Interaction of hepatocyte nuclear factors in transcriptional regulation of tissue specific hormonal expression of human multidrug resistance-associated protein 2 (abcc2)

https://doi.org/10.1016/j.taap.2008.10.005Get rights and content

Abstract

Multidrug resistance-associated protein 2 (MRP2) (ABCC2) is an ATP-binding cassette membrane protein located primarily on apical surface of hepatocytes that mediates transport of conjugated xenobiotics and endogenous compounds into bile. MRP2 is highly expressed in hepatocytes, and at lower levels in small intestines, stomach and kidney. Previous reports have characterized mammalian MRP2 promoters, but none have established the molecular mechanism(s) involved in liver enriched expression. This study aims to investigate the mechanism of hepatic MRP2 regulation.

A 2130 bp of MRP2 promoter was cloned from PAC-1 clone P108G1–7, to identify putative liver specific/hormone responsive functional DNA binding sites. Using deletion analysis, site specific mutagenesis and co-transfection studies, liver specific expression was determined.

MRP2 promoter-LUC constructs were highly expressed in liver cell lines compared to non-liver cells. The region extending from − 3 to+ 458 bp of MRP2 promoter starting from AUG contained the potential binding sites for CAAATT box enhancer binding protein (C/EBP), hepatocytes nuclear factor 1, 3 and 4 (HNF1, HNF3, and HNF4. Only HNF1 and HNF4 co-transfection with MRP2 luciferase increased expression. Site specific mutational analysis of HNF1 binding site indicated an important role for HNF1α. HNF4α induction of MRP2 was independent of HNF1 binding site. C/EBP, HNF3, and HNF6 inhibited HNF1α while HNF4α induced MRP2 luciferase expression and glucocorticoids stimulated MRP2 expression.

This study emphasizes the complex regulation of MRP2 with HNF1α and HNF4α playing a central role. The coordinated regulation of xenobiotic transporters and oxidative conjugation may determine the adaptive responses to cellular detoxification processes.

Introduction

Major functions of hepatocytes involve sinusoidal extraction, intracellular metabolism and biliary excretion of endogenous and exogenous lipophilic compounds (Kim, 2002). Xenobiotics, unconjugated bilirubin and hormones are efficiently cleared from sinusoidal blood circulation into liver by basolateral transporters, including OATP-C (SLCO1B1) and also conjugated with phase II enzymes such as UDP-glucuronyltransferase (UGT1A1), sulfotransferase or glutathione transferase (Cui et al., 2001, Hagenbuch and Meier, 2004, Kullak-Ublick et al., 2004). After conjugation, the water soluble metabolites are excreted primarily into bile by the multidrug resistance-associated protein 2 (MRP2) or ABCC2 (Paulusma and Oude Elferink, 1997). Since MRP2 mediates the rate-limiting step in biliary excretion of many drugs and conjugated bilirubin, it is not surprising that recent studies indicate that biliary excretion plays an important role in the overall bioavailability of their metabolism (Bodo et al., 2003).

The transcriptional regulation of multi drug resistance is a complex process involving several key transcriptional cascades (Eloranta et al., 2005, Geier et al., 2003). Nuclear transcriptional factors such as Y-box protein1, FXR, SHP, PXR, CAR, and HNF4α have been reported to modulate the regulation of transport and drug metabolizing genes (Eloranta et al., 2005, Geier et al., 2003, Kast et al., 2002).

MRP2 is an ATP-binding cassette membrane protein that belongs to the MRP transporter subfamily. MRP2 mediates transport of xenobiotics that are generally conjugated with glucuronide, sulphate or glutathione (Borst et al., 2000, Paulusma and Oude Elferink, 1997). MRP2 is found at the apical pole primarily of hepatocytes, but also at lower content in the intestines and kidneys (Gerk and Vore, 2002). Mutant rat strains lacking Mrp2 expression such as transport deficient (TR) Wistar rat and the Eisai hyperbilirubinemic rat (EHBR) display low biliary secretion of xenobiotic conjugates and endogenous compounds such as conjugated bilirubin (Paulusma and Oude Elferink, 1997). Similarly, hereditary deficiency of MRP2 expression in humans, known as the Dubin–Johnson syndrome, causes reduced biliary elimination of bilirubin glucuronides (Kartenbeck et al., 1996, Paulusma et al., 1997) and other organic anions except bile acids.

Regulation of hepatic expression of MRP2 is still poorly understood. In rats, down regulation of Mrp2 has been shown to be associated with administration of lipopolysacharide, ethinyl estradiol and cholestasis (Gerk and Vore, 2002). On the other hand, cellular hydration, reactive oxygen species, drugs and hormones have been demonstrated to increase Mrp2 (Fardel et al., 2001, Qadri et al., 2006, Schrenk et al., 2001). Potential molecular processes for the regulation of rat and human MRP2 have been described, but their specific sites of action and tissue specific regulation is not evident. Both rat and human MRP2 promoters have been cloned and partially characterized (Geier et al., 2003, Hisaeda et al., 2004, Kauffmann and Schrenk, 1998, Kauffmann et al., 2001, Stockel et al., 2000, Tanaka et al., 1999). Potentially important sites in the human promoter which may be involved in the regulation of MRP2 were identified at − 678 to + 197 and include putative sites such as C/EBPβ, HNF1, USF, CBF/NF-Y and Sp1 (Hisaeda et al., 2004, Kauffmann et al., 2001, Stockel et al., 2000; Tanaka et al., 1999a). However, sites for basal liver-specific expression were not demonstrated in either rat or human promoters (Kauffmann et al., 2001).

Hepatocyte nuclear factor 1α (HNF1α) was one possible candidate which may be important in regulation of liver-specific expression since putative DNA binding sites have been identified in human, rat and mouse genes (Vollrath et al., 2006). In addition, HNF1α is important in modifying hepatic expression of xenobiotic transporters such as OATP-C (SLC21A6) and in both phase I and II conjugation steps including glucuronyl transferases and cytochrome P450s (Bernard et al., 1999, Bock and Kohle, 2004, Paulson et al., 1990, Song et al., 1998). Therefore, it is speculated that HNF1α, a liver enriched transcription factor, might also be responsible for basal hepatic expression of MRP2. Since many conjugating enzymes as well as Mrp2 in rats are induced by administration of glucocorticoids (Jemnitz et al., 2002), we examined whether Dexamethasone also regulated the human MRP2 promoter.

The results in this report demonstrate that HNF1α is a key transcription factor responsible for basal liver specific MRP2 expression. In addition, we also identified a putative HNF4α DNA binding site is also involved in regulation of human MRP2 expression. In contrast, HNF3, HNF6 and C/EBP decrease both basal and HNF1 and HNF4 induced expression of MRP2 promoter. Finally, our results indicate that glucocorticoids induce the human as well as the rat MRP2 promoter in vitro through a proximal promoter site that is independent of HNF1α.

Section snippets

Materials

32P-UTP (800 Ci/mmol) was obtained from GE Healthcare (Piscataway, NJ). All chemicals were of analytical grade and cell culture grade from Sigma Chemical Co. (St. Louis, MO). Restriction enzymes were obtained from Invitrogen and Promega (Madison, WI).

Genomic cloning of MRP-2, construction of deletion plasmids; and sequence analysis of the 5′ flanking region

The PAC-1 clone P108G1-7 was isolated from human genomic library by PCR screening with two primers from the first exon of human MRP2 gene (in collaboration with Dr. David Patterson). A 2130 bp human MRP2 promoter was obtained by DNA sequencing and

Cloning and analysis of the 5′-flanking region of the human MRP2 gene

The human genomic library was screened by PCR using MRP2 primers. We isolated a genomic clone that was 7.5 kb in length. The fragment containing the MRP2 5′-flanking region was identified by PCR. A total of 2130 bp nucleotides of the MRP2 5′-flanking region (nucleotides − 1 to − 2133 with respect to the translation start site) was sequenced and analyzed. The sequence is available from the Gene Bank under the accession number AF163577.

Fig. 1 shows the sequence information for the MRP2 gene.

Discussion

This study describes the molecular cloning, characterization and regulation of a human multidrug resistance-associated protein. The experiments provide evidence for involvement of proximal DNA promoter elements in determining the constitutive activity of ABCC2 promoter selectively in liver derived cell lines. This work also describes that liver-specific expression was due in large part to cis-acting regulatory elements for HNF1α and HNF4α that govern initiation of transcription in the region of

Acknowledgments

These studies were supported by grants from the NIH DK-15851, American Liver Foundation (ALF) USA and Higher Education Commission, Pakistan (HEC) R/D 829. We thank Miss Saman Majeed for technical help and proof reading of the manuscript.

References (53)

  • JungD. et al.

    Characterization of the human OATP-C (SLC21A6) gene promoter and regulation of liver-specific OATP genes by hepatocyte nuclear factor 1 alpha.

    J. Biol. Chem.

    (2001)
  • JungD. et al.

    Hepatocyte nuclear factor 1 alpha: a key mediator of the effect of bile acids on gene expression

    Hepatology

    (2003)
  • KartenbeckJ. et al.

    Absence of the canalicular isoform of the MRP gene-encoded conjugate export pump from the hepatocytes in Dubin–Johnson syndrome

    Hepatology

    (1996)
  • KastH.R. et al.

    Regulation of multidrug resistance-associated protein 2 (ABCC2) by the nuclear receptors pregnane × receptor, farnesoid X-activated receptor, and constitutive androstane receptor.

    J. Biol. Chem.

    (2002)
  • KauffmannH.M. et al.

    Sequence analysis and functional characterization of the 5′-flanking region of the rat multidrug resistance protein 2 (mrp2) gene.

    Biochem. Biophys. Res. Commun.

    (1998)
  • KauffmannH.M. et al.

    Basal expression of the rat, but not of the human, multidrug resistance protein 2 (MRP2) gene is mediated by CBF/NF-Y and Sp1 promoter-binding sites

    Toxicology

    (2001)
  • Kullak-UblickG.A. et al.

    Enterohepatic bile salt transporters in normal physiology and liver disease

    Gastroenterology

    (2004)
  • PontoglioM. et al.

    Hepatocyte nuclear factor 1 inactivation results in hepatic dysfunction, phenylketonuria, and renal Fanconi syndrome

    Cell

    (1996)
  • SchrenkD. et al.

    Up-regulation of transporters of the MRP family by drugs and toxins

    Toxicol. Lett.

    (2001)
  • ShiaoT. et al.

    Structural and functional characterization of liver cell-specific activity of the human sodium/taurocholate cotransporter

    Genomics

    (2000)
  • SongC.S. et al.

    Tissue-specific and androgen-repressible regulation of the rat dehydroepiandrosterone sulfotransferase gene promoter.

    J. Biol. Chem.

    (1998)
  • ZollnerG. et al.

    Hepatobiliary transporter expression in percutaneous liver biopsies of patients with cholestatic liver diseases

    Hepatology

    (2001)
  • BernardP. et al.

    Activation of the mouse TATA-less and human TATA-containing UDP-glucuronosyltransferase 1A1 promoters by hepatocyte nuclear factor 1

    Mol. Pharmacol.

    (1999)
  • BerrF. et al.

    Ethynylestradiol impairs bile salt uptake and Na-K pump function of rat hepatocytes

    Am. J. Physiol.

    (1984)
  • BockK.W. et al.

    Coordinate regulation of drug metabolism by xenobiotic nuclear receptors: UGTs acting together with CYPs and glucuronide transporters

    Drug Meta. Rev.

    (2004)
  • Bodo, A., Bakos, E., Szeri, F., Varadi, A., and Sarkadi, B., 2003. The role of multidrug transporters in drug...
  • Cited by (26)

    • HNF1A inhibition induces the resistance of pancreatic cancer cells to gemcitabine by targeting ABCB1

      2019, EBioMedicine
      Citation Excerpt :

      The majority of HNF1A studies have been conducted in liver and/or pancreatic islet cells. They revealed direct expression regulatory effects of HNF1A that are tissue-specific and, sometimes, opposite effects in liver and islet cells [47]. Previous studies have identified that HNF1A contains a dimerization domain at the N-terminus, a DNA-binding domain in the middle region and a transactivation domain at the C-terminus of the protein [48].

    • Insights into the diagnosis of hepatocellular carcinomas with hepatobiliary MRI

      2016, Journal of Hepatology
      Citation Excerpt :

      We will see later that HCCs have a good prognosis when both OATP1B3 and HNF4α are upregulated [9]. HNF1α and HNF4α also play an important role in the regulation of MRP2 but no data exist in HCCs [42]. Phosphorylation of transporters by the protein kinase C (PKC) pathway modifies the function of hepatocyte transporters [43].

    • Genetic polymorphisms of UGT1A8, UGT1A9 and HNF-1α and gastrointestinal symptoms in renal transplant recipients taking mycophenolic acid

      2013, Transplant Immunology
      Citation Excerpt :

      The HNF1α binds to the proximal promoters and enhances the transcription rate of the UGT1A and 2B genes in the liver [16]. Studies have also shown an important role of HNF1α in inducing the expression of MRP2 [28,29,25,26]. However, the impact of this molecule on the MPA GI side effects in transplant patients has not been elucidated.

    • Understanding the molecular mechanism(s) of hepatitis C virus (HCV) induced interferon resistance

      2013, Infection, Genetics and Evolution
      Citation Excerpt :

      The ISDR region NS5A protein with additional 26 amino acids has defined consequence on NS5A/PKR interaction. Other mechanisms that may involve the eventual phosphorylation event of PKR may be affected by the viral induced ROS; MRP2 and fatty acids (Qadri et al., 2004, 2009, 2012). In a recent study, palmitate, a saturated fatty acid, was shown to bind PKR and consequently inhibiting its autophosphorylation at Thr451 and Thr446 (Cho et al., 2011).

    • Molecular characterization and functions of zebrafish ABCC2 in cellular efflux of heavy metals

      2011, Comparative Biochemistry and Physiology - C Toxicology and Pharmacology
    View all citing articles on Scopus
    View full text