Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Oncogene ; 29(16): 2449-56, 2010 Apr 22.
Article in English | MEDLINE | ID: mdl-20118982

ABSTRACT

Cutaneous malignant melanoma is considered one of the most deadly human cancers, based on both its penchant for metastatic spread and its typical resistance to currently available therapy. Long known to harbor oncogenic NRAS mutations, melanomas were more recently reported to be frequent bearers of activating mutations in BRAF, one of the effectors situated downstream of wild-type NRAS. NRAS and BRAF mutations are rarely found in the same melanoma, suggesting that they may possess important overlapping oncogenic activities. Here, we compare and contrast the oncogenic roles of the three major NRas downstream effectors, Raf, phosphatidylinositol 3-kinase (PI3K) and Ral guanine exchange factor (RalGEF), using genetically engineered Arf-deficient immortalized mouse melanocytes as a model system. Although no single downstream pathway could recapitulate all of the consequences of oncogenic NRas expression, our data indicate a prominent role for BRaf and PI3K in melanocyte senescence and invasiveness, respectively. More surprisingly, we discovered that constitutive RalGEF activation had a major impact on several malignant phenotypes, particularly anchorage-independent growth, indicating that this often overlooked pathway should be more carefully evaluated as a possible therapeutic target.


Subject(s)
Cell Transformation, Neoplastic , Genes, ras/physiology , Melanoma/etiology , ral Guanine Nucleotide Exchange Factor/physiology , Animals , Cyclin-Dependent Kinase Inhibitor p16/physiology , Mice , PTEN Phosphohydrolase/physiology , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins B-raf/physiology , Proto-Oncogene Proteins c-raf/physiology , Signal Transduction
2.
Blood ; 112(1): 56-63, 2008 Jul 01.
Article in English | MEDLINE | ID: mdl-18417737

ABSTRACT

The small GTP-binding protein Ral has been implicated in regulated exocytosis via its interaction with the mammalian exocyst complex. We have previously demonstrated that Ral is involved in exocytosis of Weibel-Palade bodies (WPBs). Little is known about intracellular signaling pathways that promote activation of Ral in response to ligand binding of G protein-coupled receptors. Here we show that RNAi-mediated knockdown of RalGDS, an exchange factor for Ral, results in inhibition of thrombin- and epinephrine-induced exocytosis of WPBs, while overexpression of RalGDS promotes exocytosis of WPBs. A RalGDS variant lacking its exchange domain behaves in a dominant negative manner by blocking release of WPBs. We also provide evidence that RalGDS binds calmodulin (CaM) via an amino-terminal CaM-binding domain. RalGDS association to CaM is required for Ral activation because a cell-permeable peptide comprising this RalGDS CaM-binding domain inhibits Ral activation and WPB exocytosis. Together our findings suggest that RalGDS plays a vital role in the regulation of Ral-dependent WPB exocytosis after stimulation with Ca(2+)- or cAMP-raising agonists.


Subject(s)
Exocytosis/physiology , Weibel-Palade Bodies/physiology , ral Guanine Nucleotide Exchange Factor/physiology , Amino Acid Sequence , Binding Sites/genetics , Calmodulin/metabolism , Cell Line , Endothelial Cells/drug effects , Endothelial Cells/physiology , Endothelial Cells/ultrastructure , Epinephrine/pharmacology , Exocytosis/drug effects , Genetic Variation , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Models, Molecular , Molecular Sequence Data , RNA Interference , RNA, Small Interfering/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction , Thrombin/pharmacology , Transfection , ral GTP-Binding Proteins/metabolism , ral Guanine Nucleotide Exchange Factor/antagonists & inhibitors , ral Guanine Nucleotide Exchange Factor/chemistry , ral Guanine Nucleotide Exchange Factor/genetics
3.
Mol Cell Biol ; 28(9): 2851-9, 2008 May.
Article in English | MEDLINE | ID: mdl-18285454

ABSTRACT

The Akt kinase is a key regulator of cell proliferation and survival. It is activated in part by PDK1-induced phosphorylation. Here we show that RalGDS, a Ras effector protein that activates Ral GTPases, has a second function that promotes Akt phosphorylation by PDK1 by bringing these two kinases together. In support of this conclusion is our finding that suppression of RalGDS expression in cells inhibits both epidermal growth factor and insulin-induced phosphorylation of Akt. Moreover, while PDK1 complexes with N-GDS, Akt complexes with the central region of RalGDS through an intermediary, JIP1. The biological significance of this newly discovered RalGDS function is highlighted by the observation that an N-terminally deleted mutant of RalGDS that retains the ability to activate Ral proteins but loses the ability to activate Akt also fails to promote cell proliferation. Thus, RalGDS forms a nexus that transduces growth factor signaling to both Ral GTPase and Akt-mediated signaling cascades.


Subject(s)
Proto-Oncogene Proteins c-akt/metabolism , ral Guanine Nucleotide Exchange Factor/physiology , Animals , Cell Line , Cell Proliferation , Enzyme Activation , Epidermal Growth Factor/metabolism , Humans , Insulin/metabolism , Mice , Mutation , Phosphorylation , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Signal Transduction , ral Guanine Nucleotide Exchange Factor/genetics
4.
Methods Enzymol ; 407: 115-28, 2006.
Article in English | MEDLINE | ID: mdl-16757319

ABSTRACT

The Ras superfamily of GTP-binding proteins is involved in many cellular processes, including cell proliferation, movement, and morphology. One such member, Ral GTPase, activates downstream signaling molecules after a conversion to the active state on GTP binding. The RalGDS-related (Rgr) oncogene belongs to the RalGDS family of guanine nucleotide exchange factors (GEFs). RalGEFs activate Ral by stimulating the dissociation of GDP, allowing the binding of GTP and the initiation of downstream signaling events by Ral effectors. Rgr was first identified as a fusion between the rabbit homolog of the Rad 23 gene and the Rgr gene in a rabbit squamous cell carcinoma. The Rgr portion of the fusion was demonstrated to contain the oncogenic activity. The human form of the Rgr oncogene was identified recently, and expression was detected in human T-cell malignancies. This chapter describes the analysis of rabbit and human Rgr function using various methods. These assays may be used for the study of oncogene function in other systems.


Subject(s)
Oncogenes/physiology , ral Guanine Nucleotide Exchange Factor/physiology , Animals , Humans , Leukocytes, Mononuclear/metabolism , Mice , Mice, Transgenic , NIH 3T3 Cells , Rabbits , Signal Transduction , T-Lymphocytes/metabolism , ras Proteins/metabolism
5.
Methods Enzymol ; 407: 648-60, 2006.
Article in English | MEDLINE | ID: mdl-16757359

ABSTRACT

Activation of Ras oncogene by point mutations is an early frequent event in thyroid tumorigenesis. In this chapter, we describe the use of human primary thyroid follicular epithelial cells expressing oncogenic mutant Ras by means of retroviral transduction as a biological model of human cancer initiation that provides powerful insights into thyroid tumorigenesis. We describe protocols for manipulating primary epithelial cells and describe the use of this model to dissect the signaling pathways required for Ras-induced proliferation in these cells. We also highlight the importance of studying Ras signaling in an appropriate cell context, summarizing some of the key differences identified between more widespread experimental models based on fibroblasts or rodent cell lines and primary epithelial cells.


Subject(s)
Cell Transformation, Neoplastic , Thyroid Gland/cytology , ras Proteins/physiology , Epithelial Cells/pathology , Genetic Vectors , Humans , Mitogen-Activated Protein Kinases/physiology , Models, Biological , Oncogene Protein p21(ras)/genetics , Oncogene Protein p21(ras)/physiology , Phosphatidylinositol 3-Kinases/physiology , Retroviridae/genetics , Thyroid Gland/pathology , raf Kinases/physiology , ral Guanine Nucleotide Exchange Factor/physiology , ras Proteins/genetics
6.
Mol Cell Biol ; 26(10): 3966-75, 2006 May.
Article in English | MEDLINE | ID: mdl-16648489

ABSTRACT

Hyperactivation of Ras is one of the most common abnormalities in acute myeloid leukemia. In experimental models, Ras inhibits myeloid differentiation, which is characteristic of leukemia; however, the mechanism through which it disrupts hematopoiesis is poorly understood. In multipotent FDCP-mix cells, Ras inhibits terminal neutrophil differentiation, thereby indefinitely extending their proliferative potential. Ras also strongly promotes the sensitivity of these cells to granulocyte-macrophage colony-stimulating factor (GM-CSF). Using this model, we have dissected the signaling elements downstream of Ras to determine their relative contribution to the dysregulation of hematopoiesis. Cells expressing Ras mutants selectively activating Raf (Ras*T35S) or phosphatidylinositol 3-kinase (Ras*Y40C) did not significantly affect differentiation or proliferative capacity, whereas Ras*E37G (which selectively activates RalGEFs) perpetuated proliferation and blocked neutrophil development in a manner similar to that of Ras. Correspondingly, expression of constitutively active versions of these effectors confirmed the overriding importance of Ral guanine nucleotide exchange factors. Cells expressing Ras demonstrated hyperactivation of Ral, which itself was able to exactly mimic the phenotype of Ras, including hypersensitivity to GM-CSF. Conversely, dominant negative Ral promoted spontaneous neutrophil development. Ral, in turn, appears to influence differentiation through multiple effectors. These data show, for the first time, the importance of Ral in regulating differentiation and self-renewal in hematopoietic cells.


Subject(s)
Cell Differentiation , Genes, ras , Leukemia, Myeloid/physiopathology , ral Guanine Nucleotide Exchange Factor/physiology , ras Proteins/physiology , Animals , Blotting, Western , Cell Culture Techniques , Cell Line, Tumor , Cell Proliferation , Clone Cells , Dose-Response Relationship, Drug , Enzyme Activation , Gene Expression Regulation, Leukemic , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Leukemia, Myeloid/genetics , Mice , Mutation , Neutrophils/cytology , Neutrophils/metabolism , Retroviridae/genetics , ral Guanine Nucleotide Exchange Factor/genetics
8.
Development ; 131(5): 1055-64, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14973286

ABSTRACT

The phytohormone gibberellin (GA) regulates the development and fertility of Arabidopsis flowers. The mature flowers of GA-deficient mutant plants typically exhibit reduced elongation growth of petals and stamens. In addition, GA-deficiency blocks anther development, resulting in male sterility. Previous analyses have shown that GA promotes the elongation of plant organs by opposing the function of the DELLA proteins, a family of nuclear growth repressors. However, it was not clear that the DELLA proteins are involved in the GA-regulation of stamen and anther development. We show that GA regulates cell elongation rather than cell division during Arabidopsis stamen filament elongation. In addition, GA regulates the cellular developmental pathway of anthers leading from microspore to mature pollen grain. Genetic analysis shows that the Arabidopsis DELLA proteins RGA and RGL2 jointly repress petal, stamen and anther development in GA-deficient plants, and that this function is enhanced by RGL1 activity. GA thus promotes Arabidopsis petal, stamen and anther development by opposing the function of the DELLA proteins RGA, RGL1 and RGL2.


Subject(s)
Arabidopsis Proteins/antagonists & inhibitors , Arabidopsis/growth & development , Gibberellins/metabolism , Arabidopsis/genetics , Arabidopsis Proteins/genetics , Arabidopsis Proteins/physiology , Flowers/growth & development , Genes, Plant , Gibberellins/genetics , Microscopy, Electron, Scanning , Mitosis , Mutation , Phenotype , Plant Growth Regulators/genetics , Plant Growth Regulators/physiology , Plant Proteins , Pollen/growth & development , Transcription Factors/genetics , Transcription Factors/physiology , ral Guanine Nucleotide Exchange Factor/genetics , ral Guanine Nucleotide Exchange Factor/physiology
9.
Hypertension ; 41(4): 956-62, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12642511

ABSTRACT

Ras-related GTPase (Ral) is converted to the GTP-bound form by Ral GDP dissociation stimulator (Ral-GDS), a putative effector protein of Ras. Although a number of studies indicate that Ras induces cardiac hypertrophy, the functional role of Ral-GDS/Ral signaling pathway is as yet unknown in cardiac myocytes. We investigated the role of the Ral-GDS/Ral pathway in cardiac hypertrophy. Transfection of Ral-GDS and constitutively active mutant of Ral (RalG23V) in cultured rat neonatal myocytes stimulated promoter activity of c-fos (5.4-fold and 2.6-fold, P<0.01), alpha-skeletal actin (2.7-fold and 2.1-fold, P<0.01), and beta-myosin heavy chain-luciferase (2.8-fold and 2.3-fold, P<0.01). Ral-GDS-induced or RalG23V-induced promoter activation was increased synergistically with activated Ras (RasG12V). Dominant-negative mutant of Ral (RalS28N) partially inhibited RasG12V induced promoter activation. Cardiac myocytes transfected with RalG23V showed increased cell size compared with nontransfected or vector-transfected cells (2.1-fold, P<0.01). Cardiotrophin-1 (CT-1) upregulated Ral-GDS mRNA expression and induced Ral activation. CT-1-induced Ral-GDS mRNA expression was inhibited by overexpression of the dominant-negative mutant of STAT3. Moreover, Ral activity was elevated in hypertrophied hearts (2.1-fold, P<0.01) by mechanical stress in association with increased CT-1 expression and signal transducer and activator of transcription 3 (STAT3) phosphorylation in the rat aortic banding model. Ral-GDS/Ral pathway is involved in a wide range of gene expressions and is activated by hypertrophic stimuli in vitro and in vivo. SATA3 may play a key role in Ral-GDS expression and Ral activation. Our data provide evidence that the Ral-GDS/Ral signaling pathway is a link to the process of cardiac hypertrophy.


Subject(s)
Cardiomegaly/etiology , ral GTP-Binding Proteins/physiology , ral Guanine Nucleotide Exchange Factor/physiology , Actins/genetics , Animals , Cardiomegaly/metabolism , Cells, Cultured , Cytokines/pharmacology , Mutation , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myosin Heavy Chains/genetics , Promoter Regions, Genetic , Proto-Oncogene Proteins c-fos/genetics , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Transcriptional Activation , Transfection , ral GTP-Binding Proteins/genetics , ral Guanine Nucleotide Exchange Factor/genetics , ras Proteins/metabolism
10.
Int J Oncol ; 21(3): 477-85, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12168089

ABSTRACT

Transformation by ras oncogenes induces the deregulation of intracellular signalling cascades that are critical elements in cell growth control. Ras proteins are molecular switches with the ability to interact and activate several effector molecules. Among those, Raf-1 kinase, PI3K and Ral-GDS are the best characterised. Raf activates the mitogenic MEK/ERK kinases pathway, while PI3K regulates the PKB/Akt cascade, involved in the control of proliferation, metabolism and apoptotic responses. Finally, Ral-GDS belongs to a family of guanine nucleotide exchange factors that activate Ral GTPases. While Raf and PI3K have emerged as critical elements in regulating cell growth and apoptosis, little is known about the role of the Ral-GDS family. We have previously reported that Ras proteins are critical elements in the regulation of phospholipase D (PLD), a proposed target for the Ral-GDS/RalA pathway. Physiological regulation of PLD by growth factors requires the simultaneous activation of the endogenous, wild-type Ras proteins, and a PKC-dependent mechanism. Transformation by ras oncogenes induces drastic alterations in PLD activity and the usual response to external stimuli, through a PKC-independent mechanism. Here we provide further evidence on the mechanisms by which oncogenic Ras proteins induces the deregulation of PLD and here we try to identify the specific effectors involved. A complex system for PLD regulation is unravelled which implies the existence of two positive regulatory pathways, mediated by Ral-GDS and PI3K, and two negative feedback mechanisms mediated by Raf and Ral-GDS. These results strongly support participation of PLD in Ras-mediated signalling. Furthermore, we provide evidence that oncogenic Ras proteins constitutively activate PLD by mechanisms different to those used by normal Ras proteins.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Phorbol 12,13-Dibutyrate/pharmacology , Phosphatidylinositol 3-Kinases/physiology , Phospholipase D/metabolism , Proto-Oncogene Proteins c-raf/physiology , ral Guanine Nucleotide Exchange Factor/physiology , ras Proteins/physiology , 3T3 Cells , Animals , Cell Transformation, Neoplastic/genetics , Cells, Cultured , Enzyme Activation , Fibroblasts/cytology , Fibroblasts/enzymology , Fibroblasts/physiology , Mice , Mutation , Platelet-Derived Growth Factor/pharmacology , Rats , Transfection , ras Proteins/genetics
11.
Oncogene ; 21(6): 937-46, 2002 Jan 31.
Article in English | MEDLINE | ID: mdl-11840339

ABSTRACT

Ras proteins are molecular switches that control signaling pathways critical in the onset of a variety of human cancers. The signaling pathways activated by Ras proteins are those controlled by its direct effectors such as the serine-threonine protein kinase Raf-1, the exchange factor for other GTPases Ral-GDS, and the lipid kinase PI3K. As a consequence of Ras activation, a number of additional enzymes are affected, including several members of the serine-threonine intracellular proteins kinases as well as enzymes related to phospholipid metabolism regulation such as phospholipases A2 and D, and choline kinase. The precise mechanisms by which ras oncogenes impinge into these later molecules and their relevance to the onset of the carcinogenic process is still not fully understood. Here we have investigated the mechanism of regulation of choline kinase by Ras proteins and found no direct link between PLD and choline kinase activation. We provide evidence that Ras proteins regulate the activity of choline kinase through its direct effectors Ral-GDS and PI3K, while the Raf pathways seems to be not relevant in this process. The importance of Ras-dependent activation of choline kinase is discussed.


Subject(s)
Choline Kinase/biosynthesis , ral Guanine Nucleotide Exchange Factor/physiology , ras Proteins/physiology , 3T3 Cells/metabolism , Amino Acid Substitution , Animals , Cell Line , Cell Transformation, Neoplastic/genetics , Choline Kinase/genetics , Embryo, Mammalian , Enzyme Activation , Enzyme Induction , Genes, ras , Guanosine Triphosphate/physiology , Humans , Isoenzymes/physiology , Kidney/cytology , Mice , Mutation, Missense , Phospholipase D/genetics , Phospholipase D/physiology , Phosphorylcholine/metabolism , Protein Structure, Tertiary , Proto-Oncogene Proteins c-raf/physiology , Proto-Oncogene Proteins p21(ras)/chemistry , Proto-Oncogene Proteins p21(ras)/physiology , Recombinant Fusion Proteins/physiology , Second Messenger Systems , Transfection
12.
Curr Opin Genet Dev ; 12(1): 44-6, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11790553

ABSTRACT

It is well known that upregulation of Ras activity can promote cell-cycle progression. Now recent studies indicate that a reciprocal relationship also exists; that is, the consequences of Ras signaling are dependent upon cell-cycle position. In quiescent cells stimulated with growth factors, one Ras effector, phosphatidylinositol-3-kinase, is activated twice as cells transition from G(0) into G(1) phase, and then later in G(1) phase. It is only during the later stages of G(1) phase that PI3K activity promotes entry into S-phase. In cycling cells, Ras activity is enhanced throughout the cell cycle, but is able to stimulate cyclin D1 elevation only during G(2) phase.


Subject(s)
Cell Cycle/physiology , Signal Transduction/physiology , ras Proteins/physiology , Animals , Cyclin D1/metabolism , G1 Phase , Gene Expression Regulation , Humans , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-raf/physiology , Resting Phase, Cell Cycle , S Phase , ral Guanine Nucleotide Exchange Factor/physiology , ras Proteins/metabolism
13.
Oncogene ; 20(50): 7334-41, 2001 Nov 01.
Article in English | MEDLINE | ID: mdl-11704863

ABSTRACT

Ras mutations occur at high frequency in thyroid cancer. In vitro, the effects of Ras in thyroid cells are pleiotropic in that expression of activated Ras has been reported to stimulate proliferation and apoptosis. An understanding of the factors that contribute to the survival versus demise of Ras-transformed cells is essential to our understanding of the contribution of Ras to thyroid neoplasia and other cancers. Constitutive expression of oncogenic H-Ras sensitized Wistar rat thyroid (WRT) cells to apoptosis stimulated by multiple insults. When deprived of matrix attachment, Ras-transformed cells perished by apoptotic cell death at a high frequency. In contrast, parental cells were more resistant to suspension-induced cell death. Ras effects on anchorage-independent cell death were reproduced by a mutant protein that signals selectively to Raf-1, but not by mutant Ras that preferentially binds to RalGDS. Expression of a Ras mutant that selectively activates PI3K resulted in substantial protection from detachment-induced cell death. MAPK activity was increased in adherent Ras12V- and Ras12V35S-expressing cells, but abolished upon detachment. Interestingly, impaired MAPK activity was sufficient to stimulate apoptosis in adherent Ras-transformed cells, but not in parental cells. Treatment with a PI3K inhibitor also stimulated apoptosis selectively in Ras-transformed cells. These results demonstrate that constitutive expression of activated Ras elicits differential effects on the survival of thyroid cells. Moreover, Ras expression results in a greater dependence of thyroid cells on MAPK and PI3K activity for their survival.


Subject(s)
Apoptosis/genetics , Cell Transformation, Neoplastic/genetics , Genes, ras , Proto-Oncogene Proteins p21(ras)/physiology , Thyroid Gland/cytology , Amino Acid Substitution , Animals , Cell Adhesion , Cell Culture Techniques/methods , Cell Line, Transformed/cytology , Culture Media, Serum-Free , Enzyme Activation , Enzyme Inhibitors/pharmacology , Epithelial Cells/cytology , Humans , MAP Kinase Signaling System , Mutagenesis, Site-Directed , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Point Mutation , Proto-Oncogene Proteins c-raf/physiology , Rats , Rats, Wistar , Suspensions , ral Guanine Nucleotide Exchange Factor/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...