GATA-4 upregulates glucose-dependent insulinotropic polypeptide expression in cells of pancreatic and intestinal lineage

https://doi.org/10.1016/j.mce.2008.01.024Get rights and content

Abstract

A thorough examination of glucose-dependent insulinotropic polypeptide (GIP) expression has been hampered by difficulty in isolating widely dispersed, GIP-producing enteroendocrine K-cells. To elucidate the molecular mechanisms governing the regulation of GIP expression, 14 intestinal and pancreatic cell lines were assessed for their suitability for studies examining GIP expression. Both STC-1 cells and the pancreatic cell line βTC-3 were found to express GIP mRNA and secrete biologically active GIP. However, levels of GIP mRNA and bioactive peptide and the activity of transfected GIP reporter constructs were significantly lower in βTC-3 than STC-1 cells. When βTC-3 cells were analyzed for transcription factors known to be important for GIP expression, PDX-1 and ISL-1, but not GATA-4, were detected. Double staining for GIP-1 and GATA-4 in mouse duodenum demonstrated GATA-4 expression in intestinal K-cells. Exogenous expression of GATA-4 in βTC-3 cells led to marked increases in both GIP transcription and secretion. Lastly suppression of GATA-4 via RNA interference, in GTC-1 cells, a subpopulation of STC-1 cells with high endogenous GIP expression resulted in a marked an attenuation of GIP promoter activity. Our data support the hypothesis that GATA-4 may function to augment or enhance GIP expression rather than act as an initiator of GIP transcription.

Introduction

Glucose-dependent insulinotropic polypeptide (GIP), like glucagon-like-peptide-1 (GLP-1), is an incretin, a mediator of the enteroinsular axis that helps to maintain glucose homeostasis under physiological conditions (Murphy et al., 1995). In the presence of elevated blood glucose, incretins stimulate insulin secretion from pancreatic β-cells (O’Harte et al., 1998, Usdin et al., 1993, Yip et al., 1998). Although GLP-1 appears to be the more potent pharmacological stimulator of insulin release (Gutniak et al., 1992, Nauck et al., 1997), using GIP and GLP-1 specific receptor antagonists, it has been demonstrated that GIP is the major physiological incretin, accounting for ∼80% of nutrient induced enteroinsular pancreatic β-cell stimulation (Gault et al., 2003, Tseng et al., 1996). GIP is a 42-amino acid peptide that is produced in specific enteroendocrine cells, termed K-cells, dispersed primarily within the small intestinal mucosa (Buchan et al., 1978, Cheung et al., 2000, Yeung et al., 1999) and is released into circulation in response to the ingestion of nutrients (Cataland et al., 1974, Pederson et al., 1975).

We have begun to elucidate the mechanisms by which GIP expression is regulated. Characterization of the rat GIP gene has demonstrated that transcription initiates from a promoter located in the 5′-flanking region of the gene (Boylan et al., 1997). Furthermore, deletional analysis of the GIP promoter revealed that the first 193 bp upstream of the transcription initiation site are sufficient to direct specific expression of the GIP gene in the neuroendocrine cell line STC-1 (Boylan et al., 1997). A subsequent mutation analysis of this region demonstrated that GIP expression requires the binding of three transcription factors (Boylan et al., 1997, Jepeal et al., 2003, Jepeal et al., 2005).

The region located between base pairs −193 and −182 with respect to the transcription initiation site (+1) of the GIP promoter contains a consensus binding motif for the GATA family of DNA binding proteins, WGATAR (Jepeal et al., 2003, Orkin, 1992). Previous studies in our laboratory have shown that binding of GATA-4 to this site is essential for high GIP promoter activity in STC-1 cells (Jepeal et al., 2003). A second cis-regulatory region located between base pairs −156 and −151 of the GIP promoter also has been shown to be essential for transcription of the GIP gene in STC-1 cells (Jepeal et al., 2003) The transcription factors ISL-1 and PDX-1 were found to bind to this region and appear to function in conjunction with GATA-4 to regulate GIP gene expression (Jepeal et al., 2003, Jepeal et al., 2005).

Unfortunately, the molecular analysis of GIP expression and K-cell function has been hampered by the inability to adequately purify populations of GIP-producing K-cells, and consequently has required the identification and use of surrogate cell lines as models for such in vitro studies. Most of what is known about the regulation of GIP expression has been determined using a single cell line, STC-1 (Boylan et al., 1997, Jepeal et al., 2003, Jepeal et al., 2005, Kieffer et al., 1995). STC-1 cells were derived from an intestinal tumor isolated from a transgenic mouse expressing a viral oncogene under control of the insulin promoter (Rindi et al., 1990). These cells express multiple peptides, including GIP, glucagon, somatostatin, CCK, chromatogranin, and amylin, but not insulin (Boylan et al., 1997, Jepeal et al., 2003, Kieffer et al., 1995, Rindi et al., 1990). This plurihormonal pattern of gene expression is consistent with previous studies demonstrating the expression of multiple hormones in cells of endocrine neoplasms (Brubaker et al., 1998, Philippe et al., 1987, Philippe et al., 1988, Sidhu et al., 2000).

In addition to intestinal tumors, the transgenic mouse line that gave rise to STC-1 cells also developed pancreatic neoplasms. The cell line βTC-3 was derived from one such pancreatic β-cell tumor. Because βTC-3 cells were found to possess many of the characteristics of native differentiated β-cells, including the synthesis and secretion of considerable amounts of insulin, they have been used extensively to study insulin regulation and β-cell function (D’Ambra et al., 1990, Matsuoka et al., 2003). However, βTC-3 cells do not behave entirely like native islet β-cells by virtue of their capacity to synthesize proglucagon-derived peptides in addition to insulin (Efrat et al., 1988).

In the present study, we have demonstrated the expression and secretion of biologically active GIP from βTC-3 cells. In addition, we have examined the transcriptional regulation of GIP expression in these cells and have specifically evaluated the role of the transcription factors GATA-4, ISL-1 and PDX-1.

Section snippets

Cell culture

STC-1 cells (mouse neuroendocrine tumor derived) (Boylan et al., 1997), GTC-1 cells (a high GIP-expressing subpopulation of STC-1 cells) IEC-6 cells (normal rat small intestine derived immature intestinal stem cells [ATCC, Manassas, VA]), βTC-3 cells (mouse pancreatic β-cell tumor [ATCC]) and LGIPR2 cells (provided by Dr. Ted B. Usdin, Bethesda, MD) (Usdin et al., 1993) were grown in Dulbecco's minimal essential medium (DMEM) containing 10% (v/v) fetal bovine serum (FBS) at 37 °C in an

GIP is secreted by the pancreatic cell line βTC-3

Biologically active GIP peptide was measured reliably using a sensitive and specific in vitro bioassay. The LGIPR2 cells used in this assay are mouse L-cells cells that express both the human GIP receptor and a β-galactosidase (β-gal) reporter coupled to the cyclic adenosine monophosphate (cAMP) responsive vasoactive intestinal polypeptide (VIP) promoter (Usdin et al., 1993). When LGIPR2 cells are incubated with samples containing active GIP, the peptide present binds to and activates the GIP

Discussion

GIP expression in vitro had previously been detected only in the intestinal-like cell line STC-1 (Boylan et al., 1997, Jepeal et al., 2005, Rindi et al., 1990) and cell lines derived from STC-1 cells such as GTC-1 (Cheung et al., 2000), STC6-14 (Kieffer et al., 1995), and GIP-INS (Ramshur et al., 2002). Although numerous other intestinal cell lines have been screened by northern blot analysis, GIP bioassay and RT-PCR (Boylan et al., 1997), none have been found to express GIP. However, because

References (49)

  • C.M. Yeung et al.

    Glucose-dependent insulinotropic polypeptide gene expression in the stomach: revealed by a transgenic mouse study, in situ hybridization and immunohistochemical staining

    Mol. Cell Endocrinol.

    (1999)
  • U. Ahlgren et al.

    Beta-cell-specific inactivation of the mouse Ipf1/Pdx1 gene results in loss of the beta-cell phenotype and maturity onset diabetes

    Genes Dev.

    (1998)
  • P.L. Brubaker et al.

    Regulation of glucagon-like peptide-1 synthesis and secretion in the GLUTag enteroendocrine cell line

    Endocrinology

    (1998)
  • A.M. Buchan et al.

    Electronimmunocytochemical evidence for the K cell localization of gastric inhibitory polypeptide (GIP) in man

    Histochemistry

    (1978)
  • S. Cataland et al.

    Gastric inhibitory polypeptide (GIP) stimulation by oral glucose in man

    J. Clin. Endocrinol. Metab.

    (1974)
  • A.D.B. Cheung et al.

    Glucose-dependent insulin release form genetically engineered K cells

    Science

    (2000)
  • R. D’Ambra et al.

    Regulation of insulin secretion from beta-cell lines derived from transgenic mice insulinomas resembles that of normal beta-cells

    Endocrinology

    (1990)
  • J.K. Divine et al.

    GATA-4, GATA-5, and GATA-6 activate the rat liver fatty acid binding protein gene in concert with HNF-1alpha

    Am. J. Physiol. Gastrointest. Liver Physiol.

    (2004)
  • M.R. Dusing et al.

    High-level activation by a duodenum-specific enhancer requires functional GATA binding sites

    Am. J. Physiol. Gastrointest. Liver Physiol.

    (2003)
  • S. Efrat et al.

    Beta-cell lines derived from transgenic mice expressing a hybrid insulin gene-oncogene

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

    (1988)
  • R. Fang et al.

    GATA family transcription factors activate lactase gene promoter in intestinal Caco-2 cells

    Am. J. Physiol. Gastrointest. Liver Physiol.

    (2001)
  • X. Gao et al.

    Distinct functions are implicated for the GATA-4, -5, and -6 transcription factors in the regulation of intestine epithelial cell differentiation

    Mol. Cell. Biol.

    (1998)
  • V.A. Gault et al.

    Effects of the novel (Pro(3))GIP antagonist and exendin(9–39)amide on GIP- and GLP-1-induced cyclic AMP generation, insulin secretion and postprandial insulin release in obese diabetic (ob/ob) mice: evidence that GIP is the major physiological incretin

    Diabetologia

    (2003)
  • M. Gutniak et al.

    Antidiabetogenic effect of glucagon-like peptide-1 (7–36)amide in normal subjects and patients with diabetes mellitus

    N. Engl. J. Med.

    (1992)
  • Cited by (18)

    • K-cells and Glucose-Dependent Insulinotropic Polypeptide in Health and Disease

      2010, Vitamins and Hormones
      Citation Excerpt :

      The precise mechanisms by which nutrients regulate GIP expression levels remain to be elucidated. Studies of the rodent GIP promoter revealed that the first 193 bp upstream of the transcription initiation site is sufficient to induce specific expression of GIP transcription through the binding of transcription factors including GATA4, Isl1, and Pdx1 (Boylan et al., 1997; Jepeal et al., 2003, 2005, 2008). Pdx1 is capable of binding to the second cis-regulatory element located between base pairs − 156 and − 151 of the murine GIP promoter, and overexpression of Pdx1 leads to a specific increase in the activity of GIP/Luc reporter constructs (Jepeal et al., 2005).

    View all citing articles on Scopus
    View full text