Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Fluids Barriers CNS ; 18(1): 38, 2021 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-34399800

RESUMO

BACKGROUND: Thrombin has been implicated in playing a role in hydrocephalus development following intraventricular hemorrhage (IVH). However, the mechanisms underlying the sex differences to the detrimental effects of thrombin post-IVH remain elusive. METHOD: Three-month old male and female Sprague-Dawley rats underwent unilateral intracerebroventricular (ICV) injections of 3U or 5U thrombin, or saline, to examine differences in thrombin-induced hydrocephalus and white matter injury. Mortality, and lateral ventricle volume and white matter injury were measured on magnetic resonance imaging evaluation at 24 h post-injection. In addition, male rats were pretreated with 17-ß estradiol (E2, 5 mg/kg) or vehicle at 24 and 2 h prior to ICV injection of 3U thrombin. All rats were euthanized at 24 h post-injection for histology and immunohistochemistry. RESULTS: ICV injection of 5U thrombin caused 100 and 0% mortality in female and male rats, respectively. 3U of thrombin resulted in significant ventricular dilation and white matter damage at 24 h in both male and female rats, but both were worse in females (p < 0.05). Furthermore, neutrophil infiltration into choroid plexus and periventricular white matter was enhanced in female rats and may play a critical role in the sex difference in brain injury. Pre-treating male rats with E2, increased thrombin (3U)-induced hydrocephalus, periventricular white matter injury and neutrophil infiltration into the choroid plexus and white matter. CONCLUSIONS: ICV thrombin injection induced more severe ventricular dilation and white matter damage in female rats compared to males. Estrogen appears to contribute to this difference which may involve greater neutrophil infiltration in females. Understanding sex differences in thrombin-induced brain injury may shed light on future interventions for hemorrhagic stroke.


Assuntos
Hidrocefalia/induzido quimicamente , Hidrocefalia/patologia , Neutrófilos/fisiologia , Caracteres Sexuais , Trombina/toxicidade , Substância Branca/patologia , Animais , Ventrículos Cerebrais/irrigação sanguínea , Ventrículos Cerebrais/efeitos dos fármacos , Ventrículos Cerebrais/patologia , Plexo Corióideo/irrigação sanguínea , Plexo Corióideo/efeitos dos fármacos , Plexo Corióideo/patologia , Feminino , Injeções Intraventriculares , Masculino , Ratos , Ratos Sprague-Dawley , Trombina/administração & dosagem , Substância Branca/efeitos dos fármacos , Substância Branca/lesões
2.
Inflammation ; 43(5): 1959-1970, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32705396

RESUMO

Increasing evidence has revealed that the uncontrolled thrombin-induced inflammation following intracerebral hemorrhage (ICH) plays a key role in ICH. Oxidative stress and neuroinflammatory responses are interdependent and bidirectional events. Melatonin is now recognized as an antioxidant and a free radical scavenger due to its roles in various physiological and pathological processes. The aim of this study was to explore the molecular mechanisms underlying the effects of melatonin on thrombin-induced microglial inflammation and its indirect protection of HT22 cells from p53-associated apoptosis. Melatonin treatment attenuated the expression of IL-1ß, IL-18, cleaved caspase-1, and NLRP3 and decreased the production of reactive oxygen species (ROS), revealing its inhibitory effects against ROS-NLRP3 inflammasome activation. In further experiments investigating the protection conferred by melatonin, incubating HT22 cells with conditioned medium (CM) from thrombin-stimulated microglia induced HT22 cell apoptosis, and this effect was reversed after treating CM with either melatonin or N-acetyl-L-cysteine (NAC). Additionally, the Bax/Bcl-2 ratio and the levels of cleaved caspase-3 and p53 were markedly lower in the cells cultured in thrombin + melatonin-CM than in the cells cultured in thrombin-CM. Furthermore, the levels of MMP, ROS, SOD, MDA, and GSH-PX in bystander HT22 cells suggested that melatonin decreased HT22 cell apoptosis instigated via the p53-associated apoptotic pathway. Therefore, these findings strongly indicate the anti-inflammatory properties of melatonin that may suppress ROS-NLRP3 inflammasome activation and protect HT22 cells against apoptosis by inhibiting the ROS-mediated p53-dependent mitochondrial apoptotic pathway.


Assuntos
Antioxidantes/farmacologia , Apoptose/efeitos dos fármacos , Citoproteção/efeitos dos fármacos , Mediadores da Inflamação/antagonistas & inibidores , Melatonina/farmacologia , Trombina/toxicidade , Animais , Apoptose/fisiologia , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Citoproteção/fisiologia , Relação Dose-Resposta a Droga , Mediadores da Inflamação/metabolismo , Camundongos , Microglia/efeitos dos fármacos , Microglia/metabolismo
3.
Biomed Res Int ; 2019: 2187306, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31467874

RESUMO

Thrombin plays a pivotal role in the pathogenesis of atherosclerosis. Baicalin, an active flavonoid compound, was shown to attenuate the development of atherosclerosis, but the mechanism remains elusive. In the present study, the role and mechanism of baicalin in thrombin-induced cell injury was investigated in human umbilical vein endothelial cells (HUVECs). Our results showed that baicalin significantly reduced thrombin-induced apoptosis of HUVECs. Additional experiments showed that baicalin inhibited thrombin-induced NF-κB activation and PAR-1 expression. In addition, baicalin decreased thrombin-induced PAR-1 expression by inhibiting ERK pathway. These results indicated that baicalin has protective effects on thrombin-induced cell injury in HUVECs possibly through inhibition of PAR-1 expression and its downstream NF-κB activation, which was mediated by ERK1/2 activation.


Assuntos
Aterosclerose/tratamento farmacológico , Flavonoides/farmacologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Receptor PAR-1/antagonistas & inibidores , Apoptose/efeitos dos fármacos , Aterosclerose/induzido quimicamente , Aterosclerose/patologia , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , NF-kappa B/genética , Substâncias Protetoras/farmacologia , Receptor PAR-1/genética , Trombina/toxicidade
4.
CNS Neurosci Ther ; 25(10): 1142-1150, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-30955248

RESUMO

AIMS: Previous studies indicated that intraventricular injection of thrombin would induce hydrocephalus. But how thrombin works in this process remains unclear. Since cadherin plays a critical role in hydrocephalus, we aimed to explore the mechanisms of how thrombin acted on choroid plexus vascular endothelium and how thrombin interacted with vascular endothelial-cadherin (VE-cadherin) during hydrocephalus. METHODS: There were two parts in this study. Firstly, rats received an injection of saline or thrombin into the right lateral ventricle. Magnetic resonance imaging was applied to measure the lateral ventricle volumes. Albumin leakage and Evans blue content were assessed to test the blood-brain barrier function. Immunofluorescence and Western blot were applied to detect the location and the expression of VE-cadherin. Secondly, we observed the roles of protease-activated receptors-1 (PAR1) inhibitor (SCH79797), Src inhibitor (PP2), p21-activated kinase-1 (PAK1) inhibitor (IPA3) in the thrombin-induced hydrocephalus, and their effects on the regulation of VE-cadherin. RESULTS: Our study demonstrated that intraventricular injection of thrombin caused significant downregulation of VE-cadherin in choroid plexus and dilation of ventricles. In addition, the inhibition of PAR1/p-Src/p-PAK1 pathway reversed the decrease of VE-cadherin and attenuated thrombin-induced hydrocephalus. CONCLUSIONS: Our results suggested that the thrombin-induced hydrocephalus was associated with the inhibition of VE-cadherin via the PAR1/p-Src/p-PAK1 pathway.


Assuntos
Caderinas/metabolismo , Endotélio Vascular/metabolismo , Hidrocefalia/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais/fisiologia , Trombina/administração & dosagem , Animais , Caderinas/antagonistas & inibidores , Endotélio Vascular/efeitos dos fármacos , Hidrocefalia/induzido quimicamente , Injeções Intraventriculares , Masculino , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Trombina/toxicidade
5.
Int J Cardiol ; 271: 219-227, 2018 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-29801760

RESUMO

BACKGROUND: Data with animal cells and models suggest that thrombin activates cardiac fibroblasts (Fib) to myofibroblasts (myoFib) via protease-activated receptor 1 (PAR1) cleavage, and in this way promotes adverse atrial remodeling and, thereby, atrial fibrillation (AF). OBJECTIVE: Here, we explored the effects of thrombin on human atrial Fib and whether they are antagonized by the clinically available direct thrombin inhibitor, dabigatran. METHODS: Fib isolated from atrial appendages of patients without AF undergoing elective cardiac surgery were evaluated for PAR expression and treated with thrombin with or without dabigatran. PAR1 cleavage, downstream signaling and myoFib markers were investigated by immunofluorescence and Western blot. Collagen synthesis, activity of matrix metalloprotease (MMP)-2 and proliferation were assessed by Picro-Sirius red staining, gelatinolytic zymography and BrdU incorporation, respectively. Fib function was studied as capability to contract a collagen gel and stimulate the chemotaxis of peripheral blood monocytes from healthy volunteers. RESULTS: Primary human atrial Fib expressed PAR1, while levels of the other PARs were very low. Thrombin triggered PAR1 cleavage and phosphorylation of ERK1/2, p38 and Akt, elicited a switch to myoFib enriched for αSMA, fibronectin and type I collagen, and induced paracrine/autocrine transforming growth factor beta-1, cyclooxygenase-2, endothelin-1 and chemokine (C-C motif) ligand 2 (CCL2); conversely, MMP-2 activity decreased. Thrombin-primed cells displayed enhanced proliferation, formed discrete collagen-containing cellular nodules, and stimulated the contraction of a collagen gel. Furthermore, their conditioned medium caused monocytes to migrate. All these effects were prevented by dabigatran. CONCLUSION: These results with human cells complete the knowledge about thrombin actions on cardiac Fib and strengthen the translational potential of the emerging paradigm that pharmacological blockade of thrombin may counteract molecular and cellular events underlying AF.


Assuntos
Antitrombinas/farmacologia , Dabigatrana/farmacologia , Fibroblastos/metabolismo , Átrios do Coração/metabolismo , Receptor PAR-1/antagonistas & inibidores , Trombina/toxicidade , Células Cultivadas , Fibroblastos/efeitos dos fármacos , Átrios do Coração/citologia , Átrios do Coração/efeitos dos fármacos , Humanos , Receptor PAR-1/biossíntese
6.
Int J Biol Macromol ; 82: 711-8, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26434517

RESUMO

Several studies have demonstrated that Astragalus polysaccharide (APS) has a protective effect on endothelial cells damaged by various factors. To examine the role of APS in the endothelial inflammatory response, rat bone marrow endothelial progenitor cells (EPCs) were isolated by density gradient centrifugation and identified by immunohistochemistry, then we established a model of inflammatory injury induced by thrombin and measured the effects of APS on EPC viability and proliferation by MTT assays. We also assayed the effect APS had on the inflammatory response, by examining the nuclear factor kappa B (NF-κB) signaling pathway, as well as the expression of intercellular adhesion molecule-1 (ICAM-1), vascular endothelial growth factor (VEGF) and its receptors Flt-1 and KDR. Results demonstrated that EPCs were damaged by thrombin, and APS appeared to inhibit this damage. APS suppressed thrombin-induced ICAM-1 expression by blocking NF-κB signaling in rat bone marrow EPCs, and up-regulating expression of VEGF and its receptors. We believed that APS may be used to treat and prevent EPC injury-related diseases.


Assuntos
Astrágalo/química , Células Progenitoras Endoteliais/efeitos dos fármacos , Células Progenitoras Endoteliais/metabolismo , Polissacarídeos/farmacologia , Substâncias Protetoras/farmacologia , Trombina/toxicidade , Animais , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Expressão Gênica , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/metabolismo , NF-kappa B/metabolismo , Polissacarídeos/química , Substâncias Protetoras/química , Ratos , Transdução de Sinais/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/metabolismo
7.
Acta Neurochir Suppl ; 121: 373-7, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26463977

RESUMO

Our previous studies demonstrated that thrombin is an important factor in brain injury after intracerebral and intraventricular hemorrhage. This study examined the effect of acetazolamide, a carbonic anhydrase inhibitor, on thrombin-induced hydrocephalus. There were two parts in this study. First, rats had an injection of either 50 µl saline or 3 U thrombin into the right lateral ventricle. Second, rats had an injection of 3 U thrombin into the right lateral ventricle and were treated with either vehicle or acetazolamide (30 mg/kg, intraperitoneally (IP)) at 1 h after thrombin infusion. Lateral ventricle volumes were measured in magnetic resonance imaging T2 images and the brains were used for histology analysis at 24 h later. Intraventricular injection of thrombin induced significantly larger ventricle volume (27.8 ± 3.7 vs 8.5 ± 1.3 mm(3), n = 6, p < 0.01) and more ventricular wall damage (the breakdown of the ependymal layer, 20.2 ± 3.1 vs 2.4 ± 0.8 %, n = 6, p < 0.01) compared with saline injection. Acetazolamide treatment (30 mg/kg, IP) markedly attenuated thrombin-induced hydrocephalus (16.1 ± 4.2 mm(3) vs 29.5 ± 5.3 mm(3), n = 6, p < 0.01). These results suggest decreasing CSF production by acetazolamide attenuated thrombin-induced hydrocephalus in rats.


Assuntos
Acetazolamida/farmacologia , Encéfalo/efeitos dos fármacos , Inibidores da Anidrase Carbônica/farmacologia , Epêndima/efeitos dos fármacos , Hidrocefalia/diagnóstico por imagem , Ventrículos Laterais/efeitos dos fármacos , Animais , Encéfalo/diagnóstico por imagem , Epêndima/patologia , Hemostáticos/toxicidade , Hidrocefalia/induzido quimicamente , Injeções Intraventriculares , Ventrículos Laterais/diagnóstico por imagem , Ventrículos Laterais/patologia , Imageamento por Ressonância Magnética , Masculino , Ratos , Ratos Sprague-Dawley , Trombina/toxicidade
8.
Acta Neurochir Suppl ; 121: 379-84, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26463978

RESUMO

We have previously shown that intracerebral hemorrhage-induced brain injury is less in rats bred for high aerobic capacity (high capacity runners; HCR) compared with those bred for low aerobic capacity (low capacity runners; LCRs). Thrombin, an essential component in the coagulation cascade, is produced after cerebral hemorrhage. Intraventricular injection of thrombin causes significant hydrocephalus and white matter damage. In the present study, we examined the effect of exercise capacity on thrombin-induced hydrocephalus and white matter damage. Mid-aged (13-month-old) female LCRs (n = 13) and HCRs (n = 12) rats were used in this study. Rats received an intraventricular injection of thrombin (3 U, 50 µl). All rats underwent magnetic resonance imaging (MRI) at 24 h and were then euthanized for brain histology and Western blot. The mortalities were 20 % in LCRs and 33 % in HCRs after thrombin injection (p > 0.05). No rats died after saline injection. Intraventricular thrombin injection resulted in hydrocephalus and periventricular white matter damage as determined on MRI. In LCR rats, thrombin induced significant ventricle enlargement (23.0 ± 2.3 vs12.8 ± 1.9 mm(3) in LCR saline group; p < 0.01) and white matter lesion (9.3 ± 7.6 vs 0.6 ± 0.5 mm(3) in LCR saline group, p < 0.05). In comparison, in HCR rats thrombin induced less ventricular enlargement (17.3 ± 3.9 vs 23.0 ± 2.3 mm(3) in LCRs, p < 0.01) and smaller white matter lesions (2.6 ± 1.2 mm(3) vs 9.3 ± 7.6 mm(3) in LCRs, p < 0.05). In LCR rats, there was also upregulation of heat shock protein-32, a stress marker, and microglial activation in the periventricular white matter. These changes were significantly reduced in HCR rats. Intraventricular injection of thrombin caused more white matter damage and hydrocephalus in rats with low aerobic capacity. A differential effect of thrombin may contribute to differences in the effects of cerebral hemorrhage with aerobic capacity.


Assuntos
Ventrículos Cerebrais/diagnóstico por imagem , Hidrocefalia/diagnóstico por imagem , Substância Branca/diagnóstico por imagem , Animais , Western Blotting , Encéfalo/diagnóstico por imagem , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Tolerância ao Exercício , Heme Oxigenase (Desciclizante)/efeitos dos fármacos , Heme Oxigenase (Desciclizante)/metabolismo , Hemostáticos/toxicidade , Hidrocefalia/induzido quimicamente , Hidrocefalia/metabolismo , Imuno-Histoquímica , Injeções Intraventriculares , Imageamento por Ressonância Magnética , Condicionamento Físico Animal , Ratos , Trombina/toxicidade
9.
Pharmacology ; 96(5-6): 284-9, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26502217

RESUMO

AIMS: 17ß-estradiol (E2) is a powerful neuroprotective agent in the central nervous system; however, little is known about its effects on intracerebral hemorrhage. This study examined the effects of E2 on thrombin-induced apoptosis in vitro and investigated the potential mechanisms. METHODS: Primary cultured cortical neurons were treated with E2 or vehicle and then the cells were exposed to thrombin. Neuronal apoptosis was assessed by flow cytometry. The phosphorylated c-Jun-N-terminal kinase (p-JNK), phosphorylated extracellular signal-regulated kinases 1/2 (p-ERK1/2), B-cell lymphoma 2 (Bcl-2), Bcl-2-associated X protein (Bax) and caspase-3 were assayed by western blot. RESULTS: Consequently, we found that E2 has significantly reduced the apoptosis in thrombin-treated neurons. E2 also exhibited a downregulation in the ratio of Bax/Bcl-2, caspase-3 and p-JNK. However, E2 had little effect on p-ERK1/2 proteins activation. CONCLUSION: Taken together, E2 has shown neuroprotective effects on thrombin-induced neuronal apoptosis, and the molecular mechanisms may correlate with the inhibition of the JNK signaling pathway.


Assuntos
Apoptose/efeitos dos fármacos , Córtex Cerebral/efeitos dos fármacos , Estradiol/farmacologia , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Trombina/toxicidade , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Western Blotting , Células Cultivadas , Córtex Cerebral/embriologia , Córtex Cerebral/metabolismo , Citometria de Fluxo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Cultura Primária de Células , Ratos
10.
Neuroreport ; 26(7): 416-23, 2015 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-25839175

RESUMO

(-)-Epigallocatechin-3-gallate (EGCG), the major polyphenolic component of green tea, has anti-inflammatory and antioxidant properties and provides neuroprotection against central nervous system diseases. Yet, it is not known whether EGCG may be neuroprotective against intracerebral hemorrhage. In this study, we used a simplified in-vitro model of thrombin neurotoxicity to test whether EGCG provides neuroprotection against thrombin-associated toxicity. Exposure of primary cortical neurons to thrombin (100 U/ml) caused dose-dependent and time-dependent cytotoxicity. Cell Counting Kit 8 and lactate dehydrogenase were used to monitor cell viability after exposure of neurons to thrombin or EGCG and after EGCG pretreatment. Flow cytometric analysis and western blotting demonstrated that thrombin-induced neuron degeneration occurs through apoptosis. A concentration of 25 µM EGCG significantly abolished thrombin-induced toxicity and prevented apoptosis by suppressing c-Jun-N-terminal kinase (JNK) phosphorylation, and the JNK inhibitor SP600125 reduced thrombin-induced caspase 3 activation and apoptosis. These data suggest that EGCG may have protective effects against thrombin-induced neuroapoptosis by inhibiting the activation of JNK, leading to caspase 3 cleavage. EGCG is a novel candidate neuroprotective agent against intracerebral hemorrhage-induced neurotoxicity.


Assuntos
Apoptose/efeitos dos fármacos , Catequina/análogos & derivados , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Trombina/toxicidade , Animais , Antracenos/farmacologia , Apoptose/fisiologia , Caspase 3/metabolismo , Catequina/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/fisiologia , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , L-Lactato Desidrogenase/metabolismo , MAP Quinase Quinase 4/antagonistas & inibidores , MAP Quinase Quinase 4/metabolismo , Neurônios/fisiologia , Fosforilação/efeitos dos fármacos , Ratos , Fatores de Tempo
11.
Brain Res ; 1613: 73-80, 2015 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-25862570

RESUMO

UNLABELLED: This study examines the effects of progesterone on blood-brain barrier (BBB) integrity following thrombin administration. Thrombin is expressed in many diseases which affect neural tissue and is associated with breakdown of the BBB. Progesterone has shown protective effects on the BBB in stroke and traumatic brain injury. METHODS: Mouse brain endothelial (bEnd.3) cells were treated with progesterone (20 µmol/l) for 24h before thrombin administration (60 U/ml). BBB permeability was measured by transendothelial electrical resistance (TEER), because TEER decrease is associated with BBB compromise. Tight junction (TJ) proteins (occludin, claudin-5, and zonula occludens-1) and endothelial protein C receptor (EPCR) were analyzed. RESULTS: Thrombin decreased TEER and progesterone prevented that decrease. TJ proteins and EPCR were also decreased after thrombin treatment and progesterone treatment blocked that effect. CONCLUSION: Progesterone can attenuate thrombin-induced BBB disruption by blocking the degradation of TJ proteins and EPCR in bEnd.3 cells.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Progesterona/farmacologia , Trombina/toxicidade , Animais , Isquemia Encefálica/metabolismo , Linhagem Celular , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Receptor de Proteína C Endotelial , Camundongos , Permeabilidade , Ratos Sprague-Dawley , Receptores de Superfície Celular , Acidente Vascular Cerebral/metabolismo , Proteínas de Junções Íntimas/efeitos dos fármacos , Proteínas de Junções Íntimas/metabolismo
12.
J Thorac Cardiovasc Surg ; 149(4): 982-8, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25641439

RESUMO

OBJECTIVE: Although fibrinogen- and thrombin-impregnated collagen (TachoSil; Takeda GmbH, Linz, Austria) can be applied to prevent air leakage, the impact of its use on lung healing is unknown. Therefore, we histologically evaluated the long-term healing process associated with the use of TachoSil to prevent air leakage in a canine model. METHODS: Via left thoracotomy, visceral pleural defects of 10 × 10 mm were created on each lung lobe of female beagles. After air leakage was confirmed, each pleural defect was covered with TachoSil. The repair sites were histologically evaluated on postoperative days 0, 4, 7, 14, 28, and 56. RESULTS: All animals survived, and none developed pneumothorax. Histologically, inflammatory cells infiltrated the TachoSil from the pleural defect, and pleural mesothelium comprised the regenerated outermost layer of the TachoSil soon after the surgery. Inflammatory cells, myofibroblasts, and neovascular vessels subsequently spread over the entire TachoSil. The number of inflammatory cells decreased, and myofibroblast and neovascular vessels replaced the entire TachoSil. In addition, the elastic layer started to regenerate from both edges and completely repaired the pleural defect. The lung parenchyma around the pleural defects was not influenced throughout the observational period, because these healing processes occurred only inside the TachoSil. CONCLUSIONS: TachoSil provided a mechanical scaffold on which healing could proceed, followed by biodegradation over the long term. TachoSil safely repaired the pleural defects without affecting lung parenchyma.


Assuntos
Fibrinogênio/farmacologia , Pleura/efeitos dos fármacos , Pneumotórax/prevenção & controle , Trombina/farmacologia , Cicatrização/efeitos dos fármacos , Animais , Modelos Animais de Doenças , Cães , Combinação de Medicamentos , Feminino , Fibrinogênio/toxicidade , Pleura/metabolismo , Pleura/cirurgia , Pneumotórax/patologia , Trombina/toxicidade , Fatores de Tempo
13.
Int J Clin Exp Pathol ; 8(11): 14021-7, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26823714

RESUMO

Baicalin, an extract from the dried root of Scutellaria baicalensis Georgi, was shown to be neuroprotective. However, the precise mechanisms are incompletely known. In this study, we determined the effect of baicalin on thrombin induced cell injury in SH-SY5Y cells, and explored the possible mechanisms. SH-SY5Y cells was treated with thrombin alone or pre-treated with baicalin (5, 10, 20 µM) for 2 h followed by thrombin treatment. Cells without thrombin and baicalin treatment were used as controls. Cell viability was detected by MTT assay. Cell apoptosis was analyzed by flow cytometry. Real-time PCR was performed to determine the mRNA expression of protease-activated receptor-1 (PAR-1). Western blotting was conducted to determine the protein expression of PAR-1, Caspase-3 and NF-κB. Baicalin reduced cell death following thrombin treatment in a dose-dependent manner, with concomitant inhibition of NF-κB activation and suppression of PAR-1 expression. In addition, baicalin reduced Caspase-3 expression. The above findings indicated that baicalin prevents against cell injury after thrombin stimulation possibly through inhibition of PAR-1 expression and NF-κB activation.


Assuntos
Flavonoides/farmacologia , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Trombina/toxicidade , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Citoproteção , Relação Dose-Resposta a Droga , Humanos , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Receptor PAR-1/antagonistas & inibidores , Receptor PAR-1/genética , Receptor PAR-1/metabolismo , Transdução de Sinais/efeitos dos fármacos
14.
Mol Ther ; 23(3): 445-55, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25502903

RESUMO

Recent evidence has shown that Ras homolog enriched in brain (Rheb) is dysregulated in Alzheimer's disease (AD) brains. However, it is still unclear whether Rheb activation contributes to the survival and protection of hippocampal neurons in the adult brain. To assess the effects of active Rheb in hippocampal neurons in vivo, we transfected neurons in the cornu ammonis 1 (CA1) region in normal adult rats with an adeno-associated virus containing the constitutively active human Rheb (hRheb(S16H)) and evaluated the effects on thrombin-induced neurotoxicity. Transduction with hRheb(S16H) significantly induced neurotrophic effects in hippocampal neurons through activation of mammalian target of rapamycin complex 1 (mTORC1) without side effects such as long-term potentiation impairment and seizures from the alteration of cytoarchitecture, and the expression of hRheb(S16H) prevented thrombin-induced neurodegeneration in vivo, an effect that was diminished by treatment with specific neutralizing antibodies against brain-derived neurotrophic factor (BDNF). In addition, our results showed that the basal mTORC1 activity might be insufficient to mediate the level of BDNF expression, but hRheb(S16H)-activated mTORC1 stimulated BDNF production in hippocampal neurons. These results suggest that viral vector transduction with hRheb(S16H) may have therapeutic value in the treatment of neurodegenerative diseases such as AD.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/biossíntese , Região CA1 Hipocampal/metabolismo , Proteínas Monoméricas de Ligação ao GTP/genética , Neurônios/metabolismo , Neuropeptídeos/genética , Transdução Genética/métodos , Animais , Anticorpos Neutralizantes/farmacologia , Fator Neurotrófico Derivado do Encéfalo/agonistas , Fator Neurotrófico Derivado do Encéfalo/antagonistas & inibidores , Fator Neurotrófico Derivado do Encéfalo/genética , Região CA1 Hipocampal/citologia , Região CA1 Hipocampal/efeitos dos fármacos , Dependovirus/genética , Dependovirus/metabolismo , Expressão Gênica , Vetores Genéticos/administração & dosagem , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Complexos Multiproteicos/agonistas , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neuropeptídeos/metabolismo , Proteína Enriquecida em Homólogo de Ras do Encéfalo , Ratos , Ratos Sprague-Dawley , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo , Trombina/antagonistas & inibidores , Trombina/toxicidade
15.
Can J Neurol Sci ; 41(6): 714-20, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25382279

RESUMO

BACKGROUND: Brain injury after intracerebral hemorrhage (ICH) arises from numerous contributors, of which some also play essential roles. Notably, thrombin production, needed to stop bleeding, also causes acute cell death and edema. In some rodent models of ICH, peri-hematoma neurons die over weeks. Hence we evaluated whether thrombin is responsible for this chronic degeneration. Functional impairments after ICH also result from sub-lethal damage to neurons, especially the loss of dendrites. Thus, we evaluated whether thrombin infusion alone, a reductionist model of ICH, causes similar injury. METHODS: Adult rats had a modest intra-striatal infusion of thrombin (1 U) or saline followed by a behavioral test, to verify impairment, 7 days later. After this they were euthanized and tissue stained with Golgi-Cox solution to allow the assessment of dendritic morphology in striatal neurons. In a second experiment, rats survived 7 or 60 days after thrombin infusion in order to histologically determine lesion volume. RESULTS: Thrombin caused early cell death and considerable atrophy in surviving peri-lesion neurons, which had less than half of their usual numbers of branches. However, total tissue loss was comparable at 7 (24.1 mm3) and 60 days (25.6 mm3). CONCLUSION: Thrombin infusion causes early cell death and neuronal atrophy in nearby surviving striatal neurons but thrombin does not cause chronic tissue loss. Thus, the chronic degeneration found after ICH in rats is not simply and solely due to acute thrombin production. Nonetheless, thrombin is an important contributor to behavioral dysfunction because it causes cell death and substantial dendritic injury.


Assuntos
Atrofia/induzido quimicamente , Degeneração Neural/induzido quimicamente , Neurônios/efeitos dos fármacos , Trombina/administração & dosagem , Trombina/toxicidade , Animais , Atrofia/patologia , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Hemorragia Cerebral/induzido quimicamente , Hemorragia Cerebral/patologia , Infusões Intraventriculares , Masculino , Degeneração Neural/patologia , Neurônios/patologia , Ratos , Ratos Sprague-Dawley
16.
PLoS One ; 9(6): e97946, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24959881

RESUMO

Intracerebral hemorrhage (ICH) is a devastating neurological disease with a grave prognosis. We evaluated microRNA (miRNA) expression after ICH and evaluated Let7c as a therapeutic target. We harvested hemorrhagic brain 24 hours after collagenase induced ICH in the rat. Microarray analysis was performed to compare the miRNAs expression pattern between hemorrhagic hemisphere and contralateral hemisphere. An in vitro thrombin toxicity model and blood injection ICH model were also used to evaluate miRNA expression. We selected miRNA for the therapeutic target study after reviewing target gene databases and their expression. The antagonistic sequence of the selected miRNA (antagomir) was used to evaluate its therapeutic potential in the in vitro thrombin toxicity and in vivo ICH models. Among 1,088 miRNAs analyzed, let7c was induced in the thrombin and ICH models. Let7c antagomir treatment increased cell survival in the in vitro thrombin injury model and improved neurological function at 4 weeks after ICH. Let7c antagomir decreased perihematoma edema, apoptotic cell death and inflammation around hematoma. Let7c antagomir also induced insulin like growth factor receptor 1 (IGF1R) protein and phosphorylated serine threonine kinase after ICH. This study shows a distinct miRNA expression pattern after ICH. The let7c antagomir reduced cell death and edema and enhanced neurological recovery at least in part by activating the IGF1R pro-survival pathway. This suggests blocking let7c might be a potential therapeutic target in ICH.


Assuntos
Encéfalo/patologia , Hemorragia Cerebral/genética , Hemostáticos/toxicidade , MicroRNAs/genética , Oligonucleotídeos/farmacologia , Trombina/toxicidade , Animais , Encéfalo/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular , Hemorragia Cerebral/induzido quimicamente , Hemorragia Cerebral/patologia , Hemorragia Cerebral/terapia , Colagenases/efeitos adversos , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Análise de Sequência com Séries de Oligonucleotídeos , Oligonucleotídeos/uso terapêutico , Ratos , Receptor IGF Tipo 1/metabolismo , Regulação para Cima
17.
Neurochem Res ; 39(7): 1219-31, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24706094

RESUMO

Thrombin effect increasing swelling-induced glutamate efflux was examined in cultured cortical astrocytes, cerebellar granule neurons (CGN), hippocampal and cortical neurons. Hypotonic glutamate efflux (monitored by D-[(3)H]aspartate) from cortical astrocytes was increased by thrombin (5 U/mL) to reach 16% of the cell pool in 5 min. Thrombin had lower effects in CGN, and marginal effects in hippocampal and cortical neurons. These differences were related to the magnitude of thrombin-evoked cytosolic calcium rise. The protease-activated receptor 1 is expressed in astrocytes and neurons. In astrocytes exposed to chemical ischemia (sodium iodoacetate plus sodium azide) D-[(3)H]aspartate release showed a first phase (20-40 min) of initial low efflux which progressively increases; and a second phase (40-60 min) of larger efflux coincident with cell swelling. Efflux at the first phase was 52% inhibited by the glutamate transporter blocker DL-threo-ß-benzyloxyaspartic-acid (TBOA) and 11% by the volume-sensitive pathway blocker phloretin. At the second phase, efflux was reduced 52 and 38% respectively, by these blockers. In CGN D-[(3)H]aspartate efflux increased sharply and then decreased. This efflux was 32% reduced by calcium omission, 46% by TBOA and 32% by 4-[(2-butyl-6,7dichloro-2-cyclopentyl-2,3-dihydro-1oxo-1H-inden-5-yl)oxy] butanoic-acid. Thrombin enhanced this release by 32%. Ischemic treatment increased astrocyte mortality from 4% in controls to 39 and 61% in ischemia and ischemia plus thrombin, respectively. Cell death was prevented by phloretin. CGN viability was unaffected by the treatment. These results suggest that coincidence of swelling and thrombin during ischemia elevates extracellular glutamate prominently from astrocyte efflux, which may endanger neurons in vivo.


Assuntos
Ácido Aspártico/metabolismo , Astrócitos/metabolismo , Hiponatremia/metabolismo , Neurônios/metabolismo , Trombina/toxicidade , Trítio/metabolismo , Animais , Astrócitos/efeitos dos fármacos , Hipóxia Celular/efeitos dos fármacos , Hipóxia Celular/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Neurônios/efeitos dos fármacos , Ratos , Ratos Wistar
18.
Transl Stroke Res ; 5(4): 472-5, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24323711

RESUMO

Thrombin causes blood-brain barrier disruption, and this study examined whether thrombin can cause brain hemorrhage through protease-activated receptor-1 (PAR-1). Male wild type and PAR-1 knockout mice had an intracerebral injection of thrombin or saline. Mice then underwent serial T2 magnetic resonance imaging and were euthanized for brain hemoglobin, iron, and interleukin-1ß measurements. Thrombin caused massive T2 lesions and brain hemorrhage in wild type mice. These effects were markedly reduced in PAR-1 knockout mice. Thrombin also increased brain interleukin-1ß, and this was absent in PAR-1 knockout mice. In conclusion, thrombin increases interleukin-1ß levels and induces intracerebral hemorrhage through PAR-1 activation.


Assuntos
Hemorragia Cerebral/induzido quimicamente , Hemorragia Cerebral/metabolismo , Hemostáticos/toxicidade , Receptor PAR-1/metabolismo , Trombina/toxicidade , Animais , Gânglios da Base/metabolismo , Interleucina-1beta/metabolismo , Masculino , Camundongos , Camundongos Knockout
19.
Transl Stroke Res ; 5(4): 526-33, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24347404

RESUMO

This study characterizes a new model of thromboembolic stroke of the middle cerebral artery in C57 black/6J mice, thus offering an opportunity to use the model for studying ischemic stroke in transgenic mice. Thromboembolic stroke was induced by local injection of either 1.5 or 3.0 UI of thrombin directly into the right MCA of C57 black/6J mice. Cerebral blood flow (CBF) velocity was measured continuously by laser Doppler flowmetry, which allowed documentation of both MCA occlusion and of spontaneous recanalization. After 24 h, all animals were euthanized. Cryosections were cut at 400-µm intervals and silver stained with the high-contrast method for volumetric assessment of infarct size. Interleukin (IL)-6, tumor necrosis factor-alpha (TNF-α), caspase-3 and hsp 70 protein levels were investigated by immunofluorescence. Thrombin injection resulted in clot formation in all animals. Cortical infarction occurred in 63% of the mice while 37% had a spontaneous MCA recanalization during the first 20 min following thrombin injection. In cases of successful MCA occlusion with consequent infarction, the clot was stable up to 2 h after formation. Subsequently, 20% recanalized spontaneously. Infarctions were restricted to the cortex with a mean lesion volume of 36 ± 5 for 1.5 UI and 56 ± 8 for 3.0 UI thrombin. Protein levels of IL-6, TNF-α, caspase-3, and hsp 70 were significantly increased after MCAO. The results demonstrate that the mouse thromboembolic stroke model produces cortical infarctions of consistent size in C57 black/6J mice, which is dependent upon the amount of thrombin used for clot formation. Spontaneous MCA recanalization occurs after 2 h of ischemia in 20% of mice. Thus, the thromboembolic model is an applicable stroke model for C57 black/6J mice, which mimics many of the features of human stroke, including spontaneous recanalization. However, strain differences between Swiss and C57 black/6J mice must be taken into account when using the model.


Assuntos
Modelos Animais de Doenças , Trombose Intracraniana/metabolismo , Trombose Intracraniana/patologia , Camundongos Endogâmicos C57BL , Acidente Vascular Cerebral/metabolismo , Acidente Vascular Cerebral/patologia , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Caspase 3/metabolismo , Citocinas/metabolismo , Proteínas de Choque Térmico HSP70/metabolismo , Hemostáticos/toxicidade , Infarto da Artéria Cerebral Média/induzido quimicamente , Camundongos , Trombina/toxicidade
20.
Zhejiang Da Xue Xue Bao Yi Xue Ban ; 42(3): 283-90, 2013 May.
Artigo em Chinês | MEDLINE | ID: mdl-23801616

RESUMO

OBJECTIVE: To investigate the roles of protease-activated receptors (PARs) in thrombin-induced brain injury and neurogenesis in rats. METHODS: Ninety male SD rats were randomly assigned to receive intra-hippocampus injection of NS, thrombin or specific agonists of 3 protease-activated receptors (PAR-1, PAR-3 and PAR-4), respectively. At 1,3 and 7 d after injection, the area of the hippocampus was determined with HE staining, the density and morphology of astrocyte were detected with GFAP staining, degenerated neurons were detected with Fluoro-Jade C staining, and the neurogenesis was examined with DCX staining. RESULTS: Compared to NS injection, the area of the hippocampus significantly increased at 1-3 d and decreased at 7 d after the injection of thrombin and PAR-1 agonist (P<0.05). In addition, injection of thrombin and PAR-1 agonist significantly increased the density of astrocyte and Fluoro-Jade C positive cells at 1-7 d after injection (P<0.05), and significantly increased the density of DCX positive cells at 3-7 d after injection(P<0.05). The injection of PAR-3 agonist and PAR-4 agonist had no affect on the area of the hippocampus, the density of astrocyte, Fluoro-Jade C positive cells and DCX positive cells. CONCLUSION: The activation of protease-activated receptor-1 may be related to the thrombin-induced brain injury and neurogenesis in rat hippocampus.


Assuntos
Lesões Encefálicas/patologia , Neurogênese/efeitos dos fármacos , Receptor PAR-1/agonistas , Receptor PAR-1/fisiologia , Trombina/toxicidade , Animais , Lesões Encefálicas/induzido quimicamente , Lesões Encefálicas/metabolismo , Proteína Duplacortina , Hipocampo/patologia , Masculino , Ratos , Ratos Sprague-Dawley , Receptores de Trombina/agonistas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...