201
|
Adapala N, Chan MM. Long-term use of an antiinflammatory, curcumin, suppressed type 1 immunity and exacerbated visceral leishmaniasis in a chronic experimental model. J Transl Med 2008; 88:1329-39. [PMID: 18794851 PMCID: PMC3056242 DOI: 10.1038/labinvest.2008.90] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Inflammation is considered the underlying cause of numerous disorders, and the practice of taking antiinflammatories as diet supplements has become increasingly prevalent. This study addresses the bioavailablity of a well-established dietary antiinflammatory, curcumin, and examines its effect on adaptive immunity. Visceral leishmaniasis is a major parasitic disease which protection relies on cell-mediated immunity and production of nitric oxide. We found that long-term, low-dose, oral consumption of curcumin activates peroxisome proliferator-activated receptor-gamma, deactivates type 1 response, inhibits inducible nitric oxide synthase, and interferes with adaptive immunity to exacerbate the pathogenesis of Leishmania donovani infection in vivo. These in vivo effects can be correlated to activities on infected residential macrophages in vitro. Therefore, when reactive radicals generated from inflammation play the dominant role in elimination of pathogens, excessive use of the antioxidative supplements may compromise microbial defense. Nonetheless, it should be noted with equal importance that our finding, conversely, also strengthens the prospect that curcumin may alleviate type 1 response disorders.
Collapse
|
202
|
Zurhove K, Nakajima C, Herz J, Bock HH, May P. Gamma-secretase limits the inflammatory response through the processing of LRP1. Sci Signal 2008; 1:ra15. [PMID: 19036715 DOI: 10.1126/scisignal.1164263] [Citation(s) in RCA: 106] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Inflammation is a potentially self-destructive process that needs tight control. We have identified a nuclear signaling mechanism through which the low-density lipoprotein receptor-related protein 1 (LRP1) limits transcription of lipopolysaccharide (LPS)-inducible genes. LPS increases the proteolytic processing of the ectodomain of LRP1, which results in the gamma-secretase-dependent release of the LRP1 intracellular domain (ICD) from the plasma membrane and its translocation to the nucleus, where it binds to and represses the interferon-gamma promoter. Basal transcription of LPS target genes and LPS-induced secretion of proinflammatory cytokines are increased in the absence of LRP1. The interaction between LRP1-ICD and interferon regulatory factor 3 (IRF-3) promotes the nuclear export and proteasomal degradation of IRF-3. Feedback inhibition of the inflammatory response through intramembranous processing of LRP1 thus defines a physiological role for gamma-secretase.
Collapse
Affiliation(s)
- Kai Zurhove
- Department of Medicine II, University Hospital, University of Freiburg, 79106 Freiburg, Germany
| | | | | | | | | |
Collapse
|
203
|
Imig JD, Zhao X, Dey A, Shaw M. CYP450, COX-2 and Obesity Related Renal Damage. Toxicol Mech Methods 2008; 15:125-36. [DOI: 10.1080/15376520590918856] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
204
|
Varga T, Nagy L. Nuclear receptors, transcription factors linking lipid metabolism and immunity: the case of peroxisome proliferator-activated receptor gamma. Eur J Clin Invest 2008; 38:695-707. [PMID: 18837794 DOI: 10.1111/j.1365-2362.2008.02022.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Exposure to lipids has a major effect on mammalian cells. Naturally, it has a profound impact on their metabolism, but it can also significantly alter their cellular and molecular phenotypes and responses. This latter is via specific signalling pathways leading to alterations in the expression of genes and gene networks. Multicellular organisms utilize a specialized group of proteins to detect and transduce lipid signals to the level of the expression of the genome. These proteins, termed nuclear hormone receptors, are lipid-activated transcription factors regulating gene expression upon binding of small fatty ligands. In this review, we discuss the role and contribution of peroxisome proliferator-activated receptor gamma (PPAR gamma) to macrophage and dendritic cell biology and also to gut epithelial cell function. We discuss how using different experimental systems and approaches the pathways activating the receptor and its target genes can be identified and complex biological processes unravelled. It appears that PPAR gamma is part of the macrophage's response to pathogenic lipoproteins and it coordinately regulates lipid uptake and efflux. Intriguingly, in another cell type of the immune system, dendritic cells, the receptor has overlapping, but distinct functions. In these cells, activation of PPAR gamma leads to altered immune phenotype characterized by increased phagocytic capacity, antigen processing and lipid antigen presenting capacity. This nuclear hormone receptor links lipid metabolism and immune cell function and these links provide unique insights into the regulatory logic of normal physiological responses and certain pathologies, such as atherosclerosis, chronic inflammatory diseases and autoimmunity.
Collapse
Affiliation(s)
- T Varga
- Department of Biochemistry and Molecular Biology, University of Debrecen, Debrecen, Hungary
| | | |
Collapse
|
205
|
Hong C, Tontonoz P. Coordination of inflammation and metabolism by PPAR and LXR nuclear receptors. Curr Opin Genet Dev 2008; 18:461-7. [PMID: 18782619 PMCID: PMC2641014 DOI: 10.1016/j.gde.2008.07.016] [Citation(s) in RCA: 184] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2008] [Accepted: 07/31/2008] [Indexed: 12/13/2022]
Abstract
Biological systems are integrated networks constantly responding to internal and external stimulators. Understanding the intrinsic response to an imbalanced system provides the opportunity to develop therapeutic approaches to reinstate the natural balanced state. Increasing evidence suggests that members of the nuclear receptor superfamily integrate both inflammatory and metabolic signals to maintain homeostasis in immune cells such as macrophages and lymphocytes. PPAR and LXR are nuclear receptors activated by fatty acid and cholesterol derivatives respectively that control the expression of an array of genes involved in lipid metabolism and inflammation. Recent studies have uncovered distinct mechanisms for transcriptional regulation of metabolic and inflammatory target genes by PPAR and LXR and have expanded the biology of these receptors to include roles in alternative macrophage activation and adaptive immunity.
Collapse
Affiliation(s)
- Cynthia Hong
- Howard Hughes Medical Institute, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, 675 Charles E. Young Drive, Los Angeles, CA 90049, USA
| | | |
Collapse
|
206
|
Hong C, Tontonoz P. Coordination of inflammation and metabolism by PPAR and LXR nuclear receptors. Curr Opin Genet Dev 2008. [PMID: 18782619 DOI: 10.1016/j.gde.2008.07] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Biological systems are integrated networks constantly responding to internal and external stimulators. Understanding the intrinsic response to an imbalanced system provides the opportunity to develop therapeutic approaches to reinstate the natural balanced state. Increasing evidence suggests that members of the nuclear receptor superfamily integrate both inflammatory and metabolic signals to maintain homeostasis in immune cells such as macrophages and lymphocytes. PPAR and LXR are nuclear receptors activated by fatty acid and cholesterol derivatives respectively that control the expression of an array of genes involved in lipid metabolism and inflammation. Recent studies have uncovered distinct mechanisms for transcriptional regulation of metabolic and inflammatory target genes by PPAR and LXR and have expanded the biology of these receptors to include roles in alternative macrophage activation and adaptive immunity.
Collapse
Affiliation(s)
- Cynthia Hong
- Howard Hughes Medical Institute, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, 675 Charles E. Young Drive, Los Angeles, CA 90049, USA
| | | |
Collapse
|
207
|
Abstract
The nuclear receptor PPARgamma is a ligand-activated transcription factor that plays an important role in the control of gene expression linked to a variety of physiological processes. PPARgamma was initially characterized as the master regulator for the development of adipose cells. Ligands for PPARgamma include naturally occurring fatty acids and the thiazolidinedione (TZD) class of antidiabetic drugs. Activation of PPARgamma improves insulin sensitivity in rodents and humans through a combination of metabolic actions, including partitioning of lipid stores and the regulation of metabolic and inflammatory mediators termed adipokines. PPARgamma signaling has also been implicated in the control of cell proliferation, atherosclerosis, macrophage function, and immunity. Here, we review recent advances in our understanding of the diverse biological actions of PPARgamma with an eye toward the expanding therapeutic potential of PPARgamma agonist drugs.
Collapse
Affiliation(s)
- Peter Tontonoz
- Howard Hughes Medical Institute and Department of Pathology and Laboratory Medicine, University of California-Los Angeles, CA 90095, USA.
| | | |
Collapse
|
208
|
Szanto A, Nagy L. The many faces of PPARgamma: anti-inflammatory by any means? Immunobiology 2008; 213:789-803. [PMID: 18926294 DOI: 10.1016/j.imbio.2008.07.015] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2008] [Accepted: 07/29/2008] [Indexed: 01/08/2023]
Abstract
The peroxisome proliferator-activated receptor gamma (PPARgamma) is a member of the nuclear receptor superfamily, a group of transcription factors that regulate expression of their target genes upon ligand binding. As endogenous ligands, oxidized fatty acids and prostanoids can bind to and activate the receptor. Natural and synthetic PPARgamma activators have been studied extensively in many inflammatory settings and in most instances they have been shown to be anti-inflammatory. In this review we give an overview of the different molecular mechanisms how PPARgamma and its agonists exert their anti-inflammatory effects both at the cellular level and the level of the organism. The action of PPARgamma in acute and chronic inflammatory diseases and disease models will be presented.
Collapse
Affiliation(s)
- Attila Szanto
- Department of Biochemistry and Molecular Biology, University of Debrecen, Medical and Health Science Center, Research Center for Molecular Medicine, Life Science Building, Egyetem ter 1, H-4032 Debrecen, Hungary.
| | | |
Collapse
|
209
|
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 DOI: 10.1042/cs20080022] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [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
|
210
|
Rozo AV, Vijayvargia R, Weiss HR, Ruan H. Silencing Jnk1 and Jnk2 accelerates basal lipolysis and promotes fatty acid re-esterification in mouse adipocytes. Diabetologia 2008; 51:1493-504. [PMID: 18528680 DOI: 10.1007/s00125-008-1036-6] [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: 12/05/2007] [Accepted: 04/05/2008] [Indexed: 01/04/2023]
Abstract
AIMS/HYPOTHESIS Elevated plasma levels of NEFA impair insulin action. Given the positive linear correlation between NEFA released by adipocytes and plasma NEFA levels, identification of mechanisms controlling adipocyte lipolysis and NEFA release could provide a guide to new therapies for insulin resistance and type 2 diabetes. METHODS Short hairpin RNA-mediated gene ablation was used to determine the functions of c-Jun N-terminal kinase (JNK)1 and JNK2 in adipocytes. RESULTS Combined JNK1/JNK2 deficiency drastically increased basal glycerol release, whereas individual JNK1- or JNK2-deficiency had no effect, indicating that JNK1/JNK2-deficiency enhances basal lipolysis, whereas the alternate subtype compensates for a single JNK subtype deficiency in the regulation of basal lipolysis. The profoundly increased glycerol release associated with JNK1/JNK2-deficiency was not accompanied by a concomitant increase in NEFA release over time. In addition, JNK1-deficiency, but not JNK2-deficiency, drastically decreased NEFA release as compared with that in JNK-intact cells, a result of increased NEFA re-esterification. In microarray, quantitative RT-PCR and western blotting, JNK1-, JNK2- and JNK1/JNK2-deficiencies selectively upregulated many genes involved in NEFA management, without affecting the expression of genes involved in insulin signalling. Assays using reporter genes driven by peroxisome proliferator-activated receptor gamma (PPAR-gamma)-responsive promoters indicate distinct roles for JNK1 and JNK2 in regulating the transcriptional effects of PPAR-gamma. CONCLUSIONS/INTERPRETATION While JNK1 and JNK2 have shared roles in the regulation of basal lipolysis, JNK1 has a more profound role in supporting baseline NEFA release. Inhibition of JNK1 activity in adipocytes has potential therapeutic uses for management of elevated circulating NEFA levels at the onset of insulin resistance.
Collapse
Affiliation(s)
- A V Rozo
- Department of Physiology and Biophysics, University of Medicine and Dentistry of New Jersey-Robert Wood Johnson Medical School, 683 Hoes Lane, Piscataway, NJ 08854, USA
| | | | | | | |
Collapse
|
211
|
A central role for free heme in the pathogenesis of severe malaria: the missing link? J Mol Med (Berl) 2008; 86:1097-111. [PMID: 18641963 DOI: 10.1007/s00109-008-0368-5] [Citation(s) in RCA: 149] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2008] [Revised: 04/10/2008] [Accepted: 05/09/2008] [Indexed: 12/14/2022]
Abstract
Malaria, the disease caused by Plasmodium infection, is endemic to poverty in so-called underdeveloped countries. Plasmodium falciparum, the main infectious Plasmodium species in sub-Saharan countries, can trigger the development of severe malaria, including cerebral malaria, a neurological syndrome that claims the lives of more than one million children (<5 years old) per year. Attempts to eradicate Plasmodium infection, and in particular its lethal outcomes, have so far been unsuccessful. Using well-established rodent models of malaria infection, we found that survival of a Plasmodium-infected host is strictly dependent on the host's ability to up-regulate the expression of heme oxygenase-1 (HO-1 encoded by the gene Hmox1). HO-1 is a stress-responsive enzyme that catabolizes free heme into biliverdin, via a reaction that releases Fe and generates the gas carbon monoxide (CO). Generation of CO through heme catabolism by HO-1 prevents the onset of cerebral malaria. The protective effect of CO is mediated via its binding to cell-free hemoglobin (Hb) released from infected red blood cells during the blood stage of Plasmodium infection. Binding of CO to cell-free Hb prevents heme release and thus generation of free heme, which we found to play a central role in the pathogenesis of cerebral malaria. We will address hereby how defense mechanisms that prevent the deleterious effects of free heme, including the expression of HO-1, impact on the pathologic outcome of Plasmodium infection and how these may be used therapeutically to suppress its lethal outcomes.
Collapse
|
212
|
Shan W, Palkar PS, Murray IA, McDevitt EI, Kennett MJ, Kang BH, Isom HC, Perdew GH, Gonzalez FJ, Peters JM. Ligand activation of peroxisome proliferator-activated receptor beta/delta (PPARbeta/delta) attenuates carbon tetrachloride hepatotoxicity by downregulating proinflammatory gene expression. Toxicol Sci 2008; 105:418-28. [PMID: 18622026 DOI: 10.1093/toxsci/kfn142] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Peroxisome proliferator-activated receptor (PPAR) beta/delta-null mice exhibit exacerbated hepatotoxicity in response to administration of carbon tetrachloride (CCl(4)). To determine whether ligand activation of the receptor protects against chemical toxicity in the liver, wild-type and PPARbeta/delta-null mice were administered CCl(4) with or without coadministration of the highly specific PPARbeta/delta ligand GW0742. Biomarkers of liver toxicity, including serum alanine aminotransferase (ALT) and hepatic tumor necrosis factor (TNF) alpha mRNA, were significantly higher in CCl(4)-treated PPARbeta/delta-null mice compared to wild-type mice. Hepatic expression of TNF-like weak inducer of apoptosis receptor (TWEAKr) and S100 calcium-binding protein A6 (S100A6/calcyclin), genes involved in nuclear factor kappa B signaling, was higher in the CCl(4)-treated PPARbeta/delta-null mice compared to wild-type mice. GW0742 treatment resulted in reduced serum ALT concentration and lower expression of CCl(4)-induced TNF-alpha, S100A6, monocyte chemoattractant protein-1 (MCP1), and TWEAKr in wild-type mice, and these effects were not observed in PPARbeta/delta-null mice. Expression of TNF-alpha was higher in PPARbeta/delta-null primary hepatocytes in response to interleukin-1beta treatment compared to wild-type hepatocytes, but GW0742 did not significantly modulate TNF-alpha expression in hepatocytes from either genotype. While PPARbeta/delta-null hepatic stellate exhibited higher rates of proliferation compared to wild-type cells, GW0742 did not affect alpha-smooth muscle actin expression in these cells. Combined, these findings demonstrate that ligand activation of PPARbeta/delta protects against chemically induced hepatotoxicity by downregulating expression of proinflammatory genes. Hepatocytes and hepatic stellate cells do not appear to directly mediate the inhibitory effects of ligand activation of PPARbeta/delta in liver, suggesting the involvement of paracrine and autocrine events mediated by hepatic cells.
Collapse
Affiliation(s)
- Weiwei Shan
- Department of Veterinary and Biomedical Sciences and The Center for Molecular Toxicology and Carcinogenesis, The Huck Institute of Life Sciences, The Pennsylvania State University, University Park, Pennsylvania 16802, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
213
|
Abstract
Macrophages, which belong to the immune system, are increasingly being recognized for their contribution to metabolic regulation. In two studies by Kang et al. (2008) and Odegaard et al. (2008) in this issue of Cell Metabolism, we learn that alternative activation (M2a) of resident macrophages in liver and adipose tissue depends highly on PPARdelta/beta activity, leading to improved fatty acid metabolism and insulin sensitivity.
Collapse
Affiliation(s)
- Béatrice Desvergne
- Center of Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, CH-1015 Lausanne, Switzerland.
| |
Collapse
|
214
|
Rigamonti E, Chinetti-Gbaguidi G, Staels B. Regulation of Macrophage Functions by PPAR-α, PPAR-γ, and LXRs in Mice and Men. Arterioscler Thromb Vasc Biol 2008; 28:1050-9. [DOI: 10.1161/atvbaha.107.158998] [Citation(s) in RCA: 207] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- Elena Rigamonti
- From the Institut Pasteur de Lille, Inserm, U545, and Université de Lille 2, Faculté de Pharmacie et de Médecine, Lille, France
| | - Giulia Chinetti-Gbaguidi
- From the Institut Pasteur de Lille, Inserm, U545, and Université de Lille 2, Faculté de Pharmacie et de Médecine, Lille, France
| | - Bart Staels
- From the Institut Pasteur de Lille, Inserm, U545, and Université de Lille 2, Faculté de Pharmacie et de Médecine, Lille, France
| |
Collapse
|
215
|
Posokhova EN, Khoshchenko OM, Chasovskikh MI, Pivovarova EN, Dushkin MI. Lipid synthesis in macrophages during inflammation in vivo: effect of agonists of peroxisome proliferator activated receptors alpha and gamma and of retinoid X receptors. BIOCHEMISTRY (MOSCOW) 2008; 73:296-304. [PMID: 18393765 DOI: 10.1134/s0006297908030097] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The effects of peroxisome proliferator activated receptors alpha and gamma (PPAR-alpha and PPAR-gamma) and retinoid X receptor (RXR) agonists upon synthesis and accumulation of lipids in murine C57Bl macrophages during inflammation induced by injection of zymosan and Escherichia coli lipopolysaccharide (LPS) have been studied. It is significant that intraperitoneal injection of zymosan (50 mg/kg) or LPS (0.1 mg/kg) in mice led to a dramatic increase of [14C]oleate incorporation into cholesteryl esters and triglycerides and [14C]acetate incorporation into cholesterol and fatty acids in peritoneal macrophages. Lipid synthesis reached its maximum rate 18-24 h after injection and was decreased 5-7 days later to control level after LPS injection or was still heightened after zymosan injection. In macrophages obtained in acute phase of inflammation (24 h), degradation of 125I-labeled native low density lipoprotein (NLDL) was 4-fold increased and degradation of 125I-labeled acetylated LDL (AcLDL) was 2-3-fold decreased. Addition of NLDL (50 microg/ml) or AcLDL (25 microg/ml) into the incubation medium of activated macrophages induced 9-14- and 1.25-fold increase of cholesteryl ester synthesis, respectively, compared with control. Addition of NLDL and AcLDL into the incubation medium completely inhibited cholesterol synthesis in control macrophages but had only slightly effect on cholesterol synthesis in activated macrophages. Injection of RXR, PPAR-alpha, or PPAR-gamma agonists--9-cis-retinoic acid (5 mg/kg), bezafibrate (10 mg/kg), or rosiglitazone (10 mg/kg), respectively--30 min before zymosan or LPS injection led to significant decrease of lipid synthesis. Ten hour preincubation of activated in vivo macrophages with the abovementioned agonists (5 microM) decreased cholesteryl ester synthesis induced by NLDL and AcLDL addition into the cell cultivation medium. The data suggest that RXR, PPAR-alpha, or PPAR-gamma agonists inhibited lipid synthesis and induction of cholesteryl ester synthesis in inflammatory macrophages caused by capture of native or modified LDL.
Collapse
Affiliation(s)
- E N Posokhova
- Institute of Internal Medicine, Siberian Branch of the Russian Academy of Medical Sciences, Novosibirsk, Russia
| | | | | | | | | |
Collapse
|
216
|
Lee JS, Kim KI, Baek SH. Nuclear receptors and coregulators in inflammation and cancer. Cancer Lett 2008; 267:189-96. [PMID: 18433989 DOI: 10.1016/j.canlet.2008.03.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2008] [Revised: 02/27/2008] [Accepted: 03/11/2008] [Indexed: 01/29/2023]
Abstract
Functional role of nuclear receptors and numerous coregulators have been studied in terms of regulating transcriptional control of genes that play critical roles in various pathways. There is growing evidence that nuclear receptors and their coregulators control inflammatory programs of gene expression and progression of hormone-dependent cancer. This review provides a general overview of the interrelationship between nuclear receptor signalling, inflammation and cancer. These insights provide inflammatory genes as attractive targets for the development of cancer therapeutics.
Collapse
Affiliation(s)
- Jason S Lee
- Department of Biological Sciences, Research Center for Functional Cellulomics, Seoul National University, Seoul 151-742, South Korea
| | | | | |
Collapse
|
217
|
Necela BM, Su W, Thompson EA. Toll-like receptor 4 mediates cross-talk between peroxisome proliferator-activated receptor gamma and nuclear factor-kappaB in macrophages. Immunology 2008; 125:344-58. [PMID: 18422969 DOI: 10.1111/j.1365-2567.2008.02849.x] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
The peroxisome proliferator-activated receptor gamma (PPARgamma) is expressed in macrophages and plays an important role in suppressing the inflammatory response. Lipopolysaccharides (LPS), which activate Toll-like receptor 4 (TLR4), reduced PPARgamma expression and function in peritoneal macrophages and macrophage cell lines. Moreover, pretreatment with the synthetic PPARgamma ligand, rosiglitazone did not prevent LPS-mediated downregulation of PPARgamma. Inhibition of PPARgamma expression was not blocked by cycloheximide, indicating that de novo protein synthesis is not required for LPS-mediated suppression of PPARgamma. Destabilization of PPARgamma messenger RNA (mRNA) was not observed in LPS-stimulated macrophages, suggesting that LPS regulates the synthesis of PPARgamma mRNA. LPS had no effect on PPARgamma expression in macrophages from TLR4 knockout mice, whereas LPS inhibited PPARgamma expression in cells that had been reconstituted to express functional TLR4. Targeting the TLR4 pathway with inhibitors of MEK1/2, p38, JNK and AP-1 had no effect on PPARgamma downregulation by LPS. However, inhibitors that target NEMO, IkappaB and NF-kappaB abolished LPS-mediated downregulation of PPARgamma in LPS-stimulated macrophages. Our data indicate that activation of TLR4 inhibits PPARgamma mRNA synthesis by an NF-kappaB-dependent mechanism. Low-density genomic profiling of macrophage-specific PPARgamma knockout cells indicated that PPARgamma suppresses inflammation under basal conditions, and that loss of PPARgamma expression is sufficient to induce a proinflammatory state. Our data reveal a regulatory feedback loop in which PPARgamma represses NF-kappaB-mediated inflammatory signalling in unstimulated macrophages; however, upon activation of TLR4, NF-kappaB drives down PPARgamma expression and thereby obviates any potential anti-inflammatory effects of PPARgamma in LPS-stimulated macrophages.
Collapse
Affiliation(s)
- Brian M Necela
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, FL 32224, USA.
| | | | | |
Collapse
|
218
|
Abstract
It is well known that the steroid hormone glucocorticoid and its nuclear receptor regulate the inflammatory process, a crucial component in the pathophysiological process related to human diseases that include atherosclerosis, obesity and type II diabetes, inflammatory bowel disease, Alzheimer's disease, multiple sclerosis, and liver tumors. Growing evidence demonstrates that orphan and adopted orphan nuclear receptors, such as peroxisome proliferator-activated receptors, liver x receptors, the farnesoid x receptor, NR4As, retinoid x receptors, and the pregnane x receptor, regulate the inflammatory and metabolic profiles in a ligand-dependent or -independent manner in human and animal models. This review summarizes the regulatory roles of these nuclear receptors in the inflammatory process and the underlying mechanisms.
Collapse
Affiliation(s)
- Kun Wang
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| | | |
Collapse
|
219
|
Subcellular localization of the Schlafen protein family. Biochem Biophys Res Commun 2008; 370:62-6. [PMID: 18355440 DOI: 10.1016/j.bbrc.2008.03.032] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2008] [Accepted: 03/05/2008] [Indexed: 11/23/2022]
Abstract
Although the first members of the Schlafen gene family were first described almost 10 years ago, the precise molecular/biochemical functions of the proteins they encode still remain largely unknown. Roles in cell growth, haematopoietic cell differentiation, and T cell development/maturation have, with some experimental support, been postulated, but none have been conclusively verified. Here, we have determined the subcellular localization of Schlafens 1, 2, 4, 5, 8, and 9, representing all three of the murine subgroups. We show that the proteins from subgroups I and II localize to the cytoplasm, while the longer forms in subgroup III localize exclusively to the nuclear compartment. We also demonstrate upregulation of Schlafen2 upon differentiation of haematopoietic cells and show this endogenous protein localizes to the cytoplasm. Thus, we propose the different subgroups of Schlafen proteins are likely to have functionally distinct roles, reflecting their differing localizations within the cell.
Collapse
|
220
|
Linard C, Grémy O, Benderitter M. Reduction of peroxisome proliferation-activated receptor gamma expression by gamma-irradiation as a mechanism contributing to inflammatory response in rat colon: modulation by the 5-aminosalicylic acid agonist. J Pharmacol Exp Ther 2008; 324:911-920. [PMID: 18077625 DOI: 10.1124/jpet.107.129122] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Radiation-induced intestinal injuries, including inflammation and immune response, remain a limiting factor in the effectiveness of pelvic radiotherapy and in the patient's quality of life during and after treatment. Peroxisome proliferation-activated receptor (PPAR) agonists are now emerging as therapeutic drugs for various inflammatory diseases that are characterized by impaired PPAR expression. The purpose of this study was to investigate the profile of PPAR expression in rat colonic mucosa 3 and 7 days after abdominal gamma-irradiation (10 Gy). We tested whether irradiation-induced acute inflammatory response could be modulated pharmacologically with the antiinflammatory properties of 5-aminosalicylic acid (5-ASA) (250 mg/kg/day), which is a PPAR activator. Irradiation drastically reduced mRNA and protein levels of PPARalpha and -gamma and of the heterodimer retinoid X receptor (RXR)alpha at 3 days postirradiation. 5-ASA treatment normalized both PPARgamma and RXRalpha expression at 3 days postirradiation and PPARalpha at 7 days. By promoting PPAR expression and its nuclear translocation, 5-ASA interfered with the nuclear factor (NF)-kappaB pathway, both by reducing irradiation-induced NF-kappaB p65 translocation/activation and increasing the expression of nuclear factor-kappaB inhibitor (IkappaB) mRNA and protein. Therefore, 5-ASA prevents irradiation-induced inflammatory processes as well as expression of tumor necrosis factor alpha, monocyte chemotactic protein-1, inducible nitric-oxide synthase, and macrophage infiltration. In addition, 5-ASA restores the interferon gamma/signal transducer and activator of transcription (STAT)-1 and STAT-3 concentrations that were impaired at 3 and 7 days postirradiation and are correlated with suppressor of cytokine signaling-3 repression. Collectively, these results indicate that PPAR agonists may be effective in the prevention of inflammatory processes and immune responses during and after pelvic radiotherapy.
Collapse
Affiliation(s)
- Christine Linard
- Institut de Radioprotection et de Sûreté Nucléaire, Direction de la Radioprotection de l'Homme, Laboratoire de Radiopathology, B.P. no. 17, F-92262 Fontenay-aux-Roses Cedex, France.
| | | | | |
Collapse
|
221
|
Dower K, Ellis DK, Saraf K, Jelinsky SA, Lin LL. Innate Immune Responses to TREM-1 Activation: Overlap, Divergence, and Positive and Negative Cross-Talk with Bacterial Lipopolysaccharide. THE JOURNAL OF IMMUNOLOGY 2008; 180:3520-34. [DOI: 10.4049/jimmunol.180.5.3520] [Citation(s) in RCA: 134] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
222
|
Duan SZ, Usher MG, Mortensen RM. Peroxisome Proliferator-Activated Receptor-γ–Mediated Effects in the Vasculature. Circ Res 2008; 102:283-94. [DOI: 10.1161/circresaha.107.164384] [Citation(s) in RCA: 216] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Peroxisome proliferator-activated receptor (PPAR)-γ is a nuclear receptor and transcription factor in the steroid superfamily. PPAR-γ agonists, the thiazolidinediones, are clinically used to treat type 2 diabetes. In addition to its function in adipogenesis and increasing insulin sensitivity, PPAR-γ also plays critical roles in the vasculature. In vascular endothelial cells, PPAR-γ activation inhibits endothelial inflammation by suppressing inflammatory gene expression and therefore improves endothelial dysfunction. In vascular smooth muscle cells, PPAR-γ activation inhibits proliferation and migration and promotes apoptosis. In macrophages, PPAR-γ activation suppresses inflammation by regulating gene expression and increases cholesterol uptake and efflux. A recurring theme in many cell types is the modulation of the innate immunity system particularly through altering the activity of the nuclear factor κB. This system is likely to be even more prominent in modulating disease in vascular cells. The effects of PPAR-γ in the vascular cells translate into the beneficial function of this transcription factor in vascular disorders, including hypertension and atherosclerosis. Both human genetic studies and animal studies using transgenic mice have demonstrated the importance of PPAR-γ in these disorders. However, recent clinical studies have raised significant concerns about the cardiovascular side effects of thiazolidinediones, particularly rosiglitazone. Weighing the potential benefit and harm of PPAR-γ activation and exploring the functional mechanisms may provide a balanced view on the clinical use of these compounds and new approaches to the future therapeutics of vascular disorders associated with diabetes.
Collapse
Affiliation(s)
- Sheng Zhong Duan
- From the Departments of Molecular and Integrative Physiology (S.Z.D., M.G.U., R.M.M.), Pharmacology (R.M.M.), and Internal Medicine (R.M.M.), Metabolism Endocrinology and Diabetes Division, University of Michigan Medical School, Ann Arbor
| | - Michael G. Usher
- From the Departments of Molecular and Integrative Physiology (S.Z.D., M.G.U., R.M.M.), Pharmacology (R.M.M.), and Internal Medicine (R.M.M.), Metabolism Endocrinology and Diabetes Division, University of Michigan Medical School, Ann Arbor
| | - Richard M. Mortensen
- From the Departments of Molecular and Integrative Physiology (S.Z.D., M.G.U., R.M.M.), Pharmacology (R.M.M.), and Internal Medicine (R.M.M.), Metabolism Endocrinology and Diabetes Division, University of Michigan Medical School, Ann Arbor
| |
Collapse
|
223
|
Tiwari R, Singh V, Barthwal M. Macrophages: An elusive yet emerging therapeutic target of atherosclerosis. Med Res Rev 2008; 28:483-544. [DOI: 10.1002/med.20118] [Citation(s) in RCA: 112] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
224
|
Bouhlel MA, Staels B, Chinetti-Gbaguidi G. Peroxisome proliferator-activated receptors--from active regulators of macrophage biology to pharmacological targets in the treatment of cardiovascular disease. J Intern Med 2008; 263:28-42. [PMID: 18042221 DOI: 10.1111/j.1365-2796.2007.01892.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Altered macrophage functions contribute to the pathogenesis of many infectious, immunological and inflammatory disease processes. Pharmacological modulation of macrophage activities therefore represents an important strategy for the prevention and treatment of inflammation-related diseases, such as atherosclerosis. This review focuses on recent advances on the role of the peroxisome proliferator-activated receptor transcription factor family in the modulation of lipid homeostasis and the inflammatory response in macrophages and the potential participation of these actions in the modulation of metabolic and cardiovascular disease.
Collapse
Affiliation(s)
- M A Bouhlel
- Département d'Athérosclérose, Institut Pasteur de Lille, Lille, France
| | | | | |
Collapse
|
225
|
Popovic ZV, Sandhoff R, Sijmonsma TP, Kaden S, Jennemann R, Kiss E, Tone E, Autschbach F, Platt N, Malle E, Gröne HJ. Sulfated glycosphingolipid as mediator of phagocytosis: SM4s enhances apoptotic cell clearance and modulates macrophage activity. THE JOURNAL OF IMMUNOLOGY 2007; 179:6770-82. [PMID: 17982067 DOI: 10.4049/jimmunol.179.10.6770] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Sulfoglycolipids are present on the surface of a variety of cells. The sulfatide SM4s is increased in lung, renal, and colon cancer and is associated with an adverse prognosis, possibly due to a low immunoreactivity of the tumor. As macrophages significantly contribute to the inflammatory infiltrate in malignancies, we postulated that SM4s may modulate macrophage function. We have investigated the effect of SM4s on the uptake of apoptotic tumor cells, macrophage cytokine profile, and receptor expression. Using flow cytometry and microscopic analyses, we found that coating apoptotic murine carcinoma cells from the colon and kidney with SM4s promoted their phagocytosis by murine macrophages up to 3-fold ex vivo and in vivo. This increased capacity was specifically inhibited by preincubation of macrophages with oxidized or acetylated low density lipoprotein and maleylated albumin, indicating involvement of scavenger receptors in this interaction. The uptake of SM4s-coated apoptotic cells significantly enhanced macrophage production of TGF-beta1, expression of P-selectin, and secretion of IL-6. These data suggest that SM4s within tumors may promote apoptotic cell removal and alter the phenotype of tumor-associated macrophages.
Collapse
Affiliation(s)
- Zoran V Popovic
- Department of Cellular and Molecular Pathology, German Cancer Research Center, Heidelberg, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
226
|
Abstract
Patients with type-2 diabetes mellitus (T2DM) are considered to be at particularly high risk for cardiovascular disease. Over the last decade, the members of the peroxisome proliferator-activated receptor (PPAR) subfamily of nuclear receptors have emerged as valuable pharmacological targets whose activation can normalize metabolic dysfunctions and reduce some cardiovascular risk factors associated with T2DM. PPARalpha agonists, such as the fibrates, can correct dyslipidemia. PPARgamma agonists, such as the thiazolidinediones, act as insulin sensitizers and improve insulin resistance in patients with T2DM. Because of restricted potency and certain side-effects of PPAR agonists, as well as the increasingly epidemic incidence of T2DM, there is a real need for the development of selective PPAR agonists with improved clinical efficacy. This chapter focuses on the PPAR agonists currently used in the clinic, as well as on the discovery and development of the next generation of PPAR agonists.
Collapse
Affiliation(s)
- Barbara Gross
- Institut Pasteur de Lille, 1 rue du Prof Calmette, Lille, F-59019, France
| | | |
Collapse
|
227
|
Fan Y, Wang Y, Tang Z, Zhang H, Qin X, Zhu Y, Guan Y, Wang X, Staels B, Chien S, Wang N. Suppression of pro-inflammatory adhesion molecules by PPAR-delta in human vascular endothelial cells. Arterioscler Thromb Vasc Biol 2007; 28:315-21. [PMID: 18048767 DOI: 10.1161/atvbaha.107.149815] [Citation(s) in RCA: 121] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
OBJECTIVE Endothelial activation is implicated in atherogenesis and diabetes. The role of peroxisome proliferator-activated receptor-delta (PPAR-delta) in endothelial activation remains poorly understood. In this study, we investigated the anti-inflammatory effect of PPAR-delta and the mechanism involved. METHODS AND RESULTS In human umbilical vein endothelial cells (HUVECs), the synthetic PPAR-delta ligands GW0742 and GW501516 significantly inhibited tumor necrosis factor (TNF)-alpha-induced expression of vascular cell adhesion molecule-1 and E-selectin (assayed by real-time RT-PCR and Northern blotting), as well as the ensuing endothelial-leukocyte adhesion. Activation of PPAR-delta upregulated the expression of antioxidant genes superoxide dismutase 1, catalase, and thioredoxin and decreased reactive oxygen species production in ECs. Chromatin immunoprecipitation assays showed that GW0742 switched the association of BCL-6, a transcription repressor, from PPAR-delta to the vascular cell adhesion molecule (VCAM)-1 promoter. Small interfering RNA reduced endogenous PPAR-delta expression but potentiated the suppressive effect of GW0742 on EC activation, which suggests that the nonliganded PPAR-delta may have an opposite effect. CONCLUSIONS We have demonstrated that ligand activation of PPAR-delta in ECs has a potent antiinflammatory effect, probably via a binary mechanism involving the induction of antioxidative genes and the release of nuclear corepressors. PPAR-delta agonists may have a potential for treating inflammatory diseases such as atherosclerosis and diabetes.
Collapse
Affiliation(s)
- Yanbo Fan
- Institute of Cardiovascular Science and Key Laboratory of Molecular Cardiovascular Science, Peking University Health Science Center, Beijing 100083, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
228
|
Allahtavakoli M, Shabanzadeh A, Roohbakhsh A, Pourshanazari A. Combination therapy of rosiglitazone, a peroxisome proliferator-activated receptor-gamma ligand, and NMDA receptor antagonist (MK-801) on experimental embolic stroke in rats. Basic Clin Pharmacol Toxicol 2007; 101:309-14. [PMID: 17910613 DOI: 10.1111/j.1742-7843.2007.00127.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Peroxisome proliferator-activated receptor-gamma (PPAR-gamma) agonists have been found to have potent anti-inflammatory actions and suggested as potential therapies for brain ischaemia. Glutamate is the most common excitatory neurotransmitter in the central nervous system and is released excessively during ischaemia. Stroke therapy will require combinations of drug classes, because no single drug class has yet been proven efficacious in human beings. The present study was conducted to assess whether N-methyl-d-aspartate (NMDA) receptor antagonist (MK-801) treatment can improve recovery from ischaemic brain injury and whether rosiglitazone, a PPAR-gamma ligand, can increase its neuroprotective effect in an embolic model of stroke. Stroke was induced in rats by embolizing a preformed clot into the middle cerebral artery. Rosiglitazone (0.1 mg/kg, intraperitoneally) and MK-801 (0.1 mg/kg, intravenously) were injected immediately after embolization. Forty-eight hours later, the brains were removed, sectioned and stained with triphenyltetrazolum chloride and analysed by a commercial image processing software programme. Rosiglitazone and MK-801 alone or in combination decreased infarct volume by 49.16%, 50.26% and 81.32%, respectively (P < 0.001). Moreover, the combination therapy significantly decreased the infarct volume when compared to any drug used alone (P < 0.05). MK-801 reduced the brain oedema by 68% compared to the control group (P < 0.05), but rosiglitazone or combination did not show any significant effect. The drugs alone or in combination also demonstrated improved neurological function, but combination therapy was more effective on neurological deficits improving. Our data show that the combination of MK-801 and rosiglitazone is more neuroprotective in thromboembolic stroke than given alone; this effect perhaps represents a possible additive effect in the brain infarction.
Collapse
Affiliation(s)
- Mohammad Allahtavakoli
- Department of Physiology and Pharmacology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| | | | | | | |
Collapse
|
229
|
Szanto A, Rőszer T. Nuclear receptors in macrophages: A link between metabolism and inflammation. FEBS Lett 2007; 582:106-16. [DOI: 10.1016/j.febslet.2007.11.020] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2007] [Accepted: 11/06/2007] [Indexed: 10/22/2022]
|
230
|
Madore AM, Houde L, Vézina H, Vohl MC, Pérusse L, Mior N, Connelly PW, Laberge C, Gaudet D, Laprise C. Contribution of hierarchical clustering techniques to the modeling of the geographic distribution of genetic polymorphisms associated with chronic inflammatory diseases in the Québec population. Public Health Genomics 2007; 10:218-26. [PMID: 17895627 DOI: 10.1159/000106560] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
OBJECTIVES The purpose of this project was to evaluate the potential of the downward hierarchical clustering analysis (DHCA) for studying genetic heterogeneity, i.e. differences in allele frequency in subpopulations, such as the 15 public health regions of the province of Québec (Canada). METHODS The study relied on an anonymized sample of 1,680 individuals who had participated in the Québec Heart Health Survey in 1990-1991. The genotyping of 11 variants in 8 candidate genes known to be involved in chronic inflammatory diseases, namely asthma and cardiovascular diseases, was performed using the amplification refractory mutation system and restriction fragment length polymorphism techniques. Only variants showing an allelic frequency >2% in the Québec Heart Health Survey (n = 8) were selected. DHCA techniques were then applied to model the geographical distribution of these 8 genetic variants in 15 Québec public health regions and to study genetic heterogeneity. RESULTS The DHCA allowed to group public health regions and gene variants on the basis of genetic variability. For both asthma and cardiovascular diseases, 3 significant clusters of public health regions and 1 cluster of gene variants were identified. DISCUSSION This study suggests that DHCA might be useful in studying genetic heterogeneity at the population level and for public health activities.
Collapse
Affiliation(s)
- A-M Madore
- University of Montreal Community Genomic Medicine Center, Chicoutimi University Hospital, Chicoutimi, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
231
|
Girroir EE, Hollingshead HE, Billin AN, Willson TM, Robertson GP, Sharma AK, Amin S, Gonzalez FJ, Peters JM. Peroxisome proliferator-activated receptor-beta/delta (PPARbeta/delta) ligands inhibit growth of UACC903 and MCF7 human cancer cell lines. Toxicology 2007; 243:236-43. [PMID: 18054822 DOI: 10.1016/j.tox.2007.10.023] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2007] [Revised: 10/10/2007] [Accepted: 10/30/2007] [Indexed: 12/19/2022]
Abstract
The development of peroxisome proliferator-activated receptor-beta/delta (PPARbeta/delta) ligands for the treatment of diseases including metabolic syndrome, diabetes and obesity has been hampered due to contradictory findings on their potential safety. For example, while some reports show that ligand activation of PPARbeta/delta promotes the induction of terminal differentiation and inhibition of cell growth, other reports suggest that PPARbeta/delta ligands potentiate tumorigenesis by increasing cell proliferation. Some of the contradictory findings could be due in part to differences in the ligand examined, the presence or absence of serum in cell cultures, differences in cell lines or differences in the method used to quantify cell growth. For these reasons, this study examined the effect of ligand activation of PPARbeta/delta on cell growth of two human cancer cell lines, MCF7 (breast cancer) and UACC903 (melanoma) in the presence or absence of serum using two highly specific PPARbeta/delta ligands, GW0742 or GW501516. Culturing cells in the presence of either GW0742 or GW501516 caused upregulation of the known PPARbeta/delta target gene angiopoietin-like protein 4 (ANGPTL4). Inhibition of cell growth was observed in both cell lines cultured in the presence of either GW0742 or GW501516, and the presence or absence of serum had little influence on this inhibition. Results from the present studies demonstrate that ligand activation of PPARbeta/delta inhibits the growth of both MCF7 and UACC903 cell lines and provide further evidence that PPARbeta/delta ligands are not mitogenic in human cancer cell lines.
Collapse
Affiliation(s)
- Elizabeth E Girroir
- Department of Veterinary and Biomedical Sciences and The Center of Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA 16802, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
232
|
Chinetti-Gbaguidi G, Staels B. Measuring biomarkers to assess the therapeutic effects of PPAR agonists? Pharmacogenomics 2007; 8:1567-80. [DOI: 10.2217/14622416.8.11.1567] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The metabolic syndrome is defined as a clustering of cardiovascular risk factors with insulin resistance, including dyslipidemia, coagulation disturbances and hypertension. Activators of the peroxisome proliferator-activated receptors (PPARs) modulate several of the metabolic risk factors predisposing to atherosclerosis. Fibrates are hypolipidemic drugs acting through activation of PPARα, whereas glitazones are insulin sensitizers activating PPARγ. In addition, these drugs exert pleiotropic anti-inflammatory actions. In this review, we will focus on the effects of fibrates and glitazones on biomarker modulation and their usefulness in the treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Giulia Chinetti-Gbaguidi
- Institut Pasteur de Lille, 1, Rue Calmette BP245, Lille, F-59019, France
- Inserm, U545, Lille, F-59019, France
- Université de Lille, Faculté des Sciences Pharmaceutiques et Biologiques et Faculté de Médecine, Lille, F-59006, France
| | - Bart Staels
- Institut Pasteur de Lille, 1, Rue Calmette BP245, Lille, F-59019, France
- Inserm, U545, Lille, F-59019, France
- Université de Lille, Faculté des Sciences Pharmaceutiques et Biologiques et Faculté de Médecine, Lille, F-59006, France
| |
Collapse
|
233
|
Szatmari I, Töröcsik D, Agostini M, Nagy T, Gurnell M, Barta E, Chatterjee K, Nagy L. PPARgamma regulates the function of human dendritic cells primarily by altering lipid metabolism. Blood 2007; 110:3271-80. [PMID: 17664351 DOI: 10.1182/blood-2007-06-096222] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Activation of the lipid-regulated nuclear receptor peroxisome proliferator-activated receptor gamma (PPARgamma) modifies the immunophenotype of monocyte-derived dendritic cells (DCs). However it has not been analyzed in a systematic manner how lipid metabolism and immune regulation are connected at the transcriptional level via this receptor. Here we present the genome-wide expression analyses of PPARgamma-instructed human DCs. Receptor activation was achieved by exogenous, synthetic as well as endogenous, natural means. More than 1000 transcripts are regulated during DC development by activation of PPARgamma; half of the changes are positive effects. These changes appear to enhance and modulate the robust gene expression alterations associated with monocyte to DC transition. Strikingly, only genes related to lipid metabolism are overrepresented among early induced genes. As a net consequence, lipid accumulation appears to be diminished in these cells. In contrast, genes related to immune response are regulated after 24 hours, implying the existence of indirect mechanisms of modulation. Receptor dependence was established by using DCs of patients harboring a dominant-negative mutation of PPARgamma. Our data show that PPARgamma acts as a mostly positive transcriptional regulator in human developing DCs, acting primarily through controlling genes involved in lipid metabolism and via this, indirectly modifying the immune phenotype.
Collapse
Affiliation(s)
- Istvan Szatmari
- Department of Biochemistry and Molecular Biology, Research Center for Molecular Medicine, University of Debrecen, Medical and Health Science Center, Nagyerdei krt. 98, Debrecen, Hungary
| | | | | | | | | | | | | | | |
Collapse
|
234
|
Hollingshead HE, Morimura K, Adachi M, Kennett MJ, Billin AN, Willson TM, Gonzalez FJ, Peters JM. PPARbeta/delta protects against experimental colitis through a ligand-independent mechanism. Dig Dis Sci 2007; 52:2912-9. [PMID: 17404849 DOI: 10.1007/s10620-006-9644-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2006] [Accepted: 10/09/2006] [Indexed: 12/12/2022]
Abstract
Peroxisome proliferator-activated receptors (PPARs) beta/delta and gamma have overlapping roles in the negative regulation of inflammatory response genes. Ligand activation of PPARgamma protects against experimental colitis in mice. PPARbeta/delta can negatively regulate inflammation and is highly expressed in the epithelial cells of the colon, therefore PPARbeta/delta may also have a role in experimental colitis. In these studies, colitis was induced by dextran sodium sulfate (DSS) treatment in wild-type and PPARbeta/delta-null mice, with and without the PPARbeta/delta specific ligand GW0742. PPARbeta/delta-null mice exhibited increased sensitivity to DSS-induced colitis, as shown by marked differences in body weight loss, colon length, colonic morphology, myeloperoxidase activity and increased expression of mRNAs encoding the inflammatory markers interferon gamma, tumor necrosis factor-alpha, and interleukin-6 compared to similarly treated wild-type mice. Interestingly, these differences were not affected by ligand activation of PPARbeta/delta in either genotype. These studies demonstrate that PPARbeta/delta expression in the colonic epithelium inhibits inflammation and protects against DSS-induced colitis through a ligand-independent mechanism.
Collapse
Affiliation(s)
- Holly E Hollingshead
- Department of Veterinary and Biomedical Sciences , The Pennsylvania State University, 312 Life Sciences Building, University Park, Pennsylvania 16802, USA
| | | | | | | | | | | | | | | |
Collapse
|
235
|
Pascual G, Ricote M, Hevener AL. Macrophage peroxisome proliferator activated receptor γ as a therapeutic target to combat Type 2 diabetes. Expert Opin Ther Targets 2007; 11:1503-20. [DOI: 10.1517/14728222.11.11.1503] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
236
|
Wray J, Bishop-Bailey D. Epoxygenases and peroxisome proliferator-activated receptors in mammalian vascular biology. Exp Physiol 2007; 93:148-54. [PMID: 17872966 DOI: 10.1113/expphysiol.2007.038612] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Epoxygenases, particularly of the CYP2C and CYP2J families, are important lipid-metabolizing enzymes. Epoxygenases are found throughout the cardiovascular system where their lipid products, particularly the epoxyeicosatrienoic acids (EETs), which are arachidonic acid metabolites, have the potential to regulate vascular tone, cellular proliferation, migration, inflammation and cardiac function. The receptors for EETs are, however, poorly understood. The peroxisome proliferator-activated receptors (PPARs) are a family of three (alpha, beta/delta and gamma) nuclear receptors that are activated by lipid metabolites. Activation of PPAR alpha and PPAR gamma, similar to the longer term effects of EETs, causes the inhibition of vascular cell proliferation, migration and inflammation. Interestingly, EETs and their metabolites have recently been found to active both PPAR alpha and PPAR gamma. The epoxygenase-EET-PPAR pathway may therefore represent a novel endogenous protective pathway by which short-lived lipid mediators control vascular cell activation.
Collapse
Affiliation(s)
- Jessica Wray
- Translational Medicine and Therapeutics, William Harvey Research Institute, Barts and the London, Queen Mary University London, Charterhouse Square, London EC1M 6BQ, UK
| | | |
Collapse
|
237
|
Hollingshead HE, Borland MG, Billin AN, Willson TM, Gonzalez FJ, Peters JM. Ligand activation of peroxisome proliferator-activated receptor-beta/delta (PPARbeta/delta) and inhibition of cyclooxygenase 2 (COX2) attenuate colon carcinogenesis through independent signaling mechanisms. Carcinogenesis 2007; 29:169-76. [PMID: 17893232 DOI: 10.1093/carcin/bgm209] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Cyclooxygenase (COX) 2-derived prostaglandin E(2) (PGE(2)) promotes colorectal carcinoma growth and invasion, and inhibition of COX2 by non-steroidal anti-inflammatory drugs is known to inhibit these processes. There is controversy regarding the effect of ligand activation of peroxisome proliferator-activated receptor (PPAR)-beta/delta on colon carcinogenesis, although collective evidence from independent laboratories suggest that ligand activation of PPARbeta/delta leads to the induction of terminal differentiation coupled with inhibition of cell growth in a variety of models. The present study examined the hypothesis that ligand activation of PPARbeta/delta and inhibition of COX2 attenuate colon cancer through independent mechanisms and that combining these two mechanisms will enhance this inhibition. Colon cancer was induced by administering azoxymethane to wild-type and PPARbeta/delta-null mice. Cohorts of mice were treated with GW0742 (a PPARbeta/delta ligand), nimesulide (a COX2 inhibitor) or a combination of GW0742 and nimesulide. Inhibition of COX2 by nimesulide attenuated colon cancer and ligand activation of PPARbeta/delta by GW0742 had inhibitory effects. However, the combined treatment of GW0742 and nimesulide did not cause an enhancement in the attenuation of colon cancer. Mechanistically, the effects of these compounds occurred through independent mechanisms as increased levels of differentiation markers as a result of ligand activation of PPARbeta/delta were not found with COX2 inhibition, and a reduction in PGE(2) levels resulting from COX2 inhibition was not observed in response to ligand activation of PPARbeta/delta. Results from these studies effectively dissociate COX2 inhibition and PPARbeta/delta activity during colon carcinogenesis.
Collapse
Affiliation(s)
- Holly E Hollingshead
- Department of Veterinary and Biomedical Sciences and The Center of Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA 16802, USA
| | | | | | | | | | | |
Collapse
|
238
|
Wu-Wong JR, Nakane M, Ma J, Ruan X, Kroeger PE. Elevated phosphorus modulates vitamin D receptor-mediated gene expression in human vascular smooth muscle cells. Am J Physiol Renal Physiol 2007; 293:F1592-604. [PMID: 17715259 DOI: 10.1152/ajprenal.00492.2006] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Clinical observations show that an increase in serum inorganic phosphorus (Pi) is linked to higher cardiovascular (CV) mortality, while vitamin D receptor (VDR) agonist therapy is associated with survival benefit in stage 5 chronic kidney disease. Smooth muscle cells (SMCs) play an important role in CV pathophysiology, but the interaction between Pi and the VDR signaling pathway in SMCs is not known. Real-time RT-PCR studies revealed that elevated Pi (2.06 mM) modulated VDR-mediated regulation of a panel of genes including thrombomodulin and osteopontin in SMCs. DNA microarray results demonstrated that increasing Pi from 0.9 to 2.06 mM exerted a widespread modulating effect on VDR-mediated gene expression. A total of 325 target genes were affected by paricalcitol at 0.9 mM Pi, with 195 up- and 130 downregulated. The number of target genes affected by paricalcitol at 2.06 mM Pi decreased to 86, with 55 up- and 31 downregulated. VDR-mediated gene expression in As4.1 cells (a juxtaglomerular cell-like cell line derived from kidney tumors in SV40 T-antigen transgenic mice) and peroxisome proliferator-activated receptor (PPAR)gamma-mediated gene expression in SMCs were also altered by elevated Pi, suggesting that the observation is not unique to VDR in SMCs. Mechanism analysis showed that elevated Pi had no significant effect on VDR or PPARgamma protein level but altered the cytosolic vs. nuclear distribution of NF-kappaB or nuclear receptor corepressor 1 (NCoR1). Our results demonstrate for the first time that elevated Pi affects VDR-mediated gene expression in human coronary artery SMCs and the effect is not limited to VDR in SMCs.
Collapse
|
239
|
Hollingshead HE, Killins RL, Borland MG, Girroir EE, Billin AN, Willson TM, Sharma AK, Amin S, Gonzalez FJ, Peters JM. Peroxisome proliferator-activated receptor-beta/delta (PPARbeta/delta) ligands do not potentiate growth of human cancer cell lines. Carcinogenesis 2007; 28:2641-9. [PMID: 17693664 DOI: 10.1093/carcin/bgm183] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Ligands for peroxisome proliferator-activated receptor-beta/delta (PPARbeta/delta) increase skeletal muscle fatty acid catabolism, improve insulin sensitivity, increase serum high-density lipoprotein cholesterol, elicit anti-inflammatory activity and induce terminal differentiation. Contradictory findings are also reported suggesting that PPARbeta/delta ligands potentiate tumorigenesis by increasing cell proliferation, by inhibiting apoptosis through phosphorylation of Akt and by increasing cyclooxygenase-2 (COX2) and vascular endothelial growth factor (VEGF) expression. The contradictory findings could be due to differences in the model system (cancer cell line versus in vivo), differences in cell culture conditions (with and without serum) or differences in ligands. The present study examined the effect of two different PPARbeta/delta ligands (GW0742 and GW501516) in human cancer cell lines (HT29, HCT116, LS-174T, HepG2 and HuH7) cultured in the presence or absence of serum and compared in vitro analysis with in vivo analysis. Neither PPARbeta/delta ligand increased cell growth or phosphorylation of Akt and no increase in the expression of VEGF or COX2 were detected in any cancer cell line in the presence or absence of serum. Similarly, liver, colon and colon polyps from mice administered these PPARbeta/delta ligands in vivo did not exhibit changes in these markers. Results from these studies demonstrate that serum withdrawal and/or differences in ligands do not underlie the disparity in responses reported in the literature. The quantitative nature of the present findings are inconsistent with the hypothesis that cancer cell lines respond differentially as compared with normal cells, and provide further evidence that PPARbeta/delta ligands do not potentiate tumorigenesis.
Collapse
Affiliation(s)
- Holly E Hollingshead
- Department of Veterinary and Biomedical Sciences and the Center of Molecular Toxicology and Carcinogenesis, Pennsylvania State University, University Park, PA 16802, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
240
|
Monroy MA, Opperman KK, Pucciarelli M, Yerrum S, Berg DA, Daly JM. THE PPARγ LIGAND 15d-PGJ2 MODULATES MACROPHAGE ACTIVATION AFTER INJURY IN A MURINE TRAUMA MODEL. Shock 2007; 28:186-91. [PMID: 17510607 DOI: 10.1097/shk.0b013e3180310982] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
In macrophages, peroxisome proliferator-activated receptor gamma (PPARgamma) has been shown to be important for differentiation, and it serves as a negative regulator of activation. Major trauma/injury causes a dramatic host response that disrupts cellular immune homeostasis and initiates an inflammatory cascade that predisposes the injured host to subsequent infections. In prior studies using a murine trauma model consisting of femur fracture and hemorrhage, splenic macrophages from traumatized mice had significantly enhanced LPS-induced cyclooxygenase enzyme (subtype 2) and iNOS production as well as elevated levels of inflammatory cytokines at 1 week after injury compared with uninjured controls. These up-regulated cellular responses corresponded to increased mortality when animals were challenged with LPS or Candida. In the current study, we used the injury model to determine the effect of treatment of injured mice with the endogenous PPARgamma ligand 15-deoxy-Delta(12-, 14)-PGJ2 (15d-PGJ2). It was found that in vivo 15d-PGJ2 treatment significantly reduced the levels of inflammatory mediators produced by splenic macrophages 7 days after injury. The mechanism of inhibition is dependent on PPARgamma because concomitant treatment of animals with the PPARgamma antagonist GW9662 reversed the inhibitory effect of 15d-PGJ2. Endogenous PPARgamma modulated activation of LPS-induced p38 mitogen-activated protein kinase. Furthermore, treatment of injured mice with 15d-PGJ2 conferred a significant survival advantage after infectious challenge induced by cecal ligation and puncture. Thus, this PPARgamma ligands significantly attenuate the postinjury inflammatory response and improve survival after infectious challenge.
Collapse
Affiliation(s)
- M Alexandra Monroy
- Department of Surgery, Temple University School of Medicine, 3400 North Broad Street, Philadelphia, PA 19140, USA.
| | | | | | | | | | | |
Collapse
|
241
|
Guri AJ, Hontecillas R, Ferrer G, Casagran O, Wankhade U, Noble AM, Eizirik DL, Ortis F, Cnop M, Liu D, Si H, Bassaganya-Riera J. Loss of PPAR gamma in immune cells impairs the ability of abscisic acid to improve insulin sensitivity by suppressing monocyte chemoattractant protein-1 expression and macrophage infiltration into white adipose tissue. J Nutr Biochem 2007; 19:216-28. [PMID: 17618105 DOI: 10.1016/j.jnutbio.2007.02.010] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2006] [Revised: 02/06/2007] [Accepted: 02/14/2007] [Indexed: 11/25/2022]
Abstract
Abscisic acid (ABA) is a natural phytohormone and peroxisome proliferator-activated receptor gamma (PPARgamma) agonist that significantly improves insulin sensitivity in db/db mice. Although it has become clear that obesity is associated with macrophage infiltration into white adipose tissue (WAT), the phenotype of adipose tissue macrophages (ATMs) and the mechanisms by which insulin-sensitizing compounds modulate their infiltration remain unknown. We used a loss-of-function approach to investigate whether ABA ameliorates insulin resistance through a mechanism dependent on immune cell PPARgamma. We characterized two phenotypically distinct ATM subsets in db/db mice based on their surface expression of F4/80. F4/80(hi) ATMs were more abundant and expressed greater concentrations of chemokine receptor (CCR) 2 and CCR5 when compared to F4/80(lo) ATMs. ABA significantly decreased CCR2(+) F4/80(hi) infiltration into WAT and suppressed monocyte chemoattractant protein-1 (MCP-1) expression in WAT and plasma. Furthermore, the deficiency of PPARgamma in immune cells, including macrophages, impaired the ability of ABA to suppress the infiltration of F4/80(hi) ATMs into WAT, to repress WAT MCP-1 expression and to improve glucose tolerance. We provide molecular evidence in vivo demonstrating that ABA improves insulin sensitivity and obesity-related inflammation by inhibiting MCP-1 expression and F4/80(hi) ATM infiltration through a PPARgamma-dependent mechanism.
Collapse
Affiliation(s)
- Amir J Guri
- Laboratory of Nutritional Immunology and Molecular Nutrition, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
242
|
Xu Y, Farmer SR, Smith BD. Peroxisome proliferator-activated receptor gamma interacts with CIITA x RFX5 complex to repress type I collagen gene expression. J Biol Chem 2007; 282:26046-56. [PMID: 17611194 DOI: 10.1074/jbc.m703652200] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Recent reports demonstrate that peroxisome proliferator-activated receptor gamma (PPARgamma), a member of the nuclear receptor superfamily, acts as a repressor of type I collagen synthesis. Our data demonstrate that exogenously expressed PPARgamma down-regulates collagen expression in a dose-responsive manner in human lung fibroblast cells. Silencing PPARgamma using lentiviruses expressing short hairpin RNAs partially reverses interferon-gamma (IFN-gamma)-induced repression and activates collagen mRNA levels. Previous studies indicate that IFN-gamma represses collagen gene expression and induces major histocompatibility complex II (MHC II) expression by activating the formation of a regulatory factor for X-box 5 (RFX5) complex with class II transactivator (CIITA). This report demonstrates that PPARgamma is within the RFX5.CIITA complex as judged by co-immunoprecipitation and DNA affinity precipitation studies. Most importantly, occupancy of PPARgamma on the collagen transcription start site and MHC II promoter increases with IFN-gamma treatment. The PPARgamma agonist, troglitazone, sensitizes the cells to IFN-gamma treatment by increasing recruitment of PPARgamma to collagen gene while repressing collagen expression, and these effects are blocked by the PPARgamma antagonist T0070907. PPARgamma may mediate IFN-gamma-stimulated collagen transcription down-regulation and MHC II up-regulation by interacting with CIITA as well as regulating CIITA expression. Therefore, PPARgamma is a critical target for investigations into therapeutics of diseases involving extracellular matrix remodeling and the immune response.
Collapse
Affiliation(s)
- Yong Xu
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | | | | |
Collapse
|
243
|
Liao W, Nguyen MTA, Yoshizaki T, Favelyukis S, Patsouris D, Imamura T, Verma IM, Olefsky JM. Suppression of PPAR-gamma attenuates insulin-stimulated glucose uptake by affecting both GLUT1 and GLUT4 in 3T3-L1 adipocytes. Am J Physiol Endocrinol Metab 2007; 293:E219-27. [PMID: 17389706 DOI: 10.1152/ajpendo.00695.2006] [Citation(s) in RCA: 102] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Peroxisome proliferator-activated receptor-gamma (PPAR-gamma) plays a critical role in regulating insulin sensitivity and glucose homeostasis. In this study, we identified highly efficient small interfering RNA (siRNA) sequences and used lentiviral short hairpin RNA and electroporation of siRNAs to deplete PPAR-gamma from 3T3-L1 adipocytes to elucidate its role in adipogenesis and insulin signaling. We show that PPAR-gamma knockdown prevented adipocyte differentiation but was not required for maintenance of the adipocyte differentiation state after the cells had undergone adipogenesis. We further demonstrate that PPAR-gamma suppression reduced insulin-stimulated glucose uptake without affecting the early insulin signaling steps in the adipocytes. Using dual siRNA strategies, we show that this effect of PPAR-gamma deletion was mediated by both GLUT4 and GLUT1. Interestingly, PPAR-gamma-depleted cells displayed enhanced inflammatory responses to TNF-alpha stimulation, consistent with a chronic anti-inflammatory effect of endogenous PPAR-gamma. In summary, 1) PPAR-gamma is essential for the process of adipocyte differentiation but is less necessary for maintenance of the differentiated state, 2) PPAR-gamma supports normal insulin-stimulated glucose transport, and 3) endogenous PPAR-gamma may play a role in suppression of the inflammatory pathway in 3T3-L1 cells.
Collapse
Affiliation(s)
- Wei Liao
- Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, CA 92093, USA
| | | | | | | | | | | | | | | |
Collapse
|
244
|
Lee JW, Bajwa PJ, Carson MJ, Jeske DR, Cong Y, Elson CO, Lytle C, Straus DS. Fenofibrate represses interleukin-17 and interferon-gamma expression and improves colitis in interleukin-10-deficient mice. Gastroenterology 2007; 133:108-23. [PMID: 17631136 DOI: 10.1053/j.gastro.2007.03.113] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2006] [Accepted: 03/22/2007] [Indexed: 12/02/2022]
Abstract
BACKGROUND & AIMS Interleukin-10 knockout (IL-10(-/-)) mice spontaneously develop colitis characterized by T-helper cell type 1-polarized inflammation. We tested the possible therapeutic activity of the peroxisome proliferator-activated receptor alpha (PPARalpha) ligand fenofibrate, and the PPARdelta ligand GW0742, in IL-10(-/-) mice and investigated the cellular/molecular mechanisms for fenofibrate action. METHODS The effect of fenofibrate or GW0742 on the progression of colitis in C3H.IL-10(-/-) mice was evaluated. Effects of fenofibrate on cytokine and chemokine gene expression were studied in cultured splenocytes, pathogenic T cells isolated from C3H/HeJBir mice, and HT-29 colorectal cancer cells. RESULTS Treatment of C3H.IL-10(-/-) mice with fenofibrate delayed the onset of colitis, decreased the colonic histopathology score, and decreased colonic expression of genes encoding the inflammatory cytokines interferon-gamma and interleukin (IL)-17. The target for fenofibrate, PPARalpha, was expressed in lymphocytes, macrophages, and crypt and surface epithelial cells of the colon. The mean number of lymphocytes was decreased by more than 75% in colonic sections of fenofibrate-treated as compared with control IL-10(-/-) mice, and fenofibrate repressed interferon-gamma and IL-17 expression in isolated T cells. Fenofibrate also repressed the expression of the genes encoding 3 chemokines, CXCL10, CCL2, and CCL20, and repressed CXCL10 gene promoter activity in tumor necrosis factor-alpha-treated HT-29 cells. In contrast to the beneficial effect of fenofibrate, the PPARdelta ligand GW0742 accelerated the onset of colitis in IL-10(-/-) mice. CONCLUSIONS The immunopathology observed in IL-10(-/-) mice resembles that seen in Crohn's disease. The novel therapeutic activity of fenofibrate in this mouse model suggests that it may also have activity in Crohn's disease.
Collapse
Affiliation(s)
- Jimmy W Lee
- Biomedical Sciences Division, University of California, Riverside, California 92521-0121, USA
| | | | | | | | | | | | | | | |
Collapse
|
245
|
Altmann R, Hausmann M, Spöttl T, Gruber M, Bull AW, Menzel K, Vogl D, Herfarth H, Schölmerich J, Falk W, Rogler G. 13-Oxo-ODE is an endogenous ligand for PPARgamma in human colonic epithelial cells. Biochem Pharmacol 2007; 74:612-22. [PMID: 17604003 DOI: 10.1016/j.bcp.2007.05.027] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2007] [Revised: 05/14/2007] [Accepted: 05/31/2007] [Indexed: 11/27/2022]
Abstract
BACKGROUND The ligand activated nuclear hormone receptor peroxisome proliferator-activated receptor gamma (PPARgamma) induces transcriptional repression of pro-inflammatory factors. Activation of PPARgamma is followed by amelioration of colitis in animal models of inflammatory bowel disease (IBD). A reduced expression of PPARgamma was found in epithelial cells of patients with ulcerative colitis. The eicosanoids 13-HODE and 15-HETE are products of 12/15-lipoxygenase (LOX) and endogenous ligands for PPARgamma. Dehydrogenation of 13-HODE by 13-HODE dehydrogenase results in formation of the 13-Oxo-ODE. Highest activity of 13-HODE dehydrogenase is found in colonic epithelial cells (CECs). We therefore investigated whether 13-Oxo-ODE is a new endogenous ligand of PPARgamma in CECs. METHODS LOX activity and 13-HODE dehydrogenase in CECs were investigated after stimulation with arachidonic or linoleic acid. LOX metabolites were identified by RP-18 reversed-phase HPLC. Binding of (14)C-labelled 13-Oxo-ODE was demonstrated using a His-tagged PPARgamma. RESULTS Stimulation of HT-29 and primary CECs homogenates with and without Ca-ionophor was followed by the formation of high amounts of the linoleic acid metabolite 13-Oxo-ODE (155 and 85 ng/ml). The decrease of IL-8 secretion from IEC was more pronounced after pre-incubation with 13-Oxo-ODE compared to the PPARgamma agonist troglitazone and higher as with the known PPARgamma ligands 13-HODE and 15-HETE. Binding assays with (14)C-labelled 13-Oxo-ODE clearly demonstrated a direct interaction. CONCLUSION High amounts of 13-Oxo-ODE can be induced in CECs by stimulation of linoleic acid metabolism. 13-Oxo-ODE binds to PPARgamma and has anti-inflammatory effects. 13-HODE dehydrogenase might be a therapeutic target in IBD.
Collapse
Affiliation(s)
- Reinhold Altmann
- Department of Internal Medicine I, University of Regensburg, 93042 Regensburg, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
246
|
Affiliation(s)
- Sadao Takahashi
- Third Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Japan
| | | | | |
Collapse
|
247
|
Hevener AL, Olefsky JM, Reichart D, Nguyen MA, Bandyopadyhay G, Leung HY, Watt MJ, Benner C, Febbraio MA, Nguyen AK, Folian B, Subramaniam S, Gonzalez FJ, Glass CK, Ricote M. Macrophage PPAR gamma is required for normal skeletal muscle and hepatic insulin sensitivity and full antidiabetic effects of thiazolidinediones. J Clin Invest 2007; 117:1658-69. [PMID: 17525798 PMCID: PMC1868788 DOI: 10.1172/jci31561] [Citation(s) in RCA: 386] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2007] [Accepted: 03/20/2007] [Indexed: 02/06/2023] Open
Abstract
PPAR gamma is required for fat cell development and is the molecular target of antidiabetic thiazolidinediones (TZDs), which exert insulin-sensitizing effects in adipose tissue, skeletal muscle, and liver. Unexpectedly, we found that inactivation of PPAR gamma in macrophages results in the development of significant glucose intolerance plus skeletal muscle and hepatic insulin resistance in lean mice fed a normal diet. This phenotype was associated with increased expression of inflammatory markers and impaired insulin signaling in adipose tissue, muscle, and liver. PPAR gamma-deficient macrophages secreted elevated levels of factors that impair insulin responsiveness in muscle cells in a manner that was enhanced by exposure to FFAs. Consistent with this, the relative degree of insulin resistance became more severe in mice lacking macrophage PPAR gamma following high-fat feeding, and these mice were only partially responsive to TZD treatment. These findings reveal an essential role of PPAR gamma in macrophages for the maintenance of whole-body insulin action and in mediating the antidiabetic actions of TZDs.
Collapse
Affiliation(s)
- Andrea L. Hevener
- Department of Medicine, Division of Endocrinology and Metabolism, and
Department of Cellular and Molecular Medicine, UCSD, La Jolla, California, USA.
St. Vincent’s Institute of Medical Research, Department of Medicine, The University of Melbourne, Fitzroy, Victoria, Australia.
Department of Bioengineering, UCSD, La Jolla, California, USA.
Baker Heart Institute, Cellular and Molecular Medicine Laboratory, St. Kilda, Victoria, Australia.
Laboratory of Metabolism, National Cancer Institute, Bethesda, Maryland, USA.
Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Jerrold M. Olefsky
- Department of Medicine, Division of Endocrinology and Metabolism, and
Department of Cellular and Molecular Medicine, UCSD, La Jolla, California, USA.
St. Vincent’s Institute of Medical Research, Department of Medicine, The University of Melbourne, Fitzroy, Victoria, Australia.
Department of Bioengineering, UCSD, La Jolla, California, USA.
Baker Heart Institute, Cellular and Molecular Medicine Laboratory, St. Kilda, Victoria, Australia.
Laboratory of Metabolism, National Cancer Institute, Bethesda, Maryland, USA.
Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Donna Reichart
- Department of Medicine, Division of Endocrinology and Metabolism, and
Department of Cellular and Molecular Medicine, UCSD, La Jolla, California, USA.
St. Vincent’s Institute of Medical Research, Department of Medicine, The University of Melbourne, Fitzroy, Victoria, Australia.
Department of Bioengineering, UCSD, La Jolla, California, USA.
Baker Heart Institute, Cellular and Molecular Medicine Laboratory, St. Kilda, Victoria, Australia.
Laboratory of Metabolism, National Cancer Institute, Bethesda, Maryland, USA.
Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - M.T. Audrey Nguyen
- Department of Medicine, Division of Endocrinology and Metabolism, and
Department of Cellular and Molecular Medicine, UCSD, La Jolla, California, USA.
St. Vincent’s Institute of Medical Research, Department of Medicine, The University of Melbourne, Fitzroy, Victoria, Australia.
Department of Bioengineering, UCSD, La Jolla, California, USA.
Baker Heart Institute, Cellular and Molecular Medicine Laboratory, St. Kilda, Victoria, Australia.
Laboratory of Metabolism, National Cancer Institute, Bethesda, Maryland, USA.
Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Gautam Bandyopadyhay
- Department of Medicine, Division of Endocrinology and Metabolism, and
Department of Cellular and Molecular Medicine, UCSD, La Jolla, California, USA.
St. Vincent’s Institute of Medical Research, Department of Medicine, The University of Melbourne, Fitzroy, Victoria, Australia.
Department of Bioengineering, UCSD, La Jolla, California, USA.
Baker Heart Institute, Cellular and Molecular Medicine Laboratory, St. Kilda, Victoria, Australia.
Laboratory of Metabolism, National Cancer Institute, Bethesda, Maryland, USA.
Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Ho-Yin Leung
- Department of Medicine, Division of Endocrinology and Metabolism, and
Department of Cellular and Molecular Medicine, UCSD, La Jolla, California, USA.
St. Vincent’s Institute of Medical Research, Department of Medicine, The University of Melbourne, Fitzroy, Victoria, Australia.
Department of Bioengineering, UCSD, La Jolla, California, USA.
Baker Heart Institute, Cellular and Molecular Medicine Laboratory, St. Kilda, Victoria, Australia.
Laboratory of Metabolism, National Cancer Institute, Bethesda, Maryland, USA.
Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Matthew J. Watt
- Department of Medicine, Division of Endocrinology and Metabolism, and
Department of Cellular and Molecular Medicine, UCSD, La Jolla, California, USA.
St. Vincent’s Institute of Medical Research, Department of Medicine, The University of Melbourne, Fitzroy, Victoria, Australia.
Department of Bioengineering, UCSD, La Jolla, California, USA.
Baker Heart Institute, Cellular and Molecular Medicine Laboratory, St. Kilda, Victoria, Australia.
Laboratory of Metabolism, National Cancer Institute, Bethesda, Maryland, USA.
Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Chris Benner
- Department of Medicine, Division of Endocrinology and Metabolism, and
Department of Cellular and Molecular Medicine, UCSD, La Jolla, California, USA.
St. Vincent’s Institute of Medical Research, Department of Medicine, The University of Melbourne, Fitzroy, Victoria, Australia.
Department of Bioengineering, UCSD, La Jolla, California, USA.
Baker Heart Institute, Cellular and Molecular Medicine Laboratory, St. Kilda, Victoria, Australia.
Laboratory of Metabolism, National Cancer Institute, Bethesda, Maryland, USA.
Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Mark A. Febbraio
- Department of Medicine, Division of Endocrinology and Metabolism, and
Department of Cellular and Molecular Medicine, UCSD, La Jolla, California, USA.
St. Vincent’s Institute of Medical Research, Department of Medicine, The University of Melbourne, Fitzroy, Victoria, Australia.
Department of Bioengineering, UCSD, La Jolla, California, USA.
Baker Heart Institute, Cellular and Molecular Medicine Laboratory, St. Kilda, Victoria, Australia.
Laboratory of Metabolism, National Cancer Institute, Bethesda, Maryland, USA.
Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Anh-Khoi Nguyen
- Department of Medicine, Division of Endocrinology and Metabolism, and
Department of Cellular and Molecular Medicine, UCSD, La Jolla, California, USA.
St. Vincent’s Institute of Medical Research, Department of Medicine, The University of Melbourne, Fitzroy, Victoria, Australia.
Department of Bioengineering, UCSD, La Jolla, California, USA.
Baker Heart Institute, Cellular and Molecular Medicine Laboratory, St. Kilda, Victoria, Australia.
Laboratory of Metabolism, National Cancer Institute, Bethesda, Maryland, USA.
Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Brian Folian
- Department of Medicine, Division of Endocrinology and Metabolism, and
Department of Cellular and Molecular Medicine, UCSD, La Jolla, California, USA.
St. Vincent’s Institute of Medical Research, Department of Medicine, The University of Melbourne, Fitzroy, Victoria, Australia.
Department of Bioengineering, UCSD, La Jolla, California, USA.
Baker Heart Institute, Cellular and Molecular Medicine Laboratory, St. Kilda, Victoria, Australia.
Laboratory of Metabolism, National Cancer Institute, Bethesda, Maryland, USA.
Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Shankar Subramaniam
- Department of Medicine, Division of Endocrinology and Metabolism, and
Department of Cellular and Molecular Medicine, UCSD, La Jolla, California, USA.
St. Vincent’s Institute of Medical Research, Department of Medicine, The University of Melbourne, Fitzroy, Victoria, Australia.
Department of Bioengineering, UCSD, La Jolla, California, USA.
Baker Heart Institute, Cellular and Molecular Medicine Laboratory, St. Kilda, Victoria, Australia.
Laboratory of Metabolism, National Cancer Institute, Bethesda, Maryland, USA.
Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Frank J. Gonzalez
- Department of Medicine, Division of Endocrinology and Metabolism, and
Department of Cellular and Molecular Medicine, UCSD, La Jolla, California, USA.
St. Vincent’s Institute of Medical Research, Department of Medicine, The University of Melbourne, Fitzroy, Victoria, Australia.
Department of Bioengineering, UCSD, La Jolla, California, USA.
Baker Heart Institute, Cellular and Molecular Medicine Laboratory, St. Kilda, Victoria, Australia.
Laboratory of Metabolism, National Cancer Institute, Bethesda, Maryland, USA.
Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Christopher K. Glass
- Department of Medicine, Division of Endocrinology and Metabolism, and
Department of Cellular and Molecular Medicine, UCSD, La Jolla, California, USA.
St. Vincent’s Institute of Medical Research, Department of Medicine, The University of Melbourne, Fitzroy, Victoria, Australia.
Department of Bioengineering, UCSD, La Jolla, California, USA.
Baker Heart Institute, Cellular and Molecular Medicine Laboratory, St. Kilda, Victoria, Australia.
Laboratory of Metabolism, National Cancer Institute, Bethesda, Maryland, USA.
Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Mercedes Ricote
- Department of Medicine, Division of Endocrinology and Metabolism, and
Department of Cellular and Molecular Medicine, UCSD, La Jolla, California, USA.
St. Vincent’s Institute of Medical Research, Department of Medicine, The University of Melbourne, Fitzroy, Victoria, Australia.
Department of Bioengineering, UCSD, La Jolla, California, USA.
Baker Heart Institute, Cellular and Molecular Medicine Laboratory, St. Kilda, Victoria, Australia.
Laboratory of Metabolism, National Cancer Institute, Bethesda, Maryland, USA.
Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| |
Collapse
|
248
|
Engdahl R, Monroy MA, Daly JM. 15-Deoxy-Delta12,14-prostaglandin J2 (15d-PGJ2) mediates repression of TNF-alpha by decreasing levels of acetylated histone H3 and H4 at its promoter. Biochem Biophys Res Commun 2007; 359:88-93. [PMID: 17532302 PMCID: PMC2584358 DOI: 10.1016/j.bbrc.2007.05.057] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2007] [Accepted: 05/09/2007] [Indexed: 10/23/2022]
Abstract
Prostaglandin metabolite 15-Deoxy-Delta(12,14)-prostaglandin J2 (15d-PGJ2) is known to inhibit a number of pro-inflammatory cytokines as well as being a ligand for nuclear receptor PPARgamma. We investigated the ability of 15d-PGJ2 to inhibit TNF-alpha gene expression through mechanisms that involve histone modification. Pretreatment with 15d-PGJ2 (10 microM) inhibited LPS-stimulated TNF-alpha mRNA in THP-1 monocytes or PMA-differentiated cells to nearly basal levels. A specific PPARgamma ligand, GW1929, failed to inhibit LPS-induced TNF-alpha mRNA expression nor did a PPARgamma antagonist, GW9662, alter the repression of TNF-alpha mRNA in LPS-stimulated cells pretreated with 15d-PGJ2 suggesting a PPARgamma-independent inhibition of TNF-alpha mRNA in THP-1 cells. Transfection studies with a reporter construct and subsequent treatment with 15d-PGJ2 demonstrated a dose-dependent inhibition of the TNF-alpha promoter. Additional studies demonstrated that inhibition of histone deacetylases with trichostatin A (TSA) or overexpression of histone acetyltransferase CBP could overcome 15d-PGJ2-mediated repression of the TNF-alpha promoter, suggesting that an important mechanism whereby 15d-PGJ2 suppresses a cytokine is through factors that regulate histone modifications. To examine the endogenous TNF-alpha promoter, chromatin immunoprecipitations (ChIP) were performed. ChIP assays demonstrated that LPS stimulation induced an increase in histone H3 and H4 acetylation at the TNF-alpha promoter, which was reduced in cells pretreated with 15d-PGJ2. These results highlight the ability of acetylation and deacetylation factors to affect the TNF-alpha promoter and demonstrate that an additional important mechanism whereby 15d-PGJ2 mediates TNF-alpha transcriptional repression by altering levels of acetylated histone H3 and H4 at its promoter.
Collapse
Affiliation(s)
- Ryan Engdahl
- Temple University School of Medicine, Department of Surgery, 3400 North Broad Street, Philadelphia, PA 19140, USA.
| | | | | |
Collapse
|
249
|
Lebovic DI, Mwenda JM, Chai DC, Mueller MD, Santi A, Fisseha S, D'Hooghe T. PPAR-gamma receptor ligand induces regression of endometrial explants in baboons: a prospective, randomized, placebo- and drug-controlled study. Fertil Steril 2007; 88:1108-19. [PMID: 17498714 PMCID: PMC2062531 DOI: 10.1016/j.fertnstert.2006.12.072] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2006] [Revised: 12/28/2006] [Accepted: 12/28/2006] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To determine the effects of a thiazolidinedione (TZD) agonist of peroxisome proliferator-activated receptor (PPAR)-gamma, rosiglitazone, in a baboon model of established endometriosis. DESIGN Prospective, randomized, placebo-controlled study. SETTING Experimental surgery laboratory at the Institute of Primate Research in Nairobi, Kenya. ANIMAL(S) Endometriosis was induced using intrapelvic injection of eutopic menstrual endometrium in 12 female baboons with a normal pelvis that had undergone at least one menstrual cycle since the time of captivity. INTERVENTION(S) Induction of endometriosis by laparoscopy was performed in 12 baboons with a normal pelvis. Endometrial tissue was extracted from each baboon by curettage, and a standard amount of endometrium was then seeded onto several peritoneal sites. About 34-68 days after the induction of laparoscopy, a pretreatment laparoscopy (baseline disease assessment) was performed in the baboons to record the extent of endometriotic lesions. The 12 baboons were randomized into three groups and treated from the day after the staging laparoscopy for a total duration of 30 days. They received phosphate-buffered saline tablets (n = 4, placebo control; placebo tablets once a day by mouth for 30 days), GnRH-antagonists (n = 4, active control; ganirelix acetate 125 microg/day for 30 days), or rosiglitazone (n = 4, test drug, 2 mg by mouth each day for 30 days). A third and final laparoscopy on day 30 after the start of treatment was performed to record the extent of endometriosis. The type of lesion (typical, red, white, or suspicious) was recorded. Biopsies were obtained to confirm the histological presence of endometriosis. MAIN OUTCOME MEASURE(S) A videolaparoscopy was performed 30 days after treatment to document the number and surface area of endometriotic lesions as well as to calculate the revised American Society for Reproductive Medicine score and stage. RESULT(S) The surface area of endometriotic lesions was statistically significantly lower in rosiglitazone-treated baboons when compared with the placebo group. Baboons treated with rosiglitazone or ganirelix had a greater negative relative change in surface area of peritoneal endometriotic lesions than controls. The overall weighted appearance of the lesion types suggests that rosiglitazone may deter the development of newer endometriotic lesions. CONCLUSION(S) A PPAR-gamma ligand, rosiglitazone, effectively diminishes the burden of endometriosis disease in a baboon endometriosis model. This animal model holds promise that a TZD drug may be helpful in women with endometriosis.
Collapse
Affiliation(s)
- Dan I Lebovic
- Reproductive Endocrinology Division, Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan 48109, USA.
| | | | | | | | | | | | | |
Collapse
|
250
|
Hall JM, McDonnell DP. The molecular mechanisms underlying the proinflammatory actions of thiazolidinediones in human macrophages. Mol Endocrinol 2007; 21:1756-68. [PMID: 17488971 DOI: 10.1210/me.2007-0060] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
It is hypothesized that the antiinflammatory actions of peroxisome proliferator-activated receptors (PPARs) may explain the protective effect of these receptors in diabetes, atherosclerosis, cancer, and other inflammatory diseases. However, emerging evidence for proinflammatory activities of activated PPARs is concerning in light of new studies that associate PPAR modulators with an increased incidence of both cardiovascular events in humans and the sporadic formation of tumors in rodents. In an attempt to define the role of each PPAR subtype in inflammation, we made the unexpected observation that human PPARdelta is a positive regulator of inflammatory responses in both monocytes and macrophages. Notably, TNFalpha-stimulated cells administered PPARdelta agonists express and secrete elevated levels of inflammatory cytokines. Most surprising, however, was the finding that thiazolidinediones (TZDs) and other known PPARgamma ligands display different degrees of proinflammatory activities in a PPARgamma- and PPARalpha-independent manner via their ability to augment PPARdelta signaling. A series of mechanistic studies revealed that TZDs, at clinically relevant concentrations, bind and activate the transcriptional activity of PPARdelta. Collectively, these studies suggest that the observed proinflammatory and potentially deleterious effects of PPARgamma ligands may be mediated through an off-target effect on PPARdelta. These studies highlight the need for PPAR modulators with increased receptor subtype specificity. Furthermore, they suggest that differences in systemic exposure and consequently in the activation of PPARgamma and PPARdelta may explain why TZDs can exhibit both inflammatory and antiinflammatory activities in humans.
Collapse
Affiliation(s)
- Julie M Hall
- Duke University Medical Center, Department of Pharmacology and Cancer Biology, Durham, North Carolina 27710, USA
| | | |
Collapse
|