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.
Breast Cancer Res ; 13(3): R65, 2011 Jun 20.
Article in English | MEDLINE | ID: mdl-21689417

ABSTRACT

INTRODUCTION: The c-Jun coactivator, Jun activation-domain binding protein 1 (Jab1) also known as the fifth component of the COP9 signalosome complex (CSN5), is a novel candidate oncogene whose aberrant expression contributes to the progression of breast carcinoma and other human cancers. The mechanism of Jab1 gene expression and its deregulation in cancer cells remains to be identified. We therefore investigated the transcriptional regulatory mechanisms of Jab1 expression in human breast carcinoma cells. METHODS: To identify potential regulators of Jab1 transcription, we cloned the 5' upstream region of the human Jab1 gene and mapped its transcriptional start site. We identified binding sequences for the CCAAT/enhancer binding protein (C/EBP) and GATA, as well as a signal transducer and activator of transcription-3 (Stat3) consensus sequence overlapping the C/EBP site, using 5'- deletion analysis and a gene reporter assay. Mutational analysis of these binding sites was performed to confirm their roles in promoting Jab1 transcription in breast cancer cells. We further confirmed these binding sites using electrophoretic mobility shift assays (EMSAs) and chromatin immunoprecipitation (ChIP) assays. We also analyzed whether the siRNA-mediated inactivation of Stat3 and Src could reduce Jab1-promoter activity and whether interleukine-6 (IL-6) could mediate increased Jab1 expression through Stat3 signaling. RESULTS: We identified binding sequences for C/EBP, GATA, as well as a Stat3 consensus sequence overlapping the C/EBP site in the promoter region of Jab1. C/EBP-beta2 is a potential transcriptional activator of Jab1 and mutation of the C/EBP/Stat3 binding site significantly reduced Jab1-promoter activity. In addition, inhibiting Stat3 significantly reduced Jab1-promoter activation. EMSA and ChIP assays confirmed that C/EBP, GATA1 and Stat3 bind to Jab1 promoter in breast carcinoma cells. We also found that Src, an activator of Stat3, is involved in Jab1-promoter activation. siRNA knockdown of Src reduced the Jab1-promoter activity, similar to the results seen when Stat3 was inhibited in breast carcinoma cells. Interestingly, reactivation of Stat3 in normal mammary epithelial cells (MCF-10A, MCF-10F) is sufficient to reactivate Jab1 expression. Treatment with the cytokine IL-6 resulted in increased Jab1 expression that was blocked by inhibition of Stat3. CONCLUSIONS: These findings reveal a novel mechanism of Jab1 gene regulation and provide functional and mechanistic links between the Src/Stat3 and IL-6/Stat3 signaling axes that are involved in the activation of Jab1 transcription and regulation of this novel oncogenic protein.


Subject(s)
Breast Neoplasms/metabolism , CCAAT-Enhancer-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , Intracellular Signaling Peptides and Proteins/genetics , Peptide Hydrolases/genetics , Promoter Regions, Genetic , STAT3 Transcription Factor/metabolism , Base Sequence , Binding Sites , CCAAT-Enhancer-Binding Protein-beta , COP9 Signalosome Complex , Cell Line, Tumor , Chromatin Immunoprecipitation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Electrophoretic Mobility Shift Assay , Female , GATA1 Transcription Factor/metabolism , Humans , Interleukin-6/pharmacology , Intracellular Signaling Peptides and Proteins/metabolism , Peptide Hydrolases/metabolism , RNA Interference , RNA, Small Interfering , Regulatory Sequences, Nucleic Acid , STAT3 Transcription Factor/antagonists & inhibitors , Sequence Analysis, DNA , Signal Transduction/genetics , Transcription, Genetic , Transcriptional Activation , src-Family Kinases/genetics , src-Family Kinases/metabolism
2.
Cancer Res ; 66(17): 8581-9, 2006 Sep 01.
Article in English | MEDLINE | ID: mdl-16951171

ABSTRACT

Reduced expression of p27 has been associated with poor prognosis in most human cancers, including pancreatic adenocarcinoma. Jun activation domain-binding protein 1 (JAB1), an activator protein (AP-1) coactivator, previously implicated in p27 degradation, is overexpressed in various tumors and correlates with low p27 expression. We examined JAB1 and p27 in normal and neoplastic pancreatic tissues. Increased JAB1 expression was seen in pancreatic carcinoma samples but not in paired normal pancreatic tissues. Immunohistochemical analysis using tissue microarrays showed that JAB1 was overexpressed in all 32 (100%) pancreatic adenocarcinoma samples tested, predominantly nuclear in 23 (72%) samples and predominantly cytoplasmic in 9 (28%) tumors. When 10% was used as a cutoff for p27 positivity, p27 was expressed in 11 (34%) of tumors; however, p27 expression was localized in the nuclei of tumor cells in only 4 (13%) of the samples. Overexpression of the JAB1 in the pancreatic carcinoma cell lines Panc-1, Mia PaCa-2, and Panc-28 resulted in decreased p27 expression. Conversely, down-regulation of JAB1 by short interfering RNA substantially increased p27 expression and inhibited progression from G(1) to S phase of the cell cycle. Interestingly, JAB1-mediated p27 degradation was not impaired when S-phase kinase-interacting protein 2 (Skp2), an F-box protein required for the ubiquitination and consequent degradation of p27, was silenced. Thus, JAB1 may have an Skp2-independent p27 degradation mechanism in pancreatic cancer cells. These findings suggest that JAB1 overexpression is involved in the pathogenesis of pancreatic cancer through JAB1-mediated p27 degradation and that control of JAB1 expression is a novel therapeutic target in patients with pancreatic adenocarcinomas.


Subject(s)
Adenocarcinoma/physiopathology , Cyclin-Dependent Kinase Inhibitor p27/genetics , Intracellular Signaling Peptides and Proteins/genetics , Pancreatic Neoplasms/physiopathology , Peptide Hydrolases/genetics , Adenocarcinoma/pathology , Adenocarcinoma/surgery , Adult , Aged , COP9 Signalosome Complex , Combined Modality Therapy , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Female , Homeostasis , Humans , Immunohistochemistry , Intracellular Signaling Peptides and Proteins/metabolism , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Pancreas/pathology , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/surgery , Peptide Hydrolases/metabolism
3.
FEBS Lett ; 579(18): 3932-40, 2005 Jul 18.
Article in English | MEDLINE | ID: mdl-15996662

ABSTRACT

The cyclin-dependent kinase (CDK) inhibitor p27(Kip1) (p27) is an important regulator of cell cycle progression controlling the transition from G to S-phase. Low p27 levels or accelerated p27 degradation correlate with excessive cell proliferation and poor prognosis in several forms of cancer. Phosphorylation of p27 at Thr187 by cyclin E-CDK2 is required to initiate the ubiquitination-proteasomal degradation of p27. Protecting p27 from ubiquitin-mediated proteasomal degradation may increase its potential in cancer gene therapy. Here we constructed a non-phosphorylatable, proteolysis-resistant p27 mutant containing a Thr187-to-Ala substitution (T187A) which is not degraded by ubiquitin-mediated proteasome pathway, and compared its effects on cell growth, cell-cycle control, and apoptosis with those of wild-type p27. In muristerone A-inducible cell lines overexpressing wild-type or mutant p27, the p27 mutant was more resistant to proteolysis in vivo and more potent in inducing cell-cycle arrest and other growth-inhibitory effects such as apoptosis. Transduction of p27(T187A) in breast cancer cells with a doxycycline-regulated adenovirus led to greater inhibition of proliferation, more extensive apoptosis, with a markedly reduced protein levels of cyclin E and increased accumulation of cyclin D1, compared with wild-type p27. These findings support the potential effectiveness of a degradation-resistant form of p27 in breast cancer gene therapy.


Subject(s)
Apoptosis , Breast Neoplasms/pathology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/physiology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/physiology , Adenoviridae/genetics , Alanine/chemistry , Annexin A5/chemistry , Biomarkers, Tumor , Breast Neoplasms/metabolism , CDC2-CDC28 Kinases/metabolism , Cell Cycle , Cell Cycle Proteins/chemistry , Cell Line , Cell Line, Tumor , Cell Proliferation , Cyclin D1/metabolism , Cyclin E/chemistry , Cyclin E/metabolism , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase Inhibitor p27 , Doxycycline/pharmacology , Ecdysterone/analogs & derivatives , Ecdysterone/pharmacology , Humans , Immunoblotting , Mutagenesis, Site-Directed , Mutation , Phosphorylation , Prognosis , Proteasome Endopeptidase Complex/metabolism , Proteins/chemistry , S Phase , Threonine/chemistry , Time Factors , Tumor Suppressor Proteins/chemistry , Ubiquitin/chemistry , Ubiquitin/metabolism
4.
J Biol Chem ; 278(21): 19245-56, 2003 May 23.
Article in English | MEDLINE | ID: mdl-12637505

ABSTRACT

The efficacy of cisplatin in cancer chemotherapy is limited by the development of resistance. Although the molecular mechanisms involved in chemoresistance are poorly understood, cellular response to cisplatin is known to involve activation of MAPK and other signal transduction pathways. An understanding of early signal transduction events in the response to cisplatin could be valuable for improving the efficacy of cancer therapy. We compared cisplatin-induced activation of three MAPKs, JNK, p38, and ERK, in a cisplatin-sensitive human ovarian carcinoma cell line (2008) and its resistant subclone (2008C13). The JNK and p38 pathways were activated differentially in response to cisplatin, with the cisplatin-sensitive cells showing prolonged activation (8-12 h) and the cisplatin-resistant cells showing only transient activation (1-3 h) of JNK and p38. In the sensitive cells, inhibition of cisplatin-induced JNK and p38 activation blocked cisplatin-induced apoptosis; persistent activation of JNK resulted in hyperphosphorylation of the c-Jun transcription factor, which in turn stimulated the transcription of an immediate downstream target, the death inducer Fas ligand (FasL). Sequestration of FasL by incubation with a neutralizing anti-FasL antibody inhibited cisplatin-induced apoptosis. In contrast, chemoresistance in 2008C13 cells was associated with failure to up-regulate FasL. Moreover, in these cells, selective stimulation of the JNK/p38 MAPK pathways by adenovirus-mediated delivery of recombinant MKK7 or MKK3 led to sensitization to apoptosis through reactivating FasL expression. Thus, the JNK > c-Jun > FasL > Fas pathway plays an important role in mediating cisplatin-induced apoptosis in ovarian cancer cells, and the duration of JNK activation is critical in determining whether cells survive or undergo apoptosis.


Subject(s)
Apoptosis/drug effects , Cisplatin/pharmacology , JNK Mitogen-Activated Protein Kinases , Membrane Glycoproteins/biosynthesis , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Ovarian Neoplasms/pathology , Cell Survival , Drug Resistance, Neoplasm , Enzyme Activation/drug effects , Fas Ligand Protein , Female , Gene Expression , Humans , MAP Kinase Kinase 3 , MAP Kinase Kinase 4 , MAP Kinase Kinase 7 , Membrane Glycoproteins/genetics , Mitogen-Activated Protein Kinase Kinases/genetics , Ovarian Neoplasms/metabolism , Phosphorylation , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins c-jun/metabolism , Recombinant Proteins , Transfection , Tumor Cells, Cultured , p38 Mitogen-Activated Protein Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...