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










Publication year range
1.
Carcinogenesis ; 22(1): 161-9, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11159755

ABSTRACT

The tumor suppressor protein p53 plays an important role in recognition of DNA damage and induction of subsequent cell cycle arrest. One of its target genes encodes the protein p21(WAF1), which is involved in mediation of growth arrest after DNA damage has occurred. Dibenzo[a,l]pyrene (DB[a,l]P) is a polycyclic aromatic hydrocarbon which is an exceptionally potent carcinogen. A reactive secondary metabolite of DB[a,l]P, the fjord region (-)-anti-11R,12S-dihydrodiol 13R,14S-epoxide [(-)-anti-DB[a,l]PDE] was used to investigate DNA damage via adduct formation and cell cycle arrest in human diploid fibroblast cell cultures (HDF). Synchronous HDF were exposed to increasing concentrations (0.014, 0.028 and 0.07 microM) of (-)-anti-DB[a,l]PDE and at 1, 12, 24 and 42 h after treatment cell pellets were analyzed for DNA adduct formation and cell cycle arrest. Exposure of HDF to 0.07 microM (-)-anti-DB[a,l]PDE caused a total DNA binding level of 113 pmol adducts/mg DNA (42 h after treatment). G(1) arrest was induced by this treatment, with 91% of the cells remaining in G(1) phase compared with the solvent-treated control cultures (50%) as analyzed by propidium iodide staining and flow cytometry. Further investigation of the percentage of cells in S phase by 5-bromo-2'-deoxyuridine incorporation confirmed the G(1) arrest in HDF treated with 0.07 microM (-)-anti-DB[a,l]PDE, with only 1.5% of the cells moving into S phase compared with 39% in the control 42 h after treatment. Induction of p53 and p21(WAF1) was demonstrated by western blot analysis.


Subject(s)
Benzopyrenes/toxicity , Cell Cycle/drug effects , DNA Adducts/biosynthesis , DNA/metabolism , Epoxy Compounds/toxicity , Fibroblasts/drug effects , Benzopyrenes/metabolism , Blotting, Western , Cell Line , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , Cyclins/metabolism , DNA/drug effects , DNA Damage , Diploidy , Epoxy Compounds/metabolism , Fibroblasts/metabolism , Fibroblasts/physiology , Genes, p53/drug effects , Humans , Signal Transduction/drug effects , Stereoisomerism , Tumor Suppressor Protein p53/metabolism
2.
J Biol Chem ; 275(46): 35778-85, 2000 Nov 17.
Article in English | MEDLINE | ID: mdl-10958792

ABSTRACT

The p53 tumor suppressor protein is a transcription factor that plays a major role in the DNA damage response. After DNA damage, p53 levels increase due primarily to stabilization of the protein. The molecular mechanisms leading to stabilization of p53 after DNA damage have not been completely elucidated. Recently we reported that cisplatin treatment activated extracellular signal-regulated kinase 1 and 2 (ERK1/2) and that inhibition of ERK1/2 resulted in enhanced sensitivity to cisplatin. In the present study, we examined the potential role of ERK1/2 activation in regulation of the p53 response to cisplatin. In the ovarian carcinoma cell line A2780, inhibition of ERK1/2 activation with the mitogen-activated protein kinase/ERK kinase 1 (MEK1) inhibitor PD98059 resulted in decreased p53 protein half-life and diminished accumulation of p53 protein during exposure to cisplatin. We also demonstrated that p53 protein co-immunoprecipitated with ERK1/2 protein and was phosphorylated by activated recombinant murine ERK2 in vitro. Furthermore, PD98059 decreased the phosphorylation of p53 at serine 15 during cisplatin exposure, suggesting that ERK1/2 mediates in part phosphorylation of p53 during the cisplatin DNA response. These results strongly suggest that cisplatin-induced ERK activation is an up-stream regulator of the p53 response to DNA damage caused by cisplatin.


Subject(s)
Cisplatin/pharmacology , DNA Damage/drug effects , Mitogen-Activated Protein Kinases/metabolism , Nuclear Proteins , Tumor Suppressor Protein p53/metabolism , Androstadienes/pharmacology , Animals , Caffeine/pharmacology , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Flavonoids/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Half-Life , Humans , Imidazoles/pharmacology , Mice , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Phosphorylation/drug effects , Phosphoserine/metabolism , Precipitin Tests , Protein Binding , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-mdm2 , Pyridines/pharmacology , Tumor Cells, Cultured , Wortmannin
3.
Mol Carcinog ; 28(2): 102-10, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10900467

ABSTRACT

Apigenin, a common dietary flavonoid, has been shown to induce cell cycle arrest in both epidermal and fibroblast cells and inhibit skin tumorigenesis in murine models. The present study assessed the influence of apigenin on cell growth and the cell cycle in the human colon carcinoma cell lines SW480, HT-29, and Caco-2. Treatment of each cell line with apigenin (0-80 microM) resulted in a dose-dependent reduction in both cell number and cellular protein content, compared with untreated control cultures. DNA flow cytometric analysis indicated that treatment with apigenin resulted in G2/M arrest in all three cell lines in a time- and dose-dependent manner. Apigenin treatment (80 microM) for 48 h produced maximum G2/M arrest of 64%, 42%, and 26% in SW480 cells, HT-29 cells, and Caco-2 cells, respectively, in comparison with control cells (15%). The proportion of S-phase cells was not altered by apigenin treatment in each of the three cell lines. The G2/M arrest was reversible after 48 h of apigenin treatment in the most sensitive cell line SW480. The degree of G2/M arrest by apigenin was inversely correlated with the corresponding inhibition of cell growth measurements in all three cell lines (r = -0.626 to -0.917, P

Subject(s)
Cell Division/drug effects , Colonic Neoplasms/pathology , Flavonoids/pharmacology , G2 Phase/drug effects , Mitosis/drug effects , Apigenin , CDC2 Protein Kinase/metabolism , Colonic Neoplasms/metabolism , Cyclin B/metabolism , Cyclin B1 , Humans , Tumor Cells, Cultured
4.
Kidney Int ; 57(4): 1460-71, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10760082

ABSTRACT

BACKGROUND: : Cellular proliferation is a key factor in the enlargement of renal cysts in autosomal dominant polycystic kidney disease (ADPKD). We determined the extent to which adenosine 3':5'-cyclic monophosphate (cAMP) may regulate the in vitro proliferation of cyst epithelial cells derived from human ADPKD cysts. METHODS: : Epithelial cells from cysts of individuals with ADPKD and from normal human kidney cortex (HKC) of individuals without ADPKD were cultured. The effects of agonists and inhibitors on the rate of cellular proliferation and the activation of extracellular signal-regulated kinase (ERK1/2) were determined. RESULTS: : 8-Br-cAMP (100 micromol/L) stimulated the proliferation of cells from eight different ADPKD subjects to 99.0% above baseline; proliferation was inhibited by protein kinase A (PKA) antagonists H-89 (97%) and Rp-cAMP (90%). Forskolin (10 micromol/L), which activates adenylyl cyclase, increased proliferation 124%, and receptor-mediated agonists arginine vasopressin, desmopressin, secretin, vasoactive intestinal polypeptide, and prostaglandin E2 stimulated proliferation 54.2, 56.3, 46.7, 37.1, and 48.3%, respectively. The mitogen extracellular kinase (MEK) inhibitor PD98059 completely inhibited ADPKD cell proliferation in response to cAMP agonists, but genistein, a receptor tyrosine kinase inhibitor, did not block cAMP-dependent proliferation. cAMP agonists increased the activity of ERK above control levels within five minutes. In contrast to ADPKD, proliferation and ERK activity of cells derived from normal HKC were not stimulated by cAMP agonists, although electrogenic Cl- secretion was increased by these agonists in both ADPKD and HKC cell monolayers. CONCLUSIONS: : We conclude that cAMP agonists stimulate the proliferation of ADPKD but not HKC epithelial cells through PKA activation of the ERK pathway at a locus distal to receptor tyrosine kinase. We suggest that the adenylyl cyclase signaling pathway may have a unique role in determining the rate of cyst enlargement in ADPKD through its actions to stimulate cellular proliferation and transepithelial solute and fluid secretion.


Subject(s)
Cyclic AMP/pharmacology , Kidney/pathology , Mitogen-Activated Protein Kinases/metabolism , Polycystic Kidney, Autosomal Dominant/pathology , Cell Division/drug effects , Cells, Cultured , Electric Impedance , Enzyme Activation/physiology , Epithelial Cells/enzymology , Epithelial Cells/pathology , Epithelial Cells/physiology , Humans , Kidney/enzymology , Kidney/physiopathology , Kidney Cortex/cytology , Kidney Cortex/physiology , Polycystic Kidney, Autosomal Dominant/enzymology , Polycystic Kidney, Autosomal Dominant/physiopathology
5.
Carcinogenesis ; 21(4): 633-9, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10753197

ABSTRACT

Apigenin, a naturally occurring, non-mutagenic flavonoid, has been shown to inhibit UV-induced skin tumorigenesis in mice when topically applied. In this report we have used the mouse keratinocyte 308 cell line, which contains a wild-type p53 gene, to study the effect of apigenin treatment on p53 protein levels and the expression of its downstream partner, p21/waf1. Cells were treated with 70 microM apigenin for various times and levels of p53 and p21/waf1 protein were assessed by western blot analysis. The level of p53 protein was induced 27-fold after 4 h of apigenin treatment and levels remained elevated through 10 h of exposure. After 24 h of exposure to 70 microM apigenin, p53 protein levels returned to control levels. p21/waf1 protein levels increased approximately 1. 5-2-fold after 4 h and remained elevated at 24 h. To investigate the mechanism of p53 protein accumulation, we compared the half-life of p53 protein in vehicle- and apigenin-treated cells. Cells were incubated for 4 h in the presence of apigenin, then cycloheximide was added to inhibit further protein synthesis and p53 protein levels were measured by western blot. The half-life of p53 protein was found to be increased an average of 8-fold in apigenin-treated cells compared with vehicle-treated cells (t(1/2) = 131 min versus 16 min in apigenin- versus vehicle-treated cells, respectively). The mechanism of p53 protein stabilization is currently being investigated. To determine whether p53 was transcriptionally active, we also performed gel mobility shift assays and transient transfection studies using a luciferase plasmid under the control of the p21/waf1 promoter. Both p53 DNA-binding activity and transcriptional activation peaked after 24 h of exposure to apigenin. These studies suggest that apigenin may exert anti-tumorigenic activity by stimulating the p53-p21/waf1 response pathway.


Subject(s)
Anticarcinogenic Agents/pharmacology , Flavonoids/pharmacology , Trans-Activators/physiology , Tumor Suppressor Protein p53/physiology , Animals , Apigenin , Cell Cycle/drug effects , Cell Line , DNA/metabolism , Keratinocytes/drug effects , Keratinocytes/metabolism , Mice , Mice, Inbred BALB C , Phosphorylation , RNA, Messenger/analysis , Tumor Suppressor Protein p53/chemistry
6.
Mol Carcinog ; 29(4): 219-28, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11170260

ABSTRACT

The cellular response to cisplatin involves activation of multiple signal transduction pathways, including the mitogen-activated protein (MAP) kinase pathways. In this study, we compared the cisplatin-induced activation of two MAP kinases, c-jun N-terminal kinase 1 (JNK1) and extracellular signal-regulated protein kinases 1 and 2 (ERK1/2), in the cisplatin-sensitive ovarian carcinoma cell line A2780 and its derivative cisplatin-resistant cell lines CP70 and C200. Dose-dependent and time-dependent activation of JNK1 and ERK1/2 occurred in each of the three cell lines in response to cisplatin treatment. The requirement of higher concentrations of cisplatin for induction of maximum activation of JNK1 and ERK1/2 was correlated with increased levels of cisplatin resistance. In addition, inhibition of cisplatin-induced ERK activation, using the MAP/ERK kinase 1 synthetic inhibitor PD98059, resulted in enhanced sensitivity to cisplatin in all three cell lines. These results suggest that cisplatin-induced ERK1/2 activity is not responsible for the acquired cisplatin resistance in CP70 and C200 cells but rather provides a general cytoprotective effect in both cisplatin-sensitive and cisplatin-resistant cell lines. In conclusion, different patterns of cisplatin-induced JNK1 and ERK1/2 activation are observed in cell lines with different levels of cisplatin sensitivity, and inhibition of cisplatin-induced ERK1/2 activation enhances sensitivity to cisplatin in both cisplatin-sensitive and cisplatin-resistant cell lines.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Mitogen-Activated Protein Kinases/drug effects , Ovarian Neoplasms/enzymology , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Female , Flavonoids/pharmacology , Humans , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinase 1/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Time Factors , Tumor Cells, Cultured
7.
Mol Carcinog ; 25(4): 262-72, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10449033

ABSTRACT

Exposure of mammalian cells to ultraviolet (UV) light and other DNA-damaging agents triggers the UV response which is characterized by induction of a large number of genes including c-fos, c-jun, and the genes for DNA repair enzymes and cell-cycle regulatory proteins such as p21 WAF1 and p53. Upon DNA damage, the p53 tumor suppressor protein transmits signals to restrict cell-cycle progression, thereby allowing time for DNA repair to occur. Cells also respond to genotoxic stress by activation of the jun N-terminal kinase (JNK)/stress-activated protein kinase pathway. In this report we investigated the effects of modulation of the level of wild-type and mutant p53 protein on basal and UV-inducible JNK activity. We used the A1-5 rat fibroblast cell line, which contains a p53 gene coding for a temperature-sensitive p53 protein, which allows us to regulate the relative level of wild-type and mutant p53 protein produced in a cell. We measured the relative levels of JNK activity in sham-irradiated and UV-irradiated cells by using the immune complex kinase assay and then computed the fold induction of JNK after UV exposure. We demonstrated that cells expressing p53 protein in the wild-type conformation (when grown at 32 degrees C) exhibited a very low level of JNK activity that was induced 14- to 16-fold by UVC irradiation. When cells were grown at 37 degrees C or 39 degrees C to express predominantly mutant p53 protein, basal JNK activity was significantly higher than at 32 degrees C. UVC irradiation of cells expressing mutant p53 protein resulted in JNK activation, although the overall fold-induction was only two-fold because JNK1 activity was already high in the sham-treated controls. UVB irradiation also induced JNK1 activity, although we again observed a relatively high level of basal JNK activity in sham-irradiated cells expressing mutant p53 protein compared with cells expressing wild-type p53. Control experiments confirmed that JNK1 basal activity was not affected by temperature alone. Western blot analysis of cell extracts indicated that expression of p21 WAF protein was significantly higher in cells expressing wild-type p53 protein and was associated with low basal levels of JNK1 activity. In contrast, cells expressing mutant p53 protein and very low levels of p21 WAF1 protein were found to have a higher level of basal JNK1 activity. We also observed a reduced ability to induce JNK1 after UV irradiation of several other cell lines with p53-mutant or p53-null genotypes. Our results provide evidence for a novel connection between p53 status and the basal level of JNK1, a critical enzyme in the stress-activated protein kinase family. In addition, these studies suggest that the presence of mutant p53 protein in a cell not only affects basal activity of JNK1 but also affects the ability of a cell to respond to UV-induced stress by transmitting signals via induction or activation of the JNK1 cascade.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Genes, p53 , Mitogen-Activated Protein Kinases , Mutation , Animals , Calcium-Calmodulin-Dependent Protein Kinases/biosynthesis , Cell Line , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , Enzyme Induction/radiation effects , Fibroblasts/enzymology , Fibroblasts/radiation effects , JNK Mitogen-Activated Protein Kinases , Keratinocytes/enzymology , Keratinocytes/radiation effects , Mice , Rats , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Ultraviolet Rays
8.
Clin Cancer Res ; 5(5): 1007-14, 1999 May.
Article in English | MEDLINE | ID: mdl-10353733

ABSTRACT

Cisplatin treatment activates multiple signal transduction pathways, which can lead to several cellular responses including cell cycle arrest, DNA repair, survival, or apoptosis. We investigated the response of the mitogen-activated protein kinases, extracellular signal-regulated kinases 1 and 2 (ERK1/2), c-Jun-N-terminal kinase 1 (JNK1), and p38, to cisplatin treatment in the ovarian carcinoma cell line SK-OV-3. Cisplatin caused a late and prolonged induction in a dose-dependent manner of both ERK1/2 and JNK1 activity. ERK1/2 and JNK1 activities continued to increase in magnitude up to 24 h following initiation of cisplatin treatment. In contrast, cisplatin treatment had no effect on p38 activity. Transplatin failed to induce either ERK1/2 or JNK1 at 24 h, which suggests that the activation of these kinases was dependent on cisplatin-specific DNA damage. Treatment with cycloheximide resulted in inhibition of cisplatin-induced ERK1/2 activation, demonstrating that ERK1/2 activity induced by cisplatin was dependent on de novo protein synthesis. Furthermore, inhibition of cisplatin-induced ERK1/2 activity by PD 98059 caused enhanced cisplatin cytotoxicity. Similar enhanced cytotoxic effects of cisplatin were also observed following treatment with PD 98059 in the ovarian carcinoma cell line UCI 101. These observations indicate that ERK1/2 activation induced by cisplatin partially protects cells from cisplatin cytotoxicity. Continued investigation into the mechanism by which the ERK pathway and other signal transduction pathways modulate the response to cisplatin may be helpful in the development of new strategies for improving the therapeutic use of platinum drugs.


Subject(s)
Adenocarcinoma/enzymology , Antineoplastic Agents/pharmacology , Calcium-Calmodulin-Dependent Protein Kinases/biosynthesis , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cisplatin/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Mitogen-Activated Protein Kinases , Neoplasm Proteins/biosynthesis , Ovarian Neoplasms/enzymology , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Cycloheximide/pharmacology , DNA Adducts , DNA Damage , DNA, Neoplasm/chemistry , DNA, Neoplasm/drug effects , Enzyme Activation/drug effects , Enzyme Induction/drug effects , Enzyme Inhibitors/pharmacology , Female , Flavonoids/pharmacology , Humans , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinase 1 , Mitogen-Activated Protein Kinase 3 , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Protein Synthesis Inhibitors/pharmacology , Signal Transduction/drug effects , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/enzymology , p38 Mitogen-Activated Protein Kinases
9.
Mol Carcinog ; 25(1): 14-20, 1999 May.
Article in English | MEDLINE | ID: mdl-10331740

ABSTRACT

Tumor necrosis factor-alpha (TNF alpha) can function as both an autocrine and a paracrine growth factor and may therefore play a role in ovarian tumor progression. TNF alpha initiates multiple cellular responses, many of which are mediated through the mitogen-activated protein kinase pathways, which transduce signals from the TNF alpha receptors through the cytoplasm to the nucleus, resulting in regulation of gene expression. We examined the role of c-jun N-terminal kinase 1 (JNK1) and extracellular signal-regulated protein kinase (ERK) 1 and 2 in the cellular growth response to TNF alpha in the ovarian carcinoma cell line UCI 101. JNK1 activity was increased to a maximum level ninefold above the basal level after 10-20 min of treatment with 10 ng/mL TNF alpha. A maximum threefold induction of ERK1/2 activity was observed after 1 min of treatment. At concentrations up to 100 ng/mL, TNF alpha had neither a stimulatory nor an inhibitory effect on growth of UCI 101 cells. However, inhibition of TNF alpha-induced ERK1/2 activity by the MAP/ERK kinase 1 inhibitor PD 98059 resulted in 60% inhibition of cell growth in TNF alpha-treated UCI 101 cells. This decrease in cell growth was accompanied by apoptosis, as demonstrated by the presence of a 180-bp DNA ladder. Thus, the inhibition of TNF alpha-induced ERK1/2 activity was associated with induction of apoptosis in the TNF alpha-resistant cell line UCI 101. Inhibition of TNF alpha-induced ERK1/2 activity was accompanied by a subsequent transient increase in TNF alpha-induced JNK1 activity. The significance of this increase with respect to apoptosis induction remains to be determined. These findings demonstrated that ERK1/2 activity can modulate cellular sensitivity to TNF alpha and suggested that the balance between the levels of ERK1/2 and JNK1 activation may be critical in the cellular growth response to TNF alpha.


Subject(s)
Adenocarcinoma/pathology , Apoptosis/drug effects , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases , Mitogen-Activated Protein Kinases , Ovarian Neoplasms/pathology , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Adenocarcinoma/enzymology , Blotting, Western , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Division/drug effects , Cell Survival/drug effects , DNA Fragmentation/drug effects , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Female , Flavonoids/pharmacology , Humans , JNK Mitogen-Activated Protein Kinases , MAP Kinase Kinase 1 , Mitogen-Activated Protein Kinase 1 , Mitogen-Activated Protein Kinase 3 , Ovarian Neoplasms/enzymology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Time Factors , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/antagonists & inhibitors
10.
Mol Carcinog ; 23(3): 159-67, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9833776

ABSTRACT

Exposure to solar ultraviolet (UV) radiation is believed to cause most human skin carcinomas. Despite the large body of evidence connecting UV exposure with skin cancer, the frequency and level of human exposure to repetitive doses of UV light will most likely continue for occupational and recreational reasons. By investigating the cellular response of keratinocytes to multiple, physiologically relevant doses of UV, we hope to better understand the processes involved in UV-induced skin cancer. In this study, we used a UV exposure model to investigate the cell-cycle response of keratinocytes exposed to multiple doses of UV-B/A radiation in which the UV-C component (wavelengths below 290 nm) had been filtered out. Our results indicated that exposure of asynchronous mouse keratinocytes to three doses of 200 J/m2 UV-B/A radiation at 30 min intervals produced an inhibition of DNA synthesis and S-phase arrest between 7 and 25 h after the last irradiation. The S-phase arrest was not due to a reduction in the level of cyclin E and A proteins but was accompanied by inhibition of cyclin-dependent kinase 2 (cdk2) activity. We observed a similar pattern of cdk2 inhibition induced by multiple UV-B/A irradiations in mouse embryo fibroblasts from p21WAF null mice, indicating that the inhibition of cdk2 was independent of p21WAF in these cells.


Subject(s)
CDC2-CDC28 Kinases , Keratinocytes/radiation effects , S Phase/radiation effects , Ultraviolet Rays/adverse effects , Animals , Blotting, Western , Cell Line , Cyclin A/metabolism , Cyclin E/metabolism , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/metabolism , Cyclin-Dependent Kinases/radiation effects , Cyclins/genetics , Cyclins/physiology , DNA/biosynthesis , DNA/radiation effects , Keratinocytes/cytology , Keratinocytes/metabolism , Mice , Mice, Inbred BALB C , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/radiation effects , S Phase/physiology
11.
Cancer Res ; 58(8): 1723-9, 1998 Apr 15.
Article in English | MEDLINE | ID: mdl-9563490

ABSTRACT

The biological effects of antioxidants are often considered in terms of their effects on oxygen or lipid radicals. However, antioxidants can also exert their effects through altering the cellular redox potential. Herein, we report that sulfur-containing antioxidants such as N-acetylcysteine and dimercaptopropanol induced apoptosis in several transformed cell lines and transformed primary cultures but not in normal cells. In contrast, chain-breaking antioxidants such as vitamin E lacked this activity. An increased glutathione level was not required for apoptosis; however, all apoptosis-inducing antioxidants elevated the total cellular thiol levels. Antioxidant-induced apoptosis required the p53 tumor suppressor gene. N-Acetylcysteine elevated p53 expression posttranscriptionally by increasing the rate of p53 mRNA translation rather than by altering the protein stability. The p53 induction occurred in normal cells. These observations indicate a redox sensor for p53 induction in vivo, with additional transformation-specific information being required for apoptosis. Manipulating p53-dependent apoptosis with nontoxic antioxidants may have a direct clinical application.


Subject(s)
Antioxidants/pharmacology , Apoptosis , Tumor Suppressor Protein p53/physiology , Acetylcysteine/pharmacology , Animals , Blotting, Northern , Blotting, Western , Buthionine Sulfoximine/pharmacology , Cells, Cultured , Dimercaprol/pharmacology , Dose-Response Relationship, Drug , Gene Expression Regulation, Neoplastic , Glutathione/analysis , Mice , Oxidation-Reduction , Pyrrolidonecarboxylic Acid , Sulfhydryl Compounds/analysis , Thiazoles/pharmacology , Thiazolidines , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism , Vitamin E/pharmacology
12.
Oncogene ; 16(11): 1501-5, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9525748

ABSTRACT

Jun N-terminal kinase (JNK1) is a member of a family of stress-activated protein kinases which are activated by many forms of stress including UV radiation, resulting in the phosphorylation of c-Jun, ATF-2, Elk-1 and p53. As UV-B radiation is mainly responsible for ultraviolet (UV)-induced skin cancers, we chose to elucidate JNK1 activation in keratinocytes which represent a UV-relevant cell system. We have demonstrated rapid activation of JNK1 in a keratinocyte cell line, C50, in response to multiple doses of UV-B irradiation. JNK1 activation occurred within 1 min, peaked by 10 min and returned to near basal levels within 2 h following the UV-B treatments. Our data provide the first evidence to show that keratinocytes do respond to multiple doses of the physiologically relevant UV-B radiation through rapid activation of the JNK1 pathway.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Epidermis/radiation effects , Keratinocytes/radiation effects , Mitogen-Activated Protein Kinases , Ultraviolet Rays , Cell Line, Transformed , Dose-Response Relationship, Radiation , Enzyme Activation , Epidermal Cells , Epidermis/enzymology , JNK Mitogen-Activated Protein Kinases , Keratinocytes/enzymology
13.
Mol Carcinog ; 19(2): 74-82, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9210954

ABSTRACT

Apigenin is a plant flavonoid that has been shown to significantly inhibit ultraviolet-induced mouse skin tumorigenesis when applied topically and may be an alternative sunscreen agent for humans. A long-term goal of our laboratory is to elucidate the molecular mechanism or mechanism by which apigenin inhibits skin tumorigenesis. In a previous publication, we characterized the mechanism by which apigenin induced G2/M arrest in keratinocytes. More recent studies in our laboratory have provided evidence that apigenin can induce G1 arrest in addition to arresting cells at G2/M. Here we describe the mechanism of the apigenin-induced G1 arrest in human diploid fibroblasts (HDF). Treatment of asynchronous HDF for 24 h with 10-50 microM apigenin resulted in dose-dependent cell-cycle arrest at both the G0/G1 and G2/M phases as measured by flow cytometry. The G0/G1 arrest was more clearly defined by using HDF that were synchronized in G0 and then released from quiescence by replating at subconfluent densities in medium containing 10-70 microM apigenin. The cells were analyzed for cell-cycle progression or cyclin D1 expression 24 h later. A dose of apigenin as low as 10 microM reduced the percentage of cells in S phase by 20% compared with control cultures treated with solvent alone. Western blot analysis of apigenin-treated HDF indicated that cyclin D1 was expressed at higher levels than in untreated cells, which signifies that they were arrested in G1 phase rather than in a G0 quiescent state. The G1 arrest was further studied by cyclin-dependent kinase 2 (cdk2) immune complex-kinase assays of apigenin-treated asynchronous HDF, which demonstrated a dose-dependent inhibition of cdk2 by apigenin. Inhibition of cdk2 kinase activity in apigenin-treated cells was associated with the accumulation of the hypophosphorylated form of the retinoblastoma (Rb) protein as measured by western blot analysis. The cdk inhibitor p21/WAF1 was also induced in a dose-dependent manner, with a 22-fold induction of p21/WAF1 in 70 microM apigenin-treated cells. In conclusion, apigenin treatment produced a G1 cell-cycle arrest by inhibiting cdk2 kinase activity and the phosphorylation of Rb and inducing the cdk inhibitor p21/WAF1, all of which may mediate its chemopreventive activities in vivo. To our knowledge this is the first report of a chemopreventive agent inducing p21/WAF1, a known downstream effector of the p53 tumor suppressor protein.


Subject(s)
Anticarcinogenic Agents/pharmacology , CDC2-CDC28 Kinases , Cell Cycle/drug effects , Cyclins/metabolism , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Oils, Volatile/pharmacology , Blotting, Western , Chamomile , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinases/metabolism , Dose-Response Relationship, Drug , Fibroblasts/drug effects , G1 Phase/drug effects , G2 Phase/drug effects , Genes, Retinoblastoma , Humans , Phosphorylation , Plants, Medicinal , Protein Serine-Threonine Kinases/metabolism , Resting Phase, Cell Cycle/drug effects
15.
Carcinogenesis ; 17(11): 2367-75, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8968050

ABSTRACT

Apigenin is a plant flavonoid which has been shown to significantly inhibit UV-induced mouse skin tumorigenesis when applied topically, and may represent an alternative sunscreen agent in humans. We have investigated the molecular mechanism(s) by which apigenin inhibits skin tumorigenesis. Initial studies examined the effects of apigenin on the cell cycle. DNA flow cytometric analysis indicated that culturing cells for 24 h in medium containing apigenin induced a G2/M arrest in two mouse skin derived cell lines, C50 and 308, as well as in human HL-60 cells. The G2/M arrest was fully reversible after an additional 24 h in medium without apigenin. We investigated the effects of apigenin on cyclin B1 and p34cdc2, since cyclin B1/p34cdc2 complexes regulate G2/M progression. Western blot and immune complex kinase assays using whole cell lysates from 308 and C50 cells treated for 24 h with 0-70 microM doses of apigenin demonstrated that apigenin treatment did not change the steady-state level of p34cdc2 protein, but did inhibit p34cdc2 H1 kinase activity in 308 cells. Western blot analysis showed that apigenin treatment of C50 cells and 308 cells inhibited the accumulation of cyclin B1 protein in a dose-dependent manner. The apigenin levels detected in cultured keratinocytes were relevant to those detected in epidermal cells of Sencar mice treated with tumor inhibitory doses of apigenin. In conclusion, we present evidence that apigenin induces a reversible G2/M arrest in cultured keratinocytes, the mechanism of which is in part due to inhibition of the mitotic kinase activity of p34cd2, and perturbation of cyclin B1 levels.


Subject(s)
Anticarcinogenic Agents/pharmacology , Cyclin B , Cyclin-Dependent Kinases , Flavonoids/pharmacology , G2 Phase/drug effects , Keratinocytes/cytology , Keratinocytes/drug effects , Mitosis/drug effects , Oils, Volatile/pharmacology , Animals , Anticarcinogenic Agents/pharmacokinetics , Cell Line , Chamomile , Cyclin B1 , Cyclins/drug effects , Cyclins/metabolism , Flavonoids/pharmacokinetics , HL-60 Cells , Humans , Keratinocytes/metabolism , Mice , Mice, Nude , Oils, Volatile/pharmacokinetics , Plants, Medicinal , Protein Serine-Threonine Kinases/drug effects , Protein Serine-Threonine Kinases/metabolism , Cyclin-Dependent Kinase-Activating Kinase
16.
Prog Clin Biol Res ; 395: 223-34, 1996.
Article in English | MEDLINE | ID: mdl-8895992

ABSTRACT

Considerable evidence suggests that dietary differences between populations account for a significant proportion of the variation in cancer occurrence in different parts of the world. A major problem has been identifying the particular dietary components which predispose or protect individuals against cancer. For example, the high rates of breast and colon cancer in the United States have been associated with numerous dietary patterns including high fat, high dietary energy, and low fruit and vegetable intakes. Our laboratories have attempted to identify mechanisms whereby diet may modify cancer and it is anticipated that future studies will determine which of these potential mechanisms may be relevant in humans. A promising lead in understanding the mechanism of high dietary fat/high dietary energy promotion of cancer was the impact of these diets on cellular protein kinase C (PKC). PKC is important in cellular signaling events which are critical to tumor promotion. Our studies demonstrated increased PKC activity and/or protein expression observed in epidermis and pancreatic epithelial cells of rodents fed high fat/energy diets. The inverse association between cancer at a number of sites and fruit and vegetable intake may be due to both micronutrient and non-nutrient components of fruits and vegetables. We have studied the prevention of skin tumor promotion by apigenin, a plant flavonoid. Apigenin may block several points in the process of tumor promotion, including inhibiting kinases, reducing transcription factors and regulating cell cycle. The complexity of our diets and the multitude of potential dietary effects which may be important in cancer development make this a fertile area for future study.


Subject(s)
Diet , Neoplasms/prevention & control , Anticarcinogenic Agents/therapeutic use , Diet/adverse effects , Dietary Fats/adverse effects , Dietary Fiber , Disease Susceptibility , Energy Intake , Energy Metabolism , Female , Flavonoids/therapeutic use , Fruit , Humans , Male , Neoplasms/epidemiology , Neoplasms/genetics , Neoplasms/metabolism , Pancreatic Neoplasms/chemically induced , Pancreatic Neoplasms/prevention & control , Risk , Skin Neoplasms/chemically induced , Skin Neoplasms/prevention & control , Vegetables , Vitamins/administration & dosage
17.
Oncogene ; 11(10): 1947-53, 1995 Nov 16.
Article in English | MEDLINE | ID: mdl-7478512

ABSTRACT

Exposure to solar ultraviolet (UV) light is a major cause of skin cancer, the most common human neoplasm. The earth's upper atmosphere absorbs the high energy UV-C wavelengths (100-280 nm), while allowing transmission of UV-B (280-320 nm) and UV-A (320-400 nm). It is therefore UV-B and to some extent UV-A, that contributes to most human skin malignancies. We report that the exposure of cultured keratinocytes or skin to UV-C radiation causes activation of MAP kinases (ERK and JNK). In contrast, the solar radiation associated with skin cancer (UV-B) was an ineffective activator of the ERK and JNK signal transduction pathways. Therefore, while exposure of epidermal cells to UV-C radiation under laboratory conditions causes marked activation of MAP kinase signal transduction pathways, only a low level of MAP kinase signaling is involved in the response of skin to biologically relevant solar radiation.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/radiation effects , Keratinocytes/radiation effects , Mitogen-Activated Protein Kinases , Signal Transduction/radiation effects , Ultraviolet Rays/adverse effects , Animals , CHO Cells/enzymology , CHO Cells/radiation effects , Calcium-Calmodulin-Dependent Protein Kinases/biosynthesis , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cells, Cultured , Cricetinae , Enzyme Activation/radiation effects , Enzyme Induction/radiation effects , Female , Gene Expression Regulation , Genes, ras , JNK Mitogen-Activated Protein Kinases , Keratinocytes/enzymology , Mice , Mice, Hairless , Mitogen-Activated Protein Kinase 1 , Mitogen-Activated Protein Kinase 3 , Protein-Tyrosine Kinases/metabolism , Protein-Tyrosine Kinases/radiation effects , Skin/enzymology , Skin/radiation effects , ras Proteins/biosynthesis , ras Proteins/genetics
18.
Proc Natl Acad Sci U S A ; 92(22): 10422-6, 1995 Oct 24.
Article in English | MEDLINE | ID: mdl-7479797

ABSTRACT

Mouse skin tumors contain activated c-H-ras oncogenes, often caused by point mutations at codons 12 and 13 in exon 1 and codons 59 and 61 in exon 2. Mutagenesis by the noncoding apurinic sites can produce G-->T and A-->T transversions by DNA misreplication with more frequent insertion of deoxyadenosine opposite the apurinic site. Papillomas were induced in mouse skin by several aromatic hydrocarbons, and mutations in the c-H-ras gene were determined to elucidate the relationship among DNA adducts, apurinic sites, and ras oncogene mutations. Dibenzo[a,l]pyrene (DB[a,l]P), DB[a,l]P-11,12-dihydrodiol, anti-DB[a,l]P-11,12-diol-13,14-epoxide, DB[a,l]P-8,9-dihydrodiol, 7,12-dimethylbenz[a]anthracene (DMBA), and 1,2,3,4-tetrahydro-DMBA consistently induced a CAA-->CTA mutation in codon 61 of the c-H-ras oncogene. Benzo[a]pyrene induced a GGC-->GTC mutation in codon 13 in 54% of tumors and a CAA-->CTA mutation in codon 61 in 15%. The pattern of mutations induced by each hydrocarbon correlated with its profile of DNA adducts. For example, both DB[a,l]P and DMBA primarily form DNA adducts at the N-3 and/or N-7 of deoxyadenosine that are lost from the DNA by depurination, generating apurinic sites. Thus, these results support the hypothesis that misreplication of unrepaired apurinic sites generated by loss of hydrocarbon-DNA adducts is responsible for transforming mutations leading to papillomas in mouse skin.


Subject(s)
Benz(a)Anthracenes/metabolism , Carcinogens/metabolism , DNA Adducts/metabolism , Genes, ras , Mutagenesis , Papilloma/genetics , Point Mutation , Skin Neoplasms/genetics , Skin/pathology , Animals , Apurinic Acid , Base Sequence , Benz(a)Anthracenes/toxicity , Carcinogens/toxicity , Codon/genetics , DNA Primers , DNA Replication , Exons , Female , Mice , Mice, Inbred SENCAR , Molecular Sequence Data , Papilloma/chemically induced , Polymerase Chain Reaction , Skin/drug effects , Skin Neoplasms/chemically induced , Structure-Activity Relationship
19.
Oncogene ; 10(10): 1955-60, 1995 May 18.
Article in English | MEDLINE | ID: mdl-7761096

ABSTRACT

The tumor suppressor gene p53 is involved in controlling cell cycle checkpoint or triggering apoptosis. p53 may accomplish these roles by acting as a sequence-specific transcription factor. One of the downstream targets of p53 transcription control is the WAF1/CIP1 gene, whose gene product p21 interacts with several cyclins and cyclin-dependent kinases, resulting in inhibition of these kinases. In our previous studies, we have shown that the p53 protein level in mouse keratinocytes was elevated following UV-B/A irradiation. In this paper we further investigated the consequences of increased p53 protein level by characterizing p53 DNA-binding level and WAF1/CIP1 gene expression in UV-B/A-irradiated mouse keratinocytes. Consistent with the increased level of p53 protein, both p53 DNA-binding level and steady-state level of WAF1/CIP1 mRNA were elevated. We have demonstrated that the induction of WAF1/CIP1 mRNA was mediated by p53, since no WAF1/CIP1 induction was observed in p53-deficient cells upon UV-B exposure. These observations suggest an important role for the tumor suppressor gene p53 in the response of keratinocytes to the biologically relevant UV-B/A irradiation and in suppressing UV-induced skin cancer.


Subject(s)
Cyclins/metabolism , Genes, p53/physiology , Keratinocytes/radiation effects , Tumor Suppressor Protein p53/physiology , Animals , Base Sequence , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , DNA/metabolism , Dose-Response Relationship, Radiation , Gene Deletion , Gene Expression Regulation, Neoplastic/radiation effects , Genes, p53/radiation effects , Humans , Keratinocytes/metabolism , Mice , Molecular Sequence Data , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/radiation effects
20.
Mol Carcinog ; 12(2): 66-76, 1995 Feb.
Article in English | MEDLINE | ID: mdl-7662118

ABSTRACT

Several lines of evidence that indicate that mutation of the Ha-ras oncogene is the initiating event in mouse skin carcinogenesis. Keratinocytes known to possess a mutated Ha-ras have been shown to be resistant to differentiation. Thus, overstimulation of the Ha-ras signaling pathway appears to block normal keratinocyte differentiation, and we hypothesized that for normal keratinocytes to terminally differentiate, the Ha-ras signaling cascade must be turned off. In the present studies, we measured the level and activity state of Ha-ras p21 protein in cultured keratinocytes undergoing calcium-induced differentiation. We have employed Western blot analysis to demonstrate that Ha-ras p21 protein levels remain constant during primary newborn and adult keratinocyte differentiation. The overall level of Ha-ras p21 was higher in immortalized, benign, and malignant mouse keratinocyte cell lines than in normal keratinocytes but did not change within each cell type when subjected to differentiating conditions. The percentage of Ha-ras p21 protein in its active, GTP-bound form also remained unchanged during primary adult keratinocyte differentiation and in immortalized, benign, and malignant keratinocytes subjected to differentiating conditions. Our results indicate that terminal differentiation of primary adult mouse keratinocytes occurred in the presence of constant levels of Ha-ras p21-GTP, suggesting that the Ha-ras signaling pathway may be blocked at a point distal to a step involving the Ha-ras p21 protein itself.


Subject(s)
Guanine Nucleotides/metabolism , Keratinocytes/cytology , Proto-Oncogene Proteins p21(ras)/metabolism , Animals , Animals, Newborn , Blotting, Western , Cell Differentiation , Cells, Cultured , Female , In Vitro Techniques , Keratinocytes/metabolism , Male , Mice , Mice, Inbred Strains
SELECTION OF CITATIONS
SEARCH DETAIL
...