Summary

Generation of Mosaic Mammary Organoids by Differential Trypsinization

Published: March 11, 2020
doi:

Summary

The mammary gland is a bilayered structure, comprising outer myoepithelial and inner luminal epithelial cells. Presented is a protocol to prepare organoids using differential trypsinization. This efficient method allows researchers to separately manipulate these two cell types to explore questions concerning their roles in mammary gland form and function.

Abstract

Organoids offer self-organizing, three-dimensional tissue structures that recapitulate physiological processes in the convenience of a dish. The murine mammary gland is composed of two distinct epithelial cell compartments, serving different functions: the outer, contractile myoepithelial compartment and the inner, secretory luminal compartment. Here, we describe a method by which the cells comprising these compartments are isolated and then combined to investigate their individual lineage contributions to mammary gland morphogenesis and differentiation. The method is simple and efficient and does not require sophisticated separation technologies such as fluorescence activated cell sorting. Instead, we harvest and enzymatically digest the tissue, seed the epithelium on adherent tissue culture dishes, and then use differential trypsinization to separate myoepithelial from luminal cells with ~90% purity. The cells are then plated in an extracellular matrix where they organize into bilayered, three-dimensional (3D) organoids that can be differentiated to produce milk after 10 days in culture. To test the effects of genetic mutations, cells can be harvested from wild type or genetically engineered mouse models, or they can be genetically manipulated prior to 3D culture. This technique can be used to generate mosaic organoids that allow investigation of gene function specifically in the luminal or myoepithelial compartment.

Introduction

The mammary gland (MG) is a tree-like, tubular epithelial structure embedded within an adipocyte rich stroma. The bilayered ductal epithelium comprises an outer, basal layer of contractile, myoepithelial cells (MyoECs) and an inner layer of luminal, secretory epithelial cells (LECs), encircling a central lumen1. During lactation when the outer MyoECs contract to squeeze milk from the inner alveolar LECs, the MG undergoes numerous changes that are under the control of growth factors (e.g., EGF and FGF) and hormones (e.g. progesterone, insulin, and prolactin). These changes cause the differentiation of specialized structures, alveoli, which synthesize and secrete milk during lactation1. The mammary epithelia can be experimentally manipulated using techniques in which either epithelial tissue fragments, cells, or even a single basal cell are transplanted into host mammary fat pads, precleared of endogenous mammary parenchyma, and allowed to grow out to reconstitute an entire, functional epithelial tree2,3,4,5. Transplantation is a powerful technique, but it is time-consuming and impossible if a mutation results in early embryonic lethality (prior to E14) that prevents the rescue of transplantable mammary anlage. Furthermore, investigators frequently wish to research the roles of the two different compartments, which are derived from lineage-restricted progenitor cells. While Cre-lox technology allows differential genetic manipulation of MyoECs and LECs, this is also a time-consuming and expensive undertaking. Thus, since the 1950s, investigators have used in vitro mammary organoids as a relatively easy and efficient way to address questions concerning mammary tissue structure and function6,7.

In early protocols describing the isolation and culture of primary mammary epithelial cells, investigators found that a basement membrane matrix (BME), composed of a plasma clot and chicken embryo extract, was required for MG fragments grown on a dish6. In the following decades, extracellular matrices (ECMs, collagen, and jellylike protein matrix secreted by Engelbreth-Holm-Swarm murine sarcoma cells) were developed to facilitate 3D culture and better mimic the in vivo environment7,8,9,10. Culturing cells in 3D matrices revealed by multiple criteria (morphology, gene expression, and hormone responsiveness) that such a microenvironment better models in vivo physiological processes9,10,11,12. Research using primary murine cells identified key growth factors and morphogens necessary for the extended maintenance and differentiation of organoids13. These studies have set the stage for the protocol presented here, and for the culture of human breast cells as 3D organoids, which is now a modern clinical tool, allowing for drug discovery and drug testing on patient samples14. Overall, organoid culturing highlights the self-organization capacities of primary cells and their contributions to morphogenesis and differentiation.

Presented here is a protocol to culture murine epithelia that can be differentiated into milk-producing acini. A differential trypsinization technique is used to isolate the MyoECs and LECs that comprise the two distinct MG cell compartments. These separated cell fractions can then be genetically manipulated to overexpress or knockdown gene function. Because lineage-intrinsic, self-organization is an innate property of mammary epithelial cells15,16,17, recombining these cell fractions allows researchers to generate bilayered, mosaic organoids. We begin by enzymatically digesting the adipose tissue, and then incubating the mammary fragments on a tissue culture dish for 24 h (Figure 1). The tissue fragments settle on polystyrene dishes as bilayered fragments with their in vivo organization: outer myoepithelial layer surrounding inner luminal layers. This cellular organization allows for the isolation of the outer MyoECs by trypsin-EDTA (0.05%) treatment for 3-6 min followed by a second round of trypsin-EDTA (0.05%) treatment that detaches the remaining inner LECs (Figure 2). Thus, these cell types with different trypsin sensitivity are isolated and can subsequently be mixed and plated in ECM (Figure 3). The cells undergo self-organization to form bilayered spheres, comprising an outer layer of MyoECs surrounding inner LECs. Lumen formation occurs as the cells grow in a medium containing a cocktail of growth factors (see recipes for Growth Medium)13. After 5 days, organoids can be differentiated into milk-producing acini by switching to Alveologenesis Medium (see recipes and Figure 3F) and incubated for another 5 days. Alternatively, organoids will continue to expand and branch in Growth Medium for at least 10 days. Organoids can be analyzed using immunofluorescence (Figure 3D-F) or released from the ECM using a recovery solution (see Table of Materials) and analyzed via other methods (e.g., immunoblot, RT-qPCR).

Protocol

All methods described here have been approved by the Institutional Animal Care and Use Committee (IACUC) of the University of California, Santa Cruz. 1. Day 1: Mammary gland digestion Prepare to harvest the MGs from mature female mice 10-14 weeks of age. Perform the harvesting on an open bench under aseptic conditions. Sterilize all surgical supplies, cork boards, and pins by autoclaving and soaking in 70% alcohol for 20 min prior to surgery. Anest…

Representative Results

The protocol presented here describes a method for investigating specific lineage contributions of mammary epithelial cells by making use of mosaic organoids. To obtain primary murine cells for organoids, the mammary gland epithelium must first be isolated from the surrounding adipocyte rich stroma (Figure 1). This process is described briefly here and is also described in a previously published study18. To obtain enough cells, it is r…

Discussion

Here, a method is presented detailing how researchers can generate 3D organoid cultures using primary MG cells. The difference between this and other protocols is that we detail a method to separate the two, distinct MG cell compartments: the outer basal MyoECs and inner LECs. Our method employs a two-step trypsin-EDTA (0.05%) treatment that we call differential trypsinization19. This procedure allows researchers to isolate MyoECs and LECs without using sophisticated flow cytometry and thus can be…

Divulgaciones

The authors have nothing to disclose.

Acknowledgements

We thank Ben Abrams for technical assistance and core support from the University of California, Santa Cruz (UCSC) Institute for the Biology of Stem Cells (IBSC). We thank Susan Strome and Bill Saxton for the use of their Solamere Spinning Disk Confocal Microscope. This work was supported in part by grants to UCSC from the Howard Hughes Medical Institute through the James H. Gilliam Fellowships for Advanced Study program (S.R.), from the NIH (NIH GM058903) for the initiative for maximizing student development (H.M.) and from the National Science Foundation for a graduate research fellowship (O.C. DGE 1339067) and by a grant (A18-0370) from the UC-Cancer Research Coordinating Committee (LH).

Materials

15 ml High-Clarity Polypropylene Conical Tube (BD Falcon) Fisher Scientific 352096
24 well ultra-low attachment plate (Corning) Fisher Scientific CLS3473-24EA
35 mm TC-treated Easy-Grip Style Cell Culture Dish (BD Falcon) Fisher Scientific 353001
50 ml High-Clarity polypropylene conical tube (BD Falcon) Fisher Scientific 352098
60 mm TC-treated Easy-Grip Style Cell Culture Dish (BD Falcon) Fisher Scientific 353004
70 µM nylon cell strainer (Corning) Fisher Scientific 08-771-2
Antibiotic-Antimycotic (100X) Thermo Fisher Scientific 15240062 Pen/Strep also works
B27 supplement without vitamin A (50x) Thermo Fisher Scientific 12587010
B6 ACTb-EGFP mice The Jackson Laboratory 003291
BD Insulin syringe 0.5 mL Thermo Fisher Scientific 14-826-79
Class 2 Dispase (Roche) Millipore Sigma 4942078001
Class 3 Collagenase Worthington Biochemical LS004206
Corning Cell Recovery solution Fisher Scientific 354253 Follow the guidelines for use – Extraction of Three-Dimensional Structures
from Corning Matrigel Matrix
Corning Costar Ultra-Low Attachment 6-well Fisher Scientific CLS3471
Dexamethasone Millipore Sigma D4902-25MG
DMEM/F12, no phenol red Thermo Fisher Scientific 11039-021
DNase (Deoxyribonuclease I) Worthington Biochemical LS002007
Donkey anti-Goat 647 Thermo Fisher Scientific A21447 Use at 1:500, Lot: 1608641, stock 2 mg/mL, RRID:AB_2535864
Donkey anti-Mouse 647 Jackson ImmunoResearch 715-606-150 Use at 1:1000, Lot: 140554, stock 1.4 mg/mL
Dulbecco's Modified Eagle Medium: Nutrient Mixture F-12 (DMEM/F12) Thermo Fisher Scientific 11330-057
Dulbecco's phosphate-buffered saline (DPBS) Thermo Fisher Scientific 14190-250 Without Mg2+/Ca2+
EGF Fisher Scientific AF-100-15-100ug
Fetal Bovine Serum VWR 97068–085 100% US Origin, premium grade, Lot: 059B18
Fluoromount-G (Southern Biotech) Fisher Scientific 0100-01 Referred to as mounting media in text
Gentamicin Thermo Fisher Scientific 15710064
Glycine Fisher Scientific BP381-5
Goat anti-WAP Santa Cruz Biotech SC-14832 Use at 1:250, Lot: J1011, stock 200 µg/mL, RRID:AB_677601
Hoechst 33342 AnaSpec AS-83218 Use 1:2000, stock is 20mM
Insulin Millipore Sigma I6634-100mg
KCl Fisher Scientific P217-500
KH2PO4 Fisher Scientific P285-500
KRT14–CreERtam The Jackson Laboratory 5107
Matrigel Growth Factor Reduced (GFR); Phenol Red-Free; 10 mL Fisher Scientific CB-40230C Lot: 8204010, stock concentration 8.9 mg/mL
MillexGV Filter Unit 0.22 µm Millipore Sigma SLGV033RS
Millicell EZ SLIDE 8-well glass, sterile Millipore Sigma PEZGS0816 These chamber slides are great for gasket removal but other brands can work well (e.g. Lab Tek II).
Mouse anti-SMA Millipore Sigma A2547 Use at 1:500, Lot: 128M4881V, stock 5.2 mg/mL, RRID:AB_476701
N-2 Supplement (100x) Thermo Fisher Scientific 17502048
NaCl Fisher Scientific S671-3
NaH2PO4 Fisher Scientific S468-500
Nrg1 R&D 5898-NR-050
Ovine Pituitary Prolactin National Hormone and Peptide Program Purchased from Dr. Parlow at Harbor-UCLA Research and Education Institute
Paraformaldahyde Millipore Sigma PX0055-3
Pentobarbital Millipore Sigma P3761
R26R-EYFP The Jackson Laboratory 6148
Rho inhibitor Y-27632 Tocris 1254
R-spondin Peprotech 120-38
Sodium Hydroxide Fisher Scientific S318-500
Sterile Filtered Donkey Serum Equitech-Bio Inc. SD30-0500
Sterile Filtered Donkey Serum Equitech-Bio Inc. SD30-0500
Triton X-100 Millipore Sigma x100-500ML Laboratory grade
Trypsin EDTA 0.05% Thermo Fisher Scientific 25300-062

Referencias

  1. Macias, H., Hinck, L. Mammary gland development. Wiley Interdisciplinary Reviews in Developmental Biology. 1 (4), 533-557 (2012).
  2. Daniel, C. W., De Ome, K. B., Young, J. T., Blair, P. B., Faulkin, L. J. The in vivo life span of normal and preneoplastic mouse mammary glands: a serial transplantation study. Proceedings of the National Academy of Science U S A. 61 (1), 53-60 (1968).
  3. Ip, M. M., Asch, B. B. . Methods in Mammary Gland Biology and Breast Cancer Research. , (2000).
  4. Shackleton, M., et al. Generation of a functional mammary gland from a single stem cell. Nature. 439 (7072), 84-88 (2006).
  5. Stingl, J., et al. Purification and unique properties of mammary epithelial stem cells. Nature. 439 (7079), 993-997 (2006).
  6. Lasfargues, E. Y. Cultivation and behavior in vitro of the normal mammary epithelium of the adult mouse. II. Observations on the secretory activity. Experimental Cell Research. 13 (3), 553-562 (1957).
  7. Simian, M., Bissell, M. J. Organoids: A historical perspective of thinking in three dimensions. Journal of Cell Biology. 216 (1), 31-40 (2017).
  8. Orkin, R. W., et al. A murine tumor producing a matrix of basement membrane. Journal of Experimental Medicine. 145 (1), 204-220 (1977).
  9. Lee, E. Y., Parry, G., Bissell, M. J. Modulation of secreted proteins of mouse mammary epithelial cells by the collagenous substrata. Journal of Cell Biology. 98 (1), 146-155 (1984).
  10. Lee, E. Y., Lee, W. H., Kaetzel, C. S., Parry, G., Bissell, M. J. Interaction of mouse mammary epithelial cells with collagen substrata: regulation of casein gene expression and secretion. Proceedings of the National Academy of Science U S A. 82 (5), 1419-1423 (1985).
  11. Bissell, M. J., Barcellos-Hoff, M. H. The influence of extracellular matrix on gene expression: is structure the message?. Journal of Cell Science. 8 (Suppl), 327-343 (1987).
  12. 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. Proceedings of the National Academy of Science U S A. 89 (19), 9064-9068 (1992).
  13. Jarde, T., et al. Wnt and Neuregulin1/ErbB signalling extends 3D culture of hormone responsive mammary organoids. Nature Commununications. 7, 13207 (2016).
  14. Sachs, N., et al. A Living Biobank of Breast Cancer Organoids Captures Disease Heterogeneity. Cell. 172 (1-2), 373-386 (2018).
  15. Daniel, C. W., Strickland, P., Friedmann, Y. Expression and functional role of E- and P-cadherins in mouse mammary ductal morphogenesis and growth. Biología del desarrollo. 169 (2), 511-519 (1995).
  16. Runswick, S. K., O’Hare, M. J., Jones, L., Streuli, C. H., Garrod, D. R. Desmosomal adhesion regulates epithelial morphogenesis and cell positioning. Nature Cell Biology. 3 (9), 823-830 (2001).
  17. Chanson, L., et al. Self-organization is a dynamic and lineage-intrinsic property of mammary epithelial cells. Proceedings of the National Academy of Science U S A. 108 (8), 3264-3269 (2011).
  18. Honvo-Houeto, E., Truchet, S. Indirect Immunofluorescence on Frozen Sections of Mouse Mammary Gland. Journal of Visualized Experiments. (106), e53179 (2015).
  19. Macias, H., et al. SLIT/ROBO1 signaling suppresses mammary branching morphogenesis by limiting basal cell number. Developmental Cell. 20 (6), 827-840 (2011).
  20. Welm, B. E., Dijkgraaf, G. J., Bledau, A. S., Welm, A. L., Werb, Z. Lentiviral transduction of mammary stem cells for analysis of gene function during development and cancer. Cell Stem Cell. 2 (1), 90-102 (2008).
  21. Smith, P., et al. VANGL2 regulates luminal epithelial organization and cell turnover in the mammary gland. Scientific Reports. 9 (1), 7079 (2019).
  22. Lee, G. Y., Kenny, P. A., Lee, E. H., Bissell, M. J. Three-dimensional culture models of normal and malignant breast epithelial cells. Nature Methods. 4 (4), 359-365 (2007).
  23. Campbell, J. J., Davidenko, N., Caffarel, M. M., Cameron, R. E., Watson, C. J. A multifunctional 3D co-culture system for studies of mammary tissue morphogenesis and stem cell biology. PLoS One. 6 (9), e25661 (2011).
  24. Labarge, M. A., Garbe, J. C., Stampfer, M. R. Processing of human reduction mammoplasty and mastectomy tissues for cell culture. Journal of Visualized Experiments. (71), e50011 (2013).
  25. Marlow, R., Dontu, G. Modeling the breast cancer bone metastatic niche in complex three-dimensional cocultures. Methods in Molecular Biology. 1293, 213-220 (2015).
  26. Koledova, Z., Lu, P. A 3D Fibroblast-Epithelium Co-culture Model for Understanding Microenvironmental Role in Branching Morphogenesis of the Mammary Gland. Methods in Molecular Biology. 1501, 217-231 (2017).
check_url/es/60742?article_type=t

Play Video

Citar este artículo
Rubio, S., Cazares, O., Macias, H., Hinck, L. Generation of Mosaic Mammary Organoids by Differential Trypsinization. J. Vis. Exp. (157), e60742, doi:10.3791/60742 (2020).

View Video