neural stem cells

神经干细胞
  • 文章类型: Journal Article
    本研究旨在评估和比较人沃顿果冻(WJ)间充质干细胞(MSCs)和神经前体(NPs)在实验性自身免疫性脑脊髓炎(EAE)中的细胞治疗,多发性硬化症的临床前模型。通过外植体技术从WJ中分离出MSCs,区分为NP,并在伦理批准后通过细胞计数和免疫细胞化学分析进行表征。48只大鼠由髓磷脂碱性蛋白和弗氏完全佐剂诱导EAE。48小时后,这些动物接受腹膜内注射250ng/剂百日咳博德特氏菌毒素.十四天后,将动物分为以下组:a.非诱导,诱导:b.Sham,c.WJ-MSC,d.NP,和e.WJ-MSC加NP。1×105此外,将细胞置于10µL溶液中,并通过立体定位脑室注射进行注射.十天后,H&E的组织病理学分析,Luxol,白细胞介素,进行CD4/CD8。统计分析表明,与其他组相比,Sham组的临床表现频率更高(15.66%);与Sham组相比,治疗组的脱髓鞘减少(WJ-MSCs,p=0.016;NPs,p=0.010;WJ-MSC+NP,p=0.000),与Sham组相比,治疗组的细胞死亡率较低。CD4/CD8比值<1显示与小胶质细胞活化无关(p=0.366),星形胶质细胞(p=0.247),和WJ-MSC中的细胞死亡(p=0.577)。WJ-MSCs和NPs在细胞治疗中具有免疫调节和神经保护作用,这将被翻译为脱髓鞘疾病的辅助手段。
    This study aims to evaluate and compare cellular therapy with human Wharton\'s jelly (WJ) mesenchymal stem cells (MSCs) and neural precursors (NPs) in experimental autoimmune encephalomyelitis (EAE), a preclinical model of Multiple Sclerosis. MSCs were isolated from WJ by an explant technique, differentiated to NPs, and characterized by cytometry and immunocytochemistry analysis after ethical approval. Forty-eight rats were EAE-induced by myelin basic protein and Freund\'s complete adjuvant. Forty-eight hours later, the animals received intraperitoneal injections of 250 ng/dose of Bordetella pertussis toxin. Fourteen days later, the animals were divided into the following groups: a. non-induced, induced: b. Sham, c. WJ-MSCs, d. NPs, and e. WJ-MSCs plus NPs. 1 × 105. Moreover, the cells were placed in a 10 µL solution and injected via a stereotaxic intracerebral ventricular injection. After ten days, the histopathological analysis for H&E, Luxol, interleukins, and CD4/CD8 was carried out. Statistical analyses demonstrated a higher frequency of clinical manifestation in the Sham group (15.66%) than in the other groups; less demyelination was seen in the treated groups than the Sham group (WJ-MSCs, p = 0.016; NPs, p = 0.010; WJ-MSCs + NPs, p = 0.000), and a lower cellular death rate was seen in the treated groups compared with the Sham group. A CD4/CD8 ratio of <1 showed no association with microglial activation (p = 0.366), astrocytes (p = 0.247), and cell death (p = 0.577) in WJ-MSCs. WJ-MSCs and NPs were immunomodulatory and neuroprotective in cellular therapy, which would be translated as an adjunct in demyelinating diseases.
    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
    感觉神经性听力损失(SNHL)是耳鼻喉科的常见病。一个关键的障碍是找到再生成年动物受损耳蜗毛细胞的有效策略。已开发出一种实用可靠的方法,为内耳干细胞移植治疗SNHL创造了优越的细胞源。Atoh1参与神经元的分化,肠道分泌细胞,和包括听觉毛细胞在内的机械感受器,因此在神经发生中起着重要作用。慢病毒介导的骨髓间充质干细胞(BMSCs)的转染被用来实现必需转录因子Atoh1的稳定表达,这对于发育听觉毛细胞而不损害细胞存活至关重要。通过使用抗贴壁培养改变细胞生长环境来操纵诱导条件,碱性成纤维细胞生长因子(bFGF)和表皮生长因子(EGF)的协同作用可显著提高Atoh1转染后骨髓间充质干细胞(BMSC)向神经干细胞(NSC)的分化效率,从而减少诱导时间。研究表明,新提出的转分化方法在受控环境中有效地将BMSCs转化为NSCs,提供干细胞移植促进毛细胞再生的潜在途径。
    Sensorineural hearing loss (SNHL) is a prevalent condition in otolaryngology. A key obstacle is finding effective strategies for regenerating damaged cochlear hair cells in adult animals. A practical and reliable approach has been developed to create a superior cell source for stem cell transplantation in the inner ear to treat SNHL. Atoh1 is involved in the differentiation of neurons, intestinal secretory cells, and mechanoreceptors including auditory hair cells, and thus plays an important role in neurogenesis. Lentivirus-mediated transfection of bone marrow mesenchymal stem cells (BMSCs) was utilized to achieve stable expression of the essential transcription factor Atoh1, which is crucial for developing auditory hair cells without compromising cell survival. By manipulating the induction conditions through altering the cell growth environment using anti-adherent culture, the synergistic impact of basic fibroblast growth factor (bFGF) and epidermal growth factor (EGF) was effectively applied to significantly improve the differentiation efficiency of bone marrow-derived mesenchymal stem cells (BMSC) into neural stem cells (NSCs) following Atoh1 transfection, thereby reducing the induction time. The study indicated that the newly proposed transdifferentiation method effectively transformed BMSCs into NSCs in a controlled environment, presenting a potential approach for stem cell transplantation to promote hair cell regeneration.
    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
    为了促进神经组织再生,生物材料支架已经成为有希望的候选者,为神经中断提供潜在的解决方案。在这些脚手架中,多通道水凝胶,其特点是精心设计的微米尺度通道,作为引导轴突生长和促进细胞相互作用的工具。本研究探讨了用甲基丙烯酰结构域(AMMA)修饰的人羊膜在神经干细胞(NSC)培养中的创新应用。AMMA水凝胶,具有类似于生理环境的定制柔软度,以多通道支架的形式制备,以模拟神经束的天然样微结构。AMMA水凝胶膜的初步实验展示了它们在神经应用中的潜力,表现出强大的附着力,扩散,和NSC的分化,而不需要额外的涂层。过渡到3D领域,多通道结构促进了复杂的神经元网络,引导神经突纵向延伸。此外,细胞阵列中突触小泡的存在表明功能性突触连接的建立,强调了发达神经元网络的生理相关性。这项工作有助于不断努力寻找道德,临床可翻译,以及再生神经科学的功能相关方法。
    In the pursuit of advancing neural tissue regeneration, biomaterial scaffolds have emerged as promising candidates, offering potential solutions for nerve disruptions. Among these scaffolds, multichannel hydrogels, characterized by meticulously designed micrometer-scale channels, stand out as instrumental tools for guiding axonal growth and facilitating cellular interactions. This study explores the innovative application of human amniotic membranes modified with methacryloyl domains (AMMA) in neural stem cell (NSC) culture. AMMA hydrogels, possessing a tailored softness resembling the physiological environment, are prepared in the format of multichannel scaffolds to simulate native-like microarchitecture of nerve tracts. Preliminary experiments on AMMA hydrogel films showcase their potential for neural applications, demonstrating robust adhesion, proliferation, and differentiation of NSCs without the need for additional coatings. Transitioning into the 3D realm, the multichannel architecture fosters intricate neuronal networks guiding neurite extension longitudinally. Furthermore, the presence of synaptic vesicles within the cellular arrays suggests the establishment of functional synaptic connections, underscoring the physiological relevance of the developed neuronal networks. This work contributes to the ongoing efforts to find ethical, clinically translatable, and functionally relevant approaches for regenerative neuroscience.
    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
    心室-室下区(V-SVZ)中的成年神经干细胞(aNSC)在很大程度上是静止的。这里,我们描述了细胞信号传导抑制蛋白的功能作用的潜在机制,LRIG1,在控制aNSC增殖中。使用Lrig1敲除模型,我们表明,Lrig1消融导致aNSCs增殖增加,而神经元子代无变化,而且这种过度增殖可能并非仅由表皮生长因子受体(EGFR)的激活引起.然而,LRIG1的损失,还导致转化生长因子β(TGFβ)和骨形态发生蛋白(BMP)信号传导的活化受损。生物化学,我们显示LRIG1结合TGFβ/BMP受体和TGFβ1配体。最后,我们表明,这些相互作用的后果是促进SMAD磷酸化。总的来说,这些数据表明,与胚胎神经干细胞不同,EGFR可能是主要的作用机制,在ASCs中,LRIG1和TGFβ途径共同发挥作用以实现其抑制作用。
    Adult Neural Stem Cells (aNSCs) in the ventricular-subventricular zone (V-SVZ) are largely quiescent. Here, we characterize the mechanism underlying the functional role of a cell-signalling inhibitory protein, LRIG1, in the control of aNSCs proliferation. Using Lrig1 knockout models, we show that Lrig1 ablation results in increased aNSCs proliferation with no change in neuronal progeny and that this hyperproliferation likely does not result solely from activation of the epidermal growth factor receptor (EGFR). Loss of LRIG1, however, also leads to impaired activation of transforming growth factor beta (TGFβ) and bone morphogenic protein (BMP) signalling. Biochemically, we show that LRIG1 binds TGFβ/BMP receptors and the TGFβ1 ligand. Finally, we show that the consequences of these interactions are to facilitate SMAD phosphorylation. Collectively, these data suggest that unlike in embryonic NSCs where EGFR may be the primary mechanism of action, in aNSCs, LRIG1 and TGFβ pathways function together to fulfill their inhibitory roles.
    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
    脑脊液接触神经元(CSF-cNs)代表一组独特的中间神经元,其特征是其突出的顶端球形突起穿透脊髓的中央管,其基底轴突向相邻细胞延伸。近一个世纪前确定,由于历史上缺乏明确的标记,CSF-cNs的特定作用和属性才刚刚开始出现.最近的发现已经证实,表达PKD2L1的CSF-cNs具有神经干细胞的属性,提示脊髓损伤后再生过程中的关键功能。本文旨在阐明CSF-cNs作为脊髓发育过程中潜在神经干细胞的分子标志物,并评估其在脊髓损伤后的作用。强调它们对脊髓修复的潜在治疗意义。
    Cerebrospinal fluid-contacting neurons (CSF-cNs) represent a distinct group of interneurons characterized by their prominent apical globular protrusions penetrating the spinal cord\'s central canal and their basal axons extending towards adjacent cells. Identified nearly a century back, the specific roles and attributes of CSF-cNs have just started to emerge due to the historical lack of definitive markers. Recent findings have confirmed that CSF-cNs expressing PKD2L1 possess attributes of neural stem cells, suggesting a critical function in the regeneration processes following spinal cord injuries. This review aims to elucidate the molecular markers of CSF-cNs as potential neural stem cells during spinal cord development and assess their roles post-spinal cord injury, with an emphasis on their potential therapeutic implications for spinal cord repair.
    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
    神经干细胞(NSC)具有显著的自我更新能力,并具有分化为各种神经谱系细胞的潜能,这使得它们在神经系统疾病的管理中至关重要。利用内源性NSC的内在潜力来增强神经修复和再生代表了解决神经系统疾病的最佳方法。在这项研究中,我们探索了一种新的二苯甲酮衍生物的潜力,命名为二吉西芬A(DGA),从内生真菌延胡索中分离出来。以前的实验已经广泛地识别和表征了DGA,揭示其独特的属性。我们的发现证明了DGA刺激神经干细胞增殖的显着能力,在体外和体内。此外,我们建立了放射性小脑损伤模型,以评估DGA对受损小脑内不同细胞亚群分布的影响,从而表明其在小脑修复中的有益作用。此外,我们对原代神经干细胞模型的观察表明,DGA显著增加细胞耗氧量,表明能量和代谢需求增加。通过利用各种途径抑制剂与DGA的组合,我们成功证明了其抵消AMPK和GSK3β抑制剂对NSC增殖的抑制作用的能力。总的来说,我们的研究结果强烈表明DGA,作为一种创新的化合物,通过AMPK/AKT/GSK3β通路的调节发挥其激活神经干细胞和促进损伤修复的作用。
    Neural stem cells (NSCs) exhibit a remarkable capacity for self-renewal and have the potential to differentiate into various neural lineage cells, which makes them pivotal in the management of neurological disorders. Harnessing the inherent potential of endogenous NSCs for enhancing nerve repair and regeneration represents an optimal approach to addressing diseases of the nervous system. In this study, we explored the potential of a novel benzophenone derivative named Digirseophene A (DGA), which was isolated from the endophytic fungus Corydalis tomentella. Previous experiments have extensively identified and characterized DGA, revealing its unique properties. Our findings demonstrate the remarkable capability of DGA to stimulate neural stem cell proliferation, both in vitro and in vivo. Furthermore, we established a model of radiation-induced cerebellar injury to assess the effects of DGA on the distribution of different cell subpopulations within the damaged cerebellum, thereby suggesting its beneficial role in cerebellar repair. In addition, our observations on a primary NSCs model revealed that DGA significantly increased cellular oxygen consumption, indicating increased energy and metabolic demands. By utilizing various pathway inhibitors in combination with DGA, we successfully demonstrated its ability to counteract the suppressive impacts of AMPK and GSK3β inhibitors on NSC proliferation. Collectively, our research results strongly suggest that DGA, as an innovative compound, exerts its role in activating NSCs and promoting injury repair through the regulation of the AMPK/AKT/GSK3β pathway.
    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
    脊髓损伤(SCI)是一种复杂的组织损伤,导致广泛的身体缺陷,包括永久性或进行性感觉障碍,运动和自主功能。迄今为止,当前临床治疗方案的局限性可能使SCI患者终生残疾.迫切需要开发新的疗法来重建受损的脊髓神经元-神经胶质网络并恢复与脊髓上通路的连接。神经干细胞(NSC)具有自我更新和分化为神经元和神经胶质细胞的能力,包括少突胶质细胞,它们是负责髓鞘形成和维持以及脱髓鞘轴突再生的细胞。对于这些属性,神经干细胞被认为是重建受损神经回路和促进髓鞘再生的有前途的细胞来源。在过去的十年里,已经在各种SCI临床前模型中广泛测试了神经干细胞的移植。本文旨在强调SCI的病理生理学,促进对NSCs在SCI修复治疗中的作用和病理机制的研究进展。临床前研究,以及通过NSC移植治疗策略进行SCI的临床试验。了解和掌握这些前沿更新将为建立新的治疗策略以提高SCI恢复质量铺平道路。
    Spinal cord injury (SCI) is a complex tissue injury that results in a wide range of physical deficits, including permanent or progressive disabilities of sensory, motor and autonomic functions. To date, limitations in current clinical treatment options can leave SCI patients with lifelong disabilities. There is an urgent need to develop new therapies for reconstructing the damaged spinal cord neuron-glia network and restoring connectivity with the supraspinal pathways. Neural stem cells (NSCs) possess the ability to self-renew and differentiate into neurons and neuroglia, including oligodendrocytes, which are cells responsible for the formation and maintenance of the myelin sheath and the regeneration of demyelinated axons. For these properties, NSCs are considered to be a promising cell source for rebuilding damaged neural circuits and promoting myelin regeneration. Over the past decade, transplantation of NSCs has been extensively tested in a variety of preclinical models of SCI. This review aims to highlight the pathophysiology of SCI and promote the understanding of the role of NSCs in SCI repair therapy and the current advances in pathological mechanism, pre-clinical studies, as well as clinical trials of SCI via NSC transplantation therapeutic strategy. Understanding and mastering these frontier updates will pave the way for establishing novel therapeutic strategies to improve the quality of recovery from SCI.
    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
    模仿内侧颞叶癫痫的海马癫痫导致小鼠成年神经源性小生境的严重破坏。癫痫发作引起神经干细胞转换为反应性表型(反应性神经干细胞,React-NSC)以多分支肥大形态为特征,大量激活进入有丝分裂,对称除法,并最终分化为反应性星形胶质细胞。因此,神经发生是慢性受损。这里,使用内侧颞叶癫痫的小鼠模型,我们发现表皮生长因子受体(EGFR)信号通路是诱导React-NSCs的关键,其抑制对神经源性生态位具有有益作用。我们表明,在通过单次海马内注射海人酸诱导癫痫发作后的最初几天,锌和肝素结合表皮生长因子的强烈释放,神经干细胞中EGFR信号通路的两种激活剂,是生产的。给予EGFR抑制剂吉非替尼,临床IV期的化疗药物,防止React-NSC的诱导并保留神经发生。
    Hippocampal seizures mimicking mesial temporal lobe epilepsy cause a profound disruption of the adult neurogenic niche in mice. Seizures provoke neural stem cells to switch to a reactive phenotype (reactive neural stem cells, React-NSCs) characterized by multibranched hypertrophic morphology, massive activation to enter mitosis, symmetric division, and final differentiation into reactive astrocytes. As a result, neurogenesis is chronically impaired. Here, using a mouse model of mesial temporal lobe epilepsy, we show that the epidermal growth factor receptor (EGFR) signaling pathway is key for the induction of React-NSCs and that its inhibition exerts a beneficial effect on the neurogenic niche. We show that during the initial days after the induction of seizures by a single intrahippocampal injection of kainic acid, a strong release of zinc and heparin-binding epidermal growth factor, both activators of the EGFR signaling pathway in neural stem cells, is produced. Administration of the EGFR inhibitor gefitinib, a chemotherapeutic in clinical phase IV, prevents the induction of React-NSCs and preserves neurogenesis.
    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
    静止,成人神经干细胞(NSC)的标志,需要维护NSC池以支持成年齿状回(DG)中的终生连续神经发生。长期持续的表观遗传修饰是否在成年DG中长期维持NSC静止尚不清楚。在这里,我们显示了Sett1a单倍体不足的小鼠,编码组蛋白H3K4甲基转移酶的精神分裂症风险基因,成年后发展出一个扩大的DG,有更多的齿状颗粒细胞。在成年DG的静止NSC中特异性缺失Sett1a促进其激活和神经发生,这通过抑制组蛋白去甲基酶LSD1来抵消。机械上,对培养的静止成年神经干细胞的RNA测序和CUT&RUN分析揭示了Sett1a缺失诱导的转录变化和许多Sett1a靶标。其中Bhlhe40的下调促进体内成年DG中静止的NSC激活。一起,我们的研究揭示了Sett1a依赖的表观遗传机制,该机制维持成年DG的NSC静止。
    Quiescence, a hallmark of adult neural stem cells (NSCs), is required for maintaining the NSC pool to support life-long continuous neurogenesis in the adult dentate gyrus (DG). Whether long-lasting epigenetic modifications maintain NSC quiescence over the long term in the adult DG is not well-understood. Here we show that mice with haploinsufficiency of Setd1a, a schizophrenia risk gene encoding a histone H3K4 methyltransferase, develop an enlarged DG with more dentate granule cells after young adulthood. Deletion of Setd1a specifically in quiescent NSCs in the adult DG promotes their activation and neurogenesis, which is countered by inhibition of the histone demethylase LSD1. Mechanistically, RNA-sequencing and CUT & RUN analyses of cultured quiescent adult NSCs reveal Setd1a deletion-induced transcriptional changes and many Setd1a targets, among which down-regulation of Bhlhe40 promotes quiescent NSC activation in the adult DG in vivo. Together, our study reveals a Setd1a-dependent epigenetic mechanism that sustains NSC quiescence in the adult DG.
    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:创伤性脑损伤(TBI)是年轻人和老年人群的脑损伤的主要原因之一,其心理残疾和死亡率很高。TBI的特点是广泛的细胞死亡,组织损伤和神经炎症,其症状根据从记忆丧失到不可逆转的昏迷和死亡状态的创伤的严重程度而变化。最近,对小鼠模型的临床前研究表明,创伤后成人神经干/祖细胞反应可以代表一个极好的模型,以阐明成人神经发生在损伤后的神经修复作用。细胞周期蛋白依赖性激酶抑制剂p21Waf1/Cip1在调节成年神经干细胞(aNSC)的静止/活化平衡和抑制祖细胞的增殖进程中起关键作用。基于这些考虑,这项工作的目的是评估aNSCS中p21Waf1/Cip1的条件消融如何在生理和创伤后条件下改变成人海马神经发生。
    方法:我们设计了一种新颖的条件p21Waf1/Cip1敲除小鼠模型,其中p21Waf1/Cip1(称为p21)的缺失是受时间控制的,并且发生在Nestin阳性的aNSC中,服用他莫昔芬后。对该小鼠模型(称为p21cKO小鼠)进行受控皮质冲击,以分析p21的缺失如何影响海马小生境内的创伤后神经源性反应。
    结果:数据表明,aNSCs中p21的条件性缺失诱导了海马成年齿状回中aNSCs的活化以及神经祖细胞的增殖和分化的强烈增加,导致神经发生和海马依赖性工作记忆的增强。然而,创伤性脑损伤后,p21cKO小鼠中神经源性反应的增加导致神经源性反应的快速消耗。其次是神经发生下降和海马功能受损。
    结论:这些数据首次证明了p21在调节创伤后海马神经源性反应中的基本作用,通过调节脑损伤后aNSC/祖细胞群的增殖和分化步骤。
    BACKGROUND: Traumatic Brain Injury (TBI) represents one of the main causes of brain damage in young people and the elderly population with a very high rate of psycho-physical disability and death. TBI is characterized by extensive cell death, tissue damage and neuro-inflammation with a symptomatology that varies depending on the severity of the trauma from memory loss to a state of irreversible coma and death. Recently, preclinical studies on mouse models have demonstrated that the post-traumatic adult Neural Stem/Progenitor cells response could represent an excellent model to shed light on the neuro-reparative role of adult neurogenesis following damage. The cyclin-dependent kinase inhibitor p21Waf1/Cip1 plays a pivotal role in modulating the quiescence/activation balance of adult Neural Stem Cells (aNSCs) and in restraining the proliferation progression of progenitor cells. Based on these considerations, the aim of this work is to evaluate how the conditional ablation of p21Waf1/Cip1 in the aNSCS can alter the adult hippocampal neurogenesis in physiological and post-traumatic conditions.
    METHODS: We designed a novel conditional p21Waf1/Cip1 knock-out mouse model, in which the deletion of p21Waf1/Cip1 (referred as p21) is temporally controlled and occurs in Nestin-positive aNSCs, following administration of Tamoxifen. This mouse model (referred as p21 cKO mice) was subjected to Controlled Cortical Impact to analyze how the deletion of p21 could influence the post-traumatic neurogenic response within the hippocampal niche.
    RESULTS: The data demonstrates that the conditional deletion of p21 in the aNSCs induces a strong increase in activation of aNSCs as well as proliferation and differentiation of neural progenitors in the adult dentate gyrus of the hippocampus, resulting in an enhancement of neurogenesis and the hippocampal-dependent working memory. However, following traumatic brain injury, the increased neurogenic response of aNSCs in p21 cKO mice leads to a fast depletion of the aNSCs pool, followed by declined neurogenesis and impaired hippocampal functionality.
    CONCLUSIONS: These data demonstrate for the first time a fundamental role of p21 in modulating the post-traumatic hippocampal neurogenic response, by the regulation of the proliferative and differentiative steps of aNSCs/progenitor populations after brain damage.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号