[HTML][HTML] Role of Notch signaling in cell-fate determination of human mammary stem/progenitor cells

G Dontu, KW Jackson, E McNicholas… - Breast cancer …, 2004 - Springer
G Dontu, KW Jackson, E McNicholas, MJ Kawamura, WM Abdallah, MS Wicha
Breast cancer research, 2004Springer
Introduction Notch signaling has been implicated in the regulation of cell-fate decisions such
as self-renewal of adult stem cells and differentiation of progenitor cells along a particular
lineage. Moreover, depending on the cellular and developmental context, the Notch
pathway acts as a regulator of cell survival and cell proliferation. Abnormal expression of
Notch receptors has been found in different types of epithelial metaplastic lesions and
neoplastic lesions, suggesting that Notch may act as a proto-oncogene. The vertebrate …
Introduction
Notch signaling has been implicated in the regulation of cell-fate decisions such as self-renewal of adult stem cells and differentiation of progenitor cells along a particular lineage. Moreover, depending on the cellular and developmental context, the Notch pathway acts as a regulator of cell survival and cell proliferation. Abnormal expression of Notch receptors has been found in different types of epithelial metaplastic lesions and neoplastic lesions, suggesting that Notch may act as a proto-oncogene. The vertebrate Notch1 and Notch4 homologs are involved in normal development of the mammary gland, and mutated forms of these genes are associated with development of mouse mammary tumors.
Methods
In order to determine the role of Notch signaling in mammary cell-fate determination, we have utilized a newly described in vitro system in which mammary stem/progenitor cells can be cultured in suspension as nonadherent 'mammospheres'. Notch signaling was activated using exogenous ligands, or was inhibited using previously characterized Notch signaling antagonists.
Results
Utilizing this system, we demonstrate that Notch signaling can act on mammary stem cells to promote self-renewal and on early progenitor cells to promote their proliferation, as demonstrated by a 10-fold increase in secondary mammosphere formation upon addition of a Notch-activating DSL peptide. In addition to acting on stem cells, Notch signaling is also able to act on multipotent progenitor cells, facilitating myoepithelial lineage-specific commitment and proliferation. Stimulation of this pathway also promotes branching morphogenesis in three-dimensional Matrigel cultures. These effects are completely inhibited by a Notch4 blocking antibody or a gamma secretase inhibitor that blocks Notch processing. In contrast to the effects of Notch signaling on mammary stem/progenitor cells, modulation of this pathway has no discernable effect on fully committed, differentiated, mammary epithelial cells.
Conclusion
These studies suggest that Notch signaling plays a critical role in normal human mammary development by acting on both stem cells and progenitor cells, affecting self-renewal and lineage-specific differentiation. Based on these findings we propose that abnormal Notch signaling may contribute to mammary carcinogenesis by deregulating the self-renewal of normal mammary stem cells.
Springer