1
|
Kohler KT, Kim J, Villadsen R, Rønnov-Jessen L, Petersen OW. Oncogene activated human breast luminal progenitors contribute basally located myoepithelial cells. Breast Cancer Res 2024; 26:183. [PMID: 39695857 DOI: 10.1186/s13058-024-01939-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 11/28/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Basal-like breast cancer originates in luminal progenitors, frequently with an altered PI3K pathway, and focally in close association with genetically altered myoepithelial cells at the site of tumor initiation. The exact trajectory behind this bi-lineage phenomenon remains poorly understood. METHODS AND RESULTS Here we used a breast cancer relevant transduction protocol including hTERT, shp16, shp53, and PIK3CAH1047R to immortalize FACS isolated luminal cells, and we identified a candidate multipotent progenitor. Specifically, we identified a keratin 23 (K23)+/ALDH1A3+/CALML5- ductal-like progenitor with the potential to differentiate into CALML5+ lobular-like cells. We found that the apparent luminal phenotype of these oncogene transduced progenitors was metastable giving rise to basal-like cells dependent on culture conditions. In 3D organoid culture and upon transplantation to mice the bipotent progenitor cell line organized into a bi-layered acinus-like structure reminiscent of that of the normal breast gland. CONCLUSIONS These findings provide proof of principle that progenitors within the human breast luminal epithelial compartment may serve as a source of correctly positioned myoepithelial cells. This may prove useful in assessing the role of myoepithelial cells in breast tumor progression.
Collapse
Affiliation(s)
| | - Jiyoung Kim
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - René Villadsen
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Lone Rønnov-Jessen
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Ole William Petersen
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
2
|
Lapenta C, Santini SM, Antonacci C, Donati S, Cecchetti S, Frittelli P, Catalano P, Urbani F, Macchia I, Spada M, Vitale S, Michelini Z, Corsi DC, Zeuner A, Dattilo R, Tamburo De Bella M. Anti-Tumor Immunity to Patient-Derived Breast Cancer Cells by Vaccination with Interferon-Alpha-Conditioned Dendritic Cells (IFN-DC). Vaccines (Basel) 2024; 12:1058. [PMID: 39340087 PMCID: PMC11435915 DOI: 10.3390/vaccines12091058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 09/06/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Breast cancer represents one of the leading causes of death among women. Surgery can be effective, but once breast cancer has metastasized, it becomes extremely difficult to treat. Conventional therapies are associated with substantial toxicity and poor efficacy due to tumor heterogeneity, treatment resistance and disease relapse. Moreover, immune checkpoint blockade appears to offer limited benefit in breast cancer. The poor tumor immunogenicity and the immunosuppressive tumor microenvironment result in scarce T-cell infiltration, leading to a low response rate. Thus, there is considerable interest in the development of improved active immunotherapies capable of sensitizing a patient's immune system against tumor cells. METHODS We evaluated the in vitro anti-tumor activity of a personalized vaccine based on dendritic cells generated in the presence of interferon (IFN)-α and granulocyte-macrophage colony-stimulating factor (IFN-DC) and loaded with an oxidized lysate from autologous tumor cells expanded as 3D organoid culture maintaining faithful tumor antigenic profiles. RESULTS Our findings demonstrate that stimulation of breast cancer patients' lymphocytes with autologous IFN-DC led to efficient Th1-biased response and the generation in vitro of potent cytotoxic activity toward the patients' own tumor cells. CONCLUSIONS This approach can be potentially applied in association with checkpoint blockade and chemotherapy in the design of new combinatorial therapies for breast cancer.
Collapse
Affiliation(s)
- Caterina Lapenta
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (S.D.); (F.U.); (I.M.); (S.V.); (A.Z.); (R.D.)
| | - Stefano Maria Santini
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (S.D.); (F.U.); (I.M.); (S.V.); (A.Z.); (R.D.)
| | - Celeste Antonacci
- Department of Pediatric Hematology/Oncology and Cellular and Gene Therapy, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy;
| | - Simona Donati
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (S.D.); (F.U.); (I.M.); (S.V.); (A.Z.); (R.D.)
| | - Serena Cecchetti
- Core Facilities, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Patrizia Frittelli
- Medical Oncology Unit, Fatebenefratelli-Isola Tiberina Hospital, 00186 Rome, Italy; (P.F.); (P.C.); (D.C.C.)
| | - Piera Catalano
- Medical Oncology Unit, Fatebenefratelli-Isola Tiberina Hospital, 00186 Rome, Italy; (P.F.); (P.C.); (D.C.C.)
| | - Francesca Urbani
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (S.D.); (F.U.); (I.M.); (S.V.); (A.Z.); (R.D.)
| | - Iole Macchia
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (S.D.); (F.U.); (I.M.); (S.V.); (A.Z.); (R.D.)
| | - Massimo Spada
- Center of Animal Research and Welfare, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Sara Vitale
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (S.D.); (F.U.); (I.M.); (S.V.); (A.Z.); (R.D.)
| | - Zuleika Michelini
- National Center for Global Health, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Domenico Cristiano Corsi
- Medical Oncology Unit, Fatebenefratelli-Isola Tiberina Hospital, 00186 Rome, Italy; (P.F.); (P.C.); (D.C.C.)
| | - Ann Zeuner
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (S.D.); (F.U.); (I.M.); (S.V.); (A.Z.); (R.D.)
| | - Rosanna Dattilo
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (S.D.); (F.U.); (I.M.); (S.V.); (A.Z.); (R.D.)
| | - Manuela Tamburo De Bella
- Hospital Clinical Networks Governance e DM70/15 Monitoring—AGENAS—National Agency for Regional Health Services, 00187 Rome, Italy;
| |
Collapse
|
3
|
Shu J, Deng H, Zhang Y, Wu F, He J. Cancer cell response to extrinsic and intrinsic mechanical cue: opportunities for tumor apoptosis strategies. Regen Biomater 2024; 11:rbae016. [PMID: 38476678 PMCID: PMC10932484 DOI: 10.1093/rb/rbae016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 01/31/2024] [Accepted: 02/07/2024] [Indexed: 03/14/2024] Open
Abstract
Increasing studies have revealed the importance of mechanical cues in tumor progression, invasiveness and drug resistance. During malignant transformation, changes manifest in either the mechanical properties of the tissue or the cellular ability to sense and respond to mechanical signals. The major focus of the review is the subtle correlation between mechanical cues and apoptosis in tumor cells from a mechanobiology perspective. To begin, we focus on the intracellular force, examining the mechanical properties of the cell interior, and outlining the role that the cytoskeleton and intracellular organelle-mediated intracellular forces play in tumor cell apoptosis. This article also elucidates the mechanisms by which extracellular forces guide tumor cell mechanosensing, ultimately triggering the activation of the mechanotransduction pathway and impacting tumor cell apoptosis. Finally, a comprehensive examination of the present status of the design and development of anti-cancer materials targeting mechanotransduction is presented, emphasizing the underlying design principles. Furthermore, the article underscores the need to address several unresolved inquiries to enhance our comprehension of cancer therapeutics that target mechanotransduction.
Collapse
Affiliation(s)
- Jun Shu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, PR China
| | - Huan Deng
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, PR China
| | - Yu Zhang
- College of Food and Biological Engineering, Chengdu University, Chengdu 610106, PR China
| | - Fang Wu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, PR China
| | - Jing He
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, PR China
| |
Collapse
|
4
|
Todhunter ME, Miyano M, Carlson EG, Hinz S, LaBarge MA. Sustained postconfluent culture of human mammary epithelial cells enriches for luminal and c-Kit+ subtypes. Breast Cancer Res 2023; 25:6. [PMID: 36653787 PMCID: PMC9847146 DOI: 10.1186/s13058-022-01595-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 12/16/2022] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND A challenge in human mammary epithelial cell (HMEC) culture is sustaining the representation of competing luminal, myoepithelial, and progenitor lineages over time. As cells replicate in culture, myoepithelial cells come to dominate the composition of the culture with serial passaging. This drift in composition presents a challenge for studying luminal and progenitor cells, which are prospective cells of origin for most breast cancer subtypes. METHODS We demonstrate the use of postconfluent culture on HMECs. Postconfluent culture entails culturing HMECs for 2-5 weeks without passaging but maintaining frequent feedings in low-stress M87A culture medium. In contrast, standard HMEC culture entails enzymatic subculturing every 3-5 days to maintain subconfluent density. RESULTS When compared to standard HMEC culture, postconfluent culture yields increased proportions of luminal cells and c-Kit+ progenitor cells. Postconfluent cultures develop a distinct multilayered morphology with individual cells showing decreased physical deformability as compared to cells in standard culture. Gene expression analysis of postconfluent cells shows increased expression of lineage-specific markers and extracellular matrix components. CONCLUSIONS Postconfluent culture is a novel, useful strategy for altering the lineage composition of HMECs, by increasing the proportional representation of luminal and progenitor cells. We speculate that postconfluent culture creates a microenvironment with cellular composition closer to the physiological state and eases the isolation of scarce cell subtypes. As such, postconfluent culture is a valuable tool for researchers using HMECs for breast cancer research.
Collapse
Affiliation(s)
- Michael E. Todhunter
- grid.410425.60000 0004 0421 8357Department of Population Sciences, Beckman Research Institute at City of Hope, 1500 E. Duarte Rd, Duarte, CA 91010 USA
| | - Masaru Miyano
- grid.410425.60000 0004 0421 8357Department of Population Sciences, Beckman Research Institute at City of Hope, 1500 E. Duarte Rd, Duarte, CA 91010 USA
| | - Eric G. Carlson
- grid.410425.60000 0004 0421 8357Department of Population Sciences, Beckman Research Institute at City of Hope, 1500 E. Duarte Rd, Duarte, CA 91010 USA ,grid.410425.60000 0004 0421 8357Irell and Manella Graduate School of Biological Sciences, City of Hope, 1500 E. Duarte Rd, Duarte, CA 91010 USA
| | - Stefan Hinz
- grid.410425.60000 0004 0421 8357Department of Population Sciences, Beckman Research Institute at City of Hope, 1500 E. Duarte Rd, Duarte, CA 91010 USA
| | - Mark A. LaBarge
- grid.410425.60000 0004 0421 8357Department of Population Sciences, Beckman Research Institute at City of Hope, 1500 E. Duarte Rd, Duarte, CA 91010 USA
| |
Collapse
|
5
|
Thiemann RF, Varney S, Moskwa N, Lamar J, Larsen M, LaFlamme SE. Regulation of myoepithelial differentiation. PLoS One 2022; 17:e0268668. [PMID: 35617216 PMCID: PMC9135247 DOI: 10.1371/journal.pone.0268668] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 05/04/2022] [Indexed: 12/30/2022] Open
Abstract
The salivary gland can be permanently impaired by radiation treatment for head and neck cancers. Efforts at tissue regeneration have focused on saliva-producing acinar cells. However, myoepithelial cells are also critical to gland function, but mechanisms that regulate their differentiation are poorly defined. To study myoepithelial differentiation, we employed mSG-PAC1 murine salivary gland epithelial cells. We demonstrate that mSG-PAC1 spheroids exhibit phenotypic plasticity between pro-acinar and myoepithelial cell fates. Increased expression of pro-acinar/acinar or myoepithelial RNAs was identified from spheroids cultured under different media conditions by microarray followed by gene-set enrichment analysis. Spheroids cultured with different medium components expressed proteins typical of either acinar or myoepithelial cells, as detected by immunocytochemistry. We demonstrate that the pattern of TAZ expression in the epithelial compartment of the differentiating murine salivary gland correlates with the expression of the myoepithelial marker alpha-SMA, as is the case for TAZ expression in mSG-PAC1 spheroids. Our analysis also indicates that YAP/TAZ target genes are upregulated together with myoepithelial markers. Importantly, siRNA targeting of TAZ expression in mSG-PAC1 spheroids diminished the expression of myoepithelial markers. Our results in this in vitro cell model implicate TAZ signaling in myoepithelial differentiation.
Collapse
Affiliation(s)
- Renee F. Thiemann
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, New York, United States of America
| | - Scott Varney
- Department of Surgery, Albany Medical College, Albany, New York, United States of America
| | - Nicholas Moskwa
- Department of Biological Sciences, University at Albany, State University of New York, Albany, New York, United States of America
| | - John Lamar
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York, United States of America
| | - Melinda Larsen
- Department of Biological Sciences, University at Albany, State University of New York, Albany, New York, United States of America
| | - Susan E. LaFlamme
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, New York, United States of America
- * E-mail:
| |
Collapse
|
6
|
Sigurdardottir AK, Jonasdottir AS, Asbjarnarson A, Helgudottir HR, Gudjonsson T, Traustadottir GA. Peroxidasin Enhances Basal Phenotype and Inhibits Branching Morphogenesis in Breast Epithelial Progenitor Cell Line D492. J Mammary Gland Biol Neoplasia 2021; 26:321-338. [PMID: 34964086 PMCID: PMC8858314 DOI: 10.1007/s10911-021-09507-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 12/13/2021] [Indexed: 11/17/2022] Open
Abstract
The human breast is composed of terminal duct lobular units (TDLUs) that are surrounded by stroma. In the TDLUs, basement membrane separates the stroma from the epithelial compartment, which is divided into an inner layer of luminal epithelial cells and an outer layer of myoepithelial cells. Stem cells and progenitor cells also reside within the epithelium and drive a continuous cycle of gland remodelling that occurs throughout the reproductive period. D492 is an epithelial cell line originally isolated from the stem cell population of the breast and generates both luminal and myoepithelial cells in culture. When D492 cells are embedded into 3D reconstituted basement membrane matrix (3D-rBM) they form branching colonies mimicking the TDLUs of the breast, thereby providing a well-suited in vitro model for studies on branching morphogenesis and breast development. Peroxidasin (PXDN) is a heme-containing peroxidase that crosslinks collagen IV with the formation of sulfilimine bonds. Previous studies indicate that PXDN plays an integral role in basement membrane stabilisation by crosslinking collagen IV and as such contributes to epithelial integrity. Although PXDN has been linked to fibrosis and cancer in some organs there is limited information on its role in development, including in the breast. In this study, we demonstrate expression of PXDN in breast epithelium and stroma and apply the D492 cell line to investigate the role of PXDN in cell differentiation and branching morphogenesis in the human breast. Overexpression of PXDN induced basal phenotype in D492 cells, loss of plasticity and inhibition of epithelial-to-mesenchymal transition as is displayed by complete inhibition of branching morphogenesis in 3D culture. This is supported by results from RNA-sequencing which show significant enrichment in genes involved in epithelial differentiation along with significant negative enrichment of EMT factors. Taken together, we provide evidence for a novel role of PXDN in breast epithelial differentiation and mammary gland development.
Collapse
Affiliation(s)
- Anna Karen Sigurdardottir
- Stem Cell Research Unit, Biomedical Center, Department of Anatomy, Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Arna Steinunn Jonasdottir
- Stem Cell Research Unit, Biomedical Center, Department of Anatomy, Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Arni Asbjarnarson
- Stem Cell Research Unit, Biomedical Center, Department of Anatomy, Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Hildur Run Helgudottir
- Stem Cell Research Unit, Biomedical Center, Department of Anatomy, Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Thorarinn Gudjonsson
- Stem Cell Research Unit, Biomedical Center, Department of Anatomy, Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
- Department of Laboratory Haematology, Landspitali - University Hospital, Reykjavik, Iceland
| | - Gunnhildur Asta Traustadottir
- Stem Cell Research Unit, Biomedical Center, Department of Anatomy, Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland.
| |
Collapse
|
7
|
Aquaporins implicated in the cell proliferation and the signaling pathways of cell stemness. Biochimie 2021; 188:52-60. [PMID: 33894294 DOI: 10.1016/j.biochi.2021.04.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 04/11/2021] [Accepted: 04/16/2021] [Indexed: 12/20/2022]
Abstract
Aquaporins (AQPs) are water channel proteins facilitating passive transport of water and other small molecules across biomembranes. Regulation of osmotic homeostasis via AQPs is accompanied by dynamic participation of various cellular signaling pathways. Recently emerging evidence reveals that functional roles of AQPs are further extended from the osmotic regulation via water permeation into the cell proliferation and differentiation. In particular, anomalous expression of AQPs has been demonstrated in various types of cancer cells and cancer stem-like cells and it has been proposed as markers for proliferation and progression of cancer cells. Thus, a more comprehensive view on AQPs could bring a great interest in the cell stemness accompanied by the expression of AQPs. AQPs are broadly expressed across tissues and cells in a cell type- and lineage-specific manner during development via spatiotemporal transcriptional regulation. Moreover, AQPs are expressed in various adult stem cells and cells associated with a stem cell niche as well as cancer stem-like cells. However, the expression and regulatory mechanisms of AQP expression in stem cells have not been well understood. This review highlighted the AQPs expression in stem cell niches/stem cells and the involvement of AQPs in the cell proliferation and signaling pathways associated with cell stemness.
Collapse
|
8
|
Chowdhury S, Ghosh S. Sources, Isolation and culture of stem cells? Stem Cells 2021. [DOI: 10.1007/978-981-16-1638-9_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
9
|
Budkova Z, Sigurdardottir AK, Briem E, Bergthorsson JT, Sigurdsson S, Magnusson MK, Traustadottir GA, Gudjonsson T, Hilmarsdottir B. Expression of ncRNAs on the DLK1-DIO3 Locus Is Associated With Basal and Mesenchymal Phenotype in Breast Epithelial Progenitor Cells. Front Cell Dev Biol 2020; 8:461. [PMID: 32612992 PMCID: PMC7308478 DOI: 10.3389/fcell.2020.00461] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 05/18/2020] [Indexed: 12/18/2022] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) and its reversed process mesenchymal-to-epithelial transition (MET) play a critical role in epithelial plasticity during development and cancer progression. Among important regulators of these cellular processes are non-coding RNAs (ncRNAs). The imprinted DLK1-DIO3 locus, containing numerous maternally expressed ncRNAs including the lncRNA maternally expressed gene 3 (MEG3) and a cluster of over 50 miRNAs, has been shown to be a modulator of stemness in embryonic stem cells and in cancer progression, potentially through the tumor suppressor role of MEG3. In this study we analyzed the expression pattern and functional role of ncRNAs from the DLK1-DIO3 locus in epithelial plasticity of the breast. We studied their expression in various cell types of breast tissue and revisit the role of the locus in EMT/MET using a breast epithelial progenitor cell line (D492) and its isogenic mesenchymal derivative (D492M). Marked upregulation of ncRNAs from the DLK1-DIO3 locus was seen after EMT induction in two cell line models of EMT. In addition, the expression of MEG3 and the maternally expressed ncRNAs was higher in stromal cells compared to epithelial cell types in primary breast tissue. We also show that expression of MEG3 is concomitant with the expression of the ncRNAs from the DLK1-DIO3 locus and its expression is therefore likely indicative of activation of all ncRNAs at the locus. MEG3 expression is correlated with stromal markers in normal tissue and breast cancer tissue and negatively correlated with the survival of breast cancer patients in two different cohorts. Overexpression of MEG3 using CRISPR activation in a breast epithelial cell line induced partial EMT and enriched for a basal-like phenotype. Conversely, knock down of MEG3 using CRISPR inhibition in a mesenchymal cell line reduced the mesenchymal and basal-like phenotype of the cell line. In summary our study shows that maternally expressed ncRNAs are markers of EMT and suggests that MEG3 is a novel regulator of EMT/MET in breast tissue. Nevertheless, further studies are needed to fully dissect the molecular pathways influenced by non-coding RNAs at the DLK1-DIO3 locus in breast tissue.
Collapse
Affiliation(s)
- Zuzana Budkova
- Stem Cell Research Unit, Biomedical Center, Department of Anatomy, Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Anna Karen Sigurdardottir
- Stem Cell Research Unit, Biomedical Center, Department of Anatomy, Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Eirikur Briem
- Stem Cell Research Unit, Biomedical Center, Department of Anatomy, Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Jon Thor Bergthorsson
- Department of Laboratory Hematology, Landspitali - University Hospital, Reykjavik, Iceland
| | - Snævar Sigurdsson
- Stem Cell Research Unit, Biomedical Center, Department of Anatomy, Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Magnus Karl Magnusson
- Department of Pharmacology and Toxicology, Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Gunnhildur Asta Traustadottir
- Stem Cell Research Unit, Biomedical Center, Department of Anatomy, Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Thorarinn Gudjonsson
- Stem Cell Research Unit, Biomedical Center, Department of Anatomy, Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland.,Department of Laboratory Hematology, Landspitali - University Hospital, Reykjavik, Iceland
| | - Bylgja Hilmarsdottir
- Stem Cell Research Unit, Biomedical Center, Department of Anatomy, Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland.,Department of Pathology, Landspitali - University Hospital, Reykjavik, Iceland
| |
Collapse
|
10
|
Samocha A, Doh H, Kessenbrock K, Roose JP. Unraveling Heterogeneity in Epithelial Cell Fates of the Mammary Gland and Breast Cancer. Cancers (Basel) 2019; 11:E1423. [PMID: 31554261 PMCID: PMC6826786 DOI: 10.3390/cancers11101423] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 09/22/2019] [Accepted: 09/22/2019] [Indexed: 12/14/2022] Open
Abstract
Fluidity in cell fate or heterogeneity in cell identity is an interesting cell biological phenomenon, which at the same time poses a significant obstacle for cancer therapy. The mammary gland seems a relatively straightforward organ with stromal cells and basal- and luminal- epithelial cell types. In reality, the epithelial cell fates are much more complex and heterogeneous, which is the topic of this review. Part of the complexity comes from the dynamic nature of this organ: the primitive epithelial tree undergoes extensively remodeling and expansion during puberty, pregnancy, and lactation and, unlike most other organs, the bulk of mammary gland development occurs late, during puberty. An active cell biological debate has focused on lineage commitment to basal- and luminal- epithelial cell fates by epithelial progenitor and stem cells; processes that are also relevant to cancer biology. In this review, we discuss the current understanding of heterogeneity in mammary gland and recent insights obtained through lineage tracing, signaling assays, and organoid cultures. Lastly, we relate these insights to cancer and ongoing efforts to resolve heterogeneity in breast cancer with single-cell RNAseq approaches.
Collapse
Affiliation(s)
- Alexandr Samocha
- Department of Anatomy, University of California, San Francisco, CA 94143, USA.
| | - Hanna Doh
- Department of Anatomy, University of California, San Francisco, CA 94143, USA.
| | - Kai Kessenbrock
- Department of Biological Chemistry, University of California, Irvine, CA 92697, USA.
| | - Jeroen P Roose
- Department of Anatomy, University of California, San Francisco, CA 94143, USA.
| |
Collapse
|
11
|
Sen M, Hahn F, Black TA, DeMarshall M, Porter W, Snowden E, Yee SS, Tong F, Ferguson M, Fleshman EN, Nakagawa H, Falk GW, Ginsberg GG, Kochman ML, Blaesius R, Rustgi AK, Carpenter EL. Flow based single cell analysis of the immune landscape distinguishes Barrett's esophagus from adjacent normal tissue. Oncotarget 2019; 10:3592-3604. [PMID: 31217895 PMCID: PMC6557213 DOI: 10.18632/oncotarget.26911] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 04/14/2019] [Indexed: 01/21/2023] Open
Abstract
Barrett’s esophagus (BE) is metaplasia of the squamous epithelium to a specialized columnar epithelium. BE progresses through low- and high-grade dysplasia before developing into esophageal adenocarcinoma. The BE microenvironment is not well defined. We compare 12 human clinical BE and adjacent normal squamous epithelium biopsies using single cell immunophenotyping by flow cytometry. A cassette of 19 epithelial and immune cell markers was used to detect differences between cellular compartments in normal and BE tissues. We found that the BE microenvironment has an immunological landscape distinct from adjacent normal epithelium. BE has an increased percentage of epithelial cells with a concomitant decrease in the percentage of immune cells, accompanied by a shift in the immune landscape from a predominantly T cell rich microenvironment in normal tissue to a B cell rich landscape in BE tissue. Hierarchical clustering separates BE and normal samples into two discrete groups based upon our 19-marker panel, but also reveals unexpected, shared phenotypes for three patients. Our results suggest that flow based single cell analysis may have the potential for revealing clinically relevant differences between BE and normal adjacent tissue, and that surface immunophenotypes could identify specific subpopulations from dysplastic tissue for further investigation.
Collapse
Affiliation(s)
- Moen Sen
- Division of Hematology and Oncology, Department of Medicine, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Friedrich Hahn
- Department of Genomic Sciences, BD Technologies and Innovation, Research Triangle Park, Durham, North Carolina, USA
| | - Taylor A Black
- Division of Hematology and Oncology, Department of Medicine, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Maureen DeMarshall
- Division of Gastroenterology, Department of Medicine, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Warren Porter
- Department of Genomic Sciences, BD Technologies and Innovation, Research Triangle Park, Durham, North Carolina, USA
| | - Eileen Snowden
- Department of Genomic Sciences, BD Technologies and Innovation, Research Triangle Park, Durham, North Carolina, USA
| | - Stephanie S Yee
- Division of Hematology and Oncology, Department of Medicine, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Frances Tong
- Department of Genomic Sciences, BD Technologies and Innovation, Research Triangle Park, Durham, North Carolina, USA
| | - Mitchell Ferguson
- Department of Genomic Sciences, BD Technologies and Innovation, Research Triangle Park, Durham, North Carolina, USA
| | - Emylee N Fleshman
- Division of Hematology and Oncology, Department of Medicine, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Hiroshi Nakagawa
- Division of Gastroenterology, Department of Medicine, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Gary W Falk
- Division of Gastroenterology, Department of Medicine, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Gregory G Ginsberg
- Division of Gastroenterology, Department of Medicine, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Michael L Kochman
- Division of Gastroenterology, Department of Medicine, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Rainer Blaesius
- Department of Genomic Sciences, BD Technologies and Innovation, Research Triangle Park, Durham, North Carolina, USA
| | - Anil K Rustgi
- Division of Gastroenterology, Department of Medicine, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Erica L Carpenter
- Division of Hematology and Oncology, Department of Medicine, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
12
|
Briem E, Ingthorsson S, Traustadottir GA, Hilmarsdottir B, Gudjonsson T. Application of the D492 Cell Lines to Explore Breast Morphogenesis, EMT and Cancer Progression in 3D Culture. J Mammary Gland Biol Neoplasia 2019; 24:139-147. [PMID: 30684066 DOI: 10.1007/s10911-018-09424-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 12/21/2018] [Indexed: 12/19/2022] Open
Abstract
The human female breast gland is composed of branching epithelial ducts that extend from the nipple towards the terminal duct lobular units (TDLUs), which are the functional, milk-producing units of the gland and the site of origin of most breast cancers. The epithelium of ducts and TDLUs is composed of an inner layer of polarized luminal epithelial cells and an outer layer of contractile myoepithelial cells, separated from the vascular-rich stroma by a basement membrane. The luminal- and myoepithelial cells share an origin and in recent years, there has been increasing understanding of how these cell types interact and how they contribute to breast cancer. Accumulating evidence links stem/or progenitor cells in the mammary/breast gland to breast cancer. In that regard, much knowledge has been gained from studies in mice due to specific strains that have allowed for gene knock out/in studies and lineage tracing of cellular fates. However, there is a large histologic difference between the human female breast gland and the mouse mammary gland that necessitates that research needs to be done on human material where primary cultures are important due to their close relation to the tissue of origin. However, due to difficulties of long-term cultures and lack of access to material, human cell lines are of great importance to bridge the gap between studies on mouse mammary gland and human primary breast cells. In this review, we describe D492, a breast epithelial progenitor cell line that can generate both luminal- and myoepithelial cells in culture, and in 3D culture it forms branching ducts similar to TDLUs. We have applied D492 and its daughter cell lines to explore cellular and molecular mechanisms of branching morphogenesis and cellular plasticity including EMT and MET. In addition to discussing the application of D492 in studying normal morphogenesis, we will also discuss how this cell line has been used to study breast cancer progression.
Collapse
Affiliation(s)
- Eirikur Briem
- Stem Cell Research Unit, Biomedical Center, Department of Anatomy, Faculty of Medicine, School of Health Sciences, University of Iceland, Vatnsmyrarvegi 16, 101, Reykjavík, Iceland
| | - Saevar Ingthorsson
- Stem Cell Research Unit, Biomedical Center, Department of Anatomy, Faculty of Medicine, School of Health Sciences, University of Iceland, Vatnsmyrarvegi 16, 101, Reykjavík, Iceland
| | - Gunnhildur Asta Traustadottir
- Stem Cell Research Unit, Biomedical Center, Department of Anatomy, Faculty of Medicine, School of Health Sciences, University of Iceland, Vatnsmyrarvegi 16, 101, Reykjavík, Iceland
| | - Bylgja Hilmarsdottir
- Department of Tumor Biology, The Norwegian Radium Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Thorarinn Gudjonsson
- Stem Cell Research Unit, Biomedical Center, Department of Anatomy, Faculty of Medicine, School of Health Sciences, University of Iceland, Vatnsmyrarvegi 16, 101, Reykjavík, Iceland.
- Department of Laboratory Hematology, Landspitali - University Hospital, Reykjavík, Iceland.
| |
Collapse
|
13
|
Sugathan S, Lee SJ, Shiwani S, Singh NK. Transdifferentiation of bovine epithelial towards adipocytes in the presence of myoepithelium. ASIAN-AUSTRALASIAN JOURNAL OF ANIMAL SCIENCES 2019; 33:349-359. [PMID: 31010974 PMCID: PMC6946969 DOI: 10.5713/ajas.18.0806] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 10/25/2018] [Indexed: 01/22/2023]
Abstract
Objective Orchastric changes in the mammary glands are vital, especially during lactation. The secretary epithelial cells together with the supporting myoepithelial and stromal cells function cordially to secrete milk. Increase in the number of luminal epithelial cells and a decrease in adipocytes are visible during lactation, whereas the reverse happens in the involution. However, an early involution occurs if the epithelial cells transdifferentiate towards adipocytes during the lactation period. We aimed to inhibit the adipocyte transdifferentiation of luminal cells by restraining the peroxisomal proliferator-activated receptor γ (PPARγ) pathway. Methods Linolenic acid (LA) and thiazolidinediones (TZDs) induced adipogenesis in mammary epithelial cells were conducted in monolayer, mixed culture as well as in transwell plate co-culture with mammary myoepithelial cells. Results Co-culture with myoepithelial cells showed higher adipogenic gene expression in epithelial cells under LA+TZDs treatment. Increase in the expressions of PPARγ, CCAAT/enhancer-binding protein α and vimentin in both mRNA as well as protein levels were observed. Whereas, bisphenol A diglycidyl ether treatment blocked LA+TZDs induced adipogenesis, as it could not show a significant rise in adipose related markers. Although comparative results were found in both mixed culture and monolayer conditions, co-culture technic was found to work better than the others. Conclusion Antagonizing PPARγ pathway in the presence of myoepithelial cells can significantly reduce the adipogenisis in epithelial cells, suggesting therapeutic inhibition of PPARγ can be considered to counter early involution or excessive adipogenesis in mammary epithelium in animals.
Collapse
Affiliation(s)
- Subi Sugathan
- Department of Animal Biotechnology, College of Animal Life Sciences, Kangwon National University, Chuncheon 24341, Korea
| | - Sung-Jin Lee
- Department of Animal Biotechnology, College of Animal Life Sciences, Kangwon National University, Chuncheon 24341, Korea
| | - Supriya Shiwani
- Department of Animal Biotechnology, College of Animal Life Sciences, Kangwon National University, Chuncheon 24341, Korea
| | - Naresh Kumar Singh
- Department of Veterinary Surgery and Radiology, Faculty of Veterinary and Animal Sciences, Institute of Agricultural Sciences, Banaras Hindu University,Varanasi-221005, Uttar Pradesh, India
| |
Collapse
|
14
|
Briem E, Budkova Z, Sigurdardottir AK, Hilmarsdottir B, Kricker J, Timp W, Magnusson MK, Traustadottir GA, Gudjonsson T. MiR-203a is differentially expressed during branching morphogenesis and EMT in breast progenitor cells and is a repressor of peroxidasin. Mech Dev 2019; 155:34-47. [PMID: 30508578 DOI: 10.1016/j.mod.2018.11.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 11/01/2018] [Accepted: 11/23/2018] [Indexed: 12/12/2022]
Abstract
MicroRNAs regulate developmental events such as branching morphogenesis, epithelial to mesenchymal transition (EMT) and its reverse process mesenchymal to epithelial transition (MET). In this study, we performed small RNA sequencing of a breast epithelial progenitor cell line (D492), and its mesenchymal derivative (D492M) cultured in three-dimensional microenvironment. Among the most downregulated miRNAs in D492M was miR-203a, a miRNA that plays an important role in epithelial differentiation. Increased expression of miR-203a was seen in D492, concomitant with increased complexity of branching. When miR-203a was overexpressed in D492M, a partial reversion towards epithelial phenotype was seen. Gene expression analysis of D492M and D492MmiR-203a revealed peroxidasin, a collagen IV cross-linker, as the most significantly downregulated gene in D492MmiR-203a. Collectively, we demonstrate that miR-203a expression temporally correlates with branching morphogenesis and is suppressed in D492M. Overexpression of miR-203a in D492M induces a partial MET and reduces the expression of peroxidasin. Furthermore, we demonstrate that miR-203a is a novel repressor of peroxidasin. MiR-203-peroxidasin axis may be an important regulator in branching morphogenesis, EMT/MET and basement membrane remodeling.
Collapse
Affiliation(s)
- Eirikur Briem
- Stem Cell Research Unit, Biomedical Center, Department of Anatomy, Faculty of Medicine, School of Health Sciences, University of Iceland, Iceland
| | - Zuzana Budkova
- Stem Cell Research Unit, Biomedical Center, Department of Anatomy, Faculty of Medicine, School of Health Sciences, University of Iceland, Iceland
| | - Anna Karen Sigurdardottir
- Stem Cell Research Unit, Biomedical Center, Department of Anatomy, Faculty of Medicine, School of Health Sciences, University of Iceland, Iceland
| | - Bylgja Hilmarsdottir
- Stem Cell Research Unit, Biomedical Center, Department of Anatomy, Faculty of Medicine, School of Health Sciences, University of Iceland, Iceland; Department of Tumor Biology, The Norwegian Radium Hospital, Oslo, Norway
| | - Jennifer Kricker
- Stem Cell Research Unit, Biomedical Center, Department of Anatomy, Faculty of Medicine, School of Health Sciences, University of Iceland, Iceland
| | - Winston Timp
- Department of Biomedical Engineering, Johns Hopkins University, USA
| | - Magnus Karl Magnusson
- Department of Laboratory Hematology, Landspitali - University Hospital, Iceland; Department of Pharmacology and Toxicology, Faculty of Medicine, School of Health Sciences, University of Iceland, Iceland
| | - Gunnhildur Asta Traustadottir
- Stem Cell Research Unit, Biomedical Center, Department of Anatomy, Faculty of Medicine, School of Health Sciences, University of Iceland, Iceland
| | - Thorarinn Gudjonsson
- Stem Cell Research Unit, Biomedical Center, Department of Anatomy, Faculty of Medicine, School of Health Sciences, University of Iceland, Iceland; Department of Laboratory Hematology, Landspitali - University Hospital, Iceland.
| |
Collapse
|
15
|
Age-related gene expression in luminal epithelial cells is driven by a microenvironment made from myoepithelial cells. Aging (Albany NY) 2018; 9:2026-2051. [PMID: 29016359 PMCID: PMC5680554 DOI: 10.18632/aging.101298] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 09/28/2017] [Indexed: 12/24/2022]
Abstract
Luminal epithelial cells in the breast gradually alter gene and protein expression with age, appearing to lose lineage-specificity by acquiring myoepithelial-like characteristics. We hypothesize that the luminal lineage is particularly sensitive to microenvironment changes, and age-related microenvironment changes cause altered luminal cell phenotypes. To evaluate the effects of different microenvironments on the fidelity of epigenetically regulated luminal and myoepithelial gene expression, we generated a set of lineage-specific probes for genes that are controlled through DNA methylation. Culturing primary luminal cells under conditions that favor myoepithelial propogation led to their reprogramming at the level of gene methylation, and to a more myoepithelial-like expression profile. Primary luminal cells' lineage-specific gene expression could be maintained when they were cultured as bilayers with primary myoepithelial cells. Isogenic stromal fibroblast co-cultures were unable to maintain the luminal phenotype. Mixed-age luminal-myoepithelial bilayers revealed that luminal cells adopt transcription and methylation patterns consistent with the chronological age of the myoepithelial cells. We provide evidence that the luminal epithelial phenotype is exquisitely sensitive to microenvironment conditions, and that states of aging are cell non-autonomously communicated through microenvironment cues over at least one cell diameter.
Collapse
|
16
|
Yan M, Wang Y, Wong CW, Or PMY, Wong KL, Li L, Many AM, Guan H, Khoo US, Chan AM. PTEN PDZ-binding domain suppresses mammary carcinogenesis in the MMTV-PyMT breast cancer model. Cancer Lett 2018; 430:67-78. [PMID: 29772266 DOI: 10.1016/j.canlet.2018.05.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 05/07/2018] [Accepted: 05/10/2018] [Indexed: 11/19/2022]
Abstract
Phosphatase and tension homolog (PTEN) is a potent tumor suppressor that possesses a PDZ-binding domain (PDZ-BD) at the end of its carboxyl terminus, whose functions during tumorigenesis remains unclear. Here, we crossed a mouse strain with germline deletion of PTEN PDZ-BD with MMTV-PyMT breast cancer model, and found that knockout (KO) mice display normal development of mammary glands, but have both increased breast tumorigenicity and lung metastasis. Orthotopic allograft experiments suggest the loss of PTEN PDZ-BD in breast cancer cells rather than in tumor microenvironment plays a prominent role in increasing tumor burden. Through RNA-sequencing, we observed a significant downregulation of myoepithelial marker genes in both KO primary breast cancer and orthotopic allografts. Moreover, these myoepithelial marker genes are significantly downregulated in human breast cancer tissues, and are associated with poorer clinical prognosis. In addition, several homeobox genes were also identified to be downreguated in KO breast cancer, whose expressions showed significant positive correlation with myoepithelial marker genes. Overall, our findings suggest a novel tumor suppressive role of PTEN PDZ-BD in a murine model of breast cancer, and the mechanism involves the dysregulation of homeobox genes which may result in defective myoepithelial differentiation in breast cancer cells.
Collapse
Affiliation(s)
- Mingfei Yan
- School of Biomedical Sciences, Room 705, Lo Kwee-Seong Integrated Biomedical Sciences Building, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yubing Wang
- School of Biomedical Sciences, Room 705, Lo Kwee-Seong Integrated Biomedical Sciences Building, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Chi Wai Wong
- School of Biomedical Sciences, Room 705, Lo Kwee-Seong Integrated Biomedical Sciences Building, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Penelope Mei-Yu Or
- School of Biomedical Sciences, Room 705, Lo Kwee-Seong Integrated Biomedical Sciences Building, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Kin Lok Wong
- School of Biomedical Sciences, Room 705, Lo Kwee-Seong Integrated Biomedical Sciences Building, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Lisha Li
- School of Biomedical Sciences, Room 705, Lo Kwee-Seong Integrated Biomedical Sciences Building, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Alexander M Many
- Department of Oncological Sciences, The Mount Sinai School of Medicine, New York NY10029, USA
| | - Hong Guan
- Department of Paediatrics, Medical School of Wisconsin, Milwaukee, WI WI53226, USA
| | - Ui Soon Khoo
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Andrew M Chan
- School of Biomedical Sciences, Room 705, Lo Kwee-Seong Integrated Biomedical Sciences Building, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
17
|
Kuo WL, Ueng SH, Wu CH, Lee LY, Lee YS, Yu MC, Chen SC, Yu CC, Tsai CN. Establishment of two basal-like breast cancer cell lines with extremely low tumorigenicity from Taiwanese premenopausal women. Hum Cell 2018; 31:154-166. [PMID: 29484537 PMCID: PMC5852199 DOI: 10.1007/s13577-017-0197-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Accepted: 12/24/2017] [Indexed: 11/19/2022]
Abstract
The research of carcinogenetic mechanisms of breast cancer in different ethnic backgrounds is an interesting field, as clinical features of breast cancers vary among races. High premenopausal incidence is distinctive in East-Asian breast cancer. However, human cell lines derived from Asian primary breast tumor are rare. To provide alternative cell line models with a relevant genetic background, we aimed to establish breast cancer cell lines from Taiwanese patients of Han-Chinese ethnicity. Fresh tissue from mammary tumors were digested into organoids, plated and grown in basal serum-free medium of human mammary epithelial cells (HuMEC) with supplements. Cells were further enriched by positive selection with CD326 (epithelial cell adhesion molecule; EpCAM)-coated micro-magnetic beads. Two breast cancer cell lines derived from premenopausal women were successfully established by this method, and named Chang-Gung Breast Cancer 01 (CGBC 01) and 02 (CGBC 02). These two cell lines had a similar phenotype with weak expression of estrogen receptor (ER), progesterone receptor (PR), and without amplification of receptor tyrosine protein kinase erbB-2 (HER2/neu). Genome-wide Single Nucleotide Polymorphism (SNP) array showed multiple copy number alterations in both cell lines. Based on gene expression profiles, CGBC 01 and 02 were clustered into basal-like subtype with reference to the breast cancer cell line gene expression database. The tumorigenicity of both cell lines was extremely low in both anchorage-independence assay and transplantation into the mammary fat pads of nude mice. CGBC 01 and CGBC 02 are low tumorigenic breast cancer cell lines, established from Han-Chinese premenopausal breast cancer patients, which serve as in vitro models in studying the biological features of Asian breast cancer.
Collapse
Affiliation(s)
- Wen-Ling Kuo
- Division of Breast Surgery and General Surgery, Department of Surgery, Chang Gung Memorial Hospital, Linkou, Guishan Dist., Taoyuan, 33305, Taiwan.,Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, 33302, Taiwan
| | - Shir-Hwa Ueng
- Department of Pathology, Chang Gung Memorial Hospital, Chang Gung University, Guishan Dist., Taoyuan, 33305, Taiwan
| | - Chun-Hsing Wu
- Division of Breast Surgery and General Surgery, Department of Surgery, Chang Gung Memorial Hospital, Linkou, Guishan Dist., Taoyuan, 33305, Taiwan
| | - Li-Yu Lee
- Department of Pathology, Chang Gung Memorial Hospital, Chang Gung University, Guishan Dist., Taoyuan, 33305, Taiwan
| | - Yun-Shien Lee
- Department of Biotechnology, Ming Chuan University, Guishan Dist., Taoyuan, 33348, Taiwan.,Genomic Medicine Research Core Laboratory, Chang Gung Memorial Hospital, Linkou, Guishan Dist., Taoyuan, 33305, Taiwan
| | - Ming-Chin Yu
- Division of Breast Surgery and General Surgery, Department of Surgery, Chang Gung Memorial Hospital, Linkou, Guishan Dist., Taoyuan, 33305, Taiwan
| | - Shin-Cheh Chen
- Division of Breast Surgery and General Surgery, Department of Surgery, Chang Gung Memorial Hospital, Linkou, Guishan Dist., Taoyuan, 33305, Taiwan
| | - Chi-Chang Yu
- Division of Breast Surgery and General Surgery, Department of Surgery, Chang Gung Memorial Hospital, Linkou, Guishan Dist., Taoyuan, 33305, Taiwan
| | - Chi-Neu Tsai
- Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, 33302, Taiwan. .,Department of Pediatric, Chang-Gung Memorial Hospital, LinKou, Guishan Dist., Taoyuan, 33305, Taiwan.
| |
Collapse
|
18
|
Cereser B, Jansen M, Austin E, Elia G, McFarlane T, van Deurzen CHM, Sieuwerts AM, Daidone MG, Tadrous PJ, Wright NA, Jones L, McDonald SAC. Analysis of clonal expansions through the normal and premalignant human breast epithelium reveals the presence of luminal stem cells. J Pathol 2018; 244:61-70. [PMID: 28940516 PMCID: PMC5765426 DOI: 10.1002/path.4989] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 09/06/2017] [Accepted: 09/15/2017] [Indexed: 12/12/2022]
Abstract
It is widely accepted that the cell of origin of breast cancer is the adult mammary epithelial stem cell; however, demonstrating the presence and location of tissue stem cells in the human breast has proved difficult. Furthermore, we do not know the clonal architecture of the normal and premalignant mammary epithelium or its cellular hierarchy. Here, we use deficiency in the mitochondrial enzyme cytochrome c oxidase (CCO), typically caused by somatic mutations in the mitochondrial genome, as a means to perform lineage tracing in the human mammary epithelium. PCR sequencing of laser-capture microdissected cells in combination with immunohistochemistry for markers of lineage differentiation was performed to determine the clonal nature of the mammary epithelium. We have shown that in the normal human breast, clonal expansions (defined here by areas of CCO deficiency) are typically uncommon and of limited size, but can occur at any site within the adult mammary epithelium. The presence of a stem cell population was shown by demonstrating multi-lineage differentiation within CCO-deficient areas. Interestingly, we observed infrequent CCO deficiency that was restricted to luminal cells, suggesting that niche succession, and by inference stem cell location, is located within the luminal layer. CCO-deficient areas appeared large within areas of ductal carcinoma in situ, suggesting that the rate of clonal expansion was altered in the premalignant lesion. © 2017 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Biancastella Cereser
- Clonal Dynamics in Epithelia Laboratory, Centre for Tumour BiologyBarts Cancer Institute, Queen Mary University of LondonUK
| | - Marnix Jansen
- Epithelial Stem Cell Laboratory, Centre for Tumour BiologyBarts Cancer Institute, Queen Mary University of LondonUK
| | - Emily Austin
- Centre for Histopathology Laboratory, Barts Cancer InstituteQueen Mary University of LondonUK
| | - George Elia
- Centre for Histopathology Laboratory, Barts Cancer InstituteQueen Mary University of LondonUK
| | - Taneisha McFarlane
- Department of Surgery and Cancer, Imperial College LondonCharing Cross HospitalLondonUK
| | - Carolien HM van Deurzen
- Department of Pathology, Erasmus MC Cancer InstituteErasmus University Medical CenterRotterdamThe Netherlands
| | - Anieta M Sieuwerts
- Department of Medical Oncology, Erasmus MC Cancer InstituteErasmus University Medical Center, RotterdamThe Netherlands
| | - Maria G Daidone
- Department of Experimental Oncology and Molecular MedicineFondazione IRCCS Istituto Nazionale dei TumoriMilanItaly
| | - Paul J Tadrous
- Department of Cellular PathologyNorthwick Park HospitalLondonUK
| | - Nicholas A Wright
- Epithelial Stem Cell Laboratory, Centre for Tumour BiologyBarts Cancer Institute, Queen Mary University of LondonUK
| | - Louise Jones
- Breast Cancer Laboratory, Centre for Tumour BiologyBarts Cancer Institute, Queen Mary University of LondonUK
| | - Stuart AC McDonald
- Clonal Dynamics in Epithelia Laboratory, Centre for Tumour BiologyBarts Cancer Institute, Queen Mary University of LondonUK
| |
Collapse
|
19
|
Extracellular Matrix Stiffness Exists in a Feedback Loop that Drives Tumor Progression. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1092:57-67. [PMID: 30368748 DOI: 10.1007/978-3-319-95294-9_4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cells communicate constantly with their surrounding extracellular matrix (ECM) to maintain homeostasis, using both mechanical and chemical signals. In cancer, abnormal signaling leads to stiffening of the ECM. A stiff microenvironment affects many aspects of the cell, including internal molecular signaling as well as behaviors such as motility and proliferation. Thus, cells and ECM interact in a feedback loop to drive matrix deposition and cross-linking, which alter the mechanical properties of the tissue. Stiffer tissue enhances the invasive potential of a tumor and decreases therapeutic efficacy. This chapter describes how specific molecular effects caused by an abnormally stiff tissue drive macroscopic changes that help determine disease outcome. A complete understanding may foster the generation of new cancer therapies.
Collapse
|
20
|
Characterization of primary human mammary epithelial cells isolated and propagated by conditional reprogrammed cell culture. Oncotarget 2017; 9:11503-11514. [PMID: 29545915 PMCID: PMC5837767 DOI: 10.18632/oncotarget.23817] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 10/30/2017] [Indexed: 01/16/2023] Open
Abstract
Purpose Conditional reprogramming methods allow for the inexhaustible in vitro proliferation of primary epithelial cells from human tissue specimens. This methodology has the potential to enhance the utility of primary cell culture as a model for mammary gland research. However, few studies have systematically characterized this method in generating in vitro normal human mammary epithelial cell models. Results We show that cells derived from fresh normal breast tissues can be propagated and exhibit heterogeneous morphologic features. The cultures are composed of CK18, desmoglein 3, and CK19-positive luminal cells and vimentin, p63, and CK14-positive myoepithelial cells, suggesting the maintenance of in vivo heterogeneity. In addition, the cultures contain subpopulations with different CD49f and EpCAM expression profiles. When grown in 3D conditions, cells self-organize into distinct structures that express either luminal or basal cell markers. Among these structures, CK8-positive cells enclosing a lumen are capable of differentiation into milk-producing cells in the presence of lactogenic stimulus. Furthermore, our short-term cultures retain the expression of ERα, as well as its ability to respond to estrogen stimulation. Materials and Methods We have investigated conditionally reprogrammed normal epithelial cells in terms of cell type heterogeneity, cellular marker expression, and structural arrangement in two-dimensional (2D) and three-dimensional (3D) systems. Conclusions The conditional reprogramming methodology allows generation of a heterogeneous culture from normal human mammary tissue in vitro. We believe that this cell culture model will provide a valuable tool to study mammary cell function and malignant transformation.
Collapse
|
21
|
Abstract
We have devised a culture system with conditions that allow primary breast myoepithelial cells (MEPs) to be passaged in a manner that sustains either nonmyodifferentiated or myodifferentiated cell populations without permitting contaminating luminal cells to grow. We show that progenitor activity and potency of MEPs to generate luminal cells in culture and in vivo rely on maintenance of myodifferentiation. Specific isolation and propagation of topographically distinct MEPs reveal the existence of multipotent progenitors in terminal duct lobular units. These findings have important implications for our understanding of the emergence of candidate luminal precursor cells to human breast cancer. The human breast parenchyma consists of collecting ducts and terminal duct lobular units (TDLUs). The TDLU is the site of origin of most breast cancers. The reason for such focal susceptibility to cancer remains poorly understood. Here, we take advantage of a region-specific heterogeneity in luminal progenitors to interrogate the differentiation repertoire of candidate stem cells in TDLUs. We show that stem-like activity in serial passage culture and in vivo breast morphogenesis relies on the preservation of a myoepithelial phenotype. By enrichment for region-specific progenitors, we identify bipotent and multipotent progenitors in ducts and TDLUs, respectively. We propose that focal breast cancer susceptibility, at least in part, originates from region-specific myoepithelial progenitors.
Collapse
|
22
|
Snowden E, Porter W, Hahn F, Ferguson M, Tong F, Parker JS, Middlebrook A, Ghanekar S, Dillmore WS, Blaesius R. Immunophenotyping and Transcriptomic Outcomes in PDX-Derived TNBC Tissue. Mol Cancer Res 2016; 15:429-438. [PMID: 28039356 DOI: 10.1158/1541-7786.mcr-16-0286-t] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 11/23/2016] [Accepted: 12/21/2016] [Indexed: 11/16/2022]
Abstract
Cancer tissue functions as an ecosystem of a diverse set of cells that interact in a complex tumor microenvironment. Genomic tools applied to biopsies in bulk fail to account for this tumor heterogeneity, whereas single-cell imaging methods limit the number of cells which can be assessed or are very resource intensive. The current study presents methods based on flow cytometric analysis and cell sorting using known cell surface markers (CXCR4/CD184, CD24, THY1/CD90) to identify and interrogate distinct groups of cells in triple-negative breast cancer clinical biopsy specimens from patient-derived xenograft (PDX) models. The results demonstrate that flow cytometric analysis allows a relevant subgrouping of cancer tissue and that sorting of these subgroups provides insights into cancer cell populations with unique, reproducible, and functionally divergent gene expression profiles. The discovery of a drug resistance signature implies that uncovering the functional interaction between these populations will lead to deeper understanding of cancer progression and drug response.Implications: PDX-derived human breast cancer tissue was investigated at the single-cell level, and cell subpopulations defined by surface markers were identified which suggest specific roles for distinct cellular compartments within a solid tumor. Mol Cancer Res; 15(4); 429-38. ©2016 AACR.
Collapse
Affiliation(s)
- Eileen Snowden
- BD Technologies, Research Triangle Park, Durham, North Carolina
| | - Warren Porter
- BD Technologies, Research Triangle Park, Durham, North Carolina
| | - Friedrich Hahn
- BD Technologies, Research Triangle Park, Durham, North Carolina
| | | | - Frances Tong
- BD Technologies, Research Triangle Park, Durham, North Carolina
| | - Joel S Parker
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina.,Department of Genetics, University of North Carolina, Chapel Hill, North Carolina
| | | | | | | | - Rainer Blaesius
- BD Technologies, Research Triangle Park, Durham, North Carolina.
| |
Collapse
|
23
|
Morsing M, Klitgaard MC, Jafari A, Villadsen R, Kassem M, Petersen OW, Rønnov-Jessen L. Evidence of two distinct functionally specialized fibroblast lineages in breast stroma. Breast Cancer Res 2016; 18:108. [PMID: 27809866 PMCID: PMC5093959 DOI: 10.1186/s13058-016-0769-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 10/05/2016] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The terminal duct lobular unit (TDLU) is the most dynamic structure in the human breast and the putative site of origin of human breast cancer. Although stromal cells contribute to a specialized microenvironment in many organs, this component remains largely understudied in the human breast. We here demonstrate the impact on epithelium of two lineages of breast stromal fibroblasts, one of which accumulates in the TDLU while the other resides outside the TDLU in the interlobular stroma. METHODS The two lineages are prospectively isolated by fluorescence activated cell sorting (FACS) based on different expression levels of CD105 and CD26. The characteristics of the two fibroblast lineages are assessed by immunocytochemical staining and gene expression analysis. The differentiation capacity of the two fibroblast populations is determined by exposure to specific differentiating conditions followed by analysis of adipogenic and osteogenic differentiation. To test whether the two fibroblast lineages are functionally imprinted by their site of origin, single cell sorted CD271low/MUC1high normal breast luminal epithelial cells are plated on fibroblast feeders for the observation of morphological development. Epithelial structure formation and polarization is shown by immunofluorescence and digitalized quantification of immunoperoxidase-stained cultures. RESULTS Lobular fibroblasts are CD105high/CD26low while interlobular fibroblasts are CD105low/CD26high. Once isolated the two lineages remain phenotypically stable and functionally distinct in culture. Lobular fibroblasts have properties in common with bone marrow derived mesenchymal stem cells and they specifically convey growth and branching morphogenesis of epithelial progenitors. CONCLUSIONS Two distinct functionally specialized fibroblast lineages exist in the normal human breast, of which the lobular fibroblasts have properties in common with mesenchymal stem cells and support epithelial growth and morphogenesis. We propose that lobular fibroblasts constitute a specialized microenvironment for human breast luminal epithelial progenitors, i.e. the putative precursors of breast cancer.
Collapse
Affiliation(s)
- Mikkel Morsing
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark.,Danish Stem Cell Centre, University of Copenhagen, Copenhagen, Denmark
| | - Marie Christine Klitgaard
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark.,Danish Stem Cell Centre, University of Copenhagen, Copenhagen, Denmark.,Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Abbas Jafari
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark.,Danish Stem Cell Centre, University of Copenhagen, Copenhagen, Denmark
| | - René Villadsen
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark.,Danish Stem Cell Centre, University of Copenhagen, Copenhagen, Denmark
| | - Moustapha Kassem
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark.,Danish Stem Cell Centre, University of Copenhagen, Copenhagen, Denmark.,Laboratory of Molecular Endocrinology, KMEB, Department of Endocrinology, Odense University Hospital and University of Southern Denmark, Odense, Denmark
| | - Ole William Petersen
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark.,Danish Stem Cell Centre, University of Copenhagen, Copenhagen, Denmark
| | | |
Collapse
|
24
|
Niwa O, Barcellos-Hoff MH, Globus RK, Harrison JD, Hendry JH, Jacob P, Martin MT, Seed TM, Shay JW, Story MD, Suzuki K, Yamashita S. ICRP Publication 131: Stem Cell Biology with Respect to Carcinogenesis Aspects of Radiological Protection. Ann ICRP 2016; 44:7-357. [PMID: 26637346 DOI: 10.1177/0146645315595585] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
This report provides a review of stem cells/progenitor cells and their responses to ionising radiation in relation to issues relevant to stochastic effects of radiation that form a major part of the International Commission on Radiological Protection's system of radiological protection. Current information on stem cell characteristics, maintenance and renewal, evolution with age, location in stem cell 'niches', and radiosensitivity to acute and protracted exposures is presented in a series of substantial reviews as annexes concerning haematopoietic tissue, mammary gland, thyroid, digestive tract, lung, skin, and bone. This foundation of knowledge of stem cells is used in the main text of the report to provide a biological insight into issues such as the linear-no-threshold (LNT) model, cancer risk among tissues, dose-rate effects, and changes in the risk of radiation carcinogenesis by age at exposure and attained age. Knowledge of the biology and associated radiation biology of stem cells and progenitor cells is more developed in tissues that renew fairly rapidly, such as haematopoietic tissue, intestinal mucosa, and epidermis, although all the tissues considered here possess stem cell populations. Important features of stem cell maintenance, renewal, and response are the microenvironmental signals operating in the niche residence, for which a well-defined spatial location has been identified in some tissues. The identity of the target cell for carcinogenesis continues to point to the more primitive stem cell population that is mostly quiescent, and hence able to accumulate the protracted sequence of mutations necessary to result in malignancy. In addition, there is some potential for daughter progenitor cells to be target cells in particular cases, such as in haematopoietic tissue and in skin. Several biological processes could contribute to protecting stem cells from mutation accumulation: (a) accurate DNA repair; (b) rapidly induced death of injured stem cells; (c) retention of the DNA parental template strand during divisions in some tissue systems, so that mutations are passed to the daughter differentiating cells and not retained in the parental cell; and (d) stem cell competition, whereby undamaged stem cells outcompete damaged stem cells for residence in the niche. DNA repair mainly occurs within a few days of irradiation, while stem cell competition requires weeks or many months depending on the tissue type. The aforementioned processes may contribute to the differences in carcinogenic radiation risk values between tissues, and may help to explain why a rapidly replicating tissue such as small intestine is less prone to such risk. The processes also provide a mechanistic insight relevant to the LNT model, and the relative and absolute risk models. The radiobiological knowledge also provides a scientific insight into discussions of the dose and dose-rate effectiveness factor currently used in radiological protection guidelines. In addition, the biological information contributes potential reasons for the age-dependent sensitivity to radiation carcinogenesis, including the effects of in-utero exposure.
Collapse
|
25
|
Weigand A, Boos AM, Tasbihi K, Beier JP, Dalton PD, Schrauder M, Horch RE, Beckmann MW, Strissel PL, Strick R. Selective isolation and characterization of primary cells from normal breast and tumors reveal plasticity of adipose derived stem cells. Breast Cancer Res 2016; 18:32. [PMID: 26968831 PMCID: PMC4788819 DOI: 10.1186/s13058-016-0688-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Accepted: 02/19/2016] [Indexed: 02/08/2023] Open
Abstract
Background There is a need to establish more cell lines from breast tumors in contrast to immortalized cell lines from metastatic effusions in order to represent the primary tumor and not principally metastatic biology of breast cancer. This investigation describes the simultaneous isolation, characterization, growth and function of primary mammary epithelial cells (MEC), mesenchymal cells (MES) and adipose derived stem cells (ADSC) from four normal breasts, one inflammatory and one triple-negative ductal breast tumors. Methods A total of 17 cell lines were established and gene expression was analyzed for MEC and MES (n = 42) and ADSC (n = 48) and MUC1, pan-KRT, CD90 and GATA-3 by immunofluorescence. DNA fingerprinting to track cell line identity was performed between original primary tissues and isolates. Functional studies included ADSC differentiation, tumor MES and MEC invasion co-cultured with ADSC-conditioned media (CM) and MES adhesion and growth on 3D-printed scaffolds. Results Comparative analysis showed higher gene expression of EPCAM, CD49f, CDH1 and KRTs for normal MEC lines; MES lines e.g. Vimentin, CD10, ACTA2 and MMP9; and ADSC lines e.g. CD105, CD90, CDH2 and CDH11. Compared to the mean of all four normal breast cell lines, both breast tumor cell lines demonstrated significantly lower ADSC marker gene expression, but higher expression of mesenchymal and invasion gene markers like SNAI1 and MMP2. When compared with four normal ADSC differentiated lineages, both tumor ADSC showed impaired osteogenic and chondrogenic but enhanced adipogenic differentiation and endothelial-like structures, possibly due to high PDGFRB and CD34. Addressing a functional role for overproduction of adipocytes, we initiated 3D-invasion studies including different cell types from the same patient. CM from ADSC differentiating into adipocytes induced tumor MEC 3D-invasion via EMT and amoeboid phenotypes. Normal MES breast cells adhered and proliferated on 3D-printed scaffolds containing 20 fibers, but not on 2.5D-printed scaffolds with single fiber layers, important for tissue engineering. Conclusion Expression analyses confirmed successful simultaneous cell isolations of three different phenotypes from normal and tumor primary breast tissues. Our cell culture studies support that breast-tumor environment differentially regulates tumor ADSC plasticity as well as cell invasion and demonstrates applications for regenerative medicine. Electronic supplementary material The online version of this article (doi:10.1186/s13058-016-0688-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Annika Weigand
- Department of Plastic and Hand Surgery and Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Krankenhausstr. 12, Erlangen, D-91054, Germany.
| | - Anja M Boos
- Department of Plastic and Hand Surgery and Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Krankenhausstr. 12, Erlangen, D-91054, Germany
| | - Kereshmeh Tasbihi
- Department of Plastic and Hand Surgery and Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Krankenhausstr. 12, Erlangen, D-91054, Germany
| | - Justus P Beier
- Department of Plastic and Hand Surgery and Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Krankenhausstr. 12, Erlangen, D-91054, Germany
| | - Paul D Dalton
- Department of Functional Materials in Medicine and Dentistry, University of Würzburg, Pleicherwall 2, Würzburg, Germany
| | - Michael Schrauder
- Department of Obstetrics and Gynecology, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Raymund E Horch
- Department of Plastic and Hand Surgery and Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Krankenhausstr. 12, Erlangen, D-91054, Germany
| | - Matthias W Beckmann
- Department of Obstetrics and Gynecology, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Pamela L Strissel
- Department of Obstetrics and Gynecology, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Reiner Strick
- Department of Obstetrics and Gynecology, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
26
|
Johnston RL, Wockner L, McCart Reed AE, Wiegmans A, Chenevix-Trench G, Khanna KK, Lakhani SR, Smart CE. High content screening application for cell-type specific behaviour in heterogeneous primary breast epithelial subpopulations. Breast Cancer Res 2016; 18:18. [PMID: 26861772 PMCID: PMC4748588 DOI: 10.1186/s13058-016-0681-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 01/29/2016] [Indexed: 12/26/2022] Open
Abstract
Background The complex interaction between multiple cell types and the microenvironment underlies the diverse pathways to carcinogenesis and necessitates sophisticated approaches to in vitro hypotheses testing. The combination of mixed culture format with high content immunofluorescence screening technology provides a powerful platform for observation of cell type specific behavior. Methods We have developed a versatile, high-throughput method for assessing cell-type specific responses. In addition to the specificity and sensitivity offered traditionally by immunofluorescent detection in flow cytometry, the ‘in-cell’ analysis method we describe provides the added benefits of higher throughput and the ability to analyse protein subcellular localisation in situ. Furthermore, elimination of the cell dissociation step allows for more immediate analysis of responses to specific extrinsic stimuli. We applied this method to investigate ionising radiation treatment response in normal breast epithelial cells, measuring growth rate, cell cycle response and double-strand DNA breaks. Results The ‘in-cell’ analysis approach elucidated several interesting donor and cell-type specific differences. Notably, in response to ionizing radiation we observed differential expression in luminal and basal-like cells of a member of the APOBEC enzyme family, recently identified as a critical driver of an oncogenic signature. Our findings suggest that this enzyme is active in the normal breast epithelium during DNA damage response. Conclusions We demonstrate the practical application of a new method for assessing cell-type specific change in mixed cultures, especially the analysis of normal primary cultures, overcoming a major technical issue of dissecting the response of multiple cell types in a heterogeneous population. Electronic supplementary material The online version of this article (doi:10.1186/s13058-016-0681-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Rebecca L Johnston
- The University of Queensland, UQ Centre for Clinical Research, Brisbane, Queensland, 4029, Australia. .,QIMR Berghofer Medical Research Institute, Brisbane, Queensland, 4029, Australia.
| | - Leesa Wockner
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, 4029, Australia.
| | - Amy E McCart Reed
- The University of Queensland, UQ Centre for Clinical Research, Brisbane, Queensland, 4029, Australia. .,QIMR Berghofer Medical Research Institute, Brisbane, Queensland, 4029, Australia.
| | - Adrian Wiegmans
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, 4029, Australia.
| | | | - Kum Kum Khanna
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, 4029, Australia.
| | - Sunil R Lakhani
- The University of Queensland, UQ Centre for Clinical Research, Brisbane, Queensland, 4029, Australia. .,Pathology Queensland, Royal Brisbane and Women's Hospital, Brisbane, Queensland, 4029, Australia. .,The University of Queensland, School of Medicine, Brisbane, Queensland, 4029, Australia.
| | - Chanel E Smart
- The University of Queensland, UQ Centre for Clinical Research, Brisbane, Queensland, 4029, Australia. .,QIMR Berghofer Medical Research Institute, Brisbane, Queensland, 4029, Australia.
| |
Collapse
|
27
|
Ontsouka EC, Bertschi JS, Huang X, Lüthi M, Müller S, Albrecht C. Can widely used cell type markers predict the suitability of immortalized or primary mammary epithelial cell models? Biol Res 2016; 49:1. [PMID: 26739591 PMCID: PMC4702413 DOI: 10.1186/s40659-015-0063-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 12/21/2015] [Indexed: 11/23/2022] Open
Abstract
Background Mammary cell cultures are convenient tools for in vitro studies of mammary gland biology. However, the heterogeneity of mammary cell types, e.g., glandular milk secretory epithelial or myoepithelial cells, often complicates the interpretation of cell-based data. The present study was undertaken to determine the relevance of bovine primary mammary epithelial cells isolated from American Holstein (bMECUS) or Swiss Holstein–Friesian (bMECCH) cows, and of primary bovine mammary alveolar epithelial cells stably transfected with simian virus-40 (SV-40) large T-antigen (MAC-T) for in vitro analyses. This was evaluated by testing their expression pattern of cytokeratin (CK) 7, 18, 19, vimentin, and α-smooth muscle actin (α-SMA). Results The expression of the listed markers was assessed using real-time quantitative PCR, flow cytometry and immunofluorescence microscopy. Characteristic markers of the mesenchymal (vimentin), myoepithelial (α-SMA) and glandular secretory cells (CKs) showed differential expression among the studied cell cultures, partly depending on the analytical method used. The relative mRNA expression of vimentin, CK7 and CK19, respectively, was lower (P < 0.05) in immortalized than in primary mammary cell cultures. The stain index (based on flow cytometry) of CK7 and CK19 protein was lower (P < 0.05) in MAC-T than in bMECs, while the expression of α-SMA and CK18 showed an inverse pattern. Immunofluorescence microscopy analysis mostly confirmed the mRNA data, while partly disagreed with flow cytometry data (e.g., vimentin level in MAC-T). The differential expression of CK7 and CK19 allowed discriminating between immortal and primary mammary cultures. Conclusions The expression of the selected widely used cell type markers in primary and immortalized MEC cells did not allow a clear preference between these two cell models for in vitro analyses studying aspects of milk composition. All tested cell models exhibited to a variable degree epithelial and mesenchymal features. Thus, based on their characterization with widely used cell markers, none of these cultures represent an unequivocal alveolar mammary epithelial cell model. For choosing the appropriate in vitro model additional properties such as the expression profile of specific proteins of interest (e.g., transporter proteins) should equally be taken into account.
Collapse
Affiliation(s)
- Edgar Corneille Ontsouka
- Faculty of Medicine, Institute of Biochemistry and Molecular Medicine, University of Bern, Buehlstrasse 28, 3012, Bern, Switzerland. .,Swiss National Center of Competence in Research, NCCR TransCure, University of Bern, Bern, Switzerland.
| | - Janique Sabina Bertschi
- Faculty of Medicine, Institute of Biochemistry and Molecular Medicine, University of Bern, Buehlstrasse 28, 3012, Bern, Switzerland.
| | - Xiao Huang
- Faculty of Medicine, Institute of Biochemistry and Molecular Medicine, University of Bern, Buehlstrasse 28, 3012, Bern, Switzerland.
| | - Michael Lüthi
- Faculty of Medicine, Institute of Biochemistry and Molecular Medicine, University of Bern, Buehlstrasse 28, 3012, Bern, Switzerland. .,Swiss National Center of Competence in Research, NCCR TransCure, University of Bern, Bern, Switzerland.
| | - Stefan Müller
- Department of Clinical Research, Faculty of Medicine, University of Bern, 3010, Bern, Switzerland.
| | - Christiane Albrecht
- Faculty of Medicine, Institute of Biochemistry and Molecular Medicine, University of Bern, Buehlstrasse 28, 3012, Bern, Switzerland. .,Swiss National Center of Competence in Research, NCCR TransCure, University of Bern, Bern, Switzerland.
| |
Collapse
|
28
|
Jamieson PR, Dekkers JF, Rios AC, Fu NY, Lindeman GJ, Visvader JE. Derivation of a robust mouse mammary organoid system for studying tissue dynamics. Development 2016; 144:1065-1071. [DOI: 10.1242/dev.145045] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 11/30/2016] [Indexed: 12/17/2022]
Abstract
Advances in stem cell research have enabled the generation of mini-organs or organoids that recapitulate phenotypic traits of the original biological specimen. Although organoids have been demonstrated for multiple organ systems, there are more limited options for studying mouse mammary gland formation in vitro. Here we have built upon previously described culture assays to define culture conditions that enable the efficient generation of clonal organoid structures from single-sorted basal mammary epithelial cells (MECs). Analysis of Confetti-reporter mice revealed the formation of uni-coloured structures and thus the clonal nature of these organoids. High resolution 3D imaging demonstrated that basal cell-derived, complex organoids comprised an inner compartment of polarized luminal cells with milk-producing capacity and an outer network of elongated myoepithelial cells. Conversely, structures generated from luminal MECs rarely contained basal/myoepithelial cells. Moreover, flow cytometry and 3D microscopy of organoids generated from lineage-specific reporter mice established the bipotent capacity of basal cells and the restricted potential of luminal cells. In summary, we describe optimized in vitro conditions for the efficient generation of mouse mammary organoids that recapitulate features of mammary tissue architecture and function, and can be applied to understand tissue dynamics and cell-fate decisions.
Collapse
Affiliation(s)
- Paul R. Jamieson
- Stem Cells and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Johanna F. Dekkers
- Stem Cells and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Anne C. Rios
- Stem Cells and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Nai Yang Fu
- Stem Cells and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Geoffrey J. Lindeman
- Stem Cells and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Familial Cancer Centre and Department of Medical Oncology, The Royal Melbourne Hospital, Parkville, VIC 3050, Australia
- Department of Medicine, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Jane E. Visvader
- Stem Cells and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
| |
Collapse
|
29
|
Sánchez-Céspedes R, Millán Y, Guil-Luna S, Reymundo C, Espinosa de Los Monteros A, Martín de Las Mulas J. Myoepithelial cells in canine mammary tumours. Vet J 2015; 207:45-52. [PMID: 26639832 DOI: 10.1016/j.tvjl.2015.10.035] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Revised: 10/07/2015] [Accepted: 10/10/2015] [Indexed: 02/05/2023]
Abstract
Mammary tumours are the most common neoplasms of female dogs. Compared to mammary tumours of humans and cats, myoepithelial (ME) cell involvement is common in canine mammary tumours (CMT) of any subtype. Since ME cell involvement in CMT influences both histogenetic tumour classification and prognosis, correct identification of ME cells is important. This review describes immunohistochemical methods for identification of canine mammary ME cells used in vivo. In addition, phenotypic and genotypic methods to isolate ME cells for in vitro studies to analyse tumour-suppressor protein production and gene expression are discussed. The contribution of ME cells to both histogenetic classifications and the prognosis of CMT is compared with other species and the potential use of ME cells as a method to identify carcinoma in situ is discussed.
Collapse
Affiliation(s)
| | - Yolanda Millán
- Department of Comparative Pathology, University of Córdoba, 14014 Córdoba, Spain
| | - Silvia Guil-Luna
- Department of Comparative Pathology, University of Córdoba, 14014 Córdoba, Spain
| | - Carlos Reymundo
- Department of Pathology, University of Córdoba, 14071 Córdoba, Spain
| | - Antonio Espinosa de Los Monteros
- Unit of Histology and Animal Pathology, Institute for Animal Health, Veterinary School, University of Las Palmas de Gran Canaria, 35413 Las Palmas, Spain
| | | |
Collapse
|
30
|
Chen L, Yin X, Lu S, Chen G, Dong L. Basal cytokeratin phenotypes of myoepithelial cells indicates the origin of ductal carcinomas in situ of the breast. Appl Immunohistochem Mol Morphol 2015; 23:558-64. [PMID: 26336082 DOI: 10.1097/pai.0000000000000123] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Terminal duct lobular unit (TDLU) is widely accepted as the origin of ductal carcinoma in situ of breast. The differentiation states of myoepithelial cells of breast ductal system hint the development of breast hyperplastic lesions. Basal cytokeratin (CK) phenotypes indicate the differentiation of myoepithelial cells. Using antibodies of CK5/6, CK14, and CK17, this study reports the basal CK phenotypes of myoepithelial cells in 20 foci of normal breast, 20 usual ductal hyperplasias, 36 ductal carcinomas in situ (DCIS), and 28 sclerosing adenosis (SA). The results showed that the positive staining of basal CKs of myoepithelial cells in normal ducts were significantly higher than those in normal lobules. The basal CK expression of myoepithelial cells of DCIS and usual ductal hyperplasia was similar to that of normal duct, whereas that of SA was similar to that of normal lobule. We propose a modified model of TDLU origin of intraductal carcinoma that most of DCIS originate from terminal ducts of TDLU, whereas most SA originate from lobules.
Collapse
Affiliation(s)
- Ling Chen
- *Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu †Department of Pathology, Maternity and Child Health Care Hospital, Jiaxing ‡Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou §Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China
| | | | | | | | | |
Collapse
|
31
|
Farhanji B, Latifpour M, Alizadeh AM, Khodayari H, Khodayari S, Khaniki M, Ghasempour S. Tumor suppression effects of myoepithelial cells on mice breast cancer. Eur J Pharmacol 2015; 765:171-8. [PMID: 26297304 DOI: 10.1016/j.ejphar.2015.08.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 08/12/2015] [Accepted: 08/17/2015] [Indexed: 01/20/2023]
Abstract
Several studies have assumed that myoepithelial cells (MECs) loss may contribute to epithelial tumor induction and/or progression. We adopted an in vitro assay and a syngeneic mice breast cancer model with histological and molecular characteristics resembling human lesions to evaluate tumor suppression effects of MECs. Flow cytometric, cell viability, blood chemistry, transmission electron microscope, immunohistochemistry and qRT-PCR assays were performed at the end of the study. We demonstrated that MECs could significantly suppress the viability of cancer cells at different time points (P<0.05). At the end of the fourth and fifth weeks, treated mice had smaller tumor volume compared with control animals. Average tumor volume was significantly less in treated groups than control group at days 21 (0.38±0.19 vs. 1.99±0.13 cm3), 28 (0.57±0.3 vs. 2.5±0.37 cm3) and 35 (0.7±0.35 vs. 2.65±0.4 cm3) after tumor cell injection (P<0.05). No hematological, hepatocellular, and renal toxicities were seen in MECs treated groups. Ultrastructural features revealed severe relationship between adjacent tumoral cells and loose interconnections of neoplastic cells in treated group. Immunohistochemical examinations of breast tumors showed high p63 and low alpha-smooth muscle actin protein expression in treated mice compared to control (P<0.05). MRNA expressions of TNF-α, smooth muscle-myosin heavy chain, connexin 43, and maspin were significantly up-regulated in breast tumor tissues in treated group compared to control (P<0.05). VEGF and alpha-smooth muscle actin mRNA expression were reduced in treated animals (P<0.05). The present study highlighted the potential tumor suppression effects of MECs on breast cancer in a typical animal model.
Collapse
Affiliation(s)
- Baharak Farhanji
- Iranian Tissue Bank & Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mostafa Latifpour
- Cancer Research Center, Tehran University of Medical Sciences, 1419733141 Tehran, Iran
| | - Ali Mohammad Alizadeh
- Cancer Research Center, Tehran University of Medical Sciences, 1419733141 Tehran, Iran.
| | - Hamid Khodayari
- Cancer Research Center, Tehran University of Medical Sciences, 1419733141 Tehran, Iran
| | - Saeed Khodayari
- Cancer Research Center, Tehran University of Medical Sciences, 1419733141 Tehran, Iran; Department of Pharmacology, Fasa University of Medical Sciences, Fasa, Iran
| | - Mahmood Khaniki
- Pathology Department, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sarieh Ghasempour
- Cancer Research Center, Tehran University of Medical Sciences, 1419733141 Tehran, Iran
| |
Collapse
|
32
|
Identification of Personalized Chemoresistance Genes in Subtypes of Basal-Like Breast Cancer Based on Functional Differences Using Pathway Analysis. PLoS One 2015; 10:e0131183. [PMID: 26126114 PMCID: PMC4488356 DOI: 10.1371/journal.pone.0131183] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 05/31/2015] [Indexed: 12/31/2022] Open
Abstract
Breast cancer is a highly heterogeneous disease that is clinically classified into several subtypes. Among these subtypes, basal-like breast cancer largely overlaps with triple-negative breast cancer (TNBC), and these two groups are generally studied together as a single entity. Differences in the molecular makeup of breast cancers can result in different treatment strategies and prognoses for patients with different breast cancer subtypes. Compared with other subtypes, basal-like and other ER+ breast cancer subtypes exhibit marked differences in etiologic factors, clinical characteristics and therapeutic potential. Anthracycline drugs are typically used as the first-line clinical treatment for basal-like breast cancer subtypes. However, certain patients develop drug resistance following chemotherapy, which can lead to disease relapse and death. Even among patients with basal-like breast cancer, there can be significant molecular differences, and it is difficult to identify specific drug resistance proteins in any given patient using conventional variance testing methods. Therefore, we designed a new method for identifying drug resistance genes. Subgroups, personalized biomarkers, and therapy targets were identified using cluster analysis of differentially expressed genes. We found that basal-like breast cancer could be further divided into at least four distinct subgroups, including two groups at risk for drug resistance and two groups characterized by sensitivity to pharmacotherapy. Based on functional differences among these subgroups, we identified nine biomarkers related to drug resistance: SYK, LCK, GAB2, PAWR, PPARG, MDFI, ZAP70, CIITA and ACTA1. Finally, based on the deviation scores of the examined pathways, 16 pathways were shown to exhibit varying degrees of abnormality in the various subgroups, indicating that patients with different subtypes of basal-like breast cancer can be characterized by differences in the functional status of these pathways. Therefore, these nine differentially expressed genes and their associated functional pathways should provide the basis for novel personalized clinical treatments of basal-like breast cancer.
Collapse
|
33
|
Arendt LM, Kuperwasser C. Form and function: how estrogen and progesterone regulate the mammary epithelial hierarchy. J Mammary Gland Biol Neoplasia 2015; 20:9-25. [PMID: 26188694 PMCID: PMC4596764 DOI: 10.1007/s10911-015-9337-0] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 07/08/2015] [Indexed: 12/30/2022] Open
Abstract
The mammary gland undergoes dramatic post-natal growth beginning at puberty, followed by full development occurring during pregnancy and lactation. Following lactation, the alveoli undergo apoptosis, and the mammary gland reverses back to resemble the nonparous gland. This process of growth and regression occurs for multiple pregnancies, suggesting the presence of a hierarchy of stem and progenitor cells that are able to regenerate specialized populations of mammary epithelial cells. Expansion of epithelial cell populations in the mammary gland is regulated by ovarian steroids, in particular estrogen acting through its receptor estrogen receptor alpha (ERα) and progesterone signaling through progesterone receptor (PR). A diverse number of stem and progenitor cells have been identified based on expression of cell surface markers and functional assays. Here we review the current understanding of how estrogen and progesterone act together and separately to regulate stem and progenitor cells within the human and mouse mammary tissues. Better understanding of the hierarchal organization of epithelial cell populations in the mammary gland and how the hormonal milieu affects its regulation may provide important insights into the origins of different subtypes of breast cancer.
Collapse
Affiliation(s)
- Lisa M Arendt
- Developmental, Molecular, and Chemical Biology Department, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, 136 Harrison Ave, Boston, MA, 02111, USA
- Molecular Oncology Research Institute, Tufts Medical Center, 800 Washington St, Boston, MA, 02111, USA
- Raymond and Beverly Sackler Laboratory for the Convergence of Biomedical, Physical and Engineering Sciences, Boston, MA, 02111, USA
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, 2015 Linden Dr, Madison, WI, 53706, USA
| | - Charlotte Kuperwasser
- Developmental, Molecular, and Chemical Biology Department, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, 136 Harrison Ave, Boston, MA, 02111, USA.
- Molecular Oncology Research Institute, Tufts Medical Center, 800 Washington St, Boston, MA, 02111, USA.
- Raymond and Beverly Sackler Laboratory for the Convergence of Biomedical, Physical and Engineering Sciences, Boston, MA, 02111, USA.
- Developmental, Molecular, and Chemical Biology Department, Tufts University School of Medicine, 800 Washington St, Box 5609, Boston, MA, 02111, USA.
| |
Collapse
|
34
|
Hilmarsdóttir B, Briem E, Sigurdsson V, Franzdóttir SR, Ringnér M, Arason AJ, Bergthorsson JT, Magnusson MK, Gudjonsson T. MicroRNA-200c-141 and ∆Np63 are required for breast epithelial differentiation and branching morphogenesis. Dev Biol 2015; 403:150-61. [PMID: 25967125 DOI: 10.1016/j.ydbio.2015.05.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 04/18/2015] [Accepted: 05/05/2015] [Indexed: 02/06/2023]
Abstract
The epithelial compartment of the breast contains two lineages, the luminal- and the myoepithelial cells. D492 is a breast epithelial cell line with stem cell properties that forms branching epithelial structures in 3D culture with both luminal- and myoepithelial differentiation. We have recently shown that D492 undergo epithelial to mesenchymal transition (EMT) when co-cultured with endothelial cells. This 3D co-culture model allows critical analysis of breast epithelial lineage development and EMT. In this study, we compared the microRNA (miR) expression profiles for D492 and its mesenchymal-derivative D492M. Suppression of the miR-200 family in D492M was among the most profound changes observed. Exogenous expression of miR-200c-141 in D492M reversed the EMT phenotype resulting in gain of luminal but not myoepithelial differentiation. In contrast, forced expression of ∆Np63 in D492M restored the myoepithelial phenotype only. Co-expression of miR-200c-141 and ∆Np63 in D492M restored the branching morphogenesis in 3D culture underlining the requirement for both luminal and myoepithelial elements for obtaining full branching morphogenesis in breast epithelium. Introduction of a miR-200c-141 construct in both D492 and D492M resulted in resistance to endothelial induced EMT. In conclusion, our data suggests that expression of miR-200c-141 and ∆Np63 in D492M can reverse EMT resulting in luminal- and myoepithelial differentiation, respectively, demonstrating the importance of these molecules in epithelial integrity in the human breast.
Collapse
Affiliation(s)
- Bylgja Hilmarsdóttir
- Stem Cell Research Unit, Biomedical Center, Department of Anatomy, Faculty of Medicine, School of Health Sciences, University of Iceland, Iceland; Department of Laboratory Hematology, Landspitali-University Hospital, Iceland
| | - Eirikur Briem
- Stem Cell Research Unit, Biomedical Center, Department of Anatomy, Faculty of Medicine, School of Health Sciences, University of Iceland, Iceland; Department of Laboratory Hematology, Landspitali-University Hospital, Iceland
| | - Valgardur Sigurdsson
- Stem Cell Research Unit, Biomedical Center, Department of Anatomy, Faculty of Medicine, School of Health Sciences, University of Iceland, Iceland
| | - Sigrídur Rut Franzdóttir
- Stem Cell Research Unit, Biomedical Center, Department of Anatomy, Faculty of Medicine, School of Health Sciences, University of Iceland, Iceland
| | - Markus Ringnér
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Lund University, Sweden
| | - Ari Jon Arason
- Stem Cell Research Unit, Biomedical Center, Department of Anatomy, Faculty of Medicine, School of Health Sciences, University of Iceland, Iceland; Department of Laboratory Hematology, Landspitali-University Hospital, Iceland
| | - Jon Thor Bergthorsson
- Stem Cell Research Unit, Biomedical Center, Department of Anatomy, Faculty of Medicine, School of Health Sciences, University of Iceland, Iceland; Department of Laboratory Hematology, Landspitali-University Hospital, Iceland
| | - Magnus Karl Magnusson
- Stem Cell Research Unit, Biomedical Center, Department of Anatomy, Faculty of Medicine, School of Health Sciences, University of Iceland, Iceland; Department of Laboratory Hematology, Landspitali-University Hospital, Iceland; Department of Medical Pharmacology and Toxicology, Faculty of Medicine, School of Health Sciences, University of Iceland, Iceland
| | - Thorarinn Gudjonsson
- Stem Cell Research Unit, Biomedical Center, Department of Anatomy, Faculty of Medicine, School of Health Sciences, University of Iceland, Iceland; Department of Laboratory Hematology, Landspitali-University Hospital, Iceland.
| |
Collapse
|
35
|
Hines WC, Yaswen P, Bissell MJ. Modelling breast cancer requires identification and correction of a critical cell lineage-dependent transduction bias. Nat Commun 2015; 6:6927. [PMID: 25896888 PMCID: PMC4411288 DOI: 10.1038/ncomms7927] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 03/13/2015] [Indexed: 01/15/2023] Open
Abstract
Clinically relevant human culture models are essential for developing effective therapies and exploring the biology and etiology of human cancers. Current breast tumour models, such as those from oncogenically transformed primary breast cells, produce predominantly basal-like properties, whereas the more common phenotype expressed by the vast majority of breast tumours are luminal. Reasons for this puzzling, yet important phenomenon, are not understood. We show here that luminal epithelial cells are significantly more resistant to viral transduction than their myoepithelial counterparts. We suggest that this is a significant barrier to generating luminal cell lines and experimental tumours in vivo and to accurate interpretation of results. We show that the resistance is due to lower affinity of luminal cells for virus attachment, which can be overcome by pretreating cells—or virus—with neuraminidase. We present an analytical method for quantifying transductional differences between cell types and an optimized protocol for transducing unsorted primary human breast cells in context. Clinical breast cancers predominantly present luminal features, but experimental models are essentially basal. Here the authors show that luminal cells are significantly less susceptible to viral transduction, and present methods to analyse and overcome the bias in heterogeneous populations.
Collapse
Affiliation(s)
- William C Hines
- Life Sciences Division, Lawrence Berkeley National Laboratory, Mailstop 977R225A, 1 Cyclotron Road, Berkeley, California 94720, USA
| | - Paul Yaswen
- Life Sciences Division, Lawrence Berkeley National Laboratory, Mailstop 977R225A, 1 Cyclotron Road, Berkeley, California 94720, USA
| | - Mina J Bissell
- Life Sciences Division, Lawrence Berkeley National Laboratory, Mailstop 977R225A, 1 Cyclotron Road, Berkeley, California 94720, USA
| |
Collapse
|
36
|
Functional Role of the microRNA-200 Family in Breast Morphogenesis and Neoplasia. Genes (Basel) 2014; 5:804-20. [PMID: 25216122 PMCID: PMC4198932 DOI: 10.3390/genes5030804] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Revised: 09/03/2014] [Accepted: 09/04/2014] [Indexed: 12/21/2022] Open
Abstract
Branching epithelial morphogenesis is closely linked to epithelial-to-mesenchymal transition (EMT), a process important in normal development and cancer progression. The miR-200 family regulates epithelial morphogenesis and EMT through a negative feedback loop with the ZEB1 and ZEB2 transcription factors. miR-200 inhibits expression of ZEB1/2 mRNA, which in turn can down-regulate the miR-200 family that further results in down-regulation of E-cadherin and induction of a mesenchymal phenotype. Recent studies show that the expression of miR-200 genes is high during late pregnancy and lactation, thereby indicating that these miRs are important for breast epithelial morphogenesis and differentiation. miR-200 genes have been studied intensively in relation to breast cancer progression and metastasis, where it has been shown that miR-200 members are down-regulated in basal-like breast cancer where the EMT phenotype is prominent. There is growing evidence that the miR-200 family is up-regulated in distal breast metastasis indicating that these miRs are important for colonization of metastatic breast cancer cells through induction of mesenchymal to epithelial transition. The dual role of miR-200 in primary and metastatic breast cancer is of interest for future therapeutic interventions, making it important to understand its role and interacting partners in more detail.
Collapse
|
37
|
The mammary cellular hierarchy and breast cancer. Cell Mol Life Sci 2014; 71:4301-24. [PMID: 25080108 PMCID: PMC4207940 DOI: 10.1007/s00018-014-1674-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2013] [Revised: 05/29/2014] [Accepted: 06/23/2014] [Indexed: 12/26/2022]
Abstract
Advances in the study of hematopoietic cell maturation have paved the way to a deeper understanding the stem and progenitor cellular hierarchy in the mammary gland. The mammary epithelium, unlike the hematopoietic cellular hierarchy, sits in a complex niche where communication between epithelial cells and signals from the systemic hormonal milieu, as well as from extra-cellular matrix, influence cell fate decisions and contribute to tissue homeostasis. We review the discovery, definition and regulation of the mammary cellular hierarchy and we describe the development of the concepts that have guided our investigations. We outline recent advances in in vivo lineage tracing that is now challenging many of our assumptions regarding the behavior of mammary stem cells, and we show how understanding these cellular lineages has altered our view of breast cancer.
Collapse
|
38
|
|
39
|
Cui SE, Li HM, Liu DL, Nan H, Xu KM, Zhao PR, Liang SW. Human breast adipose‑derived stem cells: characterization and differentiation into mammary gland‑like epithelial cells promoted by autologous activated platelet‑rich plasma. Mol Med Rep 2014; 10:605-14. [PMID: 24890669 DOI: 10.3892/mmr.2014.2280] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Accepted: 04/14/2014] [Indexed: 11/06/2022] Open
Abstract
Human adipose‑derived stem cells (ASCs) isolated from various body sites have been widely investigated in basic and clinical studies. However, ASCs derived from human breast tissue (hbASCs) have not been extensively investigated. In order to expand our understanding of hbASCs and examine their potential applications in stem cell research and cell‑based therapy, hbASCs were isolated from discarded surgical fat tissue following reduction mammoplasty and a comprehensive characterization of these hbASCs was performed, including analysis of their cellular morphology, growth features, cell surface protein markers and multilineage differentiation capacity. These hbASCs expressed cluster of differentiation (CD)44, CD49d, CD90 and CD105, but did not express CD31 and CD34. Subsequently, the hbASCs were differentiated into adipocytes, osteocytes and chondrocytes in vitro. In order to examine the potential applications of hbASCs in breast reconstruction, an approach to promote in vitro differentiation of hbASCs into mammary gland‑like epithelial cells (MGECs) was developed using activated autologous platelet‑rich plasma (PRP). A proliferation phase and a subsequent morphological conversion phase were observed during this differentiation process. PRP significantly promoted the growth of hbASCs in the proliferation phase and increased the eventual conversion rate of hbASCs into MGECs. Thus, to the best of our knowledge, the present study provided the first comprehensive characterization of hbASCs and validated their multipotency. Furthermore, it was revealed that activated autologous PRP was able to enhance the differentiation efficiency of hbASCs into MGECs. The present study and other studies of hbASCs may aid the development of improved breast reconstruction strategies.
Collapse
Affiliation(s)
- Shi-En Cui
- Department of Mammary Gland Surgery, Zhongshan Hospital of Sun Yat‑Sen University, Zhongshan, Guangdong 528403, P.R. China
| | - Hong-Mian Li
- Department of Plastic and Aesthetic Surgery, Zhongshan Bo'ai Hospital of Southern Medical University, Zhongshan, Guangdong 528403, P.R. China
| | - Da-Lie Liu
- Department of Plastic and Reconstructive Surgery, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510282, P.R. China
| | - Hua Nan
- Department of Plastic and Reconstructive Surgery, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510282, P.R. China
| | - Kun-Ming Xu
- Department of Plastic and Aesthetic Surgery, Zhongshan Bo'ai Hospital of Southern Medical University, Zhongshan, Guangdong 528403, P.R. China
| | - Pei-Ran Zhao
- Research Center for Tissue Engineering, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Shuang-Wu Liang
- Research Center for Tissue Engineering, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| |
Collapse
|
40
|
Wahler J, So JY, Kim YC, Liu F, Maehr H, Uskokovic M, Suh N. Inhibition of the transition of ductal carcinoma in situ to invasive ductal carcinoma by a Gemini vitamin D analog. Cancer Prev Res (Phila) 2014; 7:617-26. [PMID: 24691501 DOI: 10.1158/1940-6207.capr-13-0362] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Ductal carcinoma in situ (DCIS) is a nonmalignant lesion of the breast with the potential to progress to invasive ductal carcinoma (IDC). The disappearance and breakdown of the myoepithelial cell layer and basement membrane in DCIS have been identified as major events in the development of breast cancer. The MCF10DCIS.com cell line is a well-established model, which recapitulates the progression of breast cancer from DCIS to IDC. We have previously reported that a novel Gemini vitamin D analog, 1α,25-dihydroxy-20R-21(3-hydroxy-3-deuteromethyl-4,4,4-trideuterobutyl)-23-yne-26,27-hexafluoro-cholecalciferol (BXL0124) is a potent inhibitor of the growth of MCF10DCIS.com xenografted tumors without hypercalcemic toxicity. In this study, we utilized the MCF10DCIS.com in vivo model to assess the effects of BXL0124 on breast cancer progression from weeks 1 to 4. Upon DCIS progression to IDC from weeks 3 to 4, tumors lost the myoepithelial cell layer and basement membrane as shown by immunofluorescence staining with smooth muscle actin and laminin 5, respectively. Administration of BXL0124 maintained the critical myoepithelial cell layer as well as basement membrane, and animals treated with BXL0124 showed a 43% reduction in tumor volume by week 4. BXL0124 treatment decreased cell proliferation and maintained vitamin D receptor levels in tumors. In addition, the BXL0124 treatment reduced the mRNA levels of matrix metalloproteinases starting at week 3, contributing to the inhibition of invasive transition. Our results suggest that the maintenance of DCIS plays a significant role in the cancer preventive action of the Gemini vitamin D BXL0124 during the progression of breast lesions.
Collapse
Affiliation(s)
- Joseph Wahler
- Authors' Affiliations: Department of Chemical Biology, Ernest Mario School of Pharmacy; Center for Advanced Biotechnology and Medicine, Rutgers, The State University of New Jersey, Piscataway; and Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| | - Jae Young So
- Authors' Affiliations: Department of Chemical Biology, Ernest Mario School of Pharmacy; Center for Advanced Biotechnology and Medicine, Rutgers, The State University of New Jersey, Piscataway; and Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| | - Yeoun Chan Kim
- Authors' Affiliations: Department of Chemical Biology, Ernest Mario School of Pharmacy; Center for Advanced Biotechnology and Medicine, Rutgers, The State University of New Jersey, Piscataway; and Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| | - Fang Liu
- Authors' Affiliations: Department of Chemical Biology, Ernest Mario School of Pharmacy; Center for Advanced Biotechnology and Medicine, Rutgers, The State University of New Jersey, Piscataway; and Rutgers Cancer Institute of New Jersey, New Brunswick, New JerseyAuthors' Affiliations: Department of Chemical Biology, Ernest Mario School of Pharmacy; Center for Advanced Biotechnology and Medicine, Rutgers, The State University of New Jersey, Piscataway; and Rutgers Cancer Institute of New Jersey, New Brunswick, New JerseyAuthors' Affiliations: Department of Chemical Biology, Ernest Mario School of Pharmacy; Center for Advanced Biotechnology and Medicine, Rutgers, The State University of New Jersey, Piscataway; and Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| | - Hubert Maehr
- Authors' Affiliations: Department of Chemical Biology, Ernest Mario School of Pharmacy; Center for Advanced Biotechnology and Medicine, Rutgers, The State University of New Jersey, Piscataway; and Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| | - Milan Uskokovic
- Authors' Affiliations: Department of Chemical Biology, Ernest Mario School of Pharmacy; Center for Advanced Biotechnology and Medicine, Rutgers, The State University of New Jersey, Piscataway; and Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| | - Nanjoo Suh
- Authors' Affiliations: Department of Chemical Biology, Ernest Mario School of Pharmacy; Center for Advanced Biotechnology and Medicine, Rutgers, The State University of New Jersey, Piscataway; and Rutgers Cancer Institute of New Jersey, New Brunswick, New JerseyAuthors' Affiliations: Department of Chemical Biology, Ernest Mario School of Pharmacy; Center for Advanced Biotechnology and Medicine, Rutgers, The State University of New Jersey, Piscataway; and Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| |
Collapse
|
41
|
Skibinski A, Breindel JL, Prat A, Galván P, Smith E, Rolfs A, Gupta PB, LaBaer J, Kuperwasser C. The Hippo transducer TAZ interacts with the SWI/SNF complex to regulate breast epithelial lineage commitment. Cell Rep 2014; 6:1059-1072. [PMID: 24613358 PMCID: PMC4011189 DOI: 10.1016/j.celrep.2014.02.038] [Citation(s) in RCA: 132] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Revised: 02/20/2014] [Accepted: 02/25/2014] [Indexed: 12/18/2022] Open
Abstract
Lineage-committed cells of many tissues exhibit substantial plasticity in contexts such as wound healing and tumorigenesis, but the regulation of this process is not well understood. We identified the Hippo transducer WWTR1/TAZ in a screen of transcription factors that are able to prompt lineage switching of mammary epithelial cells. Forced expression of TAZ in luminal cells induces them to adopt basal characteristics, and depletion of TAZ in basal and/or myoepithelial cells leads to luminal differentiation. In human and mouse tissues, TAZ is active only in basal cells and is critical for basal cell maintenance during homeostasis. Accordingly, loss of TAZ affects mammary gland development, leading to an imbalance of luminal and basal populations as well as branching defects. Mechanistically, TAZ interacts with components of the SWI/SNF complex to modulate lineage-specific gene expression. Collectively, these findings uncover a new role for Hippo signaling in the determination of lineage identity through recruitment of chromatin-remodeling complexes.
Collapse
Affiliation(s)
- Adam Skibinski
- Department of Developmental, Chemical, and Molecular Biology, Tufts University, 145 Harrison Avenue, Boston, MA 02111, USA; Molecular Oncology Research Institute, Tufts Medical Center, 800 Washington Street, Boston, MA 02111, USA
| | - Jerrica L Breindel
- Department of Developmental, Chemical, and Molecular Biology, Tufts University, 145 Harrison Avenue, Boston, MA 02111, USA; Molecular Oncology Research Institute, Tufts Medical Center, 800 Washington Street, Boston, MA 02111, USA
| | - Aleix Prat
- Translational Genomics Group, Vall d'Hebron Institute of Oncology, Passeig de la Vall d'Hebron 119-129, Barcelona 08035, Spain
| | - Patricia Galván
- Translational Genomics Group, Vall d'Hebron Institute of Oncology, Passeig de la Vall d'Hebron 119-129, Barcelona 08035, Spain
| | - Elizabeth Smith
- Department of Developmental, Chemical, and Molecular Biology, Tufts University, 145 Harrison Avenue, Boston, MA 02111, USA
| | - Andreas Rolfs
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Piyush B Gupta
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Whitehead Institute for Biomedical Research, Nine Cambridge Center, Cambridge, MA 02142, USA
| | - Joshua LaBaer
- Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, 727 East Tyler Street, Tempe, AZ 85287, USA
| | - Charlotte Kuperwasser
- Department of Developmental, Chemical, and Molecular Biology, Tufts University, 145 Harrison Avenue, Boston, MA 02111, USA; Molecular Oncology Research Institute, Tufts Medical Center, 800 Washington Street, Boston, MA 02111, USA.
| |
Collapse
|
42
|
Grudzien-Nogalska E, Reed BC, Rhoads RE. CPEB1 promotes differentiation and suppresses EMT in mammary epithelial cells. J Cell Sci 2014; 127:2326-38. [PMID: 24634508 DOI: 10.1242/jcs.144956] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Downregulation of CPEB1, a sequence-specific RNA-binding protein, in a mouse mammary epithelial cell line (CID-9) causes epithelial-to-mesenchymal transition (EMT), based on several criteria. First, CPEB1 knockdown decreases protein levels of E-cadherin and β-catenin but increases those of vimentin and Twist1. Second, the motility of CPEB1-depleted cells is increased. Third, CID-9 cells normally form growth-arrested, polarized and three-dimensional acini upon culture in extracellular matrix, but CPEB1-deficient CID-9 cells form nonpolarized proliferating colonies lacking a central cavity. CPEB1 downregulates Twist1 expression by binding to its mRNA, shortening its poly(A) tract and repressing its translation. CID-9 cultures contain both myoepithelial and luminal epithelial cells. CPEB1 increases during CID-9 cell differentiation, is predominantly expressed in myoepithelial cells, and its knockdown prevents expression of the myoepithelial marker p63. CPEB1 is present in proliferating subpopulations of pure luminal epithelial cells (SCp2) and myoepithelial cells (SCg6), but its depletion increases Twist1 only in SCg6 cells and fails to downregulate E-cadherin in SCp2 cells. We propose that myoepithelial cells prevent EMT by influencing the polarity and proliferation of luminal epithelial cells in a mechanism that requires translational silencing of myoepithelial Twist1 by CPEB1.
Collapse
Affiliation(s)
- Ewa Grudzien-Nogalska
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA
| | - Brent C Reed
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA
| | - Robert E Rhoads
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA
| |
Collapse
|
43
|
Mobasheri A, Barrett-Jolley R. Aquaporin water channels in the mammary gland: from physiology to pathophysiology and neoplasia. J Mammary Gland Biol Neoplasia 2014; 19:91-102. [PMID: 24338153 PMCID: PMC3947960 DOI: 10.1007/s10911-013-9312-6] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Accepted: 11/13/2013] [Indexed: 12/21/2022] Open
Abstract
Aquaporins are membrane proteins that play fundamental roles in water and small solute transport across epithelial and endothelial barriers. Recent studies suggest that several aquaporin proteins are present in the mammary gland. Immunohistochemical techniques have confirmed the presence of aquaporin 1 (AQP1) and AQP3 water channels in rat, mouse, bovine and human mammary glands. Studies suggest that in addition to AQP1 and AQP3 AQP4, AQP5 and AQP7 proteins are expressed in different locations in the mammary gland. Aquaporins play key roles in tumor biology and are involved in cell growth, migration and formation of ascites via increased water permeability of micro-vessels. Emerging evidence suggests that expression of these proteins is altered in mammary tumors and in breast cancer cell lines although it is not yet clear whether this is a cause or a consequence of neoplastic development. This review analyzes the expression and potential functional roles of aquaporin water channels in the mammary gland. The physiological mechanisms involved in the transport of water and small solutes across mammary endothelial and epithelial barriers are discussed in the context of milk production and lactation. This paper also reviews papers from the recent cancer literature that implicate aquaporins in mammary neoplasia.
Collapse
Affiliation(s)
- Ali Mobasheri
- School of Pharmacy, University of Bradford, Richmond Road, Bradford, BD7 1DP, UK,
| | | |
Collapse
|
44
|
Abstract
Many organs respond to physiological challenges by changing tissue size or composition. Such changes may originate from tissue-specific stem cells and their supportive environment (niche). The endocrine system is a major effector and conveyor of physiological changes and as such could alter stem cell behavior in various ways. In this review, we examine how hormones affect stem cell biology in four different organs: the ovary, intestine, hematopoietic system, and mammary gland. Hormones control every stage of stem cell life, including establishment, expansion, maintenance, and differentiation. The effects can be cell autonomous or non-cell autonomous through the niche. Moreover, a single hormone can affect different stem cells in different ways or affect the same stem cell differently at various developmental times. The vast complexity and diversity of stem cell responses to hormonal cues allow hormones to coordinate the body's reaction to physiological challenges.
Collapse
Affiliation(s)
- Dana Gancz
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, 76100 Israel; ,
| | | |
Collapse
|
45
|
Sánchez-Céspedes R, Maniscalco L, Iussich S, Martignani E, Guil-Luna S, De Maria R, Martín de Las Mulas J, Millán Y. Isolation, purification, culture and characterisation of myoepithelial cells from normal and neoplastic canine mammary glands using a magnetic-activated cell sorting separation system. Vet J 2013; 197:474-82. [PMID: 23583698 DOI: 10.1016/j.tvjl.2013.03.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2012] [Revised: 02/25/2013] [Accepted: 03/02/2013] [Indexed: 01/08/2023]
Abstract
Mammary gland tumours, the most common malignant neoplasm in bitches, often display myoepithelial (ME) cell proliferation. The aim of this study was to isolate, purify, culture and characterise ME cells from normal and neoplastic canine mammary glands. Monodispersed cells from three normal canine mammary glands and five canine mammary tumours were incubated with an anti-Thy1 antibody and isolated by magnetic-activated cell sorting (MACS). Cells isolated from two normal glands (cell lines CmME-N1 and CmME-N2) and four tumours (cell lines CmME-K1 from a complex carcinoma, CmME-K2 from a simple tubulopapillary carcinoma, and CmME-K3 and CmME-K4 from two carcinomas within benign tumours) were cultured in supplemented DMEM/F12 media for 40days. Cell purity was >90%. Tumour-derived ME cell lines exhibited heterogeneous morphology, growth patterns and immunocytochemical expression of cytokeratins, whereas cell lines from normal glands retained their morphology and levels of cytokeratin expression during culture. Cell lines from normal glands and carcinomas within benign tumours grew more slowly than those from simple and complex carcinomas. This methodology has the potential to be used for in vitro analysis of the role of ME cells in the growth and progression of canine mammary tumours.
Collapse
Affiliation(s)
- R Sánchez-Céspedes
- Department of Comparative Pathology, Veterinary Faculty, University of Córdoba, Córdoba, Spain.
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Expression and functional role of sprouty-2 in breast morphogenesis. PLoS One 2013; 8:e60798. [PMID: 23573284 PMCID: PMC3616012 DOI: 10.1371/journal.pone.0060798] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2012] [Accepted: 03/03/2013] [Indexed: 12/26/2022] Open
Abstract
Branching morphogenesis is a mechanism used by many species for organogenesis and tissue maintenance. Receptor tyrosine kinases (RTKs), including epidermal growth factor receptor (EGFR) and the sprouty protein family are believed to be critical regulators of branching morphogenesis. The aim of this study was to analyze the expression of Sprouty-2 (SPRY2) in the mammary gland and study its role in branching morphogenesis. Human breast epithelial cells, breast tissue and mouse mammary glands were used for expression studies using immunoblotting, real rime PCR and immunohistochemistry. Knockdown of SPRY2 in the breast epithelial stem cell line D492 was done by lentiviral transduction of shRNA constructs targeting SPRY2. Three dimensional culture of D492 with or without endothelial cells was done in reconstituted basement membrane matrix. We show that in the human breast, SPRY2 is predominantly expressed in the luminal epithelial cells of both ducts and lobuli. In the mouse mammary gland, SPRY2 expression is low or absent in the virgin state, while in the pregnant mammary gland SPRY2 is expressed at branching epithelial buds with increased expression during lactation. This expression pattern is closely associated with the activation of the EGFR pathway. Using D492 which generates branching structures in three-dimensional (3D) culture, we show that SPRY2 expression is low during initiation of branching with subsequent increase throughout the branching process. Immunostaining locates expression of phosphorylated SPRY2 and EGFR at the tip of lobular-like, branching ends. SPRY2 knockdown (KD) resulted in increased migration, increased pERK and larger and more complex branching structures indicating a loss of negative feedback control during branching morphogenesis. In D492 co-cultures with endothelial cells, D492 SPRY2 KD generates spindle-like colonies that bear hallmarks of epithelial to mesenchymal transition. These data indicate that SPRY2 is an important regulator of branching morphogenesis and epithelial to mesenchymal transition in the mammary gland.
Collapse
|
47
|
Park JP, Raafat A, Feltracco JA, Blanding WM, Booth BW. Differential gene expression in nuclear label-retaining cells in the developing mouse mammary gland. Stem Cells Dev 2013. [PMID: 23199335 DOI: 10.1089/scd.2012.0496] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The immortal strand theory postulates stem cells protect themselves from DNA replication-associated mutations and subsequent cancer risk through selective segregation of template DNA strands. Stem cells self-renew by asymmetric cellular division. During asymmetric division, stem cells maintain their template DNA strands, while the newly synthesized DNA strands segregate to newly formed daughter cells. Previous studies have demonstrated that self-renewing mammary stem cells originate in the expanding mammary ducts during puberty-associated allometric growth. In this study, we labeled newly forming mammary stem cells with the thymidine analog 5-ethynl-2'-deoxyuridine for 2 weeks during allometric ductal expansion. Cells that incorporate and retain the nuclear label following extended chase periods are termed label-retaining cells (LRCs). A second nuclear label, 5-bromodeoxyuridine, was administered before euthanasia to identify cells traversing the cell cycle. Mammary cells collected following euthanasia were sorted based on nuclear label retention. Members of the Notch and Wnt signaling pathways were found differentially expressed by mammary LRCs. These pathways are involved in the regulation of stem cells in the mouse mammary gland. Upon further analysis, we found that in contrast to non-LRCs, Notch1 and Notch2 are expressed and localized in the nuclei of the LRCs. Expression of Notch-inducible genes, Hes1 and Hey2, was elevated in LRCs. Inhibition of Notch1 by shRNA reduced colony forming potential and label retention by mammary epithelial cells in vitro. These results indicate that genes are differentially regulated in the LRC population of mammary glands and Notch1 mediates asymmetric cell division of mammary progenitor cells.
Collapse
Affiliation(s)
- Jang Pyo Park
- Institute for Biological Interfaces of Engineering, Clemson University, Clemson, South Carolina 29634, USA
| | | | | | | | | |
Collapse
|
48
|
Shehata M, Teschendorff A, Sharp G, Novcic N, Russell IA, Avril S, Prater M, Eirew P, Caldas C, Watson CJ, Stingl J. Phenotypic and functional characterisation of the luminal cell hierarchy of the mammary gland. Breast Cancer Res 2012; 14:R134. [PMID: 23088371 PMCID: PMC4053112 DOI: 10.1186/bcr3334] [Citation(s) in RCA: 233] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2012] [Accepted: 10/15/2012] [Indexed: 01/16/2023] Open
Abstract
Introduction The organisation of the mammary epithelial hierarchy is poorly understood. Our hypothesis is that the luminal cell compartment is more complex than initially described, and that an understanding of the developmental relationships within this lineage will help in understanding the cellular context in which breast tumours occur. Methods We used fluorescence-activated cell sorting along with in vitro and in vivo functional assays to examine the growth and differentiation properties of distinct subsets of human and mouse mammary epithelial cells. We also examined how loss of steroid hormones influenced these populations in vivo. Gene expression profiles were also obtained for all the purified cell populations and correlated to those obtained from breast tumours. Results The luminal cell compartment of the mouse mammary gland can be resolved into nonclonogenic oestrogen receptor-positive (ER+) luminal cells, ER+ luminal progenitors and oestrogen receptor-negative (ER-) luminal progenitors. The ER+ luminal progenitors are unique in regard to cell survival, as they are relatively insensitive to loss of oestrogen and progesterone when compared with the other types of mammary epithelial cells. Analysis of normal human breast tissue reveals a similar hierarchical organisation composed of nonclonogenic luminal cells, and relatively differentiated (EpCAM+CD49f+ALDH-) and undifferentiated (EpCAM+CD49f+ALDH+) luminal progenitors. In addition, approximately one-quarter of human breast samples examined contained an additional population that had a distinct luminal progenitor phenotype, characterised by low expression of ERBB3 and low proliferative potential. Parent-progeny relationship experiments demonstrated that all luminal progenitor populations in both species are highly plastic and, at low frequencies, can generate progeny representing all mammary cell types. The ER- luminal progenitors in the mouse and the ALDH+ luminal progenitors in the human appear to be analogous populations since they both have gene signatures that are associated with alveolar differentiation and resemble those obtained from basal-like breast tumours. Conclusion The luminal cell compartment in the mammary epithelium is more heterogeneous than initially perceived since progenitors of varying levels of luminal cell differentiation and proliferative capacities can be identified. An understanding of these cells will be essential for understanding the origins and the cellular context of human breast tumours.
Collapse
|
49
|
Hilton HN, Graham JD, Kantimm S, Santucci N, Cloosterman D, Huschtscha LI, Mote PA, Clarke CL. Progesterone and estrogen receptors segregate into different cell subpopulations in the normal human breast. Mol Cell Endocrinol 2012; 361:191-201. [PMID: 22580007 DOI: 10.1016/j.mce.2012.04.010] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2012] [Revised: 04/18/2012] [Accepted: 04/20/2012] [Indexed: 11/21/2022]
Abstract
Progesterone is critical in normal breast development and its synthetic derivatives are emerging as major drivers of breast cancer risk. The recent demonstration that progesterone regulates the stem cell compartment in the murine mammary gland, despite the absence of progesterone receptor (PR) in mammary stem cells, highlights the fact that PR distribution in progenitor cell subsets in the human breast remains to be conclusively shown. By utilising two independent cell sorting strategies to fractionate cells into distinct subpopulations enriched for different cell lineage characteristics, we have demonstrated a consistent enrichment of PR transcripts, relative to estrogen receptor transcripts, in the bipotent progenitor subfraction in the normal human breast. We have also shown co-expression of both steroid hormone receptors with basal markers in a subset of human breast cells, and finally we have demonstrated that PR+ bipotent progenitor cells are estrogen-insensitive, and that estrogen regulates PR in mature luminal cells only.
Collapse
Affiliation(s)
- H N Hilton
- Westmead Institute for Cancer Research, Sydney Medical School-Westmead, University of Sydney at Westmead Millennium Institute, Westmead, New South Wales 2145, Australia.
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Issues to be considered when studying cancer in vitro. Crit Rev Oncol Hematol 2012; 85:95-111. [PMID: 22823950 DOI: 10.1016/j.critrevonc.2012.06.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2012] [Revised: 05/31/2012] [Accepted: 06/27/2012] [Indexed: 01/17/2023] Open
Abstract
Various cancer treatment approaches have shown promising results when tested preclinically. The results of clinical trials, however, are often disappointing. While searching for the reasons responsible for their failures, the relevance of experimental and preclinical models has to be taken into account. Possible factors that should be considered, including cell modifications during in vitro cultivation, lack of both the relevant interactions and the structural context in vitro have been summarized in the present review.
Collapse
|