neuronal death

神经元死亡
  • 文章类型: Journal Article
    颅内动脉瘤破裂引起的蛛网膜下腔出血预后较差,使这种疾病成为社会问题。SAH发作后颅内压和蛛网膜下腔血块升高引起的炎症加剧了神经元死亡和血管痉挛,导致不良结果和严重的后遗症。这里,FROUNT介导CCR2和CCR5信号作为细胞内分子与这些化学引诱受体结合,促进炎症细胞的迁移,比如巨噬细胞,在原地引发炎症。鞘内注射自体血建立大鼠蛛网膜下腔出血动物模型。FROUNT抑制剂的作用,双硫仑,关于存活率,然后检查海马或血管痉挛中的神经元死亡。双硫仑的鞘内给药可显着抑制CD68阳性巨噬细胞和髓过氧化物酶阳性中性粒细胞原位向水箱凝块的浸润。在这种情况下,双硫仑改善了大鼠蛛网膜下腔出血后动物的死亡。此外,双硫仑抑制了蛛网膜下腔出血后的两个主要事件,海马神经元死亡和血管痉挛。因此,双硫仑对CCR2和CCR5信号传导的药理学抑制可能是改善蛛网膜下腔出血结果的治疗策略。
    Subarachnoid hemorrhage due to rupture of intracranial aneurysms has a poor outcome, making this disease being the social problem. Inflammation evoked by the increase in intracranial pressure and the clot in the subarachnoid space after the onset of SAH exacerbates neuronal death and vasospasm, resulting in the poor outcome and severe aftereffects. Here, FROUNT mediates CCR2 and CCR5 signaling as an intracellular molecule binding to these chemoattractant receptors which facilitate the migration of inflammatory cells, such as macrophages, in situ to trigger inflammation there. Animal model of subarachnoid hemorrhage was established in rats through intrathecal injection of autologous blood. The effect of the FROUNT inhibitor, disulfiram, on survival rate, neuronal death in hippocampus or vasospasm was then examined. The intrathecal administration of disulfiram significantly suppressed the infiltration of CD68-positive macrophages and myeloperoxidase-positive neutrophils toward the clot in the cistern in situ. In this condition, disulfiram ameliorated the death of animals after the onset of subarachnoid hemorrhage in rats. In addition, disulfiram suppressed both the two major events after subarachnoid hemorrhage, the neuronal death in hippocampus and vasospasm. The pharmacological inhibition of CCR2 and CCR5 signaling by disulfiram could thus be the therapeutic strategy to improve the outcome of subarachnoid hemorrhage.
    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
    治疗性电刺激,例如经颅皮层刺激和周围体感刺激,用于改善中风患者的运动功能。我们假设这些刺激通过调节新的共同信号通路在缺血性卒中亚急性期发挥神经保护作用。用高清晰度(HD)-经颅交流电流刺激(tACS;20Hz,89.1A/mm2),HD-经颅直流电刺激(tDCS;强度,55A/mm2;电荷密度,66,000C/m2),或电针(EA,2Hz,1mA)在中风的早期阶段。使用行为运动功能测试评估治疗效果。使用转录组学和其他生物医学分析确定了潜在的机制。所有治疗性电动工具都减轻了缺血性中风引起的运动功能障碍。我们使用转录组分析专注于电刺激参与细胞凋亡和细胞死亡的常见基因,并选择了11个最有效的靶标(Trem2,S100a9,Lgals3,Tlr4,Myd88,NF-kB,STAT1,IL-6,IL-1β,TNF-α,和Iba1)。随后的研究表明,电刺激调节炎症细胞因子,包括IL-1β和TNF-α,通过调节STAT1和NF-kB激活,特别是在变形虫小胶质细胞中;此外,电刺激通过激活神经营养因子增强神经元存活,包括BDNF和FGF9。应用于经颅皮质或外周神经水平以促进功能恢复的治疗性电刺激可以通过调节常见的神经元死亡途径和上调神经营养因子来改善神经保护。因此,联合经颅皮质和外周体感刺激可能发挥协同神经保护作用,进一步增强对缺血性卒中患者运动功能障碍的有益作用。
    Therapeutic electrical stimulation, such as transcranial cortical stimulation and peripheral somatosensory stimulation, is used to improve motor function in patients with stroke. We hypothesized that these stimulations exert neuroprotective effects during the subacute phase of ischemic stroke by regulating novel common signaling pathways. Male C57BL/6J mouse models of ischemic stroke were treated with high-definition (HD)-transcranial alternating current stimulation (tACS; 20 Hz, 89.1 A/mm2), HD-transcranial direct current stimulation (tDCS; intensity, 55 A/mm2; charge density, 66,000 C/m2), or electroacupuncture (EA, 2 Hz, 1 mA) in the early stages of stroke. The therapeutic effects were assessed using behavioral motor function tests. The underlying mechanisms were determined using transcriptomic and other biomedical analyses. All therapeutic electrical tools alleviated the motor dysfunction caused by ischemic stroke insults. We focused on electrically stimulating common genes involved in apoptosis and cell death using transcriptome analysis and chose 11 of the most potent targets (Trem2, S100a9, Lgals3, Tlr4, Myd88, NF-kB, STAT1, IL-6, IL-1β, TNF-α, and Iba1). Subsequent investigations revealed that electrical stimulation modulated inflammatory cytokines, including IL-1β and TNF-α, by regulating STAT1 and NF-kB activation, especially in amoeboid microglia; moreover, electrical stimulation enhanced neuronal survival by activating neurotrophic factors, including BDNF and FGF9. Therapeutic electrical stimulation applied to the transcranial cortical- or periphery-nerve level to promote functional recovery may improve neuroprotection by modulating a common neuronal death pathway and upregulating neurotrophic factors. Therefore, combining transcranial cortical and peripheral somatosensory stimulation may exert a synergistic neuroprotective effect, further enhancing the beneficial effects on motor deficits in patients with 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
    准备急性脑切片会产生模拟严重穿透性脑损伤的创伤。在新生儿急性脑切片中,创伤诱导的神经元钙动力学的时空特征及其对网络活动的影响相对未知。使用多光子激光扫描显微镜对急性新生小鼠脑片(P8-12)中的体感新皮层进行了观察,我们同时对神经元Ca2动力学(GCaMP6s)和细胞毒性(碘化丙啶或PI)进行成像,以确定细胞毒性Ca2负载神经元(GCaMP填充)与不同深度和孵育时间的细胞活力之间的关系。切片表面富含PI+细胞和充满GCaMP的神经元,随深度呈指数下降。具有高PI+细胞的区域与升高的神经元和神经纤维Ca2+相关。随着长时间的孵育,PI+细胞和GCaMP填充的神经元的数量增加。GCaMP填充的神经元不参与刺激诱发或癫痫诱发的网络活动。重要的是,浅表组织,创伤引起的损伤程度更高,显示癫痫相关的神经元Ca2+反应减弱。钙蛋白酶抑制阻止了深层组织中PI细胞和GCaMP填充的神经元的增加以及延长的孵育时间。同种型特异性药理学抑制暗示钙蛋白酶-2是急性切片中创伤诱发损伤的重要因素。我们的结果表明,急性新生儿脑切片中细胞死亡与异常神经元Ca2负荷之间存在钙蛋白酶介导的时空关系。此外,我们证明,急性脑片中的神经元表现出改变的生理学,这取决于创伤诱导的损伤程度。阻断钙蛋白酶可能是预防年轻大脑创伤性脑损伤期间急性神经元死亡的治疗选择。意义陈述这是首次描述急性新生儿切片中新皮质损伤的时空动力学的研究,模仿严重的穿透性创伤性脑损伤,使用PI标记和升高的神经元Ca2+负荷作为细胞毒性的标志物。我们发现深度和时间依赖性神经元损伤,导致神经元反应改变。通过药理学抑制钙蛋白酶减轻了神经元Ca2升高和细胞毒性,参与多种细胞死亡机制的Ca2+依赖性蛋白酶家族。我们的研究为新生儿急性脑切片中损伤依赖性神经元和回路功能改变提供了证据。钙蛋白酶抑制减少了新生儿大脑中创伤诱导的细胞死亡,将它们确定为这个年龄段的潜在治疗目标。
    Preparing acute brain slices produces trauma that mimics severe penetrating brain injury. In neonatal acute brain slices, the spatiotemporal characteristics of trauma-induced calcium dynamics in neurons and its effect on network activity are relatively unknown. Using multiphoton laser scanning microscopy of the somatosensory neocortex in acute neonatal mouse brain slices (P8-12), we simultaneously imaged neuronal Ca2+ dynamics (GCaMP6s) and cytotoxicity (propidium iodide or PI) to determine the relationship between cytotoxic Ca2+ loaded neurons (GCaMP-filled) and cell viability at different depths and incubation times. PI+ cells and GCaMP-filled neurons were abundant at the surface of the slices, with an exponential decrease with depth. Regions with high PI+ cells correlated with elevated neuronal and neuropil Ca2+ The number of PI+ cells and GCaMP-filled neurons increased with prolonged incubation. GCaMP-filled neurons did not participate in stimulus-evoked or seizure-evoked network activity. Significantly, the superficial tissue, with a higher degree of trauma-induced injury, showed attenuated seizure-related neuronal Ca2+ responses. Calpain inhibition prevented the increase in PI+ cells and GCaMP-filled neurons in the deep tissue and during prolonged incubation times. Isoform-specific pharmacological inhibition implicated calpain-2 as a significant contributor to trauma-induced injury in acute slices. Our results show a calpain-mediated spatiotemporal relationship between cell death and aberrant neuronal Ca2+ load in acute neonatal brain slices. Also, we demonstrate that neurons in acute brain slices exhibit altered physiology depending on the degree of trauma-induced injury. Blocking calpains may be a therapeutic option to prevent acute neuronal death during traumatic brain injury in the young brain.
    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
    癫痫,以异常和过度的大脑神经元活动为标志,与神经元膜中L型电压门控钙通道(LTCC)的激活有关。LTCC促进钙(Ca2)和其他金属离子的进入,如锌(Zn2+)和镁(Mg2+),进入细胞质。突触前末端的这种Ca2流入触发了Zn2和谷氨酸释放到突触后末端。然后通过LTCC将Zn2+转运到突触后神经元。由此产生的Zn2+在神经元中的积累显著增加了烟酰胺腺嘌呤二核苷酸磷酸(NADPH)氧化酶亚基的表达,有助于活性氧(ROS)的产生和神经元死亡。氨氯地平(AML),通常用于高血压和冠状动脉疾病,通过抑制LTCC工作。我们探讨了AML是否可以减轻神经元中的Zn2+易位和积累,可能提供预防癫痫引起的海马神经元死亡的保护。我们通过使用毛果芸香碱建立大鼠癫痫模型并给予AML(10mg/kg,口头,每天持续7天)癫痫发作后。我们通过行为测试评估认知功能,并对Zn2+积累进行组织学分析,氧化应激,和神经元死亡。我们的研究结果表明,AML的LTCC抑制减少了过度的Zn2+积累,活性氧(ROS)的产生,癫痫发作后海马神经元死亡。这些结果表明,氨氯地平在癫痫发作管理和减轻癫痫发作的不利影响方面具有作为治疗剂的潜力。
    Epilepsy, marked by abnormal and excessive brain neuronal activity, is linked to the activation of L-type voltage-gated calcium channels (LTCCs) in neuronal membranes. LTCCs facilitate the entry of calcium (Ca2+) and other metal ions, such as zinc (Zn2+) and magnesium (Mg2+), into the cytosol. This Ca2+ influx at the presynaptic terminal triggers the release of Zn2+ and glutamate to the postsynaptic terminal. Zn2+ is then transported to the postsynaptic neuron via LTCCs. The resulting Zn2+ accumulation in neurons significantly increases the expression of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase subunits, contributing to reactive oxygen species (ROS) generation and neuronal death. Amlodipine (AML), typically used for hypertension and coronary artery disease, works by inhibiting LTCCs. We explored whether AML could mitigate Zn2+ translocation and accumulation in neurons, potentially offering protection against seizure-induced hippocampal neuronal death. We tested this by establishing a rat epilepsy model with pilocarpine and administering AML (10 mg/kg, orally, daily for 7 days) post-epilepsy onset. We assessed cognitive function through behavioral tests and conducted histological analyses for Zn2+ accumulation, oxidative stress, and neuronal death. Our findings show that AML\'s LTCC inhibition decreased excessive Zn2+ accumulation, reactive oxygen species (ROS) production, and hippocampal neuronal death following seizures. These results suggest amlodipine\'s potential as a therapeutic agent in seizure management and mitigating seizures\' detrimental effects.
    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
    癫痫发作产生的分子机制仍然难以捉摸,然而,它们对于开发有效的癫痫治疗方法至关重要。目前的研究表明,抑制c-Abl酪氨酸激酶可以防止细胞凋亡,减少树突状脊柱的损失,并在兴奋性毒性的体外模型中维持N-甲基-d-天冬氨酸(NMDA)受体亚基2B(NR2B)磷酸化。毛果芸香碱诱导的小鼠癫痫持续状态(SE)促进c-Abl磷酸化,破坏c-Abl活性导致癫痫发作减少,向SE增加延迟,改善动物生存。目前,临床上使用的c-Abl抑制剂是非选择性的,并且脑渗透性差。变构c-Abl抑制剂,neurotinib,在这里使用具有良好的效力,选择性,药代动力学,大大改善了大脑的穿透力。给予Neurotinib的小鼠在毛果芸香碱-SE诱导后癫痫发作较少,存活率提高。我们的发现揭示了c-Abl激酶的激活是发生的关键因素,并强调了其抑制作用在预防啮齿动物和人类癫痫样癫痫发作中的作用。
    The molecular mechanisms underlying seizure generation remain elusive, yet they are crucial for developing effective treatments for epilepsy. The current study shows that inhibiting c-Abl tyrosine kinase prevents apoptosis, reduces dendritic spine loss, and maintains N-methyl-d-aspartate (NMDA) receptor subunit 2B (NR2B) phosphorylated in in vitro models of excitotoxicity. Pilocarpine-induced status epilepticus (SE) in mice promotes c-Abl phosphorylation, and disrupting c-Abl activity leads to fewer seizures, increases latency toward SE, and improved animal survival. Currently, clinically used c-Abl inhibitors are non-selective and have poor brain penetration. The allosteric c-Abl inhibitor, neurotinib, used here has favorable potency, selectivity, pharmacokinetics, and vastly improved brain penetration. Neurotinib-administered mice have fewer seizures and improved survival following pilocarpine-SE induction. Our findings reveal c-Abl kinase activation as a key factor in ictogenesis and highlight the impact of its inhibition in preventing the insurgence of epileptic-like seizures in rodents and humans.
    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
    衰老是一个复杂的,多因素,不可逆的细胞周期停止,除了是衰老和神经系统疾病的重要因素外,还具有肿瘤抑制作用。DNA受损,神经炎症,氧化应激和破坏的蛋白质平衡是引起衰老的一些因素。衰老是由DNA损伤引发的,DNA损伤会引发DNA损伤反应。DNA损伤反应,其中包括含有活化H2AX的DNA损伤灶的形成,这是细胞衰老的关键因素,是由双链DNA断裂引起的。氧化应激损害认知,抑制神经发生,并具有加速老化的效果。衰老细胞产生称为衰老相关分泌表型(SASP)的促炎介质。这些促炎细胞因子和趋化因子对神经炎症有影响,神经元死亡,和细胞增殖。虽然很容易将神经退行性疾病视为加速衰老和衰老的表现,这篇综述将提供有关衰老和DNA损伤反应导致的脑衰老和神经变性的信息。
    Senescence is a complicated, multi-factorial, irreversible cell cycle halt that has a tumor-suppressing effect in addition to being a significant factor in aging and neurological diseases. Damaged DNA, neuroinflammation, oxidative stress and disrupted proteostasis are a few of the factors that cause senescence. Senescence is triggered by DNA damage which initiates DNA damage response. The DNA damage response, which includes the formation of DNA damage foci containing activated H2AX, which is a key factor in cellular senescence, is provoked by a double strand DNA break. Oxidative stress impairs cognition, inhibits neurogenesis, and has an accelerated aging effect. Senescent cells generate pro-inflammatory mediators known as senescence-associated secretory phenotype (SASP). These pro-inflammatory cytokines and chemokines have an impact on neuroinflammation, neuronal death, and cell proliferation. While it is tempting to think of neurodegenerative diseases as manifestations of accelerated aging and senescence, this review will present information on brain ageing and neurodegeneration as a result of senescence and DNA damage response.
    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
    线粒体功能障碍和谷氨酸毒性与神经疾病有关,包括脑外伤.文献综述表明,神经元谷氨酸的毒性和传递作用在空间和功能上是分开的。传递途径利用突触GluN2A受体,快速释放的谷氨酸池,Synaptotagmin1介导的谷氨酸诱发释放和星形胶质细胞调节的细胞外谷氨酸量。毒性途径利用突触外GluN2B受体和谷氨酸的细胞质池,它是由Synaptotagmin7和神经元2-酮戊二酸脱氢酶复合物(OGDHC)介导的谷氨酸自发释放的结果,三羧酸(TCA)循环酶。此外,在神经炎症时观察到的OGDHC的抑制是由于免疫细胞过度释放活性氧/氮物质。OGDHC的丢失抑制线粒体对谷氨酸的摄取,从而促进其细胞外积累和刺激毒性谷氨酸途径而不影响传播。高水平的细胞外谷氨酸导致细胞内氧化还原稳态失调并引起铁凋亡,兴奋毒性,和线粒体功能障碍。后者影响需要高能量供应的传输途径并导致细胞死亡。线粒体由于TCA循环受损而加重谷氨酸毒性,并成为谷氨酸毒性的受害者,破坏氧化磷酸化。因此,在神经系统疾病中靶向TCA周期的治疗可能比试图保持线粒体氧化磷酸化更有效.
    Mitochondrial dysfunction and glutamate toxicity are associated with neural disorders, including brain trauma. A review of the literature suggests that toxic and transmission actions of neuronal glutamate are spatially and functionally separated. The transmission pathway utilizes synaptic GluN2A receptors, rapidly released pool of glutamate, evoked release of glutamate mediated by Synaptotagmin 1 and the amount of extracellular glutamate regulated by astrocytes. The toxic pathway utilizes extrasynaptic GluN2B receptors and a cytoplasmic pool of glutamate, which results from the spontaneous release of glutamate mediated by Synaptotagmin 7 and the neuronal 2-oxoglutarate dehydrogenase complex (OGDHC), a tricarboxylic acid (TCA) cycle enzyme. Additionally, the inhibition of OGDHC observed upon neuro-inflammation is due to an excessive release of reactive oxygen/nitrogen species by immune cells. The loss of OGDHC inhibits uptake of glutamate by mitochondria, thus facilitating its extracellular accumulation and stimulating toxic glutamate pathway without affecting transmission. High levels of extracellular glutamate lead to dysregulation of intracellular redox homeostasis and cause ferroptosis, excitotoxicity, and mitochondrial dysfunction. The latter affects the transmission pathway demanding high-energy supply and leading to cell death. Mitochondria aggravate glutamate toxicity due to impairments in the TCA cycle and become a victim of glutamate toxicity, which disrupts oxidative phosphorylation. Thus, therapies targeting the TCA cycle in neurological disorders may be more efficient than attempting to preserve mitochondrial oxidative phosphorylation.
    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
    神经功能障碍和神经胶质激活在严重感染如败血症中很常见。在患者和动物模型中,对全身性炎症的反应都存在性二态性。但很少有比较研究。这里,我们研究了通过腹腔注射脂多糖(LPS)诱导的全身性炎症对雄性和雌性小鼠视网膜的影响,并确定了NLRP3炎性体的拮抗作用和凋亡的外源性途径是否对视网膜具有保护作用。
    向两个月龄的C57BL/6J雄性和雌性小鼠施用单次腹膜内注射LPS(5mg/kg)。使用视网膜电图和光谱域光学相干断层扫描在体内纵向检查视网膜。在平板上分析了视网膜神经节细胞(RGC)的存活和小胶质细胞的活化。将视网膜提取物用于CD45和CD11b阳性细胞的流式细胞术分析。通过ELISA测量促炎细胞因子的匹配血浆和视网膜水平。在用P2X7R和TNFR1拮抗剂单独或组合治疗的动物中评估视网膜功能和RGC存活。
    在LPS处理的两种性别的动物中,有一过性视网膜功能障碍,视力形成性RGC丧失,但非视力形成性RGC丧失,视网膜肿胀,小胶质细胞激活,细胞浸润,并增加TNF和IL-1β。与女性相比,男性表现出更高的视觉形成RGC死亡,功能恢复较慢,和淋巴毒素α在他们的视网膜中过度表达。P2X7R和TNFR1拮抗作用,单独或组合,获救的视力形成RGC。P2X7R拮抗作用也挽救了视网膜功能。女性对治疗的反应优于男性。
    全身性LPS在小鼠视网膜中具有神经元和性别特异性的不良反应,通过靶向NLRP3炎性体和凋亡的外在途径来抵消。我们的结果强调了在影响中枢神经系统的炎症性疾病的临床前研究中分析男性和女性的必要性。
    Neurological dysfunction and glial activation are common in severe infections such as sepsis. There is a sexual dimorphism in the response to systemic inflammation in both patients and animal models, but there are few comparative studies. Here, we investigate the effect of systemic inflammation induced by intraperitoneal administration of lipopolysaccharide (LPS) on the retina of male and female mice and determine whether antagonism of the NLRP3 inflammasome and the extrinsic pathway of apoptosis have protective effects on the retina.
    A single intraperitoneal injection of LPS (5 mg/kg) was administered to two months old C57BL/6J male and female mice. Retinas were examined longitudinally in vivo using electroretinography and spectral domain optical coherence tomography. Retinal ganglion cell (RGC) survival and microglial activation were analysed in flat-mounts. Retinal extracts were used for flow cytometric analysis of CD45 and CD11b positive cells. Matched plasma and retinal levels of proinflammatory cytokines were measured by ELISA. Retinal function and RGC survival were assessed in animals treated with P2X7R and TNFR1 antagonists alone or in combination.
    In LPS-treated animals of both sexes, there was transient retinal dysfunction, loss of vision-forming but not non-vision forming RGCs, retinal swelling, microglial activation, cell infiltration, and increases in TNF and IL-1β. Compared to females, males showed higher vision-forming RGC death, slower functional recovery, and overexpression of lymphotoxin alpha in their retinas. P2X7R and TNFR1 antagonism, alone or in combination, rescued vision-forming RGCs. P2X7R antagonism also rescued retinal function. Response to treatment was better in females than in males.
    Systemic LPS has neuronal and sex-specific adverse effects in the mouse retina, which are counteracted by targeting the NLRP3 inflammasome and the extrinsic pathway of apoptosis. Our results highlight the need to analyse males and females in preclinical studies of inflammatory diseases affecting the central nervous system.
    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
    神经元死亡是阿尔茨海默病(AD)的关键病理之一。神经元死亡是如何在AD中开始的,目前还不清楚,因此,澄清这一过程可能有助于开发有效的治疗方法。本研究收集了85个AD样本和83个对照样本的单细胞RNA测序数据,覆盖前额叶皮层,内部嗅觉皮层,上顶叶,额上回,尾内嗅皮质,体感皮层,海马体,上额叶皮质和外周血单个核细胞。此外,获得了来自6只AppNL-G-FAD小鼠和6只对照C57Bl/6J小鼠的冠状切片的空间转录组数据。在野生型和5×FAD小鼠中实验验证了主要的单细胞和空间转录组学结果。我们发现小胶质细胞亚群Mic_PTPRG可以与特定类型的神经元(尤其是兴奋性ExNeu_PRKN_VIRMA和抑制性InNeu_PRKN_VIRMA神经元亚群)进行通信,并导致它们在AD进展过程中表达PTPRG。在神经元内,PTPRG结合并上调m6A甲基转移酶VIRMA,从而抑制PRKNmRNA的翻译,通过抑制线粒体自噬来阻止神经元受损线粒体的清除。随着疾病的进展,神经元中的能量和营养代谢途径被重新编程,导致他们的死亡。始终如一,我们确定PTPTRG可以在小鼠大脑中与VIRMA物理相互作用,而PRKN在5×FAD小鼠大脑中显著上调。总之,我们的发现表明,PTPRG激活m6A甲基转移酶VIRMA,以阻断线粒体自噬介导的AD神经元死亡,这是一条潜在的途径,小胶质细胞和神经元PTPRG通过其在AD进展期间修饰大脑中的神经元连接。
    Neuronal death is one of the key pathologies in Alzheimer\'s disease (AD). How neuronal death begins in AD is far from clear, so clarifying this process may help develop effective therapies. This study collected single-cell RNA sequencing data of 85 AD samples and 83 control samples, covering the prefrontal cortex, internal olfactory cortex, superior parietal lobe, superior frontal gyrus, caudal internal olfactory cortex, somatosensory cortex, hippocampus, superior frontal cortex and peripheral blood mononuclear cells. Additionally, spatial transcriptomic data of coronal sections from 6 AppNL-G-F AD mice and 6 control C57Bl/6 J mice were acquired. The main single-cell and spatial transcriptomics results were experimentally validated in wild type and 5 × FAD mice. We found that the microglia subpopulation Mic_PTPRG can communicate with specific types of neurons (especially excitatory ExNeu_PRKN_VIRMA and inhibitory InNeu_PRKN_VIRMA neuronal subpopulations) and cause them to express PTPRG during AD progression. Within neurons, PTPRG binds and upregulates the m6A methyltransferase VIRMA, thus inhibiting translation of PRKN mRNA to prevent the clearance of damaged mitochondria in neurons through suppressing mitophagy. As the disease progresses, the energy and nutrient metabolic pathways in neurons are reprogrammed, leading to their death. Consistently, we determined that PTPTRG can physically interact with VIRMA in mouse brains and PRKN is significantly upregulated in 5 × FAD mouse brain. Altogether, our findings demonstrate that PTPRG activates the m6A methyltransferase VIRMA to block mitophagy-mediated neuronal death in AD, which is a potential pathway, through which microglia and neuronal PTPRG modify neuronal connections in the brain during AD progression.
    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
    N-甲基-D-天冬氨酸受体(NMDAR)介导的谷氨酸兴奋性毒性显着导致缺血性神经元死亡和再通后梗死扩展。尽管付出了巨大的努力,在减轻脑损伤的临床试验中,靶向NMDAR已被证明是不成功的.这里,我们发现瞬时受体电位美司他丁2(TRPM2)与蛋白激酶Cγ(PKCγ)相关的相互作用基序,并证明TRPM2-PKCγ解偶联是减轻缺血性卒中NMDAR介导的兴奋性毒性的有效治疗策略.我们证明,TRPM2-PKCγ相互作用允许TRPM2介导的Ca2+流入促进PKCγ激活,随后增强TRPM2诱导的突触外NMDAR(esNMDAR)活性的增强作用。通过鉴定TRPM2(M2PBM)上的PKCγ结合基序,与PKCγ的C2结构域直接相关,开发了一种干扰肽(TAT-M2PBM)来破坏TRPM2-PKCγ相互作用而不损害PKCγ功能。M2PBM缺失或TRPM2-PKCγ解离消除了TRPM2-PKCγ和TRPM2-esNMDAR偶联,导致兴奋性毒性神经元死亡减少,缺血性脑损伤减轻。
    N-methyl-D-aspartate receptor (NMDAR)-mediated glutamate excitotoxicity significantly contributes to ischemic neuronal death and post-recanalization infarction expansion. Despite tremendous efforts, targeting NMDARs has proven unsuccessful in clinical trials for mitigating brain injury. Here, we show the discovery of an interaction motif for transient receptor potential melastatin 2 (TRPM2) and protein kinase Cγ (PKCγ) association and demonstrate that TRPM2-PKCγ uncoupling is an effective therapeutic strategy for attenuating NMDAR-mediated excitotoxicity in ischemic stroke. We demonstrate that the TRPM2-PKCγ interaction allows TRPM2-mediated Ca2+ influx to promote PKCγ activation, which subsequently enhances TRPM2-induced potentiation of extrasynaptic NMDAR (esNMDAR) activity. By identifying the PKCγ binding motif on TRPM2 (M2PBM), which directly associates with the C2 domain of PKCγ, an interfering peptide (TAT-M2PBM) is developed to disrupt TRPM2-PKCγ interaction without compromising PKCγ function. M2PBM deletion or TRPM2-PKCγ dissociation abolishes both TRPM2-PKCγ and TRPM2-esNMDAR couplings, resulting in reduced excitotoxic neuronal death and attenuated ischemic brain injury.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号