Abstract

BackgroundThe control of the functional pancreatic β-cell mass serves the key homeostatic function of releasing the right amount of insulin to keep blood sugar in the normal range. It is not fully understood though how β-cell mass is determined.Methodology/Principal FindingsConditional chicken ovalbumin upstream promoter transcription factor II (COUP-TFII)-deficient mice were generated and crossed with mice expressing Cre under the control of pancreatic duodenal homeobox 1 (pdx1) gene promoter. Ablation of COUP-TFII in pancreas resulted in glucose intolerance. Beta-cell number was reduced at 1 day and 3 weeks postnatal. Together with a reduced number of insulin-containing cells in the ductal epithelium and normal β-cell proliferation and apoptosis, this suggests decreased β-cell differentiation in the neonatal period. By testing islets isolated from these mice and cultured β-cells with loss and gain of COUP-TFII function, we found that COUP-TFII induces the expression of the β-catenin gene and its target genes such as cyclin D1 and axin 2. Moreover, induction of these genes by glucagon-like peptide 1 (GLP-1) via β-catenin was impaired in absence of COUP-TFII. The expression of two other target genes of GLP-1 signaling, GLP-1R and PDX-1 was significantly lower in mutant islets compared to control islets, possibly contributing to reduced β-cell mass. Finally, we demonstrated that COUP-TFII expression was activated by the Wnt signaling-associated transcription factor TCF7L2 (T-cell factor 7-like 2) in human islets and rat β-cells providing a feedback loop.Conclusions/SignificanceOur findings show that COUP-TFII is a novel component of the GLP-1 signaling cascade that increases β-cell number during the neonatal period. COUP-TFII is required for GLP-1 activation of the β-catenin-dependent pathway and its expression is under the control of TCF7L2.

Highlights

  • Type 2 diabetes mellitus (T2DM) is a multifactorial disorder associated with impaired pancreatic b-cell function and insulin resistance

  • We established that COUP-TFII contributes to the control of insulin secretion through the complex HNF4a/MODY1 transcription factor network operating in b-cells [4]

  • Pdx1CRE/- COUP-TFIIFl/Fl mice are glucose intolerant Since the nuclear receptor COUP-TFII is highly expressed in pancreatic b-cells, we generated mice to study the consequences of total b-cell ablation of COUP-TFII gene expression on b-cell function and glucose homeostasis using the Cre recombinase/loxP system

Read more

Summary

Introduction

Type 2 diabetes mellitus (T2DM) is a multifactorial disorder associated with impaired pancreatic b-cell function and insulin resistance. The onset of b-cell dysfunction in T2DM is complex, involving genetic and environmental factors that lead to decreased insulin secretion and reduced b-cell mass [1]. Knockout mice with heterozygous deletion of COUP-TFII and mice with complete disruption of HNF4a in pancreatic b-cells have similar defects in insulin secretion, which led us to propose a model of transcriptional crosstalk between these two nuclear receptors. The control of the functional pancreatic b-cell mass serves the key homeostatic function of releasing the right amount of insulin to keep blood sugar in the normal range. It is not fully understood though how b-cell mass is determined

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call