Mini Review

PPARβ/δ in the Pathogenesis of Vascular Diseases

Zhanjun Jia1,2 and Shuzhen Li 1,2

1Nanjing Key Laboratory of Pediatrics, Children’s hospital of Nanjing Medical University, Nanjing 210008, China

2Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, China

*Corresponding authors: Zhanjun Jia, Nanjing Key Laboratory of Pediatrics, Children’s hospital of Nanjing Medical University, Nanjing 210008, China, Tel: +86 2583117309; Email: jiazj72@hotmail.com

Shuzhen Li, Nanjing Key Laboratory of Pediatrics, Children’s zhenleifeng@163.comhospital of Nanjing Medical University, Nanjing 210008, China, Tel: +86 13770627736; Email:

Citation: Jia Z and Li S (2018) PPARβ/δ in the Pathogenesis of Vascular Diseases. J Cardiovasc Dis Card Surg 2018: 03-06. doi:https://doi.org/10.29199/CDCS.101012

Received Date: 29 January, 2018; Accepted Date: 05 February, 2018; Published Date: 28 February, 2018

Abstract

Three members of PPAR family (PPARα, PPARγ, and PPARδ) have been widely studied in the past decades. Among three members of PPAR family, PPARα and PPARγ were better investigated and understood than PPARδ in various fields. However, in recent years, more and more studies revealed important roles of PPARδ in the physiology and pathology of many conditions including cardiovascular injury. In this mini review, we will introduce the roles of PPARδ in the pathology of vascular injury.

PPARδ is also termed as PPARβ which is expressed in many tissues [1-4], including vascular smooth muscle cells [5,6]. Recent evidence suggested that PPARβ/δ was involved in lipid metabolism in skeletal muscle [7,8]. In vascular smooth muscle cells [9,10] and cardiovascular tissues [11-13], PPARβ/δ is abundantly expressed. A recent study also suggested that PPARβ/δ contributed to a vasoprotective effect of a widely used antidiabetic drug metformin in obese mice [14]. Thus, PPARβ/δ is becoming an interesting novel target for the treatment of metabolic syndrome and metabolism disorder-associated cardiovascular diseases.

Although a number of studies have shown that PPARα and PPARγ exert anti-inflammatory, anti-proliferative, and antiangiogenic actions in cardiovascular cells, the role of PPARβ/δ in vascular pathophysiology is less understood [15,16]. In the past years, little evidence was reported on the relationship between PPARβ/δ and clinical complications. Moreover, the lack of PPARβ/δ-specific ligands on the market also hampered the research on the roles of PPARβ/δ in the disease conditions. However, in recent years, synthetic ligands of PPARβ/δ have been developed [17]. Furthermore, a novel subtype-selective and target gene-selective PPARβ/δ agonist, naturally occurring dimeric alkaloid picrasidine N (1) from picrasma quassioides was identified from a library consisting of plant extracts and natural compounds using a mammalian one-hybrid assay, which can selectively induce mRNA expression of PPARβ/δ target gene ANGPTL4 but not other PPAR target genes such as ADRP, PDK4, and CPT-1, suggesting its potential as an important compound for elucidating the mechanism of PPARβ/δ-regulated specific genes and the biological functions of PPARβ/δ [18].

ligand GW501516 also inhibited TNF-α-induced leukocyte rolling flux, adhesion, and emigration in a dose-dependent manner [29]. PPARβ/δ agonism can directly induce endothelium-dependent relaxation when used at higher concentrations [33] and can restore the endothelial function in animal models of both type 1 [34] and type 2 [35] diabetes through increasing NO bioavailability and suppressing the generation of NADPH oxidase-derived superoxide anions. However, PPARβ/δ ligand GW0742 on PPARβ/δ restored the endothelial function possibly through preserving the insulin-Akt-eNOS pathway impaired by high glucose [32]. A selective in vivo and in vitroIn endothelial cells, PPARβ/δ activation inhibits inflammation [19-21], oxidative stress [14,22], and apoptosis [23,24], but stimulates angiogenesis [25]. It is a known concept that atherosclerosis is an inflammatory disease [26-28]. Studies demonstrated that the PPARβ/δ ligands including L-165041, GW0742, and GW501516 alleviated inflammatory responses by reducing the translocation of NF-κB and the subsequent inhibition on VCAM-1, ICAM-1, E-selectin, and MCP-1 [20,29-31]. PPARβ/δ activation induced by PPARβ agonists GW0742 or L165041 lipid-related vascular injury was reported with diverse results by two independent studies. One report found that PPARβ/δ agonist GW0742 had no obvious effect on the lesion progression in fat- and cholesterol- supplemented Ldlr-/- mice, while another one detected a protective role of this agonist with a more aggressive dosing regimen [36,37], suggesting a dose-dependent effect.

Besides above evidence, more . GW0742 also could activate PPARβ/δ to restore the lipid-induced endothelial dysfunction by up-regulation of carnitine palmitoyltransferase-1 (CPT-1), thus reducing DAG accumulation and the subsequent PKC-mediated ROS production and eNOS inhibition [41]. In hypertensive rats, GW0742 prevented the development of endothelial dysfunction and hypertension by enhancing endothelial NOS activity, antioxidant genes and the regulators of G protein-coupled signaling proteins (RGS) 5, limiting NADPH oxidase activity, and reducing expressions of proinflammatory and pro-atherogenic genes [42,43]. In diet-induced obesity, treatment with GW0742 significantly prevented hypertension, vascular inflammatory and oxidative status, and endothelial dysfunction [39]. These evidences highly suggested an anti-hypertensive potential of GW0742 in the clinic.researches strongly supported a beneficial effect of PPARβ/δ agonist GW0742 on endothelial function [32,34,38,39]. Fan et al. reported that GW0742 protected endothelial cells against oxidative stress by upregulation of antioxidant enzymes, including superoxide dismutase-1, catalase and thioredoxin reductase [40]

-induced apoptosis by inducing PPARβ/δ binding to 2O2Prostacyclin (PGI2), an endogenous ligand for PPARβ/δ, protected human endothelial cells from H14-3-3ε promoter, thereby upregulating 14-3-3ε protein expression, which augmented Bad sequestration and prevented Bad-triggered apoptosis [44]. However, although PPARβ/δ agonists were shown to favorably modulate lipid metabolism and attenuate inflammation, thus reducing the susceptibility to atherosclerosis in both ApoE−/− and LDLR−/− mice [45,46], PPARβ−/− bone marrow transplants attenuated atherosclerotic lesion area in LDLR−/− mice possibly through enhancing the inflammatory suppressor Bcl-6 [47], suggesting different actions of PPARβ/δ in different cell types. Additionally, GW501516 and L-165041 enhance the recovery of blood flow in mice after hind limb ischemia and stimulate the proliferation of endothelial progenitor cells by acting on the PI3K/Akt pathway [48], indicating a protective role of PPARβ/δ under ischemic injury.

[49]. Furthermore, PPARβ/δ agonist GW0742 was also effective for the attenuation of neointimal hyperplasia by suppressing VSMC proliferation and accelerating reendothelization after arterial injury [5]. Activation of PPARβ/δ by GW0742 or adenovirus delivery of PPARβ/δ (Ad-PPARβ/δ) significantly inhibited kip1Vascular smooth muscle cell (VSMC) proliferation is the key pathogenic event of vascular proliferative diseases including atherosclerosis, restenosis, and vein graft failure. Activation of PPARβ/δ by GW501516 inhibited the PDGF-induced proliferation and migration of human pulmonary artery smooth muscle cells (HPASMC) as well as the collagen synthesis, which was associated with decreased expressions of cyclin D1, cyclin D3, CDK2, and CDK4, as well as the increased expressions of cell cycle inhibitory genes of G0S2 and P27hemoglobin-induced VSMC phenotypic switch involved in the pathophysiology of vascular injury after aneurysmal subarachnoid hemorrhage (aSAH) [50]. Thus, these solid evidence also suggested a beneficial role of PPARβ/δ in antagonizing VSMC proliferation-associated diseases [13,51-53].

In summary, PPARβ/δ activation chiefly accounts for a protection against endothelial injury and dysfunction, as well as the vascular cell proliferation via diverse mechanisms. Targeting PPARβ/δ might be a promising strategy in treating various vascular diseases including atherosclerosis and high blood pressure.

References

  1. Braissant O (1996) Differential expression of peroxisome proliferator-activated receptors (PPARs): tissue distribution of PPAR-alpha, -beta, and -gamma in the adult rat. Endocrinology 137: 354-66.
  2. Kliewer SA, Forman BM, Blumberg B, Ong ES, Borgmeyer U, et al. (1994) Differential expression and activation of a family of murine peroxisome proliferator-activated receptors. Proc Natl Acad Sci U S A 91: 7355-7359.
  3. Feige JN, Gelman L, Michalik L, Desvergne B, Wahli W, (2006) From molecular action to physiological outputs: peroxisome proliferator-activated receptors are nuclear receptors at the crossroads of key cellular functions. Prog Lipid Res 452: 120-59.
  4. Manzur M, Ganss R (2009) Regulator of G protein signaling 5: a new player in vascular remodeling. Trends Cardiovasc Med 19: 26-30.
  5. Hamaya R, Ogawa M, Suzuki JI, Kobayashi N, Hirata Y, et al. (2013) A selective peroxisome proliferator-activated receptor-beta/delta agonist attenuates neointimal hyperplasia after wire-mediated arterial injury. Expert Opin Investig Drugs 22: 1095-1106.
  6. Cheang WS, Fang X, Tian XY (2013) Pleiotropic effects of peroxisome proliferator-activated receptor γ and δ in vascular diseases. Circ J 77: 2664-2671.
  7. Lee J. and Chung WY (2011) The role played by the peroxisome proliferator-activated receptor-beta/delta (PPARbeta/delta) activator, GW501516, in control of fatty acid metabolism: a new potential therapeutic target for treating metabolic syndrome. Endocrinology 152:1742-1744.
  8. Chawla A, Lee CH, Barak Y, He W, Rosenfeld J, et al. (2003) PPARdelta is a very low-density lipoprotein sensor in macrophages. Proc Natl Acad Sci U S A 100: 1268-1273.
  9. Zhang J, Fu M, Zhu X, Xiao Y, Mou Y, et al. (2002) Peroxisome proliferator-activated receptor delta is up-regulated during vascular lesion formation and promotes post-confluent cell proliferation in vascular smooth muscle cells. J Biol Chem 277: 11505-11512.
  10. Girroir EE, Hollingshead HE, He P, Zhu B, Perdew GH, et al. (2008) Quantitative expression patterns of peroxisome proliferator-activated receptor-beta/delta (PPARbeta/delta) protein in mice. Biochem Biophys Res Commun 371: 456-461.
  11. Gilde AJ, van der Lee KA, Willemsen PH, Chinetti G, van der Leij FR, et al. (2003) Peroxisome proliferator-activated receptor (PPAR) alpha and PPARbeta/delta, but not PPARgamma, modulate the expression of genes involved in cardiac lipid metabolism. Circ Res 92: 518-524.
  12. Galatou E, Kelly T, Lazou A (2014) The PPAR β/δ agonist GW0742 modulates signaling pathways associated with cardiac myocyte growth via a non-genomic redox mechanism. Mol Cell Biochem 395: 145-154.
  13. Ehrenborg E, Skogsberg J (2013) Peroxisome proliferator-activated receptor delta and cardiovascular disease. Atherosclerosis 231: 95-106.
  14. Cheang WS, Tian XY, Wong WT, Lau CW, Lee SS, et al. (2014) Metformin protects endothelial function in diet-induced obese mice by inhibition of endoplasmic reticulum stress through 5' adenosine monophosphate-activated protein kinase-peroxisome proliferator-activated receptor δ pathway. Arterioscler Thromb Vasc Biol 34: 830-836.
  15. Chawla A (2010) Control of macrophage activation and function by PPARs. Circ Res 106: 1559-1569.
  16. Sertznig P, Seifert M, Tilgen W, Reichrath J (2007) Present concepts and future outlook: function of peroxisome proliferator-activated receptors (PPARs) for pathogenesis, progression, and therapy of cancer. J Cell Physiol 212: 1-12.
  17. Berger J, Leibowitz MD, Doebber TW, Elbrecht A, Zhang B, et al. (1999) Novel peroxisome proliferator-activated receptor (PPAR) gamma and PPARdelta ligands produce distinct biological effects. J Biol Chem 274: 6718-6725.
  18. Zhao S, Kanno Y, Li W, Wakatabi H, Sasaki T, et al. (2016) Picrasidine N Is a Subtype-Selective PPARβ/δ Agonist. J Nat Prod 79: 879-885.
  19. Bojic LA1, Sawyez CG, Telford DE, Edwards JY, Hegele RA, et al. (2012) Activation of peroxisome proliferator-activated receptor δ inhibits human macrophage foam cell formation and the inflammatory response induced by very low-density lipoprotein. Arterioscler Thromb Vasc Biol 32: 2919-2928.
  20. Xu S, Song H, Huang M, Wang K, Xu C et al. (2014) 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 34: 828-834.
  21. Woo CH, Massett MP, Shishido T, Itoh S, Ding B, et al. (2006) ERK5 activation inhibits inflammatory responses via peroxisome proliferator-activated receptor delta (PPARdelta) stimulation. J Biol Chem 281: 32164-32174.
  22. Xiao GF, Xu SH, Chao Y, Xie LD, Xu CS, et al. (2014) PPARδ activation inhibits homocysteine-induced p22(phox) expression in EA.hy926 cells through reactive oxygen species/p38MAPK pathway. Eur J Pharmacol 727: 29-34.
  23. al Yacoub N, Romanowska M, Krauss S, Schweiger S, Foerster J (2008) PPARdelta is a type 1 IFN target gene and inhibits apoptosis in T cells. J Invest Dermatol, 128: 1940-1949.
  24. Chao X et al.(2014) Activation of peroxisome proliferator-activated receptor beta/delta attenuates acute ischemic stroke on middle cerebral ischemia occlusion in rats. J Stroke Cerebrovasc Dis 23: 1396-402.
  25. Stephen RL, Gustafsson MC, Jarvis M, Tatoud R, Marshall BR, et al. (2004) Activation of peroxisome proliferator-activated receptor delta stimulates the proliferation of human breast and prostate cancer cell lines. Cancer Res 64: 3162-3170.
  26. Montecucco F, Mach F (2009) Atherosclerosis is an inflammatory disease. Semin Immunopathol 31: 1-3.
  27. Jain MK, Sangwung P, Hamik A (2014) Regulation of an inflammatory disease: Krüppel-like factors and atherosclerosis. Arterioscler Thromb Vasc Biol 34: 499-508.
  28. Hidalgo A, Tello L, Toro EF (2014) Numerical and analytical study of an atherosclerosis inflammatory disease model. J Math Biol 68: 1785-1814.
  29. Piqueras L, Sanz MJ, Perretti M, Morcillo E, Norling L, et al. (2009) Activation of PPARbeta/delta inhibits leukocyte recruitment, cell adhesion molecule expression, and chemokine release. J Leukoc Biol 86: 115-122.
  30. Liang YJ, Liu YC, Chen CY, Lai LP, Shyu KG, et al. (2010) Comparison of PPARδ and PPARγ in inhibiting the pro-inflammatory effects of C-reactive protein in endothelial cells. Int J Cardiol 143: 361-367.
  31. Luz-Crawford P, Ipseiz N, Espinosa-Carrasco G, Caicedo A, Tejedor G, et al. (2016) PPAR β/δ directs the therapeutic potential of mesenchymal stem cells in arthritis. Ann Rheum Dis 75: 2166-2174.
  32. Quintela AM, Jiménez R, Piqueras L, Gómez-Guzmán M, Haro J, et al. (2014) PPARbeta activation restores the high glucose-induced impairment of insulin signalling in endothelial cells. Br J Pharmacol 171: 3089-3102.
  33. Jiménez R, Sánchez M, Zarzuelo MJ, Romero M, Quintela AM, et al. (2010) Endothelium-dependent vasodilator effects of peroxisome proliferator-activated receptor beta agonists via the phosphatidyl-inositol-3 kinase-Akt pathway. J Pharmacol Exp Ther 332: 554-561.
  34. Quintela AM, Jiménez R, Gómez-Guzmán M, Zarzuelo MJ, Galindo P, et al. (2012) Activation of peroxisome proliferator-activated receptor-β/-δ (PPARβ/δ) prevents endothelial dysfunction in type 1 diabetic rats. Free Radic Biol Med 53: 730-741.
  35. Tian J, Wang J, Li Y, Villarreal D, Carhart R, et al. (2012) Endothelial function in patients with newly diagnosed type 2 diabetes receiving early intensive insulin therapy. Am J Hypertens 25: 1242-1248.
  36. Li AC1, Binder CJ, Gutierrez A, Brown KK, Plotkin CR, et al. (2004) Differential inhibition of macrophage foam-cell formation and atherosclerosis in mice by PPARalpha, beta/delta, and gamma. J Clin Invest 114: 1564-1576.
  37. Graham TL, Mookherjee C, Suckling KE, Palmer CN, Patel L (2005) The PPARdelta agonist GW0742X reduces atherosclerosis in LDLR(-/-) mice. Atherosclerosis 181: 29-37.
  38. Romero M, Toral M, Robles-Vera I, Sánchez M, Jiménez R, et al. (2017) Activation of Peroxisome Proliferator Activator Receptor β/δ Improves Endothelial Dysfunction and Protects Kidney in Murine Lupus. Hypertension 69: 641-650.
  39. Toral M, Gómez-Guzmán M, Jiménez R, Romero M, Zarzuelo MJ, et al. (2015) Chronic peroxisome proliferator-activated receptor β/δ agonist GW0742 prevents hypertension, vascular inflammatory and oxidative status, and endothelial dysfunction in diet-induced obesity. J Hypertens 33: 1831-1844.
  40. Fan Y, Wang Y, Tang Z, Zhang H, Qin X, et al. (2008) Suppression of pro-inflammatory adhesion molecules by PPAR-delta in human vascular endothelial cells. Arterioscler Thromb Vasc Biol 28: 315-321.
  41. Toral M, Romero M, Jiménez R, Mahmoud AM, Barroso E, et al. (2015) Carnitine palmitoyltransferase-1 up-regulation by PPAR-β/δ prevents lipid-induced endothelial dysfunction. Clin Sci (Lond) 129: 823-837.
  42. Zarzuelo MJ, Jiménez R, Galindo P, Sánchez M, Nieto A, et al. (2011) Antihypertensive effects of peroxisome proliferator-activated receptor-β activation in spontaneously hypertensive rats. Hypertension 58: 733-743.
  43. Zarzuelo MJ, Gómez-Guzmán M, Jiménez R, Quintela AM, Romero M, et al. (2013) Effects of peroxisome proliferator-activated receptor-β activation in endothelin-dependent hypertension. Cardiovasc Res 99: 622-631.
  44. Liou JY, Lee S, Ghelani D, Matijevic-Aleksic N, Wu KK (2006) Protection of endothelial survival by peroxisome proliferator-activated receptor-delta mediated 14-3-3 upregulation. Arterioscler Thromb Vasc Biol 26: 1481-1487.
  45. Barish GD, Atkins AR, Downes M, Olson P, Chong LW, et al. (2008) PPARdelta regulates multiple proinflammatory pathways to suppress atherosclerosis. Proc Natl Acad Sci U S A 105: 4271-4276.
  46. Bojic LA, Burke AC, Chhoker SS, Telford DE, Sutherland BG, et al. (2014) Peroxisome proliferator-activated receptor δ agonist GW1516 attenuates diet-induced aortic inflammation, insulin resistance, and atherosclerosis in low-density lipoprotein receptor knockout mice. Arterioscler Thromb Vasc Biol 34: 52-60.
  47. Lee CH, Chawla A, Urbiztondo N, Liao D, Boisvert WA, et al. (2003) Transcriptional repression of atherogenic inflammation: modulation by PPARdelta. Science 302: 453-457.
  48. Han JK, Lee HS, Yang HM, Hur J, Jun SI et al. (2008) Peroxisome proliferator-activated receptor-delta agonist enhances vasculogenesis by regulating endothelial progenitor cells through genomic and nongenomic activations of the phosphatidylinositol 3-kinase/Akt pathway. Circulation 118: 1021-1033.
  49. Liu G, Li X, Li Y, Tang X, Xu J et al. (2013) PPARδ agonist GW501516 inhibits PDGF-stimulated pulmonary arterial smooth muscle cell function related to pathological vascular remodeling. Biomed Res Int 2013: 903947.
  50. Zhang H, Jiang L, Guo Z, Zhong J, Wu J, et al. (2017) PPAR β/δ, a Novel Regulator for Vascular Smooth Muscle Cells Phenotypic Modulation and Vascular Remodeling after Subarachnoid Hemorrhage in Rats. Sci Rep 7: 45234.
  51. Kim HJ, Kim MY, Hwang JS, Kim HJ, Lee JH et al. (2010) PPARdelta inhibits IL-1beta-stimulated proliferation and migration of vascular smooth muscle cells via up-regulation of IL-1Ra. Cell Mol Life Sci 67: 2119-2130.
  52. Hwang JS, Ham SA, Yoo T, Lee WJ, Paek KS, et al. (2016) Sirtuin 1 Mediates the Actions of Peroxisome Proliferator-Activated Receptor δ on the Oxidized Low-Density Lipoprotein-Triggered Migration and Proliferation of Vascular Smooth Muscle Cells. Mol Pharmacol 90: 522-529.
  53. Hytönen J, Leppänen O, Braesen JH, Schunck WH, Mueller D, et al. (2016) Activation of Peroxisome Proliferator-Activated Receptor-δ as Novel Therapeutic Strategy to Prevent In-Stent Restenosis and Stent Thrombosis. Arterioscler Thromb Vasc Biol 36: 1534-1548.