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











Publication year range
2.
J Mammary Gland Biol Neoplasia ; 20(1-2): 51-62, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26286174

ABSTRACT

Tissue based research requires a background in human and veterinary pathology, developmental biology, anatomy, as well as molecular and cellular biology. This type of comparative tissue biology (CTB) expertise is necessary to tackle some of the conceptual challenges in human breast stem cell research. It is our opinion that the scarcity of CTB expertise contributed to some erroneous interpretations in tissue based research, some of which are reviewed here in the context of breast stem cells. In this article we examine the dissimilarities between mouse and human mammary tissue and suggest how these may impact stem cell studies. In addition, we consider the differences between breast ducts vs. lobules and clarify how these affect the interpretation of results in stem cell research. Lastly, we introduce a new elaboration of normal epithelial cell types in human breast and discuss how this provides a clinically useful basis for breast cancer classification.


Subject(s)
Carcinoma/pathology , Cell Differentiation , Keratins/analysis , Mammary Glands, Animal/anatomy & histology , Mammary Glands, Human/anatomy & histology , Stem Cells/chemistry , Stem Cells/cytology , Animals , Carcinoma/chemistry , Cell Lineage , Female , Flow Cytometry , Histology, Comparative , Humans , Immunohistochemistry , Mammary Glands, Animal/chemistry , Mammary Glands, Human/chemistry , Mice
3.
Methods Mol Biol ; 1293: 213-20, 2015.
Article in English | MEDLINE | ID: mdl-26040690

ABSTRACT

Despite advances in early detection, prevention and treatment of breast cancer, the mortality of breast cancer patients did not decrease considerably in the last years. Metastatic breast cancer remains incurable. There is compelling evidence that dissemination of breast cancer cells at distant sites is an early event. At the time of detection and diagnosis, patients have disseminated breast cancer cells in the bone marrow. Only in half of these patients the disseminated cells proliferate and generate metastases, typically in 3-5 years for ER negative breast tumors and 10-15 years for ER positive breast tumors. In other patients metastases never develop. The ability to predict which patients will develop metastases and to devise strategies to interfere with this process hinges on understanding the mechanisms underlying growth at the metastatic site. In turn, this requires novel experimental systems that model in vitro the survival, dormancy and proliferation of disseminated cancer cells.We have established such experimental systems that model the bone microenvironment of the breast cancer metastatic niche. These systems are based on 3D complex cultures of human bone marrow stromal cells and breast cancer cell lines in collagen biomatrices. We identified conditions in which cancer cells are dormant, and conditions in which they proliferate and we validated the results in vivo. Dormant cancer cells were able to proliferate upon transfer into supportive microenvironment or upon manipulation of signaling pathways that control dormancy. These experimental systems will be instrumental in screening new compounds for metastasis studies and particularly in studying the pathways that control cellular dormancy. We provide in this chapter detailed protocols for these complex 3D coculture systems.


Subject(s)
Bone Neoplasms/pathology , Bone Neoplasms/secondary , Breast Neoplasms/pathology , Tumor Microenvironment , Cell Culture Techniques , Cell Line , Coculture Techniques , Female , Humans , Mesenchymal Stem Cells , Spheroids, Cellular , Tumor Cells, Cultured
4.
Stem Cell Reports ; 4(4): 699-711, 2015 Apr 14.
Article in English | MEDLINE | ID: mdl-25818813

ABSTRACT

Characterization of normal breast stem cells is important for understanding their role in breast development and in breast cancer. However, the identity of these cells is a subject of controversy and their localization in the breast epithelium is not known. In this study, we utilized a novel approach to analyze the morphogenesis of mammary lobules, by combining one-dimensional theoretical models and computer-generated 3D fractals. Comparing predictions of these models with immunohistochemical analysis of tissue sections for candidate stem cell markers, we defined distinct areas where stem cells reside in the mammary lobule. An increased representation of stem cells was found in smaller, less developed lobules compared to larger, more mature lobules, with marked differences in the gland of nulliparous versus parous women and that of BRCA1/2 mutation carriers versus non-carriers.


Subject(s)
Cell Differentiation , Mammary Glands, Human , Organogenesis , Stem Cells/cytology , Stem Cells/metabolism , Biomarkers/metabolism , Epithelium/metabolism , Female , Humans , Models, Biological , Tissue Culture Techniques
5.
Breast Cancer Res ; 16(3): R52, 2014 May 27.
Article in English | MEDLINE | ID: mdl-24887554

ABSTRACT

INTRODUCTION: Although estrogen and progesterone play a key role in normal mammary development and in breast cancer, the potential for proliferation and lineage differentiation as well as origin of cells that express the estrogen receptor (ER) in normal breast epithelium are not known. Some evidence suggests that normal human mammary stem/progenitor cells are ER-, but the identity of these cells and the cellular hierarchy of breast epithelium are still subjects of controversy. It is likely that elucidation of these aspects will bring insight into the cellular origin of breast cancer subtypes. METHODS: We used fluorescence-activated cell sorting of primary human mammary epithelial cells along with in vitro and in vivo functional assays to examine the hierarchic relation between cells with aldehyde dehydrogenase enzymatic activity (ALDH+ cells) and ER+ cells in the normal human breast epithelium. We assessed the proliferation and lineage differentiation potential of these cells in vitro and in vivo. A gene reporter assay was used to separate live ER+ and ER- mammary epithelial cells. With shRNA-mediated knockdown, we investigated the role of ALDH isoforms in the functionality of mammary epithelial progenitor cells. RESULTS: We describe a cellular hierarchy in the normal human mammary gland in which ER-/ALDH+ cells with functional properties of stem/progenitor cells generate ER+ progenitor cells, which in turn give rise to cells of luminal lineage. We show that the ALDH1A1 isoform, through its function in the retinoic acid metabolism, affects the proliferation and/or early differentiation of stem/progenitor cells and is important for branching morphogenesis. CONCLUSIONS: This study presents direct evidence that ER+ cells are generated by ER-/ALDH+ stem/progenitor cells. We also show that ER+ cells are able to generate cell progeny of luminal lineage in vitro and in vivo. Loss of ALDH1A1 function impairs this process, as well as branching morphogenesis and clonogenicity in suspension culture. This latter effect is reversed by treatment with retinoic acid.


Subject(s)
Aldehyde Dehydrogenase/metabolism , Mammary Glands, Human/metabolism , Receptors, Estrogen/metabolism , Tretinoin/pharmacology , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase 1 Family , Aldehyde Oxidoreductases/metabolism , Cell Differentiation , Cell Lineage , Cell Proliferation , Cells, Cultured , Female , Flow Cytometry , Humans , Mammary Glands, Human/cytology , Protein Isoforms/genetics , RNA Interference , RNA, Small Interfering , Receptors, Estrogen/biosynthesis , Retinal Dehydrogenase , Stem Cells/cytology , Stem Cells/enzymology , Tretinoin/metabolism
6.
Stem Cell Reports ; 2(6): 780-93, 2014 Jun 03.
Article in English | MEDLINE | ID: mdl-24936466

ABSTRACT

Using in vitro and in vivo experimental systems and in situ analysis, we show that growth hormone (GH) is secreted locally by normal human mammary epithelial cells upon progesterone stimulation. GH increases proliferation of a subset of cells that express growth hormone receptor (GHR) and have functional properties of stem and early progenitor cells. In 72% of ductal carcinoma in situ lesions, an expansion of the cell population that expresses GHR was observed, suggesting that GH signaling may contribute to breast cancer development.


Subject(s)
Cell Proliferation/drug effects , Human Growth Hormone/metabolism , Mammary Glands, Human/cytology , Mammary Glands, Human/metabolism , Progesterone/pharmacology , Stem Cells/cytology , Stem Cells/metabolism , Cells, Cultured , Humans , Mammary Glands, Human/drug effects , Stem Cells/drug effects
7.
Cancer Res ; 73(23): 6886-99, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24145351

ABSTRACT

Mortality of patients with breast cancer is due overwhelmingly to metastatic spread of the disease. Although dissemination is an early event in breast cancer, extended periods of cancer cell dormancy can result in long latency of metastasis development. Deciphering the mechanisms underlying cancer cell dormancy and subsequent growth at the metastatic site would facilitate development of strategies to interfere with these processes. A challenge in this undertaking has been the lack of models for cancer cell dormancy. We have established novel experimental systems that model the bone microenvironment of the breast cancer metastatic niche. These systems are based on 3D cocultures of breast cancer cells with cell types predominant in bone marrow. We identified conditions in which cancer cells are dormant and conditions in which they proliferate. Dormant cancer cells were able to proliferate upon transfer into supportive microenvironment or upon manipulation of signaling pathways that control dormancy. These experimental systems will be instrumental for metastasis studies, particularly the study of cellular dormancy.


Subject(s)
Bone Neoplasms/secondary , Breast Neoplasms/pathology , Models, Biological , Neoplastic Stem Cells/physiology , Tumor Microenvironment/physiology , Animals , Bone Marrow Cells/physiology , Cell Cycle Checkpoints/physiology , Cells, Cultured , Female , Human Umbilical Vein Endothelial Cells/physiology , Humans , MCF-7 Cells , Mice , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/pathology , Stem Cell Niche/physiology , Stromal Cells/physiology
8.
Clin Cancer Res ; 19(6): 1512-24, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23340294

ABSTRACT

PURPOSE: Accumulating evidence supports the existence of breast cancer stem cells (BCSC), which are characterized by their capacity to self-renew and divide indefinitely and resistance to conventional therapies. The Notch pathway is important for stem cell renewal and is a potential target for BCSC-directed therapy. EXPERIMENTAL DESIGN: Using human breast tumorgraft studies, we evaluated the impact of gamma secretase inhibitors (GSI) on the BCSC population and the efficacy of combining GSI with docetaxel treatment. The mouse experimental therapy paralleled a concurrent clinical trial in patients with advanced breast cancer, designed to determine the maximum-tolerated dose of the GSI, MK-0752, administered sequentially with docetaxel, and to evaluate BCSC markers in serial tumor biopsies. RESULTS: Treatment with GSI reduced BCSCs in MC1 and BCM-2147 tumorgrafts by inhibition of the Notch pathway. GSI enhanced the efficacy of docetaxel in preclinical studies. In the clinical trial, 30 patients with advanced breast cancer were treated with escalating doses of MK-0752 plus docetaxel. Clinically, meaningful doses of both drugs were possible with manageable toxicity and preliminary evidence of efficacy. A decrease in CD44(+)/CD24(-), ALDH(+), and mammosphere-forming efficiency were observed in tumors of patients undergoing serial biopsies. CONCLUSIONS: These preclinical data show that pharmacologic inhibition of the Notch pathway can reduce BCSCs in breast tumorgraft models. The clinical trial shows feasibility of combination GSI and chemotherapy, and together these results encourage further study of Notch pathway inhibitors in combination with chemotherapy in breast cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Benzene Derivatives/administration & dosage , Breast Neoplasms/drug therapy , Propionates/administration & dosage , Sulfones/administration & dosage , Taxoids/administration & dosage , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Animals , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Benzene Derivatives/adverse effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Docetaxel , Female , Humans , Maximum Tolerated Dose , Mice , Neoplasm Staging , Propionates/adverse effects , Receptors, Notch/metabolism , Signal Transduction , Sulfones/adverse effects , Taxoids/adverse effects , Xenograft Model Antitumor Assays
9.
Sci Rep ; 2: 906, 2012.
Article in English | MEDLINE | ID: mdl-23205268

ABSTRACT

The bone marrow (BM) is a major organ of breast cancer (BC) dormancy and a common source of BC resurgence. Gap junctional intercellular communication (GJIC) between BC cells (BCCs) and BM stroma facilitates dormancy. This study reports on a hierarchy of BCCs with the most immature subset (Oct4(hi)/CD44(hi/med)/CD24(-/+)) demonstrating chemoresistance, dormancy, and stem cell properties: self-renewal, serial passaging ability, cycling quiescence, long doubling time, asymmetric division, high metastatic and invasive capability. In vitro and in vivo studies indicated that this subset was responsible for GJIC with BM stroma. Similar BCCs were detected in the blood of patients despite aggressive treatment and in a patient with a relatively large tumor but no lymph node involvement. In brief, these findings identified a novel BCC subset with stem cell properties, with preference for dormancy and in the circulation of patients. The findings establish a working cellular hierarchy of BCCs based on phenotype and functions.


Subject(s)
Biomarkers, Tumor/genetics , Bone Marrow Cells/pathology , Breast Neoplasms/pathology , Cell Communication/genetics , Gap Junctions/metabolism , Neoplastic Stem Cells/pathology , Stromal Cells/pathology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/metabolism , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , CD24 Antigen/genetics , CD24 Antigen/metabolism , Carboplatin/pharmacology , Carboplatin/therapeutic use , Cell Communication/drug effects , Cell Division , Cell Line, Tumor , Coculture Techniques , Female , Gap Junctions/genetics , Humans , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Mice , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Phenotype , Signal Transduction , Stromal Cells/drug effects , Stromal Cells/metabolism , Transfection
10.
J Natl Cancer Inst Monogr ; 2011(43): 147-51, 2011.
Article in English | MEDLINE | ID: mdl-22043063

ABSTRACT

A panel of international breast cancer experts formulated a declaration of consensus regarding many key issues in the use of primary systemic therapy (PST) either in clinical routine or research practice. The attainment of pathological complete response (pCR), defined as no residual invasive tumor in the surgical specimens both in breast and in axillary nodes, is one of the main goals of PST, and pCR can be used as the primary objective in prospective clinical trials. However, pCR is not a reliable endpoint with all treatment approaches, and alternatives such as Ki67 index of the residual invasive disease or after 2 weeks of PST are also potential endpoints. PST has several advantages: breast conservation and the unique opportunity to obtain information on the interaction between treatment and tumor biology. Changes in tumor biology after PST are an early phenomenon; so, an additional core biopsy performed after 14 days from treatment start should be considered in clinical trials.


Subject(s)
Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Neoadjuvant Therapy , Antineoplastic Agents, Hormonal/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biopsy/methods , Breast Neoplasms/pathology , Breast Neoplasms/surgery , Chemotherapy, Adjuvant , Clinical Trials as Topic , Female , Humans , Ki-67 Antigen/metabolism , Meta-Analysis as Topic , Neoadjuvant Therapy/methods , Palpation , Receptor, ErbB-2/metabolism , Remission Induction , Treatment Outcome , Ultrasonography, Mammary
11.
Clin Cancer Res ; 17(20): 6510-21, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21878538

ABSTRACT

PURPOSE: Tamoxifen (Tam) is the most prescribed hormonal agent for treatment of estrogen receptor α (ERα)-positive breast cancer patients. Using microarray analysis, we observed that metastatic breast tumors resistant to Tam therapy had elevated levels of Dicer. EXPERIMENTAL DESIGN: We overexpressed Dicer in ERα-positive MCF-7 human breast cancer cells and observed a concomitant increase in expression of the breast cancer resistance protein (BCRP). We thus hypothesized that Tam resistance associated with Dicer overexpression in ERα-positive breast cancer cells may involve BCRP. We analyzed BCRP function in Dicer-overexpressing cells using growth in soft agar and mammosphere formation and evaluated intracellular Tam efflux. RESULTS: In the presence of Tam, Dicer-overexpressing cells formed resistant colonies in soft agar, and treatment with BCRP inhibitors restored Tam sensitivity. Tumor xenograft studies confirmed that Dicer-overexpressing cells were resistant to Tam in vivo. Tumors and distant metastases could be initiated with as few as five mammosphere cells from both vector and Dicer-overexpressing cells, indicating that the mammosphere assay selected for cells with enhanced tumor-initiating and metastatic capacity. Dicer-overexpressing cells with elevated levels of BCRP effluxed Tam more efficiently than control cells, and BCRP inhibitors were able to inhibit efflux. CONCLUSION: Dicer-overexpressing breast cancer cells enriched for cells with enhanced BCRP function. We hypothesize that it is this population which may be involved in the emergence of Tam-resistant growth. BCRP may be a novel clinical target to restore Tam sensitivity.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Antineoplastic Agents, Hormonal/therapeutic use , DEAD-box RNA Helicases/pharmacology , Drug Resistance, Neoplasm/genetics , Estrogen Antagonists/therapeutic use , Neoplasm Proteins/metabolism , Ribonuclease III/pharmacology , Tamoxifen/therapeutic use , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Animals , Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Line, Tumor , Disease Models, Animal , Estrogen Receptor alpha/metabolism , Female , Humans , Mice , Mice, Nude , Neoplasms, Hormone-Dependent/genetics , Tamoxifen/pharmacology , Up-Regulation
12.
Methods Mol Biol ; 731: 471-82, 2011.
Article in English | MEDLINE | ID: mdl-21516430

ABSTRACT

Three experimental systems based on mouse models are currently used to study breast cancer: transgenic mice, carcinogen-induced models, and xenografts of breast cancers. Each of these models has advantages and limitations. This chapter focuses on xenotransplantation of breast cancers and reviews the techniques used so far in establishing this model, the advantages and limitations compared to other experimental systems, and finally, the technical questions that remain to be answered.


Subject(s)
Breast Neoplasms/pathology , Neoplasm Transplantation/methods , Transplantation, Heterologous/methods , Adipose Tissue/surgery , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic , Female , Humans , Mice , Transplantation, Heterologous/immunology
13.
Cancer Res ; 71(2): 614-24, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21224357

ABSTRACT

We have used in vitro and mouse xenograft models to examine the interaction between breast cancer stem cells (CSC) and bone marrow-derived mesenchymal stem cells (MSC). We show that both of these cell populations are organized in a cellular hierarchy in which primitive aldehyde dehydrogenase expressing mesenchymal cells regulate breast CSCs through cytokine loops involving IL6 and CXCL7. In NOD/SCID mice, labeled MSCs introduced into the tibia traffic to sites of growing breast tumor xenografts where they accelerated tumor growth by increasing the breast CSC population. With immunochemistry, we identified MSC-CSC niches in these tumor xenografts as well as in frozen sections from primary human breast cancers. Bone marrow-derived MSCs may accelerate human breast tumor growth by generating cytokine networks that regulate the CSC population.


Subject(s)
Breast Neoplasms/pathology , Cell Communication/physiology , Cytokines/metabolism , Mesenchymal Stem Cells/pathology , Neoplastic Stem Cells/pathology , Aldehyde Dehydrogenase/analysis , Aldehyde Dehydrogenase/biosynthesis , Animals , Bone Marrow Cells/cytology , Breast Neoplasms/enzymology , Breast Neoplasms/metabolism , Female , Humans , Mesenchymal Stem Cells/enzymology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/enzymology , Neoplastic Stem Cells/metabolism , Transplantation, Heterologous
14.
Breast Cancer Res ; 12(1): 301, 2010.
Article in English | MEDLINE | ID: mdl-20122290

ABSTRACT

Recent studies from Clarke's group published in the journal Cell indicate that miRNAs may be the elusive universal stem cell markers that the field of cancer stem cell biology has been seeking. Distinct profiles of miRNAs appear to reflect the state of cell differentiation not only in breast cancer cells, but also in normal mammary epithelial cells. Moreover, they are conserved across tissues and species. The authors of this work also show evidence that downregulation of miRNA-200c in normal and malignant breast stem cells and in embryonal carcinoma cells has functional relevance, being responsible for the proliferative potential of these cells in vitro and in vivo.


Subject(s)
Breast Neoplasms/pathology , MicroRNAs/physiology , Neoplastic Stem Cells/pathology , Animals , Female , Humans , MicroRNAs/antagonists & inhibitors
15.
J Clin Invest ; 120(2): 485-97, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20051626

ABSTRACT

Recent evidence suggests that breast cancer and other solid tumors possess a rare population of cells capable of extensive self-renewal that contribute to metastasis and treatment resistance. We report here the development of a strategy to target these breast cancer stem cells (CSCs) through blockade of the IL-8 receptor CXCR1. CXCR1 blockade using either a CXCR1-specific blocking antibody or repertaxin, a small-molecule CXCR1 inhibitor, selectively depleted the CSC population in 2 human breast cancer cell lines in vitro. Furthermore, this was followed by the induction of massive apoptosis in the bulk tumor population via FASL/FAS signaling. The effects of CXCR1 blockade on CSC viability and on FASL production were mediated by the FAK/AKT/FOXO3A pathway. In addition, repertaxin was able to specifically target the CSC population in human breast cancer xenografts, retarding tumor growth and reducing metastasis. Our data therefore suggest that CXCR1 blockade may provide a novel means of targeting and eliminating breast CSCs.


Subject(s)
Breast Neoplasms/pathology , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/transplantation , Receptors, Interleukin-8A/antagonists & inhibitors , Stem Cells/pathology , Animals , Cell Division/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Fas Ligand Protein/genetics , Female , Humans , Mice , RNA, Messenger/genetics , Receptors, Interleukin-8A/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Sulfonamides/pharmacology , Transplantation, Heterologous/pathology
16.
Breast Cancer Res Treat ; 122(3): 777-85, 2010 Aug.
Article in English | MEDLINE | ID: mdl-19898931

ABSTRACT

The cancer stem cell hypothesis asserts that malignancies arise in tissue stem and/or progenitor cells through the dysregulation or acquisition of self-renewal. In order to determine whether the dietary polyphenols, curcumin, and piperine are able to modulate the self-renewal of normal and malignant breast stem cells, we examined the effects of these compounds on mammosphere formation, expression of the breast stem cell marker aldehyde dehydrogenase (ALDH), and Wnt signaling. Mammosphere formation assays were performed after curcumin, piperine, and control treatment in unsorted normal breast epithelial cells and normal stem and early progenitor cells, selected by ALDH positivity. Wnt signaling was examined using a Topflash assay. Both curcumin and piperine inhibited mammosphere formation, serial passaging, and percent of ALDH+ cells by 50% at 5 microM and completely at 10 microM concentration in normal and malignant breast cells. There was no effect on cellular differentiation. Wnt signaling was inhibited by both curcumin and piperine by 50% at 5 microM and completely at 10 microM. Curcumin and piperine separately, and in combination, inhibit breast stem cell self-renewal but do not cause toxicity to differentiated cells. These compounds could be potential cancer preventive agents. Mammosphere formation assays may be a quantifiable biomarker to assess cancer preventive agent efficacy and Wnt signaling assessment can be a mechanistic biomarker for use in human clinical trials.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Breast/pathology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Aldehyde Dehydrogenase/metabolism , Alkaloids/administration & dosage , Benzodioxoles/administration & dosage , Breast/drug effects , Breast/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Curcumin/administration & dosage , Female , Humans , Immunoenzyme Techniques , Neoplastic Stem Cells/metabolism , Piperidines/administration & dosage , Polyunsaturated Alkamides/administration & dosage , Signal Transduction/drug effects , Wnt Proteins/metabolism
17.
Clin Cancer Res ; 16(1): 45-55, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-20028757

ABSTRACT

PURPOSE: To examine the role of cancer stem cells (CSC) in mediating metastasis in inflammatory breast cancer (IBC) and the association of these cells with patient outcome in this aggressive type of breast cancer. EXPERIMENTAL DESIGN: CSCs were isolated from SUM149 and MARY-X, an IBC cell line and primary xenograft, by virtue of increased aldehyde dehydrogenase (ALDH) activity as assessed by the ALDEFLUOR assay. Invasion and metastasis of CSC populations were assessed by in vitro and mouse xenograft assays. Expression of ALDH1 was determined on a retrospective series of 109 IBC patients and this was correlated with histoclinical data. All statistical tests were two sided. Log-rank tests using Kaplan-Meier analysis were used to determine the correlation of ALDH1 expression with development of metastasis and patient outcome. RESULTS: Both in vitro and xenograft assays showed that invasion and metastasis in IBC are mediated by a cellular component that displays ALDH activity. Furthermore, expression of ALDH1 in IBC was an independent predictive factor for early metastasis and decreased survival in this patient population. CONCLUSIONS: These results suggest that the metastatic, aggressive behavior of IBC may be mediated by a CSC component that displays ALDH enzymatic activity. ALDH1 expression represents the first independent prognostic marker to predict metastasis and poor patient outcome in IBC. The results illustrate how stem cell research can translate into clinical practice in the IBC field.


Subject(s)
Aldehyde Dehydrogenase/metabolism , Breast Neoplasms/enzymology , Inflammation/enzymology , Isoenzymes/metabolism , Neoplasm Metastasis , Neoplastic Stem Cells/enzymology , Aldehyde Dehydrogenase 1 Family , Animals , Biomarkers, Tumor/metabolism , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Retinal Dehydrogenase , Treatment Outcome
19.
Cancer Res ; 69(8): 3382-9, 2009 Apr 15.
Article in English | MEDLINE | ID: mdl-19336570

ABSTRACT

Although the concept that cancers originate from stem cells (SC) is becoming scientifically accepted, mechanisms by which SC contribute to tumor initiation and progression are largely unknown. For colorectal cancer (CRC), investigation of this problem has been hindered by a paucity of specific markers for identification and isolation of SC from normal and malignant colon. Accordingly, aldehyde dehydrogenase 1 (ALDH1) was investigated as a possible marker for identifying colonic SC and for tracking them during cancer progression. Immunostaining showed that ALDH1(+) cells are sparse and limited to the normal crypt bottom, where SCs reside. During progression from normal epithelium to mutant (APC) epithelium to adenoma, ALDH1(+) cells increased in number and became distributed farther up the crypt. CD133(+) and CD44(+) cells, which are more numerous and broadly distributed in normal crypts, showed similar changes during tumorigenesis. Flow cytometric isolation of cancer cells based on enzymatic activity of ALDH (Aldefluor assay) and implantation of these cells in nonobese diabetic-severe combined immunodeficient mice (a) generated xenograft tumors (Aldefluor(-) cells did not), (b) generated them after implanting as few as 25 cells, and (c) generated them dose dependently. Further isolation of cancer cells using a second marker (CD44(+) or CD133(+) serially) only modestly increased enrichment based on tumor-initiating ability. Thus, ALDH1 seems to be a specific marker for identifying, isolating, and tracking human colonic SC during CRC development. These findings also support our original hypothesis, derived previously from mathematical modeling of crypt dynamics, that progressive colonic SC overpopulation occurs during colon tumorigenesis and drives CRC development.


Subject(s)
Aldehyde Dehydrogenase/biosynthesis , Cell Transformation, Neoplastic/metabolism , Colon/enzymology , Colonic Neoplasms/enzymology , Isoenzymes/biosynthesis , Stem Cells/enzymology , AC133 Antigen , Aldehyde Dehydrogenase 1 Family , Animals , Antigens, CD/biosynthesis , Biomarkers, Tumor , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/pathology , Colon/immunology , Colon/pathology , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Epithelial Cells/enzymology , Epithelial Cells/immunology , Epithelial Cells/pathology , Flow Cytometry , Glycoproteins/biosynthesis , Humans , Hyaluronan Receptors/biosynthesis , Immunohistochemistry , Mice , Mice, Inbred NOD , Mice, SCID , Peptides , Retinal Dehydrogenase , Stem Cells/immunology , Stem Cells/pathology , Transplantation, Heterologous
20.
Cancer Res ; 69(4): 1302-13, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-19190339

ABSTRACT

Tumors may be initiated and maintained by a cellular subcomponent that displays stem cell properties. We have used the expression of aldehyde dehydrogenase as assessed by the ALDEFLUOR assay to isolate and characterize cancer stem cell (CSC) populations in 33 cell lines derived from normal and malignant mammary tissue. Twenty-three of the 33 cell lines contained an ALDEFLUOR-positive population that displayed stem cell properties in vitro and in NOD/SCID xenografts. Gene expression profiling identified a 413-gene CSC profile that included genes known to play a role in stem cell function, as well as genes such as CXCR1/IL-8RA not previously known to play such a role. Recombinant interleukin-8 (IL-8) increased mammosphere formation and the ALDEFLUOR-positive population in breast cancer cell lines. Finally, we show that ALDEFLUOR-positive cells are responsible for mediating metastasis. These studies confirm the hierarchical organization of immortalized cell lines, establish techniques that can facilitate the characterization of regulatory pathways of CSCs, and identify potential stem cell markers and therapeutic targets.


Subject(s)
Breast Neoplasms/pathology , Breast/cytology , Neoplasm Metastasis/pathology , Stem Cells/pathology , Aldehyde Dehydrogenase/genetics , Animals , Biomarkers, Tumor/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Line , Cell Line, Tumor , Female , Flow Cytometry , Gene Expression Profiling , Homeostasis , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , RNA, Neoplasm/genetics , RNA, Neoplasm/isolation & purification , Receptors, Interleukin-8A/genetics , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/cytology , Stem Cells/physiology
SELECTION OF CITATIONS
SEARCH DETAIL