Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 118
Filter
1.
Sci Rep ; 9(1): 6100, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30988343

ABSTRACT

Quantifying tumor burden is important for following the natural history of orthotopic colon cancer and therapeutic efficacy. Bioluminescence imaging (BLI) is commonly used for such assessment and has both advantages and limitations. We compared BLI and magnetic resonance imaging (MRI) for quantifying orthotopic tumors in a mouse model of colon cancer. Among sequences tested, T2-based MRI imaging ranked best overall for colon cancer border delineation, contrast, and conspicuity. Longitudinal MRI detected tumor outside the colon, indistinguished by BLI. Colon tumor weights calculated from MRI in vivo correlated highly with tumor weights measured ex vivo whereas the BLI signal intensities correlated relatively poorly and this difference in correlations was highly significant. This suggests that MRI may more accurately assess tumor burden in longitudinal monitoring of orthotopic colon cancer in this model as well as in other models.


Subject(s)
Colonic Neoplasms/diagnostic imaging , Luminescent Measurements , Magnetic Resonance Imaging , Animals , HCT116 Cells , Humans , Male , Mice , Mice, Nude , Neoplasm Transplantation
2.
Oncogene ; 31(8): 1013-23, 2012 Feb 23.
Article in English | MEDLINE | ID: mdl-21765467

ABSTRACT

Peroxisome proliferator-activated receptor γ (PPARγ) may serve as a useful target for drug development in non-diabetic diseases. However, some colorectal cancer cells are resistant to PPARγ agonists by mechanisms that are poorly understood. Here, we provide the first evidence that elevated PPARδ expression and/or activation of PPARδ antagonize the ability of PPARγ to induce colorectal carcinoma cell death. More importantly, the opposing effects of PPARδ and PPARγ in regulating programmed cell death are mediated by survivin and caspase-3. We found that activation of PPARγ results in decreased survivin expression and increased caspase-3 activity, whereas activation of PPARδ counteracts these effects. Our findings suggest that PPARδ and PPARγ coordinately regulate cancer cell fate by controlling the balance between the cell death and survival and demonstrate that inhibition of PPARδ can reprogram PPARγ ligand-resistant cells to respond to PPARγ agonists.


Subject(s)
Apoptosis/drug effects , Colorectal Neoplasms/metabolism , Inhibitor of Apoptosis Proteins/metabolism , PPAR delta/metabolism , PPAR gamma/metabolism , Caspase 3/metabolism , Cell Line, Tumor , Cell Survival , DNA Fragmentation , Gene Expression , Gene Expression Regulation, Neoplastic , Gene Knockout Techniques , Humans , Inhibitor of Apoptosis Proteins/genetics , Oxazoles/pharmacology , PPAR delta/agonists , PPAR delta/genetics , PPAR gamma/agonists , PPAR gamma/genetics , Survivin , Thiazoles/pharmacology , Tyrosine/analogs & derivatives , Tyrosine/pharmacology
3.
Oncogene ; 29(6): 781-8, 2010 Feb 11.
Article in English | MEDLINE | ID: mdl-19946329

ABSTRACT

Colorectal cancer (CRC) is a heterogeneous disease, including at least three major forms: hereditary, sporadic and colitis-associated CRC. A large body of evidence indicates that genetic mutations, epigenetic changes, chronic inflammation, diet and lifestyle are the risk factors for CRC. As elevated cyclooxygenase-2 (COX-2) expression was found in most CRC tissue and is associated with worse survival among CRC patients, investigators have sought to evaluate the effects of nonsteroidal anti-inflammatory drugs (NSAIDs) and selective COX-2 inhibitors (COXIBs) on CRC. The epidemiological studies, clinical trials and animal experiments indicate that NSAIDs are among the most promising chemopreventive agents for this disease. NSAIDs exert their anti-inflammatory and antitumor effects primarily by reducing prostaglandin production by inhibition of COX-2 activity. In this review, we highlight breakthroughs in our understanding of the roles of COX-2 in CRC and inflammatory bowel disease. These recent data provide a rationale for re-evaluating COX-2 as both the prognostic and the predictive marker in a wide variety of malignancies and for renewing the interest in evaluating relative benefits and risk of COXIBs in appropriately selected patients for cancer prevention and treatment.


Subject(s)
Colorectal Neoplasms/enzymology , Cyclooxygenase 2/metabolism , Intestinal Diseases/enzymology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/prevention & control , Dinoprostone/metabolism , Humans , Inflammation/enzymology , Inflammation/metabolism , Intestinal Diseases/metabolism
4.
Gut ; 55(1): 115-22, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16118353

ABSTRACT

Chemoprevention has been considered as a possible approach for cancer prevention. A significant effort has been made in the development of novel drugs for both cancer prevention and treatment over the past decade. Recent epidemiological studies and clinical trials indicate that long term use of aspirin and similar agents, also called non-steroidal anti-inflammatory drugs (NSAIDs), can decrease the incidence of certain malignancies, including colorectal, oesophageal, breast, lung, and bladder cancers. The best known targets of NSAIDs are cyclooxygenase (COX) enzymes, which convert arachidonic acid to prostaglandins (PGs) and thromboxane. COX-2 derived prostaglandin E(2)(PGE(2)) can promote tumour growth by binding its receptors and activating signalling pathways which control cell proliferation, migration, apoptosis, and/or angiogenesis. However, the prolonged use of high dosages of COX-2 selective inhibitors (COXIBs) is associated with unacceptable cardiovascular side effects. Thus it is crucial to develop more effective chemopreventive agents with minimal toxicity. Recent efforts to identify the molecular mechanisms by which PGE(2) promotes tumour growth and metastasis may provide opportunities for the development of safer strategies for cancer prevention and treatment.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Dinoprostone/physiology , Neoplasms/prevention & control , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Anticarcinogenic Agents/therapeutic use , Cyclooxygenase Inhibitors/therapeutic use , Humans
5.
Br J Cancer ; 88(9): 1445-52, 2003 May 06.
Article in English | MEDLINE | ID: mdl-12778076

ABSTRACT

Matrix metalloproteinases (MMPs) and cyclooxygenase-2 (COX-2) are expressed in both sporadic and familial adenomatous colonic polyps and tumours and have been independently shown to play causal roles in intestinal tumour formation in mouse models of colon cancer. The apparent roles of these enzymes in intestinal tumorigenesis led us to examine, in the Min mouse model of colon cancer, if selective COX-2 and MMP inhibitors provide additive or synergistic therapeutic benefits in intestinal tumour prevention. The broad-spectrum MMP inhibitor (A-177430; MMPI) and the selective COX-2 inhibitor (A-285969; COX-2I) both showed dose-dependent inhibition of the number of adenomas in Min mice. Using suboptimal doses, the MMPI reduced tumour multiplicity by 32%, the COX-2I by 48% and, both agents in combination resulted in a 67% decrease compared to control demonstrating a cooperative effect on intestinal tumorigenesis. Apoptosis, proliferation, and angiogenesis were assayed in tumors from each treatment group. These agents in combination allowed for a lowered dosage to be administered to achieve significant biological effects. Clinically, this could potentially reduce side effects associated with currently used MMP and COX-2 inhibitors. Together, these compounds could represent an easily tolerated chemopreventive approach.


Subject(s)
Adenoma/drug therapy , Cyclooxygenase Inhibitors/therapeutic use , Enzyme Inhibitors/therapeutic use , Intestinal Neoplasms/drug therapy , Isoenzymes/metabolism , Matrix Metalloproteinase Inhibitors , Peptides, Cyclic/therapeutic use , Prostaglandin-Endoperoxide Synthases/metabolism , Adenoma/pathology , Animals , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Intestinal Neoplasms/pathology , Mice , Mice, Inbred C57BL
6.
Neoplasia ; 3(5): 428-36, 2001.
Article in English | MEDLINE | ID: mdl-11687954

ABSTRACT

Selective COX-2 inhibitors reduce adenoma formation and cancer progression in rodent models of colorectal cancer. To assess the therapeutic activity of selective COX-2 inhibitors, we tested the effect of SC-58125 treatment on the growth of human colon carcinoma cells in nude mice. Delaying treatment by 2, 4, or 7 weeks following implantation of the carcinoma cells resulted in a significant inhibition of tumor growth. Furthermore, short-term (48 hours) treatment with SC-58125 was sufficient to attenuate tumor growth for up to 15 days. SC-58125 treatment did not alter the rate at which cells underwent apoptosis, but did result in a delayed progression through the cell cycle at the G(2)/M transition. Accordingly, p34(cdc2) protein levels and activity were decreased following SC-58125 treatment. We conclude that SC-58125 primarily exerts a cytostatic effect in vivo, which is likely to be mediated through inhibition of progression through the G(2)/M phase of the cell cycle.


Subject(s)
Cell Division/drug effects , Colorectal Neoplasms/drug therapy , Cyclooxygenase Inhibitors/therapeutic use , Isoenzymes/antagonists & inhibitors , Pyrazoles/therapeutic use , Animals , Apoptosis , Cell Division/physiology , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/pathology , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Flow Cytometry , Gene Expression Profiling , Humans , Immunoblotting , Isoenzymes/genetics , Membrane Proteins , Mice , Mice, Nude , Mitotic Index , Prostaglandin-Endoperoxide Synthases/genetics , RNA/analysis , RNA/metabolism , Tetrazolium Salts , Thiazoles , Transplantation, Heterologous , Tumor Cells, Cultured
7.
Clin Cancer Res ; 7(10): 2998-3005, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11595687

ABSTRACT

It has been proposed that Cyclooxygenase (COX)-2 inhibitors may be able to enhance the effects of chemotherapeutic or radiation treatment; however, currently few studies have been reported that define the radiation-enhancing effect of COX-2 inhibitors. We conducted in vitro radiation survival experiments using rat intestinal epithelial cells which were stably transfected with COX-2 cDNA in the sense (RIE-S) and antisense (RIE-AS) orientations to investigate the potential radiosensitizing effect of the selective COX-2 inhibitor, NS-398. Apoptosis was measured using 7-aminoactinomycin-D with flow cytometry to investigate underlying mechanisms for the effect of NS-398 on radiosensitivity. The same experiments were repeated with NCI-H460 human lung cancer cells, which express COX-2 constitutively, and HCT-116 human colon cancer cells, which lack COX-2 expression. In vivo tumor growth delay assays were also performed with tumors formed by H460 and HCT-116 cells. No difference was observed in the intrinsic radiation sensitivity of RIE-S and RIE-AS cells exposed to radiation alone. However, 150-400 microM of NS-398 enhanced radiosensitivity in a concentration-dependent manner in RIE-S cells with dose enhancement ratios of 1.2-1.9 at a surviving fraction of 0.25. However, this effect was not shown in RIE-AS cells. NS-398 enhanced radiosensitivity in H460 cells with a dose enhancement ratio of 1.8 but protected HCT-116 cells from the effects of radiation. Radiation-induced apoptosis was enhanced by NS-398 in RIE-S and H460 cells but not in RIE-AS and HCT-116 cells. Additionally, this radiation-enhancing effect in RIE-S cells seemed to be attributable to some mechanisms other than the reversal of radioresistance induced by COX-2. NS-398 (36 mg/kg) enhanced the effect of radiation on H460 tumors in vivo by an enhancement factor of 2.5; however, it did not enhance the radiosensitivity of HCT-116 tumors (enhancement factor = 1.04). These in vitro and in vivo results suggest that selective COX-2 inhibitors enhance the effect of radiation on tumors that express COX-2 but not on COX-2-lacking tumors. This effect may be attributable to enhancement of radiation-induced apoptosis. Thus, selective COX-2 inhibitors may have potential as radiosensitizers for treatment of human cancers.


Subject(s)
Cyclooxygenase Inhibitors/pharmacology , Isoenzymes/antagonists & inhibitors , Nitrobenzenes/pharmacology , Sulfonamides/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Survival/radiation effects , Cells, Cultured , Combined Modality Therapy , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Cyclooxygenase Inhibitors/therapeutic use , DNA, Antisense/genetics , Dose-Response Relationship, Radiation , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/radiation effects , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/drug effects , Intestinal Mucosa/radiation effects , Isoenzymes/genetics , Membrane Proteins , Mice , Mice, Nude , Neoplasm Transplantation , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Neoplasms, Experimental/radiotherapy , Nitrobenzenes/therapeutic use , Prostaglandin-Endoperoxide Synthases/genetics , Rats , Sulfonamides/therapeutic use , Time Factors , Transfection , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
J Biol Chem ; 276(47): 44137-45, 2001 Nov 23.
Article in English | MEDLINE | ID: mdl-11551965

ABSTRACT

Infertility and spontaneous pregnancy losses are an enduring problem to women's health. The establishment of pregnancy depends on successful implantation, where a complex series of interactions occurs between the heterogeneous cell types of the uterus and blastocyst. Although a number of genes are implicated in embryo-uterine interactions during implantation, genetic evidence suggests that only a small number of them are critical to this process. To obtain a global view and identify novel pathways of implantation, we used a dual screening strategy to analyze the expression of nearly 10,000 mouse genes by microarray analysis. Comparison of implantation and interimplantation sites by a conservative statistical approach revealed 36 up-regulated genes and 27 down-regulated genes at the implantation site. We also compared the uterine gene expression profile of progesterone-treated, delayed implanting mice to that of mice in which delayed implantation was terminated by estrogen. The results show up-regulation of 128 genes and down-regulation of 101 genes after termination of the delayed implantation. A combined analysis of these experiments showed specific up-regulation of 27 genes both at the implantation site and during uterine activation, representing a broad diversity of molecular functions. In contrast, the majority of genes that were decreased in the combined analysis were related to host immunity or the immune response, suggesting the importance of these genes in regulating the uterine environment for the implanting blastocyst. Collectively, we identified genes with recognized roles in implantation, genes with potential roles in this process, and genes whose functions have yet to be defined in this event. The identification of unique genetic markers for the onset of implantation signifies that genome-wide analysis coupled with functional assays is a promising approach to resolve the molecular pathways required for successful implantation.


Subject(s)
Biomarkers , Embryo Implantation/genetics , Gene Expression Profiling , Uterus/physiology , Animals , Female , Mice , Up-Regulation , Uterus/metabolism
9.
Best Pract Res Clin Gastroenterol ; 15(5): 787-800, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11566041

ABSTRACT

Over the past three decades studies have been conducted to determine the role of prostaglandins in normal physiology and in certain diseases. Cyclo-oxygenase (COX) or prostaglandin endoperoxide synthase (Pghs) is required for the conversion of arachidonic acid to prostaglandins. Two isoforms of this enzyme have been identified which are referred to as COX-1 and COX-2. Under most circumstances, COX-1 is produced constitutively, whereas COX-2 can be induced by several physiological stimuli and is expressed at sites of inflammation. Although these isozymes catalyze identical reactions, they are often regulated by different signalling systems. The goal of this chapter is to provide a review of the role of cyclo-oxygenase in biology and disease, and to summarize the current understanding of mechanisms for the regulation of COX-2 expression.


Subject(s)
Isoenzymes/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Prostaglandins/physiology , Arachidonic Acid/metabolism , Cyclooxygenase 1 , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Cyclooxygenase Inhibitors/therapeutic use , Humans , Membrane Proteins , Peroxidases/metabolism , Prostaglandins/biosynthesis
11.
Oncogene ; 20(33): 4450-6, 2001 Jul 27.
Article in English | MEDLINE | ID: mdl-11494140

ABSTRACT

Numerous reports suggest that use of nonsteroidal anti-inflammatory drugs (NSAIDs) decrease mortality from colorectal cancer. To better understand all of the mechanisms underlying this effect, the global pattern of gene expression in colon carcinoma cells following treatment with NS-398, a selective cyclo-oxygenase-2 inhibitor was evaluated. We utilized suppression subtractive hybridization combined with differential screening to identify genes whose expression was affected following treatment. Among the subtracted cDNA fragments confirmed as differentially expressed, there were two which are known to be involved in the regulation of cell adhesion (human FAT and proto-cadherin-7). We identified two other genes whose levels were decreased and these are known to be involved in the regulation of cell proliferation (cyclin K and p-100). We identified additional genes which are involved in different signaling pathways which regulate programmed cell death (Dynamin 2, Pdcd4 and LIP.1). These results provide evidence that some of the effects of NS-398 on carcinoma cells may be due to modulation of genes which regulate programmed cell death, cell proliferation and cell-cell communication. Additional studies are underway to determine the biological function of the novel genes that were identified.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Colonic Neoplasms/genetics , Cyclooxygenase Inhibitors/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Isoenzymes/antagonists & inhibitors , Neoplasm Proteins/antagonists & inhibitors , Nitrobenzenes/pharmacology , RNA-Binding Proteins , Sulfonamides/pharmacology , Adaptor Proteins, Signal Transducing , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Apoptosis/genetics , Apoptosis Regulatory Proteins , Blotting, Northern , Cadherins/biosynthesis , Cadherins/genetics , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Catenins , Cell Adhesion , Cyclins/biosynthesis , Cyclins/genetics , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , DNA, Complementary/genetics , Dynamin I , Dynamins , GTP Phosphohydrolases/biosynthesis , GTP Phosphohydrolases/genetics , Gene Expression Profiling , Humans , Membrane Proteins , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Phosphoproteins/biosynthesis , Phosphoproteins/genetics , Prostaglandin-Endoperoxide Synthases , Protein Biosynthesis , Proteins/genetics , Protocadherins , RNA, Messenger/genetics , RNA, Neoplasm/genetics , Rectal Neoplasms/genetics , Rectal Neoplasms/metabolism , Rectal Neoplasms/pathology , Signal Transduction/genetics , Substrate Specificity , Subtraction Technique , Tumor Cells, Cultured/drug effects , Delta Catenin
12.
Am J Physiol Gastrointest Liver Physiol ; 281(3): G688-96, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11518681

ABSTRACT

Cyclooxygenase-2 (COX-2) and inducible nitric oxide synthase (iNOS) expression has been demonstrated in inflamed intestinal mucosa. Although regulation of COX-2 and iNOS expression has been studied extensively, the interplay between these two enzymes remains unclear. Because they play crucial roles in inflammation and/or carcinogenesis, we investigated whether COX-2 regulates iNOS expression and evaluated the effects of COX-2 inhibitor and arachidonic acid (AA) on iNOS induction. The COX-2 gene coding region was stably transfected into rat intestinal epithelial cells (RIE sense cells). After interferon-gamma (IFN-gamma) and lipopolysaccharide (LPS) administration, iNOS and COX-2 expression was evaluated by Western blotting. PGE(2) was measured by the enzyme immunoassay (EIA) method. Expression of IFN response factor-1, phosphorylated extracellular signal-related kinase-1 and -2, and Ikappa-Balpha was evaluated. Activator protein-1 and nuclear factor-kappaB (NF-kappaB) were examined by gel mobility shift assay; a supershift assay was performed to identify the NF-kappaB complex components. JTE-522 or AA was added before IFN-gamma and LPS administration, and effects on iNOS and PGE(2) induction were evaluated by Western blotting or EIA. iNOS protein and mRNA expression was inhibited in RIE sense cells. Although NF-kappaB activation was suppressed and Ikappa-Balpha protein was more stable, respectively, in RIE sense cells, no difference was noted in other transcription factors. JTE-522 increased iNOS protein expression in RIE cells. We conclude that COX-2 suppressed iNOS expression in RIE cells through suppression of NF-kappaB by stabilizing Ikappa-Balpha.


Subject(s)
Epithelial Cells/enzymology , I-kappa B Proteins , Intestinal Mucosa/enzymology , Isoenzymes/metabolism , Nitric Oxide Synthase/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Animals , Arachidonic Acid/pharmacology , Benzenesulfonates/pharmacology , Blotting, Western , Cells, Cultured , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Cyclooxygenase Inhibitors/pharmacology , DNA-Binding Proteins/biosynthesis , Dinoprostone/analysis , Dinoprostone/metabolism , Down-Regulation/drug effects , Down-Regulation/physiology , Enzyme Induction/drug effects , Epithelial Cells/cytology , Epithelial Cells/drug effects , Interferon Regulatory Factor-1 , Interferon-gamma/pharmacology , Intestinal Mucosa/cytology , Intestinal Mucosa/drug effects , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Lipopolysaccharides/pharmacology , Mitogen-Activated Protein Kinase 1/biosynthesis , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/biosynthesis , NF-KappaB Inhibitor alpha , NF-kappa B/metabolism , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Oxazoles/pharmacology , Phosphoproteins/biosynthesis , Prostaglandin-Endoperoxide Synthases/genetics , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Rats , Transcription Factor AP-1/metabolism , Transfection
13.
J Biol Chem ; 276(32): 29681-7, 2001 Aug 10.
Article in English | MEDLINE | ID: mdl-11397807

ABSTRACT

Activation of the nuclear hormone peroxisome proliferator-activated receptor gamma (PPARgamma) inhibits cell growth and promotes differentiation in a broad spectrum of epithelial derived tumor cell lines. Here we utilized microarray technology to identify PPARgamma gene targets in intestinal epithelial cells. For each gene, the induction or repression was seen with two structurally distinct PPARgamma agonists, and the change in expression could be blocked by co-treatment with a specific PPARgamma antagonist. A majority of the genes could be regulated independently by a retinoid X receptor specific agonist. Genes implicated in lipid transport or storage (adipophilin and liver fatty acid-binding protein) were also activated by agonists of PPAR subtypes alpha and/or delta. In contrast, PPARgamma-selective targets included genes linked to growth regulatory pathways (regenerating gene IA), colon epithelial cell maturation (GOB-4 and keratin 20), and immune modulation (neutrophil-gelatinase-associated lipocalin). Additionally, three different genes of the carcinoembryonic antigen family were induced by PPARgamma. Cultured cells treated with PPARgamma ligands demonstrated an increase in Ca(2+)-independent, carcinoembryonic antigen-dependent homotypic aggregation, suggesting a potential role for PPARgamma in regulating intercellular adhesion. Collectively, these results will help define the mechanisms by which PPARgamma regulates intestinal epithelial cell biology.


Subject(s)
Colorectal Neoplasms/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Animals , Blotting, Northern , Blotting, Western , COS Cells , Cell Adhesion , Cell Division , Colorectal Neoplasms/genetics , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Epithelial Cells/metabolism , Humans , Intestinal Mucosa/metabolism , Ligands , Luciferases/metabolism , Mutation , Oligonucleotide Array Sequence Analysis , Receptors, Cytoplasmic and Nuclear/physiology , Receptors, Retinoic Acid/agonists , Retinoid X Receptors , Time Factors , Transcription Factors/agonists , Transcription Factors/physiology , Transfection , Tumor Cells, Cultured
14.
J Biol Chem ; 276(33): 31059-66, 2001 Aug 17.
Article in English | MEDLINE | ID: mdl-11397815

ABSTRACT

Helicobacter pylori colonization leads to epithelial cell hyperproliferation within inflamed mucosa, but levels of apoptosis vary, suggesting that imbalances between rates of cell production and loss may contribute to differences in gastric cancer risk among infected populations. Peroxisome proliferator-activated receptor gamma (PPARgamma) regulates inflammatory and growth responses of intestinal epithelial cells. We determined whether activation of PPARgamma modified H. pylori-induced apoptosis in gastric epithelial cells. PPARgamma was expressed and functionally active in gastric epithelial cell lines sensitive to H. pylori-induced apoptosis. PPARgamma ligands 15d-PGJ(2) and BRL-49653 significantly attenuated H. pylomicronri-induced apoptosis, effects that could be reversed by co-treatment with a specific PPARgamma antagonist. Cyclopentanone prostaglandins that do not bind and activate PPARgamma had no effects on H. pylori-induced apoptosis. The ability of H. pylori to activate nuclear factor (NF)-kappaB and increase levels of the NF-kappaB target IL-8 was blocked by co-treatment with PPARgamma agonists, and direct inhibition of NF-kappaB also abolished H. pylori-stimulated apoptosis. These results suggest that activation of the PPARgamma pathway attenuates the ability of H. pylori to induce NF-kappaB-mediated apoptosis in gastric epithelial cells. Because PPARgamma regulates a multitude of host responses, activation of this receptor may contribute to varying levels of cellular turnover as well as the diverse pathologic outcomes associated with chronic H. pylori colonization.


Subject(s)
Apoptosis , Gastric Mucosa/microbiology , Helicobacter pylori/physiology , NF-kappa B/physiology , Receptors, Cytoplasmic and Nuclear/physiology , Thiazolidinediones , Transcription Factors/physiology , Cell Line , Cyclooxygenase 2 , Humans , Isoenzymes/physiology , Membrane Proteins , Prostaglandin-Endoperoxide Synthases/physiology , Rosiglitazone , Species Specificity , Thiazoles/pharmacology
16.
Dig Dis Sci ; 46(4): 785-9, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11330414

ABSTRACT

Barrett's epithelium is a recognized premalignant condition for esophageal adenocarcinoma. Nonsteroidal antiinflammatory drugs (NSAIDs) decrease the relative risk of colon cancer in humans and the esophageal tumor load in carcinogen-treated mice. Previous studies provided conflicting results for COX-2 activity in Barrett's mucosa. Pinch mucosal biopsies were collected from Barrett's and adjacent normal esophageal mucosa from 17 patients with Barrett's esophagus. Low-grade dysplasia was found in seven patients. COX-2 protein was undetectable in normal esophageal mucosa. COX-1 protein expression did not vary between normal and Barrett's epithelium. Increased COX-2 protein was detected in Barrett's epithelium in seven patients (41%) but did not differ with or without dysplasia (43% vs 40%). In conclusion, COX-2 protein is increased in 41% of patients with Barrett's epithelium compared to normal esophageal mucosa but did not differ with or without dysplasia. COX-2 induction may be an early event in the development of Barrett's esophagus.


Subject(s)
Barrett Esophagus/enzymology , Isoenzymes/metabolism , Peroxidases/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Aged , Cyclooxygenase 2 , Humans , Male , Membrane Proteins , Middle Aged
17.
Gastroenterology ; 120(7): 1713-9, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11375952

ABSTRACT

BACKGROUND & AIMS: The cyclooxygenase 2 (COX-2) and ErbB/HER pathways are important modulators of cancer cell growth. We sought to determine the effects of treatment with a specific COX-2 inhibitor and/or a monoclonal antibody against the ErbB receptor subtype HER-2/neu on carcinoma cell growth. METHODS: A cell-proliferation assay was used to determine the response of HCA-7 cells to the HER-3/HER-4 ligand heregulin beta-1 (HRGbeta-1). Both in vitro and in vivo assays were used to determine the effects of the selective COX-2 inhibitor, celecoxib, and/or an anti-HER-2/neu monoclonal antibody (either Herceptin [Genetech Inc., S. San Francisco, CA] or 2C4) on cell growth. RESULTS: HCA-7 cells express HER-2/neu messenger RNA and protein, and exposure of these cells to HRGbeta-1 results in a significant stimulation of cell growth. Celecoxib or Herceptin inhibits HCA-7 cell growth in vitro and in vivo. Combination therapy with celecoxib plus Herceptin or celecoxib plus 2C4 resulted in additive effects that resulted in almost complete inhibition of tumor growth. CONCLUSIONS: Combined treatment with COX-2 and HER-2/neu inhibitors more effectively reduces colorectal carcinoma growth than either agent alone. Therefore, targeting of both the COX-2 and ErbB signaling pathways may represent a novel approach for the treatment and/or prevention of colorectal cancer in humans.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colorectal Neoplasms/drug therapy , Cyclooxygenase Inhibitors/administration & dosage , Isoenzymes/antagonists & inhibitors , Receptor, ErbB-2/antagonists & inhibitors , Sulfonamides/administration & dosage , Animals , Antibodies, Monoclonal, Humanized , Celecoxib , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Female , Humans , Isoenzymes/physiology , Membrane Proteins , Mice , Neoplasm Transplantation , Prostaglandin-Endoperoxide Synthases/physiology , Pyrazoles , Receptor, ErbB-2/physiology , Transplantation, Heterologous , Trastuzumab , Tumor Cells, Cultured
18.
Am J Physiol Gastrointest Liver Physiol ; 280(5): G805-11, 2001 May.
Article in English | MEDLINE | ID: mdl-11292587

ABSTRACT

We have previously demonstrated that after bile duct ligation hepatocytes express Bcl-2, although the mechanisms regulating Bcl-2 expression were not identified. Our aim was to determine if biliary constituents induce hepatocellular expression of Bcl-2 by a cyclooxygenase-2 (COX-2)-dependent mechanism. We used the choledocho-venous fistula (CVF) rat model for these studies and inhibited COX-2 by feeding the animals nimesulide, a selective inhibitor of COX-2 activity. Serum bile acids were 70-fold greater in CVF animals compared with controls, although liver histology and serum alanine aminotransferase values remained normal for the duration of the study. Neither Bcl-2 nor COX-2 was detected in sham-operated animals. However, Bcl-2 was expressed in hepatocytes but not in other liver cells in the CVF animals. In contrast, COX-2 protein was identified in Kupffer cells but not in hepatocytes of CVF animals. Hepatic Bcl-2 protein expression was fourfold lower in the livers from nimesulide-treated CVF rats. In conclusion, high circulating concentrations of biliary constituents are associated with stimulation of de novo hepatocyte expression of Bcl-2 and Kupffer cell expression of COX-2. These data suggest Kupffer cell-derived prostanoids may regulate Bcl-2 expression in the hepatocyte.


Subject(s)
Common Bile Duct/physiology , Gene Expression Regulation/physiology , Genes, bcl-2 , Hepatocytes/physiology , Isoenzymes/metabolism , Kupffer Cells/physiology , Liver/physiology , Prostaglandin-Endoperoxide Synthases/metabolism , Proto-Oncogene Proteins c-bcl-2/analysis , Anastomosis, Surgical , Animals , Bile Acids and Salts/blood , Cell Survival , Cells, Cultured , Common Bile Duct/surgery , Cyclooxygenase 2 , Hepatocytes/cytology , Immunohistochemistry , Kupffer Cells/cytology , Kupffer Cells/enzymology , Liver/cytology , Male , Rats , Rats, Sprague-Dawley , Renal Veins/surgery
19.
Cancer Res ; 61(6): 2670-5, 2001 Mar 15.
Article in English | MEDLINE | ID: mdl-11289146

ABSTRACT

Cyclooxygenase (COX) 2 expression is regulated via the Ras signaling pathway, and induction of mutated Ras rapidly increases COX-2 levels in intestinal epithelial cells. Protein kinase B (Akt/PKB) is an important effector of Ras signaling and a critical component of Ras-mediated transformation. Here we investigate the role of Akt/PKB in K-Ras-mediated induction of COX-2. Rat intestinal epithelial cells (IEC-6) were transfected with an inducible K-RasVal12 cDNA (IEC-iK-Ras cells). Addition of 5 mM isopropyl-1-thio-beta-D-galactopyranoside induced the expression of K-RasVal12, followed by increased activity of extracellular signal-regulated kinase and Akt/PKB. COX-2 levels were dramatically increased after induction of K-RasVal12. Inhibition of MAPK/ERK kinase activity by PD 98059 completely blocked the K-Ras-mediated induction of COX-2, whereas inhibition of PI3K/Akt/PKB activity with LY 294002 or by expressing a dominant negative Akt (Akt-K179M) partially blocked the induction of COX-2 by K-Ras. Transient transfection of cells with phosphatidylinositol 3-kinase and Akt expression vectors revealed that PI3/Akt/PKB activity predominantly regulates the stability of COX-2 mRNA. Thus, Akt/PKB activity is involved in K-Ras-induced expression of COX-2 and stabilization of COX-2 mRNA largely depends on the activation of Akt/PKB.


Subject(s)
Isoenzymes/biosynthesis , Prostaglandin-Endoperoxide Synthases/biosynthesis , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , RNA, Messenger/metabolism , ras Proteins/physiology , Animals , Cell Line , Cyclooxygenase 2 , Enzyme Activation , Enzyme Induction , Epithelial Cells/enzymology , Epithelial Cells/physiology , Gene Expression Regulation, Enzymologic/physiology , Genes, ras , Intestinal Mucosa/cytology , Intestinal Mucosa/enzymology , Intestinal Mucosa/physiology , Isoenzymes/genetics , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinases/biosynthesis , Mitogen-Activated Protein Kinases/metabolism , Prostaglandin-Endoperoxide Synthases/genetics , Protein Serine-Threonine Kinases/biosynthesis , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins c-akt , RNA, Messenger/genetics , Rats , Transfection , ras Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...