Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 161
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Circ Res ; 128(3): 386-400, 2021 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-33292062

RESUMEN

RATIONALE: Current thrombolytic agents activate plasminogen to plasmin which triggers fibrinolysis to dissolve thrombi. Since plasmin is a nonspecific proteolytic enzyme, all of the current plasmin-dependent thrombolytics lead to serious hemorrhagic complications, demanding a new class of fibrinolytic enzymes independent from plasmin activation and undesirable side effects. We speculated that the mammalian version of bacterial heat-shock proteins could selectively degrade intravascular thrombi, a typical example of a highly aggregated protein mixture. OBJECTIVE: The objective of this study is to identify enzymes that can dissolve intravascular thrombi specifically without affecting fibrinogen and fibronectin so that the wound healing processes remain uninterrupted and tissues are not damaged. In this study, HtrA (high-temperature requirement A) proteins were tested for its specific proteolytic activity on intravascular thrombi independently from plasmin activation. METHODS AND RESULTS: HtrA1 and HtrA2/Omi proteins, collectively called as HtrAs, lysed ex vivo blood thrombi by degrading fibrin polymers. The thrombolysis by HtrAs was plasmin-independent and specific to vascular thrombi without causing the systemic activation of plasminogen and preventing nonspecific proteolysis of other proteins including fibrinogen and fibronectin. As expected, HtrAs did not disturb clotting and wound healing of excised wounds from mouse skin. It was further confirmed in a tail bleeding and a rebleeding assay that HtrAs allowed normal clotting and maintenance of clot stability in wounds, unlike other thrombolytics. Most importantly, HtrAs completely dissolved blood thrombi in tail thrombosis mice, and the intravenous injection of HtrAs to mice with pulmonary embolism completely dissolved intravascular thrombi and thus rescued thromboembolism. CONCLUSIONS: Here, we identified HtrA1 and HtrA2/Omi as plasmin-independent and highly specific thrombolytics that can dissolve intravascular thrombi specifically without bleeding risk. This work is the first report of a plasmin-independent thrombolytic pathway, providing HtrA1 and HtrA2/Omi as ideal therapeutic candidates for various thrombotic diseases without hemorrhagic complications.


Asunto(s)
Fibrina/metabolismo , Fibrinólisis/efectos de los fármacos , Fibrinolíticos/farmacología , Serina Peptidasa A1 que Requiere Temperaturas Altas/farmacología , Serina Peptidasa A2 que Requiere Temperaturas Altas/farmacología , Embolia Pulmonar/tratamiento farmacológico , Trombosis/tratamiento farmacológico , Animales , Modelos Animales de Enfermedad , Femenino , Fibrinolíticos/toxicidad , Hemorragia/inducido químicamente , Serina Peptidasa A1 que Requiere Temperaturas Altas/toxicidad , Serina Peptidasa A2 que Requiere Temperaturas Altas/toxicidad , Humanos , Cinética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Embolia Pulmonar/sangre , Embolia Pulmonar/enzimología , Proteínas Recombinantes/farmacología , Trombosis/sangre , Trombosis/enzimología , Cicatrización de Heridas/efectos de los fármacos
2.
Circ Res ; 128(1): 62-75, 2021 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-33070717

RESUMEN

RATIONALE: Hemorrhagic complications represent a major limitation of intravenous thrombolysis using tPA (tissue-type plasminogen activator) in patients with ischemic stroke. The expression of tPA receptors on immune cells raises the question of what effects tPA exerts on these cells and whether these effects contribute to thrombolysis-related hemorrhagic transformation. OBJECTIVE: We aim to determine the impact of tPA on immune cells and investigate the association between observed immune alteration with hemorrhagic transformation in ischemic stroke patients and in a rat model of embolic stroke. METHODS AND RESULTS: Paired blood samples were collected before and 1 hour after tPA infusion from 71 patients with ischemic stroke. Control blood samples were collected from 27 ischemic stroke patients without tPA treatment. A rat embolic middle cerebral artery occlusion model was adopted to investigate the underlying mechanisms of hemorrhagic transformation. We report that tPA induces a swift surge of circulating neutrophils and T cells with profoundly altered molecular features in ischemic stroke patients and a rat model of focal embolic stroke. tPA exacerbates endothelial injury, increases adhesion and migration of neutrophils and T cells, which are associated with brain hemorrhage in rats subjected to embolic stroke. Genetic ablation of annexin A2 in neutrophils and T cells diminishes the effect of tPA on these cells. Decoupling the interaction between mobilized neutrophils/T cells and the neurovascular unit, achieved via a S1PR (sphingosine-1-phosphate receptor) 1 modulator RP101075 and a CCL2 (C-C motif chemokine ligand 2) synthesis inhibitor bindarit, which block lymphocyte egress and myeloid cell recruitment, respectively, attenuates hemorrhagic transformation and improves neurological function after tPA thrombolysis. CONCLUSIONS: Our findings suggest that immune invasion of the neurovascular unit represents a previously unrecognized mechanism underlying tPA-mediated brain hemorrhage, which can be overcome by precise immune modulation during thrombolytic therapy.


Asunto(s)
Accidente Cerebrovascular Embólico/tratamiento farmacológico , Fibrinolíticos/toxicidad , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Hemorragias Intracraneales/inducido químicamente , Accidente Cerebrovascular Isquémico/tratamiento farmacológico , Neutrófilos/efectos de los fármacos , Linfocitos T/efectos de los fármacos , Terapia Trombolítica , Activador de Tejido Plasminógeno/toxicidad , Animales , Anexina A2/metabolismo , Línea Celular , Quimiocina CCL2/metabolismo , Quimiotaxis de Leucocito/efectos de los fármacos , Modelos Animales de Enfermedad , Accidente Cerebrovascular Embólico/sangre , Accidente Cerebrovascular Embólico/inmunología , Femenino , Fibrinolíticos/administración & dosificación , Humanos , Infarto de la Arteria Cerebral Media/sangre , Infarto de la Arteria Cerebral Media/inmunología , Infusiones Intravenosas , Hemorragias Intracraneales/sangre , Hemorragias Intracraneales/inmunología , Accidente Cerebrovascular Isquémico/sangre , Accidente Cerebrovascular Isquémico/inmunología , Masculino , Infiltración Neutrófila/efectos de los fármacos , Neutrófilos/inmunología , Neutrófilos/metabolismo , Ratas Wistar , Receptores de Esfingosina-1-Fosfato/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Activador de Tejido Plasminógeno/administración & dosificación
3.
Arterioscler Thromb Vasc Biol ; 41(2): 668-682, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33297751

RESUMEN

OBJECTIVE: Current antiplatelet medications increase the risk of bleeding, which leads to a clear clinical need in developing novel mechanism-based antiplatelet drugs. TYMP (Thymidine phosphorylase), a cytoplasm protein that is highly expressed in platelets, facilitates multiple agonist-induced platelet activation, and enhances thrombosis. Tipiracil hydrochloride (TPI), a selective TYMP inhibitor, has been approved by the Food and Drug Administration for clinical use. We tested the hypothesis that TPI is a safe antithrombotic medication. Approach and Results: By coexpression of TYMP and Lyn, GST (glutathione S-transferase) tagged Lyn-SH3 domain or Lyn-SH2 domain, we showed the direct evidence that TYMP binds to Lyn through both SH3 and SH2 domains, and TPI diminished the binding. TYMP deficiency significantly inhibits thrombosis in vivo in both sexes. Pretreatment of platelets with TPI rapidly inhibited collagen- and ADP-induced platelet aggregation. Under either normal or hyperlipidemic conditions, treating wild-type mice with TPI via intraperitoneal injection, intravenous injection, or gavage feeding dramatically inhibited thrombosis without inducing significant bleeding. Even at high doses, TPI has a lower bleeding side effect compared with aspirin and clopidogrel. Intravenous delivery of TPI alone or combined with tissue plasminogen activator dramatically inhibited thrombosis. Dual administration of a very low dose of aspirin and TPI, which had no antithrombotic effects when used alone, significantly inhibited thrombosis without disturbing hemostasis. CONCLUSIONS: This study demonstrated that inhibition of TYMP, a cytoplasmic protein, attenuated multiple signaling pathways that mediate platelet activation, aggregation, and thrombosis. TPI can be used as a novel antithrombotic medication without the increase in risk of bleeding.


Asunto(s)
Plaquetas/efectos de los fármacos , Trombosis de las Arterias Carótidas/prevención & control , Inhibidores Enzimáticos/farmacología , Fibrinolíticos/farmacología , Activación Plaquetaria/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/farmacología , Pirrolidinas/farmacología , Timidina Fosforilasa/antagonistas & inhibidores , Timina/farmacología , Animales , Aspirina/farmacología , Plaquetas/enzimología , Células COS , Trombosis de las Arterias Carótidas/sangre , Trombosis de las Arterias Carótidas/enzimología , Trombosis de las Arterias Carótidas/genética , Chlorocebus aethiops , Modelos Animales de Enfermedad , Terapia Antiplaquetaria Doble , Inhibidores Enzimáticos/toxicidad , Femenino , Fibrinolíticos/toxicidad , Hemorragia/inducido químicamente , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Adhesividad Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/toxicidad , Unión Proteica , Pirrolidinas/toxicidad , Transducción de Señal , Timidina Fosforilasa/genética , Timidina Fosforilasa/metabolismo , Timina/toxicidad , Dominios Homologos src , Familia-src Quinasas/genética , Familia-src Quinasas/metabolismo
4.
Arterioscler Thromb Vasc Biol ; 41(1): e1-e17, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33232198

RESUMEN

OBJECTIVE: TSP-1 (thrombospondin 1) is one of the most expressed proteins in platelet α-granules and plays an important role in the regulation of hemostasis and thrombosis. Interaction of released TSP-1 with CD47 membrane receptor has been shown to regulate major events leading to thrombus formation, such as, platelet adhesion to vascular endothelium, nitric oxide/cGMP (cyclic guanosine monophosphate) signaling, platelet activation as well as aggregation. Therefore, targeting TSP-1:CD47 axis may represent a promising antithrombotic strategy. Approach and Results: A CD47-derived cyclic peptide was engineered, namely TAX2, that targets TSP-1 and selectively prevents TSP-1:CD47 interaction. Here, we demonstrate for the first time that TAX2 peptide strongly decreases platelet aggregation and interaction with collagen under arterial shear conditions. TAX2 also delays time for complete thrombotic occlusion in 2 mouse models of arterial thrombosis following chemical injury, while Thbs1-/- mice recapitulate TAX2 effects. Importantly, TAX2 administration is not associated with increased bleeding risk or modification of hematologic parameters. CONCLUSIONS: Overall, this study sheds light on the major contribution of TSP-1:CD47 interaction in platelet activation and thrombus formation while putting forward TAX2 as an innovative antithrombotic agent with high added-value.


Asunto(s)
Arteriopatías Oclusivas/prevención & control , Antígeno CD47/antagonistas & inhibidores , Fibrinolíticos/farmacología , Péptidos Cíclicos/farmacología , Inhibidores de Agregación Plaquetaria/farmacología , Agregación Plaquetaria/efectos de los fármacos , Trombosis/prevención & control , Trombospondina 1/antagonistas & inhibidores , Animales , Arteriopatías Oclusivas/sangre , Arteriopatías Oclusivas/metabolismo , Antígeno CD47/metabolismo , Colágeno/metabolismo , Modelos Animales de Enfermedad , Fibrinolíticos/toxicidad , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Péptidos Cíclicos/toxicidad , Inhibidores de Agregación Plaquetaria/toxicidad , Ratas Sprague-Dawley , Transducción de Señal , Trombosis/sangre , Trombosis/metabolismo , Trombospondina 1/genética , Trombospondina 1/metabolismo , Factores de Tiempo
5.
Biol Pharm Bull ; 44(2): 211-218, 2021 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-33281147

RESUMEN

Subtilisin NAT, a Bacillus subtilisin, is widely applied as a functional food and considered to be one of the most exploitable potential oral thrombolytic agents. Subtilisin QK, another Bacillus subtilisin, is a serine protease fermented by Bacillus subtilis 02 and has a better thrombolytic effect. Therefore, subtilisin QK is typically used for evaluating the safety of Bacillus subtilisins. Here, we conduct several good laboratory practice (GLP)-compliant studies in non-rodent animal, i.e., in Beagle dogs, including acute toxicity, subchronic toxicity, and safety pharmacology studies. No adverse effects were evident in the acute and 28-d subchronic toxicity studies at doses up to 40000 FU/kg and 16000 FU/kg/d, respectively. In evaluating the pharmacological safety of up to 2000FU/kg subtilisin QK, we found no significant differences between the electrocardiograms, blood pressures, and respiration of beagle dogs. These findings suggest the safety of Bacillus subtilisin, providing reliable pharmacological and toxicological data for its development and popularization as a functional food and drug.


Asunto(s)
Fibrinolíticos/toxicidad , Subtilisinas/toxicidad , Animales , Perros , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Evaluación Preclínica de Medicamentos , Femenino , Fibrinolíticos/administración & dosificación , Masculino , Subtilisinas/administración & dosificación , Pruebas de Toxicidad Aguda , Pruebas de Toxicidad Subcrónica
6.
Mar Drugs ; 19(4)2021 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-33921137

RESUMEN

2,5-Bis-[8-(4,8-dimethyl-nona-3,7-dienyl)-5,7-dihydroxy-8-methyl-3-keto-1,2,7,8-teraahydro-6H-pyran[a]isoindol-2-yl]-pentanoic acid (FGFC1) is a marine pyran-isoindolone derivative isolated from a rare marine microorganism Stachybotrys longispora FG216, which showed moderate antithrombotic(fibrinolytic) activity. To further enhance its antithrombotic effect, a series of new FGFC1 derivatives (F1-F7) were synthesized via chemical modification at C-2 and C-2' phenol groups moieties and C-1″ carboxyl group. Their fibrinolytic activities in vitro were evaluated. Among the derivatives, F1-F4 and F6 showed significant fibrinolytic activities with EC50 of 59.7, 87.1, 66.6, 82.8, and 42.3 µM, respectively, via enhancement of urokinase activity. Notably, derivative F6 presented the most remarkable fibrinolytic activity (2.72-fold than that of FGFC1). Furthermore, the cytotoxicity of derivative F6 was tested as well as expression of Fas/Apo-1 and IL-1 on HeLa cells. The results showed that, compared to FGFC1, derivative F6 possessed moderate cytotoxicity and apoptotic effect on HeLa cells (statistical significance p > 0.1), making F6 a potential antithrombotic agent towards clinical application.


Asunto(s)
Fibrinólisis/efectos de los fármacos , Fibrinolíticos/farmacología , Isoindoles/farmacología , Piranos/farmacología , Stachybotrys/metabolismo , Apoptosis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Fibrinolíticos/síntesis química , Fibrinolíticos/aislamiento & purificación , Fibrinolíticos/toxicidad , Células HeLa , Humanos , Isoindoles/síntesis química , Isoindoles/aislamiento & purificación , Isoindoles/toxicidad , Estructura Molecular , Piranos/síntesis química , Piranos/aislamiento & purificación , Piranos/toxicidad , Relación Estructura-Actividad
7.
Pak J Pharm Sci ; 34(5(Supplementary)): 1885-1890, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34836855

RESUMEN

The facile and efficient protocol for the synthesis of N-phenyl piperazine based di-thio-carbamates has been reported under neat conditions. A library of novel piperazine based di-thio-carbamates (3a-h) in excellent yields has been prepared. Solvent free, catalyst free and easy work up conditions make this protocol an attractive synthetic protocol to achieve novel biologically active di-thio-carbamates. The synthesized molecules have been characterized by FT-IR, 1HNMR and 13CNMR spectroscopic techniques. The pharmacological aspects of these derivatives have been evaluated via hemolysis and thrombolysis. All the target molecules (3a-h) exhibit mild to medium potential as hemolytic and thrombolytic agents. Among the synthesized derivatives, compound 3c showed least cytotoxicity and better thrombolytic potential.


Asunto(s)
Fibrinolíticos/síntesis química , Fibrinolíticos/farmacología , Tecnología Química Verde/métodos , Hemolíticos/síntesis química , Hemolíticos/farmacología , Piperazinas/síntesis química , Piperazinas/farmacología , Tiocarbamatos/síntesis química , Tiocarbamatos/farmacología , Fibrinolíticos/toxicidad , Hemólisis/efectos de los fármacos , Hemolíticos/toxicidad , Humanos , Estructura Molecular , Piperazinas/toxicidad , Relación Estructura-Actividad , Tiocarbamatos/toxicidad
8.
Cardiovasc Drugs Ther ; 34(1): 15-23, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32062793

RESUMEN

PURPOSE: Drugs inhibiting the platelet P2Y12 receptor, such as clopidogrel and prasugrel, are potent antithrombotic agents and are widely used in cardiovascular disease. However, the adverse effects of these drugs have limited their clinical use. For example, clopidogrel resistance occurs in approximately one third of patients, while prasugrel increases the risk of major bleeding. Therefore, new generations of such drugs are of clinical interest. METHODS: In this study, the pharmacodynamics of a new P2Y12 antagonist, CN-218, was compared with that of clopidogrel and prasugrel in rats and mice. The differences between CN-218 and clopidogrel include deuteration of the 7-position methyl carboxylate and the introduction of cinnamate in the 2-position of thiophene. RESULTS: CN-218 had an antiaggregatory efficacy that was at least five times more potent than that of clopidogrel but not as potent as that of prasugrel. It had a significant impact on activated partial thromboplastin time (APTT), whereby the APTT of CN-218-treated rats was approximately 9 s longer than that of the vehicle- or clopidogrel-treated group, while it had no impact on prothrombin time (PT) in rats. CN-218 had a similar potent antithrombotic effect to that of prasugrel and clopidogrel and also reduced the risk of bleeding compared to prasugrel. CONCLUSION: CN-218 may be a promising antithrombotic agent, with potent antiplatelet and significant anticoagulant activity, as well as a lower risk of bleeding compared to clopidogrel and prasugrel.


Asunto(s)
Coagulación Sanguínea/efectos de los fármacos , Plaquetas/efectos de los fármacos , Fibrinolíticos/farmacología , Piperidinas/farmacología , Inhibidores de Agregación Plaquetaria/farmacología , Agregación Plaquetaria/efectos de los fármacos , Antagonistas del Receptor Purinérgico P2Y/farmacología , Receptores Purinérgicos P2Y12/efectos de los fármacos , Tiofenos/farmacología , Trombosis/prevención & control , Animales , Plaquetas/metabolismo , Carragenina , Clopidogrel/farmacología , AMP Cíclico/sangre , Modelos Animales de Enfermedad , Fibrinolíticos/toxicidad , Hemorragia/inducido químicamente , Masculino , Ratones , Piperidinas/toxicidad , Inhibidores de Agregación Plaquetaria/toxicidad , Clorhidrato de Prasugrel/farmacología , Antagonistas del Receptor Purinérgico P2Y/toxicidad , Ratas Wistar , Receptores Purinérgicos P2Y12/sangre , Tiofenos/toxicidad , Trombosis/sangre , Trombosis/inducido químicamente
9.
Regul Toxicol Pharmacol ; 103: 282-291, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30790607

RESUMEN

The fibrinolytic enzyme produced by Mucor subtilissimus UCP 1262 was obtained by solid fermentation and purified by ion exchange chromatography using DEAE-Sephadex A50. The enzyme toxicity was evaluated using mammalian cell lineages: HEK-293, J774.A1, Sarcoma-180 and PBMCs which appeared to be viable at a level of 80%. The biochemical parameters of the mice treated with an acute dose of enzyme (2000 mg/mL) identified alterations of AST and ALT and the histomorphometric analysis of the liver showed a loss of endothelial cells (P < 0.001). However, these changes are considered minimal to affirm that there was a significant degree of hepatotoxicity. The comet assay and the micronucleus test did not identify damage in the DNA of the erythrocytes of the animals treated. The protease did not degrade the Aα and Bß chains of human and bovine fibrinogens, thus indicating that it does not act as anticoagulant, but rather as a fibrinolytic agent. The assay performed to assess blood biocompatibility shows that at dose of 0.3-5 mg/mL the hemolytic grade is considered insignificant. Moreover, the enzyme did not prolong bleeding time in mice when dosed with 1 mg/kg. These results indicate that this enzyme produced is a potential competitor for developing novel antithrombotic drugs.


Asunto(s)
Alanina Transaminasa/metabolismo , Aspartato Aminotransferasas/metabolismo , Fibrinolíticos/toxicidad , Mucor/enzimología , Péptido Hidrolasas/toxicidad , Animales , Relación Dosis-Respuesta a Droga , Células Endoteliales/efectos de los fármacos , Fibrinolíticos/administración & dosificación , Fibrinolíticos/metabolismo , Hígado/efectos de los fármacos , Hígado/patología , Ratones , Péptido Hidrolasas/administración & dosificación , Péptido Hidrolasas/metabolismo
10.
Arterioscler Thromb Vasc Biol ; 37(7): 1307-1314, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28596377

RESUMEN

OBJECTIVE: Currently prescribed antiplatelet drugs have 1 common side effect-an increased risk of hemorrhage and thrombocytopenia. On the contrary, bleeding defects associated with glycoprotein VI (GPVI) expression deficiency are usually slightly prolonged bleeding times. However, GPVI antagonists are lacking in clinic. APPROACH AND RESULTS: Using reverse-phase high-performance liquid chromatography and sequencing, we revealed the partial sequence of trowaglerix α subunit, a potent specific GPVI-targeting snaclec (snake venom C-type lectin protein). Hexapeptide (Troα6 [trowaglerix a chain hexapeptide, CKWMNV]) and decapeptide (Troα10) derived from trowaglerix specifically inhibited collagen-induced platelet aggregation through blocking platelet GPVI receptor. Computational peptide design helped to design a series of Troα6/Troα10 peptides. Protein docking studies on these decapeptides and GPVI suggest that Troα10 was bound at the lower surface of D1 domain and outer surface of D2 domain, which was at the different place of the collagen-binding site and the scFv (single-chain variable fragment) D2-binding site. The newly discovered site was confirmed by inhibitory effects of polyclonal antibodies on collagen-induced platelet aggregation. This indicates that D2 domain of GPVI is a novel and important binding epitope on GPVI-mediated platelet aggregation. Troα6/Troα10 displayed prominent inhibitory effect of thrombus formation in fluorescein sodium-induced platelet thrombus formation of mesenteric venules and ferric chloride-induced carotid artery injury thrombosis model without prolonging the in vivo bleeding time. CONCLUSIONS: We develop a novel antithrombotic peptides derived from trowaglerix that acts through GPVI antagonism with greater safety-no severe bleeding. The binding epitope of polypeptides on GPVI is novel and important. These hexa/decapeptides have therapeutic potential for developing ideal small-mass GPVI antagonists for arterial thrombogenic diseases.


Asunto(s)
Plaquetas/efectos de los fármacos , Traumatismos de las Arterias Carótidas/tratamiento farmacológico , Venenos de Crotálidos/farmacología , Fibrinolíticos/farmacología , Fragmentos de Péptidos/farmacología , Inhibidores de Agregación Plaquetaria/farmacología , Agregación Plaquetaria/efectos de los fármacos , Glicoproteínas de Membrana Plaquetaria/antagonistas & inhibidores , Trombosis/prevención & control , Animales , Sitios de Unión , Plaquetas/metabolismo , Traumatismos de las Arterias Carótidas/sangre , Traumatismos de las Arterias Carótidas/inducido químicamente , Cloruros , Diseño Asistido por Computadora , Venenos de Crotálidos/metabolismo , Venenos de Crotálidos/toxicidad , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Diseño de Fármacos , Compuestos Férricos , Fibrinolíticos/metabolismo , Fibrinolíticos/toxicidad , Fluoresceína , Hemorragia/inducido químicamente , Humanos , Lectinas Tipo C/metabolismo , Masculino , Ratones Endogámicos ICR , Simulación del Acoplamiento Molecular , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/toxicidad , Inhibidores de Agregación Plaquetaria/metabolismo , Inhibidores de Agregación Plaquetaria/toxicidad , Glicoproteínas de Membrana Plaquetaria/metabolismo , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Transducción de Señal/efectos de los fármacos , Trombosis/sangre , Trombosis/inducido químicamente
11.
J Stroke Cerebrovasc Dis ; 27(10): 2761-2767, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30056000

RESUMEN

BACKGROUND: The aim of this study was to assess the risk and the threshold of hemorrhagic transformation (HT) after treatment with recombinant tissue plasminogen activator (rtPA) under the novel oral anticoagulant, rivaroxaban. METHODS: Fifty-three spontaneous hypertensive rats were used in this study. We performed transient middle cerebral artery occlusion for 270 minutes. Placebo, 10 mg/kg or 20 mg/kg rivaroxaban were administered via a stomach tube 180 minutes after induction of ischemia, and rtPA (10 mg/kg) was administered just before reperfusion. Ninety minutes after rivaroxaban administration we measured the rivaroxaban plasma concentration and prothrombin time (PT). HT volume was assessed by hemoglobin spectrophotometry. Additionally, infarct volume, IgG leakage volume, and neurological outcome were assessed. RESULTS: Rivaroxaban plasma concentration and PT increased in a dose dependent manner but were lower than human peak levels after a once-daily dose of 20 mg rivaroxaban. HT volume increased after treatment with 20 mg/kg rivaroxaban compared with placebo treated controls or those treated with 10 mg/kg rivaroxaban (26.5 ± 5.4, 26.8 ± 8.7, and 41.4 ± 12.6 µL in placebo, 10 mg/kg, and 20 mg/kg treated groups, respectively; P < .05). CONCLUSIONS: Our results suggest that even at therapeutic plasma concentrations, rivaroxaban may increase the risk of HT after thrombolysis in some conditions, such as hypertension and/or a prolonged ischemic period.


Asunto(s)
Coagulación Sanguínea/efectos de los fármacos , Inhibidores del Factor Xa/toxicidad , Fibrinolíticos/toxicidad , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Hemorragias Intracraneales/inducido químicamente , Rivaroxabán/toxicidad , Terapia Trombolítica/efectos adversos , Activador de Tejido Plasminógeno/toxicidad , Animales , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Fibrinolíticos/administración & dosificación , Infarto de la Arteria Cerebral Media/sangre , Infarto de la Arteria Cerebral Media/patología , Infarto de la Arteria Cerebral Media/fisiopatología , Hemorragias Intracraneales/sangre , Masculino , Ratas Endogámicas SHR , Proteínas Recombinantes/toxicidad , Factores de Riesgo , Activador de Tejido Plasminógeno/administración & dosificación
12.
Neurobiol Dis ; 108: 173-182, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28830843

RESUMEN

Tissue plasminogen activator (tPA) is administered after ischemic stroke to dissolve intravascular clots, but its use can lead to hemorrhagic transformation (HT). Therapeutic strategies to reduce hemorrhagic complications of tPA might be of benefit for stroke patients. Adenosine A2b receptor (A2bR) plays pivotal roles in regulating vascular protection in peripheral organs. This study explored whether A2bR agonist BAY 60-6583 reduces hemorrhage risk after tPA usage. Using a rat transient middle cerebral artery occlusion model, we showed that mRNA and protein expression of A2bR increased to a greater extent after ischemia-reperfusion than did expression of the other three adenosine receptors (A1, A2a, and A3). tPA administration reduced A2bR expression in ischemic brain microvessels. Post-treatment with BAY 60-6583 (1mg/kg) at the start of reperfusion reduced lesion volume in the absence or presence of tPA (10mg/kg) and attenuated brain swelling, blood-brain barrier disruption, and tPA-exacerbated HT at 24h. Additionally, BAY 60-6583 mitigated sensorimotor deficits in the presence of tPA. BAY 60-6583 inhibited tPA-enhanced matrix metalloprotease-9 activation, probably through elevation of tissue inhibitor of matrix metalloproteinases-1 expression, and thereby reduced degradation of tight junction proteins. These effects would likely protect cerebrovascular integrity. A2bR agonists as an adjuvant to tPA could be a promising strategy for decreasing the risk of HT during treatment for ischemic stroke.


Asunto(s)
Agonistas del Receptor de Adenosina A2/farmacología , Isquemia Encefálica/tratamiento farmacológico , Hemorragia Cerebral/prevención & control , Fibrinolíticos/toxicidad , Fármacos Neuroprotectores/farmacología , Activador de Tejido Plasminógeno/toxicidad , Aminopiridinas/farmacología , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Hemorragia Cerebral/metabolismo , Hemorragia Cerebral/patología , Modelos Animales de Enfermedad , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Microvasos/efectos de los fármacos , Microvasos/metabolismo , Microvasos/patología , Distribución Aleatoria , Ratas Sprague-Dawley , Receptor de Adenosina A2B/metabolismo , Accidente Cerebrovascular/tratamiento farmacológico , Accidente Cerebrovascular/patología , Inhibidor Tisular de Metaloproteinasa-1/metabolismo
13.
J Neurosci Res ; 95(4): 992-999, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27546887

RESUMEN

Alzheimer's disease (AD) is a complex, progressive neurological disorder characterized by the formation of extracellular amyloid plaques composed of ß-amyloid protein (Aß), the key component in pathogenesis of AD. Peripheral administration of enoxaparin (ENO) reportedly reduces the level of Aß and the amyloid plaques in the cortex of amyloid precursor protein (APP) transgenic mice. However, the exact mechanism of these effects is unclear. Our previous studies indicated that ENO can inhibit APP processing to Aß in primary cortical cells from Tg2576 mice by downregulating BACE1 levels. This study examines whether ENO-induced reduction of amyloid load is due to the decreased APP processing to Aß in Tg2576 mice. Surprisingly, our results indicated that ENO significantly increases the Aß42/Aß40 ratio in cortex and enhances the amyloid plaque load in both cortex and hippocampus, although overall APP processing was not influenced by ENO. Moreover, ENO stimulated the aggregation of both Aß40 and Aß42 in vitro. Although ENO has been reported to improve cognition in vivo and has potential as a therapeutic agent for AD, the results from our study suggest that ENO can exacerbate the amyloid pathology, and the strategy of using ENO for the treatment of AD may require further assessment. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides/metabolismo , Encéfalo/efectos de los fármacos , Enoxaparina/toxicidad , Fibrinolíticos/toxicidad , Placa Amiloide/inducido químicamente , Proteína ADAM10/metabolismo , Enfermedad de Alzheimer/inducido químicamente , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/genética , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Ácido Aspártico Endopeptidasas/metabolismo , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Mutación/genética , Placa Amiloide/genética , Agregado de Proteínas/efectos de los fármacos , Agregado de Proteínas/genética
14.
Arterioscler Thromb Vasc Biol ; 35(3): 637-44, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25593131

RESUMEN

OBJECTIVE: Platelet hyperactivity is associated with vascular disease and contributes to the genesis of thrombotic disorders. ADP plays an important role in platelet activation and activates platelets through 2 G-protein-coupled receptors, the Gq-coupled P2Y1 receptor (P2Y1R), and the Gi-coupled P2Y12 receptor. Although the involvement of the P2Y1R in thrombogenesis is well established, there are no antagonists that are currently available for clinical use. APPROACH AND RESULTS: Our goal is to determine whether a novel antibody targeting the ligand-binding domain, ie, second extracellular loop (EL2) of the P2Y1R (EL2Ab) could inhibit platelet function and protect against thrombogenesis. Our results revealed that the EL2Ab does indeed inhibit ADP-induced platelet aggregation, in a dose-dependent manner. Furthermore, EL2Ab was found to inhibit integrin GPIIb-IIIa activation, dense and α granule secretion, and phosphatidylserine exposure. These inhibitory effects translated into protection against thrombus formation, as evident by a prolonged time for occlusion in a FeCl3-induced thrombosis model, but this was accompanied by a prolonged tail bleeding time. We also observed a dose-dependent displacement of the radiolabeled P2Y1R antagonist [(3)H]MRS2500 from its ligand-binding site by EL2Ab. CONCLUSIONS: Collectively, our findings demonstrate that EL2Ab binds to and exhibits P2Y1R-dependent function-blocking activity in the context of platelets. These results add further evidence for a role of the P2Y1R in thrombosis and validate the concept that targeting it is a relevant alternative or complement to current antiplatelet strategies.


Asunto(s)
Anticuerpos/farmacología , Plaquetas/efectos de los fármacos , Fibrinolíticos/farmacología , Activación Plaquetaria/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/farmacología , Antagonistas del Receptor Purinérgico P2Y/farmacología , Receptores Purinérgicos P2Y1/efectos de los fármacos , Animales , Anticuerpos/metabolismo , Anticuerpos/toxicidad , Sitios de Unión , Unión Competitiva , Plaquetas/metabolismo , Traumatismos de las Arterias Carótidas/sangre , Traumatismos de las Arterias Carótidas/tratamiento farmacológico , Nucleótidos de Desoxiadenina/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Epítopos , Fibrinolíticos/metabolismo , Fibrinolíticos/toxicidad , Hemorragia/inducido químicamente , Hemostasis/efectos de los fármacos , Humanos , Ligandos , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfatidilserinas/sangre , Agregación Plaquetaria/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/metabolismo , Inhibidores de Agregación Plaquetaria/toxicidad , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/antagonistas & inhibidores , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Antagonistas del Receptor Purinérgico P2Y/metabolismo , Antagonistas del Receptor Purinérgico P2Y/toxicidad , Receptores Purinérgicos P2Y1/sangre , Receptores Purinérgicos P2Y1/deficiencia , Receptores Purinérgicos P2Y1/genética , Receptores Purinérgicos P2Y1/inmunología , Receptores Purinérgicos P2Y1/metabolismo , Vesículas Secretoras/efectos de los fármacos , Vesículas Secretoras/metabolismo , Trombosis/sangre , Trombosis/prevención & control , Factores de Tiempo
15.
Blood ; 120(13): 2723-32, 2012 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-22915646

RESUMEN

von Willebrand factor (VWF) is a promising target for developing antithrombotic drugs. The absence of accessible animal models impedes the study of specific human VWF (huVWF) targeting molecules in thrombosis. huVWF is not functional in the mouse because of a lack of interaction between huVWF and murine glycoprotein Ib. Using site-directed mutagenesis, we have replaced single or multiple amino acids in huVWF with their murine counterparts to eliminate species incompatibility. Using hydrodynamic injection, we have expressed the different chimeric VWF constructs into VWF(-/-) mice. Only huVWF with a complete murine A1 domain insertion was able to correct bleeding in vivo and form occlusive thrombi in mesenteric vessels after FeCl(3) treatment. Using this model, we tested the antithrombotic effect of monoclonal antibodies against huVWF, blocking its interaction with collagens (mAbs 203 and 505) or with glycoprotein IIbIIIa (mAb 9). The 3 mAbs inhibited the thrombotic process in arterioles of VWF(-/-) mice expressing huVWFmuA1. Inhibiting VWF-interaction with collagens was more potent, emphasizing the potential of such a target as an antithrombotic tool. Our results validate our murine model as a simple in vivo tool to evaluate anti-huVWF agents.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Modelos Animales de Enfermedad , Hemorragia/prevención & control , Mutación/genética , Adhesividad Plaquetaria/efectos de los fármacos , Trombosis/prevención & control , Factor de von Willebrand/fisiología , Proteína ADAMTS13 , Animales , Cloruros/toxicidad , Colágeno/antagonistas & inhibidores , Colágeno/inmunología , Colágeno/metabolismo , Ensayo de Inmunoadsorción Enzimática , Compuestos Férricos/toxicidad , Fibrinolíticos/toxicidad , Hemorragia/genética , Humanos , Metaloendopeptidasas/sangre , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutagénesis Sitio-Dirigida , Adhesividad Plaquetaria/genética , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/antagonistas & inhibidores , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/inmunología , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Trombosis/inducido químicamente , Trombosis/genética , Factor de von Willebrand/antagonistas & inhibidores
16.
Biometrics ; 70(2): 389-97, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24571185

RESUMEN

While a general goal of early phase clinical studies is to identify an acceptable dose for further investigation, modern dose finding studies and designs are highly specific to individual clinical settings. In addition, as outcome-adaptive dose finding methods often involve complex algorithms, it is crucial to have diagnostic tools to evaluate the plausibility of a method's simulated performance and the adequacy of the algorithm. In this article, we propose a simple technique that provides an upper limit, or a benchmark, of accuracy for dose finding methods for a given design objective. The proposed benchmark is nonparametric optimal in the sense of O'Quigley et al. (2002, Biostatistics 3, 51-56), and is demonstrated by examples to be a practical accuracy upper bound for model-based dose finding methods. We illustrate the implementation of the technique in the context of phase I trials that consider multiple toxicities and phase I/II trials where dosing decisions are based on both toxicity and efficacy, and apply the benchmark to several clinical examples considered in the literature. By comparing the operating characteristics of a dose finding method to that of the benchmark, we can form quick initial assessments of whether the method is adequately calibrated and evaluate its sensitivity to the dose-outcome relationships.


Asunto(s)
Biometría/métodos , Ensayos Clínicos Fase I como Asunto/estadística & datos numéricos , Algoritmos , Antineoplásicos/administración & dosificación , Antineoplásicos/toxicidad , Teorema de Bayes , Benchmarking/estadística & datos numéricos , Ácidos Borónicos/administración & dosificación , Ácidos Borónicos/toxicidad , Bortezomib , Ensayos Clínicos Fase II como Asunto/estadística & datos numéricos , Simulación por Computador , Relación Dosis-Respuesta a Droga , Fibrinolíticos/administración & dosificación , Fibrinolíticos/toxicidad , Humanos , Linfoma/tratamiento farmacológico , Modelos Estadísticos , Pirazinas/administración & dosificación , Pirazinas/toxicidad , Accidente Cerebrovascular/tratamiento farmacológico , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico
17.
Arterioscler Thromb Vasc Biol ; 33(7): 1670-8, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23559626

RESUMEN

OBJECTIVE: During coagulation, factor IX (FIX) is activated by 2 distinct mechanisms mediated by the active proteases of either FVIIa or FXIa. Both coagulation factors may contribute to thrombosis; FXI, however, plays only a limited role in the arrest of bleeding. Therefore, therapeutic targeting of FXI may produce an antithrombotic effect with relatively low hemostatic risk. APPROACH AND RESULTS: We have reported that reducing FXI levels with FXI antisense oligonucleotides produces antithrombotic activity in mice, and that administration of FXI antisense oligonucleotides to primates decreases circulating FXI levels and activity in a dose-dependent and time-dependent manner. Here, we evaluated the relationship between FXI plasma levels and thrombogenicity in an established baboon model of thrombosis and hemostasis. In previous studies with this model, antibody-induced inhibition of FXI produced potent antithrombotic effects. In the present article, antisense oligonucleotides-mediated reduction of FXI plasma levels by ≥ 50% resulted in a demonstrable and sustained antithrombotic effect without an increased risk of bleeding. CONCLUSIONS: These results indicate that reducing FXI levels using antisense oligonucleotides is a promising alternative to direct FXI inhibition, and that targeting FXI may be potentially safer than conventional antithrombotic therapies that can markedly impair primary hemostasis.


Asunto(s)
Coagulación Sanguínea/efectos de los fármacos , Factor XI/metabolismo , Fibrinolíticos/administración & dosificación , Oligonucleótidos Antisentido/administración & dosificación , Trombosis/prevención & control , Animales , Anticuerpos Monoclonales/administración & dosificación , Derivación Arteriovenosa Quirúrgica , Tiempo de Sangría , Colágeno , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Factor XI/antagonistas & inhibidores , Factor XI/genética , Fibrinolíticos/toxicidad , Hemorragia/inducido químicamente , Macaca fascicularis , Oligonucleótidos Antisentido/toxicidad , Papio , Trombina/metabolismo , Trombosis/sangre , Trombosis/etiología , Trombosis/genética , Factores de Tiempo
18.
Drug Chem Toxicol ; 37(1): 8-16, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23834224

RESUMEN

DLBS1033 is a bioactive protein extract containing Lumbricus rubellus and has been known to have antithrombotic/thrombolytic activity. The present study was aimed to assess the safety aspect of DLBS1033 in a preclinical setting, which included observation on toxic signs after acute and repeated administrations, and the drug's effect on prenatal development and drug interaction. In acute toxicity study, a high dose level (16.2 g/kg) of DLBS1033 was well tolerated. In subchronic toxicity study, after the doses of 270, 540 and 1080 mg/kg of DLBS1033 per day, no mortality was observed and other parameters were all observed to be normal. In prenatal developmental toxicity, no observed adverse effect level (NOAEL) of DLBS1033 was observed at a moderate dose (540 mg/kg). Coadministration of DLBS1033 with clopidogrel or aspirin did not cause gastric lesions, except when all three drugs were coadministrated. Taken together, results of the present study suggested that DLBS1033 is safe for long-term administration, with a caution at a high dose used during pregnancy, and can be used in combination with one of the antiplatelet drugs.


Asunto(s)
Desarrollo Fetal/efectos de los fármacos , Fibrinolíticos/toxicidad , Oligoquetos/química , Extractos de Tejidos/toxicidad , Análisis de Varianza , Animales , Aspirina/administración & dosificación , Aspirina/toxicidad , Clopidogrel , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos/métodos , Electroforesis en Gel de Poliacrilamida , Femenino , Ratones , Nivel sin Efectos Adversos Observados , Embarazo , Ratas , Ratas Wistar , Ticlopidina/administración & dosificación , Ticlopidina/análogos & derivados , Ticlopidina/toxicidad , Extractos de Tejidos/aislamiento & purificación , Pruebas de Toxicidad Aguda
19.
J Thromb Haemost ; 22(4): 1016-1023, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38142847

RESUMEN

BACKGROUND: Antithrombotic medications carry an inherent risk of bleeding, which may be exacerbated when anticoagulant and antiplatelet therapeutics are combined. Prior studies have shown different effects of antiplatelet vs anticoagulant drugs on the structure and function of hemostatic plugs in vivo. OBJECTIVES: We examined whether dual antithrombotic treatment consisting of combined antiplatelet and anticoagulant therapeutics alters hemostatic plug structure and function differently from treatment with either therapeutic alone. METHODS: Mice were treated with the P2Y12 antagonist clopidogrel and the factor Xa inhibitor rivaroxaban across a range of doses, either alone or in combination. The hemostatic response was assessed using a mouse jugular vein puncture injury model. Platelet accumulation and fibrin deposition were evaluated using quantitative multiphoton fluorescence microscopy, and bleeding times were recorded. RESULTS: Mice treated with clopidogrel alone exhibited a decrease in platelet accumulation at the site of injury, with prolonged bleeding times only at the highest doses of clopidogrel used. Mice treated with rivaroxaban alone instead showed a reduction in fibrin deposition with no impact on bleeding. Mice treated with both clopidogrel and rivaroxaban exhibited platelet and fibrin accumulation that was similar to that with either drug given alone; however, dual antithrombotic therapy resulted in impaired hemostasis at doses that had no impact on bleeding when given in isolation. CONCLUSION: Combined administration of antiplatelet and anticoagulant therapeutics exacerbates bleeding as compared to that with either drug alone, potentially via combined loss of both adenosine 5'-diphosphate- and thrombin-mediated platelet activation. These findings enhance our understanding of the bleeding risk associated with dual antithrombotic therapy.


Asunto(s)
Hemostáticos , Inhibidores de Agregación Plaquetaria , Humanos , Inhibidores de Agregación Plaquetaria/farmacología , Fibrinolíticos/toxicidad , Clopidogrel , Rivaroxabán , Aspirina , Hemostasis , Anticoagulantes , Hemorragia/inducido químicamente , Hemorragia/tratamiento farmacológico , Fibrina
20.
J Neurosci Res ; 90(11): 2086-93, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22791305

RESUMEN

Detection and protection of the neurovascular unit (NVU) are essential for treatment of acute stroke patients, especially the use of tissue plasminogen activator (tPA). In the present study, we conducted in vivo and ex vivo optical imaging for detecting activation of matrix metalloproteinases (MMPs) and evaluated the protective effect of intracerebral transplantation of bone marrow stromal cells (BMSCs) obtained from green fluorescent protein (GFP) transgenic (Tg) mice on the NVU in tPA-mediated brain damage after transient middle cerebral artery occlusion (tMCAO) in mice. Compared with the tMCAO group, the tMCAO plus BMSC group showed significant reductions of in vivo and ex vivo fluorescent signals for MMPs at 48 hr after tMCAO, with a partial colocalization of BMSC-GFP signals. Intracerebrally transplanted BMSCs ameliorated MMP-9 activation by immunohistochemistry and Western blot with differentiation into microglial and astroglial cells. Double-immunofluorescence study revealed improved NVU disruption in the tMCAO plus BMSC group. The present study suggests that intracerebral BMSC transplantation reduced MMP activation and subsequent NVU disruption caused by tPA after tMCAO and that this MMP activation and BMSC effect were detectable with in vivo and ex vivo optical imaging.


Asunto(s)
Trasplante de Médula Ósea , Isquemia Encefálica/terapia , Fibrinolíticos/toxicidad , Trasplante de Células Madre Mesenquimatosas , Activador de Tejido Plasminógeno/toxicidad , Animales , Western Blotting , Isquemia Encefálica/metabolismo , Hemorragia Cerebral/inducido químicamente , Hemorragia Cerebral/terapia , Activación Enzimática/efectos de los fármacos , Técnica del Anticuerpo Fluorescente , Masculino , Metaloproteinasas de la Matriz/biosíntesis , Células Madre Mesenquimatosas , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Imagen Óptica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA