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 187 毫秒
1.
摘要 目的:阿尔茨海默病(AD)中小胶质细胞的免疫监测和吞噬功能逐渐减弱并发生炎症激活。以往研究报道瞬时受体电位香草素1型(TRPV1)通道的激活可以缓解3xTg小鼠脑内小胶质细胞的炎症激活和吞噬功能障碍,作用机制尚不清楚。方法:首先,通过蛋白印迹法和免疫荧光实验测量3xTg小鼠大脑细胞核内组蛋白H4的12位赖氨酸乳酸化(H4K12la)的表达水平和细胞定位情况。其次,验证TRPV1的激活是否可以调控3xTg小鼠大脑细胞核内H4K12la的表达水平。最后,使用Imaris软件和流式细胞术分析TRPV1的激活对小胶质细胞炎症激活形态和生物标志物的影响。结果:蛋白印迹法显示3xTg小鼠大脑细胞核内H4K12la的表达水平上升,免疫荧光实验证明H4K12la与小胶质细胞共定位。TRPV1的激活可以减少3xTg小鼠脑内小胶质细胞中H4K12la的表达水平,缓解3xTg小鼠脑内小胶质细胞炎症激活。结论:TRPV1可以通过抑制组蛋白H4K12la表达缓解AD小胶质细胞炎症激活。  相似文献   

2.
目的 研究PDAPP转基因小鼠脑组织内反应性星形胶质细胞的活化程度。方法 通过免疫组织化学染色方法检测小鼠脑组织内反应性星形胶质细胞表达胶质纤维酸性蛋白 (GFAP)的情况 ,比较PDAPP转基因小鼠和C5 7 BL非转基因小鼠脑组织反应性星形胶质细胞的活化程度。结果 PDAPP转基因小鼠脑组织内反应性星形胶质细胞表达GFAP的水平明显高于C5 7 BL非转基因小鼠。结论 PDAPP转基因小鼠脑组织内存在明显的神经炎症反应  相似文献   

3.
目的:研究高压氧(HBO)对大鼠创伤性脑损伤(TBI)治疗效用并观察脑组织星形胶质细胞活化及胶质细胞源性神经营养因子(GDNF)和神经生长因子(NGF)表达的变化以探讨作用机制。方法:SD雄性大鼠54只,随机分为3组(n=18):假手术组、TBI组和HBO治疗组。采用Feeney法建立大鼠TBI模型,假手术组只开放骨窗,不予打击。HBO治疗组大鼠于脑损伤后6 h采用动物高压舱,以3ATA压力纯氧治疗60 min。TBI后48 h测量神经功能,然后分离脑组织,其中18只用干湿法测定脑含水量;18只脑组织用于切片,部分进行尼氏染色后作形态学观察,部分进行免疫组织化学染色,检测星形胶质细胞标记物胶质纤维酸性蛋白(GFAP)、波形蛋白(vimentin)与S100蛋白的表达;另18只大鼠取伤侧脑半球,进行Western blot分析,观察GDNF和NGF的表达。结果:HBO治疗能减轻神经功能障碍,降低脑含水量,减少海马部位神经细胞丢失,进一步激活损伤侧皮质与海马部位GFAP、vimentin与S-100阳性表达星形胶质细胞,促进损伤侧脑组织GDNF与NGF的表达。结论:HBO对创伤性脑损伤有较好治疗效果,其机制与上调GDNF和NGF的表达有关。  相似文献   

4.
摘要 目的:探讨星形胶质细胞糖原动员是否对大脑缺血再灌注损伤有神经保护作用。方法:研究构建了星形胶质细胞特异性糖原分解代谢关键酶糖原磷酸化酶(Glycogen phosphorylase, GP)过表达转基因小鼠(GFAP-GP),并通过免疫荧光染色对GP的含量进行验证。在小鼠大脑中动脉梗死/再通模型中,利用GFAP-GP小鼠促进再灌注后累积糖原的分解(糖原动员),通过三苯基氯化四氮唑(Triphenyl tetrazolium chloride, TTC)染色分析再灌注后GFAP-GP小鼠的脑梗死面积,Corner test和Grid-walking test检测再灌注后GFAP-GP小鼠的神经行为学功能。结果:GFAP-GP小鼠中GP的含量发生了明显的增加,再灌注后GFAP-GP小鼠与野生型小鼠相比,脑糖原含量明显降低,梗死明显减少,肢体感觉与运动功能明显改善。结论:星形胶质细胞糖原动员可改善大脑缺血再灌注损伤。  相似文献   

5.
摘要 目的:探讨Ca2+激活的小电导SK3钾通道在Cu2+-Aβ复合物(Cu-Aβ)所致小胶质细胞激活中的作用及下游信号通路。方法:应用Cu-Aβ激活BV2小胶质细胞,采用ELISA和Amplex Red试剂盒检测细胞培养上清中肿瘤坏死因子(TNF-α)和过氧化氢(H2O2)的含量,应用qPCR和Western blot检测钾通道mRNA和蛋白水平及相关信号通路蛋白的磷酸化。结果:(1)应用不同离子通道阻断剂以及不同亚型钾通道阻断剂预处理的实验结果表明,SK3通道可能介导了Cu-Aβ所致的小胶质细胞激活。(2)qPCR和Western blot检测结果表明,Cu-Aβ可上调小胶质细胞内SK3 mRNA和蛋白表达。(3)通过转染SK3-siRNA下调小胶质细胞内SK3表达水平,结果表明,下调SK3表达后显著抑制Cu-Aβ所致的小胶质细胞激活。(4)应用特异性信号分子阻断剂预处理的实验结果表明,PI3K/Akt信号和 ERK信号均参与了Cu-Aβ所致的小胶质细胞激活。(5)应用相关信号分子阻断剂预处理的实验结果进一步表明,在介导Cu-Aβ诱发的小胶质细胞激活过程中,SK3通道位于PI3K/Akt-ERK信号通路的上游。结论:SK3通道通过其下游的PI3K/Akt-ERK信号通路介导Cu-Aβ所致的小胶质细胞炎症反应。  相似文献   

6.
目的:探讨创伤性脑损伤(TBI)后白细胞介素-1β(IL-1β)在神经胶质细胞中的经时定位表达情况。方法:选择72只SPF级雄性小鼠分为假手术组(sham组)、TBI 6h组、TBI 12h组、TBI 1d组、TBI 4d组与TBI 7d组,每组12只,分别在脑损伤后6h、12h、1d、4d、7d时获取血清和脑组织并且制作切片。ELISA检测损伤后炎症因子IL-1β、白细胞介素-6 (IL-6)和肿瘤坏死因子-α(TNF-α)的表达。Western blot检测钙离子结合蛋白-1(IBA-1)和胶质纤维酸性蛋白(GFAP)的表达。应用免疫荧光双重染色技术观察炎症因子IL-1β在小胶质细胞和星形胶质细胞中的定位表达情况。结果:TBI后6h-7d时炎症因子IL-1β、IL-6和TNF-α的表达量均高于sham组(P0.05)。Western blot结果显示,IBA-1的表达在损伤后6h-7d时高于sham组,GFAP的表达在损伤后1d-7d时高于sham组(P0.05)。免疫荧光双重染色技术显示,6h、12h时IL-1β主要表达在小胶质细胞中,IL-1β和IBA-1共表达细胞数量多于sham组(P0.05);1d、4d、7d时IL-1β主要表达在星形胶质细胞中,IL-1β和GFAP共表达细胞数量多于sham组(P0.05)。结论:TBI诱导了胶质细胞和炎症因子的表达,其表达随脑损伤的时间而变化,IL-1β早期定位表达于小胶质细胞,后期定位表达于星形胶质细胞中。  相似文献   

7.
目的 β分泌酶1(BACE1)是阿尔茨海默病患者脑中淀粉样蛋白(Aβ)产生的关键酶。肌养蛋白聚糖 (dystroglycan,DG)帮助星形胶质细胞的终足锚定在脑血管上,形成一道支持血脑屏障的胶质界限。一项无靶标蛋白质组学研究指出BACE1可能会下调DG的表达水平。本文旨在研究BACE1能否调控DG的蛋白质水平及其可能的调控机制。方法 利用瞬时转染法在HEK-293T细胞系和原代培养的小鼠星形胶质细胞中表达目的蛋白。通过蛋白质免疫印迹分析目标蛋白质的相对水平。利用基因荧光定量和免疫共沉淀技术探索BACE1调控DG的潜在机制。结果 在HEK-293T和原代小鼠星形胶质细胞中引入BACE1会使DG β亚基(β-DG)的蛋白质水平显著降低。在HEK-293T细胞中,β-DG蛋白水平的下降依赖于BACE1的酶活性。结论 在HEK-293T细胞和小鼠星形胶质细胞中,BACE1使β-DG的蛋白质水平下降。  相似文献   

8.
目的 探讨白质消融性白质脑病中胶质细胞选择性受累而神经元受累轻微的原因。方法EIF2B5-RNAi表达载体转染至人星形胶质细胞和人神经元,检测基础状态下及内质网应激(endoplasmic reticulum stress,ERS)后细胞凋亡和活力,检测参与ERS调控的已知和未知miRNA,筛选EIF2B5-RNAi人星形胶质细胞在ERS后miRNA变化。结果EIF2B5-RNAi人神经元相比,星形胶质细胞自发凋亡及细胞活力下降。较之神经元,更多miRNA参与星形胶质细胞ERS刺激后的调控,EIF2B5-RNAi组参与调控的miRNA数目显著减少。聚类分析发现,5条已知miRNA是通路连接的关键组分。结论 人星形胶质细胞在ERS后可能更加依赖众多促细胞增殖分化的miRNA修复,而EIF2B5-RNAi人星形胶质细胞存在自发凋亡,ERS后严重减少的miRNA可能导致细胞无法存活。  相似文献   

9.
神经病理痛是由于躯体感觉系统的损伤或疾病所引起的疼痛。胶质细胞主要包括中枢神经系统的星形胶质细胞和小胶质细胞,以及外周神经系统的施旺细胞和卫星胶质细胞。胶质细胞在神经受损后被激活,发生形态变化并上调特定蛋白表达,通过与神经元的相互作用,在神经病理痛的初始和维持阶段发挥重要作用。本文综述近年来胶质细胞参与神经病理痛的研究成果。  相似文献   

10.
摘要 目的:探究间充质干细胞外泌体对脑内小胶质细胞极化和炎症因子释放的影响及其机制。方法:收集体外培养的间充质干细胞上清,超高速冷冻离心获取外泌体。采用纳米颗粒系统和透射电子显微镜分别检测外泌体粒径大小、形态结构和功能完整性。通过免疫荧光、ELISA和细胞流式等方式检测LPS刺激下,外泌体对BV2细胞的表型极化和炎症因子释放的影响。采用Western Blot法检测间充质干细胞外泌体对BV2细胞JAK1/STAT3通路活化的影响。结果:(1)间充质干细胞分泌的外泌体粒径大小主要介于40-100 nm,透射电镜显示外泌体形态呈典型膜性"杯盘"状结构;(2)流式结果表明,相比于对照组,LPS组能显著激活M1型小胶质细胞表面标志物CD11b和M2型小胶质细胞表面标志物CD206的表达,而经外泌体处理,CD11b的表达显著被抑制,CD206显著升高。同时ELISA结果证实,相比于LPS组,外泌体组分泌的促炎症因子(IL-1β、IL-6)和NO水平显著降低(P<0.05),抗炎因子(IL-10)显著升高 (P<0.05);(3)间充质干细胞外泌体显著提高了BV2细胞JAK1/STAT3通路的磷酸化水平。结论:间充质干细胞外泌体通过激活JAK1/STAT3通路有效促进脑内小胶质细胞M1型向M2型极化。  相似文献   

11.
BackgroundChronic activation of glial cells contributes to neurodegenerative diseases. Cytochrome c (CytC) is a soluble mitochondrial protein that can act as a damage-associated molecular pattern (DAMP) when released into the extracellular space from damaged cells. CytC causes immune activation of microglia in a toll-like receptor (TLR) 4-dependent manner. The effects of extracellular CytC on astrocytes are unknown. Astrocytes, which are the most abundant glial cell type in the brain, express TLR 4 and secrete inflammatory mediators; therefore, we hypothesized that extracellular CytC can interact with the TLR 4 of astrocytes inducing their release of inflammatory molecules and cytotoxins.MethodExperiments were conducted using primary human astrocytes, U118 MG human astrocytic cells, BV-2 murine microglia, and SH-SY5Y human neuronal cells.ResultsExtracellularly applied CytC increased the secretion of interleukin (IL)-1β, granulocyte-macrophage colony stimulating factor (GM-CSF) and IL-12 p70 by cultured primary human astrocytes. Anti-TLR 4 antibodies blocked the CytC-induced secretion of IL-1β and GM-CSF by astrocytes. Supernatants from CytC-activated astrocytes were toxic to human SH-SY5Y neuronal cells. We also demonstrated CytC release from damaged glial cells by measuring CytC in the supernatants of BV-2 microglia after their exposure to cytotoxic concentrations of staurosporine, amyloid-β peptides (Aβ42) and tumor necrosis factor-α.ConclusionCytC can be released into the extracellular space from damaged glial cells causing immune activation of astrocytes in a TLR 4-dependent manner.General significanceAstrocyte activation by CytC may contribute to neuroinflammation and neuronal death in neurodegenerative diseases. Astrocyte TLR 4 could be a potential therapeutic target in these diseases.  相似文献   

12.
Unveiling the mechanisms participating in the damage and repair of traumatic brain injury (TBI) is fundamental to develop new therapies. The P2Y-like GPR17 receptor has recently emerged as a sensor of damage and a key actor in lesion remodeling/repair in the rodent brain, but its role in humans is totally unknown. Here, we characterized GPR17 expression in brain specimens from seven intensive care unit TBI patients undergoing neurosurgery for contusion removal and from 28 autoptic TBI cases (and 10 control subjects of matched age and gender) of two university hospitals. In both neurosurgery and autoptic samples, GPR17 expression was strong inside the contused core and progressively declined distally according to a spatio-temporal gradient. Inside and around the core, GPR17 labeled dying neurons, reactive astrocytes, and activated microglia/macrophages. In peri-contused parenchyma, GPR17 decorated oligodendrocyte precursor cells (OPCs) some of which had proliferated, indicating re-myelination attempts. In autoptic cases, GPR17 expression positively correlated with death for intracranial complications and negatively correlated with patients’ post-traumatic survival. Data indicate lesion-specific sequential involvement of GPR17 in the (a) death of irreversibly damaged neurons, (b) activation of microglia/macrophages remodeling the lesion, and (c) activation/proliferation of multipotent parenchymal progenitors (both reactive astrocytes and OPCs) starting repair processes. Data validate GPR17 as a target for neurorepair and are particularly relevant to setting up new therapies for TBI patients.  相似文献   

13.
Glutamate-mediated excitotoxicity is known to cause secondary brain damage following stroke and traumatic brain injury (TBI). However, clinical trials using NMDA antagonists failed. Thus, glial excitatory amino acid transporters (EAATs) might be a promising target for therapeutic intervention. METHODS AND RESULTS: We examined expression of EAAT1 (GLAST) and EAAT2 (Glt-1) in 36 TBI cases by immunohistochemistry. Cortical expression of both EAATs decreased rapidly and widespread throughout the brain (in lesional, adjacent and remote areas) following TBI. In the white matter numbers of EAAT1+ parenchymal cells increased 39-fold within 24h (p<0.001) and remained markedly elevated till later stages in the lesion (90-fold, p<0.01) and in peri-lesional regions (86-fold, p<0.01). In contrast, EAAT2+ parenchymal cells and EAAT1+ or EAAT2+ perivascular cells did not increase significantly. Within the first days following TBI mainly activated microglia and thereafter mainly reactive astrocytes expressed EAAT1. Perivascular monocytes and foamy macrophages lacked EAAT1 immunoreactivity. We conclude that following TBI i) loss of cortical EAATs contributes to secondary brain damage, ii) glial EAAT1 expression reflects a potential neuroprotective function of microglia and astrocytes, iii) microglial EAAT1 expression is restricted to an early stage of activation, iv) blood-derived monocytes do not express EAAT1 and v) pharmacological modification of glial EAAT expression might further limit neuronal damage.  相似文献   

14.
Traumatic brain injury (TBI) can result in tissue alterations distant from the site of the initial injury, which can trigger pathological changes within hippocampal circuits and are thought to contribute to long-term cognitive and neuropsychological impairments. However, our understanding of secondary injury mechanisms is limited. Astrocytes play an important role in brain repair after injury and astrocyte-mediated mechanisms that are implicated in synapse development are likely important in injury-induced synapse remodeling. Our studies suggest a new role of ephrin-B1, which is known to regulate synapse development in neurons, in astrocyte-mediated synapse remodeling following TBI. Indeed, we observed a transient upregulation of ephrin-B1 immunoreactivity in hippocampal astrocytes following moderate controlled cortical impact model of TBI. The upregulation of ephrin-B1 levels in hippocampal astrocytes coincided with a decline in the number of vGlut1-positive glutamatergic input to CA1 neurons at 3 days post injury even in the absence of hippocampal neuron loss. In contrast, tamoxifen-induced ablation of ephrin-B1 from adult astrocytes in ephrin-B1loxP/yERT2-CreGFAP mice accelerated the recovery of vGlut1-positive glutamatergic input to CA1 neurons after TBI. Finally, our studies suggest that astrocytic ephrin-B1 may play an active role in injury-induced synapse remodeling through the activation of STAT3-mediated signaling in astrocytes. TBI-induced upregulation of STAT3 phosphorylation within the hippocampus was suppressed by astrocyte-specific ablation of ephrin-B1 in vivo, whereas the activation of ephrin-B1 in astrocytes triggered an increase in STAT3 phosphorylation in vitro. Thus, regulation of ephrin-B1 signaling in astrocytes may provide new therapeutic opportunities to aid functional recovery after TBI.  相似文献   

15.
Brain metabolism is thought to be maintained by neuronal-glial metabolic coupling. Glia take up glutamate from the synaptic cleft for conversion into glutamine, triggering glial glycolysis and lactate production. This lactate is shuttled into neurons and further metabolized. The origin and role of lactate in severe traumatic brain injury (TBI) remains controversial. Using a modified weight drop model of severe TBI and magnetic resonance (MR) spectroscopy with infusion of 13C-labeled glucose, lactate, and acetate, the present study investigated the possibility that neuronal-glial metabolism is uncoupled following severe TBI. Histopathology of the model showed severe brain injury with subarachnoid and hemorrhage together with glial cell activation and positive staining for Tau at 90 min post-trauma. High resolution MR spectroscopy of brain metabolites revealed significant labeling of lactate at C-3 and C-2 irrespective of the infused substrates. Increased 13C-labeled lactate in all study groups in the absence of ischemia implied activated astrocytic glycolysis and production of lactate with failure of neuronal uptake (i.e. a loss of glial sensing for glutamate). The early increase in extracellular lactate in severe TBI with the injured neurons rendered unable to pick it up probably contributes to a rapid progression toward irreversible injury and pan-necrosis. Hence, a method to detect and scavenge the excess extracellular lactate on site or early following severe TBI may be a potential primary therapeutic measure.  相似文献   

16.
Rationale: The neuroinflammation is necessary for glial group initiation and clearance of damaged cell debris after nerve injury. However, the proinflammatory polarization of excessive microglia amplifies secondary injury via enhancing cross-talk with astrocytes and exacerbating neurological destruction after spinal cord injury (SCI). The glucagon-like peptide-1 receptor (GLP-1R) agonist has been previously shown to have a neuroprotective effect in neurodegeneration, whereas its potency in microglial inflammation after SCI is still unknown.Methods: The effect and mechanism of GLP-1R activation by exendin-4 (Ex-4) were investigated in in vitro cultured glial groups and in vivo in SCI mice. Alterations in the gene expression after GLP-1R activation in inflammatory microglia were measured using mRNA sequencing. The microglial polarization, neuroinflammatory level, and astrocyte reaction were detected by using western blotting, flow cytometry, and immunofluorescence. The recoveries of neurological histology and function were also observed using imaging and ethological examinations.Results: GLP-1R activation attenuated microglia-induced neuroinflammation by reversing M1 subtypes to M2 subtypes in vitro and in vivo. In addition, activation of GLP-1R in microglia blocked production of reactive astrocytes. We also found less neuroinflammation, reactive astrocytes, corrected myelin integrity, ameliorated histology, and improved locomotor function in SCI mice treated with Ex-4. Mechanistically, we found that Ex-4 rescued the RNA expression of Arf and Rho GAP adapter protein 3 (ARAP3). Knockdown of ARAP3 in microglia reversed activation of RhoA and the pharmacological effect of Ex-4 on anti-inflammation in vitro.Conclusion: Ex-4 exhibited a previously unidentified role in reducing reactive astrocyte activation by mediation of the PI3K/ARAP3/RhoA signaling pathway, by neuroinflammation targeting microglia, and exerted a neuroprotective effect post-SCI, implying that activation of GLP-1R in microglia was a therapeutical option for treatment of neurological injury.  相似文献   

17.
In two recent papers published in Nature Neuroscience and Cell Stem Cells, Magdalena Götz and colleagues shed new light on the in vivo response of glial cells to brain injury and characterize a highly heterogeneous behavior of astrocytes to chronic and acute brain injury.Astrocytes have important roles in the brain, for example by regulating neurotransmitter clearance, controlling the formation and maintenance of synapses, and by contributing to the blood–brain barrier (BBB; for a review see [1]). In addition, astrocytes respond to acute and chronic injury by hypertrophy and induced proliferation. Notably, astrocytes in the mammalian brain represent a highly heterogeneous population and the exact cellular identity of the astrocytic response in the damaged brain remains largely unknown (for a review see [2]). Thus, live-imaging and single-cell studies are required to unravel the complexity of astrocyte behaviour and distinguish between the good and the bad effects of astrocytic activation on brain function and tissue homeostasis in response to acute and chronic injury.It is thought that astrocytes respond to injury through hypertrophy of cell bodies and processes, upregulation of the intermediate filaments GFAP and vimentin, extension of processes, proliferation and gradual overlapping of astrocytic domains (for a review see [3]). Interestingly, it is known that although some aspects of the astrocyte response to injury can be detrimental—such as the formation of a glial scar—it can also be beneficial by limiting the invasion of immune cells into the brain parenchyma [4,5,6]. However, our understanding of the response of astrocytes to injury assumes a global homogeneous response, and an unawareness of the more complex and diverse in vivo situation. Two papers from the group of Magdalena Götz, published in Nature Neuroscience and Cell Stem Cell, begin to unmask the heterogeneity of the astrocyte response to injury through in vivo live imaging after brain injury and by using multiple lesion models and comparing their effects on astroglial behaviour and properties within the injured brain.In the first study, Bardehle et al used in vivo two-photon laser-scanning microscopy to monitor individual astrocytes for up to 28 days after a stab wound to the somatosensory cortex [7]. To visualize single cells, astrocytes were labelled using different lines: GLASTCreERT2/eGFP or Confetti reporter, labelling 60–80% of all astrocytes; Aldh1l1-eGFP mice, labelling all astrocytes; and hGFAP-eGFP mice, labelling only those astrocytes with the highest GFAP expression. The authors found that most GFP+ astrocytes maintained their morphology after injury and that only subsets showed signs of hypertrophy and polarization towards the injury site. Interestingly, only a small population of astrocytes divided, all of which had their somata apposed to blood vessels (juxtavascular) and depended on proper functioning of the small RhoGTPase Cdc42 for their proliferative response. Strikingly, none of the labelled astrocytes migrated towards the lesion site, suggesting that the increase in GFAP reactivity often seen at the site of injury is not due to astrocyte migration, but rather is due to increased GFAP expression through hypertrophy, an increased number of proliferative cells and the upregulation of GFAP in cells that might not express detectable levels of GFAP before injury. Notably, migration of other glial cells (microglia and NG2+ glia) to the injury site was observed, suggesting that the migratory properties in response to injury in the brain might not be general to all glia. Thus, the contribution of activated astrocytes to the formation of a glial scar in the brain following injury might be limited and need to be reconsidered. In addition, the location of proliferating astroglial cells at juxtavascular positions, and their limited movement, suggest that these proliferating astrocytes might be a subset that is responsible for the ‘beneficial'' astrocytic response to injury by tightening the BBB, preventing the invasion of cells into the lesioned brain parenchyma. Thus, observing the glial response after brain injury in real time within their in vivo environment identified a highly selective and cell-specific astrocyte response, challenging previously held concepts of astroglial migration and massive astrocyte proliferation after injury.In the next study, Sirko et al analysed how the astroglial response varies between different types of acute or more chronic brain injury [8]. To this end the authors used four different models of injury: MCAo lesion (invasive), stab wound (invasive), APPPS1 mutation (non-invasive) and ectopic p25 activation in neurons (non-invasive). They analysed comparative data for reactive gliosis and induction of stem cell properties in activated astroglia found after brain injury (Figure 1). Interestingly, the two non-invasive, chronic lesion models induced the least response from astrocytes, with astrocytes undergoing hypertrophy but having low levels of proliferation and virtually no neurosphere-forming capacity, indicating that chronic injury in these models does not enhance astrocyte proliferation or acquisition of stem cell properties. In contrast, a much larger astrocytic response occurred in the invasive models, in which astrocytes not only underwent hypertrophy but also had a relatively high proliferative rate and formed multipotent and self-renewing neurospheres in vitro. The authors then showed that Sonic hedgehog (SHH) levels increased dramatically, but only in invasive models, and that SHH levels correlated with in vivo astrocyte proliferation rates and in vitro stem cell potential between injury conditions. By using pharmacological and genetic gain- and loss-of-function strategies, SHH signalling could indeed be identified as a crucial mediator of injury-induced acquisition of stem cell properties in astrocytes. Thus, Sirko et al identified substantial differences with respect to glial response between chronic and acute injury models and identified a molecular pathway (SHH) that at least partly accounts for enhanced astroglial response in invasive injury models.Open in a separate windowFigure 1Glial cell response, stem cell potential and extracellular Sonic hedgehog (SHH) levels vary depending on the type of brain injury. Astrocytes (yellow), NG2+ glial cells (blue) and microglia (red) reside in the uninjured intact brain, in which only NG2+ cells usually proliferate. When this tissue is studied in vitro to measure its stem cell potential, virtually no neurospheres are formed. After different types of injury, however, morphological and proliferative changes occur to all cells and their in vitro stem cell potential can be reactivated. In six-month-old APPPS1 mice, all glial cells change their morphology, with astrocytic and NG2+ hypertrophy of cell body and processes, and hypertrophy and reduction of processes in microglia. While few astrocytes proliferate, large amounts of proliferation ocurrs in both NG2+ glia and microglia. This tissue in vitro can form a few spheres that are self-renewing and multipotent, generating astrocytes, neurons and oligodendrocytes. In a model of neuronal death (CK/p25; overexpressing p25 in the postnatal forebrain), astrocytes and microglia change their morphology as described above. Astrocytes and NG2+ glia do not have any increase in proliferation rates, whereas microglia proliferate greatly. This tissue has little stem cell potential and makes only a few primary multipotent spheres. Finally, in the more invasive stab wound injury to the cortex, all glial cells become morphologically reactive, and astrocytes, NG2+ glia and microglia all proliferate in response. This tissue has the largest stem cell potential, capable of making both primary and secondary spheres with multipotent progeny. In each situation, the levels of SHH (green) can be correlated with the proliferation rates of astrocytes and in vitro stem cell potential, such that only in stab wound injury are SHH levels significantly upregulated. APPPS1, co-expresses mutated amyloid precursor protein 1 and mutated presenilin 1; NG2+, neuron-glial antigen 2.The two papers by the Götz group shed new light on the in vivo response of glial cells to brain injury and characterize a highly heterogeneous behaviour of astrocytes to chronic and acute brain injury. Surprisingly, only subsets of astrocytes proliferate or polarize, and none of them migrate towards the lesion. The juxtavascular position of proliferating astrocytes suggests that these cells might have access to the increase in SHH after invasive injury, which can regulate their division. However, it is not clear whether this proliferation is through their de-differentiation and acquisition of neural stem cell potential, or whether it is a result of a mature astrocyte division. That the astrocyte progeny remains with the original cell at the juxtavascular location suggests that they might be acting in a positive way to limit the migration of invading immune cells into the brain. Further studies on whether the increase in juxtavascular, astroglial proliferation affects the BBB permeability or decreases the number of invading cells will be important to understand this effect. If it turns out that enhanced astroglial proliferation might be generally beneficial for the injured brain, it is also tempting to speculate that for other brain injuries where the proliferation rates and SHH levels are reduced, enhanced glial proliferation in close proximity to blood vessels might help to reduce tissue damage and to improve regeneration and repair. Thus, SHH could represent a future therapeutic target to activate glial proliferation in the context of non-invasive, chronic brain injury. In any case, the acquisition of stem cell properties allowing astrocytes to form neurospheres in vitro is not directly tied to the in vivo use of these stem cell properties (for a review, see [9]). Whether the de-differentiation of astrocytes and proliferation of stem cells in vivo is beneficial or detrimental remains unclear. However, the new data have set the cellular framework for future studies to understand injury-induced astroglial stem cell characteristics in vivo and whether this in vitro potential might be unleashed for regenerative strategies in vivo.  相似文献   

18.
Astrogliosis is a process that involves morphological and biochemical changes associated with astrocyte activation in response to cell damage in the brain. The upregulation of intermediate filament proteins including glial fibrillary acidic protein (GFAP), nestin and vimentin are often used as indicators for astrogliosis. Although connexin43 (Cx43), a channel protein widely expressed in adult astrocytes, exhibits enhanced immunoreactivity in the peri-lesion region, its role in astrogliosis is still unclear. Here, we correlated the temporal and spatial expression of Cx43 to the activation of astrocytes and microglia in response to an acute needle stab wound in vivo. We found large numbers of microglia devoid of Cx43 in the needle wound at 3 days post injury (dpi) while reactive astrocytes expressing Cx43 were present in the peripheral zone surrounding the injury site. A redistribution of Cx43 to the needle site, corresponding to the increased presence of GFAP-positive reactive astrocytes in the region, was only apparent from 6 dpi and sustained until at least 15 dpi. Interestingly, the extent of microglial activation and subsequent astrogliosis in the brain of Cx43 knockout mice was significantly larger than those of wild type, suggesting that Cx43 expression limits the degree of microgliosis. Although Cx43 is not essential for astrogliosis and microglial activation induced by a needle injury, our results demonstrate that Cx43 is a useful marker for injury induced astrogliosis due to its enhanced expression specifically within a small region of the lesion for an extended period. As a channel protein, Cx43 is a potential in vivo diagnostic tool of asymptomatic brain injury.  相似文献   

19.
Microglia account for approximately 12% of the total cellular population in the mammalian brain. While neurons and astrocytes are considered the major cell types of the nervous system, microglia play a significant role in normal brain physiology by monitoring tissue for debris and pathogens and maintaining homeostasis in the parenchyma via phagocytic activity 1,2. Microglia are activated during a number of injury and disease conditions, including neurodegenerative disease, traumatic brain injury, and nervous system infection 3. Under these activating conditions, microglia increase their phagocytic activity, undergo morpohological and proliferative change, and actively secrete reactive oxygen and nitrogen species, pro-inflammatory chemokines and cytokines, often activating a paracrine or autocrine loop 4-6. As these microglial responses contribute to disease pathogenesis in neurological conditions, research focused on microglia is warranted.Due to the cellular heterogeneity of the brain, it is technically difficult to obtain sufficient microglial sample material with high purity during in vivo experiments. Current research on the neuroprotective and neurotoxic functions of microglia require a routine technical method to consistently generate pure and healthy microglia with sufficient yield for study. We present, in text and video, a protocol to isolate pure primary microglia from mixed glia cultures for a variety of downstream applications. Briefly, this technique utilizes dissociated brain tissue from neonatal rat pups to produce mixed glial cell cultures. After the mixed glial cultures reach confluency, primary microglia are mechanically isolated from the culture by a brief duration of shaking. The microglia are then plated at high purity for experimental study.The principle and protocol of this methodology have been described in the literature 7,8. Additionally, alternate methodologies to isolate primary microglia are well described 9-12. Homogenized brain tissue may be separated by density gradient centrifugation to yield primary microglia 12. However, the centrifugation is of moderate length (45 min) and may cause cellular damage and activation, as well as, cause enriched microglia and other cellular populations. Another protocol has been utilized to isolate primary microglia in a variety of organisms by prolonged (16 hr) shaking while in culture 9-11. After shaking, the media supernatant is centrifuged to isolate microglia. This longer two-step isolation method may also perturb microglial function and activation. We chiefly utilize the following microglia isolation protocol in our laboratory for a number of reasons: (1) primary microglia simulate in vivo biology more faithfully than immortalized rodent microglia cell lines, (2) nominal mechanical disruption minimizes potential cellular dysfunction or activation, and (3) sufficient yield can be obtained without passage of the mixed glial cell cultures.It is important to note that this protocol uses brain tissue from neonatal rat pups to isolate microglia and that using older rats to isolate microglia can significantly impact the yield, activation status, and functional properties of isolated microglia. There is evidence that aging is linked with microglia dysfunction, increased neuroinflammation and neurodegenerative pathologies, so previous studies have used ex vivo adult microglia to better understand the role of microglia in neurodegenerative diseases where aging is important parameter. However, ex vivo microglia cannot be kept in culture for prolonged periods of time. Therefore, while this protocol extends the life of primary microglia in culture, it should be noted that the microglia behave differently from adult microglia and in vitro studies should be carefully considered when translated to an in vivo setting.  相似文献   

20.
摘要 目的:探究炙甘草汤加减缓解神经根型颈椎病大鼠疼痛和对炎症反应的影响及机制。方法:采用免疫组织化学对接受炙甘草汤加减治疗的大鼠的脊髓组织神经元、小胶质细胞和星形胶质细胞中sPLA2的表达进行检测。使用免疫组织化学法通过测量DNA损伤标记物8-OHG检测氧化应激的程度。结果:与在神经根受压之前进行炙甘草汤加减灌胃可显著减少脊髓炎症以及DRG中的外周氧化损伤(P<0.05)。炙甘草汤加减降低了脊髓中的小胶质细胞和星形胶质细胞的激活,差异有统计学意义(P<0.05)。与第7天神经胶质激活减少的同时,脊髓sPLA2的产生亦受到抑制,神经胶质和神经元均减少,差异有统计学意义(P<0.05)。在疼痛性神经根损伤后,氧化应激标记物8-OHG几乎只存在于脊髓神经元中。在神经创伤前立即进行炙甘草汤加减治疗可防止外周DRG神经元中DNA和RNA中8-OHG增加,差异有统计学意义(P<0.05)。结论:炙甘草汤加减可以通过减少中枢和外周神经炎症和氧化应激来预防疼痛的发展。  相似文献   

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