Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
Add more filters










Publication year range
1.
Oncogene ; 35(31): 4036-47, 2016 08 04.
Article in English | MEDLINE | ID: mdl-26657155

ABSTRACT

We previously found that the scaffold adapter GRB2-associated binding protein 2 (GAB2) is amplified and overexpressed in a subset of primary high-grade serous ovarian cancers and cell lines. Ovarian cancer cells overexpressing GAB2 are dependent on GAB2 for activation of the phosphatidylinositol 3-kinase (PI3K) pathway and are sensitive to PI3K inhibition. In this study, we show an important role of GAB2 overexpression in promoting tumor angiogenesis by upregulating expression of multiple chemokines. Specifically, we found that suppression of GAB2 by inducible small hairpin RNA in ovarian cancer cells inhibited tumor cell proliferation, angiogenesis and peritoneal tumor growth in immunodeficient mice. Overexpression of GAB2 upregulated the secretion of several chemokines from ovarian cancer cells, including CXCL1, CXCL2 and CXCL8. The secreted chemokines not only signal through endothelial CXCR2 receptor in a paracrine manner to promote endothelial tube formation, but also act as autocrine growth factors for GAB2-induced transformation of fallopian tube secretory epithelial cells and clonogenic growth of ovarian cancer cells overexpressing GAB2. Pharmacological inhibition of inhibitor of nuclear factor kappa-B kinase subunit ß (IKKß), but not PI3K, mechanistic target of rapamycin (mTOR) or mitogen-activated protein kinase (MEK), could effectively suppress GAB2-induced chemokine expression. Inhibition of IKKß augmented the efficacy of PI3K/mTOR inhibition in suppressing clonogenic growth of ovarian cancer cells with GAB2 overexpression. Taken together, these findings suggest that overexpression of GAB2 in ovarian cancer cells promotes tumor growth and angiogenesis by upregulating expression of CXCL1, CXCL2 and CXCL8 that is IKKß-dependent. Co-targeting IKKß and PI3K pathways downstream of GAB2 might be a promising therapeutic strategy for ovarian cancer that overexpresses GAB2.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Chemokines/genetics , Neovascularization, Pathologic/etiology , Ovarian Neoplasms/pathology , Animals , Chemokine CXCL1/genetics , Chemokine CXCL2/genetics , Endothelial Cells/physiology , Female , Humans , I-kappa B Kinase/physiology , Interleukin-8/genetics , Mice , NF-kappa B/physiology , Ovarian Neoplasms/blood supply , Phosphoinositide-3 Kinase Inhibitors , TOR Serine-Threonine Kinases/antagonists & inhibitors , Up-Regulation
2.
Oncogene ; 31(3): 333-41, 2012 Jan 19.
Article in English | MEDLINE | ID: mdl-21666724

ABSTRACT

Previously, our group identified a novel amplicon at chromosome 9p24 in human esophageal and breast cancers, and cloned the novel gene, GASC1 (gene amplified in squamous cell carcinoma 1, also known as JMJD2C/KDM4C), from this amplicon. GASC1 is a histone demethylase involved in the deregulation of histone methylation in cancer cells. In the current study, we aimed to comprehensively characterize the genes in the 9p24 amplicon in human breast cancer. We performed extensive genomic analyses on a panel of cancer cell lines and narrowed the shortest region of overlap to approximately 2 Mb. Based on statistical analysis of copy number increase and overexpression, the 9p24 amplicon contains six candidate oncogenes. Among these, four genes (GASC1 UHRF2, KIAA1432 and C9orf123) are overexpressed only in the context of gene amplification while two genes (ERMP1 and IL33) are overexpressed independent of the copy number increase. We then focused our studies on the UHRF2 gene, which has a potential involvement in both DNA methylation and histone modification. Knocking down UHRF2 expression inhibited the growth of breast cancer cells specifically with 9p24 amplification. Conversely, ectopic overexpression of UHRF2 in non-tumorigenic MCF10A cells promoted cell proliferation. Furthermore, we demonstrated that UHRF2 has the ability to suppress the expression of key cell-cycle inhibitors, such as p16(INK4a), p21(Waf1/Cip1) and p27(Kip1). Taken together, our studies support the notion that the 9p24 amplicon contains multiple oncogenes that may integrate genetic and epigenetic codes and have important roles in human tumorigenesis.


Subject(s)
Breast Neoplasms/genetics , Carcinoma, Squamous Cell/genetics , Chromosomes, Human, Pair 9/genetics , Gene Amplification/genetics , Carrier Proteins/genetics , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cyclin-Dependent Kinase Inhibitor p16/biosynthesis , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Cyclin-Dependent Kinase Inhibitor p27/biosynthesis , Female , Gene Knockdown Techniques , Guanine Nucleotide Exchange Factors , Humans , Jumonji Domain-Containing Histone Demethylases/genetics , Membrane Proteins/genetics , Peptide Hydrolases/genetics , Ubiquitin-Protein Ligases/genetics , Up-Regulation/genetics
3.
Oncogene ; 28(50): 4491-500, 2009 Dec 17.
Article in English | MEDLINE | ID: mdl-19784073

ABSTRACT

Earlier, mapping of the 9p23-24 amplicon in esophageal cancer cell lines led us to the positional cloning of gene amplified in squamous cell carcinoma 1 (GASC1), which encodes a nuclear protein with a Jumonji C domain that catalyzes lysine (K) demethylation of histones. However, the transforming roles of GASC1 in breast cancer remain to be determined. In this study, we identified GASC1 as one of the amplified genes for the 9p23-24 region in breast cancer, particularly in basal-like subtypes. The levels of GASC1 transcript expression were significantly higher in aggressive, basal-like breast cancers compared with nonbasal-like breast cancers. Our in vitro assays demonstrated that GASC1 induces transformed phenotypes, including growth factor-independent proliferation, anchorage-independent growth, altered morphogenesis in Matrigel, and mammosphere forming ability, when overexpressed in immortalized, nontransformed mammary epithelial MCF10A cells. Additionally, GASC1 demethylase activity regulates the expression of genes critical for stem cell self-renewal, including NOTCH1, and may be linked to the stem cell phenotypes in breast cancer. Thus, GASC1 is a driving oncogene in the 9p23-24 amplicon in human breast cancer and targeted inhibition of GASC1 histone demethylase in cancer could provide potential new avenues for therapeutic development.


Subject(s)
Breast Neoplasms/genetics , Cell Transformation, Neoplastic , Gene Amplification , Jumonji Domain-Containing Histone Demethylases/genetics , Oncogenes , Breast Neoplasms/pathology , Cell Proliferation , Chromosomes, Human, Pair 9 , Female , Humans , RNA, Messenger/analysis , Receptor, Notch1/genetics
4.
Oncogene ; 26(14): 2104-14, 2007 Mar 29.
Article in English | MEDLINE | ID: mdl-17001308

ABSTRACT

Amplification of the 8p11-12 region occurs in 15-20% of breast cancers, but the driving oncogene at this locus has yet to be definitively identified. We mapped the 8p11-12 amplicon in breast cancer cell lines and primary human breast cancers and identified the candidate oncogene human Sm-like protein (hLsm1, LSM1) based on increases in copy number and expression level relative to human mammary epithelial cells. To examine the oncogenic role of LSM1, we overexpressed this gene in MCF10A mammary epithelial cells and inhibited its production in the SUM44 breast cancer cell line, which has a natural amplification and overexpression of LSM1. Our data confirmed that LSM1 is an oncogene from the 8p11-12 amplicon by showing that hLsm1 overexpression induced growth factor-independent proliferation and soft agar colony formation in MCF10A cells, and hLsm1 inhibition in SUM44 cells dramatically reduced soft agar growth. Little is known about hLsm1 function other than its involvement in mRNA degradation; therefore, we used expression microarray analysis to investigate how hLsm1 affects cell transformation in MCF10A and SUM44 cells. We identified numerous genes altered following hLsm1 overexpression common to SUM44 breast cancer cells that play important roles in cell cycle regulation, cell proliferation and other cancer-promoting processes. Future work will continue to characterize these important changes to achieve a more complete understanding of the mechanism of hLsm1's effect on cancer progression.


Subject(s)
Breast Neoplasms/genetics , Cell Transformation, Neoplastic/genetics , Chromosomes, Human, Pair 8/genetics , Gene Amplification , Oncogenes , Proto-Oncogene Proteins/genetics , RNA-Binding Proteins/genetics , Cell Line, Tumor , Cell Proliferation , Culture Media, Conditioned , Female , Gene Dosage , Gene Expression Profiling , Humans , Insulin-Like Growth Factor I/metabolism , Proto-Oncogene Proteins/antagonists & inhibitors , RNA, Small Interfering/pharmacology , RNA-Binding Proteins/antagonists & inhibitors
5.
Oncogene ; 20(30): 4019-28, 2001 Jul 05.
Article in English | MEDLINE | ID: mdl-11494130

ABSTRACT

To gain better understanding of the molecular alterations responsible for the aggressive growth potential of epidermal growth factor receptor (EGFR)-positive breast cancers, we utilized an expression cloning strategy to seek gene products that mediate the EGF-independent growth of human breast cancer cells. A retroviral cDNA expression library was constructed from the EGFR-positive SUM-149PT cell line, and transduced into growth factor-dependent human mammary epithelial (HME) cells. Recipient cells were functionally selected for their ability to proliferate in serum-free, EGF-free medium. Library cDNAs were recovered from EGF-independent colonies by PCR amplification or by biological rescue. Clone H55a#1 contained a library insert encoding amphiregulin. This EGFR ligand was able to confer EGF independence when transduced into HME cells. SUM-149PT and H55a#1 cells overexpressed amphiregulin transcripts, and secreted moderate EGF-like activity in conditioned media, indicating a possible autocrine loop. EGFR membrane levels and constitutive phosphorylation were consistent with this hypothesis, as well as the sensitivity of the cells to an ErbB-specific kinase inhibitor. Expression of the WT1 Wilms' tumor suppressor gene, a transcriptional activator of amphiregulin, did not parallel amphiregulin transcript levels, suggesting that another factor regulates amphiregulin in SUM-149PT. Our data confirm the importance of amphiregulin in the etiology of breast cancer.


Subject(s)
Breast Neoplasms/pathology , Breast/cytology , Epidermal Growth Factor/pharmacology , Genetic Techniques , Glycoproteins/physiology , Growth Substances/physiology , Intercellular Signaling Peptides and Proteins , Amphiregulin , Cell Division/drug effects , Cell Transformation, Neoplastic/genetics , Cells, Cultured/drug effects , Clone Cells/cytology , Clone Cells/drug effects , Culture Media, Conditioned/pharmacology , Culture Media, Serum-Free , DNA, Complementary/genetics , EGF Family of Proteins , Epithelial Cells/cytology , Epithelial Cells/drug effects , ErbB Receptors/genetics , ErbB Receptors/physiology , Female , Gene Library , Genes, Wilms Tumor , Genetic Complementation Test , Genetic Vectors/genetics , Glycoproteins/genetics , Growth Substances/genetics , Humans , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/drug effects , Phenotype , Retroviridae/genetics , Transfection , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/drug effects
6.
Cell Growth Differ ; 12(7): 371-8, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11457734

ABSTRACT

Insulin-like growth factor I (IGF-I) protects cells from apoptosis primarily through the action of phosphatidylinositol-3 kinase and the downstream serine/threonine kinase Akt. The PTEN gene product, a protein which dephosphorylates phosphatidylinositol lipids, prevents activation of Akt and regulates several cellular functions, including cell cycle progression, cell migration, and survival from apoptosis. In this study, PTEN overexpression decreases IGF-I-induced Akt activity, enhances serum withdrawal-induced apoptosis, and decreases IGF-I protection and cell growth in SHEP cells. The PTEN lipid phosphatase mutant G129E fails to inhibit IGF-I-stimulated Akt activity and protection from apoptosis. The C124S mutation, which abolishes both lipid and protein phosphatase activity, fails to inhibit Akt activity and IGF-I protection against hyperosmotic-induced apoptosis but still inhibits growth and IGF-I protection against serum withdrawal-induced apoptosis. These data suggest a role for PTEN in modulating the effect of IGF-I on Akt activity, neuroblastoma cell growth, and protection against apoptotic stimuli.


Subject(s)
Apoptosis/physiology , Insulin-Like Growth Factor I/metabolism , Phosphoric Monoester Hydrolases/metabolism , Protein Serine-Threonine Kinases , Tumor Suppressor Proteins , Apoptosis/drug effects , Cell Division/physiology , Culture Media, Serum-Free , Diuretics, Osmotic/pharmacology , Flow Cytometry , Immunoblotting , Mannitol/pharmacology , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Neuroblastoma , PTEN Phosphohydrolase , Phosphoric Monoester Hydrolases/genetics , Phosphorylation , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Tumor Cells, Cultured
7.
Cancer Res ; 61(13): 5168-78, 2001 Jul 01.
Article in English | MEDLINE | ID: mdl-11431356

ABSTRACT

With the goal of identifying genes that have an expression pattern that can facilitate the diagnosis of primary breast cancers (BCs) as well as the discovery of novel drug leads for BC treatment, we used cDNA hybridization arrays to analyze the gene expression profiles (GEPs) of nine weakly invasive and four highly invasive BC cell lines. Differences in gene expression between weakly and highly invasive BC cells were identified that enabled the definition of consensus GEPs for each invasive phenotype. To determine whether the consensus GEPs, comprising 24 genes, could be used to predict the aggressiveness of previously uncharacterized cells, gene expression levels and comparative invasive and migratory characteristics of nine additional human mammary epithelial cell strains/lines were determined. The results demonstrated that the GEP of a cell line is predictive of its invasive and migratory behavior, as manifest by the morphology of its colonies when cultured on a matrix of basement membrane constituents (i.e., Matrigel). We found that the expression of keratin 19 was consistently elevated in the less aggressive BC cell lines and that vimentin and fos-related antigen-1 (FRA-1) were consistently overexpressed in the more highly aggressive BC cells. Moreover, even without these three genes, the GEP of a cell line still accurately predicted the aggressiveness of the BC cell, indicating that the expression pattern of multiple genes may be used as BC prognosticators because single markers often fail to be predictive in clinical specimens.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Gene Expression Profiling , Animals , Breast Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Humans , Mice , Neoplasm Invasiveness , Neoplasm Metastasis , Oligonucleotide Array Sequence Analysis , Predictive Value of Tests , Tumor Cells, Cultured
9.
Anal Chem ; 73(6): 1219-27, 2001 Mar 15.
Article in English | MEDLINE | ID: mdl-11305655

ABSTRACT

Nonporous (NPS) RP-HPLC has been used to rapidly separate proteins from whole cell lysates of human breast cell lines. The nonporous separation involves the use of hard-sphere silica beads of 1.5-microm diameter coated with C18, which can be used to separate proteins ranging from 5 to 90 kDa. Using only 30-40 microg of total protein, the protein molecular weights are detectable on-line using an ESI-oaTOF MS. Of hundreds of proteins detected in this mass range, approxinately 75-80 are more highly expressed. The molecular weight profiles can be displayed as a mass map analogous to a virtual "1-D gel" and differentially expressed proteins can be compared by image analysis. The separated proteins can also be detected by UV absorption and differentially expressed proteins quantified. The eluting proteins can be collected in the liquid phase and the molecular weight and peptide maps determined by MALDI-TOF MS for identification. It is demonstrated that the expressed protein profiles change during neoplastic progression and that many oncoproteins are readily detected. It is also shown that the response of premalignant cancer cells to estradiol can be rapidly screened by this method, demonstrating significant changes in response to an external agent. Ultimately, the proteins can be studied by peptide mapping to search for posttranslational modifications of the oncoproteins accompanying progression.


Subject(s)
Neoplasm Proteins/analysis , Cell Transformation, Neoplastic , Chromatography, High Pressure Liquid , Databases, Factual , Humans , Molecular Weight , Spectrometry, Mass, Electrospray Ionization , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tumor Cells, Cultured
10.
Int J Radiat Oncol Biol Phys ; 48(5): 1519-28, 2000 Dec 01.
Article in English | MEDLINE | ID: mdl-11121658

ABSTRACT

PURPOSE: Overexpression of the ErbB family of growth factor receptors is present in a wide variety of human tumors and is correlated with poor prognosis. The purpose of this study was to determine the effects of a novel small molecule ErbB tyrosine kinase inhibitor, CI-1033, in combination with ionizing radiation on breast cancer cell growth and survival. MATERIALS & METHODS: Growth assays were performed on ErbB-overexpressing human breast cancer cells developed in our laboratory in the presence of 0.1-1.0 microM CI-1033 (Parke Davis). Clonogenic survival assays were performed in the presence of ionizing radiation with or without CI-1033. For some experiments, clonogen numbers, defined as the product of surviving fraction and total number of cells, were calculated at each time point during a course of multifraction radiation. RESULTS: CI-1033 potently inhibited the growth of ErbB-overexpressing breast cancer cells. A single 48-h exposure of 1 microM CI-1033 resulted in growth inhibition for 7 days, whereas three times weekly administration resulted in sustained growth inhibition. Clonogenic survival was modestly decreased after a 7-day exposure to CI-1033. Exposure to both CI-1033 and radiation (6 Gy) yielded a 23-fold decrease in clonogenic survival compared to radiation alone. In a multifraction experiment, exposure to CI-1033 and three 5-Gy fractions of gamma radiation decreased the total number of clonogens in the population by 65-fold compared to radiation alone. CONCLUSION: CI-1033 results in potent growth inhibition and modest cytotoxicity of ErbB-overexpressing breast cancer cells, and has synergistic effects when combined with ionizing radiation. These data suggest that CI-1033 may have excellent clinical potential both alone and in combination with radiation therapy.


Subject(s)
Breast Neoplasms/radiotherapy , Enzyme Inhibitors/therapeutic use , Neoplasm Proteins/antagonists & inhibitors , Radiation-Sensitizing Agents/therapeutic use , Receptor, ErbB-2/antagonists & inhibitors , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Division/drug effects , Cell Survival/radiation effects , Dose Fractionation, Radiation , ErbB Receptors/metabolism , Female , Humans , Radiation Tolerance , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/radiation effects , Tumor Stem Cell Assay
11.
Genes Chromosomes Cancer ; 29(4): 356-62, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11066082

ABSTRACT

Breast cancer is a heterogeneous disorder in which most tumors display some degree of aneuploidy, especially those at later stages of the disease. Aneuploidy and associated chromosome instability may be important in the progression of mammary tumorigenesis. Aneuploidy is prevented during normal cell division in part through regulation of a mitotic spindle checkpoint where mitotic arrest prevents segregation of misaligned chromosomes into daughter cells at anaphase. Mitotic arrest genes, including the MAD family, which was originally characterized in yeast, help regulate normal function of the mitotic spindle checkpoint. Decreased expression of the human gene MAD2L1 was previously reported in a breast cancer cell line exhibiting chromosome instability and aneuploidy. To explore further the potential role of MAD2L1 in breast cancer, we analyzed MAD2L1 gene expression in 13 minimally to grossly aneuploid human breast cancer cell lines and found significant differences of expression in three lines. Sequence analysis of MAD2L1 cDNA in these as well as nine additional aneuploid breast cancer and five immortalized normal human mammary epithelial cell lines revealed one heterozygous frameshift (572 del A) mutation in a cancer cell line that demonstrated a high level of transcript expression. In addition, two 3'UTR sequence variants were noted in breast cancer cell lines. The 572 del A mutation creates a truncated MAD2 protein product. Further functional studies in primary breast tumors are therefore warranted to determine the potential role MAD2L1 may play in breast cancer.


Subject(s)
Breast Neoplasms/genetics , Calcium-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic/genetics , Amino Acid Substitution/genetics , Calcium-Binding Proteins/biosynthesis , Cell Cycle Proteins , Cell Line , Cell Line, Transformed , DNA Mutational Analysis , DNA, Neoplasm/analysis , Frameshift Mutation , Genes, Neoplasm , Humans , Mad2 Proteins , Repressor Proteins , Sequence Deletion , Tumor Cells, Cultured
12.
Breast Cancer Res ; 2(4): 311-20, 2000.
Article in English | MEDLINE | ID: mdl-11056689

ABSTRACT

The fibroblast growth factor receptor (FGFR)2 gene has been shown to be amplified in 5-10% of breast cancer patients. A breast cancer cell line developed in our laboratory, SUM-52PE, was shown to have a 12-fold amplification of the FGFR2 gene, and FGFR2 message was found to be overexpressed 40-fold in SUM-52PE cells as compared with normal human mammary epithelial (HME) cells. Both human breast cancer (HBC) cell lines and HME cells expressed two FGFR2 isoforms, whereas SUM-52PE cells overexpressed those two isoforms, as well as several unique FGFR2 polypeptides. SUM-52PE cells expressed exclusively FGFR2-IIIb isoforms, which are high-affinity receptors for fibroblast growth factor (FGF)-1 and FGF-7. Differences were identified in the expression of the extracellular Ig-like domains, acid box and carboxyl termini, and several variants not previously reported were isolated from these cells.


Subject(s)
Breast Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/biosynthesis , Protein Isoforms/biosynthesis , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptors, Fibroblast Growth Factor/biosynthesis , Tumor Cells, Cultured/metabolism , Alternative Splicing , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line , Cloning, Molecular , Exons/genetics , Female , Fibroblast Growth Factor 1 , Fibroblast Growth Factor 2/metabolism , Fibroblast Growth Factor 7 , Fibroblast Growth Factors/metabolism , Gene Amplification , Humans , Neoplasm Proteins/genetics , Protein Isoforms/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptor, Fibroblast Growth Factor, Type 2 , Receptors, Fibroblast Growth Factor/genetics , Recombinant Fusion Proteins/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Substrate Specificity , Transfection
13.
Radiat Res ; 154(5): 590-9, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11025655

ABSTRACT

The underlying causes for different apoptotic responses in neoplastic cells are still not fully understood. We demonstrate here that a human breast cancer cell line, MDA-MB-468, which lacks the retinoblastoma protein (RB), is particularly sensitive to low doses of ultraviolet (UV) radiation. These cells are 15-20-fold more sensitive to UV radiation than RB-positive cell lines, as measured by both apoptosis and clonogenic assays. In addition, a prostate cancer cell line that lacks functional RB, DU-145, was found to have a similar apoptotic response to low doses of UV radiation. Based on these data, we hypothesized that the lack of RB is responsible for the extreme sensitivity of these cells to UV-radiation-induced apoptosis. To further examine the role of RB in apoptosis, cells of RB-positive human breast cancer and normal cell lines were infected with the human papilloma virus type 16 (HPV-16) E7 and assessed for UV-radiation sensitivity. The HPV-16 E7 protein is known to decrease levels of free RB in cells. Infection of RB-positive human breast cancer or normal cells with E7 resulted in a 4-5-fold increase in sensitivity to UV radiation compared to controls. The above data suggest a role for the RB protein in protecting cells from undergoing apoptosis in response to UV radiation.


Subject(s)
Apoptosis , Radiation Tolerance , Retinoblastoma Protein/physiology , Ultraviolet Rays , Breast Neoplasms , Enzyme Inhibitors/pharmacology , ErbB Receptors/antagonists & inhibitors , Humans , Tumor Cells, Cultured , Tumor Stem Cell Assay
14.
Cancer Res ; 60(16): 4519-25, 2000 Aug 15.
Article in English | MEDLINE | ID: mdl-10969801

ABSTRACT

Breast cancer cell lines provide a useful starting point for the discovery and functional analysis of genes involved in breast cancer. Here, we studied 38 established breast cancer cell lines by comparative genomic hybridization (CGH) to determine recurrent genetic alterations and the extent to which these cell lines resemble uncultured tumors. The following chromosomal gains were observed: 8q (75%), 1q (61%), 20q (55%), 7p (44%), 3q (39%), 5p (39%), 7q (39%), 17q (33%), 1p (30%), and 20p (30%), and the most common losses were: 8p (58%), 18q (58%), 1p (42%), Xp (42%), Xq (42%), 4p (36%), 11q (36%), 18p (33%), 10q (30%), and 19p (28%). Furthermore, 35 recurrent high-level amplification sites were identified, most often involving 8q23 (37%), 20q13 (29%), 3q25-q26 (24%), 17q22-q23 (16%), 17q23-q24 (16%), 1p13 (11%), 1q32 (11%), 5p13 (11%), 5p14 (11%), 11q13 (11%), 17q12-q21 (11%), and 7q21-q22 (11%). A comparison of DNA copy number changes found in the cell lines with those reported in 17 published studies (698 tumors) of uncultured tumors revealed a substantial degree of overlap. CGH copy number profiles may facilitate identification of important new genes located at the hotspots of such chromosomal alterations. This was illustrated by analyzing expression levels of 1236 genes using cDNA microarrays in four of the cell lines. Several highly overexpressed genes (such as RCH1 at 17q23, TOPO II at 17q21-q22, as well as CAS and MYBL2 at 20q13) were involved in these recurrent DNA amplifications. In conclusion, DNA copy number profiles were generated by CGH for most of the publicly available breast cancer cell lines and were made available on a web site (http://www.nhgri.nih.gov/DIR/CGB/++ +CR2000). This should facilitate the correlative analysis of gene expression and copy number as illustrated here by the finding by cDNA microarrays of several overexpressed genes that were amplified.


Subject(s)
Breast Neoplasms/genetics , DNA, Complementary/genetics , DNA, Neoplasm/genetics , Gene Expression Profiling/methods , Nucleic Acid Hybridization/methods , Breast Neoplasms/metabolism , Chromosome Deletion , Female , Gene Amplification , Humans , In Situ Hybridization, Fluorescence , Male , Tumor Cells, Cultured
15.
Neoplasia ; 2(3): 251-60, 2000.
Article in English | MEDLINE | ID: mdl-10935511

ABSTRACT

Oncogenes which drive the cell cycle, such as c-myc, can sensitize cells to apoptosis. This suggests the possibility that the expression of genes such as bcl-2 or bcl-xL is required to inhibit apoptosis induced by oncogene expression. We hypothesized that inhibition of Bcl-2/Bcl-xL by the pro-apoptotic Bcl-xS protein, would result in selective induction of apoptosis in mammary carcinoma cells compared to their nontransformed counterparts. Therefore, we compared the effects of Bcl-xS expression delivered by a bcl-xS adenovirus (bcl-xS-Adv) vector, on viability and apoptosis of nontransformed versus transformed mammary epithelial cells. We report that c-myc-transformed murine mammary cells are extremely sensitive to apoptosis induced by the bcl-xS adenovirus (bcl-xS-Adv) vector, whereas immortalized, nontransformed murine mammary cells are relatively resistant to apoptosis induced by this vector. Likewise, human mammary epithelial cells transduced with c-erbB-2 were more sensitive to apoptosis induced by the bcl-xS vector than the nontransformed parental cells. Similar results were obtained when we tested the effects of bcl-xS adenoviral infection on primary normal human mammary epithelial cells and SUM-190 PT cells, (a c-erbB-2 over-expressing human mammary carcinoma cell line) grown on Matrigel. These data are consistent with the hypothesis that inhibition of Bcl-2/Bcl-xL can result in selective killing of cancer cells compared to their nontransformed counterparts.


Subject(s)
Adenoviridae/genetics , Apoptosis , Mammary Glands, Animal/cytology , Mammary Neoplasms, Experimental/therapy , Proto-Oncogene Proteins c-bcl-2/physiology , Animals , Cell Line , Cell Transformation, Neoplastic , Fluorouracil/pharmacology , Genes, myc , Humans , Paclitaxel/pharmacology , Proto-Oncogene Proteins c-bcl-2/genetics , Receptor, ErbB-2/physiology , bcl-X Protein
16.
J Clin Invest ; 105(11): 1537-45, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10841512

ABSTRACT

The PHSRN sequence of the plasma fibronectin (pFn) cell-binding domain induces human keratinocytes and fibroblasts to invade the naturally serum-free extracellular matrices of sea urchin embryos. The potency of acetylated, amidated PHSRN (Ac-PHSRN-NH(2)) is significantly increased, making it more active on a molar basis than the 120-kDa cell-binding domain of pFn. Arginine is important to this activity because PHSAN and PHSEN are inactive, as is a randomized sequence peptide, Ac-HSPNR-NH(2). One treatment with Ac-PHSRN-NH(2) stimulates reepithelialization and contraction of dermal wounds in healing-impaired, obese diabetic C57BL6/KsJ db/db mice. Wound closure is equally rapid in treated db/db and db/+ mice and may be more rapid than in untreated nondiabetic db/+ littermates. In contrast, treatment with either Ac-HSPNR-NH(2) or normal saline (NS) has no effect. Analysis of sectioned db/db wounds shows that, in contrast to treatment with Ac-HSPNR-NH(2) or NS, a single Ac-PHSRN-NH(2) treatment stimulates keratinocyte and fibroblast migration into wounds, enhances fibroplasia and vascularization in the provisional matrix, and stimulates the formation of prominent fibers that may be associated with wound contraction.


Subject(s)
Chemotactic Factors/pharmacology , Diabetes Mellitus/physiopathology , Extracellular Matrix/drug effects , Fibronectins/pharmacology , Peptide Fragments/pharmacology , Wound Healing/drug effects , Animals , Binding Sites , Cell Movement , Cells, Cultured , Fibroblasts/physiology , Keratinocytes/physiology , Mice , Mice, Inbred C57BL , Mice, Obese , Receptors, Fibronectin/physiology
17.
J Cell Physiol ; 183(3): 301-13, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10797304

ABSTRACT

The neu differentiation factors/heregulins (HRGs) comprise a family of polypeptide growth factors that activate p185(erbB-2) through direct binding to either erbB-3 or erbB-4 receptor tyrosine kinases. We have previously shown that HRG-beta is mitogenic for various human mammary epithelial cell lines that coexpress c-erbB-2 and c-erbB-3. Phosphatidylinositol 3-kinase (PI3K) is activated by p185(erbB-2) /erbB-3 heterodimers in cells stimulated by HRG, and PI3K is constitutively activated by p185(erbB-2) /erbB-3 in breast carcinoma cells that overexpress c-erbB-2. To better understand the relative abilities of HRGs, epidermal growth factor (EGF), or insulin to activate PI3K under normal physiological conditions, we compared the levels of recruitment of the 85-kDa regulatory subunit of PI3K when activated by the type I (erbB) or type II [insulin-like growth factor (IGF)] receptor tyrosine kinases in two different nontransformed human mammary epithelial cell lines. The nontransformed H16N-2 cells isolated from normal tissue express EGFR, p185(erbB-2), and erbB-3, and are highly responsive to the mitogenic effects of HRG-beta as well as to the combination of EGF and insulin in serum-free culture. We measured the stoichiometry of p85 recruited by tyrosine-phosphorylated proteins induced in H16N-2 cells by either the alpha or the beta isoform of HRG. HRG-beta was greater than 10-fold more potent in inducing p85 recruitment than was the less biologically active HRG-alpha isoform. HRG-beta was also a more potent inducer of p85 recruited by tyrosine-phosphorylated proteins than was either EGF, insulin, or EGF and insulin combined. Furthermore, erbB-3 principally mediated the direct recruitment of p85 in cells stimulated by HRG or EGF, indicating that, in addition to the high-level activation of PI3K by p185(erbB-2) / erbB-3, EGFR/erbB-3 heterodimer interaction is essential for the weak but significant level of PI3K activated by EGF in cells that express normal EGFR levels. Studies using the PI3K inhibitor wortmannin also indicated that PI3K activation was required for the proliferation of H16N-2 cells induced by either HRG-beta or EGF and insulin in serum-free culture. Finally, HRG-beta was also an especially potent inducer of PI3K in the nontransformed MCF-10A cells, which were derived spontaneously from normal reduction mammoplasty tissue. These data show, for the first time, a side-by-side quantitative comparison of the relative degree of PI3K activated by different growth factors in nontransformed growth factor-dependent cells under precisely defined conditions in culture.


Subject(s)
Breast/cytology , Epithelial Cells/physiology , Neuregulin-1/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Androstadienes/pharmacology , Breast Neoplasms , Cell Division/drug effects , Cell Line , Enzyme Activation , Epidermal Growth Factor/pharmacology , Epithelial Cells/cytology , Epithelial Cells/drug effects , ErbB Receptors/genetics , ErbB Receptors/physiology , Female , Humans , Insulin/pharmacology , Receptor, ErbB-2/genetics , Receptor, ErbB-2/physiology , Receptor, ErbB-3/genetics , Receptor, ErbB-3/physiology , Tumor Cells, Cultured , Wortmannin
18.
Br J Cancer ; 82(3): 666-74, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10682681

ABSTRACT

Amplification and overexpression of ERBB-2 in human breast cancer is thought to play a significant role in the progression of the disease; however, its precise role in the aetiology of altered phenotypes associated with human breast cancer is unknown. We have previously shown that exogenous overexpression of ERBB-2 conferred growth factor independence on human mammary epithelial cells. In this study, we show that ERBB-2 overexpression also causes the cells to acquire other characteristics exhibited by human breast cancer cells, such as anchorage-independent growth and invasion capabilities. ERBB-2-induced invasion is dependent on fibronectin and correlates with the down-regulation of cell surface alpha4 integrin. In addition ERBB-2 co-immunoprecipitates with focal adhesion kinase (FAK) in these cells. We have also shown, by use of exogenously expressed PTEN and by treatment with the PI3'-kinase inhibitor LY294002, that ERBB-2-induced invasion is dependent on the PI3'-kinase pathway; however, PTEN does not dephosphorylate FAK in these cells.


Subject(s)
Breast Neoplasms/metabolism , Breast/metabolism , Genes, erbB-2 , Neoplasm Invasiveness/genetics , Phosphatidylinositol 3-Kinases/metabolism , Breast/cytology , Breast/enzymology , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Division , Cell Survival , Humans , Phenotype , Signal Transduction
19.
Br J Cancer ; 81(8): 1328-34, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10604729

ABSTRACT

We describe a survey of genetic changes by comparative genomic hybridization (CGH) in 11 human breast cancer cell lines recently established in our laboratory. The most common gains took place at 8q (73%), 1 q (64%), 7q (64%), 3q (45%) and 7p (45%), whereas losses were most frequent at Xp (54%), 8p (45%), 18q (45%) and Xq (45%). Many of the cell lines displayed prominent, localized DNA amplifications by CGH. One-third of these loci affected breast cancer oncogenes, whose amplifications were validated with specific probes: 17q12 (two cell lines with ERBB2 amplifications), 11q13 (two with cyclin-D1), 8p11-p12 (two with FGFR1) and 10q25 (one with FGFR2). Gains and amplifications affecting 8q were the most common genetic alterations in these cell lines with the minimal, common region of involvement at 8q22-q23. No high-level MYC (at 8q24) amplifications were found in any of the cell lines. Two-thirds of the amplification sites took place at loci not associated with established oncogenes, such as 1q41-q43, 7q21-q22, 7q31, 8q23, 9p21-p23, 11p12-p14, 15q12-q14, 16q13-q21, 17q23, 20p11-p12 and 20q13. Several of these locations have not been previously reported and may harbour important genes whose amplification is selected for during cancer development. In summary, this set of breast cancer cell lines displaying prominent DNA amplifications should facilitate discovery and functional analysis of genes and signal transduction pathways contributing to breast cancer development.


Subject(s)
Breast Neoplasms/genetics , DNA, Neoplasm/genetics , Gene Amplification , Humans , Nucleic Acid Hybridization , Oncogenes , Sequence Deletion , Tumor Cells, Cultured
20.
Clin Cancer Res ; 5(9): 2511-9, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10499627

ABSTRACT

Inflammatory breast cancer is a rapidly growing, distinct form of locally advanced breast cancer that carries a guarded prognosis. To identify the genes that contribute to this aggressive phenotype, we compared under- and overexpressed sequences in an inflammatory breast tumor cell line with those of actively replicating normal human mammary epithelial cell lines using differential display. Of the 17 transcripts isolated and characterized from these experiments, overexpression of RhoC GTPase and loss of expression of a novel gene on 6q22, LIBC (lost in inflammatory breast cancer), were highly correlated (P<0.0095 and P<0.0013, respectively) with the inflammatory phenotype when a panel of archival inflammatory breast cancers was compared with noninflammatory stage III breast cancers by in situ hybridization. This study suggests two new molecular markers specific for inflammatory breast cancer.


Subject(s)
Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , GTP Phosphohydrolases/genetics , Immediate-Early Proteins , Insulin-Like Growth Factor Binding Proteins/genetics , Insulin-Like Growth Factor Binding Proteins/metabolism , Intercellular Signaling Peptides and Proteins , Neoplasm Proteins/genetics , rho GTP-Binding Proteins/genetics , Amino Acid Sequence , Animals , Biomarkers, Tumor/isolation & purification , Breast/cytology , Breast Neoplasms/enzymology , Breast Neoplasms/metabolism , CCN Intercellular Signaling Proteins , Cattle , Connective Tissue Growth Factor , Epithelial Cells/metabolism , GTP Phosphohydrolases/biosynthesis , Genes, Tumor Suppressor , Growth Substances/chemistry , Humans , Insulin-Like Growth Factor Binding Proteins/isolation & purification , Mice , Molecular Sequence Data , Neoplasm Proteins/isolation & purification , Neoplasm Proteins/metabolism , Phenotype , RNA, Messenger/isolation & purification , RNA, Messenger/metabolism , Reproducibility of Results , Swine , Transcription, Genetic , Tumor Cells, Cultured , ras Proteins , rho GTP-Binding Proteins/biosynthesis , rhoC GTP-Binding Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...