Research paper
Cytochromes CYP1A1 and CYP1B1 in the rat mammary gland: Cell-specific expression and regulation by polycyclic aromatic hydrocarbons and hormones

https://doi.org/10.1016/0303-7207(95)03668-WGet rights and content

Abstract

Cultured rat mammary cells express both CYP1A1 and CYP1B1 in response to polycyclic aromatic hydrocarbons (PAH) and 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) in a cell type-specific manner. The expression of each P450 was determined functionally (regioselective PAH metabolism), as apoprotein (immunoblots) and as mRNA (Northern hybridization). The epithelial rat mammary cells (RMEC) expressed CYP1A1, however only after PAH or TCDD treatment. CYP1B1 protein was scarcely detected in these induced RMEC but was surprisingly active as a participant in 7,12-dimethylbenz[a]anthracene (DMBA) metabolism shown through selective antibody inhibition (40% of total activity). CYP1B1 was selectively expressed in the stromal fibroblast population of rat mammary cells to the exclusion of CYP1A1. In the rat mammary fibroblasts (RMF), CYP1B1 protein and associated activity were each present at low levels constitutively and were highly induced by benz[a]anthracene (BA) to a greater extent than by TCDD (12- versus 6-fold). However, BA (10 μM) and TCDD (10 nM) stimulated the 5.2-kb CYP1B1-specific mRNA equally. These increases are consistent with the involvement of the aryl hydrocarbon (Ah) receptor in the transcription of the CYP1B1 gene and with the additional stabilization of CYP1B1 protein by BA, previously observed in embryo fibroblasts. Exactly this regulation of CYP1B1-dependent activity was seen in RMEC suggesting that this arises from exceptionally active CYP1B1 in a small proportion (5%) of residual RMF. The constitutive expression and PAH inducibility of CYP1B1 and CYP1A1 proteins in RMF and RMEC, respectively, were each substantially decreased (∼75%) by a hormonal mixture (17β-estradiol (0.2 μM) progesterone (1.5 μM) cortisol (1.5 μM) and prolactin (5 μg/ml)). Progesterone and cortisol, added singly to RMF suppressed CYP1B1 protein expression (∼80%) in both untreated and BA-induced cells, while cortisol also suppressed the 5.2-kb CYP1B1 mRNA. In contrast, 17β-estradiol stimulated constitutive expression of CYP1B1 protein (50–75%) and mRNA level (2- to 3-fold), but did not affect CYP1B1 expression in BA-treated RMF. The expression of CYP1A1 and CYP1B1 is therefore highly cell specific even though each is regulated through the Ah receptor. Each P450 exhibits a surprisingly similar pattern of hormonal regulation even though expressed in different cell types.

References (36)

  • L.H. Pottenger et al.

    Arch. Biochem. Biophys.

    (1991)
  • Ü. Savas et al.

    J. Biol. Chem.

    (1994)
  • K.K. Bhattacharyya et al.

    J. Biol. Chem.

    (1995)
  • T.R. Sutter et al.

    J. Biol. Chem.

    (1994)
  • E.B. Gehly et al.

    J. Biol. Chem.

    (1979)
  • Y. Inaguma et al.

    Dev. Biol.

    (1988)
  • D.W. Petersen et al.
  • K.S. Russel et al.

    Cancer Res.

    (1992)
  • J.M. Hall et al.

    Science

    (1990)
  • B.S. Cha et al.
  • M.S. Wolff et al.

    Reports

    (1993)
  • C.B. Huggins et al.

    Cancer Res.

    (1962)
  • C.S. Cooper et al.

    Carcinogenesis

    (1982)
  • M.N. Gould et al.

    Cancer Res.

    (1982)
  • M. Christou et al.

    Carcinogenesis

    (1987)
  • J.T. Sawicki et al.

    Cancer Res.

    (1983)
  • S. Otto et al.

    Endocrinology

    (1991)
  • L.H. Pottenger et al.

    Carcinogenesis

    (1990)
  • Cited by (105)

    • T-2 toxin upregulates the expression of human cytochrome P450 1A1 (CYP1A1) by enhancing NRF1 and Sp1 interaction

      2019, Toxicology Letters
      Citation Excerpt :

      Many cis-acting elements that bind to the promoter region of CYP1A1 have been identified, including AhR/ARNT, Sp1, nuclear factor-Y/α-actinin-4 (NF-Y/ACTN4), hypoxia-inducible factor-1α (HIF1α), pregame X receptor (PXR) and CCAAT/enhancer binding protein α (C/EBPα) (Fallone et al., 2005; Jie et al., 2008; Kim et al., 2015; Kobayashi et al., 1996; Poch et al., 2004; Shimoyama et al., 2014). A previous report showed that AhR binds to XRE and regulates the induction of CYP1A1 expression in response to TCDD (Christou et al., 1995; Nishimura et al., 2005). These findings indicate that the transcriptional network that regulates the transcription of CYP1A1 is complicated and diverse.

    • Cancer chemoprevention revisited: Cytochrome P450 family 1B1 as a target in the tumor and the microenvironment

      2018, Cancer Treatment Reviews
      Citation Excerpt :

      In the murine mammary gland CYP1A1 and CYP1B1 are expressed at low levels in physiological conditions, with CYP1A1 primarily localized in the epithelial cells and CYP1B1 in fibroblasts. The expression of epithelial CYP1A1 and fibroblast CYP1B1 could be further induced by administration of BaP or TCDD [197,198]. The analysis of normal human breast tissues also demonstrated that fibroblasts constitutively express CYP1B1 (but not CYP1A1 and CYP1A2) and that these levels can be further induced by TCDD [199].

    • CYP1B1 and hormone-induced cancer

      2012, Cancer Letters
      Citation Excerpt :

      Additionally, we hypothesize a role for γ-synuclein (SNCG), a neuronal protein required for efficient transcriptional activation of ERα [34], in CYP1B1-mediated hormonal carcinogenesis. CYP1B1 activity is regulated via several factors including aryl hydrocarbon receptor (AhR), the AhR/AhR nuclear translocator (ARNT) complex, Sp1 transcription factors, epigenetic regulation (e.g., CYP1B1 promoter methylation), endogenous oestrogen levels, ERs and BRCA-1 activity [35–37]. In non-cancerous tissues, CYP1B1 is transcriptionally activated when compounds including PAHs or dioxins bind to the AhR complex [consisting of AhR, Hsp90, X-associated protein 2 (XAP2) and p23 proteins] in the cytosol [24,38].

    View all citing articles on Scopus
    View full text