Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Stem Cell Reports ; 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38964325

ABSTRACT

Culture-acquired variants in human pluripotent stem cells (hPSCs) hinder their applications in research and clinic. However, the mechanisms that underpin selection of variants remain unclear. Here, through analysis of comprehensive karyotyping datasets from over 23,000 hPSC cultures of more than 1,500 lines, we explored how culture conditions shape variant selection. Strikingly, we identified an association of chromosome 1q gains with feeder-free cultures and noted a rise in its prevalence in recent years, coinciding with increased usage of feeder-free regimens. Competition experiments of multiple isogenic lines with and without a chromosome 1q gain confirmed that 1q variants have an advantage in feeder-free (E8/vitronectin), but not feeder-based, culture. Mechanistically, we show that overexpression of MDM4, located on chromosome 1q, drives variants' advantage in E8/vitronectin by alleviating genome damage-induced apoptosis, which is lower in feeder-based conditions. Our study explains condition-dependent patterns of hPSC aberrations and offers insights into the mechanisms of variant selection.

2.
Stem Cell Reports ; 19(1): 1-10, 2024 01 09.
Article in English | MEDLINE | ID: mdl-38157849

ABSTRACT

The expression of one or more of a small number of molecules, typically cell surface-associated antigens, or transcription factors, is widely used for identifying pluripotent stem cells (PSCs) or for monitoring their differentiation. However, none of these marker molecules are uniquely expressed by PSCs and all are expressed by stem cells that have lost the ability to differentiate. Consequently, none are indicators of pluripotency, per se. Here we summarize the nature and characteristics of several markers that are in wide use, including the cell surface antigens, stage-specific embryonic antigen (SSEA)-1, SSEA-3, SSEA-4, TRA-1-60, TRA-1-81, GCTM2, and the transcription factors POUF5/OCT4, NANOG, and SOX2, highlighting issues that must be considered when interpreting data about their expression on putative PSCs.


Subject(s)
Pluripotent Stem Cells , Pluripotent Stem Cells/metabolism , Lewis X Antigen/metabolism , Cell Differentiation , Transcription Factors/genetics , Antigens, Surface/metabolism , Octamer Transcription Factor-3/metabolism
3.
Dev Cell ; 56(17): 2455-2470.e10, 2021 09 13.
Article in English | MEDLINE | ID: mdl-34407428

ABSTRACT

The appearance of genetic changes in human pluripotent stem cells (hPSCs) presents a concern for their use in research and regenerative medicine. Variant hPSCs that harbor recurrent culture-acquired aneuploidies display growth advantages over wild-type diploid cells, but the mechanisms that yield a drift from predominantly wild-type to variant cell populations remain poorly understood. Here, we show that the dominance of variant clones in mosaic cultures is enhanced through competitive interactions that result in the elimination of wild-type cells. This elimination occurs through corralling and mechanical compression by faster-growing variants, causing a redistribution of F-actin and sequestration of yes-associated protein (YAP) in the cytoplasm that induces apoptosis in wild-type cells. YAP overexpression or promotion of YAP nuclear localization in wild-type cells alleviates their "loser" phenotype. Our results demonstrate that hPSC fate is coupled to mechanical cues imposed by neighboring cells and reveal that hijacking this mechanism allows variants to achieve clonal dominance in cultures.


Subject(s)
Cell Competition/genetics , Cell Differentiation/genetics , Cell Proliferation/genetics , Pluripotent Stem Cells/cytology , YAP-Signaling Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Cells, Cultured , Cytoplasm/metabolism , Humans , Transcription Factors/metabolism
4.
Stem Cell Reports ; 14(6): 1009-1017, 2020 06 09.
Article in English | MEDLINE | ID: mdl-32413278

ABSTRACT

Human pluripotent stem cells (PSCs) are subject to the appearance of recurrent genetic variants on prolonged culture. We have now found that, compared with isogenic differentiated cells, PSCs exhibit evidence of considerably more DNA damage during the S phase of the cell cycle, apparently as a consequence of DNA replication stress marked by slower progression of DNA replication, activation of latent origins of replication, and collapse of replication forks. As in many cancers, which, like PSCs, exhibit a shortened G1 phase and DNA replication stress, the resulting DNA damage may underlie the higher incidence of abnormal and abortive mitoses in PSCs, resulting in chromosomal non-dysjunction or cell death. However, we have found that the extent of DNA replication stress, DNA damage, and consequent aberrant mitoses can be substantially reduced by culturing PSCs in the presence of exogenous nucleosides, resulting in improved survival, clonogenicity, and population growth.


Subject(s)
DNA Replication , Genomic Instability , Nucleosides/pharmacology , Pluripotent Stem Cells/drug effects , Cell Line , Culture Media/chemistry , Culture Media/pharmacology , Cytoprotection , DNA Damage , Humans , Mitosis , Nucleosides/analysis , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism
5.
Nat Commun ; 11(1): 1528, 2020 03 23.
Article in English | MEDLINE | ID: mdl-32251294

ABSTRACT

The occurrence of repetitive genomic changes that provide a selective growth advantage in pluripotent stem cells is of concern for their clinical application. However, the effect of different culture conditions on the underlying mutation rate is unknown. Here we show that the mutation rate in two human embryonic stem cell lines derived and banked for clinical application is low and not substantially affected by culture with Rho Kinase inhibitor, commonly used in their routine maintenance. However, the mutation rate is reduced by >50% in cells cultured under 5% oxygen, when we also found alterations in imprint methylation and reversible DNA hypomethylation. Mutations are evenly distributed across the chromosomes, except for a slight increase on the X-chromosome, and an elevation in intergenic regions suggesting that chromatin structure may affect mutation rate. Overall the results suggest that pluripotent stem cells are not subject to unusually high rates of genetic or epigenetic alterations.


Subject(s)
Cell Culture Techniques/methods , Chromosomes, Human, X/genetics , DNA, Intergenic/genetics , Mutation Rate , Pluripotent Stem Cells/physiology , Cell Line , Culture Media/pharmacology , DNA Methylation , DNA Mutational Analysis , Epigenesis, Genetic , Humans , Oxidative Stress/drug effects , Oxidative Stress/genetics , Oxygen/chemistry , Oxygen/pharmacology , Sequence Analysis, RNA , Whole Genome Sequencing
6.
Stem Cell Reports ; 10(6): 1895-1907, 2018 06 05.
Article in English | MEDLINE | ID: mdl-29779895

ABSTRACT

Human embryonic stem cells (hESCs) display substantial heterogeneity in gene expression, implying the existence of discrete substates within the stem cell compartment. To determine whether these substates impact fate decisions of hESCs we used a GFP reporter line to investigate the properties of fractions of putative undifferentiated cells defined by their differential expression of the endoderm transcription factor, GATA6, together with the hESC surface marker, SSEA3. By single-cell cloning, we confirmed that substates characterized by expression of GATA6 and SSEA3 include pluripotent stem cells capable of long-term self-renewal. When clonal stem cell colonies were formed from GATA6-positive and GATA6-negative cells, more of those derived from GATA6-positive cells contained spontaneously differentiated endoderm cells than similar colonies derived from the GATA6-negative cells. We characterized these discrete cellular states using single-cell transcriptomic analysis, identifying a potential role for SOX17 in the establishment of the endoderm-biased stem cell state.


Subject(s)
Cell Self Renewal , Endoderm/cytology , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Biomarkers , Cell Differentiation/genetics , GATA6 Transcription Factor/genetics , GATA6 Transcription Factor/metabolism , Gene Expression Profiling , Genes, Reporter , Humans , Immunophenotyping , Single-Cell Analysis/methods
7.
Stem Cell Reports ; 7(5): 998-1012, 2016 11 08.
Article in English | MEDLINE | ID: mdl-27829140

ABSTRACT

Genetic changes in human pluripotent stem cells (hPSCs) gained during culture can confound experimental results and potentially jeopardize the outcome of clinical therapies. Particularly common changes in hPSCs are trisomies of chromosomes 1, 12, 17, and 20. Thus, hPSCs should be regularly screened for such aberrations. Although a number of methods are used to assess hPSC genotypes, there has been no systematic evaluation of the sensitivity of the commonly used techniques in detecting low-level mosaicism in hPSC cultures. We have performed mixing experiments to mimic the naturally occurring mosaicism and have assessed the sensitivity of chromosome banding, qPCR, fluorescence in situ hybridization, and digital droplet PCR in detecting variants. Our analysis highlights the limits of mosaicism detection by the commonly employed methods, a pivotal requirement for interpreting the genetic status of hPSCs and for setting standards for safe applications of hPSCs in regenerative medicine.


Subject(s)
Genetic Variation , Mosaicism , Pluripotent Stem Cells/metabolism , Cell Culture Techniques , Cell Line , Chromosomes, Human , Chromosomes, Human, Pair 17 , Chromosomes, Human, Pair 20 , DNA Copy Number Variations , Humans , In Situ Hybridization, Fluorescence , Karyotype , Pluripotent Stem Cells/cytology , Polymerase Chain Reaction , Trisomy
8.
Cell Stem Cell ; 19(5): 653-662, 2016 11 03.
Article in English | MEDLINE | ID: mdl-27545503

ABSTRACT

Adult neural stem cells (NSCs) are defined by their inherent capacity to self-renew and give rise to neurons, astrocytes, and oligodendrocytes. In vivo, however, hippocampal NSCs do not generate oligodendrocytes for reasons that have remained enigmatic. Here, we report that deletion of Drosha in adult dentate gyrus NSCs activates oligodendrogenesis and reduces neurogenesis at the expense of gliogenesis. We further find that Drosha directly targets NFIB to repress its expression independently of Dicer and microRNAs. Knockdown of NFIB in Drosha-deficient hippocampal NSCs restores neurogenesis, suggesting that the Drosha/NFIB mechanism robustly prevents oligodendrocyte fate acquisition in vivo. Taken together, our findings establish that adult hippocampal NSCs inherently possess multilineage potential but that Drosha functions as a molecular barrier preventing oligodendrogenesis.


Subject(s)
Adult Stem Cells/cytology , Aging/metabolism , Hippocampus/cytology , Multipotent Stem Cells/cytology , NFI Transcription Factors/metabolism , Neural Stem Cells/cytology , Ribonuclease III/metabolism , Adult Stem Cells/metabolism , Animals , Base Sequence , Cell Differentiation , Dentate Gyrus/cytology , Gene Deletion , Gene Knockdown Techniques , Mice , Mice, Knockout , Multipotent Stem Cells/metabolism , NFI Transcription Factors/genetics , Neural Stem Cells/metabolism , Neurogenesis/genetics , Oligodendroglia/cytology , Oligodendroglia/metabolism , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism
9.
Sci Rep ; 5: 11694, 2015 Jul 14.
Article in English | MEDLINE | ID: mdl-26170169

ABSTRACT

Maternal exposure during pregnancy to toxins can occasionally lead to miscarriage and malformation. It is currently thought that toxins pass through the placental barrier, albeit bi-layered in the first trimester, and damage the fetus directly, albeit at low concentration. Here we examined the responses of human embryonic stem (hES) cells in tissue culture to two metals at low concentration. We compared direct exposures with indirect exposures across a bi-layered model of the placenta cell barrier. Direct exposure caused increased DNA damage without apoptosis or a loss of cell number but with some evidence of altered differentiation. Indirect exposure caused increased DNA damage and apoptosis but without loss of pluripotency. This was not caused by metal ions passing through the barrier. Instead the hES cells responded to signalling molecules (including TNF-α) secreted by the barrier cells. This mechanism was dependent on connexin 43 mediated intercellular 'bystander signalling' both within and between the trophoblast barrier and the hES colonies. These results highlight key differences between direct and indirect exposure of hES cells across a trophoblast barrier to metal toxins. It offers a theoretical possibility that an indirectly mediated toxicity of hES cells might have biological relevance to fetal development.


Subject(s)
Cell Communication , Human Embryonic Stem Cells/metabolism , Signal Transduction , Trophoblasts/metabolism , Apoptosis/drug effects , Apoptosis/genetics , Cell Cycle Checkpoints/drug effects , Cell Differentiation/drug effects , Connexin 43/metabolism , Cytokines/biosynthesis , DNA Damage/drug effects , Fibroblasts/drug effects , Fibroblasts/metabolism , Gap Junctions/metabolism , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/drug effects , Humans , Metals, Heavy/toxicity , Trophoblasts/cytology , Trophoblasts/drug effects , Tumor Necrosis Factor-alpha/biosynthesis
10.
PLoS One ; 10(4): e0123467, 2015.
Article in English | MEDLINE | ID: mdl-25875838

ABSTRACT

We have used single cell transcriptome analysis to re-examine the substates of early passage, karyotypically Normal, and late passage, karyotypically Abnormal ('Culture Adapted') human embryonic stem cells characterized by differential expression of the cell surface marker antigen, SSEA3. The results confirmed that culture adaptation is associated with alterations to the dynamics of the SSEA3(+) and SSEA3(-) substates of these cells, with SSEA3(-) Adapted cells remaining within the stem cell compartment whereas the SSEA3(-) Normal cells appear to have differentiated. However, the single cell data reveal that these substates are characterized by further heterogeneity that changes on culture adaptation. Notably the Adapted population includes cells with a transcriptome substate suggestive of a shift to a more naïve-like phenotype in contrast to the cells of the Normal population. Further, a subset of the Normal SSEA3(+) cells expresses genes typical of endoderm differentiation, despite also expressing the undifferentiated stem cell genes, POU5F1 (OCT4) and NANOG, whereas such apparently lineage-primed cells are absent from the Adapted population. These results suggest that the selective growth advantage gained by genetically variant, culture adapted human embryonic stem cells may derive in part from a changed substate structure that influences their propensity for differentiation.


Subject(s)
Antigens, Tumor-Associated, Carbohydrate/genetics , Cell Differentiation , Stage-Specific Embryonic Antigens/genetics , Antigens, Tumor-Associated, Carbohydrate/metabolism , Cells, Cultured , Cluster Analysis , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Human Embryonic Stem Cells , Humans , Nanog Homeobox Protein , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Real-Time Polymerase Chain Reaction , Stage-Specific Embryonic Antigens/metabolism , Transcriptome
11.
Stem Cell Reports ; 3(1): 142-55, 2014 Jul 08.
Article in English | MEDLINE | ID: mdl-25068128

ABSTRACT

Using time-lapse imaging, we have identified a series of bottlenecks that restrict growth of early-passage human embryonic stem cells (hESCs) and that are relieved by karyotypically abnormal variants that are selected by prolonged culture. Only a minority of karyotypically normal cells divided after plating, and these were mainly cells in the later stages of cell cycle at the time of plating. Furthermore, the daughter cells showed a continued pattern of cell death after division, so that few formed long-term proliferating colonies. These colony-forming cells showed distinct patterns of cell movement. Increasing cell density enhanced cell movement facilitating cell:cell contact, which resulted in increased proportion of dividing cells and improved survival postplating of normal hESCs. In contrast, most of the karyotypically abnormal cells reentered the cell cycle on plating and gave rise to healthy progeny, without the need for cell:cell contacts and independent of their motility patterns.


Subject(s)
Embryonic Stem Cells/cytology , Cell Differentiation/physiology , Cells, Cultured , Embryonic Stem Cells/physiology , Humans , Time-Lapse Imaging
13.
Neuroreport ; 24(18): 1031-4, 2013 Dec 18.
Article in English | MEDLINE | ID: mdl-24089015

ABSTRACT

Human pluripotent stem cells (PSCs), whether embryonic stem cells or induced PSCs, offer enormous opportunities for regenerative medicine and other biomedical applications once we have developed the ability to harness their capacity for extensive differentiation. Central to this is our ability to identify and characterize such PSCs, but this is fraught with potential difficulties that arise from a tension between functional definitions of pluripotency and the more convenient use of 'markers', a problem exacerbated by ethical issues, our lack of knowledge of early human embryonic development, and differences from the mouse paradigm.


Subject(s)
Pluripotent Stem Cells/physiology , Animals , Embryonic Stem Cells/physiology , Embryonic Stem Cells/transplantation , Humans , Mice , Pluripotent Stem Cells/transplantation
14.
Stem Cell Res ; 11(3): 1022-36, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23941963

ABSTRACT

Genomic abnormalities may accumulate in human embryonic stem cells (hESCs) during in vitro maintenance. Characterization of the mechanisms enabling survival and expansion of abnormal hESCs is important due to consequences of genetic changes for the therapeutic utilization of stem cells. Furthermore, these cells provide an excellent model to study transformation in vitro. We report here that the histone deacetylase proteins, HDAC1 and HDAC2, are increased in karyotypically abnormal hESCs when compared to their normal counterparts. Importantly, similar to many cancer cell lines, we found that HDAC inhibitors repress proliferation of the karyotypically abnormal hESCs, whereas normal cells are more resistant to the treatment. The decreased proliferation correlates with downregulation of HDAC1 and HDAC2 proteins, induction of the proliferation inhibitor, cyclin-dependent kinase inhibitor 1A (CDKN1A), and altered regulation of tumor suppressor protein Retinoblastoma 1 (RB1). Through genome-wide transcriptome analysis we have identified genes with altered expression and responsiveness to HDAC inhibition in abnormal cells. Most of these genes are linked to severe developmental and neurological diseases and cancers. Our results highlight the importance of epigenetic mechanisms in the regulation of genomic stability of hESCs, and provide valuable candidates for targeted and selective growth inhibition of karyotypically abnormal cells.


Subject(s)
Chromosome Aberrations , Embryonic Stem Cells/drug effects , Gene Expression Regulation , Histone Deacetylase Inhibitors/pharmacology , Cell Differentiation/drug effects , Cell Line , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Down-Regulation/drug effects , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Genomic Instability , Histone Deacetylase 1/antagonists & inhibitors , Histone Deacetylase 1/genetics , Histone Deacetylase 1/metabolism , Histone Deacetylase 2/antagonists & inhibitors , Histone Deacetylase 2/metabolism , Humans , LDL-Receptor Related Proteins/genetics , LDL-Receptor Related Proteins/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Nervous System Diseases/genetics , Nervous System Diseases/metabolism , Nervous System Diseases/pathology , Osteopontin/genetics , Osteopontin/metabolism , RNA, Small Interfering/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
16.
PLoS One ; 8(2): e56893, 2013.
Article in English | MEDLINE | ID: mdl-23457636

ABSTRACT

Stella is a developmentally regulated gene highly expressed in mouse embryonic stem (ES) cells and in primordial germ cells (PGCs). In human, the gene encoding the STELLA homologue lies on chromosome 12p, which is frequently amplified in long-term cultured human ES cells. However, the role played by STELLA in human ES cells has not been reported. In the present study, we show that during retinoic acid (RA)-induced differentiation of human ES cells, expression of STELLA follows that of VASA, a marker of germline differentiation. By contrast, human embryonal carcinoma cells express STELLA at a higher level compared with both karyotypically normal and abnormal human ES cell lines. We found that over-expression of STELLA does not interfere with maintenance of the stem cell state of human ES cells, but following retinoic acid induction it leads to up-regulation of germline- and endodermal-associated genes, whereas neural markers PAX6 and NEUROD1 are down-regulated. Further, STELLA over-expression facilitates the differentiation of human ES cells into BE12-positive cells, in which the expression of germline- and endodermal-associated genes is enriched, and suppresses differentiation of the neural lineage. Taken together, this finding suggests a role for STELLA in facilitating germline and endodermal differentiation of human ES cells.


Subject(s)
Cell Lineage , Embryonic Stem Cells/cytology , Endoderm/cytology , Germ Cells/cytology , Proteins/metabolism , Cell Differentiation , Cell Line , Chromosomal Proteins, Non-Histone , Gene Expression Regulation, Developmental , Humans , Proteins/genetics , Time Factors
17.
Curr Opin Genet Dev ; 22(5): 403-8, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22868175

ABSTRACT

Stem cell biology has many roots, and the current interest in the possible medical and pharmaceutical applications of pluripotent stem cells has far removed origins in the biology of a rare but peculiar type of tumor, the teratomas. The identification of their stem cells and their relationship to the early embryo paved the way, first in the mouse and later in humans, to the development of embryonic stem (ES) and induced pluripotent stem (iPS) cells, and to approaches for controlling their differentiations. More recently, the recognition of genetic change and culture adaptation of these cells after prolonged culture has returned us to those cancer roots.


Subject(s)
Pluripotent Stem Cells/cytology , Teratoma/pathology , Animals , Cell Differentiation , Embryonic Stem Cells/cytology , Humans , Mice , Neoplastic Stem Cells/cytology
18.
Stem Cells ; 30(9): 1901-10, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22821732

ABSTRACT

Human embryonic stem cells (hESCs) tend to lose genomic integrity during long periods of culture in vitro and to acquire a cancer-like phenotype. In this study, we aim at understanding the contribution of point mutations to the adaptation process and at providing a mechanistic explanation for their accumulation. We observed that, due to the absence of p21/Waf1/Cip1, cultured hESCs lack proper cell cycle checkpoints and are vulnerable to the kind of DNA damage usually repaired by the highly versatile nucleotide excision repair (NER) pathway. In response to UV-induced DNA damage, the majority of hESCs succumb to apoptosis; however, a subpopulation continues to proliferate, carrying damaged DNA and accumulating point mutations with a typical UV-induced signature. The UV-resistant cells retain their proliferative capacity and potential for pluripotent differentiation and are markedly less apoptotic to subsequent UV exposure. These findings demonstrate that, due to deficient DNA damage response, the modest NER activity in hESCs is insufficient to prevent increased mutagenesis. This provides for the appearance of genetically aberrant hESCs, paving the way for further major genetic changes.


Subject(s)
Cell Cycle Checkpoints/genetics , DNA Damage , DNA Repair , Embryonic Stem Cells/physiology , Point Mutation , Apoptosis/genetics , Cell Growth Processes/genetics , Cells, Cultured , Embryonic Stem Cells/cytology , Humans
19.
Nat Biotechnol ; 29(12): 1132-44, 2011 Nov 27.
Article in English | MEDLINE | ID: mdl-22119741

ABSTRACT

The International Stem Cell Initiative analyzed 125 human embryonic stem (ES) cell lines and 11 induced pluripotent stem (iPS) cell lines, from 38 laboratories worldwide, for genetic changes occurring during culture. Most lines were analyzed at an early and late passage. Single-nucleotide polymorphism (SNP) analysis revealed that they included representatives of most major ethnic groups. Most lines remained karyotypically normal, but there was a progressive tendency to acquire changes on prolonged culture, commonly affecting chromosomes 1, 12, 17 and 20. DNA methylation patterns changed haphazardly with no link to time in culture. Structural variants, determined from the SNP arrays, also appeared sporadically. No common variants related to culture were observed on chromosomes 1, 12 and 17, but a minimal amplicon in chromosome 20q11.21, including three genes expressed in human ES cells, ID1, BCL2L1 and HM13, occurred in >20% of the lines. Of these genes, BCL2L1 is a strong candidate for driving culture adaptation of ES cells.


Subject(s)
Embryonic Stem Cells/cytology , Growth/genetics , Induced Pluripotent Stem Cells/cytology , RNA-Binding Proteins/metabolism , bcl-X Protein/metabolism , Cell Differentiation/genetics , Cell Line , Chromosomes, Human, Pair 20/genetics , Clonal Evolution/genetics , DNA Methylation , Ethnicity/genetics , Gene Expression Regulation, Developmental , Genetic Variation , Genotype , Humans , Inhibitor of Differentiation Protein 1/genetics , Inhibitor of Differentiation Protein 1/metabolism , Polymorphism, Single Nucleotide , RNA-Binding Proteins/genetics , Selection, Genetic/genetics , bcl-X Protein/genetics
20.
J Biomol Screen ; 16(6): 603-17, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21593487

ABSTRACT

Disentangling the complex interactions that govern stem cell fate choices of self-renewal, differentiation, or death presents a formidable challenge. Image-based phenotype-driven screening meets this challenge by providing means for rapid testing of many small molecules simultaneously. Pluripotent embryonal carcinoma (EC) cells offer a convenient substitute for embryonic stem (ES) cells in such screens because they are simpler to maintain and control. The authors developed an image-based screening assay to identify compounds that affect survival or differentiation of the human EC stem cell line NTERA2 by measuring the effect on cell number and the proportion of cells expressing a pluripotency-associated marker SSEA3. A pilot screen of 80 kinase inhibitors identified several compounds that improved cell survival or induced differentiation. The survival compounds Y-27632, HA-1077, and H-8 all strongly inhibit the kinases ROCK and PRK2, highlighting the important role of these kinases in EC cell survival. Two molecules, GF109203x and rottlerin, induced EC differentiation. The effects of rottlerin were also investigated in human ES cells. Rottlerin inhibited the self-renewal ability of ES cells, caused the cell cycle arrest, and repressed the expression of pluripotency-associated genes.


Subject(s)
Cell Differentiation/drug effects , Embryonal Carcinoma Stem Cells/cytology , Embryonal Carcinoma Stem Cells/drug effects , High-Throughput Screening Assays , Protein Kinase Inhibitors/pharmacology , Acetophenones/pharmacology , Benzopyrans/pharmacology , Cell Differentiation/genetics , Cell Survival/drug effects , Cell Survival/genetics , Embryonal Carcinoma Stem Cells/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental/drug effects , Humans , Image Processing, Computer-Assisted , Phenotype , Reproducibility of Results , Small Molecule Libraries/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...