Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Stem Cell Reports ; 11(4): 998-1008, 2018 10 09.
Article in English | MEDLINE | ID: mdl-30269954

ABSTRACT

Differentiation of astrocytes from human pluripotent stem cells (hPSCs) is a tedious and variable process. This hampers the study of hPSC-generated astrocytes in disease processes and drug development. By using CRISPR/Cas9-mediated inducible expression of NFIA or NFIA plus SOX9 in hPSCs, we developed a method to efficiently generate astrocytes in 4-7 weeks. The astrocytic identity of the induced cells was verified by their characteristic molecular and functional properties as well as after transplantation. Furthermore, we developed a strategy to generate region-specific astrocyte subtypes by combining differentiation of regional progenitors and transgenic induction of astrocytes. This simple and efficient method offers a new opportunity to study the fundamental biology of human astrocytes and their roles in disease processes.


Subject(s)
Astrocytes/cytology , Pluripotent Stem Cells/cytology , Astrocytes/metabolism , Cell Differentiation , Humans , NFI Transcription Factors/metabolism , Neuronal Outgrowth , Pluripotent Stem Cells/metabolism , SOX9 Transcription Factor/metabolism
2.
Assay Drug Dev Technol ; 15(4): 178-188, 2017.
Article in English | MEDLINE | ID: mdl-28525289

ABSTRACT

Electrophysiology of excitable cells, including muscle cells and neurons, has been measured by making direct contact with a single cell using a micropipette electrode. To increase the assay throughput, optical devices such as microscopes and microplate readers have been used to analyze electrophysiology of multiple cells. We have established a high-throughput (HTP) analysis of action potentials (APs) in highly enriched motor neurons and cardiomyocytes (CMs) that are differentiated from human induced pluripotent stem cells (iPSCs). A multichannel electric field stimulation (EFS) device enabled the ability to electrically stimulate cells and measure dynamic changes in APs of excitable cells ultra-rapidly (>100 data points per second) by imaging entire 96-well plates. We found that the activities of both neurons and CMs and their response to EFS and chemicals are readily discerned by our fluorescence imaging-based HTP phenotyping assay. The latest generation of calcium (Ca2+) indicator dyes, FLIPR Calcium 6 and Cal-520, with the HTP device enables physiological analysis of human iPSC-derived samples highlighting its potential application for understanding disease mechanisms and discovering new therapeutic treatments.


Subject(s)
High-Throughput Screening Assays , Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/cytology , Neurons/cytology , Optical Imaging , Calcium/metabolism , Cells, Cultured , Electric Stimulation/instrumentation , Electrodes , High-Throughput Screening Assays/instrumentation , Humans , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Neurons/metabolism , Optical Imaging/instrumentation , Phenotype
3.
Cell Stem Cell ; 17(2): 233-44, 2015 Aug 06.
Article in English | MEDLINE | ID: mdl-26145478

ABSTRACT

Precise temporal control of gene expression or deletion is critical for elucidating gene function in biological systems. However, the establishment of human pluripotent stem cell (hPSC) lines with inducible gene knockout (iKO) remains challenging. We explored building iKO hPSC lines by combining CRISPR/Cas9-mediated genome editing with the Flp/FRT and Cre/LoxP system. We found that "dual-sgRNA targeting" is essential for biallelic knockin of FRT sequences to flank the exon. We further developed a strategy to simultaneously insert an activity-controllable recombinase-expressing cassette and remove the drug-resistance gene, thus speeding up the generation of iKO hPSC lines. This two-step strategy was used to establish human embryonic stem cell (hESC) and induced pluripotent stem cell (iPSC) lines with iKO of SOX2, PAX6, OTX2, and AGO2, genes that exhibit diverse structural layout and temporal expression patterns. The availability of iKO hPSC lines will substantially transform the way we examine gene function in human cells.


Subject(s)
CRISPR-Cas Systems/genetics , Gene Knockout Techniques , Genetic Engineering/methods , Stem Cells/metabolism , Base Sequence , Cell Differentiation , Exons/genetics , Gene Expression Regulation , Gene Knock-In Techniques , Gene Targeting , Homozygote , Humans , Molecular Sequence Data , Mutagenesis, Insertional/genetics , Otx Transcription Factors/metabolism , Phospholipid Transfer Proteins/metabolism , Pluripotent Stem Cells/metabolism , RNA, Guide, Kinetoplastida/metabolism
4.
Sci Rep ; 5: 12189, 2015 Jul 20.
Article in English | MEDLINE | ID: mdl-26190808

ABSTRACT

Spinal muscular atrophy (SMA) presents severe muscle weakness with limited motor neuron (MN) loss at an early stage, suggesting potential functional alterations in MNs that contribute to SMA symptom presentation. Using SMA induced pluripotent stem cells (iPSCs), we found that SMA MNs displayed hyperexcitability with increased membrane input resistance, hyperpolarized threshold, and larger action potential amplitude, which was mimicked by knocking down full length survival motor neuron (SMN) in non-SMA MNs. We further discovered that SMA MNs exhibit enhanced sodium channel activities with increased current amplitude and facilitated recovery, which was corrected by restoration of SMN1 in SMA MNs. Together we propose that SMN reduction results in MN hyperexcitability and impaired neurotransmission, the latter of which exacerbate each other via a feedback loop, thus contributing to severe symptoms at an early stage of SMA.


Subject(s)
Motor Neurons/metabolism , Muscular Atrophy, Spinal/physiopathology , Cell Differentiation/genetics , Cell Line , Gene Expression Profiling , Gene Expression Regulation , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Membrane Potentials , Motor Neurons/cytology , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/metabolism , RNA, Messenger/genetics , Sodium Channels/metabolism , Survival of Motor Neuron 1 Protein/genetics , Survival of Motor Neuron 1 Protein/metabolism , Synaptic Potentials
5.
Sci Rep ; 5: 10666, 2015 Jun 08.
Article in English | MEDLINE | ID: mdl-26053681

ABSTRACT

Dorsal root avulsion results in permanent impairment of sensory functions due to disconnection between the peripheral and central nervous system. Improved strategies are therefore needed to reconnect injured sensory neurons with their spinal cord targets in order to achieve functional repair after brachial and lumbosacral plexus avulsion injuries. Here, we show that sensory functions can be restored in the adult mouse if avulsed sensory fibers are bridged with the spinal cord by human neural progenitor (hNP) transplants. Responses to peripheral mechanical sensory stimulation were significantly improved in transplanted animals. Transganglionic tracing showed host sensory axons only in the spinal cord dorsal horn of treated animals. Immunohistochemical analysis confirmed that sensory fibers had grown through the bridge and showed robust survival and differentiation of the transplants. Section of the repaired dorsal roots distal to the transplant completely abolished the behavioral improvement. This demonstrates that hNP transplants promote recovery of sensorimotor functions after dorsal root avulsion, and that these effects are mediated by spinal ingrowth of host sensory axons. These results provide a rationale for the development of novel stem cell-based strategies for functionally useful bridging of the peripheral and central nervous system.


Subject(s)
Axons/physiology , Human Embryonic Stem Cells/physiology , Nerve Regeneration/physiology , Sensory Receptor Cells/physiology , Spinal Cord Injuries/physiopathology , Spinal Nerve Roots/physiology , Stem Cells/physiology , Animals , Ganglia, Spinal/physiology , Humans , Male , Mice , Spinal Cord/physiology
6.
Nat Commun ; 6: 6626, 2015 Mar 25.
Article in English | MEDLINE | ID: mdl-25806427

ABSTRACT

Human pluripotent stem cells (hPSCs) have opened new opportunities for understanding human development, modelling disease processes and developing new therapeutics. However, these applications are hindered by the low efficiency and heterogeneity of cell types, such as motorneurons (MNs), differentiated from hPSCs as well as our inability to maintain the potency of lineage-committed progenitors. Here by using a combination of small molecules that regulate multiple signalling pathways, we develop a method to guide human embryonic stem cells to a near-pure population (>95%) of motor neuron progenitors (MNPs) in 12 days, and an enriched population (>90%) of functionally mature MNs in an additional 16 days. More importantly, the MNPs can be expanded for at least five passages so that a single MNP can be amplified to 1 × 10(4). This method is reproducible in human-induced pluripotent stem cells and is applied to model MN-degenerative diseases and in proof-of-principle drug-screening assays.


Subject(s)
Motor Neurons/cytology , Neural Stem Cells/cytology , Pluripotent Stem Cells/cytology , Cell Culture Techniques , Humans , Neuromuscular Junction/cytology
7.
J Clin Invest ; 125(3): 1033-42, 2015 Mar 02.
Article in English | MEDLINE | ID: mdl-25642771

ABSTRACT

Astrocytes are integral components of the homeostatic neural network as well as active participants in pathogenesis of and recovery from nearly all neurological conditions. Evolutionarily, compared with lower vertebrates and nonhuman primates, humans have an increased astrocyte-to-neuron ratio; however, a lack of effective models has hindered the study of the complex roles of human astrocytes in intact adult animals. Here, we demonstrated that after transplantation into the cervical spinal cords of adult mice with severe combined immunodeficiency (SCID), human pluripotent stem cell-derived (PSC-derived) neural progenitors migrate a long distance and differentiate to astrocytes that nearly replace their mouse counterparts over a 9-month period. The human PSC-derived astrocytes formed networks through their processes, encircled endogenous neurons, and extended end feet that wrapped around blood vessels without altering locomotion behaviors, suggesting structural, and potentially functional, integration into the adult mouse spinal cord. Furthermore, in SCID mice transplanted with neural progenitors derived from induced PSCs from patients with ALS, astrocytes were generated and distributed to a similar degree as that seen in mice transplanted with healthy progenitors; however, these mice exhibited motor deficit, highlighting functional integration of the human-derived astrocytes. Together, these results indicate that this chimeric animal model has potential for further investigating the roles of human astrocytes in disease pathogenesis and repair.


Subject(s)
Astrocytes/physiology , Neural Stem Cells/transplantation , Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/physiopathology , Animals , Apoptosis , Cell Differentiation , Cell Movement , Cell Proliferation , Cells, Cultured , Humans , Induced Pluripotent Stem Cells/transplantation , Mice, SCID , Motor Neurons/physiology , Muscle Strength , Spinal Cord/pathology
8.
Cell Stem Cell ; 14(6): 796-809, 2014 Jun 05.
Article in English | MEDLINE | ID: mdl-24704493

ABSTRACT

Amyotrophic lateral sclerosis (ALS) presents motoneuron (MN)-selective protein inclusions and axonal degeneration but the underlying mechanisms of such are unknown. Using induced pluripotent cells (iPSCs) from patients with mutation in the Cu/Zn superoxide dismutase (SOD1) gene, we show that spinal MNs, but rarely non-MNs, exhibited neurofilament (NF) aggregation followed by neurite degeneration when glia were not present. These changes were associated with decreased stability of NF-L mRNA and binding of its 3' UTR by mutant SOD1 and thus altered protein proportion of NF subunits. Such MN-selective changes were mimicked by expression of a single copy of the mutant SOD1 in human embryonic stem cells and were prevented by genetic correction of the SOD1 mutation in patient's iPSCs. Importantly, conditional expression of NF-L in the SOD1 iPSC-derived MNs corrected the NF subunit proportion, mitigating NF aggregation and neurite degeneration. Thus, NF misregulation underlies mutant SOD1-mediated NF aggregation and axonal degeneration in ALS MNs.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Induced Pluripotent Stem Cells/metabolism , Models, Biological , Motor Neurons/metabolism , Mutant Proteins/metabolism , Superoxide Dismutase/metabolism , Amyotrophic Lateral Sclerosis/pathology , Humans , Induced Pluripotent Stem Cells/pathology , Mutant Proteins/genetics , Mutation , Superoxide Dismutase/genetics , Superoxide Dismutase-1
9.
Stem Cells ; 32(5): 1230-8, 2014 May.
Article in English | MEDLINE | ID: mdl-24497442

ABSTRACT

Regulatable transgene expression in human pluripotent stem cells (hPSCs) and their progenies is often necessary to dissect gene function in a temporal and spatial manner. However, hPSC lines with inducible transgene expression, especially in differentiated progenies, have not been established due to silencing of randomly inserted genes during stem cell expansion and/or differentiation. Here, we report the use of transcription activator-like effector nucleases-mediated targeting to AAVS1 site to generate versatile conditional hPSC lines. Transgene (both green fluorescent protein and a functional gene) expression in hPSCs and their derivatives was not only sustained but also tightly regulated in response to doxycycline both in vitro and in vivo. We modified the donor construct so that any gene of interest can be readily inserted to produce hPSC lines with conditional transgene expression. This technology will substantially improve the way we study human stem cells.


Subject(s)
Gene Expression/genetics , Induced Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/metabolism , Transgenes/genetics , Animals , Astrocytes/cytology , Astrocytes/metabolism , Blotting, Western , Doxycycline/pharmacology , Gene Expression/drug effects , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Immunohistochemistry , Induced Pluripotent Stem Cells/cytology , Mice, SCID , Microscopy, Confocal , Neurons/cytology , Neurons/metabolism , Pluripotent Stem Cells/cytology
10.
Development ; 140(12): 2611-8, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23637338

ABSTRACT

The role of miRNAs in neuroectoderm specification is largely unknown. We screened miRNA profiles that are differentially changed when human embryonic stem cells (hESCs) were differentiated to neuroectodermal precursors (NEP), but not to epidermal (EPI) cells and found that two miRNA families, miR-200 and miR-96, were uniquely downregulated in the NEP cells. We confirmed zinc-finger E-box-binding homeobox (ZEB) transcription factors as a target of the miR-200 family members and identified paired box 6 (PAX6) transcription factor as the new target of miR-96 family members via gain- and loss-of-function analyses. Given the essential roles of ZEBs and PAX6 in neural induction, we propose a model by which miR-200 and miR-96 families coordinate to regulate neural induction.


Subject(s)
Embryonic Stem Cells/metabolism , MicroRNAs/metabolism , Neural Plate/cytology , Animals , Cell Differentiation , Cell Line , Cell Lineage , Down-Regulation , Embryonic Stem Cells/cytology , Epidermal Cells , Epidermis/metabolism , Eye Proteins/genetics , Eye Proteins/metabolism , Gene Expression Regulation , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Mice , MicroRNAs/genetics , Neural Plate/metabolism , PAX6 Transcription Factor , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Time Factors , Transcription, Genetic , Zinc Finger E-box Binding Homeobox 2
11.
Cell Rep ; 3(5): 1580-91, 2013 May 30.
Article in English | MEDLINE | ID: mdl-23643533

ABSTRACT

Postnatal and adult human and monkey fibroblasts were infected with Sendai virus containing the Yamanaka factors for 24 hr, then they were cultured in a chemically defined medium containing leukemia inhibitory factor (LIF), transforming growth factor (TGF)-ß inhibitor SB431542, and glycogen synthase kinase (GSK)-3ß inhibitor CHIR99021 at 39°C for inactivation of the virus. Induced neural progenitor (iNP) colonies appeared as early as day 13 and can be expanded for >20 passages. Under the same defined condition, no induced pluripotent stem cell (iPSC) colonies formed at either 37°C or 39°C. The iNPs predominantly express hindbrain genes and differentiate into hindbrain neurons, and when caudalized, they produced an enriched population of spinal motor neurons. Following transplantation into the forebrain, the iNP-derived cells retained the hindbrain identity. The ability to generate defined, integration-free iNPs from adult primate fibroblasts under a defined condition with predictable fate choices will facilitate disease modeling and therapeutic development.


Subject(s)
Fibroblasts/cytology , Neural Stem Cells/cytology , Animals , Benzamides/pharmacology , Cell Differentiation , Dioxoles/pharmacology , Fibroblasts/drug effects , Haplorhini , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/transplantation , Leukemia Inhibitory Factor/pharmacology , Neuroglia/cytology , Neuroglia/metabolism , Neurons/cytology , Neurons/metabolism , Prosencephalon/metabolism , Pyridines/pharmacology , Pyrimidines/pharmacology , Rhombencephalon/metabolism , Temperature
12.
Cereb Cortex ; 21(9): 2177-86, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21330470

ABSTRACT

The mechanisms by which transcription factors control stepwise lineage restriction during the specification of cortical neurons remain largely unknown. Here, we investigated the role of forebrain embryonic zinc finger like (Fezf2) in this process by generating Fezf2 knockdown and tetracycline-inducible Fezf2 overexpression mouse embryonic stem cell (mESC) lines. The overexpression of Fezf2 at early time points significantly increased the generation of rostral forebrain progenitors (Foxg1(+), Six3(+)) and inhibited the expression of transcription factors which are expressed by the midbrain and caudal diencephalon (En1(+), Irx(+)). This effect was partially achieved by the regulation of Wnt signaling during this critical early time window. The role of Fezf2 in regulating the rostrocaudal patterning was further confirmed by the significant decrease in the expression of Foxg1 and Six3 and the increase in the expression of En1 when Fezf2 was knocked down. In addition, Fezf2 overexpression at later time points had little effect on the expression of Foxg1 and Six3. Instead, Fezf2 promotes the generation of dorsal telencephalic progenitors and deep-layer cortical neurons at later stages. Collectively, our data suggest that Fezf2 controls the specification of telencephalic progenitors from mESCs through differentially regulating the expression of rostrocaudal and dorsoventral patterning genes.


Subject(s)
Cell Differentiation/genetics , Cell Differentiation/physiology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Neural Stem Cells/physiology , Telencephalon/embryology , Animals , Blotting, Western , Cell Line , DNA-Binding Proteins/biosynthesis , Eye Proteins/genetics , Forkhead Transcription Factors/genetics , Gene Knockdown Techniques , Genetic Vectors , Homeodomain Proteins/genetics , Immunohistochemistry , Lentivirus/genetics , Mice , Nerve Tissue Proteins/biosynthesis , Prosencephalon/cytology , Prosencephalon/embryology , Reverse Transcriptase Polymerase Chain Reaction , Telencephalon/cytology , Transcription Factors/genetics , Transfection , Wnt Signaling Pathway/physiology , Homeobox Protein SIX3
13.
Biochem Biophys Res Commun ; 404(1): 388-92, 2011 Jan 07.
Article in English | MEDLINE | ID: mdl-21130748

ABSTRACT

Botulinum neurotoxins (BoNTs), the most poisonous protein toxins known, represent a serious bioterrorism threat but are also used as a unique and important bio-pharmaceutical to treat an increasing myriad of neurological disorders. The only currently accepted detection method by the United States Food and Drug Administration for biological activity of BoNTs and for potency determination of pharmaceutical preparations is the mouse bioassay (MBA). Recent advances have indicated that cell-based assays using primary neuronal cells can provide an equally sensitive and robust detection platform as the MBA to reliably and quantitatively detect biologically active BoNTs. This study reports for the first time a BoNT detection assay using mouse embryonic stem cells to produce a neuronal cell culture. The data presented indicate that this assay can reliably detect BoNT/A with a similar sensitivity as the MBA.


Subject(s)
Bioterrorism , Botulinum Toxins, Type A/analysis , Embryonic Stem Cells/cytology , Neurogenesis , Neurons/chemistry , Animals , Biological Assay , Botulinum Toxins, Type A/toxicity , Cell Count , Cell Culture Techniques , Cells, Cultured , Mice , Neurons/cytology , Neurons/drug effects , Sensitivity and Specificity
14.
Cell Stem Cell ; 7(1): 90-100, 2010 Jul 02.
Article in English | MEDLINE | ID: mdl-20621053

ABSTRACT

The transcriptional regulation of neuroectoderm (NE) specification is unknown. Here we show that Pax6 is uniformly expressed in early NE cells of human fetuses and those differentiated from human embryonic stem cells (hESCs). This is in contrast to the later expression of Pax6 in restricted mouse brain regions. Knockdown of Pax6 blocks NE specification from hESCs. Overexpression of either Pax6a or Pax6b, but not Pax6triangle upPD, triggers hESC differentiation. However, only Pax6a converts hESCs to NE. In contrast, neither loss nor gain of function of Pax6 affects mouse NE specification. Both Pax6a and Pax6b bind to pluripotent gene promoters but only Pax6a binds to NE genes during human NE specification. These findings indicate that Pax6 is a transcriptional determinant of the human NE and suggest that Pax6a and Pax6b coordinate with each other in determining the transition from pluripotency to the NE fate in human by differentially targeting pluripotent and NE genes.


Subject(s)
Cell Differentiation/physiology , Eye Proteins/metabolism , Homeodomain Proteins/metabolism , Neural Plate/cytology , Neural Plate/metabolism , Paired Box Transcription Factors/metabolism , Repressor Proteins/metabolism , Animals , Cell Differentiation/genetics , Cell Line , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Eye Proteins/genetics , Homeodomain Proteins/genetics , Humans , In Vitro Techniques , Mice , Mice, SCID , Models, Biological , PAX6 Transcription Factor , Paired Box Transcription Factors/genetics , Repressor Proteins/genetics , Teratoma/pathology
15.
Methods Mol Biol ; 614: 127-34, 2010.
Article in English | MEDLINE | ID: mdl-20225040

ABSTRACT

Human Embryonic stem cells (hESCs) offer an invaluable tool for revealing human biology and a potential source of functional cells/tissues for regenerative medicine. The utility of hESCs will likely be significantly enhanced and broadened by our ability to build versatile genetically modified hESC lines. Here, we describe an efficient lentiviral vector mediated method to establish stable transgenic hESCs.


Subject(s)
Embryonic Stem Cells/metabolism , Genetic Vectors , Lentivirus/genetics , Transgenes , Cell Line , Humans , Promoter Regions, Genetic , Regenerative Medicine , Transfection
16.
Nat Protoc ; 4(11): 1614-22, 2009.
Article in English | MEDLINE | ID: mdl-19834476

ABSTRACT

We have developed a four-part protocol to differentiate human embryonic stem cells (hESCs) to oligodendrocyte progenitor cells (OPCs) according to developmental principles. In the first 2 weeks, hESCs are induced to differentiate into neuroepithelial cells, which form neural tube-like rosettes. In the following 10 d, these neuroepithelial cells are specified to OLIG2-expressing progenitors in the presence of retinoic acid (RA) and sonic hedgehog (SHH). Upon treatment with fibroblast growth factor 2 (FGF2) for another 10 d, these progenitors convert to OLIG2 and NKX2.2-expressing pre-OPCs. Finally, the pre-OPCs take 8-9 weeks to differentiate into OPCs, which express additional markers of oligodendrocytes, such as SOX10, platelet-derived growth factor receptor alpha (PDGFRalpha) and NG2. The unique aspects of the protocol are the use of FGF2 to promote the differentiation of gliogenic pre-OPCs in the third part and the removal of FGF2 during the transition of pre-OPCs to OPCs. This 3-month differentiation protocol consistently yields OPCs of high purity capable of producing myelin sheaths in vivo.


Subject(s)
Cell Culture Techniques/methods , Oligodendroglia/cytology , Pluripotent Stem Cells/cytology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/drug effects , Fibroblast Growth Factor 2/pharmacology , Hedgehog Proteins/pharmacology , Homeobox Protein Nkx-2.2 , Homeodomain Proteins/metabolism , Humans , Nerve Tissue Proteins/metabolism , Neuroepithelial Cells/cytology , Neuroepithelial Cells/drug effects , Neuroepithelial Cells/metabolism , Nuclear Proteins , Oligodendrocyte Transcription Factor 2 , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism , Transcription Factors/metabolism , Tretinoin/pharmacology , Zebrafish Proteins
17.
Stem Cells ; 27(5): 1032-41, 2009 May.
Article in English | MEDLINE | ID: mdl-19415769

ABSTRACT

To circumvent the silencing effect of transgene expression in human embryonic stem cells (hESCs), we employed the Cre recombination-mediated cassette exchange strategy to target the silencing-resistant site in the genome. We have identified new loci that sustain transgene expression during stem cell expansion and differentiation to cells representing the three germ layers in vitro and in vivo. The built-in double loxP cassette in the established master hESC lines was specifically replaced by a targeting vector containing the same loxP sites, using the cell-permeable Cre protein transduction method, resulting in successful generation of new hESC lines with constitutive functional gene expression, inducible transgene expression, and lineage-specific reporter gene expression. This strategy and the master cell lines allow for rapid production of transgenic hESC lines in ordinary laboratories.


Subject(s)
Embryonic Stem Cells/metabolism , Integrases/metabolism , Mutagenesis, Insertional , Recombination, Genetic/genetics , Transgenes/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation , Cell Line , Cell Membrane Permeability , Embryonic Stem Cells/cytology , Gene Expression Regulation , Gene Silencing , Green Fluorescent Proteins/metabolism , Humans , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/cytology , Neurons/metabolism , Oligodendrocyte Transcription Factor 2 , Organ Specificity , Transfection
18.
Development ; 136(9): 1443-52, 2009 May.
Article in English | MEDLINE | ID: mdl-19363151

ABSTRACT

Human embryonic stem cells (hESCs) offer a platform to bridge what we have learned from animal studies to human biology. Using oligodendrocyte differentiation as a model system, we show that sonic hedgehog (SHH)-dependent sequential activation of the transcription factors OLIG2, NKX2.2 and SOX10 is required for sequential specification of ventral spinal OLIG2-expressing progenitors, pre-oligodendrocyte precursor cells (pre-OPCs) and OPCs from hESC-derived neuroepithelia, indicating that a conserved transcriptional network underlies OPC specification in human as in other vertebrates. However, the transition from pre-OPCs to OPCs is protracted. FGF2, which promotes mouse OPC generation, inhibits the transition of pre-OPCs to OPCs by repressing SHH-dependent co-expression of OLIG2 and NKX2.2. Thus, despite the conservation of a similar transcriptional network across vertebrates, human stem/progenitor cells may respond differently to those of other vertebrates to certain extrinsic factors.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/cytology , Fibroblast Growth Factor 2/pharmacology , Hedgehog Proteins/metabolism , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Signal Transduction/drug effects , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cells, Cultured , Gene Expression Regulation , Hedgehog Proteins/genetics , Homeobox Protein Nkx-2.2 , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Microscopy, Electron , Myelin Sheath/metabolism , Myelin Sheath/ultrastructure , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Nuclear Proteins , Oligodendrocyte Transcription Factor 2 , Oligodendroglia/cytology , Transcription Factors/genetics , Transcription Factors/metabolism , Zebrafish Proteins
19.
Stem Cells ; 26(4): 886-93, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18238853

ABSTRACT

Specification of distinct cell types from human embryonic stem cells (hESCs) is key to the potential application of these naïve pluripotent cells in regenerative medicine. Determination of the nontarget differentiated populations, which is lacking in the field, is also crucial. Here, we show an efficient differentiation of motor neurons ( approximately 50%) by a simple sequential application of retinoid acid and sonic hedgehog (SHH) in a chemically defined suspension culture. We also discovered that purmorphamine, a small molecule that activates the SHH pathway, could replace SHH for the generation of motor neurons. Immunocytochemical characterization indicated that cells differentiated from hESCs were nearly completely restricted to the ventral spinal progenitor fate (NKX2.2+, Irx3+, and Pax7-), with the exception of motor neurons (HB9+) and their progenitors (Olig2+). Thus, the directed neural differentiation system with small molecules, even without further purification, will facilitate basic and translational studies using human motoneurons at a minimal cost.


Subject(s)
Cell Differentiation , Directed Molecular Evolution/methods , Embryonic Stem Cells/cytology , Motor Neurons/cytology , Motor Neurons/physiology , Animals , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Line , Cells, Cultured , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/physiology , Hedgehog Proteins/biosynthesis , Hedgehog Proteins/genetics , Hedgehog Proteins/physiology , Homeobox Protein Nkx-2.2 , Homeodomain Proteins , Humans , Mice , Morpholines/pharmacology , Motor Neurons/drug effects , Nuclear Proteins , Purines/pharmacology , Spinal Cord/cytology , Spinal Cord/drug effects , Spinal Cord/physiology , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/physiology , Transcription Factors , Tretinoin/pharmacology , Tretinoin/physiology
20.
Mol Cell Neurosci ; 33(4): 371-80, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17035043

ABSTRACT

To dissect out interactions between the transcription factor Olig2 and other intrinsic and extrinsic factors in neural cell fate determination, we established a mouse embryonic stem (ES) cell line with induced expression of Olig2 along neural differentiation. During neuronal differentiation, both the control and Olig2-induced groups produced a similar proportion of HB9-expressing motoneurons in the presence of retinoic acid (RA) and sonic hedgehog (SHH), but both generated few motoneurons in the absence of SHH. Induced Olig2 expression did not alter the pattern of gene transcription without SHH, suggesting that Olig2 requires cooperation with RA and SHH for motoneuron specification. During glial differentiation, the Olig2-induced group generated significantly more oligodendrocytes and fewer neurons and astrocytes than the control group. This effect was not blocked by inhibition of SHH signaling, suggesting that Olig2 bypasses the need of SHH in oligodendrocyte specification. However, treatment with ciliary neurotropic factor (CNTF) markedly increased astrocyte and decreased oligodendrocyte differentiation even when Olig2 is sustained in the nuclei, suggesting that Olig2 cannot bypass the CNTF-STAT signaling to repress astrocyte differentiation.


Subject(s)
Astrocytes/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/physiology , Gene Expression Regulation, Developmental/physiology , Motor Neurons/metabolism , Nerve Tissue Proteins/metabolism , Oligodendroglia/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation/drug effects , Cell Line , Chick Embryo , Ciliary Neurotrophic Factor/pharmacology , Embryo, Mammalian , Gene Expression Regulation, Developmental/drug effects , Hedgehog Proteins/pharmacology , Homeodomain Proteins/metabolism , Immunohistochemistry/methods , Mice , Nerve Tissue Proteins/genetics , Oligodendrocyte Transcription Factor 2 , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Stem Cells/drug effects , Stem Cells/physiology , Transcription Factors/metabolism , Transfection/methods , Tretinoin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...