Next Article in Journal
Methyl Donors, Epigenetic Alterations, and Brain Health: Understanding the Connection
Next Article in Special Issue
Cardiac Differentiation Promotes Focal Adhesions Assembly through Vinculin Recruitment
Previous Article in Journal
CARs and Drugs: Pharmacological Ways of Boosting CAR-T-Cell Therapy
Previous Article in Special Issue
Monomeric C-Reactive Protein in Atherosclerotic Cardiovascular Disease: Advances and Perspectives
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Pharmacological Utility of PPAR Modulation for Angiogenesis in Cardiovascular Disease

by
Nicole Wagner
* and
Kay-Dietrich Wagner
*
CNRS, INSERM, Université Côte d’Azur, iBV, 06107 Nice, France
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(3), 2345; https://doi.org/10.3390/ijms24032345
Submission received: 13 December 2022 / Revised: 22 January 2023 / Accepted: 23 January 2023 / Published: 25 January 2023
(This article belongs to the Special Issue Molecular Mechanisms of Cardiac Development and Disease)

Abstract

:
Peroxisome proliferator activated receptors, including PPARα, PPARβ/δ, and PPARγ, are ligand-activated transcription factors belonging to the nuclear receptor superfamily. They play important roles in glucose and lipid metabolism and are also supposed to reduce inflammation and atherosclerosis. All PPARs are involved in angiogenesis, a process critically involved in cardiovascular pathology. Synthetic specific agonists exist for all PPARs. PPARα agonists (fibrates) are used to treat dyslipidemia by decreasing triglyceride and increasing high-density lipoprotein (HDL) levels. PPARγ agonists (thiazolidinediones) are used to treat Type 2 diabetes mellitus by improving insulin sensitivity. PPARα/γ (dual) agonists are supposed to treat both pathological conditions at once. In contrast, PPARβ/δ agonists are not in clinical use. Although activators of PPARs were initially considered to have favorable effects on the risk factors for cardiovascular disease, their cardiovascular safety is controversial. Here, we discuss the implications of PPARs in vascular biology regarding cardiac pathology and focus on the outcomes of clinical studies evaluating their benefits in cardiovascular diseases.

1. Introduction

Peroxisome Proliferator Activated Receptors are ligand-activated transcription factors regulating energy homeostasis. They include PPARα, PPARβ/δ, and PPARγ. PPARα is highly expressed in skeletal muscle, the heart, the liver, and brown adipose tissue and acts as a major regulator of fatty acid oxidation (FAO), thereby lowering lipid levels. PPARβ/δ is ubiquitously expressed and involved in lipid catabolism, inflammation, and energy dissipation. PPARγ expression is highest in adipose tissue, the large intestine, and the spleen, where it regulates adipogenesis, lipid and glucose metabolism, and inflammatory pathways. Thus, PPARs coordinate a variety of metabolic pathways upon activation by endogenous ligands, mainly produced in the metabolic pathways of fatty acids or synthetic modulators. After interaction with the ligands, PPARs are translocated to the nucleus, where they change their structure and regulate gene transcription. PPAR agonists have variable properties and specificities for individual PPAR receptors, different absorption and distribution profiles, and differing gene expression profiles, which accordingly lead to distinct clinical outcomes. PPARα agonists (fibrates) are used to treat dyslipidemia by decreasing triglyceride and increasing high-density lipoprotein (HDL) levels (reviewed in [1]). The PPARβ/δ agonist GW501516 entered clinical development as a drug candidate for cardiovascular and metabolic diseases, but clinical trials had to be stopped due to rapid cancer appearance observed in animal studies [2,3]. PPARγ agonists (thiazolidinediones) improve insulin sensitivity and are used in the therapy of Type 2 diabetes [1]. The effectiveness of these PPAR activators in the treatment of metabolic disorders, such as hyperlipidemia and diabetes, could consequently be considered to improve cardiovascular outcomes. Furthermore, all PPARs modulate angiogenesis, a critical process in cardiovascular pathologies. Although many research studies suggested beneficial effects of PPAR activation in the setting of cardiovascular disease (reviewed in [4,5]), they only have limited effects on cardiovascular outcomes in clinical studies (reviewed in [6,7,8]). In this review, we intend to summarize the angiogenic functions of PPARs with respect to their implications in cardiovascular pathology and analyze their utility in pharmacological therapies of cardiac disease states.

2. Peroxisome Proliferator Activated Receptors (PPARs)

The endothelial expression of all PPARs was demonstrated 20 years ago [9]. Although some conflicting results have been reported, in general, the activation of PPARα and PPARγ is linked to antiangiogenic effects, while PPARβ/δ activators favor angiogenesis (reviewed in [8]). Vascular aging is associated with structural and functional changes to the vasculature and represents an important risk factor for cardiovascular disease. Vascular aging leads to increased oxidative stress, disturbing metabolic and hemodynamic mechanisms (reviewed in [10,11]). Although reduced telomere length resulting in DNA damage, impaired replicative capacity of cells, and upregulated vascular cell senescence is clearly a hallmark of cardiovascular aging [12], it has been shown recently that, in most organs, an age-dependent diminution of endothelial cells and pericytes precedes vascular senescence. The implication is that the vascular cell loss drives senescence. The molecular analysis of endothelial cells from different tissues with age-induced vascular cell loss compared to endothelial cells from organs without age-related vascular cell disappearance revealed inflammatory processes as a common nominator in this process [13,14]. As all PPARs are considered to suppress vascular inflammation and atherogenesis (Figure 1) and be strongly involved in vascular metabolic and hemodynamic mechanisms, they are attractive candidates in the therapy of cardiovascular diseases.

2.1. Peroxisome Proliferator Activated Receptor Alpha (PPARα)

PPARα activation in the vasculature represses inflammatory responses, which involves the inhibition of the cytokine-induced vascular cell adhesion molecule-1 (VCAM-1), thus limiting the recruitment of inflammatory cells to the activated endothelium. This is, in part, achieved through the inhibition of the nuclear factor κB (NFκB) activation [15,16]. PPARα negative interference with NF-κB and activator protein (AP)-1 transcriptional activities [17] is further implicated in cyclooxygenase-2 (COX-2), interleukin-6, prostaglandin [18], monocytic tissue factor (TF) [19], and endothelin-1 (ET-1) [20] repression. PPARα activators enhance endothelial nitric oxide (NO) synthase expression and NO release [21,22]. Thus, the regulation of the vascular tone by stimulating NO-dependent vasodilation and inhibiting ET-1-induced vasoconstriction further contributes to the vasoprotective effects of PPARα activation.
PPARα antiangiogenic functions are mediated through decreased Akt (protein kinase B) activation, COX-2 [23], prostaglandin E (PGE)(2) [24], and matrix metallopeptidase (MMP) 9 [25], as well as vascular endothelial growth factor (VEGF) expression and an increase of antiangiogenic endostatin and thrombospondin (TSP)-1 [26]. A cardiomyocyte-specific overexpression of PPARα in genetic mouse models recapitulates a diabetic heart phenotype with cardiomyopathy due to lipid accumulation in cardiomyocytes. The expression of genes involved in cardiac fatty acid uptake and oxidation pathways increased while those of genes involved in glucose transport and utilization decreased, which consequently led to higher myocardial fatty acid oxidation rates and lower glucose uptake and oxidation [27,28]. Most animal studies applying ischemia/reperfusion or myocardial infarction procedures reported beneficial effects of PPARα agonists reflected by reduced infarct sizes and improved cardiac function (reviewed in [5]). In humans, the Fenofibrate Intervention and Event Lowering in Diabetes (FIELD) study randomized 9795 patients with Type 2 diabetes mellitus and dyslipidemia to fenofibrate or a placebo. Fenofibrate did not significantly reduce the composite primary end point of nonfatal myocardial infarction and coronary heart disease mortality. However, it reduced total cardiovascular events, mainly due to fewer nonfatal myocardial infarctions and revascularizations, as a 21% reduction of coronary vascularization was observed in the fenofibrate-treated subjects [29]. Clinical trials using PPARα agonists are summarized in Table 1. The ACCORD (action to control cardiovascular risk in diabetes) trial combined statin (simvastatin) therapy with fenofibrate in patients with Type 2 diabetes mellitus. The combination of the PPARα agonist fenofibrate and simvastatin did not reduce the overall rate of fatal cardiovascular events, nonfatal myocardial infarction, or nonfatal stroke as compared with simvastatin alone [30]. A newer pharmacogenetic analysis of the ACCORD-Lipid trial identified a common variant at the PPARA locus (rs6008845, C/T). T/T homozygous patients for this variant experienced a 51% reduction in major cardiovascular events in response to fibrate treatment, even in the absence of atherogenic dyslipidemia. Also, African Americans from the ACCORD cohort and from external cohorts (ACCORD-BP, ORIGIN, and TRIUMPH) showed a significant benefit from fibrate treatment for the T/T variant [31]. Interestingly, this variant was associated with lower levels of circulating CCL11, which is a proinflammatory and atherogenic cytokine [31]. Fibrate pharmacogenomics have been reviewed in detail by House & Motsinger-Reif [32]. The identification of PPARA common variants, which are associated with a clear cardiovascular benefit from fibrate treatment, justifies additional clinical studies. Unfortunately, although sequencing technologies are more easily available in recent years, it is still not common clinical practice to prescribe widely used drugs like fibrates on the basis of identified genetic variants.
The Helsinki heart study randomized 4180 men with primary dyslipidemia to the PPARα agonist gemfibrozil or a placebo. Gemfibrozil lowered the incidence of coronary heart disease by 34% [33]. Similarly, in the Veterans Affairs High-Density Lipoprotein Cholesterol Intervention Trial, which randomized 2531 men, Gemfibrozil therapy resulted in a significant reduction (22%) in the risk of major cardiovascular events in patients with coronary disease whose primary lipid abnormality was a low HDL cholesterol level [34,35]. The recent Pemafibrate to Reduce Cardiovascular Outcomes by Reducing Triglycerides in Patients with Diabetes (PROMINENT) trial for Pemafibrate - a highly active and selective PPARα agonist - in diabetic patients [36] was stopped in April 2022. Reviewing the results of an interim analysis, the Data Safety Monitoring Board (DSMB) concluded that it was unlikely that the primary endpoints would be met. The absolute treatment effects of fibrates in the primary prevention of cardiovascular events appear to be modest, as fibrates improve cardiovascular outcomes only in specific high-risk populations (fenofibrate in diabetic patients with metabolic syndrome, as well as gemfibrozil in patients with dyslipidemia).

2.2. Peroxisome Proliferator Activated Receptor Beta/Delta (PPARβ/δ)

Similar to PPARα and PPARγ, PPARβ/δ activation inhibits major vascular inflammatory responses through the inhibition of NFκB [37], VCAM-1 [37,38,39], E-selectin [38,39], and the intercellular adhesion molecule (ICAM)-1 [38,40]. In addition, PPARβ/δ controls inflammation through ligand-dependent interaction with B-cell lymphoma (BCL)-6. In the unliganded state, PPARβ/δ binds to the transcriptional repressor BCL-6. Upon ligand activation of PPARβ/δ, BCL-6 is released and can suppress pro-inflammatory pathways responsible for atherosclerosis formation [41]. PPARβ/δ further induces transforming growth factor (TGF) β1, leading to the induction of the antithrombotic plasminogen activator inhibitor-1 and the decrease of the proinflammatory chemokine monocyte chemoattractant protein (MCP)-1 [42]. PPARβ/δ also protects vascular cells from tissue-damaging oxidant stress, which itself would enhance inflammatory reactions. PPARβ/δ agonists induce heme oxygenase-1 expression, rendering endothelial cells in culture resistant to cellular stress induced by hydrogen peroxide or leptin [43]. PPARβ/δ further increases 14-3- 3α protein [44], superoxide dismutase-1, and catalase and thioredoxin expression [38]. The expression of these antioxidant genes represses reactive oxygen species (ROS) production in vascular cells, thus orchestrating a powerful antioxidant response.
In strong contrast to PPARα and PPARγ, the activation of PPARβ/δ induces pro-angiogenic responses in vitro and in vivo [45,46,47,48,49,50,51], reviewed in [8]. Pro-angiogenic targets of PPARβ/δ include VEGF [45], Cl−intracellular channel protein (CLIC) 4 [52], which is required for endothelial tube formation [53], calcineurin, which leads to the activation of nuclear factor of activated T cells (NFAT) c3 and the induction of pro-angiogenic hypoxia-inducible factor (HIF)-1 [48], as well as platelet-derived growth factor receptor (PDGFR) β and tyrosinkinase KIT (c-Kit) [50].
PPARβ/δ is the dominant PPAR subtype expressed in cardiac tissue [54]. The cardiomyocyte-specific overexpression of PPARβ/δ increases FAO and glycolysis. The increased capacity for myocardial glucose utilization was found to be beneficial in the setting of myocardial injury due to ischemia/reperfusion [28]. The cardiomyocyte-specific overexpression of PPARβ/δ further induced cell cycle progression of cardiomyocytes in myocardial infarction models, resulting in improved cardiac function [55]. The PPARβ/δ agonist GW610742X did not change left ventricular functional parameters or infarct sizes but normalized cardiac substrate metabolism and reduced right ventricular hypertrophy and pulmonary congestion after experimental myocardial infarction in rats [56]. Indirect cardioprotective functions of PPARβ/δ have been proposed in a study investigating the beneficial effects of remote ischemic preconditioning (rIPC) for cardiac protection after myocardial infarction. The protective effects of rIPC were mediated via the phosphoinositide 3-kinase (PI3K)/Akt/glycogen synthase kinase 3β (GSK3β) signaling pathway, which induced the nuclear accumulation of β-catenin and upregulation of its downstream targets E-cadherin and PPARβ/δ, which are involved in cell survival. Although the authors observed reduced apoptosis, infarct sizes, and improved functional recovery in mice, the exact impact of PPARβ/δ is not clear [57]. However, vascularization after myocardial infarction and the pro-angiogenic effects of PPARβ/δ on the functional outcome have not been investigated in any of these experimental studies. Our group used mice with inducible vascular-specific overexpression of PPARβ/δ, in which we found a rapid increase of cardiac vascularization accompanied by a fast onset of myocardial hypertrophy and impaired cardiac function. In addition, in the setting of myocardial infarction, PPARβ/δ vessel-specific overexpression increased capillary densities but failed to improve the outcome, as reflected by bigger infarct sizes, increased fibrosis, and significantly impaired echocardiographic parameters [49]. In line with our findings, treatment with the PPARβ/δ agonist GW610742 has been reported to increase vessel densities and fibrosis after myocardial infarction in rats without having functional benefits [58]. It seems that the specific, unbalanced activation of PPARβ/δ in only the vasculature is not sufficient to protect against the cardiomyocyte damage in chronic ischemic heart disease. Consequently, for PPARβ/δ modulation in cardiovascular disease, clinical trials are rare. The effects of the PPARβ/δ agonist GW501516 were investigated in pre-clinical trials to treat metabolic syndrome and diabetes at the beginning of 2000. Trials were stopped in 2007 due to the appearance of multiple cancers in mice and rats [3]. Not surprisingly, no clinical trials for the use of selective PPARβ/δ agonists in cardiovascular disease exist. The angiotensin II receptor blocker telmisartan targets PPARβ/δ [59] as well as PPARγ [60,61]. Two clinical trials for telmisartan have been completed: TRANSCEND (Telmisartan Randomized Assessment Study in ACE-Intolerant Subjects with Cardiovascular Disease) and ONTARGET (Ongoing Telmisartan Alone and in Combination with Ramipiril Global Endpoint Trial). In terms of primary and secondary outcomes, no significant differences were observed between the groups, with the exception of female patients who showed a 20% overall risk reduction for myocardial infarction [62]. If this limited beneficial effect of telmisartan can be attributed to angiotensin II receptor blockade, PPARγ activation, or PPARβ/δ activation is difficult to conclude.

2.3. Peroxisome Proliferator Activated Receptor Gamma (PPARγ)

Like the other PPARs, PPARγ activation inhibits tissue and endothelial cell inflammatory responses. PPARγ agonists decrease expression of the adhesion molecule ICAM-1 by suppressing NF-κB/DNA binding activity [63]. They inhibit pro-inflammatory molecules such as MMP-9 [64], chemokines monocyte chemoattractant protein (MCP)-1 [65], IFN-inducible protein of 10 kDa (IP-10), monokine induced by IFN-gamma (Mig), and IFN-inducible T-cell alpha-chemoattractant (I-TAC) [66], as well as cytokine IL-8 [67] and vasoactive mediators inducible nitric oxide synthase (iNOS) [68] and ET-1 [17]).
The activation of PPARγ has mostly been demonstrated to inhibit angiogenesis [9,69]. PPARγ agonists do not only suppress transcription of VEGF [70], but also of the vascular endothelial growth factor receptor (VEGFR) 2 [71]. Antiangiogenic effects of PPARγ activation are further attributed to a pro-apoptotic mechanism due to PPARγ-mediated increased NO production [72], simultaneous activation of the p38 mitogen activated protein kinase (MAPK) pathway and inhibition of phosphorylation of p42/44 MAPK [73], and the suppression of protein kinase C (PKC)α -and C-AMP Response Element-binding protein (CREB)-mediated COX-2 expression in endothelial cells [74]. However, some in vivo studies establish angiogenic effects of PPARγ activation. In diabetic mice, blood flow recovery was impaired after hindlimb ischemia compared to nondiabetic controls. In diabetic mice, treatment with PPARγ agonists partially restored blood flow recovery and increased capillary density in ischemic hindlimbs [75,76]. Rosiglitazone increased angiogenesis, reduced apoptosis, improved functional recovery, and diminished the lesion size in animals with focal cerebral ischemia [77]. Pro-angiogenic effects of PPARγ agonist treatment in ischemic tissues are NO-dependent and related to endothelial nitric oxide synthase (eNOS) upregulation [76]. Although these studies seem to contradict the results of in vitro studies, they better take into account the interplay between different cell types, which might be responsible for the pro-angiogenic action of PPARγ activation in pathological in vivo situations. The cardiomyocyte-specific knockout of PPARγ induced cardiac hypertrophy, probably through the lack of inhibition of NF-κB [78], whereas the overexpression of PPARγ in cardiomyocytes resulted in cardiac myopathy characterized by increased lipid and glycogen storage and cardiac dysfunction [79]. Interestingly, the knockout of PPARγ in myeloid cells worsened the outcome after experimental myocardial infarction in mice, likely due to increased oxidative stress and cardiac inflammation [80]. Given the favorable anti-inflammatory and antiatherogenic effects of PPARγ activation in atherosclerosis, PPARγ agonists might lower the risk of cardiovascular disease. Accordingly, experimental animal studies using myocardial ischemia/reperfusion experiments all reported beneficial aspects with PPARγ activators (reviewed in [5]). The first small clinical trials showed some benefits of pioglitazone mainly concerning the amelioration of atherosclerotic inflammation [81,82,83,84]. Clinical trials using PPARγ agonists are summarized in Table 2. In 2005, the large PROspective pioglitAzone Clinical Trial In macroVascular Events (PROactive) trial randomized 5238 diabetic patients with macrovascular cardiovascular disease to pioglitazone or a placebo. The primary end points were all-cause mortality, nonfatal myocardial infarction, strokes, acute coronary syndromes, endovascular or surgical intervention in the coronary or leg arteries, and amputation above the ankle. No reduction in primary end points with pioglitazone could be demonstrated. However, pioglitazone reduced a composite secondary end point of all-cause mortality, nonfatal myocardial infarction, and strokes [85,86,87]. Later, in 2008, the PERISCOPE (Pioglitazone Effect on Regression of Intravascular Sonographic Coronary Obstruction Prospective Evaluation) trial compared the effects of pioglitazone and glimepiride (an antidiabetic Insulin secretagogue) in 543 patients with diabetes mellitus and coronary disease. The primary outcome was a change in the percentage of the atheroma volume from the baseline. After 18 months of follow-up, pioglitazone effectively reduced the progression of coronary atherosclerosis compared with glimepiride. The incidence of cardiovascular death and nonfatal myocardial infarction did not differ between the groups [88]. The more recent IRIS (Insulin Resistance Intervention After Stroke) trial evaluated the effectiveness of pioglitazone in patients with insulin resistance and a recent event of a stroke or a transient ischemic attack. The primary outcome was a fatal or nonfatal stroke, or a myocardial infarction. After five years, in 11.8% of the placebo group, a primary outcome event occurred. This was diminished to 9% in the pioglitazone cohort. Pioglitazone therapy was associated with higher risks of weight gain, edema, and fractures. Incident bladder cancer occurred in 12 patients in the pioglitazone group and in eight in the placebo group [89]. Thus, although pioglitazone might improve cardiovascular outcomes, there are major noncardiovascular safety concerns, such as an increased risk of bladder cancer (reviewed in [90]). In 2006, rosiglitazone was investigated in patients with impaired glucose tolerance. In the DREAM (Diabetes Reduction Assessment with Ramipiril and Rosiglitazone Medication) trial, 5962 patients were randomized to ramipiril (an angiotensin-converting enzyme inhibitor), rosiglitazone, ramipiril and rosiglitazone, and a placebo. The composite primary endpoint after three years was an incidence of diabetes, which was significantly lower with rosiglitazone. On the other hand, an increase in cardiovascular vents and congestive heart failure was observed in the patients receiving rosiglitazone [91]. In the ADOPT (A Diabetes Outcome Progression) trial, 4360 patients with diabetes were randomized to rosiglitazone, metformin, or glyburide. The primary outcome was the time to reach monotherapy failure, which was significantly reduced with rosiglitazone compared to the other two antidiabetic drugs [92]. In a meta-analysis study from 2007, 42 clinical trials were evaluated concerning the effects of rosiglitazone on the risk of myocardial infarction and death from cardiovascular causes, including the ADOPT and DREAM trials. The authors could conclude that rosiglitazone was associated with a significant increase in the risk of myocardial infarction and an increase in the risk of death from cardiovascular causes [93]. In the already ongoing RECORD (Rosiglitazone Evaluated for Cardiac Outcomes and Regulation of Glycaemia in Diabetes) trial, which assessed the addition of rosiglitazone to either metformin or sulfonylurea compared with the combination of the two in 4447 patients with diabetes mellitus, the addition of rosiglitazone to glucose-lowering therapy in patients with diabetes increased the risk of heart failure. Although the data were inconclusive about possible effects on myocardial infarction, rosiglitazone did not increase the risk of overall cardiovascular morbidity or mortality compared with standard glucose-lowering drugs [94]. A U.S. Food and Drug Administration (FDA) advisory meeting concluded that rosiglitazone carried considerable cardiovascular risks (including myocardial infarction risks [95]), leading to withdrawal in Europe and strong restrictions in the U.S.A.
As for PPARα, also PPARγ variants modulate the risk for diabetes and cardiovascular events. The PPARG Pro12Ala allele was associated with a reduced risk of myocardial infarction in a Type 2 diabetic population, while the T allele of the C1431T polymorphism was associated with increased all-cause mortality [96]. A study using a community-based cohort from Maryland did not find associations between PPAR polymorphisms and the risk of cardiovascular morbidity and mortality [97].
Pan PPAR agonists (Table 3) were designed in order to treat dyslipidemia and diabetes with a single drug (reviewed in [98]). Bezafibrate is the first and most studied PPAR pan agonist. In 1998, the SENDCAP (St. Mary’s, Ealing, Northwick Park Diabetes Cardiovascular Disease Prevention) study randomized 164 diabetes patients to bezafibrate or a placebo. Bezafibrate improved dyslipidemia but had no effect on the progress of arterial disease. Nevertheless, it significantly decreased the incidence of definite coronary heart disease events over 3 years [99]. Later, the BIP (Bezafibrate infarction Prevention) trial randomized 3090 patients with previous myocardial infarction or stable angina and hyperlipidemia to bezafibrate or a placebo. The composite endpoints were myocardial infarction or death. Bezafibrate effectively lowered triglycerides, but only an overall, not significant trend to reduce the incidence of primary end points was observed [100].
The experimental PPAR pan agonists muraglitazar, ragaglitazar, and tesaglitazar were initially promising, but serious side effects were observed during testing. Clinical trials using pan-or dual (α/γ) PPAR agonists for cardiovascular disease are summarized in Table 3. The meta-analysis of five clinical trials involving 2374 patients exposed to muraglitazar and 1351 patients exposed to comparator agents, of which 823 received pioglitazone and 528 a placebo, revealed that muraglitazar was associated with an excess incidence of death and major adverse cardiovascular events [101]. Clinical trials for ragaglitazar have been stopped after the discovery of its carcinogenic effects on rodent bladders [102]. The clinical development of tesaglitazar has been discontinued due to its serum creatinine increasing the effect in diabetic patients [103]. Later trials, starting in 2006 with the dual PPARα/γ agonist aleglitazar (Synchrony, AleCardio, and Aleprevent randomized clinical trials), showed no significant improvement of cardiovascular disease but multiple negative side effects such as oedema, hemodilution, weight gain in the Synchrony trial [104], renal dysfunction and heart failure in the AleCardio trial [105]. The Aleprevent study had to be halted prematurely after the randomization of 1999 patients, although the initially intended sample size was 19,000 patients, due to hypoglycemia, edema, and adverse muscular events [106]. Given these unfavorable results, the clinical development of dual-PPAR agonist for cardiovascular disease has been stopped. Although glitazars improved metabolic parameters, they induced cardiac failure, which is probably due to glucolipotoxicity through the combined increase of PPARγ-driven insulin sensitization and glucose uptake in the setting of higher PPARα-induced activation of fatty acid uptake and oxidation. Unfortunately, the impact of a “doubled” inhibition of angiogenesis through simultaneous activation of PPARα and γ has never been investigated, but the lack of severe cardiovascular side effects in the pan agonist trials with bezafibrate could indicate that a balanced angiogenic response by activation of pro-angiogenic PPARβ/δ and antiangiogenic PPARα and γ could avoid major cardiovascular adverse effects.
Table 3. Summary of Clinical Trials with pan-or dual (α/γ) PPAR agonists for Cardiovascular Disease.
Table 3. Summary of Clinical Trials with pan-or dual (α/γ) PPAR agonists for Cardiovascular Disease.
Study Name/DrugConditionPrimary EndpointsOutcomeTrial Identifier/Reference
St. Mary’s, Ealing, Northwick Park Diabetes Cardiovascular Disease Prevention (SENDCAP) study
Bezafibrate
164 diabetic patientsAlteration of cardiovascular outcomesNo effect on the progress of ultrasonically measured arterial disease[99]
Bezafibrate infarction Prevention (BIP) trial
Bezafibrate
3090 patients with coronary artery diseaseMyocardial infarction, sudden deathNo reduction of myocardial infarction or death[100]
Metanalysis of five clinical trials with Muraglitazar in Diabetic Patients
Muraglitazar
3725 diabetic patientsMajor adverse cardiovascular events, deathExcess incidence of death, myocardial infarction, congestive heart failure[101]
Tesaglitazar vs. Placebo in Combination With Insulin (GALLANT9) trial Tesaglitazar370 diabetic patientsAbsolute change from baseline in glycosylated hemoglobin A1c (HbA1c) Reduction of HbA1c), significant increase in serum creatinineNCT00242372 [103]
A Study of Aleglitazar in Patients With Type 2 Diabetes (SYNCHRONY) trial332 diabetic patientsAbsolute change from baseline in glycosylated hemoglobin A1c (HbA1c)Reduction of HbA1c), oedema, hemodilution, and weight gain NCT00388518 [104]
Aleglitazar to Reduce Cardiovascular Risk in (CHD) Patients With a Recent Acute Coronary Syndrome (ACS) Event and Type 2 Diabetes Mellitus (T2D) (AleCardio) trial7226 diabetic patients with myocardial infarction or unstable anginaEffect on cardiovascular death, nonfatal myocardial infarction, and nonfatal strokeNo reduction in the risk of cardiovascular outcomesNCT01042769
[105]
Aleglitazar To Reduce Cardiovascular Risk In Patients With Stable Cardiovascular Disease And Glucose Abnormalities1999 diabetic patients with cardiovascular diseaseReduction of cardiovascular morbidity and mortalityNo reduction in the risk of cardiovascular outcomesEudraCT Number: 2012-000671-16
[106]

3. Cardiovascular Disease Therapies Involving PPARs

To not completely dismiss the utility of PPARs in cardiovascular disease therapy, it is important to note that many conventional pharmacological therapies (Aspirin, Statins, Angiotensin-converting enzyme [ACE] inhibitors, and Angiotensin receptor blockers [ARBs]) for primary or secondary prevention of myocardial infarction often affect PPARs. Therefore, we include a brief review regarding their potential PPAR-related effects on cardiovascular pathophysiology.

3.1. Aspirin

Aspirin is one of the most frequently used drugs worldwide and is considered to be effective in the prevention of cardiovascular disease, mainly through its antithrombotic effects (reviewed in [107]). Additionally, it reduces cancer risk and mortality (reviewed in [108]). PPARα has recently been identified as a receptor for aspirin in the neuronal system [109]. Aspirin exerts neuroprotective functions through the stimulation of neurotrophin production, mediated through its binding to PPARα [110]. Neurotrophins are involved in the maintenance of cardiometabolic homeostasis and cardioprotection (reviewed in [111]), which might represent one aspect of the cardiovascular benefit of aspirin therapy mediated through PPARα. For aspirin, mainly antiangiogenic effects have been reported: it suppressed collateral artery growth in animal studies, possibly through cyclooxygenase (COX) inhibition [112]. Interestingly, antiangiogenic effects of PPARα activation have also been attributed to COX inhibition [23]. However, antiangiogenic effects of aspirin can’t be solely attributed to COX inhibition [113]. Aspirin reduces the production of pro-angiogenic 11-hydroxyeicosatetraenoic acid (HETE) and 15(S)-HETE [114], whereas PPARα ligands reduce the production of AA-derived epoxyeicosatrienoic acids (EETs) and increase their pro-angiogenic hydroxyl product, 11-HETE [115], possibly reflecting a balanced angiogenic response in the setting of cardiovascular and acute ischemic disease. In a small clinical study with patients with myocardial ischemia undergoing coronary artery bypass graft operation, aspirin significantly reduced VEGF release [116], an antiangiogenic mechanism also assigned to PPARα and γ activation (reviewed in [8]). It would be interesting to determine to what extent antiangiogenic effects of aspirin are mediated by PPAR activation.

3.2. Statins

Statins are frequently prescribed medications in the management of high cholesterol and associated cardiovascular complications. They lower cholesterol through inhibition of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA)-reductase [117]. Statin use is associated with a significantly reduced risk of all-cause mortality in cohorts with cardiovascular and noncardiovascular disease, including cancer [118]. Not only antiatherogenic and anti-inflammatory actions of statins have been associated with the activation of PPARα and γ ([119,120,121,122], but also cardioprotective statin effects, such as the inhibition of cardiac hypertrophy and fibrosis, can be attributed to PPARα and γ modulation [123,124,125]. Furthermore, statins have the potential to lower persistent elevated blood pressure levels through mechanisms implying PPAR activation [126]. In addition, the inhibition of platelet aggregation by statins is mediated through PPARs [127], which additionally contributes to vascular defensive effects of statins. Angiogenesis is stimulated at low (nanomolar) statin concentrations and inhibited by high (micromolar) doses [128,129,130]. Mechanisms of the pro-angiogenic effects of statins include activation of the (PI3K)/Akt pathway [130], an increase of VEGF, IL-8, Angiopoietin 1 and 2, eNOS, and heme oxygenase (HO)-1 expression [131]. This correlates quite well with the pro-angiogenic mechanisms of PPARβ/δ [132] and might indicate that angiogenic effects of statins are partially mediated by PPARs. This notion is further supported by the finding that statins induce transcriptional activation of PPARα/retinoid X receptor (RXR)α, PPARβ/δ/RXRα, and PPARγ2/RXRα [133].

3.3. Angiotensin-Converting Enzyme (ACE) Inhibitors

ACE inhibitors are commonly used in patients with hypertension, heart failure, coronary artery disease, diabetes, and chronic renal disease. They inhibit the activity of angiotensin-converting enzymes, an important component of the renin–angiotensin system that converts angiotensin I to angiotensin II (AngII). AngII promotes vascular inflammation through NF-κB-mediated induction of pro-inflammatory genes and downregulation of PPARα and γ [134]. ACE inhibitors upregulate PPAR expression [135,136]. Like ACE inhibitors, PPARs inhibit AngII synthesis [137]. Therefore, they also contribute to antiatherogenic and anti-inflammatory effects. In chronic daunorubicin-induced cardiomyopathy, ACE inhibition reestablished normal PPARβ/δ and γ levels, along with a reduction of NADPH (nicotinamide adenine dinucleotide phosphate) oxidase. However, a possible causal link between these effects has not been investigated [138]. Ang II enhances angiogenesis through angiotensin Type 1 receptor activation (AT1R) that involves VEGF/eNOS-dependent pathways [139]. Both subtypes of receptors, AT1R and AT2R, are implicated in angiogenic processes [140,141]. ACE inhibitors do not solely inhibit the activity of pro-angiogenic AngII, but also the degradation of bradykinin, which mediates pro-angiogenic functions through binding to its receptors B1 and B2 [142]. Not surprisingly, the inhibition of ACE, affecting AngII and bradykinin degradation, has different impacts on angiogenesis. ACE inhibitors were reported to promote angiogenesis in hindlimb ischemia models [143,144,145] and to promote myocardial capillary formation [146,147,148,149]. An analysis of ACE inhibitor effects on retinal and hindlimb angiogenesis in diabetic mice revealed an induction of neovascularization in the hindlimbs through the activation of bradykinin signaling, and a reduction of retinal vascularization through the inhibition of AngII [150]. In addition, in a rabbit model of VEGF-induced corneal vascularization, ACE inhibition significantly reduced angiogenesis [151]. Similarly, in exercise-induced angiogenesis, ACE inhibitors reduced VEGF expression and neovascularization [152]. Importantly, although ACE inhibitors have some pro-angiogenic effects, they reduce tumor angiogenesis [153,154,155]. A meta-analysis of randomized trials with ACE inhibitors did not find any impact on the occurrence of cancer or cancer death [156], and ACE inhibition is nowadays considered as a possible complementary treatment option for cancer patients (reviewed in [157].

3.4. Angiotensin Receptor Blockers (ARBs)

ARBs were developed to overcome some deficiencies of ACE inhibitors: competitive inhibition of ACE results in a reactive increase in renin and angiotensin I levels, which may overcome the blockade effect. ACE is a relatively nonspecific enzyme that has substrates in addition to angiotensin I, including bradykinin. Therefore, the inhibition of ACE may result in accumulation; the production of AngII can occur through non-ACE pathways. Therefore, ARBs may offer more complete AngII inhibition by interacting selectively with the angiotensin Type 1 receptor site (reviewed in [158]). Their indications are the same as those for ACE inhibitors. The ARBs irbesartan and, mostly, telmisartan induce PPARγ activity [61,159] and were, therefore, considered as highly beneficial in treating cardiovascular, metabolic, and inflammatory diseases in insulin-resistant and euglycemic states reviewed in [160]. Interestingly, telmisartan provides a triple inhibition of angiotensin II function: AT1R blockade, downregulation of AT1R expression [161], and ACE inhibition [162]. The reduced expression of AT1R and vascular ACE inhibition were attributed to PPARγ activation [161,162]. Telmisartan further elicits anti-inflammatory and antiatherosclerotic effects through the activation of PPARγ, involving reduced activity of the pro-inflammatory transcription factors NF-kB and early-growth response protein (Egr)-1 [163], promotes the proliferation of endothelial progenitor cells [164], and improves microvascular dysfunction during myocardial ischemia/reperfusion injury [165] via PPARγ dependent pathways. Telmisartan was approved by the FDA for the treatment of hypertension in November 1998. In 2009, telmisartan was the first ARB to be granted FDA approval for the reduction of cardiovascular risk in high-risk patients unable to take ACE inhibitors. In addition to telmisartan, other ARBs have been described to activate PPARγ [166,167,168,169,170,171,172], but also PPARα [173,174,175,176,177] and PPARβ/δ [175,178,179]. PPARβ/δ activation by telmisartan has been reported to reduce cardiac fibrosis in diabetic animals [179], inhibit inflammation of endothelial cells [180], ameliorate insulin resistance in skeletal muscle [178], prevent obesity [181], exert neuroprotective effects [182], and even improve symptoms of stress-induced depression [183].
In general, ARBs have antiangiogenic effects. They were shown to inhibit neoangiogenesis in ischemia hindlimb models [184], impair VEGF-induced revascularization in cardiomyopathy [185] and diabetes [140], and impede hypoxia-induced coronary angiogenesis [186]. The combined blockade of angiotensin Type 1 receptor and the activation of PPARγ by telmisartan inhibited vascularization in several models of angiogenesis [187]. However, PPARγ-independent mechanisms in the inhibition of vascular cell proliferation by telmisartan, involving repression of Akt, have been postulated [188].
Importantly, ARBs inhibit tumor angiogenesis and tumor growth, involving the repression of VEGF [189,190]. Telmisartan was shown to exert antitumor effects in vitro through PPARγ activation and subsequent downregulation of MMP9 and ICAM-1 [191,192].
Despite these encouraging experimental results, a first meta-analysis of randomized controlled trials in 2010 suggested that ARBs, mainly telmisartan, are associated with a modestly increased risk of new cancer diagnosis [193]. In a novel meta-analysis of the same year, covering 70 randomized controlled trials for antihypertensive drugs, the 5–10% relative increase in the risk of cancer or cancer-related death with the use of ARBs established in the former study was refuted [194]. Accordingly, a meta-analysis of 15 trials for different ARBs enrolling 138,769 patients did not find an excess of cancer incidence [195].
Recently, the design of tumor microenvironment-activated ARBs has been proposed to reprogram cancer associated fibroblasts to an immunosupportive state, diminishing immunosuppression and improving T lymphocyte activity in cancer [196].

4. Conclusions

In conclusion, the utility of PPAR agonists for the treatment of cardiovascular disease, beneficial modelling of angiogenesis, and myocardial infarction prevention is limited. Although they seem to reduce cardiovascular risk factors, they failed to improve cardiovascular outcomes in clinical trials, were only moderately effective in very selective subgroups of patients, or even produced severe side effects. The observed heterogenous effects of PPAR agonists in clinical studies highlight the importance of a strict post marketing surveillance. Established conventional cardiovascular medications often include PPAR-activating mechanisms, which might contribute to their beneficial low risk effects. Balanced indirect or coupled PPAR activation by these medications seems to be preferable to unique, unbalanced PPAR activation in the setting of cardiovascular diseases.

Author Contributions

Conceptualization, N.W. and K.-D.W.; writing—original draft preparation, N.W. and K.-D.W.; writing—review and editing, N.W. and K.-D.W.; funding acquisition, N.W. and K.-D.W. All authors have read and agreed to the published version of the manuscript.

Funding

The authors of this review were funded by Fondation pour la Recherche Medicale, grant number FRM DPC20170139474 (K.D.W.), Fondation ARC pour la recherche sur le cancer”, grant number n°PJA 20161204650 (N.W.), Gemluc (N.W.), and Plan Cancer INSERM (K.-D.W.)

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ACEAngiotensin Converting Enzyme Inhibitor
AktProtein Kinase B
AngII Angiotensin II
ARBAngiotensin Receptor Blocker
AP-1Activator Protein-1
AT1R Angiotensin Type 1 Receptor
BCL-6B-cell lymphoma-6
c-KitTyrosinkinase Kit
CLIC 4Cl−intracellular channel protein 4
COX-2Cyclooxygenase-2
CREBC-AMP Response Element-binding protein
EGREarly Growth Response Protein
eNOSendothelial Nitric Oxide Synthase
ET-1Endothelin-1
FAOFatty Acid Oxidation
FDA Food and Drug Administration
GSK3βglycogen synthase kinase 3β
HDLHigh Density Lipoprotein
HETEhydroxy eicosatetraenoic acid
HIF-1Hypoxia Inducible Factor 1
HMG-CoA 3-hydroxy-3-methylglutaryl coenzyme A
HOHeme Oxygenase
ICAM-1Intercellular Adhesion Molecule-1
IP-10 IFN-inducible protein of 10 kDa
I-TACIFN-inducible T-cell alpha-chemoattractant
MAPKMitogen Activated Protein Kinase
MCPMonocyte Chemoattractant Protein
MMPMatrix Metallopeptidase
Migmonokine induced by IFN-gamma
NADPHnicotinamide adenine dinucleotide phosphate
NFκBnuclear factor κB
NONitric Oxide
NFATnuclear factor of activated T cells
PDGFRPlatelet-derived Growth Factor Receptor
PI3Kphosphoinositide 3-kinase
PPARPeroxisome proliferator activated receptor
PGEProstaglandin E
PKCProtein Kinase C
rIPCRemote Ischemic Preconditioning
RXRRetinoid X Receptor
TSPThrombospondin
TFTissue Factor
TGFTransforming Growth Factor
VCAM-1Vascular Cell Adhesion Molecule 1
VEGFVascular Endothelial Growth Factor
VEGFRVascular Endothelial Growth Factor Receptor

References

  1. Montaigne, D.; Butruille, L.; Staels, B. PPAR control of metabolism and cardiovascular functions. Nat. Rev. Cardiol. 2021, 18, 809–823. [Google Scholar] [CrossRef]
  2. Sahebkar, A.; Chew, G.T.; Watts, G.F. New peroxisome proliferator-activated receptor agonists: Potential treatments for atherogenic dyslipidemia and non-alcoholic fatty liver disease. Expert. Opin. Pharmacother. 2014, 15, 493–503. [Google Scholar] [CrossRef]
  3. Mitchell, J.A.; Bishop-Bailey, D. PPARβ/δ a potential target in pulmonary hypertension blighted by cancer risk. Pulm. Circ. 2019, 9, 2053. [Google Scholar] [CrossRef] [Green Version]
  4. Khuchua, Z.; Glukhov, A.I.; Strauss, A.W.; Javadov, S. Elucidating the Beneficial Role of PPAR Agonists in Cardiac Diseases. Int. J. Mol. Sci. 2018, 19, 3464. [Google Scholar] [CrossRef] [Green Version]
  5. Wagner, K.D.; Wagner, N. PPARs and Myocardial Infarction. Int. J. Mol. Sci. 2020, 21, 9436. [Google Scholar] [CrossRef]
  6. Friedland, S.N.; Leong, A.; Filion, K.B.; Genest, J.; Lega, I.C.; Mottillo, S.; Poirier, P.; Reoch, J.; Eisenberg, M.J. The cardiovascular effects of peroxisome proliferator-activated receptor agonists. Am. J. Med. 2012, 125, 126–133. [Google Scholar] [CrossRef] [PubMed]
  7. Cheng, H.S.; Tan, W.R.; Low, Z.S.; Marvalim, C.; Lee, J.Y.H.; Tan, N.S. Exploration and Development of PPAR Modulators in Health and Disease: An Update of Clinical Evidence. Int. J. Mol. Sci. 2019, 20, 5055. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Wagner, N.; Wagner, K.D. PPARs and Angiogenesis-Implications in Pathology. Int. J. Mol. Sci. 2020, 21, 5723. [Google Scholar] [CrossRef] [PubMed]
  9. Bishop-Bailey, D.; Hla, T. Endothelial cell apoptosis induced by the peroxisome proliferator-activated receptor (PPAR) ligand 15-deoxy-Delta12, 14-prostaglandin J2. J. Biol. Chem. 1999, 274, 17042–17048. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Lakatta, E.G.; Levy, D. Arterial and cardiac aging: Major shareholders in cardiovascular disease enterprises: Part I: Aging arteries: A “set up” for vascular disease. Circulation 2003, 107, 139–146. [Google Scholar] [CrossRef] [Green Version]
  11. Lakatta, E.G.; Levy, D. Arterial and cardiac aging: Major shareholders in cardiovascular disease enterprises: Part II: The aging heart in health: Links to heart disease. Circulation 2003, 107, 346–354. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Minamino, T.; Komuro, I. Vascular aging: Insights from studies on cellular senescence, stem cell aging, and progeroid syndromes. Nat. Clin. Pract. Cardiovasc. Med. 2008, 5, 637–648. [Google Scholar] [CrossRef] [PubMed]
  13. Chen, J.; Lippo, L.; Labella, R.; Tan, S.L.; Marsden, B.D.; Dustin, M.L.; Ramasamy, S.K.; Kusumbe, A.P. Decreased blood vessel density and endothelial cell subset dynamics during ageing of the endocrine system. EMBO J. 2021, 40, e105242. [Google Scholar] [CrossRef] [PubMed]
  14. Chen, J.; Sivan, U.; Tan, S.L.; Lippo, L.; De Angelis, J.; Labella, R.; Singh, A.; Chatzis, A.; Cheuk, S.; Medhghalchi, M.; et al. High-resolution 3D imaging uncovers organ-specific vascular control of tissue aging. Sci. Adv. 2021, 7, eabd7819. [Google Scholar] [CrossRef]
  15. Marx, N.; Sukhova, G.K.; Collins, T.; Libby, P.; Plutzky, J. PPARalpha activators inhibit cytokine-induced vascular cell adhesion molecule-1 expression in human endothelial cells. Circulation 1999, 99, 3125–3131. [Google Scholar] [CrossRef] [Green Version]
  16. Chinetti, G.; Griglio, S.; Antonucci, M.; Torra, I.P.; Delerive, P.; Majd, Z.; Fruchart, J.C.; Chapman, J.; Najib, J.; Staels, B. Activation of proliferator-activated receptors alpha and gamma induces apoptosis of human monocyte-derived macrophages. J. Biol. Chem. 1998, 273, 25573–25580. [Google Scholar] [CrossRef] [Green Version]
  17. Delerive, P.; De Bosscher, K.; Besnard, S.; Vanden Berghe, W.; Peters, J.M.; Gonzalez, F.J.; Fruchart, J.C.; Tedgui, A.; Haegeman, G.; Staels, B. Peroxisome proliferator-activated receptor alpha negatively regulates the vascular inflammatory gene response by negative cross-talk with transcription factors NF-kappaB and AP-1. J. Biol. Chem. 1999, 274, 32048–32054. [Google Scholar] [CrossRef] [Green Version]
  18. Staels, B.; Koenig, W.; Habib, A.; Merval, R.; Lebret, M.; Torra, I.P.; Delerive, P.; Fadel, A.; Chinetti, G.; Fruchart, J.C.; et al. Activation of human aortic smooth-muscle cells is inhibited by PPARalpha but not by PPARgamma activators. Nature 1998, 393, 790–793. [Google Scholar] [CrossRef]
  19. Neve, B.P.; Corseaux, D.; Chinetti, G.; Zawadzki, C.; Fruchart, J.C.; Duriez, P.; Staels, B.; Jude, B. PPARalpha agonists inhibit tissue factor expression in human monocytes and macrophages. Circulation 2001, 103, 207–212. [Google Scholar] [CrossRef] [Green Version]
  20. Martin-Nizard, F.; Furman, C.; Delerive, P.; Kandoussi, A.; Fruchart, J.C.; Staels, B.; Duriez, P. Peroxisome proliferator-activated receptor activators inhibit oxidized low-density lipoprotein-induced endothelin-1 secretion in endothelial cells. J. Cardiovasc. Pharmacol. 2002, 40, 822–831. [Google Scholar] [CrossRef] [PubMed]
  21. Omura, M.; Kobayashi, S.; Mizukami, Y.; Mogami, K.; Todoroki-Ikeda, N.; Miyake, T.; Matsuzaki, M. Eicosapentaenoic acid (EPA) induces Ca(2+)-independent activation and translocation of endothelial nitric oxide synthase and endothelium-dependent vasorelaxation. FEBS Lett. 2001, 487, 361–366. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Goya, K.; Sumitani, S.; Xu, X.; Kitamura, T.; Yamamoto, H.; Kurebayashi, S.; Saito, H.; Kouhara, H.; Kasayama, S.; Kawase, I. Peroxisome proliferator-activated receptor alpha agonists increase nitric oxide synthase expression in vascular endothelial cells. Arterioscler. Thromb. Vasc. Biol. 2004, 24, 658–663. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Varet, J.; Vincent, L.; Mirshahi, P.; Pille, J.V.; Legrand, E.; Opolon, P.; Mishal, Z.; Soria, J.; Li, H.; Soria, C. Fenofibrate inhibits angiogenesis in vitro and in vivo. Cell Mol. Life Sci. 2003, 60, 810–819. [Google Scholar] [CrossRef] [PubMed]
  24. Yokoyama, Y.; Xin, B.; Shigeto, T.; Umemoto, M.; Kasai-Sakamoto, A.; Futagami, M.; Tsuchida, S.; Al-Mulla, F.; Mizunuma, H. Clofibric acid, a peroxisome proliferator-activated receptor alpha ligand, inhibits growth of human ovarian cancer. Mol. Cancer Ther. 2007, 6, 1379–1386. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Meissner, M.; Berlinski, B.; Gille, J.; Doll, M.; Kaufmann, R. Peroxisome proliferator activated receptor-α agonists suppress transforming growth factor-α-induced matrix metalloproteinase-9 expression in human keratinocytes. Clin. Exp. Dermatol. 2011, 36, 911–914. [Google Scholar] [CrossRef]
  26. Panigrahy, D.; Kaipainen, A.; Huang, S.; Butterfield, C.E.; Barnés, C.M.; Fannon, M.; Laforme, A.M.; Chaponis, D.M.; Folkman, J.; Kieran, M.W. PPARalpha agonist fenofibrate suppresses tumor growth through direct and indirect angiogenesis inhibition. Proc. Natl. Acad. Sci. USA 2008, 105, 985–990. [Google Scholar] [CrossRef] [Green Version]
  27. Finck, B.N.; Lehman, J.J.; Leone, T.C.; Welch, M.J.; Bennett, M.J.; Kovacs, A.; Han, X.; Gross, R.W.; Kozak, R.; Lopaschuk, G.D.; et al. The cardiac phenotype induced by PPARalpha overexpression mimics that caused by diabetes mellitus. J. Clin. Investig. 2002, 109, 121–130. [Google Scholar] [CrossRef]
  28. Burkart, E.M.; Sambandam, N.; Han, X.; Gross, R.W.; Courtois, M.; Gierasch, C.M.; Shoghi, K.; Welch, M.J.; Kelly, D.P. Nuclear receptors PPARbeta/delta and PPARalpha direct distinct metabolic regulatory programs in the mouse heart. J. Clin. Investig. 2007, 117, 3930–3939. [Google Scholar] [CrossRef]
  29. Keech, A.; Simes, R.J.; Barter, P.; Best, J.; Scott, R.; Taskinen, M.R.; Forder, P.; Pillai, A.; Davis, T.; Glasziou, P.; et al. Effects of long-term fenofibrate therapy on cardiovascular events in 9795 people with type 2 diabetes mellitus (the FIELD study): Randomised controlled trial. Lancet 2005, 366, 1849–1861. [Google Scholar] [CrossRef]
  30. Ginsberg, H.N.; Elam, M.B.; Lovato, L.C.; Crouse, J.R.; Leiter, L.A.; Linz, P.; Friedewald, W.T.; Buse, J.B.; Gerstein, H.C.; Probstfield, J.; et al. Effects of combination lipid therapy in type 2 diabetes mellitus. N Engl. J. Med. 2010, 362, 1563–1574. [Google Scholar] [CrossRef]
  31. Morieri, M.L.; Shah, H.S.; Sjaarda, J.; Lenzini, P.A.; Campbell, H.; Motsinger-Reif, A.A.; Gao, H.; Lovato, L.; Prudente, S.; Pandolfi, A.; et al. Polymorphism Influences the Cardiovascular Benefit of Fenofibrate in Type 2 Diabetes: Findings From ACCORD-Lipid. Diabetes 2020, 69, 771–783. [Google Scholar] [CrossRef] [PubMed]
  32. House, J.S.; Motsinger-Reif, A.A. Fibrate pharmacogenomics: Expanding past the genome. Pharmacogenomics 2020, 21, 293–306. [Google Scholar] [CrossRef] [PubMed]
  33. Frick, M.H.; Elo, O.; Haapa, K.; Heinonen, O.P.; Heinsalmi, P.; Helo, P.; Huttunen, J.K.; Kaitaniemi, P.; Koskinen, P.; Manninen, V. Helsinki Heart Study: Primary-prevention trial with gemfibrozil in middle-aged men with dyslipidemia. Safety of treatment, changes in risk factors, and incidence of coronary heart disease. N. Engl. J. Med. 1987, 317, 1237–1245. [Google Scholar] [CrossRef] [PubMed]
  34. Rubins, H.B.; Robins, S.J.; Collins, D.; Fye, C.L.; Anderson, J.W.; Elam, M.B.; Faas, F.H.; Linares, E.; Schaefer, E.J.; Schectman, G.; et al. Gemfibrozil for the secondary prevention of coronary heart disease in men with low levels of high-density lipoprotein cholesterol. Veterans Affairs High-Density Lipoprotein Cholesterol Intervention Trial Study Group. N. Engl. J. Med. 1999, 341, 410–418. [Google Scholar] [CrossRef] [PubMed]
  35. Rubins, H.B.; Robins, S.J.; Collins, D. The Veterans Affairs High-Density Lipoprotein Intervention Trial: Baseline characteristics of normocholesterolemic men with coronary artery disease and low levels of high-density lipoprotein cholesterol. Veterans Affairs Cooperative Studies Program High-Density Lipoprotein Intervention Trial Study Group. Am. J. Cardiol. 1996, 78, 572–575. [Google Scholar] [CrossRef]
  36. Pradhan, A.D.; Paynter, N.P.; Everett, B.M.; Glynn, R.J.; Amarenco, P.; Elam, M.; Ginsberg, H.; Hiatt, W.R.; Ishibashi, S.; Koenig, W.; et al. Rationale and design of the Pemafibrate to Reduce Cardiovascular Outcomes by Reducing Triglycerides in Patients with Diabetes (PROMINENT) study. Am. Heart J. 2018, 206, 80–93. [Google Scholar] [CrossRef]
  37. Rival, Y.; Benéteau, N.; Taillandier, T.; Pezet, M.; Dupont-Passelaigue, E.; Patoiseau, J.F.; Junquéro, D.; Colpaert, F.C.; Delhon, A. PPARalpha and PPARdelta activators inhibit cytokine-induced nuclear translocation of NF-kappaB and expression of VCAM-1 in EAhy926 endothelial cells. Eur. J. Pharmacol. 2002, 435, 143–151. [Google Scholar] [CrossRef]
  38. Fan, Y.; Wang, Y.; Tang, Z.; Zhang, H.; Qin, X.; Zhu, Y.; Guan, Y.; Wang, X.; Staels, B.; Chien, S.; et al. Suppression of pro-inflammatory adhesion molecules by PPAR-delta in human vascular endothelial cells. Arterioscler. Thromb. Vasc. Biol. 2008, 28, 315–321. [Google Scholar] [CrossRef]
  39. Piqueras, L.; Sanz, M.J.; Perretti, M.; Morcillo, E.; Norling, L.; Mitchell, J.A.; Li, Y.; Bishop-Bailey, D. Activation of PPARbeta/delta inhibits leukocyte recruitment, cell adhesion molecule expression, and chemokine release. J. Leukoc. Biol. 2009, 86, 115–122. [Google Scholar] [CrossRef]
  40. Moraes, L.A.; Piqueras, L.; Bishop-Bailey, D. Peroxisome proliferator-activated receptors and inflammation. Pharmacol. Ther. 2006, 110, 371–385. [Google Scholar] [CrossRef]
  41. Lee, C.H.; Chawla, A.; Urbiztondo, N.; Liao, D.; Boisvert, W.A.; Evans, R.M.; Curtiss, L.K. Transcriptional repression of atherogenic inflammation: Modulation by PPARdelta. Science 2003, 302, 453–457. [Google Scholar] [CrossRef] [PubMed]
  42. Kim, H.J.; Ham, S.A.; Kim, S.U.; Hwang, J.Y.; Kim, J.H.; Chang, K.C.; Yabe-Nishimura, C.; Seo, H.G. Transforming growth factor-beta1 is a molecular target for the peroxisome proliferator-activated receptor delta. Circ. Res. 2008, 102, 193–200. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Ali, F.; Ali, N.S.; Bauer, A.; Boyle, J.J.; Hamdulay, S.S.; Haskard, D.O.; Randi, A.M.; Mason, J.C. PPARdelta and PGC1alpha act cooperatively to induce haem oxygenase-1 and enhance vascular endothelial cell resistance to stress. Cardiovasc. Res. 2010, 85, 701–710. [Google Scholar] [CrossRef]
  44. Liou, J.Y.; Lee, S.; Ghelani, D.; Matijevic-Aleksic, N.; Wu, K.K. Protection of endothelial survival by peroxisome proliferator-activated receptor-delta mediated 14-3-3 upregulation. Arterioscler. Thromb. Vasc. Biol. 2006, 26, 1481–1487. [Google Scholar] [CrossRef] [Green Version]
  45. Piqueras, L.; Reynolds, A.R.; Hodivala-Dilke, K.M.; Alfranca, A.; Redondo, J.M.; Hatae, T.; Tanabe, T.; Warner, T.D.; Bishop-Bailey, D. Activation of PPARbeta/delta induces endothelial cell proliferation and angiogenesis. Arterioscler. Thromb. Vasc. Biol. 2007, 27, 63–69. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Bishop-Bailey, D. A Role for PPARbeta/delta in Ocular Angiogenesis. PPAR Res. 2008, 2008, 825970. [Google Scholar] [CrossRef] [Green Version]
  47. Gaudel, C.; Schwartz, C.; Giordano, C.; Abumrad, N.A.; Grimaldi, P.A. Pharmacological activation of PPARbeta promotes rapid and calcineurin-dependent fiber remodeling and angiogenesis in mouse skeletal muscle. Am. J. Physiol. Endocrinol. Metab. 2008, 295, E297–E304. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Wagner, N.; Jehl-Piétri, C.; Lopez, P.; Murdaca, J.; Giordano, C.; Schwartz, C.; Gounon, P.; Hatem, S.N.; Grimaldi, P.; Wagner, K.D. Peroxisome proliferator-activated receptor beta stimulation induces rapid cardiac growth and angiogenesis via direct activation of calcineurin. Cardiovasc. Res. 2009, 83, 61–71. [Google Scholar] [CrossRef] [Green Version]
  49. Wagner, K.D.; Vukolic, A.; Baudouy, D.; Michiels, J.F.; Wagner, N. Inducible Conditional Vascular-Specific Overexpression of Peroxisome Proliferator-Activated Receptor Beta/Delta Leads to Rapid Cardiac Hypertrophy. PPAR Res. 2016, 2016, 7631085. [Google Scholar] [CrossRef] [Green Version]
  50. Wagner, K.-D.; Du, S.; Martin, L.; Leccia, N.; Michiels, J.-F.; Wagner, N. Vascular PPARβ/δ Promotes Tumor Angiogenesis and Progression. Cells 2019, 8, 1623. [Google Scholar] [CrossRef] [Green Version]
  51. Abdollahi, A.; Schwager, C.; Kleeff, J.; Esposito, I.; Domhan, S.; Peschke, P.; Hauser, K.; Hahnfeldt, P.; Hlatky, L.; Debus, J.; et al. Transcriptional network governing the angiogenic switch in human pancreatic cancer. Proc. Natl. Acad. Sci. USA 2007, 104, 12890–12895. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Müller-Brüsselbach, S.; Kömhoff, M.; Rieck, M.; Meissner, W.; Kaddatz, K.; Adamkiewicz, J.; Keil, B.; Klose, K.J.; Moll, R.; Burdick, A.D.; et al. Deregulation of tumor angiogenesis and blockade of tumor growth in PPARbeta-deficient mice. EMBO J. 2007, 26, 3686–3698. [Google Scholar] [CrossRef] [Green Version]
  53. Bohman, S.; Matsumoto, T.; Suh, K.; Dimberg, A.; Jakobsson, L.; Yuspa, S.; Claesson-Welsh, L. Proteomic analysis of vascular endothelial growth factor-induced endothelial cell differentiation reveals a role for chloride intracellular channel 4 (CLIC4) in tubular morphogenesis. J. Biol. Chem. 2005, 280, 42397–42404. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Gilde, A.J.; van der Lee, K.A.; Willemsen, P.H.; Chinetti, G.; van der Leij, F.R.; van der Vusse, G.J.; Staels, B.; van Bilsen, M. Peroxisome proliferator-activated receptor (PPAR) alpha and PPARbeta/delta, but not PPARgamma, modulate the expression of genes involved in cardiac lipid metabolism. Circ. Res. 2003, 92, 518–524. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Magadum, A.; Ding, Y.; He, L.; Kim, T.; Vasudevarao, M.D.; Long, Q.; Yang, K.; Wickramasinghe, N.; Renikunta, H.V.; Dubois, N.; et al. Live cell screening platform identifies PPARδ as a regulator of cardiomyocyte proliferation and cardiac repair. Cell Res. 2017, 27, 1002–1019. [Google Scholar] [CrossRef] [Green Version]
  56. Jucker, B.M.; Doe, C.P.; Schnackenberg, C.G.; Olzinski, A.R.; Maniscalco, K.; Williams, C.; Hu, T.C.; Lenhard, S.C.; Costell, M.; Bernard, R.; et al. PPARdelta activation normalizes cardiac substrate metabolism and reduces right ventricular hypertrophy in congestive heart failure. J. Cardiovasc. Pharmacol. 2007, 50, 25–34. [Google Scholar] [CrossRef]
  57. Li, J.; Xuan, W.; Yan, R.; Tropak, M.B.; Jean-St-Michel, E.; Liang, W.; Gladstone, R.; Backx, P.H.; Kharbanda, R.K.; Redington, A.N. Remote preconditioning provides potent cardioprotection via PI3K/Akt activation and is associated with nuclear accumulation of β-catenin. Clin. Sci. 2011, 120, 451–462. [Google Scholar] [CrossRef] [Green Version]
  58. Park, J.R.; Ahn, J.H.; Jung, M.H.; Koh, J.S.; Park, Y.; Hwang, S.J.; Jeong, Y.H.; Kwak, C.H.; Lee, Y.S.; Seo, H.G.; et al. Effects of Peroxisome Proliferator-Activated Receptor-δ Agonist on Cardiac Healing after Myocardial Infarction. PLoS ONE 2016, 11, e0148510. [Google Scholar] [CrossRef] [Green Version]
  59. Mikami, D.; Kimura, H.; Kamiyama, K.; Torii, K.; Kasuno, K.; Takahashi, N.; Yoshida, H.; Iwano, M. Telmisartan activates endogenous peroxisome proliferator-activated receptor-δ and may have anti-fibrotic effects in human mesangial cells. Hypertens. Res. 2014, 37, 422–431. [Google Scholar] [CrossRef] [Green Version]
  60. Amano, Y.; Yamaguchi, T.; Ohno, K.; Niimi, T.; Orita, M.; Sakashita, H.; Takeuchi, M. Structural basis for telmisartan-mediated partial activation of PPAR gamma. Hypertens. Res. 2012, 35, 715–719. [Google Scholar] [CrossRef] [Green Version]
  61. Benson, S.C.; Pershadsingh, H.A.; Ho, C.I.; Chittiboyina, A.; Desai, P.; Pravenec, M.; Qi, N.; Wang, J.; Avery, M.A.; Kurtz, T.W. Identification of telmisartan as a unique angiotensin II receptor antagonist with selective PPARgamma-modulating activity. Hypertension 2004, 43, 993–1002. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Kappert, K.; Böhm, M.; Schmieder, R.; Schumacher, H.; Teo, K.; Yusuf, S.; Sleight, P.; Unger, T.; Investigators, O.T. Impact of sex on cardiovascular outcome in patients at high cardiovascular risk: Analysis of the Telmisartan Randomized Assessment Study in ACE-Intolerant Subjects With Cardiovascular Disease (TRANSCEND) and the Ongoing Telmisartan Alone and in Combination With Ramipril Global End Point Trial (ONTARGET). Circulation 2012, 126, 934–941. [Google Scholar] [CrossRef] [PubMed]
  63. Chen, N.G.; Han, X. Dual function of troglitazone in ICAM-1 gene expression in human vascular endothelium. Biochem. Biophys. Res. Commun. 2001, 282, 717–722. [Google Scholar] [CrossRef]
  64. Marx, N.; Schönbeck, U.; Lazar, M.A.; Libby, P.; Plutzky, J. Peroxisome proliferator-activated receptor gamma activators inhibit gene expression and migration in human vascular smooth muscle cells. Circ. Res. 1998, 83, 1097–1103. [Google Scholar] [CrossRef] [Green Version]
  65. Murao, K.; Imachi, H.; Momoi, A.; Sayo, Y.; Hosokawa, H.; Sato, M.; Ishida, T.; Takahara, J. Thiazolidinedione inhibits the production of monocyte chemoattractant protein-1 in cytokine-treated human vascular endothelial cells. FEBS Lett. 1999, 454, 27–30. [Google Scholar] [CrossRef] [PubMed]
  66. Marx, N.; Mach, F.; Sauty, A.; Leung, J.H.; Sarafi, M.N.; Ransohoff, R.M.; Libby, P.; Plutzky, J.; Luster, A.D. Peroxisome proliferator-activated receptor-gamma activators inhibit IFN-gamma-induced expression of the T cell-active CXC chemokines IP-10, Mig, and I-TAC in human endothelial cells. J. Immunol. 2000, 164, 6503–6508. [Google Scholar] [CrossRef] [Green Version]
  67. Lee, H.; Shi, W.; Tontonoz, P.; Wang, S.; Subbanagounder, G.; Hedrick, C.C.; Hama, S.; Borromeo, C.; Evans, R.M.; Berliner, J.A.; et al. Role for peroxisome proliferator-activated receptor alpha in oxidized phospholipid-induced synthesis of monocyte chemotactic protein-1 and interleukin-8 by endothelial cells. Circ. Res. 2000, 87, 516–521. [Google Scholar] [CrossRef] [Green Version]
  68. Ricote, M.; Li, A.C.; Willson, T.M.; Kelly, C.J.; Glass, C.K. The peroxisome proliferator-activated receptor-gamma is a negative regulator of macrophage activation. Nature 1998, 391, 79–82. [Google Scholar] [CrossRef]
  69. Xin, X.; Yang, S.; Kowalski, J.; Gerritsen, M.E. Peroxisome proliferator-activated receptor gamma ligands are potent inhibitors of angiogenesis in vitro and in vivo. J. Biol. Chem. 1999, 274, 9116–9121. [Google Scholar] [CrossRef] [Green Version]
  70. Peeters, L.L.; Vigne, J.L.; Tee, M.K.; Zhao, D.; Waite, L.L.; Taylor, R.N. PPAR gamma represses VEGF expression in human endometrial cells: Implications for uterine angiogenesis. Angiogenesis 2005, 8, 373–379. [Google Scholar] [CrossRef]
  71. Sassa, Y.; Hata, Y.; Aiello, L.P.; Taniguchi, Y.; Kohno, K.; Ishibashi, T. Bifunctional properties of peroxisome proliferator-activated receptor gamma1 in KDR gene regulation mediated via interaction with both Sp1 and Sp3. Diabetes 2004, 53, 1222–1229. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Kim, K.Y.; Cheon, H.G. Antiangiogenic effect of rosiglitazone is mediated via peroxisome proliferator-activated receptor gamma-activated maxi-K channel opening in human umbilical vein endothelial cells. J. Biol. Chem. 2006, 281, 13503–13512. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Kiec-Wilk, B.; Grzybowska-Galuszka, J.; Polus, A.; Pryjma, J.; Knapp, A.; Kristiansen, K. The MAPK-dependent regulation of the Jagged/Notch gene expression by VEGF, bFGF or PPAR gamma mediated angiogenesis in HUVEC. J. Physiol. Pharmacol. 2010, 61, 217–225. [Google Scholar] [PubMed]
  74. Scoditti, E.; Massaro, M.; Carluccio, M.A.; Distante, A.; Storelli, C.; De Caterina, R. PPARgamma agonists inhibit angiogenesis by suppressing PKCalpha- and CREB-mediated COX-2 expression in the human endothelium. Cardiovasc. Res. 2010, 86, 302–310. [Google Scholar] [CrossRef] [PubMed]
  75. Biscetti, F.; Pecorini, G.; Arena, V.; Stigliano, E.; Angelini, F.; Ghirlanda, G.; Ferraccioli, G.; Flex, A. Cilostazol improves the response to ischemia in diabetic mice by a mechanism dependent on PPARγ. Mol. Cell. Endocrinol. 2013, 381, 80–87. [Google Scholar] [CrossRef]
  76. Huang, P.H.; Sata, M.; Nishimatsu, H.; Sumi, M.; Hirata, Y.; Nagai, R. Pioglitazone ameliorates endothelial dysfunction and restores ischemia-induced angiogenesis in diabetic mice. Biomed. Pharmacother. 2008, 62, 46–52. [Google Scholar] [CrossRef]
  77. Chu, K.; Lee, S.T.; Koo, J.S.; Jung, K.H.; Kim, E.H.; Sinn, D.I.; Kim, J.M.; Ko, S.Y.; Kim, S.J.; Song, E.C.; et al. Peroxisome proliferator-activated receptor-gamma-agonist, rosiglitazone, promotes angiogenesis after focal cerebral ischemia. Brain Res. 2006, 1093, 208–218. [Google Scholar] [CrossRef] [PubMed]
  78. Duan, S.Z.; Ivashchenko, C.Y.; Russell, M.W.; Milstone, D.S.; Mortensen, R.M. Cardiomyocyte-specific knockout and agonist of peroxisome proliferator-activated receptor-gamma both induce cardiac hypertrophy in mice. Circ. Res. 2005, 97, 372–379. [Google Scholar] [CrossRef] [Green Version]
  79. Son, N.H.; Park, T.S.; Yamashita, H.; Yokoyama, M.; Huggins, L.A.; Okajima, K.; Homma, S.; Szabolcs, M.J.; Huang, L.S.; Goldberg, I.J. Cardiomyocyte expression of PPARgamma leads to cardiac dysfunction in mice. J. Clin. Investig. 2007, 117, 2791–2801. [Google Scholar] [CrossRef] [Green Version]
  80. Shen, Z.X.; Yang, Q.Z.; Li, C.; Du, L.J.; Sun, X.N.; Liu, Y.; Sun, J.Y.; Gu, H.H.; Sun, Y.M.; Wang, J.; et al. Myeloid peroxisome proliferator-activated receptor gamma deficiency aggravates myocardial infarction in mice. Atherosclerosis 2018, 274, 199–205. [Google Scholar] [CrossRef]
  81. Choo, E.H.; Han, E.J.; Kim, C.J.; Kim, S.H.; O, J.H.; Chang, K.; Seung, K.B. Effect of Pioglitazone in Combination with Moderate Dose Statin on Atherosclerotic Inflammation: Randomized Controlled Clinical Trial Using Serial FDG-PET/CT. Korean Circ. J. 2018, 48, 591–601. [Google Scholar] [CrossRef] [PubMed]
  82. Skochko, O.V.; Kaidashev, I.P. Effect of pioglitazone on insulin resistance, progression of atherosclerosis and clinical course of coronary heart disease. Wiad. Lek. 2017, 70, 881–890. [Google Scholar]
  83. Forst, T.; Hohberg, C.; Fuellert, S.D.; Lübben, G.; Konrad, T.; Löbig, M.; Weber, M.M.; Sachara, C.; Gottschall, V.; Pfützner, A. Pharmacological PPARgamma stimulation in contrast to beta cell stimulation results in an improvement in adiponectin and proinsulin intact levels and reduces intima media thickness in patients with type 2 diabetes. Horm. Metab. Res. 2005, 37, 521–527. [Google Scholar] [CrossRef] [PubMed]
  84. Ryan, K.E.; McCance, D.R.; Powell, L.; McMahon, R.; Trimble, E.R. Fenofibrate and pioglitazone improve endothelial function and reduce arterial stiffness in obese glucose tolerant men. Atherosclerosis 2007, 194, e123–e130. [Google Scholar] [CrossRef] [Green Version]
  85. Dormandy, J.; Bhattacharya, M.; van Troostenburg de Bruyn, A.R.; PROactive Investigators. Safety and tolerability of pioglitazone in high-risk patients with type 2 diabetes: An overview of data from PROactive. Drug Saf. 2009, 32, 187–202. [Google Scholar] [CrossRef]
  86. Dormandy, J.A.; Charbonnel, B.; Eckland, D.J.; Erdmann, E.; Massi-Benedetti, M.; Moules, I.K.; Skene, A.M.; Tan, M.H.; Lefèbvre, P.J.; Murray, G.D.; et al. Secondary prevention of macrovascular events in patients with type 2 diabetes in the PROactive Study (PROspective pioglitAzone Clinical Trial In macroVascular Events): A randomised controlled trial. Lancet 2005, 366, 1279–1289. [Google Scholar] [CrossRef] [PubMed]
  87. Erdmann, E.; Charbonnel, B.; Wilcox, R.G.; Skene, A.M.; Massi-Benedetti, M.; Yates, J.; Tan, M.; Spanheimer, R.; Standl, E.; Dormandy, J.A.; et al. Pioglitazone use and heart failure in patients with type 2 diabetes and preexisting cardiovascular disease: Data from the PROactive study (PROactive 08). Diabetes Care 2007, 30, 2773–2778. [Google Scholar] [CrossRef] [Green Version]
  88. Nissen, S.E.; Nicholls, S.J.; Wolski, K.; Nesto, R.; Kupfer, S.; Perez, A.; Jure, H.; De Larochellière, R.; Staniloae, C.S.; Mavromatis, K.; et al. Comparison of pioglitazone vs glimepiride on progression of coronary atherosclerosis in patients with type 2 diabetes: The PERISCOPE randomized controlled trial. JAMA 2008, 299, 1561–1573. [Google Scholar] [CrossRef] [Green Version]
  89. Kernan, W.N.; Viscoli, C.M.; Furie, K.L.; Young, L.H.; Inzucchi, S.E.; Gorman, M.; Guarino, P.D.; Lovejoy, A.M.; Peduzzi, P.N.; Conwit, R.; et al. Pioglitazone after Ischemic Stroke or Transient Ischemic Attack. N. Engl. J. Med. 2016, 374, 1321–1331. [Google Scholar] [CrossRef]
  90. Hampp, C.; Pippins, J. Pioglitazone and bladder cancer: FDA’s assessment. Pharmacoepidemiol. Drug Saf. 2017, 26, 117–118. [Google Scholar] [CrossRef]
  91. Gerstein, H.C.; Yusuf, S.; Bosch, J.; Pogue, J.; Sheridan, P.; Dinccag, N.; Hanefeld, M.; Hoogwerf, B.; Laakso, M.; Mohan, V.; et al. Effect of rosiglitazone on the frequency of diabetes in patients with impaired glucose tolerance or impaired fasting glucose: A randomised controlled trial. Lancet 2006, 368, 1096–1105. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Kahn, S.E.; Haffner, S.M.; Heise, M.A.; Herman, W.H.; Holman, R.R.; Jones, N.P.; Kravitz, B.G.; Lachin, J.M.; O’Neill, M.C.; Zinman, B.; et al. Glycemic durability of rosiglitazone, metformin, or glyburide monotherapy. N. Engl. J. Med. 2006, 355, 2427–2443. [Google Scholar] [CrossRef] [Green Version]
  93. Nissen, S.E.; Wolski, K. Effect of rosiglitazone on the risk of myocardial infarction and death from cardiovascular causes. N. Engl. J. Med. 2007, 356, 2457–2471. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Home, P.D.; Pocock, S.J.; Beck-Nielsen, H.; Curtis, P.S.; Gomis, R.; Hanefeld, M.; Jones, N.P.; Komajda, M.; McMurray, J.J.; Team, R.S. Rosiglitazone evaluated for cardiovascular outcomes in oral agent combination therapy for type 2 diabetes (RECORD): A multicentre, randomised, open-label trial. Lancet 2009, 373, 2125–2135. [Google Scholar] [CrossRef] [PubMed]
  95. Rosen, C.J. Revisiting the rosiglitazone story--lessons learned. N. Engl. J. Med. 2010, 363, 803–806. [Google Scholar] [CrossRef] [PubMed]
  96. Doney, A.S.; Fischer, B.; Leese, G.; Morris, A.D.; Palmer, C.N. Cardiovascular risk in type 2 diabetes is associated with variation at the PPARG locus: A Go-DARTS study. Arterioscler. Thromb. Vasc. Biol. 2004, 24, 2403–2407. [Google Scholar] [CrossRef] [Green Version]
  97. Gallicchio, L.; Kalesan, B.; Huang, H.Y.; Strickland, P.; Hoffman, S.C.; Helzlsouer, K.J. Genetic polymorphisms of peroxisome proliferator-activated receptors and the risk of cardiovascular morbidity and mortality in a community-based cohort in washington county, Maryland. PPAR Res. 2008, 2008, 276581. [Google Scholar] [CrossRef] [Green Version]
  98. Tenenbaum, A.; Motro, M.; Fisman, E.Z. Dual and pan-peroxisome proliferator-activated receptors (PPAR) co-agonism: The bezafibrate lessons. Cardiovasc. Diabetol. 2005, 4, 14. [Google Scholar] [CrossRef] [Green Version]
  99. Elkeles, R.S.; Diamond, J.R.; Poulter, C.; Dhanjil, S.; Nicolaides, A.N.; Mahmood, S.; Richmond, W.; Mather, H.; Sharp, P.; Feher, M.D. Cardiovascular outcomes in type 2 diabetes. A double-blind placebo-controlled study of bezafibrate: The St. Mary’s, Ealing, Northwick Park Diabetes Cardiovascular Disease Prevention (SENDCAP) Study. Diabetes Care 1998, 21, 641–648. [Google Scholar] [CrossRef]
  100. The BIP Study Group. Secondary prevention by raising HDL cholesterol and reducing triglycerides in patients with coronary artery disease. Circulation 2000, 102, 21–27. [Google Scholar] [CrossRef]
  101. Nissen, S.E.; Wolski, K.; Topol, E.J. Effect of muraglitazar on death and major adverse cardiovascular events in patients with type 2 diabetes mellitus. JAMA 2005, 294, 2581–2586. [Google Scholar] [CrossRef] [Green Version]
  102. Oleksiewicz, M.B.; Thorup, I.; Nielsen, H.S.; Andersen, H.V.; Hegelund, A.C.; Iversen, L.; Guldberg, T.S.; Brinck, P.R.; Sjogren, I.; Thinggaard, U.K.; et al. Generalized cellular hypertrophy is induced by a dual-acting PPAR agonist in rat urinary bladder urothelium in vivo. Toxicol Pathol 2005, 33, 552–560. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Ratner, R.E.; Parikh, S.; Tou, C.; Group, G.S. Efficacy, safety and tolerability of tesaglitazar when added to the therapeutic regimen of poorly controlled insulin-treated patients with type 2 diabetes. Diab. Vasc. Dis. Res. 2007, 4, 214–221. [Google Scholar] [CrossRef]
  104. Henry, R.R.; Lincoff, A.M.; Mudaliar, S.; Rabbia, M.; Chognot, C.; Herz, M. Effect of the dual peroxisome proliferator-activated receptor-alpha/gamma agonist aleglitazar on risk of cardiovascular disease in patients with type 2 diabetes (SYNCHRONY): A phase II, randomised, dose-ranging study. Lancet 2009, 374, 126–135. [Google Scholar] [CrossRef] [PubMed]
  105. Lincoff, A.M.; Tardif, J.C.; Schwartz, G.G.; Nicholls, S.J.; Rydén, L.; Neal, B.; Malmberg, K.; Wedel, H.; Buse, J.B.; Henry, R.R.; et al. Effect of aleglitazar on cardiovascular outcomes after acute coronary syndrome in patients with type 2 diabetes mellitus: The AleCardio randomized clinical trial. JAMA 2014, 311, 1515–1525. [Google Scholar] [CrossRef] [PubMed]
  106. Erdmann, E.; Califf, R.; Gerstein, H.C.; Malmberg, K.; Ruilope, L.; Schwartz, G.G.; Wedel, H.; Volz, D.; Ditmarsch, M.; Svensson, A.; et al. Effects of the dual peroxisome proliferator-activated receptor activator aleglitazar in patients with Type 2 Diabetes mellitus or prediabetes. Am. Heart J. 2015, 170, 117–122. [Google Scholar] [CrossRef]
  107. Angiolillo, D.J.; Capodanno, D. Aspirin for Primary Prevention of Cardiovascular Disease in the 21. Am. J. Cardiol. 2021, 144 (Suppl. 1), S15–S22. [Google Scholar] [CrossRef]
  108. Kolawole, O.R.; Kashfi, K. NSAIDs and Cancer Resolution: New Paradigms beyond Cyclooxygenase. Int. J. Mol. Sci. 2022, 23, 1432. [Google Scholar] [CrossRef]
  109. Patel, D.; Roy, A.; Kundu, M.; Jana, M.; Luan, C.H.; Gonzalez, F.J.; Pahan, K. Aspirin binds to PPARα to stimulate hippocampal plasticity and protect memory. Proc. Natl. Acad. Sci. USA 2018, 115, E7408–E7417. [Google Scholar] [CrossRef] [Green Version]
  110. Patel, D.; Roy, A.; Pahan, K. PPARα serves as a new receptor of aspirin for neuroprotection. J. Neurosci. Res. 2020, 98, 626–631. [Google Scholar] [CrossRef] [PubMed]
  111. Chaldakov, G. The metabotrophic NGF and BDNF: An emerging concept. Arch. Ital. Biol. 2011, 149, 257–263. [Google Scholar] [CrossRef]
  112. Hoefer, I.E.; Grundmann, S.; Schirmer, S.; van Royen, N.; Meder, B.; Bode, C.; Piek, J.J.; Buschmann, I.R. Aspirin, but not clopidogrel, reduces collateral conductance in a rabbit model of femoral artery occlusion. J. Am. Coll. Cardiol. 2005, 46, 994–1001. [Google Scholar] [CrossRef] [Green Version]
  113. Borthwick, G.M.; Johnson, A.S.; Partington, M.; Burn, J.; Wilson, R.; Arthur, H.M. Therapeutic levels of aspirin and salicylate directly inhibit a model of angiogenesis through a Cox-independent mechanism. FASEB J. 2006, 20, 2009–2016. [Google Scholar] [CrossRef]
  114. Rauzi, F.; Kirkby, N.S.; Edin, M.L.; Whiteford, J.; Zeldin, D.C.; Mitchell, J.A.; Warner, T.D. Aspirin inhibits the production of proangiogenic 15(S)-HETE by platelet cyclooxygenase-1. FASEB J. 2016, 30, 4256–4266. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Wu, L.; Wang, W.; Dai, M.; Li, H.; Chen, C.; Wang, D. PPARα ligand, AVE8134, and cyclooxygenase inhibitor therapy synergistically suppress lung cancer growth and metastasis. BMC Cancer 2019, 19, 1166. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Gerrah, R.; Fogel, M.; Gilon, D. Aspirin decreases vascular endothelial growth factor release during myocardial ischemia. Int. J. Cardiol. 2004, 94, 25–29. [Google Scholar] [CrossRef]
  117. Endo, A.; Kuroda, M.; Tanzawa, K. Competitive inhibition of 3-hydroxy-3-methylglutaryl coenzyme A reductase by ML-236A and ML-236B fungal metabolites, having hypocholesterolemic activity. FEBS Lett. 1976, 72, 323–326. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Nowak, M.M.; Niemczyk, M.; Florczyk, M.; Kurzyna, M.; Pączek, L. Effect of Statins on All-Cause Mortality in Adults: A Systematic Review and Meta-Analysis of Propensity Score-Matched Studies. J. Clin. Med. 2022, 11, 5643. [Google Scholar] [CrossRef] [PubMed]
  119. Yano, M.; Matsumura, T.; Senokuchi, T.; Ishii, N.; Murata, Y.; Taketa, K.; Motoshima, H.; Taguchi, T.; Sonoda, K.; Kukidome, D.; et al. Statins activate peroxisome proliferator-activated receptor gamma through extracellular signal-regulated kinase 1/2 and p38 mitogen-activated protein kinase-dependent cyclooxygenase-2 expression in macrophages. Circ. Res. 2007, 100, 1442–1451. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Paumelle, R.; Blanquart, C.; Briand, O.; Barbier, O.; Duhem, C.; Woerly, G.; Percevault, F.; Fruchart, J.C.; Dombrowicz, D.; Glineur, C.; et al. Acute antiinflammatory properties of statins involve peroxisome proliferator-activated receptor-alpha via inhibition of the protein kinase C signaling pathway. Circ. Res. 2006, 98, 361–369. [Google Scholar] [CrossRef]
  121. Inoue, I.; Goto, S.; Mizotani, K.; Awata, T.; Mastunaga, T.; Kawai, S.; Nakajima, T.; Hokari, S.; Komoda, T.; Katayama, S. Lipophilic HMG-CoA reductase inhibitor has an anti-inflammatory effect: Reduction of MRNA levels for interleukin-1beta, interleukin-6, cyclooxygenase-2, and p22phox by regulation of peroxisome proliferator-activated receptor alpha (PPARalpha) in primary endothelial cells. Life Sci. 2000, 67, 863–876. [Google Scholar] [CrossRef] [PubMed]
  122. Martin, G.; Duez, H.; Blanquart, C.; Berezowski, V.; Poulain, P.; Fruchart, J.C.; Najib-Fruchart, J.; Glineur, C.; Staels, B. Statin-induced inhibition of the Rho-signaling pathway activates PPARalpha and induces HDL apoA-I. J. Clin. Investig. 2001, 107, 1423–1432. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Ye, P.; Sheng, L.; Zhang, C.; Liu, Y. Atorvastatin attenuating down-regulation of peroxisome proliferator-activated receptor gamma in preventing cardiac hypertrophy of rats in vitro and in vivo. J. Pharm. Pharm. Sci. 2006, 9, 365–375. [Google Scholar]
  124. Qin, Y.W.; Ye, P.; He, J.Q.; Sheng, L.; Wang, L.Y.; Du, J. Simvastatin inhibited cardiac hypertrophy and fibrosis in apolipoprotein E-deficient mice fed a “Western-style diet” by increasing PPAR α and γ expression and reducing TC, MMP-9, and Cat S levels. Acta Pharmacol. Sin. 2010, 31, 1350–1358. [Google Scholar] [CrossRef] [PubMed]
  125. Shen, Y.; Wu, H.; Wang, C.; Shao, H.; Huang, H.; Jing, H.; Li, D. Simvastatin attenuates cardiopulmonary bypass-induced myocardial inflammatory injury in rats by activating peroxisome proliferator-activated receptor γ. Eur. J. Pharmacol. 2010, 649, 255–262. [Google Scholar] [CrossRef] [PubMed]
  126. Desjardins, F.; Sekkali, B.; Verreth, W.; Pelat, M.; De Keyzer, D.; Mertens, A.; Smith, G.; Herregods, M.C.; Holvoet, P.; Balligand, J.L. Rosuvastatin increases vascular endothelial PPARgamma expression and corrects blood pressure variability in obese dyslipidaemic mice. Eur. Heart J. 2008, 29, 128–137. [Google Scholar] [CrossRef] [PubMed]
  127. Ali, F.Y.; Armstrong, P.C.; Dhanji, A.R.; Tucker, A.T.; Paul-Clark, M.J.; Mitchell, J.A.; Warner, T.D. Antiplatelet actions of statins and fibrates are mediated by PPARs. Arterioscler. Thromb. Vasc. Biol. 2009, 29, 706–711. [Google Scholar] [CrossRef] [Green Version]
  128. Weis, M.; Heeschen, C.; Glassford, A.J.; Cooke, J.P. Statins have biphasic effects on angiogenesis. Circulation 2002, 105, 739–745. [Google Scholar] [CrossRef] [Green Version]
  129. Dulak, J.; Loboda, A.; Jazwa, A.; Zagorska, A.; Dörler, J.; Alber, H.; Dichtl, W.; Weidinger, F.; Frick, M.; Jozkowicz, A. Atorvastatin affects several angiogenic mediators in human endothelial cells. Endothelium 2005, 12, 233–241. [Google Scholar] [CrossRef] [Green Version]
  130. Urbich, C.; Dernbach, E.; Zeiher, A.M.; Dimmeler, S. Double-edged role of statins in angiogenesis signaling. Circ. Res. 2002, 90, 737–744. [Google Scholar] [CrossRef] [Green Version]
  131. Matsumura, M.; Fukuda, N.; Kobayashi, N.; Umezawa, H.; Takasaka, A.; Matsumoto, T.; Yao, E.H.; Ueno, T.; Negishi, N. Effects of atorvastatin on angiogenesis in hindlimb ischemia and endothelial progenitor cell formation in rats. J. Atheroscler. Thromb. 2009, 16, 319–326. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Zuo, X.; Xu, W.; Xu, M.; Tian, R.; Moussalli, M.J.; Mao, F.; Zheng, X.; Wang, J.; Morris, J.S.; Gagea, M.; et al. Metastasis regulation by PPARD expression in cancer cells. JCI Insight 2017, 2, e91419. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Inoue, I.; Itoh, F.; Aoyagi, S.; Tazawa, S.; Kusama, H.; Akahane, M.; Mastunaga, T.; Hayashi, K.; Awata, T.; Komoda, T.; et al. Fibrate and statin synergistically increase the transcriptional activities of PPARalpha/RXRalpha and decrease the transactivation of NFkappaB. Biochem. Biophys. Res. Commun. 2002, 290, 131–139. [Google Scholar] [CrossRef] [PubMed]
  134. Tham, D.M.; Martin-McNulty, B.; Wang, Y.X.; Wilson, D.W.; Vergona, R.; Sullivan, M.E.; Dole, W.; Rutledge, J.C. Angiotensin II is associated with activation of NF-kappaB-mediated genes and downregulation of PPARs. Physiol. Genom. 2002, 11, 21–30. [Google Scholar] [CrossRef]
  135. Da Cunha, V.; Tham, D.M.; Martin-McNulty, B.; Deng, G.; Ho, J.J.; Wilson, D.W.; Rutledge, J.C.; Vergona, R.; Sullivan, M.E.; Wang, Y.X. Enalapril attenuates angiotensin II-induced atherosclerosis and vascular inflammation. Atherosclerosis 2005, 178, 9–17. [Google Scholar] [CrossRef]
  136. Storka, A.; Vojtassakova, E.; Mueller, M.; Kapiotis, S.; Haider, D.G.; Jungbauer, A.; Wolzt, M. Angiotensin inhibition stimulates PPARgamma and the release of visfatin. Eur. J. Clin. Investig. 2008, 38, 820–826. [Google Scholar] [CrossRef]
  137. Efrati, S.; Berman, S.; Ilgiyeav, E.; Averbukh, Z.; Weissgarten, J. PPAR-gamma activation inhibits angiotensin II synthesis, apoptosis, and proliferation of mesangial cells from spontaneously hypertensive rats. Nephron. Exp. Nephrol. 2007, 106, e107–e112. [Google Scholar] [CrossRef]
  138. Cernecka, H.; Doka, G.; Srankova, J.; Pivackova, L.; Malikova, E.; Galkova, K.; Kyselovic, J.; Krenek, P.; Klimas, J. Ramipril restores PPARβ/δ and PPARγ expressions and reduces cardiac NADPH oxidase but fails to restore cardiac function and accompanied myosin heavy chain ratio shift in severe anthracycline-induced cardiomyopathy in rat. Eur. J. Pharmacol. 2016, 791, 244–253. [Google Scholar] [CrossRef]
  139. Tamarat, R.; Silvestre, J.S.; Kubis, N.; Benessiano, J.; Duriez, M.; deGasparo, M.; Henrion, D.; Levy, B.I. Endothelial nitric oxide synthase lies downstream from angiotensin II-induced angiogenesis in ischemic hindlimb. Hypertension 2002, 39, 830–835. [Google Scholar] [CrossRef] [Green Version]
  140. Jesmin, S.; Hattori, Y.; Sakuma, I.; Mowa, C.N.; Kitabatake, A. Role of ANG II in coronary capillary angiogenesis at the insulin-resistant stage of a NIDDM rat model. Am. J. Physiol. Heart. Circ. Physiol. 2002, 283, H1387–H1397. [Google Scholar] [CrossRef] [Green Version]
  141. Walther, T.; Menrad, A.; Orzechowski, H.D.; Siemeister, G.; Paul, M.; Schirner, M. Differential regulation of in vivo angiogenesis by angiotensin II receptors. FASEB J. 2003, 17, 2061–2067. [Google Scholar] [CrossRef] [PubMed]
  142. Couture, R.; Blaes, N.; Girolami, J.P. Kinin receptors in vascular biology and pathology. Curr. Vasc. Pharmacol. 2014, 12, 223–248. [Google Scholar] [CrossRef]
  143. Fabre, J.E.; Rivard, A.; Magner, M.; Silver, M.; Isner, J.M. Tissue inhibition of angiotensin-converting enzyme activity stimulates angiogenesis in vivo. Circulation 1999, 99, 3043–3049. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Silvestre, J.S.; Bergaya, S.; Tamarat, R.; Duriez, M.; Boulanger, C.M.; Levy, B.I. Proangiogenic effect of angiotensin-converting enzyme inhibition is mediated by the bradykinin B(2) receptor pathway. Circ. Res. 2001, 89, 678–683. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Li, P.; Kondo, T.; Numaguchi, Y.; Kobayashi, K.; Aoki, M.; Inoue, N.; Okumura, K.; Murohara, T. Role of bradykinin, nitric oxide, and angiotensin II type 2 receptor in imidapril-induced angiogenesis. Hypertension 2008, 51, 252–258. [Google Scholar] [CrossRef] [Green Version]
  146. Unger, T.; Mattfeldt, T.; Lamberty, V.; Bock, P.; Mall, G.; Linz, W.; Schölkens, B.A.; Gohlke, P. Effect of early onset angiotensin converting enzyme inhibition on myocardial capillaries. Hypertension 1992, 20, 478–482. [Google Scholar] [CrossRef] [Green Version]
  147. Sanchez de Miguel, L.; Neysari, S.; Jakob, S.; Petrimpol, M.; Butz, N.; Banfi, A.; Zaugg, C.E.; Humar, R.; Battegay, E.J. B2-kinin receptor plays a key role in B1-, angiotensin converting enzyme inhibitor-, and vascular endothelial growth factor-stimulated in vitro angiogenesis in the hypoxic mouse heart. Cardiovasc. Res. 2008, 80, 106–113. [Google Scholar] [CrossRef] [Green Version]
  148. Hiller, K.H.; Ruile, P.; Kraus, G.; Bauer, W.R.; Waller, C. Tissue ACE inhibition improves microcirculation in remote myocardium after coronary stenosis: MR imaging study in rats. Microvasc. Res. 2010, 80, 484–490. [Google Scholar] [CrossRef]
  149. Ziada, A.M.; Hassan, M.O.; Tahlilkar, K.I.; Inuwa, I.M. Long-term exercise training and angiotensin-converting enzyme inhibition differentially enhance myocardial capillarization in the spontaneously hypertensive rat. J. Hypertens. 2005, 23, 1233–1240. [Google Scholar] [CrossRef]
  150. Ebrahimian, T.G.; Tamarat, R.; Clergue, M.; Duriez, M.; Levy, B.I.; Silvestre, J.S. Dual effect of angiotensin-converting enzyme inhibition on angiogenesis in type 1 diabetic mice. Arterioscler. Thromb. Vasc. Biol. 2005, 25, 65–70. [Google Scholar] [CrossRef] [Green Version]
  151. Sharma, A.; Bettis, D.I.; Cowden, J.W.; Mohan, R.R. Localization of angiotensin converting enzyme in rabbit cornea and its role in controlling corneal angiogenesis in vivo. Mol. Vis. 2010, 16, 720–728. [Google Scholar] [PubMed]
  152. Amaral, S.L.; Papanek, P.E.; Greene, A.S. Angiotensin II and VEGF are involved in angiogenesis induced by short-term exercise training. Am. J. Physiol. Heart Circ. Physiol. 2001, 281, H1163–H1169. [Google Scholar] [CrossRef] [PubMed]
  153. Volpert, O.V.; Ward, W.F.; Lingen, M.W.; Chesler, L.; Solt, D.B.; Johnson, M.D.; Molteni, A.; Polverini, P.J.; Bouck, N.P. Captopril inhibits angiogenesis and slows the growth of experimental tumors in rats. J. Clin. Investig. 1996, 98, 671–679. [Google Scholar] [CrossRef]
  154. Yoshiji, H.; Kuriyama, S.; Noguchi, R.; Yoshii, J.; Ikenaka, Y.; Yanase, K.; Namisaki, T.; Kitade, M.; Yamazaki, M.; Masaki, T.; et al. Combination of vitamin K2 and the angiotensin-converting enzyme inhibitor, perindopril, attenuates the liver enzyme-altered preneoplastic lesions in rats via angiogenesis suppression. J. Hepatol. 2005, 42, 687–693. [Google Scholar] [CrossRef] [PubMed]
  155. Attoub, S.; Gaben, A.M.; Al-Salam, S.; Al Sultan, M.A.; John, A.; Nicholls, M.G.; Mester, J.; Petroianu, G. Captopril as a potential inhibitor of lung tumor growth and metastasis. Ann. N Y Acad. Sci. 2008, 1138, 65–72. [Google Scholar] [CrossRef] [PubMed]
  156. Sipahi, I.; Chou, J.; Mishra, P.; Debanne, S.M.; Simon, D.I.; Fang, J.C. Meta-analysis of randomized controlled trials on effect of angiotensin-converting enzyme inhibitors on cancer risk. Am. J. Cardiol. 2011, 108, 294–301. [Google Scholar] [CrossRef] [PubMed]
  157. Perini, M.V.; Dmello, R.S.; Nero, T.L.; Chand, A.L. Evaluating the benefits of renin-angiotensin system inhibitors as cancer treatments. Pharmacol. Ther. 2020, 211, 107527. [Google Scholar] [CrossRef]
  158. Burnier, M.; Brunner, H.R. Angiotensin II receptor antagonists. Lancet 2000, 355, 637–645. [Google Scholar] [CrossRef]
  159. Schupp, M.; Janke, J.; Clasen, R.; Unger, T.; Kintscher, U. Angiotensin type 1 receptor blockers induce peroxisome proliferator-activated receptor-gamma activity. Circulation 2004, 109, 2054–2057. [Google Scholar] [CrossRef] [Green Version]
  160. Doggrell, S.A. Telmisartan-killing two birds with one stone. Expert Opin. Pharmacother. 2004, 5, 2397–2400. [Google Scholar] [CrossRef]
  161. Imayama, I.; Ichiki, T.; Inanaga, K.; Ohtsubo, H.; Fukuyama, K.; Ono, H.; Hashiguchi, Y.; Sunagawa, K. Telmisartan downregulates angiotensin II type 1 receptor through activation of peroxisome proliferator-activated receptor gamma. Cardiovasc. Res. 2006, 72, 184–190. [Google Scholar] [CrossRef]
  162. Takai, S.; Jin, D.; Kimura, M.; Kirimura, K.; Sakonjo, H.; Tanaka, K.; Miyazaki, M. Inhibition of vascular angiotensin-converting enzyme by telmisartan via the peroxisome proliferator-activated receptor gamma agonistic property in rats. Hypertens. Res. 2007, 30, 1231–1237. [Google Scholar] [CrossRef] [Green Version]
  163. Blessing, E.; Preusch, M.; Kranzhöfer, R.; Kinscherf, R.; Marx, N.; Rosenfeld, M.E.; Isermann, B.; Weber, C.M.; Kreuzer, J.; Gräfe, J.; et al. Anti-atherosclerotic properties of telmisartan in advanced atherosclerotic lesions in apolipoprotein E deficient mice. Atherosclerosis 2008, 199, 295–303. [Google Scholar] [CrossRef]
  164. Honda, A.; Matsuura, K.; Fukushima, N.; Tsurumi, Y.; Kasanuki, H.; Hagiwara, N. Telmisartan induces proliferation of human endothelial progenitor cells via PPARgamma-dependent PI3K/Akt pathway. Atherosclerosis 2009, 205, 376–384. [Google Scholar] [CrossRef]
  165. Zeng, X.C.; Li, X.S.; Wen, H. Telmisartan protects against microvascular dysfunction during myocardial ischemia/reperfusion injury by activation of peroxisome proliferator-activated receptor γ. BMC Cardiovasc. Disord. 2013, 13, 39. [Google Scholar] [CrossRef] [Green Version]
  166. Iwai, M.; Kanno, H.; Senba, I.; Nakaoka, H.; Moritani, T.; Horiuchi, M. Irbesartan increased PPARγ activity in vivo in white adipose tissue of atherosclerotic mice and improved adipose tissue dysfunction. Biochem. Biophys. Res. Commun. 2011, 406, 123–126. [Google Scholar] [CrossRef] [PubMed]
  167. Takai, S.; Jin, D.; Miyazaki, M. Irbesartan prevents metabolic syndrome in rats via activation of peroxisome proliferator-activated receptor γ. J. Pharmacol. Sci. 2011, 116, 309–315. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  168. Villapol, S.; Yaszemski, A.K.; Logan, T.T.; Sánchez-Lemus, E.; Saavedra, J.M.; Symes, A.J. Candesartan, an angiotensin II AT₁-receptor blocker and PPAR-γ agonist, reduces lesion volume and improves motor and memory function after traumatic brain injury in mice. Neuropsychopharmacology 2012, 37, 2817–2829. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  169. Koh, E.J.; Yoon, S.J.; Lee, S.M. Losartan protects liver against ischaemia/reperfusion injury through PPAR-γ activation and receptor for advanced glycation end-products down-regulation. Br. J. Pharmacol. 2013, 169, 1404–1416. [Google Scholar] [CrossRef] [PubMed]
  170. Zhang, Z.Z.; Shang, Q.H.; Jin, H.Y.; Song, B.; Oudit, G.Y.; Lu, L.; Zhou, T.; Xu, Y.L.; Gao, P.J.; Zhu, D.L.; et al. Cardiac protective effects of irbesartan via the PPAR-gamma signaling pathway in angiotensin-converting enzyme 2-deficient mice. J Transl Med 2013, 11, 229. [Google Scholar] [CrossRef] [Green Version]
  171. Zhong, J.; Gong, W.; Lu, L.; Chen, J.; Lu, Z.; Li, H.; Liu, W.; Liu, Y.; Wang, M.; Hu, R.; et al. Irbesartan ameliorates hyperlipidemia and liver steatosis in type 2 diabetic db/db mice via stimulating PPAR-γ, AMPK/Akt/mTOR signaling and autophagy. Int. Immunopharmacol. 2017, 42, 176–184. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. Zhao, W.; Shen, F.; Yao, J.; Su, S.; Zhao, Z. Angiotensin II receptor type 1 blocker candesartan improves morphine tolerance by reducing morphine-induced inflammatory response and cellular activation of BV2 cells via the PPARγ/AMPK signaling pathway. Mol. Med. Rep. 2022, 26, 1–10. [Google Scholar] [CrossRef] [PubMed]
  173. Clemenz, M.; Frost, N.; Schupp, M.; Caron, S.; Foryst-Ludwig, A.; Böhm, C.; Hartge, M.; Gust, R.; Staels, B.; Unger, T.; et al. Liver-specific peroxisome proliferator-activated receptor alpha target gene regulation by the angiotensin type 1 receptor blocker telmisartan. Diabetes 2008, 57, 1405–1413. [Google Scholar] [CrossRef] [Green Version]
  174. Rong, X.; Li, Y.; Ebihara, K.; Zhao, M.; Kusakabe, T.; Tomita, T.; Murray, M.; Nakao, K. Irbesartan treatment up-regulates hepatic expression of PPARalpha and its target genes in obese Koletsky (fa(k)/fa(k)) rats: A link to amelioration of hypertriglyceridaemia. Br. J. Pharmacol. 2010, 160, 1796–1807. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Kajiya, T.; Ho, C.; Wang, J.; Vilardi, R.; Kurtz, T.W. Molecular and cellular effects of azilsartan: A new generation angiotensin II receptor blocker. J. Hypertens. 2011, 29, 2476–2483. [Google Scholar] [CrossRef] [PubMed]
  176. Harada, M.; Kamijo, Y.; Nakajima, T.; Hashimoto, K.; Yamada, Y.; Shimojo, H.; Gonzalez, F.J.; Aoyama, T. Peroxisome proliferator-activated receptor α-dependent renoprotection of murine kidney by irbesartan. Clin. Sci. 2016, 130, 1969–1981. [Google Scholar] [CrossRef] [PubMed]
  177. Hattori, N.; Yamada, A.; Nakatsuji, S.; Matsuda, T.; Nishiyama, N.; Shimatsu, A. Telmisartan is the most effective ARB to increase adiponectin via PPARα in adipocytes. J. Mol. Endocrinol. 2022, 69, 259–268. [Google Scholar] [CrossRef]
  178. Li, L.; Luo, Z.; Yu, H.; Feng, X.; Wang, P.; Chen, J.; Pu, Y.; Zhao, Y.; He, H.; Zhong, J.; et al. Telmisartan improves insulin resistance of skeletal muscle through peroxisome proliferator-activated receptor-δ activation. Diabetes 2013, 62, 762–774. [Google Scholar] [CrossRef] [Green Version]
  179. Chang, W.T.; Cheng, J.T.; Chen, Z.C. Telmisartan improves cardiac fibrosis in diabetes through peroxisome proliferator activated receptor δ (PPARδ): From bedside to bench. Cardiovasc. Diabetol. 2016, 15, 113. [Google Scholar] [CrossRef] [Green Version]
  180. Xu, S.; Song, H.; Huang, M.; Wang, K.; Xu, C.; Xie, L. Telmisartan inhibits the proinflammatory effects of homocysteine on human endothelial cells through activation of the peroxisome proliferator-activated receptor-δ pathway. Int. J. Mol. Med. 2014, 34, 828–834. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  181. He, H.; Yang, D.; Ma, L.; Luo, Z.; Ma, S.; Feng, X.; Cao, T.; Yan, Z.; Liu, D.; Tepel, M.; et al. Telmisartan prevents weight gain and obesity through activation of peroxisome proliferator-activated receptor-delta-dependent pathways. Hypertension 2010, 55, 869–879. [Google Scholar] [CrossRef] [Green Version]
  182. Tong, Q.; Wu, L.; Jiang, T.; Ou, Z.; Zhang, Y.; Zhu, D. Inhibition of endoplasmic reticulum stress-activated IRE1α-TRAF2-caspase-12 apoptotic pathway is involved in the neuroprotective effects of telmisartan in the rotenone rat model of Parkinson’s disease. Eur. J. Pharmacol. 2016, 776, 106–115. [Google Scholar] [CrossRef] [PubMed]
  183. Li, Y.; Cheng, K.C.; Liu, K.F.; Peng, W.H.; Cheng, J.T.; Niu, H.S. Telmisartan Activates PPARδ to Improve Symptoms of Unpredictable Chronic Mild Stress-Induced Depression in Mice. Sci. Rep. 2017, 7, 14021. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Emanueli, C.; Salis, M.B.; Stacca, T.; Pinna, A.; Gaspa, L.; Madeddu, P. Angiotensin AT(1) receptor signalling modulates reparative angiogenesis induced by limb ischaemia. Br. J. Pharmacol. 2002, 135, 87–92. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Shimizu, T.; Okamoto, H.; Chiba, S.; Matsui, Y.; Sugawara, T.; Akino, M.; Nan, J.; Kumamoto, H.; Onozuka, H.; Mikami, T.; et al. VEGF-mediated angiogenesis is impaired by angiotensin type 1 receptor blockade in cardiomyopathic hamster hearts. Cardiovasc. Res. 2003, 58, 203–212. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Rakusan, K.; Chvojkova, Z.; Oliviero, P.; Ostadalova, I.; Kolar, F.; Chassagne, C.; Samuel, J.L.; Ostadal, B. ANG II type 1 receptor antagonist irbesartan inhibits coronary angiogenesis stimulated by chronic intermittent hypoxia in neonatal rats. Am. J. Physiol. Heart Circ. Physiol. 2007, 292, H1237–H1244. [Google Scholar] [CrossRef]
  187. Nenicu, A.; Körbel, C.; Gu, Y.; Menger, M.D.; Laschke, M.W. Combined blockade of angiotensin II type 1 receptor and activation of peroxisome proliferator-activated receptor-γ by telmisartan effectively inhibits vascularization and growth of murine endometriosis-like lesions. Hum. Reprod. 2014, 29, 1011–1024. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  188. Yamamoto, K.; Ohishi, M.; Ho, C.; Kurtz, T.W.; Rakugi, H. Telmisartan-induced inhibition of vascular cell proliferation beyond angiotensin receptor blockade and peroxisome proliferator-activated receptor-gamma activation. Hypertension 2009, 54, 1353–1359. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  189. Imai, N.; Hashimoto, T.; Kihara, M.; Yoshida, S.; Kawana, I.; Yazawa, T.; Kitamura, H.; Umemura, S. Roles for host and tumor angiotensin II type 1 receptor in tumor growth and tumor-associated angiogenesis. Lab. Investig. 2007, 87, 189–198. [Google Scholar] [CrossRef] [Green Version]
  190. Suganuma, T.; Ino, K.; Shibata, K.; Kajiyama, H.; Nagasaka, T.; Mizutani, S.; Kikkawa, F. Functional expression of the angiotensin II type 1 receptor in human ovarian carcinoma cells and its blockade therapy resulting in suppression of tumor invasion, angiogenesis, and peritoneal dissemination. Clin. Cancer Res. 2005, 11, 2686–2694. [Google Scholar] [CrossRef] [Green Version]
  191. Li, J.; Chen, L.; Yu, P.; Liu, B.; Zhu, J.; Yang, Y. Telmisartan exerts anti-tumor effects by activating peroxisome proliferator-activated receptor-γ in human lung adenocarcinoma A549 cells. Molecules 2014, 19, 2862–2876. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  192. Pu, Z.; Zhu, M.; Kong, F. Telmisartan prevents proliferation and promotes apoptosis of human ovarian cancer cells through upregulating PPARγ and downregulating MMP-9 expression. Mol. Med. Rep. 2016, 13, 555–559. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Sipahi, I.; Debanne, S.M.; Rowland, D.Y.; Simon, D.I.; Fang, J.C. Angiotensin-receptor blockade and risk of cancer: Meta-analysis of randomised controlled trials. Lancet Oncol. 2010, 11, 627–636. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Bangalore, S.; Kumar, S.; Kjeldsen, S.E.; Makani, H.; Grossman, E.; Wetterslev, J.; Gupta, A.K.; Sever, P.S.; Gluud, C.; Messerli, F.H. Antihypertensive drugs and risk of cancer: Network meta-analyses and trial sequential analyses of 324,168 participants from randomised trials. Lancet Oncol. 2011, 12, 65–82. [Google Scholar] [CrossRef]
  195. ARB Trialists Collaboration. Effects of telmisartan, irbesartan, valsartan, candesartan, and losartan on cancers in 15 trials enrolling 138,769 individuals. J. Hypertens. 2011, 29, 623–635. [Google Scholar] [CrossRef]
  196. Chauhan, V.P.; Chen, I.X.; Tong, R.; Ng, M.R.; Martin, J.D.; Naxerova, K.; Wu, M.W.; Huang, P.; Boucher, Y.; Kohane, D.S.; et al. Reprogramming the microenvironment with tumor-selective angiotensin blockers enhances cancer immunotherapy. Proc. Natl. Acad. Sci. USA 2019, 116, 10674–10680. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Simplified scheme of vascular PPAR actions with relevance for cardiovascular disease. ↑ indicates increase; ↓ indicates decrease.
Figure 1. Simplified scheme of vascular PPAR actions with relevance for cardiovascular disease. ↑ indicates increase; ↓ indicates decrease.
Ijms 24 02345 g001
Table 1. Summary of Clinical Trials with PPARα agonists for Cardiovascular Disease.
Table 1. Summary of Clinical Trials with PPARα agonists for Cardiovascular Disease.
Study Name/DrugConditionPrimary EndpointsOutcomeTrial Identifier/Reference
Fenofibrate Intervention and Event Lowering in Diabetes (FIELD) study
Fenofibrate
9795 patients with diabetes and dyslipidemiaNonfatal myocardial infarction, coronary heart disease mortalityNo significant reduction of coronary eventsnumber ISRCTN 64783481
[29]
Action to control cardiovascular risk in diabetes (ACCORD) trial
Simvastatin alone or in combination with Fenofibrate
5518 patients with DiabetesNonfatal myocardial infarction, nonfatal stroke, cardiovascular deathNo reduction of nonfatal myocardial infarction, nonfatal stroke, cardiovascular deathNCT00000620
[30]
Helsinki Heart Study
Gemfibrozil
4081 male patients with hyperlipidemiaCardiac death, myocardial infarctionReduction of coronary heart disease progression[33]
Veterans Affairs High-Density Lipoprotein Cholesterol Intervention Trial
Gemfibrozil
2531 male patients with dyslipidemia and coronary artery diseaseMyocardial infarction, death from coronary artery diseaseReduction of major cardiovascular events[34,35]
Pemafibrate to Reduce Cardiovascular Outcomes by Reducing Triglycerides in Patients with Diabetes (PROMINENT) trial
Pemafibrate
10,544 patients with diabetesNonfatal myocardial infarction, nonfatal stroke, cardiovascular deathStopped in 2022 due to futility[36]
NCT03071692
Table 2. Summary of Clinical Trials with PPARγ agonists for Cardiovascular Disease.
Table 2. Summary of Clinical Trials with PPARγ agonists for Cardiovascular Disease.
Study Name/DrugConditionPrimary EndpointsOutcomeTrial Identifier/Reference
PROspective pioglitAzone Clinical Trial In macroVascular Events (PROactive) trial
Pioglitazone
5238 patients with diabetes and cardiovascular diseaseTime to first occurrence of macrovascular events or deathNo significant reduction of coronary events, higher incidence of heart failureNCT00174993
[85,86]
Insulin Resistance Intervention After Stroke Trial (IRIS) trial
Pioglitazone
3876 patients with insulin resistance and stroke or myocardial infarctionRecurrent fatal or nonfatal Stroke, or fatal or nonfatal myocardial infarctionLower risk of stroke or myocardial infarctionNCT00091949 [89]
Diabetes Reduction Assessment with Ramipiril and Rosiglitazone Medication (DREAM) trial
Rosiglitazone
5962 patients with impaired glucose toleranceIncidence of diabetesLower incidence of diabetes, increase in cardiovascular eventsNCT00095654 [91]
A Diabetes Outcome Progression (ADOPT) trial
Rosiglitazone
4360 diabetic patientsTime to monotherapy failureTime to monotherapy failure reducedNCT00279045 [92]
Rosiglitazone Evaluated for Cardiac Outcomes and Regulation of Glycaemia in Diabetes (RECORD) trial
Rosiglitazone
4447 diabetic patientsCardiovascular events, long-term glycemic controlNo reduction of overall cardiovascular morbidity or mortalityNCT00379769 [94]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Wagner, N.; Wagner, K.-D. Pharmacological Utility of PPAR Modulation for Angiogenesis in Cardiovascular Disease. Int. J. Mol. Sci. 2023, 24, 2345. https://doi.org/10.3390/ijms24032345

AMA Style

Wagner N, Wagner K-D. Pharmacological Utility of PPAR Modulation for Angiogenesis in Cardiovascular Disease. International Journal of Molecular Sciences. 2023; 24(3):2345. https://doi.org/10.3390/ijms24032345

Chicago/Turabian Style

Wagner, Nicole, and Kay-Dietrich Wagner. 2023. "Pharmacological Utility of PPAR Modulation for Angiogenesis in Cardiovascular Disease" International Journal of Molecular Sciences 24, no. 3: 2345. https://doi.org/10.3390/ijms24032345

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop