首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 15 毫秒
1.
Glial cell line-derived neurotrophic factor (GDNF) is a transforming growth factor-beta which has shown beneficial effects in rats after acute focal cerebral ischemia (FCI). To study the effects of GDNF on chronic FCI injury in conscious rats, we used fibrin glue (GDNF-fibrin glue) and fibrin glue free (GDNF-only)-GDNF topically applied to the ischemic brain after right middle cerebral artery (MCA) ligation. Infarct brain volume and functional motor deficits were measured before and after FCI injury. After FCI injury induced by right MCA ligation, rats were randomly assigned to one of four treatment groups: (a) sham, (b) control, (c) topically applied GDNF (1 mug)-only, and (d) topically applied GDNF (1 mug)-fibrin glue. The degree of ischemic brain injury was estimated by infarct volume of right MCA territory at 4 weeks after occlusion. The functional motor deficits were quantified with rotarod test and grasping power test once a week. Topically applied GDNF-fibrin glue at infarct brain tissue after 4 weeks FCI injury significantly reduced the total infarct volume by 44.3% and 36%, respectively, compared to that of control group and GDNF-only group. The mean latencies for rats to stay on the rotarod were 55.0%, 50.3%, and 92.2% (P < 0.05 vs. control group and GDNF-only group) of baseline, respectively, in the control, GDNF-only, and GDNF-fibrin glue groups at the end of the 1st week after FCI injury but 75.3%, 67.3%, and 106.6% (P < 0.05 vs. control group and GDNF-only group) of baseline at the end of the 4th week after FCI injury. The mean values of grasping power were 78.7%, 71.7%, and 101.2% (P < 0.05 vs. control group and GDNF-only group) of baseline, respectively, in the control, GDNF-only, and GDNF-fibrin glue groups at the end of 1st week after FCI injury but 89.6%, 97.6%, and 120.7% (P < 0.05 vs. control group) of baseline at the end of 4th week after FCI injury. These results indicate that GDNF-fibrin glue not only reduced the total infarct volume after FCI injury but can also improve motor deficits after FCI injury. We concluded GDNF-fibrin glue could facilitate delivery of GDNF to the damaged brain tissue with subsequent reduction of ischemic brain injury accompanied by enhancing functional recovery in rats with chronic FCI injury.  相似文献   

2.
Zhang WR  Hayashi T  Iwai M  Nagano I  Sato K  Manabe Y  Abe K 《Brain research》2001,903(1-2):253-256
Time dependent influence of glial cell line-derived neurotrophic factor (GDNF) was examined after 90 min of transient middle cerebral artery occlusion (MCAO) in rats. Treatment with GDNF significantly reduced the infarct volume stained with 2,3,5-triphenyltetrazolium chloride (TTC) when GDNF was topically applied at 0 and 1 h of reperfusion, but became insignificant at 3 h as compared to vehicle group. The protective effect of GDNF was closely related to the significant reduction of the number of terminal deoxynucleotidyl transferase-mediated dUTP-biotin in situ nick end labeling (TUNEL) positive cells as well as immunofluorescently positive cells for active forms of caspases, especially active caspase-3 but not -9. Thus, the present study showed that topical application of GDNF significantly reduced infarct size in a time-dependent manner, while the therapeutic time window was shorter than other chemical compounds such as an NMDA receptor antagonist (MK-801) and a free radical scavenger (alpha-phenyl-tert-butyl-nitrone, PBN). The effect of GDNF was stronger in suppressing active caspase-3 than active caspase-9.  相似文献   

3.
目的 研究胶质源性神经营养因子GDNF基因转导的细胞侧脑室内移植治疗局灶性缺血性脑损伤大鼠的可行性,并探讨GDNF的神经保护作用机制.方法 体外培养SH-SY5Y细胞株,并用分子克隆技术转导入GFP-GDNF基因,使其可持续分泌绿色荧光蛋白GFP和GDNF.取75只体重150~180 g的健康雄性大鼠,分为移植缺血组、注射缺血组、缺血对照组和正常组等.在立体定向仪介导下向移植缺血组大鼠侧脑室注入转导GDNF基因的SY5Y细胞,向注射缺血组大鼠侧脑室内注射入5U/10μL的GDNF,24 h后采用线栓法对移植缺血组、注射缺血组和缺血对照组动物进行手术,建立局灶性脑缺血损伤模型(即大脑中动脉阻断,MCAO),在缺血后2 d时用Longa五分法评测行为学改变,TTC染色判断梗塞体积大小,用Western-blot法检测凋亡相关蛋白Bcl-2/Bax的表达水平.结果 在缺血损伤两天后,移植缺血组动物的肢体功能恢复效果优于缺血对照组,TTC染色亦提示移植缺血组脑梗死体积小于缺血对照组和注射缺血组(P<0.01).缺血灶周围脑组织凋亡相关蛋白Bcl-2和Bax含量均明显增高,移植缺血组Bcl-2/Bax的比值高于缺血对照组和注射缺血组.结论 GD-NF对缺血性脑损伤大鼠有神经保护作用,用GDNF基因转导的细胞移植治疗是一种可行的途径,效果优于经侧脑室直接给药.GDNF可调节凋亡相关蛋白Bcl-2/Bax的表达,这可能为其神经保护作用的机制之一.  相似文献   

4.
Adult neural stem and progenitor cells (NSPCs) are important autologous transplantation tools in regenerative medicine, as they can secrete factors that protect the ischemic brain. We investigated whether adult NSPCs genetically modified to secrete more glial cell line-derived neurotrophic factor (GDNF) could protect against transient ischemia in rats. NSPCs were harvested from the subventricular zone of adult Wistar rats and cultured for 3 weeks in the presence of epidermal growth factor. The NSPCs were treated with fibre-mutant Arg-Gly-Asp adenovirus containing the GDNF gene (NSPC-GDNF) or enhanced green fluorescent protein (EGFP) gene (NSPC-EGFP; control group). In one experiment, cultured cells were transplanted into the right ischemic boundary zone of Wistar rat brains. One week later, animals underwent 90 min of intraluminal right middle cerebral artery occlusion followed by magnetic resonance imaging and behavioural tests. The NSPC-GDNF group had higher behavioural scores and lesser infarct volume than did controls at 1, 7 and 28 days postocclusion. In the second experiment, we transplanted NSPCs 3 h after ischemic insult. Compared to controls, rats receiving NSPC-GDNF had decreased infarct volume and better behavioural assessments at 7 days post-transplant. Animals were killed on day 7 and brains were collected for GDNF ELISA and morphological assessment. Compared to controls, more GDNF was secreted, more NSPC-GDNF cells migrated toward the ischemic core and more NSPC-GDNF cells expressed immature neuronal marker. Moreover, the NSPC-GDNF group showed more effective inhibition of microglial invasion and apoptosis. These findings suggest that NSPC-GDNF may be useful in treatment of cerebral ischemia.  相似文献   

5.
The therapeutic effect of a novel RNA viral vector, Sendai virus (SeV)-mediated glial cell line-derived neurotrophic factor (GDNF) gene (SeV/GDNF), on the infarct volume, was investigated after 90 minutes of transient middle cerebral artery occlusion (tMCAO) in rats with relation to nuclear translocation of apoptosis inducing factor (AIF). The topical administration of SeV/GDNF induced high level expression of GDNF protein, which effectively reduced the infarct volume when administrated 0 and 1 hours as well after the reperfusion. Twenty-four hours after ischemia, the obvious nuclear translocation of AIF was found in neurons of peri-infarct area, which significantly reduced with administration of SeV/GDNF 0 or 1 hour after reperfusion, as well as the number of TUNEL positive cells. These results demonstrate that SeV vector-mediated gene transfer of GDNF effectively reduced ischemic infarct volume after tMCAO and extended the therapeutic time window compared with previous viral vectors, and that promoting neuronal survival of GDNF might be related to the reduction of AIF nuclear translocation, indicating the high therapeutic potency of SeV/GDNF for cerebral ischemia.  相似文献   

6.
To examine a possible protective effect of exogenous glial cell line-derived neurotrophic factor (GDNF) gene expression against ischemic brain injury, a replication-defective adenoviral vector containing GDNF gene (Ad-GDNF) was directly injected into the cerebral cortex at 1 day before 90 minutes of transient middle cerebral artery occlusion (MCAO) in rats. 2,3,5-Triphenyltetrazolium chloride staining showed that infarct volume of the Ad-GDNF-injected group at 24 hours after the transient MCAO was significantly smaller than that of vehicle- or Ad-LacZ-treated group. Enzyme-linked immunosorbent assay (ELISA) for immunoreactive GDNF demonstrated that GDNF gene products in the Ad-GDNF-injected group were higher than those of vehicle-treated group at 24 hours after transient MCAO. Immunoreactive GDNF staining was obviously detected in the cortex around the needle track just before or 24 hours after MCAO in the Ad-GDNF group, whereas no or slight GDNF staining was detected in the vehicle group. The numbers of TUNEL, immunoreactive caspase-3, and cytochrome c-positive neurons induced in the ipsilateral cerebral cortex at 24 hours after transient MCAO were markedly reduced by the Ad-GDNF group. These results suggest that the successful exogenous GDNF gene transfer ameliorates ischemic brain injury after transient MCAO in association with the reduction of apoptotic signals.  相似文献   

7.
Glial cell line‐derived neurotrophic factor (GDNF) and hepatocyte growth factor (HGF) are strong neurotrophic factors, which function as antiapoptotic factors. However, the neuroprotective effect of GDNF and HGF in ameliorating ischemic brain injury via an antiautophagic effect has not been examined. Therefore, we investigated GDNF and HGF for changes of infarct size and antiapoptotic and antiautophagic effects after transient middle cerebral artery occlusion (tMCAO) in rats. For the estimation of ischemic brain injury, the infarct size was calculated at 24 hr after tMCAO by HE staining. Terminal deoxynucleotidyl transferase‐mediated dUTP‐biotin in situ nick end labeling (TUNEL) was performed for evaluating the antiapoptotic effect. Western blot analysis of microtubule‐associated protein 1 light chain 3 (LC3) and immunofluorescence analysis of LC3 and phosphorylated mTOR/Ser2448 (p‐mTOR) were performed for evaluating the antiautophagic effect. GDNF and HGF significantly reduced infarct size after cerebral ischemia. The amounts of LC3‐I plus LC3‐II (relative to β‐tubulin) were significantly increased after tMCAO, and GDNF and HGF significantly decreased them. GDNF and HGF significantly increased p‐mTOR‐positive cells. GDNF and HGF significantly decreased the numbers of TUNEL‐, LC3‐, and LC3/TUNEL double‐positive cells. LC3/TUNEL double‐positive cells accounted for about 34.3% of LC3 plus TUNEL‐positive cells. This study suggests that the protective effects of GDNF and HGF were greatly associated with not only the antiapoptotic but also the antiautophagic effects; maybe two types of cell death can occur in the same cell at the same time, and GDNF and HGF are capable of ameliorating these two pathways. © 2010 Wiley‐Liss, Inc.  相似文献   

8.
Zhang WR  Sato K  Iwai M  Nagano I  Manabe Y  Abe K 《Brain research》2002,947(1):140-145
The time dependent influence of adenovirus-mediated glial cell line-derived neurotrophic factor (GDNF) gene (Ad-GDNF) was examined after 90 min of transient middle cerebral artery occlusion (MCAO) in rats. Treatment with Ad-GDNF significantly reduced the infarct volume when immediately administered after the reperfusion, but became insignificant when administered at 1 h after the reperfusion as were the cases treated with vehicle- and adenoviral vector containing the E. coli lacZ gene (Ad-LacZ)-treated groups. The protective effect of GDNF was related to the significant reduction of the number of TUNEL positive cells as well as immunohistochemical positive cells for active caspase-3 but not -9. These results showed that exogenous GDNF gene transfer successfully reduced the infarct size in a time-dependant manner by suppressing active caspase-3 but not active caspase-9. However, the therapeutic time window was shorter than the effect of GDNF protein itself previously reported.  相似文献   

9.
Direct intracerebral administration of glial cell line-derived neurotrophic factor (GDNF) is neuroprotective against ischemia-induced cerebral injury. Utilizing viral vectors to deliver and express therapeutic genes presents an opportunity to produce GDNF within localized regions of an evolving infarct. We investigated whether a herpes simplex virus (HSV) amplicon-based vector encoding GDNF (HSVgdnf) would protect neurons against ischemic injury. In primary cortical cultures HSVgdnf reduced oxidant-induced injury compared to the control vector HSVlac. To test protective effects in vivo, HSVgdnf or HSVlac was injected into the cerebral cortex 4 days prior to, or 3 days, after a 60-min unilateral occlusion of the middle cerebral artery. Control stroke animals developed bradykinesia and motor asymmetry; pretreatment with HSVgdnf significantly reduced such motor deficits. Animals receiving HSVlac or HSVgdnf after the ischemic insult did not exhibit any behavioral improvement. Histological analyses performed 1 month after stroke revealed a reduction in ischemic tissue loss in rats pretreated with HSVgdnf. Similarly, these animals exhibited less immunostaining for glial fibrillary acidic protein and the apoptotic marker caspase-3. Taken together, our data indicate that HSVgdnf pretreatment provides protection against cerebral ischemia and supports the utilization of the HSV amplicon for therapeutic delivery of trophic factors to the CNS.  相似文献   

10.
Hepatocyte growth factor (HGF) and glial cell line-derived neurotrophic factor (GDNF) are strong neurotrophic factors. However, their potentials in neurogenesis, angiogenesis, synaptogenesis, and antifibrosis have not been compared. Therefore, we investigated these effects of HGF and GDNF in cerebral ischemia in the rat. Wistar rats were subjected to 90 min of transient middle cerebral artery occlusion (tMCAO). Immediately after reperfusion, HGF or GDNF was given by topical application. BrdU was injected intraperitoneally twice daily 1, 2, and 3 days after tMCAO. On 14 day, we histologically evaluated infarct volume, antiapoptotic effect, neurogenesis, angiogenesis, synaptogenesis, and antifibrosis. Both HGF and GDNF significantly reduced infarct size and the number of TUNEL-positive cells, but only HGF significantly increased the number of BrdU-positive cells in the subventricular zone, and 5'-bromo-2'-deoxyuridine -positive cells differentiated into mature neurons on the ischemic side. Enhancement of angiogenesis and synaptogenesis at the ischemic boundary zone was also observed only in HGF-treated rats. HGF significantly decreased the glial scar formation and scar thickness of the brain pia mater after tMCAO, but GDNF did not. Our study shows that both HGF and GDNF had significant neurotrophic effects, but only HGF can promote the neurogenesis, angiogenesis, and synaptogenesis and inhibit fibrotic change in brains after tMCAO.  相似文献   

11.
Phagocyte survival and function are enhanced by GM-CSF and G-CSF. The production of both CSFs can be induced in mesenchymal cells by tumor necrosis factor-α (TNF-α) and interleukin-1 (IL-1). We have recently demonstrated that IL-1α and β induced the production of GM-CSF and G-CSF by two human astroglial cell lines. In the present study, we examined the effects TNF-α on the production of GM-CSF and G-CSF by U87MG, a human astroglial cell line that constitutively expresses GM-CSF and G-CSF, and U373MG, a second human astroglial cell line does not produce CSF. We demonstrate that U87MG can be induced to increase its production of GM-CSF and G-CSF by exposure to TNF-α while U373MG is induced to produce GM-CSF but not G-CSF. These responses, measured by accumulation of elevated levels of CSF protein and mRNA, are rapid and sensitive. The implications of these findings to the immunopathogenesis of central nervous system infections are discussed.  相似文献   

12.
13.
目的研究P2Y1受体对缺血时星形胶质细胞产生胶质原纤维酸性蛋白(glial fibrillary acidic protein,GFAP)及胶质细胞源性神经营养因子(glial cell line—derived neurotrophic factor,GDNF)的影响及其相关信号通路。方法分别利用右侧大腑中动脉线拴阻塞及培养细胞缺氧无营养后恢复正常培养,造成体内、外缺血再灌注模型。用免疫荧光标记、实时定量RT—PCR、Western blotting、酶联免疫吸附试验观察P2Y1受体、GDNF定位,检测GFAP、GDNF及信号分子的表达变化。结果与单纯性缺血组比较,用选择性拈抗剂MRS2179阻断P2Y1受体后,可使体内、外星形胶质细胞产生的GFAP减少,同时使其产生GDNF增加。体外缺氧无营养并阻断P2Y1受体后:可使磷酸化蛋白激酶B(Akt)及cAMP反应元件结合蛋(cAMP response element binding protein,CREB)升高,而使磷酸化JAK2及STAT3(Ser727)降低;JAK2的抑制剂AG490在降低磷酸化STAT3(Ser727)的同时也降低GFAP表达水平;PI3-K的抑制剂LY294002可降低磷酸化的Akt及CREB;MEK1/2抑制剂U0126可同时降低磷酸化的JAK2、STAT3 (Ser727)、Akt及CREB。结论P2Y1受体参与短时性缺血时星形胶质细胞GFAP及GDNF的产生过程,相关信号途径分别为JAK2/STAT3和P13-K/AKT/CREB,并且两条途径存在串话。  相似文献   

14.
In the present study, we have studied the expression of glial cell line-derived neurotrophic factor (GDNF) and its receptors Ret, GFRα1, and GFRα2 in the retino-tectal system before and after optic nerve transection. Using retrograde neuronal tracing in combination with in situ hybridization, we found that Ret and GFRα1 are expressed by 13–14% of the retinal ganglion cells (RGCs). These Ret-expressing RGCs could not be identified as belonging to any particular of the RGA, RGB, and RGC sub types. Ret is co-expressed with the brain-derived neurotrophic factor receptor TrkB in these RGCs. Optic nerve transection resulted in reduced Ret mRNA levels in retina, while the levels of GDNF, GFRα1, and 2 mRNA increased. Administration of GDNF protein supported the axotomized RGCs. Analysis of normal superior colliculus (SC) did not show any expression of GDNF mRNA, yet GDNF mRNA levels in SC increased after injury. Together, these findings identify a portion of RGCs as being possible targets for pharmacological treatment with GDNF in a direct mode of action. The absence of detectable GDNF mRNA in normal SC questions the role for GDNF as being a target-derived factor produced in the SC for adult RGCs. The results support a function for GDNF locally in the retina and as part of an injury-induced system that may act to enhance neuroprotective and neuroregenerative responses both to endogenous GDNF ligands and those administered exogenously.  相似文献   

15.
背景:近年来,已有实验证实经颅直接给予胶质细胞源性神经营养因子(glial cell-derived neurotrophic factor,GDNF)或携带GDNF基因病毒载体有显著减少脑梗死体积、促进神经功能恢复的疗效,但其治疗方法有创,临床应用有限。 目的:观察GDNF基因转染的人脐血CD34+细胞经静脉移植对自发性高血压大鼠脑梗死的疗效,并探讨其机制。 方法:分离人脐血CD34+细胞,脂质体方法转染pEGFP-GDNF质粒和pEGFP空载体至CD34+细胞制备pEGFP-GDNF-CD34+和pEGFP-CD34+细胞;制备60只雄性自发性高血压大鼠大脑中动脉栓死模型并随机分为3组:pEGFP-GDNF-CD34+细胞移植组(基因细胞组)、pEGFP-CD34+细胞移植组(单纯细胞组)、生理盐水组。改良神经功能损害评分评价神经功能恢复状况;图像分析法观察TTC染色脑梗死体积;酶联免疫法检测细胞培养液与脑组织匀浆GDNF水平,荧光显微镜及免疫组织化学检测绿色荧光蛋白标记CD34+细胞及其人神经胶质纤维酸性蛋白和人神经元核抗原表达。 结果与结论:经静脉移植pEGFP-GDNF-CD34+细胞向自发性高血压大鼠脑缺血区域迁移、存活、并向神经细胞定向分化,促进神经功能恢复。GDNF基因转染CD34+细胞在脑组织存活、向神经细胞分化及对大脑神经功能保护作用优于CD34+细胞。脑组织GDNF水平可能是GDNF基因转染CD34+细胞和单纯CD34+细胞移植治疗自发性高血压大鼠局灶性脑缺血疗效差异机制之一。  相似文献   

16.
目的 确立更规范统一的制作大鼠局灶性脑缺血模型方法,使脑梗死体积更加稳定。方法 对24只大鼠使用液态硅胶涂层尼龙线栓塞大脑中动脉,分别缺血l,2,6和24h后再灌注24h,监测缺血侧局部脑血流,测定脑梗死体积及脑水肿程度。结果 缺血后所有大鼠局部脑血流均降到缺血前基值的25%以下,TTC染色显示所有动物在缺血侧皮质和尾状核均有明显的梗死灶和缺血,缺血1h组梗死体积与缺血2h以上组有显性差异,缺血2h以上各组之间梗死体积无显性差异;各组脑水肿程度无显性差异。结论 应用硅胶涂层尼龙线结合局部脑血流监测,缺血2h以上同时予以血流监测,可制作梗死体积稳定的大鼠局灶性脑缺血模型。  相似文献   

17.
Cerebral ischemia triggers secondary ischemia/reperfusion injury and endoplasmic reticulum stress initiates cell apoptosis. However, the regulatory mechanism of the signaling pathway remains unclear. We hypothesize that the regulatory mechanisms are mediated by the protein kinase-like endoplasmic reticulum kinase/eukaryotic initiation factor 2α in the endoplasmic reticulum stress signaling pathway. To verify this hypothesis, we occluded the middle cerebral artery in rats to establish focal cerebral ischemia/reperfusion model. Results showed that the expression levels of protein kinase-like endoplasmic reticulum kinase and caspase-3, as well as the phosphorylation of eukaryotic initiation factor 2α, were increased after ischemia/reperfusion. Administration of atorvastatin decreased the expression of protein kinase-like endoplasmic reticulum kinase, caspase-3 and phosphorylated eukaryotic initiation factor 2α, reduced the infarct volume and improved ultrastructure in the rat brain. After salubrinal, the specific inhibitor of phosphorylated eukaryotic initiation factor 2α was given into the rats intragastrically, the expression levels of caspase-3 and phosphorylated eukaryotic initiation factor 2α in the were decreased, a reduction of the infarct volume and less ultrastructural damage were observed than the untreated, ischemic brain. However, salubrinal had no impact on the expression of protein kinase-like endoplasmic reticulum kinase. Experimental findings indicate that atorvastatin inhibits endoplasmic reticulum stress and exerts neuroprotective effects. The underlying mechanisms of attenuating ischemia/reperfusion injury are associated with the protein kinase-like endoplasmic reticulum kinase/eukaryotic initiation factor 2α/caspase-3 pathway.  相似文献   

18.
Transplantation of human umbilical cord blood cells (HUCBC) produces reliable behavioral and morphological improvements in animal models of stroke. However, the mechanisms of action still have not been fully elucidated. The aim of the present study is the evaluation of potential neuroprotective effects produced by HUCBC in terms of reduced infarct volume and caspase-3-dependent cell death. Permanent middle cerebral artery occlusion was induced in 90 spontaneously hypertensive rats. The animals were randomly assigned to the control group (n = 49) or the verum group (n = 41). The cell suspension (8 × 106 HUCBC per kilogram bodyweight) or vehicle solution was intravenously administered 24 h after stroke onset. Fifty subjects (n = 25/25) were sacrificed after 25, 48, 72 and 96 h, and brain specimens were removed for immunohistochemistry for MAP2, cleaved caspase-3 (casp3) and GFAP. Another 42 animals (n = 26/16) were sacrificed after 0, 6, 24, 36 and 48 h and their brains processed for quantitative PCR for casp3 and survivin. The infarct volume remained stable over the entire experimental period. However, cleaved casp3 activity increased significantly in the infarct border zone within the same time frame. Numerous cleaved casp3-positive cells were colocalized with the astrocytic marker GFAP, whereas cleavage of neuronal casp3 was observed rarely. Neither the infarct volume nor casp3 activity was significantly affected by cell transplantation. Delayed systemic transplantation of HUCBC failed to produce neuroprotective effects in a permanent stroke model using premorbid subjects.  相似文献   

19.
Inflammation plays a key role in ischemic stroke pathophysiology: microglial/macrophage cells and type-1 helper cells (Th1) seem deleterious, while type-2 helper cells (Th2) and regulatory T cells (Treg) seem protective. CD4 Th0 differentiation is modulated by microglial cytokine secretion. Glatiramer Acetate (GA) is an immunomodulatory drug that has been approved for the treatment of human multiple sclerosis by means of a number of mechanisms: reduced microglial activation and pro-inflammatory cytokine production, Th0 differentiation shifting from Th2 to Th2 and Treg with anti-inflammatory cytokine production and increased neurogenesis.We induced permanent (pMCAo) or transient middle cerebral artery occlusion (tMCAo) and GA (2 mg) or vehicle was injected subcutaneously immediately after cerebral ischemia. Mice were sacrificed at D3 to measure neurological deficit, infarct volume, microglial cell density and qPCR of TNFα and IL-1β (pro-inflammatory microglial cytokines), IFNγ (Th2 cytokine), IL-4 (Th2 cytokine), TGFβ and IL-10 (Treg cytokines), and at D7 to evaluate neurological deficit, infarct volume and neurogenesis assessment.We showed that in GA-treated pMCAo mice, infarct volume, microglial cell density and cytokine secretion were not significantly modified at D3, while neurogenesis was enhanced at D7 without significant infarct volume reduction. In GA-treated tMCAo mice, microglial pro-inflammatory cytokines IL-1β and TNFα were significantly decreased without modification of microglial/macrophage cell density, cytokine secretion, neurological deficit or infarct volume at D3, or modification of neurological deficit, neurogenesis or infarct volume at D7.In conclusion, Glatiramer Acetate administered after cerebral ischemia does not reduce infarct volume or improve neurological deficit in mice despite a significant increase in neurogenesis in pMCAo and a microglial pro-inflammatory cytokine reduction in tMCAo.  相似文献   

20.
设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号