with PB. ETOH and INH had no effect on the O-deethylation of 7-ethoxycoumarin, and enzymatic activity increased by BF but decreased by PB. These results demonstrate that the cytochrome P-450-dependent monooxygenase system of hamster liver microsomes responds to treatment with ETOH and other xenobiotics in a manner that is quantitatively and, in certain respects, qualitatively different from that reported for the rat, rabbit, and mouse.


Journal of Lipid Research | 2008

Omega oxidation of 3-hydroxy fatty acids by the human CYP4F gene subfamily enzyme CYP4F11

Madhurima Dhar; Daniel W. Sepkovic; Vandana Hirani; Ronald P. Magnusson; Jerome M. Lasker

Long-chain 3-hydroxydicarboxylic acids (3-OHDCAs) are thought to arise via β-oxidation of the corresponding dicarboxylic acids (DCAs), although long-chain DCAs are neither readily transported into nor β-oxidized in mitochondria. We thus examined whether ω-hydroxylation of 3-hydroxy fatty acids (3-OHFAs), formed via incomplete mitochondrial oxidation, is a more likely pathway for 3-OHDCA production. NADPH-fortified human liver microsomes converted 3-hydroxystearate and 3-hydroxypalmitate to their ω-hydroxylated metabolites, 3,18-dihydroxystearate and 3,16-dihydroxypalmitate, respectively, as identified by GC-MS. Rates of 3,18-dihydroxystearate and 3,16-dihydroxypalmitate formation were 1.23 ± 0.5 and 1.46 ± 0.30 nmol product formed/min/mg protein, respectively (mean ± SD; n = 13). Polyspecific CYP4F antibodies markedly inhibited microsomal ω-hydroxylation of 3-hydroxystearate (68%) and 3-hydroxypalmitate (99%), whereas CYP4A11 and CYP2E1 antibodies had little effect. Upon reconstitution, CYP4F11 and, to a lesser extent, CYP4F2 catalyzed ω-hydroxylation of 3-hydroxystearate, whereas CYP4F3b, CYP4F12, and CYP4A11 exhibited negligible activity. CYP4F11 was the lone CYP4F/A enzyme that effectively oxidized 3-hydroxypalmitate. Kinetic parameters of microsomal 3-hydroxystearate metabolism were Km = 55 μM and Vmax = 8.33 min−1, whereas those for 3-hydroxypalmitate were Km = 56.4 μM and Vmax = 14.2 min−1. CYP4F11 kinetic values resembled those of native microsomes, with Km = 53.5 μM and Vmax = 13.9 min−1 for 3-hydroxystearate and Km = 105.8 μM and Vmax = 70.6 min−1 for 3-hydroxypalmitate. Our data show that 3-hydroxystearate and 3-hydroxypalmitate are converted to ω-hydroxylated 3-OHDCA precursors in human liver and that CYP4F11 is the predominant catalyst of this reaction. CYP4F11-promoted ω-hydroxylation of 3-OHFAs may modulate the disposition of these compounds in pathological states in which enhanced fatty acid mobilization or impairment of mitochondrial fatty acid β-oxidation increases circulating 3-OHFA levels.


Annals of the New York Academy of Sciences | 1987

The Microsomal Ethanol Oxidizing System and Its Interaction with Other Drugs, Carcinogens, and Vitamins

Charles S. Lieber; Jerome M. Lasker; J. Alderman; Maria Anna Leo

The interaction of ethanol with the oxidative drug-metabolizing enzymes present in liver microsomes results in a number of clinically significant side effects in the alcoholic. Following chronic ethanol consumption, the activity of the microsomal ethanol oxidizing system (MEOS) increases. This enhancement of MEOS activity is primarily due to the induction of a unique microsomal cytochrome P-450 isozyme, which has a high capacity for ethanol oxidation, as shown in reconstituted systems. Normally present in liver microsomes at low levels, this form of cytochrome P-450 increases dramatically after chronic ethanol intake in many species, including baboons. The in-vivo role of cytochrome P-450 in hepatic ethanol oxidation, especially following chronic ethanol consumption, has been conclusively demonstrated in deer-mice lacking liver ADH. Induction of microsomal cytochrome P-450 by ethanol is associated with the enhanced oxidation of other drugs as well, resulting in metabolic tolerance to these agents. There is also increased cytochrome P-450-dependent activation of known hepatotoxins such as carbon tetrachloride and acetaminophen, which may explain the greater susceptibility of alcoholics to the toxicity of industrial solvents and commonplace analgesics. In addition, the ethanol-inducible form of cytochrome P-450 has the highest capacity of all known P-450 isozymes for the activation of dimethylnitrosamine, a potent (and ubiquitous) carcinogen. Moreover, cytochrome P-450-catalyzed oxidation of retinol is accelerated in liver microsomes, which may contribute to the hepatic vitamin A depletion seen in alcoholics. In contrast to chronic ethanol consumption, acute ethanol intake inhibits the metabolism of other drugs via competition for shared microsomal oxidation pathways. Thus, the interplay between ethanol and liver microsomes has a profound impact on the way heavy drinkers respond to drugs, solvents, vitamins, and carcinogens.


Journal of Pharmacology and Experimental Therapeutics | 2011

Genistein, Resveratrol, and 5-Aminoimidazole-4-carboxamide-1-β-d-ribofuranoside Induce Cytochrome P450 4F2 Expression through an AMP-Activated Protein Kinase-Dependent Pathway

Mei-Hui Hsu; Üzen Savas; Jerome M. Lasker; Eric F. Johnson

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

Collaboration


Dive into the Jerome M. Lasker's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Charles S. Lieber

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Eric F. Johnson

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Vandana Hirani

Hackensack University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Imre Wolf

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Maria Anna Leo

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Pnina K. Powell

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Üzen Savas

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge