Neuron-restrictive silencer factor functions to suppress Sp1-mediated transactivation of human secretin receptor gene

https://doi.org/10.1016/j.bbagrm.2012.11.002Get rights and content

Abstract

In the present study, a functional neuron restrictive silencer element (NRSE) was initially identified in the 5′ flanking region (− 83 to − 67, relative to ATG) of human secretin receptor (hSCTR) gene by promoter assays coupled with scanning mutation analyses. The interaction of neuron restrictive silencer factor (NRSF) with this motif was later indicated via gel mobility shift and ChIP assays. The silencing activity of NRSF was confirmed by over-expression and also by shRNA knock-down of endogenous NRSF. These studies showed an inverse relationship between the expression levels of NRSF and hSCTR in the cells. As hSCTR gene was previously shown to be controlled by two GC-boxes which are regulated by the ratio of Sp1 to Sp3, in the present study, the functional interactions of NRSF and Sp proteins to regulate hSCTR gene was investigated. By co-immunoprecipitation assays, we found that NRSF could be co-precipitated with Sp1 as well as Sp3 in PANC-1 cells. Interestingly, co-expressions of these factors showed that NRSF could suppress Sp1-mediated, but not Sp3-mediated, transactivation of hSCTR. Taken together, we propose here that the down-regulatory effects of NRSF on hSCTR gene expression are mediated via its suppression on Sp1-mediated transactivation.

Highlights

► The binding of NRSF to hSCTR was investigated by gel shift and ChIP assays. ► The interactions of NRSF with Sp1 and Sp3 were studied by co-immunoprecipitation. ► The Sp1 to Sp3 ratio mediates the silencing effects of NRSF on hSCTR. ► Treatment of TSA relieves”” that HDAC is key to the silencer activity of NRSF.

Introduction

Secretin (SCT), a brain-gut peptide belonging to the secretin/vasoactive intestinal peptide/glucagon peptide family, functions to stimulate secretion of bicarbonate, electrolytes and water from pancreatic ductal epithelial cells [1], while its effects on other gastrointestinal tissues including intestine [2], stomach [3] and liver [4] were also suggested. Recently, the neuroactive functions of secretin were studied. It was suggested that in the cerebellum, secretin acts as a retrograde messenger to facilitate GABA release from the presynaptic basket cells, either directly or indirectly via an unknown glutamate source, resulting in potentiating evoked inhibitory postsynaptic currents (IPSCs) in Purkinje cells [5], [6]. Recent evidences have suggested that SCT is potentially a neurohypophysial factor [7]. SCT and its receptor (SCTR) overlap with the functions of angiotensin II (ANGII), and more importantly, are needed in mediating the central actions of ANGII-induced responses [8].

The activities of secretin are mediated via a class II G protein-coupled receptor, secretin receptor (SCTR). Using human pancreatic ductal carcinoma (PANC-1) and bovine pancreatic ductal (BPD-1) cells as models, we have previously identified a 106-bp core promoter element (− 263 to − 158, relative to the ATG start codon) in the 5′ flanking region of the human secretin receptor (hSCTR) gene [9]. This core promoter is controlled by the competitive binding of specificity protein 1 and 3 (Sp1 and Sp3) with two functional GC boxes (− 240 to − 226 and − 203 to − 194). In addition, the methylation status of CpG dinucleotides in the CpG island which overlaps with the core promoter was also found to be a critical factor to mediate the cell-specific expression of the hSCTR [10].

To understand further the spatial and temporal expression of hSCTR, in this report, we sought to investigate the functions of a putative neuron restrictive silencer element (NRSE) located downstream (− 83 to − 67, relative to ATG) of the hSCTR core promoter. NRSE, also known as repressor element-1 (RE-1) with a consensus sequence “NTYAGMRCCNNRGMSAG” [11], was initially identified to regulate a number of neuron-specific genes by repressing their expressions in non-neural tissues [12]. Recently, NRSE is regarded a common repressor element as it can suppress an increasing number of non-neuronal genes. A genome-wide search indicated the presence of about 1800 putative NRSE sites in both human and mouse genomes [11]. The protein factor that interacts with NRSE is neuron-restrictive silencer factor (NRSF), which is a member of the zinc-finger GLi-Krüppel family [13]. There are three domains in NRSF: a Krüppel-type zinc-finger for binding NRSE, an N-terminal repressor domain for interacting with SIN3 transcription regulator (Sin3), and a C-terminal repressor domain for recruiting the corepressor element 1 silencing transcription factor (CoREST). Both Sin3 and CoREST interact with histone deacetylase containing complex to deacetylate core histone proteins, and as a consequence, they work together to silence target genes by forming condensed chromatin structures and mediates developmental stage-specific gene expression [14], [15], [16]. An in silico analysis indicates that all members of the secretin receptor family contain at least one putative NRSE-like motif in their 5’ flanking regions [17]. With the knowledge that most of NRSF-regulated genes also possess GC-boxes in their promoter regions [18], [19], for this reason, the functional relationships between Sp-protein(s) and NRSF in controlling hSCTR expression was investigated. Findings reported here not only provide crucial information regarding the cell-specific expression of hSCTR, but also are applicable to the understanding of other GC-box/NRSF co-regulated genes, including several members of the secretin receptor family.

Section snippets

Cell culture

All cell lines were purchased from American Type Culture Collection (ATCC). PANC1-1 cells were cultured in Dulbecco's modified Eagle's medium (DMEM, Invitrogen, Carlsbad, CA) with 10% FBS. PC12 cells were cultured in DMEM with 10% horse serum and 5% fetal bovine serum (FBS). All cells were cultured at 37 °C with 5% CO2 in a medium supplemented with 100 U/ml penicillin G and 100 μg/ml streptomycin (Invitrogen).

Plasmid construction

Cloning of the hSCTR promoter was described previously [9]. The plasmid p263/158 (− 263

Identification of a functional NRSE in the 5′ flanking region of the hSCTR gene

In human and mouse SCTR genes, putative NRSE sites at − 83/− 67 and − 314/− 298, respectively, relative to the ATG codon were identified (Fig. 1A). To initially investigate whether this motif is functional in hSCTR, several DNA fragments with different 3′ regions (ranged from − 158 to − 1) were linked to the luciferase reporter gene for transient promoter assays using the human pancreatic PANC-1 cell as a model. Constructs with the core promoter and the putative NRSE (p-263/-45, p-263/-1) exhibited

NRSF and cell-specificity of secretin receptor

Secretin receptor was previously considered to be localized to classical sites such as the pancreatic and biliary ductal cells. Now it has been demonstrated that secretin receptors are expressed in specific cell types in various parts of the brain [5], [29], [30] as well as peripheral tissues [31], [32], [33]. The spatial and temporal regulation of this receptor is the key to understanding secretin's physiology, but remains largely unknown. The present study provides new information regarding

Funding

The present study was supported by the Hong Kong Government RGC grant 7696/09M to LTO Lee, CRFHKU6/CRF/11G to BKC Chow and the Committee on Research 201109176070 to LTO Lee.

Acknowledgements

We thank Prof. C. Paya, Mayo Clinic, for providing Sp1/CMV and Sp3/CMV vector, Prof. G. Suske, Institut fuör Molekularbiologie and Tumorforschung, for the Sp4/CMV vector and Dr. G. Mandel, State University of New York, for REEX1 and p73 vector.

References (51)

  • M. Mieda et al.

    Expression of the rat m4 muscarinic acetylcholine receptor gene is regulated by the neuron-restrictive silencer element/repressor element 1

    J. Biol. Chem.

    (1997)
  • F. Atouf et al.

    Expression of neuronal traits in pancreatic beta cells. Implication of neuron-restrictive silencing factor/repressor element silencing transcription factor, a neuron-restrictive silencer

    J. Biol. Chem.

    (1997)
  • S.D. Kraner et al.

    Silencing the type II sodium channel gene: a model for neural-specific gene regulation

    Neuron

    (1992)
  • D.M. Kemp et al.

    NRSF/REST confers transcriptional repression of the GPR10 gene via a putative NRSE/RE-1 located in the 5′ promoter region

    FEBS Lett.

    (2002)
  • H.J. Lee et al.

    Regulation of mGluR1 expression in human melanocytes and melanoma cells

    Biochim. Biophys. Acta

    (2012)
  • C.S. Kim et al.

    Neuron-restrictive silencer factor (NRSF) functions as a repressor in neuronal cells to regulate the mu opioid receptor gene

    J. Biol. Chem.

    (2004)
  • Y. Zhang et al.

    Dual mechanisms of regulation of transcription of luteinizing hormone receptor gene by nuclear orphan receptors and histone deacetylase complexes

    J. Steroid Biochem. Mol. Biol.

    (2003)
  • H.P. Bayliss et al.

    Mechanism of pancreatic secretion

    J. Physiol. (Lond.)

    (1902)
  • P.S. Olsen et al.

    Effect of secretin and somatostatin on secretion of epidermal growth factor from Brunner's glands in the rat

    Dig. Dis. Sci.

    (1994)
  • R. Lenzen et al.

    Secretin stimulates bile ductular secretory activity through the cAMP system

    Am. J. Physiol.

    (1992)
  • W.H. Yung et al.

    Secretin facilitates GABA transmission in the cerebellum

    J. Neurosci.

    (2001)
  • S.M. Lee et al.

    Expression and spatial distribution of secretin and secretin receptor in human cerebellum

    Neuroreport

    (2005)
  • J.Y. Chu et al.

    Secretin as a neurohypophysial factor regulating body water homeostasis

    Proc. Natl. Acad. Sci. U.S.A.

    (2009)
  • V.H. Lee et al.

    An indispensable role of secretin in mediating the osmoregulatory functions of angiotensin II

    FASEB J.

    (2010)
  • R.T. Pang et al.

    CpG methylation and transcription factors Sp1 and Sp3 regulate the expression of the human secretin receptor gene

    Mol. Endocrinol.

    (2004)
  • Cited by (3)

    • Role of nuclear factor of activated T-cells 5 in regulating hypertonic-mediated secretin receptor expression in kidney collecting duct cells

      2016, Biochimica et Biophysica Acta - Gene Regulatory Mechanisms
      Citation Excerpt :

      Hence, the precise molecular mechanism underlying the induction of SCTR gene expression in response to osmotic stress has yet to be clarified. To better understand the gene regulation of SCTR, in previous research we have identified a core promoter element of the human SCTR (hSCTR) gene [9–11] within the promoter's two functional GC boxes that interact specifically with Sp1 and Sp3. Similar to the SCT gene, Sp1 strongly activates hSCTR promoter activity, whereas Sp3 represses the promoter function by competing with Sp1 for the same binding site.

    View full text