Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Tumour Biol ; 36(11): 8617-24, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26040766

RESUMEN

Poly(ADP-ribose) polymerase 3 (PARP3), a critical player in cellular response to DNA double-strand breaks (DSBs), plays an essential role in the maintenance of genome integrity. However, the role of PARP3 in tumorigenesis especially in glioblastoma remains largely unknown. In the present study, we found that the mRNA and protein levels of PARP3 were upregulated in primary glioblastoma tissues. Knockdown of PARP3 expression by lentivirus-based shRNA decreased cell glioblastoma proliferation and inhibited tumor growth in vivo by using a xenograft mouse model. Furthermore, we found that silencing the expression of PARP3 resulted in a synergistic radiosensitizing effect when combined with radiotherapy in glioblastoma cell lines. At the molecular level, we found that PARP3 interacted with FoxM1 to enhance its transcriptional activity and conferred glioblastoma cell radioresistance. Thus, our data suggest that PARP3 could be a therapeutic target to overcome radioresistance in glioblastoma.


Asunto(s)
Proteínas de Ciclo Celular/genética , Factores de Transcripción Forkhead/genética , Glioblastoma/genética , Glioblastoma/radioterapia , Poli(ADP-Ribosa) Polimerasas/genética , Tolerancia a Radiación/genética , Animales , Carcinogénesis/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Proliferación Celular/genética , Proteína Forkhead Box M1 , Factores de Transcripción Forkhead/metabolismo , Glioblastoma/patología , Humanos , Ratones , Poli(ADP-Ribosa) Polimerasas/metabolismo , ARN Mensajero/genética , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Neurochem Res ; 40(3): 591-9, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25542238

RESUMEN

Cerebral inflammation plays a crucial role in early brain injury (EBI) after subarachnoid hemorrhage (SAH). This study investigated the effects of c-Jun N-terminal kinase (JNK) inhibitor SP600125, acetylcholine (Ach), etanercept, and anti-TNF-α on cellular apoptosis in the cerebral cortex and the hippocampus, in order to establish the role of JNK and TNF-α in EBI. The SAH model was established using an endovascular puncture protocol. The reliability of the EBI model was determined by phosphorylated-Bad (pBad) immunohistochemistry. Neurological scores were recorded and western blot was used to detect the expression of JNK and TNF-α, and TUNEL assay was used to mark apoptotic cells. The results showed that pBad positive cells were evenly distributed in the cerebral cortex at different time points. The highest expression of pBad was reached 1 day after SAH, and pJNK and TNF-α reached their peak expression at 2 days after SAH. SP600125, Ach, and etanercept significantly decreased the level of pJNK and TNF-α in the cerebral cortex and the hippocampus. In addition, SP600125 and etanercept reduced cellular apoptosis in the cerebral cortex and the hippocampus and significantly improved neurological scores at 2 days after SAH potentially via inhibition of the JNK-TNF-α pathway. Ach reduced cellular apoptosis only in the cerebral cortex. It is possible that JNK induces TNF-α expression, which in turn enhances JNK expression in EBI after SAH, leading to increased apoptosis in the cerebral cortex and the hippocampus. Thus, our results indicate that that etanercept may be a potential therapeutic agent to alleviate EBI.


Asunto(s)
Lesiones Encefálicas/tratamiento farmacológico , Etanercept/uso terapéutico , Proteínas Quinasas JNK Activadas por Mitógenos/fisiología , Hemorragia Subaracnoidea/tratamiento farmacológico , Factor de Necrosis Tumoral alfa/fisiología , Animales , Antiinflamatorios no Esteroideos/farmacología , Antiinflamatorios no Esteroideos/uso terapéutico , Lesiones Encefálicas/etiología , Lesiones Encefálicas/metabolismo , Etanercept/farmacología , Masculino , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Hemorragia Subaracnoidea/complicaciones , Hemorragia Subaracnoidea/metabolismo
3.
Cell Biol Int ; 39(7): 788-98, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25612169

RESUMEN

A rapid increase in matrix metalloproteinase-9 (MMP-9) expression by stimulated leukocytes is common in many diseases. Recent evidence suggests that the beneficial effects of statins are mediated in part by the suppression of MMP-9 release. In this study, we investigated the effect of statin on MMP-9 expression and its antagonist, tissue inhibitor of metalloproteinase-1 (TIMP-1) in LPS-stimulated leukocytes. Rat neutrophils and monocytes were stimulated with lipopolysaccharide (LPS) in the presence of simvastatin. MMP-9 secretion and mRNA expression were analyzed using ELISA and RT-PCR, respectively. Total MMP-9 protein production was measured by Western blot analysis. Potential signal transduction pathways responsible for MMP-9 production were investigated using luciferase reporter assays (NF-κB), pull-down assays (RhoA), and pharmacological inhibition. Our data show that MMP-9 and TIMP-1 expression are differentially induced by LPS in neutrophils and monocytes. We showed that rapid MMP-9 release occurred mainly via secretion from intracellular stores. Moreover, we showed that statin significantly suppressed LPS-induced MMP-9 release and mRNA expression in a time- and concentration-dependent manner. We also evaluated that simvastain postponed the rapid LPS-induced MMP-9 release for about 20 min. In conclusion, we demonstrated that the suppressive effect of simvastatin on LPS-stimulated MMP-9 release does not occur via the NF-κB pathway and the MAPKs pathway, but via the RhoA/ROCK pathway.


Asunto(s)
Anticolesterolemiantes/farmacología , Lipopolisacáridos/inmunología , Metaloproteinasa 9 de la Matriz/inmunología , Transducción de Señal/efectos de los fármacos , Simvastatina/farmacología , Animales , Células Cultivadas , Monocitos/efectos de los fármacos , Monocitos/inmunología , FN-kappa B/inmunología , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología , Ratas Sprague-Dawley , Quinasas Asociadas a rho/inmunología , Proteína de Unión al GTP rhoA/inmunología
4.
Acta Neurochir (Wien) ; 157(5): 781-92, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25697836

RESUMEN

BACKGROUND: AMP-activated protein kinase (AMPK) is a key metabolic and stress sensor/effector. Few investigations have been performed to study the role of AMPK in subarachnoid hemorrhage (SAH)-induced early brain injury (EBI). This study was undertaken to investigate the time course of AMPK activation in the early stage of SAH and to evaluate the influence of AICAR (which is known to mimic AMP and activates AMPK) and compound C (a commonly used AMPK inhibitor) on EBI in rats following SAH. METHODS: Adult male rats were divided into six groups: control, sham, SAH, SAH + vehicle, SAH + AICAR and SAH + compound C. SAHs were induced by a modified endovascular perforation method. Immunohistochemistry, real-time PCR and Western blot were used to detect the spatial and dynamic expression of AMPK after SAH. Cortical apoptosis and the expressions of apoptosis-related proteins such as FOXO3a (forkhead box, class O, 3a) and Bim (Bcl-2-interacting mediator of cell death) were detected after different drug interventions. RESULTS: We found SAH induced prolonged activation of AMPK. Treatment with AICAR markedly induced overactivation of AMPK and upregulation of FOXO3a and Bim. AICAR also significantly exacerbated cerebral apoptosis and neurological impairment following SAH. On the other hand, pre-administration of compound C attenuated EBI in this SAH model by modulating cerebral apoptosis by inhibiting FOXO3a and Bim. CONCLUSIONS: Our findings suggest that the AMPK pathway may play an important role in SAH-induced neuronal apoptosis, and the use of AMPK inhibitors can provide neuroprotection in EBI after SAH.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Transducción de Señal , Hemorragia Subaracnoidea/metabolismo , Animales , Apoptosis , Masculino , Ratas , Ratas Sprague-Dawley
5.
Int J Neurosci ; 125(3): 161-9, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24754439

RESUMEN

Ischemic postconditioning refers to controlling reperfusion blood flow during reperfusion after ischemia, which can induce an endogenous neuroprotective effect and reduce ischemia-reperfusion injury. Activation of endogenous neuroprotective mechanisms plays a key role in protecting against brain ischemia-reperfusion injury. The mechanisms of cerebral ischemic postconditioning are not completely clear, and the following aspects may be involved: downregulation of oxidative stress, attenuating mitochondrial dysfunction, attenuating endoplasmic reticulum stress, accelerating the elimination of glutamate, increasing rCBF, inhibiting apoptosis, inhibiting autophagy, and regulating signal transduction.


Asunto(s)
Investigación Biomédica , Poscondicionamiento Isquémico/métodos , Fármacos Neuroprotectores/uso terapéutico , Daño por Reperfusión/prevención & control , Animales , Isquemia Encefálica/complicaciones , Humanos , Daño por Reperfusión/etiología , Transducción de Señal/fisiología
6.
Rev Neurosci ; 25(2): 247-54, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24501156

RESUMEN

Caveolin-1 is the principal marker of caveolae in endothelial cells. It plays an important role in physiological and pathological conditions of the blood-brain barrier and serves as a mediator in drug delivery through the blood-brain barrier. Caveolin-1 is related to the diminished expression of tight junction-associated proteins and metabolic pinocytosis vesicles when the blood-brain barrier is destroyed by outside invaders or malignant stimulus. The permeability of the blood-brain barrier, regulated by types of drugs or physical irradiation, is connected with drug transportation with the participation of caveolin-1. Caveolin-1, which serves as a platform or medium for signal transduction, cooperates with several signal molecules by forming a complex. Silencing of caveolin-1 and disruption of caveolae can attenuate or remove pathological damage and even engender the opposite effects in the blood-brain barrier. This review considers the role of caveolin-1 in the blood-brain barrier that may have profound implications for central nervous system disease and drug delivery through the blood-brain barrier.


Asunto(s)
Barrera Hematoencefálica/fisiología , Permeabilidad Capilar/fisiología , Caveolina 1/metabolismo , Animales , Caveolas/metabolismo , Humanos
7.
Acta Neurochir (Wien) ; 156(11): 2103-9, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25139403

RESUMEN

BACKGROUND: Recent evidence has demonstrated that rosiglitazone can attenuate cerebral vasospasm following subarachnoid hemorrhage (SAH). Some studies have shown that rosiglitazone can suppress inflammation and immune responses after SAH. However, the precise molecular mechanisms by which cerebral vasospasm is attenuated is not clear. METHODS: In this study, SAH was created using a "double hemorrhage" injection rat model. Rats were randomly divided into three groups and treated with saline (control group), untreated (SAH group), or treated with rosiglitazone. Using immunocytochemistry, hematoxylin and eosin (HE) staining, and measurement of the basilar artery, we investigated the formation of pathologic changes in the basilar artery, measured the expression of caveolin-1 and proliferating cell nuclear antigen (PCNA), and investigated the role of rosiglitazone in vascular smooth muscle cell (VSMC) proliferation in the basilar artery after SAH. RESULTS: In this study, we observed significant pathologic changes in the basilar artery after experimental SAH. The level of vasospasm gradually increased with time during the 1st week, peaked on day 7, and almost recovered on day 14. After rosiglitazone treatment, the level of vasospasm was significantly attenuated in comparison with the SAH group. Immunocytochemistry staining showed that caveolin-1 expression was significantly increased in the rosiglitazone group, compared with the SAH group. Inversely, the expression of PCNA showed a notable decrease after rosiglitazone treatment. CONCLUSIONS: The results indicate that rosiglitazone can attenuate cerebral vasospasm following SAH. Up-regulation of caveolin-1 by rosiglitazone may be a new molecular mechanism for this response, which is to inhibit proliferation of VSMCs after SAH, and this study may provide a novel insight to prevent delayed cerebral vasospasm (DCVS).


Asunto(s)
Arteria Basilar/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Hemorragia Subaracnoidea/complicaciones , Tiazolidinedionas/farmacología , Vasoconstricción/efectos de los fármacos , Vasodilatadores/farmacología , Vasoespasmo Intracraneal/etiología , Animales , Arteria Basilar/patología , Caveolina 1/efectos de los fármacos , Caveolina 1/metabolismo , Modelos Animales de Enfermedad , Inmunohistoquímica , Masculino , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/metabolismo , Antígeno Nuclear de Célula en Proliferación/efectos de los fármacos , Antígeno Nuclear de Célula en Proliferación/metabolismo , Ratas , Ratas Sprague-Dawley , Rosiglitazona , Hemorragia Subaracnoidea/patología , Hemorragia Subaracnoidea/fisiopatología , Regulación hacia Arriba , Vasoespasmo Intracraneal/fisiopatología , Vasoespasmo Intracraneal/prevención & control
8.
Acta Neurochir (Wien) ; 156(5): 941-9; discussion 949, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24463741

RESUMEN

BACKGROUND: There are complex interactions between acetylcholine (ACh), the suppressor of cytokine signaling-3 (SOCS-3), and cytokines, however, little is known about their dynamic expression or their effects on cerebral vasospasm (CVS) after subarachnoid hemorrhage (SAH). Therefore, we aimed to describe and clarify the dynamic expression of SOCS-3 and cytokines after SAH, as well as the relationships between the levels of SOCS-3, cytokines, and ACh. METHODS: The rat model of single cisterna magna injection was used to mimic acute SAH. The degree of CVS was indicated by lumen diameter and artery wall thickness under H&E staining. A semi-quantitative immunohistochemical analysis method was used to clarify the role of SOCS-3 in the CVS after SAH. We also measured the content of IL-6 and IL-10 in cerebrospinal fluid. RESULTS: We found that SOCS-3 expression levels increased rapidly within 12 h after SAH, more slowly after 12 h, and did not reach a peak within 48 h. Interleukin 6 (IL-6) levels rapidly increased within 24 h after SAH, reached a peak 24 h after SAH, and decreased slightly at 48 h. IL-10 levels increased during the first 6 h after SAH, after which this increase tapered off. ACh treatment reduced IL-6 levels and resulted in elevated levels of SOCS-3, but had no effect on IL-10 expression. Furthermore, ACh treatment relieved basilar arterial vasospasm, whereas mecamylamine pretreatment counteracted the activity of ACh. CONCLUSIONS: Taken together, these data indicate that SOCS-3 was involved in vasospasm via an IL-6- and IL-10-related mechanism, and that CVS following SAH could be reversed by the intraventricular injection of ACh.


Asunto(s)
Arteria Basilar/metabolismo , Interleucina-10/líquido cefalorraquídeo , Interleucina-6/líquido cefalorraquídeo , Hemorragia Subaracnoidea/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Vasoespasmo Intracraneal/metabolismo , Acetilcolina/farmacología , Animales , Arteria Basilar/efectos de los fármacos , Cisterna Magna , Citocinas , Inmunohistoquímica , Inyecciones Intraventriculares , Interleucina-10/metabolismo , Interleucina-6/metabolismo , Masculino , Ratas , Hemorragia Subaracnoidea/complicaciones , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/efectos de los fármacos , Vasoconstricción/efectos de los fármacos , Vasodilatadores/farmacología , Vasoespasmo Intracraneal/etiología
9.
Metab Brain Dis ; 28(1): 33-43, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23232626

RESUMEN

Regional cerebral blood flow (rCBF) in the cerebral metabolism and energy metabolism measurements can be used to assess blood flow of brain cells and to detect cell activity. Changes of rCBF in the cerebral microcirculation and energy metabolism were determined in an experimental model of subarachnoid hemorrhage (SAH) model in 56 large-eared Japanese rabbits about 12 to 16-month old. Laser Doppler flowmetry was used to detect the blood supply to brain cells. Internal carotid artery and vein blood samples were used for duplicate blood gas analysis to assess the energy metabolism of brain cells. Cerebral blood flow (CBF) was detected by single photon emission computed tomography (SPECT) perfusion imaging using Tc-99m ethyl cysteinate dimer (Tc-99m ECD) as an imaging reagent. The percentage of injected dose per gram of brain tissue was calculated and analyzed. There were positive correlations between the percentage of radionuclide injected per gram of brain tissue and rCBF supply and cerebral metabolic rate for oxygen (P < 0.05). However, there was a negative correlation between radioactivity counts per unit volume detected on the SPECT rheoencephalogram and lactic acid concentration in the homolateral internal carotid artery and vein. In summary, this study found abnormal CBF in metabolism and utilization of brain cells after SAH, and also found that deterioration of energy metabolism of brain cells played a significant role in the development of SAH. There are matched reductions in CBF and metabolism. Thus, SPECT imaging could be used as a noninvasive method to detect CBF.


Asunto(s)
Encéfalo/metabolismo , Circulación Cerebrovascular , Microcirculación , Hemorragia Subaracnoidea/fisiopatología , Animales , Metabolismo Energético , Femenino , Masculino , Consumo de Oxígeno , Conejos , Hemorragia Subaracnoidea/patología , Tomografía Computarizada de Emisión de Fotón Único
10.
Acta Neurochir (Wien) ; 155(11): 2063-70; discussion 2069-70, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23873121

RESUMEN

BACKGROUND: Akt plays an important role in cell survival, proliferation, apoptosis and other activities. It also has been involved in maintaining smooth muscle cell contraction phenotypes in vitro and in vivo. Recent studies have focused on the inhibition of Akt in acute vasospasm and neuronal apoptosis after subarachnoid hemorrhage (SAH). However, its role in delayed cerebral vasospasm (DCVS) has not been reported. METHODS: In this study, using a "two-hemorrhage" rat model of SAH, we examined the expression of p-Akt and the formation of vasospasm in the basilar arteries. To investigate the possible role of Akt in phenotypic switching, we performed immunohistochemical staining to examine expressions of SMα-actin and proliferating cell nuclear antigen (PCNA), markers of smooth muscle phenotypic switching. RESULTS: We found that the basilar arteries exhibited vasospasm after SAH and that vasospasm became most severe on day 7 after SAH. Elevated protein expression of p-Akt was detected 4 days after SAH induction, peaked on day 7, and recovered on day 21, which was in a parallel time course to the development of DCVS. Moreover, results of immunohistochemical staining revealed enhanced expression of PCNA but gradual reduction in expression of SMα-actin from day 1 to day 7 after SAH; then, the expressions of PCNA and SMα-actin gradually recovered until day 21. CONCLUSIONS: These results support a novel mechanism in which the Akt signaling pathway plays an important role in the proliferation of smooth muscle cells (SMCs) rather than inducing phenotype switching in basilar arteries, which promotes the development of DCVS after SAH.


Asunto(s)
Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/fisiología , Hemorragia Subaracnoidea/metabolismo , Vasoespasmo Intracraneal/metabolismo , Animales , Apoptosis/fisiología , Arteria Basilar/metabolismo , Proliferación Celular , Modelos Animales de Enfermedad , Masculino , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Ratas , Ratas Sprague-Dawley
11.
Neural Regen Res ; 15(4): 667-675, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31638090

RESUMEN

Tissue plasminogen activator is usually used for the treatment of acute ischemic stroke, but the role of endogenous tissue plasminogen activator in traumatic brain injury has been rarely reported. A rat model of traumatic brain injury was established by weight-drop method. The tissue plasminogen activator inhibitor neuroserpin (5 µL, 0.25 mg/mL) was injected into the lateral ventricle. Neurological function was assessed by neurological severity score. Neuronal and axonal injuries were assessed by hematoxylin-eosin staining and Bielschowsky silver staining. Protein level of endogenous tissue plasminogen activator was analyzed by western blot assay. Apoptotic marker cleaved caspase-3, neuronal marker neurofilament light chain, astrocyte marker glial fibrillary acidic protein and microglial marker Iba-1 were analyzed by immunohistochemical staining. Apoptotic cell types were detected by immunofluorescence double labeling. Apoptotic cells in the damaged cortex were detected by terminal deoxynucleotidyl transferase-mediated digoxigenin-dUTP-biotin nick-end labeling staining. Degenerating neurons in the damaged cortex were detected by Fluoro-Jade B staining. Expression of tissue plasminogen activator was increased at 6 hours, and peaked at 3 days after traumatic brain injury. Neuronal apoptosis and axonal injury were detected after traumatic brain injury. Moreover, neuroserpin enhanced neuronal apoptosis, neuronal injury and axonal injury, and activated microglia and astrocytes. Neuroserpin further deteriorated neurobehavioral function in rats with traumatic brain injury. Our findings confirm that inhibition of endogenous tissue plasminogen activator aggravates neuronal apoptosis and axonal injury after traumatic brain injury, and activates microglia and astrocytes. This study was approved by the Biomedical Ethics Committee of Animal Experiments of Shaanxi Province of China in June 2015.

12.
Neural Regen Res ; 14(10): 1726-1733, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31169190

RESUMEN

The role of vascular endothelial growth factor A in platelet adhesion in cerebral microvessels in the early stage of subarachnoid hemorrhage remains unclear. In this study, the endovascular puncture method was used to produce a rat model of subarachnoid hemorrhage. Then, 30 minutes later, vascular endothelial growth factor A antagonist anti-vascular endothelial growth factor receptor 2 antibody, 10 µg, was injected into the right ventricle. Immunohistochemistry and western blot assay were used to assess expression of vascular endothelial growth factor A, occludin and claudin-5. Immunohistochemical double labeling was conducted to examine co-expression of GP Ia-II integrin and type IV collagen. TUNEL was used to detect apoptosis in the hippocampus. Neurological score was used to assess behavioral performance. After subarachnoid hemorrhage, the expression of vascular endothelial growth factor A increased in the hippocampus, while occludin and claudin-5 expression levels decreased. Co-expression of GP Ia-II integrin and type IV collagen and the number of apoptotic cells increased, whereas behavioral performance was markedly impaired. After treatment with anti-vascular endothelial growth factor receptor 2 antibody, occludin and claudin-5 expression recovered, while co-expression of GP Ia-II integrin and type IV collagen and the number of apoptotic cells decreased. Furthermore, behavioral performance improved notably. Our findings suggest that increased vascular endothelial growth factor A levels promote platelet adhesion and contribute to early brain injury after subarachnoid hemorrhage. This study was approved by the Biomedical Ethics Committee, Medical College of Xi'an Jiaotong University, China in December 2015.

13.
Neuropsychiatr Dis Treat ; 15: 1993-2002, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31410004

RESUMEN

Background: The use of thrombolysis with tissue-plasminogen activator (t-PA) in patients with acute ischemic stroke (AIS) is limited by increased levels of matrix metalloproteinase-9 (MMP-9) and by the increased risk of hemorrhagic transformation (HT). In this study, we investigated the effects of simvastatin pretreatment on t-PA-induced MMP-9/tissue inhibitor of metalloproteinase-1 (TIMP-1) imbalance and HT aggravation in a rat AIS model. Methods: The rat AIS model was established by autologous blood emboli. Two weeks before surgery, rats were pretreated with simvastatin (60 mg/kg/d), and three hours after surgery, t-PA (10 mg/kg) was administered. MMP-9 and TIMP-1 levels in the infarcted zone and plasma were evaluated by Western blot analysis and ELISA; the level of HT was quantified by determining the hemoglobin content. RhoA activation was determined to clarify the potential effect. Results: The results suggested that pretreatment with simvastatin suppressed the increase in t-PA-induced MMP-9 levels and neutralized the elevated MMP-9/TIMP-1 ratio, but had no effect on TIMP-1 levels. Thrombolysis with t-PA after ischemia improved neurological outcome, but increased intracranial hemorrhage. Moreover, t-PA-induced HT aggravation was reduced by simvastatin pretreatment. In addition, we showed that t-PA-induced activation of RhoA was suppressed by simvastatin, and that t-PA-induced MMP-9/TIMP-1 imbalance and hemorrhage was reduced by Rho kinases (ROCK) inhibitor Y-27632. Conclusion: In this study, we showed that simvastatin pretreatment ameliorated t-PA-induced HT and MMP-9/TIMP-1 imbalance, and demonstrated that the RhoA/ROCK pathway was implicated.

14.
Zhongguo Wei Zhong Bing Ji Jiu Yi Xue ; 20(12): 740-2, 2008 Dec.
Artículo en Zh | MEDLINE | ID: mdl-19111124

RESUMEN

OBJECTIVE: To discuss the relationship between the expression of endothelin receptor A (ETRA) in brain tissue and delayed neurological deficit (DND) after subarachnoid hemorrhage (SAH). METHODS: Thirty-one Japanese rabbits were divided into SAH group (n=15), saline group (n=10), puncture group (n=3), normal control group (n=3) randomly, and they subdivided into 1 hour, 3 days, 5 days, 7 days, 10 days subgroups in both SAH group and saline group. The SAH model was successfully reproduced by intracisternal injection of autologous arterial blood twice. When the animals were sacrificed by perfusion, brain stems were harvested and prepared for immunohistochemical staining and hematoxylin and eosin (HE) staining. The pathological changes in the brain tissue were observed with light microscope. Then positive-cell counting was used to semi-quantitative analysis. RESULTS: The expression of ETRA was not detected nearly in neurons or gliocyte either in puncture group (10.6+/-3.9) or in normal group (10.0+/-2.8), while it was detected in some cells in saline group. But no significant difference was showed among the subgroups in saline group (12.3+/-2.1 lowest vs. 13.3+/-2.6 highest, P>0.05) by variance analysis. The expression of ETRA in SAH group was slightly stronger after 1 hour (20.1+/-3.5), peaked at 3 days (48.8+/-6.7), and became weak gradually after 5 days. The ETRA positive cells at 5 days, 7 days, and 10 days were 23.1+/-2.9, 22.5+/-4.2 and 19.1+/-2.8 respectively. There were significant differences among the values at different time points (P<0.05) in SAH group, and the positive cells of ETRA were obviously increased in number in SAH group at all time points compared with saline group (all P<0.05). CONCLUSION: ETRA is expressed distinctly in the brain tissue, and it may play an important role in DND pathogenesis.


Asunto(s)
Encéfalo/metabolismo , Receptor de Endotelina A/metabolismo , Hemorragia Subaracnoidea/metabolismo , Animales , Encéfalo/patología , Modelos Animales de Enfermedad , Femenino , Masculino , Conejos , Distribución Aleatoria , Hemorragia Subaracnoidea/patología
15.
Exp Ther Med ; 15(2): 1330-1338, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29434719

RESUMEN

High mobility group box 1 (HMGB1) is a classic damage-associated molecular pattern that has an important role in the pathological inflammatory response. In vitro studies have demonstrated that the Janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) signaling pathway is involved in the regulation of HMGB1 expression, mediating the inflammatory response. Therefore, the purpose of the present study was to evaluate JAK2/STAT3 pathway involvement in the subarachnoid hemorrhage (SAH)-dependent regulation of HMGB1, using an in vivo rat model. A SAH model was established by endovascular perforation. Western blotting, immunohistochemistry and immunofluorescence were used to analyze HMGB1 expression after SAH. In addition, the effects of AG490 after SAH on JAK2/STAT3 phosphorylation, HMGB1 expression and brain damage were evaluated. The results of the present study demonstrated that JAK2/STAT3 was significantly phosphorylated (P<0.05) and the total HMGB1 protein level was significantly increased (P<0.05) after SAH. In addition, the cytosolic HMGB1 level after SAH demonstrated an initial increase followed by a decrease to the control level, while the nuclear HMGB1 level after SAH demonstrated the opposite trend, with an initial decrease and subsequent increase. AG490 administration after SAH significantly inhibited JAK2/STAT3 phosphorylation (P<0.05), suppressed the expression and translocation of HMGB1, reduced cortical apoptosis, brain edema and neurological deficits. These results demonstrated the involvement of the JAK2/STAT3 pathway in HMGB1 regulation after SAH.

16.
Zhongguo Wei Zhong Bing Ji Jiu Yi Xue ; 19(6): 329-31, 2007 Jun.
Artículo en Zh | MEDLINE | ID: mdl-17577435

RESUMEN

OBJECTIVE: To discuss the effect of drainage of the cerebrospinal fluid (CSF) at acute period after aneurysmal subarachnoid hemorrhage (SAH) on the formation of hydrocephalus. METHODS: Eighty-four patients with aneurysmal SAH were randomly divided into two groups according to therapeutic regimen. Forty-two cases in specific treatment group were given intravascular embolism at the acute period of hemorrhage after a ruptured aneurysm, then CSF was drained immediately. Forty-two cases were in conventional expectant treatment group. Clinical data and incidence of hydrocephalus of specific treatment group and conventional expectant treatment group were analyzed. RESULTS: Clinical data did not show any differences between two groups, so they could be compared (all P>0.05). The incidence rate of acute hydrocephalus in specific treatment group was 7.14% (3/42 cases), that of subacute hydrocephalus was 4.76% (2/42 cases), and that of chronic hydrocephalus was 16.67% (7/42 cases). The total incidence rate was 28.57%. In conventional expectant treatment group, the incidence rate of acute hydrocephalus was 23.81% (10/42 cases), incidence of subacute hydrocephalus was 9.52% (4/42 cases), and that of chronic hydrocephalus was 35.71% (15/42 cases), and total incidence rate was 69.05%. There was significant difference between specific treatment group and conventional expectant treatment group in incidence of acute and chronic hydrocephalus (acute chi (2)=4.46, chronic chi (2)=3.94, both P<0.05), and there was no difference in subacute hydrocephalus between two groups (chi (2)=0.72, P>0.05), but significant difference was found in total incidence rate between two groups (chi (2)=13.77, P<0.01). CONCLUSION: Embolization of the intracranial aneurysm with interventional treatment at the acute hemorrhage stage (within 7 days) for the aneurysmal SAH, followed by immediate drainage of CSF can prevent hydrocephalus or alleviate hydrocephalus, and the treatment plays a significant role in the formation and development of hydrocephalus.


Asunto(s)
Drenaje , Hidrocefalia/prevención & control , Hemorragia Subaracnoidea/cirugía , Adulto , Anciano , Aneurisma Roto/complicaciones , Aneurisma Roto/terapia , Derivaciones del Líquido Cefalorraquídeo , Embolización Terapéutica , Femenino , Humanos , Hidrocefalia/líquido cefalorraquídeo , Hidrocefalia/etiología , Aneurisma Intracraneal/complicaciones , Aneurisma Intracraneal/terapia , Masculino , Persona de Mediana Edad , Hemorragia Subaracnoidea/complicaciones
17.
Zhonghua Wai Ke Za Zhi ; 45(4): 233-6, 2007 Feb 15.
Artículo en Zh | MEDLINE | ID: mdl-17502016

RESUMEN

OBJECTIVE: To discuss the early diagnostic methods and therapeutic principles of aneurysmal subarachnoid hemorrhage (SAH), and evaluate the therapeutic efficacy objectively. METHODS: Using neuro-imaging examinations combined with case history and clinical symptoms to make the early diagnosis of 96 case with aneurysmal SAH, and Guglielmi detachable microcoil (GDC) was utilized for early intracapsular embolization in the ruptured aneurysms. Efficient symptomatic treatment was done early after operation. RESULTS: All of 96 cases were early diagnosed and successfully embolized; Among them, the aneurysmal lumen was 100% occluded in 83 cases, 95% in 8 cases, 90% in 5 cases. There were 3 cases complicating with aneurysms rupture during operation, 5 cases with cerebral vasospasm. One case was affected by microcoil terminal escape after operation, 3 recurrent cases were all cured with secondary GDC embolization. There were 9 complications associated with embolization techniques and 13 cases (13.5%) occurring permanent sequelae associated with SAH. According to the Glasgow prognosis score, 77 patients got grade I, 7 grade II, 6 grade III, 3 grade IV, and 3 grade V. The mortality rate was 3.1%. CONCLUSIONS: To make early etiological diagnosis of the SAH patients, using GDC to embolize the aneurysms, and earlier efficient symptomatic treatment are important methods to improve the curative rate and reduce the mortality rate.


Asunto(s)
Embolización Terapéutica , Aneurisma Intracraneal/diagnóstico , Aneurisma Intracraneal/terapia , Hemorragia Subaracnoidea/diagnóstico , Hemorragia Subaracnoidea/terapia , Adulto , Anciano , Aneurisma Roto/complicaciones , Aneurisma Roto/diagnóstico , Aneurisma Roto/terapia , Angiografía/métodos , Diagnóstico Precoz , Femenino , Estudios de Seguimiento , Humanos , Aneurisma Intracraneal/complicaciones , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Hemorragia Subaracnoidea/etiología , Tomografía Computarizada por Rayos X , Resultado del Tratamiento
18.
Mol Med Rep ; 15(5): 3001-3010, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28339015

RESUMEN

Diffuse axonal injury (DAI) is the most common and significant pathological features of traumatic brain injury (TBI). However, there are still no effective drugs to combat the formation and progression of DAI in affected individuals. FK506, also known as tacrolimus, is an immunosuppressive drug, which is widely used in transplantation medicine for the reduction of allograft rejection. Previous studies have identified that FK506 may play an important role in the nerve protective effect of the central nervous system. In the present study, apoptosis of neuronal cells was observed following the induction of experimental DAI. The results demonstrated that it was closely related with the upregulation of death­associated protein kinase 1 (DAPK1). It was hypothesized that FK506 may inhibit the activity of DAPK1 by inhibiting calcineurin activity, which may be primarily involved in anti­apoptosis following DAI induction. Through researching the expression of nerve regeneration associated proteins (NF­H and GAP­43) following DAI, the present study provides novel data to suggest that FK506 promotes axon formation and nerve regeneration following experimental DAI. Therefore, FK506 may be a potent therapeutic for inhibiting nerve injury, as well as promoting the nerve regeneration following DAI.


Asunto(s)
Apoptosis/efectos de los fármacos , Axones/efectos de los fármacos , Lesión Axonal Difusa/tratamiento farmacológico , Tacrolimus/farmacología , Animales , Axones/metabolismo , Axones/patología , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Lesiones Traumáticas del Encéfalo/metabolismo , Lesiones Traumáticas del Encéfalo/patología , Tronco Encefálico/efectos de los fármacos , Tronco Encefálico/patología , Región CA1 Hipocampal/efectos de los fármacos , Región CA1 Hipocampal/patología , Calcineurina/efectos de los fármacos , Proteínas Quinasas Asociadas a Muerte Celular/antagonistas & inhibidores , Proteínas Quinasas Asociadas a Muerte Celular/metabolismo , Lesión Axonal Difusa/metabolismo , Lesión Axonal Difusa/patología , Proteína GAP-43/metabolismo , Masculino , Regeneración Nerviosa/efectos de los fármacos , Proteínas de Neurofilamentos/metabolismo , Ratas , Ratas Sprague-Dawley , Regulación hacia Arriba
19.
Neural Regen Res ; 11(8): 1278-84, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27651776

RESUMEN

Delayed ischemic neurologic deficit after subarachnoid hemorrhage results from loss of neural cells. Nerve growth factor and its receptor TrkA may promote regeneration of neural cells, but their expression after subarachnoid hemorrhage remains unclear. In the present study, a rat model of subarachnoid hemorrhage was established using two injections of autologous blood into the cistern magna. Immunohisto-chemical staining suggested that the expression of nerve growth factor and TrkA in the cerebral cortex and brainstem increased at 6 hours, peaked at 12 hours and decreased 1 day after induction of subarachnoid hemorrhage, whereas the expression in the hippocampus increased at 6 hours, peaked on day 1, and decreased 3 days later. Compared with those for the rats in the sham and saline groups, neurobehavioral scores decreased significantly 12 hours and 3 days after subarachnoid hemorrhage (P < 0.05). These results suggest that the expression of nerve growth factor and its receptor TrkA is dynamically changed in the rat brain and may thus participate in neuronal survival and nerve regeneration after subarachnoid hemorrhage.

20.
Brain Res ; 1639: 108-19, 2016 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-26940841

RESUMEN

OBJECTIVE: To investigate the dynamic expression of vasopressin and its potential role in rat brain tissue after experimental subarachnoid hemorrhage (SAH). METHODS: Male Sprague-Dawley rats were divided into 10min, 1h, 6h, 24h, 48h and 72h groups. The SAH model was established by endovascular puncture. ELISA and immunohistochemistry were performed to evaluate dynamic expression of vasopressin. Immunohistochemistry of GPIIb/IIIa integrin was used to assess platelet aggregation. Double immunofluorescence labeling was carried out to observe the reaction between vasopressin and platelet. Early brain injury was evaluated by apoptotic cells counting. Neurobehavioral score was performed to assess neuroprotective role of SR 49059 (a selective antagonists of vasopressin receptor). RESULTS: In peripheral blood and hypothalamus, vasopressin increased rapidly at 6h and 24h. Expression of GPIIb/IIIa integrin peaked at 24h in cortex and hippocampus. Immunofluorescence showed that vasopressin and GPIIb/IIIa integrin located at the same site. Administration of SR 49059 significantly decreased platelet aggregation and number of apoptotic cells. The neurobehavioral score was promoted significantly after the intervention. CONCLUSION: The results indicate that rapidly increased vasopressin could induce platelet aggregation and contribute to early brain injury after SAH.


Asunto(s)
Encéfalo/metabolismo , Agregación Plaquetaria/fisiología , Hemorragia Subaracnoidea/metabolismo , Vasopresinas/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Encéfalo/efectos de los fármacos , Encéfalo/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Ensayo de Inmunoadsorción Enzimática , Antagonistas de Hormonas/farmacología , Inmunohistoquímica , Indoles/farmacología , Masculino , Fármacos Neuroprotectores/farmacología , Agregación Plaquetaria/efectos de los fármacos , Pirrolidinas/farmacología , Distribución Aleatoria , Ratas Sprague-Dawley , Hemorragia Subaracnoidea/sangre , Hemorragia Subaracnoidea/tratamiento farmacológico , Hemorragia Subaracnoidea/patología , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA