301
|
Databases and collaboration require standards for human stem cell research. Drug Discov Today 2014; 20:247-54. [PMID: 25449658 DOI: 10.1016/j.drudis.2014.10.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 09/26/2014] [Accepted: 10/20/2014] [Indexed: 11/20/2022]
Abstract
Stem cell research is at an important juncture: despite significant potential for human health and several countries with key initiatives to expedite commercialization, there are gaps in capturing and exploiting the results of past and current research. Here, we propose a concerted plan that could be taken to foster a more collaborative approach and ensure that all research efforts can be leveraged across the community. The creation of a definitive centralized database repository, or at least harmonized data repositories, for stem cell groups in academia and industry, enabling secure selective sharing of data when needed, could provide the core structure that is sought globally and protect intellectual property. The development of minimum information about stem cell experiments (MIASCE) could be key to this development.
Collapse
|
302
|
Cancer-like epigenetic derangements of human pluripotent stem cells and their impact on applications in regeneration and repair. Curr Opin Genet Dev 2014; 28:43-9. [PMID: 25461449 DOI: 10.1016/j.gde.2014.09.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 09/12/2014] [Accepted: 09/14/2014] [Indexed: 01/27/2023]
Abstract
A growing body of work has raised concern that many human pluripotent stem cell (hPSC) lines possess tumorigenic potential following differentiation to clinically relevant lineages. In this review, we highlight recent work characterizing the spectrum of cancer-like epigenetic derangements in human embryonic stem cells (hESC) and human induced pluripotent stem cells (hiPSC) that are associated with reprogramming errors or prolonged culture that may contribute to such tumorigenicity. These aberrations include cancer-like promoter DNA hypermethylation and histone marks associated with pluripotency, as well as aberrant X-chromosome regulation. We also feature recent work that suggests optimized high-fidelity reprogramming derivation methods can minimize cancer-associated epigenetic aberrations in hPSC, and thus ultimately improve the ultimate clinical utility of hiPSC in regenerative medicine.
Collapse
|
303
|
Yu H, Cheng L, Cho KS. The potential of stem cell-based therapy for retinal repair. Neural Regen Res 2014; 9:1100-3. [PMID: 25206766 PMCID: PMC4146102 DOI: 10.4103/1673-5374.135311] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/20/2014] [Indexed: 12/20/2022] Open
Affiliation(s)
- Honghua Yu
- Department of Ophthalmology, General Hospital of Guangzhou Military Command of PLA, Guangzhou, Guangdong Province, China ; Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, 20 Staniford St., Boston, MA, USA
| | - Lin Cheng
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China ; Institute of Clinical Pharmacology, Central South University; Hunan Key Laboratory of Pharmacogenetics, Changsha, Hunan Province, China ; Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, 20 Staniford St., Boston, MA, USA
| | - Kin-Sang Cho
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, 20 Staniford St., Boston, MA, USA
| |
Collapse
|
304
|
Székely T, Burrage K, Mangel M, Bonsall MB. Stochastic dynamics of interacting haematopoietic stem cell niche lineages. PLoS Comput Biol 2014; 10:e1003794. [PMID: 25188267 PMCID: PMC4154659 DOI: 10.1371/journal.pcbi.1003794] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Accepted: 07/04/2014] [Indexed: 01/02/2023] Open
Abstract
Since we still know very little about stem cells in their natural environment, it is useful to explore their dynamics through modelling and simulation, as well as experimentally. Most models of stem cell systems are based on deterministic differential equations that ignore the natural heterogeneity of stem cell populations. This is not appropriate at the level of individual cells and niches, when randomness is more likely to affect dynamics. In this paper, we introduce a fast stochastic method for simulating a metapopulation of stem cell niche lineages, that is, many sub-populations that together form a heterogeneous metapopulation, over time. By selecting the common limiting timestep, our method ensures that the entire metapopulation is simulated synchronously. This is important, as it allows us to introduce interactions between separate niche lineages, which would otherwise be impossible. We expand our method to enable the coupling of many lineages into niche groups, where differentiated cells are pooled within each niche group. Using this method, we explore the dynamics of the haematopoietic system from a demand control system perspective. We find that coupling together niche lineages allows the organism to regulate blood cell numbers as closely as possible to the homeostatic optimum. Furthermore, coupled lineages respond better than uncoupled ones to random perturbations, here the loss of some myeloid cells. This could imply that it is advantageous for an organism to connect together its niche lineages into groups. Our results suggest that a potential fruitful empirical direction will be to understand how stem cell descendants communicate with the niche and how cancer may arise as a result of a failure of such communication. Stem cells portend great potential for advances in medicine. However, these advances require detailed understanding of the dynamics of stem cells. In vitro studies are now routine and challenge our preconceptions about stem cell biology, but the dynamics of stem cells in vivo remain poorly understood. Thus, there is a real need for novel computational frameworks for general understanding and predictions about experiments on stem cells in their native environments. By implementing a stochastic model of stem cell dynamics, generically based on the bone marrow system, in a novel, fast and computationally efficient way, we show how different couplings of stem cell niche lineages lead to different predictions about homeostatic control. Understanding the demand control of stem cell systems is essential to both predicting in vivo stem cell dynamics and also how its breakdown may lead to the development of cancers of the blood system.
Collapse
Affiliation(s)
- Tamás Székely
- Department of Computer Science, University of Oxford, Oxford, United Kingdom
- * E-mail:
| | - Kevin Burrage
- Department of Computer Science, University of Oxford, Oxford, United Kingdom
- Department of Mathematics, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Marc Mangel
- Department of Applied Mathematics and Statistics, University of California Santa Cruz, Santa Cruz, California, United States of America
- Department of Biology, University of Bergen, Bergen, Norway
| | - Michael B. Bonsall
- Mathematical Ecology Research Group, Department of Zoology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
305
|
Muratore CR, Srikanth P, Callahan DG, Young-Pearse TL. Comparison and optimization of hiPSC forebrain cortical differentiation protocols. PLoS One 2014; 9:e105807. [PMID: 25165848 PMCID: PMC4148335 DOI: 10.1371/journal.pone.0105807] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 07/18/2014] [Indexed: 02/07/2023] Open
Abstract
Several protocols have been developed for human induced pluripotent stem cell neuronal differentiation. We compare several methods for forebrain cortical neuronal differentiation by assessing cell morphology, immunostaining and gene expression. We evaluate embryoid aggregate vs. monolayer with dual SMAD inhibition differentiation protocols, manual vs. AggreWell aggregate formation, plating substrates, neural progenitor cell (NPC) isolation methods, NPC maintenance and expansion, and astrocyte co-culture. The embryoid aggregate protocol, using a Matrigel substrate, consistently generates a high yield and purity of neurons. NPC isolation by manual selection, enzymatic rosette selection, or FACS all are efficient, but exhibit some differences in resulting cell populations. Expansion of NPCs as neural aggregates yields higher cell purity than expansion in a monolayer. Finally, co-culture of iPSC-derived neurons with astrocytes increases neuronal maturity by day 40. This study directly compares commonly employed methods for neuronal differentiation of iPSCs, and can be used as a resource for choosing between various differentiation protocols.
Collapse
Affiliation(s)
- Christina R. Muratore
- Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Priya Srikanth
- Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- Program in Neuroscience, Graduate School of Arts and Sciences, Harvard University, Cambridge, Massachusetts, United States of America
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, Massachusetts, United States of America
| | - Dana G. Callahan
- Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Tracy L. Young-Pearse
- Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
306
|
Koh S, Piedrahita JA. From "ES-like" cells to induced pluripotent stem cells: a historical perspective in domestic animals. Theriogenology 2014; 81:103-11. [PMID: 24274415 DOI: 10.1016/j.theriogenology.2013.09.009] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Revised: 09/05/2013] [Accepted: 09/05/2013] [Indexed: 01/10/2023]
Abstract
Pluripotent stem cells such as embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) provide great potential as cell sources for gene editing to generate genetically modified animals, as well as in the field of regenerative medicine. Stable, long-term ESCs have been established in laboratory mouse and rat; however, isolation of true pluripotent ESCs in domesticated animals such as pigs and dogs have been less successful. Initially, domesticated animal pluripotent cell lines were referred to as "embryonic stem-like" cells owing to their similar morphologic characteristics to mouse ESCs, but accompanied by a limited ability to proliferate in vitro in an undifferentiated state. That is, they shared some but not all the characteristics of true ESCs. More recently, advances in reprogramming using exogenous transcription factors, combined with the utilization of small chemical inhibitors of key biochemical pathways, have led to the isolation of iPSCs. In this review, we provide a historical perspective of the isolation of various types of pluripotent stem cells in domesticated animals. In addition, we summarize the latest progress and limitations in the derivation and application of iPSCs.
Collapse
Affiliation(s)
- Sehwon Koh
- Center for Comparative Medicine and Translational Research, North Carolina State University, Raleigh, North Carolina, USA; Genomics Program, North Carolina State University, Raleigh, North Carolina, USA
| | | |
Collapse
|
307
|
Production of neural stem cells from human pluripotent stem cells. J Biotechnol 2014; 188:122-9. [PMID: 25150215 DOI: 10.1016/j.jbiotec.2014.07.453] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Revised: 07/01/2014] [Accepted: 07/31/2014] [Indexed: 12/24/2022]
Abstract
Despite significant advances in commercially available media and kits and the differentiation approaches for human neural stem cell (NSC) generation, NSC production from the differentiation of human pluripotent stem cell (hPSC) is complicated by its time-consuming procedure, complex medium composition, and purification step. In this study, we developed a convenient and simplified NSC production protocol to meet the demand of NSC production. We demonstrated that NSCs can be generated efficiently without requirement of specific small molecules or embryoid body formation stage. Our experimental results suggest that a short suspension culture period may facilitate ectoderm lineage specification rather than endoderm or mesoderm lineage specification from hPSCs. The method developed in this study shortens the turnaround time of NSC production from both human embryonic stem cells (hESCs) and induced pluripotent stem cells (iPSCs) differentiation. It provides a straightforward and useful strategy for generating NSCs that can benefit a wide range of research applications for human brain research.
Collapse
|
308
|
Pistollato F, Louisse J, Scelfo B, Mennecozzi M, Accordi B, Basso G, Gaspar JA, Zagoura D, Barilari M, Palosaari T, Sachinidis A, Bremer-Hoffmann S. Development of a pluripotent stem cell derived neuronal model to identify chemically induced pathway perturbations in relation to neurotoxicity: effects of CREB pathway inhibition. Toxicol Appl Pharmacol 2014; 280:378-88. [PMID: 25150140 DOI: 10.1016/j.taap.2014.08.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Revised: 07/01/2014] [Accepted: 08/11/2014] [Indexed: 01/09/2023]
Abstract
According to the advocated paradigm shift in toxicology, acquisition of knowledge on the mechanisms underlying the toxicity of chemicals, such as perturbations of biological pathways, is of primary interest. Pluripotent stem cells (PSCs), such as human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs), offer a unique opportunity to derive physiologically relevant human cell types to measure molecular and cellular effects of such pathway modulations. Here we compared the neuronal differentiation propensity of hESCs and hiPSCs with the aim to develop novel hiPSC-based tools for measuring pathway perturbation in relation to molecular and cellular effects in vitro. Among other fundamental pathways, also, the cAMP responsive element binding protein (CREB) pathway was activated in our neuronal models and gave us the opportunity to study time-dependent effects elicited by chemical perturbations of the CREB pathway in relation to cellular effects. We show that the inhibition of the CREB pathway, using 2-naphthol-AS-E-phosphate (KG-501), induced an inhibition of neurite outgrowth and synaptogenesis, as well as a decrease of MAP2(+) neuronal cells. These data indicate that a CREB pathway inhibition can be related to molecular and cellular effects that may be relevant for neurotoxicity testing, and, thus, qualify the use of our hiPSC-derived neuronal model for studying chemical-induced neurotoxicity resulting from pathway perturbations.
Collapse
Affiliation(s)
| | - Jochem Louisse
- Institute for Health and Consumer Protection (IHCP), JRC, Ispra, Italy
| | - Bibiana Scelfo
- Institute for Health and Consumer Protection (IHCP), JRC, Ispra, Italy
| | - Milena Mennecozzi
- Institute for Health and Consumer Protection (IHCP), JRC, Ispra, Italy
| | - Benedetta Accordi
- Oncohematology Laboratory, Department of Woman and Child Health, University of Padova, Padova, Italy
| | - Giuseppe Basso
- Oncohematology Laboratory, Department of Woman and Child Health, University of Padova, Padova, Italy
| | - John Antonydas Gaspar
- Center of Physiology and Pathophysiology, Institute of Neurophysiology, University of Cologne, Cologne, Germany
| | - Dimitra Zagoura
- Institute for Health and Consumer Protection (IHCP), JRC, Ispra, Italy
| | - Manuela Barilari
- Institute for Health and Consumer Protection (IHCP), JRC, Ispra, Italy
| | - Taina Palosaari
- Institute for Health and Consumer Protection (IHCP), JRC, Ispra, Italy
| | - Agapios Sachinidis
- Center of Physiology and Pathophysiology, Institute of Neurophysiology, University of Cologne, Cologne, Germany
| | | |
Collapse
|
309
|
Lecht S, Gerstenhaber JA, Stabler CT, Pimton P, Karamil S, Marcinkiewicz C, Schulman ES, Lelkes PI. Heterogeneous Mixed-Lineage Differentiation of Mouse Embryonic Stem Cells Induced by Conditioned Media from A549 Cells. Stem Cells Dev 2014; 23:1923-36. [DOI: 10.1089/scd.2014.0042] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Shimon Lecht
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, Pennsylvania
| | - Jonathan A. Gerstenhaber
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, Pennsylvania
| | - Collin T. Stabler
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, Pennsylvania
| | - Pimchanok Pimton
- Department of Biology, School of Science, Walailak University, Thammarat, Thailand
| | - Seda Karamil
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, Pennsylvania
| | - Cezary Marcinkiewicz
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, Pennsylvania
| | - Edward S. Schulman
- Division of Pulmonary, Critical Care and Sleep Medicine, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Peter I. Lelkes
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, Pennsylvania
| |
Collapse
|
310
|
Sun B, Yu W, Wang F, Song W, Jin H, Sun Y. Effects of group culture on the development of discarded human embryos and the construction of human embryonic stem cell lines. J Assist Reprod Genet 2014; 31:1369-76. [PMID: 25113620 DOI: 10.1007/s10815-014-0308-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 07/21/2014] [Indexed: 10/24/2022] Open
Abstract
PURPOSE To explore the effect of group culture on the developmental potential of discarded embryos in in vitro fertilization/intracytoplasmic sperm injection (IVF/ICSI) cycles and establish the human embryonic stem cell lines for future research. METHORDS Fresh discarded embryos were collected from the IVF/ICSI-ET program in the reproductive medical center of the first affiliated hospital of Zhengzhou university in this study. All zygotes were individually cultured from Day 1 to Day 3. On Day 3, discard embryos were then cultured in group of 1-4 embryos per droplet (30 μl/droplets) with a constant culture medium until Day 5 or 6. Mechanical method was used to isolate the inner cell mass (ICM) of blastocyst from the embryo. Then we inoculated the ICM on feeder layer. After identification of those cells, the human embryonic stem cell lines (hESCs) were established. RESULTS In this study, we collected 1,223 fresh discarded embryos and they were sequential cultured to the blastocysts (18.07 %, 221/1,223), in which good quality blastocysts were 61(4.98 %, 61/1,223). There was no significant difference in the patients. The embryos from 1PN, 2PN, 3PN were sequential cultured to the blastocyst s(39.31 %,92/234;12.87 %,64/497;13.21 %,65/492),in which good quality blastocysts was 13.6 %(32/92),2.61 %(13/64), 3.04 %(15/65).1PN embryo's blastulation rate and quality embryo formation rate was significantly higher than the 2PN and 3PN embryos' (P <0.05). Three embryos group cultivation has the highest blastulation rate and quality embryo formation rate (P <0.05). In total, we successfully established 4 hESCs lines. CONCLUSION The group culture of human discard embryos can improve the blastulation rate and blastocyst quality to some extent. Three embryos group cultivate is the better culture number. Human discard embryos are good source for establishment of hESCs.
Collapse
Affiliation(s)
- Bo Sun
- Reproductive Medical Center, First Affiliated Hospital of Zhengzhou University, Constructive East Road, Zhengzhou, 450052, China
| | | | | | | | | | | |
Collapse
|
311
|
Abstract
OPINION STATEMENT Reconstitution of cardiac muscle as well as blood vessels to provide sufficient oxygenation and nutrients to the myocardium is an important component of any therapeutic approach for cardiac repair after injury. Recent reports of reprogramming somatic cells directly to cells of another lineage raised the possibility of using cell reprogramming for cardiac regenerative therapy. Here, we provide an overview of the current reprogramming strategies to generate cardiomyocytes (CMs), endothelial cells (ECs) and smooth muscle cells (SMCs), and the implications of these methods for cardiac regeneration. We also discuss the challenges and limitations that need to be addressed for the development of future therapies.
Collapse
|
312
|
Menasché P, Vanneaux V, Fabreguettes JR, Bel A, Tosca L, Garcia S, Bellamy V, Farouz Y, Pouly J, Damour O, Périer MC, Desnos M, Hagège A, Agbulut O, Bruneval P, Tachdjian G, Trouvin JH, Larghero J. Towards a clinical use of human embryonic stem cell-derived cardiac progenitors: a translational experience. Eur Heart J 2014; 36:743-50. [PMID: 24835485 DOI: 10.1093/eurheartj/ehu192] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
AIM There is now compelling evidence that cells committed to a cardiac lineage are most effective for improving the function of infarcted hearts. This has been confirmed by our pre-clinical studies entailing transplantation of human embryonic stem cell (hESC)-derived cardiac progenitors in rat and non-human primate models of myocardial infarction. These data have paved the way for a translational programme aimed at a phase I clinical trial. METHODS AND RESULTS The main steps of this programme have included (i) the expansion of a clone of pluripotent hESC to generate a master cell bank under good manufacturing practice conditions (GMP); (ii) a growth factor-induced cardiac specification; (iii) the purification of committed cells by immunomagnetic sorting to yield a stage-specific embryonic antigen (SSEA)-1-positive cell population strongly expressing the early cardiac transcription factor Isl-1; (iv) the incorporation of these cells into a fibrin scaffold; (v) a safety assessment focused on the loss of teratoma-forming cells by in vitro (transcriptomics) and in vivo (cell injections in immunodeficient mice) measurements; (vi) an extensive cytogenetic and viral testing; and (vii) the characterization of the final cell product and its release criteria. The data collected throughout this process have led to approval by the French regulatory authorities for a first-in-man clinical trial of transplantation of these SSEA-1(+) progenitors in patients with severely impaired cardiac function. CONCLUSION Although several facets of this manufacturing process still need to be improved, these data may yet provide a useful platform for the production of hESC-derived cardiac progenitor cells under safe and cost-effective GMP conditions.
Collapse
Affiliation(s)
- Philippe Menasché
- Department of Cardiovascular Surgery, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, 20, rue Leblanc, 75015 Paris, France University Paris Descartes, Sorbonne Paris Cité, Paris F-75475, France INSERM U970, Hôpital Européen Georges Pompidou, Paris, France
| | - Valérie Vanneaux
- Cell Therapy Unit and Clinical Investigation Center in Biotherapies (CBT501), Assistance Publique-Hôpitaux de Paris, Hôpital Saint-Louis, Paris, France University Paris Diderot, Sorbonne Paris Cité, Paris F-75475, France INSERM UMRS1160, Institut Universitaire d'Hématologie, Hôpital Saint-Louis, Paris, France
| | - Jean-Roch Fabreguettes
- Central Pharmacy, Clinical Trials Department, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Alain Bel
- Department of Cardiovascular Surgery, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, 20, rue Leblanc, 75015 Paris, France INSERM U970, Hôpital Européen Georges Pompidou, Paris, France
| | - Lucie Tosca
- Assistance Publique-Hôpitaux de Paris, University Paris Sud, Histology-Embryology-Cytogenetics, Hôpitaux Universitaires Paris Sud, Clamart 92141, France
| | - Sylvie Garcia
- Unité de Biologie des Populations Lymphocytaires, Department of Immunology, Institut Pasteur, CNRS-URA 1961, Paris, France
| | - Valérie Bellamy
- INSERM U970, Hôpital Européen Georges Pompidou, Paris, France
| | - Yohan Farouz
- University Paris Descartes, Sorbonne Paris Cité, Paris F-75475, France INSERM U970, Hôpital Européen Georges Pompidou, Paris, France
| | - Julia Pouly
- Department of Cardiovascular Surgery, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, 20, rue Leblanc, 75015 Paris, France
| | - Odile Damour
- Tissues and Cells Bank, Edouard Herriot Hospital, Lyon, France
| | | | - Michel Desnos
- University Paris Descartes, Sorbonne Paris Cité, Paris F-75475, France INSERM U970, Hôpital Européen Georges Pompidou, Paris, France Department of Cardiology, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Paris, France
| | - Albert Hagège
- University Paris Descartes, Sorbonne Paris Cité, Paris F-75475, France INSERM U970, Hôpital Européen Georges Pompidou, Paris, France Department of Cardiology, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Paris, France
| | - Onnik Agbulut
- Sorbonne Universités, UPMC Univ Paris 06, UMR CNRS 8256, Biological Adaptation and Ageing, Paris, France
| | - Patrick Bruneval
- University Paris Descartes, Sorbonne Paris Cité, Paris F-75475, France INSERM U970, Hôpital Européen Georges Pompidou, Paris, France Department of Pathology, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Paris, France
| | - Gérard Tachdjian
- Assistance Publique-Hôpitaux de Paris, University Paris Sud, Histology-Embryology-Cytogenetics, Hôpitaux Universitaires Paris Sud, Clamart 92141, France
| | - Jean-Hugues Trouvin
- School of Pharmacy, University Paris Descartes, Paris, France Central Pharmacy, Pharmaceutical Innovation Department, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Jérôme Larghero
- Cell Therapy Unit and Clinical Investigation Center in Biotherapies (CBT501), Assistance Publique-Hôpitaux de Paris, Hôpital Saint-Louis, Paris, France University Paris Diderot, Sorbonne Paris Cité, Paris F-75475, France INSERM UMRS1160, Institut Universitaire d'Hématologie, Hôpital Saint-Louis, Paris, France
| |
Collapse
|
313
|
Schäfer R. Does the adult stroma contain stem cells? ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2014; 129:177-89. [PMID: 23117643 DOI: 10.1007/10_2012_160] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
It is well accepted that adult mesenchymal stromal cells (MSCs) comprise subpopulations of cells sharing common phenotypical and functional properties. However, there is emerging evidence that MSC subpopulations may also feature distinct characteristics. This chapter focuses on MSC subpopulations reflecting their possible stem cell properties relative to defined pluripotent stem cells (PSCs) such as embryonic stem cells (ESCs) or induced pluripotent stem cells (iPSCs). This attempt at an ontogenetic reflection on MSCs can be useful for both basic and translational research in the field.
Collapse
Affiliation(s)
- Richard Schäfer
- Department of Neurosurgery, Stanford University School of Medicine, 1201 Welch Road, Stanford, CA, 94305-5487, USA,
| |
Collapse
|
314
|
Chmielowiec J, Borowiak M. In vitro differentiation and expansion of human pluripotent stem cell-derived pancreatic progenitors. Rev Diabet Stud 2014; 11:19-34. [PMID: 25148365 DOI: 10.1900/rds.2014.11.19] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Recent progress in understanding stem cell biology has been remarkable, especially in deciphering signals that support differentiation towards tissue-specific lineages. This achievement positions us firmly at the beginning of an era of patient-specific regenerative medicine and human disease modeling. It will be necessary to equip the progress in this era with a reliable source of self-renewing progenitor cells that differentiate into functional target cells. The generation of pancreatic progenitors that mature in vivo into functional beta-cells has raised the hope for new therapeutic options in diabetes, but key challenges still remain including the production of sufficient numbers of cells for research and transplantation. Recent approaches to this problem have shown that the presence of organ- and stage-specific mesenchyme improves the generation of progenitors, from endoderm to endocrine cells. Alternatively, utilization of three-dimensional culture may improve the efficiency and yield of directed differentiation. Here, we review the current knowledge of pancreatic directed differentiation and ex vivo expansion of pancreatic progenitors, including recent advances in differentiation strategies for the generation of pancreatic progenitors, and we discuss persistent challenges which will need to be overcome before personalized cell-based therapy becomes a practical strategy.
Collapse
Affiliation(s)
- Jolanta Chmielowiec
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Malgorzata Borowiak
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| |
Collapse
|
315
|
Sterthaus O, Feutz AC, Zhang H, Pletscher F, Bruder E, Miny P, Lezzi G, De Geyter M, De Geyter C. Gene expression profiles of similarly derived human embryonic stem cell lines correlate with their distinct propensity to exit stemness and their different differentiation behavior in culture. Cell Reprogram 2014; 16:185-95. [PMID: 24811852 DOI: 10.1089/cell.2013.0089] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Four normal-karyotype human embryonic stem cell (hESC) lines were generated using the same protocol and maintained under identical conditions. Despite these precautions, gene expression patterns were found to be dissimilar among the four lines. The observed differences were typical of each cell line, correlated with their distinct propensity to exit stemness, created heterogeneity among the cells during cell line maintenance, and correlated with their altered capacity as a source of differentiated cells. The capacity of some cell lines to give rise to more, and more mature, neurons within comparable time frames of directed differentiation reflected the distinct proportions of cells already predifferentiated at the onset. These findings demonstrate that the subsequent stages of neural differentiation were altered both in a quantitative and timely fashion. As a consequence, cell lines with apparent better and quicker ability to produce neurons were actually the less capable of reproducing proper differentiation. Previous data suggested that cell lines able to generate more neurons faster would be more suitable to clinical application. Our analysis of the differentiation process strongly suggests the opposite. The spontaneous tendency to predifferentiate of any particular hESC line should be known because it clearly impacts further experimental results.
Collapse
Affiliation(s)
- Oliver Sterthaus
- 1 Clinic of Gynecological Endocrinology and Reproductive Medicine, University of Basel , CH-4031, Basel, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
316
|
Methionine metabolism regulates maintenance and differentiation of human pluripotent stem cells. Cell Metab 2014; 19:780-94. [PMID: 24746804 DOI: 10.1016/j.cmet.2014.03.017] [Citation(s) in RCA: 394] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2013] [Revised: 01/09/2014] [Accepted: 03/11/2014] [Indexed: 12/26/2022]
Abstract
Mouse embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) are in a high-flux metabolic state, with a high dependence on threonine catabolism. However, little is known regarding amino acid metabolism in human ESCs/iPSCs. We show that human ESCs/iPSCs require high amounts of methionine (Met) and express high levels of enzymes involved in Met metabolism. Met deprivation results in a rapid decrease in intracellular S-adenosylmethionine (SAM), triggering the activation of p53-p38 signaling, reducing NANOG expression, and poising human iPSC/ESCs for differentiation, follow by potentiated differentiation into all three germ layers. However, when exposed to prolonged Met deprivation, the cells undergo apoptosis. We also show that human ESCs/iPSCs have regulatory systems to maintain constant intracellular Met and SAM levels. Our findings show that SAM is a key regulator for maintaining undifferentiated pluripotent stem cells and regulating their differentiation.
Collapse
|
317
|
Dajani R, Koo SE, Sullivan GJ, Park IH. Investigation of Rett syndrome using pluripotent stem cells. J Cell Biochem 2014; 114:2446-53. [PMID: 23744605 DOI: 10.1002/jcb.24597] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Accepted: 05/28/2013] [Indexed: 01/23/2023]
Abstract
Rett syndrome (RTT) is one of most prevalent female neurodevelopmental disorders. De novo mutations in X-linked MECP2 are mostly responsible for RTT. Since the identification of MeCP2 as the underlying cause of RTT, murine models have contributed to understanding the pathophysiology of RTT and function of MeCP2. Reprogramming is a procedure to produce induced pluripotent stem cells (iPSCs) by overexpression of four transcription factors. iPSCs obtain similar features as embryonic stem cells and are capable of self-renewing and differentiating into cells of all three layers. iPSCs have been utilized in modeling human diseases in vitro. Neurons differentiated from RTT-iPSCs showed the recapitulation of RTT phenotypes. Despite the early success, genetic and epigenetic instability upon reprogramming and ensuing maintenance of iPSCs raise concerns in using RTT-iPSCs as an accurate in vitro model. Here, we update the current iPSC-based RTT modeling, and its concerns and challenges.
Collapse
Affiliation(s)
- Rana Dajani
- Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, 10 Amistad 201B, New Haven, Connecticut, 06520; Department of Biology and Biotechnology, Hashemite University, Zarqa, P.O. Box 150459 13133, Jordan
| | | | | | | |
Collapse
|
318
|
De Paepe C, Krivega M, Cauffman G, Geens M, Van de Velde H. Totipotency and lineage segregation in the human embryo. ACTA ACUST UNITED AC 2014; 20:599-618. [DOI: 10.1093/molehr/gau027] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
319
|
Martins AM, Vunjak-Novakovic G, Reis RL. The current status of iPS cells in cardiac research and their potential for tissue engineering and regenerative medicine. Stem Cell Rev Rep 2014; 10:177-90. [PMID: 24425421 PMCID: PMC4476262 DOI: 10.1007/s12015-013-9487-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The recent availability of human cardiomyocytes derived from induced pluripotent stem (iPS) cells opens new opportunities to build in vitro models of cardiac disease, screening for new drugs, and patient-specific cardiac therapy. Notably, the use of iPS cells enables studies in the wide pool of genotypes and phenotypes. We describe progress in reprogramming of induced pluripotent stem (iPS) cells towards the cardiac lineage/differentiation. The focus is on challenges of cardiac disease modeling using iPS cells and their potential to produce safe, effective and affordable therapies/applications with the emphasis of cardiac tissue engineering. We also discuss implications of human iPS cells to biological research and some of the future needs.
Collapse
Affiliation(s)
- Ana M. Martins
- 3B’s Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Guimarães, Portugal. ICVS/3B’s-PT Government Associate Laboratory, Braga/Guimarães, Portugal. Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | | | - Rui L. Reis
- 3B’s Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Guimarães, Portugal. ICVS/3B’s-PT Government Associate Laboratory, Braga/Guimarães, Portugal. Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, AvePark, Zona Industrial da Gandra, S. Cláudio do Barco, 4806-909 Caldas das Taipas, Guimarães, Portugal
| |
Collapse
|
320
|
From pluripotency to forebrain patterning: an in vitro journey astride embryonic stem cells. Cell Mol Life Sci 2014; 71:2917-30. [PMID: 24643740 PMCID: PMC4098049 DOI: 10.1007/s00018-014-1596-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Revised: 02/17/2014] [Accepted: 02/26/2014] [Indexed: 02/07/2023]
Abstract
Embryonic stem cells (ESCs) have been used extensively as in vitro models of neural development and disease, with special efforts towards their conversion into forebrain progenitors and neurons. The forebrain is the most complex brain region, giving rise to several fundamental structures, such as the cerebral cortex, the hypothalamus, and the retina. Due to the multiplicity of signaling pathways playing different roles at distinct times of embryonic development, the specification and patterning of forebrain has been difficult to study in vivo. Research performed on ESCs in vitro has provided a large body of evidence to complement work in model organisms, but these studies have often been focused more on cell type production than on cell fate regulation. In this review, we systematically reassess the current literature in the field of forebrain development in mouse and human ESCs with a focus on the molecular mechanisms of early cell fate decisions, taking into consideration the specific culture conditions, exogenous and endogenous molecular cues as described in the original studies. The resulting model of early forebrain induction and patterning provides a useful framework for further studies aimed at reconstructing forebrain development in vitro for basic research or therapy.
Collapse
|
321
|
Holditch SJ, Terzic A, Ikeda Y. Concise review: pluripotent stem cell-based regenerative applications for failing β-cell function. Stem Cells Transl Med 2014; 3:653-61. [PMID: 24646490 DOI: 10.5966/sctm.2013-0184] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Diabetes engenders the loss of pancreatic β-cell mass and/or function, resulting in insulin deficiency relative to the metabolic needs of the body. Diabetic care has traditionally relied on pharmacotherapy, exemplified by insulin replacement to target peripheral actions of the hormone. With growing understanding of the pathogenesis of diabetic disease, alternative approaches aiming at repair and restoration of failing β-cell function are increasingly considered as complements to current diabetes therapy regimens. To this end, emphasis is placed on transplantation of exogenous pancreas/islets or artificial islets, enhanced proliferation and maturation of endogenous β cells, prevention of β-cell loss, or fortified renewal of β-like-cell populations from stem cell pools and non-β-cell sources. In light of emerging clinical experiences with human embryonic stem cells and approval of the first in-human trial with induced pluripotent stem cells, in this study we highlight advances in β-cell regeneration strategies with a focus on pluripotent stem cell platforms in the context of translational applications.
Collapse
Affiliation(s)
- Sara J Holditch
- Center for Regenerative Medicine, Department of Molecular Medicine, Division of Cardiovascular Diseases, Department of Medicine, Department of Molecular Pharmacology and Experimental Therapeutics, and Department of Medical Genetics, Mayo Clinic, Rochester, Minnesota, USA
| | | | | |
Collapse
|
322
|
Haidet-Phillips AM, Roybon L, Gross SK, Tuteja A, Donnelly CJ, Richard JP, Ko M, Sherman A, Eggan K, Henderson CE, Maragakis NJ. Gene profiling of human induced pluripotent stem cell-derived astrocyte progenitors following spinal cord engraftment. Stem Cells Transl Med 2014; 3:575-85. [PMID: 24604284 DOI: 10.5966/sctm.2013-0153] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The generation of human induced pluripotent stem cells (hiPSCs) represents an exciting advancement with promise for stem cell transplantation therapies as well as for neurological disease modeling. Based on the emerging roles for astrocytes in neurological disorders, we investigated whether hiPSC-derived astrocyte progenitors could be engrafted to the rodent spinal cord and how the characteristics of these cells changed between in vitro culture and after transplantation to the in vivo spinal cord environment. Our results show that human embryonic stem cell- and hiPSC-derived astrocyte progenitors survive long-term after spinal cord engraftment and differentiate to astrocytes in vivo with few cells from other lineages present. Gene profiling of the transplanted cells demonstrates the astrocyte progenitors continue to mature in vivo and upregulate a variety of astrocyte-specific genes. Given this mature astrocyte gene profile, this work highlights hiPSCs as a tool to investigate disease-related astrocyte biology using in vivo disease modeling with significant implications for human neurological diseases currently lacking animal models.
Collapse
Affiliation(s)
- Amanda M Haidet-Phillips
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Departments of Rehabilitation and Regenerative Medicine, Pathology and Cell Biology, Neurology, and Neuroscience, Center for Motor Neuron Biology and Disease, Columbia Stem Cell Initiative, Columbia University Medical Center, New York, New York, Stem Cell Laboratory for CNS Disease Modeling-MultiPark, Department of Experimental Medical Science, Lund University, Lund, Sweden; Howard Hughes Medical Institute, Harvard Stem Cell Institute and Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
323
|
Srikanth P, Young-Pearse TL. Stem cells on the brain: modeling neurodevelopmental and neurodegenerative diseases using human induced pluripotent stem cells. J Neurogenet 2014; 28:5-29. [PMID: 24628482 PMCID: PMC4285381 DOI: 10.3109/01677063.2014.881358] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Seven years have passed since the initial report of the generation of induced pluripotent stem cells (iPSCs) from adult human somatic cells, and in the intervening time the field of neuroscience has developed numerous disease models using this technology. Here, we review progress in the field and describe both the advantages and potential pitfalls of modeling neurodegenerative and neurodevelopmental diseases using this technology. We include tables with information on neural differentiation protocols and studies that developed human iPSC lines to model neurological diseases. We also discuss how one can: investigate effects of genetic mutations with iPSCs, examine cell fate-specific phenotypes, best determine the specificity of a phenotype, and bring in vivo relevance to this in vitro technique.
Collapse
Affiliation(s)
- Priya Srikanth
- Center for Neurologic Diseases, Brigham and Women's Hospital , Boston, Massachusetts , USA
| | | |
Collapse
|
324
|
Chan SSK, Shi X, Toyama A, Arpke RW, Dandapat A, Iacovino M, Kang J, Le G, Hagen HR, Garry DJ, Kyba M. Mesp1 patterns mesoderm into cardiac, hematopoietic, or skeletal myogenic progenitors in a context-dependent manner. Cell Stem Cell 2014; 12:587-601. [PMID: 23642367 DOI: 10.1016/j.stem.2013.03.004] [Citation(s) in RCA: 142] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2012] [Revised: 12/12/2012] [Accepted: 03/14/2013] [Indexed: 02/07/2023]
Abstract
Mesp1 is regarded as the master regulator of cardiovascular development, initiating the cardiac transcription factor cascade to direct the generation of cardiac mesoderm. To define the early embryonic cell population that responds to Mesp1, we performed pulse inductions of gene expression over tight temporal windows following embryonic stem cell differentiation. Remarkably, instead of promoting cardiac differentiation in the initial wave of mesoderm, Mesp1 binds to the Tal1 (Scl) +40 kb enhancer and generates Flk-1+ precursors expressing Etv2 (ER71) and Tal1 that undergo hematopoietic differentiation. The second wave of mesoderm responds to Mesp1 by differentiating into PDGFRα+ precursors that undergo cardiac differentiation. Furthermore, in the absence of serum-derived factors, Mesp1 promotes skeletal myogenic differentiation. Lineage tracing revealed that the majority of yolk sac and many adult hematopoietic cells derive from Mesp1+ precursors. Thus, Mesp1 is a context-dependent determination factor, integrating the stage of differentiation and the signaling environment to specify different lineage outcomes.
Collapse
Affiliation(s)
- Sunny Sun-Kin Chan
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
325
|
Abstract
Human pluripotent stem cells (hPSCs) have the potential to generate any human cell type, and one widely recognized goal is to make pancreatic β cells. To this end, comparisons between differentiated cell types produced in vitro and their in vivo counterparts are essential to validate hPSC-derived cells. Genome-wide transcriptional analysis of sorted insulin-expressing (INS(+)) cells derived from three independent hPSC lines, human fetal pancreata, and adult human islets points to two major conclusions: (i) Different hPSC lines produce highly similar INS(+) cells and (ii) hPSC-derived INS(+) (hPSC-INS(+)) cells more closely resemble human fetal β cells than adult β cells. This study provides a direct comparison of transcriptional programs between pure hPSC-INS(+) cells and true β cells and provides a catalog of genes whose manipulation may convert hPSC-INS(+) cells into functional β cells.
Collapse
|
326
|
Small-molecule induction promotes corneal epithelial cell differentiation from human induced pluripotent stem cells. Stem Cell Reports 2014; 2:219-31. [PMID: 24527395 PMCID: PMC3923224 DOI: 10.1016/j.stemcr.2013.12.014] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Revised: 12/23/2013] [Accepted: 12/23/2013] [Indexed: 12/21/2022] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) offer unique opportunities for developing novel cell-based therapies and disease modeling. In this study, we developed a directed differentiation method for hiPSCs toward corneal epithelial progenitor cells capable of terminal differentiation toward mature corneal epithelial-like cells. In order to improve the efficiency and reproducibility of our method, we replicated signaling cues active during ocular surface ectoderm development with the help of two small-molecule inhibitors in combination with basic fibroblast growth factor (bFGF) in serum-free and feeder-free conditions. First, small-molecule induction downregulated the expression of pluripotency markers while upregulating several transcription factors essential for normal eye development. Second, protein expression of the corneal epithelial progenitor marker p63 was greatly enhanced, with up to 95% of cells being p63 positive after 5 weeks of differentiation. Third, corneal epithelial-like cells were obtained upon further maturation. Small-molecule induction directs early stage differentiation Subsequent maturation yields homogeneous populations of p63-positive cells p63-positive progenitor cells are capable of terminal differentiation The serum-free and feeder-free method can be upgraded to fully defined and xeno free
Collapse
|
327
|
Cai Q, Bonfanti P, Sambathkumar R, Vanuytsel K, Vanhove J, Gysemans C, Debiec-Rychter M, Raitano S, Heimberg H, Ordovas L, Verfaillie CM. Prospectively isolated NGN3-expressing progenitors from human embryonic stem cells give rise to pancreatic endocrine cells. Stem Cells Transl Med 2014; 3:489-99. [PMID: 24493854 DOI: 10.5966/sctm.2013-0078] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Pancreatic endocrine progenitors obtained from human embryonic stem cells (hESCs) represent a promising source to develop cell-based therapies for diabetes. Although endocrine pancreas progenitor cells have been isolated from mouse pancreata on the basis of Ngn3 expression, human endocrine progenitors have not been isolated yet. As substantial differences exist between human and murine pancreas biology, we investigated whether it is possible to isolate pancreatic endocrine progenitors from differentiating hESC cultures by lineage tracing of NGN3. We targeted the 3' end of NGN3 using zinc finger nuclease-mediated homologous recombination to allow selection of NGN3eGFP(+) cells without disrupting the coding sequence of the gene. Isolated NGN3eGFP(+) cells express PDX1, NKX6.1, and chromogranin A and differentiate in vivo toward insulin, glucagon, and somatostatin single hormone-expressing cells but not to ductal or exocrine pancreatic cells or other endodermal, mesodermal, or ectodermal lineages. This confirms that NGN3(+) cells represent pancreatic endocrine progenitors in humans. In addition, this hESC reporter line constitutes a unique tool that may aid in gaining insight into the developmental mechanisms underlying fate choices in human pancreas and in developing cell-based therapies.
Collapse
Affiliation(s)
- Qing Cai
- Stamcelinstituut Leuven, Laboratory for Experimental Medicine and Endocrinology, Department of Human Genetics, and University Hospitals Leuven, KU Leuven, Leuven, Belgium; Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
328
|
Guloglu MO, Larsen A, Brundin P. Adipocytes derived from PA6 cells reliably promote the differentiation of dopaminergic neurons from human embryonic stem cells. J Neurosci Res 2014; 92:564-73. [PMID: 24482287 DOI: 10.1002/jnr.23355] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 11/26/2013] [Accepted: 11/30/2013] [Indexed: 11/09/2022]
Abstract
The PA6 stromal cell line comprises a heterogeneous population of cells that can induce both mouse and human embryonic stem cells to differentiate into dopaminergic neurons. This ability of PA6 cells has been termed stromal cell-derived inducing activity (SDIA). The level of SDIA has been found to vary considerably between and within batches of PA6 cells. Not only are the molecular mechanisms that underlie SDIA unknown but also the cell type(s) within the heterogeneous PA6 cultures that underlie SDIA remain poorly defined. In this study, we reveal that adipocytes, which are present within the heterogeneous PA6 cell population, robustly release the factors mediating SDIA. Furthermore, we report that the coculture of human embryonic stem cells with PA6-derived adipocytes reliably induces their differentiation into midbrain dopaminergic neurons.
Collapse
Affiliation(s)
- M O Guloglu
- Neuronal Survival Unit, Department of Experimental Medical Science, Wallenberg Neuroscience Center, Lund, Sweden
| | | | | |
Collapse
|
329
|
Araoka T, Mae SI, Kurose Y, Uesugi M, Ohta A, Yamanaka S, Osafune K. Efficient and rapid induction of human iPSCs/ESCs into nephrogenic intermediate mesoderm using small molecule-based differentiation methods. PLoS One 2014; 9:e84881. [PMID: 24454758 PMCID: PMC3893162 DOI: 10.1371/journal.pone.0084881] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Accepted: 11/20/2013] [Indexed: 02/07/2023] Open
Abstract
The first step in developing regenerative medicine approaches to treat renal diseases using pluripotent stem cells must be the generation of intermediate mesoderm (IM), an embryonic germ layer that gives rise to kidneys. In order to achieve this goal, establishing an efficient, stable and low-cost method for differentiating IM cells using small molecules is required. In this study, we identified two retinoids, AM580 and TTNPB, as potent IM inducers by high-throughput chemical screening, and established rapid (five days) and efficient (80% induction rate) IM differentiation from human iPSCs using only two small molecules: a Wnt pathway activator, CHIR99021, combined with either AM580 or TTNPB. The resulting human IM cells showed the ability to differentiate into multiple cell types that constitute adult kidneys, and to form renal tubule-like structures. These small molecule differentiation methods can bypass the mesendoderm step, directly inducing IM cells by activating Wnt, retinoic acid (RA), and bone morphogenetic protein (BMP) pathways. Such methods are powerful tools for studying kidney development and may potentially provide cell sources to generate renal lineage cells for regenerative therapy.
Collapse
Affiliation(s)
- Toshikazu Araoka
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Shin-ichi Mae
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Yuko Kurose
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Motonari Uesugi
- Institute for Integrated Cell–Material Sciences, Kyoto University, Kyoto, Japan
- Institute for Chemical Research, Kyoto University, Kyoto, Japan
| | - Akira Ohta
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Shinya Yamanaka
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- Gladstone Institute of Cardiovascular Disease, San Francisco, California, United States of America
| | - Kenji Osafune
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| |
Collapse
|
330
|
Nishio M, Saeki K. Differentiation of human pluripotent stem cells into highly functional classical brown adipocytes. Methods Enzymol 2014; 537:177-97. [PMID: 24480347 DOI: 10.1016/b978-0-12-411619-1.00010-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
We describe a detailed method for directed differentiation of human pluripotent stem cells, including human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs), into functional classical brown adipocytes (BAs) under serum-free and feeder-free conditions. It is a two-tiered culture system, based on very simple techniques, a floating culture and a subsequent adherent culture. It does not require gene transfer. The entire process can be carried out in about 10 days. The key point is the usage of our special hematopoietic cytokine cocktail. Almost all the differentiated cells express uncoupling protein 1, a BA-selective marker, as determined by immunostaining. The differentiated cells show characteristics of classical BA as assessed by morphology and gene/protein expression. Moreover, the expression of myoblast marker genes is transiently induced during the floating culture step. hESC/hiPSC-derived BAs show significantly higher oxygen consumption rates (OCRs) than white adipocytes generated from human mesenchymal stem cell. They also show responsiveness to adrenergic stimuli, with about twofold upregulation in OCR by β-adrenergic receptor (β-AR) agonist treatments. hESC/hiPSC-derived BAs exert in vivo calorigenic activities in response to β-AR agonist treatments as assessed by thermography. Finally, lipid and glucose metabolisms are significantly improved in hESC/hiPSC-derived BA-transplanted mice. Our system provides a highly feasible way to produce functional classical BA bearing metabolism-improving capacities from hESC/hiPSC under a feeder-free and serum-free condition without gene transfer.
Collapse
Affiliation(s)
- Miwako Nishio
- Department of Disease Control, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Kumiko Saeki
- Department of Disease Control, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan.
| |
Collapse
|
331
|
Prilutsky D, Palmer NP, Smedemark-Margulies N, Schlaeger TM, Margulies DM, Kohane IS. iPSC-derived neurons as a higher-throughput readout for autism: promises and pitfalls. Trends Mol Med 2013; 20:91-104. [PMID: 24374161 DOI: 10.1016/j.molmed.2013.11.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Revised: 11/20/2013] [Accepted: 11/21/2013] [Indexed: 12/13/2022]
Abstract
The elucidation of disease etiologies and establishment of robust, scalable, high-throughput screening assays for autism spectrum disorders (ASDs) have been impeded by both inaccessibility of disease-relevant neuronal tissue and the genetic heterogeneity of the disorder. Neuronal cells derived from induced pluripotent stem cells (iPSCs) from autism patients may circumvent these obstacles and serve as relevant cell models. To date, derived cells are characterized and screened by assessing their neuronal phenotypes. These characterizations are often etiology-specific or lack reproducibility and stability. In this review, we present an overview of efforts to study iPSC-derived neurons as a model for autism, and we explore the plausibility of gene expression profiling as a reproducible and stable disease marker.
Collapse
Affiliation(s)
- Daria Prilutsky
- Center for Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
| | - Nathan P Palmer
- Center for Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
| | | | | | - David M Margulies
- Center for Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA; Divisions of Genetics and Developmental Medicine, Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Isaac S Kohane
- Center for Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
332
|
Lammens T, D'hont I, D'Herde K, Benoit Y, Diez-Fraile A. Long non-coding RNAs in pluripotent stem cell biology. Vet Q 2013; 33:202-6. [PMID: 24256470 DOI: 10.1080/01652176.2013.866297] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Pluripotent stem cells are defined by their unlimited self-renewal capacities and potential to differentiate into any cell lineage. Many crucial determinants for the induction and maintenance of this pluripotent state have been identified. Long non-coding RNAs have recently emerged as key regulators of pluripotent stem cells and have enhanced our understanding of their potential functions in tissue regeneration. This review provides an overview of recent important insights into the roles of long non-coding RNAs as regulators and markers of pluripotency.
Collapse
Affiliation(s)
- Tim Lammens
- a Department of Pediatric Hematology-Oncology and Stem Cell Transplantation , Ghent University Hospital, 3K12D , De Pintelaan 185, 9000 Ghent , Belgium
| | | | | | | | | |
Collapse
|
333
|
Kondo Y, Iwao T, Nakamura K, Sasaki T, Takahashi S, Kamada N, Matsubara T, Gonzalez FJ, Akutsu H, Miyagawa Y, Okita H, Kiyokawa N, Toyoda M, Umezawa A, Nagata K, Matsunaga T, Ohmori S. An efficient method for differentiation of human induced pluripotent stem cells into hepatocyte-like cells retaining drug metabolizing activity. Drug Metab Pharmacokinet 2013; 29:237-43. [PMID: 24334537 DOI: 10.2133/dmpk.dmpk-13-rg-104] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The use of human induced pluripotent stem (iPS) cells would be of great value for a variety of applications involving drug development studies. Several reports have been published on the differentiation of human iPS cells into hepatocyte-like cells; however, the cells were insufficient for application in drug metabolism studies. In this study, we aimed to establish effective methods for differentiation of human iPS cells into hepatocytes. Two human iPS cell lines were differentiated by addition of activin A, dimethyl sulfoxide, hepatocyte growth factor, oncostatin M, and dexamethasone. The differentiated cells expressed hepatocyte markers and drug-metabolizing enzymes, revealing that the human iPS cells were differentiated into hepatocyte-like cells. Expression of CYP3A4 and UGT1A1 mRNAs increased with treatment with typical inducers of the enzymes, and the response of the cells against the inducers was similar to that of human hepatocytes. Furthermore, the drug-metabolizing activity of CYP3A4, as monitored by testosterone 6β-hydroxylase activity, was elevated by these inducers. In conclusion, we established methods for differentiation of hepatocyte-like cells expressing drug metabolizing activity from human iPS cells. The hepatocyte-like cells derived from human iPS cells will be useful for drug metabolism studies.
Collapse
Affiliation(s)
- Yuki Kondo
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
334
|
Huang HN, Chen SY, Hwang SM, Yu CC, Su MW, Mai W, Wang HW, Cheng WC, Schuyler SC, Ma N, Lu FL, Lu J. miR-200c and GATA binding protein 4 regulate human embryonic stem cell renewal and differentiation. Stem Cell Res 2013; 12:338-53. [PMID: 24365599 DOI: 10.1016/j.scr.2013.11.009] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2013] [Revised: 11/11/2013] [Accepted: 11/20/2013] [Indexed: 10/26/2022] Open
Abstract
Human embryonic stem cells (hESCs) are functionally unique for their self-renewal ability and pluripotency, but the molecular mechanisms giving rise to these properties are not fully understood. hESCs can differentiate into embryoid bodies (EBs) containing ectoderm, mesoderm, and endoderm. In the miR-200 family, miR-200c was especially enriched in undifferentiated hESCs and significantly downregulated in EBs. The knockdown of the miR-200c in hESCs downregulated Nanog expression, upregulated GATA binding protein 4 (GATA4) expression, and induced hESC apoptosis. The knockdown of GATA4 rescued hESC apoptosis induced by downregulation of miR-200c. miR-200c directly targeted the 3'-untranslated region of GATA4. Interestingly, the downregulation of GATA4 significantly inhibited EB formation in hESCs. Overexpression of miR-200c inhibited EB formation and repressed the expression of ectoderm, endoderm, and mesoderm markers, which could partially be rescued by ectopic expression of GATA4. Fibroblast growth factor (FGF) and activin A/nodal can sustain hESC renewal in the absence of feeder layer. Inhibition of transforming growth factor-β (TGF-β[Symbol: see text])/activin A/nodal signaling by SB431542 treatment downregulated the expression of miR-200c. Overexpression of miR-200c partially rescued the expression of Nanog/phospho-Smad2 that was downregulated by SB431542 treatment. Our observations have uncovered novel functions of miR-200c and GATA4 in regulating hESC renewal and differentiation.
Collapse
Affiliation(s)
- Hsiao-Ning Huang
- Genomics Research Center, Academia Sinica, Taipei, Taiwan; Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan
| | - Shao-Yin Chen
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Shiaw-Min Hwang
- Bioresource Collection and Research Center, Food Industry Research and Development Institute, Hsinchu, Taiwan
| | - Ching-Chia Yu
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Ming-Wei Su
- National RNAi Platform/National Core Facility Program for Biotechnology, Taipei, Taiwan; Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Wei Mai
- National RNAi Platform/National Core Facility Program for Biotechnology, Taipei, Taiwan; Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Hsei-Wei Wang
- VGH-YM Genomic Research Center, National Yang-Ming University, Taipei, Taiwan; Institute of Biomedical Informatics, National Yang-Ming University, Taipei, Taiwan; Institute of Microbiology and Immunology, National Yang-Ming University, Taipei, Taiwan; Cancer Research Center, National Yang-Ming University, Taipei, Taiwan; Department of Education and Research, Taipei City Hospital, Taipei, Taiwan
| | - Wei-Chung Cheng
- VGH-YM Genomic Research Center, National Yang-Ming University, Taipei, Taiwan; Division of Pediatric Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Scott C Schuyler
- Department of Biomedical Science, College of Medicine, Chang Gung University, Taoyuan County, Taiwan
| | - Nianhan Ma
- Institute of Systems Biology and Bioinformatics, National Central University, Taoyuan, Taiwan
| | - Frank Leigh Lu
- Department of Pediatrics, National Taiwan University Hospital, National Taiwan University Medical College, Taipei, Taiwan
| | - Jean Lu
- Genomics Research Center, Academia Sinica, Taipei, Taiwan; Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan; National RNAi Platform/National Core Facility Program for Biotechnology, Taipei, Taiwan; Institute of Microbiology and Immunology, National Yang-Ming University, Taipei, Taiwan; Genomics and System Biology Program, College of Life Science, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
335
|
Huang SXL, Islam MN, O'Neill J, Hu Z, Yang YG, Chen YW, Mumau M, Green MD, Vunjak-Novakovic G, Bhattacharya J, Snoeck HW. Efficient generation of lung and airway epithelial cells from human pluripotent stem cells. Nat Biotechnol 2013; 32:84-91. [PMID: 24291815 PMCID: PMC4101921 DOI: 10.1038/nbt.2754] [Citation(s) in RCA: 439] [Impact Index Per Article: 36.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Accepted: 10/30/2013] [Indexed: 12/14/2022]
Abstract
The ability to generate lung and airway epithelial cells from human pluripotent stem cells (hPSCs) would have applications in regenerative medicine, drug screening and modeling of lung disease, and studies of human lung development. We established, based on developmental paradigms, a highly efficient method for directed differentiation of hPSCs into lung and airway epithelial cells. Long-term differentiation in vivo and in vitro yielded basal, goblet, Clara, ciliated, type I and type II alveolar epithelial cells. Type II alveolar epithelial cells generated were capable of surfactant protein-B uptake and stimulated surfactant release, providing evidence of specific function. Inhibiting or removing agonists to signaling pathways critical for early lung development in the mouse—retinoic acid, Wnt and BMP—recapitulated defects in corresponding genetic mouse knockouts. The capability of this protocol to generate most cell types of the respiratory system suggests its utility for deriving patient-specific therapeutic cells.
Collapse
Affiliation(s)
- Sarah X L Huang
- 1] Columbia Center for Translational Immunology, Columbia University Medical Center, New York, New York, USA. [2] Department of Medicine, Columbia University Medical Center, New York, New York, USA
| | | | - John O'Neill
- Department of Biomedical Engineering, Columbia University, New York, New York, USA
| | - Zheng Hu
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, New York, USA
| | - Yong-Guang Yang
- 1] Columbia Center for Translational Immunology, Columbia University Medical Center, New York, New York, USA. [2] Department of Medicine, Columbia University Medical Center, New York, New York, USA
| | - Ya-Wen Chen
- 1] Columbia Center for Translational Immunology, Columbia University Medical Center, New York, New York, USA. [2] Department of Medicine, Columbia University Medical Center, New York, New York, USA
| | - Melanie Mumau
- 1] Columbia Center for Translational Immunology, Columbia University Medical Center, New York, New York, USA. [2] Department of Medicine, Columbia University Medical Center, New York, New York, USA
| | - Michael D Green
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, New York, USA
| | - Gordana Vunjak-Novakovic
- 1] Department of Medicine, Columbia University Medical Center, New York, New York, USA. [2] Department of Biomedical Engineering, Columbia University, New York, New York, USA
| | - Jahar Bhattacharya
- 1] Department of Medicine, Columbia University Medical Center, New York, New York, USA. [2] Department of Physiology & Cellular Biophysics, Columbia University Medical Center, New York, New York, USA
| | - Hans-Willem Snoeck
- 1] Columbia Center for Translational Immunology, Columbia University Medical Center, New York, New York, USA. [2] Department of Medicine, Columbia University Medical Center, New York, New York, USA. [3] Department of Microbiology and Immunology, Columbia University Medical Center, New York, New York, USA
| |
Collapse
|
336
|
Hazeltine LB, Selekman JA, Palecek SP. Engineering the human pluripotent stem cell microenvironment to direct cell fate. Biotechnol Adv 2013; 31:1002-19. [PMID: 23510904 PMCID: PMC3758782 DOI: 10.1016/j.biotechadv.2013.03.002] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Revised: 02/20/2013] [Accepted: 03/11/2013] [Indexed: 01/31/2023]
Abstract
Human pluripotent stem cells (hPSCs), including both embryonic stem cells and induced pluripotent stem cells, offer a potential cell source for research, drug screening, and regenerative medicine applications due to their unique ability to self-renew or differentiate to any somatic cell type. Before the full potential of hPSCs can be realized, robust protocols must be developed to direct their fate. Cell fate decisions are based on components of the surrounding microenvironment, including soluble factors, substrate or extracellular matrix, cell-cell interactions, mechanical forces, and 2D or 3D architecture. Depending on their spatio-temporal context, these components can signal hPSCs to either self-renew or differentiate to cell types of the ectoderm, mesoderm, or endoderm. Researchers working at the interface of engineering and biology have identified various factors which can affect hPSC fate, often based on lessons from embryonic development, and they have utilized this information to design in vitro niches which can reproducibly direct hPSC fate. This review highlights culture systems that have been engineered to promote self-renewal or differentiation of hPSCs, with a focus on studies that have elucidated the contributions of specific microenvironmental cues in the context of those culture systems. We propose the use of microsystem technologies for high-throughput screening of spatial-temporal presentation of cues, as this has been demonstrated to be a powerful approach for differentiating hPSCs to desired cell types.
Collapse
Affiliation(s)
| | | | - Sean P. Palecek
- Department of Chemical and Biological Engineering, University of Wisconsin – Madison 1415 Engineering Drive, Madison, WI 53706 USA
| |
Collapse
|
337
|
Abstract
Post-translational modifications (PTMs) are known to be essential mechanisms used by eukaryotic cells to diversify their protein functions and dynamically coordinate their signaling networks. Defects in PTMs have been linked to numerous developmental disorders and human diseases, highlighting the importance of PTMs in maintaining normal cellular states. Human pluripotent stem cells (hPSCs), including embryonic stem cells (hESCs) and induced pluripotent stem cells (hiPSCs), are capable of self-renewal and differentiation into a variety of functional somatic cells; these cells hold a great promise for the advancement of biomedical research and clinical therapy. The mechanisms underlying cellular pluripotency in human cells have been extensively explored in the past decade. In addition to the vast amount of knowledge obtained from the genetic and transcriptional research in hPSCs, there is a rapidly growing interest in the stem cell biology field to examine pluripotency at the protein and PTM level. This review addresses recent progress toward understanding the role of PTMs (glycosylation, phosphorylation, acetylation and methylation) in the regulation of cellular pluripotency.
Collapse
|
338
|
Nazareth EJP, Ostblom JEE, Lücker PB, Shukla S, Alvarez MM, Oh SKW, Yin T, Zandstra PW. High-throughput fingerprinting of human pluripotent stem cell fate responses and lineage bias. Nat Methods 2013; 10:1225-31. [PMID: 24141495 PMCID: PMC5061564 DOI: 10.1038/nmeth.2684] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Accepted: 08/30/2013] [Indexed: 12/22/2022]
Abstract
Populations of cells create local environments that lead to emergent heterogeneity. This is particularly evident in human pluripotent stem cells (hPSCs) where microenvironmental heterogeneity limits cell fate control. We have developed a high-throughput platform to screen hPSCs in configurable micro-environments, enabling the optimization of colony size, cell density, and additional parameters for rapid and robust cell fate responses. Single-cell protein expression profiling revealed that Oct4 and Sox2 co-staining discriminate pluripotent, neuroectoderm, primitive streak, and extraembryonic cell fates, allowing dose responses of 27 developmental factors to simultaneously delineate lineage-specific concentration optima. This platform also enabled quantification of endogenous signaling pathway activation and differentiation bias (fingerprinting). Short-term (48 h) fingerprinting is predictive of definitive endoderm induction efficiency across 12 cell lines and was used a priori to rescue long-term (>18 day) differentiation of a cell line reticent to cardiac induction. These findings facilitate high-throughput hPSC-based screening and quantification of lineage induction bias.
Collapse
Affiliation(s)
- Emanuel J P Nazareth
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | | | | | | |
Collapse
|
339
|
Yusa K. Seamless genome editing in human pluripotent stem cells using custom endonuclease-based gene targeting and the piggyBac transposon. Nat Protoc 2013. [PMID: 24071911 DOI: 10.1038/nprot.2013.126.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
I report here a detailed protocol for seamless genome editing using the piggyBac transposon in human pluripotent stem cells (hPSCs). Recent advances in custom endonucleases have enabled us to routinely perform genome editing in hPSCs. Conventional approaches use the Cre/loxP system that leaves behind residual sequences in the targeted genome. I used the piggyBac transposon to seamlessly remove a drug selection cassette and demonstrated safe genetic correction of a mutation causing α-1 antitrypsin deficiency in patient-derived hPSCs. An alternative approach to using the piggyBac transposon to correct mutations involves using single-stranded oligonucleotides, which is a faster process to complete. However, this experimental procedure is rather complicated and it may be hard to achieve homozygous modifications. In contrast, using the piggyBac transposon with drug selection-based enrichment of genetic modifications, as described here, is simple and can yield multiple correctly targeted clones, including homozygotes. Although two rounds of genetic manipulation are required to achieve homozygote modifications, the entire process takes ∼3 months to complete.
Collapse
Affiliation(s)
- Kosuke Yusa
- Wellcome Trust Sanger Institute, Cambridge, UK.
| |
Collapse
|
340
|
Yusa K. Seamless genome editing in human pluripotent stem cells using custom endonuclease-based gene targeting and the piggyBac transposon. Nat Protoc 2013; 8:2061-78. [PMID: 24071911 DOI: 10.1038/nprot.2013.126] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
I report here a detailed protocol for seamless genome editing using the piggyBac transposon in human pluripotent stem cells (hPSCs). Recent advances in custom endonucleases have enabled us to routinely perform genome editing in hPSCs. Conventional approaches use the Cre/loxP system that leaves behind residual sequences in the targeted genome. I used the piggyBac transposon to seamlessly remove a drug selection cassette and demonstrated safe genetic correction of a mutation causing α-1 antitrypsin deficiency in patient-derived hPSCs. An alternative approach to using the piggyBac transposon to correct mutations involves using single-stranded oligonucleotides, which is a faster process to complete. However, this experimental procedure is rather complicated and it may be hard to achieve homozygous modifications. In contrast, using the piggyBac transposon with drug selection-based enrichment of genetic modifications, as described here, is simple and can yield multiple correctly targeted clones, including homozygotes. Although two rounds of genetic manipulation are required to achieve homozygote modifications, the entire process takes ∼3 months to complete.
Collapse
Affiliation(s)
- Kosuke Yusa
- Wellcome Trust Sanger Institute, Cambridge, UK.
| |
Collapse
|
341
|
de Peppo GM, Marolt D. Make no bones about it: cells could soon be reprogrammed to grow replacement bones? Expert Opin Biol Ther 2013; 14:1-5. [PMID: 24053578 DOI: 10.1517/14712598.2013.840581] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Recent developments in nuclear reprogramming allow the generation of patient-matched stem cells with broad potential for applications in cell therapies, disease modeling and drug discovery. An increasing body of work is reporting the derivation of lineage-specific progenitors from human-induced pluripotent stem cells (hiPSCs), which could in the near future be used to engineer personalized tissue substitutes, including those for reconstructive therapies of bone. Although the potential clinical impact of such technology is not arguable, significant challenges remain to be addressed before hiPSC-derived progenitors can be employed to engineer bone substitutes of clinical relevance. The most important challenge is indeed the construction of personalized multicellular bone substitutes for the treatment of complex skeletal defects that integrate fast, are immune tolerated and display biofunctionality and long-term safety. As recent studies suggest, the merging of iPSC technology with advanced biomaterials and bioreactor technologies offers a way to generate bone substitutes in a controllable, automated manner with potential to meet the needs for scale-up and requirements for translation into clinical practice. It is only via the use of state-of-the-art cell culture technologies, process automation under GMP-compliant conditions, application of appropriate engineering strategies and compliance with regulatory policies that personalized lab-made bone grafts can start being used to treat human patients.
Collapse
Affiliation(s)
- Giuseppe Maria de Peppo
- The New York Stem Cell Foundation Research Institute , 1995 Broadway, NY 10032 , USA +1 212 851 5422 ; +1 212 851 5423 ;
| | | |
Collapse
|
342
|
Razak SRA, Ueno K, Takayama N, Nariai N, Nagasaki M, Saito R, Koso H, Lai CY, Murakami M, Tsuji K, Michiue T, Nakauchi H, Otsu M, Watanabe S. Profiling of microRNA in human and mouse ES and iPS cells reveals overlapping but distinct microRNA expression patterns. PLoS One 2013; 8:e73532. [PMID: 24086284 PMCID: PMC3781120 DOI: 10.1371/journal.pone.0073532] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Accepted: 07/22/2013] [Indexed: 12/19/2022] Open
Abstract
Using quantitative PCR-based miRNA arrays, we comprehensively analyzed the expression profiles of miRNAs in human and mouse embryonic stem (ES), induced pluripotent stem (iPS), and somatic cells. Immature pluripotent cells were purified using SSEA-1 or SSEA-4 and were used for miRNA profiling. Hierarchical clustering and consensus clustering by nonnegative matrix factorization showed two major clusters, human ES/iPS cells and other cell groups, as previously reported. Principal components analysis (PCA) to identify miRNAs that segregate in these two groups identified miR-187, 299-3p, 499-5p, 628-5p, and 888 as new miRNAs that specifically characterize human ES/iPS cells. Detailed direct comparisons of miRNA expression levels in human ES and iPS cells showed that several miRNAs included in the chromosome 19 miRNA cluster were more strongly expressed in iPS cells than in ES cells. Similar analysis was conducted with mouse ES/iPS cells and somatic cells, and several miRNAs that had not been reported to be expressed in mouse ES/iPS cells were suggested to be ES/iPS cell-specific miRNAs by PCA. Comparison of the average expression levels of miRNAs in ES/iPS cells in humans and mice showed quite similar expression patterns of human/mouse miRNAs. However, several mouse- or human-specific miRNAs are ranked as high expressers. Time course tracing of miRNA levels during embryoid body formation revealed drastic and different patterns of changes in their levels. In summary, our miRNA expression profiling encompassing human and mouse ES and iPS cells gave various perspectives in understanding the miRNA core regulatory networks regulating pluripotent cells characteristics.
Collapse
Affiliation(s)
- Siti Razila Abdul Razak
- Division of Molecular and Developmental Biology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
343
|
Alsanie WF, Niclis JC, Petratos S. Human embryonic stem cell-derived oligodendrocytes: protocols and perspectives. Stem Cells Dev 2013; 22:2459-76. [PMID: 23621561 PMCID: PMC3760471 DOI: 10.1089/scd.2012.0520] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Accepted: 04/26/2013] [Indexed: 12/19/2022] Open
Abstract
Oligodendrocytes play a fundamental supportive role in the mammalian central nervous system (CNS) as the myelinating-glial cells. Disruption of fast axonal transport mechanisms can occur as a consequence of mature oligodendrocyte loss following spinal cord injury, stroke, or due to neuroinflammatory conditions, such as multiple sclerosis. As a result of the limited remyelination ability in the CNS after injury or disease, human embryonic stem cells (hESCs) may prove to be a promising option for the generation and replacement of mature oligodendrocytes. Moreover, hESC-derived oligodendrocytes may be experimentally utilized to unravel fundamental questions of oligodendrocyte development, along with their therapeutic potential through growth factor support of axons and neurons. However, an intensive characterization and examination of hESC-derived oligodendrocytes prior to preclinical or clinical trials is required to facilitate greater success in their integration following cellular replacement therapy (CRT). Currently, the protocols utilized to derive oligodendrocytes from hESCs consist of significant variations in culture style, time-length of differentiation, and the provision of growth factors in culture. Further, these differing protocols also report disparate patterns in the expression of oligodendroglial markers by these derived oligodendrocytes, throughout their differentiation in culture. We have comprehensively reviewed the published protocols describing the derivation of oligodendrocytes from hESCs and the studies that examine their efficacy to remyelinate, along with the fundamental issues of their safety as a viable CRT. Additionally, this review will highlight particular issues of concern and suggestions for troubleshooting to provide investigators critical information for the future improvement of establishing in vitro hESC-derived oligodendrocytes.
Collapse
Affiliation(s)
- Walaa F Alsanie
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Australia.
| | | | | |
Collapse
|
344
|
Martinez-Fernandez A, Li X, Hartjes KA, Terzic A, Nelson TJ. Natural cardiogenesis-based template predicts cardiogenic potential of induced pluripotent stem cell lines. ACTA ACUST UNITED AC 2013; 6:462-71. [PMID: 24036272 DOI: 10.1161/circgenetics.113.000045] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Cardiac development is a complex process resulting in an integrated, multilineage tissue with developmental corruption in early embryogenesis leading to congenital heart disease. Interrogation of individual genes has provided the backbone for cardiac developmental biology, yet a comprehensive transcriptome derived from natural cardiogenesis is required to gauge innate developmental milestones. METHODS AND RESULTS Stage-specific cardiac structures were dissected from 8 distinctive mouse embryonic time points to produce genome-wide expressome analysis across cardiogenesis. With reference to this native cardiogenic expression roadmap, divergent induced pluripotent stem cell-derived cardiac expression profiles were mapped from procardiogenic 3-factor (SOX2, OCT4, KLF4) and less-cardiogenic 4-factor (plus c-MYC) reprogrammed cells. Expression of cardiac-related genes from 3-factor-induced pluripotent stem cell differentiated in vitro at days 5 and 11 and recapitulated expression profiles of natural embryos at days E7.5-E8.5 and E14.5-E18.5, respectively. By contrast, 4-factor-induced pluripotent stem cells demonstrated incomplete cardiogenic gene expression profiles beginning at day 5 of differentiation. Differential gene expression within the pluripotent state revealed 23 distinguishing candidate genes among pluripotent cell lines with divergent cardiogenic potentials. A confirmed panel of 12 genes, differentially expressed between high and low cardiogenic lines, was transformed into a predictive score sufficient to discriminate individual induced pluripotent stem cell lines according to relative cardiogenic potential. CONCLUSIONS Transcriptome analysis attuned to natural embryonic cardiogenesis provides a robust platform to probe coordinated cardiac specification and maturation from bioengineered stem cell-based model systems. A panel of developmental-related genes allowed differential prognosis of cardiogenic competency, thus prioritizing cell lines according to natural blueprint to streamline functional applications.
Collapse
Affiliation(s)
- Almudena Martinez-Fernandez
- Division of Cardiovascular Diseases, Department of Medicine, Department of Molecular Pharmacology and Experimental Therapeutics, Division of General Internal Medicine Transplant Center, Division of Biomedical Statistics and Informatics, and Center for Regenerative Medicine, Mayo Clinic, Rochester, MN
| | | | | | | | | |
Collapse
|
345
|
Mfopou JK, Bouwens L. [Differentiation of pluripotent stem cells into pancreatic lineages]. Med Sci (Paris) 2013; 29:736-43. [PMID: 24005628 DOI: 10.1051/medsci/2013298012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Diabetes mellitus is the leading metabolic disease and represents a major public health concern worldwide. Whereas the transplantation of pancreas donor-derived islets significantly improves the quality of life of diabetic patients who become insulin independent for few years, it can unfortunately be provided only to few patients in an advanced stage of the disease. This situation is related to the severe shortage in pancreas donors and has prompted the hunt for alternative sources of islet cells. Beside many other strategies aiming at producing new beta cells in vitro or in vivo, a particular focus has been on the plupiropent stem cells because of their abundant availability and their extreme plasticity. Progress in understanding small vertebrates embryonic development has tremendously contributed to the design of differentiation strategies applied to pluripotent stem cells. Nowadays, definitive endoderm and pancreatic progenitors can be efficiently induced from human embryonic stem cells and from human induced pluripotent stem cells. Although we are still lacking the knowledge required for deriving functional beta cells in vitro, transplantation experiments have demonstrated that stem cell-derived pancreas progenitors further generate this phenotype in vivo. All these findings gathered during the last decade witness the closer clinical application of pluripotent stem cell progenies in diabetes cell therapy.
Collapse
Affiliation(s)
- Josué Kunjom Mfopou
- Unité de différenciation cellulaire, Centre de recherche sur le diabète, Vrije Universiteit Brussel, Bruxelles, Belgique.
| | | |
Collapse
|
346
|
Phanthong P, Raveh-Amit H, Li T, Kitiyanant Y, Dinnyes A. Is aging a barrier to reprogramming? Lessons from induced pluripotent stem cells. Biogerontology 2013; 14:591-602. [PMID: 23963527 DOI: 10.1007/s10522-013-9455-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Accepted: 08/16/2013] [Indexed: 12/17/2022]
Abstract
The discovery of induced pluripotent stem cells (iPSCs) has the potential to revolutionize the field of regenerative medicine. In the past few years, iPSCs have been the subject of intensive research towards their application in disease modeling and drug screening. In the future, these cells may be applied in cell therapy to replace or regenerate tissues by autologous transplantation. However, two major hurdles need to be resolved in order to reach the later goal: the low reprogramming efficiency and the safety risks, such as the integration of foreign DNA into the genome of the cells and the tumor formation potential arising from transplantation of residual undifferentiated cells. Recently, aging emerged as one of the barriers that accounts, at least in part, for the low reprogramming efficiency of bona fide iPSCs. Here, we review the molecular pathways linking aging and reprogramming along with the unanswered questions in the field. We discuss whether reprogramming rejuvenates the molecular and cellular features associated with age, and present the recent advances with iPSC-based models, contributing to our understanding of physiological and premature aging.
Collapse
|
347
|
Duggal G, Heindryckx B, Warrier S, O'Leary T, Van der Jeught M, Lierman S, Vossaert L, Deroo T, Deforce D, Chuva de Sousa Lopes SM, De Sutter P. Influence of activin A supplementation during human embryonic stem cell derivation on germ cell differentiation potential. Stem Cells Dev 2013; 22:3141-55. [PMID: 23829223 DOI: 10.1089/scd.2013.0024] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Human embryonic stem cells (hESCs) are more similar to "primed" mouse epiblast stem cells (mEpiSCs). mEpiSCs, which are derived in Activin A, show an increased propensity to form primordial germ cell (PGC)-like cells in response to bone morphogenic protein 4 (BMP4). Hence, we hypothesized that hESCs derived in the presence of Activin A may be more competent in differentiating towards PGC-like cells after supplementation with BMP4 compared to standard hESC lines. We were able to successfully derive two hESC lines in the presence of Activin A, which were pluripotent and showed higher base levels of STELLA and cKIT compared to standard hESC lines derived without Activin A addition. Furthermore, upon differentiation as embryoid bodies in the presence of BMP4, we observed upregulation of VASA at day 7, both at the transcript and protein level compared to standard hESC lines, which appeared to take longer time for PGC specification. Unlike other hESC lines, nuclear pSMAD2/3 presence confirmed that Activin signalling was switched on in Activin A-derived hESC lines. They were also responsive to BMP4 based on nuclear detection of pSMAD1/5/8 and showed endodermal differentiation as a result of GATA-6 expression. Hence, our results provide novel insights into the impact of hESC derivation in the presence of Activin A and its subsequent influence on germ cell differentiation potential in vitro.
Collapse
Affiliation(s)
- Galbha Duggal
- 1 Department for Reproductive Medicine, Ghent University Hospital , Ghent, Belgium
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
348
|
Wei R, Yang J, Hou W, Liu G, Gao M, Zhang L, Wang H, Mao G, Gao H, Chen G, Hong T. Insulin-producing cells derived from human embryonic stem cells: comparison of definitive endoderm- and nestin-positive progenitor-based differentiation strategies. PLoS One 2013; 8:e72513. [PMID: 23951327 PMCID: PMC3741181 DOI: 10.1371/journal.pone.0072513] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2012] [Accepted: 07/16/2013] [Indexed: 12/26/2022] Open
Abstract
Human embryonic stem cells (hESCs) are pluripotent and capable of undergoing multilineage differentiation into highly specialized cells including pancreatic islet cells. Thus, they represent a novel alternative source for targeted therapies and regenerative medicine for diabetes. Significant progress has been made in differentiating hESCs toward pancreatic lineages. One approach is based on the similarities of pancreatic β cell and neuroepithelial development. Nestin-positive cells are selected as pancreatic β cell precursors and further differentiated to secrete insulin. The other approach is based on our knowledge of developmental biology in which the differentiation protocol sequentially reproduces the individual steps that are known in normal β cell ontogenesis during fetal pancreatic development. In the present study, the hESC cell line PKU1.1 was induced to differentiate into insulin-producing cells (IPCs) using both protocols. The differentiation process was dynamically investigated and the similarities and differences between both strategies were explored. Our results show that IPCs can be successfully induced with both differentiation strategies. The resulting IPCs from both protocols shared many similar features with pancreatic islet cells, but not mature, functional β cells. However, these differently-derived IPC cell types displayed specific morphologies and different expression levels of pancreatic islet development-related markers. These data not only broaden our outlook on hESC differentiation into IPCs, but also extend the full potential of these processes for regenerative medicine in diabetes.
Collapse
Affiliation(s)
- Rui Wei
- Department of Endocrinology and Metabolism, Peking University Third Hospital, Haidian District, Beijing, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
349
|
Asgari S, Moslem M, Bagheri-Lankarani K, Pournasr B, Miryounesi M, Baharvand H. Differentiation and transplantation of human induced pluripotent stem cell-derived hepatocyte-like cells. Stem Cell Rev Rep 2013; 9:493-504. [PMID: 22076752 DOI: 10.1007/s12015-011-9330-y] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The generation of human induced pluripotent stem cells (hiPSCs) with a high differentiation potential provided a new source for hepatocyte generation not only for drug discovery and in vitro disease models, but also for cell replacement therapy. However, the reported hiPSC-derived hepatocyte-like cells (HLCs) were not well characterized and their transplantation, as the most promising clue of cell function was not reported. Here, we performed a growth factor-mediated differentiation of functional HLCs from hiPSCs and evaluated their potential for recovery of a carbon tetrachloride (CCl4)-injured mouse liver following transplantation. The hiPSC-derived hepatic lineage cells expressed hepatocyte-specific markers, showed glycogen and lipid storage activity, secretion of albumin (ALB), alpha-fetoprotein (AFP), urea, and CYP450 metabolic activity in addition to low-density lipoprotein (LDL) and indocyanin green (ICG) uptake. Similar results were observed with human embryonic stem cell (hESC)-derived HLCs. The transplantation of hiPSC-HLCs into a CCl4-injured liver showed incorporation of the hiPSC-HLCs into the mouse liver which resulted in a significant enhancement in total serum ALB after 1 week. A reduction of total serum LDH and bilirubin was seen when compared with the control and sham groups 1 and 5 weeks post-transplantation. Additionally, we detected human serum ALB and ALB-positive transplanted cells in both the host serum and livers, respectively, which showed functional integration of transplanted cells within the mouse livers. Therefore, our results have opened up a proof of concept that functional HLCs can be generated from hiPSCs, thus improving the general condition of a CCl4-injured mouse liver after their transplantation. These results may bring new insights in the clinical applications of hiPSCs once safety issues are overcome.
Collapse
Affiliation(s)
- Samira Asgari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, PO Box 19395-4644, Tehran, Iran
| | | | | | | | | | | |
Collapse
|
350
|
Takahashi K, Yamanaka S. Induced pluripotent stem cells in medicine and biology. Development 2013; 140:2457-61. [PMID: 23715538 DOI: 10.1242/dev.092551] [Citation(s) in RCA: 188] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Differentiated cells can be reprogrammed to pluripotency and other cell fates by treatment with defined factors. The discovery of induced pluripotent stem cells (iPSCs) has opened up unprecedented opportunities in the pharmaceutical industry, in the clinic and in laboratories. In particular, the medical applications of human iPSCs in disease modeling and stem cell therapy have been progressing rapidly. The ability to induce cell fate conversion is attractive not only for these applications, but also for basic research fields, such as development, cancer, epigenetics and aging.
Collapse
Affiliation(s)
- Kazutoshi Takahashi
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan.
| | | |
Collapse
|