neuronal survival

神经元存活
  • 文章类型: Journal Article
    视网膜神经节细胞(RGC)的轴突形成视神经,将视觉信息从眼睛传递到大脑。RGC及其轴突的损伤或丢失是创伤性损伤和退行性疾病如青光眼中视觉功能缺陷的主要原因。然而,这些神经退行性疾病的神经损伤尚无有效的临床治疗方法。这里,我们报道LIM同源结构域转录因子Lhx2在多种模拟青光眼疾病的动物模型中促进RGC存活和轴突再生.此外,在N-甲基-D-天冬氨酸(NMDA)诱导的RGCs兴奋毒性损伤后,Lhx2减轻了视觉信号转导的损失。机制分析表明,Lhx2的过表达通过系统地调节再生相关基因的转录和抑制信号素3C(Sema3C)的转录来支持轴突再生。总的来说,我们的研究确定了Lhx2在促进RGC存活和轴突再生中的关键作用,为青光眼神经变性提供了一种有前途的神经修复策略。
    The axons of retinal ganglion cells (RGCs) form the optic nerve, transmitting visual information from the eye to the brain. Damage or loss of RGCs and their axons is the leading cause of visual functional defects in traumatic injury and degenerative diseases such as glaucoma. However, there are no effective clinical treatments for nerve damage in these neurodegenerative diseases. Here, we report that LIM homeodomain transcription factor Lhx2 promotes RGC survival and axon regeneration in multiple animal models mimicking glaucoma disease. Furthermore, following N-methyl-D-aspartate (NMDA)-induced excitotoxicity damage of RGCs, Lhx2 mitigates the loss of visual signal transduction. Mechanistic analysis revealed that overexpression of Lhx2 supports axon regeneration by systematically regulating the transcription of regeneration-related genes and inhibiting transcription of Semaphorin 3C (Sema3C). Collectively, our studies identify a critical role of Lhx2 in promoting RGC survival and axon regeneration, providing a promising neural repair strategy for glaucomatous neurodegeneration.
    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
    缺血再灌注(I/R)损伤是影响脑卒中预后的关键因素。炎症反应,氧化应激,神经元凋亡是影响I/R损伤进展的主要因素。Farrarrol(FAR)是一种天然化合物,可以有效抑制炎症反应和氧化应激。然而,FAR在脑I/R损伤中的作用尚不清楚。在这项研究中,我们发现FAR可以降低I/R损伤后的脑损伤和神经元活力。同时,FAR的给药也降低了脑损伤后小胶质细胞的炎症反应。机械上,FAR治疗通过增强cAMP反应元件结合蛋白(CREB)的激活来增加下游神经营养因子和抗凋亡基因的表达,从而直接减少氧糖剥夺/复氧(OGD/R)后的神经元死亡。此外,FAR降低核因子κB(NF-κB)和丝裂原活化蛋白激酶(MAPK)信号通路的激活,抑制小胶质细胞激活,并减少OGD/R处理或LPS刺激后小胶质细胞中炎性细胞因子的产生。FAR受损的炎症反应直接促进OGD/R后神经元的存活。总之,FAR通过上调CREB表达和减轻神经炎症直接减少神经元死亡,对脑I/R损伤具有保护作用。因此,FAR可能是治疗脑I/R损伤的潜在有效药物。
    Ischemia-reperfusion (I/R) injury is a key influencing factor in the outcome of stroke. Inflammatory response, oxidative stress, and neuronal apoptosis are among the main factors that affect the progression of I/R injury. Farrerol (FAR) is a natural compound that can effectively inhibit the inflammatory response and oxidative stress. However, the role of FAR in cerebral I/R injury remains unknown. In this study, we found that FAR reduced brain injury and neuronal viability after cerebral I/R injury. Meanwhile, administration of FAR also reduced the inflammatory response of microglia after brain injury. Mechanistically, FAR treatment directly reduced neuronal death after oxygen glucose deprivation/re-oxygenation (OGD/R) through enhancing cAMP-response element binding protein (CREB) activation to increase the expression of downstream neurotrophic factors and anti-apoptotic genes. Moreover, FAR decreased the activation of nuclear factor kappa-B (NF-κB) and mitogen-activated protein kinase (MAPK) signaling pathways, inhibited microglia activation, and reduced the production of inflammatory cytokines in microglia after OGD/R treatment or LPS stimulation. The compromised inflammatory response by FAR directly promoted the survival of neurons after OGD/R. In conclusion, FAR exerted a protective effect on cerebral I/R injury by directly decreasing neuronal death through upregulating CREB expression and attenuating neuroinflammation. Therefore, FAR could be a potentially effective drug for the treatment of cerebral I/R injury.
    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
    缺血性中风导致神经元丢失和长期功能缺陷。不幸的是,保护神经元和促进功能恢复的有效方法仍然不可用。少突胶质细胞(OLs),中枢神经系统中的髓鞘细胞,易受氧气和营养剥夺的影响,缺血性中风后发生变性。从技术上讲,从少突胶质前体细胞(OPCs)的分化可以产生新的OLs和髓鞘。然而,缺血性卒中后髓磷脂动力学及其功能意义仍知之甚少。这里,我们报道了在缺血性卒中的人脑切片的病变中大量的脱氮轴突,伴随着神经元密度的降低,提示神经元丢失与卒中病变的髓磷脂缺陷相关。了解卒中后髓鞘动力学的纵向变化,我们标记并追踪预先存在或新形成的髓鞘,分别,通过使用细胞特异性遗传方法。我们的结果表明,在短暂性大脑中动脉阻塞(tMCAO)小鼠模型中,中风后2周出现大量的OLs死亡和髓磷脂丢失。相比之下,卒中后4周和8周髓鞘再生仍然不足。值得注意的是,神经元损失和功能损伤进一步恶化,在老年大脑,伴随着正在消失的新一代髓鞘。了解髓鞘再生和神经元存活之间的因果关系,我们通过条件缺失Olig2(正调节因子)或毒蕈碱受体1(M1R,OPCs中的负调节器)。删除Olig2会抑制髓鞘再生,抑制tMCAO后神经元存活和功能恢复。相反,tMCAO后通过M1RcKO增强髓鞘再生或治疗前髓鞘形成药物clemastine保持白质完整性和神经元存活,加速功能恢复。一起,我们的研究结果表明,增强髓鞘形成是缺血性卒中后保护神经元和促进功能恢复的有前景的策略.
    Ischaemic stroke causes neuron loss and long-term functional deficits. Unfortunately, effective approaches to preserving neurons and promoting functional recovery remain unavailable. Oligodendrocytes, the myelinating cells in the CNS, are susceptible to oxygen and nutrition deprivation and undergo degeneration after ischaemic stroke. Technically, new oligodendrocytes and myelin can be generated by the differentiation of oligodendrocyte precursor cells (OPCs). However, myelin dynamics and their functional significance after ischaemic stroke remain poorly understood. Here, we report numerous denuded axons accompanied by decreased neuron density in sections from ischaemic stroke lesions in human brain, suggesting that neuron loss correlates with myelin deficits in these lesions. To investigate the longitudinal changes in myelin dynamics after stroke, we labelled and traced pre-existing and newly-formed myelin, respectively, using cell-specific genetic approaches. Our results indicated massive oligodendrocyte death and myelin loss 2 weeks after stroke in the transient middle cerebral artery occlusion (tMCAO) mouse model. In contrast, myelin regeneration remained insufficient 4 and 8 weeks post-stroke. Notably, neuronal loss and functional impairments worsened in aged brains, and new myelin generation was diminished. To analyse the causal relationship between remyelination and neuron survival, we manipulated myelinogenesis by conditional deletion of Olig2 (a positive regulator) or muscarinic receptor 1 (M1R, a negative regulator) in OPCs. Deleting Olig2 inhibited remyelination, reducing neuron survival and functional recovery after tMCAO. Conversely, enhancing remyelination by M1R conditional knockout or treatment with the pro-myelination drug clemastine after tMCAO preserved white matter integrity and neuronal survival, accelerating functional recovery. Together, our findings demonstrate that enhancing myelinogenesis is a promising strategy to preserve neurons and promote functional recovery after ischaemic 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: Letter
    作为唯一被FDA批准的临床溶栓药物,组织型纤溶酶原激活剂(tPA)是超早期缺血性卒中(IS)的良好标准急性治疗方法。tPA形成阿替普酶的活性成分,重组组织型纤溶酶原激活剂(rtPA),以其血管内溶栓活性而闻名。然而,tPA在中枢神经系统(CNS)中的多方面功能具有未开发的潜力。目前,越来越多的研究探索了tPA在神经系统疾病中的神经保护功能,尤其是急性缺血性卒中(AIS)。一系列研究表明,tPA具有抗兴奋性毒性,神经营养,和对神经元的抗凋亡作用;它还参与神经元可塑性,轴突再生,和脑部炎症过程,但如何深刻理解潜在的机制,最大限度地利用tPA似乎是当务之急。因此,需要更多的工作来阐明tPA在卒中发病后如何发挥更多不同的功能.在这篇评论中,我们将重点放在关于tPA为什么以及如何促进缺血性神经元存活的可能假设上。文本提供了tPA功能的整体图景,并列举了未来的考虑因素,这可能会引起更多关注tPA在AIS中的治疗潜力。
    As the only clinical thrombolytic drug approved by the FDA, tissue-type plasminogen activator (tPA) is the good standard acute treatment against ischemic stroke (IS) during the super-early stage. tPA forms the active principle of alteplase, a recombinant tissue-type plasminogen activator (rtPA), which is well known for its intravascular thrombolytic activity. However, the multifaceted functions of tPA in the central nervous system (CNS) hold untapped potential. Currently, increasing studies have explored the neuroprotective function of tPA in neurological diseases, particularly in acute ischemic stroke (AIS). A series of studies have indicated that tPA has anti-excitotoxic, neurotrophic, and anti-apoptotic effects on neurons; it is also involved in neuronal plasticity, axonal regeneration, and cerebral inflammatory processes, but how to deeply understand the underlying mechanism and take maximum advantage of tPA seems to be urgent. Therefore, more work is needed to illuminate how tPA performs with more diverse functions after stroke onset. In this comment, we focus on possible hypotheses about why and how tPA promotes ischemic neuronal survival in a comprehensive view. The text provides a holistic picture of the functions of tPA and enlists the considerations for the future, which might attract more attention toward the therapeutic potential of tPA in AIS.
    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
    本研究旨在破译糖蛋白非转移性黑色素瘤蛋白B(GPNMB)对新生儿缺氧缺血性脑病(NHIE)的影响及其可能的分子机制。建立7日龄大鼠缺氧缺血(HI)模型,然后,进行Zea-Longa评分和Nissl染色以测量HI后的脑损伤。此外,基因测序用于检测差异表达基因(DEGs),然后,基因本体论和京都百科全书的基因和基因组数据库用于确定DEG的功能。此外,在SY5Y细胞和人胎儿神经元中建立了氧葡萄糖剥夺(OGD)模型,然后,通过定量实时聚合酶链反应验证GPNMB水平.此外,GPNMB干扰后应用甲基噻唑基四唑和细胞计数试剂盒-8测定。最后,使用SpliceGrapher软件分析GPNMB表达的选择性剪接。结果表明,HI可引起大鼠明显的神经功能缺损和神经元损伤。此外,GPNMB是DEGs中最明显的上调基因。此外,OGD后,SY5Y和胎儿神经元中GPNMB显著上调,和GPNMB-si促进细胞活力和数量的增加。此外,我们发现GPNMB选择性剪接类型是选择性3\'剪接位点,在143382985:143404102中具有选择性剪接位点。在这里,GPNMB通过选择性剪接促进NHIE后神经元存活的关键调节机制。
    This study aimed to decipher the effect of glycoprotein nonmetastatic melanoma protein B (GPNMB) on neonatal hypoxic-ischemic encephalopathy (NHIE) and its potential molecular mechanism. The hypoxic-ischemic (HI) model was established in 7-day-old rats, and then, Zea-Longa scores and Nissl staining were performed to measure brain damage post-HI. In addition, gene sequencing was used to detect the differential expression genes (DEGs), and then, Gene Ontology and Kyoto Encyclopedia of Genes and Genomes databases were used to determine the function of DEGs. Furthermore, an oxygen-glucose deprivation (OGD) model was developed in SY5Y cells and human fetal neurons, and then, the level of GPNMB was verified by quantitative real-time polymerase chain reaction. In addition, methyl thiazolyl tetrazolium and cell counting kit-8 assays were applied after GPNMB interference. Finally, the alternative splicing of GPNMB expression was analyzed using Splice Grapher software. The results indicated that HI induced marked neurological impairment and neuron injury in rats. Also, GPNMB was the most obviously upregulated gene in DEGs. Additionally, GPNMB was upregulated significantly in SY5Y and fetal neurons after OGD, and GPNMB-si promoted an increase in cell viability and number. Moreover, we found that the GPNMB alternative splicing type was the Alternative 3\' splice site, with the alternative splicing site in 143382985:143404102. Herein, GPNMB promotes a crucial regulatory mechanism with alternative splicing for neuronal survival after NHIE.
    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
    帕金森病(PD)的主要特征是黑质致密质(SNpc)中多巴胺能神经元的进行性变性和过度激活的小胶质细胞和星形胶质细胞介导的神经炎症。据报道,NLRC5(核苷酸结合寡聚化结构域样受体家族caspase募集结构域)参与各种免疫疾病,但其在神经退行性疾病中的作用尚不清楚。在目前的研究中,我们发现,NLRC5的表达在1-甲基-4-苯基-1,2,3,6-四氢吡啶盐酸盐(MPTP)诱导的PD小鼠的黑质纹状体轴中增加,以及在原代星形胶质细胞中,小胶质细胞和神经元暴露于不同的神经毒性刺激。在急性MPTP诱导的PD模型中,NLRC5缺乏可显着减少多巴胺能系统变性并改善运动缺陷和纹状体炎症。此外,我们发现NLRC5缺乏降低了促炎基因IL-1β的表达,用神经炎性刺激处理的原代小胶质细胞和原代星形胶质细胞中的IL-6,TNF-α和COX2,并减少了响应于LPS处理的混合神经胶质细胞中的炎症反应。此外,NLRC5缺陷抑制了NF-κB和MAPK信号通路的激活,并增强了混合胶质细胞中AKT-GSK-3β和AMPK信号的激活。此外,NLRC5缺陷增加了用MPP+或来自LPS刺激的混合神经胶质细胞的条件培养基处理的原代神经元的存活,并促进了NF-κB和AKT信号通路的激活。此外,与健康受试者相比,PD患者血液中NLRC5的mRNA表达降低。因此,我们建议NLRC5促进PD的神经炎症和多巴胺能变性,并可能作为神经胶质激活的标志.
    Parkinson\'s disease (PD) is mainly characterized by the progressive degeneration of dopaminergic neurons in the substantia nigra pars compacta (SNpc) and neuroinflammation mediated by overactivated microglia and astrocytes. NLRC5 (nucleotide-binding oligomerization domain-like receptor family caspase recruitment domain containing 5) has been reported to participate in various immune disorders, but its role in neurodegenerative diseases remains unclear. In the current study, we found that the expression of NLRC5 was increased in the nigrostriatal axis of mice with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine hydrochloride (MPTP)-induced PD, as well as in primary astrocytes, microglia and neurons exposed to different neurotoxic stimuli. In an acute MPTP-induced PD model, NLRC5 deficiency significantly reduced dopaminergic system degeneration and ameliorated motor deficits and striatal inflammation. Furthermore, we found that NLRC5 deficiency decreased the expression of the proinflammatory genes IL-1β, IL-6, TNF-α and COX2 in primary microglia and primary astrocytes treated with neuroinflammatory stimuli and reduced the inflammatory response in mixed glial cells in response to LPS treatment. Moreover, NLRC5 deficiency suppressed activation of the NF-κB and MAPK signaling pathways and enhanced the activation of AKT-GSK-3β and AMPK signaling in mixed glial cells. Furthermore, NLRC5 deficiency increased the survival of primary neurons treated with MPP+ or conditioned medium from LPS-stimulated mixed glial cells and promoted activation of the NF-κB and AKT signaling pathways. Moreover, the mRNA expression of NLRC5 was decreased in the blood of PD patients compared to healthy subjects. Therefore, we suggest that NLRC5 promotes neuroinflammation and dopaminergic degeneration in PD and may serve as a marker of glial activation.
    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
    急性缺血性卒中(AIS)是全球范围内严重且危及生命的疾病。尽管溶栓或血管内血栓切除术,相当比例的AIS患者有不良临床结局.此外,现有的抗血小板和抗凝药物治疗二级预防策略不能充分降低缺血性卒中复发的风险.因此,探索这样做的新机制代表了预防和治疗AIS的迫切需要。最近的研究发现,蛋白质糖基化在AIS的发生和结果中起着至关重要的作用。作为一种常见的共翻译和翻译后修饰,蛋白质糖基化通过调节蛋白质或酶的活性和功能参与多种生理和病理过程。蛋白质糖基化涉及缺血性中风中脑栓塞的两个原因:动脉粥样硬化和心房颤动。缺血性中风后,脑蛋白糖基化水平变得动态调节,通过影响炎症反应显著影响卒中结局,兴奋毒性,神经元凋亡,和血脑屏障破坏。针对卒中发生和进展中糖基化的药物可能代表了一种新的治疗思路。在这次审查中,我们关注糖基化如何影响AIS的发生和结果的可能观点。然后,我们提出了糖基化作为未来AIS患者的治疗药物靶标和预后标志物的潜力。
    Acute ischemic stroke (AIS) is a serious and life-threatening disease worldwide. Despite thrombolysis or endovascular thrombectomy, a sizeable fraction of patients with AIS have adverse clinical outcomes. In addition, existing secondary prevention strategies with antiplatelet and anticoagulant drugs therapy are not able to adequately decrease the risk of ischemic stroke recurrence. Thus, exploring novel mechanisms for doing so represents an urgent need for the prevention and treatment of AIS. Recent studies have discovered that protein glycosylation plays a critical role in the occurrence and outcome of AIS. As a common co- and post-translational modification, protein glycosylation participates in a wide variety of physiological and pathological processes by regulating the activity and function of proteins or enzymes. Protein glycosylation is involved in two causes of cerebral emboli in ischemic stroke: atherosclerosis and atrial fibrillation. Following ischemic stroke, the level of brain protein glycosylation becomes dynamically regulated, which significantly affects stroke outcome through influencing inflammatory response, excitotoxicity, neuronal apoptosis, and blood-brain barrier disruption. Drugs targeting glycosylation in the occurrence and progression of stroke may represent a novel therapeutic idea. In this review, we focus on possible perspectives about how glycosylation affects the occurrence and outcome of AIS. We then propose the potential of glycosylation as a therapeutic drug target and prognostic marker for AIS patients in the future.
    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
    肌细胞增强因子2(MEF2)转录因子家族包含四个高度保守的成员,在神经系统中起重要作用。它们出现在发育中的大脑中精确定义的时间范围内,以打开和关闭影响生长的基因,修剪和神经元的存活。已知MEF2决定神经元发育,突触可塑性和限制海马突触的数量,从而影响学习和记忆的形成。在初级神经元中,已知外界刺激或应激条件对MEF2活性的负调节会诱导细胞凋亡,尽管MEF2的促凋亡或抗凋亡作用取决于神经元的成熟期。相比之下,MEF2转录活性的增强可在体外和神经退行性疾病的临床前模型中保护神经元免于凋亡死亡。越来越多的证据将这种转录因子置于与年龄依赖性神经元功能障碍或逐渐但不可逆的神经元丢失相关的许多神经病理学的中心。在这项工作中,我们讨论了MEF2在发育过程中和成年期影响神经元存活的功能改变可能与神经精神疾病有关。
    The family of myocyte enhancer factor 2 (MEF2) transcription factors comprises four highly conserved members that play an important role in the nervous system. They appear in precisely defined time frames in the developing brain to turn on and turn off genes affecting growth, pruning and survival of neurons. MEF2s are known to dictate neuronal development, synaptic plasticity and restrict the number of synapses in the hippocampus, thus affecting learning and memory formation. In primary neurons, negative regulation of MEF2 activity by external stimuli or stress conditions is known to induce apoptosis, albeit the pro or antiapoptotic action of MEF2 depends on the neuronal maturation stage. By contrast, enhancement of MEF2 transcriptional activity protects neurons from apoptotic death both in vitro and in preclinical models of neurodegenerative diseases. A growing body of evidence places this transcription factor in the center of many neuropathologies associated with age-dependent neuronal dysfunctions or gradual but irreversible neuron loss. In this work, we discuss how the altered function of MEF2s during development and in adulthood affecting neuronal survival may be linked to neuropsychiatric disorders.
    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
    已知神经营养因子是嗅鞘细胞(OEC)的神经保护作用的主要来源。然而,来自OECs的神经营养因子的形式尚不完全清楚。我们之前的研究表明,OECs可以分泌外泌体(OECs-Exo),通过切换巨噬细胞/小胶质细胞的表型提供神经保护。考虑到外泌体也可以被神经元吸收,我们探讨了OECs-Exo对神经元存活的直接影响及其潜在机制。电子显微镜,纳米交通分析,和Western印迹用于鉴定OECs-Exo。通过流式细胞术和TUNEL染色测试OECs-Exo对神经元存活的影响。采用Westernblotting和ELISA检测纯化OECs-Exo中的神经营养因子。我们首先分离了OECs-Exo,发现OECs-Exo对响应TNF-α攻击的神经元存活具有保护作用。然后在OECs-Exo,其在神经元中的受体TrkB被OECs-Exo处理激活。此外,我们证明OECs部分通过外泌体来源的BDNF阻止TNF-α诱导的神经元凋亡。我们的数据表明,OEC部分通过OEC-Exo衍生的BDNF减弱TNF-α诱导的神经元凋亡,这可能为基于OEC-Exo的治疗的神经保护作用提供新的策略。
    It is known that neurotrophic factors are a major source of the neuroprotective effects of olfactory ensheathing cells (OECs). However, the form of neurotrophic factors that originate from OECs is not fully understood. Our previous study demonstrated that OECs could secrete exosome (OECs-Exo), which provided neuroprotection by switching the phenotype of macrophages/microglia. Considering that exosomes could also be taken up by neurons, we explored the direct effect of OECs-Exo on neuronal survival and the underlying mechanism. Electron microscopy, nano-traffic analysis, and Western blotting were applied to identify the OECs-Exo. The effect of OECs-Exo on neuronal survival was tested by flow cytometry and TUNEL staining. Western blotting and ELISA were used to detect neurotrophic factors in purified OECs-Exo. We first isolated OECs-Exo and found that OECs-Exo exerted protective effects on neuronal survival in response to TNF-α challenge. Brain-derived neurotrophic factor (BDNF) was then identified in OECs-Exo, and its receptor TrkB in neurons was activated by OECs-Exo treatment. Furthermore, we demonstrated that OECs prevented TNF-α-induced apoptosis in neurons partially through exosome-derived BDNF. Our data showed that OECs attenuated TNF-α-induced apoptosis in neurons partially through OEC-Exo-derived BDNF, which might provide a novel strategy for the neuroprotective effect of OEC-Exo-based 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
    由可扩散蛋白介导的血管和神经细胞之间的细胞间通讯最近已成为神经发育的关键内在程序。然而,血管平滑肌细胞(VSMC)分泌组是否调节神经回路的连接仍然未知。这里,我们显示来自脑VSMC培养物的条件培养基增强多种神经元功能,如神经生成,神经元成熟,和生存,从而改善电路连接。然而,蛋白质通过加热变性损害了这些影响。供体VSMC分泌组和受体神经元转录组的组合组学分析显示,细胞外基质受体信号传导和粘附分子整联蛋白结合的重叠途径介导VSMC依赖性神经元发育。此外,我们发现人动脉VSMCs以多种方式促进神经元发育,包括扩大新生神经突起始的时间窗口,增加神经元密度,促进同步发射,而人脐静脉VSMC缺乏这种能力。这些体外数据表明,脑小动脉VSMC可能通过体内细胞间通讯携带神经发育的直接指导性信息。
    Intercellular communication between vascular and nerve cells mediated by diffusible proteins has recently emerged as a critical intrinsic program for neural development. However, whether the vascular smooth muscle cell (VSMC) secretome regulates the connectivity of neural circuits remains unknown. Here, we show that conditioned medium from brain VSMC cultures enhances multiple neuronal functions, such as neuritogenesis, neuronal maturation, and survival, thereby improving circuit connectivity. However, protein denaturation by heating compromised these effects. Combined omics analyses of donor VSMC secretomes and recipient neuron transcriptomes revealed that overlapping pathways of extracellular matrix receptor signaling and adhesion molecule integrin binding mediate VSMC-dependent neuronal development. Furthermore, we found that human arterial VSMCs promote neuronal development in multiple ways, including expanding the time window for nascent neurite initiation, increasing neuronal density, and promoting synchronized firing, whereas human umbilical vein VSMCs lack this capability. These in vitro data indicate that brain arteriolar VSMCs may carry direct instructive information for neural development through intercellular communication in vivo.
    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