Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Blood Cancer J ; 4: e175, 2014 Jan 17.
Article in English | MEDLINE | ID: mdl-24442204

ABSTRACT

Multiple myeloma (MM) is an incurable haematological malignancy characterised by the proliferation of mature antibody-secreting plasma B cells in the bone marrow. MM can arise from initiating translocations, of which the musculoaponeurotic fibrosarcoma (MAF) family is implicated in ∼5%. MMs bearing Maf translocations are of poor prognosis. These translocations are associated with elevated Maf expression, including c-MAF, MAFB and MAFA, and with t(14;16) and t(14;20) translocations, involving c-MAF and MAFB, respectively. c-MAF is also overexpressed in MM through MEK/ERK activation, bringing the number of MMs driven by the deregulation of a Maf gene close to 50%. Here we demonstrate that MAFB and c-MAF are phosphorylated by the Ser/Thr kinase GSK3 in human MM cell lines. We show that LiCl-induced GSK3 inhibition targets these phosphorylations and specifically decreases proliferation and colony formation of Maf-expressing MM cell lines. Interestingly, bortezomib induced stabilisation of Maf phosphorylation, an observation that could explain, at least partially, the low efficacy of bortezomib for patients carrying Maf translocations. Thus, GSK3 inhibition could represent a new therapeutic approach for these patients.

2.
Oncogene ; 25(9): 1299-310, 2006 Mar 02.
Article in English | MEDLINE | ID: mdl-16247450

ABSTRACT

Maf b-Zip transcription factors are involved in both terminal differentiation and oncogenesis. To investigate this apparent contradiction, we used two different primary cell types and performed an extensive analysis of transformation parameters induced by Maf proteins. We show that MafA and c-Maf are potent oncogenes in chicken embryo fibroblasts, while MafB appears weaker. We also provide the first evidence that MafA can confer growth factor independence and promote cell division at low density. Moreover, using MafA as a model, we identified several parameters that are critical for Maf transforming activities. Indeed, MafA ability to induce anchorage-independent cell growth was sensitive to culture conditions. In addition, the transforming activity of MafA was dependent on its phosphorylation state, since mutation on Ser65 impaired its ability to induce growth at low density and anchorage-independent growth. We next examined transforming activity of large Maf proteins in embryonic neuroretina cells, where they are known to induce differentiation. Unlike v-Jun, MafA, MafB and c-Maf did not show oncogenic activity in these cells. Moreover, they counteracted transformation induced by constitutive activation of the Ras/Raf/MEK pathway. Taken together, our results show that Maf proteins could display antagonistic functions in oncogenesis depending on the cellular context, and support a dual role for Maf as both oncogenes and tumor suppressor-like proteins.


Subject(s)
Cell Transformation, Neoplastic/genetics , Maf Transcription Factors, Large/physiology , Proto-Oncogene Proteins c-maf/physiology , Animals , Cell Culture Techniques , Cell Division , Cell Proliferation , Chick Embryo/cytology , Fibroblasts , Genes, Tumor Suppressor , Humans , Oncogenes , Phosphorylation , Plasmids , Retina/cytology , Reverse Transcriptase Polymerase Chain Reaction
3.
Cell Death Differ ; 11(10): 1084-91, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15243584

ABSTRACT

Melanocytes are cells of the epidermis that synthesize melanin, which is responsible for skin pigmentation. Transformation of melanocytes leads to melanoma, a highly aggressive neoplasm, which displays resistance to apoptosis. In this report, we demonstrate that TNF-related apoptosis-inducing ligand (TRAIL), which was thought to kill only transformed cells, promotes very efficiently apoptosis of primary human melanocytes, leading to activation of caspases 8, 9 and 3, and the cleavage of vital proteins. Further, we show that stem cell factor (SCF), a physiologic melanocyte growth factor that activates both the phosphatidyl-inositol-3 kinase (PI3K) and the extracellular regulated kinase (ERK) pathways, strongly protects melanocytes from TRAIL and staurosporine killing. Interestingly, inhibition of PI3K or its downstream target AKT completely blocks the antiapoptotic effect of SCF, while inhibition of ERK has only a moderate effect. Our data indicate that protection evoked by SCF/PI3K/AKT cascade is not mediated by an increase in the intracellular level of FLIP. Further, only a sustained PI3K activity can protect melanocytes from apoptosis, thereby indicating that the PI3K/AKT pathway plays a pivotal role in melanocyte survival. The results gathered in this report bring new information on the molecular mechanisms involved in primary melanocyte apoptosis and survival that would help to better understand the process by which melanomas acquire their resistance to apoptosis.


Subject(s)
Apoptosis/drug effects , Melanocytes/drug effects , Membrane Glycoproteins/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Apoptosis Regulatory Proteins , Cells, Cultured , Enzyme Activation , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Keratinocytes/drug effects , Keratinocytes/metabolism , Melanocytes/cytology , Melanocytes/enzymology , Melanocytes/metabolism , Melanoma/pathology , Phosphoinositide-3 Kinase Inhibitors , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Signal Transduction/drug effects , Stem Cell Factor/pharmacology , TNF-Related Apoptosis-Inducing Ligand
4.
Biol Cell ; 93(1-2): 53-62, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11730323

ABSTRACT

The Raf/MEK/ERK signaling was the first MAP kinase cascade to be characterized. It is probably one of the most well known signal transduction pathways among biologists because of its implication in a wide variety of cellular functions as diverse -and occasionally contradictory- as cell proliferation, cell-cycle arrest, terminal differentiation and apoptosis. Discovery and understanding of this pathway have benefited from the combination of both genetic studies in worms and flies and biochemical studies in mammalian cells. However, ten years after, this field is still under debate and new molecular partners in the cascade continue to increase the complexity of its regulation. This review deals with the emergence of new concepts in the activation and regulation of the Raf/MEK/ERK module. In particular, the preponderant role of B-Raf is underlined, and the role of novel regulators such as KSR is discussed.


Subject(s)
MAP Kinase Kinase Kinase 1 , MAP Kinase Signaling System , Mitogen-Activated Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Animals , Enzyme Activation , Humans
5.
Mol Cell Biol ; 21(19): 6706-17, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11533257

ABSTRACT

Signals from the extracellular matrix are essential for the survival of many cell types. Dominant-negative mutants of two members of Rho family GTPases, Rac1 and Cdc42, mimic the loss of anchorage in primary mouse fibroblasts and are potent inducers of apoptosis. This pathway of cell death requires the activation of both the p53 tumor suppressor and the extracellular signal-regulated mitogen-activated protein kinases (Erks). Here we characterize the proapoptotic Erk signal and show that it differs from the classically observed survival-promoting one by the intensity of the kinase activation. The disappearance of the GTP-bound forms of Rac1 and Cdc42 gives rise to proapoptotic, moderate activation of the Raf-MEK-Erk cascade via a signaling pathway involving the kinases phosphatidlyinositol 3-kinase and Akt. Moreover, concomitant activation of p53 and inhibition of Akt are both necessary and sufficient to signal anoikis in primary fibroblasts. Our data demonstrate that the GTPases of the Rho family control three major components of cellular signal transduction, namely, p53, Akt, and Erks, which collaborate in the induction of apoptosis due to the loss of anchorage.


Subject(s)
Anoikis , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Proto-Oncogene Proteins/physiology , rho GTP-Binding Proteins/physiology , Animals , Apoptosis , Cell Nucleus/metabolism , Cells, Cultured , Extracellular Matrix/physiology , Fibroblasts/cytology , Fibroblasts/metabolism , MAP Kinase Kinase 1 , MAP Kinase Signaling System , Mice , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinase Kinases/metabolism , Mutation , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt , Tumor Suppressor Protein p53/metabolism , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/physiology , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/physiology , rho GTP-Binding Proteins/genetics
6.
Mol Cell Biol ; 21(14): 4441-52, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11416124

ABSTRACT

We previously described the identification of quail MafA, a novel transcription factor of the Maf bZIP (basic region leucine zipper) family, expressed in the differentiating neuroretina (NR). In the present study, we provide the first evidence that MafA is phosphorylated and that its biological properties strongly rely upon phosphorylation of serines 14 and 65, two residues located in the transcriptional activating domain within a consensus for phosphorylation by mitogen-activated protein kinases and which are conserved among Maf proteins. These residues are phosphorylated by ERK2 but not by p38, JNK, and ERK5 in vitro. However, the contribution of the MEK/ERK pathway to MafA phosphorylation in vivo appears to be moderate, implicating another kinase. The integrity of serine 14 and serine 65 residues is required for transcriptional activity, since their mutation into alanine severely impairs MafA capacity to activate transcription. Furthermore, we show that the MafA S14A/S65A mutant displays reduced capacity to induce expression of QR1, an NR-specific target of Maf proteins. Likewise, the integrity of serines 14 and 65 is essential for the MafA ability to stimulate expression of crystallin genes in NR cells and to induce NR-to-lens transdifferentiation. Thus, the MafA capacity to induce differentiation programs is dependent on its phosphorylation.


Subject(s)
Leucine Zippers , Mitogen-Activated Protein Kinase 1/metabolism , Proto-Oncogene Proteins/metabolism , Trans-Activators/metabolism , Amino Acid Sequence , Animals , Binding Sites , Eye Proteins/genetics , Glycoproteins/genetics , HeLa Cells , Humans , Lectins, C-Type , Lens, Crystalline , Maf Transcription Factors, Large , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinase 7 , Mitogen-Activated Protein Kinase 8 , Mitogen-Activated Protein Kinases/metabolism , Molecular Sequence Data , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/physiology , Rabbits , Receptors, Immunologic , Serine/genetics , Serine/metabolism , Trans-Activators/genetics , Trans-Activators/physiology , Transcription, Genetic , p38 Mitogen-Activated Protein Kinases
7.
J Biol Chem ; 276(37): 34958-65, 2001 Sep 14.
Article in English | MEDLINE | ID: mdl-11418594

ABSTRACT

The p42/p44 mitogen-activated protein (MAP) kinase is stimulated by various mitogenic stimuli, and its sustained activation is necessary for cell cycle G(1) progression and G(1)/S transition. G(1) progression and G(1)/S transition also depend on sequential cyclin-dependent kinase (CDK) activation. Here, we demonstrate that MAP kinase inhibition leads to accumulation of the CDK inhibitor p27(Kip1) in NIH 3T3 cells. Blocking the proteasome-dependent degradation of p27(Kip1) impaired this accumulation, suggesting that MAP kinase does not act on p27(Kip1) protein synthesis. In the absence of extracellular signals (growth factors or cell adhesion), genetic activation of MAP kinase decreased the expression of p27(Kip1) as assessed by cotransfection experiments and by immunofluorescence detection. Importantly, MAP kinase activation also decreased the expression of a p27(Kip1) mutant, which cannot be phosphorylated by CDK2, suggesting that MAP kinase-dependent p27(Kip1) regulation is CDK2-independent. Accordingly, expression of dominant-negative CDK2 did not impair the down-regulation of p27(Kip1) induced by MAP kinase activation. These data demonstrate that the MAP kinase pathway regulates p27(Kip1) expression in fibroblasts essentially through a degradation mechanism, independently of p27(Kip1) phosphorylation by CDK2. This strengthens the role of this CDK inhibitor as a key effector of G(1) growth arrest, whose expression can be controlled by extracellular stimuli-dependent signaling pathways.


Subject(s)
CDC2-CDC28 Kinases , Cell Cycle Proteins/metabolism , Cyclin E/physiology , Cyclin-Dependent Kinases/physiology , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinases/physiology , Protein Serine-Threonine Kinases/physiology , Tumor Suppressor Proteins , 3T3 Cells , Animals , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase Inhibitor p27 , Cysteine Endopeptidases/physiology , Enzyme Activation , Mice , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinase Kinases/physiology , Multienzyme Complexes/physiology , Phosphorylation , Proteasome Endopeptidase Complex
8.
Mol Cell Biol ; 21(8): 2659-70, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11283246

ABSTRACT

Thrombopoietin (TPO) regulates growth and differentiation of megakaryocytes. We previously showed that extracellular signal-regulated kinases (ERKs) are required for TPO-mediated full megakaryocytic maturation in both normal progenitors and a megakaryoblastic cell line (UT7) expressing the TPO receptor (Mpl). In these cells, intensity and duration of TPO-induced ERK signal are controlled by several regions of the cytoplasmic domain of Mpl. In this study, we explored the signaling pathways involved in this control. We show that the small GTPases Ras and Rap1 contribute together to TPO-induced ERK activation in UT7-Mpl cells and that they do so by activating different Raf kinases as downstream effectors: a Ras-Raf-1 pathway is required to initiate ERK activation while Rap1 sustains this signal through B-Raf. Indeed, (i) in cells expressing wild-type or mutant Mpl, TPO-induced Ras and Rap1 activation correlates with early and sustained phases of ERK signal, respectively; (ii) interfering mutants of Ras and Rap1 both inhibit ERK kinase activity and ERK-dependent Elk1 transcriptional activation in response to TPO; (iii) the kinetics of activation of Raf-1 and B-Raf by TPO follow those of Ras and Rap1, respectively; (iv) RasV12-mediated Elk1 activation was modulated by the wild type or interfering mutants of Raf-1 but not those of B-Raf; (v) Elk1 activation mediated by a constitutively active mutant of Rap1 (Rap1V12) is potentiated by B-Raf and inhibited by an interfering mutant of this kinase. UT7-Mpl cells represent the second cellular model in which Ras and Rap1 act in concert to modulate the duration of ERK signal in response to a growth factor and thereby the differentiation program. This is also, to our knowledge, the first evidence suggesting that Rap1 may play an active role in megakaryocytic maturation.


Subject(s)
Mitogen-Activated Protein Kinases/metabolism , Neoplasm Proteins , Proto-Oncogene Proteins c-raf/metabolism , Receptors, Cytokine , Thrombopoietin/pharmacology , rap1 GTP-Binding Proteins/metabolism , ras Proteins/metabolism , Animals , Cell Line , Enzyme Activation/drug effects , Humans , Mice , Mutation , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-raf/genetics , Receptors, Thrombopoietin , Signal Transduction , rap1 GTP-Binding Proteins/genetics
9.
Mol Cell Biol ; 20(19): 7068-79, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10982823

ABSTRACT

Ras-induced cell transformation is mediated through distinct downstream signaling pathways, including Raf, Ral-GEFs-, and phosphatidylinositol 3-kinase (PI 3-kinase)-dependent pathways. In some cell types, strong activation of the Ras-Raf-MEK-extracellular signal-regulated kinase (ERK) cascade leads to cell cycle arrest rather than cell division. We previously reported that constitutive activation of this pathway induces sustained proliferation of primary cultures of postmitotic chicken neuroretina (NR) cells. We used this model system to investigate the respective contributions of Ras downstream signaling pathways in Ras-induced cell proliferation. Three RasV12 mutants (S35, G37, and C40) which differ by their ability to bind to Ras effectors (Raf, Ral-GEFs, and the p110 subunit of PI 3-kinase, respectively) were able to induce sustained NR cell proliferation, although none of these mutants was reported to transform NIH 3T3 cells. Furthermore, they all repressed the promoter of QR1, a neuroretina growth arrest-specific gene. Overexpression of B-Raf or activated versions of Ras effectors Rlf-CAAX and p110-CAAX also induced NR cell division. The mitogenic effect of the RasC40-PI 3-kinase pathway appears to involve Rac and RhoA GTPases but not the antiapoptotic Akt (protein kinase B) signaling. Division induced by RasG37-Rlf appears to be independent of Ral GTPase activation and presumably requires an unidentified mechanism. Activation of either Ras downstream pathway resulted in ERK activation, and coexpression of a dominant negative MEK mutant or mKsr-1 kinase domain strongly inhibited proliferation induced by the three Ras mutants or by their effectors. Similar effects were observed with dominant negative mutants of Rac and Rho. Thus, both the Raf-MEK-ERK and Rac-Rho pathways are absolutely required for Ras-induced NR cell division. Activation of these two pathways by the three distinct Ras downstream effectors possibly relies on an autocrine or paracrine loop, implicating endogenous Ras, since the mitogenic effect of each Ras effector mutant was inhibited by RasN17.


Subject(s)
MAP Kinase Signaling System/physiology , Nerve Tissue Proteins/physiology , Protein Serine-Threonine Kinases , Retina/cytology , ras Proteins/physiology , 3T3 Cells , Animals , Cell Division , Cells, Cultured , Chick Embryo , Chloramphenicol O-Acetyltransferase/biosynthesis , Eye Proteins/biosynthesis , Eye Proteins/genetics , Eye Proteins/physiology , Feedback , Genes, ras , Guanine Nucleotide Exchange Factors , MAP Kinase Signaling System/genetics , Mice , Mitogen-Activated Protein Kinase Kinases/physiology , Phosphatidylinositol 3-Kinases/physiology , Promoter Regions, Genetic , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-raf/physiology , Recombinant Fusion Proteins/biosynthesis , Retina/metabolism , Transcription Factors/physiology , Transfection , rac GTP-Binding Proteins/physiology , ral GTP-Binding Proteins/physiology , rho GTP-Binding Proteins/physiology
10.
EMBO J ; 19(12): 2900-10, 2000 Jun 15.
Article in English | MEDLINE | ID: mdl-10856235

ABSTRACT

In melanocytes and melanoma cells, cAMP activates extracellular signal-regulated kinases (ERKs) and MEK-1 by an unknown mechanism. We demonstrate that B-Raf is activated by cAMP in melanocytes. A dominant-negative mutant of B-Raf, but not of Raf-1, blocked the cAMP-induced activation of ERK, indicating that B-Raf is the MEK-1 upstream regulator mediating this cAMP effect. Studies using Clostridium sordelii lethal toxin and Clostridium difficile toxin B have suggested that Rap-1 or Ras might transduce cAMP action. We show that Ras, but not Rap-1, is activated cell-specifically and mediates the cAMP-dependent activation of ERKs, while Rap-1 is not involved in this process in melanocytes. Our results suggest a novel, cell-specific mechanism involving Ras small GTPase and B-Raf kinase as mediators of ERK activation by cAMP. Also, in melanocytes, Ras or ERK activation by cAMP is not mediated through protein kinase A activation. Neither the Ras exchange factor, Son of sevenless (SOS), nor the cAMP-responsive Rap-1 exchange factor, Epac, participate in the cAMP-dependent activation of Ras. These findings suggest the existence of a melanocyte-specific Ras exchange factor directly regulated by cAMP.


Subject(s)
Cyclic AMP/metabolism , MAP Kinase Signaling System , Melanocytes/metabolism , Mitogen-Activated Protein Kinases/metabolism , ras Proteins/metabolism , Animals , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme Activation , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , MAP Kinase Kinase Kinases/metabolism , Melanoma, Experimental , Mice , Models, Biological , PC12 Cells , Proto-Oncogene Proteins c-raf/metabolism , Rats , Son of Sevenless Proteins/genetics , Son of Sevenless Proteins/metabolism , ras Proteins/genetics
11.
Oncogene ; 19(10): 1257-65, 2000 Mar 02.
Article in English | MEDLINE | ID: mdl-10713667

ABSTRACT

CDC25 dual-specificity phosphatases are essential regulators that activate cyclin-dependent kinases (CDKs) at critical stages of the cell cycle. In human cells, CDC25A and C are involved in the control of G1/S and G2/M respectively, whereas CDC25B is proposed to act both in S phase and G2/M. Evidence for an interaction between CDC25 phosphatases and members of the 14-3-3 protein family has been obtained in vitro and in vivo in several organisms. On the basis of the work performed with CDC25C, it has been proposed that phosphorylation is required to mediate the interaction with 14-3-3. Here we have examined the molecular basis of the interaction between CDC25B phosphatases and 14-3-3 proteins. We show that in the two-hybrid assay all three splice variants of CDC25B interact similarly and strongly with 14-3-3eta, beta and zeta proteins, but poorly with epsilon and Theta. In vitro, CDC25B interacts at a low level with 14-3-3beta, epsilon, zeta, eta, and Theta isoforms. This interaction is not increased upon phosphorylation of CDC25B by CHK1 and is not abolished by dephosphorylation. In contrast, a specific, strong interaction between CDC25B and 14-3-3zeta and eta isoforms is revealed by a deletion of 288 residues in the amino-terminal region of CDC25B. This interaction requires the integrity of Ser 323, although it is independent of phosphorylation. Thus, interaction between 14-3-3 proteins and CDC25B is regulated in a manner that is different from that with CDC25C. We propose that, in addition to a low affinity binding site that is available for all 14-3-3 isoforms, post-translational modification of CDC25B in vivo exposes a high-affinity binding site that is specific for the zeta and eta14-3-3 isoforms.


Subject(s)
Cell Cycle Proteins/metabolism , Proteins/metabolism , Tyrosine 3-Monooxygenase , cdc25 Phosphatases/metabolism , 14-3-3 Proteins , Binding Sites , Checkpoint Kinase 1 , Humans , Models, Theoretical , Phosphorylation , Protein Binding , Protein Isoforms/metabolism , Protein Kinases/metabolism , Serine , Two-Hybrid System Techniques
12.
J Biol Chem ; 273(38): 24939-47, 1998 Sep 18.
Article in English | MEDLINE | ID: mdl-9733801

ABSTRACT

Members of the raf oncogene family encode serine/threonine protein kinases, which activate the mitogen-activated protein kinase kinase MEKs (MAPK or ERK kinases) through direct interaction and phosphorylation. Several recent studies have revealed interesting differences between two members of this family, Raf-1 and B-Raf, regarding their activation, regulation, and kinase activity. In particular, B-Raf was shown to display higher MEK kinase activity than Raf-1. By using both two-hybrid analysis and coimmunoprecipitation experiments, we demonstrate here that B-Raf also markedly differs from Raf-1 by a higher affinity for MEK. We previously reported that the B-raf gene encodes multiple protein isoforms resulting from complex alternative splicing of two exons (exons 8b and 10) located upstream of B-Raf kinase domain. In the present study, we show that these naturally occurring modifications within the protein sequence markedly modulate both the biochemical and oncogenic properties of B-Raf. The presence of exon 10 sequences enhances the affinity for MEK, the basal kinase activity, as well as the mitogenic and transforming properties of full-length B-Raf, whereas the presence of exon 8b sequences seems to have opposite effects. Therefore, alternative splicing represents a novel regulatory mechanism for a protein of the Raf family.


Subject(s)
Alternative Splicing , MAP Kinase Kinase Kinase 1 , Oncogenes , Proto-Oncogene Proteins c-raf/genetics , Proto-Oncogene Proteins c-raf/metabolism , 3T3 Cells , Amino Acid Sequence , Animals , Cell Division , Cells, Cultured , Chick Embryo , Enzyme Activation , Exons , Humans , Isoenzymes/biosynthesis , Isoenzymes/genetics , Isoenzymes/metabolism , Kinetics , Mice , Neurons/cytology , Neurons/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-raf/biosynthesis , Recombinant Proteins/biosynthesis , Recombinant Proteins/metabolism , Retina , Transfection
13.
Curr Biol ; 8(1): 46-55, 1998 Jan 01.
Article in English | MEDLINE | ID: mdl-9427625

ABSTRACT

BACKGROUND: Ksr (kinase supressor of Ras) was identified as a regulator of the Ras-MAP kinase (mitogen-activated protein kinase) pathway by genetic screens in Drosophila and Caenorhabditis elegans. Ksr is a kinase with similarities to the three conserved regions of Raf kinases, especially within the kinase domain. To investigate whether these structural similarities correlated with common functional properties, we examined the ability of mKsr-1, the murine homolog of Ksr, to interact with components of the vertebrate MAP kinase pathway. RESULTS: In the yeast two-hybrid interaction assay, mKsr-1 did not bind to either Ras, B-Raf or Raf-1, but interacted strongly with both MEK-1 and MEK-2, activators of MAP kinase. The Ksr-MEK interaction was confirmed by co-immunoprecipitation experiments. Ectopically expressed mKsr-1 co-precipitated with endogenous MEK-1 in COS-1 cells, and endogenous Ksr and MEK co-precipitated from PC12 cells. Phosphorylation of MEK by mKsr-1 was not detected, however. In contrast, the MEK subpopulation complexed with mKsr-1 in COS-1 cells or PC12 cells did not display kinase activity. This ability of Ksr to block MEK in an inactive form correlated with a biological response: mKsr-1 did not transform NIH3T3 cells, and, furthermore, mKsr-1 reduced Ras-induced transformation. Similarly, mKsr-1 inhibited the proliferation of embryonic neuroretina cells induced by Ras and B-Raf but not that induced by MEK. CONCLUSIONS: Our results suggest a novel mechanism for Ksr in regulating the MAP kinase pathway, at least in part through an ability to interact with MEK.


Subject(s)
Cell Transformation, Neoplastic/drug effects , Mitogen-Activated Protein Kinase Kinases , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Signal Transduction , ras Proteins/antagonists & inhibitors , 3T3 Cells , Animals , COS Cells , Cell Division/drug effects , Chick Embryo , Epidermal Growth Factor/pharmacology , MAP Kinase Kinase 1 , MAP Kinase Kinase 2 , Mice , Nerve Growth Factors/pharmacology , PC12 Cells , Proto-Oncogene Proteins c-raf/metabolism , Rats , Retina/drug effects
14.
Oncogene ; 12(10): 2213-21, 1996 May 16.
Article in English | MEDLINE | ID: mdl-8668348

ABSTRACT

Recent studies suggested the existence of Ras/B-Raf/ MEK-1 complexes and a critical role for B-Raf in regulating the MAP kinase/ERKs signalling pathway. We report, here, that both Ras and MEK-1 proteins interact physically with B-Raf proteins in the yeast two-hybrid system. In addition, by screening a mouse brain cDNA library, we isolated additional B-Raf interacting proteins. These include three members of the 14-3-3 proteins family (eta, theta and zeta) and the MEK-2 protein. We also show that c-Raf-1, previously reported to interact with beta and zeta 14-3-3 proteins, also interacts with eta and theta 14-3-3 proteins in the two-hybrid system. By using different portions of the B-Raf protein, we mapped the regions of the protein involved in these interactions. Specifically, we have characterized B-Raf specific sequences required for an efficient interaction with MEK proteins. We show that, consequently, B-Raf interacts with MEK-1 and MEK-2 with a better affinity than does c-Raf-1, thus strengthening the notion that B-Raf is a stronger MEK activator than c-Raf-l. Our results also suggest that a MEK specific sequence, not present in MAP kinase kinases which are not activated by members of the Raf family, is required for the interaction with Raf proteins.


Subject(s)
Mitogen-Activated Protein Kinase Kinases , Protein Serine-Threonine Kinases/metabolism , Protein Sorting Signals/metabolism , Proto-Oncogene Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Signal Transduction/physiology , Tyrosine 3-Monooxygenase , 14-3-3 Proteins , Amino Acid Sequence , Animals , Base Sequence , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cricetinae , Humans , MAP Kinase Kinase 1 , MAP Kinase Kinase 2 , Mice , Molecular Sequence Data , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Proteins/genetics , Proteins/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-raf , Saccharomyces cerevisiae/enzymology , Sequence Homology, Amino Acid , ras Proteins/genetics , ras Proteins/metabolism
15.
J Biol Chem ; 270(40): 23381-9, 1995 Oct 06.
Article in English | MEDLINE | ID: mdl-7559496

ABSTRACT

The c-Rmil/B-raf proto-oncogene is a member of the mil/raf family encoding serine/threonine protein kinases shown to be involved in signal transduction from the membrane to the nucleus. We isolated from a mouse brain library B-raf cDNAs containing a previously unidentified 36-base pair alternatively spliced exon located between exons 8 and 9 and, therefore, designated exon 8b. Human and mouse B-raf mRNAs also contain the 120-base pair alternatively spliced exon 10 previously described in the avian c-Rmil gene. Independent splicing of these two exons, located between the conserved region 2 (CR2) and the catalytic domain (CR3) gives rise to mRNAs potentially encoding four distinct proteins. By using specific sera generated against different portions of B-Raf, we identified at least 10 protein isoforms in adult mouse tissues. Some isoforms, in the range of 69-72 kDa, are not recognized by antisera directed against peptides encoded by exons 1 and 2, indicating the existence of B-Raf proteins with two different NH2 extremities. The other isoforms, in the range of 79-99 kDa, contain the amino acids encoded by exons 1 and 2, by either or both of the alternatively spliced exons, and, possibly, by another of the unidentified exon. Analysis of B-raf mRNA expression by reverse transcriptase-polymerase chain reaction and immunocharacterization of B-Raf proteins in different tissues of the adult mouse showed a tissue-specific pattern of B-Raf isoforms expression. Interestingly, isoforms containing amino acids encoded by exon 10 are specifically expressed in neural tissues. Taken together, these results suggest that distinct B-Raf proteins could be involved, in a tissue-specific manner, in signal transduction pathways.


Subject(s)
Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogenes , Alternative Splicing , Amino Acid Sequence , Animals , Antibody Specificity , Base Sequence , Birds , DNA Primers/genetics , DNA, Complementary/genetics , Exons , Gene Expression , Humans , Male , Mice , Molecular Sequence Data , Protein Serine-Threonine Kinases/immunology , Proto-Oncogene Mas , Proto-Oncogene Proteins/immunology , Proto-Oncogene Proteins c-raf , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Tissue Distribution
16.
Oncogene ; 10(8): 1647-51, 1995 Apr 20.
Article in English | MEDLINE | ID: mdl-7731720

ABSTRACT

The B-raf/c-Rmil proto-oncogene belongs to the raf/mil family of serine/threonine protein kinases. It encodes multiple protein isoforms resulting from alternative splicing of two exons located upstream of the kinase domain. Recent studies suggested that B-Raf could be the intermediate molecule between Ras and Mek-1 (MAP Kinase Kinase) in signalling pathways specific of neural cells. However, there has been no evidence for a direct interaction between B-Raf and Mek-1. We report here that different B-Raf isoforms can be co-immunoprecipitated with anti-Mek-1 antisera in COS-1 cells and that the kinase activity of B-Raf is not required for its interaction with Mek-1. We also show that all B-Raf isoforms tested phosphorylate Mek-1 in a time-dependent manner, whereas kinase defective mutants fail to do so. Finally, we demonstrate that the constitutively activated S218D, S222D and S218D/S222D mutants of Mek-1 interact similarly with B-Raf. However, only the S218D and S222D mutants, and not the S218D/S222D double mutant, can be phosphorylated by B-Raf isoforms. Therefore, serine residues 218 and 222, previously shown to regulate Mek-1 activity, appear to be the major phosphorylation sites by B-Raf in vitro.


Subject(s)
Mitogen-Activated Protein Kinase Kinases , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/physiology , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/physiology , Serine/metabolism , Animals , Cell Line , MAP Kinase Kinase 1 , Phosphorylation , Precipitin Tests , Protein Serine-Threonine Kinases/immunology , Protein-Tyrosine Kinases/immunology , Proto-Oncogene Proteins/immunology , Proto-Oncogene Proteins c-raf
17.
Oncogene ; 10(6): 1159-65, 1995 Mar 16.
Article in English | MEDLINE | ID: mdl-7535416

ABSTRACT

The B-raf/c-Rmil proto-oncogene belongs to the raf/mil family of serine/threonine protein kinases. It encodes multiple protein isoforms previously shown to be expressed predominantly in neural tissues. We report here that B-Raf proteins of 95 and 72 kDa are also expressed in various human and murine hematopoietic cell lines. Their relative level of expression is variable depending on the cell line examined. The highest level of expression of p95B-raf was found in UT-7 cells, a human pluripotent cell line established from a patient with a megakaryoblastic leukemia. These cells are able to differentiate toward erythroid or myeloid lineage phenotypes in presence of erythropoietin (EPO) or granulocyte-macrophage colony-stimulating factor (GM-CSF) respectively. We show that treatment of UT-7 cells with EPO, GM-CSF or stem cell factor (SCF) rapidly induces phosphorylation of p95B-raf as indicated by a shift of electrophoretic mobility. This increase in phosphorylation is correlated with a three-fold increase of B-Raf kinase activity. B-Raf activation also increases in a dose-dependent manner in response to EPO and GM-CSF. We also show that both p95B-raf and p72B-raf can be activated by IL-3 in murine BAF-3 pro-B cells and by anti-CD3 in human Jurkat cells, respectively. These observations provide the first evidence that the B-Raf kinase is involved in signal transduction pathways regulating proliferation and differentiation of hematopoietic cells of both myeloid and lymphoid lineages.


Subject(s)
Isoenzymes/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Cell Line , Enzyme Activation , Erythropoietin/pharmacology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Hematopoietic Cell Growth Factors/pharmacology , Hematopoietic Stem Cells , Humans , Isoenzymes/genetics , Leukemia/enzymology , Mice , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Mas , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-raf , Stem Cell Factor , Tumor Cells, Cultured
18.
C R Seances Soc Biol Fil ; 189(1): 71-85, 1995.
Article in French | MEDLINE | ID: mdl-7648369

ABSTRACT

The c-Rmil/B-raf proto-oncogene belongs to the mil/raf family encoding serine/threonine protein kinases shown to be involved in signal transduction from the membrane to the nucleus. We previously showed that the avian c-Rmil gene encodes two proteins of 94 and 95 kDa resulting from the alternative splicing of a 120 bp exon encoding 40 aminoacids (exon 10). We isolated from a mouse brain library B-raf cDNAs containing this exon 10 and a previously unidentified 36 bp insert which constitutes an additional alternatively spliced exon designated exon 8b. These two exons are located between the CR2 region and the catalytic domain of the protein. By using specific sera generated against different regions of the B-Raf protein, we identified 10 B-Raf isoforms and we defined their structure and their expression pattern in adult mouse tissues. The B-Raf proteins are mainly expressed in neural tissues and, interestingly, isoforms containing aminoacids encoded by exon 10 are specifically expressed in these tissues. We also show that several B-Raf isoforms interact with the Mek-1 protein (MAP kinase kinase) and phosphorylate this protein on serine residues 218 and 222.


Subject(s)
Isoenzymes/genetics , MAP Kinase Kinase Kinase 1 , Protein Serine-Threonine Kinases/genetics , Proto-Oncogenes/genetics , Animals , Blotting, Western , Drug Residues , Exons , Isoenzymes/metabolism , Mice , Phosphorylation , Precipitin Tests , Protein Serine-Threonine Kinases/metabolism , Rabbits , Serine/metabolism
19.
Folia Biol (Praha) ; 40(5): 225-35, 1994.
Article in English | MEDLINE | ID: mdl-7895853

ABSTRACT

Oncogene transduction, the process by which a cellular gene is captured by a retrovirus was mainly described in vivo. We have developed a biological system allowing stepwise analysis of transduction mechanisms in tissue culture. Avian neuroretina (NR) cells dissected at the 8th day of embryonic development rapidly cease to divide and differentiate in culture. Serial passaging of a retrovirus that does not carry an oncogene on such cultures leads with a high frequency to the emergence of new viruses that have transduced oncogenes from the mil/raf family of serine/threonine kinases. These viruses have been selected by their ability to induce NR cell division. This experimental system allowed the isolation of the following molecular intermediates generated during the successive steps of oncogene transduction: a chimeric transcript containing viral and cellular sequences joined together by an alternative splicing mechanism; then a complete retrovirus with a 5' end identical to that of chimeric RNA; finally, a retrovirus that has acquired additional gag sequences and consequently, an increased replicative capacity. Structural analysis of these molecules led us to propose a general model for oncogene transduction in which the key step is the synthesis of chimeric RNAs. This model also explains generation of the vast majority of acutely transforming retroviruses isolated in vivo.


Subject(s)
Oncogenes/genetics , Retroviridae/genetics , Animals , Avian Leukosis Virus/genetics , Base Sequence , Genes, Viral , Humans , Molecular Sequence Data , Signal Transduction , Transduction, Genetic
20.
J Virol ; 67(11): 6853-6, 1993 Nov.
Article in English | MEDLINE | ID: mdl-8411388

ABSTRACT

We previously reported that serial passaging of Rous-associated virus type 1 in nondividing chicken embryo neuroretina cells leads to reproducible generation of acutely mitogenic retroviruses that transduced the catalytic domain of c-mil/c-raf or c-Rmil/B-raf. On the basis of structural analysis of several retroviruses, we proposed that the early step of oncogene transduction is the constitution of alternatively spliced leader-delta onc-poly(A) transcripts. Here, we show that neuroretina cells do synthesize hybrid leader-delta mil and leader-delta Rmil RNAs and that these RNAs exhibit mitogenic properties and serve as templates for the generation of transducing retorviruses.


Subject(s)
Avian Leukosis Virus/genetics , Avian Leukosis/genetics , Alternative Splicing , Amino Acid Sequence , Animals , Avian Leukosis/microbiology , Base Sequence , Chickens , DNA Primers , Molecular Sequence Data , Oncogenes , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-raf , RNA, Messenger/genetics , RNA, Viral/genetics , Recombination, Genetic , Retina/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL