Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
Add more filters

Publication year range
1.
Toxicol Appl Pharmacol ; 290: 116-25, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26529667

ABSTRACT

Depleted uranium (DU) mainly accumulates in the bone over the long term. Osteoblast cells are responsible for the formation of bone, and they are sensitive to DU damage. However, studies investigating methods of reducing DU damage in osteoblasts are rarely reported. Ghrelin is a stomach hormone that stimulates growth hormones released from the hypothalamic-pituitary axis, and it is believed to play an important physiological role in bone metabolism. This study evaluates the impact of ghrelin on DU-induced apoptosis of the osteoblast MC3T3-E1 and investigates its underlying mechanisms. The results show that ghrelin relieved the intracellular oxidative stress induced by DU, eliminated reactive oxygen species (ROS) and reduced lipid peroxidation by increasing intracellular GSH levels; in addition, ghrelin effectively suppressed apoptosis, enhanced mitochondrial membrane potential, and inhibited cytochrome c release and caspase-3 activation after DU exposure. Moreover, ghrelin significantly reduced the expression of DU-induced phosphorylated p38-mitogen-activated protein kinase (MAPK). A specific inhibitor (SB203580) or specific siRNA of p38-MAPK could significantly suppress DU-induced apoptosis and related signals, whereas ROS production was not affected. In addition, ghrelin receptor inhibition could reduce the anti-apoptosis effect of ghrelin on DU and reverse the effect of ghrelin on intracellular ROS and p38-MAPK after DU exposure. These results suggest that ghrelin can suppress DU-induced apoptosis of MC3T3-E1 cells, reduce DU-induced oxidative stress by interacting with its receptor, and inhibit downstream p38-MAPK activation, thereby suppressing the mitochondrial-dependent apoptosis pathway.


Subject(s)
Ghrelin/pharmacology , MAP Kinase Signaling System , Oxidative Stress/drug effects , Protective Agents/pharmacology , Uranium/toxicity , 3T3 Cells/radiation effects , Animals , Apoptosis/drug effects , Caspase 3/genetics , Caspase 3/metabolism , Cell Survival/drug effects , Cell Survival/radiation effects , Lipid Peroxidation/drug effects , Mice , Osteoblasts/drug effects , Osteoblasts/metabolism , Phosphorylation , Reactive Oxygen Species/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
2.
BMC Musculoskelet Disord ; 13: 94, 2012 Jun 08.
Article in English | MEDLINE | ID: mdl-22682502

ABSTRACT

BACKGROUND: It has been indicated that moderate or high dose of X-irradiation could delay fracture union and cause osteoradionecrosis, in part, mediated by its effect on proliferation and differentiation of osteoblasts. However, whether low dose irradiation (LDI) has similar roles on osteoblasts is still unknown. In this study, we investigated whether and to what extent LDI could affect the proliferation, differentiation and mineralization of osteoblasts in vitro. METHODS: The MC3T3-E1 cells were exposed to single dose of X-irradiation with 0, 0.1, 0.5, 1.0 Gy respectively. Cell proliferation, apoptosis, alkaline phosphatase (ALP) activity, and mineralization was evaluated by methylthiazoletetrazolium (MTT) and bromodeoxyuridine (BrdU) assay, flow cytometry, ALP viability kit and von Kossa staining, respectively. Osteocalcin (OCN) and core-binding factor α1 (Cbfα1) expressions were measured by real time-PCR and western blot, respectively. RESULTS: The proliferation of the cells exposed to 2.0 Gy was significantly lower than those exposed to ≤1.0 Gy (p < 0.05) from Day 4 to Day 8, measured by MTT assay and BrdU incorporation. For cells exposed to ≤1.0 Gy, increasing dosages of X-irradiation had no significant effect on cell proliferation and apoptosis. Importantly, LDI of 0.5 and 1 Gy increased ALP activities and mineralized nodules of MC3T3-E1 cells. In addition, mRNA and protein expressions of OCN and Cbfα1 were also markedly increased after treatment with LDI at 0.5 and 1 Gy. CONCLUSIONS: LDI have different effects on proliferation and differentiation of osteoblasts from those of high dose of X-irradiation, which might suggest that LDI could lead to promotion of fracture healing through enhancing the differentiation and mineralization of osteoblasts.


Subject(s)
Calcification, Physiologic/radiation effects , Cell Differentiation/radiation effects , Cell Proliferation/radiation effects , Osteoblasts/radiation effects , 3T3 Cells/metabolism , 3T3 Cells/pathology , 3T3 Cells/radiation effects , Alkaline Phosphatase/metabolism , Animals , Apoptosis/radiation effects , Bromodeoxyuridine/metabolism , Calcium/analysis , Calcium/metabolism , Cell Line , Cell Survival/radiation effects , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Dose-Response Relationship, Radiation , Gene Expression/radiation effects , Mice , Osteoblasts/metabolism , Osteoblasts/pathology , Osteocalcin/genetics , Osteocalcin/metabolism , RNA, Messenger/metabolism , Tetrazolium Salts/metabolism , Thiazoles/metabolism
3.
Cells Tissues Organs ; 191(1): 21-35, 2010.
Article in English | MEDLINE | ID: mdl-19546512

ABSTRACT

The standard method for producing graftable epithelia relies on the presence of a feeder layer of lethally irradiated 3T3-J2 murine fibroblasts (Rheinwald and Green technique). Here, we studied a new keratinocyte culture system, which envisages the utilization of nonirradiated human fibroblasts embedded into a fibrin substrate, in cultures destined for a future clinical application. We tested this culture system using keratinocytes grown on a fibrin gel precoated with 3T3-J2 murine fibroblasts as a control. In order to evaluate the new technology, we compared the clonogenic potential and the proliferative, differentiative and metabolic characteristics of keratinocytes cultured on the fibrin gel under the two culture conditions. The results demonstrated that the proposed technology did not impair the behavior of cultured keratinocytes and revealed that cells maintained their proliferative potential and phenotype under the experimental conditions. In particular, the demonstration of stem cell maintenance under the adopted culture conditions is very important for acute burn treatment with skin substitutes. This work is a first step in the evaluation of a new keratinocyte culture system, which has been studied in order to take advantage of an additional human cell population (i.e. nonirradiated, growing fibroblasts) for future transplantation purposes in acute and chronic wounds. Additional research will allow us to attain (1) the removal of murine cells in the initial phase of keratinocyte cultures, and (2) the removal of other potentially dangerous animal-derived materials from the entire culture system.


Subject(s)
3T3 Cells/cytology , Cell Communication , Cell Culture Techniques , Cell Differentiation , Fibroblasts/cytology , Keratinocytes/cytology , 3T3 Cells/physiology , 3T3 Cells/radiation effects , Animals , Biocompatible Materials , Cell Proliferation , Fibrin , Fibrin Tissue Adhesive , Fibroblasts/physiology , Humans , Keratinocytes/physiology , Mice , Stem Cells/cytology , Stem Cells/physiology , Tissue Engineering
4.
In Vivo ; 24(4): 519-23, 2010.
Article in English | MEDLINE | ID: mdl-20668319

ABSTRACT

Although the advantage of ultraviolet (UV) irradiation of titanium plates for the attachment of osteoblast is known, the details of the experimental conditions have not been described in previous literature. We established optimal conditions of UV irradiation of titanium plate for the adhesion of mouse osteoblast MC3T3-E1 cells. The viable cell number was determined by MTT method. UV irradiation at two different wavelengths (253.7 and 365 nm) enhanced the cell attachment on titanium plate to comparable extents. The optimal UV exposure duration was 20 minutes and prolonged irradiation slightly reduced cell attachment. The attached cells proliferated during 24 hours, accompanied by the enhanced consumption of extracellular glutamine and arginine. The present study supports the previous reports of the efficacy of UV irradiation, and this simple and rapid assay system may be applicable for the study of the interaction of osteoblast and UV-activated titanium plates.


Subject(s)
3T3 Cells/radiation effects , Cell Adhesion/radiation effects , Osteoblasts/radiation effects , Titanium/radiation effects , 3T3 Cells/cytology , Animals , Arginine/metabolism , Cell Division/radiation effects , Glutamine/metabolism , Mice , Osteoblasts/cytology , Surface Properties , Ultraviolet Rays
5.
Oncogene ; 25(6): 940-53, 2006 Feb 09.
Article in English | MEDLINE | ID: mdl-16247471

ABSTRACT

Following genotoxic stress, p53 either rescues a damaged cell or promotes its elimination. The parameters determining a specific outcome of the p53 response are largely unknown. In mouse fibroblasts treated with different irradiation schemes, we monitored transcriptional and non-transcriptional p53 activities and identified determinants that initiate an anti- or a pro-apoptotic p53 response within the context of p53-independent stress signaling. The primary, transcription-mediated p53 response in these cells is anti-apoptotic, while induction of p53-dependent apoptosis requires an additional, transcription-independent p53 activity, provided by high intracellular levels of activated p53. High intracellular levels of p53 were selectively generated after apoptosis-inducing high-dose UV-irradiation, and correlated with a strongly delayed upregulation of Mdm2. Following high-dose UV-irradiation, p53 accumulated in the cytoplasm and led to activation of the pro-apoptotic protein Bax. As p53-dependent Bax-activation is transcription-independent, we postulated that certain transcription-deficient mutant p53 proteins might also exert this activity. Indeed we found an endogenous, transcription-inactive mutant p53 that upon genotoxic stress induced Bax-activation in vivo. Our results demonstrate the impact and in vivo relevance of non-transcriptional mechanisms for wild-type and mutant p53-mediated apoptosis.


Subject(s)
Apoptosis/genetics , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/radiation effects , 3T3 Cells/radiation effects , Animals , Apoptosis/radiation effects , Cell Survival/drug effects , Cell Survival/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p21/radiation effects , Cytosol/metabolism , Dose-Response Relationship, Radiation , Gamma Rays , Intracellular Signaling Peptides and Proteins , Mice , Mutagenicity Tests , Mutation , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/radiation effects , Proto-Oncogene Proteins c-mdm2/metabolism , Proto-Oncogene Proteins c-mdm2/radiation effects , Stress, Physiological , Tumor Suppressor Protein p53/genetics , Ultraviolet Rays , Up-Regulation , bcl-2-Associated X Protein/metabolism , bcl-2-Associated X Protein/radiation effects
6.
Mol Biol Cell ; 13(7): 2502-17, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12134086

ABSTRACT

Caveolae are vesicular invaginations of the plasma membrane. Caveolin-1 is the principal structural component of caveolae in vivo. Several lines of evidence are consistent with the idea that caveolin-1 functions as a "transformation suppressor" protein. In fact, caveolin-1 mRNA and protein expression are lost or reduced during cell transformation by activated oncogenes. Interestingly, the human caveolin-1 gene is localized to a suspected tumor suppressor locus (7q31.1). We have previously demonstrated that overexpression of caveolin-1 arrests mouse embryonic fibroblasts in the G(0)/G(1) phase of the cell cycle through activation of a p53/p21-dependent pathway, indicating a role of caveolin-1 in mediating growth arrest. However, it remains unknown whether overexpression of caveolin-1 promotes cellular senescence in vivo. Here, we demonstrate that mouse embryonic fibroblasts transgenically overexpressing caveolin-1 show: 1) a reduced proliferative lifespan; 2) senescence-like cell morphology; and 3) a senescence-associated increase in beta-galactosidase activity. These results indicate for the first time that the expression of caveolin-1 in vivo is sufficient to promote and maintain the senescent phenotype. Subcytotoxic oxidative stress is known to induce premature senescence in diploid fibroblasts. Interestingly, we show that subcytotoxic level of hydrogen peroxide induces premature senescence in NIH 3T3 cells and increases endogenous caveolin-1 expression. Importantly, quercetin and vitamin E, two antioxidant agents, successfully prevent the premature senescent phenotype and the up-regulation of caveolin-1 induced by hydrogen peroxide. Also, we demonstrate that hydrogen peroxide alone, but not in combination with quercetin, stimulates the caveolin-1 promoter activity. Interestingly, premature senescence induced by hydrogen peroxide is greatly reduced in NIH 3T3 cells harboring antisense caveolin-1. Importantly, induction of premature senescence is recovered when caveolin-1 levels are restored. Taken together, these results clearly indicate a central role for caveolin-1 in promoting cellular senescence and they suggest the hypothesis that premature senescence may represent a tumor suppressor function mediated by caveolin-1 in vivo.


Subject(s)
Caveolae/metabolism , Caveolins/metabolism , Cellular Senescence/physiology , Fibroblasts/physiology , 3T3 Cells/drug effects , 3T3 Cells/radiation effects , Animals , Antioxidants/pharmacology , Apoptosis/physiology , Caveolin 1 , Caveolins/genetics , Cell Division/physiology , Cell Size , Cells, Cultured , Cellular Senescence/drug effects , Fibroblasts/cytology , Genes, Reporter , Humans , Hydrogen Peroxide/pharmacology , Mice , Oxidants/pharmacology , Oxidative Stress , Promoter Regions, Genetic , Quercetin/pharmacology , Time Factors , Transcription, Genetic , Transgenes , Tumor Suppressor Protein p53/metabolism , Ultraviolet Rays , Up-Regulation/physiology , Vitamin E/pharmacology , beta-Galactosidase/metabolism
7.
Mol Biol Cell ; 6(9): 1197-213, 1995 Sep.
Article in English | MEDLINE | ID: mdl-8534916

ABSTRACT

The cyclin-dependent kinase (CDK) inhibitor p27 binds and inhibits the kinase activity of several CDKs. Here we report an analysis of the behavior and partners of p27 in Swiss 3T3 mouse fibroblasts during normal mitotic cell cycle progression, as well as in cells arrested at different stages in the cycle by growth factor deprivation, lovastatin treatment, or ultraviolet (UV) irradiation. We found that the level of p27 is elevated in cells arrested in G0 by growth factor deprivation or contact inhibition. In G0, p27 was predominantly monomeric, although some portion was associated with residual cyclin A.Cdk2. During G1, all of p27 was associated with cyclin D1.Cdk4 and was then redistributed to cyclin A.Cdk2 as cells entered S phase. The loss of the monomeric p27 pool as cyclins accumulate in G1 is consistent with the in vivo and in vitro data showing that p27 binds better to cyclin.CDK complexes than to monomeric CDKs. In growing cells, the majority of p27 was associated with cyclin D1 and the level of p27 was significantly lower than the level of cyclin D1. In cells arrested in G1 with lovastatin, cyclin D1 was degraded and p27 was redistributed to cyclin A.Cdk2. In contrast to p21 (which is a p27-related CDK inhibitor and is induced by UV irradiation), the level of p27 was reduced after UV irradiation, but because cyclin D1 was degraded more rapidly than p27, there was a transient increase in binding of p27 to cyclin A.Cdk2. These data suggest that cyclin D1.Cdk4 acts as a reservoir for p27, and p27 is redistributed from cyclin D1.Cdk4 to cyclin A.Cdk2 complexes during S phase, or when cells are arrested by growth factor deprivation, lovastatin treatment, or UV irradiation. It is likely that a similar principle of redistribution of p27 is used by the cell in other instances of cell cycle arrest.


Subject(s)
3T3 Cells/cytology , Cell Cycle Proteins , Cyclin-Dependent Kinases/antagonists & inhibitors , Lovastatin/pharmacology , Microtubule-Associated Proteins/metabolism , Tumor Suppressor Proteins , Ultraviolet Rays , 3T3 Cells/drug effects , 3T3 Cells/metabolism , 3T3 Cells/radiation effects , Animals , Base Sequence , Cell Compartmentation , Cell Cycle/drug effects , Cell Cycle/radiation effects , Cyclin D1 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclins/metabolism , Interphase/drug effects , Mice , Mitosis , Molecular Sequence Data , Nocodazole/pharmacology , Oncogene Proteins/metabolism
8.
Cancer Res ; 59(11): 2562-5, 1999 Jun 01.
Article in English | MEDLINE | ID: mdl-10363973

ABSTRACT

The p53 gene regulates the cell cycle response to DNA damage, which may allow time for adequate DNA repair. We asked whether p53 could directly increase the repair of defined double-strand breaks (DSBs) by nonhomologous end-joining in gamma-irradiated mouse embryonic fibroblasts with differing p53 status. By using an episomal plasmid reactivation assay, we found that presence of wild-type p53 enhanced rejoining of DSBs with short complementary ends of single-stranded DNA. p53 appeared to be directly involved in this regulation, because rejoining enhancement was dependent on the presence of nonspecific DNA binding activity as mediated by the COOH-terminal domain and was independent of transactivating function. We hypothesize that tumor cells lacking p53 and normal cells with wild-type p53 may use different pathways for repair of radiation-induced DSBs.


Subject(s)
3T3 Cells/radiation effects , DNA Damage , DNA Repair , Tumor Suppressor Protein p53/physiology , 3T3 Cells/metabolism , Animals , Chloramphenicol O-Acetyltransferase/genetics , Genes, Reporter , Mice , Transcriptional Activation , Transfection , Tumor Suppressor Protein p53/genetics
9.
Cancer Res ; 56(2): 246-50, 1996 Jan 15.
Article in English | MEDLINE | ID: mdl-8542574

ABSTRACT

AT cells are extremely sensitive to ionizing radiation. Since the AT gene has homology to phosphatidylinositol 3 kinases (PI 3-kinases), wortmannin, a specific inhibitor of PI 3-kinase, was used to determine if PI 3-kinase activity regulates radiation sensitivity. Human and murine cells exposed to wortmannin alone did not display significant cytotoxicity. Wortmannin in combination with radiation was an effective radiosensitizer of murine NIH-3T3 fibroblasts, with a sensitizer enhancement ratio of 1.8 at 10% survival, and had a similar effect on the human tumor cell lines HeLa, SW480, and MCF-7. Wortmannin inhibited the induction of p53 DNA-binding activity by actinomycin D and radiation and blocked the transcriptional activation of a p53 CAT reporter gene by actinomycin D. Wortmannin radiosensitized both wild-type (NIH-3T3 and MCF-7) and mutant (SW480 and HeLa) p53 cells, indicating that p53 induction was not required for radiosensitization by wortmannin. The results suggest that a wortmannin-sensitive pathway, possibly involving PI 3-kinase activity, may regulate the response of the cells to DNA damage.


Subject(s)
3T3 Cells/drug effects , 3T3 Cells/radiation effects , Androstadienes/pharmacology , DNA Damage , Genes, p53 , Neoplasms/drug therapy , Neoplasms/radiotherapy , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Radiation-Sensitizing Agents/pharmacology , Adenocarcinoma/drug therapy , Adenocarcinoma/radiotherapy , Androstadienes/toxicity , Animals , Base Sequence , Breast Neoplasms/drug therapy , Breast Neoplasms/radiotherapy , Cell Survival/radiation effects , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/radiotherapy , Combined Modality Therapy , DNA, Neoplasm/drug effects , DNA, Neoplasm/metabolism , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , HeLa Cells , Humans , Mice , Molecular Sequence Data , Neoplasms/metabolism , Phosphatidylinositol 3-Kinases , Radiation-Sensitizing Agents/toxicity , Tumor Suppressor Protein p53/biosynthesis , Wortmannin
10.
Cancer Res ; 57(22): 5168-78, 1997 Nov 15.
Article in English | MEDLINE | ID: mdl-9371520

ABSTRACT

DNA damage inactivates cyclin-dependent kinases (CDKs) and arrests the cell cycle. Following DNA damage, the G1-S CDKs are inhibited by a mechanism involving p53-dependent induction of p21Cip1/Waf1; but how the Cdc2 is inhibited is less apparent. We found that the signal generated by the DNA damage checkpoint in G2 was dominant over that from the spindle microtubule-assembly checkpoint, because the high Cdc2 activity present in nocodazole or Taxol-arrested cells was reduced by DNA damage. Phosphorylation of the inhibitory residues in Cdc2, Thr14, and Tyr15 coincided with the inactivation of Cdc2 after DNA damage. Interpretation of this result, however, was not straightforward due to the regulation of Thr14/Tyr15 phosphorylation by feedback loops; hence, their phosphorylation can in principle result merely from the inhibition of Cdc2 activity. Consistent with this, Thr14/Tyr15 phosphorylation was induced when Cdc2 kinase activity was inhibited with butyrolactone-I. Given these complications, we undertook a more critical analysis of the mechanisms that regulate Cdc2 after DNA damage. Caffeine reversed the DNA damage-induced inhibition of Cdc2 by causing dephosphorylation of Cdc2, and this dephosphorylation still occurred even when the Cdc2 feedback loops were blocked with butyrolactone-I. These data suggest that the DNA damage checkpoint in part acts through Thr14/Tyr15 phosphorylation by a mechanism independent of Cdc2 activity, and this phosphorylation can be accentuated by the Cdc2 feedback loops involving Thr14/Tyr15 protein kinases and phosphatases. The kinase activity of the Wee1Hu Tyr15 protein kinase was unaltered after DNA damage, but the phosphatase activity of Cdc25C was reduced. Thus, the decrease in Cdc25C activity may in part account for the DNA damage-induced increase in Thr14/Tyr15 phosphorylation.


Subject(s)
CDC2 Protein Kinase/metabolism , Cyclin A/metabolism , Cyclin B/metabolism , DNA Damage , G2 Phase/drug effects , G2 Phase/radiation effects , Threonine/metabolism , Tyrosine/metabolism , 3T3 Cells/drug effects , 3T3 Cells/radiation effects , Animals , Antineoplastic Agents/pharmacology , Caffeine/pharmacology , Cyclin B1 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , Doxorubicin/pharmacology , Electrophoresis, Polyacrylamide Gel , G1 Phase/drug effects , G1 Phase/radiation effects , Humans , Mice , Mitosis/drug effects , Nocodazole/pharmacology , Paclitaxel/pharmacology , Phosphorylation , Protein Kinases/metabolism , Tumor Cells, Cultured
11.
Cancer Res ; 52(1): 154-62, 1992 Jan 01.
Article in English | MEDLINE | ID: mdl-1530767

ABSTRACT

Mouse embryo fibroblasts (C3H 10T1/2) were exposed to 4 Gy of gamma-rays. The cells yielded 5-8 transformed foci per 10(4) surviving cells. Addition of 100 microM of either eicosapentaenoate or docosahexaenoate to the tissue culture medium reduced the number of transformed foci to 0-1.4. C3H 10T1/2 and NIH 3T3 cells were transfected with plasmid T24 containing the Harvey ras oncogene. C3H 10T1/2 cells yielded 0.85-1.1 foci/ng DNA, while NIH 3T3 cells yielded 0.12-0.14 foci/ng DNA. Foci formation was suppressed 65% in C3H 10T1/2 cells and 93% in NIH 3T3 cells when 100 microM eicosapentaenoate was present in the culture medium. Docosahexaenoate had a similar but somewhat weaker effect. Addition of arachidonate to the medium had little or no effect. Cells grown in the presence of added eicosapentaenoate or docosahexaenoate produced much less prostaglandin E when challenged with calcium ionophore A23187. This is a reflection of changes in arachidonate production or utilization that occur during transformation which are suppressed by the added omega 3 fatty acids. Addition of eicosapentaenoate or docosahexaenoate to the culture medium resulted in extensive remodeling of the molecular species of the four major phospholipid classes that were examined. In its simplest form, omega 3-fatty acid-containing species substantially replaced omega 6-fatty acid-containing species. However, many more subtle changes occurred, and the different phospholipids responded differently to different polyunsaturated fatty acids. A feature of C3H 10T1/2 cells was their preferential accumulation of molecular species of 22-carbon fatty acids such as docosapentaenoate (22:5 omega 3) and docosatetraenoate (22:4 omega 6) in preference to eicosapentaenoate (20:5 omega 3) and eicosapentaenoate (arachidonate, 20:4 omega 6). It is proposed that the protective effect of eicosapentaenoate and docosahexaenoate arises out of the changes in the composition of the fatty acids that are released from one or more phospholipids by the action of phospholipases. The changes consist of a reduced release of arachidonate, the normal substrate of cyclooxygenase and lipoxygenases, and a greatly increased release of eicosapentaenoate and docosahexaenoate, which inhibit one or more of these enzymes, or form oxygenated products which are not as active as the arachidonate-derived products. Other mechanisms are also considered.


Subject(s)
Cell Transformation, Neoplastic/drug effects , Docosahexaenoic Acids/pharmacology , Eicosapentaenoic Acid/pharmacology , Fatty Acids, Omega-3/pharmacology , Fatty Acids, Unsaturated/pharmacology , Genes, ras/genetics , Transfection , 3T3 Cells/chemistry , 3T3 Cells/drug effects , 3T3 Cells/radiation effects , Alprostadil/analogs & derivatives , Alprostadil/biosynthesis , Animals , Arachidonic Acid/metabolism , Cell Transformation, Neoplastic/chemistry , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/radiation effects , Dinoprostone/biosynthesis , Fatty Acids, Omega-6 , Fibroblasts/chemistry , Fibroblasts/drug effects , Fibroblasts/radiation effects , Mice , Phosphatidylcholines/chemistry , Phosphatidylcholines/metabolism , Phosphatidylethanolamines/chemistry , Phosphatidylethanolamines/metabolism , Phosphatidylinositols/chemistry , Phosphatidylinositols/metabolism , Phosphatidylserines/chemistry , Phosphatidylserines/metabolism , Phospholipids/chemistry , Time Factors
12.
Oncogene ; 20(23): 2889-99, 2001 May 24.
Article in English | MEDLINE | ID: mdl-11420701

ABSTRACT

Cyclin-dependent kinase 6(cdk6) is present in randomly proliferating cultures of 3T3 cells but has little detectable enzymatic activity. Significant activity is detected only during a short period in early G1 phase. To examine the possible functions of cdk6 in 3T3 cells, lines stably over-expressing cdk6 were constructed and compared to normal 3T3 cells or cell lines with reduced cdk6 levels due to expression of a dominant-negative form of the protein. Over-expression of cdk6 in cells, which led to high levels of activity even in proliferating cultures, had dramatic effects. Cell lines stably over-expressing wild-type cdk6 had a markedly reduced growth rate compared to parental 3T3 cells or lines expressing a dominant-negative form of cdk6. They also over-produced the p53 and p130 proteins and had increased sensitivity to UV-irradiation. Irradiation resulted in accumulation of the Bax protein and rapid cell death. Levels of p53 and p130 proteins were down-regulated and the growth rate of the cells was increased by introduction of the dominant-negative form of cdk6 into cells over-expressing cdk6, indicating that cdk6 is involved in the overproduction of p53 and p130. The results suggest that cdk6, through regulation of growth-suppressing molecules, may play a role in halting cellular growth when proliferation is inappropriate.


Subject(s)
Cyclin-Dependent Kinases , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-bcl-2 , Tumor Suppressor Protein p53/metabolism , 3T3 Cells/radiation effects , Animals , Cell Division/genetics , Cell Line/radiation effects , Culture Media, Serum-Free , Cyclin-Dependent Kinase 6 , Mice , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/radiation effects , Transfection , Tumor Suppressor Protein p53/genetics , bcl-2-Associated X Protein
13.
Oncogene ; 13(2): 413-8, 1996 Jul 18.
Article in English | MEDLINE | ID: mdl-8710381

ABSTRACT

In response to DNA damage, the transcriptional activity of p53 rises. This has been thought to be due to an increase in the level of p53 protein. By comparing the p53 protein level and its ability to transactivate target genes Waf1/Cip1 and mdm2 in both T22 and NIH3T3 cells irradiated with u.v., a discordance between the p53 protein level and its transcriptional activity was observed. When the cells were irradiated with 10 J/m2 of u.v., there was a substantial increase in expression of Waf1/ Cip1 and mdm2. However, little increase in Waf1/Cip1 and mdm2 expression was observed in T22 and NIH3T3 cells 8 or 9 h after exposure to 50 J/m2 of u.v., although the p53 protein level accumulated to its highest level under these conditions. Interestingly, a significant increase in Waf1/Cip1 expression was seen 24 h after irradiation in NIH3T3 cells, indicating that the inhibition of p53 transcriptional activity is reversible. Discordance between the transcriptional activity of p53 and its protein level was further studied using a cell line expressing the p53 reporter plasmid RGC delta fosLacZ. Using double immunofluorescence staining, the coexpression of p53 and beta-galactosidase from the reporter plasmid in the same cells was investigated. The observed lack of correlation between the elevated p53 and beta-galactosidase and expression in u.v. irradiated cells strongly indicates that the ability of p53 to transactivate its target genes is not simply correlated to its protein level. The results indicate that the transcriptional activity of p53 may be negatively regulated.


Subject(s)
Nuclear Proteins , Transcription, Genetic/radiation effects , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/radiation effects , Ultraviolet Rays , 3T3 Cells/metabolism , 3T3 Cells/radiation effects , Animals , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/biosynthesis , Cyclins/genetics , Gene Expression Regulation/radiation effects , Lac Operon , Male , Mice , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-mdm2 , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcriptional Activation , Tumor Suppressor Protein p53/biosynthesis
14.
Oncogene ; 16(4): 459-69, 1998 Jan 29.
Article in English | MEDLINE | ID: mdl-9484835

ABSTRACT

Transcriptional activation and stabilization of p53 is a major response of mammalian cells to U.V.-light induced genetic damages, and possibly responsible for cell damage control. We have studied here by gel mobility shift and immunoblotting assays the activation and accumulation of p53 by U.V.C. and its dependency on cell cycle, protein synthesis and protein phosphorylation. In G0/G1 synchronized cells U.V.C.-induced p53 DNA-binding activity, but not its accumulation, whereas both events took place in G1/S and S-phase cells. The kinetics of p53 activation by U.V.C. were slow requiring at least 1 h and slowly increasing thereafter with full activation observed at 6 h. Treatment of cells with cycloheximide (CHX) prevented the activation of p53 in all phases of the cell cycle and its accumulation in G1/S and S. However, removing CHX-block allowed full activation and accumulation of p53 with fast kinetics even if 4 h had lapsed since the initial U.V.C. insult. This suggests that the protein synthesis-dependent signal initiating p53 activation by U.V.C. remains continuous in the cells. The requirement of protein phosphorylation as mediator of p53 activation by U.V.C. was studied by using chemical protein kinase inhibitors. Of the tested inhibitors, only staurosporine, a known inhibitor of protein kinase C (PKC) and various other kinases, inhibited both p53 activation and accumulation, whereas specific PKC inhibitors, tyrosine kinase inhibitors and a serine/threonine kinase inhibitor did not. PKC-mediation of the p53 U.V.-response was further ruled out by the reactivity of the activated p53 to C-terminal antibody PAb 421. Kinetic studies showed that staurosporine-mediated inhibition of p53 function is an early event in cell damage response. Thus dual, kinetically different events, de novo protein synthesis and staurosporine-inhibited protein phosphorylation are required for p53 activation and accumulation in all phases of the cell cycle. Notably, in the absence of U.V.-induced accumulation in G0/G1 cells, p53 activation is still subject to inhibition of protein synthesis.


Subject(s)
DNA/metabolism , Tumor Suppressor Protein p53/radiation effects , 3T3 Cells/drug effects , 3T3 Cells/radiation effects , Animals , Antibodies/metabolism , Cycloheximide/pharmacology , DNA Replication/drug effects , Enzyme Inhibitors/pharmacology , G1 Phase/genetics , G1 Phase/physiology , Gene Expression Regulation/radiation effects , Genes, p53/radiation effects , Mice , Protein Synthesis Inhibitors/pharmacology , Resting Phase, Cell Cycle/genetics , Resting Phase, Cell Cycle/physiology , S Phase/genetics , S Phase/physiology , Staurosporine/pharmacology , Time Factors , Tumor Suppressor Protein p53/drug effects , Tumor Suppressor Protein p53/immunology , Tumor Suppressor Protein p53/metabolism
15.
Oncogene ; 17(9): 1089-96, 1998 Sep 03.
Article in English | MEDLINE | ID: mdl-9764819

ABSTRACT

p53 tumor suppressor gene controls cell response to a variety of stresses inducing growth arrest or apoptosis in damaged cells. It largely determines the sensitivity of tumor and normal cells to radiation and chemotherapy, and, therefore, defines both the efficacy and limitations of anti-cancer treatment. To determine molecular mechanisms of p53-dependent stress response in normal tissues we identified and compared the spectra of radiation-responsive genes in cells of different origin and p53 status using a cDNA array hybridization technique. The majority of genes identified were p53-dependent and cell type specific. Several of the new p53 responders encode known secreted growth inhibitory factors. This suggests that p53, in addition to its intrinsic antiproliferation activity, can cause 'bystander effect' by inducing export of growth suppressive stimuli from damaged cells to neighboring cells. Consistently, a p53-dependent accumulation of factors, which causes growth inhibitory effects in a variety of cell lines, was found after gamma irradiation in the media from established and primary cell cultures and in the urine of irradiated mice. Moreover, p53-dependent factors released by normal human fibroblasts potentiated the cytotoxic effect of a chemotherapeutic drug on co-cultivated tumor cells. This suggests a previously unknown role for normal cells in chemo- and radiation therapy of cancer.


Subject(s)
Growth Inhibitors/metabolism , Stress, Physiological/physiopathology , 3T3 Cells/cytology , 3T3 Cells/metabolism , 3T3 Cells/radiation effects , Animals , Antineoplastic Agents/pharmacology , Cell Division/drug effects , Cell Division/genetics , Cell Division/radiation effects , Cell Line , Gamma Rays , Gene Expression/drug effects , Gene Expression/genetics , Gene Expression/radiation effects , Gene Expression Regulation/radiation effects , Gene Expression Regulation, Neoplastic/radiation effects , Genes/genetics , Genes/radiation effects , Genes, Tumor Suppressor/genetics , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/metabolism , Tumor Cells, Cultured/radiation effects , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/physiology
16.
Oncogene ; 13(3): 625-32, 1996 Aug 01.
Article in English | MEDLINE | ID: mdl-8760304

ABSTRACT

N3T3 and P-3T3 cells, originally isolated from a NIH3T3 cell clone on the basis of their negative and positive transformation by v-Abl, v-Src and Bcr-Abl, were previously found to show distinct cyclin activity changes following 12-O-tetradecanoylphorbol-13-acetate (TPA) treatment, which is anti-mitogenic for N-3T3 cells and mitogenic for P-3T3 cells. We have found in this study that, while the G1/S arrest and cell death induced by serum starvation and TPA treatment in N-3T3 cells did not involve p53-mediated checkpoint or apoptosis, N-3T3 and P-3T3 cells evidently responded differently in these aspects of cell cycle regulation to DNA-damaging agents, methylmethane sulfonate (MMS) and gamma-radiation. In N-3T3 cells, DNA damages elicit cell growth arrest at G1/S transition with concomitant accumulation of p53 and p53-inducible Waf1/Cip1 proteins and also signs of apoptosis such as DNA ladder patterns and apoptotic (subgenomic) peak in flow cytograph. Conversely, P-3T3 cells treated with the DNA-damaging agents showed no cell cycle interruption nor accumulation of p53 or Waf1/Cip1. However, both P-3T3 and N-3T3 cells showed the same p53 protein half-life of 40 min or less, the same wild-type p53 DNA sequence and the same co-immunoprecipitable cellular proteins in complexes with p53, suggesting that an alteration in a signal transduction pathway upstream of p53 might account for the evasion of p53-mediated G1 checkpoint in P-3T3 cells.


Subject(s)
DNA Damage , G1 Phase/physiology , S Phase/physiology , Tumor Suppressor Protein p53/physiology , 3T3 Cells/cytology , 3T3 Cells/drug effects , 3T3 Cells/radiation effects , Animals , Apoptosis/drug effects , Apoptosis/physiology , Base Sequence , Cell Division/drug effects , Cell Division/physiology , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/biosynthesis , DNA/drug effects , DNA/radiation effects , G1 Phase/drug effects , G1 Phase/radiation effects , Genes, p53 , Methyl Methanesulfonate/pharmacology , Mice , Molecular Sequence Data , S Phase/drug effects , S Phase/radiation effects , Tetradecanoylphorbol Acetate/pharmacology
17.
Oncogene ; 19(9): 1206-14, 2000 Feb 24.
Article in English | MEDLINE | ID: mdl-10713709

ABSTRACT

The phosphoaminothiol WR1065, the active metabolite of the pro-drug amifostine (WR2721), protects cultured cells and tissues against cytotoxic exposure to radiation or chemotherapeutic agents. We show here that WR1065 and the pro-drug WR2721 activate the p53 tumor suppressor protein and induce the expression of the cyclin-dependent kinase inhibitor p21waf-1 in the breast cancer cell line MCF-7, and in the mouse fibroblast cell line balb/c 3T3. Using two MCF-7 derived cell lines, MN1 and MDD2, we show that induction of p21waf-1 is detectable in MN1 (expressing a functional p53) but not in MDD2 (p53 disabled). These effects are observed at concentrations of WR1065 (0.5 to 1 mM) identical to those required to protect against cytotoxicity by hydrogen peroxide. Induction of p53 is not prevented by addition of aminoguanidine, an inhibitor of Cu-dependent amine-oxidases which blocks the extra-cellular degradation of WR1065 into toxic metabolites. Moreover, spermidine, a natural polyamine structurally related to amifostine, does not activate p53. Induction of p53 by WR1065 results in a delay in the G1/S transition in MCF-7 and MN-1 cells, but not in the p53 disabled cells MDD2. These data indicate that WR1065, a polyamine analog with thiol anti-oxidant properties, activates a cell cycle check-point involving p53.


Subject(s)
Cell Cycle/drug effects , Cyclins/metabolism , Down-Regulation/drug effects , Mercaptoethylamines/pharmacology , Radiation-Protective Agents/pharmacology , Tumor Suppressor Protein p53/physiology , 3T3 Cells/drug effects , 3T3 Cells/radiation effects , Animals , Cell Cycle/radiation effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cyclin-Dependent Kinase Inhibitor p21 , DNA/metabolism , Gamma Rays , Humans , Mice , Mice, Inbred BALB C , Protein Binding/drug effects , Signal Transduction/drug effects , Signal Transduction/radiation effects , Spermidine/pharmacology , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/radiation effects , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/radiation effects
18.
Oncogene ; 19(37): 4221-9, 2000 Aug 31.
Article in English | MEDLINE | ID: mdl-10980595

ABSTRACT

Ribosomal S6 kinases (RSKs) are serine/threonine kinases activated by mitogenic signals through the Mitogen-Activated Protein Kinases/Extracellular Signal-Regulated Kinases (MAPK/ERK). RSKs contain two heterologous complete protein kinase domains. Phosphorylation by ERK of the C-terminal kinase domain allows activation of the N-terminal kinase domain, which mediates substrate phosphorylation. In human, there are three isoforms of RSK (RSK1, RSK2, RSK3), whose functional specificity remains undefined. Importantly, we have shown that mutations in the RSK2 gene lead to the Coffin-Lowry syndrome (CLS). In this study, we characterize two monoclonal antibodies raised against phosphorylated forms of the N- and C-terminal domain of RSK2 (P-S227 and P-T577, respectively). Using these two antibodies, we show that stress signals, such as UV light, induce phosphorylation and activation of the three RSKs to an extent which is comparable to Epidermal Growth Factor (EGF)-mediated activation. The use of specific kinase inhibitors indicates that UV-induced phosphorylation and activation of RSK2 is mediated by the MAPK/ERK pathway, but that the Stress-Activated Protein Kinase 2 (SAPK2)/p38 pathway is also involved. These results modify the view of RSKs as kinases restricted to the mitogenic response and reveal a previously unappreciated role of MAPKs in stress induced signaling. Oncogene (2000) 19, 4221 - 4229


Subject(s)
Isoenzymes/radiation effects , MAP Kinase Signaling System/radiation effects , Protein Processing, Post-Translational/radiation effects , Ribosomal Protein S6 Kinases/radiation effects , Stress, Physiological/physiopathology , Ultraviolet Rays , 3T3 Cells/enzymology , 3T3 Cells/radiation effects , Amino Acid Sequence , Animals , Antibodies, Monoclonal/immunology , COS Cells/enzymology , COS Cells/radiation effects , Cells, Cultured , Enzyme Inhibitors/pharmacology , Epidermal Growth Factor/pharmacology , Fibroblasts/enzymology , Fibroblasts/radiation effects , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , MAP Kinase Signaling System/physiology , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Mitogen-Activated Protein Kinases/physiology , Molecular Sequence Data , Phosphorylation/radiation effects , Protein Structure, Tertiary , Ribosomal Protein S6 Kinases/antagonists & inhibitors , Ribosomal Protein S6 Kinases/genetics , Ribosomal Protein S6 Kinases/immunology , Ribosomal Protein S6 Kinases/metabolism , Tetradecanoylphorbol Acetate/pharmacology , p38 Mitogen-Activated Protein Kinases
19.
Biochim Biophys Acta ; 1496(2-3): 207-20, 2000 Apr 17.
Article in English | MEDLINE | ID: mdl-10771089

ABSTRACT

Expression of the cyclin kinase inhibitor, p21, is regulated both transcriptionally and posttranscriptionally by the ubiquitin-proteasome degradation pathway. Recently, we reported that DNA damage is required for efficient p21 expression by demonstrating that enhanced p21 mRNA expression induced by DNA damage results in increased p21 protein, but enhanced p21 mRNA without DNA damage does not. In addition, we demonstrated that DNA damage suppressed the ubiquitination of p21. In this study, we analyze the link between p21 stabilization and DNA damage. Enhanced p21 protein expression in ML-1 cells resulting from 15 Gy gamma-irradiation was diminished by Wortmannin or LY294002 pretreatment of cells. However, the levels of p21 mRNA were not affected by inhibitor pretreatment. Wortmannin or LY294002 pretreatment reduces p53 expression after gamma-irradiation to a lesser degree than that of p21. In addition, we examined the involvement of DNA-PK, whose activity is inhibited by Wortmannin or LY294002, in p21 stabilization using the SCID fibroblast cell line and a DNA-PK targeting ML-1 cell line. Accumulation of p21 protein by gamma-irradiation was similar to that of DNA-PK intact cells and was reduced by Wortmannin or LY294002 pretreatment. Involvement of another DNA damage detecting enzyme, the ATM gene product, whose activity is also inhibited by Wortmannin or LY294002, was evaluated. ATM deficient cells induced p21 after gamma-irradiation, gamma-irradiation-induced p21 protein was diminished by pretreatment of cells with Wortmannin or LY294002. We conclude that the p21 stabilization mechanism functions after gamma-irradiation, was sensitive to Wortmannin or LY294002, and required neither DNA-PK nor ATM gene product for activity.


Subject(s)
Androstadienes/pharmacology , Chromones/pharmacology , Cyclins/biosynthesis , Morpholines/pharmacology , Phosphoinositide-3 Kinase Inhibitors , 3T3 Cells/radiation effects , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , DNA Damage , DNA-Binding Proteins , Dose-Response Relationship, Drug , Down-Regulation , Enzyme Inhibitors/pharmacology , Gamma Rays , Gene Expression Regulation/drug effects , Gene Expression Regulation/radiation effects , Gene Targeting , Humans , Mice , Protein Serine-Threonine Kinases/analysis , Tumor Cells, Cultured/radiation effects , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Proteins , Wortmannin
20.
Biochim Biophys Acta ; 1593(2-3): 219-29, 2003 Feb 17.
Article in English | MEDLINE | ID: mdl-12581866

ABSTRACT

Protein kinase C (PKC) isoforms play distinct roles in cellular functions. We have previously shown that ionizing radiation activates PKC isoforms (alpha, delta, epsilon, and zeta), however, isoform-specific sensitivities to radiation and its exact mechanisms in radiation mediated signal transduction are not fully understood. In this study, we showed that overexpression of PKC isoforms (alpha, delta, epsilon, and zeta) increased radiation-induced cell death in NIH3T3 cells and PKC epsilon overexpression was predominantly responsible. In addition, PKC epsilon overexpression increased ERK1/2 activation without altering other MAP-kinases such as p38 MAPK or JNK. Co-transfection of dominant negative PKC epsilon (PKC epsilon -KR) blocked both PKC epsilon -mediated ERK1/2 activation and radiation-induced cell death, while catalytically active PKC epsilon construction augmented these phenomena. When the PKC epsilon overexpressed cells were pretreated with PD98059, MEK inhibitor, radiation-induced cell death was inhibited. Co-transfection of the cells with a mutant of ERK1 or -2 (ERK1-KR or ERK2-KR) also blocked these phenomena, and co-transfection with dominant negative Ras or Raf cDNA revealed that PKC epsilon -mediated ERK1/2 activation was Ras-Raf-dependent. In conclusion, PKC epsilon -mediated ERK1/2 activation was responsible for the radiation-induced cell death.


Subject(s)
3T3 Cells/radiation effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinases/metabolism , Protein Kinase C/metabolism , 3T3 Cells/metabolism , Animals , Cell Death/drug effects , Enzyme Activation , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Mice , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/genetics , Protein Kinase C/biosynthesis , Protein Kinase C/genetics , Protein Kinase C-epsilon , Proto-Oncogene Proteins c-raf/metabolism , Transfection , ras Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL