Retinal progenitor cells

视网膜祖细胞
  • 文章类型: Journal Article
    视网膜祖细胞(RPC)是一种多能且高度增殖的群体,在器官发生过程中会产生所有视网膜细胞类型。定义它们的分子特征是确定治疗视觉障碍的合适方法的关键步骤。这里,我们在3个胚胎阶段对非洲爪的全眼进行了RNA测序,并使用差异表达分析来确定视网膜发生过程中含有增殖和分化RPCs的视组织的转录组学谱.基因本体论和KEGG途径分析表明,在视网膜早期发育的主动RPC增殖期间,与发育途径相关的基因(包括Wnt和Hedgehog信号)被上调(NieuwkoopFaberst。24和27)。发育中的眼睛具有动态表达谱,并在RPC后代规范和分化过程中转向富集代谢过程和光转导(st.35).此外,保守的成人眼再生基因也在早期视网膜发育过程中表达,包括sox2,pax6,nrl,和陷波信号组件。此处提供的眼睛转录组概况涵盖RPC增殖至视网膜发生,并包括再生能力阶段。因此,我们的数据集为揭示RPC活性的分子调节因子提供了丰富的资源,并将允许未来的研究解决眼修复和再生过程中RPC增殖的调节因子.
    Retinal progenitor cells (RPCs) are a multipotent and highly proliferative population that give rise to all retinal cell types during organogenesis. Defining their molecular signature is a key step towards identifying suitable approaches to treat visual impairments. Here, we performed RNA sequencing of whole eyes from Xenopus at three embryonic stages and used differential expression analysis to define the transcriptomic profiles of optic tissues containing proliferating and differentiating RPCs during retinogenesis. Gene Ontology and KEGG pathway analyses showed that genes associated with developmental pathways (including Wnt and Hedgehog signaling) were upregulated during the period of active RPC proliferation in early retinal development (Nieuwkoop Faber st. 24 and 27). Developing eyes had dynamic expression profiles and shifted to enrichment for metabolic processes and phototransduction during RPC progeny specification and differentiation (st. 35). Furthermore, conserved adult eye regeneration genes were also expressed during early retinal development, including sox2, pax6, nrl, and Notch signaling components. The eye transcriptomic profiles presented here span RPC proliferation to retinogenesis and include regrowth-competent stages. Thus, our dataset provides a rich resource to uncover molecular regulators of RPC activity and will allow future studies to address regulators of RPC proliferation during eye repair and regrowth.
    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
    视网膜祖细胞(RPC)是一种多能且高度增殖的群体,在器官发生过程中会产生所有视网膜细胞类型。定义它们的分子特征是确定治疗视觉障碍的合适方法的关键步骤。这里,我们在3个胚胎阶段对非洲爪的全眼进行了RNA测序,并使用差异表达分析来确定视网膜发生过程中含有增殖和分化RPCs的视组织的转录组学谱.基因本体论和KEGG途径分析表明,在视网膜早期发育的主动RPC增殖期间,与发育途径相关的基因(包括Wnt和Hedgehog信号)被上调(NieuwkoopFaberst。24和27)。发育中的眼睛具有动态表达谱,并在RPC后代规范和分化过程中转向富集代谢过程和光转导(st.35).此外,保守的成人眼再生基因也在早期视网膜发育过程中表达,包括sox2,pax6,nrl,和陷波信号组件。此处提供的眼睛转录组概况涵盖了RPC增殖到视网膜发生,并包括再生能力阶段。因此,我们的数据集为揭示RPC活性的分子调节因子提供了丰富的资源,并将允许未来的研究解决眼修复和再生过程中RPC增殖的调节因子.
    Retinal progenitor cells (RPCs) are a multipotent and highly proliferative population that give rise to all retinal cell types during organogenesis. Defining their molecular signature is a key step towards identifying suitable approaches to treat visual impairments. Here, we performed RNA-sequencing of whole eyes from Xenopus at three embryonic stages and used differential expression analysis to define the transcriptomic profiles of optic tissues containing proliferating and differentiating RPCs during retinogenesis. Gene Ontology and KEGG pathway analyses showed that genes associated with developmental pathways (including Wnt and Hedgehog signaling) were upregulated during the period of active RPC proliferation in early retinal development (Nieuwkoop Faber st. 24 and 27). Developing eyes had dynamic expression profiles and shifted to enrichment for metabolic processes and phototransduction during RPC progeny specification and differentiation (st. 35). Furthermore, conserved adult eye regeneration genes were also expressed during early retinal development including sox2, pax6, nrl, and Notch signaling components. The eye transcriptomic profiles presented here span RPC proliferation to retinogenesis and included regrowth-competent stages. Thus, our dataset provides a rich resource to uncover molecular regulators of RPC activity and will allow future studies to address regulators of RPC proliferation during eye repair and regrowth.
    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
    前神经转录因子无碱性螺旋-环-螺旋转录因子7(ATOH7)在发育中的神经视网膜的早期祖细胞中表达。在脊椎动物中,这对视网膜神经节细胞(RGC)的发育至关重要,由于突变动物几乎完全没有RGC,视神经发育不全,和视网膜血管发育的像差。人类突变是罕见的,并导致常染色体隐性视神经发育不全(ONH)或严重的血管变化,诊断为原发性玻璃体常染色体隐性持续增生(PHPVAR)。为了更好地理解ATOH7在神经视网膜发育中的作用,我们创建了ATOH7敲除和表达eGFP的ATOH7报告人诱导多能干细胞(hiPSCs),分化为早期视网膜类器官。由ATOH7调节的靶基因座通过在靶下切割和使用核酸酶的释放进行测序(CUT&RUN-seq)和通过野生型和突变型类器官衍生的报告细胞的RNA测序(RNA-seq)的差异表达来鉴定。此外,对整个类器官进行单细胞RNA测序(scRNA-seq)以鉴定细胞类型特异性基因.突变的类器官在轴突发芽中表现出实质性的缺陷,RGC的减少,以及其他细胞类型的增加。我们确定了469个差异表达的靶基因,属于轴突发育/指导和Notch信号传导的基因过度表达。一起来看,我们通过诱导RGC特异性基因,同时抑制其他细胞命运,巩固了人类ATOH7在指导祖细胞能力方面的功能。此外,我们强调了负责ATOH7相关视神经和视网膜血管异常的候选基因,这揭示了相关疾病的潜在未来治疗目标。
    The proneural transcription factor atonal basic helix-loop-helix transcription factor 7 (ATOH7) is expressed in early progenitors in the developing neuroretina. In vertebrates, this is crucial for the development of retinal ganglion cells (RGCs), as mutant animals show an almost complete absence of RGCs, underdeveloped optic nerves, and aberrations in retinal vessel development. Human mutations are rare and result in autosomal recessive optic nerve hypoplasia (ONH) or severe vascular changes, diagnosed as autosomal recessive persistent hyperplasia of the primary vitreous (PHPVAR). To better understand the role of ATOH7 in neuroretinal development, we created ATOH7 knockout and eGFP-expressing ATOH7 reporter human induced pluripotent stem cells (hiPSCs), which were differentiated into early-stage retinal organoids. Target loci regulated by ATOH7 were identified by Cleavage Under Targets and Release Using Nuclease with sequencing (CUT&RUN-seq) and differential expression by RNA sequencing (RNA-seq) of wildtype and mutant organoid-derived reporter cells. Additionally, single-cell RNA sequencing (scRNA-seq) was performed on whole organoids to identify cell type-specific genes. Mutant organoids displayed substantial deficiency in axon sprouting, reduction in RGCs, and an increase in other cell types. We identified 469 differentially expressed target genes, with an overrepresentation of genes belonging to axon development/guidance and Notch signaling. Taken together, we consolidate the function of human ATOH7 in guiding progenitor competence by inducing RGC-specific genes while inhibiting other cell fates. Furthermore, we highlight candidate genes responsible for ATOH7-associated optic nerve and retinovascular anomalies, which sheds light to potential future therapy targets for related disorders.
    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
    视网膜变性(RD),一组导致不可逆视力丧失的疾病,其特征在于视网膜色素上皮(RPE)或视网膜神经元损伤和丢失。免疫排斥和肿瘤发生的风险较低,干细胞分泌的细胞外囊泡(EV)为RD提供了一种新的无细胞治疗范式,还有待调查。人视网膜类器官衍生的视网膜祖细胞(hERO-RPC)是用于RD治疗的容易获得和先进的细胞来源。然而,hERO-RPC衍生的电动汽车需要进一步表征。这里,我们比较了来自hERO-RPCs(hRPC-EVs)的EVs和作为对照的人类胚胎干细胞(hESC)来源的EVs(hESC-EVs)的特征.基于深入的蛋白质组学分析,我们揭示了hRPC-EV和hESC-EV之间的显着差异。EV与其各自的起源细胞之间的比较表明,hRPC-EV的蛋白质负载比hESC-EV的蛋白质负载更具选择性。特别是,hESC-EV富含与血管生成和细胞周期相关的蛋白质,而hRPC-EV富含与免疫调节和视网膜发育相关的蛋白质。更重要的是,与HESC-EV相比,hRPC-EV与细胞增殖的相关性较低,并且具有调节脂质代谢的独特能力。进一步证实,hRPC-EV可能消除脂质沉积,抑制脂毒性和氧化应激,并增强油酸处理的ARPE-19细胞的吞噬作用和存活。机械上,hRPC-EV整合到油酸处理的ARPE-19细胞的线粒体网络中,并增加线粒体脂肪酸β-氧化相关蛋白的水平。因此,类器官衍生的hRPC-EV代表了RD无细胞治疗的有希望的来源,尤其是与RPE细胞脂质代谢异常相关的致盲疾病。
    Retinal degeneration (RD), a group of diseases leading to irreversible vision loss, is characterised by retinal pigment epithelium (RPE) or retinal neuron damage and loss. With fewer risks of immune rejection and tumorigenesis, stem cell-secreted extracellular vesicles (EVs) offer a new cell-free therapeutic paradigm for RD, which remains to be investigated. Human retinal organoid-derived retinal progenitor cells (hERO-RPCs) are an easily accessible and advanced cell source for RD treatment. However, hERO-RPCs-derived EVs require further characterisation. Here, we compared the characteristics of EVs from hERO-RPCs (hRPC-EVs) with those of human embryonic stem cell (hESC)-derived EVs (hESC-EVs) as controls. Based on in-depth proteomic analysis, we revealed remarkable differences between hRPC-EVs and hESC-EVs. A comparison between EVs and their respective cells of origin demonstrated that the protein loading of hRPC-EVs was more selective than that of hESC-EVs. In particular, hESC-EVs were enriched with proteins related to angiogenesis and cell cycle, whereas hRPC-EVs were enriched with proteins associated with immune modulation and retinal development. More importantly, compared with that of hESC-EVs, hRPC-EVs exhibited a lower correlation with cell proliferation and a unique capacity to regulate lipid metabolism. It was further confirmed that hRPC-EVs potentially eliminated lipid deposits, inhibited lipotoxicity and oxidative stress, and enhanced phagocytosis and survival of oleic acid-treated ARPE-19 cells. Mechanistically, hRPC-EVs are integrated into the mitochondrial network of oleic acid-treated ARPE-19 cells, and increased the level of mitochondrial fatty acid β-oxidation-related proteins. Thus, organoid-derived hRPC-EVs represent a promising source of cell-free therapy for RD, especially for blinding diseases related to abnormal lipid metabolism in RPE cells.
    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
    目的:观察低氧浓度对人诱导多能干细胞(hiPSC)来源的视网膜类器官(ROs)神经视网膜的影响。
    方法:实验使用hiPSC和三维培养方法。将生成的胚状体(EBs)随机平均分为低氧和常氧组。在第38、45和52天拍摄EB的照片,并计算相应的EB体积。同时,在这三个时间点收集样本,其次是固定,切片,和免疫荧光。
    结果:在第38天,Ki67阳性增殖细胞的比例稳定增加;这种增殖促进作用倾向于增加组织密度而不是组织体积。在第45天和第52天,两组具有相对相似的Ki67阳性细胞比率。进一步的免疫荧光分析显示,第52天,神经视网膜内SOX2阳性细胞的比例显着增加(P<0.05)。相比之下,在所有三个时间点,PAX6和CHX10阳性细胞的百分比在缺氧处理后均显着降低(P<0.01),第45天CHX10除外(P>0.05)。此外,PAX6-/TUJ1+细胞在神经视网膜内的比例显著增加(分别为P<0.01,<0.05,<0.05)。
    结论:低氧促进神经视网膜的干性和增殖,这表明低氧条件可以扩大hiPSC衍生的RO中的视网膜祖细胞池。
    OBJECTIVE: To observe the effect of low oxygen concentration on the neural retina in human induced pluripotent stem cell (hiPSC)-derived retinal organoids (ROs).
    METHODS: The hiPSC and a three-dimensional culture method were used for the experiments. Generated embryoid bodies (EBs) were randomly and equally divided into hypoxic and normoxic groups. Photographs of the EBs were taken on days 38, 45, and 52, and the corresponding volume of EBs was calculated. Simultaneously, samples were collected at these three timepoints, followed by fixation, sectioning, and immunofluorescence.
    RESULTS: The proportion of Ki67-positive proliferating cells increased steadily on day 38; this proliferation-promoting effect tended to increase tissue density rather than tissue volume. On days 45 and 52, the two groups had relatively similar ratios of Ki67-positive cells. Further immunofluorescence analysis showed that the ratio of SOX2-positive cells significantly increased within the neural retina on day 52 (P<0.05). In contrast, the percentage of PAX6- and CHX10-positive cells significantly decreased following hypoxia treatment at all three timepoints (P<0.01), except for CHX10 at day 45 (P>0.05). Moreover, the proportion of PAX6-/TUJ1+ cells within the neural retinas increased considerably (P<0.01, <0.05, <0.05 respectively).
    CONCLUSIONS: Low oxygen promotes stemness and proliferation of neural retinas, suggesting that hypoxic conditions can enlarge the retinal progenitor cell pool in hiPSC-derived ROs.
    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
    基于细胞的疗法显示出巨大的潜力,因为它们能够取代垂死的视网膜神经元细胞并保持视力。移民,视网膜祖细胞(RPCs)的增殖和分化在RPCs移植到宿主时整合到视网膜中起着至关重要的作用。我们的研究旨在探讨透明质酸(HA)-CD44相互作用对RPCs迁移调节的影响,增殖和分化,并研究潜在的监管机制。我们发现CD44在RPCs中表达,HA-CD44相互作用显着改善了RPCs的粘附和迁移。通过HA-CD44相互作用刺激microRNA-21(miR-21)表达在RPC中是蛋白激酶C(PKC)/Nanog依赖性的。用PKC或Nanog特异性ASODN或miR-21antagomir处理RPCs可有效阻断HA介导的RPCs粘附和迁移。此外,HA-CD44相互作用介导的RPCs增殖和神经元分化需要Rho-激酶(ROK)/Grb2相关结合剂(Gab-1)相关磷脂酰肌醇3激酶(PI3K)/AKT信号传导激活。我们的发现证明了HA-CD44相互作用在调节迁移中的新作用,RPCs的增殖和神经元分化。HA-CD44信号可以代表一种控制RPC命运的新方法,这些发现可能对RPCs在未来的治疗应用具有指导意义。
    Cell-based therapies have shown great potential because of their abilities to replace dying retinal neuron cells and preserve vision. The migration, proliferation and differentiation of retinal progenitor cells(RPCs) plays a vital role in the integration of the RPCs into the retina when transplanted into the host. Our study aimed to explore the effects of Hyaluronan(HA)-CD44 interactions on the regulation of RPCs migration, proliferation and differentiation, and investigate the underlying regulatory mechanisms. We found that CD44 was expressed in RPCs, and the HA-CD44 interaction markedly improved RPCs adhesion and migration. The stimulation of microRNA-21(miR-21) expression by the HA-CD44 interaction was protein kinase C (PKC)/Nanog-dependent in RPCs. Treatment of RPCs with PKC- or Nanog-specific ASODN or miR-21 antagomir effectively blocked HA-mediated RPCs adhesion and migration. Moreover, Rho-Kinase(ROK)/ Grb2-associated binders(Gab-1) associated phosphatidylinositol 3-kinase(PI3K)/AKT signalling activation was required for HA-CD44 interaction mediated RPCs proliferation and neuronal differentiation. Our findings demonstrated new roles for the HA-CD44 interaction in regulating the migration, proliferation and neuronal differentiation of RPCs. HA-CD44 signalling could represent a novel approach to controlling RPC fates, and the findings may be instructive for the application of RPCs for future therapeutic applications.
    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
    弓形虫病影响世界人口的三分之一,并以原生动物弓形虫为病原体。先天性弓形虫病(CT)可对胎儿造成严重损害,包括流产,颅内钙化,脑积水和视网膜脉络炎。CT的严重程度取决于发生感染的妊娠期,并且已经报道了视网膜发育过程中细胞水平的改变。在这项研究中,我们提出了一种小鼠CT模型来研究感染对视网膜发育的影响。
    在胚胎第10天(E10),用两个弓形虫囊肿(ME49株)对色素C57BL/6品系小鼠的怀孕雌性进行胃内感染,并在E18分析后代。
    受感染的胚胎的体型和体重明显小于PBS处理的对照组,表明胚胎发育受到影响。在视网膜上,与对照组相比,在感染小鼠的NBL顶端区域发现Ki-67阳性细胞(增殖细胞标志物)数量显著增加.支持这一点,细胞周期蛋白CyclinD3,Cdk6和pChK2在感染的视网膜中发生显着变化。有趣的是,免疫组织化学分析显示β-III-微管蛋白阳性细胞的数量显着增加,神经元分化的最早标志之一。
    我们的数据表明CT影响视网膜祖细胞的细胞周期进程,可能诱导这些细胞停滞在G2/M期。这种改变可能会影响分化,预测/增加神经元成熟,从而导致异常的视网膜形成。我们的模型模拟了眼CT中观察到的重要事件。
    UNASSIGNED: Toxoplasmosis affects one third of the world population and has the protozoan Toxoplasma gondii as etiological agent. Congenital toxoplasmosis (CT) can cause severe damage to the fetus, including miscarriages, intracranial calcification, hydrocephalus and retinochoroiditis. Severity of CT depends on the gestational period in which infection occurs, and alterations at the cellular level during retinal development have been reported. In this study, we proposed a mouse CT model to investigate the impact of infection on retinal development.
    UNASSIGNED: Pregnant females of pigmented C57BL/6 strain mice were infected intragastrically with two T. gondii cysts (ME49 strain) at embryonic day 10 (E10), and the offspring were analyzed at E18.
    UNASSIGNED: Infected embryos had significantly smaller body sizes and weights than the PBS-treated controls, indicating that embryonic development was affected. In the retina, a significant increase in the number of Ki-67-positive cells (marker of proliferating cells) was found in the apical region of the NBL of infected mice compared to the control. Supporting this, cell cycle proteins Cyclin D3, Cdk6 and pChK2 were significantly altered in infected retinas. Interestingly, the immunohistochemical analysis showed a significant increase in the population of β-III-tubulin-positive cells, one of the earliest markers of neuronal differentiation.
    UNASSIGNED: Our data suggests that CT affects cell cycle progression in retinal progenitor cells, possibly inducing the arrest of these cells at G2/M phase. Such alterations could influence the differentiation, anticipating/increasing neuronal maturation, and therefore leading to abnormal retinal formation. Our model mimics important events observed in ocular CT.
    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
    视网膜变性,以视网膜神经元的进行性丧失为特征,是无法治愈的视力障碍的主要原因。基于视网膜祖细胞(RPCs)的移植可以促进视力恢复,但是,由于RPCs的神经源性分化不精确,并破坏了被严重氧化性视网膜病变包围的移植细胞的功能,因此该过程的临床疗效受到损害。这里,结果表明,超薄碳化铌(Nb2C)MXene可以增强RPC的视网膜再生性能。具有中等光热效应的Nb2CMXene通过激活细胞内信号显着改善RPCs的视网膜神经元分化,除了通过同时清除自由基来实现高效的RPC保护之外,全面的生物医学评估和理论计算已经有力地证明了这一点。在MXene辅助RPCs视网膜下移植到典型的视网膜变性10(rd10)小鼠中时,观察到神经元分化显着增加,从而有助于视网膜结构和视觉功能的有效恢复。MXene的双重内在功能协同辅助RPC移植,这代表了视觉恢复研究领域的一个有趣的范例,并将拓宽纳米医学的多功能性领域。
    Retinal degeneration, characterized by the progressive loss of retinal neurons, is the leading cause of incurable visual impairment. Retinal progenitor cells (RPCs)-based transplantation can facilitate sight restoration, but the clinical efficacy of this process is compromised by the imprecise neurogenic differentiation of RPCs and undermining function of transplanted cells surrounded by severely oxidative retinal lesions. Here, it is shown that ultrathin niobium carbide (Nb2 C) MXene enables performance enhancement of RPCs for retinal regeneration. Nb2 C MXene with moderate photothermal effect markedly improves retinal neuronal differentiation of RPCs by activating intracellular signaling, in addition to the highly effective RPC protection by scavenging free radicals concurrently, which has been solidly evidenced by the comprehensive biomedical assessments and theoretical calculations. A dramatically increased neuronal differentiation is observed upon subretinal transplantation of MXene-assisted RPCs into the typical retinal degeneration 10 (rd10) mice, thereby contributing to the efficient restoration of retinal architecture and visual function. The dual-intrinsic function of MXene synergistically aids RPC transplantation, which represents an intriguing paradigm in vision-restoration research filed, and will broaden the multifunctionality horizon of nanomedicine.
    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
    目的:探讨人胚胎干细胞来源的视网膜前体细胞(hERO-RPCs)视网膜下移植是否能促进Müller胶质细胞去分化和转分化。从而改善视觉功能,延缓视网膜退行性进展。
    方法:将hERO-RPCs视网膜下移植到皇家外科医学院(RCS)大鼠体内。术后4周和8周进行视网膜电图(ERG)记录以评估视网膜功能。使用免疫荧光,在术后2,4和8wk观察外核层(ONL)厚度和视网膜Müller胶质的变化.为了验证hERO-RPCs对Müller胶质细胞的体外作用,我们用Transwell系统将hERO-RPCs与Müller胶质细胞共培养。共文化之后,进行Ki67染色和定量聚合酶链反应(qPCR)以分别测量Müller胶质细胞的增殖和mRNA水平。细胞迁移实验用于检测hERO-RPCs对Müller胶质细胞迁移的影响。两组之间的比较通过非配对学生t检验进行,多组间的比较采用单因素方差分析和Tukey多重比较检验。
    结果:术后4周和8周移植hERO-RPCs可明显改善RCS大鼠的视功能和ONL厚度。除了在术后4周和8周抑制胶质增生,hERO-RPCs显著增加Müller胶质细胞去分化相关转录因子的表达,并促进术后2、4和8wk的迁移,而不是这些细胞在RCS大鼠中的转分化。体外,使用Transwell系统,我们发现hERO-RPCs促进原代大鼠Müller胶质细胞的增殖和迁移,并在mRNA水平诱导其去分化。
    结论:这些结果表明,hERO-RPCs可能促进Müller胶质细胞的早期去分化,这可能为干细胞治疗和穆勒胶质细胞重编程的机制提供新的见解,有助于开发视网膜变性疾病的新疗法。
    OBJECTIVE: To explore whether the subretinal transplantation of retinal progenitor cells from human embryonic stem cell-derived retinal organoid (hERO-RPCs) could promote Müller glia dedifferentiation and transdifferentiation, thus improving visual function and delaying retinal degenerative progression.
    METHODS: hERO-RPCs were subretinally transplanted into Royal College of Surgeons (RCS) rats. Electroretinography (ERG) recording was performed at 4 and 8wk postoperation to assess retinal function. Using immunofluorescence, the changes in outer nuclear layer (ONL) thickness and retinal Müller glia were explored at 2, 4, and 8wk postoperation. To verify the effect of hERO-RPCs on Müller glia in vitro, we cocultured hERO-RPCs with Müller glia with a Transwell system. After coculture, Ki67 staining and quantitative polymerase chain reaction (qPCR) were performed to measure the proliferation and mRNA levels of Müller glia respectively. Cell migration experiment was used to detect the effect of hERO-RPCs on Müller glial migration. Comparisons between two groups were performed by the unpaired Student\'s t-test, and comparisons among multiple groups were made with one-way ANOVA followed by Tukey\'s multiple comparison test.
    RESULTS: The visual function and ONL thickness of RCS rats were significantly improved by transplantation of hERO-RPCs at 4 and 8wk postoperation. In addition to inhibiting gliosis at 4 and 8wk postoperation, hERO-RPCs significantly increased the expression of dedifferentiation-associated transcriptional factor in Müller glia and promoted the migration at 2, 4 and 8wk postoperation, but not the transdifferentiation of these cells in RCS rats. In vitro, using the Transwell system, we found that hERO-RPCs promoted the proliferation and migration of primary rat Müller glia and induced their dedifferentiation at the mRNA level.
    CONCLUSIONS: These results show that hERO-RPCs might promote early dedifferentiation of Müller glia, which may provide novel insights into the mechanisms of stem cell therapy and Müller glial reprogramming, contributing to the development of novel therapies for retinal degeneration disorders.
    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
    遗传性和年龄相关的视网膜变性是没有有效治疗的失明的最常见原因。视网膜祖细胞(RPCs),它们具有分化为各种视网膜细胞类型的多能性,被认为是视网膜退行性疾病的细胞移植治疗的有希望的来源。然而,RPCs的自限扩增导致细胞来源供应困难,限制了其临床治疗。在这项工作中,我们发现抑制RPCs中的microRNA-449a(miR-449a)可以促进RPCs的增殖和抑制其凋亡,部分通过上调Notch信号。通过内皮细胞来源的外泌体将miR-449a抑制剂转导到RPCs中的进一步优化可以促进体内移植的RPCs的存活并减少视网膜变性小鼠模型中的细胞凋亡。总之,这些研究表明,exosome-miR-449a抑制剂可以有效促进RPCs的体外扩增,增强移植的RPCs在体内的存活,这可能为将来的视网膜变性提供一种新的干预策略。
    Inherited and age-related retinal degenerations are the commonest causes of blindness without effective treatments. Retinal progenitor cells (RPCs), which have the multipotency to differentiate into various retinal cell types, are regarded as a promising source of cell transplantation therapy for retinal degenerative diseases. However, the self-limited expansion of RPCs causes difficulty in cell source supply and restrict its clinical treatment. In this work, we found that inhibition of microRNA-449a (miR-449a) in RPCs can promote proliferation and inhibit apoptosis of RPCs, partially through upregulating Notch signaling. Further optimization of transduction miR-449a inhibitor into RPCs by endothelial cell-derived exosomes can promote the survival of RPCs transplanted in vivo and reduce cell apoptosis in retinal degeneration mouse models. In summary, these studies have shown that exosome-miR-449a inhibitor can effectively promote the expansion of RPCs in vitro and enhance transplanted RPCs survival in vivo, which might provide a novel intervention strategy for retinal degenerations in the future.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号