101
|
Characterization of mammary epithelial stem/progenitor cells and their changes with aging in common marmosets. Sci Rep 2016; 6:32190. [PMID: 27558284 PMCID: PMC4997341 DOI: 10.1038/srep32190] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 08/04/2016] [Indexed: 11/21/2022] Open
Abstract
Age is the number one risk factor for breast cancer, yet the underlying mechanisms are unexplored. Age-associated mammary stem cell (MaSC) dysfunction is thought to play an important role in breast cancer carcinogenesis. Non-human primates with their close phylogenetic relationship to humans provide a powerful model system to study the effects of aging on human MaSC. In particular, the common marmoset monkey (Callithrix jacchus) with a relatively short life span is an ideal model for aging research. In the present study, we characterized for the first time the mammary epithelial stem/progenitor cells in the common marmoset. The MaSC-enriched cells formed four major types of morphologically distinct colonies when cultured on plates pre-seeded with irradiated NIH3T3 fibroblasts, and were also capable of forming mammospheres in suspension culture and subsequent formation of 3D organoids in Matrigel culture. Most importantly, these 3D organoids were found to contain stem/progenitor cells that can undergo self-renewal and multi-lineage differentiation both in vitro and in vivo. We also observed a significant decrease of luminal-restricted progenitors with age. Our findings demonstrate that common marmoset mammary stem/progenitor cells can be isolated and quantified with established in vitro and in vivo assays used for mouse and human studies.
Collapse
|
102
|
Distinct breast cancer stem/progenitor cell populations require either HIF1α or loss of PHD3 to expand under hypoxic conditions. Oncotarget 2016; 6:31721-39. [PMID: 26372732 PMCID: PMC4741635 DOI: 10.18632/oncotarget.5564] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 08/10/2015] [Indexed: 12/26/2022] Open
Abstract
The heterogeneous nature of breast cancer is a result of intrinsic tumor complexity and also of the tumor microenvironment, which is known to be hypoxic. We found that hypoxia expands different breast stem/progenitor cell populations (cells with increased aldehyde dehydrogenase activity (Aldefluor+), high mammosphere formation capacity and CD44+CD24−/low cells) both in primary normal epithelial and tumor cells. The presence of the estrogen receptor (ER) limits hypoxia-dependent CD44+CD24−/low cell expansion. We further show that the hypoxia-driven cancer stem-like cell enrichment results from a dedifferentiation process. The enhanced mammosphere formation and Aldefluor+ cell content observed in breast cancer cells relies on hypoxia-inducible factor 1α (HIF1α). In contrast, the CD44+CD24−/low population expansion is HIF1α independent and requires prolyl hydroxylase 3 (PHD3) downregulation, which mimics hypoxic conditions, leading to reduced CD24 expression through activation of NFkB signaling. These studies show that hypoxic conditions expand CSC populations through distinct molecular mechanisms. Thus, potential therapies that combine current treatments for breast cancer with drugs that target CSC should take into account the heterogeneity of the CSC subpopulations.
Collapse
|
103
|
Mammary Development and Breast Cancer: A Wnt Perspective. Cancers (Basel) 2016; 8:cancers8070065. [PMID: 27420097 PMCID: PMC4963807 DOI: 10.3390/cancers8070065] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Revised: 06/30/2016] [Accepted: 07/07/2016] [Indexed: 12/21/2022] Open
Abstract
The Wnt pathway has emerged as a key signaling cascade participating in mammary organogenesis and breast oncogenesis. In this review, we will summarize the current knowledge of how the pathway regulates stem cells and normal development of the mammary gland, and discuss how its various components contribute to breast carcinoma pathology.
Collapse
|
104
|
Jeong Y, Rhee H, Martin S, Klass D, Lin Y, Nguyen LXT, Feng W, Diehn M. Identification and genetic manipulation of human and mouse oesophageal stem cells. Gut 2016; 65:1077-86. [PMID: 25897018 DOI: 10.1136/gutjnl-2014-308491] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 03/28/2015] [Indexed: 12/08/2022]
Abstract
OBJECTIVE Human oesophageal stem cell research is hampered by the lack of an optimal assay system to study self-renewal and differentiation. We aimed to identify and characterise human and mouse oesophageal stem/progenitor cells by establishing 3-dimensional organotypic sphere culture systems for both species. DESIGN Primary oesophageal epithelial cells were freshly isolated and fluorescence-activated cell sorting (FACS)-sorted from human and mouse oesophagus and 3-dimensional organotypic sphere culture systems were developed. The self-renewing potential and differentiation status of novel subpopulations were assessed by sphere-forming ability, cell cycle analysis, immunostaining, qPCR and RNA-Seq. RESULTS Primary human and mouse oesophageal epithelial cells clonally formed esophagospheres consisting of stratified squamous epithelium. Sphere-forming cells could self-renew and form esophagospheres for over 43 passages in vitro and generated stratified squamous epithelium when transplanted under the kidney capsule of immunodeficient mice. Sphere-forming cells were 10-15-fold enriched among human CD49f(hi)CD24(low) cells and murine CD49f(+)CD24(low)CD71(low) cells compared with the most differentiated cells. Genetic elimination of p63 in mouse and human oesophageal cells dramatically decreased esophagosphere formation and basal gene expression while increasing suprabasal gene expression. CONCLUSIONS We developed clonogenic and organotypic culture systems for the quantitative analyses of human and mouse oesophageal stem/progenitor cells and identified novel cell surface marker combinations that enrich for these cells. Using this system, we demonstrate that elimination of p63 inhibits self-renewal of human oesophageal stem/progenitor cells. We anticipate that these esophagosphere culture systems will facilitate studies of oesophageal stem cell biology and may prove useful for ex vivo expansion of human oesophageal stem cells.
Collapse
Affiliation(s)
- Youngtae Jeong
- Stanford Cancer Institute and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Horace Rhee
- Stanford Cancer Institute and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, California, USA
| | - Shanique Martin
- Stanford Cancer Institute and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Daniel Klass
- Stanford Cancer Institute and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Yuan Lin
- Stanford Cancer Institute and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Le Xuan Truong Nguyen
- Stanford Cancer Institute and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Weiguo Feng
- Stanford Cancer Institute and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Maximilian Diehn
- Stanford Cancer Institute and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
105
|
Margan MM, Jitariu AA, Cimpean AM, Nica C, Raica M. Molecular Portrait of the Normal Human Breast Tissue and Its Influence on Breast Carcinogenesis. J Breast Cancer 2016; 19:99-111. [PMID: 27382385 PMCID: PMC4929267 DOI: 10.4048/jbc.2016.19.2.99] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 06/05/2016] [Indexed: 12/12/2022] Open
Abstract
Normal human breast tissue consists of epithelial and nonepithelial cells with different molecular profiles and differentiation grades. This molecular heterogeneity is known to yield abnormal clones that may contribute to the development of breast carcinomas. Stem cells that are found in developing and mature breast tissue are either positive or negative for cytokeratin 19 depending on their subtype. These cells are able to generate carcinogenesis along with mature cells. However, scientific data remains controversial regarding the monoclonal or polyclonal origin of breast carcinomas. The majority of breast carcinomas originate from epithelial cells that normally express BRCA1. The consecutive loss of the BRCA1 gene leads to various abnormalities in epithelial cells. Normal breast epithelial cells also express hypoxia inducible factor (HIF) 1α and HIF-2α that are associated with a high metastatic rate and a poor prognosis for malignant lesions. The nuclear expression of estrogen receptor (ER) and progesterone receptor (PR) in normal human breast tissue is maintained in malignant tissue as well. Several controversies regarding the ability of ER and PR status to predict breast cancer outcome remain. Both ER and PR act as modulators of cell activity in normal human breast tissue. Ki-67 positivity is strongly correlated with tumor grade although its specific role in applied therapy requires further studies. Human epidermal growth factor receptor 2 (HER2) oncoprotein is less expressed in normal human breast specimens but is highly expressed in certain malignant lesions of the breast. Unlike HER2, epidermal growth factor receptor expression is similar in both normal and malignant tissues. Molecular heterogeneity is not only found in breast carcinomas but also in normal breast tissue. Therefore, the molecular mapping of normal human breast tissue might represent a key research area to fully elucidate the mechanisms of breast carcinogenesis.
Collapse
Affiliation(s)
- Madalin Marius Margan
- Department XII-Obstetrics and Gynecology, Neonatology and Perinatal Care, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
| | - Andreea Adriana Jitariu
- Department of Microscopic Morphology/Histology, Angiogenesis Research Center, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
| | - Anca Maria Cimpean
- Department of Microscopic Morphology/Histology, Angiogenesis Research Center, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
| | - Cristian Nica
- Department of Surgery, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
| | - Marius Raica
- Department of Microscopic Morphology/Histology, Angiogenesis Research Center, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
| |
Collapse
|
106
|
Nolan E, Vaillant F, Branstetter D, Pal B, Giner G, Whitehead L, Lok SW, Mann GB, Rohrbach K, Huang LY, Soriano R, Smyth GK, Dougall WC, Visvader JE, Lindeman GJ. RANK ligand as a potential target for breast cancer prevention in BRCA1-mutation carriers. Nat Med 2016; 22:933-9. [PMID: 27322743 DOI: 10.1038/nm.4118] [Citation(s) in RCA: 197] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 05/06/2016] [Indexed: 12/19/2022]
Abstract
Individuals who have mutations in the breast-cancer-susceptibility gene BRCA1 (hereafter referred to as BRCA1-mutation carriers) frequently undergo prophylactic mastectomy to minimize their risk of breast cancer. The identification of an effective prevention therapy therefore remains a 'holy grail' for the field. Precancerous BRCA1(mut/+) tissue harbors an aberrant population of luminal progenitor cells, and deregulated progesterone signaling has been implicated in BRCA1-associated oncogenesis. Coupled with the findings that tumor necrosis factor superfamily member 11 (TNFSF11; also known as RANKL) is a key paracrine effector of progesterone signaling and that RANKL and its receptor TNFRSF11A (also known as RANK) contribute to mammary tumorigenesis, we investigated a role for this pathway in the pre-neoplastic phase of BRCA1-mutation carriers. We identified two subsets of luminal progenitors (RANK(+) and RANK(-)) in histologically normal tissue of BRCA1-mutation carriers and showed that RANK(+) cells are highly proliferative, have grossly aberrant DNA repair and bear a molecular signature similar to that of basal-like breast cancer. These data suggest that RANK(+) and not RANK(-) progenitors are a key target population in these women. Inhibition of RANKL signaling by treatment with denosumab in three-dimensional breast organoids derived from pre-neoplastic BRCA1(mut/+) tissue attenuated progesterone-induced proliferation. Notably, proliferation was markedly reduced in breast biopsies from BRCA1-mutation carriers who were treated with denosumab. Furthermore, inhibition of RANKL in a Brca1-deficient mouse model substantially curtailed mammary tumorigenesis. Taken together, these findings identify a targetable pathway in a putative cell-of-origin population in BRCA1-mutation carriers and implicate RANKL blockade as a promising strategy in the prevention of breast cancer.
Collapse
Affiliation(s)
- Emma Nolan
- ACRF Stem Cells and Cancer Division, Walter and Eliza Hall Institute of Medical Research (WEHI), Parkville, Victoria, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - François Vaillant
- ACRF Stem Cells and Cancer Division, Walter and Eliza Hall Institute of Medical Research (WEHI), Parkville, Victoria, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | | | - Bhupinder Pal
- ACRF Stem Cells and Cancer Division, Walter and Eliza Hall Institute of Medical Research (WEHI), Parkville, Victoria, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Göknur Giner
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia.,Bioinformatics Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Lachlan Whitehead
- Imaging Laboratory, Systems Biology and Personalized Medicine Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Sheau W Lok
- ACRF Stem Cells and Cancer Division, Walter and Eliza Hall Institute of Medical Research (WEHI), Parkville, Victoria, Australia.,Familial Cancer Centre, Royal Melbourne Hospital, Parkville, Victoria, Australia.,Department of Medical Oncology, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Gregory B Mann
- The Breast Service, Royal Melbourne Hospital and Royal Women's Hospital, Parkville, Victoria, Australia.,Department of Surgery, University of Melbourne, Parkville, Victoria, Australia
| | | | - Kathy Rohrbach
- Department of Pathology, Amgen Inc., Seattle, Washington, USA
| | - Li-Ya Huang
- Department of Pathology, Amgen Inc., Seattle, Washington, USA
| | - Rosalia Soriano
- Department of Pathology, Amgen Inc., Seattle, Washington, USA
| | - Gordon K Smyth
- Bioinformatics Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Mathematics and Statistics, University of Melbourne, Parkville, Victoria, Australia
| | | | - Jane E Visvader
- ACRF Stem Cells and Cancer Division, Walter and Eliza Hall Institute of Medical Research (WEHI), Parkville, Victoria, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Geoffrey J Lindeman
- ACRF Stem Cells and Cancer Division, Walter and Eliza Hall Institute of Medical Research (WEHI), Parkville, Victoria, Australia.,Familial Cancer Centre, Royal Melbourne Hospital, Parkville, Victoria, Australia.,Department of Medical Oncology, Royal Melbourne Hospital, Parkville, Victoria, Australia.,Department of Medicine, University of Melbourne, Parkville, Victoria, Australia.,Familial Cancer Centre, Victorian Comprehensive Cancer Centre, Parkville, Victoria, Australia
| |
Collapse
|
107
|
Brooks MD, Burness ML, Wicha MS. Therapeutic Implications of Cellular Heterogeneity and Plasticity in Breast Cancer. Cell Stem Cell 2016; 17:260-71. [PMID: 26340526 DOI: 10.1016/j.stem.2015.08.014] [Citation(s) in RCA: 279] [Impact Index Per Article: 34.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Cellular heterogeneity represents one of the greatest challenges in cancer therapeutics. In many malignancies, this heterogeneity is generated during tumor evolution through a combination of genetic alterations and epigenetic events that recapitulate normal developmental processes including stem cell self-renewal and differentiation. Many, if not most, tumors display similar hierarchal organization, at the apex of which are "stem-like cells" that drive tumor growth, mediate metastasis, and contribute to treatment resistance. Using breast cancer as a model, we discuss how an improved understanding of tumor cellular heterogeneity and plasticity may lead to development of more effective therapeutic strategies.
Collapse
Affiliation(s)
- Michael D Brooks
- Department of Internal Medicine, University of Michigan Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Monika L Burness
- Department of Internal Medicine, University of Michigan Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Max S Wicha
- Department of Internal Medicine, University of Michigan Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
108
|
Sigl V, Owusu-Boaitey K, Joshi PA, Kavirayani A, Wirnsberger G, Novatchkova M, Kozieradzki I, Schramek D, Edokobi N, Hersl J, Sampson A, Odai-Afotey A, Lazaro C, Gonzalez-Suarez E, Pujana MA, Cimba F, Heyn H, Vidal E, Cruickshank J, Berman H, Sarao R, Ticevic M, Uribesalgo I, Tortola L, Rao S, Tan Y, Pfeiler G, Lee EY, Bago-Horvath Z, Kenner L, Popper H, Singer C, Khokha R, Jones LP, Penninger JM. RANKL/RANK control Brca1 mutation- . Cell Res 2016; 26:761-74. [PMID: 27241552 PMCID: PMC5129883 DOI: 10.1038/cr.2016.69] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Revised: 05/24/2016] [Accepted: 05/25/2016] [Indexed: 12/18/2022] Open
Abstract
Breast cancer is the most common female cancer, affecting approximately one in eight women during their life-time. Besides environmental triggers and hormones, inherited mutations in the breast cancer 1 (BRCA1) or BRCA2 genes markedly increase the risk for the development of breast cancer. Here, using two different mouse models, we show that genetic inactivation of the key osteoclast differentiation factor RANK in the mammary epithelium markedly delayed onset, reduced incidence, and attenuated progression of Brca1;p53 mutation-driven mammary cancer. Long-term pharmacological inhibition of the RANK ligand RANKL in mice abolished the occurrence of Brca1 mutation-driven pre-neoplastic lesions. Mechanistically, genetic inactivation of Rank or RANKL/RANK blockade impaired proliferation and expansion of both murine Brca1;p53 mutant mammary stem cells and mammary progenitors from human BRCA1 mutation carriers. In addition, genome variations within the RANK locus were significantly associated with risk of developing breast cancer in women with BRCA1 mutations. Thus, RANKL/RANK control progenitor cell expansion and tumorigenesis in inherited breast cancer. These results present a viable strategy for the possible prevention of breast cancer in BRCA1 mutant patients.
Collapse
Affiliation(s)
- Verena Sigl
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna 1030, Austria
| | - Kwadwo Owusu-Boaitey
- Department of Biological Sciences, University of Maryland-Baltimore County, Baltimore, MD 21250, USA
| | - Purna A Joshi
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada M5G 1L7
| | - Anoop Kavirayani
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna 1030, Austria
| | - Gerald Wirnsberger
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna 1030, Austria
| | - Maria Novatchkova
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna 1030, Austria
| | - Ivona Kozieradzki
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna 1030, Austria
| | - Daniel Schramek
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, 600 University Avenue, Toronto, Ontario, Canada M5G 1X5.,Department of Molecular Genetics, University of Toronto, Ontario, Canada M5S 3E1
| | - Nnamdi Edokobi
- Department of Biological Sciences, University of Maryland-Baltimore County, Baltimore, MD 21250, USA
| | - Jerome Hersl
- Department of Pharmacology, University of Maryland, Baltimore, School of Medicine, Baltimore, MD 21201, USA
| | - Aishia Sampson
- Department of Pharmacology, University of Maryland, Baltimore, School of Medicine, Baltimore, MD 21201, USA
| | - Ashley Odai-Afotey
- Department of Biological Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Conxi Lazaro
- Hereditary Cancer Program, Catalan Institute of Oncology, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Catalonia, Spain
| | - Eva Gonzalez-Suarez
- Cancer Epigenetics and Biology Program, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Catalonia, Spain
| | - Miguel A Pujana
- ProCURE, Catalan Institute of Oncology, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Catalonia, Spain
| | - For Cimba
- Department of Public and Primary Care, Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge, UK
| | - Holger Heyn
- Cancer Epigenetics and Biology Program, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Catalonia, Spain
| | - Enrique Vidal
- Centre for Genomic Regulation, The Barcelona Institute of Science and Technology, University Pompeu Fabra, Barcelona, Catalonia, Spain
| | - Jennifer Cruickshank
- The Campbell Family Institute for Breast Cancer Research, University Health Network, Toronto, Ontario, Canada M5G 1Z5
| | - Hal Berman
- The Campbell Family Institute for Breast Cancer Research, University Health Network, Toronto, Ontario, Canada M5G 1Z5
| | - Renu Sarao
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna 1030, Austria
| | - Melita Ticevic
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna 1030, Austria
| | - Iris Uribesalgo
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna 1030, Austria
| | - Luigi Tortola
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna 1030, Austria
| | - Shuan Rao
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna 1030, Austria
| | - Yen Tan
- Departments of Obstetrics and Gynecology and Comprehensive Cancer Center, Medical University of Vienna, Vienna 1090, Austria
| | - Georg Pfeiler
- Departments of Obstetrics and Gynecology and Comprehensive Cancer Center, Medical University of Vienna, Vienna 1090, Austria
| | - Eva Yhp Lee
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA 92697, USA
| | - Zsuzsanna Bago-Horvath
- Department of Experimental Pathology and Pathology of Laboratory Animals, Medical University Vienna and University of Veterinary Medicine Vienna, Vienna 1090, Austria
| | - Lukas Kenner
- Department of Experimental Pathology and Pathology of Laboratory Animals, Medical University Vienna and University of Veterinary Medicine Vienna, Vienna 1090, Austria.,Ludwig Boltzmann Institute for Cancer Research (LBI-CR), Vienna, Austria
| | - Helmuth Popper
- Research Unit Molecular Lung and Pleura Pathology, Institute of Pathology, Medical University Graz, Graz 8010, Austria
| | - Christian Singer
- Departments of Obstetrics and Gynecology and Comprehensive Cancer Center, Medical University of Vienna, Vienna 1090, Austria
| | - Rama Khokha
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada M5G 1L7
| | - Laundette P Jones
- Department of Pharmacology, University of Maryland, Baltimore, School of Medicine, Baltimore, MD 21201, USA
| | - Josef M Penninger
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna 1030, Austria
| |
Collapse
|
109
|
Chiotaki R, Polioudaki H, Theodoropoulos PA. Stem cell technology in breast cancer: current status and potential applications. STEM CELLS AND CLONING-ADVANCES AND APPLICATIONS 2016; 9:17-29. [PMID: 27217783 PMCID: PMC4853137 DOI: 10.2147/sccaa.s72836] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Breast cancer, the leading cause of cancer among females, is supported by the presence of a rare subset of undifferentiated cells within the tumor, identified as breast cancer stem cells (BCSCs). BCSCs underlie the mechanisms of tumor initiation and sustenance and are implicated in the dissemination of the primary tumor to metastatic sites, as they have been found circulating in the blood of breast cancer patients. The discovery of BCSCs has generated a great amount of interest among the scientific community toward their isolation, molecular characterization, and therapeutic targeting. In this review, after summarizing the literature on molecular characterization of BCSCs and methodologies used for their isolation, we will focus on recent data supporting their molecular and functional heterogeneity. Additionally, following a synopsis of the latest approaches for BCSC targeting, we will specifically emphasize on the therapeutic use of naïve or engineered normal stem cells in the treatment of breast cancer and present contradictory findings challenging their safety.
Collapse
Affiliation(s)
- Rena Chiotaki
- Department of Biochemistry, School of Medicine, University of Crete, Heraklion, Greece
| | - Hara Polioudaki
- Department of Biochemistry, School of Medicine, University of Crete, Heraklion, Greece
| | | |
Collapse
|
110
|
Bhat V, Sun YJ, Weger S, Raouf A. Notch-Induced Expression of FZD7 Requires Noncanonical NOTCH3 Signaling in Human Breast Epithelial Cells. Stem Cells Dev 2016; 25:522-9. [PMID: 26847503 DOI: 10.1089/scd.2015.0315] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The evolutionarily conserved Notch and Wnt signaling pathways have demonstrated roles in normal mammary gland development and in breast carcinogenesis. We previously reported that in human mammary gland, signaling through NOTCH3 alone regulates the commitment of the undifferentiated bipotential progenitors to the luminal cell fate, indicating that NOTCH3 may regulate the expression of unique genes apart from the other Notch receptors. In this study, we used gain of function and loss of function experiments and found that a Wnt signaling receptor, Frizzled7 (FZD7), is a unique and nonredundant target of NOTCH3 in human breast epithelial cells. Interestingly, neither the constitutively active forms of NOTCH1-2, 4 nor loss of expression of these receptors were able to alter expression of FZD7 in human breast epithelial cells. We further show that FZD7-expressing cells are found more frequently in the luminal progenitor-enriched subpopulation of cells obtained from breast reduction samples compared with the undifferentiated bipotent progenitors. Also, we show that NOTCH3-induced expression of FZD7 occurs in the absence of CSL (CBF1-Suppressor of Hairless-Lag-1). Our data suggest that noncanonical Notch signaling through NOTCH3 could modulate Wnt signaling via FZD7 and in this way, might be involved in luminal cell differentiation.
Collapse
Affiliation(s)
- Vasudeva Bhat
- 1 Department of Immunology, Faculty of Health Sciences, University of Manitoba , Winnipeg, Manitoba, Canada .,2 Research Institute for Oncology and Hematology, CancerCare Manitoba , Winnipeg, Manitoba, Canada
| | - Yu Jia Sun
- 1 Department of Immunology, Faculty of Health Sciences, University of Manitoba , Winnipeg, Manitoba, Canada .,2 Research Institute for Oncology and Hematology, CancerCare Manitoba , Winnipeg, Manitoba, Canada
| | - Steve Weger
- 1 Department of Immunology, Faculty of Health Sciences, University of Manitoba , Winnipeg, Manitoba, Canada .,2 Research Institute for Oncology and Hematology, CancerCare Manitoba , Winnipeg, Manitoba, Canada
| | - Afshin Raouf
- 1 Department of Immunology, Faculty of Health Sciences, University of Manitoba , Winnipeg, Manitoba, Canada .,2 Research Institute for Oncology and Hematology, CancerCare Manitoba , Winnipeg, Manitoba, Canada
| |
Collapse
|
111
|
Abstract
Tumor heterogeneity impinges on prognosis, response to therapy, and metastasis. As such, heterogeneity is one of the most important and clinically relevant areas of cancer research. Breast cancer displays frequent intra- and inter-tumor heterogeneity as the result of genetic and non-genetic alterations that often enhance the vigor of cancer cells. In-depth characterization and understanding of the origin of this phenotypic and molecular diversity is paramount to improving diagnosis, the definition of prognostic and predictive biomarkers, and the design of therapeutic strategies. Here, we summarize current knowledge about sources of breast cancer heterogeneity, its consequences, and possible counter-measures. We discuss especially the impact on tumor heterogeneity of the differentiation state of the cell-of-origin, cancer cell plasticity, the microenvironment, and genetic evolution. Factors that enhance cancer cell vigor are clearly detrimental for patients.
Collapse
Affiliation(s)
- Shany Koren
- Friedrich Miescher Institute for Biomedical Research (FMI), Basel, Switzerland
| | | |
Collapse
|
112
|
Abstract
There is substantial evidence that many cancers, including breast cancer, are driven by a population of cells that display stem cell properties. These cells, termed cancer stem cells (CSCs) or tumor initiating cells, not only drive tumor initiation and growth but also mediate tumor metastasis and therapeutic resistance. In this chapter, we summarize current advances in CSC research with a major focus on breast CSCs (BCSCs). We review the prevailing methods to isolate and characterize BCSCs and recent evidence documenting their cellular origins and phenotypic plasticity that enables them to transition between mesenchymal and epithelial-like states. We describe in vitro and clinical evidence that these cells mediate metastasis and treatment resistance in breast cancer, the development of novel strategies to isolate circulating tumor cells (CTCs) that contain CSCs and the use of patient-derived xenograft (PDX) models in preclinical breast cancer research. Lastly, we highlight several signaling pathways that regulate BCSC self-renewal and describe clinical implications of targeting these cells for breast cancer treatment. The development of strategies to effectively target BCSCs has the potential to significantly improve the outcomes for patients with breast cancer.
Collapse
|
113
|
Ke J, Zhao Z, Hong SH, Bai S, He Z, Malik F, Xu J, Zhou L, Chen W, Martin-Trevino R, Wu X, Lan P, Yi Y, Ginestier C, Ibarra I, Shang L, McDermott S, Luther T, Clouthier SG, Wicha MS, Liu S. Role of microRNA221 in regulating normal mammary epithelial hierarchy and breast cancer stem-like cells. Oncotarget 2016; 6:3709-21. [PMID: 25686829 PMCID: PMC4414148 DOI: 10.18632/oncotarget.2888] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 12/09/2014] [Indexed: 12/25/2022] Open
Abstract
Increasing evidence suggests that lineage specific subpopulations and stem-like cells exist in normal and malignant breast tissues. Epigenetic mechanisms maintaining this hierarchical homeostasis remain to be investigated. In this study, we found the level of microRNA221 (miR-221) was higher in stem-like and myoepithelial cells than in luminal cells isolated from normal and malignant breast tissue. In normal breast cells, over-expression of miR-221 generated more myoepithelial cells whereas knock-down of miR-221 increased luminal cells. Over-expression of miR-221 stimulated stem-like cells in luminal type of cancer and the miR-221 level was correlated with clinical outcome in breast cancer patients. Epithelial-mesenchymal transition (EMT) was induced by overexpression of miR-221 in normal and breast cancer cells. The EMT related gene ATXN1 was found to be a miR-221 target gene regulating breast cell hierarchy. In conclusion, we propose that miR-221 contributes to lineage homeostasis of normal and malignant breast epithelium.
Collapse
Affiliation(s)
- Jia Ke
- Department of Colorectal Surgery, Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Zhiju Zhao
- Innovation Center for Cell Biology and The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science & Technology of China, Hefei, Anhui, China
| | - Su-Hyung Hong
- Department of Oral Microbiology, School of Dentistry Kyungpook National University, Jung-gu, Daegu, South Korea
| | - Shoumin Bai
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun-Yat-Sen University, Guangzhou, China
| | - Zhen He
- Department of Colorectal Surgery, Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Fayaz Malik
- Comprehensive Cancer Center, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Jiahui Xu
- Innovation Center for Cell Biology and The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science & Technology of China, Hefei, Anhui, China
| | - Lei Zhou
- Innovation Center for Cell Biology and The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science & Technology of China, Hefei, Anhui, China
| | - Weilong Chen
- Innovation Center for Cell Biology and The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science & Technology of China, Hefei, Anhui, China
| | - Rachel Martin-Trevino
- Comprehensive Cancer Center, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Xiaojian Wu
- Department of Colorectal Surgery, Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Ping Lan
- Department of Colorectal Surgery, Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yongju Yi
- Network Information Center, Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Christophe Ginestier
- Centre de Recherche en Cancérologie de Marseille, Laboratoire d'Oncologie Moléculaire, UMR891 Inserm/Institut Paoli-Calmettes, Université de la Méditerranée, Marseille, France
| | - Ingrid Ibarra
- Cold Spring Harbor Laboratory, Program in Genetics and Bioinformatics, Cold Spring Harbor, NY, USA
| | - Li Shang
- Comprehensive Cancer Center, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Sean McDermott
- Comprehensive Cancer Center, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Tahra Luther
- Comprehensive Cancer Center, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Shawn G Clouthier
- Comprehensive Cancer Center, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Max S Wicha
- Comprehensive Cancer Center, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Suling Liu
- Innovation Center for Cell Biology and The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science & Technology of China, Hefei, Anhui, China
| |
Collapse
|
114
|
Shimono Y, Mukohyama J, Nakamura SI, Minami H. MicroRNA Regulation of Human Breast Cancer Stem Cells. J Clin Med 2015; 5:jcm5010002. [PMID: 26712794 PMCID: PMC4730127 DOI: 10.3390/jcm5010002] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 12/01/2015] [Accepted: 12/21/2015] [Indexed: 12/11/2022] Open
Abstract
MicroRNAs (miRNAs) are involved in virtually all biological processes, including stem cell maintenance, differentiation, and development. The dysregulation of miRNAs is associated with many human diseases including cancer. We have identified a set of miRNAs differentially expressed between human breast cancer stem cells (CSCs) and non-tumorigenic cancer cells. In addition, these miRNAs are similarly upregulated or downregulated in normal mammary stem/progenitor cells. In this review, we mainly describe the miRNAs that are dysregulated in human breast CSCs directly isolated from clinical specimens. The miRNAs and their clusters, such as the miR-200 clusters, miR-183 cluster, miR-221-222 cluster, let-7, miR-142 and miR-214, target the genes and pathways important for stem cell maintenance, such as the self-renewal gene BMI1, apoptosis, Wnt signaling, Notch signaling, and epithelial-to-mesenchymal transition. In addition, the current evidence shows that metastatic breast CSCs acquire a phenotype that is different from the CSCs in a primary site. Thus, clarifying the miRNA regulation of the metastatic breast CSCs will further advance our understanding of the roles of human breast CSCs in tumor progression.
Collapse
Affiliation(s)
- Yohei Shimono
- Division of Molecular and Cellular Biology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan.
- Division of Medical Oncology/Hematology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan.
| | - Junko Mukohyama
- Division of Molecular and Cellular Biology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan.
| | - Shun-Ichi Nakamura
- Division of Molecular and Cellular Biology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan.
- Division of Biochemistry, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan.
| | - Hironobu Minami
- Division of Medical Oncology/Hematology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan.
| |
Collapse
|
115
|
Barcoding reveals complex clonal dynamics of de novo transformed human mammary cells. Nature 2015; 528:267-71. [PMID: 26633636 DOI: 10.1038/nature15742] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 09/22/2015] [Indexed: 02/06/2023]
Abstract
Most human breast cancers have diversified genomically and biologically by the time they become clinically evident. Early events involved in their genesis and the cellular context in which these events occur have thus been difficult to characterize. Here we present the first formal evidence of the shared and independent ability of basal cells and luminal progenitors, isolated from normal human mammary tissue and transduced with a single oncogene (KRAS(G12D)), to produce serially transplantable, polyclonal, invasive ductal carcinomas within 8 weeks of being introduced either subrenally or subcutaneously into immunodeficient mice. DNA barcoding of the initial cells revealed a dramatic change in the numbers and sizes of clones generated from them within 2 weeks, and the first appearance of many 'new' clones in tumours passaged into secondary recipients. Both primary and secondary tumours were phenotypically heterogeneous and primary tumours were categorized transcriptionally as 'normal-like'. This system challenges previous concepts that carcinogenesis in normal human epithelia is necessarily a slow process requiring the acquisition of multiple driver mutations. It also presents the first description of initial events that accompany the genesis and evolution of malignant human mammary cell populations, thereby contributing new understanding of the rapidity with which heterogeneity in their properties can develop.
Collapse
|
116
|
Sreekumar A, Roarty K, Rosen JM. The mammary stem cell hierarchy: a looking glass into heterogeneous breast cancer landscapes. Endocr Relat Cancer 2015; 22. [PMID: 26206777 PMCID: PMC4618079 DOI: 10.1530/erc-15-0263] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The mammary gland is a dynamic organ that undergoes extensive morphogenesis during the different stages of embryonic development, puberty, estrus, pregnancy, lactation and involution. Systemic and local cues underlie this constant tissue remodeling and act by eliciting an intricate pattern of responses in the mammary epithelial and stromal cells. Decades of studies utilizing methods such as transplantation and lineage-tracing have identified a complex hierarchy of mammary stem cells, progenitors and differentiated epithelial cells that fuel mammary epithelial development. Importantly, these studies have extended our understanding of the molecular crosstalk between cell types and the signaling pathways maintaining normal homeostasis that often are deregulated during tumorigenesis. While several questions remain, this research has many implications for breast cancer. Fundamental among these are the identification of the cells of origin for the multiple subtypes of breast cancer and the understanding of tumor heterogeneity. A deeper understanding of these critical questions will unveil novel breast cancer drug targets and treatment paradigms. In this review, we provide a current overview of normal mammary development and tumorigenesis from a stem cell perspective.
Collapse
Affiliation(s)
- Amulya Sreekumar
- Department of Molecular and Cellular BiologyBaylor College of Medicine, One Baylor Plaza, DeBakey Building M638, Houston, Texas 77030, USA
| | - Kevin Roarty
- Department of Molecular and Cellular BiologyBaylor College of Medicine, One Baylor Plaza, DeBakey Building M638, Houston, Texas 77030, USA
| | - Jeffrey M Rosen
- Department of Molecular and Cellular BiologyBaylor College of Medicine, One Baylor Plaza, DeBakey Building M638, Houston, Texas 77030, USA
| |
Collapse
|
117
|
Nie S, McDermott SP, Deol Y, Tan Z, Wicha MS, Lubman DM. A quantitative proteomics analysis of MCF7 breast cancer stem and progenitor cell populations. Proteomics 2015; 15:3772-83. [PMID: 26332018 DOI: 10.1002/pmic.201500002] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 07/21/2015] [Accepted: 08/26/2015] [Indexed: 12/11/2022]
Abstract
Accumulating evidence has demonstrated that breast cancers are initiated and develop from a small population of stem-like cells termed cancer stem cells (CSCs). These cells are hypothesized to mediate tumor metastasis and contribute to therapeutic resistance. However, the molecular regulatory networks responsible for maintaining CSCs in an undifferentiated state have yet to be elucidated. In this study, we used CSC markers to isolate pure breast CSCs fractions (ALDH+ and CD44+CD24- cell populations) and the mature luminal cells (CD49f-EpCAM+) from the MCF7 cell line. Proteomic analysis was performed on these samples and a total of 3304 proteins were identified. A label-free quantitative method was applied to analyze differentially expressed proteins. Using the criteria of greater than twofold changes and p value <0.05, 305, 322 and 98 proteins were identified as significantly different in three pairwise comparisons of ALDH+ versus CD44+CD24-, ALDH+ versus CD49f-EpCAM+ and CD44+CD24- versus CD49f-EpCAM+, respectively. Pathway analysis of differentially expressed proteins by Ingenuity Pathway Analysis (IPA) revealed potential molecular regulatory networks that may regulate CSCs. Selected differential proteins were validated by Western blot assay and immunohistochemical staining. The use of proteomics analysis may increase our understanding of the underlying molecular mechanisms of breast CSCs. This may be of importance in the future development of anti-CSC therapeutics.
Collapse
Affiliation(s)
- Song Nie
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Sean P McDermott
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, MI, USA.,Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Yadwinder Deol
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, MI, USA.,Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Zhijing Tan
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Max S Wicha
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, MI, USA.,Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - David M Lubman
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
118
|
Baratta M, Volpe M, Nucera D, Gabai G, Guzzo N, Faustini M, Martignani E. Differential expression of living mammary epithelial cell subpopulations in milk during lactation in dairy cows. J Dairy Sci 2015; 98:6897-904. [DOI: 10.3168/jds.2015-9369] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 06/23/2015] [Indexed: 11/19/2022]
|
119
|
Clonogenic assay allows for selection of a primitive mammary epithelial cell population in bovine. Exp Cell Res 2015; 338:245-50. [PMID: 26321394 DOI: 10.1016/j.yexcr.2015.08.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Revised: 08/25/2015] [Accepted: 08/26/2015] [Indexed: 01/23/2023]
Abstract
Adult mammary stem cells have been identified in several species including the bovine. They are responsible for the development of the gland and for cyclic remodeling during estrous cycles and pregnancy. Epithelial cell subpopulations exist within the mammary gland. We and others showed previously that the Colony Forming Cell (CFC) assay can be used to detect lineage-restricted mammary progenitors. We carried out CFCs with bovine mammary cells and manually separated colonies with specific morphologies associated with either a luminal or a myoepithelial phenotype. Expression of specific markers was assessed by immunocytochemistry or by flow cytometry to confirm that the manual separation resulted in isolation of phenotipically different cells. When transplanted in recipient immunodeficient mice, we found that only myoepithelial-like colonies gave rise to outgrowths that resembled bovine mammary alveoli, thus proving that adult stem cells were maintained during culture and segregated with myoepithelial cells. After recovery of the cells from the transplanted mice and subsequent progenitor content analysis, we found a tendency to detect a higher progenitor frequency when myoepithelial-like colonies were transplanted. We here demonstrate that bovine adult mammary stem cells can be sustained in short-term culture and that they can be enriched by manually selecting for basal-like morphology.
Collapse
|
120
|
Basak P, Chatterjee S, Weger S, Bruce MC, Murphy LC, Raouf A. Estrogen regulates luminal progenitor cell differentiation through H19 gene expression. Endocr Relat Cancer 2015; 22:505-17. [PMID: 25944846 PMCID: PMC4498491 DOI: 10.1530/erc-15-0105] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Although the role of estrogen signaling in breast cancer development has been extensively studied, the mechanisms that regulate the indispensable role of estrogen in normal mammary gland development have not been well studied. Because of the unavailability of culture system to maintain estrogen-receptor-positive (ERα(+)) cells in vitro, the molecular mechanisms that regulate estrogen/ERα signaling in the normal human breast are unknown. In the present study, we examined the effects of estrogen signaling on ERα(+) human luminal progenitors using a modified matrigel assay and found that estrogen signaling increased the expansion potential of these progenitors. Furthermore, we found that blocking ERα attenuated luminal progenitor expansion and decreased the luminal colony-forming potential of these progenitors. Additionally, blocking ERα decreased H19 expression in the luminal progenitors and led to the development of smaller luminal colonies. We further showed that knocking down the H19 gene in the luminal progenitors significantly decreased the colony-forming potential of the luminal progenitors, and this phenotype could not be rescued by the addition of estrogen. Lastly, we explored the clinical relevance of the estrogen-H19 signaling axis in breast tumors and found that ERα(+) tumors exhibited a higher expression of H19 as compared with ERα(-) tumors and that H19 expression showed a positive correlation with ERα expression in those tumors. Taken together, the present results indicate that the estrogen-ERα-H19 signaling axis plays a role in regulating the proliferation and differentiation potentials of the normal luminal progenitors and that this signaling network may also be important in the development of ER(+) breast cancer tumors.
Collapse
Affiliation(s)
- Pratima Basak
- Department of ImmunologyUniversity of Manitoba, 471 Apotex Centre 750 McDermot Avenue, Winnipeg, Manitoba, Canada R3E 0T5Manitoba Institute of Cell Biology675 McDermot Avenue, Winnipeg, Manitoba, Canada R3E 0V9Department of Biochemistry and Medical GeneticsUniversity of Manitoba, Winnipeg, Manitoba, Canada R3E 0W2 Department of ImmunologyUniversity of Manitoba, 471 Apotex Centre 750 McDermot Avenue, Winnipeg, Manitoba, Canada R3E 0T5Manitoba Institute of Cell Biology675 McDermot Avenue, Winnipeg, Manitoba, Canada R3E 0V9Department of Biochemistry and Medical GeneticsUniversity of Manitoba, Winnipeg, Manitoba, Canada R3E 0W2
| | - Sumanta Chatterjee
- Department of ImmunologyUniversity of Manitoba, 471 Apotex Centre 750 McDermot Avenue, Winnipeg, Manitoba, Canada R3E 0T5Manitoba Institute of Cell Biology675 McDermot Avenue, Winnipeg, Manitoba, Canada R3E 0V9Department of Biochemistry and Medical GeneticsUniversity of Manitoba, Winnipeg, Manitoba, Canada R3E 0W2 Department of ImmunologyUniversity of Manitoba, 471 Apotex Centre 750 McDermot Avenue, Winnipeg, Manitoba, Canada R3E 0T5Manitoba Institute of Cell Biology675 McDermot Avenue, Winnipeg, Manitoba, Canada R3E 0V9Department of Biochemistry and Medical GeneticsUniversity of Manitoba, Winnipeg, Manitoba, Canada R3E 0W2
| | - Steven Weger
- Department of ImmunologyUniversity of Manitoba, 471 Apotex Centre 750 McDermot Avenue, Winnipeg, Manitoba, Canada R3E 0T5Manitoba Institute of Cell Biology675 McDermot Avenue, Winnipeg, Manitoba, Canada R3E 0V9Department of Biochemistry and Medical GeneticsUniversity of Manitoba, Winnipeg, Manitoba, Canada R3E 0W2
| | - M Christine Bruce
- Department of ImmunologyUniversity of Manitoba, 471 Apotex Centre 750 McDermot Avenue, Winnipeg, Manitoba, Canada R3E 0T5Manitoba Institute of Cell Biology675 McDermot Avenue, Winnipeg, Manitoba, Canada R3E 0V9Department of Biochemistry and Medical GeneticsUniversity of Manitoba, Winnipeg, Manitoba, Canada R3E 0W2 Department of ImmunologyUniversity of Manitoba, 471 Apotex Centre 750 McDermot Avenue, Winnipeg, Manitoba, Canada R3E 0T5Manitoba Institute of Cell Biology675 McDermot Avenue, Winnipeg, Manitoba, Canada R3E 0V9Department of Biochemistry and Medical GeneticsUniversity of Manitoba, Winnipeg, Manitoba, Canada R3E 0W2
| | - Leigh C Murphy
- Department of ImmunologyUniversity of Manitoba, 471 Apotex Centre 750 McDermot Avenue, Winnipeg, Manitoba, Canada R3E 0T5Manitoba Institute of Cell Biology675 McDermot Avenue, Winnipeg, Manitoba, Canada R3E 0V9Department of Biochemistry and Medical GeneticsUniversity of Manitoba, Winnipeg, Manitoba, Canada R3E 0W2 Department of ImmunologyUniversity of Manitoba, 471 Apotex Centre 750 McDermot Avenue, Winnipeg, Manitoba, Canada R3E 0T5Manitoba Institute of Cell Biology675 McDermot Avenue, Winnipeg, Manitoba, Canada R3E 0V9Department of Biochemistry and Medical GeneticsUniversity of Manitoba, Winnipeg, Manitoba, Canada R3E 0W2
| | - Afshin Raouf
- Department of ImmunologyUniversity of Manitoba, 471 Apotex Centre 750 McDermot Avenue, Winnipeg, Manitoba, Canada R3E 0T5Manitoba Institute of Cell Biology675 McDermot Avenue, Winnipeg, Manitoba, Canada R3E 0V9Department of Biochemistry and Medical GeneticsUniversity of Manitoba, Winnipeg, Manitoba, Canada R3E 0W2 Department of ImmunologyUniversity of Manitoba, 471 Apotex Centre 750 McDermot Avenue, Winnipeg, Manitoba, Canada R3E 0T5Manitoba Institute of Cell Biology675 McDermot Avenue, Winnipeg, Manitoba, Canada R3E 0V9Department of Biochemistry and Medical GeneticsUniversity of Manitoba, Winnipeg, Manitoba, Canada R3E 0W2
| |
Collapse
|
121
|
Bovine CD49 positive-cell subpopulation remarkably increases in mammary epithelial cells that retain a stem-like phenotype. Res Vet Sci 2015; 102:1-6. [PMID: 26412510 DOI: 10.1016/j.rvsc.2015.07.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 06/30/2015] [Accepted: 07/05/2015] [Indexed: 01/06/2023]
Abstract
We previously proved that adult stem cells reside in the bovine mammary gland and possess an intrinsic potential to generate a functional mammary outgrowth. The aim of this study was to investigate on the immunophenotyping features retained by mammary stem-like cells detected in long term culture. Flow cytometry analysis showed different subpopulations of mammary epithelial cells emerging according to the timing of cell culture. CD49f(+)-cells significantly increased during the culture (p<0.01) and a similar trend was observed, even if less regular, for CD29(+) and ALDH1 positive cell populations. No difference during the culture was observed for CD24 positive cells but after 35 days of culture a subset of cells, CD49f positive, still retained regenerative capabilities in in vivo xenotransplants. These cells were able to form organized pseudo-alveoli when transplanted in immunodeficient mice. These results prove the presence of a multipotent cell subpopulation that retain a strong epithelial induction, confirmed in in vivo xenotransplants with a presumable in vitro expansion of the primitive population of adult mammary stem cells.
Collapse
|
122
|
Phillips S, Kuperwasser C. SLUG: Critical regulator of epithelial cell identity in breast development and cancer. Cell Adh Migr 2015; 8:578-87. [PMID: 25482617 DOI: 10.4161/19336918.2014.972740] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
SLUG, a member of the SNAIL family of transcriptional repressors, is known to play a diverse number of roles in the cell, and its deregulation has been observed in a variety of cancers including breast. Here, we focus on SLUG's role as a master regulator of mammary epithelial cell (MEC) fate and lineage commitment in the normal mammary gland, and discuss how aberrant SLUG expression can influence breast tumor formation, phenotype, and progression. Specifically, we discuss SLUG's involvement in MEC differentiation, stemness, cellular plasticity, and the epithelial to mesenchymal transition (EMT), and highlight the complex connection between these programs during development and disease progression. Undoubtedly, delineating how molecular factors influence lineage identity and cell-state dynamics in the normal mammary gland will contribute to our understanding of breast tumor heterogeneity.
Collapse
Key Words
- BCSC, Breast Cancer Stem Cell
- BM, Basement Membrane
- BRCA1, Breast Cancer Associated 1
- CK, Cytokeratin
- CSC, Cancer Stem Cell
- E-CAD, E-Cadherin
- EMT
- EMT, Epithelial to Mesenchymal Transition
- ERα, Estrogen Receptor
- HDAC, Histone Deacetylasae
- HMECs, Human Mammary Epithelial Cells
- IHC, Immunohistochemical
- LSD1, Lysine Specific Demethylase 1.
- ME, Myoepithelial
- MEC, Mammary Epithelial Cell
- MaSC, Mammary Stem Cell
- SLUG
- SMA, Smooth Muscle Actin
- SNAG, Snai.Gfi-1
- WT, Wild type
- breast cancer
- cellular plasticity
- differentiation
- mammary stem cells
Collapse
Affiliation(s)
- Sarah Phillips
- a Department of Developmental, Molecular & Chemical Biology ; Sackler School of Graduate Biomedical Sciences ; Tufts University School of Medicine ; Boston , MA USA
| | | |
Collapse
|
123
|
Joshi PA, Waterhouse PD, Kannan N, Narala S, Fang H, Di Grappa MA, Jackson HW, Penninger JM, Eaves C, Khokha R. RANK Signaling Amplifies WNT-Responsive Mammary Progenitors through R-SPONDIN1. Stem Cell Reports 2015; 5:31-44. [PMID: 26095608 PMCID: PMC4618445 DOI: 10.1016/j.stemcr.2015.05.012] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 05/14/2015] [Accepted: 05/22/2015] [Indexed: 12/29/2022] Open
Abstract
Systemic and local signals must be integrated by mammary stem and progenitor cells to regulate their cyclic growth and turnover in the adult gland. Here, we show RANK-positive luminal progenitors exhibiting WNT pathway activation are selectively expanded in the human breast during the progesterone-high menstrual phase. To investigate underlying mechanisms, we examined mouse models and found that loss of RANK prevents the proliferation of hormone receptor-negative luminal mammary progenitors and basal cells, an accompanying loss of WNT activation, and, hence, a suppression of lobuloalveologenesis. We also show that R-spondin1 is depleted in RANK-null progenitors, and that its exogenous administration rescues key aspects of RANK deficiency by reinstating a WNT response and mammary cell expansion. Our findings point to a novel role of RANK in dictating WNT responsiveness to mediate hormone-induced changes in the growth dynamics of adult mammary cells. Luminal progenitors are targets of progesterone in the adult human breast Progesterone-induced expansion of mammary epithelial subsets requires RANK RANK signaling targets WNT-responsive ER–PR– luminal progenitors and basal cells RANK controls RSPO1, which rescues defective progenitor expansion in Rank-null state
Collapse
Affiliation(s)
- Purna A Joshi
- Princess Margaret Cancer Centre, Toronto, ON M5G 1L7, Canada
| | | | - Nagarajan Kannan
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, BC V5Z1L3, Canada
| | - Swami Narala
- Princess Margaret Cancer Centre, Toronto, ON M5G 1L7, Canada
| | - Hui Fang
- Princess Margaret Cancer Centre, Toronto, ON M5G 1L7, Canada
| | | | | | - Josef M Penninger
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, 1030 Vienna, Austria
| | - Connie Eaves
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, BC V5Z1L3, Canada
| | - Rama Khokha
- Princess Margaret Cancer Centre, Toronto, ON M5G 1L7, Canada.
| |
Collapse
|
124
|
Pal B, Chen Y, Bert A, Hu Y, Sheridan JM, Beck T, Shi W, Satterley K, Jamieson P, Goodall GJ, Lindeman GJ, Smyth GK, Visvader JE. Integration of microRNA signatures of distinct mammary epithelial cell types with their gene expression and epigenetic portraits. Breast Cancer Res 2015; 17:85. [PMID: 26080807 PMCID: PMC4497411 DOI: 10.1186/s13058-015-0585-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 05/13/2015] [Indexed: 12/21/2022] Open
Abstract
INTRODUCTION MicroRNAs (miRNAs) have been implicated in governing lineage specification and differentiation in multiple organs; however, little is known about their specific roles in mammopoiesis. We have determined the global miRNA expression profiles of functionally distinct epithelial subpopulations in mouse and human mammary tissue, and compared these to their cognate transcriptomes and epigenomes. Finally, the human miRNA signatures were used to interrogate the different subtypes of breast cancer, with a view to determining miRNA networks deregulated during oncogenesis. METHODS RNA from sorted mouse and human mammary cell subpopulations was subjected to miRNA expression analysis using the TaqMan MicroRNA Array. Differentially expressed (DE) miRNAs were correlated with gene expression and histone methylation profiles. Analysis of miRNA signatures of the intrinsic subtypes of breast cancer in The Cancer Genome Atlas (TCGA) database versus those of normal human epithelial subpopulations was performed. RESULTS Unique miRNA signatures characterized each subset (mammary stem cell (MaSC)/basal, luminal progenitor, mature luminal, stromal), with a high degree of conservation across species. Comparison of miRNA and transcriptome profiles for the epithelial subtypes revealed an inverse relationship and pinpointed key developmental genes. Interestingly, expression of the primate-specific miRNA cluster (19q13.4) was found to be restricted to the MaSC/basal subset. Comparative analysis of miRNA signatures with H3 lysine modification maps of the different epithelial subsets revealed a tight correlation between active or repressive marks for the top DE miRNAs, including derepression of miRNAs in Ezh2-deficient cellular subsets. Interrogation of TCGA-identified miRNA profiles with the miRNA signatures of different human subsets revealed specific relationships. CONCLUSIONS The derivation of global miRNA expression profiles for the different mammary subpopulations provides a comprehensive resource for understanding the interplay between miRNA networks and target gene expression. These data have highlighted lineage-specific miRNAs and potential miRNA-mRNA networks, some of which are disrupted in neoplasia. Furthermore, our findings suggest that key developmental miRNAs are regulated by global changes in histone modification, thus linking the mammary epigenome with genome-wide changes in the expression of genes and miRNAs. Comparative miRNA signature analyses between normal breast epithelial cells and breast tumors confirmed an important linkage between luminal progenitor cells and basal-like tumors.
Collapse
Affiliation(s)
- Bhupinder Pal
- ACRF 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.
| | - Yunshun Chen
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia. .,Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia.
| | - Andrew Bert
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, 5000, Australia.
| | - Yifang Hu
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia.
| | - Julie M Sheridan
- ACRF 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.
| | - Tamara Beck
- ACRF 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.
| | - Wei Shi
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia. .,Department of Computing and Information Systems, The University of Melbourne, Parkville, VIC, 3010, Australia.
| | - Keith Satterley
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia.
| | - Paul Jamieson
- ACRF 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.
| | - Gregory J Goodall
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, 5000, Australia. .,School of Medicine and School of Molecular and Biomedical Science, University of Adelaide, Adelaide, SA, 5005, Australia.
| | - Geoffrey J Lindeman
- ACRF Stem Cells and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia. .,Department of Medicine, The University of Melbourne, Parkville, VIC, 3010, Australia. .,Department of Medical Oncology, The Royal Melbourne Hospital, Parkville, VIC, 3010, Australia.
| | - Gordon K Smyth
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia. .,Department of Mathematics and Statistics, The University of Melbourne, Parkville, VIC, 3010, Australia.
| | - Jane E Visvader
- ACRF 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
|
125
|
Linnemann JR, Miura H, Meixner LK, Irmler M, Kloos UJ, Hirschi B, Bartsch HS, Sass S, Beckers J, Theis FJ, Gabka C, Sotlar K, Scheel CH. Quantification of regenerative potential in primary human mammary epithelial cells. Development 2015; 142:3239-51. [PMID: 26071498 PMCID: PMC4582177 DOI: 10.1242/dev.123554] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 05/08/2015] [Indexed: 01/13/2023]
Abstract
We present an organoid regeneration assay in which freshly isolated human mammary epithelial cells are cultured in adherent or floating collagen gels, corresponding to a rigid or compliant matrix environment. In both conditions, luminal progenitors form spheres, whereas basal cells generate branched ductal structures. In compliant but not rigid collagen gels, branching ducts form alveoli at their tips, express basal and luminal markers at correct positions, and display contractility, which is required for alveologenesis. Thereby, branched structures generated in compliant collagen gels resemble terminal ductal-lobular units (TDLUs), the functional units of the mammary gland. Using the membrane metallo-endopeptidase CD10 as a surface marker enriches for TDLU formation and reveals the presence of stromal cells within the CD49f(hi)/EpCAM(-) population. In summary, we describe a defined in vitro assay system to quantify cells with regenerative potential and systematically investigate their interaction with the physical environment at distinct steps of morphogenesis.
Collapse
Affiliation(s)
- Jelena R Linnemann
- Institute of Stem Cell Research, Helmholtz Center for Health and Environmental Research Munich, Neuherberg 85764, Germany
| | - Haruko Miura
- Institute of Stem Cell Research, Helmholtz Center for Health and Environmental Research Munich, Neuherberg 85764, Germany
| | - Lisa K Meixner
- Institute of Stem Cell Research, Helmholtz Center for Health and Environmental Research Munich, Neuherberg 85764, Germany
| | - Martin Irmler
- Institute of Experimental Genetics, Helmholtz Center Munich, Neuherberg 85764, Germany
| | - Uwe J Kloos
- Institute of Stem Cell Research, Helmholtz Center for Health and Environmental Research Munich, Neuherberg 85764, Germany
| | - Benjamin Hirschi
- Institute of Stem Cell Research, Helmholtz Center for Health and Environmental Research Munich, Neuherberg 85764, Germany
| | - Harald S Bartsch
- Institute of Pathology, Medical School, Ludwig Maximilian University Munich, Munich 80337, Germany
| | - Steffen Sass
- Institute of Computational Biology, Helmholtz Center Munich, Neuherberg 85764, Germany
| | - Johannes Beckers
- Institute of Experimental Genetics, Helmholtz Center Munich, Neuherberg 85764, Germany Department of Experimental Genetics, Technical University Munich, Freising 85354, Germany
| | - Fabian J Theis
- Institute of Computational Biology, Helmholtz Center Munich, Neuherberg 85764, Germany Department of Mathematics, Technical University Munich, Garching 85747, Germany
| | - Christian Gabka
- Nymphenburg Clinic for Plastic and Aesthetic Surgery, Munich 80637, Germany
| | - Karl Sotlar
- Institute of Pathology, Medical School, Ludwig Maximilian University Munich, Munich 80337, Germany
| | - Christina H Scheel
- Institute of Stem Cell Research, Helmholtz Center for Health and Environmental Research Munich, Neuherberg 85764, Germany
| |
Collapse
|
126
|
Chiang HC, Elledge R, Larson P, Jatoi I, Li R, Hu Y. Effects of Radiation Therapy on Breast Epithelial Cells in BRCA1/2 Mutation Carriers. BREAST CANCER-BASIC AND CLINICAL RESEARCH 2015; 9:25-9. [PMID: 26052229 PMCID: PMC4454133 DOI: 10.4137/bcbcr.s26774] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 04/27/2015] [Accepted: 04/29/2015] [Indexed: 12/12/2022]
Abstract
Women carrying BRCA1 and BRCA2 mutations have significantly elevated risk of developing breast and ovarian cancers. BRCA1-associated breast cancer likely originates from progenitors of the luminal epithelial lineage. Recent studies indicate that radiation therapy (RT) for BRCA1 cancer patients is associated with lower incidence of developing subsequent ipsilateral breast cancer. In the current study, we analyzed tumor-free breast tissue procured via prophylactic bilateral mastectomy from three BRCA1 and one BRCA2 mutation carriers, who had been previously treated with RT for unilateral breast cancers. Freshly isolated breast cells from the irradiated and nonirradiated breast tissue of the same individuals were subjected to flow cytometry, using established cell-surface markers. Two out of the three BRCA1 carriers and one BRCA2 carrier exhibited significantly diminished luminal cell population in the irradiated breast versus the nonirradiated side. There was also RT-associated reduction in the colony-forming ability of the breast epithelial cells. Our finding suggests that prior RT could result in the depletion of the luminal epithelial compartment and thus reduced incidence of BRCA1/2-associated breast cancer.
Collapse
Affiliation(s)
- Huai-Chin Chiang
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Richard Elledge
- Department of Medicine, Cancer Therapy and Research Center, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Paula Larson
- Methodist Healthcare System, San Antonio, TX, USA
| | - Ismail Jatoi
- Division of Surgical Oncology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Rong Li
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Yanfen Hu
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| |
Collapse
|
127
|
Hilton HN, Clarke CL. Impact of progesterone on stem/progenitor cells in the human breast. J Mammary Gland Biol Neoplasia 2015; 20:27-37. [PMID: 26254191 DOI: 10.1007/s10911-015-9339-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 07/30/2015] [Indexed: 12/15/2022] Open
Abstract
The epithelium of the human breast is made up of a branching ductal-lobular system, which is lined by a single layer of luminal cells surrounded by a contractile basal cell layer. The co-ordinated development of stem/progenitor cells into these luminal and basal cells is fundamentally important for breast morphogenesis. The ovarian steroid hormone, progesterone, is critical in driving proliferation and normal breast development, yet progesterone analogues have also been shown to be a major driver of breast cancer risk. Studies in recent years have revealed an important role for progesterone in stimulating the mammary stem cell compartment in the mouse mammary gland, and growing evidence supports the notion that progesterone also stimulates progenitor cells in both the normal human breast and in breast cancer cells. As changes in cell type composition are one of the hallmark features of breast cancer progression, these observations have critical implications in discerning the mechanisms of how progesterone increases breast cancer risk. This review summarises recent work regarding the impact of progesterone action on the stem/progenitor cell compartment of the human breast.
Collapse
Affiliation(s)
- Heidi N Hilton
- Centre for Cancer Research, Westmead Millennium Institute, University of Sydney Medical School, Westmead, NSW, Australia.
| | - Christine L Clarke
- Centre for Cancer Research, Westmead Millennium Institute, University of Sydney Medical School, Westmead, NSW, Australia
| |
Collapse
|
128
|
Models of breast morphogenesis based on localization of stem cells in the developing mammary lobule. Stem Cell Reports 2015; 4:699-711. [PMID: 25818813 PMCID: PMC4400614 DOI: 10.1016/j.stemcr.2015.02.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 02/19/2015] [Accepted: 02/20/2015] [Indexed: 12/21/2022] Open
Abstract
Characterization of normal breast stem cells is important for understanding their role in breast development and in breast cancer. However, the identity of these cells is a subject of controversy and their localization in the breast epithelium is not known. In this study, we utilized a novel approach to analyze the morphogenesis of mammary lobules, by combining one-dimensional theoretical models and computer-generated 3D fractals. Comparing predictions of these models with immunohistochemical analysis of tissue sections for candidate stem cell markers, we defined distinct areas where stem cells reside in the mammary lobule. An increased representation of stem cells was found in smaller, less developed lobules compared to larger, more mature lobules, with marked differences in the gland of nulliparous versus parous women and that of BRCA1/2 mutation carriers versus non-carriers.
Collapse
|
129
|
Vieira AF, Ribeiro AS, Dionísio MR, Sousa B, Nobre AR, Albergaria A, Santiago-Gómez A, Mendes N, Gerhard R, Schmitt F, Clarke RB, Paredes J. P-cadherin signals through the laminin receptor α6β4 integrin to induce stem cell and invasive properties in basal-like breast cancer cells. Oncotarget 2015; 5:679-92. [PMID: 24553076 PMCID: PMC3996674 DOI: 10.18632/oncotarget.1459] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
P-cadherin is a classical cell-cell adhesion molecule that, in contrast to E-cadherin, has a positive role in breast cancer progression, being considered a poor prognostic factor in this disease. In previous reports, we have shown that this protein induces cancer stem cell and invasive properties to basal-like breast cancer cells. Here, we clarify the downstream signaling pathways that are triggered by P-cadherin to mediate these effects. We demonstrated that P-cadherin inhibition led to a significant decreased adhesion of cancer cells to the basement membrane substrate laminin, as well as to a major reduction in the expression of the laminin receptor α6β4 integrin. Remarkably, the expression of this heterodimer was required for the invasive capacity and increased mammosphere forming efficiency induced by P-cadherin expression. Moreover, we showed that P-cadherin transcriptionally up-regulates the α6 integrin subunit expression and directly interacts with the β4 integrin subunit. We still showed that P-cadherin downstream signaling, in response to laminin, involves the activation of focal adhesion (FAK), Src and AKT kinases. The association between the expression of P-cadherin, α6β4 heterodimer and the active FAK and Src phosphorylated forms was validated in vivo. Our data establish that there is a crosstalk between P-cadherin and the laminin receptor α6β4 integrin signaling pathway, which link has never been previously described. The activation of this heterodimer explains the stem cell and invasive properties induced by P-cadherin to breast cancer cells, pointing to a new molecular mechanism that may be targeted to counteract the effects induced by this adhesion molecule.
Collapse
Affiliation(s)
- André Filipe Vieira
- IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
130
|
Burleigh A, McKinney S, Brimhall J, Yap D, Eirew P, Poon S, Ng V, Wan A, Prentice L, Annab L, Barrett JC, Caldas C, Eaves C, Aparicio S. A co-culture genome-wide RNAi screen with mammary epithelial cells reveals transmembrane signals required for growth and differentiation. Breast Cancer Res 2015; 17:4. [PMID: 25572802 PMCID: PMC4322558 DOI: 10.1186/s13058-014-0510-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 12/18/2014] [Indexed: 02/01/2023] Open
Abstract
INTRODUCTION The extracellular signals regulating mammary epithelial cell growth are of relevance to understanding the pathophysiology of mammary epithelia, yet they remain poorly characterized. In this study, we applied an unbiased approach to understanding the functional role of signalling molecules in several models of normal physiological growth and translated these results to the biological understanding of breast cancer subtypes. METHODS We developed and utilized a cytogenetically normal clonal line of hTERT immortalized human mammary epithelial cells in a fibroblast-enhanced co-culture assay to conduct a genome-wide small interfering RNA (siRNA) screen for evaluation of the functional effect of silencing each gene. Our selected endpoint was inhibition of growth. In rigorous postscreen validation processes, including quantitative RT-PCR, to ensure on-target silencing, deconvolution of pooled siRNAs and independent confirmation of effects with lentiviral short-hairpin RNA constructs, we identified a subset of genes required for mammary epithelial cell growth. Using three-dimensional Matrigel growth and differentiation assays and primary human mammary epithelial cell colony assays, we confirmed that these growth effects were not limited to the 184-hTERT cell line. We utilized the METABRIC dataset of 1,998 breast cancer patients to evaluate both the differential expression of these genes across breast cancer subtypes and their prognostic significance. RESULTS We identified 47 genes that are critically important for fibroblast-enhanced mammary epithelial cell growth. This group was enriched for several axonal guidance molecules and G protein-coupled receptors, as well as for the endothelin receptor PROCR. The majority of genes (43 of 47) identified in two dimensions were also required for three-dimensional growth, with HSD17B2, SNN and PROCR showing greater than tenfold reductions in acinar formation. Several genes, including PROCR and the neuronal pathfinding molecules EFNA4 and NTN1, were also required for proper differentiation and polarization in three-dimensional cultures. The 47 genes identified showed a significant nonrandom enrichment for differential expression among 10 molecular subtypes of breast cancer sampled from 1,998 patients. CD79A, SERPINH1, KCNJ5 and TMEM14C exhibited breast cancer subtype-independent overall survival differences. CONCLUSION Diverse transmembrane signals are required for mammary epithelial cell growth in two-dimensional and three-dimensional conditions. Strikingly, we define novel roles for axonal pathfinding receptors and ligands and the endothelin receptor in both growth and differentiation.
Collapse
Affiliation(s)
- Angela Burleigh
- Department of Pathology and Laboratory Medicine, University of British Columbia, and BC Cancer Agency, 675 West 10th Avenue, Vancouver, BC, V5Z 1L3, Canada.
| | - Steven McKinney
- Department of Pathology and Laboratory Medicine, University of British Columbia, and BC Cancer Agency, 675 West 10th Avenue, Vancouver, BC, V5Z 1L3, Canada.
| | - Jazmine Brimhall
- Department of Pathology and Laboratory Medicine, University of British Columbia, and BC Cancer Agency, 675 West 10th Avenue, Vancouver, BC, V5Z 1L3, Canada.
| | - Damian Yap
- Department of Pathology and Laboratory Medicine, University of British Columbia, and BC Cancer Agency, 675 West 10th Avenue, Vancouver, BC, V5Z 1L3, Canada.
| | - Peter Eirew
- Department of Pathology and Laboratory Medicine, University of British Columbia, and BC Cancer Agency, 675 West 10th Avenue, Vancouver, BC, V5Z 1L3, Canada.
| | - Steven Poon
- Department of Pathology and Laboratory Medicine, University of British Columbia, and BC Cancer Agency, 675 West 10th Avenue, Vancouver, BC, V5Z 1L3, Canada.
| | - Viola Ng
- Department of Pathology and Laboratory Medicine, University of British Columbia, and BC Cancer Agency, 675 West 10th Avenue, Vancouver, BC, V5Z 1L3, Canada.
| | - Adrian Wan
- Department of Pathology and Laboratory Medicine, University of British Columbia, and BC Cancer Agency, 675 West 10th Avenue, Vancouver, BC, V5Z 1L3, Canada.
| | - Leah Prentice
- Department of Pathology and Laboratory Medicine, University of British Columbia, and BC Cancer Agency, 675 West 10th Avenue, Vancouver, BC, V5Z 1L3, Canada.
- Centre for Translational and Applied Genomics, BC Cancer Agency, 600 West 10th Avenue, Vancouver, BC, V5Z 4E6, Canada.
| | - Lois Annab
- Chromatin and Gene Expression Section, Research Triangle Park, Durham, NC, 27709, USA.
| | - J Carl Barrett
- Laboratory of Molecular Carcinogenesis, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, Durham, NC, 27709, USA.
| | - Carlos Caldas
- Cancer Research UK Cambridge Research Institute and Department of Oncology, University of Cambridge, Li Ka Shin Centre, Cambridge, CB2 0RE, UK.
| | - Connie Eaves
- Terry Fox Laboratory, BC Cancer Agency, Vancouver, BC, V5Z 1L3, Canada.
| | - Samuel Aparicio
- Department of Pathology and Laboratory Medicine, University of British Columbia, and BC Cancer Agency, 675 West 10th Avenue, Vancouver, BC, V5Z 1L3, Canada.
| |
Collapse
|
131
|
Mammary cancer stem cells reinitiation assessment at the metastatic niche: the lung and bone. Methods Mol Biol 2015; 1293:221-9. [PMID: 26040691 DOI: 10.1007/978-1-4939-2519-3_13] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Mammary cancer stem cells (MCSC) have been operationally defined as cells that re-form secondary tumors upon transplantation into immunodeficient mice. Building on this observation, it has also been suggested that MCSCs are responsible for metastasis as well as evasion and resistance to therapeutic treatments. MCSC reinitiating potential is usually tested by implantation of limited amounts of cells orthotopically or subcutaneously, yet this poorly recapitulates the metastatic niche where truly metastatic reinitiation will occur. Herein, we describe the implantation of small amounts of MCSC selected populations in the bone (intra tibiae injection) and the lung (intra thoracic injection) to test for their metastatic reinitiation capabilities.
Collapse
|
132
|
Abstract
Fluorescent-activated cell sorting (FACS) represents one of the key techniques that have been used to isolate and characterize stem cells, including cells from the mammary gland. A combination of approaches, including recognition of cell surface antigens and different cellular activities, has facilitated the identification of stem cells from the healthy mammary gland and from breast tumors. In this chapter we describe the protocol to use FACS to separate breast cancer stem cells, but most of the general principles discussed could be applied to sort other types of cells.
Collapse
Affiliation(s)
- Oihana Iriondo
- CIC bioGUNE, Technological Park of Bizkaia, Building 801A, 48160, Derio, Spain
| | | | | |
Collapse
|
133
|
Nguyen LV, Cox CL, Eirew P, Knapp DJHF, Pellacani D, Kannan N, Carles A, Moksa M, Balani S, Shah S, Hirst M, Aparicio S, Eaves CJ. DNA barcoding reveals diverse growth kinetics of human breast tumour subclones in serially passaged xenografts. Nat Commun 2014; 5:5871. [PMID: 25532760 PMCID: PMC4284657 DOI: 10.1038/ncomms6871] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 11/14/2014] [Indexed: 12/20/2022] Open
Abstract
Genomic and phenotypic analyses indicate extensive intra- as well as intertumoral heterogeneity in primary human malignant cell populations despite their clonal origin. Cellular DNA barcoding offers a powerful and unbiased alternative to track the number and size of multiple subclones within a single human tumour xenograft and their response to continued in vivo passaging. Using this approach we find clone-initiating cell frequencies that vary from ~1/10 to ~1/10,000 cells transplanted for two human breast cancer cell lines and breast cancer xenografts derived from three different patients. For the cell lines, these frequencies are negatively affected in transplants of more than 20,000 cells. Serial transplants reveal five clonal growth patterns (unchanging, expanding, diminishing, fluctuating or of delayed onset), whose predominance is highly variable both between and within original samples. This study thus demonstrates the high growth potential and diverse growth properties of xenografted human breast cancer cells. Cancer cells within the same tumour are heterogeneous in their tumorigenic potential, differentiation status and sensitivity to treatments. Here Nguyen et al. use a sensitive DNA barcoding method to characterize the diversity of clonal growth behaviour within human breast tumours.
Collapse
Affiliation(s)
- Long V Nguyen
- Terry Fox Laboratory, British Columbia Cancer Agency, 675 West 10th Avenue, Vancouver, British Columbia, Canada V5Z 1L3
| | - Claire L Cox
- Terry Fox Laboratory, British Columbia Cancer Agency, 675 West 10th Avenue, Vancouver, British Columbia, Canada V5Z 1L3
| | - Peter Eirew
- Department of Molecular Oncology, British Columbia Cancer Agency, 675 West 10th Avenue, Vancouver, British Columbia, Canada V5Z 1L3
| | - David J H F Knapp
- Terry Fox Laboratory, British Columbia Cancer Agency, 675 West 10th Avenue, Vancouver, British Columbia, Canada V5Z 1L3
| | - Davide Pellacani
- Terry Fox Laboratory, British Columbia Cancer Agency, 675 West 10th Avenue, Vancouver, British Columbia, Canada V5Z 1L3
| | - Nagarajan Kannan
- Terry Fox Laboratory, British Columbia Cancer Agency, 675 West 10th Avenue, Vancouver, British Columbia, Canada V5Z 1L3
| | - Annaick Carles
- Centre for High-Throughput Biology, Department of Microbiology &Immunology, University of British Columbia, 2125 East Mall, Vancouver, British Columbia, Canada V6T 1Z4
| | - Michelle Moksa
- Centre for High-Throughput Biology, Department of Microbiology &Immunology, University of British Columbia, 2125 East Mall, Vancouver, British Columbia, Canada V6T 1Z4
| | - Sneha Balani
- Terry Fox Laboratory, British Columbia Cancer Agency, 675 West 10th Avenue, Vancouver, British Columbia, Canada V5Z 1L3
| | - Sohrab Shah
- Department of Molecular Oncology, British Columbia Cancer Agency, 675 West 10th Avenue, Vancouver, British Columbia, Canada V5Z 1L3
| | - Martin Hirst
- Centre for High-Throughput Biology, Department of Microbiology &Immunology, University of British Columbia, 2125 East Mall, Vancouver, British Columbia, Canada V6T 1Z4
| | - Samuel Aparicio
- Department of Molecular Oncology, British Columbia Cancer Agency, 675 West 10th Avenue, Vancouver, British Columbia, Canada V5Z 1L3
| | - Connie J Eaves
- Terry Fox Laboratory, British Columbia Cancer Agency, 675 West 10th Avenue, Vancouver, British Columbia, Canada V5Z 1L3
| |
Collapse
|
134
|
Verbeke S, Richard E, Monceau E, Schmidt X, Rousseau B, Velasco V, Bernard D, Bonnefoi H, MacGrogan G, Iggo RD. Humanization of the mouse mammary gland by replacement of the luminal layer with genetically engineered preneoplastic human cells. Breast Cancer Res 2014; 16:504. [PMID: 25527189 PMCID: PMC4407301 DOI: 10.1186/s13058-014-0504-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 12/11/2014] [Indexed: 01/09/2023] Open
Abstract
Introduction The cell of origin for estrogen receptor α (ERα) positive breast cancer is
probably a luminal stem cell in the terminal duct lobular units. To model these
cells we have used the murine myoepithelial layer in the mouse mammary ducts as a
scaffold upon which to build a human luminal layer. To prevent squamous
metaplasia, a common artifact in genetically engineered breast cancer models, we
sought to limit activation of the epidermal growth factor receptor (EGFR) during
in vitro cell culture before grafting the
cells. Methods Human reduction mammoplasty cells were grown in
vitro in WIT medium. Epidermal growth factor (EGF) in the medium was
replaced with amphiregulin and neuregulin to decrease activation of EGFR and
increase activation of EGFR homologs 3 and 4 (ERBB3 and ERBB4). Lentiviral vectors
were used to express oncogenic transgenes and fluorescent proteins. Human mammary
epithelial cells were mixed with irradiated mouse fibroblasts and matrigel, then
injected through the nipple into the mammary ducts of immunodeficient mice.
Engrafted cells were visualized by stereomicroscopy for fluorescent proteins and
characterized by histology and immunohistochemistry. Results Growth of normal mammary epithelial cells in conditions favoring ERBB3/4
signaling prevented squamous metaplasia in
vitro. Normal human cells were quickly lost after intraductal
injection but cells infected with lentiviruses expressing CCND1, MYC, TERT, BMI1 and a
short hairpin RNA targeting TP53 were able to
engraft and progressively replace the luminal layer in the mouse mammary ducts,
resulting in the formation of an extensive network of humanized ducts. Despite
expressing multiple oncogenes, the human cells formed a morphologically normal
luminal layer. Expression of a single additional oncogene, PIK3CA-H1047R, converted the
cells into invasive cancer cells. The resulting tumors were ERα+, Ki67+ luminal B
adenocarcinomas that were resistant to treatment with fulvestrant. Conclusions Injection of preneoplastic human mammary epithelial cells into the mammary
ducts of immunodeficient mice leads to replacement of the murine luminal layer
with morphologically normal human cells. Genetic manipulation of the injected
cells makes it possible to study defined steps in the transformation of human
mammary epithelial cells in a more physiological environment than has hitherto
been possible. Electronic supplementary material The online version of this article (doi:10.1186/s13058-014-0504-9) contains supplementary material, which is available to authorized
users.
Collapse
Affiliation(s)
- Stephanie Verbeke
- INSERM U916, Bergonié Cancer Institute, University of Bordeaux, 229 cours de l'Argonne, Bordeaux, 33076, France.
| | - Elodie Richard
- INSERM U916, Bergonié Cancer Institute, University of Bordeaux, 229 cours de l'Argonne, Bordeaux, 33076, France.
| | - Elodie Monceau
- INSERM U916, Bergonié Cancer Institute, University of Bordeaux, 229 cours de l'Argonne, Bordeaux, 33076, France.
| | - Xenia Schmidt
- INSERM U916, Bergonié Cancer Institute, University of Bordeaux, 229 cours de l'Argonne, Bordeaux, 33076, France. .,School of Medicine, University of St Andrews, Medical and Biological Sciences Building, North Haugh, St Andrews, KY16 9TF, UK.
| | - Benoit Rousseau
- Animalerie A2, University of Bordeaux, 146 rue Léo Saignat, 33076, Bordeaux, France.
| | - Valerie Velasco
- INSERM U916, Bergonié Cancer Institute, University of Bordeaux, 229 cours de l'Argonne, Bordeaux, 33076, France. .,Pathology Department, Bergonié Cancer Institute, 229 cours de l'Argonne, 33076, Bordeaux, France.
| | - David Bernard
- INSERM U1052, Centre Leon Berard, University of Lyon, 28 rue Laennec, 69008, Lyon, France.
| | - Herve Bonnefoi
- INSERM U916, Bergonié Cancer Institute, University of Bordeaux, 229 cours de l'Argonne, Bordeaux, 33076, France.
| | - Gaetan MacGrogan
- INSERM U916, Bergonié Cancer Institute, University of Bordeaux, 229 cours de l'Argonne, Bordeaux, 33076, France. .,Pathology Department, Bergonié Cancer Institute, 229 cours de l'Argonne, 33076, Bordeaux, France.
| | - Richard D Iggo
- INSERM U916, Bergonié Cancer Institute, University of Bordeaux, 229 cours de l'Argonne, Bordeaux, 33076, France. .,School of Medicine, University of St Andrews, Medical and Biological Sciences Building, North Haugh, St Andrews, KY16 9TF, UK.
| |
Collapse
|
135
|
Ucar A, Ucar O, Klug P, Matt S, Brunk F, Hofmann TG, Kyewski B. Adult thymus contains FoxN1(-) epithelial stem cells that are bipotent for medullary and cortical thymic epithelial lineages. Immunity 2014; 41:257-69. [PMID: 25148026 PMCID: PMC4148705 DOI: 10.1016/j.immuni.2014.07.005] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 06/13/2014] [Indexed: 12/15/2022]
Abstract
Within the thymus, two major thymic epithelial cell (TEC) subsets—cortical and medullary TECs—provide unique structural and functional niches for T cell development and establishment of central tolerance. Both lineages are believed to originate from a common progenitor cell, yet the cellular and molecular identity of these bipotent TEC progenitors/stem cells remains ill defined. Here we identify rare stromal cells in the murine adult thymus, which under low-attachment conditions formed spheres (termed “thymospheres”). These thymosphere-forming cells (TSFCs) displayed the stemness features of being slow cycling, self-renewing, and bipotent. TSFCs could be significantly enriched based on their distinct surface antigen phenotype. The FoxN1 transcription factor was dispensable for TSFCs maintenance in situ and for commitment to the medullary and cortical TEC lineages. In summary, this study presents the characterization of the adult thymic epithelial stem cells and demonstrates the dispensability of FoxN1 function for their stemness. Adult thymic epithelium contains thymosphere-forming stem cells (TSFCs) TSFCs give rise to both thymic epithelial lineages in vitro and in vivo TSFCs are maintained in the absence of functional FoxN1 Lineage commitment in the sphere is FoxN1 independent
Collapse
Affiliation(s)
- Ahmet Ucar
- Division of Developmental Immunology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Cellular Senescence Group, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany
| | - Olga Ucar
- Division of Developmental Immunology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Paula Klug
- Division of Developmental Immunology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Sonja Matt
- Cellular Senescence Group, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany
| | - Fabian Brunk
- Division of Developmental Immunology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Thomas G Hofmann
- Cellular Senescence Group, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany.
| | - Bruno Kyewski
- Division of Developmental Immunology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| |
Collapse
|
136
|
Granit RZ, Slyper M, Ben-Porath I. Axes of differentiation in breast cancer: untangling stemness, lineage identity, and the epithelial to mesenchymal transition. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2014; 6:93-106. [PMID: 24741710 DOI: 10.1002/wsbm.1252] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Differentiation-associated regulatory programs are central in determining tumor phenotype, and contribute to heterogeneity between tumors and between individual cells within them. The transcriptional programs that control luminal and basal lineage identity in the normal mammary epithelium, as well as progenitor and stem cell function, are active in breast cancers, and show distinct associations with different disease subtypes. Also active in some tumors is the epithelial to mesenchymal transition (EMT) program, which endows carcinoma cells with mesenchymal as well as stem cell traits. The differentiation state of breast cancer cells is thus dictated by the complex combination of regulatory programs, and these can dramatically affect tumor growth and metastatic capacity. Breast cancer differentiation is often viewed along an axis between a basal–mesenchymal–stem cell state and a luminal–epithelial–differentiated state. Here we consider the links, as well as the distinctions, between the three components of this axis: basal versus luminal, mesenchymal versus epithelial, and stem cell versus differentiated identity. Analysis on a multidimensional scale, in which each of these axes is assessed separately, may offer increased resolution of tumor differentiation state. Cancer cells possessing a high degree of stemness would display increased capacity to shift between positions on such a multidimensional scale, and to acquire intermediate phenotypes on its different axes. Further molecular analysis of breast cancer cells with a focus on single-cell profiling, and the development of improved tools for dissection of the circuits controlling gene activity, are essential for the elucidation of the programs dictating breast cancer differentiation state.
Collapse
|
137
|
Arendt LM, Keller PJ, Skibinski A, Goncalves K, Naber SP, Buchsbaum RJ, Gilmore H, Come SE, Kuperwasser C. Anatomical localization of progenitor cells in human breast tissue reveals enrichment of uncommitted cells within immature lobules. Breast Cancer Res 2014; 16:453. [PMID: 25315014 PMCID: PMC4303132 DOI: 10.1186/s13058-014-0453-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 10/01/2014] [Indexed: 11/10/2022] Open
Abstract
INTRODUCTION Lineage tracing studies in mice have revealed the localization and existence of lineage-restricted mammary epithelial progenitor cells that functionally contribute to expansive growth during puberty and differentiation during pregnancy. However, extensive anatomical differences between mouse and human mammary tissues preclude the direct translation of rodent findings to the human breast. Therefore, here we characterize the mammary progenitor cell hierarchy and identify the anatomic location of progenitor cells within human breast tissues. METHODS Mammary epithelial cells (MECs) were isolated from disease-free reduction mammoplasty tissues and assayed for stem/progenitor activity in vitro and in vivo. MECs were sorted and evaluated for growth on collagen and expression of lineages markers. Breast lobules were microdissected and individually characterized based on lineage markers and steroid receptor expression to identify the anatomic location of progenitor cells. Spanning-tree progression analysis of density-normalized events (SPADE) was used to identify the cellular hierarchy of MECs within lobules from high-dimensional cytometry data. RESULTS Integrating multiple assays for progenitor activity, we identified the presence of luminal alveolar and basal ductal progenitors. Further, we show that Type I lobules of the human breast were the least mature, demonstrating an unrestricted pattern of expression of luminal and basal lineage markers. Consistent with this, SPADE analysis revealed that immature lobules were enriched for basal progenitor cells, while mature lobules consisted of increased hierarchal complexity of cells within the luminal lineages. CONCLUSIONS These results reveal underlying differences in the human breast epithelial hierarchy and suggest that with increasing glandular maturity, the epithelial hierarchy also becomes more complex.
Collapse
|
138
|
ΔNp63 promotes stem cell activity in mammary gland development and basal-like breast cancer by enhancing Fzd7 expression and Wnt signalling. Nat Cell Biol 2014; 16:1004-15, 1-13. [PMID: 25241036 PMCID: PMC4183725 DOI: 10.1038/ncb3040] [Citation(s) in RCA: 164] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2014] [Accepted: 08/18/2014] [Indexed: 02/07/2023]
Abstract
Emerging evidence suggests that cancer is populated and maintained by tumor initiating cells (TICs) with stem-like properties similar to that of adult tissue stem cells. Despite recent advances, the molecular regulatory mechanisms that may be shared between normal and malignant stem cells remain poorly understood. Here we show that the ΔNp63 isoform of the Trp63 transcription factor promotes normal mammary stem cell (MaSC) activity by increasing the expression of the Wnt receptor Fzd7, thereby enhancing Wnt signaling. Importantly, Fzd7-dependent enhancement of Wnt signaling by ΔNp63 also governs tumor initiating activity of the basal subtype of breast cancer. These findings establish ΔNp63 as a key regulator of stem cells in both normal and malignant mammary tissues and provide direct evidence that breast cancer TICs and normal MaSCs share common regulatory mechanisms.
Collapse
|
139
|
Prpar Mihevc S, Ogorevc J, Dovc P. Lineage-specific markers of goat mammary cells in primary culture. In Vitro Cell Dev Biol Anim 2014; 50:926-36. [PMID: 25213688 DOI: 10.1007/s11626-014-9796-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 07/03/2014] [Indexed: 11/29/2022]
Abstract
The objective of this study was morphological and functional characterization of cells from the primary cell culture developed from lactating goat mammary gland, focusing on distribution of lineage-specific markers. Primary cells were grown on a thin layer of basement membrane matrix, a growth surface that resembles in vivo conditions. The cells in adherent conditions rapidly proliferated and showed cobblestone morphology, typical for epithelial cells. Under non-adherent conditions, goat mammary cells formed spherical, acini-like structures that resembled alveoli of lactating mammary gland. Immunofluorescence and RNA sequencing were employed to determine expression of lineage-specific markers. Presence of markers cytokeratin 14 and 18, integrin alpha 6, vimentin, estrogen receptor, smooth muscle actin, and cytokeratin 5 was detected using immunofluorescence. The greatest expression was observed for markers typical for myoepithelial cells, luminal cells, and mesenchymal cells. Based on our characterization, we can conclude that established primary culture was composed of mainly epithelial and stromal cells. These findings demonstrate that primary mammary cells express some of the most important functional and biochemical markers needed for their characterization. First, they grow in the characteristic cobblestone morphology of epithelial cells. Second, they express classical cytoplasmic network of cytokeratin fibers. Third, they express markers typical of mammary parenchyma and stroma. The established cell culture represents a good in vitro model for studies of mammary gland development, differentiation, and lactation. We suggest that herein revealed lineage markers are suitable for characterization of mammary cells of goat and possibly other mammalian species.
Collapse
Affiliation(s)
- Sonja Prpar Mihevc
- Department of Animal Science, Biotechnical Faculty, University of Ljubljana, Groblje 3, 1230, Domzale, Slovenia
| | | | | |
Collapse
|
140
|
Bovine mammary epithelial cells retain stem-like phenotype in long-term cultures. Res Vet Sci 2014; 97:367-75. [PMID: 25189469 DOI: 10.1016/j.rvsc.2014.07.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Revised: 04/19/2014] [Accepted: 07/20/2014] [Indexed: 12/31/2022]
Abstract
The detection and characterization of bovine mammary stem cells may give a better understanding of the cyclic characteristic of mammary gland development. In turn, this could potentially offer techniques to manipulate lactation yield and for regenerative medicine. We previously demonstrated that adult stem cells reside in the bovine mammary gland and possess an intrinsic regenerative potential. In vitro maintenance and expansion of this primitive population is a challenging task that could make easier the study of adult mammary stem cells. The aim of this study is to investigate this possibility. Different subpopulations of mammary epithelial cells emerge when they are cultured in two defined culture conditions. Specific cell differentiation markers as cytokeratin 18 (CK18) and cytokeratin 14 (CK14) were expressed with significant differences according to culture conditions. Vimentin, a well-known fibroblast marker was observed to increase significantly (P < 0.5) only after day 20. In both conditions, after prolonged culture (25 days) a subset of cells still retained regenerative capabilities. These cells were able to form organized pseudo-alveoli when transplanted in immunodeficient mice as shown by the expression of cytokeratin 14 (CK14), cytokeratin 18 (CK18), p63 (a mammary basal cell layer marker) and Epithelial Cell Adhesion Molecule (EpCAM). We also were able to observe the presence of milk proteins signal in these regenerated structures, which is a specific marker of functional mammary alveoli. Progenitor content was also analyzed in vitro through Colony-Forming Cell (CFC) assays with no substantial differences among culture conditions and time points. These results demonstrate that long-term culture of a multipotent cell subpopulation with intrinsic regenerative potential is possible.
Collapse
|
141
|
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: 3.9] [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
|
142
|
Kawamura E, Fielding AB, Kannan N, Balgi A, Eaves CJ, Roberge M, Dedhar S. Identification of novel small molecule inhibitors of centrosome clustering in cancer cells. Oncotarget 2014; 4:1763-76. [PMID: 24091544 PMCID: PMC3858562 DOI: 10.18632/oncotarget.1198] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Most normal cells have two centrosomes that form bipolar spindles during mitosis, while cancer cells often contain more than two, or "supernumerary" centrosomes. Such cancer cells achieve bipolar division by clustering their centrosomes into two functional poles, and inhibiting this process then leads to cancer-specific cell death. A major problem with clinically used anti-mitotic drugs, such as paclitaxel, is their toxicity in normal cells. To discover new compounds with greater specificity for cancer cells, we established a high-content screen for agents that block centrosome clustering in BT-549 cells, a breast cancer cell line that harbors supernumerary centrosomes. Using this screen, we identified 14 compounds that inhibit centrosome clustering and induce mitotic arrest. Some of these compounds were structurally similar, suggesting a common structural motif important for preventing centrosome clustering. We next compared the effects of these compounds on the growth of several breast and other cancer cell lines, an immortalized normal human mammary epithelial cell line, and progenitor-enriched primary normal human mammary epithelial cells. From these comparisons, we found some compounds that kill breast cancer cells, but not their normal epithelial counterparts, suggesting their potential for targeted therapy. One of these compounds, N2-(3-pyridylmethyl)-5-nitro-2-furamide (Centrosome Clustering Chemical Inhibitor-01, CCCI-01), that showed the greatest differential response in this screen was confirmed to have selective effects on cancer as compared to normal breast progenitors using more precise apoptosis induction and clonogenic growth endpoints. The concentration of CCCI-01 that killed cancer cells in the clonogenic assay spared normal human bone marrow hematopoietic progenitors in the colony-forming cell assay, indicating a potential therapeutic window for CCCI-01, whose selectivity might be further improved by optimizing the compound. Immunofluorescence analysis showed that treatment with CCCI-01 lead to multipolar spindles in BT-549, while maintaining bipolar spindles in the normal primary human mammary epithelial cells. Since centrosome clustering is a complex process involving multiple pathways, the 14 compounds identified in this study provide a potentially novel means to developing non-cross-resistant anti-cancer drugs that block centrosome clustering.
Collapse
Affiliation(s)
- Eiko Kawamura
- Department of Integrative Oncology, British Columbia Cancer Agency, Vancouver, BC, Canada
| | | | | | | | | | | | | |
Collapse
|
143
|
Abstract
Based on transplantation and lineage tracing studies, a hierarchy of stem and progenitor cells has been shown to exist among the mammary epithelium. In this review, Visvader and Stingl integrate recent data on the mammary stem cell differentiation hierarchy and its control at the transcriptional and epigenetic levels. They also discuss the relevance of the evolving hierarchy to the identification of “cells of origin” of breast cancer. The mammary epithelium is highly responsive to local and systemic signals, which orchestrate morphogenesis of the ductal tree during puberty and pregnancy. Based on transplantation and lineage tracing studies, a hierarchy of stem and progenitor cells has been shown to exist among the mammary epithelium. Lineage tracing has highlighted the existence of bipotent mammary stem cells (MaSCs) in situ as well as long-lived unipotent cells that drive morphogenesis and homeostasis of the ductal tree. Moreover, there is accumulating evidence for a heterogeneous MaSC compartment comprising fetal MaSCs, slow-cycling cells, and both long-term and short-term repopulating cells. In parallel, diverse luminal progenitor subtypes have been identified in mouse and human mammary tissue. Elucidation of the normal cellular hierarchy is an important step toward understanding the “cells of origin” and molecular perturbations that drive breast cancer.
Collapse
Affiliation(s)
- 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, University of Melbourne, Parkville VIC 3010, Australia
| | - John Stingl
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge CB2 0RE, United Kingdom
| |
Collapse
|
144
|
Choudhary RK. Mammary stem cells: expansion and animal productivity. J Anim Sci Biotechnol 2014; 5:36. [PMID: 25057352 PMCID: PMC4107933 DOI: 10.1186/2049-1891-5-36] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2014] [Accepted: 07/04/2014] [Indexed: 12/12/2022] Open
Abstract
Identification and characterization of mammary stem cells and progenitor cells from dairy animals is important in the understanding of mammogenesis, tissue turnover, lactation persistency and regenerative therapy. It has been realized by many investigators that altered lactation, long dry periods (non-milking period between two consecutive lactation cycles), abrupt cessation of lactation (common in water buffaloes) and disease conditions like mastitis, greatly reduce milk yield thus render huge financial losses within the dairy sector. Cellular manipulation of specialized cell types within the mammary gland, called mammary stem cells (MaSCs)/progenitor cells, might provide potential solutions to these problems and may improve milk production. In addition, MaSCs/progenitor cells could be used in regenerative therapy against tissue damage caused by mastitis. This review discusses methods of MaSC/progenitor cell manipulation and their mechanisms in bovine and caprine animals. Author believes that intervention of MaSCs/progenitor cells could lessen the huge financial losses to the dairy industry globally.
Collapse
Affiliation(s)
- Ratan K Choudhary
- School of Animal Biotechnology, Guru Angad Dev Veterinary and Animal Science University, Ludhiana, Punjab 141004, India
| |
Collapse
|
145
|
Abstract
The very limited ability of adult podocytes to proliferate in vivo is clinically significant because podocytes form a vascular barrier that is functionally critical to the nephron, podocyte hypoplasia is a characteristic of disease, and inadequate regeneration of podocytes is a major cause of persistent podocyte hypoplasia. Excessive podocyte loss or inadequate replacement leads to glomerulosclerosis in many progressive kidney diseases. Thus, restoration of podocyte cell density almost certainly is reliant on regeneration by podocyte progenitors. However, such putative progenitors have remained elusive until recently. In this review, we describe the developmental processes leading to podocyte and parietal epithelial cell (PEC) formation during glomerulogenesis. We compare evidence that in normal human kidneys PECs expressing progenitor markers CD133 and CD24 can differentiate into podocytes in vitro and in vivo, with evidence from animal models suggesting a more limited role of the PEC's capacity to serve as a podocyte progenitor in adults. We highlight tantalizing new evidence that specialized vascular wall cells of afferent arterioles, including those that produce renin in healthy kidney, provide a novel local progenitor source of new PECs and podocytes in response to podocyte hypoplasia in the adult, and draw comparisons with glomerulogenesis.
Collapse
Affiliation(s)
- Stuart J Shankland
- Division of Nephrology, Department of Medicine, University of Washington School of Medicine, Seattle, WA.
| | - Jeffrey W Pippin
- Division of Nephrology, Department of Medicine, University of Washington School of Medicine, Seattle, WA
| | | |
Collapse
|
146
|
Honeth G, Lombardi S, Ginestier C, Hur M, Marlow R, Buchupalli B, Shinomiya I, Gazinska P, Bombelli S, Ramalingam V, Purushotham AD, Pinder SE, Dontu G. Aldehyde dehydrogenase and estrogen receptor define a hierarchy of cellular differentiation in the normal human mammary epithelium. Breast Cancer Res 2014; 16:R52. [PMID: 24887554 PMCID: PMC4095680 DOI: 10.1186/bcr3663] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Accepted: 04/29/2014] [Indexed: 12/15/2022] Open
Abstract
INTRODUCTION Although estrogen and progesterone play a key role in normal mammary development and in breast cancer, the potential for proliferation and lineage differentiation as well as origin of cells that express the estrogen receptor (ER) in normal breast epithelium are not known. Some evidence suggests that normal human mammary stem/progenitor cells are ER-, but the identity of these cells and the cellular hierarchy of breast epithelium are still subjects of controversy. It is likely that elucidation of these aspects will bring insight into the cellular origin of breast cancer subtypes. METHODS We used fluorescence-activated cell sorting of primary human mammary epithelial cells along with in vitro and in vivo functional assays to examine the hierarchic relation between cells with aldehyde dehydrogenase enzymatic activity (ALDH+ cells) and ER+ cells in the normal human breast epithelium. We assessed the proliferation and lineage differentiation potential of these cells in vitro and in vivo. A gene reporter assay was used to separate live ER+ and ER- mammary epithelial cells. With shRNA-mediated knockdown, we investigated the role of ALDH isoforms in the functionality of mammary epithelial progenitor cells. RESULTS We describe a cellular hierarchy in the normal human mammary gland in which ER-/ALDH+ cells with functional properties of stem/progenitor cells generate ER+ progenitor cells, which in turn give rise to cells of luminal lineage. We show that the ALDH1A1 isoform, through its function in the retinoic acid metabolism, affects the proliferation and/or early differentiation of stem/progenitor cells and is important for branching morphogenesis. CONCLUSIONS This study presents direct evidence that ER+ cells are generated by ER-/ALDH+ stem/progenitor cells. We also show that ER+ cells are able to generate cell progeny of luminal lineage in vitro and in vivo. Loss of ALDH1A1 function impairs this process, as well as branching morphogenesis and clonogenicity in suspension culture. This latter effect is reversed by treatment with retinoic acid.
Collapse
|
147
|
Glutathione-dependent and -independent oxidative stress-control mechanisms distinguish normal human mammary epithelial cell subsets. Proc Natl Acad Sci U S A 2014; 111:7789-94. [PMID: 24821780 DOI: 10.1073/pnas.1403813111] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Mechanisms that control the levels and activities of reactive oxygen species (ROS) in normal human mammary cells are poorly understood. We show that purified normal human basal mammary epithelial cells maintain low levels of ROS primarily by a glutathione-dependent but inefficient antioxidant mechanism that uses mitochondrial glutathione peroxidase 2. In contrast, the matching purified luminal progenitor cells contain higher levels of ROS, multiple glutathione-independent antioxidants and oxidative nucleotide damage-controlling proteins and consume O2 at a higher rate. The luminal progenitor cells are more resistant to glutathione depletion than the basal cells, including those with in vivo and in vitro proliferation and differentiation activity. The luminal progenitors also are more resistant to H2O2 or ionizing radiation. Importantly, even freshly isolated "steady-state" normal luminal progenitors show elevated levels of unrepaired oxidative DNA damage. Distinct ROS control mechanisms operating in different subsets of normal human mammary cells could have differentiation state-specific functions and long-term consequences.
Collapse
|
148
|
Phillips S, Prat A, Sedic M, Proia T, Wronski A, Mazumdar S, Skibinski A, Shirley SH, Perou CM, Gill G, Gupta PB, Kuperwasser C. Cell-state transitions regulated by SLUG are critical for tissue regeneration and tumor initiation. Stem Cell Reports 2014; 2:633-47. [PMID: 24936451 PMCID: PMC4050485 DOI: 10.1016/j.stemcr.2014.03.008] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Revised: 03/25/2014] [Accepted: 03/26/2014] [Indexed: 12/17/2022] Open
Abstract
Perturbations in stem cell activity and differentiation can lead to developmental defects and cancer. We use an approach involving a quantitative model of cell-state transitions in vitro to gain insights into how SLUG/SNAI2, a key developmental transcription factor, modulates mammary epithelial stem cell activity and differentiation in vivo. In the absence of SLUG, stem cells fail to transition into basal progenitor cells, while existing basal progenitor cells undergo luminal differentiation; together, these changes result in abnormal mammary architecture and defects in tissue function. Furthermore, we show that in the absence of SLUG, mammary stem cell activity necessary for tissue regeneration and cancer initiation is lost. Mechanistically, SLUG regulates differentiation and cellular plasticity by recruiting the chromatin modifier lysine-specific demethylase 1 (LSD1) to promoters of lineage-specific genes to repress transcription. Together, these results demonstrate that SLUG plays a dual role in repressing luminal epithelial differentiation while unlocking stem cell transitions necessary for tumorigenesis. SLUG promotes stem cell-state transitions necessary for tumorigenesis Loss of SLUG in mice leads to luminal differentiation of basal/myoepithelial cells In vitro quantitative modeling can accurately predict epithelial plasticity in vivo A SLUG/LSD1 complex epigenetically regulates mammary epithelial cell differentiation
Collapse
Affiliation(s)
- Sarah Phillips
- Department of Developmental, Molecular and Chemical Biology, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, 136 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 (VHIO), Barcelona 08035, Spain
| | - Maja Sedic
- Department of Developmental, Molecular and Chemical Biology, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA ; Molecular Oncology Research Institute, Tufts Medical Center, 800 Washington Street, Boston, MA 02111, USA
| | - Theresa Proia
- Whitehead Institute for Biomedical Research, 9 Cambridge Center, Cambridge MA, 02142, USA
| | - Ania Wronski
- Department of Developmental, Molecular and Chemical Biology, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA ; Molecular Oncology Research Institute, Tufts Medical Center, 800 Washington Street, Boston, MA 02111, USA
| | - Sohini Mazumdar
- Department of Developmental, Molecular and Chemical Biology, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA
| | - Adam Skibinski
- Department of Developmental, Molecular and Chemical Biology, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA ; Molecular Oncology Research Institute, Tufts Medical Center, 800 Washington Street, Boston, MA 02111, USA
| | - Stephanie H Shirley
- Department of Molecular Carcinogenesis, Science Park - Research Division, The University of Texas MD Anderson Cancer Center, Smithville, TX 78957, USA
| | - Charles M Perou
- Department of Genetics and Pathology, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Grace Gill
- Department of Developmental, Molecular and Chemical Biology, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA
| | - Piyush B Gupta
- Whitehead Institute for Biomedical Research, 9 Cambridge Center, Cambridge MA, 02142, USA ; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Charlotte Kuperwasser
- Department of Developmental, Molecular and Chemical Biology, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA ; Molecular Oncology Research Institute, Tufts Medical Center, 800 Washington Street, Boston, MA 02111, USA
| |
Collapse
|
149
|
Comparison of stem/progenitor cell number and transcriptomic profile in the mammary tissue of dairy and beef breed heifers. J Appl Genet 2014; 55:383-95. [PMID: 24748329 PMCID: PMC4102771 DOI: 10.1007/s13353-014-0213-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Revised: 03/26/2014] [Accepted: 03/28/2014] [Indexed: 12/25/2022]
Abstract
Bovine mammary stem cells (MaSC) are a source of ductal and lobulo-alveolar tissue during the development of the mammary gland and its remodeling in repeating lactation cycles. We hypothesize that the number of MaSC, their molecular properties, and interactions with their niche may be essential in order to determine the mammogenic potential in heifers. To verify this hypothesis, we compared the number of MaSC and the transcriptomic profile in the mammary tissue of 20-month-old, non-pregnant dairy (Holstein-Friesian, HF) and beef (Limousin, LM) heifers. For the identification and quantification of putative stem/progenitor cells in mammary tissue sections, scanning cytometry was used with a combination of MaSC molecular markers: stem cell antigen-1 (Sca-1) and fibronectin type III domain containing 3B (FNDC3B) protein. Cytometric analysis revealed a significantly higher number of Sca-1posFNDC3Bpos cells in HF (2.94 ± 0.35 %) than in LM (1.72 ± 0.20 %) heifers. In HF heifers, a higher expression of intramammary hormones, growth factors, cytokines, chemokines, and transcription regulators was observed. The model of mammary microenvironment favorable for MaSC was associated with the regulation of genes involved in MaSC maintenance, self-renewal, proliferation, migration, differentiation, mammary tissue remodeling, angiogenesis, regulation of adipocyte differentiation, lipid metabolism, and steroid and insulin signaling. In conclusion, the mammogenic potential in postpubertal dairy heifers is facilitated by a higher number of MaSC and up-regulation of mammary auto- and paracrine factors representing the MaSC niche.
Collapse
|
150
|
Nguyen L, Makarem M, Carles A, Moksa M, Kannan N, Pandoh P, Eirew P, Osako T, Kardel M, Cheung A, Kennedy W, Tse K, Zeng T, Zhao Y, Humphries R, Aparicio S, Eaves C, Hirst M. Clonal Analysis via Barcoding Reveals Diverse Growth and Differentiation of Transplanted Mouse and Human Mammary Stem Cells. Cell Stem Cell 2014; 14:253-63. [DOI: 10.1016/j.stem.2013.12.011] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 10/08/2013] [Accepted: 12/16/2013] [Indexed: 10/25/2022]
|