Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Mol Metab ; 74: 101765, 2023 08.
Article in English | MEDLINE | ID: mdl-37390979

ABSTRACT

OBJECTIVE: Obesity is a major global health problem which can be targeted with new mechanistic diverse pharmacological interventions. Here we evaluate a new long-acting secretin receptor agonist as a potential treatment for obesity. METHODS: BI-3434 was designed as a secretin analog with stabilized peptide backbone and attached fatty acid-based half-life extension group. The peptide was evaluated in vitro for its ability to stimulate cAMP accumulation in a cell line stably expressing recombinant secretin receptor. On the functional level, stimulation of lipolysis in primary adipocytes after treatment with BI-3434 was determined. The ability of BI-3434 to activate secretin receptor in vivo was assessed in a cAMP reporter CRE-Luc mouse model. Furthermore, a diet-induced obesity mouse model was used to test the effects of BI-3434 on body weight and food intake following repeated daily subcutaneous administration alone and in combination with a GLP-1R agonist. RESULTS: BI-3434 potently activated human secretin receptor. However, lipolysis was only weakly induced in primary murine adipocytes. BI-3434 had an extended half-life compared to endogenous secretin and activated target tissues like pancreas, adipose tissue, and stomach in vivo. BI-3434 did not lower food intake in lean or diet-induced obese mice, but it increased energy expenditure after daily administration. This led to a loss of fat mass, which did not translate in a significant effect on body weight. However, treatment in combination with a GLP-1R agonist led to a synergistic effect on body weight loss. CONCLUSIONS: BI-3434 is a highly potent and selective agonist of secretin receptor with an extended pharmacokinetic (PK) profile. Increased energy expenditure after daily treatment with BI-3434 suggests that secretin receptor is involved in metabolic regulation and energy homeostasis. Targeting secretin receptor alone may not be an efficient anti-obesity treatment, but could be combined with anorectic principles like GLP-1R agonists.


Subject(s)
Gastrointestinal Hormones , Secretin , Mice , Humans , Animals , Secretin/pharmacology , Secretin/therapeutic use , Obesity/drug therapy , Obesity/etiology , Obesity/metabolism , Body Weight , Peptides/pharmacology , Peptides/therapeutic use , Diet, High-Fat/adverse effects
2.
Eur J Pharmacol ; 908: 174352, 2021 Oct 05.
Article in English | MEDLINE | ID: mdl-34274340

ABSTRACT

The anorectic action of the pancreatic hormone amylin is mainly mediated through the area postrema (AP). Amylin activates AP neurons using a heterodimeric receptor (AMY) composed of the calcitonin receptor (CTR) and the receptor activity modifying protein (RAMP 1, 2 or 3). The aim of the following experiments is to test the effects of the long acting amylin analogue (LAAMA) in RAMP1/3 knock-out (KO) male mice and in neuronal CTR KO Nestin-CreCTR male mice. In vitro, LAAMA exerted an equipotent effect on CTR and AMYs that was maintained across species. Following one week of 45% high fat diet, WT, RAMP1/3 KO and Nestin-CreCTR mice were injected daily for one week with vehicle or LAAMA. LAAMA decreased body weight gain in WT and in RAMP1/3 KO mice suggesting that RAMP1/3 are not necessary for LAAMA-induced effects. However, LAAMA was not able to produce any body lowering and anorectic effects in Nestin-CreCTR mice. This was accompanied by the absence of any c-Fos signal in the AP opposite to WT control mice. Together, these results suggest that LAAMA's effects are mainly mediated through CTR rather than specific AMY. The study of LAAMA or any amylin receptor agonist in different receptor KO mouse models helps disentangle the underlying mechanisms used by these molecules.


Subject(s)
Receptors, Calcitonin , Animals , Area Postrema , Islet Amyloid Polypeptide , Mice , Proto-Oncogene Proteins c-fos
3.
Cell Signal ; 28(10): 1451-62, 2016 10.
Article in English | MEDLINE | ID: mdl-27368419

ABSTRACT

RAF family kinases are central components of the Ras-RAF-MEK-ERK cascade. Dimerization is a key mechanism of RAF activation in response to physiological, pathological and pharmacological signals. It is mediated by a dimer interface region in the RAF kinase domain that is also conserved in KSR, a scaffolding protein that binds RAF, MEK and ERK. The regulation of RAF dimerization is incompletely understood. Especially little is known about the molecular mechanism involved in the selection of the dimerization partner. Previously, we reported that Ras-dependent binding of the tumour suppressor DiRas3 to C-RAF inhibits the C-RAF:B-RAF heterodimerization. Here we show that DiRas3 binds to KSR1 independently of its interaction with activated Ras and RAF. Our data also suggest that depending on the local stoichiometry between DiRas3 and oncogenic Ras, DiRas3 can either enhance homodimerization of KSR1 or recruit KSR1 to the Ras:C-RAF complex and thereby reduce the availability of C-RAF for binding to B-RAF. This mechanism, which is shared between A-RAF and C-RAF, may be involved in the regulation of Ras12V-induced cell transformation by DiRas3.


Subject(s)
Multiprotein Complexes/metabolism , Protein Kinases/metabolism , Protein Multimerization , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins c-raf/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Cell Proliferation , Cell Transformation, Neoplastic , Intracellular Space/metabolism , Mice , NIH 3T3 Cells , Protein Binding , Protein Stability , Proto-Oncogene Proteins p21(ras)/metabolism , Subcellular Fractions/metabolism
4.
J Biol Chem ; 288(49): 35237-52, 2013 Dec 06.
Article in English | MEDLINE | ID: mdl-24158441

ABSTRACT

In metazoans, the highly conserved MAPK signaling pathway regulates cell fate decision. Aberrant activation of this pathway has been implicated in multiple human cancers and some developmental disorders. KSR1 functions as an essential scaffold that binds the individual components of the cascade and coordinates their assembly into multiprotein signaling platforms. The mechanism of KSR1 regulation is highly complex and not completely understood. In this study, we identified Tyr(728) as a novel regulatory phosphorylation site in KSR1. We show that Tyr(728) is phosphorylated by LCK, uncovering an additional and unexpected link between Src kinases and MAPK signaling. To understand how phosphorylation of Tyr(728) may regulate the role of KSR1 in signal transduction, we integrated structural modeling and biochemical studies. We demonstrate that Tyr(728) is involved in maintaining the conformation of the KSR1 kinase domain required for binding to MEK. It also affects phosphorylation and activation of MEK by RAF kinases and consequently influences cell proliferation. Moreover, our studies suggest that phosphorylation of Tyr(728) may affect the intrinsic kinase activity of KSR1. Together, we propose that phosphorylation of Tyr(728) may regulate the transition between the scaffolding and the catalytic function of KSR1 serving as a control point used to fine-tune cellular responses.


Subject(s)
Mitogen-Activated Protein Kinase Kinases/metabolism , Protein Kinases/chemistry , Protein Kinases/metabolism , Amino Acid Sequence , Animals , Cell Line , Cell Proliferation , Enzyme Activation , Humans , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , MAP Kinase Signaling System , Mice , Models, Molecular , Molecular Dynamics Simulation , Molecular Sequence Data , Mutagenesis, Site-Directed , Phosphorylation , Protein Binding , Protein Conformation , Protein Kinases/genetics , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Tyrosine/chemistry
5.
Mol Biosyst ; 9(4): 551-8, 2013 Apr 05.
Article in English | MEDLINE | ID: mdl-23212737

ABSTRACT

Raf kinases function downstream of Ras proteins to activate the MEK-ERK pathway which is deregulated in a large number of human cancers. Raf inhibitors are clinically highly effective for the treatment of cancer and melanoma in particular, but have unexpected side effects that include a paradoxical activation of the ERK pathway. These effects seem to be related to the heterodimerization of Raf-1 and B-Raf kinases. Here, we discuss the role of Raf dimerization as part of the physiological activation mechanism of Raf kinases, the mechanism of Raf dimerization induced by drugs, and the implications of dimerization for drug therapies targeting Raf kinases.


Subject(s)
Protein Multimerization , Signal Transduction , raf Kinases/chemistry , raf Kinases/metabolism , Drug Resistance, Neoplasm/genetics , Humans , Neoplasms/metabolism , Protein Binding , Protein Kinase Inhibitors , raf Kinases/genetics
6.
J Biol Chem ; 287(27): 23128-40, 2012 Jun 29.
Article in English | MEDLINE | ID: mdl-22605333

ABSTRACT

The maternally imprinted Ras-related tumor suppressor gene DiRas3 is lost or down-regulated in more than 60% of ovarian and breast cancers. The anti-tumorigenic effect of DiRas3 is achieved through several mechanisms, including inhibition of cell proliferation, motility, and invasion, as well as induction of apoptosis and autophagy. Re-expression of DiRas3 in cancer cells interferes with the signaling through Ras/MAPK and PI3K. Despite intensive research, the mode of interference of DiRas3 with the Ras/RAF/MEK/ERK signal transduction is still a matter of speculation. In this study, we show that DiRas3 associates with the H-Ras oncogene and that activation of H-Ras enforces this interaction. Furthermore, while associated with DiRas3, H-Ras is able to bind to its effector protein C-RAF. The resulting multimeric complex consisting of DiRas3, C-RAF, and active H-Ras is more stable than the two protein complexes H-Ras·C-RAF or H-Ras·DiRas3, respectively. The consequence of this complex formation is a DiRas3-mediated recruitment and anchorage of C-RAF to components of the membrane skeleton, suppression of C-RAF/B-RAF heterodimerization, and inhibition of C-RAF kinase activity.


Subject(s)
MAP Kinase Signaling System/physiology , Proto-Oncogene Proteins c-raf/chemistry , Proto-Oncogene Proteins c-raf/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , rho GTP-Binding Proteins/metabolism , Animals , COS Cells , Cell Membrane/metabolism , Chlorocebus aethiops , Cytoskeleton/metabolism , Dimerization , Genes, Tumor Suppressor/physiology , Humans , Multiprotein Complexes/metabolism , Prenylation/physiology , Proto-Oncogene Proteins c-raf/genetics , Proto-Oncogene Proteins p21(ras)/genetics , rho GTP-Binding Proteins/genetics
7.
J Biol Chem ; 286(18): 16491-503, 2011 May 06.
Article in English | MEDLINE | ID: mdl-21454547

ABSTRACT

The serine/threonine kinase RAF is a central component of the MAPK cascade. Regulation of RAF activity is highly complex and involves recruitment to membranes and association with Ras and scaffold proteins as well as multiple phosphorylation and dephosphorylation events. Previously, we identified by molecular modeling an interaction between the N-region and the RKTR motif of the kinase domain in RAF and assigned a new function to this tetrapeptide segment. Here we found that a single substitution of each basic residue within the RKTR motif inhibited catalytic activity of all three RAF isoforms. However, the inhibition and phosphorylation pattern of C-RAF and A-RAF differed from B-RAF. Furthermore, substitution of the first arginine led to hyperphosphorylation and accumulation of A-RAF and C-RAF in plasma membrane fraction, indicating that this residue interferes with the recycling process of A-RAF and C-RAF but not B-RAF. In contrast, all RAF isoforms behave similarly with respect to the RKTR motif-dependent dimerization. The exchange of the second arginine led to exceedingly increased dimerization as long as one of the protomers was not mutated, suggesting that substitution of this residue with alanine may result in similar a structural rearrangement of the RAF kinase domain, as has been found for the C-RAF kinase domain co-crystallized with a dimerization-stabilizing RAF inhibitor. In summary, we provide evidence that each of the basic residues within the RKTR motif is indispensable for correct RAF function.


Subject(s)
Cell Membrane/enzymology , Mutation, Missense , Protein Multimerization/physiology , raf Kinases/metabolism , Amino Acid Motifs , Amino Acid Substitution , Animals , COS Cells , Cell Membrane/genetics , Chlorocebus aethiops , Humans , Protein Structure, Tertiary , raf Kinases/genetics
8.
J Biol Chem ; 286(20): 17934-44, 2011 May 20.
Article in English | MEDLINE | ID: mdl-21317286

ABSTRACT

BAD (Bcl-2 antagonist of cell death) belongs to the proapoptotic BH3-only subfamily of Bcl-2 proteins. Physiological activity of BAD is highly controlled by phosphorylation. To further analyze the regulation of BAD function, we investigated the role of recently identified phosphorylation sites on BAD-mediated apoptosis. We found that in contrast to the N-terminal phosphorylation sites, the serines 124 and 134 act in an antiapoptotic manner because the replacement by alanine led to enhanced cell death. Our results further indicate that RAF kinases represent, besides PAK1, BAD serine 134 phosphorylating kinases. Importantly, in the presence of wild type BAD, co-expression of survival kinases, such as RAF and PAK1, leads to a strongly increased proliferation, whereas substitution of serine 134 by alanine abolishes this process. Furthermore, we identified BAD serine 134 to be strongly involved in survival signaling of B-RAF-V600E-containing tumor cells and found that phosphorylation of BAD at this residue is critical for efficient proliferation in these cells. Collectively, our findings provide new insights into the regulation of BAD function by phosphorylation and its role in cancer signaling.


Subject(s)
Apoptosis , Cell Proliferation , MAP Kinase Signaling System , Neoplasms/metabolism , Proto-Oncogene Proteins B-raf/metabolism , bcl-Associated Death Protein/metabolism , Amino Acid Substitution , Cell Survival/genetics , HEK293 Cells , HeLa Cells , Humans , Mutation, Missense , Neoplasms/genetics , Phosphorylation , Proto-Oncogene Proteins B-raf/genetics , bcl-Associated Death Protein/genetics , p21-Activated Kinases/genetics , p21-Activated Kinases/metabolism
9.
Biochim Biophys Acta ; 1810(2): 162-9, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21081150

ABSTRACT

BACKGROUND: BAD protein (Bcl-2 antagonist of cell death) belongs to the BH3-only subfamily of proapoptotic proteins and is proposed to function as the sentinel of the cellular health status. Physiological activity of BAD is regulated by phosphorylation, association with 14-3-3 proteins, binding to membrane lipids and pore formation. Since the functional role of the BAD C-terminal part has not been considered so far, we have investigated here the interplay of the structure and function of this region. METHODS: The structure of the regulatory C-terminal part of human BAD was analyzed by CD spectroscopy. The channel-forming activity of full-length BAD and BAD peptides was carried out by lipid bilayer measurements. Interactions between proteins and peptides were monitored by the surface plasmon resonance technique. In aqueous solution, C-terminal part of BAD exhibits a well-ordered structure and stable conformation. In a lipid environment, the helical propensity considerably increases. The interaction of the C-terminal segment of BAD with the isolated BH3 domain results in the formation of permanently open pores whereby the phosphorylation of serine 118 within the BH3 domain is necessary for effective pore formation. In contrast, phosphorylation of serine 99 in combination with 14-3-3 association suppresses formation of channels. C-terminal part of BAD controls BAD function by structural transitions, lipid binding and phosphorylation. Conformational changes of this region upon membrane interaction in conjunction with phosphorylation of the BH3 domain suggest a novel mechanism for regulation of BAD. GENERAL SIGNIFICANCE: Multiple signaling pathways mediate inhibition and activation of cell death via BAD.


Subject(s)
Lipid Bilayers/chemistry , Protein Conformation , Protein Structure, Tertiary , bcl-Associated Death Protein/chemistry , 14-3-3 Proteins/chemistry , 14-3-3 Proteins/genetics , 14-3-3 Proteins/metabolism , Amino Acid Sequence , Circular Dichroism , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Humans , Lipid Bilayers/metabolism , Molecular Sequence Data , Peptides/chemistry , Peptides/metabolism , Phosphorylation , Protein Binding , Protein Structure, Secondary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Surface Plasmon Resonance , Water/chemistry , bcl-Associated Death Protein/genetics , bcl-Associated Death Protein/metabolism
10.
Mol Cell Biol ; 30(19): 4698-711, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20679480

ABSTRACT

The Ras-RAF-mitogen-activated protein kinase (Ras-RAF-MAPK) pathway is overactive in many cancers and in some developmental disorders. In one of those disorders, namely, Noonan syndrome, nine activating C-RAF mutations cluster around Ser(259), a regulatory site for inhibition by 14-3-3 proteins. We show that these mutations impair binding of 14-3-3 proteins to C-RAF and alter its subcellular localization by promoting Ras-mediated plasma membrane recruitment of C-RAF. By presenting biophysical binding data, the 14-3-3/C-RAFpS(259) crystal structure, and cellular analyses, we indicate a mechanistic link between a well-described human developmental disorder and the impairment of a 14-3-3/target protein interaction. As a broader implication of these findings, modulating the C-RAFSer(259)/14-3-3 protein-protein interaction with a stabilizing small molecule may yield a novel potential approach for treatment of diseases resulting from an overactive Ras-RAF-MAPK pathway.


Subject(s)
14-3-3 Proteins/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Signal Transduction , ras Proteins/metabolism , 14-3-3 Proteins/chemistry , 14-3-3 Proteins/genetics , Animals , Binding Sites/genetics , Cell Line , Chlorocebus aethiops , Crystallization , Crystallography, X-Ray , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Kinetics , Microscopy, Confocal , Models, Molecular , Mutation , Noonan Syndrome/genetics , Noonan Syndrome/metabolism , Phosphorylation , Protein Binding , Protein Structure, Tertiary , Proto-Oncogene Proteins c-raf/chemistry , Proto-Oncogene Proteins c-raf/genetics , Serine/genetics , Serine/metabolism , Transfection , ras Proteins/genetics
11.
J Biol Chem ; 284(41): 28004-28020, 2009 Oct 09.
Article in English | MEDLINE | ID: mdl-19667065

ABSTRACT

BAD is a proapoptotic member of the Bcl-2 protein family that is regulated by phosphorylation in response to survival factors. Although much attention has been devoted to the identification of phosphorylation sites in murine BAD, little data are available with respect to phosphorylation of human BAD protein. Using mass spectrometry, we identified here besides the established phosphorylation sites at serines 75, 99, and 118 several novel in vivo phosphorylation sites within human BAD (serines 25, 32/34, 97, and 124). Furthermore, we investigated the quantitative contribution of BAD targeting kinases in phosphorylating serine residues 75, 99, and 118. Our results indicate that RAF kinases represent, besides protein kinase A, PAK, and Akt/protein kinase B, in vivo BAD-phosphorylating kinases. RAF-induced phosphorylation of BAD was reduced to control levels using the RAF inhibitor BAY 43-9006. This phosphorylation was not prevented by MEK inhibitors. Consistently, expression of constitutively active RAF suppressed apoptosis induced by BAD and the inhibition of colony formation caused by BAD could be prevented by RAF. In addition, using the surface plasmon resonance technique, we analyzed the direct consequences of BAD phosphorylation by RAF with respect to association with 14-3-3 and Bcl-2/Bcl-X(L) proteins. Phosphorylation of BAD by active RAF promotes 14-3-3 protein association, in which the phosphoserine 99 represented the major binding site. Finally, we show here that BAD forms channels in planar bilayer membranes in vitro. This pore-forming capacity was dependent on phosphorylation status and interaction with 14-3-3 proteins. Collectively, our findings provide new insights into the regulation of BAD function by phosphorylation.


Subject(s)
Ion Channels/chemistry , Ion Channels/metabolism , bcl-Associated Death Protein/chemistry , bcl-Associated Death Protein/metabolism , 14-3-3 Proteins/genetics , 14-3-3 Proteins/metabolism , Amino Acid Sequence , Animals , Cyclic AMP-Dependent Protein Kinases/metabolism , Humans , Ion Channels/genetics , Lipid Bilayers/metabolism , Mass Spectrometry , Mice , Molecular Sequence Data , NIH 3T3 Cells , Peptides/chemistry , Peptides/genetics , Peptides/metabolism , Phosphorylation , Protein Binding , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Sequence Alignment , bcl-Associated Death Protein/genetics , bcl-X Protein/genetics , bcl-X Protein/metabolism , p21-Activated Kinases/metabolism , raf Kinases/genetics , raf Kinases/metabolism
12.
J Biol Chem ; 284(5): 3183-3194, 2009 Jan 30.
Article in English | MEDLINE | ID: mdl-19049963

ABSTRACT

Mammalian 14-3-3 proteins play a crucial role in the activation process of RAF kinases. However, little is known about the selectivity of the mammalian 14-3-3 isoforms with respect to RAF association and activation. Using mass spectrometry, we analyzed the composition of the 14-3-3 isoforms attached to RAF kinases and found that B-RAF associates in vivo with 14-3-3 at much higher diversity than A- and C-RAF. We also examined in vitro binding of purified mammalian 14-3-3 proteins to RAF kinases using surface plasmon resonance techniques. While B- and C-RAF exhibited binding to all seven 14-3-3 isoforms, A-RAF bound with considerably lower affinities to epsilon, tau, and sigma 14-3-3. These findings indicate that 14-3-3 proteins associate with RAF isoforms in a pronounced isoform-specific manner. Because 14-3-3 proteins appear in dimeric forms, we addressed the question of whether both homo- and heterodimeric forms of 14-3-3 proteins participate in RAF signaling. For that purpose, the budding yeast Saccharomyces cerevisiae, possessing only two 14-3-3 isoforms (BMH1 and BMH2), served as testing system. By deletion of the single BMH2 gene, we found that both homo- and heterodimeric forms of 14-3-3 can participate in RAF activation. Furthermore, we show that A-, B-, and C-RAF activity is differentially regulated by its C-terminal and internal 14-3-3 binding domain. Finally, prohibitin, a scaffold protein that affects C-RAF activation in a stimulatory manner, proved to interfere with the internal 14-3-3 binding site in C-RAF. Together, our results shed more light on the complex mechanism of RAF activation, particularly with respect to activation steps that are mediated by 14-3-3 proteins and prohibitin.


Subject(s)
14-3-3 Proteins/physiology , Protein Isoforms/physiology , raf Kinases/metabolism , 14-3-3 Proteins/chemistry , 14-3-3 Proteins/metabolism , Base Sequence , Binding Sites , Biosensing Techniques , DNA Primers , Dimerization , Electrophoresis, Polyacrylamide Gel , HeLa Cells , Humans , Immunoprecipitation , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Tandem Mass Spectrometry
13.
J Biol Chem ; 283(40): 27239-54, 2008 Oct 03.
Article in English | MEDLINE | ID: mdl-18662992

ABSTRACT

In mammals the RAF family of serine/threonine kinases consists of three members, A-, B-, and C-RAF. Activation of RAF kinases involves a complex series of phosphorylations. Although the most prominent phosphorylation sites of B- and C-RAF are well characterized, little is known about regulatory phosphorylation of A-RAF. Using mass spectrometry, we identified here a number of novel in vivo phosphorylation sites in A-RAF. In particular, we found that Ser-432 participates in MEK binding and is indispensable for A-RAF signaling. On the other hand, phosphorylation within the activation segment does not contribute to epidermal growth factor-mediated activation. Furthermore, we show that the potential 14-3-3 binding domains in A-RAF are phosphorylated independently of its activation status. Of importance, we identified a novel regulatory domain in A-RAF (referred to as IH-segment) positioned between amino acids 248 and 267 that contains seven putative phosphorylation sites. Three of these sites, serines 257, 262, and 264, regulate A-RAF activation in a stimulatory manner. The spatial model of the A-RAF fragment, including residues between Ser-246 and Glu-277, revealed a switch of charge at the molecular surface of the IH-region upon phosphorylation, suggesting a mechanism in which the high accumulation of negative charges may lead to an electrostatic destabilization of protein-membrane interaction resulting in depletion of A-RAF from the plasma membrane. Together, we provide here for the first time a detailed analysis of in vivo A-RAF phosphorylation status and demonstrate that regulation of A-RAF by phosphorylation exhibits unique features compared with B- and C-RAF.


Subject(s)
Cell Membrane/metabolism , Models, Molecular , Proto-Oncogene Proteins A-raf/metabolism , Animals , COS Cells , Cell Membrane/chemistry , Cell Membrane/genetics , Chlorocebus aethiops , Enzyme Activation/genetics , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Mass Spectrometry , Phosphorylation , Proto-Oncogene Proteins A-raf/chemistry , Proto-Oncogene Proteins A-raf/genetics
14.
J Biol Chem ; 282(36): 26575-90, 2007 Sep 07.
Article in English | MEDLINE | ID: mdl-17613527

ABSTRACT

In mammals the RAF family of serine/threonine kinases consists of three members, A-, B-, and C-RAF. A prominent feature of RAF isoforms regards differences in basal and inducible kinase activities. To elucidate the nature of these differences, we studied the role of the nonconserved residues within the N-region (Negative-charge regulatory region). The nonconserved amino acids in positions -3 and +1 relative to the highly conserved serine 299 in A-RAF and serine 338 in C-RAF have so far not been considered as regulatory residues. Here we demonstrate the essential role of these residues in the RAF activation process. Substitution of tyrosine 296 in A-RAF to arginine led to a constitutively active kinase. In contrast, substitution of glycine 300 by serine (mimicking B- and C-RAF) acts in an inhibitory manner. Consistent with these data, the introduction of glycine in the analogous position of C-RAF (S339G mutant) led to a constitutively active C-RAF kinase. Based on the three-dimensional structure of the catalytic domain of B-RAF and using the sequences of the N-regions of A- and C-RAF, we searched by molecular modeling for the putative contact points between these two moieties. A tight interaction between the N-region residue serine 339 of C-RAF and arginine 398 of the catalytic domain was identified and proposed to inhibit the kinase activity of RAF proteins, because abrogation of this interaction contributes to RAF activation. Furthermore, tyrosine 296 in A-RAF favors a spatial orientation of the N-region segment, which enables a tighter contact to the catalytic domain, whereas a glutamine residue at this position in C-RAF abrogates this interaction. Considering this observation, we suggest that tyrosine 296, which is unique for A-RAF, is a major determinant of the low activating potency of this RAF isoform.


Subject(s)
Evolution, Molecular , Models, Molecular , Proto-Oncogene Proteins A-raf/chemistry , Amino Acid Substitution , Animals , COS Cells , Catalytic Domain/genetics , Chlorocebus aethiops , Enzyme Activation/genetics , Enzyme Induction/genetics , Humans , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Protein Structure, Tertiary/genetics , Proto-Oncogene Proteins A-raf/genetics , Proto-Oncogene Proteins A-raf/metabolism , Proto-Oncogene Proteins B-raf/chemistry , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins c-raf/chemistry , Proto-Oncogene Proteins c-raf/genetics , Proto-Oncogene Proteins c-raf/metabolism , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...