Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
Add more filters










Publication year range
2.
Crit Care Med ; 29(7 Suppl): S53-60; discussion S60-1, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11445735

ABSTRACT

OBJECTIVE: To review the mechanisms of action and rationale for the use of recombinant human activated protein C in the treatment of severe sepsis. Specifically, we focus on the mechanisms of action in the protein C pathway that converge to modulate the pathophysiology of severe inflammatory disease and sepsis. This analysis includes a discussion of the role of activated protein C in directly modulating cell system biology, independent of antithrombotic activity. DATA SOURCES/STUDY SELECTION: Published research and review articles relating to the protein C pathway, recombinant human protein C, and the role of protein C in sepsis. Data were also derived from broad gene profiling in model systems of endothelial dysfunction. DATA EXTRACTION AND SYNTHESIS: Relevant studies were included to support discussion of the unique mechanistic aspect of protein C and its role in the pathogenesis of severe sepsis. We discuss the potential of activated protein C as a unique system modulator for the treatment of severe sepsis and other systemic inflammatory responses that result in microvascular coagulopathy, endothelial dysfunction, and vascular bed failure. CONCLUSIONS: The protein C pathway plays a unique role in modulating vascular function. As an antithrombotic/profibrinolytic agent, it plays a clear role in maintaining vascular patency. Moreover, it has anti-inflammatory properties and appears to play a unique role as an antiapoptotic and endothelial cell survival factor. In states of systemic inflammatory activation, loss of protein C due to consumptive processes results in a compromised ability to modulate coagulation as well as inflammatory and cell survival functions. This compromise leads to vascular dysfunction, end-organ failure, and death. Replacement with recombinant human activated protein C offers a system-modulating approach to improved outcome.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiology , Fibrinolytic Agents/therapeutic use , Protein C/therapeutic use , Recombinant Proteins/therapeutic use , Sepsis/therapy , Anti-Inflammatory Agents, Non-Steroidal/immunology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Apoptosis/drug effects , Apoptosis/physiology , Fibrinolytic Agents/immunology , Fibrinolytic Agents/pharmacology , Hemostasis/drug effects , Hemostasis/physiology , Humans , Inflammation , Multiple Organ Failure/microbiology , Protein C/immunology , Protein C/pharmacology , Recombinant Proteins/immunology , Recombinant Proteins/pharmacology , Sepsis/complications , Sepsis/immunology , Sepsis/physiopathology , Thrombosis/microbiology , Treatment Outcome
4.
Dev Growth Differ ; 43(2): 115-32, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11284962

ABSTRACT

A previously identified truncated form of the human Smad 6 gene containing a unique 12 amino acid motif at its N-terminus was studied. We have named this truncated form of the gene Smad 6s, for 'short-form', to distinguish it from the full-length form (Smad 6fl). Reverse transcription-polymerase chain reaction and immunohistochemistry revealed that Smad 6s has a unique pattern of expression in human coronary tissue and is upregulated in diseased heart tissue. We used the expression of human Smad 6s in Xenopus laevis as a model system to assess Smad 6s function. Injection of Smad 6fl RNA (4-cell embryos, 2 x ventral) produced tadpoles with partial secondary axes. In contrast, Smad 6s RNA injected in a similar manner produced tadpoles with a severe 'head-only' phenotype with no morphological appearance of a secondary axis. Mutant Smad 6s RNA lacking the unique 12 amino acids at the N-terminus of the Smad 6s isoform produced no embryonic phenotype, suggesting that this region is important in conferring biological activity. Ectodermal explant assays show that Smad 6s has activity consistent with being a BMP antagonist and can synergize with and enhance the activities of the activin and fibroblast growth factor pathways, all of which are novel findings in this study.


Subject(s)
Bone Morphogenetic Proteins/antagonists & inhibitors , DNA-Binding Proteins/physiology , Trans-Activators/physiology , Animals , Bone Morphogenetic Proteins/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Gene Expression Regulation, Developmental , Humans , Immunohistochemistry , Phenotype , Smad6 Protein , Trans-Activators/chemistry , Trans-Activators/genetics , Xenopus Proteins , Xenopus laevis/embryology
5.
J Biol Chem ; 276(14): 11199-203, 2001 Apr 06.
Article in English | MEDLINE | ID: mdl-11278252

ABSTRACT

Human protein C is a natural anticoagulant factor, and a recombinant activated form of the molecule (rhAPC) is completing clinical evaluation for treatment of severe sepsis. Because of the pathophysiologic role of endothelial dysfunction in severe inflammatory disease and sepsis, we explored the possibility that rhAPC might directly modulate endothelial function, independent of its anticoagulant activity. Using broad transcriptional profiling, we show that rhAPC directly modulates patterns of endothelial cell gene expression clustering into anti-inflammatory and cell survival pathways. rhAPC directly suppressed expression of p50 and p52 NFkappaB subunits, resulting in a functional decrease in NFkappaB binding at target sites. Further, rhAPC blocked expression of downstream NFkappaB regulated genes following tumor necrosis factor alpha induction, including dose-dependent suppression of cell adhesion expression and functional binding of intracellular adhesion molecule 1, vascular cell adhesion molecule 1, and E-selectin. Further, rhAPC modulated several genes in the endothelial apoptosis pathway, including the Bcl-2 homologue protein and inhibitor of apoptosis protein. These pathway changes resulted in the ability of rhAPC to inhibit the induction of apoptosis by the potent inducer, staurosporine. This new mechanistic understanding of endothelial regulation and the modulation of tumor necrosis factor-induced endothelial dysfunction creates a novel link between coagulation, inflammation, and cell death and provides insight into the molecular basis for the efficacy of APC in systemic inflammation and sepsis.


Subject(s)
Endothelium, Vascular/physiology , Gene Expression Regulation , Protein C/genetics , Apoptosis/genetics , Endothelium, Vascular/pathology , Gene Expression Profiling , Humans , Inflammation/genetics , Inflammation/pathology
6.
Thromb Haemost ; 86(6): 1563-72, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11776328

ABSTRACT

Plasminogen activator inhibitor-1 (PAI-1) is a serine protease inhibitor (SERPIN) specific for tissue-type and urokinase-like plasminogen activators. High plasma PAI-1 activity is a risk factor for thrombotic diseases. Due to the short half-life of PAI-1, regulation of PAI-1 gene expression and secretion of active PAI-1 into the blood stream is important for hemostatic balance. We have investigated transcriptional control of PAI-1 gene expression in bovine aortic endothelial cells (BAECs) and human cell lines using PAI-1 5' promoter-luciferase reporter assays. Contrary to the cytokine-induced up-regulation of PAI-1 mRNA and protein levels, we found that only transforming growth factor-beta (TGF-beta) was efficient in inducing PAI-1 promoter activation. Tissue necrosis factor-alpha (TNF-alpha) induced a small luciferase activity with the 2.5 kb PAI-1 promoter, but not with the PAI-800/4G/5G and p3TP-lux promoters. Next we investigated whether a lack of response to TNF-alpha was due to deficient signaling pathways. BAECs responded to TNF-alpha with robust NFkappaB promoter activation. TGF-beta activated the p38 MAP kinase, while TNF-alpha activated both the SAPK/JNK and p38 MAP kinases. The ERK1/2 MAP kinases were constitutively activated in BAECs. BAEC therefore responded to TNF-alpha stimulation with activation of the MAP kinases and the NFkappaB transcriptional factors. We further measured the messenger RNA stability under the influence by TGF-beta and TNF-alpha and found no difference. PAI-1 gene activation by TNF-alpha apparently is yet to be defined for the location of the response element and/or the signaling pathway, while TGF-beta is the most important cytokine for PAI-1 transcriptional activation through its 5' proximal promoter.


Subject(s)
Endothelium, Vascular/drug effects , Gene Expression Regulation/drug effects , Plasminogen Activator Inhibitor 1/genetics , Transforming Growth Factor beta/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Cells, Cultured/drug effects , Endothelium, Vascular/metabolism , Enzyme Activation/drug effects , Genes, Reporter , Humans , JNK Mitogen-Activated Protein Kinases , Luciferases/biosynthesis , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Plasminogen Activator Inhibitor 1/biosynthesis , Promoter Regions, Genetic/drug effects , RNA, Messenger/metabolism , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/pharmacology , Transcriptional Activation , Transforming Growth Factor beta1 , p38 Mitogen-Activated Protein Kinases
7.
Endocrinology ; 141(10): 3908-11, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11014248

ABSTRACT

Raloxifene is a nonsteroidal selective estrogen receptor modulator (SERM) that mimics the effects of estrogen on some plasma lipids and may have direct effects on the vascular wall. The objective of this study was to determine the effects of 17beta-estradiol, raloxifene, and LY139,478 (a related benzothiophene SERM) on the anticoagulant protein C pathway. In human vascular endothelial cells activated with interleukin-1 (IL-1), we demonstrated decreased thrombomodulin-dependent protein C activation. 17beta-estradiol reduced the anticoagulant properties of both unstimulated and IL-1-activated endothelial cells by decreasing thrombomodulin expression. In contrast, raloxifene and LY139,478 enhanced the anticoagulant properties of both unstimulated and IL-1-activated endothelial cells through upregulation of thrombomodulin. Regulation of the protein C pathway via thrombomodulin on vascular endothelium may be a novel mechanism by which SERMs could potentially confer cardioprotective effects and reduce the thrombotic risk associated with HRT in compromised patients.


Subject(s)
Estradiol/pharmacology , Estrogen Antagonists/pharmacology , Protein C/metabolism , Raloxifene Hydrochloride/pharmacology , Thrombomodulin/metabolism , Thrombosis/prevention & control , Cells, Cultured , Dose-Response Relationship, Drug , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Humans , Protein C/physiology , Pyrrolidines/pharmacology , Thiophenes/pharmacology , Umbilical Veins/cytology , Umbilical Veins/drug effects
8.
Peptides ; 21(8): 1231-6, 2000 Aug.
Article in English | MEDLINE | ID: mdl-11035210

ABSTRACT

Because plasma levels of protein C (PC) or activated protein C (APC) are altered in certain diseases associated with vascular dysfunction, and APC has therapeutic potential in preventing microvascular coagulation in severe sepsis, potential vascular effects of PC and APC were compared to those of the vasoactive peptide, thrombin. Thrombin was a more potent relaxant agonist than contractile agonist in aorta. Unlike thrombin, cumulatively administered APC (10(-9)-10(-7) M) did not exert vascular effects in rat or rabbit aorta. Noncumulative challenge of PC (10(-7) M) and APC (8 x 10(-8) M) also did not contract rat or rabbit aortae, either with or without endothelium. Likewise, the same concentrations of PC and APC also did not relax norepinephrine-induced (10(-7) M) vascular tone in either rat or rabbit aortae. Thus, in contrast to thrombin, PC and APC failed to modulate vascular tone, suggesting that the therapeutic use of APC is unlikely to be accompanied by any direct effects on vascular motility.


Subject(s)
Blood Vessels/drug effects , Protein C/pharmacology , Thrombin/pharmacology , Animals , Aorta/drug effects , Carbachol/pharmacology , Cardiotonic Agents/pharmacology , Dose-Response Relationship, Drug , Endothelium, Vascular/drug effects , Male , Potassium Chloride/pharmacology , Protein C/metabolism , Rabbits , Rats , Rats, Sprague-Dawley , Recombinant Proteins/metabolism , Thrombin/metabolism
10.
Exp Cell Res ; 238(1): 294-8, 1998 Jan 10.
Article in English | MEDLINE | ID: mdl-9457083

ABSTRACT

Thrombomodulin (TM) is a cell-surface receptor that plays a critical role in endothelial cell anticoagulant activity through its cofactor role in the thrombin-catalyzed activation of human protein C. In this study, we examined the effect of vascular endothelial growth factor (VEGF), a potent angiogenic factor, on surface anticoagulant activity and thrombomodulin expression. We show that thrombin-dependent activation of human protein C, measured on the endothelial cell surface, increased from 50 to 80% following exposure of cells to VEGF for 24 h. The effect was concentration dependent with the half-maximal stimulatory effect at approximately 100 pM. This increase in thrombin-dependent aPC generation correlated with a proportional and concentration-dependent increase in the level of cell-surface TM antigen. Both the total cellular TM antigen and the total cellular TM mRNA levels increased approximately 2.5-fold in VEGF-treated cells suggesting that most if not all of the regulation was at the message level. We further show that VEGF blocked IL-1 beta-induced suppression of both TM surface antigen and mRNA and was similarly capable of antagonizing the down-regulation of TM by TGF-beta and from cell activation by LPS. Our data suggest that VEGF regulation of TM may contribute to mechanisms that would maintain local hemostasis during angiogenesis and revascularization and could play a role in minimizing loss of vessel anticoagulant function during inflammatory processes.


Subject(s)
Endothelial Growth Factors/pharmacology , Endothelium, Vascular/drug effects , Lymphokines/pharmacology , Protein C/biosynthesis , Thrombomodulin/biosynthesis , Anticoagulants , Aorta , Cell Membrane/physiology , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/physiology , Gene Expression Regulation/drug effects , Humans , Interleukin-1/pharmacology , RNA, Messenger/biosynthesis , Thrombin/pharmacology , Transcription, Genetic/drug effects , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
11.
Cell ; 89(7): 1165-73, 1997 Jun 27.
Article in English | MEDLINE | ID: mdl-9215638

ABSTRACT

TGFbeta signaling is initiated when the type I receptor phosphorylates the MAD-related protein, Smad2, on C-terminal serine residues. This leads to Smad2 association with Smad4, translocation to the nucleus, and regulation of transcriptional responses. Here we demonstrate that Smad7 is an inhibitor of TGFbeta signaling. Smad7 prevents TGFbeta-dependent formation of Smad2/Smad4 complexes and inhibits the nuclear accumulation of Smad2. Smad7 interacts stably with the activated TGFbeta type I receptor, thereby blocking the association, phosphorylation, and activation of Smad2. Furthermore, mutations in Smad7 that interfere with receptor binding disrupt its inhibitory activity. These studies thus define a novel function for MAD-related proteins as intracellular antagonists of the type I kinase domain of TGFbeta family receptors.


Subject(s)
Carrier Proteins/genetics , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/physiology , Trans-Activators , Transforming Growth Factor beta/physiology , Amino Acid Sequence , Animals , COS Cells , Carrier Proteins/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Endothelium, Vascular/chemistry , Endothelium, Vascular/cytology , Humans , Liver Neoplasms , Molecular Sequence Data , Phosphorylation , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Signal Transduction/drug effects , Smad2 Protein , Tumor Cells, Cultured , Umbilical Veins/cytology
13.
Blood ; 89(2): 534-40, 1997 Jan 15.
Article in English | MEDLINE | ID: mdl-9002956

ABSTRACT

Conversion by alpha-thrombin of the zymogen human protein C (HPC) to activated protein C (aPC) is an important physiologic feedback control mechanism for the coagulation cascade. Although activation of HPC by thrombomodulin-bound thrombin is relatively rapid, activation by free thrombin occurs at a significantly slower rate. Previously, we generated a "hyper-activatable" derivative of HPC (FLIN-Q3) with an increased activation rate by free alpha-thrombin in vitro. In this study, the antithrombotic efficacy of FLIN-Q3 was compared with both native zymogen and aPC in an arteriovenous shunt model of thrombosis in the guinea pig. Recombinant proteins were infused 15 minutes before and throughout a 15-minute period while blood was circulated from carotid to jugular through tubing that enclosed a thread on which fibrin was deposited. Parallel dose-dependent antithrombotic responses were observed. Under these non-steady-state conditions, the calculated infusion doses associated with a 50% reduction of thrombus mass were 2.7, 24, and 250 mg/kg/h for aPC, FLIN-Q3, and HPC, respectively. Thrombus weight correlated inversely with plasma concentration of aPC, measured amidolytically, from either direct infusion of aPC or that generated from the zymogens in the animal, and similarly correlated inversely with anticoagulant activity measured by whole blood aPTT. Neither zymogen form showed significant aPC activity before shunt circulation, suggesting a requirement for exposure to thrombin. After the infusion was discontinued for 15 minutes, a second period of thrombus formation in the shunt demonstrated the ability of zymogen forms of PC, unlike aPC, to provide "on-demand" anticoagulant responses to repeated thrombotic stimuli. Thus, a "hyper-activatable" PC molecule such as FLIN-Q3 may represent a superior form of anticoagulant therapy than either the native zymogen or aPC.


Subject(s)
Anticoagulants/administration & dosage , Protein C/administration & dosage , Thrombin/metabolism , Thrombosis/drug therapy , Animals , Guinea Pigs , Humans , Male , Protein C/analogs & derivatives , Protein C/genetics , Recombinant Proteins/administration & dosage , Recombinant Proteins/genetics , Thrombosis/metabolism
14.
J Biol Chem ; 271(37): 22285-8, 1996 Sep 13.
Article in English | MEDLINE | ID: mdl-8798383

ABSTRACT

Activated protein C (aPC) is an important feedback regulator of the clotting cascade. In vivo, the conversion of protein C (PC) from its zymogen to activated form is mediated primarily by thrombin bound to thrombomodulin (TM), an endothelial cell surface protein. Molecular modeling suggests residues Lys37-Lys38-Lys39 of protein C's serine protease domain reside in a surface-exposed loop (variable region 1) whose high concentration of positive charge might be involved in protein-protein interactions. In this study, we have examined the role of the conserved tribasic Lys37-39 charge center in human protein C activation. This sequence was changed to acidic by substitution with Asp37-Glu38-Asp39 (DED) and Glu37-Glu38-Glu39 (EEE), or to neutrality by substitution with Gly37-Gly38-Gly39 (GGG). These mutant PCs, expressed and purified from recombinant human 293 cells, appeared normal with regard to intracellular processing, ability to be secreted, and formation of a viable active site for tripeptidyl-p-nitroanilide substrate cleavage. For activation by free thrombin, wild-type (wt) and mutant PCs displayed equivalent activation rates, as well as identical calcium-dependent inhibition of such activation. Activation of wt-PC with a soluble TM-thrombin complex yielded a 2,000-fold faster rate compared with that by free thrombin at the same (physiological) calcium level. In contrast, the acidic mutants DED and EEE exhibited virtually no TM-mediated increase in activation rate, while the neutral mutant GGG was somewhat intermediate with a 30-fold stimulation of activation rate. These reductions in activation rate were independent of the presence of chondroitin sulfate on TM. Our observations represent the first identification of residues whose mutation essentially uncouples activation by the TM-thrombin complex without affecting activation by free thrombin. Further, our results suggest that VR1 residues within the zymogen form of a serine protease can be important for recognition by physiological activators.


Subject(s)
Lysine/metabolism , Protein C/metabolism , Thrombin/metabolism , Thrombomodulin/metabolism , Amino Acid Sequence , Calcium/metabolism , Electrophoresis, Polyacrylamide Gel , Enzyme Activation , Humans , Kinetics , Lysine/genetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Binding , Protein C/genetics , Sequence Homology, Amino Acid , Serine Endopeptidases/chemistry , Structure-Activity Relationship
15.
Science ; 273(5280): 1389-91, 1996 Sep 06.
Article in English | MEDLINE | ID: mdl-8703074

ABSTRACT

A modulator of the enzymatic activity of human thrombin, designated LY254603, was identified that enhances the thrombin-catalyzed generation of the anticoagulant factor activated protein C, yet inhibits thrombin-dependent fibrinogen clotting. By means of mutant substrates, it was shown that LY254603 mediates the change in enzymatic substrate specificity through an alteration in thrombin's S3 substrate recognition site, a mechanism that appeared to be independent of allosteric changes induced by either sodium ions or by thrombomodulin. This compound may represent the prototype of a class of agents that specifically modulates the balance between thrombin's procoagulant and anticoagulant functions.


Subject(s)
Fibrinogen/metabolism , Naphthalenes/pharmacology , Phenyl Ethers/pharmacology , Protein C/metabolism , Thrombin/pharmacology , Blood Coagulation/drug effects , Calcium/pharmacology , Choline/pharmacology , Dose-Response Relationship, Drug , Enzyme Activation , Humans , Naphthalenes/chemistry , Partial Thromboplastin Time , Phenyl Ethers/chemistry , Protein C/chemistry , Protein Conformation , Recombinant Proteins/metabolism , Sodium Chloride/pharmacology , Substrate Specificity/drug effects , Thrombomodulin/metabolism
16.
J Lipid Res ; 37(5): 1162-6, 1996 May.
Article in English | MEDLINE | ID: mdl-8725167

ABSTRACT

Cholesteryl ester transfer protein (CETP) mediates the exchange of phospholipids and neutral lipids between the plasma lipoproteins, and plays an important role in high density lipoprotein (HDL) metabolism. While there are reports of low-level CETP secretion from cultured cells, the lack of a good model cell line has hampered the detailed study of CETP regulation and secretion. In this study, we have found that the human liposarcoma cell line, SW872, secretes cholesteryl ester transfer protein at levels substantially higher than observed from other cell lines. The secretion of CETP from this adipose-derived cell was up-regulated by 25-OH cholesterol and by low density lipoprotein (LDL) cholesterol in a concentration-dependent manner. Analysis of both full length and exon 9-deleted CETP mRNA demonstrated increases in response to LDL and 25-OH cholesterol, providing evidence for regulation at the message level. Our results suggest that the CETP-producing SW872 cell line may provide a model in which to study the regulation of this important modulator of lipoprotein metabolism.


Subject(s)
Adipocytes/metabolism , Carrier Proteins/metabolism , Cholesterol/pharmacology , Glycoproteins/metabolism , Carrier Proteins/genetics , Cell Line , Cholesterol Ester Transfer Proteins , Humans , Hydroxycholesterols/pharmacology , Lipoproteins, LDL/pharmacology , Liposarcoma , RNA, Messenger/biosynthesis
17.
J Pharmacol Exp Ther ; 276(3): 1104-10, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8786541

ABSTRACT

Myocardial infarct size has been measured after 1 hr of mechanical occlusion of the circumflex coronary artery and 5 hr of reperfusion in control dogs infused with saline, and in dogs infused with activated protein C (aPC) (1mg/kg/hr i.v.). Infusion of aPC during reperfusion produced a sustained doubling of activated partial thromboplastin time and no change in thrombin time at a final plasma parent drug concentration of 1.25 +/- 0.11 mug/ml. aPC infusion did not alter systolic arterial pressure, cardiac rate or the rate pressure product when compared to time-related alterations observed in control dogs. ST-segment deviation and the intensity and duration of cardiac arrhythmias associated with reperfusion of ischemic myocardium also were similar between groups. Resultant infarct sizes were 34.8 +/- 3.9 and 33.2 +/- 6.2% of the left ventricular mass placed at risk of necrosis in control and aPC-treated dogs. respectively. aPC infusion was associated with a small reduction in leukocytosis in response to myocardial ischemic injury, but did not alter the localization of leukocytes within ischemic and infarcted myocardium. In vitro concentrations of aPC (0.3, 1 and 3 mug/ml), comparable to the plasma concentration that inhibited blood coagulation in dogs, did not alter superoxide production or CD11b/CD18-mediated adhesion of chemotactic factor f-Met-Leu-Phe-stimulated neutrophils. Present data indicate that aPC lacks cardioprotectant activity at an infusion rate inhibiting coagulation. Apart from inhibition of thrombin generation, no evidence of an anti-inflammatory effect of aPC was observed.


Subject(s)
Myocardial Infarction/drug therapy , Myocardial Reperfusion , Protein C/pharmacology , Animals , Blood Pressure/drug effects , Cell Adhesion/drug effects , Disease Models, Animal , Dogs , Dose-Response Relationship, Drug , Male , Time Factors
18.
J Biol Chem ; 271(9): 4589-92, 1996 Mar 01.
Article in English | MEDLINE | ID: mdl-8617716

ABSTRACT

Human apolipoprotein E is a plasma lipoprotein that appears to play an important protective role in the development of atherosclerosis. While little is known about the regulation of apoE, recent studies have shown that cytokines repress apoE synthesis both in vivo and in vitro. Furthermore, we have recently shown that the endogenous apoE gene is negatively regulated by the nuclear trans-repressor BEF-1 in the human HepG2 cell line. In this study we demonstrate that treatment of HepG2 cells with the cytokine interleukin-1 and interleukin-6 resulted in the induction of an isoform of BEF-1, designated B1. The induction of the B1 isoform could be blocked by the protein kinase inhibitor staurosporine, suggesting that B1 is a phosphorylated form of BEF-1. As further support, the B1 isoform could also be induced by phorbol ester, and subsequently inhibited by staurosporine, implicating a role for protein kinase C-mediated phosphorylation. Quantitation of the levels of the BEF-1 isoforms, and studies in the presence of cyclohexamide, provided evidence for the phosphorylation of an existing intracellular pool of BEF-1, with no change in the total intracellular level. Under conditions that generated increased levels of the B1 isoform, there was a concomitant and proportional decrease in the level of apoE mRNA. The effect did not appear to be the result of improved binding to the apoE regulatory region as the DNA binding affinity of B1 was identical to native BEF-1. Our data suggest that the regulation of apoE by BEF-1 is modulated by differential phosphorylation, possibly through the protein kinase C pathway.


Subject(s)
Apolipoproteins E/biosynthesis , Gene Expression Regulation, Neoplastic , Interleukin-1/pharmacology , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Apolipoproteins E/genetics , Carcinoma, Hepatocellular , Cell Line , Cycloheximide/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Kinetics , Liver Neoplasms , Phosphorylation , Protein Kinase C/metabolism , RNA, Messenger/biosynthesis , Regulatory Sequences, Nucleic Acid , Tumor Cells, Cultured
19.
Am J Physiol ; 270(2 Pt 2): H603-9, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8779836

ABSTRACT

Vascular smooth muscle cells produce the proteolytically activated thrombin receptor. Under certain conditions, they have been reported to synthesize thrombomodulin (TM), another thrombin receptor known to convert the specificity of thrombin from cleavage of procoagulant/proinflammatory substrates to the cleavage of the anticoagulant/anti-inflammatory factor protein C. In this study, we examined the role of TM in modulating thrombin-mediated cellular responses. Using a thrombin receptor-positive TM-negative rabbit intimal smooth muscle cell line (RIC), we isolated cells expressing varying levels of functional surface TM after transfection with an expression vector containing the cDNA for full-length TM. The parent RIC (TM negative) line responded to alpha-thrombin and to agonist peptide (SFLLRN-PNDKYEPF; abbreviated SFLL) with both mitogenic response and phosphoinositol release. However, transfected cells producing high levels of TM, equivalent to the level on rabbit aortic endothelial cells, responded to SFLL but not to alpha-thrombin. Whereas alpha-thrombin, SFLL, and the combination of SFLL and thrombin resulted in a mitogenic response in the TM-negative RIC line, the response to the agonist peptide could be blocked by thrombin in the TM-producing cell line. The degree to which thrombin receptor activation was blocked directly correlated with the level of TM on the cell surface, and high levels of thrombin could overcome the inhibitory effect. Our data demonstrate that the coexpression of TM with thrombin receptor on vascular smooth muscle cells can result in a modulation of cellular responses to thrombin, which could control thrombin-induced proliferative events following vessel injury or insult.


Subject(s)
Muscle, Smooth, Vascular/metabolism , Receptors, Thrombin/physiology , Thrombomodulin/physiology , Animals , Cell Line , Cell Membrane/metabolism , Dose-Response Relationship, Drug , Drug Combinations , Inositol Phosphates/metabolism , Mitogens/pharmacology , Muscle, Smooth, Vascular/cytology , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/pharmacology , Rabbits , Receptors, Thrombin/antagonists & inhibitors , Recombinant Proteins , Thrombin/pharmacology , Transfection
20.
J Biol Chem ; 270(26): 15447-50, 1995 Jun 30.
Article in English | MEDLINE | ID: mdl-7797534

ABSTRACT

Apolipoprotein E (apoE) is a major constituent of plasma lipoprotein that functions in lipid transport and redistribution (reverse cholesterol transport) and probably plays an important role in inhibiting the development and/or progression of atherosclerosis. While cis-acting regions involved in basal and tissue-specific control of the apoE gene have been identified by promoter mapping studies, much less is known about factors that regulate the gene. In this study, we demonstrate that the region between -94 and -84 upstream of transcriptional start site of the human apoE gene contains a binding site for the transcriptional repressor factor BEF-1, a tyrosine-phosphorylated nuclear protein that was first identified in HeLa cells. Using gel retardation assays, we show that HeLa cell-derived BEF-1 binds the apoE BEF-1 homology, and this binding can be competed with the prototype BEF-1 sequence, but not by a mutated sequence. Furthermore, we demonstrate that the apoE- producing human liver HepG2 cell produces significant levels of BEF-1, which could bind to both the prototype BEF-1 sequence and the apoE homology, and be competed equivalently with cold BEF-1 or apoE homology. To determine if BEF-1 affected the expression of apoE, we performed competition experiments using plasmids containing the intact or mutated BEF-1 homology. The introduction of the intact BEF-1 site into HepG2 cells resulted in an induction of apoE mRNA, whereas control and mutated BEF-1-containing plasmids had no significant effect. We also found that increasing the level of nuclear BEF-1 by treatment of cells with orthovanadate resulted in a reduction in the level of apoE mRNA. Overall, our data suggest that the endogenous apoE gene in the human HepG2 cell line is repressed by the trans-acting influence of nuclear factor BEF-1.


Subject(s)
Apolipoproteins E/genetics , Gene Expression Regulation , Liver/metabolism , Repressor Proteins/physiology , Base Sequence , Binding Sites , Humans , Molecular Sequence Data , RNA, Messenger/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...