Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 45
1.
Cancers (Basel) ; 13(18)2021 Sep 08.
Article En | MEDLINE | ID: mdl-34572749

Reprogramming the tumor stroma is an emerging approach to circumventing the challenges of conventional cancer therapies. This strategy, however, is hampered by the lack of a specific molecular target. We previously reported that stromal fibroblasts (FBs) with high expression of CD36 could be utilized for this purpose. These studies are now expanded to identify the secreted factors responsible for tumor suppression. Methodologies included 3D colonies, fluorescent microscopy coupled with quantitative techniques, proteomics profiling, and bioinformatics analysis. The results indicated that the conditioned medium (CM) of the CD36+ FBs caused growth suppression via apoptosis in the triple-negative cell lines of MDA-MB-231, BT549, and Hs578T, but not in the ERBB2+ SKBR3. Following the proteomics and bioinformatic analysis of the CM of CD36+ versus CD36- FBs, we determined KLF10 as one of the transcription factors responsible for growth suppression. We also identified FBLN1, SLIT3, and PENK as active ligands, where their minimum effective concentrations were determined. Finally, in MDA-MB-231, we showed that a mixture of FBLN1, SLIT3, and PENK could induce an amount of growth suppression similar to the CM of CD36+ FBs. In conclusion, our findings suggest that these ligands, secreted by CD36+ FBs, can be targeted for breast cancer treatment.

2.
Biochem Biophys Res Commun ; 526(1): 41-47, 2020 05 21.
Article En | MEDLINE | ID: mdl-32192771

Human breast tumors are not fully autonomous. They are dependent on nutrients and growth-promoting signals provided by the supporting stromal cells. Within the tumor microenvironment, one of the secreted macromolecules by tumor cells is activin A, where we show to downregulate CD36 in fibroblasts. Downregulation of CD36 in fibroblasts also increases the secretion of activin A by fibroblasts. We hypothesize that overexpression of CD36 in fibroblasts inhibits the formation of solid tumors in subtypes of breast cancer models. For the first time, we show that co-culturing organoid models of breast cancer cell lines of MDA-MB-231 (e.g., a triple-negative line) or MCF7 (e.g., a luminal-A line) with CD36+ fibroblasts inhibit the growth and normalizes basal and lateral polarities, respectively. In the long-term anchorage-independent growth assay, the rate of colony formation is also reduced for MDA-MB-231. These observations are consistent with the mechanism of tumor suppression involving the downregulation of pSMAD2/3 and YY1 expression levels. Our integrated analytical methods leverage and extend quantitative assays at cell- and colony-scales in both short- and long-term cultures using brightfield or immunofluorescent microscopy and robust image analysis. Conditioned media are profiled with the ELISA assay.


Breast Neoplasms/metabolism , Breast Neoplasms/pathology , CD36 Antigens/metabolism , Fibroblasts/metabolism , Mammary Glands, Human/pathology , Activins/pharmacology , Cell Line, Tumor , Cell Polarity/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Down-Regulation/drug effects , Female , Fibroblasts/drug effects , Fibroblasts/pathology , Humans , Phosphorylation/drug effects , Smad Proteins/metabolism , Tumor Stem Cell Assay , YY1 Transcription Factor/metabolism
3.
Breast Cancer Res Treat ; 176(1): 131-140, 2019 Jul.
Article En | MEDLINE | ID: mdl-30993572

PURPOSE: Understanding how differentiation, microenvironment, and hormonal milieu influence human breast cell susceptibility to malignant transformation will require the use of physiologically relevant in vitro systems. We sought to develop a 3D culture model that enables the propagation of normal estrogen receptor alpha (ER) + cells. METHODS: We tested soluble factors and protocols for the ability to maintain progenitor and ER + cells in cultures established from primary cells. Optimized conditions were then used to profile estrogen-induced gene expression changes in cultures from three pathology-free individuals. RESULTS: Long-term representation of ER + cells was optimal in medium that included three different TGFß/activin receptor-like kinase inhibitors. We found that omitting the BMP signaling antagonist, Noggin, enhanced the responsiveness of the PGR gene to estradiol exposure without altering the proportions of ER + cells in the cultures. Profiling of estradiol-exposed cultures showed that while all the cultures showed immediate and robust induction of PGR, LRP2, and IGFB4, other responses varied qualitatively and quantitatively across specimens. CONCLUSIONS: We successfully identified conditions for the maintenance and propagation of functional ER + cells from normal human breast tissues. We propose that these 3D cultures will overcome limitations of conventional 2D cultures of partially or fully transformed cell lines by sustaining normal endocrine function and growth regulation of the cell populations that comprise intact breasts.


Breast Neoplasms/metabolism , Receptors, Estrogen/metabolism , Biomarkers, Tumor , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic , Humans , Neoplastic Stem Cells/metabolism , Receptors, Estrogen/genetics , Spheroids, Cellular , Transcriptional Activation , Tumor Cells, Cultured
4.
SLAS Discov ; 22(3): 298-308, 2017 03.
Article En | MEDLINE | ID: mdl-27872202

Malignant melanomas often arise from nevi, which result from initial oncogene-induced hyperproliferation of melanocytes that are maintained in a CDKN2A/p16-mediated senescent state. Thus, genes that can bypass this senescence barrier are likely to contribute to melanoma development. We have performed a gain-of-function screen of 17,030 lentivirally expressed human open reading frames (ORFs) in a melanoma cell line containing an inducible p16 construct to identify such genes. Genes known to bypass p16-induced senescence arrest, including the human papilloma virus 18 E7 gene ( HPV18E7), and genes such as the p16-binding CDK6 with expected functions, as well as panel of novel genes, were identified, including high-mobility group box (HMGB) proteins. A number of these were further validated in two other models of p16-induced senescence. Tissue immunohistochemistry demonstrated higher levels of CDK6 in primary melanomas compared with normal skin and nevi. Reduction of CDK6 levels drove melanoma cells expressing functional p16 into senescence, demonstrating its contribution to bypass senescence.


Cell Cycle Checkpoints , Cyclin-Dependent Kinase 6/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Gene Expression Regulation, Neoplastic , High-Throughput Screening Assays , Melanocytes/metabolism , Cell Line, Tumor , Cellular Senescence , Cyclin-Dependent Kinase 6/metabolism , Cyclin-Dependent Kinase Inhibitor p16/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Library , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Genome, Human , HEK293 Cells , HMGB Proteins/genetics , HMGB Proteins/metabolism , Humans , Lentivirus/genetics , Lentivirus/metabolism , Melanocytes/pathology , Melanoma/genetics , Melanoma/metabolism , Melanoma/pathology , Nevus/genetics , Nevus/metabolism , Nevus/pathology , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/metabolism , Open Reading Frames , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Skin Neoplasms/pathology
5.
Nat Med ; 22(11): 1321-1329, 2016 11.
Article En | MEDLINE | ID: mdl-27775705

Triple-negative breast cancer (TNBC), in which cells lack expression of the estrogen receptor (ER), the progesterone receptor (PR) and the ERBB2 (also known as HER2) receptor, is the breast cancer subtype with the poorest outcome. No targeted therapy is available against this subtype of cancer owing to a lack of validated molecular targets. We previously reported that signaling involving MYC-an essential, pleiotropic transcription factor that regulates the expression of hundreds of genes-is disproportionally higher in triple-negative (TN) tumors than in receptor-positive (RP) tumors. Direct inhibition of the oncogenic transcriptional activity of MYC has been challenging to achieve. Here, by conducting a shRNA screen targeting the kinome, we identified PIM1, a non-essential serine-threonine kinase, in a synthetic lethal interaction with MYC. PIM1 expression was higher in TN tumors than in RP tumors and was associated with poor prognosis in patients with hormone- and HER2-negative tumors. Small-molecule PIM kinase inhibitors halted the growth of human TN tumors with elevated MYC expression in patient-derived tumor xenograft (PDX) and MYC-driven transgenic mouse models of breast cancer by inhibiting the oncogenic transcriptional activity of MYC and restoring the function of the endogenous cell cycle inhibitor, p27. Our findings warrant clinical evaluation of PIM kinase inhibitors in patients with TN tumors that have elevated MYC expression.


Carcinoma, Ductal, Breast/metabolism , Mammary Neoplasms, Experimental/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Triple Negative Breast Neoplasms/metabolism , Animals , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Female , Humans , In Situ Nick-End Labeling , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/genetics , Mice, Transgenic , Microscopy, Fluorescence , Prognosis , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-pim-1/metabolism , RNA, Small Interfering , Real-Time Polymerase Chain Reaction , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Triple Negative Breast Neoplasms/drug therapy , Xenograft Model Antitumor Assays
6.
Environ Health Perspect ; 124(5): 563-9, 2016 05.
Article En | MEDLINE | ID: mdl-26502914

BACKGROUND: Xenoestrogens are synthetic compounds that mimic endogenous estrogens by binding to and activating estrogen receptors. Exposure to estrogens and to some xenoestrogens has been associated with cell proliferation and an increased risk of breast cancer. Despite evidence of estrogenicity, parabens are among the most widely used xenoestrogens in cosmetics and personal-care products and are generally considered safe. However, previous cell-based studies with parabens do not take into account the signaling cross-talk between estrogen receptor α (ERα) and the human epidermal growth factor receptor (HER) family. OBJECTIVES: We investigated the hypothesis that the potency of parabens can be increased with HER ligands, such as heregulin (HRG). METHODS: The effects of HER ligands on paraben activation of c-Myc expression and cell proliferation were determined by real-time polymerase chain reaction, Western blots, flow cytometry, and chromatin immunoprecipitation assays in ERα- and HER2-positive human BT-474 breast cancer cells. RESULTS: Butylparaben (BP) and HRG produced a synergistic increase in c-Myc mRNA and protein levels in BT-474 cells. Estrogen receptor antagonists blocked the synergistic increase in c-Myc protein levels. The combination of BP and HRG also stimulated proliferation of BT-474 cells compared with the effects of BP alone. HRG decreased the dose required for BP-mediated stimulation of c-Myc mRNA expression and cell proliferation. HRG caused the phosphorylation of serine 167 in ERα. BP and HRG produced a synergistic increase in ERα recruitment to the c-Myc gene. CONCLUSION: Our results show that HER ligands enhanced the potency of BP to stimulate oncogene expression and breast cancer cell proliferation in vitro via ERα, suggesting that parabens might be active at exposure levels not previously considered toxicologically relevant from studies testing their effects in isolation. CITATION: Pan S, Yuan C, Tagmount A, Rudel RA, Ackerman JM, Yaswen P, Vulpe CD, Leitman DC. 2016. Parabens and human epidermal growth factor receptor ligand cross-talk in breast cancer cells. Environ Health Perspect 124:563-569; http://dx.doi.org/10.1289/ehp.1409200.


Estrogens/toxicity , Parabens/toxicity , Cell Line, Tumor , ErbB Receptors/metabolism , Estrogen Receptor alpha/metabolism , Genes, myc , Humans , Neuregulin-1/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism
7.
Mol Cancer Ther ; 14(12): 2805-17, 2015 Dec.
Article En | MEDLINE | ID: mdl-26438156

The requisite role of HER3 in HER2-amplified cancers is beyond what would be expected as a dimerization partner or effector substrate and it exhibits a substantial degree of resiliency that mitigates the effects of HER2-inhibitor therapies. To better understand the roots of this resiliency, we conducted an in-depth chemical-genetic interrogation of the signaling network downstream of HER3. A unique attribute of these tumors is the deregulation of TORC2. The upstream signals that ordinarily maintain TORC2 signaling are lost in these tumors, and instead TORC2 is driven by Akt. We find that in these cancers HER3 functions as a buffering arm of an Akt-TORC2 feed-forward loop that functions as a self-perpetuating module. This network topology alters the role of HER3 from a conditionally engaged ligand-driven upstream physiologic signaling input to an essential component of a concentric signaling throughput highly competent at preservation of homeostasis. The competence of this signaling topology is evident in its response to perturbation at any of its nodes. Thus, a critical pathophysiologic event in the evolution of HER2-amplified cancers is the loss of the input signals that normally drive TORC2 signaling, repositioning it under Akt dependency, and fundamentally altering the role of HER3. This reprogramming of the downstream network topology is a key aspect in the pathogenesis of HER2-amplified cancers and constitutes a formidable barrier in the targeted therapy of these cancers.


Breast Neoplasms/genetics , Multiprotein Complexes/genetics , Oncogene Protein v-akt/genetics , Receptor, ErbB-2/genetics , Receptor, ErbB-3/genetics , TOR Serine-Threonine Kinases/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Female , Humans , Mechanistic Target of Rapamycin Complex 2 , Multiprotein Complexes/biosynthesis , Oncogene Protein v-akt/biosynthesis , Protein Multimerization/genetics , Receptor, ErbB-2/biosynthesis , Receptor, ErbB-3/biosynthesis , Signal Transduction/genetics , TOR Serine-Threonine Kinases/biosynthesis
8.
Nature ; 526(7571): 131-5, 2015 Oct 01.
Article En | MEDLINE | ID: mdl-26416748

Despite major advances in understanding the molecular and genetic basis of cancer, metastasis remains the cause of >90% of cancer-related mortality. Understanding metastasis initiation and progression is critical to developing new therapeutic strategies to treat and prevent metastatic disease. Prevailing theories hypothesize that metastases are seeded by rare tumour cells with unique properties, which may function like stem cells in their ability to initiate and propagate metastatic tumours. However, the identity of metastasis-initiating cells in human breast cancer remains elusive, and whether metastases are hierarchically organized is unknown. Here we show at the single-cell level that early stage metastatic cells possess a distinct stem-like gene expression signature. To identify and isolate metastatic cells from patient-derived xenograft models of human breast cancer, we developed a highly sensitive fluorescence-activated cell sorting (FACS)-based assay, which allowed us to enumerate metastatic cells in mouse peripheral tissues. We compared gene signatures in metastatic cells from tissues with low versus high metastatic burden. Metastatic cells from low-burden tissues were distinct owing to their increased expression of stem cell, epithelial-to-mesenchymal transition, pro-survival, and dormancy-associated genes. By contrast, metastatic cells from high-burden tissues were similar to primary tumour cells, which were more heterogeneous and expressed higher levels of luminal differentiation genes. Transplantation of stem-like metastatic cells from low-burden tissues showed that they have considerable tumour-initiating capacity, and can differentiate to produce luminal-like cancer cells. Progression to high metastatic burden was associated with increased proliferation and MYC expression, which could be attenuated by treatment with cyclin-dependent kinase (CDK) inhibitors. These findings support a hierarchical model for metastasis, in which metastases are initiated by stem-like cells that proliferate and differentiate to produce advanced metastatic disease.


Breast Neoplasms/pathology , Disease Progression , Neoplasm Metastasis/pathology , Neoplastic Stem Cells/pathology , Single-Cell Analysis , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Cycle/drug effects , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Separation , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/pathology , Cyclin-Dependent Kinases/antagonists & inhibitors , Disease Models, Animal , Epithelial Cells/drug effects , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition/genetics , Flow Cytometry , Gene Expression Profiling , Genes, myc/genetics , Humans , Mesoderm/metabolism , Mesoderm/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Metastasis/drug therapy , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Xenograft Model Antitumor Assays
9.
Semin Cancer Biol ; 35 Suppl: S104-S128, 2015 Dec.
Article En | MEDLINE | ID: mdl-25869441

One of the hallmarks of malignant cell populations is the ability to undergo continuous proliferation. This property allows clonal lineages to acquire sequential aberrations that can fuel increasingly autonomous growth, invasiveness, and therapeutic resistance. Innate cellular mechanisms have evolved to regulate replicative potential as a hedge against malignant progression. When activated in the absence of normal terminal differentiation cues, these mechanisms can result in a state of persistent cytostasis. This state, termed "senescence," can be triggered by intrinsic cellular processes such as telomere dysfunction and oncogene expression, and by exogenous factors such as DNA damaging agents or oxidative environments. Despite differences in upstream signaling, senescence often involves convergent interdependent activation of tumor suppressors p53 and p16/pRB, but can be induced, albeit with reduced sensitivity, when these suppressors are compromised. Doses of conventional genotoxic drugs required to achieve cancer cell senescence are often much lower than doses required to achieve outright cell death. Additional therapies, such as those targeting cyclin dependent kinases or components of the PI3K signaling pathway, may induce senescence specifically in cancer cells by circumventing defects in tumor suppressor pathways or exploiting cancer cells' heightened requirements for telomerase. Such treatments sufficient to induce cancer cell senescence could provide increased patient survival with fewer and less severe side effects than conventional cytotoxic regimens. This positive aspect is countered by important caveats regarding senescence reversibility, genomic instability, and paracrine effects that may increase heterogeneity and adaptive resistance of surviving cancer cells. Nevertheless, agents that effectively disrupt replicative immortality will likely be valuable components of new combinatorial approaches to cancer therapy.


Cell Proliferation/genetics , Cellular Senescence/genetics , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/genetics , Antineoplastic Agents/therapeutic use , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/genetics , Genomic Instability/drug effects , Humans , Neoplasms/pathology , Phosphatidylinositol 3-Kinases/genetics , Signal Transduction/genetics , Telomerase/drug effects , Telomerase/genetics , Tumor Suppressor Protein p53/genetics
10.
Nat Commun ; 6: 6927, 2015 Apr 21.
Article En | MEDLINE | ID: mdl-25896888

Clinically relevant human culture models are essential for developing effective therapies and exploring the biology and etiology of human cancers. Current breast tumour models, such as those from oncogenically transformed primary breast cells, produce predominantly basal-like properties, whereas the more common phenotype expressed by the vast majority of breast tumours are luminal. Reasons for this puzzling, yet important phenomenon, are not understood. We show here that luminal epithelial cells are significantly more resistant to viral transduction than their myoepithelial counterparts. We suggest that this is a significant barrier to generating luminal cell lines and experimental tumours in vivo and to accurate interpretation of results. We show that the resistance is due to lower affinity of luminal cells for virus attachment, which can be overcome by pretreating cells--or virus--with neuraminidase. We present an analytical method for quantifying transductional differences between cell types and an optimized protocol for transducing unsorted primary human breast cells in context.


Breast Neoplasms/metabolism , Cell Lineage/physiology , Models, Biological , Breast/cytology , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic/physiology , Humans , Lentivirus , Neuraminidase/metabolism , Transduction, Genetic
11.
Cell Cycle ; 13(24): 3818-22, 2014.
Article En | MEDLINE | ID: mdl-25483053

Deregulated cell cycle progression can often be traced to intrinsic defects in specific regulatory proteins in cancer cells. Knowledge of these primary defects has led to targeted approaches that exploit the defects and spare normal cells. However, the success of such targeted approaches is still hit-or-miss. Genetic and epigenetic variability inherent in most tumors often results in phenotypic heterogeneity that, in turn, results in de novo or acquired resistance to therapeutic agents. The ability of cells to compensate and adapt to the inhibition of a specific cell cycle mediator is not remarkable. What is novel and of great potential importance is that the ability of cells to exhibit such adaptability varies markedly. "Phenotypic stability factors" that restrict the ability of cells to undergo epithelial-mesenchymal transitions (EMT) may dictate the success or failure of targeted therapies by interfering with compensatory changes such as deregulation of CDK2 activity. Identification of existing and new agents that induce and maintain phenotypic stability factors will inform and enable synergistic approaches to the eradication of even the most aggressive tumors.


Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cyclin D/genetics , Cyclin D/metabolism , Cyclin E/genetics , Cyclin E/metabolism , Cyclin-Dependent Kinase 2/metabolism , Epithelial-Mesenchymal Transition , Female , Humans , NF-kappa B/metabolism , Neoplastic Stem Cells/metabolism , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Receptors, Notch/chemistry , Receptors, Notch/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism
12.
Cancer Epidemiol Biomarkers Prev ; 21(10): 1735-44, 2012 Oct.
Article En | MEDLINE | ID: mdl-22859400

BACKGROUND: Age is the strongest breast cancer risk factor, with overall breast cancer risk increasing steadily beginning at approximately 30 years of age. However, while breast cancer risk is lower among younger women, young women's breast cancer may be more aggressive. Although, several genomic and epidemiologic studies have shown higher prevalence of aggressive, estrogen-receptor negative breast cancer in younger women, the age-related gene expression that predisposes to these tumors is poorly understood. Characterizing age-related patterns of gene expression in normal breast tissues may provide insights on etiology of distinct breast cancer subtypes that arise from these tissues. METHODS: To identify age-related changes in normal breast tissue, 96 tissue specimens from patients with reduction mammoplasty, ages 14 to 70 years, were assayed by gene expression microarray. RESULTS: Significant associations between gene expression levels and age were identified for 802 probes (481 increased, 321 decreased with increasing age). Enriched functions included "aging of cells," "shape change," and "chemotaxis," and enriched pathways included Wnt/beta-catenin signaling, Ephrin receptor signaling, and JAK/Stat signaling. Applying the age-associated genes to publicly available tumor datasets, the age-associated pathways defined two groups of tumors with distinct survival. CONCLUSION: The hazard rates of young-like tumors mirrored that of high-grade tumors in the Surveillance, Epidemiology, and End Results Program, providing a biologic link between normal aging and age-related tumor aggressiveness. IMPACT: These data show that studies of normal tissue gene expression can yield important insights about the pathways and biologic pressures that are relevant during tumor etiology and progression.


Aging/metabolism , Breast Neoplasms/etiology , Breast/metabolism , Gene Expression Regulation, Neoplastic , Adolescent , Adult , Age Factors , Aged , Breast Neoplasms/metabolism , Female , Gene Expression , Humans , Incidence , Mammaplasty , Middle Aged , Signal Transduction
13.
Proc Natl Acad Sci U S A ; 109(39): E2595-604, 2012 Sep 25.
Article En | MEDLINE | ID: mdl-22923691

Breast cancer progression involves genetic changes and changes in the extracellular matrix (ECM). To test the importance of the ECM in tumor cell dissemination, we cultured epithelium from primary human breast carcinomas in different ECM gels. We used basement membrane gels to model the normal microenvironment and collagen I to model the stromal ECM. In basement membrane gels, malignant epithelium either was indolent or grew collectively, without protrusions. In collagen I, epithelium from the same tumor invaded with protrusions and disseminated cells. Importantly, collagen I induced a similar initial response of protrusions and dissemination in both normal and malignant mammary epithelium. However, dissemination of normal cells into collagen I was transient and ceased as laminin 111 localized to the basal surface, whereas dissemination of carcinoma cells was sustained throughout culture, and laminin 111 was not detected. Despite the large impact of ECM on migration strategy, transcriptome analysis of our 3D cultures revealed few ECM-dependent changes in RNA expression. However, we observed many differences between normal and malignant epithelium, including reduced expression of cell-adhesion genes in tumors. Therefore, we tested whether deletion of an adhesion gene could induce sustained dissemination of nontransformed cells into collagen I. We found that deletion of P-cadherin was sufficient for sustained dissemination, but exclusively into collagen I. Our data reveal that metastatic tumors preferentially disseminate in specific ECM microenvironments. Furthermore, these data suggest that breaks in the basement membrane could induce invasion and dissemination via the resulting direct contact between cancer cells and collagen I.


Breast Neoplasms , Cell Movement , Gene Expression Regulation, Neoplastic , Mammary Neoplasms, Animal , Tumor Microenvironment , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Female , Humans , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/metabolism , Mammary Neoplasms, Animal/pathology , Mice , Neoplasm Invasiveness
14.
Cancer Discov ; 2(7): 638-51, 2012 Jul.
Article En | MEDLINE | ID: mdl-22728437

UNLABELLED: The transcription factor ZNF217 is a candidate oncogene in the amplicon on chromosome 20q13 that occurs in 20% to 30% of primary human breast cancers and that correlates with poor prognosis. We show that Znf217 overexpression drives aberrant differentiation and signaling events, promotes increased self-renewal capacity, mesenchymal marker expression, motility, and metastasis, and represses an adult tissue stem cell gene signature downregulated in cancers. By in silico screening, we identified candidate therapeutics that at low concentrations inhibit growth of cancer cells expressing high ZNF217. We show that the nucleoside analogue triciribine inhibits ZNF217-induced tumor growth and chemotherapy resistance and inhibits signaling events [e.g., phospho-AKT, phospho-mitogen-activated protein kinase (MAPK)] in vivo. Our data suggest that ZNF217 is a biomarker of poor prognosis and a therapeutic target in patients with breast cancer and that triciribine may be part of a personalized treatment strategy in patients overexpressing ZNF217. Because ZNF217 is amplified in numerous cancers, these results have implications for other cancers. SIGNIFICANCE: This study finds that ZNF217 is a poor prognostic indicator and therapeutic target in patients with breast cancer and may be a strong biomarker of triciribine treatment efficacy in patients. Because previous clinical trials for triciribine did not include biomarkers of treatment efficacy, this study provides a rationale for revisiting triciribine in the clinical setting as a therapy for patients with breast cancer who overexpress ZNF217.


Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Trans-Activators/genetics , Animals , Antibiotics, Antineoplastic/pharmacology , Biomarkers, Tumor/metabolism , Blotting, Western , Breast Neoplasms/pathology , Breast Neoplasms/prevention & control , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Progression , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Profiling , Humans , MCF-7 Cells , Mice , NIH 3T3 Cells , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Oligonucleotide Array Sequence Analysis , Prognosis , Reverse Transcriptase Polymerase Chain Reaction , Ribonucleosides/pharmacology , Survival Analysis , Trans-Activators/metabolism , Xenograft Model Antitumor Assays
15.
J Exp Med ; 209(4): 679-96, 2012 Apr 09.
Article En | MEDLINE | ID: mdl-22430491

Estrogen, progesterone, and HER2 receptor-negative triple-negative breast cancers encompass the most clinically challenging subtype for which targeted therapeutics are lacking. We find that triple-negative tumors exhibit elevated MYC expression, as well as altered expression of MYC regulatory genes, resulting in increased activity of the MYC pathway. In primary breast tumors, MYC signaling did not predict response to neoadjuvant chemotherapy but was associated with poor prognosis. We exploit the increased MYC expression found in triple-negative breast cancers by using a synthetic-lethal approach dependent on cyclin-dependent kinase (CDK) inhibition. CDK inhibition effectively induced tumor regression in triple-negative tumor xenografts. The proapoptotic BCL-2 family member BIM is up-regulated after CDK inhibition and contributes to this synthetic-lethal mechanism. These results indicate that aggressive breast tumors with elevated MYC are uniquely sensitive to CDK inhibitors.


Breast Neoplasms/drug therapy , Cyclin-Dependent Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-myc/physiology , Signal Transduction/physiology , Animals , Apoptosis Regulatory Proteins/analysis , Apoptosis Regulatory Proteins/physiology , Bcl-2-Like Protein 11 , Breast Neoplasms/chemistry , Breast Neoplasms/mortality , Cell Line, Tumor , Female , Humans , Membrane Proteins/analysis , Membrane Proteins/physiology , Mice , Mice, Inbred BALB C , Prognosis , Proto-Oncogene Proteins/analysis , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-myc/analysis , Proto-Oncogene Proteins c-myc/genetics , Receptor, ErbB-2/analysis , Receptors, Estrogen/analysis , Receptors, Progesterone/analysis , Xenograft Model Antitumor Assays
16.
Cell Cycle ; 11(5): 1008-13, 2012 Mar 01.
Article En | MEDLINE | ID: mdl-22333593

RB family proteins pRb, p107 and p130 have similar structures and overlapping functions, enabling cell cycle arrest and cellular senescence. pRb, but not p107 or p130, is frequently mutated in human malignancies. In human fibroblasts acutely exposed to oncogenic ras, pRb has a specific role in suppressing DNA replication, and p107 or p130 cannot compensate for the loss of this function; however, a second p53/p21-dependent checkpoint prevents escape from growth arrest. This model of oncogene-induced senescence requires the additional loss of p53/p21 to explain selection for preferential loss of pRb function in human malignancies. We asked whether similar rules apply to the role of pRb in growth arrest of human epithelial cells, the source of most cancers. In two malignant human breast cancer cell lines, we found that individual RB family proteins were sufficient for the establishment of p16-initiated senescence, and that growth arrest in G 1 was not dependent on the presence of functional pRb or p53. However, senescence induction by endogenous p16 was delayed in primary normal human mammary epithelial cells with reduced pRb but not with reduced p107 or p130. Thus, under these circumstances, despite the presence of functional p53, p107 and p130 were unable to completely compensate for pRb in mediating senescence induction. We propose that early inactivation of pRb in pre-malignant breast cells can, by itself, extend proliferative lifespan, allowing acquisition of additional changes necessary for malignant transformation.


Cyclin-Dependent Kinase Inhibitor p16/metabolism , Retinoblastoma Protein/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cells, Cultured , Cellular Senescence , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , G1 Phase Cell Cycle Checkpoints , Humans , MCF-7 Cells , RNA Interference , RNA, Small Interfering/metabolism , Retinoblastoma Protein/antagonists & inhibitors , Retinoblastoma Protein/genetics , Retinoblastoma-Like Protein p107/antagonists & inhibitors , Retinoblastoma-Like Protein p107/genetics , Retinoblastoma-Like Protein p107/metabolism , Retinoblastoma-Like Protein p130/antagonists & inhibitors , Retinoblastoma-Like Protein p130/genetics , Retinoblastoma-Like Protein p130/metabolism , Tumor Suppressor Protein p53/metabolism
17.
Breast Cancer Res ; 13(3): 308, 2011 May 25.
Article En | MEDLINE | ID: mdl-21635707

Two recent reports identify ZNF703 as an oncogene driving selection of frequent chromosome 8p12 amplifications in luminal B breast tumors. The estrogen-responsive ZNF703 gene encodes a transcriptional cofactor that, when overexpressed, induces cell proliferation and interferes with transforming growth factor beta signaling. In MCF7 cells, increased ZNF703 expression results in activation of genes involved in stem cell self-renewal - while in primary human mammary epithelial cells, ZNF703 increases the ratio of luminal to basal progenitors. Expression of the murine homolog of ZNF703 reduces cell adhesion and promotes metastasis. ZNF703 overexpression thus alters regulation of proliferation and differentiation in luminal B tumors.


Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Transforming Growth Factor beta/metabolism , Animals , Cell Adhesion/genetics , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Chromosomes, Human, Pair 8/genetics , Female , Gene Amplification , Humans , Mice , Neoplasm Metastasis , Neoplastic Stem Cells/metabolism , Signal Transduction/genetics , Transcription Factors/biosynthesis
18.
Inorg Chem ; 50(1): 271-84, 2011 Jan 03.
Article En | MEDLINE | ID: mdl-21121684

The reactions of the breast cancer drug metabolite derivatives of tamoxifen, cis and trans-hydroxytamoxifen, cis-1 and trans-2, with [Cp*Rh(L)(3)](2+) complexes (L = H(2)O or MeOH), in CH(2)Cl(2) and CH(3)OH solvents, initially provided the kinetic η(1)-N complexes, cis-4 (OTf(-), CH(3)OH) and trans-5 (OTf(-), CH(3)OH), which underwent a novel, regioselective, intramolecular N-π rearrangement to give the cis and trans-η(6)-phenol substituted complexes, cis-6 and trans-7, via η(2)-N,O, η(1)-O, and ether aromatic ring η(6) intermediates. Recent density functional theory (DFT) calculations showed a preferred ground state for η(1)-N; η(2)-N,O; η(1)-O; and the η(6) complexes, including the prominent roles of the triflate anion (OTf(-)), and solvent molecules (CH(2)Cl(2) and CH(3)OH), and provided further steric, electronic, and thermodynamic data on the mechanism of the N-π rearrangement. The η(6) complex, cis-6, was shown to be an antagonist for ERα estrogen receptor binding, in a competition experiment with the female hormone, estradiol; therefore, computer docking studies of this biologically active complex at the estrogen receptors, ERα and ERß, also provided information on the binding modes and thermodynamic parameters, while bioassay results provided growth inhibition data on both hormone dependent and independent breast cancer cell lines.


Antineoplastic Agents, Hormonal , Estrogen Receptor alpha , Estrogen Receptor beta , Organometallic Compounds , Tamoxifen/analogs & derivatives , Antineoplastic Agents, Hormonal/chemical synthesis , Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/drug therapy , Cell Line, Tumor , Cell Proliferation/drug effects , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/antagonists & inhibitors , Estrogen Receptor beta/metabolism , Estrogens/metabolism , Female , Humans , Models, Molecular , Organometallic Compounds/chemical synthesis , Organometallic Compounds/pharmacology , Protein Binding , Rhodium/chemistry , Stereoisomerism , Tamoxifen/chemistry , Tamoxifen/pharmacology , Thermodynamics
19.
Genes Dev ; 24(24): 2800-11, 2010 Dec 15.
Article En | MEDLINE | ID: mdl-21159820

Organization into polarized three-dimensional structures defines whether epithelial cells are normal or malignant. In a model of morphogenesis, we show that inhibiting key signaling pathways in human breast cancer cells leads to "phenotypic reversion" of the malignant cells. Using architecture as an endpoint, we report that, in all cases, signaling through Raf/MEK/ERK disrupted tissue polarity via matrix metalloproteinase9 (MMP9) activity. Induction of Raf or activation of an engineered, functionally inducible MMP9 in nonmalignant cells led to loss of tissue polarity, and reinitiated proliferation. Conversely, inhibition of Raf or MMP9 with small molecule inhibitors or shRNAs restored the ability of cancer cells to form polarized quiescent structures. Silencing MMP9 expression also reduced tumor growth dramatically in a murine xenograft model. LC-MS/MS analysis comparing conditioned medium from nonmalignant cells with or without active MMP9 revealed laminin 111 (LM1) as an important target of MMP9. LM1 has been implicated in acinar morphogenesis; thus, its degradation by MMP9 provides a mechanism for loss of tissue polarity and reinitiation of growth associated with MMP9 activity. These findings underscore the importance of the dynamic reciprocity between the extracellular matrix integrity, tissue polarity, and Raf/MEK/ERK and MMP9 activities, providing an axis for either tissue homeostasis or malignant progression.


Breast Neoplasms/pathology , Breast/cytology , Matrix Metalloproteinase 9/metabolism , Morphogenesis , raf Kinases/physiology , Animals , Cell Culture Techniques , Cell Polarity , Cell Proliferation , Humans , Laminin/metabolism , Mice , Neoplasms, Experimental , Signal Transduction , Transplantation, Heterologous
...