Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Cell Rep ; 10(4): 616-32, 2015 Feb 03.
Article in English | MEDLINE | ID: mdl-25640183

ABSTRACT

Human pluripotent stem cell (hPSC) lines exhibit repeated patterns of genetic variation, which can alter in vitro properties as well as suitability for clinical use. We examined associations between copy-number variations (CNVs) on chromosome 17 and hPSC mesodiencephalic dopaminergic (mDA) differentiation. Among 24 hPSC lines, two karyotypically normal lines, BG03 and CT3, and BG01V2, with trisomy 17, exhibited amplification of the WNT3/WNT9B region and rapid mDA differentiation. In hPSC lines with amplified WNT3/WNT9B, basic fibroblast growth factor (bFGF) signaling through mitogen-activated protein kinase (MAPK)/ERK amplifies canonical WNT signaling by phosphorylating LRP6, resulting in enhanced undifferentiated proliferation. When bFGF is absent, noncanonical WNT signaling becomes dominant due to upregulation of SIAH2, enhancing JNK signaling and promoting loss of pluripotency. When bFGF is present during mDA differentiation, stabilization of canonical WNT signaling causes upregulation of LMX1A and mDA induction. Therefore, CNVs in 17q21.31, a "hot spot" for genetic variation, have multiple and complex effects on hPSC cellular phenotype.


Subject(s)
Neurons/cytology , Neurons/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Wnt Proteins/metabolism , Wnt3 Protein/metabolism , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Signal Transduction , Wnt Proteins/genetics , Wnt3 Protein/genetics
2.
J Vis Exp ; (89)2014 Jul 24.
Article in English | MEDLINE | ID: mdl-25077932

ABSTRACT

Human pluripotent stem cells (hPSCs) hold great promise for regenerative medicine and biopharmaceutical applications. Currently, optimal culture and efficient expansion of large amounts of clinical-grade hPSCs are critical issues in hPSC-based therapies. Conventionally, hPSCs are propagated as colonies on both feeder and feeder-free culture systems. However, these methods have several major limitations, including low cell yields and generation of heterogeneously differentiated cells. To improve current hPSC culture methods, we have recently developed a new method, which is based on non-colony type monolayer (NCM) culture of dissociated single cells. Here, we present detailed NCM protocols based on the Rho-associated kinase (ROCK) inhibitor Y-27632. We also provide new information regarding NCM culture with different small molecules such as Y-39983 (ROCK I inhibitor), phenylbenzodioxane (ROCK II inhibitor), and thiazovivin (a novel ROCK inhibitor). We further extend our basic protocol to cultivate hPSCs on defined extracellular proteins such as the laminin isoform 521 (LN-521) without the use of ROCK inhibitors. Moreover, based on NCM, we have demonstrated efficient transfection or transduction of plasmid DNAs, lentiviral particles, and oligonucleotide-based microRNAs into hPSCs in order to genetically modify these cells for molecular analyses and drug discovery. The NCM-based methods overcome the major shortcomings of colony-type culture, and thus may be suitable for producing large amounts of homogeneous hPSCs for future clinical therapies, stem cell research, and drug discovery.


Subject(s)
Cytological Techniques/methods , Enzyme Inhibitors/pharmacology , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/physiology , rho-Associated Kinases/antagonists & inhibitors , Amides/pharmacology , Animals , Dioxanes/pharmacology , Fibroblasts/cytology , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/physiology , Laminin/pharmacology , Mice , Pluripotent Stem Cells/cytology , Pyridines/pharmacology , Pyrimidines/pharmacology , Thiazoles/pharmacology , Transfection
3.
Stem Cells Transl Med ; 3(7): 867-78, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24855277

ABSTRACT

The ability to differentiate induced pluripotent stem cells (iPSCs) into committed skeletal progenitors could allow for an unlimited autologous supply of such cells for therapeutic uses; therefore, we attempted to create novel bone-forming cells from human iPSCs using lines from two distinct tissue sources and methods of differentiation that we previously devised for osteogenic differentiation of human embryonic stem cells, and as suggested by other publications. The resulting cells were assayed using in vitro methods, and the results were compared with those obtained from in vivo transplantation assays. Our results show that true bone was formed in vivo by derivatives of several iPSC lines, but that the successful cell lines and differentiation methodologies were not predicted by the results of the in vitro assays. In addition, bone was formed equally well from iPSCs originating from skin or bone marrow stromal cells (also known as bone marrow-derived mesenchymal stem cells), suggesting that the iPSCs did not retain a "memory" of their previous life. Furthermore, one of the iPSC-derived cell lines formed verifiable cartilage in vivo, which likewise was not predicted by in vitro assays.


Subject(s)
Biological Assay/methods , Cell Differentiation , Chondrocytes/metabolism , Chondrogenesis , Induced Pluripotent Stem Cells/metabolism , Mesenchymal Stem Cells/metabolism , Osteoblasts/metabolism , Osteogenesis , Aged , Aged, 80 and over , Animals , Cell Line , Cellular Reprogramming , Chondrocytes/transplantation , Female , Gene Expression Regulation, Developmental , Humans , Induced Pluripotent Stem Cells/transplantation , Male , Mesenchymal Stem Cell Transplantation , Mice , Osteoblasts/transplantation , Phenotype , Transfection
4.
Stem Cell Res ; 12(3): 610-21, 2014 May.
Article in English | MEDLINE | ID: mdl-24603366

ABSTRACT

Human pluripotent stem cells (hPSCs) have two potentially attractive applications: cell replacement-based therapies and drug discovery. Both require the efficient generation of large quantities of clinical-grade stem cells that are free from harmful genomic alterations. The currently employed colony-type culture methods often result in low cell yields, unavoidably heterogeneous cell populations, and substantial chromosomal abnormalities. Here, we shed light on the structural relationship between hPSC colonies/embryoid bodies and early-stage embryos in order to optimize current culture methods based on the insights from developmental biology. We further highlight core signaling pathways that underlie multiple epithelial-to-mesenchymal transitions (EMTs), cellular heterogeneity, and chromosomal instability in hPSCs. We also analyze emerging methods such as non-colony type monolayer (NCM) and suspension culture, which provide alternative growth models for hPSC expansion and differentiation. Furthermore, based on the influence of cell-cell interactions and signaling pathways, we propose concepts, strategies, and solutions for production of clinical-grade hPSCs, stem cell precursors, and miniorganoids, which are pivotal steps needed for future clinical applications.


Subject(s)
Cell Differentiation , Cell Proliferation , Mammals/embryology , Pluripotent Stem Cells/cytology , Signal Transduction , Animals , Humans , Mammals/genetics , Mammals/metabolism , Pluripotent Stem Cells/metabolism
5.
Stem Cell Res ; 12(2): 376-86, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24374290

ABSTRACT

Many studies have compared the genetic and epigenetic profiles of human induced pluripotent stem cells (hiPSCs) to human embryonic stem cells (hESCs) and yet the picture remains unclear. To address this, we derived a population of neural precursor cells (NPCs) from the H1 (WA01) hESC line and generated isogenic iPSC lines by reprogramming. The gene expression and methylation profile of three lines were compared to the parental line and intermediate NPC population. We found no gene probe with expression that differed significantly between hESC and iPSC samples under undifferentiated or differentiated conditions. Analysis of the global methylation pattern also showed no significant difference between the two PSC populations. Both undifferentiated populations were distinctly different from the intermediate NPC population in both gene expression and methylation profiles. One point to note is that H1 is a male line and so extrapolation to female lines should be cautioned. However, these data confirm our previous findings that there are no significant differences between hESCs and hiPSCs at the gene expression or methylation level.


Subject(s)
Induced Pluripotent Stem Cells/physiology , Neural Stem Cells/physiology , Animals , Cell Differentiation/physiology , DNA Methylation , Female , Genomic Imprinting , Humans , Induced Pluripotent Stem Cells/metabolism , Male , Membrane Proteins/genetics , Mice , Nerve Tissue Proteins/genetics , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Nitric Oxide/metabolism , Transcriptome
6.
Stem Cell Res ; 10(1): 57-66, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23117585

ABSTRACT

Much of the excitement generated by induced pluripotent stem cell technology is concerned with the possibility of disease modeling as well as the potential for personalized cell therapy. However, to pursue this it is important to understand the 'normal' pluripotent state including its inherent variability. We have performed various molecular profiling assays for 21 hESC lines and 8 hiPSC lines to generate a comprehensive snapshot of the undifferentiated state of pluripotent stem cells. Analysis of the gene expression data revealed no iPSC-specific gene expression pattern in accordance with previous reports. We further compared cells, differentiated as embryoid bodies in 2 media proposed to initiate differentiation towards separate cell fates, as well as 20 adult tissues. From this analysis we have generated a gene list which defines pluripotency and establishes a baseline for the pluripotent state. Finally, we provide lists of genes enriched under both differentiation conditions which show the proposed bias toward independent cell fates.


Subject(s)
Databases, Factual , Pluripotent Stem Cells/metabolism , Animals , Cell Line , Gene Expression Profiling , Humans , Mice , National Institutes of Health (U.S.) , Pluripotent Stem Cells/cytology , Principal Component Analysis , United States
7.
Stem Cells ; 30(10): 2175-87, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22887864

ABSTRACT

The expression and function of several multidrug transporters (including ABCB1 and ABCG2) have been studied in human cancer cells and in mouse and human adult stem cells. However, the expression of ABCG2 in human embryonic stem cells (hESCs) remains unclear. Limited and contradictory results in the literature from two research groups have raised questions regarding its expression and function. In this study, we used quantitative real-time PCR, Northern blots, whole genome RNA sequencing, Western blots, and immunofluorescence microscopy to study ABCG2 expression in hESCs. We found that full-length ABCG2 mRNA transcripts are expressed in undifferentiated hESC lines. However, ABCG2 protein was undetectable even under embryoid body differentiation or cytotoxic drug induction. Moreover, surface ABCG2 protein was coexpressed with the differentiation marker stage-specific embryonic antigen-1 of hESCs, following constant BMP-4 signaling at days 4 and 6. This expression was tightly correlated with the downregulation of two microRNAs (miRNAs) (i.e., hsa-miR-519c and hsa-miR-520h). Transfection of miRNA mimics and inhibitors of these two miRNAs confirmed their direct involvement in the regulation ABCG2 translation. Our findings clarify the controversy regarding the expression of the ABCG2 gene and also provide new insights into translational control of the expression of membrane transporter mRNAs by miRNAs in hESCs.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Embryonic Stem Cells/metabolism , Gene Expression Regulation , MicroRNAs/genetics , Neoplasm Proteins/genetics , RNA, Messenger/biosynthesis , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/metabolism , Animals , Bone Morphogenetic Protein 4/genetics , Bone Morphogenetic Protein 4/metabolism , Cell Differentiation , Cells, Cultured , Embryonic Stem Cells/cytology , Feeder Cells , Fibroblasts , Humans , Lewis X Antigen/genetics , Lewis X Antigen/metabolism , Mice , MicroRNAs/metabolism , Neoplasm Proteins/metabolism , Protein Biosynthesis , Transfection
8.
Stem Cell Res ; 9(3): 237-48, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22910561

ABSTRACT

Regenerative medicine, relying on human embryonic stem cell (hESC) technology, opens promising new avenues for therapy of many severe diseases. However, this approach is restricted by limited production of the desired cells due to the refractory properties of hESC growth in vitro. It is further hindered by insufficient control of cellular stress, growth rates, and heterogeneous cellular states under current culture conditions. In this study, we report a novel cell culture method based on a non-colony type monolayer (NCM) growth. Human ESCs under NCM remain pluripotent as determined by teratoma assays and sustain the potential to differentiate into three germ layers. This NCM culture has been shown to homogenize cellular states, precisely control growth rates, significantly increase cell production, and enhance hESC recovery from cryopreservation without compromising chromosomal integrity. This culture system is simple, robust, scalable, and suitable for high-throughput screening and drug discovery.


Subject(s)
Cell Culture Techniques/methods , Cell Proliferation , Embryonic Stem Cells/cytology , Cell Differentiation , Cell Line , Gene Expression , Humans
9.
Nat Biotechnol ; 25(7): 803-16, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17572666

ABSTRACT

The International Stem Cell Initiative characterized 59 human embryonic stem cell lines from 17 laboratories worldwide. Despite diverse genotypes and different techniques used for derivation and maintenance, all lines exhibited similar expression patterns for several markers of human embryonic stem cells. They expressed the glycolipid antigens SSEA3 and SSEA4, the keratan sulfate antigens TRA-1-60, TRA-1-81, GCTM2 and GCT343, and the protein antigens CD9, Thy1 (also known as CD90), tissue-nonspecific alkaline phosphatase and class 1 HLA, as well as the strongly developmentally regulated genes NANOG, POU5F1 (formerly known as OCT4), TDGF1, DNMT3B, GABRB3 and GDF3. Nevertheless, the lines were not identical: differences in expression of several lineage markers were evident, and several imprinted genes showed generally similar allele-specific expression patterns, but some gene-dependent variation was observed. Also, some female lines expressed readily detectable levels of XIST whereas others did not. No significant contamination of the lines with mycoplasma, bacteria or cytopathic viruses was detected.


Subject(s)
Embryonic Stem Cells/cytology , Gene Expression Regulation, Developmental , Alkaline Phosphatase/metabolism , Antigens, CD/biosynthesis , Biotechnology/methods , Cell Differentiation , Cell Lineage , Cell Membrane/metabolism , Cells, Cultured , Cluster Analysis , Female , Gene Expression Profiling , Genotype , Glycolipids/chemistry , Humans , Membrane Glycoproteins/biosynthesis , Tetraspanin 29
10.
Ecology ; 88(5): 1232-40, 2007 May.
Article in English | MEDLINE | ID: mdl-17536409

ABSTRACT

Species may compete indirectly by altering the traits of a shared resource. For example, herbivore-induced responses in plants may make plants more resistant or susceptible to additional herbivorous insect species. Herbivore-induced plant responses can significantly affect interspecific competition and herbivore population dynamics. These herbivore-herbivore indirect interactions have been overlooked in aquatic ecosystems where previous studies used the same herbivore species to induce changes and to assess the effects of these changes. We asked whether seaweed grazing by one of two herbivorous, congeneric snail species (Littorina obtusata or Littorina littorea) with different feeding strategies and preferences would affect subsequent feeding preferences of three herbivore species (both snails and the isopod Idotea baltica) and population densities of three herbivore species (both snails and a third periwinkle snail, Lacuna vincta). In addition, we measured phlorotannin concentrations to test the hypothesis that these metabolites function as induced defenses in the Phaeophyceae. Snail herbivory induced cue-specific responses in apical tissues of the seaweed Fucus vesiculosus that affected the three herbivore species similarly. When compared to ungrazed controls, direct grazing by Littorina obtusata reduced seaweed palatability by at least 52% for both snail species and the isopod species. In contrast, direct grazing by L. littorea did not decrease seaweed palatability for any herbivore, indicating herbivore-specific responses. Previous grazing by L. obtusata reduced populations of L. littorea on outplanted seaweeds by 46% but had no effect on L. obtusata populations. Phlorotannins, a potential class of inducible chemicals in brown algae, were not more concentrated in grazed seaweed tissues, suggesting that some other trait was responsible for the induced resistance. Our results indicate that marine herbivores may compete via inducible responses in shared seaweeds. These plant-mediated interactions were asymmetric with a specialist (L. obtusata) competitively superior to a generalist (L. littorea).


Subject(s)
Ecosystem , Feeding Behavior/physiology , Seaweed , Snails/physiology , Animals , Plants, Edible/chemistry , Plants, Edible/growth & development , Plants, Edible/metabolism , Population Density , Population Dynamics , Seaweed/chemistry , Seaweed/growth & development , Seaweed/metabolism
11.
Int J Biochem Cell Biol ; 38(7): 1063-75, 2006.
Article in English | MEDLINE | ID: mdl-16469522

ABSTRACT

The culture of human embryonic stem cells (hESCs) is limited, both technically and with respect to clinical potential, by the use of mouse embryonic fibroblasts (MEFs) as a feeder layer. The concern over xenogeneic contaminants from the mouse feeder cells may restrict transplantation to humans and the variability in MEFs from batch-to-batch and laboratory-to-laboratory may contribute to some of the variability in experimental results. Finally, use of any feeder layer increases the work load and subsequently limits the large-scale culture of human ES cells. Thus, the development of feeder-free cultures will allow more reproducible culture conditions, facilitate scale-up and potentiate the clinical use of cells differentiated from hESC cultures. In this review, we describe various methods tested to culture cells in the absence of MEF feeder layers and other advances in eliminating xenogeneic products from the culture system.


Subject(s)
Coculture Techniques/methods , Embryo, Mammalian/cytology , Stem Cells/cytology , Animals , Antigens, Heterophile/analysis , Antigens, Heterophile/immunology , Cell Differentiation/drug effects , Cell Line/metabolism , Cell Proliferation/drug effects , Culture Media, Conditioned/analysis , Culture Media, Conditioned/pharmacology , Fibroblasts/metabolism , Humans , Mice , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism , Recombinant Proteins/chemistry , Stem Cells/drug effects
12.
EMBO J ; 24(1): 209-20, 2005 Jan 12.
Article in English | MEDLINE | ID: mdl-15592431

ABSTRACT

The extracellular aggregation of amyloid beta (Abeta) peptides and the intracellular hyperphosphorylation of tau at specific epitopes are pathological hallmarks of neurodegenerative diseases such as Alzheimer's disease (AD). Cdk5 phosphorylates tau at AD-specific phospho-epitopes when it associates with p25. p25 is a truncated activator, which is produced from the physiological Cdk5 activator p35 upon exposure to Abeta peptides. We show that neuronal infections with Cdk5 inhibitory peptide (CIP) selectively inhibit p25/Cdk5 activity and suppress the aberrant tau phosphorylation in cortical neurons. Furthermore, Abeta(1-42)-induced apoptosis of these cortical neurons was also reduced by coinfection with CIP. Of particular importance is our finding that CIP did not inhibit endogenous or transfected p35/Cdk5 activity, nor did it inhibit the other cyclin-dependent kinases such as Cdc2, Cdk2, Cdk4 and Cdk6. These results, therefore, provide a strategy to address, and possibly ameliorate, the pathology of neurodegenerative diseases that may be a consequence of aberrant p25 activation of Cdk5, without affecting 'normal' Cdk5 activity.


Subject(s)
Apoptosis/physiology , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/metabolism , Nerve Tissue Proteins/metabolism , Neurons/physiology , Peptides/metabolism , tau Proteins/metabolism , Amyloid beta-Peptides/metabolism , Animals , CDC2 Protein Kinase/metabolism , Caspase 3 , Caspases/metabolism , Cells, Cultured , Cyclin-Dependent Kinase 5 , Cyclin-Dependent Kinases/genetics , Embryo, Mammalian/anatomy & histology , Humans , Nerve Tissue Proteins/genetics , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Neurons/cytology , Peptide Fragments/metabolism , Peptides/genetics , Phosphorylation , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...