Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 43
1.
Int J Obes (Lond) ; 42(4): 711-720, 2018 04.
Article En | MEDLINE | ID: mdl-29104286

BACKGROUND/OBJECTIVES: Obesity (body mass index (BMI)⩾30 kg m-2) is associated with an increased risk of estrogen-dependent breast cancer after menopause. Levels of aromatase, the rate-limiting enzyme in estrogen biosynthesis, are elevated in breast tissue of obese women. Recently, the regulation of aromatase by the p53-hypoxia-inducible factor-1α (HIF1α)/pyruvate kinase M2 (PKM2) axis was characterized in adipose stromal cells (ASCs) of women with Li-Fraumeni Syndrome, a hereditary cancer syndrome that predisposes to estrogen-dependent breast cancer. The current study aimed to determine whether stimulation of aromatase by obesity-associated adipokine leptin involves the regulation of the p53-HIF1α/PKM2 axis. SUBJECTS/METHODS: Human breast ASCs were used to characterize the p53-HIF1α/PKM2-aromatase axis in response to leptin. The effect of pharmacological or genetic modulation of protein kinase C (PKC), mitogen-activated protein kinase (MAPK), p53, Aha1, Hsp90, HIF1α and PKM2 on aromatase promoter activity, expression and enzyme activity was examined. Semiquantitative immunofluorescence and confocal imaging were used to assess ASC-specific protein expression in formalin-fixed paraffin-embedded tissue sections of breast of women and mammary tissue of mice following a low-fat (LF) or high-fat (HF) diet for 17 weeks. RESULTS: Leptin-mediated induction of aromatase was dependent on PKC/MAPK signaling and the suppression of p53. This, in turn, was associated with an increase in Aha1 protein expression, activation of Hsp90 and the stabilization of HIF1α and PKM2, known stimulators of aromatase expression. Consistent with these findings, ASC-specific immunoreactivity for p53 was inversely associated with BMI in breast tissue, while HIF1α, PKM2 and aromatase were positively correlated with BMI. In mice, HF feeding was associated with significantly lower p53 ASC-specific immunoreactivity compared with LF feeding, while immunoreactivity for HIF1α, PKM2 and aromatase were significantly higher. CONCLUSIONS: Overall, findings demonstrate a novel mechanism for the obesity-associated increase in aromatase in ASCs of the breast and support the study of lifestyle interventions, including weight management, which may reduce breast cancer risk via effects on this pathway.


Aromatase/metabolism , Breast Neoplasms/metabolism , Leptin/metabolism , Obesity/metabolism , Tumor Suppressor Protein p53/metabolism , Adipocytes/metabolism , Animals , Aromatase/genetics , Body Mass Index , Breast/cytology , Breast/metabolism , Carrier Proteins/metabolism , Cells, Cultured , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mammary Glands, Animal/metabolism , Membrane Proteins/metabolism , Mice , Signal Transduction , Thyroid Hormones/metabolism , Thyroid Hormone-Binding Proteins
2.
J Clin Oncol ; 21(14): 2645-50, 2003 Jul 15.
Article En | MEDLINE | ID: mdl-12860939

PURPOSE: Preclinical studies suggest that treatment with a selective cyclo-oxygenase-2 (COX-2) inhibitor may augment the antitumor effects of chemotherapy. In this study, patients with non-small-cell lung cancer (NSCLC) were preoperatively treated with celecoxib in combination with chemotherapy. End points were toxicity, response rates, and measurement of intratumoral levels of prostaglandin E2 (PGE2). METHODS: In this phase II trial, 29 patients with stages IB to IIIA NSCLC were treated with two preoperative cycles of paclitaxel and carboplatin, as well as daily celecoxib, followed by surgical resection. Levels of PGE2 in the primary tumors and adjacent normal lung tissue were compared in 17 study patients versus 13 controls, who received preoperative paclitaxel/carboplatin without celecoxib. RESULTS: All patients completed preoperative chemotherapy, and 26 completed preoperative celecoxib. The overall clinical response rate was 65% (48% with partial response; 17% with complete response). Grade 3 or 4 neutropenia was observed in 18 patients (62%). Twenty-eight patients were explored and underwent complete resection of their tumors. There were no complete pathologic responses, but seven patients (24%) had minimal residual microscopic disease. The addition of celecoxib to a regimen of paclitaxel and carboplatin abrogated the marked increase in levels of PGE2 detected in primary tumors after treatment with paclitaxel and carboplatin alone. CONCLUSION: In comparison with historically reported response rates, these data suggest that the addition of a selective COX-2 inhibitor may enhance the response to preoperative paclitaxel and carboplatin in patients with NSCLC. Moreover, treatment with celecoxib 400 mg twice daily was sufficient to normalize the increase in PGE2 levels found in NSCLC patients after treatment with paclitaxel and carboplatin. Confirmatory trials are planned.


Carboplatin/administration & dosage , Carcinoma, Non-Small-Cell Lung/drug therapy , Cyclooxygenase Inhibitors/administration & dosage , Lung Neoplasms/drug therapy , Paclitaxel/administration & dosage , Sulfonamides/administration & dosage , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols , Carboplatin/adverse effects , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/surgery , Celecoxib , Chemotherapy, Adjuvant , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Synergism , Female , Follow-Up Studies , Humans , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Lung Neoplasms/surgery , Male , Maximum Tolerated Dose , Middle Aged , Paclitaxel/adverse effects , Pneumonectomy , Preoperative Care/methods , Pyrazoles , Sulfonamides/adverse effects , Survival Analysis , Treatment Outcome
3.
Gastroenterology ; 121(6): 1391-9, 2001 Dec.
Article En | MEDLINE | ID: mdl-11729118

BACKGROUND & AIMS: Reflux of duodenal contents including bile acids is believed to contribute to esophageal injury and Barrett's esophagus. Cyclooxygenase (COX)-2, an inducible form of COX, has been implicated in both inflammation and carcinogenesis. In this study, we investigated the effects of bile acids and duodenal reflux on COX-2 expression in cultured esophageal cells and tissue, respectively. METHODS: Immunoblotting and Northern blotting were used to assess the effects of bile acids on COX-2 expression in esophageal cell lines. Immunoblotting and immunohistochemistry were performed to evaluate the effects of duodenal reflux on COX-2 expression and cell proliferation in esophageal tissue. RESULTS: Unconjugated bile acids were about fivefold more potent inducers of COX-2 messenger RNA, COX-2 protein, and prostaglandin synthesis than conjugated bile acids. Acidifying the culture medium sensitized esophageal cells to bile acid-mediated induction of COX-2. The induction of COX-2 by bile acids was mediated by phosphatidylinositol-3 kinase and extracellular signal-regulated kinase 1/2 mitogen-activated protein kinases. In experimental animals, duodenoesophageal reflux led to esophagitis, marked thickening of the esophageal mucosa, and enhanced expression of COX-2. Increased immunoreactivity for Ki-67 and cyclin D1 indicated that enhanced cell proliferation contributed to mucosal thickening. CONCLUSIONS: Reflux of duodenal contents into the esophagus led to increased COX-2 expression and mucosal thickening. Bile acids are likely to contribute to these effects.


Duodenogastric Reflux/enzymology , Esophagus/enzymology , Isoenzymes/metabolism , Mucous Membrane/enzymology , Prostaglandin-Endoperoxide Synthases/metabolism , Animals , Bile Acids and Salts/physiology , Chenodeoxycholic Acid/pharmacology , Cyclooxygenase 2 , Duodenogastric Reflux/complications , Enzyme Induction , Esophagus/drug effects , Esophagus/pathology , Gastroesophageal Reflux/complications , Gastroesophageal Reflux/enzymology , Gastroesophageal Reflux/pathology , Glycochenodeoxycholic Acid/pharmacology , Male , Mucous Membrane/drug effects , Phosphatidylinositol 3-Kinases/physiology , Prostaglandins/biosynthesis , Rats , Rats, Sprague-Dawley , Signal Transduction , Tumor Cells, Cultured
4.
Clin Cancer Res ; 7(12): 3971-6, 2001 Dec.
Article En | MEDLINE | ID: mdl-11751489

Recently, an inducible microsomal human prostaglandin E synthase (mPGES) was identified. This enzyme converts the cyclooxygenase (COX) product prostaglandin (PG) H(2) to PGE(2), an eicosanoid that has been linked to carcinogenesis. Increased amounts of PGE(2) have been observed in many tumor types including colorectal adenomas and cancers. To further elucidate the mechanism responsible for increased levels of PGE(2) in colorectal tumors, we determined the amounts of mPGES and COX-2 in 18 paired samples (tumor and adjacent normal) of colorectal cancer. With immunoblot analysis, mPGES was overexpressed in 83% of colorectal cancers. COX-2 was also commonly up-regulated in these tumors; marked differences in the extent of up-regulation of mPGES and COX-2 were observed in individual tumors. Immunohistochemistry revealed increased mPGES immunoreactivity in neoplastic cells in both colorectal adenomas and cancers compared with adjacent normal colonic epithelium. Cell culture was used to investigate the regulation of mPGES and COX-2. Chenodeoxycholate markedly induced COX-2 but not mPGES in colorectal cancer cells. Tumor necrosis factor-alpha induced both mPGES and COX-2, but the time course and magnitude of induction differed. As reported previously for COX-2, overexpressing Ras caused a several-fold increase in mPGES promoter activity. Taken together, our results suggest that overexpression of mPGES in addition to COX-2 contributes to increased amounts of PGE(2) in colorectal adenomas and cancer. The mechanisms controlling the expression of these two enzymes are not identical.


Adenoma/enzymology , Colorectal Neoplasms/enzymology , Intramolecular Oxidoreductases/biosynthesis , Adenocarcinoma , Blotting, Western , Cell Line , Chenodeoxycholic Acid/pharmacology , Colonic Neoplasms , Colorectal Neoplasms/pathology , Colorectal Neoplasms/surgery , Cyclooxygenase 2 , Dinoprostone/metabolism , Enzyme Induction , Gene Expression Regulation, Enzymologic , Humans , Intestinal Mucosa/enzymology , Intestinal Mucosa/pathology , Intramolecular Oxidoreductases/genetics , Isoenzymes/genetics , Isoenzymes/metabolism , Membrane Proteins , Prostaglandin-E Synthases , Prostaglandin-Endoperoxide Synthases/genetics , Prostaglandin-Endoperoxide Synthases/metabolism , Recombinant Proteins/biosynthesis , Transfection , Tumor Necrosis Factor-alpha/pharmacology
5.
Clin Cancer Res ; 7(11): 3349-55, 2001 Nov.
Article En | MEDLINE | ID: mdl-11705847

We investigated the effect of thalidomide, a compound with immunomodulatory and antiangiogenic properties, on lipopolysaccharide (LPS)-mediated induction of cyclooxygenase-2 (Cox-2) and prostaglandin (PG) biosynthesis in murine macrophages. Thalidomide caused a dose-dependent inhibition of LPS-mediated induction of PGE(2) synthesis in RAW 264.7 cells. The induction of Cox-2 protein and mRNA by LPS was also suppressed by thalidomide. Based on the results of nuclear run-off assays and transient transfections, treatment with LPS stimulated Cox-2 transcription, an effect that was unaffected by thalidomide. Thalidomide decreased the stability of Cox-2 mRNA. A series of structural analogues of thalidomide also inhibited LPS-mediated induction of Cox-2 and PGE(2) synthesis. Taken together, these data provide new insights into the antineoplastic and anti-inflammatory properties of thalidomide.


Isoenzymes/drug effects , Lipopolysaccharides/pharmacology , Prostaglandin-Endoperoxide Synthases/drug effects , Thalidomide/pharmacology , Animals , Blotting, Northern , Cell Line , Cyclooxygenase 2 , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Immunoblotting , Isoenzymes/genetics , Isoenzymes/metabolism , Prostaglandin-Endoperoxide Synthases/genetics , Prostaglandin-Endoperoxide Synthases/metabolism , RNA Stability/drug effects , RNA, Messenger/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Thalidomide/analogs & derivatives
6.
J Biomol Screen ; 6(2): 101-10, 2001 Apr.
Article En | MEDLINE | ID: mdl-11689104

Cyclooxygenase-2 (COX-2) is a recognized target for cancer prevention and possibly treatment. To identify novel inhibitors of COX-2, we developed a high throughput reporter gene assay that utilizes a region of the human COX-2 promoter to drive luciferase expression. A total of 968 extracts from 266 plants were screened. Extracts from 12 plants (4.5%), including Arnebia euchroma, a medicinal plant used in the Far East to treat inflammation, inhibited the stimulation of COX-2 promoter activity. The gene promoter assay then was used to identify shikonin, a compound with known anti-inflammatory and chemopreventive properties, as an active compound in A. euchroma. To complement the gene promoter studies, we determined the effects of a mixture of shikonins on phorbol 12-myristate 13-acetate (PMA)-mediated induction of COX-2 in transformed human mammary epithelial cells. Shikonins inhibited PMA-mediated induction of COX-2 mRNA, protein, and prostaglandin E(2) synthesis. In transient transfections, PMA caused a severalfold increase in COX-2 promoter activity, an effect that was suppressed by shikonins. Shikonins also inhibited PMA-mediated stimulation of extracellular signal-regulated kinase1/2 mitogen-activated protein kinases and activator protein-1 activity. Collectively, these results demonstrate the successful development and use of a high throughput reporter gene assay for the identification of a novel inhibitor of COX-2 expression.


Drug Evaluation, Preclinical/methods , Enzyme Inhibitors/pharmacology , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Plant Extracts/pharmacology , Promoter Regions, Genetic , Prostaglandin-Endoperoxide Synthases/genetics , Transcription, Genetic , Antineoplastic Agents, Phytogenic/pharmacology , Blotting, Northern , Blotting, Western , Breast/pathology , Cell Line, Transformed , Cyclooxygenase 2 , Dinoprostone/metabolism , Dose-Response Relationship, Drug , Epithelial Cells/metabolism , Humans , Luciferases/biosynthesis , Luciferases/genetics , Membrane Proteins , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Naphthoquinones/chemistry , Naphthoquinones/pharmacology , Plasmids/metabolism , RNA, Messenger/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Transcription Factor AP-1/metabolism , Transfection , Tumor Cells, Cultured
7.
Surgery ; 130(5): 826-33, 2001 Nov.
Article En | MEDLINE | ID: mdl-11685192

BACKGROUND: Cyclooxygenase-2 (Cox-2), the inducible form of Cox, is a rate-limiting enzyme in the synthesis of prostaglandins (PGs). Prostaglandin E2 (PGE2) and other eicosanoids possess immunosuppressive properties. Previously, traumatic injury was found to stimulate the synthesis of PGs and cause immune dysfunction. In this study a murine model was used to determine the effect of trauma on the expression of Cox-2 in macrophages and to elucidate the role of Cox-2 in trauma-induced immune dysfunction. METHODS: Mice were randomized to control or trauma (femur fracture plus 40% blood volume hemorrhage) groups. One, 4, and 7 days after injury, splenic macrophages were isolated and assayed for expression of Cox-2 and production of PGE2. In addition, the effect of pharmacologically inhibiting Cox-2 or knocking out the Cox-2 gene on trauma-induced suppression of splenocyte mitogenesis was determined. RESULTS: Trauma led to increased expression of Cox-2, enhanced synthesis of PGE2, and suppressed splenocyte mitogenesis. Both pharmacologic inhibition and genetic deletion of Cox-2 abrogated trauma-mediated suppression of splenocyte mitogenesis. CONCLUSIONS: These experiments link trauma-induced increases in Cox-2 expression and PGE2 production to reduced immune function. Cox-2 represents a potential pharmacologic target to prevent or reverse trauma-induced immunosuppression.


Isoenzymes/biosynthesis , Prostaglandin-Endoperoxide Synthases/biosynthesis , Wounds and Injuries/immunology , Animals , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Cyclooxygenase Inhibitors/pharmacology , Dinoprostone/biosynthesis , Enzyme Induction , Female , Immune Tolerance , Lymphocyte Activation , Macrophages/enzymology , Mice , Mice, Inbred BALB C , Wounds and Injuries/enzymology
8.
Clin Cancer Res ; 7(9): 2669-74, 2001 Sep.
Article En | MEDLINE | ID: mdl-11555578

An inducible microsomal form of human prostaglandin E synthase (mPGES) was recently identified. This enzyme converts the cyclooxygenase (COX) product, prostaglandin (PG) H2, to PGE2, a prostanoid that has been implicated in carcinogenesis. Increased amounts of PGE2 are detected in many types of cancer, but the underlying mechanism is not fully understood. Hence, we compared amounts of mPGES in 19 paired samples (tumor and adjacent normal tissue) of non-small cell lung cancer (NSCLC). By immunoblot analysis, mPGES was overexpressed in about 80% of NSCLCs. Immunohistochemistry localized the expression of mPGES to neoplastic epithelial cells. COX-2 was also commonly up-regulated in these tumors; marked differences in the extent of up-regulation of mPGES and COX-2 were observed in individual tumors. Cell culture was used to define the underlying mechanism(s) that accounts for up-regulation of mPGES in NSCLC. As reported previously for COX-2, levels of mPGES mRNA and protein were increased in NSCLC cell lines containing mutant Ras as compared with a nontumorigenic bronchial epithelial cell line. Nuclear run-offs revealed increased rates of mPGES transcription in the transformed cell lines. Overexpression of Ras caused a severalfold increase in mPGES promoter activity in nontransformed cells. Tumor necrosis factor-alpha induced mPGES and COX-2 in NSCLC cell lines but had no effect on the expression of either enzyme in a nontumorigenic bronchial epithelial cell line. Consistent with prior observations for COX-2, these data suggest that both cellular transformation and cytokines contribute to the up-regulation of mPGES in NSCLC.


Carcinoma, Non-Small-Cell Lung/pathology , Intramolecular Oxidoreductases/genetics , Lung Neoplasms/pathology , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/genetics , Cyclooxygenase 2 , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Immunoblotting , Intramolecular Oxidoreductases/drug effects , Intramolecular Oxidoreductases/metabolism , Isoenzymes/drug effects , Isoenzymes/metabolism , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Membrane Proteins , Prostaglandin-E Synthases , Prostaglandin-Endoperoxide Synthases/drug effects , Prostaglandin-Endoperoxide Synthases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/pharmacology
10.
Endocr Relat Cancer ; 8(2): 97-114, 2001 Jun.
Article En | MEDLINE | ID: mdl-11397667

Cyclooxygenase-2 (COX-2), an inducible prostaglandin synthase, is normally expressed in parts of the kidney and brain. Aberrant COX-2 expression was first reported in colorectal carcinomas and adenomas, and has now been detected in various human cancers, including those of the breast. Strikingly, COX-2 overexpression in murine mammary gland is sufficient to cause tumour formation. To date, the role of COX-2 in tumorigenesis has been most intensively studied in the colon. Thus, the relationship between COX-2 and neoplasia can best be illustrated with reference to intestinal tumorigenesis. Here we consider the potential utility of selective COX-2 inhibitors for the prevention and treatment of breast cancer. Data for cancers of the colon and breast are compared where possible. In addition, the mechanisms by which COX-2 is upregulated in cancers and contributes to tumorigenesis are discussed. Importantly, several recent studies of mammary tumorigenesis in animal models have found selective COX-2 inhibitors to be effective in the prevention and treatment of breast cancer. Clinical trials will be needed to determine whether COX-2 inhibition represents a useful approach to preventing or treating human breast cancer.


Breast Neoplasms/drug therapy , Cyclooxygenase Inhibitors/therapeutic use , Isoenzymes/antagonists & inhibitors , Animals , Breast Neoplasms/enzymology , Breast Neoplasms/prevention & control , Celecoxib , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/prevention & control , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Isoenzymes/biosynthesis , Isoenzymes/physiology , Membrane Proteins , Prostaglandin-Endoperoxide Synthases/biosynthesis , Prostaglandin-Endoperoxide Synthases/physiology , Pyrazoles , Sulfonamides/therapeutic use , Up-Regulation
11.
J Biol Chem ; 276(15): 12440-8, 2001 Apr 13.
Article En | MEDLINE | ID: mdl-11278336

We investigated whether peroxisome proliferator-activated receptor gamma (PPARgamma) ligands (ciglitazone, troglitazone, and 15-deoxy-Delta(12,14) prostaglandin J(2)) inhibited cyclooxygenase-2 (COX-2) induction in human epithelial cells. Ligands of PPARgamma inhibited phorbol ester (phorbol 12-myristate 13-acetate, PMA)-mediated induction of COX-2 and prostaglandin E(2) synthesis. Nuclear run-offs revealed increased rates of COX-2 transcription after treatment with PMA, an effect that was inhibited by PPARgamma ligands. PMA-mediated induction of COX-2 promoter activity was inhibited by PPARgamma ligands; this suppressive effect was prevented by overexpressing a dominant negative form of PPARgamma or a PPAR response element decoy oligonucleotide. The stimulatory effects of PMA were mediated by a cyclic AMP response element in the COX-2 promoter. Treatment with PMA increased activator protein-1 (AP-1) activity and the binding of c-Jun, c-Fos, and ATF-2 to the cyclic AMP response element, effects that were blocked by PPARgamma ligands. These findings raised questions about the mechanism underlying the anti-AP-1 effect of PPARgamma ligands. The induction of c-Jun by PMA was blocked by PPARgamma ligands. Overexpression of either c-Jun or CREB-binding protein/p300 partially relieved the suppressive effect of PPARgamma ligands. When CREB-binding protein and c-Jun were overexpressed together, the ability of PPARgamma ligands to suppress PMA-mediated induction of COX-2 promoter activity was essentially abrogated. Bisphenol A diglycidyl ether, a compound that binds to PPARgamma but lacks the ability to activate transcription, also inhibited PMA-mediated induction of AP-1 activity and COX-2. Taken together, these findings are likely to be important for understanding the anti-inflammatory and anti-cancer properties of PPARgamma ligands.


Isoenzymes/genetics , Nuclear Proteins/metabolism , Prostaglandin-Endoperoxide Synthases/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Trans-Activators/metabolism , Transcription Factor AP-1/metabolism , Transcription Factors/metabolism , Transcriptional Activation , Arthritis/prevention & control , Base Sequence , CREB-Binding Protein , Cell Line , Cyclooxygenase 2 , DNA Primers , Dinoprostone/biosynthesis , Enzyme Induction , Humans , Isoenzymes/biosynthesis , Ligands , Membrane Proteins , Neoplasms/prevention & control , Promoter Regions, Genetic , Prostaglandin-Endoperoxide Synthases/biosynthesis , Tetradecanoylphorbol Acetate/pharmacology
12.
Clin Cancer Res ; 7(2): 429-34, 2001 Feb.
Article En | MEDLINE | ID: mdl-11234900

Multiple lines of evidence suggest that cyclooxygenase-2 (COX-2) is an important target for preventing epithelial malignancies. Little is known, however, about the expression of COX-2 in gynecological malignancies. By immunoblot analysis, COX-2 was detected in 12 of 13 cases of cervical cancer but was undetectable in normal cervical tissue. Immunohistochemistry revealed COX-2 in malignant epithelial cells. COX-2 was also expressed in cervical intraepithelial neoplasia. The mechanism by which COX-2 is up-regulated in cervical cancer is unknown. Because the epidermal growth factor (EGF) receptor is commonly overexpressed in cervical cancer, we investigated whether EGF could induce COX-2 in cultured human cervical carcinoma cells. Treatment with EGF markedly induced COX-2 protein, COX-2 mRNA, and stimulated COX-2 promoter activity. The induction of COX-2 by EGF was suppressed by inhibitors of tyrosine kinase activity, phosphatidylinositol 3-kinase, mitogen-activated protein kinase kinase, and p38 mitogen-activated protein kinase. Moreover, overexpressing dominant-negative forms of extracellular signal-regulated kinase 1, c-Jun NH2-terminal kinase, p38, and c-Jun blocked EGF-mediated induction of COX-2 promoter activity. Taken together, these findings suggest that deregulation of the EGF receptor signaling pathway may lead to enhanced COX-2 expression in cervical cancer.


Adenocarcinoma/enzymology , Carcinoma, Adenosquamous/enzymology , Carcinoma, Squamous Cell/enzymology , Isoenzymes/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Sarcoma/enzymology , Uterine Cervical Neoplasms/enzymology , Blotting, Northern , Blotting, Western , Cyclooxygenase 2 , Female , Genes, erbB-1/physiology , Humans , Immunoenzyme Techniques , Isoenzymes/genetics , Membrane Proteins , Mitogen-Activated Protein Kinases/metabolism , Plasmids , Prostaglandin-Endoperoxide Synthases/genetics , RNA, Messenger/metabolism , Signal Transduction , Tumor Cells, Cultured
13.
J Biol Chem ; 276(23): 20108-15, 2001 Jun 08.
Article En | MEDLINE | ID: mdl-11274170

The inducible prostaglandin synthase cyclooxygenase-2 (COX-2) is aberrantly expressed in intestinal tumors resulting from APC mutation, and is also transcriptionally up-regulated in mouse mammary epithelial cells in response to Wnt1 expression. beta-Catenin stabilization is a consequence of both APC mutation and Wnt signaling. We have previously observed coordinate regulation of the matrilysin promoter by beta-catenin and Ets family transcription factors of the PEA3 subfamily. Here we show that while beta-catenin only weakly activates the COX-2 promoter, PEA3 family transcription factors are potent activators of COX-2 transcription. Consistent with this, PEA3 is up-regulated in Wnt1-expressing mouse mammary epithelial cells, and PEA3 factors are highly expressed in tumors from Wnt1 transgenic mice, in which Cox-2 is also up-regulated. Promoter mapping experiments suggest that the NF-IL6 site in the COX-2 promoter is important for mediating PEA3 responsiveness. The NF-IL6 site is also important for COX-2 transcription in some colorectal cancer lines (Shao, J., Sheng, H., Inoue, H., Morrow, J. D., and DuBois, R. N. (2000) J. Biol. Chem. 275, 33951-33956), and PEA3 factors are highly expressed in colorectal cancer cell lines. Therefore, we speculate that PEA3 factors may contribute to the up-regulation of COX-2 expression resulting from both APC mutation and Wnt1 expression.


Gene Expression Regulation, Enzymologic , Isoenzymes/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Proto-Oncogene Proteins/metabolism , Transcription Factors/metabolism , Up-Regulation , Zebrafish Proteins , Animals , Base Sequence , Cell Line , Cyclooxygenase 2 , DNA , Humans , Isoenzymes/genetics , Membrane Proteins , Mice , Molecular Sequence Data , Promoter Regions, Genetic , Prostaglandin-Endoperoxide Synthases/genetics , Wnt Proteins , Wnt1 Protein
14.
Lancet Oncol ; 2(9): 544-51, 2001 Sep.
Article En | MEDLINE | ID: mdl-11905709

Understanding the mechanisms underlying carcinogenesis provides insights that are necessary for the development of therapeutic strategies to prevent cancer. Chemoprevention--the use of drugs or natural substances to inhibit carcinogenesis - is an important and rapidly evolving aspect of cancer research. We discuss evidence that cyclooxygenase 2 (COX 2), an inducible form of the enzyme, is a potential pharmacological target to prevent cancer. Key data implicating a causal relation between increased activity of COX 2 and carcinogenesis and possible mechanisms of action of COX 2 in this context are covered. Importantly, selective COX 2 inhibitors appear to be safe enough in human beings to allow large-scale clinical testing in healthy people. Several chemoprevention trials using selective COX 2 inhibitors are underway.


Antineoplastic Agents/therapeutic use , Isoenzymes/antagonists & inhibitors , Neoplasms/drug therapy , Neoplasms/prevention & control , Apoptosis , Cyclooxygenase 2 , Humans , Immunosuppression Therapy , Inflammation , Isoenzymes/physiology , Membrane Proteins , Neoplasm Invasiveness , Neoplasms/enzymology , Neoplasms/etiology , Neoplasms/pathology , Neovascularization, Pathologic , Prostaglandin-Endoperoxide Synthases/physiology , Prostaglandins/biosynthesis , Xenobiotics
15.
Ann N Y Acad Sci ; 952: 109-15, 2001 Dec.
Article En | MEDLINE | ID: mdl-11795429

Multiple lines of evidence suggest that cyclooxygenase-2 (COX-2), an inducible form of COX, represents a potential pharmacologic target to prevent cancer. Key data suggesting a causal relationship between increased COX-2 activity and carcinogenesis and possible mechanisms of action of COX-2 in this context will be discussed. The possibility that COX-2 represents a pharmacological target for preventing upper aerodigestive cancers (head and neck, lung) will be emphasized. Importantly, clinical trials have been initiated to assess the chemopreventive properties of selective COX-2 inhibitors.


Anticarcinogenic Agents/therapeutic use , Cyclooxygenase Inhibitors/therapeutic use , Esophageal Neoplasms/prevention & control , Isoenzymes/antagonists & inhibitors , Respiratory Tract Neoplasms/prevention & control , Animals , Apoptosis/drug effects , Barrett Esophagus/drug therapy , Cell Transformation, Neoplastic/drug effects , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Disease Progression , Drug Screening Assays, Antitumor , Esophageal Neoplasms/enzymology , Head and Neck Neoplasms/enzymology , Head and Neck Neoplasms/prevention & control , Isoenzymes/physiology , Leukoplakia, Oral/drug therapy , Lung Neoplasms/enzymology , Lung Neoplasms/prevention & control , Mice , Mice, Knockout , Neoplasms, Experimental/prevention & control , Precancerous Conditions/drug therapy , Prostaglandin-Endoperoxide Synthases/physiology , Respiratory Tract Neoplasms/enzymology , Xenograft Model Antitumor Assays
16.
Cancer Res ; 60(9): 2399-404, 2000 May 01.
Article En | MEDLINE | ID: mdl-10811116

We investigated the effects of ursolic acid, a chemopreventive agent, on the expression of cyclooxygenase-2 (COX-2) in phorbol 12-myristate 13-acetate (PMA)-treated human mammary and oral epithelial cells. Treatment with ursolic acid suppressed PMA-mediated induction of COX-2 protein and synthesis of prostaglandin E2. Ursolic acid also suppressed the induction of COX-2 mRNA by PMA. Nuclear run-offs revealed increased rates of COX-2 transcription after treatment with PMA, an effect that was inhibited by ursolic acid. Transient transfections indicated that the effects of PMA were mediated by a cyclic AMP response element in the COX-2 promoter. Ursolic acid inhibited PMA-mediated activation of protein kinase C, extracellular signal-regulated kinase 1/2, c-Jun N-terminal kinase, and p38 mitogen-activated protein kinases. Treatment with PMA increased activator protein-1 activity and the binding of c-Jun to the cyclic AMP response element of the COX-2 promoter, effects that were blocked by ursolic acid. These data are important for understanding the anticancer and anti-inflammatory properties of ursolic acid.


Breast/enzymology , JNK Mitogen-Activated Protein Kinases , Transcription, Genetic , Triterpenes/pharmacology , Blotting, Northern , Carcinoma, Squamous Cell/metabolism , Cell Line , Cyclooxygenase 2 , Dinoprostone/biosynthesis , Dose-Response Relationship, Drug , Epithelial Cells/enzymology , Female , Humans , Isoenzymes/metabolism , MAP Kinase Kinase 4 , Membrane Proteins , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Plasmids , Prostaglandin-Endoperoxide Synthases/metabolism , Protein Kinase C/metabolism , RNA, Ribosomal, 18S/metabolism , Signal Transduction , Tetradecanoylphorbol Acetate/pharmacology , Tongue Neoplasms/metabolism , Transcription, Genetic/drug effects , Transfection , Triterpenes/chemistry , Tumor Cells, Cultured , p38 Mitogen-Activated Protein Kinases , Ursolic Acid
17.
J Biol Chem ; 275(20): 14838-45, 2000 May 19.
Article En | MEDLINE | ID: mdl-10809726

We investigated whether microtubule-interfering agents (MIAs: taxol, colchicine, nocodazole, vinblastine, vincristine, 17-beta-estradiol, 2-methoxyestradiol) altered cyclooxygenase-2 (COX-2) expression in human mammary epithelial cells. MIAs enhanced prostaglandin E(2) synthesis and increased levels of COX-2 protein and mRNA. Nuclear run-off assays revealed increased rates of COX-2 transcription after treatment with MIAs. Calphostin C, an inhibitor of protein kinase C, blocked the induction of COX-2 by MIAs. The stimulation of COX-2 promoter activity by MIAs was inhibited by overexpressing dominant negative forms of Rho and Raf-1. MIAs stimulated ERK, JNK, and p38 mitogen-activated protein kinases (MAPK); pharmacological inhibitors of MAPK kinase and p38 MAPK blocked the induction of COX-2 by MIAs. Overexpressing dominant negative forms of ERK1 or p38 MAPK inhibited MIA-mediated activation of the COX-2 promoter. MIAs stimulated the binding of the activator protein-1 transcription factor complex to the cyclic AMP response element in the COX-2 promoter. A dominant negative form of c-Jun inhibited the activation of the COX-2 promoter by MIAs. Additionally, cytochalasin D, an agent that inhibits actin polymerization, stimulated COX-2 transcription by the same signaling pathway as MIAs. Thus, microtubule- or actin-interfering agents stimulated MAPK signaling and activator protein-1 activity. This led, in turn, to induction of COX-2 gene expression via the cyclic AMP response element site in the COX-2 promoter.


Isoenzymes/genetics , Microtubules/physiology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinases/metabolism , Prostaglandin-Endoperoxide Synthases/genetics , Transcription, Genetic , 2-Methoxyestradiol , Breast/cytology , Cell Line , Colchicine/pharmacology , Cyclooxygenase 2 , Epithelial Cells/cytology , Epithelial Cells/physiology , Estradiol/analogs & derivatives , Estradiol/pharmacology , Female , Humans , Kinetics , Membrane Proteins , Microtubules/drug effects , Mitogen-Activated Protein Kinase 3 , Nocodazole/pharmacology , Paclitaxel/pharmacology , Vinblastine/pharmacology , Vincristine/pharmacology , p38 Mitogen-Activated Protein Kinases
18.
J Biol Chem ; 275(7): 4949-55, 2000 Feb 18.
Article En | MEDLINE | ID: mdl-10671533

Polycyclic aromatic hydrocarbons, such as benzo[a]pyrene (B[a]P) present in tobacco smoke and tar, have been implicated in the development of atherosclerosis as well as cancer. Increased expression of cyclooxygenase-2 (COX-2) has been detected both in atherosclerotic lesions and in epithelial cancers. To determine whether polycyclic aromatic hydrocarbons might directly affect COX expression in vascular cells, we investigated the effects of B[a]P on COX-2 expression in human and rat arterial smooth muscle cells (SMC). Treatment with B[a]P increased levels of COX-2 protein and mRNA and enhanced prostaglandin synthesis. Nuclear runoff assays and transient transfections revealed increased COX-2 gene transcription after treatment with B[a]P. Experiments were done to define the signaling mechanism by which B[a]P induced COX-2. B[a]P caused a rapid increase in phosphorylation of extracellular signal-regulated kinase (ERK); pharmacologic inhibition of mitogen-activated protein kinase kinase blocked B[a]P-mediated induction of COX-2. Depletion of the intracellular antioxidant, glutathione, with buthionine sulfoximine significantly increased B[a]P-mediated induction of COX-2 while exposure to N-acetylcysteine, a precursor of glutathione, suppressed the induction of COX-2 by B[a]P. Several lines of evidence suggest that the induction of COX-2 by B[a]P is mediated, at least in part, by NF-kappaB. Treatment with B[a]P increased binding of NF-kappaB to DNA. Moreover, B[a]P-mediated stimulation of COX-2 promoter activity was blocked when a construct containing a mutagenized NF-kappaB site was used. Pharmacological inhibitors of NF-kappaB blocked the induction of COX-2 protein and the stimulation of COX-2 promoter activity by B[a]P. Taken together, these data are likely to be important for understanding the atherogenic effects of tobacco smoke.


Benzo(a)pyrene/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Isoenzymes/genetics , Mitogen-Activated Protein Kinases/metabolism , Muscle, Smooth, Vascular/drug effects , NF-kappa B/metabolism , Prostaglandin-Endoperoxide Synthases/genetics , Transcription, Genetic/drug effects , Animals , Base Sequence , Cyclooxygenase 2 , DNA Primers , Humans , Membrane Proteins , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/enzymology , Oxidative Stress , Rats
19.
J Biol Chem ; 274(41): 29138-48, 1999 Oct 08.
Article En | MEDLINE | ID: mdl-10506169

Treatment of normal human epidermal keratinocytes (NHEK) with interferon-gamma (IFN-gamma) causes a 9-fold increase in the level of cyclooxygenase-2 (COX-2) mRNA expression. Nuclear run-off assays indicate that this induction is at least partly due to increased transcription. Activation of the epidermal growth factor receptor (EGFR) signaling pathway due to the enhanced transforming growth factor alpha (TGFalpha) expression plays an important role in the induction of COX-2 by IFN-gamma. This is supported by the ability of TGFalpha to rapidly induce COX-2 and the inhibition of the IFN-gamma-mediated COX-2 mRNA induction by an EGFR antibody and EGFR-selective kinase inhibitors. Deletion and mutation analysis indicates the importance of the proximal cAMP-response element/ATF site in the transcriptional control of this gene by TGFalpha. The increase in COX-2 mRNA by TGFalpha requires activation of both the extracellular signal-regulated kinase (ERK) and p38 mitogen-activated protein kinase (MAPK) pathways. Inhibition of p38 MAPK decreases the stability of COX-2 mRNA, while inhibition of MAPK/ERK kinase (MEK) does not. These results suggest that the p38 MAPK signaling pathway controls COX-2 at the level of mRNA stability, while the ERK signaling pathway regulates COX-2 at the level of transcription. In contrast to NHEK, IFN-gamma and TGFalpha are not very effective in inducing TGFalpha or COX-2 expression in several squamous carcinoma cell lines, indicating alterations in both IFN-gamma and TGFalpha response pathways.


Gene Expression Regulation, Enzymologic/drug effects , Intercellular Signaling Peptides and Proteins , Interferon-gamma/pharmacology , Isoenzymes/metabolism , Mitogen-Activated Protein Kinases/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Transforming Growth Factor alpha/pharmacology , Amphiregulin , Carcinoma, Squamous Cell , Cyclooxygenase 1 , Cyclooxygenase 2 , Dinoprostone/metabolism , EGF Family of Proteins , Eicosanoids/metabolism , Enzyme Inhibitors/pharmacology , ErbB Receptors/genetics , Flavonoids/pharmacology , Glycoproteins/metabolism , Growth Substances/metabolism , Humans , Isoenzymes/genetics , Keratinocytes , Membrane Proteins , Promoter Regions, Genetic , Prostaglandin-Endoperoxide Synthases/genetics , RNA, Messenger/metabolism , Regulatory Sequences, Nucleic Acid , Signal Transduction/genetics , Tumor Cells, Cultured
20.
Cancer Res ; 59(10): 2347-52, 1999 May 15.
Article En | MEDLINE | ID: mdl-10344742

We investigated the mechanisms by which caffeic acid phenethyl ester (CAPE), a phenolic antioxidant, inhibited the stimulation of prostaglandin (PG) synthesis in cultured human oral epithelial cells and in an animal model of acute inflammation. Treatment of cells with CAPE (2.5 microg/ml) suppressed phorbol ester (12-O-tetradecanoylphorbol-13-acetate; TPA) and calcium ionophore (A23187)-mediated induction of PGE2 synthesis. This relatively low concentration of CAPE did not affect amounts of cyclooxygenase (COX) enzymes. CAPE nonselectively inhibited the activities of baculovirus-expressed hCOX-1 and hCOX-2 enzymes. TPA- and A23187-stimulated release of arachidonic acid from membrane phospholipids was also suppressed by CAPE (4-8 microg/ml). Higher concentrations of CAPE (10-20 microg/ml) suppressed the induction of COX-2 mRNA and protein mediated by TPA. Transient transfections using human COX-2 promoter deletion constructs were performed; the effects of TPA and CAPE were localized to a 124-bp region of the COX-2 promoter. In the rat carrageenan air pouch model of inflammation, CAPE (10-100 mg/kg) caused dose-dependent suppression of PG synthesis. Amounts of COX-2 in the pouch were markedly suppressed by 100 mg/kg CAPE but were unaffected by indomethacin. These data are important for understanding the anticancer and anti-inflammatory properties of CAPE.


Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anticarcinogenic Agents/pharmacology , Caffeic Acids/pharmacology , Cyclooxygenase Inhibitors/pharmacology , Inflammation/genetics , Isoenzymes/biosynthesis , Mouth Mucosa/cytology , Phenylethyl Alcohol/analogs & derivatives , Promoter Regions, Genetic/drug effects , Prostaglandin-Endoperoxide Synthases/biosynthesis , Air , Animals , Arachidonic Acids/metabolism , Calcimycin/antagonists & inhibitors , Calcimycin/pharmacology , Carcinoma, Squamous Cell/pathology , Carrageenan/toxicity , Cell Membrane/drug effects , Cell Membrane/metabolism , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Dinoprostone/biosynthesis , Enzyme Activation/drug effects , Enzyme Induction/drug effects , Epithelial Cells/drug effects , Epithelial Cells/enzymology , Genetic Vectors/genetics , Humans , Indomethacin/pharmacology , Inflammation/chemically induced , Inflammation/metabolism , Ionophores/antagonists & inhibitors , Ionophores/pharmacology , Isoenzymes/genetics , Male , Membrane Lipids/metabolism , Membrane Proteins , Nucleopolyhedroviruses/genetics , Phenylethyl Alcohol/pharmacology , Phospholipids/metabolism , Prostaglandin-Endoperoxide Synthases/genetics , Rats , Rats, Inbred Lew , Recombinant Fusion Proteins/biosynthesis , Tetradecanoylphorbol Acetate/antagonists & inhibitors , Tetradecanoylphorbol Acetate/pharmacology , Transfection , Tumor Cells, Cultured
...