Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Cell Biol ; 19(8): 5601-7, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10409749

ABSTRACT

We have examined structural differences between the proto-oncogene c-Myb and the cyclic AMP-responsive factor CREB that underlie their constitutive or signal-dependent activation properties. Both proteins stimulate gene expression via activating regions that articulate with a shallow hydrophobic groove in the KIX domain of the coactivator CREB-binding protein (CBP). Three hydrophobic residues in c-Myb that are conserved in CREB function importantly in cellular gene activation and in complex formation with KIX. These hydrophobic residues are assembled on one face of an amphipathic helix in both proteins, and mutations that disrupt c-Myb or CREB helicity in this region block interaction of either factor with KIX. Binding of the helical c-Myb domain to KIX is accompanied by a substantial increase in entropy that compensates for the comparatively low enthalpy of complex formation. By contrast, binding of CREB to KIX entails a large entropy cost due to a random coil-to-helix transition in CREB that accompanies complex formation. These results indicate that the constitutive and inducible activation properties of c-Myb and CREB reflect secondary structural characteristics of their corresponding activating regions that influence the thermodynamics of formation of a complex with CBP.


Subject(s)
Cyclic AMP Response Element-Binding Protein/chemistry , Gene Expression Regulation , Protein Structure, Secondary , Proto-Oncogene Proteins/chemistry , Trans-Activators/chemistry , Amino Acid Sequence , Animals , CREB-Binding Protein , Cyclic AMP Response Element-Binding Protein/physiology , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Nuclear Proteins/metabolism , Protein Binding , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-myb , Recombinant Fusion Proteins/metabolism , Regulatory Sequences, Nucleic Acid , Signal Transduction , Structure-Activity Relationship , Thermodynamics , Trans-Activators/metabolism , Trans-Activators/physiology , Transcriptional Activation , Transfection
2.
Proc Natl Acad Sci U S A ; 96(12): 7065-70, 1999 Jun 08.
Article in English | MEDLINE | ID: mdl-10359839

ABSTRACT

Interest in the guanosine triphosphatase (GTPase) reaction of Ras as a molecular drug target stems from the observation that, in a large number of human tumors, Ras is characteristically mutated at codons 12 or 61, more rarely 13. Impaired GTPase activity, even in the presence of GTPase activating proteins, has been found to be the biochemical reason behind the oncogenicity of most Gly12/Gln61 mutations, thus preventing Ras from being switched off. Therefore, these oncogenic Ras mutants remain constitutively activated and contribute to the neoplastic phenotype of tumor cells. Here, we show that the guanosine 5'-triphosphate (GTP) analogue diaminobenzophenone-phosphoroamidate-GTP (DABP-GTP) is hydrolyzed by wild-type Ras but more efficiently by frequently occurring oncogenic Ras mutants, to yield guanosine 5'-diphosphate-bound inactive Ras and DABP-Pi. The reaction is independent of the presence of Gln61 and is most dramatically enhanced with Gly12 mutants. Thus, the defective GTPase reaction of the oncogenic Ras mutants can be rescued by using DABP-GTP instead of GTP, arguing that the GTPase switch of Ras is not irreversibly damaged. An exocyclic aromatic amino group of DABP-GTP is critical for the reaction and bypasses the putative rate-limiting step of the intrinsic Ras GTPase reaction. The crystal structures of Ras-bound DABP-beta,gamma-imido-GTP show a disordered switch I and identify the Gly12/Gly13 region as the hydrophobic patch to accommodate the DABP-moiety. The biochemical and structural studies help to define the requirements for the design of anti-Ras drugs aimed at the blocked GTPase reaction.


Subject(s)
Benzophenones/analysis , Genes, ras , Guanosine Triphosphate/analogs & derivatives , Mutation , ras Proteins/genetics , ras Proteins/metabolism , Benzophenones/chemistry , Benzophenones/metabolism , Crystallography, X-Ray , Escherichia coli , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Guanosine Triphosphate/analysis , Guanosine Triphosphate/chemistry , Guanosine Triphosphate/genetics , Guanosine Triphosphate/metabolism , Humans , Hydrolysis , Molecular Sequence Data , Protein Binding
3.
FEBS Lett ; 433(3): 326-30, 1998 Aug 21.
Article in English | MEDLINE | ID: mdl-9744820

ABSTRACT

Hydrolysis of GTP, bound to members of the G-protein superfamily, terminates their downstream signaling activity. A conserved glutamine serves a critical role in this pivotal guanosine triphosphatase (GTPase) reaction. However, the role of the catalytic glutamine in GTP hydrolysis is still not well understood. We have employed substrate-assisted catalysis to probe the catalytic mechanism of Gs alpha using GTP analogues. These GTP analogues, each having different functional groups, were designed to support or refute particular putative GTPase mechanisms. We have found that a hydrogen donor group, in close proximity to the gamma-phosphate of GTP, is necessary and sufficient to substitute for the function of the catalytic glutamine in the GTPase reaction.


Subject(s)
GTP Phosphohydrolases/metabolism , GTP-Binding Protein alpha Subunits, Gs/metabolism , Guanosine Triphosphate/analogs & derivatives , Guanosine Triphosphate/metabolism , Adenosine Triphosphate/metabolism , Adenylyl Cyclases/metabolism , Animals , Cell Membrane/metabolism , GTP Phosphohydrolases/chemistry , GTP-Binding Protein alpha Subunits, Gs/chemistry , Glutamine/metabolism , Hydrolysis , Kinetics , Parotid Gland/metabolism , Rats , Substrate Specificity
4.
Eur J Biochem ; 249(1): 330-6, 1997 Oct 01.
Article in English | MEDLINE | ID: mdl-9363787

ABSTRACT

Signaling by guanine-nucleotide-binding proteins (G-proteins) occurs when they are charged with GTP, while hydrolysis of the bound nucleotide turns the signaling off. Despite a wealth of biochemical and structural information, the mechanism of GTP hydrolysis by G-proteins remains controversial. We have employed substrate-assisted catalysis as a novel approach to study catalysis by G-proteins. In these studies, we have used diaminobenzophenone-phosphonoamidate-GTP, a unique GTP analog bearing the functional groups that are missing in the GTPase-deficient [Leu227]G(s alpha) mutant. This mutant, found in various human tumors, fails to hydrolyze GTP for an extended period. In contrast, the GTP analog is hydrolyzed by this mutant and by the wild-type enzyme at the same rate. On the other hand, modification of G(s alpha) by cholera toxin, which catalyses ADP-ribosylation of Arg201 of G(s alpha), decreased the rates of hydrolysis of both GTP and its analog by 95%. These results attest to the specificity of the GTP analog as a unique substrate for the [Leu227]G(s alpha) mutant and to the essential role of Gln227 in GTP hydrolysis. Furthermore, the finding that the GTP analog was hydrolyzed at the same rate as GTP by the wild-type enzyme, favors a model in which formation of a pentavalent transition state intermediate, presumably stabilized by the catalytic glutamine, is not the rate-limiting step of the GTPase reaction.


Subject(s)
GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Point Mutation , Animals , Benzophenones/chemistry , Benzophenones/metabolism , Binding Sites/genetics , Catalysis , Cholera Toxin/pharmacology , Computer Simulation , GTP-Binding Proteins/chemistry , Guanosine Triphosphate/analogs & derivatives , Guanosine Triphosphate/chemistry , Guanosine Triphosphate/metabolism , Humans , Hydrolysis , In Vitro Techniques , Kinetics , Models, Molecular , Protein Conformation , Rats , Signal Transduction
5.
Anal Biochem ; 236(2): 302-8, 1996 May 01.
Article in English | MEDLINE | ID: mdl-8660509

ABSTRACT

Determination of microgram quantities of protein in the Bradford Coomassie brilliant blue assay is accomplished by measurement of absorbance at 590 nm. However, as intrinsic nonlinearity compromises the sensitivity and accuracy of this method. It is shown that under standard assay conditions, the ratio of the absorbances, 590 nm over 450 nm, is strictly linear with protein concentration. This simple procedure increases the accuracy and improves the sensitivity of the assay about 10-fold, permitting quantitation down to 50 ng of bovine serum albumin. Furthermore, protein assay in presence of up to 35-fold weight excess of sodium dodecyl sulfate (detergent) over bovine serum albumin (protein) can be performed. A linear equation that perfectly fits the experimental data is provided on the basis of mass action and Beer's law.


Subject(s)
Serum Albumin, Bovine/analysis , Spectrophotometry/methods , Calibration , Detergents , Indicators and Reagents , Linear Models , Microchemistry , Rosaniline Dyes , Sensitivity and Specificity , Sodium Dodecyl Sulfate
6.
Biochem J ; 306 ( Pt 1): 253-8, 1995 Feb 15.
Article in English | MEDLINE | ID: mdl-7864818

ABSTRACT

A novel photoaffinity label, m-acetylanilido-GTP (m-AcAGTP), was synthesized and used to identify GTP-binding proteins (G-proteins). This GTP analogue is easily prepared and can be used for photoaffinity labelling of G-proteins without chromatographic purification. In the presence of the beta-adrenergic agonist isoprenaline, it activates turkey erythrocyte adenylate cyclase. This activation persists even when the beta-adrenergic receptor is subsequently blocked by antagonist, indicating that the GTP analogue is resistant to hydrolysis. The apparent Ka for activation of turkey erythrocyte adenylate cyclase by m-AcAGTP was found to be 0.21 microM, a value similar to that for guanosine 5'-[beta,gamma-imido]triphosphate. m-AcAGTP also effectively inhibited the light-dependent GTPase of Musca fly eye membranes. Photoaffinity labelling of fly eye membranes with [alpha-32P]m-AcAGTP, followed by immunoprecipitation of G-protein Gq, identified a labelled protein band with the mobility of a 41.5 kDa protein on SDS/PAGE. Labelling of this protein was enhanced 9-fold in blue over red illuminated membranes, containing metarhodopsin and rhodopsin respectively. Labelling of alpha-subunits of heterotrimeric G-proteins was also demonstrated in turkey erythrocyte membranes. The ease of preparation of m-AcAGTP and the chemical properties of the photoreactive acetophenone make this affinity label an important new tool in studies of cellular phenomena mediated by guanine nucleotide-binding proteins.


Subject(s)
Affinity Labels , GTP-Binding Proteins/analysis , Guanosine Triphosphate/analogs & derivatives , Adenylyl Cyclases/metabolism , Animals , Enzyme Activation/drug effects , Erythrocytes/enzymology , GTP-Binding Proteins/metabolism , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacology , Guanosine Triphosphate/chemical synthesis , Guanosine Triphosphate/metabolism , Guanosine Triphosphate/pharmacology , Guanylyl Imidodiphosphate/pharmacology , Immunosorbent Techniques , Isoproterenol/pharmacology , Photochemistry , Propranolol/pharmacology , Turkeys/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...