Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Cancer Res ; 66(13): 6649-56, 2006 Jul 01.
Article in English | MEDLINE | ID: mdl-16818638

ABSTRACT

Prostaglandin E(2) (PGE(2)), a proinflammatory bioactive lipid, promotes cancer progression by modulating proliferation, apoptosis, and angiogenesis. PGE(2) is a downstream product of cyclooxygenase (COX) and is biochemically inactivated by prostaglandin dehydrogenase (PGDH). In the present study, we investigated the mechanisms by which PGDH is down-regulated in cancer. We show that epidermal growth factor (EGF) represses PGDH expression in colorectal cancer cells. EGF receptor (EGFR) signaling induces Snail, which binds conserved E-box elements in the PGDH promoter to repress transcription. Induction of PGE(2) catabolism through inhibition of EGFR signaling blocks cancer growth in vivo. In human colon cancers, elevated Snail expression correlates well with down-regulation of PGDH. These data indicate that PGDH may serve a tumor suppressor function in colorectal cancer and provide a possible COX-2-independent way to target PGE(2) to inhibit cancer progression.


Subject(s)
Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Dinoprostone/metabolism , Epidermal Growth Factor/pharmacology , Hydroxyprostaglandin Dehydrogenases/biosynthesis , Transcription Factors/biosynthesis , Animals , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/genetics , Disease Progression , Down-Regulation/drug effects , ErbB Receptors/metabolism , HCT116 Cells , HT29 Cells , Humans , Hydroxyprostaglandin Dehydrogenases/genetics , Mice , Mice, Inbred C57BL , Snail Family Transcription Factors , Transcription Factors/genetics , Transfection
2.
J Clin Oncol ; 23(2): 254-66, 2005 Jan 10.
Article in English | MEDLINE | ID: mdl-15637389

ABSTRACT

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 a rapidly evolving aspect of cancer research. Evidence is presented that cyclooxygenase-2 (COX-2) and epidermal growth factor receptor (EGFR) are potential pharmacologic targets to prevent cancer. In this paper, we review key data implicating a causal relationship between COX-2, EGFR, and carcinogenesis and possible mechanisms of action. We discuss evidence of crosstalk between COX-2 and EGFR in order to strengthen the rationale for combination chemoprevention, and review plans for a clinical trial that will evaluate the concept of combination chemoprevention targeting COX-2 and EGFR.


Subject(s)
Anticarcinogenic Agents/pharmacology , Antineoplastic Agents/pharmacology , Drug Delivery Systems , ErbB Receptors/antagonists & inhibitors , Isoenzymes/antagonists & inhibitors , Neoplasms/prevention & control , Clinical Trials as Topic , Cyclooxygenase 2 , Drug Therapy, Combination , ErbB Receptors/physiology , Humans , Isoenzymes/physiology , Membrane Proteins , Models, Biological , Neoplasms/drug therapy , Neoplasms/metabolism , Prostaglandin-Endoperoxide Synthases/physiology , Signal Transduction
3.
Methods Enzymol ; 407: 401-10, 2006.
Article in English | MEDLINE | ID: mdl-16757341

ABSTRACT

Oncogenic mutations in Ras (H-Ras, N-Ras, and K-Ras) are found in a wide variety of human malignancies, including adenocarcinomas of the colon, where K-Ras mutations often occur early in tumor development and strongly correlate with the transition to invasive adenocarcinoma. Our laboratory is interested in examining the interaction between Ras signaling and up-regulation of cyclooxygenase-2 (COX-2), a key regulator of prostaglandin biosynthesis. Our studies demonstrate that the Ras oncoprotein can regulate transcriptional activation and stabilization of COX-2 expression by several mechanisms. In this chapter we have outlined protocols and experimental approaches used in our laboratory to measure H-Ras up-regulation of COX-2 expression and to elaborate on more recent techniques that illustrate the importance of activation of Ras by prostaglandin E2 (PGE(2)). These methods have facilitated our understanding of the mechanisms by which the COX-2-derived PGE(2) and Ras activation of the mitogen-activated protein kinase (MAPK) signaling promotes oncogenic transformation. In light of the critical roles of both COX-2 and Ras signaling in carcinogenesis, our understanding of the complete signaling nuances between different isoforms of Ras on activation of COX-2, as well as understanding the novel mechanism whereby COX-2-derived PGE(2) constitutively activates Ras, will potentially aid in the identification of new targets for cancer therapy.


Subject(s)
Cyclooxygenase 2/metabolism , ras Proteins/physiology , Animals , Cell Line , Dinoprostone/physiology , Enzyme Activation/drug effects , Humans , MAP Kinase Signaling System , Rats , Up-Regulation
4.
Cancer Res ; 68(22): 9331-7, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-19010907

ABSTRACT

Prostaglandin E(2) (PGE(2)) promotes cancer progression by modulating proliferation, apoptosis, angiogenesis, and the immune response. Enzymatic degradation of PGE(2) involves the NAD(+)-dependent 15-hydroxyprostaglandin dehydrogenase (15-PGDH). Recent reports have shown a marked diminution of 15-PGDH expression in colorectal carcinomas (CRC). We report here that treatment of CRC cells with histone deacetylase (HDAC) inhibitors, including sodium butyrate and valproic acid, induces 15-PGDH expression. Additionally, we show that pretreatment of CRC cells with HDAC inhibitors can block epidermal growth factor-mediated or Snail-mediated transcriptional repression of 15-PGDH. We show an interaction between Snail and HDAC2 and the binding of HDAC2 to the 15-PGDH promoter. In vivo, we observe increased Hdac2 expression in Apc-deficient mouse adenomas, which inversely correlated with loss of 15-Pgdh expression. Finally, in human colon cancers, elevated HDAC expression correlated with down-regulation of 15-PGDH. These data suggest that class I HDACs, specifically HDAC2, and the transcriptional repressor Snail play a central role in the suppression of 15-PGDH expression. These results also provide a cyclooxygenase-2-independent mechanism to explain increased PGE(2) levels that contribute to progression of CRC.


Subject(s)
Colonic Neoplasms/enzymology , Histone Deacetylases/physiology , Hydroxyprostaglandin Dehydrogenases/genetics , Repressor Proteins/physiology , Transcription Factors/physiology , Animals , Enzyme Inhibitors/pharmacology , Epigenesis, Genetic , Histone Deacetylase 2 , Histone Deacetylase Inhibitors , Histone Deacetylases/genetics , Humans , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Snail Family Transcription Factors
5.
J Biol Chem ; 281(5): 2676-82, 2006 Feb 03.
Article in English | MEDLINE | ID: mdl-16293616

ABSTRACT

Many lines of research implicate cyclooxygenase 2-derived prostaglandins in tumor growth and metastasis. More specifically, we have shown that prostaglandin E2 (PGE2) promotes cell proliferation and invasion through transactivation of the epidermal growth factor receptor, initiates immune evasion through induction of decay accelerating factor, and transactivates peroxisome proliferator-activated receptor delta, leading to increased polyp size and multiplicity. We continue to identify novel PGE2 target genes in colorectal carcinoma cells and report here that an immediate early gene, nuclear factor NR4A2 (Nurr1), is induced by PGE2 that in turn regulates cell death. Originally described as a critical dopaminergic neuron growth factor receptor, NR4A2 expression is rapidly but transiently induced by PGE2 in a cAMP/protein kinase A-dependent manner. NR4A2 binds to the cognate NBRE response element and enhances transcription of a reporter construct in colorectal carcinoma cells. Furthermore, NR4A2 expression is elevated in Apc-/+ mouse adenomas and its levels were further increased following PGE2 treatment. Human colorectal cancers relative to matched normal mucosa showed increased NR4A2 expression. Although not previously described in epithelial tissues, NR4A2 protein localizes to proliferating crypts of Apc-/+ mouse intestine. Finally, functional studies reveal that PGE2-mediated protection from apoptosis is completely inhibited by a dominant-negative NR4A2 construct. Building on previous reports from our group on the peroxisome proliferator-activated receptor family of nuclear receptors, these most recent data suggest that NR4A2, a member of another family of nuclear receptors can stimulate progression of colorectal cancer downstream from cyclooxygenase 2-derived PGE2.


Subject(s)
Colorectal Neoplasms/pathology , DNA-Binding Proteins/genetics , Dinoprostone/physiology , Gene Expression Regulation, Neoplastic , Transcription Factors/genetics , Animals , Cell Line, Tumor , Colorectal Neoplasms/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/physiology , Disease Progression , Humans , Intestinal Mucosa/metabolism , Mice , Mice, Knockout , Nuclear Receptor Subfamily 4, Group A, Member 2 , Protein Transport , Transcription Factors/metabolism , Transcription Factors/physiology
6.
Gastroenterology ; 128(5): 1445-61, 2005 May.
Article in English | MEDLINE | ID: mdl-15887126

ABSTRACT

Nonsteroidal anti-inflammatory drugs (NSAIDs) are generally prescribed to ameliorate symptoms associated with acute pain and chronic inflammatory diseases such as arthritis. Recent epidemiologic studies and clinical trials indicate that use of NSAIDs and cyclooxygenase (COX)-2 selective inhibitors are associated with a reduced risk of certain malignancies, especially gastrointestinal cancer. The cyclooxygenase enzymes are the best known targets of NSAIDs; this diverse class of compounds blocks conversion of arachidonic acid to prostanoids. Prostaglandins and other eicosanoids derived from COX-1 and COX-2 are involved in a variety of physiologic and pathologic processes in the gastrointestinal tract. Recent efforts to identify the molecular mechanisms by which COX-2-derived prostanoids exert their proneoplastic effects have provided a rationale for the possible use of NSAIDs alone or in a combination with conventional or experimental anticancer agents for the treatment or prevention of gastrointestinal cancers.


Subject(s)
Eicosanoids/physiology , Gastrointestinal Neoplasms/physiopathology , Gastrointestinal Tract/physiology , Prostaglandins/physiology , Animals , Humans
7.
Nat Clin Pract Oncol ; 2(4): 202-10, 2005 Apr.
Article in English | MEDLINE | ID: mdl-16264935

ABSTRACT

Discovery of molecular pathways critical to carcinogenesis is revolutionizing the treatment and prevention of cancer. Traditional chemotherapeutic approaches usually cause 'global' cytotoxicity to both normal and carcinoma cells. Over the past decade, however, investigators have developed compounds that inhibit tumor formation more selectively by targeting specific signaling pathways, including those involving the epidermal growth factor receptor (EGFR) and cyclooxygenase 2 (COX2). COX2-derived bioactive lipids, including prostaglandin E2, are potent inflammatory mediators that promote tumor growth and metastasis through stimulation of cell proliferation, invasion, and angiogenesis. Recent work has demonstrated significant crosstalk between the COX2 and EGFR pathways, while preclinical data demonstrates a synergistic effect when both pathways are targeted simultaneously. Combination therapy, a common strategy in cancer treatment, is likely to improve outcomes in cancer prevention as well. Ongoing clinical trials designed to assess whether low doses of COX2 and EGFR inhibitors used in combination could prove more effective and result in reduced toxicity than either agent alone may provide new options for cancer prevention and treatment. We discuss advances in cancer prevention by focusing on mechanisms by which bioactive lipids contribute to tumor formation. While cancer chemoprevention is a relatively young field, we argue that this approach to malignant disease bears significant potential.


Subject(s)
Inflammation Mediators/metabolism , Neoplasms/prevention & control , Humans
8.
Oncology ; 69 Suppl 1: 28-32, 2005.
Article in English | MEDLINE | ID: mdl-16210874

ABSTRACT

Carcinoma of the colon or rectum represents one of the most common malignancies worldwide with a higher prevalence in industrialized regions. Epidemiologic studies of individuals taking non-steroidal anti-inflammatory drugs (NSAIDs) have shown a significant reduction in colorectal cancer (CRC) mortality compared to those individuals not receiving these agents. NSAIDs inhibit the enzymatic activity of both isoforms of cyclooxygenase (COX-1 and COX-2), while COX-2-selective inhibitors have shown some efficacy in reducing polyp formation. COX-2-derived bioactive lipids, including the primary prostaglandin (PG) generated in colorectal tumors, PGE(2), are known to stimulate cell migration, proliferation and tumor-associated neovascularization while inhibiting cell death. Here we briefly review the role of NSAIDs in preventing CRC, as well as the proposed mechanism by which a COX-2-derived PG, PGE(2), promotes colon cancer.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Colorectal Neoplasms/prevention & control , Dinoprostone/physiology , Animals , Antineoplastic Agents , Arachidonic Acid/metabolism , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Cyclooxygenase Inhibitors , Humans , Hydroxyprostaglandin Dehydrogenases/metabolism , Membrane Proteins , Prostaglandin-Endoperoxide Synthases/metabolism
9.
J Biol Chem ; 280(1): 476-83, 2005 Jan 07.
Article in English | MEDLINE | ID: mdl-15520008

ABSTRACT

Cyclooxygenase-derived prostaglandin E(2) (PGE(2)) stimulates tumor progression by modulating several proneoplastic pathways. The mechanisms by which PGE(2) promotes tumor growth and metastasis through stimulation of cell migration, invasion, and angiogenesis have been fairly well characterized. Much less is known, however, about the molecular mechanisms responsible for the immunosuppressive effects of PGE(2). We identified PGE(2) target genes and subsequently studied their biologic role in colorectal cancer cells. The complement regulatory protein decay-accelerating factor (DAF or CD55) was induced following PGE(2) treatment of LS174T colon cancer cells. Analysis of PGE(2)-mediated activation of the DAF promoter employing 5'-deletion luciferase constructs suggests that regulation occurs at the transcriptional level via a cyclic AMP/protein kinase A-dependent pathway. Nonsteroidal anti-inflammatory drugs blocked DAF expression in HCA-7 colon cancer cells, which could be restored by the addition of exogenous PGE(2). Finally, we observed an increase in DAF expression in the intestinal mucosa of Apc(Min+/-) mice treated with PGE(2) in vivo. In summary, these results indicate a novel immunosuppressive role for PGE(2) in the development of colorectal carcinomas.


Subject(s)
CD55 Antigens/metabolism , Colorectal Neoplasms/metabolism , Dinoprostone/metabolism , Gene Expression Regulation, Neoplastic , Animals , CD55 Antigens/genetics , Cell Line, Tumor , Colorectal Neoplasms/etiology , Cyclic AMP-Dependent Protein Kinases/metabolism , Dinoprostone/genetics , Dinoprostone/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice , Promoter Regions, Genetic , Sequence Deletion , Signal Transduction , Transcriptional Activation/drug effects
10.
J Biol Chem ; 280(5): 3217-23, 2005 Feb 04.
Article in English | MEDLINE | ID: mdl-15542609

ABSTRACT

Prostaglandin E2 (PGE2) can stimulate tumor progression by modulating several proneoplastic pathways, including proliferation, angiogenesis, cell migration, invasion, and apoptosis. Although steady-state tissue levels of PGE2 stem from relative rates of biosynthesis and breakdown, most reports examining PGE2 have focused solely on the cyclooxygenase-dependent formation of this bioactive lipid. Enzymatic degradation of PGE2 involves the NAD+-dependent 15-hydroxyprostaglandin dehydrogenase (15-PGDH). The present study examined a range of normal tissues in the human and mouse and found high levels of 15-PGDH in the large intestine. By contrast, the expression of 15-PGDH is decreased in several colorectal carcinoma cell lines and in other human malignancies such as breast and lung carcinomas. Consistent with these findings, we observe diminished 15-Pgdh expression in ApcMin+/- mouse adenomas. Enzymatic activity of 15-PGDH correlates with expression levels and the genetic disruption of 15-Pgdh completely blocks production of the urinary PGE2 metabolite. Finally, 15-PGDH expression and activity are significantly down-regulated in human colorectal carcinomas relative to matched normal tissue. In summary, these results suggest a novel tumor suppressive role for 15-PGDH due to loss of expression during colorectal tumor progression.


Subject(s)
Colorectal Neoplasms/metabolism , Colorectal Neoplasms/physiopathology , Hydroxyprostaglandin Dehydrogenases/genetics , Hydroxyprostaglandin Dehydrogenases/metabolism , Animals , Colon/enzymology , Cyclooxygenase 2 , Dinoprostone/metabolism , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Membrane Proteins , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Prostaglandin-Endoperoxide Synthases/metabolism , Tumor Cells, Cultured
11.
Cancer J ; 10(3): 145-52, 2004.
Article in English | MEDLINE | ID: mdl-15285921

ABSTRACT

The cyclooxygenase (COX) enzymes (COX-1 and COX-2) are key enzymes of prostaglandin (PG) biosynthesis. Nonselective non-steroidal anti-inflammatory drugs (NSAIDs) inhibit the enzymatic activity of both COX-1 and COX-2. Selective COX-2 inhibitors have been developed that appear to have 50% less gastrointestinal toxicity than traditional nonselective NSAIDs. Experimental evidence suggests that the COX pathway is involved in tumor promotion. Evidence to support this comes from both clinical and laboratory findings suggesting that chronic NSAID use reduces the relative risk for developing colorectal cancer (CRC). Although the precise mechanism or mechanisms by which these drugs affect tumor progression is not completely understood, it is likely that part of their anti-tumor effect is due to inhibition of the COX- 2 enzyme. COX-2 levels are increased in CRC as well as in several other solid malignancies. COX-2-derived bioactive lipid products promote tumor-associated n eovascularization, inhibit cell death, and stimulate cell proliferation and motility. Additionally, treatment with COX-2-selective inhibitors reduces polyp burden in animal models of intestinal neoplasia and in humans with familial adenomatous polyposis (FAP). Ongoing human clinical trails are under way to test the efficacy of COX-2-selective inhibitors in a number of human cancers.


Subject(s)
Colorectal Neoplasms/enzymology , Isoenzymes/drug effects , Prostaglandin-Endoperoxide Synthases/drug effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Chemoprevention , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Cyclooxygenase Inhibitors/pharmacology , Cyclooxygenase Inhibitors/therapeutic use , Drug Delivery Systems , Gene Expression Regulation, Enzymologic , Humans , Isoenzymes/metabolism , Membrane Proteins , Prostaglandin-Endoperoxide Synthases/metabolism , Prostaglandins/biosynthesis
12.
Cell Cycle ; 3(12): 1512-5, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15539957

ABSTRACT

Both Wnt and cyclooxygenase (COX-2) pathways are activated in most sporadic and familial colorectal cancers, especially in those with chromosomal instability. We have recently shown that a common target of both signaling pathways, the peroxisome proliferator-activated receptor (PPAR)-delta, is involved in intestinal adenoma growth. Activation of this receptor by synthetic agonist (GW501516) or COX-2-derived prostaglandin E2 (PGE2) accelerates intestinal adenoma growth in Apc(Min) mice. Moreover, these effects are lost in Apc(Min) mice lacking PPARdelta. These findings implicate PPARdelta as a focal point of cross-talk between the Wnt and prostaglandin signaling pathways. Based on this work it looks as if PPARdelta agonists currently in development for treatment of dyslipidemias and obesity may increase the risk of tumor formation in humans. By contrast, antagonists of PPARdelta may provide a novel approach for prevention and treatment of colorectal cancer.


Subject(s)
Adenoma/pathology , Cyclooxygenase 2/metabolism , Wnt Proteins/metabolism , Animals , Cell Proliferation , Dinoprostone/metabolism , Humans , PPAR delta/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL