Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Biochem J ; 449(2): 389-400, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23078367

ABSTRACT

Hypoxia in the microenvironment of many solid tumours is an important determinant of malignant progression. The ISR (integrated stress response) protects cells from the ER (endoplasmic reticulum) stress caused by severe hypoxia. Likewise, autophagy is a mechanism by which cancer cells can evade hypoxic cell death. In the present paper we report that the autophagy-initiating kinase ULK1 (UNC51-like kinase 1) is a direct transcriptional target of ATF4 (activating transcription factor 4), which drives the expression of ULK1 mRNA and protein in severe hypoxia and ER stress. We demonstrate that ULK1 is required for autophagy in severe hypoxia and that ablation of ULK1 causes caspase-3/7-independent cell death. Furthermore, we report that ULK1 expression is associated with a poor prognosis in breast cancer. Collectively, the findings of the present study identify transcriptional up-regulation of ULK1 as a novel arm of the ISR, and suggest ULK1 as a potentially effective target for cancer therapy.


Subject(s)
Activating Transcription Factor 4/genetics , Intracellular Signaling Peptides and Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Transcriptional Activation , Up-Regulation , Activating Transcription Factor 4/metabolism , Animals , Autophagy/genetics , Autophagy-Related Protein-1 Homolog , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Hypoxia , Cell Line, Tumor , Cell Survival/genetics , Endoplasmic Reticulum Stress/genetics , Female , Gene Expression Regulation, Neoplastic , HCT116 Cells , HT29 Cells , Humans , Intracellular Signaling Peptides and Proteins/metabolism , MCF-7 Cells , Mice , Multivariate Analysis , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Prognosis , Protein Serine-Threonine Kinases/metabolism , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Survival Analysis
2.
Biosci Rep ; 32(4): 413-22, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22668349

ABSTRACT

Inhibition of the PI3K (phosphoinositide 3-kinase)/Akt/mTORC1 (mammalian target of rapamycin complex 1) and Ras/MEK [MAPK (mitogen-activated protein kinase)/ERK (extracellular-signal-regulated kinase) kinase]/ERK pathways for cancer therapy has been pursued for over a decade with limited success. Emerging data have indicated that only discrete subsets of cancer patients have favourable responses to these inhibitors. This is due to genetic mutations that confer drug insensitivity and compensatory mechanisms. Therefore understanding of the feedback mechanisms that occur with respect to specific genetic mutations may aid identification of novel biomarkers that predict patient response. In the present paper, we show that feedback between the PI3K/Akt/mTORC1 and Ras/MEK/ERK pathways is cell-line-specific and highly dependent on the activating mutation of K-Ras or overexpression c-Met. We found that cell lines exhibited differential signalling and apoptotic responses to PD184352, a specific MEK inhibitor, and PI103, a second-generation class I PI3K inhibitor. We reveal that feedback from the PI3K/Akt/mTORC1 to the Ras/MEK/ERK pathway is present in cancer cells harbouring either K-Ras activating mutations or amplification of c-Met but not the wild-type counterparts. Moreover, we demonstrate that inhibition of protein phosphatase activity by OA (okadaic acid) restored PI103-mediated feedback in wild-type cells. Together, our results demonstrate a novel mechanism for feedback between the PI3K/Akt/mTORC1 and the Ras/MEK/ERK pathways that only occurs in K-Ras mutant and c-Met amplified cells but not the isogenic wild-type cells through a mechanism that may involve inhibition of a specific endogenous phosphatase(s) activity. We conclude that monitoring K-Ras and c-Met status are important biomarkers for determining the efficacy of PI103 and other PI3K/Akt inhibitors in cancer therapy.


Subject(s)
MAP Kinase Signaling System , Mutation , Oncogenes , Phosphatidylinositol 3-Kinases/metabolism , Apoptosis/drug effects , Benzamides/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Drug Synergism , Feedback, Physiological , Furans/pharmacology , Humans , Mechanistic Target of Rapamycin Complex 1 , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Multiprotein Complexes , Okadaic Acid/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphoprotein Phosphatases/metabolism , Phosphorylation , Protein Processing, Post-Translational , Proteins/antagonists & inhibitors , Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , Proto-Oncogene Proteins p21(ras) , Pyridines/pharmacology , Pyrimidines/pharmacology , Sirolimus/pharmacology , TOR Serine-Threonine Kinases , Up-Regulation , ras Proteins/metabolism
3.
Biochemistry ; 51(10): 2100-12, 2012 Mar 13.
Article in English | MEDLINE | ID: mdl-22352903

ABSTRACT

Evidence that elongation factor 2 kinase (eEF-2K) has potential as a target for anticancer therapy and possibly for the treatment of depression is emerging. Here the steady-state kinetic mechanism of eEF-2K is presented using a peptide substrate and is shown to conform to an ordered sequential mechanism with ATP binding first. Substrate inhibition by the peptide was observed and revealed to be competitive with ATP, explaining the observed ordered mechanism. Several small molecules are reported to inhibit eEF-2K activity with the most notable being the histidine kinase inhibitor NH125, which has been used in a number of studies to characterize eEF-2K activity in cells. While NH125 was previously reported to inhibit eEF-2K in vitro with an IC(50) of 60 nM, its mechanism of action was not established. Using the same kinetic assay, the ability of an authentic sample of NH125 to inhibit eEF-2K was assessed over a range of substrate and inhibitor concentrations. A typical dose-response curve for the inhibition of eEF-2K by NH125 is best fit to an IC(50) of 18 ± 0.25 µM and a Hill coefficient of 3.7 ± 0.14, suggesting that NH125 is a weak inhibitor of eEF-2K under the experimental conditions of a standard in vitro kinase assay. To test the possibility that NH125 is a potent inhibitor of eEF2 phosphorylation, we assessed its ability to inhibit the phosphorylation of eEF2. Under standard kinase assay conditions, NH125 exhibits a similar weak ability to inhibit the phosphorylation of eEF2 by eEF-2K. Notably, the activity of NH125 is severely abrogated by the addition of 0.1% Triton to the kinase assay through a process that can be reversed upon dilution. These studies suggest that NH125 is a nonspecific colloidal aggregator in vitro, a notion further supported by the observation that NH125 inhibits other protein kinases, such as ERK2 and TRPM7 in a manner similar to that of eEF-2K. As NH125 is reported to inhibit eEF-2K in a cellular environment, its ability to inhibit eEF2 phosphorylation was assessed in MDA-MB-231 breast cancer, A549 lung cancer, and HEK-293T cell lines using a Western blot approach. No sign of a decrease in the level of eEF2 phosphorylation was observed up to 12 h following addition of NH125 to the media. Furthermore, contrary to the previously reported literatures, NH125 induced the phosphorylation of eEF-2.


Subject(s)
Elongation Factor 2 Kinase/antagonists & inhibitors , Imidazoles/pharmacology , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Artifacts , Cell Line , Dose-Response Relationship, Drug , Elongation Factor 2 Kinase/genetics , Elongation Factor 2 Kinase/metabolism , HEK293 Cells , Humans , Imidazoles/administration & dosage , In Vitro Techniques , Kinetics , Molecular Sequence Data , Peptides/chemistry , Peptides/metabolism , Phosphorylation , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacology , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Substrate Specificity
4.
Biochemistry ; 51(11): 2232-45, 2012 Mar 20.
Article in English | MEDLINE | ID: mdl-22329831

ABSTRACT

Eukaryotic elongation factor 2 kinase (eEF-2K) is an atypical protein kinase regulated by Ca(2+) and calmodulin (CaM). Its only known substrate is eukaryotic elongation factor 2 (eEF-2), whose phosphorylation by eEF-2K impedes global protein synthesis. To date, the mechanism of eEF-2K autophosphorylation has not been fully elucidated. To investigate the mechanism of autophosphorylation, human eEF-2K was coexpressed with λ-phosphatase and purified from bacteria in a three-step protocol using a CaM affinity column. Purified eEF-2K was induced to autophosphorylate by incubation with Ca(2+)/CaM in the presence of MgATP. Analyzing tryptic or chymotryptic peptides by mass spectrometry monitored the autophosphorylation over 0-180 min. The following five major autophosphorylation sites were identified: Thr-348, Thr-353, Ser-445, Ser-474, and Ser-500. In the presence of Ca(2+)/CaM, robust phosphorylation of Thr-348 occurs within seconds of addition of MgATP. Mutagenesis studies suggest that phosphorylation of Thr-348 is required for substrate (eEF-2 or a peptide substrate) phosphorylation, but not self-phosphorylation. Phosphorylation of Ser-500 lags behind the phosphorylation of Thr-348 and is associated with the Ca(2+)-independent activity of eEF-2K. Mutation of Ser-500 to Asp, but not Ala, renders eEF-2K Ca(2+)-independent. Surprisingly, this Ca(2+)-independent activity requires the presence of CaM.


Subject(s)
Calcium/metabolism , Calmodulin/metabolism , Elongation Factor 2 Kinase/metabolism , Serine/genetics , Threonine/genetics , Amino Acid Sequence , Binding Sites , Elongation Factor 2 Kinase/genetics , Humans , Mass Spectrometry , Molecular Sequence Data , Phosphorylation , Threonine/metabolism
5.
Biochem Biophys Res Commun ; 411(2): 227-34, 2011 Jul 29.
Article in English | MEDLINE | ID: mdl-21708134

ABSTRACT

The tumour metastasis suppressor, N-myc Downstream Regulated Gene (NDRG) 1, is a by the protein kinases SGK1 and GSK3ß, but the relevance of its phosphorylation remains unclear. Analysis of HCT116 cells, either proficient or deficient for p53 revealed NDRG1 protein expression and phosphorylation by SGK1 was increased basally in p53-deficient cells. Treatment with the cell cycle inhibitors, aphidicolin or nocodazole also revealed increased NDRG1 phosphorylation in p53-deficient cells. Finally, phosphorylated NDRG1 was found to co-localise with γ-tubulin on centromeres and also to the cleavage furrow during cytokinesis. Taken together, this work demonstrates that NDRG1 phosphorylation, by the protein kinase SGK1, is temporally and spatially controlled during the cell cycle, suggesting a role for NDRG1 in successful mitosis.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Cycle , Immediate-Early Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Cell Cycle Proteins/genetics , Cell Line, Tumor , Centromere/metabolism , Down-Regulation , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mitosis , Phosphorylation , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
6.
Protein Expr Purif ; 79(2): 237-44, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21605678

ABSTRACT

The eukaryotic elongation factor 2 kinase (eEF-2K) modulates the rate of protein synthesis by impeding the elongation phase of translation by inactivating the eukaryotic elongation factor 2 (eEF-2) via phosphorylation. eEF-2K is known to be activated by calcium and calmodulin, whereas the mTOR and MAPK pathways are suggested to negatively regulate kinase activity. Despite its pivotal role in translation regulation and potential role in tumor survival, the structure, function, and regulation of eEF-2K have not been described in detail. This deficiency may result from the difficulty of obtaining the recombinant kinase in a form suitable for biochemical analysis. Here we report the purification and characterization of recombinant human eEF-2K expressed in the Escherichia coli strain Rosetta-gami 2(DE3). Successive chromatography steps utilizing Ni-NTA affinity, anion-exchange, and gel filtration columns accomplished purification. Cleavage of the thioredoxin-His(6)-tag from the N-terminus of the expressed kinase with TEV protease yielded 9 mg of recombinant (G-D-I)-eEF-2K per liter of culture. Light scattering shows that eEF-2K is a monomer of ∼85 kDa. In vitro kinetic analysis confirmed that recombinant human eEF-2K is able to phosphorylate wheat germ eEF-2 with kinetic parameters comparable to the mammalian enzyme.


Subject(s)
Cloning, Molecular/methods , Elongation Factor 2 Kinase/metabolism , Peptide Elongation Factor 2/metabolism , Plasmids/genetics , Protein Biosynthesis/genetics , Recombinant Proteins/metabolism , Amino Acid Sequence , Calcium/metabolism , Calmodulin/genetics , Calmodulin/metabolism , Chromatography, Affinity , Chromatography, Gel , Chromatography, Ion Exchange , Elongation Factor 2 Kinase/genetics , Elongation Factor 2 Kinase/isolation & purification , Endopeptidases/metabolism , Escherichia coli , Histidine/metabolism , Humans , Kinetics , Molecular Sequence Data , Oligopeptides/metabolism , Phosphorylation , Plasmids/chemistry , Plasmids/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Thioredoxins/metabolism , Transformation, Bacterial
7.
Biochemistry ; 50(18): 3660-72, 2011 May 10.
Article in English | MEDLINE | ID: mdl-21449613

ABSTRACT

The mitogen-activated protein (MAP) kinase ERK2 contains recruitment sites that engage canonical and noncanonical motifs found in a variety of upstream kinases, regulating phosphatases and downstream targets. Interactions involving two of these sites, the D-recruitment site (DRS) and the F-recruitment site (FRS), have been shown to play a key role in signal transduction by ERK/MAP kinases. The dynamic nature of these recruitment events makes NMR uniquely suited to provide significant insight into these interactions. While NMR studies of kinases in general have been greatly hindered by their large size and complex dynamic behavior leading to the suboptimal performance of standard methodologies, we have overcome these difficulties for inactive full-length ERK2 and obtained an acceptable level of backbone resonance assignments. This allowed a detailed investigation of the structural perturbations that accompany interactions involving both canonical and noncanonical recruitment events. No crystallographic information exists for the latter. We found that the chemical shift perturbations in inactive ERK2, indicative of structural changes in the presence of canonical and noncanonical motifs, are not restricted to the recruitment sites but also involve the linker that connects the N- and C-lobes and, in most cases, a gatekeeper residue that is thought to exert allosteric control over catalytic activity. We also found that, even though the canonical motifs interact with the DRS utilizing both charge-charge and hydrophobic interactions, the noncanonical interactions primarily involve the latter. These results demonstrate the feasibility of solution NMR techniques for a comprehensive analysis of docking interactions in a full-length ERK/MAP kinase.


Subject(s)
Magnetic Resonance Spectroscopy/methods , Mitogen-Activated Protein Kinase 1/chemistry , Allosteric Site , Amino Acid Motifs , Animals , Binding Sites , Crystallography, X-Ray/methods , Escherichia coli/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , MAP Kinase Signaling System , Models, Statistical , Molecular Conformation , Peptides/chemistry , Protein Binding , Protein Structure, Tertiary , Rats
8.
Biochemistry ; 50(21): 4568-78, 2011 May 31.
Article in English | MEDLINE | ID: mdl-21506533

ABSTRACT

The extracellular signal-regulated protein kinase, ERK2, fully activated by phosphorylation and without a His(6) tag, shows little tendency to dimerize with or without either calcium or magnesium ions when analyzed by light scattering or analytical ultracentrifugation. Light scattering shows that ~90% of ERK2 is monomeric. Sedimentation equilibrium data (obtained at 4.8-11.2 µM ERK2) with or without magnesium (10 mM) are well described by an ideal one-component model with a fitted molar mass of 40180 ± 240 Da (without Mg(2+) ions) or 41290 ± 330 Da (with Mg(2+) ions). These values, close to the sequence-derived mass of 41711 Da, indicate that no significant dimerization of ERK2 occurs in solution. Analysis of sedimentation velocity data for a 15 µM solution of ERK2 with an enhanced van Holde-Weischet method determined the sedimentation coefficient (s) to be ~3.22 S for activated ERK2 with or without 10 mM MgCl(2). The frictional coefficient ratio (f/f(0)) of 1.28 calculated from the sedimentation velocity and equilibrium data is close to that expected for an ~42 kDa globular protein. The translational diffusion coefficient of ~8.3 × 10(-7) cm(2) s(-1) calculated from the experimentally determined molar mass and sedimentation coefficient agrees with the value determined by dynamic light scattering in the absence and presence of calcium or magnesium ions and a value determined by NMR spectrometry. ERK2 has been proposed to homodimerize and bind only to cytoplasmic but not nuclear proteins [Casar, B., et al. (2008) Mol. Cell 31, 708-721]. Our light scattering data show, however, that ERK2 forms a strong 1:1 complex of ~57 kDa with the cytoplasmic scaffold protein PEA-15. Thus, ERK2 binds PEA-15 as a monomer. Our data provide strong evidence that ERK2 is monomeric under physiological conditions. Analysis of the same ERK2 construct with the nonphysiological His(6) tag shows substantial dimerization under the same ionic conditions.


Subject(s)
Cations, Divalent/metabolism , Cytoplasm/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Phosphoproteins/metabolism , Apoptosis Regulatory Proteins , Enzyme Activation , Humans , Light , Scattering, Radiation , Ultracentrifugation
9.
Biochemistry ; 46(32): 9174-86, 2007 Aug 14.
Article in English | MEDLINE | ID: mdl-17658891

ABSTRACT

Many substrates of ERK2 contain a D-site, a sequence recognized by ERK2 that is used to promote catalysis. Despite lacking a canonical D-site, the substrate Ets-1 is displaced from ERK2 by peptides containing one. This suggests that Ets-1 may contain a novel or cryptic D-site. To investigate this possibility a protein footprinting strategy was developed to elucidate ERK2-ligand interactions. Using this approach, single cysteine reporters were placed in the D-recruitment site (DRS) of ERK2 and the resulting ERK2 proteins subjected to alkylation by iodoacetamide. The ability of residues 1-138 of Ets-1 to protect the cysteines from alkylation was determined. The pattern of protection observed is consistent with Ets-1 occupying a hydrophobic binding site within the DRS of ERK2. Significantly, a peptide derived from the D-site of Elk-1, which is known to bind the DRS, exhibits a similar pattern of cysteine protection. This analysis expands the repertoire of the DRS on ERK2 and suggests that other targeting sequences remain to be identified. Furthermore, cysteine-footprinting is presented as a useful way to interrogate protein-ligand interactions at the resolution of a single amino acid.


Subject(s)
Cysteine/metabolism , Mitogen-Activated Protein Kinase 1/chemistry , Mitogen-Activated Protein Kinase 1/metabolism , Protein Footprinting , Proto-Oncogene Protein c-ets-1/chemistry , Proto-Oncogene Protein c-ets-1/metabolism , Alkylation , Amino Acid Sequence , Animals , Binding, Competitive/genetics , Catalysis , Mice , Mitogen-Activated Protein Kinase 1/genetics , Molecular Sequence Data , Phosphorylation , Protein Binding/genetics , Protein Interaction Mapping , Protein Transport/genetics , Rats , Substrate Specificity/genetics
10.
Biochemistry ; 46(32): 9187-98, 2007 Aug 14.
Article in English | MEDLINE | ID: mdl-17658892

ABSTRACT

PEA-15 is a small anti-apoptotic protein that is enriched in astrocytes, but expressed in a broad range of tissues. It sequesters the protein kinases ERK1 and 2 in the cytoplasm, thereby limiting their proximity to nuclear substrates. Using a fluorescence anisotropy approach, PEA-15 is shown to be a high-affinity ligand for both ERK1 and 2, exhibiting a dissociation constant in the range of Kd = 0.2-0.4 microM, regardless of their activation states. Neither the phosphorylation of PEA-15 (phospho Ser-104 and/or phospho Ser-116) nor the phosphorylation of ERK1/2 (by MKK1) significantly affects the stability of the ERK/PEA-15 interaction, and therefore it does not directly regulate the release of ERK2 to the nucleus. The extreme C-terminus of PEA-15 was previously shown by mutagenesis to be important for ERK2 binding; however, the site of binding was not established. Here it is demonstrated that the D-recruitment site (DRS) of ERK2 binds PEA-15, probably at the C-terminus, and renders PEA-15 an inhibitor of ERK2 docking interactions. Using fluorescence anisotropy competition assays it is shown that PEA-15 competes for binding to ERK1/2 with a peptide derived from the D-site of Elk-1, which binds the DRS of ERK1/2. Using modified ERK2 proteins containing single cysteine residues, PEA-15 was shown to protect single cysteines situated within the DRS from alkylation. The pattern and magnitude of protection were very similar to those induced by the binding of the peptide derived from the D-site of Elk-1. These and published data support the notion that PEA-15 binds two sites on ERK1/2 in a bidentate manner: the DRS and a site that includes the MAP kinase insert. Previous reports have suggested that PEA-15 is not an inhibitor of ERK2; however, it is shown here to potently inhibit the ability of ERK2 to phosphorylate two transcription factors, Elk-1 and Ets-1, which contain docking sites for the DRS of ERK2. Therefore, in addition to sequestering ERK1/2 in the cytoplasm, PEA-15 has the potential to modulate the activity of ERK2 in cells by competing directly with proteins that contain D-sites.


Subject(s)
Apoptosis/physiology , Intracellular Signaling Peptides and Proteins/physiology , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Phosphoproteins/physiology , Protein Kinase Inhibitors/metabolism , Amino Acid Sequence , Apoptosis/genetics , Apoptosis Regulatory Proteins , Binding Sites/physiology , Binding, Competitive/physiology , Catalysis , Cytoplasm/chemistry , Cytoplasm/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinase 3/physiology , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Phosphoproteins/biosynthesis , Phosphoproteins/metabolism , Phosphorylation , Protein Binding , Protein Kinase Inhibitors/chemistry , Protein Transport/physiology , Signal Transduction/physiology
11.
Biochemistry ; 45(46): 13719-33, 2006 Nov 21.
Article in English | MEDLINE | ID: mdl-17105191

ABSTRACT

ERK2 is a proline-directed protein kinase that displays a high specificity for a single threonine (Thr-38) on the substrate Ets-1, which lies within the consensus sequence 36phi-chi-Thr-Pro39 (where phi is typically a small hydrophobic residue and chi appears to be unrestricted). Thr-38 lies in a long flexible N-terminal tail (residues 1-52), which also contains a second potential phosphorylation site, Ser-26. How Ets-1 binds ERK2 to promote the phosphorylation of Thr-38 while simultaneously discriminating against the phosphorylation of Ser-26 is unclear. To delineate the details of the molecular recognition of Ets-1 by ERK2, the binding of various mutants and truncations of Ets-1 were analyzed by fluorescence anisotropy. The data that were obtained support the notion that the N-terminal tail contains a previously unrecognized docking site that promotes the phosphorylation of Thr-38. This new docking site helps assemble the complex of Ets-1 and ERK2 and makes a similar contribution to the stabilization of the complex as does the pointed domain of Ets-1. The in vitro activation of ERK2 by MKK1 induces a large conformational transition of the activation segment (DFG-APE), but neither induces self-association of ERK2 nor destabilizes the stability of the ERK2.Ets-1 complex. This latter observation suggests that interactions intrinsic to the active site are not important for complex assembly, a notion further supported by the observation that the substitution of a number of different amino acids for Pro-39 does not destabilize the complex. Mutagenesis of ERK2 within loop 13 suggests that Ets-1 binds the substrate-binding groove. These data suggest that ERK2 uses two weak docking interactions to specifically assemble the complex, perhaps in doing so denying Ser-26 access to the active site. Displacement of residues 1-138 of Ets-1 (EtsDelta138) from ERK2 by the peptide N-QKGKPRDLELPLSPSL-C, derived from Elk-1, suggests that Ets-1 engages the D-recruitment site (beta7-beta8 reverse turn and the alphaD-alphaE helix) of ERK2. Displacement of EtsDelta138 from ERK2 by the peptide N-AKLSFQFPS-C derived from Elk-1 shows that EtsDelta138 communicates with the F-recruitment site of ERK2 also.


Subject(s)
Mitogen-Activated Protein Kinase 1/metabolism , Proto-Oncogene Protein c-ets-1/metabolism , Signal Transduction , Amino Acid Sequence , Fluorescence Polarization , Humans , Light , Models, Molecular , Molecular Sequence Data , Phosphorylation , Proto-Oncogene Protein c-ets-1/chemistry , Scattering, Radiation , Sequence Homology, Amino Acid
12.
Biochem Biophys Res Commun ; 312(3): 623-8, 2003 Dec 19.
Article in English | MEDLINE | ID: mdl-14680810

ABSTRACT

Two ATP-competitive inhibitors-4,5,6,7-tetrabromo-benzotriazole (TBBt) and 4,5,6,7-tetrabromo-benzimidazole (TBBz) have been shown to decrease activity of CK2 holoenzyme. Surprisingly it occurs that TBBz contrary to TBBt does not inhibit free catalytic subunit CK2 [Formula: see text]. Both inhibitors are virtually inactive against RAP protein kinase. The above-mentioned protein kinases phosphorylate in vitro a set of acidic ribosomal P-proteins of the 60S ribosomal subunit. Such a modification is one of the mechanisms regulating translational activity of ribosomes in vivo. Application of these two very selective inhibitors allows us to define the role of free catalytic [Formula: see text] subunit of CK2 in phosphorylation of ribosomal proteins. It occurs that CK2 [Formula: see text] but not CK2 holoenzyme is responsible for phosphorylation of P-proteins in vivo. Moreover, elimination of both forms of protein kinase CK2 (hCK2 and CK2 [Formula: see text] ) activity in living cells led to dramatic loss of the translational activity of the ribosome.


Subject(s)
Benzimidazoles/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Protozoan Proteins , Ribosomal Proteins/metabolism , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/enzymology , Triazoles/pharmacology , Benzimidazoles/chemistry , Benzimidazoles/toxicity , Binding, Competitive , Casein Kinase II , Cell Division/drug effects , Dose-Response Relationship, Drug , Enzyme Activation , Protein Biosynthesis/drug effects , Protein Biosynthesis/physiology , Protein Serine-Threonine Kinases/chemistry , Ribosomal Proteins/chemistry , Saccharomyces cerevisiae/cytology , Structure-Activity Relationship , Triazoles/chemistry , Triazoles/toxicity
13.
Biochem Biophys Res Commun ; 307(1): 31-40, 2003 Jul 18.
Article in English | MEDLINE | ID: mdl-12849977

ABSTRACT

The 60S ribosomes from Saccharomyces cerevisiae contain a set of acidic P-proteins playing an important role in the ribosome function. Reversible phosphorylation of those proteins is a mechanism regulating translational activity of ribosomes. The key role in regulation of this process is played by specific, second messenger-independent protein kinases. The PK60S kinase was one of the enzymes phosphorylating P-proteins. The enzyme has been purified from yeast and characterised. Pure enzyme has properties similar to those reported for casein kinase type 2. Peptide mass fingerprinting (PMF) has identified the PK60S as a catalytic alpha(') subunit of casein kinase type 2 (CK2alpha(')). Protein kinase activity is inhibited by SOD1 and by highly specific CK2 inhibitor-4,5,6,7-tetrabromo-benzotriazole (TBBt). The possible mechanism of regulation of CK2alpha(') activity in stress conditions, by superoxide dismutase in regulation of 80S-ribosome activity, is discussed.


Subject(s)
Catalytic Domain , Fungal Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae/enzymology , Superoxide Dismutase/metabolism , Casein Kinase II , DNA-Binding Proteins/genetics , DNA-Binding Proteins/isolation & purification , DNA-Binding Proteins/metabolism , Fungal Proteins/genetics , Multienzyme Complexes , Peptides/genetics , Peptides/metabolism , Phosphorylation , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/isolation & purification , Ribosomes/metabolism , Saccharomyces cerevisiae/metabolism , Superoxide Dismutase-1
SELECTION OF CITATIONS
SEARCH DETAIL
...