Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Feb 09.
Article in English | MEDLINE | ID: mdl-37503155

ABSTRACT

Biallelic germline mutations in the SLC25A1 gene lead to combined D/L-2-hydroxyglutaric aciduria (D/L-2HGA), a fatal systemic disease uniquely characterized by the accumulation of both enantiomers of 2-hydroxyglutaric acid (2HG). How SLC25A1 deficiency contributes to D/L-2HGA and the role played by 2HG is unclear and no therapy exists. Both enantiomers act as oncometabolites, but their activities in normal tissues remain understudied. Here we show that mice lacking both SLC25A1 alleles exhibit developmental abnormalities that mirror human D/L-2HGA. SLC25A1 deficient cells undergo premature senescence, suggesting that loss of proliferative capacity underlies the pathogenesis of D/L-2HGA. Remarkably, D- and L-2HG directly induce senescence and treatment of zebrafish embryos with the combination of D- and L-2HG phenocopies SLC25A1 loss, leading to developmental abnormalities in an additive fashion relative to either enantiomer alone. Metabolic analyses further demonstrate that cells with dysfunctional SLC25A1 undergo mitochondrial respiratory deficit and remodeling of the metabolism and we propose several strategies to correct these defects. These results reveal for the first time pathogenic and growth suppressive activities of 2HG in the context of SLC25A1 deficiency and suggest that targeting the 2HG pathway may be beneficial for the treatment of D/L-2HGA.

2.
Mol Biomed ; 3(1): 16, 2022 May 26.
Article in English | MEDLINE | ID: mdl-35614362

ABSTRACT

Cancer metastasis is the primary cause of the high mortality rate among human cancers. Efforts to identify therapeutic agents targeting cancer metastasis frequently fail to demonstrate efficacy in clinical trials despite strong preclinical evidence. Until recently, most preclinical studies used mouse models to evaluate anti-metastatic agents. Mouse models are time-consuming and expensive. In addition, an important drawback is that mouse models inadequately model the early stages of metastasis which plausibly leads to the poor correlation with clinical outcomes.Here, we report an in vivo model based on xenografted zebrafish embryos where we select for progressively invasive subpopulations of MDA-MB-231 breast cancer cells. A subpopulation analogous to circulating tumor cells found in human cancers was selected by injection of MDA-MB-231 cells into the yolk sacs of 2 days post-fertilized zebrafish embryos and selecting cells that migrated to the tail. The selected subpopulation derived from MDA-MB-231 cells were increasingly invasive in zebrafish. Isolation of these subpopulations and propagation in vitro revealed morphological changes consistent with activation of an epithelial-mesenchymal transition program. Differential gene analysis and knockdown of genes identified gene-candidates (DDIT4, MT1X, CTSD, and SERPINE1) as potential targets for anti-metastasis therapeutics. Furthermore, RNA-splicing analysis reinforced the importance of BIRC5 splice variants in breast cancer metastasis. This is the first report using zebrafish to isolate and expand progressively invasive populations of human cancer cells. The model has potential applications in understanding the metastatic process, identification and/or development of therapeutics that specifically target metastatic cells and formulating personalized treatment strategies for individual cancer patients.

3.
Mol Cancer Ther ; 20(10): 1800-1808, 2021 10.
Article in English | MEDLINE | ID: mdl-34253593

ABSTRACT

Metastasis is the major cause of mortality in patients with breast cancer. Many signaling pathways have been linked to cancer invasiveness, but blockade of few protein components has succeeded in reducing metastasis. Thus, identification of proteins contributing to invasion that are manipulable by small molecules may be valuable in inhibiting spread of the disease. The protein kinase with no lysine (K) 1 (WNK1) has been suggested to induce migration of cells representing a range of cancer types. Analyses of mouse models and patient data have implicated WNK1 as one of a handful of genes uniquely linked to invasive breast cancer. Here, we present evidence that inhibition of WNK1 slows breast cancer metastasis. We show that depletion or inhibition of WNK1 reduces migration of several breast cancer cell lines in wound healing assays and decreases invasion in collagen matrices. Furthermore, WNK1 depletion suppresses expression of AXL, a tyrosine kinase implicated in metastasis. Finally, we demonstrate that WNK inhibition in mice attenuates tumor progression and metastatic burden. These data showing reduced migration, invasion, and metastasis upon WNK1 depletion in multiple breast cancer models suggest that WNK1 contributes to the metastatic phenotype, and that WNK1 inhibition may offer a therapeutic avenue for attenuating progression of invasive breast cancers.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/pathology , Gene Expression Regulation, Neoplastic , WNK Lysine-Deficient Protein Kinase 1/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Movement , Cell Proliferation , Female , Humans , Imidazoles/pharmacology , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Invasiveness , Pyrrolidines/pharmacology , Tumor Cells, Cultured , WNK Lysine-Deficient Protein Kinase 1/antagonists & inhibitors , WNK Lysine-Deficient Protein Kinase 1/genetics , Xenograft Model Antitumor Assays
4.
Cancer Res ; 81(16): 4230-4241, 2021 08 15.
Article in English | MEDLINE | ID: mdl-34135000

ABSTRACT

AIB1Δ4 is an N-terminally truncated isoform of the oncogene amplified in breast cancer 1 (AIB1) with increased expression in high-grade human ductal carcinoma in situ (DCIS). However, the role of AIB1Δ4 in DCIS malignant progression has not been defined. Here we CRISPR-engineered RNA splice junctions to produce normal and early-stage DCIS breast epithelial cells that expressed only AIB1Δ4. These cells showed enhanced motility and invasion in 3D cell culture. In zebrafish, AIB1Δ4-expressing cells enabled invasion of parental cells when present in a mixed population. In mouse xenografts, a subpopulation of AIB1Δ4 cells mixed with parental cells enhanced tumor growth, recurrence, and lung metastasis. AIB1Δ4 chromatin immunoprecipitation sequencing revealed enhanced binding to regions including peroxisome proliferator-activated receptor (PPAR) and glucocorticoid receptor (GR) genomic recognition sites. H3K27ac and H3K4me1 genomic engagement patterns revealed selective activation of breast cancer-specific enhancer sites by AIB1Δ4. AIB1Δ4 cells displayed upregulated inflammatory response genes and downregulated PPAR signaling gene expression patterns. In the presence of AIB1Δ4 enabler cells, parental cells increased NF-κB and WNT signaling. Cellular cross-talk was inhibited by the PPARγ agonist efatutazone but was enhanced by treatment with the GR agonist dexamethasone. In conclusion, expression of the AIB1Δ4-selective cistrome in a small subpopulation of cells triggers an "enabler" phenotype hallmarked by an invasive transcriptional program and collective malignant progression in a heterogeneous tumor population. SIGNIFICANCE: A minor subset of early-stage breast cancer cells expressing AIB1Δ4 enables bulk tumor cells to become invasive, suggesting that selective eradication of this population could impair breast cancer metastasis.


Subject(s)
Nuclear Receptor Coactivator 3/genetics , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Animals , CRISPR-Cas Systems , Cell Culture Techniques, Three Dimensional , Cell Line, Tumor , Dexamethasone/chemistry , Disease Progression , Electric Impedance , Enhancer Elements, Genetic , Female , Humans , Lung Neoplasms/pathology , Mice , Mice, SCID , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Transplantation , Nuclear Receptor Coactivator 3/chemistry , Phenotype , Protein Isoforms , RNA Splicing , Receptors, Glucocorticoid/metabolism , Signal Transduction , Thiazolidinediones/pharmacology , Zebrafish
5.
Int J Mol Sci ; 22(1)2020 Dec 24.
Article in English | MEDLINE | ID: mdl-33374314

ABSTRACT

Using a data driven analysis of a high-content screen, we have uncovered new regulators of epithelial-to-mesenchymal transition (EMT) induced cell migration. Our results suggest that increased expression of miR614 can alter cell intrinsic gene expression to enhance single cell and collective migration in multiple contexts. Interestingly, miR614 specifically increased the expression of the EMT transcription factor Slug while not altering existing epithelial character or inducing other canonical EMT regulatory factors. Analysis of two different cell lines identified a set of genes whose expression is altered by the miR614 through direct and indirect mechanisms. Prioritization driven by functional testing of 25 of the miR614 suppressed genes uncovered the mitochondrial small GTPase Miro1 and the transmembrane protein TAPT1 as miR614 suppressed genes that inhibit migration. Notably, the suppression of either Miro1 or TAPT1 was sufficient to increase Slug expression and the rate of cell migration. Importantly, reduced TAPT1 expression correlated with an increased risk of relapse in breast cancer patients. Together, our results reveal how increased miR614 expression and the suppression of TAPT1 and Miro1 modulate the EMT state and migratory properties of breast cancer cells.


Subject(s)
Breast Neoplasms/metabolism , Cell Movement , Gene Expression Regulation, Neoplastic , MicroRNAs/biosynthesis , RNA, Neoplasm/biosynthesis , Breast Neoplasms/genetics , Cell Line, Tumor , Epithelial-Mesenchymal Transition/genetics , Female , Humans , MicroRNAs/genetics , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , RNA, Neoplasm/genetics
6.
Cancer Res ; 80(18): 3933-3944, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32661136

ABSTRACT

Defining how interactions between tumor subpopulations contribute to invasion is essential for understanding how tumors metastasize. Here, we find that the heterogeneous expression of the transcription factor ΔNp63 confers distinct proliferative and invasive epithelial-to-mesenchymal transition (EMT) states in subpopulations that establish a leader-follower relationship to collectively invade. A ΔNp63-high EMT program coupled the ability to proliferate with an IL1α- and miR-205-dependent suppression of cellular protrusions that are required to initiate collective invasion. An alternative ΔNp63-low EMT program conferred cells with the ability to initiate and lead collective invasion. However, this ΔNp63-low EMT state triggered a collateral loss of fitness. Importantly, rare growth-suppressed ΔNp63-low EMT cells influenced tumor progression by leading the invasion of proliferative ΔNp63-high EMT cells in heterogeneous primary tumors. Thus, heterogeneous activation of distinct EMT programs promotes a mode of collective invasion that overcomes cell intrinsic phenotypic deficiencies to induce the dissemination of proliferative tumor cells. SIGNIFICANCE: These findings reveal how an interaction between cells in different EMT states confers properties that are not induced by either EMT program alone.


Subject(s)
Epithelial-Mesenchymal Transition/physiology , Neoplasm Invasiveness/physiopathology , Neoplasm Proteins/metabolism , Transcription Factors/metabolism , Triple Negative Breast Neoplasms/pathology , Tumor Suppressor Proteins/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Cell Culture Techniques , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell Surface Extensions , Disease Progression , Epithelial-Mesenchymal Transition/genetics , Extracellular Matrix/pathology , Female , Humans , Interleukin-1alpha/genetics , Interleukin-1alpha/metabolism , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasm Proteins/deficiency , Neoplasm Proteins/genetics , RNA, Small Interfering/metabolism , Spheroids, Cellular , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/deficiency , Transcription Factors/genetics , Triple Negative Breast Neoplasms/genetics , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics
7.
J Vis Exp ; (160)2020 06 23.
Article in English | MEDLINE | ID: mdl-32658183

ABSTRACT

Significant progress has been made in treating cancer with immunotherapy, although a large number of cancers remain resistant to treatment. A limited number of assays allow for direct monitoring and mechanistic insights into the interactions between tumor and immune cells, amongst which, T-cells play a significant role in executing the cytotoxic response of the adaptive immune system to cancer cells. Most assays are based on two-dimensional (2D) co-culture of cells due to the relative ease of use but with limited representation of the invasive growth phenotype, one of the hallmarks of cancer cells. Current three-dimensional (3D) co-culture systems either require special equipment or separate monitoring for invasion of co-cultured cancer cells and interacting T-cells. Here we describe an approach to simultaneously monitor the invasive behavior in 3D of cancer cell spheroids and T-cell cytotoxicity in co-culture. Spheroid formation is driven by enhanced cell-cell interactions in scaffold-free agarose microwell casts with U-shaped bottoms. Both T-cell co-culture and cancer cell invasion into type I collagen matrix are performed within the microwells of the agarose casts without the need to transfer the cells, thus maintaining an intact 3D co-culture system throughout the assay. The collagen matrix can be separated from the agarose cast, allowing for immunofluorescence (IF) staining and for confocal imaging of cells. Also, cells can be isolated for further growth or subjected to analyses such as for gene expression or fluorescence activated cell sorting (FACS). Finally, the 3D co-culture can be analyzed by immunohistochemistry (IHC) after embedding and sectioning. Possible modifications of the assay include altered compositions of the extracellular matrix (ECM) as well as the inclusion of different stromal or immune cells with the cancer cells.


Subject(s)
Coculture Techniques/methods , T-Lymphocytes, Cytotoxic/cytology , Cell Communication , Cell Line, Tumor , Collagen Type I/metabolism , Extracellular Matrix/metabolism , Humans , Neoplasm Invasiveness , Spheroids, Cellular/pathology
8.
J Clin Med ; 8(5)2019 May 10.
Article in English | MEDLINE | ID: mdl-31083398

ABSTRACT

Epithelial-to-mesenchymal transition (EMT) programs contribute to the acquisition of invasive properties that are essential for metastasis. It is well established that EMT programs alter cell state and promote invasive behavior. This review discusses how rather than following one specific program, EMT states are diverse in their regulation and invasive properties. Analysis across a spectrum of models using a combination of approaches has revealed how unique features of distinct EMT programs dictate whether tumor cells invade as single cells or collectively as cohesive groups of cells. It has also been shown that the mode of collective invasion is determined by the nature of the EMT, with cells in a trailblazer-type EMT state being capable of initiating collective invasion, whereas cells that have undergone an opportunist-type EMT are dependent on extrinsic factors to invade. In addition to altering cell intrinsic properties, EMT programs can influence invasion through non-cell autonomous mechanisms. Analysis of tumor subpopulations has demonstrated how EMT-induced cells can drive the invasion of sibling epithelial populations through paracrine signaling and remodeling of the microenvironment. Importantly, the variation in invasive properties controlled by EMT programs influences the kinetics and location of metastasis.

9.
Oncotarget ; 7(19): 28592-611, 2016 May 10.
Article in English | MEDLINE | ID: mdl-27081041

ABSTRACT

Tumor invasion can be induced by changes in gene expression that alter cell phenotype. The transcription factor ΔNp63α promotes basal-like breast cancer (BLBC) migration by inducing the expression of the mesenchymal genes Slug and Axl, which confers cells with a hybrid epithelial/mesenchymal state. However, the extent of the ΔNp63α regulated genes that support invasive behavior is not known. Here, using gene expression analysis, ChIP-seq, and functional testing, we find that ΔNp63α promotes BLBC motility by inducing the expression of the atypical cadherin FAT2, the vesicular binding protein SNCA, the carbonic anhydrase CA12, the lipid binding protein CPNE8 and the kinase NEK1, along with Slug and Axl. Notably, lung squamous cell carcinoma migration also required ΔNp63α dependent FAT2 and Slug expression, demonstrating that ΔNp63α promotes migration in multiple tumor types by inducing mesenchymal and non-mesenchymal genes. ΔNp63α activation of FAT2 and Slug influenced E-cadherin localization to cell-cell contacts, which can restrict spontaneous cell movement. Moreover, live-imaging of spheroids in organotypic culture demonstrated that ΔNp63α, FAT2 and Slug were essential for the extension of cellular protrusions that initiate collective invasion. Importantly, ΔNp63α is co-expressed with FAT2 and Slug in patient tumors and the elevated expression of ΔNp63α, FAT2 and Slug correlated with poor patient outcome. Together, these results reveal how ΔNp63α promotes cell migration by directly inducing the expression of a cohort of genes with distinct cellular functions and suggest that FAT2 is a new regulator of collective invasion that may influence patient outcome.


Subject(s)
Cadherins/genetics , Cell Movement/genetics , Gene Expression Regulation, Neoplastic , Neoplasms/genetics , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cadherins/metabolism , Cell Line, Tumor , Female , Gene Expression Profiling/methods , Humans , Kaplan-Meier Estimate , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Neoplasm Invasiveness , Neoplasms/metabolism , Neoplasms/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Snail Family Transcription Factors/genetics , Snail Family Transcription Factors/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology
10.
Oncotarget ; 7(12): 14708-26, 2016 Mar 22.
Article in English | MEDLINE | ID: mdl-26895102

ABSTRACT

Genes that are normally biased towards expression in the testis are often induced in tumor cells. These gametogenic genes, known as cancer-testis antigens (CTAs), have been extenstively investigated as targets for immunotherapy. However, despite their frequent detection, the degree to which CTAs support neoplastic invasion is poorly understood. Here, we find that the CTA genes SPANX-A/C/D and CTAG2 are coordinately induced in breast cancer cells and regulate distinct features of invasive behavior. Our functional analysis revealed that CTAG2 interacts with Pericentrin at the centrosome and is necessary for directional migration. Conversely, SPANX-A/C/D interacts with Lamin A/C at the inner nuclear membrane and is required for the formation of actin-rich cellular protrusions that reorganize the extracellular matrix. Importantly, SPANX-A/C/D was required for breast cancer cells to spontaneously metastasize to the lung, demonstrating that CTA reactivation can be critical for invasion dependent phenotypes in vivo. Moreover, elevated SPANX-A/C/D expression in breast cancer patient tumors correlated with poor outcome. Together, our results suggest that distinct CTAs promote tumor progression by regulating complementary cellular functions that are integrated together to induce invasive behavior.


Subject(s)
Antigens, Neoplasm/metabolism , Antigens, Surface/metabolism , Biomarkers, Tumor/metabolism , Breast Neoplasms/pathology , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , Animals , Apoptosis , Breast Neoplasms/metabolism , Cell Movement , Cell Proliferation , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Invasiveness , Neoplasm Staging , Prognosis , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
11.
Cancer Res ; 75(18): 3925-35, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26292362

ABSTRACT

Cell identity signals influence the invasive capability of tumor cells, as demonstrated by the selection for programs of epithelial-to-mesenchymal transition (EMT) during malignant progression. Breast cancer cells retain canonical epithelial traits and invade collectively as cohesive groups of cells, but the signaling pathways critical to their invasive capabilities are still incompletely understood. Here we report that the transcription factor ΔNp63α drives the migration of basal-like breast cancer (BLBC) cells by inducing a hybrid mesenchymal/epithelial state. Through a combination of expression analysis and functional testing across multiple BLBC cell populations, we determined that ΔNp63α induces migration by elevating the expression of the EMT program components Slug and Axl. Interestingly, ΔNp63α also increased the expression of miR-205, which can silence ZEB1/2 to prevent the loss of epithelial character caused by EMT induction. In clinical specimens, co-expression of various elements of the ΔNp63α pathway confirmed its implication in motility signaling in BLBC. We observed that activation of the ΔNp63α pathway occurred during the transition from noninvasive ductal carcinoma in situ to invasive breast cancer. Notably, in an orthotopic tumor model, Slug expression was sufficient to induce collective invasion of E-cadherin-expressing BLBC cells. Together, our results illustrate how ΔNp63α can drive breast cancer cell invasion by selectively engaging promigratory components of the EMT program while, in parallel, still promoting the retention of epithelial character.


Subject(s)
Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/pathology , Carcinoma, Intraductal, Noninfiltrating/pathology , Cell Transformation, Neoplastic/pathology , Epithelial-Mesenchymal Transition/physiology , Gene Expression Regulation, Neoplastic , Neoplasm Invasiveness/physiopathology , Neoplasm Proteins/physiology , Transcription Factors/physiology , Tumor Suppressor Proteins/physiology , Animals , Breast Neoplasms/mortality , Cadherins/biosynthesis , Cadherins/genetics , Cell Movement , Disease Progression , Epithelial Cells/pathology , Female , Heterografts , Humans , Kaplan-Meier Estimate , Mice , Mice, Inbred NOD , Mice, SCID , MicroRNAs/biosynthesis , MicroRNAs/genetics , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , RNA, Small Interfering/pharmacology , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptor Protein-Tyrosine Kinases/genetics , Signal Transduction/physiology , Snail Family Transcription Factors , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transfection , Tumor Cells, Cultured , Axl Receptor Tyrosine Kinase
12.
J Clin Invest ; 125(5): 1927-43, 2015 May.
Article in English | MEDLINE | ID: mdl-25844900

ABSTRACT

Tumor cells can engage in a process called collective invasion, in which cohesive groups of cells invade through interstitial tissue. Here, we identified an epigenetically distinct subpopulation of breast tumor cells that have an enhanced capacity to collectively invade. Analysis of spheroid invasion in an organotypic culture system revealed that these "trailblazer" cells are capable of initiating collective invasion and promote non-trailblazer cell invasion, indicating a commensal relationship among subpopulations within heterogenous tumors. Canonical mesenchymal markers were not sufficient to distinguish trailblazer cells from non-trailblazer cells, suggesting that defining the molecular underpinnings of the trailblazer phenotype could reveal collective invasion-specific mechanisms. Functional analysis determined that DOCK10, ITGA11, DAB2, PDFGRA, VASN, PPAP2B, and LPAR1 are highly expressed in trailblazer cells and required to initiate collective invasion, with DOCK10 essential for metastasis. In patients with triple-negative breast cancer, expression of these 7 genes correlated with poor outcome. Together, our results indicate that spontaneous conversion of the epigenetic state in a subpopulation of cells can promote a transition from in situ to invasive growth through induction of a cooperative form of collective invasion and suggest that therapeutic inhibition of trailblazer cell invasion may help prevent metastasis.


Subject(s)
Breast Neoplasms/pathology , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Genes, Neoplasm , Neoplasm Invasiveness/genetics , Neoplasm Metastasis/genetics , Neoplasm Proteins/physiology , Animals , Breast Neoplasms/mortality , Cell Line, Tumor , Cell Movement , Cell Surface Extensions/physiology , Cell Surface Extensions/ultrastructure , Epigenesis, Genetic/genetics , Epithelial-Mesenchymal Transition , Extracellular Matrix , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Invasiveness/pathology , Neoplasm Proteins/genetics , RNA Interference , Specific Pathogen-Free Organisms , Spheroids, Cellular , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/mortality , Triple Negative Breast Neoplasms/pathology
13.
Cancer Res ; 71(21): 6857-66, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21908556

ABSTRACT

Most ductal breast carcinoma cells are weakly invasive in vitro and in vivo, suggesting that components of their microenvironment may facilitate a transition from in situ to invasive stages during progression. Here, we report that coculture of mammary fibroblasts specifically triggers invasive behavior in basal-type breast cancer cells through a ligand independent mechanism. When cultured alone in organotypic culture, both basal- and luminal-type breast cancer cells formed noninvasive spheroids with characteristics of ductal carcinoma in situ (DCIS). In contrast, when cocultured with mammary fibroblasts, basal-type spheroids exhibited invasive character whereas the luminal-type spheroids retained a benign and noninvasive duct-like architecture. Real-time imaging and functional studies revealed that the specificity of invasion was linked to a unique capacity of basal-type breast cancer cells to move within spheroids. Mammary fibroblasts induced invasion by triggering basal-type breast cancer cells to convert from a noninvasive program of mammary epithelial morphogenesis to an invasive program of sprouting endothelial angiogenesis. Contrary to the existing invasion models, soluble ligands produced by the fibroblasts were not sufficient to trigger invasion. Instead, basal-type invasion relied upon a Cdc42-dependent reorganization of collagen fibers in the extracellular matrix by fibroblasts. Inhibiting basal-type cell movement with clinically relevant drugs blocked invasion both in organotypic culture and in animals, suggesting a new treatment strategy for early-stage patients. Together our findings establish that fibroblast recruitment by basal-type breast cancer cells into early-stage tumors is sufficient to trigger their conversion from a benign, noninvasive DCIS-like stage to a malignant invasive stage. Furthermore, our findings suggest that different subtypes of breast cancer may require distinct types of contributions from the microenvironment to undergo malignant progression.


Subject(s)
Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/pathology , Carcinoma, Intraductal, Noninfiltrating/pathology , Neoplasm Invasiveness/physiopathology , Tumor Microenvironment/physiology , Animals , Breast/cytology , Breast Neoplasms/classification , Carcinoma, Ductal, Breast/classification , Cell Movement/drug effects , Cell Movement/physiology , Cells, Cultured/cytology , Cells, Cultured/physiology , Coculture Techniques , Collagen/physiology , Culture Media, Conditioned/pharmacology , Extracellular Matrix/physiology , Extracellular Matrix/ultrastructure , Female , Fibroblasts/cytology , Fibroblasts/physiology , Humans , Mice , Mice, SCID , Neoplasm Invasiveness/prevention & control , Neoplasm Transplantation , Neovascularization, Pathologic/pathology , Protein Kinase Inhibitors/pharmacology , RNA, Small Interfering/pharmacology , Spheroids, Cellular/physiology , Spheroids, Cellular/transplantation , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/physiology , cdc42 GTP-Binding Protein/antagonists & inhibitors , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/physiology
14.
Breast Cancer Res ; 11(3): R29, 2009.
Article in English | MEDLINE | ID: mdl-19457236

ABSTRACT

INTRODUCTION: Epithelial tumors, including breast cancer, are being identified and treated at earlier stages of tumor development because of technological advances in screening and detection methods. It is likely that early-stage epithelial tumors, such as mammary ductal carcinoma in situ (DCIS), will be amenable to new and more efficacious diagnostic tests and forms of therapy. However, our limited understanding of the underlying molecular mechanisms of early-stage epithelial tumor growth has hampered the development of new forms treatment and preventative therapy. METHODS: The Raf-MEK1/2-ERK1/2 mitogen-activated protein kinase module is activated by stimuli complicit in mammary neoplastic progression. We have recently demonstrated that the activation of ERK1/2 induces a non-invasive form of motility, where cells can track along the basement membrane and adjacent epithelial cells, but do not become invasive over time, using real-time imaging of a mammary epithelial organotypic culture model. Using this novel approach combined with traditional biochemical techniques, we have analyzed at the molecular level how ERK1/2 induces this new non-invasive form of motility as well as proliferation and cell survival. RESULTS: We find that the activation of Raf:ER in the differentiated epithelium of fully formed acini promotes proliferation and cell survival, which are characteristic features of pre-invasive DCIS lesions. The activation of ERK1/2 correlated with induction of c-Fos, a transcriptional regulator of proliferation and reduced expression of the pro-apoptotic BH3-only protein BIM. Both ERK1/2 and PI-3 kinase-dependent effector pathways were required for activated Raf:ER to reduce expression of p27 and promote proliferation. In addition, PI-3K activity was necessary for the induction of non-invasive motility induced by ERK1/2. CONCLUSIONS: ERK1/2 activation is sufficient to induce cell behaviors in organotypic culture that could promote recurrent and invasive growth in DCIS patients. Interestingly, PI-3K activity is necessary for two of these behaviors, proliferation and cell motility. Collectively, our results suggest that the relationship between the activity state of the ERK1/2 and PI-3K signaling pathways and recurrent growth in DCIS patients should be investigated.


Subject(s)
Breast/metabolism , Epithelium/metabolism , Gene Expression Regulation, Enzymologic , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Apoptosis , Cell Line , Cell Movement , Cell Proliferation , Cell Survival , Disease Progression , Epithelium/pathology , Humans , MAP Kinase Signaling System , Microscopy, Fluorescence/methods
15.
J Cell Biol ; 179(7): 1555-67, 2007 Dec 31.
Article in English | MEDLINE | ID: mdl-18166657

ABSTRACT

To determine how extracellular signal-regulated kinases (ERK) 1/2 promote mammary tumorigenesis, we examined the real-time behavior of cells in an organotypic culture of the mammary glandular epithelium. Inducible activation of ERK1/2 in mature acini elicits cell motility and disrupts epithelial architecture in a manner that is reminiscent of ductal carcinoma in situ; however, motile cells do not invade through the basement membrane and branching morphogenesis does not take place. ERK1/2-induced motility causes cells to move both within the cell monolayer that contacts the basement membrane surrounding the acinus and through the luminal space of the acinus. E-cadherin expression is reduced after ERK1/2 activation, but motility does not involve an epithelial-mesenchymal transition. Cell motility and the disruption of epithelial architecture require a Rho kinase- and myosin light chain kinase-dependent increase in the phosphorylation of myosin light chain 2. Our results identify a new mechanism for the disruption of architecture in epithelial acini and suggest that ERK1/2 can promote noninvasive motility in preinvasive mammary tumors.


Subject(s)
Breast Neoplasms/physiopathology , Breast/physiopathology , Carcinoma/physiopathology , Cell Movement , Epithelial Cells/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Breast/metabolism , Breast/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cadherins/metabolism , Carcinoma/metabolism , Carcinoma/pathology , Cardiac Myosins/metabolism , Cell Adhesion , Cell Line , Cell Line, Tumor , Enzyme Activation/drug effects , Epithelial Cells/pathology , Female , Humans , Mitogen-Activated Protein Kinase 3/drug effects , Myosin Light Chains/metabolism , Myosin-Light-Chain Kinase/metabolism , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/physiopathology , Organ Culture Techniques , Phosphorylation , rho-Associated Kinases/metabolism
16.
Arch Biochem Biophys ; 449(1-2): 8-16, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16626623

ABSTRACT

We showed previously that p90 RSK was activated in cells expressing an activated mutant of MEK5, the activator of the MAP kinase ERK5. Based on the following evidence, we suggest that ERK5 can directly activate RSK in cells. ERK5 binds to RSK in vitro and co-immunoprecipitates from cell extracts; activation of ERK5 weakens its binding to RSK, suggesting that RSK is released upon activation. Phosphorylation of RSK by ERK5 in vitro causes its activation, indicating that RSK is a substrate of ERK5. In cells activation of ERK5 but not p38 or the c-Jun N-terminal kinase is associated with RSK activation. The large C-terminal domain of ERK5 is not required for binding or activation of RSK by ERK5; however, the common docking or CD domain of ERK5 and the docking or D domain of RSK are important for their association.


Subject(s)
Mitogen-Activated Protein Kinase 7/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Animals , Binding Sites , COS Cells , Chlorocebus aethiops , Extracellular Signal-Regulated MAP Kinases/metabolism , HeLa Cells , Humans , Phosphorylation , Protein Binding
17.
Mol Cell Biol ; 26(8): 3039-47, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16581779

ABSTRACT

Cells integrate signals to select the appropriate response from an array of possible outcomes. Signal integration causes the reorganization of signaling pathways by undescribed events. To analyze the molecular changes in signaling pathways that elicit different responses, we focused on the interaction between cyclic AMP (cAMP) and growth factors. We show that the activation of extracellular signal-regulated kinase 5 (ERK5), but not ERK1/2, by growth factors is disrupted by cAMP through cAMP-dependent protein kinase (PKA). Activation of MEKK2, a mitogen-activated protein (MAP) kinase kinase kinase upstream of ERK5 that is required for growth factor activation of ERK5, is also disrupted by PKA. Transcription of c-Jun is induced by ERK5, and like ERK5, c-Jun induction is also blocked by cAMP. Transcription from the serum response element, like activation of ERK1/2, is not blocked by cAMP. Collectively, these results support a model in which cAMP shapes the growth factor-induced cellular response through PKA-dependent uncoupling of selected MAP kinase cascades from activating signals.


Subject(s)
Cyclic AMP/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Signal Transduction/drug effects , 1-Methyl-3-isobutylxanthine/pharmacology , Animals , Cell Line , Colforsin/pharmacology , Cyclic AMP/metabolism , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Epidermal Growth Factor/pharmacology , Fibroblast Growth Factors/pharmacology , Gene Expression Regulation, Enzymologic , HeLa Cells , Humans , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinase 7/metabolism , Mitogen-Activated Protein Kinases/genetics , NIH 3T3 Cells , Proto-Oncogene Proteins c-jun/metabolism , Response Elements/genetics , Transcription, Genetic
18.
J Biol Chem ; 277(50): 48094-8, 2002 Dec 13.
Article in English | MEDLINE | ID: mdl-12297510

ABSTRACT

ERK5 activity is increased by agents known to activate receptor tyrosine kinases, G-protein coupled receptors, and stress response pathways. We now find a role for cAMP in the regulation of ERK5. ERK5 is activated by forskolin, isoproterenol, and epinephrine in NIH3T3 cells and C2C12 myoblasts. ERK1/2 are also activated by cAMP in NIH3T3 cells, but not in C2C12 myoblasts, demonstrating differential regulation of ERK5 and ERK1/2 by cAMP. We examined the effect of cell context on activation of ERK5 and discovered ERK5 activity is inhibited, rather than activated, by cAMP in confluent, serum-deprived NIH3T3 cells and C2C12 myoblasts. Our results suggest that regulation of MAP kinase pathways by cAMP is not only dictated by cell type, but also by cell context.


Subject(s)
Cyclic AMP/physiology , Mitogen-Activated Protein Kinases/metabolism , 3T3 Cells , Animals , Colforsin/pharmacology , Enzyme Activation , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinase 7 , Mitogen-Activated Protein Kinases/antagonists & inhibitors , PC12 Cells , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...