101
|
Johnston RL, Wockner L, McCart Reed AE, Wiegmans A, Chenevix-Trench G, Khanna KK, Lakhani SR, Smart CE. High content screening application for cell-type specific behaviour in heterogeneous primary breast epithelial subpopulations. Breast Cancer Res 2016; 18:18. [PMID: 26861772 PMCID: PMC4748588 DOI: 10.1186/s13058-016-0681-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 01/29/2016] [Indexed: 12/26/2022] Open
Abstract
Background The complex interaction between multiple cell types and the microenvironment underlies the diverse pathways to carcinogenesis and necessitates sophisticated approaches to in vitro hypotheses testing. The combination of mixed culture format with high content immunofluorescence screening technology provides a powerful platform for observation of cell type specific behavior. Methods We have developed a versatile, high-throughput method for assessing cell-type specific responses. In addition to the specificity and sensitivity offered traditionally by immunofluorescent detection in flow cytometry, the ‘in-cell’ analysis method we describe provides the added benefits of higher throughput and the ability to analyse protein subcellular localisation in situ. Furthermore, elimination of the cell dissociation step allows for more immediate analysis of responses to specific extrinsic stimuli. We applied this method to investigate ionising radiation treatment response in normal breast epithelial cells, measuring growth rate, cell cycle response and double-strand DNA breaks. Results The ‘in-cell’ analysis approach elucidated several interesting donor and cell-type specific differences. Notably, in response to ionizing radiation we observed differential expression in luminal and basal-like cells of a member of the APOBEC enzyme family, recently identified as a critical driver of an oncogenic signature. Our findings suggest that this enzyme is active in the normal breast epithelium during DNA damage response. Conclusions We demonstrate the practical application of a new method for assessing cell-type specific change in mixed cultures, especially the analysis of normal primary cultures, overcoming a major technical issue of dissecting the response of multiple cell types in a heterogeneous population. Electronic supplementary material The online version of this article (doi:10.1186/s13058-016-0681-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Rebecca L Johnston
- The University of Queensland, UQ Centre for Clinical Research, Brisbane, Queensland, 4029, Australia. .,QIMR Berghofer Medical Research Institute, Brisbane, Queensland, 4029, Australia.
| | - Leesa Wockner
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, 4029, Australia.
| | - Amy E McCart Reed
- The University of Queensland, UQ Centre for Clinical Research, Brisbane, Queensland, 4029, Australia. .,QIMR Berghofer Medical Research Institute, Brisbane, Queensland, 4029, Australia.
| | - Adrian Wiegmans
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, 4029, Australia.
| | | | - Kum Kum Khanna
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, 4029, Australia.
| | - Sunil R Lakhani
- The University of Queensland, UQ Centre for Clinical Research, Brisbane, Queensland, 4029, Australia. .,Pathology Queensland, Royal Brisbane and Women's Hospital, Brisbane, Queensland, 4029, Australia. .,The University of Queensland, School of Medicine, Brisbane, Queensland, 4029, Australia.
| | - Chanel E Smart
- The University of Queensland, UQ Centre for Clinical Research, Brisbane, Queensland, 4029, Australia. .,QIMR Berghofer Medical Research Institute, Brisbane, Queensland, 4029, Australia.
| |
Collapse
|
102
|
Fate Mapping Mammalian Corneal Epithelia. Ocul Surf 2016; 14:82-99. [PMID: 26774909 DOI: 10.1016/j.jtos.2015.11.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 11/16/2015] [Accepted: 11/18/2015] [Indexed: 02/07/2023]
Abstract
The anterior aspect of the cornea consists of a stratified squamous epithelium, thought to be maintained by a rare population of stem cells (SCs) that reside in the limbal transition zone. Although migration of cells that replenish the corneal epithelium has been studied for over a century, the process is still poorly understood and not well characterized. Numerous techniques have been employed to examine corneal epithelial dynamics, including visualization by light microscopy, the incorporation of vital dyes and DNA labels, and transplantation of genetically marked cells that have acted as cell and lineage beacons. Modern-day lineage tracing utilizes molecular methods to determine the fate of a specific cell and its progeny over time. Classically employed in developmental biology, lineage tracing has been used more recently to track the progeny of adult SCs in a number of organs to pin-point their location and understand their movement and influence on tissue regeneration. This review highlights key discoveries that have led researchers to develop cutting-edge genetic tools to effectively and more accurately monitor turnover and displacement of cells within the mammalian corneal epithelium. Collating information on the basic biology of SCs will have clinical ramifications in furthering our knowledge of the processes that govern their role in homeostasis, wound-healing, transplantation, and how we can improve current unsatisfactory SC-based therapies for patients suffering blinding corneal disease.
Collapse
|
103
|
Nair NU, Hunter L, Shao M, Grnarova P, Lin Y, Bucher P, E Moret BM. A maximum-likelihood approach for building cell-type trees by lifting. BMC Genomics 2016; 17 Suppl 1:14. [PMID: 26819094 PMCID: PMC4895258 DOI: 10.1186/s12864-015-2297-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In cell differentiation, a less specialized cell differentiates into a more specialized one, even though all cells in one organism have (almost) the same genome. Epigenetic factors such as histone modifications are known to play a significant role in cell differentiation. We previously introduce cell-type trees to represent the differentiation of cells into more specialized types, a representation that partakes of both ontogeny and phylogeny. RESULTS We propose a maximum-likelihood (ML) approach to build cell-type trees and show that this ML approach outperforms our earlier distance-based and parsimony-based approaches. We then study the reconstruction of ancestral cell types; since both ancestral and derived cell types can coexist in adult organisms, we propose a lifting algorithm to infer internal nodes. We present results on our lifting algorithm obtained both through simulations and on real datasets. CONCLUSIONS We show that our ML-based approach outperforms previously proposed techniques such as distance-based and parsimony-based methods. We show our lifting-based approach works well on both simulated and real data.
Collapse
Affiliation(s)
- Nishanth Ulhas Nair
- School of Computer and Communication Sciences, École Polytechnique Fédérale de Lausanne (EPFL), EPFL IC IIF LCBB, INJ 211 (Batiment INJ), Station 14, Lausanne, CH-1015, Switzerland.
| | - Laura Hunter
- Computer Science Department, Stanford University, Stanford, USA.
| | - Mingfu Shao
- School of Computer and Communication Sciences, École Polytechnique Fédérale de Lausanne (EPFL), EPFL IC IIF LCBB, INJ 211 (Batiment INJ), Station 14, Lausanne, CH-1015, Switzerland.
| | - Paulina Grnarova
- School of Computer and Communication Sciences, École Polytechnique Fédérale de Lausanne (EPFL), EPFL IC IIF LCBB, INJ 211 (Batiment INJ), Station 14, Lausanne, CH-1015, Switzerland.
| | - Yu Lin
- Department of Computer Science and Engineering, University of California, San Diego, San Diego, USA.
| | - Philipp Bucher
- School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
- Swiss Institute of Bioinformatics, Lausanne, Switzerland.
| | - Bernard M E Moret
- School of Computer and Communication Sciences, École Polytechnique Fédérale de Lausanne (EPFL), EPFL IC IIF LCBB, INJ 211 (Batiment INJ), Station 14, Lausanne, CH-1015, Switzerland.
- Swiss Institute of Bioinformatics, Lausanne, Switzerland.
| |
Collapse
|
104
|
Arévalo Turrubiarte M, Perruchot MH, Finot L, Mayeur F, Dessauge F. Phenotypic and functional characterization of two bovine mammary epithelial cell lines in 2D and 3D models. Am J Physiol Cell Physiol 2015; 310:C348-56. [PMID: 26659725 DOI: 10.1152/ajpcell.00261.2015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 11/30/2015] [Indexed: 02/04/2023]
Abstract
Immortalized bovine mammary epithelial cells (BME-UV1) and immortalized bovine mammary alveolar cells (MAC-T) have been extensively used as in vitro cell models to understand milk production in dairy cows. Precise knowledge about their phenotype and performance remains, however, unknown. This study aims to characterize MAC-T and BME-UV1 profiles when cultured in two-dimensional adherent, three-dimensional adherent (Matrigel), and three-dimensional no adherent [ultralow attachment (ULA)] supports. MAC-T and BME-UV1 were compared according to their proliferation capacities and to specific cell surface markers CD24, CD326 [epithelial cell adhesion molecule (EpCAM)], CD10, and integrin CD49f (α-6). Cytokeratin (CK14 and CK19), signal transducer and activator of transcription 5, and other proteins (occludin and cadherin-1) were analyzed. BME-UV1 in ULA support expressed higher CD49f marker. A different intensity of CD49 staining allowed the discrimination between the two cell lines in adherent condition. CD10, EpCAM, and CK19 expressions show that BME-UV1 cells have luminal capacity, while MAC-T has a myoepithelial profile with a high expression of CK14. BME-UV1 cells possess a closer committed progenitor profile due to their higher expression in aldehyde dehydrogenase and EpCAM. We observed that BME-UV1 cells have a better capacity to form spherical structures, mammospheres, in Matrigel than MAC-T, which was confirmed by the higher mammosphere area. In the ULA condition, BME-UV1 proliferated over the 6 days of culture. Taken together, our results clearly confirm the BME-UV1 luminal profile and MAC-T ductal/myoepithelial-like phenotype.
Collapse
Affiliation(s)
- Magdalena Arévalo Turrubiarte
- Institut National de la Recherche Agronomique, UMR1348 Pegase, Saint-Gilles, France; and Agrocampus Ouest, UMR1348 Pegase, Rennes, France
| | - Marie-Hélène Perruchot
- Institut National de la Recherche Agronomique, UMR1348 Pegase, Saint-Gilles, France; and Agrocampus Ouest, UMR1348 Pegase, Rennes, France
| | - Laurence Finot
- Institut National de la Recherche Agronomique, UMR1348 Pegase, Saint-Gilles, France; and Agrocampus Ouest, UMR1348 Pegase, Rennes, France
| | - Frédérique Mayeur
- Institut National de la Recherche Agronomique, UMR1348 Pegase, Saint-Gilles, France; and Agrocampus Ouest, UMR1348 Pegase, Rennes, France
| | - Frédéric Dessauge
- Institut National de la Recherche Agronomique, UMR1348 Pegase, Saint-Gilles, France; and Agrocampus Ouest, UMR1348 Pegase, Rennes, France
| |
Collapse
|
105
|
Fridriksdottir AJ, Kim J, Villadsen R, Klitgaard MC, Hopkinson BM, Petersen OW, Rønnov-Jessen L. Propagation of oestrogen receptor-positive and oestrogen-responsive normal human breast cells in culture. Nat Commun 2015; 6:8786. [PMID: 26564780 PMCID: PMC4660059 DOI: 10.1038/ncomms9786] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 10/04/2015] [Indexed: 02/07/2023] Open
Abstract
Investigating the susceptibility of oestrogen receptor-positive (ERpos) normal human breast epithelial cells (HBECs) for clinical purposes or basic research awaits a proficient cell-based assay. Here we set out to identify markers for isolating ERpos cells and to expand what appear to be post-mitotic primary cells into exponentially growing cultures. We report a robust technique for isolating ERpos HBECs from reduction mammoplasties by FACS using two cell surface markers, CD166 and CD117, and an intracellular cytokeratin marker, Ks20.8, for further tracking single cells in culture. We show that ERpos HBECs are released from growth restraint by small molecule inhibitors of TGFβ signalling, and that growth is augmented further in response to oestrogen. Importantly, ER signalling is functionally active in ERpos cells in extended culture. These findings open a new avenue of experimentation with normal ERpos HBECs and provide a basis for understanding the evolution of human breast cancer. Culturing normal primary breast cells that express the oestrogen receptor is difficult. Here, the authors isolate oestrogen receptor positive normal breast cells using the cell surface markers CD166 and CD117, and show that the cultures can be repeatedly passaged and retain oestrogen receptor protein expression.
Collapse
Affiliation(s)
- Agla J Fridriksdottir
- Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark.,Danish Stem Cell Centre, Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Jiyoung Kim
- Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark.,Danish Stem Cell Centre, Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - René Villadsen
- Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark.,Danish Stem Cell Centre, Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Marie Christine Klitgaard
- Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark.,Danish Stem Cell Centre, Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark.,Department of Biology, University of Copenhagen, DK-2100 Copenhagen Ø, Denmark
| | - Branden M Hopkinson
- Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark.,Danish Stem Cell Centre, Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Ole William Petersen
- Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark.,Danish Stem Cell Centre, Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Lone Rønnov-Jessen
- Department of Biology, University of Copenhagen, DK-2100 Copenhagen Ø, Denmark
| |
Collapse
|
106
|
Lin H, Sun G, He H, Botsford B, Li M, Elisseeff JH, Yiu SC. Three-Dimensional Culture of Functional Adult Rabbit Lacrimal Gland Epithelial Cells on Decellularized Scaffold. Tissue Eng Part A 2015; 22:65-74. [PMID: 26414959 DOI: 10.1089/ten.tea.2015.0286] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Aqueous tear-deficient dry eye disease is a multifactorial chronic disorder, in which the lacrimal gland fails to produce enough tears to maintain a healthy ocular surface. Some severe cases may develop corneal damage and significant vision loss. Treatment primarily involves palliation using ocular surface lubricants, but can only provide temporary relief. Construction of a bioengineered lacrimal gland having functional secretory epithelial cells is a potentially promising option for providing long-term relief to severe dry eye patients. Using sphere-forming culture techniques, we cultured adult rabbit lacrimal gland progenitor cells and prepared a lacrimal gland scaffold by decellularization. When progenitor cells were seeded onto the decellularized scaffold, they formed duct- and acinar-like structures in the three-dimensional culture system. Lacrimal gland epithelial cells showed good cell viability, cell differentiation, and secretory function in decellularized lacrimal gland matrix, as indicated by morphology, immunostaining, and β-hexosaminidase secretion assay. This study demonstrated the potential suitability of utilizing tissue-specific progenitor cells and a tissue-derived bioscaffold for lacrimal gland restoration.
Collapse
Affiliation(s)
- Hui Lin
- 1 Department of Ophthalmology, Wilmer Eye Institute, School of Medicine, Johns Hopkins University , Baltimore, Maryland
| | - Guoying Sun
- 1 Department of Ophthalmology, Wilmer Eye Institute, School of Medicine, Johns Hopkins University , Baltimore, Maryland
| | - Hong He
- 1 Department of Ophthalmology, Wilmer Eye Institute, School of Medicine, Johns Hopkins University , Baltimore, Maryland
| | | | - Mackenzie Li
- 3 University of British Columbia , Vancouver, British Columbia, Canada
| | - Jennifer H Elisseeff
- 1 Department of Ophthalmology, Wilmer Eye Institute, School of Medicine, Johns Hopkins University , Baltimore, Maryland.,4 Translational Tissue Engineering Center, Johns Hopkins University , Baltimore, Maryland
| | - Samuel C Yiu
- 1 Department of Ophthalmology, Wilmer Eye Institute, School of Medicine, Johns Hopkins University , Baltimore, Maryland
| |
Collapse
|
107
|
Voutsadakis IA. The network of pluripotency, epithelial-mesenchymal transition, and prognosis of breast cancer. BREAST CANCER-TARGETS AND THERAPY 2015; 7:303-19. [PMID: 26379447 PMCID: PMC4567227 DOI: 10.2147/bctt.s71163] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Breast cancer is the leading female cancer in terms of prevalence. Progress in molecular biology has brought forward a better understanding of its pathogenesis that has led to better prognostication and treatment. Subtypes of breast cancer have been identified at the genomic level and guide therapeutic decisions based on their biology and the expected benefit from various interventions. Despite this progress, a significant percentage of patients die from their disease and further improvements are needed. The cancer stem cell theory and the epithelial-mesenchymal transition are two comparatively novel concepts that have been introduced in the area of cancer research and are actively investigated. Both processes have their physiologic roots in normal development and common mediators have begun to surface. This review discusses the associations of these networks as a prognostic framework in breast cancer.
Collapse
Affiliation(s)
- Ioannis A Voutsadakis
- Division of Medical Oncology, Department of Internal Medicine, Sault Area Hospital, Sault Ste Marie, ON, Canada ; Division of Clinical Sciences, Northern Ontario School of Medicine, Sudbury, ON, Canada
| |
Collapse
|
108
|
Gordon N, Skinner AM, Pommier RF, Schillace RV, O'Neill S, Peckham JL, Muller P, Condron ME, Donovan C, Naik A, Hansen J, Pommier SJ. Gene expression signatures of breast cancer stem and progenitor cells do not exhibit features of Warburg metabolism. Stem Cell Res Ther 2015; 6:157. [PMID: 26316122 PMCID: PMC4552365 DOI: 10.1186/s13287-015-0153-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 06/05/2015] [Accepted: 08/11/2015] [Indexed: 11/10/2022] Open
Abstract
INTRODUCTION Cancers are believed to adapt to continual changes in glucose and oxygen availability by relying almost exclusively on glycolytic metabolism for energy (i.e. the Warburg effect). The process by which breast cancers sustain growth in avascular tissue is thought to be mediated via aberrant hypoxia response with ensuing shifts in glycolytic metabolism. Given their role in initiating and perpetuating tumors, we sought to determine whether breast cancer stem and progenitor cells play an instrumental role in this adaptive metabolic response. METHODS Breast cancer stem/progenitor cells were isolated from invasive ductal carcinomas, and benign stem cells (SC) were isolated from reduction mammoplasty tissues. Relative expression of 33 genes involved in hypoxia and glucose metabolism was evaluated in flow cytometrically isolated stem and progenitor cell populations. Significance between cohorts and cell populations was determined using Student's 2-tailed t test. RESULTS While benign stem/progenitor cells exhibited few significant inter-group differences in expression of genes involved in hypoxia regulation or glucose metabolism, breast cancer stem/progenitor cells demonstrated significant inter-group variability. Breast cancer stem/progenitor cells adapted to microenvironments through changes in stem cell numbers and transcription of glycolytic genes. One of four breast cancer stem/progenitor cells subpopulations exhibited an aerobic glycolysis gene expression signature. This subpopulation comprises the majority of the tumor and therefore best reflects invasive ductal carcinoma tumor biology. Although PI3K/AKT mutations are associated with increased proliferation of breast cancer cells, mutations in breast cancer stem/progenitor cells subpopulations did not correlate with changes in metabolic gene expression. CONCLUSIONS The adaptive capacity of breast cancer stem/progenitor cells may enable tumors to survive variable conditions encountered during progressive stages of cancer growth.
Collapse
Affiliation(s)
- Nicole Gordon
- Department of Surgery, Division of Surgical Oncology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Mail Code L619, Portland, OR, 97239, USA.
| | - Amy M Skinner
- Department of Surgery, Division of Surgical Oncology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Mail Code L619, Portland, OR, 97239, USA.
| | - Rodney F Pommier
- Department of Surgery, Division of Surgical Oncology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Mail Code L619, Portland, OR, 97239, USA.
| | - Robynn V Schillace
- Department of Surgery, Division of Surgical Oncology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Mail Code L619, Portland, OR, 97239, USA.
| | - Steven O'Neill
- Department of Surgery, Division of Surgical Oncology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Mail Code L619, Portland, OR, 97239, USA.
| | - Jennifer L Peckham
- Department of Surgery, Division of Surgical Oncology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Mail Code L619, Portland, OR, 97239, USA.
| | - Patrick Muller
- Department of Surgery, Division of Surgical Oncology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Mail Code L619, Portland, OR, 97239, USA.
| | - Mary E Condron
- Department of Surgery, Division of Surgical Oncology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Mail Code L619, Portland, OR, 97239, USA.
| | - Cory Donovan
- Department of Surgery, Division of Surgical Oncology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Mail Code L619, Portland, OR, 97239, USA.
| | - Arpana Naik
- Department of Surgery, Division of Surgical Oncology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Mail Code L619, Portland, OR, 97239, USA.
| | - Juliana Hansen
- Department of Surgery, Division of Plastic & Reconstructive Surgery, Oregon Health & Science University, Portland, OR, USA.
| | - SuEllen J Pommier
- Department of Surgery, Division of Surgical Oncology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Mail Code L619, Portland, OR, 97239, USA.
| |
Collapse
|
109
|
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
|
110
|
Finlay-Schultz J, Sartorius CA. Steroid hormones, steroid receptors, and breast cancer stem cells. J Mammary Gland Biol Neoplasia 2015; 20:39-50. [PMID: 26265122 PMCID: PMC4666507 DOI: 10.1007/s10911-015-9340-5] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 07/31/2015] [Indexed: 12/14/2022] Open
Abstract
The ovarian hormones progesterone and estrogen play important roles in breast cancer etiology, proliferation, and treatment. Androgens may also contribute to breast cancer risk and progression. In recent years, significant advances have been made in defining the roles of these steroid hormones in stem cell homeostasis in the breast. Stem cells are potential origins of breast cancer and may dictate tumor phenotype. At least a portion of breast cancers are proposed to be driven by cancer stem cells (CSCs), cells that mimic the self-renewing and repopulating properties of normal stem cells, and can confer drug resistance. Progesterone has been identified as the critical hormone regulating normal murine mammary stem cell (MaSC) populations and normal human breast stem cells. Synthetic progestins increase human breast cancer risk; one theory speculates that this occurs through increased stem cells. Progesterone treatment also increases breast CSCs in established breast cancer cell lines. This is mediated in part through progesterone regulation of transcription factors, signal transduction pathways, and microRNAs. There is also emerging evidence that estrogens and androgens can regulate breast CSC numbers. The evolving concept that a breast CSC phenotype is dynamic and can be influenced by cell signaling and external cues emphasizes that steroid hormones could be crucial players in controlling CSC number and function. Here we review recent studies on steroid hormone regulation of breast CSCs, and discuss mechanisms by which this occurs.
Collapse
Affiliation(s)
- Jessica Finlay-Schultz
- Department of Pathology, University of Colorado Anschutz Medical Campus, 12801 East 17th Avenue; MS 8104, Aurora, CO, 80045, USA.
| | - Carol A Sartorius
- Department of Pathology, University of Colorado Anschutz Medical Campus, 12801 East 17th Avenue; MS 8104, Aurora, CO, 80045, USA
| |
Collapse
|
111
|
Dontu G, Ince TA. Of mice and women: a comparative tissue biology perspective of breast stem cells and differentiation. J Mammary Gland Biol Neoplasia 2015; 20:51-62. [PMID: 26286174 PMCID: PMC4595531 DOI: 10.1007/s10911-015-9341-4] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 08/10/2015] [Indexed: 12/12/2022] Open
Abstract
Tissue based research requires a background in human and veterinary pathology, developmental biology, anatomy, as well as molecular and cellular biology. This type of comparative tissue biology (CTB) expertise is necessary to tackle some of the conceptual challenges in human breast stem cell research. It is our opinion that the scarcity of CTB expertise contributed to some erroneous interpretations in tissue based research, some of which are reviewed here in the context of breast stem cells. In this article we examine the dissimilarities between mouse and human mammary tissue and suggest how these may impact stem cell studies. In addition, we consider the differences between breast ducts vs. lobules and clarify how these affect the interpretation of results in stem cell research. Lastly, we introduce a new elaboration of normal epithelial cell types in human breast and discuss how this provides a clinically useful basis for breast cancer classification.
Collapse
Affiliation(s)
- Gabriela Dontu
- Stem Cell Group, Breakthrough Breast Cancer Research Unit, Research Oncology, King's College London School of Medicine, 3rd Floor Bermondsey Wing, Guy's Hospital, London, SE1 9RT, UK
| | - Tan A Ince
- Sylvester Comprehensive Cancer Center, Braman Family Breast Cancer Institute, Interdisciplinary Stem Cell Institute and Department of Pathology, University of Miami Miller School of Medicine, 1501 NW 10th Ave., Miami, 33136, FL, USA.
| |
Collapse
|
112
|
Arendt LM, Kuperwasser C. Form and function: how estrogen and progesterone regulate the mammary epithelial hierarchy. J Mammary Gland Biol Neoplasia 2015; 20:9-25. [PMID: 26188694 PMCID: PMC4596764 DOI: 10.1007/s10911-015-9337-0] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 07/08/2015] [Indexed: 12/30/2022] Open
Abstract
The mammary gland undergoes dramatic post-natal growth beginning at puberty, followed by full development occurring during pregnancy and lactation. Following lactation, the alveoli undergo apoptosis, and the mammary gland reverses back to resemble the nonparous gland. This process of growth and regression occurs for multiple pregnancies, suggesting the presence of a hierarchy of stem and progenitor cells that are able to regenerate specialized populations of mammary epithelial cells. Expansion of epithelial cell populations in the mammary gland is regulated by ovarian steroids, in particular estrogen acting through its receptor estrogen receptor alpha (ERα) and progesterone signaling through progesterone receptor (PR). A diverse number of stem and progenitor cells have been identified based on expression of cell surface markers and functional assays. Here we review the current understanding of how estrogen and progesterone act together and separately to regulate stem and progenitor cells within the human and mouse mammary tissues. Better understanding of the hierarchal organization of epithelial cell populations in the mammary gland and how the hormonal milieu affects its regulation may provide important insights into the origins of different subtypes of breast cancer.
Collapse
Affiliation(s)
- Lisa M Arendt
- Developmental, Molecular, and Chemical Biology Department, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, 136 Harrison Ave, Boston, MA, 02111, USA
- Molecular Oncology Research Institute, Tufts Medical Center, 800 Washington St, Boston, MA, 02111, USA
- Raymond and Beverly Sackler Laboratory for the Convergence of Biomedical, Physical and Engineering Sciences, Boston, MA, 02111, USA
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, 2015 Linden Dr, Madison, WI, 53706, USA
| | - Charlotte Kuperwasser
- Developmental, Molecular, and Chemical Biology Department, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, 136 Harrison Ave, Boston, MA, 02111, USA.
- Molecular Oncology Research Institute, Tufts Medical Center, 800 Washington St, Boston, MA, 02111, USA.
- Raymond and Beverly Sackler Laboratory for the Convergence of Biomedical, Physical and Engineering Sciences, Boston, MA, 02111, USA.
- Developmental, Molecular, and Chemical Biology Department, Tufts University School of Medicine, 800 Washington St, Box 5609, Boston, MA, 02111, USA.
| |
Collapse
|
113
|
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
|
114
|
Hilmarsdóttir B, Briem E, Sigurdsson V, Franzdóttir SR, Ringnér M, Arason AJ, Bergthorsson JT, Magnusson MK, Gudjonsson T. MicroRNA-200c-141 and ∆Np63 are required for breast epithelial differentiation and branching morphogenesis. Dev Biol 2015; 403:150-61. [PMID: 25967125 DOI: 10.1016/j.ydbio.2015.05.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 04/18/2015] [Accepted: 05/05/2015] [Indexed: 02/06/2023]
Abstract
The epithelial compartment of the breast contains two lineages, the luminal- and the myoepithelial cells. D492 is a breast epithelial cell line with stem cell properties that forms branching epithelial structures in 3D culture with both luminal- and myoepithelial differentiation. We have recently shown that D492 undergo epithelial to mesenchymal transition (EMT) when co-cultured with endothelial cells. This 3D co-culture model allows critical analysis of breast epithelial lineage development and EMT. In this study, we compared the microRNA (miR) expression profiles for D492 and its mesenchymal-derivative D492M. Suppression of the miR-200 family in D492M was among the most profound changes observed. Exogenous expression of miR-200c-141 in D492M reversed the EMT phenotype resulting in gain of luminal but not myoepithelial differentiation. In contrast, forced expression of ∆Np63 in D492M restored the myoepithelial phenotype only. Co-expression of miR-200c-141 and ∆Np63 in D492M restored the branching morphogenesis in 3D culture underlining the requirement for both luminal and myoepithelial elements for obtaining full branching morphogenesis in breast epithelium. Introduction of a miR-200c-141 construct in both D492 and D492M resulted in resistance to endothelial induced EMT. In conclusion, our data suggests that expression of miR-200c-141 and ∆Np63 in D492M can reverse EMT resulting in luminal- and myoepithelial differentiation, respectively, demonstrating the importance of these molecules in epithelial integrity in the human breast.
Collapse
Affiliation(s)
- Bylgja Hilmarsdóttir
- Stem Cell Research Unit, Biomedical Center, Department of Anatomy, Faculty of Medicine, School of Health Sciences, University of Iceland, Iceland; Department of Laboratory Hematology, Landspitali-University Hospital, Iceland
| | - Eirikur Briem
- Stem Cell Research Unit, Biomedical Center, Department of Anatomy, Faculty of Medicine, School of Health Sciences, University of Iceland, Iceland; Department of Laboratory Hematology, Landspitali-University Hospital, Iceland
| | - Valgardur Sigurdsson
- Stem Cell Research Unit, Biomedical Center, Department of Anatomy, Faculty of Medicine, School of Health Sciences, University of Iceland, Iceland
| | - Sigrídur Rut Franzdóttir
- Stem Cell Research Unit, Biomedical Center, Department of Anatomy, Faculty of Medicine, School of Health Sciences, University of Iceland, Iceland
| | - Markus Ringnér
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Lund University, Sweden
| | - Ari Jon Arason
- Stem Cell Research Unit, Biomedical Center, Department of Anatomy, Faculty of Medicine, School of Health Sciences, University of Iceland, Iceland; Department of Laboratory Hematology, Landspitali-University Hospital, Iceland
| | - Jon Thor Bergthorsson
- Stem Cell Research Unit, Biomedical Center, Department of Anatomy, Faculty of Medicine, School of Health Sciences, University of Iceland, Iceland; Department of Laboratory Hematology, Landspitali-University Hospital, Iceland
| | - Magnus Karl Magnusson
- Stem Cell Research Unit, Biomedical Center, Department of Anatomy, Faculty of Medicine, School of Health Sciences, University of Iceland, Iceland; Department of Laboratory Hematology, Landspitali-University Hospital, Iceland; Department of Medical Pharmacology and Toxicology, Faculty of Medicine, School of Health Sciences, University of Iceland, Iceland
| | - Thorarinn Gudjonsson
- Stem Cell Research Unit, Biomedical Center, Department of Anatomy, Faculty of Medicine, School of Health Sciences, University of Iceland, Iceland; Department of Laboratory Hematology, Landspitali-University Hospital, Iceland.
| |
Collapse
|
115
|
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
|
116
|
Lee JK, Garbe JC, Vrba L, Miyano M, Futscher BW, Stampfer MR, LaBarge MA. Age and the means of bypassing stasis influence the intrinsic subtype of immortalized human mammary epithelial cells. Front Cell Dev Biol 2015; 3:13. [PMID: 25815289 PMCID: PMC4356162 DOI: 10.3389/fcell.2015.00013] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 02/17/2015] [Indexed: 11/23/2022] Open
Abstract
Based on molecular features, breast cancers are grouped into intrinsic subtypes that have different prognoses and therapeutic response profiles. With increasing age, breast cancer incidence increases, with hormone receptor-positive and other luminal-like subtype tumors comprising a majority of cases. It is not known at what stage of tumor progression subtype specification occurs, nor how the process of aging affects the intrinsic subtype. We examined subtype markers in immortalized human mammary epithelial cell lines established following exposure of primary cultured cell strains to a two-step immortalization protocol that targets the two main barriers to immortality: stasis (stress-associated senescence) and replicative senescence. Cell lines derived from epithelial cells obtained from non-tumorous pre- and post-menopausal breast surgery tissues were compared. Additionally, comparisons were made between lines generated using two different genetic interventions to bypass stasis: transduction of either an shRNA that down-regulated p16INK4A, or overexpressed constitutive active cyclin D1/CDK2. In all cases, the replicative senescence barrier was bypassed by transduction of c-Myc. Cells from all resulting immortal lines exhibited normal karyotypes. Immunofluorescence, flow cytometry, and gene expression analyses of lineage-specific markers were used to categorize the intrinsic subtypes of the immortalized lines. Bypassing stasis with p16 shRNA in young strains generated cell lines that were invariably basal-like, but the lines examined from older strains exhibited some luminal features such as keratin 19 and estrogen receptor expression. Overexpression of cyclin D1/CDK2 resulted in keratin 19 positive, luminal-like cell lines from both young and old strains, and the lines examined from older strains exhibited estrogen receptor expression. Thus age and the method of bypassing stasis independently influence the subtype of immortalized human mammary epithelial cells.
Collapse
Affiliation(s)
- Jonathan K Lee
- Life Sciences Division, Lawrence Berkeley National Laboratory Berkeley, CA, USA
| | - James C Garbe
- Life Sciences Division, Lawrence Berkeley National Laboratory Berkeley, CA, USA
| | - Lukas Vrba
- Arizona Cancer Center, The University of Arizona Tucson, AZ, USA ; College of Pharmacy, Department of Pharmacology and Toxicology, The University of Arizona Tucson, AZ, USA
| | - Masaru Miyano
- Life Sciences Division, Lawrence Berkeley National Laboratory Berkeley, CA, USA
| | - Bernard W Futscher
- Arizona Cancer Center, The University of Arizona Tucson, AZ, USA ; College of Pharmacy, Department of Pharmacology and Toxicology, The University of Arizona Tucson, AZ, USA
| | - Martha R Stampfer
- Life Sciences Division, Lawrence Berkeley National Laboratory Berkeley, CA, USA ; Arizona Cancer Center, The University of Arizona Tucson, AZ, USA
| | - Mark A LaBarge
- Life Sciences Division, Lawrence Berkeley National Laboratory Berkeley, CA, USA
| |
Collapse
|
117
|
Kin K, Nnamani MC, Lynch VJ, Michaelides E, Wagner GP. Cell-type phylogenetics and the origin of endometrial stromal cells. Cell Rep 2015; 10:1398-409. [PMID: 25732829 DOI: 10.1016/j.celrep.2015.01.062] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 12/23/2014] [Accepted: 01/28/2015] [Indexed: 12/23/2022] Open
Abstract
A challenge of genome annotation is the identification of genes performing specific biological functions. Here, we propose a phylogenetic approach that utilizes RNA-seq data to infer the historical relationships among cell types and to trace the pattern of gene-expression changes on the tree. The hypothesis is that gene-expression changes coincidental with the origin of a cell type will be important for the function of the derived cell type. We apply this approach to the endometrial stromal cells (ESCs), which are critical for the initiation and maintenance of pregnancy. Our approach identified well-known regulators of ESCs, PGR and FOXO1, as well as genes not yet implicated in female fertility, including GATA2 and TFAP2C. Knockdown analysis confirmed that they are essential for ESC differentiation. We conclude that phylogenetic analysis of cell transcriptomes is a powerful tool for discovery of genes performing cell-type-specific functions.
Collapse
Affiliation(s)
- Koryu Kin
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT 06511, USA; Yale Systems Biology Institute, Yale University, New Haven, CT 06516, USA
| | - Mauris C Nnamani
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT 06511, USA; Yale Systems Biology Institute, Yale University, New Haven, CT 06516, USA
| | - Vincent J Lynch
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT 06511, USA; Yale Systems Biology Institute, Yale University, New Haven, CT 06516, USA; Department of Human Genetics, University of Chicago, Chicago, IL 60637, USA
| | | | - Günter P Wagner
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT 06511, USA; Yale Systems Biology Institute, Yale University, New Haven, CT 06516, USA.
| |
Collapse
|
118
|
Sani M, Hosseini SM, Salmannejad M, Aleahmad F, Ebrahimi S, Jahanshahi S, Talaei-Khozani T. Origins of the breast milk-derived cells; an endeavor to find the cell sources. Cell Biol Int 2015; 39:611-8. [PMID: 25572907 DOI: 10.1002/cbin.10432] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2014] [Accepted: 12/26/2014] [Indexed: 12/25/2022]
Abstract
Fresh human breast milk consists of a heterogeneous population of cells that may offer a non-invasive source of cells for therapeutic proposes. The aims of this study were to characterize the breast milk-derived cells cultured in vitro. To do this, the cells from human breast milk were cultured and the expression of the CD markers along with the embryonic stem cell markers, endothelial and luminal mammary epithelial cell markers was evaluated by flow cytometry and immunofluorescence. The presence of fetal microchimerism among the isolated cells was also determined by the presence of SRY gene. They were also differentiated into adipocytes and osteoblasts. The results showed that a remarkable number of cells expressed the mesenchymal stem cell (MSC) markers such as CD90, CD44, CD271, and CD146. A subpopulation of the human breast milk-derived cells (HBMDC) also expressed the embryonic stem cell markers, such as TRA 60-1, Oct4, Nanog and Sox2 but not SSEA1 or 4. The frequencies of the cells which expressed the endothelial, hematopoietic cell markers were negligible. SRY gene was not detected in the breast milk isolated cells. A subpopulation of the cells also expressed cytokeratin 18, the marker of luminal mammary epithelial cells. These cells showed the capability to differentiate into adipocytes and osteoblasts. In conclusion, these finding highlighted the presence of cells with various sources in the breast milk. Different stem cells including MSCs or embryonic stem cell-like cell along with the exfoliated cells from luminal epithelial cells were found among the isolated cells. The breast milk-derived stem cells might be considered as a non-invasive source of the stem cells for therapeutic purpose.
Collapse
Affiliation(s)
- Mahsa Sani
- Anatomy Department, Laboratory for stem cell research, Shiraz Medical School, Shiraz University of Medical Sciences, Shiraz, Iran; Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran; Cellular and Molecular Research Club, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | | | | | | | | | | |
Collapse
|
119
|
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
|
120
|
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
|
121
|
Gligorich KM, Vaden RM, Shelton DN, Wang G, Matsen CB, Looper RE, Sigman MS, Welm BE. Development of a screen to identify selective small molecules active against patient-derived metastatic and chemoresistant breast cancer cells. Breast Cancer Res 2014; 15:R58. [PMID: 23879992 PMCID: PMC4028696 DOI: 10.1186/bcr3452] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Revised: 04/19/2013] [Accepted: 07/23/2013] [Indexed: 12/13/2022] Open
Abstract
Introduction High failure rates of new investigational drugs have impaired the development of breast cancer therapies. One challenge is that excellent activity in preclinical models, such as established cancer cell lines, does not always translate into improved clinical outcomes for patients. New preclinical models, which better replicate clinically-relevant attributes of cancer, such as chemoresistance, metastasis and cellular heterogeneity, may identify novel anti-cancer mechanisms and increase the success of drug development. Methods Metastatic breast cancer cells were obtained from pleural effusions of consented patients whose disease had progressed. Normal primary human breast cells were collected from a reduction mammoplasty and immortalized with human telomerase. The patient-derived cells were characterized to determine their cellular heterogeneity and proliferation rate by flow cytometry, while dose response curves were performed for chemotherapies to assess resistance. A screen was developed to measure the differential activity of small molecules on the growth and survival of patient-derived normal breast and metastatic, chemoresistant tumor cells to identify selective anti-cancer compounds. Several hits were identified and validated in dose response assays. One compound, C-6, was further characterized for its effect on cell cycle and cell death in cancer cells. Results Patient-derived cells were found to be more heterogeneous, with reduced proliferation rates and enhanced resistance to chemotherapy compared to established cell lines. A screen was subsequently developed that utilized both tumor and normal patient-derived cells. Several compounds were identified, which selectively targeted tumor cells, but not normal cells. Compound C-6 was found to inhibit proliferation and induce cell death in tumor cells via a caspase-independent mechanism. Conclusions Short-term culture of patient-derived cells retained more clinically relevant features of breast cancer compared to established cell lines. The low proliferation rate and chemoresistance make patient-derived cells an excellent tool in preclinical drug development.
Collapse
|
122
|
Balk-Møller E, Kim J, Hopkinson B, Timmermans-Wielenga V, Petersen OW, Villadsen R. A marker of endocrine receptor-positive cells, CEACAM6, is shared by two major classes of breast cancer: luminal and HER2-enriched. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:1198-208. [PMID: 24655379 DOI: 10.1016/j.ajpath.2013.12.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Revised: 12/11/2013] [Accepted: 12/12/2013] [Indexed: 12/19/2022]
Abstract
Elucidating the phenotypic evolution of breast cancer through distinct subtypes relies heavily on defining a lineage blueprint of the normal human breast. Here, we show that in normal breast, within the luminal epithelial lineage, a subset of cells characterized by strong staining for endocrine receptors are also characterized by expression of the surface marker CEACAM6. Topographically, this pattern of staining predominates in terminal ductal lobular units, rather than in interlobular ducts. In culture, CEACAM6-expressing cells remain essentially postmitotic under conditions in which the other cells of luminal epithelial lineage are highly proliferative. We examined the pattern of expression among three major breast cancer subtypes: luminal, HER2-enriched, and basal-like. In 104 biopsies, the luminal and HER2-enriched subtypes showed a high proportion of CEACAM6(+) tumors (78% and 83%, respectively); the basal-like subtype showed a low proportion (28%). Further accentuation of this pattern was observed in 13 established breast cancer cell lines. When differentiation was induced by all-trans retinoic acid, CEACAM6 expression strongly correlated with luminal-like differentiation. Furthermore, CEACAM6(+) cancer cells were less proliferative than CEACAM6(-) cells in tumorsphere assays and were less tumorigenic in nude mice. Based on these observations, we propose that luminal and HER2-enriched breast cancers are more closely related than previously thought and may share a common cell of origin.
Collapse
Affiliation(s)
- Emilie Balk-Møller
- Department of Cellular and Molecular Medicine, the Panum Institute, the Center for Biological Disease Analysis, and the Danish Stem Cell Center, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jiyoung Kim
- Department of Cellular and Molecular Medicine, the Panum Institute, the Center for Biological Disease Analysis, and the Danish Stem Cell Center, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Branden Hopkinson
- Department of Cellular and Molecular Medicine, the Panum Institute, the Center for Biological Disease Analysis, and the Danish Stem Cell Center, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Ole W Petersen
- Department of Cellular and Molecular Medicine, the Panum Institute, the Center for Biological Disease Analysis, and the Danish Stem Cell Center, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - René Villadsen
- Department of Cellular and Molecular Medicine, the Panum Institute, the Center for Biological Disease Analysis, and the Danish Stem Cell Center, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
123
|
Differential in vivo tumorigenicity of distinct subpopulations from a luminal-like breast cancer xenograft. PLoS One 2014; 9:e113278. [PMID: 25419568 PMCID: PMC4242648 DOI: 10.1371/journal.pone.0113278] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 10/22/2014] [Indexed: 01/16/2023] Open
Abstract
Intratumor heterogeneity caused by genetic, phenotypic or functional differences between cancer cell subpopulations is a considerable clinical challenge. Understanding subpopulation dynamics is therefore central for both optimization of existing therapy and for development of new treatment. The aim of this study was to isolate subpopulations from a primary tumor and by comparing molecular characteristics of these subpopulations, find explanations to their differing tumorigenicity. Cell subpopulations from two patient derived in vivo models of primary breast cancer, ER+ and ER-, were identified. EpCAM+ cells from the ER+ model gave rise to tumors independently of stroma cell support. The tumorigenic fraction was further divided based on SSEA-4 and CD24 expression. Both markers were expressed in ER+ breast cancer biopsies. FAC-sorted cells based on EpCAM, SSEA-4 and CD24 expression were subsequently tested for differences in functionality by in vivo tumorigenicity assay. Three out of four subpopulations of cells were tumorigenic and showed variable ability to recapitulate the marker expression of the original tumor. Whole genome expression analysis of the sorted populations disclosed high similarity in the transcriptional profiles between the tumorigenic populations. Comparing the non-tumorigenic vs the tumorigenic populations, 44 transcripts were, however, significantly differentially expressed. A subset of these, 26 identified and named genes, highly expressed in the non-tumorigenic population, predicted longer overall survival (N = 737, p<0.0001) and distant metastasis free survival (DMFS) (N = 1379, p<0.0001) when performing Kaplan-Meier survival analysis using the GOBO online database. The 26 gene set correlated with longer DMFS in multiple breast cancer subgroups. Copy number profiling revealed no aberrations that could explain the observed differences in tumorigenicity. This study emphasizes the functional variability among cell populations that are otherwise genomically similar, and that the risk of breast cancer recurrence can only be eliminated if the tumorigenic abilities in multiple cancer cell subpopulations are inhibited.
Collapse
|
124
|
Hassiotou F, Hartmann PE. At the dawn of a new discovery: the potential of breast milk stem cells. Adv Nutr 2014; 5:770-8. [PMID: 25398739 PMCID: PMC4224213 DOI: 10.3945/an.114.006924] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Breast milk contains bioactive molecules that provide a multitude of immunologic, developmental and nutritional benefits to the infant. Less attention has been placed on the cellular nature of breast milk, which contains thousands to millions of maternal cells in every milliliter that the infant ingests. What are the properties and roles of these cells? Most studies have examined breast milk cells from an immunologic perspective, focusing specifically on the leukocytes, mainly in the early postpartum period. In the past decade, research has taken a multidimensional approach to investigating the cells of human milk. Technologic advances in single cell analysis and imaging have aided this work, which has resulted in the breakthrough discovery of stem cells in breast milk with multilineage potential that are transferred to the offspring during breastfeeding. This has generated numerous implications for both infant and maternal health and regenerative medicine. This review summarizes the latest knowledge on breast milk stem cells, and discusses their known in vitro and in vivo attributes as well as potential functions and applications.
Collapse
Affiliation(s)
- Foteini Hassiotou
- School of Chemistry and Biochemistry, Faculty of Science, The University of Western Australia, Crawley, Australia
| | | |
Collapse
|
125
|
Rennó AL, Alves-Júnior MJ, Rocha RM, De Souza PC, de Souza VB, Jampietro J, Vassallo J, Hyslop S, Anhê GF, de Moraes Schenka NG, Soares FA, Schenka AA. Decreased Expression of Stem Cell Markers by Simvastatin in 7,12-dimethylbenz(a)anthracene (DMBA)–induced Breast Cancer. Toxicol Pathol 2014; 43:400-10. [DOI: 10.1177/0192623314544707] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Simvastatin, a competitive inhibitor of HMG-CoA reductase widely used in the treatment and prevention of hyperlipidemia-related diseases, has recently been associated to in vitro anticancer stem cell (CSC) actions. However, these effects have not been confirmed in vivo. To assess in vivo anti-CSC effects of simvastatin, female Sprague-Dawley rats with 7,12-dimethyl-benz(a)anthracene (DMBA)–induced mammary cancer and control animals were treated for 14 days with either simvastatin (20 or 40 mg/kg/day) or soybean oil ( N = 60). Tumors and normal breast tissues were removed for pathologic examination and immunodetection of CSC markers. At 40 mg/kg/day, simvastatin significantly reduced tumor growth and the expression of most CSC markers. The reduction in tumor growth (80%) could not be explained solely by the decrease in CSCs, since the latter accounted for less than 10% of the neoplasia (differentiated cancer cells were also affected). Stem cells in normal, nonneoplastic breast tissues were not affected by simvastatin. Simvastatin was also associated with a significant decrease in proliferative activity but no increase in cell death. In conclusion, this is the first study to confirm simvastatin anti-CSC actions in vivo, further demonstrating that this effect is specific for neoplastic cells, but not restricted to CSCs, and most likely due to inhibition of cell proliferation.
Collapse
Affiliation(s)
- André Lisboa Rennó
- Department of Pharmacology, School of Medical Sciences, State University of Campinas (Unicamp), São Paulo, Brazil
| | - Marcos José Alves-Júnior
- Department of Pharmacology, School of Medical Sciences, State University of Campinas (Unicamp), São Paulo, Brazil
| | - Rafael Malagoli Rocha
- Laboratory of Investigative Pathology, Department of Anatomic Pathology, Hospital AC Camargo, São Paulo, Brazil
| | - Philipi Coutinho De Souza
- Department of Pharmacology, School of Medical Sciences, State University of Campinas (Unicamp), São Paulo, Brazil
| | - Valéria Barbosa de Souza
- Department of Pharmacology, School of Medical Sciences, State University of Campinas (Unicamp), São Paulo, Brazil
| | - Juliano Jampietro
- Laboratory of Investigative Pathology, Department of Anatomic Pathology, Hospital AC Camargo, São Paulo, Brazil
| | - José Vassallo
- Laboratory of Investigative Pathology, Department of Anatomic Pathology, Hospital AC Camargo, São Paulo, Brazil
- Laboratory of Investigative and Molecular Pathology, Center for Investigation in Pediatrics (Ciped), São Paulo, Brazil
| | - Stephen Hyslop
- Department of Pharmacology, School of Medical Sciences, State University of Campinas (Unicamp), São Paulo, Brazil
| | - Gabriel Forato Anhê
- Department of Pharmacology, School of Medical Sciences, State University of Campinas (Unicamp), São Paulo, Brazil
| | | | - Fernando Augusto Soares
- Laboratory of Investigative Pathology, Department of Anatomic Pathology, Hospital AC Camargo, São Paulo, Brazil
| | - André Almeida Schenka
- Department of Pharmacology, School of Medical Sciences, State University of Campinas (Unicamp), São Paulo, Brazil
- Laboratory of Investigative Pathology, Department of Anatomic Pathology, Hospital AC Camargo, São Paulo, Brazil
- Laboratory of Investigative and Molecular Pathology, Center for Investigation in Pediatrics (Ciped), São Paulo, Brazil
| |
Collapse
|
126
|
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
|
127
|
Tang J, Fernandez-Garcia I, Vijayakumar S, Martinez-Ruis H, Illa-Bochaca I, Nguyen DH, Mao JH, Costes SV, Barcellos-Hoff MH. Irradiation of juvenile, but not adult, mammary gland increases stem cell self-renewal and estrogen receptor negative tumors. Stem Cells 2014; 32:649-61. [PMID: 24038768 DOI: 10.1002/stem.1533] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Revised: 06/30/2013] [Accepted: 07/24/2013] [Indexed: 02/06/2023]
Abstract
Children exposed to ionizing radiation have a substantially greater breast cancer risk than adults; the mechanism for this strong age dependence is not known. Here we show that pubertal murine mammary glands exposed to sparsely or densely ionizing radiation exhibit enrichment of mammary stem cell and Notch pathways, increased mammary repopulating activity indicative of more stem cells, and propensity to develop estrogen receptor (ER) negative tumors thought to arise from stem cells. We developed a mammary lineage agent-based model (ABM) to evaluate cell inactivation, self-renewal, or dedifferentiation via epithelial-mesenchymal transition (EMT) as mechanisms by which radiation could increase stem cells. ABM rejected cell inactivation and predicted increased self-renewal would only affect juveniles while dedifferentiation could act in both juveniles and adults. To further test self-renewal versus dedifferentiation, we used the MCF10A human mammary epithelial cell line, which recapitulates ductal morphogenesis in humanized fat pads, undergoes EMT in response to radiation and transforming growth factor β (TGFβ) and contains rare stem-like cells that are Let-7c negative or express both basal and luminal cytokeratins. ABM simulation of population dynamics of double cytokeratin cells supported increased self-renewal in irradiated MCF10A treated with TGFβ. Radiation-induced Notch concomitant with TGFβ was necessary for increased self-renewal of Let-7c negative MCF10A cells but not for EMT, indicating that these are independent processes. Consistent with these data, irradiating adult mice did not increase mammary repopulating activity or ER-negative tumors. These studies suggest that irradiation during puberty transiently increases stem cell self-renewal, which increases susceptibility to developing ER-negative breast cancer.
Collapse
Affiliation(s)
- Jonathan Tang
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley California, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
128
|
Coradini D, Boracchi P, Oriana S, Biganzoli E, Ambrogi F. Differential expression of genes involved in the epigenetic regulation of cell identity in normal human mammary cell commitment and differentiation. CHINESE JOURNAL OF CANCER 2014; 33:501-10. [PMID: 25223915 PMCID: PMC4198753 DOI: 10.5732/cjc.014.10066] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
The establishment and maintenance of mammary epithelial cell identity depends on the activity of a group of proteins, collectively called maintenance proteins, that act as epigenetic regulators of gene transcription through DNA methylation, histone modification, and chromatin remodeling. Increasing evidence indicates that dysregulation of these crucial proteins may disrupt epithelial cell integrity and trigger breast tumor initiation. Therefore, we explored in silico the expression pattern of a panel of 369 genes known to be involved in the establishment and maintenance of epithelial cell identity and mammary gland remodeling in cell subpopulations isolated from normal human mammary tissue and selectively enriched in their content of bipotent progenitors, committed luminal progenitors, and differentiated myoepithelial or differentiated luminal cells. The results indicated that, compared to bipotent cells, differentiated myoepithelial and luminal subpopulations were both characterized by the differential expression of 4 genes involved in cell identity maintenance: CBX6 and PCGF2, encoding proteins belonging to the Polycomb group, and SMARCD3 and SMARCE1, encoding proteins belonging to the Trithorax group. In addition to these common genes, the myoepithelial phenotype was associated with the differential expression of HDAC1, which encodes histone deacetylase 1, whereas the luminal phenotype was associated with the differential expression of SMARCA4 and HAT1, which encode a Trithorax protein and histone acetylase 1, respectively. The luminal compartment was further characterized by the overexpression of ALDH1A3 and GATA3, and the down-regulation of NOTCH4 and CCNB1, with the latter suggesting a block in cell cycle progression at the G2 phase. In contrast, myoepithelial differentiation was associated with the overexpression of MYC and the down-regulation of CCNE1, with the latter suggesting a block in cell cycle progression at the G1 phase.
Collapse
Affiliation(s)
- Danila Coradini
- Department of Clinical Sciences and Community Health, Medical Statistics, Biometry and Bioinformatics, University of Milan, Via Vanzetti 5, Milan 20133, Italy;
| | | | | | | | | |
Collapse
|
129
|
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
|
130
|
Functional Role of the microRNA-200 Family in Breast Morphogenesis and Neoplasia. Genes (Basel) 2014; 5:804-20. [PMID: 25216122 PMCID: PMC4198932 DOI: 10.3390/genes5030804] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Revised: 09/03/2014] [Accepted: 09/04/2014] [Indexed: 12/21/2022] Open
Abstract
Branching epithelial morphogenesis is closely linked to epithelial-to-mesenchymal transition (EMT), a process important in normal development and cancer progression. The miR-200 family regulates epithelial morphogenesis and EMT through a negative feedback loop with the ZEB1 and ZEB2 transcription factors. miR-200 inhibits expression of ZEB1/2 mRNA, which in turn can down-regulate the miR-200 family that further results in down-regulation of E-cadherin and induction of a mesenchymal phenotype. Recent studies show that the expression of miR-200 genes is high during late pregnancy and lactation, thereby indicating that these miRs are important for breast epithelial morphogenesis and differentiation. miR-200 genes have been studied intensively in relation to breast cancer progression and metastasis, where it has been shown that miR-200 members are down-regulated in basal-like breast cancer where the EMT phenotype is prominent. There is growing evidence that the miR-200 family is up-regulated in distal breast metastasis indicating that these miRs are important for colonization of metastatic breast cancer cells through induction of mesenchymal to epithelial transition. The dual role of miR-200 in primary and metastatic breast cancer is of interest for future therapeutic interventions, making it important to understand its role and interacting partners in more detail.
Collapse
|
131
|
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
|
132
|
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
|
133
|
Duss S, Brinkhaus H, Britschgi A, Cabuy E, Frey DM, Schaefer DJ, Bentires-Alj M. Mesenchymal precursor cells maintain the differentiation and proliferation potentials of breast epithelial cells. Breast Cancer Res 2014; 16:R60. [PMID: 24916766 PMCID: PMC4095576 DOI: 10.1186/bcr3673] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Accepted: 05/22/2014] [Indexed: 12/16/2022] Open
Abstract
INTRODUCTION Stromal-epithelial interactions play a fundamental role in tissue homeostasis, controlling cell proliferation and differentiation. Not surprisingly, aberrant stromal-epithelial interactions contribute to malignancies. Studies of the cellular and molecular mechanisms underlying these interactions require ex vivo experimental model systems that recapitulate the complexity of human tissue without compromising the differentiation and proliferation potentials of human primary cells. METHODS We isolated and characterized human breast epithelial and mesenchymal precursors from reduction mammoplasty tissue and tagged them with lentiviral vectors. We assembled heterotypic co-cultures and compared mesenchymal and epithelial cells to cells in corresponding monocultures by analyzing growth, differentiation potentials, and gene expression profiles. RESULTS We show that heterotypic culture of non-immortalized human primary breast epithelial and mesenchymal precursors maintains their proliferation and differentiation potentials and constrains their growth. We further describe the gene expression profiles of stromal and epithelial cells in co-cultures and monocultures and show increased expression of the tumor growth factor beta (TGFβ) family member inhibin beta A (INHBA) in mesenchymal cells grown as co-cultures compared with monocultures. Notably, overexpression of INHBA in mesenchymal cells increases colony formation potential of epithelial cells, suggesting that it contributes to the dynamic reciprocity between breast mesenchymal and epithelial cells. CONCLUSIONS The described heterotypic co-culture system will prove useful for further characterization of the molecular mechanisms mediating interactions between human normal or neoplastic breast epithelial cells and the stroma, and will provide a framework to test the relevance of the ever-increasing number of oncogenomic alterations identified in human breast cancer.
Collapse
Affiliation(s)
- Stephan Duss
- Friedrich Miescher Institute for Biomedical Research, Mechanisms of Cancer, Maulbeerstrasse 66, CH-4058 Basel, Switzerland
| | - Heike Brinkhaus
- Friedrich Miescher Institute for Biomedical Research, Mechanisms of Cancer, Maulbeerstrasse 66, CH-4058 Basel, Switzerland
| | - Adrian Britschgi
- Friedrich Miescher Institute for Biomedical Research, Mechanisms of Cancer, Maulbeerstrasse 66, CH-4058 Basel, Switzerland
| | - Erik Cabuy
- Friedrich Miescher Institute for Biomedical Research, Mechanisms of Cancer, Maulbeerstrasse 66, CH-4058 Basel, Switzerland
| | - Daniel M Frey
- Department of Surgery, University Hospital of Basel, Spitalstrasse 21, CH-4058 Basel, Switzerland
| | - Dirk J Schaefer
- Department of Surgery, University Hospital of Basel, Spitalstrasse 21, CH-4058 Basel, Switzerland
| | - Mohamed Bentires-Alj
- Friedrich Miescher Institute for Biomedical Research, Mechanisms of Cancer, Maulbeerstrasse 66, CH-4058 Basel, Switzerland
| |
Collapse
|
134
|
Pelissier FA, Garbe JC, Ananthanarayanan B, Miyano M, Lin C, Jokela T, Kumar S, Stampfer MR, Lorens JB, LaBarge MA. Age-related dysfunction in mechanotransduction impairs differentiation of human mammary epithelial progenitors. Cell Rep 2014; 7:1926-39. [PMID: 24910432 DOI: 10.1016/j.celrep.2014.05.021] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Revised: 04/15/2014] [Accepted: 05/08/2014] [Indexed: 11/29/2022] Open
Abstract
Dysfunctional progenitor and luminal cells with acquired basal cell properties accumulate during human mammary epithelial aging for reasons not understood. Multipotent progenitors from women aged <30 years were exposed to a physiologically relevant range of matrix elastic modulus (stiffness). Increased stiffness causes a differentiation bias towards myoepithelial cells while reducing production of luminal cells and progenitor maintenance. Lineage representation in progenitors from women >55 years is unaffected by physiological stiffness changes. Efficient activation of Hippo pathway transducers YAP and TAZ is required for the modulus-dependent myoepithelial/basal bias in younger progenitors. In older progenitors, YAP and TAZ are activated only when stressed with extraphysiologically stiff matrices, which bias differentiation towards luminal-like phenotypes. In vivo YAP is primarily active in myoepithelia of younger breasts, but localization and activity increases in luminal cells with age. Thus, aging phenotypes of mammary epithelia may arise partly because alterations in Hippo pathway activation impair microenvironment-directed differentiation and lineage specificity.
Collapse
Affiliation(s)
- Fanny A Pelissier
- Life Science Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA; Center for Cancer Biomarkers, Department of Biomedicine, University of Bergen, Bergen 5009, Norway
| | - James C Garbe
- Life Science Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | | | - Masaru Miyano
- Life Science Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - ChunHan Lin
- Life Science Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA; Department of Comparative Biochemistry, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Tiina Jokela
- Life Science Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA; Center for Cancer Biomarkers, Department of Biomedicine, University of Bergen, Bergen 5009, Norway
| | - Sanjay Kumar
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Martha R Stampfer
- Life Science Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - James B Lorens
- Center for Cancer Biomarkers, Department of Biomedicine, University of Bergen, Bergen 5009, Norway
| | - Mark A LaBarge
- Life Science Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA.
| |
Collapse
|
135
|
Lombardi S, Honeth G, Ginestier C, Shinomiya I, Marlow R, Buchupalli B, Gazinska P, Brown J, Catchpole S, Liu S, Barkan A, Wicha M, Purushotham A, Burchell J, Pinder S, Dontu G. Growth hormone is secreted by normal breast epithelium upon progesterone stimulation and increases proliferation of stem/progenitor cells. Stem Cell Reports 2014; 2:780-93. [PMID: 24936466 PMCID: PMC4050343 DOI: 10.1016/j.stemcr.2014.05.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Revised: 05/06/2014] [Accepted: 05/07/2014] [Indexed: 01/16/2023] Open
Abstract
Using in vitro and in vivo experimental systems and in situ analysis, we show that growth hormone (GH) is secreted locally by normal human mammary epithelial cells upon progesterone stimulation. GH increases proliferation of a subset of cells that express growth hormone receptor (GHR) and have functional properties of stem and early progenitor cells. In 72% of ductal carcinoma in situ lesions, an expansion of the cell population that expresses GHR was observed, suggesting that GH signaling may contribute to breast cancer development.
Collapse
Affiliation(s)
- Sara Lombardi
- Research Oncology, King's College London, London SE1 9RT, UK
| | | | - Christophe Ginestier
- Centre de Recherche et Cancérologie, Marseille, Inserm, CRCM, U1068, France ; Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Rebecca Marlow
- Research Oncology, King's College London, London SE1 9RT, UK
| | | | | | - John Brown
- Research Oncology, King's College London, London SE1 9RT, UK
| | | | - Suling Liu
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ariel Barkan
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Max Wicha
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Joy Burchell
- Research Oncology, King's College London, London SE1 9RT, UK
| | - Sarah Pinder
- Research Oncology, King's College London, London SE1 9RT, UK
| | - Gabriela Dontu
- Research Oncology, King's College London, London SE1 9RT, UK
| |
Collapse
|
136
|
Chen F, Li A, Gao S, Hollern D, Williams M, Liu F, VanSickle EA, Andrechek E, Zhang C, Yang C, Luo R, Xiao H. Tip30 controls differentiation of murine mammary luminal progenitor to estrogen receptor-positive luminal cell through regulating FoxA1 expression. Cell Death Dis 2014; 5:e1242. [PMID: 24853420 PMCID: PMC4047867 DOI: 10.1038/cddis.2014.224] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 04/11/2014] [Accepted: 04/17/2014] [Indexed: 12/21/2022]
Abstract
Estrogen receptor-alpha positive (ER+) breast cancers comprise the majority of human breast cancers, but molecular mechanisms underlying this subtype of breast cancers remain poorly understood. Here, we show that ER+ mammary luminal tumors arising in Tip30−/−MMTV-Neu mice exhibited increased enrichment of luminal progenitor gene signature. Deletion of the Tip30 gene increased proportion of mammary stem and progenitor cell populations, and raised susceptibility to ER+ mammary luminal tumors in female Balb/c mice. Moreover, Tip30−/− luminal progenitors displayed increases in propensity to differentiate to mature ER+ luminal cells and FoxA1 expression. Knockdown of FoxA1 expression in Tip30−/− progenitors by shRNA specific for FoxA1 reduced their differentiation toward ER+ mature luminal cells. Taken together, our results suggest that TIP30 is a key regulator for maintaining ER+ and ER−luminal pools in the mammary luminal lineage, and loss of it promotes expansion of ER+ luminal progenitors and mature cells and ER+ mammary tumorigenesis.
Collapse
Affiliation(s)
- F Chen
- 1] Department of Physiology, Michigan State University, East Lansing, MI, USA [2] Cancer Center, Southern Medical University, Guangzhou, China [3] Traditional Chinese Medicine-Integrated Hospital, Southern Medical University, Southern Medical University, Guangzhou, China
| | - A Li
- 1] Department of Physiology, Michigan State University, East Lansing, MI, USA [2] Cancer Center, Southern Medical University, Guangzhou, China [3] Traditional Chinese Medicine-Integrated Hospital, Southern Medical University, Southern Medical University, Guangzhou, China
| | - S Gao
- 1] Department of Physiology, Michigan State University, East Lansing, MI, USA [2] Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
| | - D Hollern
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - M Williams
- 1] Department of Physiology, Michigan State University, East Lansing, MI, USA [2] Cell and Molecular Biology Program, Michigan State University, East Lansing, MI, USA
| | - F Liu
- 1] Department of Physiology, Michigan State University, East Lansing, MI, USA [2] Traditional Chinese Medicine-Integrated Hospital, Southern Medical University, Southern Medical University, Guangzhou, China
| | - E A VanSickle
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - E Andrechek
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - C Zhang
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - C Yang
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - R Luo
- 1] Cancer Center, Southern Medical University, Guangzhou, China [2] Traditional Chinese Medicine-Integrated Hospital, Southern Medical University, Southern Medical University, Guangzhou, China
| | - H Xiao
- 1] Department of Physiology, Michigan State University, East Lansing, MI, USA [2] Cell and Molecular Biology Program, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
137
|
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
|
138
|
Rios AC, Fu NY, Lindeman GJ, Visvader JE. In situ identification of bipotent stem cells in the mammary gland. Nature 2014; 506:322-7. [DOI: 10.1038/nature12948] [Citation(s) in RCA: 400] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2013] [Accepted: 12/12/2013] [Indexed: 12/29/2022]
|
139
|
Progesterone stimulates progenitor cells in normal human breast and breast cancer cells. Breast Cancer Res Treat 2014; 143:423-33. [DOI: 10.1007/s10549-013-2817-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Accepted: 12/18/2013] [Indexed: 01/16/2023]
|
140
|
|
141
|
|
142
|
Mukhopadhyay C, Zhao X, Maroni D, Band V, Naramura M. Distinct effects of EGFR ligands on human mammary epithelial cell differentiation. PLoS One 2013; 8:e75907. [PMID: 24124521 PMCID: PMC3790811 DOI: 10.1371/journal.pone.0075907] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Accepted: 08/16/2013] [Indexed: 11/18/2022] Open
Abstract
Based on gene expression patterns, breast cancers can be divided into subtypes that closely resemble various developmental stages of normal mammary epithelial cells (MECs). Thus, understanding molecular mechanisms of MEC development is expected to provide critical insights into initiation and progression of breast cancer. Epidermal growth factor receptor (EGFR) and its ligands play essential roles in normal and pathological mammary gland. Signals through EGFR is required for normal mammary gland development. Ligands for EGFR are over-expressed in a significant proportion of breast cancers, and elevated expression of EGFR is associated with poorer clinical outcome. In the present study, we examined the effect of signals through EGFR on MEC differentiation using the human telomerase reverse transcriptase (hTERT)-immortalized human stem/progenitor MECs which express cytokeratin 5 but lack cytokeratin 19 (K5+K19- hMECs). As reported previously, these cells can be induced to differentiate into luminal and myoepithelial cells under appropriate culture conditions. K5+K19- hMECs acquired distinct cell fates in response to EGFR ligands epidermal growth factor (EGF), amphiregulin (AREG) and transforming growth factor alpha (TGFα) in differentiation-promoting MEGM medium. Specifically, presence of EGF during in vitro differentiation supported development into both luminal and myoepithelial lineages, whereas cells differentiated only towards luminal lineage when EGF was replaced with AREG. In contrast, substitution with TGFα led to differentiation only into myoepithelial lineage. Chemical inhibition of the MEK-Erk pathway, but not the phosphatidylinositol 3-kinase (PI3K)-AKT pathway, interfered with K5+K19- hMEC differentiation. The present data validate the utility of the K5+K19- hMEC cells for modeling key features of human MEC differentiation. This system should be useful in studying molecular/biochemical mechanisms of human MEC differentiation.
Collapse
Affiliation(s)
- Chandrani Mukhopadhyay
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Xiangshan Zhao
- Department of Genetics, Cell Biology and Anatomy, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Dulce Maroni
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Vimla Band
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Department of Genetics, Cell Biology and Anatomy, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Mayumi Naramura
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Department of Genetics, Cell Biology and Anatomy, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- * E-mail:
| |
Collapse
|
143
|
Epithelial-to-mesenchymal transition: what is the impact on breast cancer stem cells and drug resistance. Cancer Treat Rev 2013; 40:341-8. [PMID: 24090504 DOI: 10.1016/j.ctrv.2013.09.008] [Citation(s) in RCA: 185] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Revised: 09/03/2013] [Accepted: 09/06/2013] [Indexed: 12/24/2022]
Abstract
There is increasing interest in cancer stem cells (CSCs) and their role in cancer progression. Recently, CSCs have been identified in brain, skin, and intestinal tumors and it has been suggested that these CSCs are responsible for tumor growth and metastasis. In breast cancer fatality is often due to the development of metastatic disease (MBC). Almost 30% of early breast cancer patients eventually develop MBC and in 90% of these multi-drug resistance (MDR) occurs. This could be attributed to the presence of breast cancer stem cells (BCSCs). Epithelial-to-mesenchymal transition (EMT) is a process known to contribute to metastasis in cancer and it is mainly characterized by loss of E-cadherin expression. The TGF-β signaling pathway has an established role in promoting EMT by down-regulating E-cadherin via a number of transcription factors, such as Twist, Snail and Slug. EMT has also been reported to produce cells with stem cell-like properties. Definition of the exact molecular mechanisms that are involved in the generation of stem cells through EMT could lead to the identification of new potential therapeutic targets and enable the development of more efficient strategies for particular patient groups. In this review we discuss what is known about the relationship between EMT, BCSCs and MDR.
Collapse
|
144
|
Abstract
Many organs respond to physiological challenges by changing tissue size or composition. Such changes may originate from tissue-specific stem cells and their supportive environment (niche). The endocrine system is a major effector and conveyor of physiological changes and as such could alter stem cell behavior in various ways. In this review, we examine how hormones affect stem cell biology in four different organs: the ovary, intestine, hematopoietic system, and mammary gland. Hormones control every stage of stem cell life, including establishment, expansion, maintenance, and differentiation. The effects can be cell autonomous or non-cell autonomous through the niche. Moreover, a single hormone can affect different stem cells in different ways or affect the same stem cell differently at various developmental times. The vast complexity and diversity of stem cell responses to hormonal cues allow hormones to coordinate the body's reaction to physiological challenges.
Collapse
Affiliation(s)
- Dana Gancz
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, 76100 Israel; ,
| | | |
Collapse
|
145
|
Castro DJ, Maurer J, Hebbard L, Oshima RG. ROCK1 inhibition promotes the self-renewal of a novel mouse mammary cancer stem cell. Stem Cells 2013; 31:12-22. [PMID: 22961723 DOI: 10.1002/stem.1224] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2012] [Accepted: 08/10/2012] [Indexed: 12/20/2022]
Abstract
The differentiation of stem-like tumor cells may contribute to the cellular heterogeneity of breast cancers. We report the propagation of highly enriched mouse mammary cancer stem cells that retain the potential to differentiate both in vivo and in culture and their use to identify chemical compounds that influence both self-renewal and differentiation. We identify epithelial tumor-initiating cells (ETICs) that express lineage markers of both basal and luminal mammary cell lineages and retain the potential, from even single cells, to generate heterogeneous tumors similar to the tumor of origin. ETICs can progress through a Rho-associated coiled-coil containing protein kinase 1 dependent, epithelial to mesenchymal transition to generate mesenchymal tumor-initiating cells capable of initiating tumors of limited heterogeneity. The propagation of ETICs may allow for the identification of new therapeutic compounds that may inhibit or prevent progression of some types of breast cancer.
Collapse
Affiliation(s)
- David J Castro
- Cancer Research Center, Sanford-Burnham Medical Research Institute, La Jolla, California, USA
| | | | | | | |
Collapse
|
146
|
Prabhakaran P, Hassiotou F, Blancafort P, Filgueira L. Cisplatin induces differentiation of breast cancer cells. Front Oncol 2013; 3:134. [PMID: 23761858 PMCID: PMC3669802 DOI: 10.3389/fonc.2013.00134] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Accepted: 05/13/2013] [Indexed: 12/25/2022] Open
Abstract
Breast tumors are heterogeneous including cells with stem cell properties and more differentiated cells. This heterogeneity is reflected into the molecular breast cancer subtypes. Breast cancer stem cells are resistant to chemotherapy, thus recent efforts are focusing on identifying treatments that shift them toward a more differentiated phenotype, making them more susceptible to chemotherapy. We examined whether the drug cisplatin induces differentiation in breast cancer cell lines that represent different breast cancer subtypes. We used three cell lines representing triple-negative breast cancers, BT-549 and MDA-MB-231 (claudin-low), and MDA-MB-468 (basal-like), along with estrogen and progesterone receptor positive MCF-7 cells (luminal). Cisplatin was applied at 2.5, 5, 10, and 20 μM, and cell viability and proliferation were measured using MTS and BrdU assays, respectively. The effect of cisplatin on the cellular hierarchy was examined by flow cytometry, immunofluorescence and qRT-PCR. Cisplatin treatment of 10 and 20 μM reduced cell viability by 36–51% and proliferation capacity by 36–67%. Treatment with cisplatin resulted in 12–67% down-regulation of stem cell markers (CD49f, SSEA4) and 10–130% up-regulation of differentiation markers (CK18, SMA, β-tubulin). At the mRNA level, CD49f was down-regulated whilst β-tubulin was up-regulated in the claudin-low cell lines. SSEA4 protein expression decreased upon cisplatin treatment, but SSEA4 mRNA expression increased indicating a differential regulation of cisplatin at the post-transcriptional level. It is concluded that cisplatin reduces breast cancer cell survival and induces differentiation of stem/progenitor cell subpopulations within breast cancer cell lines. These effects indicate the potential of this drug to target specific chemotherapy-resistant cells within a tumor.
Collapse
Affiliation(s)
- Praseetha Prabhakaran
- School of Anatomy, Physiology and Human Biology, The University of Western Australia , Crawley, Perth, WA , Australia ; Faculty of Biosciences and Bioengineering, Universiti Teknologi Malaysia , Skudai, Johor , Malaysia
| | | | | | | |
Collapse
|
147
|
Šale S, Lafkas D, Artavanis-Tsakonas S. Notch2 genetic fate mapping reveals two previously unrecognized mammary epithelial lineages. Nat Cell Biol 2013; 15:451-60. [PMID: 23604318 DOI: 10.1038/ncb2725] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Accepted: 03/06/2013] [Indexed: 12/20/2022]
Abstract
Notch signalling is implicated in stem and progenitor cell fate control in numerous organs. Using conditional in vivo genetic labelling we traced the fate of cells expressing the Notch2 receptor paralogue and uncovered the existence of two previously unrecognized mammary epithelial cell lineages that we term S (Small) and L (Large). S cells appear in a bead-on-a-string formation and are embedded between the luminal and basal/myoepithelial layers in a unique reiterative pattern, whereas single or paired L cells appear among ductal and alveolar cells. Long-term lineage tracing and functional studies indicate that S and L cells regulate ipsi- and contralateral spatial placement of tertiary branches and formation of alveolar clusters. Our findings revise present models of mammary epithelial cell hierarchy, reveal a hitherto undescribed mechanism regulating branching morphogenesis and may have important implications for identification of the cell-of-origin of distinct breast cancer subtypes.
Collapse
Affiliation(s)
- Sanja Šale
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | |
Collapse
|
148
|
Expression and functional role of sprouty-2 in breast morphogenesis. PLoS One 2013; 8:e60798. [PMID: 23573284 PMCID: PMC3616012 DOI: 10.1371/journal.pone.0060798] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2012] [Accepted: 03/03/2013] [Indexed: 12/26/2022] Open
Abstract
Branching morphogenesis is a mechanism used by many species for organogenesis and tissue maintenance. Receptor tyrosine kinases (RTKs), including epidermal growth factor receptor (EGFR) and the sprouty protein family are believed to be critical regulators of branching morphogenesis. The aim of this study was to analyze the expression of Sprouty-2 (SPRY2) in the mammary gland and study its role in branching morphogenesis. Human breast epithelial cells, breast tissue and mouse mammary glands were used for expression studies using immunoblotting, real rime PCR and immunohistochemistry. Knockdown of SPRY2 in the breast epithelial stem cell line D492 was done by lentiviral transduction of shRNA constructs targeting SPRY2. Three dimensional culture of D492 with or without endothelial cells was done in reconstituted basement membrane matrix. We show that in the human breast, SPRY2 is predominantly expressed in the luminal epithelial cells of both ducts and lobuli. In the mouse mammary gland, SPRY2 expression is low or absent in the virgin state, while in the pregnant mammary gland SPRY2 is expressed at branching epithelial buds with increased expression during lactation. This expression pattern is closely associated with the activation of the EGFR pathway. Using D492 which generates branching structures in three-dimensional (3D) culture, we show that SPRY2 expression is low during initiation of branching with subsequent increase throughout the branching process. Immunostaining locates expression of phosphorylated SPRY2 and EGFR at the tip of lobular-like, branching ends. SPRY2 knockdown (KD) resulted in increased migration, increased pERK and larger and more complex branching structures indicating a loss of negative feedback control during branching morphogenesis. In D492 co-cultures with endothelial cells, D492 SPRY2 KD generates spindle-like colonies that bear hallmarks of epithelial to mesenchymal transition. These data indicate that SPRY2 is an important regulator of branching morphogenesis and epithelial to mesenchymal transition in the mammary gland.
Collapse
|
149
|
Yoshioka T, Otero J, Chen Y, Kim YM, Koutcher JA, Satagopan J, Reuter V, Carver B, de Stanchina E, Enomoto K, Greenberg NM, Scardino PT, Scher HI, Sawyers CL, Giancotti FG. β4 Integrin signaling induces expansion of prostate tumor progenitors. J Clin Invest 2013; 123:682-99. [PMID: 23348745 PMCID: PMC3561800 DOI: 10.1172/jci60720] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2012] [Accepted: 10/25/2012] [Indexed: 02/03/2023] Open
Abstract
The contextual signals that regulate the expansion of prostate tumor progenitor cells are poorly defined. We found that a significant fraction of advanced human prostate cancers and castration-resistant metastases express high levels of the β4 integrin, which binds to laminin-5. Targeted deletion of the signaling domain of β4 inhibited prostate tumor growth and progression in response to loss of p53 and Rb function in a mouse model of prostate cancer (PB-TAg mice). Additionally, it suppressed Pten loss-driven prostate tumorigenesis in tissue recombination experiments. We traced this defect back to an inability of signaling-defective β4 to sustain self-renewal of putative cancer stem cells in vitro and proliferation of transit-amplifying cells in vivo. Mechanistic studies indicated that mutant β4 fails to promote transactivation of ErbB2 and c-Met in prostate tumor progenitor cells and human cancer cell lines. Pharmacological inhibition of ErbB2 and c-Met reduced the ability of prostate tumor progenitor cells to undergo self-renewal in vitro. Finally, we found that β4 is often coexpressed with c-Met and ErbB2 in human prostate cancers and that combined pharmacological inhibition of these receptor tyrosine kinases exerts antitumor activity in a mouse xenograft model. These findings indicate that the β4 integrin promotes prostate tumorigenesis by amplifying ErbB2 and c-Met signaling in tumor progenitor cells.
Collapse
Affiliation(s)
- Toshiaki Yoshioka
- Cell Biology Program, Sloan-Kettering Institute for Cancer Research, Memorial Sloan-Kettering Cancer Center (MSKCC), New York, New York, USA.
Departments of Molecular Pathology and Tumor Pathology, Akita University Graduate School of Medicine, Akita, Japan.
Human Oncology and Pathogenesis Program,
Department of Medicine,
Department of Medical Physics,
Department of Epidemiology and Biostatistics, and
Department of Pathology, Memorial Hospital, MSKCC, New York, New York, USA.
Antitumor Assessment Core, MSKCC, New York, New York, USA.
Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.
Department of Surgery, Memorial Hospital, MSKCC, New York, New York, USA
| | - Javier Otero
- Cell Biology Program, Sloan-Kettering Institute for Cancer Research, Memorial Sloan-Kettering Cancer Center (MSKCC), New York, New York, USA.
Departments of Molecular Pathology and Tumor Pathology, Akita University Graduate School of Medicine, Akita, Japan.
Human Oncology and Pathogenesis Program,
Department of Medicine,
Department of Medical Physics,
Department of Epidemiology and Biostatistics, and
Department of Pathology, Memorial Hospital, MSKCC, New York, New York, USA.
Antitumor Assessment Core, MSKCC, New York, New York, USA.
Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.
Department of Surgery, Memorial Hospital, MSKCC, New York, New York, USA
| | - Yu Chen
- Cell Biology Program, Sloan-Kettering Institute for Cancer Research, Memorial Sloan-Kettering Cancer Center (MSKCC), New York, New York, USA.
Departments of Molecular Pathology and Tumor Pathology, Akita University Graduate School of Medicine, Akita, Japan.
Human Oncology and Pathogenesis Program,
Department of Medicine,
Department of Medical Physics,
Department of Epidemiology and Biostatistics, and
Department of Pathology, Memorial Hospital, MSKCC, New York, New York, USA.
Antitumor Assessment Core, MSKCC, New York, New York, USA.
Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.
Department of Surgery, Memorial Hospital, MSKCC, New York, New York, USA
| | - Young-Mi Kim
- Cell Biology Program, Sloan-Kettering Institute for Cancer Research, Memorial Sloan-Kettering Cancer Center (MSKCC), New York, New York, USA.
Departments of Molecular Pathology and Tumor Pathology, Akita University Graduate School of Medicine, Akita, Japan.
Human Oncology and Pathogenesis Program,
Department of Medicine,
Department of Medical Physics,
Department of Epidemiology and Biostatistics, and
Department of Pathology, Memorial Hospital, MSKCC, New York, New York, USA.
Antitumor Assessment Core, MSKCC, New York, New York, USA.
Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.
Department of Surgery, Memorial Hospital, MSKCC, New York, New York, USA
| | - Jason A. Koutcher
- Cell Biology Program, Sloan-Kettering Institute for Cancer Research, Memorial Sloan-Kettering Cancer Center (MSKCC), New York, New York, USA.
Departments of Molecular Pathology and Tumor Pathology, Akita University Graduate School of Medicine, Akita, Japan.
Human Oncology and Pathogenesis Program,
Department of Medicine,
Department of Medical Physics,
Department of Epidemiology and Biostatistics, and
Department of Pathology, Memorial Hospital, MSKCC, New York, New York, USA.
Antitumor Assessment Core, MSKCC, New York, New York, USA.
Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.
Department of Surgery, Memorial Hospital, MSKCC, New York, New York, USA
| | - Jaya Satagopan
- Cell Biology Program, Sloan-Kettering Institute for Cancer Research, Memorial Sloan-Kettering Cancer Center (MSKCC), New York, New York, USA.
Departments of Molecular Pathology and Tumor Pathology, Akita University Graduate School of Medicine, Akita, Japan.
Human Oncology and Pathogenesis Program,
Department of Medicine,
Department of Medical Physics,
Department of Epidemiology and Biostatistics, and
Department of Pathology, Memorial Hospital, MSKCC, New York, New York, USA.
Antitumor Assessment Core, MSKCC, New York, New York, USA.
Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.
Department of Surgery, Memorial Hospital, MSKCC, New York, New York, USA
| | - Victor Reuter
- Cell Biology Program, Sloan-Kettering Institute for Cancer Research, Memorial Sloan-Kettering Cancer Center (MSKCC), New York, New York, USA.
Departments of Molecular Pathology and Tumor Pathology, Akita University Graduate School of Medicine, Akita, Japan.
Human Oncology and Pathogenesis Program,
Department of Medicine,
Department of Medical Physics,
Department of Epidemiology and Biostatistics, and
Department of Pathology, Memorial Hospital, MSKCC, New York, New York, USA.
Antitumor Assessment Core, MSKCC, New York, New York, USA.
Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.
Department of Surgery, Memorial Hospital, MSKCC, New York, New York, USA
| | - Brett Carver
- Cell Biology Program, Sloan-Kettering Institute for Cancer Research, Memorial Sloan-Kettering Cancer Center (MSKCC), New York, New York, USA.
Departments of Molecular Pathology and Tumor Pathology, Akita University Graduate School of Medicine, Akita, Japan.
Human Oncology and Pathogenesis Program,
Department of Medicine,
Department of Medical Physics,
Department of Epidemiology and Biostatistics, and
Department of Pathology, Memorial Hospital, MSKCC, New York, New York, USA.
Antitumor Assessment Core, MSKCC, New York, New York, USA.
Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.
Department of Surgery, Memorial Hospital, MSKCC, New York, New York, USA
| | - Elisa de Stanchina
- Cell Biology Program, Sloan-Kettering Institute for Cancer Research, Memorial Sloan-Kettering Cancer Center (MSKCC), New York, New York, USA.
Departments of Molecular Pathology and Tumor Pathology, Akita University Graduate School of Medicine, Akita, Japan.
Human Oncology and Pathogenesis Program,
Department of Medicine,
Department of Medical Physics,
Department of Epidemiology and Biostatistics, and
Department of Pathology, Memorial Hospital, MSKCC, New York, New York, USA.
Antitumor Assessment Core, MSKCC, New York, New York, USA.
Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.
Department of Surgery, Memorial Hospital, MSKCC, New York, New York, USA
| | - Katsuhiko Enomoto
- Cell Biology Program, Sloan-Kettering Institute for Cancer Research, Memorial Sloan-Kettering Cancer Center (MSKCC), New York, New York, USA.
Departments of Molecular Pathology and Tumor Pathology, Akita University Graduate School of Medicine, Akita, Japan.
Human Oncology and Pathogenesis Program,
Department of Medicine,
Department of Medical Physics,
Department of Epidemiology and Biostatistics, and
Department of Pathology, Memorial Hospital, MSKCC, New York, New York, USA.
Antitumor Assessment Core, MSKCC, New York, New York, USA.
Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.
Department of Surgery, Memorial Hospital, MSKCC, New York, New York, USA
| | - Norman M. Greenberg
- Cell Biology Program, Sloan-Kettering Institute for Cancer Research, Memorial Sloan-Kettering Cancer Center (MSKCC), New York, New York, USA.
Departments of Molecular Pathology and Tumor Pathology, Akita University Graduate School of Medicine, Akita, Japan.
Human Oncology and Pathogenesis Program,
Department of Medicine,
Department of Medical Physics,
Department of Epidemiology and Biostatistics, and
Department of Pathology, Memorial Hospital, MSKCC, New York, New York, USA.
Antitumor Assessment Core, MSKCC, New York, New York, USA.
Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.
Department of Surgery, Memorial Hospital, MSKCC, New York, New York, USA
| | - Peter T. Scardino
- Cell Biology Program, Sloan-Kettering Institute for Cancer Research, Memorial Sloan-Kettering Cancer Center (MSKCC), New York, New York, USA.
Departments of Molecular Pathology and Tumor Pathology, Akita University Graduate School of Medicine, Akita, Japan.
Human Oncology and Pathogenesis Program,
Department of Medicine,
Department of Medical Physics,
Department of Epidemiology and Biostatistics, and
Department of Pathology, Memorial Hospital, MSKCC, New York, New York, USA.
Antitumor Assessment Core, MSKCC, New York, New York, USA.
Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.
Department of Surgery, Memorial Hospital, MSKCC, New York, New York, USA
| | - Howard I. Scher
- Cell Biology Program, Sloan-Kettering Institute for Cancer Research, Memorial Sloan-Kettering Cancer Center (MSKCC), New York, New York, USA.
Departments of Molecular Pathology and Tumor Pathology, Akita University Graduate School of Medicine, Akita, Japan.
Human Oncology and Pathogenesis Program,
Department of Medicine,
Department of Medical Physics,
Department of Epidemiology and Biostatistics, and
Department of Pathology, Memorial Hospital, MSKCC, New York, New York, USA.
Antitumor Assessment Core, MSKCC, New York, New York, USA.
Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.
Department of Surgery, Memorial Hospital, MSKCC, New York, New York, USA
| | - Charles L. Sawyers
- Cell Biology Program, Sloan-Kettering Institute for Cancer Research, Memorial Sloan-Kettering Cancer Center (MSKCC), New York, New York, USA.
Departments of Molecular Pathology and Tumor Pathology, Akita University Graduate School of Medicine, Akita, Japan.
Human Oncology and Pathogenesis Program,
Department of Medicine,
Department of Medical Physics,
Department of Epidemiology and Biostatistics, and
Department of Pathology, Memorial Hospital, MSKCC, New York, New York, USA.
Antitumor Assessment Core, MSKCC, New York, New York, USA.
Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.
Department of Surgery, Memorial Hospital, MSKCC, New York, New York, USA
| | - Filippo G. Giancotti
- Cell Biology Program, Sloan-Kettering Institute for Cancer Research, Memorial Sloan-Kettering Cancer Center (MSKCC), New York, New York, USA.
Departments of Molecular Pathology and Tumor Pathology, Akita University Graduate School of Medicine, Akita, Japan.
Human Oncology and Pathogenesis Program,
Department of Medicine,
Department of Medical Physics,
Department of Epidemiology and Biostatistics, and
Department of Pathology, Memorial Hospital, MSKCC, New York, New York, USA.
Antitumor Assessment Core, MSKCC, New York, New York, USA.
Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.
Department of Surgery, Memorial Hospital, MSKCC, New York, New York, USA
| |
Collapse
|
150
|
Abstract
Mammary glands are crucial to the reproductive strategy of mammals, and the milk of domesticated ruminants serves as an important source of nutrients for the human population. The majority of mammary gland development occurs postnatally, and the mammary gland undergoes cyclical periods of growth, differentiation, lactation, and regression that are coordinated to provide nutrients for offspring or are driven by strategies to manage reproduction and milk production of domesticated species. Growth and maintenance of the mammary epithelium depends on the function of mammary stem cells and progenitor cells. In this review, we provide an overview of postnatal mammary gland development, cyclical phases of mammary gland regression (regression during lactation and between successive lactations), and mammary stem cells and progenitor cells. Where possible, these processes are related to animal production and compared across species, particularly bovine, porcine, murine, and human.
Collapse
Affiliation(s)
- Anthony V Capuco
- Bovine Functional Genomics Laboratory, US Department of Agriculture, Agricultural Research Service, Beltsville, Maryland 20705;
| | | |
Collapse
|