Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters











Publication year range
1.
Cancer Gene Ther ; 17(5): 307-14, 2010 May.
Article in English | MEDLINE | ID: mdl-20075984

ABSTRACT

As we have recently found a novel oncogene, the cancer-upregulated gene 2 (CUG2), which was elevated in a variety of tumor tissues such as the ovary, liver, lung and pancreas, we examined whether reovirus could efficiently induce cytolysis in cancer cells expressing CUG2 and thus be used as a potential cancer therapeutic agent. In this study, we describe experiments in which we use reovirus to treat NIH3T3 cells stably expressing either CUG2 (NIH-CUG2) or vector only (NIH-Vec). NIH-CUG2 cells readily support reoviral proliferation and undergo apoptosis, whereas NIH-Vec cells are highly resistant to reoviral infection and virus-induced apoptosis. This notable result may be explained by the observation that CUG2 expression inhibits PKR activation, leading to reoviral proliferation in nonpermissive NIH3T3 cells. Furthermore, reovirus infection results in almost complete regression of tumorgenic NIH-CUG2 cells in transplanted nude mice. As we found that CUG2 enhances activation of MAPK (ERK, JNK and p38), Src kinase and Ras, we examined whether CUG2 confers reoviral replication independent of the Ras or p38 MAPK signaling pathway. From these experiments we found that either inhibition of p38 MAPK or Ras blocks reoviral proliferation even in the presence of CUG2 but inhibition of ERK, JNK and Src kinase does not, indicating that activation of p38 MAPK and Ras has critical roles in reoviral replication in CUG2-expressing tumor cells. Accordingly, we propose that reovirus can be useful in the treatment of transformed cells expressing CUG2, which is commonly detected in various tumor tissues.


Subject(s)
Apoptosis/physiology , Nuclear Proteins/metabolism , Reoviridae/physiology , Signal Transduction/physiology , p38 Mitogen-Activated Protein Kinases/metabolism , ras Proteins/metabolism , Animals , Apoptosis/genetics , Blotting, Western , Cell Line , Chromosomal Proteins, Non-Histone , Immunohistochemistry , In Situ Nick-End Labeling , Male , Mice , Mice, Nude , NIH 3T3 Cells , Nuclear Proteins/genetics , Proliferating Cell Nuclear Antigen/metabolism , Reoviridae/growth & development , p38 Mitogen-Activated Protein Kinases/genetics , ras Proteins/genetics
2.
Cancer Gene Ther ; 17(5): 365-72, 2010 May.
Article in English | MEDLINE | ID: mdl-20075985

ABSTRACT

Many oncolytic viruses are currently being tested as potential cancer therapeutic agents. To be effective, these viruses must replicate and propagate efficiently through the tumor mass. However, it is possible that the hypoxia that characterizes many tumors may be an obstacle to viral therapy because of its inhibition of viral replication and propagation. We, therefore, decided to test how oncolytic reovirus and its target cells respond to hypoxia. We found that reovirus infection suppresses hypoxia inducible factor (HIF)-1alpha protein levels (but not transcript abundance) in colon cancer HCT116 cells under CoCl(2) or hypoxia. Reovirus infection was able to reduce HIF-1alpha levels in both von Hippel Lindau (VHL)-/- renal carcinoma A498 and p53-/- HCT116 cells, indicating that the decrease of HIF-1alpha mediated by reovirus requires neither VHL nor p53 proteins. However, treatment with the inhibitor MG132 restored HIF-1alpha levels, suggesting that reovirus-induced HIF-1alpha decrease needs proteosomal activity. A498 VHL-/- cells with constitutive expression of HIF-1alpha were relatively resistant to reovirus-induced apoptosis when compared with A498 VHL+/+ cells. However, we found that the use of YC-1 to target HIF-1alpha promoted reovirus-induced apoptosis in A498 VHL-/- cells. Accordingly, we propose that reovirus may be used together with YC-1 as a potential therapeutic agent against chemoresistant or radioresistant tumors that are hypoxic and show increased levels of HIF-1alpha.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Oncolytic Viruses/physiology , Tumor Suppressor Protein p53/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Animals , Apoptosis/genetics , Apoptosis/physiology , Blotting, Western , Cell Hypoxia/genetics , Cell Hypoxia/physiology , Cell Line, Tumor , Down-Regulation , HCT116 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mice , Oncolytic Viruses/genetics , Reverse Transcriptase Polymerase Chain Reaction
3.
Cancer Gene Ther ; 16(5): 453-61, 2009 May.
Article in English | MEDLINE | ID: mdl-19096445

ABSTRACT

Although reovirus has been used in tests as a potential cancer therapeutic agent against a variety of cancer cells, its application to hepatocellular carcinoma cells, in which the hepatitis B virus (HBV) X (HBX) protein of HBV plays a primary role, has not yet been explored. Here, we describe experiments in which we use reovirus to treat Chang liver carcinoma cells expressing either a vector only (Chang-vec) or a vector encoding HBX protein (Chang-HBX). Although Chang-vec cells readily support reoviral proliferation and undergo apoptosis, Chang-HBX cells are highly resistant to reoviral infection and virus-induced apoptosis, even though HBX protein induces activation of Ras and inactivation of PKR, which are normally thought to enhance reoviral oncolysis. The resistance of Chang-HBX cells to reovirus may instead be explained by HBX-induced downregulation of death receptor 5 and activation of Stat1. Phosphorylated Stat1 activates interferon (IFN)-stimulated regulatory element (ISRE)- and IFN-gamma-activated sequence (GAS)-mediated transcription, leading to the production of IFN-beta, whereas the reduced expression of Stat1 with its siRNA results in a decrease in IFN-beta production, by which Chang-HBX cells eventually succumb to reovirus infection. This result further indicates that HBX induces the establishment of an antiviral state through Stat1 activation. Thus, it appears that active Ras does not override the antiviral effect mediated by the activation of Stat1. Accordingly, we report that HBX, an oncoprotein of HBV, can prevent reoviral oncolysis of hepatocellular carcinoma. This suggests there may be limits to the practical application of reovirus in the treatment of human cancers already expressing other oncoviral proteins.


Subject(s)
Carcinoma, Hepatocellular/therapy , Gene Expression Regulation , Liver Neoplasms/therapy , Reoviridae/metabolism , Trans-Activators/metabolism , Animals , Apoptosis , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation , Hepatitis B virus/genetics , Hepatitis B virus/metabolism , Humans , Interferon-alpha/immunology , Interferon-alpha/metabolism , Liver Neoplasms/pathology , Mice , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Reoviridae/genetics , Signal Transduction/physiology , TNF-Related Apoptosis-Inducing Ligand/metabolism , Trans-Activators/genetics , Viral Regulatory and Accessory Proteins , eIF-2 Kinase/metabolism , ras Proteins/metabolism
5.
J Biol Chem ; 276(27): 24645-53, 2001 Jul 06.
Article in English | MEDLINE | ID: mdl-11323430

ABSTRACT

Activation of Ras signaling by growth factors has been associated with gene regulation and cell proliferation. Here we characterize the contributory role of cytosolic phospholipase A(2) in the oncogenic Ha-Ras(V12) signaling pathway leading to activation of c-fos serum response element (SRE) and transformation in Rat-2 fibroblasts. Using a c-fos SRE-luciferase reporter gene, we showed that the transactivation of SRE by Ha-Ras(V12) is mainly via a Rac-linked cascade, although the Raf-mitogen-activated protein kinase cascade is required for full activation. In addition, Ha-Ras(V12)-induced DNA synthesis was significantly attenuated by microinjection of recombinant Rac(N17), a dominant negative mutant of Rac1. To identify the mediators downstream of Rac in the Ha-Ras(V12) signaling, we investigated the involvement of cytosolic phospholipase A(2). Oncogenic Ha-Ras(V12)-induced SRE activation was significantly inhibited by either pretreatment with mepacrine, a phospholipase A(2) inhibitor, or cotransfection with the antisense oligonucleotide of cytosolic phospholipase A(2). We also found cytosolic phospholipase A(2) to be situated downstream of Ha-Ras(V12) in a signal pathway leading to transformation. Together, these results are indicative of mediatory roles of Rac and cytosolic phospholipase A(2) in the signaling pathway by which Ha-Ras(V12) transactivates c-fos SRE and transformation. Our findings point to cytosolic phospholipase A(2) as a novel potential target for suppressing oncogenic Ha-Ras(V12) signaling in the cell.


Subject(s)
Genes, ras/physiology , Phospholipases A/metabolism , Signal Transduction , ras Proteins/physiology , Animals , Base Sequence , DNA Replication/drug effects , DNA-Binding Proteins/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Genes, Reporter , Luciferases/genetics , Microinjections , Molecular Sequence Data , Nuclear Proteins/metabolism , Oligonucleotides, Antisense/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phospholipases A2 , Quinacrine/pharmacology , Rats , Serum Response Factor , Transcriptional Activation/drug effects , Transfection , rac1 GTP-Binding Protein/metabolism
6.
Mol Endocrinol ; 14(6): 915-25, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10847592

ABSTRACT

ASC-2 was recently discovered as a cancer-amplified transcription coactivator molecule of nuclear receptors, which interacts with multifunctional transcription integrators steroid receptor coactivator-1 (SRC-1) and CREB-binding protein (CBP)/p300. Herein, we report the identification of three mitogenic transcription factors as novel target molecules of ASC-2. First, the C-terminal transactivation domain of serum response factor (SRF) was identified among a series of ASC-2-interacting proteins from the yeast two-hybrid screening. Second, ASC-2 specifically interacted with the activating protein-1 (AP-1) components c-Jun and c-Fos as well as the nuclear factor-kappaB (NFkappaB) components p50 and p65, as demonstrated by the glutathione S-transferase pull-down assays as well as the yeast two-hybrid tests. In cotransfection of mammalian cells, ASC-2 potentiated transactivations by SRF, AP-1, and NFkappaB in a dose-dependent manner, either alone or in conjunction with SRC-1 and p300. In addition, ASC-2 efficiently relieved the previously described transrepression between nuclear receptors and either AP-1 or NFkappaB. Overall, these results suggest that the nuclear receptor coactivator ASC-2 also mediates transactivations by SRF, AP-1, and NFkappaB, which may contribute to the putative, ASC-2-mediated tumorigenesis.


Subject(s)
DNA-Binding Proteins/pharmacology , Intracellular Signaling Peptides and Proteins , NF-kappa B/pharmacology , Neoplasms/metabolism , Nuclear Proteins/pharmacology , Transcription Factor AP-1/pharmacology , Transcription Factors/pharmacology , 3T3 Cells , Animals , Binding Sites , DNA/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drug Synergism , Gene Expression , Genes, fos , Genes, jun , Glutathione Transferase/genetics , HeLa Cells , Humans , Mice , NF-kappa B/genetics , NF-kappa B/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nuclear Receptor Coactivators , Recombinant Fusion Proteins/metabolism , Serum Response Factor , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation/drug effects , Transfection
7.
J Biol Chem ; 274(48): 34283-93, 1999 Nov 26.
Article in English | MEDLINE | ID: mdl-10567404

ABSTRACT

Many transcription coactivators interact with nuclear receptors in a ligand- and C-terminal transactivation function (AF2)-dependent manner. We isolated a nuclear factor (designated ASC-2) with such properties by using the ligand-binding domain of retinoid X receptor as a bait in a yeast two-hybrid screening. ASC-2 also interacted with other nuclear receptors, including retinoic acid receptor, thyroid hormone receptor, estrogen receptor alpha, and glucocorticoid receptor, basal factors TFIIA and TBP, and transcription integrators CBP/p300 and SRC-1. In transient cotransfections, ASC-2, either alone or in conjunction with CBP/p300 and SRC-1, stimulated ligand-dependent transactivation by wild type nuclear receptors but not mutant receptors lacking the AF2 domain. Consistent with an idea that ASC-2 is essential for the nuclear receptor function in vivo, microinjection of anti-ASC-2 antibody abrogated the ligand-dependent transactivation of retinoic acid receptor, and this repression was fully relieved by coinjection of ASC-2-expression vector. Surprisingly, ASC-2 was identical to a gene previously identified during a search for genes amplified and overexpressed in breast and other human cancers. From these results, we concluded that ASC-2 is a bona fide transcription coactivator molecule of nuclear receptors, and its altered expression may contribute to the development of cancers.


Subject(s)
Intracellular Signaling Peptides and Proteins , Receptors, Cytoplasmic and Nuclear/physiology , Trans-Activators/physiology , Transcription Factors/physiology , Transcriptional Activation/genetics , Amino Acid Sequence , Animals , Cloning, Molecular , Gene Amplification , Gene Expression , Gene Expression Regulation, Neoplastic , Histone Acetyltransferases , Humans , Ligands , Molecular Sequence Data , Neoplasms/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nuclear Receptor Coactivator 1 , Nuclear Receptor Coactivators , Oocytes/metabolism , Protein Binding , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Cells, Cultured , Two-Hybrid System Techniques , Xenopus
8.
J Biol Chem ; 274(40): 28491-6, 1999 Oct 01.
Article in English | MEDLINE | ID: mdl-10497212

ABSTRACT

Bcl3, an IkappaB protein, was originally isolated as a putative proto-oncogene in a subset of B cell chronic lymphocytic leukemias. Bcl3 was subsequently shown to associate tightly with and transactivate the NFkappaB p50 or p52 homodimer. Herein, we show that Bcl3 stimulates the activating protein-1 (AP-1) transactivation, either alone or in conjunction with transcription integrators steroid receptor coactivator-1 and CREB-binding protein/p300. The C-terminal 158 residues of Bcl3 exhibited an autonomous transactivation function and interacted with specific subregions of the AP-1 components c-Jun and c-Fos, CREB-binding protein/p300, and steroid receptor coactivator-1, as demonstrated by the yeast and mammalian two-hybrid tests as well as glutathione S-transferase pull-down assays. In addition, anti-HA antibody co-precipitated c-Jun from HeLa cells co-expressing c-Jun and HA-tagged Bcl3, consistent with the idea that Bcl3 directly associates with AP-1 in vivo. Furthermore, microinjection of Bcl3 expression vector into Rat-1 fibroblast cells significantly enhanced DNA synthesis and expression of c-jun, one of the cellular target genes of AP-1. These results suggest that Bcl3 may directly participate in the tumorigenesis processes as a novel transcription coactivator of the mitogenic transcription factor AP-1 in vivo.


Subject(s)
Cell Division/physiology , I-kappa B Proteins/metabolism , Proto-Oncogene Proteins/physiology , Transcription Factor AP-1/genetics , Transcriptional Activation/physiology , Animals , B-Cell Lymphoma 3 Protein , Cell Line , DNA Replication , Humans , Protein Binding , Proto-Oncogene Mas , Proto-Oncogene Proteins/metabolism , Transcription Factors
9.
Mol Cell Biol ; 19(9): 6323-32, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10454579

ABSTRACT

Activating signal cointegrator 1 (ASC-1) harbors an autonomous transactivation domain that contains a putative zinc finger motif which provides binding sites for basal transcription factors TBP and TFIIA, transcription integrators steroid receptor coactivator 1 (SRC-1) and CBP-p300, and nuclear receptors, as demonstrated by the glutathione S-transferase pull-down assays and the yeast two-hybrid tests. The ASC-1 binding sites involve the hinge domain but not the C-terminal AF2 core domain of nuclear receptors. Nonetheless, ASC-1 appears to require the AF2-dependent factors to function (i.e., CBP-p300 and SRC-1), as suggested by the ability of ASC-1 to coactivate nuclear receptors, either alone or in cooperation with SRC-1 and p300, as well as its inability to coactivate a mutant receptor lacking the AF2 core domain. By using indirect immunofluorescence, we further show that ASC-1, a nuclear protein, is localized to the cytoplasm under conditions of serum deprivation but is retained in the nucleus when it is serum starved in the presence of ligand or coexpressed CBP or SRC-1. These results suggest that ASC-1 is a novel coactivator molecule of nuclear receptors which functions in conjunction with CBP-p300 and SRC-1 and may play an important role in establishing distinct coactivator complexes under different cellular conditions.


Subject(s)
Receptors, Cytoplasmic and Nuclear/metabolism , Trans-Activators/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cell Line , Cell Nucleus/metabolism , Culture Media, Serum-Free , Cytosol/metabolism , DNA, Complementary/genetics , DNA, Complementary/isolation & purification , HeLa Cells , Histone Acetyltransferases , Humans , Models, Biological , Molecular Sequence Data , Nuclear Proteins/metabolism , Nuclear Receptor Coactivator 1 , Receptors, Retinoic Acid/metabolism , Receptors, Steroid/metabolism , Retinoid X Receptors , Sequence Homology, Amino Acid , Signal Transduction , Trans-Activators/chemistry , Trans-Activators/genetics , Transcription Factors/metabolism , Zinc Fingers
10.
Exp Mol Med ; 31(2): 76-82, 1999 Jun 30.
Article in English | MEDLINE | ID: mdl-10410306

ABSTRACT

The chronic myelogenous leukemic K562 cell line carrying Bcr-Abl tyrosine kinase is considered as pluripotent hematopoietic progenitor cells expressing markers for erythroid, granulocytic, monocytic, and megakaryocytic lineages. Here we investigated the signaling modulations required for induction of erythroid differentiation of K562 cells. When the K562 cells were treated with herbimycin A (an inhibitor of protein tyrosine kinase), ras antisense oligonucleotide, and PD98059 (a specific inhibitor of MEK), inhibition of ERK/MAPK activity and cell growth, and induction of erythroid differentiation were observed. The ras mutant, pZIPRas61leu-transfected cells, K562-Ras61leu, have shown a markedly decreased cell proliferation rate with approximately 2-fold doubling time, compared with the parental K562 cells, and about 60% of these cells have shown the phenotype of erythroid differentiation. In addition, herbimycin A inhibited the growth rate and increased the erythroid differentiation, but did not affect the elevated activity of ERK/MAPK in the K562-Ras61leu cells. On the other hand, effects of PD98059 on the growth and differentiation of K562-Ras61leu cells were biphasic. At low concentration of PD98059, which inhibited the elevated activity of ERK/MAPK to the level of parental cells, the growth rate increased and the erythroid differentiation decreased slightly, and at high concentration of PD98059, which inhibited the elevated activity of ERK/MAPK below that of the parental cells, the growth rate turned down and the erythroid differentiation was restored to the untreated control level. Taken together, these results suggest that an appropriate activity of ERK/MAPK is required to maintain the rapid growth and transformed phenotype of K562 cells.


Subject(s)
Erythroid Precursor Cells/physiology , Erythropoiesis , ras Proteins/metabolism , Androstadienes/pharmacology , Benzoquinones , Calcium-Calmodulin-Dependent Protein Kinases , Cell Differentiation/drug effects , Enzyme Inhibitors/pharmacology , Erythroid Precursor Cells/cytology , Flavonoids/pharmacology , Humans , K562 Cells , Lactams, Macrocyclic , Leukemia, Myeloid/pathology , Oligonucleotides, Antisense/pharmacology , Quinones/pharmacology , Rifabutin/analogs & derivatives , Wortmannin
11.
Mol Cells ; 8(1): 90-5, 1998 Feb 28.
Article in English | MEDLINE | ID: mdl-9571637

ABSTRACT

Rac, a member of the Rho family GTPases, has been implicated in the regulation of a wide range of biological processes including actin remodeling, cell transformation, G1 cell cycle progression, and gene expression. To determine whether Rac GTPase activity is required for epidermal growth factor-induced mitogenesis, Rat-2 stable cells expressing a dominant-negative Rac1 mutant, RacN17, were prepared. Exposure to EGF exhibited a significantly restricted growth response in Rat-2-RacN17 cells compared to Rat-2 parental cells, suggesting an essential role of Rac in EGF-induced mitogenesis. In contrast, addition of lysophosphatidic acid exerted the same level of growth in Rat-2 and Rat-2-RacN17 cells. To gain further evidence for the essential role of Rac in EGF-induced mitogenesis, we performed the microinjection experiment. EGF-induced DNA synthesis was significantly blocked by microinjection of recombinant RacN17 protein, and not control IgG. Our further study to analyze the downstream mediator of Rac in EGF-signaling to mitogenesis demonstrated that Rac-activated phospholipase A2 plays a critical role. Taken together, our results suggest that the "Rac and Rac-activated PLA2" cascade is one of the major mitogenic pathways induced by EGF.


Subject(s)
Epidermal Growth Factor/pharmacology , GTP Phosphohydrolases/metabolism , GTP-Binding Proteins/metabolism , Mitogens/pharmacology , Mitosis/drug effects , Animals , Cell Division/drug effects , Cells, Cultured , DNA/biosynthesis , DNA/drug effects , Enzyme Activation , Fibroblasts , GTP Phosphohydrolases/physiology , GTP-Binding Proteins/genetics , Growth Inhibitors/physiology , Microinjections , Mitosis/genetics , Phospholipases A/metabolism , Phospholipases A/physiology , Phospholipases A2 , Rats , Recombinant Proteins/pharmacology , rac GTP-Binding Proteins
12.
Arch Pharm Res ; 20(4): 297-305, 1997 Aug.
Article in English | MEDLINE | ID: mdl-18975169

ABSTRACT

A new serum-free defined medium was developed that supports the growth of normal rat mammary epithelial cells. Mammary organoids from the glands of female F344 rats were cultured in a serum-free medium. Monolayer culture colonies developed within a week and remained viable for months in culture. Upon subculture of one-week-old primary colonies, almost the same morphology of colonies was developed. The scrape loading/dye transfer technique showed that most of colonies that developed in a serum-free medium containing EGF, human transferrin, insulin, and hydrocortisone (basal serum-free medium, BSFM) failed to show cell-cell communication. However, colonies cultured in BSFM supplemented with prolactin, E(2), and progesterone (complete hormone serum-free medium, CHSFM) showed cell-cell communication at 14 days of primary culture or of subculture. By flow cytometry with FITC-PNA and PE-anti-Thy-1.1 monoclonal antibody, we distinguished four RMEC subpopulations in cultures in both media: Thy-1.1+ cells, PNA+ cells, cells negative to both reagents and cells positive to both reagents. It is likely that combined prolactin, cortisol, and insulin in CHSFM stimulate terminal differentiation of clonogenic cells.

13.
J Biol Chem ; 270(32): 19151-7, 1995 Aug 11.
Article in English | MEDLINE | ID: mdl-7642582

ABSTRACT

Potential signaling substrates for the insulin-like growth factor I (IGF-I) receptor are SH2 domain proteins including the p85 subunit of phosphatidylinositol 3-kinase, the tyrosine phosphatase Syp, GTPase activating protein (GAP), and phospholipase C-gamma (PLC-gamma). In this study, we demonstrate an association between the IGF-I receptor and p85, Syp, and GAP, but not with PLC-gamma in lysates of cells overexpressing the human IGF-I receptor. We further investigated these interactions using glutathione S-transferase (GST) fusion proteins containing the amino-terminal SH2 domains of p85 or GAP, or both SH2 domains of Syp or PLC-gamma to precipitate the IGF-I receptor from purified receptor preparations and from whole cell lysates. p85-, Syp-, and GAP-GSTs precipitated the IGF-I receptor, whereas the PLC-gamma-GST did not. Using phosphopeptides corresponding to IGF-I receptor phosphorylation sites, we determined that the p85- and Syp-GST association with the IGF-I receptor could be inhibited by a carboxyl-terminal peptide containing pY1316 and that the GAP-GST association could be inhibited by a NPXY domain peptide. The GAP-GST binding site was confirmed by showing that a mutant IGF-I receptor with a deletion of the NPXY domain including tyrosine 950 was poorly precipitated by the GAP-GST. We conclude that p85 and Syp may bind directly to the IGF-I receptor at tyrosine 1316, and that GAP may bind to the IGF-I receptor at and PLC-gamma was not evident. p85, Syp, and GAP are potential modulators of IGF-I receptor signal transduction.


Subject(s)
Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Tyrosine Phosphatases/metabolism , Proteins/metabolism , Receptor, IGF Type 1/metabolism , Amino Acid Sequence , Animals , Arsenicals/pharmacology , Binding Sites , CHO Cells , Cricetinae , GTPase-Activating Proteins , Insulin-Like Growth Factor I/pharmacology , Intracellular Signaling Peptides and Proteins , Molecular Sequence Data , Phosphatidylinositol 3-Kinases , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Protein Tyrosine Phosphatase, Non-Receptor Type 6 , SH2 Domain-Containing Protein Tyrosine Phosphatases , Type C Phospholipases/metabolism , Tyrosine/metabolism
14.
Stem Cells ; 13(4): 380-92, 1995 Jul.
Article in English | MEDLINE | ID: mdl-7549897

ABSTRACT

Protein-tyrosine kinases (PTKs) are of vital importance in a variety of cell functions. Recent studies have provided considerable insight into the binding of growth factors to tyrosine kinase receptors and the consequent induction of signal pathways that lead to a biologic response. Future studies will further delineate the signals that result in a proliferative response and those that induce a differentiation response. Current studies, reviewed here, indicate an important biologic role for PTKs in the regulation of megakaryocyte development and maturation. Whether PTKs function in megakaryocytes in signaling pathways that are similar to pathways in other cells will need to be examined in future studies.


Subject(s)
Hematopoiesis/physiology , Megakaryocytes/enzymology , Protein-Tyrosine Kinases/metabolism , Animals , Cell Differentiation , Cell Division , Cytokines/genetics , Cytoplasm/enzymology , Gene Expression Regulation , Humans , Megakaryocytes/cytology , Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction
15.
Biochemistry ; 34(25): 7996-8004, 1995 Jun 27.
Article in English | MEDLINE | ID: mdl-7540866

ABSTRACT

We examined the signal transduction pathway leading to insulin stimulation of two immediate early genes, c-fos, and the early growth response gene, Egr-1. In Rat 1 fibroblasts overexpressing normal human insulin receptors (HIRc-B), insulin and IGF-I rapidly and transiently induced the expression of both c-fos and Egr-1 mRNA with maximum accumulation at 30 min, declining to basal levels at 120 min. Insulin (100 ng/mL) increased c-fos and Egr-1 mRNA expression 10-fold (EC50 = 20 ng/mL), whereas IGF-I (100 ng/mL) and serum (20%) led to a 3- and 11.5-fold increase, respectively. Insulin-stimulated c-fos protein expression was maximal at 1 h postinduction and undetectable at 4 h. The effects of insulin and IGF-I on both c-fos mRNA and protein expression were absent in Rat 1 fibroblasts expressing tyrosine kinase-defective human insulin receptors (A/K1018). In cells expressing insulin receptors in which the two C-terminal tyrosines are mutated to phenylalanine (Y/F2 cells), the insulin stimulated increase in Egr-1 and c-fos mRNA was comparable to that of HIRc cells, whereas, in cells expressing C-terminal truncated receptors (delta CT cells), the insulin induced increase in Egr-1 mRNA was normal, but the c-fos mRNA response was severely blunted. As expected, the insulin effect to increase ras GTP formation and MAP kinase activity was negligible in A/K1018 cells but normal, or supernormal, in Y/F2 cells. Importantly, stimulation of ras GTP was increased in delta CT cells, whereas stimulation of MAP kinase activity was almost absent.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Gene Expression Regulation/drug effects , Genes, Immediate-Early/drug effects , Immediate-Early Proteins , Insulin/pharmacology , Signal Transduction , Animals , Base Sequence , Blotting, Northern , Cell Line , DNA-Binding Proteins/genetics , Early Growth Response Protein 1 , Fibroblasts/metabolism , Genes, fos , Guanosine Triphosphate/metabolism , Kinetics , Molecular Sequence Data , Polymerase Chain Reaction , Proto-Oncogene Proteins p21(ras)/metabolism , RNA, Messenger , RNA-Directed DNA Polymerase , Rats , Transcription Factors/genetics , Transcriptional Activation
16.
J Biol Chem ; 270(16): 9661-6, 1995 Apr 21.
Article in English | MEDLINE | ID: mdl-7536744

ABSTRACT

We have cloned a protein tyrosine kinase, MATK, which is expressed abundantly in megakaryocytes and the brain. We investigated whether MATK participates in the c-Kit ligand/stem cell factor (KL/SCF) signaling pathway in the megakaryocytic cell line CMK. After KL/SCF stimulation, five major proteins of molecular masses of 145, 113, 92, 76, and 63 kDa were rapidly and transiently tyrosine-phosphorylated in a time-dependent manner, peaking within 5 min, and returning to basal levels within 60 min. To study the role of MATK in the KL/SCF signaling pathway, glutathione S-transferase (GST) fusion proteins containing SH2 and SH3 domains of MATK were cloned, expressed in Escherichia coli, and purified. MATK-SH2, but not MATK-SH3, precipitated the tyrosine-phosphorylated c-Kit (molecular mass of 145 kDa) in KL/SCF-stimulated CMK cells. Other GST fusion proteins containing the SH2 domain of p85 of phosphatidylinositol 3-kinase, phospholipase C gamma-1, and ras-GAP also precipitated c-Kit. The tyrosine-phosphorylated c-Kit was co-immunoprecipitated with anti-MATK and anti-p85 antibodies in KL/SCF-stimulated CMK cells, but not in granulocyte-macrophage colony stimulating factor or interleukin-6-stimulated cells, suggesting receptor specificity. These results indicate that MATK associates with the c-Kit receptor following specific stimulation by KL/SCF via its SH2 domain and likely participates in transduction of growth signals induced by this cytokine in megakaryocytes.


Subject(s)
Megakaryocytes/enzymology , Protein-Tyrosine Kinases/physiology , Proto-Oncogene Proteins pp60(c-src) , Proto-Oncogene Proteins/physiology , Receptor Protein-Tyrosine Kinases/physiology , Receptors, Colony-Stimulating Factor/physiology , GTPase-Activating Proteins , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Hematopoietic Cell Growth Factors/pharmacology , Humans , Interleukin-6/pharmacology , Proteins/physiology , Proto-Oncogene Proteins c-kit , Stem Cell Factor , ras GTPase-Activating Proteins
17.
J Biol Chem ; 270(8): 3938-43, 1995 Feb 24.
Article in English | MEDLINE | ID: mdl-7876140

ABSTRACT

GLUT4, the major insulin-responsive glucose transporter isoform in rat adipocytes, rapidly recycles between the cell surface and an intracellular pool with two first order rate constants, one for internalization (kin) and the other for externalization (kex). Insulin decreases kin by 2.8-fold and increases kex by 3.3-fold, thus increasing the steady-state cell surface GLUT4 level by approximately 8-fold (Jhun, B. H., Rampal, A. L., Liu, H., Lachaal, M., and Jung, C. (1992) J. Biol. Chem. 267, 17710-17715). To gain an insight into the biochemical mechanisms that modulate these rate constants, we studied the effects upon them of okadaic acid (OKA), a phosphatase inhibitor that exerts a insulin-like effect on glucose transport in adipocytes. OKA stimulated 3-O-methylglucose transport maximally 3.1-fold and increased the cell surface GLUT4 level 3.4-fold. When adipocytes were pulse-labeled with an impermeant, covalently reactive glucose analog, [3H]1,3-bis-(3-deoxy-D-glucopyranose-3-yloxy)-2-propyl 4-benzoylbenzoate, and the time course of labeled GLUT4 recycling was followed, the kex was found to increase 2.8-fold upon maximal stimulation by OKA, whereas the kin remained unchanged within experimental error. These findings demonstrate that OKA mimics the insulin effect on only GLUT4 externalization and suggest that insulin stimulates GLUT4 externalization by increasing the phosphorylation state of a serine/threonine phosphoprotein, probably by inhibiting protein phosphatase 1 or 2A.


Subject(s)
Adipocytes/drug effects , Ethers, Cyclic/pharmacology , Glucose/metabolism , Monosaccharide Transport Proteins/metabolism , Muscle Proteins , Adipocytes/metabolism , Animals , Biological Transport , Blotting, Western , Cell Membrane/drug effects , Cell Membrane/metabolism , Cells, Cultured , Glucose Transporter Type 4 , Insulin/pharmacology , Kinetics , Okadaic Acid , Phosphorylation , Rats , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism
18.
Mol Cell Biol ; 14(11): 7466-75, 1994 Nov.
Article in English | MEDLINE | ID: mdl-7935461

ABSTRACT

We have investigated the functional role of the SH2 domain of the 85-kDa subunit (p85) of the phosphatidylinositol 3-kinase in the insulin signal transduction pathway. Microinjection of a bacterial fusion protein containing the N-terminal SH2 domain of p85 inhibited insulin- and other growth factor-induced DNA synthesis by 90% and c-fos protein expression by 80% in insulin-responsive rat fibroblasts. The specificity of the fusion protein was examined by in vitro precipitation experiments, which showed that the SH2 domain of p85 can independently associate with both insulin receptor substrate 1 and the insulin receptor itself in the absence of detectable binding to other phosphoproteins. The microinjection results were confirmed through the use of an affinity-purified antibody directed against p85, which gave the same phenotype. Additional studies were carried out in another cell line expressing mutant insulin receptors which lack the cytoplasmic tyrosine residues with which p85 interacts. Microinjection of the SH2 domain fusion protein also inhibited insulin signaling in these cells, suggesting that association of p85 with insulin receptor substrate 1 is a key element in insulin-mediated cell cycle progression. In addition, coinjection of purified p21ras protein with the p85 fusion protein or the antibody restored DNA synthesis, suggesting that ras function is either downstream or independent of p85 SH2 domain interaction.


Subject(s)
DNA/biosynthesis , Genes, fos/drug effects , Insulin/pharmacology , Phosphotransferases (Alcohol Group Acceptor)/pharmacology , Animals , Antibodies/administration & dosage , Cell Line , Humans , Insulin Receptor Substrate Proteins , Microinjections , Molecular Weight , Phosphatidylinositol 3-Kinases , Phosphoproteins/metabolism , Phosphotransferases (Alcohol Group Acceptor)/administration & dosage , Phosphotransferases (Alcohol Group Acceptor)/chemistry , Protein Conformation , Proto-Oncogene Proteins c-fos/biosynthesis , Proto-Oncogene Proteins p21(ras)/administration & dosage , Rats , Receptor, Insulin/metabolism , Recombinant Fusion Proteins/administration & dosage , Signal Transduction/drug effects
19.
J Biol Chem ; 269(18): 13689-94, 1994 May 06.
Article in English | MEDLINE | ID: mdl-7513704

ABSTRACT

Shc proteins contain a single SH2 domain, lack catalytic activity, and are substrates for activated receptors for insulin, insulin-like growth factor-1 (IGF-1), and epidermal growth factor (EGF). Treatment with these growth factors induced rapid tyrosine phosphorylation of Shc. We investigated the potential role of Shc in mitogenic signaling. Affinity-purified antibodies were microinjected into living Rat1 fibroblasts overexpressing human insulin receptors. Bromodeoxyuridine incorporation into newly synthesized DNA was subsequently studied to assess the importance of Shc. Cellular microinjection of anti-Shc antibody inhibited BrdU incorporation induced by insulin, IGF-1, and EGF, but did not affect cells stimulated by fetal calf serum. Microinjection of an oncogenic p21ras protein (T24) into quiescent cells produced constitutively active mitogenic signaling, and comicroinjection of T24 with the anti-Shc antibody restored insulin and EGF stimulation of DNA synthesis. Immunoprecipitates of Shc from lysates of insulin-stimulated cells removed 70-80% of guanine nucleotide-releasing factor activity. These results indicate that Shc is an important component in a mitogenic signal transduction pathway that is shared by insulin, IGF-1, and EGF. The functional locus of Shc is either upstream of p21ras or lies on a distinct branch of the pathway leading to cell cycle progression.


Subject(s)
Epidermal Growth Factor/pharmacology , Insulin-Like Growth Factor I/pharmacology , Insulin/pharmacology , Mitogens/pharmacology , Proteins/physiology , Proto-Oncogene Proteins pp60(c-src) , Signal Transduction , Animals , Cell Line , Humans , Phosphorylation , Proteins/chemistry , Rats , Receptor, Insulin/genetics , Transfection
20.
J Biol Chem ; 269(8): 5699-704, 1994 Feb 25.
Article in English | MEDLINE | ID: mdl-8119907

ABSTRACT

We have investigated the role of cellular p21ras protein in insulin and insulin-like growth factor-I (IGF-I) signaling pathways. Insulin stimulation increased Ras-GTP formation in Rat-1 fibroblasts overexpressing normal human insulin receptors (HIRc-B), far greater than in parental Rat-1 fibroblasts, indicating that competent insulin receptors mediate this response. Cellular microinjection of a dominant-negative mutant p21ras protein (N17 ras) or anti-p21ras monoclonal antibody (Y13-259) into HIRc-B cells reduced insulin- and IGF-I-stimulated DNA synthesis by 75-90%. Insulin-induced c-fos protein expression was also inhibited by 74%. Microinjection of oncogenic p21ras (T-24 ras) into HIRc-B cells activated the mitogenic pathway, and coinjection of N17 ras and T-24 ras showed that oncogenic p21ras rescued the cells from the N17 ras blockade. This later finding indicates that T-24 ras acts downstream of N17 ras. In conclusion, 1) microinjection of a dominant interferring ras mutant into quiescent cells abrogated subsequent insulin and IGF-I mitogenic signaling; 2) oncogenic ras protein rescued cells from the N17 ras blockade, indicating that T24 ras action is downstream of the site of N17 inhibition; and 3) p21ras is an intermediate signaling molecule in the insulin/IGF-I signal transduction pathway and is required for gene expression and DNA synthesis.


Subject(s)
Insulin-Like Growth Factor I/metabolism , Insulin/metabolism , Oncogene Protein p21(ras)/physiology , Signal Transduction , Animals , Antibodies/pharmacology , Cell Line , DNA/biosynthesis , Guanosine Triphosphate/metabolism , Humans , Microinjections , Oncogene Protein p21(ras)/immunology , Proto-Oncogene Proteins c-fos/biosynthesis , Rats
SELECTION OF CITATIONS
SEARCH DETAIL