Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Curr Biol ; 21(7): 563-8, 2011 Apr 12.
Article in English | MEDLINE | ID: mdl-21458265

ABSTRACT

RAF kinase inhibitors can induce ERK cascade signaling by promoting dimerization of RAF family members in the presence of oncogenic or normally activated RAS. This interaction is mediated by a dimer interface region in the RAF kinase domain that is conserved in members of the ERK cascade scaffold family, kinase suppressor of RAS (KSR). In this study, we find that most RAF inhibitors also induce the binding of KSR1 to wild-type and oncogenic B-RAF proteins, including V600E B-RAF, but promote little complex formation between KSR1 and C-RAF. The inhibitor-induced KSR1/B-RAF interaction requires direct binding of the drug to B-RAF and is dependent on conserved dimer interface residues in each protein, but, unexpectedly, is not dependent on binding of B-RAF to activated RAS. Inhibitor-induced KSR/B-RAF complex formation can occur in the cytosol and is observed in normal mouse fibroblasts, as well as a variety of human cancer cell lines. Strikingly, we find that KSR1 competes with C-RAF for inhibitor-induced binding to B-RAF and, as a result, alters the effect of the inhibitors on ERK cascade signaling.


Subject(s)
MAP Kinase Signaling System/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Proto-Oncogene Proteins B-raf/metabolism , raf Kinases/antagonists & inhibitors , Animals , Carcinoma, Non-Small-Cell Lung , Cell Line, Tumor , Dimerization , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Melanoma , Mice , Protein Binding , Protein Kinase Inhibitors/metabolism , Protein Kinases/genetics , Proto-Oncogene Proteins B-raf/genetics , Pyridines/metabolism , Pyridines/pharmacology , raf Kinases/metabolism
2.
Small GTPases ; 2(5): 276-281, 2011 Sep.
Article in English | MEDLINE | ID: mdl-22292131

ABSTRACT

The Ras, Raf, MEK and ERK proteins form an essential signal transduction pathway that is aberrantly activated in many human cancers. Kinase suppressor of Ras (KSR) is a conserved positive modulator of this pathway, and since its discovery, there has been a concerted effort to elucidate KSR function in both normal and aberrant Ras/ERK signaling. The KSR proteins possess a C-terminal region that is closely related to the Raf family kinase domain; however, mammalian KSR proteins lack a key catalytic residue, suggesting a role as a pseudokinase. Like many other pseudokinases, KSR has scaffolding activities and interacts with Raf, MEK and ERK to provide spatio-temporal regulation of ERK activation. Recently, significant advances have been made that further our understanding of how KSR proteins function in normal and oncogenic signaling. The newly solved KSR2/MEK1 structure has revealed important mechanistic details for how KSR regulates MEK activation and has raised questions regarding KSR kinase activity. In addition, KSR expression levels have been found to alter the effects of Raf inhibitors on oncogenic Ras/ERK signaling. Specifically, KSR1 competes with C-Raf for inhibitor-induced binding to B-Raf and in doing so attenuates the paradoxical activating effect of these drugs on ERK signaling.

3.
Methods Mol Biol ; 661: 323-34, 2010.
Article in English | MEDLINE | ID: mdl-20811992

ABSTRACT

ERK cascade scaffolds serve as docking platforms to coordinate the assembly of multiprotein complexes that contribute to the spatial and temporal control of ERK signaling. Given that protein-protein interactions are essential for scaffold function, determining the full repertoire of scaffold binding partners will likely provide new insight into the regulation and activities of the ERK cascade scaffolds. In this chapter, we describe methods to identify scaffold interacting proteins using a proteomics approach. This protocol is based on the affinity purification of scaffold complexes from tissue culture cells and utilizes mass spectrometry to identify the protein constituents of the complex.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Signaling System , Proteomics/methods , Extracellular Signal-Regulated MAP Kinases/isolation & purification , HEK293 Cells , Humans , Mass Spectrometry , Resins, Synthetic
4.
Proc Natl Acad Sci U S A ; 106(27): 11022-7, 2009 Jul 07.
Article in English | MEDLINE | ID: mdl-19541618

ABSTRACT

Scaffold proteins contribute to the spatiotemporal control of MAPK signaling and KSR1 is an ERK cascade scaffold that localizes to the plasma membrane in response to growth factor treatment. To better understand the molecular mechanisms of KSR1 function, we examined the interaction of KSR1 with each of the ERK cascade components, Raf, MEK, and ERK. Here, we identify a hydrophobic motif within the proline-rich sequence (PRS) of MEK1 and MEK2 that is required for constitutive binding to KSR1 and find that MEK binding and residues in the KSR1 CA1 region enable KSR1 to form a ternary complex with B-Raf and MEK following growth factor treatment that enhances MEK activation. We also find that docking of active ERK to the KSR1 scaffold allows ERK to phosphorylate KSR1 and B-Raf on feedback S/TP sites. Strikingly, feedback phosphorylation of KSR1 and B-Raf promote their dissociation and result in the release of KSR1 from the plasma membrane. Together, these findings provide unique insight into the signaling dynamics of the KSR1 scaffold and reveal that through regulated interactions with Raf and ERK, KSR1 acts to both potentiate and attenuate ERK cascade activation, thus regulating the intensity and duration of ERK cascade signaling emanating from the plasma membrane during growth factor signaling.


Subject(s)
Multiprotein Complexes/metabolism , Protein Kinases/metabolism , Signal Transduction , Amino Acid Motifs , Amino Acid Sequence , Animals , Cell Line , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Feedback, Physiological , Humans , Mice , Mitogen-Activated Protein Kinase Kinases/chemistry , Mitogen-Activated Protein Kinase Kinases/metabolism , Molecular Sequence Data , Phosphorylation , Proline-Rich Protein Domains , Protein Binding , Proto-Oncogene Proteins B-raf/metabolism
5.
J Biol Chem ; 282(36): 26225-34, 2007 Sep 07.
Article in English | MEDLINE | ID: mdl-17613518

ABSTRACT

Kinase suppressor of Ras 1 (KSR1) is a protein scaffold that facilitates ERK cascade activation at the plasma membrane, a critical step in the signal transduction process that allows cells to respond to survival, proliferative, and differentiative cues. Here, we report that KSR1 undergoes caspase-dependent cleavage in apoptotic cells and that cleavage destroys the scaffolding function of the full-length KSR1 protein and generates a stable C-terminal fragment that can inhibit ERK activation. KSR1 is cleaved in response to multiple apoptotic stimuli and occurs in vivo during the involution of mouse mammary tissues, a morphogenic process requiring cellular apoptosis. In addition, we find that in comparison with KSR1(-/-) mouse embryonic fibroblasts expressing wild type KSR1 (WT-KSR1), cells expressing a cleavage-resistant KSR1 protein (DEVA-KSR1) exhibit reduced apoptotic signaling in response to tumor necrosis factor-alpha/cycloheximide treatment. The effect of DEVA-KSR1 expression was found to correlate with increased levels of active phosphoERK and could be significantly reversed by treating cells with the MEK inhibitor U0126. In contrast, reduced phosphoERK levels and enhanced apoptotic signaling were observed in cells constitutively expressing the C-terminal KSR1 fragment (CTF-KSR1). Moreover, we find that cleavage of WT-KSR1 correlates with a dramatic reduction in active phosphoERK levels. These findings identify KSR1 as a caspase target and suggest that cleavage of the KSR1 scaffold represents another mechanism whereby caspases down-regulate ERK survival signaling to promote cellular apoptosis.


Subject(s)
Apoptosis/physiology , Caspases/metabolism , Embryo, Mammalian/enzymology , Fibroblasts/enzymology , MAP Kinase Signaling System/physiology , Mammary Glands, Animal/enzymology , Protein Kinases/metabolism , Animals , Apoptosis/drug effects , Butadienes/pharmacology , Caspases/genetics , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Membrane/enzymology , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/physiology , Embryo, Mammalian/cytology , Female , Fibroblasts/cytology , MAP Kinase Signaling System/drug effects , Mammary Glands, Animal/cytology , Mice , Mice, Knockout , Nitriles/pharmacology , Protein Kinases/genetics
6.
Traffic ; 5(2): 102-16, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14690499

ABSTRACT

GGAs comprise a family of Arf-dependent coat proteins or adaptors that regulate vesicle traffic from the trans-Golgi network (TGN). GGAs bind activated Arf, cargo, and additional components necessary for vesicle budding through interactions with their four functional domains: VHS, GAT, hinge, and GAE. We identified three sites of phosphorylation in GGA1 by tandem mass spectrometry: S268 and T270 in the GAT domain and S480 in the hinge. Expression of HA-GGA1 in mammalian cells and comparison to endogenous GGA1 confirmed their localization to late Golgi compartments. In contrast, mutations that mimic the phosphoprotein (HA-GGA1[S268D] or HA-GGA1[T270D]) at either of the sites in the GAT domain caused a decrease in the colocalization with markers of the Golgi and TGN and an increase in puncta in cytoplasm. Quantitative comparisons of the extent of colocalization of GGA1 proteins with the known components of GGA1 vesicles revealed that the composition of those markers tested in HA-GGA1[S268D] and HA-GGA1[T270D] vesicles were indistinguishable from those of HA-GGA1 vesicles. We conclude that phosphorylation of the GAT domain can stabilize the coat proteins bound and thus regulate the rate of coat protein dissociation.


Subject(s)
ADP-Ribosylation Factors/metabolism , Adaptor Proteins, Vesicular Transport , Carrier Proteins/metabolism , Subcellular Fractions/metabolism , ADP-Ribosylation Factors/genetics , Animals , Biomarkers , COS Cells , Carrier Proteins/genetics , Chlorocebus aethiops , DNA-Binding Proteins/metabolism , Golgi Apparatus/metabolism , HeLa Cells , Humans , Mutation , Phosphorylation , Protein Structure, Tertiary , Serine/metabolism , Subcellular Fractions/chemistry , Threonine/metabolism , Vesicular Transport Proteins/metabolism , rab GTP-Binding Proteins/metabolism , trans-Golgi Network/metabolism
7.
Arch Biochem Biophys ; 404(2): 197-209, 2002 Aug 15.
Article in English | MEDLINE | ID: mdl-12147257

ABSTRACT

We have cloned and overexpressed a truncated, recombinant form of beta-carbonic anhydrase from Arabidopsis thaliana. The wild-type enzyme and two site-directed variants, H216N and Y212F, have been kinetically characterized both at steady state by stopped-flow spectrophotometry and at chemical equilibrium by (18)O isotope exchange methods. The wild-type enzyme has a maximal k(cat) for CO2 hydration of 320 ms(-1) and is rate limited by proton transfer involving two residues with apparent pK(a) values of 6.0 and 8.7. The mutant enzyme H216N has a maximal k(cat) at high pH that is 43% that of wild type, but is only 5% that of wild type at pH 7.0. (18)O exchange studies reveal that the effect of the mutations H216N or Y212F is primarily on proton transfer steps in the catalytic mechanism and not in the rate of CO2-HCO3- exchange. These results suggest that residues His-216 and Tyr-212 are both important for efficient proton transfer in A. thaliana carbonic anhydrase.


Subject(s)
Arabidopsis/enzymology , Carbonic Anhydrases/chemistry , Carbonic Anhydrases/genetics , Amino Acid Substitution , Arabidopsis Proteins/chemistry , Arabidopsis Proteins/genetics , Binding Sites/physiology , Carbon Dioxide/chemistry , Catalysis , Enzyme Activation/physiology , Escherichia coli/genetics , Hydrogen-Ion Concentration , Imidazoles/chemistry , Isoenzymes/chemistry , Isoenzymes/genetics , Kinetics , Molecular Weight , Mutagenesis, Site-Directed , Oxygen Isotopes , Protons , Substrate Specificity , Zinc/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...