Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 131
Filter
1.
Exp Cell Res ; 269(1): 64-72, 2001 Sep 10.
Article in English | MEDLINE | ID: mdl-11525640

ABSTRACT

The p53-regulated stress-inducible gene GADD45 has been shown to participate in cellular response to DNA damage, including cell cycle checkpoint, apoptosis, and DNA repair. However, the regulation of GADD45 expression is complex and may involve both p53-dependent and -independent pathways. Recent findings have demonstrated that the p53-independent induction of GADD45 is mainly regulated by the transcription factors Oct-1 and NF-YA, which directly bind to their consensus motifs located at the GADD45 promoter region. Here, we report that mitogen-activated protein (MAP) kinases are involved in the induction of the GADD45 promoter after DNA damage. Inhibition of JNK1 and ERK kinase activities either by expression of the dominant negative mutant JNK1 or by treatment with a selective chemical inhibitor of ERK (PD098059) substantially abrogates the UV induction of the GADD45 promoter. In contrast, a p38 kinase inhibitor (SB203580) has little effect on GADD45 induction by UV. In addition, the GADD45 promoter is strongly activated following expression of JNK1; Raf-1, which is an upstream activator of the ERK pathway; or MEK1, an upstream activator of both the ERK and the JNK pathways. Activation of the GADD45 promoter by MAP kinases does not require normal p53 function. Interestingly, the MAP kinase-regulatory effect appears to be mediated via OCT-1 and CAAT motifs since disruption of these sites abrogates activation of the GADD45 promoter by MAP kinases. Therefore, these findings indicate that the MAP kinase pathways are involved in the regulation of the p53-independent induction of the GADD45 promoter, probably via interaction with transcription factors that directly bind to OCT-1 and CAAT motifs.


Subject(s)
DNA Damage/physiology , Gene Expression Regulation/physiology , MAP Kinase Signaling System/physiology , Promoter Regions, Genetic/physiology , Proteins/genetics , Transcription Factors/metabolism , Amino Acid Motifs/physiology , Amino Acid Motifs/radiation effects , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , DNA Damage/radiation effects , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation/radiation effects , Host Cell Factor C1 , Humans , Intracellular Signaling Peptides and Proteins , MAP Kinase Signaling System/radiation effects , Mitogen-Activated Protein Kinase 8 , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Mitogen-Activated Protein Kinases/radiation effects , Octamer Transcription Factor-1 , Promoter Regions, Genetic/radiation effects , Protein Structure, Tertiary/genetics , Proteins/metabolism , Proteins/radiation effects , Proto-Oncogene Proteins c-raf/genetics , Proto-Oncogene Proteins c-raf/metabolism , Regulatory Sequences, Nucleic Acid/physiology , Stress, Physiological/genetics , Stress, Physiological/metabolism , Transcription Factors/genetics , Transcription Factors/radiation effects , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/radiation effects , Ultraviolet Rays/adverse effects , GADD45 Proteins
2.
Mol Cell Biol ; 21(17): 5889-98, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11486028

ABSTRACT

Cellular aging is accompanied by alterations in gene expression patterns. Here, using two models of replicative senescence, we describe the influence of the RNA-binding protein HuR in regulating the expression of several genes whose expression decreases during senescence. We demonstrate that HuR levels, HuR binding to target mRNAs encoding proliferative genes, and the half-lives of such mRNAs are lower in senescent cells. Importantly, overexpression of HuR in senescent cells restored a "younger" phenotype, while a reduction in HuR expression accentuated the senescent phenotype. Our studies highlight a critical role for HuR during the process of replicative senescence.


Subject(s)
Aging/genetics , Antigens, Surface , Gene Expression Regulation , RNA-Binding Proteins/metabolism , Aged , Aging/metabolism , Cell Line , Cellular Senescence , ELAV Proteins , ELAV-Like Protein 1 , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Phenotype , RNA, Messenger , Skin/cytology
3.
J Biol Chem ; 276(30): 28364-71, 2001 Jul 27.
Article in English | MEDLINE | ID: mdl-11350969

ABSTRACT

Phospholipase C-gamma1 (PLC-gamma1) is rapidly activated in response to growth factor stimulation and plays an important role in regulating cell proliferation and differentiation through the generation of the second messengers diacylglycerol and inositol 1,4,5-trisphosphate, leading to the activation of protein kinase C (PKC) and increased levels of intracellular calcium, respectively. Given the existing overlap between signaling pathways that are activated in response to oxidant injury and those involved in responding to proliferative stimuli, we investigated the role of PLC-gamma1 during the cellular response to oxidative stress. Treatment of normal mouse embryonic fibroblasts (MEF) with H2O2 resulted in time- and concentration-dependent tyrosine phosphorylation of PLC-gamma1. Phosphorylation could be blocked by pharmacological inhibitors of Src family tyrosine kinases or the epidermal growth factor receptor tyrosine kinase, but not by inhibitors of the platelet-derived growth factor receptor or phosphatidylinositol 3-kinase. To investigate the physiologic relevance of H2O2-induced tyrosine phosphorylation of PLC-gamma1, we compared survival of normal MEF and PLC-gamma1-deficient MEF following exposure to H2O2. Treatment of PLC-gamma1-deficient MEF with H2O2 resulted in rapid cell death, whereas normal MEF were resistant to the stress. Pretreatment of normal MEF with a selective pharmacological inhibitor of PLC-gamma1, or inhibitors of inositol trisphosphate receptors and PKC, increased their sensitivity to H2O2, whereas treatment of PLC-gamma1-deficient MEF with agents capable of directly activating PKC and enhancing calcium mobilization significantly improved their survival. Finally, reconstitution of PLC-gamma1 protein expression in PLC-gamma1-deficient MEF restored cell survival following H2O2 treatment. These findings suggest an important protective function for PLC-gamma1 activation during the cellular response to oxidative stress.


Subject(s)
Isoenzymes/metabolism , JNK Mitogen-Activated Protein Kinases , Oxidative Stress , Type C Phospholipases/metabolism , Alleles , Animals , Cell Death , Cell Differentiation , Cell Division , Cell Line , Cell Survival , Dose-Response Relationship, Drug , Enzyme Activation , Enzyme Inhibitors/pharmacology , Fibroblasts/metabolism , Hydrogen Peroxide/pharmacology , Immunoblotting , MAP Kinase Kinase 4 , Mice , Mitogen-Activated Protein Kinase Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phospholipase C gamma , Phosphorylation , Precipitin Tests , Protein Kinase C/metabolism , Signal Transduction , Time Factors , Tyrosine/metabolism
4.
J Biol Chem ; 276(30): 28546-53, 2001 Jul 27.
Article in English | MEDLINE | ID: mdl-11352915

ABSTRACT

c-Jun, a member of the activation protein 1 (AP-1) family of transcription factors, has been implicated in the regulation of many important biological processes including cell cycle progression, transformation, differentiation, and apoptosis. Accordingly, its expression and function are upregulated in response to diverse stimuli including mitogens and a wide range of stresses. Transcriptional activation of the c-Jun protein is dependent on its phosphorylation at Ser-63 and Ser-73, a process mediated by c-Jun N-terminal kinase. Active c-Jun is required for AP-1 transactivation and c-Jun-mediated transformation, but its role during stress remains unclear as both pro-apoptotic and pro-survival effects of c-Jun have been observed. Here we investigated the importance of c-Jun N-terminal phosphorylation in influencing the sensitivity of human T98G glioblastoma cells to a variety of cytotoxic agents. Stable expression of a nonphosphorylatable dominant negative protein c-Jun(S63A,S73A) markedly inhibited the activation of AP-1-driven transcription and greatly increased the cytotoxic effects of DNA-damaging agents associated with enhanced apoptosis. However, the same cells expressing the mutant Jun protein did not differ from parental cells in their sensitivity to several non-DNA-damaging cytotoxic agents. Our results suggest that activated c-Jun has a selective role in protecting human tumor cells from apoptosis induced by DNA damage.


Subject(s)
DNA Damage , Proto-Oncogene Proteins c-jun/physiology , Antineoplastic Agents/pharmacology , Apoptosis , Chloramphenicol O-Acetyltransferase/metabolism , Cisplatin/pharmacology , Flow Cytometry , Humans , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinases/metabolism , Mutagens , Phosphorylation , Serine/chemistry , Stress, Physiological , Time Factors , Transcription Factor AP-1/metabolism , Transcription, Genetic , Transcriptional Activation , Transfection , Tumor Cells, Cultured
5.
Mol Cell Biol ; 21(8): 2743-54, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11283254

ABSTRACT

The p53 tumor suppressor protein plays a key role in the regulation of stress-mediated growth arrest and apoptosis. Stress-induced phosphorylation of p53 tightly regulates its stability and transcriptional activities. Mass spectrometry analysis of p53 phosphorylated in 293T cells by active Jun NH2-terminal kinase (JNK) identified T81 as the JNK phosphorylation site. JNK phosphorylated p53 at T81 in response to DNA damage and stress-inducing agents, as determined by phospho-specific antibodies to T81. Unlike wild-type p53, in response to JNK stimuli p53 mutated on T81 (T81A) did not exhibit increased expression or concomitant activation of transcriptional activity, growth inhibition, and apoptosis. Forced expression of MKP5, a JNK phosphatase, in JNK kinase-expressing cells decreased T81 phosphorylation while reducing p53 transcriptional activity and p53-mediated apoptosis. Similarly transfection of antisense JNK 1 and -2 decreased T81 phosphorylation in response to UV irradiation. More than 180 human tumors have been reported to contain p53 with mutations within the region that encompasses T81 and the JNK binding site (amino acids 81 to 116). Our studies identify an additional mechanism for the regulation of p53 stability and functional activities in response to stress.


Subject(s)
Mitogen-Activated Protein Kinases/metabolism , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/metabolism , Animals , Base Sequence , Binding Sites , Cell Division , Cell Line , DNA Primers/genetics , Drug Stability , Dual-Specificity Phosphatases , Genes, p53 , Humans , Intracellular Signaling Peptides and Proteins , JNK Mitogen-Activated Protein Kinases , MAP Kinase Kinase 4 , Mass Spectrometry , Mice , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinase Phosphatases , Mitogen-Activated Protein Kinases/genetics , Mutation , Neoplasms/genetics , Neoplasms/metabolism , Oligonucleotides, Antisense/pharmacology , Phosphorylation , Protein Tyrosine Phosphatases/metabolism , Stress, Physiological/genetics , Stress, Physiological/metabolism , Threonine/chemistry , Transcription, Genetic , Tumor Suppressor Protein p53/genetics , Ultraviolet Rays
7.
Mol Cell Biol ; 21(4): 1249-59, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11158311

ABSTRACT

gadd153, also known as chop, is a highly stress-inducible gene that is robustly expressed following disruption of homeostasis in the endoplasmic reticulum (ER) (so-called ER stress). Although all reported types of ER stress induce expression of Gadd153, its role in the stress response has remained largely undefined. Several studies have correlated Gadd153 expression with cell death, but a mechanistic link between Gadd153 and apoptosis has never been demonstrated. To address this issue we employed a cell model system in which Gadd153 is constitutively overexpressed, as well as two cell lines in which Gadd153 expression is conditional. In all cell lines, overexpression of Gadd153 sensitized cells to ER stress. Investigation of the mechanisms contributing to this effect revealed that elevated Gadd153 expression results in the down-regulation of Bcl2 expression, depletion of cellular glutathione, and exaggerated production of reactive oxygen species. Restoration of Bcl2 expression in Gadd153-overexpressing cells led to replenishment of glutathione and a reduction in levels of reactive oxygen species, and it protected cells from ER stress-induced cell death. We conclude that Gadd153 sensitizes cells to ER stress through mechanisms that involve down-regulation of Bcl2 and enhanced oxidant injury.


Subject(s)
CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Endoplasmic Reticulum/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Animals , Base Sequence , Cell Line , DNA Primers/genetics , Down-Regulation , Gene Expression , Genes, bcl-2 , Glutathione/metabolism , HeLa Cells , Humans , Mice , Mutagenesis, Site-Directed , Oxidation-Reduction , Oxidative Stress , Proto-Oncogene Proteins c-bcl-2/genetics , Transcription Factor CHOP
8.
Free Radic Biol Med ; 29(11): 1166-76, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11121725

ABSTRACT

While the Ku complex, comprised of Ku70 and Ku80, is primarily involved in the repair of DNA double-strand breaks, it is also believed to participate in additional cellular processes. Here, treatment of embryo fibroblasts (MEFs) derived from either wild-type or Ku80-null (Ku80(-/-)) mice with various stress agents revealed that hydrogen peroxide (H(2)O(2)) was markedly more cytotoxic for Ku80(-/-) MEFs and led to their long-term accumulation in the G2 phase. This differential response was not due to differences in DNA repair, since H(2)O(2)-triggered DNA damage was repaired with comparable efficiency in both Wt and Ku80(-/-) MEFs, but was associated with differences in the expression of important cell cycle regulatory genes. Our results support the notion that Ku80-mediated cytoprotection and G2-progression are not only dependent on the cell's DNA repair but also may reflect Ku80's influence on additional cellular processes such as gene expression.


Subject(s)
Antigens, Nuclear , DNA Helicases , DNA Repair , DNA-Binding Proteins/deficiency , G2 Phase/drug effects , Hydrogen Peroxide/pharmacology , Nuclear Proteins/deficiency , Animals , Cell Line , Cell Survival/drug effects , Colony-Forming Units Assay , Cyclins/genetics , DNA Damage , DNA-Binding Proteins/physiology , Embryo, Mammalian , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Flow Cytometry , Free Radicals , Gamma Rays , Immunosorbent Techniques , Ku Autoantigen , Mice , Mice, Knockout , Nuclear Proteins/physiology
9.
Nature ; 408(6809): 239-47, 2000 Nov 09.
Article in English | MEDLINE | ID: mdl-11089981

ABSTRACT

Living in an oxygenated environment has required the evolution of effective cellular strategies to detect and detoxify metabolites of molecular oxygen known as reactive oxygen species. Here we review evidence that the appropriate and inappropriate production of oxidants, together with the ability of organisms to respond to oxidative stress, is intricately connected to ageing and life span.


Subject(s)
Aging/physiology , Oxidants/physiology , Oxidative Stress , Animals , Cellular Senescence/physiology , Energy Intake , Humans , Longevity , Reactive Oxygen Species , Signal Transduction
10.
Biochem J ; 352 Pt 1: 219-25, 2000 Nov 15.
Article in English | MEDLINE | ID: mdl-11062076

ABSTRACT

Peroxynitrite is a potent oxidizing and nitrating species formed in a diffusion-limited reaction between nitrogen monoxide and superoxide. It induces apoptosis through unknown mechanisms and is believed to interfere with receptor tyrosine kinase signalling through nitration of tyrosine residues. One pathway emanating from receptor tyrosine kinases is that leading to activation of the anti-apoptotic kinase Akt. In the present study we provide evidence that peroxynitrite, administered to cells using two different delivery systems, results in the dose- and time-dependent activation of Akt. Akt activation is rapid and followed by phosphorylation of glycogen synthase kinase-3, an established substrate of Akt. Akt activation is inhibited in the presence of the phosphoinositide 3-kinase (PI-3K) inhibitors wortmannin and LY294002, and by treatment with the platelet-derived growth factor (PDGF) receptor (PDGFR) inhibitor AG1295, indicating a requirement for PDGFR and PI-3K in mediating peroxynitrite-induced Akt activation. Accordingly, the PDGFR-A and PDGFR-B isoforms were shown to undergo rapid tyrosine phosphorylation on treatment with peroxynitrite. Prior exposure of cells to peroxynitrite interferes with PDGF-induced Akt phosphorylation. Our findings suggest that Akt activation occurs as an acute response to peroxynitrite treatment and could play an important role in influencing cell survival and/or alter the cellular response to other growth regulatory signals.


Subject(s)
Fibroblasts/enzymology , Nitrates/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins , Skin/enzymology , Androstadienes/pharmacology , Apoptosis , Blotting, Western , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cells, Cultured , Chromones/pharmacology , Dose-Response Relationship, Drug , Enzyme Activation , Enzyme Inhibitors/pharmacology , Glycogen Synthase Kinase 3 , Glycogen Synthase Kinases , Humans , Molsidomine/analogs & derivatives , Molsidomine/pharmacology , Morpholines/pharmacology , Oxidative Stress , Phosphorylation , Precipitin Tests , Protein Isoforms , Proto-Oncogene Proteins c-akt , Receptors, Platelet-Derived Growth Factor/chemistry , Receptors, Platelet-Derived Growth Factor/metabolism , Signal Transduction , Time Factors , Tyrosine/metabolism , Tyrphostins/pharmacology , Wortmannin
11.
Mol Cell Biol ; 20(21): 7903-13, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11027261

ABSTRACT

Prostaglandin A(2) (PGA(2)), an experimental chemotherapeutic agent, causes growth arrest associated with decreased cyclin D1 expression in several cancer cell lines. Here, using human non-small-cell lung carcinoma H1299 cells, we investigated the mechanisms whereby PGA(2) down-regulates cyclin D1 expression. Transcription rates of the cyclin D1 gene, studied using a cyclin D1 promoter-luciferase construct and nuclear run-on assays, were not affected by PGA(2) treatment. Instead, the cyclin D1 mRNA was rendered unstable after exposure to PGA(2). Since the stability of labile mRNA is modulated through binding of proteins to specific mRNA sequences, we sought to identify protein(s) recognizing the cyclin D1 mRNA. In electrophoretic mobility-shift assays using radiolabeled RNA probes derived from different regions of cyclin D1 mRNA, we observed that (i) lysates prepared from PGA(2)-treated cells exhibited enhanced protein-cyclin D1 RNA complex formation; (ii) the kinetics of complex formation correlated closely with that of cyclin D1 mRNA loss; and (iii) binding occurred within a 390-base cyclin D1 3' untranslated region (UTR) (K12). This binding activity could be cross-linked, revealing proteins ranging from 30 to 47 kDa. The RNA-binding protein AUF1, previously associated with the degradation of target mRNAs, bound cyclin D1 mRNA, because anti-AUF1 antibodies were capable of supershifting or immunoprecipitating cyclin D1 mRNA-protein complexes. Finally, insertion of K12 in the 3'UTR of reporter genes markedly reduced the expression and half-life of the resulting chimeric mRNAs in transfected, PGA(2)-treated cells. Our data demonstrate that PGA(2) down-regulates cyclin D1 expression by decreasing cyclin D1 mRNA stability and implicates a 390-base element in the 3'UTR in this regulation.


Subject(s)
Cyclin D1/genetics , Cyclin D1/metabolism , Down-Regulation , Heterogeneous-Nuclear Ribonucleoprotein D , Prostaglandins A/metabolism , RNA, Messenger/metabolism , 3' Untranslated Regions , Blotting, Northern , Blotting, Western , Cell Division/drug effects , Cell Nucleus/metabolism , Cross-Linking Reagents/pharmacology , Cytoplasm/metabolism , Dose-Response Relationship, Drug , Genes, Reporter , Heterogeneous Nuclear Ribonucleoprotein D0 , Humans , Kinetics , Models, Genetic , Precipitin Tests , Promoter Regions, Genetic , Prostaglandins A/genetics , Protein Binding , RNA/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Subcellular Fractions , Time Factors , Transcription, Genetic , Transcriptional Activation , Transfection , Tumor Cells, Cultured
12.
J Biol Chem ; 275(50): 39435-43, 2000 Dec 15.
Article in English | MEDLINE | ID: mdl-10993883

ABSTRACT

Cisplatin activates multiple signal transduction pathways involved in coordinating cellular responses to stress. Here we demonstrate a requirement for extracellular signal-regulated protein kinase (ERK), a member of the mitogen-activated protein kinase family in mediating cisplatin-induced apoptosis of human cervical carcinoma HeLa cells. Cisplatin treatment resulted in dose- and time- dependent activation of ERK. That elevated ERK activity contributed to cell death by cisplatin was supported by several observations: 1) PD98059 and U0126, chemical inhibitors of the MEK/ERK signaling pathway, prevented apoptosis; 2) pretreatment of cells with TPA, an activator of the ERK pathway, enhanced their sensitivity to cisplatin; 3) suramin, a growth factor receptor antagonist that greatly suppressed ERK activation, likewise inhibited cisplatin-induced apoptosis; and, finally, 4) HeLa cell variants selected for cisplatin resistance showed reduced activation of ERK following cisplatin treatment. Cisplatin-induced apoptosis was associated with cytochrome c release and subsequent caspase-3 activation, both of which could be prevented by treatment with the MEK inhibitors. However, the caspase inhibitor benzyloxycarbonyl-Val-Ala-Asp-fluoromethylketone protected HeLa cells against apoptosis without affecting ERK activation. Taken together, our findings suggest that ERK activation plays an active role in mediating cisplatin-induced apoptosis of HeLa cells and functions upstream of caspase activation to initiate the apoptotic signal.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cisplatin/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Amino Acid Chloromethyl Ketones/pharmacology , Blotting, Western , Butadienes/pharmacology , Caspase 3 , Caspases/metabolism , Cell Line , Cell Survival/drug effects , Cysteine Proteinase Inhibitors/pharmacology , Cytochrome c Group/metabolism , Dose-Response Relationship, Drug , Enzyme Activation , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , HeLa Cells , Humans , Indoles/pharmacology , MAP Kinase Kinase 1 , MAP Kinase Kinase 2 , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Models, Biological , Nitriles/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Signal Transduction , Suramin/pharmacology , Tetradecanoylphorbol Acetate/pharmacology , Time Factors
13.
J Biol Chem ; 275(19): 14624-31, 2000 May 12.
Article in English | MEDLINE | ID: mdl-10799549

ABSTRACT

The serine/threonine kinase Akt (also known as protein kinase B) is activated in response to various stimuli by a mechanism involving phosphoinositide 3-kinase (PI3-K). Akt provides a survival signal that protects cells from apoptosis induced by growth factor withdrawal, but its function in other forms of stress is less clear. Here we investigated the role of PI3-K/Akt during the cellular response to oxidant injury. H(2)O(2) treatment elevated Akt activity in multiple cell types in a time- (5-30 min) and dose (400 microM-2 mm)-dependent manner. Expression of a dominant negative mutant of p85 (regulatory component of PI3-K) and treatment with inhibitors of PI3-K (wortmannin and LY294002) prevented H(2)O(2)-induced Akt activation. Akt activation by H(2)O(2) also depended on epidermal growth factor receptor (EGFR) signaling; H(2)O(2) treatment led to EGFR phosphorylation, and inhibition of EGFR activation prevented Akt activation by H(2)O(2). As H(2)O(2) causes apoptosis of HeLa cells, we investigated whether alterations of PI3-K/Akt signaling would affect this response. Wortmannin and LY294002 treatment significantly enhanced H(2)O(2)-induced apoptosis, whereas expression of exogenous myristoylated Akt (an activated form) inhibited cell death. Constitutive expression of v-Akt likewise enhanced survival of H(2)O(2)-treated NIH3T3 cells. These results suggest that H(2)O(2) activates Akt via an EGFR/PI3-K-dependent pathway and that elevated Akt activity confers protection against oxidative stress-induced apoptosis.


Subject(s)
Cell Survival , ErbB Receptors/metabolism , Oxidative Stress , Retroviridae Proteins, Oncogenic/metabolism , Animals , Apoptosis/drug effects , Enzyme Activation , Humans , Hydrogen Peroxide/pharmacology , Oncogene Protein v-akt , Phosphatidylinositol 3-Kinases/metabolism
14.
J Biol Chem ; 275(32): 24767-75, 2000 Aug 11.
Article in English | MEDLINE | ID: mdl-10825181

ABSTRACT

The c-Jun N-terminal kinase/stress-activated protein kinase (JNK/SAPK) pathway is activated by numerous cellular stresses. Although it has been implicated in mediating apoptosis and growth factor signaling, its role in regulating cell growth is not yet clear. Here, the influence of JNK on basal (unstimulated) growth of human tumor glioblastoma T98G cells was investigated using highly specific JNK antisense oligonucleotides to inhibit JNK expression. Transient depletion of either JNK1 or JNK2 suppressed cell growth associated with an inhibition of DNA synthesis and cell cycle arrest in S phase. The growth-inhibitory potency of JNK2 antisense ((JNK)2 IC(50) = 0.14 micrometer) was greater than that of JNK1 antisense ((JNK)1 IC(50) = 0.37 micrometer), suggesting that JNK2 plays a dominant role in regulating growth of T98G cells. Indeed, JNK2 antisense-treated populations exhibited greater inhibition of DNA synthesis and accumulation of S-phase cells than did the JNK1 antisense-treated cultures, with a significant proportion of these cells detaching from the tissue culture plate. JNK2 (but not JNK1) antisense-treated cultures exhibited marked elevation in the expression of the cyclin-dependent kinase inhibitor p21(cip1/waf1) accompanied by inhibition of Cdk2/Cdc2 kinase activities. Taken together, these results indicate that JNK is required for growth of T98G cells in nonstress conditions and that p21(cip1/waf1) may contribute to the sustained growth arrest of JNK2-depleted T98G cultures.


Subject(s)
Cell Cycle/drug effects , Cell Division/physiology , Mitogen-Activated Protein Kinases/genetics , Oligodeoxyribonucleotides, Antisense/pharmacology , Transcription, Genetic/drug effects , Cell Division/drug effects , DNA, Neoplasm/biosynthesis , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma , Humans , JNK Mitogen-Activated Protein Kinases , Kinetics , Mitogen-Activated Protein Kinase 8 , Mitogen-Activated Protein Kinase 9 , Mitogen-Activated Protein Kinases/metabolism , S Phase , Thionucleotides , Tumor Cells, Cultured
15.
J Gerontol A Biol Sci Med Sci ; 55(3): B125-34, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10795716

ABSTRACT

Numerous studies have demonstrated that the proliferative capacity of cells declines with age. Using rat primary hepatocytes as a model system, we recently demonstrated that this age-related decline in the proliferative response to mitogenic stimulation is associated with decreased activities of both extracellular signal-regulated kinase (ERK) and p70 S6 kinase (p70(S6k)). To unravel the molecular basis for age-related defects in the ERK pathway, we have now characterized the upstream signaling events that occur after epidermal growth factor (EGF) stimulation in young and aged hepatocytes. As previously noted for ERK, the activities of both MEK (the kinase immediately upstream of ERK) and Ras following EGF stimulation were significantly lower in aged hepatocytes. An examination of the EGF receptor (EGFR) revealed a similar amount of EGFR in the two age groups. Likewise, EGFR and Shc, an adaptor protein that plays a crucial role in linking EGFR to Ras activation, underwent tyrosine phosphorylation to a similar degree in both young and aged hepatocytes. However, in aged cells Shc was unable to form stable complexes with EGFR after EGF stimulation. Our results suggest that a decrease in the association between Shc and EGFR in aged cells underlies the age-related declines in the ERK signaling cascade and in proliferative capacity.


Subject(s)
Adaptor Proteins, Signal Transducing , Cellular Senescence/physiology , ErbB Receptors/metabolism , MAP Kinase Signaling System/physiology , Ribosomal Protein S6 Kinases/metabolism , ras Proteins/metabolism , Animals , Cells, Cultured , GRB2 Adaptor Protein , Liver/chemistry , Liver/cytology , Liver/enzymology , MAP Kinase Kinase 1 , Male , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Proteins/metabolism , Rats , Rats, Wistar , Tyrosine/metabolism
16.
J Biol Chem ; 275(24): 18234-42, 2000 Jun 16.
Article in English | MEDLINE | ID: mdl-10748131

ABSTRACT

Transforming growth factor (TGF)-beta1, a pleiotropic cytokine involved in regulating growth and differentiation, can exert both pro-apoptotic and anti-apoptotic effects depending on the cell type or circumstances. We observed that TGF-beta1 blocked apoptosis resulting from serum withdrawal in A549 human lung carcinoma cells. This was associated with suppression of JNK activation that occurs concomitant with the onset of apoptosis in the absence of TGF-beta1, suggesting that JNK plays an active role in the death process and that TGF-beta1 exerts its protective influence by altering JNK activity. Overexpression of a dominant negative mutant form of SEK1, an upstream activator of JNK, likewise suppressed JNK activation and inhibited apoptosis. Investigation of early events following TGF-beta1 treatment revealed an early induction and phosphorylation of c-Jun that was absent in cells subjected to serum withdrawal alone. That TGF-beta1-induced expression of c-Jun is important for survival was supported by the finding that overexpression of non-phosphosphorylatable dominant negative mutant c-Jun, c-Jun(S73A), attenuated the protective influence of TGF-beta1. Our findings suggest that JNK activation is a late but essential event in serum deprivation-induced apoptosis in A549 cells. TGF-beta1 prevents apoptosis, in part, through the early induction and phosphorylation of c-Jun, which in turn results in attenuated JNK activation.


Subject(s)
Apoptosis/drug effects , MAP Kinase Kinase 4 , Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Transforming Growth Factor beta/pharmacology , Alanine/metabolism , Blood , Cell Survival , Cells, Cultured , Culture Media , Humans , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinase Kinases/biosynthesis , Phosphorylation , Transfection
17.
Mol Cell Biol ; 20(5): 1713-22, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10669748

ABSTRACT

c-Jun N-terminal kinase (JNK) plays a critical role in coordinating the cellular response to stress and has been implicated in regulating cell growth and transformation. To investigate the growth-regulatory functions of JNK1 and JNK2, we used specific antisense oligonucleotides (AS) to inhibit their expression. A survey of several human tumor cell lines revealed that JNKAS treatment markedly inhibited the growth of cells with mutant p53 status but not that of cells with normal p53 function. To further examine the influence of p53 on cell sensitivity to JNKAS treatment, we compared the responsiveness of RKO, MCF-7, and HCT116 cells with normal p53 function to that of RKO E6, MCF-7 E6, and HCT116 p53(-/-), which were rendered p53 deficient by different methods. Inhibition of JNK2 (and to a lesser extent JNK1) expression dramatically reduced the growth of p53-deficient cells but not that of their normal counterparts. JNK2AS-induced growth inhibition was correlated with significant apoptosis. JNK2AS treatment induced the expression of the cyclin-dependent kinase inhibitor p21(Cip1/Waf1) in parental MCF-7, RKO, and HCT116 cells but not in the p53-deficient derivatives. That p21(Cip1/Waf1) expression contributes to the survival of JNK2AS-treated cells was supported by additional experiments demonstrating that p21(Cip1/Waf1) deficiency in HCT116 cells also results in heightened sensitivity to JNKAS treatment. Our results indicate that perturbation of JNK2 expression adversely affects the growth of otherwise nonstressed cells. p53 and its downstream effector p21(Cip1/Waf1) are important in counteracting these detrimental effects and promoting cell survival.


Subject(s)
Apoptosis , Mitogen-Activated Protein Kinases , Protein Kinases/genetics , Tumor Suppressor Protein p53/genetics , Apoptosis/genetics , Cell Division/genetics , Cell Survival/genetics , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Mitogen-Activated Protein Kinase 9 , Protein Kinases/biosynthesis , Signal Transduction/genetics , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism
18.
Oncogene ; 18(44): 6021-8, 1999 Oct 28.
Article in English | MEDLINE | ID: mdl-10557091

ABSTRACT

All cells depend on sterols and isoprenoids derived from mevalonate (MVA) for growth, differentiation, and maintenance of homeostatic functions. In plants, environmental insults like heat and sunlight trigger the synthesis of isoprene, also derived from MVA, and this phenomenon has been associated with enhanced tolerance to heat. Here, we show that in human prostate adenocarcinoma PC-3M cells heat shock leads to activation of the MVA pathway. This is characterized by a dose- and time-dependent elevation in 3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMGR) activity, enhanced sterol and isoprenoid synthesis, and increased protein prenylation. Furthermore, prenylation and subsequent membrane localization of Ras, a central player in cell signaling, was rapidly induced following heat stress. These effects were dose-dependent, augmented with repeated insults, and were prevented by culturing cells in the presence of lovastatin, a competitive inhibitor of HMGR. Enhanced Ras maturation by heat stress was also associated with a heightened activation of extracellular signal-regulated kinase (ERK), a key mediator of both mitogenic and stress signaling pathways, in response to subsequent growth factor stimulation. Thus, activation of the MVA pathway may constitute an important adaptive host response to stress, and have significant implications to carcinogenesis.


Subject(s)
Adenocarcinoma/metabolism , Cholesterol/metabolism , Genes, ras , Hydroxymethylglutaryl CoA Reductases/metabolism , Prostatic Neoplasms/metabolism , Stress, Physiological/metabolism , Adenocarcinoma/drug therapy , Adenocarcinoma/etiology , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Diterpenes/metabolism , Farnesol/metabolism , Heat-Shock Response/genetics , Humans , Hydroxymethylglutaryl CoA Reductases/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Hydroxymethylglutaryl-CoA-Reductases, NADP-dependent , Lovastatin/pharmacology , Male , Mevalonic Acid/metabolism , Mitogen-Activated Protein Kinase 1 , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/etiology , Protein Prenylation , Sterols/biosynthesis , Stress, Physiological/complications , ras Proteins/genetics , ras Proteins/metabolism
19.
J Biol Chem ; 274(42): 29599-602, 1999 Oct 15.
Article in English | MEDLINE | ID: mdl-10514426

ABSTRACT

Cells respond to environmental stress with activation of c-Jun N-terminal kinase (JNK) and p38. Recent studies have implicated Gadd45 and two related proteins, MyD118/Gadd45beta and CR6/Gadd45gamma, as initiators of JNK/p38 signaling via their interaction with an upstream kinase MTK1. It was proposed that stress-induced expression of the Gadd45-related proteins leads to MTK1 activation and subsequent JNK/p38 activation. Using embryo fibroblasts from gadd45-null mice, we have addressed the requirement for Gadd45 in mediating JNK/p38 activation during acute stress. Comparison of JNK/p38 activities in response to methyl methanesulfonate, hydrogen peroxide, UVC irradiation, sorbitol, and anisomycin treatment of gadd45(+/+) and gadd45(-/-) fibroblasts revealed no deficiency in JNK/p38 activation in gadd45(-/-) fibroblasts. In addition, in wild type cells, JNK and p38 activation significantly preceded gadd45 induction with all stresses. Examination of myd118/gadd45beta and cr6/gadd45gamma expression in gadd45(+/+) and gadd45(-/-) fibroblasts revealed similar induction patterns in the two cell types, which, like gadd45 expression, was delayed relative to JNK/p38 activation. We conclude that gadd45 expression is not required for activation of JNK/p38 by environmental stresses, nor are stress-induced increases in myd118/gadd45beta and cr6/gadd45gamma expression necessary for kinase activation in response to such insults.


Subject(s)
Mitogen-Activated Protein Kinases/metabolism , Oxidative Stress , Proteins/metabolism , Animals , Cell Line , Enzyme Activation , Humans , Intracellular Signaling Peptides and Proteins , JNK Mitogen-Activated Protein Kinases , Mice , Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , p38 Mitogen-Activated Protein Kinases , GADD45 Proteins
20.
Oncogene ; 18(28): 4153-9, 1999 Jul 15.
Article in English | MEDLINE | ID: mdl-10435597

ABSTRACT

Both DR4 and DR5 have recently been identified as membrane death receptors that are activated by their ligand TRAIL to engage the intracellular apoptotic machinery. TRID (also named as TRAIL-R3) is an antagonist decoy receptor and lacks the cytoplasmic death domain. TRID protects from TRAIL-induced apoptosis by competing with DR4 and DR5 for binding to TRAIL. TRID has been shown to be overexpressed in normal human tissues but not in malignantly transformed cell lines. DR5 is a p53-regulated gene and we have recently reported that DR5 expression is induced in response to genotoxic stress in both a p53-dependent and independent manner (Sheikh et al., 1998). In the current study, we demonstrate that TRID gene expression is also induced by the genotoxic agents ionizing radiation and methyl methanesulfonate (MMS) in predominantly p53 wild-type cells, whereas UV-irradiation does not induce TRID gene expression. Consistent with these results, exogenous wild-type p53 also upregulates the expression of endogenous TRID in p53-null cells. Thus, TRID appears to be a p53 target gene that is regulated by genotoxic stress in a p53-dependent manner. Using primary gastrointestinal tract (GIT) tumors and their matching normal tissue, we also demonstrate for the first time that TRID expression is enhanced in primary tumors of the GIT. It is, therefore, possible that TRID overexpressing GIT tumors may gain a selective growth advantage by escaping from TRAIL-induced apoptosis.


Subject(s)
Apoptosis/genetics , Carcinoma/genetics , DNA Damage , DNA Repair/genetics , DNA, Neoplasm/genetics , Gastrointestinal Neoplasms/genetics , Gene Expression Regulation, Neoplastic/genetics , Neoplasm Proteins/genetics , Receptors, Tumor Necrosis Factor/genetics , Tumor Suppressor Protein p53/physiology , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Apoptosis Regulatory Proteins , Carcinoma/metabolism , Carcinoma/pathology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , DNA, Neoplasm/drug effects , DNA, Neoplasm/radiation effects , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Female , GPI-Linked Proteins , Gamma Rays , Gastrointestinal Neoplasms/metabolism , Gastrointestinal Neoplasms/pathology , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/radiation effects , Genes, p53 , Humans , Male , Membrane Glycoproteins/metabolism , Methyl Methanesulfonate/pharmacology , Middle Aged , Mutagens/pharmacology , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/physiology , Organ Specificity , Protein Conformation , Receptors, Tumor Necrosis Factor/biosynthesis , Receptors, Tumor Necrosis Factor/physiology , Receptors, Tumor Necrosis Factor, Member 10c , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Stress, Physiological/genetics , TNF-Related Apoptosis-Inducing Ligand , Temperature , Tumor Cells, Cultured , Tumor Necrosis Factor Decoy Receptors , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...