101
|
NO-Donating NSAIDs, PPARdelta, and Cancer: Does PPARdelta Contribute to Colon Carcinogenesis? PPAR Res 2011; 2008:919572. [PMID: 18528523 PMCID: PMC2408682 DOI: 10.1155/2008/919572] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2008] [Revised: 04/25/2008] [Accepted: 05/06/2008] [Indexed: 12/30/2022] Open
Abstract
The chemopreventive NO-donating NSAIDs (NO-NSAIDs; NSAIDs with an NO-releasing moiety) modulate PPARδ and offer the opportunity to revisit the controversial role of PPARδ in carcinogenesis (several papers report that PPARδ either promotes or inhibits cancer). This review summarizes the pharmacology of NO-NSAIDs, PPARδ cancer biology, and the relationship between the two. In particular, a study of the chemopreventive effect of two isomers of NO-aspirin on intestinal neoplasia in Min mice showed that, compared to wild-type controls, PPARδ is overexpressed in the intestinal mucosa of Min mice; PPARδ responds to
m- and p-NO-ASA proportionally to their antitumor effect (p- > m-). This effect is accompanied by the induction of epithelial cell death, which correlates with the antineoplastic effect of NO-aspirin; and NO-aspirin's effect on PPARδ is specific (no changes in PPARα or PPARγ). Although these data support the notion that PPARδ promotes intestinal carcinogenesis and its inhibition could be therapeutically useful, more work is needed before a firm conclusion is reached.
Collapse
|
102
|
PPARgamma and PPARdelta as Modulators of Neoplasia and Cell Fate. PPAR Res 2011; 2008:247379. [PMID: 18566686 PMCID: PMC2430014 DOI: 10.1155/2008/247379] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2008] [Accepted: 05/22/2008] [Indexed: 12/25/2022] Open
Abstract
PPARγ and PPARδ agonists represent unique classes of drugs that act through their ability to modulate gene transcription associated with intermediary metabolism, differentiation, tumor suppression, and in some instances proliferation and cell adhesion. PPARγ agonists are used by millions of people each year to treat type 2 diabetes but may also find additional utility as relatively nontoxic potentiators of chemotherapy. PPARδ agonists produce complex actions as shown by their tumor promoting effects in rodents and their cholesterol-lowering action in dyslipidemias. There is now emerging evidence that PPARs regulate tumor suppressor genes and developmental pathways associated with transformation and cell fate determination. This review discusses the role of PPARγ and PPARδ agonists as modulators of these processes.
Collapse
|
103
|
Peroxisome proliferator-activated receptors and progression of colorectal cancer. PPAR Res 2011; 2008:931074. [PMID: 18551185 PMCID: PMC2422873 DOI: 10.1155/2008/931074] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2008] [Accepted: 04/29/2008] [Indexed: 12/18/2022] Open
Abstract
The peroxisome proliferator-activated receptors (PPARs) are members of the nuclear hormone receptor superfamily. These receptors are also ligand-dependent transcription factors responsible for the regulation of cellular events that range from glucose and lipid homeostases to cell differentiation and apoptosis. The importance of these receptors in lipid homeostasis and energy balance is well established. In addition to these metabolic and anti-inflammatory properties, emerging evidence indicates that PPARs can function as either tumor suppressors or accelerators, suggesting that these receptors are potential candidates as drug targets for cancer prevention and treatment. However, conflicting results have emerged regarding the role of PPARs on colon carcinogenesis. Therefore, further investigation is warranted prior to considering modulation of PPARs as an efficacious therapy for colorectal cancer chemoprevention and treatment.
Collapse
|
104
|
Multiple Interactions between Peroxisome Proliferators-Activated Receptors and the Ubiquitin-Proteasome System and Implications for Cancer Pathogenesis. PPAR Res 2011; 2008:195065. [PMID: 18551186 PMCID: PMC2423003 DOI: 10.1155/2008/195065] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2008] [Accepted: 04/29/2008] [Indexed: 12/31/2022] Open
Abstract
The peroxisome proliferator-activated receptors (PPAR) α, β/δ, and γ are ligand-activated nuclear receptors involved in a number of physiological processes, including lipid and glucose homeostasis, inflammation, cell growth, differentiation, and death. PPAR agonists are used in the treatment of human diseases, like type 2 diabetes and dyslipidemia, and PPARs appear as promising therapeutic targets in other conditions, including cancer. A better understanding of the functions and regulation of PPARs in normal and pathological processes is of primary importance to devise appropriate therapeutic strategies. The ubiquitin-proteasome system (UPS) plays an important role in controlling level and activity of many nuclear receptors and transcription factors. PPARs are subjected to UPS-dependent regulation. Interestingly, the three PPAR isotypes are differentially regulated by the UPS in response to ligand-dependent activation, a phenomenon that may be intrinsically connected to their distinct cellular functions and behaviors. In addition to their effects ongene expression, PPARs appear to affect protein levels and downstream pathways also by modulating the activity of the UPS in target-specific manners. Here we review the current knowledge of the interactions between the UPS and PPARs in light of the potential implications for their effects on cell fate and tumorigenesis.
Collapse
|
105
|
Abstract
Peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors that belong to the nuclear hormone receptor superfamily. PPARalpha is mainly expressed in the liver, where it activates fatty acid catabolism. PPARalpha activators have been used to treat dyslipidemia, causing a reduction in plasma triglyceride and elevation of high-density lipoprotein cholesterol. PPARdelta is expressed ubiquitously and is implicated in fatty acid oxidation and keratinocyte differentiation. PPARdelta activators have been proposed for the treatment of metabolic disease. PPARgamma2 is expressed exclusively in adipose tissue and plays a pivotal role in adipocyte differentiation. PPARgamma is involved in glucose metabolism through the improvement of insulin sensitivity and represents a potential therapeutic target of type 2 diabetes. Thus PPARs are molecular targets for the development of drugs treating metabolic syndrome. However, PPARs also play a role in the regulation of cancer cell growth. Here, we review the function of PPARs in tumor growth.
Collapse
|
106
|
|
107
|
Keith RL, Blatchford PJ, Kittelson J, Minna JD, Kelly K, Massion PP, Franklin WA, Mao J, Wilson DO, Merrick DT, Hirsch FR, Kennedy TC, Bunn PA, Geraci MW, Miller YE. Oral iloprost improves endobronchial dysplasia in former smokers. Cancer Prev Res (Phila) 2011; 4:793-802. [PMID: 21636546 PMCID: PMC3251330 DOI: 10.1158/1940-6207.capr-11-0057] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
There are no established chemopreventive agents for lung cancer, the leading cause of cancer death in the United States. Prostacyclin levels are low in lung cancer and supplementation prevents lung cancer in preclinical models. We carried out a multicenter double-blind, randomized, phase II placebo-controlled trial of oral iloprost in current or former smokers with sputum cytologic atypia or endobronchial dysplasia. Bronchoscopy was performed at study entry and after completion of six months of therapy. Within each subject, the results were calculated by using the average score of all biopsies (Avg), the worst biopsy score (Max), and the dysplasia index (DI). Change in Avg was the primary end point, evaluated in all subjects, as well as in current and former smokers. The accrual goal of 152 subjects was reached and 125 completed both bronchoscopies (60/75 iloprost, 65/77 placebo). Treatment groups were well matched for age, tobacco exposure, and baseline histology. Baseline histology was significantly worse for current smokers (Avg 3.0) than former smokers (Avg 2.1). When compared with placebo, former smokers receiving oral iloprost exhibited a significantly greater improvement in Avg (0.41 units better, P = 0.010), in Max (1.10 units better, P = 0.002), and in DI (12.45%, P = 0.006). No histologic improvement occurred in current smokers. Oral iloprost significantly improves endobronchial histology in former smokers and deserves further study to determine if it can prevent the development of lung cancer.
Collapse
Affiliation(s)
- Robert L Keith
- Division of Pulmonary Sciences and Critical Care Medicine, Eastern Colorado VA Healthcare System, University of Colorado Denver, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
108
|
Yoshinaga M, Taki K, Somada S, Sakiyama Y, Kubo N, Kaku T, Tsuruta S, Kusumoto T, Sakai H, Nakamura K, Takayanagi R, Muto Y. The expression of both peroxisome proliferator-activated receptor delta and cyclooxygenase-2 in tissues is associated with poor prognosis in colorectal cancer patients. Dig Dis Sci 2011; 56:1194-200. [PMID: 20824502 DOI: 10.1007/s10620-010-1389-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2009] [Accepted: 08/05/2010] [Indexed: 01/31/2023]
Abstract
BACKGROUND The role of peroxisome proliferator-activated receptor delta (PPAR δ) in the development and progression of colorectal cancer (CRC) remains controversial. AIMS We investigated the impact of PPAR δ expression in tissues on liver metastasis of CRC. METHODS We analyzed samples of primary CRC and matched normal adjacent tissues from 52 patients for the expression of PPAR δ, cyclooxygenase (COX)-2, vascular endothelial growth factor (VEGF)-A, and CXC chemokine receptor 4 (CXCR4). Correlations of the molecules expressions with clinical characteristics and prognosis of patients were studied. RESULTS The number of patients positive for PPAR δ, COX-2, CXCR4, and VEGF-A was 25, 33, 18, and 19, respectively. Among the PPAR δ (+)/COX-2 (+), PPAR δ (-)/COX-2 (+), PPAR δ (+)/COX-2 (-), and PPAR δ (-)/COX-2 (-) patient groups, PPAR δ (+)/COX-2 (+) patients had the highest incidence of liver metastasis (p<0.01). PPAR δ (+)/COX-2 (+) expression was a significant independent prognostic factor (HR=7.108, 95% CI 1.231-41.029, p=0.0283) by Cox proportional analysis. PPAR δ (+)/COX-2 (+) patients had the highest positivity for CXCR4 or VEGF-A in tissues (p<0.01). Among the patients in the CXCR4 (+)/VEGF-A (+), CXCR4 (+)/VEGF-A (-), CXCR4 (-)/VEGF-A (+), and CXCR4 (-)/VEGF-A (-) groups, CXCR4 (+)/VEGF-A (+) patients had the highest incidence of liver metastasis (p<0.01). CONCLUSIONS The expression of both PPAR δ and COX-2 in tissues may lead to liver metastasis and consequent poor prognosis in CRC patients.
Collapse
Affiliation(s)
- Masahiro Yoshinaga
- Department of Gastroenterology, Hepatology, and Nutrition, National Hospital Organization Beppu Medical Center, 1473 Uchikamado, Beppu, Oita, 874-0011, Japan.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
109
|
Omiecinski CJ, Vanden Heuvel JP, Perdew GH, Peters JM. Xenobiotic metabolism, disposition, and regulation by receptors: from biochemical phenomenon to predictors of major toxicities. Toxicol Sci 2011; 120 Suppl 1:S49-75. [PMID: 21059794 PMCID: PMC3145385 DOI: 10.1093/toxsci/kfq338] [Citation(s) in RCA: 243] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2010] [Accepted: 11/01/2010] [Indexed: 02/07/2023] Open
Abstract
To commemorate the 50th anniversary of the Society of Toxicology, this special edition article reviews the history and current scope of xenobiotic metabolism and transport, with special emphasis on the discoveries and impact of selected "xenobiotic receptors." This overall research realm has witnessed dynamic development in the past 50 years, and several of the key milestone events that mark the impressive progress in these areas of toxicological sciences are highlighted. From the initial observations regarding aspects of drug metabolism dating from the mid- to late 1800's, the area of biotransformation research witnessed seminal discoveries in the mid-1900's and onward that are remarkable in retrospect, including the discovery and characterization of the phase I monooxygenases, the cytochrome P450s. Further research uncovered many aspects of the biochemistry of xenobiotic metabolism, expanding to phase II conjugation and phase III xenobiotic transport. This led to hallmark developments involving integration of genomic technologies to elucidate the basis for interindividual differences in response to xenobiotic exposures and discovery of nuclear and soluble receptor families that selectively "sense" the chemical milieu of the mammalian cell and orchestrate compensatory changes in gene expression programming to accommodate complex xenobiotic exposures. This review will briefly summarize these developments and investigate the expanding roles of xenobiotic receptor biology in the underlying basis of toxicological response to chemical agents.
Collapse
Affiliation(s)
- Curtis J Omiecinski
- Department of Veterinary and Biomedical Sciences, Center for Molecular Toxicology and Carcinogenesis, Penn State University, University Park, Pennsylvania 16802, USA.
| | | | | | | |
Collapse
|
110
|
Green DE, Sutliff RL, Hart CM. Is peroxisome proliferator-activated receptor gamma (PPARγ) a therapeutic target for the treatment of pulmonary hypertension? Pulm Circ 2011; 1:33-47. [PMID: 21547012 PMCID: PMC3085428 DOI: 10.4103/2045-8932.78101] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Pulmonary hypertension (PH), a progressive disorder associated with significant morbidity and mortality, is caused by complex pathways that culminate in structural and functional alterations of the pulmonary circulation and increases in pulmonary vascular resistance and pressure. Diverse genetic, pathological, or environmental triggers stimulate PH pathogenesis culminating in vasoconstriction, cell proliferation, vascular remodeling, and thrombosis. We conducted a thorough literature review by performing MEDLINE searches via PubMed to identify articles pertaining to PPARγ as a therapeutic target for the treatment of PH. This review examines basic and preclinical studies that explore PPARγ and its ability to regulate PH pathogenesis. Despite the current therapies that target specific pathways in PH pathogenesis, including prostacyclin derivatives, endothelin-receptor antagonists, and phosphodiesterase type 5 inhibitors, morbidity and mortality related to PH remain unacceptably high, indicating the need for novel therapeutic approaches. Consequently, therapeutic targets that simultaneously regulate multiple pathways involved in PH pathogenesis have gained attention. This review focuses on peroxisome proliferator-activated receptor gamma (PPARγ), a member of the nuclear hormone receptor superfamily of ligand-activated transcription factors. While the PPARγ receptor is best known as a master regulator of lipid and glucose metabolism, a growing body of literature demonstrates that activation of PPARγ exerts antiproliferative, antithrombotic, and vasodilatory effects on the vasculature, suggesting its potential efficacy as a PH therapeutic target.
Collapse
Affiliation(s)
- David E. Green
- Department of Medicine, Emory University, Atlanta Veterans Affairs Medical Center, Decatur, Georgia, USA
| | - Roy L. Sutliff
- Department of Medicine, Emory University, Atlanta Veterans Affairs Medical Center, Decatur, Georgia, USA
| | - C. Micheal Hart
- Department of Medicine, Emory University, Atlanta Veterans Affairs Medical Center, Decatur, Georgia, USA
| |
Collapse
|
111
|
Pollock CB, Rodriguez O, Martin PL, Albanese C, Li X, Kopelovich L, Glazer RI. Induction of metastatic gastric cancer by peroxisome proliferator-activated receptorδ activation. PPAR Res 2010; 2010:571783. [PMID: 21318167 PMCID: PMC3026990 DOI: 10.1155/2010/571783] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2010] [Accepted: 11/16/2010] [Indexed: 01/24/2023] Open
Abstract
Peroxisome proliferator-activated receptorδ (PPARδ) regulates a multiplicity of physiological processes associated with glucose and lipid metabolism, inflammation, and proliferation. One or more of these processes likely create risk factors associated with the ability of PPARδ agonists to promote tumorigenesis in some organs. In the present study, we describe a new gastric tumor mouse model that is dependent on the potent and highly selective PPARδ agonist GW501516 following carcinogen administration. The progression of gastric tumorigenesis was rapid as determined by magnetic resonance imaging and resulted in highly metastatic squamous cell carcinomas of the forestomach within two months. Tumorigenesis was associated with gene expression signatures indicative of cell adhesion, invasion, inflammation, and metabolism. Increased PPARδ expression in tumors correlated with increased PDK1, Akt, β-catenin, and S100A9 expression. The rapid development of metastatic gastric tumors in this model will be useful for evaluating preventive and therapeutic interventions in this disease.
Collapse
Affiliation(s)
- Claire B. Pollock
- Department of Oncology, Lombardi Comprehensive Cancer Center, Washington, DC 20057, USA
| | - Olga Rodriguez
- Department of Oncology, Lombardi Comprehensive Cancer Center, Washington, DC 20057, USA
| | - Philip L. Martin
- Center for Advanced Preclinical Research, SAIC/NCI-Frederick, Frederick, MD 21702, USA
| | - Chris Albanese
- Department of Oncology, Lombardi Comprehensive Cancer Center, Washington, DC 20057, USA
| | - Xin Li
- Department of Biostatistics, Bioinformatics, and Biomathematics, Lombardi Comprehensive Cancer Center, Washington, DC 20057, USA
| | - Levy Kopelovich
- Chemoprevention Agent Development and Research Group, Division of Cancer Prevention, National Cancer Institute, Bethesda, MD 20814, USA
| | - Robert I. Glazer
- Department of Oncology, Lombardi Comprehensive Cancer Center, Washington, DC 20057, USA
| |
Collapse
|
112
|
Sutliff RL, Kang BY, Hart CM. PPARgamma as a potential therapeutic target in pulmonary hypertension. Ther Adv Respir Dis 2010; 4:143-60. [PMID: 20530063 DOI: 10.1177/1753465809369619] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Pulmonary hypertension (PH) is a progressive disorder of the pulmonary circulation associated with significant morbidity and mortality. The pathobiology of PH involves a complex series of derangements causing endothelial dysfunction, vasoconstriction and abnormal proliferation of pulmonary vascular wall cells that lead to increases in pulmonary vascular resistance and pressure. Recent evidence indicates that the ligand-activated transcription factor, peroxisome proliferator-activated receptor gamma (PPARgamma) can have a favorable impact on a variety of pathways involved in the pathogenesis of PH. This review summarizes PPARgamma biology and the emerging evidence that therapies designed to activate this receptor may provide novel approaches to the treatment of PH. Mediators of PH that are regulated by PPARgamma are reviewed to provide insights into potential mechanisms underlying therapeutic effects of PPARgamma ligands in PH.
Collapse
Affiliation(s)
- Roy L Sutliff
- Division of Pulmonary, Allergy and Critical Care Medicine, Atlanta VA Medical Center, Decatur, GA 30033, USA.
| | | | | |
Collapse
|
113
|
Wang D, DuBois RN. Therapeutic potential of peroxisome proliferator-activated receptors in chronic inflammation and colorectal cancer. Gastroenterol Clin North Am 2010; 39:697-707. [PMID: 20951925 PMCID: PMC2957674 DOI: 10.1016/j.gtc.2010.08.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Peroxisome proliferatoreactivated receptors (PPARs) are members of the nuclear hormone receptor superfamily and have been implicated in a variety of physiologic and pathologic processes, such as nutrient metabolism, energy homeostasis, inflammation, and cancer. This article highlights breakthroughs in our understanding of the potential roles of PPARs in inflammatory bowel disease and colorectal cancer. PPARs might hold the key to some of the questions that are pertinent to the pathophysiology of inflammatory diseases and colorectal cancer and could possibly serve as drug targets for new antiinflammatory therapeutic and anticancer agents.
Collapse
Affiliation(s)
- Dingzhi Wang
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Unit 1014; S7.8316A, 6767 Bertner Avenue, Houston, TX 77030, USA
| | | |
Collapse
|
114
|
Peroxisome Proliferator-Activated Receptors Protect against Apoptosis via 14-3-3. PPAR Res 2010; 2010. [PMID: 20862376 PMCID: PMC2938460 DOI: 10.1155/2010/417646] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2010] [Accepted: 07/14/2010] [Indexed: 01/17/2023] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) were reported to prevent cells from stress-induced apoptosis and protect tissues against ischemia-reperfusion injury. The underlying transcriptional mechanism is unclear. Recent reports indicate that the antiapoptotic actions of ligand-activated PPARδ and PPARγ are mediated through enhanced binding of PPAR to the promoter of 14-3-3ε and upregulation of 14-3-3ε expression. We propose that ligand-activated PPARα exerts its anti-apoptotic actions via the identical pathway. The PPAR to 14-3-3 transcriptional axis plays an important role in protection of cell and tissue integrity and is a target for drug discovery.
Collapse
|
115
|
Prostacyclin inhibits non-small cell lung cancer growth by a frizzled 9-dependent pathway that is blocked by secreted frizzled-related protein 1. Neoplasia 2010; 12:244-53. [PMID: 20234818 DOI: 10.1593/neo.91690] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2009] [Revised: 01/04/2010] [Accepted: 01/05/2010] [Indexed: 12/14/2022] Open
Abstract
The goal of this study was to assess the ability of iloprost, an orally active prostacyclin analog, to inhibit transformed growth of human non-small cell lung cancer (NSCLC) and to define the mechanism of iloprost's tumor suppressive effects. In a panel of NSCLC cell lines, the ability of iloprost to inhibit transformed cell growth was not correlated with the expression of the cell surface receptor for prostacyclin, but instead was correlated with the presence of Frizzled 9 (Fzd 9) and the activation of peroxisome proliferator-activated receptor-gamma (PPARgamma). Silencing of Fzd 9 blocked PPARgamma activation by iloprost, and expression of Fzd 9 in cells lacking the protein resulted in iloprost's activation of PPARgamma and inhibition of transformed growth. Interestingly, soluble Frizzled-related protein-1, a well-known inhibitor of Wnt/Fzd signaling, also blocked the effects of iloprost and Fzd 9. Moreover, mice treated with iloprost had reduced lung tumors and increased Fzd 9 expression. These studies define a novel paradigm, linking the eicosanoid pathway and Wnt signaling. In addition, these data also suggest that prostacyclin analogs may represent a new class of therapeutic agents in the treatment of NSCLC where the restoration of noncanonical Wnt signaling maybe important for the inhibition of transformed cell growth.
Collapse
|
116
|
Iyer A, Fairlie DP, Prins JB, Hammock BD, Brown L. Inflammatory lipid mediators in adipocyte function and obesity. Nat Rev Endocrinol 2010; 6:71-82. [PMID: 20098448 DOI: 10.1038/nrendo.2009.264] [Citation(s) in RCA: 208] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Survival of multicellular organisms depends on their ability to fight infection, metabolize nutrients, and store energy for times of need. Unsurprisingly, therefore, immunoregulatory and metabolic mechanisms interact in human conditions such as obesity. Both infiltrating immunoinflammatory cells and adipocytes play critical roles in the modulation of metabolic homeostasis, so it is important to understand factors that regulate both adipocyte and immune cell function. A currently favored paradigm for obesity-associated metabolic dysfunction is that chronic macronutrient and/or lipid overload (associated with adiposity) induces cellular stress that initiates and perpetuates an inflammatory cycle and pathophysiological signaling of immunoinflammatory cells and adipocytes. Many lipid mediators exert their biological effects by binding to cognate receptors, such as G-protein-coupled receptors and Toll-like receptors. This process is tightly regulated under normal physiological conditions, and any disruption can initiate disease processes. Observations that cellular lipid loading (associated with adiposity) initiates inflammatory events has encouraged studies on the role of lipid mediators. In this review, we speculate that lipid mediators act on important immune receptors to induce low-grade tissue inflammation, which leads to adipocyte and metabolic dysfunction.
Collapse
Affiliation(s)
- Abishek Iyer
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | | | | | | | | |
Collapse
|
117
|
Naruhn S, Meissner W, Adhikary T, Kaddatz K, Klein T, Watzer B, Müller-Brüsselbach S, Müller R. 15-hydroxyeicosatetraenoic acid is a preferential peroxisome proliferator-activated receptor beta/delta agonist. Mol Pharmacol 2010; 77:171-84. [PMID: 19903832 DOI: 10.1124/mol.109.060541] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2025] Open
Abstract
Peroxisome proliferator-activated receptor (PPARs) modulate target gene expression in response to unsaturated fatty acid ligands, such as arachidonic acid (AA). Here, we report that the AA metabolite 15-hydroxyeicosatetraenoic acid (15-HETE) activates the ligand-dependent activation domain (AF2) of PPARbeta/delta in vivo, competes with synthetic agonists in a PPARbeta/delta ligand binding assay in vitro, and triggers the interaction of PPARbeta/delta with coactivator peptides. These agonistic effects were also seen with PPARalpha and PPARgamma, but to a significantly weaker extent. We further show that 15-HETE strongly induces the expression of the bona fide PPAR target gene Angptl4 in a PPARbeta/delta-dependent manner and, conversely, that inhibition of 15-HETE synthesis reduces PPARbeta/delta transcriptional activity. Consistent with its function as an agonistic ligand, 15-HETE triggers profound changes in chromatin-associated PPARbeta/delta complexes in vivo, including the recruitment of the coactivator cAMP response element-binding protein binding protein. Both 15R-HETE and 15S-HETE are similarly potent at inducing PPARbeta/delta coactivator binding and transcriptional activation, indicating that 15-HETE enantiomers generated by different pathways function as PPARbeta/delta agonists.
Collapse
Affiliation(s)
- Simone Naruhn
- Institut für Molekularbiologie und Tumorforschung, Philipps-Universität Marburg, 35032 Marburg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
118
|
Wagner KD, Wagner N. Peroxisome proliferator-activated receptor beta/delta (PPARbeta/delta) acts as regulator of metabolism linked to multiple cellular functions. Pharmacol Ther 2009; 125:423-35. [PMID: 20026355 DOI: 10.1016/j.pharmthera.2009.12.001] [Citation(s) in RCA: 143] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2009] [Accepted: 12/02/2009] [Indexed: 12/14/2022]
Abstract
Peroxisome proliferator-activated receptors (PPARs) are nuclear receptors. They function as ligand activated transcription factors. They exist in three isoforms, PPARalpha, PPARbeta (formerly PPARdelta), and PPARgamma. For all PPARs lipids are endogenous ligands, linking them directly to metabolism. PPARs form heterodimers with retinoic X receptors, and, upon ligand binding, modulate gene expression of downstream target genes dependent on the presence of co-repressors or co-activators. This results in cell-type specific complex regulations of proliferation, differentiation and cell survival. Specific synthetic agonists for all PPARs are available. PPARalpha and PPARgamma agonists are already in clinical use for the treatment of hyperlipidemia and type 2 diabetes, respectively. More recently, PPARbeta activation came into focus as an interesting novel approach for the treatment of metabolic syndrome and associated cardiovascular diseases. Although the initial notion was that PPARbeta is expressed ubiquitously, more recently extensive investigations have been performed demonstrating high PPARbeta expression in a variety of tissues, e.g. skin, skeletal muscle, adipose tissue, inflammatory cells, heart, and various types of cancer. In addition, in vitro and in vivo studies using specific PPARbeta agonists, tissue-specific over-expression or knockout mouse models have demonstrated a variety of functions of PPARbeta in adipose tissue, muscle, skin, inflammation, and cancer. We will focus here on functions of PPARbeta in adipose tissue, skeletal muscle, heart, angiogenesis and cancer related to modifications in metabolism and the identified underlying molecular mechanisms.
Collapse
|
119
|
Peters JM, Gonzalez FJ. Sorting out the functional role(s) of peroxisome proliferator-activated receptor-beta/delta (PPARbeta/delta) in cell proliferation and cancer. BIOCHIMICA ET BIOPHYSICA ACTA 2009; 1796:230-41. [PMID: 19505534 PMCID: PMC2762441 DOI: 10.1016/j.bbcan.2009.06.002] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/07/2009] [Revised: 06/01/2009] [Accepted: 06/02/2009] [Indexed: 12/19/2022]
Abstract
Peroxisome proliferator-activated receptor-beta/delta (PPARbeta/delta) has many beneficial physiological functions ranging from enhancing fatty acid catabolism, improving insulin sensitivity, inhibiting inflammation and increasing oxidative myofibers allowing for improved athletic performance. Thus, given the potential for targeting PPARbeta/delta for the prevention and/or treatment of diseases including diabetes, dyslipidemias, metabolic syndrome and cancer, it is critical to clarify the functional role of PPARbeta/delta in cell proliferation and associated disorders such as cancer. However, there is considerable controversy whether PPARbeta/delta stimulates or inhibits cell proliferation. This review summarizes the literature describing the influence of PPARbeta/delta on cell proliferation, with an emphasis toward dissecting the data that give rise to opposing hypotheses. Suggestions are offered to standardize measurements associated with these studies so that interlaboratory comparisons can be accurately assessed.
Collapse
Affiliation(s)
- Jeffrey M Peters
- Department of Veterinary and Biomedical Sciences and The Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA 16802, USA.
| | | |
Collapse
|
120
|
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.
Collapse
Affiliation(s)
- D Wang
- Department of Cancer Biology and GI Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030-4009, USA
| | | |
Collapse
|
121
|
Kashfi K. Anti-inflammatory agents as cancer therapeutics. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2009; 57:31-89. [PMID: 20230759 DOI: 10.1016/s1054-3589(08)57002-5] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cancer prevention sometimes referred to as tertiary prevention or chemoprevention makes use of specific xenobiotics or drugs to prevent, delay, or retard the development of cancer. Over the last two decades or so cancer prevention has made significant strides. For example, prevention of lung cancer through smoking cessation; cervical cancer prevention through regular Pap smear tests; colon cancer prevention through screening colonoscopy; and prostate cancer reductions by prostate-specific antigen measurements in conjunction with regular prostate examinations. The seminal epidemiological observation that nonsteroidal anti-inflammatory drugs (NSAIDs) prevent colon and other cancers has provided the impetus to develop novel chemoprevention approaches against cancer. To that end, a number of "designer drugs" have been synthesized that are in different stages of development, evaluation, and deployment. Some include the cyclooxygenase-2-specific inhibitors (coxibs), nitric oxide-releasing NSAIDs (NO-NSAIDs and NONO-NSAIDs), hydrogen sulfide-releasing NSAIDs, modulators of the lipoxygenase pathway, prostanoid receptor blockers, and chemokine receptor antagonists. In addition to these novel agents, there are also a host of naturally occurring compounds/micronutrients that have chemopreventive properties. This chapter reviews these classes of compounds, their utility and mechanism(s) of action against the background of mediators that link inflammation and cancer.
Collapse
Affiliation(s)
- Khosrow Kashfi
- Department of Physiology and Pharmacology, Sophie Davis School of Biomedical Education, The City College of The City University of New York, New York 10031, USA
| |
Collapse
|
122
|
Knockdown of peroxisome proliferator-activated receptor-beta induces less differentiation and enhances cell-fibronectin adhesion of colon cancer cells. Oncogene 2009; 29:516-26. [PMID: 19935699 DOI: 10.1038/onc.2009.370] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The role of peroxisome proliferator-activated receptor-beta/delta (PPAR-beta/delta) in the pathogenesis of colon cancer remains highly controversial. This study specifically silenced the PPAR-beta expression in three colon cancer cell lines with different metastatic potentials. Although PPAR-beta knockdown resulted in more malignant morphological changes, bigger colony sizes and lower carcinoembryonic antigen (CEA) secretion, and enhanced the cell-fibronectin adhesion, cell invasion and migration were unaffected. These effects were stronger in poorly metastatic cell lines compared with highly metastatic ones. Simultaneously, PPAR-beta knockdown decreased the mRNAs encoding adipocyte differentiation-related protein and liver fatty acid binding protein, and increased the mRNA of ILK, whereas the mRNAs encoding integrin-beta1 and angiopoietin-like 4 were unchanged. Using immunohistochemistry, we determined that the intensity of PPAR-beta expression was stronger in rectal cancers with better differentiation than in those with poor differentiation, and was stronger in early-stage tumors than in advanced ones. Together, these findings consistently indicate that PPAR-beta may facilitate differentiation and inhibit the cell-fibronectin adhesion of colon cancer, having a role as an inhibitor in the carcinogenesis and progression of colorectal cancer. Interestingly, PPAR-beta seems to have a more important role in poorly metastatic cells than in highly metastatic ones.
Collapse
|
123
|
Fang F, Kang Z, Wong C. Vitamin E tocotrienols improve insulin sensitivity through activating peroxisome proliferator-activated receptors. Mol Nutr Food Res 2009; 54:345-52. [DOI: 10.1002/mnfr.200900119] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
124
|
Kwak H, Hwang I, Kim JH, Kim MY, Yang JS, Jeong S. Modulation of transcription by the peroxisome proliferator-activated receptor delta--binding RNA aptamer in colon cancer cells. Mol Cancer Ther 2009; 8:2664-73. [PMID: 19723884 DOI: 10.1158/1535-7163.mct-09-0214] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Peroxisome proliferator-activated receptor delta (PPAR-delta), one of three PPAR subtypes, is a lipid-sensing nuclear receptor that has been implicated in multiple processes, including inflammation and cancer. To directly establish the role of PPAR-delta in colon cancer development and progression, we selected high-affinity RNA aptamers and expressed them in several colon cancer cell lines. Nuclear-expressed aptamers efficiently inhibited PPAR-delta-dependent transcription from a synthetic peroxisome proliferator response element-driven luciferase reporter. PPAR-delta-specific aptamers suppressed transcription from natural promoters of vascular endothelial cell growth factor-A and cyclooxygenase-2. Moreover, vascular endothelial cell growth factor-A and cyclooxygenase-2 mRNA levels were significantly reduced by the PPAR-delta-specific aptamers in colon cancer cells. Most significantly, HCT116 colon cancer cells with high-level expression of PPAR-delta-specific aptamers exhibited a striking loss of tumorigenic potential. Further study on these RNA aptamers could provide an opportunity to modulate PPAR-delta-mediated colon cancer development and progression. Taken together, our results establish an important role for PPAR-delta in transcription of tumor-promoting genes, which can be specifically modulated by high-affinity RNA intramers in colon cancer cells. The RNA intramers may be further developed as specific inhibitors for cancer therapeutic strategies.
Collapse
Affiliation(s)
- Hoyun Kwak
- Department of Molecular Biology, Institute of Nanosensor and Biotechnology, Dankook University, Yongin-si, Gyeonggi-do 448-701, Republic of Korea
| | | | | | | | | | | |
Collapse
|
125
|
Ishizuka M, Sawada T, Okada T, Nagata H, Takagi K, Horie T, Kubota K. Influence of Tumor Peroxisome Proliferator-Activated Receptor γ and δ Expression on Postoperative Mortality of Patients Undergoing Colorectal Cancer Surgery. J INVEST SURG 2009; 22:105-11. [DOI: 10.1080/08941930802713084] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
126
|
Robinson E, Grieve DJ. Significance of peroxisome proliferator-activated receptors in the cardiovascular system in health and disease. Pharmacol Ther 2009; 122:246-63. [PMID: 19318113 DOI: 10.1016/j.pharmthera.2009.03.003] [Citation(s) in RCA: 106] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2009] [Accepted: 03/03/2009] [Indexed: 01/12/2023]
Abstract
Peroxisome proliferator-activated receptors (PPARs) are ligand-activated nuclear transcription factors that belong to the nuclear receptor superfamily. Three isoforms of PPAR have been identified, alpha, delta and gamma, which play distinct roles in the regulation of key metabolic processes, such as glucose and lipid redistribution. PPARalpha is expressed predominantly in the liver, kidney and heart, and is primarily involved in fatty acid oxidation. PPARgamma is mainly associated with adipose tissue, where it controls adipocyte differentiation and insulin sensitivity. PPARdelta is abundantly and ubiquitously expressed, but as yet its function has not been clearly defined. Activators of PPARalpha (fibrates) and gamma (thiazolidinediones) have been used clinically for a number of years in the treatment of hyperlipidaemia and to improve insulin sensitivity in diabetes. More recently, PPAR activation has been found to confer additional benefits on endothelial function, inflammation and thrombosis, suggesting that PPAR agonists may be good candidates for the treatment of cardiovascular disease. In this regard, it has been demonstrated that PPAR activators are capable of reducing blood pressure and attenuating the development of atherosclerosis and cardiac hypertrophy. This review will provide a detailed discussion of the current understanding of basic PPAR physiology, with particular reference to the cardiovascular system. It will also examine the evidence supporting the involvement of the different PPAR isoforms in cardiovascular disease and discuss the current and potential future clinical applications of PPAR activators.
Collapse
Affiliation(s)
- Emma Robinson
- Centre for Vision and Vascular Science, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, 3rd Floor, Medical Biology Centre, 97 Lisburn Road, Belfast, BT9 7BL UK
| | | |
Collapse
|
127
|
Zuo X, Peng Z, Moussalli MJ, Morris JS, Broaddus RR, Fischer SM, Shureiqi I. Targeted genetic disruption of peroxisome proliferator-activated receptor-delta and colonic tumorigenesis. J Natl Cancer Inst 2009; 101:762-7. [PMID: 19436036 DOI: 10.1093/jnci/djp078] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Peroxisome proliferator-activated receptor-delta (PPAR-delta) is overexpressed in human colon cancer, but its contribution to colonic tumorigenesis is controversial. We generated a mouse model in which PPAR-delta was genetically disrupted in colonic epithelial cells by targeted deletion of exon 4. Elimination of colon-specific PPAR-delta expression was confirmed by real-time reverse transcription-polymerase chain reaction (real-time RT-PCR), immunoblotting, and activity assays. Mice with and without targeted PPAR-delta genetic disruption (10-11 mice per group) were tested for incidence of azoxymethane-induced colon tumors. The effects of targeted PPAR-delta deletion on vascular endothelial growth factor expression were determined by real-time RT-PCR. Targeted PPAR-delta genetic disruption inhibited colonic carcinogenesis: Mice with PPAR-delta((-/-)) colons developed 98.5% fewer tumors than wild-type mice (PPAR-delta((-/-)) vs wild-type, mean = 0.1 tumors per mouse vs 6.6 tumors per mouse, difference = 6.5 tumors per mouse, 95% confidence interval = 4.9 to 8.0 tumors per mouse, P < .001, two-sided test). Increased expression of vascular endothelial growth factor in colon tumors vs normal colon was suppressed by loss of PPAR-delta expression. These findings indicate that PPAR-delta has a crucial role in promoting colonic tumorigenesis.
Collapse
Affiliation(s)
- Xiangsheng Zuo
- Department of Clinical Cancer Prevention, Unit 1360, The University of Texas MD Anderson Cancer Center, Houston, TX 77030-4009, USA
| | | | | | | | | | | | | |
Collapse
|
128
|
Ondrey F. Peroxisome proliferator-activated receptor gamma pathway targeting in carcinogenesis: implications for chemoprevention. Clin Cancer Res 2009; 15:2-8. [PMID: 19118026 DOI: 10.1158/1078-0432.ccr-08-0326] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The peroxisome proliferator-activated receptor (PPAR) gamma is one member of the nuclear receptor superfamily that contains in excess of 80 described receptors. PPARgamma activators are a diverse group of agents that range from endogenous fatty acids or derivatives (linolenic, linoleic, and 15-deoxy-Delta(12,14)-prostaglandin J(2)) to Food and Drug Administration-approved thiazolidinedione drugs [pioglitazone (Actos) and rosiglitazone (Avandia)] for the treatment of diabetes. Once activated, PPARgamma will preferentially bind with retinoid X receptor alpha and signal antiproliferative, antiangiogenic, and prodifferentiation pathways in several tissue types, thus making it a highly useful target for down-regulation of carcinogenesis. Although PPAR-gamma activators show many anticancer effects on cell lines, their advancement into human advanced cancer clinical trials has met with limited success. This article will review translational findings in PPARgamma activation and targeting in carcinogenesis prevention as they relate to the potential use of PPARgamma activators clinically as cancer chemoprevention strategies.
Collapse
Affiliation(s)
- Frank Ondrey
- Department of Otolaryngology and University of Minnesota Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota 55455, USA.
| |
Collapse
|
129
|
Yoshinaga M, Kitamura Y, Chaen T, Yamashita S, Tsuruta S, Hisano T, Ikeda Y, Sakai H, Nakamura K, Takayanagi R, Muto Y. The simultaneous expression of peroxisome proliferator-activated receptor delta and cyclooxygenase-2 may enhance angiogenesis and tumor venous invasion in tissues of colorectal cancers. Dig Dis Sci 2009; 54:1108-14. [PMID: 18720000 DOI: 10.1007/s10620-008-0465-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2008] [Accepted: 07/16/2008] [Indexed: 12/09/2022]
Abstract
We conducted this study to evaluate the impact of the expression of peroxisome proliferator-activated receptor delta on angiogenesis in tissue samples of colorectal cancer. We examined 52 samples of primary human colorectal carcinomas and matched normal adjacent tissues to evaluate the expression of peroxisome proliferator-activated receptor delta, cyclooxygenase-2, vascular endothelial growth factor-A, and CD34 through immunohistochemical analysis. Peroxisome proliferator-activated receptor delta was expressed in 25 (48.1%), and cyclooxygenase-2 was expressed in 26 (50.0%) of total colorectal cancer tissues. Tissue samples were divided into four groups, according to the expression of peroxisome proliferator-activated receptor delta and cyclooxygenase-2. The positive rate of vascular endothelial growth factor-A, the levels of microvascular density, and the incidence of venous vessel invasion in peroxisome proliferator-activated receptor delta (+)/cyclooxygenase-2 (+) samples exceeded significantly those in the other three groups of tissue samples (P<0.05). The results suggest that the axis of the cyclooxygenase-2/peroxisome proliferator-activated receptor delta signal pathway might play a crucial role in the development of colorectal cancers by enhancing angiogenesis.
Collapse
Affiliation(s)
- Masahiro Yoshinaga
- Department of Gastroenterology, Hepathology, and Nutrition, National Hospital Organization Beppu Medical Center, 1473 Uchikamado, Beppu, Oita 874-0011, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
130
|
Chong HC, Tan MJ, Philippe V, Tan SH, Tan CK, Ku CW, Goh YY, Wahli W, Michalik L, Tan NS. Regulation of epithelial-mesenchymal IL-1 signaling by PPARbeta/delta is essential for skin homeostasis and wound healing. ACTA ACUST UNITED AC 2009; 184:817-31. [PMID: 19307598 PMCID: PMC2699156 DOI: 10.1083/jcb.200809028] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Skin morphogenesis, maintenance, and healing after wounding require complex epithelial–mesenchymal interactions. In this study, we show that for skin homeostasis, interleukin-1 (IL-1) produced by keratinocytes activates peroxisome proliferator–activated receptor β/δ (PPARβ/δ) expression in underlying fibroblasts, which in turn inhibits the mitotic activity of keratinocytes via inhibition of the IL-1 signaling pathway. In fact, PPARβ/δ stimulates production of the secreted IL-1 receptor antagonist, which leads to an autocrine decrease in IL-1 signaling pathways and consequently decreases production of secreted mitogenic factors by the fibroblasts. This fibroblast PPARβ/δ regulation of the IL-1 signaling is required for proper wound healing and can regulate tumor as well as normal human keratinocyte cell proliferation. Together, these findings provide evidence for a novel homeostatic control of keratinocyte proliferation and differentiation mediated via PPARβ/δ regulation in dermal fibroblasts of IL-1 signaling. Given the ubiquitous expression of PPARβ/δ, other epithelial–mesenchymal interactions may also be regulated in a similar manner.
Collapse
Affiliation(s)
- Han Chung Chong
- School of Biological Sciences, Nanyang Technological University, Singapore 637551
| | | | | | | | | | | | | | | | | | | |
Collapse
|
131
|
Abstract
PURPOSE OF REVIEW Eicosanoids are products of arachidonic acid metabolism which have important roles in renal homeostasis and disease. In recent years the development of genetically modified animals and new drugs targeting eicosanoids producing enzymes and receptors has unveiled new roles for eicosanoids in kidney function. This review provides an overview of eicosanoid biosynthesis and receptors and discusses recent findings on their role in acute and chronic renal diseases and in renal transplantation. RECENT FINDINGS Products of the cyclooxygenases, 5-lipoxygenase, and cytochrome P450 pathways of arachidonic acid metabolism act through distinct receptors presented at different segment of the nephron. Apart from its role in renal physiology and hemodynamic, eicosanoids actively participate in the pathogenesis of acute and chronic renal diseases and have immunoregulatory role in kidney transplantation. SUMMARY The new discoveries on the role of eicosanoids in kidney functions and the development of drugs targeting eicosanoids synthesis or action should help to envisage novel therapeutic approaches for patients suffering from renal diseases.
Collapse
|
132
|
Abstract
Organotypic cultures help to unravel how a transcription factor modulates crosstalk between different layers of the skin.
Collapse
|
133
|
Ghosh M, Ai Y, Narko K, Wang Z, Peters JM, Hla T. PPARdelta is pro-tumorigenic in a mouse model of COX-2-induced mammary cancer. Prostaglandins Other Lipid Mediat 2009; 88:97-100. [PMID: 19101649 PMCID: PMC2662999 DOI: 10.1016/j.prostaglandins.2008.11.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2008] [Revised: 11/08/2008] [Accepted: 11/08/2008] [Indexed: 02/07/2023]
Abstract
Cyclooxygenase-2 (COX-2), overexpressed in inflammatory conditions and cancer, regulates angiogenesis and tumorigenesis via the production of biologically active prostanoids. Previously, we showed that COX-2 over-expression in the mammary gland of transgenic mice induces an angiogenic switch and transforms the mammary epithelium into invasive mammary carcinoma. Since COX-2-derived prostanoids can activate the nuclear receptor PPARdelta, we crossed Ppardelta(-/-) mice with COX-2 transgenic mice in the FVB/N background. PPARdelta was expressed constitutively in the mammary gland of virgin, pregnant and lactating mice. Mammary hyperplasia and tumorigenesis in the COX-2 transgenic mice was markedly reduced in the Ppardelta(-/-) mice compared to their wild type counterparts. Analysis of the mammary tissues indicated that immunoreactive Ki-67, cyclin D1 and phosphorylated histone 3 (Phospho H3) were reduced in Ppardelta(-/-) mice, suggesting that PPARdelta activation regulates cell proliferation in the mammary gland. We postulate that activation of the nuclear receptor PPARdelta by COX-2-derived prostanoids may be involved in the proliferation of mammary epithelial cells and therefore contribute to mammary cancer development.
Collapse
Affiliation(s)
- Mallika Ghosh
- Center for Vascular Biology, University of Connecticut Health Center, Farmington, CT 06030-3501, United States
| | - Youxi Ai
- Center for Vascular Biology, University of Connecticut Health Center, Farmington, CT 06030-3501, United States
| | - Kirsi Narko
- Center for Vascular Biology, University of Connecticut Health Center, Farmington, CT 06030-3501, United States
| | - Zhenglong Wang
- Vascular Biology Center, Yale University School of Medicine, New Haven, CT 06510, United States
| | - Jeffrey M. Peters
- Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, PA 16802, United States
| | - Timothy Hla
- Center for Vascular Biology, University of Connecticut Health Center, Farmington, CT 06030-3501, United States
| |
Collapse
|
134
|
PPARδ activity in cardiovascular diseases: A potential pharmacological target. PPAR Res 2009; 2009:745821. [PMID: 19325917 PMCID: PMC2659552 DOI: 10.1155/2009/745821] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2008] [Revised: 12/21/2008] [Accepted: 02/12/2009] [Indexed: 11/17/2022] Open
Abstract
Activation of peroxisome proliferator-activated receptors (PPARs), and particularly of
PPARα and PPARγ, using selective agonists, is currently used in the treatment of metabolic diseases such as hypertriglyceridemia and type 2 diabetes mellitus. PPARα and PPARγ anti-inflammatory, antiproliferative and antiangiogenic properties in cardiovascular cells were
extensively clarified in a variety of in vitro and in vivo models. In contrast, the role of PPARδ in cardiovascular system is poorly understood. Prostacyclin, the predominant prostanoid released by
vascular cells, is a putative endogenous agonist for PPARδ, but only recently PPARδ selective synthetic agonists were found, improving studies about the physiological and pathophysiological roles of PPARδ activation. Recent reports suggest that the PPARδ activation may play a pivotal role to
regulate inflammation, apoptosis, and cell proliferation, suggesting that this transcriptional factor could become an interesting pharmacological target to regulate cardiovascular cell apoptosis, proliferation, inflammation, and metabolism.
Collapse
|
135
|
Wang JB, Qi LL, Zheng SD, Wang HZ, Wu TX. Curcumin suppresses PPARδ expression and related genes in HT-29 cells. World J Gastroenterol 2009; 15:1346-52. [PMID: 19294764 PMCID: PMC2658832 DOI: 10.3748/wjg.15.1346] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effects of curcumin on the expression of peroxisome proliferator-activated receptorδ (PPARδ) and related genes in HT-29 cells.
METHODS: HT-29 cells were treated with curcumin (0-80 &mgr;mol/L) for 24 h. The effects of curcumin on the morphology of HT-29 cells were studied by Hoechst 33342 staining. The activity of caspase-3 was determined using DEVD-pNA as substrate. The levels of peroxisome PPARδ, 14-3-3epsilon and vascular endothelial growth factor (VEGF) in HT-29 cells were determined by Western blotting analysis and their mRNA expression was determined by real-time quantitative RT-PCR.
RESULTS: Treatment with 10-80 &mgr;mol/L curcumin induced typical features of apoptosis and activated the caspase-3 in HT-29 cells. The expression of PPARδ, 14-3-3epsilon and VEGF was reduced and the activity of β-catenin/Tcf-4 signaling was inhibited by curcumin treatment.
CONCLUSION: Curcumin can induce apoptosis of HT-29 cells and down-regulate the expression of PPARδ, 14-3-3epsilon and VEGF in HT-29.
Collapse
|
136
|
The PPARδ ligand GW501516 reduces growth but not apoptosis in mouse inner medullary collecting duct cells. PPAR Res 2009; 2009:706283. [PMID: 19266055 PMCID: PMC2651001 DOI: 10.1155/2009/706283] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2008] [Revised: 12/12/2008] [Accepted: 01/05/2009] [Indexed: 11/18/2022] Open
Abstract
The collecting duct (CD) expresses considerable amounts of PPARδ. While its role is unknown in the CD, in other renal cells it has been shown to regulate both growth and apoptosis. We thus hypothesized that PPARδ reduces apoptotic responses and stimulates cell
growth in the mouse CD, and examined the effect of GW501516, a synthetic PPARδ ligand, on these responses in mouse IMCD-K2 cells. High doses of GW501516 decreased both DNA and protein synthesis in these cells by 80%, but had no overall effect on cell viability. Although anisomycin treatment resulted in an increase of caspase-3 levels of about 2.59-fold of control, GW501516 did not affect anisomycin-induced changes in active caspase-3 levels. These results show that a
PPARδ ligand inhibits growth but does not affect anisomycin-apoptosis in a mouse IMCD cell line. This could have therapeutic implications for renal diseases associated with increased CD growth responses.
Collapse
|
137
|
Hassan MR, Hossain MM, Bailey J, Macintyre G, Ho JWK, Ramamohanarao K. A voting approach to identify a small number of highly predictive genes using multiple classifiers. BMC Bioinformatics 2009; 10 Suppl 1:S19. [PMID: 19208118 PMCID: PMC2648737 DOI: 10.1186/1471-2105-10-s1-s19] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Background Microarray gene expression profiling has provided extensive datasets that can describe characteristics of cancer patients. An important challenge for this type of data is the discovery of gene sets which can be used as the basis of developing a clinical predictor for cancer. It is desirable that such gene sets be compact, give accurate predictions across many classifiers, be biologically relevant and have good biological process coverage. Results By using a new type of multiple classifier voting approach, we have identified gene sets that can predict breast cancer prognosis accurately, for a range of classification algorithms. Unlike a wrapper approach, our method is not specialised towards a single classification technique. Experimental analysis demonstrates higher prediction accuracies for our sets of genes compared to previous work in the area. Moreover, our sets of genes are generally more compact than those previously proposed. Taking a biological viewpoint, from the literature, most of the genes in our sets are known to be strongly related to cancer. Conclusion We show that it is possible to obtain superior classification accuracy with our approach and obtain a compact gene set that is also biologically relevant and has good coverage of different biological processes.
Collapse
Affiliation(s)
- Md Rafiul Hassan
- Department of Computer Science and Software Engineering, The University of Melbourne, Victoria 3010, Australia.
| | | | | | | | | | | |
Collapse
|
138
|
Abstract
Peroxisome proliferator-activated receptors belong to the superfamily of ligand-dependent nuclear receptor transcription factors, which include three subtypes: PPAR-α, β/δ, and γ. PPAR-δ, play important roles in the regulation of cell growth and differentiation as well as tissue wound and repair. Emerging evidence has also demonstrated that PPAR-δ is implicated in lipids and glucose metabolism. Most recently, the direct effects of PPAR-δ on cardiovascular processes such as endothelial function and angiogenesis have also been investigated. Therefore, it is suggested that PPAR-δ may have critical roles in cardiovascular pathophysiology and is a potential target for therapeutic intervention of cardiovascular disorders such as atherosclerosis.
Collapse
|
139
|
Cyclooxygenase inhibitors induce colon cancer cell apoptosis Via PPARdelta --> 14-3-3epsilon pathway. Methods Mol Biol 2009; 512:295-307. [PMID: 19347284 DOI: 10.1007/978-1-60327-530-9_16] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Non-steroidal anti-inflammatory drugs (NSAIDs) and selective cyclooxygenase-2 (COX-2) inhibitors (COXIBs) induce cancer cell apoptosis via several signaling pathways. There is evidence that they induce colon cancer cell apoptosis by suppressing peroxisome proliferator-activated receptor delta (PPARdelta) through inhibition of COX-2-derived prostacyclin (PGI2). PGI2 activates PPARdelta resulting in binding of PPARdelta to specific PPAR response elements (PPRE) of target genes. We have identified 14-3-3epsilon as one of the genes that are upregulated by PPARdelta. Elevated 14-3-3epsilon proteins in cytosol enhance sequestration of Bad and reduce mitochondrial damage by Bad and thereby control apoptosis. NSAIDs and COXIBs block PGI(2) production, thereby reducing PPARdelta DNA binding activity and abrogating 14-3-3e upregulation. Furthermore, the COX-2 inhibitors suppress PPARdelta expression. Suppression of PPARdelta leads to reduced 14-3-3e and hence a decline in Bad sequestration, resulting in an increased Bad-induced apoptosis via the mitochondrial death pathway.
Collapse
|
140
|
Michalik L, Wahli W. PPARs Mediate Lipid Signaling in Inflammation and Cancer. PPAR Res 2008; 2008:134059. [PMID: 19125181 PMCID: PMC2606065 DOI: 10.1155/2008/134059] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2008] [Accepted: 09/17/2008] [Indexed: 02/06/2023] Open
Abstract
Lipid mediators can trigger physiological responses by activating nuclear hormone receptors, such as the peroxisome proliferator-activated receptors (PPARs). PPARs, in turn, control the expression of networks of genes encoding proteins involved in all aspects of lipid metabolism. In addition, PPARs are tumor growth modifiers, via the regulation of cancer cell apoptosis, proliferation, and differentiation, and through their action on the tumor cell environment, namely, angiogenesis, inflammation, and immune cell functions. Epidemiological studies have established that tumor progression may be exacerbated by chronic inflammation. Here, we describe the production of the lipids that act as activators of PPARs, and we review the roles of these receptors in inflammation and cancer. Finally, we consider emerging strategies for therapeutic intervention.
Collapse
Affiliation(s)
- Liliane Michalik
- Center for Integrative Genomics, National Research Center Frontiers in Genetics, University of Lausanne, 1015 Lausanne, Switzerland
| | - Walter Wahli
- Center for Integrative Genomics, National Research Center Frontiers in Genetics, University of Lausanne, 1015 Lausanne, Switzerland
| |
Collapse
|
141
|
Jouzeau JY, Moulin D, Koufany M, Sebillaud S, Bianchi A, Netter P. [Pathophysiological relevance of peroxisome proliferators activated receptors (PPAR) to joint diseases - the pro and con of agonists]. ACTA ACUST UNITED AC 2008; 202:289-312. [PMID: 19094928 DOI: 10.1051/jbio:2008034] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Peroxisome proliferators activated receptors (PPAR) are ligand-inducible nuclear transacting factors comprising three subtypes, PPARalpha, PPARbeta/delta and PPARgamma, which play a key role in lipids and glucose homeostasis. All PPAR subtypes have been identified in joint or inflammatory cells and their activation resulted in a transcriptional repression of pro-inflammatory cytokines (IL-1, TNFalpha), early inflammatory genes (NOS(2), COX-2, mPGES-1) or matrix metalloproteases (MMP-1, MMP-13), at least for the gamma subtype. PPAR full agonists were also shown to stimulate IL-1 receptor antagonist (IL-1Ra) production by cytokine-stimulated articular cells in a subtype-dependent manner. These anti-inflammatory and anti-catabolic properties were confirmed in animal models of joint diseases where PPAR agonists reduced synovial inflammation while preventing cartilage destruction or inflammatory bone loss, although many effects required much higher doses than needed to restore insulin sensitivity or to lower circulating lipid levels. However, these promising effects of PPAR full agonists were hampered by their ability to reduce the growth factor-dependent synthesis of extracellular matrix components or to induce chondrocyte apoptosis, by the possible contribution of immunosuppressive properties to their anti-arthritic effects, by the increased adipocyte differentiation secondary to prolonged stimulation of PPARgamma, and by a variable contribution of PPAR subtypes depending on the system. Clinical data are scarce in rheumatoid arthritis (RA) patients whereas thousands of patients worldwilde, treated with PPAR agonists for type 2 diabetes or dyslipidemia, are paradoxically prone to suffer from osteoarthritis (OA). Whereas high dosage of full agonists may expose RA patients to cardiovascular adverse effects, the proof of concept that PPAR agonists have therapeutical relevance to OA may benefit from an epidemiological follow-up of joint lesions in diabetic or hyperlipidemic patients treated for long periods of time with glitazones or fibrates. Additionally, cellular and animal studies are required to assess whether partial agonists of PPAR (SPPARMs) may preserve therapeutical properties with potentially less safety concern.
Collapse
|
142
|
The Potential Applications of Peroxisome Proliferator-Activated Receptor delta Ligands in Assisted Reproductive Technology. PPAR Res 2008; 2008:794814. [PMID: 19096716 PMCID: PMC2603272 DOI: 10.1155/2008/794814] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2008] [Revised: 06/27/2008] [Accepted: 09/12/2008] [Indexed: 12/23/2022] Open
Abstract
Peroxisome proliferator-activated receptor δ (PPARδ, also known as PPARβ) has ubiquitous distribution and extensive biological functions. The reproductive function of PPARδ was first revealed in the uterus at the implantation site. Since then, PPARδ and its ligand have been discovered in all reproductive tissues, including the gametes and the preimplantation embryos. PPARδ in preimplantation embryos is normally activated by oviduct-derived PPARδ ligand. PPARδ activation is associated with an increase in embryonic cell proliferation and a decrease in programmed cell death (apoptosis). On the other hand, the role of PPARδ and its ligand in gamete formation and function is less well understood. This review will summarize the reproductive functions of PPARδ and project its potential applications in assisted reproductive technology.
Collapse
|
143
|
Jacobs W, Vonk-Noordegraaf A. Epoprostenol in pulmonary arterial hypertension. Expert Opin Drug Metab Toxicol 2008; 5:83-90. [DOI: 10.1517/17425250802622962] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
144
|
He P, Borland MG, Zhu B, Sharma AK, Amin S, El-Bayoumy K, Gonzalez FJ, Peters JM. Effect of ligand activation of peroxisome proliferator-activated receptor-beta/delta (PPARbeta/delta) in human lung cancer cell lines. Toxicology 2008; 254:112-7. [PMID: 18950674 PMCID: PMC2653217 DOI: 10.1016/j.tox.2008.09.023] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2008] [Revised: 09/22/2008] [Accepted: 09/23/2008] [Indexed: 11/21/2022]
Abstract
There is compelling evidence that peroxisome proliferator-activated receptor-beta/delta (PPARbeta/delta) mediates terminal differentiation and is associated with inhibition of cell growth. However, it was recently suggested that growth of two human lung cancer cell lines is enhanced by PPARbeta/delta. The goal of the present study was to provide insight in resolving this controversy. Therefore, the effect of ligand activation of PPARbeta/delta in A549 and H1838 human lung cancer cell lines was examined using two high affinity PPARbeta/delta ligands. Ligand activation of PPARbeta/delta caused up-regulation of a known PPARbeta/delta target gene, angiopoietin-like 4 (Angptl4) but did not influence expression of phosphatase and tensin homolog (PTEN) or phosphorylation of protein kinase B (Akt), and did not affect cell growth. Results from this study demonstrate that two human lung cancer cell lines respond to ligand activation of PPARbeta/delta by modulation of target gene expression (Angptl4), but fail to exhibit significant modulation of cell proliferation.
Collapse
Affiliation(s)
- Pengfei He
- Department of Veterinary and Biomedical Sciences and The Center of Molecular Toxicology and Carcinogenesis
| | - Michael G. Borland
- Department of Veterinary and Biomedical Sciences and The Center of Molecular Toxicology and Carcinogenesis
- Graduate Program in Biochemistry, Microbiology, and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania 16802
| | - Bokai Zhu
- Department of Veterinary and Biomedical Sciences and The Center of Molecular Toxicology and Carcinogenesis
| | - Arun K. Sharma
- Department of Pharmacology, The Pennsylvania State University, Milton S. Hershey Medical Center, Hershey, Pennsylvania 17033
| | - Shantu Amin
- Department of Pharmacology, The Pennsylvania State University, Milton S. Hershey Medical Center, Hershey, Pennsylvania 17033
| | - Karam El-Bayoumy
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, Milton S. Hershey Medical Center, Hershey, Pennsylvania 17033
| | - Frank J. Gonzalez
- Laboratory of Metabolism, National Cancer Institute, Bethesda, Maryland 20892 USA
| | - Jeffrey M. Peters
- Department of Veterinary and Biomedical Sciences and The Center of Molecular Toxicology and Carcinogenesis
- Graduate Program in Biochemistry, Microbiology, and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania 16802
| |
Collapse
|
145
|
Pedchenko TV, Gonzalez AL, Wang D, DuBois RN, Massion PP. Peroxisome proliferator-activated receptor beta/delta expression and activation in lung cancer. Am J Respir Cell Mol Biol 2008; 39:689-96. [PMID: 18566335 PMCID: PMC3159083 DOI: 10.1165/rcmb.2007-0426oc] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2007] [Accepted: 06/12/2008] [Indexed: 12/13/2022] Open
Abstract
Peroxisome proliferator-activated receptor beta/delta (PPARbeta/delta) is a ligand-binding inducible transcriptional factor linked to carcinogenesis. Important functions of PPARbeta/delta were demonstrated in series of human epithelial cancers; however, its role in lung cancer remains controversial. We investigated the differential expression level and localization of PPARbeta/delta in tumors and adjacent normal lung tissue, and the effect of PPARbeta/delta activation on lung cancer cell proliferation and apoptosis. PPARbeta/delta was expressed in all studied human non-small cell lung cancers, and strong PPARbeta/delta immunoreactivity was observed in epithelial cells of more than 75% of studied lung tumors. PPARbeta/delta expression was consistently limited to the cancer cells in tumor tissue, while in adjacent normal lung tissue it was limited predominantly to the mononuclear cells. We found that ligand-binding activation of PPARbeta/delta stimulates cell proliferation (an effect that was blocked by a dominant-negative construct of PPARbeta/delta), stimulates anchorage-independent cell growth, and inhibits apoptosis in lung cancer cell lines. Importantly, the activation of PPARbeta/delta induces Akt phosphorylation correlated with up-regulation of PDK1, down-regulation of PTEN, and increased expression of Bcl-xL and COX-2. These findings indicate that PPARbeta/delta exerts proliferative and anti-apoptotic effects via PI3K/Akt1 and COX-2 pathways. In conclusion, PPARbeta/delta is strongly expressed in the majority of lung cancers, and its activation induces proliferative and survival response in non-small cell lung cancer.
Collapse
Affiliation(s)
- Tetyana V. Pedchenko
- Division of Allergy, Pulmonary, and Critical Care Medicine, Division of Pathology, Department of Medicine, and Vanderbilt-Ingram Comprehensive Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Adriana L. Gonzalez
- Division of Allergy, Pulmonary, and Critical Care Medicine, Division of Pathology, Department of Medicine, and Vanderbilt-Ingram Comprehensive Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - DingZhi Wang
- Division of Allergy, Pulmonary, and Critical Care Medicine, Division of Pathology, Department of Medicine, and Vanderbilt-Ingram Comprehensive Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Raymond N. DuBois
- Division of Allergy, Pulmonary, and Critical Care Medicine, Division of Pathology, Department of Medicine, and Vanderbilt-Ingram Comprehensive Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Pierre P. Massion
- Division of Allergy, Pulmonary, and Critical Care Medicine, Division of Pathology, Department of Medicine, and Vanderbilt-Ingram Comprehensive Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee
| |
Collapse
|
146
|
The Role of PPARs in the Endothelium: Implications for Cancer Therapy. PPAR Res 2008; 2008:904251. [PMID: 19043612 PMCID: PMC2586817 DOI: 10.1155/2008/904251] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2008] [Revised: 07/28/2008] [Accepted: 09/09/2008] [Indexed: 12/18/2022] Open
Abstract
The growth and metastasis of cancers intimately involve the vasculature and in particular the endothelial cell layer. Tumours require new blood vessel formation via angiogenesis to support growth. In addition, inflammation, coagulation, and platelet activation are common signals in the growth and metastasis of tumour cells. The endothelium plays a central role in the homeostatic control of inflammatory cell recruitment, regulating platelet activation and coagulation pathways. PPARalpha, -beta/delta, and -gamma are all expressed in endothelial cells. This review will discuss the roles of PPARs in endothelial cells in relation to angiogenesis, inflammation, coagulation, and platelet control pathways. In particular, we will discuss the recent evidence that supports the hypothesis that PPARalpha and PPARgamma are antiangiogenic receptors, while PPARbeta/delta is proangiogenic.
Collapse
|
147
|
Valcárcel M, Arteta B, Jaureguibeitia A, Lopategi A, Martínez I, Mendoza L, Muruzabal FJ, Salado C, Vidal-Vanaclocha F. Three-dimensional growth as multicellular spheroid activates the proangiogenic phenotype of colorectal carcinoma cells via LFA-1-dependent VEGF: implications on hepatic micrometastasis. J Transl Med 2008; 6:57. [PMID: 18844982 PMCID: PMC2579286 DOI: 10.1186/1479-5876-6-57] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2008] [Accepted: 10/09/2008] [Indexed: 11/10/2022] Open
Abstract
Background The recruitment of vascular stromal and endothelial cells is an early event occurring during cancer cell growth at premetastatic niches, but how the microenvironment created by the initial three-dimensional (3D) growth of cancer cells affects their angiogenesis-stimulating potential is unclear. Methods The proangiogenic profile of CT26 murine colorectal carcinoma cells was studied in seven-day cultured 3D-spheroids of <300 μm in diameter, produced by the hanging-drop method to mimic the microenvironment of avascular micrometastases prior to hypoxia occurrence. Results Spheroid-derived CT26 cells increased vascular endothelial growth factor (VEGF) secretion by 70%, which in turn increased the in vitro migration of primary cultured hepatic sinusoidal endothelium (HSE) cells by 2-fold. More importantly, spheroid-derived CT26 cells increased lymphocyte function associated antigen (LFA)-1-expressing cell fraction by 3-fold; and soluble intercellular adhesion molecule (ICAM)-1, given to spheroid-cultured CT26 cells, further increased VEGF secretion by 90%, via cyclooxygenase (COX)-2-dependent mechanism. Consistent with these findings, CT26 cancer cells significantly increased LFA-1 expression in non-hypoxic avascular micrometastases at their earliest inception within hepatic lobules in vivo; and angiogenesis also markedly increased in both subcutaneous tumors and hepatic metastases produced by spheroid-derived CT26 cells. Conclusion 3D-growth per se enriched the proangiogenic phenotype of cancer cells growing as multicellular spheroids or as subclinical hepatic micrometastases. The contribution of integrin LFA-1 to VEGF secretion via COX-2 was a micro environmental-related mechanism leading to the pro-angiogenic activation of soluble ICAM-1-activated colorectal carcinoma cells. This mechanism may represent a new target for specific therapeutic strategies designed to block colorectal cancer cell growth at a subclinical micrometastatic stage within the liver.
Collapse
Affiliation(s)
- María Valcárcel
- Dept, Cell Biology and Histology, Basque Country University School of Medicine & Dentistry, Bizkaia-48940, Spain.
| | | | | | | | | | | | | | | | | |
Collapse
|
148
|
Nemenoff R, Meyer AM, Hudish TM, Mozer AB, Snee A, Narumiya S, Stearman RS, Winn RA, Weiser-Evans M, Geraci MW, Keith RL. Prostacyclin prevents murine lung cancer independent of the membrane receptor by activation of peroxisomal proliferator--activated receptor gamma. Cancer Prev Res (Phila) 2008; 1:349-56. [PMID: 19138979 PMCID: PMC2680197 DOI: 10.1158/1940-6207.capr-08-0145] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Overexpression of prostacyclin synthase (PGIS) decreases lung tumor multiplicity in chemical- and cigarette-smoke-induced murine lung cancer models. Prostacyclin signals through a single G-protein-coupled receptor (IP), which signals through cyclic AMP. To determine the role of this receptor in lung cancer chemoprevention by prostacyclin, PGIS-overexpressing mice were crossed to mice that lack the IP receptor [IP(-/-)]. Carcinogen-induced lung tumor incidence was similar in IP(+/+), IP(+/-), and IP(-/-) mice, and overexpression of PGIS gave equal protection in all three groups, indicating that the protective effects of prostacyclin are not mediated through activation of IP. Because prostacyclin can activate members of the peroxisomal proliferator-activated receptor (PPAR) family of nuclear receptors, we examined the role of PPARgamma in the protection of prostacyclin against lung tumorigenesis. Iloprost, a stable prostacyclin analogue, activated PPARgamma in nontransformed bronchial epithelial cells and in a subset of human non-small-cell lung cancer cell lines. Iloprost-impregnated chow fed to wild-type mice resulted in elevated lung macrophages and decreased lung tumor formation. Transgenic animals with lung-specific PPARgamma overexpression also developed fewer lung tumors. This reduction was not enhanced by administration of supplemental iloprost. These studies indicate that PPARgamma is a critical target for prostacyclin-mediated lung cancer chemoprevention and may also have therapeutic activity.
Collapse
Affiliation(s)
- Raphael Nemenoff
- Division of Renal Diseases and Hypertension, University of Colorado Health Sciences Center, 4200 E. Ninth Ave, Box C272, Denver, CO 80262
| | - Amy M. Meyer
- Division of Renal Diseases and Hypertension, University of Colorado Health Sciences Center, 4200 E. Ninth Ave, Box C272, Denver, CO 80262
| | - Tyler M. Hudish
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Denver VA Medical Center, 1055 Clermont St. Box 111A, Denver, CO 80220
| | - Anthony B. Mozer
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Denver VA Medical Center, 1055 Clermont St. Box 111A, Denver, CO 80220
| | - Amy Snee
- Division of Renal Diseases and Hypertension, University of Colorado Health Sciences Center, 4200 E. Ninth Ave, Box C272, Denver, CO 80262
| | - Shuh Narumiya
- Department of Pharmacology, Kyoto University Faculty of Medicine, and Department of Physiological Chemistry, Kyoto University Faculty of Pharmaceutical Sciences, Kyoto, Japan
| | - Robert S. Stearman
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Health Sciences Center, 4200 E. Ninth Ave, Box C272, Denver, CO 80262
| | - Robert A. Winn
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Health Sciences Center, 4200 E. Ninth Ave, Box C272, Denver, CO 80262
| | - Mary Weiser-Evans
- Division of Renal Diseases and Hypertension, University of Colorado Health Sciences Center, 4200 E. Ninth Ave, Box C272, Denver, CO 80262
| | - Mark W. Geraci
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Health Sciences Center, 4200 E. Ninth Ave, Box C272, Denver, CO 80262
| | - Robert L. Keith
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Health Sciences Center, 4200 E. Ninth Ave, Box C272, Denver, CO 80262
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Denver VA Medical Center, 1055 Clermont St. Box 111A, Denver, CO 80220
| |
Collapse
|
149
|
Wang D, DuBois RN. Pro-inflammatory prostaglandins and progression of colorectal cancer. Cancer Lett 2008; 267:197-203. [PMID: 18406516 PMCID: PMC2553688 DOI: 10.1016/j.canlet.2008.03.004] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2008] [Revised: 03/18/2008] [Accepted: 03/03/2008] [Indexed: 12/25/2022]
Abstract
Chronic inflammation is a risk factor for several gastrointestinal malignancies, including esophageal, gastric, hepatic, pancreatic and colorectal cancer. It has long been known that long-term use of nonsteroidal anti-inflammatory drugs (NSAIDs) reduces the relative risk of developing colorectal cancer. NSAIDs exert their anti-inflammatory and anti-tumor effects primarily by inhibiting activity of cyclooxygenase (COX) enzymes. Cyclooxygenase enzymes catalyze the conversion of arachidonic acid into prostanoids, including prostaglandins (PGs) and thromboxanes (TXs). Emerging evidence demonstrates that prostaglandins play an important role in inflammation and cancer. In this review, we highlight recent breakthroughs in our understanding of the roles of the different prostaglandins in colorectal cancer (CRC) and inflammatory bowel disease (IBD). These findings may provide a rationale for the development of new anti-inflammatory therapeutic approaches to cancer prevention and/or treatment.
Collapse
Affiliation(s)
- Dingzhi Wang
- Departments of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Raymond N. DuBois
- Cancer Biology, MD Anderson Cancer Center, The University of Texas, Houston, TX 77030-4009
| |
Collapse
|
150
|
Peters JM, Hollingshead HE, Gonzalez FJ. Role of peroxisome-proliferator-activated receptor beta/delta (PPARbeta/delta) in gastrointestinal tract function and disease. Clin Sci (Lond) 2008; 115:107-27. [PMID: 18616431 PMCID: PMC2705117 DOI: 10.1042/cs20080022] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PPARbeta/delta (peroxisome-proliferator-activated receptor beta/delta) is one of three PPARs in the nuclear hormone receptor superfamily that are collectively involved in the control of lipid homoeostasis among other functions. PPARbeta/delta not only acts as a ligand-activated transcription factor, but also affects signal transduction by interacting with other transcription factors such as NF-kappaB (nuclear factor kappaB). Constitutive expression of PPARbeta/delta in the gastrointestinal tract is very high compared with other tissues and its potential physiological roles in this tissue include homoeostatic regulation of intestinal cell proliferation/differentiation and modulation of inflammation associated with inflammatory bowel disease and colon cancer. Analysis of mouse epithelial cells in the intestine and colon has clearly demonstrated that ligand activation of PPARbeta/delta induces terminal differentiation. The PPARbeta/delta target genes mediating this effect are currently unknown. Emerging evidence suggests that PPARbeta/delta can suppress inflammatory bowel disease through PPARbeta/delta-dependent and ligand-independent down-regulation of inflammatory signalling. However, the role of PPARbeta/delta in colon carcinogenesis remains controversial, as conflicting evidence suggests that ligand activation of PPARbeta/delta can either potentiate or attenuate this disease. In the present review, we summarize the role of PPARbeta/delta in gastrointestinal physiology and disease with an emphasis on findings in experimental models using both high-affinity ligands and null-mouse models.
Collapse
Affiliation(s)
- Jeffrey M Peters
- Department of Veterinary and Biomedical Sciences and The Center of Molecular Toxicology and Carcinogenesis, Pennsylvania State University, University Park, PA 16802, USA.
| | | | | |
Collapse
|