Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters











Publication year range
1.
Acta Cir Bras ; 34(1): e20190010000006, 2019 Feb 14.
Article in English | MEDLINE | ID: mdl-30785507

ABSTRACT

PURPOSE: To investigate whether hirudin exerts its antithrombin action to decrease the ratio of Human Microvascular Endothelial Cells (HMVECs) apoptosis. METHODS: Human microvascular endothelial cells (HMVECs) cultured in the third and fifth generations were used. HMVECs were divided into normal group, thrombin group (T group), natrual hirudin group (H group), thrombin + natrual hirudin group (T + H group), AG490 group, thrombin + AG490 group (T + AG490 group), natrual hirudin + AG490 group (H + AG490 group), thrombin + natural hirudin + AG490 (T + H + AG490 group).Apart from the normal group, the other groups were exposed to the relevant drugs for 24 hours.HMVEC apoptosis was assessed by flow cytometric and double Immunofluorescence of phosphorylation of JAK (P-JAK2) and TUNEL assay. RESULTS: Compared with the normal group, in thrombin group the HMVECs apoptosis rate were significantly increased (P<0.05).The results indicated that the index of apoptosis and the apoptosis rate were improved in cultures treated by natural hirudin (T + H group), relative to cultures with thrombin only (T group). We found that the index of apoptosis and the apoptosis rate in the AG490 + thrombin group were higher than that in the hirudin + thrombin group (P<0.05). Double Immunofluorescence of p-JAK2 and TUNEL assays showed that cells were double positive for P-JAK2 uptake and TUNEL detection liquid binding. CONCLUSION: The natural hirudin and JAK2/STATs signal inhibitor AG490 could block the effects of thrombin. Natural hirudin could attenuate HMVECs apoptosis via antagonizing thrombin and it is suggested that this effect may occur by blocking the JAK2/STATs signaling pathway and this signaling pathways appears to be not the only pathway.


Subject(s)
Antithrombins/pharmacology , Apoptosis/drug effects , Endothelial Cells/drug effects , Hirudins/pharmacology , Protein Kinase Inhibitors/pharmacology , Thrombin/drug effects , Cell Proliferation/drug effects , Humans , Microvessels/drug effects , Microvessels/metabolism , Signal Transduction/drug effects
2.
Acta cir. bras ; Acta cir. bras;34(1): e20190010000006, 2019. graf
Article in English | LILACS | ID: biblio-983690

ABSTRACT

Abstract Purpose: To investigate whether hirudin exerts its antithrombin action to decrease the ratio of Human Microvascular Endothelial Cells (HMVECs) apoptosis. Methods: Human microvascular endothelial cells (HMVECs) cultured in the third and fifth generations were used. HMVECs were divided into normal group, thrombin group (T group), natrual hirudin group (H group), thrombin + natrual hirudin group (T + H group), AG490 group, thrombin + AG490 group (T + AG490 group), natrual hirudin + AG490 group (H + AG490 group), thrombin + natural hirudin + AG490 (T + H + AG490 group).Apart from the normal group, the other groups were exposed to the relevant drugs for 24 hours.HMVEC apoptosis was assessed by flow cytometric and double Immunofluorescence of phosphorylation of JAK (P-JAK2) and TUNEL assay. Results: Compared with the normal group, in thrombin group the HMVECs apoptosis rate were significantly increased (P<0.05).The results indicated that the index of apoptosis and the apoptosis rate were improved in cultures treated by natural hirudin (T + H group), relative to cultures with thrombin only (T group). We found that the index of apoptosis and the apoptosis rate in the AG490 + thrombin group were higher than that in the hirudin + thrombin group (P<0.05). Double Immunofluorescence of p-JAK2 and TUNEL assays showed that cells were double positive for P-JAK2 uptake and TUNEL detection liquid binding. Conclusion: The natural hirudin and JAK2/STATs signal inhibitor AG490 could block the effects of thrombin. Natural hirudin could attenuate HMVECs apoptosis via antagonizing thrombin and it is suggested that this effect may occur by blocking the JAK2/STATs signaling pathway and this signaling pathways appears to be not the only pathway.


Subject(s)
Humans , Thrombin/drug effects , Antithrombins/pharmacology , Hirudins/pharmacology , Apoptosis/drug effects , Endothelial Cells/drug effects , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Cell Proliferation/drug effects , Microvessels/drug effects , Microvessels/metabolism
3.
Rev. méd. Chile ; 144(9): 1103-1111, set. 2016. graf, tab
Article in Spanish | LILACS | ID: biblio-830618

ABSTRACT

Background: Atrial fibrillation (AF) generates a hypercoagulable state with an increased thrombin generation and raised levels of thrombin-antithrombin complexes, which results in a high risk of stroke and thromboembolism. Aim: To evaluate the anticoagulant effect of rivaroxaban by anti-Xa factor activity and its correlation with thrombin-antithrombin complexes, thrombin generation and prothrombin time in patients newly diagnosed with non-valvular AF. Patients and Methods: Prospective study in patients with indication of anticoagulation. Demographic variables, cardiovascular risk factors, CHA2DS2-VASc and HAS-BLED scores were recorded. Blood samples were taken at baseline, at 3 and 24 hours after the administration of the drug and at 30 days. Rivaroxaban levels, anti-Xa activity, prothrombin time, thrombin generation and plasma levels of thrombin-antithrombin complexes were determined. Results: We studied 20 patients aged 76.3 ± 8.0 years (60% female) with a CHA2DS2-VASc score > 2 points. The anti-Xa factor activity correlated with rivaroxaban plasma levels at 3 hours (r = 0.61, p < 0.01), at 24 hours (r = 0.85, p < 0.01) and at 30 days (r = 0.99, p < 0.01), with prothrombin time at 3 hours (r = -0.86, p = 0.019) and at 30 days (r = -0.63, p = 0.02) and with a sustained decrease in thrombin generation at 30 days of follow-up (r = -0.74, p < 0.01). There was no correlation with thrombin-antithrombin complexes (r = -0.02, p = 0.83). Conclusions: Rivaroxaban consistently inhibited the mild pro-coagulant state found in newly diagnosed non-valvular AF patients through the first 24 hours and this effect was maintained at 30 days. Plasma levels of the drug correlated with anti-Xa factor activity, thrombin generation and prothrombin time


Subject(s)
Humans , Male , Female , Aged , Aged, 80 and over , Peptide Hydrolases/drug effects , Atrial Fibrillation/blood , Thrombin/drug effects , Factor Xa/drug effects , Antithrombin III/drug effects , Factor Xa Inhibitors/pharmacology , Rivaroxaban/pharmacology , Prothrombin Time , Time Factors , Thrombin/metabolism , Factor Xa/metabolism , Administration, Oral , Prospective Studies
4.
Rev Med Chil ; 144(9): 1103-1111, 2016 Sep.
Article in Spanish | MEDLINE | ID: mdl-28060970

ABSTRACT

BACKGROUND: Atrial fibrillation (AF) generates a hypercoagulable state with an increased thrombin generation and raised levels of thrombin-antithrombin complexes, which results in a high risk of stroke and thromboembolism. AIM: To evaluate the anticoagulant effect of rivaroxaban by anti-Xa factor activity and its correlation with thrombin-antithrombin complexes, thrombin generation and prothrombin time in patients newly diagnosed with non-valvular AF. PATIENTS AND METHODS: Prospective study in patients with indication of anticoagulation. Demographic variables, cardiovascular risk factors, CHA2DS2-VASc and HAS-BLED scores were recorded. Blood samples were taken at baseline, at 3 and 24 hours after the administration of the drug and at 30 days. Rivaroxaban levels, anti-Xa activity, prothrombin time, thrombin generation and plasma levels of thrombin-antithrombin complexes were determined. RESULTS: We studied 20 patients aged 76.3 ± 8.0 years (60% female) with a CHA2DS2-VASc score > 2 points. The anti-Xa factor activity correlated with rivaroxaban plasma levels at 3 hours (r = 0.61, p < 0.01), at 24 hours (r = 0.85, p < 0.01) and at 30 days (r = 0.99, p < 0.01), with prothrombin time at 3 hours (r = -0.86, p = 0.019) and at 30 days (r = -0.63, p = 0.02) and with a sustained decrease in thrombin generation at 30 days of follow-up (r = -0.74, p < 0.01). There was no correlation with thrombin-antithrombin complexes (r = -0.02, p = 0.83). CONCLUSIONS: Rivaroxaban consistently inhibited the mild pro-coagulant state found in newly diagnosed non-valvular AF patients through the first 24 hours and this effect was maintained at 30 days. Plasma levels of the drug correlated with anti-Xa factor activity, thrombin generation and prothrombin time.


Subject(s)
Antithrombin III/drug effects , Atrial Fibrillation/blood , Factor Xa Inhibitors/pharmacology , Factor Xa/drug effects , Peptide Hydrolases/drug effects , Rivaroxaban/pharmacology , Thrombin/drug effects , Administration, Oral , Aged , Aged, 80 and over , Factor Xa/metabolism , Female , Humans , Male , Prospective Studies , Prothrombin Time , Thrombin/metabolism , Time Factors
5.
Arch Biochem Biophys ; 482(1-2): 25-32, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19101499

ABSTRACT

alphaIIbbeta3 is an integrin that is involved in platelet adhesion and aggregation. This receptor may be inhibited by cysteine-rich peptides known as disintegrins. We isolated two disintegrins from Bothrops jararaca venom called jarastatin and jararacin. We evaluated the structural characteristics and the effects on human platelet aggregation of these disintegrins. Inhibitory profiles were compared to six distinct peptides synthesized based on their RGD hairpin loop primary sequences. Both jarastatin and jararacin inhibited ADP and thrombin induction. Conversely, none of the cyclic peptides showed high-quality activity in assays induced by ADP or thrombin. We constructed homology models for all of these molecules, and theoretically evaluated their interaction with the alphaIIbbeta3 crystal structure using a molecular modeling approach. These results support the observations that the cyclic peptides had little effects, and also reinforce the observation that residues outside the disintegrin RGD sequence are required for interactions with receptor.


Subject(s)
Crotalid Venoms/toxicity , Integrins/antagonists & inhibitors , Oligopeptides/chemistry , Oligopeptides/pharmacology , Platelet Aggregation/drug effects , Amino Acid Sequence , Animals , Bothrops , Chromatography, High Pressure Liquid , Crotalid Venoms/chemistry , Disintegrins/chemistry , Disintegrins/isolation & purification , Disintegrins/pharmacology , Humans , Oligopeptides/isolation & purification , Peptides/chemical synthesis , Peptides/chemistry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Thrombin/biosynthesis , Thrombin/drug effects
6.
J Thromb Thrombolysis ; 24(3): 275-82, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17486300

ABSTRACT

The Porthidium genus is represented by the P. lansbergii rozei and P. lansbergii hutmanni (Plh) subspecies in Venezuela. The venom components of these have been little studied, probably due to the low incidence of reported accidents, although acute and serious local effects such as invasive edema and disseminated ecchymosis are present during human envenonation. The aim of this work was to characterize the in vitro effects of crude P. l. hutmanni venom, and its fractions, on platelet aggregation triggered by two physiologic agonists: thrombin and collagen. The effects of thrombin and collagen were observed on a platelet-rich plasma (PRP) solution (3 x 10(5) platelets/microL) using serial dilutions of P. l. hutmanni venom (0.625-40 microg). The crude venom was fractionated by anionic exchange chromatography and two peaks obtained. Crude venom and both fractions were highly inhibitory on platelet aggregation mediated by the two agonists. The anti-aggregating dose (AD(50)) for both agonists was determined. PRP collagen-triggered aggregation was most inhibited by the crude venom (AD(50) = 0.67 microg) when compared with PRP thrombin-triggered aggregation (AD(50) = 4.92 microg). Collagen-induced aggregation was more intensely inhibited by venom than thrombin-induced aggregation. In conclusion, to specify the inhibition mechanisms involved for each of the active components in the venom from these subspecies, we must characterize and purify the inhibitors of aggregation from P. l. hutmanni venom, with the purpose of suggesting new pharmacological substances to be incorporated into the therapeutic arsenal to treat hemostatic pathologies related to high levels of platelet aggregation.


Subject(s)
Crotalid Venoms/pharmacology , Fibrinolytic Agents/pharmacology , Platelet Aggregation/drug effects , Chromatography, Ion Exchange , Collagen/drug effects , Crotalid Venoms/isolation & purification , Fibrinolytic Agents/isolation & purification , Humans , Snake Bites/blood , Thrombin/drug effects , Venezuela
7.
Biochim Biophys Acta ; 1770(5): 810-9, 2007 May.
Article in English | MEDLINE | ID: mdl-17306461

ABSTRACT

Venoms of Colubridae snakes are a rich source of novel compounds, which may have applications in medicine and biochemistry. In the present study, we describe the purification and characterization of a metalloproteinase (patagonfibrase), the first protein to be isolated from Philodryas patagoniensis (Colubridae) snake venom. Patagonfibrase is a single-chain protein, showing a molecular mass of 53,224 Da and an acidic isoelectric point (5.8). It hydrolyzed selectively the Aalpha-chain of fibrinogen and when incubated with fibrinogen or plasma, the thrombin clotting time was prolonged. Prominent hemorrhage developed in mouse skin after intradermal injection of patagonfibrase. When administered into mouse gastrocnemius muscle, it induced local hemorrhage and necrosis, and systemic bleeding in lungs. Patagonfibrase showed proteolytic activity toward azocasein, which was enhanced by Ca(2+) and inhibited by Zn(2+), cysteine, dithiothreitol and Na(2)EDTA. Patagonfibrase impaired platelet aggregation induced by collagen and ADP. Thus, patagonfibrase may play a key role in the pathogenesis of disturbances that occur in P. patagoniensis envenomation, and may be used as a biological tool to explore many facets of hemostasis.


Subject(s)
Blood Coagulation/drug effects , Colubridae , Fibrinogen/drug effects , Metalloproteases/isolation & purification , Metalloproteases/toxicity , Snake Venoms/toxicity , Animals , Blood Platelets/drug effects , Edema/chemically induced , Edema/pathology , Electrophoresis, Polyacrylamide Gel , Fibrinogen/metabolism , Hemorrhage/chemically induced , Humans , Hydrogen-Ion Concentration , Inflammation/chemically induced , Inflammation/pathology , Isoelectric Focusing , Mass Spectrometry , Mice , Muscle, Skeletal/drug effects , Necrosis/chemically induced , Necrosis/pathology , Platelet Aggregation/drug effects , Snake Venoms/enzymology , Temperature , Thrombin/drug effects , Thrombin/metabolism , Thrombin Time
8.
J Thromb Haemost ; 4(9): 2022-7, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16961610

ABSTRACT

BACKGROUND: Antiplatelet drugs constitute the therapy of choice for acute coronary syndromes, but bleeding can be a side-effect requiring treatment. Restoration of normal platelet activity is also mandatory before urgent surgery. This study investigated: (a) whether a regimen of aspirin or clopidogrel plus aspirin significantly inhibited platelet thrombin generation (TG); and (b) the reversal of this inhibition by recombinant activated factor VII (rFVIIa). METHODS AND RESULTS: TG was evaluated by the lag time, time to peak, peak of TG, and area under the curve after 35 min of assay (AUC(0 --> 35 min)). These measures were examined by the calibrated automated thrombography method in 22 healthy volunteers, 22 volunteers after a 100 mg day(-1) aspirin intake (200 mg first day) for 5-7 days, and 22 healthy volunteers after aspirin 100 mg day(-1) (200 mg first day) plus clopidogrel 75 mg day(-1) (300 mg first day) for 4-7 days. The TG parameters were measured under basal conditions and after platelet stimulation by sodium arachidonate (AA), adenosine 5'-diphosphate (ADP), collagen and rFVIIa in normal non-aspirinated as well as in vivo aspirinated platelet-rich plasma (PRP) or aspirin plus clopidogrel PRP. Lag time was shorter (P < 0.05), and peak of TG and AUC(0 --> 35 min) were significantly greater (P < 0.01 for both), in PRP activated with ADP, collagen, AA or FVIIa than in non-activated PRP from normal subjects. Both non-activated PRP and activated PRP prepared from platelets obtained from volunteers after aspirin intake showed significant prolongation of the time parameters but there was less effect on peak of TG and AUC(0 --> 35 min). For most parameters, aspirin plus clopidogrel administration showed to be more effective compared with the effect obtained by aspirin alone. When rFVIIa was added to ASA-PRP or ASA + Clop PRP, lag time (P < 0.001 for all) and time to peak (P < 0.001-0.017) were significantly shortened, indicating that rFVIIa reverses the inhibitory effect of these anti-aggregating agents. CONCLUSION: Platelets activated by AA, ADP, collagen or FVIIa triggered TG. This effect was inhibited by aspirin plus clopidogrel, suggesting an additional benefit of this drug combination for preventing thrombosis. rFVIIa reverses the inhibitory effect of aspirin or aspirin plus clopidogrel, and could be useful for bleeding complications or when acute surgery is needed during treatment with these antiplatelet drugs.


Subject(s)
Aspirin/administration & dosage , Factor VIIa/administration & dosage , Thrombin/drug effects , Ticlopidine/analogs & derivatives , Adult , Aged , Blood Coagulation Tests , Clopidogrel , Drug Interactions , Drug Therapy, Combination , Female , Humans , Male , Middle Aged , Platelet Activation/drug effects , Platelet Function Tests , Recombinant Proteins , Thrombin/biosynthesis , Ticlopidine/administration & dosage
9.
An. acad. bras. ciênc ; 77(2): 275-280, June 2005. ilus, tab
Article in English | LILACS | ID: lil-399101

ABSTRACT

A alta especificidade das proteases da coagulação tem sido atribuída não somente aos resíduos que cercam o sítio ativo, mas também a outros domínios de superfície que estão envolvidos no reconhecimento e interação com substratos macromoleculares e inibidores. Inibidores específicos da coagulação sanguínea obtidos de fontes exógenas como glândulas salivares de animais hematófagos e venenos de serpentes têm sido identificados. Alguns desses inibidores interagem com os exosítios das enzimas da coagulação. Dois exemplos são discutidos nesta curta revisão. A Botrojaracina é uma proteína derivada de veneno de serpente que se liga aos exosítios 1 e 2 da trombina. A formação do complexo impede várias atividades da trombina dependentes do exosítio 1 incluindo a clivagem do fibrinogênio e a ativação plaquetária. A Botrojaracina também interage com o proexosítio 1 da protrombina diminuindo a ativação do zimogênio pelo complexo protrombinase (FXa/FVa). O ixolaris é um inibidor com dois domínios Kunitz obtido da glândula salivar de carrapato, homólogo ao inibidor da via do fator tecidual. Recentemente foi demonstrado que o ixolaris se liga ao exosítio de ligação à heparina do FXa, impedindo o reconhecimento da protrombina pela enzima. Além disso, o ixolaris interage com o FX, possivelmente através do proexosítio de ligação à heparina. Diferentemente do FX, o complexo ixolaris-FX não é reconhecido como substrato pelo complexo tenase intrínseco (FIXa/FVIIIa). Nós concluimos que esses inibidores podem servir como ferramentas para o estudo dos exosítios da coagulação, assim como protótipos para novas drogas anticoagulantes.


Subject(s)
Animals , Anticoagulants/pharmacology , Blood Coagulation/drug effects , Crotalid Venoms/enzymology , Crotalid Venoms/pharmacology , Salivary Proteins and Peptides/pharmacology , Thrombin/drug effects , Anticoagulants/isolation & purification , Bothrops , Crotalid Venoms/chemistry , Crotalid Venoms/isolation & purification , Enzyme Inhibitors/pharmacology , Factor X/drug effects , Factor Xa/drug effects , Salivary Proteins and Peptides/isolation & purification
10.
An Acad Bras Cienc ; 77(2): 275-80, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15895163

ABSTRACT

The high specificity of blood coagulation proteases has been attributed not only to residues surrounding the active site but also to other surface domains that are involved in recognizing and interacting with macromolecular substrates and inhibitors. Specific blood coagulation inhibitors obtained from exogenous sources such as blood sucking salivary glands and snake venoms have been identified. Some of these inhibitors interact with exosites on coagulation enzymes. Two examples are discussed in this short revision. Bothrojaracin is a snake venom-derived protein that binds to thrombin exosites 1 and 2. Complex formation impairs several exosite-dependent activities of thrombin including fibrinogen cleavage and platelet activation. Bothrojaracin also interacts with proexosite 1 on prothrombin thus decreasing the zymogen activation by the prothrombinase complex (FXa/FVa). Ixolaris is a two Kunitz tick salivary gland inhibitor, that is homologous to tissue factor pathway inhibitor. Recently it was demonstrated that ixolaris binds to heparin-binding exosite of FXa, thus impairing the recognition of prothrombin by the enzyme. In addition, ixolaris interacts with FX possibly through the heparin-binding proexosite. Differently from FX, the ixolaris-FX complex is not recognized as substrate by the intrinsic tenase complex (FIXa/FVIIIa). We conclude that these inhibitors may serve as tools for the study of coagulation exosites as well as prototypes for new anticoagulant drugs.


Subject(s)
Anticoagulants/pharmacology , Blood Coagulation/drug effects , Crotalid Venoms/enzymology , Crotalid Venoms/pharmacology , Salivary Proteins and Peptides/pharmacology , Thrombin/drug effects , Animals , Anticoagulants/isolation & purification , Bothrops , Crotalid Venoms/chemistry , Crotalid Venoms/isolation & purification , Enzyme Inhibitors/pharmacology , Factor X/drug effects , Factor Xa/drug effects , Salivary Proteins and Peptides/isolation & purification , Serine Endopeptidases/drug effects
11.
Toxicon ; 45(4): 527-34, 2005 Mar 15.
Article in English | MEDLINE | ID: mdl-15733575

ABSTRACT

Duvernoy's gland secretion of Philodryas patagoniensis exhibits high hemorrhagic activity, containing enzymes that are able to degrade the vascular wall. In this work we aim to determine if the secretion can also affect the hemostatic system by causing changes in blood coagulation. Procoagulant and coagulant activities were evaluated on plasma and fibrinogen, respectively. The delay in the thrombin clotting time of fibrinogen previously incubated with the secretion was also determined. Specific hydrolysis of fibrinogen and fibrin incubated with the secretion at different time intervals was shown by electrophoresis on polyacrylamide gel. To determine the structural characteristics of the enzymes degrading fibrinogen and fibrin, secretion were incubated in the presence of 45 mM Na(2)EDTA, 40 mM Benzamidine, and/or 2 mM PMSF before the incubation with fibrinogen or fibrin, respectively. The effect in vivo was investigated in adult male rats injected with different dose of secretion, aliquots of blood were withdrawn at different time intervals, and the fibrinogen concentration was determined. Duvernoy's gland secretion of P. patagoniensis did not clot plasma or fibrinogen. It exhibited a potent fibrinogenolytic activity degrading the Aalpha-chain faster than the Bbeta-chain, whereas gamma-chain was resistant. This latter corresponded with a strong delay in the thrombin clotting time of fibrinogen (4 mg/ml) pre-incubated with the secretion, being 9.53 microg the amount of protein from Duvernoy's gland secretion that increased the thrombin clotting time from 20 to 60 s. In vivo, the loss of rat plasma fibrinogen was proportional to the amount of secretion injected. The secretion also hydrolyzed fibrin degrading the alpha-monomer. Inhibition studies with Na(2)EDTA, Benzamidine, and/or PMSF showed that metalloproteinases and serinoproteinases are the main enzymes responsible for the hydrolyzing activity on fibrinogen and fibrin. All these results demonstrate that Duvernoy's gland secretion of P. patagoniensis possesses enzymes able to hydrolyze plasma components playing a relevant role in the blood coagulation. These hydrolyzing activities and those acting on the wall of blood vessels let the secretion exhibit a high hemorrhagic activity, which may result in permanent sequelae or even cause the death of the victims bitten by this colubrid snake.


Subject(s)
Blood Coagulation/drug effects , Colubridae , Exocrine Glands/metabolism , Snake Venoms/toxicity , Analysis of Variance , Animals , Argentina , Electrophoresis, Polyacrylamide Gel , Fibrinogen/drug effects , Fibrinogen/metabolism , Male , Rats , Rats, Sprague-Dawley , Snake Venoms/enzymology , Snake Venoms/metabolism , Thrombin/drug effects , Thrombin/metabolism , Time Factors
12.
J Biochem Mol Toxicol ; 16(4): 189-96, 2002.
Article in English | MEDLINE | ID: mdl-12242688

ABSTRACT

Heptachlor is a persistent organochlorine insecticide that has been detected in human tissues and fluids. The ability of heptachlor to interfere with platelet phosphoinositides metabolism and related signaling events stimulated by thrombin was evaluated. In vitro incubations with a concentration range of 1-100 microM heptachlor, prior to platelets activation, were performed. Experiments showed that 10 microM increased protein Kinase C (PKC) activity and phosphatidylinositolbiphosphate and phosphatidic acid phosphorylation. Simultaneously phosphatidylcholine and phosphatidylethanolamine breakdown were prevented. Similar effects were observed with HC 1 microM. However, heptachlor 100 microM increased phosphatidylinositolbiphosphate phosphorylation but reduced serine/threonine kinases activity. We propose that signal transduction steps downstream phospholipase C (PLC) are unphysiologically activated by heptachlor and facilitated by the increase in phosphatidylinositolbiphosphate, the substrate for PLC activity, thus producing an accumulation of phosphatidic acid. The elevated level of this compound itself or the transient increase in diacylglycerol produced may cause calcium mobilization and the activation of PKC. In contrast with the alterations observed in phospholipids and protein phosphorylation, no changes in aggregation properties were observed.


Subject(s)
Blood Platelets/drug effects , Heptachlor/pharmacology , Insecticides/pharmacology , Signal Transduction/drug effects , Thrombin/drug effects , Blood Platelets/enzymology , Blood Platelets/metabolism , Blood Proteins/metabolism , Humans , Platelet Aggregation/drug effects , Protein Kinase C/metabolism , Thrombin/metabolism
13.
Thromb Res ; 105(2): 183-8, 2002 Jan 15.
Article in English | MEDLINE | ID: mdl-11958811

ABSTRACT

Quercetin 3-acetyl-7,3',4'-trisulphate (ATS) and quercetin 3,7,3',4'-tetrasulphate (QTS) obtained from Flaveria bidentis (Asteraceae) were investigated in vitro for anticoagulant activity. Three different concentrations of each flavonoid were assayed at different incubation times, showing at 1 mM significant prolongation on the activated partial thromboplastin time (APTT), less on the prothrombin time (PT), and no effect on the thrombin time (TT). In order to define the action mechanism of the anticoagulant activity, all coagulation factors were evaluated and no important activity decrease was observed, indicating that another mechanism is involved. Thus, thrombin inhibition mediated by antithrombin III (ATIII) and heparin cofactor II (HCII) activation was investigated in comparison to the physiological activators, heparin and dermatan sulphate (DS), respectively. As a conclusion, no activation on ATIII for neither flavonoids was observed. On the contrary, QTS much more than ATS produced an activation on HCII comparable to the one of DS, indicating that these flavonoids act as agonists of this inhibitor. A plausible explanation of the effects of both flavonoids could be due to the different degree of sulphation of these molecules. According to the results obtained, and taking in account the high solubility of these natural products in aqueous media and the nontoxic nature of this family of compounds, further investigation on the antithrombotic effects of these flavonoids are merited.


Subject(s)
Anticoagulants/pharmacology , Asteraceae/chemistry , Flavonoids/pharmacology , Quercetin/analogs & derivatives , Anticoagulants/chemistry , Blood Coagulation Tests , Factor Xa/drug effects , Flavonoids/chemistry , Humans , Molecular Structure , Plants, Medicinal/chemistry , Quercetin/chemistry , Quercetin/pharmacology , Static Electricity , Sulfates , Thrombin/drug effects
15.
FEBS Lett ; 458(1): 11-6, 1999 Sep 10.
Article in English | MEDLINE | ID: mdl-10518924

ABSTRACT

The recombinant phage antibody system pCANTAB 5E has been used to display functionally active leech-derived tryptase inhibitor (LDTI) on the tip of the filamentous M13 phage. A limited combinatorial library of 5.2 x 10(4) mutants was created with a synthetic LDTI gene, using a degenerated oligonucleotide and the pCANTAB 5E phagemid. The mutations were restricted to the P1-P4' positions of the reactive site. Fusion phages and appropriate host strains containing the phagemids were selected after binding to thrombin and DNA sequencing. The variants LDTI-2T (K8R, I9V, S10, K11W, P12A), LDTI-5T (K8R, I9V, S10, K11S, P12L) and LDTI-10T (K8R, I9L, S10, K11D, P12I) were produced with a Saccharomyces cerevisiae expression system. The new inhibitors, LDTI-2T and -5T, prolong the blood clotting time, inhibit thrombin (Ki 302 nM and 28 nM) and trypsin (Ki 6.4 nM and 2.1 nM) but not factor Xa, plasma kallikrein or neutrophil elastase. The variant LDTI-10T binds to thrombin but does not inhibit it. The relevant reactive site sequences of the thrombin inhibiting variants showed a strong preference for arginine in position P1 (K8R) and for valine in P1' (I9V). The data indicate further that LDTI-5T might be a model candidate for generation of active-site directed thrombin inhibitors and that LDTI in general may be useful to generate specific inhibitors suitable for a better understanding of enzyme-inhibitor interactions.


Subject(s)
Bacteriophages/chemistry , Peptide Library , Proteins/chemistry , Proteins/pharmacokinetics , Serine Endopeptidases/chemistry , Thrombin/antagonists & inhibitors , Amino Acid Sequence , Base Sequence , Chymases , Cloning, Molecular , Dose-Response Relationship, Drug , Humans , Kinetics , Models, Genetic , Molecular Sequence Data , Mutagenesis, Insertional , Saccharomyces cerevisiae/chemistry , Thrombin/drug effects , Tryptases
SELECTION OF CITATIONS
SEARCH DETAIL