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










Publication year range
1.
Biomaterials ; 136: 1-11, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28505596

ABSTRACT

Cell replacement therapies have broad biomedical potential; however, low cell survival and poor functional integration post-transplantation are major hurdles that hamper clinical benefit. For example, following striatal transplantation of midbrain dopaminergic (mDA) neurons for the treatment of Parkinson's disease (PD), only 1-5% of the neurons typically survive in preclinical models and in clinical trials. In general, resource-intensive generation and implantation of larger numbers of cells are used to compensate for the low post-transplantation cell-survival. Poor graft survival is often attributed to adverse biochemical, mechanical, and/or immunological stress that cells experience during and after implantation. To address these challenges, we developed a functionalized hyaluronic acid (HA)-based hydrogel for in vitro maturation and central nervous system (CNS) transplantation of human pluripotent stem cell (hPSC)-derived neural progenitors. Specifically, we functionalized the HA hydrogel with RGD and heparin (hep) via click-chemistry and tailored its stiffness to encourage neuronal maturation, survival, and long-term maintenance of the desired mDA phenotype. Importantly, ∼5 times more hydrogel-encapsulated mDA neurons survived after transplantation in the rat striatum, compared to unencapsulated neurons harvested from commonly used 2D surfaces. This engineered biomaterial may therefore increase the therapeutic potential and reduce the manufacturing burden for successful neuronal implantation.


Subject(s)
Dopaminergic Neurons/cytology , Dopaminergic Neurons/transplantation , Embryonic Stem Cells/cytology , Hyaluronic Acid/chemistry , Hydrogels/chemistry , Tissue Scaffolds/chemistry , Animals , Cell Line , Cell Survival , Cells, Cultured , Female , Heparin/chemistry , Humans , Mesencephalon/cytology , Neural Stem Cells/cytology , Neural Stem Cells/transplantation , Neurogenesis , Oligopeptides/chemistry , Rats, Inbred F344
2.
Theranostics ; 6(3): 291-301, 2016.
Article in English | MEDLINE | ID: mdl-26909106

ABSTRACT

Stem cell therapies have enormous potential for treating many debilitating diseases, including heart failure, stroke and traumatic brain injury. For maximal efficacy, these therapies require targeted cell delivery to specific tissues followed by successful cell engraftment. However, targeted delivery remains an open challenge. As one example, it is common for intravenous deliveries of mesenchymal stem cells (MSCs) to become entrapped in lung microvasculature instead of the target tissue. Hence, a robust, quantitative imaging method would be essential for developing efficacious cell therapies. Here we show that Magnetic Particle Imaging (MPI), a novel technique that directly images iron-oxide nanoparticle-tagged cells, can longitudinally monitor and quantify MSC administration in vivo. MPI offers near-ideal image contrast, depth penetration, and robustness; these properties make MPI both ultra-sensitive and linearly quantitative. Here, we imaged, for the first time, the dynamic trafficking of intravenous MSC administrations using MPI. Our results indicate that labeled MSC injections are immediately entrapped in lung tissue and then clear to the liver within one day, whereas standard iron oxide particle (Resovist) injections are immediately taken up by liver and spleen. Longitudinal MPI-CT imaging also indicated a clearance half-life of MSC iron oxide labels in the liver at 4.6 days. Finally, our ex vivo MPI biodistribution measurements of iron in liver, spleen, heart, and lungs after injection showed excellent agreement (R(2) = 0.943) with measurements from induction coupled plasma spectrometry. These results demonstrate that MPI offers strong utility for noninvasively imaging and quantifying the systemic distribution of cell therapies and other therapeutic agents.


Subject(s)
Diagnostic Imaging/methods , Ferric Compounds/analysis , Magnetics , Mesenchymal Stem Cell Transplantation , Administration, Intravenous , Animals , Female , Humans , Mice , Nanoparticles/analysis , Rats, Inbred F344 , Staining and Labeling , Tissue Distribution
3.
Sci Rep ; 5: 14055, 2015 Sep 11.
Article in English | MEDLINE | ID: mdl-26358296

ABSTRACT

We demonstrate that Magnetic Particle Imaging (MPI) enables monitoring of cellular grafts with high contrast, sensitivity, and quantitativeness. MPI directly detects the intense magnetization of iron-oxide tracers using low-frequency magnetic fields. MPI is safe, noninvasive and offers superb sensitivity, with great promise for clinical translation and quantitative single-cell tracking. Here we report the first MPI cell tracking study, showing 200-cell detection in vitro and in vivo monitoring of human neural graft clearance over 87 days in rat brain.


Subject(s)
Cell Tracking , Magnetic Resonance Imaging , Magnetite Nanoparticles , Animals , Cell Differentiation , Cell Tracking/methods , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Female , Humans , Iron/metabolism , Magnetic Resonance Imaging/methods , Rats , Reproducibility of Results , Sensitivity and Specificity , Staining and Labeling
4.
Development ; 142(10): 1885-92, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25968319

ABSTRACT

Neural stem cells (NSCs) are defined by their ability to self-renew and to differentiate into mature neuronal and glial cell types. NSCs are the subject of intense investigation, owing to their crucial roles in neural development and adult brain function and because they present potential targets for gene and cell replacement therapies following injury or disease. Approaches to specifically genetically perturb or modulate NSC function would be valuable for either motivation. Unfortunately, most gene delivery vectors are incapable of efficient or specific gene delivery to NSCs in vivo. Vectors based on adeno-associated virus (AAV) present a number of advantages and have proven increasingly successful in clinical trials. However, natural AAV variants are inefficient in transducing NSCs. We previously engineered a novel AAV variant (AAV r3.45) capable of efficient transduction of adult NSCs in vitro. Here, to build upon the initial promise of this variant, we investigated its in vitro and in vivo infectivity. AAV r3.45 was more selective for NSCs than mature neurons in a human embryonic stem cell-derived culture containing a mixture of cell types, including NSCs and neurons. It was capable of more efficient and selective transduction of rat and mouse NSCs in vivo than natural AAV serotypes following intracranial vector administration. Delivery of constitutively active ß-catenin yielded insights into mechanisms by which this key regulator modulates NSC function, indicating that this engineered AAV variant can be harnessed for preferential modulation of adult NSCs in the hippocampus. The capacity to rapidly genetically modify these cells might greatly accelerate in vivo investigations of adult neurogenesis.


Subject(s)
Dependovirus/genetics , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Animals , Cell Line , Gene Transfer Techniques , Humans , Mice , Rats , beta Catenin/metabolism
5.
Biomaterials ; 35(3): 941-8, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24172856

ABSTRACT

Stem cell differentiation is regulated by complex repertoires of signaling ligands which often use multivalent interactions, where multiple ligands tethered to one entity interact with multiple cellular receptors to yield oligomeric complexes. One such ligand is Sonic hedgehog (Shh), whose posttranslational lipid modifications and assembly into multimers enhance its biological potency, potentially through receptor clustering. Investigations of Shh typically utilize recombinant, monomeric protein, and thus the impact of multivalency on ligand potency is unexplored. Among its many activities, Shh is required for ventralization of the midbrain and forebrain and is therefore critical for the development of midbrain dopaminergic (mDA) and forebrain gamma-aminobutyric acid (GABA) inhibitory neurons. We have designed multivalent biomaterials presenting Shh in defined spatial arrangements and investigated the role of Shh valency in ventral specification of human embryonic stem cells (hESCs) into these therapeutically relevant cell types. Multivalent Shh conjugates with optimal valencies, compared to the monomeric Shh, increased the percentages of neurons belonging to mDA or forebrain GABAergic fates from 33% to 60% or 52% to 86%, respectively. Thus, multivalent Shh bioconjugates can enhance neuronal lineage commitment of pluripotent stem cells and thereby facilitate efficient derivation of neurons that could be used to treat Parkinson's and epilepsy patients.


Subject(s)
Biocompatible Materials/metabolism , Dopaminergic Neurons/cytology , Embryonic Stem Cells/cytology , GABAergic Neurons/cytology , Hedgehog Proteins/metabolism , Animals , Biocompatible Materials/chemistry , Cell Differentiation , Cell Line , Dopaminergic Neurons/metabolism , Embryonic Stem Cells/metabolism , GABAergic Neurons/metabolism , Hedgehog Proteins/chemistry , Humans , Hyaluronic Acid/chemistry , Hyaluronic Acid/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism
6.
Neurobiol Dis ; 62: 62-72, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24055772

ABSTRACT

Alzheimer's disease (AD) is among the most prevalent forms of dementia affecting the aging population, and pharmacological therapies to date have not been successful in preventing disease progression. Future therapeutic efforts may benefit from the development of models that enable basic investigation of early disease pathology. In particular, disease-relevant models based on human pluripotent stem cells (hPSCs) may be promising approaches to assess the impact of neurotoxic agents in AD on specific neuronal populations and thereby facilitate the development of novel interventions to avert early disease mechanisms. We implemented an efficient paradigm to convert hPSCs into enriched populations of cortical glutamatergic neurons emerging from dorsal forebrain neural progenitors, aided by modulating Sonic hedgehog (Shh) signaling. Since AD is generally known to be toxic to glutamatergic circuits, we exposed glutamatergic neurons derived from hESCs to an oligomeric pre-fibrillar forms of Aß known as "globulomers", which have shown strong correlation with the level of cognitive deficits in AD. Administration of such Aß oligomers yielded signs of the disease, including cell culture age-dependent binding of Aß and cell death in the glutamatergic populations. Furthermore, consistent with previous findings in postmortem human AD brain, Aß-induced toxicity was selective for glutamatergic rather than GABAeric neurons present in our cultures. This in vitro model of cortical glutamatergic neurons thus offers a system for future mechanistic investigation and therapeutic development for AD pathology using human cell types specifically affected by this disease.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/toxicity , Glutamic Acid/metabolism , Neurons/drug effects , Neurons/physiology , Pluripotent Stem Cells/cytology , Age Factors , Amyloid beta-Peptides/metabolism , Animals , Cell Death , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Cerebral Cortex/physiology , Female , GABAergic Neurons/drug effects , GABAergic Neurons/pathology , GABAergic Neurons/physiology , Hedgehog Proteins/metabolism , Humans , Neurons/pathology , Rats, Inbred F344
7.
Nat Nanotechnol ; 8(11): 831-8, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24141540

ABSTRACT

There is broad interest in designing nanostructured materials that can interact with cells and regulate key downstream functions. In particular, materials with nanoscale features may enable control over multivalent interactions, which involve the simultaneous binding of multiple ligands on one entity to multiple receptors on another and are ubiquitous throughout biology. Cellular signal transduction of growth factor and morphogen cues (which have critical roles in regulating cell function and fate) often begins with such multivalent binding of ligands, either secreted or cell-surface-tethered to target cell receptors, leading to receptor clustering. Cellular mechanisms that orchestrate ligand-receptor oligomerization are complex, however, so the capacity to control multivalent interactions and thereby modulate key signalling events within living systems is currently very limited. Here, we demonstrate the design of potent multivalent conjugates that can organize stem cell receptors into nanoscale clusters and control stem cell behaviour in vitro and in vivo. The ectodomain of ephrin-B2, normally an integral membrane protein ligand, was conjugated to a soluble biopolymer to yield multivalent nanoscale conjugates that potently induce signalling in neural stem cells and promote their neuronal differentiation both in culture and within the brain. Super-resolution microscopy analysis yielded insights into the organization of the receptor-ligand clusters at the nanoscale. We also found that synthetic multivalent conjugates of ephrin-B1 strongly enhance human embryonic and induced pluripotent stem cell differentiation into functional dopaminergic neurons. Multivalent bioconjugates are therefore powerful tools and potential nanoscale therapeutics for controlling the behaviour of target stem cells in vitro and in vivo.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/cytology , Ephrin-B2/pharmacology , Nanoconjugates/chemistry , Neural Stem Cells/cytology , Animals , Brain/cytology , Brain/drug effects , Cells, Cultured , Humans , Ligands , Mice , Neurons/cytology , Neurons/drug effects , Receptors, Eph Family/metabolism , Recombinant Proteins/metabolism , Signal Transduction
8.
Aging (Albany NY) ; 5(5): 357-72, 2013 May.
Article in English | MEDLINE | ID: mdl-23793469

ABSTRACT

This work builds upon our findings that proteins secreted by hESCs exhibit pro-regenerative activity, and determines that hESC-conditioned medium robustly enhances the proliferation of both muscle and neural progenitor cells. Importantly, this work establishes that it is the proteins that bind heparin which are responsible for the pro-myogenic effects of hESC-conditioned medium, and indicates that this strategy is suitable for enriching the potentially therapeutic factors. Additionally, this work shows that hESC-secreted proteins act independently of the mitogen FGF-2, and suggests that FGF-2 is unlikely to be a pro-aging molecule in the physiological decline of old muscle repair. Moreover, hESC-secreted factors improve the viability of human cortical neurons in an Alzheimer's disease (AD) model, suggesting that these factors can enhance the maintenance and regeneration of multiple tissues in the aging body.


Subject(s)
Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental/physiology , Heparin/metabolism , Aging/metabolism , Aging/physiology , Animals , Culture Media, Conditioned/chemistry , Culture Media, Conditioned/pharmacology , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Heparin/chemistry , Humans , Mice , Mice, Inbred C57BL , Muscle Development/drug effects , Muscle, Skeletal/injuries , Muscle, Skeletal/physiology , Neurons/drug effects , Neurons/physiology , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/drug effects , Satellite Cells, Skeletal Muscle/physiology
9.
Mol Ther ; 20(2): 329-38, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22108859

ABSTRACT

Efficient approaches for the precise genetic engineering of human pluripotent stem cells (hPSCs) can enhance both basic and applied stem cell research. Adeno- associated virus (AAV) vectors are of particular interest for their capacity to mediate efficient gene delivery to and gene targeting in various cells. However, natural AAV serotypes offer only modest transduction of human embryonic and induced pluripotent stem cells (hESCs and hiPSCs), which limits their utility for efficiently manipulating the hPSC genome. Directed evolution is a powerful means to generate viral vectors with novel capabilities, and we have applied this approach to create a novel AAV variant with high gene delivery efficiencies (~50%) to hPSCs, which are importantly accompanied by a considerable increase in gene-targeting frequencies, up to 0.12%. While this level is likely sufficient for numerous applications, we also show that the gene-targeting efficiency mediated by an evolved AAV variant can be further enhanced (>1%) in the presence of targeted double- stranded breaks (DSBs) generated by the co-delivery of artificial zinc finger nucleases (ZFNs). Thus, this study demonstrates that under appropriate selective pressures, AAV vectors can be created to mediate efficient gene targeting in hPSCs, alone or in the presence of ZFN- mediated double-stranded DNA breaks.


Subject(s)
Dependovirus/genetics , Directed Molecular Evolution , Gene Targeting , Genetic Vectors/genetics , Pluripotent Stem Cells/metabolism , Transduction, Genetic , Base Sequence , Capsid Proteins/genetics , Cell Line , DNA Breaks, Double-Stranded , Dependovirus/physiology , Endonucleases/genetics , Gene Expression , Gene Library , Gene Order , Gene Transfer Techniques , Humans , Molecular Sequence Data , Mutation , Viral Tropism , Zinc Fingers/genetics
10.
Mol Ther ; 19(4): 667-75, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21224831

ABSTRACT

Gene delivery to, and gene targeting in, stem cells would be a highly enabling technology for basic science and biomedical application. Adeno-associated viral (AAV) vectors have demonstrated the capacity for efficient delivery to numerous cells, but their application to stem cells has been limited by low transduction efficiency. Due to their considerable advantages, however, engineering AAV delivery systems to enhance gene delivery to stem cells may have an impact in stem cell biology and therapy. Therefore, using several diverse AAV capsid libraries-including randomly mutagenized, DNA shuffled, and random peptide insertion variants-we applied directed evolution to create a "designer" AAV vector with enhanced delivery efficiency for neural stem cells (NSCs). A novel AAV variant, carrying an insertion of a selected peptide sequence on the surface of the threefold spike within the heparin-binding site, emerged from this evolution. Importantly, this evolved AAV variant mediated efficient gene delivery to rat, mouse, and human NSCs, as well as efficient gene targeting within adult NSCs, and it is thus promising for applications ranging from basic stem cell biology to clinical translation.


Subject(s)
Dependovirus/genetics , Neural Stem Cells/virology , Animals , Cells, Cultured , Chromatography , Female , Fluorescent Antibody Technique , Gene Transfer Techniques , Polymerase Chain Reaction , Rats , Rats, Inbred F344
11.
Restor Neurol Neurosci ; 28(4): 589-603, 2010.
Article in English | MEDLINE | ID: mdl-20714081

ABSTRACT

The greatest therapeutic promise of human embryonic stem cells (hESC) is to generate specialized cells to replace damaged tissue in patients suffering from various degenerative diseases. However, the signaling mechanisms involved in lineage restriction of ESC to adopt various cellular phenotypes are still under investigation. Furthermore, for progression of hESC-based therapies towards clinical applications, appropriate culture conditions must be developed to generate genetically stable homogenous populations of cells, to hinder possible adverse effects following transplantation. Other critical challenges that must be addressed for successful cell implantation include problems related to survival and functional efficacy of the grafted cells. This review initially describes the derivation of hESC and focuses on recent advances in generation, characterization, and maintenance of these cells. We also give an overview of original and emerging differentiation strategies used to convert hESC to different cell types. Finally, we will discuss transplantation studies of hESC-derived cells with respect to safety and functional recovery.


Subject(s)
Embryonic Stem Cells/physiology , Cell Differentiation , Cell Lineage , Cells, Cultured , Embryonic Stem Cells/transplantation , Humans , Pluripotent Stem Cells/physiology
12.
Trends Biotechnol ; 28(3): 117-24, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20042248

ABSTRACT

The stem cell niche is an anatomical site that contains a reservoir of multipotent stem cells (SCs) that can maintain normal tissue, or replenish injured or aged cell populations, in response to mechanisms that regulate whether they should remain quiescent, undergo self-renewal, or differentiate. The choice among these hallmark SC behaviors is governed by intricate soluble and "solid phase" signals that are systemic or presented by the local niche cells. In this review, we discuss the progress achieved in understanding the mechanisms and principles that govern microenvironmental regulation of SC behavior, and focus on novel approaches that have been developed to synthesize this basic information to engineer creative strategies for harnessing and controlling SCs ex vivo and in vivo.


Subject(s)
Cell Culture Techniques/methods , Models, Biological , Stem Cells/physiology , Tissue Engineering/methods , Animals , Biomimetic Materials , Drosophila/cytology , Humans , Microfluidic Analytical Techniques/instrumentation , Microfluidic Analytical Techniques/methods , Stem Cells/cytology
13.
Restor Neurol Neurosci ; 27(4): 359-70, 2009.
Article in English | MEDLINE | ID: mdl-19738328

ABSTRACT

PURPOSE: Human embryonic stem cells (hESCs) which express a reporter gene consistently during all phases of differentiation would be valuable for basic research on cell transplantation. In this study, we describe karyotypically-abnormal variant hESCs, BGO1V2-EFG, which express hrGFP driven by the EF1 promoter. METHODS: BGO1V2-EFG cells were analyzed by using immunocytochemistry, single cell-based confocal image, and in vitro differentiation, including dopaminergic differentiation. RESULTS: Undifferentiated BGO1V2-EFG cells expressed pluripotent ESC markers and retained the ability to differentiate into cell types of all three germ layers. BGO1V2-EFG cells maintained stable and robust hrGFP expression in vitro in the undifferentiated state and during differentiation. The EF1 promoter retained activity during dopaminergic differentiation, as 76% of tyrosine hydroxlase (TH)-positive cells co-expressed hrGFP by confocal analysis. Treated with sodium butyrate (0.02 mM to 2.0 mM), an inhibitor of histone deacetylase (HDAC), during differentiation did not affect hrGFP expression, although TH expression was reduced by higher concentrations of sodium butyrate. CONCLUSION: BGO1V2-EFG cells maintain stable and robust hrGFP expression in the undifferentiated state and during neural differentiation. Especially, the EF1 promoter was effective in driving hrGFP expression during dopaminergic differentiation. BGO1V2-EFG cells may be useful for transplantation studies in Parkinson disease animal models.


Subject(s)
Cell Differentiation/physiology , Dopamine/metabolism , Embryonic Stem Cells/physiology , Green Fluorescent Proteins/genetics , Butyrates/pharmacology , Cell Differentiation/drug effects , Cell Line , Dopamine/genetics , Dose-Response Relationship, Drug , Embryonic Stem Cells/drug effects , Enzyme Inhibitors , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/genetics , Humans , Microscopy, Confocal/methods , Peptide Elongation Factor 1/genetics , Promoter Regions, Genetic , Transfection/methods , Tyrosine 3-Monooxygenase/metabolism
14.
PLoS One ; 4(9): e6997, 2009 Sep 14.
Article in English | MEDLINE | ID: mdl-19750008

ABSTRACT

With recent findings on the role of reprogramming factors on stem cells, in vitro screening assays for studying (de)-differentiation is of great interest. We developed a miniaturized stem cell screening chip that is easily accessible and provides means of rapidly studying thousands of individual stem/progenitor cell samples, using low reagent volumes. For example, screening of 700,000 substances would take less than two days, using this platform combined with a conventional bio-imaging system. The microwell chip has standard slide format and consists of 672 wells in total. Each well holds 500 nl, a volume small enough to drastically decrease reagent costs but large enough to allow utilization of standard laboratory equipment. Results presented here include weeklong culturing and differentiation assays of mouse embryonic stem cells, mouse adult neural stem cells, and human embryonic stem cells. The possibility to either maintain the cells as stem/progenitor cells or to study cell differentiation of stem/progenitor cells over time is demonstrated. Clonality is critical for stem cell research, and was accomplished in the microwell chips by isolation and clonal analysis of single mouse embryonic stem cells using flow cytometric cell-sorting. Protocols for practical handling of the microwell chips are presented, describing a rapid and user-friendly method for the simultaneous study of thousands of stem cell cultures in small microwells. This microwell chip has high potential for a wide range of applications, for example directed differentiation assays and screening of reprogramming factors, opening up considerable opportunities in the stem cell field.


Subject(s)
Oligonucleotide Array Sequence Analysis , Stem Cells/cytology , Animals , Cell Culture Techniques , Cell Differentiation , Cell Separation/methods , Embryonic Stem Cells/cytology , Equipment Design , Female , Flow Cytometry/methods , Humans , Mice , Mice, Inbred C57BL , Neurons/metabolism
15.
PLoS One ; 4(8): e6606, 2009 Aug 12.
Article in English | MEDLINE | ID: mdl-19672298

ABSTRACT

BACKGROUND: Stromal-Derived Inducing Activity (SDIA) is one of the most efficient methods of generating dopaminergic (DA) neurons from embryonic stem cells (ESC). DA neuron induction can be achieved by co-culturing ESC with the mouse stromal cell lines PA6 or MS5. The molecular nature of this effect, which has been termed "SDIA" is so far unknown. Recently, we found that factors secreted by PA6 cells provided lineage-specific instructions to induce DA differentiation of human ESC (hESC). METHODOLOGY/PRINCIPAL FINDINGS: In the present study, we compared PA6 cells to various cell lines lacking the SDIA effect, and employed genome expression analysis to identify differentially-expressed signaling molecules. Among the factors highly expressed by PA6 cells, and known to be associated with CNS development, were stromal cell-derived factor 1 (SDF-1/CXCL12), pleiotrophin (PTN), insulin-like growth factor 2 (IGF2), and ephrin B1 (EFNB1). When these four factors, the combination of which was termed SPIE, were applied to hESC, they induced differentiation to TH-positive neurons in vitro. RT-PCR and western blot analysis confirmed the expression of midbrain specific markers, including engrailed 1, Nurr1, Pitx3, and dopamine transporter (DAT) in cultures influenced by these four molecules. Electrophysiological recordings showed that treatment of hESC with SPIE induced differentiation of neurons that were capable of generating action potentials and forming functional synaptic connections. CONCLUSIONS/SIGNIFICANCE: The combination of SDF-1, PTN, IGF2, and EFNB1 mimics the DA phenotype-inducing property of SDIA and was sufficient to promote differentiation of hESC to functional midbrain DA neurons. These findings provide a method for differentiating hESC to form DA neurons, without a requirement for the use of animal-derived cell lines or products.


Subject(s)
Carrier Proteins/physiology , Cell Differentiation/physiology , Chemokine CXCL12/physiology , Cytokines/physiology , Dopamine/physiology , Embryonic Stem Cells/cytology , Ephrin-B1/physiology , Insulin-Like Growth Factor II/physiology , Neurons/cytology , Animals , Blotting, Western , Coculture Techniques , Embryonic Stem Cells/metabolism , Humans , Mice , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction
16.
Restor Neurol Neurosci ; 26(6): 447-58, 2008.
Article in English | MEDLINE | ID: mdl-19096132

ABSTRACT

BACKGROUND AND PURPOSE: Human embryonic stem cells (hESC) are considered a renewable source of dopamine producing neurons, and are of particular interest for their potential clinical use in Parkinson's disease. In this study, we characterized human dopaminergic neurons generated by stromal-derived inducing activity (SDIA) from BG01V2, a strain of human embryonic stem cell line, BG01, characterized by a chromosome 17 trisomy. Similar chromosomal changes have been repeatedly observed in hESC cultures in different laboratories, indicating the importance of chromosome 17 for growth and adaptation of hESC to culture. METHODS: We investigated in vitro proliferation of differentiating cells using a BrDU incorporation assay, and monitored the cell population in long term cultures. Despite the cytogenetic abnormality, TH+ neurons were postmitotic at all stages of differentiation. After 30 days of differentiation, cell division ceased in 91% of the overall population of cells in the culture, indicating intact cell cycle regulation. RESULTS: Expression of midbrain specific marker genes (Otx2, Pax5, Msx-1) showed differentiation of hESC-derived neural progenitor cells into midbrain specific dopamine neurons. These neurons expressed the dopamine transporter (DAT), and displayed functional DAT activity and electrical excitability. CONCLUSIONS: TH+ cells derived from the BG01V2 hESC line using SDIA are postmitotic and have functional characteristics of normal dopaminergic neurons.


Subject(s)
Cell Differentiation/physiology , Dopamine/metabolism , Embryonic Stem Cells/physiology , Neurons/physiology , Actins/metabolism , Bromodeoxyuridine/metabolism , Cell Line , Cell Proliferation , Chromosomes, Human, Pair 17 , Cocaine/analogs & derivatives , Cocaine/pharmacokinetics , Coculture Techniques , Dopamine Plasma Membrane Transport Proteins/metabolism , Humans , Membrane Potentials/physiology , Microtubule-Associated Proteins/metabolism , Nerve Tissue Proteins/metabolism , Otx Transcription Factors/metabolism , Patch-Clamp Techniques/methods , Protein Binding/drug effects , Time Factors , Tritium/pharmacokinetics , Tyrosine 3-Monooxygenase/metabolism
17.
Stem Cells ; 26(6): 1517-25, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18388303

ABSTRACT

Producing dopaminergic (DA) neurons is a major goal of human embryonic stem cell (hESC) research. DA neurons can be differentiated from hESC by coculture with the mouse PA6 stromal cell line; this differentiation-inducing effect is termed stromal-derived inducing activity (SDIA). The molecular and biochemical nature of SDIA is, however, unknown. Various studies have suggested that SDIA involves either a fixation-resistant component located on the PA6 cell surface or factors secreted into the medium by PA6 cells. To address this question, hESC were cocultured with PA6 cells for 12 days and then further differentiated with sonic hedgehog homolog, fibroblast growth factor-8, and glial cell line-derived neurotrophic factor. After 18 days, 34% of cells were tyrosine hydroxylase (TH)+. When PA6 cells were fixed or irradiated, the number of TH+ cells was decreased by threefold, whereas mitomycin-c treatment of feeder cells decreased the number of TH+ cells by 32%. The neural-inducing effect of PA6 cells, as monitored by beta-III-tubulin expression, was minimally affected by mitomycin-c treatment or fixation but was decreased 50% by irradiation. Medium conditioned by PA6 cells was ineffective in differentiating TH+ cells when used alone. Conditioned medium combined with heparin and/or fixed PA6 cells produced TH+ cell differentiation, although less effectively than PA6 cell coculture. Thus, PA6 cell surface activity is required for neural differentiation of hESC, but secreted factors are required for the specific DA neuron-inducing effect. Disclosure of potential conflicts of interest is found at the end of this article.


Subject(s)
Cell Differentiation/physiology , Dopamine/physiology , Embryonic Stem Cells/cytology , Neurons/cytology , Neurons/physiology , Stromal Cells/cytology , Animals , Cell Culture Techniques/methods , Coculture Techniques , Culture Media , Embryonic Stem Cells/physiology , Humans , Immunohistochemistry , Mice , Stromal Cells/physiology
18.
PLoS One ; 3(1): e1422, 2008 Jan 09.
Article in English | MEDLINE | ID: mdl-18183302

ABSTRACT

BACKGROUND: We initiated differentiation of human embryonic stem cells (hESCs) into dopamine neurons, obtained a purified population of neuronal precursor cells by cell sorting, and determined patterns of gene transcription. METHODOLOGY: Dopaminergic differentiation of hESCs was initiated by culturing hESCs with a feeder layer of PA6 cells. Differentiating cells were then sorted to obtain a pure population of PSA-NCAM-expressing neuronal precursors, which were then analyzed for gene expression using Massive Parallel Signature Sequencing (MPSS). Individual genes as well as regions of the genome which were activated were determined. PRINCIPAL FINDINGS: A number of genes known to be involved in the specification of dopaminergic neurons, including MSX1, CDKN1C, Pitx1 and Pitx2, as well as several novel genes not previously associated with dopaminergic differentiation, were expressed. Notably, we found that a specific region of the genome located on chromosome 11p15.5 was highly activated. This region contains several genes which have previously been associated with the function of dopaminergic neurons, including the gene for tyrosine hydroxylase (TH), the rate-limiting enzyme in catecholamine biosynthesis, IGF2, and CDKN1C, which cooperates with Nurr1 in directing the differentiation of dopaminergic neurons. Other genes in this region not previously recognized as being involved in the functions of dopaminergic neurons were also activated, including H19, TSSC4, and HBG2. IGF2 and CDKN1C were also found to be highly expressed in mature human TH-positive dopamine neurons isolated from human brain samples by laser capture. CONCLUSIONS: The present data suggest that the H19-IGF2 imprinting region on chromosome 11p15.5 is involved in the process through which undifferentiated cells are specified to become neuronal precursors and/or dopaminergic neurons.


Subject(s)
Chromosomes, Human, Pair 11 , Dopamine/metabolism , Embryonic Stem Cells/cytology , Gene Expression Profiling , Neurons/cytology , Cell Differentiation/genetics , Cell Line , Cell Separation , Flow Cytometry , Humans , Immunohistochemistry , Neurons/metabolism , Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...