Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
Add more filters










Publication year range
1.
Oncogene ; 31(43): 4630-8, 2012 Oct 25.
Article in English | MEDLINE | ID: mdl-22231449

ABSTRACT

While Ras is well-known to function on the plasma membrane (PM) to mediate growth factor signaling, increasing evidence suggests that Ras has complex roles in the cytoplasm. To uncover these roles, we screened a cDNA library and isolated H-Ras-binding proteins that also influence Ras functions. Many isolated proteins regulate trafficking involving endosomes; CHMP6/VPS20 and VPS4A, which interact with ESCRT-III (Endosomal Sorting Complex Required for Transport-III), were chosen for further study. We showed that the binding is direct and occurs in endosomes. Furthermore, the binding is most efficient when H-Ras has a functional effector-binding loop, and is GTP-bound and ubiquitylated. CHMP6 and VPS4A also bound to N-Ras but not K-Ras. Repressing CHMP6 and VPS4A blocked Ras-induced transformation, which correlated with inefficient Ras localization to the PM as measured by cell fractionation and photobleaching. Moreover, silencing CHMP6 and VPS4A also blocked epidermal growth factor receptor (EGFR) recycling. These data suggest that Ras interacts with key ESCRT-III components to promote recycling of itself and EGFR back to the PM to create a positive feedback loop to enhance growth factor signaling.


Subject(s)
Endosomal Sorting Complexes Required for Transport/physiology , Genes, ras , Intercellular Signaling Peptides and Proteins/metabolism , Signal Transduction , Vacuolar Proton-Translocating ATPases/physiology , ATPases Associated with Diverse Cellular Activities , Cell Membrane/metabolism , ErbB Receptors/metabolism , Humans , Ubiquitination
2.
Gene Ther ; 10(2): 163-71, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12571645

ABSTRACT

Intratracheal (IT) injection of manganese superoxide dismutase-plasmid/liposome (MnSOD-PL) complexes prior to whole lung irradiation of C57BL/6J mice provides significant protection from acute and chronic irradiation damage. We determined the duration of increased MnSOD biochemical activity and differential expression of a hemagglutinin (HA) epitope-tagged MnSOD transgene. HA-MnSOD-PL was IT injected at doses of 0-1000 microg, and mice were killed 1,2,3 or 4 days later. Other groups of mice were irradiated to 20 Gy to the pulmonary cavity 24 h after injection and killed at the same time points as non-irradiated mice. Both non-irradiated and irradiated groups of mice showed increased MnSOD biochemical activity with plasmid dose that plateaued at 100 microg of MnSOD plasmid DNA. In control mice, MnSOD biochemical activity decreased at 2, 3 or 4 days after injection. In irradiated mice, MnSOD biochemical activity decreased at day 2 but increased on days 3 and 4. HA-MnSOD expression decreased in broncheoalveolar macrophages and alveolar type-II cells 3 days after injection in non-irradiated and irradiated mice, but remained elevated in endothelial and epithelial cells past 4 days. The data provide a rationale for every second-day administration of intrapulmonary MnSOD-PL in clinical trials of radioprotective gene therapy. This should be sufficient to provide radioprotection during radiation treatments.


Subject(s)
Genetic Therapy/methods , Lung/enzymology , Lung/radiation effects , Radiation Injuries, Experimental/prevention & control , Radiation-Protective Agents , Superoxide Dismutase/genetics , Animals , Endothelium, Vascular/enzymology , Epithelial Cells/enzymology , Gene Expression , Hemagglutinins/genetics , Injections , Liposomes , Macrophages, Alveolar/enzymology , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Pulmonary Alveoli/enzymology , Radiation Injuries, Experimental/enzymology , Time Factors
3.
Mol Cell ; 8(4): 759-69, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11684012

ABSTRACT

The regulation of protein tyrosine phosphatase (PTPase) SHP-2 is proposed to involve tyrosine phosphorylation on two tail tyrosine residues. Using "expressed protein ligation", nonhydrolyzable phosphotyrosine analogs were introduced at known phosphorylation sites in SHP-2. Biochemical analysis suggests that a phosphonate at Tyr542 interacts intramolecularly with the N-terminal SH2 domain to relieve basal inhibition of the PTPase, whereas a phosphonate at Tyr-580 stimulates the PTPase activity by interaction with the C-terminal SH2 domain. Microinjection experiments indicate that a single phosphorylation of Tyr-542 of SHP-2 is sufficient to activate the MAP kinase pathway in living cells. These studies support a novel mechanism explaining how tyrosine phosphorylation of a PTPase is important in signal transduction.


Subject(s)
Adaptor Proteins, Signal Transducing , Phosphotyrosine/metabolism , Protein Tyrosine Phosphatases/metabolism , Signal Transduction/physiology , Amino Acid Sequence , Culture Media, Serum-Free , Enzyme Activation , GRB2 Adaptor Protein , Intracellular Signaling Peptides and Proteins , Microinjections , Microscopy, Fluorescence , Molecular Sequence Data , Molecular Structure , Phosphorylation , Phosphotyrosine/chemistry , Point Mutation , Protein Conformation , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Protein Tyrosine Phosphatase, Non-Receptor Type 6 , Protein Tyrosine Phosphatases/chemistry , Protein Tyrosine Phosphatases/genetics , Proteins/genetics , Proteins/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , SH2 Domain-Containing Protein Tyrosine Phosphatases , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Subtilisin/metabolism
4.
Int J Cancer ; 96(4): 221-31, 2001 Aug 20.
Article in English | MEDLINE | ID: mdl-11474496

ABSTRACT

Intraesophageal administration of manganese superoxide dismutase-plasmid/liposome (MnSOD-PL) prior to single fraction radiation has been shown to protect mice from lethal esophagitis. In our study, C3H/HeNsd mice received fractionated radiation in two protocols: (i) 18 Gy daily for four days with MnSOD-PL administration 24 hr prior to the first and third fraction, or (ii) 12 Gy daily for six days with MnSOD-PL 24 hr prior to the first, third, and fifth fraction. Control radiated mice received either no liposomes only or LacZ (bacterial beta-galactosidase gene)-plasmid/liposome (LacZ-PL) by the same schedules. We measured thiol depletion and lipid peroxidation (LP) in whole esophagus and tested the effectiveness of a new plasmid, hemagglutinin (HA) epitope-tagged MnSOD (HA-MnSOD). In fractionation protocols, mice receiving MnSOD-PL, but not LacZ-PL (200 microl of plasmid/liposomes containing 200 microg of plasmid DNA), showed a significant reduction in morbidity, decreased weight loss, and improved survival. Four and seven days after 37 Gy single fraction radiation, the esophagus demonstrated a significant increase in peroxidized lipids and reduction in overall antioxidant levels, reduced thiols, and decreased glutathione (GSH). These reductions were modulated by MnSOD-PL administration. The HA-MnSOD plasmid product was detected in the basal layers of the esophageal epithelium 24 hr after administration and provided significant radiation protection compared to glutathione peroxidase-plasmid/liposome (GPX-PL), or liposomes containing MnSOD protein, vitamin E, co-enzyme Q10, or 21-aminosteroid. Thus, MnSOD-PL administration significantly improved tolerance to fractionated radiation and modulated radiation effects on levels of GSH and lipid peroxidation (LP). These studies provide further support for translation of MnSOD-PL treatment into human esophageal radiation protection.


Subject(s)
Esophagitis/etiology , Esophagitis/prevention & control , Liposomes/therapeutic use , Plasmids/therapeutic use , Superoxide Dismutase/therapeutic use , Animals , Biomarkers , Cells, Cultured , Chromatography, High Pressure Liquid , Dinoprost/metabolism , Dose-Response Relationship, Drug , Epitopes/metabolism , Fatty Acids, Unsaturated/metabolism , Female , Hemagglutinins/metabolism , Lac Operon , Lipid Metabolism , Lipid Peroxidation/radiation effects , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Radiotherapy/adverse effects , Superoxide Dismutase/chemistry , Time Factors
5.
Proc Natl Acad Sci U S A ; 98(16): 9014-9, 2001 Jul 31.
Article in English | MEDLINE | ID: mdl-11470914

ABSTRACT

The activity of the small GTPase, Rac1, plays a role in various cellular processes including cytoskeletal rearrangement, gene transcription, and malignant transformation. In this report constitutively active Rac1 (Rac V12) is shown to stimulate the activation of STAT3, a member of the family of signal transducers and activators of transcription (STATs). The activity of Rac1 leads to STAT3 translocation to the nucleus coincident with STAT3-dependent gene expression. The expression of Vav (Delta1-187), a constitutively active guanine nucleotide exchange factor for the Rho GTPases, or activated forms of Ras or Rho family members, leads to STAT3-specific activation. The activation of STAT3 requires tyrosine phosphorylation at residue 705, but is not dependent on phosphorylation of Ser-727. Our studies indicate that Rac1 induces STAT3 activation through an indirect mechanism that involves the autocrine production and action of IL-6, a known mediator of STAT3 response. Rac V12 expression results in the induction of the IL-6 and IL-6 receptor genes and neutralizing antibodies directed against the IL-6 receptor block Rac1-induced STAT3 activation. Furthermore, inhibition of the nuclear factor-kappaB activation or disruption of IL-6-mediated signaling through the expression of IkappaBalpha S32AS36A and suppressor of cytokine signaling 3, respectively, blocks Rac1-induced STAT3 activation. These findings elucidate a mechanism dependent on the induction of an autocrine IL-6 activation loop through which Rac1 mediates STAT3 activation establishing a link between oncogenic GTPase activity and Janus kinase/STAT signaling.


Subject(s)
DNA-Binding Proteins/metabolism , Interleukin-6/physiology , Trans-Activators/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Base Sequence , Cell Line , DNA Primers , GTP Phosphohydrolases/metabolism , Gene Expression , Humans , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor , Signal Transduction
6.
J Biol Chem ; 276(29): 27629-37, 2001 Jul 20.
Article in English | MEDLINE | ID: mdl-11333268

ABSTRACT

Ras GTPases function as binary switches in signaling pathways controlling cell growth and differentiation. The guanine nucleotide exchange factor Sos mediates the activation of Ras in response to extracellular signals. We have previously solved the crystal structure of nucleotide-free Ras in complex with the catalytic domain of Sos (Boriack-Sjodin, P. A., Margarit, S. M., Bar-Sagi, D., and Kuriyan, J. (1998) Nature 394, 337-343). The structure demonstrates that Sos induces conformational changes in two loop regions of Ras known as switch 1 and switch 2. In this study, we have employed site-directed mutagenesis to investigate the functional significance of the conformational changes for the catalytic function of Sos. Switch 2 of Ras is held in a very tight embrace by Sos, with almost every external side chain coordinated by Sos. Mutagenesis of contact residues at the switch 2-Sos interface shows that only a small set of side chains affect binding, with the most important contact being mediated by tyrosine 64, which is buried in a hydrophobic pocket of Sos in the Ras.Sos complex. Substitutions of Ras and Sos side chains that are inserted into the Mg(2+)- and nucleotide phosphate-binding site of switch 2 (Ras Ala(59) and Sos Leu(938) and Glu(942)) have no effect on the catalytic function of Sos. These results indicate that the interaction of Sos with switch 2 is necessary for tight binding, but is not the critical driving force for GDP displacement. The structural distortion of switch 1 induced by Sos is mediated by a small number of specific contacts between highly conserved residues on both Ras and Sos. Mutations of a subset of these residues (Ras Tyr(32) and Tyr(40)) result in an increase in the intrinsic rate of nucleotide dissociation from Ras and impair the binding of Ras to Sos. Based on this analysis, we propose that the interactions of Sos with the switch 1 and switch 2 regions of Ras have distinct functional consequences: the interaction with switch 2 mediates the anchoring of Ras to Sos, whereas the interaction with switch 1 leads to disruption of the nucleotide-binding site and GDP dissociation.


Subject(s)
Guanosine Diphosphate/metabolism , Oncogene Protein p21(ras)/physiology , Amino Acid Substitution , Humans , Magnesium/metabolism , Models, Molecular , Mutagenesis, Site-Directed , Oncogene Protein p21(ras)/chemistry , Oncogene Protein p21(ras)/metabolism , Phosphates/metabolism , Protein Conformation
7.
J Biol Chem ; 276(19): 15609-15, 2001 May 11.
Article in English | MEDLINE | ID: mdl-11278702

ABSTRACT

Ras proteins are key regulators of cell growth and differentiation. Mammalian cells express three closely related Ras proteins: Ha-Ras, K-Ras, and N-Ras. We have compared the abilities of the Ha-Ras and K-Ras isoforms to activate the Rac effector pathway, using three Rac-dependent readouts: induction of membrane ruffling and pinocytosis, stimulation of cell motility, and Pak binding. The total surface area of membrane ruffles induced by K-RasV12 was 2-fold greater than that induced by Ha-RasV12. Likewise, the number of K-RasV12-induced pinocytic vesicles per cell was approximately 2-fold greater than that induced by Ha-RasV12. In a wound healing assay, K-RasV12-injected cells migrated twice as fast as Ha-RasV12-injected cells. Moreover, the Pak binding activity of Rac, which is indicative of the amount of GTP-bound Rac, was higher in K-RasV12-expressing cells than Ha-RasV12-expressing cells. These results suggest that K-Ras activates Rac more efficiently than Ha-Ras. The preferential activation of Rac by K-Ras is dependent on the mode of membrane anchoring and impacts on the ability of K-Ras to regulate cell survival.


Subject(s)
Cell Membrane/physiology , Cell Membrane/ultrastructure , rac GTP-Binding Proteins/metabolism , ras Proteins/metabolism , Animals , Apoptosis , COS Cells , Cell Line , Chlorocebus aethiops , Humans , Mammals , Pinocytosis , Recombinant Proteins/metabolism , Transfection
11.
Science ; 288(5467): 870-4, 2000 May 05.
Article in English | MEDLINE | ID: mdl-10797012

ABSTRACT

The c-Jun NH2-terminal kinase (JNK) is activated when cells are exposed to ultraviolet (UV) radiation. However, the functional consequence of JNK activation in UV-irradiated cells has not been established. It is shown here that JNK is required for UV-induced apoptosis in primary murine embryonic fibroblasts. Fibroblasts with simultaneous targeted disruptions of all the functional Jnk genes were protected against UV-stimulated apoptosis. The absence of JNK caused a defect in the mitochondrial death signaling pathway, including the failure to release cytochrome c. These data indicate that mitochondria are influenced by proapoptotic signal transduction through the JNK pathway.


Subject(s)
Apoptosis , Cytochrome c Group/metabolism , Mitogen-Activated Protein Kinases/metabolism , Protein Serine-Threonine Kinases , Animals , Apoptotic Protease-Activating Factor 1 , Caspase 3 , Caspase 9 , Caspases/metabolism , Cell Count , Cell Division , Cells, Cultured , DNA Fragmentation , Enzyme Activation , Fibroblasts , Gene Targeting , JNK Mitogen-Activated Protein Kinases , MAP Kinase Signaling System , Methyl Methanesulfonate/pharmacology , Mice , Mitochondria/metabolism , Mitogen-Activated Protein Kinases/genetics , NF-kappa B/metabolism , Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-bcl-2/metabolism , Tumor Suppressor Protein p53/metabolism , Ultraviolet Rays
12.
J Virol ; 74(5): 2084-93, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10666238

ABSTRACT

The adenovirus type 5 (Ad5) E4-6/7 protein interacts directly with different members of the E2F family and mediates the cooperative and stable binding of E2F to a unique pair of binding sites in the Ad5 E2a promoter region. This induction of E2F DNA binding activity strongly correlates with increased E2a transcription when analyzed using virus infection and transient expression assays. Here we show that while different adenovirus isolates express an E4-6/7 protein that is capable of induction of E2F dimerization and stable DNA binding to the Ad5 E2a promoter region, not all of these viruses carry the inverted E2F binding site targets in their E2a promoter regions. The Ad12 and Ad40 E2a promoter regions bind E2F via a single binding site. However, these promoters bind adenovirus-induced (dimerized) E2F very weakly. The Ad3 E2a promoter region binds E2F very poorly, even via a single binding site. A possible explanation of these results is that the Ad E4-6/7 protein evolved to induce cellular gene expression. Consistent with this notion, we show that infection with different adenovirus isolates induces the binding of E2F to an inverted configuration of binding sites present in the cellular E2F-1 promoter. Transient expression of the E4-6/7 protein alone in uninfected cells is sufficient to induce transactivation of the E2F-1 promoter linked to chloramphenicol acetyltransferase or green fluorescent protein reporter genes. Further, expression of the E4-6/7 protein in the context of adenovirus infection induces E2F-1 protein accumulation. Thus, the induction of E2F binding to the E2F-1 promoter by the E4-6/7 protein observed in vitro correlates with transactivation of E2F-1 promoter activity in vivo. These results suggest that adenovirus has evolved two distinct mechanisms to induce the expression of the E2F-1 gene. The E1A proteins displace repressors of E2F activity (the Rb family members) and thus relieve E2F-1 promoter repression; the E4-6/7 protein complements this function by stably recruiting active E2F to the E2F-1 promoter to transactivate expression.


Subject(s)
Adenoviridae/metabolism , Adenovirus E4 Proteins/pharmacology , Carrier Proteins , Cell Cycle Proteins , Promoter Regions, Genetic , Transcription Factors/genetics , Adenoviridae/genetics , Amino Acid Sequence , Binding Sites , Cell Line , Cloning, Molecular , DNA/metabolism , DNA-Binding Proteins/genetics , E2F Transcription Factors , E2F1 Transcription Factor , Electrophoresis, Agar Gel , Gene Expression Regulation, Viral/drug effects , HeLa Cells , Humans , Immunohistochemistry , Molecular Sequence Data , Recombinant Proteins/biosynthesis , Retinoblastoma-Binding Protein 1 , Sequence Alignment , Transcription Factor DP1 , Transcription Factors/analysis , Transcription Factors/biosynthesis
13.
Nat Cell Biol ; 1(1): 20-6, 1999 May.
Article in English | MEDLINE | ID: mdl-10559859

ABSTRACT

The Ink4/Arf locus encodes two tumour-suppressor proteins, p16Ink4a and p19Arf, that govern the antiproliferative functions of the retinoblastoma and p53 proteins, respectively. Here we show that Arf binds to the product of the Mdm2 gene and sequesters it into the nucleolus, thereby preventing negative-feedback regulation of p53 by Mdm2 and leading to the activation of p53 in the nucleoplasm. Arf and Mdm2 co-localize in the nucleolus in response to activation of the oncoprotein Myc and as mouse fibroblasts undergo replicative senescence. These topological interactions of Arf and Mdm2 point towards a new mechanism for p53 activation.


Subject(s)
ADP-Ribosylation Factors/metabolism , Cell Nucleolus/metabolism , Nuclear Proteins , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , 3T3 Cells , ADP-Ribosylation Factors/genetics , Animals , Cell Nucleolus/ultrastructure , Cellular Senescence , Feedback , Fibroblasts/cytology , Fibroblasts/physiology , Gene Expression Regulation , Genes, myc , Genes, p53 , Mice , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-mdm2 , Recombinant Proteins/metabolism , Transfection
14.
Mol Cell Biol ; 19(9): 5892-901, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10454536

ABSTRACT

Ras is an essential component of signal transduction pathways that control cell proliferation, differentiation, and survival. In this study we have examined the cellular responses to high-intensity Ras signaling. Expression of increasing amounts of the oncogenic form of human HRas, HRasV12, results in a dose-dependent induction of apoptosis in both primary and immortalized cells. The induction of apoptosis by HRasV12 is blocked by activated Rac and potentiated by dominant interfering Rac. The ability of Rac to suppress Ras-induced apoptosis is dependent on effector pathway(s) controlled by the insert region and is linked to the activation of NF-kappaB. The apoptotic effect of HRasV12 requires the activation of both the ERK and JNK mitogen-activated protein kinase cascade and is independent of p53. These results demonstrate a role for Rac in controlling signals that are necessary for cell survival, and suggest a mechanism by which Rac activity can confer growth advantage to cells transformed by the ras oncogene.


Subject(s)
Apoptosis/physiology , Proteins/physiology , ras Proteins/physiology , Animals , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Cycle Proteins/metabolism , Cell Line , Cell Survival/physiology , GTP-Binding Proteins/metabolism , GTPase-Activating Proteins , Gene Expression , Growth Substances/physiology , Humans , Mice , NF-kappa B/metabolism , Plasmids/genetics , Proteins/genetics , Rats , Signal Transduction , Tumor Suppressor Protein p53/metabolism , cdc42 GTP-Binding Protein , ras GTPase-Activating Proteins , ras Proteins/genetics
15.
Ann N Y Acad Sci ; 886: 122-31, 1999.
Article in English | MEDLINE | ID: mdl-10667210

ABSTRACT

RAS interacts with multiple targets in the cell and controls at least two signaling pathways, one regulating extracellular signal-regulated kinase (ERK) activation and the other controlling membrane ruffling formation. These two pathways appear to act synergistically to cause transformation. Human smooth muscle alpha-actin promoter is repressed in RAS-transformed cells and derepressed in revertant cell lines, suggesting that it is a sensitive marker to follow phenotypic changes in fibroblast cells. SCH 51344 is a pyrazoloquinoline derivative identified on the basis of its ability to derepress alpha-actin promoter in RAS-transformed cells. Previous studies have shown that SCH 51344 is a potent inhibitor of RAS transformation. However, SCH 51344 had very little effect on the activities of proteins in the ERK pathway, suggesting that it inhibits RAS transformation by a novel mechanism. Recently, we have demonstrated that SCH 51344 specifically blocks membrane ruffling induced by activated forms of H-RAS, K-RAS, N-RAS, and RAC. Treatment of fibroblast cells with this compound had very little effect on RAS-mediated activation of ERK and JUN kinase activities. SCH 51344 was effective in inhibiting the anchorage-independent growth of Rat-2 fibroblast cells transformed by the three forms of oncogenic RAS and RAC V12. These results indicate that SCH 51344 inhibits a critical component of the membrane ruffling pathway downstream from RAC and suggest that targeting this pathway may be an effective approach to inhibiting transformation by RAS and other oncogenes.


Subject(s)
Aminoquinolines/pharmacology , Anticarcinogenic Agents/pharmacology , Pyrazoles/pharmacology , rac GTP-Binding Proteins/antagonists & inhibitors , ras Proteins/antagonists & inhibitors , Actins/genetics , Enzyme Activation , Genes, Reporter , Humans , Promoter Regions, Genetic , Protein Kinases/metabolism
16.
Cell ; 95(2): 259-68, 1998 Oct 16.
Article in English | MEDLINE | ID: mdl-9790532

ABSTRACT

Proteins containing Dbl homology (DH) domains activate Rho-family GTPases by functioning as specific guanine nucleotide exchange factors. All known DH domains have associated C-terminal pleckstrin homology (PH) domains that are implicated in targeting and regulatory functions. The crystal structure of a fragment of the human Son of sevenless protein containing the DH and PH domains has been determined at 2.3 A resolution. The entirely alpha-helical DH domain is unrelated in architecture to other nucleotide exchange factors. The active site of the DH domain, identified on the basis of sequence conservation and structural features, lies near the interface between the DH and PH domains. The structure suggests that ligation of the PH domain will be coupled structurally to the GTPase binding site.


Subject(s)
Blood Proteins/chemistry , Blood Proteins/genetics , Phosphoproteins , Proteins/chemistry , Proteins/genetics , Binding Sites , Crystallography , Guanine Nucleotide Exchange Factors , Humans , Molecular Sequence Data , Protein Structure, Secondary , Protein Structure, Tertiary , Sequence Homology, Amino Acid
17.
Nature ; 394(6691): 337-43, 1998 Jul 23.
Article in English | MEDLINE | ID: mdl-9690470

ABSTRACT

The crystal structure of human H-Ras complexed with the Ras guanine-nucleotide-exchange-factor region of the Son of sevenless (Sos) protein has been determined at 2.8 A resolution. The normally tight interaction of nucleotides with Ras is disrupted by Sos in two ways. First, the insertion into Ras of an alpha-helix from Sos results in the displacement of the Switch 1 region of Ras, opening up the nucleotide-binding site. Second, side chains presented by this helix and by a distorted conformation of the Switch 2 region of Ras alter the chemical environment of the binding site for the phosphate groups of the nucleotide and the associated magnesium ion, so that their binding is no longer favoured. Sos does not impede the binding sites for the base and the ribose of GTP or GDP, so the Ras-Sos complex adopts a structure that allows nucleotide release and rebinding.


Subject(s)
Membrane Proteins/metabolism , ras Proteins/metabolism , Binding Sites , Catalysis , Cloning, Molecular , Crystallography, X-Ray , Escherichia coli , GTP Phosphohydrolase-Linked Elongation Factors/metabolism , Guanine/metabolism , Guanosine Triphosphate/metabolism , Humans , Membrane Proteins/chemistry , Models, Molecular , Molecular Sequence Data , Mutation , Peptide Elongation Factor Tu/chemistry , Peptide Elongation Factor Tu/metabolism , Peptide Elongation Factors/chemistry , Peptide Elongation Factors/metabolism , Protein Binding , Protein Conformation , Son of Sevenless Proteins , Structure-Activity Relationship , ras Proteins/chemistry , ras Proteins/genetics
18.
J Biol Chem ; 273(29): 17991-4, 1998 Jul 17.
Article in English | MEDLINE | ID: mdl-9660749

ABSTRACT

The Rac GTP-binding protein controls signal transduction pathways that are critical for mitogenesis and oncogenesis (1,2). The biochemical nature of these signaling pathways is presently unknown. Here we report that a region in Rac1 (residues 124-135), previously defined as the insert region (3), is essential for its mitogenic activity. Deletion of this region does not interfere with the ability of Rac1 to induce cytoskeletal changes or to activate the Jun kinase mitogen-activated protein kinase cascade but abrogates Rac1-induced stimulation of DNA synthesis and Rac1-mediated superoxide production in quiescent fibroblasts. Treatment of cells with agents that abolish superoxide generation inhibits specifically the mitogenic effect of Rac1. Our results identify an effector site in Rac1 that is necessary for mitogenic signaling and implicate superoxide generation as a candidate effector pathway of Rac1-dependent cell growth.


Subject(s)
GTP-Binding Proteins/metabolism , Mitogen-Activated Protein Kinases , Mitosis , Superoxides/metabolism , Animals , Binding Sites , COS Cells , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , DNA Replication , GTP-Binding Proteins/genetics , JNK Mitogen-Activated Protein Kinases , Mutagenesis, Insertional , NADPH Oxidases/metabolism , Proto-Oncogene Proteins c-raf/metabolism , rac GTP-Binding Proteins
19.
J Biol Chem ; 273(13): 7743-8, 1998 Mar 27.
Article in English | MEDLINE | ID: mdl-9516483

ABSTRACT

Kinase suppressor of Ras (KSR) is a loss-of-function allele that suppresses the rough eye phenotype of activated Ras in Drosophila and the multivulval phenotype of activated Ras in Caenorhabditis elegans. Genetic and biochemical studies suggest that KSR is a positive regulator of Ras signaling that functions between Ras and Raf or in a pathway parallel to Raf. We examined the effect of mammalian KSR expression on the activation of extracellular ligand-regulated (ERK) mitogen-activated protein (MAP) kinase in fibroblasts. Ectopic expression of KSR inhibited the activation of ERK MAP kinase by insulin, phorbol ester, or activated alleles of Ras, Raf, and mitogen and extracellular-regulated kinase. Expression of deletion mutants of KSR demonstrated that the KSR kinase domain was necessary and sufficient for the inhibitory effect of KSR on ERK MAP kinase activity. KSR inhibited cell transformation by activated RasVal-12 but had no effect on the ability of RasVal-12 to induce membrane ruffling. These data indicate that KSR is a potent modulator of a signaling pathway essential to normal and oncogenic cell growth and development.


Subject(s)
Growth Substances/metabolism , Mitogen-Activated Protein Kinase Kinases , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Signal Transduction , ras Proteins/metabolism , 3T3 Cells , Animals , Caenorhabditis elegans , Cells, Cultured , Drosophila , Enzyme Activation , Insulin/pharmacology , MAP Kinase Kinase 1 , Mice , Tetradecanoylphorbol Acetate/pharmacology
20.
Mol Cell Biol ; 18(2): 880-6, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9447984

ABSTRACT

The guanine nucleotide exchange factor Sos mediates the coupling of receptor tyrosine kinases to Ras activation. To investigate the mechanisms that control Sos activity, we have analyzed the contribution of various domains to its catalytic activity. Using human Sos1 (hSos1) truncation mutants, we show that Sos proteins lacking either the amino or the carboxyl terminus domain, or both, display a guanine nucleotide exchange activity that is significantly higher compared with that of the full-length protein. These results demonstrate that both the amino and the carboxyl terminus domains of Sos are involved in the negative regulation of its catalytic activity. Furthermore, in vitro Ras binding experiments suggest that the amino and carboxyl terminus domains exert negative allosteric control on the interaction of the Sos catalytic domain with Ras. The guanine nucleotide exchange activity of hSos1 was not augmented by growth factor stimulation, indicating that Sos activity is constitutively maintained in a downregulated state. Deletion of both the amino and the carboxyl terminus domains was sufficient to activate the transforming potential of Sos. These findings suggest a novel negative regulatory role for the amino terminus domain of Sos and indicate a cooperation between the amino and the carboxyl terminus domains in the regulation of Sos activity.


Subject(s)
Eukaryotic Initiation Factor-2/physiology , Proteins/physiology , Animals , COS Cells , Catalysis , ErbB Receptors/metabolism , Eukaryotic Initiation Factor-2/genetics , Growth Substances/metabolism , Guanine Nucleotide Exchange Factors , Humans , Mutagenesis, Site-Directed , Proteins/genetics , Signal Transduction , Structure-Activity Relationship , ras Guanine Nucleotide Exchange Factors , ras Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...