Abstract

Cyclin A1 (Ccna1), a member of the mammalian A type cyclins, is most abundantly expressed in spermatocytes and is essential for spermatogenesis in the mouse. Ccna1- deficient spermatocytes arrest at late meiotic prophase and undergo apoptosis. To further delineate the mechanisms and key factors involved in this process, we have examined changes in expression of genes involved in both intrinsic and extrinsic signaling pathways that trigger apoptosis in the mutant spermatocytes. Our results show that both pathways are involved, and that the factors involved in the intrinsic pathway were expressed earlier than those involved in the extrinsic pathway. We have also begun to identify in vivo Ccna1-interacting proteins, using an unbiased biochemical approach, and identified 14-3-3, a key regulator of apoptosis, as a Ccna1-interacting protein. Expression levels of 14-3-3 proteins remain unchanged between wild type and mutant testes but there were differences in the subcellular distribution. In wild type control, 14-3-3 is detected in both cytosolic and nuclear fractions whereas it is restricted to the cytoplasm in mutant testes. This differential distribution of 14-3-3 may contribute to the induction of apoptosis in Ccna1-deficient spermatocytes. These results provide insight into the apoptotic mechanisms and pathways that are triggered when progression through the meiotic cell cycle is defective in male gametogenesis.

Highlights

  • Apoptosis is an evolutionarily conserved mechanism of programed cell death that occurs in response to certain physiological stimuli, cell injury, or stress, and is a vital component of various developmental processes in metazoans [1, 2]

  • To identify the factors and pathways involved in the induction of apoptosis that occurs in late prophase cyclin A1 (Ccna1)-/- spermatocytes [13], we performed a qRT-PCR array analysis using the Super Array (SA biosciences, # PAMM012Z), which measures the expression of 84 apoptosisrelated mouse genes

  • These three time points were chosen based on previous TUNEL assays showing that in the mutant testes, there were few apoptotic spermatocytes at postnatal day (PND) 17, that they increased significantly at PND 22, and that they were robustly detected at PND 28 [15]

Read more

Summary

Introduction

Apoptosis is an evolutionarily conserved mechanism of programed cell death that occurs in response to certain physiological stimuli, cell injury, or stress, and is a vital component of various developmental processes in metazoans [1, 2]. Massive apoptosis occurs during the first wave of spermatogenesis to regulate the number of germ cells and their ratio relative to the number of Sertoli cells [5]. Apoptosis is triggered when defects in complex processes such as synapsis, recombination, and segregation of homologues compromise the quality of the spermatocyte [6]. The normal occurrence of these processes is strictly regulated by meiotic checkpoints that create a network of signaling mechanisms that modulates the activity and progression of meiosis. Depletion of essential survival factors and persistent meiotic defects are sensed by these highly conserved meiotic checkpoints, which cause an arrest of cellular progression of the defective cells and an induction of the apoptotic program [7]

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