Abstract

Hepatic gluconeogenesis is essential to maintain blood glucose levels, and its abnormal activation leads to hyperglycemia and type 2 diabetes. However, the molecular mechanisms in the regulation of hepatic gluconeogenesis remain to be fully defined. In this study, using murine hepatocytes and a liver-specific knockout mouse model, we explored the physiological role of nuclear factor Y (NF-Y) in regulating hepatic glucose metabolism and the underlying mechanism. We found that NF-Y targets the gluconeogenesis pathway in the liver. Hepatic NF-Y expression was effectively induced by cAMP, glucagon, and fasting in vivo Lentivirus-mediated NF-Y overexpression in Hepa1-6 hepatocytes markedly raised the gluconeogenic gene expression and cellular glucose production compared with empty vector control cells. Conversely, CRISPR/Cas9-mediated knockdown of NF-Y subunit A (NF-YA) attenuated gluconeogenic gene expression and glucose production. We also provide evidence indicating that CRE-loxP-mediated, liver-specific NF-YA knockout compromises hepatic glucose production. Mechanistically, luciferase reporter gene assays and ChIP analysis indicated that NF-Y activates transcription of the gluconeogenic genes Pck1 and G6pc, by encoding phosphoenolpyruvate carboxykinase (PEPCK) and the glucose-6-phosphatase catalytic subunit (G6Pase), respectively, via directly binding to the CCAAT regulatory sequence motif in their promoters. Of note, NF-Y enhanced gluconeogenesis by interacting with cAMP-responsive element-binding protein (CREB). Overall, our results reveal a previously unrecognized physiological function of NF-Y in controlling glucose metabolism by up-regulating the gluconeogenic genes Pck1 and G6pc Modulation of hepatic NF-Y expression may therefore offer an attractive therapeutic approach to manage type 2 diabetes.

Highlights

  • Hepatic gluconeogenesis is essential to maintain blood glucose levels, and its abnormal activation leads to hyperglycemia and type 2 diabetes

  • In this study, using murine hepatocytes and a liverspecific knockout mouse model, we explored the physiological role of nuclear factor Y (NF-Y) in regulating hepatic glucose metabolism and the underlying mechanism

  • To further confirm that NF-Y is involved into the cAMP axis in vivo, we treated mice with 300 ␮g/kg glucagon, a hormone produced by ␣ cells activates the hepatic cAMP signaling pathway [24]

Read more

Summary

Introduction

Hepatic gluconeogenesis is essential to maintain blood glucose levels, and its abnormal activation leads to hyperglycemia and type 2 diabetes. Gluconeogenesis in the liver is largely regulated at the transcriptional level by control of PEPCK3 and G6Pase, which are the major rate-limiting enzymes for this glucose-generating pathway [6] These enzymes catalyze the conversion of oxaloacetate and glucose-6-phosphate to phosphoenolpyruvate and glucose, respectively. Abundant evidence indicates that the cAMP signaling pathway is central to the hormonal control of gluconeogenesis [8], and cAMP-responsive element (CRE)-binding protein (CREB) is a important effector of the cAMP pathway in liver [9, 10] Both Pck and G6pc genes encoding PEPCK and G6Pase, respectively, possess CRE in their promoter regions, and binding of CREB to CRE increases the transcriptional activity of these genes. Hepatocytes lacking PGC-1␣ retain some ability to respond to cAMP [12], indicating that other pathways contribute to control of gluconeogenic gene expression by the cAMP axis

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