Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
Breast Cancer Res ; 23(1): 66, 2021 06 13.
Article in English | MEDLINE | ID: mdl-34120626

ABSTRACT

BACKGROUND: Normal human breast tissues are a heterogeneous mix of epithelial and stromal subtypes in different cell states. Delineating the spectrum of cellular heterogeneity will provide new insights into normal cellular properties within the breast tissue that might become dysregulated in the initial stages of cancer. Investigation of surface marker expression provides a valuable approach to resolve complex cell populations. However, the majority of cell surface maker expression of primary breast cells have not been investigated. METHODS: To determine the differences in expression of a range of uninvestigated cell surface markers between the normal breast cell subpopulations, primary human breast cells were analysed using high-throughput flow cytometry for the expression of 242 cell surface proteins in conjunction with EpCAM/CD49f staining. RESULTS: We identified 35 surface marker proteins expressed on normal breast epithelial and/or stromal subpopulations that were previously unreported. We also show multiple markers were equally expressed in all cell populations (e.g. CD9, CD59, CD164) while other surface markers were confirmed to be enriched in different cell lineages: CD24, CD227 and CD340 in the luminal compartment, CD10 and CD90 in the basal population, and CD34 and CD140b on stromal cells. CONCLUSIONS: Our dataset of CD marker expression in the normal breast provides better definition for breast cellular heterogeneity.


Subject(s)
Breast/metabolism , Membrane Proteins/metabolism , Biomarkers/metabolism , Breast/cytology , Cells, Cultured , Epithelial Cells/metabolism , Female , Humans , Stem Cells/metabolism , Stromal Cells/metabolism
2.
Methods Mol Biol ; 1501: 261-276, 2017.
Article in English | MEDLINE | ID: mdl-27796958

ABSTRACT

The mammary epithelium is composed of a variety of specialized cell types that function in a coordinated fashion to produce and eject milk through multiple cycles of pregnancy. The ability to identify and purify these subsets of cells in order to interrogate their growth and differentiation capacities, as well as to characterize the molecular mechanisms that regulate their behavior, is essential in identifying the processes associated with breast cancer initiation and progression. This methods chapter outlines the step-by-step methods for dissociating human breast reduction specimens to a single cell suspension of viable cells. As well, strategies for purifying four distinct subsets of epithelial cells by using fluorescence-activated cell sorting and protocols for interrogating the growth and differentiation properties of these purified cells at clonal densities in adherent culture are also described.


Subject(s)
Breast/cytology , Epithelial Cells/cytology , Mammary Glands, Human/cytology , Animals , Breast Neoplasms/pathology , Cell Culture Techniques/methods , Cell Differentiation/physiology , Cell Proliferation/physiology , Cells, Cultured , Epithelium/physiology , Female , Humans , Mice , NIH 3T3 Cells
3.
Nat Commun ; 6: 8487, 2015 Oct 29.
Article in English | MEDLINE | ID: mdl-26511661

ABSTRACT

The cycling properties of mammary stem and progenitor cells is not well understood. To determine the division properties of these cells, we administered synthetic nucleosides for varying periods of time to mice at different stages of postnatal development and monitored the rate of uptake of these nucleosides in the different mammary cell compartments. Here we show that most cell division in the adult virgin gland is restricted to the oestrogen receptor-expressing luminal cell lineage. Our data also demonstrate that the oestrogen receptor-expressing, milk and basal cell subpopulations have telomere lengths and cell division kinetics that are not compatible with these cells being hierarchically organized; instead, our data indicate that in the adult homeostatic gland, each cell type is largely maintained by its own restricted progenitors. We also observe that transplantable stem cells are largely quiescent during oestrus, but are cycling during dioestrus when progesterone levels are high.


Subject(s)
Cell Self Renewal , Mammary Glands, Animal/growth & development , Stem Cells/cytology , Animals , Female , Kinetics , Mammary Glands, Animal/chemistry , Mammary Glands, Animal/cytology , Mammary Glands, Animal/metabolism , Mice , Mice, Inbred C57BL , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Stem Cells/chemistry , Stem Cells/metabolism
5.
Nature ; 523(7560): 313-7, 2015 Jul 16.
Article in English | MEDLINE | ID: mdl-26153859

ABSTRACT

Progesterone receptor (PR) expression is used as a biomarker of oestrogen receptor-α (ERα) function and breast cancer prognosis. Here we show that PR is not merely an ERα-induced gene target, but is also an ERα-associated protein that modulates its behaviour. In the presence of agonist ligands, PR associates with ERα to direct ERα chromatin binding events within breast cancer cells, resulting in a unique gene expression programme that is associated with good clinical outcome. Progesterone inhibited oestrogen-mediated growth of ERα(+) cell line xenografts and primary ERα(+) breast tumour explants, and had increased anti-proliferative effects when coupled with an ERα antagonist. Copy number loss of PGR, the gene coding for PR, is a common feature in ERα(+) breast cancers, explaining lower PR levels in a subset of cases. Our findings indicate that PR functions as a molecular rheostat to control ERα chromatin binding and transcriptional activity, which has important implications for prognosis and therapeutic interventions.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Estrogen Receptor alpha/metabolism , Gene Expression Regulation, Neoplastic , Receptors, Progesterone/metabolism , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Chromatin/drug effects , Chromatin/genetics , Chromatin/metabolism , DNA Copy Number Variations/genetics , Disease Progression , Estrogen Receptor alpha/antagonists & inhibitors , Estrogens/metabolism , Estrogens/pharmacology , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Ligands , Mice , Progesterone/metabolism , Progesterone/pharmacology , Protein Binding/drug effects , Receptors, Progesterone/genetics , Transcription, Genetic/drug effects , Xenograft Model Antitumor Assays
6.
Cell Stem Cell ; 16(6): 712-24, 2015 Jun 04.
Article in English | MEDLINE | ID: mdl-26004780

ABSTRACT

Heterogeneity within the self-renewal durability of adult hematopoietic stem cells (HSCs) challenges our understanding of the molecular framework underlying HSC function. Gene expression studies have been hampered by the presence of multiple HSC subtypes and contaminating non-HSCs in bulk HSC populations. To gain deeper insight into the gene expression program of murine HSCs, we combined single-cell functional assays with flow cytometric index sorting and single-cell gene expression assays. Through bioinformatic integration of these datasets, we designed an unbiased sorting strategy that separates non-HSCs away from HSCs, and single-cell transplantation experiments using the enriched population were combined with RNA-seq data to identify key molecules that associate with long-term durable self-renewal, producing a single-cell molecular dataset that is linked to functional stem cell activity. Finally, we demonstrated the broader applicability of this approach for linking key molecules with defined cellular functions in another stem cell system.


Subject(s)
Gene Expression Regulation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Single-Cell Analysis/methods , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Cell Proliferation , Clone Cells , Gene Expression Profiling , Genome , Hematopoietic Stem Cell Transplantation , Humans , Mice, Inbred C57BL
7.
Nat Commun ; 6: 5987, 2015 Jan 09.
Article in English | MEDLINE | ID: mdl-25574598

ABSTRACT

Triple-negative breast cancer (TNBC) has poor prognostic outcome compared with other types of breast cancer. The molecular and cellular mechanisms underlying TNBC pathology are not fully understood. Here, we report that the transcription factor BCL11A is overexpressed in TNBC including basal-like breast cancer (BLBC) and that its genomic locus is amplified in up to 38% of BLBC tumours. Exogenous BCL11A overexpression promotes tumour formation, whereas its knockdown in TNBC cell lines suppresses their tumourigenic potential in xenograft models. In the DMBA-induced tumour model, Bcl11a deletion substantially decreases tumour formation, even in p53-null cells and inactivation of Bcl11a in established tumours causes their regression. At the cellular level, Bcl11a deletion causes a reduction in the number of mammary epithelial stem and progenitor cells. Thus, BCL11A has an important role in TNBC and normal mammary epithelial cells. This study highlights the importance of further investigation of BCL11A in TNBC-targeted therapies.


Subject(s)
Carrier Proteins/metabolism , Gene Expression Regulation, Neoplastic , Nuclear Proteins/metabolism , Stem Cells/metabolism , Triple Negative Breast Neoplasms/diagnosis , Triple Negative Breast Neoplasms/metabolism , 9,10-Dimethyl-1,2-benzanthracene/chemistry , Animals , Cell Line, Tumor , Cell Proliferation , Cell Survival , DNA-Binding Proteins , Female , Humans , Immunohistochemistry , Mammary Glands, Animal/metabolism , Mice , Neoplasm Transplantation , Oligonucleotide Array Sequence Analysis , Prognosis , Repressor Proteins
8.
Gut ; 64(1): 11-9, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24572143

ABSTRACT

OBJECTIVE: Knowledge of the cellular mechanisms involved in homeostasis of human squamous oesophagus in the steady state and following chronic injury is limited. We aimed to better understand these mechanisms by using a functional 3D approach. DESIGN: Proliferation, mitosis and the expression of progenitor lineage markers were assessed in normal squamous oesophagus from 10 patients by immunofluorescence on 3D epithelial whole mounts. Cells expressing differential levels of epithelial and progenitor markers were isolated using flow cytometry sorting and characterised by qPCR and IF. Their self-renewing potential was investigated by colony forming cells assays and in vitro organotypic culture models. RESULTS: Proliferation and mitotic activity was highest in the interpapillary basal layer and decreased linearly towards the tip of the papilla (p<0.0001). The orientation of mitosis was random throughout the basal layer, and asymmetric divisions were not restricted to specific cell compartments. Cells sorted into distinct populations based on the expression of epithelial and progenitor cell markers (CD34 and EpCAM) showed no difference in self-renewal in 2D culture, either as whole populations or as single cells. In 3D organotypic cultures, all cell subtypes were able to recapitulate the architecture of the tissue of origin and the main factor determining the success of the 3D culture was the number of cells plated, rather than the cell type. CONCLUSIONS: Oesophageal epithelial cells demonstrate remarkable plasticity for self-renewal. This situation could be viewed as an ex vivo wounding response and is compatible with recent findings in murine models.


Subject(s)
Cell Differentiation , Epithelial Cells/physiology , Esophagus/cytology , Cell Proliferation , Cells, Cultured , Clone Cells , Humans , Mitosis
9.
Nat Cell Biol ; 16(10): 942-50, 1-7, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25173976

ABSTRACT

Contractile myoepithelial cells dominate the basal layer of the mammary epithelium and are considered to be differentiated cells. However, we observe that up to 54% of single basal cells can form colonies when seeded into adherent culture in the presence of agents that disrupt actin-myosin interactions, and on average, 65% of the single-cell-derived basal colonies can repopulate a mammary gland when transplanted in vivo. This indicates that a high proportion of basal myoepithelial cells can give rise to a mammary repopulating unit (MRU). We demonstrate that myoepithelial cells, flow-sorted using two independent myoepithelial-specific reporter strategies, have MRU capacity. Using an inducible lineage-tracing approach we follow the progeny of myoepithelial cells that express α-smooth muscle actin and show that they function as long-lived lineage-restricted stem cells in the virgin state and during pregnancy.


Subject(s)
Epithelial Cells/cytology , Mammary Glands, Animal/cytology , Myocytes, Smooth Muscle/metabolism , Stem Cells/cytology , Actins/metabolism , Animals , Benzamides/pharmacology , Cell Proliferation/drug effects , Cells, Cultured , Dioxoles/pharmacology , Epithelial Cells/metabolism , Female , Flow Cytometry , Gene Expression Profiling , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Interleukin Receptor Common gamma Subunit/deficiency , Interleukin Receptor Common gamma Subunit/genetics , Mammary Glands, Animal/metabolism , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Microscopy, Fluorescence , Myocytes, Smooth Muscle/cytology , Oligonucleotide Array Sequence Analysis , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/metabolism
10.
Breast Cancer Res ; 16(4): 411, 2014 Jul 24.
Article in English | MEDLINE | ID: mdl-25056669

ABSTRACT

INTRODUCTION: Lineage tracing using inducible genetic labeling has emerged to be a powerful method for interrogating the developmental fate of cells in intact tissues. A common induction mechanism is the use of tamoxifen-dependent Cre recombinase (CreER and CreERT2), but the effects of tamoxifen at doses normally used in lineage-tracing studies on normal adult mammary gland homeostasis are not known. METHODS: We used flow cytometry and immunostaining of intact glands to determine whether varying doses of tamoxifen skew the distribution and the apoptosis and proliferation status of different types of mammary epithelial cells in vivo. We also examined how tamoxifen influences the number of progenitor and mammary repopulating units (MRUs). RESULTS: Our results indicate that ≥5 mg/25 g body weight of tamoxifen induces a transient increase in cell proliferation and in the number of basal cells in the adult mammary epithelium up to 7 days after tamoxifen administration. However, in the medium term (3 weeks), all doses of tamoxifen≥1 mg/25 g body weight result in a decrease in the number of basal and EpCAM+CD49b- luminal cells and a decrease in progenitor cell function. Tamoxifen at doses≥5 mg/25 g body weight induced a transient increase in caspase-3-mediated apoptotic cell death within the mammary epithelium. However, mammary epithelial cell numbers in all subpopulations were restored to their original levels by 8 weeks. No long-lasting effects of tamoxifen on MRU numbers or on pubertal ductal development were observed. CONCLUSION: Tamoxifen can skew the distribution of mammary cell types in a dose-dependent manner, and thus caution must be taken when interpreting lineage-tracing studies using high doses of tamoxifen, particularly when short-duration analyses of a quantitative nature are being performed.


Subject(s)
Homeostasis/drug effects , Mammary Glands, Animal/drug effects , Mammary Glands, Animal/physiology , Tamoxifen/pharmacology , Animals , Cell Proliferation , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Female , Mice , Sexual Maturation/drug effects , Tamoxifen/administration & dosage
11.
Genes Dev ; 28(11): 1143-58, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24888586

ABSTRACT

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.


Subject(s)
Cell Differentiation , Mammary Glands, Animal/cytology , Mammary Glands, Human/cytology , Stem Cells/cytology , Animals , Cell Lineage , Female , Humans , Stem Cell Transplantation
12.
Breast Cancer Res ; 15(5): 313, 2013.
Article in English | MEDLINE | ID: mdl-24103450

ABSTRACT

The European Network for Breast Development and Cancer (ENBDC) Workshop on 'Methods in Mammary Gland Development and Cancer' has grown into the essential, international technical discussion forum for scientists with interests in the normal and neoplastic breast. The fifth ENBDC meeting was held in Weggis, Switzerland in April, 2013, and focussed on emerging, state-of-the-art techniques for the study of non-coding RNA, lineage tracing, tumor heterogeneity, metastasis and metabolism.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Lineage , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , RNA, Untranslated , Animals , Breast Neoplasms/pathology , Female , Humans , Neoplasm Metastasis
13.
PLoS Biol ; 11(8): e1001630, 2013.
Article in English | MEDLINE | ID: mdl-23966837

ABSTRACT

Many normal adult tissues contain rare stem cells with extensive self-maintaining regenerative potential. During development, the stem cells of the hematopoietic and neural systems undergo intrinsically specified changes in their self-renewal potential. In the mouse, mammary stem cells with transplantable regenerative activity are first detectable a few days before birth. They share some phenotypic properties with their adult counterparts but are enriched in a subpopulation that displays a distinct gene expression profile. Here we show that fetal mammary epithelial cells have a greater direct and inducible growth potential than their adult counterparts. The latter feature is revealed in a novel culture system that enables large numbers of in vitro clonogenic progenitors as well as mammary stem cells with serially transplantable activity to be produced within 7 days from single fetal or adult input cells. We further show that these responses are highly dependent on novel factors produced by fibroblasts. These findings provide new avenues for elucidating mechanisms that regulate normal mammary epithelial stem cell properties at the single-cell level, how these change during development, and how their perturbation may contribute to transformation.


Subject(s)
Epithelial Cells/cytology , Mammary Glands, Animal/cytology , 3T3 Cells , Animals , Epithelial Cells/physiology , Female , Immunohistochemistry , Mammary Glands, Animal/physiology , Mice
15.
Cell Rep ; 3(2): 342-9, 2013 Feb 21.
Article in English | MEDLINE | ID: mdl-23403292

ABSTRACT

Estrogen receptor-α (ER) is the driving transcription factor in most breast cancers, and its associated proteins can influence drug response, but direct methods for identifying interacting proteins have been limited. We purified endogenous ER using an approach termed RIME (rapid immunoprecipitation mass spectrometry of endogenous proteins) and discovered the interactome under agonist- and antagonist-liganded conditions in breast cancer cells, revealing transcriptional networks in breast cancer. The most estrogen-enriched ER interactor is GREB1, a potential clinical biomarker with no known function. GREB1 is shown to be a chromatin-bound ER coactivator and is essential for ER-mediated transcription, because it stabilizes interactions between ER and additional cofactors. We show a GREB1-ER interaction in three xenograft tumors, and using a directed protein-protein approach, we find GREB1-ER interactions in half of ER(+) primary breast cancers. This finding is supported by histological expression of GREB1, which shows that GREB1 is expressed in half of ER(+) cancers, and predicts good clinical outcome. These findings reveal an unexpected role for GREB1 as an estrogen-specific ER cofactor that is expressed in drug-sensitive contexts.


Subject(s)
Estrogen Receptor alpha/metabolism , Neoplasm Proteins/metabolism , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Chromatin/metabolism , Chromatin Immunoprecipitation , Female , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Mice , Mice, SCID , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Protein Interaction Maps , RNA Interference , RNA, Small Interfering/metabolism , Transcription, Genetic , Transplantation, Heterologous
16.
Development ; 140(5): 1079-89, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23404106

ABSTRACT

Nuclear receptor interacting protein (Nrip1), also known as RIP140, is a co-regulator for nuclear receptors that plays an essential role in ovulation by regulating the expression of the epidermal growth factor-like family of growth factors. Although several studies indicate a role for RIP140 in breast cancer, its role in the development of the mammary gland is unclear. By using RIP140-null and RIP140 transgenic mice, we demonstrate that RIP140 is an essential factor for normal mammary gland development and that it functions by mediating oestrogen signalling. RIP140-null mice exhibit minimal ductal elongation with no side-branching, whereas RIP140-overexpressing mice show increased cell proliferation and ductal branching with age. Tissue recombination experiments demonstrate that RIP140 expression is required in both the mammary epithelial and stromal compartments for ductal elongation during puberty and that loss of RIP140 leads to a catastrophic loss of the mammary epithelium, whereas RIP140 overexpression augments the mammary basal cell population and shifts the progenitor/differentiated cell balance within the luminal cell compartment towards the progenitors. For the first time, we present a genome-wide global view of oestrogen receptor-α (ERα) binding events in the developing mammary gland, which unravels 881 ERα binding sites. Unbiased evaluation of several ERα binding sites for RIP140 co-occupancy reveals selectivity and demonstrates that RIP140 acts as a co-regulator with ERα to regulate directly the expression of amphiregulin (Areg), the progesterone receptor (Pgr) and signal transducer and activator of transcription 5a (Stat5a), factors that influence key mitogenic pathways that regulate normal mammary gland development.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Growth Substances/genetics , Mammary Glands, Animal/growth & development , Nuclear Proteins/physiology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cells, Cultured , Estradiol/pharmacology , Female , Gene Expression Regulation, Developmental/drug effects , Growth Substances/metabolism , Mammary Glands, Animal/drug effects , Mammary Glands, Animal/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Morphogenesis/drug effects , Morphogenesis/genetics , Morphogenesis/physiology , NIH 3T3 Cells , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nuclear Receptor Interacting Protein 1 , Signal Transduction/drug effects , Signal Transduction/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription Factors/physiology
17.
Methods Mol Biol ; 946: 395-409, 2013.
Article in English | MEDLINE | ID: mdl-23179846

ABSTRACT

Evidence is emerging that the mouse mammary epithelium is arranged as a hierarchy that spans from stem cells to lineage-restricted progenitor cells to differentiated luminal and myoepithelial cells. The use of fluorescence-activated cell sorting (FACS) in combination with quantitative functional clonal assays represents a powerful tool for studying the properties of mouse mammary stem and progenitor cells. This chapter outlines the experimental procedures for generating single viable cell suspensions of mouse mammary epithelial cells, immunostaining cells for flow cytometry, in vitro assays for the detection and enumeration of mouse mammary progenitor cells, and in vivo assays for the detection and enumeration of mouse mammary stem cells.


Subject(s)
Cell Culture Techniques/methods , Collagenases/metabolism , Flow Cytometry/methods , Hyaluronoglucosaminidase/metabolism , Mammary Glands, Animal/cytology , Mammary Glands, Animal/metabolism , Animals , Colony-Forming Units Assay , Female , Mice , NIH 3T3 Cells , Single-Cell Analysis , Stem Cell Transplantation , Stem Cells/cytology , Stromal Cells/cytology , Suspensions
18.
Breast Cancer Res ; 14(5): R134, 2012 Oct 22.
Article in English | MEDLINE | ID: mdl-23088371

ABSTRACT

INTRODUCTION: The organisation of the mammary epithelial hierarchy is poorly understood. Our hypothesis is that the luminal cell compartment is more complex than initially described, and that an understanding of the developmental relationships within this lineage will help in understanding the cellular context in which breast tumours occur. METHODS: We used fluorescence-activated cell sorting along with in vitro and in vivo functional assays to examine the growth and differentiation properties of distinct subsets of human and mouse mammary epithelial cells. We also examined how loss of steroid hormones influenced these populations in vivo. Gene expression profiles were also obtained for all the purified cell populations and correlated to those obtained from breast tumours. RESULTS: The luminal cell compartment of the mouse mammary gland can be resolved into nonclonogenic oestrogen receptor-positive (ER+) luminal cells, ER+ luminal progenitors and oestrogen receptor-negative (ER-) luminal progenitors. The ER+ luminal progenitors are unique in regard to cell survival, as they are relatively insensitive to loss of oestrogen and progesterone when compared with the other types of mammary epithelial cells. Analysis of normal human breast tissue reveals a similar hierarchical organisation composed of nonclonogenic luminal cells, and relatively differentiated (EpCAM+CD49f+ALDH-) and undifferentiated (EpCAM+CD49f+ALDH+) luminal progenitors. In addition, approximately one-quarter of human breast samples examined contained an additional population that had a distinct luminal progenitor phenotype, characterised by low expression of ERBB3 and low proliferative potential. Parent-progeny relationship experiments demonstrated that all luminal progenitor populations in both species are highly plastic and, at low frequencies, can generate progeny representing all mammary cell types. The ER- luminal progenitors in the mouse and the ALDH+ luminal progenitors in the human appear to be analogous populations since they both have gene signatures that are associated with alveolar differentiation and resemble those obtained from basal-like breast tumours. CONCLUSION: The luminal cell compartment in the mammary epithelium is more heterogeneous than initially perceived since progenitors of varying levels of luminal cell differentiation and proliferative capacities can be identified. An understanding of these cells will be essential for understanding the origins and the cellular context of human breast tumours.


Subject(s)
Mammary Glands, Animal/cytology , Mammary Glands, Animal/metabolism , Mammary Glands, Human/cytology , Mammary Glands, Human/metabolism , Phenotype , Animals , Antigens, Surface/metabolism , Cell Differentiation , Colony-Forming Units Assay , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelium/metabolism , Female , Humans , Immunophenotyping , Mice , Receptors, Estrogen/metabolism , Stem Cells/metabolism
19.
Nat Commun ; 3: 1055, 2012.
Article in English | MEDLINE | ID: mdl-22968701

ABSTRACT

The role of transforming growth factor-beta (TGFß) in the progression of different molecular subtypes of breast cancer has not been clarified. Here we show that TGFß increases breast tumour-initiating cell (BTIC) numbers but only in claudin(low) breast cancer cell lines by orchestrating a specific gene signature enriched in stem cell processes that predicts worse clinical outcome in breast cancer patients. NEDD9, a member of the Cas family of integrin scaffold proteins, is necessary to mediate these TGFß-specific effects through a positive feedback loop that integrates TGFß/Smad and Rho-actin-SRF-dependent signals. In normal human mammary epithelium, TGFß induces progenitor activity only in the basal/stem cell compartment, where claudin(low) cancers are presumed to arise. These data show opposing responses to TGFß in both breast malignant cell subtypes and normal mammary epithelial cell subpopulations and suggest therapeutic strategies for a subset of human breast cancers.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Claudins/metabolism , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/drug effects , Transforming Growth Factor beta/pharmacology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Chromatin Immunoprecipitation , Claudins/genetics , Female , Humans , Mammary Glands, Human/cytology , Mammary Glands, Human/drug effects , Mammary Glands, Human/metabolism , Mice , Neoplastic Stem Cells/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/metabolism , Tumor Cells, Cultured
20.
J Mammary Gland Biol Neoplasia ; 17(2): 91-7, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22644112

ABSTRACT

Isolation of mammary epithelial subpopulations, including stem and progenitor cells, has become a standard technique in recent years. However, a number of methods and approaches for this have developed and the relative benefits of the different approaches, and the reason for their development, have not always been clear. Here, three of the leading laboratories working on the separation of mammary cell subpopulations have summarised their methods, highlighted their differences and similarities and also discussed the reasoning behind the approaches they have taken. This article will assist workers establishing mammary cell separation protocols in their laboratories to make informed choices about the methods they should use.


Subject(s)
Adult Stem Cells/cytology , Cell Separation/methods , Epithelium/metabolism , Mammary Glands, Animal/cytology , Adult Stem Cells/metabolism , Animals , Antibody Specificity , Antigens, Surface/metabolism , Cell Differentiation , Cell Separation/instrumentation , Cells, Cultured , Female , Mammary Glands, Animal/metabolism , Mice , Organoids/cytology , Organoids/metabolism , Staining and Labeling/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...