Wnt and Neuregulin1/ErbB signalling extends 3D culture of hormone responsive mammary organoids

The development of in vitro culture systems quantitatively and qualitatively recapitulating normal breast biology is key to the understanding of mammary gland biology. Current three-dimensional mammary culture systems have not demonstrated concurrent proliferation and functional differentiation ex vivo in any system for longer than 2 weeks. Here, we identify conditions including Neuregulin1 and R-spondin 1, allowing maintenance and expansion of mammary organoids for 2.5 months in culture. The organoids comprise distinct basal and luminal compartments complete with functional steroid receptors and stem/progenitor cells able to reconstitute a complete mammary gland in vivo. Alternative conditions are also described that promote enrichment of basal cells organized into multiple layers surrounding a keratinous core, reminiscent of structures observed in MMTV-Wnt1 tumours. These conditions comprise a unique tool that should further understanding of normal mammary gland development, the molecular mechanism of hormone action and signalling events whose deregulation leads to breast tumourigenesis.

[1]  J. Rosen,et al.  On hormone action in the mammary gland. , 2012, Cold Spring Harbor perspectives in biology.

[2]  J. Visvader,et al.  Notch signaling regulates mammary stem cell function and luminal cell-fate commitment. , 2008, Cell stem cell.

[3]  C. Clarke,et al.  Progesterone induces adult mammary stem cell expansion , 2010, Nature.

[4]  M. Bissell,et al.  Three-dimensional cultures of mouse mammary epithelial cells. , 2013, Methods in molecular biology.

[5]  D. Edwards,et al.  Progesterone receptor and Stat5 signaling cross talk through RANKL in mammary epithelial cells. , 2013, Molecular endocrinology.

[6]  M. Smalley Isolation, culture and analysis of mouse mammary epithelial cells. , 2010, Methods in molecular biology.

[7]  Mina J Bissell,et al.  Collagen-IV and laminin-1 regulate estrogen receptor α expression and function in mouse mammary epithelial cells , 2003, Journal of Cell Science.

[8]  H. Clevers,et al.  Single Lgr5 stem cells build crypt–villus structures in vitro without a mesenchymal niche , 2009, Nature.

[9]  Hans Clevers,et al.  In vitro expansion of single Lgr5+ liver stem cells induced by Wnt-driven regeneration , 2013, Nature.

[10]  Andrew J. Ewald,et al.  Collective Invasion in Breast Cancer Requires a Conserved Basal Epithelial Program , 2013, Cell.

[11]  Jane E. Visvader,et al.  In situ identification of bipotent stem cells in the mammary gland , 2014, Nature.

[12]  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.

[13]  L. Ellisen,et al.  Basal cell signaling by p63 controls luminal progenitor function and lactation via NRG1. , 2014, Developmental cell.

[14]  L. Hennighausen,et al.  Mouse mammary epithelial cells express the Na-K-Cl cotransporter, NKCC1: characterization, localization, and involvement in ductal development and morphogenesis. , 2002, Molecular endocrinology.

[15]  J. Visvader,et al.  Control of mammary stem cell function by steroid hormone signalling , 2010, Nature.

[16]  K. Badani,et al.  Single luminal epithelial progenitors can generate prostate organoids in culture , 2014, Nature Cell Biology.

[17]  E. Cuppen,et al.  Identification of Multipotent Luminal Progenitor Cells in Human Prostate Organoid Cultures , 2014, Cell.

[18]  Andrew J Ewald,et al.  Collective epithelial migration and cell rearrangements drive mammary branching morphogenesis. , 2008, Developmental cell.

[19]  Mark D. Aupperlee,et al.  Progestin-regulated luminal cell and myoepithelial cell-specific responses in mammary organoid culture. , 2008, Endocrinology.

[20]  R. Nusse,et al.  Wnt proteins are self-renewal factors for mammary stem cells and promote their long-term expansion in culture. , 2010, Cell stem cell.

[21]  M J Bissell,et al.  The interplay of matrix metalloproteinases, morphogens and growth factors is necessary for branching of mammary epithelial cells. , 2001, Development.

[22]  A. Ashworth,et al.  Dissociation of estrogen receptor expression and in vivo stem cell activity in the mammary gland , 2007, The Journal of cell biology.

[23]  J. Hell,et al.  NS21: Re-defined and modified supplement B27 for neuronal cultures , 2008, Journal of Neuroscience Methods.

[24]  S. Nandi,et al.  Serum-free growth of normal and tumor mouse mammary epithelial cells in primary culture. , 1982, Proceedings of the National Academy of Sciences of the United States of America.

[25]  Q. Lin,et al.  R-spondins function as ligands of the orphan receptors LGR4 and LGR5 to regulate Wnt/β-catenin signaling , 2011, Proceedings of the National Academy of Sciences.

[26]  A. Ashworth,et al.  Pregnancy in the mature adult mouse does not alter the proportion of mammary epithelial stem/progenitor cells , 2009, Breast Cancer Research.

[27]  G. Brewer,et al.  Optimized survival of hippocampal neurons in B27‐supplemented neurobasal™, a new serum‐free medium combination , 1993, Journal of neuroscience research.

[28]  T. Dale,et al.  Wnt signalling in murine postnatal mammary gland development , 2012, Acta physiologica.

[29]  M. O'hare,et al.  Differentiation of Separated Mouse Mammary Luminal Epithelial and Myoepithelial Cells Cultured on EHS Matrix Analyzed by Indirect Immunofluorescence of Cytoskeletal Antigens , 1999, The journal of histochemistry and cytochemistry : official journal of the Histochemistry Society.

[30]  J. Rosen,et al.  Distinct roles of fibroblast growth factor receptor 1 and 2 in regulating cell survival and epithelial-mesenchymal transition. , 2007, Molecular endocrinology.

[31]  B. Katzenellenbogen,et al.  Progesterone receptor regulation in T47D human breast cancer cells: analysis by density labeling of progesterone receptor synthesis and degradation and their modulation by progestin. , 1988, Endocrinology.

[32]  A. Ashworth,et al.  Dynamic expression of Erbb pathway members during early mammary gland morphogenesis. , 2008, The Journal of investigative dermatology.

[33]  Alan Mackay,et al.  Identification of cellular and genetic drivers of breast cancer heterogeneity in genetically engineered mouse tumour models , 2014, The Journal of pathology.

[34]  J. Russo,et al.  Pattern of distribution of cells positive for estrogen receptor α and progesterone receptor in relation to proliferating cells in the mammary gland , 1999, Breast Cancer Research and Treatment.

[35]  A. Ashworth,et al.  CD24 staining of mouse mammary gland cells defines luminal epithelial, myoepithelial/basal and non-epithelial cells , 2005, Breast Cancer Research.

[36]  G. Feng,et al.  In vivo and in vitro models for the therapeutic targeting of Wnt signaling using a Tet-OΔN89β-catenin system , 2012, Oncogene.

[37]  F. Reyal,et al.  Luminal Progenitors Restrict Their Lineage Potential during Mammary Gland Development , 2015, PLoS biology.

[38]  E. Hrabeta-Robinson,et al.  Estrogen-triggered delays in mammary gland gene expression during the estrous cycle: evidence for a novel timing system. , 2006, The Journal of endocrinology.

[39]  C. Birchmeier,et al.  The breast proto-oncogene, HRGα regulates epithelial proliferation and lobuloalveolar development in the mouse mammary gland , 2002, Oncogene.

[40]  Rory Stark,et al.  Progesterone receptor modulates estrogen receptor-α action in breast cancer , 2015, Nature.

[41]  Hua Yu,et al.  R-spondin1 is a novel hormone mediator for mammary stem cell self-renewal , 2014, Genes & development.

[42]  L. Young,et al.  Influence of cell division on an aging process. Life span of mouse mammary epithelium during serial propagation in vivo. , 1971, Experimental Cell Research.

[43]  L. Cantley,et al.  A neu acquaintance for ErbB3 and ErbB4: A role for receptor heterodimerization in growth signaling , 1994, Cell.

[44]  D. Powell,et al.  A Versatile Strategy for Isolating a Highly Enriched Population of Intestinal Stem Cells , 2016, Stem cell reports.

[45]  W. Birchmeier,et al.  Sequential requirement of hepatocyte growth factor and neuregulin in the morphogenesis and differentiation of the mammary gland , 1995, The Journal of cell biology.

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

[47]  Hans Clevers,et al.  Unlimited in vitro expansion of adult bi-potent pancreas progenitors through the Lgr5/R-spondin axis , 2013, The EMBO journal.

[48]  Hans Clevers,et al.  Negative Feedback Loop of Wnt Signaling through Upregulation of Conductin/Axin2 in Colorectal and Liver Tumors , 2002, Molecular and Cellular Biology.

[49]  B. Groner,et al.  c-Kit is required for growth and survival of the cells of origin of Brca1-mutation-associated breast cancer , 2012, Oncogene.

[50]  J. Graham,et al.  DNA replication licensing and progenitor numbers are increased by progesterone in normal human breast. , 2009, Endocrinology.

[51]  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.

[52]  E. Betzig,et al.  A Localized Wnt Signal Orients Asymmetric Stem Cell Division in Vitro , 2013, Science.

[53]  Andrew J. Ewald,et al.  Three-dimensional organotypic culture: experimental models of mammalian biology and disease , 2014, Nature Reviews Molecular Cell Biology.

[54]  Hans Clevers,et al.  Lgr5(+ve) stem cells drive self-renewal in the stomach and build long-lived gastric units in vitro. , 2010, Cell stem cell.