Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Breast Cancer Res ; 26(1): 11, 2024 01 16.
Article in English | MEDLINE | ID: mdl-38229104

ABSTRACT

BACKGROUND: Human breast cancer most frequently originates within a well-defined anatomical structure referred to as the terminal duct lobular unit (TDLU). This structure is endowed with its very own lobular fibroblasts representing one out of two steady-state fibroblast subtypes-the other being interlobular fibroblasts. While cancer-associated fibroblasts (CAFs) are increasingly appreciated as covering a spectrum of perturbed states, we lack a coherent understanding of their relationship-if any-with the steady-state fibroblast subtypes. To address this, we here established two autologous CAF lines representing inflammatory CAFs (iCAFs) and myofibroblast CAFs (myCAFs) and compared them with already established interlobular- and lobular fibroblasts with respect to their origin and impact on tumor formation. METHODS: Primary breast tumor-derived CAFs were transduced to express human telomerase reverse transcriptase (hTERT) and sorted into CD105low and CD105high populations using fluorescence-activated cell sorting (FACS). The two populations were tested for differentiation similarities to iCAF and myCAF states through transcriptome-wide RNA-Sequencing (RNA-Seq) including comparison to an available iCAF-myCAF cell state atlas. Inference of origin in interlobular and lobular fibroblasts relied on RNA-Seq profiles, immunocytochemistry and growth characteristics. Osteogenic differentiation and bone formation assays in culture and in vivo were employed to gauge for origin in bone marrow-derived mesenchymal stem cells (bMSCs). Functional characteristics were assessed with respect to contractility in culture and interaction with tumor cells in mouse xenografts. The cells' gene expression signatures were tested for association with clinical outcome of breast cancer patients using survival data from The Cancer Genome Atlas database. RESULTS: We demonstrate that iCAFs have properties in common with interlobular fibroblasts while myCAFs and lobular fibroblasts are related. None of the CAFs qualify as bMSCs as revealed by lack of critical performance in bone formation assays. Functionally, myCAFs and lobular fibroblasts are almost equally tumor promoting as opposed to iCAFs and interlobular fibroblasts. A myCAF gene signature is found to associate with poor breast cancer-specific survival. CONCLUSIONS: We propose that iCAFs and myCAFs originate in interlobular and lobular fibroblasts, respectively, and more importantly, that the tumor-promoting properties of lobular fibroblasts render the TDLU an epicenter for breast cancer evolution.


Subject(s)
Breast Neoplasms , Cancer-Associated Fibroblasts , Humans , Mice , Animals , Female , Breast Neoplasms/pathology , Osteogenesis , Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Breast/pathology , Tumor Microenvironment
2.
NPJ Breast Cancer ; 8(1): 81, 2022 Jul 12.
Article in English | MEDLINE | ID: mdl-35821504

ABSTRACT

Normal breast luminal epithelial progenitors have been implicated as cell of origin in basal-like breast cancer, but their anatomical localization remains understudied. Here, we combine collection under the microscope of organoids from reduction mammoplasties and single-cell mRNA sequencing (scRNA-seq) of FACS-sorted luminal epithelial cells with multicolor imaging to profile ducts and terminal duct lobular units (TDLUs) and compare them with breast cancer subtypes. Unsupervised clustering reveals eleven distinct clusters and a differentiation trajectory starting with keratin 15+ (K15+) progenitors enriched in ducts. Spatial mapping of luminal progenitors is confirmed at the protein level by staining with critical duct markers. Comparison of the gene expression profiles of normal luminal cells with those of breast cancer subtypes suggests a strong correlation between normal breast ductal progenitors and basal-like breast cancer. We propose that K15+ basal-like breast cancers originate in ductal progenitors, which emphasizes the importance of not only lineages but also cellular position within the ductal-lobular tree.

3.
Commun Biol ; 5(1): 219, 2022 03 10.
Article in English | MEDLINE | ID: mdl-35273332

ABSTRACT

The myoepithelial (MEP) lineage of human breast comprises bipotent and multipotent progenitors in ducts and terminal duct lobular units (TDLUs). We here assess whether this heterogeneity impacts on oncogenic PIK3CA transformation. Single cell RNA sequencing (scRNA-seq) and multicolor imaging reveal that terminal ducts represent the most enriched source of cells with ductal MEP markers including α-smooth muscle actin (α-SMA), keratin K14, K17 and CD200. Furthermore, we find neighboring CD200high and CD200low progenitors within terminal ducts. When sorted and kept in ground state conditions, their CD200low and CD200high phenotypes are preserved. Upon differentiation, progenitors remain multipotent and bipotent, respectively. Immortalized progenitors are transduced with mutant PIK3CA on an shp53 background. Upon transplantation, CD200low MEP progenitors distinguish from CD200high by the formation of multilayered structures with a hyperplastic inner layer of luminal epithelial cells. We suggest a model with spatially distributed MEP progenitors as founder cells of biphasic breast lesions with implications for early detection and prevention strategies.


Subject(s)
Breast Neoplasms , Neoplastic Stem Cells , Oncogenes , Breast , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Class I Phosphatidylinositol 3-Kinases/genetics , Epithelial Cells/pathology , Female , Humans
4.
Sci Rep ; 11(1): 17232, 2021 08 26.
Article in English | MEDLINE | ID: mdl-34446796

ABSTRACT

Full term pregnancy at an early age is the only factor known to consistently protect against breast cancer. Because hormone receptor positive progenitors in the human breast relay endocrine signaling, we here sought to determine whether an experimental mimicry of the third trimester surge of hormones would change their susceptibility to growth stimulation. Hormone receptor positive, reduction mammoplasty-derived human breast epithelial progenitors were exposed to a short-term, pregnancy-level of estradiol, and their subsequent response to estradiol stimulation was analyzed. Exposure to pregnancy-level of estradiol results in subsequent lower sensitivity to estrogen-induced proliferation. Expression array and immunoblotting reveal upregulation of S100A7 and down-regulation of p27, both associated with parity and epithelial differentiation. Notably, we find that the epithelial differentiation is accompanied by upregulation of E-cadherin and down-regulation of vimentin as well as by diminished migration and more mature luminal epithelial differentiation in a mouse transplantation model. Our findings are in support of a de-sensitization mechanism for pregnancy-induced prevention against breast cancer.


Subject(s)
Breast/drug effects , Estradiol/pharmacology , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Animals , Breast/cytology , Breast/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cadherins/metabolism , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cells, Cultured , Estrogens/pharmacology , Female , Gene Expression/drug effects , Humans , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Pregnancy , Receptors, Estrogen/genetics , Receptors, Progesterone/genetics , S100 Calcium Binding Protein A7/genetics , S100 Calcium Binding Protein A7/metabolism
5.
Breast Cancer Res ; 22(1): 102, 2020 09 29.
Article in English | MEDLINE | ID: mdl-32993755

ABSTRACT

BACKGROUND: Breast cancer arises within specific regions in the human breast referred to as the terminal duct lobular units (TDLUs). These are relatively dynamic structures characterized by sex hormone driven cyclic epithelial turnover. TDLUs consist of unique parenchymal entities embedded within a fibroblast-rich lobular stroma. Here, we established and characterized a new human breast lobular fibroblast cell line against its interlobular counterpart with a view to assessing the role of region-specific stromal cues in the control of TDLU dynamics. METHODS: Primary lobular and interlobular fibroblasts were transduced to express human telomerase reverse transcriptase (hTERT). Differentiation of the established cell lines along lobular and interlobular pathways was determined by immunocytochemical staining and genome-wide RNA sequencing. Their functional properties were further characterized by analysis of mesenchymal stem cell (MSC) differentiation repertoire in culture and in vivo. The cells' physiological relevance for parenchymal differentiation was examined in heterotypic co-culture with fluorescence-activated cell sorting (FACS)-purified normal breast primary luminal or myoepithelial progenitors. The co-cultures were immunostained for quantitative assessment of epithelial branching morphogenesis, polarization, growth, and luminal epithelial maturation. In extension, myoepithelial progenitors were tested for luminal differentiation capacity in culture and in mouse xenografts. To unravel the significance of transforming growth factor-beta (TGF-ß)-mediated crosstalk in TDLU-like morphogenesis and differentiation, fibroblasts were incubated with the TGF-ß signaling inhibitor, SB431542, prior to heterotypic co-culture with luminal cells. RESULTS: hTERT immortalized fibroblast cell lines retained critical phenotypic traits in culture and linked to primary fibroblasts. Cell culture assays and transplantation to mice showed that the origin of fibroblasts determines TDLU-like and ductal-like differentiation of epithelial progenitors. Whereas lobular fibroblasts supported a high level of branching morphogenesis by luminal cells, interlobular fibroblasts supported ductal-like myoepithelial characteristics. TDLU-like morphogenesis, at least in part, relied on intact TGF-ß signaling. CONCLUSIONS: The significance of the most prominent cell type in normal breast stroma, the fibroblast, in directing epithelial differentiation is largely unknown. Through establishment of lobular and interlobular fibroblast cell lines, we here demonstrate that epithelial progenitors are submitted to stromal cues for site-specific differentiation. Our findings lend credence to considering stromal subtleties of crucial importance in the development of normal breast and, in turn, breast cancer.


Subject(s)
Breast Neoplasms/pathology , Breast/cytology , Cell Differentiation , Epithelial Cells/cytology , Fibroblasts/cytology , Stem Cells/metabolism , Stromal Cells/cytology , Adult , Animals , Breast/metabolism , Breast Neoplasms/metabolism , Cell Line , Coculture Techniques , Epithelial Cells/metabolism , Female , Fibroblasts/metabolism , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Stem Cells/cytology , Stromal Cells/metabolism , Xenograft Model Antitumor Assays , Young Adult
6.
Breast Cancer Res ; 21(1): 141, 2019 12 11.
Article in English | MEDLINE | ID: mdl-31829259

ABSTRACT

Organoid cultures are increasingly used to model human cancers experimentally with a view to tailoring personalized medicine and predicting drug responses. Breast cancer is no exception, but in particular, primary breast cancer poses some inherent difficulties due to the frequent presence of residual non-malignant cells in the biopsies. We originally developed an assay for the distinction between malignant and non-malignant structures in primary breast cancer organoid cultures (Petersen et al., Proc Natl Acad Sci (USA) 89(19):9064-8, 1992). Here, we apply this assay to assess the frequency of normal-like organoids in primary breast carcinoma cultures and the cellular composition as a consequence of passaging. We find that in consecutively collected samples of primary human breast cancers, residual non-malignant tissues were observed histologically in five out of ten biopsies. Based on relevant morphogenesis and correct polarization as recorded by expression in luminal epithelial cells of mucin 1 (Muc1), occludin, and keratin 19 (K19) and expression in basal cells of integrin ß4, p63, and K14, non-malignant organoids were present in all primary human breast cancer-derived cultures. Furthermore, passaging in a contemporary culture medium was in favor of the selective expansion of basal-like cells. We conclude that organoid cultures of human breast cancers are most representative of the tissue origin in primary culture.


Subject(s)
Breast Neoplasms/pathology , Organoids/pathology , Biomarkers , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Fluorescent Antibody Technique , Genetic Variation , Humans , Immunohistochemistry , Tissue Culture Techniques , Tumor Cells, Cultured , Whole Genome Sequencing
7.
Mol Cancer ; 17(1): 171, 2018 12 10.
Article in English | MEDLINE | ID: mdl-30526604

ABSTRACT

Tumorigenesis is increasingly considered to rely on subclones of cells poised to undergo an epithelial to mesenchymal transition (EMT) program. We and others have provided evidence, however, that the tumorigenesis of human breast cancer is not always restricted to typical EMT cells but is also somewhat paradoxically conveyed by subclones of apparently differentiated, non-EMT cells. Here we characterize such non-EMT-like and EMT-like subclones. Through a loss-of-function screen we found that a member of the E3 ubiquitin ligase complexes, FBXO11, specifically fuels tumor formation of a non-EMT-like clone by restraining the p53/p21 pathway. Interestingly, in the related EMT-like clone, FBXO11 operates through the BCL2 pathway with little or no impact on tumorigenesis. These data command caution in attempts to assess tumorigenesis prospectively based on EMT profiling, and they emphasize the importance of next generation subtyping of tumors, that is at the level of clonal composition.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Epithelial-Mesenchymal Transition/genetics , F-Box Proteins/genetics , Protein-Arginine N-Methyltransferases/genetics , Carcinogenesis/genetics , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/genetics , Female , Humans , MCF-7 Cells , Proto-Oncogene Proteins c-bcl-2/genetics , Signal Transduction/genetics , Tumor Suppressor Protein p53/genetics , Ubiquitin-Protein Ligases/genetics
8.
Curr Biol ; 28(20): 3220-3228.e6, 2018 10 22.
Article in English | MEDLINE | ID: mdl-30293715

ABSTRACT

Cancers develop in a complex mutational landscape. Genetic models of tumor formation have been used to explore how combinations of mutations cooperate to promote tumor formation in vivo. Here, we identify lactate dehydrogenase (LDH), a key enzyme in Warburg effect metabolism, as a cooperating factor that is both necessary and sufficient for epidermal growth factor receptor (EGFR)-driven epithelial neoplasia and metastasis in a Drosophila model. LDH is upregulated during the transition from hyperplasia to neoplasia, and neoplasia is prevented by LDH depletion. Elevated LDH is sufficient to drive this transition. Notably, genetic alterations that increase glucose flux, or a high-sugar diet, are also sufficient to promote EGFR-driven neoplasia, and this depends on LDH activity. We provide evidence that increased LDHA expression promotes a transformed phenotype in a human primary breast cell culture model. Furthermore, analysis of publically available cancer data showed evidence of synergy between elevated EGFR and LDHA activity linked to poor clinical outcome in a number of human cancers. Altered metabolism has generally been assumed to be an enabling feature that accelerates cancer cell proliferation. Our findings provide evidence that sugar metabolism may have a more profound role in driving neoplasia than previously appreciated.


Subject(s)
Drosophila Proteins/metabolism , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic , Hydro-Lyases/metabolism , Neoplasms, Glandular and Epithelial/metabolism , Neoplasms, Glandular and Epithelial/physiopathology , Neoplasms/metabolism , Neoplasms/physiopathology , Receptors, Invertebrate Peptide/metabolism , Animals , Cell Line, Tumor , Disease Models, Animal , Drosophila melanogaster , Humans
9.
Proc Natl Acad Sci U S A ; 114(47): E10102-E10111, 2017 11 21.
Article in English | MEDLINE | ID: mdl-29109259

ABSTRACT

The human breast parenchyma consists of collecting ducts and terminal duct lobular units (TDLUs). The TDLU is the site of origin of most breast cancers. The reason for such focal susceptibility to cancer remains poorly understood. Here, we take advantage of a region-specific heterogeneity in luminal progenitors to interrogate the differentiation repertoire of candidate stem cells in TDLUs. We show that stem-like activity in serial passage culture and in vivo breast morphogenesis relies on the preservation of a myoepithelial phenotype. By enrichment for region-specific progenitors, we identify bipotent and multipotent progenitors in ducts and TDLUs, respectively. We propose that focal breast cancer susceptibility, at least in part, originates from region-specific myoepithelial progenitors.


Subject(s)
Cell Transformation, Neoplastic/genetics , Epithelial Cells/cytology , Mammary Glands, Human/cytology , Multipotent Stem Cells/cytology , Muscle Cells/cytology , Adolescent , Adult , Antigens, CD/genetics , Antigens, CD/metabolism , Biomarkers/metabolism , Cell Culture Techniques , Cell Differentiation , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Epithelial Cells/metabolism , Female , Gene Expression , Humans , Keratin-19/genetics , Keratin-19/metabolism , Mammary Glands, Human/metabolism , Middle Aged , Multipotent Stem Cells/metabolism , Muscle Cells/metabolism , Myoepithelioma/diagnosis , Myoepithelioma/genetics , Myoepithelioma/metabolism , Myoepithelioma/pathology , Organ Specificity , Primary Cell Culture , Prognosis
10.
BMC Cancer ; 17(1): 19, 2017 01 05.
Article in English | MEDLINE | ID: mdl-28056857

ABSTRACT

BACKGROUND: One of the hallmarks of cancer is an altered energy metabolism, and here, mitochondria play a central role. Previous studies have indicated that some mitochondrial ribosomal proteins change their expression patterns upon transformation. METHOD: In this study, we have used the selection of recombinant antibody libraries displayed on the surface of filamentous bacteriophage as a proteomics discovery tool for the identification of breast cancer biomarkers. A small subpopulation of breast cells expressing both cytokeratin 19 and cytokeratin 14 was targeted using a novel selection procedure. RESULTS: We identified the mitochondrial ribosomal protein s18a (Mrps18a) as a protein which is upregulated in breast cancer. However, Mrps18a was not homogeneously upregulated in all cancer cells, suggesting the existence of sub-populations within the tumor. The upregulation was not confined to cytokeratin 19 and cytokeratin 14 double positive cells. CONCLUSION: This study illustrates how phage display can be applied towards the discovery of proteins which exhibit changes in their expression patterns. We identified the mitochondrial protein Mrps18a as being upregulated in human breast cancer cells compared to normal breast cells.


Subject(s)
Biomarkers, Tumor/analysis , Breast Neoplasms/metabolism , Mitochondrial Proteins/biosynthesis , Ribosomal Proteins/biosynthesis , Blotting, Western , Breast Neoplasms/pathology , Cell Surface Display Techniques , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunohistochemistry , Mitochondrial Proteins/analysis , Mitochondrial Ribosomes/metabolism , Proteomics , Ribosomal Proteins/analysis , Up-Regulation
11.
Oncotarget ; 8(6): 10580-10593, 2017 Feb 07.
Article in English | MEDLINE | ID: mdl-28076334

ABSTRACT

Understanding human cancer increasingly relies on insight gained from subtype specific comparisons between malignant and non-malignant cells. The most frequent subtype in breast cancer is the luminal. By far the most frequently used model for luminal breast cancer is the iconic estrogen receptor-positive (ERpos) MCF7 cell line. However, luminal specific comparisons have suffered from the lack of a relevant non-malignant counterpart. Our previous work has shown that transforming growth factor-ß receptor (TGFßR) inhibition suffices to propagate prospectively isolated ERpos human breast luminal cells from reduction mammoplasties (HBEC). Here we demonstrate that transduction of these cells with hTERT/shp16 renders them immortal while remaining true to the luminal lineage including expression of functional ER (iHBECERpos). Under identical culture conditions a major difference between MCF7 and normal-derived cells is the dependence of the latter on TGFßR inhibition for ER expression. In a breast fibroblast co-culture model we further show that whereas MCF7 proliferate concurrently with ER expression, iHBECERpos form correctly polarized acini, and segregate into proliferating and ER expressing cells. We propose that iHBECERpos may serve to shed light on hitherto unappreciated differences in ER regulation and function between normal breast and breast cancer.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Cell Separation/methods , Mammary Glands, Human/metabolism , Receptors, Estrogen/metabolism , Biomarkers, Tumor/genetics , Breast Neoplasms/classification , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Communication , Cell Line, Transformed , Cell Proliferation , Cellular Microenvironment , Coculture Techniques , Estradiol/pharmacology , Female , Fibroblasts/metabolism , Genotype , Humans , MCF-7 Cells , Mammary Glands, Human/drug effects , Phenotype , Receptors, Estrogen/drug effects , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Telomerase/genetics , Telomerase/metabolism , Transduction, Genetic
12.
Breast Cancer Res ; 18(1): 108, 2016 11 03.
Article in English | MEDLINE | ID: mdl-27809866

ABSTRACT

BACKGROUND: The terminal duct lobular unit (TDLU) is the most dynamic structure in the human breast and the putative site of origin of human breast cancer. Although stromal cells contribute to a specialized microenvironment in many organs, this component remains largely understudied in the human breast. We here demonstrate the impact on epithelium of two lineages of breast stromal fibroblasts, one of which accumulates in the TDLU while the other resides outside the TDLU in the interlobular stroma. METHODS: The two lineages are prospectively isolated by fluorescence activated cell sorting (FACS) based on different expression levels of CD105 and CD26. The characteristics of the two fibroblast lineages are assessed by immunocytochemical staining and gene expression analysis. The differentiation capacity of the two fibroblast populations is determined by exposure to specific differentiating conditions followed by analysis of adipogenic and osteogenic differentiation. To test whether the two fibroblast lineages are functionally imprinted by their site of origin, single cell sorted CD271low/MUC1high normal breast luminal epithelial cells are plated on fibroblast feeders for the observation of morphological development. Epithelial structure formation and polarization is shown by immunofluorescence and digitalized quantification of immunoperoxidase-stained cultures. RESULTS: Lobular fibroblasts are CD105high/CD26low while interlobular fibroblasts are CD105low/CD26high. Once isolated the two lineages remain phenotypically stable and functionally distinct in culture. Lobular fibroblasts have properties in common with bone marrow derived mesenchymal stem cells and they specifically convey growth and branching morphogenesis of epithelial progenitors. CONCLUSIONS: Two distinct functionally specialized fibroblast lineages exist in the normal human breast, of which the lobular fibroblasts have properties in common with mesenchymal stem cells and support epithelial growth and morphogenesis. We propose that lobular fibroblasts constitute a specialized microenvironment for human breast luminal epithelial progenitors, i.e. the putative precursors of breast cancer.


Subject(s)
Fibroblasts/cytology , Fibroblasts/metabolism , Mammary Glands, Human/cytology , Mammary Glands, Human/metabolism , Stromal Cells/cytology , Stromal Cells/metabolism , Biomarkers , Cell Differentiation , Cell Lineage , Cluster Analysis , Female , Flow Cytometry , Gene Expression Profiling , Humans , Immunophenotyping , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Phenotype
13.
Cancer Genomics Proteomics ; 13(1): 21-30, 2016.
Article in English | MEDLINE | ID: mdl-26708596

ABSTRACT

BACKGROUND/AIM: Primary tumors display a great level of intra-tumor heterogeneity in breast cancer. The current lack of prognostic and predictive biomarkers limits accurate stratification and the ability to predict response to therapy. The aim of the present study was to select recombinant antibody fragments specific against breast cancer subpopulations, aiding the discovery of novel biomarkers. MATERIALS AND METHODS: Recombinant antibody fragments were selected by phage display. A novel shadowstick technology enabled the direct selection using tissue sections of antibody fragments specific against small subpopulations of breast cancer cells. Selections were performed against a subpopulation of breast cancer cells expressing CD271+, as these previously have been indicated to be potential breast cancer stem cells. The selected antibody fragments were screened by phage ELISA on both breast cancer and myoepithelial cells. The antibody fragments were validated and evaluated by immunohistochemistry experiments. RESULTS: Our study revealed an antibody fragment, LH8, specific for breast cancer cells. Immunohistochemistry results indicate that this particular antibody fragment binds an antigen that exhibits differential expression in different breast cancer subpopulations. CONCLUSION: Further studies characterizing this antibody fragment, the subpopulation it binds and the cognate antigen may unearth novel biomarkers of clinical relevance.


Subject(s)
Bacteriophages/immunology , Breast Neoplasms/immunology , Antibody Specificity , Antigens, Neoplasm/immunology , Biomarkers, Tumor/immunology , Breast Neoplasms/pathology , Cells, Cultured , Female , Humans , Immunohistochemistry , Peptide Library
14.
Nat Commun ; 6: 8786, 2015 Nov 13.
Article in English | MEDLINE | ID: mdl-26564780

ABSTRACT

Investigating the susceptibility of oestrogen receptor-positive (ER(pos)) 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 ER(pos) cells and to expand what appear to be post-mitotic primary cells into exponentially growing cultures. We report a robust technique for isolating ER(pos) 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 ER(pos) HBECs are released from growth restraint by small molecule inhibitors of TGFß signalling, and that growth is augmented further in response to oestrogen. Importantly, ER signalling is functionally active in ER(pos) cells in extended culture. These findings open a new avenue of experimentation with normal ER(pos) HBECs and provide a basis for understanding the evolution of human breast cancer.


Subject(s)
Breast/cytology , Epithelial Cells/cytology , Estrogens/metabolism , Flow Cytometry/methods , Receptors, Estrogen/metabolism , Breast/metabolism , Cell Culture Techniques , Cell Proliferation , Cells, Cultured , Epithelial Cells/metabolism , Female , Humans
15.
Immunol Res ; 62(3): 263-72, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25963139

ABSTRACT

Breast cancer tumors are composed of heterogeneous cell populations. These populations display a high variance in morphology, growth and metastatic propensity. They respond differently to therapeutic interventions, and some may be more prone to cause recurrence. Studying individual subpopulations of breast cancer may provide crucial knowledge for the development of individualized therapy. However, this process is challenged by the availability of biomarkers able to identify subpopulations specifically. Here, we demonstrate an approach for phage display selection of recombinant antibody fragments on cryostat sections of human breast cancer tissue. This method allows for selection of recombinant antibodies binding to antigens specifically expressed in a small part of the tissue section. In this case, a CD271(+) subpopulation of breast cancer cells was targeted, and these may be potential breast cancer stem cells. We isolated an antibody fragment LH 7, which in immunohistochemistry experiments demonstrates specific binding to breast cancer subpopulations. The selection of antibody fragments directly on small defined areas within a larger section of malignant tissue is a novel approach by which it is possible to better target cellular heterogeneity in proteomic studies. The identification of novel biomarkers is relevant for our understanding and intervention in human diseases. The selection of the breast cancer-specific antibody fragment LH 7 may reveal novel subpopulation-specific biomarkers, which has the potential to provide new insight and treatment strategies for breast cancer.


Subject(s)
Antigens, Neoplasm/immunology , Breast Neoplasms/immunology , Cell Surface Display Techniques/methods , Nerve Tissue Proteins/immunology , Receptors, Nerve Growth Factor/immunology , Single-Domain Antibodies/immunology , Antibodies, Neoplasm/immunology , Antibody Specificity/immunology , Biomarkers, Tumor/analysis , Breast Neoplasms/genetics , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Female , Humans , Staining and Labeling/methods
16.
Proc Natl Acad Sci U S A ; 109(16): 6124-9, 2012 Apr 17.
Article in English | MEDLINE | ID: mdl-22454501

ABSTRACT

The majority of human breast cancers exhibit luminal epithelial differentiation. However, most aggressive behavior, including invasion and purported cancer stem cell activity, are considered characteristics of basal-like cells. We asked the following questions: Must luminal-like breast cancer cells become basal-like to initiate tumors or to invade? Could luminally differentiated cells within a basally initiated hierarchy also be tumorigenic? To answer these questions, we used rare and mutually exclusive lineage markers to isolate subsets of luminal-like and basal-like cells from human breast tumors. We enriched for populations with or without prominent basal-like traits from individual tumors or single cell cloning from cell lines and recovered cells with a luminal-like phenotype. Tumor cells with basal-like traits mimicked phenotypic and functional behavior associated with stem cells assessed by gene expression, mammosphere formation and lineage markers. Luminal-like cells without basal-like traits, surprisingly, were fully capable of initiating invasive tumors in NOD SCID gamma (NSG) mice. In fact, these phenotypically pure luminal-like cells generated larger and more invasive tumors than their basal-like counterparts. The tumorigenicity and invasive potential of the luminal-like cancer cells relied strongly on the expression of the gene GCNT1, which encodes a key glycosyltransferase controlling O-glycan branching. These findings demonstrate that basal-like cells, as defined currently, are not a requirement for breast tumor aggressiveness, and that within a single tumor there are multiple "stem-like" cells with tumorigenic potential casting some doubt on the hypothesis of hierarchical or differentiative loss of tumorigenicity.


Subject(s)
Breast Neoplasms/pathology , Cell Differentiation , Mammary Neoplasms, Experimental/pathology , Neoplastic Stem Cells/pathology , Adapalene , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Interleukin Receptor Common gamma Subunit/deficiency , Interleukin Receptor Common gamma Subunit/genetics , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, Knockout , Mice, Nude , Mucin-1/metabolism , N-Acetylglucosaminyltransferases/genetics , N-Acetylglucosaminyltransferases/metabolism , Naphthalenes/metabolism , Neoplasm Invasiveness , Neoplastic Stem Cells/metabolism , Oligonucleotide Array Sequence Analysis , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Heterologous , Tumor Cells, Cultured
17.
PLoS One ; 6(9): e23833, 2011.
Article in English | MEDLINE | ID: mdl-21915264

ABSTRACT

Epithelial to mesenchymal transition (EMT) is a critical event in cancer progression and is closely linked to the breast epithelial cancer stem cell phenotype. Given the close interaction between the vascular endothelium and cancer cells, especially at the invasive front, we asked whether endothelial cells might play a role in EMT. Using a 3D culture model we demonstrate that endothelial cells are potent inducers of EMT in D492 an immortalized breast epithelial cell line with stem cell properties. Endothelial induced mesenchymal-like cells (D492M) derived from D492, show reduced expression of keratins, a switch from E-Cadherin (E-Cad) to N-Cadherin (N-Cad) and enhanced migration. Acquisition of cancer stem cell associated characteristics like increased CD44(high)/CD24(low) ratio, resistance to apoptosis and anchorage independent growth was also seen in D492M cells. Endothelial induced EMT in D492 was partially blocked by inhibition of HGF signaling. Basal-like breast cancer, a vascular rich cancer with stem cell properties and adverse prognosis has been linked with EMT. We immunostained several basal-like breast cancer samples for endothelial and EMT markers. Cancer cells close to the vascular rich areas show no or decreased expression of E-Cad and increased N-Cad expression suggesting EMT. Collectively, we have shown in a 3D culture model that endothelial cells are potent inducers of EMT in breast epithelial cells with stem cell properties. Furthermore, we demonstrate that basal-like breast cancer contains cells with an EMT phenotype, most prominently close to vascular rich areas of these tumors. We conclude that endothelial cells are potent inducers of EMT and may play a role in progression of basal-like breast cancer.


Subject(s)
Epithelial Cells/cytology , Epithelial Cells/metabolism , Apoptosis/physiology , Blotting, Western , Cell Dedifferentiation , Cell Line , Cell Line, Tumor , Cells, Cultured , Epidermal Growth Factor/metabolism , Epithelial-Mesenchymal Transition , Female , Fibroblast Growth Factors/metabolism , Flow Cytometry , Hepatocyte Growth Factor/metabolism , Humans , Immunochemistry , In Vitro Techniques , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/metabolism , Transforming Growth Factor beta1/metabolism
18.
Cold Spring Harb Perspect Biol ; 2(5): a003160, 2010 May.
Article in English | MEDLINE | ID: mdl-20452965

ABSTRACT

The origins of the epithelial cells participating in the development, tissue homeostasis, and cancer of the human breast are poorly understood. However, emerging evidence suggests a role for adult tissue-specific stem cells in these processes. In a hierarchical manner, these generate the two main mammary cell lineages, producing an increasing number of cells with distinct properties. Understanding the biological characteristics of human breast stem cells and their progeny is crucial in attempts to compare the features of normal stem cells and cancer precursor cells and distinguish these from nonprecursor cells and cells from the bulk of a tumor. A historical overview of research on human breast stem cells in primary tissue and in culture reveals the progress that has been made in this area, whereas a focus on the cell-of-origin and reprogramming that occurs during neoplastic conversion provides insight into the enigmatic way in which human breast cancers are skewed toward the luminal epithelial lineage.


Subject(s)
Breast/cytology , Stem Cells/cytology , Breast/growth & development , Breast/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line , Female , Humans , Keratins/metabolism
19.
Ugeskr Laeger ; 169(36): 2968-72, 2007 Sep 03.
Article in Danish | MEDLINE | ID: mdl-17953870

ABSTRACT

A hallmark of cancer is tumour heterogeneity. This is generally attributed to genomic instability and phenotypic drifting of transformed cells. In recent years another source of tumour heterogeneity has emerged--the "cancer initiating cells" or "cancer stem cells". These are very rare cancer cells that are believed to fuel the bulk of the epithelial cells composing the tumour. Here we discuss the possible existence of normal and cancer stem cells in the human breast and in breast cancer and their potential role in the evolution of this disease.


Subject(s)
Breast Neoplasms/etiology , Animals , Breast/cytology , Breast/pathology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Female , Hematopoietic Stem Cells/pathology , Humans , Leukemia, Myeloid, Acute/pathology , Mice , Stem Cells/pathology
20.
Toxicol Sci ; 93(2): 357-68, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16757570

ABSTRACT

We examined the effect of 17beta-estradiol (E2) and soy isoflavones' exposure on morphogenesis and global gene expression in the murine mammary gland. Three exposure regimens were applied: isoflavones added to the diet throughout either the lactational period (via the dams) or the postweaning period and E2 administered orally during the lactational period. Whole mounts of mammary glands were evaluated both in juvenile and adult animals with respect to branching morphogenesis and terminal end bud (TEB) formation. At postnatal day (PND) 28, we observed a significant increase in branching morphogenesis in all treated groups with the most pronounced effect after E2 exposure. For the E2-treated animals there was also a significant increase in TEB formation. At PNDs 42-43 the postweaning isoflavone and the E2 groups showed a transient reduction in the number of TEBs. A similar response after isoflavone and E2 exposure was further substantiated by changes in gene expression, since the same groups of genes were up- and downregulated, particularly in the E2 and postweaning isoflavone regimen. All changes in gene expression correlated with changes in the cellular composition of the gland, i.e., more and larger TEBs and ducts. The results suggest an estrogenic response of physiological doses of isoflavones on mammary gland development at both the morphological and molecular level, which resembled that induced by puberty.


Subject(s)
Estradiol/pharmacology , Glycine max , Isoflavones/pharmacology , Mammary Glands, Animal/drug effects , Morphogenesis/drug effects , Animals , Estradiol/blood , Estrogen Receptor alpha/genetics , Female , Gene Expression Profiling , In Situ Hybridization , Isoflavones/blood , Male , Mammary Glands, Animal/growth & development , Mammary Glands, Animal/metabolism , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...