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.
Oncogene ; 33(19): 2454-63, 2014 May 08.
Article in English | MEDLINE | ID: mdl-23752186

ABSTRACT

Despite frequent KRAS mutation, the early molecular mechanisms of pancreatic ductal adenocarcinoma (PDAC) development have not been fully elucidated. By tracking a potential regulator of another feature of PDAC precursors, acquisition of foregut or gastric epithelial gene signature, we herein report that aberrant overexpression of ecotropic viral integration site 1 (EVI1) oncoprotein, which is usually absent in normal pancreatic duct, is a widespread marker across the full spectrum of human PDAC precursors and PDAC. In pancreatic cancer cells, EVI1 depletion caused remarkable inhibition of cell growth and migration, indicating its oncogenic roles. Importantly, we found that EVI1 upregulated KRAS expression through suppression of a potent KRAS suppressor, miR-96, in pancreatic cancer cells. Collectively, the present findings suggest that EVI1 overexpression and KRAS mutation converge on activation of the KRAS pathway in early phases of pancreatic carcinogenesis and propose EVI1 and/or miR-96 as early markers and therapeutic targets in this dismal disease.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , DNA-Binding Proteins/genetics , MicroRNAs/genetics , Pancreatic Neoplasms/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogenes/genetics , Transcription Factors/genetics , ras Proteins/genetics , Blotting, Western , Carcinoma, Pancreatic Ductal/metabolism , Cell Line, Tumor , Chromatin Immunoprecipitation , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic/genetics , Humans , Immunohistochemistry , MDS1 and EVI1 Complex Locus Protein , Mutation , Oligonucleotide Array Sequence Analysis , Oncogenes , Pancreatic Neoplasms/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins p21(ras) , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Transcription Factors/metabolism , Up-Regulation , ras Proteins/metabolism
2.
Br J Cancer ; 102(1): 206-12, 2010 Jan 05.
Article in English | MEDLINE | ID: mdl-19997106

ABSTRACT

BACKGROUND: Tumour-initiating cells (TICs) or cancer stem cells can exist as a small population in malignant tissues. The signalling pathways activated in TICs that contribute to tumourigenesis are not fully understood. METHODS: Several breast cancer cell lines were sorted with CD24 and CD44, known markers for enrichment of breast cancer TICs. Tumourigenesis was analysed using sorted cells and total RNA was subjected to gene expression profiling and gene set enrichment analysis (GSEA). RESULTS: We showed that several breast cancer cell lines have a small population of CD24(-/low)/CD44(+) cells in which TICs may be enriched, and confirmed the properties of TICs in a xenograft model. GSEA revealed that CD24(-/low)/CD44(+) cell populations are enriched for genes involved in transforming growth factor-beta, tumour necrosis factor, and interferon response pathways. Moreover, we found the presence of nuclear factor-kappaB (NF-kappaB) activity in CD24(-/low)/CD44(+) cells, which was previously unrecognised. In addition, NF-kappaB inhibitor dehydroxymethylepoxyquinomicin (DHMEQ) prevented tumourigenesis of CD24(-/low)/CD44(+) cells in vivo. CONCLUSION: Our findings suggest that signalling pathways identified using GSEA help to identify molecular targets and biomarkers for TIC-like cells.


Subject(s)
Breast Neoplasms/pathology , CD24 Antigen/analysis , Cell Separation/methods , Gene Expression Profiling , Hyaluronan Receptors/analysis , Neoplasm Proteins/analysis , Neoplastic Stem Cells/physiology , Animals , Benzamides/pharmacology , Benzamides/therapeutic use , Biomarkers , Breast Neoplasms/genetics , Cyclohexanones/pharmacology , Cyclohexanones/therapeutic use , Female , Genetic Vectors/pharmacology , Humans , Mice , Mice, Inbred NOD , Mice, SCID , NF-kappa B/antagonists & inhibitors , NF-kappa B/physiology , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/physiology , Neoplastic Stem Cells/chemistry , Neoplastic Stem Cells/transplantation , Oligonucleotide Array Sequence Analysis , Signal Transduction/physiology , Transduction, Genetic , Xenograft Model Antitumor Assays
3.
Proc Natl Acad Sci U S A ; 98(1): 125-9, 2001 Jan 02.
Article in English | MEDLINE | ID: mdl-11134534

ABSTRACT

The role of PKN, a fatty acid- and Rho small GTPase-activated protein kinase, in cell-cycle regulation was analyzed. Microinjection of the active form of PKN into a Xenopus embryo caused cleavage arrest, whereas normal cell division proceeded in the control embryo microinjected with buffer or the inactive form of PKN. Exogenous addition of the active form of PKN delayed mitotic timing in Xenopus egg cycling extracts judging by morphology of sperm nuclei and Cdc2/cyclin B histone H1 kinase activity. The kinase-negative form of PKN did not affect the timing, suggesting that delayed mitotic timing depends on the kinase activity of PKN. The dephosphorylation of Tyr-15 of Cdc2 was also delayed in correlation with Cdc2/cyclin B histone H1 kinase activation in extracts containing active PKN. The Cdc25C activity for the dephosphorylation of Tyr-15 in Cdc2 was suppressed by pretreatment with the active form of PKN. Furthermore, PKN efficiently phosphorylated Cdc25C in vitro, indicating that PKN directly inhibits Cdc25C activity by phosphorylation. These results suggest that PKN plays a significant role in the control of mitotic timing by inhibition of Cdc25C.


Subject(s)
Cell Cycle Proteins , Mitosis/drug effects , Nuclear Proteins , Protein Serine-Threonine Kinases/pharmacology , Protein-Tyrosine Kinases/pharmacology , ras-GRF1/antagonists & inhibitors , Animals , CDC2 Protein Kinase/metabolism , Cell Extracts , Cell Nucleus/metabolism , Cyclin B/metabolism , Enzyme Activation/drug effects , Female , Male , Microinjections , Oocytes/cytology , Oocytes/enzymology , Oocytes/metabolism , Phosphorylation/drug effects , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein Tyrosine Phosphatases/metabolism , Protein-Tyrosine Kinases/metabolism , Recombinant Fusion Proteins , Spermatozoa/cytology , Xenopus/embryology , Xenopus/metabolism , Xenopus Proteins , ras-GRF1/metabolism
4.
J Biol Chem ; 276(13): 10025-31, 2001 Mar 30.
Article in English | MEDLINE | ID: mdl-11104762

ABSTRACT

For the phosphorylation state of microtubule-associated protein, tau plays a pivotal role in regulating microtubule networks in neurons. Tau promotes the assembly and stabilization of microtubules. The potential for tau to bind to microtubules is down-regulated after local phosphorylation. When we investigated the effects of PKN activation on tau phosphorylation, we found that PKN triggers disruption of the microtubule array both in vitro and in vivo and predominantly phosphorylates tau in microtubule binding domains (MBDs). PKN has a catalytic domain highly homologous to protein kinase C (PKC), a kinase that phosphorylates Ser-313 (= Ser-324, the number used in this study) in MBDs. Thus, we identified the phosphorylation sites of PKN and PKC subtypes (PKC-alpha, -betaI, -betaII, -gamma, -delta, -epsilon, -zeta, and -lambda) in MBDs. PKN phosphorylates Ser-258, Ser-320, and Ser-352, although all PKC subtypes phosphorylate Ser-258, Ser-293, Ser-324, and Ser-352. There is a PKN-specific phosphorylation site, Ser-320, in MBDs. HIA3, a novel phosphorylation-dependent antibody recognizing phosphorylated tau at Ser-320, showed immunoreactivity in Chinese hamster ovary cells expressing tau and the active form of PKN, but not in Chinese hamster ovary cells expressing tau and the inactive form of PKN. The immunoreactivity for phosphorylated tau at Ser-320 increased in the presence of a phosphatase inhibitor, FK506 treatment, which means that calcineurin (protein phosphatase 2B) may be involved in dephosphorylating tau at Ser-320 site. We also noted that PKN reduces the phosphorylation recognized by the phosphorylation-dependent antibodies AT8, AT180, and AT270 in vivo. Thus PKN serves as a regulator of microtubules by specific phosphorylation of tau, which leads to disruption of tubulin assembly.


Subject(s)
Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/physiology , Protein-Tyrosine Kinases/metabolism , Protein-Tyrosine Kinases/physiology , tau Proteins/metabolism , Amino Acid Sequence , Animals , Binding Sites , CHO Cells , Calcineurin/metabolism , Catalytic Domain , Cricetinae , Down-Regulation , Glutathione Transferase/metabolism , Humans , Immunoblotting , Immunosuppressive Agents/pharmacology , Microscopy, Fluorescence , Microtubules/metabolism , Molecular Sequence Data , Phosphorylation , Protein Binding , Protein Kinase C/metabolism , Protein Structure, Tertiary , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Serine/metabolism , Tacrolimus/pharmacology , Time Factors , Transfection , Tubulin/metabolism , Tumor Cells, Cultured
5.
J Biol Chem ; 275(44): 34592-6, 2000 Nov 03.
Article in English | MEDLINE | ID: mdl-10945988

ABSTRACT

Protein kinase C (PKC) family requires phosphorylation of itself to become competent for responding to second messengers. Much attention has been focused on elucidating the role of phosphorylation in PKC activity; however, it remains unknown where this modification takes place in the cells. This study examines whether anchoring protein is involved in the regulation of PKC phosphorylation. A certain population of PKC epsilon in rat brain extracts as well as that expressed in COS7 cells was associated with an endogenous anchoring protein CG-NAP (centrosome and Golgi localized PKN- associated protein). Pulse chase experiments revealed that the associated PKC epsilon was an immature species at the hypophosphorylated state. In vitro binding studies confirmed that non- or hypophosphorylated PKC epsilon directly bound to CG-NAP via its catalytic domain, whereas sufficiently phosphorylated PKC epsilon did not. PKC epsilon mutant at a potential phosphorylation site of Thr-566 or Ser-729 to Ala, possessing almost no catalytic activity, was associated and co-localized with CG-NAP at Golgi/centrosome area. On the other hand, wild type and a phosphorylation-mimicking mutant at Thr-566 were mainly distributed in cytosol and represented second messenger-dependent catalytic activation. These results suggest that CG-NAP anchors hypophosphorylated PKCepsilon at the Golgi/centrosome area during maturation and serves as a scaffold for the phosphorylation reaction.


Subject(s)
Adaptor Proteins, Signal Transducing , Carrier Proteins/metabolism , Cytoskeletal Proteins , Isoenzymes/metabolism , Protein Kinase C/metabolism , A Kinase Anchor Proteins , Animals , COS Cells , Phosphorylation , Protein Binding , Protein Kinase C-epsilon , Second Messenger Systems
6.
Biochem Biophys Res Commun ; 273(1): 209-12, 2000 Jun 24.
Article in English | MEDLINE | ID: mdl-10873588

ABSTRACT

We analyzed the effects of PKNalpha and protein kinase C (PKC) on phosphorylation of tau protein by glycogen synthase kinase (GSK)-3beta using monoclonal antibodies (AT8, AT180, and AT270). These antibodies are highly specific for phosphorylated tau in Alzheimer paired helical filaments, and recognize phosphorylated Ser202/Thr205, Thr231, and Thr181 of tau protein, respectively. Immunoblot analysis demonstrated that PKNalpha and PKC did not directly phosphorylate their sites, whereas GSK-3beta efficiently did so. Incubating GSK-3beta with PKNalpha or PKC subtypes inhibited subsequent GSK-3beta-induced AT8 and AT270 immunoreactivity. However, the constitutive active form of the GSK-3beta(S9A) mutant was almost totally inert to each enzyme. Incubating tau with PKNalpha increased the GSK-3beta-induced AT180 immunoreactivity, which was further enhanced when the S9A mutant was used instead of the wild type GSK-3beta. These results suggest that PKNalpha and PKC directly inhibit GSK-3beta activity at least in part by phosphorylating Ser9 of GSK-3beta, and that they indirectly suppress GSK-3beta-stimulated phosphorylation of tau at amino acids Ser202/Thr205 and Thr181, but enhanced phosphorylation at Thr231 through phosphorylation at other sites of tau.


Subject(s)
Bacterial Proteins , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Protein Kinase C/metabolism , Protein Kinases/metabolism , tau Proteins/metabolism , Amino Acid Sequence , Animals , Antibodies, Monoclonal/immunology , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Epitopes/immunology , Glycogen Synthase Kinase 3 , Glycogen Synthase Kinases , Humans , Molecular Sequence Data , Mutation/genetics , Phosphoproteins/chemistry , Phosphoproteins/immunology , Phosphoproteins/metabolism , Phosphorylation , Phosphoserine/immunology , Phosphoserine/metabolism , Phosphotyrosine/immunology , Phosphotyrosine/metabolism , Protein Kinase C/chemistry , Protein Kinases/chemistry , Protein Kinases/genetics , Rats , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , tau Proteins/chemistry , tau Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...