Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Nat Protoc ; 19(2): 565-594, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38087082

ABSTRACT

To produce abundant cell culture samples to generate large, standardized image datasets of human induced pluripotent stem (hiPS) cells, we developed an automated workflow on a Hamilton STAR liquid handler system. This was developed specifically for culturing hiPS cell lines expressing fluorescently tagged proteins, which we have used to study the principles by which cells establish and maintain robust dynamic localization of cellular structures. This protocol includes all details for the maintenance, passage and seeding of cells, as well as Matrigel coating of 6-well plastic plates and 96-well optical-grade, glass plates. We also developed an automated image-based hiPS cell colony segmentation and feature extraction pipeline to streamline the process of predicting cell count and selecting wells with consistent morphology for high-resolution three-dimensional (3D) microscopy. The imaging samples produced with this protocol have been used to study the integrated intracellular organization and cell-to-cell variability of hiPS cells to train and develop deep learning-based label-free predictions from transmitted-light microscopy images and to develop deep learning-based generative models of single-cell organization. This protocol requires some experience with robotic equipment. However, we provide details and source code to facilitate implementation by biologists less experienced with robotics. The protocol is completed in less than 10 h with minimal human interaction. Overall, automation of our cell culture procedures increased our imaging samples' standardization, reproducibility, scalability and consistency. It also reduced the need for stringent culturist training and eliminated culturist-to-culturist variability, both of which were previous pain points of our original manual pipeline workflow.


Subject(s)
Induced Pluripotent Stem Cells , Humans , Microscopy , Reproducibility of Results , Cell Culture Techniques/methods , Automation
2.
Sci Rep ; 11(1): 15845, 2021 08 04.
Article in English | MEDLINE | ID: mdl-34349150

ABSTRACT

We performed a comprehensive analysis of the transcriptional changes occurring during human induced pluripotent stem cell (hiPSC) differentiation to cardiomyocytes. Using single cell RNA-seq, we sequenced > 20,000 single cells from 55 independent samples representing two differentiation protocols and multiple hiPSC lines. Samples included experimental replicates ranging from undifferentiated hiPSCs to mixed populations of cells at D90 post-differentiation. Differentiated cell populations clustered by time point, with differential expression analysis revealing markers of cardiomyocyte differentiation and maturation changing from D12 to D90. We next performed a complementary cluster-independent sparse regression analysis to identify and rank genes that best assigned cells to differentiation time points. The two highest ranked genes between D12 and D24 (MYH7 and MYH6) resulted in an accuracy of 0.84, and the three highest ranked genes between D24 and D90 (A2M, H19, IGF2) resulted in an accuracy of 0.94, revealing that low dimensional gene features can identify differentiation or maturation stages in differentiating cardiomyocytes. Expression levels of select genes were validated using RNA FISH. Finally, we interrogated differences in cardiac gene expression resulting from two differentiation protocols, experimental replicates, and three hiPSC lines in the WTC-11 background to identify sources of variation across these experimental variables.


Subject(s)
Biomarkers/metabolism , Cell Differentiation , Gene Expression Regulation , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Transcriptome , Humans , Induced Pluripotent Stem Cells/cytology , RNA-Seq
3.
Cell Syst ; 12(6): 670-687.e10, 2021 06 16.
Article in English | MEDLINE | ID: mdl-34043964

ABSTRACT

Although some cell types may be defined anatomically or by physiological function, a rigorous definition of cell state remains elusive. Here, we develop a quantitative, imaging-based platform for the systematic and automated classification of subcellular organization in single cells. We use this platform to quantify subcellular organization and gene expression in >30,000 individual human induced pluripotent stem cell-derived cardiomyocytes, producing a publicly available dataset that describes the population distributions of local and global sarcomere organization, mRNA abundance, and correlations between these traits. While the mRNA abundance of some phenotypically important genes correlates with subcellular organization (e.g., the beta-myosin heavy chain, MYH7), these two cellular metrics are heterogeneous and often uncorrelated, which suggests that gene expression alone is not sufficient to classify cell states. Instead, we posit that cell state should be defined by observing full distributions of quantitative, multidimensional traits in single cells that also account for space, time, and function.


Subject(s)
Induced Pluripotent Stem Cells , Cell Differentiation/genetics , Humans , Myocytes, Cardiac/metabolism , Transcriptome/genetics
4.
Cell Stem Cell ; 20(1): 120-134, 2017 01 05.
Article in English | MEDLINE | ID: mdl-28094016

ABSTRACT

During human brain development, multiple signaling pathways generate diverse cell types with varied regional identities. Here, we integrate single-cell RNA sequencing and clonal analyses to reveal lineage trees and molecular signals underlying early forebrain and mid/hindbrain cell differentiation from human embryonic stem cells (hESCs). Clustering single-cell transcriptomic data identified 41 distinct populations of progenitor, neuronal, and non-neural cells across our differentiation time course. Comparisons with primary mouse and human gene expression data demonstrated rostral and caudal progenitor and neuronal identities from early brain development. Bayesian analyses inferred a unified cell-type lineage tree that bifurcates between cortical and mid/hindbrain cell types. Two methods of clonal analyses confirmed these findings and further revealed the importance of Wnt/ß-catenin signaling in controlling this lineage decision. Together, these findings provide a rich transcriptome-based lineage map for studying human brain development and modeling developmental disorders.


Subject(s)
Brain/embryology , Cell Lineage , Embryonic Development , Human Embryonic Stem Cells/cytology , Single-Cell Analysis/methods , Animals , Brain/metabolism , Cell Line , Cell Lineage/genetics , Clone Cells , Embryonic Development/genetics , Humans , Mice , Models, Biological , Neurons/cytology , Neurons/metabolism , Reproducibility of Results , Sequence Analysis, RNA , Transcription Factors/metabolism , Transcriptome/genetics , Wnt Signaling Pathway/genetics
5.
Nat Methods ; 13(1): 87-93, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26524239

ABSTRACT

The diverse progenitors that give rise to the human neocortex have been difficult to characterize because progenitors, particularly radial glia (RG), are rare and are defined by a combination of intracellular markers, position and morphology. To circumvent these problems, we developed Fixed and Recovered Intact Single-cell RNA (FRISCR), a method for profiling the transcriptomes of individual fixed, stained and sorted cells. Using FRISCR, we profiled primary human RG that constitute only 1% of the midgestation cortex and classified them as ventricular zone-enriched RG (vRG) that express ANXA1 and CRYAB, and outer subventricular zone-localized RG (oRG) that express HOPX. Our study identified vRG and oRG markers and molecular profiles, an essential step for understanding human neocortical progenitor development. FRISCR allows targeted single-cell profiling of any tissues that lack live-cell markers.


Subject(s)
Brain/cytology , Neuroglia/cytology , Transcriptome , Humans , Single-Cell Analysis
6.
Nucleic Acids Res ; 43(10): e65, 2015 May 26.
Article in English | MEDLINE | ID: mdl-25765640

ABSTRACT

Isogenic pluripotent stem cells are critical tools for studying human neurological diseases by allowing one to study the effects of a mutation in a fixed genetic background. Of particular interest are the spectrum of autism disorders, some of which are monogenic such as Timothy syndrome (TS); others are multigenic such as the microdeletion and microduplication syndromes of the 16p11.2 chromosomal locus. Here, we report engineered human embryonic stem cell (hESC) lines for modeling these two disorders using locus-specific endonucleases to increase the efficiency of homology-directed repair (HDR). We developed a system to: (1) computationally identify unique transcription activator-like effector nuclease (TALEN) binding sites in the genome using a new software program, TALENSeek, (2) assemble the TALEN genes by combining golden gate cloning with modified constructs from the FLASH protocol, and (3) test the TALEN pairs in an amplification-based HDR assay that is more sensitive than the typical non-homologous end joining assay. We applied these methods to identify, construct, and test TALENs that were used with HDR donors in hESCs to generate an isogenic TS cell line in a scarless manner and to model the 16p11.2 copy number disorder without modifying genomic loci with high sequence similarity.


Subject(s)
Cell Engineering , Child Development Disorders, Pervasive/genetics , Embryonic Stem Cells , Models, Genetic , Autistic Disorder , Binding Sites , Cell Line , Deoxyribonucleases/metabolism , Gene Targeting , Genome, Human , Humans , Long QT Syndrome/genetics , Recombinational DNA Repair , Software , Syndactyly/genetics
7.
Proc Natl Acad Sci U S A ; 111(12): 4484-9, 2014 Mar 25.
Article in English | MEDLINE | ID: mdl-24623855

ABSTRACT

The naïve pluripotent state has been shown in mice to lead to broad and more robust developmental potential relative to primed mouse epiblast cells. The human naïve ES cell state has eluded derivation without the use of transgenes, and forced expression of OCT4, KLF4, and KLF2 allows maintenance of human cells in a naïve state [Hanna J, et al. (2010) Proc Natl Acad Sci USA 107(20):9222-9227]. We describe two routes to generate nontransgenic naïve human ES cells (hESCs). The first is by reverse toggling of preexisting primed hESC lines by preculture in the histone deacetylase inhibitors butyrate and suberoylanilide hydroxamic acid, followed by culture in MEK/ERK and GSK3 inhibitors (2i) with FGF2. The second route is by direct derivation from a human embryo in 2i with FGF2. We show that human naïve cells meet mouse criteria for the naïve state by growth characteristics, antibody labeling profile, gene expression, X-inactivation profile, mitochondrial morphology, microRNA profile and development in the context of teratomas. hESCs can exist in a naïve state without the need for transgenes. Direct derivation is an elusive, but attainable, process, leading to cells at the earliest stage of in vitro pluripotency described for humans. Reverse toggling of primed cells to naïve is efficient and reproducible.


Subject(s)
Embryonic Stem Cells/cytology , Animals , Cell Lineage , Cells, Cultured , Embryonic Stem Cells/metabolism , Gene Expression Profiling , Glycogen Synthase Kinase 3/antagonists & inhibitors , Histone Deacetylase Inhibitors/pharmacology , Humans , Kruppel-Like Factor 4 , Mice , Protein Kinase Inhibitors/pharmacology , Transgenes , X Chromosome Inactivation
8.
Prostate ; 72(13): 1453-63, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22314551

ABSTRACT

BACKGROUND: CD90(+) prostate cancer-associated (CP) stromal cells represent a diseased cell type found only in tumor tissue. They differ from their normal counterpart in gene expression and inductive signaling. Genetic reprogramming by induced pluripotent stem (iPS) cell technology can effectively change adult cells into stem-like cells through wholesale alteration of the gene expression program. This technology might be used to 'erase' the abnormal gene expression of diseased cells. The resultant iPS cells would no longer express the disease phenotype, and behave like stem cells. METHODS: CP stromal cells, isolated from tumor tissue of a surgically resected prostate by anti-CD90-mediated sorting and cultured in vitro, were transfected with in vitro packaged lentiviral expression vectors containing stem cell transcription factor genes POU5F1, LIN28, NANOG, and SOX2. RESULTS: Alkaline phosphatase-positive iPS cells were obtained in about 3 weeks post-transfection at a frequency of 10(-4) . Their colony morphology was indistinguishable from that of human embryonic stem (ES) cells. Transcriptome analysis showed a virtually complete match in gene expression between the iPS and ES cells. CONCLUSIONS: Genes of CP stromal cells could be fully inactivated by genetic reprogramming. As a consequence, the disease phenotype was 'cured'.


Subject(s)
Embryonic Stem Cells/cytology , Induced Pluripotent Stem Cells/cytology , Prostatic Neoplasms/pathology , Stromal Cells/pathology , Embryonic Stem Cells/metabolism , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Male , Nanog Homeobox Protein , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Stromal Cells/metabolism , Transcriptome
9.
PLoS Genet ; 6(10): e1001181, 2010 Oct 28.
Article in English | MEDLINE | ID: mdl-21060811

ABSTRACT

Each unit of the D4Z4 macrosatellite repeat contains a retrotransposed gene encoding the DUX4 double-homeobox transcription factor. Facioscapulohumeral dystrophy (FSHD) is caused by deletion of a subset of the D4Z4 units in the subtelomeric region of chromosome 4. Although it has been reported that the deletion of D4Z4 units induces the pathological expression of DUX4 mRNA, the association of DUX4 mRNA expression with FSHD has not been rigorously investigated, nor has any human tissue been identified that normally expresses DUX4 mRNA or protein. We show that FSHD muscle expresses a different splice form of DUX4 mRNA compared to control muscle. Control muscle produces low amounts of a splice form of DUX4 encoding only the amino-terminal portion of DUX4. FSHD muscle produces low amounts of a DUX4 mRNA that encodes the full-length DUX4 protein. The low abundance of full-length DUX4 mRNA in FSHD muscle cells represents a small subset of nuclei producing a relatively high abundance of DUX4 mRNA and protein. In contrast to control skeletal muscle and most other somatic tissues, full-length DUX4 transcript and protein is expressed at relatively abundant levels in human testis, most likely in the germ-line cells. Induced pluripotent (iPS) cells also express full-length DUX4 and differentiation of control iPS cells to embryoid bodies suppresses expression of full-length DUX4, whereas expression of full-length DUX4 persists in differentiated FSHD iPS cells. Together, these findings indicate that full-length DUX4 is normally expressed at specific developmental stages and is suppressed in most somatic tissues. The contraction of the D4Z4 repeat in FSHD results in a less efficient suppression of the full-length DUX4 mRNA in skeletal muscle cells. Therefore, FSHD represents the first human disease to be associated with the incomplete developmental silencing of a retrogene array normally expressed early in development.


Subject(s)
Gene Expression Profiling , Homeodomain Proteins/genetics , Muscular Dystrophy, Facioscapulohumeral/genetics , RNA Splicing , Adult , Animals , Blotting, Western , Cell Line , Chromosomes, Human, Pair 4/genetics , Female , Fluorescent Antibody Technique , Gene Expression Regulation , HCT116 Cells , Humans , Induced Pluripotent Stem Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, SCID , Molecular Sequence Data , Muscle Cells/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Facioscapulohumeral/pathology , Repetitive Sequences, Nucleic Acid/genetics , Retroelements/genetics , Reverse Transcriptase Polymerase Chain Reaction
10.
Cell Stem Cell ; 4(4): 359-69, 2009 Apr 03.
Article in English | MEDLINE | ID: mdl-19341625

ABSTRACT

Recent evidence indicates that mouse and human embryonic stem cells (ESCs) are fixed at different developmental stages, with the former positioned earlier. We show that a narrow concentration of the naturally occurring short-chain fatty acid, sodium butyrate, supports the extensive self-renewal of mouse and human ESCs, while promoting their convergence toward an intermediate stem cell state. In response to butyrate, human ESCs regress to an earlier developmental stage characterized by a gene expression profile resembling that of mouse ESCs, preventing precocious Xist expression while retaining the ability to form complex teratomas in vivo. Other histone deacetylase inhibitors (HDACi) also support human ESC self-renewal. Our results indicate that HDACi can promote ESC self-renewal across species, and demonstrate that ESCs can toggle between alternative states in response to environmental factors.


Subject(s)
Butyrates/pharmacology , Cell Differentiation , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/physiology , Histone Deacetylase Inhibitors , Animals , Embryonic Stem Cells/enzymology , Enzyme Inhibitors/pharmacology , Gene Expression Profiling , Histone Deacetylases/metabolism , Humans , Mice , RNA, Long Noncoding , RNA, Untranslated/metabolism
11.
Exp Cell Res ; 314(16): 2930-40, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18692044

ABSTRACT

Despite a growing body of literature concerning the hematopoietic differentiation of human embryonic stem cells (hESCs), the full hematopoietic potential of the majority of existing hESC lines remains unknown. In this study, the hematopoietic response of five NIH-approved hESC lines (H1, hSF6, BG01, BG02, and BG03) was compared. Our data show that despite expressing similar hESC markers under self-renewing conditions and initiating mesodermal differentiation under spontaneous differentiation conditions, marked differences in subsequent hematopoietic differentiation potential among these lines existed. A high degree of hematopoietic differentiation was attained only by H1 and BG02, whereas this process appeared to be abortive in nature for hSF6, BG01, and BG03. This difference in hematopoietic differentiation predisposition was readily apparent during spontaneous differentiation, and further augmented under hematopoietic-inducing conditions. This predisposition appeared to be intrinsic to the specific hESC line and independent of passage number or gender karyotype. Interestingly, H1 and BG02 displayed remarkable similarities in their kinetics of hematopoietic marker expression, hematopoietic colony formation, erythroid differentiation, and globin expression, suggesting that a similar, predetermined differentiation sequence is followed. The identification of intrinsic and extrinsic factors governing the hematopoietic differentiation potential of hESCs will be of great importance for the putative clinical utility of hESC lines.


Subject(s)
Cell Differentiation/physiology , Embryonic Stem Cells/physiology , Hematopoiesis/physiology , Hematopoietic Stem Cells/physiology , Animals , Biomarkers/metabolism , Cell Line , Cell Lineage , Coculture Techniques , Embryonic Stem Cells/cytology , GATA1 Transcription Factor/genetics , GATA1 Transcription Factor/metabolism , Gene Expression Profiling , Hematopoietic Stem Cells/cytology , Humans , Mice , Phenotype
12.
Stem Cells ; 26(10): 2496-505, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18583537

ABSTRACT

We used massively parallel pyrosequencing to discover and characterize microRNAs (miRNAs) expressed in human embryonic stem cells (hESC). Sequencing of small RNA cDNA libraries derived from undifferentiated hESC and from isogenic differentiating cultures yielded a total of 425,505 high-quality sequence reads. A custom data analysis pipeline delineated expression profiles for 191 previously annotated miRNAs, 13 novel miRNAs, and 56 candidate miRNAs. Further characterization of a subset of the novel miRNAs in Dicer-knockdown hESC demonstrated Dicer-dependent expression, providing additional validation of our results. A set of 14 miRNAs (9 known and 5 novel) was noted to be expressed in undifferentiated hESC and then strongly downregulated with differentiation. Functional annotation analysis of predicted targets of these miRNAs and comparison with a null model using non-hESC-expressed miRNAs identified statistically enriched functional categories, including chromatin remodeling and lineage-specific differentiation annotations. Finally, integration of our data with genome-wide chromatin immunoprecipitation data on OCT4, SOX2, and NANOG binding sites implicates these transcription factors in the regulation of nine of the novel/candidate miRNAs identified here. Comparison of our results with those of recent deep sequencing studies in mouse and human ESC shows that most of the novel/candidate miRNAs found here were not identified in the other studies. The data indicate that hESC express a larger complement of miRNAs than previously appreciated, and they provide a resource for additional studies of miRNA regulation of hESC physiology. Disclosure of potential conflicts of interest is found at the end of this article.


Subject(s)
Embryonic Stem Cells/metabolism , Gene Expression Profiling , Gene Library , MicroRNAs/genetics , Sequence Analysis, RNA , Base Sequence , Cell Differentiation , Cell Line , Databases, Genetic , Embryonic Stem Cells/cytology , Embryonic Stem Cells/enzymology , Expressed Sequence Tags , Gene Expression Regulation, Developmental , Humans , MicroRNAs/chemistry , Molecular Sequence Data , Multipotent Stem Cells/cytology , Multipotent Stem Cells/metabolism , Nucleic Acid Conformation , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , RNA, Small Interfering/chemistry , RNA, Small Interfering/genetics , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction , Ribonuclease III/metabolism , Transcription Factors/metabolism
13.
Blood ; 110(12): 4111-9, 2007 Dec 01.
Article in English | MEDLINE | ID: mdl-17761519

ABSTRACT

Despite progress in developing defined conditions for human embryonic stem cell (hESC) cultures, little is known about the cell-surface receptors that are activated under conditions supportive of hESC self-renewal. A simultaneous interrogation of 42 receptor tyrosine kinases (RTKs) in hESCs following stimulation with mouse embryonic fibroblast (MEF) conditioned medium (CM) revealed rapid and prominent tyrosine phosphorylation of insulin receptor (IR) and insulin-like growth factor-1 receptor (IGF1R); less prominent tyrosine phosphorylation of epidermal growth factor receptor (EGFR) family members, including ERBB2 and ERBB3; and trace phosphorylation of fibroblast growth factor receptors. Intense IGF1R and IR phosphorylation occurred in the absence of MEF conditioning (NCM) and was attributable to high concentrations of insulin in the proprietary KnockOut Serum Replacer (KSR). Inhibition of IGF1R using a blocking antibody or lentivirus-delivered shRNA reduced hESC self-renewal and promoted differentiation, while disruption of ERBB2 signaling with the selective inhibitor AG825 severely inhibited hESC proliferation and promoted apoptosis. A simple defined medium containing an IGF1 analog, heregulin-1beta (a ligand for ERBB2/ERBB3), fibroblast growth factor-2 (FGF2), and activin A supported long-term growth of multiple hESC lines. These studies identify previously unappreciated RTKs that support hESC proliferation and self-renewal, and provide a rationally designed medium for the growth and maintenance of pluripotent hESCs.


Subject(s)
Cell Proliferation , Embryonic Stem Cells/metabolism , Pluripotent Stem Cells/metabolism , Receptor, ErbB-2/metabolism , Receptor, IGF Type 2/metabolism , Signal Transduction/physiology , Animals , Antibodies, Monoclonal/pharmacology , Apoptosis/drug effects , Apoptosis/physiology , Benzothiazoles/pharmacology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Line , Cell Proliferation/drug effects , Culture Media, Conditioned , Embryonic Stem Cells/cytology , Fibroblast Growth Factor 2/pharmacology , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Mice , Neuregulin-1/pharmacology , Phosphorylation/drug effects , Pluripotent Stem Cells/cytology , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-3/antagonists & inhibitors , Receptor, ErbB-3/metabolism , Receptor, IGF Type 2/antagonists & inhibitors , Receptor, Insulin/antagonists & inhibitors , Receptor, Insulin/metabolism , Signal Transduction/drug effects , Tyrphostins/pharmacology
14.
Stem Cells ; 24(12): 2677-84, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16916927

ABSTRACT

In October 2003, the NIH established three extramural "Exploratory Centers for Human Embryonic Stem Cell Research." Our center acquired 15 of the 22 NIH-approved cell lines. Lines were tested for: (a) freedom from mycoplasma contamination; (b) appropriate pattern of gene expression during self-renewal and differentiation; (c) ability to adapt to uniform culture conditions; (d) ability to grow at clonal densities; (e) karyotype; (f) growth efficiency; and (g) efficiency of stable transfection following electroporation. One line harbored mycoplasma. Ten lines were converted to uniform conditions. Nine lines were fully characterized. Human ESC (hESC) lines varied markedly with respect to growth efficiency as measured by the amount of time it took to plate and double (31-57 hours), cloning efficiency (0.8%-9.2%), and stable transfection rates following electroporation (0%-53% relative to a standard mouse ESC line). One hESC line had an unstable karyotype at an early passage. Modifications of the proposed Material Transfer Agreements with hESC suppliers were required to improve accessibility to hESC lines by local researchers. The NIH-approved hESC lines vary in their behavior in culture. Many hESC lines can be maintained using culture conditions less onerous than those recommended by their suppliers. Intellectual property issues pose a significant obstacle to research using NIH-approved hESC lines.


Subject(s)
Embryonic Stem Cells/cytology , National Institutes of Health (U.S.) , Animals , Biomarkers , Cell Culture Techniques , Cell Differentiation , Cell Survival , Clone Cells , Embryonic Stem Cells/microbiology , Humans , Karyotyping , Mice , Mycoplasma/isolation & purification , Phenotype , Transfection , United States
15.
Blood ; 108(5): 1515-23, 2006 Sep 01.
Article in English | MEDLINE | ID: mdl-16645170

ABSTRACT

Human embryonic stem cells are a promising tool to study events associated with the earliest ontogenetic stages of hematopoiesis. We describe the generation of erythroid cells from hES (H1) by subsequent processing of cells present at early and late stages of embryoid body (EB) differentiation. Kinetics of hematopoietic marker emergence suggest that CD45+ hematopoiesis peaks at late D14EB differentiation stages, although low-level CD45- erythroid differentiation can be seen before that stage. By morphologic criteria, hES-derived erythroid cells were of definitive type, but these cells both at mRNA and protein levels coexpressed high levels of embryonic (epsilon) and fetal (gamma) globins, with little or no adult globin (beta). This globin expression pattern was not altered by the presence or absence of fetal bovine serum, vascular endothelial growth factor, Flt3-L, or coculture with OP-9 during erythroid differentiation and was not culture time dependent. The coexpression of both embryonic and fetal globins by definitive-type erythroid cells does not faithfully mimic either yolk sac embryonic or their fetal liver counterparts. Nevertheless, the high frequency of erythroid cells coexpressing embryonic and fetal globin generated from embryonic stem cells can serve as an invaluable tool to further explore molecular mechanisms.


Subject(s)
Erythrocytes/physiology , Fetal Hemoglobin/physiology , Hematopoiesis/physiology , Stem Cells/cytology , Stem Cells/physiology , Cell Differentiation , DNA Primers , Embryo, Mammalian , Erythrocytes/cytology , Fetal Hemoglobin/genetics , Gene Expression Regulation , Globins/genetics , Humans , Kinetics , Polymerase Chain Reaction , RNA, Messenger/genetics , fms-Like Tyrosine Kinase 3/genetics
16.
Biotechniques ; 38(6): 879-80, 882-3, 2005 Jun.
Article in English | MEDLINE | ID: mdl-16018548

ABSTRACT

A significant obstacle to using human embryonic stem cells (hESCs) arises from extremely poor survival associated with freezing, typically in the range of 1%. This report describes a slow controlled-rate freezing technique commonly used for mammalian embryo cryopreservation. Using a combination of surviving colony number and colony diameter; survival was determined relative to untreated hESCs. Using a dimethyl sulfoxide (DMSO) cryoprotectant and either a homemade controlled-rate freezing device or a commercial freezing device, survival rates of 20%-80% were obtained. To achieve the highest levels of survival, the critical factors were an ice crystal seed (at -7 degrees to -10 degrees C), a freeze rate between 0.3 degrees and 1.8 degrees C/min, and a rapid thaw rate using room temperature water. Slow controlled-rate cooling allows a rapid, simple, and reproducible means of cryopreserving hESCs.


Subject(s)
Embryo, Mammalian/cytology , Freezing , Stem Cells/cytology , Cell Survival , Cryopreservation , Cytoprotection , Dimethyl Sulfoxide , Humans , Karyotyping
17.
Reprod Biol Endocrinol ; 1: 43, 2003 May 16.
Article in English | MEDLINE | ID: mdl-12823859

ABSTRACT

BACKGROUND: Ablation of the low-affinity receptor subunit for leukemia inhibitory factor (LIFR) causes multi-systemic defects in the late gestation fetus. Because corticosterone is known to have a broad range of effects and LIF function has been associated with the hypothalamo-pituitary-adrenal axis, this study was designed to determine the role for LIFR in the fetus when exposed to the elevated maternal glucocorticoid levels of late gestation. Uncovering a requirement for LIFR in appropriate glucocorticoid response will further understanding of control of glucocorticoid function. METHODS: Maternal adrenalectomy or RU486 administration were used to determine the impact of the maternal glucocorticoid surge on fetal development in the absence of LIFR. The mice were analyzed by a variety of histological techniques including immunolabeling and staining techniques (hematoxylin and eosin, Alizarin red S and alcian blue). Plasma corticosterone was assayed using radioimmunoassay. RESULTS: Maternal adrenalectomy does not improve the prognosis for LIFR null pups and exacerbates the effects of LIFR loss. RU486 noticeably improves many of the tissues affected by LIFR loss: bone density, skeletal muscle integrity and glial cell formation. LIFR null pups exposed during late gestation to RU486 in utero survive natural delivery, unlike LIFR null pups from untreated litters. But RU486 treated LIFR null pups succumb within the first day after birth, presumably due to neural deficit resulting in an inability to suckle. CONCLUSION: LIFR plays an integral role in modulating the fetal response to elevated maternal glucocorticoids during late gestation. This role is likely to be mediated through the glucocorticoid receptor and has implications for adult homeostasis as a direct tie between immune, neural and hormone function.


Subject(s)
Abnormalities, Multiple/genetics , Fetus/physiology , Proteins/physiology , Receptors, Cytokine/physiology , Abnormalities, Multiple/embryology , Adrenalectomy , Adrenocorticotropic Hormone/analysis , Animals , Bone Diseases, Metabolic/embryology , Bone Diseases, Metabolic/genetics , Bone Diseases, Metabolic/prevention & control , Corticosterone/blood , Female , Fetal Diseases/embryology , Fetal Diseases/genetics , Fetal Diseases/prevention & control , Genes, Lethal , Gestational Age , Homeostasis , Hormone Antagonists/pharmacology , Hypothalamo-Hypophyseal System/physiology , Interleukin-6 , Leukemia Inhibitory Factor , Leukemia Inhibitory Factor Receptor alpha Subunit , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mifepristone/pharmacology , Muscle, Skeletal/embryology , Muscle, Skeletal/pathology , Neuroglia/drug effects , Neuroimmunomodulation/physiology , Pituitary-Adrenal System/physiology , Pregnancy , Receptors, Cytokine/deficiency , Receptors, Cytokine/genetics , Receptors, Glucocorticoid/physiology , Receptors, OSM-LIF , Specific Pathogen-Free Organisms , Spinal Cord/embryology , Spinal Cord/pathology
18.
Transgenic Res ; 12(6): 743-6, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14713204

ABSTRACT

Inbred ES lines, though useful for generating targeted mutations in mice, are used infrequently. To appreciate the relative efficiency of inbred ES lines, a C57BL/6 ES line was compared with 129 strain ES lines for effectiveness in chimera formation leading to the establishment of targeted mutations in mice. Data from a transgenic facility spanning 7 years were collected. C57BL/6 ES cells injected into Balb/c embryos results in lower coat color chimerism than do 129 ES cells injected into C57BL/6 embryos. Combined data indicate that five independent targeted C57BL/6 clones should be injected as compared to three independent 129 clones to generate enough chimeras to effectively test for germ-line transmission. Thus, although less efficient than 129 ES lines, the C57BL/6 ES line is a relatively competent line and useful for the routine generation of targeted mutations in mice on a defined genetic background.


Subject(s)
Mutation , Stem Cells/cytology , Animals , Cell Culture Techniques/methods , Cells, Cultured , Chimera , Embryo Transfer , Embryo, Mammalian/cytology , Female , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Microinjections
SELECTION OF CITATIONS
SEARCH DETAIL
...