Unraveling Heterogeneity in Epithelial Cell Fates of the Mammary Gland and Breast Cancer
Abstract
:1. Introduction into Mammary Gland Structure, Function and Early Development
2. Luminal Cells and Luminal-Specific Progenitors
3. Basal Cells and Progenitors
4. Lineage Tracing to Identify Restricted and Bipotent Progenitor Cells
5. Three-Dimensional Spheroid Cultures
6. EGFR Family and Ligands
7. EGFR during Mammary Gland Development
8. EGFR Signal Strength, Downstream Effector Kinases, Cell Fate
9. EGFR and Other Receptor Tyrosine Kinases Signaling and Ductal Morphogenesis
10. ERFR in Breast Cancer and Oncogenic PI3K Signals That Switch Fate
11. The Mammary Stem Cell Conundrum: More Questions than Answers
12. Organoids to Assess Stem- and Progenitor-Potential
13. Reconstructing Mammary Epithelial Cell Types and States Using Single-Cell Genomics
14. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Macias, H.; Hinck, L. Mammary gland development. Wiley Interdiscip. Rev. Dev. Biol. 2012, 1, 533–557. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hogg, N.A.; Harrison, C.J.; Tickle, C. Lumen formation in the developing mouse mammary gland. J. Embryol. Exp. Morphol. 1983, 73, 39–57. [Google Scholar] [PubMed]
- Visvader, J.E. Keeping abreast of the mammary epithelial hierarchy and breast tumorigenesis. Genes Dev. 2009, 23, 2563–2577. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hinck, L.; Silberstein, G.B. Key stages in mammary gland development: the mammary end bud as a motile organ. Breast Cancer Res. 2005, 7, 245–251. [Google Scholar] [CrossRef] [PubMed]
- Sternlicht, M.D.; Kouros-Mehr, H.; Lu, P.; Werb, Z. Hormonal and local control of mammary branching morphogenesis. Differentiation 2006, 74, 365–381. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Williams, J.M.; Daniel, C.W. Mammary ductal elongation: differentiation of myoepithelium and basal lamina during branching morphogenesis. Dev. Biol. 1983, 97, 274–290. [Google Scholar] [CrossRef]
- Visvader, J.E.; Stingl, J. Mammary stem cells and the differentiation hierarchy: Current status and perspectives. Genes Dev. 2014, 28, 1143–1158. [Google Scholar] [CrossRef] [PubMed]
- Oakes, S.R.; Naylor, M.J.; Asselin-Labat, M.L.; Blazek, K.D.; Gardiner-Garden, M.; Hilton, H.N.; Kazlauskas, M.; Pritchard, M.A.; Chodosh, L.A.; Pfeffer, P.L.; et al. The Ets transcription factor Elf5 specifies mammary alveolar cell fate. Genes Dev. 2008, 22, 581–586. [Google Scholar] [CrossRef] [Green Version]
- Chakrabarti, R.; Wei, Y.; Romano, R.A.; DeCoste, C.; Kang, Y.; Sinha, S. Elf5 regulates mammary gland stem/progenitor cell fate by influencing notch signaling. Stem Cells 2012, 30, 1496–1508. [Google Scholar] [CrossRef]
- Lee, H.J.; Gallego-Ortega, D.; Ledger, A.; Schramek, D.; Joshi, P.; Szwarc, M.M.; Cho, C.; Lydon, J.P.; Khokha, R.; Penninger, J.M.; et al. Progesterone drives mammary secretory differentiation via RankL-mediated induction of Elf5 in luminal progenitor cells. Development 2013, 140, 1397–1401. [Google Scholar] [CrossRef] [Green Version]
- Kouros-Mehr, H.; Slorach, E.M.; Sternlicht, M.D.; Werb, Z. GATA-3 maintains the differentiation of the luminal cell fate in the mammary gland. Cell 2006, 127, 1041–1055. [Google Scholar] [CrossRef] [PubMed]
- Yamaji, D.; Na, R.; Feuermann, Y.; Pechhold, S.; Chen, W.; Robinson, G.W.; Hennighausen, L. Development of mammary luminal progenitor cells is controlled by the transcription factor STAT5A. Genes Dev. 2009, 23, 2382–2387. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Asselin-Labat, M.L.; Sutherland, K.D.; Barker, H.; Thomas, R.; Shackleton, M.; Forrest, N.C.; Hartley, L.; Robb, L.; Grosveld, F.G.; van der Wees, J.; et al. Gata-3 is an essential regulator of mammary-gland morphogenesis and luminal-cell differentiation. Nat. Cell Biol. 2007, 9, 201–209. [Google Scholar] [CrossRef] [PubMed]
- Shehata, M.; Teschendorff, A.; Sharp, G.; Novcic, N.; Russell, I.A.; Avril, S.; Prater, M.; Eirew, P.; Caldas, C.; Watson, C.J.; et al. Phenotypic and functional characterisation of the luminal cell hierarchy of the mammary gland. Breast Cancer Res. 2012, 14, e134. [Google Scholar] [CrossRef] [PubMed]
- Regan, J.L.; Kendrick, H.; Magnay, F.A.; Vafaizadeh, V.; Groner, B.; Smalley, M.J. c-Kit is required for growth and survival of the cells of origin of Brca1-mutation-associated breast cancer. Oncogene 2012, 31, 869–883. [Google Scholar] [CrossRef] [PubMed]
- Asselin-Labat, M.L.; Sutherland, K.D.; Vaillant, F.; Gyorki, D.E.; Wu, D.; Holroyd, S.; Breslin, K.; Ward, T.; Shi, W.; Bath, M.L.; et al. Gata-3 negatively regulates the tumor-initiating capacity of mammary luminal progenitor cells and targets the putative tumor suppressor caspase-14. Mol. Cell Biol. 2011, 31, 4609–4622. [Google Scholar] [CrossRef] [PubMed]
- Nassour, M.; Idoux-Gillet, Y.; Selmi, A.; Come, C.; Faraldo, M.L.; Deugnier, M.A.; Savagner, P. Slug controls stem/progenitor cell growth dynamics during mammary gland morphogenesis. PLoS ONE 2012, 7, e53498. [Google Scholar] [CrossRef] [PubMed]
- Guo, W.; Keckesova, Z.; Donaher, J.L.; Shibue, T.; Tischler, V.; Reinhardt, F.; Itzkovitz, S.; Noske, A.; Zurrer-Hardi, U.; Bell, G.; et al. Slug and Sox9 cooperatively determine the mammary stem cell state. Cell 2012, 148, 1015–1028. [Google Scholar] [CrossRef]
- Mills, A.A.; Zheng, B.; Wang, X.J.; Vogel, H.; Roop, D.R.; Bradley, A. p63 is a p53 homologue required for limb and epidermal morphogenesis. Nature 1999, 398, 708–713. [Google Scholar] [CrossRef]
- Yalcin-Ozuysal, O.; Fiche, M.; Guitierrez, M.; Wagner, K.U.; Raffoul, W.; Brisken, C. Antagonistic roles of Notch and p63 in controlling mammary epithelial cell fates. Cell Death Differ. 2010, 17, 1600–1612. [Google Scholar] [CrossRef] [Green Version]
- Bouras, T.; Pal, B.; Vaillant, F.; Harburg, G.; Asselin-Labat, M.L.; Oakes, S.R.; Lindeman, G.J.; Visvader, J.E. Notch signaling regulates mammary stem cell function and luminal cell-fate commitment. Cell Stem Cell 2008, 3, 429–441. [Google Scholar] [CrossRef]
- Cicalese, A.; Bonizzi, G.; Pasi, C.E.; Faretta, M.; Ronzoni, S.; Giulini, B.; Brisken, C.; Minucci, S.; Di Fiore, P.P.; Pelicci, P.G. The tumor suppressor p53 regulates polarity of self-renewing divisions in mammary stem cells. Cell 2009, 138, 1083–1095. [Google Scholar] [CrossRef] [PubMed]
- Van Keymeulen, A.; Rocha, A.S.; Ousset, M.; Beck, B.; Bouvencourt, G.; Rock, J.; Sharma, N.; Dekoninck, S.; Blanpain, C. Distinct stem cells contribute to mammary gland development and maintenance. Nature 2011, 479, 189–193. [Google Scholar] [CrossRef] [PubMed]
- Rios, A.C.; Fu, N.Y.; Lindeman, G.J.; Visvader, J.E. In situ identification of bipotent stem cells in the mammary gland. Nature 2014, 506, 322–327. [Google Scholar] [CrossRef] [PubMed]
- Simian, M.; Bissell, M.J. Organoids: A historical perspective of thinking in three dimensions. J. Cell Biol. 2017, 216, 31–40. [Google Scholar] [CrossRef] [PubMed]
- Orkin, R.W.; Gehron, P.; McGoodwin, E.B.; Martin, G.R.; Valentine, T.; Swarm, R. A murine tumor producing a matrix of basement membrane. J. Exp. Med. 1977, 145, 204–220. [Google Scholar] [CrossRef]
- Barcellos-Hoff, M.H.; Aggeler, J.; Ram, T.G.; Bissell, M.J. Functional differentiation and alveolar morphogenesis of primary mammary cultures on reconstituted basement membrane. Development 1989, 105, 223–235. [Google Scholar] [PubMed]
- Petersen, O.W.; Ronnov-Jessen, L.; Howlett, A.R.; Bissell, M.J. Interaction with basement membrane serves to rapidly distinguish growth and differentiation pattern of normal and malignant human breast epithelial cells. Proc. Natl. Acad. Sci. USA 1992, 89, 9064–9068. [Google Scholar] [CrossRef]
- Wang, F.; Weaver, V.M.; Petersen, O.W.; Larabell, C.A.; Dedhar, S.; Briand, P.; Lupu, R.; Bissell, M.J. Reciprocal interactions between beta1-integrin and epidermal growth factor receptor in three-dimensional basement membrane breast cultures: a different perspective in epithelial biology. Proc. Natl. Acad. Sci. USA 1998, 95, 14821–14826. [Google Scholar] [CrossRef]
- Pechoux, C.; Gudjonsson, T.; Ronnov-Jessen, L.; Bissell, M.J.; Petersen, O.W. Human mammary luminal epithelial cells contain progenitors to myoepithelial cells. Dev. Biol. 1999, 206, 88–99. [Google Scholar] [CrossRef]
- Dontu, G.; Abdallah, W.M.; Foley, J.M.; Jackson, K.W.; Clarke, M.F.; Kawamura, M.J.; Wicha, M.S. In vitro propagation and transcriptional profiling of human mammary stem/progenitor cells. Genes Dev. 2003, 17, 1253–1270. [Google Scholar] [CrossRef] [Green Version]
- Eccles, S.A. The epidermal growth factor receptor/Erb-B/HER family in normal and malignant breast biology. Int. J. Dev. Biol. 2011, 55, 685–696. [Google Scholar] [CrossRef]
- Shi, F.; Telesco, S.E.; Liu, Y.; Radhakrishnan, R.; Lemmon, M.A. ErbB3/HER3 intracellular domain is competent to bind ATP and catalyze autophosphorylation. Proc. Natl. Acad. Sci. USA 2010, 107, 7692–7697. [Google Scholar] [CrossRef] [Green Version]
- Hynes, N.E.; Watson, C.J. Mammary gland growth factors: roles in normal development and in cancer. Cold Spring Harb. Perspect. Biol. 2010, 2, e003186. [Google Scholar] [CrossRef]
- Normanno, N.; Bianco, C.; De Luca, A.; Salomon, D.S. The role of EGF-related peptides in tumor growth. Front. Biosci. 2001, 6, 685–707. [Google Scholar] [CrossRef]
- Riese, D.J.; Kim, E.D.; Elenius, K.; Buckley, S.; Klagsbrun, M.; Plowman, G.D.; Stern, D.F. The epidermal growth factor receptor couples transforming growth factor-alpha, heparin-binding epidermal growth factor-like factor, and amphiregulin to Neu, ErbB-3, and ErbB-4. J. Biol. Chem. 1996, 271, 20047–20052. [Google Scholar] [CrossRef]
- Moriki, T.; Maruyama, H.; Maruyama, I.N. Activation of preformed EGF receptor dimers by ligand-induced rotation of the transmembrane domain. J. Mol. Biol. 2001, 311, 1011–1026. [Google Scholar] [CrossRef]
- Sternlicht, M.D.; Sunnarborg, S.W. The ADAM17-amphiregulin-EGFR axis in mammary development and cancer. J. Mammary Gland Biol. Neoplasia 2008, 13, 181–194. [Google Scholar] [CrossRef]
- Miettinen, P.J.; Berger, J.E.; Meneses, J.; Phung, Y.; Pedersen, R.A.; Werb, Z.; Derynck, R. Epithelial immaturity and multiorgan failure in mice lacking epidermal growth factor receptor. Nature 1995, 376, 337–341. [Google Scholar] [CrossRef]
- Threadgill, D.W.; Dlugosz, A.A.; Hansen, L.A.; Tennenbaum, T.; Lichti, U.; Yee, D.; LaMantia, C.; Mourton, T.; Herrup, K.; Harris, R.C.; et al. Targeted disruption of mouse EGF receptor: effect of genetic background on mutant phenotype. Science 1995, 269, 230–234. [Google Scholar] [CrossRef]
- Luetteke, N.C.; Phillips, H.K.; Qiu, T.H.; Copeland, N.G.; Earp, H.S.; Jenkins, N.A.; Lee, D.C. The mouse waved-2 phenotype results from a point mutation in the EGF receptor tyrosine kinase. Genes Dev. 1994, 8, 399–413. [Google Scholar] [CrossRef]
- Fowler, K.J.; Walker, F.; Alexander, W.; Hibbs, M.L.; Nice, E.C.; Bohmer, R.M.; Mann, G.B.; Thumwood, C.; Maglitto, R.; Danks, J.A.; 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. Proc. Natl. Acad. Sci. USA 1995, 92, 1465–1469. [Google Scholar] [CrossRef]
- Sebastian, J.; Richards, R.G.; Walker, M.P.; Wiesen, J.F.; Werb, Z.; Derynck, R.; Hom, Y.K.; Cunha, G.R.; DiAugustine, R.P. Activation and function of the epidermal growth factor receptor and erbB-2 during mammary gland morphogenesis. Cell Growth Differ. 1998, 9, 777–785. [Google Scholar]
- Xie, W.; Paterson, A.J.; Chin, E.; Nabell, L.M.; Kudlow, J.E. Targeted expression of a dominant negative epidermal growth factor receptor in the mammary gland of transgenic mice inhibits pubertal mammary duct development. Mol. Endocrinol. 1997, 11, 1766–1781. [Google Scholar] [CrossRef]
- Sternlicht, M.D.; Sunnarborg, S.W.; Kouros-Mehr, H.; Yu, Y.; Lee, D.C.; Werb, Z. Mammary ductal morphogenesis requires paracrine activation of stromal EGFR via ADAM17-dependent shedding of epithelial amphiregulin. Development 2005, 132, 3923–3933. [Google Scholar] [CrossRef] [Green Version]
- Wiesen, J.F.; Young, P.; Werb, Z.; Cunha, G.R. Signaling through the stromal epidermal growth factor receptor is necessary for mammary ductal development. Development 1999, 126, 335–344. [Google Scholar]
- Andrechek, E.R.; White, D.; Muller, W.J. Targeted disruption of ErbB2/Neu in the mammary epithelium results in impaired ductal outgrowth. Oncogene 2005, 24, 932–937. [Google Scholar] [CrossRef]
- Jackson-Fisher, A.J.; Bellinger, G.; Ramabhadran, R.; Morris, J.K.; Lee, K.F.; Stern, D.F. ErbB2 is required for ductal morphogenesis of the mammary gland. Proc. Natl. Acad. Sci. USA 2004, 101, 17138–17143. [Google Scholar] [CrossRef] [Green Version]
- Pasic, L.; Eisinger-Mathason, T.S.; Velayudhan, B.T.; Moskaluk, C.A.; Brenin, D.R.; Macara, I.G.; Lannigan, D.A. Sustained activation of the HER1-ERK1/2-RSK signaling pathway controls myoepithelial cell fate in human mammary tissue. Genes Dev. 2011, 25, 1641–1653. [Google Scholar] [CrossRef]
- Mukhopadhyay, C.; Zhao, X.; Maroni, D.; Band, V.; Naramura, M. Distinct effects of EGFR ligands on human mammary epithelial cell differentiation. PLoS ONE 2013, 8, e75907. [Google Scholar] [CrossRef]
- Marshall, C.J. Specificity of receptor tyrosine kinase signaling: transient versus sustained extracellular signal-regulated kinase activation. Cell 1995, 80, 179–185. [Google Scholar] [CrossRef]
- Metzger, R.J.; Klein, O.D.; Martin, G.R.; Krasnow, M.A. The branching programme of mouse lung development. Nature 2008, 453, 745–750. [Google Scholar] [CrossRef] [Green Version]
- Schedin, P.; Keely, P.J. Mammary gland ECM remodeling, stiffness, and mechanosignaling in normal development and tumor progression. Cold Spring Harb. Perspect. Biol. 2011, 3, e003228. [Google Scholar] [CrossRef]
- Brisken, C.; O’Malley, B. Hormone action in the mammary gland. Cold Spring Harb. Perspect. Biol. 2010, 2, e003178. [Google Scholar] [CrossRef]
- Bocchinfuso, W.P.; Korach, K.S. Mammary gland development and tumorigenesis in estrogen receptor knockout mice. J. Mammary Gland Biol. Neoplasia 1997, 2, 323–334. [Google Scholar] [CrossRef]
- Daniel, C.W.; Silberstein, G.B.; Strickland, P. Direct action of 17 beta-estradiol on mouse mammary ducts analyzed by sustained release implants and steroid autoradiography. Cancer Res. 1987, 47, 6052–6057. [Google Scholar]
- Zhang, H.Z.; Bennett, J.M.; Smith, K.T.; Sunil, N.; Haslam, S.Z. Estrogen mediates mammary epithelial cell proliferation in serum-free culture indirectly via mammary stroma-derived hepatocyte growth factor. Endocrinology 2002, 143, 3427–3434. [Google Scholar] [CrossRef]
- Coleman, S.; Silberstein, G.B.; Daniel, C.W. Ductal morphogenesis in the mouse mammary gland: evidence supporting a role for epidermal growth factor. Dev. Biol. 1988, 127, 304–315. [Google Scholar] [CrossRef]
- Gallego, M.I.; Binart, N.; Robinson, G.W.; Okagaki, R.; Coschigano, K.T.; Perry, J.; Kopchick, J.J.; Oka, T.; Kelly, P.A.; Hennighausen, L. Prolactin, growth hormone, and epidermal growth factor activate Stat5 in different compartments of mammary tissue and exert different and overlapping developmental effects. Dev. Biol. 2001, 229, 163–175. [Google Scholar] [CrossRef]
- Kleinberg, D.L.; Feldman, M.; Ruan, W. IGF-I: an essential factor in terminal end bud formation and ductal morphogenesis. J. Mammary Gland Biol. Neoplasia 2000, 5, 7–17. [Google Scholar] [CrossRef]
- Xu, X.; Weinstein, M.; Li, C.; Naski, M.; Cohen, R.I.; Ornitz, D.M.; Leder, P.; Deng, C. Fibroblast growth factor receptor 2 (FGFR2)-mediated reciprocal regulation loop between FGF8 and FGF10 is essential for limb induction. Development 1998, 125, 753–765. [Google Scholar]
- Lu, P.; Ewald, A.J.; Martin, G.R.; Werb, Z. Genetic mosaic analysis reveals FGF receptor 2 function in terminal end buds during mammary gland branching morphogenesis. Dev. Biol. 2008, 321, 77–87. [Google Scholar] [CrossRef] [Green Version]
- Parsa, S.; Ramasamy, S.K.; De Langhe, S.; Gupte, V.V.; Haigh, J.J.; Medina, D.; Bellusci, S. Terminal end bud maintenance in mammary gland is dependent upon FGFR2b signaling. Dev. Biol. 2008, 317, 121–131. [Google Scholar] [CrossRef]
- Jackson-Fisher, A.J.; Bellinger, G.; Breindel, J.L.; Tavassoli, F.A.; Booth, C.J.; Duong, J.K.; Stern, D.F. ErbB3 is required for ductal morphogenesis in the mouse mammary gland. Breast Cancer Res. 2008, 10, e96. [Google Scholar] [CrossRef]
- Tidcombe, H.; Jackson-Fisher, A.; Mathers, K.; Stern, D.F.; Gassmann, M.; Golding, J.P. Neural and mammary gland defects in ErbB4 knockout mice genetically rescued from embryonic lethality. Proc Natl. Acad. Sci. USA 2003, 100, 8281–8286. [Google Scholar] [CrossRef] [Green Version]
- Schlessinger, J. Ligand-induced, receptor-mediated dimerization and activation of EGF receptor. Cell 2002, 110, 669–672. [Google Scholar] [CrossRef]
- Zhang, X.; Gureasko, J.; Shen, K.; Cole, P.A.; Kuriyan, J. An allosteric mechanism for activation of the kinase domain of epidermal growth factor receptor. Cell 2006, 125, 1137–1149. [Google Scholar] [CrossRef]
- Jura, N.; Shan, Y.; Cao, X.; Shaw, D.E.; Kuriyan, J. Structural analysis of the catalytically inactive kinase domain of the human EGF receptor 3. Proc. Natl. Acad. Sci. USA 2009, 106, 21608–21613. [Google Scholar] [CrossRef] [Green Version]
- Holbro, T.; Beerli, R.R.; Maurer, F.; Koziczak, M.; Barbas, C.F.; Hynes, N.E. The ErbB2/ErbB3 heterodimer functions as an oncogenic unit: ErbB2 requires ErbB3 to drive breast tumor cell proliferation. Proc. Natl. Acad. Sci. USA 2003, 100, 8933–8938. [Google Scholar] [CrossRef] [Green Version]
- Huebner, R.J.; Neumann, N.M.; Ewald, A.J. Mammary epithelial tubes elongate through MAPK-dependent coordination of cell migration. Development 2016, 143, 983–993. [Google Scholar] [CrossRef] [Green Version]
- Koren, S.; Bentires-Alj, M. Breast tumor heterogeneity: Source of fitness, hurdle for therapy. Mol. Cell 2015, 60, 537–546. [Google Scholar] [CrossRef]
- Koren, S.; Reavie, L.; Couto, J.P.; De Silva, D.; Stadler, M.B.; Roloff, T.; Britschgi, A.; Eichlisberger, T.; Kohler, H.; Aina, O.; et al. PIK3CA(H1047R) induces multipotency and multi-lineage mammary tumours. Nature 2015, 525, 114–118. [Google Scholar] [CrossRef]
- Van Keymeulen, A.; Lee, M.Y.; Ousset, M.; Brohee, S.; Rorive, S.; Giraddi, R.R.; Wuidart, A.; Bouvencourt, G.; Dubois, C.; Salmon, I.; et al. Reactivation of multipotency by oncogenic PIK3CA induces breast tumour heterogeneity. Nature 2015, 525, 119–123. [Google Scholar] [CrossRef]
- Kaimala, S.; Bisana, S.; Kumar, S. Mammary gland stem cells: More puzzles than explanations. J. Biosci. 2012, 37, 349–358. [Google Scholar] [CrossRef]
- Moraes, R.C.; Zhang, X.; Harrington, N.; Fung, J.Y.; Wu, M.F.; Hilsenbeck, S.G.; Allred, D.C.; Lewis, M.T. Constitutive activation of smoothened (SMO) in mammary glands of transgenic mice leads to increased proliferation, altered differentiation and ductal dysplasia. Development 2007, 134, 1231–1242. [Google Scholar] [CrossRef] [Green Version]
- Shackleton, M.; Vaillant, F.; Simpson, K.J.; Stingl, J.; Smyth, G.K.; Asselin-Labat, M.L.; Wu, L.; Lindeman, G.J.; Visvader, J.E. Generation of a functional mammary gland from a single stem cell. Nature 2006, 439, 84–88. [Google Scholar] [CrossRef]
- Lim, E.; Vaillant, F.; Wu, D.; Forrest, N.C.; Pal, B.; Hart, A.H.; Asselin-Labat, M.L.; Gyorki, D.E.; Ward, T.; Partanen, A.; et al. Aberrant luminal progenitors as the candidate target population for basal tumor development in BRCA1 mutation carriers. Nat. Med. 2009, 15, 907–913. [Google Scholar] [CrossRef]
- Sleeman, K.E.; Kendrick, H.; Ashworth, A.; Isacke, C.M.; Smalley, M.J. CD24 staining of mouse mammary gland cells defines luminal epithelial, myoepithelial/basal and non-epithelial cells. Breast Cancer Res. 2006, 8, e7. [Google Scholar] [CrossRef]
- Jones, C.; Mackay, A.; Grigoriadis, A.; Cossu, A.; Reis-Filho, J.S.; Fulford, L.; Dexter, T.; Davies, S.; Bulmer, K.; Ford, E.; et al. Expression profiling of purified normal human luminal and myoepithelial breast cells: identification of novel prognostic markers for breast cancer. Cancer Res. 2004, 64, 3037–3045. [Google Scholar] [CrossRef]
- Kawase, Y.; Yanagi, Y.; Takato, T.; Fujimoto, M.; Okochi, H. Characterization of multipotent adult stem cells from the skin: Transforming growth factor-beta (TGF-beta) facilitates cell growth. Exp. Cell Res. 2004, 295, 194–203. [Google Scholar] [CrossRef]
- Tai, M.H.; Chang, C.C.; Kiupel, M.; Webster, J.D.; Olson, L.K.; Trosko, J.E. Oct4 expression in adult human stem cells: evidence in support of the stem cell theory of carcinogenesis. Carcinogenesis 2005, 26, 495–502. [Google Scholar] [CrossRef]
- Liu, B.Y.; McDermott, S.P.; Khwaja, S.S.; Alexander, C.M. The transforming activity of Wnt effectors correlates with their ability to induce the accumulation of mammary progenitor cells. Proc. Natl. Acad. Sci. USA 2004, 101, 4158–4163. [Google Scholar] [CrossRef] [Green Version]
- Bai, L.; Rohrschneider, L.R. s-SHIP promoter expression marks activated stem cells in developing mouse mammary tissue. Genes Dev. 2010, 24, 1882–1892. [Google Scholar] [CrossRef] [Green Version]
- Barker, N.; Tan, S.; Clevers, H. Lgr proteins in epithelial stem cell biology. Development 2013, 140, 2484–2494. [Google Scholar] [CrossRef] [Green Version]
- Plaks, V.; Brenot, A.; Lawson, D.A.; Linnemann, J.R.; Van Kappel, E.C.; Wong, K.C.; de Sauvage, F.; Klein, O.D.; Werb, Z. Lgr5-expressing cells are sufficient and necessary for postnatal mammary gland organogenesis. Cell Rep. 2013, 3, 70–78. [Google Scholar] [CrossRef]
- de Visser, K.E.; Ciampricotti, M.; Michalak, E.M.; Tan, D.W.; Speksnijder, E.N.; Hau, C.S.; Clevers, H.; Barker, N.; Jonkers, J. Developmental stage-specific contribution of LGR5(+) cells to basal and luminal epithelial lineages in the postnatal mammary gland. J. Pathol. 2012, 228, 300–309. [Google Scholar] [CrossRef]
- Fu, N.Y.; Rios, A.C.; Pal, B.; Law, C.W.; Jamieson, P.; Liu, R.; Vaillant, F.; Jackling, F.; Liu, K.H.; Smyth, G.K.; et al. Identification of quiescent and spatially restricted mammary stem cells that are hormone responsive. Nat. Cell Biol. 2017, 19, 164–176. [Google Scholar] [CrossRef]
- Makarem, M.; Kannan, N.; Nguyen, L.V.; Knapp, D.J.; Balani, S.; Prater, M.D.; Stingl, J.; Raouf, A.; Nemirovsky, O.; Eirew, P.; et al. Developmental changes in the in vitro activated regenerative activity of primitive mammary epithelial cells. PLoS Biol. 2013, 11, e1001630. [Google Scholar] [CrossRef]
- Spike, B.T.; Engle, D.D.; Lin, J.C.; Cheung, S.K.; La, J.; Wahl, G.M. A mammary stem cell population identified and characterized in late embryogenesis reveals similarities to human breast cancer. Cell Stem Cell 2012, 10, 183–197. [Google Scholar] [CrossRef]
- Pal, B.; Bouras, T.; Shi, W.; Vaillant, F.; Sheridan, J.M.; Fu, N.; Breslin, K.; Jiang, K.; Ritchie, M.E.; Young, M.; et al. Global changes in the mammary epigenome are induced by hormonal cues and coordinated by Ezh2. Cell Rep. 2013, 3, 411–426. [Google Scholar] [CrossRef]
- Kaanta, A.S.; Virtanen, C.; Selfors, L.M.; Brugge, J.S.; Neel, B.G. Evidence for a multipotent mammary progenitor with pregnancy-specific activity. Breast Cancer Res. 2013, 15, e65. [Google Scholar] [CrossRef]
- Pal, B.; Chen, Y.; Vaillant, F.; Jamieson, P.; Gordon, L.; Rios, A.C.; Wilcox, S.; Fu, N.; Liu, K.H.; Jackling, F.C.; et al. Construction of developmental lineage relationships in the mouse mammary gland by single-cell RNA profiling. Nat. Commun. 2017, 8, e1627. [Google Scholar] [CrossRef]
- Raouf, A.; Sun, Y.; Chatterjee, S.; Basak, P. The biology of human breast epithelial progenitors. Semin. Cell Dev. Biol. 2012, 23, 606–612. [Google Scholar] [CrossRef]
- Clevers, H. Modeling Development and disease with organoids. Cell 2016, 165, 1586–1597. [Google Scholar] [CrossRef]
- Sato, T.; Clevers, H. Growing self-organizing mini-guts from a single intestinal stem cell: mechanism and applications. Science 2013, 340, 1190–1194. [Google Scholar] [CrossRef]
- Muranen, T.; Selfors, L.M.; Worster, D.T.; Iwanicki, M.P.; Song, L.; Morales, F.C.; Gao, S.; Mills, G.B.; Brugge, J.S. Inhibition of PI3K/mTOR leads to adaptive resistance in matrix-attached cancer cells. Cancer Cell 2012, 21, 227–239. [Google Scholar] [CrossRef]
- Schwank, G.; Clevers, H. Gastrointestinal Physiology and Diseases; Humana Press: New York, NY, USA, 2016; pp. 3–11. [Google Scholar]
- Takebe, T.; Sekine, K.; Enomura, M.; Koike, H.; Kimura, M.; Ogaeri, T.; Zhang, R.R.; Ueno, Y.; Zheng, Y.W.; Koike, N.; et al. Vascularized and functional human liver from an iPSC-derived organ bud transplant. Nature 2013, 499, 481–484. [Google Scholar] [CrossRef]
- Lancaster, M.A.; Knoblich, J.A. Organogenesis in a dish: Modeling development and disease using organoid technologies. Science 2014, 345, e1247125. [Google Scholar] [CrossRef]
- Fatehullah, A.; Tan, S.H.; Barker, N. Organoids as an in vitro model of human development and disease. Nat. Cell Biol. 2016, 18, 246–254. [Google Scholar] [CrossRef]
- Mroue, R.; Bissell, M.J. Epithelial Cell Culture Protocols; Humana Press: Totowa, NJ, USA, 2012; pp. 221–250. [Google Scholar]
- Ewald, A.J.; Brenot, A.; Duong, M.; Chan, B.S.; Werb, Z. Collective epithelial migration and cell rearrangements drive mammary branching morphogenesis. Dev. Cell 2008, 14, 570–581. [Google Scholar] [CrossRef]
- Nusse, R. Wnt signaling and stem cell control. Cell Res. 2008, 18, 523–527. [Google Scholar] [CrossRef] [Green Version]
- Mao, B.; Wu, W.; Li, Y.; Hoppe, D.; Stannek, P.; Glinka, A.; Niehrs, C. LDL-receptor-related protein 6 is a receptor for Dickkopf proteins. Nature 2001, 411, 321–325. [Google Scholar] [CrossRef]
- Behrens, J.; von Kries, J.P.; Kuhl, M.; Bruhn, L.; Wedlich, D.; Grosschedl, R.; Birchmeier, W. Functional interaction of beta-catenin with the transcription factor LEF-1. Nature 1996, 382, 638–642. [Google Scholar] [CrossRef]
- Molenaar, M.; van de Wetering, M.; Oosterwegel, M.; Peterson-Maduro, J.; Godsave, S.; Korinek, V.; Roose, J.; Destree, O.; Clevers, H. XTcf-3 transcription factor mediates beta-catenin-induced axis formation in Xenopus embryos. Cell 1996, 86, 391–399. [Google Scholar] [CrossRef]
- Roose, J.; Molenaar, M.; Peterson, J.; Hurenkamp, J.; Brantjes, H.; Moerer, P.; van de Wetering, M.; Destree, O.; Clevers, H. The Xenopus Wnt effector XTcf-3 interacts with Groucho-related transcriptional repressors. Nature 1998, 395, 608–612. [Google Scholar] [CrossRef]
- Cavallo, R.A.; Cox, R.T.; Moline, M.M.; Roose, J.; Polevoy, G.A.; Clevers, H.; Peifer, M.; Bejsovec, A. Drosophila Tcf and groucho interact to repress wingless signalling activity. Nature 1998, 395, 604–608. [Google Scholar] [CrossRef]
- Waltzer, L.; Bienz, M. The control of beta-catenin and TCF during embryonic development and cancer. Cancer Metastasis Rev. 1999, 18, 231–246. [Google Scholar] [CrossRef]
- Roose, J.; Clevers, H. TCF transcription factors: molecular switches in carcinogenesis. Biochim. Biophys. Acta 1999, 1424, M23–M37. [Google Scholar] [CrossRef]
- Leung, C.; Tan, S.H.; Barker, N. Recent advances in Lgr5(+) stem cell research. Trends Cell Biol. 2018, 28, 380–391. [Google Scholar] [CrossRef]
- De Lau, W.; Barker, N.; Low, T.Y.; Koo, B.K.; Li, V.S.; Teunissen, H.; Kujala, P.; Haegebarth, A.; Peters, P.J.; van de Wetering, M.; et al. Lgr5 homologues associate with Wnt receptors and mediate R-spondin signalling. Nature 2011, 476, 293–297. [Google Scholar] [CrossRef]
- Janda, C.Y.; Dang, L.T.; You, C.; Chang, J.; de Lau, W.; Zhong, Z.A.; Yan, K.S.; Marecic, O.; Siepe, D.; Li, X.; et al. Surrogate Wnt agonists that phenocopy canonical Wnt and beta-catenin signalling. Nature 2017, 545, 234–237. [Google Scholar] [CrossRef]
- Sachs, N.; de Ligt, J.; Kopper, O.; Gogola, E.; Bounova, G.; Weeber, F.; Balgobind, A.V.; Wind, K.; Gracanin, A.; Begthel, H.; et al. A living biobank of breast cancer organoids captures disease heterogeneity. Cell 2018, 172, 373–386.e10. [Google Scholar] [CrossRef]
- Sato, T.; Clevers, H. SnapShot: Growing organoids from stem cells. Cell 2015, 161, 1700–1701. [Google Scholar] [CrossRef]
- Yang, Y.; Spitzer, E.; Meyer, D.; Sachs, M.; Niemann, C.; Hartmann, G.; Weidner, K.M.; Birchmeier, C.; Birchmeier, W. Sequential requirement of hepatocyte growth factor and neuregulin in the morphogenesis and differentiation of the mammary gland. J. Cell Biol. 1995, 131, 215–226. [Google Scholar] [CrossRef]
- Liu, X.; Ory, V.; Chapman, S.; Yuan, H.; Albanese, C.; Kallakury, B.; Timofeeva, O.A.; Nealon, C.; Dakic, A.; Simic, V.; et al. ROCK inhibitor and feeder cells induce the conditional reprogramming of epithelial cells. Am. J. Pathol. 2012, 180, 599–607. [Google Scholar] [CrossRef]
- Tanay, A.; Regev, A. Scaling single-cell genomics from phenomenology to mechanism. Nature 2017, 541, 331–338. [Google Scholar] [CrossRef] [Green Version]
- Nguyen, Q.H.; Pervolarakis, N.; Blake, K.; Ma, D.; Davis, R.T.; James, N.; Phung, A.T.; Willey, E.; Kumar, R.; Jabart, E.; et al. Profiling human breast epithelial cells using single cell RNA sequencing identifies cell diversity. Nat. Commun. 2018, 9, e2028. [Google Scholar] [CrossRef]
- Bach, K.; Pensa, S.; Grzelak, M.; Hadfield, J.; Adams, D.J.; Marioni, J.C.; Khaled, W.T. Differentiation dynamics of mammary epithelial cells revealed by single-cell RNA sequencing. Nat. Commun. 2017, 8, e2128. [Google Scholar] [CrossRef]
- Chen, W.; Morabito, S.J.; Kessenbrock, K.; Enver, T.; Meyer, K.B.; Teschendorff, A.E. Teschendorff. Single-cell landscape in mammary epithelium reveals bipotent-like cells associated with breast cancer risk and outcome. Commun. Biol. 2019, 1–13. [Google Scholar]
- Chung, W.; Eum, H.H.; Lee, H.O.; Lee, K.M.; Lee, H.B.; Kim, K.T.; Ryu, H.S.; Kim, S.; Lee, J.E.; Park, Y.H.; et al. Single-cell RNA-seq enables comprehensive tumour and immune cell profiling in primary breast cancer. Nat. Commun. 2017, 8, e15081. [Google Scholar] [CrossRef]
- Gao, R.; Kim, C.; Sei, E.; Foukakis, T.; Crosetto, N.; Chan, L.K.; Srinivasan, M.; Zhang, H.; Meric-Bernstam, F.; Navin, N. Nanogrid single-nucleus RNA sequencing reveals phenotypic diversity in breast cancer. Nat. Commun. 2017, 8, e228. [Google Scholar] [CrossRef]
- Azizi, E.; Carr, A.J.; Plitas, G.; Cornish, A.E.; Konopacki, C.; Prabhakaran, S.; Nainys, J.; Wu, K.; Kiseliovas, V.; Setty, M.; et al. Single-cell map of diverse immune phenotypes in the breast tumor microenvironment. Cell 2018, 174, 1293–1308.e136. [Google Scholar] [CrossRef]
- Hu, Q.T.; Hong, Y.; Qi, P.; Lu, G.Q.; Mai, X.Y.; Xu, S.; He, X.Y.; Guo, Y.; Gao, L.L.; Jing, Z.Y.; et al. An atlas of infiltrated B-lymphocytes in breast cancer revealed by paired single-cell RNA-sequencing and antigen receptor profiling. BioRxiv 2019, e695601. [Google Scholar]
- Wang, J.; Xu, R.; Yuan, H.; Zhang, Y.; Cheng, S. Single-cell RNA sequencing reveals novel gene expression signatures of trastuzumab treatment in HER2+ breast cancer: A pilot study. Medicine 2019, 98, e15872. [Google Scholar] [CrossRef]
- Buenrostro, J.D.; Wu, B.; Chang, H.Y.; Greenleaf, W.J. ATAC-seq: A method for assaying chromatin accessibility genome-wide. Curr. Protoc. Mol. Biol. 2015, 109, 21–29. [Google Scholar] [CrossRef]
- Buenrostro, J.D.; Wu, B.; Litzenburger, U.M.; Ruff, D.; Gonzales, M.L.; Snyder, M.P.; Chang, H.Y.; Greenleaf, W.J. Single-cell chromatin accessibility reveals principles of regulatory variation. Nature 2015, 523, 486–490. [Google Scholar] [CrossRef]
- Cusanovich, D.A.; Daza, R.; Adey, A.; Pliner, H.A.; Christiansen, L.; Gunderson, K.L.; Steemers, F.J.; Trapnell, C.; Shendure, J. Multiplex single cell profiling of chromatin accessibility by combinatorial cellular indexing. Science 2015, 348, 910–914. [Google Scholar] [CrossRef]
- Chung, C.Y.; Ma, Z.; Dravis, C.; Preissl, S.; Poirion, O.; Luna, G.; Hou, X.M.; Giraddi, R.R.; Ren, B.; Wahl, G.M. Single-cell chromatin accessibility analysis of mammary gland development reveals cell state transcriptional regulators and cellular lineage relationships. BioRxiv 2019, e624957. [Google Scholar]
- Pervolarakis, N.; Sun, P.; Gutierrez, G.; Nguyen, Q.H.; Jhutty, D.; Zheng, G.X.Y.; Nemec, C.M.; Dai, X.; Watanabe, K.; Kessenbrock, K. Integrated single-cell transcriptomics and chromatin accessibility analysis reveals novel regulators of mammary epithelial cell identity. Cell Rep. 2019, 1–39. [Google Scholar] [CrossRef]
EGFR Family | EGFR (ErbB1/Her1) | EGFR2 (ErbB2/Her2) | EGFR3 (ErbB3/Her3) | EGFR4 (ErbB4/Her4) |
---|---|---|---|---|
EGF | X | |||
TGF | X | |||
AREG | X | |||
EPG | X | |||
BTC | X | X | ||
HB-EGF | X | X | ||
EPR | X | X | ||
NRG1 | X | X | ||
NRG2 | X | X | ||
NRG3 | X |
© 2019 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Samocha, A.; Doh, H.; Kessenbrock, K.; Roose, J.P. Unraveling Heterogeneity in Epithelial Cell Fates of the Mammary Gland and Breast Cancer. Cancers 2019, 11, 1423. https://doi.org/10.3390/cancers11101423
Samocha A, Doh H, Kessenbrock K, Roose JP. Unraveling Heterogeneity in Epithelial Cell Fates of the Mammary Gland and Breast Cancer. Cancers. 2019; 11(10):1423. https://doi.org/10.3390/cancers11101423
Chicago/Turabian StyleSamocha, Alexandr, Hanna Doh, Kai Kessenbrock, and Jeroen P. Roose. 2019. "Unraveling Heterogeneity in Epithelial Cell Fates of the Mammary Gland and Breast Cancer" Cancers 11, no. 10: 1423. https://doi.org/10.3390/cancers11101423
APA StyleSamocha, A., Doh, H., Kessenbrock, K., & Roose, J. P. (2019). Unraveling Heterogeneity in Epithelial Cell Fates of the Mammary Gland and Breast Cancer. Cancers, 11(10), 1423. https://doi.org/10.3390/cancers11101423