Abstract

BackgroundThe adult mammary epithelium is composed of basal and luminal cells. The luminal lineage comprises two major cell populations, positive and negative for estrogen and progesterone receptors (ER and PR, respectively), both containing clonogenic progenitor cells. Deregulated ER/PR− luminal progenitor cells are suspected to be at the origin of basal-type triple-negative (TNBC) breast cancers, a subtype frequently associated with loss of P53 function and MET signaling hyperactivation. Using mouse models, we recently reported that p53 restricts luminal progenitor cell amplification whereas paracrine Met activation stimulates their growth and favors a luminal-to-basal switch. Here, we analyzed how these two critical pathways interact to control luminal progenitor function.MethodsWe have (i) established and analyzed the gene expression profile of luminal progenitors isolated by ICAM-1, a robust surface marker we previously identified; (ii) purified luminal progenitors from p53-deficient and p53-proficient mouse mammary epithelium to compare their functional and molecular characteristics; and (iii) analyzed their response to HGF, the major Met ligand, in three-dimensional cultures.ResultsWe found that luminal progenitors, compared to non-clonogenic luminal cells, overexpress Trp53 and numerous p53 target genes. In vivo, loss of Trp53 induced the expansion of luminal progenitors, affecting expression of several important p53 target genes including those encoding negative regulators of cell cycle progression. Consistently, p53-deficient luminal progenitors displayed increased proliferative and self-renewal activities in culture. However, they did not exhibit perturbed expression of luminal-specific markers and major regulators, such as Hey1, Elf5, and Gata3. Moreover, although expressing Met at higher level than p53-proficient luminal progenitors, p53-deficient luminal progenitors failed to acquire basal-specific features when stimulated by HGF, showing that p53 promotes the plastic behavior of luminal progenitors downstream of Met activation.ConclusionsOur study reveals a crosstalk between Met- and p53-mediated signaling pathways in the regulation of luminal progenitor function. In particular, it shows that neither p53 loss alone nor p53 loss combined with Met signaling activation caused an early detectable cell fate alteration in luminal progenitors. Conceivably, additional events are required to confer basal-specific characteristics to luminal-derived TNBCs.

Highlights

  • The adult mammary epithelium is composed of basal and luminal cells

  • Molecular profiles of the luminal subsets separated by ICAM-1 We recently identified ICAM-1 as a robust surface marker allowing the separation of clonogenic luminal progenitors from non-clonogenic Estrogen receptor (ER)/Progesterone receptor (PR)+ luminal cells in the adult mouse mammary epithelium [8]

  • Luminal progenitors identified by ICAM-1 express Met and Trp53 at high level We previously reported that the ICAM1+ luminal progenitor-enriched population is a target of hepatocyte growth factor (HGF)/Met signaling [8]

Read more

Summary

Introduction

The adult mammary epithelium is composed of basal and luminal cells. The luminal lineage comprises two major cell populations, positive and negative for estrogen and progesterone receptors (ER and PR, respectively), both containing clonogenic progenitor cells. The luminal lineage, characterized by its expression of keratins 8/18/19, comprises ductal hormone-sensing cells, positive for estrogen and progesterone receptors (ER, PR), and ductal and alveolar cells lacking ER and PR expression [1, 5, 6] Both ER/PR+ and ER/PR− luminal cell fractions contain clonogenic stem/progenitor cells that drive the expansion of the luminal cell population during puberty and gestation and ensure its maintenance at homeostasis [7,8,9,10,11]. ER/PR− luminal stem/progenitor cells are suspected to be at the origin of basal-like, triple-negative breast cancers (TNBCs), a subtype lacking ER, PR, and amplified HER2, displaying a basal-type gene signature [12,13,14,15,16]. Luminal progenitors seem able to express basal-specific genes under pathological conditions, indicating that they display phenotypic plasticity, a property contributing to the complex intra-tumoral heterogeneity of TNBCs [17]

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