Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
2.
Mol Cell Biol ; 20(22): 8480-8, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11046144

ABSTRACT

The transcription factor c-Jun is critically involved in the regulation of proliferation and differentiation as well as cellular transformation induced by oncogenic Ras. The signal transduction pathways that couple Ras activation to c-Jun phosphorylation are still partially elusive. Here we show that an activated version of the Ras effector Rlf, a guanine nucleotide exchange factor (GEF) of the small GTPase Ral, can induce the phosphorylation of serines 63 and 73 of c-Jun. In addition, we show that growth factor-induced, Ras-mediated phosphorylation of c-Jun is abolished by inhibitory mutants of the RalGEF-Ral pathway. These results suggest that the RalGEF-Ral pathway plays a major role in Ras-dependent c-Jun phosphorylation. Ral-dependent regulation of c-Jun phosphorylation includes JNK, a still elusive JNKK, and possibly Src.


Subject(s)
Mitogen-Activated Protein Kinases/metabolism , ral Guanine Nucleotide Exchange Factor/metabolism , ras Proteins/metabolism , Animals , Cell Differentiation/drug effects , Cell Division/drug effects , Cell Line/drug effects , Enzyme Activation , Guanine Nucleotide Exchange Factors , Humans , Insulin/metabolism , Insulin/pharmacology , JNK Mitogen-Activated Protein Kinases , MAP Kinase Kinase 4 , Mice , Mitogen-Activated Protein Kinase Kinases/metabolism , Mutation , Phosphorylation , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Serine , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism , ral GTP-Binding Proteins/metabolism , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism
3.
J Biol Chem ; 275(38): 29761-6, 2000 Sep 22.
Article in English | MEDLINE | ID: mdl-10889189

ABSTRACT

Ral is a ubiquitously expressed Ras-like small GTPase. Several guanine nucleotide exchange factors for Ral have been identified, including members of the RalGDS family, which exhibit a Ras binding domain and are regulated by binding to RasGTP. Here we describe a novel type of RalGEF, RalGEF2. This guanine nucleotide exchange factor has a characteristic Cdc25-like catalytic domain at the N terminus and a pleckstrin homology (PH) domain at the C terminus. RalGEF2 is able to activate Ral both in vivo and in vitro. Deletion of the PH domain results in an increased cytoplasmic localization of the protein and a corresponding reduction in activity in vivo, suggesting that the PH domain functions as a membrane anchor necessary for optimal activity in vivo.


Subject(s)
ral GTP-Binding Proteins/metabolism , ral Guanine Nucleotide Exchange Factor/analysis , ral Guanine Nucleotide Exchange Factor/metabolism , Amino Acid Sequence , Animals , Blood Proteins/genetics , Blood Proteins/metabolism , COS Cells , Enzyme Activation , Molecular Sequence Data , Phosphoproteins/genetics , Phosphoproteins/metabolism , Sequence Alignment , Substrate Specificity , ral Guanine Nucleotide Exchange Factor/genetics
4.
J Cell Biol ; 148(6): 1151-8, 2000 Mar 20.
Article in English | MEDLINE | ID: mdl-10725328

ABSTRACT

Integrin-mediated leukocyte adhesion is a critical aspect of leukocyte function that is tightly regulated by diverse stimuli, including chemokines, antigen receptors, and adhesion receptors. How cellular signals from CD31 and other adhesion amplifiers are integrated with those from classical mitogenic stimuli to regulate leukocyte function remains poorly understood. Here, we show that the cytoplasmic tail of CD31, an important integrin adhesion amplifier, propagates signals that induce T cell adhesion via beta1 (VLA-4) and beta2 (LFA-1) integrins. We identify the small GTPase, Rap1, as a critical mediator of this effect. Importantly, CD31 selectively activated the small Ras-related GTPase, Rap1, but not Ras, R-Ras, or Rap2. An activated Rap1 mutant stimulated T lymphocyte adhesion to intercellular adhesion molecule (ICAM) and vascular cell adhesion molecule (VCAM), as did the Rap1 guanine nucleotide exchange factor C3G and a catalytically inactive mutant of RapGAP. Conversely, negative regulators of Rap1 signaling blocked CD31-dependent adhesion. These findings identify a novel important role for Rap1 in regulating ligand-induced cell adhesion and suggest that Rap1 may play a more general role in coordinating adhesion-dependent signals during leukocyte migration and extravasation. Our findings also suggest an alternative mechanism, distinct from interference with Ras-proximal signaling, by which Rap1 might mediate transformation reversion.


Subject(s)
Cell Adhesion Molecules/metabolism , Cell Adhesion/physiology , Integrins/physiology , Lymphocyte Function-Associated Antigen-1/physiology , Platelet Endothelial Cell Adhesion Molecule-1/physiology , Receptors, Lymphocyte Homing/physiology , rap1 GTP-Binding Proteins/metabolism , Antigens, CD/physiology , Humans , Integrin alpha4beta1 , Jurkat Cells , Platelet Endothelial Cell Adhesion Molecule-1/chemistry , Recombinant Fusion Proteins/metabolism , Signal Transduction , T-Lymphocytes/physiology , Transfection , Vascular Cell Adhesion Molecule-1/metabolism
5.
Oncogene ; 18(31): 4435-9, 1999 Aug 05.
Article in English | MEDLINE | ID: mdl-10442634

ABSTRACT

Previous work by us and others has implicated a role for Ral guanine exchange factors (RalGEFs) in Ras-induced cell growth and oncogenic transformation. Here we show for the first time that RalGEFs are involved in Ras-induced differentiation as well. Expression of oncogenic Ras in F9 embryonal carcinoma (EC) cells is known to induce differentiation to a primitive endoderm (PrE)-like phenotype, but the downstream signal transduction mechanisms involved are unclear. We found that PrE differentiation is induced by the Ras effector domain mutants, RasV12G37 and RasV12E38, but not by RasV12C40. Accordingly, expression of constitutively active forms of RalGEF (Rlf-CAAX) or Rafl (Raf-CAAX) is sufficient to induce differentiation. Inhibition of RalGEF activity by expression of dominant negative Ral completely abolishes Rlf-CAAX- and RasV12G37-induced differentiation, while it reduces differentiation by RasV12 and Raf-CAAX. Finally, while Rlf-CAAX does not increase Erk activity, inhibition of MEK blocks both Ras- as well as Rlf-CAAX-induced differentiation, suggesting that RalGEFs induce PrE differentiation in a manner depending on basal MEK or Erk activity. Based on these results we conclude that Ras induces PrE differentiation of F9 EC cells via an interplay of Erk-and RalGEF-mediated pathways.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Endoderm/cytology , GTP-Binding Proteins/metabolism , Protein Kinases/metabolism , Proto-Oncogene Proteins c-raf , ras Proteins/metabolism , Amino Acid Substitution , Animals , Carcinoma, Embryonal , Cell Differentiation , Enzyme Activation , Mutagenesis, Site-Directed , Phenotype , Recombinant Fusion Proteins/metabolism , Retroviridae Proteins, Oncogenic/metabolism , Signal Transduction , Transfection , Tumor Cells, Cultured , rap GTP-Binding Proteins , ras Proteins/genetics
6.
J Biol Chem ; 274(31): 21847-52, 1999 Jul 30.
Article in English | MEDLINE | ID: mdl-10419502

ABSTRACT

We have measured the activation of the small GTPase Ral in human neutrophils after stimulation with fMet-Leu-Phe (fMLP), platelet activating factor (PAF), and granulocyte macrophage-colony stimulating factor and compared it with the activation of two other small GTPases, Ras and Rap1. We found that fMLP and PAF, but not granulocyte macrophage-colony stimulating factor, induce Ral activation. All three stimuli induce the activation of both Ras and Rap1. Utilizing specific inhibitors we demonstrate that fMLP-induced Ral activation is mediated by pertussis toxin-sensitive G-proteins and partially by Src-like kinases, whereas fMLP-induced Ras activation is independent of Src-like kinases. PAF-induced Ral activation is mediated by pertussis toxin-insensitive proteins, Src-like kinases and phosphatidylinositol 3-kinase. Phosphatidylinositol 3-kinase is not involved in PAF-induced Ras activation. The calcium ionophore ionomycin activates Ral, but calcium depletion partially inhibits fMLP- and PAF-induced Ral activation, whereas Ras activation was not affected. In addition, 12-O-tetradecanoylphorbol-13-acetate-induced activation of Ral is completely abolished by inhibitors of protein kinase C, whereas 12-O-tetradecanoylphorbol-13-acetate-induced Ras activation is largely insensitive. We conclude that in neutrophils Ral activation is mediated by multiple pathways, and that fMLP and PAF induce Ral activation differently.


Subject(s)
GTP-Binding Proteins/blood , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophils/physiology , Platelet Activating Factor/pharmacology , Signal Transduction/physiology , Calcium/blood , Calcium-Calmodulin-Dependent Protein Kinases/blood , Enzyme Activation , GTP Phosphohydrolases/blood , GTPase-Activating Proteins , Humans , In Vitro Techniques , Kinetics , Mitogen-Activated Protein Kinase 1 , Neutrophils/drug effects , Phosphorylation , Protein Kinase C/blood , Proteins/metabolism , Signal Transduction/drug effects , Tetradecanoylphorbol Acetate/pharmacology , ral GTP-Binding Proteins , rap GTP-Binding Proteins , ras GTPase-Activating Proteins
7.
J Biol Chem ; 274(13): 8737-45, 1999 Mar 26.
Article in English | MEDLINE | ID: mdl-10085114

ABSTRACT

In search for effectors of the Ras-related GTPase Rap2, we used the yeast two-hybrid method and identified the C-terminal Ras/Rap interaction domain of the Ral exchange factors (RalGEFs) Ral GDP dissociation stimulator (RalGDS), RalGDS-like (RGL), and RalGDS-like factor (Rlf). These proteins, which also interact with activated Ras and Rap1, are effectors of Ras and mediate the activation of Ral in response to the activation of Ras. Here we show that the full-length RalGEFs interact with the GTP-bound form of Rap2 in the two-hybrid system as well as in vitro. When co-transfected in HeLa cells, an activated Rap2 mutant (Rap2Val-12) but not an inactive protein (Rap2Ala-35) co-immunoprecipitates with RalGDS and Rlf; moreover, Rap2-RalGEF complexes can be isolated from the particulate fraction of transfected cells and were localized by confocal microscopy to the resident compartment of Rap2, i.e. the endoplasmic reticulum. However, the overexpression of activated Rap2 neither leads to the activation of the Ral GTPase via RalGEFs nor inhibits Ras-dependent Ral activation in vivo. Several hypotheses that could explain these results, including compartmentalization of proteins involved in signal transduction, are discussed. Our results suggest that in cells, the interaction of Rap2 with RalGEFs might trigger other cellular responses than activation of the Ral GTPase.


Subject(s)
GTP-Binding Proteins/genetics , Animals , Fluorescent Antibody Technique , GTP Phosphohydrolases/genetics , Gene Expression Regulation/genetics , HeLa Cells , Humans , Mice , Mutation/genetics , Precipitin Tests , Protein Binding/genetics , Signal Transduction/genetics , Transfection , ral Guanine Nucleotide Exchange Factor , rap GTP-Binding Proteins
8.
Curr Opin Genet Dev ; 9(1): 112-7, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10072355

ABSTRACT

The Ral guanine nucleotide exchange factors are direct targets of Ras, providing a mechanism for Ral activation by extracellular signals. In addition, Ral can be activated by a Ras-independent pathway. Ral guanine nucleotide exchange factors contribute to cellular transformation induced by oncogenic Ras through an Erk-independent mechanism which may involve activation of transcription.


Subject(s)
ras Proteins/physiology , Enzyme Activation , GTP Phosphohydrolases/metabolism , GTP-Binding Proteins/metabolism , Guanine Nucleotide Exchange Factors , Guanosine Triphosphate/metabolism , Humans , Proteins/metabolism , ral GTP-Binding Proteins , rap GTP-Binding Proteins , ras Guanine Nucleotide Exchange Factors
9.
J Biol Chem ; 274(3): 1487-94, 1999 Jan 15.
Article in English | MEDLINE | ID: mdl-9880524

ABSTRACT

The formation of parietal endoderm (PE) is one of the first differentiation processes during mouse development and can be studied in vitro using F9 embryonal carcinoma (EC) cells. Treatment of F9 EC cells with retinoic acid (RA) induces differentiation toward primitive endoderm (PrE), while differentiation toward PE is induced by subsequent addition of parathyroid hormone (PTH) or PTH-related peptide (PTHrP). The signal transduction mechanisms involved in this two-step process are largely unclear. We show that the RA-induced differentiation toward PrE is accompanied by a sustained increase in Ras activity and that ectopic expression of oncogenic Ha-Ras is sufficient to induce PrE differentiation. Ras activity subsequently decreases upon PTH-induced differentiation toward PE. This is a necessary event, since expression of oncogenic Ha-Ras in PrE-like cells prevents PTH-induced PE differentiation. Expression of active PKA in PrE-like F9 cells mimics PTH-induced PE differentiation and is again prevented by oncogenic Ha-Ras. The effect of oncogenic Ras on both differentiation steps is abolished by the MEK inhibitor PD98059 and can be mimicked by constitutively active forms of Raf and MEK. In conclusion, our data suggest that activation of the Ras/Erk is sufficient to induce differentiation to PrE and to prevent subsequent differentiation toward PE. Activation of PKA down-regulates Ras activity, resulting in disappearance of this blockade and transmission of signal(s) triggering PE differentiation.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/physiology , Endoderm/physiology , ras Proteins/physiology , Animals , Carcinoma, Embryonal/pathology , Cell Differentiation/drug effects , Cell Transformation, Neoplastic , Cyclic AMP-Dependent Protein Kinases/metabolism , Down-Regulation , Enzyme Activation , Mice , Parathyroid Hormone/pharmacology , Rats , Tretinoin/pharmacology , Tumor Cells, Cultured
10.
EMBO J ; 17(20): 5905-12, 1998 Oct 15.
Article in English | MEDLINE | ID: mdl-9774335

ABSTRACT

The small GTPase Rap1 has been implicated in both negative and positive control of Ras-mediated signalling events. We have investigated which extracellular signals can activate Rap1 and whether this activation leads to a modulation of Ras effector signalling, i.e. the activation of ERK and the small GTPase Ral. We found that Rap1 is rapidly activated following stimulation of a large variety of growth factor receptors. These receptors include receptor tyrosine kinases for platelet-derived growth factor (PDGF) and epithelial growth factor (EGF), and G protein-coupled receptors for lysophosphatidic acid (LPA), thrombin and endothelin. At least three distinct pathways may transduce a signal towards Rap1 activation: increase in intracellular calcium, release of diacylglycerol and cAMP synthesis. Surprisingly, activation of endogenous Rap1 fails to affect Ras-dependent ERK activation. In addition, we found that although overexpression of active Rap1 is able to activate the Ral pathway, activation of endogenous Rap1 in fibroblasts does not result in Ral activation. Rap1 also does not negatively influence Ras-mediated Ral activation. We conclude that activation of Rap1 is a common event upon growth factor treatment and that the physiological function of Rap1 is likely to be different from modulation of Ras effector signalling.


Subject(s)
GTP-Binding Proteins/physiology , Proto-Oncogene Proteins/physiology , Signal Transduction/physiology , ras Proteins/physiology , 3T3 Cells/drug effects , 3T3 Cells/metabolism , Animals , Blotting, Western , COS Cells/drug effects , COS Cells/metabolism , Cells, Cultured , Epidermal Growth Factor/pharmacology , Lysophospholipids/pharmacology , Mice , Mitogen-Activated Protein Kinases/metabolism , PC12 Cells/drug effects , PC12 Cells/metabolism , Platelet-Derived Growth Factor/pharmacology , Rats , Type C Phospholipases/drug effects , Type C Phospholipases/physiology , ral GTP-Binding Proteins , rap GTP-Binding Proteins
11.
Biochemistry ; 37(39): 13453-62, 1998 Sep 29.
Article in English | MEDLINE | ID: mdl-9753431

ABSTRACT

Ral-specific guanine nucleotide exchange factors RalGDS, Rgl, and Rlf have been suggested to function as intermediates between Ras and Ral pathways by being able to bind Ras proteins through their C-terminal Ras-binding domains (RBD). The RBDs of RalGDS and of the Ser/Thr kinase c-Raf-1 have been shown to have the same tertiary structure. In contrast to the RBDs of Raf and RalGDS, which bind either Ras or Rap with high affinity, Rlf-RBD has a similar affinity for both GTP-binding proteins. To be able to compare these RBDs on a structural level, we have solved the three-dimensional structure of Rlf-RBD by NMR spectroscopy. The overall tertiary structure of Rlf-RBD shows the betabetaalphabetabetaalphabeta-fold of the ubiquitin superfamily and is very similar to that of RalGDS-RBD. The binding interface of Rlf-RBD to Ras was mapped using chemical shift analysis and indicated a binding mode similar to that in the case of Rap.Raf-RBD. However, comparison of the putatively interacting regions revealed structural differences which are proposed to be responsible for the different substrate affinities of Rlf-, RalGDS-, and Raf-RBD.


Subject(s)
GTP-Binding Proteins/chemistry , Transcription Factors/chemistry , ras Proteins/metabolism , Amino Acid Sequence , Animals , Crystallography, X-Ray , GTP-Binding Proteins/metabolism , Guanine Nucleotide Exchange Factors , Guanylyl Imidodiphosphate/chemistry , Macromolecular Substances , Mice , Models, Molecular , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Protein Folding , Protein Structure, Secondary , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Substrate Specificity , Transcription Factors/metabolism , ral Guanine Nucleotide Exchange Factor , rap GTP-Binding Proteins
12.
Curr Biol ; 8(8): 471-4, 1998 Apr 09.
Article in English | MEDLINE | ID: mdl-9550702

ABSTRACT

The small GTPase Ral is a Ras-like GTPase [1] that has been implicated in growth-factor-induced and Ras-induced DNA synthesis [2-4], and Ras-induced oncogenic transformation [3,5]. Recently, we and others found that three different Ral guanine nucleotide exchange factors (Ral GEFs) - Ral GDS, Rgl and Rlf - bind specifically to the GTP-bound form of several Ras-like GTPases [6-9]. Although oncogenic Ras is able to activate these Ral GEFs [2,5,10], it is unknown whether growth factors can induce the activation of Ral and, if so, which small GTPase is involved in this process. Here, we show that stimulation of various growth factor receptors, including receptor tyrosine kinases and serpentine receptors, results in rapid activation of Ral. This activation correlates with the activation of Ras, and dominant-negative Ras completely inhibits Ral activation induced by insulin and epidermal growth factor (EGF). From these results, we conclude that Ral activation is a direct downstream effect of growth-factor-induced Ras activation.


Subject(s)
GTP Phosphohydrolases/metabolism , GTP-Binding Proteins/metabolism , Genes, ras/physiology , 3T3 Cells , Animals , Cell Line , Enzyme Activation , Epidermal Growth Factor/pharmacology , Fibroblasts , Insulin/pharmacology , Lysophospholipids/pharmacology , Mice , Mutation , Rats , Receptors, Endothelin/metabolism , Receptors, Platelet-Derived Growth Factor/metabolism , Recombinant Fusion Proteins , ral GTP-Binding Proteins
13.
Mol Cell Biol ; 18(5): 2486-91, 1998 May.
Article in English | MEDLINE | ID: mdl-9566869

ABSTRACT

Ral is a ubiquitously expressed Ras-like small GTPase which is abundantly present in human platelets. The biological function of Ral and the signaling pathway in which Ral is involved are largely unknown. Here we describe a novel method to measure Ral activation utilizing the Ral binding domain of the putative Ral effector RLIP76 as an activation-specific probe. With this assay we investigated the signaling pathway that leads to Ral activation in human platelets. We found that Ral is rapidly activated after stimulation with various platelet agonists, including alpha-thrombin. In contrast, the platelet antagonist prostaglandin I2 inhibited alpha-thrombin-induced Ral activation. Activation of Ral by alpha-thrombin could be inhibited by depletion of intracellular Ca2+, whereas the induction of intracellular Ca2+ resulted in the activation of Ral. Our results show that Ral can be activated by extracellular stimuli. Furthermore, we show that increased levels of intracellular Ca2+ are sufficient for Ral activation in platelets. This activation mechanism correlates with the activation mechanism of the small GTPase Rap1, a putative upstream regulator of Ral guanine nucleotide exchange factors.


Subject(s)
ATP-Binding Cassette Transporters , Blood Platelets/enzymology , GTP Phosphohydrolases/metabolism , GTP-Binding Proteins/metabolism , GTPase-Activating Proteins , Platelet Activation/physiology , Calcium/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Enzyme Activation , Epoprostenol/pharmacology , Guanosine Triphosphate/metabolism , Humans , Peptide Fragments/genetics , Peptide Fragments/metabolism , Platelet Activation/drug effects , Protein Binding , Signal Transduction , Thrombin/pharmacology , ral GTP-Binding Proteins , rap GTP-Binding Proteins
14.
EMBO J ; 16(22): 6748-61, 1997 Nov 17.
Article in English | MEDLINE | ID: mdl-9362489

ABSTRACT

Rlf is a ubiquitously expressed distinct relative of RalGDS that interacts with active Ras in vitro. We now demonstrate that Rlf, when co-expressed with Ras mutants, associates in vivo with RasV12 and the effector-domain mutant RasV12G37, but not with RasV12E38 or RasV12C40. Rlf exhibits guanine nucleotide exchange activity towards the small GTPase Ral and, importantly, Rlf-induced Ral activation is stimulated by active Ras. In addition, RasV12 and RasV12G37 synergize with Rlf in the transcriptional activation of the c-fos promoter. Rlf, when targeted to the plasma membrane using the Ras farnesyl attachment site (Rlf-CAAX), is constitutively active, inducing both Ral activation and c-fos promoter activity. Rlf-CAAX-induced gene expression is insensitive to dominant negative Ras and the MEK inhibitor PD98059, and involves activation of the serum response element. Furthermore, expression of Rlf-CAAX is sufficient to induce proliferation of NIH 3T3 cells under low-serum conditions. These data demonstrate that Rlf is an effector of Ras which functions as an exchange factor for Ral. Rlf mediates a distinct Ras-induced signalling pathway to gene induction. Finally, a constitutively active form of Rlf can stimulate transcriptional activation and cell growth.


Subject(s)
Cell Transformation, Neoplastic , Gene Expression Regulation , Mitogen-Activated Protein Kinase Kinases , Proto-Oncogene Proteins c-fos/biosynthesis , Transcription Factors/metabolism , ras Proteins/metabolism , 3T3 Cells , Animals , COS Cells , Cell Compartmentation , Cell Division , Cell Membrane , GTP-Binding Proteins/metabolism , Guanine Nucleotide Exchange Factors , Guanine Nucleotides/metabolism , MAP Kinase Kinase 1 , Mice , Protein Prenylation , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Proteins/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Transcriptional Activation , ral GTP-Binding Proteins , ras Guanine Nucleotide Exchange Factors
15.
Oncogene ; 13(2): 353-62, 1996 Jul 18.
Article in English | MEDLINE | ID: mdl-8710374

ABSTRACT

The small GTPase Rap 1A is a close relative of Ras that, when overexpressed, is able to revert oncogenic transformation induced by active Ras. We screened a mouse embryonic cDNA library using the yeast two-hybrid system and isolated the cDNA of a novel Rap 1A-interacting protein. The open reading frame encodes for an 84 kDa protein with a Cdc25-homology domain which shares approximately 30% identity with Ral guanine nucleotide dissociation stimulator (RalGDS) and RalGDS-like (Rg1). The C-terminal region reveals a striking conservation of sequences with the Ras-binding domain of RalGDS. We designated this protein Rlf, for RalGDS-like factor. In the yeast system, Rlf interacts with Rap 1A, H-Ras and R-Ras, but not with Rac and Rho. In addition, we found that Rlf interacts with Rap 1Aval12 but not with Rap 1AAsn17. In vitro binding studies revealed that a C-terminally located 91 amino acid region of Rlf is sufficient for direct association with the GTP-bound form of Ras and Rap 1A. The observed dissociation constants are 0.6 microM and 0.4 microM, respectively. No significant association with Ras-GDP or Rap 1A-GDP could be detected. These binding characteristics indicate that Rlf is a putative effector for Ras and Rap 1A.


Subject(s)
DNA, Complementary/genetics , DNA, Complementary/isolation & purification , GTP-Binding Proteins/genetics , GTP-Binding Proteins/isolation & purification , Guanine Nucleotide Exchange Factors , Transcription Factors/genetics , Transcription Factors/isolation & purification , ras Proteins/genetics , ras Proteins/isolation & purification , ras-GRF1 , Amino Acid Sequence , Animals , Base Sequence , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cloning, Molecular , Fungal Proteins/genetics , Fungal Proteins/metabolism , GTP-Binding Proteins/metabolism , Guanosine Triphosphate/metabolism , Mice , Molecular Sequence Data , Protein Binding , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins , Sequence Homology, Amino Acid , Transcription Factors/metabolism , ral Guanine Nucleotide Exchange Factor , rap GTP-Binding Proteins , ras Proteins/metabolism
16.
Biochim Biophys Acta ; 1177(2): 160-6, 1993 Jun 06.
Article in English | MEDLINE | ID: mdl-8388736

ABSTRACT

We demonstrate that exposure of human epidermoid carcinoma A431 cells to epidermal growth factor (EGF) results in phosphorylation of eIF-4B within minutes after addition of EGF. The EGF-induced phosphorylation of eIF-4B is not caused by the EGF receptor tyrosine kinase itself, since no tyrosine-phosphorylated eIF-4B could be detected upon immunoprecipitation using an anti-phosphotyrosine antibody. Enhanced phosphorylation of eIF-4B was also detected upon exposure of the cells to phorbol 12-myristate 13-acetate (PMA), an activator of protein kinase C (PKC), suggesting that eIF-4B may be a substrate of PKC. However, down-regulation of PKC did not influence the EGF-induced eIF-4B phosphorylation, which indicates that eIF-4B is phosphorylated by an as yet unknown kinase, activated early in the EGF-induced signal transduction cascade.


Subject(s)
Epidermal Growth Factor/pharmacology , ErbB Receptors/drug effects , Eukaryotic Initiation Factors , Peptide Initiation Factors/metabolism , Protein Kinase C/metabolism , Enzyme Activation , ErbB Receptors/metabolism , Humans , Phosphorylation/drug effects , Precipitin Tests , Protein-Tyrosine Kinases/metabolism , Signal Transduction , Tetradecanoylphorbol Acetate , Time Factors , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...