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.
Tumori ; 102(2): 144-9, 2016.
Article in English | MEDLINE | ID: mdl-26917410

ABSTRACT

PURPOSE: Because the peptidyl-prolyl isomerase PIN1 interacts with multiple protein kinases and phosphoproteins into a network orchestrating the cellular response to various stimuli, there is an increasing interest in exploiting its potential as therapeutic target. In the present study, the effect of targeting PIN1 was investigated in 2 human cancer cell lines characterized by increased aggressive potential, high expression of erbB receptor family members, and defective p53. METHODS: PIN1 silencing was carried out in skin squamous cell carcinoma A431 cells displaying elevated EGFR/HER1 levels and in ovarian adenocarcinoma SKOV-3 cells displaying high levels of erbB2 (HER2). Nonoverlapping siRNA duplexes targeting different regions of PIN1 mRNA were transfected in tumor cells, which were analyzed using Western blotting for the expression of selected proteins. In vivo tumorigenicity studies were carried out in athymic nude mice. RESULTS: A431 and SKOV-3 cell systems were found to be a source of cells with increased aggressive potential, i.e., cancer stem cell-like cells, as defined by the capability to grow as spheres. A marked decrease of PIN1 levels and of sphere-forming capability was observed in PIN1-silenced cells. The expression of phospho-p38 decreased following PIN1 silencing in A431 and SKOV-3 cells, as well as phospho-EGFR levels in A431 - silenced cells. PIN1 inhibition prolonged latency and reduced tumor take and growth of SKOV-3 cells in nude mice. CONCLUSIONS: Our results support that PIN1 may be a valuable target to hit in cancer cells characterized by increased aggressive potential, overexpression of erbB receptor family members, and defective p53.


Subject(s)
ErbB Receptors/metabolism , Gene Silencing , Neoplasms/metabolism , Peptidylprolyl Isomerase/genetics , Peptidylprolyl Isomerase/metabolism , Tumor Suppressor Protein p53/metabolism , Adenocarcinoma/metabolism , Animals , Blotting, Western , Carcinogenicity Tests , Carcinoma, Squamous Cell/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Nude , NIMA-Interacting Peptidylprolyl Isomerase , Ovarian Neoplasms/metabolism , Skin Neoplasms/metabolism , Up-Regulation
2.
J Cell Physiol ; 228(4): 871-8, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23018838

ABSTRACT

Many squamous cell carcinomas (SCCs) are characterized by high levels of EGFR and by overexpression of the ΔNp63α isoform. Here, we investigated the regulation of ΔNp63α expression upon EGFR activation and the role of the EGFR-ΔNp63α axis in proliferation of SCC tumor-initiating cells (TICs). SCC cell lines A-431, Cal-27, and SCC-25 treated with EGF showed a time-dependent increase in ΔNp63α expression at the protein and mRNA levels, which was blocked by the tyrosine kinase inhibitor (TKI) Lapatinib. RNA interference experiments suggested the role of STAT3 in regulating ΔNp63α expression downstream of EGFR. Inactivation of EGFR by the monoclonal antibody Cetuximab and RNA interference against STAT3 or ΔNp63α impaired the TICs ability to grow under non-differentiating conditions. Radiation treatment, which triggers EGFR activation, induced ΔNp63α accumulation without affecting TICs proliferation, whereas the combination Cetuximab plus radiation significantly reduced TICs growth under non-differentiating conditions. Together, our findings provide evidence that ΔNp63α expression is regulated by EGFR activation through STAT3 and that the EGFR-ΔNp63α axis is crucial for proliferation of TICs present in SCCs.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , ErbB Receptors/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , STAT3 Transcription Factor/metabolism , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Carcinoma, Squamous Cell/genetics , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cetuximab , ErbB Receptors/genetics , Humans , Lapatinib , MCF-7 Cells , Neoplastic Stem Cells/drug effects , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , RNA, Messenger/genetics , STAT3 Transcription Factor/genetics
3.
Cancer Lett ; 296(2): 206-15, 2010 Oct 28.
Article in English | MEDLINE | ID: mdl-20435407

ABSTRACT

There is indication that tumor growth is sustained by subpopulation of cells with stem-like features but little is known on their genomic characterization and their genetic stability. We report a detailed molecular cytogenetic characterization using Spectral Karyotyping and fluorescent in situ hybridization of parental serum-cultured adherent cells and their sphere-growing stem-like counterpart before and after differentiation from six cell lines established from solid tumors. Our findings indicate increased cytogenetic complexity in sphere-growing stem-like and their differentiated adherent cells compared to parental adherent component suggesting the existence within cell lines of heterogeneous and genetically unstable subpopulations of cells endowed with stem-like features.


Subject(s)
Cell Line, Tumor/pathology , Stem Cells/pathology , Antigens, CD/analysis , Biomarkers, Tumor/analysis , Cell Adhesion , Cell Culture Techniques/methods , Cell Differentiation , Cell Division , Chromosome Aberrations , Flow Cytometry , Gene Expression Regulation, Neoplastic , Humans , In Situ Hybridization, Fluorescence , Karyotyping/methods , Neoplasms/genetics
4.
Clin Cancer Res ; 15(6): 2010-21, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19276287

ABSTRACT

PURPOSE: The existence of tumor-initiating cells in breast cancer has profound implications for cancer therapy. In this study, we investigated the sensitivity of tumor-initiating cells isolated from human epidermal growth factor receptor type 2 (HER2)-overexpressing carcinoma cell lines to trastuzumab, a compound used for the targeted therapy of breast cancer. EXPERIMENTAL DESIGN: Spheres were analyzed by indirect immunofluorescence for HER2 cell surface expression and by real-time PCR for HER2 mRNA expression in the presence or absence of the Notch1 signaling inhibitor (GSI) or Notch1 small interfering RNA. Xenografts of HER2-overexpressing breast tumor cells were treated with trastuzumab or doxorubicin. The sphere-forming efficiency (SFE) and serial transplantability of tumors were assessed. RESULTS: In HER2-overexpressing carcinoma cell lines, cells with tumor-initiating cell properties presented increased HER2 levels compared with the bulk cell population without modification in HER2 gene amplification. HER2 levels were controlled by Notch1 signaling, as shown by the reduction of HER2 cell surface expression and lower SFE following gamma-secretase inhibition or Notch1 specific silencing. We also show that trastuzumab was able to effectively target tumor-initiating cells of HER2-positive carcinoma cell lines, as indicated by the significant decrease in SFE and the loss of serial transplantability, following treatment of HER2-overexpressing xenotransplants. CONCLUSIONS: Here, we provide evidence for the therapeutic efficacy of trastuzumab in debulking and in targeting tumor-initiating cells of HER2-overexpressing tumors. We also propose that Notch signaling regulates HER2 expression, thereby representing a critical survival pathway of tumor-initiating cells.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/analysis , Animals , Antibodies, Monoclonal, Humanized , Cell Line, Tumor , Humans , Lapatinib , Mice , Quinazolines/pharmacology , RNA, Messenger/analysis , Receptor, ErbB-2/genetics , Receptor, Notch1/physiology , Trastuzumab , Xenograft Model Antitumor Assays
5.
Cancer Res ; 67(11): 5308-17, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17545611

ABSTRACT

In some HER2-positive breast tumors, cell surface overexpression of HER2 is not associated with gene amplification but may instead rest in altered gene transcription, half-life, or recycling of the oncoprotein. Here, we show that HER2 overexpression in HER2 2+ carcinomas is associated with neither an increase in gene transcription nor a deregulation in the ubiquitin-dependent pathways, but instead seems to be regulated by protein kinase Calpha (PKCalpha) activity. The stimulation of PKCalpha up-regulated HER2 expression, whereas PKCalpha inhibition by pharmacologic treatments and PKCalpha-specific small interfering RNA led to a dramatic down-regulation of HER2 levels only in breast cancer cells HER2 2+. Consistent with the in vitro data, our biochemical analysis of HER2 2+ human primary breast specimens revealed significantly higher levels of phosphorylated PKCalpha compared with HER2-negative tumors. Inhibition of HER2 activation by the tyrosine kinase inhibitor lapatinib led to decreased levels of PKCalpha phosphorylation, clearly indicating a cross-talk between PKCalpha and HER2 molecules. These data suggest that HER2 overexpression in HER2 2+ carcinomas is due to an accumulation of the recycled oncoprotein to the cell surface induced by activated PKCalpha.


Subject(s)
Breast Neoplasms/enzymology , Protein Kinase C-alpha/metabolism , Receptor, ErbB-2/biosynthesis , Breast Neoplasms/genetics , Carbazoles/pharmacology , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , Epidermal Growth Factor/pharmacology , Gene Amplification , Humans , Immunoprecipitation , Indoles/pharmacology , Protein Kinase C-alpha/antagonists & inhibitors , Protein Kinase C-alpha/biosynthesis , Protein Kinase C-alpha/genetics , Proto-Oncogene Proteins c-cbl/biosynthesis , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Receptor, ErbB-2/genetics , Transfection
6.
Proc Natl Acad Sci U S A ; 103(50): 18981-6, 2006 Dec 12.
Article in English | MEDLINE | ID: mdl-17142325

ABSTRACT

The tumor suppressor gene FHIT is inactivated by genetic and epigenetic changes in the majority of common human cancers. The human Fhit protein undergoes phosphorylation on tyrosine residue 114 by Src and related kinases both in vitro and in vivo. Src is a key cytoplasmic tyrosine kinase downstream to several growth factor receptors, including those of the EGF receptor family, which are overexpressed and activated in about one-third of human breast and ovarian carcinomas. However, the biological significance of Fhit phosphorylation by Src has remained elusive. In the present study, we demonstrate that FHIT acts as a checkpoint in cell proliferation mediated by activated tyrosine kinase receptors that recruit Src. Activation of EGF receptor family members induced Fhit phosphorylation by Src and the subsequent proteasome degradation of the phosphorylated Fhit protein. Indeed, the use of the Fhit mutant Y114F, which carries a phenylalanine instead of a tyrosine at position 114, unable to be phosphorylated on tyrosine 114 by Src, prevents Fhit degradation. Moreover, Fhit protein reduction is transient and occurs in a specific temporal window. During the signaling pathway of activated tyrosine kinase receptors, the phosphorylation of Fhit induces its degradation and the subsequent reduction in Fhit protein levels allows the transmission of the mitogenic signal; immediately thereafter, Fhit protein levels are restored. Such a scenario would suggest a key role for Fhit in the balance of proliferation/survival/apoptosis signals.


Subject(s)
Acid Anhydride Hydrolases/metabolism , ErbB Receptors/metabolism , Mitosis , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Proteasome Endopeptidase Complex/metabolism , Acid Anhydride Hydrolases/genetics , Cell Line , Enzyme Activation , Epidermal Growth Factor/metabolism , ErbB Receptors/classification , Humans , Mitogens/pharmacology , Mitosis/drug effects , Models, Biological , Neoplasm Proteins/genetics , Neoplasms/genetics , Phosphorylation , Proto-Oncogene Proteins pp60(c-src)/metabolism , Signal Transduction
7.
J Biol Chem ; 278(44): 43169-77, 2003 Oct 31.
Article in English | MEDLINE | ID: mdl-12907674

ABSTRACT

Cbl proteins have RING finger-dependent ubiquitin ligase (E3) activity that is essential for down-regulation of tyrosine kinases. Here we establish that two WW domain HECT E3s, Nedd4 and Itch, bind Cbl proteins and target them for proteasomal degradation. This is dependent on the E3 activity of the HECT E3s but not on that of Cbl. Consistent with these observations, in cells expressing the epidermal growth factor receptor, Nedd4 reverses Cbl-b effects on receptor down-regulation, ubiquitylation, and proximal events in signaling. Cbl-b also targets active Src for degradation in cells, and Nedd4 similarly reverses Cbl-mediated Src degradation. These findings establish that RING finger E3s can be substrates, not only for autoubiquitylation but also for ubiquitylation by HECT E3s and suggest an additional level of regulation for Cbl substrates including protein-tyrosine kinases.


Subject(s)
Cysteine Endopeptidases/metabolism , Multienzyme Complexes/metabolism , Proto-Oncogene Proteins/metabolism , Cell Line , Down-Regulation , Endosomal Sorting Complexes Required for Transport , Escherichia coli/metabolism , Glutathione Transferase/metabolism , Humans , Immunoblotting , Jurkat Cells , Nedd4 Ubiquitin Protein Ligases , Plasmids/metabolism , Precipitin Tests , Proteasome Endopeptidase Complex , Protein Binding , Protein Structure, Tertiary , Proto-Oncogene Proteins c-cbl , Recombinant Fusion Proteins/metabolism , Repressor Proteins/metabolism , Time Factors , Transfection , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...