Regular Article
Involvement of Hepatocyte Nuclear Factor 4alpha in Transcriptional Regulation of the Human Pregnane X Receptor Gene in the Human Liver

https://doi.org/10.2133/dmpk.23.59Get rights and content

Summary:

Pregnane X receptor (PXR; NR1I2), a key transcriptional factor that regulates genes encoding drug-metabolizing enzymes and drug transporters, is abundantly expressed in the human liver. However, studies on the molecular mechanism of human PXR gene regulation are limited. In this study, we examined the involvement of hepatocyte nuclear factor 4alpha (HNF4α; NR2A1) in the transcriptional regulation of the human PXR gene in the human liver. The activities of the human PXR promoter containing the direct repeat 1 (DR1) element located at  88/− 76 of the promoter were significantly increased by co-expression of HNF4α in the human hepatocellular carcinoma cell line. In addition, introduction of mutation into the DR1 element abolished the transcriptional activation of the human PXR promoter by exogenous HNF4α. The results of gel mobility shift assays and chromatin immunoprecipitation assays showed that HNF4α was bound to the promoter region containing the DR1 element. A knock-down of HNF4α by siRNA significantly decreased expression levels of endogenous PXR mRNA in HepG2 cells. Furthermore, expression levels of PXR mRNA positively correlated with those of HNF4α mRNA in 18 human liver samples. These results suggested that HNF4α transactivated the human PXR gene by binding to the DR1 element located at − 88/− 76 of the promoter and was involved in the expression of PXR in the human liver.

References (33)

  • B. Blumberg et al.

    M., Ong, E. S. and Evans, R. M.: SXR, a novel steroid and xenobiotic-sensing nuclear receptor

    Genes Dev.

    (1998)
  • J.M. Lehmann et al.

    The human orphan nuclear receptor PXR is activated by compounds that regulate CYP3A4 gene expression and cause drug interactions

    J. Clin. Invest.

    (1998)
  • B. Goodwin et al.

    The orphan human pregnane X receptor mediates the transcriptional activation of CYP3A4 by rifampicin through a distal enhancer module

    Mol. Pharmacol.

    (1999)
  • B. Goodwin et al.

    Regulation of the human CYP2B6 gene by the nuclear pregnane X receptor

    Mol. Pharmacol.

    (2001)
  • Y. Chen et al.

    Induction of human CYP2C9 by rifampicin, hyperforin, and phenobarbital is mediated by the pregnane X receptor

    J. Pharmacol. Exp. Ther.

    (2004)
  • J.M. Pascussi et al.

    Dexamethasone induces pregnane X receptor and retinoid X receptor-alpha expression in human hepatocytes: synergistic increase of CYP3A4 induction by pregnane X receptor activators

    Mol. Pharmacol.

    (2000)
  • Cited by (19)

    • RNA interference screen identifies NAA10 as a regulator of PXR transcription

      2019, Biochemical Pharmacology
      Citation Excerpt :

      Following the screening data analysis, the genes that repressed PXR mRNA after knockdown to equal to or greater than PXR siRNA controls were selected as hits from each plate (n = 115) for a confirmatory screen. Expected hits included PXR itself and HNF4A (HNF4A siRNA was indicated as a blue dot in Fig. 1B and C), a previously described regulator of PXR [13,14], thereby validating our experimental approach. To minimize false positives due to off-target effects, we assembled a new library containing pooled siRNAs, with each pool containing four individual siRNAs targeting each of the genes identified in the primary screen.

    • Developmental regulation of CYP3A4 and CYP3A7 in Chinese Han population

      2016, Drug Metabolism and Pharmacokinetics
      Citation Excerpt :

      On the other hand, HNF4A is also a key transcription factor of a coordinating nuclear receptor-mediated response to CYP3A4 through activation of the PXR gene. Iwazaki confirmed that HNF4A transactivated the PXR gene by binding to the DR1 element located at −88/−76 of the promoter and enhanced the expression level of PXR in the human liver [25]. Furthermore, during rifampin-mediated PXR activation, PXR simultaneously inhibited SHP gene transcription and subsequently promoted recruitment of transcriptional co-activators to the CYP3A4 chromatin.

    • Crosstalk of HNF4α with extracellular and intracellular signaling pathways in the regulation of hepatic metabolism of drugs and lipids

      2016, Acta Pharmaceutica Sinica B
      Citation Excerpt :

      HNF4α is required for FXR expression in the fetal liver but not in the adult liver2. HNF4α can activate the human PXR promoter by binding to the DR1 motif in hepatoma cells64. In mice, HNF4α is essential for the expression of PXR in fetal liver by binding to the PXR promoter65.

    • Identification and interplay of sequence specific DNA binding proteins involved in regulation of human Pregnane and Xenobiotic Receptor gene

      2015, Experimental Cell Research
      Citation Excerpt :

      Statistical analysis was done by two way Student's t-test and asterisks (*) signify values that differed significantly from the control experiment with p-value less than 0.05 (p<0.05 in Student's t-test). Studies from our laboratory along with others have shown that expression of PXR is driven by proximal promoter region [33,34,39,40]. In a recent report, we have identified the regulatory sequences within the 5′-proximal promoter that tightly regulate PXR expression in liver cells [34].

    • Pregnane and Xenobiotic Receptor gene expression in liver cells is modulated by Ets-1 in synchrony with transcription factors Pax5, LEF-1 and c-jun

      2015, Experimental Cell Research
      Citation Excerpt :

      The human PXR-promoter homology with mouse promoter, 100 bp 5′-upstream region from the transcription initiation site of PXR gene is highly conserved in both the species [18]. From gene regulation studies, it is apparent that NR family members like glucocorticoid receptor (GR), peroxisome-proliferator activated receptor (PPARα) and hepatocyte nuclear factor (HNF4α) activate the transcription of human PXR which is predominantly mediated by its proximal promoter [17,19,20]. Because of its important physiologic and pathologic roles, understanding the mechanism of PXR gene expression is fundamental to the control of PXR expression and to the development of new therapeutic strategies for preventing drug–drug interaction or PXR-related cancer conditions.

    View all citing articles on Scopus

    This study was supported by a grant-in-aid from The Japan Health Sciences Foundation (Research on Health Sciences focusing on Drug Innovation) and the Ministry of Health Labor and Welfare of Japan (Research in Regulatory Science of Pharmaceutical and Medical Devices)

    View full text