13 research outputs found

    Unterschiede und Ähnlichkeiten in der Regulierung der RAF Isoformen : Identifizierung der neuen A-RAF Phosphorylierungsstellen

    No full text
    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. The physiological role and the function of these sites were investigated subsequently by amino acid exchange at the relevant positions. In particular, we found that S432 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. Regarding regulation of A-RAF activity by 14-3-3 proteins, we show that A-RAF activity is regulated differentially by its C-terminal and internal 14-3-3 binding domain. Furthermore, by use of SPR technique, we found that 14-3-3 proteins associate with RAF in an isoform-specific manner. Of importance, we identified a novel regulatory domain in A-RAF (referred to as IH-segment) positioned between amino acids 248 and 267, which 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 S246 and E277 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. Activation of B- and C-RAF is regulated by phosphorylation at conserved residues within the negative-charge regulatory region (N-region). Identification of phosphopeptides covering the sequence of the N-region led to the conclusion that, similar to B- and C-RAF, kinase activity of A-RAF is regulated by phosphorylation of the N-region. Abrogation of A-RAF activity by S299A substitution and elevated activity of the A-RAF-Y301D-Y302D mutant confirmed this conclusion. In addition, we studied the role of the non-conserved residues within the N-region in the activation process of RAF kinases. The non-conserved amino acids in positions –3 and +1 relative to the highly conserved S299 in A-RAF and S338 in C-RAF have so far not been considered as regulatory residues. Here, we demonstrate that Y296R substitution in A-RAF led to a constitutively active kinase. In contrast, G300S substitution (mimicking B- and C-RAF) acts in an inhibitory manner. These data were confirmed by analogous mutations in C-RAF. Based on the three-dimensional structure of the catalytic domain of B-RAF, a tight interaction between the N-region residue S339 and the catalytic domain residue R398 was identified in C-RAF and proposed to inhibit the kinase activity of RAF proteins. Furthermore, Y296 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 Y296, which is unique for A-RAF, is a major determinant of the low activating potency of this RAF isoform. Finally, the residues R359 in A-RAF and R398 in C-RAF, which interact with the N-region, are also involved in binding of phosphatidic acid. Substitution of this conserved arginine by alanine resulted in accumulation of hyper-phosphorylated form of RAF, suggesting that this residue play a crucial role in phosphorylation-mediated feedback regulation of A- and C-RAF. Collectively, 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.Die Protein-Familie der Serin/Threonin-spezifischen RAF-Kinasen umfasst in SĂ€ugetieren drei Mitglieder, A-, B- und C-RAF. Bei der Aktivierung dieser Kinasen spielen Phosphorylierungs-ereignisse eine entscheidende Rolle. Im Gegensatz zu B- und C-RAF, deren Phosphorylierungsstellen ausgiebig charakterisiert sind, blieb die Phosphorylierung von A-RAF weitgehend unerforscht. In der vorliegenden Arbeit wurden unter Verwendung der massenspektrometrischen Analyse zahlreiche neue in vivo A-RAF-Phosphorylierungsstellen identifiziert. Die physiologische Relevanz und die Funktion dieser Stellen wurden anschließend durch AminosĂ€urenaustausch an den relevanten Positionen untersucht. Dabei wurde festgestellt, dass S432 in der A-RAF-Bindung zu MEK involviert und fĂŒr die Signalweiterleitung unverzichtbar ist. Hingegen ist die EGF-bedingte A-RAF-Aktivierung nicht von der Phosphorylierung innerhalb des Aktivierungssegments abhĂ€ngig. Hinsichtlich der Regulation von A-RAF-Aktivierung durch 14-3-3-Proteine, wurde hier gezeigt, dass die katalytische AktivitĂ€t von A-RAF durch die C-terminale und die interne 14-3-3-BindungsdomĂ€nen unterschiedlich reguliert wird. Weiterhin wurde mittels SPR-Verfahren festgestellt, dass die Interaktion von 14-3-3-Proteinen mit RAF-Kinasen einen isoformspezifischen Charakter trĂ€gt. Von entscheidender Bedeutung war die Entdeckung einer neuen regulatorischen DomĂ€ne (hier als IH-Segment bezeichnet), die in der A-RAF-Sequenz die AminosĂ€uren 248 bis 267 umfasst und sieben A-RAF-spezifische Phosphorylierungsstellen enthĂ€lt. Drei dieser Stellen, S257, S262 und S264, erwiesen sich als positive Regulatoren der A-RAF-Aktivierung. Das rĂ€umliche Modell dieses A-RAF-Fragments deckte eine „Ladungsumkehr“ an der molekularen OberflĂ€che der IH-Region infolge der Phosphorylierung auf. Dieser Befund begrĂŒndete den Vorschlag eines Regulations-mechanismus, in dem die starke Akkumulierung der negativen Ladungen zu einer elektrostatischen Destabilisierung der Protein-Membran-Interaktion fĂŒhrt, was die VerdrĂ€ngung der A-RAF-Kinase von der Plasma-Membran zur Folge haben könnte. Die Aktivierung von B- und C-RAF wird durch Phosphorylierung der sogenannten „negativ geladenen“ Region (N-Region) reguliert. Die Identifizierung mehrerer Phosphopeptide aus der N-Region von A-RAF veranlasste die Schlussfolgerung, dass die A-RAF-AktivitĂ€t ebenfalls durch die Phosphorylierung innerhalb dieser Region gesteuert werden könnte. In der Tat, die Aufhebung der A-RAF-AktivitĂ€t durch die S299A-Substitution und die erhöhte AktivitĂ€t der A-RAF-Y301D-Y302D-Mutante bestĂ€tigen diese Aussage. DarĂŒberhinaus wurde die Rolle der nichtkonservierten AminosĂ€uren an den Positionen –3 und +1 relativ zum S299 in A-RAF und S338 in C-RAF im Aktivierungsprozess der RAF-Kinasen untersucht, nachdem diese ursprĂŒnglich nicht als regulatorische Stellen erkannt wurden. Es wird hier demonstriert, dass Y296R-Substitution der A-RAF-Kinase eine konstitutive AktivitĂ€t verleiht. Hingegen wirkte die G300S-Substitution, die von B- und C-RAF abgeleitet wurde, inhibitorisch. Diese Befunde wurden durch die analogen Mutationen in C-RAF bestĂ€tigt. Basierend auf der dreidimensionalen Struktur der katalytischen DomĂ€ne von B-RAF wurde eine Interaktion zwischen der N-Region und der katalytischen DomĂ€ne in A- und C-RAF festgestellt, die zu einer Inhibierung der AktivitĂ€t fĂŒhren soll. DarĂŒberhinaus wurde gezeigt, dass die rĂ€umliche Ausrichtung von Y296 in der N-Region von A-RAF einen engen Kontakt mit der katalytischen DomĂ€ne ermöglicht; dagegen hebt Glutamin in dieser Position die Interaktion auf. In Anbetracht dieser Befunde wurde vorschlagen, dass das A-RAF-spezifische Y296 das niedrige Aktivierungs-potential dieser RAF-Isoform determiniert. In diesem Zusammenhang wurde auch gefunden, dass die AminosĂ€uren R359 in A-RAF und R398 in C-RAF eine duale Funktion besitzen, indem sie sowohl mit der N-Region als auch mit Lipiden in Wechselwirkung treten können. Substitution dieser konservierten Arginine durch Alanin fĂŒhrte zur Akkumulierung der hyperphosphorylierten Formen der RAF-Kinasen, was die Schlussfolgerung erlaubt, dass diese Reste eine wichtige Rolle in der ERK-vermittelten Feedback-Regulation von A- und C-RAF spielen. Insgesamt wird hier zum ersten Mal eine detaillierte Analyse der in vivo A-RAF-Phosphorylierung geliefert und gezeigt, dass die phosphorylierungsvermittelte Regulation von A-RAF einzigartige Merkmale innerhalb der Familie von RAF-Kinasen aufweist

    It takes two to tango—signalling by dimeric Raf kinases

    Get PDF
    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.SFI Grant No. 06/CE/B1129Deposited by bulk impor

    The calcitonin receptor is the main mediator of LAAMA's body weight lowering effects in male mice

    Get PDF
    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

    Stabilization of C-RAF:KSR1 complex by DiRas3 reduces availability of C-RAF for dimerization with B-RAF

    Get PDF
    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.Science Foundation IrelandGerman Research Foundatio

    BAD Contributes to RAF-mediated Proliferation and Cooperates with B-RAF-V600E in Cancer Signaling*

    No full text
    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

    Single Substitution within the RKTR Motif Impairs Kinase Activity but Promotes Dimerization of RAF Kinase*

    No full text
    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

    Regulation of RAF activity by 14-3-3 proteins: RAF kinases associate functionally with both homo- and heterodimeric forms of 14-3-3 proteins

    No full text
    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

    Impaired Binding of 14-3-3 to C-RAF in Noonan Syndrome Suggests New Approaches in Diseases with Increased Ras Signaling▿

    No full text
    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 Ser259, 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-RAFpS259 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-RAFSer259/14-3-3 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
    corecore