Abstract

The Nr4a subfamily of nuclear receptor comprises three members in mammalian cells: Nur77/Nr4a1, Nurr1/Nr4a2, and Nor1/Nr4a3. Nr4a proteins play key roles in the regulation of glucose homeostasis in peripheral metabolic tissues. However, their biological functions in β-cells remain relatively uncharacterized. Here we sought to investigate the potential role of Nor1 in the regulation of β-cell mass and, in particular, β-cell survival/apoptosis. We used histological analysis to examine the consequences of genetic deletion of either Nur77 and Nor1 on β-cell mass, investigated the expression patterns of Nr4as in human islets and INS cells and performed gain- and loss-of-function experiments to further characterize the role of Nor1 in β-cell apoptosis. Surprisingly, Nor1 knockout mice displayed increased β-cell mass, whereas mice with genetic deletion of Nur77 did not exhibit any significant differences compared with their WT littermates. The increase in β-cell mass in Nor1 knockout mice was accompanied by improved glucose tolerance. A gene expression study performed in both human islets and INS cells revealed that Nor1 expression is significantly increased by pro-inflammatory cytokines and, to a lesser extent, by elevated concentrations of glucose. Nor1 overexpression in both INS and human islet cells caused apoptosis, whereas siRNA-mediated Nor1 knockdown prevented cytokine-induced β-cell death. Finally, Nor1 expression was up-regulated in islets of individuals with type 2 diabetes. Altogether, our results uncover that Nor1 negatively regulates β-cell mass. Nor1 represents a promising molecular target in diabetes treatment to prevent β-cell destruction.

Highlights

  • The Nr4a subfamily of nuclear receptor comprises three members in mammalian cells: Nur77/Nr4a1, Nurr1/Nr4a2, and Nor1/Nr4a3

  • The potential involvement of Nr4as in metabolic diseases has received increasing attention. This stems from several key observations: Nr4as expression varies in animal models of obesity and/or diabetes (18 –20), their expression is increased in adipose tissue of obese patients compared with lean subjects [21], Nr4a expression is modulated by physical activity and nutritional interventions [16, 22,23,24], and Nr4as have been shown to exert tissue-specific functions, resulting in the regulation of glucose homeostasis [18, 20, 25,26,27,28,29,30,31,32]

  • To investigate the potential roles of Nr4a members in the regulation of ␤-cell mass, we measured the cross-sectional ␤-cell areas in pancreata harvested from whole-body Nor1 or Nur77 knockout mice compared with WT littermates

Read more

Summary

ARTICLE cro

The potential involvement of Nr4as in metabolic diseases has received increasing attention (reviewed in Ref. 16) This stems from several key observations: Nr4as expression varies in animal models of obesity and/or diabetes (18 –20), their expression is increased in adipose tissue of obese patients compared with lean subjects [21], Nr4a expression is modulated by physical activity and nutritional interventions [16, 22,23,24], and Nr4as have been shown to exert tissue-specific functions, resulting in the regulation of glucose homeostasis [18, 20, 25,26,27,28,29,30,31,32]. This prompted us to further characterize the function of Nor in ␤-cells by studying its regulation and potential implication in ␤-cell death

Results
Discussion
Experimental procedures
Cell culture
Human islets
Western blotting
Cytochrome c release
Statistical analysis
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