Get 20M+ Full-Text Papers For Less Than $1.50/day. Start a 14-Day Trial for You or Your Team.

Learn More →

Protective Actions of PPAR- Activation in Renal Endothelium

Protective Actions of PPAR- Activation in Renal Endothelium Protective Actions of PPAR-γ Activation in Renal Endothelium <meta name="citation_title" content="Protective Actions of PPAR- γ Activation in Renal Endothelium" /> //// Hindawi Publishing Corporation Home Journals About Us About this Journal Submit a Manuscript Table of Contents Journal Menu Abstracting and Indexing Aims and Scope Article Processing Charges Articles in Press Author Guidelines Bibliographic Information Contact Information Editorial Board Editorial Workflow Free eTOC Alerts Reviewers Acknowledgment Subscription Information Open Focus Issues Focus Issue Guidelines Open Special Issues Published Special Issues Special Issue Guidelines Abstract Full-Text PDF Full-Text HTML Linked References How to Cite this Article PPAR Research Volume 2008 (2008), Article ID 635680, 9 pages doi:10.1155/2008/635680 Review Article Protective Actions of PPAR- 𝛾 Activation in Renal Endothelium Peter E. Westerweel 1,2 and Marianne C. Verhaar 1 1 Department of Vascular Medicine, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands 2 Department of Internal Medicine, St. Antonius Hospital, Koekoekslaan 1, 3435 CM Nieuwegein, The Netherlands Received 15 September 2008; Accepted 8 December 2008 Academic Editor: Nigel Brunskill Copyright © 2008 Peter E. Westerweel and Marianne C. Verhaar. This is an open access article distributed under the Creative Commons Attribution License , which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Abstract Renal endothelial damage is pivotal in the initiation and progression of renal disease. Damaged renal endothelium may be regenerated through proliferation of local endothelium and circulation-derived endothelial progenitor cells. Activation of the PPAR- 𝛾 -receptors present on endothelial cells affects their cellular behavior. Proliferation, apoptosis, migration, and angiogenesis by endothelial cells are modulated, but may involve both stimulation and inhibition depending on the specific circumstances. PPAR- 𝛾 -receptor activation stimulates the production of nitric oxide, C-type natriuretic peptide, and superoxide dismutase, while endothelin-1 production is inhibited. Together, they augment endothelial function, resulting in blood pressure lowering and direct renoprotective effects. The presentation of adhesion molecules and release of cytokines recruiting inflammatory cells are inhibited by PPAR- 𝛾 -agonism. Finally, PPAR- 𝛾 -receptors are also found on endothelial progenitor cells and PPAR- 𝛾 -agonists stimulate progenitor-mediated endothelial repair. Together, the stimulatory effects of PPAR- 𝛾 -agonism on endothelium make an important contribution to the beneficial actions of PPAR- 𝛾 -agonists on renal disease. 1. Introduction PPAR- γ -agonists are widely used for their insulin-sensitizing actions in the treatment of type 2 diabetes mellitus, but have additional therapeutic potential beyond the metabolic effects. Currently, the clinically most used PPAR- γ -agonistic drugs are the thiazolidinediones (TZDs). PPAR- γ -agonists may favorably affect the course of renal disease in both diabetic and nondiabetic conditions [ 1 , 2 ]. In nondiabetic animals, beneficial effects have been shown for anti-GBM antibody-induced crescentic glomerulonephritis [ 3 ], passive Heymann nephritis [ 4 ], the development of glomerulosclerosis after 5/6 nephrectomy [ 5 ], renal ischemia-reperfusion induced damage [ 6 ], and anti-Thy-1-glomerulonephritis [ 7 ]. One potential mechanism is the modulation of endothelial cell function through activation of PPAR- γ receptors, which are expressed on glomerular endothelium [ 8 , 9 ]. In response to injury, the endothelial expression of PPAR- γ -receptors may be increased, for example, as transiently occurs after ischemia- reperfusion [ 8 , 9 ]. Treatment with PPAR- γ -agonists also increases the expression of PPAR- γ -receptors on renal endothelium in both the glomerulus and the capillary endothelium of the medullary vasa recta [ 10 ]. The relevance for renal disease of activating the PPAR- γ -receptors on renal endothelium is becoming increasingly clear and is the focus of this review. 2. The Role of Endothelium in Renal Disease Renal microvascular endothelial injury is a pivotal pathogenic factor for various renal diseases. Renal disease conditions involving prominent endothelial damage include ischemic nephropathy, glomerulonephritis, interstitial nephritis, and allograft rejection [ 11 , 12 ]. Endothelial dysfunction and attenuated angiogenesis contribute to declining renal function with ageing [ 13 ] and the pathogenesis and progression of chronic kidney disease [ 11 ]. The microvascular endothelium, by the release of endothelium-derived factors such as nitric oxide (NO) and as a critical component of the glomerular filtration barrier, exerts important protection against progressive renal damage. Endothelial dysfunction results in increased permeability causing passage of macromolecules (microalbuminuria), which is considered to be the earliest renal sign of vascular dysfunction [ 14 , 15 ]. Endothelial dysfunction has been shown to predict susceptibility to renal damage in a rat renal injury model [ 16 ]. Progression of renal disease does not only depend on the degree of microvascular endothelial injury, but also on the effectiveness of endothelial repair. Impaired glomerular capillary repair was found to be associated with the development of glomerulosclerosis and renal failure [ 17 ]. During experimental glomerulonephritis, angiogenic factors such as VEGF and bFGF are released, which stimulate endothelial regeneration [ 18 – 20 ]. Blocking the VEGF-induced endothelial repair with a VEGF-antagonist interferes with renal recovery and results in progressive renal impairment [ 21 ]. Consistently, progressive renal disease is associated with reduced expression of angiogenic growth factors and enhanced expression of antiangiogenic factors [ 22 , 23 ]. The glomerular endothelium can recover from injury by replacing lost or damaged endothelial cells, in part through proliferation of local endothelium, stimulated by the release of angiogenic growth factors [ 11 , 12 ]. We [ 24 – 26 ] and others [ 27 ] have observed in both human and experimental animal studies that damaged glomerular endothelium may also be regenerated from circulating bone marrow-derived endothelial progenitor cells. Endothelial progenitor cells incorporate into the damaged glomerulus, differentiate into mature endothelial cells, and eventually fully integrate into the resident endothelium [ 26 ]. Enhancing renal endothelial repair offers therapeutic potential. Stimulating angiogenesis with VEGF-treatment augments capillary repair and renal recovery after glomerulonephritis [ 19 ]. Enhancing NO production by supplementing the substrate L-arginine improves the clinical course of anti-Thy-1-glomerulonephritis [ 28 ]. Infusion of unselected bone marrow cells ameliorates experimental progressive glomerulosclerosis [ 29 ]. Intrarenal administration of endothelial progenitor cells attenuates endothelial injury and mesangial activation in experimental glomerulonephritis [ 30 ]. 3. The Role of PPAR- γ -Receptors in Vascular Development and Remodeling Phenotypical studies in humans and animals with genetic mutations in the PPAR- γ -receptor imply a role in the regulation of vascular function and remodeling. In humans with dominant-negative heterozygous mutations in the PPAR- γ -receptor causing an impaired capacity for transcriptional activation, diabetes and also hypertension occur at an unusually young age [ 31 ]. Heterozygous knockout mice have increased insulin sensitivity [ 32 ] and decreased fat mass [ 33 ], but do not have a vascular phenotype [ 34 ]. However, but PPAR- γ -null mice are embryonically lethal due to placental dysfunction, characterized by defective trophoblast differentiation and markedly impaired placental vascularization [ 34 ]. A surviving PPAR- γ -knockout mouse that was supplemented with a wild-type placenta developed an apparently normal vascular system during further embryogenesis, but died some days after birth due to a combination of pathologies, including severe lipodystrophic changes and hemorrhages [ 34 ]. In a conditional knockout model using a cre-lox system to save floxed PPAR- γ -knockout mice from embryonic lethality by preserving PPAR- γ -function in the trophoblast marked lipodystrophy was also observed, together with insulin resistance. Surprisingly, these mice were hypotensive and showed increased endothelium-dependent relaxation in response to acetylcholine [ 35 ]. In the latter study, PPAR- γ -function was deficient in all cell types. Mice in which PPAR- γ -function was selectively knocked out in endothelial cells only using again a cre-lox system were found to be hypertensive when fed a high-fat diet [ 36 ]. Also, in mice with a nonlethal-dominant negative mutation in the PPAR- γ -receptor, endothelium-dependent blood vessel dilatation is impaired, while the endothelium is more sensitive to endothelin-1-induced vasoconstriction [ 37 ]. Superoxide levels in these mice are elevated and treatment with a superoxide scavenger can reverse the impaired endothelial vasodilation, highlighting the pivotal role of increased radical formation [ 37 ]. Cerebral arterioles were hypertrophied with a decrease in luminal diameters, indicative of adverse inward vascular remodeling [ 37 ]. Of note, dominant negative PPAR- γ -mutations were found not to be fully selective, therefore, it cannot be excluded that some of the effects observed in the knockout systems are attributable to PPAR- γ -receptor-independent signaling, in particular through the alternative PPAR-receptors [ 38 ]. 4. Effects of PPAR- γ -Activation on Endothelial Proliferation, Migration, Angiogenesis, and Apoptosis Reports on the direct effects of PPAR- γ -agonist treatment on proliferation, migration, and angiogenic network formation of cultured endothelial cells showed variable results. Fukunaga et al. demonstrated increased proliferation with both troglitazone and pioglitazone in four different endothelial cell lines [ 39 ]. In contrast, two other studies found troglitazone to inhibit proliferation of macrovascular endothelial cells [ 40 ] and human umbilical vein endothelial cells [ 41 ]. Rosiglitazone was shown also to inhibit proliferation in one study [ 42 ], while another study found no effect at all [ 40 ]. VEGF-induced endothelial migration was found to be inhibited by PPAR- γ -agonists troglitazone and ciglitazone, which was mediated by inhibition of Akt [ 43 ]. This is in line with the reduced migration and inhibition of angiogenic network formation by PPAR- γ -agonism observed by Xin et al [ 44 ]. However, in the study by Fukunaga et al., troglitazone stimulated both endothelial cell migration and proliferation, resulting in accelerated coverage of a disrupted endothelial monolayer in a wound healing assay [ 39 ]. Also, Biscetti et al. found stimulation of endothelial network formation with PPAR- γ activation using the GW1929 compound, mainly through a VEGF-dependent mechanism [ 45 ]. In vitro effects of PPAR- γ -agonism on endothelial cell apoptosis have been similarly variable. Both spontaneous and TNF-alpha-induced endothelial apoptosis were shown to be inhibited by various thiazolidinedione PPAR- γ -agonists [ 46 ] and troglitazone was found to markedly reduce apoptosis in serum-starved endothelial cells [ 47 ], while another study observed induction of apoptosis using both ciglitazone and PPAR- γ -receptor overexpression [ 48 ]. In several studies, the endogenous PPAR- γ ligand 15-deoxy-delta12,14-prostaglandin J2 (15d-PGJ2)-induced endothelial cell apoptosis [ 46 , 48 , 49 ], but it is important to note that in vascular endothelial cells, the effects of 15d-PGJ2 may be independent of PPAR- γ -receptor activation [ 49 , 50 ]. Taken together, it is clear that PPAR- γ -agonists may modulate endothelial proliferation, migration, angiogenesis, and apoptosis in vitro, but that this may result in both stimulation and inhibition. The factors that determine whether treatment with a PPAR- γ -agonist results in a stimulatory or inhibitory effect on endothelial cells remain largely unidentified. This may involve PPAR- γ -receptor-independent effects as reported for 15d-PGJ2 [ 49 , 50 ] and also with the various TZDs [ 45 ]. Fukunaga et al. posed that a concentration dependency may explain some of the observed discrepancies, as they observed stimulation of DNA-synthesis at low PPAR- γ -agonist dosages and inhibition at higher dosages [ 39 ]. However, this cannot explain all of the divergent findings. In vivo studies in diabetic animals with impaired angiogenesis in response to peripheral ischemia showed that treatment with PPAR- γ -agonist pioglitazone augmented the angiogenic response and increased blood flow recovery [ 51 ]. In rats with experimental focal cerebral ischemia, PPAR- γ -agonist treatment stimulated local angiogenesis and improved functional neurological recovery [ 52 ]. In contrast, PPAR- γ -agonist treatment inhibited pathological choroidial and retinal neovascularization [ 53 ] and suppressed tumor growth and metastasis by inhibiting tumor angiogenesis in several primary tumors, in part through decreasing VEGF-production by tumor cells and blocking the production of angiogenic ELR+CXC-chemokines, mediated through antagonizing NF-kappaB activation [ 54 , 55 ]. These findings suggest that PPAR- γ -agonism may differentially affect neovascularization with inhibition of pathological neovascularization and augmentation of physiological neovascularization. This is in line with the in vitro observations of both stimulatory and inhibitory actions. 5. Effects of PPAR- γ -Activation on Endothelial Dysfunction Endothelial integrity does not only rely on the number of cells, but also on their function. NO-production is a key component of endothelial function. NO stimulates vasodilation, inhibits inflammation, prevents platelet activation, and scavenges radicals [ 56 ]. In vitro studies on the effect of PPAR- γ -activation have consistently shown a stimulating effect on NO production by endothelial cells [ 57 – 60 ]. The observation that siRNA against PPAR- γ blocked the increase in NO-production confirmed that this effect is PPAR- γ -receptor mediated [ 59 ]. In both experimental animal studies and in humans, PPAR- γ -activation has been shown to augment systemic NO-production and endothelial function. In diabetic rats, PPAR- γ -agonist treatment restored impaired endothelium-dependent arterial relaxation by increasing NO-production while reducing oxidative stress [ 61 ]. In healthy human subjects, a single dose of troglitazone increased NO-dependent endothelial function measured by venous occlusion plethysmography of the forearm and nitrite levels [ 62 ]. In nondiabetic patients with hypertension or hypercholesterolemia, endothelial function was improved with pioglitazone [ 63 ]. In type 2 diabetic patients, vascular resistance was shown to be reduced with troglitazone treatment [ 64 , 65 ] and in type 2 diabetic patients with angina pectoris, troglitazone reduced the frequency of angina pectoris with improving endothelial function [ 66 ]. Rosiglitazone attenuated the detrimental effects of the presence of diabetes on NO-production measured directly using an intravital probe and by assessing blood flow in the peripheral skin [ 67 ]. Long-term treatment with pioglitazone resulted in a reduced pulse wave velocity, indicative of reduced vascular stiffness [ 68 ]. In renal transplant recipients, PPAR- γ -agonist treatment enhanced endothelial function [ 69 ]. Importantly, the observed beneficial effects on systemic endothelial function were found to extend to improving intrarenal NO-production. In human type 2 diabetic patients, rosiglitazone treatment increased intrarenal NO levels, which was associated with improvement of renal hemodynamics and reduction of proteinuria [ 70 ]. This is in line with animal studies. In obese Zucker rats, PPAR- γ -agonist treatment lowered blood pressure and ameliorated abnormal pressure natriuresis in association with increased renal NO-metabolite nitrite/nitrate production [ 71 ]. In obese hypertensive Sprague-Dawley rats, PPAR- γ -agonist treatment also reduced blood pressure, increased NO-metabolite nitrite/nitrate excretion, and reduced excretion of oxidative-stress associated urinary isoprostanes and lipid peroxides [ 72 ]. In the kidneys of these rats, eNOS expression was increased while the pathological increase in p47phox and gp91phox associated with obesity was attenuated [ 72 ]. Differences have been observed between the various PPAR- γ -activating compounds in the signaling level at which NO-production is stimulated. The NO producing enzyme endothelial nitric oxide synthase (eNOS) is not only regulated at the level of transcription and translation, but also has multiple phosphorylation sites for activation and deactivation of the enzyme and requires translocation to the caveolae to associate with its cofactors and effectively produce NO [ 73 , 74 ]. Troglitazone has been shown to enhance NO-production by increasing eNOS transcription and eNOS protein translation in isolated endothelial cells [ 58 ], while no effect was found at the transcriptional level for 15d-PGJ2 [ 57 , 58 ], pioglitazone [ 58 ], and ciglitazone [ 57 ]. 15d-PGJ2 and rosiglitazone, but not ciglitazone, were found to stimulate eNOS-phophorylation at activation site ser-1177, thought to be mediated through increasing heat shock protein (hsp)-90 association with eNOS [ 59 ]. Interestingly, there is a cross-talk between NO and PPAR- γ pathways as NO has been shown to rapidly and dose-dependently increase PPAR- γ -binding, mediated by p38 MAPK activation [ 60 ]. In vivo, the stimulatory effect of PPAR- γ -agonists rosiglitazone and pioglitazone on endothelial NO-production was associated with increased eNOS-phosphorylation, while eNOS mRNA and total protein levels were not affected [ 51 , 75 ]. Rosiglitazone treatment has been shown to enhance NO-production through enhancement of cellular transport of arginine, the substrate for NO [ 76 ]. This is particularly relevant for renal disease, as arginine transport was found to be markedly impaired in uremic conditions [ 77 – 79 ]. PPAR- γ -agonism favorably affects other endothelium-derived factors that act in conjunction with NO to maintain endothelial homeostasis. The release of C-type natriuretic peptide, another vasodilatory peptide, by endothelial cells is increased by PPAR- γ -agonist treatment [ 39 ]. In addition, PPAR- γ -agonist treatment increased Cu 2+ and Zn 2+ -superoxide dismutase expression in cultured endothelial cells, thereby increasing their potential for oxygen radical scavenging [ 80 , 81 ]. Also, PPAR- γ -agonist treatment decreases the production of radical oxygen species in endothelial cells [ 81 , 82 ], in part through decreasing the expression of subunits of NADPH-oxidase [ 80 , 81 ]. 6. Antihypertensive Effects of PPAR- γ -Agonism Mediated by the Endothelium Improving NO-availability is thought to be a major mechanism mediating the blood pressure lowering effect of PPAR- γ -agonist treatment. Pioglitazone treatment prevented hypertension and renal oxidative stress both by reducing free-radical production and by increasing nitric oxide production [ 72 ]. No blood pressure reduction was seen with PPAR- γ -agonist treatment in rats also receiving NO-inhibitor L-NAME [ 83 ]. However, other mechanisms may play a role in the anti-hypertensive effect of PPAR- γ -agonist treatment, such as effects on the contractility and proliferation of smooth muscle cells [ 84 ]. A naturally occurring antagonist of NO is the vasoconstrictor endothelin-1, which is involved in atherosclerosis and hypertension [ 85 ]. PPAR- γ -activation inhibits the production of endothelin-1 from endothelial cells in vitro [ 39 , 86 – 88 ]. In type 2 diabetic patients, pioglitazone treatment reduced urinary endothelin-1 secretion, along with decreasing microalbuminuria [ 89 ]. Interestingly, in this study, serum endothelin-1 levels were not affected, suggesting a specific pathogenic role for endothelin-1 secretion in the kidney [ 89 ]. In DOCA-salt hypertensive rats, TZD treatment reduced endothelin-1 production and blunted radical oxygen species production with diminished hypertension progression and vascular remodeling [ 90 ]. Several studies show that the inhibitory effect of PPAR- γ -activation on endothelin-1 secretion takes place at the transcriptional level [ 86 , 87 ] and it was found to be NO-dependent [ 87 ]. Delerive et al. demonstrated that PPAR- γ -activation negatively interferes with the activator protein-1 signaling cascade, resulting in inhibition of thrombin-induced transcription of endothelin-1 [ 88 ]. In hypertension, increased production and secondary effects of angiotensin-II play a major role. In endothelial cells, angiotensin-II is a strong inducer of NADPH-oxidase, resulting in the production of radical oxygen species [ 91 ]. Interestingly, there is a cross-talk between the Angiotensin-II and PPAR- γ signaling pathways. Infusion of angiotensin-II downregulates PPAR- γ -receptor expression in the vascular wall [ 92 ] and treatment with PPAR- γ -agonists abrogates many of the angiotensin-II pathophysiological effects [ 93 ]. Of note, angiotensin-II-receptor-1 antagonists have been shown to act as partial PPAR- γ -agonists [ 94 ]. 7. Effects of PPAR- γ -Activation on Inflammatory Cell Recruitment to the Endothelium Endothelial cells form an important barrier between the blood and peripheral tissues and regulate homing, adhesion, and transmigration of inflammatory cells. Upon activation, endothelial cells may further release inflammatory cytokines and express adhesion molecules to attract inflammatory cells. PPAR- γ -agonist treatment inhibits the increased expression of adhesion molecules such as VCAM-1, ICAM-1, and E-selectin and release of inflammatory cytokines upon stimulation of endothelial cells with PMA [ 95 , 96 ], TNF- 𝛼 [ 95 , 97 ], IFN- γ [ 98 ], IL-1 β [ 99 ], LPS [ 96 ], microparticles [ 100 ], and high glucose [ 101 ]. In vitro studies confirmed that this resulted in decreased adhesion of inflammatory cells to the endothelium [ 96 , 97 , 101 , 102 ]. In addition, a recent study showed that in activated primary human brain endothelial cells, PPAR- γ -activation resulted in a marked reduction of monocyte adhesion and in vitro transendothelial migration, mediated by inhibition of Rac1 and RhoA GTPases [ 102 ]. In vivo, troglitazone and 15d-PGJ2 reduced ICAM-1 and VCAM-1 expression on endothelial cells and reduced inflammatory cell homing to atherosclerotic plaques in a mouse model [ 103 ]. 8. Effects of PPAR- γ -Activation on Endothelial Progenitor Cells A recently uncovered effect of PPAR- γ -agonists is the capacity to augment the level and function of endothelial progenitor cells. PPAR- γ -agonist treatment increases endothelial progenitor cell levels in mice [ 104 , 105 ] and humans [ 104 , 106 , 107 ]. A stimulatory effect on endothelial progenitor cell outgrowth was also observed when cultured peripheral blood mononuclear cells were exposed to PPAR- γ -agonist treatment ex vivo [ 104 , 105 , 108 , 109 ], suggesting an effect on endothelial progenitor cell survival, adhesion, or differentiation. PPAR- γ -agonist treatment largely prevented apoptosis of endothelial progenitor cells induced by CRP [ 109 ] and H 2 O 2 [ 105 ]. Based on marker expression patterns, the differentiation of endothelial progenitor cells toward the endothelial lineage appears to be stimulated [ 104 , 108 ], while expression of smooth muscle cell markers is inhibited [ 104 ]. Pioglitazone inhibited detrimental effects of angiotensin-II on endothelial progenitor cells, including inhibiting the induction of cellular senescence of endothelial progenitor cells via downregulation of the expression of angiotensin-II-receptor-1 and limiting the angiotensin-II-induced increased generation of peroxynitrate and superoxide by the NADPH-oxidase subunit gp91phox [ 110 ]. Functionally, PPAR- γ -agonist treatment was shown to stimulate angiogenic network formation by endothelial progenitor cells in vitro [ 109 ] and in vivo [ 105 ]. The adhesion capacity may be increased upon TZD-treatment [ 108 , 109 ]. In addition, endothelial progenitor cell-mediated reendothelialization of a denuded segment of the femoral artery in mice was accelerated in PPAR- γ -agonist treated animals [ 104 ]. As endothelial progenitor cells may also participate in regeneration of damaged renal endothelium [ 26 ], we investigated a potential role for enhanced endothelial progenitor cell homing and glomerular incorporation by PPAR- γ -agonist treatment [ 7 ]. However, using a rat allogenic bone marrow transplantation model, we could not detect an effect on the number of incorporated circulation-derived glomerular endothelial cells in the recovering glomerulus with rosiglitazone treatment, although rosiglitazone did attenuate the clinical course of glomerulonephritis. 9. Conclusions and Perspectives Activation of the PPAR- γ -receptors present on renal endothelial cells affects their cellular behavior. Proliferation, apoptosis, migration, and angiogenesis by endothelial cells are modulated, but may involve both stimulation and inhibition depending on the specific circumstances. It remains unclear how this bimodal potential of PPAR- γ -agonists is regulated. An important consequence of PPAR- γ -receptor activation is the enhanced production of nitric oxide and C-type natriuretic peptide and superoxide dismutase, while endothelin-1 production is inhibited. Together, this improves the capacity of the endothelium to exert vasodilatory, anti-inflammatory, and antioxidative actions, resulting in blood pressure lowering and direct renoprotective effects. In addition, the presentation of adhesion molecules and release of cytokines aimed at recruiting inflammatory cells to activated endothelium is inhibited by PPAR- γ -agonism. This may also in part account for the anti-inflammatory effects of PPAR- γ -agonists, supplementary to direct effects on the inflammatory cells themselves. Finally, PPAR- γ -receptors are also found on endothelial progenitor cells and PPAR- γ -agonist stimulate progenitor-mediated endothelial repair, although definitive evidence that this occurs in the kidney is currently lacking. Activation of other PPAR-receptors besides PPAR- γ may also have beneficial effects on the endothelium. Recently, selective agonists for PPAR- β / δ were developed [ 111 ]. PPAR- β / δ receptors are present on endothelium and stimulation with a pharmacological PPAR- β / δ -agonist was found to increase endothelial cell proliferation and angiogenesis in vitro and in vivo, mainly mediated through VEGF [ 112 ]. Like PPAR- γ -agonists, PPAR- β / δ -agonists increase circulating endothelial progenitor cell levels, augment endothelial progenitor cell function in vitro, and improve endothelial progenitor cell-mediated neovascularization of ischemic tissue [ 113 ]. Also, much like PPAR- γ -agonists, PPAR- β / δ -agonist treatment reduced the expression of adhesion molecules and monocyte binding to activated endothelial cells [ 114 ]. Currently, PPAR- γ -agonist treatment is not standard clinical practice in nondiabetic renal patients. As shown in this review, experimental studies indicate that stimulatory effects of PPAR- γ -agonism on endothelium may provide additional benefit in nondiabetic renal disease. Together with other potentially therapeutic effects independent of the insulin-sensitizing action such as the anti-inflammatory actions, this provides a rationale for further clinical evaluation in nondiabetic renal patients. For patients with diabetic kidney disease, a pressing question is whether glycemic control with a TZD is superior to other antidiabetic drugs for preventing the decline of renal function. To date, only retrospective studies, post hoc analyses, and pilot studies are available to help answer this question. Trials designed to specifically evaluate this question have yet to be performed. Acknowledgments P. E. Westerweel is supported by ZonMw AGIKO Grant no. 2007/12579. M. C. Verhaar is supported by NWO Vidi Grant no. 016.096.359. <h4>References</h4> Y. Guan and M. D. Breyer, “ Peroxisome proliferator-activated receptors (PPARs): novel therapeutic targets in renal disease ,” Kidney International , vol. 60, no. 1, pp. 14–30, 2001. P. A. Sarafidis and G. L. Bakris, “ Protection of the kidney by thiazolidinediones: an assessment from bench to bedside ,” Kidney International , vol. 70, no. 7, pp. 1223–1233, 2006. K. Haraguchi, H. Shimura, and T. Onaya, “ Suppression of experimental crescentic glomerulonephritis by peroxisome proliferator-activated receptor (PPAR) γ activators ,” Clinical and Experimental Nephrology , vol. 7, no. 1, pp. 27–32, 2003. A. Benigni, C. Zoja, S. Tomasoni, et al., “ Transcriptional regulation of nephrin gene by peroxisome proliferator-activated receptor- γ agonist: molecular mechanism of the antiproteinuric effect of pioglitazone ,” Journal of the American Society of Nephrology , vol. 17, no. 6, pp. 1624–1632, 2006. L.-J. Ma, C. Marcantoni, M. F. Linton, S. Fazio, and A. B. Fogo, “ Peroxisome proliferator-activated receptor- γ agonist troglitazone protects against nondiabetic glomerulosclerosis in rats ,” Kidney International , vol. 59, no. 5, pp. 1899–1910, 2001. A. Sivarajah, P. K. Chatterjee, N. S. A. Patel, et al., “ Agonists of peroxisome-proliferator activated receptor-gamma reduce renal ischemia/reperfusion injury ,” The American Journal of Nephrology , vol. 23, no. 4, pp. 267–276, 2003. P. E. Westerweel, K. den Ouden, T. Q. Nguyen, R. Goldschmeding, J. A. Joles, and M. C. Verhaar, “ Amelioration of anti-Thy1-glomerulonephritis by PPAR- γ agonism without increase of endothelial progenitor cell homing ,” The American Journal of Physiology , vol. 294, no. 2, pp. F379–F384, 2008. M. Matsuyama, R. Yoshimura, T. Hase, et al., “ Expression of peroxisome proliferator-activated receptor- γ in renal ischemia-reperfusion injury ,” Transplantation Proceedings , vol. 37, no. 4, pp. 1684–1685, 2005. R. Yoshimura, M. Matsuyama, Y. Segawa, et al., “ Study of peroxisome proliferator-activated receptor (PPAR)- γ in renal ischemia-reperfusion injury ,” Transplantation Proceedings , vol. 36, no. 7, pp. 1946–1948, 2004. Y. Guan, Y. Zhang, A. Schneider, L. Davis, R. M. Breyer, and M. D. Breyer, “Peroxisome proliferator-activated receptor- γ activity is associated with renal microvasculature,” The American Journal of Physiology , vol. 281, no. 6, pp. F1036–F1046, 2001. N. Yamanaka and A. Shimizu, “ Role of glomerular endothelial damage in progressive renal disease ,” Kidney and Blood Pressure Research , vol. 22, no. 1-2, pp. 13–20, 1999. M. E. J. Reinders, T. J. Rabelink, and D. M. Briscoe, “ Angiogenesis and endothelial cell repair in renal disease and allograft rejection ,” Journal of the American Society of Nephrology , vol. 17, no. 4, pp. 932–942, 2006. D. A. Long, W. Mu, K. L. Price, and R. J. Johnson, “ Blood vessels and the aging kidney ,” Nephron Experimental Nephrology , vol. 101, no. 3, pp. e95–e99, 2005. J. P. Garg and G. L. Bakris, “ Microalbuminuria: marker of vascular dysfunction, risk factor for cardiovascular disease ,” Vascular Medicine , vol. 7, no. 1, pp. 35–43, 2002. G. L. Bakris, “ Clinical importance of microalbuminuria in diabetes and hypertension ,” Current Hypertension Reports , vol. 6, no. 5, pp. 352–356, 2004. S. Gschwend, H. Buikema, G. Navis, R. H. Henning, D. de Zeeuw, and R. P. E. van Dokkum, “ Endothelial dilatory function predicts individual susceptibility to renal damage in the 5/6 nephrectomized rat ,” Journal of the American Society of Nephrology , vol. 13, no. 12, pp. 2909–2915, 2002. A. Shimizu, H. Kitamura, Y. Masuda, M. Ishizaki, Y. Sugisaki, and N. Yamanaka, “Rare glomerular capillary regeneration and subsequent capillary regression with endothelial cell apoptosis in progressive glomerulonephritis,” The American Journal of Pathology , vol. 151, no. 5, pp. 1231–1239, 1997. L. Iruela-Arispe, K. Gordon, C. Hugo, et al., “Participation of glomerular endothelial cells in the capillary repair of glomerulonephritis,” The American Journal of Pathology , vol. 147, no. 6, pp. 1715–1727, 1995. Y. Masuda, A. Shimizu, T. Mori, et al., “Vascular endothelial growth factor enhances glomerular capillary repair and accelerates resolution of experimentally induced glomerulonephritis,” The American Journal of Pathology , vol. 159, no. 2, pp. 599–608, 2001. K. Miyamoto, Y. Kitamoto, H. Tokunaga, et al., “ Protective effect of vascular endothelial growth factor/vascular permeability factor 165 and 121 on glomerular endothelial cell injury in the rat ,” Laboratory Investigation , vol. 84, no. 9, pp. 1126–1136, 2004. T. Ostendorf, U. Kunter, F. Eitner, et al., “ VEGf 165 mediates glomerular endothelial repair ,” The Journal of Clinical Investigation , vol. 104, no. 7, pp. 913–923, 1999. D.-H. Kang, J. Hughes, M. Mazzali, G. F. Schreiner, and R. J. Johnson, “Impaired angiogenesis in the remnant kidney model—part II: vascular endothelial growth factor administration reduces renal fibrosis and stabilizes renal function,” Journal of the American Society of Nephrology , vol. 12, no. 7, pp. 1448–1457, 2001. D. H. Kang, A. H. Joly, S. W. Oh, et al., “Impaired angiogenesis in the remnant kidney model—part I: potential role of vascular endothelial growth factor and thrombospondin-1,” Journal of the American Society of Nephrology , vol. 12, no. 7, pp. 1434–1447, 2001. M. B. Rookmaaker, H. Tolboom, R. Goldschmeding, J.-J. Zwaginga, T. J. Rabelink, and M. C. Verhaar, “ Bone marrow-derived cells contribute to endothelial repair after thrombotic microangiopathy ,” Blood , vol. 99, no. 3, p. 1095, 2002. M. B. Rookmaaker, A. M. Smits, H. Tolboom, et al., “Bone-marrow-derived cells contribute to glomerular endothelial repair in experimental glomerulonephritis,” The American Journal of Pathology , vol. 163, no. 2, pp. 553–562, 2003. M. B. Rookmaaker, M. C. Verhaar, A. J. Van Zonneveld, and T. J. Rabelink, “ Progenitor cells in the kidney: biology and therapeutic perspectives ,” Kidney International , vol. 66, no. 2, pp. 518–522, 2004. E. L. Lagaaij, G. F. Cramer-Knijnenburg, F. J. van Kemenade, L. A. van Es, J. A. Bruijn, and J. H. J. M. van Krieken, “ Endothelial cell chimerism after renal transplantation and vascular rejection ,” The Lancet , vol. 357, no. 9249, pp. 33–37, 2001. H. Peters, U. Daig, S. Martini, et al., “ No mediates antifibrotic actions of L-arginine supplementation following induction of anti-thy1 glomerulonephritis ,” Kidney International , vol. 64, no. 2, pp. 509–518, 2003. B. Li, T. Morioka, M. Uchiyama, and T. Oite, “ Bone marrow cell infusion ameliorates progressive glomerulosclerosis in an experimental rat model ,” Kidney International , vol. 69, no. 2, pp. 323–330, 2006. H. Uchimura, T. Marumo, O. Takase, et al., “ Intrarenal injection of bone marrow-derived angiogenic cells reduces endothelial injury and mesangial cell activation in experimental glomerulonephritis ,” Journal of the American Society of Nephrology , vol. 16, no. 4, pp. 997–1004, 2005. I. Barroso, M. Gurnell, V. E. F. Crowley, et al., “ Dominant negative mutations in human PPAR γ associated with severe insulin resistance, diabetes mellitus and hypertension ,” Nature , vol. 402, no. 6764, pp. 880–883, 1999. P. D. G. Miles, Y. Barak, W. He, R. M. Evans, and J. M. Olefsky, “ Improved insulin-sensitivity in mice heterozygous for PPAR- γ deficiency ,” The Journal of Clinical Investigation , vol. 105, no. 3, pp. 287–292, 2000. T. Yamauchi, J. Kamon, H. Waki, et al., “ The mechanisms by which both heterozygous peroxisome proliferator-activated receptor γ (PPAR γ ) deficiency and PPAR γ agonist improve insulin resistance ,” The Journal of Biological Chemistry , vol. 276, no. 44, pp. 41245–41254, 2001. Y. Barak, M. C. Nelson, E. S. Ong, et al., “ PPAR γ is required for placental, cardiac, and adipose tissue development ,” Molecular Cell , vol. 4, no. 4, pp. 585–595, 1999. S. Z. Duan, C. Y. Ivashchenko, S. E. Whitesall, et al., “ Hypotension, lipodystrophy, and insulin resistance in generalized PPAR γ -deficient mice rescued from embryonic lethality ,” The Journal of Clinical Investigation , vol. 117, no. 3, pp. 812–822, 2007. C. J. Nicol, M. Adachi, T. E. Akiyama, and F. J. Gonzalez, “ PPAR γ in endothelial cells influences high fat diet-induced hypertension ,” The American Journal of Hypertension , vol. 18, no. 4, pp. 549–556, 2005. A. M. Beyer, G. L. Baumbach, C. M. Halabi, et al., “ Interference with PPAR γ signaling causes cerebral vascular dysfunction, hypertrophy, and remodeling ,” Hypertension , vol. 51, no. 4, pp. 867–871, 2008. R. K. Semple, A. Meirhaeghe, A. J. Vidal-Puig, et al., “ A dominant negative human peroxisome proliferator-activated receptor (PPAR) α is a constitutive transcriptional corepressor and inhibits signaling through all PPAR isoforms ,” Endocrinology , vol. 146, no. 4, pp. 1871–1882, 2005. Y. Fukunaga, H. Itoh, K. Doi, et al., “ Thiazolidinediones, peroxisome proliferator-activated receptor γ agonists, regulate endothelial cell growth and secretion of vasoactive peptides ,” Atherosclerosis , vol. 158, no. 1, pp. 113–119, 2001. S. T. de Dios, K. M. Hannan, R. J. Dilley, M. A. Hill, and P. J. Little, “ Troglitazone, but not rosiglitazone, inhibits Na/H exchange activity and proliferation of macrovascular endothelial cells ,” Journal of Diabetes and Its Complications , vol. 15, no. 3, pp. 120–127, 2001. H. K. Hong, Y. M. Cho, K.-H. Park, C.-T. Lee, H. K. Lee, and K. S. Park, “ Peroxisome proliferator-activated receptor gamma mediated inhibition of plasminogen activator inhibitor type 1 production and proliferation of human umbilical vein endothelial cells ,” Diabetes Research and Clinical Practice , vol. 62, no. 1, pp. 1–8, 2003. W. H.-H. Sheu, H.-C. Ou, F.-P. Chou, T.-M. Lin, and C.-H. Yang, “ Rosiglitazone inhibits endothelial proliferation and angiogenesis ,” Life Sciences , vol. 78, no. 13, pp. 1520–1528, 2006. S. Goetze, F. Eilers, A. Bungenstock, et al., “ PPAR activators inhibit endothelial cell migration by targeting Akt ,” Biochemical and Biophysical Research Communications , vol. 293, no. 5, pp. 1431–1437, 2002. X. Xin, S. Yang, J. Kowalski, and M. E. Gerritsen, “ Peroxisome proliferator-activated receptor γ ligands are potent inhibitors of angiogenesis in vitro and in vivo ,” The Journal of Biological Chemistry , vol. 274, no. 13, pp. 9116–9121, 1999. F. Biscetti, E. Gaetani, A. Flex, et al., “ Selective activation of peroxisome proliferator-activated receptor (PPAR) α and PPAR γ induces neoangiogenesis through a vascular endothelial growth factor-dependent mechanism ,” Diabetes , vol. 57, no. 5, pp. 1394–1404, 2008. M. Artwohl, T. Hölzebein, C. Fürnsinn, et al., “ Thiazolidinediones inhibit apoptosis and heat shock protein 60 expression in human vascular endothelial cells ,” Thrombosis and Haemostasis , vol. 93, no. 5, pp. 810–815, 2005. K. M. Hannan, R. J. Dilley, S. T. de Dios, and P. J. Little, “ Troglitazone stimulates repair of the endothelium and inhibits neointimal formation in denuded rat aorta ,” Arteriosclerosis, Thrombosis, and Vascular Biology , vol. 23, no. 5, pp. 762–768, 2003. D. Bishop-Bailey and T. Hla, “ Endothelial cell apoptosis induced by the peroxisome proliferator-activated receptor (PPAR) ligand 15-deoxy- Δ 12 , 14 -prostaglandin J 2 ,” The Journal of Biological Chemistry , vol. 274, no. 24, pp. 17042–17048, 1999. A.-L. Levonen, D. A. Dickinson, D. R. Moellering, R. T. Mulcahy, H. J. Forman, and V. M. Darley-Usmar, “ Biphasic effects of 15-deoxy- Δ 12 , 14 -prostaglandin J 2 on glutathione induction and apoptosis in human endothelial cells ,” Arteriosclerosis, Thrombosis, and Vascular Biology , vol. 21, no. 11, pp. 1846–1851, 2001. H. Migita and J. Morser, “ 15-deoxy- Δ 12 , 14 -postaglandin J 2 (15d-PGJ2) signals through retinoic acid receptor-related orphan receptor- α but not peroxisome proliferator-activated receptor- γ in human vascular endothelial cells: the effect of 15d-PGJ2 on tumor necrosis factor- α -induced gene expression ,” Arteriosclerosis, Thrombosis, and Vascular Biology , vol. 25, no. 4, pp. 710–716, 2005. P.-H. Huang, M. Sata, H. Nishimatsu, M. Sumi, Y. Hirata, and R. Nagai, “ Pioglitazone ameliorates endothelial dysfunction and restores ischemia-induced angiogenesis in diabetic mice ,” Biomedicine and Pharmacotherapy , vol. 62, no. 1, pp. 46–52, 2008. K. Chu, S.-T. Lee, J.-S. Koo, et al., “ Peroxisome proliferator-activated receptor- γ -agonist, rosiglitazone, promotes angiogenesis after focal cerebral ischemia ,” Brain Research , vol. 1093, no. 1, pp. 208–218, 2006. T. Murata, S. He, M. Hangai, et al., “Peroxisome proliferator-activated receptor- γ ligands inhibit choroidal neovascularization,” Investigative Ophthalmology and Visual Science , vol. 41, no. 8, pp. 2309–2317, 2000. D. Panigrahy, S. Singer, L. Q. Shen, et al., “ PPAR γ ligands inhibit primary tumor growth and metastasis by inhibiting angiogenesis ,” The Journal of Clinical Investigation , vol. 110, no. 7, pp. 923–932, 2002. V. G. Keshamouni, D. A. Arenberg, R. C. Reddy, M. J. Newstead, S. Anthwal, and T. J. Standiford, “ PPAR- γ activation inhibits angiogenesis by blocking ELR+CXC chemokine production in non-small cell lung cancer ,” Neoplasia , vol. 7, no. 3, pp. 294–301, 2005. M. C. Verhaar and T. J. Rabelink, “ Endothelial function: strategies for early intervention ,” Cardiovascular Drugs and Therapy , vol. 12, pp. 125–134, 1998. D. S. Calnek, L. Mazzella, S. Roser, J. Roman, and C. M. Hart, “ Peroxisome proliferator-activated receptor γ ligands increase release of nitric oxide from endothelial cells ,” Arteriosclerosis, Thrombosis, and Vascular Biology , vol. 23, no. 1, pp. 52–57, 2003. K. Goya, S. Sumitani, M. Otsuki, et al., “ The thiazolidinedione drug troglitazone up-regulates nitric oxide synthase expression in vascular endothelial cells ,” Journal of Diabetes and Its Complications , vol. 20, no. 5, pp. 336–342, 2006. J. A. Polikandriotis, L. J. Mazzella, H. L. Rupnow, and C. M. Hart, “ Peroxisome proliferator-activated receptor γ ligands stimulate endothelial nitric oxide production through distinct peroxisome proliferator-activated receptor γ -dependent mechanisms ,” Arteriosclerosis, Thrombosis, and Vascular Biology , vol. 25, no. 9, pp. 1810–1816, 2005. A. Ptasinska, S. Wang, J. Zhang, R. A. Wesley, and R. L. Danner, “ Nitric oxide activation of peroxisome proliferator-activated receptor gamma through a p38 MAPK signaling pathway ,” The FASEB Journal , vol. 21, no. 3, pp. 950–961, 2007. J. B. Majithiya, A. N. Paramar, and R. Balaraman, “ Pioglitazone, a PPAR γ agonist, restores endothelial function in aorta of streptozotocin-induced diabetic rats ,” Cardiovascular Research , vol. 66, no. 1, pp. 150–161, 2005. S. Fujishima, Y. Ohya, Y. Nakamura, U. Onaka, I. Abe, and M. Fujishima, “ Troglitazone, an insulin sensitizer, increases forearm blood flow in humans ,” The American Journal of Hypertension , vol. 11, no. 9, pp. 1134–1137, 1998. U. Campia, L. A. Matuskey, and J. A. Panza, “ Peroxisome proliferator-activated receptor- γ activation with pioglitazone improves endothelium-dependent dilation in nondiabetic patients with major cardiovascular risk factors ,” Circulation , vol. 113, no. 6, pp. 867–875, 2006. M. N. Ghazzi, J. E. Perez, T. K. Antonucci, et al., “ Cardiac and glycemic benefits of troglitazone treatment in NIDDM. The Troglitazone Study Group ,” Diabetes , vol. 46, no. 3, pp. 433–439, 1997. B. H. Sung, J. L. Izzo, Jr., P. Dandona, and M. F. Wilson, “Vasodilatory effects of troglitazone improve blood pressure at rest and during mental stress in type 2 diabetes mellitus,” Hypertension , vol. 34, no. 1, pp. 83–88, 1999. T. Murakami, S. Mizuno, K. Ohsato, et al., “ Effects of troglitazone on frequency of coronary vasospastic-induced angina pectoris in patients with diabetes mellitus ,” The American Journal of Cardiology , vol. 84, no. 1, pp. 92–94, 1999. A. I. Vinik, K. B. Stansberry, and P. M. Barlow, “ Rosiglitazone treatment increases nitric oxide production in human peripheral skin: a controlled clinical trial in patients with type 2 diabetes mellitus ,” Journal of Diabetes and Its Complications , vol. 17, no. 5, pp. 279–285, 2003. T. Nakamura, T. Matsuda, Y. Kawagoe, et al., “ Effect of pioglitazone on carotid intima-media thickness and arterial stiffness in type 2 diabetic nephropathy patients ,” Metabolism , vol. 53, no. 10, pp. 1382–1386, 2004. M. H. Voytovich, C. Simonsen, T. Jenssen, J. Hjelmesæth, A. Åsberg, and A. Hartmann, “ Short-term treatment with rosiglitazone improves glucose tolerance, insulin sensitivity and endothelial function in renal transplant recipients ,” Nephrology Dialysis Transplantation , vol. 20, no. 2, pp. 413–418, 2005. F. Pistrosch, K. Herbrig, B. Kindel, J. Passauer, S. Fischer, and P. Gross, “ Rosiglitazone improves glomerular hyperfiltration, renal endothelial dysfunction, and microalbuminuria of incipient diabetic nephropathy in patients ,” Diabetes , vol. 54, no. 7, pp. 2206–2211, 2005. K. Fujiwara, K. Hayashi, H. Matsuda, et al., “Altered pressure-natriuresis in obese Zucker rats,” Hypertension , vol. 33, no. 6, pp. 1470–1475, 1999. A. D. Dobrian, S. D. Schriver, A. A. Khraibi, and R. L. Prewitt, “ Pioglitazone prevents hypertension and reduces oxidative stress in diet-induced obesity ,” Hypertension , vol. 43, no. 1, pp. 48–56, 2004. R. Govers and T. J. Rabelink, “Cellular regulation of endothelial nitric oxide synthase,” The American Journal of Physiology , vol. 280, no. 2, pp. F193–F206, 2001. B. Braam and M. C. Verhaar, “ Understanding eNOS for pharmacological modulation of endothelial function: a translational view ,” Current Pharmaceutical Design , vol. 13, no. 17, pp. 1727–1740, 2007. A. T. Gonon, A. Bulhak, F. Labruto, P.-O. Sjöquist, and J. Pernow, “ Cardioprotection mediated by rosiglitazone, a peroxisome proliferator activated receptor gamma ligand, in relation to nitric oxide ,” Basic Research in Cardiology , vol. 102, no. 1, pp. 80–89, 2007. M. Ingbir, I. F. Schwartz, A. Shtabsky, et al., “ Rosiglitazone improves aortic arginine transport, through inhibition of PKC α , in uremic rats ,” The American Journal of Physiology , vol. 295, no. 2, pp. F471–F477, 2008. I. F. Schwartz, R. Ayalon, T. Chernichovski, et al., “ Arginine uptake is attenuated through modulation of cationic amino-acid transporter-1, in uremic rats ,” Kidney International , vol. 69, no. 2, pp. 298–303, 2006. L. Wagner, J. D. Klein, J. M. Sands, and C. Baylis, “ Urea transporters are distributed in endothelial cells and mediate inhibition of L-arginine transport ,” The American Journal of Physiology , vol. 283, no. 3, pp. F578–F582, 2002. S. Xiao, L. Wagner, J. Mahaney, and C. Baylis, “Uremic levels of urea inhibit L-arginine transport in cultured endothelial cells,” The American Journal of Physiology , vol. 280, no. 6, pp. F989–F995, 2001. I. Inoue, S.-I. Goto, T. Matsunaga, et al., “ The ligands/activators for peroxisome proliferator-activated receptor α (PPAR α ) and PPAR γ increase Cu 2 + , Zn 2 + -superoxide dismutase and decrease p22phox message expressions in primary endothelial cells ,” Metabolism , vol. 50, no. 1, pp. 3–11, 2001. J. Hwang, D. J. Kleinhenz, B. Lassègue, K. K. Griendling, S. Dikalov, and C. M. Hart, “ Peroxisome proliferator-activated receptor- γ ligands regulate endothelial membrane superoxide production ,” The American Journal of Physiology , vol. 288, no. 4, pp. C899–C905, 2005. J. L. Mehta, B. Hu, J. Chen, and D. Li, “ Pioglitazone inhibits LOX-1 expression in human coronary artery endothelial cells by reducing intracellular superoxide radical generation ,” Arteriosclerosis, Thrombosis, and Vascular Biology , vol. 23, no. 12, pp. 2203–2208, 2003. J. B. Majithiya, A. N. Parmar, C. J. Trivedi, and R. Balaraman, “ Effect of pioglitazone on L-NAME induced hypertension in diabetic rats ,” Vascular Pharmacology , vol. 43, no. 4, pp. 260–266, 2005. P. A. Sarafidis and A. N. Lasaridis, “ Actions of peroxisome proliferator-activated receptors- γ agonists explaining a possible blood pressure-lowering effect ,” The American Journal of Hypertension , vol. 19, no. 6, pp. 646–653, 2006. E. L. Schiffrin, “ Role of endothelin-1 in hypertension and vascular disease ,” The American Journal of Hypertension , vol. 14, no. 6, part 2, pp. 83S–89S, 2001. F. Martin-Nizard, C. Furman, P. Delerive, et al., “ Peroxisome proliferator-activated receptor activators inhibit oxidized low-density lipoprotein-induced endothelin-1 secretion in endothelial cells ,” Journal of Cardiovascular Pharmacology , vol. 40, no. 6, pp. 822–831, 2002. H. Satoh, K. Tsukamoto, Y. Hashimoto, et al., “ Thiazolidinediones suppress endothelin-1 secretion from bovine vascular endothelial cells: a new possible role of PPAR γ on vascular endothelial function ,” Biochemical and Biophysical Research Communications , vol. 254, no. 3, pp. 757–763, 1999. P. Delerive, F. Martin-Nizard, G. Chinetti, et al., “Peroxisome proliferator-activated receptor activators inhibit thrombin-induced endothelin-1 production in human vascular endothelial cells by inhibiting the activator protein-1 signaling pathway,” Circulation Research , vol. 85, no. 5, pp. 394–402, 1999. T. Nakamura, C. Ushiyama, N. Shimada, K. Hayashi, I. Ebihara, and H. Koide, “ Comparative effects of pioglitazone, glibenclamide, and voglibose on urinary endothelin-1 and albumin excretion in diabetes patients ,” Journal of Diabetes and Its Complications , vol. 14, no. 5, pp. 250–254, 2000. M. Iglarz, R. M. Touyz, F. Amiri, M.-F. Lavoie, Q. N. Diep, and E. L. Schiffrin, “ Effect of peroxisome proliferator-activated receptor- α and - γ activators on vascular remodeling in endothelin-dependent hypertension ,” Arteriosclerosis, Thrombosis, and Vascular Biology , vol. 23, no. 1, pp. 45–51, 2003. T. Münzel, U. Hink, T. Heitzer, and T. Meinertz, “ Role for NADPH/NADH oxidase in the modulation of vascular tone ,” Annals of the New York Academy of Sciences , vol. 874, pp. 386–400, 1999. D. M. Tham, B. Martin-McNulty, Y.-X. Wang, et al., “Angiotensin II is associated with activation of NF- κ B-mediated genes and downregulation of PPARs,” Physiological Genomics , vol. 11, no. 1, pp. 21–30, 2003. Q. N. Diep, M. El Mabrouk, J. S. Cohn, et al., “ Structure, endothelial function, cell growth, and inflammation in blood vessels of angiotensin II-infused rats: role of peroxisome proliferator-activated receptor- γ ,” Circulation , vol. 105, no. 19, pp. 2296–2302, 2002. U. Kintscher and T. Unger, “ Vascular protection in diabetes: a pharmacological view of angiotensin II type 1 receptor blockers ,” Acta Diabetologica , vol. 42, pp. S26–S32, 2005. N. Wang, L. Verna, N.-G. Chen, et al., “ Constitutive activation of peroxisome proliferator-activated receptor- γ suppresses pro-inflammatory adhesion molecules in human vascular endothelial cells ,” The Journal of Biological Chemistry , vol. 277, no. 37, pp. 34176–34181, 2002. S. M. Jackson, F. Parhami, X.-P. Xi, et al., “Peroxisome proliferator-activated receptor activators target human endothelial cells to inhibit leukocyte-endothelial cell interaction,” Arteriosclerosis, Thrombosis, and Vascular Biology , vol. 19, no. 9, pp. 2094–2104, 1999. Y. Jung, S. Song, and C. Choi, “ Peroxisome proliferator activated receptor γ agonists suppress TNF α -induced ICAM-1 expression by endothelial cells in a manner potentially dependent on inhibition of reactive oxygen species ,” Immunology Letters , vol. 117, no. 1, pp. 63–69, 2008. N. Marx, F. Mach, A. Sauty, et al., “Peroxisome proliferator-activated receptor- γ activators inhibit IFN- γ -induced expression of the T cell-active CXC chemokines IP-10, Mig, and I-TAC in human endothelial cells,” The Journal of Immunology , vol. 164, no. 12, pp. 6503–6508, 2000. E. Imamoto, N. Yoshida, K. Uchiyama, et al., “ Inhibitory effect of pioglitazone on expression of adhesion molecules on neutrophils and endothelial cells ,” BioFactors , vol. 20, no. 1, pp. 37–47, 2004. A. Tesse, G. Al-Massarani, R. Wangensteen, S. Reitenbach, M. C. Martínez, and R. Andriantsitohaina, “ Rosiglitazone, a peroxisome proliferator-activated receptor- γ agonist, prevents microparticle-induced vascular hyporeactivity through the regulation of proinflammatory proteins ,” Journal of Pharmacology and Experimental Therapeutics , vol. 324, no. 2, pp. 539–547, 2008. E. Verrier, L. Wang, C. Wadham, et al., “ PPAR γ agonists ameliorate endothelial cell activation via inhibition of diacylglycerol-protein kinase C signaling pathway: role of diacylglycerol kinase ,” Circulation Research , vol. 94, no. 11, pp. 1515–1522, 2004. S. H. Ramirez, D. Heilman, B. Morsey, R. Potula, J. Haorah, and Y. Persidsky, “Activation of peroxisome proliferator-activated receptor gamma (PPARgamma) suppresses Rho GTPases in human brain microvascular endothelial cells and inhibits adhesion and transendothelial migration of HIV-1 infected monocytes,” The Journal of Immunology , vol. 180, no. 3, pp. 1854–1865, 2008. V. Pasceri, H. D. Wu, J. T. Willerson, and E. T. H. Yeh, “Modulation of vascular inflammation in vitro and in vivo by peroxisome proliferator-activated receptor- γ activators,” Circulation , vol. 101, no. 3, pp. 235–238, 2000. C.-H. Wang, N. Ciliberti, S.-H. Li, et al., “ Rosiglitazone facilitates angiogenic progenitor cell differentiation toward endothelial lineage: a new paradigm in glitazone pleiotropy ,” Circulation , vol. 109, no. 11, pp. 1392–1400, 2004. C. Gensch, Y. P. Clever, C. Werner, M. Hanhoun, M. Böhm, and U. Laufs, “ The PPAR- γ agonist pioglitazone increases neoangiogenesis and prevents apoptosis of endothelial progenitor cells ,” Atherosclerosis , vol. 192, no. 1, pp. 67–74, 2007. F. Pistrosch, K. Herbrig, U. Oelschlaegel, et al., “ PPAR γ -agonist rosiglitazone increases number and migratory activity of cultured endothelial progenitor cells ,” Atherosclerosis , vol. 183, no. 1, pp. 163–167, 2005. C. Werner, C. H. Kamani, C. Gensch, M. Böhm, and U. Laufs, “ The peroxisome proliferator-activated receptor- γ agonist pioglitazone increases number and function of endothelial progenitor cells in patients with coronary artery disease and normal glucose tolerance ,” Diabetes , vol. 56, no. 10, pp. 2609–2615, 2007. S. Redondo, M. Hristov, D. Gümbel, T. Tejerina, and C. Weber, “ Biphasic effect of pioglitazone on isolated human endothelial progenitor cells: involvement of peroxisome proliferator-activated receptor- γ and transforming growth factor- β 1 ,” Thrombosis and Haemostasis , vol. 97, no. 6, pp. 979–987, 2007. S. Verma, M. A. Kuliszewski, S.-H. Li, et al., “ C-reactive protein attenuates endothelial progenitor cell survival, differentiation, and function: further evidence of a mechanistic link between C-reactive protein and cardiovascular disease ,” Circulation , vol. 109, no. 17, pp. 2058–2067, 2004. T. Imanishi, K. Kobayashi, A. Kuroi, H. Ikejima, and T. Akasaka, “ Pioglitazone inhibits angiotensin II-induced senescene of endothelial progenitor cell ,” Hypertension Research , vol. 31, no. 4, pp. 757–765, 2008. D. Bishop-Bailey, “ Peroxisome proliferator-activated receptor β / δ goes vascular ,” Circulation Research , vol. 102, no. 2, pp. 146–147, 2008. L. Piqueras, A. R. Reynolds, K. M. Hodivala-Dilke, et al., “ Activation of PPAR β / δ induces endothelial cell proliferation and angiogenesis ,” Arteriosclerosis, Thrombosis, and Vascular Biology , vol. 27, no. 1, pp. 63–69, 2007. J. K. Han, H. S. Lee, H. M. Yang, et al., “ Peroxisome proliferator-activated receptor-delta agonist enhances vasculogenesis by regulating endothelial progenitor cells through genomic and nongenomic activations of the phosphatidylinositol 3-kinase/Akt pathway ,” Circulation , vol. 118, no. 10, pp. 1021–1033, 2008. Y. Rival, N. Benéteau, T. Taillandier, et al., “ PPAR α and PPAR δ activators inhibit cytokine-induced nuclear translocation of NF- κ B and expression of VCAM-1 in EAhy926 endothelial cells ,” European Journal of Pharmacology , vol. 435, no. 2-3, pp. 143–151, 2002. // http://www.deepdyve.com/assets/images/DeepDyve-Logo-lg.png PPAR Research Hindawi Publishing Corporation

Protective Actions of PPAR- Activation in Renal Endothelium

PPAR Research , Volume 2008 (2008) – Feb 26, 2009

Loading next page...
 
/lp/hindawi-publishing-corporation/protective-actions-of-ppar-activation-in-renal-endothelium-f6iIJdkiwK

References

References for this paper are not available at this time. We will be adding them shortly, thank you for your patience.

Publisher
Hindawi Publishing Corporation
Copyright
Copyright © 2008 Peter E. Westerweel and Marianne C. Verhaar.
ISSN
1687-4757
eISSN
1687-4765
Publisher site
See Article on Publisher Site

Abstract

Protective Actions of PPAR-γ Activation in Renal Endothelium <meta name="citation_title" content="Protective Actions of PPAR- γ Activation in Renal Endothelium" /> //// Hindawi Publishing Corporation Home Journals About Us About this Journal Submit a Manuscript Table of Contents Journal Menu Abstracting and Indexing Aims and Scope Article Processing Charges Articles in Press Author Guidelines Bibliographic Information Contact Information Editorial Board Editorial Workflow Free eTOC Alerts Reviewers Acknowledgment Subscription Information Open Focus Issues Focus Issue Guidelines Open Special Issues Published Special Issues Special Issue Guidelines Abstract Full-Text PDF Full-Text HTML Linked References How to Cite this Article PPAR Research Volume 2008 (2008), Article ID 635680, 9 pages doi:10.1155/2008/635680 Review Article Protective Actions of PPAR- 𝛾 Activation in Renal Endothelium Peter E. Westerweel 1,2 and Marianne C. Verhaar 1 1 Department of Vascular Medicine, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands 2 Department of Internal Medicine, St. Antonius Hospital, Koekoekslaan 1, 3435 CM Nieuwegein, The Netherlands Received 15 September 2008; Accepted 8 December 2008 Academic Editor: Nigel Brunskill Copyright © 2008 Peter E. Westerweel and Marianne C. Verhaar. This is an open access article distributed under the Creative Commons Attribution License , which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Abstract Renal endothelial damage is pivotal in the initiation and progression of renal disease. Damaged renal endothelium may be regenerated through proliferation of local endothelium and circulation-derived endothelial progenitor cells. Activation of the PPAR- 𝛾 -receptors present on endothelial cells affects their cellular behavior. Proliferation, apoptosis, migration, and angiogenesis by endothelial cells are modulated, but may involve both stimulation and inhibition depending on the specific circumstances. PPAR- 𝛾 -receptor activation stimulates the production of nitric oxide, C-type natriuretic peptide, and superoxide dismutase, while endothelin-1 production is inhibited. Together, they augment endothelial function, resulting in blood pressure lowering and direct renoprotective effects. The presentation of adhesion molecules and release of cytokines recruiting inflammatory cells are inhibited by PPAR- 𝛾 -agonism. Finally, PPAR- 𝛾 -receptors are also found on endothelial progenitor cells and PPAR- 𝛾 -agonists stimulate progenitor-mediated endothelial repair. Together, the stimulatory effects of PPAR- 𝛾 -agonism on endothelium make an important contribution to the beneficial actions of PPAR- 𝛾 -agonists on renal disease. 1. Introduction PPAR- γ -agonists are widely used for their insulin-sensitizing actions in the treatment of type 2 diabetes mellitus, but have additional therapeutic potential beyond the metabolic effects. Currently, the clinically most used PPAR- γ -agonistic drugs are the thiazolidinediones (TZDs). PPAR- γ -agonists may favorably affect the course of renal disease in both diabetic and nondiabetic conditions [ 1 , 2 ]. In nondiabetic animals, beneficial effects have been shown for anti-GBM antibody-induced crescentic glomerulonephritis [ 3 ], passive Heymann nephritis [ 4 ], the development of glomerulosclerosis after 5/6 nephrectomy [ 5 ], renal ischemia-reperfusion induced damage [ 6 ], and anti-Thy-1-glomerulonephritis [ 7 ]. One potential mechanism is the modulation of endothelial cell function through activation of PPAR- γ receptors, which are expressed on glomerular endothelium [ 8 , 9 ]. In response to injury, the endothelial expression of PPAR- γ -receptors may be increased, for example, as transiently occurs after ischemia- reperfusion [ 8 , 9 ]. Treatment with PPAR- γ -agonists also increases the expression of PPAR- γ -receptors on renal endothelium in both the glomerulus and the capillary endothelium of the medullary vasa recta [ 10 ]. The relevance for renal disease of activating the PPAR- γ -receptors on renal endothelium is becoming increasingly clear and is the focus of this review. 2. The Role of Endothelium in Renal Disease Renal microvascular endothelial injury is a pivotal pathogenic factor for various renal diseases. Renal disease conditions involving prominent endothelial damage include ischemic nephropathy, glomerulonephritis, interstitial nephritis, and allograft rejection [ 11 , 12 ]. Endothelial dysfunction and attenuated angiogenesis contribute to declining renal function with ageing [ 13 ] and the pathogenesis and progression of chronic kidney disease [ 11 ]. The microvascular endothelium, by the release of endothelium-derived factors such as nitric oxide (NO) and as a critical component of the glomerular filtration barrier, exerts important protection against progressive renal damage. Endothelial dysfunction results in increased permeability causing passage of macromolecules (microalbuminuria), which is considered to be the earliest renal sign of vascular dysfunction [ 14 , 15 ]. Endothelial dysfunction has been shown to predict susceptibility to renal damage in a rat renal injury model [ 16 ]. Progression of renal disease does not only depend on the degree of microvascular endothelial injury, but also on the effectiveness of endothelial repair. Impaired glomerular capillary repair was found to be associated with the development of glomerulosclerosis and renal failure [ 17 ]. During experimental glomerulonephritis, angiogenic factors such as VEGF and bFGF are released, which stimulate endothelial regeneration [ 18 – 20 ]. Blocking the VEGF-induced endothelial repair with a VEGF-antagonist interferes with renal recovery and results in progressive renal impairment [ 21 ]. Consistently, progressive renal disease is associated with reduced expression of angiogenic growth factors and enhanced expression of antiangiogenic factors [ 22 , 23 ]. The glomerular endothelium can recover from injury by replacing lost or damaged endothelial cells, in part through proliferation of local endothelium, stimulated by the release of angiogenic growth factors [ 11 , 12 ]. We [ 24 – 26 ] and others [ 27 ] have observed in both human and experimental animal studies that damaged glomerular endothelium may also be regenerated from circulating bone marrow-derived endothelial progenitor cells. Endothelial progenitor cells incorporate into the damaged glomerulus, differentiate into mature endothelial cells, and eventually fully integrate into the resident endothelium [ 26 ]. Enhancing renal endothelial repair offers therapeutic potential. Stimulating angiogenesis with VEGF-treatment augments capillary repair and renal recovery after glomerulonephritis [ 19 ]. Enhancing NO production by supplementing the substrate L-arginine improves the clinical course of anti-Thy-1-glomerulonephritis [ 28 ]. Infusion of unselected bone marrow cells ameliorates experimental progressive glomerulosclerosis [ 29 ]. Intrarenal administration of endothelial progenitor cells attenuates endothelial injury and mesangial activation in experimental glomerulonephritis [ 30 ]. 3. The Role of PPAR- γ -Receptors in Vascular Development and Remodeling Phenotypical studies in humans and animals with genetic mutations in the PPAR- γ -receptor imply a role in the regulation of vascular function and remodeling. In humans with dominant-negative heterozygous mutations in the PPAR- γ -receptor causing an impaired capacity for transcriptional activation, diabetes and also hypertension occur at an unusually young age [ 31 ]. Heterozygous knockout mice have increased insulin sensitivity [ 32 ] and decreased fat mass [ 33 ], but do not have a vascular phenotype [ 34 ]. However, but PPAR- γ -null mice are embryonically lethal due to placental dysfunction, characterized by defective trophoblast differentiation and markedly impaired placental vascularization [ 34 ]. A surviving PPAR- γ -knockout mouse that was supplemented with a wild-type placenta developed an apparently normal vascular system during further embryogenesis, but died some days after birth due to a combination of pathologies, including severe lipodystrophic changes and hemorrhages [ 34 ]. In a conditional knockout model using a cre-lox system to save floxed PPAR- γ -knockout mice from embryonic lethality by preserving PPAR- γ -function in the trophoblast marked lipodystrophy was also observed, together with insulin resistance. Surprisingly, these mice were hypotensive and showed increased endothelium-dependent relaxation in response to acetylcholine [ 35 ]. In the latter study, PPAR- γ -function was deficient in all cell types. Mice in which PPAR- γ -function was selectively knocked out in endothelial cells only using again a cre-lox system were found to be hypertensive when fed a high-fat diet [ 36 ]. Also, in mice with a nonlethal-dominant negative mutation in the PPAR- γ -receptor, endothelium-dependent blood vessel dilatation is impaired, while the endothelium is more sensitive to endothelin-1-induced vasoconstriction [ 37 ]. Superoxide levels in these mice are elevated and treatment with a superoxide scavenger can reverse the impaired endothelial vasodilation, highlighting the pivotal role of increased radical formation [ 37 ]. Cerebral arterioles were hypertrophied with a decrease in luminal diameters, indicative of adverse inward vascular remodeling [ 37 ]. Of note, dominant negative PPAR- γ -mutations were found not to be fully selective, therefore, it cannot be excluded that some of the effects observed in the knockout systems are attributable to PPAR- γ -receptor-independent signaling, in particular through the alternative PPAR-receptors [ 38 ]. 4. Effects of PPAR- γ -Activation on Endothelial Proliferation, Migration, Angiogenesis, and Apoptosis Reports on the direct effects of PPAR- γ -agonist treatment on proliferation, migration, and angiogenic network formation of cultured endothelial cells showed variable results. Fukunaga et al. demonstrated increased proliferation with both troglitazone and pioglitazone in four different endothelial cell lines [ 39 ]. In contrast, two other studies found troglitazone to inhibit proliferation of macrovascular endothelial cells [ 40 ] and human umbilical vein endothelial cells [ 41 ]. Rosiglitazone was shown also to inhibit proliferation in one study [ 42 ], while another study found no effect at all [ 40 ]. VEGF-induced endothelial migration was found to be inhibited by PPAR- γ -agonists troglitazone and ciglitazone, which was mediated by inhibition of Akt [ 43 ]. This is in line with the reduced migration and inhibition of angiogenic network formation by PPAR- γ -agonism observed by Xin et al [ 44 ]. However, in the study by Fukunaga et al., troglitazone stimulated both endothelial cell migration and proliferation, resulting in accelerated coverage of a disrupted endothelial monolayer in a wound healing assay [ 39 ]. Also, Biscetti et al. found stimulation of endothelial network formation with PPAR- γ activation using the GW1929 compound, mainly through a VEGF-dependent mechanism [ 45 ]. In vitro effects of PPAR- γ -agonism on endothelial cell apoptosis have been similarly variable. Both spontaneous and TNF-alpha-induced endothelial apoptosis were shown to be inhibited by various thiazolidinedione PPAR- γ -agonists [ 46 ] and troglitazone was found to markedly reduce apoptosis in serum-starved endothelial cells [ 47 ], while another study observed induction of apoptosis using both ciglitazone and PPAR- γ -receptor overexpression [ 48 ]. In several studies, the endogenous PPAR- γ ligand 15-deoxy-delta12,14-prostaglandin J2 (15d-PGJ2)-induced endothelial cell apoptosis [ 46 , 48 , 49 ], but it is important to note that in vascular endothelial cells, the effects of 15d-PGJ2 may be independent of PPAR- γ -receptor activation [ 49 , 50 ]. Taken together, it is clear that PPAR- γ -agonists may modulate endothelial proliferation, migration, angiogenesis, and apoptosis in vitro, but that this may result in both stimulation and inhibition. The factors that determine whether treatment with a PPAR- γ -agonist results in a stimulatory or inhibitory effect on endothelial cells remain largely unidentified. This may involve PPAR- γ -receptor-independent effects as reported for 15d-PGJ2 [ 49 , 50 ] and also with the various TZDs [ 45 ]. Fukunaga et al. posed that a concentration dependency may explain some of the observed discrepancies, as they observed stimulation of DNA-synthesis at low PPAR- γ -agonist dosages and inhibition at higher dosages [ 39 ]. However, this cannot explain all of the divergent findings. In vivo studies in diabetic animals with impaired angiogenesis in response to peripheral ischemia showed that treatment with PPAR- γ -agonist pioglitazone augmented the angiogenic response and increased blood flow recovery [ 51 ]. In rats with experimental focal cerebral ischemia, PPAR- γ -agonist treatment stimulated local angiogenesis and improved functional neurological recovery [ 52 ]. In contrast, PPAR- γ -agonist treatment inhibited pathological choroidial and retinal neovascularization [ 53 ] and suppressed tumor growth and metastasis by inhibiting tumor angiogenesis in several primary tumors, in part through decreasing VEGF-production by tumor cells and blocking the production of angiogenic ELR+CXC-chemokines, mediated through antagonizing NF-kappaB activation [ 54 , 55 ]. These findings suggest that PPAR- γ -agonism may differentially affect neovascularization with inhibition of pathological neovascularization and augmentation of physiological neovascularization. This is in line with the in vitro observations of both stimulatory and inhibitory actions. 5. Effects of PPAR- γ -Activation on Endothelial Dysfunction Endothelial integrity does not only rely on the number of cells, but also on their function. NO-production is a key component of endothelial function. NO stimulates vasodilation, inhibits inflammation, prevents platelet activation, and scavenges radicals [ 56 ]. In vitro studies on the effect of PPAR- γ -activation have consistently shown a stimulating effect on NO production by endothelial cells [ 57 – 60 ]. The observation that siRNA against PPAR- γ blocked the increase in NO-production confirmed that this effect is PPAR- γ -receptor mediated [ 59 ]. In both experimental animal studies and in humans, PPAR- γ -activation has been shown to augment systemic NO-production and endothelial function. In diabetic rats, PPAR- γ -agonist treatment restored impaired endothelium-dependent arterial relaxation by increasing NO-production while reducing oxidative stress [ 61 ]. In healthy human subjects, a single dose of troglitazone increased NO-dependent endothelial function measured by venous occlusion plethysmography of the forearm and nitrite levels [ 62 ]. In nondiabetic patients with hypertension or hypercholesterolemia, endothelial function was improved with pioglitazone [ 63 ]. In type 2 diabetic patients, vascular resistance was shown to be reduced with troglitazone treatment [ 64 , 65 ] and in type 2 diabetic patients with angina pectoris, troglitazone reduced the frequency of angina pectoris with improving endothelial function [ 66 ]. Rosiglitazone attenuated the detrimental effects of the presence of diabetes on NO-production measured directly using an intravital probe and by assessing blood flow in the peripheral skin [ 67 ]. Long-term treatment with pioglitazone resulted in a reduced pulse wave velocity, indicative of reduced vascular stiffness [ 68 ]. In renal transplant recipients, PPAR- γ -agonist treatment enhanced endothelial function [ 69 ]. Importantly, the observed beneficial effects on systemic endothelial function were found to extend to improving intrarenal NO-production. In human type 2 diabetic patients, rosiglitazone treatment increased intrarenal NO levels, which was associated with improvement of renal hemodynamics and reduction of proteinuria [ 70 ]. This is in line with animal studies. In obese Zucker rats, PPAR- γ -agonist treatment lowered blood pressure and ameliorated abnormal pressure natriuresis in association with increased renal NO-metabolite nitrite/nitrate production [ 71 ]. In obese hypertensive Sprague-Dawley rats, PPAR- γ -agonist treatment also reduced blood pressure, increased NO-metabolite nitrite/nitrate excretion, and reduced excretion of oxidative-stress associated urinary isoprostanes and lipid peroxides [ 72 ]. In the kidneys of these rats, eNOS expression was increased while the pathological increase in p47phox and gp91phox associated with obesity was attenuated [ 72 ]. Differences have been observed between the various PPAR- γ -activating compounds in the signaling level at which NO-production is stimulated. The NO producing enzyme endothelial nitric oxide synthase (eNOS) is not only regulated at the level of transcription and translation, but also has multiple phosphorylation sites for activation and deactivation of the enzyme and requires translocation to the caveolae to associate with its cofactors and effectively produce NO [ 73 , 74 ]. Troglitazone has been shown to enhance NO-production by increasing eNOS transcription and eNOS protein translation in isolated endothelial cells [ 58 ], while no effect was found at the transcriptional level for 15d-PGJ2 [ 57 , 58 ], pioglitazone [ 58 ], and ciglitazone [ 57 ]. 15d-PGJ2 and rosiglitazone, but not ciglitazone, were found to stimulate eNOS-phophorylation at activation site ser-1177, thought to be mediated through increasing heat shock protein (hsp)-90 association with eNOS [ 59 ]. Interestingly, there is a cross-talk between NO and PPAR- γ pathways as NO has been shown to rapidly and dose-dependently increase PPAR- γ -binding, mediated by p38 MAPK activation [ 60 ]. In vivo, the stimulatory effect of PPAR- γ -agonists rosiglitazone and pioglitazone on endothelial NO-production was associated with increased eNOS-phosphorylation, while eNOS mRNA and total protein levels were not affected [ 51 , 75 ]. Rosiglitazone treatment has been shown to enhance NO-production through enhancement of cellular transport of arginine, the substrate for NO [ 76 ]. This is particularly relevant for renal disease, as arginine transport was found to be markedly impaired in uremic conditions [ 77 – 79 ]. PPAR- γ -agonism favorably affects other endothelium-derived factors that act in conjunction with NO to maintain endothelial homeostasis. The release of C-type natriuretic peptide, another vasodilatory peptide, by endothelial cells is increased by PPAR- γ -agonist treatment [ 39 ]. In addition, PPAR- γ -agonist treatment increased Cu 2+ and Zn 2+ -superoxide dismutase expression in cultured endothelial cells, thereby increasing their potential for oxygen radical scavenging [ 80 , 81 ]. Also, PPAR- γ -agonist treatment decreases the production of radical oxygen species in endothelial cells [ 81 , 82 ], in part through decreasing the expression of subunits of NADPH-oxidase [ 80 , 81 ]. 6. Antihypertensive Effects of PPAR- γ -Agonism Mediated by the Endothelium Improving NO-availability is thought to be a major mechanism mediating the blood pressure lowering effect of PPAR- γ -agonist treatment. Pioglitazone treatment prevented hypertension and renal oxidative stress both by reducing free-radical production and by increasing nitric oxide production [ 72 ]. No blood pressure reduction was seen with PPAR- γ -agonist treatment in rats also receiving NO-inhibitor L-NAME [ 83 ]. However, other mechanisms may play a role in the anti-hypertensive effect of PPAR- γ -agonist treatment, such as effects on the contractility and proliferation of smooth muscle cells [ 84 ]. A naturally occurring antagonist of NO is the vasoconstrictor endothelin-1, which is involved in atherosclerosis and hypertension [ 85 ]. PPAR- γ -activation inhibits the production of endothelin-1 from endothelial cells in vitro [ 39 , 86 – 88 ]. In type 2 diabetic patients, pioglitazone treatment reduced urinary endothelin-1 secretion, along with decreasing microalbuminuria [ 89 ]. Interestingly, in this study, serum endothelin-1 levels were not affected, suggesting a specific pathogenic role for endothelin-1 secretion in the kidney [ 89 ]. In DOCA-salt hypertensive rats, TZD treatment reduced endothelin-1 production and blunted radical oxygen species production with diminished hypertension progression and vascular remodeling [ 90 ]. Several studies show that the inhibitory effect of PPAR- γ -activation on endothelin-1 secretion takes place at the transcriptional level [ 86 , 87 ] and it was found to be NO-dependent [ 87 ]. Delerive et al. demonstrated that PPAR- γ -activation negatively interferes with the activator protein-1 signaling cascade, resulting in inhibition of thrombin-induced transcription of endothelin-1 [ 88 ]. In hypertension, increased production and secondary effects of angiotensin-II play a major role. In endothelial cells, angiotensin-II is a strong inducer of NADPH-oxidase, resulting in the production of radical oxygen species [ 91 ]. Interestingly, there is a cross-talk between the Angiotensin-II and PPAR- γ signaling pathways. Infusion of angiotensin-II downregulates PPAR- γ -receptor expression in the vascular wall [ 92 ] and treatment with PPAR- γ -agonists abrogates many of the angiotensin-II pathophysiological effects [ 93 ]. Of note, angiotensin-II-receptor-1 antagonists have been shown to act as partial PPAR- γ -agonists [ 94 ]. 7. Effects of PPAR- γ -Activation on Inflammatory Cell Recruitment to the Endothelium Endothelial cells form an important barrier between the blood and peripheral tissues and regulate homing, adhesion, and transmigration of inflammatory cells. Upon activation, endothelial cells may further release inflammatory cytokines and express adhesion molecules to attract inflammatory cells. PPAR- γ -agonist treatment inhibits the increased expression of adhesion molecules such as VCAM-1, ICAM-1, and E-selectin and release of inflammatory cytokines upon stimulation of endothelial cells with PMA [ 95 , 96 ], TNF- 𝛼 [ 95 , 97 ], IFN- γ [ 98 ], IL-1 β [ 99 ], LPS [ 96 ], microparticles [ 100 ], and high glucose [ 101 ]. In vitro studies confirmed that this resulted in decreased adhesion of inflammatory cells to the endothelium [ 96 , 97 , 101 , 102 ]. In addition, a recent study showed that in activated primary human brain endothelial cells, PPAR- γ -activation resulted in a marked reduction of monocyte adhesion and in vitro transendothelial migration, mediated by inhibition of Rac1 and RhoA GTPases [ 102 ]. In vivo, troglitazone and 15d-PGJ2 reduced ICAM-1 and VCAM-1 expression on endothelial cells and reduced inflammatory cell homing to atherosclerotic plaques in a mouse model [ 103 ]. 8. Effects of PPAR- γ -Activation on Endothelial Progenitor Cells A recently uncovered effect of PPAR- γ -agonists is the capacity to augment the level and function of endothelial progenitor cells. PPAR- γ -agonist treatment increases endothelial progenitor cell levels in mice [ 104 , 105 ] and humans [ 104 , 106 , 107 ]. A stimulatory effect on endothelial progenitor cell outgrowth was also observed when cultured peripheral blood mononuclear cells were exposed to PPAR- γ -agonist treatment ex vivo [ 104 , 105 , 108 , 109 ], suggesting an effect on endothelial progenitor cell survival, adhesion, or differentiation. PPAR- γ -agonist treatment largely prevented apoptosis of endothelial progenitor cells induced by CRP [ 109 ] and H 2 O 2 [ 105 ]. Based on marker expression patterns, the differentiation of endothelial progenitor cells toward the endothelial lineage appears to be stimulated [ 104 , 108 ], while expression of smooth muscle cell markers is inhibited [ 104 ]. Pioglitazone inhibited detrimental effects of angiotensin-II on endothelial progenitor cells, including inhibiting the induction of cellular senescence of endothelial progenitor cells via downregulation of the expression of angiotensin-II-receptor-1 and limiting the angiotensin-II-induced increased generation of peroxynitrate and superoxide by the NADPH-oxidase subunit gp91phox [ 110 ]. Functionally, PPAR- γ -agonist treatment was shown to stimulate angiogenic network formation by endothelial progenitor cells in vitro [ 109 ] and in vivo [ 105 ]. The adhesion capacity may be increased upon TZD-treatment [ 108 , 109 ]. In addition, endothelial progenitor cell-mediated reendothelialization of a denuded segment of the femoral artery in mice was accelerated in PPAR- γ -agonist treated animals [ 104 ]. As endothelial progenitor cells may also participate in regeneration of damaged renal endothelium [ 26 ], we investigated a potential role for enhanced endothelial progenitor cell homing and glomerular incorporation by PPAR- γ -agonist treatment [ 7 ]. However, using a rat allogenic bone marrow transplantation model, we could not detect an effect on the number of incorporated circulation-derived glomerular endothelial cells in the recovering glomerulus with rosiglitazone treatment, although rosiglitazone did attenuate the clinical course of glomerulonephritis. 9. Conclusions and Perspectives Activation of the PPAR- γ -receptors present on renal endothelial cells affects their cellular behavior. Proliferation, apoptosis, migration, and angiogenesis by endothelial cells are modulated, but may involve both stimulation and inhibition depending on the specific circumstances. It remains unclear how this bimodal potential of PPAR- γ -agonists is regulated. An important consequence of PPAR- γ -receptor activation is the enhanced production of nitric oxide and C-type natriuretic peptide and superoxide dismutase, while endothelin-1 production is inhibited. Together, this improves the capacity of the endothelium to exert vasodilatory, anti-inflammatory, and antioxidative actions, resulting in blood pressure lowering and direct renoprotective effects. In addition, the presentation of adhesion molecules and release of cytokines aimed at recruiting inflammatory cells to activated endothelium is inhibited by PPAR- γ -agonism. This may also in part account for the anti-inflammatory effects of PPAR- γ -agonists, supplementary to direct effects on the inflammatory cells themselves. Finally, PPAR- γ -receptors are also found on endothelial progenitor cells and PPAR- γ -agonist stimulate progenitor-mediated endothelial repair, although definitive evidence that this occurs in the kidney is currently lacking. Activation of other PPAR-receptors besides PPAR- γ may also have beneficial effects on the endothelium. Recently, selective agonists for PPAR- β / δ were developed [ 111 ]. PPAR- β / δ receptors are present on endothelium and stimulation with a pharmacological PPAR- β / δ -agonist was found to increase endothelial cell proliferation and angiogenesis in vitro and in vivo, mainly mediated through VEGF [ 112 ]. Like PPAR- γ -agonists, PPAR- β / δ -agonists increase circulating endothelial progenitor cell levels, augment endothelial progenitor cell function in vitro, and improve endothelial progenitor cell-mediated neovascularization of ischemic tissue [ 113 ]. Also, much like PPAR- γ -agonists, PPAR- β / δ -agonist treatment reduced the expression of adhesion molecules and monocyte binding to activated endothelial cells [ 114 ]. Currently, PPAR- γ -agonist treatment is not standard clinical practice in nondiabetic renal patients. As shown in this review, experimental studies indicate that stimulatory effects of PPAR- γ -agonism on endothelium may provide additional benefit in nondiabetic renal disease. Together with other potentially therapeutic effects independent of the insulin-sensitizing action such as the anti-inflammatory actions, this provides a rationale for further clinical evaluation in nondiabetic renal patients. For patients with diabetic kidney disease, a pressing question is whether glycemic control with a TZD is superior to other antidiabetic drugs for preventing the decline of renal function. To date, only retrospective studies, post hoc analyses, and pilot studies are available to help answer this question. Trials designed to specifically evaluate this question have yet to be performed. Acknowledgments P. E. Westerweel is supported by ZonMw AGIKO Grant no. 2007/12579. M. C. Verhaar is supported by NWO Vidi Grant no. 016.096.359. <h4>References</h4> Y. Guan and M. D. Breyer, “ Peroxisome proliferator-activated receptors (PPARs): novel therapeutic targets in renal disease ,” Kidney International , vol. 60, no. 1, pp. 14–30, 2001. P. A. Sarafidis and G. L. Bakris, “ Protection of the kidney by thiazolidinediones: an assessment from bench to bedside ,” Kidney International , vol. 70, no. 7, pp. 1223–1233, 2006. K. Haraguchi, H. Shimura, and T. Onaya, “ Suppression of experimental crescentic glomerulonephritis by peroxisome proliferator-activated receptor (PPAR) γ activators ,” Clinical and Experimental Nephrology , vol. 7, no. 1, pp. 27–32, 2003. A. Benigni, C. Zoja, S. Tomasoni, et al., “ Transcriptional regulation of nephrin gene by peroxisome proliferator-activated receptor- γ agonist: molecular mechanism of the antiproteinuric effect of pioglitazone ,” Journal of the American Society of Nephrology , vol. 17, no. 6, pp. 1624–1632, 2006. L.-J. Ma, C. Marcantoni, M. F. Linton, S. Fazio, and A. B. Fogo, “ Peroxisome proliferator-activated receptor- γ agonist troglitazone protects against nondiabetic glomerulosclerosis in rats ,” Kidney International , vol. 59, no. 5, pp. 1899–1910, 2001. A. Sivarajah, P. K. Chatterjee, N. S. A. Patel, et al., “ Agonists of peroxisome-proliferator activated receptor-gamma reduce renal ischemia/reperfusion injury ,” The American Journal of Nephrology , vol. 23, no. 4, pp. 267–276, 2003. P. E. Westerweel, K. den Ouden, T. Q. Nguyen, R. Goldschmeding, J. A. Joles, and M. C. Verhaar, “ Amelioration of anti-Thy1-glomerulonephritis by PPAR- γ agonism without increase of endothelial progenitor cell homing ,” The American Journal of Physiology , vol. 294, no. 2, pp. F379–F384, 2008. M. Matsuyama, R. Yoshimura, T. Hase, et al., “ Expression of peroxisome proliferator-activated receptor- γ in renal ischemia-reperfusion injury ,” Transplantation Proceedings , vol. 37, no. 4, pp. 1684–1685, 2005. R. Yoshimura, M. Matsuyama, Y. Segawa, et al., “ Study of peroxisome proliferator-activated receptor (PPAR)- γ in renal ischemia-reperfusion injury ,” Transplantation Proceedings , vol. 36, no. 7, pp. 1946–1948, 2004. Y. Guan, Y. Zhang, A. Schneider, L. Davis, R. M. Breyer, and M. D. Breyer, “Peroxisome proliferator-activated receptor- γ activity is associated with renal microvasculature,” The American Journal of Physiology , vol. 281, no. 6, pp. F1036–F1046, 2001. N. Yamanaka and A. Shimizu, “ Role of glomerular endothelial damage in progressive renal disease ,” Kidney and Blood Pressure Research , vol. 22, no. 1-2, pp. 13–20, 1999. M. E. J. Reinders, T. J. Rabelink, and D. M. Briscoe, “ Angiogenesis and endothelial cell repair in renal disease and allograft rejection ,” Journal of the American Society of Nephrology , vol. 17, no. 4, pp. 932–942, 2006. D. A. Long, W. Mu, K. L. Price, and R. J. Johnson, “ Blood vessels and the aging kidney ,” Nephron Experimental Nephrology , vol. 101, no. 3, pp. e95–e99, 2005. J. P. Garg and G. L. Bakris, “ Microalbuminuria: marker of vascular dysfunction, risk factor for cardiovascular disease ,” Vascular Medicine , vol. 7, no. 1, pp. 35–43, 2002. G. L. Bakris, “ Clinical importance of microalbuminuria in diabetes and hypertension ,” Current Hypertension Reports , vol. 6, no. 5, pp. 352–356, 2004. S. Gschwend, H. Buikema, G. Navis, R. H. Henning, D. de Zeeuw, and R. P. E. van Dokkum, “ Endothelial dilatory function predicts individual susceptibility to renal damage in the 5/6 nephrectomized rat ,” Journal of the American Society of Nephrology , vol. 13, no. 12, pp. 2909–2915, 2002. A. Shimizu, H. Kitamura, Y. Masuda, M. Ishizaki, Y. Sugisaki, and N. Yamanaka, “Rare glomerular capillary regeneration and subsequent capillary regression with endothelial cell apoptosis in progressive glomerulonephritis,” The American Journal of Pathology , vol. 151, no. 5, pp. 1231–1239, 1997. L. Iruela-Arispe, K. Gordon, C. Hugo, et al., “Participation of glomerular endothelial cells in the capillary repair of glomerulonephritis,” The American Journal of Pathology , vol. 147, no. 6, pp. 1715–1727, 1995. Y. Masuda, A. Shimizu, T. Mori, et al., “Vascular endothelial growth factor enhances glomerular capillary repair and accelerates resolution of experimentally induced glomerulonephritis,” The American Journal of Pathology , vol. 159, no. 2, pp. 599–608, 2001. K. Miyamoto, Y. Kitamoto, H. Tokunaga, et al., “ Protective effect of vascular endothelial growth factor/vascular permeability factor 165 and 121 on glomerular endothelial cell injury in the rat ,” Laboratory Investigation , vol. 84, no. 9, pp. 1126–1136, 2004. T. Ostendorf, U. Kunter, F. Eitner, et al., “ VEGf 165 mediates glomerular endothelial repair ,” The Journal of Clinical Investigation , vol. 104, no. 7, pp. 913–923, 1999. D.-H. Kang, J. Hughes, M. Mazzali, G. F. Schreiner, and R. J. Johnson, “Impaired angiogenesis in the remnant kidney model—part II: vascular endothelial growth factor administration reduces renal fibrosis and stabilizes renal function,” Journal of the American Society of Nephrology , vol. 12, no. 7, pp. 1448–1457, 2001. D. H. Kang, A. H. Joly, S. W. Oh, et al., “Impaired angiogenesis in the remnant kidney model—part I: potential role of vascular endothelial growth factor and thrombospondin-1,” Journal of the American Society of Nephrology , vol. 12, no. 7, pp. 1434–1447, 2001. M. B. Rookmaaker, H. Tolboom, R. Goldschmeding, J.-J. Zwaginga, T. J. Rabelink, and M. C. Verhaar, “ Bone marrow-derived cells contribute to endothelial repair after thrombotic microangiopathy ,” Blood , vol. 99, no. 3, p. 1095, 2002. M. B. Rookmaaker, A. M. Smits, H. Tolboom, et al., “Bone-marrow-derived cells contribute to glomerular endothelial repair in experimental glomerulonephritis,” The American Journal of Pathology , vol. 163, no. 2, pp. 553–562, 2003. M. B. Rookmaaker, M. C. Verhaar, A. J. Van Zonneveld, and T. J. Rabelink, “ Progenitor cells in the kidney: biology and therapeutic perspectives ,” Kidney International , vol. 66, no. 2, pp. 518–522, 2004. E. L. Lagaaij, G. F. Cramer-Knijnenburg, F. J. van Kemenade, L. A. van Es, J. A. Bruijn, and J. H. J. M. van Krieken, “ Endothelial cell chimerism after renal transplantation and vascular rejection ,” The Lancet , vol. 357, no. 9249, pp. 33–37, 2001. H. Peters, U. Daig, S. Martini, et al., “ No mediates antifibrotic actions of L-arginine supplementation following induction of anti-thy1 glomerulonephritis ,” Kidney International , vol. 64, no. 2, pp. 509–518, 2003. B. Li, T. Morioka, M. Uchiyama, and T. Oite, “ Bone marrow cell infusion ameliorates progressive glomerulosclerosis in an experimental rat model ,” Kidney International , vol. 69, no. 2, pp. 323–330, 2006. H. Uchimura, T. Marumo, O. Takase, et al., “ Intrarenal injection of bone marrow-derived angiogenic cells reduces endothelial injury and mesangial cell activation in experimental glomerulonephritis ,” Journal of the American Society of Nephrology , vol. 16, no. 4, pp. 997–1004, 2005. I. Barroso, M. Gurnell, V. E. F. Crowley, et al., “ Dominant negative mutations in human PPAR γ associated with severe insulin resistance, diabetes mellitus and hypertension ,” Nature , vol. 402, no. 6764, pp. 880–883, 1999. P. D. G. Miles, Y. Barak, W. He, R. M. Evans, and J. M. Olefsky, “ Improved insulin-sensitivity in mice heterozygous for PPAR- γ deficiency ,” The Journal of Clinical Investigation , vol. 105, no. 3, pp. 287–292, 2000. T. Yamauchi, J. Kamon, H. Waki, et al., “ The mechanisms by which both heterozygous peroxisome proliferator-activated receptor γ (PPAR γ ) deficiency and PPAR γ agonist improve insulin resistance ,” The Journal of Biological Chemistry , vol. 276, no. 44, pp. 41245–41254, 2001. Y. Barak, M. C. Nelson, E. S. Ong, et al., “ PPAR γ is required for placental, cardiac, and adipose tissue development ,” Molecular Cell , vol. 4, no. 4, pp. 585–595, 1999. S. Z. Duan, C. Y. Ivashchenko, S. E. Whitesall, et al., “ Hypotension, lipodystrophy, and insulin resistance in generalized PPAR γ -deficient mice rescued from embryonic lethality ,” The Journal of Clinical Investigation , vol. 117, no. 3, pp. 812–822, 2007. C. J. Nicol, M. Adachi, T. E. Akiyama, and F. J. Gonzalez, “ PPAR γ in endothelial cells influences high fat diet-induced hypertension ,” The American Journal of Hypertension , vol. 18, no. 4, pp. 549–556, 2005. A. M. Beyer, G. L. Baumbach, C. M. Halabi, et al., “ Interference with PPAR γ signaling causes cerebral vascular dysfunction, hypertrophy, and remodeling ,” Hypertension , vol. 51, no. 4, pp. 867–871, 2008. R. K. Semple, A. Meirhaeghe, A. J. Vidal-Puig, et al., “ A dominant negative human peroxisome proliferator-activated receptor (PPAR) α is a constitutive transcriptional corepressor and inhibits signaling through all PPAR isoforms ,” Endocrinology , vol. 146, no. 4, pp. 1871–1882, 2005. Y. Fukunaga, H. Itoh, K. Doi, et al., “ Thiazolidinediones, peroxisome proliferator-activated receptor γ agonists, regulate endothelial cell growth and secretion of vasoactive peptides ,” Atherosclerosis , vol. 158, no. 1, pp. 113–119, 2001. S. T. de Dios, K. M. Hannan, R. J. Dilley, M. A. Hill, and P. J. Little, “ Troglitazone, but not rosiglitazone, inhibits Na/H exchange activity and proliferation of macrovascular endothelial cells ,” Journal of Diabetes and Its Complications , vol. 15, no. 3, pp. 120–127, 2001. H. K. Hong, Y. M. Cho, K.-H. Park, C.-T. Lee, H. K. Lee, and K. S. Park, “ Peroxisome proliferator-activated receptor gamma mediated inhibition of plasminogen activator inhibitor type 1 production and proliferation of human umbilical vein endothelial cells ,” Diabetes Research and Clinical Practice , vol. 62, no. 1, pp. 1–8, 2003. W. H.-H. Sheu, H.-C. Ou, F.-P. Chou, T.-M. Lin, and C.-H. Yang, “ Rosiglitazone inhibits endothelial proliferation and angiogenesis ,” Life Sciences , vol. 78, no. 13, pp. 1520–1528, 2006. S. Goetze, F. Eilers, A. Bungenstock, et al., “ PPAR activators inhibit endothelial cell migration by targeting Akt ,” Biochemical and Biophysical Research Communications , vol. 293, no. 5, pp. 1431–1437, 2002. X. Xin, S. Yang, J. Kowalski, and M. E. Gerritsen, “ Peroxisome proliferator-activated receptor γ ligands are potent inhibitors of angiogenesis in vitro and in vivo ,” The Journal of Biological Chemistry , vol. 274, no. 13, pp. 9116–9121, 1999. F. Biscetti, E. Gaetani, A. Flex, et al., “ Selective activation of peroxisome proliferator-activated receptor (PPAR) α and PPAR γ induces neoangiogenesis through a vascular endothelial growth factor-dependent mechanism ,” Diabetes , vol. 57, no. 5, pp. 1394–1404, 2008. M. Artwohl, T. Hölzebein, C. Fürnsinn, et al., “ Thiazolidinediones inhibit apoptosis and heat shock protein 60 expression in human vascular endothelial cells ,” Thrombosis and Haemostasis , vol. 93, no. 5, pp. 810–815, 2005. K. M. Hannan, R. J. Dilley, S. T. de Dios, and P. J. Little, “ Troglitazone stimulates repair of the endothelium and inhibits neointimal formation in denuded rat aorta ,” Arteriosclerosis, Thrombosis, and Vascular Biology , vol. 23, no. 5, pp. 762–768, 2003. D. Bishop-Bailey and T. Hla, “ Endothelial cell apoptosis induced by the peroxisome proliferator-activated receptor (PPAR) ligand 15-deoxy- Δ 12 , 14 -prostaglandin J 2 ,” The Journal of Biological Chemistry , vol. 274, no. 24, pp. 17042–17048, 1999. A.-L. Levonen, D. A. Dickinson, D. R. Moellering, R. T. Mulcahy, H. J. Forman, and V. M. Darley-Usmar, “ Biphasic effects of 15-deoxy- Δ 12 , 14 -prostaglandin J 2 on glutathione induction and apoptosis in human endothelial cells ,” Arteriosclerosis, Thrombosis, and Vascular Biology , vol. 21, no. 11, pp. 1846–1851, 2001. H. Migita and J. Morser, “ 15-deoxy- Δ 12 , 14 -postaglandin J 2 (15d-PGJ2) signals through retinoic acid receptor-related orphan receptor- α but not peroxisome proliferator-activated receptor- γ in human vascular endothelial cells: the effect of 15d-PGJ2 on tumor necrosis factor- α -induced gene expression ,” Arteriosclerosis, Thrombosis, and Vascular Biology , vol. 25, no. 4, pp. 710–716, 2005. P.-H. Huang, M. Sata, H. Nishimatsu, M. Sumi, Y. Hirata, and R. Nagai, “ Pioglitazone ameliorates endothelial dysfunction and restores ischemia-induced angiogenesis in diabetic mice ,” Biomedicine and Pharmacotherapy , vol. 62, no. 1, pp. 46–52, 2008. K. Chu, S.-T. Lee, J.-S. Koo, et al., “ Peroxisome proliferator-activated receptor- γ -agonist, rosiglitazone, promotes angiogenesis after focal cerebral ischemia ,” Brain Research , vol. 1093, no. 1, pp. 208–218, 2006. T. Murata, S. He, M. Hangai, et al., “Peroxisome proliferator-activated receptor- γ ligands inhibit choroidal neovascularization,” Investigative Ophthalmology and Visual Science , vol. 41, no. 8, pp. 2309–2317, 2000. D. Panigrahy, S. Singer, L. Q. Shen, et al., “ PPAR γ ligands inhibit primary tumor growth and metastasis by inhibiting angiogenesis ,” The Journal of Clinical Investigation , vol. 110, no. 7, pp. 923–932, 2002. V. G. Keshamouni, D. A. Arenberg, R. C. Reddy, M. J. Newstead, S. Anthwal, and T. J. Standiford, “ PPAR- γ activation inhibits angiogenesis by blocking ELR+CXC chemokine production in non-small cell lung cancer ,” Neoplasia , vol. 7, no. 3, pp. 294–301, 2005. M. C. Verhaar and T. J. Rabelink, “ Endothelial function: strategies for early intervention ,” Cardiovascular Drugs and Therapy , vol. 12, pp. 125–134, 1998. D. S. Calnek, L. Mazzella, S. Roser, J. Roman, and C. M. Hart, “ Peroxisome proliferator-activated receptor γ ligands increase release of nitric oxide from endothelial cells ,” Arteriosclerosis, Thrombosis, and Vascular Biology , vol. 23, no. 1, pp. 52–57, 2003. K. Goya, S. Sumitani, M. Otsuki, et al., “ The thiazolidinedione drug troglitazone up-regulates nitric oxide synthase expression in vascular endothelial cells ,” Journal of Diabetes and Its Complications , vol. 20, no. 5, pp. 336–342, 2006. J. A. Polikandriotis, L. J. Mazzella, H. L. Rupnow, and C. M. Hart, “ Peroxisome proliferator-activated receptor γ ligands stimulate endothelial nitric oxide production through distinct peroxisome proliferator-activated receptor γ -dependent mechanisms ,” Arteriosclerosis, Thrombosis, and Vascular Biology , vol. 25, no. 9, pp. 1810–1816, 2005. A. Ptasinska, S. Wang, J. Zhang, R. A. Wesley, and R. L. Danner, “ Nitric oxide activation of peroxisome proliferator-activated receptor gamma through a p38 MAPK signaling pathway ,” The FASEB Journal , vol. 21, no. 3, pp. 950–961, 2007. J. B. Majithiya, A. N. Paramar, and R. Balaraman, “ Pioglitazone, a PPAR γ agonist, restores endothelial function in aorta of streptozotocin-induced diabetic rats ,” Cardiovascular Research , vol. 66, no. 1, pp. 150–161, 2005. S. Fujishima, Y. Ohya, Y. Nakamura, U. Onaka, I. Abe, and M. Fujishima, “ Troglitazone, an insulin sensitizer, increases forearm blood flow in humans ,” The American Journal of Hypertension , vol. 11, no. 9, pp. 1134–1137, 1998. U. Campia, L. A. Matuskey, and J. A. Panza, “ Peroxisome proliferator-activated receptor- γ activation with pioglitazone improves endothelium-dependent dilation in nondiabetic patients with major cardiovascular risk factors ,” Circulation , vol. 113, no. 6, pp. 867–875, 2006. M. N. Ghazzi, J. E. Perez, T. K. Antonucci, et al., “ Cardiac and glycemic benefits of troglitazone treatment in NIDDM. The Troglitazone Study Group ,” Diabetes , vol. 46, no. 3, pp. 433–439, 1997. B. H. Sung, J. L. Izzo, Jr., P. Dandona, and M. F. Wilson, “Vasodilatory effects of troglitazone improve blood pressure at rest and during mental stress in type 2 diabetes mellitus,” Hypertension , vol. 34, no. 1, pp. 83–88, 1999. T. Murakami, S. Mizuno, K. Ohsato, et al., “ Effects of troglitazone on frequency of coronary vasospastic-induced angina pectoris in patients with diabetes mellitus ,” The American Journal of Cardiology , vol. 84, no. 1, pp. 92–94, 1999. A. I. Vinik, K. B. Stansberry, and P. M. Barlow, “ Rosiglitazone treatment increases nitric oxide production in human peripheral skin: a controlled clinical trial in patients with type 2 diabetes mellitus ,” Journal of Diabetes and Its Complications , vol. 17, no. 5, pp. 279–285, 2003. T. Nakamura, T. Matsuda, Y. Kawagoe, et al., “ Effect of pioglitazone on carotid intima-media thickness and arterial stiffness in type 2 diabetic nephropathy patients ,” Metabolism , vol. 53, no. 10, pp. 1382–1386, 2004. M. H. Voytovich, C. Simonsen, T. Jenssen, J. Hjelmesæth, A. Åsberg, and A. Hartmann, “ Short-term treatment with rosiglitazone improves glucose tolerance, insulin sensitivity and endothelial function in renal transplant recipients ,” Nephrology Dialysis Transplantation , vol. 20, no. 2, pp. 413–418, 2005. F. Pistrosch, K. Herbrig, B. Kindel, J. Passauer, S. Fischer, and P. Gross, “ Rosiglitazone improves glomerular hyperfiltration, renal endothelial dysfunction, and microalbuminuria of incipient diabetic nephropathy in patients ,” Diabetes , vol. 54, no. 7, pp. 2206–2211, 2005. K. Fujiwara, K. Hayashi, H. Matsuda, et al., “Altered pressure-natriuresis in obese Zucker rats,” Hypertension , vol. 33, no. 6, pp. 1470–1475, 1999. A. D. Dobrian, S. D. Schriver, A. A. Khraibi, and R. L. Prewitt, “ Pioglitazone prevents hypertension and reduces oxidative stress in diet-induced obesity ,” Hypertension , vol. 43, no. 1, pp. 48–56, 2004. R. Govers and T. J. Rabelink, “Cellular regulation of endothelial nitric oxide synthase,” The American Journal of Physiology , vol. 280, no. 2, pp. F193–F206, 2001. B. Braam and M. C. Verhaar, “ Understanding eNOS for pharmacological modulation of endothelial function: a translational view ,” Current Pharmaceutical Design , vol. 13, no. 17, pp. 1727–1740, 2007. A. T. Gonon, A. Bulhak, F. Labruto, P.-O. Sjöquist, and J. Pernow, “ Cardioprotection mediated by rosiglitazone, a peroxisome proliferator activated receptor gamma ligand, in relation to nitric oxide ,” Basic Research in Cardiology , vol. 102, no. 1, pp. 80–89, 2007. M. Ingbir, I. F. Schwartz, A. Shtabsky, et al., “ Rosiglitazone improves aortic arginine transport, through inhibition of PKC α , in uremic rats ,” The American Journal of Physiology , vol. 295, no. 2, pp. F471–F477, 2008. I. F. Schwartz, R. Ayalon, T. Chernichovski, et al., “ Arginine uptake is attenuated through modulation of cationic amino-acid transporter-1, in uremic rats ,” Kidney International , vol. 69, no. 2, pp. 298–303, 2006. L. Wagner, J. D. Klein, J. M. Sands, and C. Baylis, “ Urea transporters are distributed in endothelial cells and mediate inhibition of L-arginine transport ,” The American Journal of Physiology , vol. 283, no. 3, pp. F578–F582, 2002. S. Xiao, L. Wagner, J. Mahaney, and C. Baylis, “Uremic levels of urea inhibit L-arginine transport in cultured endothelial cells,” The American Journal of Physiology , vol. 280, no. 6, pp. F989–F995, 2001. I. Inoue, S.-I. Goto, T. Matsunaga, et al., “ The ligands/activators for peroxisome proliferator-activated receptor α (PPAR α ) and PPAR γ increase Cu 2 + , Zn 2 + -superoxide dismutase and decrease p22phox message expressions in primary endothelial cells ,” Metabolism , vol. 50, no. 1, pp. 3–11, 2001. J. Hwang, D. J. Kleinhenz, B. Lassègue, K. K. Griendling, S. Dikalov, and C. M. Hart, “ Peroxisome proliferator-activated receptor- γ ligands regulate endothelial membrane superoxide production ,” The American Journal of Physiology , vol. 288, no. 4, pp. C899–C905, 2005. J. L. Mehta, B. Hu, J. Chen, and D. Li, “ Pioglitazone inhibits LOX-1 expression in human coronary artery endothelial cells by reducing intracellular superoxide radical generation ,” Arteriosclerosis, Thrombosis, and Vascular Biology , vol. 23, no. 12, pp. 2203–2208, 2003. J. B. Majithiya, A. N. Parmar, C. J. Trivedi, and R. Balaraman, “ Effect of pioglitazone on L-NAME induced hypertension in diabetic rats ,” Vascular Pharmacology , vol. 43, no. 4, pp. 260–266, 2005. P. A. Sarafidis and A. N. Lasaridis, “ Actions of peroxisome proliferator-activated receptors- γ agonists explaining a possible blood pressure-lowering effect ,” The American Journal of Hypertension , vol. 19, no. 6, pp. 646–653, 2006. E. L. Schiffrin, “ Role of endothelin-1 in hypertension and vascular disease ,” The American Journal of Hypertension , vol. 14, no. 6, part 2, pp. 83S–89S, 2001. F. Martin-Nizard, C. Furman, P. Delerive, et al., “ Peroxisome proliferator-activated receptor activators inhibit oxidized low-density lipoprotein-induced endothelin-1 secretion in endothelial cells ,” Journal of Cardiovascular Pharmacology , vol. 40, no. 6, pp. 822–831, 2002. H. Satoh, K. Tsukamoto, Y. Hashimoto, et al., “ Thiazolidinediones suppress endothelin-1 secretion from bovine vascular endothelial cells: a new possible role of PPAR γ on vascular endothelial function ,” Biochemical and Biophysical Research Communications , vol. 254, no. 3, pp. 757–763, 1999. P. Delerive, F. Martin-Nizard, G. Chinetti, et al., “Peroxisome proliferator-activated receptor activators inhibit thrombin-induced endothelin-1 production in human vascular endothelial cells by inhibiting the activator protein-1 signaling pathway,” Circulation Research , vol. 85, no. 5, pp. 394–402, 1999. T. Nakamura, C. Ushiyama, N. Shimada, K. Hayashi, I. Ebihara, and H. Koide, “ Comparative effects of pioglitazone, glibenclamide, and voglibose on urinary endothelin-1 and albumin excretion in diabetes patients ,” Journal of Diabetes and Its Complications , vol. 14, no. 5, pp. 250–254, 2000. M. Iglarz, R. M. Touyz, F. Amiri, M.-F. Lavoie, Q. N. Diep, and E. L. Schiffrin, “ Effect of peroxisome proliferator-activated receptor- α and - γ activators on vascular remodeling in endothelin-dependent hypertension ,” Arteriosclerosis, Thrombosis, and Vascular Biology , vol. 23, no. 1, pp. 45–51, 2003. T. Münzel, U. Hink, T. Heitzer, and T. Meinertz, “ Role for NADPH/NADH oxidase in the modulation of vascular tone ,” Annals of the New York Academy of Sciences , vol. 874, pp. 386–400, 1999. D. M. Tham, B. Martin-McNulty, Y.-X. Wang, et al., “Angiotensin II is associated with activation of NF- κ B-mediated genes and downregulation of PPARs,” Physiological Genomics , vol. 11, no. 1, pp. 21–30, 2003. Q. N. Diep, M. El Mabrouk, J. S. Cohn, et al., “ Structure, endothelial function, cell growth, and inflammation in blood vessels of angiotensin II-infused rats: role of peroxisome proliferator-activated receptor- γ ,” Circulation , vol. 105, no. 19, pp. 2296–2302, 2002. U. Kintscher and T. Unger, “ Vascular protection in diabetes: a pharmacological view of angiotensin II type 1 receptor blockers ,” Acta Diabetologica , vol. 42, pp. S26–S32, 2005. N. Wang, L. Verna, N.-G. Chen, et al., “ Constitutive activation of peroxisome proliferator-activated receptor- γ suppresses pro-inflammatory adhesion molecules in human vascular endothelial cells ,” The Journal of Biological Chemistry , vol. 277, no. 37, pp. 34176–34181, 2002. S. M. Jackson, F. Parhami, X.-P. Xi, et al., “Peroxisome proliferator-activated receptor activators target human endothelial cells to inhibit leukocyte-endothelial cell interaction,” Arteriosclerosis, Thrombosis, and Vascular Biology , vol. 19, no. 9, pp. 2094–2104, 1999. Y. Jung, S. Song, and C. Choi, “ Peroxisome proliferator activated receptor γ agonists suppress TNF α -induced ICAM-1 expression by endothelial cells in a manner potentially dependent on inhibition of reactive oxygen species ,” Immunology Letters , vol. 117, no. 1, pp. 63–69, 2008. N. Marx, F. Mach, A. Sauty, et al., “Peroxisome proliferator-activated receptor- γ activators inhibit IFN- γ -induced expression of the T cell-active CXC chemokines IP-10, Mig, and I-TAC in human endothelial cells,” The Journal of Immunology , vol. 164, no. 12, pp. 6503–6508, 2000. E. Imamoto, N. Yoshida, K. Uchiyama, et al., “ Inhibitory effect of pioglitazone on expression of adhesion molecules on neutrophils and endothelial cells ,” BioFactors , vol. 20, no. 1, pp. 37–47, 2004. A. Tesse, G. Al-Massarani, R. Wangensteen, S. Reitenbach, M. C. Martínez, and R. Andriantsitohaina, “ Rosiglitazone, a peroxisome proliferator-activated receptor- γ agonist, prevents microparticle-induced vascular hyporeactivity through the regulation of proinflammatory proteins ,” Journal of Pharmacology and Experimental Therapeutics , vol. 324, no. 2, pp. 539–547, 2008. E. Verrier, L. Wang, C. Wadham, et al., “ PPAR γ agonists ameliorate endothelial cell activation via inhibition of diacylglycerol-protein kinase C signaling pathway: role of diacylglycerol kinase ,” Circulation Research , vol. 94, no. 11, pp. 1515–1522, 2004. S. H. Ramirez, D. Heilman, B. Morsey, R. Potula, J. Haorah, and Y. Persidsky, “Activation of peroxisome proliferator-activated receptor gamma (PPARgamma) suppresses Rho GTPases in human brain microvascular endothelial cells and inhibits adhesion and transendothelial migration of HIV-1 infected monocytes,” The Journal of Immunology , vol. 180, no. 3, pp. 1854–1865, 2008. V. Pasceri, H. D. Wu, J. T. Willerson, and E. T. H. Yeh, “Modulation of vascular inflammation in vitro and in vivo by peroxisome proliferator-activated receptor- γ activators,” Circulation , vol. 101, no. 3, pp. 235–238, 2000. C.-H. Wang, N. Ciliberti, S.-H. Li, et al., “ Rosiglitazone facilitates angiogenic progenitor cell differentiation toward endothelial lineage: a new paradigm in glitazone pleiotropy ,” Circulation , vol. 109, no. 11, pp. 1392–1400, 2004. C. Gensch, Y. P. Clever, C. Werner, M. Hanhoun, M. Böhm, and U. Laufs, “ The PPAR- γ agonist pioglitazone increases neoangiogenesis and prevents apoptosis of endothelial progenitor cells ,” Atherosclerosis , vol. 192, no. 1, pp. 67–74, 2007. F. Pistrosch, K. Herbrig, U. Oelschlaegel, et al., “ PPAR γ -agonist rosiglitazone increases number and migratory activity of cultured endothelial progenitor cells ,” Atherosclerosis , vol. 183, no. 1, pp. 163–167, 2005. C. Werner, C. H. Kamani, C. Gensch, M. Böhm, and U. Laufs, “ The peroxisome proliferator-activated receptor- γ agonist pioglitazone increases number and function of endothelial progenitor cells in patients with coronary artery disease and normal glucose tolerance ,” Diabetes , vol. 56, no. 10, pp. 2609–2615, 2007. S. Redondo, M. Hristov, D. Gümbel, T. Tejerina, and C. Weber, “ Biphasic effect of pioglitazone on isolated human endothelial progenitor cells: involvement of peroxisome proliferator-activated receptor- γ and transforming growth factor- β 1 ,” Thrombosis and Haemostasis , vol. 97, no. 6, pp. 979–987, 2007. S. Verma, M. A. Kuliszewski, S.-H. Li, et al., “ C-reactive protein attenuates endothelial progenitor cell survival, differentiation, and function: further evidence of a mechanistic link between C-reactive protein and cardiovascular disease ,” Circulation , vol. 109, no. 17, pp. 2058–2067, 2004. T. Imanishi, K. Kobayashi, A. Kuroi, H. Ikejima, and T. Akasaka, “ Pioglitazone inhibits angiotensin II-induced senescene of endothelial progenitor cell ,” Hypertension Research , vol. 31, no. 4, pp. 757–765, 2008. D. Bishop-Bailey, “ Peroxisome proliferator-activated receptor β / δ goes vascular ,” Circulation Research , vol. 102, no. 2, pp. 146–147, 2008. L. Piqueras, A. R. Reynolds, K. M. Hodivala-Dilke, et al., “ Activation of PPAR β / δ induces endothelial cell proliferation and angiogenesis ,” Arteriosclerosis, Thrombosis, and Vascular Biology , vol. 27, no. 1, pp. 63–69, 2007. J. K. Han, H. S. Lee, H. M. Yang, et al., “ Peroxisome proliferator-activated receptor-delta agonist enhances vasculogenesis by regulating endothelial progenitor cells through genomic and nongenomic activations of the phosphatidylinositol 3-kinase/Akt pathway ,” Circulation , vol. 118, no. 10, pp. 1021–1033, 2008. Y. Rival, N. Benéteau, T. Taillandier, et al., “ PPAR α and PPAR δ activators inhibit cytokine-induced nuclear translocation of NF- κ B and expression of VCAM-1 in EAhy926 endothelial cells ,” European Journal of Pharmacology , vol. 435, no. 2-3, pp. 143–151, 2002. //

Journal

PPAR ResearchHindawi Publishing Corporation

Published: Feb 26, 2009

There are no references for this article.