induced pluripotent stem cells

诱导多能干细胞
  • 文章类型: Journal Article
    中枢神经系统(CNS)通过交感神经和副交感神经在调节心率和心肌收缩力方面发挥作用,心脏可以通过反馈信号影响中枢神经系统的功能平衡。虽然心脑疾病往往共存,相互影响,心脏疾病和脑部疾病之间的潜在联系仍不清楚,因为缺乏可靠的模型。诱导多能干细胞(iPSCs),可以分化为多种功能细胞类型,干细胞生物学和再生医学可能提供工具来阐明这些关系的机制,并促进有效治疗药物的筛选。因为钙离子在调节心血管和神经系统中起着至关重要的作用,这篇综述探讨了最近的iPSC疾病模型如何揭示细胞内钙的失调可能是心脑疾病之间关系的共同病理因素.
    The central nervous system (CNS) plays a role in regulating heart rate and myocardial contractility through sympathetic and parasympathetic nerves, and the heart can impact the functional equilibrium of the CNS through feedback signals. Although heart and brain diseases often coexist and mutually influence each other, the potential links between heart and brain diseases remain unclear due to a lack of reliable models of these relationships. Induced pluripotent stem cells (iPSCs), which can differentiate into multiple functional cell types, stem cell biology and regenerative medicine may offer tools to clarify the mechanisms of these relationships and facilitate screening of effective therapeutic agents. Because calcium ions play essential roles in regulating both the cardiovascular and nervous systems, this review addresses how recent iPSC disease models reveal how dysregulation of intracellular calcium might be a common pathological factor underlying the relationships between heart and brain 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
    非酒精性脂肪性肝病(NAFLD)的发病率,或代谢功能障碍相关的脂肪肝疾病(MAFLD),在成人和儿童中正在增加。不幸的是,有效的药物治疗仍然不可用。含patatin类磷脂酶结构域的蛋白质(PNPLA3I148M)中的单核苷酸多态性(SNP)在疾病进展的所有阶段与疾病具有最重要的遗传关联。确定PNPLA3诱导的NAFLD的潜在治疗方法的障碍是缺乏概括PNPLA3I148M介导的脂质积累开始的人细胞平台。从PNPLA3-/-和PNPLA3I148M/M诱导的多能干细胞(iPSC)产生肝细胞样细胞。通过用BODIPY493/503染色测量脂质水平,发现在PNPLA3变体iPSC衍生的肝细胞中增加。小分子筛选鉴定了靶向Src/PI3K/Akt信号传导并且可以根除这些细胞中的脂质积累的多种化合物。我们发现,目前在临床试验中针对相同途径的癌症治疗药物也减少了PNPLA3变异细胞中的脂质积累。
    The incidence of nonalcoholic fatty liver disease (NAFLD), or metabolic dysfunction-associated fatty liver disease (MAFLD), is increasing in adults and children. Unfortunately, effective pharmacological treatments remain unavailable. Single nucleotide polymorphisms (SNPs) in the patatin-like phospholipase domain-containing protein (PNPLA3 I148M) have the most significant genetic association with the disease at all stages of its progression. A roadblock to identifying potential treatments for PNPLA3-induced NAFLD is the lack of a human cell platform that recapitulates the PNPLA3 I148M-mediated onset of lipid accumulation. Hepatocyte-like cells were generated from PNPLA3-/- and PNPLA3I148M/M-induced pluripotent stem cells (iPSCs). Lipid levels were measured by staining with BODIPY 493/503 and were found to increase in PNPLA3 variant iPSC-derived hepatocytes. A small-molecule screen identified multiple compounds that target Src/PI3K/Akt signaling and could eradicate lipid accumulation in these cells. We found that drugs currently in clinical trials for cancer treatment that target the same pathways also reduced lipid accumulation in PNPLA3 variant cells.
    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
    常染色体显性视神经萎缩(ADOA)是一种罕见的进行性疾病,主要由OPA1突变引起,OPA1是一种编码线粒体蛋白的核基因,在线粒体动力学中起着至关重要的作用。细胞存活,氧化磷酸化,和mtDNA维护。ADOA的特征在于视网膜神经节细胞(RGCs)的变性。这会导致视力丧失,这在许多情况下可能导致法律失明。如今,ADOA没有有效的治疗方法。在这篇文章中,我们使用iPSC技术和基因组编辑工具CRISPR/Cas9从先前生成的具有致病性变异NM_015560.3:c.1861C>T的ADOA加患者的iPSC细胞系中建立了ADOA的等基因人类RGC模型(p.Gln621Ter)在OPA1中的杂合。为此,已经采用了基于向iPSC培养基补充几种小分子和试图模拟胚胎发育的定义因子的方案。随后,创建的模型经过验证,确认基因组间通信缺陷的存在,线粒体呼吸受损,以及细胞凋亡和ROS产生的增加。最后,我们建议通过qPCR分析OPA1表达作为一种简单的读出方法,用于使用创建的RGC模型进行未来的药物筛选研究。总之,该模型为进一步研究ADOAplus的潜在病理生理机制以及测试具有潜在药理作用的化合物提供了有用的平台.
    Autosomal dominant optic atrophy (ADOA) is a rare progressive disease mainly caused by mutations in OPA1, a nuclear gene encoding for a mitochondrial protein that plays an essential role in mitochondrial dynamics, cell survival, oxidative phosphorylation, and mtDNA maintenance. ADOA is characterized by the degeneration of retinal ganglion cells (RGCs). This causes visual loss, which can lead to legal blindness in many cases. Nowadays, there is no effective treatment for ADOA. In this article, we have established an isogenic human RGC model for ADOA using iPSC technology and the genome editing tool CRISPR/Cas9 from a previously generated iPSC line of an ADOA plus patient harboring the pathogenic variant NM_015560.3: c.1861C>T (p.Gln621Ter) in heterozygosis in OPA1. To this end, a protocol based on supplementing the iPSC culture media with several small molecules and defined factors trying to mimic embryonic development has been employed. Subsequently, the created model was validated, confirming the presence of a defect of intergenomic communication, impaired mitochondrial respiration, and an increase in apoptosis and ROS generation. Finally, we propose the analysis of OPA1 expression by qPCR as an easy read-out method to carry out future drug screening studies using the created RGC model. In summary, this model provides a useful platform for further investigation of the underlying pathophysiological mechanisms of ADOA plus and for testing compounds with potential pharmacological action.
    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
    鉴于神经元群体在各种生物过程中的关键作用,评估他们的集体输出对于理解神经系统的复杂功能至关重要。基于我们先前开发的螺旋扫描机制,用于从单细胞快速获取拉曼光谱,并结合机器学习用于无标签评估细胞状态,我们调查了PaintRamanExpress光谱系统(PRESS)是否可以评估神经元活动。我们通过检查谷氨酸能神经元作为单个神经元和自主神经神经节作为源自人诱导的多能干细胞的神经元群体的化学反应来检验这一假设。PRESS在几秒钟内成功获得了单个神经元和神经节的拉曼光谱,实现足够详细分析的信噪比。为了评估诱导神经元和神经节的配体反应性,拉曼光谱进行了主成分和偏最小二乘判别分析。PRESS检测到神经元对谷氨酸和尼古丁的反应,在没有钙的情况下不存在。此外,PRESS诱导的剂量依赖性神经元活动变化。这些发现强调了PRESS评估个体神经元活动和阐明神经元种群动态和药理学反应的能力。预示着药物发现和再生医学发展的新机会。
    Given the pivotal role of neuronal populations in various biological processes, assessing their collective output is crucial for understanding the nervous system\'s complex functions. Building on our prior development of a spiral scanning mechanism for the rapid acquisition of Raman spectra from single cells and incorporating machine learning for label-free evaluation of cell states, we investigated whether the Paint Raman Express Spectroscopy System (PRESS) can assess neuronal activities. We tested this hypothesis by examining the chemical responses of glutamatergic neurons as individual neurons and autonomic neuron ganglia as neuronal populations derived from human-induced pluripotent stem cells. The PRESS successfully acquired Raman spectra from both individual neurons and ganglia within a few seconds, achieving a signal-to-noise ratio sufficient for detailed analysis. To evaluate the ligand responsiveness of the induced neurons and ganglia, the Raman spectra were subjected to principal component and partial least squares discriminant analyses. The PRESS detected neuronal activity in response to glutamate and nicotine, which were absent in the absence of calcium. Additionally, the PRESS induced dose-dependent neuronal activity changes. These findings underscore the capability of the PRESS to assess individual neuronal activity and elucidate neuronal population dynamics and pharmacological responses, heralding new opportunities for drug discovery and regenerative medicine advancement.
    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
    脉络膜,位于视网膜和巩膜之间的血管化组织,在维持眼稳态中起着至关重要的作用。尽管意义重大,脉络膜异常的研究和有效体外模型的建立一直受到限制。在这项研究中,我们通过人诱导多能干细胞(hiPSC)来源的内皮细胞(ECs)和小鼠脉络膜成纤维细胞(msCFs)与hiPSC来源的视网膜色素上皮(RPE)细胞的共培养,建立了体外脉络膜模型。这个模型,包括EC在内,CFs,和RPE细胞,表现出与体内脉络膜血管的相似性,通过细胞外基质标志物和血管相关标志物的免疫组织化学证实,以及脉络膜血管生成发芽测定分析。我们的体外模型在评估由靶向血管调节的药物诱导的血管变化方面的有效性得到了证明。我们的模型为了解脉络膜发育和脉络膜血管疾病进展的病理机制提供了有价值的工具。
    The choroid, a vascularized tissue situated between the retina and the sclera, plays a crucial role in maintaining ocular homeostasis. Despite its significance, research on choroidal abnormalities and the establishment of effective in vitro models have been limited. In this study, we developed an in vitro choroid model through the co-culture of human induced pluripotent stem cells (hiPSC)-derived endothelial cells (ECs) and mouse choroidal fibroblasts (msCFs) with hiPSC-derived retinal pigment epithelial (RPE) cells via a permeable membrane. This model, inclusive of ECs, CFs, and RPE cells, exhibited similarities with in vivo choroidal vessels, as confirmed through immunohistochemistry of extracellular matrix markers and vascular-related markers, as well as choroid angiogenesis sprouting assay analysis. The effectiveness of our in vitro model was demonstrated in assessing vascular changes induced by drugs targeting vasoregulation. Our model offers a valuable tool for gaining insights into the pathological mechanisms underlying choroid development and the progression of choroidal vascular 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
    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
    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
    生物电信号具有在胚胎发生和组织水平再生期间强有力地控制和操纵模式的能力。内生局部和全局电场作为空间“预模式”,控制细胞命运和组织尺度解剖边界;然而,促进这些稳健的多尺度结果的机制特征不佳。计算模型解决了预测体外图案化行为的需求,并进一步阐明了细胞生物电信号传导成分在图案化结果中的作用。这里,我们修改了先前设计的图像模式识别算法,以区分不同细胞培养条件下模拟的非兴奋生物电模式的独特空间特征。该算法应用于跨培养的人iPSC集落的膜电位(Vmem)的模拟模式和实验显微镜图像之间的比较。此外,我们将预测扩展到一种新的共培养条件,其中模拟了具有不同离子通量的细胞亚群;定义的空间特征在体外用基因修饰的菌落进行了概括。这些结果共同为多细胞系统中出现的多尺度空间特征建模策略提供了信息。表征非兴奋细胞中膜电位异质性的分子贡献,并实现下游工程生物电组织设计。
    Bioelectric signals possess the ability to robustly control and manipulate patterning during embryogenesis and tissue-level regeneration. Endogenous local and global electric fields function as a spatial \'pre-pattern\', controlling cell fates and tissue-scale anatomical boundaries; however, the mechanisms facilitating these robust multiscale outcomes are poorly characterized. Computational modeling addresses the need to predict in vitro patterning behavior and further elucidate the roles of cellular bioelectric signaling components in patterning outcomes. Here, we modified a previously designed image pattern recognition algorithm to distinguish unique spatial features of simulated non-excitable bioelectric patterns under distinct cell culture conditions. This algorithm was applied to comparisons between simulated patterns and experimental microscopy images of membrane potential (Vmem) across cultured human iPSC colonies. Furthermore, we extended the prediction to a novel co-culture condition in which cell sub-populations possessing different ionic fluxes were simulated; the defining spatial features were recapitulated in vitro with genetically modified colonies. These results collectively inform strategies for modeling multiscale spatial characteristics that emerge in multicellular systems, characterizing the molecular contributions to heterogeneity of membrane potential in non-excitable cells, and enabling downstream engineered bioelectrical tissue design.
    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
    我们介绍了营养不良性大疱性表皮松解症细胞疗法(DEBCT),一种可扩展的平台,可产生自体器官型iPS细胞衍生的诱导皮肤复合(iSC)移植物,用于最终治疗。临床级制造将CRISPR介导的基因校正与重编程整合到一个步骤中,加速从患者中衍生COL7A1编辑的iPS细胞。分化为表皮,真皮和黑素细胞祖细胞随后是CD49f富集,将成熟异质性降至最低。来自4名具有不同突变的患者的iSC的小鼠异种移植在1个月时显示疾病修饰活性。下一代测序,生物分布和致瘤性测定在1-9个月时建立了良好的安全性。单细胞转录组学显示,iSC由主要的皮肤细胞谱系组成,并包括角质形成细胞的突出的全克隆干细胞样特征,以及最近描述的成纤维细胞的Gibbin依赖性特征。后者与iSC的增强的可嫁接性相关。总之,DEBCT克服了制造和安全障碍,并建立了可重复的,安全,与cGMP相容的治疗方法可以治愈DEB患者的病变。
    We present Dystrophic Epidermolysis Bullosa Cell Therapy (DEBCT), a scalable platform producing autologous organotypic iPS cell-derived induced skin composite (iSC) grafts for definitive treatment. Clinical-grade manufacturing integrates CRISPR-mediated genetic correction with reprogramming into one step, accelerating derivation of COL7A1-edited iPS cells from patients. Differentiation into epidermal, dermal and melanocyte progenitors is followed by CD49f-enrichment, minimizing maturation heterogeneity. Mouse xenografting of iSCs from four patients with different mutations demonstrates disease modifying activity at 1 month. Next-generation sequencing, biodistribution and tumorigenicity assays establish a favorable safety profile at 1-9 months. Single cell transcriptomics reveals that iSCs are composed of the major skin cell lineages and include prominent holoclone stem cell-like signatures of keratinocytes, and the recently described Gibbin-dependent signature of fibroblasts. The latter correlates with enhanced graftability of iSCs. In conclusion, DEBCT overcomes manufacturing and safety roadblocks and establishes a reproducible, safe, and cGMP-compatible therapeutic approach to heal lesions of DEB patients.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号