Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Mol Microbiol ; 100(5): 860-76, 2016 06.
Article in English | MEDLINE | ID: mdl-26878111

ABSTRACT

Cells possess stress-activated protein kinase (SAPK) signalling pathways, which are activated practically in response to any cellular insult, regulating responses for survival and adaptation to harmful environmental changes. To understand the function of SAPK pathways in T. atroviride, mutants lacking the MAPKK Pbs2 and the MAPK Tmk3 were analysed under several cellular stresses, and in their response to light. All mutants were highly sensitive to cellular insults such as osmotic and oxidative stress, cell wall damage, high temperature, cadmium, and UV irradiation. Under oxidative stress, the Tmk3 pathway showed specific roles during development, which in conidia are essential for tolerance to oxidant agents and appear to play a minor role in mycelia. The function of this pathway was more evident in Δpbs2 and Δtmk3 mutant strains when combining oxidative stress or cell wall damage with light. Light stimulates tolerance to osmotic stress through Tmk3 independently of the photoreceptor Blr1. Strikingly, photoconidiation and expression of blue light regulated genes was severally affected in Δtmk3 and Δpbs2 strains, indicating that this pathway regulates light responses. Furthermore, Tmk3 was rapidly phosphorylated upon light exposure. Thus, our data indicate that Tmk3 signalling cooperates with the Blr photoreceptor complex in the activation of gene expression.


Subject(s)
Fungal Proteins/metabolism , Light , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Stress, Physiological , Trichoderma/genetics , Trichoderma/metabolism , Cell Wall/metabolism , Fungal Proteins/genetics , Gene Expression Regulation, Fungal , Mitogen-Activated Protein Kinase 8/genetics , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinase Kinases/radiation effects , Mycelium/growth & development , Mycelium/metabolism , Osmotic Pressure , Phosphorylation , Signal Transduction/genetics , Signal Transduction/radiation effects , Spores, Fungal/metabolism , Spores, Fungal/radiation effects , Trichoderma/radiation effects
2.
Head Neck ; 35(2): 220-8, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22302684

ABSTRACT

BACKGROUND: Irradiation-induced signaling via the 2 pathways, Raf-MEK-ERK and PI3K-Akt, is known to be closely associated with a limited response to radiotherapy. In the present study we analyzed the relevance of constitutively active K-Ras for postradiogenic pathway stimulation and the option of coordinated inhibition to overcome these rescue mechanisms. METHODS: We used 2 epithelial tumor cell lines as a model system, one of them harboring a G12S K-Ras mutation. Cells were irradiated and the effect of combined treatment with ionizing radiation and inhibitors on the expression of pERK and pAkt was determined by Western blotting. Additionally, clonogenic assays were performed to functionally analyze survival of the cell lines. RESULTS: Compared with the nonmutated cells we observed the G12S cell line showing a clearly reduced response to inhibitor treatment under irradiation. In the case of pharmacologic inhibition of 1 of the pathways a compensatory upregulation of the second cascade leading to increased clonogenic survival seems feasible. However, there was a good functional response of this cell line to double inhibition with both compounds represented by minimized colony forming ability. The activation of ERK and Akt after irradiation was confirmed in xenotransplants showing elevated postradiogenic protein levels. CONCLUSION: With our data we confirmed our hypothesis of postradiogenic constitutive activation of the 2 pathways both required for Ras-mediated radioresistance in epithelial cells. If this effect should prove itself as a general mechanism in Ras-mutated tumors, application of specific inhibitors to block both cascades in parallel could contribute to enhance radiosensitivity in these types of cancer.


Subject(s)
Genes, ras/genetics , Mitogen-Activated Protein Kinase Kinases/radiation effects , Phosphatidylinositol 3-Kinases/radiation effects , Radiation Tolerance/genetics , Blotting, Western , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Electrophoresis, Polyacrylamide Gel , Epithelial Cells/radiation effects , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Humans , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinase Kinases/genetics , Phosphatidylinositol 3-Kinases/genetics , Polymerase Chain Reaction/methods , Sensitivity and Specificity , Signal Transduction/radiation effects , Up-Regulation
4.
J Biol Chem ; 279(20): 21628-36, 2004 May 14.
Article in English | MEDLINE | ID: mdl-14970211

ABSTRACT

Beta-amyloid precursor protein (APP) is the precursor of beta-amyloid (Abeta), which is implicated in Alzheimer's disease pathogenesis. APP complements amyloid precursor-like protein 2 (APLP2), and together they play essential physiological roles. Phosphorylation at the Thr(668) residue of APP (with respect to the numbering conversion for the APP 695 isoform) and the Thr(736) residue of APLP2 (with respect to the numbering conversion for the APLP2 763 isoform) in their cytoplasmic domains acts as a molecular switch for their protein-protein interaction and is implicated in neural function(s) and/or Alzheimer's disease pathogenesis. Here we demonstrate that both APP and APLP2 can be phosphorylated by JNK at the Thr(668) and Thr(736) residues, respectively, in response to cellular stress. X11-like (X11L, also referred to as X11beta and Mint2), which is a member of the mammalian LIN-10 protein family and a possible regulator of Abeta production, elevated APP and APLP2 phosphorylation probably by facilitating JNK-mediated phosphorylation, whereas other members of the family, X11 and X11L2, did not. These observations revealed an involvement of X11L in the phosphorylation of APP family proteins in cellular stress and suggest that X11L protein may be important in the physiology of APP family proteins as well as in the regulation of Abeta production.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Cadherins/metabolism , Carrier Proteins/metabolism , JNK Mitogen-Activated Protein Kinases , Nerve Tissue Proteins/metabolism , Adaptor Proteins, Signal Transducing , Amyloid beta-Protein Precursor/radiation effects , Anisomycin/pharmacology , Binding Sites , Cadherins/radiation effects , Carrier Proteins/radiation effects , Cell Line , Humans , Kidney , MAP Kinase Kinase 4 , Mitogen-Activated Protein Kinase Kinases/drug effects , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinase Kinases/radiation effects , Nerve Tissue Proteins/radiation effects , Phosphorylation , Protein Isoforms/metabolism , Recombinant Proteins/metabolism , Sorbitol/pharmacology , Threonine , Transfection , Ultraviolet Rays
5.
Oncogene ; 22(37): 5885-96, 2003 Sep 01.
Article in English | MEDLINE | ID: mdl-12947395

ABSTRACT

Within the last 15 years, multiple new signal transduction pathways within cells have been discovered. Many of these pathways belong to what is now termed 'the mitogen-activated protein kinase (MAPK) superfamily.' These pathways have been linked to the growth factor-mediated regulation of diverse cellular events such as proliferation, senescence, differentiation and apoptosis. Based on currently available data, exposure of cells to ionizing radiation and a variety of other toxic stresses induces simultaneous compensatory activation of multiple MAPK pathways. These signals play critical roles in controlling cell survival and repopulation effects following irradiation, in a cell-type-dependent manner. Some of the signaling pathways activated following radiation exposure are those normally activated by mitogens, such as the 'classical' MAPK (also known as the ERK) pathway. Other MAPK pathways activated by radiation include those downstream of death receptors and procaspases, and DNA-damage signals, including the JNK and P38 MAPK pathways. The expression and release of autocrine growth factor ligands, such as (transforming growth factor alpha) and TNF-alpha, following irradiation can also enhance the responses of MAPK pathways in cells and, consequently, of bystander cells. Thus, the ability of radiation to activate MAPK signaling pathways may depend on the expression of multiple growth factor receptors, autocrine factors and Ras mutation. Enhanced basal signaling by proto-oncogenes such as K-/H-/N-RAS may provide a radioprotective and growth-promoting signal. In many cell types, this may be via the PI3K pathway; in others, this may occur through nuclear factor-kappa B or multiple MAPK pathways. This review will describe the enzymes within the known MAPK signaling pathways and discuss their activation and roles in cellular radiation responses.


Subject(s)
Mitogen-Activated Protein Kinases/radiation effects , Signal Transduction/radiation effects , Animals , Cell Survival , Humans , JNK Mitogen-Activated Protein Kinases , MAP Kinase Kinase 5 , Mitogen-Activated Protein Kinase Kinases/radiation effects , Phosphatidylinositol 3-Kinases/radiation effects , p38 Mitogen-Activated Protein Kinases
6.
Cell Death Differ ; 9(9): 963-71, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12181747

ABSTRACT

MEK/ERK-mediated signals have recently been found to inhibit Fas-mediated cell death through inhibition of caspase-8 activity. It remains unknown whether MEK/ERK-mediated signals affect ionizing radiation (IR)-induced cell death. Here we demonstrate that MEK/ERK-mediated signals selectively inhibit IR-induced loss of mitochondrial membrane potential (DeltaPsi(m)) and subsequent cell death. In Jurkat cells, TPA strongly activated ERK and inhibited the IR-induced caspase-8/Bid cleavage and the loss of DeltaPsi(m). The inhibitory effect of TPA was mostly abrogated by pretreatment of a specific MEK inhibitor PD98059, indicating that the effect depends upon MEK/ERK-mediated signals. Moreover, BAF-B03 transfectants expressing IL-2 receptor (IL-2R) beta(c) chain lacking the acidic region, which is responsible for MEK/ERK-mediated signals, revealed higher sensitivity to IR than the transfectants expressing wild-type IL-2R. Interestingly, the signals could neither protect the DeltaPsi(m) loss nor cell death in UV-irradiated cells. These data imply that the anti-apoptotic effect of MEK/ERK-mediated signals appears to selectively inhibit the IR-induced cell death through protection of the DeltaPsi(m) loss. Our data enlighten an anti-apoptotic function of MEK/ERK pathway against IR-induced apoptosis, thereby implying its contribution to radioresistance.


Subject(s)
Cell Death/radiation effects , Intracellular Membranes/enzymology , Jurkat Cells/enzymology , Mitochondria/enzymology , Mitogen-Activated Protein Kinase Kinases/radiation effects , Mitogen-Activated Protein Kinases/radiation effects , Protein Serine-Threonine Kinases/radiation effects , BH3 Interacting Domain Death Agonist Protein , Carrier Proteins/drug effects , Carrier Proteins/metabolism , Carrier Proteins/radiation effects , Caspase Inhibitors , Caspases/metabolism , Caspases/radiation effects , Cell Death/drug effects , Cell Death/physiology , Enzyme Inhibitors/pharmacology , Humans , Immunohistochemistry , Interleukin-2/pharmacology , Intracellular Membranes/drug effects , Intracellular Membranes/radiation effects , Jurkat Cells/drug effects , Jurkat Cells/radiation effects , MAP Kinase Kinase 1 , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membrane Potentials/radiation effects , Microscopy, Electron , Mitochondria/drug effects , Mitochondria/radiation effects , Mitogen-Activated Protein Kinase Kinases/drug effects , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/drug effects , Mitogen-Activated Protein Kinases/metabolism , Protein Serine-Threonine Kinases/drug effects , Protein Serine-Threonine Kinases/metabolism , Radiation Tolerance/drug effects , Radiation Tolerance/physiology , Radiation, Ionizing , Tetradecanoylphorbol Acetate/pharmacology , Ultraviolet Rays
7.
Mol Cell Biol ; 22(13): 4522-34, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12052862

ABSTRACT

Retinoids exhibit antineoplastic activities that may be linked to retinoid receptor-mediated transrepression of activating protein 1 (AP1), a heterodimeric transcription factor composed of fos- and jun-related proteins. Here we show that transcriptional activation of an AP1-regulated gene through the mitogen-activated protein kinase (MAPK)-extracellular signal-regulated kinase (ERK) pathway (MAPK(ERK)) is characterized, in intact cells, by a switch from a fra2-junD dimer to a junD-fosB dimer loading on its promoter and by simultaneous recruitment of ERKs, CREB-binding protein (CBP), and RNA polymerase II. All-trans-retinoic acid (atRA) receptor (RAR) was tethered constitutively to the AP1 promoter. AP1 transrepression by retinoic acid was concomitant to glycogen synthase kinase 3 activation, negative regulation of junD hyperphosphorylation, and to decreased RNA polymerase II recruitment. Under these conditions, fra1 loading to the AP1 response element was strongly increased. Importantly, CBP and ERKs were excluded from the promoter in the presence of atRA. AP1 transrepression by retinoids was RAR and ligand dependent, but none of the functions required for RAR-mediated transactivation was necessary for AP1 transrepression. These results indicate that transrepressive effects of retinoids are mediated through a mechanism unrelated to transcriptional activation, involving the RAR-dependent control of transcription factors and cofactor assembly on AP1-regulated promoters.


Subject(s)
MAP Kinase Kinase 4 , Mitogen-Activated Protein Kinases/metabolism , Nuclear Proteins/metabolism , Promoter Regions, Genetic , Receptors, Retinoic Acid/metabolism , Trans-Activators/metabolism , Transcription Factor AP-1/metabolism , Anisomycin/pharmacology , CREB-Binding Protein , Calcium-Calmodulin-Dependent Protein Kinases/drug effects , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , DNA/metabolism , Dimerization , Enzyme Inhibitors/pharmacology , Glycogen Synthase Kinase 3 , Glycogen Synthase Kinases , HeLa Cells/drug effects , HeLa Cells/radiation effects , Humans , MAP Kinase Kinase 1 , Mitogen-Activated Protein Kinase Kinases/drug effects , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinase Kinases/radiation effects , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/genetics , Mutation , Nuclear Proteins/genetics , Nuclear Receptor Co-Repressor 1 , Protein Serine-Threonine Kinases/drug effects , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/radiation effects , Receptors, Retinoic Acid/drug effects , Receptors, Retinoic Acid/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Response Elements , Retinoic Acid Receptor alpha , Retinoid X Receptors , Tetradecanoylphorbol Acetate/pharmacology , Trans-Activators/genetics , Transcription Factor AP-1/drug effects , Transcription Factor AP-1/genetics , Transcription Factors/drug effects , Transcription Factors/genetics , Transcription Factors/metabolism , Tretinoin/pharmacology , Ultraviolet Rays
8.
Pigment Cell Res ; 14(2): 103-9, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11310789

ABSTRACT

UV-induced melanogenesis is a well known physiological response of human skin exposed to solar radiation; however, the signaling molecules involved in the stimulation of melanogenesis in melanocytes following UV exposure remain unclear. In this study we induced melanogenesis in vitro in normal human epidermal melanocytes using a single irradiation with UVA at 1 kJ/m2 and examined the potential involvement of mitogen-activated protein kinases (MAPK) as UVA-responsive signaling molecules in those cells. UVA irradiation did not affect the proliferation of melanocytes, but it did increase tyrosinase mRNA expression, which reached a maximum level 4 hr after UVA irradiation. The amount of tyrosinase protein, as quantitated by immunoblotting, was also increased at 24 hr following UVA irradiation. Among the MAPK examined, extracellular signal-related kinase (ERK) 1/2 was phosphorylated within 15 min of UVA irradiation, but no such phosphorylation was observed for c-Jun N-terminal kinases (JNK) or p38. Accordingly, the activity of ERK1/2 was also increased shortly after UVA irradiation. These responses of ERK1/2 to UVA irradiation were markedly inhibited when cells were pre-treated with N-acetyl-L-cysteine, an antioxidant, or with suramin, a tyrosine kinase receptor inhibitor. The formation of (6-4)photoproducts or cyclobutane pyrimidine dimers was not detected in cellular DNA after UVA irradiation. These findings suggest that a single UVA irradiation-induced melanogenesis is associated with the activation of ERK1/2 by upstream signals that originate from reactive oxygen species or from activated tyrosine kinase receptors, but not from damaged DNA.


Subject(s)
Epidermal Cells , Melanocytes/metabolism , Melanocytes/radiation effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinases/metabolism , Ultraviolet Rays , Acetylcysteine/pharmacology , Cell Survival/radiation effects , Cells, Cultured , DNA Damage/radiation effects , Enzyme Activation/radiation effects , Epidermis/metabolism , Epidermis/radiation effects , Free Radical Scavengers/pharmacology , Humans , Melanins/metabolism , Melanins/radiation effects , Melanocytes/drug effects , Mitogen-Activated Protein Kinase 1/drug effects , Mitogen-Activated Protein Kinase 1/radiation effects , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinase Kinases/radiation effects , Mitogen-Activated Protein Kinases/drug effects , Mitogen-Activated Protein Kinases/radiation effects , Monophenol Monooxygenase/genetics , Monophenol Monooxygenase/metabolism , Monophenol Monooxygenase/radiation effects , Phosphorylation/radiation effects , Reactive Oxygen Species/metabolism
9.
J Neurosci Res ; 62(6): 799-808, 2000 Dec 15.
Article in English | MEDLINE | ID: mdl-11107164

ABSTRACT

The protein serine/threonine kinase Akt is a target of phosphatidylinositol 3-kinase that mediates many of the trophic actions of growth factors on cells. In PC12 cells, complete removal of serum leads to rapid stimulation of the cJun N-terminal kinase (JNK) pathway. Inclusion of insulin-like growth factor-1, a stimulator of Akt in PC12 cells, inhibits JNK activation in this setting, whereas addition of wortmannin to PC12 cells in the presence of serum stimulates JNK activity, suggesting that growth factor-mediated signaling through the phosphatidylinositol 3-kinase/Akt pathway chronically inhibits the JNK pathway in PC12 cells. To explore the possible role of Akt as a negative regulator of JNK activity in PC12 cells, a myristoylated, gain-of-function Akt polypeptide (Myr-Akt) was expressed by retrovirus-mediated gene transfer. Stimulation of JNK activity by serum withdrawal or UV irradiation in PC12 cell clones stably expressing Myr-Akt was inhibited approximately 95% or 50%, respectively, relative to control transfected PC12 cells. Phosphorylation of both JNKs and a proximal activator, MAP kinase kinase 4 (MKK4), in response to UV irradiation was inhibited in Myr-Akt-expressing PC12 cells. Furthermore, transient expression of Myr-Akt strongly inhibited cJun transactivation mediated by MEKK1 or MKK7-JNK3, a gain-of-function MKK7-JNK fusion protein. Interestingly, inhibited JNK activation in the Myr-Akt-expressing PC12 cells is associated with marked induction of JNK-interacting protein-1 (JIP-1). We propose that negative regulation of the JNK pathway through Akt-dependent induction of specfic JIP proteins contributes to the antiapoptotic actions of Akt in neuronal cell types.


Subject(s)
Adaptor Proteins, Signal Transducing , Carrier Proteins/metabolism , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinase Kinases/metabolism , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins c-jun/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Apoptosis/physiology , Chromaffin Cells/metabolism , Insulin-Like Growth Factor I/pharmacology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/radiation effects , Mitogen-Activated Protein Kinase Kinases/drug effects , Mitogen-Activated Protein Kinase Kinases/radiation effects , Neurons/metabolism , PC12 Cells/drug effects , PC12 Cells/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-jun/drug effects , Proto-Oncogene Proteins c-jun/radiation effects , Rats , Signal Transduction/drug effects , Signal Transduction/physiology , Transfection/methods
10.
Oncogene ; 19(39): 4480-90, 2000 Sep 14.
Article in English | MEDLINE | ID: mdl-11002421

ABSTRACT

The activation of cell cycle checkpoints in response to genotoxic stressors is essential for the maintenance of genomic integrity. Although most prior studies of cell cycle effects of UV irradiation have used UVC, this UV range does not penetrate the earth's atmosphere. Thus, we have investigated the mechanisms of ultraviolet B (UVB) irradiation-induced cell cycle arrest in a biologically relevant target cell type, the early stage human melanoma cell line, WM35. Irradiation of WM35 cells with UVB resulted in arrests throughout the cell cycle: at the G1/S transition, in S phase and in G2. G1 arrest was accompanied by increased association of p21 with cyclin E/cdk2 and cyclin A/cdk2, increased binding of p27 to cyclin E/cdk2 and inhibition of these kinases. A loss of Cdc25A expression was associated with an increased inhibitory phosphotyrosine content of cyclin E- and cyclin A-associated cdk2 and may also contribute to G1 arrest following UVB irradiation. The association of Cdc25A with 14-3-3 was increased by UVB. Reduced cyclin D1 protein and increased binding of p21 and p27 to cyclin D1/cdk4 complexes were also observed. The loss of cyclin D1 could not be attributed to inhibition of either MAPK or PI3K/PKB pathways, since both were activated by UVB. Cdc25B levels fell and the remaining protein showed an increased association with 14-3-3 in response to UVB. Losses in cyclin B1 expression and an increased binding of p21 to cyclin B1/cdk1 complexes also contributed to inhibition of this kinase activity, and G2/M arrest. Oncogene (2000) 19, 4480 - 4490.


Subject(s)
Cell Cycle/radiation effects , Melanoma/pathology , Muscle Proteins , Tyrosine 3-Monooxygenase , 14-3-3 Proteins , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinases/metabolism , Cyclin-Dependent Kinases/radiation effects , Cyclins/metabolism , Cyclins/radiation effects , Humans , Melanoma/metabolism , Melanoma/radiotherapy , Microfilament Proteins/metabolism , Microfilament Proteins/radiation effects , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinase Kinases/radiation effects , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/radiation effects , Protein Kinases/metabolism , Protein Kinases/radiation effects , Proteins/metabolism , Proteins/radiation effects , Retinoblastoma Protein/metabolism , Retinoblastoma Protein/radiation effects , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/radiation effects , Ultraviolet Rays , cdc25 Phosphatases/metabolism , cdc25 Phosphatases/radiation effects
11.
Mol Cell Biol ; 20(13): 4543-52, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10848581

ABSTRACT

The p38 group of kinases belongs to the mitogen-activated protein (MAP) kinase superfamily with structural and functional characteristics distinguishable from those of the ERK, JNK (SAPK), and BMK (ERK5) kinases. Although there is a high degree of similarity among members of the p38 group in terms of structure and activation, each member appears to have a unique function. Here we show that activation of p38gamma (also known as ERK6 or SAPK3), but not the other p38 isoforms, is required for gamma-irradiation-induced G(2) arrest. Activation of the MKK6-p38gamma cascade is sufficient to induce G(2) arrest in cells, and expression of dominant negative alleles of MKK6 or p38gamma allows cells to escape the DNA damage-induce G(2) delay. Activation of p38gamma is dependent on ATM and leads to activation of Cds1 (also known as Chk2). These data suggest a model in which activation of ATM by gamma irradiation leads to the activation of MKK6, p38gamma, and Cds1 and that activation of both MKK6 and p38gamma is essential for the proper regulation of the G(2) checkpoint in mammalian cells.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/radiation effects , Cell Cycle/radiation effects , Mitogen-Activated Protein Kinases/metabolism , Ataxia Telangiectasia Mutated Proteins , CDC2 Protein Kinase/drug effects , CDC2 Protein Kinase/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/radiation effects , Cell Death/radiation effects , Checkpoint Kinase 2 , DNA Damage/radiation effects , DNA-Binding Proteins , Enzyme Inhibitors/pharmacology , Fibroblasts/cytology , Fibroblasts/radiation effects , G2 Phase/radiation effects , Gamma Rays , HeLa Cells/radiation effects , Humans , Imidazoles/pharmacology , Isoenzymes , MAP Kinase Kinase 1 , MAP Kinase Kinase 5 , MAP Kinase Kinase 6 , MAP Kinase Kinase 7 , Mitogen-Activated Protein Kinase 7 , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinase Kinases/radiation effects , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/radiation effects , Phosphorylation , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/radiation effects , Pyridines/pharmacology , Signal Transduction , Tumor Suppressor Proteins , cdc25 Phosphatases/metabolism , cdc25 Phosphatases/radiation effects , p38 Mitogen-Activated Protein Kinases
12.
Int J Oncol ; 16(2): 413-22, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10639586

ABSTRACT

The ability of low dose ionizing radiation (2 Gy) to modulate the activities of the mitogen activated protein kinase (MAPK) and c-Jun NH2-terminal kinase (JNK1) cascades in human monocytic leukemia (U937/pREP4) cells and in cells over-expressing dominant negative c-Jun (TAM67) (U937/TAM67) was investigated. Radiation exposure caused prolonged ( approximately 1 h) MAPK activations in U937 cells. In contrast, low dose irradiation weakly modulated JNK1 activity in these cells. Inhibition of the MAPK pathway by use of the specific MEK1/2 inhibitor (10 microM PD98059) in both U937/pREP4 and U937/TAM67 cells prior to radiation exposure permitted strong prolonged radiation-induced activations of JNK1. Expression of TAM67 decreased the ability of radiation to cause apoptosis compared to control transfected cells. However, combined MEK1/2 inhibition and radiation exposure in both cell types caused a large decrease in suspension culture growth and a large increase in apoptosis, when compared to either treatment alone. Reduced proliferation after combined irradiation and PD98059 treatment in both cell types correlated with prolonged cell cycle arrest in G2/M phase. Prolonged growth arrest was abolished when MEK1/2 inhibitor was removed 6 h following irradiation, which was associated with a reduction in apoptosis. The ability of MEK1/2 inhibition to cause prolonged G2/M growth arrest was reduced in U937 cells stably transfected with a p21Cip-1/WAF1 antisense construct (U937/p21AS). This data correlated with an enhancement of radiation-induced apoptosis and a reduced ability of MEK1/2 inhibition to potentiate apoptosis. Collectively our data demonstrate that inhibition of MEK1/2 function increases the radiation sensitivity of U937 cells, independently of c-Jun function, and decreases the ability of these cells to recover from the radiation-induced G2/M cell cycle checkpoint arrest. In addition, our data also demonstrate that the ability of MEK1/2 inhibition to potentiate radiation-induced cell death in U937 cells in part requires an ability of cells to express low levels of p21Cip-1/WAF1.


Subject(s)
Apoptosis/radiation effects , G2 Phase/radiation effects , Mitogen-Activated Protein Kinase Kinases/radiation effects , Mitogen-Activated Protein Kinases/radiation effects , Mitosis/radiation effects , Apoptosis/physiology , Cell Cycle/physiology , Cell Cycle/radiation effects , G2 Phase/physiology , Humans , JNK Mitogen-Activated Protein Kinases , Leukemia, Myeloid/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Mitosis/physiology , U937 Cells/radiation effects
SELECTION OF CITATIONS
SEARCH DETAIL
...