151
|
Shibata N, Kawaguchi-Niida M, Yamamoto T, Toi S, Hirano A, Kobayashi M. Effects of the PPARgamma activator pioglitazone on p38 MAP kinase and IkappaBalpha in the spinal cord of a transgenic mouse model of amyotrophic lateral sclerosis. Neuropathology 2008; 28:387-98. [PMID: 18312546 DOI: 10.1111/j.1440-1789.2008.00890.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Emerging evidence suggests the involvement of programmed cell death and inflammation in amyotrophic lateral sclerosis (ALS). To assess molecular pathological effects of the anti-inflammatory peroxisome proliferator-activated receptor-gamma (PPARgamma) agonist pioglitazone in ALS, we verified changes in the population of neurons, astrocytes, and microglia in the ventral horns of spinal cord lumbar segments from the pioglitazone-treated and non-treated groups of mice carrying a transgene for G93A mutant human superoxide dismutase-1 (SOD1) (ALS mice) and non-transgenic littermates (control mice), performed immunohistochemical and immunoblot analyses of PPARgamma, active form of phosphorylated p38 mitogen-activated protein kinase (p-p38) and inhibitor of nuclear factor-kappaB (NF-kappaB)-alpha (IkappaBalpha) in the spinal cords, and compared the results between the different groups. Image analysis revealed that optical density of NeuN-immunoreactive neurons was significantly lower in the non-treated groups of presymptomatic and advanced ALS mice than in the non-treated groups of age-matched control mice and was recovered with pioglitazone treatment, and that optical densities of GFAP-immunoreactive astrocytes and Iba1-immunoreactive microglia were significantly higher in the non-treated group of advanced ALS mice than in the non-treated group of control mice and were recovered with pioglitazone treatment. Immunohistochemical analysis demonstrated that immunoreactivities for PPARgamma and p-p38 were mainly localized in neurons, and that IkappaBalpha immunoreactivity was mainly localized in astrocytes and microglia. Immunoblot analysis showed that pioglitazone treatment resulted in no significant change in nuclear PPARgamma-immunoreactive density, a significant decrease in cytosolic p-p38-immunoreactive density, and a significant increase in cytosolic IkappaBalpha-immunoreactive density. Our results suggest that pioglitazone protects motor neurons against p38-mediated neuronal death and NF-kappaB-mediated glial inflammation via a PPARgamma-independent mechanism.
Collapse
Affiliation(s)
- Noriyuki Shibata
- Department of Pathology, Tokyo Woman's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, Japan.
| | | | | | | | | | | |
Collapse
|
152
|
Jiang Q, Heneka M, Landreth GE. The role of peroxisome proliferator-activated receptor-gamma (PPARgamma) in Alzheimer's disease: therapeutic implications. CNS Drugs 2008; 22:1-14. [PMID: 18072811 DOI: 10.2165/00023210-200822010-00001] [Citation(s) in RCA: 131] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Alzheimer's disease is a complex neurodegenerative disorder, with aging, genetic and environmental factors contributing to its development and progression. The complexity of Alzheimer's disease presents substantial challenges for the development of new therapeutic agents. Alzheimer's disease is typified by pathological depositions of beta-amyloid peptides and neurofibrillary tangles within the diseased brain. It has also been demonstrated to be associated with a significant microglia-mediated inflammatory component, dysregulated lipid homeostasis and regional deficits in glucose metabolism within the brain. The peroxisome proliferator-activated receptor-gamma (PPARgamma) is a prototypical ligand-activated nuclear receptor that coordinates lipid, glucose and energy metabolism, and is found in elevated levels in the brains of individuals with Alzheimer's disease. A recently appreciated physiological function of this type of receptor is its ability to modulate inflammatory responses. In animal models of Alzheimer's disease, PPARgamma agonist treatment results in the reduction of amyloid plaque burden, reduced inflammation and reversal of disease-related behavioural impairment. In a recent phase II clinical trial, the use of the PPARgamma agonist rosiglitazone was associated with improved cognition and memory in patients with mild to moderate Alzheimer's disease. Thus, PPARgamma may act to modulate multiple pathophysiological mechanisms that contribute to Alzheimer's disease, and represents an attractive therapeutic target for the treatment of the disease.
Collapse
Affiliation(s)
- Qingguang Jiang
- Department of Neurosciences, Alzheimer Research Laboratory, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | | | | |
Collapse
|
153
|
Ringseis R, Gahler S, Eder K. Conjugated linoleic acid isomers inhibit platelet-derived growth factor-induced NF-kappaB transactivation and collagen formation in human vascular smooth muscle cells. Eur J Nutr 2008; 47:59-67. [PMID: 18264811 DOI: 10.1007/s00394-008-0697-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2007] [Accepted: 01/22/2008] [Indexed: 10/22/2022]
Abstract
BACKGROUND Atherosclerosis is characterized by extensive thickening of the arterial intima partially resulting from deposition of collagen by vascular smooth muscle cells (SMCs). Polyunsaturated fatty acids stimulate collagen formation through NF-kappaB activation. AIM OF THE STUDY The present study aimed to explore the effect of conjugated linoleic acids (CLAs) which are known to inhibit NF-kappaB activation on collagen formation by SMCs. METHODS Vascular SMCs were cultured with 50 micromol/l of CLA isomers (c9t11-CLA, t10c12-CLA) or linoleic acid (LA) and analysed for collagen formation and NF-kappaB p50 transactivation. RESULTS Treatment with CLA isomers but not LA significantly reduced PDGF-stimulated [(3)H] proline incorporation into cell layer protein of SMCs without altering cell proliferation. Simultaneous treatment with the PPARgamma inhibitor T0070907 abrogated this effect. Treatment of SMCs with c9t11-CLA and t10c12-CLA significantly reduced PDGF-induced NF-kappaB p50 activation. CONCLUSIONS CLA isomers inhibit PDGF-stimulated collagen production by vascular SMCs, which is considered to be a hallmark of atherosclerosis, in a PPARgamma-dependent manner. Whether inhibition of the NF-kappaB-pathway is of significance for the reduction of collagen formation by CLA isomers needs further investigation.
Collapse
Affiliation(s)
- Robert Ringseis
- Institut für Agrar- und Ernährungswissenschaften, Martin-Luther-Universität, Halle-Wittenberg, Emil-Abderhalden-Strasse 26, 06108, Halle/Saale, Germany.
| | | | | |
Collapse
|
154
|
Gerry JM, Pascual G. Narrowing in on Cardiovascular Disease: The Atheroprotective Role of Peroxisome Proliferator–Activated Receptor γ. Trends Cardiovasc Med 2008; 18:39-44. [DOI: 10.1016/j.tcm.2007.12.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2007] [Revised: 12/02/2007] [Accepted: 12/04/2007] [Indexed: 02/02/2023]
|
155
|
Kuboki S, Shin T, Huber N, Eismann T, Galloway E, Schuster R, Blanchard J, Zingarelli B, Lentsch AB. Peroxisome proliferator-activated receptor-gamma protects against hepatic ischemia/reperfusion injury in mice. Hepatology 2008; 47:215-24. [PMID: 18085707 DOI: 10.1002/hep.21963] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
UNLABELLED The function of peroxisome proliferator-activated receptor-gamma (PPARgamma) in hepatic inflammation and injury is unclear. In this study, we sought to determine the role of PPARgamma in hepatic ischemia/reperfusion injury in mice. Male mice were subjected to 90 minutes of partial hepatic ischemia followed by up to 8 hours of reperfusion. PPARgamma was found to be constitutively activated in hepatocytes but not in nonparenchymal cells. Upon induction of ischemia, hepatic PPARgamma activation rapidly decreased and remained suppressed throughout the 8-hour reperfusion period. This reduced activation was not a result of decreased protein availability as hepatic nuclear PPARgamma, retinoid X receptor-alpha (RXRalpha), and PPARgamma/RXRalpha heterodimer expression was maintained. Accompanying the decrease in PPARgamma activation was a decrease in the expression of the natural ligand 15-deoxy-Delta(12,14)-prostaglandin J(2). This was associated with reduced interaction of PPARgamma and the coactivator, p300. To determine whether PPARgamma activation is hepatoprotective during hepatic ischemia/reperfusion injury, mice were treated with the PPARgamma agonists, rosiglitazone and connecting peptide. These treatments increased PPARgamma activation and reduced liver injury compared to untreated mice. Furthermore, PPARgamma-deficient mice had more liver injury after ischemia/reperfusion than their wild-type counterparts. CONCLUSION These data suggest that PPARgamma is an important endogenous regulator of, and potential therapeutic target for, ischemic liver injury.
Collapse
Affiliation(s)
- Satoshi Kuboki
- Laboratory of Trauma, Sepsis & Inflammation Research, Department of Surgery, University of Cincinnati, Cincinnati, OH 45267-0558, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
156
|
Authier H, Cassaing S, Coste A, Balard P, Gales A, Berry A, Bans V, Bessières MH, Pipy B. Interleukin-13 primes iNO synthase expression induced by LPS in mouse peritoneal macrophages. Mol Immunol 2008; 45:235-43. [PMID: 17568676 DOI: 10.1016/j.molimm.2007.04.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2007] [Revised: 04/02/2007] [Accepted: 04/06/2007] [Indexed: 01/22/2023]
Abstract
Th2 cytokines such as interleukin-13 (IL-13) have both, stimulatory and inhibitory effects on effector functions of macrophages. Reactive nitrogen species are classically induced in Th1 cytokines and/or lipopolysaccharides (LPS) activated macrophages and this response is inhibited by IL-13. In contrast, IL-13 primes macrophages to produce NO in response to LPS when IL-13 treatment happens prior to LPS exposure. This mechanism occurs through a complex signalling pathway, which involves the scavenger receptor CD36, the LPS receptor CD14 and the nuclear receptor PPARgamma. The enhancement of NO production is the consequence of iNOS induction at mRNA and protein levels. The increase of the NO production induced by LPS in IL-13 pre-treated macrophages is found to potentiate the inhibition of Toxoplasma gondii intracellular replication. These results reveal a novel IL-13 signalling pathway that primes the antimicrobial activity of macrophages induced by LPS caused by overexpression of the iNOS-NO axis.
Collapse
Affiliation(s)
- Hélène Authier
- Laboratoire des macrophages, Médiateurs de l'Inflammation et Interactions Cellulaires, Université Paul Sabatier Toulouse III, EA2405, INSERM IFR31 BP84225, 31432 Toulouse, Cedex 4, France
| | | | | | | | | | | | | | | | | |
Collapse
|
157
|
Uchida K, Shibata T. 15-Deoxy-Delta(12,14)-prostaglandin J2: an electrophilic trigger of cellular responses. Chem Res Toxicol 2007; 21:138-44. [PMID: 18052108 DOI: 10.1021/tx700177j] [Citation(s) in RCA: 125] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Electrophilic molecules are endogenously generated and are causally involved in many pathophysiological effects. Prostaglandin D (20 (PGD (2)), a major cyclooxygenase product in a variety of tissues, readily undergoes dehydration to yield the cyclopentenone-type PGs of the J (2)-series such as 15-deoxy-Delta (12,14)-PGJ (2) (15d-PGJ (2)). 15d-PGJ (2) is an electrophile, which can covalently react via the Michael addition reaction with nucleophiles, such as the free sulfhydryls of glutathione and cysteine residues in cellular proteins that play an important role in the control of the redox cell-signaling pathways. Covalent binding of 15d-PGJ (2) to cellular proteins may be one of the mechanisms by which 15d-PGJ (2) induces a cellular response involved in most of the pathophysiological effects associated with inflammation. In the present perspective, we provide a comprehensive summary of 15d-PGJ (2) as an electrophilic mediator of cellular responses.
Collapse
Affiliation(s)
- Koji Uchida
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan.
| | | |
Collapse
|
158
|
Kaplan J, Cook JA, O'Connor M, Zingarelli B. PEROXISOME PROLIFERATOR-ACTIVATED RECEPTOR γ IS REQUIRED FOR THE INHIBITORY EFFECT OF CIGLITAZONE BUT NOT 15-DEOXY-Δ12,14-PROSTAGLANDIN J2 ON THE NFκB PATHWAY IN HUMAN ENDOTHELIAL CELLS. Shock 2007; 28:722-726. [PMID: 17621259 DOI: 10.1097/shk.0b013e318055683a] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Peroxisome proliferator-activated receptor gamma (PPARgamma) is a ligand-activated nuclear receptor with effects on inflammation, atherosclerosis, and apoptosis. The endogenous PPARgamma ligand, 15-deoxy-Delta12,14-PGJ2 (15d-PGJ2), and the synthetic ligand, ciglitazone, have anti-inflammatory properties in endothelial cells. In addition to PPARgamma-dependent effects on the anti-inflammatory process, it has been proposed that PPARgamma ligands may also inhibit the nuclear transcription factor kappaB (NFkappaB) pathway in a PPARgamma-independent manner. The purpose of this study was to compare the effects of 15d-PGJ2 and ciglitazone on the cytokine-induced activation of the NFkappaB pathway. Human umbilical vein endothelial cells (HUVECs) were transiently transfected with NFkappaB-luciferase or PPARgamma elements-luciferase reporter constructs for 48 h. The HUVECs were pretreated with 15d-PGJ2 or ciglitazone (30 microM) for 1 h, followed by a 4-h stimulation with tumor necrosis factor alpha (100 U/mL). Luciferase assay was performed to determine reporter activity. Additionally, HUVECs were transiently transfected with a dominant-negative mutant, which retains ligand and DNA binding but exhibits markedly reduced transactivation. Stimulation of HUVEC with tumor necrosis factor alpha increased NFkappaB activation while decreasing PPARgamma activity. Overexpression of a dominant-negative PPARgamma mutant prevented the inhibitory effect of ciglitazone on cytokine-induced NFkappaB activation in transfected human endothelial cells. Conversely, 15d-PGJ2 inhibited the cytokine-induced NFkappaB activation even in the absence of PPARgamma. Our data suggest that 15d-PGJ2 exerts direct inhibitory effects on the NFkappaB pathway through a PPARgamma-independent mechanism. On the contrary, the inhibitory effect of ciglitazone on the NFkappaB pathway seems to require PPARgamma activation.
Collapse
Affiliation(s)
- Jennifer Kaplan
- Cincinnati Children's Hospital Medical Center, 3229 Burnet Avenue, Cincinnati, OH 45229, USA
| | | | | | | |
Collapse
|
159
|
Peebles KA, Lee JM, Mao JT, Hazra S, Reckamp KL, Krysan K, Dohadwala M, Heinrich EL, Walser TC, Cui X, Baratelli FE, Garon E, Sharma S, Dubinett SM. Inflammation and lung carcinogenesis: applying findings in prevention and treatment. Expert Rev Anticancer Ther 2007; 7:1405-21. [PMID: 17944566 DOI: 10.1586/14737140.7.10.1405] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Lung carcinogenesis is a complex process requiring the acquisition of genetic mutations that confer the malignant phenotype as well as epigenetic alterations that may be manipulated in the course of therapy. Inflammatory signals in the lung cancer microenvironment can promote apoptosis resistance, proliferation, invasion, metastasis, and secretion of proangiogenic and immunosuppressive factors. Here, we discuss several prototypical inflammatory mediators controlling the malignant phenotype in lung cancer. Investigation into the detailed molecular mechanisms underlying the tumor-promoting effects of inflammation in lung cancer has revealed novel potential drug targets. Cytokines, growth factors and small-molecule inflammatory mediators released in the developing tumor microenvironment pave the way for epithelial-mesenchymal transition, the shift from a polarized, epithelial phenotype to a highly motile mesenchymal phenotype that becomes dysregulated during tumor invasion. Inflammatory mediators within the tumor microenvironment are derived from neoplastic cells as well as stromal and inflammatory cells; thus, lung cancer develops in a host environment in which the deregulated inflammatory response promotes tumor progression. Inflammation-related metabolic and catabolic enzymes (prostaglandin E(2) synthase, prostaglandin I(2) synthase and 15-hydroxyprostaglandin dehydrogenase), cell-surface receptors (E-type prostaglandin receptors) and transcription factors (ZEB1, SNAIL, PPARs, STATs and NF-kappaB) are differentially expressed in lung cancer cells compared with normal lung epithelial cells and, thus, may contribute to tumor initiation and progression. These newly discovered molecular mechanisms in the pathogenesis of lung cancer provide novel opportunities for targeted therapy and prevention in lung cancer.
Collapse
Affiliation(s)
- Katherine A Peebles
- David Geffen School of Medicine at UCLA, Division of Pulmonary & Critical Care Medicine & Hospitalists, Department of Medicine, UCLA Lung Cancer Research Program, CA, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
160
|
Zingarelli B, Hake PW, Mangeshkar P, O'Connor M, Burroughs TJ, Piraino G, Denenberg A, Wong HR. DIVERSE CARDIOPROTECTIVE SIGNALING MECHANISMS OF PEROXISOME PROLIFERATOR-ACTIVATED RECEPTOR-γ LIGANDS, 15-DEOXY-Δ12,14-PROSTAGLANDIN J2 AND CIGLITAZONE, IN REPERFUSION INJURY. Shock 2007; 28:554-63. [PMID: 17589386 DOI: 10.1097/shk.0b013e31804f56b9] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Peroxisome proliferator-activated receptor-gamma (PPAR-gamma) is a nuclear receptor that regulates diverse biological functions including inflammation. The PPARgamma ligands have been reported to exert cardioprotective effects and attenuate myocardial reperfusion injury. Here, we examined the molecular mechanisms of their anti-inflammatory effects. Male Wistar rats were subjected to myocardial ischemia and reperfusion and were treated with the PPAR-gamma ligands, 15-deoxy-Delta-prostaglandin J2 (15d-PGJ2) or ciglitazone, or with vehicle only, in the absence or presence of the selective PPAR-gamma antagonist GW-9662. In vehicle-treated rats, myocardial injury was associated with elevated tissue activity of myeloperoxidase, indicating infiltration of neutrophils, and elevated plasma levels of creatine kinase and tumor necrosis factor-alpha. These events were preceded by activation of the nuclear factor-kappaB pathway. The PPAR-gamma DNA binding was also increased in the heart after reperfusion. Treatment with ciglitazone or 15d-PGJ2 reduced myocardial damage and neutrophil infiltration and blunted creatine kinase levels and cytokine production. The beneficial effects of both ligands were associated with enhancement of PPAR-gamma DNA binding and reduction of nuclear factor-kappaB activation. Treatment with 15d-PGJ2, but not ciglitazone, enhanced DNA binding of heat shock factor 1 and upregulated the expression of the cardioprotective heat shock protein 70. Treatment with 15d-PGJ2, but not ciglitazone, also induced a significant increase in nuclear phosphorylation of the prosurvival kinase Akt. The cardioprotection afforded by ciglitazone was attenuated by the PPAR-gamma antagonist GW-9662. In contrast, GW-9662 did not affect the beneficial effects afforded by 15d-PGJ2. Thus, our data suggest that treatment with these chemically unrelated PPAR-gamma ligands results in diverse anti-inflammatory mechanisms.
Collapse
Affiliation(s)
- Basilia Zingarelli
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, The University of Cincinnati College of Medicine, Cincinnati, Ohio 45229, USA.
| | | | | | | | | | | | | | | |
Collapse
|
161
|
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]
|
162
|
McIntyre RS, Soczynska JK, Woldeyohannes HO, Lewis GF, Leiter LA, MacQueen GM, Miranda A, Fulgosi D, Konarski JZ, Kennedy SH. Thiazolidinediones: novel treatments for cognitive deficits in mood disorders? Expert Opin Pharmacother 2007; 8:1615-28. [PMID: 17685880 DOI: 10.1517/14656566.8.11.1615] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The aim of this review is to provide a rationale for evaluating thiazolidinediones (TZDs) as putative treatments for cognitive deficits in individuals with mood disorders. A MedLine search of all English-language articles published between January 1966 and August 2006 was conducted. The search terms were: the non-proprietary names of TZDs (e.g., rosiglitazone and pioglitazone), peroxisome proliferator-activated receptor, cognition, neuroprotection, inflammation, oxidative stress, cellular metabolism and excitotoxicity cross-referenced with the individual names of mood (e.g., major depressive disorder and bipolar disorder) and dementing disorders (e.g., Alzheimer's disease) as defined in the Diagnostic and Statistical Manual of Mental Disorders third edition, revised/fourth edition, text revision (DSM-III-R/IV-TR). The search was augmented with a manual review of article reference lists. Articles selected for review were based on adequacy of sample size, the use of standardized experimental procedures, validated assessment measures and overall manuscript quality. Contemporary pathophysiologic models of mood disorders emphasize alterations in neuronal plasticity, metabolism and cytoarchitecture with associated regional abnormalities in neuronal (and glial) density and morphology. These abnormalities are hypothesized to subserve cognitive deficits and other clinical features of mood disorders. TZDs may attenuate, abrogate and/or reverse the neurotoxic effects of depressive illness by means of disparate mechanisms, notably insulin signaling, anti-inflammation, glucocorticoid activity and cellular metabolism. Extant data provide the basis for formulating a hypothesis that TZDs may be salutary for cognitive deficits and several aspects of somatic health (e.g., cardiovascular disease) associated with mood disorders.
Collapse
Affiliation(s)
- Roger S McIntyre
- University of Toronto, Department of Psychiatry, Toronto, ON, Canada.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
163
|
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
|
164
|
Majai G, Sarang Z, Csomós K, Zahuczky G, Fésüs L. PPARgamma-dependent regulation of human macrophages in phagocytosis of apoptotic cells. Eur J Immunol 2007; 37:1343-54. [PMID: 17407194 DOI: 10.1002/eji.200636398] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Macrophages acquire their capacity for efficient phagocytosis of apoptotic cells during their differentiation from monocytes. The peroxisome proliferator-activated receptor gamma (PPARgamma) is highly up-regulated during this maturation program. We report that addition of PPARgamma antagonist during differentiation of human monocytes to macrophages significantly reduced the capacity of macrophages to engulf apoptotic neutrophils, but did not influence phagocytosis of opsonized bacteria. Macrophage-specific deletion of PPARgamma in mice also resulted in decreased uptake of apoptotic cells. The antagonist acted in a dose-dependent manner during the differentiation of human macrophages and could also reverse the previously observed augmentation of phagocytosis by glucocorticoids. Blocking activation of PPARgamma led to down-regulation of molecular elements (CD36, AXL, TG2 and PTX3) of the engulfment process. Inhibition of PPARgamma-dependent gene expression did not block the anti-inflammatory effect of apoptotic neutrophils or synthetic glucocorticoid, but significantly decreased production of IL-10 induced by LPS. Our results suggest that during differentiation of macrophages natural ligands of PPARgamma are formed, regulating the expression of genes responsible for effective clearance of apoptotic cells and macrophage-mediated inflammatory responses.
Collapse
Affiliation(s)
- Gyöngyike Majai
- Department of Biochemistry and Molecular Biology, Signalling and Apoptosis Research Group of the Hungarian Academy of Sciences, Research Centre for Molecular Medicine, University of Debrecen, Medical and Health Science Centre, Debrecen, Hungary
| | | | | | | | | |
Collapse
|
165
|
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
|
166
|
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
|
167
|
Yi JH, Park SW, Kapadia R, Vemuganti R. Role of transcription factors in mediating post-ischemic cerebral inflammation and brain damage. Neurochem Int 2007; 50:1014-27. [PMID: 17532542 PMCID: PMC2040388 DOI: 10.1016/j.neuint.2007.04.019] [Citation(s) in RCA: 169] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2007] [Revised: 04/16/2007] [Accepted: 04/18/2007] [Indexed: 12/20/2022]
Abstract
Inflammation is a known precipitator of neuronal death after cerebral ischemia. The mechanisms that promote or curtail the start and spread of inflammation in brain are still being debated. By virtue of their capability to modulate gene expression, several transcription factors induced in the ischemic brain can modulate the post-ischemic inflammation. While the induction of transcription factors such as IRF1, NF-kappaB, ATF-2, STAT3, Egr1 and C/EBPbeta is thought to promote post-ischemic inflammation, activation of transcription factors such as HIF-1, CREB, c-fos, PPARalpha, PPARgamma and p53 is thought to prevent post-ischemic inflammation and neuronal damage. Of these, PPARgamma which is a ligand-activated transcription factor was recently shown to prevent inflammatory gene expression in several animal models CNS disorders. This review article discusses some of the molecular mechanisms of PPARgamma induction by its agonists following focal cerebral ischemia.
Collapse
Affiliation(s)
- Jae-Hyuk Yi
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Seung-Won Park
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
- Department of Neurological Surgery (SP), Chung-Ang University, Seoul, Korea
| | - Ramya Kapadia
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
- Neuroscience Training Program, University of Wisconsin, Madison, WI, USA
| | - Raghu Vemuganti
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
- Neuroscience Training Program, University of Wisconsin, Madison, WI, USA
- Cardiovascular Research Center, University of Wisconsin, Madison, WI, USA
- Regenerative Medicine Program, University of Wisconsin, Madison, WI, USA
| |
Collapse
|
168
|
Ponferrada A, Caso JR, Alou L, Colón A, Sevillano D, Moro MA, Lizasoain I, Menchén P, Gómez-Lus ML, Lorenzo P, Cos E, Leza JC, Menchén L. The role of PPARgamma on restoration of colonic homeostasis after experimental stress-induced inflammation and dysfunction. Gastroenterology 2007; 132:1791-803. [PMID: 17484875 DOI: 10.1053/j.gastro.2007.02.032] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2006] [Accepted: 01/18/2007] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Psychological stress has been implicated in the clinical course of several gastrointestinal diseases, but the mechanisms implicated and the effects of stress on the normal colon are not yet fully understood. METHODS Male Wistar rats were exposed to various immobilization periods as a stress paradigm. Colon was processed to assess myeloperoxidase activity, nitric oxide synthase 2, cyclooxygenase 2, and peroxisome proliferator-activated receptor gamma (PPARgamma) expression and production of prostaglandins. Colonic permeability, bacterial translocation, tight junctions ultrastructure, and immunoglobulin (Ig) A levels were also evaluated. RESULTS Exposure to acute (6 hours) immobilization stress produced an increase in myeloperoxidase activity and nitric oxide synthase 2 and cyclooxygenase 2 expression. All these parameters remained increased after 5 days of repeated stress exposure, showing a trend to normalize after 10 days. Levels of the anti-inflammatory eicosanoid 15-deoxy-Delta(12,14)-prostaglandin J(2) (15d-PGJ(2)) and expression of PPARgamma run parallel with these changes. Colonic epithelial barrier was altered after stress exposure, and a significant decrease in colonic IgA levels after acute stress exposure was observed. Pretreatment with PPARgamma agonists 15d-PGJ(2) and rosiglitazone prevented colonic inflammation and barrier dysfunction as well as the decrease of IgA production induced after acute stress; PPARgamma specific antagonist T0070907 reverted these effects. CONCLUSIONS Activation of PPARgamma in rat colon in vivo seems to counteract colonic inflammation and dysfunction induced by stress. On the other hand, PPARgamma ligands may be therapeutically useful in conditions in which inflammation and barrier dysfunction takes place in colon after exposure to stress.
Collapse
Affiliation(s)
- Angel Ponferrada
- Servicio de Aparato Digestivo, Hospital General Universitario Gregorio Marañón, Madrid
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
169
|
Ricote M, Glass CK. PPARs and molecular mechanisms of transrepression. Biochim Biophys Acta Mol Cell Biol Lipids 2007; 1771:926-35. [PMID: 17433773 PMCID: PMC1986735 DOI: 10.1016/j.bbalip.2007.02.013] [Citation(s) in RCA: 413] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2006] [Revised: 02/26/2007] [Accepted: 02/27/2007] [Indexed: 12/21/2022]
Abstract
In the last few years, PPARs have emerged as key regulators of inflammatory and immune responses. However, the mechanistic basis of the anti-inflammatory effects of peroxisome proliferator-activated receptors (PPARs) remains poorly understood. Accumulating evidence suggests that these effects result from inhibition of signal-dependent transcription factors that mediate inflammatory programs of gene activation. Several mechanisms underlying negative regulation of gene expression by PPARs have been described. Recent studies, using siRNA, microarray analysis and macrophage-specific knockout mice, have highlighted PPARs molecular transrepression mechanism in macrophages. Identification of their mechanism of action should help promote the understanding of the physiologic roles that PPARs play in immunity and contribute to the development of new therapeutic agents.
Collapse
Affiliation(s)
- Mercedes Ricote
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain.
| | | |
Collapse
|
170
|
Jones DC, Manning BM, Daynes RA. A role for the peroxisome proliferator-activated receptor α in T-cell physiology and ageing immunobiology. Proc Nutr Soc 2007; 61:363-9. [PMID: 12296295 DOI: 10.1079/pns2002173] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Peroxisome proliferator-activated receptor (PPAR) α represents an important member of the nuclear hormone receptor superfamily that can be activated by a variety of natural fatty acids, some of their metabolites and by commonly-used anti-lipidaemic drugs. We recently demonstrated PPARα expression in T lymphocytes, where it controls the initiation of transcription of T-box expressed in T-cells (T-bet) independent of added agonist. T-bet is an activation-inducible transcription factor regulator of interleukin 2 (suppression) and interferon γ (stimulation) synthesis. A suppressed ability to produce interleukin 2 and an enhanced production of interferon γ occurs in activated T-cells from PPARα-/- mice, as well as in T-cells from wild-type aged animals whose lymphocytes express lowered basal levels of PPARα. The dysregulated expression and/or function of cytokines, glucocorticoids or leptin that occurs with advanced age could all be responsible for the reduced expression of PPARα. Dietary supplementation of aged mice with vitamin E, or supplementation with known agonists of PPARα, was associated with elevation of lymphocyte expression of this nuclear hormone receptor, restoration of control over T-bet expression and elimination of the dysregulated production of interleukin 2 and interferon γ following lymphocyte activation. Interleukin 2 and interferon γ play very important roles in the initiation and/or regulation of immune, inflammatory and autoimmune disease states. Thus, the mechanisms that control the timing, magnitude and duration of specific cytokine production by activated T lymphocytes need clarification before appropriate nutritional or therapeutic strategies can be devised to treat disease conditions where cytokine expression and/or activities are deemed to be dysregulated and responsible.
Collapse
Affiliation(s)
- Dallas C Jones
- Department of Pathology, University of Utah School of Medicine, Salt Lake City 84132, USA
| | | | | |
Collapse
|
171
|
Abstract
Currently, there are no disease-modifying therapies available for Alzheimer's disease (AD). Acetylcholinesterase inhibitors and memantine are licensed for AD and have moderate symptomatic benefits. Epidemiological studies have suggested that NSAIDs, estrogen, HMG-CoA reductase inhibitors (statins) or tocopherol (vitamin E) can prevent AD. However, prospective, randomised studies have not convincingly been able to demonstrate clinical efficacy. Major progress in molecular medicine suggests further drug targets. The metabolism of the amyloid-precursor protein and the aggregation of its Abeta fragment are the focus of current studies. Abeta peptides are produced by the enzymes beta- and gamma-secretase. Inhibition of gamma-secretase has been shown to reduce Abeta production. However, gamma-secretase activity is also involved in other vital physiological pathways. Involvement of gamma-secretase in cell differentiation may preclude complete blockade of gamma-secretase for prolonged times in vivo. Inhibition of beta-secretase seems to be devoid of serious adverse effects according to studies with knockout animals. However, targeting beta-secretase is hampered by the lack of suitable inhibitors to date. Other approaches focus on enzymes that cut inside the Abeta sequence such as alpha-secretase and neprilysin. Stimulation of the expression or activity of alpha-secretase or neprilysin has been shown to enhance Abeta degradation. Furthermore, inhibitors of Abeta aggregation have been described and clinical trials have been initiated. Peroxisome proliferator activated receptor-gamma agonists and selected NSAIDs may be suitable to modulate both Abeta production and inflammatory activation. On the basis of autopsy reports, active immunisation against Abeta in humans seems to have proven its ability to clear amyloid deposits from the brain. However, a first clinical trial with active vaccination against the full length Abeta peptide has been halted because of adverse effects. Further trials with vaccination or passive transfer of antibodies are planned.
Collapse
Affiliation(s)
- Michael Hüll
- Department of Psychiatry and Psychotherapy, University of Freiburg, Hauptstrasse 5, D-79108 Freiburg, Germany.
| | | | | |
Collapse
|
172
|
Lee KS, Park SJ, Kim SR, Min KH, Jin SM, Lee HK, Lee YC. Modulation of airway remodeling and airway inflammation by peroxisome proliferator-activated receptor gamma in a murine model of toluene diisocyanate-induced asthma. THE JOURNAL OF IMMUNOLOGY 2007; 177:5248-57. [PMID: 17015710 DOI: 10.4049/jimmunol.177.8.5248] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Toluene diisocyanate (TDI) is a leading cause of occupational asthma. Although considerable controversy remains regarding its pathogenesis, TDI-induced asthma is an inflammatory disease of the airways characterized by airway remodeling. Peroxisome proliferator-activated receptor gamma (PPARgamma) has been shown to play a critical role in the control of airway inflammatory responses. However, no data are available on the role of PPARgamma in TDI-induced asthma. We have used a mouse model for TDI-induced asthma to determine the effect of PPARgamma agonist, rosiglitazone, or pioglitazone, and PPARgamma on TDI-induced bronchial inflammation and airway remodeling. This study with the TDI-induced model of asthma revealed the following typical pathophysiological features: increased numbers of inflammatory cells of the airways, airway hyperresponsiveness, increased levels of Th2 cytokines (IL-4, IL-5, and IL-13), adhesion molecules (ICAM-1 and VCAM-1), chemokines (RANTES and eotaxin), TGF-beta1, and NF-kappaB in nuclear protein extracts. In addition, the mice exposed to TDI developed features of airway remodeling, including thickening of the peribronchial smooth muscle layer, subepithelial collagen deposition, and increased airway mucus production. Administration of PPARgamma agonists or adenovirus carrying PPARgamma2 cDNA reduced the pathophysiological symptoms of asthma and decreased the increased levels of Th2 cytokines, adhesion molecules, chemokines, TGF-beta1, and NF-kappaB in nuclear protein extracts after TDI inhalation. In addition, inhibition of NF-kappaB activation decreased the increased levels of Th2 cytokines, adhesion molecules, chemokines, and TGF-beta1 after TDI inhalation. These findings demonstrate a protective role of PPARgamma in the pathogenesis of the TDI-induced asthma phenotype.
Collapse
Affiliation(s)
- Kyung Sun Lee
- Department of Internal Medicine, Airway Remodeling Laboratory, Chonbuk National University Medical School, Jeonju, South Korea.
| | | | | | | | | | | | | |
Collapse
|
173
|
Phulwani NK, Feinstein DL, Gavrilyuk V, Akar C, Kielian T. 15-deoxy-Delta12,14-prostaglandin J2 (15d-PGJ2) and ciglitazone modulate Staphylococcus aureus-dependent astrocyte activation primarily through a PPAR-gamma-independent pathway. J Neurochem 2007; 99:1389-1402. [PMID: 17074064 PMCID: PMC2423669 DOI: 10.1111/j.1471-4159.2006.04183.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Brain abscesses arise from a focal parenchymal infection by various pathogens, particularly Staphylococcus aureus. We have shown that astrocytes are activated upon exposure to S. aureus and may contribute to the excessive tissue damage characteristic of brain abscess. Therefore, modulating astrocyte activation may facilitate a reduction in brain abscess severity. Peroxisome proliferator activated receptor-gamma (PPAR-gamma) agonists are potent inhibitors of microglial activation; however, the effects of these compounds on S. aureus-dependent astrocyte activation have not yet been examined. Here, we demonstrate that two chemically distinct PPAR-gamma agonists, 15-deoxy-delta12,14-prostaglandin J2 (15d-PGJ2) and ciglitazone, suppress the production of several pro-inflammatory molecules in S. aureus-stimulated astrocytes including interleukin-1beta and nitric oxide (NO). Interestingly, 15d-PGJ2 attenuated Toll-like receptor 2 (TLR2) and inducible nitric oxide synthase expression, but failed to modulate macrophage inflammatory protein-2 (MIP-2/CXCL2) production, suggesting that 15d-PGJ2 is not a global inhibitor of astrocyte activation. Another novel finding of this study was the fact that both 15d-PGJ2 and ciglitazone were capable of attenuating pre-existing astrocyte activation, indicating their potential benefit in a therapeutic setting. Importantly, 15d-PGJ2 and ciglitazone were still capable of inhibiting S. aureus-induced pro-inflammatory mediator release in PPAR-gamma-deficient astrocytes, supporting PPAR-gamma-independent effects of these compounds. Collectively, these results suggest that 15d-PGJ2 and ciglitazone exert their anti-inflammatory actions on astrocytes primarily independent of the PPAR-gamma pathway.
Collapse
Affiliation(s)
- Nirmal K. Phulwani
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Douglas L. Feinstein
- Department of Anesthesiology, University of Illinois and Jesse Brown Veterans Affairs, Chicago, Illinois, USA
| | - Vitaliy Gavrilyuk
- Department of Anesthesiology, University of Illinois and Jesse Brown Veterans Affairs, Chicago, Illinois, USA
| | - Candan Akar
- Department of Anesthesiology, University of Illinois and Jesse Brown Veterans Affairs, Chicago, Illinois, USA
| | - Tammy Kielian
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| |
Collapse
|
174
|
|
175
|
Abstract
PURPOSE OF REVIEW In diabetes, oxidative stress plays a key role in the pathogenesis of vascular complications; therefore an antioxidant therapy would be of great interest in this disease. RECENT FINDINGS Hyperglycemia directly promotes an endothelial dysfunction--inducing process of overproduction of superoxide at the mitochondrial level. This is the first and key event able to activate all the pathways involved in the development of vascular complications of diabetes. It has recently been shown that statins, angiotensin-converting enzyme inhibitors, angiotensin II type 1 blockers, calcium channel blockers, and thiazolidinediones have a strong intracellular antioxidant activity. SUMMARY Classic antioxidants, such as vitamin E, failed to show beneficial effects on diabetic complications probably because their action is only "symptomatic". The preventive activity against hyperglycemia-induced oxidative stress shown by statins, angiotensin-converting enzyme inhibitors, angiotensin II type 1 blockers, calcium channel blockers, and thiazolidinediones justifies use of these compounds for preventing complications in patients with diabetes, in whom antioxidant defences have been shown to be defective.
Collapse
Affiliation(s)
- Antonio Ceriello
- Warwick Medical School, Clinical Science Research Institute, University Hospital-Walsgrave Campus, University of Warwick, Clifford Bridge Road, Coventry CV2 2DX, UK.
| |
Collapse
|
176
|
Tobin JF, Freedman LP. Nuclear receptors as drug targets in metabolic diseases: new approaches to therapy. Trends Endocrinol Metab 2006; 17:284-90. [PMID: 16870465 DOI: 10.1016/j.tem.2006.07.004] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2006] [Revised: 07/10/2006] [Accepted: 07/12/2006] [Indexed: 10/24/2022]
Abstract
Nuclear receptors represent novel targets for the development of therapeutic agents for the treatment of numerous diseases, including type 2 diabetes, obesity dyslipidemia, atherosclerosis and the metabolic syndrome. There have been many recent advances in the development of new therapeutic agents for a subset of these receptors, including the peroxisome proliferator-activated receptors, the liver X receptors and the farnesoid X receptor. To date, the synthesis of selective modulators that regulate the activity of these receptors has been empirical. However, a detailed understanding of the molecular basis for selective modulation, as well as new insights into the biology of these receptors, might open the door to the rational design of a new generation of therapeutic agents with improved safety and efficacy.
Collapse
Affiliation(s)
- James F Tobin
- Department of Cardiovascular and Metabolic Diseases, Wyeth Research, 200 Cambridge Park Drive, Cambridge, MA 02140, USA.
| | | |
Collapse
|
177
|
Rollins MD, Sudarshan S, Firpo MA, Etherington BH, Hart BJ, Jackson HH, Jackson JD, Emerson LL, Yang DT, Mulvihill SJ, Glasgow RE. Anti-inflammatory effects of PPAR-gamma agonists directly correlate with PPAR-gamma expression during acute pancreatitis. J Gastrointest Surg 2006; 10:1120-30. [PMID: 16966031 DOI: 10.1016/j.gassur.2006.04.027] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2006] [Revised: 04/27/2006] [Accepted: 04/28/2006] [Indexed: 01/31/2023]
Abstract
Peroxisome proliferator-activated receptors (PPARs) are ligand-inducible transcription factors that regulate cellular energy and lipid metabolism. PPAR-gamma agonists also have potent anti-inflammatory properties through down-regulation of early inflammatory response genes. The role of PPAR-gamma in acute pancreatitis has not been adequately examined. In this study, we determined the effect of PPAR-gamma agonists on the severity of pancreatitis and sought to correlate PPAR-gamma expression in pancreatic acinar cells and the severity of acute pancreatitis in vivo. Acute pancreatitis was induced in mice by hyperstimulation with the cholecystokinin analog, cerulein. PPAR-gamma agonists were administered by intraperitoneal injection 15-30 minutes before induction of pancreatitis (pretreatment) or at various times after induction of pancreatitis (treatment). Pancreata and serum were harvested over the course of 24 hours. Serum amylase activity and glucose levels were measured. Pancreata were used for histological evaluation as well as protein and mRNA analysis. Pretreatment of mice with the PPAR-gamma agonists 15-deoxy-Delta12, 14-prostaglandin J(2), or troglitazone significantly reduced the severity of pancreatitis in a dose-dependent manner. This reduction was indicated by reduced serum amylase activity and histological damage (leukocyte infiltration, vacuolization, and necrosis). Although cerulein decreased PPAR-gamma expression in the pancreas, pretreatment with agonists maintained PPAR-gamma expression early in acute pancreatitis. The expression of PPAR-gamma inversely correlated with pancreatitis severity and expression of the proinflammatory cytokines, interleukin-6, and tumor necrosis factor-alpha. Treatment with troglitazone after the induction of pancreatitis reduced serum amylase activity. The results suggest that PPAR-gamma plays a direct role in the inflammatory cascade during the early events of acute pancreatitis. Our data are the first to demonstrate that PPAR-gamma agonists represent a promising therapeutic strategy for acute pancreatitis.
Collapse
Affiliation(s)
- Michael D Rollins
- Department of Surgery, University of Utah School of Medicine, Salt Lake City, Utah 84132, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
178
|
Ou Z, Zhao X, Labiche LA, Strong R, Grotta JC, Herrmann O, Aronowski J. Neuronal expression of peroxisome proliferator-activated receptor-gamma (PPARγ) and 15d-prostaglandin J2—Mediated protection of brain after experimental cerebral ischemia in rat. Brain Res 2006; 1096:196-203. [PMID: 16725118 DOI: 10.1016/j.brainres.2006.04.062] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2006] [Revised: 04/04/2006] [Accepted: 04/10/2006] [Indexed: 12/29/2022]
Abstract
Existing experimental evidence suggests that PPARgamma may play a beneficial role in neuroprotection from various brain pathologies. Here we found that focal cerebral ischemia induced by middle cerebral/common carotid arteries occlusion (MCA/CCAo) induced up-regulation of PPARgamma messenger RNA in the ischemic hemisphere as early as 6 h after the ischemic event. The increased PPARgamma mRNA expression was primarily associated with neurons in the ischemic penumbra, suggesting an important role for PPARgamma in neurons after ischemia. Intraventricular injection of 15d-Delta(12,14)-prostaglandin J(2) (15d-PGJ(2)), a proposed endogenous PPARgamma agonist, into the ischemic rat brains significantly increased the PPARgamma-DNA-binding activity and reduced infarction volume at 24 h after reperfusion. We propose that PPARgamma up-regulation in response to ischemia may contribute to PPARgamma activation in the presence of PPARgamma agonists. Activation of PPARgamma in neurons at an early stage after ischemia may represent a pro-survival mechanism against ischemic injury.
Collapse
Affiliation(s)
- Zhishuo Ou
- University of Texas Health Science Center-Houston, Medical School, Department of Neurology, Stroke Program, Houston, 77030, USA
| | | | | | | | | | | | | |
Collapse
|
179
|
Zhao X, Zhang Y, Strong R, Grotta JC, Aronowski J. 15d-Prostaglandin J2 activates peroxisome proliferator-activated receptor-gamma, promotes expression of catalase, and reduces inflammation, behavioral dysfunction, and neuronal loss after intracerebral hemorrhage in rats. J Cereb Blood Flow Metab 2006; 26:811-20. [PMID: 16208315 DOI: 10.1038/sj.jcbfm.9600233] [Citation(s) in RCA: 187] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Peroxisome proliferator-activated receptor-gamma (PPARgamma) is a transcription factor that regulates the expression of various gene products that are essential in lipid and glucose metabolism, as well as that of the peroxisome-enriched antioxidant enzyme, catalase. Activation of PPARgamma is linked to anti-inflammatory activities and is beneficial for cardiovascular diseases. However, little is known about its role in intracerebral hemorrhage (ICH). 15-Deoxy-Delta(12,14)-prostaglandin J2 (15d-PGJ2) acts as a physiologic agonist for PPARgamma. In this study, we found that injection of 15d-PGJ2 into the locus of striatal hematoma increased PPARgamma-deoxyribonucleic acid (DNA) binding activity and the expression of catalase messenger ribonucleic acid (mRNA) and protein in the perihemorrhagic area. Additionally, 15d-PGJ2 significantly reduced nuclear factor-kappaB (NF-kappaB) activation and prevented neutrophil infiltration measured by myeloperoxidase (MPO) immunoassay, and also reduced cell apoptosis measured by terminal deoxynucleotide transferase dUTP nick-end labeling (TUNEL). In addition, 15d-PGJ2 reduced behavioral dysfunction produced by the ICH. Altogether, our findings indicate that injection of 15d-PGJ2 at the onset of ICH is associated with activation of PPARgamma and elevation of catalase expression, suppression of NF-kappaB activity, and restricted neutrophil infiltration. All these events predicted reduced behavioral deficit and neuronal damage.
Collapse
Affiliation(s)
- Xiurong Zhao
- Stroke Program, Department of Neurology, University of Texas - Houston Medical School, Houston, Texas 77030, USA
| | | | | | | | | |
Collapse
|
180
|
Sung B, Park S, Yu BP, Chung HY. Amelioration of age-related inflammation and oxidative stress by PPARgamma activator: suppression of NF-kappaB by 2,4-thiazolidinedione. Exp Gerontol 2006; 41:590-9. [PMID: 16716549 DOI: 10.1016/j.exger.2006.04.005] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2004] [Revised: 03/31/2006] [Accepted: 04/04/2006] [Indexed: 11/30/2022]
Abstract
Peroxisome proliferator-activated receptors (PPARs) are members of the nuclear hormone receptor superfamily of transcription factors and are key regulators in various pathophysiological processes related to energy metabolism including lipid and carbohydrate metabolism and inflammation. PPARgamma signaling pathways are reported to exert anti-inflammatory effects by inhibition of NF-kappaB. We previously reported that age-related oxidative stress and inflammatory reactions cause reduced PPARgamma during the aging process. In present study, we investigated the action of 2,4-thiazolidinedione (2,4-TZD), a well-known PPARgamma activator, on aging process using kidneys from Fischer 344 rats, young (9-month-old), old (22-month-old) and old-2,4-TZD fed (4 mg/kg for 10 days). The results showed that the 2,4-TZD treatment brought about several major changes, decrease of: (1) age-related oxidative stress; (2) p65 translocation and NF-kappaB binding activity; (3) NF-kappaB-regulated gene expression, inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), and inflammatory mediators such as interleukin-1beta (IL-1beta), IL-6, adhesion molecules, VCAM-1 and P-selectin; and (4) age-related disturbance of the redox-status. Therefore, we concluded that 2,4-TZD exerted significant anti-oxidative and anti-inflammatory effects in aged rats, most likely by its ability to attenuate oxidative stress. We propose that 2,4-TZD or other potent PPARgamma activators may be useful in the therapy against age-related inflammation.
Collapse
Affiliation(s)
- Bokyung Sung
- College of Pharmacy, Pusan National University, Jangjeon-dong, Geumjeong-gu, Busan 609-735, South Korea
| | | | | | | |
Collapse
|
181
|
Grau R, Punzón C, Fresno M, Iñiguez M. Peroxisome-proliferator-activated receptor alpha agonists inhibit cyclo-oxygenase 2 and vascular endothelial growth factor transcriptional activation in human colorectal carcinoma cells via inhibition of activator protein-1. Biochem J 2006; 395:81-8. [PMID: 16343055 PMCID: PMC1409694 DOI: 10.1042/bj20050964] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Recent evidence indicates that PPAR (peroxisome-proliferator-activated receptor) alpha ligands possess anti-inflammatory and antitumoural properties owing to their inhibitory effects on the expression of genes that are involved in the inflammatory response. However, the precise molecular mechanisms underlying these effects are poorly understood. In the present study, we show that tumour promoter PMA-mediated induction of genes that are significantly associated with inflammation, tumour growth and metastasis, such as COX-2 (cyclo-oxygenase 2) and VEGF (vascular endothelial growth factor), is inhibited by PPARalpha ligands in the human colorectal carcinoma cell line SW620. PPARalpha activators LY-171883 and WY-14,643 were able to diminish transcriptional induction of COX-2 and VEGF by inhibiting AP-1 (activator protein-1)-mediated transcriptional activation induced by PMA or by c-Jun overexpression. The actions of these ligands on AP-1 activation and COX-2 and VEGF transcriptional induction were found to be dependent on PPARalpha expression. Our studies demonstrate the existence of a negative cross-talk between the PPARalpha- and AP-1-dependent signalling pathways in these cells. PPARalpha interfered with at least two steps within the pathway leading to AP-1 activation. First, PPARalpha activation impaired AP-1 binding to a consensus DNA sequence. Secondly, PPARalpha ligands inhibited c-Jun transactivating activity. Taken together, these findings provide new insight into the anti-inflammatory and anti-tumoural properties of PPARalpha activation, through the inhibition of the induction of AP-1-dependent genes that are involved in inflammation and tumour progression.
Collapse
Affiliation(s)
- Raquel Grau
- Centro de Biología Molecular “Severo Ochoa”, Departamento de Biología Molecular. Universidad Autónoma de Madrid, Cantoblanco, 28049 Madrid, Spain
| | - Carmen Punzón
- Centro de Biología Molecular “Severo Ochoa”, Departamento de Biología Molecular. Universidad Autónoma de Madrid, Cantoblanco, 28049 Madrid, Spain
| | - Manuel Fresno
- Centro de Biología Molecular “Severo Ochoa”, Departamento de Biología Molecular. Universidad Autónoma de Madrid, Cantoblanco, 28049 Madrid, Spain
| | - Miguel A. Iñiguez
- Centro de Biología Molecular “Severo Ochoa”, Departamento de Biología Molecular. Universidad Autónoma de Madrid, Cantoblanco, 28049 Madrid, Spain
- To whom correspondence should be addressed (email )
| |
Collapse
|
182
|
Coyle AT, Kinsella BT. Synthetic peroxisome proliferator-activated receptor γ agonists rosiglitazone and troglitazone suppress transcription by promoter 3 of the human thromboxane A2 receptor gene in human erythroleukemia cells. Biochem Pharmacol 2006; 71:1308-23. [PMID: 16499875 DOI: 10.1016/j.bcp.2006.01.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2005] [Revised: 01/20/2006] [Accepted: 01/20/2006] [Indexed: 10/25/2022]
Abstract
The human thromboxane (TX)A2 receptor (TP) gene encodes two TP isoforms, TPalpha and TPbeta, that are regulated by distinct promoters designated promoter Prm1 and Prm3, respectively. Previous studies established that 15d-Delta12,14-prostaglandin J2 (15d-PGJ2) selectively inhibits Prm3 activity and TPbeta expression through a peroxisome proliferator-activated receptor (PPAR)gamma mechanism without affecting Prm1 activity or TPalpha expression in human megakaryocytic erythroleukemia (HEL) 92.1.7 cells. Herein, we investigated the effect of synthetic thiazolidinedione (TZD) PPARgamma ligands rosiglitazone and troglitazone on TP gene expression in HEL cells. Like 15d-PGJ2, both TZDs suppressed Prm3 activity, TPbeta mRNA expression and TP-mediated calcium mobilization without affecting Prm1 or TPalpha mRNA expression. However, unlike 15d-PGJ2, both TZDs mediated their PPARgamma-dependent effects through trans-repression of an activator protein-1 (AP-1) element, a site previously found to be critical for basal Prm3 activity. These data provide further evidence for the role of PPARgamma in regulating the human TP gene; they highlight further differences in TPalpha and TPbeta expression/regulation and point to essential differences between natural and synthetic PPARgamma agonists in mediating those effects.
Collapse
MESH Headings
- Cell Line
- Cell Line, Tumor
- Chromans/pharmacology
- Genes, Reporter
- Humans
- Leukemia, Erythroblastic, Acute
- Luciferases
- PPAR gamma/agonists
- Promoter Regions, Genetic/genetics
- Prostaglandin D2/analogs & derivatives
- Prostaglandin D2/pharmacology
- Protein Isoforms/genetics
- Protein Isoforms/metabolism
- RNA, Messenger/metabolism
- Receptors, Thromboxane A2, Prostaglandin H2/genetics
- Receptors, Thromboxane A2, Prostaglandin H2/metabolism
- Retinoid X Receptor alpha
- Rosiglitazone
- Thiazolidinediones/pharmacology
- Transcription Factor AP-1
- Transcription, Genetic
- Troglitazone
Collapse
Affiliation(s)
- Adrian T Coyle
- School of Biomolecular and Biomedical Science, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | | |
Collapse
|
183
|
Kasper LH, Fukuyama T, Biesen MA, Boussouar F, Tong C, de Pauw A, Murray PJ, van Deursen JMA, Brindle PK. Conditional knockout mice reveal distinct functions for the global transcriptional coactivators CBP and p300 in T-cell development. Mol Cell Biol 2006; 26:789-809. [PMID: 16428436 PMCID: PMC1347027 DOI: 10.1128/mcb.26.3.789-809.2006] [Citation(s) in RCA: 168] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The global transcriptional coactivators CREB-binding protein (CBP) and the closely related p300 interact with over 312 proteins, making them among the most heavily connected hubs in the known mammalian protein-protein interactome. It is largely uncertain, however, if these interactions are important in specific cell lineages of adult animals, as homozygous null mutations in either CBP or p300 result in early embryonic lethality in mice. Here we describe a Cre/LoxP conditional p300 null allele (p300flox) that allows for the temporal and tissue-specific inactivation of p300. We used mice carrying p300flox and a CBP conditional knockout allele (CBPflox) in conjunction with an Lck-Cre transgene to delete CBP and p300 starting at the CD4- CD8- double-negative thymocyte stage of T-cell development. Loss of either p300 or CBP led to a decrease in CD4+ CD8+ double-positive thymocytes, but an increase in the percentage of CD8+ single-positive thymocytes seen in CBP mutant mice was not observed in p300 mutants. T cells completely lacking both CBP and p300 did not develop normally and were nonexistent or very rare in the periphery, however. T cells lacking CBP or p300 had reduced tumor necrosis factor alpha gene expression in response to phorbol ester and ionophore, while signal-responsive gene expression in CBP- or p300-deficient macrophages was largely intact. Thus, CBP and p300 each supply a surprising degree of redundant coactivation capacity in T cells and macrophages, although each gene has also unique properties in thymocyte development.
Collapse
Affiliation(s)
- Lawryn H Kasper
- Department of Biochemistry, St. Jude Children's Research Hospital, 332 N. Lauderdale, Memphis, TN 38105, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
184
|
Abstract
In an era marked by the increasing prevalence of obesity, diabetes, and cardiovascular disease, the nuclear receptor peroxisome proliferator-activated receptor gamma (PPARgamma) has emerged as a transcriptional regulator of metabolism whose activity can be modulated by direct binding of small molecules. As the master regulator of fat-cell formation, PPARgamma is required for the accumulation of adipose tissue and hence contributes to obesity. Yet PPARgamma ligands are clinically effective antidiabetic drugs, although side effects limit their utility. Can PPARgamma be targeted with greater benefit and with less risk to patients? The answer depends upon the basic biology of PPARgamma, and the possibility of selectively modulating the activity of this nuclear receptor in a tissue- and target-gene-specific manner.
Collapse
Affiliation(s)
- Michael Lehrke
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, and The Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | |
Collapse
|
185
|
Belvisi MG, Hele DJ, Birrell MA. Peroxisome proliferator-activated receptor gamma agonists as therapy for chronic airway inflammation. Eur J Pharmacol 2006; 533:101-9. [PMID: 16458290 DOI: 10.1016/j.ejphar.2005.12.048] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/13/2005] [Indexed: 12/21/2022]
Abstract
Peroxisome proliferator-activated receptor gamma (PPARgamma) is a ligand-activated transcription factor belonging to the nuclear hormone receptor superfamily. PPARgamma regulates several metabolic pathways by binding to sequence-specific PPAR response elements in the promoter region of target genes, including lipid biosynthesis and glucose metabolism. Synthetic PPARgamma agonists have been developed, such as the thiazolidinediones rosiglitazone and pioglitazone. These act as insulin sensitizers and are used in the treatment of type 2 diabetes. Recently however, PPARgamma ligands have been implicated as regulators of cellular inflammatory and immune responses. They are thought to exert anti-inflammatory effects by negatively regulating the expression of pro-inflammatory genes. Several studies have demonstrated that PPARgamma ligands possess anti-inflammatory properties and that these properties may prove helpful in the treatment of inflammatory diseases of the airways. This review will outline the anti-inflammatory effects of synthetic and endogenous PPARgamma ligands and discuss their potential therapeutic effects in animal models of inflammatory airway disease.
Collapse
Affiliation(s)
- Maria G Belvisi
- Respiratory Pharmacology, Airway Diseases, National Heart and Lung Institute, Faculty of Medicine, Imperial College, Guy Scadding Building, Dovehouse Street, London, SW3 6LY UK.
| | | | | |
Collapse
|
186
|
Pereira MP, Hurtado O, Cárdenas A, Boscá L, Castillo J, Dávalos A, Vivancos J, Serena J, Lorenzo P, Lizasoain I, Moro MA. Rosiglitazone and 15-deoxy-Delta12,14-prostaglandin J2 cause potent neuroprotection after experimental stroke through noncompletely overlapping mechanisms. J Cereb Blood Flow Metab 2006; 26:218-229. [PMID: 16034372 DOI: 10.1038/sj.jcbfm.9600182] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Stroke triggers an inflammatory cascade which contributes to a delayed cerebral damage, thus implying that antiinflammatory strategies might be useful in the treatment of acute ischaemic stroke. Since two unrelated peroxisome proliferator-activated receptor-gamma (PPARgamma) agonists, the thiazolidinedione rosiglitazone (RSG) and the cyclopentenone prostaglandin 15-deoxy-Delta(12,14)-prostaglandin J2 (15d-PGJ2), have been shown to possess antiinflammatory properties, we have tested their neuroprotective effects in experimental stroke. Rosiglitazone or 15d-PGJ2 were administered to rats 10 mins or 2 h after permanent middle cerebral artery occlusion (MCAO). Stroke outcome was evaluated by determination of infarct volume and assessment of neurological scores. Brains were collected for protein expression, gene array analyses and gene shift assays. Our results show that both compounds decrease MCAO-induced infarct size and improve neurological scores. At late times, the two compounds converge in the inhibition of MCAO-induced brain expression of inducible NO synthase and the matrix metalloproteinase 9. Interestingly, at early times, complementary DNA microarrays and gene shift assays show that different mechanisms are recruited. Analysis of early nuclear p65 and late cytosolic IkappaBalpha protein levels shows that both compounds inhibit nuclear factor-kappaB signalling, although at different levels. All these results suggest both PPARgamma-dependent and independent pathways, and might be useful to design both therapeutic strategies and prognostic markers for stroke.
Collapse
Affiliation(s)
- Marta P Pereira
- Departamento de Farmacología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
187
|
Feige JN, Gelman L, Michalik L, Desvergne B, Wahli W. From molecular action to physiological outputs: peroxisome proliferator-activated receptors are nuclear receptors at the crossroads of key cellular functions. Prog Lipid Res 2006; 45:120-59. [PMID: 16476485 DOI: 10.1016/j.plipres.2005.12.002] [Citation(s) in RCA: 578] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Peroxisome proliferator-activated receptors (PPARs) compose a family of three nuclear receptors which act as lipid sensors to modulate gene expression. As such, PPARs are implicated in major metabolic and inflammatory regulations with far-reaching medical consequences, as well as in important processes controlling cellular fate. Throughout this review, we focus on the cellular functions of these receptors. The molecular mechanisms through which PPARs regulate transcription are thoroughly addressed with particular emphasis on the latest results on corepressor and coactivator action. Their implication in cellular metabolism and in the control of the balance between cell proliferation, differentiation and survival is then reviewed. Finally, we discuss how the integration of various intra-cellular signaling pathways allows PPARs to participate to whole-body homeostasis by mediating regulatory crosstalks between organs.
Collapse
Affiliation(s)
- Jérôme N Feige
- Center for Integrative Genomics, NCCR Frontiers in Genetics, Le Génopode, University of Lausanne, CH-1015 Lausanne, Switzerland
| | | | | | | | | |
Collapse
|
188
|
Liu D, Zeng BX, Zhang SH, Wang YL, Zeng L, Geng ZL, Zhang SF. Rosiglitazone, a peroxisome proliferator-activated receptor-gamma agonist, reduces acute lung injury in endotoxemic rats. Crit Care Med 2005; 33:2309-16. [PMID: 16215386 DOI: 10.1097/01.ccm.0000183161.81503.7d] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Rosiglitazone, a potent agonist of peroxisome proliferator-activated receptor (PPAR)-gamma, exerts anti-inflammatory effects in vitro and in vivo. This study was designated to determine the effects of rosiglitazone on endotoxin-induced acute lung injury in rats. DESIGN Prospective, experimental study. SETTING University research laboratory. SUBJECTS Thirty-six male Wistar rats. INTERVENTIONS All the animals were randomly assigned to one of six groups (n = 6 per group) and were given either lipopolysaccharide (6 mg/kg intravenously) or saline, pretreated with rosiglitazone (0.3 mg/kg intravenously) or vehicle (10% dimethyl sulphoxide) 30 mins before lipopolysaccharide. The selective PPAR-gamma antagonist GW9662 (0.3 mg/kg intravenously) or its vehicle (10% dimethyl sulphoxide) was given 20 mins before rosiglitazone. MEASUREMENTS AND MAIN RESULTS Endotoxemia for 4 hrs induced evident lung histologic injury and edema, both of which were significantly attenuated by rosiglitazone pretreatment. The protective effects of rosiglitazone were correlated with the reduction by 71% of the increase of myeloperoxidase activity and the reduction by 84% of the increase of malondialdehyde in the lung tissue. The pulmonary hyperproduction of nitric oxide was reduced by 82% of the increase related to lipopolysaccharide challenge. Pretreatment with rosiglitazone also markedly suppressed lipopolysaccharide-induced expression of inducible nitric oxide synthase messenger RNA and protein in the lung, as demonstrated by reverse transcription-polymerase chain reaction or Western blot analysis. Immunohistochemical analysis revealed that rosiglitazone inhibited the formation of nitrotyrosine, a marker for peroxynitrite reactivity, in the lung tissue. In addition, the specific PPAR-gamma antagonist GW9662 antagonized the effects of rosiglitazone. CONCLUSIONS This study provides evidence, for the first time, that the PPAR-gamma agonist rosiglitazone significantly reduces endotoxin-induced acute lung injury in rats.
Collapse
Affiliation(s)
- Dong Liu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | | | | | | | | | | |
Collapse
|
189
|
Lu D, Cottam HB, Corr M, Carson DA. Repression of beta-catenin function in malignant cells by nonsteroidal antiinflammatory drugs. Proc Natl Acad Sci U S A 2005; 102:18567-71. [PMID: 16352713 PMCID: PMC1317972 DOI: 10.1073/pnas.0509316102] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Activation of the Wnt/beta-catenin pathway promotes the development of several cancers and is an attractive target for chemopreventive and chemotherapeutic agents. Nonsteroidal antiinflammatory drugs (NSAIDs) have been reported to antagonize beta-catenin function, but their mechanism of action is not known. We demonstrate here that interference with beta-catenin function by NSAIDs does not correlate with cyclooxygenase (COX) inhibition. Instead, NSAID inhibition of beta-catenin requires the high level expression of peroxisome proliferator-activated receptor gamma (PPAR-gamma) and its co-receptor retinoid-X-receptor alpha (RXR-alpha). Immunoprecipitation experiments show that beta-catenin interacts with RXR-alpha and PPAR-gamma in some malignant cells. Repression of beta-catenin-dependent transcription by NSAIDs is thus indirect and depends on the coexpression of other nuclear receptors.
Collapse
Affiliation(s)
- Desheng Lu
- Rebecca and John Moores Cancer Center, University of California at San Diego, La Jolla, CA 92093, USA.
| | | | | | | |
Collapse
|
190
|
Ogawa S, Lozach J, Benner C, Pascual G, Tangirala RK, Westin S, Hoffmann A, Subramaniam S, David M, Rosenfeld MG, Glass CK. Molecular determinants of crosstalk between nuclear receptors and toll-like receptors. Cell 2005; 122:707-21. [PMID: 16143103 PMCID: PMC1430687 DOI: 10.1016/j.cell.2005.06.029] [Citation(s) in RCA: 496] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2005] [Revised: 05/08/2005] [Accepted: 06/24/2005] [Indexed: 12/14/2022]
Abstract
Nuclear receptors (NRs) repress transcriptional responses to diverse signaling pathways as an essential aspect of their biological activities, but mechanisms determining the specificity and functional consequences of transrepression remain poorly understood. Here, we report signal- and gene-specific repression of transcriptional responses initiated by engagement of toll-like receptors (TLR) 3, 4, and 9 in macrophages. The glucocorticoid receptor (GR) represses a large set of functionally related inflammatory response genes by disrupting p65/interferon regulatory factor (IRF) complexes required for TLR4- or TLR9-dependent, but not TLR3-dependent, transcriptional activation. This mechanism requires signaling through MyD88 and enables the GR to differentially regulate pathogen-specific programs of gene expression. PPARgamma and LXRs repress overlapping transcriptional targets by p65/IRF3-independent mechanisms and cooperate with the GR to synergistically transrepress distinct subsets of TLR-responsive genes. These findings reveal combinatorial control of homeostasis and immune responses by nuclear receptors and suggest new approaches for treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Sumito Ogawa
- Department of Cellular and Molecular Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
191
|
Zingarelli B, Cook JA. Peroxisome proliferator-activated receptor-gamma is a new therapeutic target in sepsis and inflammation. Shock 2005; 23:393-9. [PMID: 15834303 DOI: 10.1097/01.shk.0000160521.91363.88] [Citation(s) in RCA: 121] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Peroxisome proliferator-activated receptor-gamma (PPARgamma) is a member of the nuclear receptor superfamily and a ligand-activated transcription factor with pleiotropic effects on lipid metabolism, inflammation, and cell proliferation. PPARgamma forms a heterodimer with the retinoid X receptor and upon ligand-activation binds to the PPAR response element in the promoter of genes to allow transcription. The class of insulin-sensitizing drugs known as thiazolidinediones have been identified as specific PPARgamma agonists that have allowed the characterization of many genes regulated by PPARgamma. Thiazolidinediones include rosiglitazone, pioglitazone, troglitazone, and ciglitazone. In addition to these synthetic agonists, cyclopentenone prostaglandins of the J2 series have been identified as natural ligands for PPARgamma. Several in vitro and in vivo studies have demonstrated that pharmacological activation of PPARgamma by 15-deoxy-Delta(12,14)-PGJ2 (15d-PGJ2) or thiazolidinediones has anti-inflammatory effects. This article provides an overview of the role of PPARgamma in regulating the inflammatory response and emphasizes the potential efficacy of PPARgamma ligands as novel therapeutic approaches beyond diabetes in sepsis, inflammation, and reperfusion injury.
Collapse
Affiliation(s)
- Basilia Zingarelli
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center and the College of Medicine, University of Cincinnati, Cincinnati, Ohio 45229, USA.
| | | |
Collapse
|
192
|
Coyle AT, O'Keeffe MB, Kinsella BT. 15-deoxy Delta12,14-prostaglandin J2 suppresses transcription by promoter 3 of the human thromboxane A2 receptor gene through peroxisome proliferator-activated receptor gamma in human erythroleukemia cells. FEBS J 2005; 272:4754-73. [PMID: 16156795 DOI: 10.1111/j.1742-4658.2005.04890.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
In humans, thromboxane (TX) A2 signals through two receptor isoforms, thromboxane receptor (TP)alpha and TPbeta, which are transcriptionally regulated by distinct promoters, Prm1 and Prm3, respectively, within the single TP gene. The aim of the current study was to investigate the ability of the endogenous peroxisome proliferator-activated receptor (PPAR)gamma ligand 15-deoxy-Delta12,14-prostaglandin J2 (15d-PGJ2) to regulate expression of the human TP gene and to ascertain its potential effects on the individual TPalpha and TPbeta isoforms. 15d-PGJ2 suppressed Prm3 transcriptional activity and TPbeta mRNA expression in the platelet progenitor megakaryocytic human erythroleukemia (HEL) 92.1.7 cell line but had no effect on Prm1 or Prm2 activity or on TPalpha mRNA expression. 15d-PGJ2 also resulted in reductions in the overall level of TP protein expression and TP-mediated intracellular calcium mobilization in HEL cells. 15d-PGJ2 suppression of Prm3 transcriptional activity and TPbeta mRNA expression was found to occur through a novel mechanism involving direct binding of PPARgamma-retinoic acid X receptor (RXR) heterodimers to a PPARgamma response element (PPRE) composed of two imperfect hexameric direct repeat (DR) sequences centred at -159 and -148, respectively, spaced by five nucleotides (DR5). These data provide direct evidence for the role of PPARgamma in the regulation of human TP gene expression within the vasculature and point to further critical differences in the modes of transcriptional regulation of TPalpha and TPbeta in humans. Moreover, these data highlight a further link between enhanced risk of cardiovascular disease in diabetes mellitus associated with increased synthesis and action of thromboxane A2 (TXA2).
Collapse
MESH Headings
- Binding Sites
- Cell Line, Tumor
- Gene Expression Regulation
- Humans
- Leukemia, Erythroblastic, Acute/pathology
- Ligands
- PPAR gamma/metabolism
- PPAR gamma/physiology
- Promoter Regions, Genetic
- Prostaglandin D2/analogs & derivatives
- Prostaglandin D2/physiology
- Protein Isoforms
- RNA, Messenger/analysis
- RNA, Messenger/genetics
- Receptors, Thromboxane A2, Prostaglandin H2/analysis
- Receptors, Thromboxane A2, Prostaglandin H2/genetics
- Response Elements
- Transcription, Genetic
Collapse
Affiliation(s)
- Adrian T Coyle
- Department of Biochemistry, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Ireland
| | | | | |
Collapse
|
193
|
|
194
|
Kim J, Oh YS, Shinn SH. Troglitazone reverses the inhibition of nitric oxide production by high glucose in cultured bovine retinal pericytes. Exp Eye Res 2005; 81:65-70. [PMID: 15978256 DOI: 10.1016/j.exer.2005.01.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2004] [Revised: 01/12/2005] [Accepted: 01/16/2005] [Indexed: 12/01/2022]
Abstract
In the retinal microcirculation, there is a basal release of nitric oxide (NO) which maintains the retinal blood flow. The proportions of endothelial cells and pericytes in the retinal capillaries are almost equal, so pericytes appear to play a important role in the regulation of microcirculatory hemodynamics in the retina. It has been suggested that the pathogenesis of early diabetic retinopathy may involve a reduced bioavailability or diminished production of NO. In this study, we investigated the role of troglitazone, a potent agonist of peroxisome proliferator activated receptor-gamma (PPARgamma) used for the treatment of diabetes, on the NO release and the effect of exposure to high glucose on the production of NO in cultured bovine retinal pericytes. Troglitazone significantly increased NO production and iNOS expression after 24hr in a dose-and PPARgamma-dependent manner. Elevation of D-glucose, but not L-glucose, from 5.5 to 30 mm for 24 hr decreased NO production, but co-treatment with troglitazone reversed high glucose-induced inhibition of NO production as well as iNOS expression. In conclusion, high glucose inhibits iNOS expression and subsequently NO synthesis in cultured bovine retinal pericytes, and troglitazone restores the NO production.
Collapse
Affiliation(s)
- Jaetaek Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Chung-Ang University, Seoul, South Korea.
| | | | | |
Collapse
|
195
|
Pereira MP, Hurtado O, Cárdenas A, Alonso-Escolano D, Boscá L, Vivancos J, Nombela F, Leza JC, Lorenzo P, Lizasoain I, Moro MA. The nonthiazolidinedione PPARgamma agonist L-796,449 is neuroprotective in experimental stroke. J Neuropathol Exp Neurol 2005; 64:797-805. [PMID: 16141790 DOI: 10.1097/01.jnen.0000178852.83680.3c] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Some agonists of the peroxisome proliferator-activated receptor gamma (PPARgamma) belonging to the thiazolidinedione (TZD) family, as well as the cyclopentenone prostaglandin 15-dPGJ2, have been shown to cause neuroprotection in animal models of stroke. We have tested whether the TZD-unrelated PPARgamma agonist L-796,449 is neuroprotective after permanent middle cerebral artery occlusion (MCAO) in the rat brain. Our results show that L-796,449 decreases MCAO-induced infarct size and improves neurologic scores. This protection is concomitant to inhibition of MCAO-induced brain expression of inducible NO synthase (iNOS) and the matrix metalloproteinase MMP-9 and to upregulation of the cytoprotective stress protein heme oxygenase-1 (HO-1). Analysis of the NF-kappaB p65 monomer and the NF-kappaB inhibitor IkappaBalpha protein levels as well as gel mobility shift assays indicate that L-796,449 inhibits NF-kappaB signaling, and that it may be recruiting both PPARgamma-dependent and independent pathways. In summary, our results provide new insights for stroke treatment.
Collapse
Affiliation(s)
- Marta P Pereira
- Departamento de Farmacología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
196
|
Abstract
ATP binding cassette transporter A1 (ABCA1) mediates the cellular efflux of phospholipids and cholesterol to lipid-poor apolipoprotein A1 (apoA1) and plays a significant role in high density lipoprotein (HDL) metabolism. ABCA1's role in the causation of Tangier disease, characterized by absent HDL and premature atherosclerosis, has implicated this transporter and its regulators liver-X-receptoralpha (LXRalpha) and peroxisome proliferator activated receptorgamma (PPARgamma) as new candidates potentially influencing the progression of atherosclerosis. In addition to lipid regulation, these genes are involved in apoptosis and inflammation, processes thought to be central to atherosclerotic plaque progression. A Medline-based review of the literature was carried out. Tangier disease and human heterozygotes with ABCA1 mutations provide good evidence that ABCA1 is a major candidate influencing atherosclerosis. Animal and in vitro experiments suggest that ABCA1 not only mediates cholesterol and phospholipid efflux, but is also involved in the regulation of apoptosis and inflammation. The complex and beneficial interactions between apoA1 and ABCA1 seem to be pivotal for cholesterol efflux. The expression of the ABCA1 is tightly regulated. Furthermore the plaque microenvironment could potentially promote ABCA1 protein degradation thus compromising cholesterol efflux. PPAR-LXR-ABCA1 interactions are integral to cholesterol homeostasis and these nuclear receptors have proven anti-inflammatory and anti-matrix metalloproteinase activity. Therapeutic manipulation of the ABCA1 transporter is feasible using PPAR and LXR agonists. PPAR agonists like glitazones and ABCA1 protein stabilization could potentially modify the clinical progression of atherosclerotic lesions.
Collapse
Affiliation(s)
- S Soumian
- Department of Vascular Surgery, Faculty of Medicine, Imperial College, Charing Cross Hospital, London, UK.
| | | | | | | |
Collapse
|
197
|
Pascual G, Fong AL, Ogawa S, Gamliel A, Li AC, Perissi V, Rose DW, Willson T, Rosenfeld MG, Glass CK. A SUMOylation-dependent pathway mediates transrepression of inflammatory response genes by PPAR-gamma. Nature 2005; 437:759-63. [PMID: 16127449 PMCID: PMC1464798 DOI: 10.1038/nature03988] [Citation(s) in RCA: 1005] [Impact Index Per Article: 50.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2005] [Accepted: 06/30/2005] [Indexed: 12/12/2022]
Abstract
Peroxisome proliferator-activated receptor-gamma (PPAR-gamma) has essential roles in adipogenesis and glucose homeostasis, and is a molecular target of insulin-sensitizing drugs. Although the ability of PPAR-gamma agonists to antagonize inflammatory responses by transrepression of nuclear factor kappa B (NF-kappaB) target genes is linked to antidiabetic and antiatherogenic actions, the mechanisms remain poorly understood. Here we report the identification of a molecular pathway by which PPAR-gamma represses the transcriptional activation of inflammatory response genes in mouse macrophages. The initial step of this pathway involves ligand-dependent SUMOylation of the PPAR-gamma ligand-binding domain, which targets PPAR-gamma to nuclear receptor corepressor (NCoR)-histone deacetylase-3 (HDAC3) complexes on inflammatory gene promoters. This in turn prevents recruitment of the ubiquitylation/19S proteosome machinery that normally mediates the signal-dependent removal of corepressor complexes required for gene activation. As a result, NCoR complexes are not cleared from the promoter and target genes are maintained in a repressed state. This mechanism provides an explanation for how an agonist-bound nuclear receptor can be converted from an activator of transcription to a promoter-specific repressor of NF-kappaB target genes that regulate immunity and homeostasis.
Collapse
Affiliation(s)
- Gabriel Pascual
- Department of Cellular and Molecular Medicine
- Biomedical Sciences Graduate ProgramUniversity of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093
| | - Amy L. Fong
- Department of Cellular and Molecular Medicine
- Biomedical Sciences Graduate ProgramUniversity of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093
| | | | | | | | | | | | - Timothy Willson
- GlaxoSmithKline, 5 Moore Drive, PO Box 13398, Research Triangle Park, NC 27709
| | | | - Christopher K. Glass
- Department of Cellular and Molecular Medicine
- Department of Medicine
- *To whom correspondence should be addressed Office: 1 858 534 6011, Fax: 1 858 822 2127,
| |
Collapse
|
198
|
Feinstein DL, Spagnolo A, Akar C, Weinberg G, Murphy P, Gavrilyuk V, Dello Russo C. Receptor-independent actions of PPAR thiazolidinedione agonists: is mitochondrial function the key? Biochem Pharmacol 2005; 70:177-88. [PMID: 15925327 DOI: 10.1016/j.bcp.2005.03.033] [Citation(s) in RCA: 220] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2005] [Revised: 03/17/2005] [Accepted: 03/23/2005] [Indexed: 12/17/2022]
Abstract
Agonists of the peroxisome proliferator activated receptor gamma (PPAR(gamma)) are currently used for treatment of type 2 diabetes due to their insulin sensitizing and glucose metabolism stabilizing effects. More recently some of these same agonists were shown to exert anti-inflammatory and anti-proliferative effects as well. Although PPAR(gamma) agonists can operate via receptor-mediated events occurring at the genomic level, thereby causing long lasting changes in gene expression patterns, recent studies demonstrate non-genomic as well as genomic actions, and receptor-dependent as well as receptor-independent effects of the thiazolidinedione (TZD) class of PPAR(gamma) agonists. In this review we will summarize data describing some of these novel, receptor independent actions of TZDs, review evidence that TZDs directly influence mitochondrial function, and attempt to reconcile how changes in mitochondrial function could contribute to other receptor-independent actions of these drugs.
Collapse
Affiliation(s)
- D L Feinstein
- Department of Anesthesiology, University of Illinois, VA Chicago Health Care System, Research & Development, Chicago, IL 60612, USA.
| | | | | | | | | | | | | |
Collapse
|
199
|
Crosby MB, Svenson J, Gilkeson GS, Nowling TK. A novel PPAR response element in the murine iNOS promoter. Mol Immunol 2005; 42:1303-10. [PMID: 15950726 DOI: 10.1016/j.molimm.2004.12.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2004] [Accepted: 12/17/2004] [Indexed: 12/20/2022]
Abstract
The nuclear hormone receptor peroxisome proliferation activated receptor gamma (PPARgamma) is a modulator of inflammation including down-regulation of inducible nitric oxide synthase (iNOS) and nitric oxide (NO) production. PPARgamma agonists reduce iNOS expression and NO production in a dose-dependent manner in macrophages, mesangial cells and other inflammatory cells. However, the mechanisms involved in the inhibition of iNOS expression by PPARgamma and its agonists are not fully understood. Here we show that the PPARgamma agonist ciglitazone dose-dependently inhibited a murine iNOS-luciferase reporter construct by up to 50% in transfected mesangial cells. Blocking de novo protein synthesis in mesangial cells had no effect on PPARgamma agonist activity, indicating that ciglitazone acts directly to inhibit iNOS transcription. We identified a novel PPAR response element (PPRE) in the murine iNOS promoter that is homologous to the PPRE consensus sequence. In binding assays PPARgamma directly binds to this response element in vitro and can function as a positive element in response to PPARgamma agonists when placed in front of a reporter gene. Site-directed mutagenesis of this PPRE in a murine iNOS promoter/reporter construct did not block the inhibitory activity of a synthetic PPARgamma agonist on the iNOS promoter/reporter construct in transfected mesangial cells. However, the mutated construct exhibited lower basal expression, and higher expression in response to inflammatory stimuli compared to the intact construct. These data suggest that the iNOS PPRE contributes to positive basal expression and negative expression of iNOS in response to inflammatory stimuli. The PPRE is not necessary, however, for synthetic PPARgamma agonists to inhibit iNOS expression.
Collapse
Affiliation(s)
- Michelle B Crosby
- Department of Medicine, Division of Rheumatology, Medical University of South Carolina, 96 Jonathon Lucas Street, Ste 912 CSB, Charleston, SC 29425, USA
| | | | | | | |
Collapse
|
200
|
Berry EBE, Keelan JA, Helliwell RJA, Gilmour RS, Mitchell MD. Nanomolar and micromolar effects of 15-deoxy-delta 12,14-prostaglandin J2 on amnion-derived WISH epithelial cells: differential roles of peroxisome proliferator-activated receptors gamma and delta and nuclear factor kappa B. Mol Pharmacol 2005; 68:169-78. [PMID: 15821150 DOI: 10.1124/mol.104.009449] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
15-Deoxy delta(12,14)-prostaglandin J(2) (15d-PGJ(2)), an activator of peroxisome proliferator-activated receptor (PPAR)-gamma and -delta, is a prostanoid metabolite with anti-inflammatory actions. In intrauterine tissues, proinflammatory cytokines and prostaglandins have been identified as playing key roles in the maintenance of pregnancy and the onset of labor. We investigated and compared the early (<3 h) effects of 15d-PGJ(2) with rosiglitazone (PPAR-gamma ligand) and 2-methyl-4-((4-methyl-2-(4-trifluoromethylphenyl)-1,3-thiazol-5-yl)-methylsulfanyl)phenoxy-acetic acid (GW501516) (PPAR-delta ligand) on interleukin (IL)-1beta-induced prostaglandin and cytokine production by amnion-derived WISH cells. We show that 15d-PGJ(2) exerts differential effects depending on concentration. At low concentrations (<0.1 microM), 15d-PGJ(2) inhibited IL-1beta-stimulated prostaglandin E(2) (PGE(2)) but not cytokine (IL-6/IL-8) production or cyclooxygenase-2 (COX-2) expression. This effect was attenuated by a PPAR-gamma inhibitor [2-chloro-5-nitro-N-phenyl-benzamide (GW9662)], by transfection with a dominant-negative PPAR construct, and was reproduced by the PPAR-gamma ligand rosiglitazone. At higher concentrations (1-10 microM), 15d-PGJ(2) inhibited IL-1beta-stimulated PGE(2) and cytokine production and COX-2 expression, and this effect was not blocked by GW9662. Rosiglitazone at high concentrations (1-10 microM) stimulated PGE(2) production in the absence or presence of the dominant-negative PPAR. The PPAR-delta ligand GW501516 also inhibited IL-1beta-stimulated PGE(2) production but only at high concentrations (1 microM). IL-1beta-induced nuclear factor-kappaB (NF-kappaB) DNA binding activity was significantly inhibited by 15d-PGJ(2) (10 microM) and GW501516 (1 microM) but increased with 10 microM rosiglitazone. We conclude that 1) at low concentrations, 15d-PGJ(2) acts through a PPAR-gamma signaling pathway; b) at higher concentrations, its actions are mediated most likely through other pathways such as activation of PPAR-delta and/or inhibition of NF-kappaB; and 3) rosiglitazone exerts PPAR-independent effects at high concentrations (>1 microM).
Collapse
Affiliation(s)
- Elicia B E Berry
- Liggins Institute, University of Auckland, Faculty of Medical & Health Sciences, Auckland, New Zealand
| | | | | | | | | |
Collapse
|