Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Oncogenesis ; 12(1): 20, 2023 Apr 12.
Article in English | MEDLINE | ID: mdl-37045830

ABSTRACT

Spry2 is a molecular modulator of tyrosine kinase receptor signaling pathways that has cancer-type-specific effects. Mammalian Spry2 protein undergoes tyrosine and serine phosphorylation in response to growth factor stimulation. Spry2 expression is distinctly altered in various cancer types. Inhibition of the proteasome functionality results in reduced intracellular Spry2 degradation. Using in vitro and in vivo assays, we show that protein kinase D (PKD) phosphorylates Spry2 at serine 112 and interacts in vivo with the C-terminal half of this protein. Importantly, missense mutation of Ser112 decreases the rate of Spry2 intracellular protein degradation. Either knocking down the expression of all three mammalian PKD isoforms or blocking their kinase activity with a specific inhibitor contributes to the stabilization of Spry2 wild-type protein. Downregulation of CSN3, a component of the COP9/Signalosome that binds PKD, significantly increases the half-life of Spry2 wild-type protein but does not affect the stability of a Spry2 after mutating Ser112 to the non-phosphorylatable residue alanine. Our data demonstrate that both PKD and the COP9/Signalosome play a significant role in control of Spry2 intracellular stability and support the consideration of the PKD/COP9 complex as a potential therapeutic target in tumors where Spry2 expression is reduced.

2.
Biochim Biophys Acta Rev Cancer ; 1874(2): 188445, 2020 12.
Article in English | MEDLINE | ID: mdl-33035641

ABSTRACT

SOS1 and SOS2 are the most universal and widely expressed family of guanine exchange factors (GEFs) capable or activating RAS or RAC1 proteins in metazoan cells. SOS proteins contain a sequence of modular domains that are responsible for different intramolecular and intermolecular interactions modulating mechanisms of self-inhibition, allosteric activation and intracellular homeostasis. Despite their homology, analyses of SOS1/2-KO mice demonstrate functional prevalence of SOS1 over SOS2 in cellular processes including proliferation, migration, inflammation or maintenance of intracellular redox homeostasis, although some functional redundancy cannot be excluded, particularly at the organismal level. Specific SOS1 gain-of-function mutations have been identified in inherited RASopathies and various sporadic human cancers. SOS1 depletion reduces tumorigenesis mediated by RAS or RAC1 in mouse models and is associated with increased intracellular oxidative stress and mitochondrial dysfunction. Since WT RAS is essential for development of RAS-mutant tumors, the SOS GEFs may be considered as relevant biomarkers or therapy targets in RAS-dependent cancers. Inhibitors blocking SOS expression, intrinsic GEF activity, or productive SOS protein-protein interactions with cellular regulators and/or RAS/RAC targets have been recently developed and shown preclinical and clinical effectiveness blocking aberrant RAS signaling in RAS-driven and RTK-driven tumors.


Subject(s)
Mutation , Neoplasms/genetics , Son of Sevenless Proteins/genetics , Son of Sevenless Proteins/metabolism , Allosteric Regulation , Animals , Homeostasis , Humans , Mice , Neoplasms/metabolism , rac1 GTP-Binding Protein/metabolism , ras Proteins/metabolism
3.
Oncogenesis ; 8(1): 2, 2019 Jan 04.
Article in English | MEDLINE | ID: mdl-30631038

ABSTRACT

Sos1 is an universal, widely expressed Ras guanine nucleotide-exchange factor (RasGEF) in eukaryotic cells. Its N-terminal HD motif is known to be involved in allosteric regulation of Sos1 GEF activity through intramolecular interaction with the neighboring PH domain. Here, we searched for other cellular proteins also able to interact productively with the Sos1 HD domain. Using a yeast two-hybrid system, we identified the interaction between the Sos1 HD region and CSN3, the third component of the COP9 signalosome, a conserved, multi-subunit protein complex that functions in the ubiquitin-proteasome pathway to control degradation of many cellular proteins. The interaction of CSN3 with the HD of Sos1 was confirmed in vitro by GST pull-down assays using truncated mutants and reproduced in vivo by co-immunoprecipitation with the endogenous, full-length cellular Sos1 protein. In vitro kinase assays showed that PKD, a COP9 signalosome-associated-kinase, is able to phosphorylate Sos1. The intracellular levels of Sos1 protein were clearly diminished following CSN3 or PKD knockdown. A sizable fraction of the endogenous Sos1 protein was found ubiquitinated in different mammalian cell types. A significant reduction of RasGTP formation upon growth factor stimulation was also observed in CSN3-silenced as compared with control cells. Our data suggest that the interaction of Sos1 with the COP9 signalosome and PKD plays a significant role in maintenance of cellular Sos1 protein stability and homeostasis under physiological conditions and raises the possibility of considering the CSN/PKD complex as a potential target for design of novel therapeutic drugs.

6.
PLoS One ; 9(12): e114837, 2014.
Article in English | MEDLINE | ID: mdl-25514808

ABSTRACT

The Shoc2 protein has been implicated in the positive regulation of the Ras-ERK pathway by increasing the functional binding interaction between Ras and Raf, leading to increased ERK activity. Here we found that Shoc2 overexpression induced sustained ERK phosphorylation, notably in the case of EGF stimulation, and Shoc2 knockdown inhibited ERK activation. We demonstrate that ectopic overexpression of human Shoc2 in PC12 cells significantly promotes neurite extension in the presence of EGF, a stimulus that induces proliferation rather than differentiation in these cells. Finally, Shoc2 depletion reduces both NGF-induced neurite outgrowth and ERK activation in PC12 cells. Our data indicate that Shoc2 is essential to modulate the Ras-ERK signaling outcome in cell differentiation processes involved in neurite outgrowth.


Subject(s)
Epidermal Growth Factor/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Neurites/metabolism , Animals , Cell Line, Tumor , Enzyme Activation/genetics , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/biosynthesis , MAP Kinase Signaling System , PC12 Cells , Phosphorylation , RNA Interference , RNA, Small Interfering , Rats , ras Proteins/genetics , ras Proteins/metabolism
7.
J Med Chem ; 53(18): 6618-28, 2010 Sep 23.
Article in English | MEDLINE | ID: mdl-20722422

ABSTRACT

Mitogen-activated protein kinase-interacting kinases 1 and 2 (MNK1 and MNK2) phosphorylate the oncogene eIF4E on serine 209. This phosphorylation has been reported to be required for its oncogenic activity. To investigate if pharmacological inhibition of MNK1 could be useful for the treatment of cancers, we pursued a comprehensive virtual screening approach to rapidly identify pharmacological tools for target validation and to find optimal starting points for a plausible medicinal chemistry project. A collection of 1236 compounds, selected from a library of 42 168 compounds and a database of 18.8 million structures, were assayed. Of the identified hits, 26 were found to have IC(50) values less than 10 µM (2.10% hit rate). The most potent compound had an IC(50) value of 117 nM, and 73.1% of these hits were fragments. The hits were characterized by a high ligand efficiency (0.32-0.52 kcal/mol per heavy atom). Ten different chemical scaffolds were represented, giving a chemotype/hit ratio of 0.38.


Subject(s)
Antineoplastic Agents/chemical synthesis , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Crystallography, X-Ray , Databases, Factual , Drug Screening Assays, Antitumor , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Models, Molecular , Protein Conformation , Protein Serine-Threonine Kinases/chemistry , Quantitative Structure-Activity Relationship
8.
J Pharmacol Exp Ther ; 324(3): 1093-101, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18089842

ABSTRACT

Melanoma is the most aggressive skin cancer and a serious health problem worldwide because of its increasing incidence and the lack of satisfactory chemotherapy for late stages of the disease. The marine depsipeptide Aplidin (plitidepsin) is an antitumoral agent under phase II clinical development against several neoplasias, including melanoma. We report that plitidepsin has a dual effect on the human SK-MEL-28 and UACC-257 melanoma cell lines; at low concentrations (

Subject(s)
Apoptosis/physiology , Cell Cycle/physiology , Depsipeptides/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , Melanoma/enzymology , rac1 GTP-Binding Protein/metabolism , Apoptosis/drug effects , Cell Cycle/drug effects , Depsipeptides/therapeutic use , Enzyme Activation/drug effects , Enzyme Activation/physiology , Humans , Melanoma/drug therapy , Melanoma/pathology , Peptides, Cyclic , Tumor Cells, Cultured
9.
Cell Signal ; 19(11): 2277-85, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17689925

ABSTRACT

Sprouty2 has been widely implicated in the negative regulation of the fibroblast growth factor receptor-extracellular regulated kinase (ERK) pathway. Sprouty2 directly interacts with the adapter protein Grb2, member of the receptor tyrosine kinase-induced signaling pathways. In considering the functional role of Grb2, we investigated whether the interaction with this protein was responsible for ERK pathway inhibition. We found that the binding between Sprouty2 and Grb2 is constitutive, independent of Sprouty2 tyrosine phosphorylation, although it is increased when fibroblast growth factor receptor is activated. This connection is mediated by the N-terminal SH3 domain of Grb2 and two Sprouty2 proline-rich stretches (residues 59-64 and 303-307). Most importantly, a double Sprouty2 mutant (hSpry2 P59AP304A), which is unable to bind Grb2, developed at a similar inhibition level of fibroblast growth factor receptor-ERK pathway than that which originated from Sprouty2 wt. These results are evidence that the Sprouty2 mechanism of ERK inhibition is independent of Grb2 binding.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , GRB2 Adaptor Protein/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Proline/metabolism , Amino Acid Sequence , Animals , Binding Sites , Fluorescence Resonance Energy Transfer , GRB2 Adaptor Protein/chemistry , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Membrane Proteins , Mice , Molecular Sequence Data , Mutation/genetics , NIH 3T3 Cells , Protein Binding , Protein Interaction Mapping , Protein Transport , Signal Transduction , ras Proteins/metabolism , src Homology Domains
10.
Mol Pharmacol ; 70(5): 1654-63, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16928956

ABSTRACT

Plitidepsin (aplidin) is a marine cyclic depsipeptide in phase II clinical development against several neoplasias. Plitidepsin is a potent inducer of apoptosis through the sustained activation of Jun N-terminal kinase (JNK). We have reported that this activation depends on the early induction of oxidative stress, activation of Rac1 small GTPase, and the later down-regulation of MKP-1 phosphatase. Using Scatchard and saturation binding analyses, we have found that (14)C-labeled plitidepsin binds to a moderately high-affinity receptor (K(d) of 44.8 +/- 3.1 and 35.5 +/- 4.8 nM, respectively) in MDA-MB-231 breast cancer cells. Two minutes after addition to cells, half of the drug was membrane-bound and was subsequently found in the cytosolic fraction. At 4 degrees C, plitidepsin cellular binding was around 10-fold lower than at 37 degrees C but sufficed to induce cell death, suggesting that this process is triggered from the membrane. Depletion of plasma membrane cholesterol by short treatment with methyl-beta-cyclodextrin diminished plitidepsin binding and Rac1 and JNK activation. Rac1 is targeted to the plasma membrane by plitidepsin as shown by subcellular fractioning and immunofluorescence analysis followed by confocal microscopy. Methyl-beta-cyclodextrin blocked this effect. A subline of HeLa cells (HeLa-R), partially resistant to plitidepsin, showed similar affinity (K(d) of 79.5 +/- 2.5 versus 37.7 +/- 8.2 nM) but 7.5-fold lower binding capacity than wild-type HeLa cells. Moreover, HeLa-R cells had lower total (71%) and membrane (67%) cholesterol content and membrane-bound Rac1, and showed no Rac1 activation upon plitidepsin treatment. In conclusion, cellular plitidepsin uptake and induction of apoptosis via activation of the Rac1-JNK pathway is membrane-cholesterol dependent.


Subject(s)
Cell Membrane/drug effects , Cholesterol/metabolism , Depsipeptides/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , rac1 GTP-Binding Protein/metabolism , Cholesterol/deficiency , Depsipeptides/pharmacokinetics , Drug Resistance, Neoplasm , Enzyme Activation/drug effects , HeLa Cells , Humans , Peptides, Cyclic , Protein Transport/drug effects , Tumor Cells, Cultured
11.
Mol Biol Cell ; 17(8): 3591-7, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16760435

ABSTRACT

hSos1 is a Ras guanine-nucleotide exchange factor. It was suggested that the carboxyl-terminal region of hSos1 down-regulates hSos1 functionality and that the intrinsic guanine-nucleotide exchange activity of this protein may be different before and after stimulation of tyrosine kinase receptors. Using different myristoylated hSos1 full-length and carboxyl-terminal truncated mutants, we show that Grb2 function accounts not only for recruitment of hSos1 to the plasma membrane but also for modulation of hSos1 activity. Our results demonstrate that the first two canonical Grb2 binding sites, inside the carboxyl-terminal region of hSos1, are responsible for this regulation. Following different approaches, such as displacement of Grb2 from the hSos1-Grb2 complex or depletion of Grb2 levels by small interfering RNA, we found that the full-length Grb2 proteins mediate negative regulation of the intrinsic Ras guanine-nucleotide exchange activity of hSos1.


Subject(s)
Down-Regulation/genetics , GRB2 Adaptor Protein/metabolism , SOS1 Protein/metabolism , Animals , COS Cells , Cells, Cultured , Chlorocebus aethiops , GRB2 Adaptor Protein/deficiency , HeLa Cells , Humans , Mice , Mutant Proteins/metabolism , NIH 3T3 Cells , SOS1 Protein/chemistry
12.
Carcinogenesis ; 27(2): 328-36, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16113051

ABSTRACT

The effect of prostaglandins on the development of papillomas has been investigated in mice receiving prostaglandins E2 (PGE2) or the cyclopentenone 15-deoxy-delta(12,14)-PGJ2 (15dPGJ2) topically, using the 7,12-dimethylbenz[a]anthracene (DMBA)-induced tetradecanoylphorbol acetate (TPA)-promoted model of skin carcinogenesis. The presence of 15dPGJ2 during DMBA and TPA treatment inhibited apoptosis and increased the rate, number, size and vascularization of the papillomas, some of them progressing into carcinomas. Moreover, skin sections from mice treated for one week with DMBA and 15dPGJ2 showed a much reduced rate of apoptotic cells, and an enhanced expression of vascular epithelial growth factor when compared with animals receiving DMBA, with or without PGE2. The analysis of molecular events in the MCA3D keratinocyte cell line showed that 15dPGJ2 activated Ras and improved cell viability by inhibiting DMBA-dependent apoptosis. In addition to this, cell adhesion was impaired in MCA3D keratinocytes co-treated with 15dPGJ2 and DMBA, at the same time when the expression of cyclooxygenase-2 (COX-2) was observed under these conditions. These effects mediated by 15dPGJ2 might contribute to understand the role of COX-2 metabolites in carcinogenesis, leading to an increase of cell viability after mutagenic injury and therefore in the progression of tumors.


Subject(s)
Immunologic Factors/pharmacology , Papilloma/chemically induced , Papilloma/physiopathology , Prostaglandin D2/analogs & derivatives , Skin Neoplasms/chemically induced , 9,10-Dimethyl-1,2-benzanthracene/administration & dosage , Administration, Topical , Animals , Apoptosis , Carcinogens/administration & dosage , Cell Transformation, Neoplastic , Cyclooxygenase 2/metabolism , Disease Progression , Drug Interactions , Female , Keratinocytes/drug effects , Mice , Prostaglandin D2/pharmacology , Skin Neoplasms/physiopathology
13.
J Biol Chem ; 279(32): 33480-91, 2004 Aug 06.
Article in English | MEDLINE | ID: mdl-15181015

ABSTRACT

Ras proteins (H-, N-, and K-Ras) operate as molecular switches in signal transduction cascades controlling cell proliferation, differentiation, or apoptosis. The interaction of Ras with its effectors is mediated by the effector-binding loop, but different data about Ras location to plasma membrane subdomains and new roles for some docking/scaffold proteins point to signaling specificities of the different Ras proteins. To investigate the molecular mechanisms for these specificities, we compared an effector loop mutation (P34G) of three Ras isoforms (H-, N-, and K-Ras4B) for their biological and biochemical properties. Although this mutation diminished the capacity of Ras proteins to activate the Raf/ERK and the phosphatidylinositol 3-kinase/AKT pathways, the H-Ras V12G34 mutant retained the ability to cause morphological transformation of NIH 3T3 fibroblasts, whereas both the N-Ras V12G34 and the K-Ras4B V12G34 mutants were defective in this biological activity. On the other hand, although both the N-Ras V12G34 and the K-Ras4B V12G34 mutants failed to promote activation of the Ral-GDS/Ral A/PLD and the Ras/Rac pathways, the H-Ras V12G34 mutant retained the ability to activate these signaling pathways. Interestingly, the P34G mutation reduced specifically the N-Ras and K-Ras4B in vitro binding affinity to Ral-GDS, but not in the case of H-Ras. Thus, independently of Ras location to membrane subdomains, there are marked differences among Ras proteins in the sensitivity to an identical mutation (P34G) affecting the highly conserved effector-binding loop.


Subject(s)
Genes, ras/genetics , Mutation , ras Proteins/genetics , Animals , Binding Sites/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Enzyme Activation/drug effects , Gene Expression , Humans , Mice , Mitogen-Activated Protein Kinase 1 , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , NIH 3T3 Cells , Phosphatidylinositol 3-Kinases/metabolism , Phospholipase D/metabolism , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-raf/metabolism , Signal Transduction , Structure-Activity Relationship , Transcription Factors/genetics , Transcription Factors/physiology , Transfection , ets-Domain Protein Elk-1 , rac1 GTP-Binding Protein/metabolism , ral GTP-Binding Proteins/metabolism , ral Guanine Nucleotide Exchange Factor/metabolism , ras Proteins/chemistry , ras Proteins/pharmacology
14.
J Biol Chem ; 277(46): 44171-9, 2002 Nov 15.
Article in English | MEDLINE | ID: mdl-12223473

ABSTRACT

The protein hSos1 is a Ras guanine nucleotide exchange factor. In the present study, we investigated the function of the amino-terminal region of the hSos1 protein, corresponding to the first 600 residues, which includes the Dbl and pleckstrin homology (DH and PH) domains. We demonstrated, using a series of truncated mutants, that this region is absolutely necessary for hSos1 activity. Our results suggest that the first 200 residues (upstream of DH domain), which we called the HF motif on the basis of their homology with histone H2A, may exert negative control over the functional activity of the whole hSos1 protein. In vitro binding analysis showed that the HF motif is able to interact specifically with the PH domain of hSos1. The amino-terminal region of hSos1 may be associated in vivo with an expressed HF motif. These findings document the existence of the HF motif located upstream of the DH domain in the hSos1 protein. This motif may be responsible for the negative control of hSos1, probably by intramolecular binding with the PH domain.


Subject(s)
SOS1 Protein/chemistry , SOS1 Protein/physiology , 3T3 Cells , Amino Acid Motifs , Animals , COS Cells , Cell Line , Gene Deletion , Genes, Reporter , Humans , MAP Kinase Signaling System , Mice , Models, Molecular , Peptides , Plasmids/metabolism , Protein Binding , Protein Structure, Tertiary , Recombinant Fusion Proteins/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...