Regular Article
Effects of CYP1A2 on Disposition of 2,3,7,8-Tetrachlorodibenzo-p-dioxin, 2,3,4,7,8-Pentachlorodibenzofuran, and 2,2′,4,4′,5,5′-Hexachlorobiphenyl in CYP1A2 Knockout and Parental (C57BL/6N and 129/Sv) Strains of Mice

https://doi.org/10.1006/taap.1999.8720Get rights and content

Abstract

TCDD is the prototype and most potent member of the highly lipophilic polyhalogenated aromatic hydrocarbons (PHAHs), which are persistent and ubiquitous environmental contaminants. In both acute and subchronic animal studies, there is a specific accumulation of TCDD in liver greater than in adipose tissue. The inducible hepatic binding protein responsible for this hepatic sequestration of TCDD and its congeners has been shown by our laboratory to be CYP1A2 (J. J. Diliberto, D. Burgin, and L. S. Birnbaum, 1997, Biochem. Biophys. Res. Commun. 236, 431–433). The present study was conducted using knockout (KO) mice lacking expression of CYP1A2 (CYP1A2−/−) in order to investigate the role of CYP1A2 gene on the disposition of TCDD, 4-PeCDF (a dioxin-like PHAH), and PCB 153 (a nondioxin-like PCB) in KO (CYP1A2−/−) mice and age-matched parental mice strains (C57BL/6N: CYP1A2+/+, Ahb/b and 129/Sv: CYP1A2+/+, Ahd/d). Mice were dosed (25 μg [3H]TCDD/kg, 300 μg [14C]4-PeCDF/kg, or 35.8 mg [14C]PCB 153/kg bw in a corn oil vehicle) orally and terminated after 4 days. Residues of administered compounds in collected tissues and daily excreta were quantitated using 3H or 14C activity. Results demonstrated differential effects in disposition for the various treatments within the three genetically different groups of mice. In KO mice, TCDD, 4-PeCDF, and PCB 153 had very little hepatic localization of chemical, and the major depot was adipose tissue. In contrast, parental strains demonstrated hepatic sequestration of TCDD and 4-PeCDF, whereas disposition of PCB 153 in parental strains was similar to that in KO mice. Another difference between KO mice and parental strains was the enhanced urinary excretion of 4-PeCDF. This study demonstrates the importance of CYP1A2 in pharmacokinetic behavior and mechanistic issues for TCDD and related compounds.

References (76)

  • J.J. Diliberto et al.

    Role of CYP1A2 in hepatic sequestration of dioxin: Studies using CYP1A2 knock-out mice

    Biochem. Biophys. Res. Commun.

    (1997)
  • J.J. Diliberto et al.

    Comparison of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) disposition following pulmonary, oral, dermal, and parenteral exposures to rats

    Toxicol. Appl. Pharmacol.

    (1996)
  • F.M. Farin et al.

    Expression of cytochrome P450s and microsomal epoxide hydrolase in primary cultures of human umbilical vein endothelial cells

    Toxicol. Appl. Pharmacol.

    (1994)
  • F.J. Gonzalez et al.

    Xenobiotic receptor knockout mice

    Toxicol. Lett.

    (1995)
  • M. Gupta et al.

    Ah receptor-independent induction of CYP1A2 gene expression in genetically inbred mice

    Environ. Toxicol. Pharmacol.

    (1998)
  • J.T. Hamm et al.

    Methoxyresorufin: An inappropriate substrate for CYP1A2 in the mouse

    Biochem. Pharmacol.

    (1998)
  • J.A. Jackson et al.

    Estimation of octanol-water partition coefficients and correlation with dermal absorption for several polyhalogenated aromatic hydrocarbons

    Fundam. Appl. Toxicol.

    (1993)
  • A.A. Jensen

    Polychlorobiphenyls (PCBs), polychlorodibenzo-p-dioxins (PCDDs) and polychlorodibenzofurans (PCDFs) in human milk, blood and adipose tissue

    Sci. Total Environ.

    (1987)
  • L.B. Kedderis et al.

    Disposition of 2,3,7,8-tetrabromodibenzo-p-dioxin and 2,3,7,8-tetrachlorodibenzo-p-dioxin in the rat: Biliary excretion and induction of cytochromes CYP1A1 and CYP1A2

    Toxicol. Appl. Pharmacol.

    (1991)
  • L.B. Kedderis et al.

    A physiologically based pharmacokinetic model for 2,3,7,8-tetrabromodibenzo-p-dioxin (TBDD) in the rat: Tissue distribution and CYP1A induction

    Toxicol. Appl. Pharmacol.

    (1993)
  • F.G. King et al.

    Physiological model for the pharmacokinetics of 2,3,7,8-tetrachlorodibenzofuran in several species

    Toxicol. Appl. Pharmacol.

    (1983)
  • J. Kuroki et al.

    High affinity of 2,3,4,7,8-pentachlorodibenzofuran to cytochrome P-450 in the hepatic microsomes of rats

    Chemosphere

    (1986)
  • G.P. Lahvis et al.

    Ahr null alleles: Distinctive or different?

    Biochem. Pharmacol.

    (1998)
  • G. Liu et al.

    Role of cytochrome P450 1A2 in acetanilide 4-hydroxylation as determined with cDNA expression and monoclonal antibodies

    Arch. Biochem. Biophys.

    (1991)
  • W.M. McDonnell et al.

    Induction of cytochrome P450IA genes (CYP1A) by omeprazole in the human alimentary tract

    Gastroenterology

    (1992)
  • J.R. Olson et al.

    Tissue distribution, excretion, and metabolism of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) in the Golden Syrian Hamster

    Toxicol. Appl. Pharmacol.

    (1980)
  • L.C. Quattrochi et al.

    The human CYP1A2 gene and induction by 3-methylcholanthrene

    J. Biol. Chem.

    (1994)
  • J.C. Ramsey et al.

    The in vivo biotransformation of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) in the rat

    Toxicol. Appl. Pharmacol.

    (1982)
  • M.J. Santostefano et al.

    Subcellular localization of TCDD differs between the liver, lungs and kidneys after acute and subchronic exposure: Species/dose comparisons and possible mechanism

    Fundam. Appl. Toxicol.

    (1996)
  • T. Sawahata et al.

    Identification of metabolites of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) formed on incubation with isolated rat hepatocytes

    Biochem. Biophys. Res. Commun.

    (1982)
  • D. Sesardic et al.

    The inducibility and catalytic activity of cytochromes P450c (P4501A1) and P450d (P4501A2) in rat tissues

    Biochem. Pharmacol.

    (1990)
  • H. Thoma et al.

    Comparison of the polychlorinated dibenzo-p-dioxin and dibenzofuran in human tissue and human liver

    Chemosphere

    (1990)
  • A.P.J.M. van Birgelen et al.

    Interactive effects between 2,3,7,8-tetrachlorodibenzo-p-dioxin and 2,2′,4,4′,5,5′-hexachlorobiphenyl in female B6C3F1 mice: Tissue distribution and tissue-specific enzyme induction

    Fundam. Appl. Toxicol.

    (1996)
  • M. Van den Berg et al.

    Hepatic retention of PCDDs and PCDFs in C57B1/6 and DBA/2 mice

    Chemosphere

    (1989)
  • R. Voorman et al.

    Specific binding of polyhalogenated aromatic hydrocarbon inducers of cytochrome P-450d to the cytochrome and inhibition of its estradiol 2-hydroxylase activity

    Toxicol. Appl. Pharmacol.

    (1987)
  • H. Zaher et al.

    Effect of phenobarbital on hepatic CYP1A1 and CYP1A2 in the Ahr-null mouse

    Biochem. Pharmacol.

    (1998)
  • K. Abraham et al.

    Pharmacokinetics and biological activity of 2,3,7,8-tetrachlorodibenzo-p-dioxin: 1. Dose-dependent tissue distribution and induction of hepatic ethoxyresorufin O-deethylase in rats following a single injection

    Arch. Toxicol.

    (1988)
  • Cited by (100)

    • 2,3,4,7,8-Pentachlorodibenzofuran is far less potent than 2,3,7,8-tetrachlorodibenzo-p-dioxin in disrupting the pituitary-gonad axis of the rat fetus

      2014, Toxicology and Applied Pharmacology
      Citation Excerpt :

      PnCDF is known to accumulate in the liver of dams more abundantly than other dioxins (Chen et al., 2001), and it is highly likely that hepatic CYP1A2 plays a role in retaining PnCDF. This is supported by the observation that knocking out the mouse Cyp1a2 gene markedly reduces the hepatic content of PnCDF (Diliberto et al., 1999). From a study focusing on TCDD, maternal Cyp1a2 but not Cyp1a1/1b1 appears to protect fetuses from dioxin-produced disorders (Dragin et al., 2006).

    View all citing articles on Scopus

    This document has been reviewed in accordance with U.S. Environmental Protection Agency policy and approved for publication. Approval does not signify that the contents necessarily reflect the view and policies of the Agency nor does mention of trade names or commercial products constitute endorsement or recommendation for use.

    2

    To whom correspondence and reprint requests should be addressed at MD-74, PKB/ETD/NHEERL, U.S. EPA, Research Triangle Park, NC 27711.

    View full text