neuronal differentiation

神经元分化
  • 文章类型: Journal Article
    为了研究生理相关背景下神经元分化的细胞行为,分化神经元必须在其天然组织环境中进行研究。这里,我们描述了一种可访问的协议,用于离体胚胎鸡脊髓切片培养物中分化神经元的荧光活成像,这有助于长期观察发育组织内的单个细胞。
    To investigate the cell behavior underlying neuronal differentiation in a physiologically relevant context, differentiating neurons must be studied in their native tissue environment. Here, we describe an accessible protocol for fluorescent live imaging of differentiating neurons within ex vivo embryonic chicken spinal cord slice cultures, which facilitates long-term observation of individual cells within developing tissue.
    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
    干细胞疗法有可能解决组织修复和再生中尚未解决的问题,特别是在神经组织中。然而,尚未找到最佳来源。越来越多的证据表明,间充质干细胞(MSC)在体内产生的积极作用可能不仅归因于它们的可塑性,而且归因于分泌的分子,包括细胞外囊泡(EV)和细胞外基质(ECM)。MSCs产生的营养效应可能揭示了开发有效组织修复策略的关键。包括基于脑植入物或其他可植入神经电极的方法。在这个意义上,MSC将在未来变得越来越有价值和需要。胎盘是用于保护和支持胎儿的临时器官。同时,胎盘代表了丰富且极其方便的MSCs来源。尽管如此,与从其他来源分离的MSCs相比,胎盘来源的MSCs(P-MSCs)仍未得到充分研究。这篇综述概述了有限的文献,描述了P-MSC衍生的生物材料的神经再生作用,并倡导利用这种尚未开发的来源对人类再生疗法的潜力。
    Stem cell therapy has the potential to meet unsolved problems in tissue repair and regeneration, particularly in the neural tissues. However, an optimal source has not yet been found. Growing evidence indicates that positive effects produced in vivo by mesenchymal stem cells (MSCs) can be due not only to their plasticity but also to secreted molecules including extracellular vesicles (EVs) and the extracellular matrix (ECM). Trophic effects produced by MSCs may reveal the key to developing effective tissue-repair strategies, including approaches based on brain implants or other implantable neural electrodes. In this sense, MSCs will become increasingly valuable and needed in the future. The placenta is a temporary organ devoted to protecting and supporting the fetus. At the same time, the placenta represents an abundant and extremely convenient source of MSCs. Nonetheless, placenta-derived MSCs (P-MSCs) remain understudied as compared to MSCs isolated from other sources. This review outlines the limited literature describing the neuroregenerative effects of P-MSC-derived biomaterials and advocates for exploiting the potential of this untapped source for human regenerative therapies.
    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
    乙二醛酶1(Glo1)是解毒甲基乙二醛(MGO)的必需酶,糖酵解的细胞毒性副产物。越来越多的研究表明,Glo1在调节皮质发育和神经发生中具有重要作用,可能导致自闭症谱系障碍(ASD)受损时的发病机制。我们以前已经表明,产前暴露于非凋亡低剂量甲基汞(MeHg),一种环境污染物,在啮齿动物模型中诱导过早的皮质神经发生和ASD样行为。在这项研究中,我们旨在确定介导产前MeHg诱导的神经元早熟分化和异常神经发育的潜在分子机制。使用单细胞RNA测序(scRNA-seq)和实时定量PCR(RT-qPCR),我们发现,非凋亡剂量的产前甲基汞暴露显著降低了胚胎培养的放射状神经胶质前体(RGP)的Glo1基因表达.在培养的RGP中,Glo1表达的敲低增加了神经元的产生,以培养的RGP群体为代价,而Glo1的过表达使MeHg诱导的神经元分化恢复到正常水平。此外,我们发现,与MeHg和多种MGO清除剂或CREB抑制剂(iCREB)共同治疗可减轻MeHg诱导的过早神经元分化,增强Glo1和CREB在介导MeHg诱导的神经元分化中的作用。我们的研究结果表明,在皮质发育中,甲基汞暴露与ASD风险基因Glo1的表达之间存在直接联系。支持基因-环境相互作用在神经发育障碍的病因中的重要作用,比如ASD。
    Glyoxalase 1 (Glo1) is an essential enzyme to detoxify methylglyoxal (MGO), a cytotoxic byproduct of glycolysis. Accumulating studies have shown an important role of Glo1 in regulating cortical development and neurogenesis, potentially contributing to the pathogenesis of autism spectrum disorder (ASD) when impaired. We have previously shown that prenatal exposure to non-apoptotic low-dose methylmercury (MeHg), an environmental pollutant, induces premature cortical neurogenesis and ASD-like behaviors in a rodent model. In this study, we aimed to determine the underlying molecular mechanisms that mediate prenatal MeHg-induced premature neuronal differentiation and abnormal neurodevelopment. Using single-cell RNA sequencing (scRNA-seq) and real-time quantitative PCR (RT-qPCR), we found that prenatal MeHg exposure at a non-apoptotic dose significantly reduced Glo1 gene expression in embryonic cultured radial glia precursors (RGPs). In cultured RGPs, the knockdown of Glo1 expression increased neuronal production at the expense of the cultured RGPs population, while overexpression of Glo1 restored MeHg-induced neuronal differentiation back to normal levels. Furthermore, we found that co-treatment with both MeHg and multiple MGO scavengers or a CREB inhibitor (iCREB) mitigated MeHg-induced premature neuronal differentiation, reinforcing the role of Glo1 and CREB in mediating MeHg-induced neuronal differentiation. Our findings demonstrate a direct link between MeHg exposure and expression of an ASD risk gene Glo1 in cortical development, supporting the important role of gene-environment interaction in contributing to the etiology of neural developmental disorders, such as ASD.
    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:对神经元的研究是揭示神经系统复杂性的基础。来自啮齿动物的原代神经元培养物长期以来一直是实验研究的基石,然而,与非人性和伦理问题相关的限制促使了替代方案的发展。近年来,从人类诱导的多能干细胞(hiPSCs)中衍生神经元已成为一种强大的选择,为各种应用提供可扩展的细胞来源。来自hiPSCs的神经祖细胞(NPCs)可以有效地分化为功能性神经元,提供了体外研究人类神经生理和病理的平台。然而,在整个实验环境中,在实现一致和可重复的结果方面仍然存在挑战.
    方法:我们的目标是提供一个循序渐进的方法协议,用额外的指令和参数扩充现有的程序,指导研究人员取得可重复的结果。
    结果:我们概述了hiPSC衍生的NPCs分化为有电能力的神经元的程序,包括初始细胞密度,形态学,维护,和差异化。我们还描述了用于评估神经元表型的特定标记的分析,与电生理分析一起评估神经元兴奋性的生物物理特性。此外,我们对三种不同的化学方法进行了比较分析——氯化钾,N-甲基-D-天冬氨酸(NMDA),和双核蛋白-诱导神经元去极化,并评估它们对快速和慢速翻译后诱导的影响,转录,和转录后反应。
    结论:我们的方案为产生具有确定的电生理特性的可靠的人类神经元培养物提供了明确的指导,以研究体外神经元分化和模型疾病。
    BACKGROUND: The study of neurons is fundamental to unraveling the complexities of the nervous system. Primary neuronal cultures from rodents have long been a cornerstone of experimental studies, yet limitations related to their non-human nature and ethical concerns have prompted the development of alternatives. In recent years, the derivation of neurons from human-induced pluripotent stem cells (hiPSCs) has emerged as a powerful option, offering a scalable source of cells for diverse applications. Neural progenitor cells (NPCs) derived from hiPSCs can be efficiently differentiated into functional neurons, providing a platform to study human neural physiology and pathology in vitro. However, challenges persist in achieving consistent and reproducible outcomes across experimental settings.
    METHODS: Our aim is to provide a step-by-step methodological protocol, augmenting existing procedures with additional instructions and parameters, to guide researchers in achieving reproducible results.
    RESULTS: We outline procedures for the differentiation of hiPSC-derived NPCs into electrically competent neurons, encompassing initial cell density, morphology, maintenance, and differentiation. We also describe the analysis of specific markers for assessing neuronal phenotype, along with electrophysiological analysis to evaluate biophysical properties of neuronal excitability. Additionally, we conduct a comparative analysis of three different chemical methods-KCl, N-methyl-D-aspartate (NMDA), and bicuculline-to induce neuronal depolarization and assess their effects on the induction of both fast and slow post-translational, transcriptional, and post-transcriptional responses.
    CONCLUSIONS: Our protocol provides clear instructions for generating reliable human neuronal cultures with defined electrophysiological properties to investigate neuronal differentiation and model diseases in vitro.
    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
    神经干/祖细胞(NSPCs)在哺乳动物的整个生命中持续存在,响应各种病理生理刺激,在中枢神经系统修复中起着至关重要的作用。尽管许多研究已经阐明了斯ninocalcin2(STC2)在调节细胞分化过程中的作用,其在NSPCs分化中的具体功能尚不清楚。明确STC2在NSPC中的作用对于设计新策略以增强损伤后脑再生的内在潜力至关重要。我们的研究揭示了STC2在C57BL/6N小鼠脑室下区(SVZ)的NSPCs中的表达。在培养的SVZ衍生的NSPC中,STC2处理显著增加了Tuj1和DCX阳性细胞的数量。此外,侧脑室注射STC2促进NSPCs的神经元分化和向嗅球的迁移。相反,STC2敲低产生相反的效果。进一步的研究表明,STC2处理增强了培养的NSPCs中的AKT磷酸化,而STC2抑制阻碍AKT激活。值得注意的是,STC2诱导的神经元分化被AKT抑制剂GSK690693阻断,而AKT激活剂SC79逆转STC2敲低对神经元分化的影响.我们的发现表明,增强SVZ衍生的NSPCs中的STC2表达促进神经元分化,AKT调节可能是STC2信号传导的关键细胞内靶标。
    Neural stem/progenitor cells (NSPCs) persist in the mammalian subventricular zone (SVZ) throughout life, responding to various pathophysiological stimuli and playing a crucial role in central nervous system repair. Although numerous studies have elucidated the role of stanniocalcin 2 (STC2) in regulating cell differentiation processes, its specific function in NSPCs differentiation remains poorly understood. Clarifying the role of STC2 in NSPCs is essential for devising novel strategies to enhance the intrinsic potential for brain regeneration postinjury. Our study revealed the expression of STC2 in NSPCs derived from the SVZ of the C57BL/6N mouse. In cultured SVZ-derived NSPCs, STC2 treatment significantly increased the number of Tuj1 and DCX-positive cells. Furthermore, STC2 injection into the lateral ventricle promoted the neuronal differentiation of NSPCs and migration to the olfactory bulb. Conversely, the STC2 knockdown produced the opposite effect. Further investigation showed that STC2 treatment enhanced AKT phosphorylation in cultured NSPCs, whereas STC2 inhibition hindered AKT activation. Notably, the neuronal differentiation induced by STC2 was blocked by the AKT inhibitor GSK690693, whereas the AKT activator SC79 reversed the impact of STC2 knockdown on neuronal differentiation. Our findings indicate that enhancing STC2 expression in SVZ-derived NSPCs facilitates neuronal differentiation, with AKT regulation potentially serving as a key intracellular target of STC2 signaling.
    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
    通过重编程产生人诱导多能干细胞(iPSCs)是再生医学领域中的转化性变化,其导致了药物发现和细胞替代疗法的新可能性。已经建立了几种方案来将hiPSC分化为神经元谱系。然而,低分化效率是这些方法的主要缺点之一。这里,我们比较了在两种不同培养基中培养的iPSC的两种神经元分化方法的效率,StemFlex培养基(SFM)和基本8培养基(E8M)。结果表明,与在SFM中从iPSC产生的那些相比,在E8M中培养的iPSC在更短的时间内有效地产生不同类型的神经元,并且在培养物中没有未分化的非神经元细胞的生长。此外,通过确认成熟神经元标志物的表达,这些神经元在免疫细胞化学上被验证为功能单位(即,中子,β微管蛋白,和突触素I),和全细胞膜片钳记录。长读单细胞RNA测序证实,在第30天的神经元培养物中,除了少量GABA能神经元外,还存在上层和深层皮质层兴奋性和抑制性神经元亚型。通路分析表明,我们的方案触发了对体内神经元分化过程重要的信号传导转录网络。
    Generation of human induced pluripotent stem cells (iPSCs) through reprogramming was a transformational change in the field of regenerative medicine that led to new possibilities for drug discovery and cell replacement therapy. Several protocols have been established to differentiate hiPSCs into neuronal lineages. However, low differentiation efficiency is one of the major drawbacks of these approaches. Here, we compared the efficiency of two methods of neuronal differentiation from iPSCs cultured in two different culture media, StemFlex Medium (SFM) and Essential 8 Medium (E8M). The results indicated that iPSCs cultured in E8M efficiently generated different types of neurons in a shorter time and without the growth of undifferentiated non-neuronal cells in the culture as compared to those generated from iPSCs in SFM. Furthermore, these neurons were validated as functional units immunocytochemically by confirming the expression of mature neuronal markers (i.e., NeuN, Beta tubulin, and Synapsin I), and whole-cell patch-clamp recordings. Long-read single-cell RNA sequencing confirms the presence of upper and deep layer cortical layer excitatory and inhibitory neuronal subtypes in addition to small populations of GABAergic neurons in day 30 neuronal cultures. Pathway analysis indicated that our protocol triggers the signaling transcriptional networks important for the process of neuronal differentiation in vivo.
    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
    环状RNA(circularRNAs)在神经发生期间上调。circRNAs在哪里以及如何定位,以及它们在此过程中扮演的角色仍然难以捉摸。比较H9细胞和H9衍生的前脑(FB)神经元之间的核和细胞质circRNAs,我们发现,富含腺苷(A)的circRNAs的一个子集被限制在H9细胞核中,但在分化后输出到细胞溶胶中。circRNAs的这种亚细胞重新定位由poly(A)结合蛋白PABPC1调节。在H9核中,新产生的富含(A)的circRNAs被PABPC1结合并被核篮蛋白TPR捕获以阻止它们的输出。调节circRNAs中富含(A)的基序改变了它们的亚细胞定位,并且在H9胞质中引入富含(A)的circRNAs导致mRNA翻译抑制。此外,神经元分化后减少的核PABPC1能够输出富含(A)的circRNAs,包括circRTN4(2,3),这是神经突生长所必需的。这些发现揭示了circRNAs的亚细胞定位特征,将它们在神经发生过程中的加工和功能联系起来。
    Circular RNAs (circRNAs) are upregulated during neurogenesis. Where and how circRNAs are localized and what roles they play during this process have remained elusive. Comparing the nuclear and cytoplasmic circRNAs between H9 cells and H9-derived forebrain (FB) neurons, we identify that a subset of adenosine (A)-rich circRNAs are restricted in H9 nuclei but exported to cytosols upon differentiation. Such a subcellular relocation of circRNAs is modulated by the poly(A)-binding protein PABPC1. In the H9 nucleus, newly produced (A)-rich circRNAs are bound by PABPC1 and trapped by the nuclear basket protein TPR to prevent their export. Modulating (A)-rich motifs in circRNAs alters their subcellular localization, and introducing (A)-rich circRNAs in H9 cytosols results in mRNA translation suppression. Moreover, decreased nuclear PABPC1 upon neuronal differentiation enables the export of (A)-rich circRNAs, including circRTN4(2,3), which is required for neurite outgrowth. These findings uncover subcellular localization features of circRNAs, linking their processing and function during neurogenesis.
    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
    用于生物应用的量子点的关键要求之一是它们的表面修饰以用于非常特异性和增强的生物识别和摄取。为此,我们呈现的是明亮的绿色合成,源自芒果叶提取物的红色发射碳量子点(mQDs)。这些mQD与多巴胺静电缀合以形成mQDs-多巴胺(mQDs:DOPA)生物缀合物。mQDs的亮红色荧光用于癌细胞和非癌细胞系的生物成像和摄取,组织,和斑马鱼等体内模型。与心脏相比,mQDs在脑组织中的摄取最高,肾,还有肝脏.mQD:DOPA缀合物杀死乳腺癌细胞并增加上皮RPE-1细胞和斑马鱼的摄取。此外,mQDs:DOPA促进SH-SY5Y细胞向分化神经元的神经元分化。mQDs和mQDs:DOPA表现出更高的集体细胞迁移的潜力,暗示了他们作为下一代先进生物和生物医学应用工具的未来潜力。
    One of the crucial requirements of quantum dots for biological applications is their surface modification for very specific and enhanced biological recognition and uptake. Toward this end, we present the green synthesis of bright, red-emitting carbon quantum dots derived from mango leaf extract (mQDs). These mQDs are conjugated electrostatically with dopamine to form mQDs-dopamine (mQDs:DOPA) bioconjugates. Bright-red fluorescence of mQDs was used for bioimaging and uptake in cancerous and noncancerous cell lines, tissues, and in vivo models like zebrafish. mQDs exhibited the highest uptake in brain tissue compared to the heart, kidney, and liver. mQD:DOPA conjugates killed breast cancer cells and increased uptake in epithelial RPE-1 cells and zebrafish. Additionally, mQDs:DOPA promoted neuronal differentiation of SH-SY5Y cells to differentiated neurons. Both mQDs and mQDs:DOPA exhibited the potential for higher collective cell migrations, implicating their future potential as next-generation tools for advanced biological and biomedical applications.
    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
    抑制因子-1沉默转录因子(REST)是形成成熟神经元所必需的。REST失调是与神经系统疾病相关的神经变性的关键机制的基础。然而,导致REST介导的关键神经发生基因沉默改变的机制尚不清楚.这里,我们显示BRCA1关联ATM激活器1(BRAT1),与神经退行性疾病相关的基因,是神经元分化过程中激活REST响应基因所必需的。我们发现整合子复合物的INTS11和INTS9亚基作为独特的三聚体复合物与BRAT1相互作用,以在分化过程中激活关键的神经元基因。BRAT1耗竭导致在关键神经元基因上的REST驻留的持久性,破坏NT2细胞分化为星形胶质细胞和神经元细胞。我们确定了BRAT1和INTS11共同占据这些基因的启动子区域,并指出了BRAT1在招募INTS11到其启动子中的作用。BRAT1中的致病突变减少了其与INTS11/INTS9的关联,将疾病表型的表现与BRAT1/INTS11/INTS9复合物的关键神经元基因的转录激活缺陷联系起来。最后,小鼠胚胎干细胞中Brat1的缺失导致神经元分化测定的缺陷。重要的是,而与野生型BRAT1重建恢复神经元分化,添加BRAT1突变体不能与INTS11/INTS9相关联,也不能挽救神经元表型.一起来看,我们的研究强调了BRAT1与INTS11和INTS9在神经系统发育中的相关性.
    Repressor element-1 silencing transcription factor (REST) is required for the formation of mature neurons. REST dysregulation underlies a key mechanism of neurodegeneration associated with neurological disorders. However, the mechanisms leading to alterations of REST-mediated silencing of key neurogenesis genes are not known. Here, we show that BRCA1 Associated ATM Activator 1 (BRAT1), a gene linked to neurodegenerative diseases, is required for the activation of REST-responsive genes during neuronal differentiation. We find that INTS11 and INTS9 subunits of Integrator complex interact with BRAT1 as a distinct trimeric complex to activate critical neuronal genes during differentiation. BRAT1 depletion results in persistence of REST residence on critical neuronal genes disrupting the differentiation of NT2 cells into astrocytes and neuronal cells. We identified BRAT1 and INTS11 co-occupying the promoter region of these genes and pinpoint a role for BRAT1 in recruiting INTS11 to their promoters. Disease-causing mutations in BRAT1 diminish its association with INTS11/INTS9, linking the manifestation of disease phenotypes with a defect in transcriptional activation of key neuronal genes by BRAT1/INTS11/INTS9 complex. Finally, loss of Brat1 in mouse embryonic stem cells leads to a defect in neuronal differentiation assay. Importantly, while reconstitution with wild-type BRAT1 restores neuronal differentiation, the addition of a BRAT1 mutant is unable to associate with INTS11/INTS9 and fails to rescue the neuronal phenotype. Taken together, our study highlights the importance of BRAT1 association with INTS11 and INTS9 in the development of the 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: Systematic Review
    脊髓损伤(SCI)是一种严重的医学疾病。寻找有效的治疗方法仍然是一个持续的挑战。目前的治疗方法,不幸的是,无法充分改善神经功能,往往导致终身残疾。本系统综述和荟萃分析使用犬模型评估干细胞治疗SCI的有效性。它还探索了实施干细胞治疗的最佳方案。从2000年到2022年10月进行了全面的研究搜索。这项研究集中在五个结果:运动功能评分,组织病理学,IHC,westernblot,和SEP。结果表明,在接受干细胞治疗的狗中,SCI后的运动得到了显着改善。与对照组相比,该疗法还导致治疗的狗的Olby评分平均增加3.15分。这些发现凸显了干细胞治疗作为一种有前途的SCI治疗的潜力。荟萃分析表明,使用骨髓干细胞,在体外进行神经分化,应用手术植入或鞘内给药途径,将基质胶与干细胞结合起来,在开始治疗前等待两周可以提高SCI治疗的有效性。
    Spinal cord injury (SCI) is a serious medical condition. The search for an effective cure remains a persistent challenge. Current treatments, unfortunately, are unable to sufficiently improve neurological function, often leading to lifelong disability. This systematic review and meta-analysis evaluated the effectiveness of stem cell therapy for SCI using canine models. It also explored the optimal protocol for implementing stem cell therapy. A comprehensive search of studies was conducted from 2000 to October 2022. This study focused on five outcomes: motor function score, histopathology, IHC, western blot, and SEP. The results demonstrated a significant improvement in locomotion post-SCI in dogs treated with stem cell therapy. The therapy also led to an average increase of 3.15 points in the Olby score of the treated dogs compared to the control group. These findings highlights stem cell therapy\'s potential as a promising SCI treatment. The meta-analysis suggests that using bone marrow stem cells, undergoing neural differentiation in vitro, applying a surgical implantation or intrathecal route of administration, associating matrigel in combination with stem cells, and a waiting period of two weeks before starting treatment can enhance SCI treatment effectiveness.
    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