Elsevier

Toxicology Letters

Volume 179, Issue 2, 30 June 2008, Pages 71-77
Toxicology Letters

Tumor necrosis factor alpha partially contributes to lipopolysaccharide-induced downregulation of CYP3A in fetal liver: Its repression by a low dose LPS pretreatment

https://doi.org/10.1016/j.toxlet.2008.04.005Get rights and content

Abstract

With embryonic development, fetal hepatocytes gradually express various types of cytochromes P450 (CYPs) that play a key role in the detoxification of xenobiotics. In the present study, we showed that maternal lipopolysaccharide (LPS) exposure downregulated cyp3a11 mRNA and CYP3A protein in fetal liver. The increased level of TNF-α protein in fetal liver, transferred from either the maternal circulation or amniotic fluid, seems to be associated with LPS-induced downregulation of cyp3a11 mRNA and CYP3A protein in fetal liver. Interestingly, a low dose LPS (10 μg/kg) pretreatment attenuated LPS-induced downregulation of cyp3a11 mRNA and CYP3A protein in fetal liver. Correspondingly, a low dose LPS pretreatment attenuated LPS-induced downregulation of pregnane X receptor (pxr) in fetal liver. Additional experiment showed that a low dose LPS pretreatment decreased the level of TNF-α in maternal serum and amniotic fluid and counteracted LPS-induced expression of TNF-α mRNA in maternal liver and placenta. Although a low dose LPS pretreatment alleviated LPS-induced increase in TNF-α in fetal liver, it had little effect on TNF-α mRNA in fetal liver. These results suggest that a low dose LPS pretreatment protects fetuses against LPS-induced downregulation of hepatic cyp3a11 and pxr expression through the repression of maternally sourced TNF-α production.

Introduction

The liver is the major organ of amino acid and lipid metabolism, gluconeogenesis, synthesis of serum proteins, and xenobiotic detoxification. The fetal liver functions as the major hematopoietic organ in the mid- to late fetal stage (Dzierzak and Medvinsky, 1995, Hardy and Hayakawa, 2001). With embryonic development, fetal hepatocytes gradually express various types of cytochromes P450 (CYPs) that play a key role in the detoxification of drug or other xenobiotics (Hulla and Juchau, 1989, Krauer and Dayer, 1991, Rich and Boobis, 1997, de Wildt et al., 1999, Hines and McCarver, 2002). CYP3A is a member of the cytochrome P-450 monooxygenase superfamily. In human, CYP3A4 and CYP3A5 account for 30–40% of the total cytochrome P450 in the adult liver, which is responsible for the oxidative metabolism of numerous clinically used drugs and toxicants (Goodwin et al., 2002). Although CYP3A4 and CYP3A5 are not detectable in fetal liver, fetal hepatocytes express CYP3A7 as early as gestational day 50–60 (Stevens et al., 2003). In mice, cyp3a11 and cyp3a13 are major members of cyp3a subfamily in the adult liver (Stevens et al., 2003, Anakk et al., 2003). In the developing mouse embryo, the amount of cyp3a11 and cyp3a13 expression gradually increases with the advancement of embryonic development (Choudhary et al., 2003).

Lipopolysaccharide (LPS) is a toxic component of cell walls of Gram-negative bacteria and is widely present in the digestive tracts of humans and animals. Humans are constantly exposed to low levels of LPS through infection. Gastrointestinal distress and alcohol drinking often increase permeability of LPS from gastrointestinal tract into blood (Zhou et al., 2003). Numerous studies have demonstrated that LPS decreases the levels of CYP3A in livers of rats and mice (Morgan et al., 2002, Aitken et al., 2006). On the other hand, maternal LPS exposure results in intra-uterine fetal death and intra-uterine growth retardation in animals (Xu et al., 2005a, Xu et al., 2006, Xu et al., 2007, Chen et al., 2006). Recently, we found that the expression of hepatic cyp3a11 mRNA was significantly decreased in fetuses from dams that were exposed to LPS during pregnancy (Xu et al., 2005b). In the present study, we investigated the role of TNF-α in LPS-induced downregulation of CYP3A in fetal liver. Our results indicate that the increased level of TNF-α in fetal liver, possibly sourced from maternal circulation and perhaps from amniotic fluid, contributes, at least partially, to LPS-induced downregulation of CYP3A in fetal liver. A low dose LPS pretreatment protects fetuses against LPS-induced downregulation of CYP3A through inhibiting the release of maternally sourced TNF-α.

Section snippets

Chemicals

Lipopolysaccharide (Escherichia coli LPS, serotype 0127:B8) and pentoxifylline (PTX) were purchased from Sigma Chemical Co. (St. Louis, MO). All other reagents were from Sigma or as indicated in the specified methods.

Animals and treatments

The ICR mice (8–10-week-old; male mice: 28–30 g; female mice: 24–26 g) were purchased from Beijing Vital River (Beijing, China). The animals were allowed free access to food and water at all times and were maintained on a 12-h light:12-h dark cycle in a controlled temperature (20–25 

Ontogeny of CYP3A in fetal liver

The ontogeny of cyp3a11 mRNA and CYP3A protein in mouse fetal liver is presented in Fig. 1. The level of cyp3a11 mRNA and CYP3A protein was very low in fetal liver from gd 14 to 16. On gd 18, the expression level of CYP3A in fetal liver was about 25% of that in adult liver, whereas the level of cyp3a11 mRNA in fetal liver was about 55% of that in adult liver. The level of cyp3a11 mRNA and CYP3A protein was greatly increased after birth. On postnatal day 4, the level of cyp3a11 mRNA and CYP3A

Discussion

CYP3A is a member of the cytochrome P-450 monooxygenase superfamily, which is responsible for the oxidative metabolism of numerous clinically used drugs (Goodwin et al., 2002). The present study showed that the level of cyp3a11 mRNA and CYP3A protein was very low in fetal liver from gd 14 to 16. The amount of CYP3A gradually increased with the advancement of embryonic development. On gd 18, CYP3A protein was about 25% of that in adult liver, whereas cyp3a11 mRNA in fetal liver was about 55% of

Acknowledgements

This project was supported by National Natural Science Foundation of China (30371667, 30572223, 30671786) and Anhui Provincial Natural Science Foundation (050430714).

References (38)

  • D.X. Xu et al.

    Effects of low-dose lipopolysaccharide (LPS) pretreatment on LPS-induced intra-uterine fetal death and preterm labor

    Toxicology

    (2007)
  • Z. Zhou et al.

    A critical involvement of oxidative stress in acute alcohol-induced hepatic TNF-alpha production

    Am. J. Pathol.

    (2003)
  • Z. Abdel-Razzak et al.

    Cytokines down-regulate expression of major cytochrome P-450 enzymes in adult human hepatocytes in primary culture

    Mol. Pharmacol.

    (1993)
  • A.E. Aitken et al.

    Regulation of drug-metabolizing enzymes and transporters in inflammation

    Annu. Rev. Pharmacol. Toxicol.

    (2006)
  • A.E. Aitken et al.

    Gene-specific effects of inflammatory cytokines on cytochrome P4502C, 2B6 and 3A4 mRNA levels in human hepatocytes

    Drug Metab. Dispos.

    (2007)
  • S. Anakk et al.

    Genomic characterization and regulation of CYP3a13: role of xenobiotics and nuclear receptors

    FASEB J.

    (2003)
  • H. Ashdown et al.

    The role of cytokines in mediating effects of prenatal infection on the fetus: implications for schizophrenia

    Mol. Psychiatry

    (2006)
  • T. Ashino et al.

    Involvement of interleukin-6 and tumor necrosis factor alpha in CYP3A11 and 2C29 down-regulation by Bacillus Calmette-Guerin and lipopolysaccharide in mouse liver

    Drug Metab. Dispos.

    (2004)
  • Y.H. Chen et al.

    Melatonin protects against lipopolysaccharide-induced intra-uterine fetal death and growth retardation in mice

    J. Pineal Res.

    (2006)
  • Cited by (19)

    • Maternal-placental-fetal drug metabolism is altered by late gestation undernutrition in the pregnant ewe

      2022, Life Sciences
      Citation Excerpt :

      Despite this diversity, the majority of clinically prescribed drugs are metabolised by three distinct CYP isoenzymes: CYP3A, CYP1A2 and CYP2D6. Although the mRNA expression of CYP1A2, CYP2D6 and CYP3A have been reported in early- and late-gestation placentae of humans and other eutherian mammals and in the late gestation human fetal liver [20–24], the functional activity of these enzymes has not been fully investigated. Several factors contribute to dysfunction of CYP activity including altered concentrations of steroid hormones, increased reactive oxygen species (ROS) production, and abnormal levels of inflammatory cytokines [25].

    • Down-regulation of carboxylesterases 1 and 2 plays an important role in prodrug metabolism in immunological liver injury rats

      2015, International Immunopharmacology
      Citation Excerpt :

      Certain cytokines, especially IL-1β, IL-6 and TNF-α, are thought to down-regulate the expressions of many drug-metabolizing enzymes during inflammation and infection, with most studies focused on cytochrome P450 [30]. Abdel-Razzak et al. [30] found that IL-6 and TNF-α managed to inhibit the expressions of several CYP enzymes mainly by attenuating those of the upstream pathways of pregnane X receptor and constitutively activated receptor [31,32]. Pregnane X receptor and constitutively activated receptor are also the upstream pathways regulating carboxylesterases, which has been extensively characterized by numerous studies [33,34].

    View all citing articles on Scopus
    1

    These authors contributed equally to this work.

    View full text