Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Exp Anim ; 70(3): 344-354, 2021 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-33731549

RESUMO

In the current study, we established a novel murine ischemic brain damage model using a photochemical reaction to evaluate the recovery of neurological dysfunction and brain repair reactions. In this model, reproducible damage was induced in the frontal lobe of the cortex, which was accompanied by neurological dysfunction. Sequential changes in damage size, microglial accumulation, astrocyte activation, and neurological dysfunction were studied in C57BL/6J and BALB/c mouse strains. Although the initial size of damage was comparable in both strains, the extent of damage was later reduced to a greater extent in C57BL/6J mice than that in BALB/c mice. In addition, C57BL/6J mice showed later edema clearance until day 7, less microglial accumulation, and relatively more astrocyte activation on day 7. Neurologic dysfunction was evaluated by three behavioral tests: the von Frey test, the balance beam test, and the tail suspension test. The behavioral abnormalities evaluated by these tests were remarkable following the induction of damage and recovered by day 21 in both strains. However, the abnormalities were more prominent and the recovery was later in C57BL/6J mice. These findings demonstrate that our novel ischemic stroke model is useful for evaluating brain repair reactions and the recovery of neurological dysfunction in mice with different genetic backgrounds. In addition, we found that both the brain repair reactions and the recovery of neurological dysfunction after comparable ischemic brain damage varied between strains; in that, they both occurred later in C57BL/6J mice.


Assuntos
Isquemia Encefálica/fisiopatologia , AVC Isquêmico/fisiopatologia , Animais , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Especificidade da Espécie
2.
Exp Anim ; 67(4): 479-486, 2018 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-29806621

RESUMO

It is observed that the increase in blood-brain barrier (BBB) permeability (BBBP) is associated with ischemic stroke and thought to trigger neuronal damage and deteriorate ischemic infarction, even though there is no experimental proof. Here, we investigated the effect of BBBP increase on brain damage, using a combination of photochemically-induced thrombotic brain damage (PIT-BD) model, a focal brain ischemic model, and transient bilateral carotid artery occlusion model (CAO, a whole brain ischemic model), in mice. In PIT-BD, BBBP increased in the region surrounding the ischemic damage from 4 h till 24 h with a peak at 8 h. On day 4, the damaged did not expand to the region with BBBP increase in mice with PIT-BD alone or with 30 min CAO at 1 h before PIT-BD, but expanded in mice with 30 min CAO at 3.5 h after PIT-BD. This expansion was paralleled with the increase in the number of apoptotic cells. These findings indicate that increase in BBBP does not cause direct neuronal death, but it facilitates ischemic neuronal loss, which was attributed, at least partially, to acceleration of apoptotic cell death.


Assuntos
Apoptose , Barreira Hematoencefálica/metabolismo , Isquemia Encefálica/etiologia , Isquemia Encefálica/patologia , Encéfalo/citologia , Encéfalo/patologia , Permeabilidade Capilar/fisiologia , Neurônios/patologia , Animais , Modelos Animais de Doenças , Masculino , Camundongos Endogâmicos C57BL
3.
Front Cell Neurosci ; 10: 2, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26834557

RESUMO

Cerebrovascular homeostasis is maintained by the blood-brain barrier (BBB), which forms a mechanical and functional barrier between systemic circulation and the central nervous system (CNS). In patients with ischemic stroke, the recombinant tissue-type plasminogen activator (rt-PA) is used to accelerate recanalization of the occluded vessels. However, rt-PA is associated with a risk of increasing intracranial bleeding (ICB). This effect is thought to be caused by the increase in cerebrovascular permeability though various factors such as ischemic reperfusion injury and the activation of matrix metalloproteinases (MMPs), but the detailed mechanisms are unknown. It was recently found that rt-PA treatment enhances BBB permeability not by disrupting the BBB, but by activating the vascular endothelial growth factor (VEGF) system. The VEGF regulates both the dissociation of endothelial cell (EC) junctions and endothelial endocytosis, and causes a subsequent increase in vessel permeability through the VEGF receptor-2 (VEGFR-2) activation in ECs. Here, we review the possibility that rt-PA increases the penetration of toxic molecules derived from the bloodstream including rt-PA itself, without disrupting the BBB, and contributes to these detrimental processes in the cerebral parenchyma.

4.
PLoS One ; 10(8): e0135836, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26284361

RESUMO

Human dynactin-associated protein (dynAP) is a transmembrane protein that promotes AktSer473 phosphorylation. Here, we report the oncogenic properties of dynAP. In contrast to control NIH3T3 cells expressing LacZ (NIH3T3LacZ), NIH3T3dynAP cells vigorously formed foci in two-dimensional culture, colonies on soft agar, and spheroids in anchorage-deficient three-dimensional culture. NIH3T3dynAP cells injected into nude mice produced tumors with abundant blood vessels and weak cell-cell contacts. Expression of dynAP elevated the level of rictor (an essential subunit of mTORC2) and promoted phosphorylation of FOXO3aSer253. FOXO3a is a transcriptional factor that stimulates expression of pro-apoptotic genes and phosphorylation of FOXO3a abrogates its function, resulting in promoted cell survival. Knockdown of rictor in NIH3T3dynAP cells reduced AktSer473 phosphorylation and formation of foci, colony in soft agar and spheroid, indicating that dynAP-induced activation of the mTORC2/AktSer473 pathway for cell survival contributes to cell transformation. E-cadherin and its mRNA were markedly reduced upon expression of dynAP, giving rise to cells with higher motility, which may be responsible for the weak cell-cell adhesion in tumors. Thus, dynAP could be a new oncoprotein and a target for cancer therapy.


Assuntos
Comunicação Celular , Transformação Celular Neoplásica , Proteínas Associadas aos Microtúbulos/genética , Neoplasias/irrigação sanguínea , Neoplasias/patologia , Neovascularização Patológica/genética , Animais , Complexo Dinactina , Feminino , Humanos , Óperon Lac/genética , Camundongos , Células NIH 3T3 , Neoplasias/genética , Regulação para Cima
5.
J Cereb Blood Flow Metab ; 35(12): 2021-31, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26219596

RESUMO

Recombinant tissue-type plasminogen activator (rt-PA) modulates cerebrovascular permeability and exacerbates brain injury in ischemic stroke, but its mechanisms remain unclear. We studied the involvement of vascular endothelial growth factor (VEGF)-mediated endocytosis in the increase of blood-brain barrier (BBB) permeability potentiated by rt-PA after ischemic stroke. The rt-PA treatment at 4 hours after middle cerebral artery occlusion induced a transient increase in BBB permeability after ischemic stroke in mice, which was suppressed by antagonists of either low-density lipoprotein receptor families (LDLRs) or VEGF receptor-2 (VEGFR-2). In immortalized bEnd.3 endothelial cells, rt-PA treatment upregulated VEGF expression and VEGFR-2 phosphorylation under ischemic conditions in an LDLR-dependent manner. In addition, rt-PA treatment increased endocytosis and transcellular transport in bEnd.3 monolayers under ischemic conditions, which were suppressed by the inhibition of LDLRs, VEGF, or VEGFR-2. The rt-PA treatment also increased the endocytosis of endothelial cells in the ischemic brain region after stroke in mice. These findings indicate that rt-PA increased BBB permeability via induction of VEGF, which at least partially mediates subsequent increase in endothelial endocytosis. Therefore, inhibition of VEGF induction may have beneficial effects after thrombolytic therapy with rt-PA treatment after stroke.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Isquemia Encefálica/patologia , Endocitose/efeitos dos fármacos , Fibrinolíticos/farmacologia , Ativador de Plasminogênio Tecidual/farmacologia , Fator A de Crescimento do Endotélio Vascular/farmacologia , Animais , Isquemia Encefálica/tratamento farmacológico , Linhagem Celular , Células Endoteliais/efeitos dos fármacos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Artéria Cerebral Média/patologia , Receptores de Lipoproteínas/efeitos dos fármacos , Proteínas Recombinantes , Soroalbumina Bovina/metabolismo , Acidente Vascular Cerebral/tratamento farmacológico , Acidente Vascular Cerebral/patologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/efeitos dos fármacos
6.
Life Sci ; 93(2-3): 89-95, 2013 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-23770230

RESUMO

AIMS: We investigated the pathophysiological changes in mice lacking α2-antiplasmin (α2-AP) and plasminogen activator inhibitor type-1 (PAI-1) genes, and elucidated the involvement of these inhibitors for fibrinolysis in immune response. MAIN METHODS: The pathophysiological changes induced by a lack of both α2-AP and PAI-1 were investigated using double knockout (KO) mice. The lung, liver, kidney and spleen tissues from α2-AP/PAI-1-double KO mice were compared with those from wild-type (WT) mice. Furthermore, the bone marrow cells from α2-AP/PAI-1-double KO mice were transplanted into 10-Gy X ray irradiated WT mice, and then the effects of the transplantation were studied. KEY FINDINGS: Plasma IgE levels in the α2-AP/PAI-1-double KO mice increased with age and exceeded 1000 ng/mL after 6 months of age. The plasma cells that produced IgE were detected in perivascular assembled lymphocytes. In the α2-AP/PAI-1-double KO mice, perivascular lymphocyte infiltration was observed in the lung, liver, and kidneys and peribronchial lymphocyte infiltration was present in the lung. When the bone marrow cells from α2-AP/PAI-1-double KO mice were transplanted into 10-Gy X ray irradiated WT mice, the phenotypes of the recipients were similar to those of α2-AP/PAI-1-double KO mice. SIGNIFICANCE: The simultaneous expression of both the α2-AP and PAI-1 genes contributes to the maintenance of immunological functions that are related to IgE. Moreover, it is suggested that both α2-AP and PAI-1 are involved in the recruitment of lymphocytes in the peripheral tissues.


Assuntos
Imunoglobulina E/metabolismo , Inibidor 1 de Ativador de Plasminogênio/genética , alfa 2-Antiplasmina/genética , Fatores Etários , Animais , Transplante de Medula Óssea , Citocinas/sangue , Fibrinólise , Imunoglobulina E/genética , Rim/metabolismo , Rim/patologia , Fígado/metabolismo , Fígado/patologia , Linfócitos/fisiologia , Camundongos , Camundongos Knockout , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Baço/metabolismo , Baço/patologia , alfa 2-Antiplasmina/metabolismo
7.
PLoS One ; 7(2): e32342, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22384223

RESUMO

BACKGROUND: There is an increasing need for animal disease models for pathophysiological research and efficient drug screening. However, one of the technical barriers to the effective use of the models is the difficulty of non-invasive and sequential monitoring of the same animals. Micro-CT is a powerful tool for serial diagnostic imaging of animal models. However, soft tissue contrast resolution, particularly in the brain, is insufficient for detailed analysis, unlike the current applications of CT in the clinical arena. We address the soft tissue contrast resolution issue in this report. METHODOLOGY: We performed contrast-enhanced CT (CECT) on mouse models of experimental cerebral infarction and hepatic ischemia. Pathological changes in each lesion were quantified for two weeks by measuring the lesion volume or the ratio of high attenuation area (%HAA), indicative of increased vascular permeability. We also compared brain images of stroke rats and ischemic mice acquired with micro-CT to those acquired with 11.7-T micro-MRI. Histopathological analysis was performed to confirm the diagnosis by CECT. PRINCIPAL FINDINGS: In the models of cerebral infarction, vascular permeability was increased from three days through one week after surgical initiation, which was also confirmed by Evans blue dye leakage. Measurement of volume and %HAA of the liver lesions demonstrated differences in the recovery process between mice with distinct genetic backgrounds. Comparison of CT and MR images acquired from the same stroke rats or ischemic mice indicated that accuracy of volumetric measurement, as well as spatial and contrast resolutions of CT images, was comparable to that obtained with MRI. The imaging results were also consistent with the histological data. CONCLUSIONS: This study demonstrates that the CECT scanning method is useful in rodents for both quantitative and qualitative evaluations of pathologic lesions in tissues/organs including the brain, and is also suitable for longitudinal observation of the same animals.


Assuntos
Isquemia Encefálica/patologia , Encéfalo/patologia , Isquemia/patologia , Fígado/patologia , Acidente Vascular Cerebral/patologia , Microtomografia por Raio-X/métodos , Animais , Pressão Sanguínea , Meios de Contraste/farmacologia , Modelos Animais de Doenças , Encefalopatia Hipertensiva , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Permeabilidade , Ratos , Fatores de Tempo
8.
Neuroreport ; 22(9): 424-7, 2011 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-21602651

RESUMO

To study spatiotemporal differences in vascular permeability, we histologically analysed tracer extravasation, neovessels and reactive astrocytes in a mouse ischaemic brain damage model. On day 1 after damage induction, the extravasation was not associated with the distribution of neovessels or reactive astrocytes. On day 7, the extravasation was limited within the infarct region in which neovessels, but not reactive astrocytes, were observed. However, the extravasation was not observed at peri-infarct region in which both neovessels and reactive astrocytes were observed, suggesting that neovessels had high permeability and reactive astrocytes prevented the extravasation from neovessels. Furthermore, the extravasation was denser in the regions near the surface than in those further in the infarct region, suggesting a spatial heterogeneity in neovascular permeability.


Assuntos
Barreira Hematoencefálica/patologia , Isquemia Encefálica/patologia , Permeabilidade Capilar , Neovascularização Fisiológica , Animais , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL
9.
J Pharmacol Sci ; 116(1): 25-9, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21498957

RESUMO

Previous studies have shown that the risk of intracranial hemorrhage (ICH) associated with the treatment of ischemic stroke is mainly attributable to antithrombotic agents. On the basis of clinical trials, only tissue-type plasminogen activator (t-PA) has been approved for treating acute ischemic strokes, but delayed treatment with t-PA is associated with the risk of cerebral hemorrhagic transformation of ischemic stroke. t-PA converts plasminogen to plasmin, which participates primarily in clot lysis via fibrin degradation and, to some extent, in tissue remodeling via degradation of various extracellular matrix proteins, either directly or via activation of matrix metalloproteinases (MMPs). MMPs mediate the pathogenesis of ischemic-stroke-associated ICH by causing the disruption of vasculature. In particular, the binding of t-PA with one of its receptors leads to the activation of low-density lipoprotein receptor-related protein (LRP), which in turn results in the release of MMP-3 by endothelial cells. LRP production is reported to be upregulated in endothelial cells exposed to ischemia, and elevated LRP levels have been implicated in the increased ICH risk associated with delayed t-PA treatment. This implies that the t-PA / LRP / MMP-3 pathway may be a suitable target for developing strategies to improve the therapeutic efficacy of t-PA in acute ischemic stroke.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Endotélio Vascular/fisiopatologia , Fibrinolíticos/efeitos adversos , Hemorragias Intracranianas/prevenção & controle , Acidente Vascular Cerebral/tratamento farmacológico , Terapia Trombolítica , Ativador de Plasminogênio Tecidual/efeitos adversos , Animais , Isquemia Encefálica/fisiopatologia , Endotélio Vascular/efeitos dos fármacos , Fibrinolisina/metabolismo , Humanos , Hemorragias Intracranianas/induzido quimicamente , Proteínas Relacionadas a Receptor de LDL/metabolismo , Metaloproteinases da Matriz/metabolismo , Terapia de Alvo Molecular , Traumatismo por Reperfusão/induzido quimicamente , Traumatismo por Reperfusão/prevenção & controle , Acidente Vascular Cerebral/fisiopatologia
10.
J Thromb Thrombolysis ; 32(2): 195-200, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21258842

RESUMO

Fibrinolytic system impairment contributes to the development of thrombotic disease such as cardiovascular disease and stroke. Therefore, an agent that increases fibrinolytic activity may be useful for the prevention of these diseases. In this study, to explore novel profibrinolytic agents, we examined the profibrinolytic effect of Enzamin, an extract of metabolic products from Bacillus subtilis AK and Lactobacillus in vitro and in vivo. Enzamin directly enhanced plasmin activity generated by tissue-type plasminogen activator (t-PA) by twofold but not by urokinase-type plasminogen activator (u-PA) in vitro, which was measured employing both the chromogenic substrate H-D: -Val-Leu-Lys-pNA (S-2251) and fibrin plate. Enzamin also increased plasmin activity generated by t-PA in the cell lysate and culture medium of endothelial cells, measured by fibrin zymography. Furthermore, the oral administration of a 1% concentration of Enzamin increased plasmin activity generated by t-PA by 1.7-fold but not by u-PA in the euglobulin fraction of mouse plasma. In conclusion, Enzamin has a unique ability to enhance the fibrinolytic activity through an increase in endogenous plasmin activity generated by t-PA released from endothelial cells, and may be a beneficial supplement for the prevention of thrombotic episodes.


Assuntos
Bacillus subtilis/química , Misturas Complexas/farmacologia , Fibrinólise/efeitos dos fármacos , Fibrinolíticos/farmacologia , Fibrinolíticos/farmacocinética , Lactobacillus/química , Animais , Testes de Coagulação Sanguínea , Linhagem Celular , Misturas Complexas/química , Avaliação Pré-Clínica de Medicamentos , Fibrinolisina/metabolismo , Fibrinolíticos/química , Camundongos , Trombose/sangue , Trombose/tratamento farmacológico , Ativador de Plasminogênio Tecidual/sangue
11.
Cell Tissue Res ; 343(2): 303-17, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21190118

RESUMO

Although the tissue plasminogen activator/plasminogen system contributes to numerous brain functions, such as learning, memory, and anxiety behavior, little attention has as yet been given to the localization of plasminogen in the brain. We have investigated the localization of plasminogen in the adult mouse brain by using immunohistochemistry. In the hippocampus, plasminogen immunoreactivity was seen in the pyramidal cell layer as numerous punctate structures in neuronal somata. An electron-microscopic study further demonstrated that the plasminogen-immunoreactive punctate structures represented secretory vesicles and/or vesicle clusters. In the cerebral cortex, plasminogen immunoreactivity was evident in the somata of the layer II/III and V neurons. A quantitative analysis revealed that parvalbumin (PV)-positive neurons had more plasminogen-immunoreactive puncta compared with those of PV-negative neurons in the hippocampus and cerebral cortex. Plasminogen immunoreactivity was present throughout the hypothalamus, being particularly prominent in the neuronal somata of the organum vasculosum laminae terminalis, ventromedial preoptic nucleus, supraoptic nucleus, subfornical organ, medial part of the paraventricular nucleus (PVN), posterior part of the PVN, and arcuate hypothalamic nucleus. Thus, plasminogen is highly expressed in specific populations of hippocampal, cortical, and hypothalamic neurons, and plasminogen-containing vesicles are mainly observed at neuronal somata.


Assuntos
Córtex Cerebral/química , Hipocampo/química , Hipotálamo/química , Plasminogênio/análise , Animais , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/química , Núcleo Hipotalâmico Paraventricular/química , Plasminogênio/metabolismo
12.
Neuroreport ; 21(8): 575-9, 2010 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-20431496

RESUMO

As stem cells can regenerate damaged tissue, their therapeutic potential on brain damage has been investigated. In this study, the effects of embryonic stem cell transplantation on brain damage were investigated by using a photochemically induced thrombotic brain damage model. Mice with systemic transplantation of embryonic stem cells expressing enhanced green fluorescence protein on day 1 showed a smaller brain lesion size on day 8 than the control mice. The smaller lesion was accompanied by the increase in the number of microvessels at the border of the damaged area. Inside and around the damaged lesion, no EGFP-positive cells were observed. These findings suggested that embryonic stem cell transplantation reduced the brain lesion through the acceleration of angiogenesis by endogenous endothelial cells.


Assuntos
Infarto Encefálico/cirurgia , Células-Tronco Embrionárias/transplante , Trombose Intracraniana/cirurgia , Neovascularização Fisiológica/fisiologia , Regeneração Nervosa/fisiologia , Transplante de Células-Tronco/métodos , Animais , Edema Encefálico/etiologia , Edema Encefálico/fisiopatologia , Edema Encefálico/cirurgia , Infarto Encefálico/patologia , Infarto Encefálico/fisiopatologia , Capilares/citologia , Capilares/fisiologia , Técnicas de Cultura de Células , Linhagem Celular , Modelos Animais de Doenças , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/fisiologia , Corantes Fluorescentes/efeitos da radiação , Corantes Fluorescentes/toxicidade , Trombose Intracraniana/patologia , Trombose Intracraniana/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microcirculação/fisiologia , Neoplasias/etiologia , Neoplasias/patologia , Neoplasias/fisiopatologia , Estimulação Luminosa/efeitos adversos , Complicações Pós-Operatórias/etiologia , Complicações Pós-Operatórias/patologia , Complicações Pós-Operatórias/fisiopatologia , Medição de Risco , Rosa Bengala/efeitos da radiação , Rosa Bengala/toxicidade , Transplante de Células-Tronco/efeitos adversos , Transplante Homólogo/efeitos adversos , Transplante Homólogo/métodos
13.
J Exp Med ; 207(4): 867-80, 2010 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-20231377

RESUMO

Platelet-derived growth factor CC (PDGF-CC) is the third member of the PDGF family discovered after more than two decades of studies on the original members of the family, PDGF-AA and PDGF-BB. The biological function of PDGF-CC remains largely to be explored. We report a novel finding that PDGF-CC is a potent neuroprotective factor that acts by modulating glycogen synthase kinase 3beta (GSK3beta) activity. In several different animal models of neuronal injury, such as axotomy-induced neuronal death, neurotoxin-induced neuronal injury, 6-hydroxydopamine-induced Parkinson's dopaminergic neuronal death, and ischemia-induced stroke, PDGF-CC protein or gene delivery protected different types of neurons from apoptosis in both the retina and brain. On the other hand, loss-of-function assays using PDGF-C null mice, neutralizing antibody, or short hairpin RNA showed that PDGF-CC deficiency/inhibition exacerbated neuronal death in different neuronal tissues in vivo. Mechanistically, we revealed that the neuroprotective effect of PDGF-CC was achieved by regulating GSK3beta phosphorylation and expression. Our data demonstrate that PDGF-CC is critically required for neuronal survival and may potentially be used to treat neurodegenerative diseases. Inhibition of the PDGF-CC-PDGF receptor pathway for different clinical purposes should be conducted with caution to preserve normal neuronal functions.


Assuntos
Apoptose/fisiologia , Encéfalo/citologia , Sobrevivência Celular/fisiologia , Quinase 3 da Glicogênio Sintase/metabolismo , Linfocinas/fisiologia , Neurônios/citologia , Fator de Crescimento Derivado de Plaquetas/fisiologia , Retina/citologia , Animais , Anticorpos/imunologia , Anticorpos/farmacologia , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose/genética , Encéfalo/irrigação sanguínea , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Isquemia Encefálica/patologia , Isquemia Encefálica/fisiopatologia , Isquemia Encefálica/prevenção & controle , Permeabilidade Capilar/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Córtex Cerebral/irrigação sanguínea , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/patologia , Feminino , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/genética , Perfilação da Expressão Gênica , Quinase 3 da Glicogênio Sintase/genética , Glicogênio Sintase Quinase 3 beta , Peróxido de Hidrogênio/farmacologia , Linfocinas/antagonistas & inibidores , Linfocinas/farmacologia , Linfocinas/uso terapêutico , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , N-Metilaspartato/farmacologia , Neovascularização Patológica/induzido quimicamente , Fatores de Crescimento Neural/genética , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Traumatismos do Nervo Óptico/tratamento farmacológico , Traumatismos do Nervo Óptico/patologia , Doença de Parkinson Secundária/induzido quimicamente , Doença de Parkinson Secundária/tratamento farmacológico , Doença de Parkinson Secundária/metabolismo , Doença de Parkinson Secundária/patologia , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Fator de Crescimento Derivado de Plaquetas/farmacologia , Fator de Crescimento Derivado de Plaquetas/uso terapêutico , Ratos , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/imunologia , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética
14.
Brain Res ; 1322: 109-17, 2010 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-20138161

RESUMO

The severity of an ischemic stroke is variable in patients, because the occlusion position on the artery and the territory of distal vessels are individual. However, the relationship between the extent of initial brain lesion and the subsequent pathophysiological responses is poorly understood. Here, we studied the effects of the initial brain lesion size on the subsequent pathophysiological responses by using a photochemically induced thrombotic brain damage (PIT-BD) model, in which the brain lesion size can be well-reproducibly controlled than that induced by a middle cerebral artery occlusion (MCA-O) model. In the PIT-BD model, a large lesion, which comprised 4.9% of the whole brain on day 3, showed a 56% reduction until day 7. However, a small lesion, which comprised 1.3% of the whole brain, showed a 30% reduction. In addition, on day 5, the activation of both microglia and astrocytes was lesser in mice with small lesions than in mice with large lesions. Furthermore, we found that, smaller lesions in mice lacking gene of urokinase-receptor (uPAR(-/-)) than wild type (uPAR(+/+)) mice on day 3 showed less reduction until day 7 in MCA-O model, whereas lesions with comparable size in uPAR(-/-) mice showed comparable reduction with uPAR(+/+) mice in PIT-BD model. Thus it was indicated that the less reduction of the lesions in uPAR(-/-) mice in the MCA-O model did not result from the deficient gene but the difference of the initial lesion size. These findings suggested that the more severe the brain damage, the stronger the subsequent pathophysiological responses.


Assuntos
Infarto Encefálico/patologia , Infarto Encefálico/fisiopatologia , Gliose/patologia , Gliose/fisiopatologia , Trombose Intracraniana/patologia , Trombose Intracraniana/fisiopatologia , Animais , Astrócitos/metabolismo , Astrócitos/patologia , Encéfalo/irrigação sanguínea , Encéfalo/patologia , Encéfalo/fisiopatologia , Infarto Encefálico/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Regulação da Expressão Gênica/genética , Gliose/metabolismo , Trombose Intracraniana/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/metabolismo , Microglia/patologia , Estimulação Luminosa , Fotoquímica , Receptores de Ativador de Plasminogênio Tipo Uroquinase/genética
15.
Blood Coagul Fibrinolysis ; 21(3): 272-8, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20160640

RESUMO

The fibrinolytic activity of blood is regulated by expressing tissue-type plasminogen activator (t-PA) and its specific inhibitor, type-1 plasminogen activator inhibitor (PAI-1), from vascular endothelial cells. Since t-PA is a major plasminogen activator in blood, it is considered that the binding protein for t-PA, which exists on endothelial cell membrane, immobilizes t-PA on the surface of endothelial cells and enhances their antithrombotic property. Recently, we have found a new t-PA binding protein in endothelial cells. Its amino acid sequence has matched that of human adenine nucleotide translocase-1 (ANT1). The aims of this study are to confirm the binding of t-PA to ANT1, and to clarify the effect of ANT1 on fibrinolytic activity around endothelial cells. ANT1 is prepared from recombinant glutathione S-transferase (GST)-ANT1 fusion protein, and reveals t-PA binding activity in a ligand blot assay. In addition, ANT1 is exclusively expressed on endothelial cell membrane by using pDisplay vector. Interaction of t-PA with ANT1, which is expressed on the surface of endothelial cells, is confirmed by IAsys binding analysis and chromogenic assay. The heterologous expression of ANT1 on endothelial cell membrane enhances the t-PA binding ability of endothelial cells and the effect of ANT1 expression on fibrinolytic activity is demonstrated by increasing t-PA-catalyzed plasminogen activation. These results suggest that a novel t-PA-binding protein, ANT1, may concentrate t-PA on the surface of cells and enhance fibrinolytic properties around endothelial cells; therefore, ANT1 can be a powerful tool for regulating the plasminogen activation system in the vessel.


Assuntos
Translocador 1 do Nucleotídeo Adenina/metabolismo , Células Endoteliais/metabolismo , Fibrinólise , Ativador de Plasminogênio Tecidual/metabolismo , Translocador 1 do Nucleotídeo Adenina/genética , Linhagem Celular , Humanos , Ligação Proteica , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
16.
Thromb Res ; 125(5): e214-21, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20064655

RESUMO

INTRODUCTION: Although the involvement of plasminogen in liver repair has been reported, its roles are still poorly understood. Here, we investigated the role of plasminogen in accumulations of macrophages and neutrophils after liver injury in mice with gene deficient of plasminogen (Plg(-/-)) or its wild type (Plg(+/+)). MATERIALS AND METHODS: Mice received traumatic liver injury caused by stabbing on the lobe or hepatic ischemia-reperfusion, and the damaged sites were histologically analyzed. RESULTS: After the traumatic liver injury, both the stab wound and the damaged tissue were decreased until day 7 in the Plg(+/+) mice. In contrast, both the stab wound and the damaged tissue were still remained until day 7 in the Plg(-/-) mice. On day 4 after traumatic liver injury, macrophages were abundant at the surrounding area of the damaged site in the Plg(+/+) mice. However, the macrophage accumulation was impaired in the Plg(-/-) mice. After hepatic ischemia-reperfusion injury, macrophage accumulation and decrease in the damaged tissue were also observed in the Plg(+/+) mice until day 7. In contrast, these responses were also impaired in the Plg(-/-) mice. Furthermore, neutrophil accumulation at the surrounding area of the damaged site was also impaired in the Plg(-/-) mice on day 4 after both liver traumatic liver injury and hepatic ischemia-reperfusion injury. CONCLUSIONS: Our data indicate that plasminogen plays a crucial role in macrophage accumulation together with the neutrophil accumulation after liver injury in both models, which may be essential for triggering the subsequent healing responses including decrease in the damaged tissue.


Assuntos
Fígado/lesões , Fígado/metabolismo , Macrófagos/metabolismo , Macrófagos/patologia , Plasminogênio/metabolismo , Ferimentos Perfurantes/metabolismo , Ferimentos Perfurantes/patologia , Animais , Fígado/patologia , Camundongos , Camundongos Knockout , Plasminogênio/genética , Regeneração/fisiologia
17.
Pathophysiology ; 17(2): 141-7, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19500952

RESUMO

Recent studies have remarkably clarified the physiological roles of fibrinolytic system components on memory formation, neuronal plasticity, neuronal migration, neuronal cell death and axonal regeneration, together with their pathophysiological roles in ischemic stroke, intracerebral hemorrhage and Alzheimer disease in the central nervous system. Fibrinolytic system components also have various roles in the peripheral and autonomic nervous system, not only under physiological conditions but also under pathophysiological conditions. In these roles, fibrinolytic system components work not only through the extracellular proteolytic cascade but also through activation of their own receptors. This paper reviews the roles of fibrinolytic system components in the nervous system.

18.
Blood ; 114(15): 3352-8, 2009 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-19608750

RESUMO

Tissue-type plasminogen activator (t-PA) is approved for treatment of ischemic stroke patients, but it increases the risk of intracranial bleeding (ICB). Previously, we have shown in a mouse stroke model that stromelysin-1 (matrix metalloproteinase-3 [MMP-3]) induced in endothelial cells was critical for ICB induced by t-PA. In the present study, using bEnd.3 cells, a mouse brain-derived endothelial cell line, we showed that MMP-3 was induced by both ischemic stress and t-PA treatment. This induction by t-PA was prevented by inhibition either of low-density lipoprotein receptor-related protein (LRP) or of nuclear factor-kappaB activation. LRP was up-regulated by ischemic stress, both in bEnd.3 cells in vitro and in endothelial cells at the ischemic damage area in the mouse stroke model. Furthermore, inhibition of LRP suppressed both MMP-3 induction in endothelial cells and the increase in ICB by t-PA treatment after stroke. These findings indicate that t-PA deteriorates ICB via MMP-3 induction in endothelial cells, which is regulated through the LRP/nuclear factor-kappaB pathway.


Assuntos
Isquemia Encefálica/enzimologia , Células Endoteliais/enzimologia , Proteínas Relacionadas a Receptor de LDL/metabolismo , Metaloproteinase 3 da Matriz/biossíntese , Acidente Vascular Cerebral/enzimologia , Ativador de Plasminogênio Tecidual/farmacologia , Animais , Isquemia Encefálica/genética , Linhagem Celular Transformada , Modelos Animais de Doenças , Indução Enzimática/efeitos dos fármacos , Humanos , Proteínas Relacionadas a Receptor de LDL/genética , Masculino , Metaloproteinase 3 da Matriz/genética , Camundongos , NF-kappa B/metabolismo , Acidente Vascular Cerebral/genética
19.
Proc Natl Acad Sci U S A ; 106(15): 6152-7, 2009 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-19369214

RESUMO

VEGF-B, a homolog of VEGF discovered a long time ago, has not been considered an important target in antiangiogenic therapy. Instead, it has received little attention from the field. In this study, using different animal models and multiple types of vascular cells, we revealed that although VEGF-B is dispensable for blood vessel growth, it is critical for their survival. Importantly, the survival effect of VEGF-B is not only on vascular endothelial cells, but also on pericytes, smooth muscle cells, and vascular stem/progenitor cells. In vivo, VEGF-B targeting inhibited both choroidal and retinal neovascularization. Mechanistically, we found that the vascular survival effect of VEGF-B is achieved by regulating the expression of many vascular prosurvival genes via both NP-1 and VEGFR-1. Our work thus indicates that the function of VEGF-B in the vascular system is to act as a "survival," rather than an "angiogenic" factor and that VEGF-B inhibition may offer new therapeutic opportunities to treat neovascular diseases.


Assuntos
Neovascularização Patológica , Fator B de Crescimento do Endotélio Vascular/metabolismo , Animais , Sobrevivência Celular , Células Cultivadas , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Genoma , Membro Posterior/irrigação sanguínea , Isquemia/genética , Isquemia/metabolismo , Camundongos , Camundongos Knockout , Ratos , Retina/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Regulação para Cima , Fator B de Crescimento do Endotélio Vascular/deficiência , Fator B de Crescimento do Endotélio Vascular/genética
20.
Thromb Res ; 122(5): 668-73, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18571222

RESUMO

The potential prothrombotic effect of the cyclooxygenase-2 (COX-2) inhibitor Rofecoxib (Vioxx) was investigated using murine thrombosis models. In a jugular vein thrombosis model (photochemically induced injury) in lean wild-type mice, Rofecoxib treatment for 4 weeks induced a mild prothrombotic tendency, as indicated by a shorter occlusion time as compared to placebo (median of 12 min versus 36 min; p < 0.05). Thrombus size was somewhat, but not significantly, enhanced after Rofecoxib treatment. In a femoral artery thrombosis model (FeCl3 induced injury) Rofecoxib did not cause an enhanced thrombotic tendency in mice with nutritionally induced or genetically determined (ob/ob) obesity. The occlusion time was comparable for obese wild-type mice with (8.8+/-0.7 min) or without (7.8+/-2.1 min) Rofecoxib treatment, as well as for ob/ob mice (8.5+/-0.7 min versus 6.8+/-3.0 min). Thus, an enhanced prothrombotic effect of Rofecoxib was detected when using a venous thrombosis model in lean mice, but not when using an arterial thrombosis model in obese mice.


Assuntos
Inibidores de Ciclo-Oxigenase 2/toxicidade , Lactonas/toxicidade , Sulfonas/toxicidade , Trombose Venosa/induzido quimicamente , Animais , Modelos Animais de Doenças , Artéria Femoral , Veias Jugulares , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Obesos , Obesidade/sangue , Obesidade/complicações , Agregação Plaquetária/efeitos dos fármacos , Trombose/sangue , Trombose/induzido quimicamente , Trombose/etiologia , Trombose Venosa/sangue , Trombose Venosa/etiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...