Abstract

Peripheral insulin resistance contributes to the development of type 2 diabetes. TCF7L2 has been tightly associated with this disease, although the exact mechanism was largely elusive. Here we propose a novel role of TCF7L2 in hepatic glucose metabolism in mammals. Expression of medium and short isoforms of TCF7L2 was greatly diminished in livers of diet-induced and genetic mouse models of insulin resistance, prompting us to delineate the functional role of these isoforms in hepatic glucose metabolism. Knockdown of hepatic TCF7L2 promoted increased blood glucose levels and glucose intolerance with increased gluconeogenic gene expression in wild-type mice, in accordance with the PCR array data showing that only the gluconeogenic pathway is specifically up-regulated upon depletion of hepatic TCF7L2. Conversely, overexpression of a nuclear isoform of TCF7L2 in high-fat diet-fed mice ameliorated hyperglycemia with improved glucose tolerance, suggesting a role of this factor in hepatic glucose metabolism. Indeed, we observed a binding of TCF7L2 to promoters of gluconeogenic genes; and expression of TCF7L2 inhibited adjacent promoter occupancies of CREB, CRTC2, and FoxO1, critical transcriptional modules in hepatic gluconeogenesis, to disrupt target gene transcription. Finally, haploinsufficiency of TCF7L2 in mice displayed higher glucose levels and impaired glucose tolerance, which were rescued by hepatic expression of a nuclear isoform of TCF7L2 at the physiological level. Collectively, these data suggest a crucial role of TCF7L2 in hepatic glucose metabolism; reduced hepatic expression of nuclear isoforms of this factor might be a critical instigator of hyperglycemia in type 2 diabetes.

Highlights

  • Dysregulation of hepatic glucose metabolism is a major predicament for the development of type 2 diabetes

  • The direct involvement of TCF7L2 on hepatic glucose metabolism has been elusive to date

  • We found that hepatic expression of nuclear isoforms of TCF7L2 was reduced in mouse models of insulin resistance

Read more

Summary

Introduction

Dysregulation of hepatic glucose metabolism is a major predicament for the development of type 2 diabetes. Hepatic gluconeogenic gene expression is mainly controlled by two major transcriptional machineries, namely cAMP response element binding protein (CREB) Regulated Transcription Activator 2 (CRTC2, known as TORC2) – CREB and Peroxisome Proliferation Activating Receptor Co-activator 1 alpha (PGC-1a) – FoxO1. Recent paper by Lu et al [19] showed the data suggesting that insulin could regulate hepatic gluconeogenic gene expression via FoxO1-independent manner, contesting the current model regarding the critical role of this factor as a regulatory target of insulin signaling pathways in the liver. Two groups reported the contrasting results using the independent lines of knockout mice for CRTC2 [20,21] These data collectively suggest that disruption of single transcriptional machinery might not be enough to affect hepatic glucose metabolism in vivo, and the transcriptional circuits are tightly interwoven with each other for the fine tuning of glucose homeostasis

Methods
Results
Discussion
Conclusion
Full Text
Paper version not known

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

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.