Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Carcinog ; 48(6): 532-44, 2009 Jun.
Article in English | MEDLINE | ID: mdl-18973190

ABSTRACT

Transforming growth factor beta (TGF-beta) exerts an important role in the late steps of carcinogenesis by cooperating with Ras to induce cell motility and tumor invasion. The transcription complex AP-1 has been implicated in the regulation of genes involved in motility and invasion, by mechanisms not yet delineated. We utilized a model of immortalized human hepatocytes (IHH) overexpressing c-Fos (IHH-Fos) or not (IHH-C) to investigate the role of c-Fos on cell motility in response to a prolonged treatment with TGF-beta, EGF or a combination of both. Cotreatment with EGF and TGF-beta, but neither cytokine alone, induced the conversion of hepatocytes to a fibroblastoid phenotype and increased their motility in Boyden chambers. EGF/TGF-beta cotreatment induced a higher effect on ERK phosphorylation compared to TGF-beta treatment alone. It also induced an increase in total and phosphorylated Ser(178) paxillin, a protein previously implicated in cell motility. This response was inhibited by two specific MEK inhibitors, indicating the involvement of the ERK pathway in paxillin activation. Overexpression of c-Fos correlated with increased cell scattering and motility, higher levels of ERK activation and phospho Ser(178) paxillin, increased levels of EGF receptor (EGF-R) mRNA and higher EGF-R phosphorylation levels following EGF/TGF-beta cotreatment. Conversely, siRNA-mediated invalidation of c-Fos delayed the appearance of fibroblastoid cells, decreased EGF-R mRNA and downregulated ERK and Ser(178) paxillin phosphorylations, indicating that c-Fos activates hepatocyte motility through an EGF-R/ERK/paxillin pathway. Since c-Fos is frequently overexpressed in hepatocarcinomas, this newly identified mechanism might be involved in the progression of hepatic tumors in vivo.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Hepatocytes/cytology , Paxillin/metabolism , Proto-Oncogene Proteins c-fos/physiology , Serine/metabolism , Up-Regulation , Base Sequence , Cell Line, Transformed , DNA Primers , Enzyme Activation , Epidermal Growth Factor/pharmacology , Fibroblasts/cytology , Humans , Microscopy, Confocal , Microscopy, Fluorescence , Paxillin/chemistry , Phosphorylation , Polymerase Chain Reaction , Signal Transduction/drug effects , Transforming Growth Factor beta/pharmacology
2.
World J Gastroenterol ; 14(41): 6339-46, 2008 Nov 07.
Article in English | MEDLINE | ID: mdl-19009649

ABSTRACT

AIM: To investigate the effect of stable c-Fos overexpression on immortalized human hepatocyte (IHH) proliferation. METHODS: IHHs stably transfected with c-Fos (IHH-Fos) or an empty vector (IHH-C) were grown in medium supplemented with 1% serum or stimulated with 10% serum. Cell proliferation was assessed by cell counts, 3H-thymidine uptake and flow cytometry analyses. The levels of cell cycle regulatory proteins (Cyclin D1, E, A) cyclin dependent kinases (cdk) cdk2, cdk4, cdk6, and their inhibitors p15, p16, p21, p27, total and phosphorylated GSK-3beta and epidermal growth factor receptor (EGF-R) were assayed by Western blotting. Analysis of Cyclin D1 mRNA levels was performed by reverse transcription-polymerase chain reaction and real-time polymerase chain reaction (PCR) analysis. Stability of Cyclin D1 was studied by cycloheximide blockade experiments. RESULTS: Stable c-Fos overexpression increased cell proliferation under low serum conditions and resulted in a two-fold increase in [3H]-thymidine incorporation following serum addition. Cell cycle analysis by flow cytometry showed that c-Fos accelerated the cell cycle kinetics. Following serum stimulation, Cyclin D1 was more abundantly expressed in c-Fos overexpressing cells. Cyclin D1 accumulation did not result from increased transcriptional activation, but from nuclear stabilization. Overexpression of c-Fos correlated with higher nuclear levels of inactive phosphorylated GSK-3beta, a kinase involved in Cyclin D1 degradation and higher levels of EGF-R mRNA, and EGF-R protein compared to IHH-C both in serum starved, and in serum stimulated cells. Abrogation of EGF-R signalling in IHH-Fos by treatment with AG1478, a specific EGF-R tyrosine kinase inhibitor, prevented the phosphorylation of GSK-3beta induced by serum stimulation and decreased Cyclin D1 stability in the nucleus. CONCLUSION: Our results clearly indicate a positive role for c-Fos in cell cycle regulation in hepatocytes. Importantly, we delineate a new mechanism by which c-Fos could contribute to hepatocarcinogenesis through stabilization of Cyclin D1 within the nucleus, evoking a new feature to c-Fos implication in hepatocellular carcinoma.


Subject(s)
Cell Nucleus/metabolism , Cell Proliferation , Cyclin D1/metabolism , Hepatocytes/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Cell Cycle , Cell Nucleus/drug effects , Cell Nucleus/enzymology , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/metabolism , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor Proteins/metabolism , Cyclin-Dependent Kinases/metabolism , Cycloheximide/pharmacology , ErbB Receptors/metabolism , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Hepatocytes/drug effects , Hepatocytes/enzymology , Humans , Kinetics , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Protein Stability , Protein Synthesis Inhibitors/pharmacology , Proto-Oncogene Proteins c-fos/genetics , Quinazolines , RNA, Messenger/metabolism , Transfection , Tyrphostins/pharmacology , Up-Regulation
3.
Carcinogenesis ; 29(3): 536-43, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18174238

ABSTRACT

Nuclear factor kappaB (NF-kappaB) and activator protein 1 are transcription factors involved in the regulation of cell proliferation that play important roles in tumorigenesis. We investigated whether these two factors cooperate for transcriptional regulation of cyclin D1 (CCND1), a gene whose deregulation is critical during carcinogenesis. We demonstrate that overexpression of JunD in human hepatocarcinoma cells strongly activates transcription mediated by the kappaB2 site of the CCND1 promoter in reporter assays, in a manner strictly dependent on the presence of NF-kappaB proteins. Serum stimulation increased the expression of p65, p50, c-Fos, c-Jun and JunD and induced the recruitment of p65, p50 and JunD to the kappaB2 site of the promoter in DNA pull-down assays. Chromatin immunoprecipitation (ChIP) analysis confirmed the serum-induced recruitment of JunD to the promoter in vivo and showed that the presence of JunD was dependent on the presence of p65 and p50, indicating a protein-protein-dependent mechanism of JunD recruitment. Serum-induced activation of protein binding to kappaB2 correlated with high levels of phosphoinositide-dependent protein kinase-1 (PDK-1) phosphorylation. Both LY294002, a specific inhibitor of phosphatidylinositol 3-kinase (PI3K), and overexpression of a dominant-negative form of PDK-1 inhibited the JunD-stimulating effect in reporter assays. LY294002 also prevented the serum-induced recruitment of JunD, but not p65 or p50 to the promoter in ChIP assay. JunD-p65 complexes, identified in vivo by co-immunoprecipitation, were decreased by LY294002 and by small interfering RNA inhibition of PDK-1. Taken together, our data demonstrate a PI3K/PDK-1-dependent functional cooperation of NF-kappaB and JunD in the transcriptional regulation of CCND1 by serum.


Subject(s)
Cyclin D1/genetics , Phosphatidylinositol 3-Kinases/physiology , Promoter Regions, Genetic , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins c-jun/physiology , Transcription Factor RelA/physiology , Base Sequence , Binding Sites , Blood , Chromatin Immunoprecipitation , Chromones/pharmacology , Cyclin D1/chemistry , Cyclin D1/metabolism , DNA Primers , Electrophoretic Mobility Shift Assay , Enzyme Inhibitors/pharmacology , Humans , Immunoprecipitation , Morpholines/pharmacology , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Transcription Factor RelA/metabolism
4.
J Hepatol ; 46(6): 1075-88, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17399843

ABSTRACT

BACKGROUND/AIMS: Endoplasmic reticulum (ER)-related unfolded protein response (UPR) is mediated by PKR-like ER kinase (PERK), ATF6 and IRE1. PERK phosphorylates eukaryotic translation initiation factor-2alpha (eIF2alpha) to attenuate protein synthesis, including in NF-kappaB-dependent antiapoptotic proteins. We hypothesized that an altered UPR in the liver may sensitize cirrhotic livers to LPS-induced, TNFalpha-mediated apoptosis. Thus, we examined in vivo UPR and NF-kappaB activity in livers from cirrhotic and normal LPS-challenged rats. METHODS: Livers were harvested in rats that did or did not receive LPS. RESULTS: Under baseline conditions, no UPR was found in normal livers while PERK/eIF2alpha and ATF6 pathways were activated in cirrhotic livers. After LPS, in normal livers, the PERK/eIF2alpha pathway was transiently activated. ATF6 and IRE1 were activated. In cirrhotic livers, the PERK/eIF2alpha pathway remained elevated. ATF6 and IRE1 pathways were altered. LPS-induced, NF-kappaB-dependent antiapoptotic proteins increased in normal livers whereas their expression was blunted at the posttranscriptional level in cirrhotic livers. CONCLUSIONS: Cirrhotic livers exhibit partial UPR activation in the basal state and full UPR, although altered, after LPS challenge. Sustained eIF2alpha phosphorylation, a hallmark of cirrhotic liver UPR, is associated with a lack of LPS-induced accumulation of NF-kappaB-dependent antiapoptotic proteins which may sensitize cirrhotic livers to LPS/TNFalpha-mediated apoptosis.


Subject(s)
Apoptosis , Fibrosis/pathology , Lipopolysaccharides/metabolism , Liver/pathology , Animals , Caspase 3/metabolism , Endoplasmic Reticulum/metabolism , Eukaryotic Initiation Factor-2/metabolism , Liver/metabolism , Male , Protein Denaturation , Protein Folding , Rats , Rats, Sprague-Dawley , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
5.
World J Gastroenterol ; 12(18): 2895-900, 2006 May 14.
Article in English | MEDLINE | ID: mdl-16718815

ABSTRACT

AIM: To investigate the role of Beclin 1 on the susceptibility of HepG2 cells to undergo apoptosis after anti-Fas antibody or doxorubicin treatment. METHODS: Beclin 1 silencing was achieved using RNA interference. DNA ploidy, the percentage of apoptotic cells and the mitochondrial membrane potential were assessed by flow cytometry. Levels of Beclin 1, Bcl-X(L) and cytochrome c, and the cleavage of poly (ADP-ribose) polymerase (PARP) were assayed by using Western blots. RESULTS: Beclin 1 expression decreased by 75% 72 h after Beclin 1 siRNA transfection. Partial Beclin 1 silencing significantly increased the percentage of subG1 cells 24 and 40 h after treatment with doxorubicin or anti-Fas antibody, respectively, and this potentiation was abrogated by treatment with a pan-caspase inhibitor. Partial Beclin 1 silencing also increased PARP cleavage, mitochondrial membrane depolarization and cytosolic cytochrome c. The pro-apoptotic consequences of partial Beclin 1 silencing were not associated with a decline in Bcl-X(L) expression. CONCLUSION: Partial Beclin 1 silencing aggravates mitochondrial permeabilization and apoptosis in HepG2 cells treated with an anti-Fas antibody or with doxorubicin.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Antibodies, Monoclonal/pharmacology , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/physiology , Apoptosis/drug effects , Carcinoma, Hepatocellular/pathology , Doxorubicin/pharmacology , Gene Silencing/physiology , Liver Neoplasms/pathology , Membrane Proteins/genetics , Membrane Proteins/physiology , Antibodies, Monoclonal, Murine-Derived , Apoptosis Regulatory Proteins/analysis , Beclin-1 , Blotting, Western , Carcinoma, Hepatocellular/chemistry , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Cytochromes c/analysis , DNA, Neoplasm/analysis , DNA, Neoplasm/genetics , Flow Cytometry , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/chemistry , Liver Neoplasms/genetics , Membrane Proteins/analysis , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/physiology , Permeability , RNA Interference , RNA, Messenger/analysis , RNA, Messenger/genetics , RNA, Small Interfering , Transfection , bcl-X Protein/analysis
6.
Am J Respir Cell Mol Biol ; 32(5): 470-7, 2005 May.
Article in English | MEDLINE | ID: mdl-15677771

ABSTRACT

Keratinocyte growth factor (KGF) is secreted by fibroblasts and protects from pulmonary fibrosis in animal models. Interleukin (IL)-1beta is the most potent inducer of KGF in fibroblasts, acting through the c-Jun pathway. We evaluated in vitro KGF production by human lung fibroblasts from patients with idiopathic pulmonary fibrosis (IPF, n = 10) and from control subjects (n = 7) at baseline and after IL-1beta stimulation. Basal KGF secretion by IPF fibroblasts was similar to controls. In fibroblasts from control subjects, IL-1beta increased c-Jun expression, c-Jun activation, and KGF secretion. SP600125, a specific c-Jun N-terminal kinase (JNK) inhibitor, inhibited the effect of IL-1beta. By contrast, in IPF fibroblasts, IL-1beta did not increase c-Jun expression and c-Jun activation, and weakly increased KGF secretion, whereas SP600125 had no effect. IL-1beta similarly increased JunB expression in fibroblasts from patients with IPF and control subjects. Total JNK content was not different in either unstimulated or IL-1beta-stimulated IPF and control fibroblasts. IL-1beta increased phosphorylated JNK in control and IPF fibroblasts, but this increase was weaker and heterogeneous in IPF. Altogether, our results demonstrate a dysregulation of KGF secretion by IPF fibroblasts. The weak response to IL-1beta is associated with a defect of c-Jun expression and activation and a defect of JNK activation.


Subject(s)
Fibroblast Growth Factors/metabolism , Fibroblasts/metabolism , Pulmonary Fibrosis/pathology , Adult , Aged , Animals , Anthracenes/metabolism , Cells, Cultured , Enzyme Activation , Fibroblast Growth Factor 7 , Fibroblast Growth Factors/genetics , Fibroblasts/cytology , Humans , Interleukin-1/metabolism , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System/physiology , Middle Aged , Phosphorylation , Proto-Oncogene Proteins c-jun/metabolism , Pulmonary Fibrosis/metabolism
7.
Oncogene ; 23(27): 4735-44, 2004 Jun 10.
Article in English | MEDLINE | ID: mdl-15122334

ABSTRACT

The dual Ser/Thr kinase MKK4 and its downstream targets JNK and p38 regulate critical cellular functions during embryogenesis and development. MKK4 has been identified as a putative tumor-suppressor gene in human solid tumors of breast, prostate and pancreas. To clarify the mechanisms underlying the transforming potential of molecular defects targeting MKK4, we have generated totipotent embryonic stem (ES) cells expressing the dominant-negative mutant DN-MKK4(Ala), S257A/T261A. Stably transfected DN-MKK4-ES cells exhibit a transformed fibroblast-like morphology, reduced proliferation rate, were no more submitted to cell contact inhibition, were growing in soft agar, and were much more tumorigenic than parental ES cells in athymic nude mice. These phenotypic changes: (i) are consistent with the protection of DN-MKK4-transfected ES cells from spontaneous, cell density-dependent, and stress-induced apoptosis (DAPI staining and poly (ADP-ribose) polymerase (PARP) cleavage) and (ii) correlated with alterations in JNK, p38, and Erk-1/-2 MAPK/SAPK signaling. Taken together, our data provide a new mechanism linking the MKK4 signaling pathways to cancer progression and identify MKK4 as a tumor-suppressor gene implicated in several transforming functions.


Subject(s)
Genes, Tumor Suppressor , Mitogen-Activated Protein Kinase Kinases/physiology , Signal Transduction/genetics , Stem Cells/enzymology , Animals , Apoptosis/physiology , Cell Division/genetics , Cell Line , Cell Survival/genetics , Embryo, Mammalian/cytology , MAP Kinase Signaling System/genetics , Mice , Mice, Nude , Mitogen-Activated Protein Kinases/metabolism , Mutation , Neoplasm Transplantation , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases , Proteins/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Stem Cells/cytology , Time Factors , Transfection , p38 Mitogen-Activated Protein Kinases
8.
Hepatology ; 36(5): 1070-8, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12395316

ABSTRACT

In cirrhosis, lipopolysaccharide (LPS, a product of Gram-negative bacteria) in the blood may cause septic shock. LPS-elicited induction of arterial inducible nitric oxide synthase (iNOS) results in nitric oxide (NO)-induced vasodilation, which causes arterial hypotension and hyporeactivity to alpha(1)-adrenergic constrictors. In vitro studies have suggested that vasopressin inhibits iNOS expression in cultured vascular smooth muscle cells exposed to LPS. Thus, the aim of this study was to investigate the effects of terlipressin administration (a vasopressin analog) on in vivo LPS-induced aortic iNOS in rats with cirrhosis. LPS (1 mg/kg, intravenously) was administered followed by the intravenous administration of terlipressin (0.05 mg/kg, intravenously) or placebo 1 hour later. Arterial pressure was measured, and contractions to phenylephrine (an alpha(1)-adrenoceptor agonist), iNOS activity, and iNOS expressions (mRNA and protein) were investigated in isolated aortas. LPS-induced arterial hypotension and aortic hyporeactivity to phenylephrine were abolished in rats that received terlipressin. LPS-induced aortic iNOS activity and expression were suppressed in terlipressin-treated rats. In conclusion, in LPS-challenged rats with cirrhosis, terlipressin administration inhibits in vivo LPS-induced aortic iNOS expression. Terlipressin administration may be a novel approach for the treatment of arterial hypotension and hyporeactivity to alpha(1)-adrenergic constrictors in patients with cirrhosis and septic shock.


Subject(s)
Liver Cirrhosis, Biliary/metabolism , Lypressin/analogs & derivatives , Lypressin/pharmacology , Lysine/analogs & derivatives , Nitric Oxide Synthase/metabolism , Shock, Septic/drug therapy , Vasoconstrictor Agents/pharmacology , Alanine Transaminase/blood , Animals , Aorta, Thoracic/enzymology , Blood Pressure/drug effects , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Hypotension/chemically induced , Hypotension/drug therapy , Hypotension/metabolism , Lipopolysaccharides , Liver Cirrhosis, Biliary/drug therapy , Lysine/pharmacology , Male , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Phenylephrine/pharmacology , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Shock, Septic/chemically induced , Shock, Septic/metabolism , Terlipressin , Tumor Necrosis Factor-alpha/metabolism
9.
J Hepatol ; 37(1): 63-71, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12076863

ABSTRACT

BACKGROUND/AIMS: Hepatitis B and C viruses, two inducers of hepatocarcinomas, have been shown to activate AP-1, NF-kappa B and STAT in vitro, but no detailed information on the activity of these transcription factors in vivo have been provided. METHODS: We have measured the DNA binding activity of these transcription factors in the peri-tumoral and the tumoral parts of 15 primary liver cancers, of viral or non-viral etiologies, and in five hepatic metastases using electrophoretic mobility shift assays. RESULTS: AP-1, NF-kappa B and STAT binding activities were increased in the peritumoral tissue, compared with histologically normal livers in 73, 87 and 70%, respectively, of the cases. A further activation of AP-1, NF-kappa B, but not STAT binding in the tumoral parts was detected in 40 and 80%, respectively, of the cases. A close correlation was found between JunD and c-Jun levels and AP-1 binding activity at the tumoral stage. By contrast, AP-1 and NF-kappa B binding activities were low or only slightly elevated in the peri-tumoral and the tumoral tissue of metastases. CONCLUSIONS: Early activation of AP-1, NF-kappa B and STAT contributes probably to the acquisition of a transformed phenotype during hepatocarcinogenesis, whatever the etiology.


Subject(s)
Carcinoma, Hepatocellular/physiopathology , DNA-Binding Proteins/metabolism , Liver Neoplasms/physiopathology , NF-kappa B/metabolism , Trans-Activators/metabolism , Transcription Factor AP-1/metabolism , Gene Expression Regulation, Neoplastic , Humans , Proliferating Cell Nuclear Antigen/genetics , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-jun/genetics , RNA, Messenger/analysis , STAT1 Transcription Factor
SELECTION OF CITATIONS
SEARCH DETAIL
...