Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters











Database
Language
Publication year range
1.
Cancer Res ; 70(6): 2296-306, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20215510

ABSTRACT

Epithelial-mesenchymal transition (EMT), a switch of polarized epithelial cells to a migratory, fibroblastoid phenotype, is considered a key process driving tumor cell invasiveness and metastasis. Using breast cancer cell lines as a model system, we sought to discover gene expression signatures of EMT with clinical and mechanistic relevance. A supervised comparison of epithelial and mesenchymal breast cancer lines defined a 200-gene EMT signature that was prognostic across multiple breast cancer cohorts. The immunostaining of LYN, a top-ranked EMT signature gene and Src-family tyrosine kinase, was associated with significantly shorter overall survival (P = 0.02) and correlated with the basal-like ("triple-negative") phenotype. In mesenchymal breast cancer lines, RNAi-mediated knockdown of LYN inhibited cell migration and invasion, but not proliferation. Dasatinib, a dual-specificity tyrosine kinase inhibitor, also blocked invasion (but not proliferation) at nanomolar concentrations that inhibit LYN kinase activity, suggesting that LYN is a likely target and that invasion is a relevant end point for dasatinib therapy. Our findings define a prognostically relevant EMT signature in breast cancer and identify LYN as a mediator of invasion and a possible new therapeutic target (and theranostic marker for dasatinib response), with particular relevance to clinically aggressive basal-like breast cancer.


Subject(s)
Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Thiazoles/pharmacology , src-Family Kinases/genetics , Breast Neoplasms/genetics , Cell Line, Tumor , Dasatinib , Epithelial Cells/pathology , Female , Gene Expression Profiling , Humans , Mesoderm/pathology , Prognosis , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/biosynthesis
2.
PLoS One ; 4(7): e6146, 2009 Jul 03.
Article in English | MEDLINE | ID: mdl-19582160

ABSTRACT

BACKGROUND: Breast cancer cell lines have been used widely to investigate breast cancer pathobiology and new therapies. Breast cancer is a molecularly heterogeneous disease, and it is important to understand how well and which cell lines best model that diversity. In particular, microarray studies have identified molecular subtypes-luminal A, luminal B, ERBB2-associated, basal-like and normal-like-with characteristic gene-expression patterns and underlying DNA copy number alterations (CNAs). Here, we studied a collection of breast cancer cell lines to catalog molecular profiles and to assess their relation to breast cancer subtypes. METHODS: Whole-genome DNA microarrays were used to profile gene expression and CNAs in a collection of 52 widely-used breast cancer cell lines, and comparisons were made to existing profiles of primary breast tumors. Hierarchical clustering was used to identify gene-expression subtypes, and Gene Set Enrichment Analysis (GSEA) to discover biological features of those subtypes. Genomic and transcriptional profiles were integrated to discover within high-amplitude CNAs candidate cancer genes with coordinately altered gene copy number and expression. FINDINGS: Transcriptional profiling of breast cancer cell lines identified one luminal and two basal-like (A and B) subtypes. Luminal lines displayed an estrogen receptor (ER) signature and resembled luminal-A/B tumors, basal-A lines were associated with ETS-pathway and BRCA1 signatures and resembled basal-like tumors, and basal-B lines displayed mesenchymal and stem/progenitor-cell characteristics. Compared to tumors, cell lines exhibited similar patterns of CNA, but an overall higher complexity of CNA (genetically simple luminal-A tumors were not represented), and only partial conservation of subtype-specific CNAs. We identified 80 high-level DNA amplifications and 13 multi-copy deletions, and the resident genes with concomitantly altered gene-expression, highlighting known and novel candidate breast cancer genes. CONCLUSIONS: Overall, breast cancer cell lines were genetically more complex than tumors, but retained expression patterns with relevance to the luminal-basal subtype distinction. The compendium of molecular profiles defines cell lines suitable for investigations of subtype-specific pathobiology, cancer stem cell biology, biomarkers and therapies, and provides a resource for discovery of new breast cancer genes.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Profiling , Base Sequence , Blotting, Western , Breast Neoplasms/pathology , Cell Line, Tumor , DNA Primers , Female , Genes, BRCA1 , Humans , Immunohistochemistry , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , RNA, Messenger/genetics , Transcription, Genetic
3.
Mol Oncol ; 2(4): 327-39, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19383354

ABSTRACT

Breast cancer exhibits clinical and molecular heterogeneity, where expression profiling studies have identified five major molecular subtypes. The basal-like subtype, expressing basal epithelial markers and negative for estrogen receptor (ER), progesterone receptor (PR) and HER2, is associated with higher overall levels of DNA copy number alteration (CNA), specific CNAs (like gain on chromosome 10p), and poor prognosis. Discovering the molecular genetic basis of tumor subtypes may provide new opportunities for therapy. To identify the driver oncogene on 10p associated with basal-like tumors, we analyzed genomic profiles of 172 breast carcinomas. The smallest shared region of gain spanned just seven genes at 10p13, including calcium/calmodulin-dependent protein kinase ID (CAMK1D), functioning in intracellular signaling but not previously linked to cancer. By microarray, CAMK1D was overexpressed when amplified, and by immunohistochemistry exhibited elevated expression in invasive carcinomas compared to carcinoma in situ. Engineered overexpression of CAMK1D in non-tumorigenic breast epithelial cells led to increased cell proliferation, and molecular and phenotypic alterations indicative of epithelial-mesenchymal transition (EMT), including loss of cell-cell adhesions and increased cell migration and invasion. Our findings identify CAMK1D as a novel amplified oncogene linked to EMT in breast cancer, and as a potential therapeutic target with particular relevance to clinically unfavorable basal-like tumors.


Subject(s)
Breast Neoplasms/pathology , Calcium-Calmodulin-Dependent Protein Kinase Type 1/genetics , Epithelial Cells/pathology , Gene Amplification , Mesenchymal Stem Cells/pathology , Calcium-Calmodulin-Dependent Protein Kinase Type 1/analysis , Cell Adhesion/genetics , Cell Proliferation , Chromosomes, Human, Pair 10/genetics , Female , Humans , Immunohistochemistry , Mesenchymal Stem Cells/classification , Neoplasm Invasiveness/genetics , Oligonucleotide Array Sequence Analysis
4.
Gastroenterology ; 132(2): 633-44, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17258738

ABSTRACT

BACKGROUND & AIMS: Colon cancers with high-frequency microsatellite instability (MSI-H) develop frameshift mutations in tumor suppressors as part of their pathogenesis. ACVR2 is mutated at its exon 10 polyadenine tract in >80% of MSI-H colon cancers, coinciding with loss of protein. ACVR2 transmits the growth effects of activin via phosphorylation of SMAD proteins to affect gene transcription. The functional effect of activin in colon cancers has not been studied. We developed and characterized a cell model in which we studied how activin signaling affects growth. METHODS: hMLH1 and ACVR2 mutant HCT116 cells were previously stably transferred with chromosome 2 (HCT116+chr2), restoring a single regulated copy of wild-type ACVR2 but not hMLH1. Both HCT116+chr2 and parental HCT116 cells (as well as HEC59 and ACVR2 and hMSH2 complemented HEC59+chr2 cells) were assessed for genetic complementation and biologic function. RESULTS: HCT116+chr2 cells and HEC59+chr2 cells, but not ACVR2-mutant HCT116 or HEC59 cells, acquired wild-type ACVR2 as well as expression of ACVR2 wild-type messenger RNA. Complemented ACVR2 protein complexed with ACVR1 with activin treatment, generating nuclear phosphoSMAD2 and activin-specific gene transcription. ACVR2-restored cells showed decreased growth and reduced S phase but increased cellular migration following activin treatment. ACVR2 small interfering RNA reversed these effects in complemented cells. CONCLUSIONS: ACVR2-complemented MSI-H colon cancers restore activin-SMAD signaling, decrease growth, and slow their cell cycle following ligand stimulation but show increased cellular migration. Activin is growth suppressive and enhances migration similar to transforming growth factor beta in colon cancer, indicating that abrogation of the effects of activin contribute to the pathogenesis of MSI-H colon cancers.


Subject(s)
Activin Receptors, Type II/metabolism , Cell Movement , Cell Proliferation , Colonic Neoplasms/metabolism , Microsatellite Instability , Signal Transduction , Active Transport, Cell Nucleus , Activin Receptors, Type I/metabolism , Activin Receptors, Type II/drug effects , Activin Receptors, Type II/genetics , Activins/metabolism , Activins/pharmacology , Adaptor Proteins, Signal Transducing , Autocrine Communication , Carrier Proteins/metabolism , Cell Movement/drug effects , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Chromosomes, Human, Pair 2/genetics , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Dose-Response Relationship, Drug , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , MutL Protein Homolog 1 , Mutation , Nuclear Proteins/metabolism , Phosphorylation , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction/drug effects , Smad2 Protein/metabolism , Time Factors , Transcriptional Activation , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL