1
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Establishment and long-term culture of mouse mammary stem cell organoids and breast tumor organoids

      research-article
      1 , 2 , , 1 , 2 , 3 , 4 , ∗∗
      STAR Protocols
      Elsevier
      Cancer, Cell culture, Organoids, Stem Cells

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Summary

          Mammary stem cells (MaSCs) contribute to mammary epithelium development and homeostasis. They have been proposed as cells of origin for breast cancer. Here, we describe an organoid culture protocol for ex vivo expansion of MaSCs from mouse tissues. These organoids maintain the self-renewal of gland-reconstituting MaSCs and can be used to model tumorigenesis by introducing patient-relevant cancer drivers and mutations. Similar organoid culture can be used for long-term expansion of luminal stem/progenitor cells from normal glands and tumor-initiating cells from mammary tumors.

          For complete details on the use and execution of this protocol, please refer to Christin et al. (2020) and Zhang et al. (2016).

          Graphical abstract

          Highlights

          • Protocol to isolate epithelial cells from mammary glands and tumors

          • Detailed procedure to generate and expand MaSCs and tumor organoids

          • Organoids can be used for genetic modification and biological analyses

          Abstract

          Mammary stem cells (MaSCs) contribute to mammary epithelium development and homeostasis. They have been proposed as cells of origin for breast cancer. Here, we describe an organoid culture protocol for ex vivo expansion of MaSCs from mouse tissues. These organoids maintain the self-renewal of gland-reconstituting MaSCs and can be used to model tumorigenesis by introducing patient-relevant cancer drivers and mutations. Similar organoid culture can be used for long-term expansion of luminal stem/progenitor cells from normal glands and tumor-initiating cells from mammary tumors.

          Related collections

          Most cited references3

          • Record: found
          • Abstract: found
          • Article: not found

          Slug and Sox9 cooperatively determine the mammary stem cell state.

          Regulatory networks orchestrated by key transcription factors (TFs) have been proposed to play a central role in the determination of stem cell states. However, the master transcriptional regulators of adult stem cells are poorly understood. We have identified two TFs, Slug and Sox9, that act cooperatively to determine the mammary stem cell (MaSC) state. Inhibition of either Slug or Sox9 blocks MaSC activity in primary mammary epithelial cells. Conversely, transient coexpression of exogenous Slug and Sox9 suffices to convert differentiated luminal cells into MaSCs with long-term mammary gland-reconstituting ability. Slug and Sox9 induce MaSCs by activating distinct autoregulatory gene expression programs. We also show that coexpression of Slug and Sox9 promotes the tumorigenic and metastasis-seeding abilities of human breast cancer cells and is associated with poor patient survival, providing direct evidence that human breast cancer stem cells are controlled by key regulators similar to those operating in normal murine MaSCs. Copyright © 2012 Elsevier Inc. All rights reserved.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: found
            Is Open Access

            Stem Cell Determinant SOX9 Promotes Lineage Plasticity and Progression in Basal-like Breast Cancer

            SUMMARY Lineage plasticity is important for the development of basal-like breast cancer (BLBC), an aggressive cancer subtype. While BLBC is likely to originate from luminal progenitor cells, it acquires substantial basal cell features and contains a heterogenous collection of cells exhibiting basal, luminal, and hybrid phenotypes. Why luminal progenitors are prone to BLBC transformation and what drives luminal-to-basal reprogramming remain unclear. Here, we show that the transcription factor SOX9 acts as a determinant for estrogen-receptor-negative (ER−) luminal stem/progenitor cells (LSPCs). SOX9 controls LSPC activity in part by activating both canonical and non-canonical nuclear factor κB (NF-κB) signaling. Inactivation of TP53 and RB via expression of SV40 TAg in a BLBC mouse tumor model leads to upregulation of SOX9, which drives luminal-to-basal reprogramming in vivo. Furthermore, SOX9 deletion inhibits the progression of ductal carcinoma in situ (DCIS)-like lesions to invasive carcinoma. These data show that ER− LSPC determinant SOX9 acts as a lineage plasticity driver for BLBC progression.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Mammary-Stem-Cell-Based Somatic Mouse Models Reveal Breast Cancer Drivers Causing Cell Fate Dysregulation

              Cancer genomics have provided an unprecedented opportunity for understanding genetic causes of human cancer. However, distinguishing which mutations are functionally relevant to cancer pathogenesis remains a major challenge. We describe here a mammary stem cell (MaSC) organoid-based approach for rapid generation of somatic GEMMs (genetically engineered mouse models). By using RNAi and CRISPR-mediated genome engineering in MaSC-GEMMs, we have discovered that inactivation of Ptpn22 or Mll3, two genes mutated in human breast cancer, greatly accelerated PI3K-driven mammary tumorigenesis. Using these tumor models, we have also identified genetic alterations promoting tumor metastasis and causing resistance to PI3K-targeted therapy. Both Ptpn22 and Mll3 inactivation resulted in disruption of mammary gland differentiation and an increase in stem cell activity. Mechanistically, Mll3 deletion enhanced stem cell activity through activation of the HIF pathway. Thus, our study established a robust in vivo platform for functional cancer genomics and discovered functional breast cancer mutations.
                Bookmark

                Author and article information

                Contributors
                Journal
                STAR Protoc
                STAR Protoc
                STAR Protocols
                Elsevier
                2666-1667
                29 May 2021
                18 June 2021
                29 May 2021
                : 2
                : 2
                : 100577
                Affiliations
                [1 ]Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA
                [2 ]Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
                Author notes
                []Corresponding author jihong.cui@ 123456einsteinmed.org
                [∗∗ ]Corresponding author wenjun.guo@ 123456einsteinmed.org
                [3]

                Technical contact

                [4]

                Lead contact

                Article
                S2666-1667(21)00284-7 100577
                10.1016/j.xpro.2021.100577
                8173303
                34124696
                ec451045-c3c9-49e7-b399-ee1451fecc5a
                © 2021 The Author(s)

                This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

                History
                Categories
                Protocol

                cancer,cell culture,organoids,stem cells
                cancer, cell culture, organoids, stem cells

                Comments

                Comment on this article