Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
J Cell Physiol ; 223(2): 309-16, 2010 May.
Article in English | MEDLINE | ID: mdl-20082306

ABSTRACT

Neurotensin (NT) stimulates protein kinase D1 (PKD1), extracellular signal regulated kinase (ERK), c-Jun N-terminal Kinase (JNK), and DNA synthesis in the human pancreatic adenocarcinoma cell line PANC-1. To determine the effect of PKD1 overexpression on these biological responses, we generated inducible stable PANC-1 clones that express wild-type (WT) or kinase-dead (K618N) forms of PKD1 in response to the ecdysone analog ponasterone-A (PonA). NT potently stimulated c-Jun Ser(63) phosphorylation in both wild type and clonal derivatives of PANC-1 cells. PonA-induced expression of WT, but not K618N PKD1, rapidly blocked NT-mediated c-Jun Ser(63) phosphorylation either at the level of or upstream of MKK4, a dual-specificity kinase that leads to JNK activation. This is the first demonstration that PKD1 suppresses NT-induced JNK/cJun activation in PANC-1 cells. In contrast, PKD1 overexpression markedly increased the duration of NT-induced ERK activation in these cells. The reciprocal influence of PKD1 signaling on pro-mitogenicERK and pro-apopotic JNK/c-Jun pathways prompted us to examine whether PKD1 overexpression promotes DNA synthesis and proliferation of PANC-1 cells. Our results show that PKD1 overexpression increased DNA synthesis and cell numbers of PANC-1 cells cultured in regular dishes or in polyhydroxyethylmethacrylate [Poly-(HEMA)]-coated dishes to eliminate cell adhesion (anchorage-independent growth). Furthermore, PKD1 overexpression markedly enhanced DNA synthesis induced by NT (1-10 nM). These results indicate that PKD1 mediates mitogenic signaling in PANC-1 and suggests that this enzyme could be a novel target for the development of therapeutic drugs that restrict the proliferation of these cells.


Subject(s)
Carcinoma/enzymology , Cell Transformation, Neoplastic/metabolism , Growth Substances/metabolism , Pancreatic Neoplasms/enzymology , Protein Kinase C/metabolism , Signal Transduction/physiology , Carcinoma/drug therapy , Carcinoma/genetics , Cell Count , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/genetics , DNA/biosynthesis , DNA/drug effects , DNA Replication/drug effects , Ecdysterone/analogs & derivatives , Ecdysterone/pharmacology , Extracellular Signal-Regulated MAP Kinases/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/physiology , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/physiology , Growth Substances/genetics , Humans , JNK Mitogen-Activated Protein Kinases/drug effects , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Kinase 4/drug effects , MAP Kinase Kinase 4/metabolism , Neurotensin/antagonists & inhibitors , Neurotensin/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Phosphorylation/drug effects , Protein Kinase C/genetics , Signal Transduction/drug effects
2.
Biochem Biophys Res Commun ; 302(4): 800-4, 2003 Mar 21.
Article in English | MEDLINE | ID: mdl-12646240

ABSTRACT

Protein kinase D (PKD) has been established as a negative modulator of the c-Jun N-terminal kinase (JNK) signaling pathway. We previously demonstrated that induced expression of constitutively active PKD (PKD-S744/748E) that mimics phosphorylation by PKC is sufficient to attenuate epidermal growth factor (EGF) stimulated c-Jun Ser 63 phosphorylation, a natural substrate of JNK, in HEK 293 cells. Because the JNK pathway has been implicated in sustaining both lung and pancreatic cancerous phenotypes, we have utilized stable inducible expression of PKD-S744/748E in clones of A549 non-small cell lung cancer (NSCLC) and Panc1, pancreatic cancer cells to determine its effects on JNK signaling in the context of the cancerous phenotype. In contrast to HEK 293 cells, induced expression of PKD-S744/748E in either A549 NSCLC or Panc1 cells failed to attenuate EGF dependent phosphorylation of c-Jun, indicating that EGF stimulated JNK phosphorylation of c-Jun is uncoupled from PKD suppression in these cancer cells.


Subject(s)
Epidermal Growth Factor/metabolism , Mitogen-Activated Protein Kinases/metabolism , Protein Kinase C/metabolism , Signal Transduction/physiology , Ecdysterone/analogs & derivatives , Ecdysterone/metabolism , Humans , JNK Mitogen-Activated Protein Kinases , Phosphorylation , Serine/metabolism , Tumor Cells, Cultured
3.
Oncogene ; 21(14): 2154-60, 2002 Mar 28.
Article in English | MEDLINE | ID: mdl-11948398

ABSTRACT

Protein kinase D (PKD), a downstream effector of protein kinase C (PKC), is implicated in suppression of the c-Jun N-terminal kinase (JNK) signaling pathway, however, its mechanism of action is unclear. Transphosphorylation of the PKD activation loop at serines 744/748 by a PKC mediated signal transduction pathway enhances its catalytic activity. Here we show that PKD activation loop phosphorylation at serines 744/748 via PKC, or mutation of these serines to glutamic acid (PKD-S744/748E) also results in complex formation with JNK, indicating that suppression of JNK signaling by PKD involves a direct interaction with JNK. Because catalytically active PKD associates with JNK we determined whether it could phosphorylate the c-Jun N-terminus as a potential mechanism by which it suppresses c-Jun Ser 63 phosphorylation when it complexes with JNK. Purified human PKD and either wild-type PKD from phorbol 12, 13-dibutyrate (PDB)-stimulated cells or unstimulated constitutively active PKD (PKD-S744/748E), phosphorylated the c-Jun N-terminus between amino acids 1-89 at sites distinct from those phosphorylated by JNK. These results demonstrate, for the first time, phosphorylation dependent association of PKD with another signaling molecule and reveal a potential mechanism by which PKD could modulate the ability of JNK to phosphorylate c-Jun by phosphorylating alternative sites in the c-Jun N-terminus when it is complexed with JNK.


Subject(s)
Mitogen-Activated Protein Kinases/metabolism , Protein Kinase C/metabolism , Proto-Oncogene Proteins c-jun/chemistry , Proto-Oncogene Proteins c-jun/metabolism , Animals , Blotting, Western , COS Cells , Cell Line , Chromatography, Thin Layer , Humans , Isoenzymes/metabolism , JNK Mitogen-Activated Protein Kinases , Macromolecular Substances , Mitogen-Activated Protein Kinase 1/metabolism , Mutation , Phosphopeptides/analysis , Phosphopeptides/metabolism , Phosphorylation , Precipitin Tests , Protein Binding , Protein Kinase C/genetics , Protein Kinase C-epsilon , Signal Transduction , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...