Abstract

Elucidation of non-canonical protein functions can identify novel tissue homeostasis pathways. Herein, we describe a role for the Bcl-2 family member BAD in postnatal mammary gland morphogenesis. In Bad3SA knock-in mice, where BAD cannot undergo phosphorylation at 3 key serine residues, pubertal gland development is delayed due to aberrant tubulogenesis of the ductal epithelium. Proteomic and RPPA analyses identify that BAD regulates focal adhesions and the mRNA translation repressor, 4E-BP1. These results suggest that BAD modulates localized translation that drives focal adhesion maturation and cell motility. Consistent with this, cells within Bad3SA organoids contain unstable protrusions with decreased compartmentalized mRNA translation and focal adhesions, and exhibit reduced cell migration and tubulogenesis. Critically, protrusion stability is rescued by 4E-BP1 depletion. Together our results confirm an unexpected role of BAD in controlling localized translation and cell migration during mammary gland development.

Highlights

  • Elucidation of non-canonical protein functions can identify novel tissue homeostasis pathways

  • This work identifies BAD as a modulator of mammary gland morphogenesis and shows a regulatory link to localized translation that is critical for pubertal stage-specific cell migration

  • We used 3 genetic engineered mouse models to explore the role of BAD in postnatal mammary gland development; knock-out Bad−/−, knock-in BadS155A and Bad3SA where 3SA indicates alanine substitutions at S112/136/155 of the endogenous Bad allele[7,8,10]

Read more

Summary

Introduction

Elucidation of non-canonical protein functions can identify novel tissue homeostasis pathways. Together our results confirm an unexpected role of BAD in controlling localized translation and cell migration during mammary gland development. The BH3-only Bcl-2-associated agonist of death protein BAD1 is best known as an apoptosis sensitizer, but has complex roles in vivo[2,3,4] It is regulated by coordinated phosphorylation of three serine (S) residues S112, 136, and 155 (75, 99, and 118 in humans) that differentially modulate activity in a tissue-specific manner[5,6]. Unbiased proteomic screens identify that Bad3SA dysregulates the mRNA translational repressor protein 4E-BP1, focal adhesion and actin binding components, which together suggest defects to the cell motility machinery. This work identifies BAD as a modulator of mammary gland morphogenesis and shows a regulatory link to localized translation that is critical for pubertal stage-specific cell migration

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