Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
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
2.
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
3.
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
4.
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
5.
Magn Reson Med ; 71(3): 1221-30, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23640714

ABSTRACT

PURPOSE: CatalyCEST MRI compares the detection of an enzyme-responsive chemical exchange saturation transfer (CEST) agent with the detection of an unresponsive "control" CEST agent that accounts for other conditions that influence CEST. The purpose of this study was to investigate the feasibility of in vivo catalyCEST MRI. METHODS: CEST agents that were responsive and unresponsive to the activity of urokinase plasminogen activator were shown to have negligible interaction with each other. A CEST-fast imaging with steady state precession (FISP) MRI protocol was used to acquire MR CEST spectroscopic images with a Capan-2 pancreatic tumor model after intravenous injection of the CEST agents. A function of (super)-Lorentzian line shapes was fit to CEST spectra of a region-of-interest that represented the tumor. RESULTS: The CEST effects from each agent showed the same initial uptake into tumor tissues, indicating that both agents had the same pharmacokinetic transport rates. Starting 5 min after injection, CEST from the enzyme-responsive agent disappeared more quickly than CEST from the unresponsive agent, indicating that the enzyme responsive agent was being catalyzed by urokinase plasminogen activator, while both agents also experienced net pharmacokinetic washout from the tumor. CONCLUSION: CatalyCEST MRI demonstrates that dynamic tracking of enzyme-responsive and unresponsive CEST agents during the same in vivo MRI study is feasible.


Subject(s)
Heterocyclic Compounds, 1-Ring/pharmacokinetics , Magnetic Resonance Imaging/methods , Magnetic Resonance Spectroscopy/methods , Molecular Imaging/methods , Pancreatic Neoplasms/enzymology , Urokinase-Type Plasminogen Activator/metabolism , Animals , Catalysis , Cell Line, Tumor , Enzyme Activation , Feasibility Studies , Mice , Pancreatic Neoplasms/pathology , Radiopharmaceuticals/pharmacokinetics , Reproducibility of Results , Sensitivity and Specificity
6.
Pancreas ; 41(5): 773-81, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22273698

ABSTRACT

OBJECTIVES: We investigated the signaling pathways activated in response to interleukin 6 (IL-6) in pancreatic cell lines, with a focus on signal transducer and activator of transcription 3 (STAT3) and proto-oncogene serine/threonine-protein (Pim-1) kinase. METHODS: Interleukin 6 receptor (IL-6R) expression and IL-6-induced cell signaling was measured by Western blotting in human pancreatic cell lines. Cucurbitacin I was used as a pharmacological tool to investigate the role of STAT3 in Pim-1 activation. Stably overexpressing Pim-1 kinase cell lines were characterized for their response to IL-6 in vitro and for their growth rate as flank tumors in scid mice. RESULTS: Interleukin 6 receptor was expressed across multiple cancer cell lines. In Panc-1 cells, IL-6 treatment increased expression of phosphorylation of signal transducer and activator of transcription 3 and Pim-1 kinase. Cucurbitacin I treatment alone increased pErk1/2 expression in wild-type and Pim-1-overexpressing cell lines and resulted in exaggerated Pim-1 kinase protein levels in control and IL-6-stimulated cells, suggesting that up-regulation of Pim-1 may be partially STAT3 independent. Pim-1 overexpression did not significantly affect growth rate in vitro or in vivo in Panc-1 or MiaPaCa2 cell lines. CONCLUSIONS: Interleukin 6 activates STAT3 and stimulates Pim-1 kinase in pancreatic cell line models. The regulation and consequence of Pim-1 expression seems to be highly context dependent.


Subject(s)
Interleukin-6/pharmacology , Pancreatic Neoplasms/genetics , Proto-Oncogene Proteins c-pim-1/genetics , STAT3 Transcription Factor/genetics , Animals , Apoptosis/drug effects , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mice , Mice, SCID , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Proto-Oncogene Mas , Proto-Oncogene Proteins c-pim-1/metabolism , Receptors, Interleukin-6/genetics , Receptors, Interleukin-6/metabolism , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor/metabolism , Transplantation, Heterologous , Triterpenes/pharmacology , Tumor Burden/genetics
7.
Chem Biol ; 15(12): 1328-38, 2008 Dec 22.
Article in English | MEDLINE | ID: mdl-19101477

ABSTRACT

Fungal polyketides with the resorcylic acid lactone (RAL) scaffold are of interest for growth stimulation, the treatment of cancer, and neurodegenerative diseases. The RAL radicicol is a nanomolar inhibitor of the chaperone Hsp90, whose repression leads to a combinatorial blockade of cancer-causing pathways. Clustered genes for radicicol biosynthesis were identified and functionally characterized from the endophytic fungus Chaetomium chiversii, and compared to recently described RAL biosynthetic gene clusters. Radicicol production is abolished upon targeted inactivation of a putative cluster-specific regulator, or either of the two polyketide synthases that are predicted to collectively synthesize the radicicol polyketide core. Genomic evidence supports the existence of flavin-dependent halogenases in fungi: inactivation of such a putative halogenase from the C. chiversii radicicol locus yields dechloro-radicicol (monocillin I). Inactivation of a cytochrome P450 epoxidase furnishes pochonin D, a deepoxy-dihydro radicicol analog.


Subject(s)
Chaetomium/chemistry , Chaetomium/genetics , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Macrolides , Zearalenone/biosynthesis , Cloning, Molecular , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , HSP90 Heat-Shock Proteins/genetics , Lactones/chemistry , Lactones/metabolism , Macrolides/chemistry , Macrolides/metabolism , Macrolides/pharmacology , Models, Biological , Molecular Structure , Multigene Family , Zearalenone/chemistry , Zearalenone/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...