neuronal autophagy

  • 文章类型: Journal Article
    背景:益气活血汤(YQHXD),中药配方,在脑出血(ICH)的临床治疗中已经证明了十多年的疗效。然而,YQHXD能够穿透脑组织的精确药物治疗化合物和YQHXD的药理学基础仍然不明确.
    方法:采用超高效液相色谱-四极杆飞行时间质谱分析大鼠脑内YQHXD的活性成分。潜在的目标,网络药理学预测了YQHXD改善ICH损伤的途径和生物学进展。此外,胶原酶诱导的ICH大鼠模型,应用血红素暴露和分子对接的原代皮质神经元验证了YQHXD的分子机制。
    结果:在脑内鉴定出YQHXD的11种活性成分。使用基因本体论(GO)和京都基因和基因组百科全书(KEGG)数据库,我们的研究集中在自噬和BDNF/TrkB信号通路在药理学背景下的作用.药理结果表明,YQHXD减轻了神经功能障碍,脑含水量,脑肿胀,和ICH引起的病理损伤。同时,YQHXD在体内抑制自噬内流和自噬体,并在体内和体外调节皮质神经元自噬和TrkB/BDNF通路。随后,N-乙酰血清素(NAS),一种选择性的TrkB激动剂,用于证实BDNF/TrkB途径在该过程中的重要性。NAS和YQHXD的组合没有进一步增强YQHXD在ICH大鼠中的保护功效。此外,分子对接分析结果显示,YQHXD的9种化合物对TrkB具有潜在的调节作用。
    结论:同侧神经元自噬和BDNF/TrkB通路在ICH后72h被激活。YQHXD有效抵抗ICH诱导的损伤,与通过BDNF/TrkB通路抑制同侧神经元自噬有关。这项研究提供了在ICH治疗背景下中药治疗机制的新见解。
    BACKGROUND: Yi-Qi-Huo-Xue Decoction (YQHXD), a traditional Chinese medicine formula, has demonstrated efficacy in the clinical treatment of intracerebral hemorrhage (ICH) for over a decade. Nevertheless, the precise pharmacotherapeutic compounds of YQHXD capable of penetrating into cerebral tissue and the pharmacological underpinnings of YQHXD remain ambiguous.
    METHODS: The active components of YQHXD in rat brains was analyzed by ultra-performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry. The potential targets, pathways and biological progresses of YQHXD ameliorating ICH induced injury was predicted by network pharmacology. Moreover, collagenase-induced ICH rat model, primary cortex neurons exposed to hemin and molecular docking were applied to validate the molecular mechanisms of YQHXD.
    RESULTS: Eleven active components of YQHXD were identified within the brains. Employing the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) databases, our investigation concentrated on the roles of autophagy and the BDNF/TrkB signaling pathway in the pharmacological context. The pharmacological results revealed that YQHXD alleviated neurological dysfunction, brain water content, brain swelling, and pathological injury caused by ICH. Meanwhile, YQHXD inhibited autophagy influx and autophagosome in vivo, and regulated cortex neuronal autophagy and TrkB/BDNF pathway both in vivo and in vitro. Subsequently, N-acetyl serotonin (NAS), a selective TrkB agonist, was employed to corroborate the significance of the BDNF/TrkB pathway in this process. The combination of NAS and YQHXD did not further enhance the protective efficacy of YQHXD in ICH rats. Additionally, outcomes of molecular docking analysis revealed that nine compounds of YQHXD exhibited potential regulatory effects on TrkB.
    CONCLUSIONS: Ipsilateral neuronal autophagy and BDNF/TrkB pathway were activated 72 h after ICH. YQHXD effectively resisted injury induced by ICH, which was related with suppression of ipsilateral neuronal autophagy via BDNF/TrkB pathway. This study provides novel insights into the therapeutic mechanisms of traditional Chinese medicine in the context of ICH treatment.
    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:G蛋白偶联受体40(GPR40)是阿尔茨海默病(AD)的潜在药物靶标,其激动剂GW9508通过静脉给药改善认知障碍。
    目的:本研究旨在研究GW9508对链脲佐菌素(STZ)诱导的AD小鼠认知功能障碍的影响。
    方法:将70只雄性ICR小鼠随机分为7组:载体假手术组,模型,多奈哌齐,GW9508-L,GW9508-M,GW9508-H,和GW1100+GW9508-H组,并分别在第1天和第3天通过侧脑室内注射每天一次(上午9:00)给予载体(人工脑脊液[aCSF])或STZ(在载体中3mg/kg)。经过两周的恢复,所有小鼠均给予药物治疗。行为实验用于测试小鼠的识别和空间记忆,而蛋白质印迹等分子生物学实验,ELISA,和Nissl染色检测信号通路的相应变化。
    结果:腹膜内施用GW9508可预防STZ诱导的认知障碍,并降低血浆和大脑中p-tau和Aβ1-42的水平。GW9508上调肠脑肽如PYY的表达,CCK,IGF-1和GLP-1均在血液循环和脑中通过激活Akt/mTOR信号通路下调自噬相关蛋白的表达水平。同时,GPR40拮抗剂GW1100明显逆转GW9508的治疗效果。
    结论:GW9508的外周给药具有神经保护作用,它可能是一种有希望的AD治疗方法。GW9508的神经保护机制是基于促进肠脑肽分泌,激活Akt/mTOR信号通路,调节神经元自噬。
    BACKGROUND: G protein-coupled receptor 40 (GPR40) is a potential drug target for Alzheimer\'s disease (AD), and its agonist GW9508 ameliorates cognitive impairment by intravenous administration.
    OBJECTIVE: The present study was conducted to investigate the efficacy of GW9508 administered peripherally on cognitive dysfunction in streptozotocin (STZ)-induced AD mice.
    METHODS: Seventy male ICR mice were randomly divided into seven groups: vehicle sham group, model, Donepezil, GW9508-L, GW9508-M, GW9508-H, and GW1100 + GW9508-H groups, and administered either vehicle (artificial cerebrospinal fluid [aCSF]) or STZ (3 mg/kg in the vehicle) once a day (9:00 a.m.) by intracerebroventricular injection bilaterally on day 1 and day 3, respectively. After 2 weeks of recovery, all mice were given drug treatment. Behavioral experiments were applied to test the recognition and spatial memory of mice, while molecular biology experiments such as Western blot, ELISA, and Nissl staining were used to detect the corresponding changes of signaling pathways.
    RESULTS: Intraperitoneal administration of GW9508 prevented STZ-induced cognitive impairment as well as decreased the level of p-tau and Aβ1-42 in plasma and brain. GW9508 upregulated the expression of gut-brain peptides like PYY, CCK, IGF-1, and GLP-1 both in blood circulation and brain and downregulated the expression level of autophagy-related proteins through activating Akt/mTOR signaling pathway. Meanwhile, the treatment effect of GW9508 was reversed by GPR40 antagonist GW1100 significantly.
    CONCLUSIONS: Peripheral administration of GW9508 exhibits neuroprotective effects, and it could be a promising therapy for AD. The neuroprotective mechanism of GW9508 was based on promoting gut-brain peptide secretion, activating Akt/mTOR signal pathway, and regulating neuronal autophagy.
    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
    神经元CX3CL1通过与小胶质细胞上表达的受体CX3CR1结合抑制小胶质细胞炎症。脑缺血显著激活神经元自噬,而自噬神经元中CX3CL1的表达则相反下调,加剧了小胶质细胞炎症.因此,这项研究旨在研究缺血激活的小胶质细胞炎症是否可以通过减弱神经元自噬促进CX3CL1表达来抑制.免疫荧光显示自噬主要发生在神经元中,但很少发生在小胶质细胞中。蛋白质印迹和免疫荧光表明,减弱HT22自噬显著增加其CX3CL1表达,随后减轻BV2介导的炎症反应,炎症因子NF-κB-p65、IL-6、IL-1β水平降低,TNF-α,和PGE2。同时,CCK-8,Nissl染色,和FJC染色显示,CX3CL1抑制的小胶质细胞炎症极大地减轻了OGD(氧糖原剥夺)产生的神经元损伤。相反,升高HT22自噬显著降低其CX3CL1表达,从而恶化小胶质细胞炎症和神经元损伤。我们的数据表明,减弱神经元自噬可能是减轻缺血性卒中后小胶质细胞炎症损伤的有效方法。
    Neuronal CX3CL1 suppressed microglial inflammation by binding to its receptor CX3CR1 expressed on microglia. Neuronal autophagy was prominently activated by cerebral ischemia, whereas CX3CL1 expression in autophagic neurons was conversely down-regulated to exacerbate microglial inflammation. Accordingly, this study was meant to investigate whether ischemia-activated microglial inflammation could be repressed by promoting CX3CL1 expression via the attenuation of neuronal autophagy. Immunofluorescence showed that autophagy predominantly occurred in neurons but barely in microglia. Western blot and immunofluorescence demonstrated that attenuating HT22 autophagy significantly increased its CX3CL1 expression and subsequently mitigated the BV2-mediated inflammatory responses, as indicated by decreased inflammatory factors of NF-κB-p65, IL-6, IL-1β, TNF-α, and PGE2. Meanwhile, CCK-8, Nissl staining, and FJC staining showed that an OGD (Oxygen-glycogen deprivation)-created neuronal injury was greatly alleviated by CX3CL1-suppressed microglial inflammation. Contrarily, elevating HT22 autophagy markedly decreased its CX3CL1 expression, which consequently worsened microglial inflammation and the neuronal injury. Our data suggests that attenuating neuronal autophagy may be an effective method to alleviate a microglial inflammatory injury after an ischemic stroke.
    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
    巨自噬/自噬是进化上高度保守的分解代谢过程,对于清除胞浆内容物以维持细胞稳态和存活很重要。最近的发现指出自噬在大脑功能中的关键作用,不仅通过保持神经元健康,尤其是通过控制神经元发育和功能的不同方面。与此相符,自噬相关基因的突变与神经发育障碍(NDD)的各种关键特征和症状有关,包括自闭症,小头/大头畸形,和癫痫。然而,自噬相关基因突变引起的NDD组相对较小.相当比例的NDD与编码调节基因表达的表观遗传调节蛋白的基因突变有关。所谓的色素病。有趣的是,一些NDD连锁的色素病变基因已被证明可以调节自噬相关基因,尽管在非神经元环境中。从这些研究中可以明显看出,自噬相关基因的严格转录调节对于控制自噬活性至关重要。这开启了一个令人兴奋的可能性,即异常的自噬调节可能会在表观遗传调节受到干扰的NDD中导致神经系统受损。我们在这里总结了NDD相关的色素病变基因,这些基因已知可以调节自噬相关基因的转录调节。因此,我们希望强调自噬是NDD相关色素病变的候选关键中枢机制.缩写:ADNP:活性依赖性神经保护者同源异型框;ASD:自闭症谱系障碍;ATG:AuttophaGy相关;CpG:胞嘧啶鸟嘌呤二核苷酸;DNMT:DNA甲基转移酶;EHMT:全色组蛋白赖氨酸甲基转移酶;EP300:E1A结合蛋白p300;EZH2:zeste2多梳抑制复合物2的增强子;H3Ktristone4me3-9di-,或三甲基化;H3K27me2/3:组蛋白3赖氨酸27二-,或三甲基化;hiPSCs:人类诱导的多能干细胞;HSP:遗传性痉挛性截瘫;ID:智力残疾;KANSL1:KAT8调节NSL复合物亚基1;KAT8:赖氨酸乙酰转移酶8;KDM1A/LSD1:赖氨酸脱甲基酶1A;MAP1LC3B:微管相关蛋白1;NAPSEPHT-1;YAPRapamycamycin1
    Macroautophagy/autophagy is an evolutionarily highly conserved catabolic process that is important for the clearance of cytosolic contents to maintain cellular homeostasis and survival. Recent findings point toward a critical role for autophagy in brain function, not only by preserving neuronal health, but especially by controlling different aspects of neuronal development and functioning. In line with this, mutations in autophagy-related genes are linked to various key characteristics and symptoms of neurodevelopmental disorders (NDDs), including autism, micro-/macrocephaly, and epilepsy. However, the group of NDDs caused by mutations in autophagy-related genes is relatively small. A significant proportion of NDDs are associated with mutations in genes encoding epigenetic regulatory proteins that modulate gene expression, so-called chromatinopathies. Intriguingly, several of the NDD-linked chromatinopathy genes have been shown to regulate autophagy-related genes, albeit in non-neuronal contexts. From these studies it becomes evident that tight transcriptional regulation of autophagy-related genes is crucial to control autophagic activity. This opens the exciting possibility that aberrant autophagic regulation might underly nervous system impairments in NDDs with disturbed epigenetic regulation. We here summarize NDD-related chromatinopathy genes that are known to regulate transcriptional regulation of autophagy-related genes. Thereby, we want to highlight autophagy as a candidate key hub mechanism in NDD-related chromatinopathies.Abbreviations: ADNP: activity dependent neuroprotector homeobox; ASD: autism spectrum disorder; ATG: AutTophaGy related; CpG: cytosine-guanine dinucleotide; DNMT: DNA methyltransferase; EHMT: euchromatic histone lysine methyltransferase; EP300: E1A binding protein p300; EZH2: enhancer of zeste 2 polycomb repressive complex 2 subunit; H3K4me3: histone 3 lysine 4 trimethylation; H3K9me1/2/3: histone 3 lysine 9 mono-, di-, or trimethylation; H3K27me2/3: histone 3 lysine 27 di-, or trimethylation; hiPSCs: human induced pluripotent stem cells; HSP: hereditary spastic paraplegia; ID: intellectual disability; KANSL1: KAT8 regulatory NSL complex subunit 1; KAT8: lysine acetyltransferase 8; KDM1A/LSD1: lysine demethylase 1A; MAP1LC3B: microtubule associated protein 1 light chain 3 beta; MTOR: mechanistic target of rapamycin kinase; MTORC1: mechanistic target of rapamycin complex 1; NDD: neurodevelopmental disorder; PHF8: PHD finger protein 8; PHF8-XLID: PHF8-X linked intellectual disability syndrome; PTM: post-translational modification; SESN2: sestrin 2; YY1: YY1 transcription factor; YY1AP1: YY1 associated protein 1.
    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
    Ferroptosis是一种新描述的调节坏死细胞死亡形式,参与与中风相关的病理性细胞死亡,导致脑缺血再灌注(I/R)损伤。因此,本研究旨在阐明GATA6在脑I/R损伤中神经元自噬和铁性凋亡中的作用。对脑I/R损伤相关差异表达基因(DEGs)以及GATA6的下游因子进行了生物信息学预测。此外,GATA6与miR-193b之间的关系以及miR-193b与ATG7之间的关系通过染色质免疫沉淀和双荧光素酶报告基因测定进行了评估.此外,神经元用氧糖剥夺(OGD)处理,随后是GATA6,miR-193b的过表达,和ATG7单独或组合以评估神经元自噬和铁凋亡。最后,在大脑中动脉闭塞(MCAO)刺激的脑I/R损伤大鼠模型中,进行体内实验以探讨GATA6/miR-193b/ATG7对神经元自噬和铁性凋亡的影响。发现GATA6和miR-193b在脑I/R损伤中表达较差。GATA6转录激活miR-193b下调ATG7。此外,GATA6介导的miR-193b激活通过抑制ATG7抑制OGD处理的神经元中的神经元自噬和铁凋亡。此外,GATA6/miR-193b通过下调体内ATG7抑制神经元自噬和铁性凋亡减轻了脑I/R损伤。总之,GATA6可能通过miR-193b/ATG7轴阻止神经元自噬和铁凋亡减轻脑I/R损伤。
    Ferroptosis is a newly described form of regulated necrotic cell death, which is engaged in the pathological cell death related to stroke, contributing to cerebral ischemia-reperfusion (I/R) injury. Therefore, we performed this study to clarify the role of GATA6 in neuronal autophagy and ferroptosis in cerebral I/R injury. The cerebral I/R injury-related differentially expressed genes (DEGs) as well as the downstream factors of GATA6 were predicted bioinformatically. Moreover, the relations between GATA6 and miR-193b and that between miR-193b and ATG7 were evaluated by chromatin immunoprecipitation and dual-luciferase reporter assays. Besides, neurons were treated with oxygen-glucose deprivation (OGD), followed by overexpression of GATA6, miR-193b, and ATG7 alone or in combination to assess neuronal autophagy and ferroptosis. At last, in vivo experiments were performed to explore the impacts of GATA6/miR-193b/ATG7 on neuronal autophagy and ferroptosis in a rat model of middle cerebral artery occlusion (MCAO)-stimulated cerebral I/R injury. It was found that GATA6 and miR-193b were poorly expressed in cerebral I/R injury. GATA6 transcriptionally activated miR-193b to downregulate ATG7. Additionally, GATA6-mediated miR-193b activation suppressed neuronal autophagy and ferroptosis in OGD-treated neurons by inhibiting ATG7. Furthermore, GATA6/miR-193b relieved cerebral I/R injury by restraining neuronal autophagy and ferroptosis via downregulation of ATG7 in vivo. In summary, GATA6 might prevent neuronal autophagy and ferroptosis to alleviate cerebral I/R injury via the miR-193b/ATG7 axis.
    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:羟基红花黄色素A(HSYA)是从植物红花中分离出的主要生物活性化合物,据报道对各种神经系统疾病具有神经保护作用,包括创伤性脑损伤(TBI)。然而,HSYA介导的TBI神经保护作用的具体分子和细胞机制尚不清楚.
    目的:本研究探讨HSYA对TBI小鼠自噬和NLRP3炎性体的影响及相关机制。
    方法:对小鼠进行TBI并用或不用HSYA处理。神经系统严重程度评分,首先进行LDH测定和细胞凋亡检测以评估HSYA在患有TBI的小鼠中的作用。然后进行RNA-seq以探索有助于HSYA介导的神经保护的机制。ELISA,西方印迹,并通过免疫荧光技术进一步探讨神经炎症和自噬的机制。此外,3-甲基腺嘌呤(3-MA),自噬抑制剂,用于确定自噬与NLRP3炎性体之间的联系。
    结果:HSYA显著降低神经系统严重程度评分,TBI小鼠血清LDH水平与细胞凋亡。在HSYA处理的TBI小鼠的皮层中鉴定出总共921个差异表达的基因,并且在炎症反应和自噬方面显着富集。此外,HSYA治疗显著降低炎性细胞因子水平和星形胶质细胞活化。重要的是,HSYA抑制神经元NLRP3炎性体激活,如NLRP3,ASC和裂解的caspase-1水平降低以及NLRP3神经元数量减少所示。它增加了自噬,改善了自噬通量功能障碍,如LC3II/LC3I水平升高和P62水平降低所证明。3-MA消除了HSYA对NLRP3炎性体的影响。机械上,HSYA可能通过AMPK/mTOR信号增强自噬。
    结论:HSYA通过触发AMPK/mTOR信号通路增强神经元自噬,导致抑制NLRP3炎性体以改善TBI后的神经系统恢复。
    BACKGROUND: Hydroxysafflor yellow A (HSYA) is the principal bioactive compound isolated from the plant Carthamus tinctorius L. and has been reported to exert neuroprotective effects against various neurological diseases, including traumatic brain injury (TBI). However, the specific molecular and cellular mechanisms underlying HSYA-mediated neuroprotection against TBI are unclear.
    OBJECTIVE: This study explored the effects of HSYA on autophagy and the NLRP3 inflammasome in mice with TBI and the related mechanisms.
    METHODS: Mice were subjected to TBI and treated with or without HSYA. Neurological severity scoring, LDH assays and apoptosis detection were first performed to assess the effects of HSYA in mice with TBI. RNA-seq was then conducted to explore the mechanisms that contributed to HSYA-mediated neuroprotection. ELISA, western blotting, and immunofluorescence were performed to further investigate the mechanisms of neuroinflammation and autophagy. Moreover, 3-methyladenine (3-MA), an autophagy inhibitor, was applied to determine the connection between autophagy and the NLRP3 inflammasome.
    RESULTS: HSYA significantly decreased the neurological severity score, serum LDH levels and apoptosis in mice with TBI. A total of 921 differentially expressed genes were identified in the cortices of HSYA-treated mice with TBI and were significantly enriched in the inflammatory response and autophagy. Furthermore, HSYA treatment markedly reduced inflammatory cytokine levels and astrocyte activation. Importantly, HSYA suppressed neuronal NLRP3 inflammasome activation, as indicated by decreased levels of NLRP3, ASC and cleaved caspase-1 and a reduced NLRP3+ neuron number. It increased autophagy and ameliorated autophagic flux dysfunction, as evidenced by increased LC3 II/LC3 I levels and decreased P62 levels. The effects of HSYA on the NLRP3 inflammasome were abolished by 3-MA. Mechanistically, HSYA may enhance autophagy through AMPK/mTOR signalling.
    CONCLUSIONS: HSYA enhanced neuronal autophagy by triggering the AMPK/mTOR signalling pathway, leading to inhibition of the NLRP3 inflammasome to improve neurological recovery after TBI.
    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
    最近对果蝇大脑发育过程中两种不同神经元类型中自噬的作用进行了表征,揭示了两种不同的调节突触形成的机制。在感光神经元中,在突触形成丝足中的自噬体形成会使假定的突触接触不稳定,从而限制了不正确的突触伙伴关系。在背侧簇神经元中,在轴突分支过程中,自噬被积极抑制以保持成熟突触的稳定。这些发现表明,不同的神经元类型可能需要在同一发育期激活或抑制突触自噬,以确保适当的突触形成和大脑连接。
    A recent characterization of the role of autophagy in two different neuron types during brain development in Drosophila revealed two different mechanisms to regulate synapse formation. In photoreceptor neurons, autophagosome formation in synaptogenic filopodia destabilizes presumptive synaptic contacts and thereby restricts incorrect synaptic partnerships. In dorsal cluster neurons, autophagy is actively suppressed to keep mature synapses stable during axonal branching. These findings indicate that different neuron types can require activation or suppression of synaptic autophagy during the same developmental period to ensure proper synapse formation and brain connectivity.
    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
    目的:最近的研究已经证明了针刺在临床上治疗神经系统疾病的神经保护作用。例如创伤性脑损伤(TBI)。由于越来越多的证据表明神经元自噬参与TBI的多个阶段,本研究旨在阐明针刺对TBI大鼠神经保护作用的自噬介导机制。
    方法:进行三个实验以检测神经元自噬的变化,并确定针刺治疗TBI的神经保护作用的潜在分子机制。采用Feeney自由落体硬膜外撞击法建立中度TBI大鼠模型,采用改良神经严重程度评分法(mNSS)进行神经功能恢复评价。Nissl和HE染色用于检查组织病理学改变。免疫荧光法检测LC3阳性细胞率。透射电镜(TEM)观察自噬体的形态和数量。蛋白质印迹法检测LC3、p62、beclin1、mTOR、ULK1,p-mTOR,和p-ULK1。定量实时聚合酶链反应(qRT-PCR)用于LC3mRNA和p62mRNA的基因表达分析。免疫共沉淀(CO-IP)方法用于鉴定mTOR和ULK1的蛋白质相互作用。
    结果:在TBI后第3天,针刺通过促进神经元自噬加速受损细胞结构的清除;在TBI后第7天和第14天,针刺抑制神经元自噬,防止过度自噬,从而减轻神经损伤。此外,在TBI后的不同阶段同时用3-MA或雷帕霉素治疗可减弱针刺的效果。
    结论:针刺对TBI不同阶段的神经元自噬具有良性调节作用,可能通过mTOR/ULK1途径。
    Recent investigations have already proved the neuroprotective efficacy of acupuncture in clinical practice in the treatment of neurological diseases, such as traumatic brain injury (TBI). Since growing evidence has suggested that neuronal autophagy was involved in multiple stages of TBI, this study aims to clarify the autophagy mediating mechanism underlying the neuroprotective effect of acupuncture in TBI rats.
    Three experiments were carried out to detect changes in neuronal autophagy and identify the potential molecular mechanism underlying the neuroprotective effect of acupuncture for TBI treatment. Feeney\'s free-falling epidural impingement method was used to establish the moderate TBI rat model; modified neurological severity scoring (mNSS) was used for neurological recovery evaluation. Nissl and HE staining were used to examine the histopathological changes. Immunofluorescence was used to detect the LC3-positive cell rate. The transmission electron microscope (TEM) was used to investigate the morphology and quantity of autophagosomes. Western blotting was used to determine the protein expressions of LC3, p62, beclin1, mTOR, ULK1, p-mTOR, and p-ULK1. Quantitative real-time polymerase chain reaction (qRT-PCR) was used for gene expressions analysis of LC3 mRNA and p62 mRNA. Co-immunoprecipitation (CO-IP) method was used to identify the protein interaction of mTOR and ULK1.
    On Day 3 after TBI, acupuncture accelerated the removal of damaged cellular structures by promoting neuronal autophagy; on Day 7 and Day 14 after TBI, acupuncture inhibited neuronal autophagy, preventing excessive autophagy and thus alleviated nerve damage. In addition, the simultaneous treatment with 3-MA or rapamycin at different stages after TBI attenuated the effect of acupuncture.
    Acupuncture has a benign regulatory effect on neuronal autophagy in different stages of TBI, possibly through the mTOR/ULK1 pathway.
    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
    在显示局部凋亡和线粒体应激迹象并经历神经免疫突触修剪的突触中,突触前蛋白水平的增加,可以观察到神经元特异性septin-3。Septin-3是SeptinGTP酶家族的成员,能够形成多聚体并有助于细胞骨架。然而,septin-3的功能仍然难以捉摸。这里,我们提供的证据表明,septin-3能够结合研究最多的自噬蛋白Atg8同源微管相关蛋白1轻链3B(LC3B),除了另一个同源物,GABA受体相关蛋白样2(GABARAPL2)。此外,我们证明,在原代神经元细胞中,化学自噬诱导后,septin-3和LC3B的共定位增加.Septin-3在自噬增强或阻断后在原代神经元中积累,类似于自噬蛋白。使用电子显微镜,我们还显示septin-3定位于LC3B阳性膜,并且可以在线粒体中找到。然而,septin-3和早期线粒体自噬标记PTEN诱导的激酶1(PINK1)的共定位结果不支持septin-3与线粒体的结合与线粒体自噬有关。我们得出结论,septin-3与突触/神经元自噬相关,结合Atg8并定位到可以通过化学自噬诱导增强的自噬膜。根据我们的结果,升高的septin-3水平可能表明神经元自噬增强或受阻。
    In synapses that show signs of local apoptosis and mitochondrial stress and undergo neuro-immunological synapse pruning, an increase in the levels of the presynaptic protein, neuronal-specific septin-3 can be observed. Septin-3 is a member of the septin GTPase family with the ability to form multimers and contribute to the cytoskeleton. However, the function of septin-3 remains elusive. Here, we provide evidence that septin-3 is capable of binding the most-studied autophagy protein Atg8 homolog microtubule-associated protein 1 light chain 3B (LC3B), besides another homolog, GABA receptor-associated protein-like 2 (GABARAPL2). Moreover, we demonstrate that colocalization of septin-3 and LC3B increases upon chemical autophagy induction in primary neuronal cells. Septin-3 is accumulated in primary neurons upon autophagy enhancement or blockade, similar to autophagy proteins. Using electron microscopy, we also show that septin-3 localizes to LC3B positive membranes and can be found at mitochondria. However, colocalization results of septin-3 and the early mitophagy marker PTEN-induced kinase 1 (PINK1) do not support that binding of septin-3 to mitochondria is mitophagy related. We conclude that septin-3 correlates with synaptic/neuronal autophagy, binds Atg8 and localizes to autophagic membranes that can be enhanced with chemical autophagy induction. Based on our results, elevated septin-3 levels might indicate enhanced or impeded autophagy in neurons.
    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
    阿尔茨海默病(AD)是一种普遍存在的有害神经退行性疾病,其特征是认知能力的不可逆和进行性损害以及脑中淀粉样蛋白β(Aβ)斑块和神经原纤维缠结(NFT)的形成。到目前为止,AD的确切机制尚不完全清楚,目前尚无有效减缓疾病进展的干预措施.自噬是一种保守的降解途径,通过靶向受损的细胞器来维持细胞内稳态至关重要。病原体,和疾病倾向的蛋白质聚集到溶酶体降解。新的证据表明,功能失调的自噬清除途径是受影响的神经元中AD发病机理的潜在细胞机制。在此,我们总结了目前在AD病理生理学中自噬功能障碍的证据,并讨论了自噬在调节AD相关蛋白降解和神经元和神经胶质细胞神经炎症中的作用。最后,我们回顾了在AD模型治疗中报道的自噬调节剂,并讨论了新型自噬激活剂用于AD治疗的潜在临床应用的障碍和机会。
    Alzheimer\'s disease (AD) is a prevalent and deleterious neurodegenerative disorder characterized by an irreversible and progressive impairment of cognitive abilities as well as the formation of amyloid β (Aβ) plaques and neurofibrillary tangles (NFTs) in the brain. By far, the precise mechanisms of AD are not fully understood and no interventions are available to effectively slow down progression of the disease. Autophagy is a conserved degradation pathway that is crucial to maintain cellular homeostasis by targeting damaged organelles, pathogens, and disease-prone protein aggregates to lysosome for degradation. Emerging evidence suggests dysfunctional autophagy clearance pathway as a potential cellular mechanism underlying the pathogenesis of AD in affected neurons. Here we summarize the current evidence for autophagy dysfunction in the pathophysiology of AD and discuss the role of autophagy in the regulation of AD-related protein degradation and neuroinflammation in neurons and glial cells. Finally, we review the autophagy modulators reported in the treatment of AD models and discuss the obstacles and opportunities for potential clinical application of the novel autophagy activators for AD therapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号