Induced pluripotent stem cells

合集下载

2020脓毒症致心肌损伤模型的研究进展

2020脓毒症致心肌损伤模型的研究进展
2007 年,Takahashi 等[22]利用病毒载体将四个转录因子转入人成体 细胞,使其重编程而得到诱导多潜能干细胞(induced pluripotent stem cells,iPSCs)。这些干细胞在特定微环境或小分子诱导剂的作用下[23], 可以向心肌细胞分化,即 hPSC-CMs,这一突破为心血管疾病人源化细胞 模型的建立奠定坚实基础。Yücel 等[24]用 LPS 处理 hPSC-CMs,6 h 后 促炎因子和趋化因子产生显著增多;而处理 48 h 后抗炎因子开始升高。 同时还发现,LPS 影响 hPSC-CMs 离子通道蛋白表达,延长动作电位时 长,导致心肌细胞电生理功能异常,证实 hPSC-CMs 具有内毒素诱导的 炎症反应系统,可以用来模拟某些脓毒症或细菌感染诱导的心肌细胞炎症 反应。 4 各模型优缺点
目前,SIMI 动物模型的研究与应用已经相对成熟。制备 SIMI 的动物 模型主要有三种方法:宿主屏障破坏,毒素注射和病原体注射。 2.1 宿主屏障破坏模型
盲肠结扎穿孔术(cecal ligation and puncture,CLP)即通过人为的盲 肠结扎和穿刺诱发多菌性腹膜炎,动物会出现典型脓毒症症状。在此脓毒
1976 年,Kimes 等[15]从胚胎期 BDIX 大鼠心室组织中获得可以体外 培养增殖的 H9c2 心肌细胞株。H9c2 心肌细胞可以传代分化,在低血清 培养基中向骨骼肌分化,而在维甲酸诱导下则呈现心肌细胞表型,但此株 细胞即使分化,依然缺乏心肌细胞样的节律性搏动。研究发现体外培养的 H9c2 细胞在 LPS 处理下出现明显的炎症反应,为利用 H9c2 细胞建立 SIMI 模型提供基础。此后研究发现黄芪多糖通过下调 miR-127、大黄素 通过下调 miR-223 的表达来改善 LPS 诱导的心肌损伤,提示 microRNA 可能是 SIMI 治疗的一个重要靶点[16]。 3.2.2 小鼠心房 HL-1 细胞株

当今干细胞研究方面地10位顶尖科学家

当今干细胞研究方面地10位顶尖科学家

排名第一:Shinya Yamanaka和JamesA.Thomson博士是建立了可诱导的万能干细胞,在干细胞再生和分化重排机理上做出了最具突破性的进展性工作;这毫无疑问是诺贝尔奖级的工作,其他几位平时工作很杰出,可是没有这种级别的工作,只好屈居次位;排名第二:Rudolf Jaenisch博士长期从事于干细胞核的替代重组和干细胞的表观遗传修饰工作,卓有成绩,这也是培养诱导干细胞的核心工作,重要性无人能代替;排名第三: Rebort lanza博士领导和指挥着全球最领先的干细胞生物技术公司,独创和建立了分离和培养单个胚胎干细胞的方法和技术。

主编了所有重要的干细胞参考书籍。

每一相重大干细胞技术的出现,美欧主流媒体都要听他的意见,可谓干细胞领域的大腕人物;排名第四:Alan Trounson博士是国际免疫学和干细胞研究的先驱者,领导和指挥原澳大利亚Monash大学免疫学和干细胞研究实验室,使Monash大学成为世界上最成功的大学之一。

手下的弟子Martin Perl博士出任南加州大学第一界干细胞和系统生物学所所长。

2007年成为美国眼下资金最多,实力最强的加州再生医学研究研究所所长,成为美国干细胞研究中最大的老板;排名第五:哈佛大学干细胞研究所所长Douglas A. Melton博士和斯坦福大学干细胞和再生医学研究所所长Irving L. Weissman博士两人都是干细胞研究领域的顶尖高手,又各了带领着东西两岸这两所美国奈至全世界的顶尖学府的干细胞研究的竟赛。

排名第六:哈佛大学干细胞研究所共同所长David T. Scadden, 博士和密西根大学干细胞中心主Sean Morrison博士两人是干细胞研究的中青年骨干,专长于干细胞分化再生的微环境调控机理的研究,. Scadden, 博士是麻省总医院再生医学研究所所长,侧重于干细胞的临床应用。

Morrison博士则是休斯医学研究所研究员,是美国中西部大学中干细胞研究的顶级人物。

诱导型多能干细胞鉴定标准

诱导型多能干细胞鉴定标准

诱导型多能干细胞鉴定标准介绍在过去的几十年里,科学家们一直努力寻找一种能够在实验室中培养的多能干细胞,以代替胚胎干细胞的应用。

诱导型多能干细胞(induced pluripotent stem cells, iPSCs)的发现,为此目标的实现提供了新的可能性。

然而,为了确保iPSCs的正确鉴定和应用,需要制定一套严格的鉴定标准。

重要性鉴定标准的制定对于保证iPSCs研究的准确性和可重复性至关重要。

只有通过严格的鉴定标准,才能确保iPSCs的多能性、稳定性和可应用性。

此外,鉴定标准的制定还可以促进iPSCs研究的国际合作和信息共享,推动该领域的进一步发展。

与鉴定标准相关的因素制定iPSCs的鉴定标准需要考虑以下几个因素:多能性指标多能性是iPSCs的关键特征之一。

因此,鉴定标准需要包括对多能性的检测方法,如基因表达分析、细胞分化能力评估等。

克隆稳定性iPSCs的克隆稳定性是其应用的基础。

鉴定标准应包括对iPSCs的克隆稳定性的评估方法,如基因组稳定性分析、细胞系传代能力评估等。

高质量的细胞文库鉴定标准还应包括对iPSCs的质量要求。

只有在具备一定质量基础上的iPSCs才能被广泛应用。

因此,鉴定标准应包括细胞文库的建立和管理方法。

鉴定标准的可追溯性为了确保鉴定标准的有效性和可追溯性,需要制定明确的实验流程和数据分析方法。

此外,鉴定标准的制定还应包括数据的共享和存储规范。

诱导型多能干细胞鉴定标准的制定为了确保诱导型多能干细胞鉴定标准的全面性和有效性,应采取以下步骤:成立专家组应成立一个包括多个相关领域专家的工作组,以确保鉴定标准的全面性和权威性。

该工作组应包括基础研究、临床应用和伦理等各个方面的专家。

文献综述和经验总结工作组应对已有的文献进行综述和总结,了解当前iPSCs鉴定方法的优缺点,为鉴定标准的制定提供参考。

此外,工作组还应汇总各个实验室的经验和实践,了解当前实验室中iPSCs鉴定的常见问题和挑战。

iPS细胞简述

iPS细胞简述

iPS细胞基本概念诱导多能干细胞(induced pluripotent stem cells, iPS cells)最初是日本人山中申弥(Shinya Yamanaka)于2006年利用病毒载体将四个转录因子(Oct4, Sox2, Klf4 和c-Myc)的组合转入分化的体细胞中,使其重编程而得到的类似胚胎干细胞的一种细胞类型[1]。

随后世界各地不同科学家陆续发现其他方法同样也可以制造这种细胞。

细胞可分为全能干细胞、多能干细胞和单能干细胞,诱导多能干细胞即通过向体细胞中导入诱导基因,使体细胞重编程获得具有胚胎干细胞样特性的多能干细胞,也称为去分化。

发展趋势自2008年开始,小鼠和人的ips细胞研究取得了极大的发展,并在体细胞的选择、转录因子的组合和数量、病毒载体、筛选条件和小分子化合物等方面进行了完善,还逐步向疾病的治疗方面深入,镰刀形红细胞贫血症治疗的研究,从理论和实践上为人类单基因遗传病的治疗奠定基础。

此外,猴、大鼠和猪ips细胞的建立在人类疾病研究上具有重大应用价值,既可构建人类疾病的动物模型,又能进行细胞移植实验,为将来的实验细胞替代治疗奠定基础。

2009年3月,Nagy小组和Kaji小组采用转座子法取代病毒载体的基因投递的方法,高效率制备了基因组无病毒整合的鼠iPS细胞,获得iPS细胞后,他们又成功将先前导入的转录因子基因从iPS细胞中移除。

随着越来越多研究模型的建立,越来越多的体细胞被成功诱导为多能干细胞,以及无病毒整合技术的发现和有害基因的成功敲除,使得诱导多能干细胞向更加系统,更加安全,更加高效的方向发展。

伴随着ips细胞被诱导分化成为多巴胺神经元、能分泌胰岛素的细胞、心脏实质细胞、视网膜的前驱细胞和神经前体细胞等的成功,越来越多的体细胞得意诱导分化成功,为患帕金森综合症、糖尿病、心血管疾病和视网膜病变等疾病的患者提供了治愈的可能。

面临问题第一,诱导体细胞重编程的分子机制至今仍不清楚。

ipsc分化为rpe的方法

ipsc分化为rpe的方法

ipsc分化为rpe的方法IPSC分化为RPE的方法简介在干细胞研究领域,将人诱导多能干细胞(induced pluripotent stem cells,简称iPSC)分化为视网膜色素上皮(retinal pigment epithelium,简称RPE)细胞是一个具有重要意义的研究方向。

RPE细胞在视网膜功能维持中起着重要的作用。

本文将介绍一些常用的方法,用于IPSC分化为RPE细胞。

方法一:原代细胞培养在过去的研究中,研究人员采用了原代细胞培养的方法来分化IPSC为RPE细胞。

这种方法的优点在于可以直接从组织中获得RPE细胞,并且维持了RPE细胞的天然特性。

但是,原代细胞培养方法存在许多挑战,如细胞来源有限、细胞增殖缓慢等。

方法二:向下分化法向下分化法是一种常用的方法,可将IPSC分化为RPE细胞。

该方法利用特定的培养条件和信号通路调控,将IPSC逐步分化为RPE细胞。

这种方法的优点在于可以控制分化过程中的细胞品质和数量,但是也需要精确调控培养条件和信号通路。

单因子向下分化法单因子向下分化法是指通过添加特定的因子或生长因子,来促进IPSC向RPE细胞的分化。

常用的因子包括视黄醇、肝细胞生长因子等。

这些因子的选择和时间控制对分化的效果有重要影响。

多因子向下分化法多因子向下分化法是通过联合使用多个因子,来实现更精确的IPSC向RPE细胞的分化。

这种方法的优点在于可以模拟胚胎发育过程,提高分化效率和细胞品质。

方法三:遗传改造法遗传改造法是通过基因编辑技术,对IPSC进行遗传改造,使其能够直接分化为RPE细胞。

常用的基因编辑技术包括CRISPR-Cas9、ZFN、TALEN等。

这种方法的优点在于可以直接转录特定的RPE细胞转录因子,从而实现高效的分化。

方法四:重编程法重编程法是将IPSC重新编程为RPE细胞的一种方法。

通过转录因子的表达,在IPSC中激活RPE细胞特定基因的表达,从而实现分化。

该方法的优点在于不需要添加外源性因子,但是需要精确控制转录因子的表达时间和水平。

干细胞的研究进展及应用前景

干细胞的研究进展及应用前景

医药·保健干细胞的研究进展及应用前景王晓瑞1李薇1顾恩妍2张慧1胡桂1(1、昆明医科大学海源学院,云南昆明6501062、北京吉源干细胞医学研究院,北京101318)现今,干细胞的研究越来越被重视,干细胞技术发展迅速,已从基础医学研究扩展到了临床应用研究,在生殖系统疾病、神经系统疾病、组织损伤性疾病等的治疗方面已取得了显著的进展[1]。

干细胞是一种特殊细胞,它具有自我更新能力、多向分化能力、可植入能力及组织重建能力等特征,它既可以通过细胞分裂维持自身群体的稳定,又可以分化成为不同类型细胞,进而构成机体各种复杂的组织器官[2]。

干细胞的研究不仅为生物学和基础医学提供了更深入的视角,而且为临床上对于很多疾病的治疗提供了新的思路,带来了新的希望。

1干细胞的定义及特点目前,根据干细胞的来源可将干细胞分为胚胎干细胞和成体干细胞两大类。

胚胎干细胞,被誉为全能性干细胞,理论上讲,无论在体内还是体外环境都可以诱导分化为机体中的所有细胞类型,在适当的条件下它们甚至可以发育为一个有机体。

成体干细胞,是存在于发育成熟个体内已分化组织中的未分化细胞,它具有自我更新能力并能分化为其所在组织起源的所有细胞类型。

而诱导性多能干细胞(iPS 细胞)是源于成熟体细胞诱导演变成具有胚胎干细胞的全能分化潜能细胞,归在哪一类尚存争议。

1.1胚胎干细胞(embryonic stem cell ,ESCs ,简称ES 或EK 细胞),是由胚胎内细胞团或原始生殖细胞经体外抑制培养而筛选出的细胞,它具有体外培养无限增殖、自我更新和多向分化的特性,此外,胚胎干细胞保持着高的端粒酶活性和正常细胞信号传导途径,可以快速增殖。

1.2成体干细胞,是存在于发育成熟个体内已分化组织中的未分化细胞,它具有自我更新能力并能分化为其所在组织起源的所有细胞类型。

有造血干细胞、神经干细胞、间充质干细胞等多种类型。

最新的研究表明成体干细胞不仅能分化为特定谱系细胞,还能分化成为在发育上无关的其他谱系细胞,这提示成体干细胞具有较大的分化潜能,可在组织修复等多种疾病的治疗中发挥重要的作用[3]。

Induction of Pluripotent Stem Cells from Mouse Embryonic and Adult Fibroblast Cultures by Defined Fa

Induction of Pluripotent Stem Cells from Mouse Embryonic and Adult Fibroblast Cultures by Defined Fa

实验摘要
Differentiated cells can be reprogrammed to an embryonic-like state by transfer of nuclear contentsinto oocytes or by fusion with embryonic stem (ES) cells. Little is known about factors that induce this reprogramming. Here, we demonstrate induction of pluripotent stem cells from mouse embryonic or adult fibroblasts by introducing four factors, Oct3/4, Sox2, c-Myc,and Klf4, under ES cell culture conditions. Unexpectedly, Nanog was dispensable. These cells, which we designated iPS (induced pluripotent stem) cells, exhibit the morphology and growth properties of ES cells and express ES cell marker genes. Subcutaneous transplantation of iPS cells into nude mice resulted in tumors containing a variety of tissues from all three germ layers. Following injection into blastocysts, iPS cells contributed to mouse embryonic development. These data demonstrate that pluripotent stem cells can be directly generated from fibroblast cultures by the addition of only a few defined factors.

干细胞英语词汇汇总

干细胞英语词汇汇总

干细胞英语词汇汇总Stem Cell Terminology and Key Concepts.Stem cells are a fascinating area of biomedicalresearch with巨大的潜力 for treating various diseases and conditions. To understand stem cell research and its applications, it's important to familiarize oneself withthe terminology and key concepts related to this field.Here is a comprehensive glossary of stem cell-related terms:1. Stem Cell: A cell with the ability to renew itself through mitotic cell division and differentiate into a diverse range of specialized cell types.2. Embryonic Stem Cell (ESC): Stem cells derived fromthe inner cell mass of a blastocyst, which is a very early stage embryo. ESCs have the ability to proliferate indefinitely while maintaining their pluripotent potential.3. Induced Pluripotent Stem Cell (iPSC): A type of stemcell generated from a differentiated cell through genetic reprogramming, usually by introducing specific transcription factors. iPSCs share many properties with embryonic stem cells.4. Adult Stem Cell: Stem cells found in adult tissues that maintain and repair the tissue throughout life. These cells are typically multipotent, meaning they can differentiate into a limited number of cell types.5. Pluripotent Stem Cell: A stem cell that can differentiate into any cell type in the body, except for the placental tissues. Embryonic stem cells and induced pluripotent stem cells are examples of pluripotent stem cells.6. Totipotent Stem Cell: A stem cell that has the potential to develop into an entire organism. This type of stem cell is only found in the very earliest stages of embryonic development.7. Differentiation: The process by which a stem celltransforms into a specialized cell type with a distinct function and structure.8. Self-Renewal: The ability of stem cells to divide and produce more stem cells of the same type. This property maintains the stem cell pool and allows for continuous tissue regeneration.9. Lineage Commitment: The point at which a stem cell irreversibly commits to differentiating into a particular cell type or lineage.10. Germline Stem Cell: A stem cell that has the potential to contribute to the germline, which gives rise to eggs and sperm. These cells are responsible for passing genetic information from one generation to the next.11. Tissue Stem Cell: A stem cell that maintains and repairs a specific tissue in the adult body. Tissue stem cells are typically found in specialized niches within the tissue where they reside.12. Stem Cell Niche: The microenvironment within a tissue that supports stem cell maintenance, proliferation, and differentiation. The niche provides signals that regulate stem cell behavior.13. Pluripotency Factors: Genes or proteins that are essential for maintaining the pluripotent state of stem cells. These factors are often transcription factors that regulate gene expression.14. Ethical Considerations: The ethical implications of stem cell research and therapy, including issues related to embryo destruction, informed consent, and the use of stem cells in medical applications.15. Regenerative Medicine: The field of medicine that aims to replace or regenerate damaged or diseased cells, tissues, and organs using stem cells and other biological materials.16. Stem Cell Therapy: The use of stem cells to treat diseases or conditions by replacing damaged cells,secreting therapeutic factors, or modulating the immune system.17. Transplantation: The process of introducing stem cells into a patient's body to treat a disease or condition. Transplantation can involve autologous (patient's own) or allogeneic (donor) stem cells.18. Cellular Therapies: Therapeutic approaches that involve the use of cells, either alone or in combinationwith other therapeutic modalities, to treat diseases or conditions. Stem cell-based therapies are a subset ofcellular therapies.19. Cell Culture: The process of growing andmaintaining cells in a controlled environment outside ofthe body, typically in a laboratory setting. Cell cultureis essential for stem cell research and therapy development.20. Genetic Engineering: The manipulation of genetic material to modify the traits or characteristics of an organism. In stem cell research, genetic engineering can beused to create induced pluripotent stem cells or modify stem cell behavior.These terms provide a foundation for understanding stem cell biology, research, and potential applications in medicine. As the field continues to evolve, it's important to stay up-to-date with new terminology and concepts that emerge.。

最新induced pluripotent stem (ips) cells诱导多能干细胞(ips细

最新induced pluripotent stem (ips) cells诱导多能干细胞(ips细

The fusion experiments by Tada, Surani and colleagues clearly showed that ES cells and embryonic germ cells contain factors that can induce reprogramming and pluripotency in somatic cells*.
Before 2006, the prevailing view was that nuclear reprogramming to a pluripotent state is a highly complex process that entail the cooperation of up to 100 factors**.
Gene-Expression Profiles of iPS Cells
Induction of Pluripotent Stem Cells from Mouse Embryonic and Adult Fibroblast Cultures by Defined Factors Kazutoshi Takahashi and Shinya Yamanaka Cell 126, 663–676, August 25, 2006
Generation of germline-competent induced pluripotent stem cells Keisuke Okita, Tomoko Ichisaka & Shinya Yamanaka Vol 448| 19 July 2007| doi:10.1038/nature05934
they are different with regards to gene expression and DNA methylation patterns, and fail to produce adult chimaeras.

当今干细胞研究方面的10位顶尖科学家

当今干细胞研究方面的10位顶尖科学家

排名第一:ShinyaYamanaka和JamesA.Thomson博士是建立了可诱导的万能干细胞,在干细胞再生和分化重排机理上做出了最具突破性的进展性工作;这毫无疑问是诺贝尔奖级的工作,其他几位平时工作很杰出,可是没有这种级别的工作,只好屈居次位;排名第二:RudolfJaenisch博士长期从事于干细胞核的替代重组和干细胞的表观遗传修饰工作,卓有成绩,这也是培养诱导干细胞的核心工作,重要性无人能代替;排名第三:Rebortlanza博士领导和指挥着全球最领先的干细胞生物技术公司,独创和建立了分离和培养单个胚胎干细胞的方法和技术。

主编了所有重要的干细胞参考书籍。

每一相重大干细胞技术的出现,美欧主流媒体都要听他的意见,可谓干细胞领域的大腕人物;排名第四:AlanTrounson博士是国际免疫学和干细胞研究的先驱者,领导和指挥原澳大利亚Monash 大学免疫学和干细胞研究实验室,使Monash大学成为世界上最成功的大学之一。

手下的弟子MartinPerl博士出任南加州大学第一界干细胞和系统生物学所所长。

2007年成为美国眼下资金最多,实力最强的加州再生医学研究研究所所长,成为美国干细胞研究中最大的老板;排名第五:哈佛大学干细胞研究所所长DouglasA.Melton博士和斯坦福大学干细胞和再生医学研究所所长IrvingL.Weissman博士两人都是干细胞研究领域的顶尖高手,又各了带领着东西两岸这两所美国奈至全世界的顶尖学府的干细胞研究的竟赛。

排名第六:哈佛大学干细胞研究所共同所长DavidT.Scadden,博士和密西根大学干细胞中心主SeanMorrison博士两人是干细胞研究的中青年骨干,专长于干细胞分化再生的微环境调控机理的研究,.Scadden,博士是麻省总医院再生医学研究所所长,侧重于干细胞的临床应用。

Morrison博士则是休斯医学研究所研究员,是美国中西部大学中干细胞研究的顶级人物。

关于诱导性多能干细胞

关于诱导性多能干细胞

诱导性多能干细胞【关键词】干细胞; 细胞分化; 转录因子诱导性多能干细胞(induced pluripotent stem cells, iPS)是通过基因转染技术(gene transfection)将某些转录因子导入动物或人的体细胞, 使体细胞直接重构成为胚胎干细胞(embryonic stem cell, ES)细胞样的多潜能细胞。

iPS细胞不仅在细胞形态、生长特性、干细胞标志物表达等方面与ES细胞非常相似, 而且在DNA甲基化方式、基因表达谱、染色质状态、形成嵌合体动物等方面也与ES细胞几乎完全相同。

iPS细胞的研究受到人们广泛的关注, 是目前细胞生物学和分子生物学领域的研究热点。

iPS细胞技术诞生还不到2年, 却为干细胞的基础研究和临床疾病治疗研究带来了前所未有的希望, iPS细胞技术的出现使人们从ES细胞和治疗性克隆等激烈的伦理学争论中解脱出来。

但是, 目前制备iPS细胞的方法在安全性方面还存在一定问题, 因此探索一种高效、安全的iPS细胞的制备方法显得十分必要。

1 iPS细胞的制备方法2006年T akahashi等[1]研究小组利用分别携带Oct4、Sox2、Myc和Klf4转录因子的4种逆转录病毒载体感染小鼠胚胎成纤维细胞(mouse embryonic fibroblasts, MEFs), 经过G418药物筛选成功获得第1批iPS细胞。

但是这批iPS细胞系中DNA甲基化的方式与自然存在的ES细胞不同, 而且这批iPS细胞不能形成畸胎瘤。

Okita等[2]研究小组报道了第2批iPS细胞的产生。

他们采用与制备首批iPS细胞相同的方法, 但是采用了不同的筛选基因。

第2批iPS细胞系DNA甲基化的方式与自然存在的ES细胞的甲基化方式相同, 并且能形成畸胎瘤。

2007年末, Takahashi和Yu等[3, 4]两研究小组分别在细胞和科学杂志上报道关于iPS研究里程碑的实验结果, 他们都成功获得了人的iPS细胞系。

诱导多能干细胞技术

诱导多能干细胞技术
forebrain neurons.
Figure 4 | Dysregulation of neuronal transcriptome encoding a subset of
presynaptic proteins, DISC1-interacting proteins and mental-disorder associated proteins in human forebrain neurons carrying the DISC1 mutation.
捷易的优势1—取材灵活(外周血、尿液)
捷易的优势2—非整合型质粒电转(无整合)
捷易的优势3—Xeno-free培养(无异源性物质)
捷易的优势4—CRISPR/Cas9基因编辑
特色: 1)Cas9 mRNA和sgRNA电转 2)单链DNA 做Doner 3)Cas9蛋白和sgRNA电转
Recovery or Construction of disease model in iPSCs
Figure 2 | Defects of glutamatergic synapses in forebrain neurons carrying
the DISC1 mutation.
Figure 3 | A causal role of the DISC1 mutation in regulating synapse formation in human
建立DISC1突变家系来源的iPS细胞系(包括正常和突变),并分化成神经元, 比较差异;
用基因编辑技术确认DISC1的作用; 二代测序寻找机制
Figure 1 | Normal neural differentiation, but markedly reduced total

FDA Warns About Stem Cell Claims中英对照

FDA Warns About Stem Cell Claims中英对照

FDA Warns About Stem Cell ClaimsFDA提醒有关干细胞的研究Stem cell therapies offer the potential to treat diseases or conditions for which few treatments exist.干细胞疗法提供了潜在去治疗疾病或条件的方法存在一些Stem cells, sometimes called the body’s “master cells,” are the precursor cells that develop into blood, brain, bones and all of your organs. Their promise in medical treatments is that they have the potential to repair, restore, replace and regenerate cells that could then be used to treat many medical conditions and diseases.干细胞,有时被称为人体的“主人细胞”是最先的细胞发展成血液,大脑,骨头和所有的人体器官。

它们在医学治疗方面的保证是它们有潜在的能力去修复,恢复,代替和再生能治疗医学方面的情况和疾病的细胞。

But the Food and Drug Administration (FDA) is concerned that the hope that patients have for cures not yet available may leave them vulnerable to unscrupulous providers of stem cell treatments that are illegal and potentially harmful.但是食物和药品管理部门(FDA)担心病人对于治好病的希望不会实现,这可能会让他们容易受到不合法的也是有伤害的干细胞治疗。

ips cell

ips cell

诱导多能干细胞(iPS)的研究热点和潜在应用NIH四月份宣布要从紧张的经费中拨款,筹建诱导多能干细胞(iPS,Induced Pluripotent Stem Cells)中心。

最近NIH也许因为在招人,我比较密集地听了一些iPS相关的讲座,所以想稍微总结一下,给有生物背景的科学工作者增加一些印象。

细胞核转移技术诞生的多利羊曾经在中国掀起一阵“克隆”热。

iPS技术的诞生则大大地更进了一步,它直接证明,用四个转录因子就可以让终末分化的体细胞回到原初的多能干细胞状态。

iPS不仅在生物学理论上有突破,在伦理学上绕过了胚胎干细胞,在实际应用上它也具有非常大的潜力。

1Cell Based Therapy: 在异体细胞或者器官移植过程中,免疫排斥一直是个难题。

理论上自身的体细胞(比如皮肤细胞或者血液细胞)可以在体外重编排成iPS细胞,而iPS细胞具有多能性,它能分化成任何其它组织特异性的终末分化细胞(比如神经细胞)或者干细胞,这些细胞如果移植到自身体内,则基本上不会产生免疫排斥的问题。

通过移植功能性的细胞(或干细胞),有可能使组织的损伤得到修复更新。

比如一些退行性疾病,心脏病,脊柱损伤等就有可能得到治疗。

2iPS细胞治疗的一个好处是,一些有遗传缺陷的细胞可以在体外修复,然后重新植入体内来修复组织或者器官。

3Disease Modeling: 在体内,胚胎初期的干细胞分化出三胚层,胚层再分化成各种各样的细胞。

在体外,多能干细胞能很大程度上重演这个过程。

以往研究一个疾病的成因,需要依赖老鼠的模型,而老鼠建模本身很困难,模型和实际的人的疾病也可能相差甚远。

直接用病人的的细胞在体外重编程为iPS细胞,然后让iPS 细胞分化成相关的有疾病的细胞,用正常iPS细胞做对照,观察这个过程中病人的iPS有哪些缺陷,发生哪些变化,可以为了解这个疾病的发生提供新的工具。

除了体外观察以外,疾病相关的iPS植入没有免疫力的小鼠,还可以在体内观察这些变化。

人诱导干细胞ipsc检测标准_概述说明以及解释

人诱导干细胞ipsc检测标准_概述说明以及解释

人诱导干细胞ipsc检测标准概述说明以及解释1. 引言1.1 概述人诱导干细胞(induced pluripotent stem cells,iPSC)是一种在体细胞经过基因重编程而得到的多能干细胞,在医学和生物学领域引起了广泛的兴趣和研究。

iPSC具有与胚胎干细胞相似的自我更新和分化潜能,同时又避免了使用胚胎来源的争议。

这使得它们成为研究人类发育、疾病机制以及药物筛选与治疗等方面的理想模型。

1.2 文章结构本文旨在对人诱导干细胞检测标准进行全面、系统性的概述和解释。

文章分为五个部分:引言、人诱导干细胞的概述、人诱导干细胞检测的重要性与挑战、已有的人诱导干细胞检测标准概述与解释以及结论与展望。

在第二部分中,我们将介绍人诱导干细胞(iPSC)的定义、背景、特点和应用,以及制备方法和技术发展历程。

这将为后续对其检测标准进行分析和评估提供必要的背景知识。

第三部分将讨论人诱导干细胞检测的重要性和意义,以及当前面临的问题和挑战。

我们还会对未来人诱导干细胞检测标准的发展进行展望,探讨可能出现的改进方向和趋势。

在第四部分中,我们将概述已有的国际标准和相关文献,并介绍主流实验方法和技术指南。

同时,我们也会分析这些检测标准在实际应用中存在的局限性,并提出改进措施建议。

最后,在结论与展望部分,我们将对人诱导干细胞检测标准进行总结和评价,并展望其未来的研究方向与发展趋势。

同时,我们还将讨论人诱导干细胞检测标准对相关领域的影响和应用前景。

1.3 目的本文旨在全面了解并解释人诱导干细胞(iPSC)的概念、特点及其制备方法,并重点关注目前人诱导干细胞检测所面临的挑战和问题。

通过梳理已有的检测标准,分析其在实际应用中的限制,并提出改进措施建议。

最后,对人诱导干细胞检测标准未来的发展方向和应用前景进行展望,为相关研究领域提供指导与参考。

2. 人诱导干细胞(iPSC)的概述2.1 iPSC的定义和背景人诱导干细胞(induced pluripotent stem cells,简称iPSC)是一种通过重编程成体细胞回退到多能性状态而获得的多潜能干细胞。

ipsc细胞gmp级别团体标准

ipsc细胞gmp级别团体标准

ipsc细胞gmp级别团体标准IPSC(诱导多能性干细胞,Induced Pluripotent Stem Cells)是通过重新编程成年细胞而产生的多能性干细胞。

在生物医学研究和临床应用中,为确保安全性和质量,常常要求生产和应用符合一定标准的细胞,包括 GMP(Good Manufacturing Practice)级别的标准。

GMP是一种质量管理体系,确保在生产和制造过程中符合一系列的标准,以确保最终产品的质量、安全性和一致性。

GMP级别的细胞生产涉及到各个环节,包括设备、人员培训、质量控制、文件管理等。

针对IPSC细胞的GMP级别团体标准可能因地区、国家或组织而有所不同,以下是一些可能涉及到的方面:
1. 设备与环境: GMP要求在生产环境中使用符合要求的设备,确保细胞的生产不受外界环境的污染。

这包括洁净室、恒温恒湿设备等。

2. 培养基和试剂物质:使用符合GMP要求的培养基和试剂,确保对细胞生长的影响最小化,同时避免可能的污染。

3. 人员培训:保证从事IPSC生产的人员受过专业的培训,了解GMP要求,严格执行相关的操作规程。

4. 文件管理:记录和存储所有与IPSC生产相关的数据,确保可追溯性,以及对生产流程的透明度。

5. 质量控制:设立质量控制标准,对细胞的质量、纯度、活性等进行监测和测试,确保最终产品符合规定的标准。

6. 监管审批:遵守相关的监管要求,确保生产过程和产品得到批准和监管机构的认可。

这些要求旨在确保生产的IPSC细胞符合高质量和安全性的标准,以满足科研和临床应用的需求。

在具体的应用中,建议在当地或相关领域的专业机构、监管机构或国际组织指南的基础上,制定符合具体实践的GMP级别团体标准。

Transwell 接触共培养促进单散iPSCs 生长及分化

Transwell 接触共培养促进单散iPSCs 生长及分化

Transwell 接触共培养促进单散iPSCs 生长及分化刘庆;郭永龙;郭晓令;连瑞玲;陈建苏【摘要】目的:观察Transwell接触共培养促进单散人诱导多能干细胞(inducedpluripotentstemcells, iPSCs)生长及分化的作用。

方法:将1~2代牛角膜内皮细胞(corneal endothelial cells, CECs)接种在Transwell 小室底面培养8 h后,应用Accutase消化及40μm过滤处理获得单散iPSCs,将其接种到已有CECs的Transwell小室内共培养14 d,前3 d使用mTeSR1培养基,第4天开始用含10%胎牛血清的低糖DMEM培养基。

分别进行实时荧光定量聚合酶链式反应( real-time fluorescence quantitative polymerase chain reaction , qPCR )、免疫荧光、死活细胞染色及碱性磷酸酶( alkaline phosphatase , ALP)染色,对iPSCs多能特性表达及分化进行鉴定。

设定单散iPSCs共培养组为实验组,常规培养iPSCs组为对照组(一),非共培养单散iPSCs组为对照组(二)。

结果:培养牛CECs形态呈典型的六边形铺路石样外观。

人iPSCs呈克隆样生长,共培养3 d后iPSCs贴壁呈单散细胞生长,免疫荧光检测未分化标志Nanog和Oct4呈阳性。

qPCR检测Nanog、Oct4和Sox2 mRNA表达,实验组与对照组(一)比较差异无统计学意义(P>0.05)。

死活细胞染色显示,实验组死细胞明显减少,与对照组(二)比较差异有统计学意义(P <0.01)。

共培养14 d后,人iPSCs形态比较均一,呈多边形,体积增大,无明显克隆团块;ALP染色阴性;免疫荧光染色ZO-1、AQP1和CD31表达阳性,CD34和CD133表达阴性。

qPCR检测Oct4、Nanog和Sox2 mRNA表达明显下调,与对照组(一)比较差异有统计学意义(P<0.01)。

  1. 1、下载文档前请自行甄别文档内容的完整性,平台不提供额外的编辑、内容补充、找答案等附加服务。
  2. 2、"仅部分预览"的文档,不可在线预览部分如存在完整性等问题,可反馈申请退款(可完整预览的文档不适用该条件!)。
  3. 3、如文档侵犯您的权益,请联系客服反馈,我们会尽快为您处理(人工客服工作时间:9:00-18:30)。

Dharmendra Kumar , Thirumala R Talluri, Taruna Anand, Wilfried A Kues, Institute of Farm Animal Genetics, Friedrich-Loeffler-Institute, 31535 Mariensee, GermanyDharmendra Kumar, Animal Physiology and Reproduction Division, Central Institute for Research on Buffaloes, Hisar 125001, Haryana, IndiaThirumala R T alluri, T aruna Anand, National Research Centre on Equines, Hisar 125001, Haryana, IndiaAuthor contributions: Kumar D and Kues WA drafted and wrote the rewiew; Kumar D designed the figures; Talluri TR and Anand T contributed specific chapters; all authors proof-read the final version.Supported by CREST fellowship from Department of Biote-chnology, Ministry of Science and Technology, Government of India (DK); International fellowship for PhD from ICAR (TRT), Government of India; International training in generation of iPS cells from NAIP , ICAR, Government of India (TA).Conflict-of-interest: The authors declare there are no competing interests.Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY -NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: /licenses/by-nc/4.0/Correspondence to: Dr . Wilfried A Kues, PhD, Institute of Farm Animal Genetics, Friedrich-Loeffler-Institute, Höltystr. 10, 31535 Mariensee, Germany. wilfried.kues@fli.bund.de Telephone: +49-5034-871120Fax: +49-5034-871101Received: July 15, 2014Peer-review started: July 17, 2014First decision: August 14, 2014Revised: December 3, 2014 Accepted: December 16, 2014Article in press: December 18, 2014Published online: March 26, 2015AbstractPluripotent stem cells are unspecialized cells withu nlimited self-renewal, and they can be triggered to differentiate into desired specialized cell types. These features provide the basis for an unlimited cell source for innovative cell therapies. Pluripotent cells also allow to study developmental pathways, and to employ them or their differentiated cell derivatives in pharmaceutical testing and biotechnological applications. Via blastocyst complementation, pluripotent cells are a favoured tool for the generation of genetically modified mice. The recently established technology to generate an induced pluripotency status by ectopic co-expression of the transcription factors Oct4, Sox2, Klf4 and c-Myc allows to extending these applications to farm animal species, for which the derivation of genuine embryonic stem cells was not successful so far . Most induced pluripotent stem (iPS) cells are generated by retroviral or lentiviral transduction of reprogramming factors. Multiple viral integrations into the genome may cause insertional mutagenesis and may increase the risk of tumour formation. Non-integration methods have been reported to overcome the safety concerns associated with retro and lentiviral-derived iPS cells, such as transient expression of the reprogramming factors using episomal plasmids, and direct delivery of reprogramming mRNAs or proteins. In this review, we focus on the mechanisms of cellular reprogramming and current methods used to induce pluripotency. We also highlight problems associated with the generation of iPS cells. An increased understanding of the fundamental mechanisms underlying pluripotency and refining the methodology of iPS cell generation will have a profound impact on future development and application in regenerative medicine and reproductive biotechnology of farm animals. Key words: Reprogramming; Large animal models; Stemness; Chimera; Germline transmission; Induced pluripotent stem cells; Gene delivery© The Author(s) 2015. Published by Baishideng Publishing Group Inc. All rights reserved.Core tip: The generation of an induced status of pluripotencyREVIEWSubmit a Manuscript: /esps/Help Desk: /esps/helpdesk.aspx DOI: 10.4252/wjsc.v7.i2.315World J Stem Cells 2015 March 26; 7(2): 315-328ISSN 1948-0210 (online)© 2015 Baishideng Publishing Group Inc. All rights reserved.Induced pluripotent stem cells: Mechanisms, achievements and perspectives in farm animalsDharmendra Kumar , Thirumala R Talluri, Taruna Anand, Wilfried A Kuesin somatic cells by ectopic expression of core transcription factors allows to extending advanced genetic modifications and reproductive techniques to species, for which the derivation of genuine embryonic stem cells was not successful till now. The commonly employed viral gene transfer may be genotoxic and therefore non-viral methods for iPS cell derivation are intensively studied. In this review, we focus on the mechanisms of cellular reprogramming and current methods used to induce pluripotency.Kumar D, Talluri TR, Anand T, Kues WA. Induced pluripotent stem cells: Mechanisms, achievements and perspectives in farm animals. World J Stem Cells2015; 7(2): 315-328 Available from: URL: /1948-0210/full/v7/i2/315.htm DOI: /10.4252/wjsc.v7.i2.315INTRODUCTIONInduced pluripotent stem (iPS) cells are defined as differentiated cells that have been experimentally reprogrammed to an embryonic stem (ES) cell-like state. The first generation of murine iPS cells was achieved[1]by retroviral transduction of four core reprogramming factors: Oct4, Sox2, Klf4, and c-Myc. Subsequently, human iPS cells were produced by viral transduction of adult fibroblasts[2,3]. Also a combination of Oct4, Sox2, Nanog and Lin28, was effective for the generation of human iPS cells[4]. An overview of reprogramming cells into iPS cells is shown in Figure 1.Subsequently, the core reprogramming factors have been successfully used to derive pluripotent cells in various other species, including rhesus monkey[5], rat[6], pig[7], dog[8], cattle[9], horse[10], sheep[11], goat[12] and buffalo[13]. A summary of the generation of iPS cells from different species of livestock is enumerated in T able 1. Importantly, iPS cells could be isolated from several species, in which the isolation of authentic ES cells was not successful despite several attempts since many years[14,15]. In particular, for economically important species, such as farm animals, the availability of authentic iPS cells would have important consequences for reproductive biology and approaches for genetic modification. For agricultural purposes, iPS cells from farm animal species can serve as a valuable genetic engineering tool to boost the generation of livestock with advantageous genes that are important for economic, reproductive and disease resistant traits, or for the study of functional genomics in mammals.So far, iPS cells have been successfully produced from fibroblasts[16], pancreas cells[17], leukocytes[18], hepatocytes[19], keratinocytes[20], neural stem cells[21], cord blood cells[22], and other cell types. T ogether these data suggest that most cell types can be reprogrammed to a pluripotent state, and that the unidirectional lineage commitment can be experimentally overwritten. Certain cell types, such as neuronal progenitors, which exhibit basal expression of one or more of the core reprogramming factors, seem to be ideal for reprogramming[21].Rodent iPS cells are almost identical to their ES cell counterparts, sharing typical hallmarks of pluripotency such as colony morphology, unlimited self-renewal, in vitro and in vivo differentiation potentials, and contribution to the germline[23,24]. Most iPS lines from farm animal species have not been tested in chimera complementation assays; however some preliminary reports suggest that chimeras and germline transmission can be achieved in sheep and pig[25,26]. iPS cells derived from rodents, humans, monkeys and farm animals share the features of high telomerase activity, expression of alkaline phosphatase, and expression of stemness genes, such as OCT4, SOX2, UTF1 and REX1. The epigenetic status of murine iPS cells has been analysed by bisulfite sequencing and chromatin immuno-precipitation DNA-Sequencing (ChIP-Seq)[27]. Thus the hallmarks for iPS cell characterisation can be enumerated as (1) unlimited self-renewal;(2)in vitro differentiation capacity; (3) in vivo differentiation capacity; (4) chimera contribution; and (5) subsequently germline transmission.Apart from scientific and ethical hindrances, religious concerns restricted the derivation of human ES cells. To circumvent these concerns, alternative approaches to generate pluripotent cells have been assessed. The alternative approaches include culture of somatic cells with cell extracts isolated from ES cells[28] or oocytes[29], and fusion of somatic cell with pluripotent cell[30]. However, extremely low efficiencies, high technical difficulties and aberrant ploidies of the resulting cells[31,32] did reduce the enthusiasm for these attempts. At the moment, the derivation of iPS cells from human tissues seems to be the most promising alternative. Prior to clinical application of iPS-derivatives, cell survival, functional integration of the cellular transplant and safety of the cell products have to be assessed in informative animal models.The progress in iPS cell development in farm animals lags behind those in rodents, but large mammalian models may be instrumental for pre-clinical tests of novel cell therapies (T able 2), enhanced pharmaceutical studies and regenerative studies, including the restoration of fertility. HISTORICAL PERSPECTIVEOntogenesis of an organism and cellular differ-entiation were thought to be a unidirectional process, where stem and progenitor cells progressively develop to terminally differentiated cells, for example neurons, muscle, and epithelial cells. During ontogenesis the nuclear DNA of most cell types is unchanged, but different epigenetic marks, such as DNA methylation and histon modifications, are set, and lock theKumar D et al. IPS cells from farm animals speciescellular potency and cell lineage commitment. This is depicted by the “epigenetic landscape” proposed byWaddington [33].Already in 1962, Gurdon [34]questioned this view by amphibian cloning; he transplanted nuclei from intestinal cells into irradiated oocytes and obtained vital tadpoles. More than three decades later , the successful cloning of a sheep (Dolly) by SCNT of a mammary epithelial cell to an enucleated oocyte, showed thateven mammalian cells can be reprogrammed [35]. This success demonstrated that differentiated cells contain the genetic information to direct ontogenesis of an entire mammalian organisms, and that enucleated oocytes contain pivotal factors for reprogramming of differentiated cell nuclei. However , the identity of the oocyte reprogramming factors remained elusive.The discoveries that ectopic expression of Antennapedia-a transcription factor was able andsufficient to induce leg structures in Drosophila [36], and that ectopic expression of the mammalian transcriptionfactor MyoD1 converted fibroblasts into myocytes [37]led to the concept of ‘‘master genes’’. A master gene was defined as a key transcription factor that in a hierarchical manner regulates a cascade of critical genes, which in a concerted action induce the cell commitment.DISCOVERY OF INDUCED PLURIPOTENCYIn 2006, T akahashi et al [1]proved that not a singlemaster factor , but a a combination of four repro-gramming factors, Oct4, Sox2, Klf4 and c-Myc, was sufficient to induce the pluripotent status in somatic mammalian cells. The resulting cells were called iPScells [1]. This discovery offers new opportunities to study developmental biology , regenerative medicine, as well as reproductive biology and biotechnology of farm animals.IPS cells from farm animals will likely serve as a bridging link between well developed rodent iPS and poorly characterised human iPS (T able 2), supporting the translation of innovative cell therapies from experimental studies to curative treatments. At the moment, human iPS cell application seems to be too risky because of basic lack of knowledge and ethical consideration which forbid certain tests such as chimera assays.In contrast, research on iPS cells derived from farm animal species is not tainted with ethical concerns. Furthermore, the methodology for generation of iPS cells is relative simple and and is thought to be easily transferable to other mammalian species. Thus farm animal models may turn out to be ideally suited to determine required cell doses, to assess long-term performance, tumorigenicity , applications methods andfate of transplanted cells [38-41].Recent advances in genetic engineering of farm animalsallow the generation of precise genetic modifications [42-47],such as the production of immunodeficient pigs [48]which will be instrumental for further advances in preclinical testings of new cell therapies. A boost of recentSomatic cells: (1) uni-to multipotent (2) limited differentiation (3) no teratoma capabilityReprogramming factorsOCT4, SOX2, NANOG, KLF4, C-MYC, LIN28, GLIS1Viral Non-viralRetrovirus Lentivirus Adenovirus Recombinant proteinsModified RNAs Minicircle DNA Episomal plasmids piggyBac transposon Sleeping Beauty transposonSendai virusiPS cells: (1) pluripotent(2) in vitro differentiation(3) in vivo differentiation (teratoma) (4) chimera contribution (5) germline transmissionEndogenous expression of core reprogramming factorsERK inhibitors MEK inhibitors Epigenetic modifiersSmall molecules Somatic stem cellsFigure 1 Methodological toolbox for generating induced pluripotent stem cells. iPS: Induced pluripotent stem.Kumar D et al . IPS cells from farm animals speciesVirally-induced iPS cellsThere has been extensive amount of work carried out to obtain virally-derived iPS cells employing either retroviruses, lentiviruses, and non-integrating viruses. The first iPS cells have been generated throughretroviral transduction of Oct4, Sox2, Klf4 and c-Myc [1]. Disarmed, optimized retro- or lentiviruses can infect mammalian cells with high efficiencies. The use of the pantropic vesicular stomatitis virus G protein (VSVG) was instrumental for viral transduction of a broad spectrum of receptive cells. Interestingly , unstimulated T cells, B cells and hematopoietic stem cells could notbe efficiently transduced with the VSVG lentiviruses [77].Retro- and lentiviruses integrate into the host genome allowing for high expression of the encoded cargo genes. The expression can be temporally confined by employing viral promoters, such as the 5’ long terminal repeat, which are usually silenced by epigenetic mechanisms. Disadvantages of the the viral approach include the limited cargo capacity of maximally 7 kb, the induction of immune responses and potential genotoxic effects. Retro- and lentiviral integrations do not happen randomly in the genome, but show a strong bias for promoter and exonic regions, which may result in dysregulation of endogenous genes. In a retrovirus-based clinical gene therapy of the X-linkedpublications describe iPS cells from buffalo [13], cattle [9,49-53],dog [8,54-56], goat [11,57], horse [10,58-62], pig [7,63-71], rabbit [72-74]and sheep [11,75,76]. The majority of these iPS cells from farm animals showed typical hallmarks of pluripotency , such as differentiation in vivo and teratoma formation. However , most farm animal iPS cultures were not assessed for chimera contribution so far . Preliminary results that porcine iPS cells can contribute to chimera formation in blastocystcomplementation were provided recently [71]. Similarly , ovine iPS cells contributed moderately to chimeric lambs after injection into eight-cell stage embryos orblastocysts [25]. These experiments represent an important step in the understanding of mechanistic nature of pluripotency in farm animals. The iPS technology may become instrumental for advanced transgenesis in large mammals (Figure 2).METHODS TO DERIVE IPS CELLSIn recent years, several methods have been established for iPS cell generation (Figure 1), employing the core reprogramming factors as genes, mRNAs or proteins, and auxillary chemical agents, which infer with the involved signalling pathways. Here, the main approaches for the generation of iPS cells are summarized.A: DMEM, ESC FBS, L-glutamine, NEAA, β-Me, bFGF, LIF and MEFs; B: DMEM, KSR, L-glutamine, NEAA, β-Me, bFGF and MEFs; C: DMEM/F12 + N2 and Neurobasal with B27, L-glutamine, hLIF, PD0325901, CHIR99021 and MEFs; D: KO-DMEM, SR, L-glutamine, NEAA, 2-Me, human bFGF and MEFs; E: DMEM, FBS, L-Glutamine, NEAA, β-Me, Sodium Pyruvate, LIF, bFGF, Doxycycline, CHIR99021, PD0325901, A83-01, Thiazovivin, B431542 and 1:1 MEFs and EFFs; F: α-MEM, FBS, deoxyribonucleosides, ribonucleoside, glutamax, NEAA, β-Me, ITS, human LIF, βFGF, EGF and MEFs; G: DMEM/F12, KSR, L-glutamine, NEAA, β-Me, FGF and MEFs; H: KO DMEM, KSR, glutamax-L, NEAA, 2-Me, pLIF, forskolin and collagen I; I: DMEM/F12, KSR, L-glutamine, NEAA, β-Me, bFGF and MEFs or gelatinized plates; J: KO DMEM, ESC FBS, bFGF, hLIF and MEFs; K: DMEM/F12, KSR, bFGF, hLIF, PD0325901, CHIR99021 and MEFs. DMEM: Dulbecco’s modified Eagle´s medium; LIF: Leukemia inhibitory factor; IGF1: Insulin-like growth factor 1; NEAA: Nonessential amino acids; FBS: Fetal bovine serum; KO: Knockout; MEM: Minimum essential medium; ITS: Insulin-transferring selenium; bFGF: Basic fibroblastic growth factor; DOX: Doxycycline; EB: Embryonic body; FCS: Fetal calf serum; hSCF: Human stem cell factor; KSR: Knockout serum replacement; MEFs: Mouse embryonic fibroblasts; OKSM: Oct-4, Klf4, Sox2, and c-Myc; OKSMLN: Oct-4, Klf4, Sox2, c-Myc, Lin28 and Nanog; VPA: Valproic acid; Me: Mercaptoethanol.Kumar D et al . IPS cells from farm animals speciessevere combined immunodeficiency (X-SCID), two of the treated children independently developed T-cell lymphomas due to viral integration in the neighborhood of the LIM domain only 2 gene[78]. These data highlight the risks of viral-based therapies[78]. Somatic cells derived from retrovirally reprogrammed iPS cells are apparently inconspicious, provided that the c-Myc transgene is silenced[19,79]. Retroviral reprogramming may evoke an immunogenicity of iPS cells[80]. Human iPS cell-like cells can be formed through transduction with lentiviruses, which do not carry reprogramming factors. The “pseudo” iPS cells were induced by viral encoded microRNA expression[81].Alternative to integrating retroviruses, non-integrating adenoviruses can be used for reprogramming[17,82]. Another non-integrating virus is represented by the Sendai virus system. Sendai viruses enable efficient production of iPS cells and later on elimination of the viral vector[83]. Though viral mediated gene transfer offers high efficiency in generation of iPS cells, they require specific safety conditions for their handling. Non-virally-derived iPS cellsThe generation of iPS cells without viral transduction is preferable for regenerative medicine. Non-viral methods of reprogramming include episomal vectors[84], minicircle DNAs[85], plasmid vectors[86], small molecules[87], mRNAs[88], recombinant proteins[89] and transposons like piggyBac[90] and Sleeping Beauty[91]. In comparison to viral systems, non-viral approaches such as transposons are able to carry large DNA cargo into the host cell, they are non-infectious and do not evoke immune responses.Episomal vectorsEpisomal vectors for reprogramming of somatic cells were recently described[84]. In this method, reprogramming of fibroblasts was carried out by transfecting with the episomal vector oriP/Epstein-Barr nuclear antigen-1. This vector was chosen because it can be removed after reprogramming by a drug selection method. The iPS cells generated through this method show similar morphology and expression patterns to ES cells. Further, they were able to form√√: Fully proven; √/-: Partially proven; --: Not achieved yet; 1The first clinical study was recently initiated (http://www. riken.jp/en/pr/press/2013/20130730_1). iPS: Induced pluripotent stem.EnucleatedoocyteGenetic modification(A) Nuclear transferGenetically modified iPS cellsElectrofusion and oocyte activationIn vitro cultureBlastocystTransgenic offspring Embryo transferinto surrogate(B) Blastocyst complementationiPS cellsReconstructed embryoFigure 2 Application of induced pluripotentstem cells for advanced generation oftransgenic animals. iPS: Induced pluripotentstem.Kumar D et al. IPS cells from farm animals speciesteratomas in immunocompromised mice. As there was no integration into the host genome, transgene free iPS cells may be selected through further sub-cloning. Despite these advantages, this method yields low reprogramming efficiency in human fibroblasts at about three to six iPS colonies per 106 input cells[84]. Minicircle vectorsMinicircle vectors are produced by the recombinatorial elimination of the bacterial backbone of the original plasmids. Minicircles containing the four reprogramming factors Oct4, Nanog, Lin28, and Sox2 in addition to an enhanced green fluorescent protein were used to obtain human iPS cells[85]. The group excised the bacterial backbone from the plasmid by taking advantage of the PhiC31-based intramolecular recombination system, which cleaves away the undesired bacterial plasmid backbones, leaving minicircle DNA to be purified containing the desired reprogramming factors[85]. It was claimed that minicircle DNA benefited from higher transfection efficiency compared to the parental plasmids. They also have longer ectopic expression, which is due to the lower activation of exogenous silencing mechanisms. Later, other groups reproduced the minicircle approach for reprogramming[92,93].Small moleculesNowadays, small molecules and chemicals are assessed to enhance reprogramming efficiency and iPS cell generation. The idea behind their use is to substitute core reprogramming factors with small molecules, which will serve to enhance the reprogramming. Shi et al[94] showed that neural progenitor cells, which endogenously express Sox2, were reprogrammed only by ectopic expression of Oct4 and Klf4. They also showed that this process was supported by the G9a histone methyltransferase inhibitor, BIX-01294 (BIX). Ichida et al[95] used small molecules (RepSox2) for replacing transcription factors (Sox2) by inhibiting transforming growth factor-β signalling. In this direction, Lee et al[96] used magnetic nanoparticle-based transfection method that employs biodegradable cationic polymer PEI-coated super paramagnetic nanoparticles for iPS cells generation. Recently, the L-channel calcium agonist, BayK8644, was assessed to improve generation of iPS cells[87]and it was claimed that BayK8644 does not directly cause epigenetic modifications as it works upstream in cell signalling pathways and can therefore avoid unwanted modifications. A more comprehensive list of small molecules involved in the iPS cells generation and their mechanism has been reviewed recently[97].Transposon systemsThe recent development of hyperactive transposase enzymes makes transposon systems an interesting alternative to viral based methods. The commonly employed Sleeping Beauty, piggyBac and Tol2 transposon systems are relatively simple organized, and the essential components can be separated on two plasmids. One plasmid carries the inverted terminal repeats (ITR) flanking the transgene, the other plasmid carries an expression cassette for the respective transposase enzyme. Upon co-transfer of both plasmids into a cell, the transposase becomes expressed and subsequently transposes the ITR-flanked transgene into the genome. Importantly, only the desired transgenes becomes integrated by a cut-and-paste mechanism, whereas the plasmid backbones are degraded. On a genomic scale transposon integrations appear to happen at random, without a bias for promoter and gene-containing regions. The integrated transposon can be removed seamlessly by supplying the transposase in trans[98], which makes the system more attractive and relevant in producing the safe and clean iPS cells. Up to six reprogramming factors have been connected by self-cleaving peptide sequences allowing for coexpression from a single cassette[91,99-103]. Individual proteins are then produced by the self-cleaving peptide[104-106].Reprogramming with protein factorsThe discussed transposon and episomal systems still require the introduction of cargo DNA into the cells[106]. Delivery of reprogramming factors as proteins is an obvious alternative. In 2009, transgene-free iPS cells were produced with proteins of reprogramming factors[107]. Therefor recombinant reprogramming proteins were produced as fusion proteins containing cell penetrating peptides. Repeated supplementation of the culture media of fibroblasts converted them to iPS cells. However, the protein-based reprogramming approach has not found widespread use, mainly due to relative low reprogramming efficiencies, and high costs for repeated treatments with protein factors.mRNAs and microRNAsThe most recent trend in the field of non-viral iPS generation is reprogramming by using RNA molecules. Recently, modified mRNAs encoding reprogramming factors were employed to generate iPS cells with high efficiency[108]. Messenger RNAs are an ideal vehicle for reprogramming, because they do not bear the risk of integrational mutagenesis, they can be transduced to cells with high efficiency, and they can be combined in desired ratios of the individual factor encoding transcripts[108]. Disadvantages of mRNAs are the short half-life of -10 h, and that innate immune responses must be inhibited to allow for the full effects[109].Recently, it was shown that micro RNAs (miR) expression is sufficient to induce pluripotency[110-112]. T wo independent groups reported iPS cell generation by delivery of miR302, or miR200c, miR302, and miR369[113,114]. These miR-derived iPS cells were indistinguishable from conventionally generated iPS cells. MicroR reprogramming seems to have advantagesKumar D et al. IPS cells from farm animals speciesfor cellular reprogramming[114-116], for example it avoids the need of transducing proto-oncogenic transcription factors[117,118]. However, it needs to be assessed whether this approach will be successful in other species, since the underlying mechanisms are not well understood[119]. MOLECULAR FACTORS REGULATING REPROGRAMMINGThe core factors for reprogramming are Oct4, Nanog, Sox2, Klf4, c-Myc and Lin28. These genetic factors reprogram cells by regulating critical signalling pathways, epigenetic modifications and micro RNAs[114]. Reprogramming by core transcription factorsOct4 is the best studied regulator of pluripotency. Oct4 expression is confined to early embryonic cells, germ line cells and cultured pluripotent stem cells, where it activates the gene transcription of stemness gene[120]. Oct4 protein cooperates with stemness factors such as Nanog and Sox2, but it also interacts with Polycomb group proteins[120], which are important repressors of transcription. Sox2 is a transcription factor that acts as coactivator of Oct4[121]. Binding of Oct4/Sox2 dimers to the promoter sequences of Oct4 and Nanog genes upregulate their transcription[122]. Nanog is a homeobox-containing transcription factor stabilizing the stemness network[122]. Klf4 is a zinc finger-containing transcription factor which regulates the expression of Oct4, Sox2 and Nanog[123-125]. Over-expression of Klf4 in ES cells increased the expression of Oct4 which further improve the self-renewal ability[126]. c-Myc enhances the efficiency and speed of reprogramming[127]. LIN28 promotes the expression of Oct4 at the posttranscriptional level by direct binding to its mRNA[128]. Recently, Glis1 has been identified as a substitute for c-Myc[129]. Glis1 transactivate the genes of Wnt ligands, Lin28a, Nanog, Mycn, Mycl1, and Foxa2[129].The aspect of whether the species-specificity of reprogramming factors is relevant for proper reprogramming, is not well understood. In principle, the essential domains of the reprogramming factors are highly conserved between mammalian species, and several publications showed successful repro-gramming with human and murine sequences in other species[5-13,130].APPLICATIONS OF IPS CELLSModeling of human diseases and preclinical trialsThe potential applications of iPS cells will impact regenerative medicine, pharmaceutical industry, and animal biotechnology[131]. Human iPS cells could be utilized for curative treatments, to studying onset and disease progression in vitro, and to test potential therapeutic in high throughput screens[114,131,132]. The production of disease-specific iPS cells has found widespread use in recent years[133-136]. Disease-specific iPS cells provide a unique resource to obtain a molecular understanding of disease onset and progression[131,132]. Induced PS-derived differentiated cells will allow to carry out in vitro drug screening (Figure 3), and to test therapeutic interventions[131]. In mice, Fanconi anemia and sickle cell anemia have been successfully corrected by using iPS cells[131,133-136].However with regard to potential curative treatments, the functionality, safety, and lack of tumorigenicity of iPS-derived cells have to be assessed in appropriate animal models bearing significant physiological and anatomical similarities to humans (T able 2). Hence, animal models could be contributed tremendously to a better understanding of disease mechanisms and therapeutic interventions. In addition, iPS cells from monkey[5], porcine[41,26], canine[8] and cattle[9] would be useful in animal biotechnology such as making precise genetic engineering for improved production traits and products[137,138].Advanced transgenesis in large mammalsTransgenic farm animals can serve as excellent models of human diseases and during the past few years transgenic farm animals have gained renewed popularity. This is due to the availability of annotated genome depositories of the major domestic species and other organisms (for example: www.ensembl. org; or /genome), and due the introduction of active methods of transgenesis, which dramatically increased the success rates[42,43]. The repertoire of molecular tools now allows the precise modification of large mammalian genomes at rapid pace and has led to a recent boost in this area. The development of genuine iPS cells from domestic species will contribute to these advances and allow to perform desired genetic modifications via high throughput screens in vitro, and then use either SCNT[47]or blastocyst complementation for the generation of transgenic offspring (Figure 3). However at the moment most of the iPS cells cultures from different domestic species have not been tested for their capability to contribute to chimera formation, and only preliminary data are available[25,26]. Thus reinforced efforts to assess the potential of current livestock iPS cell lines for chimera contribution and germ cell differentiation are required. The majority of current livestock iPS cell lines are generated with retro- or lentiviral reprogramming approaches (T able 1), and the opportunities to assess alternative non-viral approaches are not widely assessed[10,56,106]. Also the potential of auxillary small molecular inhibitors of stemness signaling pathway is not exploited for livestock iPS cells. Potentially, high throughput screens to identify small molecules with species-specific activity are required. It is anticipated that these approaches will lead to livestock iPS cells, which will make a significant impact for future genetic modifications of these species.Kumar D et al. IPS cells from farm animals species。

相关文档
最新文档