Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 99
Filter
1.
Phys Rev Lett ; 105(2): 022002, 2010 Jul 09.
Article in English | MEDLINE | ID: mdl-20867699

ABSTRACT

Charged-hadron transverse-momentum and pseudorapidity distributions in proton-proton collisions at square root of s = 7 TeV are measured with the inner tracking system of the CMS detector at the LHC. The charged-hadron yield is obtained by counting the number of reconstructed hits, hit pairs, and fully reconstructed charged-particle tracks. The combination of the three methods gives a charged-particle multiplicity per unit of pseudorapidity dN(ch)/dη|(|η|<0.5) = 5.78 ± 0.01(stat) ± 0.23(syst) for non-single-diffractive events, higher than predicted by commonly used models. The relative increase in charged-particle multiplicity from square root of s = 0.9 to 7 TeV is [66.1 ± 1.0(stat) ± 4.2(syst)]%. The mean transverse momentum is measured to be 0.545 ± 0.005(stat) ± 0.015(syst) GeV/c. The results are compared with similar measurements at lower energies.

2.
Phys Rev Lett ; 105(3): 032001, 2010 Jul 16.
Article in English | MEDLINE | ID: mdl-20867758

ABSTRACT

Bose-Einstein correlations have been measured using samples of proton-proton collisions at 0.9 and 2.36 TeV center-of-mass energies, recorded by the CMS experiment at the CERN Large Hadron Collider. The signal is observed in the form of an enhancement of pairs of same-sign charged particles with small relative four-momentum. The size of the correlated particle emission region is seen to increase significantly with the particle multiplicity of the event.

3.
Phys Rev Lett ; 105(21): 211801, 2010 Nov 19.
Article in English | MEDLINE | ID: mdl-21231289

ABSTRACT

A search for narrow resonances in the dijet mass spectrum is performed using data corresponding to an integrated luminosity of 2.9 pb⁻¹ collected by the CMS experiment at the Large Hadron Collider. Upper limits at the 95% confidence level are presented on the product of the resonance cross section, branching fraction into dijets, and acceptance, separately for decays into quark-quark, quark-gluon, or gluon-gluon pairs. The data exclude new particles predicted in the following models at the 95% confidence level: string resonances, with mass less than 2.50 TeV, excited quarks, with mass less than 1.58 TeV, and axigluons, colorons, and E6 diquarks, in specific mass intervals. This extends previously published limits on these models.

4.
Phys Rev Lett ; 105(26): 262001, 2010 Dec 31.
Article in English | MEDLINE | ID: mdl-21231646

ABSTRACT

A search for quark compositeness in the form of quark contact interactions, based on hadronic jet pairs (dijets) produced in proton-proton collisions at √s=7 TeV, is described. The data sample of the study corresponds to an integrated luminosity of 2.9 pb(-1) collected with the CMS detector at the LHC. The dijet centrality ratio, which quantifies the angular distribution of the dijets, is measured as a function of the invariant mass of the dijet system and is found to agree with the predictions of the standard model. A statistical analysis of the data provides a lower limit on the energy scale of quark contact interactions. The sensitivity of the analysis is such that the expected limit is 2.9 TeV; because the observed value of the centrality ratio at high invariant mass is below the expectation, the observed limit is 4.0 TeV at the 95% confidence level.

5.
Mol Biol (Mosk) ; 37(4): 688-95, 2003.
Article in Russian | MEDLINE | ID: mdl-12942642

ABSTRACT

Androgen-dependent human prostate adenocarcinoma cell line LNCaP was used to study the effect of androgen deprivation on the cell response to TNF-related cytokines. Several signaling pathways were implicated in cell survival in the absence of androgens. In androgen-deprived LNCaP cells, TNF-alpha and TRAIL stimulated the cell growth and activated the mitogenic and antiapoptotic signaling pathways involving NF-kappa B, STAT3, PI3K, and beta-catenin. The results suggested a role of cytokines in the survival of prostate adenocarcinoma cells deprived of androgens in vitro.


Subject(s)
Androgens/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Signal Transduction , Acetylcysteine/pharmacology , Androstadienes/pharmacology , Apoptosis Regulatory Proteins , Cell Division/physiology , Cell Survival/physiology , Cytokines/metabolism , Cytokines/pharmacology , Cytoplasm/drug effects , Cytoplasm/metabolism , Cytoskeletal Proteins/drug effects , Cytoskeletal Proteins/metabolism , DNA-Binding Proteins/drug effects , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , Lymphoid Enhancer-Binding Factor 1 , Male , Membrane Glycoproteins/drug effects , Membrane Glycoproteins/metabolism , NF-kappa B/drug effects , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Prostatic Neoplasms/drug therapy , STAT3 Transcription Factor , TNF-Related Apoptosis-Inducing Ligand , Thymidine/metabolism , Trans-Activators/drug effects , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/drug effects , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Wortmannin , beta Catenin
6.
Cancer Chemother Pharmacol ; 45(6): 441-9, 2000.
Article in English | MEDLINE | ID: mdl-10854130

ABSTRACT

PURPOSE: We have previously found that microinjection of activated MEK (mitogen activated kinase kinase) and ERK (mitogen-activated protein; MAP kinase) fails to induce oocyte maturation, but that maturation, induced by oncogenic ras-p21 and insulin-activated cell ras-p21, is blocked by peptides from the ras-binding domain of raf. We also found that jun kinase (JNK), on the stress-activated protein (SAP) pathway, which is critical to the oncogenic ras-p21 signal transduction pathway, is a strong inducer of oocyte maturation. Our purpose in this study was to determine the role of the raf-MEK-MAP kinase pathway in oocyte maturation and how it interacts with JNK from the SAP pathway. METHODS: We microinjected raf dominant negative mutant mRNA (DN-raf) and the MEK-specific phosphatase, MKP-T4, either together with oncogenic p21 or c-raf mRNA, into oocytes or into oocytes incubated with insulin to determine the effects of these raf-MEK-MAP kinase pathway inhibitors. RESULTS: We found that oocyte maturation induced by both oncogenic and activated normal p21 is inhibited by both DN-raf and by MKP-T4. The latter more strongly blocks the oncogenic pathway. Also an mRNA encoding a constitutively activated MEK strongly induces oocyte maturation that is not inhibited by DN-raf or by MKP-T4. Surprisingly, we found that oocyte maturation induced by JNK is blocked both by DN-raf and MKP-T4. Furthermore, we discovered that c-raf induces oocyte maturation that is inhibited by glutathione-S-transferase (GST), which we have found to be a potent and selective inhibitor of JNK. CONCLUSION: We conclude that there is a strong reciprocal interaction between the SAP pathway involving JNK and the raf-MEK-MAP kinase pathway and that oncogenic ras-p21 can be preferentially inhibited by MEK inhibitors. The results imply that blockade of both MEK and JNK-oncogenic ras-p21 interactions may constitute selective synergistic combination chemotherapy against oncogenic ras-induced tumors.


Subject(s)
Mitogen-Activated Protein Kinases/metabolism , Oocytes/growth & development , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins p21(ras)/physiology , Signal Transduction , Animals , Female , JNK Mitogen-Activated Protein Kinases , Models, Biological , Proto-Oncogene Proteins c-raf/metabolism , Xenopus laevis
7.
Cancer Res ; 60(4): 896-900, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10706102

ABSTRACT

p53 phosphorylation and association with proteins is implicated in its stability and activity. We have compared the association of DNA-bound and overall pools of p53 with murine double minute 2 (Mdm2), c-Jun NH2-terminal kinase (JNK), p300/CBP, and p14ARF during cell cycle progression. Whereas DNA-bound p53 associates with JNK at G0-G1 and with Mdm2 and p300 during S and G2-M phases, the general pool of p53 was found in complex with JNK and Mdm2 almost throughout the cell cycle. Phosphorylation of p53 at serines 9, 15, and 20 is at the highest levels at G1 and at serines 37 and 392 during G2-M phase. Whereas a high dose of UV irradiation was required for phosphorylation of serines 15 and 392 between 8 and 24 h after treatment, a low dose caused immediate phosphorylation on serines 9, 20, and 372. These dynamic changes in the phosphorylation of p53 are expected to play a pivotal role in p53 association, stability, and function.


Subject(s)
Mitogen-Activated Protein Kinases/metabolism , Nuclear Proteins/metabolism , Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Trans-Activators/metabolism , Tumor Suppressor Protein p53/metabolism , Cell Cycle , Humans , JNK Mitogen-Activated Protein Kinases , Phosphorylation , Proto-Oncogene Proteins c-mdm2 , Tumor Suppressor Protein p14ARF , Ultraviolet Rays
8.
Ann Clin Lab Sci ; 30(1): 57-64, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10678584

ABSTRACT

We have identified the intracellular detoxification enzyme, glutathione-S-transferase (GST), as a potent inhibitor of the activation of jun by its kinase, jun-N-terminal kinase (JNK), in vitro. All three major isozymes (alpha, mu, and pi) bind to JNK-jun complexes and inhibit activation of jun by JNK. We now find that GST inhibits JNK-induced oocyte maturation in vivo and strongly inhibits oocyte maturation induced by oncogenic ras-p21 protein, but not by insulin-activated normal cellular p21 protein. These results correlate with the finding that oncogenic, but not insulin-activated normal, p21 induces high levels of activated JNK. GST also strongly blocks induction of oocyte maturation by protein kinase C (PKC) which is a critical downstream target of oncogenic but not normal ras-p21. Thus, we have established a new function for GST as a potent physiological inhibitor of the ras-JNK-jun pathway.


Subject(s)
Glutathione Transferase/pharmacology , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinase Kinases/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Signal Transduction/physiology , Animals , Dose-Response Relationship, Drug , Enzyme Activation/physiology , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , MAP Kinase Kinase 4 , Mitogens/metabolism , Oocytes/cytology , Oocytes/enzymology , Oocytes/growth & development , Phosphorylation , Phosphotyrosine/analysis , Protein Kinase C/metabolism , Signal Transduction/drug effects , Xenopus laevis
9.
J Mol Biol ; 295(4): 1009-21, 2000 Jan 28.
Article in English | MEDLINE | ID: mdl-10656807

ABSTRACT

Identification of Mdm2 and JNK as proteins that target degradation of wt p53 prompted us to examine their effect on mutant p53, which exhibits a prolonged half-life. Of five mutant p53 forms studied for association with the targeting molecules, two no longer bound to Mdm2 and JNK. Three mutant forms, which exhibit high expression levels, showed lower affinity for association with Mdm2 and JNK in concordance with greater affinity to p14(ARF), which is among the stabilizing p53 molecules. Monitoring mutant p53 stability in vitro confirmed that, while certain forms of mutant p53 are no longer affected by either JNK or Mdm2, others are targeted for degradation by JNK/Mdm2, albeit at lower efficiency when compared with wt p53. Expression of wt p53 in tumor cells revealed a short half-life, suggesting that the targeting molecules are functional. Forced expression of mutant p53 in p53 null cells confirmed pattern of association with JNK/Mdm2 and prolonged half-life, as found in the tumor cells. Over-expression of Mdm2 in either tumor (which do express endogenous functional Mdm2) or in p53 null cells decreased the stability of mutant p53 suggesting that, despite its expression, Mdm2/JNK are insufficient (amount/affinity) for targeting mutant p53 degradation. Based on both in vitro and in vivo analyses, we conclude that the prolonged half-life of mutant p53 depends on the nature of the mutation, which either alters association with targeting molecules, ratio between p53 and targeting/stabilizing molecules or targeting efficiency.


Subject(s)
Mitogen-Activated Protein Kinases/metabolism , Nuclear Proteins , Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/metabolism , Cell Membrane/metabolism , Fibroblasts , Genes, p53 , Half-Life , Humans , JNK Mitogen-Activated Protein Kinases , Kinetics , Protein Conformation , Proto-Oncogene Proteins c-mdm2 , Recombinant Proteins/metabolism , Transfection , Tumor Cells, Cultured , Tumor Suppressor Protein p14ARF , Tumor Suppressor Protein p53/genetics , Zinc Fingers
10.
Oncogene ; 18(45): 6104-11, 1999 Nov 01.
Article in English | MEDLINE | ID: mdl-10557101

ABSTRACT

Stress-activated signaling cascades are affected by altered redox potential. Key contributors to altered redox potential are reactive oxygen species (ROS) which are formed, in most cases, by exogenous genotoxic agents including irradiation, inflammatory cytokines and chemical carcinogens. ROS and altered redox potential can be considered as the primary intracellular changes which regulate protein kinases, thereby serving as an important cellular component linking external stimuli with signal transduction in stress response. The mechanisms, which underlie the ROS-mediated response, involve direct alteration of kinases and transcription factors, and indirect modulation of cysteine-rich redox-sensitive proteins exemplified by thioredoxin and glutathione S-transferase. This review summarizes the current understanding of the mechanisms contributing to ROS-related changes in key stress activated signaling cascades.


Subject(s)
Oxidative Stress/physiology , Reactive Oxygen Species/metabolism , Signal Transduction , Animals , Gene Expression Regulation , Glutathione Transferase/metabolism , Humans , Oxidation-Reduction , Protein Kinases/genetics , Protein Kinases/metabolism , Thioredoxins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
11.
Biochim Biophys Acta ; 1450(3): 434-43, 1999 Jul 08.
Article in English | MEDLINE | ID: mdl-10395954

ABSTRACT

Phosphatidylinositol 3-kinase (PI-3 kinase) has been implicated in the regulation of many cellular processes, including growth and transformation. We describe the effect of glucocorticoids on cell growth, phosphoinositide formation and PI-3 kinase activity in Rous sarcoma virus-transformed hamster fibroblasts (HET-SR). Using a prolonged dexamethasone treatment of HET-SR cells we have selected a new glucocorticoid receptor-positive cell subline, HET-SR(h), that was resistant to growth inhibitory action of dexamethasone and/or non-hormonal drugs (vinblastine, adriamycin) and was characterized by higher levels of phosphoinositide formation and increased PI-3 kinase activity. Study of the short-term hormone action has shown that both dexamethasone-sensitive and -resistant sublines responded to hormone by a decrease in phospholipid turnover rate. At the same time, in both cell lines activation of PI-3 kinase after dexamethasone addition was revealed. Dexamethasone-dependent activation of PI-3 kinase was more significant and maintained for a longer period in HET-SR(h) cells than in parent HET-SR cells. Finally, by transfecting p110*, a constitutively active catalytic subunit of PI-3 kinase, into hormone-sensitive HET-SR cells, we have found a marked increase in cell resistance to growth inhibitory dexamethasone action. These results suggest that PI-3 kinase may serve as one of the factors providing cell resistance to cytostatic drugs.


Subject(s)
Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Animals , Cell Division/drug effects , Cell Line, Transformed , Cell Survival , Cricetinae , Doxorubicin/pharmacology , Drug Resistance , Enzyme Activation/drug effects , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositols/analysis , Receptors, Glucocorticoid/analysis , Time Factors , Transfection , Vinblastine/pharmacology
12.
EMBO J ; 18(5): 1321-34, 1999 Mar 01.
Article in English | MEDLINE | ID: mdl-10064598

ABSTRACT

Studies of low basal Jun N-terminal kinase (JNK) activity in non-stressed cells led us to identify a JNK inhibitor that was purified and identified as glutathione S-transferase Pi (GSTp) and was characterized as a JNK-associated protein. UV irradiation or H2O2 treatment caused GSTp oligomerization and dissociation of the GSTp-JNK complex, indicating that it is the monomeric form of GSTp that elicits JNK inhibition. Addition of purified GSTp to the Jun-JNK complex caused a dose-dependent inhibition of JNK activity. Conversely, immunodepleting GSTp from protein extracts attenuated JNK inhibition. Furthermore, JNK activity was increased in the presence of specific GSTp inhibitors and a GSTp-derived peptide. Forced expression of GSTp decreased MKK4 and JNK phosphorylation which coincided with decreased JNK activity, increased c-Jun ubiquitination and decreased c-Jun-mediated transcription. Co-transfection of MEKK1 and GSTp restored MKK4 phosphorylation but did not affect GSTp inhibition of JNK activity, suggesting that the effect of GSTp on JNK is independent of the MEKK1-MKK4 module. Mouse embryo fibroblasts from GSTp-null mice exhibited a high basal level of JNK activity that could be reduced by forced expression of GSTp cDNA. In demonstrating the relationships between GSTp expression and its association with JNK, our findings provide new insight into the regulation of stress kinases.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Glutathione Transferase/metabolism , MAP Kinase Kinase 4 , Mitogen-Activated Protein Kinase Kinases , Mitogen-Activated Protein Kinases , 3T3 Cells , Animals , Enzyme Inhibitors/pharmacology , Free Radical Scavengers/pharmacology , Gene Expression Regulation/genetics , Glutathione Transferase/genetics , JNK Mitogen-Activated Protein Kinases , Mice , Mice, Knockout , Oligopeptides/pharmacology , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Transfection , Ubiquitins/metabolism , Ultraviolet Rays
13.
Mol Carcinog ; 24(1): 64-9, 1999 Jan.
Article in English | MEDLINE | ID: mdl-10029412

ABSTRACT

The activity of phosphatidylinositol 3-kinase (PI3K), a key component of multiple signal transduction pathways, was investigated in early- and late-stage melanoma cells that have varying degrees of radiation resistance. Analysis of PI3K biproducts (PI-3,4-P2 and PI-3,4,5-triphosphate) revealed a direct correlation between radiation resistance and levels of PI3K activity. Treating melanoma cells with wortmanin or LY294002, two different PI3K inhibitors, decreased PI3K activity and caused a dose-dependent decrease in resistance to ultraviolet radiation. Lower resistance to radiation elicited by LY294002 coincided with increased apoptosis. To further establish the role of PI3K in radiation resistance, we transfected early-stage melanoma cells with the cDNA of p85, the regulatory subunit of PI3K. Clones that constitutively overexpressed p85 exhibited a higher degree of PI-3,4-P2 synthesis and a corresponding increase in their resistance to ultraviolet radiation. The results of this study point to the role of PI3K and its biproducts in radiation resistance of human melanoma cells.


Subject(s)
Melanoma/enzymology , Phosphatidylinositol 3-Kinases/metabolism , Radiation Tolerance , Ultraviolet Rays , Androstadienes/pharmacology , Apoptosis , Cell Line , Chromones/pharmacology , Dose-Response Relationship, Radiation , Enzyme Inhibitors/pharmacology , Humans , Morpholines/pharmacology , Phosphatidylinositols/metabolism , Radiation Tolerance/drug effects , Signal Transduction/drug effects , Signal Transduction/radiation effects , Skin/cytology , Skin/enzymology , Skin/radiation effects , Tumor Cells, Cultured , Wortmannin
14.
J Protein Chem ; 18(8): 859-66, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10839622

ABSTRACT

We have recently found that the glutathione-S-transferase pi-isozyme (GST-pi), a cellular detoxification enzyme, potently and selectively inhibits activation of jun protein by its upstream kinase, jun kinase (JNK). This newly identified regulatory activity of GST-pi is strongly inhibited by a group of agents that inhibit its enzymatic activity. Since loss of enzymatic activity in general does not correlate with loss of regulatory activity, it is likely that inhibitor binding induces changes in the structure of one or more domains of GST that block its interaction with JNK. To identify regions of GST that change conformation on the binding of inhibitors, we have performed molecular dynamics calculations on GST-pi to compute its average structure in the presence and absence of the inhibitor, glutathione sulfonate. Superposition of the two average structures reveals that several regions change local structure depending upon whether the inhibitor is bound or not bound. Two of these regions, residues 36-50 and 194-201, are highly exposed. We have synthesized peptides corresponding to these two segments and find that the 194-201 sequence strongly inhibits the ability of GST-pi to block the in vitro phosphorylation of jun by JNK. These results suggest that this region of GST-pi is critical to its functioning as a newly discovered regulator of signal transduction.


Subject(s)
Glutathione Transferase/chemistry , Glutathione Transferase/metabolism , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Amino Acid Sequence , Crystallography, X-Ray , Glutathione/analogs & derivatives , Glutathione/chemistry , Glutathione/metabolism , Glutathione S-Transferase pi , Glutathione Transferase/antagonists & inhibitors , Isoenzymes/antagonists & inhibitors , Isoenzymes/chemistry , Isoenzymes/metabolism , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinases/metabolism , Models, Molecular , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Conformation
15.
Oncogene ; 17(19): 2543-7, 1998 Nov 12.
Article in English | MEDLINE | ID: mdl-9824166

ABSTRACT

Key to p53 ability to mediate its multiple cellular functions lies in its stability. In the present study we have elucidated the mechanism by which Mdm2 regulates p53 degradation. Using in vitro and in vivo ubiquitination assays we demonstrate that Mdm2 association with p53 targets p53 ubiquitination. Exposure of cells to UV-irradiation inhibits this targeting. Mdm2 which is deficient in p53 binding failed to target p53 ubiquitination, suggesting that the association is essential for Mdm2 targeting ability. While mdm2-p53 complex is found in non-stressed cells, the amount of p53-bound mdm2 is decreased after UV-irradiation, further pointing to the relationship between mdm2 binding and p53 level. Similar to Swiss 3T3 cells, the dissociation of mdm2-p53 complex was also found in UV-treated Scid cells, lacking functional DNA-PK, suggesting that DNA-PK is not sufficient for dissociating mdm2 from p53. Together our studies point to the role of Mdm2, as one of p53-associated proteins, in targeting p53 ubiquitination.


Subject(s)
DNA-Binding Proteins , Nuclear Proteins , Proto-Oncogene Proteins/physiology , Tumor Suppressor Protein p53/metabolism , 3T3 Cells , Animals , DNA Damage , DNA-Activated Protein Kinase , Fibroblasts , Macromolecular Substances , Mice , Mice, Inbred BALB C , Mice, SCID , Phosphorylation , Protein Conformation , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-mdm2 , Recombinant Fusion Proteins/metabolism , Tumor Suppressor Protein p53/radiation effects , Ubiquitins/metabolism , Ultraviolet Rays
16.
Genes Dev ; 12(17): 2658-63, 1998 Sep 01.
Article in English | MEDLINE | ID: mdl-9732264

ABSTRACT

In this study we elucidated the role of nonactive JNK in regulating p53 stability. The amount of p53-JNK complex was inversely correlated with p53 level. A peptide corresponding to the JNK binding site on p53 efficiently blocked ubiquitination of p53. Similarly, p53 lacking the JNK binding site exhibits a longer half-life than p53(wt). Outcompeting JNK association with p53 increased the level of p53, whereas overexpression of a phosphorylation mutant form of JNK inhibited p53 accumulation. JNK-p53 and Mdm2-p53 complexes were preferentially found in G0/G1 and S/G2M phases of the cell cycle, respectively. Altogether, these data indicate that JNK is an Mdm2-independent regulator of p53 stability in nonstressed cells.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Mitogen-Activated Protein Kinases , Nuclear Proteins , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Ubiquitins/metabolism , 3T3 Cells , Amino Acid Sequence , Animals , Calcium-Calmodulin-Dependent Protein Kinases/chemistry , Cell Line , JNK Mitogen-Activated Protein Kinases , Mice , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Phosphorylation , Proto-Oncogene Proteins c-mdm2 , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Reticulocytes/metabolism , Sequence Deletion , Spodoptera , Transfection , Tumor Suppressor Protein p53/chemistry
17.
Proc Natl Acad Sci U S A ; 95(18): 10541-6, 1998 Sep 01.
Article in English | MEDLINE | ID: mdl-9724739

ABSTRACT

Activation of the tumor suppressor p53 by stress and damage stimuli often correlates with induction of stress kinases, Jun-NH2 kinase (JNK). As JNK association with p53 plays an important role in p53 stability, in the present study we have elucidated the relationship between the JNK-signaling pathway and p53 stability and activity. Expression of a constitutively active form of JNKK upstream kinase, mitogen-activated protein kinase kinase kinase (DeltaMEKK1), increased the level of the exogenously transfected form of p53 in p53 null (10.1) cells as well as of endogenous p53 in MCF7 breast cancer cells. Increased p53 level by forced expression of DeltaMEKK1 coincided with a decrease in p53 ubiquitination in vivo and with prolonged p53 half-life. Computerized modeling of the JNK-binding site (amino acids 97-116; p7 region) enabled us to design mutations of exposed residues within this region. Respective mutations (p53(101-5-8)) and deletion (p53(Deltap7)) forms of p53 did not exhibit the same increase in p53 levels upon DeltaMEKK1 expression. In vitro phosphorylation of p53 by JNK abolished Mdm2 binding and targeting of p53 ubiquitination. Similarly, DeltaMEKK1 expression increased p53 phosphorylation by immunopurified JNK and dissociated p53-Mdm2 complexes. Transcriptional activity of p53, as measured via mdm2 promoter-driven luciferase, exhibited a substantial increase in DeltaMEKK1-expressing cells. Cotransfection of p53 and DeltaMEKK1 into p53 null cells potentiated p53-dependent apoptosis, suggesting that MEKK1 effectors contribute to the ability of p53 to mediate programmed cell death. Our results point to the role of MEKK1-JNK signaling in p53 stability, transcriptional activities, and apoptotic capacity as part of the cellular response to stress.


Subject(s)
MAP Kinase Kinase Kinase 1 , Nuclear Proteins , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Apoptosis , Cell Line, Transformed , Cells, Cultured , Computer Simulation , Humans , Phosphorylation , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-mdm2 , Transcriptional Activation , Ubiquitins/metabolism
18.
Proc Natl Acad Sci U S A ; 95(5): 2284-9, 1998 Mar 03.
Article in English | MEDLINE | ID: mdl-9482877

ABSTRACT

Treatment of mouse or human cells with the protein kinase C (PKC) inhibitors H7 or bisindolylmaleimide I induced an increase in the lifetime of p53, leading to its accumulation. In inhibitor-treated cells, p53 translocated to the nuclei and bound to DNA but was not competent to induce transcription. However, transactivation could be induced by subsequent DNA damage. Phorbol ester, a potent activator of PKC, significantly inhibited the accumulation of p53 after DNA damage. Therefore, constitutive PKC-dependent phosphorylation of p53 itself, or of a protein that interacts with p53, is required for the rapid degradation of p53 in untreated cells. Furthermore, an increase in the lifetime of p53 is not accompanied necessarily by its activation. Treatment with the PKC inhibitors decreased the overall level of p53 phosphorylation but led to the appearance of a phosphopeptide not seen in tryptic digests of p53 from untreated cells. Therefore, the lifetime and activities of p53 are likely to be regulated by distinct alterations of the phosphorylation pattern of p53, probably caused by the actions of different kinases.


Subject(s)
1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Enzyme Inhibitors/pharmacology , Tumor Suppressor Protein p53/metabolism , Amino Acid Sequence , Animals , Cell Line , Cell Nucleus/metabolism , Doxorubicin/pharmacology , Humans , Indoles/pharmacology , Kinetics , Maleimides/pharmacology , Mice , Molecular Sequence Data , Peptide Fragments/chemistry , Phosphorylation , Protein Kinase C/antagonists & inhibitors , RNA, Messenger/biosynthesis , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Transcription, Genetic/drug effects , Transcription, Genetic/radiation effects , Transfection , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/chemistry , Ultraviolet Rays
19.
Oncogene ; 16(4): 523-31, 1998 Jan 29.
Article in English | MEDLINE | ID: mdl-9484842

ABSTRACT

We have previously identified a U.V.-response element (URE; TGACAACA) and its bound proteins, members of the AP1 and ATF transcription factor families, in melanoma cells. Using a mutant form of cylic AMP response element binding (CREB), we found that CREB-associated-URE-bound proteins conferred characteristic melanoma phenotypes, including radiation resistance (Oncogene 12: 2223, 1996). In the present study we sought to determine which of the CREB-associated proteins confers radiation resistance on human melanoma cells. To this end we purified and identified via microsequencing ATF2 as a major URE- bound and CREB-associated protein in MeWo cells--a late stage human melanoma cell line. To determine the contribution of ATF2 to radiation resistance, MeWo cells were transfected with ATF2 cDNA lacking the trans-activation domain (ATF2(delta1-195)). MeWo cells that stably express ATF2(delta1-195) showed weaker transcriptional activities and an altered pattern of homo/hetero dimers. ATF2(delta1-195) clones exhibited up to tenfold lower resistance to irradiation by either U.V. or X-rays. The degree of resistance to radiation in the ATF2(delta1-195)-expressing clones could be increased upon transient transfection with ATF2(wt), but not with phosphorylation-defective mutant ATF2(69,71). Similarly, transfection of ATF2(wt) to WM3211, an early stage human melanoma cells line, increased resistance to radiation. Finally, changes elicited through ATF2(delta1-195) also led to reduced drug resistance, as shown for MMC, araC and cisplatinum. Our results suggest that ATF2 is a regulator of radiation and drug resistance in melanomas, and that tumor targeted ATF2 modulators may be useful sensitizers in the treatment of tumors of this type.


Subject(s)
Cyclic AMP Response Element-Binding Protein/physiology , Melanoma , Neoplasm Proteins/physiology , Radiation Tolerance/physiology , Transcription Factors/physiology , Activating Transcription Factor 2 , Cell Division/radiation effects , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Drug Resistance, Neoplasm , Humans , Melanoma/genetics , Melanoma/pathology , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasm Staging , Oligonucleotides/chemistry , Oligonucleotides/metabolism , Phosphorylation , Radiation Tolerance/genetics , Recombinant Fusion Proteins/genetics , Saccharomyces cerevisiae Proteins , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic , Transfection , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/radiation effects
20.
Proc Natl Acad Sci U S A ; 94(5): 1686-91, 1997 Mar 04.
Article in English | MEDLINE | ID: mdl-9050839

ABSTRACT

Phosphorylation of the p53 tumor suppressor protein is known to modulate its functions. Using bacterially produced glutathione S-transferase (GST)-p53 fusion protein and baculovirus-expressed histidine-tagged p53 ((His)p53), we have determined human p53 phosphorylation by purified forms of jun-N-kinase (JNK), protein kinase A (PKA), and beta subunit of casein kinase II (CKIIbeta) as well as by kinases present in whole cell extracts (WCEs). We demonstrate that PKA is potent p53 kinase, albeit, in a conformation- and concentration-dependent manner, as concluded by comparing full-length with truncated forms of p53. We further demonstrate JNK interaction with GST-p53 and the ability of JNK to phosphorylate truncated forms of GST-p53 or full-length (His)p53. Dependence of phosphorylation on conformation of p53 is further supported by the finding that the wild-type form of p53 (p53wt) undergoes better phosphorylation by CKIIbeta and by WCE kinases than mutant forms of p53 at amino acid 249 (p53(249)) or 273 (p53(273)). Moreover, shifting the kinase reaction's temperature from 37 degrees C to 18 degrees C reduces the phosphorylation of mutant p53 to a greater extent than of p53wt. Comparing truncated forms of p53 revealed that the ability of CKIIbeta, PKA, or WCE kinases to phosphorylate p53 requires amino acids 97-155 within the DNA-binding domain region. Among three 20-aa peptides spanning this region we have identified residues 97-117 that increase p53 phosphorylation by CKIIbeta while inhibiting p53 phosphorylation by PKA or WCE kinases. The importance of this region is further supported by computer modeling studies, which demonstrated that mutant p53(249) exhibits significant changes to the conformation of p53 within amino acids 97-117. In summary, phosphorylation-related analysis of different p53 forms in vitro indicates that conformation of p53 is a key determinant in its availability as a substrate for different kinases, as for the phosphorylation pattern generated by the same kinase.


Subject(s)
Mitogen-Activated Protein Kinases , Protein Conformation , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/metabolism , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Casein Kinase II , Cyclic AMP-Dependent Protein Kinases/metabolism , DNA/metabolism , Electrophoresis, Polyacrylamide Gel , Humans , JNK Mitogen-Activated Protein Kinases , Models, Molecular , Molecular Sequence Data , Peptide Fragments/chemical synthesis , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Peptide Mapping , Phosphorylation , Point Mutation , Protein Serine-Threonine Kinases/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Tumor Suppressor Protein p53/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...