Distinct Effects of EGFR Ligands on Human Mammary Epithelial Cell Differentiation

Based on gene expression patterns, breast cancers can be divided into subtypes that closely resemble various developmental stages of normal mammary epithelial cells (MECs). Thus, understanding molecular mechanisms of MEC development is expected to provide critical insights into initiation and progression of breast cancer. Epidermal growth factor receptor (EGFR) and its ligands play essential roles in normal and pathological mammary gland. Signals through EGFR is required for normal mammary gland development. Ligands for EGFR are over-expressed in a significant proportion of breast cancers, and elevated expression of EGFR is associated with poorer clinical outcome. In the present study, we examined the effect of signals through EGFR on MEC differentiation using the human telomerase reverse transcriptase (hTERT)-immortalized human stem/progenitor MECs which express cytokeratin 5 but lack cytokeratin 19 (K5+K19- hMECs). As reported previously, these cells can be induced to differentiate into luminal and myoepithelial cells under appropriate culture conditions. K5+K19- hMECs acquired distinct cell fates in response to EGFR ligands epidermal growth factor (EGF), amphiregulin (AREG) and transforming growth factor alpha (TGFα) in differentiation-promoting MEGM medium. Specifically, presence of EGF during in vitro differentiation supported development into both luminal and myoepithelial lineages, whereas cells differentiated only towards luminal lineage when EGF was replaced with AREG. In contrast, substitution with TGFα led to differentiation only into myoepithelial lineage. Chemical inhibition of the MEK-Erk pathway, but not the phosphatidylinositol 3-kinase (PI3K)-AKT pathway, interfered with K5+K19- hMEC differentiation. The present data validate the utility of the K5+K19- hMEC cells for modeling key features of human MEC differentiation. This system should be useful in studying molecular/biochemical mechanisms of human MEC differentiation.

[1]  G. Merlo,et al.  Expression of transforming growth factor alpha, amphiregulin and cripto-1 in human breast carcinomas. , 1994, British Journal of Cancer.

[2]  Lothar Hennighausen,et al.  Information networks in the mammary gland , 2005, Nature Reviews Molecular Cell Biology.

[3]  P. Cohen,et al.  Sustained activation of the mitogen-activated protein (MAP) kinase cascade may be required for differentiation of PC12 cells. Comparison of the effects of nerve growth factor and epidermal growth factor. , 1992, The Biochemical journal.

[4]  G. Mann,et al.  A mutation in the epidermal growth factor receptor in waved-2 mice has a profound effect on receptor biochemistry that results in impaired lactation. , 1995, Proceedings of the National Academy of Sciences of the United States of America.

[5]  Wenjun Guo,et al.  Slug and Sox9 Cooperatively Determine the Mammary Stem Cell State , 2012, Cell.

[6]  V. Band,et al.  Human papilloma virus DNAs immortalize normal human mammary epithelial cells and reduce their growth factor requirements. , 1990, Proceedings of the National Academy of Sciences of the United States of America.

[7]  A. Ashworth,et al.  BRCA1 basal-like breast cancers originate from luminal epithelial progenitors and not from basal stem cells. , 2010, Cell stem cell.

[8]  N. Hynes,et al.  Key signalling nodes in mammary gland development and cancer: Myc , 2009, Breast Cancer Research.

[9]  S. Bates,et al.  Expression of transforming growth factor alpha and its messenger ribonucleic acid in human breast cancer: its regulation by estrogen and its possible functional significance. , 1988, Molecular endocrinology.

[10]  R. Nusse,et al.  Developmental stage and time dictate the fate of Wnt/β-catenin-responsive stem cells in the mammary gland. , 2012, Cell Stem Cell.

[11]  L. Chodosh,et al.  Autocrine prolactin induced by the Pten-Akt pathway is required for lactation initiation and provides a direct link between the Akt and Stat5 pathways. , 2012, Genes & development.

[12]  Christian A. Rees,et al.  Molecular portraits of human breast tumours , 2000, Nature.

[13]  S. Green,et al.  PC12 cell neuronal differentiation is associated with prolonged p21ras activity and consequent prolonged ERK activity , 1992, Neuron.

[14]  C. Watson,et al.  Key signalling nodes in mammary gland development and cancer. Signalling downstream of PI3 kinase in mammary epithelium: a play in 3 Akts , 2010, Breast Cancer Research.

[15]  S. Sunnarborg,et al.  The ADAM17–amphiregulin–EGFR Axis in Mammary Development and Cancer , 2008, Journal of Mammary Gland Biology and Neoplasia.

[16]  D. Goeddel,et al.  Synthesis of messenger RNAs for transforming growth factors alpha and beta and the epidermal growth factor receptor by human tumors. , 1987, Cancer research.

[17]  R. Tibshirani,et al.  Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications , 2001, Proceedings of the National Academy of Sciences of the United States of America.

[18]  C. Perou,et al.  Phenotypic evaluation of the basal-like subtype of invasive breast carcinoma , 2006, Modern Pathology.

[19]  H. Clevers,et al.  Developmental stage‐specific contribution of LGR5+ cells to basal and luminal epithelial lineages in the postnatal mammary gland , 2012, The Journal of pathology.

[20]  W. Hahn,et al.  Human breast cancer cells generated by oncogenic transformation of primary mammary epithelial cells. , 2001, Genes & development.

[21]  C. Eaves,et al.  Characterization of bipotent mammary epithelial progenitor cells in normal adult human breast tissue , 2001, Breast Cancer Research and Treatment.

[22]  A. Rocha,et al.  Distinct stem cells contribute to mammary gland development and maintenance , 2011, Nature.

[23]  R. Tibshirani,et al.  Repeated observation of breast tumor subtypes in independent gene expression data sets , 2003, Proceedings of the National Academy of Sciences of the United States of America.

[24]  David C. Lee,et al.  Targeted inactivation of the EGF and amphiregulin genes reveals distinct roles for EGF receptor ligands in mouse mammary gland development. , 1999, Development.

[25]  Mads Lerdrup,et al.  Differential Effects of EGFR Ligands on Endocytic Sorting of the Receptor , 2009, Traffic.

[26]  V. Band,et al.  Telomerase-immortalized human mammary stem/progenitor cells with ability to self-renew and differentiate , 2010, Proceedings of the National Academy of Sciences.

[27]  M. Vivanco,et al.  Corrigendum to “Growth and differentiation of progenitor/stem cells derived from the human mammary gland” [Exp. Cell Res. 297 (2004) 444–460] , 2004 .

[28]  E. Lander,et al.  Genetic predisposition directs breast cancer phenotype by dictating progenitor cell fate. , 2011, Cell stem cell.

[29]  M. Wicha,et al.  Regulation of Mammary Stem/Progenitor Cells by PTEN/Akt/β-Catenin Signaling , 2009, PLoS biology.

[30]  L. Hennighausen,et al.  Conditional loss of PTEN leads to precocious development and neoplasia in the mammary gland. , 2002, Development.

[31]  V. Band,et al.  Distinctive traits of normal and tumor-derived human mammary epithelial cells expressed in a medium that supports long-term growth of both cell types. , 1989, Proceedings of the National Academy of Sciences of the United States of America.

[32]  L. Luttrell,et al.  Essential role of c-Cbl in amphiregulin-induced recycling and signaling of the endogenous epidermal growth factor receptor. , 2009, Biochemistry.

[33]  A. Trumpp,et al.  Negative Regulation of Neural Stem/Progenitor Cell Proliferation by the Pten Tumor Suppressor Gene in Vivo , 2001, Science.

[34]  J. Fata,et al.  The MAPK(ERK-1,2) pathway integrates distinct and antagonistic signals from TGFalpha and FGF7 in morphogenesis of mouse mammary epithelium. , 2007, Developmental biology.

[35]  A. Gown,et al.  Immunohistochemical and Clinical Characterization of the Basal-Like Subtype of Invasive Breast Carcinoma , 2004, Clinical Cancer Research.

[36]  J. Rich,et al.  Growth factor receptors define cancer hierarchies. , 2013, Cancer cell.

[37]  F. Martin,et al.  Mitogen-activated protein kinase signalling in experimental models of breast cancer progression and in mammary gland development , 2009 .

[38]  C. Eaves,et al.  Deciphering the Mammary Epithelial Cell Hierarchy , 2006, Cell cycle.

[39]  C. Moskaluk,et al.  Sustained activation of the HER1-ERK1/2-RSK signaling pathway controls myoepithelial cell fate in human mammary tissue. , 2011, Genes & development.

[40]  M. Okabe,et al.  Activation of Akt signaling is sufficient to maintain pluripotency in mouse and primate embryonic stem cells , 2006, Oncogene.

[41]  M. Vivanco,et al.  Growth and differentiation of progenitor/stem cells derived from the human mammary gland. , 2004, Experimental cell research.

[42]  Dulce Maroni,et al.  TGFB1 disrupts the angiogenic potential of microvascular endothelial cells of the corpus luteum , 2011, Journal of Cell Science.

[43]  Helmut Dotzlaw,et al.  Epidermal growth factor gene expression in human breast cancer biopsy samples: relationship to estrogen and progesterone receptor gene expression. , 1990, Cancer research.

[44]  Mina J. Bissell,et al.  Evidence for a stem cell hierarchy in the adult human breast , 2007, The Journal of cell biology.

[45]  A. Harris,et al.  Amphiregulin, epidermal growth factor receptor, and estrogen receptor expression in human primary breast cancer. , 1993, Cancer research.

[46]  S. Naber,et al.  Akt1 ablation inhibits, whereas Akt2 ablation accelerates, the development of mammary adenocarcinomas in mouse mammary tumor virus (MMTV)-ErbB2/neu and MMTV-polyoma middle T transgenic mice. , 2007, Cancer research.

[47]  S. Fox,et al.  Aberrant luminal progenitors as the candidate target population for basal tumor development in BRCA1 mutation carriers , 2009, Nature Medicine.

[48]  Robert A. Weinberg,et al.  Creation of human tumour cells with defined genetic elements , 1999, Nature.