Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
Add more filters










Publication year range
1.
BMC Plant Biol ; 19(1): 386, 2019 Sep 05.
Article in English | MEDLINE | ID: mdl-31488054

ABSTRACT

BACKGROUND: Consumers purchase fresh strawberries all year long. Extending the fruiting season for new strawberry cultivars is a common breeding goal. Understanding the inheritance of repeat fruiting is key to improving breeding efficiency. Several independent research groups using multiple genotypes and analytic approaches have all identified a single genomic region in strawberry associated with repeat fruiting. Markers mapped to this region were used to evaluate breeding parents from the United States Department of Agriculture - Agricultural Research Service (USDA-ARS) strawberry breeding program at Beltsville, Maryland. RESULTS: Markers mapped to repeat fruiting identified once-fruiting genotypes but not repeat-fruiting genotypes. Eleven of twenty-three breeding parents with repeat-fruiting marker profiles were actually once fruiting, indicating at least one additional locus acting epistatically to suppress repeat fruiting. Family segregation ratios could not be predicted reliably by the combined use of parental phenotypes and marker profiles, when using a single-gene model. Expected segregation ratios were calculated for all phenotypic and marker-profile combinations possible from the mapped locus combined with a hypothetical dominant or recessive suppressor locus. Segregation ratios specific to an epistatic suppressor acting on the mapped locus were observed in four families. The segregation ratios for two families were best explained by a dominant suppressor acting on the mapped locus, and, for the other two, by a recessive suppressor. Not all of the observed ratios could be explained by one model or the other, and when multiple families with a common parent were compared, there was no predicted genotype for the common parent that would lead to all of the observed segregation ratios. CONCLUSIONS: Considering all lines of evidence in this study and others, repeat-fruiting in commercial strawberry is controlled primarily by a dominant allele at a single locus, previously mapped by multiple groups. At least two additional genes, one dominant and one recessive, exist that act epistatically to suppress repeat fruiting. Environmental effects and/or incomplete penetrance likely affect phenotype through the suppressor loci, rather than the primary mapped locus. One of the dominant suppressors acts only in the first year, the year the plant is germinated from seed, and not after the plant has experienced a winter.


Subject(s)
Epistasis, Genetic , Fragaria/genetics , Fruit/growth & development , Plant Breeding , Fragaria/growth & development , Fruit/genetics , Genotype , Phenotype
2.
Org Biomol Chem ; 14(2): 711-715, 2016 Jan 14.
Article in English | MEDLINE | ID: mdl-26565618

ABSTRACT

Frequently present in pancreatic, colorectal and non-small cell lung carcinomas, oncogenic mutant K-Ras must be localised to the plasma membrane (PM) to be functional. Inhibitors of K-Ras PM localisation are therefore putative cancer chemotherapeutics. By screening a microbial extract library in a high content cell-based assay we detected the rare oligomycin class of Streptomyces polyketides as inhibitors of K-Ras PM localisation. Cultivation and fractionation of three unique oligomycin producing Streptomyces strains yielded oligomycins A-E (1-5) and 21-hydroxy-oligomycin A (6), together with the new 21-hydroxy-oligomycin C (7) and 40-hydroxy-oligomycin B (8). Structures for 1-8 were assigned by detailed spectroscopic analysis. Cancer cell viability screening confirmed 1-8 were cytotoxic to human colorectal carcinoma cells (IC50 > 3 µM), and were inhibitors of the ABC transporter efflux pump P-glycoprotein (P-gp), with 5 being comparable in potency to the positive control verapamil. Significantly, oligomycins 1-8 proved to be exceptionally potent inhibitors of K-Ras PM localisation (Emax 0.67-0.75 with an IC50 ~ 1.5-14 nM).


Subject(s)
Cell Membrane/drug effects , Cell Membrane/enzymology , Oligomycins/pharmacology , ras Proteins/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/pathology , Dogs , Dose-Response Relationship, Drug , Humans , Madin Darby Canine Kidney Cells , Oligomycins/chemical synthesis , Oligomycins/chemistry , Protein Transport/drug effects , Structure-Activity Relationship , ras Proteins/antagonists & inhibitors
3.
Oncogene ; 35(17): 2186-96, 2016 04 28.
Article in English | MEDLINE | ID: mdl-26212011

ABSTRACT

In the absence of extracellular stimulation the adaptor protein growth factor receptor-bound protein (Grb2) and the phospholipase Plcγ1 compete for the same binding site on fibroblast growth factor receptor 2 (FGFR2). Reducing cellular Grb2 results in upregulation of Plcγ1 and depletion of the phospholipid PI(4,5)P2. The functional consequences of this event on signaling pathways are unknown. We show that the decrease in PI(4,5)P2 level under non-stimulated conditions inhibits PTEN activity leading to the aberrant activation of the oncoprotein Akt. This results in excessive cell proliferation and tumor progression in a xenograft mouse model. As well as defining a novel mechanism of Akt phosphorylation with important therapeutic consequences, we also demonstrate that differential expression levels of FGFR2, Plcγ1 and Grb2 correlate with patient survival. Oncogenesis through fluctuation in the expression levels of these proteins negates extracellular stimulation or mutation and defines them as novel prognostic markers in ovarian cancer.


Subject(s)
GRB2 Adaptor Protein/genetics , Oncogene Protein v-akt/genetics , Ovarian Neoplasms/genetics , PTEN Phosphohydrolase/genetics , Phospholipase C gamma/genetics , Receptor, Fibroblast Growth Factor, Type 2/genetics , Animals , Carcinogenesis/genetics , Cell Proliferation/genetics , Female , GRB2 Adaptor Protein/antagonists & inhibitors , Gene Expression Regulation, Neoplastic , Humans , Mice , Ovarian Neoplasms/pathology , PTEN Phosphohydrolase/antagonists & inhibitors , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositols/metabolism , Phospholipase C gamma/biosynthesis , Prognosis , Receptor, Fibroblast Growth Factor, Type 2/biosynthesis , Signal Transduction
4.
Biochim Biophys Acta ; 1746(3): 274-83, 2005 Dec 30.
Article in English | MEDLINE | ID: mdl-16039730

ABSTRACT

Ras proteins are compartmentalized by dynamic interactions with both plasma membrane microdomains and intracellular membranes. The mechanisms underlying Ras compartmentalization involve a series of protein/lipid, lipid/lipid and cytoskeleton interactions, resulting in the generation of discrete microdomains from which Ras operates. Segregation of Ras proteins to these different platforms regulates the formation of Ras signaling complexes and the generation of discrete signal outputs. This temporal and spatial modulation of Ras signal transduction provides a mechanism for the generation of different biological outcomes from different Ras isoforms, as well as flexibility in the signal output from a single activated isoform.


Subject(s)
Cell Membrane/metabolism , Signal Transduction , ras Proteins/physiology , Animals , Humans , Intracellular Membranes/metabolism , Membrane Microdomains/metabolism , Protein Isoforms/physiology , Protein Transport
5.
Oncogene ; 20(30): 3949-58, 2001 Jul 05.
Article in English | MEDLINE | ID: mdl-11494123

ABSTRACT

Raf-1 activation is a complex process which involves plasma membrane recruitment, phosphorylation, protein-protein and lipid-protein interactions. We now show that PP1 and PP2A serine-threonine phosphatases also have a positive role in Ras dependent Raf-1 activation. General serine-threonine phosphatase inhibitors such sodium fluoride, or ss-glycerophosphate and sodium pyrophosphate, or specific PP1 and PP2A inhibitors including microcystin-LR, protein phosphatase 2A inhibitor I(1) or protein phosphatase inhibitor 2 all abrogate H-Ras and K-Ras dependent Raf-1 activation in vitro. A critical Raf-1 target residue for PP1 and PP2A is S259. Serine phosphatase inhibitors block the dephosphorylation of S259, which accompanies Raf-1 activation, and Ras dependent activation of mutant Raf259A is relatively resistant to serine phosphatase inhibitors. Sucrose gradient analysis demonstrates that serine phosphatase inhibition increases the total amount of 14-3-3 and Raf-1 associated with the plasma membrane and significantly alters the distribution of 14-3-3 and Raf-1 across different plasma membrane microdomains. These observations suggest that dephosphorylation of S259 is a critical early step in Ras dependent Raf-1 activation which facilitates 14-3-3 displacement. Inhibition of PP1 and PP2A therefore causes plasma membrane accumulation of Raf-1/14-3-3 complexes which cannot be activated.


Subject(s)
Carrier Proteins , Intracellular Signaling Peptides and Proteins , Phosphoprotein Phosphatases/physiology , Proto-Oncogene Proteins c-raf/metabolism , Tyrosine 3-Monooxygenase/physiology , 14-3-3 Proteins , Amino Acid Substitution , Animals , Binding Sites , COS Cells , Cell Membrane/metabolism , Chlorocebus aethiops , Diphosphates/pharmacology , Enzyme Activation , Enzyme Inhibitors/pharmacology , Genes, ras , Glycerophosphates/pharmacology , Isoenzymes/metabolism , Macromolecular Substances , Marine Toxins , Microcystins , Models, Biological , Mutation, Missense , Peptides, Cyclic/pharmacology , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphorylation , Protein Phosphatase 2 , Protein Processing, Post-Translational , Protein Structure, Tertiary , Proteins/pharmacology , Proto-Oncogene Proteins c-raf/chemistry , Proto-Oncogene Proteins c-raf/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , RNA-Binding Proteins/pharmacology , Sodium Fluoride/pharmacology , Transfection
6.
Methods Enzymol ; 333: 172-83, 2001.
Article in English | MEDLINE | ID: mdl-11400335

ABSTRACT

Experimental protocols that allow confident assignment of signaling proteins to specific subdomains of the plasma membrane are essential for a full understanding of the complexities of signal transduction. This is especially relevant for Ras proteins, where the different membrane anchors of the Ras isoforms target them to functionally distinct microdomains that in turn allow quantitatively different signal outputs from otherwise highly homologous proteins. The methods outlined in this chapter, in addition to being invaluable in addressing Ras function, should also have wide utility in the study of many mammalian signal transduction pathways.


Subject(s)
Caveolins/metabolism , ras Proteins/physiology , Animals , Caveolin 1 , Caveolins/chemistry , Caveolins/genetics , Cell Membrane/chemistry , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Cells, Cultured , Cricetinae , Immunohistochemistry , Microscopy, Electron , Mutation , Protein Structure, Tertiary , Transfection , ras Proteins/chemistry , ras Proteins/metabolism
7.
J Cell Sci ; 114(Pt 9): 1603-8, 2001 May.
Article in English | MEDLINE | ID: mdl-11309191

ABSTRACT

The Ras GTPases operate as molecular switches that link extracellular stimuli with a diverse range of biological outcomes. Although many studies have concentrated on the protein-protein interactions within the complex signaling cascades regulated by Ras, it is becoming clear that the spatial orientation of different Ras isoforms within the plasma membrane is also critical for their function. H-Ras, N-Ras and K-Ras use different membrane anchors to attach to the plasma membrane. Recently it has been shown that these anchors also act as trafficking signals that direct palmitoylated H-Ras and N-Ras through the exocytic pathway to the cell surface but divert polybasic K-Ras around the Golgi to the plasma membrane via an as yet-unidentified-route. Once at the plasma membrane, H-Ras and K-Ras operate in different microdomains. K-Ras is localized predominantly to the disordered plasma membrane, whereas H-Ras exists in a GTP-regulated equilibrium between disordered plasma membrane and cholesterol-rich lipid rafts. These observations provide a likely explanation for the increasing number of biological differences being identified between the otherwise highly homologous Ras isoforms and raise interesting questions about the role membrane microlocalization plays in determining the interactions of Ras with its effectors and exchange factors.


Subject(s)
Cell Compartmentation , ras Proteins/metabolism , Amino Acid Sequence , Animals , Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Molecular Sequence Data , Protein Transport , Sequence Homology, Amino Acid , ras Proteins/chemistry
8.
J Virol ; 75(9): 4150-7, 2001 May.
Article in English | MEDLINE | ID: mdl-11287564

ABSTRACT

The initial step in viral infection is the attachment of the virus to the host cell via an interaction with its receptor. We have previously shown that a receptor for human papillomavirus is the alpha6 integrin. The alpha6 integrin is involved in the attachment of epithelial cells with the basement membrane, but recent evidence suggests that ligation of many integrins results in intracellular signaling events that influence cell proliferation. Here we present evidence that exposure of A431 human epithelial cells to human papillomavirus type 6b L1 virus-like particles (VLPs) results in a dose-dependent increase in cell proliferation, as measured by bromodeoxyuridine incorporation. This proliferation is lost if VLPs are first denatured or incubated with a monoclonal antibody against L1 protein. The MEK1 inhibitor PB98059 inhibits the VLP-mediated increase in cell proliferation, suggesting involvement of the Ras-MAP kinase pathway. Indeed, VLP binding results in rapid phosphorylation of the beta4 integrin upon tyrosine residues and subsequent recruitment of the adapter protein Shc to beta4. Within 30 min, the activation of Ras, Raf, and Erk2 was observed. Finally, the upregulation of c-myc mRNA was observed at 60 min. These data indicate that human papillomavirus type 6b is able to signal cells via the Ras-MAP kinase pathway to induce cell proliferation. We hypothesize that such a mechanism would allow papillomaviruses to infect hosts more successfully by increasing the potential pool of cells they are able to infect via the initiation of proliferation in resting keratinocyte stem and suprabasal cells.


Subject(s)
Adaptor Proteins, Signal Transducing , Adaptor Proteins, Vesicular Transport , Capsid Proteins , MAP Kinase Signaling System , Oncogene Proteins, Viral/metabolism , Papillomaviridae/metabolism , Antigens, CD/metabolism , Cell Division , Cell Extracts , Cell Line , Enzyme Activation , Gene Expression , Humans , Integrin beta4 , MAP Kinase Kinase 1 , MAP Kinase Kinase 2 , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Oncogene Proteins, Viral/genetics , Papillomaviridae/genetics , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Proteins/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-raf/genetics , Proto-Oncogene Proteins c-raf/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Shc Signaling Adaptor Proteins , Src Homology 2 Domain-Containing, Transforming Protein 1 , Tyrosine/metabolism , Viral Proteins , Virion , ras Proteins/metabolism
9.
Nat Cell Biol ; 3(4): 368-75, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11283610

ABSTRACT

Different sites of plasma membrane attachment may underlie functional differences between isoforms of Ras. Here we show that palmitoylation and farnesylation targets H-ras to lipid rafts and caveolae, but that the interaction of H-ras with these membrane subdomains is dynamic. GTP-loading redistributes H-ras from rafts into bulk plasma membrane by a mechanism that requires the adjacent hypervariable region of H-ras. Release of H-ras-GTP from rafts is necessary for efficient activation of Raf. By contrast, K-ras is located outside rafts irrespective of bound nucleotide. Our studies identify a novel protein determinant that is required for H-ras function, and show that the GTP/GDP state of H-ras determines its lateral segregation on the plasma membrane.


Subject(s)
Guanosine Triphosphate/metabolism , Membrane Microdomains/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Recombinant Fusion Proteins/metabolism , Animals , Cell Line , Cell Membrane/metabolism , Cricetinae , Enzyme Activation , Lipid Metabolism , Microscopy, Immunoelectron , Proto-Oncogene Proteins c-raf/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Recombinant Fusion Proteins/genetics
10.
J Biol Chem ; 275(41): 32260-7, 2000 Oct 13.
Article in English | MEDLINE | ID: mdl-10918068

ABSTRACT

Ras proteins operate as molecular switches in signal transduction pathways downstream of tyrosine kinases and G-protein-coupled receptors. Ras is switched from the inactive GDP-bound state to the active GTP-bound state by guanine nucleotide exchange factors (GEFs). We report here the cloning and characterization of RasGRP2, a longer alternatively spliced form of the recently cloned RapGEF, CalDAG-GEFI. A unique feature of RasGRP2 is that it is targeted to the plasma membrane by a combination of N-terminal myristoylation and palmitoylation. In vivo, RasGRP2 selectively catalyzes nucleotide exchange on N- and Ki-Ras, but not Ha-Ras. RasGRP2 also catalyzes nucleotide exchange on Rap1, but this RapGEF activity is less potent than that associated with CalDAG-GEFI. The nucleotide exchange activity of RasGRP2 toward N-Ras is stimulated by diacylglycerol and inhibited by calcium. The effects of diacylglycerol and calcium are additive but are not accompanied by any detectable change in the subcellular localization of RasGRP2. In contrast, CalDAG-GEFI is localized predominantly to the cytosol and lacks Ras exchange activity in vivo. However, prolonged exposure to phorbol esters, or growth in serum, results in localization of CalDAG-GEFI to the cell membrane and restoration of Ras exchange activity. Expression of RasGRP2 or CalDAG-GEFI in NIH3T3 cells transfected with wild type N-Ras results in an accelerated growth rate but not morphologic transformation. Thus, under appropriate growth conditions, CalDAG-GEFI and RasGRP2 are dual specificity Ras and Rap exchange factors.


Subject(s)
Cell Membrane/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Membrane Proteins/metabolism , rap GTP-Binding Proteins/metabolism , ras Proteins/metabolism , Alternative Splicing/genetics , Amino Acid Sequence , Animals , Calcium/pharmacology , Cell Line , Cell Membrane/chemistry , Cloning, Molecular , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Diglycerides/pharmacology , Fluorescent Antibody Technique , Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Guanine Nucleotide Exchange Factors/chemistry , Guanine Nucleotide Exchange Factors/genetics , Humans , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/chemistry , Membrane Proteins/genetics , Molecular Sequence Data , Protein Processing, Post-Translational , Protein Transport/drug effects , Recombinant Fusion Proteins , Substrate Specificity , Tetradecanoylphorbol Acetate/pharmacology , Transfection , ras Proteins/genetics
11.
Mol Cell Biol ; 20(7): 2475-87, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10713171

ABSTRACT

Ras proteins must be localized to the inner surface of the plasma membrane to be biologically active. The motifs that effect Ras plasma membrane targeting consist of a C-terminal CAAX motif plus a second signal comprising palmitoylation of adjacent cysteine residues or the presence of a polybasic domain. In this study, we examined how Ras proteins access the cell surface after processing of the CAAX motif is completed in the endoplasmic reticulum (ER). We show that palmitoylated CAAX proteins, in addition to being localized at the plasma membrane, are found throughout the exocytic pathway and accumulate in the Golgi region when cells are incubated at 15 degrees C. In contrast, polybasic CAAX proteins are found only at the cell surface and not in the exocytic pathway. CAAX proteins which lack a second signal for plasma membrane targeting accumulate in the ER and Golgi. Brefeldin A (BFA) significantly inhibits the plasma membrane accumulation of newly synthesized, palmitoylated CAAX proteins without inhibiting their palmitoylation. BFA has no effect on the trafficking of polybasic CAAX proteins. We conclude that H-ras and K-ras traffic to the cell surface through different routes and that the polybasic domain is a sorting signal diverting K-Ras out of the classical exocytic pathway proximal to the Golgi. Farnesylated Ras proteins that lack a polybasic domain reach the Golgi but require palmitoylation in order to traffic further to the cell surface. These data also indicate that a Ras palmitoyltransferase is present in an early compartment of the exocytic pathway.


Subject(s)
Cell Membrane/metabolism , ras Proteins/metabolism , Animals , Brefeldin A/pharmacology , Cell Line , Cricetinae , Endoplasmic Reticulum/metabolism , Exocytosis , Golgi Apparatus/metabolism , Green Fluorescent Proteins , Luminescent Proteins , Microscopy, Electron , Microscopy, Fluorescence , Paclitaxel/pharmacology , Palmitic Acid/metabolism , Protein Sorting Signals/chemistry , Transfection , ras Proteins/chemistry
12.
Nat Cell Biol ; 1(2): 98-105, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10559881

ABSTRACT

The plasma membrane pits known as caveolae have been implicated both in cholesterol homeostasis and in signal transduction. CavDGV and CavKSY, two dominant-negative amino-terminal truncation mutants of caveolin, the major structural protein of caveolae, significantly inhibited caveola-mediated SV40 infection, and were assayed for effects on Ras function. We find that CavDGV completely blocked Raf activation mediated by H-Ras, but not that mediated by K-Ras. Strikingly, the inhibitory effect of CavDGV on H-Ras signalling was completely reversed by replenishing cell membranes with cholesterol and was mimicked by cyclodextrin treatment, which depletes membrane cholesterol. These results provide a crucial link between the cholesterol-trafficking role of caveolin and its postulated role in signal transduction through cholesterol-rich surface domains. They also provide direct evidence that H-Ras and K-Ras, which are targeted to the plasma membrane by different carboxy-terminal anchors, operate in functionally distinct microdomains of the plasma membrane.


Subject(s)
Caveolins , Cell Membrane/physiology , Cholesterol/metabolism , Membrane Lipids/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Sequence Deletion , 3T3 Cells , Animals , Caveolin 1 , Cell Line , Chlorocebus aethiops , Cricetinae , Genetic Vectors , Membrane Proteins/chemistry , Mice , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Recombinant Proteins/metabolism , Signal Transduction , Simian virus 40 , Transfection
13.
Oncogene ; 18(26): 3862-9, 1999 Jul 01.
Article in English | MEDLINE | ID: mdl-10445849

ABSTRACT

Activation of Raf-1 occurs at the plasma membrane. We recently showed that 14-3-3 must be complexed with Raf-1 for efficient recruitment to the plasma membrane and activation by Ras, but that 14-3-3 is completely displaced from Raf-1 following plasma membrane binding. We show here that the Raf-1 zinc finger is not absolutely required for 14-3-3 binding but is required to stabilize the interaction between Raf-1 and 14-3-3. Incubation of Raf-1 with phosphatidylserine, an inner plasma membrane phospholipid, results in removal of 14-3-3 and an increase in Raf-1 kinase activity, whereas removal of 14-3-3 from Raf-1 using specific phosphopeptides substantially reduces Raf-1 basal kinase activity. Displacement of 14-3-3 from activated Raf-1 by phosphopeptides has no effect on kinase activity if Raf-1 is first removed from solution, but completely eradicates kinase activity of soluble activated Raf-1. These results suggest a mechanism for the removal of 14-3-3 from Raf-1 at the plasma membrane and show that removal of 14-3-3 from Raf-1 has markedly different effects depending on experimental conditions.


Subject(s)
Phosphatidylserines/metabolism , Proteins/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Tyrosine 3-Monooxygenase , 14-3-3 Proteins , Animals , Binding, Competitive , COS Cells , Cattle , Chlorocebus aethiops , Cysteine/metabolism , Humans , Protein Binding , Proto-Oncogene Proteins c-raf/chemistry , Recombinant Fusion Proteins/metabolism , Transfection , Zinc Fingers/physiology
14.
J Biol Chem ; 273(37): 24052-6, 1998 Sep 11.
Article in English | MEDLINE | ID: mdl-9727023

ABSTRACT

Ha-, N-, and Ki-Ras are ubiquitously expressed in mammalian cells and can all interact with the same set of effector proteins. We show here, however, that in vivo there are marked quantitative differences in the ability of Ki- and Ha-Ras to activate Raf-1 and phosphoinositide 3-kinase. Thus, Ki-Ras both recruits Raf-1 to the plasma membrane more efficiently than Ha-Ras and is a more potent activator of membrane-recruited Raf-1 than Ha-Ras. In contrast, Ha-Ras is a more potent activator of phosphoinositide 3-kinase than Ki-Ras. Interestingly, the ability of Ha-Ras to recruit Raf-1 to the plasma membrane is significantly increased when the Ha-Ras hypervariable region is shortened so that the spacing of the Ha-Ras GTPase domains from the inner surface of the plasma membrane mimicks that of Ki-Ras. Importantly, these data show for the first time that the activation of different Ras isoforms can have distinct biochemical consequences for the cell. The mutation of specific Ras isoforms in different human tumors can, therefore, also be rationalized.


Subject(s)
Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Amino Acid Sequence , Animals , COS Cells , Cell Membrane/metabolism , Enzyme Activation , Genetic Variation , Humans , Kinetics , Molecular Sequence Data , Proto-Oncogene Proteins p21(ras)/chemistry , Proto-Oncogene Proteins p21(ras)/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Transfection
15.
Mol Cell Biol ; 18(7): 3947-55, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9632779

ABSTRACT

14-3-3 proteins complex with many signaling molecules, including the Raf-1 kinase. However, the role of 14-3-3 in regulating Raf-1 activity is unclear. We show here that 14-3-3 is bound to Raf-1 in the cytosol but is totally displaced when Raf-1 is recruited to the plasma membrane by oncogenic mutant Ras, in vitro and in vivo. 14-3-3 is also displaced when Raf-1 is targeted to the plasma membrane. When serum-starved cells are stimulated with epidermal growth factor, some recruitment of 14-3-3 to the plasma membrane is evident, but 14-3-3 recruitment correlates with Raf-1 dissociation and inactivation, not with Raf-1 recruitment. In vivo, overexpression of 14-3-3 potentiates the specific activity of membrane-recruited Raf-1 without stably associating with the plasma membrane. In vitro, Raf-1 must be complexed with 14-3-3 for efficient recruitment and activation by oncogenic Ras. Recombinant 14-3-3 facilitates Raf-1 activation by membranes containing oncogenic Ras but reduces the amount of Raf-1 that associates with the membranes. These data demonstrate that the interaction of 14-3-3 with Raf-1 is permissive for recruitment and activation by Ras, that 14-3-3 is displaced upon membrane recruitment, and that 14-3-3 may recycle Raf-1 to the cytosol. A model that rationalizes many of the apparently discrepant observations on the role of 14-3-3 in Raf-1 activation is proposed.


Subject(s)
Oncogene Protein p21(ras)/metabolism , Proteins/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Tyrosine 3-Monooxygenase , 14-3-3 Proteins , Animals , COS Cells , Cell Line , Cell Membrane/metabolism , Cricetinae , Enzyme Activation , Epidermal Growth Factor/pharmacology , Oncogene Protein p21(ras)/genetics , Proto-Oncogene Proteins c-raf/genetics , Time Factors
16.
Cancer Res ; 57(23): 5399-405, 1997 Dec 01.
Article in English | MEDLINE | ID: mdl-9393766

ABSTRACT

Point mutations, deletions, and recombinations of the RET proto-oncogene are associated with several inherited human diseases of neural crest-derived cells: Hirschsprung's disease, familial medullary thyroid carcinoma, and the multiple endocrine neoplasia (MEN) syndromes, types 2A and 2B. RET expression is restricted to normal and malignant cells of neural crest origin, such as human neuroblastoma cells. To better understand the role of the activated RET oncogene in neural crest cells, we transfected two adherent human neuroblastoma tumor cell lines with oncogenic MEN2 mutant RET cDNAs. Transfectant clones from both cell lines overexpressing MEN2B RET demonstrated a marked increase in the cell fraction growing in suspension. Both control and MEN2B cells formed tumors at the site of injection in all cases. However, mice injected with MEN2B cells developed lung metastases at a much higher frequency than control mice. Only RET protein derived from MEN2A transfectant cells had increased autokinase activity, whereas MEN2B transfectant cells demonstrated selective activation of the mitogen-activated protein kinase, Jun kinase-1 (Jnk1). These results indicate a biochemical signaling pathway that may link oncogenic RET with the metastatic process.


Subject(s)
Drosophila Proteins , Lung Neoplasms/secondary , Multiple Endocrine Neoplasia Type 2b/genetics , Multiple Endocrine Neoplasia Type 2b/pathology , Neuroblastoma/pathology , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogenes , Receptor Protein-Tyrosine Kinases/biosynthesis , Amino Acid Substitution , Animals , COS Cells , Cell Division , Humans , Lung Neoplasms/pathology , Mice , Mice, Nude , Multiple Endocrine Neoplasia Type 2a/genetics , Multiple Endocrine Neoplasia Type 2b/metabolism , Multiple Endocrine Neoplasia Type 2b/secondary , Neoplasm Metastasis , Neural Crest/cytology , Neural Crest/metabolism , Point Mutation , Polymerase Chain Reaction , Proto-Oncogene Mas , Proto-Oncogene Proteins c-ret , Recombinant Proteins/biosynthesis , Transfection , Transplantation, Heterologous , Tumor Cells, Cultured
17.
J Biol Chem ; 272(32): 20139-45, 1997 Aug 08.
Article in English | MEDLINE | ID: mdl-9242688

ABSTRACT

Ras recruits Raf to the plasma membrane for activation by a combination of tyrosine phosphorylation and other as yet undefined mechanism(s). We show here that the Raf zinc finger is not required for plasma membrane recruitment of Raf by Ras but is essential for full activation of Raf at the plasma membrane. Membrane targeting cannot compensate for the absence of the zinc finger. One facet of the zinc finger activation defect is revealed using a constitutively activated Raf mutant. Targeting Raf Y340D,Y341D to the plasma membrane increments activity, but full activation requires coexpression with activated Ras. This sensitivity to regulation by Ras at the plasma membrane is abrogated by mutations in the Raf zinc finger but is unaffected by mutation of the minimal Ras binding domain. These data show for the first time that Ras has two separate roles in Raf activation: recruitment of Raf to the plasma membrane through an interaction with the minimal Ras binding domain and activation of membrane-localized Raf via a mechanism that requires the Raf zinc finger.


Subject(s)
Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Zinc Fingers , ras Proteins/metabolism , Animals , Binding Sites , COS Cells , Cell Membrane/metabolism , Mutagenesis, Site-Directed , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-raf , Tyrosine/metabolism
18.
Blood ; 85(6): 1509-16, 1995 Mar 15.
Article in English | MEDLINE | ID: mdl-7888672

ABSTRACT

We have identified six novel types of mutation that cause factor XI deficiency, an inherited bleeding disorder. Two are point mutations that interfere with the normal splicing of exons in the mRNA and four are point mutations that result in amino acid substitutions. One of these amino acid substitutions (Asp 16-->His) is near the amino terminal end of the protein. The other three amino acid substitutions (Leu 302-->Pro, Thr 304-->Ile, and Glu 323-->Lys) are in the fourth apple domain, a region that mediates dimerization of identical subunits of factor XI. All four amino acid substitutions cause a reduction in the amount of factor XI secreted from cells grown in vitro.


Subject(s)
Factor XI Deficiency/genetics , Factor XI/genetics , Point Mutation , Amino Acid Sequence , Base Sequence , Cells, Cultured , Humans , Molecular Sequence Data
SELECTION OF CITATIONS
SEARCH DETAIL
...