14-3-3 pathway

14-3-3 pathway

G Model ARTICLE IN PRESS PRO 6114 1–9 Prostaglandins & other Lipid Mediators xxx (2015) xxx–xxx Contents lists available at ScienceDirect Prostag...

569KB Sizes 0 Downloads 43 Views

G Model

ARTICLE IN PRESS

PRO 6114 1–9

Prostaglandins & other Lipid Mediators xxx (2015) xxx–xxx

Contents lists available at ScienceDirect

Prostaglandins and Other Lipid Mediators

1

Invited review

2

Prostacyclin protects vascular integrity via PPAR/14-3-3 pathway

3

Q1

Ling-yun Chu a , Jun-Yang Liou a,b , Kenneth K. Wu a,b,c,∗ a

Metabolomic Medicine Research Center, China Medical University, Taichung, Taiwan Institute of Cell and System Medicine, National Health Research Institute, Chunan, Taiwan c Department of Medical Sciences, National Tsing-Hua University, Hsin-chu, Taiwan

4

b

5 6 7

a r t i c l e

8 20

i n f o

a b s t r a c t

9

Article history: Received 19 January 2015 Received in revised form 25 March 2015 Accepted 13 April 2015 Available online xxx

10 11 12 13 14 15

19

Keywords: Prostacyclin PPAR 14-3-3

21

Contents

16 17 18

1. 2. 3. 4. 5. 6. 7. 8. 9. 10. 11. 12. 13.

22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38

Vascular integrity is protected by the lining endothelial cells (ECs) through structural and molecular protective mechanisms. In response to external stresses, ECs are dynamic in producing protective molecules such as prostacyclin (PGI2 ). PGI2 is known to inhibit platelet aggregation and controls smooth muscle cell contraction via IP receptors. Recent studies indicate that PGI2 defends endothelial survival and protects vascular smooth muscle cell from apoptosis via peroxisome-proliferator activated receptors (PPAR). PPAR activation results in 14-3-3 upregulation. Increase in cytosolic 14-3-3␧ or 14-3-3␤ enhances binding and sequestration of Akt-mediated phosphorylated Bad and reduces Bad-mediated apoptosis via the mitochondrial pathway. Experimental data indicate that administration of PGI2 analogs or augmentation of PGI2 production by gene transfer attenuates endothelial damage and organ infarction caused by ischemia–reperfusion injury. The protective effect of PGI2 is attributed in part to preserving endothelial integrity. © 2015 Published by Elsevier Inc.

Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Endothelial cell is a major source of PGI2 production . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . PGI2 inhibits thromboxane A2 (TXA2 )-induced platelet aggregation and SMC contraction via IP signaling pathway . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . PGI2 prevents stress-induced EC apoptosis via peroxisome-proliferator activated receptors (PPARs) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . PGI2 controls vascular SMC apoptosis via PPAR␣ . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . PPAR␦-mediated 14-3-3␧ upregulation confers resistance to apoptosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . PGI2 protects VSMCs from H2 O2 -induced apoptosis by activating PPAR␣ → 14-3-3␤ transcriptional pathway . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . PGI2 prevents vascular cell apoptosis through multiple mechanisms . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Prostacyclin analogs protect endothelial barrier function via cyclic AMP pathway . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Endothelial damage predisposes heart and brain to ischemia reperfusion injury . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . PGI2 and 15d-PGJ2 protect against ischemia–reperfusion injury . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Physiological and therapeutic considerations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Conclusion and future perspective . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Conflict of interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Author and contributions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Acknowledgement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .

00 00 00 00 00 00 00 00 00 00 00 00 00 00 00 00 00

39

1. Introduction

40

41Q3

Blood vessels form a closed transportation system for effective oxygen and nutrient delivery and CO2 and waste disposal. A typical

∗ Corresponding author at: Metabolomic Medicine Research Center, China Medical Q2 University, Taichung, Taiwan. Tel.: +886 713 500 6800. E-mail address: [email protected] (K.K. Wu).

medium-size artery comprises three layers of tissues and a central lumen. Facing the lumen is the intimal layer (tunica intima) which is composed of a single layer of cells, i.e. the vascular endothelial cells and subendothelial connective tissues. The medial layer (tunica media) which is separated from the intimal layer by a dense band of elastic tissue, i.e. internal elastic lamina, is composed of smooth muscle cells and supportive tissues. The adventitia layer (tunica externa) which is separated from media by external elastic lamina contains fibroblasts, nerve innovation and other cell types.

http://dx.doi.org/10.1016/j.prostaglandins.2015.04.006 1098-8823/© 2015 Published by Elsevier Inc.

Please cite this article in press as: Chu L-y, et al. Prostacyclin protects vascular integrity via PPAR/14-3-3 pathway. Prostaglandins Other Lipid Mediat (2015), http://dx.doi.org/10.1016/j.prostaglandins.2015.04.006

42 43 44 45 46 47 48 49 50

G Model PRO 6114 1–9 2 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65 66 67 68 69 70 71 72 73 74 75 76 77 78 79 80 81 82 83 84 85 86 87 88

89

90 91 92 93 94 95 96 97 98 99 100 101 102 103 104 105 106 107 108 109 110 111 112 113 114

ARTICLE IN PRESS L.-y. Chu et al. / Prostaglandins & other Lipid Mediators xxx (2015) xxx–xxx

Each layer possesses special properties which contribute to vascular integrity and function. The intimal layer provides structural barrier to control vascular permeability and produce vasoprotective molecules to defend against invading toxins and insulting agents. The chief function of the medial layer is to provide regulated vascular contractility. Adventitia is a gateway for blood vessels to communicate with the outside world. Circulating in the lumen of blood vessel are normal blood constituents including blood cells, proteins, small-molecule nutrients and wastes, electrolytes and metals. Under physiological conditions, blood constituents do not interact with the lining endothelium and their passage through the vascular wall is tightly controlled. However, foreign microorganisms and toxins as well as endogenously produced cytokines and immune mediators often place endothelium under stresses which threaten to disrupt barrier, damage endothelial cells and perturb vascular integrity. Blood vessels are resilient and able to withstand the stresses thanks to dynamic innate protective mechanisms. Vascular endothelial cells (EC) are a key player of the innate protection. They are endowed with cellular and molecular ammunitions to defend against the insulting factors. ECs possess intercellular tight junctions to serve as a barrier and constitutively express thrombomodulin and protein C receptors at the luminal surface to prevent coagulation and thrombosis. Furthermore, they contain dynamic metabolic pathways which respond to the environmental insults by producing biologically active molecules to protect the vascular wall. Two pathways have been well characterized: (1) l-arginine catabolism to generate nitric oxide (NO) and (2) arachidonic acid (AA) metabolism to generate protective eicosanoids notably prostacyclin (PGI2 ) and epoxyeicosatrienoic acids (EETs). In response to vascular injury, NO and PGI2 are concurrently generated and act synergistically to block platelet activation and aggregation and relax vascular smooth muscle. Earlier investigations have centered on the actions of PGI2 and NO on blood platelets and vascular smooth muscle cell (SMC) contraction. Recent studies indicate that they protect ECs and SMCs from apoptosis and defend against inflammation and tissue injury. This review will focus on the anti-apoptotic actions of PGI2 and the underlying mechanisms. 2. Endothelial cell is a major source of PGI2 production In response to the environmental stresses, AA metabolism is initiated with activation of phospholipase A2 (PLA2 ). PLA2 is translocated from plasma membrane to the outer surface of endoplasmic reticulum (ER) and nuclear envelope (NE) where it catalyzes the liberation of AA from membrane phospholipids notably phosphatidylcholine. Free AA is converted to diverse metabolites by three major pathways: (1) cyclooxygenase (COX) pathway; (2) lipoxygenase (LOX) pathway and (3) cytochrome p450 (CYP) oxygenase pathway. Within each pathway, multiple metabolites are synthesized by their specific enzymes. The COX pathway generates several structure-related prostaglandins, thromboxane and prostacyclin (PGI2 ), while the LOX pathway produces leukotrienes, 5-, 12- and 15-HETEs (hydroxyeicosatetraenoic acid). The CYP pathway produces EETs and 20-HETE. All the AA metabolites share a 20-carbon unsaturated fatty acid backbone and hence are collectively called eicosanoids. Eicosanoids have diverse biological activities and play a broad spectrum of physiological roles. They are involved in myriad pathophysiological processes. Each cell type expresses a specific set of enzymes for synthesis of a selective list of eicosanoids. Vascular ECs produce eicosanoids from all three metabolic pathways among which prostacyclin (PGI2 ) is a predominant COX metabolite with vasoprotective actions. Prostacyclin was discovered as a metabolite of prostaglandin endoperoxide produced by vascular wall [1,2]. Its synthetic

PGI2 PGIS PLA2

ER or NE membrane

COX AA

PGI2 PGH2

Fig. 1. Schematic illustration of functional coupling of PGI2 synthetic enzymes. Abbreviations: PLA2 , phospholipase A2 ; AA, arachidonic acid; COX, cyclooxygenase; PGIS, PGI2 (prostacyclin) synthase; ER, endoplasmic reticulum; NE, nuclear envelope.

enzyme, prostacyclin synthase (PGIS) was isolated from arteries [3] which was subsequently cloned [4,5]. Biochemical characterization of PGI synthase reveals that it belongs to cytochrome p450 (CYP450) superfamily. However, it is an atypical CYP450 as it does not possess oxygenase activity but acts as an isomerase [6,7]. It anchors to the outer membrane of ER and NE by a single transmembranous domain and its substrate channel is attached to the outer membrane of ER and NE by hydrophobic interactions [8]. As illustrated in Fig. 1, PGI synthase is thought to be functionally coupled to the upstream enzymes at ER and NE membranes to facilitate PGI2 synthesis. A majority of the produced PGI2 is released into the extracellular milieu. A fraction of PGI2 is thought to enter nucleus. However, this presumption has not been proved by experimental data. Blood vessels are the principal source of PGI2 production. Early studies have provided quantitative data about PGI2 production by vascular cells [9]. Endothelial cells (EC) have a robust synthesis of PGI2 . Vascular smooth muscle cells (SMC) produce a smaller amount, about 1/7–1/10 of that produced by EC. Vascular adventitial fibroblasts produce only a trace amount of PGI2 . PGI2 production by ECs is regulated at the COX step. There are two COX isoforms in ECs. COX-1 is a house keeping enzyme which catalyzes the production of a basal level of PGI2 . By contrast, COX2 is highly responsive to exogenous stimuli. Shear stress as well as chemical stimuli such as cytokines, endotoxins, environmental toxins, immune and pro-inflammatory mediators induces COX-2 expression at the transcriptional level resulting in production of abundant PGI2 [10–13]. Stress-coupled PGI2 production is considered to play an important protective role [14]. 3. PGI2 inhibits thromboxane A2 (TXA2 )-induced platelet aggregation and SMC contraction via IP signaling pathway Effects of PGI2 on platelet reactivity and SMC contractility have been extensively investigated. PGI2 inhibits platelet aggregation induced by various physiological agonists. Of particularly importance is its inhibition of platelet aggregation induced by thromboxane A2 (TXA2 ). TXA2 was identified as a metabolite of prostaglandin endoperoxide [15] which is produced in platelets by a specific enzyme, thromboxane synthase [16,17]. It was initially identified as an autacoid to induce platelet aggregation and subsequently reported to constrict arteries. Interestingly, PGI2 has actions opposite to TXA2 : it inhibits TXA2 -induced platelet aggregation and vaso-constriction. The yin-yang relationship of PGI2 vs. TXA2 proves to be physiologically relevant [18] and pharmacologically important. It is suggested that the cardiovascular complications of selective COX-2 inhibitors are attributable to loss of the vaso-protective PGI2 , leaving the actions of TXA2 un-opposed with consequent increase in the risk of vascular thrombosis and excessive vasoconstriction [19]. PGI2 inhibits platelet aggregation and vascular SMC contractility by binding to a specific membrane G-protein coupled receptor, i.e. the I-type prostaglandin (IP) receptor, which activates adenylyl

Please cite this article in press as: Chu L-y, et al. Prostacyclin protects vascular integrity via PPAR/14-3-3 pathway. Prostaglandins Other Lipid Mediat (2015), http://dx.doi.org/10.1016/j.prostaglandins.2015.04.006

115 116 117 118 119 120 121 122 123 124 125 126 127 128 129 130 131 132 133 134 135 136 137 138 139 140 141 142 143

144 145

146 147 148 149 150 151 152 153 154 155 156 157 158 159 160 161 162 163 164 165

G Model PRO 6114 1–9

ARTICLE IN PRESS L.-y. Chu et al. / Prostaglandins & other Lipid Mediators xxx (2015) xxx–xxx

166 167 168 169 170 171 172

173 174

175 176 177 178 179 180 181 182 183 184 185 186 187 188 189 190 191 192 193 194 195 196 197 198 199 200 201 202 203 204 205 206 207 208 209 210 211 212 213 214 215 216 217 218 219 220 221 222 223 224 225 226 227 228 229

cyclase and generates cyclic AMP [20,21]. Cyclic AMP serves as a second messenger to activate protein kinase A (PKA) which mediates relaxation of vascular SMCs and inhibition of platelet reactivity. The IP signaling pathway is essential for defending against thrombosis and vascular diseases as genetic deletion of IP receptors in mice increases risk of thrombi formation, intimal hyperplasia and atherogenesis [22–24]. 4. PGI2 prevents stress-induced EC apoptosis via peroxisome-proliferator activated receptors (PPARs) It was reported that stable analogs of PGI2 protect myocardial cells from adriamycin-induced apoptosis and hypertonicityinduced renal epithelial cell apoptosis [25,26]. But the mechanism was unclear. It was subsequently noted that PGI2 analogs protect against apoptosis via PPAR receptors. PPAR belong to the nuclear receptor superfamily. Three members: PPAR␣, PPAR␥ and PPAR␦ (homologous to PPAR␤ in chicken cells) have been detected in mammalian cells [27]. Their natural ligands in cells are not entirely clear but are thought to include fatty acid derivatives such as PGI2 . Kliewer et al. and Forman et al. were the first to report that stable PGI2 analogs such as iloprost and carbaprostacyclin (cPGI2 ) bind and activate PPAR␦ and PPAR␣ [28,29]. Liou et al. reported that pretreatment of human umbilical vein EC (HUVEC) with cPGI2 prevents H2 O2 -induced apoptosis and silencing of PPAR␦ abrogates the anti-apoptotic action of cPGI2 [30]. Furthermore, repetitive lowlevel H2 O2 stress was reported to protect HUVEC from apoptosis via PPAR␦ activation [31]. Besides protection against H2 O2 -induced apoptosis, PPAR␦ agonists prevent endothelial dysfunction induced by ␤-amyloid precursor proteins or cigarette smoke [32,33]. These findings are in agreement with the concept that external addition of synthetic PGI2 analogs protect ECs and enhance EC functions through a mechanism involving PPAR␦ [34]. It is interesting to note that the effective anti-apoptotic concentrations of cPGI2 are 50–100 ␮M which are much higher than the effective concentrations in controlling platelet activation and SMC contractility. Due to a short half-life of PGI2 in aqueous solution, it is difficult to determine whether authentic PGI2 binds and activates PPARs by in vitro assays. To provide evidence that endogenously produced PGI2 acts through activation of PPARs, the effect of amplified PGI2 production by gene transfer on EC survival is evaluated. We have reported that transfection of HUVEC with an adenoviral vector containing a bicistronic COX-1 and PGIS construct (Ad-COPI) augments PGI2 production while suppresses the synthesis of other eicosanoids due to metabolic shift to the COX → PGIS pathway [35]. Ad-COPI transfected HUVECs are resistant to H2 O2 -induced apoptosis which is abrogated by silencing of PPAR␦ expression with specific siRNA [30]. Ad-COPI transfection into renal cells defends against gentamicin-induced renal cell apoptosis via PPAR␣ [36]. These results suggest that authentic PGI2 is effective in protecting ECs from apoptosis via PPAR␦ and/or PPAR␣. A majority of PGI2 produced by Ad-COPI are secreted into extracellular milieu. It is possible that a fraction of the PGI2 produced enter nucleus to bind and activate PPAR␦ and/or PPAR␣. PGI2 released into the extracellular milieu acts in a paracrine manner to protect surrounding ECs from apoptosis. It remain unclear how the extracellular PGI2 or PGI2 agonists penetrates plasma membrane and traverses cytoplasm to reach nucleus to target PPAR␦ and/or PPAR␣. Activation of PPAR␦ in ECs by synthetic agonists not only protects EC from apoptosis but also promotes EC proliferation and angiogenesis [37]. Furthermore, it reduces stress-induced expression of VCAM-1 and E-selectin and increases the expression of superoxide dismutase 1, catalase and thioredoxin and consequently suppresses reactive oxygen species (ROS) generation [38]. It is unclear how PGI2 controls the expression of pro-inflammatory genes and increases the expression of anti-oxidant enzymes.

3

PPAR␣ proteins are detected in ECs. It was reported that PPAR␣ activation by synthetic agonists protects HUVEC from apoptosis [39]. In fact, PPAR␣ activation by agonists protect against apoptosis in a number of cell types including renal cells [36], cardiomyocytes [40] and hepatocytes [41]. Genetic deletion of PPAR␣ in mice renders EC dysfunctional and unable to protect against ischemia–reperfusion injury [42]. The reported data suggest that PPAR␦ and PPAR␣ are functionally important in protecting ECs which appear to be depend on the type of insulting agents. 5. PGI2 controls vascular SMC apoptosis via PPAR␣ SMC apoptosis not only reduces cell mass and weakens atherosclerotic plaques but also induces inflammatory responses [43]. Furthermore, vascular SMC apoptosis is associated with increased SMC migration, proliferation and extracellular matrix production [44]. Vascular MSC apoptosis is induced by reactive oxygen species and pro-inflammatory cytokines. PGI2 was reported to protect SMC from oxidant-induced apoptosis [44]. cPGI2 as well as Ad-COPI transfection increases resistance to H2 O2 -induced apoptosis of neonatal rat aortic SMC [45]. It is interesting that the anti-apoptotic effect of PGI2 is abolished by PPAR␣ inhibitors but not PPAR␦ or PPAR␥ inhibitors. Importance of PPAR␣ in defending against apoptosis is supported by experimental results which show that PPAR␣ agonists protect SMCs from apoptosis. PPAR␣ overexpression in SMCs by transient transfection renders SMCs resistant to H2 O2 -induced apoptosis. Thus, in contrast with the predominate role of PPAR␦ in controlling EC apoptosis, PPAR␣ is pivotal in suppressing stress-induced SMC apoptosis. It is unclear why PGI2 targets different PPAR isoforms in EC vs SMC. One possible reason is differential quantitative expression of PPAR␣ and PPAR␦ in these two vascular cells. Current available data are insufficient to address this possibility. It is important to recognize that PPAR activation is regulated by IP receptor signaling. It has been reported that IP receptor activation by PGI2 analogs upregulates PPAR␦ in adipocytes [46,47]. Although PGI2 and its analogs do not directly activate PPAR␥, they activate PPAR␥ by IP receptor activation and the consequent cyclic AMP-PKA signaling [48]. In vascular SMCs, iloprost was reported to bind IP receptors and activate PKA whereby it induces COX-2 expression and PGI2 production [49]. It was proposed that the endogenously produced PGI2 activates PPAR␣ or ␦ to protect vascular SMC survival and contractile property [50]. Thus, there is a close relationship between IP receptor activation and PPAR activation in cells expressing both receptors such as vascular SMCs. It remains to be investigated whether IP receptor-mediated posttranslational modification of PPAR␣ or ␦ alters its selection of transcriptional targets. 6. PPAR␦-mediated 14-3-3␧ upregulation confers resistance to apoptosis Many genes harbor PPAR response elements (PPRE) in the promoter region. Activated PPAR␦ or PPAR␣ forms heterodimer complex with retinoid X receptor (RXR) [51] which binds to PPRE and activate or repress the expression of diverse classes of genes. Gene expression profiling by microarray assay has revealed a complex set of gene expression regulated by PPAR␣ or PPAR␦ [52]. However, only a few reports have identified relevant genes which are involved in cell survival and anti-apoptosis. Di-Poi et al. reported that PPAR␦ activation in keratinocytes upregulates ILK (integrin-linked kinase) and PDK-1 (phosphoinositide-dependent kinase-1) which activate Akt [53]. Akt protects cell survival by increasing sequestration of the pro-apoptotic Bcl-2 family proteins. To identify downstream target of PPAR␦, we screened a number of anti-apoptotic genes in HUVEC and found that 14-3-3␧ is

Please cite this article in press as: Chu L-y, et al. Prostacyclin protects vascular integrity via PPAR/14-3-3 pathway. Prostaglandins Other Lipid Mediat (2015), http://dx.doi.org/10.1016/j.prostaglandins.2015.04.006

230 231 232 233 234 235 236 237 238

239

240 241 242 243 244 245 246 247 248 249 250 251 252 253 254 255 256 257 258 259 260 261 262 263 264 265 266 267 268 269 270 271 272 273 274

275 276

277 278 279 280 281 282 283 284 285 286 287 288 289 290

G Model PRO 6114 1–9 4 291 292 293 294 295 296 297 298 299 300 301 302 303 304 305 306 307 308 309 310 311 312 313 314 315 316 317 318 319 320 321 322 323 324 325 326 327 328 329 330 331 332 333 334 335 336 337 338 339 340 341 342 343 344 345 346 347 348

ARTICLE IN PRESS L.-y. Chu et al. / Prostaglandins & other Lipid Mediators xxx (2015) xxx–xxx

upregulated by PPAR␦ [30]. 14-3-3␧ is a member of the 14-3-3 family which comprises seven members in mammalian cells. All seven isoforms of 14-3-3 proteins are detected in HUVEC. PGI2 analogs and Ad-COPI selectively upregulate 14-3-3␧ expression [30]. Activation of PPAR␦ with L-165041, a synthetic PPAR␦ agonist, also selectively upregulates 14-3-3␧. Furthermore, PGI2 -induced 143-3␧ upregulation is abolished by silencing of PPAR␦ with siRNA. To confirm that PGI2 upregulates 14-3-3␧ at the transcriptional level through induction of PPAR␦ binding to 14-3-3␧, we searched human 14-3-3␧ promoter region for PPAR␦ binding motifs. Human 14-3-3␧ gene harbors several putative PPREs. We found that PPAR␦-RXR binds to PPRE sites located at -1426 to -1477 of human 14-3-3␧. Deletion of this region abolished PPAR␦-mediated 14-33␧ upregulation without affecting the basal 14-3-3␧ expression [30]. These findings indicate that PGI2 and PPAR␦ agonists activate PPAR␦ which is complexed with RXR and binds to the 14-3-3␧ promoter region to enhance 14-3-3␧ transcription. 14-3-3 proteins function as scaffold proteins to facilitate biochemical reactions, protein interactions or sequestration. The 14-3-3 family proteins share sequence homology, functional characteristics and structural similarities [54]. However, their expressions vary in different cell types which influence their physiological roles. A major function of 14-3-3 proteins is to bind phosphorylated Bad or Bax, sequester them in the cytosol and thereby reduce Bad/Bax-induced apoptosis [55,56]. In HUVEC at basal state, constitutively expressed 14-3-3␧ binds and sequesters Bad in the cytosol with a small amount of Bad detected in mitochondria. H2 O2 treatment results in a large increase of Bad translocation to mitochondria where it triggers apoptosis by perturbing mitochondrial membrane potential. PGI2 is capable of cutting down H2 O2 -induced Bad translocation to mitochondria by enhancing Bad sequestration in cytosol through PPAR␦-mediated 14-3-3␧ upregulation and Akt activation. It appears that selective increase in cytosolic 14-3-3␧ proteins is sufficient to exert a significant augmentation of Bad binding and sequestration in the cytosol. Selective COX-2 inhibitors (coxibs) and a number of nonsteroidal anti-inflammatory drugs (NSAIDs) are associated with cardiovascular complications [57,58]. The exact mechanism by which COX-2 inhibition increases the risk of myocardial infarction has not been clearly elucidated but is thought to be related to shutdown of PGI2 production. Liou et al. reported that NSAIDs such as sulindac induces endothelial apoptosis by inhibiting the expression of PPAR␦ and 14-3-3␧ [59]. Neither PGI2 nor synthetic PPAR␦ agonists can rescue HUVEC because of deficiency of PPAR␦. These results may explain why NSAIDs increase risk of myocardial infarction (MI). Furthermore, they provide additional evidence to support the crucial role that the PPAR␦ → 14-3-3␧ pathway plays in mediating the anti-apoptotic effect of PGI2. The transcriptional pathway via which PGI2 confers resistance to apoptosis in ECs is summarized in Fig. 2. In brief, within the nucleus, PGI2 binds and activates PPAR␦ which forms a heterodimer with RXR. PPAR␦-RXR complex binds to specific sites on 14-3-3␧ promoter to enhance 14-3-3␧ transcription resulting in an increase in cytosolic 14-3-3␧ proteins. H2 O2 triggers Bad mobilization to mitochondria in HUVECs where Bad binds and inactivates Bcl-2 resulting in apoptosis via the mitochondrial pathway. PGI2 suppresses the pro-apoptotic effect of Bad by activation of Akt and upregulation of 14-3-3␧ which increases Bad sequestration in cytosol.

Endothelial Cells Nucleus

Cytosol PGI2 PPARδ

PDK-1 and ILK Akt

PPARδ-RXR

p-Bad

14-3-3ε transcripon 14-3-3ε/Bad Bad sequestraon mitochondria damage apoptosis

Fig. 2. Transcriptional pathways via which PGI2 protects against endothelial cell apoptosis. Dashed line denotes the potential pathway for increased expression of PDK-1 and ILK. Abbreviations: PDK-1, phosphoinositide-dependent kinase-1; ILK, integrin-linked kinase; RXR, retinoid X receptor.

14-3-3 expression and regulation in VSMCs, we analyzed 14-3-3 expression with Western blotting. All the 14-3-3 family proteins except 14-3-3␴ are detected. Ad-PGIS transfection or cPGI2 treatment results in upregulation predominantly of 14-3-3␤ with a lesser increase in 14-3-3␪ and 14-3-3␧ [45]. PPAR␣ agonists such as WY14643 and GW9578 upregulates primarily 14-3-3␤ with a minor increase in 14-3-3␧. Thus, PGI2 and PPAR␣ agonists predominantly increase 14-3-3␤ protein expression. Interestingly, H2 O2 treatment of VSMCs results in 14-3-3␤ degradation by caspase 3 and PGI2 rescues 14-3-3␤ through PPAR␣-induced 14-3-3␤ upregulation. It is surprising that 14-3-3␧ does not play a significant role in the anti-apoptotic action of PGI2 in SMCs. 14-3-3␤ upregulation increases Bad binding and sequestration and consequently retards Bad translocation to mitochondria. H2 O2 -induced caspase 3 in VSMCs is suppressed by 14-3-3␤ overexpression but not 14-3-3␧ or 14-3-3␪ overexpression. Conversely, 14-3-3␤ siRNA abrogates the anti-apoptotic effect of PGI2 . Thus, PGI2 protects VSMCs via a different PPAR → 14-3-3 transcriptional pathway. As illustrated in Fig. 3, H2 O2 induces 14-3-3␤ degradation which weakens Bad sequestration and consequently enhances Bad-induced apoptosis. It is likely that PGI2 -activated PPAR␣ forms heterodimer with RXR and binds to PPAR response elements at the promoter region of 14-3-3␤ to augment 14-3-3␤ expression. Robust 14-3-3␤ expression overwhelms caspase-3 induced 14-33␤ degradation and raises 14-3-3␤ protein levels. 14-3-3␤ binds and sequesters Bad and attenuates Bad-induced apoptosis.

Vascular Smooth Muscle Cells H2O2

PGI2

Caspase 3

PPARα

14-3-3β degradaon

14-3-3β

14-3-3β 349 350

351 352 353

7. PGI2 protects VSMCs from H2 O2 -induced apoptosis by activating PPAR␣ → 14-3-3␤ transcriptional pathway In view of the involvement of PPAR␦ → 14-3-3␧ in regulating EC apoptosis, we were curious whether this pathway is operative in regulating apoptosis in SMCs. Since little was known about

Bad sequestraon Bad-induced apoptosis Fig. 3. PGI2 protects against H2 O2 -induced apoptosis in vascular SMC via PPAR␣/143-3␤.

Please cite this article in press as: Chu L-y, et al. Prostacyclin protects vascular integrity via PPAR/14-3-3 pathway. Prostaglandins Other Lipid Mediat (2015), http://dx.doi.org/10.1016/j.prostaglandins.2015.04.006

354 355 356 357 358 359 360 361 362 363 364 365 366 367 368 369 370 371 372 373 374 375 376 377 378 379

G Model PRO 6114 1–9

ARTICLE IN PRESS L.-y. Chu et al. / Prostaglandins & other Lipid Mediators xxx (2015) xxx–xxx

380 381

382 383 384 385 386 387 388 389 390 391 392 393 394 395 396 397 398 399 400 401 402 403 404 405 406 407 408 409 410 411 412 413 414 415 416 417 418 419 420 421

422 423

424 425 426 427 428 429 430 431 432 433 434 435 436 437 438 439 440 441

8. PGI2 prevents vascular cell apoptosis through multiple mechanisms The mechanisms by which PGI2 defends vascular cells against apoptosis are not restricted to the Akt and the PPAR → 14-3-3 pathways. It was reported that PGI2 increases XIAP proteins in ECs as a result of blocking XIAP ubiquitination and proteasome degradation [60]. XIAP is a member of the IAP family proteins which inhibit caspase 9, caspase 7 and caspase 3. It is well characterized that the cellular XIAP level is tightly regulated by degradation via ubiquitin-proteasome system. Prevention of XIAP degradation by PGI2 preserves XIAP level to defend against apoptosis. PGI2 was reported to protect VSMCs via transforming growth factors ␤ (TGF␤) pathway. Kim et al. reported that PPAR␦ activation by ligand GW501516 attenuates VSMC apoptosis induced by oxidized low-density lipoprotein (oxLDL) by a mechanism involving extracellular matrix (ECM) [61]. They show that activated PPAR␦ binds and activates the promoters of two collagen genes, i.e. COL3A1 and COL1A1, which depends on TGF␤ → Smad 3 pathway. Smad 3 binds to the collagen promoter and together with PPAR␦ increases ECM production. Interestingly, collagen and ECM accumulation plays an essential role in protecting VSMCs from oxLDL and elastase-induced apoptosis, as silencing of COL3A1 or 1A1 abrogates the protective effect of GW501516. Pro-inflammatory cytokines notably tumor necrosis factor ␣ (TNF␣) induce apoptosis and convert EC from a protective phenotype to inflammatory phenotype. Pro-inflammatory mediators trigger oxidative pathways to generate ROS. ROS damage cells and cause apoptosis as well as cell death via necrosis. PPAR␦ agonists were reported to suppress EC inflammatory switch and ROS production. PGI2 may suppress EC ROS production and inflammation via PPAR␦ activation, which further contributes to protection against vascular cell death. Apoptotic cell debris is cleared primarily by macrophages. PPAR␦ was reported to be critical for clearance of apoptotic cells by macrophages [62]. In a murine model with PPAR␦ deletion, macrophages are found to be dysfunctional and defective in clearing apoptosis cells as well as production of pro-inflammatory cytokines. Defect in clearing apoptotic cells leads to accumulation of cell debris which induce immune and inflammatory responses, further aggregating cell damage and apoptosis. PPAR␦ agonists rescue the clearance of apoptotic cells by macrophages and hence avoid the dire consequence of inflammation and cell death.

9. Prostacyclin analogs protect endothelial barrier function via cyclic AMP pathway Vascular endothelium possesses tight intercellular junctions to restrict the passage of proteins, circulating cells and small molecules into the vascular wall. Vascular permeability at the intercellular junctions is highly regulated at the level of adherens junction (AJ) and tight junction. The barrier is one of the fundamental functions of endothelial cells. Disruption of the barrier by environmental insults such as hypoxia, LPS and TNF␣ results in drastic changes in the blood vessel including subendothelial edema, expression of adhesive molecules on EC luminal surface which facilitate transendothelial migration of blood cells and vascular wall inflammation [58]. Severe vascular changes lead to endothelial detachment which exposes the subendothelial tissues to circulating platelets, coagulation factors with consequent thrombosis and intimal hyperplasia. Prostacyclin analogs were reported to enhance endothelial barrier function by upregulating VE-cadherin in a cyclic AMPdependent manner [63]. VE-cadherin is a key constituent of the AJ protein complex. VE-cadherin upregulation increases EC cell

5

LPS / cytokines / Hypoxia

VE-cadherin Vascular permeability

Subendothelial edema

Endothelial Cells

PGI2 cyclic AMP

Endothelial inflammaon

Endothelial detachment

Platelet thrombosis

Inmal inflammaon

Tissue damage, organ infarcon Fig. 4. Schematic illustration of the influence of endothelial cell-derived PGI2 on controlling endothelial permeability, vascular inflammation, endothelial detachment and the consequent tissue damage and organ infarction.

to cell contact and enhances the barrier function. VEGF, LPS and TNF␣ disrupt the barrier function by causing degradation of VEcadherin and thereby increase the permeability to allow passage of blood proteins and cells to the subendothelial region [64]. Iloprost, a PGI2 analog was reported to prevent LPS-induced disintegration of VE-cadherin and preserve the barrier function in endothelial cells by a cAMP-dependent suppression of NF-␬B activation [65]. Hypoxia also causes barrier disruption and increases endothelial permeability through generation of ROS which trigger transendothelial migration of polymorphonuclear cells and inflammation [66]. Inflammation in turn increases endothelial permeability via cytokines such as IL-6 [67]. Continuous hypoxia induces acute endothelial damage as manifested by vascular wall edema, and inflammation which eventually leads to endothelial detachment and denudation. Loss of PGI2 production aggravates the vascular damage. Increase in PGI2 production by transfer of PGI synthase gene protects vascular endothelium and inhibits neointimal formation in animal models [68,69]. The reported data suggest that PGI2 produced by endothelial cells plays an important role in maintaining the physiological state of barrier function and protecting the barrier from disruption by environmental insults and pro-inflammatory cytokines. Continuous exposure to severe insults results in endothelial barrier disruption and the consequent vascular wall edema, expression of pro-inflammatory adhesive molecules on endothelial cells and endothelial detachment. Loss of endothelium and progressive inflammation leads to platelet thrombosis, intimal hyperplasia, and the consequent tissue damage and organ infarction. Supply of PGI2 either by infusion of PGI2 analogs or by gene transfer at an early stage of barrier disruption may rescue the barrier function and prevents acute vascular damage and endothelial detachment via a cyclic AMP-dependent mechanism (Fig. 4). 10. Endothelial damage predisposes heart and brain to ischemia reperfusion injury Ischemia–reperfusion (IR) is a major cause of severe human diseases including MI and ischemic stroke. IR is due to temporary occlusion of an artery such as coronary artery followed by opening of the arterial lumen and reperfusion of myocardial tissues. During the arterial occlusion period, the arterial endothelium suffers from severe hypoxia and ischemia and loses the endothelial barrier function resulting in an increase in vascular permeability

Please cite this article in press as: Chu L-y, et al. Prostacyclin protects vascular integrity via PPAR/14-3-3 pathway. Prostaglandins Other Lipid Mediat (2015), http://dx.doi.org/10.1016/j.prostaglandins.2015.04.006

442 443 444 445 446 447 448 449 450 451 452 453 454 455 456 457 458 459 460 461 462 463 464 465 466 467 468 469 470 471 472 473

474 475

476 477 478 479 480 481 482

G Model

ARTICLE IN PRESS

PRO 6114 1–9 6 483 484 485 486 487 488 489 490 491 492 493 494 495 496 497 498 499 500 501 502 503 504 505 506 507 508 509 510 511 512 513 514 515 516

517 518

519 520 521 522 523 524 525 526 527 528 529 530 531 532 533 534 535 536 537 538 539 540 541 542 543 544 545

L.-y. Chu et al. / Prostaglandins & other Lipid Mediators xxx (2015) xxx–xxx

with the development of subendothelial edema, inflammation and endothelial detachment. Endothelial damage is aggravated by ROS and inflammation produced by reperfusion. Time course experiments in animal models have revealed that endothelial damage preceded myocardial damage following left anterior descending coronary artery occlusion and reperfusion [70–72]. Morphological examination of endothelium with transmission electron microscopy showed evidence of endothelial damage [70,71]. These results suggest that endothelial damage and its consequent loss of vasodilating and vasoprotective eicosanoids during early phase of IR plays an important role in IR-induced MI. Arterial occlusion-reperfusion causes endothelial damage by several possible mechanisms including mechanical stresses due to turbulent blood flow at the occlusion-reperfusion site, generation of reactive oxygen species and pro-inflammatory cytokines. It is possible that endothelial damage leads to EC apoptosis and consequently endothelial denudation. Loss of EC barrier function and protective molecules results in subendothelial inflammation and extracellular matrix accumulation. Pro-inflammatory cytokines and chemokines and growth factors attract SMCs to the intima which contribute to intimal hyperplasia and inflammation. SMC apoptosis due to hypoxia, ROS or inflammation aggravates inflammation and increases SMC proliferation and migration. Robust cytokine/chemokine productions generate systemic inflammation which damages tissues and is thought to be a major pathogenetic factor for post-MI heart failure [73]. Large amounts of ROS are generated during the vicious cycle of inflammation and tissue damage. ROS represents a master trigger of endothelial dysfunction and apoptosis and a key mediator of cellular and tissue damage. Tabernero et al. reported that endothelial and myocardial PPAR␣ are vital for controlling ROS-induced endothelial damage by promoting expression of anti-oxidant enzymes [42]. As PGI2 ligates PPAR␣, it is possible that PGI2 protects endothelium against oxidant-induced damage and endothelial dysfunction by PPAR␣ as well as PPAR␦.

11. PGI2 and 15d-PGJ2 protect against ischemia–reperfusion injury Arterial occlusion followed by reperfusion creates complex biochemical changes which cause damage to the tissues supplied by the involved arteries. IR not only causes tissue infarction but also damages the downstream arterial endothelium resulting in loss of protective molecules including PGI2 [74]. A number of laboratories have reported beneficial effects of stable PGI2 analogs on protecting against cerebral ischemia in animal models [75,76]. Augmentation of PGI2 production by administration of Ad-COPI to a rat stroke model via intracerebral ventricle infusion shows increased brain levels of 6-keto-PGF1␣ (a degradation product of PGI2 ) and suppression of thromboxane, and leukotrienes [77]. It reduced IRinduced cerebral infarct size. These experimental data prove the principle that restoration of PGI2 production is effective in alleviating IR injury. PGI2 protects against IR injury not only by preventing endothelial and neuronal apoptosis but also by control of vasoconstriction and promoting angiogenesis. PGI2 defends against IR-induced EC and tissue damage via several transcriptional pathways. Besides the 14-3-3 upregulation, it was reported that PPAR␦ agonist GW0742 attenuates I/R induced myocardial infarction size by suppressing NF-␬B, COX-2, inducible nitric oxide synthase as well as the glycogen synthase kinase-3␤ (GSK-3␤) pathway [78]. Chen et al. [79] reported that PGI2 protects against I/R-induced renal damage via PPAR␣. They demonstrated that PGI2 reduces NF-␬␤ activation resulting in reduction of TNF␣ and apoptosis [79]. I/R-induced renal damage worsens in PPAR␣ knockout mice whereas PPAR␣ agonists alleviate the I/R renal damage. These findings are consistent with the interpretation that PGI2

is capable of controlling apoptosis and tissue damage by multiple signaling and transcriptional pathways. 15-Deoxy-12,14 PGJ2 (15d-PGJ2 ) is a non-enzymatic degradation product of PGD2 . It binds PPAR␥ and activates PPAR␥ via which it confers anti-inflammatory protection [80]. Lin et al. reported that Ad-COX-1 administration to the rat stroke model increases PGD2 and 15d-PGJ2 which might cooperate with PGI2 to reduce IR-induced brain infarction and neuronal apoptosis [77]. Direct infusion of 15d-PGJ2 reduced the IR-induced cerebral infarction size [81]. Rosiglitazone, a synthetic PPAR␥ agonist protects brain from IR-induced infarction which was abrogated by PPAR␥ siRNA [82]. Proteomic analysis identified 14-3-3␧ as the key target of PPAR␥. Rosiglitazone treatment resulted in more than 5-fold increase in 14-3-3␧ proteins in the rat brain. In vitro studies reveal that rosiglitazone increased 14-3-3␧ protein expression in neuronal cells which confers resistance to apoptosis via binding and sequestering Bad. Thus PPAR␥ activation, like PPAR␦ activation in ECs enhances 14-3-3␧ promoter activity and increases 14-3-3␧ protein expression which contributes to protection against apoptosis. Rosiglitazone was reported to protect mitochondrial membrane potential and upregulate the expression of Bcl-2 which augment the protection against IR-induced brain infarction [83]. These reported data indicate that 15d-PGJ2 protects against IR injury via PPAR␥ and the protective effect of PPAR␥ may be mediated by 143-3 upregulation. 14-3-3 upregulation may be one of the universal mechanism to protect cells from apoptosis. 12. Physiological and therapeutic considerations Endothelial integrity is vital to healthy blood vessels. Disruption of endothelial integrity leads to vascular dysfunction, endothelial cell apoptosis and endothelial detachment which set the stage for serious vascular diseases, such as atherosclerosis and restenosis. Vascular endothelial integrity is protected by multiple molecules whose production is coupled to stress signals. Blood flow shear stress maintains basal vascular homeostasis by stimulating the production of PGI2 , and NO [84,85]. Based on in vitro cell experiments, it has been presumed that endothelium is resilient and able to withstand diverse insults and stresses by rapid responses to the external stress signals resulting in robust production of protective molecules such as PGI2 via induction of COX-2 expression [14]. These presumptions are validated in part by gene transfer experiments but to a large extent unproven. The quantitative and temporal relationship between the severity of environmental stresses and the extent of vasoprotective molecule production remains to be determined. Environmental stresses such as LPS and cytokines alter the EC properties converting it from protective to pro-inflammatory phenotypes. Pro-inflammatory eicosanoids notably leukotrienes and 20-HETE are produced which act against PGI2 . It is unclear how the balance between protective eicosanoids notably PGI2 and possibly EETs (epoxyeicosatrienoic acid) and pro-inflammatory eicosanoids is regulated. The temporal changes in this balance are also not well understood. Understanding of these issues will advance the use of PGI2 stable analogs in prevention and treatment of vascular diseases and tissue damage. PGI2 and its stable prostacyclin analogs were reported to be effective in treating human pulmonary artery hypertension (PAH) in 1980s and remain a mainstay in the treatment of PAH [86–88]. Several PGI2 analogs including iloprost, beraprost, epoprostenol and treprostinil are available for clinical treatment of this serious vascular disease. Their clinical effects are attributed to control of pulmonary arterial SMC proliferation and vascular remodeling [89,90]. By contrast, PGI2 analogs have not been proven to be effective in alleviating other types of vascular diseases [91]. Previous clinical trials on peripheral vascular diseases were designed to treat advanced diseases focusing on demonstration of improvement in

Please cite this article in press as: Chu L-y, et al. Prostacyclin protects vascular integrity via PPAR/14-3-3 pathway. Prostaglandins Other Lipid Mediat (2015), http://dx.doi.org/10.1016/j.prostaglandins.2015.04.006

546 547 548 549 550 551 552 553 554 555 556 557 558 559 560 561 562 563 564 565 566 567 568 569 570 571

572

573 574 575 576 577 578 579 580 581 582 583 584 585 586 587 588 589 590 591 592 593 594 595 596 597 598 599 600 601 602 603 604 605 606 607 608 609

G Model

ARTICLE IN PRESS

PRO 6114 1–9

L.-y. Chu et al. / Prostaglandins & other Lipid Mediators xxx (2015) xxx–xxx

628

blood flow and relief of ischemic symptoms. Given the new findings that PGI2 protects vascular integrity, the therapeutic strategy should be shifted to prevention and early treatment to protect endothelial integrity. Earlier studies with gene transfer of PGI2 synthetic enzymes have provided evidence that enrichment of PGI2 at early arterial injury is effective in preventing arterial thrombosis [92] and attenuating IR-induced cerebral infarction [77] in animal models. The experimental data from animal experiments provide proof of principle but their translation to human therapy is challenging. One strategy is using biomarkers to identify and select patients with evidence of early arterial wall injury for PGI2 gene transfer therapy or chemoprevention. This remains an extremely difficult challenge since there are no reliable biomarkers available. Although gene therapy for hereditary diseases is promising, the application to vascular diseases required additional experiments. Chemoprevention of chronic vascular diseases such as atherosclerosis and restenosis with PGI2 and its analogs has been hampered by lacking efficacious PGI2 analogs that can be taken orally for a long time without adverse effects.

629

13. Conclusion and future perspective

610 611 612 613 614 615 616 617 618 619 620 621 622 623 624 625 626 627

7

endothelial cells via different signaling pathways. It is not entirely clear whether production and actions of those molecules are coordinated. A new therapeutic approach based on combination of the diverse group of vasoprotective molecules should be considered. Conflict of interest We did not receive payment or services at any time from a third party for any aspect of the submitted work. We declare no financial relationships with entities that could be perceived to influence, or that give the appearance of potentially influencing. Author and contributions Ling-Yun Chu: preparing and final approval of the manuscript, Jun-Yang Liou: preparing and final approval of the manuscript and Kenneth K. Wu: drafting, revising and final approval of the manuscript. Acknowledgement

630 631 632 633 634 635 636 637 638 639 640 641 642 643 644 645 646 647 648 649 650 651 652 653 654 655 656 657 658 659 660 661 662 663 664 665 666 667 668 669 670 671 672

Endothelial cells are highly responsive to the environmental insults. When attacked by chemical or physical insults, EC arachidonic metabolism is activated resulting in generation of protective eicosanoids such as PGI2 . PGI2 plays multiple roles in protecting endothelial survival and vascular integrity through interaction with two types of receptors: IP receptors and PPAR (␣ and ␦). PGI2 and several synthetic analogs defend against vascular thrombosis and vasoconstriction via IP receptors while protects EC and SMC from apoptosis primarily via PPAR␦ and PPAR␣. We have identified 143-3 upregulation as an important target of PPAR␣ and ␦. PPARs complex with RXR and bind to PPAR response elements on 143-3␧ in ECs and 14-3-3␤ in SMC to promote the expression of these two types of 14-3-3 isoform proteins in EC and SMC, respectively. Elevated 14-3-3␧ or ␤ facilitates Bad sequestration in the cytosol. Through protecting EC and SMC survival, PGI2 is pivotal in maintaining vascular integrity thereby defending against stressinduced vascular damage. Once endothelium is damaged and ECs are defective in generating the protective molecules such as PGI2 , inflammatory cell accumulation, smooth muscle cell migration and proliferation and lipid deposition occur resulting in development of atherosclerosis, intimal hyperplasia and restenosis. EC damage by ischemia–reperfusion injury contributes to tissue damage and organ infarction. Preservation of EC function and its ability to produce PGI2 is crucial in preventing vascular diseases and I/R-induced organ infarction such as myocardial infarction, and ischemic stroke. There have been numerous attempts to treat vascular diseases and I/R-induced organ infarction with stable PGI2 analogs [86]. Several PGI2 analogs were found to be efficient in treating human pulmonary artery hypertension (PAH) and are now used in clinical treatment of PAH patients. Although the effects of PGI2 analogs are attributed to control VSMC proliferation, preservation of endothelial function is an important attribute. Treatment of other vascular diseases such as peripheral artery disease with PGI2 analogs has not produced consistent results and the adverse effects of the drugs have hampered their clinical use. A new perspective is to target protection of vascular cell survival and vascular integrity. There are major challenges ahead with this approach. We need precise biomarkers to predict early vascular injury and safe oral PGI2 analogs in order to selective appropriate patients for chemoprevention and/or early treatment of vascular diseases and I/R tissue damage. Besides PGI2 , ECs produce nitric oxide, EETs and other metabolites such as 5-methoxytryptophan (5-MTP) [93] which protect

This work is supported in part by grants from Ministry of Science Q4 and Technology, Taiwan (NSC102-2321-B-400-012; 102-2321-B039-009). References [1] Bunting S, Gryglewski R, Moncada S, Vane JR. Arterial walls generate from prostaglandin endoperoxides a substance (prostaglandin X) which relaxes strips of mesenteric and coeliac ateries and inhibits platelet aggregation. Prostaglandins 1976;12:897–913. [2] Whittaker N, Bunting S, Salmon J, Moncada S, Vane JR, Johnson RA, et al. The chemical structure of prostaglandin X (prostacyclin). Prostaglandins 1976;12:915–28. [3] Moncada S, Gryglewski R, Bunting S, Vane JR. An enzyme isolated from arteries transforms prostaglandin endoperoxides to an unstable substance that inhibits platelet aggregation. Nature 1976;263:663–5. [4] Hara S, Miyata A, Yokoyama C, Inoue H, Brugger R, Lottspeich F, et al. Isolation and molecular cloning of prostacyclin synthase from bovine endothelial cells. J Biol Chem 1994;269:19897–903. [5] Pereira B, Wu KK, Wang LH. Molecular cloning and characterization of bovine prostacyclin synthase. Biochem Biophys Res Commun 1994;203:59–66. [6] Hecker M, Ullrich V. On the mechanism of prostacyclin and thromboxane A2 biosynthesis. J Biol Chem 1989;264:141–50. [7] Wu KK, Liou JY. Cellular and molecular biology of prostacyclin synthase. Biochem Biophys Res Commun 2005;338:45–52. [8] Ruan KH, Deng H, Wu J, So SP. The N-terminal membrane anchor domain of the membrane-bound prostacyclin synthase involved in the substrate presentation of the coupling reaction with cyclooxygenase. Arch Biochem Biophys 2005;435:372–81. [9] Baenziger NL, Becherer PR, Majerus PW. Characterization of prostacyclin synthesis in cultured human arterial smooth muscle cells, venous endothelial cells and skin fibroblasts. Cell 1979;16:967–74. [10] Wu KK. Inducible cyclooxygenase and nitric oxide synthase. Adv Pharmacol 1995;33:179–207. [11] Topper JN, Cai J, Falb D, Gimbrone Jr MA. Identification of vascular endothelia genes differentially responsive to fluid mechanical stimuli: cyclooxygenase-2, manganese superoxide dismutase, and endothelial cell nitric oxide synthase are selectively up-regulated by steady laminar shear stress. Proc Natl Acad Sci U S A 1996;93:10417–22. [12] Weksler BB, Ley CW, Jaffe EA. Stimulation of endothelial cell prostacyclin production by thrombin, trypsin, and the ionophore A 23187. J Clin Invest 1978;62:923–30. [13] Shroer K, Zhu Y, Saunders MA, Deng WG, Xu XM, Meyer-Kirchrath, et al. Obligatory role of cyclic adenosine monophosphate response element in cyclooxygenase-2 promoter induction and feedback regulation by inflammatory mediators. Circulation 2002;105:2760–5. [14] Wu KK. Injury-coupled induction of endothelial eNOS and COX-2 genes: a paradigm for thromboresistant gene therapy. Proc Assoc Am Phys 1998;110:163–70. [15] Samuelsson B, Goldyne M, Granström E, Hamberg M, Hammarström S, Malmsten C. Prostaglandins and thromboxanes. Annu Rev Biochem 1978;47:997–1029. [16] Ohashi K, Ruan KH, Kulmacz RJ, Wu KK, Wang LH. Primary structure of human thromboxane synthase determined from the cDNA sequence. J Biol Chem 1992;267:789–93.

Please cite this article in press as: Chu L-y, et al. Prostacyclin protects vascular integrity via PPAR/14-3-3 pathway. Prostaglandins Other Lipid Mediat (2015), http://dx.doi.org/10.1016/j.prostaglandins.2015.04.006

673 674 675 676

677

678 679 680 681

682

683 684 685 686

687

688 689 690

691

692 693 694 695 696 697 698 699 700 701 702 703 704 705 706 707 708 709 710 711 712 713 714 715 716 717 718 719 720 721 722 723 724 725 726 727 728 729 730 731 732 733 734 735 736 737 738 739 740

G Model PRO 6114 1–9 8 741 742 743 744 745 746 747 748 749 750 751 752 753 754 755 756 757 758 759 760 761 762 763 764 765 766 767 768 769 770 771 772 773 774 775 776 777 778 779 780 781 782 783 784 785 786 787 788 789 790 791 792 793 794 795 796 797 798 799 800 801 802 803 804 805 806 807 808 809 810 811 812 813 814 815 816 817 818 819 820 821 822 823 824 825

ARTICLE IN PRESS L.-y. Chu et al. / Prostaglandins & other Lipid Mediators xxx (2015) xxx–xxx

[17] Tanabe T, Yokoyama C, Miyata A, Ihara H, Kosaka T, Suzuki K, et al. Molecular cloning and expression of human thromboxane synthase. J Lipid Mediat 1993;6:139–44. [18] Moncada S, Vane JR. Arachidonic acid metabolites and the interactions between platelets and blood-vessel walls. N Engl J Med 1979;300:1142–7. [19] Cheng Y, Austin SC, Rocca B, Koller BH, Coffman TM, Grosser T, et al. Role of prostacyclin in the cardiovascular response to thromboxane A2. Science 2002;296:539–41. [20] Narumiya S, Sugimoto Y, Ushikubi F. Prostanoid receptors: structures, properties, and functions. Physiol Rev 1999;79:1193–226. [21] Gorman RR, Bunting S, Miller OV. Modulation of human platelet adenylate cyclas by prostacyclin (PGX). Prostaglandins 1977;13:377–88. [22] Rudic RD, Brinster D, Cheng Y, Fries S, Song WL, Austin S, et al. COX-2-derived prostacyclin modulates vascular remodeling. Circ Res 2005;96:1240–7. [23] Falcetti E, Hall SM, Phillips PG, Patel J, Morrell NW, Haworth SG, et al. Smooth muscle proliferation and role of the prostacyclin (IP) receptor in idiopathic pulmonary arterial hypertension. Am J Respir Crit Care Med 2010;182: 1161–70. [24] Murata T, Ushikubi F, Matsuoka T, Hirata M, Yamasaki A, Sugimoto Y, et al. Altered pain perception and inflammatory response in mice lacking prostacyclin receptor. Nature 1997;388:678–82. [25] Adderley SR, Fitzgerald DJ. Oxidative damage of cardiomyocytes is limited by extracellular regulated kinases 1/2-mediated induction of cyclooxygenase-2. J Biol Chem 1999;274:5038–46. [26] Hao CM, Kömhoff M, Guan Y, Redha R, Breyer MD. Selective targeting of cyclooxygenase-2 reveals its role in renal medullary interstitial cell survival. Am J Physiol 1999;277:F352–9. [27] Kersten S, Desvergne B, Wahli W. Roles of PPARs in health and disease. Nature 2000;405:421–4. [28] Kliewer SA, Sundseth SS, Jones SA, Brown PJ, Wisely GB, Koble CS, et al. Fatty acids and eicosanoids regulate gene expression through direct interactions with peroxisome proliferator-activated receptors alpha and gamma. Proc Natl Acad Sci U S A 1997;94:4318–23. [29] Forman BM, Chen J, Evans RM. Hypolipidemic drugs, polyunsaturated fatty acids, and eicosanoids are ligands for peroxisome proliferator-activated receptors alpha and delta. Proc Natl Acad Sci U S A 1997;94:4312–7. [30] Liou JY, Lee S, Ghelani D, Matijevic-Aleksic N, Wu KK. Protection of endothelial survival by peroxisome proliferator-activated receptor-delta mediated 14-3-3 upregulation. Arterioscler Thromb Vasc Biol 2006;26:1481–7. [31] Jiang B, Liang P, Zhang B, Huang X, Xiao X. Enhancement of PPAR-beta activity by repetitive low-grade H2 O2 stress protects human umbilical vein endothelial cells from subsequent oxidative stress-induced apoptosis. Free Radic Biol Med 2009;46:555–63. [32] Nana-Sinkam SP, Lee JD, Sotto-Santiago S, Stearman RS, Keith RL, Choudhury Q, et al. Prostacyclin prevents pulmonary endothelial cell apoptosis induced by cigarette smoke. Am J Respir Crit Care Med 2007;175:676–85. [33] d’Uscio LV, Das P, Santhanam AV, He T, Younkin SG, Katusic ZS. Activation of PPAR␦ prevents endothelial dysfunction induced by overexpression of amyloid-␤ precursor protein. Cardiovasc Res 2012;96:504–12. [34] Katusic ZS, Santhanam AV, He T. Vascular effects of prostacyclin: does activation of PPAR␦ play a role? Trends Pharmacol Sci 2012;33:559–64. [35] Shyue SK, Tsai MJ, Liou JY, Willerson JT, Wu KK. Selective augmentation of prostacyclin production by combined prostacyclin synthase and cyclooxygenase-1 gene transfer. Circulation 2001;103:2090–5. [36] Hsu YH, Chen CH, Hou CC, Sue YM, Cheng CY, Cheng TH, et al. Prostacyclin protects renal tubular cells from gentamicin-induced apoptosis via a PPARalpha-dependent pathway. Kidney Int 2008;73:578–87. [37] Piqueras L, Reynolds AR, Hodivala-Dilke KM, Alfranca A, Redondo JM, Hatae T, et al. Activation of PPARbeta/delta induces endothelial cell proliferation and angiogenesis. Arterioscler Thromb Vasc Biol 2007;27:63–9. [38] Fan Y, Wang Y, Tang Z, Zhang H, Qin X, Zhu Y, et al. Suppression of pro-inflammatory adhesion molecules by PPAR-delta in human vascular endothelial cells. Arterioscler Thromb Vasc Biol 2008;28:315–21. [39] Inoue I, Hayashi K, Yagasaki F, Nakamura K, Matsunaga T, Xu H, et al. Apoptosis of endothelial cells may be mediated by genes of peroxisome proliferator-activated receptor gamma 1 (PPARgamma 1) and PPARalpha genes. J Atheroscler Thromb 2003;10:99–108. [40] Yeh CH, Chen TP, Lee CH, Wu YC, Lin YM, Lin PJ. Cardiomyocytic apoptosis following global cardiac ischemia and reperfusion can be attenuated by peroxisome proliferator-activated receptor alpha but not gamma activators. Shock 2006;26:262–70. [41] Roberts RA, James NH, Woodyatt NJ, Macdonald N, Tugwood JD. Evidence for the suppression of apoptosis by the peroxisome proliferator activated receptor alpha (PPAR alpha). Carcinogenesis 1998;19:43–8. [42] Tabernero A, Schoonjans K, Jesel L, Carpusca I, Auwerx J, Andriantsitohaina R. Activation of the peroxisome proliferator-activated receptor alpha protects against myocardial ischaemic injury improves endothelial vasodilatation. BMC Pharmacol 2002;2:10. [43] Clarke MC, Figg N, Maguire JJ, Davenport AP, Goddard M, Littlewood TD, et al. Apoptosis of vascular smooth muscle cells induces features of plaque vulnerability in atherosclerosis. Nat Med 2006;12:1075–80. [44] Yu H, Clarke MC, Figg N, Littlewood TD, Bennett MR. Smooth muscle cell apoptosis promotes vessel remodeling and repair via activation of cell migration, proliferation, and collagen synthesis. Arterioscler Thromb Vasc Biol 2011;31:2402–9.

[45] Chen YC, Chu LY, Yang SF, Chen HL, Yet SF, Wu KK. Prostacyclin and PPAR␣ agonists control vascular smooth muscle cell apoptosis and phenotypic switch through distinct 14-3-3 isoforms. PLoS ONE 2013;8:e69702. [46] Aubert J, Saint-Marc P, Belmonte N, Dani C, Négrel R, Ailhaud G. Prostacyclin IP receptor up-regulates the early expression of C/EBPbeta and C/EBPdelta in preadipose cells. Mol Cell Endocrinol 2000;160:149–56. [47] Lazennec G, Canaple L, Saugy D, Wahli W. Activation of peroxisome proliferator-activated receptors (PPARs) by their ligands and protein kinase A activators. Mol Endocrinol 2000;14:1962–75. [48] Falcetti E, Flavell DM, Staels B, Tinker A, Haworth SG, Clapp LH. IP receptordependent activation of PPARgamma by stable prostacyclin analogues. Biochem Biophys Res Commun 2007;360:821–7. [49] Fetalvero KM, Shyu M, Nomikos AP, Chiu YF, Wagner RJ, Powell RJ, et al. The prostacyclin receptor induces human vascular smooth muscle cell differentiation via the protein kinase A pathway. Am J Physiol Heart Circ Physiol 2006;290:H1337–46. [50] Kasza Z, Fetalvero KM, Ding M, Wagner RJ, Acs K, Guzman AK, et al. Novel signaling pathways promote a paracrine wave of prostacyclin-induced vascular smooth muscle differentiation. J Mol Cell Cardiol 2009;46:682–94. [51] Plutzky J. The PPAR-RXR transcriptional complex in the vasculature: energy in the balance. Circ Res 2011;108:1002–16. [52] Cherkaoui-Malki M, Meyer K, Cao WQ, Latruffe N, Yeldandi AV, Rao MS, et al. Identification of novel peroxisome proliferator-activated receptor alpha (PPARalpha) target genes in mouse liver using cDNA microarray analysis. Gene Expr 2001;9:291–304. [53] Di-Poï N, Tan NS, Michalik L, Wahli W, Desvergne B. Antiapoptotic role of PPARbeta in keratinocytes via transcriptional control of the Akt1 signaling pathway. Mol Cell 2002;10:721–33. [54] Tzivion G, Avruch J. 14-3-3 proteins: active cofactors in cellular regulation by serine/threonine phosphorylation. J Biol Chem 2002;277:3061–4. [55] Datta SR, Katsov A, Hu L, Petros A, Fesik SW, Yaffe MB, et al. 14-3-3 proteins and survival kinases cooperate to inactivate BAD by BH3 domain phosphorylation. Mol Cell 2000;6:41–51. [56] Masters SC, Yang H, Datta SR, Greenberg ME, Fu H. 14-3-3 inhibits Badinduced cell death through interaction with serine-136. Mol Pharmacol 2001;60:1325–31. [57] Mukherjee D, Nissen SE, Topol EJ. Risk of cardiovascular events associated with selective COX-2 inhibitors. J Am Med Assoc 2001;286:954–9. [58] Kimmel SE, Berlin JA, Reilly M, Jaskowiak J, Kishel L, Chittams J, et al. The effects of nonselective non-aspirin non-steroidal anti-inflammatory medications on the risk of nonfatal myocardial infarction and their interaction with aspirin. J Am Coll Cardiol 2004;43:985–90. [59] Liou JY, Wu CC, Chen BR, Yen LB, Wu KK. Nonsteroidal anti-inflammatory drugs induced endothelial apoptosis by perturbing peroxisome proliferator-activated receptor-delta transcriptional pathway. Mol Pharmacol 2008;74:1399–406. [60] Liou JY, Matijevic-Aleksic N, Lee S, Wu KK. Prostacyclin inhibits endothelial cell XIAP ubiquitination and degradation. J Cell Physiol 2007;212:840. [61] Kim HJ, Kim MY, Jin H, Kim HJ, Kang SS, Kim HJ, et al. Peroxisome proliferatoractivated receptor ␦ regulates extracellular matrix and apoptosis of vascular smooth muscle cells through the activation of transforming growth factor␤1/Smad3. Circ Res 2009;105:16–24. [62] Mukundan L, Odegaard JI, Morel CR, Heredia JE, Mwangi JW, Ricardo-Gonzalez RR, et al. PPAR-delta senses and orchestrates clearance of apoptotic cells to promote tolerance. Nat Med 2009;15:1266–72. [63] Fukuhara S, Sakurai A, Sano H, Yamagishi A, Somekawa S, Takakura N, et al. Cyclic AMP potentiates vascular endothelial cadherin-mediated cell-cell contact to enhance endothelial barrier function through an Epac-Rap1 signaling pathway. Mol Cell Biol 2005;25:136–46. [64] Dejana E, Tournier-Lasserve E, Weinstein BM. The control of vascular integrity by endothelial cell junctions: molecular basis and pathological implications. Dev Cell 2009;16:209–21. [65] Birukova AA, Wu T, Tian Y, Meliton A, Sarich N, Tian X, et al. Iloprost improves endothelial barrier function in lipopolysaccharide-induced lung injury. Eur Respir 2013;41:165–76. [66] Eltzschig HK, Faigle M, Knapp S, Karhausen J, Ibla J, Rosenberger P, et al. Endothelial catabolism of extracellular adenosine during hypoxia: the role of surface adenosine deaminase and CD26. Blood 2006;108:1602–10. [67] Pearlstein DP, Ali MH, Mungai PT, Hynes KL, Gewertz BL, Schumacker PT. Role of mitochondrial oxidant generation in endothelial cell responses to hypoxia. Arterioscler Thromb Vasc Biol 2002;22:566–73. [68] Yamada M, Numaguchi Y, Okumura K, Harada M, Naruse K, Matsui H, et al. Prostacyclin synthase gene transfer modulates cyclooxygenase-2-derived prostanoid synthesis and inhibits neointimal formation in rat ballon-injured arteries. Arterioscler Thromb Vasc Biol 2002;22:256–62. [69] Numaguchi Y, Naruse K, Harada M, Osanai H, Mokuno S, Murase K, et al. Prostacyclin synthase gene transfer accelerates reendothelialization and inhibits neointimal formation in rat carotid arteries after balloon injury. Arterioscler Thromb Vasc Biol 1999;19:727–33. [70] Van Benthuysen KM, McMurtry IF, Horwitz LD. Reperfusion after acute coronary occlusion in dogs impairs endothelium-dependent relaxation to acetylcholine and augments contractile reactivity in vitro. J Clin Invest 1987;79:265–74. [71] Mehta JL, Nichols WW, Donnelly WH, Lawson DL, Saldeen TG. Impaired canine coronary vasodilator response to acetylcholine and bradykinin after occlusionreperfusion. Circ Res 1989;64:43–54.

Please cite this article in press as: Chu L-y, et al. Prostacyclin protects vascular integrity via PPAR/14-3-3 pathway. Prostaglandins Other Lipid Mediat (2015), http://dx.doi.org/10.1016/j.prostaglandins.2015.04.006

826 827 828 829 830 831 832 833 834 835 836 837 838 839 840 841 842 843 844 845 846 847 848 849 850 851 852 853 854 855 856 857 858 859 860 861 862 863 864 865 866 867 868 869 870 871 872 873 874 875 876 877 878 879 880 881 882 883 884 885 886 887 888 889 890 891 892 893 894 895 896 897 898 899 900 901 902 903 904 905 906 907 908 909 910

G Model PRO 6114 1–9

ARTICLE IN PRESS L.-y. Chu et al. / Prostaglandins & other Lipid Mediators xxx (2015) xxx–xxx

911 912 913 914 915 916 917 918 919 920 921 922 923 924 925 926 927 928 929 930 931 932 933 934 935 936 937 938 939 940 941 942 943

[72] Tsao PS, Aoki N, Lefer DJ, Johnson G, Lefer AM. Time course of endothelial dysfunction and myocardial injury during myocardial ischemia and reperfusion in the cat. Circulation 1990;82:1402–12. [73] Frangogiannis NG. Regulation of the inflammatory response in cardiac repair. Circ Res 2012;110:159–73. [74] Collard CD, Gelman S. Pathophysiology, clinical manifestations, and prevention of ischemia–reperfusion injury. Anesthesiology 2001;94:1133–8. [75] Matsuda S, Wen TC, Karasawa Y, Araki H, Otsuka H, Ishihara K, et al. Protective effect of a prostaglandin I2 analog, TEI-7165, on ischemic neuronal damage in gerbils. Brain Res 1997;769:321–8. [76] Martin JF, Hamdy N, Nicholl J, Lewtas N, Bergvall U, Owen P, et al. Doubleblind controlled trial of prostacyclin in cerebral infarction. Stroke 1985;16: 386–90. [77] Lin H, Lin TN, Cheung WM, Nian GM, Tseng PH, Chen SF, et al. Cyclooxygenase1 and bicistronic cyclooxygenase-1/prostacyclin synthase gene transfer protect against ischemic cerebral infarction. Circulation 2002;105: 1962–9. [78] Kapoor A, Collino M, Castiglia S, Fantozzi R, Thiemermann C. Activation of peroxisome proliferator-activated receptor-beta/delta attenuates myocardial ischemia/reperfusion injury in the rat. Shock 2010;34:117–24. [79] Chen HH, Chen TW, Lin H. Prostacyclin-induced peroxisome proliferatoractivated receptor-alpha translocation attenuates NF-kappaB and TNF-alpha activation after renal ischemia–reperfusion injury. Am J Physiol Renal Physiol 2009;297:F1109–18. [80] Ricote M, Li AC, Willson TM, Kelly CJ, Glass CK. The peroxisome proliferatoractivated receptor-gamma is a negative regulator of macrophage activation. Nature 1998;391:79–82. [81] Lin TN, Cheung WM, Wu JS, Chen JJ, Lin H, Chen JJ, et al. 15d-prostaglandin J2 protects brain from ischemia–reperfusion injury. Arterioscler Thromb Vasc Biol 2006;26:481–7. [82] Wu JS, Cheung WM, Tsai YS, Chen YT, Fong WH, Tsai HD, et al. Ligand-activated peroxisome proliferator-activated receptor-gamma protects against ischemic cerebral infarction and neuronal apoptosis by 14-3-3 epsilon upregulation. Circulation 2009;119:1124–34.

9

[83] Wu JS, Lin TN, Wu KK. Rosiglitazone and PPAR-gamma overexpression protect mitochondrial membrane potential and prevent apoptosis by upregulating anti-apoptotic Bcl-2 family proteins. J Cell Physiol 2009;220:58–71. [84] Okahara K, Sun B, Kambayashi J. Upregulation of prostacyclin synthesis-related gene expression by shear stress in vascular endothelial cells. Arterioscler Thromb Vasc Biol 1998;18:1922–6. [85] Tsao PS, Lewis NP, Alpert S, Cooke JP. Exposure to shear stress alters endothelial adhesiveness. Role of nitric oxide. Circulation 1995;92:3513–9. [86] Gomberg-Maitland M, Olschewski H. Prostacyclin therapies for the treatment of pulmonary arterial hypertension. Eur Respir J 2008;31:891–901. [87] Rubin LJ, Mendoza J, Hood M, McGoon M, Barst R, Williams WB, et al. Treatment of primary pulmonary hypertension with continuous intravenous prostacyclin (epoprostenol). Results of a randomized trial. Ann Intern Med 1990;112:485–91. [88] Clapp LH, Finney P, Turcato S, Tran S, Rubin LJ, Tinker A. Differential effects of stable prostacyclin analogs on smooth muscle proliferation and cyclic AMP generation in human pulmonary artery. Am J Respir Cell Mol Biol 2002;26(194):201. [89] Yang J, Li X, Al-Lamki RS, Southwood M, Zhao J, Lever AM, et al. Smad-dependent and smad-independent induction of id1 by prostacyclin analogues inhibits proliferation of pulmonary artery smooth muscle cells in vitro and in vivo. Circ Res 2010;107:252–62. [90] Wharton J, Davie N, Upton PD, Yacoub MH, Polak JM, Morrell NW. Prostacyclin analogues differentially inhibit growth of distal and proximal human pulmonary artery smooth muscle cells. Circulation 2000;102:3130–6. [91] Loosemore TM, Chalmers TC, Dormandy JA. A meta-analysis of randomized placebo control trials in Fontaine stages III and IV peripheral occlusive arterial disease. Int Angiol 1994;13:133–42. [92] Zoldhelyi P, McNatt J, Xu XM, Loose-Mitchell D, Meidell RS, Clubb Jr FJ, et al. Prevention of arterial thrombosis by adenovirus-mediated transfer of cyclooxygenase gene. Circulation 1996;93:10–7. [93] Cheng HH, Kuo CC, Yan JL, Chen HL, Lin WC, Wang KH, et al. Control of cyclooxygenase-2 expression and tumorigenesis by endogenous 5-methoyxtryptophan. Proc Natl Acad Sci U S A 2012;109:13231–6.

Please cite this article in press as: Chu L-y, et al. Prostacyclin protects vascular integrity via PPAR/14-3-3 pathway. Prostaglandins Other Lipid Mediat (2015), http://dx.doi.org/10.1016/j.prostaglandins.2015.04.006

944 945 946 947 948 949 950 951 952 953 954 955 956 957 958 959 960 961 962 963 964 965 966 967 968 969 970 971 972 973 974 975 976 977