Pluripotent Stem Cells

多能干细胞
  • 文章类型: Journal Article
    尽管患者预后有所改善,儿科癌症仍然是儿童非意外死亡的主要原因.最近对儿科癌症患者的遗传分析表明,种系遗传易感性和癌症特异性体细胞驱动突变都具有重要作用。越来越多,有证据表明,起源癌细胞转化的发育时间点对于肿瘤身份和治疗反应至关重要.因此,未来的治疗发展将通过使用忠实地概括遗传背景的疾病模型来支持,细胞起源,和儿童癌症的脆弱性发展窗口。人类干细胞有可能将所有这些特征整合到儿科癌症模型中,同时作为快速遗传和药理测试的平台。在这次审查中,我们描述了人类干细胞是如何用于儿科癌症模型的,以及这些模型与其他儿科癌症模型模式的比较.
    今天,儿童癌症是儿童非意外死亡的主要原因.为了进一步改善结果,对于研究人员和临床医生来说,认识到小儿癌症与成人癌症的区别非常重要。遗传的癌症风险可能在儿科癌症风险中发挥更大的作用,和随后的肿瘤特异性获得性驱动突变启动肿瘤形成。然而,遗传和获得性突变之间存在实质性的相互作用,这支持同时考虑两者。生物技术的最新进展,改善了早期发育细胞和儿科癌细胞之间的匹配,尽管某些儿童中枢神经系统肿瘤的细胞来源仍然难以捉摸。越来越多,证据,特别是在小儿髓母细胞瘤中,证明了癌细胞起源转化的发育时间点对于肿瘤身份和治疗反应至关重要。因此,未来的治疗发展将通过使用忠实地概括遗传背景的疾病模型来支持,细胞起源,和儿科癌症的发育窗口。人类干细胞有可能将所有这些特征整合到儿科癌症模型中,同时作为快速遗传和药理测试的平台。在这次审查中,我们描述了如何使用人类干细胞来模拟儿科癌症,这些模型与其他儿科癌症模型相比,以及未来如何改进这些模型。
    Despite improvements in patient outcomes, pediatric cancer remains a leading cause of non-accidental death in children. Recent genetic analysis of patients with pediatric cancers indicates an important role for both germline genetic predisposition and cancer-specific somatic driver mutations. Increasingly, evidence demonstrates that the developmental timepoint at which the cancer cell-of-origin transforms is critical to tumor identity and therapeutic response. Therefore, future therapeutic development would be bolstered by the use of disease models that faithfully recapitulate the genetic context, cell-of-origin, and developmental window of vulnerability in pediatric cancers. Human stem cells have the potential to incorporate all of these characteristics into a pediatric cancer model, while serving as a platform for rapid genetic and pharmacological testing. In this review, we describe how human stem cells have been used to model pediatric cancers and how these models compare to other pediatric cancer model modalities.
    Today, pediatric cancer is a leading cause of non-accidental death in children. In order to further improve outcomes, it is important for researchers and clinicians alike to recognize how pediatric cancers are distinct from adult cancers. Inherited risk of cancer may play a greater role in pediatric cancer risk, and subsequent tumor-specific acquired driver mutations initiate tumor formation. However, there is substantial interaction between inherited and acquired mutations, which supports consideration of both simultaneously. Recent advancements in biotechnology, have improved matching between early cells of development and pediatric cancer cells, although cell-of-origin for certain pediatric central nervous system tumors remain elusive. Increasingly, evidence, particularly in pediatric medulloblastoma, demonstrates that the developmental timepoint at which the cancer cell-of-origin transforms is critical to tumor identity and therapeutic response. Therefore, future therapeutic development would be bolstered by the use of disease models that faithfully recapitulate the genetic context, cell-of-origin, and developmental window of pediatric cancers. Human stem cells have the potential to incorporate all of these characteristics into a pediatric cancer model, while serving as a platform for rapid genetic and pharmacological testing. In this review, we describe how human stem cells have been used to model pediatric cancers, how human these models compare to other pediatric cancer model modalities, and how these models can be improved in the future.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Case Reports
    Brugada综合征(BrS)是一种遗传的常染色体显性遗传心脏通道病。SCN5A基因的致病性罕见突变,编码电压依赖性心脏Na+通道蛋白(Nav1.5)的α亚基,在20%的BrS患者中被发现,影响通道的正确功能。迄今为止,尽管数百种SCN5A变种与BrS相关,在大多数情况下,潜在的致病机制仍不清楚。因此,SCN5ABrS罕见变异体的功能特征仍然是一个主要障碍,是确认其致病作用的基础.由多能干细胞(PSC)分化的人心肌细胞(CM)已被广泛证明是研究心脏病的可靠平台。能够概括疾病的特定特征,包括心律失常事件和传导异常。基于此,在这项研究中,我们对BrS家族性罕见变异NM_198056.2:c.3673G>A(NP_932173.1:p。Glu1225Lys),以前从未在心脏相关的背景下进行功能表征,作为人类心肌细胞。使用编码GFP标记的SCN5A基因的特定慢病毒载体,该基因携带特定的c.3673G>A变体和与对照PSC(PSC-CM)分化的CM,我们证明了突变的Nav1.5受损,因此表明罕见的BrS检测变体的致病性。更广泛地说,我们的工作支持PSC-CMs用于评估基因变异的致病性,由于下一代测序方法的进步及其在基因检测中的大量使用,其鉴定呈指数级增长。
    Brugada syndrome (BrS) is an inherited autosomal dominant cardiac channelopathy. Pathogenic rare mutations in the SCN5A gene, encoding the alpha-subunit of the voltage-dependent cardiac Na+ channel protein (Nav1.5), are identified in 20% of BrS patients, affecting the correct function of the channel. To date, even though hundreds of SCN5A variants have been associated with BrS, the underlying pathogenic mechanisms are still unclear in most cases. Therefore, the functional characterization of the SCN5A BrS rare variants still represents a major hurdle and is fundamental to confirming their pathogenic effect. Human cardiomyocytes (CMs) differentiated from pluripotent stem cells (PSCs) have been extensively demonstrated to be reliable platforms for investigating cardiac diseases, being able to recapitulate specific traits of disease, including arrhythmic events and conduction abnormalities. Based on this, in this study, we performed a functional analysis of the BrS familial rare variant NM_198056.2:c.3673G>A (NP_932173.1:p.Glu1225Lys), which has been never functionally characterized before in a cardiac-relevant context, as the human cardiomyocyte. Using a specific lentiviral vector encoding a GFP-tagged SCN5A gene carrying the specific c.3673G>A variant and CMs differentiated from control PSCs (PSC-CMs), we demonstrated an impairment of the mutated Nav1.5, thus suggesting the pathogenicity of the rare BrS detected variant. More broadly, our work supports the application of PSC-CMs for the assessment of the pathogenicity of gene variants, the identification of which is increasing exponentially due to the advances in next-generation sequencing methods and their massive use in genetic testing.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • DOI:
    文章类型: News
    用常规动物模型或二维(2D)培养的细胞系研究人类生物学一直是具有挑战性的。干细胞生物学的最新进展使体外培养干细胞成为可能,导致建立称为类器官的体外三维(3D)器官样结构。类器官是模拟体内对应物的组织结构和功能的自组织3D微型组织。目前,可以为多种组织如肠建立类器官,大脑,肾,前列腺,胰腺,肝脏,膀胱,心,和视网膜,来自多能干细胞(PSC),包括胚胎干细胞(ESCs)和诱导多能干细胞(iPSCs),或成体干细胞(AdSC)。除了正常的类器官,已经从各种人类肿瘤如胰腺,结直肠,乳房,肝脏,前列腺,和膀胱肿瘤。此外,生物工程技术,包括生物材料和支架制造,生物打印,和微流体最近已被应用于体外创建更成熟和复杂的类器官和微型组织。结合最近先进的计算分析,包括多组学分析和生物信息学进一步促进了使用人类类器官作为人类疾病建模的新平台的过程。药物筛选,以确定潜在的靶标和新疗法,以及精准医学和再生疗法的发展。
    Studying human biology has been challenging with conventional animal models or two-dimensional (2D) cultured cell lines. Recent advances in stem cell biology have made it possible to culture stem cells in vitro, leading to the establishment of in vitro three-dimensional (3D) organ-like structures known as organoids. Organoids are self-organizing 3D miniature tissues that mimic the tissue architecture and functionality of in vivo counterparts. Currently, organoids can be established for multiple tissues such as the intestine, brain, kidney, prostate, pancreas, liver, bladder, heart, and retina, either from pluripotent stem cells (PSCs), including embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), or adult stem cells (AdSCs). In addition to normal organoids, patient-derived tumor organoids have been established from various human tumors such as pancreatic, colorectal, breast, liver, prostate, and bladder tumors. Also, bioengineering technologies including biomaterial and scaffold fabrication, bioprinting, and microfluidics have been recently applied to create more mature and complex organoids and miniature tissues in vitro. Incorporating recently advanced computational analyses including multi-omics profiling and bioinformatics further facilitated the process of using human organoids as a novel platform for human disease modeling, drug screening to identify potential targets and novel therapeutics, and the development of precision medicine and regenerative therapies. [BMB Reports 2023; 56(1): 1].
    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
    Charcot-Marie-Tooth型4B3(CMT4B3)是与MTMR5/SBF1基因变异相关的遗传性神经病的一种罕见亚型。在这里,我们报道了一名12岁意大利女孩的hiPSC线的产生和表征,该女孩患有运动和轴突受累的早期严重多发性神经病,在MTMR5/SBF1基因中具有双等位基因变体。使用非整合性游离质粒重编程成纤维细胞,和iPSC成功通过了干性和多能性测试。产生患者特异性hiPSC以获得用于研究这种罕见状况的疾病模型。
    Charcot-Marie-Tooth type 4B3 (CMT4B3) is a rare subtype of hereditary neuropathy associated with variants in the MTMR5/SBF1 gene. Herein, we report the generation and characterization of a hiPSC line from a 12-year-old Italian girl with early onset severe polyneuropathy with motor and axonal involvement, harboring biallelic variants in the MTMR5/SBF1 gene. Fibroblasts were reprogrammed using non-integrating episomal plasmids, and iPSCs successfully passed the stemness and pluripotency tests. Patient-specific hiPSCs were produced to obtain a disease model for the study of this rare condition.
    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
    Nodal is a transforming growth factor-β (TGF-β) superfamily member that plays a number of critical roles in mammalian embryonic development. Nodal is essential for the support of the peri-implantation epiblast in the mouse embryo and subsequently acts to specify mesendodermal fate at the time of gastrulation and, later, left-right asymmetry. Maintenance of human pluripotent stem cells (hPSCs) in vitro is dependent on Nodal signaling. Because it has proven difficult to prepare a biologically active form of recombinant Nodal protein, Activin or TGFB1 are widely used as surrogates for NODAL in hPSC culture. Nonetheless, the expression of the components of an endogenous Nodal signaling pathway in hPSC provides a potential autocrine pathway for the regulation of self-renewal in this system. Here we review recent studies that have clarified the role of Nodal signaling in pluripotent stem cell populations, highlighted spatial restrictions on Nodal signaling, and shown that Nodal functions in vivo as a heterodimer with GDF3, another TGF-β superfamily member expressed by hPSC. We discuss the role of this pathway in the maintenance of the epiblast and hPSC in light of these new advances.
    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
    基于细胞的疗法必须以可重复和具有成本效益的制造来实现临床疗效和安全性。这项研究使用基于人类多能干细胞的多巴胺能神经元细胞治疗产品的范例解决了过程开发问题。早期识别和纠正产品安全和制造过程的风险减少了后期开发中昂贵且耗时的桥接研究。新产品介绍图用于确定特定于产品的开发要求。本文以系统性风险分析为例。基于当前价值的预期优先级指导有关过程研究顺序以及是否以及是否应尽早放弃进一步研究的决策。这三种工具的应用使发展研究的优先次序得以确定。
    Cell-based therapies must achieve clinical efficacy and safety with reproducible and cost-effective manufacturing. This study addresses process development issues using the exemplar of a human pluripotent stem cell-based dopaminergic neuron cell therapy product. Early identification and correction of risks to product safety and the manufacturing process reduces the expensive and time-consuming bridging studies later in development. A New Product Introduction map was used to determine the developmental requirements specific to the product. Systematic Risk Analysis is exemplified here. Expected current value-based prioritization guides decisions about the sequence of process studies and whether and if an early abandonment of further research is appropriate. The application of the three tools enabled prioritization of the development studies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Case Reports
    Carcinosarcoma is a clonal tumor developed through sarcomatoid changes in a carcinoma via the epithelial-mesenchymal transition (EMT). Here, we present an extremely rare case of pulmonary carcinosarcoma characterized by components suggesting pluripotency, namely neuroendocrine, myogenic, and chondrogenic differentiation, based on immunohistochemical analysis. A 42-year-old Japanese man was admitted to our hospital. Analysis of tumor tissue after right upper lobe lobectomy revealed a transition between carcinomatous and sarcomatous components. Immunohistochemical analysis suggested immortality owing to complete loss of p53 and diffuse expression of p16 in both the carcinomatous and sarcomatous components. There were also scattered cell groups expressing aldehyde dehydrogenase 1 family member A1, SOX2, CD133, and c-kit, suggesting the possible presence of cancer stem cells. Our findings in this case suggested that the EMT may play a key role in mediating the immortality of tumor cells in carcinosarcoma and facilitating the pluripotency of cancer stem cells.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    源自人多能干细胞(hPSC)衍生物的三维类器官已经广泛用于体外模型中,用于研究发育和疾病。它们在体外形态发生过程中概括正常人类发育方面的能力产生了具有前所未有的仿生能力的组织结构。当前的类器官衍生方案主要依赖于在3-D球形细胞聚集体中发生的自发形态发生过程,而很少或没有外源控制。这产生了含有仿生组织的微尺度区域的类器官,但在宏观尺度(即100微米到毫米),类器官形态,细胞结构,和细胞组成是非仿生和可变的。目前在微观尺度上缺乏对体外类器官形态发生的控制,这阻碍了实现该技术的潜力,可重复地形成解剖学上正确的人体组织单位,可以作为最佳的人体体外模型,甚至移植。这里,我们回顾了组织工程方法,这些方法可用于开发强大的方法来指导多尺度,3-D人类类器官形态发生。迫切需要这种技术合并,以利用类器官形态发生作为具有仿生解剖学和生理学的功能性人体组织工程的工具。
    人类PSC衍生的3-D类器官正在彻底改变生物医学科学。它们能够在患者特定的遗传背景和前所未有的仿生组织微环境中研究发育和疾病。然而,他们不受控制,微尺度的自发形态发生产生了宏观类器官形态的不一致,细胞结构,和细胞组成限制了它们的标准化和应用。将组织工程方法与类器官衍生方案相结合,可以使我们通过指导标准化的体外形态发生来利用其潜力,从而在所有规模上产生具有仿生能力的类器官。这样的进步将使得能够使用类器官作为“下一代”功能性组织工程的基础,解剖学模拟人体组织和潜在的新型器官移植。这里,我们讨论了类器官形态发生的关键方面,其中创新的组织工程方法的应用将在实现这一目标方面取得重大进展。
    Three-dimensional organoids derived from human pluripotent stem cell (hPSC) derivatives have become widely used in vitro models for studying development and disease. Their ability to recapitulate facets of normal human development during in vitro morphogenesis produces tissue structures with unprecedented biomimicry. Current organoid derivation protocols primarily rely on spontaneous morphogenesis processes to occur within 3-D spherical cell aggregates with minimal to no exogenous control. This yields organoids containing microscale regions of biomimetic tissues, but at the macroscale (i.e. 100\'s of microns to millimeters), the organoids\' morphology, cytoarchitecture, and cellular composition are non-biomimetic and variable. The current lack of control over in vitro organoid morphogenesis at the microscale induces aberrations at the macroscale, which impedes realization of the technology\'s potential to reproducibly form anatomically correct human tissue units that could serve as optimal human in vitro models and even transplants. Here, we review tissue engineering methodologies that could be used to develop powerful approaches for instructing multiscale, 3-D human organoid morphogenesis. Such technological mergers are critically needed to harness organoid morphogenesis as a tool for engineering functional human tissues with biomimetic anatomy and physiology.
    Human PSC-derived 3-D organoids are revolutionizing the biomedical sciences. They enable the study of development and disease within patient-specific genetic backgrounds and unprecedented biomimetic tissue microenvironments. However, their uncontrolled, spontaneous morphogenesis at the microscale yields inconsistences in macroscale organoid morphology, cytoarchitecture, and cellular composition that limits their standardization and application. Integration of tissue engineering methods with organoid derivation protocols could allow us to harness their potential by instructing standardized in vitro morphogenesis to generate organoids with biomimicry at all scales. Such advancements would enable the use of organoids as a basis for \'next-generation\' tissue engineering of functional, anatomically mimetic human tissues and potentially novel organ transplants. Here, we discuss critical aspects of organoid morphogenesis where application of innovative tissue engineering methodologies would yield significant advancement towards this goal.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    从受影响的胚泡或从患者来源的体细胞产生的人多能干细胞(PSC)是用于疾病建模和药物发现的新兴平台。脆性X综合征(FXS),遗传性智力残疾的主要原因,是在胚胎干细胞和诱导的PCSs中建模的首批疾病之一,并且可以作为在人类疾病研究中利用人类PSC的示例性病例。在过去的十年里,FXS-PSC已用于解决关于FXS的病理生理学的基本问题。在这篇综述中,我们总结了FXS-PSC的生成方法,讨论它们与现有建模系统相比的优缺点,并描述它们在FXS发病机制研究和靶向治疗开发中的应用。
    Human pluripotent stem cells (PSCs) generated from affected blastocysts or from patient-derived somatic cells are an emerging platform for disease modeling and drug discovery. Fragile X syndrome (FXS), the leading cause of inherited intellectual disability, was one of the first disorders modeled in both embryonic stem cells and induced PCSs and can serve as an exemplary case for the utilization of human PSCs in the study of human diseases. Over the past decade, FXS-PSCs have been used to address the fundamental questions regarding the pathophysiology of FXS. In this review we summarize the methodologies for generation of FXS-PSCs, discuss their advantages and disadvantages compared with existing modeling systems and describe their utilization in the study of FXS pathogenesis and in the development of targeted treatment.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Letter
    暂无摘要。
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号