neuronal differentiation

神经元分化
  • 文章类型: Journal Article
    神经发育障碍包括一系列疾病,如智力障碍,自闭症谱系障碍,罕见的遗传病和发育性和癫痫性脑病,都表现在童年。超过1,500个基因参与各种信号通路,包括许多转录调节因子,剪接体元素,已经认识到染色质修饰复合物和从头变体在这些疾病中的重要作用。随着新的机器学习工具应用于神经成像,这些发现有助于基因诊断,提供对神经病理学机制的关键见解并帮助预后,精准医学。此外,新发现强调了理解蛋白质编码基因之外的遗传贡献的重要性,并强调了RNA和非编码DNA分子以及新参与者的作用,如转座因子,其失调会导致基因功能破坏,表观遗传改变,和基因组不稳定性。最后,分析神经影像学的最新进展现在提供了在体内表征神经元细胞结构的可能性,为传统的验尸研究提供了一个可行的替代方案。随着最近推出的人类胎儿大脑发育数字地图集,这些新方法将允许回答复杂的生物学问题,有关胎儿在儿童时期认知功能的起源。在这次审查中,我们提出了十大引人入胜的主题,这些主题在去年取得了重大进展。
    Neurodevelopmental disorders encompass a range of conditions such as intellectual disability, autism spectrum disorder, rare genetic disorders and developmental and epileptic encephalopathies, all manifesting during childhood. Over 1,500 genes involved in various signaling pathways, including numerous transcriptional regulators, spliceosome elements, chromatin-modifying complexes and de novo variants have been recognized for their substantial role in these disorders. Along with new machine learning tools applied to neuroimaging, these discoveries facilitate genetic diagnoses, providing critical insights into neuropathological mechanisms and aiding in prognosis, and precision medicine. Also, new findings underscore the importance of understanding genetic contributions beyond protein-coding genes and emphasize the role of RNA and non-coding DNA molecules but also new players, such as transposable elements, whose dysregulation generates gene function disruption, epigenetic alteration, and genomic instability. Finally, recent developments in analyzing neuroimaging now offer the possibility of characterizing neuronal cytoarchitecture in vivo, presenting a viable alternative to traditional post-mortem studies. With a recently launched digital atlas of human fetal brain development, these new approaches will allow answering complex biological questions about fetal origins of cognitive function in childhood. In this review, we present ten fascinating topics where major progress has been made in the last year.
    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
    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: Journal Article
    联合治疗是开发治疗复杂疾病的有前途的方法之一。如阿尔茨海默病(AD)。在泰国传统药物中,临床应用通常包括多种植物性药物作为制剂。植物药物在联合治疗中的协同相互作用被认为具有几个优点。包括提高治疗效果,减少毒性和/或不良反应。这项研究旨在探索传统多草药配方中两种植物药物的植物混合制剂(BHP)的治疗功能。以1:9w/w的特定比例说明了龙血树(DCS)和椭圆形紫菜(AEF)的BHP在神经保护和抗炎方面的协同作用。在培养的PC12细胞中,DCS和AEF的BHP在诱导神经元分化方面表现出协同作用,以神经丝表达和神经突生长为特征。此外,DCS和AEF的BHP在抑制Aβ聚集方面表现出协同作用,AD病理学的标志。LPS诱导的活化的BV2小胶质细胞被DCS和AEF的BHP协同抑制,通过促炎标志物的表达评估,包括TNF-α,IL-1β,和iNOS,以及小胶质细胞的形态变化。研究结果表明,DCS和AEF的BHP在1:9w/w的特定比例下比单个植物药物的作用更大,以增强神经保护和抗炎功能。
    Combination therapy is one of the promising approaches in developing therapeutics to cure complex diseases, such as Alzheimer\'s disease (AD). In Thai traditional medicines, the clinical application often comprises multiple botanical drugs as a formulation. The synergistic interactions between botanical drugs in combination therapies are proposed to have several advantages, including increased therapeutic efficacy, and decreased toxicity and/or adverse effects. This study aimed to explore the therapeutic functions of a botanical hybrid preparation (BHP) of two botanical drugs within a traditional multi-herbal formulation. The synergistic actions of BHP of Dracaena cochinchinensis stemwood (DCS) and Ardisia elliptica fruit (AEF) at a specific ratio of 1:9 w/w were illustrated in neuroprotection and anti-inflammation. In cultured PC12 cells, BHP of DCS and AEF showed synergistic functions in inducing neuronal differentiation, characterized by neurofilament expression and neurite outgrowth. In addition, BHP of DCS and AEF exhibited a synergistic effect in inhibiting the aggregation of Aβ, a hallmark of AD pathology. The activated BV2 microglial cells induced by LPS were synergistically suppressed by the BHP of DCS and AEF, as evaluated by the expression of pro-inflammatory markers, including TNF-α, IL-1β, and iNOS, as well as the morphological change of microglial cells. The findings suggested that the effects of BHP of DCS and AEF were greater than individual botanical drugs in a specific ratio of 1:9 w/w to enhance neuroprotective and anti-inflammatory functions.
    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
    神经系统疾病,从中风和创伤性脑损伤等急性形式到痴呆等神经退行性疾病,是全球残疾调整寿命年(DALYs)的主要原因。解决这些情况和促进神经系统再生的有希望的方法是使用神经肽制剂脑活素,已在临床和临床前研究中被证明是有效的。尽管几种肽制剂声称具有相似的临床疗效和安全性,之前已经提出了对其通用成分和功效的担忧。根据这些报告,我们分析了几种据称类似于脑活素的肽制剂的肽组成和神经营养活性,并在一些国家批准用于治疗神经系统疾病。我们的结果表明,这些制剂缺乏相关的生物活性,并且肽组成与脑活素明显不同。肽。
    Neurological disorders, ranging from acute forms such as stroke and traumatic brain injury to neurodegenerative diseases like dementia, are the leading cause of disability-adjusted life years (DALYs) worldwide. A promising approach to address these conditions and promote nervous system regeneration is the use of the neuropeptide preparation Cerebrolysin, which has been shown to be effective in both clinical and preclinical studies. Despite claims of similar clinical efficacy and safety by several peptide preparations, concerns regarding their generic composition and efficacy have been previously raised. Based on these reports, we analyzed the peptide composition and neurotrophic activity of several peptide preparations allegedly similar to Cerebrolysin and approved in some countries for treating neurological diseases. Our results demonstrate that these preparations lack relevant biological activity and that the peptide composition is significantly different from Cerebrolysin. peptide.
    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
    脂肪间充质干细胞(ASCs)是成人多能干细胞,能够向中胚层谱系细胞以外的神经元分化。这项研究的目的是使用褪黑激素结合胶质细胞的条件培养基(CM)测试ASC神经分化。从健康捐赠者的脂肪抽吸物中分离出来,在经历神经分化程序之前,将ASC在基础生长培养基中扩增。为此,使用获自嗅鞘细胞和施旺细胞的CM。在一些样本中,加入1μM的褪黑激素。培养1天和7天后,使用免疫细胞化学和流式细胞术研究细胞以评估神经标记物的表达(Nestin,MAP2,突触素I,GFAP)在不同条件下。结果证实,成功的神经分化是通过神经胶质CM实现的,而单独添加褪黑激素并没有引起明显的变化。当褪黑激素与CM结合使用时,ASC神经分化增强,正如神经元标记表达的进一步改善所证明的那样,而神经胶质分化减弱。还观察到动态调制,测试褪黑素受体的表达。总之,我们的数据表明,褪黑素的神经分化能力可被有效利用,以获得神经元样分化的ASCs,用于潜在的治疗策略.
    Adipose-derived mesenchymal stem cells (ASCs) are adult multipotent stem cells, able to differentiate toward neural elements other than cells of mesodermal lineage. The aim of this research was to test ASC neural differentiation using melatonin combined with conditioned media (CM) from glial cells. Isolated from the lipoaspirate of healthy donors, ASCs were expanded in a basal growth medium before undergoing neural differentiation procedures. For this purpose, CM obtained from olfactory ensheathing cells and from Schwann cells were used. In some samples, 1 µM of melatonin was added. After 1 and 7 days of culture, cells were studied using immunocytochemistry and flow cytometry to evaluate neural marker expression (Nestin, MAP2, Synapsin I, GFAP) under different conditions. The results confirmed that a successful neural differentiation was achieved by glial CM, whereas the addition of melatonin alone did not induce appreciable changes. When melatonin was combined with CM, ASC neural differentiation was enhanced, as demonstrated by a further improvement of neuronal marker expression, whereas glial differentiation was attenuated. A dynamic modulation was also observed, testing the expression of melatonin receptors. In conclusion, our data suggest that melatonin\'s neurogenic differentiation ability can be usefully exploited to obtain neuronal-like differentiated ASCs for potential therapeutic strategies.
    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
    胚胎干细胞(ESCs)发育为神经元需要复杂的转录调控,拼接,翻译,但是这些过程如何互连还不清楚。我们发现多嘧啶束结合蛋白1(PTBP1)控制DPF2的剪接,DPF2是BRG1/BRM相关因子(BAF)染色质重塑复合物的亚基。Dpf2外显子7剪接被PTBP1抑制以在发育早期产生DPF2-S同工型。在神经元分化过程中,PTBP1的缺失允许外显子7包含和DPF2-L表达。通过这些替代的DPF2同种型诱导不同的细胞表型和基因表达程序。我们确定了每个DPF2亚型富集的染色质结合位点,以及两者绑定的网站。在ESC,DPF2-S优先位点被多能性因子结合。在神经元祖细胞中,DPF2-S位点与核因子I(NFI)结合,而DPF2-L位点被CCCTC结合因子(CTCF)结合。DPF2-S位点表现出增强子修饰,而DPF2-L位点显示启动子修饰。因此,选择性剪接重定向BAF复合物靶向以影响神经元发育过程中的染色质组织。
    Development of embryonic stem cells (ESCs) into neurons requires intricate regulation of transcription, splicing, and translation, but how these processes interconnect is not understood. We found that polypyrimidine tract binding protein 1 (PTBP1) controls splicing of DPF2, a subunit of BRG1/BRM-associated factor (BAF) chromatin remodeling complexes. Dpf2 exon 7 splicing is inhibited by PTBP1 to produce the DPF2-S isoform early in development. During neuronal differentiation, loss of PTBP1 allows exon 7 inclusion and DPF2-L expression. Different cellular phenotypes and gene expression programs were induced by these alternative DPF2 isoforms. We identified chromatin binding sites enriched for each DPF2 isoform, as well as sites bound by both. In ESC, DPF2-S preferential sites were bound by pluripotency factors. In neuronal progenitors, DPF2-S sites were bound by nuclear factor I (NFI), while DPF2-L sites were bound by CCCTC-binding factor (CTCF). DPF2-S sites exhibited enhancer modifications, while DPF2-L sites showed promoter modifications. Thus, alternative splicing redirects BAF complex targeting to impact chromatin organization during neuronal development.
    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
    缺血性卒中是一种主要的脑血管疾病,具有很高的发病率和死亡率,缺血性卒中相关神经功能障碍的有效治疗方法尚待开发.在这项研究中,我们从人类白细胞抗原主要位点基因纯合诱导的多能干细胞(hiPSC-NPCs)产生神经祖细胞,并评估其对缺血性卒中的治疗效果.在亚急性或急性期,将hiPSC-NPCs脑内移植到短暂大脑中动脉闭塞产生的大鼠缺血脑中,以及它们在体内的存活,分化,并评估神经功能障碍功能改善的疗效。hiPSC-NPCs在宿主脑组织中进行组织学鉴定,并显示神经元分化为vGLUT阳性谷氨酸能神经元,神经突延伸到同侧梗死和对侧健康半球,急性和亚急性期移植后12周形成突触结构。当在亚急性阶段用γ-分泌酶抑制剂预处理移植时,它们还改善了神经功能。然而,它们的影响是适度的,并不显著,表明在急性期移植中细胞可能存在保持未分化和未成熟状态的风险.这些结果表明,hiPSC-NPC在缺血性卒中受损的神经组织中显示出细胞替代作用,但其疗效不足以改善急性或亚急性移植后的神经功能。需要进一步优化细胞制备方法和移植时机,以平衡hiPSC-NPC移植的功效和安全性。
    Ischemic stroke is a major cerebrovascular disease with high morbidity and mortality rates; however, effective treatments for ischemic stroke-related neurological dysfunction have yet to be developed. In this study, we generated neural progenitor cells from human leukocyte antigen major loci gene-homozygous-induced pluripotent stem cells (hiPSC-NPCs) and evaluated their therapeutic effects against ischemic stroke. hiPSC-NPCs were intracerebrally transplanted into rat ischemic brains produced by transient middle cerebral artery occlusion at either the subacute or acute stage, and their in vivo survival, differentiation, and efficacy for functional improvement in neurological dysfunction were evaluated. hiPSC-NPCs were histologically identified in host brain tissues and showed neuronal differentiation into vGLUT-positive glutamatergic neurons, extended neurites into both the ipsilateral infarct and contralateral healthy hemispheres, and synaptic structures formed 12 weeks after both acute and subacute stage transplantation. They also improved neurological function when transplanted at the subacute stage with γ-secretase inhibitor pretreatment. However, their effects were modest and not significant and showed a possible risk of cells remaining in their undifferentiated and immature status in acute-stage transplantation. These results suggest that hiPSC-NPCs show cell replacement effects in ischemic stroke-damaged neural tissues, but their efficacy is insufficient for neurological functional improvement after acute or subacute transplantation. Further optimization of cell preparation methods and the timing of transplantation is required to balance the efficacy and safety of hiPSC-NPC transplantation.
    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
    选择性RNA剪接是神经元分化和突触成熟中必不可少的动态过程,这个过程的失调与神经退行性疾病有关。最近的研究揭示了RNA结合蛋白在神经元剪接程序调节中的重要性。然而,这些剪接调节因子调控的分子机制尚不清楚.这里我们展示了KIS,一种在发育大脑中上调的激酶,在小鼠的神经元分化过程中,外显子的使用会引起全基因组的改变。KIS包含剪接体组分和磷酸化PTBP2共有的蛋白质识别结构域,抵消了该剪接因子在外显子排斥中的作用。在分子水平上,PTBP2内非结构化结构域的磷酸化导致其与两个共调节因子解离,Matrin3和hnRNPM,并阻碍复合物的RNA结合能力。此外,KIS和PTBP2在突触脊柱出现和成熟中显示出强烈且相反的功能相互作用。一起来看,我们的数据揭示了剪接调节因子的翻译后控制,这些调节因子将神经元发育中的转录和替代外显子使用程序联系起来。
    Alternative RNA splicing is an essential and dynamic process in neuronal differentiation and synapse maturation, and dysregulation of this process has been associated with neurodegenerative diseases. Recent studies have revealed the importance of RNA-binding proteins in the regulation of neuronal splicing programs. However, the molecular mechanisms involved in the control of these splicing regulators are still unclear. Here, we show that KIS, a kinase upregulated in the developmental brain, imposes a genome-wide alteration in exon usage during neuronal differentiation in mice. KIS contains a protein-recognition domain common to spliceosomal components and phosphorylates PTBP2, counteracting the role of this splicing factor in exon exclusion. At the molecular level, phosphorylation of unstructured domains within PTBP2 causes its dissociation from two co-regulators, Matrin3 and hnRNPM, and hinders the RNA-binding capability of the complex. Furthermore, KIS and PTBP2 display strong and opposing functional interactions in synaptic spine emergence and maturation. Taken together, our data uncover a post-translational control of splicing regulators that link transcriptional and alternative exon usage programs in neuronal development.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号