首页 > 最新文献

Current Protocols in Stem Cell Biology最新文献

英文 中文
Development of a Three-Dimensional Bioengineering Technology to Generate Lung Tissue for Personalized Disease Modeling 三维生物工程技术生成肺组织用于个性化疾病建模的发展
Q2 Biochemistry, Genetics and Molecular Biology Pub Date : 2018-06-07 DOI: 10.1002/cpsc.56
Dan C. Wilkinson, Michael Mellody, Luisa K. Meneses, Ashley C. Hope, Bruce Dunn, Brigitte N. Gomperts

This unit describes a protocol for generation of lung organoids. A lung organoid is a 3D cell/hydrogel composite that resembles the morphology and cellular composition of the human distal lung. These tissue-engineered constructs provide an in vitro model of human lung and are best suited for disease modeling applications. The organoid generation methodology is flexible, allowing for easy scalability in the number of organoids produced and in the ability to accommodate a wide range of cell types. © 2018 by John Wiley & Sons, Inc.

本单元描述了一种生成肺类器官的方案。肺类器官是一种3D细胞/水凝胶复合材料,类似于人类远端肺的形态和细胞组成。这些组织工程构建物提供了人体肺的体外模型,最适合用于疾病建模应用。类器官生成方法是灵活的,允许在产生的类器官数量上容易扩展,并且能够适应广泛的细胞类型。©2018 by John Wiley &儿子,Inc。
{"title":"Development of a Three-Dimensional Bioengineering Technology to Generate Lung Tissue for Personalized Disease Modeling","authors":"Dan C. Wilkinson,&nbsp;Michael Mellody,&nbsp;Luisa K. Meneses,&nbsp;Ashley C. Hope,&nbsp;Bruce Dunn,&nbsp;Brigitte N. Gomperts","doi":"10.1002/cpsc.56","DOIUrl":"10.1002/cpsc.56","url":null,"abstract":"<p>This unit describes a protocol for generation of lung organoids. A lung organoid is a 3D cell/hydrogel composite that resembles the morphology and cellular composition of the human distal lung. These tissue-engineered constructs provide an <i>in vitro</i> model of human lung and are best suited for disease modeling applications. The organoid generation methodology is flexible, allowing for easy scalability in the number of organoids produced and in the ability to accommodate a wide range of cell types. © 2018 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":"2018-06-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1002/cpsc.56","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"36242684","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: 108
Cancer Stem Cell Migration in Three-Dimensional Aligned Collagen Matrices 肿瘤干细胞在三维排列的胶原基质中的迁移
Q2 Biochemistry, Genetics and Molecular Biology Pub Date : 2018-05-24 DOI: 10.1002/cpsc.57
Arja Ray, Rachel K. Morford, Paolo P. Provenzano

Cell migration is strongly influenced by the organization of the surrounding 3-D extracellular matrix. In particular, within fibrous solid tumors, carcinoma cell invasion may be directed by patterns of aligned collagen in the extra-epithelial space. Thus, studying the interactions of heterogeneous populations of cancer cells that include the stem/progenitor-like cancer stem cell subpopulation and aligned collagen networks is critical to our understanding of carcinoma dissemination. Here, we describe a robust method to generate aligned collagen matrices in vitro that mimic in vivo fiber organization. Subsequently, a protocol is presented for seeding aligned matrices with distinct carcinoma cell subpopulations and performing live cell imaging and quantitative analysis of cell migration. Together, the engineered constructs and the imaging techniques laid out here provide a platform to study cancer stem cell migration in 3-D anisotropic collagen with real-time visualization of cellular interactions with the fibrous matrix. © 2018 by John Wiley & Sons, Inc.

细胞迁移受到周围三维细胞外基质组织的强烈影响。特别是,在纤维性实体瘤中,癌细胞的侵袭可能是由上皮外空间排列的胶原蛋白模式所引导的。因此,研究包括干细胞/祖细胞样癌症干细胞亚群和排列的胶原网络在内的异质癌细胞群的相互作用对我们理解癌症传播至关重要。在这里,我们描述了一种强大的方法来产生对齐的胶原基质在体外,模拟体内纤维组织。随后,提出了一种方案,用于播种具有不同癌细胞亚群的排列矩阵,并进行活细胞成像和细胞迁移的定量分析。总之,工程结构和成像技术提供了一个平台来研究癌症干细胞在三维各向异性胶原中的迁移,并实时可视化细胞与纤维基质的相互作用。©2018 by John Wiley &儿子,Inc。
{"title":"Cancer Stem Cell Migration in Three-Dimensional Aligned Collagen Matrices","authors":"Arja Ray,&nbsp;Rachel K. Morford,&nbsp;Paolo P. Provenzano","doi":"10.1002/cpsc.57","DOIUrl":"10.1002/cpsc.57","url":null,"abstract":"<p>Cell migration is strongly influenced by the organization of the surrounding 3-D extracellular matrix. In particular, within fibrous solid tumors, carcinoma cell invasion may be directed by patterns of aligned collagen in the extra-epithelial space. Thus, studying the interactions of heterogeneous populations of cancer cells that include the stem/progenitor-like cancer stem cell subpopulation and aligned collagen networks is critical to our understanding of carcinoma dissemination. Here, we describe a robust method to generate aligned collagen matrices <i>in vitro</i> that mimic <i>in vivo</i> fiber organization. Subsequently, a protocol is presented for seeding aligned matrices with distinct carcinoma cell subpopulations and performing live cell imaging and quantitative analysis of cell migration. Together, the engineered constructs and the imaging techniques laid out here provide a platform to study cancer stem cell migration in 3-D anisotropic collagen with real-time visualization of cellular interactions with the fibrous matrix. © 2018 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":"2018-05-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1002/cpsc.57","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"36243500","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: 5
Fluorescence Endomicroscopy Imaging of Mesenchymal Stem Cells in the Rat Lung 大鼠肺间充质干细胞的荧光内镜成像
Q2 Biochemistry, Genetics and Molecular Biology Pub Date : 2018-05-04 DOI: 10.1002/cpsc.52
Jessica R. Perez, Norma Ybarra, Frederic Chagnon, Olivier Lesur, Jan Seuntjens, Issam El Naqa

Stem cell therapy has shown great promise for organ repair and regeneration. In the context of lung disease, such as radiation-induced lung damage (RILD) in cancer radiotherapy, mesenchymal stem cells (MSCs) have shown the ability to reduce damage possibly due to their immunomodulatory properties and other unknown mechanisms. However, once MSCs are transplanted into the body, little is known as to their localization or their mechanisms of action. In this work, we proposed, implemented, and validated a fluorescence endomicroscopy (FE) imaging technique that allows for the real-time detection and quantification of transplanted pre-labeled MSCs in vivo and tracking in a rat model. This protocol covers aspects related to MSCs extraction, labeling, FE imaging, and image analysis developed in a RILD rat model but applicable to other biological systems. © 2018 by John Wiley & Sons, Inc.

干细胞治疗在器官修复和再生方面显示出巨大的希望。在肺部疾病的背景下,如癌症放疗中的辐射诱导肺损伤(RILD),间充质干细胞(MSCs)已显示出减少损伤的能力,可能是由于其免疫调节特性和其他未知机制。然而,一旦骨髓间充质干细胞移植到体内,对其定位或作用机制知之甚少。在这项工作中,我们提出、实施并验证了一种荧光内镜(FE)成像技术,该技术可以实时检测和定量体内移植的预标记间充质干细胞,并在大鼠模型中进行跟踪。本协议涵盖了在RILD大鼠模型中开发的MSCs提取、标记、FE成像和图像分析相关的方面,但适用于其他生物系统。©2018 by John Wiley &儿子,Inc。
{"title":"Fluorescence Endomicroscopy Imaging of Mesenchymal Stem Cells in the Rat Lung","authors":"Jessica R. Perez,&nbsp;Norma Ybarra,&nbsp;Frederic Chagnon,&nbsp;Olivier Lesur,&nbsp;Jan Seuntjens,&nbsp;Issam El Naqa","doi":"10.1002/cpsc.52","DOIUrl":"10.1002/cpsc.52","url":null,"abstract":"<p>Stem cell therapy has shown great promise for organ repair and regeneration. In the context of lung disease, such as radiation-induced lung damage (RILD) in cancer radiotherapy, mesenchymal stem cells (MSCs) have shown the ability to reduce damage possibly due to their immunomodulatory properties and other unknown mechanisms. However, once MSCs are transplanted into the body, little is known as to their localization or their mechanisms of action. In this work, we proposed, implemented, and validated a fluorescence endomicroscopy (FE) imaging technique that allows for the real-time detection and quantification of transplanted pre-labeled MSCs <i>in vivo</i> and tracking in a rat model. This protocol covers aspects related to MSCs extraction, labeling, FE imaging, and image analysis developed in a RILD rat model but applicable to other biological systems. © 2018 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":"2018-05-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1002/cpsc.52","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"36338591","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: 1
Generation of Human Induced Pluripotent Stem Cells Using a Defined, Feeder-Free Reprogramming System 使用一种明确的、无馈源的重编程系统生成人类诱导多能干细胞
Q2 Biochemistry, Genetics and Molecular Biology Pub Date : 2018-05-04 DOI: 10.1002/cpsc.48
Seonmi Park, Gustavo Mostoslavsky

Human induced pluripotent stem cells (hiPSCs) offer great opportunities for the study of human development and disease modeling and have enormous potential for use in future clinical cell-based therapies. However, most current systems to create hiPSCs often expose the cells to animal feeder layers or xenogeneic reagents; this raises safety concerns about using hiPSC-derived cells for therapeutic purposes. Here, we describe protocols to generate hiPSCs without exposing the cells to xenogeneic materials that uses a defined, feeder-free reprogramming system. With this method, we were able to successfully reprogram not only patient-derived peripheral blood mononuclear cells but also amniocytes from the amniotic fluid of stillborn fetuses using two independent reprogramming platforms. Importantly, hiPSCs generated in this fashion expressed pluripotent markers and had normal karyotypes. The protocols allowed us to generate and culture hiPSCs under Good Manufacturing Practice–like conditions, a necessary step for the future clinical application of these cells. © 2018 by John Wiley & Sons, Inc.

人类诱导多能干细胞(hiPSCs)为人类发育和疾病建模研究提供了巨大的机会,在未来的临床细胞治疗中具有巨大的潜力。然而,目前大多数制造hipsc的系统通常将细胞暴露于动物饲养层或异种试剂中;这引起了使用hipsc衍生细胞用于治疗目的的安全性担忧。在这里,我们描述了在不将细胞暴露于异种材料的情况下生成hipsc的方案,该方案使用定义的,无馈线的重编程系统。通过这种方法,我们不仅能够成功地对患者外周血单核细胞进行重编程,还能够使用两个独立的重编程平台对死胎羊水中的羊膜细胞进行重编程。重要的是,以这种方式产生的hiPSCs表达多能性标记物,并且具有正常的核型。该方案允许我们在类似良好生产规范的条件下生成和培养hipsc,这是这些细胞未来临床应用的必要步骤。©2018 by John Wiley &儿子,Inc。
{"title":"Generation of Human Induced Pluripotent Stem Cells Using a Defined, Feeder-Free Reprogramming System","authors":"Seonmi Park,&nbsp;Gustavo Mostoslavsky","doi":"10.1002/cpsc.48","DOIUrl":"10.1002/cpsc.48","url":null,"abstract":"<p>Human induced pluripotent stem cells (hiPSCs) offer great opportunities for the study of human development and disease modeling and have enormous potential for use in future clinical cell-based therapies. However, most current systems to create hiPSCs often expose the cells to animal feeder layers or xenogeneic reagents; this raises safety concerns about using hiPSC-derived cells for therapeutic purposes. Here, we describe protocols to generate hiPSCs without exposing the cells to xenogeneic materials that uses a defined, feeder-free reprogramming system. With this method, we were able to successfully reprogram not only patient-derived peripheral blood mononuclear cells but also amniocytes from the amniotic fluid of stillborn fetuses using two independent reprogramming platforms. Importantly, hiPSCs generated in this fashion expressed pluripotent markers and had normal karyotypes. The protocols allowed us to generate and culture hiPSCs under Good Manufacturing Practice–like conditions, a necessary step for the future clinical application of these cells. © 2018 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":"2018-05-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1002/cpsc.48","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"36338590","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: 13
Derivation of Epithelial-Only Airway Organoids from Human Pluripotent Stem Cells 从人多能干细胞中衍生上皮性气道类器官
Q2 Biochemistry, Genetics and Molecular Biology Pub Date : 2018-05-04 DOI: 10.1002/cpsc.51
Katherine B. McCauley, Finn Hawkins, Darrell N. Kotton

New protocols to efficiently generate functional airway epithelial organoids from human pluripotent stem cells (PSCs) would represent a major advance towards effective disease modeling, drug screening and cell based therapies for lung disorders. This unit describes an approach using stage-specific signaling pathway manipulation to differentiate cells to proximal airway epithelium via key developmental intermediates. Cells are directed via definitive endoderm (DE) to anterior foregut, and then specified to NKX2-1+ lung epithelial progenitors. These lung progenitors are purified using cell surface marker sorting and replated in defined culture conditions to form three-dimensional, epithelial-only airway organoids. This directed differentiation approach using serum-free, defined media also includes protocols for evaluation of DE induction, intracellular FACS analysis of NKX2-1 specification efficiency and enrichment, and approaches for characterization and expansion of airway organoids. Taken together, this represents an efficient and reproducible approach to generate expandable airway organoids from human PSCs for use in numerous downstream applications. © 2018 by John Wiley & Sons, Inc.

从人类多能干细胞(PSCs)中高效生成功能性气道上皮类器官的新方案将代表着有效的疾病建模、药物筛选和基于细胞的肺部疾病治疗的重大进展。本单元描述了一种使用阶段特异性信号通路操纵的方法,通过关键的发育中间体将细胞分化为近端气道上皮。细胞通过最终内胚层(DE)被引导到前前肠,然后被指定到NKX2-1+肺上皮祖细胞。使用细胞表面标记分选纯化这些肺祖细胞,并在规定的培养条件下复制,形成三维的、仅上皮的气道类器官。这种使用无血清的定向分化方法还包括评估DE诱导的方案,细胞内FACS分析NKX2-1的规范效率和富集,以及表征和扩展气道类器官的方法。综上所述,这代表了一种有效且可重复的方法,可以从人类psc中生成可扩展的气道类器官,用于许多下游应用。©2018 by John Wiley &儿子,Inc。
{"title":"Derivation of Epithelial-Only Airway Organoids from Human Pluripotent Stem Cells","authors":"Katherine B. McCauley,&nbsp;Finn Hawkins,&nbsp;Darrell N. Kotton","doi":"10.1002/cpsc.51","DOIUrl":"10.1002/cpsc.51","url":null,"abstract":"<p>New protocols to efficiently generate functional airway epithelial organoids from human pluripotent stem cells (PSCs) would represent a major advance towards effective disease modeling, drug screening and cell based therapies for lung disorders. This unit describes an approach using stage-specific signaling pathway manipulation to differentiate cells to proximal airway epithelium via key developmental intermediates. Cells are directed via definitive endoderm (DE) to anterior foregut, and then specified to NKX2-1+ lung epithelial progenitors. These lung progenitors are purified using cell surface marker sorting and replated in defined culture conditions to form three-dimensional, epithelial-only airway organoids. This directed differentiation approach using serum-free, defined media also includes protocols for evaluation of DE induction, intracellular FACS analysis of NKX2-1 specification efficiency and enrichment, and approaches for characterization and expansion of airway organoids. Taken together, this represents an efficient and reproducible approach to generate expandable airway organoids from human PSCs for use in numerous downstream applications. © 2018 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":"2018-05-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1002/cpsc.51","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"36337446","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: 40
Methods for Generating Vascularized Islet-Like Organoids Via Self-Condensation 血管化胰岛样器官的自凝生成方法
Q2 Biochemistry, Genetics and Molecular Biology Pub Date : 2018-05-04 DOI: 10.1002/cpsc.49
Yoshinobu Takahashi, Takanori Takebe, Hideki Taniguchi

Despite the promise of emerging organoid-based approaches, building additional complexity, such as the vascular network, remains a major challenge toward regenerative therapy. Recently, we developed a complex organoid engineering method by "self-condensation," wherein mesenchymal cell–dependent contraction enables large-scale condensation from heterotypic multiple progenitors. Here, we describe the adaptation of this protocol for generating three-dimensional (3D) pancreatic condensates from dissociated β cell lines (MIN6) together with blood vessel–forming progenitors. This protocol achieves 3D pancreatic islet-like organoid self-organization with endothelialized networks through mesenchymal stem cell–dependent contraction. Transplantation of pancreatic islet-like organoids treats diabetes in mice effectively. Given the donor shortage associated with clinical islet transplantation, our approach offers a promising alternative toward therapeutic organoid transplantation. © 2018 by John Wiley & Sons, Inc.

尽管新兴的基于类器官的方法有希望,但建立额外的复杂性,如血管网络,仍然是再生治疗的主要挑战。最近,我们通过“自缩聚”开发了一种复杂的类器官工程方法,其中间充质细胞依赖性收缩能够从异型多个祖细胞大规模缩聚。在这里,我们描述了该方案的适应性,从解离的β细胞系(MIN6)和血管形成祖细胞一起产生三维(3D)胰腺凝析液。该方案通过间充质干细胞依赖性收缩实现具有内皮化网络的三维胰岛样器官自组织。胰岛样器官移植可有效治疗小鼠糖尿病。鉴于与临床胰岛移植相关的供体短缺,我们的方法为治疗性类器官移植提供了一个有希望的替代方法。©2018 by John Wiley &儿子,Inc。
{"title":"Methods for Generating Vascularized Islet-Like Organoids Via Self-Condensation","authors":"Yoshinobu Takahashi,&nbsp;Takanori Takebe,&nbsp;Hideki Taniguchi","doi":"10.1002/cpsc.49","DOIUrl":"10.1002/cpsc.49","url":null,"abstract":"<p>Despite the promise of emerging organoid-based approaches, building additional complexity, such as the vascular network, remains a major challenge toward regenerative therapy. Recently, we developed a complex organoid engineering method by \"self-condensation,\" wherein mesenchymal cell–dependent contraction enables large-scale condensation from heterotypic multiple progenitors. Here, we describe the adaptation of this protocol for generating three-dimensional (3D) pancreatic condensates from dissociated β cell lines (MIN6) together with blood vessel–forming progenitors. This protocol achieves 3D pancreatic islet-like organoid self-organization with endothelialized networks through mesenchymal stem cell–dependent contraction. Transplantation of pancreatic islet-like organoids treats diabetes in mice effectively. Given the donor shortage associated with clinical islet transplantation, our approach offers a promising alternative toward therapeutic organoid transplantation. © 2018 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":"2018-05-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1002/cpsc.49","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"36338596","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: 21
CRISPR/Cas9-based Targeted Genome Editing for the Development of Monogenic Diseases Models with Human Pluripotent Stem Cells 基于CRISPR/ cas9的靶向基因组编辑用于人类多能干细胞单基因疾病模型的开发
Q2 Biochemistry, Genetics and Molecular Biology Pub Date : 2018-04-26 DOI: 10.1002/cpsc.50
Navin Gupta, Koichiro Susa, Yoko Yoda, Joseph V. Bonventre, M. Todd Valerius, Ryuji Morizane

Human pluripotent stem cells (hPSCs) represent a formidable tool for disease modeling, drug discovery, and regenerative medicine using human cells and tissues in vitro. Evolving techniques of targeted genome editing, specifically the CRISPR/Cas9 system, allow for the generation of cell lines bearing gene-specific knock-outs, knock-in reporters, and precise mutations. However, there are increasing concerns related to the transfection efficiency, cell viability, and maintenance of pluripotency provided by genome-editing techniques. The procedure presented here employs transient antibiotic selection that overcomes reduced transfection efficiency, avoids cytotoxic flow sorting for increased viability, and generates multiple genome-edited pluripotent hPSC lines expanded from a single parent cell. Avoidance of xenogeneic contamination from feeder cells and reduced operator workload, owing to single-cell passaging rather than clump passaging, are additional benefits. The outlined methods may enable researchers with limited means and technical experience to create human stem cell lines containing desired gene-specific mutations. © 2018 by John Wiley & Sons, Inc.

人类多能干细胞(hPSCs)是一种强大的工具,用于疾病建模、药物发现和体外使用人类细胞和组织的再生医学。不断发展的靶向基因组编辑技术,特别是CRISPR/Cas9系统,允许产生具有基因特异性敲除、敲入报告基因和精确突变的细胞系。然而,人们越来越关注基因组编辑技术提供的转染效率、细胞活力和多能性的维持。本文介绍的程序采用瞬时抗生素选择,克服了转染效率降低的问题,避免了细胞毒性流动分选以提高生存能力,并从单个亲本细胞扩增产生多个基因组编辑的多能性hPSC系。由于采用单细胞传代而不是成团传代,避免了饲养细胞的异种污染,减少了操作员的工作量,这些都是额外的好处。概述的方法可能使研究人员在有限的手段和技术经验下创建包含所需基因特异性突变的人类干细胞系。©2018 by John Wiley &儿子,Inc。
{"title":"CRISPR/Cas9-based Targeted Genome Editing for the Development of Monogenic Diseases Models with Human Pluripotent Stem Cells","authors":"Navin Gupta,&nbsp;Koichiro Susa,&nbsp;Yoko Yoda,&nbsp;Joseph V. Bonventre,&nbsp;M. Todd Valerius,&nbsp;Ryuji Morizane","doi":"10.1002/cpsc.50","DOIUrl":"10.1002/cpsc.50","url":null,"abstract":"<p>Human pluripotent stem cells (hPSCs) represent a formidable tool for disease modeling, drug discovery, and regenerative medicine using human cells and tissues <i>in vitro</i>. Evolving techniques of targeted genome editing, specifically the CRISPR/Cas9 system, allow for the generation of cell lines bearing gene-specific knock-outs, knock-in reporters, and precise mutations. However, there are increasing concerns related to the transfection efficiency, cell viability, and maintenance of pluripotency provided by genome-editing techniques. The procedure presented here employs transient antibiotic selection that overcomes reduced transfection efficiency, avoids cytotoxic flow sorting for increased viability, and generates multiple genome-edited pluripotent hPSC lines expanded from a single parent cell. Avoidance of xenogeneic contamination from feeder cells and reduced operator workload, owing to single-cell passaging rather than clump passaging, are additional benefits. The outlined methods may enable researchers with limited means and technical experience to create human stem cell lines containing desired gene-specific mutations. © 2018 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":"2018-04-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1002/cpsc.50","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"36337445","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: 9
Stem Cell–Derived Retinal Pigment Epithelial Layer Model from Adult Human Globes Donated for Corneal Transplants 角膜移植捐献成人眼球干细胞源性视网膜色素上皮层模型
Q2 Biochemistry, Genetics and Molecular Biology Pub Date : 2018-04-26 DOI: 10.1002/cpsc.53
Marie Fernandes, Brian McArdle, Lauren Schiff, Timothy A. Blenkinsop

An adult human retinal pigment epithelial layer (ahRPE) model derived from stem cells isolated from native RPE monolayers (ahRPE-SCs) exhibits key physiological characteristics of native tissue and therefore provides the means to create a human “disease in a dish” model to study RPE diseases. Traditionally, RPE lines are established from whole globes dedicated to research. Here we describe a new technique for establishing primary RPE lines from the posterior poles of globes used for corneal transplants. Since tissues from corneal transplants are derived from younger and healthier donors than those used for research, we have hypothesized that RPE cells isolated from corneal transplantation globes will result in improved primary RPE line establishment. Our new procedure increases the rate of establishing successful RPE cultures and improves the total cell number yield. Use of this advanced methodology can provide a new source of high-quality primary RPE line cultures. © 2018 by John Wiley & Sons, Inc.

成人视网膜色素上皮层(ahRPE)模型来源于从天然视网膜色素单层(ahRPE- scs)分离的干细胞,显示出天然组织的关键生理特征,因此提供了创建人类“培养皿疾病”模型来研究RPE疾病的手段。传统上,RPE生产线是在全球范围内建立的,致力于研究。在这里,我们描述了一种从用于角膜移植的球体后极建立原发性RPE线的新技术。由于角膜移植的组织来自于比研究中使用的更年轻、更健康的供体,我们假设从角膜移植球中分离的RPE细胞将改善初级RPE系的建立。我们的新程序增加了建立成功的RPE培养的比率,并提高了总细胞数量的产量。使用这种先进的方法可以提供高质量的原生RPE系培养的新来源。©2018 by John Wiley &儿子,Inc。
{"title":"Stem Cell–Derived Retinal Pigment Epithelial Layer Model from Adult Human Globes Donated for Corneal Transplants","authors":"Marie Fernandes,&nbsp;Brian McArdle,&nbsp;Lauren Schiff,&nbsp;Timothy A. Blenkinsop","doi":"10.1002/cpsc.53","DOIUrl":"10.1002/cpsc.53","url":null,"abstract":"<p>An adult human retinal pigment epithelial layer (ahRPE) model derived from stem cells isolated from native RPE monolayers (ahRPE-SCs) exhibits key physiological characteristics of native tissue and therefore provides the means to create a human “disease in a dish” model to study RPE diseases. Traditionally, RPE lines are established from whole globes dedicated to research. Here we describe a new technique for establishing primary RPE lines from the posterior poles of globes used for corneal transplants. Since tissues from corneal transplants are derived from younger and healthier donors than those used for research, we have hypothesized that RPE cells isolated from corneal transplantation globes will result in improved primary RPE line establishment. Our new procedure increases the rate of establishing successful RPE cultures and improves the total cell number yield. Use of this advanced methodology can provide a new source of high-quality primary RPE line cultures. © 2018 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":"2018-04-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1002/cpsc.53","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"36337447","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: 12
Derivation of Multipotent Mesenchymal Stromal Cells from Ovine Bone Marrow 绵羊骨髓多能间充质间质细胞的制备
Q2 Biochemistry, Genetics and Molecular Biology Pub Date : 2018-02-28 DOI: 10.1002/cpsc.43
Daniel Vivas, Marta Caminal, Irene Oliver-Vila, Joaquim Vives

In the field of orthopedics, translational research of novel therapeutic approaches involves the use of large animal models (such as sheep, goat, pig, dog, and horse) due to the similarities with humans in weight, size, joint structure, and bone/cartilage healing mechanisms. Particularly in the development of cell-based therapies, the lack of manageable immunocompromised preclinical large animal models prevents the use of human cells, which makes it necessary to produce equivalent homologous cell types for the study of their pharmacodynamics, pharmacokinetics, and toxicology. The methods described herein allow for the isolation, expansion, manipulation, and characterization of fibroblastic-like ovine bone marrow–derived multipotent mesenchymal stromal cells (BM-MSC) that, similar to human BM-MSC, adhere to standard plastic surfaces; express specific surface markers such as CD44, CD90, CD140a, CD105, and CD166; and display trilineage differentiation potential in vitro. Homogeneous cell cultures result from a 3-week bioprocess yielding cell densities in the range of 2–4 × 104 MSC/cm2 at passage 2, which corresponds to ∼8 cumulative population doublings. Large quantities of BM-MSC resulting from following this methodology can be readily used in proof of efficacy and safety studies in the preclinical development stage. © 2018 by John Wiley & Sons, Inc.

在骨科领域,新型治疗方法的转化研究涉及使用大型动物模型(如绵羊、山羊、猪、狗和马),因为它们在体重、大小、关节结构和骨/软骨愈合机制方面与人类相似。特别是在细胞疗法的发展中,缺乏可管理的免疫功能受损的临床前大型动物模型阻碍了人类细胞的使用,这使得有必要生产等效的同源细胞类型以研究其药效学,药代动力学和毒理学。本文描述的方法允许分离、扩增、操作和表征成纤维样羊骨髓来源的多能间充质间质细胞(BM-MSC),类似于人BM-MSC,粘附在标准塑料表面;表达特异性表面标记物,如CD44、CD90、CD140a、CD105和CD166;并在体外表现出三龄分化潜能。经过3周的生物过程,均匀的细胞培养在传代2时产生2 - 4 × 104 MSC/cm2范围内的细胞密度,相当于8倍的累积群体。遵循这种方法产生的大量的骨髓间质干细胞可以很容易地用于临床前开发阶段的有效性和安全性研究的证明。©2018 by John Wiley &儿子,Inc。
{"title":"Derivation of Multipotent Mesenchymal Stromal Cells from Ovine Bone Marrow","authors":"Daniel Vivas,&nbsp;Marta Caminal,&nbsp;Irene Oliver-Vila,&nbsp;Joaquim Vives","doi":"10.1002/cpsc.43","DOIUrl":"10.1002/cpsc.43","url":null,"abstract":"<p>In the field of orthopedics, translational research of novel therapeutic approaches involves the use of large animal models (such as sheep, goat, pig, dog, and horse) due to the similarities with humans in weight, size, joint structure, and bone/cartilage healing mechanisms. Particularly in the development of cell-based therapies, the lack of manageable immunocompromised preclinical large animal models prevents the use of human cells, which makes it necessary to produce equivalent homologous cell types for the study of their pharmacodynamics, pharmacokinetics, and toxicology. The methods described herein allow for the isolation, expansion, manipulation, and characterization of fibroblastic-like ovine bone marrow–derived multipotent mesenchymal stromal cells (BM-MSC) that, similar to human BM-MSC, adhere to standard plastic surfaces; express specific surface markers such as CD44, CD90, CD140a, CD105, and CD166; and display trilineage differentiation potential in vitro. Homogeneous cell cultures result from a 3-week bioprocess yielding cell densities in the range of 2–4 × 10<sup>4</sup> MSC/cm<sup>2</sup> at passage 2, which corresponds to ∼8 cumulative population doublings. Large quantities of BM-MSC resulting from following this methodology can be readily used in proof of efficacy and safety studies in the preclinical development stage. © 2018 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":"2018-02-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1002/cpsc.43","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"35889246","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: 21
Conditional Manipulation of Gene Function in Human Cells with Optimized Inducible shRNA 利用优化的可诱导shRNA条件操纵人细胞的基因功能
Q2 Biochemistry, Genetics and Molecular Biology Pub Date : 2018-02-28 DOI: 10.1002/cpsc.45
Alessandro Bertero, Loukia Yiangou, Stephanie Brown, Daniel Ortmann, Matthias Pawlowski, Ludovic Vallier

The difficulties involved in conditionally perturbing complex gene expression networks represent major challenges toward defining the mechanisms controlling human development, physiology, and disease. We developed an OPTimized inducible KnockDown (OPTiKD) platform that addresses the limitations of previous approaches by allowing streamlined, tightly-controlled, and potent loss-of-function experiments for both single and multiple genes. The method relies on single-step genetic engineering of the AAVS1 genomic safe harbor with an optimized tetracycline-responsive cassette driving one or more inducible short hairpin RNAs (shRNAs). OPTiKD provides homogeneous, dose-responsive, and reversible gene knockdown. When implemented in human pluripotent stem cells (hPSCs), the approach can be then applied to a broad range of hPSC-derived mature cell lineages that include neurons, cardiomyocytes, and hepatocytes. Generation of OPTiKD hPSCs in commonly used culture conditions is simple (plasmid based), rapid (two weeks), and highly efficient (>95%). Overall, this method facilitates the functional annotation of the human genome in health and disease. © 2018 by John Wiley & Sons, Inc.

有条件地干扰复杂基因表达网络所涉及的困难代表了定义控制人类发育、生理和疾病的机制的主要挑战。我们开发了一种优化的可诱导敲除(OPTiKD)平台,通过简化、严格控制和有效的单基因和多基因功能丧失实验,解决了以前方法的局限性。该方法依赖于AAVS1基因组安全港的单步基因工程,利用优化的四环素反应盒驱动一个或多个诱导短发夹rna (shRNAs)。OPTiKD提供均匀、剂量响应和可逆的基因敲除。当在人类多能干细胞(hPSCs)中实施时,该方法可以应用于广泛的hpsc衍生的成熟细胞系,包括神经元、心肌细胞和肝细胞。在常用的培养条件下,OPTiKD hPSCs的生成简单(质粒为基础)、快速(两周)和高效(95%)。总的来说,这种方法促进了人类基因组在健康和疾病中的功能注释。©2018 by John Wiley &儿子,Inc。
{"title":"Conditional Manipulation of Gene Function in Human Cells with Optimized Inducible shRNA","authors":"Alessandro Bertero,&nbsp;Loukia Yiangou,&nbsp;Stephanie Brown,&nbsp;Daniel Ortmann,&nbsp;Matthias Pawlowski,&nbsp;Ludovic Vallier","doi":"10.1002/cpsc.45","DOIUrl":"10.1002/cpsc.45","url":null,"abstract":"<p>The difficulties involved in conditionally perturbing complex gene expression networks represent major challenges toward defining the mechanisms controlling human development, physiology, and disease. We developed an OPTimized inducible KnockDown (OPTiKD) platform that addresses the limitations of previous approaches by allowing streamlined, tightly-controlled, and potent loss-of-function experiments for both single and multiple genes. The method relies on single-step genetic engineering of the AAVS1 genomic safe harbor with an optimized tetracycline-responsive cassette driving one or more inducible short hairpin RNAs (shRNAs). OPTiKD provides homogeneous, dose-responsive, and reversible gene knockdown. When implemented in human pluripotent stem cells (hPSCs), the approach can be then applied to a broad range of hPSC-derived mature cell lineages that include neurons, cardiomyocytes, and hepatocytes. Generation of OPTiKD hPSCs in commonly used culture conditions is simple (plasmid based), rapid (two weeks), and highly efficient (&gt;95%). Overall, this method facilitates the functional annotation of the human genome in health and disease. © 2018 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":"2018-02-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1002/cpsc.45","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"35890119","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: 9
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