Abstract

During the luteinization after ovulation in mammalian ovary, the containing cells undergo an energy consuming function re-determination process to differentiate into luteal cells under avascular environment. Previous evidences have delineated the contribution of autophagy to the cell differentiation and the catabolic homeostasis in various types of mammalian cells, whereas few interest had been focused on the involvement of autophagy in the luteinization of granulosa cells during the formation of early corpus luteum. Herein, the present study investigated that expression and contribution of autophagy during granulosa cell luteinization and early luteal development through in vivo and in vitro experiments. The results clearly demonstrated that HIF-1α/BNIP3-mediated autophagy plays a vital role in the luteinization of granulosa cells during the early luteal formation in vivo and in vitro. In the neonatal corpus luteum, HIF-1α up-regulated BNIP3 expressions, which contributed to the autophagic initiation by disrupting beclin1 from Bcl-2/beclin1 complex and protected cells from apoptosis by curbing the skew of mitochondria balance under avascular niche. Notably, Inhibition of HIF-1α activity by echinomycin enhanced the levels of cytoplasmic cytochrome c and cell apoptosis in the nascent corpus luteum. These findings revealed that HIF-1α/BNIP3-mediated autophagy enabled the process of granulosa cell luteinization and protected the granulosa-lutein cells from further apoptosis under hypoxia niche. To our knowledge, the present study firstly clarified that HIF-1α/BNIP3-mediated autophagy contributes to the luteinization of granulosa cells during the formation of pregnant corpus luteum, which will help us further understanding the luteal biology and provide us new clues for the treatment of luteal insufficiency.

Highlights

  • Corpus luteum is an ephemeral endocrine gland responsible for the secretion of sexual hormones and the maintenance of mammalian pregnancy (Galvão et al, 2013)

  • These findings indicated that hypoxia-inducible factor (HIF)-1α was involved in the luteinization of granulosa cells in vivo and in vitro

  • Targeting BNIP3 obviously reduced the expression levels of LC3II and StAR in these granulosa-lutein cells with no significant changes of HIF-1α (Figures 3A–D). These results of in vivo study revealed that inhibition of HIF-1α binding activity by echinomycin curbed the expression levels of BNIP3 and LC-3II in luteal cells (Figures 3E,F). These results clearly demonstrated that HIF-1α/BNIP3 pathway is involved in the regulation of autophagy initiation and the luteinization of granulosa cells in vivo and in vitro

Read more

Summary

Introduction

Corpus luteum is an ephemeral endocrine gland responsible for the secretion of sexual hormones and the maintenance of mammalian pregnancy (Galvão et al, 2013). The contracted corpus luteum undergoes a rapid and remarkable amount of tissue remodeling and angiogenesis, the formation of luxurious blood vessel network consumes a few days after ovulation (Davis et al, 2003; Stocco et al, 2007). Both the differentiation of granulosa cell and the formation of corpus luteum are progressed under low oxygen conditions (Huey et al, 1999). By which mechanism granulosa cells overcome hypoxia condition to initiate the program of cell differentiation still remains elusive

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