Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 7(10): e47876, 2012.
Article in English | MEDLINE | ID: mdl-23077670

ABSTRACT

Expression of the heparin-binding growth factor, pleiotrophin (PTN) in the mammary gland has been reported but its function during mammary gland development is not known. We examined the expression of PTN and its receptor ALK (Anaplastic Lymphoma Kinase) at various stages of mouse mammary gland development and found that their expression in epithelial cells is regulated in parallel during pregnancy. A 30-fold downregulation of PTN mRNA expression was observed during mid-pregnancy when the mammary gland undergoes lobular-alveolar differentiation. After weaning of pups, PTN expression was restored although baseline expression of PTN was reduced significantly in mammary glands of mice that had undergone multiple pregnancies. We found PTN expressed in epithelial cells of the mammary gland and thus used a monoclonal anti-PTN blocking antibody to elucidate its function in cultured mammary epithelial cells (MECs) as well as during gland development. Real-time impedance monitoring of MECs growth, migration and invasion during anti-PTN blocking antibody treatment showed that MECs motility and invasion but not proliferation depend on the activity of endogenous PTN. Increased number of mammospheres with laminin deposition after anti-PTN blocking antibody treatment of MECs in 3D culture and expression of progenitor markers suggest that the endogenously expressed PTN inhibits the expansion and differentiation of epithelial progenitor cells by disrupting cell-matrix adhesion. In vivo, PTN activity was found to inhibit ductal outgrowth and branching via the inhibition of phospho ERK1/2 signaling in the mammary epithelial cells. We conclude that PTN delays the maturation of the mammary gland by maintaining mammary epithelial cells in a progenitor phenotype and by inhibiting their differentiation during mammary gland development.


Subject(s)
Carrier Proteins , Cytokines , Epithelial Cells/metabolism , Gene Expression Regulation , Mammary Glands, Animal/metabolism , Anaplastic Lymphoma Kinase , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Culture Techniques , Cell Differentiation , Cell-Matrix Junctions/metabolism , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Female , Humans , MAP Kinase Signaling System , Pregnancy , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism
2.
Breast Cancer Res Treat ; 116(2): 225-37, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19418218

ABSTRACT

AIB1 (amplified in breast cancer 1), also called SRC-3 and NCoA-3, is a member of the p160 nuclear receptor co-activator family and is considered an important oncogene in breast cancer. Increased AIB1 levels in human breast cancer have been correlated with poor clinical prognosis. Overexpression of AIB1 in conjunction with members of the epidermal growth factor receptor (EGF/HER) tyrosine kinase family, such as HER2, is associated with resistance to tamoxifen therapy and decreased disease-free survival. A number of functional studies in cell culture and in rodents indicate that AIB1 has a pleiotropic role in breast cancer. Initially AIB1 was shown to have a role in the estrogen-dependent proliferation of breast epithelial cells. However, AIB1 also affects the growth of hormone-independent breast cancer and AIB1 levels are limiting for IGF-1-, EGF- and heregulin-stimulated biological responses in breast cancer cells and consequently the PI3 K/Akt/mTOR and other EGFR/HER2 signaling pathways are controlled by changes in AIB1 protein levels. The cellular levels and activity of AIB1 are in turn regulated at the levels of transcription, mRNA stability, post-translational modification, and by a complex control of protein half life. In particular, AIB1 activity as well as its half-life is modulated through a number of post-translational modifications including serine, threonine and tyrosine phosphorylation via kinases that are components of multiple signal transduction pathways. This review summarizes the possible mechanisms of how dysregulation of AIB1 at multiple levels can lead to the initiation and progression of breast cancer as well as its role as a predictor of response to breast cancer therapy, and as a possible therapeutic target.


Subject(s)
Breast Neoplasms/genetics , Histone Acetyltransferases/metabolism , Trans-Activators/metabolism , Animals , Biomarkers, Tumor/analysis , Breast Neoplasms/metabolism , Drug Resistance, Neoplasm/genetics , Female , Gene Expression , Gene Expression Regulation, Neoplastic , Humans , Nuclear Receptor Coactivator 3
3.
Methods ; 38(4): 253-62, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16513366

ABSTRACT

Gene expression in diseased tissues can indicate the contribution to a disease process and potentially guide therapeutic decision-making. Archival tissues with associated clinical outcome may be useful to discover or validate the role of a candidate gene in a disease process or the response to therapy. Such archival tissues are commonly formalin-fixed and paraffin-embedded, restricting the methods available for gene expression analysis. Obviously, the detection of proteins in tissues requires adaptation for each protein and the detection of secreted proteins can prove difficult or of reduced value since the protein detected may not reflect the total amount produced. Thus, we describe here a reliable method for the detection of mRNA in archival tissues. The method for mRNA in situ hybridization (ISH) was adapted by us for >15 different genes and applied to several hundred tissue microarrays (TMAs) and full sections generating >10,000 expression data points. We also discuss the utility of TMAs to simultaneously analyze several hundred tissue samples on one slide to minimize variability and preserve valuable tissue samples. Experimental protocols are provided that can be implemented without major hurdles in a typical molecular pathology laboratory and we discuss quantitative analysis as well as advantages and limitations of ISH with a special focus on secreted proteins. We conclude that ISH is a reliable and cost effective approach to gene expression analysis in archival tissues that is amenable to screening of series of tissues or of genes of interest.


Subject(s)
Formaldehyde/pharmacology , Gene Expression Profiling/instrumentation , Gene Expression Profiling/methods , RNA, Messenger/metabolism , Animals , Fixatives , Formaldehyde/chemistry , Gene Expression , Humans , In Situ Hybridization , Oligonucleotide Probes/chemistry , Oligonucleotides, Antisense/chemistry , Paraffin/chemistry , Paraffin/pharmacology , Paraffin Embedding , Polymerase Chain Reaction , Protein Array Analysis , Proteins/analysis , Proteins/metabolism , RNA/metabolism , Reproducibility of Results
4.
Cancer Res ; 66(2): 1191-8, 2006 Jan 15.
Article in English | MEDLINE | ID: mdl-16424058

ABSTRACT

The activity of growth factors is crucial for tumor progression. We previously characterized a secreted fibroblast growth factor-binding protein (FGF-BP1) as a chaperone molecule, which enhances the biological functions of FGFs by releasing FGFs from the extracellular matrix. Here, we characterize the frequency and pattern of FGF-BP1 expression during the malignant progression of pancreas and colorectal carcinoma. For this, we generated monoclonal antibodies that detect FGF-BP1 protein in formalin-fixed, paraffin-embedded tissues and applied in situ hybridization to detect FGF-BP1 mRNA in adjacent tissue sections. FGF-BP1 protein and mRNA were found up-regulated (>70% positive) in parallel (r = 0.70, P < 0.0001) in colon adenoma (n = 9) as well as primary (n = 46) and metastatic (n = 71) colorectal cancers relative to normal colon epithelia (all P < 0.0001, versus normal). Similarly, pancreatitis (n = 17), pancreatic intraepithelial neoplasia (n = 80), and pancreatic adenocarcinoma (n = 67) showed a significant up-regulation of FGF-BP1 compared with normal pancreas (n = 42; all P < 0.0001, relative to normal). Furthermore, the biological activity of FGF-BP1 is neutralized by one of the antibodies, suggesting the potential for antibody-based therapeutic targeting. We propose that the up-regulation of the secreted FGF-BP1 protein during initiation of pancreas and colon neoplasia could make this protein a possible serum marker indicating the presence of high-risk premalignant lesions.


Subject(s)
Adenocarcinoma/genetics , Carrier Proteins/biosynthesis , Colonic Neoplasms/genetics , Pancreatic Neoplasms/genetics , Adenocarcinoma/pathology , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Carrier Proteins/physiology , Cell Transformation, Neoplastic , Colonic Neoplasms/pathology , Disease Progression , Gene Expression Profiling , Humans , In Situ Hybridization , Intercellular Signaling Peptides and Proteins , Pancreatic Neoplasms/pathology , Pancreatitis/genetics , Pancreatitis/pathology , Pancrelipase/physiology , RNA, Messenger/biosynthesis , Risk Assessment , Tumor Cells, Cultured , Up-Regulation
5.
Mol Endocrinol ; 19(3): 644-56, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15550471

ABSTRACT

Amplified in breast cancer 1 (AIB1, also known as ACTR, SRC-3, RAC-3, TRAM-1, p/CIP) is a member of the p160 nuclear receptor coactivator family involved in transcriptional regulation of genes activated through steroid receptors, such as estrogen receptor alpha (ER(alpha)). The AIB1 gene and a more active N-terminally deleted isoform (AIB1-Delta3) are overexpressed in breast cancer. To determine the role of AIB1-Delta3 in breast cancer pathogenesis, we generated transgenic mice with human cytomegalovirus immediate early gene 1 (hCMVIE1) promoter-driven over-expression of human AIB1/ACTR-Delta3 (CMVAIB1/ACTR-Delta3 mice). AIB1/ACTR-Delta3 transgene mRNA expression was confirmed in CMV-AIB1/ACTR-Delta3 mammary glands by in situ hybridization. These mice demonstrated significantly increased mammary epithelial cell proliferation (P < 0.003), cyclin D1 expression (P = 0.002), IGF-I receptor protein expression (P = 0.026), mammary gland mass (P < 0.05), and altered expression of CCAAT/enhancer binding protein isoforms (P = 0.029). At 13 months of age, mammary ductal ectasia was found in CMV-AIB1/ACTR-Delta3 mice, but secondary and tertiary branching patterns were normal. There were no changes in the expression patterns of either ER(alpha) or Stat5a, a downstream mediator of prolactin signaling. Serum IGF-I levels were not altered in the transgenic mice. These data indicate that overexpression of the AIB1/ACTR-Delta3 isoform resulted in altered mammary epithelial cell growth. The observed changes in cell proliferation and gene expression are consistent with alterations in growth factor signaling that are thought to contribute to either initiation or progression of breast cancer. These results are consistent with the hypothesis that the N-terminally deleted isoform of AIB1 can play a role in breast cancer development and/or progression.


Subject(s)
Transcription Factors/chemistry , Alternative Splicing , Animals , Antigens, Viral/genetics , Blotting, Southern , Blotting, Western , Breast Neoplasms/embryology , Cell Proliferation , Cyclin D1/metabolism , DNA/metabolism , DNA-Binding Proteins/chemistry , Enzyme-Linked Immunosorbent Assay , Epithelial Cells/metabolism , Estrogen Receptor alpha/metabolism , Gene Expression Regulation , Genotype , Humans , Immediate-Early Proteins/genetics , Immunoblotting , Immunohistochemistry , In Situ Hybridization , Insulin-Like Growth Factor I/biosynthesis , Insulin-Like Growth Factor I/metabolism , Mammary Glands, Animal , Mammary Glands, Human/metabolism , Mammary Neoplasms, Animal , Mice , Mice, Transgenic , Milk Proteins/chemistry , Models, Genetic , Nuclear Receptor Coactivator 3 , Promoter Regions, Genetic , Protein Isoforms , Receptor, IGF Type 1/metabolism , Reverse Transcriptase Polymerase Chain Reaction , STAT5 Transcription Factor , Signal Transduction , Trans-Activators/chemistry , Transcription Factors/biosynthesis , Transgenes , Tumor Suppressor Proteins
6.
Cancer Res ; 63(7): 1696-705, 2003 Apr 01.
Article in English | MEDLINE | ID: mdl-12670924

ABSTRACT

The fibroblast growth factor-binding protein (FGF-BP) binds and activates fibroblast growth factors in the extracellular matrix, and can have a rate-limiting role in tumor angiogenesis. Here we demonstrate high levels of FGF-BP expression in invasive human breast cancer, relative to normal breast and in situ carcinoma, and in MDA-MB-468 human breast cancer cells. In these cells, FGF-BP was up-regulated by treatment with epidermal growth factor (EGF), dependent on protein kinase C and p38 mitogen-activated protein kinase signaling. Mutational analysis revealed that the activator protein 1 and CCAAT/enhancer binding protein (C/EBP) sites on the FGF-BP gene promoter were required for the EGF effect, whereas deletion of the C/EBP site resulted in a significant increase in promoter basal activity indicating a basal repressive control mechanism. These data suggest that the C/EBP site is a central regulatory element for the regulation of FGF-BP promoter activity in MDA-MB-468 cells. We found that MDA-MB-468 cells express high endogenous levels of both the activating (LAP) and repressive (LIP) isoforms of C/EBPbeta. Overexpression of C/EBPbeta-LAP in MDA-MB-468 cells resulted in a large 80-fold increase in FGF-BP promoter basal activity, which was reversed by coexpression of LIP. Gel-shift analysis revealed that four LIP- and LAP-containing complexes (a-d) bind to the C/EBP site. DNA binding of the LIP and LAP-containing c complex and the b complex in the presence of EGF was modulated by inhibition of p38 mitogen-activated protein kinase, suggesting a role for these complexes in the EGF induction of the FGF-BP promoter. This study suggests that along with its well-defined role in mammary gland development, C/EBPbeta may well play a role in the pathology of breast cancer, in particular in the control of angiogenesis in the invasive phenotype.


Subject(s)
Breast Neoplasms/metabolism , CCAAT-Enhancer-Binding Protein-beta/physiology , Carrier Proteins/biosynthesis , Binding Sites , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carrier Proteins/genetics , Epidermal Growth Factor/pharmacology , Epidermal Growth Factor/physiology , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/physiology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinases/metabolism , Neoplasm Invasiveness , Promoter Regions, Genetic , Protein Kinase C/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Transcription Factor AP-1/metabolism , Transfection , Tumor Cells, Cultured , p38 Mitogen-Activated Protein Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...