当前位置: 首页 > 医学版 > 期刊论文 > 内科学 > 循环研究杂志 > 2005年 > 第3期 > 正文
编号:11257873
Central Role of PKC in Neointimal Expansion Triggered by Acute Arterial Injury
     the Departments of Surgery (M.A., D.B., E.H., Y.S.Z., A.M.S., S.-F.Y.) and Pathology (S.D.Y.), College of Physicians and Surgeons, Columbia University, New York

    Max Planck Institute of Experimental Endocrinology (M.L.), Hannover, Germany

    the Departments of Cardiology (H.A.K.) and Medicine (P.P.N.), University of Heidelberg, Heidelberg, Germany.

    Abstract

    We tested the hypothesis that PKC contributes to vascular smooth muscle cell (SMC) migration and proliferation; processes central to the pathogenesis of restenosis consequent to vascular injury. Homozygous PKC null (eC/eC) mice or wild-type mice fed the PKC inhibitor, ruboxistaurin, displayed significantly decreased neointimal expansion in response to acute femoral artery endothelial denudation injury compared with controls. In vivo and in vitro analyses demonstrated that PKCII is critically linked to SMC activation, at least in part via regulation of ERK1/2 MAP kinase and early growth response-1. These data highlight novel roles for PKC in the SMC response to acute arterial injury and suggest that blockade of PKC may represent a therapeutic strategy to limit restenosis.

    Key Words: arterial injury PKC smooth muscle cell activation inhibitor neointima

    Introduction

    Key evidence for the inducibility of early growth response-1 (Egr-1) in acute arterial injury first emerged from studies in the rat aorta. Egr-1 and a number of its target genes were induced at the wound margins.1eC2 Application of a DNA enzyme that specifically cleaves Egr-1 mRNA in rats and pigs subjected to arterial injury attenuated neointima formation.3eC4 However, these studies did not elucidate the pathway by which Egr-1 was upregulated after vascular injury, nor did they delineate the downstream implications of this molecule in vascular repair.

    Previous studies provided insights into a mechanism by which Egr-1 was upregulated by activation of PKC, particularly the II isoform, in acute hypoxic stress.5eC6 Hypoxia induces rapid activation of PKCII, leading to activation of Raf, mitogen-activated protein kinase/extracellular signal regulated protein kinase kinase and mitogen-activated protein kinases7: events central to PKC-induced transcriptional regulation of Egr-1. In a murine model of single lung ischemia/reperfusion (I/R), PKC null (eC/eC) mice displayed enhanced protection against the adverse effects of I/R compared with Egr-1eC/eC or wild-type mice.8

    Increased activation of the diacylglycerol (DAG)-PKC signal transduction pathway has been identified in vascular tissues retrieved from human subjects and animals with experimental diabetes and in vascular cells incubated in elevated concentrations of glucose.9 Administration of the PKC inhibitor ruboxistaurin (LY333531) to animals with diabetes resulted in significant improvement in microvascular complications in key target organs of diabetes, the retina, kidney, nerve, and heart.10eC12 In this study, we tested the hypothesis that PKCII centrally modulated the response to chronic vascular stress, even in euglycemia. We performed femoral arterial endothelial denudation in mice and show for the first time that PKCII is critically linked to smooth muscle cell (SMC) activation and pathological expansion of the neointima triggered by acute arterial injury in euglycemia, at least in part, via activation of ERK1/2 MAP kinases and regulation of Egr-1.

    Materials and Methods

    Animals and Induction of Vascular Injury

    All procedures were approved by the IACUC at Columbia University. Male C57BL/6 mice (age, 8 to 12 weeks; Jackson Laboratories, Bar Harbor, Maine) were anesthetized by intraperitoneal injection of ketamine (50 mg/kg) and xylazine (5 mg/kg). The femoral artery injury was performed as published.13 Egr-1eC/eC mice,14 a generous gift of Dr Jeffrey Milbrandt (Washington University School of Medicine, St. Louis, Mo), and PKCeC/eC mice,15 were backcrossed 10 generations into C57BL/6. Littermates were used as controls.

    C57BL/6 mice were fed PKC inhibitor ruboxistaurin-containing chow from 3 days before injury to day 7 or 28 after injury. Ruboxistaurin was generously supplied by Dr Louis Vignati (Eli Lilly & Company; Indianapolis, Ind). For BrdU labeling, mice received two intraperitoneal injections of BrdU (2.5 mg/injection; Sigma-Aldrich), 12 hours and 1 hour before euthanasia. Tissues were fixed and processed as described later.

    Histology

    Harvesting of vessel segments was performed as published.13 The section (5 e) at the midportion of each femoral artery was treated with Van Giesson staining kit (Sigma-Aldrich), and the degree of intimal thickening was analyzed quantitatively using a Zeiss microscope and image analysis system (Media Cybernetics Inc). Three types of measurements were made, including luminal area, the area encircled by the internal elastic lamina (IEL) and the area encircled by the external elastic lamina (EEL). All measurements were performed by one of the investigators blinded to the experimental protocol. Intimal area was calculated by subtracting the area encircled by IEL from that encircled by the EEL.

    Representative sections were stained with monoclonal anti-smooth muscle actin IgG (1:100, Sigma-Aldrich). Sections were deparaffinized and blocked with hydrogen peroxide (3%) in methanol for 10 minutes. For BrdU staining, slides were immersed in citrate buffer (0.01 mol/L, pH 6) and microwaved twice for 5 minutes. Blocking was performed with goat serum (4%) and bovine serum albumin (1%) in PBS. Primary antibodies were added to slides and incubated overnight at 4°C. They were incubated with secondary affinity-purified peroxidase-conjugated goat anti-mouse IgG (Sigma).

    SMC replication in the media and intima of arterial segments was evaluated by staining sections with rat monoclonal anti-BrdU IgG (1:100; Sigma). Numbers of stained and total nuclei were counted, and BrdU labeling index was calculated (BrdU-labeled nuclei/total nucleix100%).

    Analysis of Myeloperoxidase Activity

    Two femoral vessel segments per mouse were pooled and homogenized in hexadecyltrimethylammonium bromide followed by three freeze/thaw cycles to release myeloperoxidase from leukocyte granules. Myeloperoxidase activity was measured as described.16

    Western Blotting

    Femoral artery segments were snap frozen in liquid nitrogen, pooled, and stored at eC80°C. Cytosolic and membrane proteins were prepared from six pooled arteries as described.17 To isolate total protein extracts, two pooled arteries, or cultured cells, were homogenized and incubated in ice-cold lysis buffer (Cell Signaling Technology). Lysate protein concentration was determined by Bio-Rad protein assay (Bio-Rad Laboratories). Equal amounts of protein were subjected to SDS-PAGE (7.5% or 12%) followed by electrophoretic transfer to nitrocellulose membranes. Nonspecific binding was blocked by incubation of membranes with nonfat dry milk or BSA (5%) for 1 hour at RT or overnight at 4°C. Blots were incubated with anti-PKCI IgG, anti-PKCII IgG, anti-PKC IgG, anti-PKC IgG, or anti-PKC IgG; and antieCphospho-ERK1/2 IgG, antieCtotal-ERK1/2 IgG, antieCphospho-JNK IgG, or antieCtotal-JNK IgG (Cell Signaling Technology); and antieCphospho-Jak2 IgG (Affinity BioReagents Inc), anti-Jak2 IgG (Santa Cruz Biotechnology Inc), antieCphospho-Stat3 IgG (Cell Signaling Technology), or anti-Stat3 IgG (Santa Cruz Biotechnology Inc); respectively, each at a dilution of 1:1000 for 1 to 3 hours or overnight according to the manufacturer’s instructions. HRP-conjugated donkey anti-rabbit IgG secondary antibody (1:1000, Amersham Biosciences) was used to identify sites of binding of primary antibody.

    RNA Extraction and Real-Time PCR

    Femoral artery segments were snap frozen in liquid nitrogen. Total RNA was extracted from at least four vessel segments using Trizol reagent (Life Technologies Inc). Total RNA (1 e) was processed directly to cDNA synthesis using the TaqMan Reverse Transcription Reagents kit (Applied Biosystem) according to the manufacturer’s protocol. All PCR primers and TaqMan probes were designed using software PrimerExpress (Applied Biosystem) and published sequence data from the NCBI database. The sequences of forward and backward primer for mouse Egr-1 or -actin are 5'-GCCTCGTGAGCATGACCAAT-3' and 5'-GCAGAGGAAGACGATGAAGCA-3'; 5'CCTGAGCGCAAGTACTCTGTGT-3' and 5'-GCTGATCCACATCTGCTGGAA-3'; respectively. The sequence of TaqMan probes for mouse Egr-1 or -actin is 5'-CTCCGACCTCTTCATCCTCGGCG-3' or 5'-CGGTGGCTCCATCTTGGCCTCAC-3'. Primers were synthesized, and TaqMan probes for mouse Egr-1 or -actin were labeled with the reporter dye 6FAM or VIC in the 5' end, and quencher dye TAMRA in both of the 3' end from Applied Biosystem. Primers and probe for 18s rRNA were purchased from Applied Biosystem. All reactions were performed in triplicate in ABI PRISM 7900HT Sequence Detection System; 18sRNA or -actin was used as an endogenous control. Data are calculated by 2eCCT method18 and are presented as the relative proportion induction of mRNA for Egr-1 in injured arteries normalized to 18s rRNA or -actin, compared with uninjured arteries.

    Cell Culture and In Vitro Assays on Cultured SMCs

    Human vascular SMCs (T/G HAVSMC) from American Type Culture Collection (Manassas, Va) were cultured in F12K media with supplements per ATCC protocol. Mouse primary vascular SMCs were cultured from aortas using a modification of the procedure of Tarvo and Barret.19 Experiments were conducted on SMCs after five to eight passages in culture. Cells were >95% SMCs based on SM-actin immunostaining. SMCs were seeded at a density of 2x104 cells/well in 24-well tissue culture-treated plates and incubated in serum-free DMEM for 48 hours. After 60 minutes preincubation with the PKC inhibitor LY379196, cells were exposed to serum-free DMEM containing the prototypic stimulus of PKC, PMA (100 ng/mL), along with [3H]-thymidine (1 e藽i/well, Perkin Elmer). After 12 hours, cells were harvested and cellular proliferation was determined based on the incorporation of [3H]-thymidine. Migration assays were performed as described.20 LY379196 was generously provided by Dr Louis Vignati (Eli Lilly & Company). Northern and Western analysis of transcripts and protein for Egr-1 were performed as described.6

    Statistical Analysis

    All data are expressed as the mean±SEM. All analyses were performed using the Statview Statistical package (version 5.0.1). Values of P<0.05 were considered statistically significant.

    Results

    Effects of Genetic Deletion of PKC on Neointimal Expansion

    To test the premise that PKC, particularly the II isoform, would be activated by acute arterial injury, we assessed membrane localization of distinct PKC isoforms after endothelial denudation in mice. Activation of PKC after denuding injury was rapid; increased PKCII antigen was observed in the membranous fraction of injured femoral arteries compared with sham (7.5 times higher than sham, P<0.001), with a peak at 30 minutes after denuding injury (Figure 1a). In contrast, immunoblotting with an antibody specific to the PKCI isoform showed no change (not shown). By 30 minutes after denudation, no changes in PKC, PKC, and PKC isoforms were detected in the membranous fraction from wild-type and PKCeC/eC mice versus sham (Figure 1b through 1d). Similar findings were observed at 60 minutes and 3 days after arterial denudation (data not shown).

    To elucidate the proximate triggers that led to rapid activation of PKCII, we examined the production of reactive oxygen species (ROS).21 Previous studies established that on denuding arterial injury, polymorphonuclear leukocytes (PMNs) rapidly adhere to the injured wall.13 Consistent with these concepts, by 25 minutes after denuding injury, myeloperoxidase (MPO) activity, an enzyme located in PMNs and a source for generation of ROS, was approximately three times higher in injured wild-type PKC+/+ femoral artery segments versus sham-treated animals (P=0.006; Figure 1e). In PKCeC/eC mice, MPO activity, although significantly higher than that observed in sham-treated vessels in wild-type mice (P=0.008), was not significantly different than that observed in injured wild-type mice (Figure 1e). These data suggest that PKC does not contribute to generation of ROS in arterial injury.

    Based on these findings, we tested the impact of acute arterial injury in homozygous PKCeC/eC mice.15 PKCeC/eC animals displayed significantly lower intima/media (I/M) ratio on day 28 after injury compared with wild-type mice (P<0.0001; Figure 2a, 2c through 2e). In Figure 2d, the adjacent section to that shown in Figure 2c was stained with an antibody to smooth muscle actin, thus identifying that the principal cells forming the expanding neointima in wild-type mice were SMCs. A representative sham-treated vessel (wild-type mice) is shown in Figure 2b.

    Because of the known changes in humoral immune function in PKCeC/eC mice,15 it was important to use a distinct strategy to test the impact of this pathway in acute arterial injury. We used pharmacological inhibition of PKC to suppress its effects in vivo. Selective inhibitors of PKC (ruboxistaurin and LY379196) were tested for their ability to modulate neointimal expansion and SMC properties. In wild-type mice subjected to femoral artery injury, daily administration of ruboxistaurin resulted in significantly less neointimal expansion on day 28 compared with vehicle-treated controls (P=0.02; Figure 2f, 2h, and 2i). Figure 2g depicts a representative sham-treated vessel retrieved from wild-type mice.

    PKC Modulates SMC Proliferation

    To address the mechanisms by which blockade/deletion of PKC resulted in reduced neointimal expansion, we assessed the expanding neointima at an early time after injury, at which point SMC proliferation was previously found to be accelerated.13 Incorporation of Bromodeoxyuridine (BrdU) was significantly decreased in SMCs of the expanding neointima in PKCeC/eC versus wild-type mice on day 7 (Figure 3a, 3c, and 3d) (32% versus 60%; P<0.0001). In wild-type mice subjected to femoral artery injury, administration of ruboxistaurin decreased incorporation of BrdU on day 7 compared with vehicle (Figure 3e, 3g, and 3h) (30% versus 60%; P<0.0001). Figure 3b and 3f indicate that BrdU incorporation in sham-treated vessels was essentially not detected.

    Downstream Targets of PKC and the Role of Egr-1 in SMCs

    Egr-1 was previously found to be regulated, at least in part, by PKCII in acute hypoxic stress. On acute femoral artery endothelial denudation in C57BL/6 mice, a time-dependent increase in mRNA transcripts encoding Egr-1 was found, with a peak 1 to 2 hours after injury (Figure 4a). By 3 and 6 hours, transcripts for Egr-1 had significantly declined (Figure 4a).

    To definitively assess the role of Egr-1 in the response to vessel injury, we used mice deficient in Egr-1 (Egr-1eC/eC). Compared with wild-type mice, Egr-1eC/eC mice displayed significantly less neointimal expansion on day 28 after denuding injury (Figure 4b, 4d, and 4e). Figure 4c represents sham-treated wild-type vessels.

    Consistent with an essential role for PKC in regulation of Egr-1 in arterial injury, arteries retrieved from PKCeC/eC mice displayed significantly lower transcript levels for Egr-1 compared with wild-type mice (Figure 4f). No differences in Egr-1 transcripts were observed at baseline between the two groups of mice (Figure 4f). Furthermore, immunohistochemistry revealed that the principal Egr-1eCexpressing cells after acute arterial injury were SMCs (Figure 4h), as demonstrated by colocalization with anti-smooth muscle actin IgG (Figure 4i). In contrast, sham control arteries from wild-type (Figure 4g), PKCeC/eC (Figure 4j) mice, and injured arteries from PKCeC/eC mice (Figure 4k) did not express detectable levels of Egr-1 antigen. Immunohistochemistry at multiple time points for detection of mononuclear phagocytes, using rat anti-mouse anti-F4/80 IgG, failed to identify significant numbers of these cells (not shown), consistent with published studies.13

    PKCII-Mediated SMC Activation: In Vitro Analyses

    To delineate the mechanisms underlying the impact of PKCII on regulation of Egr-1 after arterial injury, we studied primary cultures of SMCs and used a prototypic stimulus for PKC, phorbol myristate acetate (PMA). The PKC inhibitor LY379196 was used to inhibit the effect of this PKC isoform. At concentrations less than 600 nmol/L, this inhibitor is selective for the PKCI and II isoforms.22

    Both transcripts and protein for Egr-1 were lower in primary cultures of human aortic SMCs exposed to PMA, 100 ng/mL in the presence of the PKC inhibitor LY379196 (Figure 5a and 5b, respectively). Similarly, in primary cultures of murine SMCs, PMA-triggered upregulation of transcripts for Egr-1 was suppressed by pretreatment with LY379196 (Figure 5c).

    Next, we examined the impact of PMA/PKC on two central functional properties of SMCs, proliferation and migration. In cultured human aortic SMCs, incubation with PMA resulted in increased incorporation of tritiated thymidine (P<0.0001), a process suppressed by LY379196 (Figure 5d). Incubation of primary murine aortic SMCs with PMA triggered increased incorporation of tritiated thymidine (P<0.0001), in a manner reduced by LY379196 (Figure 5e). Further, we studied the role of PKC in mediating cellular migration, a key property of SMCs in the expanding neointima after injury. In modified Boyden chambers, addition of PMA to the lower compartment significantly increased the number of migrating primary murine aortic SMCs (P<0.0001), a process significantly suppressed by LY379196 (Figure 5f).

    These findings established that Egr-1 was a downstream target of PKC in acute arterial injury and that proliferation and migration of SMCs were modulated, at least in part, via PKC.

    Signal Transduction Pathways Mediating the Impact of PKC in Acute Arterial Injury: In Vivo Analyses

    Previous studies linked a range of signaling mechanisms, including the mitogen-activated (MAP) kinase pathway, especially extracellular signal-regulated protein kinase (ERK1/2), Janus kinase (Jak) 2, and signal transducer and activator of transcription (Stat) 3, to the response to arterial injury in SMCs.23eC24 Homogenates of injured arteries harvested 15 minutes after denudation revealed markedly increased phosphorylated ERK1/2 and phospho-JNK compared with sham-treated arterial segments in wild-type mice (P<0.0001; Figure 6a and 6b). In contrast, PKCeC/eC mice displayed only a small increase in phosphorylation of ERK1/2 and JNK above sham (P<0.0001; Figure 6a and 6b).

    In addition, we tested the potential role of PKC on the Jak2/Stat3 pathway on day 7 after injury. Although phospho-Jak2 and phospho-Stat3 were significantly increased in injured versus sham mice on day 7 (Figure 6c and 6d), there were no significant differences between phospho-Jak2 and phospho-Stat3 between wild-type versus PKCeC/eC mice after injury. No differences in phospho-Jak2 or phospho-Stat3 were observed between injured versus sham-treated arteries at 24 hours, 3 days, or 5 days after denudation (not shown).

    Signal Transduction Pathways Mediating the Impact of PKC in SMCs: In Vitro Analyses

    As it is not feasible to chronically administer inhibitors of ERK 1/2 and JNK MAP kinases in vivo, we dissected these pathways in vitro using primary cultures of murine aortic SMCs. Incubation of primary murine SMCs with PMA (100 ng/mL) for 15 minutes resulted in a striking increase in phosphorylation of ERK1/2 and JNK (Figure 6e and 6f). Real-time quantitative PCR analysis of RNA from mouse aortic SMCs incubated with PMA demonstrated an 7.7 times increase in expression of Egr-1 transcripts compared with untreated SMCs, in a manner suppressed by PD98059, but not by the JNK inhibitor SP600125 (Figure 6g). These findings suggested that PKC-mediated regulation of Egr-1 was due, at least in part, to phosphorylation of ERK1/2, but not via phosphorylation of JNK.

    Lastly, we examined the impact of PKC on proliferation of murine aortic SMCs. Although PMA caused a significant increase in tritiated thymidine incorporation in wild-type SMCs, pretreatment with PD98059 strikingly suppressed this effect, whereas SP600125 caused a statistically significant attenuation in proliferation, albeit to a degree less than that observed by blockade of ERK1/2 MAP kinase (Figure 6h).

    Discussion

    These findings reinforce the concept that pathways leading to restenosis are programmed by key alterations in the vessel wall that occur within minutes to hours after the initial insult. Activation of PKC is underway within the first 15 minutes of arterial injury. A key question that arises is by what biochemical/molecular mechanism does acute arterial injury cause rapid activation of PKC Although previous work suggested that monocyte recruitment to the vessel wall is not a prominent feature of arterial injury in C57BL/6 mice, it has been shown that polymorphonuclear leukocytes are rapidly recruited to the injured vessel.13 One consequence of neutrophil activation is generation of oxidant stress, in part by myeloperoxidase (MPO) activity.25 Our studies suggest that increased MPO activity is measurable rapidly after arterial injury compared with sham controls in C57BL/6 mice and provides a mechanism by which PKC, and especially the isoform, may be activated by acute endothelial denudation. Specifically, one consequence of MPO activity, generation of advanced glycation endproducts (AGEs), has been linked to PKC activation.26eC27 These considerations highlight the concept that arterial injury acutely perturbs critical biochemical pathways leading to rapid recruitment of signaling pathways that significantly modify the vascular milieu.

    Previous studies have linked the PKC isoform to stimulation of SMCs. In those studies, however, the specific stimuli to activation of PKC were distinct from those studied here. In vitro, Li and colleagues28 showed that mechanical stress induced SMC migration in cell culture, in a pathways dependent on PKC. In vivo, Leitges and colleagues29 showed that vein graft arteriosclerosis was significantly attenuated in homozygous PKCeC/eC mice versus littermates. We conclude that these stimuli are distinct from those studied in the present work, as our data showed clearly that there was no evidence of activation of PKC triggered by endothelial denudation in the murine femoral artery. These considerations highlight novel and intricate means of regulation of SMC properties that are dependent, at least in part, on an array of PKC isoforms and single subtypes.

    We previously reported that hypoxia/reoxygenation (H/R) treatment of alveolar mononuclear phagocytes triggered rapid activation of PKC; downstream consequences of PKC activation in these cells included upregulation of Egr-1 transcripts.8 Pretreatment of the alveolar mononuclear phagocytes with inhibitors of PKC, ERK1/2 MAP kinase, and JNK MAP kinase before H/R suppressed upregulation of Egr-1.8 In the present studies, however, pretreatment of SMCs with inhibitors of PKC and ERK1/2 MAP kinase before exposure to PMA attenuated upregulation of Egr-1. Inhibitors of JNK MAP kinase did not affect Egr-1 regulation. These findings underscore the concept that the impact of PKC may vary in distinct cell types and in response to different stimuli. Although the in vitro stimulus to activation of PKC used here, PMA, is not specific for activation of PKC isoform, the same is true of other stimuli used to trigger activation of this enzyme, such as high concentrations of glucose in cell culture medium. Thus, experiments using LY379196 and PKC null mice in the present work confirm the central importance of the PKC pathway in triggering neointimal expansion stimulated by endothelial denudation.

    In terms of clinical translation, our data highlight the potentially critical difference between strategies such as sirolimus-coated stents versus blockade of PKC in restenosis. Certainly, sirolimus-coated stents demonstrated striking efficacy in reduction of restenosis,30eC31 yet the beneficial effect may not be complete at least in certain subsets of subjects.32 The recent observations that parenterally administered sirolimus provided only marginal benefit, even at high doses, strongly suggested that more effective targets for untoward neointimal expansion in restenosis and atherosclerosis will be needed in the future.33eC34

    Taken together, these findings highlight pivotal roles for PKC in the SMC response to acute arterial injury and the development of pathological neointimal expansion.

    Acknowledgments

    The authors gratefully acknowledge support from the LeDucq Foundation, Surgical Research Fund of Columbia University, Burroughs Wellcome Fund, and grants from the USPHS, National Institutes of Health (RO1 HL075529eC01). We thank Dr Jeffrey Milbrandt for the generous gift of Egr-1eC/eC mice.

    References

    Khachigian L, Lindner V, Williams A, Collins T. Egr-1 induced endothelial gene expression: a common theme in vascular injury. Science. 1996; 271: 1427eC1431.

    Santiago F, Lowe H, Day F, Chesterman C, Khachigian L. Egr-1 induction by injury is triggered by release and paracrine activation by FGF-2. Am J Pathol. 1999; 154: 937eC944.

    Santiago FS, Lowe HC, Kavurma MM, Chesterman CN, Baker A, Atkins DG, Khachigian LM. New DNA enzyme targeting Egr-1 mRNA inhibits vascular smooth muscle proliferation and regrowth after injury. Nat Med. 1999; 5: 1264eC1269.

    Lowe HC, Fahmy RG, Kavurma MM, Baker A, Chesterman CN, Khachigian LM. Catalytic oligodeoxynucleotides define a key regulatory role for early growth response-factor 1 in the porcine model of coronary in stent restenosis. Circ Res. 2001; 89: 670eC677.

    Yan SF, Lu J, Zou YS, Soh-Won J, Cohen DM, Buttrick PM, Cooper DR, Steinberg SF, Mackman N, Pinsky DJ, Stern DM. Hypoxia-associated induction of early growth response-1 gene expression. J Biol Chem. 1999; 274: 15030eC15040.

    Yan SF, Fujita T, Lu J, Okada K, Zou YS, Mackman N, Pinsky DJ, Stern DM. Egr-1, a master switch coordinating upregulation of divergent gene families underlying ischemic stress. Nat Med. 2000; 6: 1355eC1361.

    Yan SF, Lu J, Zou YS, Kisiel W, Mackman N, Leitges M, Steinberg S, Pinsky D, Stern D. Protein kinase C and oxygen deprivation. J Biol Chem. 2000; 275: 11921eC11928.

    Fujita T, Asai T, Andrassy M, Stern DM, Pinsky DJ, Zou YS, Okada M, Naka Y, Schmidt AM, Yan SF. PKC regulates ischemia/reperfusion injury in the lung. J Clin Invest. 2004; 113: 1615eC1623.

    Way KJ, Natai N, King GL. Protein kinase C and the development of diabetic vascular complications. Diabetes Med. 2001; 18: 945eC959.

    Danis RP, Bingaman DP, Jirousek M, Yang Y. Inhibition of intraocular neovascularization caused by retinal ischemia in pigs by PKC inhibition with LY333531. Invest Ophthalmol Vis Sci. 1998; 39: 171eC179.

    Koya D, Haneda M, Nakagawa H, Isshiki K, Sato H, Maeda S, Sugimoto T, Kashiwagi A, Ways DK, King GL, Kikkawa R. Amelioration of accelerated diabetic mesangial expansion by treatment with a PKC beta inhibitor in diabetic db/db mice, a rodent model for type 2 diabetes. FASEB J. 2000; 14: 439eC447.

    Nakamura J, Kato K, Hamada Y, Nakayama M, Chaya S, Nakashima E, Naruse K, Kasuya Y, Mizubayashi R, Miwa K, Yasuda Y, Kamiya H, Ienaga K, Sakakibara F, Koh N, Hotta N. A protein kinase C beta selective inhibitor ameliorates neural dysfunction in streptozotocin-induced diabetic rats. Diabetes. 1999; 48: 2090eC2095.

    Roque M, Fallon JT, Badimon JJ, Zhang WX, Taubman MB, Reis ED. Mouse model of femoral artery denudation injury associated with the rapid accumulation of adhesion molecules on the luminal surface and recruitment of neutrophils. Arterioscler Thromb Vasc Biol. 2000; 20: 335eC342.

    Lee SL, Wang Y, Milbrandt J. Unimpaired macrophage differentiation and activation in mice lacking the zinc finger transplantation factor NGFI-A (EGR1). Mol Cell Biol. 1996; 16: 4566eC4572.

    Leitges M, Schmedt C, Guinamard R, Davoust J, Schaal S, Stabel S, Tarakhovsky A. Immunodeficiency in protein kinase C deficient mice. Science. 1996; 273: 788eC791.

    Goldblum S, Wu K, Jay M. Lung myeloperoxidase as a measure of pulmonary leukostasis in rabbits. J App Physiol. 1985; 59: 1978eC1985.

    Goldberg M, Zhang HL, Steinberg SF. Hypoxia alters the subcellular distribution of protein kinase C isoforms in neonatal rat ventricular myocytes. J Clin Invest. 1997; 99: 55eC61.

    Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2eCCT method. Methods. 2001; 25: 402eC408.

    Tarvo P, Barret G. Differences in proliferation of primary cultures of vascular smooth muscle cells taken from male abd female rats. Blood Vessels. 1980; 17: 110eC116.

    Sakaguchi T, Yan SF, Yan SD, Belov D, Rong LL, Sousa M, Andrassy M, Marso SP, Duda S, Arnold B, Liliensiek B, Nawroth PP, Stern DM, Schmidt AM, Naka Y. Centeral role of RAGE-dependent neointimal expansion in arterial restenosis. J Clin Invest. 2003; 111: 959eC972.

    Scivittaro V, Ganz MB, Weiss MF. AGEs induce oxidative stress and activate protein kinase C II in neonatal mesangial cells. Am J Physiol Renal Physiol. 2000; 278: F676eCF683.

    Slosberg ED, Yao Y, Xing F, Ikui A, Jirousek MR, Weinstein IB. The protein kinase C -specific inhibitor LY379196 blocks TPA-induced monocytic differentiation of HL60 cells. Mol Carcinog. 2000; 27: 166eC176.

    Koyama H, Olson N, Dastva F, Reidy M. Cell replication in the arterial wall. Circ Res. 1998; 82: 713eC721.

    Seki Y, Shibata R, Nagata T, Yasukawa H, Yoshimura A, Imaizumi T. Role of JAK/STAT pathway in rat carotid artery remodeling after vascular injury. Circ Res. 2000; 87: 12eC18.

    Jansson G. Oestrogen-induced enhancement of myeloperoxidase activity in polymorphonuclear leukocytes: a possible cause of oxidative stress in inflammatory cells. Free Radic Res Commun. 1991; 14: 195eC208.

    Anderson MM, Requena JR, Crowley JR, Thorpe SR, Heinecke J. The myeloperoxidase system of human phagocytes generates Nepsilon-(carboxymethyl)lysine on proteins: a mechanism for producing advanced glycation endproducts at sites of inflammation. J Clin Invest. 1999; 104: 103eC113.

    Ido Y, Chang KC, Lejeune WS, Bjercke RJ, Reiser KM, Williamson JR, Tilton RG. Vascular dysfunction induced by AGE is mediated by VEGF via mechanisms involving reactive oxygen species, guanylate cyclase and protein kinase C. Microcirculation. 2001; 8: 251eC263.

    Li C, Wernig F, Leitges M, Hu Y, Xu Q. Mechanical stress-activated PKC delta regulates smooth muscle migration. FASEB J. 2003; 17: 2106eC2108.

    Leitges M, Mayr M, Braun U, Mayr U, Li C, Pfister G, Ghafari-Tabrizi Baier G, Hu Y, Xu Q. Exacerbated vein graft arteriosclerosis in protein kinase C delta mice. J Clin Invest. 2001; 108: 1505eC1512.

    Morice MC, Serruys PW, Sousa JE, Fajadet J, Ban Hayashi E, Perin M, Colombo A, Schuler G, Barragan P, Guagliumi G, Molnar F, Falotico R; RAVEL Study Group. Randomized study with the sirolimus-coated Bx velocity balloon-expandable stent in the treatment of patients with de novo native coronary artery lesions. N Engl J Med. 2002; 346: 1773eC1780.

    Moses JW, Leon MB, Popma JJ, Fitzgerald PJ, Holmes DR, O’Shaughnessy C, Caputo RP, Kereiakes DJ, Williams DO, Teirstein PS, Jaeger JL, Kuntz RE; SIRIUS Investigators. Sirolimus-eluting stents versus standard stents in patients with stenosis in a native coronary artery. N Engl J Med. 2003; 349: 1315eC1323.

    Lemos PA, Saia F, Ligthart JM, Arampatzis CA, Sianos G, Tanabe K, Hoye A, Degertekin M, Daemen J, McFadden E, Hofma S, Smits PC, de Feyter P, van der Giessen WJ, van Domburg RT, Serruys PW. Coronary restenosis after sirolimus-eluting stent implantation: morphological description and mechanistic analysis from a consecutive series of cases. Circulation. 2003; 108: 257eC260.

    Brara PS, Moussavian M, Grise MA, Reilly JP, Fernandez M, Schatz RA, Teirstein PS. Pilot trial of oral rapamycin for recalcitrant restenosis. Circulation. 2003; 107: 1722eC1724.

    Rodriguez AE, Alemparte MR, Vigo CF, Pereira CF, Llaurado C, Russo M, Virmani R, Ambrose JA. Pilot study of oral rapamycin to prevent restenosis in patients undergoing coronary stent therapy: Argentina Single-Center Study (ORAR Trial). J Invasive Cardiol. 2003; 15: 581eC584.(Martin Andrassy, Dmitry B)