Abstract

Decidualization refers to the functional differentiation of endometrial stromal cells and plays a significant role in embryo implantation and pregnancy. C-peptide is excreted in equimolar concentrations as that of insulin during the metabolism of proinsulin in pancreatic beta-cells. High levels of C-peptide are correlated with hyperinsulinemia and polycystic ovarian syndrome, which show a defect in decidualization. However, the role of C-peptide in decidualization has not yet been studied. Here, we identified C-peptide as an endogenous antideciduogenic factor. This inhibitory function was confirmed by the reduced expression of decidual markers, including prolactin, insulin-like growth factor-binding protein-1, and Forkhead box protein O1 as well as by the fibroblastic morphological change in the presence of C-peptide. C-peptide also enhanced cellular senescence and decreased the proportion of apoptotic cells during decidualization. In addition, C-peptide potentiated the inhibitory effects of both insulin and palmitic acid in an AKT- and autophagy-independent manner, respectively. Furthermore, C-peptide augmented protein phosphatase 1 (PP1) activity, leading to a reduction in the inhibitory phosphorylation of glycogen synthase kinase (GSK)3β, which resulted in enhanced cellular senescence and decreased apoptosis during decidualization. Taken together, our findings suggest that C-peptide is an antideciduogenic factor acting via the regulation between PP1 and GSK3β in patients with hyperinsulinemia.

Highlights

  • The human proinsulin connecting peptide (C-peptide) is composed of 31 amino acids, which are cleaved from proinsulin during proteolytic processing (Bhatt et al, 2014; Leno-Duran et al, 2014)

  • The cells were differentiated in the presence of 8-Br-cAMP as shown by increased mRNA expression of decidualization markers, including PRL, insulin-like growth factor-binding protein-1 (IGFBP1), and Forkhead box protein O1 (FOXO1) (Figure 1A)

  • This reversal in the presence of C-peptide was comparable to the observations made in the absence of C-peptide (Figure 1D; second bar vs. third bar), indicating that C-peptide did not facilitate the restoration of the growth status of human eSCs

Read more

Summary

Introduction

The human proinsulin connecting peptide (C-peptide) is composed of 31 amino acids, which are cleaved from proinsulin during proteolytic processing (Bhatt et al, 2014; Leno-Duran et al, 2014). It has been shown to stimulate mitogen-activated protein kinase (MAPK), leading to the activation of Na+-K+ATPase and numerous transcription factors, including cAMP response element-binding protein (CREB), nuclear factor kappa beta (NF-κB), and activating transcription factor 1 (ATF1) These functions may play some protective roles in diabetic vascular dysfunction, micro- and/or macro-vascular damage, and diabetic neuropathy (Wahren et al, 2000). C-peptide induces cyclin D1 expression, promoting the critical transition of cell proliferation from G1- to S-phase via the activation of retinoblastoma protein phosphorylation in smooth muscles of both rats and humans (Walcher et al, 2006) These results suggest that C-peptide plays several functional roles in diverse tissues and biological contexts, in addition to its role in diabetes. The potential effects of C-peptide signaling on endometrium differentiation during embryo implantation and pregnancy maintenance have not yet been investigated

Methods
Results
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.