Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Sci Rep ; 10(1): 4788, 2020 Mar 11.
Article in English | MEDLINE | ID: mdl-32161318

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

2.
Sci Rep ; 9(1): 14843, 2019 10 16.
Article in English | MEDLINE | ID: mdl-31619692

ABSTRACT

Human breast cancer is believed to arise in luminal progenitors within the normal breast. A subset of these are double positive (DP) for basal and luminal keratins and localizes to a putative stem cell zone within ducts. We here present a new protocol based on a combination of CD146 with CD117 and CD326 which provides an up to thirty fold enrichment of the DP cells. We show by expression profiling, colony formation, and morphogenesis that CD146high/CD117high/CD326high DP cells belong to a luminal progenitor compartment. While these DP cells are located quite uniformly in ducts, with age a variant type of DP (vDP) cells, which is mainly CD146-negative, accumulates in lobules. Intriguingly, in specimens with BRCA1 mutations known to predispose for cancer, higher frequencies of lobular vDP cells are observed. We propose that vDP cells are strong candidates for tracing the cellular origin of breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Carcinogenesis , Keratin-14/metabolism , Keratin-19/metabolism , Mammary Glands, Human/metabolism , Neoplastic Stem Cells/metabolism , Adolescent , Adult , Breast Neoplasms/pathology , CD146 Antigen/metabolism , Cells, Cultured , Female , Healthy Volunteers , Humans , Mammary Glands, Human/pathology , Middle Aged , Neoplastic Stem Cells/pathology , Young Adult
3.
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
4.
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
5.
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
6.
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
7.
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
8.
Cancer Res ; 72(14): 3687-701, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22552289

ABSTRACT

Women older than 50 years account for 75% of new breast cancer diagnoses, and the majority of these tumors are of a luminal subtype. Although age-associated changes, including endocrine profiles and alterations within the breast microenvironment, increase cancer risk, an understanding of the cellular and molecular mechanisms that underlies these observations is lacking. In this study, we generated a large collection of normal human mammary epithelial cell strains from women ages 16 to 91 years, derived from primary tissues, to investigate the molecular changes that occur in aging breast cells. We found that in finite lifespan cultured and uncultured epithelial cells, aging is associated with a reduction of myoepithelial cells and an increase in luminal cells that express keratin 14 and integrin-α6, a phenotype that is usually expressed exclusively in myoepithelial cells in women younger than 30 years. Changes to the luminal lineage resulted from age-dependent expansion of defective multipotent progenitors that gave rise to incompletely differentiated luminal or myoepithelial cells. The aging process therefore results in both a shift in the balance of luminal/myoepithelial lineages and to changes in the functional spectrum of multipotent progenitors, which together increase the potential for malignant transformation. Together, our findings provide a cellular basis to explain the observed vulnerability to breast cancer that increases with age.


Subject(s)
Aging , Cell Differentiation , Cellular Senescence , Mammary Glands, Human/cytology , Multipotent Stem Cells/physiology , Aged , Aged, 80 and over , Cells, Cultured , Epithelial Cells/cytology , Female , Humans , Middle Aged , Phenotype
9.
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
10.
Int J Dev Biol ; 55(7-9): 719-29, 2011.
Article in English | MEDLINE | ID: mdl-22161829

ABSTRACT

Distinct subsets of cells, including cells with stem cell-like properties, have been proposed to exist in normal human breast epithelium and breast carcinomas. The cellular origins of epithelial cells contributing to gland development, tissue homeostasis and cancer are, however, still poorly understood. The mouse is a widely used model of mammary gland development, both directly by studying the mouse mammary epithelial cells themselves and indirectly, by studying development, morphogenesis, differentiation and carcinogenesis of xenotransplanted human breast epithelium in vivo. While in early studies, human or mouse epithelium was implanted as fragments into the mouse gland, more recent technical progress has allowed the self-renewal capacity and differentiation potential of distinct cell populations or even individual cells to be interrogated. Here, we review and discuss similarities and differences between mouse and human gland development with particular emphasis on the identity and localization of stem cells, and the influence of the surrounding microenvironment. It is concluded that while recent advances in the field have contributed immense insight into how the normal mammary gland develops and is maintained, significant discrepancies exist between the mouse and human gland which should be taken into consideration in current and future models of mammary stem cell biology.


Subject(s)
Breast/cytology , Mammary Glands, Animal/cytology , Stem Cells/cytology , Animals , Breast/growth & development , Breast Neoplasms/pathology , Cell Differentiation , Cell Lineage , Female , Humans , Mammary Glands, Animal/growth & development , Mice , Neoplastic Stem Cells/pathology , Signal Transduction , Stem Cell Niche , Stem Cell Transplantation , Stem Cells/physiology , Transplantation, Heterologous , Tumor Microenvironment
11.
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
12.
In Vitro Cell Dev Biol Anim ; 47(2): 139-48, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21082277

ABSTRACT

An epithelial cell line, referred to as A163, was established from breast carcinoma derived from a patient with a strong family history of breast cancer but no known breast cancer susceptibility mutation. A163 was propagated in a serum-free culture medium including the epidermal growth factor. Immunophenotypic characterization demonstrated a mixed luminal and basal-like phenotype. When epidermal growth factor was excluded from the culture medium, A163 entered a quiescent period followed by a period of increased cell proliferation in a subpopulation of the cells. The epidermal growth factor-independent subpopulation retained the basal-like phenotype of the parental cell line. Karyotype and fluorescent in situ hybridization analysis showed an amplification of epidermal growth factor receptor on 7q in A163-S1 only, resulting in high expression of total and phosphorylated epidermal growth factor receptor. The A163-S1 sub-line piles up in culture, indicating a loss of contact inhibition. When grown on transwell filters, A163 shows basal expression of P63 and cytokeratin 14, whereas A163-S1 expresses P63 ubiquitously, and has lost the basal specific expression of cytokeratin 14, indicating a loss of polarity. Furthermore, when cultured in reconstituted basement membrane matrix, A163 form polarized normal like acini. In contrast, A163-S1 form large disorganized structures with lack of polarity. These cell lines may prove useful to understand molecular changes in breast cancer progression, in particular basal-like breast cancer subtype with bad prognosis and no current treatment options.


Subject(s)
Breast Neoplasms/pathology , Carcinoma/pathology , Cell Line, Tumor , ErbB Receptors/genetics , Gene Amplification , Neoplasms, Basal Cell/pathology , Breast Neoplasms/genetics , Carcinoma/genetics , Cell Proliferation , Culture Media, Serum-Free/pharmacology , Epidermal Growth Factor/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/pathology , Female , Humans , Middle Aged , Neoplasms, Basal Cell/genetics , Selection, Genetic
13.
BMC Res Notes ; 3: 184, 2010 Jul 07.
Article in English | MEDLINE | ID: mdl-20609224

ABSTRACT

BACKGROUND: Epithelial-stromal interaction provides regulatory signals that maintain correct histoarchitecture and homeostasis in the normal breast and facilitates tumor progression in breast cancer. However, research on the regulatory role of the endothelial component in the normal and malignant breast gland has largely been neglected. The aim of the study was to investigate the effects of endothelial cells on growth and differentiation of human breast epithelial cells in a three-dimensional (3D) co-culture assay. METHODS: Breast luminal and myoepithelial cells and endothelial cells were isolated from reduction mammoplasties. Primary cells and established normal and malignant breast cell lines were embedded in reconstituted basement membrane in direct co-culture with endothelial cells and by separation of Transwell filters. Morphogenic and phenotypic profiles of co-cultures was evaluated by phase contrast microscopy, immunostaining and confocal microscopy. RESULTS: In co-culture, endothelial cells stimulate proliferation of both luminal- and myoepithelial cells. Furthermore, endothelial cells induce a subpopulation of luminal epithelial cells to form large acini/ducts with a large and clear lumen. Endothelial cells also stimulate growth and cloning efficiency of normal and malignant breast epithelial cell lines. Transwell and gradient co-culture studies show that endothelial derived effects are mediated - at least partially - by soluble factors. CONCLUSION: Breast endothelial cells - beside their role in transporting nutrients and oxygen to tissues - are vital component of the epithelial microenvironment in the breast and provide proliferative signals to the normal and malignant breast epithelium. These growth promoting effects of endothelial cells should be taken into consideration in breast cancer biology.

14.
Am J Pathol ; 176(3): 1229-40, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20133812

ABSTRACT

Tumor cells can activate stroma, yet the implication of this activation in terms of reciprocal induction of gene expression in tumor cells is poorly understood. Epithelial Stromal Interaction 1 (EPSTI1) is an interferon response gene originally isolated from heterotypic recombinant cultures of human breast cancer cells and activated breast myofibroblasts. Here we describe the first immunolocalization of EPSTI1 in normal and cancerous breast tissue, and we provide evidence for a role of this molecule in the regulation of tumor cell properties and epithelial-mesenchymal transition. In general, no EPSTI1 staining was observed in normal breast epithelial cells from reduction mammoplasties (n=25). However, in carcinomas, staining was positive in 22 of 40 biopsies and inversely correlated with the level of differentiation. To address the function of EPSTI1, we expressed EPSTI1 ectopically in one cell line and silenced endogenous EPSTI1 by RNA interference in another. Irrespective of the experimental approach, EPSTI1 expression led to an increase in tumorsphere formation-a property associated with breast stem/progenitor cells. Most remarkably, we show that EPSTI1, by conveying spread of tumor cells, can replace peritumoral activated fibroblasts in a tumor environment assay. These observations implicate EPSTI1 as a hitherto unappreciated regulator of tumor cell properties.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Neoplasm Proteins/metabolism , Animals , Biological Assay , Cell Line, Tumor , Cell Nucleus/metabolism , Epithelium/metabolism , Epithelium/pathology , Female , Humans , Mesoderm/metabolism , Mesoderm/pathology , Mice , Organ Specificity , RNA Interference
15.
Integr Biol (Camb) ; 1(1): 70-9, 2009 Jan.
Article in English | MEDLINE | ID: mdl-20023793

ABSTRACT

In adult tissues, multi-potent progenitor cells are some of the most primitive members of the developmental hierarchies that maintain homeostasis. That progenitors and their more mature progeny share identical genomes, suggests that fate decisions are directed by interactions with extrinsic soluble factors, ECM, and other cells, as well as physical properties of the ECM. To understand regulation of fate decisions, therefore, would require a means of understanding carefully choreographed combinatorial interactions. Here we used microenvironment protein microarrays to functionally identify combinations of cell-extrinsic mammary gland proteins and ECM molecules that imposed specific cell fates on bipotent human mammary progenitor cells. Micropatterned cell culture surfaces were fabricated to distinguish between the instructive effects of cell-cell versus cell-ECM interactions, as well as constellations of signaling molecules; and these were used in conjunction with physiologically relevant 3 dimensional human breast cultures. Both immortalized and primary human breast progenitors were analyzed. We report on the functional ability of those proteins of the mammary gland that maintain quiescence, maintain the progenitor state, and guide progenitor differentiation towards myoepithelial and luminal lineages.


Subject(s)
Cell Culture Techniques/methods , Mammary Glands, Human/cytology , Mammary Glands, Human/physiology , Protein Array Analysis/methods , Proteome/metabolism , Stem Cells/cytology , Stem Cells/physiology , Cell Differentiation , Cell Separation/methods , Cell Survival , Cells, Cultured , Humans
16.
J Cell Biol ; 177(1): 87-101, 2007 Apr 09.
Article in English | MEDLINE | ID: mdl-17420292

ABSTRACT

Cellular pathways that contribute to adult human mammary gland architecture and lineages have not been previously described. In this study, we identify a candidate stem cell niche in ducts and zones containing progenitor cells in lobules. Putative stem cells residing in ducts were essentially quiescent, whereas the progenitor cells in the lobules were more likely to be actively dividing. Cells from ducts and lobules collected under the microscope were functionally characterized by colony formation on tissue culture plastic, mammosphere formation in suspension culture, and morphogenesis in laminin-rich extracellular matrix gels. Staining for the lineage markers keratins K14 and K19 further revealed multipotent cells in the stem cell zone and three lineage-restricted cell types outside this zone. Multiparameter cell sorting and functional characterization with reference to anatomical sites in situ confirmed this pattern. The proposal that the four cell types are indeed constituents of an as of yet undescribed stem cell hierarchy was assessed in long-term cultures in which senescence was bypassed. These findings identify an adult human breast ductal stem cell activity and its earliest descendants.


Subject(s)
Breast/cytology , Cell Lineage , Stem Cells/classification , Biomarkers/metabolism , Breast/metabolism , Cell Differentiation , Cell Line , Female , Humans , Keratins/metabolism , Mammary Glands, Human/cytology , Oncogene Proteins, Viral/genetics , Papillomavirus E7 Proteins , Repressor Proteins/genetics , Stem Cells/metabolism , Transduction, Genetic
17.
In Vitro Cell Dev Biol Anim ; 42(10): 332-40, 2006.
Article in English | MEDLINE | ID: mdl-17316068

ABSTRACT

Attempts to study endothelial-epithelial interactions in the human breast have been hampered by lack of protocols for long-term cultivation of breast endothelial cells (BRENCs). The aim of this study was to establish long-term cultures of BRENCs and to compare their phenotypic traits with the tissue of origin. Microvasculature was localized in situ by immunohistochemistry in breast samples. From this tissue, collagen-rich stroma and adipose tissue were dissected mechanically and further disaggregated to release microvessel organoids. BRENCs were cultured from these organoids in endothelial specific medium and characterized by staining for endothelial markers. Microvessels were a prominent feature of intralobular tissue as evidenced by immunostaining against endothelial specific markers such as CD31, VE-cadherin, and von Willebrand factor (VWF). Double staining against VE-cadherin and lymphatic vessel endothelial hyaluronan receptor-1 (LYVE-1) showed that blood and lymphatic vessels could be distinguished. An antibody against CD31 was used to refine protocols for isolation of microvasculature from reduction mammoplasties. BRENCs retained critical traits even at high passage, including uptake of low-density lipoprotein, and had E-selectin induced upon treatment with tumor necrosis factor-alpha. The first signs of senescence in passage 14 were accompanied by gain of trisomy 11. At passage 18 cells showed chromosomal aberrations and growth arrest as revealed by beta-galactosidase staining. We demonstrate here that breast microvasculature may serve as a large-scale source for expansion of BRENCs with molecular and functional traits preserved. These cells will form the basis for studies on the role of endothelial cells in breast morphogenesis.


Subject(s)
Breast/blood supply , Breast/cytology , Cellular Senescence/physiology , Endothelial Cells/cytology , Cell Separation , Cells, Cultured , Chromosomal Instability , Endothelial Cells/metabolism , Female , Humans , Karyotyping , Phenotype , Time Factors
18.
J Mammary Gland Biol Neoplasia ; 10(1): 61-74, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15886887

ABSTRACT

Recent genome-wide expression analysis of breast cancer has brought new life to the classical idea of tumors as caricatures of the process of tissue renewal as envisioned by Pierce and Speers (Cancer Res 1988;48:1996-2004) more than a decade ago. The search for a cancer founder cell or different cancer founder cells is only possible if a hierarchy of differentiation has been established for the particular tissue in question. In the human breast, the luminal epithelial and myoepithelial lineages have been characterized extensively in situ by increasingly elaborate panel of markers, and methods to isolate, culture, and clone different subpopulations have improved dramatically. Comparisons have been made with the mouse mammary gland in physiological three-dimensional culture assays of morphogenesis, and the plasticity of breast epithelial cells has been challenged by immortalization and transformation. As a result of these efforts, several candidate progenitor cells have been proposed independently of each other, and some of their features have been compared. This research has all been done to better understand breast tissue homeostasis, cell-type diversification in general and breast cancer evolution in particular. The present review discusses the current approaches to address these issues and the measures taken to unravel and maintain cell type diversification for further investigation.


Subject(s)
Breast/cytology , Cell Differentiation , Animals , Breast/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Homeostasis , Humans , Stem Cells/cytology , Stem Cells/metabolism
19.
In Vitro Cell Dev Biol Anim ; 41(10): 337-42, 2005.
Article in English | MEDLINE | ID: mdl-16448223

ABSTRACT

Germ line mutations in BRCA1 and BRCA2 account for a large proportion of inherited breast and ovarian cancer. Both genes are involved in DNA repair by homologous recombination and are thought to play a vital role in maintaining genomic stability. A major drawback for long-term functional studies of BRCA in general and BRCA2 in particular has been a lack of representative human breast epithelial cell lines. In the present study, we have established three cell lines from two patients harboring the 999del5 germ line founder mutation in the BRCA2 gene. Primary cultures were established from cellular outgrowth of explanted tissue and subsequently transfected with a retroviral construct containing the HPV-16 E6 and E7 oncogenes. Paired cancer-derived and normal-derived cell lines were established from one patient referred to as BRCA2-999del5-2T and BRCA2-999del5-2N, respectively. In addition, one cell line was derived from cancer-associated normal tissue from another patient referred to as BRCA2-999del5-1N. All three cell lines showed characteristics of breast epithelial cells as evidenced by expression of breast epithelial specific cytokeratins. Cytogenetic analysis showed marked chromosomal instability with tetraploidy and frequent telomeric associations. In conclusion, we have established three breast epithelial cell lines from two patients carrying the BRCA2 Icelandic 999del5 founder mutation. These cell lines form the basis for further studies on carcinogenesis and malignant progression of breast cancer on a defined genetic background.


Subject(s)
Breast/cytology , Cell Line, Tumor , Epithelial Cells/cytology , Genes, BRCA2 , Mutation/genetics , Chromosomal Instability/genetics , Cytogenetic Analysis , DNA Mutational Analysis , DNA Primers , Female , Genetic Vectors/genetics , Humans , Iceland , Immunohistochemistry , Karyotyping , Keratins/metabolism , Oncogenes/genetics , Retroviridae
SELECTION OF CITATIONS
SEARCH DETAIL
...