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.
BMC Cancer ; 16: 614, 2016 08 08.
Article in English | MEDLINE | ID: mdl-27501846

ABSTRACT

BACKGROUND: Circulating tumor cells (CTCs) have shown prognostic relevance in many cancer types. However, the majority of current CTC capture methods rely on positive selection techniques that require a priori knowledge about the surface protein expression of disseminated CTCs, which are known to be a dynamic population. METHODS: We developed a microfluidic CTC capture chip that incorporated a nanoroughened glass substrate for capturing CTCs from blood samples. Our CTC capture chip utilized the differential adhesion preference of cancer cells to nanoroughened etched glass surfaces as compared to normal blood cells and thus did not depend on the physical size or surface protein expression of CTCs. RESULTS: The microfluidic CTC capture chip was able to achieve a superior capture yield for both epithelial cell adhesion molecule positive (EpCAM+) and EpCAM- cancer cells in blood samples. Additionally, the microfluidic CTC chip captured CTCs undergoing transforming growth factor beta-induced epithelial-to-mesenchymal transition (TGF-ß-induced EMT) with dynamically down-regulated EpCAM expression. In a mouse model of human breast cancer using EpCAM positive and negative cell lines, the number of CTCs captured correlated positively with the size of the primary tumor and was independent of their EpCAM expression. Furthermore, in a syngeneic mouse model of lung cancer using cell lines with differential metastasis capability, CTCs were captured from all mice with detectable primary tumors independent of the cell lines' metastatic ability. CONCLUSIONS: The microfluidic CTC capture chip using a novel nanoroughened glass substrate is broadly applicable to capturing heterogeneous CTC populations of clinical interest independent of their surface marker expression and metastatic propensity. We were able to capture CTCs from a non-metastatic lung cancer model, demonstrating the potential of the chip to collect the entirety of CTC populations including subgroups of distinct biological and phenotypical properties. Further exploration of the biological potential of metastatic and presumably non-metastatic CTCs captured using the microfluidic chip will yield insights into their relevant differences and their effects on tumor progression and cancer outcomes.


Subject(s)
Cell Separation/methods , Epithelial Cell Adhesion Molecule/metabolism , Microfluidic Analytical Techniques/methods , Neoplasms/metabolism , Neoplastic Cells, Circulating/pathology , Transforming Growth Factor beta/pharmacology , A549 Cells , Animals , Cell Adhesion , Cell Line, Tumor , Epithelial-Mesenchymal Transition/drug effects , Female , Genetic Heterogeneity , Humans , MCF-7 Cells , Mice , Neoplasm Metastasis , Neoplasm Transplantation , Neoplasms/pathology , Neoplastic Cells, Circulating/drug effects , Neoplastic Cells, Circulating/metabolism
2.
Carcinogenesis ; 35(6): 1292-300, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24510113

ABSTRACT

In cancer cells, the process of epithelial-mesenchymal transition (EMT) confers migratory and invasive capacity, resistance to apoptosis, drug resistance, evasion of host immune surveillance and tumor stem cell traits. Cells undergoing EMT may represent tumor cells with metastatic potential. Characterizing the EMT secretome may identify biomarkers to monitor EMT in tumor progression and provide a prognostic signature to predict patient survival. Utilizing a transforming growth factor-ß-induced cell culture model of EMT, we quantitatively profiled differentially secreted proteins, by GeLC-tandem mass spectrometry. Integrating with the corresponding transcriptome, we derived an EMT-associated secretory phenotype (EASP) comprising of proteins that were differentially upregulated both at protein and mRNA levels. Four independent primary tumor-derived gene expression data sets of lung cancers were used for survival analysis by the random survival forests (RSF) method. Analysis of 97-gene EASP expression in human lung adenocarcinoma tumors revealed strong positive correlations with lymph node metastasis, advanced tumor stage and histological grade. RSF analysis built on a training set (n = 442), including age, sex and stage as variables, stratified three independent lung cancer data sets into low-, medium- and high-risk groups with significant differences in overall survival. We further refined EASP to a 20 gene signature (rEASP) based on variable importance scores from RSF analysis. Similar to EASP, rEASP predicted survival of both adenocarcinoma and squamous carcinoma patients. More importantly, it predicted survival in the early-stage cancers. These results demonstrate that integrative analysis of the critical biological process of EMT provides mechanism-based and clinically relevant biomarkers with significant prognostic value.


Subject(s)
Epithelial-Mesenchymal Transition , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Phenotype , Adult , Aged , Cell Line, Tumor , Cluster Analysis , Computational Biology , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression , Gene Expression Profiling , Humans , Lung Neoplasms/mortality , Lung Neoplasms/therapy , Male , Middle Aged , Neoplasm Grading , Neoplasm Staging , Prognosis , Proteomics
3.
J Thorac Oncol ; 6(11): 1784-92, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21964532

ABSTRACT

BACKGROUND: Acquisition of mesenchymal phenotype by epithelial cells by means of epithelial-mesenchymal transition (EMT) is considered as an early event in the multistep process of tumor metastasis. Therefore, inhibition of EMT might be a rational strategy to prevent metastasis. METHODS: Using the global gene expression profile from a cell culture model of transforming growth factor-ß (TGF-ß)-induced EMT, we identified potential EMT inhibitors. We used a publicly available database (www.broad.mit.edu/cmap) comprising gene expression profiles obtained from multiple different cell lines in response to various drugs to derive negative correlations to EMT gene expression profile using Connectivity Map, a pattern matching tool. RESULTS: Experimental validation of the identified compounds showed rapamycin as a novel inhibitor of TGF-ß signaling along with 17-AAG, a known modulator of TGF-ß pathway. Both of these compounds completely blocked EMT and the associated migratory and invasive phenotype. The other identified compound, LY294002, demonstrated a selective inhibition of mesenchymal markers, cell migration and invasion, without affecting the loss of E-cadherin expression or Smad phosphorylation. CONCLUSIONS: Our data reveal that rapamycin is a novel modulator of TGF-ß signaling, and along with 17-AAG and LY294002, could be used as therapeutic agent for inhibiting EMT. This study demonstrates the potential of a systems approach in identifying novel modulators of a complex biological process.


Subject(s)
Adenocarcinoma, Bronchiolo-Alveolar/pathology , Adenocarcinoma/pathology , Benzoquinones/pharmacology , Cell Movement/drug effects , Enzyme Inhibitors/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Lactams, Macrocyclic/pharmacology , Lung Neoplasms/pathology , Transforming Growth Factor beta/pharmacology , Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Adenocarcinoma, Bronchiolo-Alveolar/drug therapy , Adenocarcinoma, Bronchiolo-Alveolar/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Cadherins/metabolism , Chromones/pharmacology , Gene Expression Profiling , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Humans , Immunosuppressive Agents/pharmacology , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Morpholines/pharmacology , Oligonucleotide Array Sequence Analysis , Phosphoinositide-3 Kinase Inhibitors , Signal Transduction , Sirolimus/pharmacology , Smad Proteins/metabolism , Transcription, Genetic , Tumor Cells, Cultured
4.
Lung Cancer ; 72(2): 154-9, 2011 May.
Article in English | MEDLINE | ID: mdl-21354647

ABSTRACT

Peroxisome proliferator-activated receptors (PPAR)-γ belongs to the nuclear hormone receptor superfamily of ligand-dependent transcription factors. It is a mediator of adipocyte differentiation, regulates lipid metabolism and macrophage function. The ligands of PPAR-γ have long been in the clinic for the treatment of type II diabetes and have a very low toxicity profile. Activation of PPAR-γ was shown to modulate various hallmarks of cancer through its pleiotropic affects on multiple different cell types in the tumor microenvironment. An overwhelming number of preclinical-studies demonstrate the efficacy of PPAR-γ ligands in the control of tumor progression through their affects on various cellular processes, including cell proliferation, apoptosis, angiogenesis, inflammation and metastasis. A variety of signaling pathways have been implicated as potential mechanisms of action. This review will focus on the molecular basis of these mechanisms; primarily PPAR-γ cross-regulation with other signaling pathways and its relevance to lung cancer therapy will be discussed.


Subject(s)
Antineoplastic Agents/therapeutic use , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , PPAR gamma/agonists , Receptor Cross-Talk , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Drug Therapy/trends , Humans , Inflammation , Lung Neoplasms/pathology , Neoplasm Metastasis , Receptor Cross-Talk/drug effects , Signal Transduction/drug effects
5.
Mol Cancer Ther ; 9(12): 3221-32, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21159608

ABSTRACT

Epithelial-mesenchymal transition (EMT) was shown to confer tumor cells with abilities essential for metastasis, including migratory phenotype, invasiveness, resistance to apoptosis, evading immune surveillance, and tumor stem cell traits. Therefore, inhibition of EMT can be an important therapeutic strategy to inhibit tumor metastasis. Here, we show that activation of peroxisome proliferator-activated receptor γ (PPAR-γ) inhibits transforming growth factor ß (TGF-ß)-induced EMT in lung cancer cells and prevents metastasis by antagonizing Smad3 function. Activation of PPAR-γ by synthetic ligands (troglitazone and rosiglitazone) or by a constitutively active form of PPAR-γ prevents TGF-ß-induced loss of E-cadherin expression and inhibits the induction of mesenchymal markers (vimentin, N-cadherin, fibronectin) and matrix metalloproteases. Consistently, activation of PPAR-γ also inhibited EMT-induced migration and invasion of lung cancer cells. Furthermore, effects of PPAR-γ ligands were attenuated by siRNA-mediated knockdown of PPAR-γ, indicating that the ligand-induced responses are PPAR-γ dependent. Selective knockdown of Smad2 and Smad3 by siRNA showed that TGF-ß-induced EMT is Smad3 dependent in lung cancer cells. Activation of PPAR-γ inhibits TGF-ß-induced Smad transcriptional activity but had no effect on the phosphorylation or nuclear translocation of Smads. Consistently, PPAR-γ activation prevented TGF-ß-induced transcriptional repression of E-cadherin promoter and inhibited transcriptional activation of N-cadherin promoter. Finally, treatment of mice with troglitazone or knockdown of Smad3 in tumor cells significantly inhibited TGF-ß-induced experimental metastasis in SCID-Beige mice. Together, with the low toxicity profile of PPAR-γ ligands, our data show that these ligands may serve as potential therapeutic agents to inhibit metastasis.


Subject(s)
Epithelial-Mesenchymal Transition , PPAR gamma/metabolism , Smad3 Protein/metabolism , Animals , Cadherins/genetics , Cadherins/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Epithelial-Mesenchymal Transition/drug effects , Gene Knockdown Techniques , Humans , Ligands , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Neoplasm Metastasis , Phenotype , Phosphorylation/drug effects , Promoter Regions, Genetic/genetics , Protein Transport/drug effects , Smad2 Protein/metabolism , Smad3 Protein/genetics , Transcription, Genetic/drug effects , Transforming Growth Factor beta/pharmacology , Xenograft Model Antitumor Assays
6.
J Proteome Res ; 8(1): 35-47, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19118450

ABSTRACT

To gain insights into how TGF-beta regulates epithelial-mesenchymal transition (EMT), we assessed the time course of proteins and mRNAs during EMT by multiplex iTRAQ labeling and 2D-LC-MS/MS, and by hybridization, respectively. Temporal iTRAQ analysis identified 66 proteins as differentially expressed during EMT, including newly associated proteins calpain, fascin and macrophage-migration inhibitory factor (MIF). Comparing protein and mRNA expression overtime showed that all the 14 up-regulated proteins involved in the actin-cytoskeleton remodeling were accompanied by increases in corresponding mRNA expression. Interestingly, siRNA mediated knockdown of cofilin1 potentiated TGF-beta-induced EMT. Further analysis of cofilin1 and beta-actin revealed an increase in their mRNA stability in response to TGF-beta, contributing to the observed increase in mRNA and protein expression. These results are the first demonstration of post-transcriptional regulation of cytoskeletal remodelling and a key role for cofilin1 during TGF-beta-induced EMT.


Subject(s)
Actins/metabolism , Chromatography, Liquid/methods , Cytoskeleton/metabolism , Epithelium/drug effects , Mass Spectrometry/methods , Mesoderm/drug effects , Proteomics/methods , RNA Processing, Post-Transcriptional , RNA, Messenger/metabolism , Transforming Growth Factor beta/metabolism , Cell Differentiation/drug effects , Cell Line, Tumor , Cofilin 1/metabolism , Gene Expression Regulation , Humans , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL