1
|
Kong Y, Gao Y, Lan D, Zhang Y, Zhan R, Liu M, Zhu Z, Zeng G, Huang Q. Trans-repression of NFκB pathway mediated by PPARγ improves vascular endothelium insulin resistance. J Cell Mol Med 2018; 23:216-226. [PMID: 30398029 PMCID: PMC6307800 DOI: 10.1111/jcmm.13913] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 07/23/2018] [Accepted: 08/20/2018] [Indexed: 02/07/2023] Open
Abstract
Previous study has shown that thiazolidinediones (TZDs) improved endothelium insulin resistance (IR) induced by high glucose concentration (HG)/hyperglycaemia through a PPARγ‐dependent‐NFκB trans‐repression mechanism. However, it is unclear, whether changes in PPARγ expression affect the endothelium IR and what the underlying mechanism is. In the present study, we aimed to address this issue. HG‐treated human umbilical vascular endothelial cells (HUVEC) were transfected by either PPARγ‐overexpressing (Ad‐PPARγ) or PPARγ‐shRNA‐containing (Ad‐PPARγ‐shRNA) adenoviral vectors. Likewise, the rats fed by high‐fat diet (HFD) were infected by intravenous administration of Ad‐PPARγ or Ad‐PPARγ‐shRNA. The levels of nitric oxide (NO), endothelin‐1 (ET‐1) and cytokines (TNFα, IL‐6, sICAM‐1 and sVCAM‐1) and the expression levels of PPARγ, eNOS, AKT, p‐AKT, IKKα/β and p‐IKKα/β and IκBα were examined; and the interaction between PPARγ and NFκB‐P65 as well as vascular function were evaluated. Our present results showed that overexpression of PPARγ notably increased the levels of NO, eNOS, p‐AKT and IκBα as well as the interaction of PPARγ and NFκB‐P65, and decreased the levels of ET‐1, p‐IKKα/β, TNFα, IL‐6, sICAM‐1 and sVCAM‐1. In contrast, down‐expression of PPARγ displayed the opposite effects. The results demonstrate that the overexpression of PPARγ improves while the down‐expression worsens the endothelium IR via a PPARγ‐mediated NFκB trans‐repression dependent manner. The findings suggest PPARγ is a potential therapeutic target for diabetic vascular complications.
Collapse
Affiliation(s)
- Ying Kong
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, China.,Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang, Jiangxi, China
| | - Yan Gao
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, China.,Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang, Jiangxi, China
| | - Dongyi Lan
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Ying Zhang
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, China.,Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang, Jiangxi, China
| | - Rixin Zhan
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, China.,Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang, Jiangxi, China
| | - Meiqi Liu
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, China.,Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang, Jiangxi, China
| | - Zhouan Zhu
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Guohua Zeng
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, China.,Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang, Jiangxi, China
| | - Qiren Huang
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, China.,Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
2
|
Osman I, Fairaq A, Segar L. Pioglitazone Attenuates Injury-Induced Neointima Formation in Mouse Femoral Artery Partially through the Activation of AMP-Activated Protein Kinase. Pharmacology 2017; 100:64-73. [PMID: 28482342 DOI: 10.1159/000471769] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 03/16/2017] [Indexed: 12/15/2022]
Abstract
BACKGROUND/AIMS Pioglitazone (PIO), an antidiabetic drug, has been shown to attenuate vascular smooth muscle cell (VSMC) proliferation, which is a major event in atherosclerosis and restenosis after angioplasty. Till date, the likely contributory role of AMP-activated protein kinase (AMPK) toward PIO inhibition of VSMC proliferation has not been examined in vivo. This study is aimed at determining whether pharmacological inhibition of AMPK would prevent the inhibitory effect of PIO on neointima formation in a mouse model of arterial injury. METHODS Male CJ57BL/6J mice were subjected to femoral artery injury using guidewire. PIO (20 mg/kg/day) was administered orally 1 day before surgery and for 3 weeks until sacrifice in the absence or presence of compound C (an AMPK inhibitor). Injured femoral arteries were used for morphometric analysis of neointima formation. Aortic tissue lysates were used for immunoblot analysis of phosphorylated AMPK. RESULTS PIO treatment resulted in a significant decrease in intima-to-media ratio by ∼50.3% (p < 0.05, compared with vehicle control; n = 6), which was accompanied by enhanced phosphorylation of AMPK by ∼85% in the vessel wall. Compound C treatment led to a marked reduction in PIO-mediated inhibition of neointima formation. CONCLUSION PIO attenuates injury-induced neointima formation, in part, through the activation of AMPK.
Collapse
Affiliation(s)
- Islam Osman
- Center for Pharmacy and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta University, Augusta, GA, USA
| | | | | |
Collapse
|
3
|
Kubota T, Kubota N, Sato H, Inoue M, Kumagai H, Iwamura T, Takamoto I, Kobayashi T, Moroi M, Terauchi Y, Tobe K, Ueki K, Kadowaki T. Pioglitazone Ameliorates Smooth Muscle Cell Proliferation in Cuff-Induced Neointimal Formation by Both Adiponectin-Dependent and -Independent Pathways. Sci Rep 2016; 6:34707. [PMID: 27703271 PMCID: PMC5050439 DOI: 10.1038/srep34707] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 08/15/2016] [Indexed: 01/11/2023] Open
Abstract
The aim of this study is to elucidate to what degree adiponectin is involved in TZD-mediated amelioration of neointimal formation. We investigated the effect of 3- or 8-weeks' pioglitazone on cuff-induced neointimal formation in adiponectin-deficient (APN-KO) and wild-type (WT) mice. Pioglitazone for 3 weeks reduced neointimal formation in the WT mice with upregulation of the plasma adiponectin levels, but failed to reduce neointimal formation in the APN-KO mice, suggesting that pioglitazone suppressed neointimal formation by adiponectin-dependent mechanisms. Pioglitazone for 3 weeks suppressed vascular smooth muscle cell (VSMC) proliferation and increased AdipoR2 expression in the WT mice. In vitro, globular adiponectin activated AMPK through both AdipoR1 and AdipoR2, resulting in the inhibition of VSMC proliferation. Interestingly, 8-weeks' pioglitazone was reduced neointimal formation in APN-KO mice to degree similar to that seen in the WT mice, suggesting that pioglitazone can also suppress neointimal formation via a mechanism independent of adiponectin. Pioglitazone for 8 weeks completely abrogated the increased VSMC proliferation, along with a reduction of cyclin B1 and cyclin D1 expressions and cardiovascular risk profile in the APN-KO mice. In vitro, pioglitazone suppressed these expressions, leading to inhibition of VSMC proliferation. Pioglitazone suppresses neointimal formation via both adiponectin-dependent and adiponectin-independent mechanisms.
Collapse
Affiliation(s)
- Tetsuya Kubota
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, University of Tokyo, Tokyo 113-8655, Japan.,Laboratory for Metabolic Homeostasis, RIKEN Center for Integrative Medical Sciences, Kanagawa, 230-0045, Japan.,Department of Clinical Nutrition, National Institute of Health and Nutrition, Tokyo 162-8636, Japan.,Division of Cardiovascular Medicine, Toho University Ohashi Medical Center, Tokyo 153-8515, Japan
| | - Naoto Kubota
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, University of Tokyo, Tokyo 113-8655, Japan.,Laboratory for Metabolic Homeostasis, RIKEN Center for Integrative Medical Sciences, Kanagawa, 230-0045, Japan.,Department of Clinical Nutrition, National Institute of Health and Nutrition, Tokyo 162-8636, Japan.,Department of Clinical Nutrition Therapy, University of Tokyo, Tokyo 113-8655, Japan
| | - Hiroyuki Sato
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, University of Tokyo, Tokyo 113-8655, Japan
| | - Mariko Inoue
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, University of Tokyo, Tokyo 113-8655, Japan.,Department of Clinical Nutrition, National Institute of Health and Nutrition, Tokyo 162-8636, Japan
| | - Hiroki Kumagai
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, University of Tokyo, Tokyo 113-8655, Japan
| | - Tomokatsu Iwamura
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, University of Tokyo, Tokyo 113-8655, Japan
| | - Iseki Takamoto
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, University of Tokyo, Tokyo 113-8655, Japan
| | - Tsuneo Kobayashi
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University, Tokyo 142-8501, Japan
| | - Masao Moroi
- Division of Cardiovascular Medicine, Toho University Ohashi Medical Center, Tokyo 153-8515, Japan
| | - Yasuo Terauchi
- Department of Diabetes and Endocrinology, Yokohama City University, School of Medicine, Kanagawa 236-0004, Japan
| | - Kazuyuki Tobe
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, Toyama, 930-0194, Japan
| | - Kohjiro Ueki
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, University of Tokyo, Tokyo 113-8655, Japan
| | - Takashi Kadowaki
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, University of Tokyo, Tokyo 113-8655, Japan
| |
Collapse
|
4
|
Goltsman I, Khoury EE, Winaver J, Abassi Z. Does Thiazolidinedione therapy exacerbate fluid retention in congestive heart failure? Pharmacol Ther 2016; 168:75-97. [PMID: 27598860 DOI: 10.1016/j.pharmthera.2016.09.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The ever-growing global burden of congestive heart failure (CHF) and type 2 diabetes mellitus (T2DM) as well as their co-existence necessitate that anti-diabetic pharmacotherapy will modulate the cardiovascular risk inherent to T2DM while complying with the accompanying restrictions imposed by CHF. The thiazolidinedione (TZD) family of peroxisome proliferator-activated receptor γ (PPARγ) agonists initially provided a promising therapeutic option in T2DM owing to anti-diabetic efficacy combined with pleiotropic beneficial cardiovascular effects. However, the utility of TZDs in T2DM has declined in the past decade, largely due to concomitant adverse effects of fluid retention and edema formation attributed to salt-retaining effects of PPARγ activation on the nephron. Presumably, the latter effects are potentially deleterious in the context of pre-existing fluid retention in CHF. However, despite a considerable body of evidence on mechanisms responsible for TZD-induced fluid retention suggesting that this class of drugs is rightfully prohibited from use in CHF patients, there is a paucity of experimental and clinical studies that investigate the effects of TZDs on salt and water homeostasis in the CHF setting. In an attempt to elucidate whether TZDs actually exacerbate the pre-existing fluid retention in CHF, our review summarizes the pathophysiology of fluid retention in CHF. Moreover, we thoroughly review the available data on TZD-induced fluid retention and proposed mechanisms in animals and patients. Finally, we will present recent studies challenging the common notion that TZDs worsen renal salt and water retention in CHF.
Collapse
Affiliation(s)
- Ilia Goltsman
- Department of Physiology, Biophysics and Systems Biology, The Bruce Rappaport, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Emad E Khoury
- Department of Physiology, Biophysics and Systems Biology, The Bruce Rappaport, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Joseph Winaver
- Department of Physiology, Biophysics and Systems Biology, The Bruce Rappaport, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Zaid Abassi
- Department of Physiology, Biophysics and Systems Biology, The Bruce Rappaport, Rappaport Faculty of Medicine, Technion, Haifa, Israel; Department of Laboratory Medicine, Rambam Human Health Care Campus, Haifa, Israel.
| |
Collapse
|
5
|
Desouza CV, Murthy SN, Diez J, Dunne B, Matta AS, Fonseca VA, McNamara DB. Differential Effects of Peroxisome Proliferator Activator Receptor-α and γ Ligands on Intimal Hyperplasia After Balloon Catheter-Induced Vascular Injury in Zucker Rats. J Cardiovasc Pharmacol Ther 2016; 8:297-305. [PMID: 14740079 DOI: 10.1177/107424840300800407] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Background: Patients with type 2 diabetes mellitus have a higher rate of restenosis following angioplasty. Peroxisome proliferator activator receptor-x (PPAR) and y ligands such as fenofibrate and rosiglitazone, respectively, have been shown to have protective effects on the vessel wall. We studied the effect of fenofibrate and rosiglitazone on intimal hyperplasia in the Zucker rat, a model for insulin resistance and type 2 diabetes mellitus, following balloon catheter-induced injury. Methods and Results: Three groups of 13-week-old female fatty Zucker rats were administered an aqueous suspension of either 3 mg/kg/d rosiglitazone (n = 7) or 150 mg/kg/d fenofibrate (n = 6) by gavage, or served as controls (n = 9). In addition, two groups of 13-week-old female lean Zucker rats were either administered 3 mg/kg/d rosiglitazone (n = 6) or served as controls (n = 6). Carotid balloon injury was induced 1 week after the drugs were started. The drug administration was continued for 3 weeks. A 2-mm balloon catheter was introduced through the femoral artery to the left carotid. The balloon was inflated to 4 atmospheres for 20 seconds and then was deflated to 2 atmospheres and dragged down to the aorta. The rats were killed 3 weeks after the injury. The carotid intima/media ratio was calculated. Intimal hyperplasia after carotid balloon-induced injury in the fatty Zucker rats was significantly reduced in the group treated with rosiglitazone (0.18 ± 0.29) compared with the untreated group (0.97 ± 0.13; P < .01). Plasma glucose, triglyceride, and insulin levels were elevated, indicative of the presence of insulin resistance; rosiglitazone treatment significantly reduced insulin and triglyceride levels without decreasing glucose. Rosiglitazone treatment also reduced, but to a lesser extent, the intimal hyperplasia in the lean Zucker rats (0.57 ± 0.10 vs 1.06 ± 0.12 treated and untreated, respectively; P < .01); however, it had no effect on insulin, triglyceride, or glucose levels in this group. The intimal hyperplasia in the fatty Zucker rats treated with fenofibrate was not reduced compared with controls (0.84 ± 0.26 vs 0.97 ± 0.13, respectively); fenofibrate reduced insulin and triglyceride, but not glucose levels, in these animals. Conclusions: The PPAR-y ligand rosiglitazone, but not the PPAR-x ligand fenofibrate, decreases intimal hyperplasia following balloon injury in both fatty and lean Zucker rats. This effect of the PPAR-y ligand was independent of glycemia, insulin, and lipid levels, and was more pronounced in insulin-resistant rats.
Collapse
Affiliation(s)
- Cyrus V Desouza
- Department of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | | | | | | | | | | | | |
Collapse
|
6
|
Osman I, Segar L. Pioglitazone, a PPARγ agonist, attenuates PDGF-induced vascular smooth muscle cell proliferation through AMPK-dependent and AMPK-independent inhibition of mTOR/p70S6K and ERK signaling. Biochem Pharmacol 2015; 101:54-70. [PMID: 26643070 DOI: 10.1016/j.bcp.2015.11.026] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 11/25/2015] [Indexed: 02/08/2023]
Abstract
Pioglitazone (PIO), a PPARγ agonist that improves glycemic control in type 2 diabetes through its insulin-sensitizing action, has been shown to exhibit beneficial effects in the vessel wall. For instance, it inhibits vascular smooth muscle cell (VSMC) proliferation, a major event in atherosclerosis and restenosis after angioplasty. Although PPARγ-dependent and PPARγ-independent mechanisms have been attributed to its vasoprotective effects, the signaling events associated with PIO action in VSMCs are not fully understood. To date, the likely intermediary role of AMP-activated protein kinase (AMPK) toward PIO inhibition of VSMC proliferation has not been examined. Using human aortic VSMCs, the present study demonstrates that PIO activates AMPK in a sustained manner thereby contributing in part to inhibition of key proliferative signaling events. In particular, PIO at 30μM concentration activates AMPK to induce raptor phosphorylation, which diminishes PDGF-induced mTOR activity as evidenced by decreased phosphorylation of p70S6K, 4E-BP1, and S6 and increased accumulation of p27(kip1), a cell cycle inhibitor. In addition, PIO inhibits the basal phosphorylation of ERK in VSMCs. Downregulation of endogenous AMPK by target-specific siRNA reveals an AMPK-independent effect for PIO inhibition of ERK, which contributes in part to diminutions in cyclin D1 expression and Rb phosphorylation and the suppression of VSMC proliferation. Furthermore, AMPK-dependent inhibition of mTOR/p70S6K and AMPK-independent inhibition of ERK signaling occur regardless of PPARγ expression/activation in VSMCs as evidenced by gene silencing and pharmacological inhibition of PPARγ. Strategies that utilize nanoparticle-mediated PIO delivery at the lesion site may limit restenosis after angioplasty without inducing PPARγ-mediated systemic adverse effects.
Collapse
Affiliation(s)
- Islam Osman
- Center for Pharmacy and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, GA, USA; Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - Lakshman Segar
- Center for Pharmacy and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, GA, USA; Charlie Norwood VA Medical Center, Augusta, GA, USA; Vascular Biology Center, Department of Pharmacology and Toxicology, Georgia Regents University, Augusta, GA, USA; Department of Medicine, Pennsylvania State University College of Medicine, Hershey, PA, USA.
| |
Collapse
|
7
|
Transcriptional Control of Vascular Smooth Muscle Cell Proliferation by Peroxisome Proliferator-Activated Receptor-gamma: Therapeutic Implications for Cardiovascular Diseases. PPAR Res 2011; 2008:429123. [PMID: 18288288 PMCID: PMC2225465 DOI: 10.1155/2008/429123] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2007] [Accepted: 10/24/2007] [Indexed: 12/14/2022] Open
Abstract
Proliferation of vascular smooth muscle cells (SMCs) is a critical process for the development of atherosclerosis and complications of procedures used to treat atherosclerotic diseases, including postangioplasty restenosis, vein graft failure, and transplant vasculopathy. Peroxisome proliferator-activated receptor (PPAR) gamma is a member of the nuclear hormone receptor superfamily and the molecular target for the thiazolidinediones (TZD), used clinically to treat insulin resistance in patients with type 2 diabetes. In addition to their efficacy to improve insulin sensitivity, TZD exert a broad spectrum of pleiotropic beneficial effects on vascular gene expression programs. In SMCs, PPARgamma is prominently upregulated during neointima formation and suppresses the proliferative response to injury of the arterial wall. Among the molecular target genes regulated by PPARgamma in SMCs are genes encoding proteins involved in the regulation of cell-cycle progression, cellular senescence, and apoptosis. This inhibition of SMC proliferation is likely to contribute to the prevention of atherosclerosis and postangioplasty restenosis observed in animal models and proof-of-concept clinical studies. This review will summarize the transcriptional target genes regulated by PPARgamma in SMCs and outline the therapeutic implications of PPARgamma activation for the treatment and prevention of atherosclerosis and its complications.
Collapse
|
8
|
Chen Z, Hasegawa T, Tanaka A, Okita Y, Okada K. Pioglitazone preserves vein graft integrity in a rat aortic interposition model. J Thorac Cardiovasc Surg 2010; 140:408-416.e1. [DOI: 10.1016/j.jtcvs.2009.11.067] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2009] [Revised: 11/10/2009] [Accepted: 11/27/2009] [Indexed: 02/07/2023]
|
9
|
Yamasaki Y, Katakami N, Furukado S, Kitagawa K, Nagatsuka K, Kashiwagi A, Daida H, Kawamori R, Kaku K. Long-term effects of pioglitazone on carotid atherosclerosis in Japanese patients with type 2 diabetes without a recent history of macrovascular morbidity. J Atheroscler Thromb 2010; 17:1132-40. [PMID: 20686324 DOI: 10.5551/jat.4663] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
AIM No previous studies have evaluated the long-term anti-atherosclerotic effects of pioglitazone in Asian patients with type 2 diabetes. Therefore, the present study investigated the protective effects of pioglitazone on the progression of carotid intima-media thickness (IMT), an established surrogate marker of cardiovascular events in Japanese type 2 diabetic patients without a recent history of cardiovascular morbidity. METHODS This 2.5-4-year, randomized, open-label, blinded endpoint study was conducted in 6 centers across Japan. Patients received pioglitazone with or without other oral glucose-lowering drugs (excluding another thiazolidinedione) (n=89) or oral glucose-lowering drugs, excluding thiazolidinediones (n=97). Treatment was adjusted to achieve HbA(1c) <6.5%. The primary endpoints of the study were the absolute changes from the baseline to final visit in max- and mean-IMT in the average of bilateral common carotid arteries. RESULTS Pioglitazone induced carotid IMT regression compared to baseline measurements (from 1.060 ± 0.2368 to 0.992 ± 0.1921 mm; p=0.0042 in max-IMT and from 0.839 ± 0.1873 to 0.780 ± 0.1571 mm; p=0.0019 in mean-IMT). Although the between-group difference did not reach statistical significance, the regression of carotid IMT values was greater in the pioglitazone-treatment group than in the non-pioglitazone group, (max-IMT: -0.069 ± 0.2199 mm vs -0.031 ± 0.2327 mm, respectively; p=NS, mean-IMT: -0.058 ± 0.1718 mm vs -0.043 ± 0.1644 mm, respectively; p=NS). CONCLUSIONS Pioglitazone induced and maintained the long-term regression of carotid IMT in Japanese type 2 diabetic patients. This suggests that pioglitazone may inhibit the progression of atherosclerosis in this patient group. Further studies are required to verify these findings.
Collapse
Affiliation(s)
- Yoshimitsu Yamasaki
- Department of Internal Medicine and Therapeutics, Osaka University Graduate School of Medicine, Osaka, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Dorafshar AH, Moodley K, Khoe M, Lyon C, Bryer-Ash M. Pioglitazone improves superoxide dismutase mediated vascular reactivity in the obese Zucker rat. Diab Vasc Dis Res 2010; 7:20-7. [PMID: 20368229 DOI: 10.1177/1479164109341688] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To test the hypothesis that the thiazolidinedione agent, pioglitazone, mediates its chronic BP lowering action via improving vascular reactivity. METHODS AND RESULTS Lean (Fa/fa) and obese (fa/fa) Zucker rats were treated with or without pioglitazone (20 mg/ kg/day) for 4 weeks (n=8 animals per group). Pioglitazone treatment was associated with a significant improvement in oral glucose tolerance in the obese animals (p<0.05 compared with untreated obese). Pioglitazone prevented the development of hypertension seen in obese untreated rats (SBP 126+/-1 versus 138+/-1 mmHg; p<0.0001). Aortic ring preparations from pioglitazone-treated obese rats showed improved relaxation responsiveness (ED(50) 0.28 versus 1.15 U/ ml, p<0.001) to SOD, a NO potentiator, compared with untreated obese animals. CONCLUSIONS SOD-mediated vasorelaxation may contribute to the chronic antihypertensive effect and/or the improvement in insulin sensitivity following pioglitazone treatment.
Collapse
Affiliation(s)
- Amir H Dorafshar
- Department of Medicine, University of California Los Angeles, California, USA
| | | | | | | | | |
Collapse
|
11
|
Sarafidis PA, Nilsson PM. The effects of thiazolidinediones on blood pressure levels – A systematic review. Blood Press 2009; 15:135-50. [PMID: 16864155 DOI: 10.1080/08037050600853720] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Insulin resistance has been proposed to be the underlying disorder of the so-called metabolic or insulin resistance syndrome, which represents the clustering in the same individual of several cardiovascular risk factors, such as type 2 diabetes mellitus, hypertension, abdominal obesity, elevated triglycerides and low high-density lipoprotein-cholesterol. As far as the connection of insulin resistance and compensatory hyperinsulinaemia with hypertension is concerned, a number of mechanisms possibly linking these disturbances have been described, such as activation of sympathetic nervous system, enhancement of renal sodium reabsorption, or impairment of endothelium-dependent vasodilatation. Thiazolidinediones (TZDs) constitute a class of oral antihyperglycaemic agents that act by decreasing insulin resistance, and apart from their action on glycaemic control, they have been also reported to exert beneficial effects on other parameters of the metabolic syndrome. In particular, during recent years a considerable number of animal and human studies have shown that the use of TZDs was associated with usually small but significant reductions of blood pressure (BP) levels. Since a possible beneficial action of these compounds on BP could be of particular value for patients with the metabolic syndrome, this review aimed to summarize and evaluate the literature data in the field, derived either from studies that just examined BP levels among other parameters or from studies that were specifically designed to determine the effect of a TZD on BP.
Collapse
Affiliation(s)
- Pantelis A Sarafidis
- 1st Department of Medicine, AHEPA University Hospital, Aristotle University, Thessaloniki, Greece.
| | | |
Collapse
|
12
|
Villacorta L, Schopfer FJ, Zhang J, Freeman BA, Chen YE. PPARgamma and its ligands: therapeutic implications in cardiovascular disease. Clin Sci (Lond) 2009; 116:205-18. [PMID: 19118492 PMCID: PMC2677102 DOI: 10.1042/cs20080195] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The relevance of PPARgamma (peroxisome-proliferator-activated receptor gamma) as an important therapeutic target for the treatment of diabetes arises from its hypoglycaemic effects in diabetic patients and also from the critical role in the regulation of cardiovascular functions. From a clinical perspective, differences between current FDA (Food and Drug Administration)-approved PPARgamma drugs have been observed in terms of atherosclerosis and cardiac and stroke events. The adverse effects of PPARgamma-specific treatments that hamper their cardiovascular protective roles, affirm the strong need to evaluate the efficacy of the current drugs. Therefore active research is directed towards high-throughput screening and pharmacological testing of a plethora of newly identified natural or synthetic compounds. In the present review we describe the rationale behind drug design strategies targeting PPARgamma, based on current knowledge regarding the effects of such drugs in experimental animal models, as well as in clinical practice. Regarding endogenous PPARgamma ligands, several fatty acid derivatives bind PPARgamma with different affinities, although the physiological relevance of these interactions is not always evident. Recently, NO-derived unsaturated fatty acids were found to be potent agonists of PPARs, with preferential affinity for PPARgamma, compared with oxidized fatty acid derivatives. Nitroalkenes exert important bioactivities of relevance for the cardiovascular system including anti-inflammatory and antiplatelet actions, and are important mediators of vascular tone. A new generation of insulin sensitizers with PPARgamma function for the treatment of diabetes may serve to limit patients from the increased cardiovascular burden of this disease.
Collapse
Affiliation(s)
- Luis Villacorta
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | | | | | | | | |
Collapse
|
13
|
Thethi TK, Singh S, Fonseca V. Insulin Sensitizers and Cardiovascular Disease. Cardiovasc Endocrinol 2008. [DOI: 10.1007/978-1-59745-141-3_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
14
|
Orasanu G, Ziouzenkova O, Devchand PR, Nehra V, Hamdy O, Horton ES, Plutzky J. The peroxisome proliferator-activated receptor-gamma agonist pioglitazone represses inflammation in a peroxisome proliferator-activated receptor-alpha-dependent manner in vitro and in vivo in mice. J Am Coll Cardiol 2008; 52:869-81. [PMID: 18755353 PMCID: PMC2633943 DOI: 10.1016/j.jacc.2008.04.055] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2008] [Revised: 04/22/2008] [Accepted: 04/29/2008] [Indexed: 01/14/2023]
Abstract
OBJECTIVES Our aim was to investigate if the peroxisome proliferator-activated receptor (PPAR)-gamma agonist pioglitazone modulates inflammation through PPARalpha mechanisms. BACKGROUND The thiazolidinediones (TZDs) pioglitazone and rosiglitazone are insulin-sensitizing PPARgamma agonists used to treat type 2 diabetes (T2DM). Despite evidence for TZDs limiting inflammation and atherosclerosis, questions exist regarding differential responses to TZDs. In a double-blinded, placebo-controlled 16-week trial among recently diagnosed T2DM subjects (n = 34), pioglitazone-treated subjects manifested lower triglycerides and lacked the increase in soluble vascular cell adhesion molecules (sVCAM)-1 evident in the placebo group. Previously we reported PPARalpha but not PPARgamma agonists could repress VCAM-1 expression. Since both triglyceride-lowering and VCAM-1 repression characterize PPARalpha activation, we studied pioglitazone's effects via PPARalpha. METHODS Pioglitazone effects on known PPARalpha responses--ligand binding domain activation and PPARalpha target gene expression--were tested in vitro and in vivo, including in wild-type and PPARalpha-deficient cells and mice, and compared with the effects of other PPARgamma (rosiglitazone) and PPARalpha (WY14643) agonists. RESULTS Pioglitazone repressed endothelial TNFalpha-induced VCAM-1 messenger ribonucleic acid expression and promoter activity, and induced hepatic IkappaBalpha in a manner dependent on both pioglitazone exposure and PPARalpha expression. Pioglitazone also activated the PPARalpha ligand binding domain and induced PPARalpha target gene expression, with in vitro effects that were most pronounced in endothelial cells. In vivo, pioglitazone administration modulated sVCAM-1 levels and IkappaBalpha expression in wild-type but not PPARalpha-deficient mice. CONCLUSIONS Pioglitazone regulates inflammatory target genes in hepatic (IkappaBalpha) and endothelial (VCAM-1) settings in a PPARalpha-dependent manner. These data offer novel mechanisms that may underlie distinct TZD responses.
Collapse
Affiliation(s)
- Gabriela Orasanu
- Cardiovascular Division, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Ouliana Ziouzenkova
- Cardiovascular Division, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Pallavi R. Devchand
- Cardiovascular Division, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Vedika Nehra
- Cardiovascular Division, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Osama Hamdy
- Clinical Research Center, Joslin Diabetes Center, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Edward S. Horton
- Clinical Research Center, Joslin Diabetes Center, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Jorge Plutzky
- Cardiovascular Division, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
15
|
Ruan X, Zheng F, Guan Y. PPARs and the kidney in metabolic syndrome. Am J Physiol Renal Physiol 2008; 294:F1032-47. [PMID: 18234957 DOI: 10.1152/ajprenal.00152.2007] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The metabolic syndrome (MetS) is defined by a set of metabolic risk factors, including insulin resistance, central obesity, dyslipidemia, hyperglycemia, and hypertension for type 2 diabetes and cardiovascular disease. Although both retrospective and prospective clinical studies have revealed that MetS is associated with chronic renal disease, even with a nondiabetic cause, the cellular and molecular mechanisms in this association remain largely uncharacterized. Recently, increasing evidence suggests that peroxisome proliferator-activated receptors (PPARs), a subgroup of the nuclear hormone receptor superfamily of ligand-activated transcription factors, may play an important role in the pathogenesis of MetS. All three members of the PPAR nuclear receptor subfamily, PPARalpha, -beta/delta, and -gamma, are critical in regulating insulin sensitivity, adipogenesis, lipid metabolism, inflammation, and blood pressure. PPARs have also been implicated in many renal pathophysiological conditions, including diabetic nephropathy and glomerulosclerosis. Ligands for PPARs such as hypolipidemic PPARalpha activators, and antidiabetic thiazolidinedione PPARgamma agonists affect not only diverse aspects of MetS but also renal disease progression. Emerging data suggest that PPARs may be potential therapeutic targets for MetS and its related renal complications. This review focuses on current knowledge of the role of PPARs in MetS and discusses the potential therapeutic utility of PPAR modulators in the treatment of kidney diseases associated with MetS.
Collapse
Affiliation(s)
- Xiongzhong Ruan
- Center for Nephrology, University College of London, London, United Kingdom
| | | | | |
Collapse
|
16
|
Onuta G, Rienstra H, de Boer JF, Boer MW, Roks AJM, Klatter FA, Uges DRA, Navis G, Rozing J, Hillebrands JL. Rosiglitazone attenuates transplant arteriosclerosis after allogeneic aorta transplantation in rats. Transplantation 2007; 84:517-26. [PMID: 17713437 DOI: 10.1097/01.tp.0000276983.91892.99] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Transplant arteriosclerosis is a leading cause of chronic transplant dysfunction and is characterized by occlusive neointima formation in intragraft arteries. Development of transplant arteriosclerosis is refractory to conventional immunosuppressive drugs and adequate therapy is not available. In this study, we determined the efficacy of the synthetic peroxisome proliferator-activated receptor (PPAR)-gamma agonist rosiglitazone to attenuate the development of transplant arteriosclerosis in rat aortic allografts. METHODS Lewis aortic allografts were transplanted into Brown Norway recipient rats. Recipient rats received either approximately 5 mg rosiglitazone/day (starting 1 week before transplantation until the end of the experiment) or were left untreated. Transplant arteriosclerosis was quantified using morphometric analysis. Alloreactivity was measured in vitro using mixed lymphocyte reactions. Regulatory T cell frequency and function were analyzed using flow cytometry and in vitro suppression assays, respectively. Intragraft gene expression was analyzed using real-time polymerase chain reaction. Finally, medial and neointimal vascular smooth muscle cell proliferation was analyzed in vitro. RESULTS Rosiglitazone significantly reduced transplant arteriosclerosis development 8 weeks after transplantation (P<0.01 vs. nontreated). Rosiglitazone reduced T cell alloreactivity which was not mediated through modulation of CD4+CD25+FoxP3+ regulatory T cells. Reduced development of transplant arteriosclerosis coincided with reduced intragraft expression of stromal-derived factor-1alpha and platelet-derived growth factor receptor-beta. Finally, rosiglitazone reduced growth-factor-driven proliferation of both medial and neointimal vascular smooth muscle cells in vitro, which was not mediated through PPARgamma. CONCLUSION PPARgamma agonists may offer a new therapeutic strategy in clinical transplantation to attenuate the development of transplant arteriosclerosis and thereby chronic transplant dysfunction.
Collapse
MESH Headings
- Animals
- Aorta, Abdominal/metabolism
- Aorta, Abdominal/pathology
- Aorta, Abdominal/transplantation
- Arteriosclerosis/pathology
- Arteriosclerosis/prevention & control
- Cell Proliferation/drug effects
- Chemokine CXCL12
- Chemokines, CXC/metabolism
- Forkhead Transcription Factors/metabolism
- Male
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- PPAR gamma/agonists
- Rats
- Rats, Inbred BN
- Rats, Inbred Lew
- Rats, Inbred WF
- Receptor, Platelet-Derived Growth Factor beta/metabolism
- Rosiglitazone
- T-Lymphocytes, Regulatory/metabolism
- T-Lymphocytes, Regulatory/pathology
- Thiazolidinediones/therapeutic use
- Transplantation Immunology/physiology
- Transplantation, Homologous
Collapse
Affiliation(s)
- Geanina Onuta
- Department of Cell Biology, Section of Immunology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Verma S, Szmitko PE. The vascular biology of peroxisome proliferator-activated receptors: modulation of atherosclerosis. Can J Cardiol 2007; 22 Suppl B:12B-17B. [PMID: 16498507 PMCID: PMC2780844 DOI: 10.1016/s0828-282x(06)70981-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Accumulating evidence suggests that peroxisome proliferator-activated receptor (PPAR) agonists possess powerful antiatherosclerotic properties, by both directly affecting the vascular wall and indirectly affecting systemic inflammation and insulin sensitivity. The PPARs are ligand-activated transcription factors, which play a number of important physiological roles in lipid and glucose homeostasis. Activation of PPARgamma appears to exert a vasculoprotective effect by limiting endothelial dysfunction, impairing atherogenesis and preventing restenosis, while simultaneously and favourably modulating adipokine expression and lipid metabolism. Several experimental and clinical studies have demonstrated the potential of the PPAR agonist drug class in terms of treating atherosclerotic disease. In the present review, the vascular biology of PPARs, and how the modulation of these molecular pathways may serve as a therapeutic strategy to prevent atherosclerosis, vascular inflammation and restenosis are discussed.
Collapse
Affiliation(s)
- Subodh Verma
- Division of Cardiac Surgery, St. Michael's Hospital, Toronto, Ontario.
| | | |
Collapse
|
18
|
Abstract
Atherosclerosis and cardiovascular disease are the major causes of morbidity and mortality in patients with diabetes and those with insulin resistance and the metabolic syndrome. Both conditions profoundly accelerate the development of atherosclerosis and increase the morbidity and mortality of cardiovascular events. The question, therefore, is what are the molecular/biochemical mechanisms that underlie the potentiating influence of diabetes, the metabolic syndrome and/or insulin resistance on the development and progression of atherosclerosis? The following review will focus on the molecular mechanism whereby hyperglycaemia and/or hyperinsulinemia either directly or indirectly promote atherosclerosis.
Collapse
Affiliation(s)
- Jane E-B Reusch
- Research Service of Department of Veterans Affairs, and Department of Medicine, University of Colorado Health Sciences Center, Denver, CO, USA
| | | |
Collapse
|
19
|
|
20
|
Blaschke F, Caglayan E, Hsueh WA. Peroxisome proliferator-activated receptor gamma agonists: their role as vasoprotective agents in diabetes. Endocrinol Metab Clin North Am 2006; 35:561-74, ix. [PMID: 16959586 DOI: 10.1016/j.ecl.2006.06.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Both type 1 and type 2 diabetes melitius are associated with increased cardiovascular morbidity, and now affect more than 170 million individuals worldwide. The incidence of type 2 diabetes is growing rapidly and now accounts for 90 to 95 percent of all diabetes cases. Thiazolidinediones (TZDs) a class of insulin sensitizing agents commonly used in the treatment of patients who have type 2 diabetes, improve endothelial dysfunction and exert beneficial effects on lipid profiles by activating the nuclear receptor peroxisome proliferator-activated receptor gamma (PPAR-gamma). In addition, TZDs exhibit antiatherogenic effects, independent of their antidiabetic and lipid-lowering properties, by attenuating proinflammatory processes. The combination of increased insulin sensitivity, improved lipid profile, and reduced inflammation may explain the cardiovascular benefits of this class of drugs
Collapse
Affiliation(s)
- Florian Blaschke
- Department of Endocrinology, Diabetes and Metabolism, Division of Endocrinology, Diabetes and Hypertension, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | | | | |
Collapse
|
21
|
Blaschke F, Spanheimer R, Khan M, Law RE. Vascular effects of TZDs: New implications. Vascul Pharmacol 2006; 45:3-18. [PMID: 16740417 DOI: 10.1016/j.vph.2005.11.009] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2005] [Revised: 11/01/2005] [Accepted: 11/01/2005] [Indexed: 12/18/2022]
Abstract
The incidence of diabetes, now affecting more than 170 million individuals is growing rapidly. Type 2 diabetes, which accounts for 90% of all diabetes cases, is associated with increased cardiovascular morbidity and mortality. Thiazolidinediones (TZDs), used for the treatment of patients with type 2 diabetes improve insulin sensitivity and endothelial dysfunction and exert beneficial effects on the lipid profile by activating the peroxisome proliferator-activated receptor gamma (PPAR-gamma). Moreover, a large body of evidence indicates that TZDs exhibit antiatherogenic effects independent of their antidiabetic and lipid-lowering properties by modulating inflammatory processes. This review will focus on the role of PPAR-gamma agonists in the vessel wall and summarize their effects on C-reactive protein (CRP), plasminogen activator inhibitor type-1 (PAI-1), matrix metalloproteinase-9 (MMP-9), adiponectin and ATP-binding cassette transporter A1 (ABCA1) and their implications for treatment of advanced stages of atherosclerosis, particularly in a setting of type 2 diabetes.
Collapse
Affiliation(s)
- Florian Blaschke
- Division of Endocrinology, Diabetes and Hypertension, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | | | | | | |
Collapse
|
22
|
Chetty VT, Sharma AM. Can PPARgamma agonists have a role in the management of obesity-related hypertension? Vascul Pharmacol 2006; 45:46-53. [PMID: 16713364 DOI: 10.1016/j.vph.2005.11.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2005] [Revised: 11/01/2005] [Accepted: 11/01/2005] [Indexed: 01/11/2023]
Abstract
Peroxisome proliferator-activated receptors (PPAR) are ligand-activated transcription factors belonging to the nuclear hormone receptor superfamily. PPARgamma is the most extensively studied amongst the three subtypes (alpha, delta and gamma). This receptor is a key modulator of lipid and glucose homeostasis and is predominantly expressed in adipose tissue. Expression of PPARgamma is also found in non-adipose tissues including heart, kidney, spleen, and interestingly, in all relevant components of the vasculature: endothelial and smooth muscle cells. These receptors may therefore also play a role in the regulation of vascular tone and blood pressure. Genetic variants of PPARgamma have also been associated with features of the metabolic syndrome, including obesity and increased blood pressure. The discovery of synthetic ligands for PPARgamma, the Thiazolidinediones (TZDs) has greatly enhanced our understanding of their ligand dependent activation and more importantly their role in vascular pathobiology. Approximately 10 years ago, serendipitous animal experiments demonstrated that despite causing sodium retention, the TZDs actually lowered blood pressure. This review will highlight the role of TZDs in various models of hypertension and discuss their potential role in the management of obesity-related hypertension.
Collapse
Affiliation(s)
- Vasudhevan T Chetty
- Michael deGroote School of Medicine, McMaster University, Hamilton, Ontario, Canada.
| | | |
Collapse
|
23
|
Rodríguez-Yáñez M, Castellanos M, Blanco M, Mosquera E, Castillo J. Vascular protection in brain ischemia. Cerebrovasc Dis 2006; 21 Suppl 2:21-9. [PMID: 16651811 DOI: 10.1159/000091700] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Vascular damage occurring after cerebral ischemia may lead to a worse outcome in patients with ischemic stroke, as it facilitates edema formation and hemorrhagic transformation. There are several phases in the development of vascular injury (acute, subacute and chronic) and different mediators act in each one. Therapeutic options to avoid vascular injury must be focused on acting in each phase. However, even though experimental studies have demonstrated the benefit of therapeutic interventions both in the acute and chronic phases of cerebral ischemia, only the chronic phase offers a therapeutic window sufficiently wide enough to provide vascular protection in clinical practice. Several drugs including erythropoietin and HMG-CoA reductase inhibitors (statins), antihypertensive (angiotensin modulators), antibiotics (minocycline) and antihyperglycemic drugs (thiazolidinediones) have been proved to provide vascular protection in patients with ischemic stroke. Anti-inflammatory, antioxidant, and antiapoptotic actions are responsible for the vascular protective effect related to these drugs.
Collapse
Affiliation(s)
- Manuel Rodríguez-Yáñez
- Department of Neurology, Division of Vascular Neurology, Hospital Clínico Universitario, University of Santiago de Compostela, Spain
| | | | | | | | | |
Collapse
|
24
|
Wang K, Zhou Z, Zhang M, Fan L, Forudi F, Zhou X, Qu W, Lincoff AM, Schmidt AM, Topol EJ, Penn MS. Peroxisome proliferator-activated receptor gamma down-regulates receptor for advanced glycation end products and inhibits smooth muscle cell proliferation in a diabetic and nondiabetic rat carotid artery injury model. J Pharmacol Exp Ther 2006; 317:37-43. [PMID: 16368901 DOI: 10.1124/jpet.105.095125] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Diabetes is associated with an increase in circulating advanced glycosylation end products (AGEs) and the increased expression of the receptor for AGEs (RAGE). Inhibition of AGE/RAGE binding through the administration of soluble RAGE (sRAGE) has been shown to decrease neointimal hyperplasia. Peroxisome proliferator-activated receptor gamma (PPARgamma), which inhibits neointimal hyperplasia, has been shown to decrease RAGE expression in cultured endothelial cells. We hypothesized that PPARgamma agonists inhibit neointimal hyperplasia via down-regulation of RAGE in vivo. Pretreatment of rat aortic smooth muscle cells (SMCs) with PPARgamma agonist rosiglitazone significantly down-regulated RAGE expression and inhibited SMC proliferation in response to the RAGE agonist S100/calgranulins. In vivo studies showed that rosiglitazone decreased RAGE expression and SMC proliferation at 7 days following carotid arterial injury in both diabetic and nondiabetic rats. At 21 days following injury, neointimal formation was significantly decreased in both diabetic and nondiabetic animals that received rosiglitazone. To determine whether inhibition of neointimal formation by PPARgamma activation could fully be accounted for by its down-regulation of RAGE, we compared the results obtained in animals treated with sRAGE, PPARgamma activator, and sRAGE + PPARgamma activator. Consistent with PPARgamma working through its effects on RAGE, we found that the addition of PPARgamma activator to sRAGE did not result in any further decrease in neointimal formation. These data demonstrate for the first time that PPARgamma agonists inhibit RAGE expression at sites of arterial injury and suggest that down-regulation of RAGE by the PPARgamma activation inhibits neointimal formation in response to arterial injury.
Collapse
Affiliation(s)
- Kai Wang
- Department of Cardiovascular Medicine, Cleveland Clinic Foundation, OH 44195, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Rodríguez Bernardino A, Cebrián Blanco S, Durán García S. Pioglitazona. Revisión de sus efectos metabólicos y sistémicos. Rev Clin Esp 2005; 205:610-20. [PMID: 16527183 DOI: 10.1016/s0014-2565(05)72655-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Type 2 diabetes mellitus has become a true epidemic and significant growth is expected in the next decades. Thus it could be expected that the impact it may have on the incidence and prevalence of cardiovascular morbidity-mortality will have considerable magnitudes. It has been demonstrated that adequate metabolic control (glycemic and lipid) of these patients, beginning with diet and exercise programs and then with drug measures, decreases the risk of complications. However, several studies have shown that metabolic control deteriorates over time regardless of the treatment used. In recent years, a new drug family has been incorporated into the therapeutic armamentarium to treat type 2 diabetes mellitus. These are thiazolidinediones or glitazones, which have differential aspects regarding other drugs. In this article, the metabolic and systemic effects of pioglitazone, that have recently demonstrated a positive effect in the secondary prevention of cardiovascular episodes in the PROactive study have been reviewed.
Collapse
|
26
|
Jawa AA, Fonseca VA. Role of insulin secretagogues and insulin sensitizing agents in the prevention of cardiovascular disease in patients who have diabetes. Cardiol Clin 2005; 23:119-38. [PMID: 15694742 DOI: 10.1016/j.ccl.2004.06.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
In the absence of clinical trial evidence to compare the secretagogues with sensitizers, it is difficult to make recommendations about which class of drug is more important to prescribe for the prevention of cardiovascular disease in diabetes mellitus. Epidemiologic data supports insulin resistance as a major factor in cardiovascular disease through a variety of mechanisms. Because sensitizers improve insulin sensitivity and correct many of the vascular abnormalities that are associated with insulin resistance, it is tempting to suggest that they may be superior for this purpose. Conversely, meeting the goals that are recommended for glycemia also are important and achieving them may not be always possible with sensitizers, particularly in the later stages of the disease when insulin levels are not high,despite insulin resistance. In such situations,combination therapy may be needed with both types of drugs. No data are available on the cardiovascular effects of such combinations;some retrospective data suggest a possibility of increased events with the combination of sulfonylureas and metformin. Thus, further prospective studies in this area are necessary.
Collapse
Affiliation(s)
- Ali A Jawa
- Department of Medicine, Section of Endocrinology, Tulane University Medical Center, SL-53, 1430 Tulane Avenue, New Orleans, LA 70112-2699, USA
| | | |
Collapse
|
27
|
Kunhiraman BP, Jawa A, Fonseca VA. Potential cardiovascular benefits of insulin sensitizers. Endocrinol Metab Clin North Am 2005; 34:117-35. [PMID: 15752925 DOI: 10.1016/j.ecl.2004.11.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
A multiple risk factor approach is needed in patients who have type 2 diabetes. Because many risk factors are linked with IR, treatment with insulin sensitizers has the potential to modulate these risk factors favorably. TZDs 'have many important effects beyond lowering blood glucose. By targeting IR, they improve many cardiovascular risk factors that are associated with the IR syndrome. In particular, they increase HDL-C, have anti-inflammatory effects, improve endothelial function and fibrinolysis, and decrease carotid intimal thickness; however, no evidence-based studies on cardiovascular outcomes are available to substantiate the potential cardioprotective effects of TZDs. Several clinical trials that were designed to investigate the effect that these agents have on reducing cardiovascular events are well under way.
Collapse
Affiliation(s)
- Biju P Kunhiraman
- Section of Endocrinology, Diabetes, and Metabolism, Tulane University and Hospital, 1430 Tulane Avenue, New Orleans, LA 70112, USA
| | | | | |
Collapse
|
28
|
Bruemmer D, Blaschke F, Law RE. New targets for PPARgamma in the vessel wall: implications for restenosis. Int J Obes (Lond) 2005; 29 Suppl 1:S26-30. [PMID: 15711579 DOI: 10.1038/sj.ijo.0802910] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Peroxisome proliferator-activated receptor {gamma} (PPARgamma), the nuclear receptor that binds the insulin-sensitizing thiazolidinediones (TZDs), is prominently upregulated in intimal vascular smooth muscle cells (VSMC) after mechanical injury to the vessel wall. Several TZD PPARgamma ligands have been shown to inhibit neointima formation in both normal and insulin-resistant vasculature. The suppression of intimal hyperplasia by TZD PPARgamma ligands probably results from their activity to inhibit VSMC growth and promote apoptosis. TZDs prevent VSMC proliferation by blocking the activity of regulatory proteins, such as phosphorylation of the retinoblastoma protein (Rb). Rb functions as a G(1) gatekeeper by controlling S phase gene expression mediated by the E2F transcription factor. Consistent with their effect on Rb phosphorylation, PPARgamma ligands inhibit the mitogenic induction of minichromosome maintenance (MCM) proteins 6 and 7, two E2F-regulated S phase genes essential for DNA replication. PPARgamma ligands also induced apoptosis in VSMC, which correlated with a potent induction of GADD45, a gene implicated in controlling cell growth and survival. A constitutively active form of PPARgamma targeted the same cell cycle regulators as did PPARgamma ligands, consistent with a nuclear-receptor-dependent mechanism of action. This review will summarize mechanisms through which PPARgamma modulates VSMC proliferation and apoptosis suggesting that PPARgamma itself is a novel important regulator of cell cycle and apoptosis and may provide a new therapeutic approach to prevent restenosis.
Collapse
Affiliation(s)
- D Bruemmer
- Division of Endocrinology and Molecular Medicine, University of Kentucky College of Medicine, Lexington, KY, USA.
| | | | | |
Collapse
|
29
|
Seki N, Bujo H, Jiang M, Shibasaki M, Takahashi K, Hashimoto N, Saito Y. A potent activator of PPARα and γ reduces the vascular cell recruitment and inhibits the intimal thickning in hypercholesterolemic rabbits. Atherosclerosis 2005; 178:1-7. [PMID: 15585194 DOI: 10.1016/j.atherosclerosis.2004.08.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2004] [Revised: 07/18/2004] [Accepted: 08/10/2004] [Indexed: 11/28/2022]
Abstract
Peroxisome proliferator-activated receptors (PPARs) regulate the vascular cell functions as well as systemic lipid and glucose metabolism. Here, we studied the effect of TAK-559, a newly developed potent activator both for PPARalpha and gamma, on the vascular cell recruitment. TNF-alpha- or interleukin-1beta (IL-1beta)-induced THP-1 cell attachment to cultured endothelial cells was significantly reduced in the presence of 10 microM TAK-559 (P < 0.05). The secretion of monocyte chemoattractant protein-1 (MCP-1) from endothelial cells is reduced by 36% in the presence of 10 microM TAK-559, accompanied with the decreased mRNA expression in the cells. The proliferation and migration of cultured smooth muscle cells (SMCs) were significantly decreased in the presence of TAK-559 (P < 0.05). TAK-559-treated hypercholesterolemic rabbits showed the significant reduction of intimal thickning after balloon catheterization by 51% compared with control (P < 0.05), although the plasma lipid and glucose level was not changed between them. The numbers of macrophage and SMCs were decreased to 34% and 49% in the hyperplastic intima of arteries from TAK-559-treated rabbits compared to those from control, respectively. These results suggest that the PPARalpha and gamma activator inhibits the recruitment of macrophages and SMCs in intima, possibly leading to the reduction of intimal hyperplasia in hypercholesterolemia.
Collapse
Affiliation(s)
- Naoto Seki
- Department of Clinical Cell Biology (F5), Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
| | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
BACKGROUND Outcome studies are used to measure clinically meaningful primary end points, such as mortality and cardiovascular morbidity. However, few outcome trials have been conducted exclusively in people with diabetes; the majority of conventional diabetes trials use surrogate end points that may or may not translate into clinical benefits. Our current knowledge of the effects of pharmacotherapies on cardiovascular risk in patients with diabetes has been gained from subgroups included in large-scale studies. Several trials with lipid-modifying, antiplatelet and/or antihypertensive therapy, for example the recent Collaborative AtoRvastatin Diabetes Study, have included sufficient numbers of patients with diabetes to indicate that effective management can reduce cardio vascular risk in this patient population. The United Kingdom Diabetes Study and the Diabetes Control and Complications Trial provide important, but inconclusive data on the impact of glucose-lowering therapy on the incidence of cardiovascular complications in diabetes. Thiazolidinediones have only become available during the past few years, thus their effects were not assessed in these landmark trials. Ongoing studies in diabetic populations at high risk for further macrovascular events, such as the PROspective pioglitAzone Clinical Trial In macroVascular Events, have been designed to assess the effect of thiazolidinediones on cardiovascular outcome in patients with diabetes and should help to reinforce the importance of broad-based treatment of the multiple metabolic risk factors for cardiovascular disease in people with diabetes. SCOPE This paper (based upon MEDLINE and EMBASE literature searches in the year range 1990-2005) reviews what we have learned from outcome studies up to the end of 2004 and looks at what we hope to learn from ongoing studies.
Collapse
Affiliation(s)
- M Hanefeld
- Centre for Clinical Studies, GWT Technical University, Dresden, Germany.
| |
Collapse
|
31
|
Abstract
The sulphonylureas act by triggering insulin release from the pancreatic beta cell. A specific site on the adenosine triphosphate (ATP)-sensitive potassium channels is occupied by sulphonylureas leading to closure of the potassium channels and subsequent opening of calcium channels. This results in exocytosis of insulin. The meglitinides are not sulphonylureas but also occupy the sulphonylurea receptor unit coupled to the ATP-sensitive potassium channel. Glibenclamide (glyburide), gliclazide, glipizide and glimepiride are the primary sulphonylureas in current clinical use for type 2 diabetes mellitus. Glibenclamide has a higher frequency of hypoglycaemia than the other agents. With long-term use, there is a progressive decrease in the effectiveness of sulphonylureas. This loss of effect is the result of a reduction in insulin-producing capacity by the pancreatic beta cell and is also seen with other antihyperglycaemic agents. The major adverse effect of sulphonylureas is hypoglycaemia. There is a theoretical concern that sulphonylureas may affect cardiac potassium channels resulting in a diminished response to ischaemia. There are now many choices for initial therapy of type 2 diabetes in addition to sulphonylureas. Metformin and thiazolidinediones affect insulin sensitivity by independent mechanisms. Disaccharidase inhibitors reduce rapid carbohydrate absorption. No single agent appears capable of achieving target glucose levels in the majority of patients with type 2 diabetes. Combinations of agents are successful in lowering glycosylated haemoglobin levels more than with a single agent. Sulphonylureas are particularly beneficial when combined with agents such as metformin that decrease insulin resistance. Sulphonylureas can also be given with a basal insulin injection to provide enhanced endogenous insulin secretion after meals. Sulphonylureas will continue to be used both primarily and as part of combined therapy for most patients with type 2 diabetes.
Collapse
Affiliation(s)
- Marc Rendell
- Creighton Diabetes Center, 601 North 30th Street, Omaha, NE 68131, USA.
| |
Collapse
|
32
|
Nakamura T, Matsuda T, Kawagoe Y, Ogawa H, Takahashi Y, Sekizuka K, Koide H. Effect of pioglitazone on carotid intima-media thickness and arterial stiffness in type 2 diabetic nephropathy patients. Metabolism 2004; 53:1382-6. [PMID: 15375799 DOI: 10.1016/j.metabol.2004.05.013] [Citation(s) in RCA: 79] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Atherosclerosis is the major cause of morbidity and mortality in patients with type 2 diabetes, and pioglitazone has been reported to have anti-inflammatory and potential antiatherogenic effects. The aim of the present study was to determine whether pioglitazone, glibenclamide, or voglibose affects carotid intima-media thickness (IMT), pulse wave velocity (PWV), and urinary albumin excretion (UAE) in normotensive type 2 diabetic nephropathy patients. Forty-five normotensive type 2 diabetes patients with microalbuminuria were randomized to 12-month treatment with pioglitazone (30 mg/d, n = 15), glibenclamide (5 mg/d, n = 15), or voglibose (0.6 mg/d, n = 15). Pre- and posttreatment UAE, PWV, and IMT values were compared between treatment groups and a group of age-matched healthy control subjects (n = 30). Pretreatment PWV, IMT, and UAE values differed little between the 3 groups, but UAE was greater in the 45 type 2 diabetes patients (132.5 +/- 36.4 microg/min) than in the control subjects (6.2 +/- 1.8 microg/min, P < .001). IMT (0.76 +/- 0.12 mm) was significantly greater in the diabetics than in the controls (0.60 +/- 0.08 mm, P < .01). PWV (1,840 +/- 320 cm/s) was also significantly greater in the diabetics than in the controls (1,350 +/- 225 cm/s, P < .01). After 6 and 12 months, UAE, IMT, and PWV in the pioglitazone treatment group were significantly lower than those in the glibenclamide treatment group and voglibose treatment group (UAE: 6 months, P < .05 and 12 months, P < .01; IMT and PWV: 6 months, P < .05 and 12 months, P < .05). Pioglitazone, but not glibenclamide or voglibose, appears to be effective in reducing UAE, IMT, and PWV in normotensive type 2 diabetes patients with microalbuminuria.
Collapse
Affiliation(s)
- Tsukasa Nakamura
- Department of Medicine, Shinmatsudo Central General Hospital, Chiba, Japan
| | | | | | | | | | | | | |
Collapse
|
33
|
Zegarska J, Paczek L, Pawlowska M, Bartlomiejczyk I, Rowinski W, Kosieradzki M, Malanowski P, Kwiatkowski A, Grochowiecki T, Szmidt J. Extracellular matrix proteins, proteolytic enzymes, and TGF-beta1 in the renal arterial wall of chronically rejected renal allografts. Transplant Proc 2004; 35:2193-5. [PMID: 14529885 DOI: 10.1016/s0041-1345(03)00789-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Chronic rejection (CR) is the leading cause of long-term failure of transplanted kidneys. The vascular hallmark is intimal hyperplasia, accompanied by macrophage, foam cell, and T-cell infiltration. Intimal thickening results from the migration and proliferation of smooth muscle cells and increased deposits of extracellular matrix (ECM) proteins, due to release of growth factors and cytokines as well as altered ECM protein turnover. We assessed the content of fibronectin (FN) and transforming growth factor-beta1 (TGF-beta1) as well as the activities of collagenase and cathepsin B and L in renal artery walls of chronically rejected human renal allografts. We investigated renal artery samples from 8 patients with CR undergoing graftectomy, 12 patients undergoing nephrectomy, and 7 organ donors. The results were related to the DNA content of homogenates. Cathepsin B and L activities were significantly higher among those with compared with donors (P =.022). There was a trend toward higher collagenase activity in CR compared with donors and the nephrectomy group. TGF-beta1 was significantly enhanced in CR compared with donors (P =.010), and showed a trend toward higher concentrations in CR compared with the nephrectomy group. The trend was toward lower FN concentrations in CR compared with the nephrectomy group and toward higher concentrations compared with donors. Summarizing, renal CR is accompanied by enhanced proteinase activity, alterations of ECM proteins, and increased TGF-beta1 in the renal artery wall. We conclude that ECM turnover and cytokines play an important role in neointimal formation and CR pathogenesis.
Collapse
Affiliation(s)
- J Zegarska
- Department of Immunology, Transplant Medicine and Internal Diseases, Medical University of Warsaw, Warsaw, Poland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Blaschke F, Bruemmer D, Law RE. Will the potential of peroxisome proliferator-activated receptor agonists be realized in the clinical setting? Clin Cardiol 2004; 27:IV3-10. [PMID: 15470905 PMCID: PMC6654069 DOI: 10.1002/clc.4960271603] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Drugs targeting both peroxisome proliferator-activated receptor-gamma (PPAR-gamma) agonists (the thiazolidinediones) and PPAR-alpha (the fibrates) have already been developed for clinical use. However, the thiazolidinediones, currently prescribed to treat hyperglycemia and improve peripheral insulin resistance, may also have cardiovascular benefits that have yet to be fully realized. Animal models of atherosclerosis have shown that the thiazolidinediones reduce the extent of atherosclerotic lesions and inhibit macrophage accumulation. Clinical studies have also shown that these drugs improve the lipid profile of patients at risk of developing atherosclerosis and reduce circulating levels of inflammatory markers. This combination of lower lipid concentrations and reduced inflammation may explain the cardiovascular benefits of this class of drugs. Early trials in patients with coronary stents have reported promising findings, with restenosis rates being greatly reduced with thiazolidinedione therapy. It is hoped that the results of future clinical trials will continue to be encouraging, so that the thiazolidinediones' cardiovascular benefits can be fully realized in the clinic.
Collapse
Affiliation(s)
- Florian Blaschke
- Department of Medicine/Cardiology, German Heart Institute, Berlin, Germany
- Division of Endocrinology, Diabetes, and Hypertension, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Dennis Bruemmer
- Division of Endocrinology, Diabetes, and Hypertension, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Ronald E. Law
- Division of Endocrinology, Diabetes, and Hypertension, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| |
Collapse
|
35
|
Charbonnel B, Dormandy J, Erdmann E, Massi-Benedetti M, Skene A. The prospective pioglitazone clinical trial in macrovascular events (PROactive): can pioglitazone reduce cardiovascular events in diabetes? Study design and baseline characteristics of 5238 patients. Diabetes Care 2004; 27:1647-53. [PMID: 15220241 DOI: 10.2337/diacare.27.7.1647] [Citation(s) in RCA: 171] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE The PROspective pioglitAzone Clinical Trial In macroVascular Events (PROactive) assesses the effect of pioglitazone, a peroxisome proliferator-activated receptor agonist, with anti-inflammatory and vascular properties, on the secondary prevention of macrovascular events in type 2 diabetes. RESEARCH DESIGN AND METHODS PROactive is an on-going randomized, double-blind outcome study in patients with type 2 diabetes managed with diet and/or oral blood glucose-lowering drugs (combination of oral agents with insulin is permitted) who have a history of macrovascular disease. Patients are randomized to receive pioglitazone (forced titration from 15 to 30 to 45 mg, depending on tolerability) or placebo in addition to existing therapy. The primary end point is the time from randomization to occurrence of a new macrovascular event or death. Follow-up is estimated to span 4 years. RESULTS A total of 5238 patients have been randomized from 19 countries. At entry into the study, patients enrolled are a mean age of 61.8 years, with type 2 diabetes for a mean of 9.5 years; 60.9 and 61.5% are taking metformin or a sulfonylurea, respectively; and 33.6% are using insulin in addition to oral glucose-lowering drugs. The majority of patients are men (66.1%). Patients are required to meet one or more of entry criteria, as follows: >6 months' history of myocardial infarction (46.7%); coronary artery revascularization (30.8%), stroke (18.8%), or acute coronary syndrome for >3 months (13.7%); other evidence of coronary artery disease (48.1%); or peripheral arterial occlusive disease (19.9%). One-half (48.5%) of the patients have two or more of these risk factors. Three-quarters (75.4%) have hypertension, and 58.8% are current or previous smokers. CONCLUSIONS The cohort of patients enrolled in PROactive is a typical type 2 diabetic population at high risk of further macrovascular events. The characteristics of this population are ideal for assessing the ability of pioglitazone to reduce the cardiovascular risk of patients with type 2 diabetes.
Collapse
|
36
|
Ueta M, Wakisaka M, Ago T, Kitazono T, Nakamura U, Yoshinari M, Iwase M, Iida M. PPARγ ligands attenuate mesangial contractile dysfunction in high glucose. Kidney Int 2004; 65:961-71. [PMID: 14871416 DOI: 10.1111/j.1523-1755.2004.00474.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND To elucidate the regulation of peroxisome proliferator-activated receptor gamma (PPARgamma) and its roles in mesangial cells, we examined the expression of PPARgamma1 and effects of its ligands on cell phenotypes and angiotensin II-induced contractile response in cultured rat mesangial cells under a high (20 mmol/L) glucose condition. METHODS The effects of tumor necrosis factor alpha (TNFalpha), protein kinase C (PKC) activation, antisense DNA for PPARgamma1, PPARgamma ligands and PD98059 were examined in mesangial cells cultured in either 5 mmol/L or 20 mmol/L glucose. The expressions of PPARgamma1 protein and alpha-smooth muscle actin (alphaSMA) as a marker of phenotype of cells were determined by Western blot. The expression of PPARgamma1 mRNA was determined by a reverse transcription-polymerase chain reaction method. The reduction of cell surface area in response to angiotensin II was measured by microscope to determine cellular contraction. RESULTS PKC activation, TNFalpha, and 20 mmol/L glucose decreased PPARgamma1 at both protein and mRNA levels, which was inhibited by PD98059, a specific inhibitor of mitogen-activated protein kinase (MAPK). Decreases of PPARgamma1 protein and contractile response and an increase of alphaSMA occurred simultaneously in the cells treated with 20 mmol/L glucose after 5 days, which were attenuated to the normal levels by PPARgamma ligands. The antisense DNA also induced the decrease of PPARgamma1 protein, contractile dysfunction, and increase of alphaSMA. CONCLUSION MAPK suppresses PPARgamma1 at the transcriptional level, and the reduction of PPARgamma1 in cultured rat mesangial cells under the high glucose condition induces phenotypic change and loss of contractile function. PPARgamma ligands recover both reductions of PPARgamma 1 protein and contractile response.
Collapse
Affiliation(s)
- Maki Ueta
- Department of Medicine and Clinical Science, Graduate School of Medical Science, Kyushu University, Fukuoka, Japan
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Ward JE, Gould H, Harris T, Bonacci JV, Stewart AG. PPAR gamma ligands, 15-deoxy-delta12,14-prostaglandin J2 and rosiglitazone regulate human cultured airway smooth muscle proliferation through different mechanisms. Br J Pharmacol 2004; 141:517-25. [PMID: 14718259 PMCID: PMC1574213 DOI: 10.1038/sj.bjp.0705630] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The influence of two peroxisome proliferator-activated receptor gamma (PPARgamma) ligands, a thiazolidinedione, rosiglitazone (RG) and the prostaglandin D2 metabolite 15-deoxy-Delta(12,14)-prostaglandin J2 (15d-PGJ2) on the proliferation of human cultured airway smooth muscle (HASM) was examined. The increases in HASM cell number in response to basic fibroblast growth factor (bFGF, 300 pm) or thrombin (0.3 U ml-1) were significantly inhibited by either RG (1-10 microM) or 15d-PGJ2 (1-10 microM). The effects of RG, but not 15d-PGJ2, were reversed by the selective PPARgamma antagonist GW9662 (1 microM). Neither RG nor 15d-PGJ2 (10 microM) decreased cell viability, or induced apoptosis, suggesting that the regulation of cell number was due to inhibition of proliferation, rather than increased cell death. Flow-cytometric analysis of HASM cell cycle distribution 24 h after bFGF addition showed that RG prevented the progression of cells from G1 to S phase. In contrast, 15d-PGJ2 caused an increase in the proportion of cells in S phase, and a decrease in G2/M, compared to bFGF alone. Neither RG nor 15d-PGJ2 inhibited ERK phosphorylation measured 6 h post mitogen addition. The bFGF-mediated increase in cyclin D1 protein levels after 8 h was reduced in the presence of 15d-PGJ2, but not RG. Although both RG and 15d-PGJ2 can inhibit proliferation of HASM irrespective of the mitogen used, only the antiproliferative effects of RG appear to be PPARgamma-dependent. The different antimitogenic mechanisms of 15d-PGJ2 and synthetic ligands for PPARgamma may be exploited to optimise the potential for these compounds to inhibit airway remodelling in asthma. British Journal of Pharmacology (2004) 141, 517-525. doi:10.1038/sj.bjp.0705630
Collapse
Affiliation(s)
- Jane E Ward
- Department of Pharmacology, University of Melbourne, Melbourne, Victoria 3010, Australia.
| | | | | | | | | |
Collapse
|
38
|
Abstract
Vascular smooth muscle cells (VSMCs) in the media of adult arteries are normally quiescent, proliferate at low frequency, and are arrested in the G(0)/G(1) phase of the cell cycle. Proliferation of VSMCs occurs in response to arterial injury and plays a crucial role in the atherosclerotic process and in the pathogenesis of restenosis. Patients with type 2 diabetes mellitus are at increased risk for postangioplasty restenosis, which results from excessive intimal hyperplasia. Insulin sensitizers of the thiazolidinedione (TZD) class inhibit growth of VSMCs by attenuating the activity of important cell-cycle regulators. The TZDs inhibit progression from G(1) to S phase in the cell cycle by blocking growth factor-induced phosphorylation of retinoblastoma tumor suppressor protein (Rb). In animal models of restenosis, TZDs inhibit intimal hyperplasia after mechanical injury in both insulin-sensitive and insulin-resistant vessels. Preliminary clinical studies using troglitazone demonstrate less intimal hyperplasia with this TZD after implantation of coronary stents in individuals with type 2 diabetes. Further large trials are needed to confirm that treatment with a TZD can protect against postangioplasty restenosis.
Collapse
MESH Headings
- Blood Vessel Prosthesis Implantation
- Cell Cycle Proteins/drug effects
- Cell Cycle Proteins/metabolism
- Cell Division/drug effects
- Coronary Artery Disease/physiopathology
- Coronary Artery Disease/therapy
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/physiopathology
- Humans
- Insulin Resistance/physiology
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/physiopathology
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/drug effects
- Risk Factors
- Stents
- Thiazolidinediones/pharmacology
Collapse
Affiliation(s)
- Dennis Bruemmer
- Division of Endocrinology, Diabetes, and Hypertension, David Geffen School of Medicine at the University of California-Los Angeles, Los Angeles, California 90095, USA
| | | |
Collapse
|
39
|
Abstract
Patients with diabetes have a greatly increased relative risk of developing cardiovascular disease when compared with patients without diabetes. Much of this risk is related to insulin resistance and is associated with both traditional and nontraditional cardiovascular risk factors. Therapy for diabetes must address these risk factors in an attempt to prevent and adequately treat cardiovascular disease. Pharmacologic therapy directed toward dyslipidemia and hypertension has a beneficial effect on risk factors and has been shown to decrease cardiovascular events. The effects of insulin and oral hypoglycemic agents on insulin resistance are variable, and their direct effect on cardiovascular disease is less clear. Metformin is the only oral hypoglycemic agent shown to decrease cardiovascular events independent of glycemia. The thiazolidinediones directly improve insulin resistance, decrease plasma insulin concentration, and have the potential to decrease the risk of cardiovascular disease in patients with diabetes. A number of studies have demonstrated that the thiazolidinediones produce changes in several cardiovascular risk factors associated with the insulin resistance syndrome, including lowering blood pressure, correcting diabetic dyslipidemia, improving fibrinolysis, and decreasing carotid artery intima-medial thickness. These agents bind a newly described class of receptors, peroxisome proliferator-activated receptors, which may have implications for atherosclerosis. Although these drugs increase low-density lipoprotein (LDL) cholesterol, they induce a favorable change in the LDL particle size and susceptibility to oxidation. Long-term clinical trials are being conducted to determine the effect that thiazolidinediones have on cardiovascular events in individuals with type 2 diabetes.
Collapse
Affiliation(s)
- Vivian A Fonseca
- Department of Medicine, and Pharmacology, Tulane University Health Sciences Center, New Orleans, Lousiana 70112, USA.
| |
Collapse
|
40
|
Takagi T, Yamamuro A, Tamita K, Yamabe K, Katayama M, Mizoguchi S, Ibuki M, Tani T, Tanabe K, Nagai K, Shiratori K, Morioka S, Yoshikawa J. Pioglitazone reduces neointimal tissue proliferation after coronary stent implantation in patients with type 2 diabetes mellitus: an intravascular ultrasound scanning study. Am Heart J 2003; 146:E5. [PMID: 12891212 DOI: 10.1016/s0002-8703(03)00146-7] [Citation(s) in RCA: 118] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND It has been reported that pioglitazone reduces neointimal hyperplasia after balloon-induced vascular injury in an experimental model. METHODS To determine whether pioglitazone reduces neointimal tissue proliferation after coronary stent implantation in patients with type 2 diabetes mellitus, we studied 44 stented lesions in 44 patients with diabetes mellitus who underwent successful coronary stent implantation. Study patients were randomized into 2 groups: the pioglitazone group (23 patients with 23 lesions) and the control group (21 patients with 21 lesions). All patients underwent serial quantitative coronary angiography and serial intravascular ultrasound scanning studies. With a motorized pullback system, multiple image slices within the stent were obtained at every 1 mm. The stent area and lumen area were measured, and the neointimal area was calculated. Measurements were averaged over the number of selected image slices. The neointimal index was calculated as the averaged neointimal area divided by the averaged stent area multiplied by 100 (%). RESULTS After 6 months of treatment, angiographic in-stent restenosis (17% vs 43%, respectively, P =.0994) and target lesion revascularization (13% vs 38%, respectively, P =.0835) were less frequent in the pioglitazone group than the control group; however, these differences did not reach significance. The intravascular ultrasound scanning study demonstrated that the neointimal index in the pioglitazone group was significantly smaller than that in the control group (28% +/- 9% vs 48% +/- 15%, respectively, P <.0001). CONCLUSION A serial intravascular ultrasound scanning assessment demonstrated that pioglitazone reduces neointimal tissue proliferation after coronary stent implantation in patients with type 2 diabetes mellitus.
Collapse
|
41
|
Kawabata H, Ishikawa K. Cardioprotection with pioglitazone is abolished by nitric oxide synthase inhibitor in ischemic rabbit hearts--comparison of the effects of pioglitazone and metformin. Diabetes Metab Res Rev 2003; 19:299-305. [PMID: 12879407 DOI: 10.1002/dmrr.379] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND The effects of two drugs representing different classes of antidiabetic pharmacology (pioglitazone, a thiazolidinedione; and metformin, a biguanide) on the myocardial metabolism in the ischemia are poorly understood. METHODS To test the hypothesis that cardioprotection of pioglitazone and metformin is associated with nitric oxide (NO), we studied the high energy phosphate metabolism by 31P-nuclear magnetic resonance (NMR) in isolated rabbit hearts. Forty-five minutes of continuous normothermic global ischemia was carried out. Pioglitazone or metformin was administered at the beginning, 60 min prior to the global ischemia, with or without the nitric oxide synthase inhibitor, L-NAME, administered 5 min or 60 min prior to the ischemia. In the first experiment, whether NO was produced or not by administration of pioglitazone, for the prevention of myocardial ischemic injury, was investigated. Hearts of male Japanese white rabbits were divided into 4 experimental groups: the control (C) group, the P group consisting of pioglitazone treatment, the P + L5 group consisting of pioglitazone treatment with L-NAME (5 min before ischemia), and the P + L60 group consisting of pioglitazone treatment with L-NAME (60 min before ischemia). In the next experiment, a comparison between the effects of pioglitazone and metformin in preventing ischemic injury were studied. The hearts were divided into 4 experimental groups: the control (C) group, the P group consisting of pioglitazone treatment, the P + L5 group consisting of pioglitazone treatment with L-NAME (5 min before ischemia), the M group consisting of metformin treatment, and the M + L5 group consisting of metformin treatment with L-NAME (5 min before ischemia). RESULTS In the first experiment, the decrease in adenosine triphosphate (ATP) during ischemia was significantly inhibited in the P group in comparison with the C group (P < 0.01). However, the decrease in ATP was not inhibited in the P + L5 group during ischemia. In contrast, in the P + L60 group, the decrease in ATP was not inhibited during a part of ischemia. In the next experiment, a comparison between the effects of pioglitazone and metformin in preventing ischemic injury was studied. As a result of administration of either pioglitazone or metformin, there was no difference between groups with and without L-NAME. CONCLUSION These results suggest that pioglitazone has a significant beneficial effect on improving the myocardial energy metabolism during ischemia. This cardioprotection may be dependent on nitric oxide (NO) synthase during ischemia more than preischemia. Furthermore, the present findings suggest that both pioglitazone and metformin have equal cardioprotective effects mediated by NO on myocardial ischemic injury in rabbits.
Collapse
Affiliation(s)
- Hitoshi Kawabata
- Department of Cardiology, Kinki University School of Medicine, Osakasayama, Japan
| | | |
Collapse
|
42
|
Affiliation(s)
- Rasa Kazlauskaite
- Department of Medicine, Rush Medical College, Chicago, Illinois, USA
| | | |
Collapse
|
43
|
Bruemmer D, Yin F, Liu J, Berger JP, Kiyono T, Chen J, Fleck E, Van Herle AJ, Forman BM, Law RE. Peroxisome proliferator-activated receptor gamma inhibits expression of minichromosome maintenance proteins in vascular smooth muscle cells. Mol Endocrinol 2003; 17:1005-18. [PMID: 12677008 DOI: 10.1210/me.2002-0410] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Using a cDNA array consisting only of cell cycle genes, we found that a novel nonthiazolidinedione partial peroxisome proliferator-activated receptor gamma (PPARgamma) agonist (nTZDpa) inhibited expression of minichromosome maintenance (MCM) proteins 6 and 7 in vascular smooth muscle cells. MCM proteins are required for the initiation and elongation stages of DNA replication and are regulated by the transcription factor E2F. Mitogen-induced MCM6 and MCM7 mRNA expression was potently inhibited by nTZDpa and to a lesser degree by the full PPARgamma agonist, rosiglitazone. Inhibition of MCM6 and MCM7 expression by nTZDpa and rosiglitazone paralleled their effect to inhibit phosphorylation of the retinoblastoma protein and cell proliferation. Transient transfection experiments revealed that the nTZDpa inhibited mitogen-induced MCM6 and MCM7 promoter activity, implicating a transcriptional mechanism. Adenoviral-mediated E2F overexpression reversed the suppressive effect of nTZDpa on MCM6 and MCM7 expression. Furthermore, activity of a luciferase reporter plasmid driven by multiple E2F elements was inhibited by nTZDpa, indicating that their down-regulation by nTZDpa involves an E2F-dependent mechanism. Overexpression of dominant-negative PPARgamma or addition of a PPARgamma antagonist, GW 9662, blocked nTZDpa inhibition of MCM7 transcription. Adenovirus-mediated overexpression of constitutively active PPARgamma inhibited MCM7 expression in a similar manner as the nTZDpa. These findings provide strong evidence that activation of PPARgamma attenuates MCM7 transcription and support the important role of this nuclear receptor in regulating vascular smooth muscle cell proliferation.
Collapse
Affiliation(s)
- Dennis Bruemmer
- Division of Endocrinology, Diabetes and Hypertension and The Gonda (Goldschmied) Diabetes Center, David Geffen School of Medicine, University of California, Los Angeles, California 90095, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Martens FMAC, Visseren FLJ, Lemay J, de Koning EJP, Rabelink TJ. Metabolic and additional vascular effects of thiazolidinediones. Drugs 2002; 62:1463-80. [PMID: 12093315 DOI: 10.2165/00003495-200262100-00004] [Citation(s) in RCA: 211] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Several cardiovascular risk factors (dyslipidaemia, hypertension, glucose intolerance, hypercoagulability, obesity, hyperinsulinaemia and low-grade inflammation) cluster in the insulin resistance syndrome. Treatment of these individual risk factors reduces cardiovascular complications. However, targeting the underlying pathophysiological mechanisms of the insulin resistance syndrome is a more rational treatment strategy to further improve cardiovascular outcome. Our understanding of the so-called cardiovascular dysmetabolic syndrome has been improved by the discovery of nuclear peroxisome proliferator-activated receptors (PPARs). PPARs are ligand-activated transcription factors belonging to the nuclear receptor superfamily. As transcription factors, PPARs regulate the expression of numerous genes and affect glycaemic control, lipid metabolism, vascular tone and inflammation. Activation of the subtype PPAR-gamma improves insulin sensitivity. Expression of PPAR-gamma is present in several cell types involved in the process of atherosclerosis. Thus, modulation of PPAR-gamma activity is an interesting therapeutic approach to reduce cardiovascular events. Thiazolidinediones are PPAR-gamma agonists and constitute a new class of pharmacological agents for the treatment of type 2 (non-insulin-dependent) diabetes mellitus. Two such compounds are currently available for clinical use: rosiglitazone and pioglitazone. Thiazolidinediones improve insulin sensitivity and glycaemic control in patients with type 2 diabetes. In addition, improvement in endothelial function, a decrease in inflammatory conditions, a decrease in plasma levels of free fatty acids and lower blood pressure have been observed, which may have important beneficial effects on the vasculature. Several questions remain to be answered about PPAR-gamma agonists, particularly with respect to the role of PPAR-gamma in vascular pathophysiology. More needs to be known about the adverse effects of thiazolidinediones, such as hepatotoxicity, increased low-density lipoprotein cholesterol levels and increased oedema. The paradox of adipocyte differentiation with weight gain concurring with the insulin-sensitising effect of thiazolidinediones is not completely understood. The decrease in blood pressure induced by thiazolidinedione treatment seems incompatible with an increase in the plasma volume, and the discrepancy between the stimulation of the expression of CD36 and the antiatherogenic effects of the thiazolidinediones also needs further explanation. Long-term clinical trials of thiazolidinediones with cardiovascular endpoints are currently in progress. In conclusion, studying the effects of thiazolidinediones may shed more light on the mechanisms involved in the insulin resistance syndrome. Furthermore, thiazolidinediones could have specific, direct effects on processes involved in the development of vascular abnormalities.
Collapse
Affiliation(s)
- Fabrice M A C Martens
- Department of Internal Medicine, Section of Vascular Medicine and Diabetology, University Medical Center Utrecht, Room F.02.126, Heidelberglaan 100, PO Box 85500, 3508 GA Utrecht, The Netherlands.
| | | | | | | | | |
Collapse
|
45
|
Abstract
In the last few years there has been an explosion of research that has improved our understanding of the pathogenesis of Type 2 diabetes mellitus (DM-2) and has led to the development of new oral antidiabetic drugs. Thiazolidinediones (TZDs) are the newest of these antidiabetic agents. TZDs are insulin sensitisers that depend on the presence of insulin for their action. They target insulin resistance, which is thought to play a central role in DM-2 and the associated metabolic syndrome characterised by central obesity, hypertension, dyslipidemia and hypercoagulability, all leading to increased cardiovascular morbidity and mortality. As a result, TZDs have the potential to improve other conditions associated with the metabolic syndrome, in addition to their glycaemic action. TZDs act by activating peroxisome proliferator-activated receptor (PPAR) phi a nuclear receptor implicated not only in lipid and glucose metabolism but other physiological functions as well. TZDs may have wide clinical applications beyond DM-2, as they can potentially be used to treat other conditions associated with insulin resistance and PPAR-phi receptors, such as impaired glucose tolerance, polycystic ovarian syndrome and HIV lipodystrophy.
Collapse
Affiliation(s)
- Anastassios G Pittas
- Tufts University School of Medicine, New England Medical Center, Division of Endocrinology, Diabetes, Metabolism and Molecular Medicine, 750 Washington Street, NEMC#268, Boston, MA 02111, USA.
| | | |
Collapse
|
46
|
Jozkowicz A, Dulak J, Nanobashvili J, Prager M, Huk I. Role of Peroxisome Proliferator-Activated Receptor gamma Ligands in the Vessel Wall. Eur Surg 2002. [DOI: 10.1046/j.1563-2563.2002.02032.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
47
|
Jozkowicz A, Dulak J, Nigisch A, Weigel G, Sporn E, Fugl A, Huk I. Ciglitazone, Ligand of Peroxisome Proliferator-Activated Receptor-gamma, Inhibits Vascular Endothelial Growth Factor Activity. Eur Surg 2002. [DOI: 10.1046/j.1563-2563.2002.02024.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
48
|
|
49
|
Abstract
Pharmaceutical interventions targeting proteins that regulate VSMC growth and movement are promising new approach to treat diabetes-associated cardiovascular disease. Peroxisome proliferator-activated receptor-gamma (PPARgamma) is a ligand-activated transcription factor in the nuclear receptor superfamily. Thiazolidineodione (TZT) insulin sensitizers are pharmacologic ligands for PPARgamma. All of the major cells in the vasculature express PPARgamma, including endothelial cells. VSMCs, and monocytes/macrophages. PPARgamma ligands may protect the vasculature against injury by inhibiting cell growth and movement, improving endothelial function, and suppressing tissue inflammation. Antiproliferative effects of PPARgamma ligands are mediated by targeting critical cell cycle regulators, including Rb and p27(Kip1), that regulate the progression of cells from G1 phase into S phase to conduct DNA synthesis. Pharmacologic activation of PPARgamma in vascular cells may provide a novel therapeutic approach to retard diabetes-associated vascular disease.
Collapse
Affiliation(s)
- Shu Wakino
- Division of Endocrinology, Diabetes and Hypertension, School of Medicine, University of California, Los Angeles, Warren Hall, Second Floor, Suite 24-130, 900 Veteran Avenue, Box 957073, Los Angeles, CA 90095, USA
| | | | | |
Collapse
|
50
|
Molavi B, Rasouli N, Mehta JL. Peroxisome proliferator-activated receptor ligands as antiatherogenic agents: panacea or another Pandora's box? J Cardiovasc Pharmacol Ther 2002; 7:1-8. [PMID: 12000972 DOI: 10.1177/107424840200700i101] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Peroxisome proliferator activated receptors (PPARs) are members of the nuclear receptor super family that modulate gene expression upon ligand activation. They are 3 major subtypes of PPARs: alpha, delta (also called beta), and gamma. PPAR-gamma is widely expressed in the cardiovascular system and is involved in the regulation of tissue inflammation and smooth muscle cell growth pathways as well as in lipoprotein metabolism and coagulation cascades. PPAR-gamma ligands of (e.g., rosigitazone and pioglitazone) have been shown to exert antiatherogenic effects both in vitro and in vivo. PPAR-alpha ligands (e.g., clofibrate and benzofibrate) modulate lipoprotein metabolism, and affect inflammation and coagulation cascade. These effects may be helpful in resolving the dilemma arising from studies that showed significant mortality and morbidity benefits of fibrates in the face of minimal changes in HDL-cholesterol levels. The role of PPAR-delta in atherogenesis remains largely unknown, although it appears that PPAR-delta activation affects lipoprotein metabolism. PPAR ligands appear to be promising agents in limiting atherosclerosis; however, large-scale clinical trials are required to assess their safety and efficacy before they can be added to the clinicians' arsenal of antiatherosclerotic agents.
Collapse
|