Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 94
Filtrar
1.
Int J Mol Sci ; 22(2)2021 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-33477998

RESUMEN

Stachybotrys microspora triprenyl phenol (SMTP) is a large family of small molecules derived from the fungus S. microspora. SMTP acts as a zymogen modulator (specifically, plasminogen modulator) that alters plasminogen conformation to enhance its binding to fibrin and subsequent fibrinolysis. Certain SMTP congeners exert anti-inflammatory effects by targeting soluble epoxide hydrolase. SMTP congeners with both plasminogen modulation activity and anti-inflammatory activity ameliorate various aspects of ischemic stroke in rodents and primates. A remarkable feature of SMTP efficacy is the suppression of hemorrhagic transformation, which is exacerbated by conventional thrombolytic treatments. No drug with such properties has been developed yet, and SMTP would be the first to promote thrombolysis but suppress disease-associated bleeding. On the basis of these findings, one SMTP congener is under clinical study and development. This review summarizes the discovery, mechanism of action, pharmacological activities, and development of SMTP.


Asunto(s)
Benzopiranos/farmacología , Inflamación/tratamiento farmacológico , Accidente Cerebrovascular Isquémico/tratamiento farmacológico , Pirrolidinonas/farmacología , Terapia Trombolítica/métodos , Animales , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Benzopiranos/uso terapéutico , Isquemia Encefálica/sangre , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/patología , Epóxido Hidrolasas/efectos de los fármacos , Epóxido Hidrolasas/metabolismo , Fibrinólisis/efectos de los fármacos , Fibrinolíticos/farmacología , Fibrinolíticos/uso terapéutico , Humanos , Inflamación/sangre , Inflamación/patología , Accidente Cerebrovascular Isquémico/sangre , Accidente Cerebrovascular Isquémico/patología , Plasminógeno/efectos de los fármacos , Plasminógeno/metabolismo , Pirrolidinonas/uso terapéutico , Stachybotrys/química , Stachybotrys/metabolismo
2.
Acta Pharmacol Sin ; 32(2): 239-44, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21293476

RESUMEN

AIM: To evaluate the effects of the fibrinolytic enzyme FII(a) from Agkistrodon acutus venom on acute pulmonary thromboembolism (APT) in animal models. METHODS: Both rabbit and dog APT models were used. For the rabbit APT model, the thrombi weight before and after administration was measured. Central venous pressure (CVP) and mean arterial pressure (MAP) were measured before and 15, 30, 60, and 120 min after the injection of the blood clot. Partial thromboplastin time (APTT), prothrombin time (PT), platelet count, and fibrinogen concentration were measured using auto analyzers. Plasminogen activity was measured based on chromogenic substrates. In the dog APT model, pulmonary blood flow was recorded using pulmonary angiography. RESULTS: Intravenous administration of FIIa (0.1-5.0 mg/kg) improved the APT-induced hemodynamic derangements and reduced thrombi weight. The angiography evidence also showed that the pulmonary emboli had almost disappeared after FII(a) infusion. FII(a) (0.1, 0.5, or 1.0 mg/kg) did not impair the coagulation pathways, although very high doses of FII(a) (5.0 mg/kg) could stimulate the production of plasminogen and result in impairment of the pathways. CONCLUSION: FII(a) could effectively protect against APT via degradation of thrombi with less activation of plasminogen, and may provide a novel fibrinolytic enzyme for targeting the main pathological processes of the disease.


Asunto(s)
Agkistrodon , Venenos de Crotálidos/farmacología , Fibrinolíticos/farmacología , Embolia Pulmonar/tratamiento farmacológico , Enfermedad Aguda , Angiografía/métodos , Animales , Venenos de Crotálidos/aislamiento & purificación , Modelos Animales de Enfermedad , Perros , Relación Dosis-Respuesta a Droga , Fibrinolíticos/aislamiento & purificación , Masculino , Metaloendopeptidasas , Plasminógeno/efectos de los fármacos , Plasminógeno/metabolismo , Embolia Pulmonar/fisiopatología , Conejos , Factores de Tiempo
3.
Acta Pol Pharm ; 66(1): 37-40, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19226966

RESUMEN

The effect of epsilon-aminocaproyl-S-benzyl-L-cysteine on the activation of plasminogen by t-PA. streptokinase and urokinase has been examined using fibrinolytic method. The obtained results have been compared with the obtained results for epsilon-aminocaproic acid and trans-4-(aminomethyl)cyclohexanecarboxylic acid. The inhibition of the plasminogen activation determined with the use of epsilon-aminocaproyl-S-benzyl-L-cysteine was weaker than the inhibition determined by using antifibrinolytic aminoacids.


Asunto(s)
Aminocaproatos , Antifibrinolíticos/farmacología , Cisteína/análogos & derivados , Fibrinolíticos/farmacología , Plasminógeno/efectos de los fármacos , Ácido Aminocaproico/farmacología , Cisteína/farmacología , Plasminógeno/metabolismo , Estreptoquinasa/farmacología , Activador de Tejido Plasminógeno/farmacología , Ácido Tranexámico/farmacología , Activador de Plasminógeno de Tipo Uroquinasa/farmacología
4.
Drug Des Devel Ther ; 13: 881-896, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30880920

RESUMEN

BACKGROUND: Sucrose allyl ether (SAE) containing hemostatic drugs and a photoinitiator was established to treat mild postpartum hemorrhage or long-term continuous abnormal uterine bleeding in minimally invasive surgery (MIS) using a photopolymerization method. METHODS AND RESULTS: Real-time infrared spectroscopy and rheological experiments showed that the SAE monomer with shear-thinning characteristics could polymerize rapidly into a transparent membrane. Cytotoxicity experiments in vitro showed that this system could elicit a long-term hemostatic effect. Tissue adhesion was also evaluated. The photo-stability of four delivered antifibrinolytic drugs (6-aminocaproic acid, ethylenediaminediacetic acid, tranexamic acid and p-(aminomethyl) benzoic acid) was tested by ultraviolet-photolysis experiments and illustrated by time-dependent density functional theory. Sustained-release experiments revealed that the formed film could be used as a drug carrier. Molecular docking and molecular dynamics were done to investigate the binding mechanism between hemostatic drugs as ligands and the human plasminogen kringle-1 (1HPK) as a target. CONCLUSION: It has been suggested that SAE with tranexamic acid could be a drug-release system of microchannel transport used in MIS. This system could tackle the dilemma of fluidity and adhesion in MIS. The photo-stable tranexamic acid was the most suitable drug according to its satisfactory binding energy, good photo-stability, and sustained release.


Asunto(s)
Antifibrinolíticos/farmacología , Liberación de Fármacos , Hemostasis , Kringles/efectos de los fármacos , Procedimientos Quirúrgicos Mínimamente Invasivos , Plasminógeno/efectos de los fármacos , Teoría Funcional de la Densidad , Portadores de Fármacos/química , Humanos , Ligandos , Modelos Moleculares , Factores de Tiempo
5.
J Trauma Acute Care Surg ; 86(1): 101-107, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30575685

RESUMEN

BACKGROUND: Both tissue plasminogen activator (tPA) in the circulation and urokinase (uPA) in tissues cleave plasminogen (PLG) to plasmin to promote clot lysis. Tranexamic acid (TXA) blocks both the tPA-dependent generation of plasmin on blood clots as well as active plasmin binding to polymerized fibrin, and is commonly administered for bleeding in trauma to limit fibrinolysis. In addition to lysing clots, however, active plasmin also cleaves complement proteins, potentially enhancing inflammation. Because TXA does not block uPA-dependent plasmin generation from PLG and instead augments it, we hypothesized that administration of TXA could enhance or inhibit proinflammatory C5a formation in a PLG activator-dependent manner. METHODS: Citrate platelet-poor plasma (PPP) and PPP depleted of complement protein C3 or PLG were obtained from healthy donors and commercial sources. Platelet-poor plasma was treated ex vivo with or without TXA and either with or without tPA or with or without uPA. Clotting was then induced by calcium and thrombin in clotted PPP experiments, while unclotted PPP experiments were treated with vehicle controls. C5a levels were measured via enzyme-linked immunosorbent assay. Data were expressed as mean ± SEM. RESULTS: Plasmin-mediated fibrinolysis by tPA in clotted PPP led to an approximately threefold increase in C5a production (p < 0.0001), which was significantly inhibited by TXA (p < 0.001). Paradoxically, when fibrinolysis was induced by uPA, TXA treatment led to further increases in C5a production beyond uPA alone (p < 0.0001). Furthermore, clotting was not required for C5a generation from uPA + TXA. C3 depletion had no effect on C5a production, while depletion of PLG eliminated it. CONCLUSIONS: Tranexamic acid administration can have proinflammatory or anti-inflammatory effects through regulating C5a generation by plasmin, depending on the predominating PLG activator. Tranexamic acid may cause significant inflammatory C5a elevations in injured tissues by augmenting uPA-mediated plasmin generation in a fibrin-independent manner. In contrast, TXA reduces C5a generation during tPA-mediated fibrinolysis that may reduce inflammatory responses. In vivo validation of these novel ex vivo findings is warranted and may have important clinical consequences.


Asunto(s)
Antiinflamatorios/metabolismo , Antifibrinolíticos/farmacología , Complemento C5a/metabolismo , Mediadores de Inflamación/metabolismo , Ácido Tranexámico/farmacología , Adulto , Antifibrinolíticos/administración & dosificación , Coagulación Sanguínea/efectos de los fármacos , Coagulación Sanguínea/fisiología , Complemento C5a/efectos de los fármacos , Femenino , Fibrinolisina/metabolismo , Fibrinólisis/efectos de los fármacos , Fibrinólisis/fisiología , Hemorragia/tratamiento farmacológico , Hemorragia/etiología , Humanos , Masculino , Persona de Mediana Edad , Plasminógeno/efectos de los fármacos , Plasminógeno/metabolismo , Transducción de Señal/efectos de los fármacos , Trombina/metabolismo , Activador de Tejido Plasminógeno/metabolismo , Ácido Tranexámico/administración & dosificación , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Heridas y Lesiones/complicaciones
6.
Toxicon ; 52(5): 628-37, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18760294

RESUMEN

Two activators of coagulation factor X, 58kDa VAFXA-I and 70kDa VAFXA-II, were purified from the venom of long-nosed viper (Vipera ammodytes ammodytes) by chromatography on gel filtration, affinity, ion-exchange and hydroxyapatite media. Both enzymes are glycoproteins composed of a heavy chain and two C-type lectin-like light chains all joined by disulphide bonds. LC-MS and LC-MS/MS analysis of their tryptic fragments demonstrated that the heavy chain consists of three domains, metalloproteinase, disintegrin-like and cysteine-rich domains. The partial amino acid sequences of VAFXAs are very similar to those of the known factor X activators, RVV-X from Vipera russelli and VLFXA from Vipera lebetina venoms, as well as to other members of the reprolysin family of metalloproteinases. The VAFXAs activate factor X in a Ca(2+)-dependent manner with the same specificity as physiological activators. The activators weakly hydrolyzed insulin B-chain, fibrinogen and some components of the extracellular matrix in vitro, but did not activate prothrombin or plasminogen. VAFXAs inhibit collagen-induced platelet aggregation in vitro. They activate coagulation factor X to Xa without toxic effects. Their application in treating patients with dysfunctional factors IXa or VIIa to restore the normal blood coagulation process is thus promising.


Asunto(s)
Factor X/metabolismo , Hemostáticos/química , Metaloendopeptidasas/química , Proteínas de Reptiles/química , Venenos de Víboras/enzimología , Secuencia de Aminoácidos , Animales , Membrana Basal/efectos de los fármacos , Coagulación Sanguínea/efectos de los fármacos , Colágeno , Combinación de Medicamentos , Factor IXa/metabolismo , Factor VIIa/metabolismo , Fibrinógeno/efectos de los fármacos , Fibrinógeno/metabolismo , Hemostáticos/aislamiento & purificación , Hemostáticos/farmacología , Humanos , Hidrólisis , Laminina , Metaloendopeptidasas/aislamiento & purificación , Metaloendopeptidasas/farmacología , Datos de Secuencia Molecular , Plasminógeno/efectos de los fármacos , Plasminógeno/metabolismo , Agregación Plaquetaria/efectos de los fármacos , Proteoglicanos , Protrombina/efectos de los fármacos , Protrombina/metabolismo , Proteínas de Reptiles/aislamiento & purificación , Proteínas de Reptiles/farmacología , Alineación de Secuencia , Especificidad por Sustrato
7.
Life Sci ; 82(7-8): 413-8, 2008 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-18206177

RESUMEN

Conditions in which serum or tissue acrolein levels are high (e.g.: renal failure, heavy smoking, oxidative stress) are also associated with increased thrombogenicity. Another emerging cardiovascular risk factor is homocysteine, and its derivative, homocysteine thiolactone. Antithrombin is one of the most important inhibitors of blood coagulation Since its activation by heparin binding requires critical interactions involving 3 Lys residues; we hypothesized that acrolein or homocysteine thiolactone impair antithrombin activity. When we incubated human antithrombin with increasing concentrations of acrolein (0-2 mmol/L) over a short period of time (0-4 h), a time and concentration dependent loss of activity was apparent (IC(50)=0.25 mmol/L). At 2 mmol/L, maximum inhibition (60%) is achieved at 1 h. This loss of activity was mirrored by changes in the electrophoretic pattern (homogeneity of the native antithrombin band as well as polymerization). In the same conditions, homocysteine thiolactone produces a significant, yet far less pronounced effect; acrolein being 3 times more potent than homocysteine thiolactone. When antithrombin was co-incubated with acrolein and cysteine, only less than 10% of antithrombin activity was lost. Aminoguanidine or carnosine displayed a significant yet, minor protective effect. The results suggest that in conditions where circulating or local acrolein concentrations are increased (atheroma plaque, thrombosis, sites of lipoperoxidation, smokers), acrolein-mediated loss of antithrombin activity could be a plausible phenomenon. This could contribute to explain increased thrombogenicity in smokers and in other conditions, as well as pointing at dietary intervention or the use of thiol-conserving reducing compounds as putative coadjuvant therapeutic measures.


Asunto(s)
Acroleína/farmacología , Antitrombinas/efectos de los fármacos , Cisteína/farmacología , Homocisteína/análogos & derivados , Relación Dosis-Respuesta a Droga , Electroforesis en Gel de Poliacrilamida , Homocisteína/farmacología , Humanos , Plasminógeno/efectos de los fármacos , Factores de Tiempo
8.
Blood Coagul Fibrinolysis ; 19(1): 60-5, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18180617

RESUMEN

Fucoidan and chondroitin-6-sulfate were oversulfated using chlorosulfonic acid-pyridine complex and were isolated as the sodium salt. Infrared analysis of oversulfated compounds showed introduction of sulfate groups in new positions, and in-vitro studies of the compounds showed a significant increase in the anticoagulant property. Addition of 28.6 microg/ml oversulfated compound gave a two-fold to four-fold increase in the rate of enhancement of activation of glutamic plasminogen by tissue plasminogen activator using 0.05 mol/l Tris buffer (pH 7.35) containing physiological concentrations of NaCl (0.9%). Under these conditions, unfractionated heparin was not active and the native compounds gave less than 30% enhancement. In the present study, the effect of lysine or cyanogen bromide-treated fibrinogen, alone or in combination with the oversulfated compounds, on the activation of glutamic plasminogen by tissue plasminogen activator was investigated. Addition of 16.2 mmol/l L-lysine gave three-fold to four-fold enhancement of activation, which was further enhanced to five-fold to six-fold by addition of 2.86 microg/ml oversulfated chondroitin-6-sulfate or oversulfated fucoidan. Cyanogen bromide-treated fibrinogen (50 microg/ml) gave a 10-fold enhancement of activation by itself, and addition of 2.86 microg/ml oversulfated compounds amplified this to 15-fold. A 25-fold to 35-fold enhancement of activation of glutamic plasminogen was obtained when 2.86 microg/ml oversulfated compounds were combined with 16.2 mmol/l lysine and 50 microg/ml cyanogen bromide-treated fibrinogen. Dilution studies showed that the amplification of the enhancement of lysine by 2.86 microg/ml oversulfated compound was related to interaction of the cofactors with both glutamic plasminogen and tissue plasminogen activator.


Asunto(s)
Sulfatos de Condroitina/farmacología , Lisina/farmacología , Plasminógeno/efectos de los fármacos , Polisacáridos/farmacología , Sulfatos de Condroitina/química , Bromuro de Cianógeno/química , Bromuro de Cianógeno/farmacología , Interacciones Farmacológicas , Fibrinógeno/química , Fibrinógeno/fisiología , Humanos , Plasminógeno/fisiología , Polisacáridos/química , Unión Proteica , Relación Estructura-Actividad , Sulfatos/química , Activador de Tejido Plasminógeno/efectos de los fármacos , Activador de Tejido Plasminógeno/metabolismo
9.
Blood Coagul Fibrinolysis ; 19(6): 483-7, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18685430

RESUMEN

Chondroitin-4-sulfate was oversulfated using chlorosulfonic acid-pyridine complex and was isolated as the sodium salt. A comparison of the infrared analysis of the native (N-2) and oversulfated (S-2) compounds showed that the two spectra were identical except for a new peak in S-2 at 825 cm corresponding to the equatorial C-6 position of galactosamine. There was a 2.7-fold increase of sulfate content in S-2 and a generation of a significant anticoagulant activity as measured by doubling of the prothrombin time of normal citrated human plasma using 7.5 microg, while N-2 was inactive even at 2,000 microg. The result of the in-vitro studies of the activation of glutamic plasminogen by tissue plasminogen activator (t-PA) or by high-molecular-weight urokinase using 0.05 mol/l Tris buffer (pH 7.35) containing a physiological concentration of NaCl (0.9%) showed that 28.6 microg/ml S-2 enhanced the activation by three-fold to four-fold by t-PA or by urokinase, while the same concentrations of N-2 or unfractionated heparin gave less than 30% enhancement of t-PA and no enhancement of urokinase. The mechanism of enhancement by S-2 was investigated by dilution studies. The results showed that S-2 interacted with both urokinase or t-PA and glutamic plasminogen favoring a template model, while N-2 or unfractionated heparin interacted only with t-PA.


Asunto(s)
Sulfatos de Condroitina/química , Sulfatos/análisis , Coagulación Sanguínea/efectos de los fármacos , Interacciones Farmacológicas , Activación Enzimática/efectos de los fármacos , Fibrinolisina/biosíntesis , Heparina/farmacología , Humanos , Plasma , Plasminógeno/efectos de los fármacos , Plasminógeno/metabolismo , Tiempo de Protrombina , Piridinas/farmacología , Espectrofotometría Infrarroja , Ácidos Sulfónicos/farmacología , Tromboplastina/farmacología , Activador de Tejido Plasminógeno/farmacología , Activador de Plasminógeno de Tipo Uroquinasa/farmacología
10.
PLoS Negl Trop Dis ; 12(4): e0006446, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29677188

RESUMEN

BACKGROUND: Serine proteases are important virulence factors for many pathogens. Recently, we discovered a group of trypsin-like serine proteases with domain organization unique to flatworm parasites and containing a thrombospondin type 1 repeat (TSR-1). These proteases are recognized as antigens during host infection and may prove useful as anthelminthic vaccines, however their molecular characteristics are under-studied. Here, we characterize the structural and proteolytic attributes of serine protease 2 (SmSP2) from Schistosoma mansoni, one of the major species responsible for the tropical infectious disease, schistosomiasis. METHODOLOGY/PRINCIPAL FINDINGS: SmSP2 comprises three domains: a histidine stretch, TSR-1 and a serine protease domain. The cleavage specificity of recombinant SmSP2 was determined using positional scanning and multiplex combinatorial libraries and the determinants of specificity were identified with 3D homology models, demonstrating a trypsin-like endopeptidase mode of action. SmSP2 displayed restricted proteolysis on protein substrates. It activated tissue plasminogen activator and plasminogen as key components of the fibrinolytic system, and released the vasoregulatory peptide, kinin, from kininogen. SmSP2 was detected in the surface tegument, esophageal glands and reproductive organs of the adult parasite by immunofluorescence microscopy, and in the excretory/secretory products by immunoblotting. CONCLUSIONS/SIGNIFICANCE: The data suggest that SmSP2 is secreted, functions at the host-parasite interface and contributes to the survival of the parasite by manipulating host vasodilatation and fibrinolysis. SmSP2 may be, therefore, a potential target for anti-schistosomal therapy.


Asunto(s)
Hemostáticos/antagonistas & inhibidores , Schistosoma mansoni/enzimología , Esquistosomiasis mansoni/parasitología , Serina Endopeptidasas/farmacología , Secuencia de Aminoácidos , Animales , Coagulación Sanguínea/efectos de los fármacos , Presión Sanguínea/efectos de los fármacos , Femenino , Fibrinólisis/efectos de los fármacos , Proteínas del Helminto/química , Proteínas del Helminto/genética , Proteínas del Helminto/farmacología , Masculino , Modelos Moleculares , Plasminógeno/efectos de los fármacos , Dominios Proteicos , Proteolisis/efectos de los fármacos , Proteínas Recombinantes , Serina Endopeptidasas/química , Serina Endopeptidasas/genética , Activador de Tejido Plasminógeno/efectos de los fármacos , Vasodilatación/efectos de los fármacos
11.
J Thromb Haemost ; 5(5): 1047-54, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17459007

RESUMEN

BACKGROUND: Prourokinase (prouPA) is unstable in plasma at therapeutic concentrations. A mutant form, M5, made more stable by reducing its intrinsic activity was therefore developed. Activation to two-chain M5 (tcM5) induced a higher catalytic activity than two-chain urokinase plasminogen activator (tcuPA), implicating an active site functional difference. Consistent with this, an unusual tcM5 complex with plasma C1-inhibitor was recently described in dog and human plasma. The effect of C1-inhibitor on fibrinolysis and fibrinogenolysis by M5 is the subject of this study. METHODS AND RESULTS: Zymograms of tcM5 and tcuPA incubated in plasma revealed prominent tcM5-C1-inhibitor complexes, which formed within 5 min. The inhibition rate by purified human C1-inhibitor (250 microg mL(-1)) was about 7-fold faster for tcM5 than it was for tcuPA (10 microg mL(-1)). The effect of the inhibitor on the stability of M5 and prouPA was determined by incubating them in plasma at high concentrations (10-20 microg mL(-1)) +/- C1-inhibitor supplementation. Above 10 microg mL(-1), depletion of all plasma plasminogen occurred, indicating plasmin generation and tcM5/tcuPA formation. With supplemental C1-inhibitor, M5 stability was restored but not prouPA stability. Clot lysis by M5 +/- supplemental C1-inhibitor showed no attenuation of the rate of fibrinolysis, whereas fibrinogenolysis was prevented by C1-inhibitor. Moreover, because of higher dose-tolerance, the rate of fibrin-specific lysis reached that achievable by non-specific fibrinolysis without inhibitor. CONCLUSIONS: Plasma C1-inhibitor stabilized M5 in its proenzyme configuration in plasma by inhibiting tcM5 and thereby non-specific plasminogen activation. At the same time, fibrin-specific plasminogen activation remained unimpaired. This unusual dissociation of effects has significant implications for improving the safety and efficacy of fibrinolysis.


Asunto(s)
Proteínas Inactivadoras del Complemento 1/farmacología , Fibrina/metabolismo , Plasminógeno/efectos de los fármacos , Serpinas/farmacología , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Proteína Inhibidora del Complemento C1 , Electroforesis en Gel de Poliacrilamida , Fibrinólisis , Humanos , Plasminógeno/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/genética
12.
Arch Oral Biol ; 52(7): 663-8, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17296163

RESUMEN

OBJECTIVE: Chronic inflammation in periodontal disease is associated with increased plasminogen activation and elevated levels of chemokines. It is unknown whether chemokines can regulate the activation of plasminogen via modulation of plasminogen activators (PA) and the corresponding plasminogen activator inhibitors (PAI) in periodontal tissue. DESIGN: To establish a link between chemokines and activation of plasminogen, human periodontal ligament fibroblasts (PDL) and gingival fibroblasts (GF) were incubated with IL-8, monocyte chemoattractant protein-1, macrophage inflammatory protein-1alpha, and platelet factor-4, either alone or in the presence of the inflammatory mediators TGF-beta and IL-1. The potential of the cell lysates to activate plasminogen was based on kinetic studies with the substrate casein. Casein zymography was performed to determine the molecular sizes of the PA. Total PAI-1 in the cell-conditioned medium was quantified by immunoassay. RESULTS: We report that the chemokines did not affect activation of plasminogen by PDL and GF. Even in the presence of TGF-beta which suppressed, and IL-1 which stimulated plasminogen activation, the chemokines had no direct effect. Inhibition of PA and plasmin, but not of matrix metalloproteinases and cysteine proteinases prevented caseinolysis. The plasminogen activation capacity of the cell lysates was represented by a single band with features of uPA. The immunoassay showed that the release of PAI-1 in PDL and GF remained unaffected by the chemokines, also when stimulated with TGF-beta. CONCLUSIONS: These results suggest that plasminogen activation by PDL and GF is not directly affected by the chemokines even in the presence of the inflammatory mediators TGF-beta and IL-1.


Asunto(s)
Quimiocinas/farmacología , Fibroblastos/metabolismo , Encía/metabolismo , Ligamento Periodontal/metabolismo , Plasminógeno/metabolismo , Caseínas/metabolismo , Células Cultivadas , Quimiocina CCL2/farmacología , Quimiocina CCL3/farmacología , Medios de Cultivo Condicionados , Inhibidores de Cisteína Proteinasa/farmacología , Fibroblastos/efectos de los fármacos , Encía/citología , Encía/efectos de los fármacos , Humanos , Mediadores de Inflamación/farmacología , Interleucina-1/farmacología , Interleucina-8/farmacología , Inhibidores de la Metaloproteinasa de la Matriz , Ligamento Periodontal/citología , Ligamento Periodontal/efectos de los fármacos , Plasminógeno/efectos de los fármacos , Activadores Plasminogénicos/análisis , Activadores Plasminogénicos/efectos de los fármacos , Inactivadores Plasminogénicos/metabolismo , Factor Plaquetario 4/farmacología , Factor de Crecimiento Transformador beta/farmacología , Activador de Plasminógeno de Tipo Uroquinasa/análisis
13.
J Am Heart Assoc ; 6(2)2017 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-28159824

RESUMEN

BACKGROUND: Thrombolytic therapy for acute thrombosis is limited by life-threatening side effects such as major bleeding and neurotoxicity. New treatment options with enhanced fibrinolytic potential are therefore required. Here, we report the development of a new thrombolytic molecule that exploits key features of thrombosis. We designed a recombinant microplasminogen modified to be activated by the prothrombotic serine-protease thrombin (HtPlg), fused to an activation-specific anti-glycoprotein IIb/IIIa single-chain antibody (SCE5), thereby hijacking the coagulation system to initiate thrombolysis. METHODS AND RESULTS: The resulting fusion protein named SCE5-HtPlg shows in vitro targeting towards the highly abundant activated form of the fibrinogen receptor glycoprotein IIb/IIIa expressed on activated human platelets. Following thrombin formation, SCE5-HtPlg is activated to contain active microplasmin. We evaluate the effectiveness of our targeted thrombolytic construct in two models of thromboembolic disease. Administration of SCE5-HtPlg (4 µg/g body weight) resulted in effective thrombolysis 20 minutes after injection in a ferric chloride-induced model of mesenteric thrombosis (48±3% versus 92±5% for saline control, P<0.01) and also reduced emboli formation in a model of pulmonary embolism (P<0.01 versus saline). Furthermore, at these effective therapeutic doses, the SCE5-HtPlg did not prolong bleeding time compared with saline (P=0.99). CONCLUSIONS: Our novel fusion molecule is a potent and effective treatment for thrombosis that enables in vivo thrombolysis without bleeding time prolongation. The activation of this construct by thrombin generated within the clot itself rather than by a plasminogen activator, which needs to be delivered systemically, provides a novel targeted approach to improve thrombolysis.


Asunto(s)
Coagulación Sanguínea/efectos de los fármacos , Fibrinolíticos/uso terapéutico , Fragmentos de Péptidos/biosíntesis , Plasminógeno/biosíntesis , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/uso terapéutico , Anticuerpos de Cadena Única/uso terapéutico , Terapia Trombolítica/métodos , Trombosis/tratamiento farmacológico , Plaquetas/efectos de los fármacos , Western Blotting , Citometría de Flujo , Humanos , Fragmentos de Péptidos/efectos de los fármacos , Plasminógeno/efectos de los fármacos , Activación Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/inmunología , Anticuerpos de Cadena Única/inmunología , Trombosis/sangre
14.
Circ Res ; 90(7): 757-63, 2002 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-11964367

RESUMEN

Prourokinase (proUK) is a zymogenic plasminogen activator that at pharmacological doses is prone to nonspecific activation to urokinase. This has handicapped therapeutic exploitation of its fibrin-specific physiological properties. To attenuate this susceptibility without compromising specific activation of proUK on a fibrin clot, a Lys300-->His mutation (M5) was developed. M5 had a lower intrinsic activity and, therefore, remained stable in plasma at a 4-fold higher concentration than did proUK. M5 had a higher 2-chain activity and induced more rapid plasminogen activation and fibrin-specific clot lysis in vitro. Sixteen dogs embolized with radiolabeled clots were infused with saline, proUK, tissue plasminogen activator, or M5. The lower intrinsic activity allowed a higher infusion rate with M5, which induced the most rapid and efficient clot lysis (50% clot lysis by approximately 600 microg/kg M5 versus approximately 1200 microg/kg proUK). In association with this, M5 caused neither a significant increase in the primary bleeding time nor secondary bleeding (total blood loss). By contrast, these measurements increased 4-fold and 5-fold, respectively, with proUK and >5-fold and 8-fold, respectively, with tissue plasminogen activator. Clot lysis by M5 and hemostasis were further evaluated in 6 rhesus monkeys. M5 again induced rapid clot lysis without a significant increase in the primary bleeding time, and secondary bleeding did not occur. In conclusion, a site-directed mutation designed to improve the stability of proUK in blood at therapeutic concentrations induced superior clot lysis in vitro and in vivo without causing significant interference with hemostasis.


Asunto(s)
Coagulación Sanguínea/efectos de los fármacos , Fibrinólisis/efectos de los fármacos , Hemostasis/efectos de los fármacos , Proteínas Recombinantes/farmacología , Terapia Trombolítica/métodos , Trombosis/tratamiento farmacológico , Activador de Plasminógeno de Tipo Uroquinasa/farmacología , Sustitución de Aminoácidos , Animales , Tiempo de Sangría , Modelos Animales de Enfermedad , Perros , Evaluación Preclínica de Medicamentos , Estabilidad de Medicamentos , Activación Enzimática/efectos de los fármacos , Activación Enzimática/genética , Vena Femoral/efectos de los fármacos , Fibrina/efectos de los fármacos , Fibrina/metabolismo , Hemorragia/prevención & control , Humanos , Macaca mulatta , Masculino , Mutagénesis Sitio-Dirigida , Plasma/efectos de los fármacos , Plasma/metabolismo , Plasminógeno/efectos de los fármacos , Plasminógeno/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Activador de Tejido Plasminógeno/farmacología , Activador de Plasminógeno de Tipo Uroquinasa/genética , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo
15.
Postepy Biochem ; 52(1): 80-6, 2006.
Artículo en Polaco | MEDLINE | ID: mdl-16869305

RESUMEN

Staphylokinase is a 135 amino acid protein produced by certain strains of Staphylococcus aureus. It belongs to fibrin-specific plasminogen activator. Staphylokinase converts plasminogen--the inactive proenzyme--to the plasmin, which dissolves the fibrin of a blood clots. This review will focus on the biochemical and thrombolytic properties of staphylokinase and its derivatives, which would make use of treatment in acute myocardial infarction and other cardiovascular diseases.


Asunto(s)
Fibrinolíticos/farmacología , Metaloendopeptidasas/farmacología , Activador de Tejido Plasminógeno/farmacología , Animales , Fibrinolisina/metabolismo , Fibrinólisis/efectos de los fármacos , Humanos , Metaloendopeptidasas/uso terapéutico , Infarto del Miocardio/tratamiento farmacológico , Plasminógeno/efectos de los fármacos , Plasminógeno/metabolismo , Proteínas Recombinantes/uso terapéutico , Estreptoquinasa/farmacología , Estreptoquinasa/uso terapéutico , Terapia Trombolítica , Activador de Tejido Plasminógeno/uso terapéutico
16.
Biochim Biophys Acta ; 1205(2): 258-61, 1994 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-8155706

RESUMEN

We examined the effects of 5,6-trans-prostaglandin E2 (trans-PG E2) on fibrinolysis and plasminogen activation by either urokinase or streptokinase. trans-PG E2 was found to enhance fibrinolysis induced by urokinase and inhibit the one by streptokinase. These effects were also appeared in the method using synthetic chromogenic substrate S-2251, which suggested that the effects of trans-PG E2 were induced in the circumstances without coagulation factors such as fibrinogen, thrombin or fibrin. Moreover, the enhancement effect of trans-PG E2 on fibrinolysis by urokinase was investigated. The result of SDS-PAGE indicated that plasmin formation rate from plasminogen by urokinase was accelerated in the presence of trans-PG E2. As trans-PG E2 increased the hydrolyzing rate of S-2288 by urokinase, trans-PG E2 directly interacted with urokinase. Therefore, the enhancement effect of trans-PG E2 on plasminogen activation by urokinase could be explained, at least in part, as follows: at first trans-PG E2 directly exerts its effect on urokinase, then it causes the increase of generation rate of plasmin from plasminogen.


Asunto(s)
Dinoprostona/farmacología , Plasminógeno/metabolismo , Estreptoquinasa/farmacología , Activador de Plasminógeno de Tipo Uroquinasa/farmacología , Secuencia de Aminoácidos , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Isomerismo , Datos de Secuencia Molecular , Plasminógeno/efectos de los fármacos
17.
J Am Coll Cardiol ; 19(5): 1085-90, 1992 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-1532403

RESUMEN

Although initially developed to reduce the risk of bleeding, second-generation (clot-selective) thrombolytic agents have been found to induce more prompt and frequent recanalization than do nonselective, first-generation agents. To determine whether they do so in part by preserving clot-associated plasminogen, human whole blood clots formed in Chandler tubes were studied. Addition of suprapharmacologic concentrations of recombinant tissue-type plasminogen activator (rt-PA) to the media bathing mature clots led to a paradoxic impairment of clot lysis and a concomitant concentration-dependent depletion of clot-associated plasminogen (Western blot analysis). In contrast, supplementation of the plasma with plasminogen (0.27 mg/ml) led to significant conservation of both plasma and clot-associated plasminogen (p less than or equal to 0.05, n = 4), and prevented the diminution of clot lysis (p less than or equal to 0.05; n = 4). Fibrinogen degradation products did not account for the attenuation of lysis with the highest concentrations of rt-PA. In concentrations equivalent to those that were induced by the highest concentrations of rt-PA evaluated, fibrinogen degradation products potentiated rather than inhibited lysis (p less than or equal to 0.05, n = 4), probably by stimulating rt-PA activity directly. When preformed clots were incubated with plasminogen-depleted plasma plus 1,000 ng/ml rt-PA, the plasminogen content in residual clot declined (9.36 +/- 0.46 versus 12.39 +/- 0.69 ng/mg clot found in nondepleted plasma; p less than or equal to 0.05; n = 6). Furthermore, clot lysis was attenuated completely.(ABSTRACT TRUNCATED AT 250 WORDS)


Asunto(s)
Coagulación Sanguínea/efectos de los fármacos , Fibrinólisis/efectos de los fármacos , Plasminógeno/efectos de los fármacos , Activador de Tejido Plasminógeno/farmacología , Coagulación Sanguínea/fisiología , Fibrinógeno/efectos de los fármacos , Fibrinógeno/fisiología , Fibrinolisina/análisis , Fibrinólisis/fisiología , Humanos , Técnicas In Vitro , Plasminógeno/análisis , Plasminógeno/fisiología , Proteínas Recombinantes/farmacología
18.
FASEB J ; 17(2): 262-4, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12490545

RESUMEN

Vascular endothelial growth factor (VEGF) is not only essential for vasculogenesis and angiogenesis but also is a potent inducer of vascular permeability. Although a dissection of the molecular pathways between angiogenesis- and vascular permeability-inducing properties would be desirable for the development of angiogenic and anti-angiogenic therapies, such mechanisms have not been identified yet. Here we provide evidence for a role of the p38 MAPK as the signaling molecule that separates these two processes. Inhibition of p38 MAPK activity enhances VEGF-induced angiogenesis in vitro and in vivo, a finding that was accompanied by prolonged Erk1/2 MAPK activation, increased endothelial survival, and plasminogen activation. Conversely, the same inhibitors abrogate VEGF-induced vascular permeability in vitro and in vivo. These dualistic properties of p38 MAPK are relevant not only for therapeutic angiogenesis but also for reducing edema formation and enhancing tissue repair in ischemic diseases.


Asunto(s)
Permeabilidad Capilar/efectos de los fármacos , Factores de Crecimiento Endotelial/farmacología , Péptidos y Proteínas de Señalización Intercelular/farmacología , Linfocinas/farmacología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neovascularización Patológica/enzimología , Alantoína/fisiología , Animales , Supervivencia Celular/efectos de los fármacos , Embrión de Pollo , Corion/irrigación sanguínea , Corion/fisiología , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/fisiopatología , Inhibidores Enzimáticos/farmacología , Factor 2 de Crecimiento de Fibroblastos/farmacología , Humanos , Imidazoles/farmacología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Neovascularización Fisiológica/efectos de los fármacos , Fosforilación/efectos de los fármacos , Plasminógeno/efectos de los fármacos , Plasminógeno/metabolismo , Piridinas/farmacología , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular , Proteínas Quinasas p38 Activadas por Mitógenos
19.
FEBS Lett ; 418(1-2): 58-62, 1997 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-9414095

RESUMEN

Staplabin (0.3-0.6 mM), a fungal triprenyl phenol, enhanced 3-fold the plasminogen activator-catalyzed activation of Glu-plasminogen and Lys-plasminogen as well as their binding to fibrin. Staplabin was not stimulatory to the amidolytic activity of plasmin and plasminogen activators. Even in the presence of epsilon-aminocaproic acid (EACA) and fibrinogen fragments, allosteric effectors for Glu-plasminogen, staplabin increased the activation of both forms of plasminogen. In size-exclusion chromatography of Glu-plasminogen and Lys-plasminogen, the molecular elution time, which varies as the conformation of a protein changes, was shortened by staplabin. These results suggest that staplabin causes plasminogens to be more susceptible to activation and fibrin binding by inducing a conformational change that is, at least in part, different from that induced by EACA and fibrinogen fragments.


Asunto(s)
Benzopiranos/farmacología , Fibrina/metabolismo , Fibrinolisina/metabolismo , Fragmentos de Péptidos/metabolismo , Activadores Plasminogénicos/farmacología , Plasminógeno/metabolismo , Conformación Proteica/efectos de los fármacos , Pirrolidinonas/farmacología , Regulación Alostérica , Ácido Aminocaproico/farmacología , Activación Enzimática , Fibrinógeno/metabolismo , Humanos , Cinética , Fragmentos de Péptidos/química , Fragmentos de Péptidos/farmacología , Plasminógeno/química , Plasminógeno/efectos de los fármacos , Trombina/metabolismo
20.
Thromb Haemost ; 65(3): 280-5, 1991 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-1904654

RESUMEN

The binding of t-PA to fibrin is mediated both by its "finger" (F) and its "kringle 2" (K2) domain. In addition, these domains are involved in the stimulation of t-PA activity by fibrin. We analyzed the kinetic characteristics of Glu-plasminogen activation by t-PA and a set of t-PA deletion mutants in the absence and the presence of desA-fibrin. In the absence of desA-fibrin, the activity of t-PA (variants) is determined by the presence of the protease domain, irrespective of the composition of the amino-terminal heavy chain. In the presence of the cofactor desA-fibrin, the activity of t-PA (variants) is dependent on the domain composition of the heavy chain. The activity of t-PA is stimulated 2,400 fold by desA-fibrin, whereas the activity of the mutant lacking the K1 domain (del. K1) increases 936 fold in the presence of this cofactor. Mutants lacking either the K2 domain (del. K2) or the F domain (del. F) exhibit an enhanced activity upon desA-fibrin addition of 200 and 210 fold, respectively. DesA-fibrin has no stimulatory effect on the activity of the mutant containing only the serine-protease domain (del.FE K1 K2) nor on the activity of the variant containing only the K1 domain and the serine-protease domain (del. FE K2). Furthermore, we determined the relative fibrin affinity of each t-PA variant, which is similarly dictated by the composition of the heavy chain.


Asunto(s)
Plasminógeno/efectos de los fármacos , Activador de Tejido Plasminógeno/farmacología , Secuencia de Aminoácidos , Fibrina/metabolismo , Cinética , Datos de Secuencia Molecular , Mutación , Unión Proteica , Activador de Tejido Plasminógeno/genética , Activador de Tejido Plasminógeno/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA