iPSC

iPSC
  • 文章类型: Journal Article
    人诱导多能干细胞衍生的感觉神经元(iPSC-dSN)模型是研究神经毒性的宝贵资源,但受复制性和可重复性差的影响。通常是由于缺乏优化。这里,我们确定了与培养条件相关的实验因素,这些因素会对体外细胞药物反应产生重大影响,并确定了改善复制性和可重复性的最佳条件。治疗时间和细胞接种密度都是重要因素,而细胞系的差异也促成了变异。在暴露于多西他赛或紫杉醇48小时后,证明了活力的可复制剂量反应。此外,证明了神经突生长的可复制剂量依赖性减少,证明该模型对其他表型检查的适用性。总的来说,我们建立了一个优化的iPSC-dSN模型,用于研究紫杉烷诱导的神经毒性。
    Human induced pluripotent stem cell-derived sensory neuron (iPSC-dSN) models are a valuable resource for the study of neurotoxicity but are affected by poor replicability and reproducibility, often due to a lack of optimization. Here, we identify experimental factors related to culture conditions that substantially impact cellular drug response in vitro and determine optimal conditions for improved replicability and reproducibility. Treatment duration and cell seeding density were both found to be significant factors, while cell line differences also contributed to variation. A replicable dose-response in viability was demonstrated after 48-h exposure to docetaxel or paclitaxel. Additionally, a replicable dose-dependent reduction in neurite outgrowth was demonstrated, demonstrating the applicability of the model for the examination of additional phenotypes. Overall, we have established an optimized iPSC-dSN model for the study of taxane-induced neurotoxicity.
    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
    新烟碱是合成的,尼古丁来源的杀虫剂在全球范围内用于保护农作物和家畜免受害虫侵害。报道的证据表明,它们也能够与哺乳动物尼古丁受体(nAChRs)相互作用,在培养的神经元中触发有害反应。在胎儿期暴露于高新烟碱水平会在动物模型中引起神经毒性。考虑到这些杀虫剂的持续暴露和nAChRs在大脑发育中的关键作用,它们对哺乳动物中枢神经系统(CNS)的潜在神经毒性需要进一步研究.我们在这里研究了不同世代的新烟碱对小鼠胎儿大脑和原代培养物以及从人类诱导多能干细胞(iPSC)获得的神经元细胞和类器官的中枢神经系统细胞的神经发育作用。新烟碱显著影响神经元的活力,吡虫啉(IMI)诱导突触蛋白表达的相关改变,神经丝结构,和体外小胶质细胞激活,在产前暴露的小鼠胎儿的大脑中。IMI还对发育中的人iPSC衍生的神经元和皮质类器官诱导神经毒性作用。总的来说,目前的研究结果表明,新烟碱类可能在小鼠和人类中枢神经系统细胞的神经/免疫发育过程中引起损害,并为此类农药暴露风险的表征提供了新的见解.
    Neonicotinoids are synthetic, nicotine-derived insecticides used worldwide to protect crops and domestic animals from pest insects. The reported evidence shows that they are also able to interact with mammalian nicotine receptors (nAChRs), triggering detrimental responses in cultured neurons. Exposure to high neonicotinoid levels during the fetal period induces neurotoxicity in animal models. Considering the persistent exposure to these insecticides and the key role of nAChRs in brain development, their potential neurotoxicity on mammal central nervous system (CNS) needs further investigations. We studied here the neurodevelopmental effects of different generations of neonicotinoids on CNS cells in mouse fetal brain and primary cultures and in neuronal cells and organoids obtained from human induced pluripotent stem cells (iPSC). Neonicotinoids significantly affect neuron viability, with imidacloprid (IMI) inducing relevant alterations in synaptic protein expression, neurofilament structures, and microglia activation in vitro, and in the brain of prenatally exposed mouse fetuses. IMI induces neurotoxic effects also on developing human iPSC-derived neurons and cortical organoids. Collectively, the current findings show that neonicotinoids might induce impairment during neuro/immune-development in mouse and human CNS cells and provide new insights in the characterization of risk for the exposure to this class of pesticides.
    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:肺癌仍然是全球癌症相关死亡率的主要原因。尽管最近的治疗进展提供了有针对性的治疗方法,耐药性和全身毒性的发展仍然是人们关注的主要问题.细胞外囊泡(EV),特别是那些来自间充质基质细胞(MSC),作为有前途的药物递送系统获得了关注,提供生物相容性和最小的免疫反应。认识到常规2D细胞培养系统在模拟肿瘤微环境方面的局限性,本研究旨在描述在肺癌和正常肺组织中使用患者特异性类器官模型的原理验证方法,以及在个性化医疗方法中使用源自诱导多能干细胞(iPSC)-MSC的自体EVs的可行性.
    方法:首先,我们将健康的成纤维细胞重新编程为iPSC。接下来,我们将患者来源的iPSC分化为分支肺类器官(BLO),并从患者来源的肿瘤组织中产生患者匹配的肺癌类器官(LCO).我们展示了从iPSC分化MSC和从iPSC-MSC分离EV的简化过程,用0.07µg/mL的细胞毒性剂顺铂封装,并应用于两个类器官模型。用LDH和CCK8试验记录顺铂和负载顺铂的EV的细胞毒性。
    结果:成纤维细胞来源的iPSC显示正常核型,多能性染色,和三系分化。iPSC来源的BLO显示肺标志物的表达,如TMPRSS2和MUC5A,而患者匹配的LCO显示Napsin和CK5的表达。接下来,我们比较了在肺癌类器官模型和健康肺类器官模型中,负载顺铂的iPSC-MSC来源的EV与空EV和单用顺铂的效果.不出所料,当LCO用20µg/mL顺铂治疗时,我们发现细胞毒性作用.用空EV治疗LCO和BLO在24小时后产生细胞毒性作用。然而,装载0.07µg/mL顺铂的EV在两个类器官模型中均未诱导任何细胞毒性作用。
    结论:我们报告了未来使用自体或同种异体iPSC-MSCEVs作为肺癌药物递送试验的原理证明。然而,由于时间和劳动力密集的过程,我们得出的结论是,目前这种管道对于个性化方法可能不可行。
    BACKGROUND: Lung cancer remains a leading cause of cancer-related mortality globally. Although recent therapeutic advancements have provided targeted treatment approaches, the development of resistance and systemic toxicity remain primary concerns. Extracellular vesicles (EVs), especially those derived from mesenchymal stromal cells (MSC), have gained attention as promising drug delivery systems, offering biocompatibility and minimal immune responses. Recognizing the limitations of conventional 2D cell culture systems in mimicking the tumor microenvironment, this study aims to describe a proof-of-principle approach for using patient-specific organoid models for both lung cancer and normal lung tissue and the feasibility of employing autologous EVs derived from induced pluripotent stem cell (iPSC)-MSC in personalized medicine approaches.
    METHODS: First, we reprogrammed healthy fibroblasts into iPSC. Next, we differentiated patient-derived iPSC into branching lung organoids (BLO) and generated patient-matched lung cancer organoids (LCO) from patient-derived tumor tissue. We show a streamlined process of MSC differentiation from iPSC and EV isolation from iPSC-MSC, encapsulated with 0.07 µg/mL of cytotoxic agent cisplatin and applied to both organoid models. Cytotoxicity of cisplatin and cisplatin-loaded EVs was recorded with LDH and CCK8 tests.
    RESULTS: Fibroblast-derived iPSC showed a normal karyotype, pluripotency staining, and trilineage differentiation. iPSC-derived BLO showed expression of lung markers, like TMPRSS2 and MUC5A while patient-matched LCO showed expression of Napsin and CK5. Next, we compared the effects of iPSC-MSC derived EVs loaded with cisplatin against empty EVs and cisplatin alone in lung cancer organoid and healthy lung organoid models. As expected, we found a cytotoxic effect when LCO were treated with 20 µg/mL cisplatin. Treatment of LCO and BLO with empty EVs resulted in a cytotoxic effect after 24 h. However, EVs loaded with 0.07 µg/mL cisplatin failed to induce any cytotoxic effect in both organoid models.
    CONCLUSIONS: We report on a proof-of-principle pipeline towards using autologous or allogeneic iPSC-MSC EVs as drug delivery tests for lung cancer in future. However, due to the time and labor-intensive processes, we conclude that this pipeline might not be feasible for personalized approaches at the moment.
    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
    Krabbe病(Kd)是由溶酶体半乳糖神经酰胺酶(GALC)的缺乏引起的溶酶体贮积症(LSD),该酶裂解富含髓磷脂的脂质半乳糖神经酰胺(GalCer)。积累的GalCer被分解代谢成细胞毒性脂质精神病,导致髓鞘细胞死亡和脱髓鞘,从而募集小胶质细胞/巨噬细胞,这些小胶质细胞/巨噬细胞无法消化髓鞘碎片并成为球形细胞。这里,为了了解Kd的病理机制,我们使用来自Kd患者的诱导多能干细胞(iPSCs)产生髓鞘形成类器官和小胶质细胞.我们表明Kd类器官在神经发生中没有明显的缺陷,天体发生,和少突发生,但表现出早期髓鞘形成缺陷。具体来说,Kd类器官在髓鞘形成高峰时显示出较短但与对照组相似的髓鞘节间数量,并且在较晚的时间点显示出减少的数量和较短的节间。有趣的是,在缺乏自噬和mTOR通路失调的情况下,髓鞘受到影响,提示缺乏溶酶体功能障碍,这使得这种类器官模型成为研究驱动Kd脱髓鞘的早期事件的非常有价值的工具。KdiPSC衍生的小胶质细胞在正常培养条件下显示出球状细胞形成的边缘速率,在GalCer取食后急剧增加。在正常培养条件下,Kd小胶质细胞表示出轻微的LAMP1含量降低和轻微的自噬卵白LC3B增加。在GalCer进食后,Kd细胞显示自噬蛋白的积累和强大的LAMP1减少,在稍后的时间点恢复,显示球形细胞的补偿能力。总之,这支持了我们的培养物作为研究驱动球形细胞形成的机制和克服GalCer在Kd中积累的代偿机制的工具的价值。
    Krabbe disease (Kd) is a lysosomal storage disorder (LSD) caused by the deficiency of the lysosomal galactosylceramidase (GALC) which cleaves the myelin enriched lipid galactosylceramide (GalCer). Accumulated GalCer is catabolized into the cytotoxic lipid psychosine that causes myelinating cells death and demyelination which recruits microglia/macrophages that fail to digest myelin debris and become globoid cells. Here, to understand the pathological mechanisms of Kd, we used induced pluripotent stem cells (iPSCs) from Kd patients to produce myelinating organoids and microglia. We show that Kd organoids have no obvious defects in neurogenesis, astrogenesis, and oligodendrogenesis but manifest early myelination defects. Specifically, Kd organoids showed shorter but a similar number of myelin internodes than Controls at the peak of myelination and a reduced number and shorter internodes at a later time point. Interestingly, myelin is affected in the absence of autophagy and mTOR pathway dysregulation, suggesting lack of lysosomal dysfunction which makes this organoid model a very valuable tool to study the early events that drive demyelination in Kd. Kd iPSC-derived microglia show a marginal rate of globoid cell formation under normal culture conditions that is drastically increased upon GalCer feeding. Under normal culture conditions, Kd microglia show a minor LAMP1 content decrease and a slight increase in the autophagy protein LC3B. Upon GalCer feeding, Kd cells show accumulation of autophagy proteins and strong LAMP1 reduction that at a later time point are reverted showing the compensatory capabilities of globoid cells. Altogether, this supports the value of our cultures as tools to study the mechanisms that drive globoid cell formation and the compensatory mechanism in play to overcome GalCer accumulation in Kd.
    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
    心力衰竭(HF)是全球住院和死亡的主要原因之一,已尝试多种干细胞疗法来加速梗死区的再生.已经出现了多种策略来建立用于再生心脏病学的心肌细胞谱系的细胞候选物。这篇文章阐述了对新兴技术的重要见解,目前的方法,和翻译承诺编程不同类型的细胞用于心脏再生。
    With heart failure (HF) being one of the leading causes of hospitalization and death worldwide, multiple stem cell therapies have been attempted to accelerate the regeneration of the infarct zone. Versatile strategies have emerged to establish the cell candidates of cardiomyocyte lineage for regenerative cardiology. This article illustrates critical insights into the emerging technologies, current approaches, and translational promises on the programming of diverse cell types for cardiac 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
    CRISPR-Cas9介导的基因编辑在基础和临床研究中具有广泛的应用,并且是几种疾病的有希望的工具。我们的实验室以前开发了一种非整合RNA病毒,麻疹病毒(MeV),通过用诱导多能干细胞生成的重编程因子替换病毒附着蛋白作为单周期重编程载体。受MeV重编程矢量效率的鼓舞,在这项研究中,我们开发了一种单周期MeV载体,将gRNA和Cas9核酸酶传递给人类细胞,以进行有效的基因编辑。我们证明MeV载体在人细胞中实现了报道基因(mCherry)和内源基因(HBB和FANCD1)的靶基因编辑。此外,MeV载体通过使用单链寡核苷酸供体的同源定向修复实现了精确的敲入。MeV载体是人类细胞中基因敲除和敲入修饰的新的灵活平台,能够在新技术发展的过程中融入它们。
    CRISPR-Cas9-mediated gene editing has vast applications in basic and clinical research and is a promising tool for several disorders. Our lab previously developed a non-integrating RNA virus, measles virus (MeV), as a single-cycle reprogramming vector by replacing the viral attachment protein with the reprogramming factors for induced pluripotent stem cell generation. Encouraged by the MeV reprogramming vector efficiency, in this study, we develop a single-cycle MeV vector to deliver the gRNA(s) and Cas9 nuclease to human cells for efficient gene editing. We show that the MeV vector achieved on-target gene editing of the reporter (mCherry) and endogenous genes (HBB and FANCD1) in human cells. Additionally, the MeV vector achieved precise knock-in via homology-directed repair using a single-stranded oligonucleotide donor. The MeV vector is a new and flexible platform for gene knock-out and knock-in modifications in human cells, capable of incorporating new technologies as they are developed.
    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
    初级纤毛是指状感觉器官,从大多数细胞类型的身体延伸出来,并具有与质膜不同的脂质和蛋白质组成。这种分配是由限制非纤毛蛋白进入的扩散屏障维持的,并允许选择性进入具有睫状靶向序列(CTS)的蛋白质。然而,CTSs不是定型的,并且先前报道的序列不足以驱动跨不同细胞类型的有效纤毛定位。这里,我们描述了一个短的肽序列,有效地将跨膜蛋白靶向所有测试细胞类型的初级纤毛,包括人类神经元。我们产生了稳定表达跨膜构建体的人诱导多能干细胞(hiPSC)系,该构建体带有细胞外HaloTag和胞内荧光蛋白,这使得光明,神经元和其他细胞类型中初级纤毛的特异性标记,以促进纤毛在健康和疾病中的研究。我们通过开发用于自动测量初级纤毛的图像分析管道来检测与信号传导或疾病状态改变相关的长度变化,从而证明了该资源的实用性。
    Primary cilia are finger-like sensory organelles that extend from the bodies of most cell types and have a distinct lipid and protein composition from the plasma membrane. This partitioning is maintained by a diffusion barrier that restricts the entry of non-ciliary proteins, and allows the selective entry of proteins harboring a ciliary targeting sequence (CTS). However, CTSs are not stereotyped and previously reported sequences are insufficient to drive efficient ciliary localisation across diverse cell types. Here, we describe a short peptide sequence that efficiently targets transmembrane proteins to primary cilia in all tested cell types, including human neurons. We generate human-induced pluripotent stem cell (hiPSC) lines stably expressing a transmembrane construct bearing an extracellular HaloTag and intracellular fluorescent protein, which enables the bright, specific labeling of primary cilia in neurons and other cell types to facilitate studies of cilia in health and disease. We demonstrate the utility of this resource by developing an image analysis pipeline for the automated measurement of primary cilia to detect changes in their length associated with altered signaling or disease state.
    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
    过度的炎症反应和氧化应激是自闭症中公认的分子发现,这些过程可能会影响表观遗传景观或受其影响。尽管如此,没有足够的治疗方法,因为用于个体化治疗的患者特异性脑分子标志物仍然具有挑战性。
    方法:我们使用了自闭症患者的iPSC来源的神经元和星形胶质细胞对照组(5/组),以检查他们是否复制了自闭症的死后脑表达/表观遗传学改变。此外,分析10个死后脑样品(5个/组)的DNA甲基化的PSC衍生细胞中受影响的基因。
    结果:我们发现TGFB1,TGFB2,IL6和IFI16的过度表达和HAP1,SIRT1,NURR1,RELN,孤独症患者星形胶质细胞中的GPX1,EN2,SLC1A2和SLC1A3,随着TGFB2,IL6,TNFA和EN2基因启动子的DNA低甲基化以及自闭症患者星形胶质细胞中HAP1启动子5-羟甲基化的减少。在神经元中,HAP1和IL6表达趋势相似。虽然HAP1启动子在神经元中高度甲基化,IFI16和SLC1A3启动子被低甲基化,并且TGFB2表现出增加的启动子5-羟基甲基化。我们还发现神经元乔化减少,脊柱尺寸,增长率,和移民,但是自闭症患者的星形胶质细胞大小增加,生长速度降低。在死后的大脑样本中,我们发现TGFB2和IFI16启动子区的DNA低甲基化,但自闭症中HAP1和SLC1A2启动子的DNA甲基化。
    结论:iPSC来源的细胞中自闭症相关的表达/表观遗传学改变复制了文献中报道的那些,使它们成为研究疾病发病机理或患者特异性疗法的适当替代品。
    Excessive inflammatory reactions and oxidative stress are well-recognized molecular findings in autism and these processes can affect or be affected by the epigenetic landscape. Nonetheless, adequate therapeutics are unavailable, as patient-specific brain molecular markers for individualized therapies remain challenging.
    METHODS: We used iPSC-derived neurons and astrocytes of patients with autism vs. controls (5/group) to examine whether they replicate the postmortem brain expression/epigenetic alterations of autism. Additionally, DNA methylation of 10 postmortem brain samples (5/group) was analyzed for genes affected in PSC-derived cells.
    RESULTS: We found hyperexpression of TGFB1, TGFB2, IL6 and IFI16 and decreased expression of HAP1, SIRT1, NURR1, RELN, GPX1, EN2, SLC1A2 and SLC1A3 in the astrocytes of patients with autism, along with DNA hypomethylation of TGFB2, IL6, TNFA and EN2 gene promoters and a decrease in HAP1 promoter 5-hydroxymethylation in the astrocytes of patients with autism. In neurons, HAP1 and IL6 expression trended alike. While HAP1 promoter was hypermethylated in neurons, IFI16 and SLC1A3 promoters were hypomethylated and TGFB2 exhibited increased promoter 5-hydroxymethlation. We also found a reduction in neuronal arborization, spine size, growth rate, and migration, but increased astrocyte size and a reduced growth rate in autism. In postmortem brain samples, we found DNA hypomethylation of TGFB2 and IFI16 promoter regions, but DNA hypermethylation of HAP1 and SLC1A2 promoters in autism.
    CONCLUSIONS: Autism-associated expression/epigenetic alterations in iPSC-derived cells replicated those reported in the literature, making them appropriate surrogates to study disease pathogenesis or patient-specific therapeutics.
    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:哺乳动物脑中神经干细胞(NSC)的增殖和分化在出生后降低到最低水平。然而,在啮齿动物的成年皮质和海马中存在神经源性生态位,灵长类动物和人类,成人NSC分化与发展共享关键的监管机制。成人神经发生损伤与阿尔茨海默病(AD)病理有关。通过使用神经营养因子来解决这些损伤是基于神经发生的治疗干预的有希望的新途径。然而,使用体内模型进行筛查的技术困难阻碍了这种可能性,包括使用成人大脑中稀缺的神经干细胞以及人类和小鼠模型之间的差异或伦理限制。
    方法:这里,我们使用小鼠和人类干细胞模型的组合,对一种新的神经源性化合物进行全面的体外表征,关注脑源性神经营养因子(BDNF)通路。ENT-A011,一种甾体脱氢表雄酮衍生物,通过蛋白质印迹在NIH-3T3细胞中测试酪氨酸受体激酶B(TrkB)受体的激活,并通过增殖评估其神经源性和神经保护作用,小鼠原代成年海马神经干细胞的细胞死亡和淀粉样β(Aβ)毒性测定,小鼠胚胎皮质神经干细胞和神经祖细胞(NPC)从三个人诱导多能干细胞系从健康和AD供体分化。RNA-seq谱分析用于评估化合物是否通过与人NPC中的BDNF相同的基因网络起作用。
    结果:ENT-A011能够增加小鼠原代成年海马神经干细胞和胚胎皮质神经干细胞的增殖,在没有EGF/FGF的情况下,同时减少Aβ诱导的细胞死亡,有选择地通过TrkB激活。该化合物能够增加参与NSC维持的星形细胞基因标记,保护海马神经元免受Αβ毒性并防止Aβ治疗后的突触丢失。ENT-A011成功诱导增殖并防止人NPC中Aβ毒性后的细胞死亡,通过与BDNF共享的核心基因网络起作用,如通过RNA-seq所示。
    结论:我们的工作表征了一种新型BDNF模拟物,通过基于干细胞的筛选,在阿尔茨海默病中具有较好的药理特性和神经源性和神经保护作用。证明干细胞系统有望入围竞争候选人进行进一步测试。
    BACKGROUND: Neural stem cell (NSC) proliferation and differentiation in the mammalian brain decreases to minimal levels postnatally. Nevertheless, neurogenic niches persist in the adult cortex and hippocampus in rodents, primates and humans, with adult NSC differentiation sharing key regulatory mechanisms with development. Adult neurogenesis impairments have been linked to Alzheimer\'s disease (AD) pathology. Addressing these impairments by using neurotrophic factors is a promising new avenue for therapeutic intervention based on neurogenesis. However, this possibility has been hindered by technical difficulties of using in-vivo models to conduct screens, including working with scarce NSCs in the adult brain and differences between human and mouse models or ethical limitations.
    METHODS: Here, we use a combination of mouse and human stem cell models for comprehensive in-vitro characterization of a novel neurogenic compound, focusing on the brain-derived neurotrophic factor (BDNF) pathway. The ability of ENT-A011, a steroidal dehydroepiandrosterone derivative, to activate the tyrosine receptor kinase B (TrkB) receptor was tested through western blotting in NIH-3T3 cells and its neurogenic and neuroprotective action were assessed through proliferation, cell death and Amyloid-β (Aβ) toxicity assays in mouse primary adult hippocampal NSCs, mouse embryonic cortical NSCs and neural progenitor cells (NPCs) differentiated from three human induced pluripotent stem cell lines from healthy and AD donors. RNA-seq profiling was used to assess if the compound acts through the same gene network as BDNF in human NPCs.
    RESULTS: ENT-A011 was able to increase proliferation of mouse primary adult hippocampal NSCs and embryonic cortical NSCs, in the absence of EGF/FGF, while reducing Aβ-induced cell death, acting selectively through TrkB activation. The compound was able to increase astrocytic gene markers involved in NSC maintenance, protect hippocampal neurons from Αβ toxicity and prevent synapse loss after Aβ treatment. ENT-A011 successfully induces proliferation and prevents cell death after Aβ toxicity in human NPCs, acting through a core gene network shared with BDNF as shown through RNA-seq.
    CONCLUSIONS: Our work characterizes a novel BDNF mimetic with preferable pharmacological properties and neurogenic and neuroprotective actions in Alzheimer\'s disease via stem cell-based screening, demonstrating the promise of stem cell systems for short-listing competitive candidates for further testing.
    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:遗传性视网膜营养不良(IRD)是全球范围内无法治愈的失明的主要原因之一。IRD是由编码视网膜必需蛋白的基因突变引起的,导致光感受器退化和视觉功能丧失。由于缺乏对其病理生理学的重要部分的了解,IRD产生了巨大的全球财务负担,分子诊断,以及几乎没有非姑息治疗方案。用于IRD的患者来源的诱导多能干细胞(iPSC)似乎是解决这些问题的绝佳选择,作为IRD病理生理学深入研究和测试新治疗方法的特殊工具。
    方法:从8名与PROM1相关的IRD患者的队列中,我们确定了3名患者携带相同的变体(c.1354dupT),但表达三种不同的IRD表型:锥形和杆状营养不良(CORD),色素性视网膜炎(RP),和Stargardt病4型(STGD4)。这三个目标患者,每个人都有一个健康的亲戚,接受了全面的眼科检查,并通过临床外显子组测序(CES)扩展了他们的遗传小组研究。随后,产生非整合性患者来源的iPSC,并对其进行充分表征.使用CRISPR/Cas9进行c.1354dupT突变的校正,并且在患者来源的iPSC系中通过流式细胞术和蛋白质印迹确认PROM1基因的遗传恢复。
    结果:CES显示,2名具有c.1354dupT突变的目标患者在与补体系统或光感受器分化和过氧化物酶体生物发生障碍相关的基因中呈现单等位基因变异,分别。证实了患者来源的iPSC细胞系的多能性和功能性,目标突变的校正完全恢复了基因修复的患者来源的iPSC系中编码Prominin-1(CD133)的能力。
    结论:PROM1基因的c.1354dupT突变与IRD的三种不同的AR表型相关。这种多向效应可能与视网膜营养不良相关的其他基因中单等位基因变体的影响有关。然而,需要提供进一步的证据。未来的实验应该包括基因编辑的患者来源的iPSC,因为它有潜力作为疾病建模工具来阐明这一问题。
    BACKGROUND: Inherited retinal dystrophies (IRD) are one of the main causes of incurable blindness worldwide. IRD are caused by mutations in genes that encode essential proteins for the retina, leading to photoreceptor degeneration and loss of visual function. IRD generates an enormous global financial burden due to the lack of understanding of a significant part of its pathophysiology, molecular diagnosis, and the near absence of non-palliative treatment options. Patient-derived induced pluripotent stem cells (iPSC) for IRD seem to be an excellent option for addressing these questions, serving as exceptional tools for in-depth studies of IRD pathophysiology and testing new therapeutic approaches.
    METHODS: From a cohort of 8 patients with PROM1-related IRD, we identified 3 patients carrying the same variant (c.1354dupT) but expressing three different IRD phenotypes: Cone and rod dystrophy (CORD), Retinitis pigmentosa (RP), and Stargardt disease type 4 (STGD4). These three target patients, along with one healthy relative from each, underwent comprehensive ophthalmic examinations and their genetic panel study was expanded through clinical exome sequencing (CES). Subsequently, non-integrative patient-derived iPSC were generated and fully characterized. Correction of the c.1354dupT mutation was performed using CRISPR/Cas9, and the genetic restoration of the PROM1 gene was confirmed through flow cytometry and western blotting in the patient-derived iPSC lines.
    RESULTS: CES revealed that 2 target patients with the c.1354dupT mutation presented monoallelic variants in genes associated with the complement system or photoreceptor differentiation and peroxisome biogenesis disorders, respectively. The pluripotency and functionality of the patient-derived iPSC lines were confirmed, and the correction of the target mutation fully restored the capability of encoding Prominin-1 (CD133) in the genetically repaired patient-derived iPSC lines.
    CONCLUSIONS: The c.1354dupT mutation in the PROM1 gene is associated to three distinct AR phenotypes of IRD. This pleotropic effect might be related to the influence of monoallelic variants in other genes associated with retinal dystrophies. However, further evidence needs to be provided. Future experiments should include gene-edited patient-derived iPSC due to its potential as disease modelling tools to elucidate this matter in question.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号