Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
J Med Chem ; 56(19): 7527-35, 2013 Oct 10.
Article in English | MEDLINE | ID: mdl-24010887

ABSTRACT

Activated thrombin-activatable fibrinolysis inhibitor (TAFIa) is a zinc-containing carboxypeptidase and significantly inhibits fibrinolysis. TAFIa inhibitors are thus expected to act as profibrinolytic agents. We recently reported the design and synthesis of selenium-containing inhibitors of TAFIa and their inhibitory activity. Here we report the crystal structures of potent selenium-, sulfur-, and phosphinic acid-containing inhibitors bound to porcine pancreatic carboxypeptidase B (ppCPB). ppCPB is a TAFIa homologue and is surrogate TAFIa for crystallographic analysis. Crystal structures of ppCPB complexed with selenium compound 1a, its sulfur analogue 2, and phosphinic acid derivative EF6265 were determined at 1.70, 2.15, and 1.90 Å resolution, respectively. Each inhibitor binds to the active site of ppCPB in a similar manner to that observed for previously reported inhibitors. Thus, in complexes, selenium, sulfur, and phosphinic acid oxygen coordinate to zinc in ppCPB. This is the first observation and report of selenium coordinating to zinc in CPB.


Subject(s)
Carboxypeptidase B/antagonists & inhibitors , Organoselenium Compounds/chemistry , Phosphinic Acids/chemistry , Zinc/chemistry , Animals , Carboxypeptidase B/chemistry , Catalytic Domain , Crystallography, X-Ray , Molecular Docking Simulation , Protein Binding , Stereoisomerism , Swine
2.
Thromb Haemost ; 109(6): 1060-9, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23572161

ABSTRACT

Thrombin-activatable fibrinolysis inhibitor (TAFI, carboxypeptidase B2) is a 58-kDa plasma glycoprotein secreted by hepatocytes as an inactive form. TAFI is activated by the thrombin-thrombomodulin complex, and activated TAFI (TAFIa) plays an important role in regulating the balance between coagulation and fibrinolysis through inhibition of fibrinolysis. It has been suggested that high levels of TAFI in circulating plasma increase the risks of cardiovascular death and acute phase in ischaemic stroke. However, the mechanisms of regulating TAFI expression have been unclear. The present study investigated the effects of nobiletin (a polymethoxy flavonoid contained in the rind of citrus fruits) on TAFI gene (CPB2) and TAFI antigen expression in cultured human hepatoma HepG2 cells. Nobiletin decreased the release of TAFI antigen from HepG2 cells into conditioned medium in parallel with decreased levels of CPB2 mRNA and antigen. The half-life time of CPB2 mRNA in nobiletin-treated cells was unchanged compared to that of untreated control cells. Using nobiletin-treated cells that were transfected with a luciferase CPB2 promoter reporter plasmid, activity decreased to half of that in untreated control cells. A series of luciferase reporter constructs containing 5´-flanking region deletions of the human CPB2 gene showed that the sequences from -150 bp to -50 bp were essential for transcription of CPB2 and contained an AP-1 binding sequence at ~ -119 bp to - 99 bp in the CPB2 promoter. The amount of complexed nuclear protein and sequences from ~ -119 bp to -99 bp was decreased in nobiletin-treated cells. ChIP assays showed that c-Jun bound to the ~ -119 bp to -99 bp region of the CPB2 promoter and that the amount of the immunocomplex decreased after nobiletin treatment. Therefore, nobiletin-induced repression of CPB2 transcription might involve AP-1 inhibition and/or prevention of AP-1 binding in a specific region on the CPB2 gene in HepG2 cells.


Subject(s)
Antioxidants/chemistry , Carboxypeptidase B2/metabolism , Citrus/chemistry , Flavones/chemistry , Gene Expression Regulation , Anticoagulants , Binding Sites , Blood Coagulation , Fibrinolysis , Fruit/chemistry , Hep G2 Cells , Humans , Plasmids/metabolism , Promoter Regions, Genetic , Protein Binding , RNA, Messenger/metabolism , Signal Transduction , Time Factors , Transcription Factor AP-1/metabolism , Transcription, Genetic
3.
J Mol Cell Cardiol ; 56: 72-80, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23274063

ABSTRACT

Hyperphosphatemia has emerged as a cardiovascular risk factor that stimulates calcification in vessels. We explored molecules that were induced by inorganic phosphate (Pi) at an early stage in vascular smooth muscle cells (VSMC). In the present study, we examined the role of thrombomodulin (TM) in Pi-induced VSMC calcification based on the results of DNA microarray analysis. Both mRNA and protein expression of TM were markedly augmented in Pi-induced calcification. Conversely, knockdown of TM by siRNA significantly inhibited calcification, in addition to Pi-induced apoptosis which plays critical roles in VSMC calcification. We further found that TM suppressed both of mRNA and protein expression of growth arrest-specific gene 6 (Gas6), a key molecule regulating apoptosis. Recombinant extracellular epidermal growth factor (EGF)-repeat domain of TM exaggerated calcification and this effect was abrogated by a neutralizing antibody for EGF receptor, suggesting that the cleaved and secreted form of TM may activate EGF receptor. We also found that downregulation of Gas6 by TM/EGF receptor axis was mediated by ERK in VSMC calcification. In the aorta of adenine-fed rat, a typical medial calcification model with hyperphosphatemia, we found that TM expression was increased. Furthermore, in human calcified aorta, increased TM expression was also observed. These results indicate that TM is a novel molecule that promotes apoptosis and vascular calcification by regulation of Gas6, presumably via EGF receptor/ERK axis.


Subject(s)
Myocytes, Smooth Muscle/metabolism , Thrombomodulin/metabolism , Vascular Calcification/metabolism , Adult , Aged, 80 and over , Animals , Aorta/pathology , Apoptosis , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cells, Cultured , Down-Regulation , ErbB Receptors/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Expression , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , MAP Kinase Signaling System , Male , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Phosphates/pharmacology , Phosphates/physiology , Rats , Rats, Wistar , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology , Thrombomodulin/genetics , Thrombomodulin/physiology , Vascular Calcification/pathology
4.
J Med Chem ; 55(17): 7696-705, 2012 Sep 13.
Article in English | MEDLINE | ID: mdl-22891675

ABSTRACT

Available therapies for thromboembolic disorders include thrombolytics, anticoagulants, and antiplatelets, but these are associated with complications such as bleeding. To develop an alternative drug which is clinically safe, we focused on activated thrombin-activatable fibrinolysis inhibitor (TAFIa) as the target molecule. TAFIa is a zinc-containing carboxypeptidase that significantly inhibits fibrinolysis. Here we designed and synthesized selenium-containing compounds 5-13 to discover novel TAFIa inhibitors having a superior zinc-coordinating group. Compounds 5-13 significantly inhibited TAFIa activity (IC(50) 2.2 × 10(-12) M - 2.6 × 10(-6) M). We found that selenol is a better functional group than thiol for coordinating to zinc at the active site of TAFIa. Furthermore, compound 12, which has an amino-chloro-pyridine ring, was found to be a potent and selective TAFIa inhibitor that lacks carboxypeptidase N inhibitory activity. Therefore, compound 12 is a promising candidate for the treatment of thromboembolic disorders. This is the first report of a selenium-containing inhibitor for TAFIa.


Subject(s)
Carboxypeptidase B2/pharmacology , Drug Design , Selenium/analysis , Carboxypeptidase B2/chemistry , Magnetic Resonance Spectroscopy , Mass Spectrometry
5.
Thromb Res ; 130(4): e222-8, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22795218

ABSTRACT

Thrombin-activatable fibrinolysis inhibitor (TAFI) is a plasma zymogen that is activated by thrombin in plasma. In fibrinolytic processes, carboxy-terminal lysine (Lys) residues in partially degraded fibrin are important sites for plasminogen binding and activation, and an active form of TAFI (TAFIa) inhibits fibrinolysis by eliminating these residues proteolytically. We synthesized DD2 [7-Amino-2-(sulfanylmethyl)heptanoic acid], a Lys analogue containing sulfur, as an inhibitor of TAFIa and investigated its pharmacological profile and pathophysiological role in thrombolysis via in vitro and in vivo studies. DD2 specifically inhibited plasma TAFIa activity with an apparent IC(50) (50% inhibitory concentration) value of 3.4×10(-8)M under the present experimental condition and enhanced tissue plasminogen activator-mediated clot lysis in a concentration-dependent manner. In order to study tissue factor (TF)-induced microthrombosis in an animal model, rats were given intravenous injection (2.5mg/kg and higher) or oral administration (10mg/kg and higher) of DD2. This attenuated TF-induced glomerular fibrin deposition and increased the plasma levels of fibrin degradation products and D-dimer in a dose-dependent manner. A DD2 dose approximately 4X higher than the dose used in intravenous injections was required to achieve an equivalent thrombolytic effect to that seen following oral administration. Moreover, the oral absorption efficiency of DD2 into the vasculature was 29.8%. These results indicate that both intravenous and oral administration of DD2 enhanced endogenous fibrinolysis and reduced thrombi in a TF-induced microthrombosis model.


Subject(s)
Amino Acids/therapeutic use , Carboxypeptidase B2/antagonists & inhibitors , Fibrinolysis/drug effects , Fibrinolytic Agents/therapeutic use , Thrombosis/drug therapy , Administration, Intravenous , Administration, Oral , Amino Acids/administration & dosage , Amino Acids/chemistry , Animals , Carboxypeptidase B2/blood , Fibrinolytic Agents/administration & dosage , Fibrinolytic Agents/chemistry , Humans , Lysine/administration & dosage , Lysine/analogs & derivatives , Lysine/therapeutic use , Male , Rats , Sulfur Compounds/administration & dosage , Sulfur Compounds/chemistry , Sulfur Compounds/therapeutic use , Thromboplastin , Thrombosis/chemically induced , Thrombosis/pathology
6.
Thromb Haemost ; 108(1): 74-85, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22535242

ABSTRACT

Thrombin-activatable fibrinolysis inhibitor (TAFI) (carboxypeptidase B2) is a plasma zymogen that is biosynthesised in the liver and released into the circulation. Activated TAFI is a prothrombotic factor which inhibits fibrin clot lysis. Cultured human hepatoma HepG2 cells were treated with peroxisome proliferator-activated receptor (PPAR)α, ß or γ agonists, and the levels of TAFI antigen and mRNA (here, termed CPB2 mRNA) were measured. HepG2 cells treated with the PPARα agonist WY14643, but not agonists for PPARß or PPARγ, decreased their release of TAFI antigen into the conditioned medium. In parallel, there were decreased levels of CPB2 mRNA and TAFI antigen in the cells. The WY14643-mediated decrease in CPB2 mRNA levels was accelerated by overexpression of PPARα and abolished by RNA interference of PPAR A mRNA. CPB2 gene promoter activity was not influenced by treatment of the cells with WY14643. The half-life of the CPB2 transcript was shortened by treatment with WY14643 as compared with that of the control, and the decreased half-life of mRNA returned to control levels by treatment with a PPARα antagonist MK886 or transfection of PPAR A-specific siRNA to WY14643-treated HepG2 cells. The present results suggest that PPARα agonists not only play a hypolipidaemic role, but also decrease the expression of TAFI, a prothrombotic factor, by decreasing stability of CPB2 transcripts.


Subject(s)
Carboxypeptidase B2/antagonists & inhibitors , Fibrinolysis/drug effects , PPAR alpha/agonists , RNA Stability , Transcription, Genetic , Carboxypeptidase B/genetics , Carboxypeptidase B/metabolism , Carboxypeptidase B2/analysis , Carboxypeptidase B2/genetics , Cell Line, Tumor , Gene Expression Regulation , Hep G2 Cells , Humans , Indoles/pharmacology , PPAR alpha/genetics , PPAR gamma/agonists , PPAR-beta/agonists , Pyrimidines/pharmacology , RNA Precursors/genetics , RNA, Messenger/analysis , RNA, Messenger/genetics , RNA, Small Interfering/genetics
7.
Arch Toxicol ; 85(11): 1429-40, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21365311

ABSTRACT

The cytotoxic effects of hydroxylated fullerenes, also termed fullerenols or fullerols [C(60)(OH)( n )], which are known nanomaterials and water-soluble fullerene derivatives, were studied in freshly isolated rat hepatocytes. The exposure of hepatocytes to C(60)(OH)(24) caused not only concentration (0-0.25 mM)- and time (0-3 h)-dependent cell death accompanied by the formation of cell blebs, loss of cellular ATP, reduced glutathione (GSH), and protein thiol levels, but also the accumulation of glutathione disulfide and malondialdehyde, indicating lipid peroxidation. Of the other analogues examined, the cytotoxic effects of C(60)(OH)(12) and fullerene C(60) at a concentration of 0.125 mM were less than those of C(60)(OH)(24). The loss of mitochondrial membrane potential and generation of oxygen radical species in hepatocytes incubated with C(60)(OH)(24) were greater than those with C(60)(OH)(12) and fullerene C(60). In the oxygen consumption of mitochondria isolated from rat liver, the ratios of state-3/state-4 respiration were more markedly decreased by C(60)(OH)(24) and C(60)(OH)(12) compared with C(60). In addition, C(60)(OH)(24) and C(60)(OH)(12) resulted in the induction of the mitochondrial permeability transition (MPT), and the effects of C(60)(OH)(12) were less than those of C(60)(OH)(24). Taken collectively, these results indicate that (a) mitochondria are target organelles for fullerenols, which elicit cytotoxicity through mitochondrial failure related to the induction of the MPT, mitochondrial depolarization, and inhibition of ATP synthesis in the early stage and subsequently oxidation of GSH and protein thiols, and lipid peroxidation through oxidative stress at a later stage; and (b) the toxic effects of fullerenols may depend on the number of hydroxyl groups participating in fullerene in rat hepatocytes.


Subject(s)
Cytotoxins/toxicity , Fullerenes/toxicity , Hepatocytes/drug effects , Mitochondria, Liver/drug effects , Animals , Cell Death , Cell Respiration/drug effects , Cells, Cultured , Glutathione/metabolism , Glutathione Disulfide/metabolism , Hepatocytes/cytology , Hydroxylation , Lipid Peroxidation/drug effects , Male , Malondialdehyde/metabolism , Oxidative Stress , Rats , Rats, Inbred F344 , Reactive Oxygen Species/metabolism , Sulfhydryl Compounds/metabolism
8.
Chem Biol Interact ; 188(3): 393-403, 2010 Dec 05.
Article in English | MEDLINE | ID: mdl-20833158

ABSTRACT

The cytotoxic effects and biotransformation of harmine and harmaline, which are known ß-carboline alkaloids and potent hallucinogens, were studied in freshly isolated rat hepatocytes. The exposure of hepatocytes to harmine caused not only concentration (0-0.50mM)- and time (0-3h)-dependent cell death accompanied by the formation of cell blebs and the loss of cellular ATP, reduced glutathione, and protein thiols but also the accumulation of glutathione disulfide. Of the other analogues examined, the cytotoxic effects of harmaline and harmol (a metabolite of harmine) at a concentration of 0.5mM were less than those of harmine. The loss of mitochondrial membrane potential and generation of oxygen radical species in hepatocytes treated with harmine were greater than those with harmaline and harmol. In the oxygen consumption of mitochondria isolated from rat liver, the ratios of state-3/state-4 respiration of these ß-carbolines were decreased in a concentration-dependent manner. In addition, harmine resulted in the induction of the mitochondrial permeability transition (MPT), and the effects of harmol and harmaline were less than those of harmine. At a weakly toxic level of harmine (0.25mM), it was metabolized to harmol and its monoglucuronide and monosulfate conjugates, and the amounts of sulfate rather than glucuronide predominantly increased with time. In the presence of 2,5-dichloro-4-nitrophenol (50µM; an inhibitor of sulfotransferase), harmine-induced cytotoxicity was enhanced, accompanied by decrease in the amount of harmol-sulfate conjugate, due to an increase in the amount of unconjugated harmol and the inhibition of harmine loss. Taken collectively, these results indicate that (a) mitochondria are target organelles for harmine, which elicits cytotoxicity through mitochondrial failure related to the induction of the MPT, mitochondrial depolarization, and inhibition of ATP synthesis; and (b) the toxic effects of harmine are greater than those of either its metabolite harmol or its analogue harmaline, suggesting that the onset of harmine-induced cytotoxicity may depend on the initial and/or residual concentrations of harmine rather than on those of its metabolites.


Subject(s)
Harmaline/metabolism , Harmaline/toxicity , Harmine/metabolism , Harmine/toxicity , Hepatocytes/cytology , Mitochondria/drug effects , Mitochondria/metabolism , Animals , Biotransformation , Cell Respiration/drug effects , Glutathione/metabolism , Glutathione Disulfide/metabolism , Hallucinogens/metabolism , Hallucinogens/toxicity , Harmine/analogs & derivatives , Hepatocytes/drug effects , Hepatocytes/metabolism , Male , Malondialdehyde/metabolism , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/metabolism , Nitrophenols/pharmacology , Permeability/drug effects , Rats , Sulfhydryl Compounds/metabolism
9.
Thromb Haemost ; 102(6): 1204-11, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19967152

ABSTRACT

Thrombin-activatable fibrinolysis inhibitor (TAFI), a carboxypeptidase B-like proenzyme, is predominantly biosynthesised in the liver and released into circulating plasma. Activated TAFI has a role in maintaining the balance between blood coagulation and fibrinolysis. We investigated the regulation of TAFI expression in cultured human hepatoma HepG2 cells. Stimulation of the cells with forskolin and dibutyryl cyclic AMP (DBcAMP) increased TAFI antigen levels in the cells in parallel with TAFI mRNA levels and antigen release from the cells into the conditioned medium. The elevated TAFI expression was abolished by pretreatment of the cells with KT5720, a protein kinase A (PKA) inhibitor. The promoter activity of the TAFI gene and the half-life of the TAFI transcript in DBcAMP-stimulated HepG2 cells increased to 1.5-fold and 2.0-fold, respectively, of those in the control cells. The increased promoter activity and the prolonged half-life were abolished by pretreatment of the cells with KT5720. These results suggest that an increase in intracellular cAMP levels up-regulates TAFI expression in the cells in accompaniment with an elevation of TAFI mRNA levels, and that the elevated mRNA levels are derived from both transcriptional and post-transcriptional regulations of the TAFI gene mediated by activation of the AMP/PKA signaling pathway.


Subject(s)
Carboxypeptidase B2/genetics , Carboxypeptidase B2/metabolism , Cyclic AMP/metabolism , Base Sequence , Bucladesine/pharmacology , Carbazoles/pharmacology , Cell Line , Colforsin/pharmacology , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Cycloheximide/pharmacology , DNA Primers/genetics , Dactinomycin/pharmacology , Humans , Promoter Regions, Genetic , Protein Kinase Inhibitors/pharmacology , Pyrroles/pharmacology , RNA Processing, Post-Transcriptional/drug effects , RNA Stability , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction/drug effects , Transcription, Genetic/drug effects , Transfection , Up-Regulation/drug effects
10.
Chem Biol Interact ; 180(1): 89-97, 2009 Jun 15.
Article in English | MEDLINE | ID: mdl-19428348

ABSTRACT

The biotransformation and cytotoxic effects of hydroxychavicol (HC; 1-allyl-3,4-dihydroxybenzene), which is a catecholic component in piper betel leaf and a major intermediary metabolite of safrole in rats and humans, was studied in freshly isolated rat hepatocytes. The exposure of hepatocytes to HC caused not only concentration (0.25-1.0mM)- and time (0-3h)-dependent cell death accompanied by the loss of cellular ATP, adenine nucleotide pools, reduced glutathione, and protein thiols, but also the accumulation of glutathione disulfide and malondialdehyde, indicating lipid peroxidation. At a concentration of 1mM, the cytotoxic effects of safrole were less than those of HC. The loss of mitochondrial membrane potential and generation of oxygen radical species assayed using 2',7'-dichlorodihydrofluoresein diacetate (DCFH-DA) in hepatocytes treated with HC were greater than those with safrole. HC at a weakly toxic level (0.25 and/or 0.50mM) was metabolized to monoglucuronide, monosulfate, and monoglutathione conjugates, which were identified by mass spectra and/or (1)H nuclear magnetic resonance spectra. The amounts of sulfate rather than glucuronide or glutathione conjugate predominantly increased, accompanied by a loss of the parent compound, with time. In hepatocytes pretreated with either diethyl maleate or salicylamide, HC-induced cytotoxicity was enhanced, accompanied by a decrease in the formation of these conjugates and by the inhibition of HC loss. Taken collectively, our results indicate that (a) mitochondria are target organelles for HC, which elicits cytotoxicity through mitochondrial failure related to mitochondrial membrane potential at an early stage and subsequently lipid peroxidation through oxidative stress at a later stage; (b) the onset of cytotoxicity depends on the initial and residual concentrations of HC rather than those of its metabolites; (c) the toxicity of HC is greater than that of safrole, suggesting the participation of a catecholic intermediate in safrole cytotoxicity in rat hepatocytes.


Subject(s)
Anti-Infective Agents/toxicity , Eugenol/analogs & derivatives , Hepatocytes/drug effects , Safrole/chemistry , Animals , Anti-Infective Agents/metabolism , Apoptosis/drug effects , Biotransformation , Cells, Cultured , Chromatography, High Pressure Liquid , Eugenol/metabolism , Eugenol/toxicity , Male , Mitochondria/drug effects , Mitochondria/enzymology , Oxidative Stress/drug effects , Rats , Safrole/metabolism , Spectrometry, Mass, Electrospray Ionization
11.
Crit Care Med ; 37(5): 1744-9, 2009 May.
Article in English | MEDLINE | ID: mdl-19325462

ABSTRACT

OBJECTIVE: Although thrombin-activatable fibrinolysis inhibitor (TAFI) has been implicated as a negative regulator of fibrinolysis, its pathophysiological significance remains to be unveiled. We performed the pharmacologic study to assess the effect of EF6265, a specific inhibitor of activated form of TAFI (TAFIa) on sepsis-induced organ dysfunction models. DESIGN: A controlled, in vivo laboratory study. SETTING: Company research laboratory. SUBJECTS: Wistar and Sprague-Dawley rats. INTERVENTIONS: Endotoxemia and sepsis models were induced by intravenous injection of lipopolysaccharide and Pseudomonas aeruginosa, respectively. MEASUREMENTS AND MAIN RESULTS: In the endotoxemia model, posttreatment (1 hour) with EF6265 reduced fibrin deposits in the kidney and liver accompanied by no significant changes in platelet count and fibrinogen concentration in plasma. This compound also significantly decreased levels of plasma lactate dehydrogenase and aspartate aminotransferase, markers of organ dysfunction. In the sepsis model, EF6265, simultaneously administered with ceftazidime (CAZ) 2 hours after Pseudomonas aeruginosa injection, showed no influence on the antibiotic activity of CAZ. Meanwhile, it dramatically potentiated the interleukin-6-reducing effect of CAZ in plasma, suggesting that inhibition of TAFIa leads to the reduction in systemic inflammatory response associated with bacterial infection. This combined treatment also lowered plasma lactate dehydrogenase and blood urea nitrogen more potently than single treatment with CAZ. CONCLUSIONS: These results clearly suggest that TAFI plays an important role in the deterioration of organ dysfunction in sepsis and the inhibitor of TAFIa protects against sepsis-induced tissue damage through regulation of fibrinolysis and inflammation.


Subject(s)
Amino Acids/pharmacology , Carboxypeptidase B2/pharmacology , Multiple Organ Failure/prevention & control , Phosphinic Acids/pharmacology , Pseudomonas Infections/drug therapy , Sepsis/drug therapy , Analysis of Variance , Animals , Blood Chemical Analysis , Ceftazidime/pharmacology , Disease Models, Animal , Fibrinogen/drug effects , Fibrinogen/metabolism , Heparin/pharmacology , Interleukin-6/blood , Leukocyte Count , Lipopolysaccharides/pharmacology , Male , Multiple Organ Failure/drug therapy , Platelet Count , Probability , Pseudomonas Infections/blood , Random Allocation , Rats , Rats, Sprague-Dawley , Rats, Wistar , Sensitivity and Specificity , Sepsis/blood , Stem Cells
12.
Biol Pharm Bull ; 32(2): 179-85, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19182372

ABSTRACT

Cell malignancy is negatively correlated with the expression of thrombomodulin (TM), a thrombin receptor expressed on the surface of various cells, including tumor cells. TM accelerates thrombin-activatable fibrinolysis inhibitor (TAFI) activation catalyzed by thrombin. The active form of TAFI (TAFIa) contributes to inhibition of plasmin formation through its carboxypeptidase B (CPB)-like activity. Here, we examined whether TM- and tumor cell-dependent TAFI activation participates in controlling pericellular fibrinolysis and cell invasion. Human fibrosarcoma HT1080 cells retained the ability to activate both prothrombin and plasminogen, but did not express TM. HT1080 cells mediated activation of TAFI in plasma in the presence of soluble-type TM (sTM) through cell-dependent prothrombin activation. HT1080 cells stably expressing TM (TM-HT1080) mediated plasma TAFI activation without added sTM, but HT1080 (wild-type) and Mock-transfected HT1080 cells (Mock) did not. Production of TAFIa suppressed cell-mediated plasminogen activation. Matrigel invasion by wild-type and Mock cells was enhanced two-fold by diluted plasma (10%), whereas the plasma-induced invasion of TM-HT1080 cells (65% of wild-type invasion) was lower than those of wild-type and Mock cells. Cell invasion by TM-HT1080 was partially enhanced by addition of a TAFIa/CPB inhibitor. These results suggest that TM suppresses pericellular fibrinolysis and plasma-induced tumor cell invasion, and that it is mediated, at least in part, by plasma TAFI activation.


Subject(s)
Antineoplastic Agents , Carboxypeptidase B2/metabolism , Plasminogen Activators/antagonists & inhibitors , Thrombin/pharmacology , Thrombomodulin/physiology , Annexin A2/metabolism , Carboxypeptidase B2/drug effects , Cell Line, Tumor , Humans , Neoplasm Invasiveness , Prothrombin/metabolism , Receptors, Cell Surface
13.
Arch Toxicol ; 83(1): 69-80, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18553070

ABSTRACT

The amphetamine-derived designer drugs have been illegally used worldwide as recreational drugs, some of which are known to be hepatotoxic in humans. To compare their cytotoxic effects, 3,4-methylenedioxy-N-methamphetamine (MDMA) and its related analogues, N-methyl-1-(3,4-methylenedioxyphenyl)-2-butanamine (MBDB), 3,4-(methylenedioxyphenyl)-2-butanamine (BDB) and 2-methylamino-1-(3,4-methylenedioxyphenyl)-propane-1-one (methylone) were studied in freshly isolated rat hepatocytes. MBDB caused not only concentration (0-4.0 mM)- and time (0-2 h)-dependent cell death accompanied by the formation of cell blebs, and the loss of cellular ATP and adenine nucleotide pools, and reduced glutathione levels, but also the accumulation of oxidized glutathione. Of the other analogues examined, the cytotoxicity of MBDB and BDB was greater than that of MDMA and methylone, suggesting that hepatotoxicity is generally induced by these drugs. In addition, DNA damage and the induction of reactive oxygen species were greater after the incubation of hepatocytes with MBDB (2 and 4 mM) than after that with MDMA. In isolated liver mitochondria, MBDB/BDB resulted in a greater increase in the rate of state 4 oxygen consumption than did MDMA/methylone, indicating an uncoupling effect and a decrease in the rate of state 3 oxygen consumption in a concentration dependent manner. Furthermore, MBDB resulted in mitochondrial swelling dependent on the mitochondrial permeability transition (MPT); the effect of MDMA was less than that of MBDB. Taken collectively, these results suggest that (1) the onset of cytotoxicity caused by designer drugs such as MBDB and MDMA is linked to mitochondrial failure dependent upon the induction of the MPT accompanied by mitochondrial depolarization and depletion of ATP through uncoupling of oxidative phosphorylation in rat hepatocytes, and (2) MBDB and MDMA elicit DNA damage, suggesting that nuclei as well as mitochondria are target sites of these compounds.


Subject(s)
Hepatocytes/drug effects , Mitochondria, Liver/drug effects , N-Methyl-3,4-methylenedioxyamphetamine/toxicity , 3,4-Methylenedioxyamphetamine/administration & dosage , 3,4-Methylenedioxyamphetamine/analogs & derivatives , 3,4-Methylenedioxyamphetamine/toxicity , Adenosine Triphosphate/metabolism , Animals , Cell Death/drug effects , DNA Damage/drug effects , Hallucinogens/administration & dosage , Hallucinogens/toxicity , Hepatocytes/metabolism , Male , Methamphetamine/administration & dosage , Methamphetamine/analogs & derivatives , Methamphetamine/toxicity , Mitochondria, Liver/metabolism , N-Methyl-3,4-methylenedioxyamphetamine/administration & dosage , Oxidative Phosphorylation/drug effects , Rats , Rats, Inbred F344 , Time Factors
14.
Biochem Pharmacol ; 75(7): 1504-14, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18261712

ABSTRACT

Nobiletin is a citrus polymethoxylated flavonoid extracted from Citrus depressa, and has several reported biological effects. In this study, we investigated the effect of nobiletin on bacterial lipopolysaccharide (LPS)-induced expression of tissue factor (TF), a trigger protein for the blood coagulation cascade, and studied the possible mechanism of TF transcriptional regulation. THP-1 monocytic cells stimulated with LPS showed an increased expression of both TF protein and mRNA levels. However, pretreatment with nobiletin resulted in inhibition of LPS-induced expression of both TF protein and mRNA in a dose-dependent manner. Electrophoretic mobility shift assays revealed that binding of nuclear proteins from LPS-stimulated THP-1 cells to the NF-kappaB or AP-1 binding motif was increased as compared to non-stimulated control cells. Such increased binding activities were significantly reduced by pretreatment with nobiletin. Binding activity of nuclear proteins to the Sp1 binding motif was observed irrespective of LPS stimulation, but Sp1 activation was inhibited by nobiletin treatment of the cells. Treatment of THP-1 cells with Sp1-specific small interfering RNA (Sp1 siRNA) abolished the ability of LPS to induce TF activity. A similar reduction in the level of TF mRNA was also observed upon treatment of cells with Sp1 siRNA. These studies reveal that constitutive Sp1 activation is an essential event for transcriptional activation of TF, and nobiletin prevents LPS-induced TF expression by inhibiting NF-kappaB, AP-1, and Sp1 activation.


Subject(s)
Flavones/pharmacology , Monocytes/metabolism , NF-kappa B/antagonists & inhibitors , Sp1 Transcription Factor/antagonists & inhibitors , Thromboplastin/biosynthesis , Transcription Factor AP-1/antagonists & inhibitors , Animals , Cells, Cultured , Citrus , Flavones/isolation & purification , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Humans , Lipopolysaccharides/pharmacology , Monocytes/drug effects , NF-kappa B/metabolism , Plant Extracts/isolation & purification , Plant Extracts/pharmacology , Rabbits , Sp1 Transcription Factor/metabolism , Thromboplastin/genetics , Transcription Factor AP-1/metabolism
15.
Nephron Exp Nephrol ; 106(4): e113-21, 2007.
Article in English | MEDLINE | ID: mdl-17622739

ABSTRACT

BACKGROUND/AIM: Plasma carboxypeptidase B is a physiological fibrinolysis inhibitor. In the present study, the effects of EF6265, a novel specific plasma carboxypeptidase B inhibitor, on renal dysfunction in a rat thrombotic glomerulonephritis model were examined. METHODS: The model was induced by injection of anti-glomerular basement membrane serum and lipopolysaccharide to rats. Renal microthrombosis was histologically evaluated by phosphotungstic acid-hematoxylin staining for fibrin thrombi. Renal dysfunction was evaluated on the basis of plasma levels of blood urea nitrogen as well as renal edemas and urine volume. RESULTS: The glomerular microthrombi observed in a positive control group were significantly reduced after a short-term treatment (4 h) with EF6265 at a dose which enhanced fibrinolysis. The elevation of blood urea nitrogen and renal edema formation decreased, and the reduction of the urine volume improved after a long-term treatment (21 h) with EF6265. In addition, EF6265 had a protective activity against multiple organ dysfunction, because it reduced plasma lactate dehydrogenase and alanine aminotransferase levels and mortality in this model. CONCLUSION: EF6265, which inhibits plasma carboxypeptidase B, showed a protective effect on thrombotic renal dysfunction in thrombotic glomerulonephritis through enhancing the fibrinolysis.


Subject(s)
Amino Acids/administration & dosage , Carboxypeptidase B/antagonists & inhibitors , Carboxypeptidase B/blood , Fibrinolysis/drug effects , Glomerulonephritis/enzymology , Kidney/physiopathology , Phosphinic Acids/administration & dosage , Thrombosis/enzymology , Animals , Cytoprotection/drug effects , Dose-Response Relationship, Drug , Glomerulonephritis/drug therapy , Glomerulonephritis/pathology , Injections, Intravenous , Kidney/drug effects , Male , Rats , Rats, Wistar , Thrombosis/drug therapy , Thrombosis/pathology
16.
Chem Biol Interact ; 164(1-2): 93-101, 2006 Dec 01.
Article in English | MEDLINE | ID: mdl-17056023

ABSTRACT

The relationship between cytotoxicity induced by N-nitrosofenfluramine and mitochondrial or glycolytic adenosine triphosphate (ATP) synthesis-dependent intracellular bioenergetics was studied in isolated rat hepatocytes. The supplementation of fructose, an ATP-generating glycolytic substrate, to hepatocyte suspensions prevented N-nitrosofenfluramine-induced cell injury accompanied by the formation of cell blebs, abrupt loss of intracellular ATP and reduced glutathione and mitochondrial membrane potential (DeltaPsi), and the accumulation of oxidized glutathione and malondialdehyde, indicating lipid peroxidation, during a 2h incubation period. Fructose (1-20mM) resulted in concentration-dependent protection against the cytotoxicity of N-nitrosofenfluramine at a concentration of 0.6mM, a low toxic dose. Pretreatment with xylitol, another glycolytic substrate, at concentration of 15mM also prevented the cytotoxicity caused by the nitroso compound, but neither glucose nor sucrose exhibited protective effects. In addition, fructose inhibited N-nitrosofenfluramine (0.5 and 0.6mM)-induced DNA damage, as evaluated in the comet assay, indicating that nuclei as well as mitochondria are target sites of the compound. These results indicate that (a) the onset of N-nitrosofenfluramine-induced cytotoxicity in rat hepatocytes is linked to mitochondrial failure, and that (b) the insufficient supply of ATP in turn limits the activities of all energy-requiring reactions and consequently leads to acute cell death.


Subject(s)
Adenosine Triphosphate/pharmacology , Fenfluramine/analogs & derivatives , Glycolysis , Hepatocytes/drug effects , Membrane Potentials/drug effects , Mitochondria, Liver/drug effects , Animals , Cell Death/drug effects , Cells, Cultured , Comet Assay , DNA Damage , Dose-Response Relationship, Drug , Fenfluramine/metabolism , Fenfluramine/toxicity , Glutathione/metabolism , Hepatocytes/metabolism , Male , Membrane Potentials/physiology , Mitochondria, Liver/metabolism , Rats , Rats, Inbred F344 , Time Factors
17.
Biochim Biophys Acta ; 1762(9): 835-43, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16930953

ABSTRACT

Verotoxin (VT)-producing Escherichia coli (E. coli) O157:H7 infections are frequently complicated by thrombotic angiopathy, hemolytic uremic syndrome (HUS) and neurological symptoms. The present data demonstrate that VT-1 (Shiga toxin) stimulation of macrophage-like THP-1 cells up-regulates the activity, antigen and mRNA levels of tissue factor (TF), a key cofactor of the coagulation-inflammation-thrombosis circuit. This up-regulation is accompanied by phosphorylation of phosphatidylinositol 3-kinase (PI3-kinase), IkappaB kinase beta (IKKbeta) and extracellular signal-regulated kinase 2 (ERK2). Changes in TF mRNA levels were in parallel with the activation of NF-kappaB/Rel and Egr-1 activation, but not with AP-1. Inhibition of PI3-kinase attenuated VT-1-induced phosphorylation of IKKbeta and ERK2, and the up-regulation of TF mRNA levels. VT-1 stimulation rapidly activated c-Yes tyrosine kinase, a member of the Src family. Treatment of the cells with c-Yes antisense oligos attenuated the VT-1-induced phosphorylation of PI3-kinase, IKKbeta and ERK2, activations of NF-kappaB/Rel and Egr-1, and up-regulation of TF mRNA levels. These results suggest that VT-1-induced macrophage stimulation activates c-Yes, which then up-regulates TF expression through activation of the IKKbeta/proteasome/NF-kappaB/Rel and MEK/ERK2/Egr-1 pathways via activation of PI3-kinase. Induction of macrophage TF expression by VT-1 may play an important role in the acceleration of the coagulation-inflammation-thrombosis circuit during infections by VT-producing E. coli.


Subject(s)
Macrophages/metabolism , Proto-Oncogene Proteins c-yes/metabolism , RNA, Messenger/metabolism , Shiga Toxin 1/pharmacology , Thromboplastin/metabolism , Up-Regulation , Cell Differentiation , Cell Line, Tumor , Humans , Leukemia, Monocytic, Acute , Macrophages/drug effects , Signal Transduction , Transcription Factors/metabolism , Transcription, Genetic
18.
Bioorg Med Chem ; 14(15): 5099-109, 2006 Aug 01.
Article in English | MEDLINE | ID: mdl-16713268

ABSTRACT

9-cis-Retinoic acid (RA) analogues devised to lock the 9-cis double bond by ring formation were synthesized using two stereoselective carbon-carbon bond formation reactions as key steps. The palladium-mediated Suzuki reaction was adopted to construct a 7E-double bond (RA numbering) and the Horner-Emmons olefination was employed for stereoselective 11E-double bond (RA numbering) formation. The synthesized 9-cis-RA analogues that are locked by five-membered ring systems (cyclopentene, dihydrofuran, and dihydrothiophene) were shown to have comparable thrombomodulin induction activities to that of 9-cis RA. Conformational analysis of these compounds showed their similarity to 9-cis RA in the spatial orientation of the side chain and the terminal carboxy group.


Subject(s)
Endothelial Cells/drug effects , Endothelial Cells/metabolism , Thrombomodulin/biosynthesis , Tretinoin/chemical synthesis , Tretinoin/pharmacology , Alitretinoin , Catalysis , Cells, Cultured , Humans , Molecular Conformation , Palladium/chemistry , Stereoisomerism , Structure-Activity Relationship , Tretinoin/chemistry
19.
Eur J Pharmacol ; 529(1-3): 164-71, 2006 Jan 04.
Article in English | MEDLINE | ID: mdl-16316651

ABSTRACT

Factor Xa plays an important role in blood coagulation and is widely regarded as an attractive target for antithrombotic drug development. M55551 and M55165 (1-arylsulfonyl-3-piperazinone derivatives) are novel synthetic factor Xa inhibitors. In vitro, M55551 and M55165 competitively inhibited factor Xa with K(i) values of 3.2 nM and 2.3 nM, respectively, and prolonged clotting time in human and rat plasma. Pharmacokinetic analysis of these compounds revealed that M55551 was intravenously active with a short half-life (0.2 h) and that M55165 exhibited good bioavailability (31%) with a long half-life (3.9 h). Therefore, the antithrombotic effects of M55551 and M55165 were compared with those of the intravenous anticoagulant argatroban and the oral anticoagulant warfarin. Intravenous administration of M55551 and oral administration of M55165 inhibited thrombus formation at 0.3 mg/kg and 10 mg/kg, respectively, without significant prolongation of bleeding time. In contrast, although argatroban (0.3 mg/kg) and warfarin (1 mg/kg) also inhibited thrombus formation, significant prolongation of bleeding time was observed at dosages of 3 mg/kg and 1 mg/kg, respectively. These results suggest that M55551 and M55165 are potent factor Xa inhibitors that are active upon intravenous and oral administration, respectively, and that may prove clinically useful for the treatment of thrombosis while minimizing bleeding risks.


Subject(s)
Anticoagulants , Factor Xa Inhibitors , Piperazines , Venous Thrombosis/drug therapy , Administration, Oral , Animals , Anticoagulants/pharmacokinetics , Anticoagulants/pharmacology , Anticoagulants/therapeutic use , Biological Availability , Bleeding Time , Disease Models, Animal , Half-Life , Humans , Injections, Intravenous , Male , Piperazines/pharmacokinetics , Piperazines/pharmacology , Piperazines/therapeutic use , Rats , Rats, Wistar , Venous Thrombosis/blood
20.
J Pharmacol Exp Ther ; 309(2): 607-15, 2004 May.
Article in English | MEDLINE | ID: mdl-14762098

ABSTRACT

Plasma procarboxypeptidase B, also known as thrombin-activatable fibrinolysis inhibitor (TAFI), is converted by thrombin into the active enzyme, carboxypeptidase B (CPB)/activated TAFI. Plasma CPB down-regulates fibrinolysis by removing carboxy-terminal lysines, the ligands for plasminogen and tissue-type plasminogen activator (tPA), from partially degraded fibrin. To target thrombosis in a new way, we have identified and optimized a phosphinic acid-containing inhibitor of CPB, EF6265 [(S)-7-amino-2-[[[(R)-2-methyl-1-(3-phenylpropanoylamino) propyl]hydroxyphosphinoyl]methyl]heptanoic acid] and determined both the pharmacological profile and pathophysiological role of CPB in rat thrombolysis. EF6265 specifically inhibited plasma CPB activity with an IC(50) (50% inhibitory concentration) of 8.3 nM and enhanced tPA-mediated clot lysis in a concentration-dependent manner. EF6265 decreased detectable thrombi (percentage of glomerular fibrin deposition; control, 98 +/- 1.1; EF6265, 0.1 mg/kg, 27 +/- 9.1) that had been generated by tissue factor in a rat microthrombosis model with concomitant increases in plasma D-dimer concentration (control, <0.5 microg/ml; EF6265, 0.1 mg/kg, 15 +/- 3.5 microg/ml). EF6265 reduced plasma alpha2-antiplasmin activity to a lesser extent than tPA. In an arteriovenous shunt model, EF6265 (1 mg/kg) enhanced exogenous tPA-mediated thrombolysis under the same conditions that neither EF6265 nor tPA (600 kIU/kg) alone reduced thrombi. EF6265 (1 and 30 mg/kg) did not affect the bleeding time in rats. Moreover, it did not prolong the bleeding time evoked by tPA (600 kIU/kg). These results confirm that circulating procarboxypeptidase B functions as a fibrinolysis inhibitor's zymogen and validates the use of CPB inhibitors as both an enhancer of physiological fibrinolysis in microcirculation and as a novel adjunctive agent to tPA for thromboembolic diseases while maintaining a small effect on primary hemostasis.


Subject(s)
Amino Acids/pharmacology , Carboxypeptidase B2/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Fibrinolysis/drug effects , Phosphinic Acids/pharmacology , Amino Acids/pharmacokinetics , Amino Acids/therapeutic use , Animals , Arteriovenous Shunt, Surgical , Bleeding Time , Carboxypeptidase B2/metabolism , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/therapeutic use , Humans , Injections, Intravenous , Male , Models, Biological , Phosphinic Acids/pharmacokinetics , Phosphinic Acids/therapeutic use , Rats , Rats, Wistar , Thromboplastin , Thrombosis/chemically induced , Thrombosis/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...