1
|
Characterization of Endoplasmic Reticulum (ER) in Human Pluripotent Stem Cells Revealed Increased Susceptibility to Cell Death upon ER Stress. Cells 2020; 9:cells9051078. [PMID: 32357563 PMCID: PMC7291192 DOI: 10.3390/cells9051078] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 04/22/2020] [Accepted: 04/23/2020] [Indexed: 12/19/2022] Open
Abstract
Human pluripotent stem cells (hPSCs), such as embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), have a well-orchestrated program for differentiation and self-renewal. However, the structural features of unique proteostatic-maintaining mechanisms in hPSCs and their features, distinct from those of differentiated cells, in response to cellular stress remain unclear. We evaluated and compared the morphological features and stress response of hPSCs and fibroblasts. Compared to fibroblasts, electron microscopy showed simpler/fewer structures with fewer networks in the endoplasmic reticulum (ER) of hPSCs, as well as lower expression of ER-related genes according to meta-analysis. As hPSCs contain low levels of binding immunoglobulin protein (BiP), an ER chaperone, thapsigargin treatment sharply increased the gene expression of the unfolded protein response. Thus, hPSCs with decreased chaperone function reacted sensitively to ER stress and entered apoptosis faster than fibroblasts. Such ER stress-induced apoptotic processes were abolished by tauroursodeoxycholic acid, an ER-stress reliever. Hence, our results revealed that as PSCs have an underdeveloped structure and express fewer BiP chaperone proteins than somatic cells, they are more susceptible to ER stress-induced apoptosis in response to stress.
Collapse
|
2
|
Jeon RH, Lee WJ, Son YB, Bharti D, Shivakumar SB, Lee SL, Rho GJ. PPIA, HPRT1, and YWHAZ Genes Are Suitable for Normalization of mRNA Expression in Long-Term Expanded Human Mesenchymal Stem Cells. BIOMED RESEARCH INTERNATIONAL 2019; 2019:3093545. [PMID: 31240211 PMCID: PMC6556274 DOI: 10.1155/2019/3093545] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 05/09/2019] [Indexed: 12/15/2022]
Abstract
Long-term expansion of mesenchymal stem cells (MSCs) under defined culture conditions is necessary in human stem cell therapy. However, it alters the characteristics of MSCs. Since quantitative real time polymerase chain reaction (qRT-PCR) is widely used as one of the key analytical methods for comparative characterization, the validation of reference genes (RGs) for normalization under each experimental condition is important to achieve reliable qRT-PCR results. Therefore, the most stable RGs for long-term expanded bone marrow- and umbilical cord blood-derived MSCs (BM-MSCs and UCB-MSCs) under defined culture conditions for up to 20 passages were evaluated. The more apparent alterations in characteristics such as differentiation capacity, proliferation, senescence, and the expression of RGs were noted in BM-MSCs than UCB-MSCs during long-term expansion. The RG validation programs (GeNorm and NormFinder) suggested that PPIA, HPRT1, and YWHAZ were suitable for normalization in qRT-PCR regardless of MSC types and long-term culture expansion, and the traditional RGs (ACTB and GAPDH) were less stable in long-term expanded MSCs. In addition, the use of these RGs for normalization of OCT4 expression in long-term expanded BM-MSCs showed that a less stable RG (GAPDH) showed contrasting data compared to other RGs. Therefore, the use of RGs such as PPIA, HPRT1, and YWHAZ for normalization in qRT-PCR experiments is highly recommended for long-term expanded MSCs to generate accurate and reliable data.
Collapse
Affiliation(s)
- Ryoung-Hoon Jeon
- College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Won-Jae Lee
- College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Young-Bum Son
- College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Dinesh Bharti
- College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea
| | | | - Sung-Lim Lee
- College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Gyu-Jin Rho
- College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea
| |
Collapse
|
3
|
Leukemia Inhibitory Factor Increases Survival of Pluripotent Stem Cell-Derived Cardiomyocytes. J Cardiovasc Transl Res 2017; 11:1-13. [PMID: 29019149 DOI: 10.1007/s12265-017-9769-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 10/02/2017] [Indexed: 01/22/2023]
Abstract
Leukemia inhibitory factor (LIF) is a growth factor with pleiotropic biological functions. It has been reported that LIF acts at different stages during mesoderm development. Also, it has been shown that LIF has a cytoprotective effect on neonatal murine cardiomyocytes (CMs) in culture, but little is known about the role of LIF during human cardiogenesis. Thus, we analyzed the effects of LIF on human pluripotent stem cells (PSC) undergoing cardiac differentiation. We first showed that LIF is expressed in the human heart during early development. We found that the addition of LIF within a precise time window during the in vitro differentiation process significantly increased CMs viability. This finding was associated to a decrease in the expression of pro-apoptotic protein Bax, which coincides with a reduction of the apoptotic rate. Therefore, the addition of LIF may represent a promising strategy for increasing CMs survival derived from PSCs.
Collapse
|
4
|
Zhang P, Chen X, Zheng Y, Zhu J, Qin Y, Lv Y, Zeng W. Long-Term Propagation of Porcine Undifferentiated Spermatogonia. Stem Cells Dev 2017; 26:1121-1131. [PMID: 28474535 DOI: 10.1089/scd.2017.0018] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Spermatogonial stem cells (SSCs) provide the foundation for spermatogenesis and fertility throughout the adult life of a male. Genetic manipulations of SSCs combined with germ cell transplantation present a novel approach for gene therapy and production of genetically modified animals. However, the rarity of SSCs within mammalian testes remains an impediment to related applications, making in vitro expansion of SSCs a prerequisite. Nevertheless, long-term culture systems of SSCs from large animals have not been established yet. In this study, we developed an optimized in vitro culture condition for porcine undifferentiated spermatogonia. The germ cells were isolated and enriched from 7-day-old porcine testes by an optimized differential planting. We tested different feeder layers and found that neonatal autologous Sertoli cells acted better than the SIM mouse embryo-derived thioguanine- and ouabain-resistant (STO) cell line and adult Sertoli cells. The effects of several growth factors were also investigated. Using neonatal Sertoli cells as feeder and Dulbecco's modified eagle medium: nutrient mixture F-12 (DMEM/F12) culture medium supplemented with 10% KSR and four cytokines, the undifferentiated spermatogonia can proliferate in vitro for at least 2 months without loss of stemness. The expression of SSC markers indicated that the cultured cells maintained SSC expression profiles. Moreover, xenotransplantation and in vitro induction showed that the long-term cultured cells preserved the capacity to colonize in vivo and differentiate in vitro, respectively, demonstrating the presence of SSCs in the cultured cells. In conclusion, the conditions described in this study can support the normal proliferation of porcine undifferentiated spermatogonia with stemness and normal karyotype for at least 2 months. This culture system will serve as a basic refinement in the future studies and facilitate studies on SSC biology and genetic manipulation of male germ cells.
Collapse
Affiliation(s)
- Pengfei Zhang
- 1 College of Animal Science and Technology, Northwest A&F University , Shaanxi, China
| | - Xiaoxu Chen
- 1 College of Animal Science and Technology, Northwest A&F University , Shaanxi, China
| | - Yi Zheng
- 2 Center for Reproductive Medicine, Amsterdam Research Institute Reproduction and Development, Academic Medical Centre, University of Amsterdam , Amsterdam, the Netherlands
| | - Jinshen Zhu
- 1 College of Animal Science and Technology, Northwest A&F University , Shaanxi, China
| | - Yuwei Qin
- 1 College of Animal Science and Technology, Northwest A&F University , Shaanxi, China
| | - Yinghua Lv
- 1 College of Animal Science and Technology, Northwest A&F University , Shaanxi, China
| | - Wenxian Zeng
- 1 College of Animal Science and Technology, Northwest A&F University , Shaanxi, China
| |
Collapse
|
5
|
Zhao Z, Ma Y, Chen Z, Liu Q, Li Q, Kong D, Yuan K, Hu L, Wang T, Chen X, Peng Y, Jiang W, Yu Y, Liu X. Effects of Feeder Cells on Dopaminergic Differentiation of Human Embryonic Stem Cells. Front Cell Neurosci 2016; 10:291. [PMID: 28066186 PMCID: PMC5168467 DOI: 10.3389/fncel.2016.00291] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 12/05/2016] [Indexed: 01/30/2023] Open
Abstract
Mouse embryonic fibroblasts (MEFs) and human foreskin fibroblasts (HFFs) are used for the culture of human embryonic stem cells (hESCs). MEFs and HFFs differed in their capacity to support the proliferation and pluripotency of hESCs and could affect cardiac differentiation potential of hESCs. The aim of this study was to evaluate the effect of MEFs and HFFs feeders on dopaminergic differentiation of hESCs lines. To minimize the impact of culture condition variation, two hESCs lines were cultured on mixed feeder cells (MFCs, MEFs: HFFs = 1:1) and HFFs feeder, respectively, and then were differentiated into dopaminergic (DA) neurons under the identical protocol. Dopaminergic differentiation was evaluated by immunocytochemistry, quantitative fluorescent real-time PCR, transmission and scanning electron microscopy, and patch clamp. Our results demonstrated that these hESCs-derived neurons were genuine and functional DA neurons. However, compared to hESCs line on MFCs feeder, hESCs line on HFFs feeder had a higher proportion of tyrosine hydroxylase (TH) positive cells and expressed higher levels of FOXA2, PITX3, NURR1, and TH genes. In addition, the values of threshold intensity and threshold membrane potential of DA neurons from hESCs line on HFFs feeder were lower than those of DA neurons from hESCs line on the MFCs feeder. In conclusion, HFFs feeder not only facilitated the differentiation of hESCs cells into dopaminergic neurons, but also induced hESCs-derived DA neurons to express higher electrophysiological excitability. Therefore, feeder cells could affect not only dopaminergic differentiation potential of different hESCs lines, but also electrophysiological properties of hESCs-derived DA neurons.
Collapse
Affiliation(s)
- Zhenqiang Zhao
- Department of Neurology, Jinling Hospital, Southern Medical UniversityNanjing, China; Department of Neurology, First Affiliated Hospital, Hainan Medical UniversityHaikou, China
| | - Yanlin Ma
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical UniversityGuangzhou, China; Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Hainan Reproductive Medical Center, First Affiliated Hospital, Hainan Medical UniversityHaikou, China
| | - Zhibin Chen
- Department of Neurology, First Affiliated Hospital, Hainan Medical University Haikou, China
| | - Qian Liu
- Department of Neurology, Jinling Hospital, Southern Medical University Nanjing, China
| | - Qi Li
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Hainan Reproductive Medical Center, First Affiliated Hospital, Hainan Medical University Haikou, China
| | - Deyan Kong
- Department of Neurology, Jinling Hospital, Southern Medical UniversityNanjing, China; Department of Neurology, Affiliated Ruikang Hospital, Guangxi Traditional Chinese Medical UniversityNanning, China
| | - Kunxiong Yuan
- Department of Neurology, Jinling Hospital, Southern Medical UniversityNanjing, China; Department of Neurology, Central HospitalShenzhen, China
| | - Lan Hu
- Department of Laboratory Medicines, First Affiliated Hospital, Hainan Medical University Haikou, China
| | - Tan Wang
- Department of Neurology, First Affiliated Hospital, Hainan Medical University Haikou, China
| | - Xiaowu Chen
- Department of Neurology, First Affiliated Hospital, Hainan Medical University Haikou, China
| | - Yanan Peng
- Department of Neurology, First Affiliated Hospital, Hainan Medical University Haikou, China
| | - Weimin Jiang
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Hainan Reproductive Medical Center, First Affiliated Hospital, Hainan Medical University Haikou, China
| | - Yanhong Yu
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University Guangzhou, China
| | - Xinfeng Liu
- Department of Neurology, Jinling Hospital, Southern Medical University Nanjing, China
| |
Collapse
|
6
|
Hogg K, Western PS. Differentiation of Fetal Male Germline and Gonadal Progenitor Cells Is Disrupted in Organ Cultures Containing Knockout Serum Replacement. Stem Cells Dev 2015; 24:2899-911. [PMID: 26393524 DOI: 10.1089/scd.2015.0196] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Germ cells occupy a unique place in development through their requirement to maintain underlying totipotency while producing highly differentiated gametes. This is reflected in the expression of key regulators of pluripotency in the fetal germline and the ability of these cells to form pluripotent stem cells and germ cell tumors. Culture of whole fetal testes, including germ cells, provides a key model for studying gonad and germline development, but it is critical that such models mimic physiological development as closely as possible. We aimed to determine the effects of differing culture conditions, including serum-free and serum-containing conditions, on fetal germ cell and testis development. We tested a commonly used model that employs knockout serum replacement (KSR) to provide more defined culture conditions than media containing fetal bovine serum (FBS). In FBS conditions, cell cycle parameters in germ and Sertoli cells closely resembled normal development. In contrast, KSR significantly inhibited male germ cell entry into mitotic arrest, a key milestone in male germline development. Moreover, KSR disrupted molecular control of cell cycle and inhibited the transcription of a range of male germ cell differentiation markers. In the somatic compartment, KSR stimulated proliferation and inhibited differentiation in Sertoli cells. These data demonstrate that KSR substantially alters germ and somatic cell differentiation in fetal testis culture and should not be used to replicate normal gonadal development. In contrast, basal media with or without serum support germ and supporting cell differentiation and cell cycle dynamics that are in line with in vivo characteristics.
Collapse
Affiliation(s)
- Kirsten Hogg
- 1 Centre for Genetic Diseases, Hudson Institute of Medical Research , Clayton, Victoria, Australia .,2 Department of Molecular and Translational Science, Monash University , Melbourne, Victoria, Australia
| | - Patrick S Western
- 1 Centre for Genetic Diseases, Hudson Institute of Medical Research , Clayton, Victoria, Australia .,2 Department of Molecular and Translational Science, Monash University , Melbourne, Victoria, Australia
| |
Collapse
|
7
|
Sterthaus O, Feutz AC, Zhang H, Pletscher F, Bruder E, Miny P, Lezzi G, De Geyter M, De Geyter C. Gene expression profiles of similarly derived human embryonic stem cell lines correlate with their distinct propensity to exit stemness and their different differentiation behavior in culture. Cell Reprogram 2014; 16:185-95. [PMID: 24811852 DOI: 10.1089/cell.2013.0089] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Four normal-karyotype human embryonic stem cell (hESC) lines were generated using the same protocol and maintained under identical conditions. Despite these precautions, gene expression patterns were found to be dissimilar among the four lines. The observed differences were typical of each cell line, correlated with their distinct propensity to exit stemness, created heterogeneity among the cells during cell line maintenance, and correlated with their altered capacity as a source of differentiated cells. The capacity of some cell lines to give rise to more, and more mature, neurons within comparable time frames of directed differentiation reflected the distinct proportions of cells already predifferentiated at the onset. These findings demonstrate that the subsequent stages of neural differentiation were altered both in a quantitative and timely fashion. As a consequence, cell lines with apparent better and quicker ability to produce neurons were actually the less capable of reproducing proper differentiation. Previous data suggested that cell lines able to generate more neurons faster would be more suitable to clinical application. Our analysis of the differentiation process strongly suggests the opposite. The spontaneous tendency to predifferentiate of any particular hESC line should be known because it clearly impacts further experimental results.
Collapse
Affiliation(s)
- Oliver Sterthaus
- 1 Clinic of Gynecological Endocrinology and Reproductive Medicine, University of Basel , CH-4031, Basel, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Shaltouki A, Peng J, Liu Q, Rao MS, Zeng X. Efficient generation of astrocytes from human pluripotent stem cells in defined conditions. Stem Cells 2014; 31:941-52. [PMID: 23341249 DOI: 10.1002/stem.1334] [Citation(s) in RCA: 200] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Accepted: 12/24/2012] [Indexed: 01/07/2023]
Abstract
Astrocytes can be generated from various tissue sources including human pluripotent stem cells (PSC). In this manuscript, we describe a chemically defined xeno-free medium culture system for rapidly generating astrocytes from neural stem cells derived from PSC. We show that astrocyte development in vitro, mimics normal development in vivo, and also passes through a CD44(+) astrocyte precursor stage. Astrocytes generated by our method display similar gene expression patterns, morphological characteristics and functional properties to primary astrocytes, and they survive and integrate after xenotransplantation. Whole genome expression profiling of astrocyte differentiation was performed at several time points of differentiation, and the results indicate the importance of known regulators and identify potential novel regulators and stage-specific lineage markers.
Collapse
|
9
|
Lesniak D, Sabri S, Xu Y, Graham K, Bhatnagar P, Suresh M, Abdulkarim B. Spontaneous epithelial-mesenchymal transition and resistance to HER-2-targeted therapies in HER-2-positive luminal breast cancer. PLoS One 2013; 8:e71987. [PMID: 23991019 PMCID: PMC3753362 DOI: 10.1371/journal.pone.0071987] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Accepted: 07/05/2013] [Indexed: 01/05/2023] Open
Abstract
Resistance to trastuzumab, a rationally designed HER-2-targeting antibody, remains a major hurdle in the management of HER-2-positive breast cancer. Preclinical studies suggest the mechanisms of trastuzumab resistance are numerous. Unfortunately, the majority of these studies are based around HER-2-positive (HER-2+) luminal cell lines. The role of epithelial to mesenchymal transition (EMT), a genetic program that confers a basal phenotype, may represent a novel mechanism of escape for HER-2+ luminal cells from trastuzumab treatment. Here we investigated this possibility using a model of clonal selection in HER-2+ luminal breast cancer cells. Following a random isolation and expansion of "colony clusters" from SKBR-3 cell lines, several colony clusters underwent a spontaneous EMT in-vitro. In addition to expression of conventional EMT markers, all mesenchymal colony clusters displayed a predominant CD44+/CD24- phenotype with decreased HER-2 expression and elevated levels of a β1-integrin isoform with a high degree of N-glycosylation. Treatment with a β1-integrin function-blocking antibody, AIIB2, preferentially decreased the N-glycosylated form of β1-integrin, impaired mammosphere formation and restored epithelial phenotype in mesenchymal colony clusters. Using this model we provide the first clear evidence that resistance to trastuzumab (and lapatinib) can occur spontaneously as HER-2+ cells shift from a luminal to a basal/mesenchymal phenotype following EMT. While the major determinant of trastuzumab resistance in mesenchymal colony clusters is likely the down regulation of the HER-2 protein, our evidence suggests that multiple factors may contribute, including expression of N-glycosylated β1-integrin.
Collapse
MESH Headings
- Antibodies, Monoclonal, Humanized/pharmacology
- Antineoplastic Agents/pharmacology
- Blotting, Western
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- CD24 Antigen/metabolism
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Epithelial-Mesenchymal Transition/genetics
- Female
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic/drug effects
- Glycosylation/drug effects
- Humans
- Hyaluronan Receptors/metabolism
- Integrin beta1/genetics
- Integrin beta1/metabolism
- Lapatinib
- Molecular Targeted Therapy/methods
- Quinazolines/pharmacology
- Receptor, ErbB-2/antagonists & inhibitors
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Trastuzumab
Collapse
Affiliation(s)
- David Lesniak
- Department of Experimental Oncology, Cross Cancer Institute and University of Alberta, Edmonton, Alberta, Canada
| | - Siham Sabri
- Department of Radiation Oncology, Research Institute of McGill University Health Center, McGill University, Montréal, Quebec, Canada
| | - Yaoxian Xu
- Department of Radiation Oncology, Research Institute of McGill University Health Center, McGill University, Montréal, Quebec, Canada
| | - Kathryn Graham
- Department of Experimental Oncology, Cross Cancer Institute and University of Alberta, Edmonton, Alberta, Canada
| | - Pravin Bhatnagar
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Mavanur Suresh
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Bassam Abdulkarim
- Department of Radiation Oncology, Research Institute of McGill University Health Center, McGill University, Montréal, Quebec, Canada
- * E-mail:
| |
Collapse
|
10
|
Duggal G, Heindryckx B, Warrier S, O'Leary T, Van der Jeught M, Lierman S, Vossaert L, Deroo T, Deforce D, Chuva de Sousa Lopes SM, De Sutter P. Influence of activin A supplementation during human embryonic stem cell derivation on germ cell differentiation potential. Stem Cells Dev 2013; 22:3141-55. [PMID: 23829223 DOI: 10.1089/scd.2013.0024] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Human embryonic stem cells (hESCs) are more similar to "primed" mouse epiblast stem cells (mEpiSCs). mEpiSCs, which are derived in Activin A, show an increased propensity to form primordial germ cell (PGC)-like cells in response to bone morphogenic protein 4 (BMP4). Hence, we hypothesized that hESCs derived in the presence of Activin A may be more competent in differentiating towards PGC-like cells after supplementation with BMP4 compared to standard hESC lines. We were able to successfully derive two hESC lines in the presence of Activin A, which were pluripotent and showed higher base levels of STELLA and cKIT compared to standard hESC lines derived without Activin A addition. Furthermore, upon differentiation as embryoid bodies in the presence of BMP4, we observed upregulation of VASA at day 7, both at the transcript and protein level compared to standard hESC lines, which appeared to take longer time for PGC specification. Unlike other hESC lines, nuclear pSMAD2/3 presence confirmed that Activin signalling was switched on in Activin A-derived hESC lines. They were also responsive to BMP4 based on nuclear detection of pSMAD1/5/8 and showed endodermal differentiation as a result of GATA-6 expression. Hence, our results provide novel insights into the impact of hESC derivation in the presence of Activin A and its subsequent influence on germ cell differentiation potential in vitro.
Collapse
Affiliation(s)
- Galbha Duggal
- 1 Department for Reproductive Medicine, Ghent University Hospital , Ghent, Belgium
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Crapnell K, Blaesius R, Hastings A, Lennon DP, Caplan AI, Bruder SP. Growth, differentiation capacity, and function of mesenchymal stem cells expanded in serum-free medium developed via combinatorial screening. Exp Cell Res 2013; 319:1409-18. [DOI: 10.1016/j.yexcr.2013.04.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Revised: 04/03/2013] [Accepted: 04/05/2013] [Indexed: 12/15/2022]
|
12
|
Ojala M, Rajala K, Pekkanen-Mattila M, Miettinen M, Huhtala H, Aalto-Setälä K. Culture conditions affect cardiac differentiation potential of human pluripotent stem cells. PLoS One 2012; 7:e48659. [PMID: 23119085 PMCID: PMC3485380 DOI: 10.1371/journal.pone.0048659] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Accepted: 09/28/2012] [Indexed: 12/21/2022] Open
Abstract
Human pluripotent stem cells (hPSCs), including human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs), are capable of differentiating into any cell type in the human body and thus can be used in studies of early human development, as cell models for different diseases and eventually also in regenerative medicine applications. Since the first derivation of hESCs in 1998, a variety of culture conditions have been described for the undifferentiated growth of hPSCs. In this study, we cultured both hESCs and hiPSCs in three different culture conditions: on mouse embryonic fibroblast (MEF) and SNL feeder cell layers together with conventional stem cell culture medium containing knockout serum replacement and basic fibroblast growth factor (bFGF), as well as on a Matrigel matrix in mTeSR1 medium. hPSC lines were subjected to cardiac differentiation in mouse visceral endodermal-like (END-2) co-cultures and the cardiac differentiation efficiency was determined by counting both the beating areas and Troponin T positive cells, as well as studying the expression of OCT-3/4, mesodermal Brachyury T and NKX2.5 and endodermal SOX-17 at various time points during END-2 differentiation by q-RT-PCR analysis. The most efficient cardiac differentiation was observed with hPSCs cultured on MEF or SNL feeder cell layers in stem cell culture medium and the least efficient cardiac differentiation was observed on a Matrigel matrix in mTeSR1 medium. Further, hPSCs cultured on a Matrigel matrix in mTeSR1 medium were found to be more committed to neural lineage than hPSCs cultured on MEF or SNL feeder cell layers. In conclusion, culture conditions have a major impact on the propensity of the hPSCs to differentiate into a cardiac lineage.
Collapse
Affiliation(s)
- Marisa Ojala
- Institute of Biomedical Technology, University of Tampere, Tampere, Finland
- BioMediTech, University of Tampere, Tampere, Finland
| | - Kristiina Rajala
- Institute of Biomedical Technology, University of Tampere, Tampere, Finland
- BioMediTech, University of Tampere, Tampere, Finland
| | - Mari Pekkanen-Mattila
- Institute of Biomedical Technology, University of Tampere, Tampere, Finland
- BioMediTech, University of Tampere, Tampere, Finland
| | - Marinka Miettinen
- Institute of Biomedical Technology, University of Tampere, Tampere, Finland
- BioMediTech, University of Tampere, Tampere, Finland
| | - Heini Huhtala
- School of Health Sciences, University of Tampere, Tampere, Finland
| | - Katriina Aalto-Setälä
- Institute of Biomedical Technology, University of Tampere, Tampere, Finland
- BioMediTech, University of Tampere, Tampere, Finland
- Heart Center, Tampere University Hospital, Tampere, Finland
| |
Collapse
|
13
|
Harvey AJ, Mao S, Lalancette C, Krawetz SA, Brenner CA. Transcriptional differences between rhesus embryonic stem cells generated from in vitro and in vivo derived embryos. PLoS One 2012; 7:e43239. [PMID: 23028448 PMCID: PMC3445581 DOI: 10.1371/journal.pone.0043239] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Accepted: 07/18/2012] [Indexed: 01/16/2023] Open
Abstract
Numerous studies have focused on the transcriptional signatures that underlie the maintenance of embryonic stem cell (ESC) pluripotency. However, it remains unclear whether ESC retain transcriptional aberrations seen in in vitro cultured embryos. Here we report the first global transcriptional profile comparison between ESC generated from either in vitro cultured or in vivo derived primate embryos by microarray analysis. Genes involved in pluripotency, oxygen regulation and the cell cycle were downregulated in rhesus ESC generated from in vitro cultured embryos (in vitro ESC). Significantly, several gene differences are similarly downregulated in preimplantation embryos cultured in vitro, which have been associated with long term developmental consequences and disease predisposition. This data indicates that prior to derivation, embryo quality may influence the molecular signature of ESC lines, and may differentially impact the physiology of cells prior to or following differentiation.
Collapse
Affiliation(s)
- Alexandra J Harvey
- Department of Physiology, Wayne State University, Detroit, Michigan, United States of America.
| | | | | | | | | |
Collapse
|
14
|
Chung TL, Turner JP, Thaker NY, Kolle G, Cooper-White JJ, Grimmond SM, Pera MF, Wolvetang EJ. Ascorbate promotes epigenetic activation of CD30 in human embryonic stem cells. Stem Cells 2011; 28:1782-93. [PMID: 20715184 DOI: 10.1002/stem.500] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Human embryonic stem cells (hESCs) and induced pluripotent stem cells have the ability to adapt to various culture conditions. Phenotypic and epigenetic changes brought about by the culture conditions can, however, have significant impacts on their use in research and in clinical applications. Here, we show that diploid hESCs start to express CD30, a biomarker for malignant cells in Hodgkin's disease and embryonal carcinoma cells, when cultured in knockout serum replacement (KOSR)-based medium, but not in fetal calf serum containing medium. We identify the commonly used medium additive, ascorbate, as the sole medium component in KOSR responsible for CD30 induction. Our data show that this epigenetic activation of CD30 expression in hESCs by ascorbate occurs through a dramatic loss of DNA methylation of a CpG island in the CD30 promoter. Analysis of the phenotype and transcriptome of hESCs that overexpress the CD30 signaling domain reveals that CD30 signaling leads to inhibition of apoptosis, enhanced single-cell growth, and transcriptome changes that are associated with cell signaling, lipid metabolism, and tissue development. Collectively, our data show that hESC culture media that contain ascorbate trigger CD30 expression through an epigenetic mechanism and that this provides a survival advantage and transcriptome changes that may help adapt hESCs to in vitro culture conditions.
Collapse
Affiliation(s)
- Tung-Liang Chung
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland, Australia
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Ström S, Rodriguez-Wallberg K, Holm F, Bergström R, Eklund L, Strömberg AM, Hovatta O. No relationship between embryo morphology and successful derivation of human embryonic stem cell lines. PLoS One 2010; 5:e15329. [PMID: 21217828 PMCID: PMC3013107 DOI: 10.1371/journal.pone.0015329] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2010] [Accepted: 11/08/2010] [Indexed: 11/18/2022] Open
Abstract
Background The large number (30) of permanent human embryonic stem cell (hESC) lines and additional 29 which did not continue growing, in our laboratory at Karolinska Institutet have given us a possibility to analyse the relationship between embryo morphology and the success of derivation of hESC lines. The derivation method has been improved during the period 2002–2009, towards fewer xeno-components. Embryo quality is important as regards the likelihood of pregnancy, but there is little information regarding likelihood of stem cell derivation. Methods We evaluated the relationship of pronuclear zygote stage, the score based on embryo morphology and developmental rate at cleavage state, and the morphology of the blastocyst at the time of donation to stem cell research, to see how they correlated to successful establishment of new hESC lines. Results Derivation of hESC lines succeeded from poor quality and good quality embryos in the same extent. In several blastocysts, no real inner cell mass (ICM) was seen, but permanent well growing hESC lines could be established. One tripronuclear (3PN) zygote, which developed to blastocyst stage, gave origin to a karyotypically normal hESC line. Conclusion Even very poor quality embryos with few cells in the ICM can give origin to hESC lines.
Collapse
Affiliation(s)
- Susanne Ström
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden.
| | | | | | | | | | | | | |
Collapse
|
16
|
Galán A, Montaner D, Póo ME, Valbuena D, Ruiz V, Aguilar C, Dopazo J, Simón C. Functional genomics of 5- to 8-cell stage human embryos by blastomere single-cell cDNA analysis. PLoS One 2010; 5:e13615. [PMID: 21049019 PMCID: PMC2964308 DOI: 10.1371/journal.pone.0013615] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2010] [Accepted: 10/02/2010] [Indexed: 12/26/2022] Open
Abstract
Blastomere fate and embryonic genome activation (EGA) during human embryonic development are unsolved areas of high scientific and clinical interest. Forty-nine blastomeres from 5- to 8-cell human embryos have been investigated following an efficient single-cell cDNA amplification protocol to provide a template for high-density microarray analysis. The previously described markers, characteristic of Inner Cell Mass (ICM) (n = 120), stemness (n = 190) and Trophectoderm (TE) (n = 45), were analyzed, and a housekeeping pattern of 46 genes was established. All the human blastomeres from the 5- to 8-cell stage embryo displayed a common gene expression pattern corresponding to ICM markers (e.g., DDX3, FOXD3, LEFTY1, MYC, NANOG, POU5F1), stemness (e.g., POU5F1, DNMT3B, GABRB3, SOX2, ZFP42, TERT), and TE markers (e.g., GATA6, EOMES, CDX2, LHCGR). The EGA profile was also investigated between the 5-6- and 8-cell stage embryos, and compared to the blastocyst stage. Known genes (n = 92) such as depleted maternal transcripts (e.g., CCNA1, CCNB1, DPPA2) and embryo-specific activation (e.g., POU5F1, CDH1, DPPA4), as well as novel genes, were confirmed. In summary, the global single-cell cDNA amplification microarray analysis of the 5- to 8-cell stage human embryos reveals that blastomere fate is not committed to ICM or TE. Finally, new EGA features in human embryogenesis are presented.
Collapse
Affiliation(s)
- Amparo Galán
- Valencia Node of The National Stem Cell Bank, Centro de Investigación Príncipe Felipe (CIPF), Valencia, Spain
| | - David Montaner
- Department of Bioinformatics and Genomics, Centro de Investigación Príncipe Felipe (CIPF), Valencia, Spain
| | - M. Eugenia Póo
- Valencia Node of The National Stem Cell Bank, Centro de Investigación Príncipe Felipe (CIPF), Valencia, Spain
| | - Diana Valbuena
- Valencia Node of The National Stem Cell Bank, Centro de Investigación Príncipe Felipe (CIPF), Valencia, Spain
| | - Verónica Ruiz
- Valencia Node of The National Stem Cell Bank, Centro de Investigación Príncipe Felipe (CIPF), Valencia, Spain
| | - Cristóbal Aguilar
- Valencia Node of The National Stem Cell Bank, Centro de Investigación Príncipe Felipe (CIPF), Valencia, Spain
| | - Joaquín Dopazo
- Department of Bioinformatics and Genomics, Centro de Investigación Príncipe Felipe (CIPF), Valencia, Spain
| | - Carlos Simón
- Valencia Node of The National Stem Cell Bank, Centro de Investigación Príncipe Felipe (CIPF), Valencia, Spain
- Fundación Instituto Valenciano de Infertilidad (FIVI), Instituto Universitario IVI (IUIVI), University of Valencia, Valencia, Spain
- * E-mail:
| |
Collapse
|
17
|
Schulze HG, Konorov SO, Caron NJ, Piret JM, Blades MW, Turner RFB. Assessing differentiation status of human embryonic stem cells noninvasively using Raman microspectroscopy. Anal Chem 2010; 82:5020-7. [PMID: 20481517 DOI: 10.1021/ac902697q] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Raman microspectroscopy is an attractive approach for chemical imaging of biological specimens, including live cells, without the need for chemi-selective stains. Using a microspectrometer, near-infrared Raman spectra throughout the range 663 cm(-1) to 1220 cm(-1) were obtained from colonies of CA1 human embryonic stem cells (hESCs) and CA1 cells that had been stimulated to differentiate for 3 weeks by 10% fetal bovine serum on gelatin. Distributions and intensities of spectral bands attributed to proteins varied significantly between undifferentiated and differentiated cells. Importantly, compared to proteins and lipids, the band intensities of nucleic acids were dominant in undifferentiated cells with a dominance-reversal in differentiated cells. Thus, we could identify intensity ratios of particular protein-related bands (e.g., 757 cm(-1) tryptophan) to nucleic acid bands (784 cm(-1) DNA/RNA composite) that were effective in discriminating between spectra of undifferentiated and differentiated cells. We observed no discernible negative effects due to the laser exposure in terms of morphology, proliferation, or pluripotency of the stem cells. We conclude that Raman microscopy and complementary data processing procedures provide a rapid, noninvasive approach that can distinguish hESCs from differentiated cells. This is the first report to identify specific Raman markers for the differentiation status of hESCs.
Collapse
Affiliation(s)
- H Georg Schulze
- Michael Smith Laboratories, The University of British Columbia, 2185 East Mall, Vancouver, BC, Canada, V6T 1Z4
| | | | | | | | | | | |
Collapse
|
18
|
Kitajima H, Niwa H. Clonal expansion of human pluripotent stem cells on gelatin-coated surface. Biochem Biophys Res Commun 2010; 396:933-8. [DOI: 10.1016/j.bbrc.2010.05.026] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2010] [Accepted: 05/06/2010] [Indexed: 11/28/2022]
|
19
|
Niemann H, Carnwath JW, Herrmann D, Wieczorek G, Lemme E, Lucas-Hahn A, Olek S. DNA methylation patterns reflect epigenetic reprogramming in bovine embryos. Cell Reprogram 2010; 12:33-42. [PMID: 20132011 DOI: 10.1089/cell.2009.0063] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
To understand the epigenetic alterations associated with assisted reproduction technology (ART) and the reprogramming of gene expression that follows somatic cell nuclear transfer (SCNT), we screened a panel of 41 amplicons representing 25 developmentally important genes on 15 different chromosomes (a total of 1079 CpG sites). Methylation analysis was performed on DNA from pools of 80 blastocysts representing three classes of embryos. This revealed a subset of amplicons that distinguish between embryos developing in vivo, produced in vitro, or reconstructed by SCNT. Following SCNT, we observed massive epigenetic reprogramming evidenced by reduced levels of methylation in the resultant embryos. Analysis of data from the 28 most informative amplicons (hotspot loci), representing more than 523 individual CpG sites, we discovered subsets of amplicons with methylation patterns that were unique to each class of embryo and may indicate metastable epialleles. Analysis of eight genes with respect to mRNA expression did not reveal a direct correlation with DNA methylation levels. In conclusion, this approach revealed a subset of amplicons that can be used to evaluate blastocyst quality and reprogramming following SCNT, and can also be employed for the localization of the epigenetic control regions within individual genes and for more general studies of stem cell differentiation.
Collapse
Affiliation(s)
- Heiner Niemann
- Institute of Farm Animal Genetics (FLI), Mariensee, Neustadt, Germany.
| | | | | | | | | | | | | |
Collapse
|
20
|
Fernandes AM, Meletti T, Guimarães R, Stelling MP, Marinho PAN, Valladão AS, Rehen SK. Worldwide Survey of Published Procedures to Culture Human Embryonic Stem Cells. Cell Transplant 2010; 19:509-23. [DOI: 10.3727/096368909x485067] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Since their derivation 11 years ago, human embryonic stem (hES) cells have become a powerful tool in both basic biomedical research and developmental biology. Their capacity for self-renewal and differentiation into any tissue type has also brought interest from fields such as cell therapy and drug screening. We conducted an extensive analysis of 750 papers (51% of the total published about hES cells between 1998 and 2008) to present a spectrum of hES cell research including culture protocols developed worldwide. This review may stimulate discussions about the importance of having unvarying methods to culture hES cells, in order to facilitate comparisons among data obtained by research groups elsewhere, especially concerning preclinical studies. Moreover, the description of the most widely used cell lines, reagents, and procedures adopted internationally will help newcomers on deciding the best strategies for starting their own studies. Finally, the results will contribute with the efforts of stem cell researchers on comparing the performance of different aspects related to hES cell culture methods.
Collapse
Affiliation(s)
- A. M. Fernandes
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - T. Meletti
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - R. Guimarães
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - M. P. Stelling
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - P. A. N. Marinho
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Programa de Engenharia Química/COPPE, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - A. S. Valladão
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - S. K. Rehen
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
21
|
Mahlstedt MM, Anderson D, Sharp JS, McGilvray R, Muñoz MDB, Buttery LD, Alexander MR, Rose FRAJ, Denning C. Maintenance of pluripotency in human embryonic stem cells cultured on a synthetic substrate in conditioned medium. Biotechnol Bioeng 2010; 105:130-40. [PMID: 19718698 DOI: 10.1002/bit.22520] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Realizing the potential clinical and industrial applications of human embryonic stem cells (hESCs) is limited by the need for costly, labile, or undefined growth substrates. Here we demonstrate that trypsin passaging of the hESC lines, HUES7 and NOTT1, on oxygen plasma etched tissue culture polystyrene (PE-TCPS) in conditioned medium is compatible with pluripotency. This synthetic culture surface is stable at room temperature for at least a year and is readily prepared by placing polystyrene substrates in a radio frequency oxygen plasma generator for 5 min. Modification of the polystyrene surface chemistry by plasma etching was confirmed by X-ray photoelectron spectroscopy (XPS) and time-of-flight secondary ion mass spectrometry (ToF-SIMS), which identified elemental and molecular changes as a result of the treatment. Pluripotency of hESCs cultured on PE-TCPS was gauged by consistent proliferation during serial passage, expression of stem cell markers (OCT4, TRA1-60, and SSEA-4), stable karyotype and multi-germlayer differentiation in vitro, including to pharmacologically responsive cardiomyocytes. Generation of cost-effective, easy-to-handle synthetic, defined, stable surfaces for hESC culture will expedite stem cell use in biomedical applications.
Collapse
|
22
|
Shaw ML, Williams EJ, Hawes S, Saffery R. Characterisation of histone variant distribution in human embryonic stem cells by transfection of in vitro transcribed mRNA. Mol Reprod Dev 2010; 76:1128-42. [PMID: 19606468 DOI: 10.1002/mrd.21077] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Recent studies, primarily in mouse embryonic stem cells, have highlighted the unique chromatin state of pluripotent stem cells, including the incorporation of histone variants into specific genomic locations, and its role in facilitating faithful expression of genes during development. However, there is little information available on the expression and subcellular localisation of histone variants in human embryonic stem cells (hESCs). In this study, we confirmed the expression of a panel of histone variant genes in several hESC lines and demonstrated the utility of transfection of in vitro transcribed, epitope-tagged mRNAs to characterise the subcellular localisation of these proteins. The subcellular localisations of variant histone H3 (CENP-A, H3.3), H2A (MACROH2A, H2AX, H2AZ, H2ABBD) and H1 (H1A, HB, H1C, H1D) were examined, revealing distinct nuclear localisation profiles for each protein. These data highlight the differences between murine (m) ESCs and hESCs, including the presence of a MACROH2A-enriched inactive X chromosome in undifferentiated XX hESC lines. We also provide the first evidence for MACROH2A accumulation on the Y-chromosome in XY hESCs.
Collapse
Affiliation(s)
- Margaret L Shaw
- Developmental Epigenetics, Department of Paediatrics, Murdoch Childrens Research Institute, Royal Children's Hospital, University of Melbourne, Parkville, Melbourne, Victoria, Australia
| | | | | | | |
Collapse
|
23
|
Peiffer I, Barbet R, Hatzfeld A, Li ML, Hatzfeld JA. Optimization of physiological xenofree molecularly defined media and matrices to maintain human embryonic stem cell pluripotency. Methods Mol Biol 2010; 584:97-108. [PMID: 19907973 DOI: 10.1007/978-1-60761-369-5_5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
We describe in this chapter the development of a xenofree molecularly defined medium, SBX, associated with xenofree matrices, to maintain human embryonic stem cell (hESC) pluripotency as determined by phenotypic, functional and TLDA studies. This simple, inexpensive, and more physiological culture condition has been chosen because (1) it is xenofree and molecularly defined; it is devoid of albumin, which is a carrier of undefined molecules; (2) it maintains pluripotency, but very significantly reduces differentiation gene expression during hESC self-renewal, as compared to the widely used culture conditions tested so far; and (3) it can be further improved by replacing high concentrations of expensive additives by physiological concentrations of new factors. Xenofree molecularly defined media and matrices represent valuable tools for elucidating still unknown functions of numerous embryonic genes using more physiological culture conditions. These genes encode potential new factors controlling hESC self-renewal and pluripotency.
Collapse
Affiliation(s)
- Isabelle Peiffer
- Centre National de la Recherche Scientifique, Institut André Lwoff, Villejuif, France
| | | | | | | | | |
Collapse
|
24
|
Aranda P, Agirre X, Ballestar E, Andreu EJ, Román-Gómez J, Prieto I, Martín-Subero JI, Cigudosa JC, Siebert R, Esteller M, Prosper F. Epigenetic signatures associated with different levels of differentiation potential in human stem cells. PLoS One 2009; 4:e7809. [PMID: 19915669 PMCID: PMC2771914 DOI: 10.1371/journal.pone.0007809] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2009] [Accepted: 10/14/2009] [Indexed: 01/01/2023] Open
Abstract
Background The therapeutic use of multipotent stem cells depends on their differentiation potential, which has been shown to be variable for different populations. These differences are likely to be the result of key changes in their epigenetic profiles. Methodology/Principal Findings to address this issue, we have investigated the levels of epigenetic regulation in well characterized populations of pluripotent embryonic stem cells (ESC) and multipotent adult stem cells (ASC) at the trancriptome, methylome, histone modification and microRNA levels. Differences in gene expression profiles allowed classification of stem cells into three separate populations including ESC, multipotent adult progenitor cells (MAPC) and mesenchymal stromal cells (MSC). The analysis of the PcG repressive marks, histone modifications and gene promoter methylation of differentiation and pluripotency genes demonstrated that stem cell populations with a wider differentiation potential (ESC and MAPC) showed stronger representation of epigenetic repressive marks in differentiation genes and that this epigenetic signature was progressively lost with restriction of stem cell potential. Our analysis of microRNA established specific microRNA signatures suggesting specific microRNAs involved in regulation of pluripotent and differentiation genes. Conclusions/Significance Our study leads us to propose a model where the level of epigenetic regulation, as a combination of DNA methylation and histone modification marks, at differentiation genes defines degrees of differentiation potential from progenitor and multipotent stem cells to pluripotent stem cells.
Collapse
Affiliation(s)
- Pablo Aranda
- Hematology Department and Area of Cell Therapy, Clínica Universidad de Navarra, Foundation for Applied Medical Research, University of Navarra, Pamplona, Spain
| | - Xabier Agirre
- Hematology Department and Area of Cell Therapy, Clínica Universidad de Navarra, Foundation for Applied Medical Research, University of Navarra, Pamplona, Spain
| | - Esteban Ballestar
- Cancer Epigenetics and Biology Program (PEBC), The Bellvitge Institute for Biomedical Research (IDIBELL-ICO), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Enrique J. Andreu
- Hematology Department and Area of Cell Therapy, Clínica Universidad de Navarra, Foundation for Applied Medical Research, University of Navarra, Pamplona, Spain
| | | | - Inés Prieto
- Hematology Department and Area of Cell Therapy, Clínica Universidad de Navarra, Foundation for Applied Medical Research, University of Navarra, Pamplona, Spain
| | - José Ignacio Martín-Subero
- Institute of Human Genetics, University Hospital Schleswig-Holstein Campus Kiel/Christian-Albrechts University, Kiel, Germany
| | - Juan Cruz Cigudosa
- Molecular Cytogenetics Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Reiner Siebert
- Institute of Human Genetics, University Hospital Schleswig-Holstein Campus Kiel/Christian-Albrechts University, Kiel, Germany
| | - Manel Esteller
- Cancer Epigenetics and Biology Program (PEBC), The Bellvitge Institute for Biomedical Research (IDIBELL-ICO), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Felipe Prosper
- Hematology Department and Area of Cell Therapy, Clínica Universidad de Navarra, Foundation for Applied Medical Research, University of Navarra, Pamplona, Spain
- * E-mail:
| |
Collapse
|
25
|
Pal R, Totey S, Mamidi MK, Bhat VS, Totey S. Propensity of human embryonic stem cell lines during early stage of lineage specification controls their terminal differentiation into mature cell types. Exp Biol Med (Maywood) 2009; 234:1230-43. [PMID: 19546356 DOI: 10.3181/0901-rm-38] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Human embryonic stem cells (hESCs) are able to stably maintain their characteristics for an unlimited period; nevertheless, substantial differences among cell lines in gene and protein expression not manifested during the undifferentiated state may appear when cells differentiate. It is widely accepted that developing an efficient protocol to control the differentiation of hESCs will enable us to produce adequate numbers of desired cell types with relative ease for diverse applications ranging from basic research to cell therapy and drug screening. Hence of late, there has been considerable interest in understanding whether and how hESC lines are equivalent or different to each other in their in vitro developmental tendencies. In this study, we compared the developmental competences of two hESC lines (HUES-9 and HUES-7) at molecular, cellular and functional levels, upon spontaneous differentiation without any added inducing agents. Both cell lines generated the three embryonic germ layers, extra-embryonic tissues and primordial germ cells during embryoid body (EB) formation. However HUES-9 showed a stronger propensity towards formation of neuroectodermal lineages, whereas HUES-7 differentiated preferentially into mesoderm and endoderm. Upon further differentiation, HUES-9 generated largely neural cells (neurons, oligodendrocytes, astrocytes and gangliosides) whereas HUES-7 formed mesendodermal derivatives, including cardiomyocytes, skeletal myocytes, endothelial cells, hepatocytes and pancreatic cells. Overall, our findings endorse the hypothesis that independently-derived hESCs biologically differ among themselves, thereby displaying varying differentiation propensity. These subtle differences not only highlight the importance of screening and deriving lines for lineage-specific differentiation but also indicate that individual lines may possess a repertoire of capabilities that is unique.
Collapse
Affiliation(s)
- Rajarshi Pal
- Manipal Institute of Regenerative Medicine, Manipal University Branch Campus, Bangalore 560071, India
| | | | | | | | | |
Collapse
|
26
|
Kumar N, Hinduja I, Nagvenkar P, Pillai L, Zaveri K, Mukadam L, Telang J, Desai S, Mangoli V, Mangoli R, Padgaonkar S, Kaur G, Puri C, Bhartiya D. Derivation and Characterization of Two Genetically Unique Human Embryonic Stem Cell Lines on In-House–Derived Human Feeders. Stem Cells Dev 2009; 18:435-45. [DOI: 10.1089/scd.2008.0234] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Neeraj Kumar
- Stem Cell Biology Department, National Institute for Research in Reproductive Health, Mumbai, India
| | | | - Punam Nagvenkar
- Stem Cell Biology Department, National Institute for Research in Reproductive Health, Mumbai, India
| | - Lakshmi Pillai
- Stem Cell Biology Department, National Institute for Research in Reproductive Health, Mumbai, India
| | | | | | - Jyoti Telang
- Stem Cell Biology Department, National Institute for Research in Reproductive Health, Mumbai, India
| | | | | | | | | | - Gurvinder Kaur
- Department of Transplant Immunology and Immunogenetics, All India Institute of Medical Sciences, New Delhi, India
| | - Chander Puri
- Stem Cell Biology Department, National Institute for Research in Reproductive Health, Mumbai, India
| | - Deepa Bhartiya
- Stem Cell Biology Department, National Institute for Research in Reproductive Health, Mumbai, India
| |
Collapse
|
27
|
Chen HF, Chuang CY, Shieh YK, Chang HW, Ho HN, Kuo HC. Novel autogenic feeders derived from human embryonic stem cells (hESCs) support an undifferentiated status of hESCs in xeno-free culture conditions. Hum Reprod 2009; 24:1114-25. [DOI: 10.1093/humrep/dep003] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
28
|
Schneider A, Spitkovsky D, Riess P, Molcanyi M, Kamisetti N, Maegele M, Hescheler J, Schaefer U. "The good into the pot, the bad into the crop!"--a new technology to free stem cells from feeder cells. PLoS One 2008; 3:e3788. [PMID: 19023443 PMCID: PMC2582950 DOI: 10.1371/journal.pone.0003788] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2008] [Accepted: 10/22/2008] [Indexed: 01/11/2023] Open
Abstract
A variety of embryonic and adult stem cell lines require an initial co-culturing with feeder cells for non-differentiated growth, self renewal and maintenance of pluripotency. However for many downstream ES cell applications the feeder cells have to be considered contaminations that might interfere not just with the analysis of experimental data but also with clinical application and tissue engineering approaches. Here we introduce a novel technique that allows for the selection of pure feeder-freed stem cells, following stem cell proliferation on feeder cell layers. Complete and reproducible separation of feeder and embryonic stem cells was accomplished by adaptation of an automated cell selection system that resulted in the aspiration of distinct cell colonies or fraction of colonies according to predefined physical parameters. Analyzing neuronal differentiation we demonstrated feeder-freed stem cells to exhibit differentiation potentials comparable to embryonic stem cells differentiated under standard conditions. However, embryoid body growth as well as differentiation of stem cells into cardiomyocytes was significantly enhanced in feeder-freed cells, indicating a feeder cell dependent modulation of lineage differentiation during early embryoid body development. These findings underline the necessity to separate stem and feeder cells before the initiation of in vitro differentiation. The complete separation of stem and feeder cells by this new technology results in pure stem cell populations for translational approaches. Furthermore, a more detailed analysis of the effect of feeder cells on stem cell differentiation is now possible, that might facilitate the identification and development of new optimized human or genetically modified feeder cell lines.
Collapse
Affiliation(s)
- Annette Schneider
- Institute for Research in Operative Medicine (IFOM), Faculty of Medicine, University of Witten/Herdecke, Witten, Germany
| | - Dimitry Spitkovsky
- Center of Physiology, Institute of Neurophysiology, Medical Faculty, University of Cologne, Cologne, Germany
| | - Peter Riess
- Department of Trauma and Orthopedic Surgery, Faculty of Medicine, University of Witten-Herdecke at the Hospital Merheim, Cologne, Germany
| | - Marek Molcanyi
- Clinic of Neurosurgery and 2nd Department of Neurophysiology, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Naidu Kamisetti
- Center of Physiology, Institute of Neurophysiology, Medical Faculty, University of Cologne, Cologne, Germany
| | - Marc Maegele
- Department of Trauma and Orthopedic Surgery, Faculty of Medicine, University of Witten-Herdecke at the Hospital Merheim, Cologne, Germany
| | - Jürgen Hescheler
- Center of Physiology, Institute of Neurophysiology, Medical Faculty, University of Cologne, Cologne, Germany
| | - Ute Schaefer
- Department of Experimental Neurotraumatology, Medical University Graz, Graz, Austria
- * E-mail:
| |
Collapse
|
29
|
Ye X, Lotan R. Potential misinterpretation of data on differential gene expression in normal and malignant cells in vitro. BRIEFINGS IN FUNCTIONAL GENOMICS AND PROTEOMICS 2008; 7:322-6. [DOI: 10.1093/bfgp/eln021] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
30
|
Abbott DE, Bailey CM, Postovit LM, Seftor EA, Margaryan N, Seftor REB, Hendrix MJC. The epigenetic influence of tumor and embryonic microenvironments: how different are they? CANCER MICROENVIRONMENT 2008; 1:13-21. [PMID: 19308681 PMCID: PMC2654360 DOI: 10.1007/s12307-008-0004-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2007] [Accepted: 01/22/2008] [Indexed: 12/13/2022]
Abstract
The microenvironment is being increasingly recognized as a critical component in tumor progression and metastases. As such, the bi-directional signaling of extracellular mediators that promote tumor growth within the microenvironment is a focus of intense scrutiny. Interestingly, there are striking similarities between the phenotypes of aggressive tumor and embryonic stem cells, particularly with respect to specific signaling pathways underlying their intriguing plasticity. Here, we demonstrate the epigenetic influence of the hESC microenvironment on the reprogramming of aggressive melanoma cells using an innovative 3-D model. Specifically, our laboratory has previously demonstrated the redifferentiation of these melanoma cells to a more melanocyte-like phenotype (Postovit et al., Stem Cells 24(3):501–505, 2006), and now we show the loss of VE-Cadherin expression (indicative of a plastic vasculogenic phenotype) and the loss of Nodal expression (a plasticity stem cell marker) in tumor cells exposed to the hESC microenvironment. Further studies with the 3-D culture model revealed the epigenetic influence of aggressive melanoma cells on hESCs resulting in the down-regulation of plasticity markers and the emergence of phenotype-specific genes. Additional studies with the aggressive melanoma conditioned matrix microenvironment demonstrated the transdifferentiation of normal melanocytes into melanoma-like cells exhibiting a vasculogenic phenotype. Collectively, these studies have advanced our understanding of the epigenetic influence associated with the microenvironments of hESCs and aggressive melanoma cells, and shed new light on their therapeutic implications. Moreover, we have a better appreciation of the convergence of embryonic and tumorigenic signaling pathways that might stimulate further consideration of targeting Nodal in aggressive tumor cells resulting in a down-regulation of tumorigenic potential and plasticity.
Collapse
Affiliation(s)
- Daniel E. Abbott
- Children’s Memorial Research Center, Department of Surgery, Northwestern University/Feinberg School of Medicine, Chicago, IL 60614 USA
| | - Caleb M. Bailey
- Cancer Biology and Epigenomics Program, Children’s Memorial Research Center, Chicago, IL 60614 USA
| | - Lynne-Marie Postovit
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, Medical Sciences Building, Rm. 438, London, ON N6A 5C1 Canada
| | - Elisabeth A. Seftor
- Cancer Biology and Epigenomics Program, Children’s Memorial Research Center, Chicago, IL 60614 USA
| | - Naira Margaryan
- Cancer Biology and Epigenomics Program, Children’s Memorial Research Center, Chicago, IL 60614 USA
| | - Richard E. B. Seftor
- Cancer Biology and Epigenomics Program, Children’s Memorial Research Center, Robert H. Lurie Comprehensive Cancer Center, Northwestern University/Feinberg School of Medicine, 2430 North Halsted Street, P.O. Box 222, Chicago, IL 60614 USA
| | - Mary J. C. Hendrix
- Cancer Biology and Epigenomics Program, Children’s Memorial Research Center, Robert H. Lurie Comprehensive Cancer Center, Northwestern University/Feinberg School of Medicine, 2430 North Halsted Street, P.O. Box 222, Chicago, IL 60614 USA
| |
Collapse
|
31
|
Derivation, maintenance and cryostorage of human embryonic stem cells. DRUG DISCOVERY TODAY. TECHNOLOGIES 2008; 5:e105-48. [PMID: 24125545 DOI: 10.1016/j.ddtec.2010.04.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
32
|
Pannetier M, Feil R. Epigenetic stability of embryonic stem cells and developmental potential. Trends Biotechnol 2007; 25:556-62. [PMID: 17983676 DOI: 10.1016/j.tibtech.2007.09.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2007] [Revised: 09/10/2007] [Accepted: 09/10/2007] [Indexed: 11/25/2022]
Abstract
Recent studies highlight the tremendous potential of human embryonic stem (ES) cells and their derivatives as therapeutic tools for degenerative diseases. However, derivation and culture of ES cells can induce epigenetic alterations, which can have long lasting effects on gene expression and phenotype. Research on human and mouse stem cells indicates that developmental, cancer-related genes, and genes regulated by genomic imprinting are particularly susceptible to changes in DNA methylation. Together with the occurrence of genetic alterations, epigenetic instability needs to be monitored when considering human stem cells for therapeutic and technological purposes. Here, we discuss the maintenance of epigenetic information in cultured stem cells and embryos and how this influences their developmental potential.
Collapse
Affiliation(s)
- Maëlle Pannetier
- Institute of Molecular Genetics, CNRS, 1919, route de Mende, 34293 Montpellier, France
| | | |
Collapse
|
33
|
Tan SM, Wang ST, Hentze H, Dröge P. A UTF1-based selection system for stable homogeneously pluripotent human embryonic stem cell cultures. Nucleic Acids Res 2007; 35:e118. [PMID: 17855398 PMCID: PMC2094078 DOI: 10.1093/nar/gkm704] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Undifferentiated transcription factor 1 (UTF1) was identified first in mouse embryonic stem cells and is also expressed in human embryonic and adult stem cells. UTF1 transcription ceases at the onset of differentiation, which clearly distinguishes it from less sensitive pluripotency markers, such as Oct4 or Nanog. We present here two transgenic hESC lines, named ZUN. Each line harbors one copy of the UTF1 promoter/enhancer driving a resistance gene and yielded highly homogeneous cultures under selection pressure, with a larger proportion of Oct4 and Sox2 positive cells. While ZUN cultures, like parental HUES8 cultures, retained the capacity to differentiate into tissues of all three germ layers using a SICD mouse teratoma model, they surprisingly exhibited an increased refractoriness to various differentiation cues in vitro. Together with its small size of only 2.4 kb for the entire cassette, these features render our selection system a powerful novel tool for many stem cell applications and human somatic cell reprogramming strategies.
Collapse
Affiliation(s)
- Shen Mynn Tan
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, 637551 and ES Cell International Pte Ltd, 11 Biopolis Way, #05-06 Helios, 138667 Singapore
| | - Siew Tein Wang
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, 637551 and ES Cell International Pte Ltd, 11 Biopolis Way, #05-06 Helios, 138667 Singapore
| | - Hannes Hentze
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, 637551 and ES Cell International Pte Ltd, 11 Biopolis Way, #05-06 Helios, 138667 Singapore
| | - Peter Dröge
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, 637551 and ES Cell International Pte Ltd, 11 Biopolis Way, #05-06 Helios, 138667 Singapore
- *To whom correspondence should be addressed. +65 6316 2809+65 6791 3856 or
| |
Collapse
|
34
|
Chase LG, Firpo MT. Development of serum-free culture systems for human embryonic stem cells. Curr Opin Chem Biol 2007; 11:367-72. [PMID: 17692558 DOI: 10.1016/j.cbpa.2007.06.421] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2007] [Revised: 06/18/2007] [Accepted: 06/19/2007] [Indexed: 01/08/2023]
Abstract
Human embryonic stem cells, because of their unique combination of long-term self-renewal properties and pluripotency, are providing new avenues of investigation of stem cell biology and human development and show promise in providing a new source of human cells for transplantation therapies and pharmaceutical testing. Current methods of propagating these cells using combinations of mouse fibroblast feeder cultures and bovine serum components are inexpensive and, in general, useful. However, the systematic investigation of the regulation of self-renewal and the production of safer sources of cells for transplantation depends on the elimination of animal products and the use of defined culture conditions. Both goals are served by the development of serum-free culture methods for human embryonic stem cells.
Collapse
Affiliation(s)
- Lucas G Chase
- Department of Medicine, Division of Endocrinology and Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, United States
| | | |
Collapse
|
35
|
Mannello F, Tonti GA. Concise Review: No Breakthroughs for Human Mesenchymal and Embryonic Stem Cell Culture: Conditioned Medium, Feeder Layer, or Feeder-Free; Medium with Fetal Calf Serum, Human Serum, or Enriched Plasma; Serum-Free, Serum Replacement Nonconditioned Medium, or Ad Hoc Formula? All That Glitters Is Not Gold! Stem Cells 2007; 25:1603-9. [PMID: 17395775 DOI: 10.1634/stemcells.2007-0127] [Citation(s) in RCA: 233] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The choice of an optimal strategy of stem cell culture is at the moment an impossible task, and the elaboration of a culture medium adapted to the production of embryonic and adult mesenchymal stem cells for the clinical application of cell therapy remains a crucial matter. To make an informed choice, it is crucial to not underestimate the theoretical health risk of using xenogenic compounds, to limit the immunological reactions once stem cells are transplanted, to not overestimate the controversial results obtained with human serum, plasma, and blood derivatives, as well as to carefully examine the pros and cons of serum-free and ad hoc formulation strategies; besides that, to also maintain multipotentiality, self-renewal, and transplantability. The extent to which we are able to achieve effective cell therapies will depend on assimilating a rapidly developing base of scientific knowledge with the practical considerations of design, delivery, and host response. Although clinical studies have already started, many questions remain unsolved, and concomitantly even more evidence on suitable and safe off-the-shelf products (mainly xeno-free) for embryonic and mesenchymal stem cells is cropping up, even though there should be no rush to enter the clinical stage while the underlying basic research is still not so solid; this solely will lead to high-quality translational research, without making blunders stemming from the assumption that all that glitters is not gold. Disclosure of potential conflicts of interest is found at the end of this article.
Collapse
Affiliation(s)
- Ferdinando Mannello
- Institute of Histology and Laboratory Analysis, Faculty of Sciences and Technologies, University of Urbino Carlo Bo, Via O. Ubaldini 7, 61029 Urbino (PU), Italy.
| | | |
Collapse
|
36
|
Ferreira LS, Gerecht S, Shieh HF, Watson N, Rupnick MA, Dallabrida SM, Vunjak-Novakovic G, Langer R. Vascular progenitor cells isolated from human embryonic stem cells give rise to endothelial and smooth muscle like cells and form vascular networks in vivo. Circ Res 2007; 101:286-94. [PMID: 17569886 DOI: 10.1161/circresaha.107.150201] [Citation(s) in RCA: 174] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We report that human embryonic stem cells contain a population of vascular progenitor cells that have the ability to differentiate into endothelial-like and smooth muscle (SM)-like cells. Vascular progenitor cells were isolated from EBs grown in suspension for 10 days and were characterized by expression of the endothelial/hematopoietic marker CD34 (CD34+ cells). When these cells are subsequently cultured in EGM-2 (endothelial growth medium) supplemented with vascular endothelial growth factor-165 (50 ng/mL), they give rise to endothelial-like cells characterized by a cobblestone cell morphology, expression of endothelial markers (platelet endothelial cell-adhesion molecule-1, CD34, KDR/Flk-1, vascular endothelial cadherin, von Willebrand factor), incorporation of acetylated low-density lipoprotein, and formation of capillary-like structures when placed in Matrigel. In contrast, when CD34+ cells are cultured in EGM-2 supplemented with platelet-derived growth factor-BB (50 ng/mL), they give rise to SM-like cells characterized by spindle-shape morphology, expression of SM cell markers (alpha-SM actin, SM myosin heavy chain, calponin, caldesmon, SM alpha-22), and the ability to contract and relax in response to common pharmacological agents such as carbachol and atropine but rarely form capillary-like structures when placed in Matrigel. Implantation studies in nude mice show that both cell types contribute to the formation of human microvasculature. Some microvessels contained mouse blood cells, which indicates functional integration with host vasculature. Therefore, the vascular progenitors isolated from human embryonic stem cells using methods established in the present study could provide a means to examine the mechanisms of endothelial and SM cell development, and they could also provide a potential source of cells for vascular tissue engineering.
Collapse
MESH Headings
- Animals
- Antigens, CD34/biosynthesis
- Becaplermin
- Biomarkers
- Blood Vessels/cytology
- Blood Vessels/ultrastructure
- Cattle
- Cell Differentiation/drug effects
- Cell Lineage
- Cells, Cultured/cytology
- Cells, Cultured/drug effects
- Cells, Cultured/metabolism
- Cells, Cultured/transplantation
- Collagen
- Culture Media/pharmacology
- Drug Combinations
- Embryonic Stem Cells/cytology
- Embryonic Stem Cells/drug effects
- Embryonic Stem Cells/metabolism
- Embryonic Stem Cells/transplantation
- Endothelial Cells/cytology
- Fetal Blood
- Gene Expression Profiling
- Gene Expression Regulation, Developmental
- Humans
- Injections, Subcutaneous
- Laminin
- Mice
- Mice, Nude
- Morphogenesis
- Muscle Proteins/biosynthesis
- Myocytes, Smooth Muscle/cytology
- Neovascularization, Physiologic
- Organ Specificity
- Platelet-Derived Growth Factor/pharmacology
- Proteoglycans
- Proto-Oncogene Proteins c-sis
- Tissue Engineering
- Vascular Endothelial Growth Factor A/pharmacology
Collapse
Affiliation(s)
- Lino S Ferreira
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Buhr N, Carapito C, Schaeffer C, Hovasse A, Van Dorsselaer A, Viville S. Proteome analysis of the culture environment supporting undifferentiated mouse embryonic stem and germ cell growth. Electrophoresis 2007; 28:1615-23. [PMID: 17436335 DOI: 10.1002/elps.200600497] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The therapeutical interest of pluripotent cells and ethical issues related to the establishment of human embryonic stem cell (ESC) or embryonic germ cell (EGC) lines raise the understanding of the mechanism underlying pluripotency to a fundamental issue. Establishing a protein pluripotency signature for these cells can be complicated by the presence of unrelated proteins produced by the culture environment. Here, we have analyzed the environment supporting ESC and EGC growth, and established 2-D reference maps for each constituent present in this culture environment: mouse embryonic fibroblast feeder cells, culture medium (CM) and gelatin. The establishment of these reference maps is essential prior to the study of ESC and EGC specific proteomes. Indeed, these maps can be subtracted from ESC or EGC maps to allow focusing on spots specific for ESCs or EGCs. Our study led to the identification of 110 unique proteins from fibroblast feeder cells and 23 unique proteins from the CM, which represent major contaminants of ESC and EGC proteomes. For gelatin, no collagen-specific proteins were identified, most likely due to difficulties in resolution and low quantities. Furthermore, no differences were observed between naive and conditioned CM. Finally, we compared these reference maps to ESC 2-D gels and isolated 17 ESC specific spots. Among these spots, proteins that had already been identified in previous human and mouse ESC proteomes were identified but no apparent ESC-specific pluripotency marker could be identified. This work represents an essential step in furthering the knowledge of environmental factors supporting ESC and EGC growth.
Collapse
Affiliation(s)
- Nicolas Buhr
- IGBMC (Institute of Genetics and Molecular and Cellular Biology), Département de Biologie du Dévelopment, Illkirch, France
| | | | | | | | | | | |
Collapse
|
38
|
Allegrucci C, Wu YZ, Thurston A, Denning CN, Priddle H, Mummery CL, Ward-van Oostwaard D, Andrews PW, Stojkovic M, Smith N, Parkin T, Jones ME, Warren G, Yu L, Brena RM, Plass C, Young LE. Restriction landmark genome scanning identifies culture-induced DNA methylation instability in the human embryonic stem cell epigenome. Hum Mol Genet 2007; 16:1253-68. [PMID: 17409196 DOI: 10.1093/hmg/ddm074] [Citation(s) in RCA: 134] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Widespread provision of human embryonic stem cells (hESCs) for therapeutic use, drug screening and disease modelling will require cell lines sustainable over long periods in culture. Since the short-term, in vitro culture of mammalian embryos can result in DNA methylation changes, the epigenetic stability of hESCs warrants investigation. Existing hESC lines have been derived and cultured under diverse conditions, providing the potential for programming differential changes into the epigenome that may result in inter-line variability over and above that inherited from the embryo. By examining the DNA methylation profiles of > 2000 genomic loci by Restriction Landmark Genome Scanning, we identified substantial inter-line epigenetic distance between six independently derived hESC lines. Lines were found to inherit further epigenetic changes over time in culture, with most changes arising in the earliest stages post-derivation. The loci affected varied between lines. The majority of culture-induced changes (82.3-87.5%) were stably inherited both within the undifferentiated cells and post-differentiation. Adapting a line to a serum-free culture system resulted in additional epigenetic instability. Overall 80.5% of the unstable loci uncovered in hESCs have been associated previously with an adult tumour phenotype. Our study shows that current methods of hESC propagation can rapidly programme stable and unpredictable epigenetic changes in the stem cell genome. This highlights the need for (i) novel screening strategies to determine the experimental utility and biosafety of hESCs and (ii) optimization and standardization of procedures for the derivation and culture of hESC lines that minimize culture-induced instability.
Collapse
Affiliation(s)
- Cinzia Allegrucci
- Division of Obstetrics and Gynaecology and Wolfson Centre for Stem Cells, Tissue Engineering and Modelling (STEM), University of Nottingham, Queens Medical Centre, Nottingham NG7 2UH, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Assou S, Le Carrour T, Tondeur S, Ström S, Gabelle A, Marty S, Nadal L, Pantesco V, Réme T, Hugnot JP, Gasca S, Hovatta O, Hamamah S, Klein B, De Vos J. A meta-analysis of human embryonic stem cells transcriptome integrated into a web-based expression atlas. Stem Cells 2007; 25:961-73. [PMID: 17204602 PMCID: PMC1906587 DOI: 10.1634/stemcells.2006-0352] [Citation(s) in RCA: 269] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Microarray technology provides a unique opportunity to examine gene expression patterns in human embryonic stem cells (hESCs). We performed a meta-analysis of 38 original studies reporting on the transcriptome of hESCs. We determined that 1,076 genes were found to be overexpressed in hESCs by at least three studies when compared to differentiated cell types, thus composing a "consensus hESC gene list." Only one gene was reported by all studies: the homeodomain transcription factor POU5F1/OCT3/4. The list comprised other genes critical for pluripotency such as the transcription factors NANOG and SOX2, and the growth factors TDGF1/CRIPTO and Galanin. We show that CD24 and SEMA6A, two cell surface protein-coding genes from the top of the consensus hESC gene list, display a strong and specific membrane protein expression on hESCs. Moreover, CD24 labeling permits the purification by flow cytometry of hESCs cocultured on human fibroblasts. The consensus hESC gene list also included the FZD7 WNT receptor, the G protein-coupled receptor GPR19, and the HELLS helicase, which could play an important role in hESCs biology. Conversely, we identified 783 genes downregulated in hESCs and reported in at least three studies. This "consensus differentiation gene list" included the IL6ST/GP130 LIF receptor. We created an online hESC expression atlas, http://amazonia.montp.inserm.fr, to provide an easy access to this public transcriptome dataset. Expression histograms comparing hESCs to a broad collection of fetal and adult tissues can be retrieved with this web tool for more than 15,000 genes.
Collapse
Affiliation(s)
- Said Assou
- Centre Hospitalier Universitaire de Montpellier, Institute for Research in Biotherapy, Hôpital Saint-Eloi, 80 Avenue Augustin Fliche, 34295 Montpellier Cedex 5, France.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Nieto A, Cobo F, Barroso-Deljesús A, Barnie AH, Catalina P, Cabrera CM, Cortes JL, Montes RM, Concha A. Embryonic stem cell bank: a work proposal. ACTA ACUST UNITED AC 2007; 2:117-26. [PMID: 17237550 DOI: 10.1007/s12015-006-0018-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/1999] [Revised: 11/30/1999] [Accepted: 11/30/1999] [Indexed: 12/30/2022]
Abstract
Human embryonic stem cells (hESCs) have an unlimited capacity to proliferate by a self-renewal process and can be differentiated in the three germ layers, opening doors to new clinical therapies to replace missing or damaged cells. The number of research groups and projects using human stem cells has increased largely in the last 5 yr. The creation of stem cell banks is another important step to support the advance of research in this field. Banks must be operated within the strict regulatory famework of good manufacturing practices and good laboratory practices that assure the highest quality standards and must implement a quality system that complies with international quality systems standards. It may also be appropriate to aim at an accreditation in order to assure correct laboratory practices at all times. Stem cell banks should receive the lines previously derived by other groups and hESCs should be provided for groups that justify their use in a research project previously approved by an ethical committee. The assays generally accepted as typical of hESCs together with the microbiological analysis should be performed in order to assure a consistent, reliable, and safe line for the researchers. In this article, the Andalusian Stem Cell Bank proposes a model of a stem cell banking process in order to create a flow diagram of hESC lines and, following the international initiatives in stem cells research, to achieve the full characterization of cells and a standardization of protocols that would simplify the hESCs culture.
Collapse
Affiliation(s)
- A Nieto
- Andalusian Stem Cells, Andalusian Stem Cell, Bank, Granada, Spain.
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Nat R, Nilbratt M, Narkilahti S, Winblad B, Hovatta O, Nordberg A. Neurogenic neuroepithelial and radial glial cells generated from six human embryonic stem cell lines in serum-free suspension and adherent cultures. Glia 2007; 55:385-99. [PMID: 17152062 DOI: 10.1002/glia.20463] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The great potential of human embryonic stem (hES) cells offers the opportunity both for studying basic developmental processes in vitro as well as for drug screening, modeling diseases, or future cell therapy. Defining protocols for the generation of human neural progenies represents a most important prerequisite. Here, we have used six hES cell lines to evaluate defined conditions for neural differentiation in suspension and adherent culture systems. Our protocol does not require fetal serum, feeder cells, or retinoic acid at any step, to induce neural fate decisions in hES cells. We monitored neurogenesis in differentiating cultures using morphological (including on-line follow up), immunocytochemical, and RT-PCR assays. For each hES cell line, in suspension or adherent culture, the same longitudinal progression of neural differentiation occurs. We showed the dynamic transitions from hES cells to neuroepithelial (NE) cells, to radial glial (RG) cells, and to neurons. Thus, 7 days after neural induction the majority of cells were NE, expressing nestin, Sox1, and Pax6. During neural proliferation and differentiation, NE cells transformed in RG cells, which acquired vimentin, BLBP, GLAST, and GFAP, proliferated and formed radial scaffolds. gamma-Aminobutyric acid (GABA)-positive and glutamate positive neurons, few oligodendrocyte progenitors and astrocytes were formed in our conditions and timing. Our system successfully generates human RG cells and could be an effective source for neuronal replacement, since RG cells predominantly generate neurons and provide them with support and guidance.
Collapse
Affiliation(s)
- Roxana Nat
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | | | | | | | | | | |
Collapse
|
42
|
Ahmad S, Stewart R, Yung S, Kolli S, Armstrong L, Stojkovic M, Figueiredo F, Lako M. Differentiation of human embryonic stem cells into corneal epithelial-like cells by in vitro replication of the corneal epithelial stem cell niche. Stem Cells 2007; 25:1145-55. [PMID: 17255521 DOI: 10.1634/stemcells.2006-0516] [Citation(s) in RCA: 146] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Human embryonic stem cells (hESCs) are pluripotent cells capable of differentiating into any cell type of the body. It has long been known that the adult stem cell niche is vital for the maintenance of adult stem cells. The cornea at the front of the eye is covered by a stratified epithelium that is renewed by stem cells located at its periphery in a region known as the limbus. These so-called limbal stem cells are maintained by factors within the limbal microenvironment, including collagen IV in basement membrane and limbal fibroblasts in the stroma. Because this niche is very specific to the stem cells (rather than to the more differentiated cells) of the corneal epithelium, it was hypothesized that replication of these factors in vitro would result in hESC differentiation into corneal epithelial-like cells. Indeed, here we show that culturing of hESC on collagen IV using medium conditioned by the limbal fibroblasts results in the loss of pluripotency and differentiation into epithelial-like cells. Further differentiation results in the formation of terminally differentiated epithelial-like cells not only of the cornea but also of skin. Scanning electron microscopy shows that some differences exist between hESC-derived and adult limbal epithelial-like cells, necessitating further investigation using in vivo animal models of limbal stem cell deficiency. Such a model of hESC differentiation is useful for understanding the early events of epithelial lineage specification and to the eventual potential application of epithelium differentiated from hESC for clinical conditions of epithelial stem cell loss. Disclosure of potential conflicts of interest is found at the end of this article.
Collapse
Affiliation(s)
- Sajjad Ahmad
- Centre for Stem Cell Biology and Developmental Genetics, University of Newcastle, Newcastle upon Tyne, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Rajala K, Hakala H, Panula S, Aivio S, Pihlajamäki H, Suuronen R, Hovatta O, Skottman H. Testing of nine different xeno-free culture media for human embryonic stem cell cultures. Hum Reprod 2007; 22:1231-8. [PMID: 17251355 DOI: 10.1093/humrep/del523] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Human embryonic stem cells (hESC) are excellent candidates for cell replacement therapies. However, currently used culture conditions contain animal-derived components that bear a risk of transmitting animal pathogens and incorporation of non-human immunogenic molecules to hESC. METHODS Nine xeno-free culture media were compared with the conventional serum replacement (ko-SR) containing media in the culture of hESC on human feeder cells. Cultured hESC were characterized immunocytochemically and by fluorescence-activated cell sorter analysis. The differentiation potential of hESC cultured with xeno-free media was determined with the RT-PCR analysis. RESULTS The hESC cultured in xeno-free media differentiated or the proliferation decreased substantially. Under some test conditions, the morphology of the feeder cells was altered considerably. The hESC cultured with human serum underwent excessive differentiation in the beginning of culture, but a fraction of hESC was able to adapt to culture conditions containing 20% of human serum. CONCLUSIONS None of the studied xeno-free media was able to maintain the undifferentiated growth of hESC. The medium containing 20% human serum was found to sustain undifferentiated hESC proliferation to some extent, yet was inferior to the conventional ko-SR-containing medium.
Collapse
Affiliation(s)
- Kristiina Rajala
- REGEA, Institute for Regenerative Medicine, University of Tampere, Tampere University Hospital, Tampere, Finland.
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Glover CH, Marin M, Eaves CJ, Helgason CD, Piret JM, Bryan J. Meta-analysis of differentiating mouse embryonic stem cell gene expression kinetics reveals early change of a small gene set. PLoS Comput Biol 2006; 2:e158. [PMID: 17121458 PMCID: PMC1664699 DOI: 10.1371/journal.pcbi.0020158] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2006] [Accepted: 10/06/2006] [Indexed: 11/19/2022] Open
Abstract
Stem cell differentiation involves critical changes in gene expression. Identification of these should provide endpoints useful for optimizing stem cell propagation as well as potential clues about mechanisms governing stem cell maintenance. Here we describe the results of a new meta-analysis methodology applied to multiple gene expression datasets from three mouse embryonic stem cell (ESC) lines obtained at specific time points during the course of their differentiation into various lineages. We developed methods to identify genes with expression changes that correlated with the altered frequency of functionally defined, undifferentiated ESC in culture. In each dataset, we computed a novel statistical confidence measure for every gene which captured the certainty that a particular gene exhibited an expression pattern of interest within that dataset. This permitted a joint analysis of the datasets, despite the different experimental designs. Using a ranking scheme that favored genes exhibiting patterns of interest, we focused on the top 88 genes whose expression was consistently changed when ESC were induced to differentiate. Seven of these (103728_at, 8430410A17Rik, Klf2, Nr0b1, Sox2, Tcl1, and Zfp42) showed a rapid decrease in expression concurrent with a decrease in frequency of undifferentiated cells and remained predictive when evaluated in additional maintenance and differentiating protocols. Through a novel meta-analysis, this study identifies a small set of genes whose expression is useful for identifying changes in stem cell frequencies in cultures of mouse ESC. The methods and findings have broader applicability to understanding the regulation of self-renewal of other stem cell types.
Collapse
Affiliation(s)
- Clive H Glover
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
| | - Michael Marin
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Statistics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Connie J Eaves
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Cheryl D Helgason
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Cancer Endocrinology, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - James M Piret
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Chemical and Biological Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jennifer Bryan
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Statistics, University of British Columbia, Vancouver, British Columbia, Canada
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
45
|
Burridge PW, Anderson D, Priddle H, Barbadillo Muñoz MD, Chamberlain S, Allegrucci C, Young LE, Denning C. Improved human embryonic stem cell embryoid body homogeneity and cardiomyocyte differentiation from a novel V-96 plate aggregation system highlights interline variability. Stem Cells 2006; 25:929-38. [PMID: 17185609 DOI: 10.1634/stemcells.2006-0598] [Citation(s) in RCA: 219] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Although all human ESC (hESC) lines have similar morphology, express key pluripotency markers, and can differentiate toward primitive germ layers in vitro, the lineage-specific developmental potential may vary between individual lines. In the current study, four hESC lines were cultured in the same feeder-free conditions to provide a standardized platform for interline analysis. A high-throughput, forced-aggregation system involving centrifugation of defined numbers of hESCs in V-96 plates (V-96FA) was developed to examine formation, growth, and subsequent cardiomyocyte differentiation from >22,000 EBs. Homogeneity of EBs formed by V-96FA in mouse embryo fibroblast-conditioned medium was significantly improved compared with formation in mass culture (p < .02; Levene's test). V-96FA EB formation was successful in all four lines, although significant differences in EB growth were observed during the first 6 days of differentiation (p = .044 to .001; one-way analysis of variance [ANOVA]). Cardiomyocyte differentiation potential also varied; 9.5% +/- 0.9%, 6.6% +/- 2.4%, 5.2% +/- 3.1%, and 1.6% +/- 1.0% beating EBs were identified for HUES-7, NOTT2, NOTT1, and BG01, respectively (p = .008; one-way ANOVA). Formation of HUES-7 V-96FA EBs in defined medium containing activin A and basic fibroblast growth factor resulted in 23.6% +/- 3.6% beating EBs, representing a 13.1-fold increase relative to mass culture (1.8% +/- 0.7%), consistent with an observed 14.8-fold increase in MYH6 (alphaMHC) expression by real-time polymerase chain reaction. In contrast, no beating areas were derived from NOTT1-EBs and BG01-EBs formed in defined medium. Thus, the V-96FA system highlighted interline variability in EB growth and cardiomyocyte differentiation but, under the test conditions described, identified HUES-7 as a line that can respond to cardiomyogenic stimulation.
Collapse
Affiliation(s)
- Paul W Burridge
- Wolfson Centre for Stem Cells, Tissue Engineering and Modelling, School of Human Development, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Chen HF, Kuo HC, Chien CL, Shun CT, Yao YL, Ip PL, Chuang CY, Wang CC, Yang YS, Ho HN. Derivation, characterization and differentiation of human embryonic stem cells: comparing serum-containing versus serum-free media and evidence of germ cell differentiation. Hum Reprod 2006; 22:567-77. [PMID: 17071820 DOI: 10.1093/humrep/del412] [Citation(s) in RCA: 111] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND This study was designed to establish human embryonic stem cell (hESC) lines, to identify the differences when maintained in serum-containing versus serum-free medium and to test their potential of in vitro differentiation. METHODS Procedures including immunosurgery were performed on 11 donated human blastocysts to establish hESC lines. The cell lines were characterized and maintained using either serum-free or serum-containing media to compare their morphology, Oct-4 expression, apoptosis and growth speed. Differentiation of these lines was evaluated by the morphology and the expression of genes belonging to the three embryonic germ layers and the germ cell lineage. RESULTS Three hESC lines were established, and they grew at similar speed in both media (serum-containing or serum-free), but hESC cultured in serum-containing medium yielded significantly higher percentages of morphologically good colonies and cells expressing Oct-4. These cell lines differentiated spontaneously in vitro into cells expressing markers belonging to all three embryonic germ layers and germ cell markers, including c-Kit, STELLA, VASA and growth differentiation factor 9 (GDF9), in directly adherent culture. CONCLUSIONS Three hESC lines with Taiwanese ancestry have been established, and they retain the in vitro differentiation potential with or without embryoid body (EB) formation. The data support that hESC may be capable of differentiation into germ cells although further confirmation is needed. It is also suggested that strategies such as stepwise adaptation will be needed before implementing a serum-free culture condition for hESC lines that have previously been derived in a medium containing serum.
Collapse
Affiliation(s)
- H-F Chen
- Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, College of Medicine and Hospital, National Taiwan University, Taipei, Taiwan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
One Successful Series Begets Another. Stem Cells 2006. [DOI: 10.1634/stemcells.2006-0549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
48
|
Aghajanova L, Skottman H, Strömberg AM, Inzunza J, Lahesmaa R, Hovatta O. Expression of leukemia inhibitory factor and its receptors is increased during differentiation of human embryonic stem cells. Fertil Steril 2006; 86:1193-209. [PMID: 16949591 DOI: 10.1016/j.fertnstert.2005.12.081] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2005] [Revised: 12/22/2005] [Accepted: 12/22/2005] [Indexed: 11/16/2022]
Abstract
OBJECTIVE To investigate gene expression profiles during the early spontaneous differentiation of human embryonic stem cells (hESCs), with particular emphasis on leukemia inhibitory factor (LIF)-induced pathways and the ultrastructural surface morphology of the undifferentiated and spontaneously differentiated hESCs. DESIGN Prospective experimental study. SETTING University laboratory. PATIENT(S) Four hESC cell lines. INTERVENTION(S) The effect of LIF on receptor expression level was studied in cultures. MAIN OUTCOME MEASURE(S) Gene expression in the hESC line HS237 was analyzed using microarrays. Real-time reverse-transcription polymerase chain reaction was used to validate the microarray results in four hESC lines (HS181, HS235, HS237, HS293). Immunohistochemistry was used to assay LIF, LIF receptor, and gp130 protein expression. Cell surface morphology was studied using scanning electron microscopy. RESULT(S) The expression of LIF, LIF receptor, and gp130 messenger RNA and protein was increased in spontaneously differentiated HS237 cells compared with undifferentiated cells, with high expression of an inhibitor of LIF-mediated signaling, suppressor of cytokine signaling-1, in undifferentiated hESCs. Genes, those expressed specifically and those shared in undifferentiated hESCs, differentiated cells, and in fibroblasts, were identified. Supplementation with LIF did not affect the LIF receptor expression. CONCLUSION(S) The expression of LIF and its receptors is low in undifferentiated hESCs but increases during differentiation. Added LIF does not prevent spontaneous differentiation. Suppressor of cytokine signaling-1 may prevent LIF signaling in hESCs.
Collapse
Affiliation(s)
- Lusine Aghajanova
- Department of Obstetrics and Gynecology, CLINTEC, Karolinska Institute, Karolinska University Hospital Huddinge, Stockholm, Sweden.
| | | | | | | | | | | |
Collapse
|
49
|
Abstract
The promise of human embryonic stem cell (hESC) lines for treating injuries and degenerative diseases, for understanding early human development, for disease modelling and for drug discovery, has brought much excitement to scientific communities as well as to the public. Although all of the lines derived worldwide share the expression of characteristic pluripotency markers, many differences are emerging between lines that may be more associated with the wide range of culture conditions in current use than the inherent genetic variation of the embryos from which embryonic stem cells were derived. Thus, the validity of many comparisons between lines published thus far is difficult to interpret. This article reviews the evidence for differences between lines, focusing on studies of pluripotency marker molecules, transcriptional profiling, genetic stability and epigenetic stability, for which there is most evidence. Recognition and assessment of environmentally induced differences will be important to facilitate the development of culture systems that maximize stability in culture and provide lines with maximal potential for safety and success in the range of possible applications.
Collapse
Affiliation(s)
- C Allegrucci
- School of Human Development, University of Nottingham, Queens Medical Centre, Nottingham, UK
| | | |
Collapse
|
50
|
Armstrong L, Hughes O, Yung S, Hyslop L, Stewart R, Wappler I, Peters H, Walter T, Stojkovic P, Evans J, Stojkovic M, Lako M. The role of PI3K/AKT, MAPK/ERK and NFkappabeta signalling in the maintenance of human embryonic stem cell pluripotency and viability highlighted by transcriptional profiling and functional analysis. Hum Mol Genet 2006; 15:1894-913. [PMID: 16644866 DOI: 10.1093/hmg/ddl112] [Citation(s) in RCA: 309] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Understanding the molecular mechanism by which pluripotency is maintained in human embryonic stem cells (hESC) is important for the development of improved methods to derive, culture and differentiate these into cells of potential therapeutic use. Large-scale transcriptional comparison of the hES-NCL1 line derived from a day 8 embryo with H1 line derived from a day 5 embryo (WiCell Inc.) showed that only 0.52% of the transcripts analysed varied significantly between the two cell lines. This is within the variability range that has been reported when hESC derived from days 5-6 embryos have been compared with each other. This implies that transcriptional differences between the cell lines are likely to reflect their genetic profile rather than the embryonic stage from which they were derived. Bioinformatic analysis of expression changes observed when these cells were induced to differentiate as embryoid bodies suggested that quite a few of the downregulated genes were components of signal transduction networks. Subsequent analysis using western blotting, flow cytometry and antibody arrays implicated components of the PI3K/AKT kinase, MAPK/ERK and NFkappabeta pathways and confirmed that these components are decreased upon differentiation. Disruption of these pathways in isolation using specific inhibitors resulted in loss of pluripotency and/or loss of viability suggesting the importance of such signalling pathways in embryonic stem cell maintenance.
Collapse
Affiliation(s)
- Lyle Armstrong
- Centre for Stem Cell Biology and Developmental Genetics, Institute of Human Genetics, University of Newcastle, Central Parkways, Newcastle upon Tyne, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|