Human induced pluripotent stem cells (hiPSCs)

人诱导多能干细胞 ( hiPSCs )
  • 文章类型: Journal Article
    人类诱导多能干细胞(hiPSC)衍生的运动神经元(MN)的产生提供了一种前所未有的方法来模拟运动障碍,例如肌张力障碍和肌萎缩性侧索硬化症。然而,在培养诱导的MNs时,实现存活构成了重大挑战,特别是当目标是达到成熟后期阶段时。利用hiPSC衍生的运动神经元和原代小鼠星形胶质细胞,我们组装了两种类型的共培养系统:神经元与星形胶质细胞的直接共培养,和使用物理分离神经元和星形胶质细胞的培养插入物的间接共培养。两个系统都显著增强神经元存活。与这两个系统相比,神经发育没有显著差异,成熟,在3周内存活,允许在成熟期准备神经元。使用间接共培养系统,我们在成熟后期从hiPSC获得了高纯度的MNs。hiPSC衍生的MN的转录组学研究显示,从早期未成熟阶段到晚期成熟阶段,基因表达存在典型的神经发育转换。与神经发育和突触发生相关的成熟基因在晚期阶段高度富集在MNs中。证明这些神经元实现了成熟。这项研究介绍了一种用于制备高纯度hiPSC衍生神经元的新工具,能够通过生化方法在疾病发作后期确定神经疾病的发病机理,这通常需要高纯度的神经元。这种进步在建模与年龄相关的神经变性中特别重要。意义陈述实现存活对长期神经细胞培养提出了重大挑战。利用hiPSC衍生的运动神经元和原代小鼠星形胶质细胞,我们建立了一个间接共培养系统,使用培养插入物物理分离神经元和星形胶质细胞,从而促进神经元成熟。转录组学研究揭示了从早期未成熟阶段到晚期成熟阶段的基因表达中典型的神经发育转换,表明用培养插入物制备的神经元的高质量和成熟。这项研究介绍了一种用于制备高纯度hiPSC衍生神经元的新工具,能够通过生化方法在疾病发作后期确定神经疾病的发病机理,这通常需要高纯度的神经元。这种进步在建模与年龄相关的神经变性中特别重要。
    Generation of human induced pluripotent stem cell (hiPSC)-derived motor neurons (MNs) offers an unprecedented approach to modeling movement disorders such as dystonia and amyotrophic lateral sclerosis. However, achieving survival poses a significant challenge when culturing induced MNs, especially when aiming to reach late maturation stages. Utilizing hiPSC-derived motor neurons and primary mouse astrocytes, we assembled two types of coculture systems: direct coculturing of neurons with astrocytes and indirect coculture using culture inserts that physically separate neurons and astrocytes. Both systems significantly enhance neuron survival. Compared with these two systems, no significant differences in neurodevelopment, maturation, and survival within 3 weeks, allowing to prepare neurons at maturation stages. Using the indirect coculture system, we obtained highly pure MNs at the late mature stage from hiPSCs. Transcriptomic studies of hiPSC-derived MNs showed a typical neurodevelopmental switch in gene expression from the early immature stage to late maturation stages. Mature genes associated with neurodevelopment and synaptogenesis are highly enriched in MNs at late stages, demonstrating that these neurons achieve maturation. This study introduces a novel tool for the preparation of highly pure hiPSC-derived neurons, enabling the determination of neurological disease pathogenesis in neurons at late disease onset stages through biochemical approaches, which typically necessitate highly pure neurons. This advancement is particularly significant in modeling age-related neurodegeneration.
    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:X连锁幼年视网膜裂(XLRS)是一种由RS1基因突变引起的遗传性疾病,导致视网膜分裂和视力障碍.RS1相关视网膜变性的机制尚不完全清楚。此外,XLRS动物模型在XLRS研究中存在局限性。这里,我们使用人类诱导多能干细胞(hiPSC)衍生的视网膜类器官(ROs)来研究XLRS的疾病机制和潜在治疗方法.
    方法:从两个RS1突变体(E72K)XLRS患者的外周血单核细胞重编程的hiPSCs分化为ROs。随后,我们探讨RS1突变是否会影响RO发育,并探讨RS1基因增强治疗的有效性.
    结果:来自RS1(E72K)突变hiPSCs的RO在光感受器中表现出发育延迟,视网膜裂素(RS1)缺乏症,与对照RO相比,自发活动改变。此外,发育延迟与杆特异性前体标记(NRL)和光感受器特异性标记(RCVRN)表达降低相关.腺相关病毒(AAV)介导的RS1基因增强在光感受器未成熟阶段挽救了具有RS1(E72K)突变的RO中的杆状光感受器发育延迟。
    结论:RS1(E72K)突变导致ROs的光感受器发育延迟,RS1基因增强治疗可以部分挽救。
    BACKGROUND: X-linked juvenile retinoschisis (XLRS) is an inherited disease caused by RS1 gene mutation, which leads to retinal splitting and visual impairment. The mechanism of RS1-associated retinal degeneration is not fully understood. Besides, animal models of XLRS have limitations in the study of XLRS. Here, we used human induced pluripotent stem cell (hiPSC)-derived retinal organoids (ROs) to investigate the disease mechanisms and potential treatments for XLRS.
    METHODS: hiPSCs reprogrammed from peripheral blood mononuclear cells of two RS1 mutant (E72K) XLRS patients were differentiated into ROs. Subsequently, we explored whether RS1 mutation could affect RO development and explore the effectiveness of RS1 gene augmentation therapy.
    RESULTS: ROs derived from RS1 (E72K) mutation hiPSCs exhibited a developmental delay in the photoreceptor, retinoschisin (RS1) deficiency, and altered spontaneous activity compared with control ROs. Furthermore, the delays in development were associated with decreased expression of rod-specific precursor markers (NRL) and photoreceptor-specific markers (RCVRN). Adeno-associated virus (AAV)-mediated gene augmentation with RS1 at the photoreceptor immature stage rescued the rod photoreceptor developmental delay in ROs with the RS1 (E72K) mutation.
    CONCLUSIONS: The RS1 (E72K) mutation results in the photoreceptor development delay in ROs and can be partially rescued by the RS1 gene augmentation therapy.
    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
    脊髓类器官模拟疾病状态在脊髓相关疾病研究中具有重要价值,从而促进新疗法的开发。然而,脊髓结构和生理功能的复杂性,随着人类诱导成分的缺乏,在人脊髓类器官的体外构建中提出了挑战。这里,我们介绍了一种新型人脱细胞胎盘来源的细胞外基质水凝胶(DPECMH),结合新的诱导方案,成功构建人脊髓类器官。人胎盘来源的脱细胞细胞外基质(dECM),通过苏木精和伊红染色验证,DNA定量,免疫荧光染色,保留了必要的ECM成分,如弹性蛋白,纤连蛋白,I型胶原蛋白,层粘连蛋白,等等。由人胎盘dECM制成的温度敏感水凝胶显示出良好的生物相容性,并促进人诱导多能干细胞(hiPSCs)衍生的脊髓类器官向神经元的分化。与Matrigel相比,它显示出增强的层状标志物表达,与Matrigel相比,它显示出更高的层状标志物表达。加速脊髓类器官的成熟过程,并证明其作为类器官培养基质的潜力。DPECMH有潜力取代Matrigel成为人类脊髓类器官的标准添加剂,从而推进脊髓类器官培养方案的发展及其在脊髓相关疾病体外建模中的应用。
    Spinal cord organoids are of significant value in the research of spinal cord-related diseases by simulating disease states, thereby facilitating the development of novel therapies. However, the complexity of spinal cord structure and physiological functions, along with the lack of human-derived inducing components, presents challenges in the in vitro construction of human spinal cord organoids. Here, we introduce a novel human decellularized placenta-derived extracellular matrix hydrogel (DPECMH) and, combined with a new induction protocol, successfully construct human spinal cord organoids. The human placenta-sourced decellularized extracellular matrix (dECM), verified through hematoxylin and eosin staining, DNA quantification, and immunofluorescence staining, retained essential ECM components such as elastin, fibronectin, type I collagen, laminin, and so forth. The temperature-sensitive hydrogel made from human placenta dECM demonstrated good biocompatibility and promoted the differentiation of human induced pluripotent stem cell (hiPSCs)-derived spinal cord organoids into neurons. It displayed enhanced expression of laminar markers in comparison to Matrigel and showed higher expression of laminar markers compared to Matrigel, accelerating the maturation process of spinal cord organoids and demonstrating its potential as an organoid culture substrate. DPECMH has the potential to replace Matrigel as the standard additive for human spinal cord organoids, thus advancing the development of spinal cord organoid culture protocols and their application in the in vitro modeling of spinal cord-related 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: Review
    神经系统肿瘤,特别是脑肿瘤,代表儿童中最常见的肿瘤和成人中最致命的肿瘤之一。尽管经过几十年的研究,这些癌症几乎没有有效的治疗方法。尽管人类神经系统肿瘤细胞和基因工程小鼠模型已成为药物发现和临床前测试的绝佳平台,它们在准确概括自发发生的人类肿瘤病理生物学的重要方面方面存在局限性。出于这个原因,注意力已经转向涉及人类胚胎或诱导多能干细胞的人类干细胞工程的部署,其中可以引入与神经系统癌症相关的遗传改变。这些干细胞可用于创建自组装的三维大脑器官,这些器官保留了发育中的人脑的关键特征。此外,干细胞工程系适合异种移植到小鼠中作为研究肿瘤细胞起源的平台,发现癌症进化轨迹并确定治疗漏洞。在这篇文章中,我们回顾了神经系统肿瘤的人类干细胞模型的现状,讨论它们的优点和缺点,并为未来的研究提供共识建议。
    Nervous system tumors, particularly brain tumors, represent the most common tumors in children and one of the most lethal tumors in adults. Despite decades of research, there are few effective therapies for these cancers. Although human nervous system tumor cells and genetically engineered mouse models have served as excellent platforms for drug discovery and preclinical testing, they have limitations with respect to accurately recapitulating important aspects of the pathobiology of spontaneously arising human tumors. For this reason, attention has turned to the deployment of human stem cell engineering involving human embryonic or induced pluripotent stem cells, in which genetic alterations associated with nervous system cancers can be introduced. These stem cells can be used to create self-assembling three-dimensional cerebral organoids that preserve key features of the developing human brain. Moreover, stem cell-engineered lines are amenable to xenotransplantation into mice as a platform to investigate the tumor cell of origin, discover cancer evolutionary trajectories and identify therapeutic vulnerabilities. In this article, we review the current state of human stem cell models of nervous system tumors, discuss their advantages and disadvantages, and provide consensus recommendations for future research.
    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:基于原发性小鼠的血脑屏障(BBB)模型,牛,和猪脑毛细血管内皮细胞培养一直被认为是具有适当特性的可靠模型,以检查小分子的功能运输。然而,物种差异有时会使将这些模型的结果转换为人类环境变得复杂。在过去的十年里,已经从干细胞来源产生了脑毛细血管内皮样细胞(BCECs),以体外模拟人BBB。本研究的目的是使用来自hIPSC系SBAD0201的人诱导多能干细胞(hiPSC)衍生的BCECs来建立和表征人BBB模型。
    方法:使用转录组学评估模型,蛋白质组学,免疫细胞化学,跨内皮电阻(TEER)测量,and,最后,用于评估选定转运蛋白和受体(GLUT-1,LAT-1,P-gp和LRP-1)的功能的转运测定法。
    结果:所得的BBB模型显示出平均TEER为5474±167Ω·cm2,并且在紧密连接区形成了claudin-5,ZO-1和occludin表达的细胞单层。细胞单层表达典型的BBB标记VE-cadherin,VWF,PECAM-1转录组学和定量靶向绝对蛋白质组学分析显示,溶质载体(SLC)转运蛋白的丰度很高,而外排转运蛋白的表达相对较低。使用GLUT-1,LAT-1和LRP-1底物和抑制剂的转运测定证实了这些转运蛋白和受体在模型中的功能活性。转运试验表明,P-gp在模型中没有功能性表达,尽管抗体染色显示P-gp位于腔膜。
    结论:结论:新的SBAD0201衍生的BBB模型形成紧密的单层,被证明可用于研究GLUT-1,LAT-1和LRP-1介导的跨BBB转运.然而,该模型不表达功能性P-gp,因此不适合进行药物外排P-gp相关研究。
    BACKGROUND: Blood-brain barrier (BBB) models based on primary murine, bovine, and porcine brain capillary endothelial cell cultures have long been regarded as robust models with appropriate properties to examine the functional transport of small molecules. However, species differences sometimes complicate translating results from these models to human settings. During the last decade, brain capillary endothelial-like cells (BCECs) have been generated from stem cell sources to model the human BBB in vitro. The aim of the present study was to establish and characterize a human BBB model using human induced pluripotent stem cell (hiPSC)-derived BCECs from the hIPSC line SBAD0201.
    METHODS: The model was evaluated using transcriptomics, proteomics, immunocytochemistry, transendothelial electrical resistance (TEER) measurements, and, finally, transport assays to assess the functionality of selected transporters and receptor (GLUT-1, LAT-1, P-gp and LRP-1).
    RESULTS: The resulting BBB model displayed an average TEER of 5474 ± 167 Ω·cm2 and cell monolayer formation with claudin-5, ZO-1, and occludin expression in the tight junction zones. The cell monolayers expressed the typical BBB markers VE-cadherin, VWF, and PECAM-1. Transcriptomics and quantitative targeted absolute proteomics analyses revealed that solute carrier (SLC) transporters were found in high abundance, while the expression of efflux transporters was relatively low. Transport assays using GLUT-1, LAT-1, and LRP-1 substrates and inhibitors confirmed the functional activities of these transporters and receptors in the model. A transport assay suggested that P-gp was not functionally expressed in the model, albeit antibody staining revealed that P-gp was localized at the luminal membrane.
    CONCLUSIONS: In conclusion, the novel SBAD0201-derived BBB model formed tight monolayers and was proven useful for studies investigating GLUT-1, LAT-1, and LRP-1 mediated transport across the BBB. However, the model did not express functional P-gp and thus is not suitable for the performance of drug efflux P-gp reletated studies.
    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
    脑类器官是自我组织的,来自多能干细胞的三维(3D)聚集体,其细胞类型和细胞结构类似于发育中的人脑。随着这种体外模型的引入,目前对人脑发育过程和神经系统疾病的理解已经显着提高。脑类器官充当二维(2D)培养物和体内模型之间的翻译链接,该模型模仿早期和晚期阶段的神经管形成以及全脑区域化的神经上皮分化。此外,区域特异性脑类器官的产生使得研究获得性和遗传性脑疾病的致病和病因学方面以及药物发现和药物毒性测试成为可能。在这篇评论文章中,我们首先概述了用于生成脑类器官的现有方法和平台及其局限性,然后讨论了脑类器官技术的最新进展。此外,我们讨论了如何使用脑类器官来模拟神经发育和神经退行性疾病的各个方面,包括自闭症谱系障碍(ASD),Rett综合征,寨卡病毒相关的小头畸形,阿尔茨海默病(AD),帕金森病(PD),和亨廷顿病(HD)。
    Brain organoids are self-organized, three-dimensional (3D) aggregates derived from pluripotent stem cells that have cell types and cellular architectures resembling those of the developing human brain. The current understanding of human brain developmental processes and neurological disorders has advanced significantly with the introduction of this in vitro model. Brain organoids serve as a translational link between two-dimensional (2D) cultures and in vivo models which imitate the neural tube formation at the early and late stages and the differentiation of neuroepithelium with whole-brain regionalization. In addition, the generation of region-specific brain organoids made it possible to investigate the pathogenic and etiological aspects of acquired and inherited brain disease along with drug discovery and drug toxicity testing. In this review article, we first summarize an overview of the existing methods and platforms used for generating brain organoids and their limitations and then discuss the recent advancement in brain organoid technology. In addition, we discuss how brain organoids have been used to model aspects of neurodevelopmental and neurodegenerative diseases, including autism spectrum disorder (ASD), Rett syndrome, Zika virus-related microcephaly, Alzheimer\'s disease (AD), Parkinson\'s disease (PD), and Huntington\'s disease (HD).
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Preprint
    神经元兴奋过度是癫痫发作的标志。最近在癫痫发作的啮齿动物模型中显示小胶质细胞,大脑的固有免疫细胞,可以对神经元兴奋性做出反应并调节。然而,人类小胶质细胞如何与人类神经元相互作用以调节在人类患者中发现的由癫痫引起的基因突变介导的兴奋过度仍然未知.SCN2A遗传基因座负责编码电压门控钠通道Nav1.2,被认为是单基因癫痫的主要贡献者之一。以前,我们证明,在一名男性供者的hiPSC来源皮质神经元模型中,在癫痫患者中发现的Nav1.2-L1342P重复突变导致兴奋过度.虽然小胶质细胞在大脑中发挥重要作用,这些细胞源自不同的谱系(卵黄囊),因此不天然存在于hiPSC衍生的神经元培养物中。为了研究小胶质细胞如何对患病的神经元做出反应并影响神经元的兴奋性,我们建立了包含hiPSC来源的神经元和小胶质细胞的共培养模型.我们发现,当与携带Nav1.2-L1342P突变的神经元共培养时,小胶质细胞的形态改变,分支长度增加,钙信号增强。此外,小胶质细胞的存在显著降低了携带突变的神经元的动作电位放电。有趣的是,我们进一步证明,在存在小胶质细胞的情况下,携带癫痫相关突变的神经元中钠通道的电流密度降低.一起来看,我们的工作揭示了人类iPSCs来源的小胶质细胞在感知和抑制由人类神经元中存在的癫痫引起的突变介导的过度兴奋中的关键作用,强调神经元-小胶质细胞相互作用在人类病理生理学中的重要性。
    小鼠模型中的癫痫研究强调了小胶质细胞在调节神经元活动中的作用,特别是促进或抑制癫痫发作。然而,在理解人类小胶质细胞对携带癫痫相关致病突变的内在过度兴奋神经元的影响方面仍然存在差距。这项研究通过研究人类小胶质细胞及其对神经元功能的影响来解决这一差距。我们的发现表明,在存在携带癫痫相关SCN2A突变的神经元的情况下,小胶质细胞表现出动态的形态学改变和钙波动。此外,小胶质细胞抑制了患病过度兴奋神经元的兴奋性,暗示潜在的有益作用。这项研究强调了小胶质细胞在调节异常神经元活动中的作用,提供与兴奋过度相关的神经系统疾病的治疗策略的见解。
    Neuronal hyperexcitability is a hallmark of seizures. It has been recently shown in rodent models of seizures that microglia, the brain\'s resident immune cells, can respond to and modulate neuronal excitability. However, how human microglia interacts with human neurons to regulate hyperexcitability mediated by epilepsy-causing genetic mutation found in human patients remains unknown. The SCN2A genetic locus is responsible for encoding the voltage-gated sodium channel Nav1.2, recognized as one of the leading contributors to monogenic epilepsies. Previously, we demonstrated that the recurring Nav1.2-L1342P mutation identified in patients with epilepsy leads to hyperexcitability in a hiPSC-derived cortical neuron model from a male donor. While microglia play an important role in the brain, these cells originate from a different lineage (yolk sac) and thus are not naturally present in hiPSCs-derived neuronal culture. To study how microglia respond to diseased neurons and influence neuronal excitability, we established a co-culture model comprising hiPSC-derived neurons and microglia. We found that microglia display altered morphology with increased branch length and enhanced calcium signal when co-cultured with neurons carrying the Nav1.2-L1342P mutation. Moreover, the presence of microglia significantly lowers the action potential firing of neurons carrying the mutation. Interestingly, we further demonstrated that the current density of sodium channels in neurons carrying the epilepsy-associated mutation was reduced in the presence of microglia. Taken together, our work reveals a critical role of human iPSCs-derived microglia in sensing and dampening hyperexcitability mediated by an epilepsy-causing mutation present in human neurons, highlighting the importance of neuron-microglia interactions in human pathophysiology.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: English Abstract
    人诱导多能干细胞(hiPSCs)在再生医学中具有广阔的应用前景。然而,多能干细胞(PSC)可能形成癌变组织的团块,这是PSC治疗中的主要安全性问题。雷帕霉素是安全且广泛使用的免疫抑制药物,其通过FKBP12和FRB片段的异二聚化起作用。这里,我们的目的是在安全港AAVS1位点插入雷帕霉素诱导的caspase9(riC9)基因,以保护药物诱导的hiPSCs治疗.含有EF1α启动子的供体载体,构建FRB-FKBP-Caspase9(CARD结构域)融合蛋白和嘌呤霉素抗性基因,并与sgRNA/Cas9载体共转染到hiPSC中。在用嘌呤霉素筛查一到两周后,收集单克隆进行基因型和表型分析.最后,雷帕霉素用于诱导caspase9的同源二聚化,以激活工程细胞的凋亡。转染hiPSCs后,再进行嘌呤霉素筛选,收集五个细胞克隆。基因组扩增和测序显示供体DNA已在内源AAVS1位点被精确敲除。工程化的hiPSC显示出正常的多能性和增殖能力。雷帕霉素诱导半胱天冬酶9活化,导致所有工程hiPSCs及其分化细胞对药物敏感性不同的凋亡。总之,我们通过caspase9的同源二聚化诱导细胞凋亡,产生了雷帕霉素可控的hiPSCs存活。为保证hiPSCs治疗的安全性提供了新的策略。
    Human induced pluripotent stem cells (hiPSCs) are promising in regenerative medicine. However, the pluripotent stem cells (PSCs) may form clumps of cancerous tissue, which is a major safety concern in PSCs therapies. Rapamycin is a safe and widely used immunosuppressive pharmaceutical that acts through heterodimerization of the FKBP12 and FRB fragment. Here, we aimed to insert a rapamycin inducible caspase 9 (riC9) gene in a safe harbor AAVS1 site to safeguard hiPSCs therapy by drug induced homodimerization. The donor vector containing an EF1α promoter, a FRB-FKBP-Caspase 9 (CARD domain) fusion protein and a puromycin resistant gene was constructed and co-transfected with sgRNA/Cas9 vector into hiPSCs. After one to two weeks screening with puromycin, single clones were collected for genotype and phenotype analysis. Finally, rapamycin was used to induce the homodimerization of caspase 9 to activate the apoptosis of the engineered cells. After transfection of hiPSCs followed by puromycin screening, five cell clones were collected. Genome amplification and sequencing showed that the donor DNA has been precisely knocked out at the endogenous AAVS1 site. The engineered hiPSCs showed normal pluripotency and proliferative capacity. Rapamycin induced caspase 9 activation, which led to the apoptosis of all engineered hiPSCs and its differentiated cells with different sensitivity to drugs. In conclusion, we generated a rapamycin-controllable hiPSCs survival by homodimerization of caspase 9 to turn on cell apoptosis. It provides a new strategy to guarantee the safety of the hiPSCs therapy.
    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
    唐氏综合征(DS)是由21号染色体(HSA21)三体性引起的,是迄今为止最常见的染色体疾病,伴有神经发育障碍和先天性心脏病。这里,我们从两名DS患者中产生了两个诱导多能干细胞(iPSC)系.这两条线表现出正常的形态,21三体核型,多能性和分化能力为三个胚层的衍生物。患者特异性iPSC细胞系是研究中的宝贵资源,用于模拟DS相关的细胞和分子病理学,并测试DS的可能治疗策略。
    Down syndrome (DS) is caused by trisomy of Homo sapiens chromosome 21 (HSA21) and is by far the most common chromosomal disorder accompanied by neurodevelopmental disorders and congenital heart disease. Here, we generated two induced pluripotent stem cell (iPSC) lines from two patients with DS. These two lines exhibited normal morphology, trisomy 21 karyotype, pluripotency and differentiation capability into derivatives of three germ layers. The patient-specific iPSC lines arean invaluable resource in research to model DS-related cellular and molecular pathologies and test possible therapeutic strategies for DS.
    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
    遗传性心肌病是一组遗传性疾病,其中心肌结构和功能受损。这些病变中的许多是罕见的,并且存在异质表型,因此,需要个性化模型来完全揭示其病理机制并开发有价值的治疗策略。心肌细胞和成纤维细胞,从患者特异性人诱导多能干细胞分化,代表了在遗传性心肌病背景下研究最多的人类心脏细胞模型。虽然内皮功能障碍已被认为是一种可能的发病机制,人类诱导多能干细胞来源的内皮细胞研究较少,尽管它们构成了合适的模型,以专门剖析功能失调的内皮在这些病理的发展和进展中的作用。在这次审查中,我们总结了人类诱导多能干细胞来源的内皮细胞用于研究基于遗传的心肌病的内皮功能障碍的主要研究,以突出可用于治疗干预的新潜在靶标。我们讨论鼓励这个方向研究的新观点。
    Genetic cardiomyopathies are a group of inherited disorders in which myocardial structure and function are damaged. Many of these pathologies are rare and present with heterogenous phenotypes, thus personalized models are required to completely uncover their pathological mechanisms and develop valuable therapeutic strategies. Both cardiomyocytes and fibroblasts, differentiated from patient-specific human induced pluripotent stem cells, represent the most studied human cardiac cell models in the context of genetic cardiomyopathies. While endothelial dysfunction has been recognized as a possible pathogenetic mechanism, human induced pluripotent stem cell-derived endothelial cells are less studied, despite they constitute a suitable model to specifically dissect the role of the dysfunctional endothelium in the development and progression of these pathologies. In this review, we summarize the main studies in which human induced pluripotent stem cell-derived endothelial cells are used to investigate endothelial dysfunction in genetic-based cardiomyopathies to highlight new potential targets exploitable for therapeutic intervention, and we discuss novel perspectives that encourage research in this direction.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号