Literature
Home医源资料库在线期刊动脉硬化血栓血管生物学杂志2004年第24卷第1期

Thrombin and Phenotypic Modulation of the Endothelium

来源:动脉硬化血栓血管生物学杂志
摘要:eduAbstractThrombinsignalingintheendotheliumislinkedtomultiplephenotypicchanges,includingalterationsinpermeability,vasomotortone,andleukocytetrafficking。ThrombinandEndothelialCellPhenotypesThrombinisamultifunctionalserineproteasethatisinvolvednotonlyinmediati......

点击显示 收起

From the Research Center for Advanced Science and Technology (T.M., A.S., T.K.), University of Tokyo, Tokyo, Japan, and the Division of Molecular and Vascular Medicine (S.W., R.A., W.C.A.), Beth Israel–Deaconess Medical Center/Harvard Medical School, Boston, Mass.

Correspondence to William C. Aird, Beth Israel Deaconess Medical Center, RW-663, Boston, MA 02215. E-mail waird@bidmc.harvard.edu

    Abstract

Thrombin signaling in the endothelium is linked to multiple phenotypic changes, including alterations in permeability, vasomotor tone, and leukocyte trafficking. The thrombin signal is transduced, at least in part, at the level of gene transcription. In this review, we focus on the role of thrombin signaling and transcriptional networks in mediating downstream gene expression and endothelial phenotype. In addition, we report the results of DNA microarrays in control and thrombin-treated endothelial cells. We conclude that (1) thrombin induces the upregulation and downregulation of multiple genes in the endothelium, (2) thrombin-mediated gene expression involves a multitude of transcription factors, and (3) future breakthroughs in the field will depend on a better understanding of the spatial and temporal dynamics of these transcriptional networks.

Key Words: thrombin ? transcription ? DNA microarrays ? endothelial cells

    Introduction

Vascular diseases are among the most common causes of morbidity and mortality in the Western world. A remarkable feature of these disorders is the focal nature of their distribution. Indeed, there does not exist a single disease state that affects virtually every blood vessel type in the body. An important goal in vascular biology is to delineate mechanisms of vascular bed–specific pathology. One clue to these patterns is found within the endothelium. The endothelium serves as a "barometer" of the microenvironment, constantly sensing changes within the extracellular compartment and responding in ways that are usually beneficial, but sometimes harmful, to the host. As an extension of this model, the endothelium has been likened to a circuit board, 1 that is hard-wired to meet the demands of the underlying tissue and 1 that is highly vulnerable to "short-circuiting" as a mechanism of focal, vasculopathic disease states.1 The overall goal of this review is to explore thrombin signaling as a paradigm of endothelial cell circuitry. To that end, we will summarize the published literature, provide new insights into thrombin-associated transcriptional networks, and propose ways in which to integrate this information into the spatial and temporal context of the endothelium.

    Thrombin and Endothelial Cell Phenotypes

Thrombin is a multifunctional serine protease that is involved not only in mediating the cleavage of fibrinogen to fibrin in the coagulation cascade but also in activating a variety of cell types, including platelets and endothelial cells. Thrombin signaling in the endothelium might result in a multitude of phenotypic changes, including alterations in cell shape, permeability, vasomotor tone, leukocyte trafficking, migration, DNA synthesis, angiogenesis, and hemostasis (Table 1).

   TABLE 1. Thrombin and Endothelial Cell Phenotypes

Thrombin Signaling in Endothelial Cells

Thrombin signaling in the endothelium is mediated by a family of 7-transmembrane G protein–coupled receptors, termed protease-activated receptors (PARs).2 Currently, 4 members of the PAR family have been identified (PAR-1 through PAR-4; Table 2). PAR-1 and PAR-3 are thrombin receptors.3,4 Thrombin activation of PAR-4 requires PAR-3 as a thrombin-binding cofactor.5 Human umbilical vein endothelial cells (HUVECs) have been reported to express PAR-1, PAR-2, and, to a lesser extent, PAR-3, but not PAR-4.6,7 One study provided evidence for the existence of functional PAR-4 receptors (as well as those for PAR-1 and PAR-2 but not PAR-3) in the endothelium of human coronary artery ring segments.8 Of the various PAR family members, PAR-1 is the predominant thrombin receptor in endothelial cells.6 Thrombin activates PAR-1 by binding to a unique site in the extracellular domain of the receptor, resulting in cleavage between Arg41 and Ser42 and consequent exposure of a new N-terminus. The unmasked tethered ligand (SFLLRN) interacts with the extracellular loop 2 of the receptor (amino acids 248 to 268), resulting in receptor activation.9

   TABLE 2. Thrombin Receptors in Endothelial Cells (ECs)

Once activated, PAR-1 is coupled to a family of heterotrimeric G proteins, consisting of an -subunit and a ?-dimer. The G proteins are in turn linked to a number of signal intermediates that include, but are not limited to, mitogen-activated protein kinase (MAPK), protein kinase C (PKC), phosphatidyl inositol 3-kinase (PI3K), and Akt (Table 3). Thrombin signaling might result in posttranscriptional changes, including calcium influx, cytoskeletal reorganization, and release of soluble mediators, growth factors, and matrix metalloproteinases (Table 4). In addition, thrombin signaling results in changes in downstream gene transcription (Table 5). For example, under in vitro conditions, thrombin has been shown to increase the expression of genes that are involved in cell proliferation, inflammation, leukocyte adhesion, vasomotor tone, and hemostasis (see Table 5 for references).

   TABLE 3. Thrombin Signaling in Endothelial Cells

   TABLE 4. Thrombin and Posttranscriptional Changes in Endothelial Cells

   TABLE 5. Thrombin and Transcriptional Changes in Endothelial Cells (Previously Published)

Uncovering Thrombin-Responsive Gene Programs

Most published studies have reported the effect of thrombin on specific target genes or clusters of genes. To more systematically catalog thrombin-responsive gene programs, we carried out DNA microarray experiments in control and thrombin-treated endothelial cells (please see online Figure I at http://atvb.ahajournals.org). In these experiments, HUVECs at passage 3 were serum-starved for 12 hours in medium containing 0.5% fetal bovine serum and then treated with 1.5 U/mL thrombin for varying periods of time. All microarray studies were carried out in duplicate with HUVECs from different donors. Only those genes that changed 2-fold in both experiments were considered significant. Several important themes emerged from these studies. First, although thrombin increased the expression of certain genes (74 of 8794 genes), thrombin signaling resulted in the downregulation of other transcripts (20 of 8794). Second, our studies revealed a time-dependent effect of thrombin on gene transcription. For example, of the genes that were induced by thrombin, 34 peaked at 1 hour, 21 at 4 hours, and 19 at 18 hours. It is noteworthy that 12 of the 34 thrombin-responsive genes that peaked at 1 hour were transcription factors (online Figure I; names denoted in red), compared with only 4 of 21 and 0 of 19 genes peaking at 4 and 18 hours, respectively. These results suggest that thrombin induces an early, temporally regulated transcriptional cascade. Third, the microarray results confirmed most, but not all, previous reports of thrombin-mediated gene expression. For example, the data support the results of a previous study that showed thrombin-mediated downregulation of endothelial nitric oxide synthase (eNOS) mRNA.10 Moreover, the microarray experiments demonstrated increased levels of angiopoeitin-2,11 platelet-derived growth factor (PDGF)-A,12 interleukin (IL)-8,13 intracellular adhesion molecule-1 (ICAM-1),14 vascular cell adhesion molecule-1 (VCAM-1),15 E-selectin,16 decay accelerating factor (DAF),17 early growth-response factor (Egr)-1,18 plasminogen activator inhibitor-1 (PAI-1),19 IL-6,13 monocyte chemoattractant protein-1 (MCP-1),13 and cyclooxygenase-2 (COX-2).20 Consistent with the published literature, the induction of Egr-1 was early18 and that of DAF, late.17 However, in contrast to previous reports,10,21–26 thrombin failed to induce Flk-1/KDR, Flt-1, endothelin-1, tissue factor (TF), endothelial protein C receptor (EPCR), and endothelin-converting enzyme-1, despite the use of duplicate microarray samples across multiple time points. Not all studies have shown a link between thrombin signaling and Flt-1 expression.21 Moreover, thrombin-mediated induction of Flk-1/KDR has been shown to require the presence of vascular endothelial growth factor (VEGF).21 Thrombin was reported to increase EPCR mRNA levels and/or EPCR promoter activity in rat and bovine aortic endothelial cells.25,26 The absence of EPCR induction in our studies might reflect differences in species and or vascular bed of origin. The discrepancy in TF response is more difficult to explain. Using ribonuclease protection assays, we have found that thrombin induces a high level of TF mRNA in multiple subtypes of human endothelial cells (including those derived from umbilical vein, pulmonary artery, and coronary artery), with maximal levels (>6-fold) occurring at 2 hours (Figure 1). Along with the published data, these latter findings suggest that the TF results in the microarray experiments represent a false-negative.

   Figure 1. Ribonuclease protection assay of thrombin-treated endothelial cells. Endothelial cells were grown on 6-well plates and starved overnight in EBM-2 medium containing 0.5% fetal bovine serum, as described in the online supplement (please see http://atvb.ahajournals.org). The cells were treated in the absence or presence of 1.5 U/mL thrombin for the times indicated and were subsequently harvested for total RNA. RNase protection assays were carried out with [-32P]UTP-labeled TF, Egr-1, ICAM-1, urokinase-type plasminogen activator and ?-actin probes, as described in the online supplement. Results are representative of 3 independent experiments and were virtually identical in human pulmonary artery endothelial cells and HUVECs.

Finally, thrombin stimulated the expression of certain genes that have not previously been reported to be thrombin-responsive. Although the results will require validation by real-time polymerase chain reaction and/or RNase protection assays, the data reveal a number of interesting candidate genes, including CBP/p300-interacting transactivator with ED-rich tail 2 (CITED2), a negative regulator of hypoxia-inducible factor-1,27 which, to our knowledge, has not been previously identified in endothelial cells; a disintegrin and metalloprotease with thrombospondin motifs 1 (ADAMTS1), a metalloprotease that has been shown to inhibit angiogenesis28,29; tumor necrosis factor (TNF) receptor superfamily member 12A, which has been reported to induce angiogenesis and migration30; and the CX3C chemokine fractalkine, a potent agonist for chemotaxis and adhesion of monocytes and lymphocytes.31

Two other groups have used DNA microarrays to analyze thrombin or thrombin-receptor signaling in endothelial cells. In 1 study, treatment of serum-starved, confluent HUVECs with a PAR-1 agonist peptide (TFLLRNPNDK) for 90 minutes resulted in a reproducible upregulation of 1% of the 7000 genes represented on the HG-U95Av2 array (Affymetrix).32 There was no mention in this study of PAR-1–associated gene repression. In the other report, custom microarrays of 300 vascular cell–related gene fragments were used to probe the effect of various extracellular mediators, including 5 U/mL thrombin, on confluent HUVECs for 2, 6, and 24 hours.33 However, the authors did not comment on the nature and/or level of the thrombin-responsive transcripts.33

Thrombin-Mediated Transcriptional Networks

In theory, the effect of thrombin on mRNA expression in endothelial cells might be mediated at the level of mRNA stability or rate of transcription. Little is known about the effect of extracellular signals on mRNA stability in the endothelium. In contrast, there has been an increased understanding of the role for transcription factors in transducing the thrombin signal at the level of gene promoters (Figure 2 and Table 6).

   Figure 2. Schemata of thrombin-responsive transcriptional networks in endothelial cells. Shown are signaling pathways that couple thrombin with SRF–Egr-1, p65–NF-B with ICAM-1, p65–NF-B, GATA-2 with VCAM-1, and dbpB with PDGF-B and EPCR Abbreviations are as defined in text.

   TABLE 6. Thrombin and Transcriptional Networks in Endothelial Cells

DNA Binding Protein B (dbpB)

The first evidence for the existence of a thrombin-responsive transcriptional network in endothelial cells was derived from studies of the PDGF-B promoter. Thrombin-mediated expression of PDGF-B was shown to involve the inducible binding of a Y-box binding transcription factor (termed dbpB) to a 9-bp thrombin-response element (TRE) in the upstream promoter region.34,35 In response to thrombin, dbpB is cleaved and released from mRNA in the cytoplasm, resulting in nuclear translocation and transcriptional activation of downstream target gene(s).35 In addition to PDGF-B, dbpB has been implicated in the thrombin-mediated regulation of TF35 and EPCR genes.25 Recently, Esmon and colleagues used homologous recombination to delete the dbpB consensus motif from the endogenous EPCR gene. They showed that wild-type, but not mutant, EPCR was responsive to thrombin under in vivo conditions (C.T. Esmon, personal communication). These latter data support a role for the dbpB transcription factor in mediating thrombin response in the intact endothelium.

Promoters of other thrombin-responsive genes (eg, VCAM-1, thrombomodulin [TM], and Flk-1/KDR) contain consensus elements for dbpB. However, evidence for their functional role in thrombin signaling is lacking. For example, although the VCAM-1 promoter contains a putative TRE at -376 relative to the start site of transcription, a mutation of this site did not affect the ability of thrombin to induce promoter activity.15 Moreover, in mobility shift assays, thrombin did not induce the binding of nuclear protein to a radiolabeled probe that spans the motif.15 In DNA microarrays, thrombin failed to increase mRNA levels of Flk-1/KDR or TM, a finding that has been confirmed in Northern blot assays (C. Seguin and W.C. Aird, unpublished observations). These observations do not rule out a role for the TRE in mediating the thrombin response of VCAM-1, Flk-1/KDR, and/or TM under certain as-yet-undefined spatial and temporal conditions. However, on the basis of these findings, as well as those discussed later, we believe that the dbpb consensus motif should be considered as 1 member of an expansive family of TREs.

Nuclear Factor-B

There is increasing evidence for the importance of nuclear factor (NF)-B in mediating thrombin response in the endothelium. NF-B is a proinflammatory, rapidly inducible transcription factor that upregulates several target genes involved in endothelial cell activation. The NF-B/Rel family consists of 5 distinct DNA-binding proteins, including p50, p52, p65 (RelA), c-Rel, and RelB. In quiescent cells, NF-B is sequestered in the cytoplasm, complexed to the inhibitory protein IB. Stimulation of cells with inflammatory mediators results in the release of NF-B from IB. Once released, NF-B translocates into the nucleus, where it binds as heterodimers or homodimers to NF-B elements in the promoter regions of target genes.

Thrombin treatment of endothelial cells results in rapid translocation of NF-B to the nucleus (please see online Figure II at http://atvb.ahajournals.org). Thrombin-mediated induction of ICAM-1 has been shown to be mediated by the binding of p65 homodimers to a single NF-B site in the upstream promoter region.36 Similarly, thrombin stimulation of VCAM-1 involves the inducible binding of p65 NF-B to a tandem NF-B motif in the 5' flanking region. When the VCAM-1 NF-B motifs are linked in tandem to a thymidine kinase (TK) heterologous core promoter, they retain thrombin responsiveness.37 Taken together, these findings suggest that in the case of VCAM-1 and ICAM-1, p65 NF-B is both sufficient and necessary for transducing the thrombin response. These observations contrast to the more commonly reported involvement of p50-p65 heterodimers in mediating NF-B–inducible gene expression.38 In fact, the p50 subunit has been proposed to function as a negative transcriptional regulator of the VCAM-1 gene by virtue of its ability to inhibit the transcriptional potential of p65.39 RelB has also been shown to inhibit p65 DNA binding.40 Interestingly, the microarray studies of thrombin-treated endothelial cells demonstrated significant induction of p50 NF-B and RelB mRNA at 4 hours (online Figure IA), suggesting a possible negative-feedback mechanism in the thrombin–VCAM-1 signaling axis.

It is interesting to speculate that thrombin signaling is unique in its ability to induce binding of p65 homodimers to downstream target genes. An alternative explanation is that p65 homodimers are involved in transducing many signals at the level of the VCAM-1 and ICAM-1 promoters but not those of other endothelial cell genes. In support of this latter hypothesis, the addition of thrombin to porcine aortic endothelial cells was reported to induce the binding of both p65 and p50 to the NF-B binding site of the IB- promoter.19

A previous study showed that NF-B binding to the VCAM-1 promoter, but not to the E-selectin promoter, was dependent on the redox state of the endothelial cell.41 Although the latter study did not address the effect of reactive oxygen species on thrombin-mediated binding of NF-B, the results are important in that they demonstrate gene/promoter-specific differences in NF-B signaling. Given the potential influence of the NF-B consensus element or a surrounding promoter sequence in directing the binding of specific homodimers, heterodimers, and/or coactivators, it seems prudent to carry out studies of NF-B–mediated gene regulation with NF-B elements that are derived from the promoter of interest.

In studies of the VCAM-1 gene, thrombin has been shown to induce p65 homodimer binding through a PI3K-, PKC-–dependent, Akt-independent signaling pathway (Figure 2).15,37 PKC- was also implicated in thrombin-mediated induction of p65 NF-B binding to the ICAM-1 promoter. However, in contrast to VCAM-1, thrombin stimulation of ICAM-1 was shown to involve parallel PI3K and PKC- pathways, both converging at the level of Akt.42 It is not clear whether these differences reflect gene-specific signaling pathways or rather differences in experimental technique/design.

GATA-2

GATA-binding proteins were initially characterized as constitutively active transcription factors involved in mediating cell type–specific gene expression and lineage determination.43,44 However, recent studies have uncovered a potentially important role for GATA proteins in temporal gene regulation. GATA DNA-binding activity and/or GATA mRNA expression have been shown to increase in response to a number of mediators, including insulinlike growth factor 1,45 follicle-stimulating hormone,46 endothelin-1,47 IL-3,48 and IL-4.49 GATA activity has been reported to decrease in response to other mediators, such as estrogen50 and transforming growth factor (TGF)-?.51

Thrombin-mediated induction of VCAM-1 was shown to involve the inducible binding of GATA-2 to a tandem GATA motif in the upstream promoter.15 The GATA elements were both sufficient and necessary for thrombin-mediated induction of the VCAM-1 gene. Interestingly, the effect of thrombin on GATA-2 DNA binding and transcriptional activity was found to be mediated by a PI3K-, PKC-–dependent signaling pathway37 (Figure 2). The mechanism by which thrombin induces the binding of GATA-2 to the VCAM-1 promoter remains to be established. In preliminary studies, the addition of thrombin to endothelial cells did not induce GATA-2 mRNA and protein level, nor did it alter the total level of serine phosphorylation (T. Minami and W.C. Aird, unpublished results). Of note, thrombin treatment of HUVECs reduced the expression of GATA-3 mRNA (online Figure I). Whether the relative levels of GATA-2 and GATA-3 play a role in mediating the thrombin response of VCAM-1 and the extent to which GATA transcription factors are involved in mediating the thrombin response of other genes remains to be determined.

Early Growth-Response Factor-1

Egr-1 (also known as zif268, TIS 8, NFGI-A, and Krox 24) is a member of the immediate-early gene family that includes c-fos, c-jun, and early growth-response genes.52–57 Egr-1, which encodes a serum-inducible zinc finger nuclear phosphoprotein, is rapidly induced in cultured cells by a wide variety of mitogenic and nonmitogenic stimuli. In endothelial cells, Egr-1 has been shown to be activated by acidic fibroblast growth factor,58 basic fibroblast growth factor,59,60 VEGF,61 epidermal growth factor (EGF),62,63 shear stress,64–66 cyclical strain,67 and hypoxia.68,69 Under in vivo conditions, the administration of VEGF and EGF results in vascular bed–specific changes in Egr-1 expression.63 Moreover, elevated Egr-1 levels have been reported in human atherosclerotic lesions.70

The murine and human Egr-1 promoters contain 5 functional serum-response elements (SREs) organized into 2 clusters: a 5' cluster of 3 SREs and a 3' cluster of 2 SREs. Collectively, the 5 SREs are responsible for transducing most, if not all, signals that activate Egr-1 expression. The 5' SRE cluster has been shown to mediate response to the majority of extracellular signals, including EGF,62 shear stress,64 growth hormone,71 urea,72 hypotonicity, lipopolysaccharide (LPS),73 granulocyte-colony stimulating factor,74 and hypoxia.69 In nonendothelial cells, the 3' cluster has been implicated in the response to glucose-induced depolarization75 and granulocyte-macrophage stimulating factor.76,77

Recently, thrombin has been shown to induce the expression Egr-1 via the most proximal SRE (SRE-1).18 Although previous studies have shown that the response of the 5' cluster of SREs to extracellular signals is mediated by the coordinate action of serum-response factor (SRF) and ternary complex factor,64,69,71,73 SRE-1 was shown to transduce the thrombin signal by an SRF-dependent, TCF-independent mechanism.18

There is a growing appreciation that thrombin signaling contributes to a proinflammatory state. Egr-1 has been reported to induce a number of downstream genes, including PDGF-A, PDGF-B, TF, Flt-1, TGF-?, TNF-, urokinase-type plasminogen activator, and metalloproteinases.61,78–80 Endothelial expression of Egr-1 is increased in response to injury.81 The effect of Egr-1 on an individual target gene is likely to vary according to the cell type and extracellular signal.82 An important goal for future studies will be to characterize the nature of the Egr-1–responsive gene program in thrombin-treated endothelial cells and to determine the extent to which these gene products contribute to the cellular phenotype.

Other Transcriptional Networks

Thrombin has been shown to induce the activity of activator protein-1 in endothelial cells in some, but not all, studies.23,83–85 Moreover, thrombin signaling in endothelial cells results in phosphorylation of the forkhead transcription factors FKHR and AFX (please see online Figure III at http://atvb.ahajournals.org). The results of the microarray experiments reveal upregulation and downregulation of many transcription factors (online Figure I). These results await validation at the level of both mRNA and DNA binding activity. Thrombin signaling in vascular smooth muscle cells (VSMCs) has been linked to NF-B, Sp1, cAMP-responsive element binding protein,86,87 activator protein-1,88 hypoxia-inducible factor-,89,90 signal transducers and activators of transcription (STATs),91 and nuclear factor of activated T cells92; in fibroblasts to Elk-1 and cyclicAMP regulatory element-binding protein (CBP)/p30093,94; and in mesangial cells to STAT-1 and -3.95 A role for these latter transcription factors in transducing the thrombin signal in endothelial cells has yet to be established.

Transcriptional Network Clustering

Common bioinformatic approaches for analyzing the results of microarray experiments include unsupervised learning techniques (eg, hierarchical clustering) and supervised learning techniques, such as assigning genes to predefined biologically meaningful classes (functional clustering). An alternative approach is to link the microarray data with an existing genomic database to gain insight into thrombin-responsive transcriptional networks. This strategy entails 2 steps: (1) identifying promoter sequences of genes that are induced by thrombin and (2) scanning a predetermined length of the promoter for established or conserved DNA motifs. In preliminary studies, we have used such an approach to analyze our microarray data (Figure 3). A preponderance of promoters of genes that were maximally upregulated at 1 hour contained SREs, which is what one would expect for an immediate-early gene. In contrast, there was a greater representation of NF-B, Egr-1, and GATA-binding elements in the promoters of genes that were induced at 4 hours. Finally, consensus dbpB binding elements were found on the promoters of growth related oncogen ? (GRO-?) and Egr-3 (maximally induced at 1 hour); VCAM-1, ICAM-1, fractalkine, and NF-B1 (maximally induced at 4 hours); and DAF, chemokine (CXC motif) receptor 4, myosin IE, PDGF-A, PDGF-B, and TNF (ligand) superfamily member 15 (maximally induced at 18 hours).

   Figure 3. Scanning the promoters of highly induced genes for established or conserved transcription factor–binding motifs. Promoter sequences (-500 to +100) from 8 of the highest thrombin-induced genes at 1 and 4 hours were retrieved from GenBank and analyzed with Chip2 promoter version 1.0 of GenomatixSuite software (CTC Laboratory systems). Please see http://atvb.ahajournals.org for additional details. GLI indicates glioma; MGSA indicates melanoma growth stimulating activity; and TRAF indicates TNF- receptor-associated factor. Other abbreviations are as defined in text.

Cell Type–Specific Nature of Thrombin Signaling

Thrombin receptors are present on many different cell types, including endothelial cells, platelets, VSMCs,96–100 fibroblasts,93,101,102 mast cells,103,104 macrophages,104 microglia,105 tumor cells, keratinocytes,106 and leukocytes.107,108

Thrombin signaling varies between cell types. For example, whereas thrombin induces expression of VCAM-1 in endothelial cells, it has no such effect in VSMCs.37 In contrast, IL-4 increased VCAM-1 mRNA levels in VSMCs, suggesting that the VCAM-1 gene is inducible under other conditions. Moreover, thrombin (as well as the PAR-1 agonist peptide) induced the expression of TF in VSMCs, confirming that VSMCs possess functional thrombin receptors.37

The discordance in thrombin response between endothelial cells and other cell types is not unique to VCAM-1. For example, although previous studies have shown that thrombin and/or PAR-1 singling induces VEGF expression in fibroblasts, endometrial cells, tumor cells, mesothelial cells, and VSMCs,100,101,109–111 there are no reports of thrombin stimulation of VEGF in endothelial cells, an observation that is supported by our microarray data. In contrast, other extracellular signals, such as hypoxia, have been shown to increase VEGF mRNA levels in endothelial cells (largely through stabilizing mRNA).112,113 We have recently demonstrated that thrombin induces expression of PDGF and ICAM-1 in cultured endothelial cells, but not in VSMCs (S.Q. Wu and W.C. Aird, unpublished observations). Other studies have suggested that PAR-1 levels (and therefore, sensitization to thrombin) are differentially regulated in endothelial cells and VSMCs.114,115 Differences also exist in PAR-1 signaling between endothelial cells and platelets. For example, in platelets, thrombin stimulates cdc42 and Rac1 activity, whereas in endothelial cells, thrombin has no effect on cdc42 activity and inhibits Rac1.116

Taken together, these results suggest that thrombin signaling varies between different cell types and that these differences are mediated by differential expression and/or activity of PARs or downstream signal intermediates. It follows that thrombin signaling should be studied in the context of the cell type of interest and that data obtained from 1 cell lineage should not be extrapolated to another.

Not All Inflammatory Mediators Are Created Equal

Endothelial cell activation is not an all-or-nothing phenomenon. Indeed, the notion of the endothelium existing in a binary state (quiescent or activated) has given way to an appreciation of the endothelium as a nonlinear system that displays phenotypic heterogeneity in space and time.117 Any notion that inflammatory mediators have identical effects on endothelial cells is an oversimplification.

Although there are similarities in the way inflammatory mediators, such as thrombin, TNF-, IL-1, and LPS, modulate endothelial cell phenotype, there are also important qualitative and quantitative differences. For example, in 1 study with DNA microarrays, treatment of HUVECs with TNF- and IL-1? resulted in overlapping but nonidentical patterns of gene expression.33 Thrombin, but not TNF- or LPS, induced PDGF-A in human endothelial cells derived from umbilical vein, pulmonary artery, or coronary artery (S.Q. Wu and W.C. Aird, unpublished observations). Compared with TNF-, thrombin signaling in porcine aortic endothelial cells results in lower and delayed induction of NF-B binding activity.19 In HUVECs, TNF-–and IL-?–mediated induction of ICAM-1 peaks at 2 hour, whereas thrombin stimulation is maximal at 4 hours.36 Moreover, in the same cell type, thrombin induces ICAM-1 via a PKC-–p65–NF-B signaling pathway,36 whereas TNF-–mediated induction of the ICAM-1 gene involves a PKC-–p65–NF-B signaling pathway.118 Inhibitors of mitogen-activated protein kinase p38 completely abrogate thrombin-, but not TNF-–, induced leukocyte recruitment in HUVECs.119 As a final example of mediator-specific responses, treatment of HUVECs with TNF-, but not LPS, resulted in a biphasic change in PAR-1 mRNA levels, with an initial decrease and a subsequent rebound above baseline.120 Together, these and other studies emphasize that although inflammatory mediators induce overlapping changes in endothelial phenotypes, each mediator engages the endothelium in its own unique way.

Temporal and Spatial Control of Thrombin Signaling

There is no question that in vitro studies of thrombin signaling have advanced our understanding of this important mediator. However, cell culture studies have their limitations. When endothelial cells are removed from their native tissue environment, they are uncoupled from critical extracellular cues and undergo phenotypic drift. As a result, signaling pathways and the transcriptional control machinery might change in ways that are difficult to control or account for. Under in vivo conditions, endothelial cells are differentially regulated in space and time.121 Stated another way, the intact endothelium represents a nonlinear dynamic, in that the whole is far greater than the sum of the parts.117 Studies of the individual parts (eg, cultured endothelial cells), however important and informative they might be, do not provide insight into higher-order behavior.

Consider, for example, the thrombin–VCAM-1 signaling pathway.15,37 The results of in vitro studies are interesting in that they (1) establish a role for GATA-2 as a signal transducer in endothelial cells, (2) imply a novel link between an atypical PKC isoform and GATA-2, and (3) provide a potentially valuable model for dissecting the mechanisms of cell type–specific thrombin signaling (endothelial cells vs VSMCs). However, there are many gaps in our knowledge. Our results are derived from cells (namely, HUVECs) that originate from a vascular bed, which, although readily accessible and historically favored, is poorly understood in health and disease. In the context of the blood vessel wall, the intact endothelium is not surrounded by tissue-culture medium and plastic but rather by free-flowing blood on 1 side and extracellular matrix and parenchymal cells on the other. How does the thrombin–VCAM-1 signaling pathway normally behave under in vivo conditions? Or, for that matter, under states of activation, such as sepsis, in which blood might be acidic, hypoxic, and delivered under low perfusion pressures; in which there might be local accumulation of inflammatory mediators and thrombin and secondary changes in leukocyte trafficking, fibrin deposition, and barrier function, all of which might impact in their own way on endothelial cell phenotype?122

In addition to these temporal considerations and in keeping with the theme of endothelial cell heterogeneity, there is every reason to believe that the expression and/or activity of the various components of the thrombin–VCAM-1 signaling axis vary between different sites of the vascular tree. For example, at any given point in time, regional differences might exist in the amount of thrombin being generated in the intravascular space, the number or ratio of PARs on the cell surface,123–125 the expression and/or activity of PKC- and - isoforms, and/or the level or transcriptional potential of p65 NF-B and GATA-2.126

In summary, current in vitro models of endothelial cell biology fall short in capturing the complex, dynamic, and emergent nature of the intact endothelium. At best, the results of the in vitro studies provide a first approximation of the true biology and might be used as a road map for exploring the system in vivo. An important goal for the future will be to learn how to better leverage the advantages that are inherent in the reductionist (in vitro) and holistic (in vivo) approaches for both mechanistic and therapeutic gain.

Thrombin Signaling—A 2-Edged Sword

Although our understanding of the molecular basis of thrombin–PAR-1 interactions and downstream signaling is rapidly evolving, we have relatively little insight about the role of PAR signaling in health and disease. As a general rule, thrombin is considered to activate endothelial cells, leading to cellular "dysfunction." Thrombin is present in increased concentrations at sites of vascular injury,127 in the vicinity of a thrombus,127 in patients with acute coronary syndromes,128 and in primate models of sepsis.129 Thrombin and/or PAR signaling has been implicated in rheumatoid (or collagen-induced) arthritis130,131 and preeclampsia.108 Despite these associations, it has been difficult to prove a cause-and-effect relation between thrombin generation and pathology or to quantify the relative contributions of thrombin-induced fibrin formation versus thrombin-mediated activation of platelets, endothelial cells, or other cell types.

There is increasing evidence that thrombin signaling also plays a protective role in endothelial cell biology. PAR-1–null mice might be rescued with endothelial cell–specific expression of PAR-1, suggesting that activation of PAR-1 and its signaling pathway in endothelial cells are essential for vascular integrity.132 Moreover, thrombin signaling might play an important role in angiogenesis.22,133,134 Thrombin also promotes the production and secretion of extracellular matrix proteins135 and positively influences remodeling processes.136,137 Other studies have shown that thrombin protects against cell death.138,139 In a recent report, PAR-1 stimulation in HUVECs resulted in increased expression of the antiapoptotic genes BCL2-related protein A1 and inhibitor of apoptosis 1, and a variety of negative regulators of proinflammatory pathways.32 Taken together, these studies demonstrate the multifaceted role of thrombin signaling at the level of the endothelium.

If one is to believe that thrombin (and/or PAR) signaling is a 2-edged sword (as we do), then it should theoretically be possible to tease out signaling pathways or transcriptional networks whose inhibition will lead to a preferential loss of proinflammatory response while retaining the protective function. To test this hypothesis, we are currently using microarray experiments of control or thrombin-treated HUVECs that have been pretreated in the absence or presence of various inhibitors of signal intermediates or transcription factors. By mining the resulting data, we will be able to assign downstream thrombin target genes to distinct signaling pathways. Our goal is to identify signal transduction components that are amenable to selective therapeutic targeting.

    Conclusions

Thrombin not only represents an interface between coagulation and inflammation but is also a powerful tool for exploring "gray" areas in vascular biology, namely, (1) the spatial and temporal dynamics of endothelial cell phenotypes, (2) the nonbinary nature of endothelial cell activation, (3) the double-edged sword of endothelial cell signaling, and (4) the emergent properties of the endothelium. Continued research in thrombin signaling promises to advance our understanding in each of these areas.

    Acknowledgments

 

This work was supported by National Institutes of Health grants HL60585, HL63609, HL65216, and HL36028.

References

Aird WC. Vascular bed-specific hemostasis: role of endothelium in sepsis pathogenesis. Crit Care Med. 2001; 29: S28–S35.

Coughlin SR. Thrombin signalling and protease-activated receptors. Nature. 2000; 407: 258–264.

Vu TK, Hung DT, Wheaton VI, Coughlin SR. Molecular cloning of a functional thrombin receptor reveals a novel proteolytic mechanism of receptor activation. Cell. 1991; 64: 1057–1068.

Ishihara H, Connolly AJ, Zeng D, Kahn ML, Zheng YW, Timmons C, Tram T, Coughlin SR. Protease-activated receptor 3 is a second thrombin receptor in humans. Nature. 1997; 386: 502–506.

Nakanishi-Matsui M, Zheng YW, Sulciner DJ, Weiss EJ, Ludeman MJ, Coughlin SR. PAR3 is a cofactor for PAR4 activation by thrombin. Nature. 2000; 404: 609–613.

O’Brien PJ, Prevost N, Molino M, Hollinger MK, Woolkalis MJ, Woulfe DS, Brass LF. Thrombin responses in human endothelial cells: contributions from receptors other than PAR1 include the transactivation of PAR2 by thrombin-cleaved PAR1. J Biol Chem. 2000; 275: 13502–13509.

Schmidt VA, Nierman WC, Maglott DR, Cupit LD, Moskowitz KA, Wainer JA, Bahou WF. The human proteinase-activated receptor-3 (PAR-3) gene: identification within a Par gene cluster and characterization in vascular endothelial cells and platelets. J Biol Chem. 1998; 273: 15061–15068.

Hamilton JR, Frauman AG, Cocks TM. Increased expression of protease-activated receptor-2 (PAR2) and PAR4 in human coronary artery by inflammatory stimuli unveils endothelium-dependent relaxations to PAR2 and PAR4 agonists. Circ Res. 2001; 89: 92–98.

Gerszten RE, Chen J, Ishii M, Ishii K, Wang L, Nanevicz T, Turck CW, Vu TK, Coughlin SR. Specificity of the thrombin receptor for agonist peptide is defined by its extracellular surface. Nature. 1994; 368: 648–651.

Eto M, Barandier C, Rathgeb L, Kozai T, Joch H, Yang Z, Luscher TF. Thrombin suppresses endothelial nitric oxide synthase and upregulates endothelin-converting enzyme-1 expression by distinct pathways: role of Rho/ROCK and mitogen-activated protein kinase. Circ Res. 2001; 89: 583–590.

Huang YQ, Li JJ, Hu L, Lee M, Karpatkin S. Thrombin induces increased expression and secretion of angiopoietin-2 from human umbilical vein endothelial cells. Blood. 2002; 99: 1646–1650.

Shankar R, de la Motte CA, Poptic EJ, DiCorleto PE. Thrombin receptor-activating peptides differentially stimulate platelet-derived growth factor production, monocytic cell adhesion, and E-selectin expression in human umbilical vein endothelial cells. J Biol Chem. 1994; 269: 13936–13941.

Marin V, Farnarier C, Gres S, Kaplanski S, Su MS, Dinarello CA, Kaplanski G. The p38 mitogen-activated protein kinase pathway plays a critical role in thrombin-induced endothelial chemokine production and leukocyte recruitment. Blood. 2001; 98: 667–673.

Kaplanski G, Marin V, Fabrigoule M, Boulay V, Benoliel AM, Bongrand P, Kaplanski S, Farnarier C. Thrombin-activated human endothelial cells support monocyte adhesion in vitro following expression of intercellular adhesion molecule-1 (ICAM-1; CD54) and vascular cell adhesion molecule-1 (VCAM-1; CD106). Blood. 1998; 92: 1259–1267.

Minami T, Aird WC. Thrombin stimulation of the vascular cell adhesion molecule-1 promoter in endothelial cells is mediated by tandem nuclear factor-B and GATA motifs. J Biol Chem. 2001; 276: 47632–47641.

Kaplanski G, Fabrigoule M, Boulay V, Dinarello CA, Bongrand P, Kaplanski S, Farnarier C. Thrombin induces endothelial type II activation in vitro: IL-1 and TNF--independent IL-8 secretion and E-selectin expression. J Immunol. 1997; 158: 5435–5441.

Lidington EA, Haskard DO, Mason JC. Induction of decay-accelerating factor by thrombin through a protease-activated receptor 1 and protein kinase C-dependent pathway protects vascular endothelial cells from complement-mediated injury. Blood. 2000; 96: 2784–2792.

Wu S, Minami T, Donovan DJ, Aird WC. The proximal serum response element in the egr-1 promoter mediates response to thrombin in primary human endothelial cells. Blood. 2002; 100: 4454–4461.

Anrather D, Millan MT, Palmetshofer A, Robson SC, Geczy C, Ritchie AJ, Bach FH, Ewenstein BM. Thrombin activates nuclear factor-B and potentiates endothelial cell activation by TNF. J Immunol. 1997; 159: 5620–5628.

Houliston RA, Keogh RJ, Sugden D, Dudhia J, Carter TD, Wheeler-Jones CP. Protease-activated receptors upregulate cyclooxygenase-2 expression in human endothelial cells. Thromb Haemost. 2002; 88: 321–328.

Wang J, Morita I, Onodera M, Murota SI. Induction of KDR expression in bovine arterial endothelial cells by thrombin: involvement of nitric oxide. J Cell Physiol. 2002; 190: 238–250.

Tsopanoglou NE, Maragoudakis ME. On the mechanism of thrombin-induced angiogenesis: potentiation of vascular endothelial growth factor activity on endothelial cells by up-regulation of its receptors. J Biol Chem. 1999; 274: 23969–23976.

Delerive P, Martin-Nizard F, Chinetti G, Trottein F, Fruchart JC, Najib J, Duriez P, Staels B. Peroxisome proliferator-activated receptor activators inhibit thrombin-induced endothelin-1 production in human vascular endothelial cells by inhibiting the activator protein-1 signaling pathway. Circ Res. 1999; 85: 394–402.

Takeya H, Gabazza EC, Aoki S, Ueno H, Suzuki K. Synergistic effect of sphingosine 1-phosphate on thrombin-induced tissue factor expression in endothelial cells. Blood. 2003; 102: 1693–1700.

Gu JM, Fukudome K, Esmon CT. Characterization and regulation of the 5'-flanking region of the murine endothelial protein C receptor gene. J Biol Chem. 2000; 275: 12481–12488.

Gu JM, Katsuura Y, Ferrell GL, Grammas P, Esmon CT. Endotoxin and thrombin elevate rodent endothelial cell protein C receptor mRNA levels and increase receptor shedding in vivo. Blood. 2000; 95: 1687–1693.

Yin Z, Haynie J, Yang X, Han B, Kiatchoosakun S, Restivo J, Yuan S, Prabhakar NR, Herrup K, Conlon RA, Hoit BD, Watanabe M, Yang YC. The essential role of Cited2, a negative regulator for HIF-1, in heart development and neurulation. Proc Natl Acad Sci U S A. 2002; 99: 10488–10493.

Luque A, Carpizo DR, Iruela-Arispe ML. ADAMTS1/METH1 inhibits endothelial cell proliferation by direct binding and sequestration of VEGF 165. J Biol Chem. 2003; 278: 23656–23665.

Abbaszade I, Liu RQ, Yang F, Rosenfeld SA, Ross OH, Link JR, Ellis DM, Tortorella MD, Pratta MA, Hollis JM, Wynn R, Duke JL, George HJ, Hillman MC Jr, Murphy K, Wiswall BH, Copeland RA, Decicco CP, Bruckner R, Nagase H, Itoh Y, Newton RC, Magolda RL, Trzaskos JM, Burn TC, et al. Cloning and characterization of ADAMTS11, an aggrecanase from the ADAMTS family. J Biol Chem. 1999; 274: 23443–23450.

Wiley SR, Cassiano L, Lofton T, Davis-Smith T, Winkles JA, Lindner V, Liu H, Daniel TO, Smith CA, Fanslow WC. A novel TNF receptor family member binds TWEAK and is implicated in angiogenesis. Immunity. 2001; 15: 837–846.

Goda S, Imai T, Yoshie O, Yoneda O, Inoue H, Nagano Y, Okazaki T, Imai H, Bloom ET, Domae N, Umehara H. CX3C-chemokine, fractalkine-enhanced adhesion of THP-1 cells to endothelial cells through integrin-dependent and -independent mechanisms. J Immunol. 2000; 164: 4313–4320.

Riewald M, Petrovan RJ, Donner A, Mueller BM, Ruf W. Activation of endothelial cell protease activated receptor 1 by the protein C pathway. Science. 2002; 296: 1880–1882.

Dekker RJ, van Soest S, Fontijn RD, Salamanca S, de Groot PG, VanBavel E, Pannekoek H, Horrevoets AJ. Prolonged fluid shear stress induces a distinct set of endothelial cell genes, most specifically lung Kruppel-like factor (KLF2). Blood. 2002; 100: 1689–1698.

Stenina OI, Poptic EJ, DiCorleto PE. Thrombin activates a Y box-binding protein (DNA-binding protein B) in endothelial cells. J Clin Invest. 2000; 106: 579–587.

Stenina OI, Shaneyfelt KM, DiCorleto PE. Thrombin induces the release of the Y-box protein dbpB from mRNA: a mechanism of transcriptional activation. Proc Natl Acad Sci U S A. 2001; 98: 7277–7282.

Rahman A, Anwar KN, True AL, Malik AB. Thrombin-induced p65 homodimer binding to downstream NF-B site of the promoter mediates endothelial ICAM-1 expression and neutrophil adhesion. J Immunol. 1999; 162: 5466–5476.

Minami T, Abid MR, Zhang J, King G, Kodama T, Aird WC. Thrombin stimulation of vascular adhesion molecule-1 in endothelial cells is mediated by protein kinase C (PKC)--NF-B and PKC--GATA signaling pathways. J Biol Chem. 2003; 278: 6976–6984.

De Martin R, Hoeth M, Hofer-Warbinek R, Schmid JA. The transcription factor NF-B and the regulation of vascular cell function. Arterioscler Thromb Vasc Biol. 2000; 20: e83–e88.

Ahmad M, Marui N, Alexander RW, Medford RM. Cell type-specific transactivation of the VCAM-1 promoter through an NF-B enhancer motif. J Biol Chem. 1995; 270: 8976–8983.

Marienfeld R, May MJ, Berberich I, Serfling E, Ghosh S, Neumann M. RelB forms transcriptionally inactive complexes with RelA/p65. J Biol Chem. 2003; 278: 19852–19860.

Faruqi RM, Poptic EJ, Faruqi TR, De La Motte C, DiCorleto PE. Distinct mechanisms for N-acetylcysteine inhibition of cytokine-induced E-selectin and VCAM-1 expression. Am J Physiol. 1997; 273: H817–H826.

Rahman A, True AL, Anwar KN, Ye RD, Voyno-Yasenetskaya TA, Malik AB. G(q) and G? regulate PAR-1 signaling of thrombin-induced NF-B activation and ICAM-1 transcription in endothelial cells. Circ Res. 2002; 91: 398–405.

Charron F, Nemer M. GATA transcription factors and cardiac development. Semin Cell Dev Biol. 1999; 10: 85–91.

Weiss MJ, Orkin SH. GATA transcription factors: key regulators of hematopoiesis. Exp Hematol. 1995; 23: 99–107.

Musaro A, McCullagh KJ, Naya FJ, Olson EN, Rosenthal N. IGF-1 induces skeletal myocyte hypertrophy through calcineurin in association with GATA-2 and NF-ATc1. Nature. 1999; 400: 581–585.

Heikinheimo M, Ermolaeva M, Bielinska M, Rahman NA, Narita N, Huhtaniemi IT, Tapanainen JS, Wilson DB. Expression and hormonal regulation of transcription factors GATA-4 and GATA-6 in the mouse ovary. Endocrinology. 1997; 138: 3505–3514.

Morin S, Paradis P, Aries A, Nemer M. Serum response factor-GATA ternary complex required for nuclear signaling by a G-protein-coupled receptor. Mol Cell Biol. 2001; 21: 1036–1044.

Towatari M, May GE, Marais R, Perkins GR, Marshall CJ, Cowley S, Enver T. Regulation of GATA-2 phosphorylation by mitogen-activated protein kinase and interleukin-3. J Biol Chem. 1995; 270: 4101–4107.

Lee YW, Kuhn H, Kaiser S, Hennig B, Daugherty A, Toborek M. Interleukin 4 induces transcription of the 15-lipoxygenase I gene in human endothelial cells. J Lipid Res. 2001; 42: 783–791.

Blobel GA, Sieff CA, Orkin SH. Ligand-dependent repression of the erythroid transcription factor GATA-1 by the estrogen receptor. Mol Cell Biol. 1995; 15: 3147–3153.

Minami T, Rosenberg RD, Aird WC. Transforming growth factor-?1-mediated inhibition of the flk-1/KDR gene is mediated by a 5'-untranslated region palindromic GATA site. J Biol Chem. 2001; 276: 5395–5402.

Christy BA, Lau LF, Nathans D. A gene activated in mouse 3T3 cells by serum growth factors encodes a protein with ‘zinc finger’ sequences. Proc Natl Acad Sci U S A. 1988; 85: 7857–7861.

Gashler A, Sukhatme VP. Early growth response protein 1 (Egr-1): prototype of a zinc-finger family of transcription factors. Prog Nucleic Acid Res Mol Biol. 1995; 50: 191–224.

Lemaire P, Revelant O, Bravo R, Charnay P. Two mouse genes encoding potential transcription factors with identical DNA-binding domains are activated by growth factors in cultured cells. Proc Natl Acad Sci U S A. 1988; 85: 4691–4695.

Milbrandt J. A nerve growth factor-induced gene encodes a possible transcriptional regulatory factor. Science. 1987; 238: 797–799.

Sukhatme VP, Cao XM, Chang LC, Tsai-Morris CH, Stamenkovich D, Ferreira PC, Cohen DR, Edwards SA, Shows TB, Curran T, et al. A zinc finger-encoding gene coregulated with c-fos during growth and differentiation, and after cellular depolarization. Cell. 1988; 53: 37–43.

Waters CM, Hancock DC, Evan GI. Identification and characterisation of the egr-1 gene product as an inducible, short-lived, nuclear phosphoprotein. Oncogene. 1990; 5: 669–674.

Delbridge GJ, Khachigian LM. FGF-1-induced platelet-derived growth factor-A chain gene expression in endothelial cells involves transcriptional activation by early growth response factor-1. Circ Res. 1997; 81: 282–288.

Ko Y, Totzke G, Schiermeyer B, Zeitler H, Schmitz U, Vetter H, Sachinidis A. Reverse transcriptase-polymerase chain reaction (RT-PCR): a sensitive method to examine basic fibroblast growth factor-induced expression of the early growth response gene-1 (egr-1) in human umbilical arterial endothelial cells. Mol Cell Probes. 1995; 9: 215–222.

Santiago FS, Lowe HC, Day FL, Chesterman CN, Khachigian LM. Early growth response factor-1 induction by injury is triggered by release and paracrine activation by fibroblast growth factor-2. Am J Pathol. 1999; 154: 937–944.

Mechtcheriakova D, Wlachos A, Holzmuller H, Binder BR, Hofer E. Vascular endothelial cell growth factor-induced tissue factor expression in endothelial cells is mediated by EGR-1. Blood. 1999; 93: 3811–3823.

Tsai JC, Liu L, Guan J, Aird WC. The Egr-1 gene is induced by epidermal growth factor in ECV304 cells and primary endothelial cells. Am J Physiol Cell Physiol. 2000; 279: C1414–C1424.

Liu L, Tsai JC, Aird WC. Egr-1 gene is induced by the systemic administration of the vascular endothelial growth factor and the epidermal growth factor. Blood. 2000; 96: 1772–1781.

Schwachtgen JL, Houston P, Campbell C, Sukhatme V, Braddock M. Fluid shear stress activation of egr-1 transcription in cultured human endothelial and epithelial cells is mediated via the extracellular signal-related kinase 1/2 mitogen-activated protein kinase pathway. J Clin Invest. 1998; 101: 2540–2549.

Chiu JJ, Wung BS, Hsieh HJ, Lo LW, Wang DL. Nitric oxide regulates shear stress-induced early growth response-1: expression via the extracellular signal–regulated kinase pathway in endothelial cells. Circ Res. 1999; 85: 238–246.

Houston P, Dickson MC, Ludbrook V, White B, Schwachtgen JL, McVey JH, Mackman N, Reese JM, Gorman DG, Campbell C, Braddock M. Fluid shear stress induction of the tissue factor promoter in vitro and in vivo is mediated by Egr-1. Arterioscler Thromb Vasc Biol. 1999; 19: 281–289.

Wung BS, Cheng JJ, Chao YJ, Hsieh HJ, Wang DL. Modulation of Ras/Raf/extracellular signal–regulated kinase pathway by reactive oxygen species is involved in cyclic strain–induced early growth response-1 gene expression in endothelial cells. Circ Res. 1999; 84: 804–812.

Lo LW, Cheng JJ, Chiu JJ, Wung BS, Liu YC, Wang DL. Endothelial exposure to hypoxia induces Egr-1 expression involving PKC-mediated Ras/Raf-1/ERK1/2 pathway. J Cell Physiol. 2001; 188: 304–312.

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: 15030–15040.

McCaffrey TA, Fu C, Du B, Eksinar S, Kent KC, Bush H Jr, Kreiger K, Rosengart T, Cybulsky MI, Silverman ES, Collins T. High-level expression of Egr-1 and Egr-1-inducible genes in mouse and human atherosclerosis. J Clin Invest. 2000; 105: 653–662.

Clarkson RW, Shang CA, Levitt LK, Howard T, Waters MJ. Ternary complex factors Elk-1 and Sap-1a mediate growth hormone-induced transcription of egr-1 (early growth response factor-1) in 3T3-F442A preadipocytes. Mol Endocrinol. 1999; 13: 619–631.

Cohen DM, Gullans SR, Chin WW. Urea inducibility of egr-1 in murine inner medullary collecting duct cells is mediated by the serum response element and adjacent Ets motifs. J Biol Chem. 1996; 271: 12903–12908.

Guha M, O’Connell MA, Pawlinski R, Hollis A, McGovern P, Yan SF, Stern D, Mackman N. Lipopolysaccharide activation of the MEK-ERK1/2 pathway in human monocytic cells mediates tissue factor and tumor necrosis factor- expression by inducing Elk-1 phosphorylation and Egr-1 expression. Blood. 2001; 98: 1429–1439.

Mora-Garcia P, Sakamoto KM. Granulocyte colony-stimulating factor induces Egr-1 up-regulation through interaction of serum response element-binding proteins. J Biol Chem. 2000; 275: 22418–22426.

Bernal-Mizrachi E, Wice B, Inoue H, Permutt MA. Activation of serum response factor in the depolarization induction of Egr-1 transcription in pancreatic islet ?-cells. J Biol Chem. 2000; 275: 25681–25689.

Sakamoto KM, Fraser JK, Lee HJ, Lehman E, Gasson JC. Granulocyte-macrophage colony-stimulating factor and interleukin-3 signaling pathways converge on the CREB-binding site in the human egr-1 promoter. Mol Cell Biol. 1994; 14: 5975–5985.

Watanabe S, Kubota H, Sakamoto KM, Arai K. Characterization of cis-acting sequences and trans-acting signals regulating early growth response 1 and c-fos promoters through the granulocyte-macrophage colony-stimulating factor receptor in BA/F3 cells. Blood. 1997; 89: 1197–1206.

Khachigian LM, Williams AJ, Collins T. Interplay of Sp1 and Egr-1 in the proximal platelet-derived growth factor A-chain promoter in cultured vascular endothelial cells. J Biol Chem. 1995; 270: 27679–27686.

Vidal F, Aragones J, Alfranca A, de Landazuri MO. Up-regulation of vascular endothelial growth factor receptor Flt-1 after endothelial denudation: role of transcription factor Egr-1. Blood. 2000; 95: 3387–3395.

Haas TL, Stitelman D, Davis SJ, Apte SS, Madri JA. Egr-1 mediates extracellular matrix-driven transcription of membrane type 1 matrix metalloproteinase in endothelium. J Biol Chem. 1999; 274: 22679–22685.

Khachigian LM, Lindner V, Williams AJ, Collins T. Egr-1-induced endothelial gene expression: a common theme in vascular injury. Science. 1996; 271: 1427–1431.

Mechtcheriakova D, Schabbauer G, Lucerna M, Clauss M, De Martin R, Binder BR, Hofer E. Specificity, diversity, and convergence in VEGF and TNF- signaling events leading to tissue factor up-regulation via EGR-1 in endothelial cells. FASEB J. 2001; 15: 230–242.

Kitazumi K, Tasaka K. The role of c-Jun protein in thrombin-stimulated expression of preproendothelin-1 mRNA in porcine aortic endothelial cells. Biochem Pharmacol. 1993; 46: 455–464.

Takata M, Urakaze M, Temaru R, Yamazaki K, Nakamura N, Nobata Y, Kishida M, Sato A, Kobayashi M. Pravastatin suppresses the interleukin-8 production induced by thrombin in human aortic endothelial cells cultured with high glucose by inhibiting the p44/42 mitogen activated protein kinase. Br J Pharmacol. 2001; 134: 753–762.

Pendurthi UR, Williams JT, Rao LV. Inhibition of tissue factor gene activation in cultured endothelial cells by curcumin: suppression of activation of transcription factors Egr-1, AP-1, and NF-B. Arterioscler Thromb Vasc Biol. 1997; 17: 3406–3413.

Edmead C, Kanthou C, Benzakour O. Thrombin activates transcription factors sp1, NF-B, and CREB: importance of the use of phosphatase inhibitors during nuclear protein extraction for the assessment of transcription factor DNA-binding activities. Anal Biochem. 1999; 275: 180–186.

Tokunou T, Ichiki T, Takeda K, Funakoshi Y, Iino N, Shimokawa H, Egashira K, Takeshita A. Thrombin induces interleukin-6 expression through the cAMP response element in vascular smooth muscle cells. Arterioscler Thromb Vasc Biol. 2001; 21: 1759–1763.

Rao GN, Katki KA, Madamanchi NR, Wu Y, Birrer MJ. JunB forms the majority of the AP-1 complex and is a target for redox regulation by receptor tyrosine kinase and G protein-coupled receptor agonists in smooth muscle cells. J Biol Chem. 1999; 274: 6003–6010.

Richard DE, Berra E, Pouyssegur J. Nonhypoxic pathway mediates the induction of hypoxia-inducible factor 1 in vascular smooth muscle cells. J Biol Chem. 2000; 275: 26765–26771.

Gorlach A, Diebold I, Schini-Kerth VB, Berchner-Pfannschmidt U, Roth U, Brandes RP, Kietzmann T, Busse R. Thrombin activates the hypoxia-inducible factor-1 signaling pathway in vascular smooth muscle cells: role of the p22(phox)-containing NADPH oxidase. Circ Res. 2001; 89: 47–54.

Pelletier S, Duhamel F, Coulombe P, Popoff MR, Meloche S. Rho family GTPases are required for activation of Jak/STAT signaling by G protein-coupled receptors. Mol Cell Biol. 2003; 23: 1316–1333.

Yellaturu CR, Ghosh SK, Rao RK, Jennings LK, Hassid A, Rao GN. A potential role for nuclear factor of activated T-cells in receptor tyrosine kinase and G-protein-coupled receptor agonist-induced cell proliferation. Biochem J. 2002; 368: 183–190.

Li QJ, Vaingankar S, Sladek FM, Martins-Green M. Novel nuclear target for thrombin: activation of the Elk1 transcription factor leads to chemokine gene expression. Blood. 2000; 96: 3696–3706.

Li QJ, Yang SH, Maeda Y, Sladek FM, Sharrocks AD, Martins-Green M. MAP kinase phosphorylation-dependent activation of Elk-1 leads to activation of the co-activator p300. EMBO J. 2003; 22: 281–291.

Chen X, Liu W, Wang J, Wang X, Yu Z. STAT1 and STAT3 mediate thrombin-induced expression of TIMP-1 in human glomerular mesangial cells. Kidney Int. 2002; 61: 1377–1382.

Okazaki H, Majesky MW, Harker LA, Schwartz SM. Regulation of platelet-derived growth factor ligand and receptor gene expression by -thrombin in vascular smooth muscle cells. Circ Res. 1992; 71: 1285–1293.

McNamara CA, Sarembock IJ, Bachhuber BG, Stouffer GA, Ragosta M, Barry W, Gimple LW, Powers ER, Owens GK. Thrombin and vascular smooth muscle cell proliferation: implications for atherosclerosis and restenosis. Semin Thromb Hemost. 1996; 22: 139–144.

Bachhuber BG, Sarembock IJ, Gimple LW, McNamara CA, Owens GK. Thrombin-induced mitogenesis in cultured aortic smooth muscle cells requires prolonged thrombin exposure. Am J Physiol. 1995; 268: C1141–C1147.

Bachhuber BG, Sarembock IJ, Gimple LW, Owens GK. -Thrombin induces transforming growth factor-?1 mRNA and protein in cultured vascular smooth muscle cells via a proteolytically activated receptor. J Vasc Res. 1997; 34: 41–48.

Bassus S, Herkert O, Kronemann N, Gorlach A, Bremerich D, Kirchmaier CM, Busse R, Schini-Kerth VB. Thrombin causes vascular endothelial growth factor expression in vascular smooth muscle cells: role of reactive oxygen species. Arterioscler Thromb Vasc Biol. 2001; 21: 1550–1555.

Huang YQ, Li JJ, Hu L, Lee M, Karpatkin S. Thrombin induces increased expression and secretion of VEGF from human FS4 fibroblasts, DU145 prostate cells and CHRF megakaryocytes. Thromb Haemost. 2001; 86: 1094–1098.

Shin H, Kitajima I, Nakajima T, Shao Q, Tokioka T, Takasaki I, Hanyu N, Kubo T, Maruyama I. Thrombin receptor mediated signals induce expressions of interleukin 6 and granulocyte colony stimulating factor via NF-B activation in synovial fibroblasts. Ann Rheum Dis. 1999; 58: 55–60.

D’Andrea MR, Rogahn CJ, Andrade-Gordon P. Localization of protease-activated receptors-1 and -2 in human mast cells: indications for an amplified mast cell degranulation cascade. Biotech Histochem. 2000; 75: 85–90.

D’Andrea MR, Derian CK, Santulli RJ, Andrade-Gordon P. Differential expression of protease-activated receptors-1 and -2 in stromal fibroblasts of normal, benign, and malignant human tissues. Am J Pathol. 2001; 158: 2031–2041.

Ryu J, Pyo H, Jou I, Joe E. Thrombin induces NO release from cultured rat microglia via protein kinase C, mitogen-activated protein kinase, and NF-B. J Biol Chem. 2000; 275: 29955–29959.

Algermissen B, Sitzmann J, Nurnberg W, Laubscher JC, Henz BM, Bauer F. Distribution and potential biologic function of the thrombin receptor PAR-1 on human keratinocytes. Arch Dermatol Res. 2000; 292: 488–495.

Bizios R, Lai L, Fenton JW 2nd, Malik AB. Thrombin-induced chemotaxis and aggregation of neutrophils. J Cell Physiol. 1986; 128: 485–490.

Wang Y, Gu Y, Lucas MJ. Expression of thrombin receptors in endothelial cells and neutrophils from normal and preeclamptic pregnancies. J Clin Endocrinol Metab. 2002; 87: 3728–3734.

Yin YJ, Salah Z, Maoz M, Ram SC, Ochayon S, Neufeld G, Katzav S, Bar-Shavit R. Oncogenic transformation induces tumor angiogenesis: a role for PAR1 activation. FASEB J. 2003; 17: 163–174.

Lockwood CJ, Krikun G, Koo AB, Kadner S, Schatz F. Differential effects of thrombin and hypoxia on endometrial stromal and glandular epithelial cell vascular endothelial growth factor expression. J Clin Endocrinol Metab. 2002; 87: 4280–4286.

Mandl-Weber S, Cohen CD, Haslinger B, Kretzler M, Sitter T. Vascular endothelial growth factor production and regulation in human peritoneal mesothelial cells. Kidney Int. 2002; 61: 570–578.

Claffey KP, Shih SC, Mullen A, Dziennis S, Cusick JL, Abrams KR, Lee SW, Detmar M. Identification of a human VPF/VEGF 3' untranslated region mediating hypoxia-induced mRNA stability. Mol Biol Cell. 1998; 9: 469–481.

Levy NS, Chung S, Furneaux H, Levy AP. Hypoxic stabilization of vascular endothelial growth factor mRNA by the RNA-binding protein HuR. J Biol Chem. 1998; 273: 6417–6423.

Herkert O, Kuhl H, Sandow J, Busse R, Schini-Kerth VB. Sex steroids used in hormonal treatment increase vascular procoagulant activity by inducing thrombin receptor (PAR-1) expression: role of the glucocorticoid receptor. Circulation. 2001; 104: 2826–2831.

Capers QT, Laursen JB, Fukui T, Rajagopalan S, Mori I, Lou P, Freeman BA, Berrington WR, Griendling KK, Harrison DG, Runge MS, Alexander RW, Taylor WR. Vascular thrombin receptor regulation in hypertensive rats. Circ Res. 1997; 80: 838–844.

Vouret-Craviari V, Bourcier C, Boulter E, van Obberghen-Schilling E. Distinct signals via Rho GTPases and Src drive shape changes by thrombin and sphingosine-1-phosphate in endothelial cells. J Cell Sci. 2002; 115: 2475–2484.

Aird WC. Endothelial cell dynamics and complexity theory. Crit Care Med. 2002; 30: S180–S185.

Rahman A, Anwar KN, Malik AB. Protein kinase C- mediates TNF--induced ICAM-1 gene transcription in endothelial cells. Am J Physiol Cell Physiol. 2000; 279: C906–C914.

Kaur J, Woodman RC, Kubes P. P38 MAPK: critical molecule in thrombin-induced NF-B-dependent leukocyte recruitment. Am J Physiol Heart Circ Physiol. 2003; 284: H1095–H1103.

Shinohara T, Suzuki K, Takada K, Okada M, Ohsuzu F. Regulation of proteinase-activated receptor 1 by inflammatory mediators in human vascular endothelial cells. Cytokine. 2002; 19: 66–75.

Aird WC. Endothelial cell heterogeneity. Crit Care Med. 2003; 31: S221–S230.

Aird WC. The role of the endothelium in severe sepsis and the multiple organ dysfunction syndrome. Blood. 2003; 101: 3765–3777.

Camerer E, Kataoka H, Kahn M, Lease K, Coughlin SR. Genetic evidence that protease-activated receptors mediate factor Xa signaling in endothelial cells. J Biol Chem. 2002; 277: 16081–16087.

Wang J, Zheng H, Ou X, Fink LM, Hauer-Jensen M. Deficiency of microvascular thrombomodulin and up-regulation of protease-activated receptor-1 in irradiated rat intestine: possible link between endothelial dysfunction and chronic radiation fibrosis. Am J Pathol. 2002; 160: 2063–2072.

Jin E, Fujiwara M, Pan X, Ghazizadeh M, Arai S, Ohaki Y, Kajiwara K, Takemura T, Kawanami O. Protease-activated receptor (PAR)-1 and PAR-2 participate in the cell growth of alveolar capillary endothelium in primary lung adenocarcinomas. Cancer. 2003; 97: 703–713.

Hajra L, Evans AI, Chen M, Hyduk SJ, Collins T, Cybulsky MI. The NF-B signal transduction pathway in aortic endothelial cells is primed for activation in regions predisposed to atherosclerotic lesion formation. Proc Natl Acad Sci U S A. 2000; 97: 9052–9057.

Hatton MW, Moar SL, Richardson M. Deendothelialization in vivo initiates a thrombogenic reaction at the rabbit aorta surface: correlation of uptake of fibrinogen and antithrombin III with thrombin generation by the exposed subendothelium. Am J Pathol. 1989; 135: 499–508.

Merlini PA, Bauer KA, Oltrona L, Ardissino D, Cattaneo M, Belli C, Mannucci PM, Rosenberg RD. Persistent activation of coagulation mechanism in unstable angina and myocardial infarction. Circulation. 1994; 90: 61–68.

Levi M, ten Cate H, Bauer KA, van der Poll T, Edgington TS, Buller HR, van Deventer SJ, Hack CE, ten Cate JW, Rosenberg RD. Inhibition of endotoxin-induced activation of coagulation and fibrinolysis by pentoxifylline or by a monoclonal anti-tissue factor antibody in chimpanzees. J Clin Invest. 1994; 93: 114–120.

Marty I, Peclat V, Kirdaite G, Salvi R, So A, Busso N. Amelioration of collagen-induced arthritis by thrombin inhibition. J Clin Invest. 2001; 107: 631–640.

Varisco PA, Peclat V, van Ness K, Bischof-Delaloye A, So A, Busso N. Effect of thrombin inhibition on synovial inflammation in antigen induced arthritis. Ann Rheum Dis. 2000; 59: 781–787.

Griffin CT, Srinivasan Y, Zheng YW, Huang W, Coughlin SR. A role for thrombin receptor signaling in endothelial cells during embryonic development. Science. 2001; 293: 1666–1670.

Haralabopoulos GC, Grant DS, Kleinman HK, Maragoudakis ME. Thrombin promotes endothelial cell alignment in Matrigel in vitro and angiogenesis in vivo. Am J Physiol. 1997; 273: C239–C245.

Herbert JM, Dupuy E, Laplace MC, Zini JM, Bar Shavit R, Tobelem G. Thrombin induces endothelial cell growth via both a proteolytic and a non-proteolytic pathway. Biochem J. 1994; 303: 227–231.

Papadimitriou E, Manolopoulos VG, Hayman GT, Maragoudakis ME, Unsworth BR, Fenton JW 2nd, Lelkes PI. Thrombin modulates vectorial secretion of extracellular matrix proteins in cultured endothelial cells. Am J Physiol. 1997; 272: C1112–C1122.

Cheung WM, D’Andrea MR, Andrade-Gordon P, Damiano BP. Altered vascular injury responses in mice deficient in protease-activated receptor-1. Arterioscler Thromb Vasc Biol. 1999; 19: 3014–3024.

Duhamel-Clerin E, Orvain C, Lanza F, Cazenave JP, Klein-Soyer C. Thrombin receptor-mediated increase of two matrix metalloproteinases, MMP-1 and MMP-3, in human endothelial cells. Arterioscler Thromb Vasc Biol. 1997; 17: 1931–1938.

Chinni C, de Niese MR, Tew DJ, Jenkins AL, Bottomley SP, Mackie EJ. Thrombin, a survival factor for cultured myoblasts. J Biol Chem. 1999; 274: 9169–9174.

Vaughan PJ, Pike CJ, Cotman CW, Cunningham DD. Thrombin receptor activation protects neurons and astrocytes from cell death produced by environmental insults. J Neurosci. 1995; 15: 5389–5401.

Stasek JE Jr, Patterson CE, Garcia JG. Protein kinase C phosphorylates caldesmon77 and vimentin and enhances albumin permeability across cultured bovine pulmonary artery endothelial cell monolayers. J Cell Physiol. 1992; 153: 62–75.

Rabiet MJ, Plantier JL, Rival Y, Genoux Y, Lampugnani MG, Dejana E. Thrombin-induced increase in endothelial permeability is associated with changes in cell-to-cell junction organization. Arterioscler Thromb Vasc Biol. 1996; 16: 488–496.

Harrington EO, Brunelle JL, Shannon CJ, Kim ES, Mennella K, Rounds S. Role of protein kinase C isoforms in rat epididymal microvascular endothelial barrier function. Am J Respir Cell Mol Biol. 2003; 28: 626–636.

Vanhauwe JF, Thomas TO, Minshall RD, Tiruppathi C, Li A, Gilchrist A, Yoon EJ, Malik AB, Hamm HE. Thrombin receptors activate G0 proteins in endothelial cells to regulate intracellular calcium and cell shape changes. J Biol Chem. 2002; 277: 34143–34149.

Cioffi DL, Moore TM, Schaack J, Creighton JR, Cooper DM, Stevens T. Dominant regulation of interendothelial cell gap formation by calcium-inhibited type 6 adenylyl cyclase. J Cell Biol. 2002; 157: 1267–1278.

Aschner JL, Lum H, Fletcher PW, Malik AB. Bradykinin- and thrombin-induced increases in endothelial permeability occur independently of phospholipase C but require protein kinase C activation. J Cell Physiol. 1997; 173: 387–396.

Garcia JG, Siflinger-Birnboim A, Bizios R, Del Vecchio PJ, Fenton JW 2nd, Malik AB. Thrombin-induced increase in albumin permeability across the endothelium. J Cell Physiol. 1986; 128: 96–104.

Nguyen LT, Lum H, Tiruppathi C, Malik AB. Site-specific thrombin receptor antibodies inhibit Ca2+ signaling and increased endothelial permeability. Am J Physiol. 1997; 273: C1756–C1763.

Sandoval R, Malik AB, Naqvi T, Mehta D, Tiruppathi C. Requirement for Ca2+ signaling in the mechanism of thrombin-induced increase in endothelial permeability. Am J Physiol Lung Cell Mol Physiol. 2001; 280: L239–L247.

De Mey JG, Claeys M, Vanhoutte PM. Endothelium-dependent inhibitory effects of acetylcholine, adenosine triphosphate, thrombin and arachidonic acid in the canine femoral artery. J Pharmacol Exp Ther. 1982; 222: 166–173.

Luscher TF, Diederich D, Siebenmann R, Lehmann K, Stulz P, von Segesser L, Yang ZH, Turina M, Gradel E, Weber E, et al. Difference between endothelium-dependent relaxation in arterial and in venous coronary bypass grafts. N Engl J Med. 1988; 319: 462–467.

Hamilton JR, Nguyen PB, Cocks TM. Atypical protease-activated receptor mediates endothelium-dependent relaxation of human coronary arteries. Circ Res. 1998; 82: 1306–1311.

Toothill VJ, Van Mourik JA, Niewenhuis HK, Metzelaar MJ, Pearson JD. Characterization of the enhanced adhesion of neutrophil leukocytes to thrombin-stimulated endothelial cells. J Immunol. 1990; 145: 283–291.

Zimmerman GA, McIntyre TM, Prescott SM. Thrombin stimulates the adherence of neutrophils to human endothelial cells in vitro. J Clin Invest. 1985; 76: 2235–2246.

Zimmerman BJ, Paulson JC, Arrhenius TS, Gaeta FC, Granger DN. Thrombin receptor peptide-mediated leukocyte rolling in rat mesenteric venules: roles of P-selectin and sialyl Lewis X. Am J Physiol. 1994; 267: H1049–H1053.

Vergnolle N, Derian CK, D’Andrea MR, Steinhoff M, Andrade-Gordon P. Characterization of thrombin-induced leukocyte rolling and adherence: a potential proinflammatory role for proteinase-activated receptor-4. J Immunol. 2002; 169: 1467–1473.

Kaur J, Woodman RC, Ostrovsky L, Kubes P. Selective recruitment of neutrophils and lymphocytes by thrombin: a role for NF-B. Am J Physiol Heart Circ Physiol. 2001; 281: H784–H795.

Maragoudakis ME, Tsopanoglou NE. On the mechanism(s) of thrombin induced angiogenesis. Adv Exp Med Biol. 2000; 476: 47–55.

Pankonin G, Teuscher E. Stimulation of endothelial cell migration by thrombin. Biomed Biochim Acta. 1991; 50: 1073–1078.

Bartha K, Domotor E, Lanza F, Adam-Vizi V, Machovich R. Identification of thrombin receptors in rat brain capillary endothelial cells. J Cereb Blood Flow Metab. 2000; 20: 175–182.

Cheng T, Liu D, Griffin JH, Fernandez JA, Castellino F, Rosen ED, Fukudome K, Zlokovic BV. Activated protein C blocks p53-mediated apoptosis in ischemic human brain endothelium and is neuroprotective. Nat Med. 2003; 9: 338–342.

Copple BL, Moulin F, Hanumegowda UM, Ganey PE, Roth RA. Thrombin and protease-activated receptor-1 agonists promote lipopolysaccharide-induced hepatocellular injury in perfused livers. J Pharmacol Exp Ther. 2003; 305: 417–425.

Grandaliano G, Di Paolo S, Monno R, Stallone G, Ranieri E, Pontrelli P, Gesualdo L, Schena FP. Protease-activated receptor 1 and plasminogen activator inhibitor 1 expression in chronic allograft nephropathy: the role of coagulation and fibrinolysis in renal graft fibrosis. Transplantation. 2001; 72: 1437–1443.

Hamilton JR, Moffatt JD, Tatoulis J, Cocks TM. Enzymatic activation of endothelial protease-activated receptors is dependent on artery diameter in human and porcine isolated coronary arteries. Br J Pharmacol. 2002; 136: 492–501.

Mirza H, Yatsula V, Bahou WF. The proteinase activated receptor-2 (PAR-2) mediates mitogenic responses in human vascular endothelial cells. J Clin Invest. 1996; 97: 1705–1714.

Camerer E, Huang W, Coughlin SR. Tissue factor- and factor X-dependent activation of protease-activated receptor 2 by factor VIIa. Proc Natl Acad Sci U S A. 2000; 97: 5255–5260.

Riewald M, Ruf W. Mechanistic coupling of protease signaling and initiation of coagulation by tissue factor. Proc Natl Acad Sci U S A. 2001; 98: 7742–7747.

Sobey CG, Moffatt JD, Cocks TM. Evidence for selective effects of chronic hypertension on cerebral artery vasodilatation to protease-activated receptor-2 activation. Stroke. 1999; 30: 1933–1940;discussion 1941.

Ferrell WR, Lockhart JC, Kelso EB, Dunning L, Plevin R, Meek SE, Smith AJ, Hunter GD, McLean JS, McGarry F, Ramage R, Jiang L, Kanke T, Kawagoe J. Essential role for proteinase-activated receptor-2 in arthritis. J Clin Invest. 2003; 111: 35–41.

Cicala C, Pinto A, Bucci M, Sorrentino R, Walker B, Harriot P, Cruchley A, Kapas S, Howells GL, Cirino G. Protease-activated receptor-2 involvement in hypotension in normal and endotoxemic rats in vivo. Circulation. 1999; 99: 2590–2597.

Vergnolle N, Derian CK, D’Andrea MR, Steinhoff M, Andrade-Gordon P. Characterization of thrombin-induced leukocyte rolling and adherence: a potential proinflammatory role for proteinase-activated receptor-4. J Immunol. 2002; 169: 1467–1473.

Gilchrist A, Vanhauwe JF, Li A, Thomas TO, Voyno-Yasenetskaya T, Hamm HE. G minigenes expressing C-terminal peptides serve as specific inhibitors of thrombin-mediated endothelial activation. J Biol Chem. 2001; 276: 25672–25679.

Grand RJ, Turnell AS, Grabham PW. Cellular consequences of thrombin-receptor activation. Biochem J. 1996; 313: 353–368.

Yan W, Tiruppathi C, Lum H, Qiao R, Malik AB. Protein kinase C-? regulates heterologous desensitization of thrombin receptor (PAR-1) in endothelial cells. Am J Physiol. 1998; 274: C387–C395.

Sandoval R, Malik AB, Minshall RD, Kouklis P, Ellis CA, Tiruppathi C. Ca2+ signalling and PKC activate increased endothelial permeability by disassembly of VE-cadherin junctions. J Physiol. 2001; 533: 433–445.

Rahman A, Anwar KN, Uddin S, Xu N, Ye RD, Platanias LC, Malik AB. Protein kinase C- regulates thrombin-induced ICAM-1 gene expression in endothelial cells via activation of p38 mitogen-activated protein kinase. Mol Cell Biol. 2001; 21: 5554–5565.

Ellis CA, Malik AB, Gilchrist A, Hamm H, Sandoval R, Voyno-Yasenetskaya T, Tiruppathi C. Thrombin induces proteinase-activated receptor-1 gene expression in endothelial cells via activation of Gi-linked Ras/mitogen-activated protein kinase pathway. J Biol Chem. 1999; 274: 13718–13727.

Eto M, Kozai T, Cosentino F, Joch H, Luscher TF. Statin prevents tissue factor expression in human endothelial cells: role of Rho/Rho-kinase and Akt pathways. Circulation. 2002; 105: 1756–1759.

Vouret-Craviari V, Boquet P, Pouyssegur J, Van Obberghen-Schilling E. Regulation of the actin cytoskeleton by thrombin in human endothelial cells: role of Rho proteins in endothelial barrier function. Mol Biol Cell. 1998; 9: 2639–2653.

Sah VP, Seasholtz TM, Sagi SA, Brown JH. The role of Rho in G protein-coupled receptor signal transduction. Annu Rev Pharmacol Toxicol. 2000; 40: 459–489.

Mehta D, Rahman A, Malik AB. Protein kinase C- signals rho-guanine nucleotide dissociation inhibitor phosphorylation and rho activation and regulates the endothelial cell barrier function. J Biol Chem. 2001; 276: 22614–22620.

Essler M, Amano M, Kruse HJ, Kaibuchi K, Weber PC, Aepfelbacher M. Thrombin inactivates myosin light chain phosphatase via Rho and its target Rho kinase in human endothelial cells. J Biol Chem. 1998; 273: 21867–21874.

van Nieuw Amerongen GP, van Delft S, Vermeer MA, Collard JG, van Hinsbergh VW. Activation of RhoA by thrombin in endothelial hyperpermeability: role of Rho kinase and protein tyrosine kinases. Circ Res. 2000; 87: 335–340.

Wojciak-Stothard B, Potempa S, Eichholtz T, Ridley AJ. Rho and Rac but not Cdc42 regulate endothelial cell permeability. J Cell Sci. 2001; 114: 1343–1355.

Qiao J, Huang F, Lum H. PKA inhibits RhoA activation: a protection mechanism against endothelial barrier dysfunction. Am J Physiol Lung Cell Mol Physiol. 2003; 284: L972–L980.

Keogh RJ, Houliston RA, Wheeler-Jones CP. Thrombin-stimulated Pyk2 phosphorylation in human endothelium is dependent on intracellular calcium and independent of protein kinase C and Src kinases. Biochem Biophys Res Commun. 2002; 294: 1001–1008.

Houliston RA, Pearson JD, Wheeler-Jones CP. Agonist-specific cross talk between ERKs and p38(mapk) regulates PGI2 synthesis in endothelium. Am J Physiol Cell Physiol. 2001; 281: C1266–C1276.

Olivot JM, Estebanell E, Lafay M, Brohard B, Aiach M, Rendu F. Thrombomodulin prolongs thrombin-induced extracellular signal-regulated kinase phosphorylation and nuclear retention in endothelial cells. Circ Res. 2001; 88: 681–687.

Garcia JG, Fenton JW 2nd, Natarajan V. Thrombin stimulation of human endothelial cell phospholipase D activity: regulation by phospholipase C, protein kinase C, and cyclic adenosine 3'5'-monophosphate. Blood. 1992; 79: 2056–2067.

Tiruppathi C, Naqvi T, Sandoval R, Mehta D, Malik AB. Synergistic effects of tumor necrosis factor- and thrombin in increasing endothelial permeability. Am J Physiol Lung Cell Mol Physiol. 2001; 281: L958–L968.

Shi S, Verin AD, Schaphorst KL, Gilbert-McClain LI, Patterson CE, Irwin RP, Natarajan V, Garcia JG. Role of tyrosine phosphorylation in thrombin-induced endothelial cell contraction and barrier function. Endothelium. 1998; 6: 153–171.

Garcia JG, Verin AD, Schaphorst K, Siddiqui R, Patterson CE, Csortos C, Natarajan V. Regulation of endothelial cell myosin light chain kinase by Rho, cortactin, and p60(src). Am J Physiol. 1999; 276: L989–L998.

Borbiev T, Verin AD, Shi S, Liu F, Garcia JG. Regulation of endothelial cell barrier function by calcium/calmodulin-dependent protein kinase II. Am J Physiol Lung Cell Mol Physiol. 2001; 280: L983–L990.

Maragoudakis ME, Tsopanoglou NE, Andriopoulou P. Mechanism of thrombin-induced angiogenesis. Biochem Soc Trans. 2002; 30: 173–177.

Klarenbach SW, Chipiuk A, Nelson RC, Hollenberg MD, Murray AG. Differential actions of PAR2 and PAR1 in stimulating human endothelial cell exocytosis and permeability: the role of Rho-GTPases. Circ Res. 2003; 92: 272–278.

de Leeuw HP, Fernandez-Borja M, Reits EA, Romani de Wit T, Wijers-Koster PM, Hordijk PL, Neefjes J, van Mourik JA, Voorberg J. Small GTP-binding protein Ral modulates regulated exocytosis of von Willebrand factor by endothelial cells. Arterioscler Thromb Vasc Biol. 2001; 21: 899–904.

Yang Z, Arnet U, Bauer E, von Segesser L, Siebenmann R, Turina M, Luscher TF. Thrombin-induced endothelium-dependent inhibition and direct activation of platelet-vessel wall interaction: role of prostacyclin, nitric oxide, and thromboxane A2. Circulation. 1994; 89: 2266–2272.

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: 923–930.

Lorant DE, Patel KD, McIntyre TM, McEver RP, Prescott SM, Zimmerman GA. Coexpression of GMP-140 and PAF by endothelium stimulated by histamine or thrombin: a juxtacrine system for adhesion and activation of neutrophils. J Cell Biol. 1991; 115: 223–234.

Tsopanoglou NE, Andriopoulou P, Maragoudakis ME. On the mechanism of thrombin-induced angiogenesis: involvement of v?3-integrin. Am J Physiol Cell Physiol. 2002; 283: C1501–C1510.

Gelehrter TD, Sznycer-Laszuk R. Thrombin induction of plasminogen activator-inhibitor in cultured human endothelial cells. J Clin Invest. 1986; 77: 165–169.

Marin V, Montero-Julian FA, Gres S, Boulay V, Bongrand P, Farnarier C, Kaplanski G. The IL-6-soluble IL-6R autocrine loop of endothelial activation as an intermediate between acute and chronic inflammation: an experimental model involving thrombin. J Immunol. 2001; 167: 3435–3442.

Nobata Y, Urakaze M, Temaru R, Sato A, Nakamura N, Yamazaki K, Kishida M, Takata M, Kobayashi M. -Tocopherol inhibits IL-8 synthesis induced by thrombin and high glucose in endothelial cells. Horm Metab Res. 2002; 34: 49–54.

Colotta F, Sciacca FL, Sironi M, Luini W, Rabiet MJ, Mantovani A. Expression of monocyte chemotactic protein-1 by monocytes and endothelial cells exposed to thrombin. Am J Pathol. 1994; 144: 975–985.

Grandaliano G, Valente AJ, Abboud HE. A novel biologic activity of thrombin: stimulation of monocyte chemotactic protein production. J Exp Med. 1994; 179: 1737–1741.

Golden CL, Nick HS, Visner GA. Thrombin regulation of endothelin-1 gene in isolated human pulmonary endothelial cells. Am J Physiol. 1998; 274: L854–L863.

Kaneider NC, Egger P, Dunzendorfer S, Noris P, Balduini CL, Gritti D, Ricevuti G, Wiedermann CJ. Reversal of thrombin-induced deactivation of CD39/ATPDase in endothelial cells by HMG-CoA reductase inhibition: effects on Rho-GTPase and adenosine nucleotide metabolism. Arterioscler Thromb Vasc Biol. 2002; 22: 894–900.


 

作者: Takashi Minami; Akira Sugiyama; Sheng-Qian Wu; Ruh 2007-5-18
医学百科App—中西医基础知识学习工具
  • 相关内容
  • 近期更新
  • 热文榜
  • 医学百科App—健康测试工具