51
|
Mazzola C, Medalie J, Scherma M, Panlilio LV, Solinas M, Tanda G, Drago F, Cadet JL, Goldberg SR, Yasar S. Fatty acid amide hydrolase (FAAH) inhibition enhances memory acquisition through activation of PPAR-alpha nuclear receptors. Learn Mem 2009; 16:332-7. [PMID: 19403796 DOI: 10.1101/lm.1145209] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Inhibitors of fatty acid amide hydrolase (FAAH) increase endogenous levels of anandamide (a cannabinoid CB(1)-receptor ligand) and oleoylethanolamide and palmitoylethanolamide (OEA and PEA, ligands for alpha-type peroxisome proliferator-activated nuclear receptors, PPAR-alpha) when and where they are naturally released in the brain. Using a passive-avoidance task in rats, we found that memory acquisition was enhanced by the FAAH inhibitor URB597 or by the PPAR-alpha agonist WY14643, and these enhancements were blocked by the PPAR-alpha antagonist MK886. These findings demonstrate novel mechanisms for memory enhancement by activation of PPAR-alpha, either directly by administering a PPAR-alpha agonist or indirectly by administering a FAAH inhibitor.
Collapse
Affiliation(s)
- Carmen Mazzola
- Preclinical Pharmacology Section, Behavioral Neuroscience Research Branch, Intramural Research Program, NIDA, NIH, DHHS, Baltimore, Maryland 21224, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
52
|
MK-886, an inhibitor of the 5-lipoxygenase-activating protein, inhibits cyclooxygenase-1 activity and suppresses platelet aggregation. Eur J Pharmacol 2009; 608:84-90. [DOI: 10.1016/j.ejphar.2009.02.023] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2008] [Revised: 02/03/2009] [Accepted: 02/12/2009] [Indexed: 01/09/2023]
|
53
|
Combination Therapy of PPARgamma Ligands and Inhibitors of Arachidonic Acid in Lung Cancer. PPAR Res 2009; 2008:750238. [PMID: 19277204 PMCID: PMC2652614 DOI: 10.1155/2008/750238] [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] [Received: 06/17/2008] [Revised: 11/26/2008] [Accepted: 12/03/2008] [Indexed: 12/27/2022] Open
Abstract
Lung cancer is the leading cause of cancer death in the United States and five-year survival remains low. Numerous studies have shown that chronic inflammation may lead to progression of carcinogenesis. As a result of inflammatory stimulation, arachidonic acid (AA) metabolism produces proliferation mediators through complex and dynamic interactions of the products of the LOX/COX enzymes. One important mediator in the activation of the AA pathways is the nuclear protein PPARgamma. Targeting LOX/COX enzymes and inducing activation of PPARgamma have resulted in significant reduction of cell growth in lung cancer cell lines. However, specific COX-inhibitors have been correlated with an increased cardiovascular risk. Clinical applications are still being explored with a novel generation of dual LOX/COX inhibitors. PPARgamma activation through synthetic ligands (TZDs) has revealed a great mechanistic complexity since effects are produced through PPARgamma-dependent and -independent mechanisms. Furthermore, PPARgamma could also be involved in regulation of COX-2. Overexpression of PPARgamma has reported to play a role in control of invasion and differentiation. Exploring the function of PPARgamma, in this new context, may provide a better mechanistic model of its role in cancer and give an opportunity to design a more efficient therapeutic approach in combination with LOX/COX inhibitors.
Collapse
|
54
|
Cheng G, Zhang X, Gao D, Jiang X, Dong W. Resveratrol inhibits MMP-9 expression by up-regulating PPAR alpha expression in an oxygen glucose deprivation-exposed neuron model. Neurosci Lett 2008; 451:105-8. [PMID: 19135132 DOI: 10.1016/j.neulet.2008.12.045] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2008] [Revised: 12/20/2008] [Accepted: 12/23/2008] [Indexed: 12/20/2022]
Abstract
Resveratrol (trans-3,4',5-trihydroxystilbene, Res) is a natural polyphenol. A recent experiment confirmed that Res can selectively activate both peroxisome proliferators-activated receptors (PPAR) alpha and gamma. In addition, Res can protect neurons by matrix metalloproteinase-9 (MMP-9) down-regulation. The relationship between Res, MMP-9 and PPAR alpha or gamma was studied in an oxygen glucose deprivation-exposed neuron model. It showed that Res can inhibit mRNA and protein expression of MMP-9, while it up-regulates the expression of PPAR alpha and gamma. The effect of Res on both PPAR alpha and MMP-9 can be offset partially by MK886. However, PPAR gamma antagonist GW9662 had little effect on MMP-9 expression. These results suggest that Res can inhibit MMP-9 expression by up-regulating PPAR alpha.
Collapse
Affiliation(s)
- Gang Cheng
- Institute of Neurosurgery, Xijing Hospital, Fourth Military Medical University, 15# West Chang Le Road, Xi'an, Shaanxi Province 710032, PR China
| | | | | | | | | |
Collapse
|
55
|
Kimura T, Mogi C, Tomura H, Kuwabara A, Im DS, Sato K, Kurose H, Murakami M, Okajima F. Induction of scavenger receptor class B type I is critical for simvastatin enhancement of high-density lipoprotein-induced anti-inflammatory actions in endothelial cells. THE JOURNAL OF IMMUNOLOGY 2008; 181:7332-40. [PMID: 18981156 DOI: 10.4049/jimmunol.181.10.7332] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Changes in plasma lipoprotein profiles, especially low levels of high-density lipoprotein (HDL), are a common biomarker for several inflammatory and immune diseases, including atherosclerosis and rheumatoid arthritis. We examined the effect of simvastatin on HDL-induced anti-inflammatory actions. HDL and sphingosine 1-phosphate (S1P), a bioactive lipid component of the lipoprotein, inhibited TNF alpha-induced expression of VCAM-1, which was associated with NO synthase (NOS) activation, in human umbilical venous endothelial cells. The HDL- but not S1P-induced anti-inflammatory actions were enhanced by a prior treatment of the cells with simvastatin in a manner sensitive to mevalonic acid. Simvastatin stimulated the expression of scavenger receptor class B type I (SR-BI) and endothelial NOS. As for S1P receptors, however, the statin inhibited the expression of S1P(3) receptor mRNA but caused no detectable change in S1P(1) receptor expression. The reconstituted HDL, a stimulator of SR-BI, mimicked HDL actions in a simvastatin-sensitive manner. The HDL- and reconstituted HDL-induced actions were blocked by small interfering RNA specific to SR-BI regardless of simvastatin treatment. The statin-induced expression of SR-BI was attenuated by constitutively active RhoA and small interfering RNA specific to peroxisome proliferator-activated receptor-alpha. Administration of simvastatin in vivo stimulated endothelial SR-BI expression, which was accompanied by the inhibition of the ex vivo monocyte adhesion in aortas from TNF alpha-injected mice. In conclusion, simvastatin induces endothelial SR-BI expression through a RhoA- and peroxisome proliferator-activated receptor-alpha-dependent mechanism, thereby enhancing the HDL-induced activation of NOS and the inhibition of adhesion molecule expression.
Collapse
Affiliation(s)
- Takao Kimura
- Laboratory of Signal Transduction, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
56
|
Cwinn MA, Jones SP, Kennedy SW. Exposure to perfluorooctane sulfonate or fenofibrate causes PPAR-alpha dependent transcriptional responses in chicken embryo hepatocytes. Comp Biochem Physiol C Toxicol Pharmacol 2008; 148:165-71. [PMID: 18565798 DOI: 10.1016/j.cbpc.2008.05.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2007] [Revised: 05/04/2008] [Accepted: 05/05/2008] [Indexed: 01/07/2023]
Abstract
Perfluorooctane sulfonate (PFOS) is a globally distributed environmental contaminant that is detected in the serum and liver of numerous mammalian and avian species. PFOS acts as a peroxisome proliferator in rodents, which occurs subsequent to activation of the nuclear receptor peroxisome proliferator activated receptor-alpha (PPAR-alpha). Activated PPAR-alpha up-regulates PPAR-alpha target genes, most of which are involved in lipid metabolism. Although several studies have investigated the effects of PFOS exposure on mammalian gene expression, there are few studies in avian species. To determine if PFOS is capable of activating avian PPAR-alpha, we exposed chicken embryo primary hepatocyte cultures (N=3 independent cell cultures) to PFOS or fenofibrate, a mammalian PPAR-alpha agonist, and examined the expression of PPAR-alpha and PPAR-alpha target genes using quantitative real-time PCR. The target genes examined were peroxisomal acyl-CoA oxidase (ACOX), liver fatty acid binding protein (L-FABP), enoyl-Coenzyme A, hydratase/3-hydroxyacyl Coenzyme A dehydrogenase bifunctional enzyme (BIEN), peroxisomal 3-ketoacyl thiolase (PKT), and malic enzyme (ME). All five target genes were induced in response to PFOS exposure and all of the target genes, except L-FABP, were induced in response to fenofibrate. PPAR-alpha mRNA expression was not altered by PFOS or fenofibrate. This study provides the first evidence that PFOS can induce PPAR-alpha-dependent transcriptional responses in an avian species and provides the first characterization of fenofibrate induced transcriptional responses in chicken embryo hepatocyte cultures.
Collapse
Affiliation(s)
- Matthew A Cwinn
- Department of Biology and Centre for Advanced Research in Environmental Genomics, University of Ottawa, Ottawa, ON, Canada K1N 6N5
| | | | | |
Collapse
|
57
|
Sasaki N, Egashira Y, Sanada H. Down-regulation of alpha-amino-beta-carboxymuconate-epsilon-semialdehyde decarboxylase by polyunsaturated fatty acids in hepatocytes is not mediated by PPARalpha. Eur J Nutr 2008; 47:80-6. [PMID: 18320257 DOI: 10.1007/s00394-008-0699-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2007] [Accepted: 02/13/2008] [Indexed: 01/11/2023]
Abstract
BACKGROUND alpha-Amino-beta-carboxymuconate-epsilon-semialdehyde decarboxylase (ACMSD) is a key enzyme in NAD biosynthesis from tryptophan. Dietary polyunsaturated fatty acids (PUFA) have been shown to suppress hepatic ACMSD activity and its mRNA level in rat. However the mechanism of the suppressive action has not been clarified yet. Although the phenomena that fatty acids suppress the expression of ACMSD in rat liver have been established in vivo experiment, it is still obscure whether the effect of fatty acids on the expression of the enzyme is caused by its direct or indirect action, because there have been very few investigations performed in vitro. AIM OF THE STUDY In this study, to examine whether down-regulation of ACMSD mRNA by PUFA involves peroxisome proliferator-activated receptor (PPAR) alpha mediated mechanism or not, we investigated the effect of PUFA on the ACMSD expression by using primary cultured rat hepatocytes. METHODS For this purpose we investigated the effect of PUFA (linoleic acid and eicosapentanoic acid) on the ACMSD mRNA level in primary-cultured rat hepatocytes and compared its effect with that of WY-14,643 (a PPARalpha agonist). After the incubation of hepatocytes with fatty acids, WY-14,643 and/or MK886 (a PPARalpha antagonist), mRNA levels of ACMSD and a peroxisome marker enzyme acyl-CoA oxidase (ACO) were determined by competitive reverse transcription-polymerase chain reaction (RT-PCR) method. RESULTS ACMSD mRNA level in primary hepatocytes were decreased by the incubation with high concentrations of linoleic acid, eicosapentaenoic acid (EPA) and WY-14,643. The appearance of ACO mRNA by WY-14,643 was remarkably increased, and those by linoleic acid and EPA were increased less than that by WY-14,643. Moreover, the suppression of ACMSD mRNA and the augmentation of ACO mRNA by WY-14,643 were inhibited by MK886, but the suppression by PUFA was not substantially affected by MK886. CONCLUSIONS The present study suggesting that the mechanism of decrease in ACMSD mRNA level by PUFA was different from that by WY-14,643, and that there would be any pathway other than PPARalpha mediated one for PUFA to regulate ACMSD expression.
Collapse
Affiliation(s)
- Naho Sasaki
- Graduate School of Science and Technology, Chiba University, 648 Mastudo, Mastudo-shi, Chiba 271 8510, Japan.
| | | | | |
Collapse
|
58
|
Andersson C, Zaman MM, Jones AB, Freedman SD. Alterations in immune response and PPAR/LXR regulation in cystic fibrosis macrophages. J Cyst Fibros 2008; 7:68-78. [PMID: 17889625 DOI: 10.1016/j.jcf.2007.05.004] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2006] [Revised: 04/06/2007] [Accepted: 05/11/2007] [Indexed: 10/22/2022]
Abstract
BACKGROUND Cystic fibrosis (CF) is characterized by an excessive inflammatory response in epithelial cells and macrophages. In CF mice, lung inflammation can be abrogated by oral treatment with docosahexaenoic acid (DHA). Since PPARs and LXRs are important regulators of inflammation and fatty acid metabolism in macrophages, we hypothesized that these pathways are dysregulated in CF macrophages and are corrected with DHA treatment. METHODS Peritoneal macrophages were obtained from wild type and cftr(-/-) mice. LPS induced cytokine secretion and NFkappaB activity were analyzed with and without oral DHA treatment. The expression and activity of PPARalpha,gamma, delta and LXRalpha were analyzed by RT-PCR and EMSA. RESULTS LPS induced TNFalpha and IL-6 secretion and NFkappaB p65 activity were increased in CF macrophages. This was associated with low basal PPARgamma expression and attenuated LPS induced induction of PPARdelta, LXRalpha and ABCA1. DHA pretreatment in vivo decreased TNFalpha secretion and p65 activity, and increased PPARalpha and gamma expression and function. The effects of DHA could be reproduced by PPAR agonists and blocked by a PPARalpha antagonist. CONCLUSION Impaired regulation of nuclear receptors may contribute to the abnormal LPS induced signaling in CF macrophages and is reversed by DHA.
Collapse
Affiliation(s)
- Charlotte Andersson
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston 02215, MA, United States
| | | | | | | |
Collapse
|
59
|
Sheldrick E, Derecka K, Marshall E, Chin E, Hodges L, Wathes D, Abayasekara D, Flint A. Peroxisome-proliferator-activated receptors and the control of levels of prostaglandin-endoperoxide synthase 2 by arachidonic acid in the bovine uterus. Biochem J 2007; 406:175-83. [PMID: 17516915 PMCID: PMC1948980 DOI: 10.1042/bj20070089] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Arachidonic acid is a potential paracrine agent released by the uterine endometrial epithelium to induce PTGS2 [PG (prostaglandin)-endoperoxide synthase 2] in the stroma. In the present study, bovine endometrial stromal cells were used to determine whether PTGS2 is induced by arachidonic acid in stromal cells, and to investigate the potential role of PPARs (peroxisome-proliferator-activated receptors) in this effect. Arachidonic acid increased PTGS2 levels up to 7.5-fold within 6 h. The cells expressed PPARalpha and PPARdelta (also known as PPARbeta) (but not PPARgamma). PTGS2 protein level was increased by PPAR agonists, including polyunsaturated fatty acids, synthetic PPAR ligands, PGA1 and NSAIDs (non-steroidal anti-inflammatory drugs) with a time course resembling that of arachidonic acid. Use of agonists and antagonists indicated PPARalpha (but not PPARdelta or PPARgamma) was responsible for PTGS2 induction. PTGS2 induction by arachidonic acid did not require PG synthesis. PTGS2 levels were increased by the PKC (protein kinase C) activators 4beta-PMA and PGF(2alpha), and the effects of arachidonic acid, NSAIDs, synthetic PPAR ligands and 4beta-PMA were blocked by PKC inhibitors. This is consistent with PPAR phosphorylation by PKC. Induction of PTGS2 protein by 4beta-PMA in the absence of a PPAR ligand was decreased by the NF-kappaB (nuclear factor kappaB) inhibitors MG132 and parthenolide, suggesting that PKC acted through NF-kappaB in addition to PPAR phosphorylation. Use of NF-kappaB inhibitors allowed the action of arachidonic acid as a PPAR agonist to be dissociated from an effect through PKC. The results are consistent with the hypothesis that arachidonic acid acts via PPARalpha to increase PTGS2 levels in bovine endometrial stromal cells.
Collapse
Affiliation(s)
- E. Linda R. Sheldrick
- *Division of Animal Physiology, School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough LE12 5RD, U.K
| | - Kamila Derecka
- *Division of Animal Physiology, School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough LE12 5RD, U.K
| | - Elaine Marshall
- *Division of Animal Physiology, School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough LE12 5RD, U.K
| | - Evonne C. Chin
- †Reproduction and Development Group, Royal Veterinary College, Hawkshead Lane, North Mymms, Hatfield AL9 7TA, U.K
| | - Louise Hodges
- †Reproduction and Development Group, Royal Veterinary College, Hawkshead Lane, North Mymms, Hatfield AL9 7TA, U.K
| | - D. Claire Wathes
- †Reproduction and Development Group, Royal Veterinary College, Hawkshead Lane, North Mymms, Hatfield AL9 7TA, U.K
| | - D. Robert E. Abayasekara
- †Reproduction and Development Group, Royal Veterinary College, Hawkshead Lane, North Mymms, Hatfield AL9 7TA, U.K
| | - Anthony P. F. Flint
- *Division of Animal Physiology, School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough LE12 5RD, U.K
- To whom correspondence should be addressed (email )
| |
Collapse
|
60
|
Zhang X, Li L, Prabhakaran K, Zhang L, Leavesley H, Borowitz J, Isom G. Uncoupling protein-2 up-regulation and enhanced cyanide toxicity are mediated by PPARalpha activation and oxidative stress. Toxicol Appl Pharmacol 2007; 223:10-9. [PMID: 17573087 PMCID: PMC1994772 DOI: 10.1016/j.taap.2007.05.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2007] [Revised: 05/07/2007] [Accepted: 05/09/2007] [Indexed: 12/17/2022]
Abstract
Uncoupling protein 2 (UCP-2) is an inner mitochondrial membrane proton carrier that modulates mitochondrial membrane potential (DeltaPsi(m)) and uncouples oxidative phosphorylation. We have shown that up-regulation of UCP-2 by Wy14,643, a selective peroxisome proliferator-activated receptor-alpha (PPARalpha) agonist, enhances cyanide cytotoxicity. The pathway by which Wy14,643 up-regulates UCP-2 was determined in a dopaminergic cell line (N27 cells). Since dopaminergic mesencephalic cells are a primary brain target of cyanide, the N27 immortalized mesencephalic cell was used in this study. Wy14,643 produced a concentration- and time-dependent up-regulation of UCP-2 that was linked to enhanced cyanide-induced cell death. MK886 (PPARalpha antagonist) or PPARalpha knock-down by RNA interference (RNAi) inhibited PPARalpha activity as shown by the peroxisome proliferator response element-luciferase reporter assay, but only partially decreased up-regulation of UCP-2. The role of oxidative stress as an alternative pathway to UCP-2 up-regulation was determined. Wy14,643 induced a rapid surge of ROS generation and loading cells with glutathione ethyl ester (GSH-EE) or pre-treatment with vitamin E attenuated up-regulation of UCP-2. On the other hand, RNAi knockdown of PPARalpha did not alter ROS generation, suggesting a PPARalpha-independent component to the response. Co-treatment with PPARalpha-RNAi and GSH-EE blocked both the up-regulation of UCP-2 by Wy14,643 and the cyanide-induced cell death. It was concluded that a PPARalpha-mediated pathway and an oxidative stress pathway independent of PPARalpha mediate the up-regulation of UCP-2 and subsequent increased vulnerability to cyanide-induced cytotoxicity.
Collapse
Affiliation(s)
- X. Zhang
- Department of Medicinal Chemistry & Molecular Pharmacology, Purdue University, West Lafayette, Indiana 47907-1333, USA
| | - L. Li
- Department of Medicinal Chemistry & Molecular Pharmacology, Purdue University, West Lafayette, Indiana 47907-1333, USA
| | - K. Prabhakaran
- Department of Medicinal Chemistry & Molecular Pharmacology, Purdue University, West Lafayette, Indiana 47907-1333, USA
| | - L. Zhang
- Department of Medicinal Chemistry & Molecular Pharmacology, Purdue University, West Lafayette, Indiana 47907-1333, USA
| | - H.B. Leavesley
- Department of Medicinal Chemistry & Molecular Pharmacology, Purdue University, West Lafayette, Indiana 47907-1333, USA
| | - J.L. Borowitz
- Department of Medicinal Chemistry & Molecular Pharmacology, Purdue University, West Lafayette, Indiana 47907-1333, USA
| | - G.E. Isom
- Department of Medicinal Chemistry & Molecular Pharmacology, Purdue University, West Lafayette, Indiana 47907-1333, USA
| |
Collapse
|
61
|
Wallace VCJ, Segerdahl AR, Lambert DM, Vandevoorde S, Blackbeard J, Pheby T, Hasnie F, Rice ASC. The effect of the palmitoylethanolamide analogue, palmitoylallylamide (L-29) on pain behaviour in rodent models of neuropathy. Br J Pharmacol 2007; 151:1117-28. [PMID: 17558434 PMCID: PMC2042941 DOI: 10.1038/sj.bjp.0707326] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND AND PURPOSE Cannabinoids are associated with analgesia in acute and chronic pain states. A spectrum of central cannabinoid (CB(1)) receptor-mediated motor and psychotropic side effects limit their therapeutic potential. Here, we investigate the analgesic effect of the palmitoylethanolamide (PEA) analogue, palmitoylallylamide (L-29), which via inhibition of fatty acid amide hydrolase (FAAH) may potentiate endocannabinoids thereby avoiding psychotropic side effects. EXPERIMENTAL APPROACH The in vivo analysis of the effect of L-29 on measures of pain behaviour in three rat models of neuropathic pain. KEY RESULTS Systemically administered L-29 (10 mg kg(-1)) reduced hypersensitivity to mechanical and thermal stimuli in the partial sciatic nerve injury (PSNI) model of neuropathic pain; and mechanical hypersensitivity in a model of antiretroviral (ddC)-associated hypersensitivity and a model of varicella zoster virus (VZV)-associated hypersensitivity. The effects of L-29 were comparable to those of gabapentin (50 mg kg(-1)). The CB(1) receptor antagonist SR141716a (1 mg kg(-1)) and the CB(2) receptor antagonist SR144528 (1 mg kg(-1)) reduced the effect of L-29 on hypersensitivity in the PSNI and ddC models, but not in the VZV model. The peroxisome proliferator-activated receptor-alpha antagonist, MK-886 (1 mg kg(-1)), partially attenuated the effect of L-29 on hypersensitivity in the PSNI model. L-29 (10 mg kg(-1)) significantly attenuated thigmotactic behaviour in the open field arena without effect on locomotor activity. CONCLUSIONS AND IMPLICATIONS L-29 produces analgesia in a range of neuropathic pain models. This presents L-29 as a novel analgesic compound that may target the endogenous cannabinoid system while avoiding undesirable side effects associated with direct cannabinoid receptor activation.
Collapse
Affiliation(s)
- V C J Wallace
- Pain Research Group, Department of Anaesthetics, Pain Medicine and Intensive Care, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital Campus London, UK
| | - A R Segerdahl
- Pain Research Group, Department of Anaesthetics, Pain Medicine and Intensive Care, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital Campus London, UK
| | - D M Lambert
- Unite de Chimie Pharmaceutique et de Radiopharmacie, Universite Catholique de Louvain, Avenue Mounier Brussels, Belgium
| | - S Vandevoorde
- Unite de Chimie Pharmaceutique et de Radiopharmacie, Universite Catholique de Louvain, Avenue Mounier Brussels, Belgium
| | - J Blackbeard
- Pain Research Group, Department of Anaesthetics, Pain Medicine and Intensive Care, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital Campus London, UK
| | - T Pheby
- Pain Research Group, Department of Anaesthetics, Pain Medicine and Intensive Care, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital Campus London, UK
| | - F Hasnie
- Pain Research Group, Department of Anaesthetics, Pain Medicine and Intensive Care, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital Campus London, UK
| | - A S C Rice
- Pain Research Group, Department of Anaesthetics, Pain Medicine and Intensive Care, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital Campus London, UK
- Author for correspondence:
| |
Collapse
|
62
|
Dubrac S, Stoitzner P, Pirkebner D, Elentner A, Schoonjans K, Auwerx J, Saeland S, Hengster P, Fritsch P, Romani N, Schmuth M. Peroxisome proliferator-activated receptor-alpha activation inhibits Langerhans cell function. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2007; 178:4362-72. [PMID: 17371993 DOI: 10.4049/jimmunol.178.7.4362] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Epidermal Langerhans cells (LC) play a pivotal role in initiating and maintaining primary immune responses in the skin. In the present study, we asked whether peroxisome proliferator-activated receptor-alpha (PPARalpha) activation modulates LC function. Our results show that PPARalpha is expressed in immature LC and is down-regulated in mature LC suggesting that an early decrease of PPARalpha expression in LC may allow them to mature after contact with an Ag. We further show that pharmacologic PPARalpha activation inhibits LC maturation, migratory capacity, cytokine expression, and the ability to drive T cell proliferation. Moreover, PPARalpha activation inhibits NF-kappaB but not stress-activated protein kinase/JNK, p38MAPK, and ERK1/2. In conclusion, PPARalpha activation by endogenous ligands may provide a molecular signal that allows LC to remain in an immature state within the epidermis for extended periods of time despite minor environmental stimuli.
Collapse
Affiliation(s)
- Sandrine Dubrac
- Department of Dermatology, Innsbruck Medical University, Anichstrasse 35, A-6020 Innsbruck, Austria.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
63
|
Becker J, Delayre-Orthez C, Frossard N, Pons F. Regulation of inflammation by PPARs: a future approach to treat lung inflammatory diseases? Fundam Clin Pharmacol 2007; 20:429-47. [PMID: 16968414 DOI: 10.1111/j.1472-8206.2006.00425.x] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Lung inflammatory diseases, such as acute lung injury (ALI), asthma, chronic obstructive pulmonary disease (COPD) and lung fibrosis, represent a major health problem worldwide. Although glucocorticoids are the most potent anti-inflammatory drug in asthma, they exhibit major side effects and have poor activity in lung inflammatory disorders such as ALI or COPD. Therefore, there is growing need for the development of alternative or new therapies to treat inflammation in the lung. Peroxisome proliferator-activated receptors (PPARs), including the three isotypes PPARalpha, PPARbeta (or PPARdelta) and PPARgamma, are transcription factors belonging to the nuclear hormone receptor superfamily. PPARs, and in particular PPARalpha and PPARgamma, are well known for their critical role in the regulation of energy homeostasis by controlling expression of a variety of genes involved in lipid and carbohydrate metabolism. Synthetic ligands of the two receptor isotypes, the fibrates and the thiazolidinediones, are clinically used to treat dyslipidaemia and type 2 diabetes, respectively. Recently however, PPARalpha and PPARgamma have been shown to exert a potent anti-inflammatory activity, mainly through their ability to downregulate pro-inflammatory gene expression and inflammatory cell functions. The present article reviews the current knowledge of the role of PPARalpha and PPARgamma in controlling inflammation, and presents different findings suggesting that PPARalpha and PPARgamma activators may be helpful in the treatment of lung inflammatory diseases.
Collapse
Affiliation(s)
- Julien Becker
- EA 3771 Inflammation et environnement dans l'asthme, Faculté de Pharmacie, Université Louis Pasteur-Strasbourg I, Illkirch, France
| | | | | | | |
Collapse
|
64
|
Kim J, Ahn JH, Kim JH, Yu YS, Kim HS, Ha J, Shinn SH, Oh YS. Fenofibrate regulates retinal endothelial cell survival through the AMPK signal transduction pathway. Exp Eye Res 2007; 84:886-93. [PMID: 17343853 DOI: 10.1016/j.exer.2007.01.009] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2006] [Revised: 12/20/2006] [Accepted: 01/09/2007] [Indexed: 01/07/2023]
Abstract
Fenofibrate, a widely used hypolipidemic drug, has anti-inflammatory and anti-atherosclerotic effects in the vessel wall. In the present study, we report an anti-apoptotic property of fenofibrate in human retinal endothelial cells (HRECs) and describe an underlying molecular mechanism. Treatment with fenofibrate protected HRECs from apoptosis in response to serum deprivation in a dose-dependent manner. This inhibition of apoptosis by fenofibrate was not altered by peroxisome proliferator-activated receptor alpha (PPARalpha) antagonist MK 886, and selective agonist for PPARalpha, WY-14643 had no beneficial effects on serum deprivation-induced cell death. Fenofibrate potently induced a sustained activation of AMP-activated protein kinase (AMPK) and vascular endothelial growth factor (VEGF) mRNA expression. Furthermore, compound C, a specific AMPK inhibitor, almost completely blocked the fenofibrate-induced survival effect as well as VEGF mRNA expression. Taken together, these results suggest that fenofibrate prevents apoptotic cell death induced by serum deprivation through PPARalpha-independent, but AMPK-dependent pathway. Thus fenofibrate may have a novel therapeutic property that can control unwanted cell death found in diabetic retinopathy.
Collapse
Affiliation(s)
- Jaetaek Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Chung-Ang University, 224-1 Heuk Seok-dong, Dongjak-ku, Seoul 156-755, South Korea.
| | | | | | | | | | | | | | | |
Collapse
|
65
|
Magro AM, Magro AD, Cunningham C, Miller MR. Down-regulation of vinculin upon MK886-induced apoptosis in LN18 glioblastoma cells. Neoplasma 2007; 54:517-26. [PMID: 17949236 PMCID: PMC4320946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Glioblastomas are a type of malignant brain tumor and are among the most difficult cancers to treat. One strategy to treat aggressive cancers is the use of drugs that target multiple signaling pathways. MK886 is a drug known to inhibit both 5- lipoxygenase-activating-protein (FLAP) and peroxisome proliferator activated receptor-alpha (PPAR-alpha). The objectives of this study were to investigate the ability of MK886 to induce apoptotic cell death in LN18 glioblastoma cells and to characterize the cell death mechanisms. MK886 induced massive apoptotic LN18 cell death that was manifested by the release of nucleosomes, annexinV binding to phosphatidylserine in the absence of nuclear staining, and changes in the fluorescent intensity of Mito Tracker Deep Red 633 indicating changes in mitochondrial oxidative function and mass. The alteration of the mitochondrial function implied that MK886 induced apoptosis in LN18 cells via a mitochondrial pathway. The broad caspases inhibitor ZVAD-FMK inhibited MK886-induced nucleosome release, but not annexinV binding or MK886-altered mitochondrial function. Real time RT-PCR demonstrated that LN18 cells expressed significant levels of FLAP and PPAR- alpha mRNAs. A low level of arachidonate 5-lipoxygenase (ALOX-5) mRNA was detected, but little, if any, arachidonate 12- lipoxygenase (ALOX-12) mRNA was present. In addition, MK886-induced apoptosis in LN18 cells was accompanied by a decrease in the protein and mRNA levels of vinculin, but not other focal adhesion proteins. In summary, the data presented here indicate that disruption of the actin-vinculin-cell-cytoskeleton matrix of the LN18 glioblastoma is a component of the MK886 induced apoptosis. In addition, MK886 treated LN18 cells could provide one model in which to investigate drugs that target lipoxygenase and PPAR-alpha pathways in the chemotherapeutic treatment of glioblastomas.
Collapse
Affiliation(s)
- A M Magro
- Department of Biology, Fairmont University, Fairmont, WV 26554, USA.
| | | | | | | |
Collapse
|
66
|
Michalik L, Auwerx J, Berger JP, Chatterjee VK, Glass CK, Gonzalez FJ, Grimaldi PA, Kadowaki T, Lazar MA, O'Rahilly S, Palmer CNA, Plutzky J, Reddy JK, Spiegelman BM, Staels B, Wahli W. International Union of Pharmacology. LXI. Peroxisome proliferator-activated receptors. Pharmacol Rev 2006; 58:726-41. [PMID: 17132851 DOI: 10.1124/pr.58.4.5] [Citation(s) in RCA: 739] [Impact Index Per Article: 38.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The three peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors of the nuclear hormone receptor superfamily. They share a high degree of structural homology with all members of the superfamily, particularly in the DNA-binding domain and ligand- and cofactor-binding domain. Many cellular and systemic roles have been attributed to these receptors, reaching far beyond the stimulation of peroxisome proliferation in rodents after which they were initially named. PPARs exhibit broad, isotype-specific tissue expression patterns. PPARalpha is expressed at high levels in organs with significant catabolism of fatty acids. PPARbeta/delta has the broadest expression pattern, and the levels of expression in certain tissues depend on the extent of cell proliferation and differentiation. PPARgamma is expressed as two isoforms, of which PPARgamma2 is found at high levels in the adipose tissues, whereas PPARgamma1 has a broader expression pattern. Transcriptional regulation by PPARs requires heterodimerization with the retinoid X receptor (RXR). When activated by a ligand, the dimer modulates transcription via binding to a specific DNA sequence element called a peroxisome proliferator response element (PPRE) in the promoter region of target genes. A wide variety of natural or synthetic compounds was identified as PPAR ligands. Among the synthetic ligands, the lipid-lowering drugs, fibrates, and the insulin sensitizers, thiazolidinediones, are PPARalpha and PPARgamma agonists, respectively, which underscores the important role of PPARs as therapeutic targets. Transcriptional control by PPAR/RXR heterodimers also requires interaction with coregulator complexes. Thus, selective action of PPARs in vivo results from the interplay at a given time point between expression levels of each of the three PPAR and RXR isotypes, affinity for a specific promoter PPRE, and ligand and cofactor availabilities.
Collapse
Affiliation(s)
- Liliane Michalik
- Center for Integrative Genomics, National Research Centre "Frontiers in Genetics," University of Lausanne, Lausanne, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
67
|
Huang THW, Li Y, Razmovski-Naumovski V, Tran VH, Li GQ, Duke CC, Roufogalis BD. Gypenoside XLIX isolated from Gynostemma pentaphyllum inhibits nuclear factor-kappaB activation via a PPAR-alpha-dependent pathway. J Biomed Sci 2006; 13:535-48. [PMID: 16525884 DOI: 10.1007/s11373-006-9076-8] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2005] [Accepted: 01/26/2006] [Indexed: 01/10/2023] Open
Abstract
Nuclear factor (NF)-kappaB is important in the generation of inflammation. Besides regulating lipid metabolism, peroxisome proliferator-activated receptor (PPAR)-alpha activators also reduce NF-kappaB activation to terminate activation of inflammatory pathways. Gynostemma pentaphyllum (GP) has been used to treat various inflammatory diseases and hyperlipidemia. Here, we demonstrate that GP extract and one of its main components, Gypenoside XLIX (Gyp-XLIX) inhibited LPS-induced NF-kappaB activation in murine macrophages. Furthermore, Gyp-XLIX restored the LPS- and TNF-alpha-induced decrease in cytosolic I-kappaBalpha protein expression and inhibited the translocation of NF-kappaB(p65) to the nucleus in THP-1 monocyte and HUVEC cells. The inhibition of LPS- and TNF-alpha-induced NF-kappaB luciferase activity in macrophages was abolished by MK-886, a selective PPAR-alpha antagonist. GP extract and Gyp-XLIX (EC(50): 10.1 microM) enhanced PPAR-alpha luciferase activity in HEK293 cells transfected with the tK-PPREx3-Luc reporter plasmid and expression vectors for PPAR-alpha. Additionally, Gyp-XLIX specifically enhanced PPAR-alpha mRNA and protein expression in THP-1-derived macrophage cells. The selectivity of Gyp-XLIX for PPAR-alpha was demonstrated by the activation of only PPAR-alpha in HEK293 cells transfected with expression vectors for PPAR-alpha, PPAR-beta/delta or PPAR-gamma1 plasmids and in THP-1-derived macrophage naturally expressing all three PPAR isoforms. The present study demonstrates that Gyp-XLIX, a naturally occurring gynosaponin, inhibits NF-kappaB activation via a PPAR-alpha-dependent pathway.
Collapse
Affiliation(s)
- Tom Hsun-Wei Huang
- Pharmaceutical Chemistry Discipline and Herbal Medicines Research and Education Centre, Faculty of Pharmacy, University of Sydney, A15 S322, Sydney, NSW 2006, Australia
| | | | | | | | | | | | | |
Collapse
|
68
|
Sabirsh A, Bristulf J, Karlsson U, Owman C, Haeggström JZ. Non-specific effects of leukotriene synthesis inhibitors on HeLa cell physiology. Prostaglandins Leukot Essent Fatty Acids 2005; 73:431-40. [PMID: 16171988 DOI: 10.1016/j.plefa.2005.08.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2005] [Revised: 08/05/2005] [Accepted: 08/07/2005] [Indexed: 10/25/2022]
Abstract
We examined the effects of various leukotriene synthesis inhibitors on calcium signalling in HeLa cells, before and after transfection with BLT(1). All of the inhibitors studied were found to reduce increases in intracellular calcium concentration induced by BLT(1), but also by an ionophore or activation of various G-protein coupled receptors, regardless of BLT(1) expression. In order to explore the mechanism of these apparently general effects we examined HeLa cell expression of leukotriene receptors and biosynthetic enzymes and found that the genes for key leukotriene synthesis enzymes and all of the leukotriene receptors were not expressed. Leukotrienes are involved in the pathology of a variety of cancers, and for HeLa cells leukotrienes have been reported to be important for aspects of the carcinogenic phenotype. We find that leukotriene synthesis inhibitors have non-specific effects, so careful controls are necessary to avoid interpreting non-specific effects as evidence for leukotriene involvement.
Collapse
Affiliation(s)
- Alan Sabirsh
- Division of Physiological Chemistry II, Department of Medical Biochemistry, Biophysics, Karolinska Institute, Sweden.
| | | | | | | | | |
Collapse
|
69
|
Huang THW, Peng G, Kota BP, Li GQ, Yamahara J, Roufogalis BD, Li Y. Pomegranate flower improves cardiac lipid metabolism in a diabetic rat model: role of lowering circulating lipids. Br J Pharmacol 2005; 145:767-74. [PMID: 15880139 PMCID: PMC1576197 DOI: 10.1038/sj.bjp.0706245] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Excess triglyceride (TG) accumulation and increased fatty acid (FA) oxidation in the diabetic heart contribute to cardiac dysfunction. Punica granatum flower (PGF) is a traditional antidiabetic medicine. Here, we investigated the effects and mechanisms of action of PGF extract on abnormal cardiac lipid metabolism both in vivo and in vitro. Long-term oral administration of PGF extract (500 mg kg(-1)) reduced cardiac TG content, accompanied by a decrease in plasma levels of TG and total cholesterol in Zucker diabetic fatty (ZDF) rats, indicating improvement by PGF extract of abnormal cardiac TG accumulation and hyperlipidemia in this diabetic model. Treatment of ZDF rats with PGF extract lowered plasma FA levels. Furthermore, the treatment suppressed cardiac overexpression of mRNAs encoding for FA transport protein, peroxisome proliferator-activated receptor (PPAR)-alpha, carnitine palmitoyltransferase-1, acyl-CoA oxidase and 5'-AMP-activated protein kinase alpha2, and restored downregulated cardiac acetyl-CoA carboxylase mRNA expression in ZDF rats, whereas it showed little effect in Zucker lean rats. The results suggest that PGF extract inhibits increased cardiac FA uptake and oxidation in the diabetic condition. PGF extract and its component oleanolic acid enhanced PPAR-alpha luciferase reporter gene activity in human embryonic kidney 293 cells, and this effect was completely suppressed by a selective PPAR-alpha antagonist MK-886, consistent with the presence of PPAR-alpha activator activity in the extract and this component. Our findings suggest that PGF extract improves abnormal cardiac lipid metabolism in ZDF rats by activating PPAR-alpha and thereby lowering circulating lipid and inhibiting its cardiac uptake.
Collapse
Affiliation(s)
- Tom Hsun-Wei Huang
- Herbal Medicines Research and Education Centre, Faculty of Pharmacy, The University of Sydney, NSW, 2006, Australia
| | - Gang Peng
- Herbal Medicines Research and Education Centre, Faculty of Pharmacy, The University of Sydney, NSW, 2006, Australia
| | - Bhavani Prasad Kota
- Herbal Medicines Research and Education Centre, Faculty of Pharmacy, The University of Sydney, NSW, 2006, Australia
| | - George Qian Li
- Herbal Medicines Research and Education Centre, Faculty of Pharmacy, The University of Sydney, NSW, 2006, Australia
| | | | - Basil D Roufogalis
- Herbal Medicines Research and Education Centre, Faculty of Pharmacy, The University of Sydney, NSW, 2006, Australia
| | - Yuhao Li
- Herbal Medicines Research and Education Centre, Faculty of Pharmacy, The University of Sydney, NSW, 2006, Australia
- Author for correspondence:
| |
Collapse
|
70
|
Wang Y, Wang Y, Yang Q, Yan JT, Zhao C, Cianflone K, Wang DW. Effects of bezafibrate on the expression of endothelial nitric oxide synthase gene and its mechanisms in cultured bovine endothelial cells. Atherosclerosis 2005; 187:265-73. [PMID: 16256120 DOI: 10.1016/j.atherosclerosis.2005.09.008] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2005] [Revised: 08/16/2005] [Accepted: 09/09/2005] [Indexed: 01/13/2023]
Abstract
OBJECTIVE Peroxisome proliferator-activated receptors alpha (PPARalpha) is a target gene for atherosclerosis and cardiovascular diseases. However, effects of PPARalpha on endothelial nitric oxide synthase (eNOS) remain unknown. We investigated the eNOS regulation by bezafibrate, a ligand of PPARalpha, and involved signaling pathways. METHODS AND RESULTS Firstly, in cultured bovine aorta endothelial cells (BAEC), bezafibrate significantly upregulated eNOS at protein, mRNA levels and NO production, respectively, in a concentration-dependent fashion (50-200muM). Next, the effects of bezafibrate on signal pathways and eNOS mRNA stability in BAEC were investigated. Results showed that bezafibrate induced phosphorylation of MAPK. Inhibitors of PPARalpha, PI3 kinase and MAPK, respectively, markedly attenuate bezafibrate-induced upregulation of eNOS. Bezafibrate incubation increased eNOS mRNA half-life, activated eNOS promoter, enhanced phosphorylation of eNOS ser-1179 site, and decreased phosphorylation of eNOS thr-497 site via activating ERK and Akt. CONCLUSIONS Bezafibrate can upregulate eNOS expression, enhance phosphorylation of eNOS ser-1179, increase NO production and transcription level and stability of eNOS mRNA through pathway dependent of PPARalpha and nongenomic effects mediated by MAPK and PI3K pathways. Hence, PPARalpha ligands exert direct benefits on vessel endothelial functions through an increase in eNOS expression level and phosphorylation of eNOS ser-1179. This mechanism provides additional anti-atherosclerotic and anti-hypertension benefits of bezafibrate in addition of lipid-lowering effects.
Collapse
Affiliation(s)
- Yan Wang
- Cardiovascular Division of Internal Medicine, Department and The Institute of Hypertension, Tongji Hospital, Tongji Medical College of Huazhong University of Science & Technology, Wuhan 430030, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
71
|
Atarod EB, Kehrer JP. Dissociation of oxidant production by peroxisome proliferator-activated receptor ligands from cell death in human cell lines. Free Radic Biol Med 2004; 37:36-47. [PMID: 15183193 DOI: 10.1016/j.freeradbiomed.2004.04.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2003] [Revised: 03/31/2004] [Accepted: 04/16/2004] [Indexed: 01/16/2023]
Abstract
Ligands of peroxisome proliferator-activated receptors (PPARs) come from a diverse group of chemicals that include pharmaceutical drugs, phthalate plasticizers, steroids, and pesticides. PPAR ligands exhibit a number of effects, including an ability to induce apoptosis in some systems. The mechanism(s) underlying the induction of apoptosis is not known. The current study examined the ability of Wy14643, a fibrate and PPARalpha agonist, and ciglitazone, a thiazolidinedione and PPARgamma agonist, to induce apoptosis as well as the production of oxidants in human Jurkat T cells that express all PPAR isoforms. Treatment with increasing doses of Wy14643 caused a substantial time-dependent increase in the overall oxidant status (as reflected by increased dichlorofluorescein fluorescence) of Jurkat cells without any change in viability except at the highest dose and longest time. Ciglitazone also caused a dose- and time-dependent increase in oxidant production. However, although the extent of this production was less than that seen with Wy14643, ciglitazone caused a dose- and time-dependent increase in apoptosis that could not be inhibited by antioxidants. Confocal micrographs of Jurkat cells loaded with dichlorofluorescein diacetate or dihydrorhodamine 123 and treated with Wy14643 or ciglitazone revealed a punctate pattern of fluorescence at early time points suggestive of a mitochondrial origin for these oxidants. Rotenone and antimycin A prevented Wy14643- but not ciglitazone-induced oxidant production. Other relatively specific PPARgamma agonists (15delta-PGJ2, and troglitazone), but not nonspecific agonists (bezafibrate and conjugated linoleic acid), were also able to induce oxidant production in Jurkat cells. These data, as well as the findings that oxidant production could be induced by Wy14643 in A549 cells that lack PPARalpha, and could not be blocked in Jurkat cells by the PPARalpha inhibitor MK886, indicate oxidant formation is unrelated to PPARalpha. These data also suggest that oxidant production induced by PPARalpha ligands originates in the mitochondria.
Collapse
Affiliation(s)
- Elisa B Atarod
- Division of Pharmacology and Toxicology, College of Pharmacy, University of Texas at Austin, Austin, TX 78712-0125, USA
| | | |
Collapse
|
72
|
Bellini MJ, Polo MP, de Alaniz MJT, de Bravo MG. Effect of simvastatin on the uptake and metabolic conversion of palmitic, dihomo-gamma-linoleic and alpha-linolenic acids in A549 cells. Prostaglandins Leukot Essent Fatty Acids 2003; 69:351-7. [PMID: 14580370 DOI: 10.1016/s0952-3278(03)00149-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
It is well known that simvastatin affects cholesterol synthesis. Furthermore it inhibits growth and proliferation and perturbs fatty acid metabolism in some cell lines. We have studied the effects of simvastatin on the uptake and metabolism of exogenous fatty acid in the human lung adenocarcinoma A549 cells. Simvastatin inhibited the proliferation of A549, and caused an increment in phospholipid/cholesterol ratio due to an increment in phospholipid content without affecting cholesterol content. All the fatty acids were uptaken and metabolized in both control and treated cells. The conversion of palmitic, linoleic and dihomo-gamma-linoleic acids to their metabolites and products/precursor ratios for the desaturation and elongation reactions showed that simvastatin enhanced the Delta5 desaturation step and altered some elongating steps. The machinery for unsaturated fatty acid synthesis in A549 is quite sensitive to simvastatin and its effects could have important implication taking into account that highly unsaturated fatty acids are involved in the regulation of diverse cellular functions by themselves or through their metabolites.
Collapse
Affiliation(s)
- M J Bellini
- Instituto de Investigaciones Bioqui;micas de La Plata (INIBIOLP), CONICET-UNLP, Facultad de Ciencias Médicas, Calle 60 y 120, La Plata 1900, Argentina
| | | | | | | |
Collapse
|
73
|
Tong Z, Wu X, Kehrer JP. Increased expression of the lipocalin 24p3 as an apoptotic mechanism for MK886. Biochem J 2003; 372:203-10. [PMID: 12614196 PMCID: PMC1223388 DOI: 10.1042/bj20021696] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2002] [Revised: 02/04/2003] [Accepted: 03/04/2003] [Indexed: 01/06/2023]
Abstract
MK886, a strong proapoptotic agent, is an inhibitor of 5-lipoxygenase (LOX) through binding to the 5-LOX-activating protein (FLAP). Although MK886-induced apoptosis is through a FLAP-independent pathway, the precise mechanisms are not understood. In the present study, a possible role of 24p3, a lipocalin, in MK886-induced apoptosis was investigated. Exposure of murine prolymphoid progenitor cells (FL5.12) to 20 microM MK886 for 16 h dramatically increased 24p3 mRNA and protein expression. Induction could also be achieved with another FLAP inhibitor, MK591. The induction of 24p3 by MK886 was dose- and time-dependent. The up-regulated 24p3 mRNA expression by MK886 was enhanced a further 3.1-fold by WY14643, an activator of peroxisome-proliferator-activated receptor alpha, whereas ciglitazone, an activator of peroxisome-proliferator-activated receptor gamma attenuated the MK886-induced 24p3 expression by more than 50%. Neither WY14643 nor ciglitazone alone had any effect on the expression of 24p3. The induction of 24p3 by MK886 was dependent on the synthesis of new protein(s), since cycloheximide, an inhibitor of protein synthesis, prevented this effect. In all cases, including the inhibition of MK886-induced 24p3 protein expression by stable transfection with antisense cDNA of 24p3, the extent of apoptosis closely paralleled 24p3 levels. Apoptosis induced by MK886, or enhanced by WY14643, was accompanied by the cleavage and activation of caspase-3. The overexpression of bcl-2 or bcl-x(L) in FL5.12 cells inhibited apoptosis induced by MK886 as well as the enhancement of apoptosis by WY14643. Thus 24p3 is an MK886-inducible gene and may play an important role in MK886-induced apoptosis.
Collapse
Affiliation(s)
- Zhimin Tong
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712-0125, U.S.A
| | | | | |
Collapse
|
74
|
Sciulli MG, Seta F, Tacconelli S, Capone ML, Ricciotti E, Pistritto G, Patrignani P. Effects of acetaminophen on constitutive and inducible prostanoid biosynthesis in human blood cells. Br J Pharmacol 2003; 138:634-41. [PMID: 12598417 PMCID: PMC1573695 DOI: 10.1038/sj.bjp.0705078] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
1. Acetaminophen, an analgesic and antipyretic drug with weak antiinflammatory properties, has been suggested to act as a tissue-selective inhibitor of prostaglandin H synthases (PGHSs) (e.g. COX-1 and COX-2) through its reducing activity, that is influenced by the different cellular levels of peroxides. 2. We have studied the effects of acetaminophen on inducible and constitutive prostanoid biosynthesis in monocytes and platelets in vitro. To discriminate between the inhibitory effect of the drug on PGHS-isozymes vs PGE-synthases (PGESs), parallel measurements of PGE(2) and thromboxane (TX) B(2) were carried out. Since antioxidant enzymes and cofactors, present in plasma, may affect acetaminophen-dependent inhibition of prostanoids, comparative experiments in whole blood vs isolated monocytes were performed. 3. Acetaminophen inhibited LPS-induced whole blood PGE(2) and TXB(2) production, in a concentration-dependent fashion [IC(50) microM (95% confidence intervals): 44 (27-70) and 94 (79-112), respectively]. Therapeutic plasma concentrations (100 and 300 microM) of the drug more profoundly reduced PGE(2) than TXB(2) (71 +/- 3 vs 54 +/- 4 and 95 +/- 0.8 vs 78 +/- 2%, respectively, mean +/- s.e.mean, n = 6, P < 0.01). 4. Differently, in isolated monocytes stimulated with LPS, both PGE(2) and TXB(2) production was maximally reduced by only 60%. 5 At 100 and 300 microM, the drug caused a similar and incomplete inhibition of platelet PGE(2) and TXB(2) production during whole blood clotting (45 +/- 4 vs 54 +/- 4 and 75 +/- 2 vs 75 +/- 1%, respectively, mean +/- s.e.mean, n = 4). 6 In conclusion, therapeutic concentrations of acetaminophen caused an incomplete inhibition of platelet COX-1 and monocyte COX-2 but in the presence of plasma, the drug almost completely suppressed inducible PGE(2) biosynthesis through its inhibitory effects on both COX-2 and inducible PGES.
Collapse
Affiliation(s)
- Maria G Sciulli
- Centre of Excellence on Aging, ‘G. D'Annunzio' University, School of Medicine, Chieti, Italy
| | - Francesca Seta
- Centre of Excellence on Aging, ‘G. D'Annunzio' University, School of Medicine, Chieti, Italy
| | - Stefania Tacconelli
- Centre of Excellence on Aging, ‘G. D'Annunzio' University, School of Medicine, Chieti, Italy
| | - Marta L Capone
- Centre of Excellence on Aging, ‘G. D'Annunzio' University, School of Medicine, Chieti, Italy
| | - Emanuela Ricciotti
- Centre of Excellence on Aging, ‘G. D'Annunzio' University, School of Medicine, Chieti, Italy
| | - Giuseppa Pistritto
- Centre of Excellence on Aging, ‘G. D'Annunzio' University, School of Medicine, Chieti, Italy
| | - Paola Patrignani
- Centre of Excellence on Aging, ‘G. D'Annunzio' University, School of Medicine, Chieti, Italy
- Author for correspondence:
| |
Collapse
|
75
|
Wilmer WA, Dixon CL, Hebert C, Lu L, Rovin BH. PPAR-alpha ligands inhibit H2O2-mediated activation of transforming growth factor-beta1 in human mesangial cells. Antioxid Redox Signal 2002; 4:877-84. [PMID: 12573136 DOI: 10.1089/152308602762197416] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Transforming growth factor-beta1 (TGF-beta1) mediates the development of glomerulosclerosis by stimulating mesangial cell production of extracellular matrix (ECM) proteins. TGF-beta1 and several ECM genes are regulated by promoter O-tetradecanoylphorbol 13-acetate-responsive elements (TREs) that are transactivated by the activator protein-1 (AP-1) transcription factor complex. AP-1-TRE interactions are regulated by redox changes. Recently, peroxisome proliferator-activated receptors (PPARs) were shown to negatively regulate several transcription factor families. In these studies, we postulated that PPAR-alpha could antagonize TGF-beta1 expression by cultured human mesangial cells (HMC). A TGF-beta1 luciferase expression plasmid was transduced into HMC via recombinant deficient adenoviral vectors. The TGF-beta1 promoter activity increased twofold (209%) following 18-h treatments with H(2)O(2) (1,000 micro M). Using RT-PCR, we demonstrated that HMC possess PPAR-alpha RNA, and PPAR-alpha protein was identified by immunohistochemistry. Pretreatment of cells with the PPAR-alpha ligands WY14643 (100-500 micro M) or clofibrate (100-500 micro M) dose-dependently inhibited oxidant-mediated induction of TGF-beta1. This inhibition occurred without affecting the H(2)O(2)-mediated activation of the mitogen-activated protein kinase (MAPK) pathways extracellular regulated kinase, p38 MAPK, or Jun N-terminal kinase, which are responsible for the regulation of AP-1 phosphorylation. These studies are the first to identify PPAR-alpha expression by HMC. The results of these studies suggest that TGF-beta1 expression mediated by oxidant stress may be suppressible by PPAR-alpha activation.
Collapse
Affiliation(s)
- William A Wilmer
- Division of Nephrology, The Department of Internal Medicine, Dorothy M. Davis Heart-Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH 43210, USA.
| | | | | | | | | |
Collapse
|
76
|
Thuillier P, Brash AR, Kehrer JP, Stimmel JB, Leesnitzer LM, Yang P, Newman RA, Fischer SM. Inhibition of peroxisome proliferator-activated receptor (PPAR)-mediated keratinocyte differentiation by lipoxygenase inhibitors. Biochem J 2002; 366:901-10. [PMID: 12069687 PMCID: PMC1222830 DOI: 10.1042/bj20020377] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2002] [Revised: 04/24/2002] [Accepted: 06/17/2002] [Indexed: 12/20/2022]
Abstract
Lipoxygenase (LOX) metabolites from arachidonic acid and linoleic acid have been implicated in atherosclerosis, inflammation, keratinocyte differentiation and tumour progression. We previously showed that peroxisome proliferator-activated receptors (PPARs) play a role in keratinocyte differentiation and that the PPARalpha ligand 8S-hydroxyeicosatetraenoic acid is important in this process. We hypothesized that blocking LOX activity would block PPAR-mediated keratinocyte differentiation. Three LOX inhibitors, nordihydroguaiaretic acid, quercetin and morin, were studied for their effects on primary keratinocyte differentiation and PPAR activity. All three LOX inhibitors blocked calcium-induced expression of the differentiation marker keratin 1. In addition, activity of a PPAR-responsive element was inhibited in the presence of all three inhibitors, and this effect was mediated primarily through PPARalpha and PPARgamma. LOX inhibitors decreased the activity of a chimaeric PPAR-Gal4-ligand-binding domain reporter system and this effect was reversed by addition of PPAR ligands. Ligand-binding studies revealed that the LOX inhibitors bind directly to PPARs and demonstrate a novel mechanism for these inhibitors in altering PPAR-mediated gene expression.
Collapse
Affiliation(s)
- Philippe Thuillier
- Department of Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Smithville 78957, USA
| | | | | | | | | | | | | | | |
Collapse
|
77
|
Holness MJ, Smith ND, Bulmer K, Hopkins T, Gibbons GF, Sugden MC. Evaluation of the role of peroxisome-proliferator-activated receptor alpha in the regulation of cardiac pyruvate dehydrogenase kinase 4 protein expression in response to starvation, high-fat feeding and hyperthyroidism. Biochem J 2002; 364:687-94. [PMID: 12049632 PMCID: PMC1222617 DOI: 10.1042/bj20011841] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Inactivation of cardiac pyruvate dehydrogenase complex (PDC) after prolonged starvation and in response to hyperthyroidism is associated with enhanced protein expression of pyruvate dehydrogenase kinase (PDK) isoform 4. The present study examined the potential role of peroxisome-proliferator-activated receptor alpha (PPARalpha) in adaptive modification of cardiac PDK4 protein expression after starvation and in hyperthyroidism. PDK4 protein expression was analysed by immunoblotting in homogenates of hearts from fed or 48 h-starved rats, rats rendered hyperthyroid by subcutaneous injection of tri-iodothyronine and a subgroup of euthyroid rats maintained on a high-fat/low-carbohydrate diet, with or without treatment with the PPARalpha agonist WY14,643. In addition, PDK4 protein expression was analysed in hearts from fed, 24 h-starved or 6 h-refed wild-type or PPARalpha-null mice. PPARalpha activation by WY14,643 in vivo over the timescale of the response to starvation failed to up-regulate cardiac PDK4 protein expression in rats maintained on standard diet (WY14,643, 1.1-fold increase; starvation, 1.8-fold increase) or influence the cardiac PDK4 response to starvation. By contrast, PPARalpha activation by WY14,643 in vivo significantly enhanced cardiac PDK4 protein expression in rats maintained on a high-fat diet, which itself increased cardiac PDK4 protein expression. PPARalpha deficiency did not abolish up-regulation of cardiac PDK4 protein expression in response to starvation (2.9-fold increases in both wild-type and PPARalpha-null mice). Starvation and hyperthyroidism exerted additive effects on cardiac PDK4 protein expression, but PPARalpha activation by WY14,643 did not influence the response of cardiac PDK4 protein expression to hyperthyroidism in either the fed or starved state. Our data support the hypothesis that cardiac PDK4 protein expression is regulated, at least in part, by a fatty acid-dependent, PPARalpha-independent mechanism and strongly implicate a fall in insulin in either initiating or facilitating the response of cardiac PDK4 protein expression to starvation.
Collapse
Affiliation(s)
- Mark J Holness
- Department of Diabetes and Metabolic Medicine, St Bartholomew's and the Royal London School of Medicine and Dentistry, Medical Sciences Building, Queen Mary, University of London, London E1 4NS, UK
| | | | | | | | | | | |
Collapse
|
78
|
Abstract
Thia substituted fatty acids are saturated fatty acids which are modified by insertion of a sulfur atom at specific positions in the carbon backbone. During the last few years pleiotropic effects of the 3-thia fatty acid tetradecylthioacetic acid have been revealed. The biological responses to tetradecylthioacetic acid include mitochondrial proliferation, increased catabolism of fatty acids, antiadiposity, improvement in insulin sensitivity, antioxidant properties, reduced proliferation and induction of apoptosis in rapidly proliferating cells, cell differentiation and antiinflammatory action. These biological responses indicate that tetradecylthioacetic acid changes the plasma profile from atherogenic to cardioprotective. As a pan-peroxisome proliferator-activated receptor ligand, tetradecylthioacetic acid regulates the adipose tissue mass and the expression of lipid metabolizing enzymes, particularly those involved in catabolic pathways. In contrast, circumstantial evidences suggest that peroxisome proliferator-activated receptor-independent metabolic pathways may be of importance for the antioxidant, antiproliferative and antiinflammatory action of tetradecylthioacetic acid.
Collapse
Affiliation(s)
- Rolf K Berge
- Department of Clinical Biochemistry, Haukeland Hospital, University of Bergen, Norway.
| | | | | | | | | | | |
Collapse
|
79
|
Kocarek TA, Shenoy SD, Mercer-Haines NA, Runge-Morris M. Use of dominant negative nuclear receptors to study xenobiotic-inducible gene expression in primary cultured hepatocytes. J Pharmacol Toxicol Methods 2002; 47:177-87. [PMID: 12628309 DOI: 10.1016/s1056-8719(03)00002-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
INTRODUCTION To determine the feasibility of using dominant negative nuclear receptors to dissect the regulation of inducible gene expression in primary cultured hepatocytes, a series of dominant negative nuclear receptor expression plasmids were designed with truncated AF-2 subdomains. METHODS Plasmids expressing dominant negative or wild-type constitutive androstane receptor (CAR), pregnane X receptor (PXR), farnesoid X receptor (FXR), liver X receptor (LXR), or peroxisome proliferator-activated receptor alpha (PPARalpha) were transiently cotransfected into primary cultured rat hepatocytes, together with an appropriate reporter plasmid. RESULTS Treatment with prototypic inducers, 10(-4) M phenobarbital (CAR activator), 10(-5) M pregnenolone 16alpha-carbonitrile (PXR activator), 3x10(-5) M chenodeoxycholate (FXR activator), or 10(-4) M ciprofibrate (PPARalpha activator), significantly activated expression from the corresponding reporter plasmid. Treatment with 22(R)-hydroxycholesterol (LXR activator) only weakly activated the LXR-responsive reporter, while pregnenolone 16alpha-carbonitrile treatment significantly activated this reporter. Cotransfection with wild-type LXRalpha strongly enhanced 22(R)-hydroxycholesterol-inducible expression from the LXR-responsive reporter. Cotransfection of hepatocyte cultures with each of the dominant negative nuclear receptor plasmids significantly inhibited inducible expression of the corresponding reporter while, with one exception (LXRalpha), cotransfection with the wild-type receptor moderately enhanced or had little effect on reporter expression. When each dominant negative nuclear receptor was cross-examined against all inducer-reporter pairs, effects on multiple inducer-reporter pairs were frequently observed. However, in general, only cotransfection with the appropriate dominant negative inhibited inducible reporter expression to a greater extent than did cotransfection with the corresponding wild-type receptor. DISCUSSION We suggest that the application of dominant negative nuclear receptors has utility in transient transfection studies aimed at discerning the regulatory role of individual nuclear receptor transcription factors in inducible hepatic gene expression, provided that appropriate controls are employed.
Collapse
Affiliation(s)
- Thomas A Kocarek
- Institute of Environmental Health Sciences, Wayne State University, Room 4000, 2727 Second Avenue, Detroit, MI 48201, USA.
| | | | | | | |
Collapse
|
80
|
Abstract
It is well established that fatty acid metabolites of cyclooxygenase, lipoxygenase (LOX), and cytochrome P450 are implicated in essential aspects of cellular signaling including the induction of programmed cell death. Here we review the roles of enzymatic and non-enzymatic products of polyunsaturated fatty acids in controlling cell growth and apoptosis. Also, the spontaneous oxidation of polyunsaturated fatty acids yields reactive aldehydes and other products of lipid peroxidation that are potentially toxic to cells and may also signal apoptosis. Significant conflicting data in terms of the role of LOX enzymes are highlighted, prompting a re-evaluation of the relationship between LOX and prostate cancer cell survival. We include new data showing that LNCaP, PC3, and Du145 cells express much lower levels of 5-LOX mRNA and protein compared with normal prostate epithelial cells (NHP2) and primary prostate carcinoma cells (TP1). Although the 5-LOX activating protein inhibitor MK886 killed these cells, another 5-LOX inhibitor AA861 hardly showed any effect. These observations suggest that 5-LOX is unlikely to be a prostate cancer cell survival factor, implying that the mechanisms by which LOX inhibitors induce apoptosis are more complex than expected. This review also suggests several mechanisms involving peroxisome proliferator activated receptor activation, BCL proteins, thiol regulation, and mitochondrial and kinase signaling by which cell death may be produced in response to changes in non-esterified and non-protein bound fatty acid levels. Overall, this review provides a context within which the effects of fatty acids and fatty acid oxidation products on signal transduction pathways, particularly those involved in apoptosis, can be considered in terms of their overall importance relative to the much better studied protein or peptide signaling factors.
Collapse
Affiliation(s)
- Dean G Tang
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas, Austin 78712, USA
| | | | | | | |
Collapse
|
81
|
Scholz-Pedretti K, Gans A, Beck KF, Pfeilschifter J, Kaszkin M. Potentiation of TNF-alpha-stimulated group IIA phospholipase A(2) expression by peroxisome proliferator-activated receptor alpha activators in rat mesangial cells. J Am Soc Nephrol 2002; 13:611-620. [PMID: 11856764 DOI: 10.1681/asn.v133611] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Natural activators of peroxisome proliferator-activated receptors (PPAR) are lipid metabolites, including those produced by phospholipases A(2) (PLA(2)). In glomerular mesangial cells, the secreted group IIA PLA(2) (sPLA(2)-IIA), which is thought to be a crucial factor in pathologic processes in the kidney, may provide free fatty acids and eicosanoids directly or indirectly, by activating a cytosolic PLA(2). The scope of this study was to investigate whether synthetic PPAR(alpha) activators have an effect on sPLA(2)-IIA mRNA expression in rat mesangial cells, thus constituting a feedback modulation of sPLA(2)-IIA transcription. In the presence of tumor necrosis factor-alpha (TNF-alpha), the PPAR(alpha) agonists WY14643 and LY171883 as well as the lipid-lowering compound clofibrate potentiated expression, secretion, and activity of group IIA sPLA(2) in mesangial cells. MK886, known as a noncompetitive inhibitor of PPAR(alpha), completely abolished the potentiation of sPLA(2)-IIA secretion and activity by WY14643, thus indicating that the effect of WY14643 is specifically mediated by PPAR(alpha). When cells were transfected with different constructs of the rat sPLA(2)-IIA promoter fused to a luciferase reporter gene, a stimulation with TNF-alpha in the presence of the PPAR(alpha) activators caused an enhanced promoter activity compared with that induced by TNF-alpha alone. Site-directed mutagenesis of a putative PPRE site in the sPLA(2)-IIA promoter abolished the potentiating effect of PPAR(alpha) agonists, thus strongly indicating its contribution to the enhanced promoter activity. In summary, this study shows that the rat sPLA(2)-IIA promoter is sensitive to PPAR(alpha) agonists, which act synergistically with cytokines, resulting in an enhanced expression of sPLA(2)-IIA in rat mesangial cells.
Collapse
Affiliation(s)
- Kirsten Scholz-Pedretti
- Pharmazentrum Frankfurt, Klinikum der Johann Wolfgang Goethe-Universität, Frankfurt am Main, Germany
| | - Annette Gans
- Pharmazentrum Frankfurt, Klinikum der Johann Wolfgang Goethe-Universität, Frankfurt am Main, Germany
| | - Karl-Friedrich Beck
- Pharmazentrum Frankfurt, Klinikum der Johann Wolfgang Goethe-Universität, Frankfurt am Main, Germany
| | - Josef Pfeilschifter
- Pharmazentrum Frankfurt, Klinikum der Johann Wolfgang Goethe-Universität, Frankfurt am Main, Germany
| | - Marietta Kaszkin
- Pharmazentrum Frankfurt, Klinikum der Johann Wolfgang Goethe-Universität, Frankfurt am Main, Germany
| |
Collapse
|
82
|
Adamson DJA, Frew D, Tatoud R, Wolf CR, Palmer CNA. Diclofenac antagonizes peroxisome proliferator-activated receptor-gamma signaling. Mol Pharmacol 2002; 61:7-12. [PMID: 11752200 DOI: 10.1124/mol.61.1.7] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Although nonsteroidal anti-inflammatory drugs (NSAIDs) are used as cancer chemopreventative agents, their mechanism is unclear because NSAIDs have cyclooxygenase-independent actions. We investigated an alternative target for NSAIDs, peroxisome proliferator-activated receptor-gamma (PPARgamma), activation of which decreases cancer cell proliferation. NSAIDs have been shown to activate this receptor, but only at high concentrations. Here, we have examined binding of diclofenac to PPARgamma using a cis-parinaric acid displacement assay and studied the effect of diclofenac effect on PPARgamma trans-activation in a COS-1 cell reporter assay. Unexpectedly, diclofenac bound PPARgamma at therapeutic concentrations (K(i) = 700 nM) but induced only 2-fold activation of PPARgamma at a concentration of 25 microM and antagonized PPARgamma trans-activation by rosiglitazone. This antagonism was overcome with increasing rosiglitazone concentrations, indicating that diclofenac is a partial agonist. No effect of diclofenac was seen without exogenous receptor, confirming that it was working through a PPARgamma-specific mechanism. This is the first description of an NSAID that can antagonize PPARgamma. In addition, this is the first time that an NSAID has been shown to bind this receptor at clinically meaningful concentrations. The physiological relevance of these findings was tested using adipocyte differentiation and cancer cell proliferation assays. Diclofenac decreased PPARgamma-mediated adipose cell differentiation by 60% and inhibited the action of rosiglitazone on the prostate cancer cell line, DU-145, allowing a 3-fold increase in proliferation. This work shows that standard doses of diclofenac may have pharmacodynamic interactions with rosiglitazone and this has therapeutic implications, both in the management of type 2 diabetes and during cancer treatment.
Collapse
Affiliation(s)
- Douglas J A Adamson
- Imperial Cancer Research Fund Molecular Pharmacology Unit, Biomedical Research Centre, Ninewells Hospital, Dundee, Scotland, United Kingdom
| | | | | | | | | |
Collapse
|