1
|
Maqbool S, Baloch MF, Khan MAK, Khalid A, Naimat K. Autologous hematopoietic stem cell transplantation conditioning regimens and chimeric antigen receptor T cell therapy in various diseases. World J Transplant 2024; 14:87532. [PMID: 38576761 PMCID: PMC10989471 DOI: 10.5500/wjt.v14.i1.87532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 10/21/2023] [Accepted: 01/08/2024] [Indexed: 03/15/2024] Open
Abstract
Conditioning regimens employed in autologous stem cell transplantation have been proven useful in various hematological disorders and underlying malig nancies; however, despite being efficacious in various instances, negative consequences have also been recorded. Multiple conditioning regimens were extracted from various literature searches from databases like PubMed, Google scholar, EMBASE, and Cochrane. Conditioning regimens for each disease were compared by using various end points such as overall survival (OS), progression free survival (PFS), and leukemia free survival (LFS). Variables were presented on graphs and analyzed to conclude a more efficacious conditioning regimen. In multiple myeloma, the most effective regimen was high dose melphalan (MEL) given at a dose of 200/mg/m2. The comparative results of acute myeloid leukemia were presented and the regimens that proved to be at an admirable position were busulfan (BU) + MEL regarding OS and BU + VP16 regarding LFS. In case of acute lymphoblastic leukemia (ALL), BU, fludarabine, and etoposide (BuFluVP) conferred good disease control not only with a paramount improvement in survival rate but also low risk of recurrence. However, for ALL, chimeric antigen receptor (CAR) T cell therapy was preferred in the context of better OS and LFS. With respect to Hodgkin's lymphoma, mitoxantrone (MITO)/MEL overtook carmustine, VP16, cytarabine, and MEL in view of PFS and vice versa regarding OS. Non-Hodgkin's lymphoma patients were administered MITO (60 mg/m2) and MEL (180 mg/m2) which showed promising results. Lastly, amyloidosis was considered, and the regimen that proved to be competent was MEL 200 (200 mg/m2). This review article demonstrates a comparison between various conditioning regimens employed in different diseases.
Collapse
Affiliation(s)
- Shahzaib Maqbool
- Department of Medicine, Holy Family Hospital, Rawalpindi 46000, Pakistan
| | - Maryam Farhan Baloch
- Department of Community Medicine, Allama Iqbal Medical College, Lahore 45000, Pakistan
| | | | - Azeem Khalid
- Department of Medicine, Allama lqbal Medical College, Lahore 45000, Pakistan
| | - Kiran Naimat
- Department of MedicineLiaquat University of Medical and Health Sciences, Karachi 43000, Pakistan
| |
Collapse
|
2
|
Abstract
The derivation of induced pluripotent stem cells (iPSCs) over a decade ago sparked widespread enthusiasm for the development of new models of human disease, enhanced platforms for drug discovery and more widespread use of autologous cell-based therapy. Early studies using directed differentiation of iPSCs frequently uncovered cell-level phenotypes in monogenic diseases, but translation to tissue-level and organ-level diseases has required development of more complex, 3D, multicellular systems. Organoids and human-rodent chimaeras more accurately mirror the diverse cellular ecosystems of complex tissues and are being applied to iPSC disease models to recapitulate the pathobiology of a broad spectrum of human maladies, including infectious diseases, genetic disorders and cancer.
Collapse
|
3
|
Global MicroRNA Profiling Uncovers miR-206 as a Negative Regulator of Hematopoietic Commitment in Human Pluripotent Stem Cells. Int J Mol Sci 2019; 20:ijms20071737. [PMID: 30965622 PMCID: PMC6479521 DOI: 10.3390/ijms20071737] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 04/02/2019] [Accepted: 04/06/2019] [Indexed: 02/08/2023] Open
Abstract
Although human pluripotent stem cells (hPSCs) can theoretically differentiate into any cell type, their ability to produce hematopoietic cells is highly variable from one cell line to another. The underlying mechanisms of this heterogeneity are not clearly understood. Here, using a whole miRNome analysis approach in hPSCs, we discovered that their hematopoietic competency was associated with the expression of several miRNAs and conversely correlated to that of miR-206 specifically. Lentiviral-based miR-206 ectopic expression in H1 hematopoietic competent embryonic stem (ES) cells markedly impaired their differentiation toward the blood lineage. Integrative bioinformatics identified a potential miR-206 target gene network which included hematopoietic master regulators RUNX1 and TAL1. This work sheds light on the critical role of miR-206 in the generation of blood cells off hPSCs. Our results pave the way for future genetic manipulation of hPSCs aimed at increasing their blood regenerative potential and designing better protocols for the generation of bona fide hPSC-derived hematopoietic stem cells.
Collapse
|
4
|
β-catenin coordinates with Jup and the TCF1/GATA6 axis to regulate human embryonic stem cell fate. Dev Biol 2017; 431:272-281. [PMID: 28943339 DOI: 10.1016/j.ydbio.2017.09.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 09/02/2017] [Accepted: 09/04/2017] [Indexed: 12/22/2022]
Abstract
β-catenin-mediated signaling has been extensively studied in regard to its role in the regulation of human embryonic stem cells (hESCs). However, the results are controversial and the mechanism by which β-catenin regulates the hESC fate remains unclear. Here, we report that β-catenin and γ-catenin are functionally redundant in mediating hESC adhesion and are required for embryoid body formation, but both genes are dispensable for hESC maintenance, as the undifferentiated state of β-catenin and γ-catenin double deficient hESCs can be maintained. Overexpression of β-catenin induces rapid hESC differentiation. Functional assays revealed that TCF1 plays a crucial role in hESC differentiation mediated by β-catenin. Forced expression of TCF1, but not other LEF1/TCF family members, resulted in hESC differentiation towards the definitive endoderm. Conversely, knockdown of TCF1 or inhibition of the interaction between TCF1 and β-catenin delayed hESC exit from pluripotency. Furthermore, we demonstrated that GATA6 plays a predominant role in TCF1-mediated hESC differentiation. Knockdown of GATA6 completely eliminated the effect of TCF1, while forced expression of GATA6 induced hESC differentiation. Our data thus reveal more detailed mechanisms for β-catenin in regulating hESC fate decisions and will expand our understanding of the self-renewal and differentiation circuitry in hESCs.
Collapse
|
5
|
Gupta P, Hourigan K, Jadhav S, Bellare J, Verma P. Effect of lactate and pH on mouse pluripotent stem cells: Importance of media analysis. Biochem Eng J 2017. [DOI: 10.1016/j.bej.2016.11.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
6
|
Lee JB, Graham M, Collins TJ, Lee JH, Hong SH, Mcnicol AJ, Shapovalova Z, Bhatia M. Reversible lineage-specific priming of human embryonic stem cells can be exploited to optimize the yield of differentiated cells. Stem Cells 2016; 33:1142-52. [PMID: 25639500 PMCID: PMC4413029 DOI: 10.1002/stem.1952] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Revised: 11/09/2014] [Accepted: 11/21/2014] [Indexed: 12/17/2022]
Abstract
The clinical use of human embryonic stem cells (hESCs) requires efficient cellular expansion that must be paired with an ability to generate specialized progeny through differentiation. Self-renewal and differentiation are deemed inherent hallmarks of hESCs and a growing body of evidence suggests that initial culture conditions dictate these two aspects of hESC behavior. Here, we reveal that defined culture conditions using commercial mTeSR1 media augment the expansion of hESCs and enhance their capacity for neural differentiation at the expense of hematopoietic lineage competency without affecting pluripotency. This culture-induced modification was shown to be reversible, as culture in mouse embryonic fibroblast-conditioned media (MEF-CM) in subsequent passages allowed mTeSR1-expanded hESCs to re-establish hematopoietic differentiation potential. Optimal yield of hematopoietic cells can be achieved by expansion in mTeSR1 followed by a recovery period in MEF-CM. Furthermore, the lineage propensity to hematopoietic and neural cell types could be predicted via analysis of surrogate markers expressed by hESCs cultured in mTeSR1 versus MEF-CM, thereby circumventing laborious in vitro differentiation assays. Our study reveals that hESCs exist in a range of functional states and balance expansion with differentiation potential, which can be modulated by culture conditions in a predictive and quantitative manner.
Collapse
Affiliation(s)
- Jung Bok Lee
- McMaster Stem Cell and Cancer Research Institute, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, Ontario, Canada
| | | | | | | | | | | | | | | |
Collapse
|
7
|
Park B, Yoo KH, Kim C. Hematopoietic stem cell expansion and generation: the ways to make a breakthrough. Blood Res 2015; 50:194-203. [PMID: 26770947 PMCID: PMC4705045 DOI: 10.5045/br.2015.50.4.194] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 12/11/2015] [Accepted: 12/16/2015] [Indexed: 12/28/2022] Open
Abstract
Hematopoietic stem cell transplantation (HSCT) is the first field where human stem cell therapy was successful. Flooding interest on human stem cell therapy to cure previously incurable diseases is largely indebted to HSCT success. Allogeneic HSCT has been an important modality to cure various diseases including hematologic malignancies, various non-malignant hematologic diseases, primary immunodeficiency diseases, and inborn errors of metabolism, while autologous HSCT is generally performed to rescue bone marrow aplasia following high-dose chemotherapy for solid tumors or multiple myeloma. Recently, HSCs are also spotlighted in the field of regenerative medicine for the amelioration of symptoms caused by neurodegenerative diseases, heart diseases, and others. Although the demand for HSCs has been growing, their supply often fails to meet the demand of the patients needing transplant due to a lack of histocompatible donors or a limited cell number. This review focuses on the generation and large-scale expansion of HSCs, which might overcome current limitations in the application of HSCs for clinical use. Furthermore, current proof of concept to replenish hematological homeostasis from non-hematological origin will be covered.
Collapse
Affiliation(s)
- Bokyung Park
- Department of Bioscience and Biotechnology, Sejong University, Korea
| | - Keon Hee Yoo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Korea.; Department of Medical Device Management and Research, SAIHST, Sungkyunkwan University, Seoul, Korea
| | - Changsung Kim
- Department of Bioscience and Biotechnology, Sejong University, Korea
| |
Collapse
|
8
|
Production of human pluripotent stem cell therapeutics under defined xeno-free conditions: progress and challenges. Stem Cell Rev Rep 2015; 11:96-109. [PMID: 25077810 DOI: 10.1007/s12015-014-9544-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Recent advances on human pluripotent stem cells (hPSCs), including human embryonic stem cells (hESCs) and induced pluripotent stem cells (hiPSCs) have brought us closer to the realization of their clinical potential. Nonetheless, tissue engineering and regenerative medicine applications will require the generation of hPSC products well beyond the laboratory scale. This also mandates the production of hPSC therapeutics in fully-defined, xeno-free systems and in a reproducible manner. Toward this goal, we summarize current developments in defined media free of animal-derived components for hPSC culture. Bioinspired and synthetic extracellular matrices for the attachment, growth and differentiation of hPSCs are also reviewed. Given that most progress in xeno-free medium and substrate development has been demonstrated in two-dimensional rather than three dimensional culture systems, translation from the former to the latter poses unique difficulties. These challenges are discussed in the context of cultivation platforms of hPSCs as aggregates, on microcarriers or after encapsulation in biocompatible scaffolds.
Collapse
|
9
|
Zandi M, Muzaffar M, Shah SM, Kumar Singh M, Palta P, Kumar Singla S, Manik R, Chauhan MS. Optimization of Buffalo (Bubalus bubalis) Embryonic Stem Cell Culture System. CELL JOURNAL 2015. [PMID: 26199905 PMCID: PMC4503840 DOI: 10.22074/cellj.2016.3728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
OBJECTIVE In order to retain an undifferentiated pluripotent state, embryonic stem (ES) cells have to be cultured on feeder cell layers. However, use of feeder layers limits stem cell research, since experimental data may result from a combined ES cell and feeder cell response to various stimuli. MATERIALS AND METHODS In this experimental study, a buffalo ES cell line was established from in vitro derived blastocysts and characterized by the Alkaline phosphatase (AP) and immunoflourescence staining of various pluripotency markers. We examined the effect of various factors like fibroblast growth factor 2 (FGF-2), leukemia inhibitory factor (LIF) and Y-27632 to support the growth and maintenance of bubaline ES cells on gelatin coated dishes, in order to establish feeder free culture systems. We also analyzed the effect of feeder-conditioned media on stem cell growth in gelatin based cultures both in the presence as well as in the absence of the growth factors. RESULTS The results showed that Y-27632, in the presence of FGF-2 and LIF, resulted in higher colony growth and increased expression of Nanog gene. Feeder-Conditioned Medium resulted in a significant increase in growth of buffalo ES cells on gelatin coated plates, however, feeder layer based cultures produced better results than gelatin based cultures. Feeder layers from buffalo fetal fibroblast cells can support buffalo ES cells for more than two years. CONCLUSION We developed a feeder free culture system that can maintain buffalo ES cells in the short term, as well as feeder layer based culture that can support the long term maintenance of buffalo ES cells.
Collapse
Affiliation(s)
- Mohammad Zandi
- Department of Animal and Poultry Science and Fisheries, Agricultural Institute, Iranian Research Organisation for Science and Technology (IROST), Tehran, Iran
| | - Musharifa Muzaffar
- Embryo Biotechnology Laboratory, Animal Biotechnology Centre, National Dairy Research Institute (NDRI), Karnal, India
| | - Syed Mohmad Shah
- Embryo Biotechnology Laboratory, Animal Biotechnology Centre, National Dairy Research Institute (NDRI), Karnal, India
| | - Manoj Kumar Singh
- Embryo Biotechnology Laboratory, Animal Biotechnology Centre, National Dairy Research Institute (NDRI), Karnal, India
| | - Prabhat Palta
- Embryo Biotechnology Laboratory, Animal Biotechnology Centre, National Dairy Research Institute (NDRI), Karnal, India
| | - Suresh Kumar Singla
- Embryo Biotechnology Laboratory, Animal Biotechnology Centre, National Dairy Research Institute (NDRI), Karnal, India
| | - Radheysham Manik
- Embryo Biotechnology Laboratory, Animal Biotechnology Centre, National Dairy Research Institute (NDRI), Karnal, India
| | - Manmohan Singh Chauhan
- Embryo Biotechnology Laboratory, Animal Biotechnology Centre, National Dairy Research Institute (NDRI), Karnal, India
| |
Collapse
|
10
|
Conway MK, Gerger MJ, Balay EE, O'Connell R, Hanson S, Daily NJ, Wakatsuki T. Scalable 96-well Plate Based iPSC Culture and Production Using a Robotic Liquid Handling System. J Vis Exp 2015:e52755. [PMID: 26068617 DOI: 10.3791/52755] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Continued advancement in pluripotent stem cell culture is closing the gap between bench and bedside for using these cells in regenerative medicine, drug discovery and safety testing. In order to produce stem cell derived biopharmaceutics and cells for tissue engineering and transplantation, a cost-effective cell-manufacturing technology is essential. Maintenance of pluripotency and stable performance of cells in downstream applications (e.g., cell differentiation) over time is paramount to large scale cell production. Yet that can be difficult to achieve especially if cells are cultured manually where the operator can introduce significant variability as well as be prohibitively expensive to scale-up. To enable high-throughput, large-scale stem cell production and remove operator influence novel stem cell culture protocols using a bench-top multi-channel liquid handling robot were developed that require minimal technician involvement or experience. With these protocols human induced pluripotent stem cells (iPSCs) were cultured in feeder-free conditions directly from a frozen stock and maintained in 96-well plates. Depending on cell line and desired scale-up rate, the operator can easily determine when to passage based on a series of images showing the optimal colony densities for splitting. Then the necessary reagents are prepared to perform a colony split to new plates without a centrifugation step. After 20 passages (~3 months), two iPSC lines maintained stable karyotypes, expressed stem cell markers, and differentiated into cardiomyocytes with high efficiency. The system can perform subsequent high-throughput screening of new differentiation protocols or genetic manipulation designed for 96-well plates. This technology will reduce the labor and technical burden to produce large numbers of identical stem cells for a myriad of applications.
Collapse
|
11
|
Abstract
The onset of hematopoiesis in mammals is defined by generation of primitive erythrocytes and macrophage progenitors in embryonic yolk sac. Laboratories have met the challenge of transient and swiftly changing specification events from ventral mesoderm through multipotent progenitors and maturing lineage-restricted hematopoietic subtypes, by developing powerful in vitro experimental models to interrogate hematopoietic ontogeny. Most importantly, studies of differentiating embryonic stem cell derivatives in embryoid body and stromal coculture systems have identified crucial roles for transcription factor networks (e.g. Gata1, Runx1, Scl) and signaling pathways (e.g. BMP, VEGF, WNT) in controlling stem and progenitor cell output. These and other relevant pathways have pleiotropic biological effects, and are often associated with early embryonic lethality in knockout mice. Further refinement in subsequent studies has allowed conditional expression of key regulatory genes, and isolation of progenitors via cell surface markers (e.g. FLK1) and reporter-tagged constructs, with the purpose of measuring their primitive and definitive hematopoietic potential. These observations continue to inform attempts to direct the differentiation, and augment the expansion, of progenitors in human cell culture systems that may prove useful in cell replacement therapies for hematopoietic deficiencies. The purpose of this review is to survey the extant literature on the use of differentiating murine embryonic stem cells in culture to model the developmental process of yolk sac hematopoiesis.
Collapse
|
12
|
Del Pino A, Ligero G, López MB, Navarro H, Carrillo JA, Pantoll SC, Díaz de la Guardia R. Morphology, cell viability, karyotype, expression of surface markers and plasticity of three human primary cell line cultures before and after the cryostorage in LN2 and GN2. Cryobiology 2014; 70:1-8. [PMID: 25445570 DOI: 10.1016/j.cryobiol.2014.10.011] [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: 04/25/2014] [Revised: 09/23/2014] [Accepted: 10/29/2014] [Indexed: 01/09/2023]
Abstract
Primary cell line cultures from human skin biopsies, adipose tissue and tumor tissue are valuable samples for research and therapy. In this regard, their derivation, culture, storage, transport and thawing are important steps to be studied. Towards this end, we wanted to establish the derivation, and identify the culture characteristics and the loss of viability of three human primary cell line cultures (human adult dermal fibroblasts (hADFs), human adult mesenchymal stem cells (hMSCs), and primary culture of tumor cells from lung adenocarcinoma (PCTCLA)). Compared to fresh hADFs, hMSCs and PCTCLA, thawed cells stored in a cryogenic Dewar tanks with liquid nitrogen (LN2), displayed 98.20% ± 0.99, 95.40% ± 1.41 and 93.31% ± 3.83 of cell viability, respectively. Thawed cells stored in a Dry Vapor Shipper container with gas phase (GN2), for 20 days, in addition displayed 4.61% ± 2.78, 3.70% ± 4.09 and 9.13% ± 3.51 of average loss of cells viability, respectively, showing strong correlation between the loss of viability in hADFs and the number of post-freezing days in the Dry Vapor Shipper. No significant changes in morphological characteristics or in the expression of surface markers (being hADFs, hMSCs and PCTCLA characterized by positive markers CD73+; CD90+; CD105+; and negative markers CD14-; CD20-; CD34-; and CD45-; n=2) were found. Chromosome abnormalities in the karyotype were not found. In addition, under the right conditions hMSCs were differentiated into adipogenic, osteogenic and chondrogenic lineages in vitro. In this paper, we have shown the characteristics of three human primary cell line cultures when they are stored in LN2 and GN2.
Collapse
Affiliation(s)
- Alberto Del Pino
- Biobanco del Sistema Sanitario Público de Andalucía (BBSSPA), Centro de Investigaciones Biomédicas, Consejería de Salud - Universidad de Granada, Granada, Spain
| | - Gertrudis Ligero
- Biobanco del Sistema Sanitario Público de Andalucía (BBSSPA), Centro de Investigaciones Biomédicas, Consejería de Salud - Universidad de Granada, Granada, Spain
| | - María B López
- Department of Physiology, Institute of Nutrition and Food Technology, University of Granada, Granada, Spain
| | - Héctor Navarro
- Biobanco del Sistema Sanitario Público de Andalucía (BBSSPA), Centro de Investigaciones Biomédicas, Consejería de Salud - Universidad de Granada, Granada, Spain
| | - Jose A Carrillo
- Biobanco del Sistema Sanitario Público de Andalucía (BBSSPA), Centro de Investigaciones Biomédicas, Consejería de Salud - Universidad de Granada, Granada, Spain
| | - Siobhan C Pantoll
- Biobanco del Sistema Sanitario Público de Andalucía (BBSSPA), Centro de Investigaciones Biomédicas, Consejería de Salud - Universidad de Granada, Granada, Spain
| | - Rafael Díaz de la Guardia
- Biobanco del Sistema Sanitario Público de Andalucía (BBSSPA), Centro de Investigaciones Biomédicas, Consejería de Salud - Universidad de Granada, Granada, Spain.
| |
Collapse
|
13
|
Kim C. Disease modeling and cell based therapy with iPSC: future therapeutic option with fast and safe application. Blood Res 2014; 49:7-14. [PMID: 24724061 PMCID: PMC3974965 DOI: 10.5045/br.2014.49.1.7] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 03/18/2014] [Accepted: 03/19/2014] [Indexed: 12/20/2022] Open
Abstract
Induced pluripotent stem cell (iPSC) technology has shown us great hope to treat various human diseases which have been known as untreatable and further endows personalized medicine for future therapy without ethical issues and immunological rejection which embryonic stem cell (hES) treatment has faced. It has been agreed that iPSCs knowledge can be harnessed from disease modeling which mimics human pathological development rather than trials utilizing conventional rodent and cell lines. Now, we can routinely generate iPSC from patient specific cell sources, such as skin fibroblast, hair follicle cells, patient blood samples and even urine containing small amount of epithelial cells. iPSC has both similarity and dissimilarity to hES. iPSC is similar enough to regenerate tissue and even full organism as ES does, however what we want for therapeutic advantage is limited to regenerated tissue and lineage specific differentiation. Depending on the lineage and type of cells, both tissue memory containing (DNA rearrangement/epigenetics) and non-containing iPSC can be generated. This makes iPSC even better choice to perform disease modeling as well as cell based therapy. Tissue memory containing iPSC from mature leukocytes would be beneficial for curing cancer and infectious disease. In this review, the benefit of iPSC for translational approaches will be presented.
Collapse
Affiliation(s)
- Changsung Kim
- Department of Bioscience and Biotechnology, Sejong University, Seoul, Korea
| |
Collapse
|
14
|
Peterson H, Abu Dawud R, Garg A, Wang Y, Vilo J, Xenarios I, Adjaye J. Qualitative modeling identifies IL-11 as a novel regulator in maintaining self-renewal in human pluripotent stem cells. Front Physiol 2013; 4:303. [PMID: 24194720 PMCID: PMC3809568 DOI: 10.3389/fphys.2013.00303] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Accepted: 10/02/2013] [Indexed: 11/13/2022] Open
Abstract
Pluripotency in human embryonic stem cells (hESCs) and induced pluripotent stem cells (iPSCs) is regulated by three transcription factors—OCT3/4, SOX2, and NANOG. To fully exploit the therapeutic potential of these cells it is essential to have a good mechanistic understanding of the maintenance of self-renewal and pluripotency. In this study, we demonstrate a powerful systems biology approach in which we first expand literature-based network encompassing the core regulators of pluripotency by assessing the behavior of genes targeted by perturbation experiments. We focused our attention on highly regulated genes encoding cell surface and secreted proteins as these can be more easily manipulated by the use of inhibitors or recombinant proteins. Qualitative modeling based on combining boolean networks and in silico perturbation experiments were employed to identify novel pluripotency-regulating genes. We validated Interleukin-11 (IL-11) and demonstrate that this cytokine is a novel pluripotency-associated factor capable of supporting self-renewal in the absence of exogenously added bFGF in culture. To date, the various protocols for hESCs maintenance require supplementation with bFGF to activate the Activin/Nodal branch of the TGFβ signaling pathway. Additional evidence supporting our findings is that IL-11 belongs to the same protein family as LIF, which is known to be necessary for maintaining pluripotency in mouse but not in human ESCs. These cytokines operate through the same gp130 receptor which interacts with Janus kinases. Our finding might explain why mESCs are in a more naïve cell state compared to hESCs and how to convert primed hESCs back to the naïve state. Taken together, our integrative modeling approach has identified novel genes as putative candidates to be incorporated into the expansion of the current gene regulatory network responsible for inducing and maintaining pluripotency.
Collapse
Affiliation(s)
- Hedi Peterson
- Quretec Ltd. Tartu, Estonia ; Faculty of Mathematics and Computer Science, Institute of Computer Science, University of Tartu Tartu, Estonia ; Faculty of Science and Technology, Institute of Molecular and Cellular Biology, University of Tartu Tartu, Estonia
| | | | | | | | | | | | | |
Collapse
|
15
|
Kook SH, Jeon YM, Park SS, Lee JC. Periodontal fibroblasts modulate proliferation and osteogenic differentiation of embryonic stem cells through production of fibroblast growth factors. J Periodontol 2013; 85:645-54. [PMID: 23805819 DOI: 10.1902/jop.2013.130252] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
BACKGROUND Periodontal ligament fibroblasts (PLFs) maintain homeostasis of periodontal ligaments by producing paracrine factors that affect various functions of stem-like cells. It is hypothesized that PLFs induce proliferation and differentiation of stem cells more effectively than gingival fibroblasts (GFs) and skin fibroblasts (SFs). METHODS PLFs and GFs were isolated from extracted teeth and cultured in the presence and absence of osteogenesis-inducing factors. Mouse embryonic stem (mES) cells and SFs were purchased commercially. mES cells were incubated with culture supernatants of these fibroblasts or cocultured directly with the cells. Proliferation and mineralization in mES cells were determined at various times of incubation. Immunostaining and polymerase chain reaction were performed. The activity of mitogen-activated protein kinase and alkaline phosphatase (ALP) was also measured. RESULTS In cocultures, PLFs stimulated proliferation of mES cells more effectively than GFs or SFs. Similarly, the addition of culture supernatant of PLFs induced the most prominent proliferation of mES cells, and this was significantly inhibited by treatment with antibody against fibroblast growth factor (FGF)4 or the c-Jun N-terminal kinase inhibitor SP600125 (anthra[1,9-cd]pyrazol-6(2H)-one). Supplementation with culture supernatant from the fibroblasts induced osteogenic differentiation of mES cells in the order PLFs > GFs > SFs. These activities of PLFs were related to their potential to produce osteogenic markers, such as ALP and runt-related transcription factor-2 (Runx2), and to secrete FGF7. Pretreatment of mES cells with the extracellular signal-regulated kinase inhibitor PD98059 [2-(2-amino-3-methyoxyphenyl)-4H-1-benzopyran-4-one] or SP600125 clearly attenuated mineralization induced by culture supernatant of PLF with attendant decreases in mRNA levels of Runx2, bone sialoprotein, osteocalcin, and osteopontin. CONCLUSION PLFs regulate the proliferation and osteogenic differentiation of mES cells more strongly than GFs and SFs via the secretion of FGF through a mechanism that involves mitogen-activated protein kinase-mediated signaling.
Collapse
Affiliation(s)
- Sung-Ho Kook
- Institute of Oral Biosciences and School of Dentistry, Chonbuk National University, Jeonju, South Korea
| | | | | | | |
Collapse
|
16
|
Liberski AR, Al-Noubi MN, Rahman ZH, Halabi NM, Dib SS, Al-Mismar R, Billing AM, Krishnankutty R, Ahmad FS, Raynaud CM, Rafii A, Engholm-Keller K, Graumann J. Adaptation of a commonly used, chemically defined medium for human embryonic stem cells to stable isotope labeling with amino acids in cell culture. J Proteome Res 2013; 12:3233-45. [PMID: 23734825 DOI: 10.1021/pr400099j] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Metabolic labeling with stable isotopes is a prominent technique for comparative quantitative proteomics, and stable isotope labeling with amino acids in cell culture (SILAC) is the most commonly used approach. SILAC is, however, traditionally limited to simple tissue culture regimens and only rarely employed in the context of complex culturing conditions as those required for human embryonic stem cells (hESCs). Classic hESC culture is based on the use of mouse embryonic fibroblasts (MEFs) as a feeder layer, and as a result, possible xenogeneic contamination, contribution of unlabeled amino acids by the feeders, interlaboratory variability of MEF preparation, and the overall complexity of the culture system are all of concern in conjunction with SILAC. We demonstrate a feeder-free SILAC culture system based on a customized version of a commonly used, chemically defined hESC medium developed by Ludwig et al. and commercially available as mTeSR1 [mTeSR1 is a trade mark of WiCell (Madison, WI) licensed to STEMCELL Technologies (Vancouver, Canada)]. This medium, together with adjustments to the culturing protocol, facilitates reproducible labeling that is easily scalable to the protein amounts required by proteomic work flows. It greatly enhances the usability of quantitative proteomics as a tool for the study of mechanisms underlying hESCs differentiation and self-renewal. Associated data have been deposited to the ProteomeXchange with the identifier PXD000151.
Collapse
|
17
|
Kingham E, Oreffo ROC. Embryonic and induced pluripotent stem cells: understanding, creating, and exploiting the nano-niche for regenerative medicine. ACS NANO 2013; 7:1867-81. [PMID: 23414366 PMCID: PMC3610401 DOI: 10.1021/nn3037094] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Accepted: 01/25/2013] [Indexed: 05/26/2023]
Abstract
Embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) have the capacity to differentiate into any specialized cell type of the human body, and therefore, ESC/iPSC-derived cell types offer great potential for regenerative medicine. However, key to realizing this potential requires a strong understanding of stem cell biology, techniques to maintain stem cells, and strategies to manipulate cells to efficiently direct cell differentiation toward a desired cell type. As nanoscale science and engineering continues to produce novel nanotechnology platforms, which inform, infiltrate, and impinge on many aspects of everyday life, it is no surprise that stem cell research is turning toward developments in nanotechnology to answer research questions and to overcome obstacles in regenerative medicine. Here we discuss recent advances in ESC and iPSC manipulation using nanomaterials and highlight future challenges within this area of research.
Collapse
Affiliation(s)
- Emmajayne Kingham
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Human Development and Health, Institute of Developmental Sciences, University of Southampton, Southampton SO16 6YD, United Kingdom.
| | | |
Collapse
|
18
|
He JQ, January CT, Thomson JA, Kamp TJ. Human embryonic stem cell-derived cardiomyocytes: drug discovery and safety pharmacology. Expert Opin Drug Discov 2013; 2:739-53. [PMID: 23488962 DOI: 10.1517/17460441.2.5.739] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Human embryonic stem cells (hESCs) can provide potentially unlimited quantities of a wide range of human cell types that can be used in drug discovery and development, basic research and regenerative medicine. In this review, the authors describe the differentiation of hESCs into cardiomyocytes and outline the properties of hESC-derived cardiomyocytes (hESC-CMs), including their cardiac-type action potentials and contractile characteristics. In vitro cellular assays using hESC-CMs, which can be genetically engineered to create target-specific reporters as well as human disease models, will have applications at multiple stages of the drug discovery process. Furthermore, cardiac safety pharmacology assays evaluating the risk of proarrhythmic side effects associated with QT prolongation may be enhanced in their predictive value with the use of hESC-CMs.
Collapse
Affiliation(s)
- Jia-Qiang He
- Cellular Dynamics International, Inc., 525 Science Drive, Suite 200, Madison, WI 53711, USA +1 608 263 4856 ; +1 608 263 0405 ;
| | | | | | | |
Collapse
|
19
|
Mesenchymal stem cells as an appropriate feeder layer for prolonged in vitro culture of human induced pluripotent stem cells. Mol Biol Rep 2013; 40:3023-31. [DOI: 10.1007/s11033-012-2376-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2012] [Accepted: 12/17/2012] [Indexed: 12/24/2022]
|
20
|
Abstract
A significant barrier to the therapeutic application of pluripotent stem cells (PSCs) is the risk associated with the presence of undefined, animal-derived elements that are routinely used to culture these cells. Originally, PSCs were derived on mouse feeder cells in media containing fetal calf serum. Such conditions could expose potential patients to animal pathogens or lead to immune rejection. Substantial efforts have been made to remove these components and successfully maintain these cells in a completely defined, xeno-free environment. In this chapter, we examine substrates consisting of animal-derived proteins, purified human proteins, recombinant human proteins, and synthetic polymers and their ability to maintain the undifferentiated growth of various pluripotent stem cell lines in a variety of supplemented media.
Collapse
Affiliation(s)
- Alexandria Sams
- Primary and Stem Cell Systems, Life Technologies, Frederick, MD, USA
| | | |
Collapse
|
21
|
Li L, Bennett SAL, Wang L. Role of E-cadherin and other cell adhesion molecules in survival and differentiation of human pluripotent stem cells. Cell Adh Migr 2012; 6:59-70. [PMID: 22647941 PMCID: PMC3364139 DOI: 10.4161/cam.19583] [Citation(s) in RCA: 145] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The survival, proliferation, self-renewal and differentiation of human pluripotent stem cells (hPSCs, including human embryonic stem cells and human induced pluripotent stem cells) involve a number of processes that require cell-cell and cell-matrix interactions. The cell adhesion molecules (CAMs), a group of cell surface proteins play a pivotal role in mediating such interactions. Recent studies have provided insights into the essential roles and mechanisms of CAMs in the regulation of hPSC fate decisions. Here, we review the latest research progress in this field and focus on how E-cadherin and several other important CAMs including classic cadherins, Ig-superfamily CAMs, integrins and heparin sulfate proteoglycans control survival and differentiation of hPSCs.
Collapse
Affiliation(s)
- Li Li
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON Canada
| | | | | |
Collapse
|
22
|
Frank S, Zhang M, Schöler HR, Greber B. Small molecule-assisted, line-independent maintenance of human pluripotent stem cells in defined conditions. PLoS One 2012; 7:e41958. [PMID: 22860038 PMCID: PMC3408405 DOI: 10.1371/journal.pone.0041958] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Accepted: 06/28/2012] [Indexed: 02/04/2023] Open
Abstract
Human pluripotent stem cells (hPSCs) are conventionally grown in a mouse feeder cell-dependent manner. Chemically defined culture conditions are, however, desirable not only for potential medically oriented applications but also for investigating mechanisms of self-renewal and differentiation. In light of the rather high complexity and cost of existing defined hPSC culture systems, we have systematically evaluated over 20 potential media ingredients. Only components that reproducibly gave beneficial effects were ultimately combined to yield a simple and cost-effective formulation termed FTDA. This xeno-free medium is based on mimicking self-renewal factor activities present in mouse embryonic fibroblast-conditioned medium, at minimal dosages. Additionally, small molecule inhibitors of BMP and WNT signaling served to specifically suppress typical types of spontaneous differentiation seen in hPSC cultures. FTDA medium was suitable for the generation of human induced pluripotent stem cells and enabled robust long-term maintenance of diverse hPSC lines including hard-to-grow ones. Comparisons with existing defined media suggested reduced spontaneous differentiation rates in FTDA. Our results imply that using supportive factors at minimal concentrations may still promote robust self-renewal and preserve pluripotency of hPSCs.
Collapse
Affiliation(s)
- Stefan Frank
- Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | | | | | | |
Collapse
|
23
|
Ebihara Y, Ma F, Tsuji K. Generation of red blood cells from human embryonic/induced pluripotent stem cells for blood transfusion. Int J Hematol 2012; 95:610-6. [PMID: 22648827 DOI: 10.1007/s12185-012-1107-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2012] [Revised: 05/14/2012] [Accepted: 05/14/2012] [Indexed: 12/19/2022]
Abstract
Red blood cell (RBC) transfusion is necessary for many patients with emergency or hematological disorders. However, to date the supply of RBCs remains labile and dependent on voluntary donations. In addition, the transmission of infectious disease via blood transfusion from unspecified donors remains a risk. Establishing a large quantity of safe RBCs would help to address this issue. Human embryonic stem (hES) cells and the recently established human induced pluripotent stem (hiPS) cells represent potentially unlimited sources of donor-free RBCs for blood transfusion, as they can proliferate indefinitely in vitro. Extensive research has been done to efficiently generate transfusable RBCs from hES/iPS cells. Nevertheless, a number of challenges must be overcome before the clinical usage of hES/iPS cell-derived RBCs can become a reality.
Collapse
Affiliation(s)
- Yasuhiro Ebihara
- Division of Stem Cell Processing, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | | | | |
Collapse
|
24
|
Kumar De A, Malakar D, Akshey YS, Jena MK, Dutta R. Isolation and characterization of embryonic stem cell-like cells from in vitro produced goat (Capra hircus) embryos. Anim Biotechnol 2012; 22:181-96. [PMID: 22132812 DOI: 10.1080/10495398.2011.622189] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The aim of the present study was to isolate and characterize goat embryonic stem cell-like cells from in vitro produced goat embryos. Inner cell mass (ICM) cells were isolated either mechanically or by enzymatic digestion from 150 blastocysts and 35 hatched blastocysts whereas 100 morulae were used for blastomeres isolation mechanically. The ICM derived cells or blastomeres were cultured on a feeder layer. The primary colony formation was significantly higher (P < 0.01) for hatched blastocysts (77.14%) than early/expanded blastocysts (54%) or morula (14%). When ICMs were isolated mechanically the primary colony formation for hatched blastocysts (90%) as well as blastocysts (66%) were significantly more than when ICMs were isolated by enzymatic digestion (60% and 30%, respectively). The colonies were disaggregated either mechanically or by enzymatic digestion for further subculture. When mechanical method was followed, the colonies remained undifferentiated up to 15 passages and three ES cell-like cell lines were produced (gES-1, gES-2, and gES-3). However, enzymatic disaggregation resulted in differentiation. The undifferentiated cells showed stem cell like morphological features, normal karyotype, and expressed stem cell specific surface markers like alkaline phosphatase, TRA-1-61, TRA-1-81, and intracellular markers Oct4, Sox2, and Nanog. Following prolonged culture of the ES cell-like cells were differentiated into several types of cells including neuron like and epithelium-like cells. In conclusion, goat embryonic stem cell-like cells can be isolated from in vitro produced goat embryos and can be maintained for long periods in culture.
Collapse
Affiliation(s)
- Arun Kumar De
- Animal Biotechnology Centre, National Dairy Research Institute, Karnal, Haryana, India
| | | | | | | | | |
Collapse
|
25
|
Joubin K, Richardson A, Novoa N, Tu E, Tomishima MJ. The endothelial cell line bEnd.3 maintains human pluripotent stem cells. Stem Cells Dev 2012; 21:2312-21. [PMID: 22224974 DOI: 10.1089/scd.2011.0501] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Endothelial cells line blood vessels and coordinate many aspects of vascular biology. More recent work has shown that endothelial cells provide a key niche in vivo for neural stem cells. In vitro, endothelial cells secrete a factor that expands neural stem cells while inhibiting their differentiation. Here, we show that a transformed mouse endothelial cell line (bEnd.3) maintains human pluripotent stem cells in an undifferentiated state. bEnd.3 cells have a practical advantage over mouse embryonic fibroblasts for pluripotent stem cell maintenance since they can be expanded in vitro and engineered to express genes of interest. We demonstrate this capability by producing fluorescent and drug-resistant feeder cells. Further, we show that bEnd.3 secretes an activity that maintains human embryonic stem cells without direct contact.
Collapse
Affiliation(s)
- Katherine Joubin
- Developmental Biology Program, SKI Stem Cell Research Facility, Center for Stem Cell Biology, Sloan-Kettering Institute, New York, New York, USA
| | | | | | | | | |
Collapse
|
26
|
Yang C, Ji L, Yue W, Shi SS, Wang RY, Li YH, Xie XY, Xi JF, He LJ, Nan X, Pei XT. Human fetal liver stromal cells expressing erythropoietin promote hematopoietic development from human embryonic stem cells. Cell Reprogram 2012; 14:88-97. [PMID: 22313114 DOI: 10.1089/cell.2011.0013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Blood cells transfusion and hematopoietic stem cells (HSCs) transplantation are important methods for cell therapy. They are widely used in the treatment of incurable hematological disorder, infectious diseases, genetic diseases, and immunologic deficiency. However, their availability is limited by quantity, capacity of proliferation and the risk of blood transfusion complications. Recently, human embryonic stem cells (hESCs) have been shown to be an alternative resource for the generation of hematopoietic cells. In the current study, we describe a novel method for the efficient production of hematopoietic cells from hESCs. The stable human fetal liver stromal cell lines (hFLSCs) expressing erythropoietin (EPO) were established using the lentiviral system. We observed that the supernatant from the EPO transfected hFLSCs could induce the hESCs differentiation into hematopoietic cells, especially erythroid cells. They not only expressed fetal and embryonic globins but also expressed the adult-globin chain on further maturation. In addition, these hESCs-derived erythroid cells possess oxygen-transporting capacity, which indicated hESCs could generate terminally mature progenies. This should be useful for ultimately developing an animal-free culture system to generate large numbers of erythroid cells from hESCs and provide an experimental model to study early human erythropoiesis.
Collapse
Affiliation(s)
- Chao Yang
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Transfusion Medicine, Beijing, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Sánchez L, Gutierrez-Aranda I, Ligero G, Martín M, Ayllón V, Real PJ, Ramos-Mejía V, Bueno C, Menendez P. Maintenance of human embryonic stem cells in media conditioned by human mesenchymal stem cells obviates the requirement of exogenous basic fibroblast growth factor supplementation. Tissue Eng Part C Methods 2012; 18:387-96. [PMID: 22136131 DOI: 10.1089/ten.tec.2011.0546] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Despite the improvements in the human embryonic stem cell (hESC) culture systems, very similar conditions to those used to maintain hESCs on mouse feeders are broadly applied to culture methods based on human feeders. Indeed, basic fibroblast growth factor (bFGF), a master hESC-sustaining factor, is still added in nearly all medium formulations for hESC propagation. Human foreskin fibroblasts (HFFs) and mesenchymal stem cells (MSCs) used as feeders have recently been reported to support hESC growth without exogenous bFGF. However, whether hESCs may be maintained undifferentiated without exogenous bFGF using media conditioned (CM) by human feeders remains elusive. We hypothesize that HFFs and hMSCs are likely to be functionally different and therefore the mechanisms by which HFF-CM and MSC-CM support undifferentiated growth of hESCs may differ. We have thus determined whether HFF-CM and/or MSC-CM sustain feeder-free undifferentiated growth of hESC without exogenous supplementation of bFGF. We report that hMSCs synthesize higher levels of endogenous bFGF than HFFs. Accordingly and in contrast to HFF-CM, MSC-CM produced without the addition of exogenous bFGF supports hESC pluripotency and culture homeostasis beyond 20 passages without the need of bFGF supplementation. hESCs maintained without exogenous bFGF in MSC-CM retained hESC morphology and expression of pluripotency surface markers and transcription factors, formed teratomas, and showed spontaneous and lineage-directed in vitro differentiation capacity. Our data indicate that MSC-CM, but not HFF-CM, provides microenvironment cues supporting feeder-free long-term maintenance of pluripotent hESCs and obviates the requirement of exogenous bFGF at any time.
Collapse
Affiliation(s)
- Laura Sánchez
- GENYO-Centre Pfizer-University of Granada-Government of Andalucía for Genomic and Oncological Research, Avda de la Ilustración, Granada, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Basic fibroblast growth factor supports expansion of mouse compact bone-derived mesenchymal stem cells (MSCs) and regeneration of bone from MSC in vivo. J Mol Histol 2011; 43:223-33. [PMID: 22203245 DOI: 10.1007/s10735-011-9385-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2011] [Accepted: 12/17/2011] [Indexed: 10/14/2022]
Abstract
Some progress has been made in development of methods to regenerate bone from cultured cells, however no method is put to practical use. Here, we developed methods to isolate, purify, and expand mesenchymal stem cells (MSCs) from mouse compact bone that may be used to regenerate bone in vivo. These cells were maintained in long-term culture and were capable of differentiating along multiple lineages, including chondrocyte, osteocyte, and adipocyte trajectories. We used standard cell isolation and culture methods to establish cell cultures from mouse compact bone and bone marrow. Cultures were grown in four distinct media to determine the optimal composition of culture medium for bone-derived MSCs. Putative MSCs were subjected to flow cytometry, alkaline phosphatase assays, immunohistochemical staining, and several differentiation assays to assess cell identity, protein expression, and developmental potential. Finally, we used an in vivo bone formation assay to determine whether putative MSCs were capable of regenerating bone. We found that compact bone of mice was a better source of MCSs than the bone marrow, that growth in plastic flasks served to purify MSCs from hematopoietic cells, and that MSCs grown in basic fibroblast growth factor (bFGF)-conditioned medium were, based on multiple criteria, superior to those grown in leukemia inhibitory factor-conditioned medium. Moreover, we found that the MSCs isolated from compact bone and grown in bFGF-conditioned medium were capable of supporting bone formation in vivo. The methods and results described here have implications for understanding MSC biology and for clinical purpose.
Collapse
|
29
|
Zhu WZ, Van Biber B, Laflamme MA. Methods for the derivation and use of cardiomyocytes from human pluripotent stem cells. Methods Mol Biol 2011; 767:419-31. [PMID: 21822893 DOI: 10.1007/978-1-61779-201-4_31] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The availability of human cardiomyocytes derived from embryonic stem cells (ESCs) has generated -considerable excitement, as these cells are an excellent model system for studying myocardial development and may have eventual application in cell-based cardiac repair. Cardiomyocytes derived from the related induced pluripotent stem cells (iPSCs) have similar properties, but also offer the prospects of patient-specific disease modeling and cell therapies. Unfortunately, the methods by which cardiomyocytes have been historically generated from pluripotent stem cells are unreliable and typically result in preparations of low cardiac purity (typically <1% cardiomyocytes). We detail here the methods for a recently reported directed cardiac differentiation protocol, which involves the serial application of two growth factors known to be involved in early embryonic heart development, activin A, and bone morphogenetic protein-4 (BMP-4). This protocol reliably yields preparations of 30-60% cardiomyocytes, which can then be further enriched to >90% cardiomyocytes using straightforward physical methods.
Collapse
Affiliation(s)
- Wei-Zhong Zhu
- Laflamme Lab, Pathology, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | | | | |
Collapse
|
30
|
Lee WY, Kim J, Gil CH, Lee JH, Song H, Kim JH, Chung HM. Maintenance of human pluripotent stem cells using 4SP-hFGF2-secreting STO cells. Stem Cell Res 2011; 7:210-8. [DOI: 10.1016/j.scr.2011.05.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2010] [Revised: 05/11/2011] [Accepted: 05/16/2011] [Indexed: 02/02/2023] Open
|
31
|
Dutta R, Malakar D, Khate K, Sahu S, Akshey Y, Mukesh M. A comparative study on efficiency of adult fibroblast, putative embryonic stem cell and lymphocyte as donor cells for production of handmade cloned embryos in goat and characterization of putative ntES cells obtained from these embryos. Theriogenology 2011; 76:851-63. [DOI: 10.1016/j.theriogenology.2011.03.025] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2010] [Revised: 02/22/2011] [Accepted: 03/04/2011] [Indexed: 10/18/2022]
|
32
|
Wang S, Tian R, Li L, Figeys D, Wang L. An enhanced chemically defined SILAC culture system for quantitative proteomics study of human embryonic stem cells. Proteomics 2011; 11:4040-6. [PMID: 21770031 DOI: 10.1002/pmic.201100052] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2011] [Revised: 04/04/2011] [Accepted: 05/03/2011] [Indexed: 01/25/2023]
Abstract
Stable isotope labeling by SILAC-based quantitative proteomics analysis provides an unprecedented tool for the study of mechanisms underlying the self-renewal and differentiation of human embryonic stem cells (hESCs). While we recently reported a chemically defined SILAC culture system specific for a rare cell proteomic reactor (R. Tian et al., Mol. Cell. Proteomics 2011, 10, M110.000679), total hESC yield, prolonged self-renewal capacity (i.e.<12 days), and laborious procedure remain substantial hurdles for its conventional application in hESC studies. Here, we devised an enhanced SILAC culture system consisting of a new chemically defined SILAC-medium and a novel culture protocol. As a result, with much less culture maneuvers, approximately 40-fold greater hESCs were produced than the system reported previously. Moreover, the enhanced SILAC culture system was sufficient to support the self-renewal of hESCs for >60 days and was also highly reproducible. As such, it provides a new platform that can be readily adapted by general laboratory for further comprehensive SILAC-based proteomics analysis of hESCs and induced pluripotent stem cells.
Collapse
Affiliation(s)
- Shuai Wang
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | | | | | | | | |
Collapse
|
33
|
Ramos-Mejia V, Bueno C, Roldan M, Sanchez L, Ligero G, Menendez P, Martin M. The adaptation of human embryonic stem cells to different feeder-free culture conditions is accompanied by a mitochondrial response. Stem Cells Dev 2011; 21:1145-55. [PMID: 21671728 DOI: 10.1089/scd.2011.0248] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The mitochondrial contribution to the maintenance of human embryonic stem cell (hESC) pluripotency and culture homeostasis remains poorly understood. Here, we sought to determine whether hESC adaptation to different feeder-free culture conditions is linked to a mitochondrial adaptation. The expression of ESC pluripotency factors and parameters of mitochondrial contribution including mitochondrial membrane potential, mtDNA content, and the expression of master mitochondrial genes implicated in replication, transcription, and biogenesis were determined in 8 hESC lines maintained in 2 distinct human feeders-conditioned media (CM): human foreskin fibroblast-CM (HFF-CM) and mesenchymal stem cell-CM (MSC-CM). We show a robust parallel trend between the expression of ESC pluripotency factors and the mitochondrial contribution depending on the culture conditions employed to maintain the hESCs, with those in MSC-CM consistently displaying increased levels of pluripotency markers associated to an enhanced mitochondrial contribution. The differences in the mitochondrial status between hESCs maintained in MSC-CM versus HFF-CM respond to coordinated changes in mitochondrial gene expression and biogenesis. Importantly, the culture conditions determine the mitochondrial distribution within the stage-specific embryonic antigen 3 positive (SSEA3(+)) and negative (SSEA3(-)) isolated cell subsets. hESC colonies in MSC-CM display an "intrinsic" high mitochondrial status which may suffice to support undifferentiated growth, whereas hESC colonies maintained in HFF-CM show low mitochondrial status, possibly relying on the production of autologous niche with higher mitochondrial status to support pluripotency and culture homeostasis. Pluripotency markers and mitochondrial status are concomitantly reverted on changing the culture conditions, supporting an unrecognized role of the mitochondria in response to hESC culture adaptation. We provide the first evidence supporting that hESCs adaptation to different feeder-free culture systems relies on a mitochondrial response.
Collapse
Affiliation(s)
- Verónica Ramos-Mejia
- Andalusian Stem Cell Bank, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain
| | | | | | | | | | | | | |
Collapse
|
34
|
Kumar De A, Malakar D, Dutta R, Sahu S, Jena MK. Effect of leukaemia inhibitory factor and different types of feeder layers on growth and pluripotent nature of embryonic stem cells from in vitro produced goat (Capra hircus) blastocysts. JOURNAL OF APPLIED ANIMAL RESEARCH 2011. [DOI: 10.1080/09712119.2011.607898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Affiliation(s)
- Arun Kumar De
- a Animal Biotechnology Centre , National Dairy Research Institute , Karnal, Haryana, India
| | - Dhruba Malakar
- a Animal Biotechnology Centre , National Dairy Research Institute , Karnal, Haryana, India
| | - Rahul Dutta
- a Animal Biotechnology Centre , National Dairy Research Institute , Karnal, Haryana, India
| | - Shailendra Sahu
- a Animal Biotechnology Centre , National Dairy Research Institute , Karnal, Haryana, India
| | - Manoj Kumar Jena
- a Animal Biotechnology Centre , National Dairy Research Institute , Karnal, Haryana, India
| |
Collapse
|
35
|
Wu L, Leijten JCH, Georgi N, Post JN, van Blitterswijk CA, Karperien M. Trophic effects of mesenchymal stem cells increase chondrocyte proliferation and matrix formation. Tissue Eng Part A 2011; 17:1425-36. [PMID: 21247341 DOI: 10.1089/ten.tea.2010.0517] [Citation(s) in RCA: 211] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Previous studies showed that coculture of primary chondrocytes (PCs) with various sources of multipotent cells results in a higher relative amount of cartilage matrix formation than cultures containing only chondrocytes. The aim of this study was to investigate the mechanism underlying this observation. We used coculture pellet models of human mesenchymal stem cells (hMSCs) and human PCs or bovine PCs (bPCs) and studied the fate and the contribution to cartilage formation of the individual cell populations during coculture. Enhanced cartilage matrix deposition was confirmed by histology and quantification of total glycosaminoglycan deposition. Species-specific quantitative polymerase chain reaction demonstrated that cartilage matrix gene expression was mainly from bovine origin when bPCs were used. Short tandem repeat analysis and species-specific quantitative polymerase chain reaction analysis of genomic DNA demonstrated the near-complete loss of MSCs in coculture pellets after 4 weeks of culture. In coculture pellets of immortalized MSCs and bPCs, chondrocyte proliferation was increased, which was partly mimicked using conditioned medium, and simultaneously preferential apoptosis of immortalized MSCs was induced. Taken together, our data clearly demonstrate that in pellet cocultures of MSCs and PCs, the former cells disappear over time. Increased cartilage formation in these cocultures is mainly due to a trophic role of the MSCs in stimulating chondrocyte proliferation and matrix deposition by chondrocytes rather than MSCs actively undergoing chondrogenic differentiation.
Collapse
Affiliation(s)
- Ling Wu
- Department of Tissue Regeneration, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | | | | | | | | | | |
Collapse
|
36
|
Wianny F, Bourillot PY, Dehay C. Embryonic stem cells in non-human primates: An overview of neural differentiation potential. Differentiation 2011; 81:142-52. [PMID: 21296479 DOI: 10.1016/j.diff.2011.01.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2010] [Revised: 12/18/2010] [Accepted: 01/11/2011] [Indexed: 12/11/2022]
Abstract
Non-human primate (NHP) embryonic stem (ES) cells show unlimited proliferative capacities and a great potential to generate multiple cell lineages. These properties make them an ideal resource both for investigating early developmental processes and for assessing their therapeutic potential in numerous models of degenerative diseases. They share the same markers and the same properties with human ES cells, and thus provide an invaluable transitional model that can be used to address the safety issues related to the clinical use of human ES cells. Here, we review the available information on the derivation and the specific features of monkey ES cells. We comment on the capacity of primate ES cells to differentiate into neural lineages and the current protocols to generate self-renewing neural stem cells. We also highlight the signalling pathways involved in the maintenance of these neural cell types. Finally, we discuss the potential of monkey ES cells for neuronal differentiation.
Collapse
Affiliation(s)
- Florence Wianny
- Inserm, U846, Stem Cell and Brain Research Institute, 18 Avenue Doyen Lépine, 69500 Bron, France.
| | | | | |
Collapse
|
37
|
Manton KJ, Richards S, Van Lonkhuyzen D, Cormack L, Leavesley D, Upton Z. A chimeric vitronectin: IGF-I protein supports feeder-cell-free and serum-free culture of human embryonic stem cells. Stem Cells Dev 2011; 19:1297-305. [PMID: 20128657 DOI: 10.1089/scd.2009.0504] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The therapeutic use of human embryonic stem (hES) cells is severely limited by safety concerns regarding their culture in media containing animal-derived or nondefined factors and on animal-derived feeder cells. Thus, there is a pressing need to develop culture techniques that are xeno-free, fully defined, and synthetic. Our laboratory has discovered that insulin-like growth factor (IGF) and vitronectin (VN) bind to each other resulting in synergistic short-term functional effects in several cell types, including keratinocytes and breast epithelial cells. We have further refined this complex into a single chimeric VN:IGF-I protein that functionally mimics the effects obtained upon binding of IGF-I to VN. The aim of the current study was to determine whether hES cells can be serially propagated in feeder-cell-free and serum-free conditions using medium containing our novel chimeric VN:IGF-I protein. Here we demonstrate that hES cells can be serially propagated and retain their undifferentiated state in vitro for up to 35 passages in our feeder-cell-free, serum-free, chemically defined media. We have utilized real-time polymerase chain reaction (PCR), immunofluorescence, and fluorescence-activated cell sorter (FACS) analysis to show that the hES cells have maintained an undifferentiated phenotype. In vitro differentiation assays demonstrated that the hES cells retain their pluripotent potential and the karyotype of the hES cells remains unchanged. This study demonstrates that the novel, fully defined, synthetic VN:IGF-I chimera-containing medium described herein is a viable alternative to media containing serum, and that in conjunction with laminin-coated plates facilitates feeder-cell-free and serum-free growth of hES.
Collapse
Affiliation(s)
- Kerry J Manton
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, Australia.
| | | | | | | | | | | |
Collapse
|
38
|
Red blood cell production from immortalized progenitor cell line. Int J Hematol 2010; 93:5-9. [PMID: 21184289 DOI: 10.1007/s12185-010-0742-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2010] [Revised: 11/24/2010] [Accepted: 11/30/2010] [Indexed: 10/18/2022]
Abstract
The supply of transfusable red blood cells (RBCs) is not sufficient in many countries. If immortalized erythroid progenitor cell lines able to produce transfusable RBCs in vitro were established, they would be valuable resources. However, such cell lines have not been established. We have developed a robust method to establish immortalized erythroid progenitor cell lines following the induction of hematopoietic differentiation of mouse embryonic stem (ES) cells and have established many immortalized erythroid progenitor cell lines so far. Although their precise characteristics varied among cell lines, each of these lines could differentiate in vitro into more mature erythroid cells, including enucleated RBCs. Following transplantation of these erythroid cells into mice suffering from acute anemia, the cells proliferated transiently, subsequently differentiated into functional RBCs, and significantly ameliorated the acute anemia. Considering the number of human ES cell lines that have been established so far and the number of induced pluripotent stem cell lines that will be established in future, the intensive testing of a number of these lines for establishing immortalized erythroid progenitor cell lines may allow the establishment of such cell lines similar to the mouse erythroid progenitor cell lines.
Collapse
|
39
|
Hartung O, Huo H, Daley GQ, Schlaeger TM. Clump passaging and expansion of human embryonic and induced pluripotent stem cells on mouse embryonic fibroblast feeder cells. CURRENT PROTOCOLS IN STEM CELL BIOLOGY 2010; Chapter 1:Unit 1C.10. [PMID: 20814935 DOI: 10.1002/9780470151808.sc01c10s14] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The ability of human embryonic stem cells (hESCs) to differentiate into essentially all somatic cell types has made them a valuable tool for studying human development and has positioned them for broad applications in toxicology, regenerative medicine, and drug discovery. This unit describes a protocol for the large-scale expansion and maintenance of hESCs in vitro. hESC cultures must maintain a balance between the cellular states of pluripotency and differentiation; thus, researchers must use care when growing these technically demanding cells. The culture system is based largely on the use of a proprietary serum-replacement product and basic fibroblast growth factor (bFGF), with mouse embryonic fibroblasts as a feeder layer. These conditions provide the basis for relatively inexpensive maintenance and expansion of hESCs, as well as their engineered counterparts, human induced pluripotent stem cells (hiPSCs).
Collapse
Affiliation(s)
- Odelya Hartung
- Stem Cell Program, Children's Hospital Boston, Boston, Massachusetts, USA
| | | | | | | |
Collapse
|
40
|
Blanco-Gelaz MA, Suarez-Alvarez B, Ligero G, Sanchez L, Vidal-Castiñeira JR, Coto E, Moore H, Menendez P, Lopez-Larrea C. Endoplasmic reticulum stress signals in defined human embryonic stem cell lines and culture conditions. Stem Cell Rev Rep 2010; 6:462-72. [PMID: 20352530 DOI: 10.1007/s12015-010-9135-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Human embryonic stem cells (hESCs) are especially resistant to several cellular stresses, but the existence and induction of Endoplasmic Reticulum (ER) stress by culture conditions are unknown. Using qPCR, here, we investigated the behavior of the principal sensors of ER stress and their relation with the feeder layer, the type of conditioned media used in feeder free systems and the upregulation of several differentiation markers. We observed the preservation of pluripotency, and detected differential expression of differentiation markers in HS181 and SHEF1 hESCs growing on Adipose-derived mesenchymal stem cells (ASCs) and feeder-free system with different conditioned media (HEF-CM and ASC-CM). Taken together, these results demonstrate evidence of ER stress events that cells must resolve to survive and maintenance of markers of pluripotency. The early differentiation status defined could progress into a more differentiated state, and may be influenced by culture conditions.
Collapse
Affiliation(s)
- Miguel Angel Blanco-Gelaz
- Histocompatibility and Transplantation Unit, Hospital Universitario Central de Asturias, 33006, Oviedo, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Salli U, Fox TE, Carkaci-Salli N, Sharma A, Robertson GP, Kester M, Vrana KE. Propagation of undifferentiated human embryonic stem cells with nano-liposomal ceramide. Stem Cells Dev 2010; 18:55-65. [PMID: 18393629 DOI: 10.1089/scd.2007.0271] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Human embryonic stem (hES) cells, located on the periphery of the colonies, express the neuroectodermal markers nestin and Tuj1, suggesting a prematurely differentiated subgroup of cells. Here, we report that ceramide, a bioactive sphingolipid, selectively eliminates hES cells differentially expressing nestin and Tuj1. In contrast, undifferentiated cells are resistant to the apoptotic effects of ceramide. Ceramide-resistant hES cells express higher levels of the messenger RNA for ceramide-metabolizing enzymes that convert ceramide into pro-mitogenic metabolites. Based on these findings, we conducted long-term studies to determine whether liposomal ceramide can be used to maintain undifferentiated hES cells free of feeder cells. We continuously cultured hES cells on matrigel for 4 months with liposomal ceramide in a feeder cell-free system. Human ES cells treated with liposomal ceramide maintained their pluripotent state as determined by in vivo and in vitro differentiation studies and contained no chromosomal abnormalities. In conclusion, our findings suggest that exposure to ceramide provides a viable strategy to prevent premature hES cell differentiation and to maintain pluripotent stem cell populations in the absence of feeder cells.
Collapse
Affiliation(s)
- Ugur Salli
- Department of Pharmacology, Pennsylvania State University, College of Medicine, Hershey, Pennsylvania 17033-0850, USA
| | | | | | | | | | | | | |
Collapse
|
42
|
Suspension culture of undifferentiated human embryonic and induced pluripotent stem cells. Stem Cell Rev Rep 2010; 6:248-59. [PMID: 20431964 DOI: 10.1007/s12015-010-9149-y] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Alongside their contribution to research, human embryonic stem cells (hESC) may also prove valuable for cell-based therapies. Traditionally, these cells have been grown in adhesion culture either with or without feeder cells, allowing for their continuous growth as undifferentiated cells. However, to be applicable in therapy and industry they must be produced in a scalable and controlled process. Here we present for the first time a suspension culture system for undifferentiated hESC and induced pluripotent stem cells (iPSC), based on medium supplemented with the IL6RIL6 chimera (interleukin-6 receptor fused to interleukin-6), and basic fibroblast growth factor. Four hESC lines cultured in this system maintained all ESC features after 20 passages, including stable karyotype and pluripotency. Similar results were obtained when hESC were replaced with iPSC from two different cell lines. We demonstrate that the IL6RIL6 chimera supports the self-renewal and expansion of undifferentiated hESC and iPSC in suspension, and thus present another efficient system for large-scale propagation of undifferentiated pluripotent cells for clinical and translational applications.
Collapse
|
43
|
Zheng Z, de Iongh RU, Rathjen PD, Rathjen J. A requirement for FGF signalling in the formation of primitive streak-like intermediates from primitive ectoderm in culture. PLoS One 2010; 5:e12555. [PMID: 20838439 PMCID: PMC2933233 DOI: 10.1371/journal.pone.0012555] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2009] [Accepted: 08/06/2010] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Embryonic stem (ES) cells hold considerable promise as a source of cells with therapeutic potential, including cells that can be used for drug screening and in cell replacement therapies. Differentiation of ES cells into the somatic lineages is a regulated process; before the promise of these cells can be realised robust and rational methods for directing differentiation into normal, functional and safe cells need to be developed. Previous in vivo studies have implicated fibroblast growth factor (FGF) signalling in lineage specification from pluripotent cells. Although FGF signalling has been suggested as essential for specification of mesoderm and endoderm in vivo and in culture, the exact role of this pathway remains unclear. METHODOLOGY/PRINCIPAL FINDINGS Using a culture model based on early primitive ectoderm-like (EPL) cells we have investigated the role of FGF signalling in the specification of mesoderm. We were unable to demonstrate any mesoderm inductive capability associated with FGF1, 4 or 8 signalling, even when the factors were present at high concentrations, nor any enhancement in mesoderm formation induced by exogenous BMP4. Furthermore, there was no evidence of alteration of mesoderm sub-type formed with addition of FGF1, 4 or 8. Inhibition of endogenous FGF signalling, however, prevented mesoderm and favoured neural differentiation, suggesting FGF signalling was required but not sufficient for the differentiation of primitive ectoderm into primitive streak-like intermediates. The maintenance of ES cell/early epiblast pluripotent marker expression was also observed in cultures when FGF signalling was inhibited. CONCLUSIONS/SIGNIFICANCE FGF signalling has been shown to be required for the differentiation of primitive ectoderm to neurectoderm. This, coupled with our observations, suggest FGF signalling is required for differentiation of the primitive ectoderm into the germ lineages at gastrulation.
Collapse
Affiliation(s)
- Zhiqiang Zheng
- Department of Zoology, University of Melbourne, Parkville, Australia
| | - Robb U. de Iongh
- Department of Anatomy and Cell Biology, University of Melbourne, Parkville, Australia
| | - Peter D. Rathjen
- Department of Zoology, University of Melbourne, Parkville, Australia
| | - Joy Rathjen
- Department of Zoology, University of Melbourne, Parkville, Australia
| |
Collapse
|
44
|
Li L, Wang BH, Wang S, Moalim-Nour L, Mohib K, Lohnes D, Wang L. Individual cell movement, asymmetric colony expansion, rho-associated kinase, and E-cadherin impact the clonogenicity of human embryonic stem cells. Biophys J 2010; 98:2442-51. [PMID: 20513387 DOI: 10.1016/j.bpj.2010.02.029] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2009] [Revised: 02/11/2010] [Accepted: 02/12/2010] [Indexed: 02/07/2023] Open
Abstract
Clonality is, at present, the only means by which the self-renewal potential of a given stem cell can be determined. To assess the clonality of human embryonic stem cells (hESC), a protocol involving seeding wells at low cell densities is commonly used to surmount poor cloning efficiencies. However, factors influencing the accuracy of such an assay have not been fully elucidated. Using clonogenic assays together with time-lapse microscopy, numerical analyses, and regulated gene expression strategies, we found that individual and collective cell movements are inherent properties of hESCs and that they markedly impact the accuracy of clonogenic assays. Analyses of cell motility using mean-square displacement and paired migration correlation indicated that cell movements become more straight-line or ballistic and less random-walk as separation distance decreases. Such motility-induced reaggregation (rather than a true clone) occurs approximately 70% of the time if the distance between two hESCs is <6.4 mum, and is not observed if the distance is >150 mum. Furthermore, newly formed small hESC colonies have a predisposition toward the formation of larger colonies through asymmetric colony expansion and movement, which would not accurately reflect self-renewal and proliferative activity of a true hESC clone. Notably, inhibition of Rho-associated kinase markedly upregulated hESC migration and reaggregation, producing considerable numbers of false-positive colonies. Conversely, E-cadherin upregulation significantly augmented hESC clonogenicity via improved survival of single hESCs without influencing cell motility. This work reveals that individual cell movement, asymmetric colony expansion, Rho-associated kinase, and E-cadherin all work together to influence hESC clonogenicity, and provides additional guidance for improvement of clonogenic assays in the analysis of hESC self-renewal.
Collapse
Affiliation(s)
- Li Li
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|
45
|
Tian R, Wang S, Elisma F, Li L, Zhou H, Wang L, Figeys D. Rare cell proteomic reactor applied to stable isotope labeling by amino acids in cell culture (SILAC)-based quantitative proteomics study of human embryonic stem cell differentiation. Mol Cell Proteomics 2010; 10:M110.000679. [PMID: 20530636 DOI: 10.1074/mcp.m110.000679] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
The molecular basis governing the differentiation of human embryonic stem cells (hESCs) remains largely unknown. Systems-level analysis by proteomics provides a unique approach to tackle this question. However, the requirement of a large number of cells for proteomics analysis (i.e. 10(6)-10(7) cells) makes this assay challenging, especially for the study of rare events during hESCs lineage specification. Here, a fully integrated proteomics sample processing and analysis platform, termed rare cell proteomic reactor (RCPR), was developed for large scale quantitative proteomics analysis of hESCs with ∼50,000 cells. hESCs were completely extracted by a defined lysis buffer, and all of the proteomics sample processing procedures, including protein preconcentration, reduction, alkylation, and digestion, were integrated into one single capillary column with a strong cation exchange monolith matrix. Furthermore, on-line two-dimensional LC-MS/MS analysis was performed directly using RCPR as the first dimension strong cation exchange column. 2,281 unique proteins were identified on this system using only 50,000 hESCs. For stable isotope labeling by amino acids in cell culture (SILAC)-based quantitative study, a ready-to-use and chemically defined medium and an in situ differentiation procedure were developed for complete SILAC labeling of hESCs with well characterized self-renewal and differentiation properties. Mesoderm-enriched differentiation was studied by RCPR using 50,000 hESCs, and 1,086 proteins were quantified with a minimum of two peptides per protein. Of these, 56 proteins exhibited significant changes during mesoderm-enriched differentiation, and eight proteins were demonstrated for the first time to be overexpressed during early mesoderm development. This work provides a new platform for the study of rare cells and in particular for further elucidating proteins that govern the mesoderm lineage specification of human pluripotent stem cells.
Collapse
Affiliation(s)
- Ruijun Tian
- Ottawa Institute of Systems Biology, Ottawa, Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|
46
|
Li L, Wang S, Jezierski A, Moalim-Nour L, Mohib K, Parks RJ, Retta SF, Wang L. A unique interplay between Rap1 and E-cadherin in the endocytic pathway regulates self-renewal of human embryonic stem cells. Stem Cells 2010; 28:247-57. [PMID: 20039365 DOI: 10.1002/stem.289] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Regulatory mechanisms pertaining to the self-renewal of stem cells remain incompletely understood. Here, we show that functional interactions between small GTPase Rap1 and the adhesion molecule E-cadherin uniquely regulate the self-renewal of human embryonic stem cells (hESCs). Inhibition of Rap1 suppresses colony formation and self-renewal of hESCs, whereas overexpression of Rap1 augments hESC clonogenicity. Rap1 does not directly influence the expression of the pluripotency genes Oct4 and Nanog. Instead, it affects the endocytic recycling pathway involved in the formation and maintenance of E-cadherin-mediated cell-cell cohesion, which is essential for the colony formation and self-renewal of hESCs. Conversely, distinct from epithelial cells, disruption of E-cadherin mediated cell-cell adhesions induces lysosome delivery and degradation of Rap1. This in turn leads to a further downregulation of E-cadherin function and a subsequent reduction in hESC clonogenic capacity. These findings provide the first demonstration that the interplay between Rap1 and E-cadherin along the endocytic recycling pathway serves as a timely and efficient mechanism to regulate hESC self-renewal. Given the availability of specific activators for Rap1, this work provides a new perspective to enable better maintenance of human pluripotent stem cells.
Collapse
Affiliation(s)
- Li Li
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Yoon BS, Jun EK, Park G, Jun Yoo S, Moon JH, Soon Baik C, Kim A, Kim H, Kim JH, Young Koh G, Taek Lee H, You S. Optimal Suppression of Protein Phosphatase 2A Activity Is Critical for Maintenance of Human Embryonic Stem Cell Self-Renewal. Stem Cells 2010; 28:874-884. [DOI: 10.1002/stem.412] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Abstract
The self-renewal of embryonic stem cells involves a balance between processes governed by crosstalk between intrinsic and extrinsic factors. We hypothesized that protein serine/threonine phosphatase 2A (PP2A) may play a central role in the signaling pathways that regulate human embryonic stem cell (hESC) self-renewal. Biochemical analyses revealed that PP2A activity gradually increases over the course of hESC differentiation; PP2A/C and PP2A/A levels also increased. The overexpression of PP2A/C or the addition of PP2A activator C2-ceramide promoted hESC differentiation. Accordingly, the addition of PP2A inactivator okadaic acid (OA) maintained hESC self-renewal in the absence of basic fibroblast growth factor (bFGF). The hESCs maintained with OA expressed pluripotency markers and exhibited substantial telomerase activity with normal karyotypes. The hESCs were able to differentiate into derivatives of the three germ layers, both in vitro and in vivo. Furthermore, the addition of OA and bFGF enabled the maintenance of hESC self-renewal without feeder cells, even in chemically defined xeno-free media. These findings shed a light on the role of PP2A in hESC differentiation and provide a novel strategy for maintaining the self-renewal capability of hESC in bFGF-free, feeder cell-free, and xeno-free media through the optimal suppression of PP2A activity using OA.
Collapse
Affiliation(s)
- Byung Sun Yoon
- Laboratories of Cell Function Regulation, Korea University, Seoul, Korea
- Institute of Life Science and Natural Resources, Korea University, Seoul, Korea
| | - Eun Kyoung Jun
- Laboratories of Cell Function Regulation, Korea University, Seoul, Korea
| | - Gyuman Park
- Research Institute for Skin Image, Korea University Guro Hospital, Seoul, Korea
| | - Seung Jun Yoo
- Laboratories of Cell Function Regulation, Korea University, Seoul, Korea
| | - Jai-Hee Moon
- Laboratories of Cell Function Regulation, Korea University, Seoul, Korea
| | | | - Aeree Kim
- Department of Pathology, College of Medicine, Korea University Guro Hospital, Seoul, Korea
| | - Hyunggee Kim
- Laboratories of Cell Function Regulation, Korea University, Seoul, Korea
| | - Jong-Hoon Kim
- Stem Cell Biology, College of Life Sciences and Biotechnology, Korea University, Seoul, Korea
| | - Gou Young Koh
- National Research Laboratory of Vascular Biology, Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Korea
| | - Hoon Taek Lee
- Department of Animal Biotechnology, Bio-Organ Research Center/Animal Resources Research Center, Konkuk University, Seoul, Korea
| | - Seungkwon You
- Laboratories of Cell Function Regulation, Korea University, Seoul, Korea
| |
Collapse
|
48
|
Vo E, Hanjaya-Putra D, Zha Y, Kusuma S, Gerecht S. Smooth-Muscle-Like Cells Derived from Human Embryonic Stem Cells Support and Augment Cord-Like Structures In Vitro. Stem Cell Rev Rep 2010; 6:237-47. [DOI: 10.1007/s12015-010-9144-3] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
49
|
Yamaoka H, Nishizawa S, Asawa Y, Fujihara Y, Ogasawara T, Yamaoka K, Nagata S, Takato T, Hoshi K. Involvement of fibroblast growth factor 18 in dedifferentiation of cultured human chondrocytes. Cell Prolif 2010; 43:67-76. [PMID: 19909293 PMCID: PMC6496437 DOI: 10.1111/j.1365-2184.2009.00655.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2008] [Accepted: 05/11/2009] [Indexed: 12/26/2022] Open
Abstract
OBJECTIVE Chondrocytes inevitably decrease production of cartilaginous matrices during long-term cultures with repeated passaging; this is termed dedifferentiation. To learn more concerning prevention of dedifferentiation, we have focused here on the fibroblast growth factor (FGF) family that influences chondrocyte proliferation or differentiation. MATERIALS AND METHODS We have compared gene expression between differentiated cells in passage 3 (P3) and dedifferentiated ones in P8 of human cultured chondrocytes. We also performed ligand administration of the responsive factor or its gene silencing, using small interfering RNA (siRNA). RESULTS FGFs 1, 5, 10, 13 and 18 were higher at P8 compared to P3, while FGFs 9 and 14 were lower. Especially, FGF18 showed a 10-fold increase by P8. Ligand administration of FGF18 in the P3 cells, or its gene silencing using siRNA in the P8 cells, revealed dose-dependent increase and decrease respectively in type II collagen/type I collagen ratio. Exogenous FGF18 also upregulated expression of transforming growth factor beta (TGF-beta), the anabolic factor of chondrocytes, in P3 chondrocytes, but P8 cells maintained a low level of TGF-beta expression, suggesting a decrease in responsiveness of TGF-beta to FGF18 stimulation in the dedifferentiated chondrocytes. CONCLUSION FGF18 seems to play a role in maintenance of chondrocyte properties, although its expression was rather high in dedifferentiated chondrocytes. Upregulation of FGF18 in dedifferentiated chondrocytes implied that it may be a marker of dedifferentiation.
Collapse
Affiliation(s)
- H. Yamaoka
- Departments of Cartilage & Bone Regeneration (Fujisoft), Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Plastic, Oral and Maxillofacial Surgery, Teikyo University School of Medicine, Tokyo, Japan
| | - S. Nishizawa
- Departments of Cartilage & Bone Regeneration (Fujisoft), Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Pharmacy, Teikyo University Hospital, Tokyo, Japan
| | - Y. Asawa
- Departments of Cartilage & Bone Regeneration (Fujisoft), Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Y. Fujihara
- Departments of Cartilage & Bone Regeneration (Fujisoft), Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - T. Ogasawara
- Departments of Cartilage & Bone Regeneration (Fujisoft), Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - K. Yamaoka
- Department of Pharmacy, Teikyo University Hospital, Tokyo, Japan
| | - S. Nagata
- Nagata Microtia and Reconstructive Plastic Surgery Clinic, Saitama, Japan
| | - T. Takato
- Departments of Sensory & Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - K. Hoshi
- Departments of Cartilage & Bone Regeneration (Fujisoft), Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
50
|
Lin G, Martins-Taylor K, Xu RH. Human embryonic stem cell derivation, maintenance, and differentiation to trophoblast. Methods Mol Biol 2010; 636:1-24. [PMID: 20336513 DOI: 10.1007/978-1-60761-691-7_1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Since the first report of derivation of human embryonic stem cell (hESC) lines in 1998, many progresses have been achieved to reliably and efficiently derive, maintain, and differentiate this therapeutically promising cell type. This chapter introduces some basic and widely recognized methods that we use in our hESC core laboratory. Specifically, it includes methods for (1) deriving hESC lines without using enzyme and antibody to isolate the inner cell mass; (2) sustaining hESC self-renewal under feeder-dependent, feeder-conditioned, and defined conditions as well as pluripotency validation and quality control assays; and (3) inducing hESC differentiation to trophoblast with BMP4.
Collapse
Affiliation(s)
- Ge Lin
- Department of Genetics and Developmental Biology, Stem Cell Institute, University of Connecticut Health Center, Farmington, CT, USA
| | | | | |
Collapse
|