Microglia

小胶质细胞
  • 文章类型: Journal Article
    脂滴(LD)是动态脂质储存细胞器。它们与新陈代谢紧密相连,可以发挥保护功能,使他们成为健康和疾病的重要参与者。大多数体内LD研究依赖于染色方法,仅提供快照。因此,我们通过用tdTomato标记内源性LD外壳蛋白perilipin2(PLIN2)来开发LD报告小鼠,在活的和固定的组织和细胞中实现无染色的荧光LD可视化。在这里,我们在标准和高脂肪饮食条件下验证了这个模型,并证明LD在健康大脑的各种细胞类型中都是高度丰富的,包括神经元,星形胶质细胞,室管膜细胞,神经干/祖细胞和小胶质细胞。此外,我们还表明,LD在大脑发育过程中非常丰富,并且可以使用胚胎切片的实时成像进行可视化。一起来看,我们的tdTom-Plin2小鼠是研究LDs及其在所有表达Plin2的组织中的生理和患病条件下的动力学的新工具。
    Lipid droplets (LDs) are dynamic lipid storage organelles. They are tightly linked to metabolism and can exert protective functions, making them important players in health and disease. Most LD studies in vivo rely on staining methods, providing only a snapshot. We therefore developed a LD-reporter mouse by labelling the endogenous LD coat protein perilipin 2 (PLIN2) with tdTomato, enabling staining-free fluorescent LD visualisation in living and fixed tissues and cells. Here we validate this model under standard and high-fat diet conditions and demonstrate that LDs are highly abundant in various cell types in the healthy brain, including neurons, astrocytes, ependymal cells, neural stem/progenitor cells and microglia. Furthermore, we also show that LDs are abundant during brain development and can be visualized using live imaging of embryonic slices. Taken together, our tdTom-Plin2 mouse serves as a novel tool to study LDs and their dynamics under both physiological and diseased conditions in all tissues expressing Plin2.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    神经免疫相互作用介导细胞间通讯并构成关键脑功能的基础。小胶质细胞,中枢神经系统驻留的巨噬细胞,通过直接的物理相互作用和分子分泌来调节大脑。一种这样的分泌因子,补体蛋白C1q,在发育和疾病模型中都有助于补体介导的突触消除,然而,大脑C1q蛋白水平在整个衰老过程中显著增加。这里,我们报道C1q与神经元核糖核蛋白(RNP)复合物以年龄依赖性方式相互作用.纯化的C1q蛋白在体外经历RNA依赖性液-液相分离(LLPS),C1q与神经元RNP复合物在体内的相互作用依赖于RNA和内吞作用。缺乏C1q的小鼠在体内神经元蛋白质合成中具有年龄特异性的改变,并且恐惧记忆消失受损。一起,我们的发现揭示了C1q的生物物理特性,该特性是RNA和年龄依赖性神经元相互作用的基础,并证明了C1q在关键的细胞内神经元过程中的作用。
    Neuroimmune interactions mediate intercellular communication and underlie critical brain functions. Microglia, CNS-resident macrophages, modulate the brain through direct physical interactions and the secretion of molecules. One such secreted factor, the complement protein C1q, contributes to complement-mediated synapse elimination in both developmental and disease models, yet brain C1q protein levels increase significantly throughout aging. Here, we report that C1q interacts with neuronal ribonucleoprotein (RNP) complexes in an age-dependent manner. Purified C1q protein undergoes RNA-dependent liquid-liquid phase separation (LLPS) in vitro, and the interaction of C1q with neuronal RNP complexes in vivo is dependent on RNA and endocytosis. Mice lacking C1q have age-specific alterations in neuronal protein synthesis in vivo and impaired fear memory extinction. Together, our findings reveal a biophysical property of C1q that underlies RNA- and age-dependent neuronal interactions and demonstrate a role of C1q in critical intracellular neuronal processes.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    背景:由于神经损伤引起的神经性疼痛(NP),通过触发炎症介质的释放破坏神经可塑性。除了神经炎症导致这种破坏的假设之外,穿心莲内酯(Andro),一种来自穿心莲的传统生物活性化合物,因其有效的抗炎特性而受到关注。然而,Andro是否可以通过调节神经炎症来改善NP仍然未知。
    目的:本研究旨在探讨Andro是否以及如何调节神经炎症和缓解NP。
    方法:使用脊神经结扎(SNL)和福尔马林大鼠模型评估Andro对NP的镇痛作用。网络药理学的结合,RNA测序,和实验验证被用来阐明Andro的镇痛作用背后的潜在机制。此外,各种技术,如功能超声,免疫组织化学,定量实时聚合酶链反应(qPCR),膜片钳,和电子显微镜被用来研究特定的神经细胞类型,神经功能,以及受Andro影响的神经可塑性变化。
    结果:网络药理学分析揭示了Andro和疼痛的共同靶标在调节疼痛相关炎症中的关键作用,包括小胶质细胞激活,神经炎症,免疫调节,和突触传递。此外,我们证实了Andro在缓解疼痛方面优于传统镇痛药物,加巴喷丁.在这些模型中,观察到Andro调节由SNL触发的血液动力学反应。转录组分析和分子对接研究表明主要组织相容性复合物II类(MHCII)基因(Db1,Da,和Bb)。电子显微镜显示突触超微结构的改善,电生理研究表明,在接受Andro治疗后,神经病大鼠的谷氨酸能传递选择性减少。系统药理学分析和生物学验证的整合共同证明了疼痛缓解的机制涉及免疫调节,增强突触可塑性,和兴奋性神经传递的精确调节。
    结论:结论:这项研究表明Andro,通过靶向MHCII基因,可能作为一个有希望的治疗候选神经性疼痛。
    BACKGROUND: Neuropathic pain (NP) due to nerve injury, disrupts neural plasticity by triggering the release of inflammatory mediators. Alongside the hypothesis that neuro-inflammation contributes to this disruption, Andrographolide (Andro), a traditional bioactive compound derived from Andrographis paniculata, has garnered attention for its potent anti-inflammatory properties. However, whether Andro could ameliorate NP by regulating neuroinflammation remains unknown.
    OBJECTIVE: This study aimed to investigate whether and how Andro regulates neuroinflammation and alleviates NP.
    METHODS: The analgesic effects of Andro on NP were evaluated using both the spinal nerve ligation (SNL) and formalin rat models. A combination of network pharmacology, RNA sequencing, and experimental validation was employed to elucidate the underlying mechanism behind Andro\'s analgesic effects. Additionally, various techniques such as functional ultrasound, immunohistochemistry, quantitative real-time polymerase chain reaction (qPCR), patch clamp, and electron microscopy were employed to investigate the specific neural cell types, neural functions, and changes in neural plasticity influenced by Andro.
    RESULTS: Network pharmacology analysis unveiled the crucial roles played by shared targets of Andro and pain in regulating pain-related inflammation, including microglia activation, neuroinflammation, immune modulation, and synaptic transmission. Furthermore, we confirmed Andro\'s superior efficacy in pain relief compared to the traditional analgesic drug, Gabapentin. In these models, Andro was observed to modulate the haemodynamic response triggered by SNL. Transcriptome analysis and molecular docking studies indicated the involvement of major histocompatibility complex class II (MHCII) genes (Db1, Da, and Bb). Electron microscopy revealed improvements in synaptic ultrastructure, and electrophysiological investigations showed a selective reduction in glutamatergic transmission in neuropathic rats after following Andro treatment. The integration of systems pharmacology analysis and biological validation collectively demonstrated that the mechanism of pain relief involves immune modulation, enhancement of synaptic plasticity, and precise regulation of excitatory neurotransmission.
    CONCLUSIONS: In conclusion, this study has demonstrated that Andro, by targeting MHCII genes, may serve as a promising therapeutic candidate for neuropathic pain.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    TMEM16F是一种钙激活磷脂杂乱酶和非选择性离子通道,这允许脂质双向移动穿过质膜。虽然TMEM16F的功能已在多种细胞类型中得到广泛表征,TMEM16F在中枢神经系统中的作用目前尚不清楚.这里,我们试图研究大脑中的TMEM16F如何参与神经变性。使用表达病理性P301S人tau的小鼠模型(PS19小鼠),我们发现缺乏TMEM16F的6至7月龄PS19小鼠的tau蛋白病变和小胶质细胞增生减少.此外,这种病理的减少可以在从神经元中去除TMEM16F的PS19小鼠中进行概括,而在这个时间点,从PS19小鼠的小胶质细胞中去除TMEM16F并没有显着影响tau蛋白病变。此外,TMEM16F在具有磷酸-tau负荷的神经元中介导异常磷脂酰丝氨酸暴露。这些研究提出了在神经元中靶向TMEM16F作为神经变性的潜在治疗的前景。
    TMEM16F is a calcium-activated phospholipid scramblase and nonselective ion channel, which allows the movement of lipids bidirectionally across the plasma membrane. While the functions of TMEM16F have been extensively characterized in multiple cell types, the role of TMEM16F in the central nervous system remains largely unknown. Here, we sought to study how TMEM16F in the brain may be involved in neurodegeneration. Using a mouse model that expresses the pathological P301S human tau (PS19 mouse), we found reduced tauopathy and microgliosis in 6- to 7-mo-old PS19 mice lacking TMEM16F. Furthermore, this reduction of pathology can be recapitulated in the PS19 mice with TMEM16F removed from neurons, while removal of TMEM16F from microglia of PS19 mice did not significantly impact tauopathy at this time point. Moreover, TMEM16F mediated aberrant phosphatidylserine exposure in neurons with phospho-tau burden. These studies raise the prospect of targeting TMEM16F in neurons as a potential treatment of neurodegeneration.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    缺血性卒中患者血糖水平升高与预后较差相关。本研究旨在探讨高血糖是否通过增加急性缺血性卒中模型的氧提取率来促进小胶质细胞凋亡。将经历大脑中动脉闭塞的C57BL/6小鼠用于评估血糖水平和神经功能。脑氧提取率(CERO2),测定氧消耗率(OCR)和脑组织氧分压(PbtO2)。探讨NOD样受体蛋白3(NLRP3)炎症小体的意义,使用NLRP3-/-小鼠,和NLRP3,caspase‑1,全长gasderminD(GSDMD‑FL)的表达水平,GSDMD‑N域(GSDMD‑N),评估IL‑1β和IL‑18。此外,Z‑YVAD‑FMK,caspase-1抑制剂,用于治疗小胶质细胞,以确定是否需要激活NLRP3炎性体来增强高血糖对焦亡的作用。提示高血糖加速急性缺血性卒中模型的脑损伤,下降的潜伏期减少和足断层的百分比证明了这一点。高血糖通过增加氧提取率来加重缺氧,正如CERO2和OCR增加所证明的那样,和响应高糖治疗的PbtO2降低。此外,通过检测caspase‑1,GSDMD‑N水平升高证实了高血糖诱导的小胶质细胞焦亡,IL‑1β和IL‑18以及GSDMD‑FL水平降低。然而,NLRP3的敲除减弱了这些作用。caspase-1的药理学抑制也降低了GSDMD-N的表达水平,小胶质细胞中的IL‑1β和IL‑18。这些结果表明,高血糖通过增加氧提取速率刺激NLRP3炎性体激活,从而导致缺血性中风后的焦度加重。
    Elevated levels of blood glucose in patients with ischemic stroke are associated with a worse prognosis. The present study aimed to explore whether hyperglycemia promotes microglial pyroptosis by increasing the oxygen extraction rate in an acute ischemic stroke model. C57BL/6 mice that underwent middle cerebral artery occlusion were used for assessment of blood glucose level and neurological function. The cerebral oxygen extraction ratio (CERO2), oxygen consumption rate (OCR) and partial pressure of brain tissue oxygen (PbtO2) were measured. To investigate the significance of the NOD‑like receptor protein 3 (NLRP3) inflammasome, NLRP3‑/‑ mice were used, and the expression levels of NLRP3, caspase‑1, full‑length gasdermin D (GSDMD‑FL), GSDMD‑N domain (GSDMD‑N), IL‑1β and IL‑18 were evaluated. In addition, Z‑YVAD‑FMK, a caspase‑1 inhibitor, was used to treat microglia to determine whether activation of the NLRP3 inflammasome was required for the enhancing effect of hyperglycemia on pyroptosis. It was revealed that hyperglycemia accelerated cerebral injury in the acute ischemic stroke model, as evidenced by decreased latency to fall and the percentage of foot fault. Hyperglycemia aggravated hypoxia by increasing the oxygen extraction rate, as evidenced by increased CERO2 and OCR, and decreased PbtO2 in response to high glucose treatment. Furthermore, hyperglycemia‑induced microglial pyroptosis was confirmed by detection of increased levels of caspase‑1, GSDMD‑N, IL‑1β and IL‑18 and a decreased level of GSDMD‑FL. However, the knockout of NLRP3 attenuated these effects. Pharmacological inhibition of caspase‑1 also reduced the expression levels of GSDMD‑N, IL‑1β and IL‑18 in microglial cells. These results suggested that hyperglycemia stimulated NLRP3 inflammasome activation by increasing the oxygen extraction rate, thus leading to the aggravation of pyroptosis following ischemic stroke.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    目的:大多数视神经脊髓炎谱系障碍(NMOSD)患者的水通道蛋白4(AQP4)特异性抗体(也称为视神经脊髓炎免疫球蛋白G抗体(NMO-IgG))呈血清阳性。尽管NMO-IgG可以诱导中枢神经系统(CNS)的病理变化,中枢神经系统和外周组织的免疫学变化在很大程度上仍然未知。我们研究了NMO-IgG是否与表达AQP4的组织结合并诱导外周组织和CNS的免疫学变化。
    方法:将C57BL/6雌性小鼠分配到NMOSD或对照组中。通过免疫染色和流式细胞术测量外周组织和中枢神经系统的病理和免疫学变化,分别。运动障碍通过开放场测试进行测量。
    结果:我们发现NMO-IgG确实与表达星形细胞和AQP4的外周组织结合,但仅在中枢神经系统中诱导神经胶质原纤维酸性蛋白和AQP4丢失。NMO-IgG诱导小胶质细胞的激活并调节小胶质细胞向经典(M1)表型的极化,但不影响外周免疫系统中的先天或适应性免疫细胞,比如巨噬细胞,中性粒细胞,Th17/Th1或产生IL-10的B细胞。此外,NMOSD小鼠在开放场中表现出明显更少的总行进距离和更高的不动时间。
    结论:我们发现注射人NMO-IgG导致星形细胞病变,中枢神经系统中的小胶质细胞活化。然而,外周组织无明显病理或免疫学改变。
    OBJECTIVE: The majority of neuromyelitis optica spectrum disorders (NMOSD) patients are seropositive for aquaporin-4 (AQP4)-specific antibodies [also named neuromyelitis optica immunoglobulin G antibodies (NMO-IgG)]. Although NMO-IgG can induce pathological changes in the central nervous system (CNS), the immunological changes in the CNS and peripheral tissue remain largely unknown. We investigated whether NMO-IgG binds to tissue expressing AQP4 and induces immunological changes in the peripheral tissue and CNS.
    METHODS: C57BL/6 female mice were assigned into an NMOSD or control group. Pathological and immunological changes in peripheral tissue and CNS were measured by immunostaining and flow cytometry, respectively. Motor impairment was measured by open-field test.
    RESULTS: We found that NMO-IgG did bind to astrocyte- and AQP4-expressing peripheral tissue, but induced glial fibrillary acidic protein and AQP4 loss only in the CNS. NMO-IgG induced the activation of microglia and modulated microglia polarization toward the classical (M1) phenotype, but did not affect innate or adaptive immune cells in the peripheral immune system, such as macrophages, neutrophils, Th17/Th1, or IL-10-producing B cells. In addition, NMOSD mice showed significantly less total distance traveled and higher immobility time in the open field.
    CONCLUSIONS: We found that injection of human NMO-IgG led to astrocytopathic lesions with microglial activation in the CNS. However, there were no significant pathological or immunological changes in the peripheral tissues.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • DOI:
    文章类型: English Abstract
    目的探讨TFEB激活剂1(TA1)促进小胶质细胞中寡聚淀粉样β(oAβ)自噬降解的机制,并探讨TA1对阿尔茨海默病(AD)小胶质细胞体外模型的治疗作用。原代小胶质细胞分别暴露于1μmol/LoAβ0、3、12和24h,构建AD小胶质细胞体外模型。为了探讨TA1的治疗效果,将原代小胶质细胞与1μmol/LoAβ和1μmol/LTA1共同治疗12h。上述细胞在固定前进一步用100nmol/L巴弗洛霉素A1处理1h。荧光探针用于检测小胶质细胞对oAβ1-42的内吞或降解。通过感染慢病毒mCherry-EGFP-LC3测定自噬通量。核TFEB强度,自噬体数量,oAβ1-42与溶酶体相关膜蛋白1(LAMP1)或微管相关蛋白轻链3(LC3)的共定位比,用免疫荧光法检测。自噬相关基因的表达,包括Lamp1、Atg5和Map1lc3b,通过qRT-PCR检测。结果表明,长时间的oAβ暴露抑制了小胶质细胞对oAβ的内吞和降解。同时,oAβ处理12h后,小胶质细胞的自噬体数量和自噬通量减少。我们进一步发现,在oAβ暴露12h后,自噬调节因子TFEB的核表达降低,导致自噬基因的减少,从而导致oAβ自噬降解的损伤。因此,长期oAβ暴露被认为可以构建AD中小胶质细胞的体外模型。TA1治疗后,细胞中TFEB的核表达明显上调。TA1处理上调自噬相关基因的表达,导致自噬通量的恢复。TA1还恢复了小胶质细胞对oAβ的内吞和降解。总之,TA1可以通过上调小胶质细胞TFEB介导的自噬来提高AD患者小胶质细胞对oAβ的清除,提示TA1是AD的潜在治疗药物。
    The purpose of the study was to investigate the mechanism of TFEB activator 1 (TA1) improving the autophagic degradation of oligomeric amyloid-β (oAβ) in microglia, and to explore the therapeutic effect of TA1 on an in vitro model of microglia in Alzheimer\'s disease (AD). Primary microglia were exposed to 1 μmol/L oAβ for 0, 3, 12, and 24 h respectively to construct the in vitro model of microglia in AD. In order to explore the therapeutic effect of TA1, primary microglia were co-treated with 1 μmol/L oAβ and 1 μmol/L TA1 for 12 h. To determine the autophagy flux, the above cells were further treated with 100 nmol/L Bafilomycin A1 for 1 h before fixation. Fluorescent probes were used to detect the endocytosis or degradation of oAβ1-42 by microglia. The autophagic flux was determined by infection of lentivirus mCherry-EGFP-LC3. The nuclear TFEB intensity, the autophagosomes number, and the colocalization ratio of oAβ1-42 with lysosome-associated membrane protein 1 (LAMP1) or microtubule-associated protein light chain 3 (LC3), were detected by immunofluorescence assay. Expressions of autophagy-related-genes, including Lamp1, Atg5, and Map1lc3b, were detected by qRT-PCR. Results showed that prolonged oAβ exposure inhibited the endocytosis and degradation of oAβ by microglia. Meanwhile, the number of autophagosomes and autophagy flux in microglia decreased after 12 h of oAβ treatment. We further found that the nuclear expression of autophagy regulator TFEB decreased after 12 h of oAβ exposure, resulting in the decrease of autophagy genes, thus leading to the damage of autophagic degradation of oAβ. Therefore, long-term oAβ exposure was considered to construct the in vitro model of microglia in AD. After TA1 treatment, the nuclear expression of TFEB in cells was obviously upregulated. TA1 treatment upregulated the expressions of autophagy-related genes, leading to the recovery of autophagy flux. TA1 also recovered the endocytosis and degradation of oAβ by microglia. In conclusion, TA1 could improve oAβ clearance by microglia in AD by upregulating microglial TFEB-mediated autophagy, suggesting TA1 as a potential therapeutic drug for AD.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    斑马鱼具有再生死亡神经元的能力,以响应视网膜损伤,穆勒胶质细胞和小胶质细胞在这一反应中起着不可或缺的作用。居民Müller胶质细胞通过重编程和经历不对称细胞分裂以产生神经元祖细胞来响应损伤,它继续增殖并分化为丢失的神经元。相比之下,小胶质细胞变得反应,吞噬死亡的细胞,并在损伤后向周围组织释放炎症信号。近年来,人们越来越关注阐明小胶质细胞在调节视网膜再生中的作用.在这里,我们证明炎症细胞因子在视网膜再生过程中差异表达,光损伤后不久,促炎细胞因子基因亚群的表达上调,而细胞因子基因不同亚群的表达随后增加。我们证明,细胞因子IL-1β和IL-10对于光损伤的视网膜中的Müller胶质细胞增殖都是必不可少的。虽然IL-1β足以在未受损的视网膜中诱导Müller胶质细胞增殖,IL-10在未受损视网膜中的表达仅诱导Müller胶质细胞表达胶质细胞标志物。一起,这些发现证明了炎性细胞因子IL-1β和IL-10对成年斑马鱼光损伤后Müller胶质细胞增殖的重要作用。
    Zebrafish possess the ability to regenerate dying neurons in response to retinal injury, with both Müller glia and microglia playing integral roles in this response. Resident Müller glia respond to damage by reprogramming and undergoing an asymmetric cell division to generate a neuronal progenitor cell, which continues to proliferate and differentiate into the lost neurons. In contrast, microglia become reactive, phagocytose dying cells, and release inflammatory signals into the surrounding tissue following damage. In recent years, there has been increased attention on elucidating the role that microglia play in regulating retinal regeneration. Here we demonstrate that inflammatory cytokines are differentially expressed during retinal regeneration, with the expression of a subset of pro-inflammatory cytokine genes upregulated shortly after light damage and the expression of a different subset of cytokine genes subsequently increasing. We demonstrate that both cytokine IL-1β and IL-10 are essential for Müller glia proliferation in the light-damaged retina. While IL-1β is sufficient to induce Müller glia proliferation in an undamaged retina, expression of IL-10 in undamaged retinas only induces Müller glia to express gliotic markers. Together, these findings demonstrate the essential role of inflammatory cytokines IL-1β and IL-10 on Müller glia proliferation following light damage in adult zebrafish.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    细胞外囊泡(EV)是用于疾病检测和监测的潜在有用的生物标志物。开发用于成像和区分来自不同细胞类型和细胞状态的小体积EV的无标记技术将具有很大的价值。这里,我们设计了一种方法,使用飞行时间二次离子质谱(ToF-SIMS)和机器学习(ML)来探索与神经炎症相关的电动汽车化学变化.与对照组相比,质谱成像能够识别和区分脂多糖(LPS)刺激后小胶质细胞释放的EV。此过程所需的样品尺寸(1微升)比其他分子分析方法(最高50微升)小得多。引人注目的是,我们看到了游离半胱氨酸硫醇(与神经炎症相关的细胞氧化应激的标志)在用LPS处理的小胶质细胞的EV中的减少,与实验测量的降低的细胞游离硫醇水平一致。这证实了ToF-SIMS和ML的协同组合是一种灵敏和有价值的技术,用于以高分辨率收集和分析来自电动汽车的分子数据。
    Extracellular vesicles (EVs) are potentially useful biomarkers for disease detection and monitoring. Development of a label-free technique for imaging and distinguishing small volumes of EVs from different cell types and cell states would be of great value. Here, we have designed a method to explore the chemical changes in EVs associated with neuroinflammation using Time-of-Flight Secondary Ion Mass spectrometry (ToF-SIMS) and machine learning (ML). Mass spectral imaging was able to identify and differentiate EVs released by microglia following lipopolysaccharide (LPS) stimulation compared to a control group. This process requires a much smaller sample size (1 µL) than other molecular analysis methods (up to 50 µL). Conspicuously, we saw a reduction in free cysteine thiols (a marker of cellular oxidative stress associated with neuroinflammation) in EVs from microglial cells treated with LPS, consistent with the reduced cellular free thiol levels measured experimentally. This validates the synergistic combination of ToF-SIMS and ML as a sensitive and valuable technique for collecting and analysing molecular data from EVs at high resolution.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:探讨一种新型蛋白激酶Cδ(PKCδ)在阿尔茨海默病(AD)神经炎症中的作用。
    方法:我们分析了AD和正常对照组的脑脊液(CSF)中的PKCδ和炎性细胞因子水平,以及它们的相关性。通过定量实时聚合酶链反应(qRT-PCR)评估PKCδ的细胞表达模式和PKCδ调节对小胶质细胞介导的神经炎症的影响。westernblot,RNA测序(RNA-seq),和免疫荧光染色。
    结果:PKCδ在AD患者脑脊液中显著升高,与细胞因子呈正相关。PKCδ主要在脑中的小胶质细胞中表达。淀粉样β(Aβ)刺激增加PKCδ的表达和分泌,这导致核因子κB(NF-κB)途径的上调和促炎细胞因子的过度产生。PKCδ的下调或抑制减弱了AD小鼠模型中Aβ诱导的小胶质细胞反应并改善了认知功能。
    结论:我们的研究将PKCδ确定为AD中小胶质细胞介导的神经炎症的潜在生物标志物和治疗靶点。
    结论:阿尔茨海默病(AD)患者脑脊液(CSF)中蛋白激酶Cδ(PKCδ)水平升高,并且与人类受试者中炎症细胞因子的升高呈正相关。PKCδ在体内主要在小胶质细胞中表达,而淀粉样β(Aβ)刺激增加PKCδ的表达和分泌,引起核因子κB(NF-κB)途径的上调和炎性细胞因子的产生。PKCδ的下调或抑制减弱小胶质细胞中Aβ增强的NF-κB信号传导和细胞因子产生,并改善AD小鼠的认知功能。PKCδ作为AD中小胶质细胞介导的神经炎症的潜在生物标志物和治疗靶标。
    BACKGROUND: To investigate the role of a novel type of protein kinase C delta (PKCδ) in the neuroinflammation of Alzheimer\'s disease (AD).
    METHODS: We analyzed PKCδ and inflammatory cytokines levels in cerebrospinal fluid (CSF) of AD and normal controls, as well as their correlations. The cellular expression pattern of PKCδ and the effects of PKCδ modulation on microglia-mediated neuroinflammation were evaluated by quantitative real-time polymerase chain reaction (qRT-PCR), western blot, RNA sequencing (RNA-seq), and immunofluorescence staining.
    RESULTS: PKCδ levels were increased dramatically in the CSF of AD patients and positively correlated with cytokines. PKCδ is expressed mainly in microglia in the brain. Amyloid beta (Aβ) stimulation increased PKCδ expression and secretion, which led to upregulation of the nuclear factor kappa B (NF-κB) pathway and overproduction of proinflammatory cytokines. Downregulation or inhibition of PKCδ attenuated Aβ-induced microglial responses and improved cognitive function in an AD mouse model.
    CONCLUSIONS: Our study identifies PKCδ as a potential biomarker and therapeutic target for microglia-mediated neuroinflammation in AD.
    CONCLUSIONS: Protein kinase C delta (PKCδ) levels increase in cerebrospinal fluid (CSF) of patients with Alzheimer\'s disease (AD), and positively correlate with elevated inflammatory cytokines in human subjects. PKCδ is expressed mainly in microglia in vivo, whereas amyloid beta (Aβ) stimulation increases PKCδ expression and secretion, causing upregulation of the nuclear factor kappa B (NF-κB) pathway and production of inflammatory cytokines. Downregulation or inhibition of PKCδ attenuates Aβ-enhanced NF-κB signaling and cytokine production in microglia and improves cognitive function in AD mice. PKCδ serves as a potential biomarker and therapeutic target for microglia-mediated neuroinflammation in AD.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号