Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Pharmacokinet Pharmacodyn ; 40(3): 401-18, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23197248

RESUMO

The current gap between animal research and clinical development of analgesic drugs presents a challenge for the application of translational PK-PD modeling and simulation. First, animal pain models lack predictive and construct validity to accurately reflect human pain etiologies and, secondly, clinical pain is a multidimensional sensory experience that can't always be captured by objective and robust measures. These challenges complicate the use of translational PK-PD modeling to project PK-PD data generated in preclinical species to a plausible range of clinical doses. To date only a few drug targets identified in animal studies have shown to be successful in the clinic. PK-PD modeling of biomarkers collected during the early phase of clinical development can bridge animal and clinical pain research. For drugs with novel mechanism of actions understanding of the target pharmacology is essential in order to increase the success of clinical development. There is a specific interest in the application of human pain models that can mimic different aspects of acute/chronic pain symptoms and serves as link between animal and clinical pain research. In early clinical development the main objective of PK-PD modeling is to characterize the relationship between target site binding and downstream biomarkers that have a potential link to the clinical endpoint (e.g. readouts from the human pain models) so as to facilitate the selection of doses for proof of concept studies. In patient studies, the role of PK-PD modeling and simulation is to characterize and confirm patient populations in terms of responder profiles with the aim to find the right dose for the right patient.


Assuntos
Analgésicos , Modelos Biológicos , Dor/tratamento farmacológico , Analgésicos/farmacocinética , Analgésicos/farmacologia , Analgésicos/uso terapêutico , Analgésicos Opioides/farmacocinética , Analgésicos Opioides/farmacologia , Analgésicos Opioides/uso terapêutico , Animais , Simulação por Computador , Humanos , Dor/sangue , Distribuição Tecidual , Resultado do Tratamento
2.
J Pharmacol Sci ; 114(1): 119-22, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20710117

RESUMO

To assess the pharmacological profile of AS1892802, a novel and selective Rho kinase (ROCK) inhibitor, we examined the effects of repeated dosing with AS1892802 on models of monoiodoacetate-induced arthritis and streptozotocin-induced neuropathy. Although single dosing of AS1892802 exerted a short-acting, moderate analgesic effect, repeated dosing exhibited a long-lasting and more potent analgesic effect in both models. Furthermore, the analgesic effect was sustained for seven days after the last administration. These results suggest that peripheral ROCK plays a crucial role in chronic pain maintenance and that AS1892802 may be useful in treating chronic pain.


Assuntos
Modelos Animais de Doenças , Medição da Dor/efeitos dos fármacos , Dor/tratamento farmacológico , Compostos de Fenilureia/administração & dosagem , Piridinas/administração & dosagem , Quinases Associadas a rho/antagonistas & inibidores , Analgésicos/administração & dosagem , Animais , Doença Crônica , Preparações de Ação Retardada , Masculino , Dor/enzimologia , Medição da Dor/métodos , Ratos , Ratos Sprague-Dawley , Quinases Associadas a rho/metabolismo
3.
Brain Res ; 1254: 99-108, 2009 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-19101526

RESUMO

The aim of this study was to evaluate the effect of tacrolimus on recombinant tissue-plasminogen activator (rt-PA)-induced hemorrhagic transformation, and to characterize its suppressive action for hemorrhage. Thrombotic occlusion of the middle cerebral artery (MCA) was induced by photochemical reaction in spontaneously hypertensive rats, and hemorrhagic scores and brain damage were measured 24 h after MCA occlusion. Administration of rt-PA 3 h after MCA occlusion significantly worsened spontaneous hemorrhagic changes and tended to aggravate brain damage. Hematoma was observed in 7 of 15 rats treated with rt-PA, and 0 of 15 rats in the control group. Tacrolimus alone administered intravenously 3 h after MCA occlusion did not produce any hemorrhagic changes. The combined treatment of tacrolimus followed by rt-PA significantly decreased the incidence of hematoma and brain damage in comparison with that of the rt-PA treated group. Permeability of the blood-brain-barrier (BBB) detected by extravasations of Evans blue was investigated 6 h after MCA occlusion, as was the integrity of microvascular endothelial cells as determined by immunohistochemical assessment of the prevalence of platelet endothelial cell adhesion molecule-1 (PECAM-1/CD31). Combined treatment of rt-PA with tacrolimus reduced the rt-PA-induced extravasation of Evans blue and preserved CD31-positive cells in the ischemic hemisphere. Thus, tacrolimus was able to reduce the rt-PA-induced hemorrhagic transformation, which might be due to the protective effects on cerebral microvascular endothelial cells after thrombotic cerebral ischemia during the acute phase of cerebral ischemia. In conclusion, the combination of rt-PA with tacrolimus may be useful for decreasing the risk of thrombolytic therapy.


Assuntos
Encéfalo/efeitos dos fármacos , Encéfalo/fisiopatologia , Infarto da Artéria Cerebral Média/tratamento farmacológico , Tacrolimo/farmacologia , Ativador de Plasminogênio Tecidual/farmacologia , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Células Endoteliais/metabolismo , Azul Evans , Imuno-Histoquímica , Imunossupressores/farmacologia , Infarto da Artéria Cerebral Média/fisiopatologia , Masculino , Microvasos/metabolismo , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Ratos , Ratos Endogâmicos SHR , Proteínas Recombinantes/farmacologia
4.
Brain Res ; 1149: 181-90, 2007 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-17391653

RESUMO

The immunosuppressant cyclosporin A (CsA) has been shown to exert potent neuroprotective effects, possibly via the inhibition of calcineurin and mitochondrial permeability transition pore formation. Here, we investigated the neuroprotective profile of a novel derivative of CsA, FR901459, by evaluating its effects against in vitro mitochondrial damage and in vivo brain damage in transient global or focal cerebral ischemia models, in comparison with those of CsA. Efficacy of calcineurin inhibition was estimated from its immunosuppressive effect on the mixed lymphocyte reaction. Results showed that the immunosuppressive effect of FR901459 was approximately 7-fold less potent than that of CsA. In contrast, FR901459 suppressed Ca(2+)-induced mitochondrial swelling measured in isolated liver mitochondria with greater potency than CsA. Further, FR901459 showed approximately 30-fold greater neuroprotective potency than CsA against neuronal cell damage induced by thapsigargin in SH-SY5Y cells. In a transient global cerebral ischemia model in gerbils, FR901459 showed the dose-dependent suppression of neuronal cell death, while FR901459 was less efficacious than CsA. In a rat transient focal ischemia model, FR901459 tended to reduce brain damage on both intravenous injection as well as intracerebroventricular infusion, but with less efficacy than CsA which significantly reduced the damage. These findings suggest that FR901459 exerts a potent neuroprotective effect by inhibiting mitochondrial damage in vitro, but that in in vivo transient cerebral ischemia, its immunosuppressive component which possibly acts via the inhibition of calcineurin may play a more important role in attenuating brain damage than its inhibitory effect against mitochondrial damage.


Assuntos
Ciclosporina/farmacologia , Hipóxia Encefálica/prevenção & controle , Ataque Isquêmico Transitório/tratamento farmacológico , Mitocôndrias/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Animais , Linhagem Celular Tumoral , Gerbillinae , Humanos , Hipóxia Encefálica/etiologia , Técnicas In Vitro , Ataque Isquêmico Transitório/complicações , Teste de Cultura Mista de Linfócitos , Mitocôndrias/patologia , Ratos
5.
Biol Pharm Bull ; 30(2): 313-7, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17268072

RESUMO

We investigated the neuroprotective effect of tacrolimus (FK506) on the ischemia-reperfusion injury caused by transient focal brain ischemia induced by middle cerebral artery (MCA) occlusion for 60 min in rats. Neuronal damage visualized as a decrease of MAP2 immunoreactivity was observed in the cerebral cortex at 9 h after MCA occlusion and further expanded at 24 h. Hypoxic areas visualized with an immunohistochemical reaction for 2-nitroimidazole, a hypoxia marker (hypoxyprobe-1), and accumulation of granulocytes and platelets were also observed at 9 h and 24 h after MCA occlusion. Tacrolimus (1 mg/kg, i.v.), administered immediately after MCA occlusion, attenuated cortical damage and decreased the hypoxyprobe-1 positive area, as well as the number of granulocytes and platelets at 24 h after MCA occlusion. Immunohistochemical analysis showed that tacrolimus reduced the number of blood vessels positively stained for ICAM-1, E-selectin and P-selection. These results suggested that tacrolimus limited attachment of granulocytes and platelets to blood vessels by inhibiting the expression of adhesion molecules and protected neuronal tissue from hypoxic insults.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Infarto da Artéria Cerebral Média/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Tacrolimo/farmacologia , Animais , Plaquetas/efeitos dos fármacos , Plaquetas/fisiologia , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Adesão Celular/efeitos dos fármacos , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Granulócitos/efeitos dos fármacos , Granulócitos/fisiologia , Infarto da Artéria Cerebral Média/metabolismo , Infarto da Artéria Cerebral Média/patologia , Molécula 1 de Adesão Intercelular/metabolismo , Masculino , Proteínas Associadas aos Microtúbulos/metabolismo , Nitroimidazóis/metabolismo , Ratos , Ratos Wistar
6.
Exp Neurol ; 204(1): 138-46, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17169359

RESUMO

Tacrolimus (FK506), an immunosuppressive drug, has been shown to exert a potent neuroprotective activity when administered immediately after occlusion of the middle cerebral artery (MCA) in a nonhuman primate model of stroke. Here, we assessed the neuroprotective efficacy of tacrolimus with delayed treatment using the same model and compared with that of recombinant tissue plasminogen activator (rt-PA). Ischemic insult was induced by photochemically induced thrombotic occlusion of MCA in cynomolgus monkeys, and tacrolimus (0.2 mg/kg) and/or rt-PA (1.0 mg/kg) was intravenously administered 2 h after MCA occlusion. In another experiment, tacrolimus (0.1 mg/kg) was administered 4 h after MCA occlusion. Neurological deficits were monitored for 28 days after the ischemic insult and cerebral infarct volumes were measured with brain slices. With drug administration 2 h after the ischemic insult, tacrolimus significantly reduced neurological deficits and infarct volumes in the cerebral cortex without affecting the recanalization pattern in the MCA, however, rt-PA did not significantly improve neurological deficits or infarct volumes, even though it increased the recanalization rate of the occluded MCA. Combined treatment with tacrolimus and rt-PA exerted additional protection. Administration of tacrolimus 4 h after the ischemic insult still showed significant amelioration of neurological deficits. These results suggested that tacrolimus had a wider therapeutic time window than rt-PA in the nonhuman primate stroke model.


Assuntos
Fibrinolíticos/farmacologia , Fármacos Neuroprotetores/farmacologia , Acidente Vascular Cerebral/patologia , Acidente Vascular Cerebral/fisiopatologia , Tacrolimo/farmacologia , Ativador de Plasminogênio Tecidual/farmacologia , Animais , Encéfalo/patologia , Circulação Cerebrovascular/efeitos dos fármacos , Esquema de Medicação , Combinação de Medicamentos , Injeções Intravenosas , Macaca fascicularis , Masculino , Sistema Nervoso/efeitos dos fármacos , Sistema Nervoso/fisiopatologia , Fármacos Neuroprotetores/administração & dosagem , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/farmacologia , Tacrolimo/administração & dosagem , Ativador de Plasminogênio Tecidual/administração & dosagem
7.
J Vet Med Sci ; 68(8): 803-7, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16953079

RESUMO

Intravascular accumulation of blood cells after brain ischemia-reperfusion can cause obstruction of cerebral blood flow and tissue hypoxia/ischemia as a consequence. In the present study, we examined temporal and topographic changes of tissue hypoxia/ischemia after occlusion of the middle cerebral artery (MCA) for 60 min in rats with immunohistochemical staining for hypoxia (2-nitroimidazole hypoxia marker: hypoxyprobe-1 adducts). Our results showed that tissue hypoxia expressed as positive staining for hypoxyprobe-1 adducts preceded neuronal degeneration. Platelets and granulocytes were detected close to the hypoxyprobe-1 adducts positive area. These results suggested that the hypoxic environment could persist even after reperfusion of MCA, because of vascular obstruction with accumulation of platelets and granulocytes.


Assuntos
Isquemia Encefálica/fisiopatologia , Encéfalo/metabolismo , Traumatismo por Reperfusão/patologia , Animais , Encéfalo/irrigação sanguínea , Encéfalo/patologia , Regulação Enzimológica da Expressão Gênica , Integrina beta3/metabolismo , Masculino , Peroxidase/metabolismo , Ratos , Ratos Wistar , Fatores de Tempo
8.
Exp Neurol ; 199(1): 92-102, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16712840

RESUMO

Adult neurogenesis is modulated by growth factors, physical conditions, and other alterations in the physical microenvironment. We studied the effects of focal ischemia on neurogenesis in the subventricular zone (SVZ), olfactory bulb (OB), and hippocampal dentate gyrus (DG) (known to be persistent neurogenic regions) in the adult non-human primate, the cynomolgus monkey. Three monkeys underwent middle cerebral artery occlusion-induced focal ischemia and were given multiple BrdU injections during the first 2 weeks after ischemia. Twenty-eight days later, the animals were perfused. The number of new neurons (3182 +/- 408/mm3) in the ipsilateral DG of ischemic monkeys was 4.7-fold that in the DG of non-operated monkeys. The number of new neurons (9176 +/- 2295/mm3) in the ipsilateral olfactory bulb of ischemic monkeys was 18.0-fold that in normal olfactory bulb. These observations suggest an increase in the number of new OB neurons, as well as new DG neurons, after focal ischemia in a primate. This substantial increase in new neurons after focal ischemia could result from the enhancement of cell proliferation rather than a change in the rate of cell commitment. Of the three monkeys subjected to ischemia, only one animal possessed a unique progenitor cell type at the most anterior aspect of the ipsilateral SVZ. Within this region, a short migration (approximately 500 microm) of doublecortin-expressing immature neuronal progenitor cells was observed.


Assuntos
Isquemia Encefálica/patologia , Isquemia Encefálica/fisiopatologia , Hipocampo/patologia , Neurônios/fisiologia , Bulbo Olfatório/patologia , Animais , Bromodesoxiuridina/metabolismo , Contagem de Células/métodos , Ventrículos Cerebrais/patologia , Modelos Animais de Doenças , Proteínas do Domínio Duplacortina , Lateralidade Funcional/fisiologia , Imuno-Histoquímica/métodos , Macaca fascicularis , Proteínas Associadas aos Microtúbulos/metabolismo , Exame Neurológico/métodos , Neuropeptídeos/metabolismo , Fosfopiruvato Hidratase/metabolismo , Fatores de Tempo
9.
J Neurotrauma ; 22(11): 1362-73, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16305324

RESUMO

Intracerebral hemorrhage (ICH) is the most serious side effect of antithrombotic agents, especially in cases of cerebrovascular disease. In the present study, we compared the exacerbation of ICH and prolongation of bleeding time (BT) in guinea pigs with recombinant tissue plasminogen activator (rt-PA), heparin, aspirin, and FK419, a novel nonpeptide platelet glycoprotein (GP) IIb/IIIa receptor antagonist. ICH was induced by injection of bacterial collagenase into the caudate nucleus; BT was measured with a Simplate R device. Neither heparin nor aspirin prolonged BT. In contrast, rt-PA at the highest dose used in the study did prolong BT, and FK419 caused a dose-dependent prolongation of BT. Moreover, rt-PA and heparin increased the degree of ICH in a dose-dependent manner, leading to death in more than half of the animals treated with higher doses of these drugs. These findings show that the prohemorrhagic mechanisms underlying the prolongation of BT differ from those in collagenase-induced ICH, and that the risk of an agent with antithrombotic effects potentiating hemorrhage in the collagenase-induced model of ICH more closely parallels that in stroke patients than does the effect of the agent on BT. The findings also suggest that antiplatelet agents, including FK419, may be safer than thrombolytic or anticoagulant agents for use in patients at risk for ICH, such as those with stroke or cerebral aneurysm.


Assuntos
Aspirina/farmacologia , Fibrinolíticos/farmacologia , Heparina/farmacologia , Hemorragias Intracranianas/induzido quimicamente , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/antagonistas & inibidores , Ativador de Plasminogênio Tecidual/farmacologia , Animais , Tempo de Sangramento , Colagenases/farmacologia , Relação Dose-Resposta a Droga , Cobaias , Hemorragias Intracranianas/mortalidade , Masculino , Piperidinas/farmacologia , Agregação Plaquetária/efeitos dos fármacos , Propionatos/farmacologia , Proteínas Recombinantes/farmacologia , Tempo de Coagulação do Sangue Total
10.
J Neurosci Methods ; 146(1): 106-15, 2005 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-15935227

RESUMO

In an attempt to establish a thrombotic middle cerebral artery (MCA) occlusion model using cynomolgus monkeys, we measured the blood flow in the main MCA tract and cerebral cortex, brain damage, and neurological deficits, and compared them with those of mechanical MCA occlusion model. Thrombotic occlusion was induced photochemically by green light application on the MCA following rose bengal treatment; mechanical occlusion was induced by MCA clipping for 3h. Patency of the main MCA tract showed two patterns in the thrombotic model: permanent occlusion or cyclical flow reduction (CFR). Regional cerebral blood flow (rCBF) decreased during occlusion followed by post-ischemic hyperperfusion in the clipping model, whereas rCBF reduction expanded time-dependently in the thrombotic occlusion model. Brain infarction and neurological scores in the thrombotic occlusion model were significantly larger than those in the clipping occlusion model. In histological assessment, microthrombi containing myeloperoxidase- and fibrinogen-positive cells were observed in the cortex following the thrombotic but not clipping occlusion. These results collectively suggest that this thrombotic MCA occlusion model, because it shows impairment of cerebral microcirculation, could provide a vital platform for understanding progressive ischemia as well as for evaluating potential therapeutic drugs.


Assuntos
Infarto Cerebral/diagnóstico por imagem , Transtornos Cerebrovasculares/diagnóstico por imagem , Modelos Animais de Doenças , Infarto da Artéria Cerebral Média/diagnóstico por imagem , Trombose Intracraniana/diagnóstico por imagem , Artéria Cerebral Média/diagnóstico por imagem , Animais , Biomarcadores/metabolismo , Córtex Cerebral/irrigação sanguínea , Córtex Cerebral/diagnóstico por imagem , Córtex Cerebral/fisiopatologia , Infarto Cerebral/fisiopatologia , Circulação Cerebrovascular/fisiologia , Transtornos Cerebrovasculares/fisiopatologia , Progressão da Doença , Fibrinogênio/metabolismo , Infarto da Artéria Cerebral Média/fisiopatologia , Trombose Intracraniana/fisiopatologia , Macaca fascicularis , Masculino , Artéria Cerebral Média/fisiopatologia , Peroxidase/metabolismo , Estimulação Luminosa/métodos , Tomografia por Emissão de Pósitrons , Rosa Bengala/efeitos da radiação , Instrumentos Cirúrgicos
11.
Brain Res ; 1014(1-2): 120-30, 2004 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-15212998

RESUMO

While the immunosuppressant tacrolimus (FK506) is known to be neuroprotective following cerebral ischemia, the mechanisms underlying its neuroprotective properties are not fully understood. To determine the mode of action by which tacrolimus ameliorates neurodegeneration after transient focal ischemia, we therefore evaluated the effect of tacrolimus on DNA damage, release of cytochrome c, activation of microglia and infiltration of neutrophils following a 60-min occlusion of the middle cerebral artery (MCA) in rats. In this model, cortical brain damage gradually expanded until 24 h after reperfusion, whereas brain damage in the caudate putamen was fully developed within 5 h. Tacrolimus (1 mg/kg) administered immediately after MCA occlusion significantly reduced ischemic damage in the cerebral cortex, but not in the caudate putamen. Tacrolimus decreased both apoptotic and necrotic cell death at 24 h and reduced the number of cytochrome c immunoreactive cells at 8 h after reperfusion in the ischemic penumbra in the cerebral cortex. In contrast, tacrolimus did not show significant neuroprotection for necrotic cell death and reduction of cytochrome c immunoreactive cells in the caudate putamen. Tacrolimus also significantly decreased microglial activation at 8 h and inflammatory markers (cytokine-induced neutrophil chemoattractant and myeloperoxidase [MPO] activity) at 24 h after reperfusion in the ischemic cortex but not in the caudate putamen. These results collectively suggest that tacrolimus ameliorates the gradually expanded brain damage by inhibiting both apoptotic and necrotic cell death, as well as suppressing inflammatory reactions.


Assuntos
Ataque Isquêmico Transitório/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Tacrolimo/farmacologia , Animais , Apoptose/efeitos dos fármacos , Núcleo Caudado/patologia , Quimiocinas CXC/metabolismo , Citocromos c/metabolismo , Imuno-Histoquímica , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Ataque Isquêmico Transitório/metabolismo , Ataque Isquêmico Transitório/patologia , Masculino , Microglia/metabolismo , Peroxidase/metabolismo , Ratos , Ratos Wistar
12.
J Cereb Blood Flow Metab ; 23(10): 1183-94, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14526229

RESUMO

Tacrolimus (FK506), an immunosuppressive drug, is known to have potent neuroprotective activity and attenuate cerebral infarction in experimental models of stroke. Here we assess the neuroprotective efficacy of tacrolimus in a nonhuman primate model of stroke, photochemically induced thrombotic occlusion of the middle cerebral artery (MCA) in cynomolgus monkeys. In the first experiment, tacrolimus (0.01, 0.032, or 0.1 mg/kg) was intravenously administered immediately after MCA occlusion, and neurologic deficits and cerebral infarction volumes were assessed 24 hours after the ischemic insult. Tacrolimus dose-dependently reduced neurologic deficits and infarction volume in the cerebral cortex, with statistically significant amelioration of neurologic deficits at 0.032 and 0.1 mg/kg and significant reduction of infarction at 0.1 mg/kg. In the second experiment, the long-term efficacy of tacrolimus on neurologic deficits and cerebral infarction was assessed. Vehicle-treated monkeys exhibited persistent and severe deficits in motor and sensory function for up to 28 days. A single intravenous bolus injection of tacrolimus (0.1 or 0.2 mg/kg) produced long-lasting amelioration of neurologic deficits and significant reduction of infarction volume. In conclusion, we have provided compelling evidence that a single dose of tacrolimus not only reduces brain infarction but also ameliorates long-term neurologic deficits in a nonhuman primate model of stroke, strengthening the view that tacrolimus might be beneficial in treating stroke patients.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Imunossupressores/farmacocinética , Fármacos Neuroprotetores/farmacocinética , Tacrolimo/farmacocinética , Animais , Pressão Sanguínea/efeitos dos fármacos , Isquemia Encefálica/patologia , Isquemia Encefálica/fisiopatologia , Circulação Cerebrovascular/efeitos dos fármacos , Modelos Animais de Doenças , Frequência Cardíaca/efeitos dos fármacos , Imunossupressores/sangue , Infarto da Artéria Cerebral Média/tratamento farmacológico , Infarto da Artéria Cerebral Média/patologia , Infarto da Artéria Cerebral Média/fisiopatologia , Macaca fascicularis , Masculino , Exame Neurológico , Fármacos Neuroprotetores/sangue , Recuperação de Função Fisiológica/efeitos dos fármacos , Tacrolimo/sangue
13.
Brain Res ; 965(1-2): 137-45, 2003 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-12591130

RESUMO

Tacrolimus (FK506), a potent immunosuppressive drug, is effective in attenuating brain infarction after cerebral ischemia. However, there has been no report characterizing the neuroprotective action and therapeutic time window of tacrolimus systematically using different types of stroke models and extended observation periods. Therefore, we evaluated the neuroprotective effect of tacrolimus in three different animal models of cerebral ischemia: transient and permanent focal ischemia in rats and transient global ischemia in gerbils. Tacrolimus at doses higher than 0.1 mg/kg (i.v.) produced a statistically significant reduction in ischemic brain damage following permanent and transient focal ischemia in rats when administered immediately after the onset of ischemia. Tacrolimus (1 mg/kg, i.v.) demonstrated similar neuroprotective activity even after delayed administration (2 h after permanent or 1 h after transient focal ischemia). The neuroprotective effect of tacrolimus was still present 2 weeks after transient focal ischemia and 1 week after permanent focal ischemia. After transient global ischemia in gerbils, tacrolimus (1 mg/kg, i.v.) given immediately after reperfusion also produced long-lasting neuroprotective effects with a protective time-window of 1-2 h. Taken together, the results clearly indicate that tacrolimus exerts potent, long-term neuroprotective effects with a favorable therapeutic time-window, regardless of the model of cerebral ischemia. These results strengthen the notion that tacrolimus might be of clinical value for the treatment of acute stroke.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Tacrolimo/uso terapêutico , Animais , Isquemia Encefálica/patologia , Contagem de Células/métodos , Relação Dose-Resposta a Droga , Masculino , Neurônios/efeitos dos fármacos , Neurônios/patologia , Fármacos Neuroprotetores/farmacologia , Ratos , Ratos Sprague-Dawley , Tacrolimo/farmacologia , Fatores de Tempo
14.
J Cereb Blood Flow Metab ; 22(10): 1205-11, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12368659

RESUMO

The authors evaluated the therapeutic efficacy of tacrolimus (FK506), administered alone or in combination with recombinant tissue plasminogen activator (t-PA), on brain infarction following thrombotic middle cerebral artery (MCA) occlusion. Thrombotic occlusion of the MCA was induced by a photochemical reaction between rose bengal and green light in Sprague-Dawley rats, and the volume of ischemic brain damage was determined 24 hours later. Intravenous administration of tacrolimus or t-PA dose-dependently reduced the volume of ischemic brain infarction, whether administered immediately or 1 hour after MCA occlusion. When tacrolimus or t-PA was administered 2 hours after MCA occlusion, each drug showed a tendency to reduce ischemic brain damage. However, combined treatment with both drugs resulted in a significant reduction in ischemic brain damage. On administration 3 hours after MCA occlusion, tacrolimus alone showed no effect, and t-PA tended to worsen ischemic brain damage. However, the combined treatment with both drugs not only ameliorated the worsening trend seen with t-PA alone, but also tended to reduce ischemic brain damage. In conclusion, tacrolimus, used in combination with t-PA, augmented therapeutic efficacy on brain damage associated with focal ischemia and extended the therapeutic time window compared to single-drug treatments.


Assuntos
Circulação Cerebrovascular/efeitos dos fármacos , Trombose Intracraniana/tratamento farmacológico , Ataque Isquêmico Transitório/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Tacrolimo/uso terapêutico , Ativador de Plasminogênio Tecidual/uso terapêutico , Animais , Velocidade do Fluxo Sanguíneo/efeitos dos fármacos , Circulação Cerebrovascular/fisiologia , Modelos Animais de Doenças , Quimioterapia Combinada , Trombose Intracraniana/patologia , Ataque Isquêmico Transitório/patologia , Masculino , Artéria Cerebral Média/efeitos dos fármacos , Artéria Cerebral Média/patologia , Fármacos Neuroprotetores/administração & dosagem , Ratos , Ratos Sprague-Dawley , Tacrolimo/administração & dosagem , Fatores de Tempo , Ativador de Plasminogênio Tecidual/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...