Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
In Vitro Cell Dev Biol Anim ; 56(7): 505-510, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32812205

ABSTRACT

Cleft lip and palate are the most common congenital abnormalities that occur early in pregnancy. The majority of cranial mesenchyme is derived from cranial neural crest cells that differentiate into odontoblasts, cartilage, craniofacial bone, and connective tissue. A subset of these cells differentiates into cranial ganglia. We have previously reported an induction protocol of cranial neural crest cell-like cells from human pluripotent stem cells. This study tested detection of the cytotoxic sensitivities of dental materials, including titanium ions, palladium ions, and hydroxyethyl methacrylate, on the cell viability of induced cranial neural crest cell-like cells (iNC-LCs) derived from Tic human induced pluripotent stem cell (hiPSC) line. Further, the sensitivity was compared with those of human fetal lung fibroblastic cell line MRC-5, which is origin of Tic hiPSC, and osteoblastic cell line MC3T3-E1 which was derived from mouse calvaria. The results suggested that this cell-based assay system using iNC-LCs is a potential method for in vitro screening as an alternative to animal testing to predict toxic effects of dental materials on early craniofacial development.


Subject(s)
Biological Assay/methods , Induced Pluripotent Stem Cells/cytology , Models, Biological , Neural Crest/cytology , Skull/cytology , Cell Death/drug effects , Cell Line , Cell Survival/drug effects , Humans , Methacrylates/pharmacology , Palladium/pharmacology , Titanium/pharmacology
2.
Methods Mol Biol ; 1965: 35-48, 2019.
Article in English | MEDLINE | ID: mdl-31069667

ABSTRACT

Cranial neural crest cells (NCCs) migrate to the branchial arches and give rise to the majority of cranial mesenchyme that eventually differentiates into odontoblasts, cartilage, craniofacial bone, and connective tissue; a subset of these cells differentiate into cranial ganglia. Here we present a protocol that describes directed differentiation method of human pluripotent stem cells into cranial NCC-like cells and a cytotoxicity assay using hPSC-derived cranial NCC-like cells. This cell-based assay system allows for high-sensitive cytotoxicity detection of test chemicals. These methods can be applied to predict drug/chemical toxicity effect on early craniofacial development.


Subject(s)
Brain/cytology , Neural Crest/cytology , Pluripotent Stem Cells/cytology , Animals , Cell Culture Techniques , Cell Differentiation , Cells, Cultured , Humans , Models, Biological , Toxicity Tests
3.
Adv Exp Med Biol ; 1123: 71-94, 2019.
Article in English | MEDLINE | ID: mdl-31016596

ABSTRACT

Pluripotent stem cells (PSCs), including embryonic stem cells and induced pluripotent stem cells, show heterogeneity with respect to their pluripotency, self-renewal ability, and other traits. PSC heterogeneity may exist among cell lines, among cells within a line, and among temporal states of individual cells. Both genetic and epigenetic factors can cause heterogeneity among cell lines. Heterogeneity among cells within a cell line may arise during long-term culturing even when a PSC cell line is derived from a single cell. Moreover, the expression levels of genes and proteins in PSCs fluctuate continuously at a frequency ranging from a few hours to a few days. Such heterogeneity decreases the reproducibility of research. Thus, methods related to the detection, reduction, and control of heterogeneity in experiments involving human PSCs need to be developed. Further, the presupposition that PSCs are highly heterogeneous should be taken into account by all researchers not only when they plan their own studies but also when they review the studies of other researchers in this field.


Subject(s)
Pluripotent Stem Cells/cytology , Cell Differentiation , Cell Line , Embryonic Stem Cells , Humans , Induced Pluripotent Stem Cells
4.
Int J Dev Biol ; 62(9-10): 613-621, 2018.
Article in English | MEDLINE | ID: mdl-30378385

ABSTRACT

Cell morphology is recognized as an important hallmark of neural cells. During the differentiation of human pluripotent stem cells (hPSCs) into neural cells, cell morphology changes dynamically. Therefore, characterization of the morphology of cells during this period is important to improve our understanding of the differentiation and development of neural cells. General methods for the directed induction of hPSCs include the steps of multi-cellular aggregation or high-density cell culture, particularly at the early phase of neural differentiation, and therefore, the morphology of each differentiating cell is difficult to recognize. Here, we have developed a new method for the directed differentiation of neuroepithelial-like cells (NELCs) from hPSCs at a low cell density in an adherent monolayer culture, as well as an image-processing algorithm to evaluate the cell morphology of differentiating NELCs, in order to follow cell morphology during the differentiation of hPSCs into NELCs. Using these methods, the morphological transition of differentiating cells was observed in real time using phase contrast imaging and then quantified. Because cell morphology is also considered an inherent biological marker of neural cells cultured in vitro, this method is potentially useful to study the mechanisms underlying neural cell differentiation.


Subject(s)
Cell Differentiation , Induced Pluripotent Stem Cells/cytology , Neuroepithelial Cells/cytology , Neurogenesis , Neurons/cytology , Biomarkers/metabolism , Cell Culture Techniques , Humans , Induced Pluripotent Stem Cells/metabolism , Neuroepithelial Cells/metabolism , Neurons/metabolism
5.
J Biosci Bioeng ; 126(3): 379-388, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29681444

ABSTRACT

Spatial pattern formation is a critical step in embryogenesis. Bone morphogenetic protein 4 (BMP4) and its inhibitors are major factors for the formation of spatial patterns during embryogenesis. However, spatial patterning of the human embryo is unclear because of ethical issues and isotropic culture environments resulting from conventional culture dishes. Here, we utilized human pluripotent stem cells (hiPSCs) and a simple anisotropic (unidirectional perfusion) culture chamber, which creates unidirectional conditions, to measure the cell community effect. The influence of cell density on BMP4-induced differentiation was explored during static culture using a conventional culture dish. Immunostaining of the early differentiation marker SSEA-1 and the mesendoderm marker BRACHYURY revealed that high cell density suppressed differentiation, with small clusters of differentiated and undifferentiated cells formed. Addition of five-fold higher concentration of BMP4 showed similar results, suggesting that suppression was not caused by depletion of BMP4 but rather by high cell density. Quantitative RT-PCR array analysis showed that BMP4 induced multi-lineage differentiation, which was also suppressed under high-density conditions. We fabricated an elongated perfusion culture chamber, in which proteins were transported unidirectionally, and hiPSCs were cultured with BMP4. At low density, the expression was the same throughout the chamber. However, at high density, SSEA-1 and BRACHYURY were expressed only in upstream cells, suggesting that some autocrine/paracrine factors inhibited the action of BMP4 in downstream cells to form the spatial pattern. Human iPSCs cultured in a perfusion culture chamber might be useful for studying in vitro macroscopic pattern formation in human embryogenesis.


Subject(s)
Bone Morphogenetic Protein 4/pharmacology , Cell Culture Techniques/methods , Cell Differentiation/drug effects , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/physiology , Cell Adhesion/drug effects , Cell Count , Cells, Cultured , Embryo, Mammalian/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/physiology , Pluripotent Stem Cells/drug effects , Spatial Analysis , Surface Properties
6.
Exp Cell Res ; 352(2): 333-345, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28215634

ABSTRACT

Limited growth potential, narrow ranges of sources, and difference in variability and functions from batch to batch of primary hepatocytes cause a problem for predicting drug-induced hepatotoxicity during drug development. Human pluripotent stem cell (hPSC)-derived hepatocyte-like cells in vitro are expected as a tool for predicting drug-induced hepatotoxicity. Several studies have already reported efficient methods for differentiating hPSCs into hepatocyte-like cells, however its differentiation process is time-consuming, labor-intensive, cost-intensive, and unstable. In order to solve this problem, expansion culture for hPSC-derived hepatic progenitor cells, including hepatic stem cells and hepatoblasts which can self-renewal and differentiate into hepatocytes should be valuable as a source of hepatocytes. However, the mechanisms of the expansion of hPSC-derived hepatic progenitor cells are not yet fully understood. In this study, to isolate hPSC-derived hepatic progenitor cells, we tried to develop serum-free growth factor defined culture conditions using defined components. Our culture conditions were able to isolate and grow hPSC-derived hepatic progenitor cells which could differentiate into hepatocyte-like cells through hepatoblast-like cells. We have confirmed that the hepatocyte-like cells prepared by our methods were able to increase gene expression of cytochrome P450 enzymes upon encountering rifampicin, phenobarbital, or omeprazole. The isolation and expansion of hPSC-derived hepatic progenitor cells in defined culture conditions should have advantages in terms of detecting accurate effects of exogenous factors on hepatic lineage differentiation, understanding mechanisms underlying self-renewal ability of hepatic progenitor cells, and stably supplying functional hepatic cells.


Subject(s)
Cellular Reprogramming Techniques/methods , Embryonic Stem Cells/cytology , Hepatocytes/cytology , Pluripotent Stem Cells/cytology , Cell Differentiation , Cells, Cultured , Culture Media, Serum-Free/pharmacology , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism
7.
In Vitro Cell Dev Biol Anim ; 53(1): 83-91, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27573412

ABSTRACT

Human pluripotent stem cells (hPSCs) provide a good model system for studying human development and are expected as a source for both cell-based medical and pharmaceutical research application. However, stable maintenance of undifferentiated hPSCs is yet challenging, and thus routine characterization is required. Flow-cytometry is one of the popular quantitative characterization tools for hPSCs, but it has drawback of spatial information loss of the cells in the culture. Here, we have applied a two-dimensional imaging cytometry that examines undifferentiated state of hPSCs to analyze localization and morphological information of immunopositive cells in the culture. The whole images of cells in a culture vessel were acquired and analyzed by an image analyzer, IN Cell Analyzer 2000, and determined staining intensity of the cells with their positional information. We have compared the expression of five hPSC-markers in four hPSC lines using the two-dimensional imaging cytometry and flow cytometry. The results showed that immunopositive ratios analyzed by the imaging cytometry had good correlation with those by the flow cytometry. Furthermore, the imaging cytometry revealed spatially heterogenic expression of hPSC-markers in undifferentiated hPSCs. Imaging cytometry is capable of reflecting minute aberrance without losing spatial and morphological information of the cells. It would be a powerful, useful, and time-efficient tool for characterizing hPSC colonies.


Subject(s)
Biomarkers/metabolism , Cell Differentiation , Flow Cytometry/methods , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Cell Shape , Colony-Forming Units Assay , Gene Expression Profiling , Gene Expression Regulation , Humans
8.
Stem Cells Dev ; 25(24): 1884-1897, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27733097

ABSTRACT

Functional hepatocytes derived from human pluripotent stem cells (hPSCs) have potential as tools for predicting drug-induced hepatotoxicity in the early phases of drug development. However, the propensity of hPSC lines to differentiate into specific lineages is reported to differ. The ability to predict low propensity of hPSCs to differentiate into hepatocytes would facilitate the selection of useful hPSC clones and substantially accelerate development of hPSC-derived hepatocytes for pharmaceutical research. In this study, we compared the expression of genes associated with hepatic differentiation in five hPSC lines including human ES cell line, H9, which is known to differentiate into hepatocytes, and an hPSC line reported with a poor propensity for hepatic differentiation. Genes distinguishing between undifferentiated hPSCs, hPSC-derived hepatoblast-like differentiated cells, and primary human hepatocytes were drawn by two-way cluster analysis. The order of expression levels of genes in undifferentiated hPSCs was compared with that in hPSC-derived hepatoblast-like cells. Three genes were selected as predictors of low propensity for hepatic differentiation. Expression of these genes was investigated in 23 hPSC clones. Review of representative cells by induction of hepatic differentiation suggested that low prediction scores were linked with low hepatic differentiation. Thus, our model using gene expression ranking and bioinformatic analysis could reasonably predict poor differentiation propensity of hPSC lines.


Subject(s)
Cell Differentiation/genetics , Gene Expression Regulation , Hepatocytes/cytology , Pluripotent Stem Cells/cytology , Cell Line , Cell Lineage/genetics , Cluster Analysis , Endoderm/cytology , Gene Expression Profiling , Genetic Association Studies , Hepatocytes/metabolism , Humans , Pluripotent Stem Cells/metabolism
9.
Sci Rep ; 6: 34009, 2016 Sep 26.
Article in English | MEDLINE | ID: mdl-27667091

ABSTRACT

Given the difficulties inherent in maintaining human pluripotent stem cells (hPSCs) in a healthy state, hPSCs should be routinely characterized using several established standard criteria during expansion for research or therapeutic purposes. hPSC colony morphology is typically considered an important criterion, but it is not evaluated quantitatively. Thus, we designed an unbiased method to evaluate hPSC colony morphology. This method involves a combination of automated non-labelled live-cell imaging and the implementation of morphological colony analysis algorithms with multiple parameters. To validate the utility of the quantitative evaluation method, a parent cell line exhibiting typical embryonic stem cell (ESC)-like morphology and an aberrant hPSC subclone demonstrating unusual colony morphology were used as models. According to statistical colony classification based on morphological parameters, colonies containing readily discernible areas of differentiation constituted a major classification cluster and were distinguishable from typical ESC-like colonies; similar results were obtained via classification based on global gene expression profiles. Thus, the morphological features of hPSC colonies are closely associated with cellular characteristics. Our quantitative evaluation method provides a biological definition of 'hPSC colony morphology', permits the non-invasive monitoring of hPSC conditions and is particularly useful for detecting variations in hPSC heterogeneity.

10.
Int J Dev Biol ; 60(1-3): 21-8, 2016.
Article in English | MEDLINE | ID: mdl-26934293

ABSTRACT

Neural crest (NC) cells are a group of cells located in the neural folds at the boundary between the neural and epidermal ectoderm. Cranial NC cells migrate to the branchial arches and give rise to the majority of the craniofacial region, whereas trunk and tail NC cells contribute to the heart, enteric ganglia of the gut, melanocytes, sympathetic ganglia, and adrenal chromaffin cells. Positional information is indispensable for the regulation of cranial or trunk and tail NC cells. However, the mechanisms underlying the regulation of positional information during human NC induction have yet to be fully elucidated. In the present study, supplementation of bone morphogenetic protein (BMP) 4 in defined serum-free culture conditions including fibroblast growth factor-2 and Wnt3a from day 8 after NC specification induced the expression of cranial NC markers, AP2alpha, MSX1, and DLX1, during NC cell differentiation from human pluripotent stem cells. On the other hand, the proportion of cells expressing p75(NTR) or HNK1 decreased compared with that of cells cultured without BMP4, whereas gene expression analysis demonstrated that the expression levels of cranial NC-associated genes increased in BMP4-treated NC cells. These BMP4-treated NC cells were capable of differentiation into osteocytes and chondrocytes. The results of the present study indicate that BMP4 regulates cranial positioning during NC development.


Subject(s)
Bone Morphogenetic Protein 4/pharmacology , Cell Differentiation/drug effects , Neural Crest/cytology , Pluripotent Stem Cells/cytology , Transcriptional Activation/drug effects , Cell Differentiation/genetics , Cell Line , Chondrocytes/cytology , Chondrocytes/metabolism , Facial Bones/cytology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Immunohistochemistry , MSX1 Transcription Factor/genetics , MSX1 Transcription Factor/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Crest/metabolism , Osteocytes/cytology , Osteocytes/metabolism , Pluripotent Stem Cells/metabolism , Receptors, Nerve Growth Factor/genetics , Receptors, Nerve Growth Factor/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Skull/cytology , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation/genetics , Transcriptome/drug effects , Transcriptome/genetics
11.
Stem Cells Transl Med ; 5(3): 275-81, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26819254

ABSTRACT

Mitochondrial oxidative phosphorylation is a major source of cellular ATP. Its usage as an energy source varies, not only according to the extracellular environment, but also during development and differentiation, as indicated by the reported changes in the flux ratio of glycolysis to oxidative phosphorylation during embryonic stem (ES) cell differentiation. The fluorescent probe JC-1 allows visualization of changes in the mitochondrial membrane potential produced by oxidative phosphorylation. Strong JC-1 signals were localized in the differentiated cells located at the edge of H9 ES colonies that expressed vimentin, an early differentiation maker. The JC-1 signals were further intensified when individual adjacent colonies were in contact with each other. Time-lapse analyses revealed that JC-1-labeled H9 cells under an overconfluent condition were highly differentiated after subculture, suggesting that monitoring oxidative phosphorylation in live cells might facilitate the prediction of induced pluripotent stem cells, as well as ES cells, that are destined to lose their undifferentiated potency.


Subject(s)
Cell Differentiation/genetics , Human Embryonic Stem Cells/metabolism , Induced Pluripotent Stem Cells/metabolism , Oxidative Phosphorylation , Adenosine Triphosphate , Cell Line , Cell Tracking/methods , Energy Metabolism , Fluorescent Dyes/metabolism , Human Embryonic Stem Cells/cytology , Humans , Induced Pluripotent Stem Cells/cytology , Membrane Potential, Mitochondrial , Mitochondria/metabolism , Vimentin/biosynthesis
12.
J Biol Chem ; 290(33): 20071-85, 2015 Aug 14.
Article in English | MEDLINE | ID: mdl-26100630

ABSTRACT

We have generated a mouse monoclonal antibody (R-17F, IgG1 subtype) specific to human induced pluripotent stem (hiPS)/embryonic stem (ES) cells by using a hiPS cell line as an antigen. Triple-color confocal immunostaining images of hiPS cells with R-17F indicated that the R-17F epitope was expressed exclusively and intensively on the cell membranes of hiPS cells and co-localized partially with those of SSEA-4 and SSEA-3. Lines of evidence suggested that the predominant part of the R-17F epitope was a glycolipid. Upon TLC blot of total lipid extracts from hiPS cells with R-17F, one major R-17F-positive band was observed at a slow migration position close to that of anti-blood group H1(O) antigen. MALDI-TOF-MS and MS(n) analyses of the purified antigen indicated that the presumptive structure of the R-17F antigen was Fuc-Hex-HexNAc-Hex-Hex-Cer. Glycan microarray analysis involving 13 different synthetic oligosaccharides indicated that R-17F bound selectively to LNFP I (Fucα1-2Galß1-3GlcNAcß1-3Galß1-4Glc). A critical role of the terminal Fucα1-2 residue was confirmed by the selective disappearance of R-17F binding to the purified antigen upon α1-2 fucosidase digestion. Most interestingly, R-17F, when added to hiPS/ES cell suspensions, exhibited potent dose-dependent cytotoxicity. The cytotoxic effect was augmented markedly upon the addition of the secondary antibody (goat anti-mouse IgG1 antibody). R-17F may be beneficial for safer regenerative medicine by eliminating residual undifferentiated hiPS cells in hiPS-derived regenerative tissues, which are considered to be a strong risk factor for carcinogenesis.


Subject(s)
Antibodies/immunology , Cytotoxicity, Immunologic , Induced Pluripotent Stem Cells/metabolism , Oligosaccharides/immunology , Carbohydrate Sequence , Cell Line , Humans , Molecular Sequence Data , Oligosaccharides/chemistry
13.
Stem Cells Transl Med ; 4(7): 720-30, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25972146

ABSTRACT

UNLABELLED: : Cell growth is an important criterion for determining healthy cell conditions. When somatic cells or cancer cells are dissociated into single cells for passaging, the cell numbers can be counted at each passage, providing information on cell growth as an indicator of the health conditions of these cells. In the case of human pluripotent stem cells (hPSCs), because the cells are usually dissociated into cell clumps of ∼50-100 cells for passaging, cell counting is time-consuming. In the present study, using a time-lapse imaging system, we developed a method to determine the growth of hPSCs from nonlabeled live cell phase-contrast images without damaging these cells. Next, the hPSC colony areas and number of nuclei were determined and used to derive equations to calculate the cell number in hPSC colonies, which were assessed on time-lapse images acquired using a culture observation system. The relationships between the colony areas and nuclei numbers were linear, although the equation coefficients were dependent on the cell line used, colony size, colony morphology, and culture conditions. When the culture conditions became improper, the change in cell growth conditions could be detected by analysis of the phase-contrast images. This method provided real-time information on colony growth and cell growth rates without using treatments that can damage cells and could be useful for basic research on hPSCs and cell processing for hPSC-based therapy. SIGNIFICANCE: This is the first study to use a noninvasive method using images to systemically determine the growth of human pluripotent stem cells (hPSCs) without damaging or wasting cells. This method would be useful for quality control during cell culture of clinical hPSCs.

14.
In Vitro Cell Dev Biol Anim ; 51(8): 769-75, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25898826

ABSTRACT

Neural differentiation is an important target of human embryonic stem cells, which provide a source for cell-based therapy, developmental biology, and pharmaceutical research. Previous studies revealed that inhibition of the bone morphogenetic protein is required for neural induction from human embryonic stem cells. On the contrary, the functions of fibroblast growth factors and Activin/Nodal signaling are controversial. Fibroblast growth factor-2 and Activin/Nodal pathways exert divergent influences on human embryonic stem cell concerning the maintenance of both pluripotency and cellular differentiation. We hypothesized that the combination of fibroblast growth factor-2 and Activin A at various concentrations synergistically exerts diverse effects on cell differentiation. To determine the effects of fibroblast growth factor-2 and Activin A on cellular differentiation into neural lineages, we examined the expression of neural differentiation markers in human embryonic stem cells treated with fibroblast growth factor-2 and/or Activin A at various concentrations in a growth factor-defined serum-free medium in short-term culture. In this study, we provide evidence that fibroblast growth factor-2 and Activin A synergistically regulated the initiation of human embryonic stem cell differentiation into neural cell lineages even though human embryonic stem cells autonomously differentiate into neural cell lineages.


Subject(s)
Activins/pharmacology , Cell Differentiation/drug effects , Fibroblast Growth Factor 2/pharmacology , Pluripotent Stem Cells/drug effects , Cell Differentiation/physiology , Cell Lineage/drug effects , Cell Lineage/physiology , Dose-Response Relationship, Drug , Drug Synergism , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/physiology , Humans , Neurons/cytology , Neurons/drug effects , Neurons/physiology , Pluripotent Stem Cells/physiology
15.
Sci Rep ; 4: 4646, 2014 Apr 11.
Article in English | MEDLINE | ID: mdl-24721898

ABSTRACT

Enzymes used for passaging human pluripotent stem cells (hPSCs) digest cell surface proteins, resulting in cell damage. Moreover, cell dissociation using divalent cation-free solutions causes apoptosis. Here we report that Mg(2+) and Ca(2+) control cell-fibronectin and cell-cell binding of hPSCs, respectively, under feeder- and serum-free culture conditions without enzyme. The hPSCs were detached from fibronectin-, vitronectin- or laminin-coated dishes in low concentrations of Mg(2+) and remained as large colonies in high concentrations of Ca(2+). Using enzyme-free solutions containing Ca(2+) without Mg(2+), we successfully passaged hPSCs as large cell clumps that showed less damage than cells passaged using a divalent cation-free solution or dispase. Under the same conditions, the undifferentiated and early-differentiated cells could also be harvested as a cell sheet without being split off. Our enzyme-free passage of hPSCs under a serum- and feeder-free culture condition reduces cell damage and facilitates easier and safer cultures of hPSCs.


Subject(s)
Cations, Divalent/metabolism , Pluripotent Stem Cells/cytology , Calcium/chemistry , Calcium/metabolism , Cations, Divalent/chemistry , Cell Differentiation , Cell Line , Cell Proliferation , Endopeptidases/metabolism , Fibronectins/metabolism , Humans , Karyotyping , Magnesium/chemistry , Magnesium/metabolism , Pluripotent Stem Cells/metabolism
16.
Stem Cells Dev ; 23(18): 2180-9, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-24410631

ABSTRACT

Epithelial-mesenchymal transition (EMT) has been thought to occur during early embryogenesis, and also the differentiation process of human embryonic stem (hES) cells. Spontaneous differentiation is sometimes observed at the peripheral of the hES cell colonies in conventional culture conditions, indicating that EMT occurs in hES cell culture. However, the triggering mechanism of EMT is not yet fully understood. The balance between self-renewal and differentiation of human pluripotent stem (hPS) cells is controlled by various signal pathways, including the fibroblast growth factor (FGF)-2. However, FGF-2 has a complex role for self-renewal of hES cells. FGF-2 activates phosphatidylinositol-3 kinase/AKT, mitogen-activated protein kinase/extracellular signal-regulated kinase-1/2 kinase, and also protein kinase C (PKC). Here, we showed that a PKC rapidly induced an early growth response protein-1 (EGR-1) in hES cells, which was followed by upregulation of EMT-related genes. Before the induction of EMT-related genes, EGR-1 was translocated into the nucleus, and then bound directly to the promoter region of SNAIL, which is a master regulator of EMT. SNAIL expression was attenuated by knockdown of EGR-1, but upregulated by ectopic expression of EGR-1. EGR-1 as the downstream signal of PKC might play a key role in EMT initiation during early differentiation of hES cells. This study would lead to a more robust understanding of the mechanisms underlying the balance between self-renewal and initiation of differentiation in hPS cells.


Subject(s)
Early Growth Response Protein 1/metabolism , Embryonic Stem Cells/metabolism , Epithelial-Mesenchymal Transition , Promoter Regions, Genetic , Protein Kinase C/metabolism , Transcription Factors/genetics , Cell Line , Embryonic Stem Cells/drug effects , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation/drug effects , Humans , Protein Binding/drug effects , Snail Family Transcription Factors , Tetradecanoylphorbol Acetate/pharmacology , Transcription Factors/metabolism
17.
Int J Dev Biol ; 57(9-10): 715-24, 2013.
Article in English | MEDLINE | ID: mdl-24307297

ABSTRACT

Mouse embryonic stem (mES) cells and mouse induced pluripotent stem (miPS) cells are commonly maintained on inactivated mouse embryonic fibroblast feeder cells in medium supplemented with fetal bovine serum or proprietary replacements. An undefined medium containing unknown quantities of reagents has limited the development of applications for pluripotent cells because of the relative lack of knowledge regarding cell responses to differentiating growth factors. Therefore we developed a serum-free medium, designated ESF7, in which mES cells can be maintained in an undifferentiated state without feeder cells. The medium was tested for culturing miPS cells. The miPS cells have been maintained in ESF7 medium for more than 3 years with an undifferentiated phenotype manifested by the expression of pluripotency marker genes and alkaline phosphatase, and these cells exhibited largely normal karyotypes. Furthermore, we found that fibroblast growth factor-2 (FGF-2) with heparin induced miPS cell differentiation into neuronal cells, both in an adherent monolayer and in embryoid body suspension culture. Moreover, we found that FGF-2 with bone morphogenetic protein 2 induced miPS cell differentiation into cardiomyocytes in embryoid body suspension culture. Furthermore, we transplanted subcutaneously miPS cells maintained in ESF7 into the dorsal flanks of SCID mice; all of the transplants produced tumors with tissues derived from all three embryonic germ layers. As this simple serum-free adherent monoculture system supports the long-term propagation of pluripotent iPS cells in vitro, it will allow us to elucidate cell responses to growth factors under defined conditions, and it should provide useful information for differentiation protocols for human iPS cells.


Subject(s)
Cell Culture Techniques , Cell Transformation, Neoplastic/drug effects , Embryonic Stem Cells/drug effects , Induced Pluripotent Stem Cells/drug effects , Muscle Development/drug effects , Neurogenesis/drug effects , Animals , Cell Proliferation , Cells, Cultured , Culture Media, Serum-Free , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Fibroblast Growth Factor 2/pharmacology , Heparin/pharmacology , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Mice , Mice, SCID , Myocytes, Cardiac/metabolism , Neurons/metabolism
18.
PLoS One ; 8(1): e54122, 2013.
Article in English | MEDLINE | ID: mdl-23349801

ABSTRACT

BACKGROUND: The self-renewal of human pluripotent stem (hPS) cells including embryonic stem and induced pluripotent stem cells have been reported to be supported by various signal pathways. Among them, fibroblast growth factor-2 (FGF-2) appears indispensable to maintain self-renewal of hPS cells. However, downstream signaling of FGF-2 has not yet been clearly understood in hPS cells. METHODOLOGY/PRINCIPAL FINDINGS: In this study, we screened a kinase inhibitor library using a high-throughput alkaline phosphatase (ALP) activity-based assay in a minimal growth factor-defined medium to understand FGF-2-related molecular mechanisms regulating self-renewal of hPS cells. We found that in the presence of FGF-2, an inhibitor of protein kinase C (PKC), GF109203X (GFX), increased ALP activity. GFX inhibited FGF-2-induced phosphorylation of glycogen synthase kinase-3ß (GSK-3ß), suggesting that FGF-2 induced PKC and then PKC inhibited the activity of GSK-3ß. Addition of activin A increased phosphorylation of GSK-3ß and extracellular signal-regulated kinase-1/2 (ERK-1/2) synergistically with FGF-2 whereas activin A alone did not. GFX negated differentiation of hPS cells induced by the PKC activator, phorbol 12-myristate 13-acetate whereas Gö6976, a selective inhibitor of PKCα, ß, and γ isoforms could not counteract the effect of PMA. Intriguingly, functional gene analysis by RNA interference revealed that the phosphorylation of GSK-3ß was reduced by siRNA of PKCδ, PKCε, and ζ, the phosphorylation of ERK-1/2 was reduced by siRNA of PKCε and ζ, and the phosphorylation of AKT was reduced by PKCε in hPS cells. CONCLUSIONS/SIGNIFICANCE: Our study suggested complicated cross-talk in hPS cells that FGF-2 induced the phosphorylation of phosphatidylinositol-3 kinase (PI3K)/AKT, mitogen-activated protein kinase/ERK-1/2 kinase (MEK), PKC/ERK-1/2 kinase, and PKC/GSK-3ß. Addition of GFX with a MEK inhibitor, U0126, in the presence of FGF-2 and activin A provided a long-term stable undifferentiated state of hPS cells even though hPS cells were dissociated into single cells for passage. This study untangles the cross-talk between molecular mechanisms regulating self-renewal and differentiation of hPS cells.


Subject(s)
Cell Proliferation , Pluripotent Stem Cells/metabolism , Protein Kinase C-delta/metabolism , Protein Kinase C-epsilon/metabolism , Protein Kinase C/metabolism , Activins/pharmacology , Alkaline Phosphatase/metabolism , Blotting, Western , Carbazoles/pharmacology , Cell Line , Chromones/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblast Growth Factor 2/pharmacology , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Immunohistochemistry , Indoles/pharmacology , Maleimides/pharmacology , Morpholines/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Pluripotent Stem Cells/cytology , Protein Kinase C/genetics , Protein Kinase C-delta/genetics , Protein Kinase C-epsilon/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Signal Transduction/drug effects , Tetradecanoylphorbol Acetate/pharmacology
19.
Int J Dev Biol ; 55(2): 181-7, 2011.
Article in English | MEDLINE | ID: mdl-21305471

ABSTRACT

Human bone marrow-derived mesenchymal stem cells (hMSCs) are potential cellular sources of therapeutic stem cells as they have the ability to proliferate and differentiate into a wide array of mesenchymal cell types such as osteoblasts, chondroblasts and adipocytes. hMSCs have been used clinically to treat patients with graft vs. host disease, osteogenesis imperfect, or alveolar cleft, suggesting that transplantation of hMSCs is comparatively safe as a stem cell-based therapy. However, conventional culture medium for hMSCs contains fetal bovine serum (FBS). In the present study, we developed a growth factor-defined, serum-free medium for culturing hMSCs. Under these conditions, TGF-beta1 promoted proliferation of hMSCs. The expanded hMSC population expressed the human pluripotency markers SSEA-3, -4, NANOG, OCT3/4 and SOX2. Furthermore, double positive cells for SSEA-3 and a mesenchymal cell marker, CD105, were detected in the population. The potential to differentiate into osteoblasts and adipocytes was confirmed. This work provides a useful tool to understand the basic biological properties of hMSCs in culture.


Subject(s)
Cell Proliferation , Intercellular Signaling Peptides and Proteins/pharmacology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Pluripotent Stem Cells/metabolism , Tissue Culture Techniques , Antigens, CD/biosynthesis , Antigens, Tumor-Associated, Carbohydrate/biosynthesis , Cell Differentiation , Cells, Cultured , Culture Media, Serum-Free , Endoglin , Homeodomain Proteins/biosynthesis , Humans , Nanog Homeobox Protein , Octamer Transcription Factor-3/biosynthesis , Receptors, Cell Surface/biosynthesis , SOXB1 Transcription Factors/biosynthesis , Stage-Specific Embryonic Antigens/biosynthesis , Transforming Growth Factor beta/pharmacology
20.
Mol Ther ; 19(2): 400-7, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21102561

ABSTRACT

Human embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) have the potential to differentiate into all cell lineages, including hepatocytes, in vitro. Induced hepatocytes have a wide range of potential application in biomedical research, drug discovery, and the treatment of liver disease. However, the existing protocols for hepatic differentiation of PSCs are not very efficient. In this study, we developed an efficient method to induce hepatoblasts, which are progenitors of hepatocytes, from human ESCs and iPSCs by overexpression of the HEX gene, which is a homeotic gene and also essential for hepatic differentiation, using a HEX-expressing adenovirus (Ad) vector under serum/feeder cell-free chemically defined conditions. Ad-HEX-transduced cells expressed α-fetoprotein (AFP) at day 9 and then expressed albumin (ALB) at day 12. Furthermore, the Ad-HEX-transduced cells derived from human iPSCs also produced several cytochrome P450 (CYP) isozymes, and these P450 isozymes were capable of converting the substrates to metabolites and responding to the chemical stimulation. Our differentiation protocol using Ad vector-mediated transient HEX transduction under chemically defined conditions efficiently generates hepatoblasts from human ESCs and iPSCs. Thus, our methods would be useful for not only drug screening but also therapeutic applications.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Genes, Homeobox/physiology , Hepatocytes/cytology , Homeodomain Proteins/physiology , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Transcription Factors/physiology , Adenoviridae/genetics , Cell Differentiation/genetics , Cell Differentiation/physiology , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Genes, Homeobox/genetics , Genetic Vectors/genetics , Homeodomain Proteins/genetics , Humans , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...