201
|
Peroxisome proliferator-activated receptors and the vascular system: beyond their metabolic effects. ACTA ACUST UNITED AC 2008; 2:227-38. [DOI: 10.1016/j.jash.2007.12.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2007] [Revised: 12/28/2007] [Accepted: 12/28/2007] [Indexed: 12/19/2022]
|
202
|
The effects of rosiglitazone on aortic atherosclerosis of cholesterol-fed rabbits. Thromb Res 2008; 123:281-7. [PMID: 18561986 DOI: 10.1016/j.thromres.2008.04.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2008] [Revised: 04/16/2008] [Accepted: 04/24/2008] [Indexed: 11/23/2022]
Abstract
INTRODUCTION Thiazolidinedione (TZD) is widely used a drug for the treatment of type 2 diabetes and protects against cardiovascular events in human. However, it is not clear whether TZD can directly inhibit the progression of atherosclerosis. To test the hypothesis whether administration of TZD could reduce the development of atherosclerosis, we studied the effects of rosiglitazone on aortic atherosclerosis of rabbits fed a cholesterol diet. MATERIALS AND METHODS Male Japanese White rabbits were fed a diet containing either 0.3% cholesterol diet (control group, n=10) or 0.3% cholesterol with rosiglitazone (TZD-treated group, n=12) for 16 weeks. We compared the plasma lipids and the extent of aortic atherosclerosis between two groups. RESULTS AND CONCLUSIONS TZD treatment significantly resulted in the reduction of aortic atherosclerosis by 21% in the aortic arch (p<0.01), 20% in the thoracic aorta (p=0.14), and 28% in the abdominal aorta (p=0.25), without affecting the plasma levels of triglycerides, total cholesterol, high-density lipoprotein cholesterol, glucose and insulin. Immunohistochemical staining showed that the cellular components (macrophages and smooth muscle cells) of the lesions of TZD-treated rabbits were unchanged compared to those of control rabbits. In addition, TZD treatment also led to dramatic improvement of fatty liver in cholesterol-fed rabbits. Our results suggest that the activation of PPARgamma can be beneficial for the treatment of atherosclerosis and fatty liver independent upon the improvement of plasma lipids and glucose metabolism.
Collapse
|
203
|
Effects of rosiglitazone on contralateral iliac artery after vascular injury in hypercholesterolemic rabbits. Thromb J 2008; 6:4. [PMID: 18485218 PMCID: PMC2396603 DOI: 10.1186/1477-9560-6-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2007] [Accepted: 05/16/2008] [Indexed: 11/10/2022] Open
Abstract
Background The objective was to evaluate the effects of rosiglitazone on iliac arteries of hypercholesterolemic rabbits undergoing balloon catheter injury in the contralateral iliac arteries. Methods White male rabbits were fed a hypercholesterolemic diet for 6 weeks and divided into two groups as follows: rosiglitazone group, 14 rabbits treated with rosiglitazone (3 mg/Kg body weight/day) during 6 weeks; and control group, 18 rabbits without rosiglitazone treatment. All animals underwent balloon catheter injury of the right iliac artery on the fourteenth day of the experiment. Results There was no significant difference in intima/media layer area ratio between the control group and the rosiglitazone group. Rosiglitazone did not reduce the probability of lesions types I, II, or III (72.73% vs. 92.31%; p = 0.30) and types IV or V (27.27% vs. 7.69%; p = 0.30). There were no differences in the extent of collagen type I and III deposition or in the percentage of animals with macrophages in the intima layer. The percentage of rabbits with smooth muscle cells in the intima layer was higher in rosiglitazone group (p = 0.011). Conclusion These findings demonstrate that rosiglitazone given for 6 weeks did not prevent atherogenesis at a vessel distant from the injury site.
Collapse
|
204
|
Kim YH, Choi SY, Suh JH, Kim TK, Seung KB, Wang YP, Chang K. The effect of Chlamydia pneumoniae on the expression of peroxisome proliferator-activated receptor-gamma in vascular smooth muscle cells. Yonsei Med J 2008; 49:230-6. [PMID: 18452259 PMCID: PMC2615309 DOI: 10.3349/ymj.2008.49.2.230] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
PURPOSE This study was designed to investigate the change of peroxisome proliferator-activated receptor gamma (PPARgamma) after the infection of the human coronary artery smooth muscle cells (HCSMCs) with Chlamydia pneumoniae (C. pneumoniae) and the effect of PPARgamma agonist on the expression of PPARgamma of C. pneumoniae-infected HCSMCs. MATERIALS AND METHODS To determine the effect of PPARgamma agonist on the proliferation of C. pneumoniae-infected HCSMCs, rosiglitazone at various concentrations was applied 1 hour before inoculation of HCSMCs. RESULTS The expression of PPARgamma mRNA in HCSMCs increased from 3 hours after C. pneumoniae infection and reached that of noninfected HCSMCs at 24 hours (p<0.05). The expression of PPARgamma protein in HCSMCs also increased from 3 hours after C. pneumoniae and persisted until 24 hours as compared with that of noninfected HCSMCs (p<0.05). The pretreatment of HCSMCs with rosiglitazone followed by the infection with C. pneumoniae augmented the expression of PPARgamma mRNA and protein (p<0.05) and decreased cell proliferation. CONCLUSION Our results showed that the expression of PPARgamma increases in response to C. pneumoniae infection and rosiglitazone further augmented the expression of PPARgamma. It is suggested that rosiglitazone could ameliorate the chronic inflammation in the vessel wall induced by C. pneumoniae by augmenting PPARgamma expression.
Collapse
MESH Headings
- Blotting, Western
- Cell Line
- Cell Proliferation/drug effects
- Chlamydophila pneumoniae/growth & development
- Chlamydophila pneumoniae/physiology
- Gene Expression Regulation/drug effects
- Humans
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/microbiology
- PPAR gamma/genetics
- PPAR gamma/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Rosiglitazone
- Thiazolidinediones/pharmacology
Collapse
Affiliation(s)
- Yong-Hwan Kim
- Department of Cardiothoracic Surgery, Catholic University College of Medicine, Seoul, Korea
| | - Si-Young Choi
- Department of Cardiothoracic Surgery, Catholic University College of Medicine, Seoul, Korea
| | - Jong-Hui Suh
- Department of Cardiothoracic Surgery, Catholic University College of Medicine, Seoul, Korea
| | - Tae-Kyun Kim
- Department of Internal Medicine, Catholic University College of Medicine, Seoul, Korea
| | - Ki-Bae Seung
- Department of Internal Medicine, Catholic University College of Medicine, Seoul, Korea
| | - Young-Pil Wang
- Department of Cardiothoracic Surgery, Catholic University College of Medicine, Seoul, Korea
| | - Kiyuk Chang
- Department of Internal Medicine, Catholic University College of Medicine, Seoul, Korea
| |
Collapse
|
205
|
Rizos CV, Liberopoulos EN, Mikhailidis DP, Elisaf MS. Pleiotropic effects of thiazolidinediones. Expert Opin Pharmacother 2008; 9:1087-108. [DOI: 10.1517/14656566.9.7.1087] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
206
|
Tian L, Luo N, Klein RL, Chung BH, Garvey WT, Fu Y. Adiponectin reduces lipid accumulation in macrophage foam cells. Atherosclerosis 2008; 202:152-61. [PMID: 18511057 DOI: 10.1016/j.atherosclerosis.2008.04.011] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2007] [Revised: 03/31/2008] [Accepted: 04/02/2008] [Indexed: 01/23/2023]
Abstract
Adiponectin is one of several, important metabolically active cytokines secreted from adipocytes. Low circulating levels of this adipokine have been associated epidemiologically with obesity, insulin resistance, type II diabetes, and cardiovascular disease. To determine if adiponectin can modulate lipid metabolism in macrophages, we expressed the adiponectin gene in human THP-1 macrophage foam cells using a lentiviral vector expression system and demonstrated that macrophages transduced with the adiponectin gene had decreased lipid accumulation compared with control macrophages transduced with the LacZ gene. Macrophages transduced with the adiponectin gene also exhibited decreased oxidized low-density lipoprotein (oxLDL) uptake and increased HDL-mediated cholesterol efflux. Additional studies suggest two potential mechanisms for the reduced lipid accumulation in these adiponectin-transduced macrophage foam cells. The first mechanism involves the PPARgamma and LXR signaling pathways which up-regulate the expression of ABCA1 and promote lipid efflux from these cells. The second mechanism involves decreased lipid uptake and increased lipid hydrolysis which may result from decreased SR-AI and increased SR-BI and HSL gene activities in the transformed macrophage foam cells. We also demonstrated that the expression of two proatherogenic cytokines, MCP-1 and TNFalpha, were decreased in the adiponectin-transduced macrophage foam cells. These results suggest that adiponectin may modulate multiple pathways of lipid metabolism in macrophages. Our studies provide new insights into potential mechanisms of adiponectin-mediated alterations in lipid metabolism and macrophage foam cell formation which may impact the development of atherosclerosis.
Collapse
Affiliation(s)
- Ling Tian
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL 35294-3360, United States
| | | | | | | | | | | |
Collapse
|
207
|
Kovesdy CP, Kalantar-Zadeh K. Novel targets and new potential: developments in the treatment of inflammation in chronic kidney disease. Expert Opin Investig Drugs 2008; 17:451-67. [PMID: 18363512 DOI: 10.1517/13543784.17.4.451] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Patients with chronic kidney disease (CKD) of all stages experience extremely high mortality, with cardiovascular causes accounting for about half of all their deaths. Traditional risk factors such as hypertension, hypercholesterolemia and diabetes mellitus cannot explain the excessively high cardiovascular mortality in CKD. Chronic inflammation is one of the novel risk factors that appear to contribute to the increased mortality seen in patients with CKD. Therapeutic interventions targeting chronic inflammation in CKD may lead to improved outcomes. OBJECTIVES To describe the role of inflammation in CKD and to review anti-inflammatory pharmacologic therapies that could have a role in its therapy. METHODS A review of the literature was carried out and expert opinion expressed. RESULTS/CONCLUSION Inflammation is a common and significant problem in CKD. There are currently no approved pharmacologic anti-inflammatory therapies in CKD but several agents are being studied in early clinical trials, while others could become viable alternatives in the future.
Collapse
Affiliation(s)
- Csaba P Kovesdy
- Salem VAMC (111D), 1970 Roanoke Blvd., Salem, VA 24153, USA.
| | | |
Collapse
|
208
|
Boon Yin K, Najimudin N, Muhammad TST. The PPARgamma coding region and its role in visceral obesity. Biochem Biophys Res Commun 2008; 371:177-9. [PMID: 18413145 DOI: 10.1016/j.bbrc.2008.04.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2008] [Accepted: 04/04/2008] [Indexed: 11/15/2022]
Abstract
Peroxisome proliferator-activated receptor gamma (PPARgamma) is a ligand activated transcription factor, plays many essential roles of biological function in higher organisms. The PPARgamma is mainly expressed in adipose tissue. It regulates the transcriptional activity of genes by binding with other transcription factor. The PPARgamma coding region has been found to be closest to that of monkey in ours and other research groups. Thus, monkey is a more suitable animal model for future PPARgamma studying, although mice and rat are frequently being used. The PPARgamma is involved in regulating alterations of adipose tissue masses result from changes in mature adipocyte size and/or number through a complex interplay process called adipogenesis. However, the role of PPARgamma in negatively regulating the process of adipogenesis remains unclear. This review may help we investigate the differential expression of key transcription factor in adipose tissue in response to visceral obesity-induced diet in vivo. The study may also provide valuable information to define a more appropriate physiological condition in adipogenesis which may help to prevent diseases cause by negative regulation of the transcription factors in adipose tissue.
Collapse
Affiliation(s)
- Khoo Boon Yin
- School of Biological Sciences, Universiti Sains Malaysia, 11800 Penang, Malaysia.
| | | | | |
Collapse
|
209
|
Sertznig P, Seifert M, Tilgen W, Reichrath J. Peroxisome proliferator-activated receptors (PPARs) and the human skin: importance of PPARs in skin physiology and dermatologic diseases. Am J Clin Dermatol 2008; 9:15-31. [PMID: 18092840 DOI: 10.2165/00128071-200809010-00002] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Peroxisome proliferator-activated receptors (PPARs) are members of the nuclear receptor superfamily that regulate lipid, glucose, and amino acid metabolism. More recently, PPARs and corresponding ligands have been shown in skin and other organs to regulate important cellular functions, including cell proliferation and differentiation, as well as inflammatory responses. These new functions identify PPARs and corresponding ligands as potential targets for the treatment of various skin diseases and other disorders. It has been shown that in inflammatory skin disorders, including hyperproliferative psoriatic epidermis and the skin of patients with atopic dermatitis, the expression of both PPARalpha and PPARgamma is decreased. This observation suggests the possibility that PPARalpha and PPARgamma activators, or compounds that positively regulate PPAR gene expression, may represent novel NSAIDs for the topical or systemic treatment of common inflammatory skin diseases such as atopic dermatitis, psoriasis, and allergic contact dermatitis. Moreover, recent findings indicate that PPAR-signaling pathways may act as a promising therapeutic target for the treatment of hyperproliferative skin diseases including skin malignancies. Studies in non-diabetic patients suggest that oral thiazolidinediones, which are synthetic ligands of PPARgamma, not only exert an antidiabetic effect but also may be beneficial for moderate chronic plaque psoriasis by suppressing proliferation and inducing differentiation of keratinocytes; furthermore, they may even induce cell growth arrest, apoptosis, and terminal differentiation in various human malignant tumors. It has been reported that PPARalpha immunoreactivity is reduced in human keratinocytes of squamous cell carcinoma (SCC) and actinic keratosis (AK), while PPARdelta appears to be upregulated. Additionally, the microvessel density is significantly higher in AK and SCC that express high levels of PPARdelta. PPARdelta has been demonstrated to have an anti-apoptotic role and to maintain survival and differentiation of epithelial cells, whereas PPARalpha and PPARgamma activators induce differentiation and inhibit proliferation and regulate apoptosis. In melanoma, the growth inhibitory effect of PPARgamma activation is independent of apoptosis and seems to occur primarily through induction of cell cycle arrest in the G1 phase of the cell cycle or induction of re-differentiation. PPARalpha activation causes inhibition of migration of melanoma cells and anchorage-independent growth, whereas primary tumor growth remains unaltered. In clinical trials of gemfibrozil, a PPARalpha ligand, significantly fewer patients treated with this lipid-lowering drug were diagnosed with melanoma as compared to those in the control group. In conclusion, an increasing body of evidence indicates that PPAR signaling pathways may represent interesting therapeutic targets for a broad variety of skin disorders, including inflammatory skin diseases such as psoriasis and atopic dermatitis, and skin malignancies.
Collapse
Affiliation(s)
- Pit Sertznig
- Department of Dermatology, The Saarland University Hospital, Homburg/Saar, Germany
| | | | | | | |
Collapse
|
210
|
Amoruso A, Bardelli C, Gunella G, Ribichini F, Brunelleschi S. A novel activity for substance P: stimulation of peroxisome proliferator-activated receptor-gamma protein expression in human monocytes and macrophages. Br J Pharmacol 2008; 154:144-52. [PMID: 18278062 DOI: 10.1038/bjp.2008.50] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND AND PURPOSE Substance P (SP) and peroxisome proliferator-activated receptor-gamma (PPAR-gamma) play important roles in different inflammatory conditions and are both expressed in human monocytes and macrophages. However, it is not known whether or not they interact. This study was undertaken to evaluate the effects of SP on PPAR-gamma protein expression in monocytes and macrophages (MDMs: monocyte-derived macrophages) from healthy smokers and non-smokers. EXPERIMENTAL APPROACH PPAR-gamma protein was detected by western blot and quantified by calculating the ratio between PPAR-gamma and beta-actin protein expression. Constitutive tachykinin NK(1) receptor expression in monocytes and MDMs from healthy smokers and non-smokers was evaluated by western blot. Cytokine release was evaluated by ELISA. KEY RESULTS In the concentration range 10(-10)-10(-6) M, SP stimulated PPAR-gamma protein expression in monocytes and MDMs, being more effective in cells from healthy smokers. Moreover, in these cells there was a constitutively increased expression of NK(1) receptors. SP-induced expression of the PPAR-gamma protein was receptor-mediated, as it was reproduced by the NK(1) selective agonist [Sar(9)Met(O(2))(11)]SP and reversed by the competitive NK(1) antagonist GR71251. SP-induced maximal effects were similar to those evoked by 15-deoxy-Delta(12,14)-prostaglandin J(2); an endogenous PPAR-gamma agonist, and were significantly reduced by a PPAR-gamma antagonist. NK(1) and PPAR-gamma agonists exerted opposite effects on TNF-alpha release from monocytes and MDMs. CONCLUSIONS AND IMPLICATIONS Enhancement of PPAR-gamma protein expression represents a novel activity for SP, which could contribute to a range of chronic inflammatory disorders.
Collapse
Affiliation(s)
- A Amoruso
- Department of Medical Sciences, School of Medicine, University of Piemonte Orientale A Avogadro, Novara, Italy
| | | | | | | | | |
Collapse
|
211
|
Duan SZ, Usher MG, Mortensen RM. Peroxisome Proliferator-Activated Receptor-γ–Mediated Effects in the Vasculature. Circ Res 2008; 102:283-94. [DOI: 10.1161/circresaha.107.164384] [Citation(s) in RCA: 216] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Peroxisome proliferator-activated receptor (PPAR)-γ is a nuclear receptor and transcription factor in the steroid superfamily. PPAR-γ agonists, the thiazolidinediones, are clinically used to treat type 2 diabetes. In addition to its function in adipogenesis and increasing insulin sensitivity, PPAR-γ also plays critical roles in the vasculature. In vascular endothelial cells, PPAR-γ activation inhibits endothelial inflammation by suppressing inflammatory gene expression and therefore improves endothelial dysfunction. In vascular smooth muscle cells, PPAR-γ activation inhibits proliferation and migration and promotes apoptosis. In macrophages, PPAR-γ activation suppresses inflammation by regulating gene expression and increases cholesterol uptake and efflux. A recurring theme in many cell types is the modulation of the innate immunity system particularly through altering the activity of the nuclear factor κB. This system is likely to be even more prominent in modulating disease in vascular cells. The effects of PPAR-γ in the vascular cells translate into the beneficial function of this transcription factor in vascular disorders, including hypertension and atherosclerosis. Both human genetic studies and animal studies using transgenic mice have demonstrated the importance of PPAR-γ in these disorders. However, recent clinical studies have raised significant concerns about the cardiovascular side effects of thiazolidinediones, particularly rosiglitazone. Weighing the potential benefit and harm of PPAR-γ activation and exploring the functional mechanisms may provide a balanced view on the clinical use of these compounds and new approaches to the future therapeutics of vascular disorders associated with diabetes.
Collapse
Affiliation(s)
- Sheng Zhong Duan
- From the Departments of Molecular and Integrative Physiology (S.Z.D., M.G.U., R.M.M.), Pharmacology (R.M.M.), and Internal Medicine (R.M.M.), Metabolism Endocrinology and Diabetes Division, University of Michigan Medical School, Ann Arbor
| | - Michael G. Usher
- From the Departments of Molecular and Integrative Physiology (S.Z.D., M.G.U., R.M.M.), Pharmacology (R.M.M.), and Internal Medicine (R.M.M.), Metabolism Endocrinology and Diabetes Division, University of Michigan Medical School, Ann Arbor
| | - Richard M. Mortensen
- From the Departments of Molecular and Integrative Physiology (S.Z.D., M.G.U., R.M.M.), Pharmacology (R.M.M.), and Internal Medicine (R.M.M.), Metabolism Endocrinology and Diabetes Division, University of Michigan Medical School, Ann Arbor
| |
Collapse
|
212
|
Barlic J, Murphy PM. An oxidized lipid-peroxisome proliferator-activated receptor gamma-chemokine pathway in the regulation of macrophage-vascular smooth muscle cell adhesion. Trends Cardiovasc Med 2008; 17:269-74. [PMID: 18021937 DOI: 10.1016/j.tcm.2007.09.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2007] [Revised: 08/31/2007] [Accepted: 09/07/2007] [Indexed: 01/16/2023]
Abstract
Recent genetic studies have implicated pro-inflammatory chemokines and chemokine receptors in atherogenesis. Studies at the molecular and cellular levels have suggested specific atherogenic mechanisms for two chemokine-chemokine receptor pairs, CCL2-CCR2 and CX3CL1-CX3CR1, involving differential receptor regulation by the transcription factor peroxisome proliferator-activated receptor gamma. This pathway is triggered by oxidized proatherogenic lipids, such as oxidized low-density lipoprotein and linoleic acid derivatives, which promote differentiation of CCR2(hi)CX3CR1(lo) human monocytes to CCR2(lo)CX3CR1(hi) macrophages that adhere to coronary artery smooth muscle cells in a CX3CR1- and peroxisome proliferator-activated receptor gamma-dependent manner. Switching CX3CR1 on and CCR2 off in vivo may result in cessation of CCR2-dependent migration and activation of CX3CR1-dependent retention that together may promote foam cell accumulation in the vessel wall.
Collapse
Affiliation(s)
- Jana Barlic
- Molecular Signaling Section, Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | |
Collapse
|
213
|
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: 0.9] [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]
|
214
|
Ho TC, Yang YC, Chen SL, Kuo PC, Sytwu HK, Cheng HC, Tsao YP. Pigment epithelium-derived factor induces THP-1 macrophage apoptosis and necrosis by the induction of the peroxisome proliferator-activated receptor gamma. Mol Immunol 2008; 45:898-909. [PMID: 17870167 DOI: 10.1016/j.molimm.2007.08.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2007] [Revised: 08/08/2007] [Accepted: 08/09/2007] [Indexed: 01/28/2023]
Abstract
Pigment epithelial-derived factor (PEDF) is a potent anti-angiogenic factor, partially through the induction of endothelial cell apoptosis. Here we report that PEDF can also induce the apoptosis of human THP-1 monocytic leukemia cell line-derived macrophage cells (THP-1 macrophages) and peroxisome proliferator-activated receptor gamma (PPARgamma), a pleiotropic transcriptional factor is involved in the signaling. TUNEL and propidium iodide permeability assays demonstrated that PEDF dose- and time-dependently induces both apoptosis and necrosis of THP-1 macrophages while inducing the cleavages of procaspase-9, -3, the release of cytochrome c and the overexpression of p53. All these PEDF effects can be attenuated by either PPARgamma inhibitor GW9662 or PPARgamma small interfering RNA. The effects of PEDF can be reproduced by transient expression of PPARgamma by a PPARgamma-expression plasmid transfection. PEDF increased the expression and transcriptional activity of PPARgamma in THP-1 macrophages. In addition, PEDF also induced apoptosis in primary human monocyte-derived macrophages (MDMs) while inducing the expression of PPARgamma. Our observations indicate that PEDF induces macrophage apoptosis and necrosis through the signaling of PPARgamma. This suggests a novel mechanism through which PEDF can modulate inflammation.
Collapse
Affiliation(s)
- Tsung-Chuan Ho
- Department of Medical Research, Mackay Memorial Hospital, Taipei, Taiwan
| | | | | | | | | | | | | |
Collapse
|
215
|
Xing B, Xin T, Hunter RL, Bing G. Pioglitazone inhibition of lipopolysaccharide-induced nitric oxide synthase is associated with altered activity of p38 MAP kinase and PI3K/Akt. J Neuroinflammation 2008; 5:4. [PMID: 18205920 PMCID: PMC2254610 DOI: 10.1186/1742-2094-5-4] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2007] [Accepted: 01/18/2008] [Indexed: 12/19/2022] Open
Abstract
Background Previous studies have suggested that peroxisome proliferator activated receptor-gamma (PPAR-γ)-mediated neuroprotection involves inhibition of microglial activation and decreased expression and activity of inducible nitric oxide synthase (iNOS); however, the underlying molecular mechanisms have not yet been well established. In the present study we explored: (1) the effect of the PPAR-γ agonist pioglitazone on lipopolysaccharide (LPS)-induced iNOS activity and nitric oxide (NO) generation by microglia; (2) the differential role of p38 mitogen-activated protein kinase (p38 MAPK), c-Jun NH(2)-terminal kinase (JNK), and phosphoinositide 3-kinase (PI3K) on LPS-induced NO generation; and (3) the regulation of p38 MAPK, JNK, and PI3K by pioglitazone. Methods Mesencephalic neuron-microglia mixed cultures, and microglia-enriched cultures were treated with pioglitazone and/or LPS. The protein levels of iNOS, p38 MAPK, JNK, PPAR-γ, PI3K, and protein kinase B (Akt) were measured by western blot. Different specific inhibitors of iNOS, p38MAPK, JNK, PI3K, and Akt were used in our experiment, and NO generation was measured using a nitrite oxide assay kit. Tyrosine hydroxylase (TH)-positive neurons were counted in mesencephalic neuron-microglia mixed cultures. Results Our results showed that pioglitazone inhibits LPS-induced iNOS expression and NO generation, and inhibition of iNOS is sufficient to protect dopaminergic neurons against LPS insult. In addition, inhibition of p38 MAPK, but not JNK, prevented LPS-induced NO generation. Further, and of interest, pioglitazone inhibited LPS-induced phosphorylation of p38 MAPK. Wortmannin, a specific PI3K inhibitor, enhanced p38 MAPK phosphorylation upon LPS stimulation of microglia. Elevations of phosphorylated PPAR-γ, PI3K, and Akt levels were observed with pioglitazone treatment, and inhibition of PI3K activity enhanced LPS-induced NO production. Furthermore, wortmannin prevented the inhibitory effect of pioglitazone on the LPS-induced NO increase. Conclusion We demonstrate that pioglitazone protects dopaminergic neurons against LPS insult at least via inhibiting iNOS expression and NO generation, which is potentially mediated via inhibition of p38 MAPK activity. In addition, the PI3K pathway actively participates in the negative regulation of LPS-induced NO production. Our findings suggest that PPAR-γ activation may involve differential regulation of p38 MAPK and of the PI3K/Akt pathway in the regulation of the inflammatory process.
Collapse
Affiliation(s)
- Bin Xing
- Department of Anatomy and Neurobiology, 310 Davis Mills Building, University of Kentucky, Chandler Medical Center, 800 Rose Street, Lexington, KY 40536-0298, USA.
| | | | | | | |
Collapse
|
216
|
Ringseis R, Schulz N, Saal D, Eder K. Troglitazone but not conjugated linoleic acid reduces gene expression and activity of matrix-metalloproteinases-2 and -9 in PMA-differentiated THP-1 macrophages. J Nutr Biochem 2007; 19:594-603. [PMID: 18155510 DOI: 10.1016/j.jnutbio.2007.08.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2007] [Revised: 08/06/2007] [Accepted: 08/07/2007] [Indexed: 12/17/2022]
Abstract
Gene expression and activity of matrix-metalloproteinases (MMP)-2 and -9 in macrophages are reduced through peroxisome proliferator-activated receptor gamma (PPARgamma)-dependent inhibition of NF-kappaB. Since conjugated linoleic acids (CLAs) are PPARgamma ligands and known to inhibit NF-kappaB via PPARgamma, we studied whether CLA isomers are capable of reducing gene expression and gelatinolytic activity of MMP-2 and -9 in PMA-differentiated THP-1 macrophages, which has not yet been investigated. Incubation of PMA-differentiated THP-1 cells with either c9t11-CLA, t10c12-CLA or linoleic acid (LA), as a reference fatty acid, resulted in a significant incorporation of the respective fatty acids into total cell lipids relative to control cells (P<.05). Treatment of PMA-differentiated THP-1 cells with 10 and 20 micromol/L troglitazone but not with 10 or 100 micromol/L c9t11-CLA, t10c12-CLA or LA reduced relative mRNA concentrations and activity of MMP-2 and MMP-9 compared to control cells (P<.05). DNA-binding activity of NF-kappaB and PPARgamma and mRNA expression of the NF-kappaB target gene cPLA2 were not influenced by treatment with CLA. In contrast, treatment of PMA-differentiated THP-1 cells with troglitazone significantly increased transactivation of PPARgamma and decreased DNA-binding activity of NF-kappaB and relative mRNA concentration of cPLA2 relative to control cells (P<.05). In conclusion, the present study revealed that CLA isomers, in contrast to troglitazone, did not reduce gene expression and activity of MMP-2 and -9 in PMA-differentiated THP-1 macrophages, which is probably explained by the observation that CLA isomers neither activated PPARgamma nor reduced DNA-binding activity of NF-kappaB. This suggests that CLA isomers are ineffective in MMP-associated extracellular matrix degradation which is thought to contribute to the progression and rupture of advanced atherosclerotic plaques.
Collapse
Affiliation(s)
- Robert Ringseis
- Institut für Agrar-und Ernährungswissenschaften, Martin-Luther-Universität Halle-Wittenberg, Emil-Abderhalden-Strasse 26, D-06108 Halle, Saale, Germany.
| | | | | | | |
Collapse
|
217
|
Toth B, Hornung D, Scholz C, Djalali S, Friese K, Jeschke U. Peroxisome proliferator-activated receptors: new players in the field of reproduction. Am J Reprod Immunol 2007; 58:289-310. [PMID: 17681045 DOI: 10.1111/j.1600-0897.2007.00514.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPAR) are members of the nuclear hormone receptor superfamily. Synthetic ligands to one family member, PPARgamma, are currently widely used as treatment for chronic diseases such as diabetes type II and other insulin resistances, e.g. as seen in polycystic ovary syndrome (PCOS). Moreover, novel approaches employing knock-out mice demonstrated that PPARgamma seems to play a key role in placental and fetal development. This review describes recent insights into the role of PPARs in human reproduction with specific reference to infertility, placental maturation and fetal development as well as disturbed pregnancy. Further, we highlight the current knowledge on synthetic ligands to PPARgamma used as a treatment in women with PCOS.
Collapse
Affiliation(s)
- Bettina Toth
- Department of Obstetrics and Gynecology, Ludwig-Maximilians-University, Grosshadern, Munich, Germany.
| | | | | | | | | | | |
Collapse
|
218
|
Szanto A, Rőszer T. Nuclear receptors in macrophages: A link between metabolism and inflammation. FEBS Lett 2007; 582:106-16. [DOI: 10.1016/j.febslet.2007.11.020] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2007] [Accepted: 11/06/2007] [Indexed: 10/22/2022]
|
219
|
Neuroprotection with pioglitazone against LPS insult on dopaminergic neurons may be associated with its inhibition of NF-kappaB and JNK activation and suppression of COX-2 activity. J Neuroimmunol 2007; 192:89-98. [PMID: 17976742 DOI: 10.1016/j.jneuroim.2007.09.029] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2007] [Revised: 08/22/2007] [Accepted: 09/17/2007] [Indexed: 01/29/2023]
Abstract
Increasing evidence links neuroinflammation to Parkinson's disease. Microglia are mediators of neuroinflammation. Overactivation of microglia contributes to the release of cyclooxygenase 2 and prostaglandin E(2) during neuronal insults. We have previously shown that pioglitazone, a peroxisome proliferator-activated receptor gamma agonist, inhibits microglia activation, reduces proinflammatory factors, and protects dopaminergic neurons. Here, we demonstrated that pioglitazone protects dopaminergic neurons by inhibiting abnormal microglia activation, interfering with phosphorylation of jun N-terminal kinase and nuclear factor kappa-B, and by suppressing cyclooxygenase 2 expression and the subsequent prostaglandin E(2) synthesis. Therefore, the anti-inflammatory properties of pioglitazone may be useful for ameliorating the progression of Parkinson's disease.
Collapse
|
220
|
Ravaux L, Denoyelle C, Monne C, Limon I, Raymondjean M, El Hadri K. Inhibition of interleukin-1beta-induced group IIA secretory phospholipase A2 expression by peroxisome proliferator-activated receptors (PPARs) in rat vascular smooth muscle cells: cooperation between PPARbeta and the proto-oncogene BCL-6. Mol Cell Biol 2007; 27:8374-87. [PMID: 17908795 PMCID: PMC2169168 DOI: 10.1128/mcb.00623-07] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The inflammation that occurs during atherosclerosis is characterized by the release of large amounts of group IIA secretory phospholipase A2 (sPLA2-IIA). This study was designed to define the function of the three peroxisome proliferator-activated receptors (PPARs) on sPLA2 expression in vascular smooth muscle cells (VSMCs). We found that PPAR ligands decreased sPLA2-IIA activity and inhibited mRNA accumulation under inflammatory conditions. Furthermore, interleukin-1beta-induced sPLA2-IIA promoter activity was inhibited by the three PPAR ligands and in a similar way when cells were cotransfected with PPARalpha, PPARbeta, or PPARgamma, plus retinoid X receptor alpha (RXRalpha). Our study revealed that the regulation of sPLA2-IIA gene transcription by PPARalpha/RXR and PPARgamma/RXR heterodimers requires an interaction with a PPAR response element (PPRE) of the sPLA2-IIA promoter. In contrast, PPARbeta operates through a PPRE-independent mechanism. In addition, we demonstrated that VSMCs expressed the transcriptional repressor BCL-6. Overexpression of BCL-6 markedly reduced sPLA2-IIA promoter activity in VSMCs, while a dominant negative form of BCL-6 abrogated sPLA2 repression by PPARbeta. The PPARbeta agonist induced a BCL-6 binding to the sPLA2 promoter in VSMCs under inflammatory conditions. The knockdown of BCL-6 by short interfering RNA abolished the inhibitory effect of the PPARbeta ligand on sPLA2 activity and prostaglandin E2 release. Thus, the inhibition of sPLA2-IIA activity by PPARbeta agonists may provide a promising approach to impacting the initiation and progression of atherosclerosis.
Collapse
MESH Headings
- Animals
- Cattle
- Enzyme Induction/drug effects
- Gene Expression Regulation, Enzymologic/drug effects
- Group II Phospholipases A2/biosynthesis
- Group II Phospholipases A2/genetics
- Interleukin-1beta/pharmacology
- Ligands
- Male
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/enzymology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/enzymology
- PPAR-beta/metabolism
- Protein Binding/drug effects
- Proto-Oncogene Proteins c-bcl-6/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA, Small Interfering/metabolism
- Rats
- Rats, Wistar
- Repressor Proteins/metabolism
- Response Elements
- Retinoid X Receptors/metabolism
- Sequence Deletion
Collapse
Affiliation(s)
- Lucas Ravaux
- UMR Physiologie et Physiopathologie, Université Pierre et Marie Curie, CNRS, 7 quai Saint-Bernard, 75252 Paris, France
| | | | | | | | | | | |
Collapse
|
221
|
Liu Y, Yuan Z, Liu Y, Zhang J, Yin P, Wang D, Wang Y, Kishimoto C, Ma A. PPARgamma gene C161T substitution is associated with reduced risk of coronary artery disease and decreased proinflammatory cytokine expression. Am Heart J 2007; 154:718-24. [PMID: 17892998 DOI: 10.1016/j.ahj.2007.06.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2005] [Accepted: 06/15/2007] [Indexed: 12/21/2022]
Abstract
BACKGROUND Peroxisome proliferator-activated receptor gamma (PPARgamma) is a transcription factor implicated in the expression of proinflammatory cytokines in atheroma-associated cells, and the expression of proinflammatory cytokines, such as tumor necrosis factor alpha (TNF-alpha) and matrix metalloproteinases (MMPs), represents a critical step in atherogenesis and atherosclerosis. In this study, we test the hypothesis of whether a polymorphism corresponding to C161T substitution in exon 6 of the PPARgamma gene may affect the expression of proinflammatory cytokines and the onset of coronary artery diseases (CADs) in a Chinese population. METHODS We have studied distribution of the PPARgamma C161T substitution at exon 6 in 247 patients with CAD and 214 patients with chest pain syndrome. The plasma concentrations of MMP-9 and TNF-alpha were measured by enzyme-linked immunosorbent assay. RESULTS The results showed that the frequencies of the CC, CT, and TT genotypes were 61.9%, 34.0%, and 4.1% in CAD, and 51.4%, 45.3%, and 3.3% in chest pain syndrome, respectively. There was a significant association between the PPARgamma C161T polymorphism and CAD. The T allele carriers (CT + TT) had significantly reduced CAD risk compared with the CC homozygotes (odds ratio 0.547, 95% CI 0.327-0.831, P = .012) in a logistic regression model after controlling known independent factors for CAD. The CC homozygotes also had increased MMP-9 and TNF-alpha levels compared with T allele carriers. Moreover, the CC homozygotes were more susceptible to acute coronary syndrome than T allele carriers. CONCLUSIONS PPARgamma C161T polymorphism was associated with angiographically documented CAD in a Chinese population. The T allele of the PPARgamma gene might have a protective effect on the progression of CAD and reduce the onset of acute coronary syndrome, which might be associated with the decreased expression of MMP-9 and TNF-alpha in patients with CAD.
Collapse
Affiliation(s)
- Yu Liu
- Department of Cardiovascular Medicine, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | | | | | | | | | | | | | | | | |
Collapse
|
222
|
Schmitz G, Grandl M. Role of redox regulation and lipid rafts in macrophages during Ox-LDL-mediated foam cell formation. Antioxid Redox Signal 2007; 9:1499-518. [PMID: 17600463 DOI: 10.1089/ars.2007.1663] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Hyperlipidemias and small dense LDLs in patients with high-triglyceride low-HDL syndromes lead to a prolonged half life of apoB-containing particles. This is associated with reactive oxygen species (ROS) activation and leads to formation of oxidized LDL (Ox-LDL). Generators of ROS in macrophages (MACs) include myeloperoxidase (MPO)-mediated respiratory burst and raft-associated NADPH-oxidase. The intracellular oxidant milieu is involved in cellular signaling pathways, like ion-transport systems, protein phosphorylation, and gene expression. Lipid oxidation through ROS can amplify foam cell formation through Ox-LDL uptake, leading to formation of ceramide (Cer)-rich lipid membrane microdomains, and is associated with expansion of the lysosomal compartment and an upregulation of ABCA1 and other genes of the AP3 secretory pathway. Ox-LDL may also affect cell-surface turnover of Cer-backbone sphingolipids and apoE-mediated uptake by LRP-family members. In contrast, HDL-mediated lipid efflux causes disruption of lipid membrane microdomains and prevents foam cell formation. Oxidation of HDL through MPO leads to a failure of lipid efflux and enhancement of MAC loading. Therefore, lipid rafts and oxidation processes are important in regulation of MAC foam cell formation and atherosclerosis, and the balance between oxidant and antioxidant intracellular systems is critically important for efficient MAC function.
Collapse
Affiliation(s)
- Gerd Schmitz
- Institute for Clinical Chemistry and Laboratory Medicine, University of Regensburg, Franz-Josef-Strauss-Allee 11, 93042 Regensburg, Germany.
| | | |
Collapse
|
223
|
Khanolkar MP, Morris RHK, Thomas AW, Bolusani H, Roberts AW, Geen J, Jackson SK, Evans LM. Rosiglitazone produces a greater reduction in circulating platelet activity compared with gliclazide in patients with type 2 diabetes mellitus--an effect probably mediated by direct platelet PPARgamma activation. Atherosclerosis 2007; 197:718-24. [PMID: 17765245 DOI: 10.1016/j.atherosclerosis.2007.07.020] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2007] [Revised: 06/26/2007] [Accepted: 07/11/2007] [Indexed: 11/24/2022]
Abstract
AIMS Type 2 diabetes mellitus (T2DM) is associated with enhanced platelet activation. We conducted a randomised double-blind study to compare the effects of combination metformin and rosiglitazone or metformin and gliclazide therapy on platelet function in persons with T2DM. METHODS Fifty subjects on metformin monotherapy received either rosiglitazone 4 mg or gliclazide 80 mg. HbA1c, HOMA-R, markers of platelet activation, inflammation, endothelial activation and oxidative stress were measured at baseline and after 24 weeks of treatment. Separate in vitro platelet function studies were conducted on platelets pre-incubated with rosiglitazone and gliclazide. RESULTS A significantly greater reduction in platelet aggregation was observed in the rosiglitazone treated group compared to gliclazide. HbA1c and markers of endothelial activation were reduced to a similar extent in both groups. A significant reduction in HOMA-R, markers of inflammation and oxidative stress was only observed with rosiglitazone. Reduction in platelet aggregation with rosiglitazone correlated with reduction in oxidative stress. In the in vitro study, rosiglitazone produced significantly greater reduction in platelet aggregation compared with gliclazide. CONCLUSION Greater reduction in platelet activity observed with rosiglitazone may be related to reduced oxidative stress and a possible direct PPARgamma mediated effect on platelet function.
Collapse
Affiliation(s)
- M P Khanolkar
- Llandough Hospital Diabetes Center, Department of Diabetes, Penarth, Cardiff, UK.
| | | | | | | | | | | | | | | |
Collapse
|
224
|
Ahmed W, Ziouzenkova O, Brown J, Devchand P, Francis S, Kadakia M, Kanda T, Orasanu G, Sharlach M, Zandbergen F, Plutzky J. PPARs and their metabolic modulation: new mechanisms for transcriptional regulation? J Intern Med 2007; 262:184-98. [PMID: 17645586 DOI: 10.1111/j.1365-2796.2007.01825.x] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Peroxisome proliferator-activated receptors (PPARs) as ligand-activated nuclear receptors involved in the transcriptional regulation of lipid metabolism, energy balance, inflammation, and atherosclerosis are at the intersection of key pathways involved in the pathogenesis of diabetes and cardiovascular disease. Synthetic PPAR agonists like fibrates (PPAR-alpha) and thiazolidinediones (PPAR-gamma) are in therapeutic use to treat dyslipidaemia and diabetes. Despite strong encouraging in vitro, animal model, and human surrogate marker studies with these agents, recent prospective clinical cardiovascular trials have yielded mixed results, perhaps explained by concomitant drug use, study design, or a lack of efficacy of these agents on cardiovascular disease (independent of their current metabolic indications). The use of PPAR agents has also been limited by untoward effects. An alternative strategy to PPAR therapeutics is better understanding PPAR biology, the nature of natural PPAR agonists, and how these molecules are generated. Such insight might also provide valuable information about pathways that protect against the metabolic problems for which PPAR agents are currently indicated. This approach underscores the important distinction between the effects of synthetic PPAR agonists and the unequivocal biologic role of PPARs as key transcriptional regulators of metabolic and inflammatory pathways relevant to diabetes and atherosclerosis.
Collapse
Affiliation(s)
- W Ahmed
- Cardiovascular Division, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
225
|
Amoruso A, Bardelli C, Gunella G, Fresu LG, Ferrero V, Brunelleschi S. Quantification of PPAR-gamma protein in monocyte/macrophages from healthy smokers and non-smokers: a possible direct effect of nicotine. Life Sci 2007; 81:906-15. [PMID: 17765929 DOI: 10.1016/j.lfs.2007.07.017] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2007] [Revised: 07/09/2007] [Accepted: 07/18/2007] [Indexed: 11/26/2022]
Abstract
Previous observations demonstrated that Peroxisome Proliferator-Activated Receptor-gamma (PPAR-gamma), a key regulator of adipocyte differentiation, is expressed in a large variety of cells, including cells of the monocyte/macrophage lineage. This study was aimed to quantify both the constitutive and ligand-induced PPAR-gamma expression in monocytes and monocyte-derived macrophages (MDM) isolated from healthy smokers and non-smokers, and to evaluate the possible direct effect of nicotine. PPAR-gamma protein was detected by Western blot and quantification was performed by calculating the ratio between PPAR-gamma and beta-actin protein expression. Cytokine release was measured with enzyme-linked immunoassay kits. Constitutive PPAR-gamma protein was detected in human monocytes and its expression was up-regulated along with differentiation to MDM. The endogenous ligand 15-deoxy-delta(12,14)-prostaglandin J(2) and the synthetic agonist ciglitazone enhanced PPAR-gamma expression, the former being effective also at low micromolar concentrations. Both agonists significantly inhibited the basal secretion of pro-inflammatory cytokines (e.g., TNF-alpha, IL-6), ciglitazone being more potent. Monocytes and MDM from healthy smokers presented a significantly enhanced (4-fold and 2.5-fold, respectively) constitutive PPAR-gamma expression, as compared to those from healthy non-smokers. However, ligand-induced PPAR-gamma expression and inhibition of cytokine secretion were similar in healthy smokers and non-smokers. Nicotine dose-dependently enhanced PPAR-gamma expression with a maximum at 10 muM, and inhibited release of pro-inflammatory cytokines; these effects were reversed by alpha-bungarotoxin. Nicotine and PPAR-gamma agonists did not exert synergistic effects. In conclusion, monocytes and MDM from healthy smokers present a constitutively enhanced PPAR-gamma expression; this effect is reproduced, to some extent, by nicotine in vitro.
Collapse
Affiliation(s)
- Angela Amoruso
- Department of Medical Sciences, School of Medicine, University of Piemonte Orientale A. Avogadro, Via Solaroli, 17 - 28100 Novara, Italy
| | | | | | | | | | | |
Collapse
|
226
|
Radhika A, Jacob SS, Sudhakaran PR. Influence of oxidatively modified LDL on monocyte-macrophage differentiation. Mol Cell Biochem 2007; 305:133-43. [PMID: 17660956 DOI: 10.1007/s11010-007-9536-0] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2007] [Accepted: 06/21/2007] [Indexed: 10/23/2022]
Abstract
Transendothelial migration of peripheral blood mononuclear cells (PBMCs) and their subsequent interaction with the subendothelial matrix lead to their differentiation to macrophages (mphis). To study whether preexposure of monocytes in circulation to modified proteins influences their differentiation to mphis, an in vitro model system using isolated PBMC in culture was used. The effect of modified proteins such as oxidatively modified LDL (ox-LDL), acetylated and non-enzymatically glycated-BSA (NEG-BSA) on the differentiation process was studied by monitoring the upregulation of mphi specific functions such as endocytosis, production of matrix metalloproteinases (MMPs), expression of surface antigen, activity of beta-glucuronidase and down regulation of monocyte specific myeloperoxidase activity. Rate of endocytosis, production of MMPs and beta-glucuronidase activity were significantly greater in cells treated with modified proteins irrespective of the nature of modification. Both CuSO4 ox-LDL and HOCl ox-LDL increased the rate of expression of the mphi specific functions. FACS analysis showed that the rate of upregulation of mphi specific CD71 and down regulation of monocyte specific CD14 were high in cells supplemented with modified proteins. Studies using PPARgamma antagonist and agonist suggest its involvement in CuSO4 ox-LDL induced monocyte-macrophage (mo-mphi) differentiation whereas the expression of macrophage specific functions in cells exposed to other modified proteins was independent of PPARgamma. PBMC isolated from hypercholesterolemic rabbits in culture expressed mphi specific functions at a faster rate compared to normal controls indicating that these observations are relevant in vivo. These results indicate that preexposure of monocytes to modified proteins promote their differentiation to mphis and may serve as a feed forward type control for clearing modified proteins.
Collapse
Affiliation(s)
- Achuthan Radhika
- Department of Biochemistry, University of Kerala, Kariavattom, Trivandrum, 695 581, India
| | | | | |
Collapse
|
227
|
Sertznig P, Seifert M, Tilgen W, Reichrath J. Present concepts and future outlook: function of peroxisome proliferator-activated receptors (PPARs) for pathogenesis, progression, and therapy of cancer. J Cell Physiol 2007; 212:1-12. [PMID: 17443682 DOI: 10.1002/jcp.20998] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Peroxisome proliferator-activated receptors (PPARs) are members of the nuclear receptor superfamily of transcriptional regulators that regulate lipid, glucose, and amino acid metabolism. In recent studies it also has been shown that these receptors are implicated in tumor progression, cellular differentiation, and apoptosis and modulation of their function is therefore considered as a potential target for cancer prevention and treatment. PPAR ligands and other agents influencing PPAR signalling pathways have been shown to reveal chemopreventive potential by mediating tumor suppressive activities in a variety of human cancers and could represent a potential novel strategy to inhibit tumor carcinogenesis and progression. This review summarizes the currently available data on the roles of PPARs in relation to the processes of cell differentiation and carcinogenesis as well as their role as promising future therapeutic targets.
Collapse
Affiliation(s)
- P Sertznig
- Department of Dermatology, The Saarland University Hospital, Homburg/Saar, Germany
| | | | | | | |
Collapse
|
228
|
Voutsadakis IA. Peroxisome proliferator-activated receptor γ (PPARγ) and colorectal carcinogenesis. J Cancer Res Clin Oncol 2007; 133:917-28. [PMID: 17659359 DOI: 10.1007/s00432-007-0277-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2006] [Accepted: 06/28/2007] [Indexed: 01/09/2023]
Abstract
Peroxisome proliferator-activated receptor gamma (PPARgamma) a member of the nuclear transcription factor superfamily is playing a role in colon carcinogenesis. Although not all in vivo models agree, PPARgamma seems to have suppressive effects in this process favoring apoptosis and inhibiting the cell cycle by inducing expression of apoptosis and senescence proteins. With the recent discovery that anti-diabetic class of drugs thiazolidinediones act through activation of PPARgamma, interest in this transcription factor has increased as it can now be pharmacologically activated in order to obtain tumor suppression. In addition, thiazolidinediones and other PPARgamma agonists possess PPARgamma-independent anti-tumor effects. Although PPARgamma agonists may not by themselves be capable to induce clinical tumor regression, their combination with chemotherapy drugs or other targeted therapies is worth pursuing in the treatment of colorectal carcinoma.
Collapse
Affiliation(s)
- Ioannis A Voutsadakis
- Division of Medical Oncology, University Hospital of Larissa, Larissa 41110, Greece.
| |
Collapse
|
229
|
Spears M, McSharry C, Thomson NC. Peroxisome proliferator-activated receptor-gamma agonists as potential anti-inflammatory agents in asthma and chronic obstructive pulmonary disease. Clin Exp Allergy 2007; 36:1494-504. [PMID: 17177672 DOI: 10.1111/j.1365-2222.2006.02604.x] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Inhaled corticosteroids are the most effective therapy for chronic persistent asthma and have a role in the treatment of chronic obstructive pulmonary disease (COPD). However, corticosteroids have reduced efficacy in some patients with asthma and fail to halt the progressive deterioration in lung function characteristic of COPD. Additional or alternative drug treatments to corticosteroids are required to improve control of inflammation in patients with therapy resistant airway disease. Peroxisome proliferator-activated receptor-gamma (PPAR-gamma) agonists have displayed potent anti-inflammatory properties in experimental models of asthma and other airway diseases and as a result have the potential to become an additional treatment for asthma and COPD. We review the evidence from these experimental models and their applicability to asthma and COPD and the requirements for future clinical and experimental research.
Collapse
Affiliation(s)
- M Spears
- Department of Respiratory Medicine, Division of Immunology, Infection and Inflammation, University of Glasgow, Glasgow, UK
| | | | | |
Collapse
|
230
|
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: 6.4] [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
|
231
|
Ivashchenko CY, Duan SZ, Usher MG, Mortensen RM. PPAR-gamma knockout in pancreatic epithelial cells abolishes the inhibitory effect of rosiglitazone on caerulein-induced acute pancreatitis. Am J Physiol Gastrointest Liver Physiol 2007; 293:G319-26. [PMID: 17463185 DOI: 10.1152/ajpgi.00056.2007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Peroxisome proliferator-activated receptor-gamma (PPAR-gamma) agonists, such as the thiazolidinediones (TZDs), decrease acute inflammation in both pancreatic cell lines and mouse models of acute pancreatitis. Since PPAR-gamma agonists have been shown to exert some of their actions independent of PPAR-gamma, the role of PPAR-gamma in pancreatic inflammation has not been directly tested. Furthermore, the differential role of PPAR-gamma in endodermal derivatives (acini, ductal cells, and islets) as opposed to the endothelial or inflammatory cells is unknown. To determine whether the effects of a TZD, rosiglitazone, on caerulein-induced acute pancreatitis are dependent on PPAR-gamma in the endodermal derivatives, we created a cell-type specific knock out of PPAR-gamma in pancreatic acini, ducts, and islets. PPAR-gamma knockout animals show a greater response in some inflammatory genes after caerulein challenge. The anti-inflammatory effect of rosiglitazone on edema, macrophage infiltration, and expression of the proinflammatory cytokines is significantly decreased in pancreata of the knockout animals compared with control animals. However, rosiglitazone retains its effect in the lungs of the pancreatic-specific PPAR-gamma knockout animals, likely due to direct anti-inflammatory effect on lung parenchyma. These data show that the PPAR-gamma in the pancreatic epithelia and islets is important in suppressing inflammation and is required for the anti-inflammatory effects of TZDs in acute pancreatitis.
Collapse
Affiliation(s)
- C Y Ivashchenko
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, 1150 W. Medical Center Drive, Ann Arbor, MI 48109, USA
| | | | | | | |
Collapse
|
232
|
Seto SW, Lam TY, Leung GPH, Au ALS, Ngai SM, Chan SW, Kwan YW. Comparison of vascular relaxation, lipolysis and glucose uptake by peroxisome proliferator-activated receptor-gamma activation in +db/+m and +db/+db mice. Eur J Pharmacol 2007; 572:40-8. [PMID: 17603034 DOI: 10.1016/j.ejphar.2007.05.070] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2007] [Revised: 05/22/2007] [Accepted: 05/23/2007] [Indexed: 11/21/2022]
Abstract
In this study, we determined the in vitro effect of peroxisome proliferator-activated receptor-gamma (PPAR-gamma) activation on the aortic relaxation, lipolysis and insulin-induced [(3)H]-glucose uptake of the abdominal (omental) adipocytes of the non-diabetic (+db/+m) and obese/diabetic (+db/+db) mice. The expression of PPAR-gamma (mRNA and protein) in aorta and adipose tissues was evaluated and compared. Cumulative application of ciglitazone, pioglitazone and troglitazone (PPAR-gamma agonists) caused a concentration-dependent aortic relaxation (sensitive to 2-chloro-5-nitro-N-phenylbenzamide (GW9662) (1 microM, a selective PPAR-gamma antagonist) and N(omega)-nitro-l-arginine methyl ester (l-NAME) (20 microM, a nitric oxide synthase inhibitor)) with a maximum relaxation of approximately 30% (3 microM) in +db/+m mice, whereas no relaxation was observed in +db/+db mice. All PPAR-gamma agonists examined did not alter the basal lipolysis of both species, but forskolin caused a concentration-dependent lipolysis, with a greater magnitude observed in +db/+m mice. Insulin (0.1 and 1 microM) caused an enhancement of [(3)H]-glucose uptake into adipocytes with a greater magnitude in +db/+m mice. In contrast, none of the PPAR-gamma agonists tested (0.1, 1 and 10 microM) altered the basal and the insulin (0.1 microM)-induced [(3)H]-glucose uptake into adipocytes of both species. In addition, there was no difference in PPAR-gamma expression (mRNA and protein) in the aorta and adipose tissues between the species. In conclusion, our results demonstrate that PPAR-gamma is present in the abdominal (omental) adipose tissue and thoracic aorta. An acute activation of PPAR-gamma produced a small ( approximately 30%) aortic relaxation (nitric oxide/endothelium-dependent) of +db/+m mice. However, all PPAR-gamma agonists examined have no acute effect on lipolysis and the insulin-induced glucose uptake into adipocytes of both +db/+m and +db/+db mice.
Collapse
MESH Headings
- Abdominal Fat/metabolism
- Adipocytes/drug effects
- Adipocytes/metabolism
- Animals
- Aorta, Thoracic/drug effects
- Aorta, Thoracic/physiopathology
- Carbohydrate Metabolism/drug effects
- Chromans/pharmacology
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/physiopathology
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/physiopathology
- Female
- Glucose/metabolism
- Hypoglycemic Agents/pharmacology
- In Vitro Techniques
- Insulin/pharmacology
- Lipid Metabolism
- Lipolysis/drug effects
- Mice
- Mice, Inbred C57BL
- Mice, Mutant Strains
- Obesity/metabolism
- Obesity/physiopathology
- PPAR gamma/agonists
- PPAR gamma/biosynthesis
- Pioglitazone
- RNA, Messenger/agonists
- RNA, Messenger/biosynthesis
- Thiazolidinediones/pharmacology
- Troglitazone
- Vasodilation/drug effects
Collapse
Affiliation(s)
- Sai Wang Seto
- Department of Pharmacology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong
| | | | | | | | | | | | | |
Collapse
|
233
|
Marx N, Walcher D. Vascular effects of PPARgamma activators - from bench to bedside. Prog Lipid Res 2007; 46:283-96. [PMID: 17637478 DOI: 10.1016/j.plipres.2007.05.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2007] [Revised: 05/21/2007] [Accepted: 05/29/2007] [Indexed: 10/23/2022]
Abstract
Activation of the nuclear transcription factor peroxisome proliferator-activated receptor-gamma (PPARgamma) plays an important role in adipogenesis, insulin resistance, and glucose homeostasis. Activators of PPARgamma include the anti-diabetic thiazolidinediones (TZDs), drugs that are in clinical use to treat patients with type 2 diabetes mellitus. Experimental as well as clinical data gathered over the last decade suggest that PPARgamma activators may exert direct modulatory function in the vasculature in addition to their metabolic effects. PPARgamma is expressed in all vascular cells, where its activators exhibit anti-inflammatory and anti-atherogenic properties, suggesting that PPARgamma ligands could influence important processes in all phases of atherogenesis. Results from clinical trials demonstrated that TZDs reduce blood levels of inflammatory biomarkers of arteriosclerosis, improve endothelial function, and directly influence lesion morphology and plaque stability, underscoring that PPAR activators may have direct effects in the vasculature in humans. This review will focus on the vascular effects of PPARgamma activators and summarize the current knowledge of their modulatory function on atherogenesis and vascular disease.
Collapse
Affiliation(s)
- Nikolaus Marx
- Department of Internal Medicine II - Cardiology, University of Ulm, Robert-Koch-Str. 8, D-89081 Ulm, Germany.
| | | |
Collapse
|
234
|
Hontecillas R, Bassaganya-Riera J. Peroxisome proliferator-activated receptor gamma is required for regulatory CD4+ T cell-mediated protection against colitis. THE JOURNAL OF IMMUNOLOGY 2007; 178:2940-9. [PMID: 17312139 DOI: 10.4049/jimmunol.178.5.2940] [Citation(s) in RCA: 133] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Peroxisome proliferator-activated receptor (PPAR) gamma activation has been implicated in the prevention of immunoinflammatory disorders; however, the mechanisms of regulation of effector and regulatory CD4+ T cell functions by endogenously activated PPAR-gamma remain unclear. We have used PPAR-gamma-deficient CD4+ T cells obtained from tissue-specific PPAR-gamma null mice (i.e., PPAR-gamma fl/fl; MMTV-Cre+) to investigate the role of endogenous PPAR-gamma on regulatory T cell (Treg) and effector CD4+ T cell function. Overall, we show that the loss of PPAR-gamma results in enhanced Ag-specific proliferation and overproduction of IFN-gamma in response to IL-12. These findings correlate in vivo with enhanced susceptibility of tissue-specific PPAR-gamma null mice to trinitrobenzene sulfonic acid-induced colitis. Furthermore, the transfer of purified PPAR-gamma null CD4+ T cells into SCID recipients results in enteric disease. To test the assertion that the deficiency of PPAR-gamma in Treg impairs their ability to prevent effector T cell-induced colitis, we performed cotransfer studies. These studies demonstrate that PPAR-gamma-expressing, but not PPAR-gamma null Treg, prevent colitis induced by transfer of naive CD4+ T cells into SCID recipients. In line with these findings, the production of IFN-gamma by spleen and mesenteric lymph node-derived CD4+ T cells was down-regulated following transfer of PPAR-gamma-expressing, but not PPAR-gamma null, Treg. In conclusion, our data suggest that endogenous PPAR-gamma activation represents a Treg intrinsic mechanism of down-regulation of effector CD4+ T cell function and prevention of colitis.
Collapse
Affiliation(s)
- Raquel Hontecillas
- Laboratory of Nutritional Immunology and Molecular Nutrition, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA.
| | | |
Collapse
|
235
|
Talbert DR, Allred CD, Zaytseva YY, Kilgore MW. Transactivation of ERalpha by Rosiglitazone induces proliferation in breast cancer cells. Breast Cancer Res Treat 2007; 108:23-33. [PMID: 17453334 DOI: 10.1007/s10549-007-9575-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2007] [Accepted: 03/16/2007] [Indexed: 10/23/2022]
Abstract
In the present study, we demonstrate that Rosiglitazone (Rosi), a thiazolidinedione and PPARgamma agonist, induces ERE (Estrogen Receptor Response Element) reporter activity, pS2 (an endogenous ER gene target) expression, and proliferation of ER positive breast cancer (MCF-7) cells. By performing a dose-response assay, we determined that high concentrations of Rosi inhibit proliferation, while low concentrations of Rosi induce proliferation. Using the anti-estrogen ICI, ER negative breast cancer (MDA-MB-231) cells, and a prostate cancer cell line (22Rv1) deficient in both ERalpha and PPARgamma, we determined that Rosiglitazone-induced ERE reporter activation and proliferation is through an ERalpha dependent mechanism. Rosiglitazone-induced ERE activation is also dependent on activation of the Extracellular Signal-Regulated Kinase-Mitogen Activated Protein Kinase (ERK-MAPK) pathway, since it is inhibited by co-treatment with U0126, a specific inhibitor of this pathway. We also demonstrate that when ERalpha and PPARgamma are both present, they compete for Rosi, inhibiting each others transactivation. To begin to unravel the pharmacological mechanism of Rosi-induced ER activation, sub-maximally effective concentrations of E(2) were used in combination with increasing concentrations of Rosi in luciferase reporter assays. From these assays it appears that E(2) and Rosi both activate ERalpha via similar pharmacological mechanisms. Furthermore sub-maximally effective concentrations of E(2) and Rosi additively increase both ERE reporter activity and MCF-7 cell proliferation. The results of this study may have clinical relevancy for Rosi's use both as an anti-diabetic in post-menopausal women and as an anti-cancer drug in women with ER positive breast cancer.
Collapse
Affiliation(s)
- Dominique R Talbert
- Department of Molecular and Biomedical Pharmacology, University of Kentucky College of Medicine, MS 305, Chandler Medical Center, Lexington, KY 40536, USA
| | | | | | | |
Collapse
|
236
|
Peiris M, Monteith GR, Roberts-Thomson SJ, Cabot PJ. A model of experimental autoimmune encephalomyelitis (EAE) in C57BL/6 mice for the characterisation of intervention therapies. J Neurosci Methods 2007; 163:245-54. [PMID: 17477973 DOI: 10.1016/j.jneumeth.2007.03.013] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2006] [Revised: 03/15/2007] [Accepted: 03/16/2007] [Indexed: 12/31/2022]
Abstract
Multiple sclerosis (MS) and its different forms are studied in the animal model experimental autoimmune encephalomyelitis (EAE). Relapsing-remitting MS, the most common form of the disease can be induced in mice where clinical symptoms fluctuate in severity over time. However, the animal model does not experience periods of recovery where clinical signs are absent, unlike the human disease. We have developed a novel model of relapsing-remitting EAE in C57BL/6 mice immunised with myelin oligodendrocyte glycoprotein (MOG) peptide and Quil A as adjuvant. These animals have relapses that are followed by periods of recovery, during which time the animals do not exhibit illness. Furthermore, administration of the PPARgamma agonist pioglitazone prior to a predicted relapse prevents the expected development of symptoms in a dose-dependent fashion. Immune cell infiltration into white matter of the CNS and decreased production of inflammatory cytokine IFN-gamma in treated animals were also observed. Our model will be a valuable tool in assessing intervention therapies for RR-MS sufferers.
Collapse
MESH Headings
- Adjuvants, Immunologic/pharmacology
- Animals
- Central Nervous System/drug effects
- Central Nervous System/immunology
- Central Nervous System/physiopathology
- Chemotaxis, Leukocyte/drug effects
- Chemotaxis, Leukocyte/immunology
- Disability Evaluation
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Encephalomyelitis, Autoimmune, Experimental/chemically induced
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Female
- Hypoglycemic Agents/pharmacology
- Interferon-gamma/immunology
- Interferon-gamma/metabolism
- Mice
- Mice, Inbred C57BL
- Multiple Sclerosis/drug therapy
- Multiple Sclerosis/immunology
- Multiple Sclerosis/physiopathology
- Myelin Proteins
- Myelin-Associated Glycoprotein/immunology
- Myelin-Associated Glycoprotein/pharmacology
- Myelin-Oligodendrocyte Glycoprotein
- PPAR gamma/antagonists & inhibitors
- PPAR gamma/immunology
- Pioglitazone
- Quillaja Saponins
- Saponins/immunology
- Saponins/pharmacology
- Secondary Prevention
- Thiazolidinediones/pharmacology
- Treatment Outcome
- Vaccination/methods
Collapse
Affiliation(s)
- Madusha Peiris
- The School of Pharmacy, Steele Building, University of Queensland, Brisbane, Queensland 4072, Australia.
| | | | | | | |
Collapse
|
237
|
Michalik L, Auwerx J, Berger JP, Chatterjee VK, Glass CK, Gonzalez FJ, Grimaldi PA, Kadowaki T, Lazar MA, O'Rahilly S, Palmer CNA, Plutzky J, Reddy JK, Spiegelman BM, Staels B, Wahli W. International Union of Pharmacology. LXI. Peroxisome proliferator-activated receptors. Pharmacol Rev 2007; 58:726-41. [PMID: 17132851 DOI: 10.1124/pr.58.4.5] [Citation(s) in RCA: 726] [Impact Index Per Article: 42.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The three peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors of the nuclear hormone receptor superfamily. They share a high degree of structural homology with all members of the superfamily, particularly in the DNA-binding domain and ligand- and cofactor-binding domain. Many cellular and systemic roles have been attributed to these receptors, reaching far beyond the stimulation of peroxisome proliferation in rodents after which they were initially named. PPARs exhibit broad, isotype-specific tissue expression patterns. PPARalpha is expressed at high levels in organs with significant catabolism of fatty acids. PPARbeta/delta has the broadest expression pattern, and the levels of expression in certain tissues depend on the extent of cell proliferation and differentiation. PPARgamma is expressed as two isoforms, of which PPARgamma2 is found at high levels in the adipose tissues, whereas PPARgamma1 has a broader expression pattern. Transcriptional regulation by PPARs requires heterodimerization with the retinoid X receptor (RXR). When activated by a ligand, the dimer modulates transcription via binding to a specific DNA sequence element called a peroxisome proliferator response element (PPRE) in the promoter region of target genes. A wide variety of natural or synthetic compounds was identified as PPAR ligands. Among the synthetic ligands, the lipid-lowering drugs, fibrates, and the insulin sensitizers, thiazolidinediones, are PPARalpha and PPARgamma agonists, respectively, which underscores the important role of PPARs as therapeutic targets. Transcriptional control by PPAR/RXR heterodimers also requires interaction with coregulator complexes. Thus, selective action of PPARs in vivo results from the interplay at a given time point between expression levels of each of the three PPAR and RXR isotypes, affinity for a specific promoter PPRE, and ligand and cofactor availabilities.
Collapse
Affiliation(s)
- Liliane Michalik
- Center for Integrative Genomics, National Research Centre "Frontiers in Genetics," University of Lausanne, Lausanne, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
238
|
Libby P, Plutzky J. Inflammation in diabetes mellitus: role of peroxisome proliferator-activated receptor-alpha and peroxisome proliferator-activated receptor-gamma agonists. Am J Cardiol 2007; 99:27B-40B. [PMID: 17307056 DOI: 10.1016/j.amjcard.2006.11.004] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Patients with type 2 diabetes mellitus and/or the metabolic syndrome have considerable cardiovascular risk. Treatment with 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors (statins) and with antihypertensive and some antihyperglycemic agents reduces this risk, but residual macrovascular morbidity and mortality persist, even in patients assigned to intensive multifactorial intervention programs. Therapeutic strategies that target inflammation and lipid abnormalities not well addressed by statins may offer additional opportunities for improving the prognosis of these patients. Inflammation, a key mechanism of atherogenesis, appears to have particular relevance to diabetic vascular complications, as well as in the development of diabetes itself. Oxidative stress and hyperglycemia also figure among the pathogenic factors that promote cardiovascular complications in patients with the metabolic syndrome and/or diabetes and may augment the ongoing inflammation. Peroxisome proliferator-activated receptor (PPAR)-alpha and PPAR-gamma, members of the nuclear receptor family, form ligand-activated transcription factors that regulate key important metabolic pathways. PPARs have become therapeutic targets through the use of the fibrate class of antidyslipidemic drugs (PPAR-alpha) and the insulin-sensitizing thiazolidinediones (PPAR-gamma). The activation of these PPARs may also suppress inflammation and atherosclerosis. Recent clinical trials (Fenofibrate Intervention and Event Lowering in Diabetes [FIELD], Prospective Pioglitazone Clinical Trial in Macrovascular Events [PROactive]) have considered the impact of these PPAR agonists on cardiovascular disease, with mixed effects that require careful analysis, especially given ongoing trials and additional PPAR agonists in development.
Collapse
Affiliation(s)
- Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA.
| | | |
Collapse
|
239
|
Klotz L, Dani I, Edenhofer F, Nolden L, Evert B, Paul B, Kolanus W, Klockgether T, Knolle P, Diehl L. Peroxisome Proliferator-Activated Receptor γ Control of Dendritic Cell Function Contributes to Development of CD4+ T Cell Anergy. THE JOURNAL OF IMMUNOLOGY 2007; 178:2122-31. [PMID: 17277116 DOI: 10.4049/jimmunol.178.4.2122] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
There is increasing evidence that dendritic cell (DC) immunogenicity is not only positively regulated by ligands of pattern recognition receptors, but also negatively by signals that prevent DC activation and full functional maturation. Depending on their activation status, DCs can induce either immunity or tolerance. In this study, we provide molecular evidence that the transcription factor peroxisome proliferator-activated receptor gamma (PPARgamma) is a negative regulator of DC maturation and function. Sustained PPARgamma activation in murine DCs reduced maturation-induced expression of costimulatory molecules and IL-12, and profoundly inhibited their capacity to prime naive CD4(+) T cells in vitro. Using PPARgamma-deficient DCs, generated by Cre-mediated ablation of the PPARgamma gene, agonist-mediated suppression of maturation-induced functional changes were abrogated. Moreover, absence of PPARgamma increased DC immunogenicity, suggesting a constitutive regulatory function of PPARgamma in DCs. Adoptive transfer of PPARgamma-activated Ag-presenting DCs induced CD4(+) T cell anergy, characterized by impaired differentiation resulting in absent Th1 and Th2 cytokine production and failure of secondary clonal expansion upon restimulation. Collectively, our data support the notion that PPARgamma is an efficient regulator of DC immunogenicity that may be exploited to deliberately target CD4(+) T cell-mediated immune responses.
Collapse
Affiliation(s)
- Luisa Klotz
- Institute of Molecular Medicine and Experimental Immunology, University of Bonn, Bonn, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
240
|
Sharma AM, Staels B. Review: Peroxisome proliferator-activated receptor gamma and adipose tissue--understanding obesity-related changes in regulation of lipid and glucose metabolism. J Clin Endocrinol Metab 2007; 92:386-95. [PMID: 17148564 DOI: 10.1210/jc.2006-1268] [Citation(s) in RCA: 360] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
CONTEXT Adipose tissue is a metabolically dynamic organ, serving as a buffer to control fatty acid flux and a regulator of endocrine function. In obese subjects, and those with type 2 diabetes or the metabolic syndrome, adipose tissue function is altered (i.e. adipocytes display morphological differences alongside aberrant endocrine and metabolic function and low-grade inflammation). EVIDENCE ACQUISITION Articles on the role of peroxisome proliferator-activated receptor gamma (PPARgamma) in adipose tissue of healthy individuals and those with obesity, metabolic syndrome, or type 2 diabetes were sourced using MEDLINE (1990-2006). EVIDENCE SYNTHESIS Articles were assessed to provide a comprehensive overview of how PPARgamma-activating ligands improve adipose tissue function, and how this links to improvements in insulin resistance and the progression to type 2 diabetes and atherosclerosis. CONCLUSIONS PPARgamma is highly expressed in adipose tissue, where its activation with thiazolidinediones alters fat topography and adipocyte phenotype and up-regulates genes involved in fatty acid metabolism and triglyceride storage. Furthermore, PPARgamma activation is associated with potentially beneficial effects on the expression and secretion of a range of factors, including adiponectin, resistin, IL-6, TNFalpha, plasminogen activator inhibitor-1, monocyte chemoattractant protein-1, and angiotensinogen, as well as a reduction in plasma nonesterified fatty acid supply. The effects of PPARgamma also extend to macrophages, where they suppress production of inflammatory mediators. As such, PPARgamma activation appears to have a beneficial effect on the relationship between the macrophage and adipocyte that is distorted in obesity. Thus, PPARgamma-activating ligands improve adipose tissue function and may have a role in preventing progression of insulin resistance to diabetes and endothelial dysfunction to atherosclerosis.
Collapse
Affiliation(s)
- Arya M Sharma
- Canada Research Chair for Cardiovascular Obesity Research and Management, McMaster University, Hamilton General Hospital, 237 Barton Street East, Hamilton, Ontario, Canada L8L 2X2.
| | | |
Collapse
|
241
|
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
|
242
|
Oliveira ACP, Bertollo CM, Rocha LTS, Nascimento EB, Costa KA, Coelho MM. Antinociceptive and antiedematogenic activities of fenofibrate, an agonist of PPAR alpha, and pioglitazone, an agonist of PPAR gamma. Eur J Pharmacol 2007; 561:194-201. [PMID: 17343847 DOI: 10.1016/j.ejphar.2006.12.026] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2006] [Revised: 12/20/2006] [Accepted: 12/21/2006] [Indexed: 10/23/2022]
Abstract
Peroxisome proliferator activated receptors (PPAR) are ligand-regulated transcription factors that control the expression of many genes. The antiinflammatory activity of fibrates, PPARalpha agonists, and thiazolidinediones, PPARgamma agonists, has been demonstrated in many in vitro and a few in vivo studies. In the present study, we evaluated the effect of acute (100 or 300 mg/kg, p.o.) or prolonged (100 or 300 mg/kg day, 7 days, p.o.) treatment with fenofibrate and acute treatment with pioglitazone (doses ranging from 1 to 50 mg/kg, i.p.), PPARalpha and PPARgamma agonists, respectively, on experimental models of nociception and edema, in order to expand the knowledge of their potential antiinflammatory activities. Fenofibrate and pioglitazone did not inhibit the nociceptive response in the hot-plate model and the first phase of formaldehyde induced nociceptive response in mice. However, treatment with pioglitazone and prolonged treatment with fenofibrate inhibited the second phase of this response. Mechanical allodynia induced by carrageenan in rats was inhibited by prolonged treatment with fenofibrate, but not by acute treatment with pioglitazone or fenofibrate. Both drugs inhibited paw edema induced by carrageenan in rats. Fenofibrate did not inhibit mechanical allodynia or paw edema induced by phorbol-12,13-didecanoate (PDD), a protein kinase C activator, in rats. Pioglitazone inhibited paw edema, but not mechanical allodynia, induced by PDD. The results represent the first demonstration of the antinociceptive and antiedematogenic activities of fenofibrate and pioglitazone and give further support to the potential use of PPAR agonists in the treatment of different inflammatory diseases.
Collapse
Affiliation(s)
- Antônio Carlos P Oliveira
- Faculdade de Farmácia, Universidade Federal de Minas Gerais, Avenida Antônio Carlos 6627 31270-91 Belo Horizonte, MG, Brazil
| | | | | | | | | | | |
Collapse
|
243
|
Wild GE, Drozdowski L, Tartaglia C, Clandinin MT, Thomson ABR. Nutritional modulation of the inflammatory response in inflammatory bowel disease- From the molecular to the integrative to the clinical. World J Gastroenterol 2007; 13:1-7. [PMID: 17206749 PMCID: PMC4065867 DOI: 10.3748/wjg.v13.i1.1] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Nutrient deficiencies are common in patients with inflammatory bowel disease (IBD). Both total parenteral and enteral nutrition provide important supportive therapy for IBD patients, but in adults these are not useful for primary therapy. Dietary intervention with omega-3 polyunsaturated fatty acids contained in fish oil may be useful for the care of IBD patients, and recent studies have stressed the role of PPAR on NFκB activity on the potential beneficial effect of dietary lipids on intestinal function.
Collapse
|
244
|
Victor NA, Wanderi EW, Gamboa J, Zhao X, Aronowski J, Deininger K, Lust WD, Landreth GE, Sundararajan S. Altered PPARgamma expression and activation after transient focal ischemia in rats. Eur J Neurosci 2007; 24:1653-63. [PMID: 17004929 DOI: 10.1111/j.1460-9568.2006.05037.x] [Citation(s) in RCA: 114] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Stroke is a devastating disease with limited treatment options. Recently, we found that the peroxisome proliferator-activated receptor-gamma (PPARgamma) agonists troglitazone and pioglitazone reduce injury and inflammation in a rat model of transient cerebral ischemia. The mechanism of this protection is unclear, as these agents can act through PPAR-gamma activation or through PPAR-gamma-independent mechanisms. Therefore, we examined PPAR-gamma expression, DNA binding and transcriptional activity following stroke. In addition, we used a PPAR-gamma antagonist, T0070907, to determine the role of PPAR-gamma during ischemia. Using immunohistochemical techniques and real-time PCR, we found low levels of PPAR-gamma mRNA and PPAR-gamma immunoreactivity in nonischemic brain; however, PPAR-gamma expression dramatically increased in ischemic neurons, peaking 24 h following middle cerebral artery occlusion. Interestingly, we found that in both vehicle- and agonist-treated brains, DNA binding was reduced in the ischemic hemisphere relative to the contralateral hemisphere. Expression of a PPAR-gamma target gene, lipoprotein lipase, was also reduced in ischemic relative to nonischemic brain. Both DNA binding and lipoprotein lipase expression were increased by the addition of the PPAR-gamma agonist rosiglitazone. Finally, we found that rosiglitazone-mediated protection after stroke was reversed by the PPAR-gamma antagonist T0070907. Interestingly, infarction size was also increased by T0070907 in the absence of PPAR-gamma agonist, suggesting that endogenous PPAR-gamma ligands may mitigate the effects of cerebral ischemia.
Collapse
Affiliation(s)
- N A Victor
- Department of Neurology, Case Western Reserve University, 11100 Euclid Ave., Cleveland, Ohio 44106, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
245
|
Joner M, Farb A, Cheng Q, Finn AV, Acampado E, Burke AP, Skorija K, Creighton W, Kolodgie FD, Gold HK, Virmani R. Pioglitazone Inhibits In-Stent Restenosis in Atherosclerotic Rabbits by Targeting Transforming Growth Factor-β and MCP-1. Arterioscler Thromb Vasc Biol 2007; 27:182-9. [PMID: 17068283 DOI: 10.1161/01.atv.0000251021.28725.e8] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective—
Although emerging data from preclinical and clinical studies suggests a reduction of in-stent restenosis with peroxisome proliferator-activated receptor (PPAR)-γ agonists, the reduction of neointimal growth via anti-inflammatory mechanisms has not been explored.
Methods and Results—
Hypercholesterolemic New Zealand White rabbits (n=45) received bilateral balloon-expandable stents implanted into atherosclerotic iliac arteries. Animals were randomized to oral pioglitazone 3 (low dose) or 10 mg/kg per day (high dose) started on the day of stent implantation; control rabbits received placebo. Tissue harvest was performed 28 days after stenting, and stented segments underwent histology, morphometry, immunostaining for macrophages, and scanning electron microscopy. In selected animals, stented arterial segments were placed in organoid culture for 48 hours, and the conditioned media was assayed for 23 different cytokines. There was a 21% reduction in neointimal area for high-dose pioglitazone treated versus placebo rabbits (
P
<0.005), which was associated with a significant reduction of neointimal macrophages. Analysis of conditioned media revealed an 82% and 74% reduction in the release of monocyte chemoattractant protein-1 (MCP-1) (
P
<0.007) and transforming growth factor (TGF)-β1 (
P
<0.01), respectively, in stented segments from animals treated with 10 mg/kg per day pioglitazone versus placebo.
Conclusions—
Oral pioglitazone suppresses in-stent neointimal growth by limiting local inflammatory pathways and may be useful as an adjunctive therapy in patients undergoing percutaneous interventions.
Collapse
|
246
|
Takahashi T, Ono H, Ono Y, Ishimitsu T, Matsuoka H. Combination Therapy With Telmisartan and Spironolactone Alleviates L-NAME Exacerbated Nephrosclerosis With an Increase in PPAR-.GAMMA. and Decrease in TGF-.BETA.1. Int Heart J 2007; 48:637-47. [DOI: 10.1536/ihj.48.637] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Toshiaki Takahashi
- Department of Hypertension and Cardiorenal Disease, Dokkyo Medical University
| | - Hidehiko Ono
- Department of Hypertension and Cardiorenal Disease, Dokkyo Medical University
| | - Yuko Ono
- Department of Pathology, Dokkyo Medical University
| | - Toshihiko Ishimitsu
- Department of Hypertension and Cardiorenal Disease, Dokkyo Medical University
| | - Hiroaki Matsuoka
- Department of Hypertension and Cardiorenal Disease, Dokkyo Medical University
| |
Collapse
|
247
|
Billiet L, Furman C, Larigauderie G, Copin C, Page S, Fruchart JC, Brand K, Rouis M. Enhanced VDUP-1 gene expression by PPARγ agonist induces apoptosis in human macrophage. J Cell Physiol 2007; 214:183-91. [PMID: 17579352 DOI: 10.1002/jcp.21179] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The fate and phenotype of lesion macrophages is regulated by cellular oxidative stress. Thioredoxin-1 (Trx-1) plays a major role in the regulation of cellular redox balance, with resultant effects on gene expression and cellular responses including cell growth and death. Trx-1 activity is inhibited by interaction with vitamin D-upregulated protein-1 (VDUP-1). Peroxisome proliferator-activated receptor gamma (PPARgamma) is expressed by human monocyte-derived macrophages (HMDM) and PPARgamma agonism has been reported to decrease expression of inflammatory genes and to promote apoptosis of these cells. To determine whether VDUP-1 may be involved in regulating the effects of PPARgamma agonists in macrophages, we investigated the effect of a synthetic PPARgamma agonist (GW929) on the expression of VDUP-1 in HMDM. GW929 concentration-dependently increased HMDM expression of VDUP-1 (mRNA and protein). Transfection of different fragments of the VDUP-1 promoter as well as gel shift analysis revealed the presence of functional PPARgamma response elements (PPRE) in the promoter. Under conditions in which PPAR agonism altered levels of VDUP-1, caspase-3 activity, and macrophage apoptosis were also elevated. The results suggest that PPARgamma activation stimulates apoptosis in human macrophages by altering the cellular redox balance via regulation of VDUP-1.
Collapse
|
248
|
Ríos-Vázquez R, Marzoa-Rivas R, Gil-Ortega I, Kaski JC. Peroxisome proliferator-activated receptor-gamma agonists for management and prevention of vascular disease in patients with and without diabetes mellitus. Am J Cardiovasc Drugs 2006; 6:231-42. [PMID: 16913824 DOI: 10.2165/00129784-200606040-00003] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Inflammation is known to have a pathogenic role in atherosclerosis and the genesis of acute coronary syndromes. The peroxisome proliferator-activated receptor (PPAR)-gamma, which is expressed in many constituent cells of atheromatous plaques, inhibits the activation of several proinflammatory genes responsible for atheromatous plaque development and maturation. Agonists of this receptor, such as rosiglitazone and pioglitazone, are currently available for the treatment of type 2 diabetes mellitus, and several lines of evidence have shown that these drugs have antiatherogenic effects. Insulin resistance is associated with inflammation and has a key role in atherogenesis. The antiatherogenic and insulin sensitizing effects of the thiazolidinediones in patients with type 2 diabetes mellitus may be associated with this action. However, in recent years there has been growing evidence that the antiatherogenic effects of PPAR-gamma agonists are not confined to patients with diabetes mellitus. PPAR-gamma agonists have been shown to downregulate the expression of endothelial activation markers, reduce circulating platelet activity, improve flow-mediated dilatation and attenuate atheromatous plaque progression in patients without diabetes mellitus. These effects of PPAR-gamma agonists appear to result from both insulin sensitization and a direct modulation of transcriptional activity in the vessel wall. This review summarizes the current understanding of the role of PPAR-gamma agonists in atherogenesis and discusses their potential role in the treatment of coronary artery disease in patients with type 2 diabetes mellitus and in nondiabetic patients.
Collapse
Affiliation(s)
- Ramón Ríos-Vázquez
- Cardiovascular Biology Research Centre, Division of Cardiac and Vascular Sciences, University of London, London, UK
| | | | | | | |
Collapse
|
249
|
Henry JR, Li Y, Warshawsky AM, Brozinick JT, Hawkins ED, Misener EA, Briere DA, Montrose-Rafizadeh C, Zink RW, Yumibe NP, Ajamie RT, Wilken B, Devanarayan V. Tetrahydroisoquinoline PPARγ agonists: Design of novel, highly selective non-TZD antihyperglycemic agents. Bioorg Med Chem Lett 2006; 16:6293-7. [PMID: 17005393 DOI: 10.1016/j.bmcl.2006.09.028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2006] [Revised: 09/01/2006] [Accepted: 09/07/2006] [Indexed: 11/25/2022]
Abstract
Novel tetrahydroisoquinolines have been developed as potent PPAR ligands. Evaluation of these compounds in PPARgamma responsive models of type 2 diabetes is described.
Collapse
Affiliation(s)
- James R Henry
- Lilly Research Laboratories, A Division of Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN 46285, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
250
|
Pakala R, Dilcher C, Baffour R, Hellinga D, Seabron R, Joner M, Kolodgie F, Virmani R, Waksman R. Peroxisome Proliferator-Activated Receptor γ Ligand Pioglitazone Alters Neointimal Composition in a Balloon-Denuded and Radiated Hypercholesterolemic Rabbit. J Cardiovasc Pharmacol 2006; 48:299-305. [PMID: 17204909 DOI: 10.1097/01.fjc.0000249891.40714.2a] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Peroxisome proliferator-activated receptor (PPAR)-gamma activation suppresses inflammatory response, monocyte recruitment, and vascular cell proliferation. Because inflammation, deregulated growth, and migration of monocytes and vascular smooth muscle cells (VSMC) play important roles in the development of neointima, we tested the effect of pioglitazone, a high-affinity ligand, for PPAR-gamma on neointima formation in the iliac arteries of a balloon-denuded and radiated hypercholesterolemic rabbit. Rabbits were fed a 1.0% cholesterol diet for 7 days followed by denudation of endothelial layer and continued on a 0.15% cholesterol diet. On day 32, animals were divided into 2 groups. One group received a 0.15% cholesterol diet (n = 7) and the other group received a 0.15% cholesterol diet supplemented with 400 mg of pioglitazone per kilogram. On day 35, the balloon-denuded area was radiated. Four weeks after radiation, animals were sacrificed and arterial segments were processed for morphometry and immunohistochemistry. Data analysis showed that the pioglitazone group had smaller neointima (0.85 +/- 0.36 vs. 1.41 +/- 0.56, P < 0.05), with more cells positive for VSMC (23.07 +/- 6.16 vs. 18.33 +/- 5.19, P = 0.04), less for monocytes (16.01 +/- 5.33 vs. 21.29 +/- 4.33, P < 0.05), and fewer cells expressing metalloproteinase (MMP)-1 and MMP-9 (3.69 +/- 0.47 vs. 4.82 +/- 0.93, P < 0.05 and 3.24 +/- 0.71 vs. 4.29 +/- 0.74, P < 0.05, respectively). Pioglitazone reduced neointimal area and modified its composition in a balloon-denuded and radiated hypercholesterolemic rabbit model.
Collapse
Affiliation(s)
- Rajbabu Pakala
- Cardiovascular Research Institute, Washington Hospital Center, Washington, DC, USA
| | | | | | | | | | | | | | | | | |
Collapse
|