neural stem cell (NSC)

  • 文章类型: Journal Article
    神经毒物是可导致对神经系统不利的结构或功能影响的物质。这些可能是化学的,生物,或物理试剂,可以穿过血脑屏障损伤神经元或干扰神经系统和其他器官之间的复杂相互作用。考虑到社会政策,公共卫生,和医学,有必要确保严格的神经毒性测试。虽然最常见的神经毒性测试涉及使用动物模型,向基于干细胞的平台的转变可能在临床和药物研究中提供更准确的生物学替代方案。考虑到这一点,本文的目的是回顾使用基于干细胞的方法评估神经毒物的当前技术和最新进展,强调发育神经毒物(DNTs),因为它们最有可能导致不可逆的脑功能严重损害。在下一节中,讨论了开发用于研究神经细胞命运和发育神经毒性的新型预测模型方法的尝试。最后,本文最后讨论了未来在发育神经毒性测试中使用计算机方法,以及监管机构在促进空间内进步方面的作用。
    Neurotoxicants are substances that can lead to adverse structural or functional effects on the nervous system. These can be chemical, biological, or physical agents that can cross the blood brain barrier to damage neurons or interfere with complex interactions between the nervous system and other organs. With concerns regarding social policy, public health, and medicine, there is a need to ensure rigorous testing for neurotoxicity. While the most common neurotoxicity tests involve using animal models, a shift towards stem cell-based platforms can potentially provide a more biologically accurate alternative in both clinical and pharmaceutical research. With this in mind, the objective of this article is to review both current technologies and recent advancements in evaluating neurotoxicants using stem cell-based approaches, with an emphasis on developmental neurotoxicants (DNTs) as these have the most potential to lead to irreversible critical damage on brain function. In the next section, attempts to develop novel predictive model approaches for the study of both neural cell fate and developmental neurotoxicity are discussed. Finally, this article concludes with a discussion of the future use of in silico methods within developmental neurotoxicity testing, and the role of regulatory bodies in promoting advancements within the space.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Editorial
    暂无摘要。
    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
    干细胞的行为受到其小生境中的机械提示的调节,这些机械提示由于细胞外基质(ECM)重塑而不断变化,血流施加的脉动机械应力,和/或细胞迁移。然而,目前还不清楚机械线索的动力学如何影响干细胞谱系的承诺,尤其是在3D微环境中,机械传感与2D微环境不同。在本研究中,我们研究了随时间变化的机械信号如何调节早期生长反应1基因(Egr1)的表达,我们最近发现它是机械敏感性神经干细胞(NSC)谱系承诺的3D基质特异性介质。具体来说,我们暂时控制了Ras同源家族成员A(RhoA)的活性,众所周知,这在机械传导中起着核心作用,使用我们以前开发的拟南芥隐色素2光活化系统。有趣的是,脉冲RhoA激活仅在坚硬的3D凝胶中诱导Egr1上调,而静态光刺激诱导Egr1表达在广泛的3D凝胶硬度范围内增加。肌动蛋白组装抑制限制了RhoA激活后的Egr1上调,暗示RhoA信号传导需要一个肌动蛋白参与的过程来上调Egr1。始终如一,静态光RhoA激活而不是脉冲光激活限制了软凝胶中的神经发生。我们的发现表明,RhoA激活的动力学以依赖于基质刚度的方式影响3D基质中Egr1介导的干细胞命运。
    The behavior of stem cells is regulated by mechanical cues in their niche that continuously vary due to extracellular matrix (ECM) remodeling, pulsated mechanical stress exerted by blood flow, and/or cell migration. However, it is still unclear how dynamics of mechanical cues influence stem cell lineage commitment, especially in a 3D microenvironment where mechanosensing differs from that in a 2D microenvironment. In the present study, we investigated how temporally varying mechanical signaling regulates expression of the early growth response 1 gene (Egr1), which we recently discovered to be a 3D matrix-specific mediator of mechanosensitive neural stem cell (NSC) lineage commitment. Specifically, we temporally controlled the activity of Ras homolog family member A (RhoA), which is known to have a central role in mechanotransduction, using our previously developed Arabidopsis thaliana cryptochrome-2-based optoactivation system. Interestingly, pulsed RhoA activation induced Egr1 upregulation in stiff 3D gels only, whereas static light stimulation induced an increase in Egr1 expression across a wide range of 3D gel stiffnesses. Actin assembly inhibition limited Egr1 upregulation upon RhoA activation, implying that RhoA signaling requires an actin-involved process to upregulate Egr1. Consistently, static-light RhoA activation rather than pulsed-light activation restricted neurogenesis in soft gels. Our findings indicate that the dynamics of RhoA activation influence Egr1-mediated stem cell fate within 3D matrices in a matrix stiffness-dependent manner.
    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
    神经干细胞(NSC)具有自我更新和多潜能特性。成年神经干细胞位于成年大脑的两个神经源性区域:侧脑室的脑室-脑室下区(V-SVZ)和海马齿状回的颗粒下区。成年神经干细胞的维持和分化受到内在和外在信号的调节,这些信号可以通过成年神经干细胞中一些关键因素的表达来整合。已显示许多转录因子在成人大脑中NSC细胞命运转变的转录调节中起着重要作用。表观遗传调节剂也已成为NSC监管的关键参与者,神经祖细胞及其通过表观遗传修饰(包括DNA甲基化)分化的后代,组蛋白修饰,染色质重塑和RNA介导的转录调控。这篇小型综述主要集中在成人神经发生过程中成人神经干细胞的表观遗传调控。与这些过程中的转录调控相结合。
    Neural stem cells (NSCs) exhibit self-renewing and multipotential properties. Adult NSCs are located in two neurogenic regions of adult brain: the ventricular-subventricular zone (V-SVZ) of the lateral ventricle and the subgranular zone of the dentate gyrus in the hippocampus. Maintenance and differentiation of adult NSCs are regulated by both intrinsic and extrinsic signals that may be integrated through expression of some key factors in the adult NSCs. A number of transcription factors have been shown to play essential roles in transcriptional regulation of NSC cell fate transitions in the adult brain. Epigenetic regulators have also emerged as key players in regulation of NSCs, neural progenitor cells and their differentiated progeny via epigenetic modifications including DNA methylation, histone modifications, chromatin remodeling and RNA-mediated transcriptional regulation. This minireview is primarily focused on epigenetic regulations of adult NSCs during adult neurogenesis, in conjunction with transcriptional regulation in these processes.
    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
    神经干/祖细胞生活在复杂的细胞环境中,神经源性生态位,支持它们的功能并实现神经发生。生态位由多种细胞类型组成,包括神经元,神经胶质和脉管系统,它们能够向神经干/祖细胞发出信号并在结构上围绕神经干/祖细胞组织。虽然重点是单个细胞类型如何向神经干/祖细胞发出信号并影响其行为,实际上,人们对利基在多个细胞起源的发育过程中是如何组装的知之甚少,以及生成的拓扑在这些单元上的作用。这篇综述提出了这一新兴研究领域的最新图景,目的是从不同的动物模型(小鼠,斑马鱼和果蝇)。我们将跨越它的多个方面,从地方的存在和重要性,通过仔细组装各种细胞和无细胞成分,粘附相互作用可能会出现更大规模的拓扑特性。
    Neural stem/progenitor cells live in an intricate cellular environment, the neurogenic niche, which supports their function and enables neurogenesis. The niche is made of a diversity of cell types, including neurons, glia and the vasculature, which are able to signal to and are structurally organised around neural stem/progenitor cells. While the focus has been on how individual cell types signal to and influence the behaviour of neural stem/progenitor cells, very little is actually known on how the niche is assembled during development from multiple cellular origins, and on the role of the resulting topology on these cells. This review proposes to draw a state-of-the art picture of this emerging field of research, with the aim to expose our knowledge on niche architecture and formation from different animal models (mouse, zebrafish and fruit fly). We will span its multiple aspects, from the existence and importance of local, adhesive interactions to the potential emergence of larger-scale topological properties through the careful assembly of diverse cellular and acellular components.
    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
    双相情感障碍(BD)是一种慢性精神疾病,其特征是躁狂症和抑郁症的反复发作,并与社交和认知障碍有关。环境因素,比如母亲吸烟和童年创伤,被认为可以调节风险基因型,并有助于BD的发病机理,提示在神经发育过程中的表观遗传调控中的关键作用。5-羟甲基胞嘧啶(5hmC)是一种特别感兴趣的表观遗传变异体,因为它在大脑中高度表达,并与神经发育有关,以及精神和神经系统疾病。
    从两名患有双相情感障碍的青少年患者及其同性别年龄匹配的未受影响的兄弟姐妹(n=4)的白细胞中产生了诱导多能干细胞(iPSC)。Further,将iPSC分化成神经元干细胞(NSC)并使用免疫荧光表征纯度。我们使用减少代表性羟甲基化分析(RRHP)来进行iPSC和NSC的全基因组5hmC分析,模拟神经元分化过程中5hmC的变化,并评估其对BD风险的影响。用在线工具DAVID进行具有分化的5hmC基因座的基因的功能注释和富集测试。
    大约200万个站点被绘制和量化,大多数(68.8%)位于基因区域,每个站点观察到3个UTR的5hmC水平升高,外显子,和CpG群岛的2kb海岸线。iPSC和NSC细胞系之间的归一化5hmC计数的配对t检验显示,在NSC中整体低羟甲基化,以及与质膜相关的基因中差异羟甲基化位点的富集(FDR=9.1×10-12)和轴突导向(FDR=2.1×10-6)。在其他神经元过程中。对于KCNK9基因的转录因子结合位点观察到最显著的差异(p=8.8×10-6),编码参与神经元活动和迁移的钾通道蛋白。蛋白质-蛋白质相互作用(PPI)网络显示,由具有高度分化的5hmC位点的基因编码的蛋白质之间存在显着的连通性(p=3.2×10-10)。与轴突导向和离子跨膜转运相关的基因形成不同的亚簇。BD病例和未受影响的兄弟姐妹的NSC比较揭示了羟甲基化水平的其他分化模式,包括与突触形成和调节相关的基因中的位点,如CUX2(p=2.4×10-5)和DOK-7(p=3.6×10-3),以及与细胞外基质有关的基因的富集(FDR=1.0×10-8)。
    一起,这些初步结果为5hmC在早期神经元分化和BD风险中的潜在作用提供了证据,通过后续研究可以实现验证和更全面的表征。
    UNASSIGNED: Bipolar disorder (BD) is a chronic mental illness characterized by recurrent episodes of mania and depression and associated with social and cognitive disturbances. Environmental factors, such as maternal smoking and childhood trauma, are believed to modulate risk genotypes and contribute to the pathogenesis of BD, suggesting a key role in epigenetic regulation during neurodevelopment. 5-hydroxymethylcytosine (5hmC) is an epigenetic variant of particular interest, as it is highly expressed in the brain and is implicated in neurodevelopment, and psychiatric and neurological disorders.
    UNASSIGNED: Induced pluripotent stem cells (iPSCs) were generated from the white blood cells of two adolescent patients with bipolar disorder and their same-sex age-matched unaffected siblings (n = 4). Further, iPSCs were differentiated into neuronal stem cells (NSCs) and characterized for purity using immuno-fluorescence. We used reduced representation hydroxymethylation profiling (RRHP) to perform genome-wide 5hmC profiling of iPSCs and NSCs, to model 5hmC changes during neuronal differentiation and assess their impact on BD risk. Functional annotation and enrichment testing of genes harboring differentiated 5hmC loci were performed with the online tool DAVID.
    UNASSIGNED: Approximately 2 million sites were mapped and quantified, with the majority (68.8%) located in genic regions, with elevated 5hmC levels per site observed for 3\' UTRs, exons, and 2-kb shorelines of CpG islands. Paired t-tests of normalized 5hmC counts between iPSC and NSC cell lines revealed global hypo-hydroxymethylation in NSCs and enrichment of differentially hydroxymethylated sites within genes associated with plasma membrane (FDR = 9.1 × 10-12) and axon guidance (FDR = 2.1 × 10-6), among other neuronal processes. The most significant difference was observed for a transcription factor binding site for the KCNK9 gene (p = 8.8 × 10-6), encoding a potassium channel protein involved in neuronal activity and migration. Protein-protein-interaction (PPI) networking showed significant connectivity (p = 3.2 × 10-10) between proteins encoded by genes harboring highly differentiated 5hmC sites, with genes involved in axon guidance and ion transmembrane transport forming distinct sub-clusters. Comparison of NSCs of BD cases and unaffected siblings revealed additional patterns of differentiation in hydroxymethylation levels, including sites in genes with functions related to synapse formation and regulation, such as CUX2 (p = 2.4 × 10-5) and DOK-7 (p = 3.6 × 10-3), as well as an enrichment of genes involved in the extracellular matrix (FDR = 1.0 × 10-8).
    UNASSIGNED: Together, these preliminary results lend evidence toward a potential role for 5hmC in both early neuronal differentiation and BD risk, with validation and more comprehensive characterization to be achieved through follow-up study.
    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
    成人神经发生是成人大脑某些区域的神经干细胞(NSC)分化为神经元和神经胶质细胞的过程。神经发生缺陷会导致神经退行性疾病,精神障碍,和其他疾病。这个过程是由转录因子定向调控的,Wnt和Notch通路,细胞外基质,和各种生长因子。压力等外部因素,体育锻炼,饮食,药物,等。,影响神经发生和肠道微生物群。肠道微生物群可能通过迷走神经影响神经干细胞,免疫和化学途径,和其他途径。中药已被证明可以影响神经干细胞的增殖和分化,并可以调节肠道微生物的丰度和代谢产物。然而,这些因子通过肠道微生物群调节神经发生的潜在机制尚不完全清楚.在这次审查中,我们描述了最近关于肠道菌群在神经发生中的作用的证据.此外,我们假设基于细菌门的微生物群-肠-脑轴的特征,包括微生物群的代谢物,以及神经元和免疫途径,同时对中药通过调节肠道菌群对成人神经发生的潜在影响提供了展望。
    Adult neurogenesis is the process of differentiation of neural stem cells (NSCs) into neurons and glial cells in certain areas of the adult brain. Defects in neurogenesis can lead to neurodegenerative diseases, mental disorders, and other maladies. This process is directionally regulated by transcription factors, the Wnt and Notch pathway, the extracellular matrix, and various growth factors. External factors like stress, physical exercise, diet, medications, etc., affect neurogenesis and the gut microbiota. The gut microbiota may affect NSCs through vagal, immune and chemical pathways, and other pathways. Traditional Chinese medicine (TCM) has been proven to affect NSCs proliferation and differentiation and can regulate the abundance and metabolites produced by intestinal microorganisms. However, the underlying mechanisms by which these factors regulate neurogenesis through the gut microbiota are not fully understood. In this review, we describe the recent evidence on the role of the gut microbiota in neurogenesis. Moreover, we hypothesize on the characteristics of the microbiota-gut-brain axis based on bacterial phyla, including microbiota\'s metabolites, and neuronal and immune pathways while providing an outlook on TCM\'s potential effects on adult neurogenesis by regulating gut microbiota.
    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
    在阿尔茨海默病(AD)期间,由于生产与生产的不平衡,细胞外基质中淀粉样β肽沉积物的增加是具有里程碑意义的间隙。淀粉样β沉积物的这种积累触发了小胶质细胞的活化。小胶质细胞在AD中发挥双重作用,通过清除淀粉样β肽的沉积增加吞噬反应(CD163,IGF-1或BDNF)和细胞毒性作用的保护作用,释放自由基(ROS或NO)和促炎细胞因子(TNF-α,IL-1β)响应于淀粉样β聚集体激活的反应性神经胶质增生。小胶质细胞激活与KV1.3通道表达增加相关,蛋白质水平和电流密度。一些研究强调了KV1.3在激活炎症反应和抑制神经祖细胞增殖和神经元分化中的重要性。然而,关于这种激活在神经干细胞分化和增殖中的途径以及在淀粉样蛋白β积累中的作用知之甚少。在最近的研究中,使用来自小鼠模型的体外细胞,已经证明,KV1.3阻断剂抑制培养物中小胶质细胞介导的神经毒性,通过NF-kB和p38MAPK途径减少促炎细胞因子IL-1β和TNF-α的表达和产生。总的来说,我们得出结论,KV1.3阻断剂改变了AD的发展过程,减少小胶质细胞毒性激活和增加神经干细胞分化。然而,需要进一步的研究来确定具体的途径,并验证这种阻断剂作为治疗阿尔茨海默症患者的治疗方法.
    Increase of deposits of amyloid β peptides in the extracellular matrix is landmark during Alzheimer\'s Disease (AD) due to the imbalance in the production vs. clearance. This accumulation of amyloid β deposits triggers microglial activation. Microglia plays a dual role in AD, a protective role by clearing the deposits of amyloid β peptides increasing the phagocytic response (CD163, IGF-1 or BDNF) and a cytotoxic role, releasing free radicals (ROS or NO) and proinflammatory cytokines (TNF-α, IL-1β) in response to reactive gliosis activated by the amyloid β aggregates. Microglia activation correlated with an increase KV1.3 channels expression, protein levels and current density. Several studies highlight the importance of KV1.3 in the activation of inflammatory response and inhibition of neural progenitor cell proliferation and neuronal differentiation. However, little is known about the pathways of this activation in neural stem cells differentiation and proliferation and the role in amyloid β accumulation. In recent studies using in vitro cells derived from mice models, it has been demonstrated that KV1.3 blockers inhibit microglia-mediated neurotoxicity in culture reducing the expression and production of the pro-inflammatory cytokines IL-1β and TNF-α through the NF-kB and p38MAPK pathway. Overall, we conclude that KV1.3 blockers change the course of AD development, reducing microglial cytotoxic activation and increasing neural stem cell differentiation. However, further investigations are needed to establish the specific pathway and to validate the use of this blocker as therapeutic treatment in Alzheimer patients.
    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
    研究神经系统疾病的遗传病因可以为基因诊断和靶向治疗提供指导。在过去,种系突变,从父母传播或从父母生殖细胞新产生,被认为是神经系统疾病的主要遗传原因。然而,最近的证据表明,大脑中的体细胞突变,它可能是在发育过程中或过度衰老的神经干细胞产生的,解释了大量的大脑疾病,从神经发育,神经退行性疾病,和神经精神肿瘤疾病。此外,对致病体细胞突变或突变基因的鉴定为分子发病机制提供了新的见解,并揭示了治疗几乎没有神经系统疾病的潜在治疗靶点,或者没有,治疗选择。RNA疗法,包括反义寡核苷酸(ASO)和小干扰RNA(siRNA),正在成为治疗遗传性神经系统疾病的有希望的治疗工具。随着与种系突变相关的神经系统疾病的批准和研究性ASO和siRNA药物数量的增加,它们也可能被证明是治疗由体细胞突变引起的神经系统疾病的有吸引力的方法.从这个角度来看,我们重点介绍了几种由脑细胞突变引起的神经系统疾病,并讨论了RNA治疗在这些疾病中的潜在作用。
    Research into the genetic etiology of a neurological disorder can provide directions for genetic diagnosis and targeted therapy. In the past, germline mutations, which are transmitted from parents or newly arise from parental germ cells, were considered as major genetic causes of neurological disorders. However, recent evidence has shown that somatic mutations in the brain, which can arise from neural stem cells during development or over aging, account for a significant number of brain disorders, ranging from neurodevelopmental, neurodegenerative, and neuropsychiatric to neoplastic disease. Moreover, the identification of disease-causing somatic mutations or mutated genes has provided new insights into molecular pathogenesis and unveiled potential therapeutic targets for treating neurological disorders that have few, or no, therapeutic options. RNA therapeutics, including antisense oligonucleotide (ASO) and small interfering RNA (siRNA), are emerging as promising therapeutic tools for treating genetic neurological disorders. As the number of approved and investigational ASO and siRNA drugs for neurological disorders associated with germline mutations increases, they may also prove to be attractive modalities for treating neurologic disorders resulting from somatic mutations. In this perspective, we highlight several neurological diseases caused by brain somatic mutations and discuss the potential role of RNA therapeutics in these conditions.
    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
    未经证实:缺氧缺血性脑损伤(HIBD)会显著影响婴儿的神经发育,并且是新生儿严重神经系统发病率和死亡率的主要原因。我们先前的研究发现,光生物调节疗法(PBMT)可以改善HIBD大鼠模型的空间学习和记忆能力。然而,PBMT在HIBD中的神经保护机制尚不清楚。
    未经批准:在本研究中,HIBD模型大鼠在黑暗中每天用5mW/cm2的PBMT治疗14天(每天10分钟),氧糖剥夺(OGD)后的原代神经干细胞(NSC)用PBMT在黑暗中以1、5、10和20mW/cm2处理10分钟。PBMT在体内和体外促进海马神经干细胞(NSC)的增殖。
    未经批准:机械,PBMT上调磷脂酰肌醇3激酶(PI3K),磷酸化蛋白激酶B(p-AKT),磷酸化糖原合成酶激酶3β(p-GSK-3β),β-连环蛋白,和细胞周期蛋白D1在体内和体外的表达,促进NSC扩散。此外,LY294002(PI3K抑制剂)和IWR-1(Wnt/β-catenin抑制剂)均抑制OGD后PBMT促进NSC增殖,并在体外抑制β-catenin和cyclinD1表达。
    未经证实:PBMT通过AKT/GSK-3β/β-catenin通路改善HIBD大鼠的空间学习记忆能力,促进海马NSC增殖。
    UNASSIGNED: Hypoxic-ischemic brain damage (HIBD) significantly affects neurodevelopment in infants and is a leading cause of severe neurological morbidity and mortality in neonates. Our previous study found that photobiomodulation therapy (PBMT) improves the impaired spatial learning and memory of HIBD rat models. However, the neuroprotective mechanism conferred by PBMT in HIBD is unclear.
    UNASSIGNED: In the present study, HIBD model rats were treated with PBMT at 5 mW/cm2 per day in the dark for 14 days (10 min each day), and primary neural stem cells (NSCs) after oxygen-glucose deprivation (OGD) were treated with PBMT for 10 min at 1, 5, 10, and 20 mW/cm2 in the dark. PBMT promoted hippocampal neural stem cell (NSC) proliferation in vivo and in vitro.
    UNASSIGNED: Mechanistically, PBMT upregulated phosphatidylinositol 3 kinase (PI3K), phosphorylated protein kinase B (p-AKT), phosphorylated glycogen synthase kinase 3 beta (p-GSK-3β), β-catenin, and cyclin D1 expression in vivo and in vitro, promoting NSC proliferation. Furthermore, both LY294002 (a PI3K inhibitor) and IWR-1 (a Wnt/β-catenin inhibitor) inhibited the PBMT promotion of NSC proliferation after OGD and suppressed β-catenin and cyclin D1 expression in vitro.
    UNASSIGNED: PBMT improved the spatial learning and memory of HIBD rats and promoted hippocampal NSC proliferation through the AKT/GSK-3β/β-catenin pathway.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号