Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Cell ; 141(5): 872-83, 2010 May 28.
Article in English | MEDLINE | ID: mdl-20471072

ABSTRACT

The presence of two active X chromosomes (XaXa) is a hallmark of the ground state of pluripotency specific to murine embryonic stem cells (ESCs). Human ESCs (hESCs) invariably exhibit signs of X chromosome inactivation (XCI) and are considered developmentally more advanced than their murine counterparts. We describe the establishment of XaXa hESCs derived under physiological oxygen concentrations. Using these cell lines, we demonstrate that (1) differentiation of hESCs induces random XCI in a manner similar to murine ESCs, (2) chronic exposure to atmospheric oxygen is sufficient to induce irreversible XCI with minor changes of the transcriptome, (3) the Xa exhibits heavy methylation of the XIST promoter region, and (4) XCI is associated with demethylation and transcriptional activation of XIST along with H3K27-me3 deposition across the Xi. These findings indicate that the human blastocyst contains pre-X-inactivation cells and that this state is preserved in vitro through culture under physiological oxygen.


Subject(s)
Chromosomes, Human, X/metabolism , Embryonic Stem Cells/metabolism , Oxygen/metabolism , X Chromosome Inactivation , Animals , Cell Differentiation , Female , Histones/metabolism , Humans , Karyotyping , Male , Mice , Oxidative Stress , Pluripotent Stem Cells/metabolism
2.
Cell ; 136(5): 964-77, 2009 Mar 06.
Article in English | MEDLINE | ID: mdl-19269371

ABSTRACT

Induced pluripotent stem cells (iPSCs) derived from somatic cells of patients represent a powerful tool for biomedical research and may provide a source for replacement therapies. However, the use of viruses encoding the reprogramming factors represents a major limitation of the current technology since even low vector expression may alter the differentiation potential of the iPSCs or induce malignant transformation. Here, we show that fibroblasts from five patients with idiopathic Parkinson's disease can be efficiently reprogrammed and subsequently differentiated into dopaminergic neurons. Moreover, we derived hiPSCs free of reprogramming factors using Cre-recombinase excisable viruses. Factor-free hiPSCs maintain a pluripotent state and show a global gene expression profile, more closely related to hESCs than to hiPSCs carrying the transgenes. Our results indicate that residual transgene expression in virus-carrying hiPSCs can affect their molecular characteristics and that factor-free hiPSCs therefore represent a more suitable source of cells for modeling of human disease.


Subject(s)
Parkinson Disease/metabolism , Pluripotent Stem Cells/pathology , Cell Differentiation , Cellular Reprogramming , Dopamine/metabolism , Fibroblasts/metabolism , Humans , Neurons/metabolism
3.
Eur J Neurosci ; 52(7): 3667-3678, 2020 10.
Article in English | MEDLINE | ID: mdl-32579729

ABSTRACT

Microglia are the primary immune cells of the central nervous system and crucial to proper development and maintenance of the brain. Microglia have been recognized to be associated with neurodegenerative diseases and neuroinflammatory disorders. CX3C chemokine receptor 1 (CX3CR1), which is specifically expressed in microglia, regulates microglia homeostatic functions such as microglial activation and is downregulated in aged brain and disease-associated microglia in rodents, yet its role in human microglia is not fully understood. In this study, we investigated the function of CX3CR1 in human microglia using human induced pluripotent stem (iPS) cell-derived microglia-like cells. Human iPS cell-derived microglia-like cells expressed microglial markers and showed an activated state and phagocytic activity. Using CRISPR/Cas9 genome editing, we deleted CX3CR1 in human iPS cells and found increased inflammatory responses and phagocytic activity in mutant as compared to wild-type microglia-like cells. In addition, the CX3C chemokine ligand 1 (CX3CL1, a ligand for CX3CR1) significantly decreased the upregulation of IL-6 by lipopolysaccharide stimulation in human iPS cell-derived microglia-like cells. These results suggest that CX3CR1 in human microglia may contribute to microglial homeostasis by regulating inflammatory response and phagocytosis.


Subject(s)
Induced Pluripotent Stem Cells , Microglia , Aged , Brain/metabolism , CX3C Chemokine Receptor 1/genetics , CX3C Chemokine Receptor 1/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Microglia/metabolism , Phagocytosis
4.
Neuron ; 112(12): 1943-1958.e10, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38697112

ABSTRACT

Mutations in the methyl-DNA-binding protein MECP2 cause the neurodevelopmental disorder Rett syndrome (RTT). How MECP2 contributes to transcriptional regulation in normal and disease states is unresolved; it has been reported to be an activator and a repressor. We describe here the first integrated CUT&Tag, transcriptome, and proteome analyses using human neurons with wild-type (WT) and mutant MECP2 molecules. MECP2 occupies CpG-rich promoter-proximal regions in over four thousand genes in human neurons, including a plethora of autism risk genes, together with RNA polymerase II (RNA Pol II). MECP2 directly interacts with RNA Pol II, and genes occupied by both proteins showed reduced expression in neurons with MECP2 patient mutations. We conclude that MECP2 acts as a positive cofactor for RNA Pol II gene expression at many neuronal genes that harbor CpG islands in promoter-proximal regions and that RTT is due, in part, to the loss of gene activity of these genes in neurons.


Subject(s)
Methyl-CpG-Binding Protein 2 , Neurons , RNA Polymerase II , Transcription, Genetic , RNA Polymerase II/metabolism , RNA Polymerase II/genetics , Methyl-CpG-Binding Protein 2/metabolism , Methyl-CpG-Binding Protein 2/genetics , Humans , Neurons/metabolism , Promoter Regions, Genetic , Rett Syndrome/genetics , Rett Syndrome/metabolism , CpG Islands/genetics , Mutation , Gene Expression Regulation/genetics
5.
Proc Natl Acad Sci U S A ; 106(1): 157-62, 2009 Jan 06.
Article in English | MEDLINE | ID: mdl-19109433

ABSTRACT

Directed reprogramming of somatic cells by defined factors provides a novel method for the generation of patient-specific stem cells with the potential to bypass both the practical and ethical concerns associated with somatic cell nuclear transfer (SCNT) and human embryonic stem (hES) cells. Although the generation of induced pluripotent stem (iPS) cells has proven a robust technology in mouse and human, a major impediment to the use of iPS cells for therapeutic purposes has been the viral-based delivery of the reprogramming factors because multiple proviral integrations pose the danger of insertional mutagenesis. Here we report a novel approach to reduce the number of viruses necessary to reprogram somatic cells by delivering reprogramming factors in a single virus using 2A "self-cleaving" peptides, which support efficient polycistronic expression from a single promoter. We find that up to four reprogramming factors (Oct4, Sox2, Klf4, and c-Myc) can be expressed from a single virus to generate iPS cells in both embryonic and adult somatic mouse cells and we show that a single proviral copy is sufficient to generate iPS cells from mouse embryonic fibroblasts. In addition we have generated human induced pluripotent stem (hiPS) cell lines from human keratinocytes, demonstrating that a single polycistronic virus can reprogram human somatic cells.


Subject(s)
Cellular Reprogramming/genetics , Fibroblasts/cytology , Genetic Vectors/genetics , Keratinocytes/cytology , Pluripotent Stem Cells/cytology , Transgenes/genetics , Animals , Cells , Cells, Cultured , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Mice , Octamer Transcription Factor-3/genetics , Proto-Oncogene Proteins c-myc/genetics , SOXB1 Transcription Factors/genetics , Transfection/methods
6.
Nat Mater ; 9(9): 768-78, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20729850

ABSTRACT

Both human embryonic stem cells and induced pluripotent stem cells can self-renew indefinitely in culture; however, present methods to clonally grow them are inefficient and poorly defined for genetic manipulation and therapeutic purposes. Here we develop the first chemically defined, xeno-free, feeder-free synthetic substrates to support robust self-renewal of fully dissociated human embryonic stem and induced pluripotent stem cells. Material properties including wettability, surface topography, surface chemistry and indentation elastic modulus of all polymeric substrates were quantified using high-throughput methods to develop structure-function relationships between material properties and biological performance. These analyses show that optimal human embryonic stem cell substrates are generated from monomers with high acrylate content, have a moderate wettability and employ integrin alpha(v)beta(3) and alpha(v)beta(5) engagement with adsorbed vitronectin to promote colony formation. The structure-function methodology employed herein provides a general framework for the combinatorial development of synthetic substrates for stem cell culture.


Subject(s)
Biocompatible Materials/chemistry , Combinatorial Chemistry Techniques/methods , Induced Pluripotent Stem Cells/cytology , Cell Differentiation , Cells, Cultured , Humans , Induced Pluripotent Stem Cells/metabolism
7.
Methods Mol Biol ; 331: 55-76, 2006.
Article in English | MEDLINE | ID: mdl-16881509

ABSTRACT

The derivation of human embryonic stem (hES) cells is a challenging procedure. The isolation and maintenance of hES is visually and manually complicated, involving mechanical or enzymatic passaging using either collagenase or trypsin. This chapter describes detailed protocols that have been used for the derivation, maintenance, and characterization of hES cells in vitro along with protocols to test their differentiation potential in vivo. When used as a guideline, these protocols will assist researchers in setting up a hES cell laboratory.


Subject(s)
Cell Culture Techniques/methods , Cell Separation/methods , Pluripotent Stem Cells/cytology , Biomarkers , Cell Culture Techniques/standards , Cell Differentiation , Cell Separation/standards , Coculture Techniques/methods , Cryopreservation , Fibroblasts/cytology , Humans , Immunohistochemistry , Karyotyping , Laboratories , Quality Control
8.
Nat Med ; 22(11): 1358-1367, 2016 11.
Article in English | MEDLINE | ID: mdl-27668937

ABSTRACT

Microglia, the only lifelong resident immune cells of the central nervous system (CNS), are highly specialized macrophages that have been recognized to have a crucial role in neurodegenerative diseases such as Alzheimer's, Parkinson's and adrenoleukodystrophy (ALD). However, in contrast to other cell types of the human CNS, bona fide microglia have not yet been derived from cultured human pluripotent stem cells. Here we establish a robust and efficient protocol for the rapid production of microglia-like cells from human (h) embryonic stem (ES) and induced pluripotent stem (iPS) cells that uses defined serum-free culture conditions. These in vitro pluripotent stem cell-derived microglia-like cells (termed pMGLs) faithfully recapitulate the expected ontogeny and characteristics of their in vivo counterparts, and they resemble primary fetal human and mouse microglia. We generated these cells from multiple disease-specific cell lines and find that pMGLs derived from an hES model of Rett syndrome are smaller than their isogenic controls. We further describe a platform to study the integration and live behavior of pMGLs in organotypic 3D cultures. This modular differentiation system allows for the study of microglia in highly defined conditions as they mature in response to developmentally relevant cues, and it provides a framework in which to study the long-term interactions of microglia residing in a tissue-like environment.


Subject(s)
Cell Differentiation , Human Embryonic Stem Cells/cytology , Induced Pluripotent Stem Cells/cytology , Microglia/cytology , Humans , In Vitro Techniques , Microglia/immunology , Organ Culture Techniques , Rett Syndrome/immunology
9.
BMC Dev Biol ; 5: 15, 2005 Jul 21.
Article in English | MEDLINE | ID: mdl-16033656

ABSTRACT

BACKGROUND: Pluripotent human embryonic stem cells (hESCs) have the potential to form every cell type in the body. These cells must be appropriately characterized prior to differentiation studies or when defining characteristics of the pluripotent state. Some developmentally regulated cell surface antigens identified by monoclonal antibodies in a variety of species and stem cell types have proven to be side chains of membrane glycolipids and glycoproteins. Therefore, to examine hESC surfaces for other potential pluripotent markers, we used a panel of 14 lectins, which were chosen based on their specificity for a variety of carbohydrates and carbohydrate linkages, along with stage specific embryonic antigen-4 (SSEA-4), to determine binding quantitation by flow cytometry and binding localization in adherent colonies by immunocytochemistry. RESULTS: Enriching cells for SSEA-4 expression increased the percentage of SSEA-4 positive cells to 98-99%. Using enriched high SSEA-4-expressing hESCs, we then analyzed the binding percentages of selected lectins and found a large variation in binding percentages ranging from 4% to 99% binding. Lycopersicon (tomato)esculetum lectin (TL), Ricinus communis agglutinin (RCA), and Concanavalin A (Con A) bound to SSEA-4 positive regions of hESCs and with similar binding percentages as SSEA-4. In contrast, we found Dolichos biflorus agglutinin (DBA) and Lotus tetragonolobus lectin (LTL) did not bind to hESCs while Phaseolus vulgaris leuco-agglutinin (PHA-L), Vicia villosa agglutinin (VVA), Ulex europaeus agglutinin (UEA), Phaseolus vulgaris erythro-agglutinin (PHA-E), and Maackia amurensis agglutinin (MAA) bound partially to hESCs. These binding percentages correlated well with immunocytochemistry results. CONCLUSION: Our results provide information about types of carbohydrates and carbohydrate linkages found on pluripotent hESC surfaces. We propose that TL, RCA and Con A may be used as markers that are associated with the pluripotent state of hESCs because binding percentages and binding localization of these lectins are similar to those of SSEA-4. Non-binding lectins, DBA and LTL, may identify differentiated cell types; however, we did not find these lectins to bind to pluripotent SSEA-4 positive hESCs. This work represents a fundamental base to systematically classify pluripotent hESCs, and in future studies these lectins may be used to distinguish differentiated hESC types based on glycan presentation that accompanies differentiation.


Subject(s)
Antigens, Surface/analysis , Glycosphingolipids/analysis , Lectins , Pluripotent Stem Cells/cytology , Carbohydrates/analysis , Embryo, Mammalian/cytology , Flow Cytometry , Humans , Immunohistochemistry , Protein Binding , Stage-Specific Embryonic Antigens
10.
BMC Neurosci ; 4: 27, 2003 Oct 22.
Article in English | MEDLINE | ID: mdl-14572319

ABSTRACT

BACKGROUND: We have developed a culture system for the efficient and directed differentiation of human embryonic stem cells (HESCs) to neural precursors and neurons.HESC were maintained by manual passaging and were differentiated to a morphologically distinct OCT-4+/SSEA-4- monolayer cell type prior to the derivation of embryoid bodies. Embryoid bodies were grown in suspension in serum free conditions, in the presence of 50% conditioned medium from the human hepatocarcinoma cell line HepG2 (MedII). RESULTS: A neural precursor population was observed within HESC derived serum free embryoid bodies cultured in MedII conditioned medium, around 7-10 days after derivation. The neural precursors were organized into rosettes comprised of a central cavity surrounded by ring of cells, 4 to 8 cells in width. The central cells within rosettes were proliferating, as indicated by the presence of condensed mitotic chromosomes and by phosphoHistone H3 immunostaining. When plated and maintained in adherent culture, the rosettes of neural precursors were surrounded by large interwoven networks of neurites. Immunostaining demonstrated the expression of nestin in rosettes and associated non-neuronal cell types, and a radial expression of Map-2 in rosettes. Differentiated neurons expressed the markers Map-2 and Neurofilament H, and a subpopulation of the neurons expressed tyrosine hydroxylase, a marker for dopaminergic neurons. CONCLUSION: This novel directed differentiation approach led to the efficient derivation of neuronal cultures from HESCs, including the differentiation of tyrosine hydroxylase expressing neurons. HESC were morphologically differentiated to a monolayer OCT-4+ cell type, which was used to derive embryoid bodies directly into serum free conditions. Exposure to the MedII conditioned medium enhanced the derivation of neural precursors, the first example of the effect of this conditioned medium on HESC.


Subject(s)
Cell Differentiation/physiology , Neurons/cytology , Stem Cells/cytology , Stem Cells/physiology , Animals , Antigens, Differentiation/biosynthesis , Cell Culture Techniques/methods , Cell Differentiation/drug effects , Cell Division/drug effects , Cell Division/physiology , Cells, Cultured , Coculture Techniques/methods , Culture Media, Conditioned/pharmacology , Culture Media, Serum-Free/pharmacology , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Mice , Neurons/drug effects , Neurons/metabolism , Stem Cells/drug effects , Time Factors
11.
Stem Cell Reports ; 2(6): 866-80, 2014 Jun 03.
Article in English | MEDLINE | ID: mdl-24936472

ABSTRACT

Niemann-Pick type C (NPC) disease is a fatal inherited lipid storage disorder causing severe neurodegeneration and liver dysfunction with only limited treatment options for patients. Loss of NPC1 function causes defects in cholesterol metabolism and has recently been implicated in deregulation of autophagy. Here, we report the generation of isogenic pairs of NPC patient-specific induced pluripotent stem cells (iPSCs) using transcription activator-like effector nucleases (TALENs). We observed decreased cell viability, cholesterol accumulation, and dysfunctional autophagic flux in NPC1-deficient human hepatic and neural cells. Genetic correction of a disease-causing mutation rescued these defects and directly linked NPC1 protein function to impaired cholesterol metabolism and autophagy. Screening for autophagy-inducing compounds in disease-affected human cells showed cell type specificity. Carbamazepine was found to be cytoprotective and effective in restoring the autophagy defects in both NPC1-deficient hepatic and neuronal cells and therefore may be a promising treatment option with overall benefit for NPC disease.


Subject(s)
Autophagy/physiology , Cholesterol/metabolism , Hepatocytes/metabolism , Induced Pluripotent Stem Cells/cytology , Neurons/metabolism , Niemann-Pick Disease, Type C/metabolism , Adult , Cells, Cultured , Child , Child, Preschool , Hepatocytes/cytology , Humans , Neurons/cytology
13.
Cell Stem Cell ; 13(4): 446-58, 2013 Oct 03.
Article in English | MEDLINE | ID: mdl-24094325

ABSTRACT

Rett syndrome (RTT) is caused by mutations of MECP2, a methyl CpG binding protein thought to act as a global transcriptional repressor. Here we show, using an isogenic human embryonic stem cell model of RTT, that MECP2 mutant neurons display key molecular and cellular features of this disorder. Unbiased global gene expression analyses demonstrate that MECP2 functions as a global activator in neurons but not in neural precursors. Decreased transcription in neurons was coupled with a significant reduction in nascent protein synthesis and lack of MECP2 was manifested as a severe defect in the activity of the AKT/mTOR pathway. Lack of MECP2 also leads to impaired mitochondrial function in mutant neurons. Activation of AKT/mTOR signaling by exogenous growth factors or by depletion of PTEN boosted protein synthesis and ameliorated disease phenotypes in mutant neurons. Our findings indicate a vital function for MECP2 in maintaining active gene transcription in human neuronal cells.


Subject(s)
Embryonic Stem Cells/pathology , Methyl-CpG-Binding Protein 2/metabolism , Neurons/pathology , Protein Biosynthesis/genetics , Rett Syndrome/genetics , Rett Syndrome/pathology , Transcription, Genetic/genetics , Cells, Cultured , Embryonic Stem Cells/metabolism , Humans , Mutation , Neurons/metabolism
14.
Science ; 342(6161): 983-7, 2013 Nov 22.
Article in English | MEDLINE | ID: mdl-24158904

ABSTRACT

The induced pluripotent stem (iPS) cell field holds promise for in vitro disease modeling. However, identifying innate cellular pathologies, particularly for age-related neurodegenerative diseases, has been challenging. Here, we exploited mutation correction of iPS cells and conserved proteotoxic mechanisms from yeast to humans to discover and reverse phenotypic responses to α-synuclein (αsyn), a key protein involved in Parkinson's disease (PD). We generated cortical neurons from iPS cells of patients harboring αsyn mutations, who are at high risk of developing PD dementia. Genetic modifiers from unbiased screens in a yeast model of αsyn toxicity led to identification of early pathogenic phenotypes in patient neurons. These included nitrosative stress, accumulation of endoplasmic reticulum (ER)-associated degradation substrates, and ER stress. A small molecule identified in a yeast screen (NAB2), and the ubiquitin ligase Nedd4 it affects, reversed pathologic phenotypes in these neurons.


Subject(s)
Benzimidazoles/pharmacology , Neurons/drug effects , Parkinson Disease/metabolism , alpha-Synuclein/metabolism , Animals , Benzimidazoles/chemistry , Endoplasmic Reticulum Stress/drug effects , Female , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Mutation , Neurogenesis , Neurons/metabolism , Neurons/pathology , Parkinson Disease/genetics , Rats , alpha-Synuclein/genetics
15.
Cell Stem Cell ; 11(1): 75-90, 2012 Jul 06.
Article in English | MEDLINE | ID: mdl-22770242

ABSTRACT

Although human induced pluripotent stem cells (hiPSCs) have enormous potential in regenerative medicine, their epigenetic variability suggests that some lines may not be suitable for human therapy. There are currently few benchmarks for assessing quality. Here we show that X-inactivation markers can be used to separate hiPSC lines into distinct epigenetic classes and that the classes are phenotypically distinct. Loss of XIST expression is strongly correlated with upregulation of X-linked oncogenes, accelerated growth rate in vitro, and poorer differentiation in vivo. Whereas differences in X-inactivation potential result in epigenetic variability of female hiPSC lines, male hiPSC lines generally resemble each other and do not overexpress the oncogenes. Neither physiological oxygen levels nor HDAC inhibitors offer advantages to culturing female hiPSC lines. We conclude that female hiPSCs may be epigenetically less stable in culture and caution that loss of XIST may result in qualitatively less desirable stem cell lines.


Subject(s)
Gene Expression Profiling , Genes, Neoplasm/genetics , Induced Pluripotent Stem Cells/metabolism , Neoplasms/genetics , Sex Characteristics , Animals , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line , Cell Proliferation/drug effects , Chromosomes, Human, X/genetics , Female , Genome, Human/genetics , Histone Deacetylase Inhibitors/pharmacology , Humans , Induced Pluripotent Stem Cells/drug effects , Male , Mice , Neoplasms/pathology , Oligonucleotide Array Sequence Analysis , Oxygen/pharmacology , RNA, Long Noncoding , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , Up-Regulation/drug effects , Up-Regulation/genetics , X Chromosome Inactivation/drug effects , X Chromosome Inactivation/genetics
16.
Cell Stem Cell ; 4(6): 513-24, 2009 Jun 05.
Article in English | MEDLINE | ID: mdl-19427283

ABSTRACT

Embryonic stem cells (ESCs) are isolated from the inner cell mass (ICM) of blastocysts, whereas epiblast stem cells (EpiSCs) are derived from the postimplantation epiblast and display a restricted developmental potential. Here we characterize pluripotent states in the nonobese diabetic (NOD) mouse strain, which prior to this study was considered "nonpermissive" for ESC derivation. We find that NOD stem cells can be stabilized by providing constitutive expression of Klf4 or c-Myc or small molecules that can replace these factors during in vitro reprogramming. The NOD ESCs and iPSCs appear to be "metastable," as they acquire an alternative EpiSC-like identity after removal of the exogenous factors, while their reintroduction converts the cells back to ICM-like pluripotency. Our findings suggest that stem cells from different genetic backgrounds can assume distinct states of pluripotency in vitro, the stability of which is regulated by endogenous genetic determinants and can be modified by exogenous factors.


Subject(s)
Hemostasis , Pluripotent Stem Cells/cytology , Animals , Cell Dedifferentiation , Embryonic Stem Cells/cytology , Germ Layers/cytology , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Mice , Mice, Inbred NOD , Mice, Transgenic , Proto-Oncogene Proteins c-myc/genetics
17.
Nat Biotechnol ; 27(9): 851-7, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19680244

ABSTRACT

Realizing the full potential of human embryonic stem cells (hESCs) and induced pluripotent stem cells (hiPSCs) requires efficient methods for genetic modification. However, techniques to generate cell type-specific lineage reporters, as well as reliable tools to disrupt, repair or overexpress genes by gene targeting, are inefficient at best and thus are not routinely used. Here we report the highly efficient targeting of three genes in human pluripotent cells using zinc-finger nuclease (ZFN)-mediated genome editing. First, using ZFNs specific for the OCT4 (POU5F1) locus, we generated OCT4-eGFP reporter cells to monitor the pluripotent state of hESCs. Second, we inserted a transgene into the AAVS1 locus to generate a robust drug-inducible overexpression system in hESCs. Finally, we targeted the PITX3 gene, demonstrating that ZFNs can be used to generate reporter cells by targeting non-expressed genes in hESCs and hiPSCs.


Subject(s)
Deoxyribonucleases/metabolism , Embryonic Stem Cells/physiology , Gene Targeting/methods , Pluripotent Stem Cells/physiology , Zinc Fingers/physiology , Cell Line , Deoxyribonucleases/genetics , Gene Expression , Gene Silencing , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Immunohistochemistry , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
18.
Nano Lett ; 8(10): 3126-30, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18754690

ABSTRACT

We develop biodegradable polymeric nanoparticles to facilitate nonviral gene transfer to human embryonic stem cells (hESCs). Small (approximately 200 nm), positively charged (approximately 10 mV) particles are formed by the self assembly of cationic, hydrolytically degradable poly(beta-amino esters) and plasmid DNA. By varying the end group of the polymer, we can tune the biophysical properties of the resulting nanoparticles and their gene-delivery efficacy. We created an OCT4-driven GFP hES cell line to allow the rapid identification of nanoparticles that facilitate gene transfer while maintaining an hESC undifferentiated state. Using this cell system, we synthesized nanoparticles that have gene delivery efficacy that is up to 4 times higher than that of the leading commercially available transfection agent, Lipofectamine 2000. Importantly, these materials have minimal toxicity and do not adversely affect hESC colony morphology or cause nonspecific differentiation.


Subject(s)
Embryonic Stem Cells/cytology , Gene Transfer Techniques , Genetic Vectors/chemistry , Animals , Biocompatible Materials/chemistry , Cations , Cell Differentiation , Flow Cytometry , Genetic Techniques , Green Fluorescent Proteins/metabolism , Hydrolysis , Mice , Nanotechnology/methods , Octamer Transcription Factor-3/metabolism , Polymers/chemistry
19.
Cell Stem Cell ; 3(3): 346-353, 2008 Sep 11.
Article in English | MEDLINE | ID: mdl-18786421

ABSTRACT

Current approaches to reprogram human somatic cells to pluripotent iPSCs utilize viral transduction of different combinations of transcription factors. These protocols are highly inefficient because only a small fraction of cells carry the appropriate number and stoichiometry of proviral insertions to initiate the reprogramming process. Here we have generated genetically homogeneous "secondary" somatic cells, which carry the reprogramming factors as defined doxycycline (DOX)-inducible transgenes. These cells were obtained by infecting fibroblasts with DOX-inducible lentiviruses, isolating "primary" iPSCs in the presence of the drug, and finally differentiating to "secondary" fibroblasts. When "secondary" fibroblast lines were cultured in the presence of DOX without further viral infection, up to 2% of the cells were reprogrammed to pluripotent "secondary" human iPSCs. This system will facilitate the characterization of the reprogramming process and provides a unique platform for genetic or chemical screens to enhance reprogramming or replace individual factors.


Subject(s)
Cell Dedifferentiation , Cellular Reprogramming , Cytological Techniques , Pluripotent Stem Cells/metabolism , Doxycycline/metabolism , Fibroblasts/metabolism , Genetic Techniques , Genetic Vectors/metabolism , Humans , Lentivirus/genetics
20.
Stem Cells ; 24(1): 125-38, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16100006

ABSTRACT

Research on the cell fate determination of embryonic stem cells is of enormous interest given the therapeutic potential in regenerative cell therapy. Human embryonic stem cells (hESCs) have the ability to renew themselves and differentiate into all three germ layers. The main focus of this study was to examine factors affecting derivation and further proliferation of multipotent neuroepithelial (NEP) cells from hESCs. hESCs cultured in serum-deprived defined medium developed distinct tube structures and could be isolated either by dissociation or adherently. Dissociated cells survived to form colonies of cells characterized as NEP when conditioned medium from human hepatocellular carcinoma HepG2 cell line (MEDII) was added. However, cells isolated adherently developed an enriched population of NEP cells independent of MEDII medium. Further characterization suggested that they were NEP cells because they had a similar phenotype profile to in vivo NEP cells and expression SOX1, SOX2, and SOX3 genes. They were positive for Nestin, a neural intermediate filament protein, and Musashi-1, a neural RNA-binding protein, but few cells expressed further differentiation markers, such as PSNCAM, A2B5, MAPII, GFAP, or O4, or other lineage markers, such as muscle actin, alpha fetoprotein, or the pluripotent marker Oct4. Further differentiation of these putative NEP cells gave rise to a mixed population of progenitors that included A2B5-positive and PSNCAM-positive cells and postmitotic neurons and astrocytes. To proliferate and culture these derived NEP cells, ideal conditions were obtained using neurobasal medium supplemented with B27 and basic fibroblast growth factor in 5% oxygen. NEP cells were continuously propagated for longer than 6 months without losing their multipotent cell characteristics and maintained a stable chromosome number.


Subject(s)
Cell Proliferation/drug effects , Neuroepithelial Cells/physiology , Stem Cells/physiology , Tissue Culture Techniques/methods , Animals , Cell Adhesion , Cell Count , Cell Differentiation/drug effects , Cell Line , Cell Survival/drug effects , Culture Media, Conditioned , Growth Substances/pharmacology , Humans , Mice , Oxygen/pharmacology , Rosette Formation
SELECTION OF CITATIONS
SEARCH DETAIL