首页 > 最新文献

Current Protocols in Stem Cell Biology最新文献

英文 中文
Human Adipose-Derived Stromal Cell Isolation Methods and Use in Osteogenic and Adipogenic In Vivo Applications 人脂肪来源的基质细胞分离方法及其在成骨和脂肪生成中的体内应用
Q2 Biochemistry, Genetics and Molecular Biology Pub Date : 2018-02-13 DOI: 10.1002/cpsc.41
Elizabeth Brett, Ruth Tevlin, Adrian McArdle, Eun Young Seo, Charles K.F. Chan, Derrick C. Wan, Michael T. Longaker

Adipose tissue represents an abundant and easily accessible source of multipotent cells, which may serve as excellent building blocks for tissue engineering. This article presents a newly described protocol for isolating adipose-derived stromal cells (ASCs) from human lipoaspirate, compared to the standard protocol for harvesting ASCs established in 2001.

Human ASC isolation is performed using two methods, and resultant cells are compared through cell yield, cell viability, cell proliferation and regenerative potential. The osteogenic and adipogenic potential of ASCs isolated using both protocols are assessed in vitro and gene expression analysis is performed. The focus of this series of protocols is the regenerative potential of both cell populations in vivo. As such, the two in vivo animal models described are fat graft retention (soft tissue reconstruction) and calvarial defect healing (bone regeneration). The techniques described comprise fat grafting with cell assisted lipotransfer, and calvarial defect creation healed with cell-seeded scaffolds. © 2017 by John Wiley & Sons, Inc.

脂肪组织是一种丰富且易于获取的多能细胞来源,可作为组织工程的优秀构建块。本文介绍了一种从人抽脂液中分离脂肪源性基质细胞(ASCs)的新方法,与2001年建立的脂肪源性基质细胞的标准方法进行了比较。采用两种方法分离人类ASC,并通过细胞产量、细胞活力、细胞增殖和再生潜能对分离得到的细胞进行比较。在体外评估两种方法分离的ASCs的成骨和成脂潜能,并进行基因表达分析。这一系列方案的重点是两种细胞群在体内的再生潜力。因此,描述的两种体内动物模型是脂肪移植物保留(软组织重建)和颅骨缺损愈合(骨再生)。所描述的技术包括细胞辅助脂肪移植的脂肪移植,以及细胞植入支架修复的颅骨缺损。©2017 by John Wiley &儿子,Inc。
{"title":"Human Adipose-Derived Stromal Cell Isolation Methods and Use in Osteogenic and Adipogenic In Vivo Applications","authors":"Elizabeth Brett,&nbsp;Ruth Tevlin,&nbsp;Adrian McArdle,&nbsp;Eun Young Seo,&nbsp;Charles K.F. Chan,&nbsp;Derrick C. Wan,&nbsp;Michael T. Longaker","doi":"10.1002/cpsc.41","DOIUrl":"10.1002/cpsc.41","url":null,"abstract":"<p>Adipose tissue represents an abundant and easily accessible source of multipotent cells, which may serve as excellent building blocks for tissue engineering. This article presents a newly described protocol for isolating adipose-derived stromal cells (ASCs) from human lipoaspirate, compared to the standard protocol for harvesting ASCs established in 2001.</p><p>Human ASC isolation is performed using two methods, and resultant cells are compared through cell yield, cell viability, cell proliferation and regenerative potential. The osteogenic and adipogenic potential of ASCs isolated using both protocols are assessed <i>in vitro</i> and gene expression analysis is performed. The focus of this series of protocols is the regenerative potential of both cell populations <i>in vivo</i>. As such, the two <i>in vivo</i> animal models described are fat graft retention (soft tissue reconstruction) and calvarial defect healing (bone regeneration). The techniques described comprise fat grafting with cell assisted lipotransfer, and calvarial defect creation healed with cell-seeded scaffolds. © 2017 by John Wiley &amp; Sons, Inc.</p>","PeriodicalId":53703,"journal":{"name":"Current Protocols in Stem Cell Biology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2018-02-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1002/cpsc.41","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"35555094","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 6
Isolation of Ready-to-Use Adipose-Derived Stem Cell (ASC) Pellet for Clinical Applications and a Comparative Overview of Alternate Methods for ASC Isolation 用于临床应用的即食脂肪源性干细胞(ASC)颗粒的分离和ASC分离替代方法的比较综述
Q2 Biochemistry, Genetics and Molecular Biology Pub Date : 2018-02-13 DOI: 10.1002/cpsc.29
Edoardo Raposio, Nicolò Bertozzi

Current literature does not offer a standardized method to isolate adipose-derived stem cells (ASCs) for clinical applications and hence clinical studies using ASCs often show inconsistent results. Most of these studies borrow laboratory or benchside-derived protocols, which are complex, time consuming, and involve the use of chemical, animal-derived reagents. In this unit we describe a relatively simple and faster isolation protocol that allows collection of a ready-to-use ASC pellet for clinical application. All steps are performed in a closed circuit in order to guarantee maximum process sterility. Once the adipose tissue is harvested by means of a standard liposuction procedure, it undergoes a first centrifugation in order to remove the oil and serous fractions. Then ASCs are released by enzymatic digestion from the surrounding connective tissue scaffold. Finally a double series of washing and centrifugation allows one to obtain the ASC pellet alone. We usually graft this ASC pellet onto the skin edge and to the bottom of chronic skin ulcers as ASCs proved to be effective in promoting wound healing processes. Moreover, an increasing number of clinical studies are currently ongoing to test their potential in every medical field, from orthopedics to cardiology, oncology, autoimmune diseases, and tissue engineering. © 2017 by John Wiley & Sons, Inc.

目前的文献没有提供一种标准化的方法来分离用于临床应用的脂肪源性干细胞(ASCs),因此使用ASCs进行的临床研究往往显示出不一致的结果。这些研究大多采用实验室或台式衍生方案,这些方案复杂、耗时,并涉及使用化学动物衍生试剂。在本单元中,我们描述了一个相对简单和快速的分离方案,允许收集准备使用的ASC颗粒用于临床应用。所有步骤都在封闭回路中进行,以保证最大限度的工艺无菌性。一旦脂肪组织通过标准吸脂程序被收集,它将经历第一次离心,以去除油和浆液部分。然后,ASCs通过酶消化从周围结缔组织支架中释放出来。最后,双系列洗涤和离心可以单独获得ASC颗粒。我们通常将ASC颗粒移植到慢性皮肤溃疡的皮肤边缘和底部,因为ASC被证明能有效促进伤口愈合过程。此外,目前正在进行越来越多的临床研究,以测试它们在各个医学领域的潜力,从骨科到心脏病学、肿瘤学、自身免疫性疾病和组织工程。©2017 by John Wiley &儿子,Inc。
{"title":"Isolation of Ready-to-Use Adipose-Derived Stem Cell (ASC) Pellet for Clinical Applications and a Comparative Overview of Alternate Methods for ASC Isolation","authors":"Edoardo Raposio,&nbsp;Nicolò Bertozzi","doi":"10.1002/cpsc.29","DOIUrl":"10.1002/cpsc.29","url":null,"abstract":"<p>Current literature does not offer a standardized method to isolate adipose-derived stem cells (ASCs) for clinical applications and hence clinical studies using ASCs often show inconsistent results. Most of these studies borrow laboratory or benchside-derived protocols, which are complex, time consuming, and involve the use of chemical, animal-derived reagents. In this unit we describe a relatively simple and faster isolation protocol that allows collection of a ready-to-use ASC pellet for clinical application. All steps are performed in a closed circuit in order to guarantee maximum process sterility. Once the adipose tissue is harvested by means of a standard liposuction procedure, it undergoes a first centrifugation in order to remove the oil and serous fractions. Then ASCs are released by enzymatic digestion from the surrounding connective tissue scaffold. Finally a double series of washing and centrifugation allows one to obtain the ASC pellet alone. We usually graft this ASC pellet onto the skin edge and to the bottom of chronic skin ulcers as ASCs proved to be effective in promoting wound healing processes. Moreover, an increasing number of clinical studies are currently ongoing to test their potential in every medical field, from orthopedics to cardiology, oncology, autoimmune diseases, and tissue engineering. © 2017 by John Wiley &amp; Sons, Inc.</p>","PeriodicalId":53703,"journal":{"name":"Current Protocols in Stem Cell Biology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2018-02-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1002/cpsc.29","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"35001879","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 41
Neural Stem Cell or Human Induced Pluripotent Stem Cell–Derived GABA-ergic Progenitor Cell Grafting in an Animal Model of Chronic Temporal Lobe Epilepsy 神经干细胞或人诱导多能干细胞衍生的gaba能祖细胞移植在慢性颞叶癫痫动物模型中的应用
Q2 Biochemistry, Genetics and Molecular Biology Pub Date : 2018-02-13 DOI: 10.1002/cpsc.9
Dinesh Upadhya, Bharathi Hattiangady, Geetha A. Shetty, Gabriele Zanirati, Maheedhar Kodali, Ashok K. Shetty

Grafting of neural stem cells (NSCs) or GABA-ergic progenitor cells (GPCs) into the hippocampus could offer an alternative therapy to hippocampal resection in patients with drug-resistant chronic epilepsy, which afflicts >30% of temporal lobe epilepsy (TLE) cases. Multipotent, self-renewing NSCs could be expanded from multiple regions of the developing and adult brain, human embryonic stem cells (hESCs), and human induced pluripotent stem cells (hiPSCs). On the other hand, GPCs could be generated from the medial and lateral ganglionic eminences of the embryonic brain and from hESCs and hiPSCs. To provide comprehensive methodologies involved in testing the efficacy of transplantation of NSCs and GPCs in a rat model of chronic TLE, NSCs derived from the rat medial ganglionic eminence (MGE) and MGE-like GPCs derived from hiPSCs are taken as examples in this unit. The topics comprise description of the required materials, reagents and equipment, methods for obtaining rat MGE-NSCs and hiPSC-derived MGE-like GPCs in culture, generation of chronically epileptic rats, intrahippocampal grafting procedure, post-grafting evaluation of the effects of grafts on spontaneous recurrent seizures and cognitive and mood impairments, analyses of the yield and the fate of graft-derived cells, and the effects of grafts on the host hippocampus. © 2016 by John Wiley & Sons, Inc.

将神经干细胞(NSCs)或gaba -能祖细胞(GPCs)移植到海马体中可以为耐药慢性癫痫患者提供海马切除术的替代治疗方法,30%的颞叶癫痫(TLE)患者患有慢性癫痫。多能、自我更新的NSCs可以从发育和成人大脑的多个区域、人胚胎干细胞(hESCs)和人诱导多能干细胞(hiPSCs)中扩增。另一方面,GPCs可以从胚胎脑的内侧和外侧神经节突起以及hESCs和hiPSCs中产生。为了提供全面的方法来测试NSCs和GPCs移植在大鼠慢性TLE模型中的疗效,本单元以大鼠内侧神经节隆起(MGE)来源的NSCs和hipsc来源的MGE样GPCs为例。主题包括所需材料、试剂和设备的描述,在培养中获得大鼠MGE-NSCs和hipsc衍生的mge样GPCs的方法,慢性癫痫大鼠的产生,海马内移植手术,移植后对移植物对自发性复发性癫痫发作和认知和情绪障碍的影响的评估,移植物衍生细胞的产量和命运的分析,以及移植物对宿主海马的影响。©2016 by John Wiley &儿子,Inc。
{"title":"Neural Stem Cell or Human Induced Pluripotent Stem Cell–Derived GABA-ergic Progenitor Cell Grafting in an Animal Model of Chronic Temporal Lobe Epilepsy","authors":"Dinesh Upadhya,&nbsp;Bharathi Hattiangady,&nbsp;Geetha A. Shetty,&nbsp;Gabriele Zanirati,&nbsp;Maheedhar Kodali,&nbsp;Ashok K. Shetty","doi":"10.1002/cpsc.9","DOIUrl":"10.1002/cpsc.9","url":null,"abstract":"<p>Grafting of neural stem cells (NSCs) or GABA-ergic progenitor cells (GPCs) into the hippocampus could offer an alternative therapy to hippocampal resection in patients with drug-resistant chronic epilepsy, which afflicts &gt;30% of temporal lobe epilepsy (TLE) cases. Multipotent, self-renewing NSCs could be expanded from multiple regions of the developing and adult brain, human embryonic stem cells (hESCs), and human induced pluripotent stem cells (hiPSCs). On the other hand, GPCs could be generated from the medial and lateral ganglionic eminences of the embryonic brain and from hESCs and hiPSCs. To provide comprehensive methodologies involved in testing the efficacy of transplantation of NSCs and GPCs in a rat model of chronic TLE, NSCs derived from the rat medial ganglionic eminence (MGE) and MGE-like GPCs derived from hiPSCs are taken as examples in this unit. The topics comprise description of the required materials, reagents and equipment, methods for obtaining rat MGE-NSCs and hiPSC-derived MGE-like GPCs in culture, generation of chronically epileptic rats, intrahippocampal grafting procedure, post-grafting evaluation of the effects of grafts on spontaneous recurrent seizures and cognitive and mood impairments, analyses of the yield and the fate of graft-derived cells, and the effects of grafts on the host hippocampus. © 2016 by John Wiley &amp; Sons, Inc.</p>","PeriodicalId":53703,"journal":{"name":"Current Protocols in Stem Cell Biology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2018-02-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1002/cpsc.9","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"34670819","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 23
Visualization and Modeling of the In Vivo Distribution of Mesenchymal Stem Cells 间充质干细胞体内分布的可视化和建模
Q2 Biochemistry, Genetics and Molecular Biology Pub Date : 2018-02-13 DOI: 10.1002/cpsc.39
Haolu Wang, Camilla A. Thorling, Zhi Ping Xu, Darrell H. G. Crawford, Xiaowen Liang, Xin Liu, Michael S. Roberts

This unit describes a protocol for elucidating the in vivo disposition of administered mesenchymal stem cells (MSCs). Specifically, direct visualization of donor cell spatiotemporal distribution and assessment of donor cell quantity in recipient organs are described. Protocols for data analysis are suggested, with the goal of developing a model to characterize and predict the physiological kinetics of administered MSCs. The use of this model is described, suggesting that it can be applied to abnormal conditions and has potential interspecies and inter-route predictive capability. These universal methods can be employed, regardless of the type of stem cell or disease, to guide future experiments and design treatment protocols. © 2017 by John Wiley & Sons, Inc.

本单元描述了一个方案,阐明体内处置的管理间充质干细胞(MSCs)。具体来说,描述了直接可视化供体细胞的时空分布和评估供体细胞在受体器官中的数量。提出了数据分析的方案,目的是建立一个模型来表征和预测给药MSCs的生理动力学。结果表明,该模型可以应用于异常条件,具有潜在的种间和路线间预测能力。无论干细胞或疾病的类型如何,这些通用方法都可以用于指导未来的实验和设计治疗方案。©2017 by John Wiley &儿子,Inc。
{"title":"Visualization and Modeling of the In Vivo Distribution of Mesenchymal Stem Cells","authors":"Haolu Wang,&nbsp;Camilla A. Thorling,&nbsp;Zhi Ping Xu,&nbsp;Darrell H. G. Crawford,&nbsp;Xiaowen Liang,&nbsp;Xin Liu,&nbsp;Michael S. Roberts","doi":"10.1002/cpsc.39","DOIUrl":"10.1002/cpsc.39","url":null,"abstract":"<p>This unit describes a protocol for elucidating the <i>in vivo</i> disposition of administered mesenchymal stem cells (MSCs). Specifically, direct visualization of donor cell spatiotemporal distribution and assessment of donor cell quantity in recipient organs are described. Protocols for data analysis are suggested, with the goal of developing a model to characterize and predict the physiological kinetics of administered MSCs. The use of this model is described, suggesting that it can be applied to abnormal conditions and has potential interspecies and inter-route predictive capability. These universal methods can be employed, regardless of the type of stem cell or disease, to guide future experiments and design treatment protocols. © 2017 by John Wiley &amp; Sons, Inc.</p>","PeriodicalId":53703,"journal":{"name":"Current Protocols in Stem Cell Biology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2018-02-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1002/cpsc.39","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"35555092","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 3
Mesenchymal Stem Cell Preparation and Transfection-free Ferumoxytol Labeling for MRI Cell Tracking 间充质干细胞制备和无转染阿魏木糖醇标记用于MRI细胞跟踪
Q2 Biochemistry, Genetics and Molecular Biology Pub Date : 2018-02-13 DOI: 10.1002/cpsc.38
Li Liu, Chien Ho

Mesenchymal stem cells (MSCs) are multipotent cells and are the most widely studied cell type for stem cell therapies. In vivo cell tracking of MSCs labeled with an FDA-approved superparamagnetic iron-oxide (SPIO) particle by magnetic resonance imaging (MRI) provides essential information, e.g., MSC engraftment, survival, and fate, thus improving cell therapy accuracy. However, current methodology for labeling MSCs with Ferumoxytol (Feraheme®), the only FDA-approved SPIO particle, needs transfection agents. This unit describes a new “bio-mimicry” protocol to prepare more native MSCs by using more “in vivo environment” of MSCs, so that the phagocytic activity of cultured MSCs is restored and expanded MSCs can be labeled with Ferumoxytol, without the need for transfection agents and/or electroporation. Moreover, MSCs re-size to a more native size, reducing from 32.0 to 19.5 μm. The MSCs prepared from this protocol retain more native properties and would be useful for biomedical applications and MSC-tracking studies by MRI. © 2017 by John Wiley & Sons, Inc.

间充质干细胞(MSCs)是一种多能细胞,是干细胞治疗中研究最广泛的细胞类型。通过磁共振成像(MRI)对经fda批准的超顺磁性氧化铁(SPIO)颗粒标记的间充质干细胞进行体内细胞跟踪,可提供必要的信息,例如间充质干细胞的植入、存活和命运,从而提高细胞治疗的准确性。然而,目前用fda唯一批准的SPIO颗粒Ferumoxytol (Feraheme®)标记MSCs的方法需要转染剂。本单元描述了一种新的“生物模拟”方案,通过使用更多的MSCs的“体内环境”来制备更多的天然MSCs,从而恢复培养的MSCs的吞噬活性并扩增MSCs,而无需转染剂和/或电穿孔。此外,MSCs的尺寸从32.0 μm减小到19.5 μm。该方法制备的间质干细胞保留了更多的天然特性,可用于生物医学应用和MRI对间质干细胞的跟踪研究。©2017 by John Wiley &儿子,Inc。
{"title":"Mesenchymal Stem Cell Preparation and Transfection-free Ferumoxytol Labeling for MRI Cell Tracking","authors":"Li Liu,&nbsp;Chien Ho","doi":"10.1002/cpsc.38","DOIUrl":"10.1002/cpsc.38","url":null,"abstract":"<p>Mesenchymal stem cells (MSCs) are multipotent cells and are the most widely studied cell type for stem cell therapies. <i>In vivo</i> cell tracking of MSCs labeled with an FDA-approved superparamagnetic iron-oxide (SPIO) particle by magnetic resonance imaging (MRI) provides essential information, e.g., MSC engraftment, survival, and fate, thus improving cell therapy accuracy. However, current methodology for labeling MSCs with Ferumoxytol (Feraheme<sup>®</sup>), the only FDA-approved SPIO particle, needs transfection agents. This unit describes a new “bio-mimicry” protocol to prepare more native MSCs by using more “<i>in vivo</i> environment” of MSCs, so that the phagocytic activity of cultured MSCs is restored and expanded MSCs can be labeled with Ferumoxytol, without the need for transfection agents and/or electroporation. Moreover, MSCs re-size to a more native size, reducing from 32.0 to 19.5 μm. The MSCs prepared from this protocol retain more native properties and would be useful for biomedical applications and MSC-tracking studies by MRI. © 2017 by John Wiley &amp; Sons, Inc.</p>","PeriodicalId":53703,"journal":{"name":"Current Protocols in Stem Cell Biology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2018-02-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1002/cpsc.38","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"35555093","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 11
CRISPR/Cas9-Based Safe-Harbor Gene Editing in Rhesus iPSCs 基于CRISPR/ cas9的安全港基因编辑在恒河猴iPSCs中的应用
Q2 Biochemistry, Genetics and Molecular Biology Pub Date : 2018-02-13 DOI: 10.1002/cpsc.37
Ravi Chandra Yada, John W. Ostrominski, Ilker Tunc, So Gun Hong, Jizhong Zou, Cynthia E. Dunbar

NHP iPSCs provide a unique opportunity to test safety and efficacy of iPSC-derived therapies in clinically relevant NHP models. To monitor these cells in vivo, there is a need for safe and efficient labeling methods. Gene insertion into genomic safe harbors (GSHs) supports reliable transgene expression while minimizing the risk the modification poses to the host genome or target cell. Specifically, this protocol demonstrates targeting of the adeno-associated virus site 1 (AAVS1), one of the most widely used GSH loci in the human genome, with CRISPR/Cas9, allowing targeted marker or therapeutic gene insertion in rhesus macaque induced pluripotent stem cells (RhiPSCs). Furthermore, detailed instructions for screening targeted clones and a tool for assessing potential off-target nuclease activity are provided. © 2017 by John Wiley & Sons, Inc.

NHP诱导多能干细胞提供了一个独特的机会,在临床相关的NHP模型中测试ipsc衍生疗法的安全性和有效性。为了在体内监测这些细胞,需要安全有效的标记方法。基因插入基因组安全港(GSHs)支持可靠的转基因表达,同时将修饰对宿主基因组或靶细胞的风险降至最低。具体来说,该方案展示了利用CRISPR/Cas9靶向人类基因组中最广泛使用的GSH位点之一腺相关病毒位点1 (AAVS1),允许在恒河猴诱导的多能干细胞(RhiPSCs)中靶向标记或治疗性基因插入。此外,详细说明筛选目标克隆和评估潜在脱靶核酸酶活性的工具提供。©2017 by John Wiley &儿子,Inc。
{"title":"CRISPR/Cas9-Based Safe-Harbor Gene Editing in Rhesus iPSCs","authors":"Ravi Chandra Yada,&nbsp;John W. Ostrominski,&nbsp;Ilker Tunc,&nbsp;So Gun Hong,&nbsp;Jizhong Zou,&nbsp;Cynthia E. Dunbar","doi":"10.1002/cpsc.37","DOIUrl":"10.1002/cpsc.37","url":null,"abstract":"<p>NHP iPSCs provide a unique opportunity to test safety and efficacy of iPSC-derived therapies in clinically relevant NHP models. To monitor these cells in vivo, there is a need for safe and efficient labeling methods. Gene insertion into genomic safe harbors (GSHs) supports reliable transgene expression while minimizing the risk the modification poses to the host genome or target cell. Specifically, this protocol demonstrates targeting of the adeno-associated virus site 1 (AAVS1), one of the most widely used GSH loci in the human genome, with CRISPR/Cas9, allowing targeted marker or therapeutic gene insertion in rhesus macaque induced pluripotent stem cells (RhiPSCs). Furthermore, detailed instructions for screening targeted clones and a tool for assessing potential off-target nuclease activity are provided. © 2017 by John Wiley &amp; Sons, Inc.</p>","PeriodicalId":53703,"journal":{"name":"Current Protocols in Stem Cell Biology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2018-02-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1002/cpsc.37","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"35252302","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 6
Rapid Screening of the Endodermal Differentiation Potential of Human Pluripotent Stem Cells 人多能干细胞内胚层分化潜能的快速筛选
Q2 Biochemistry, Genetics and Molecular Biology Pub Date : 2018-02-13 DOI: 10.1002/cpsc.36
Richard Siller, Gareth J. Sullivan

Human pluripotent stem cells (hPSCs) hold tremendous promise for regenerative medicine, disease modeling, toxicology screening, and developmental biology. These applications are hindered due to inherent differences in differentiation potential observed among different hPSC lines. This is particularly true for the differentiation of hPSCs toward the endodermal lineage. Several groups have developed methods to screen hPSCs for their endodermal differentiation potential (EP). Particularly notable studies include (i) the use of WNT3A expression as a predictive biomarker, (ii) an embryoid body–based screen, and (iii) a transcriptomics-based approach. We recently developed a rapid screen to access the EP of hPSCs solely based on morphological analysis. The screen takes 4 days to perform and yields results that are easy to interpret. As the screen is based on our recently developed small molecule protocol for hepatocyte like cell (HLC) differentiation of hPSCs, this method is extremely cost-effective compared to the aforementioned approaches. © 2017 by John Wiley & Sons, Inc.

人类多能干细胞(hPSCs)在再生医学、疾病建模、毒理学筛选和发育生物学方面有着巨大的前景。由于在不同的hPSC系中观察到的分化电位的固有差异,这些应用受到阻碍。对于向内胚层谱系分化的人造血干细胞来说尤其如此。一些研究小组已经开发出了筛选人造血干细胞的内胚层分化潜能(EP)的方法。特别值得注意的研究包括(i)使用WNT3A表达作为预测性生物标志物,(ii)基于胚胎体的筛选,以及(iii)基于转录组学的方法。我们最近开发了一种基于形态学分析的快速筛选方法来获取人造血干细胞的EP。该屏幕需要4天的时间来执行,并产生易于解释的结果。由于筛选是基于我们最近开发的肝细胞样细胞(HLC)分化的小分子方案,与上述方法相比,该方法具有极高的成本效益。©2017 by John Wiley &儿子,Inc。
{"title":"Rapid Screening of the Endodermal Differentiation Potential of Human Pluripotent Stem Cells","authors":"Richard Siller,&nbsp;Gareth J. Sullivan","doi":"10.1002/cpsc.36","DOIUrl":"10.1002/cpsc.36","url":null,"abstract":"<p>Human pluripotent stem cells (hPSCs) hold tremendous promise for regenerative medicine, disease modeling, toxicology screening, and developmental biology. These applications are hindered due to inherent differences in differentiation potential observed among different hPSC lines. This is particularly true for the differentiation of hPSCs toward the endodermal lineage. Several groups have developed methods to screen hPSCs for their endodermal differentiation potential (EP). Particularly notable studies include (i) the use of <i>WNT3A</i> expression as a predictive biomarker, (ii) an embryoid body–based screen, and (iii) a transcriptomics-based approach. We recently developed a rapid screen to access the EP of hPSCs solely based on morphological analysis. The screen takes 4 days to perform and yields results that are easy to interpret. As the screen is based on our recently developed small molecule protocol for hepatocyte like cell (HLC) differentiation of hPSCs, this method is extremely cost-effective compared to the aforementioned approaches. © 2017 by John Wiley &amp; Sons, Inc.</p>","PeriodicalId":53703,"journal":{"name":"Current Protocols in Stem Cell Biology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2018-02-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1002/cpsc.36","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"35252304","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 11
Generation of Oligodendrogenic Spinal Neural Progenitor Cells From Human Induced Pluripotent Stem Cells 从人诱导多能干细胞生成少突性脊神经祖细胞
Q2 Biochemistry, Genetics and Molecular Biology Pub Date : 2017-08-14 DOI: 10.1002/cpsc.31
Mohamad Khazaei, Christopher S. Ahuja, Michael G. Fehlings

This unit describes protocols for the efficient generation of oligodendrogenic neural progenitor cells (o-NPCs) from human induced pluripotent stem cells (hiPSCs). Specifically, detailed methods are provided for the maintenance and differentiation of hiPSCs, human induced pluripotent stem cell-derived neural progenitor cells (hiPS-NPCs), and human induced pluripotent stem cell-oligodendrogenic neural progenitor cells (hiPSC-o-NPCs) with the final products being suitable for in vitro experimentation or in vivo transplantation. Throughout, cell exposure to growth factors and patterning morphogens has been optimized for both concentration and timing, based on the literature and empirical experience, resulting in a robust and highly efficient protocol. Using this derivation procedure, it is possible to obtain millions of oligodendrogenic-NPCs within 40 days of initial cell plating which is substantially shorter than other protocols for similar cell types. This protocol has also been optimized to use translationally relevant human iPSCs as the parent cell line. The resultant cells have been extensively characterized both in vitro and in vivo and express key markers of an oligodendrogenic lineage. © 2017 by John Wiley & Sons, Inc.

本单元描述了从人诱导多能干细胞(hiPSCs)高效生成少突胶质神经祖细胞(o- npc)的方法。具体而言,提供了hipsc、人诱导多能干细胞衍生神经祖细胞(hiPS-NPCs)和人诱导多能干细胞-少突神经祖细胞(hiPSC-o-NPCs)的维持和分化的详细方法,最终产品适合体外实验或体内移植。在整个过程中,基于文献和经验经验,细胞暴露于生长因子和模式形态因子的浓度和时间都得到了优化,从而形成了一个强大而高效的方案。使用这种衍生程序,可以在初始细胞镀后40天内获得数百万个少突胶质细胞,这比类似细胞类型的其他方案要短得多。该方案还经过优化,可以使用翻译相关的人类iPSCs作为亲本细胞系。由此产生的细胞在体外和体内都得到了广泛的表征,并表达了一个少突细胞谱系的关键标记。©2017 by John Wiley &儿子,Inc。
{"title":"Generation of Oligodendrogenic Spinal Neural Progenitor Cells From Human Induced Pluripotent Stem Cells","authors":"Mohamad Khazaei,&nbsp;Christopher S. Ahuja,&nbsp;Michael G. Fehlings","doi":"10.1002/cpsc.31","DOIUrl":"10.1002/cpsc.31","url":null,"abstract":"<p>This unit describes protocols for the efficient generation of oligodendrogenic neural progenitor cells (o-NPCs) from human induced pluripotent stem cells (hiPSCs). Specifically, detailed methods are provided for the maintenance and differentiation of hiPSCs, human induced pluripotent stem cell-derived neural progenitor cells (hiPS-NPCs), and human induced pluripotent stem cell-oligodendrogenic neural progenitor cells (hiPSC-o-NPCs) with the final products being suitable for in vitro experimentation or in vivo transplantation. Throughout, cell exposure to growth factors and patterning morphogens has been optimized for both concentration and timing, based on the literature and empirical experience, resulting in a robust and highly efficient protocol. Using this derivation procedure, it is possible to obtain millions of oligodendrogenic-NPCs within 40 days of initial cell plating which is substantially shorter than other protocols for similar cell types. This protocol has also been optimized to use translationally relevant human iPSCs as the parent cell line. The resultant cells have been extensively characterized both in vitro and in vivo and express key markers of an oligodendrogenic lineage. © 2017 by John Wiley &amp; Sons, Inc.</p>","PeriodicalId":53703,"journal":{"name":"Current Protocols in Stem Cell Biology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2017-08-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1002/cpsc.31","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"35268003","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 15
Generation of Xeno-Free, cGMP-Compliant Patient-Specific iPSCs from Skin Biopsy 从皮肤活检中生成无xeno、符合cgmp的患者特异性iPSCs
Q2 Biochemistry, Genetics and Molecular Biology Pub Date : 2017-08-14 DOI: 10.1002/cpsc.30
Luke A. Wiley, Kristin R. Anfinson, Cathryn M. Cranston, Emily E. Kaalberg, Malia M. Collins, Robert F. Mullins, Edwin M. Stone, Budd A. Tucker

This unit describes protocols for the generation of clinical-grade patient-specific induced pluripotent stem cell (iPSC)–derived retinal cells from patients with inherited retinal degenerative blindness. Specifically, we describe how, using xeno-free reagents in an ISO class 5 environment, one can isolate and culture dermal fibroblasts, generate iPSCs, and derive autologous retinal cells via 3-D differentiation. The universal methods described herein for the isolation of dermal fibroblasts and generation of iPSCs can be employed regardless of disease, tissue, or cell type of interest. © 2017 by John Wiley & Sons, Inc.

本单元描述了从遗传性视网膜退行性失明患者中产生临床级患者特异性诱导多能干细胞(iPSC)衍生视网膜细胞的方案。具体来说,我们描述了如何在ISO 5级环境中使用无xeno试剂,分离和培养真皮成纤维细胞,生成iPSCs,并通过3-D分化获得自体视网膜细胞。本文描述的用于分离真皮成纤维细胞和生成iPSCs的通用方法可用于任何疾病、组织或感兴趣的细胞类型。©2017 by John Wiley &儿子,Inc。
{"title":"Generation of Xeno-Free, cGMP-Compliant Patient-Specific iPSCs from Skin Biopsy","authors":"Luke A. Wiley,&nbsp;Kristin R. Anfinson,&nbsp;Cathryn M. Cranston,&nbsp;Emily E. Kaalberg,&nbsp;Malia M. Collins,&nbsp;Robert F. Mullins,&nbsp;Edwin M. Stone,&nbsp;Budd A. Tucker","doi":"10.1002/cpsc.30","DOIUrl":"10.1002/cpsc.30","url":null,"abstract":"<p>This unit describes protocols for the generation of clinical-grade patient-specific induced pluripotent stem cell (iPSC)–derived retinal cells from patients with inherited retinal degenerative blindness. Specifically, we describe how, using xeno-free reagents in an ISO class 5 environment, one can isolate and culture dermal fibroblasts, generate iPSCs, and derive autologous retinal cells via 3-D differentiation. The universal methods described herein for the isolation of dermal fibroblasts and generation of iPSCs can be employed regardless of disease, tissue, or cell type of interest. © 2017 by John Wiley &amp; Sons, Inc.</p>","PeriodicalId":53703,"journal":{"name":"Current Protocols in Stem Cell Biology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2017-08-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1002/cpsc.30","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"35268005","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 16
Functional Stem Cell Integration into Neural Networks Assessed by Organotypic Slice Cultures 器官型切片培养评估功能性干细胞与神经网络的整合
Q2 Biochemistry, Genetics and Molecular Biology Pub Date : 2017-08-14 DOI: 10.1002/cpsc.34
David Forsberg, Charoensri Thonabulsombat, Johan Jäderstad, Linda Maria Jäderstad, Petri Olivius, Eric Herlenius

Re-formation or preservation of functional, electrically active neural networks has been proffered as one of the goals of stem cell–mediated neural therapeutics. A primary issue for a cell therapy approach is the formation of functional contacts between the implanted cells and the host tissue. Therefore, it is of fundamental interest to establish protocols that allow us to delineate a detailed time course of grafted stem cell survival, migration, differentiation, integration, and functional interaction with the host. One option for in vitro studies is to examine the integration of exogenous stem cells into an existing active neural network in ex vivo organotypic cultures. Organotypic cultures leave the structural integrity essentially intact while still allowing the microenvironment to be carefully controlled. This allows detailed studies over time of cellular responses and cell-cell interactions, which are not readily performed in vivo. This unit describes procedures for using organotypic slice cultures as ex vivo model systems for studying neural stem cell and embryonic stem cell engraftment and communication with CNS host tissue. © 2017 by John Wiley & Sons, Inc.

重构或保存功能,电活性神经网络已被提供作为干细胞介导的神经治疗的目标之一。细胞治疗方法的一个主要问题是在植入细胞和宿主组织之间形成功能性接触。因此,建立能够描述移植干细胞存活、迁移、分化、整合以及与宿主功能相互作用的详细时间过程的方案具有重要意义。体外研究的一个选择是在体外器官型培养中检查外源干细胞与现有活性神经网络的整合。有机型培养基本上保持结构完整性,同时仍然允许微环境被仔细控制。这使得随着时间的推移,细胞反应和细胞间相互作用的详细研究成为可能,这在体内是不容易进行的。本单元描述了使用器官型切片培养作为体外模型系统来研究神经干细胞和胚胎干细胞的植入以及与中枢神经系统宿主组织的交流。©2017 by John Wiley &儿子,Inc。
{"title":"Functional Stem Cell Integration into Neural Networks Assessed by Organotypic Slice Cultures","authors":"David Forsberg,&nbsp;Charoensri Thonabulsombat,&nbsp;Johan Jäderstad,&nbsp;Linda Maria Jäderstad,&nbsp;Petri Olivius,&nbsp;Eric Herlenius","doi":"10.1002/cpsc.34","DOIUrl":"10.1002/cpsc.34","url":null,"abstract":"<p>Re-formation or preservation of functional, electrically active neural networks has been proffered as one of the goals of stem cell–mediated neural therapeutics. A primary issue for a cell therapy approach is the formation of functional contacts between the implanted cells and the host tissue. Therefore, it is of fundamental interest to establish protocols that allow us to delineate a detailed time course of grafted stem cell survival, migration, differentiation, integration, and functional interaction with the host. One option for in vitro studies is to examine the integration of exogenous stem cells into an existing active neural network in ex vivo organotypic cultures. Organotypic cultures leave the structural integrity essentially intact while still allowing the microenvironment to be carefully controlled. This allows detailed studies over time of cellular responses and cell-cell interactions, which are not readily performed in vivo. This unit describes procedures for using organotypic slice cultures as ex vivo model systems for studying neural stem cell and embryonic stem cell engraftment and communication with CNS host tissue. © 2017 by John Wiley &amp; Sons, Inc.</p>","PeriodicalId":53703,"journal":{"name":"Current Protocols in Stem Cell Biology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2017-08-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1002/cpsc.34","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"35267931","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 7
期刊
Current Protocols in Stem Cell Biology
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1