Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
2.
Cell Stem Cell ; 28(2): 343-355.e5, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33545081

ABSTRACT

Human pluripotent stem cells show considerable promise for applications in regenerative medicine, including the development of cell replacement paradigms for the treatment of Parkinson's disease. Protocols have been developed to generate authentic midbrain dopamine (mDA) neurons capable of reversing dopamine-related deficits in animal models of Parkinson's disease. However, the generation of mDA neurons at clinical scale suitable for human application remains an important challenge. Here, we present an mDA neuron derivation protocol based on a two-step WNT signaling activation strategy that improves expression of midbrain markers, such as Engrailed-1 (EN1), while minimizing expression of contaminating posterior (hindbrain) and anterior (diencephalic) lineage markers. The resulting neurons exhibit molecular, biochemical, and electrophysiological properties of mDA neurons. Cryopreserved mDA neuron precursors can be successfully transplanted into 6-hydroxydopamine (6OHDA) lesioned rats to induce recovery of amphetamine-induced rotation behavior. The protocol presented here is the basis for clinical-grade mDA neuron production and preclinical safety and efficacy studies.


Subject(s)
Dopaminergic Neurons , Human Embryonic Stem Cells , Animals , Cell Differentiation , Mesencephalon , Rats , Wnt Signaling Pathway
3.
Nat Commun ; 9(1): 4345, 2018 10 19.
Article in English | MEDLINE | ID: mdl-30341316

ABSTRACT

Environmental and genetic risk factors contribute to Parkinson's Disease (PD) pathogenesis and the associated midbrain dopamine (mDA) neuron loss. Here, we identify early PD pathogenic events by developing methodology that utilizes recent innovations in human pluripotent stem cells (hPSC) and chemical sensors of HSP90-incorporating chaperome networks. We show that events triggered by PD-related genetic or toxic stimuli alter the neuronal proteome, thereby altering the stress-specific chaperome networks, which produce changes detected by chemical sensors. Through this method we identify STAT3 and NF-κB signaling activation as examples of genetic stress, and phospho-tyrosine hydroxylase (TH) activation as an example of toxic stress-induced pathways in PD neurons. Importantly, pharmacological inhibition of the stress chaperome network reversed abnormal phospho-STAT3 signaling and phospho-TH-related dopamine levels and rescued PD neuron viability. The use of chemical sensors of chaperome networks on hPSC-derived lineages may present a general strategy to identify molecular events associated with neurodegenerative diseases.


Subject(s)
Dopaminergic Neurons/metabolism , HSP90 Heat-Shock Proteins/metabolism , Mesencephalon/metabolism , Biosensing Techniques , HSP90 Heat-Shock Proteins/physiology , Mesencephalon/pathology , NF-kappa B/metabolism , STAT3 Transcription Factor/metabolism , Stress, Physiological
4.
Stem Cell Reports ; 9(1): 355-365, 2017 07 11.
Article in English | MEDLINE | ID: mdl-28602613

ABSTRACT

Human pluripotent stem cells (PSCs) provide an unlimited cell source for cell therapies and disease modeling. Despite their enormous power, technical aspects have hampered reproducibility. Here, we describe a modification of PSC workflows that eliminates a major variable for nearly all PSC experiments: the quality and quantity of the PSC starting material. Most labs continually passage PSCs and use small quantities after expansion, but the "just-in-time" nature of these experiments means that quality control rarely happens before use. Lack of quality control could compromise PSC quality, sterility, and genetic integrity, which creates a variable that might affect results. This method, called CryoPause, banks PSCs as single-use, cryopreserved vials that can be thawed and immediately used in experiments. Each CryoPause bank provides a consistent source of PSCs that can be pre-validated before use to reduce the possibility that high levels of spontaneous differentiation, contamination, or genetic integrity will compromise an experiment.


Subject(s)
Cryopreservation/methods , Pluripotent Stem Cells/cytology , Animals , Biological Specimen Banks , Cell Differentiation , Cell Line , Cell- and Tissue-Based Therapy , Gene Editing , Humans , Mice , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/transplantation
5.
Mol Ther Methods Clin Dev ; 4: 72-82, 2017 Mar 17.
Article in English | MEDLINE | ID: mdl-28344993

ABSTRACT

Cell replacement therapy in the nervous system has a rich history, with ∼40 years of research and ∼30 years of clinical experience. There is compelling evidence that appropriate cells can integrate and function in the dysfunctioning human nervous system, but the clinical results are mixed in practice. A number of factors conspire to vary patient outcome: the indication, cell source, patient selection, and team performing transplantation are all variables that can affect efficacy. Most early clinical trials have used fetal cells, a limited cell source that resists scale and standardization. Direct fetal cell transplantation creates significant challenges to commercialization that is the ultimate goal of an effective cell therapy. One approach to help scale and standardize fetal cell preparations is the expansion of neural cells in vitro. Expansion is achieved by transformation or through the application of mitogens before cryopreservation. Recently, neural cells derived from pluripotent stem cells have provided a scalable alternative. Pluripotent stem cells are desirable for manufacturing but present alternative concerns and manufacturing obstacles. All cell sources require robust and reproducible manufacturing to make nervous system cell replacement therapy an option for patients. Here, we discuss the challenges and opportunities for cell replacement in the nervous system. In this review, we give an overview of completed and ongoing neural cell transplantation clinical trials, and we discuss the challenges and opportunities for future cell replacement trials with a particular focus on pluripotent stem cell-derived therapies.

6.
Stem Cell Reports ; 6(6): 858-872, 2016 06 14.
Article in English | MEDLINE | ID: mdl-27304916

ABSTRACT

Human pluripotent stem cells (hPSCs) provide an unlimited cell source for regenerative medicine. Hormone-producing cells are particularly suitable for cell therapy, and hypopituitarism, a defect in pituitary gland function, represents a promising therapeutic target. Previous studies have derived pituitary lineages from mouse and human ESCs using 3D organoid cultures that mimic the complex events underlying pituitary gland development in vivo. Instead of relying on unknown cellular signals, we present a simple and efficient strategy to derive human pituitary lineages from hPSCs using monolayer culture conditions suitable for cell manufacturing. We demonstrate that purified placode cells can be directed into pituitary fates using defined signals. hPSC-derived pituitary cells show basal and stimulus-induced hormone release in vitro and engraftment and hormone release in vivo after transplantation into a murine model of hypopituitarism. This work lays the foundation for future cell therapy applications in patients with hypopituitarism.


Subject(s)
Corticotrophs/metabolism , Embryonic Stem Cells/metabolism , Hypopituitarism/therapy , Pluripotent Stem Cells/metabolism , Thyrotrophs/metabolism , Adrenocorticotropic Hormone/biosynthesis , Adrenocorticotropic Hormone/metabolism , Animals , Benzamides/pharmacology , Biomarkers/metabolism , Bone Morphogenetic Protein 4/pharmacology , Cell Culture Techniques , Cell Differentiation/drug effects , Cell- and Tissue-Based Therapy , Corticotrophs/cytology , Corticotrophs/drug effects , Dioxoles/pharmacology , Disease Models, Animal , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Fibroblast Growth Factors/pharmacology , Follicle Stimulating Hormone/biosynthesis , Follicle Stimulating Hormone/metabolism , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/metabolism , Gene Expression , Growth Hormone/biosynthesis , Growth Hormone/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Hypopituitarism/genetics , Hypopituitarism/metabolism , Hypopituitarism/pathology , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Pituitary Gland/metabolism , Pituitary Gland/pathology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/drug effects , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/metabolism , Thyrotrophs/cytology , Thyrotrophs/drug effects , Transcription Factor AP-2/genetics , Transcription Factor AP-2/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
7.
Methods Mol Biol ; 1307: 329-43, 2016.
Article in English | MEDLINE | ID: mdl-24301074

ABSTRACT

The neural crest (NC) is a transient population of multipotent cells giving rise to the peripheral nervous system, skin pigmentation, heart, and facial mesenchyme. The broad cell fate potential of NC makes it an attractive cell fate to derive from human pluripotent stem cells (hPSCs) for exploring embryonic development, modeling disease, and generating cells for transplantation. Here, we discuss recent publications and methods for efficiently differentiating hPSCs into NC. We also provide methods to direct NC into two different terminal fates: melanocytes and sensory neurons.


Subject(s)
Cell Culture Techniques/methods , Neural Crest/cytology , Pluripotent Stem Cells/cytology , Smad Proteins/antagonists & inhibitors , Wnt Proteins/metabolism , Animals , Cell Count , Cell Differentiation , Cell Lineage , Cells, Cultured , Humans , Melanocytes/cytology , Mice , Pluripotent Stem Cells/metabolism , Sensory Receptor Cells/cytology , Smad Proteins/metabolism
8.
Curr Protoc Stem Cell Biol ; 33: 1H.9.1-1H.9.9, 2015 May 01.
Article in English | MEDLINE | ID: mdl-26344232

ABSTRACT

Neural crest (NC) cells are migratory multipotent progenitors that delaminate from the neural tube during embryonic development and give rise to various cell types in different organs. These cells are a transient embryonic cell population and therefore difficult to obtain from primary sources. Deriving NC from human pluripotent stem cells offers an alternative way to provide large-scale human NC cells for developmental and disease-related studies. In recent years, the protocols to make these cells have matured, incorporating the efficient conversion of pluripotent stem cells to neural cells through dual SMAD inhibition and early Wnt activation to increase the yield of NC cells. Here, we provide a minor variation to this NC protocol that has been successful for many in our laboratories.


Subject(s)
Neural Crest , Pluripotent Stem Cells , Smad Proteins/antagonists & inhibitors , Smad Proteins/metabolism , Animals , Humans , Neural Crest/cytology , Neural Crest/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism
9.
Stem Cells ; 33(2): 367-77, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25336442

ABSTRACT

The predominant view of pluripotency regulation proposes a stable ground state with coordinated expression of key transcription factors (TFs) that prohibit differentiation. Another perspective suggests a more complexly regulated state involving competition between multiple lineage-specifying TFs that define pluripotency. These contrasting views were developed from extensive analyses of TFs in pluripotent cells in vitro. An experimentally validated, genome-wide repertoire of the regulatory interactions that control pluripotency within the in vivo cellular contexts is yet to be developed. To address this limitation, we assembled a TF interactome of adult human male germ cell tumors (GCTs) using the Algorithm for the Accurate Reconstruction of Cellular Pathways (ARACNe) to analyze gene expression profiles of 141 tumors comprising pluripotent and differentiated subsets. The network (GCT(Net)) comprised 1,305 TFs, and its ingenuity pathway analysis identified pluripotency and embryonal development as the top functional pathways. We experimentally validated GCT(Net) by functional (silencing) and biochemical (ChIP-seq) analysis of the core pluripotency regulatory TFs POU5F1, NANOG, and SOX2 in relation to their targets predicted by ARACNe. To define the extent of the in vivo pluripotency network in this system, we ranked all TFs in the GCT(Net) according to sharing of ARACNe-predicted targets with those of POU5F1 and NANOG using an odds-ratio analysis method. To validate this network, we silenced the top 10 TFs in the network in H9 embryonic stem cells. Silencing of each led to downregulation of pluripotency and induction of lineage; 7 of the 10 TFs were identified as pluripotency regulators for the first time.


Subject(s)
Algorithms , Models, Biological , Neoplasm Proteins/metabolism , Neoplasms, Germ Cell and Embryonal/metabolism , Pluripotent Stem Cells/metabolism , Transcription Factors/metabolism , Adult , Cell Line, Tumor , Humans , Male , Neoplasm Proteins/genetics , Neoplasms, Germ Cell and Embryonal/genetics , Neoplasms, Germ Cell and Embryonal/pathology , Pluripotent Stem Cells/pathology , Transcription Factors/genetics
10.
Stem Cells Dev ; 23(10): 1149-56, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24383669

ABSTRACT

MicroRNAs (miRNAs) regulate myriad biological processes; however, their role in cell fate choice is relatively unexplored. Pluripotent NT2/D1 embryonal carcinoma cells differentiate into an epithelial/smooth muscle phenotype when treated with bone morphogenetic protein-2 (BMP-2). To identify miRNAs involved in epithelial cell development, we performed miRNA profiling of NT2/D1 cells treated with BMP-2 at 6, 12, and 24 h, and on days 6 and 10. Integration of the miRNA profiling data with previously obtained gene expression profiling (GEP) data of NT2/D1 cells treated with BMP-2 at the same time points identified miR-18b and miR-518b as the top two miRNAs with the highest number of up-regulated predicted targets with known functions in epithelial lineage development. Silencing of miR-18b and miR-518b in NT2/D1 cells revealed several up-regulated TFs with functions in epithelial lineage development; among these, target prediction programs identified FOXN1 as the only direct target of both miRNAs. FOXN1 has previously been shown to play an important role in keratinocyte differentiation and epithelial cell proliferation. NT2/D1 and H9 human embryonic stem cells with silenced miR-18b and miR-518b showed up-regulation of FOXN1 and the epithelial markers CDH1, EPCAM, KRT19, and KRT7. A 3'UTR luciferase assay confirmed FOXN1 to be a target of the two miRNAs, and up-regulation of FOXN1 in NT2/D1 cells led to the expression of epithelial markers. Overexpression of the two miRNAs in BMP-2-treated NT2/D1 cells led to down-regulation of FOXN1 and epithelial lineage markers. These results show that miR-18b and miR-518b are upstream controllers of FOXN1-directed epithelial lineage development.


Subject(s)
Cell Differentiation/physiology , Epithelial Cells/metabolism , Forkhead Transcription Factors/biosynthesis , MicroRNAs/metabolism , Pluripotent Stem Cells/metabolism , Cell Line , Cell Proliferation/physiology , Down-Regulation/physiology , Epithelial Cells/cytology , Forkhead Transcription Factors/genetics , Humans , MicroRNAs/genetics , Pluripotent Stem Cells/cytology , Up-Regulation/physiology
11.
Mol Cell ; 53(1): 19-31, 2014 Jan 09.
Article in English | MEDLINE | ID: mdl-24289922

ABSTRACT

Fragile X syndrome (FXS) is caused by a CGG repeat expansion in the FMR1 gene that appears to occur during oogenesis and during early embryogenesis. One model proposes that repeat instability depends on the replication fork direction through the repeats such that (CNG)n hairpin-like structures form, causing DNA polymerase to stall and slip. Examining DNA replication fork progression on single DNA molecules at the endogenous FMR1 locus revealed that replication forks stall at CGG repeats in human cells. Furthermore, replication profiles of FXS human embryonic stem cells (hESCs) compared to nonaffected hESCs showed that fork direction through the repeats is altered at the FMR1 locus in FXS hESCs, such that predominantly the CCG strand serves as the lagging-strand template. This is due to the absence of replication initiation that would typically occur upstream of FMR1, suggesting that altered replication origin usage combined with fork stalling promotes repeat instability during early embryonic development.


Subject(s)
DNA Replication , Embryonic Stem Cells/metabolism , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/embryology , Genetic Loci , Trinucleotide Repeats , Embryonic Development/genetics , Embryonic Stem Cells/pathology , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Fragile X Syndrome/pathology , Humans
12.
Cell Stem Cell ; 13(6): 691-705, 2013 Dec 05.
Article in English | MEDLINE | ID: mdl-24315443

ABSTRACT

Reprogramming somatic cells to induced pluripotent stem cells (iPSCs) resets their identity back to an embryonic age and, thus, presents a significant hurdle for modeling late-onset disorders. In this study, we describe a strategy for inducing aging-related features in human iPSC-derived lineages and apply it to the modeling of Parkinson's disease (PD). Our approach involves expression of progerin, a truncated form of lamin A associated with premature aging. We found that expression of progerin in iPSC-derived fibroblasts and neurons induces multiple aging-related markers and characteristics, including dopamine-specific phenotypes such as neuromelanin accumulation. Induced aging in PD iPSC-derived dopamine neurons revealed disease phenotypes that require both aging and genetic susceptibility, such as pronounced dendrite degeneration, progressive loss of tyrosine hydroxylase (TH) expression, and enlarged mitochondria or Lewy-body-precursor inclusions. Thus, our study suggests that progerin-induced aging can be used to reveal late-onset age-related disease features in hiPSC-based disease models.


Subject(s)
Aging/pathology , Induced Pluripotent Stem Cells/metabolism , Models, Biological , Nuclear Proteins/metabolism , Protein Precursors/metabolism , Adult , Age of Onset , Aged , Aged, 80 and over , Animals , Biomarkers/metabolism , Cell Differentiation , Cellular Reprogramming , Cellular Senescence , Child , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Dopaminergic Neurons/transplantation , Dopaminergic Neurons/ultrastructure , Fibroblasts/metabolism , Humans , Lamin Type A , Mesencephalon/pathology , Mice , Middle Aged , Parkinson Disease/pathology , Phenotype , Tissue Donors
13.
Cell Rep ; 3(4): 1140-52, 2013 Apr 25.
Article in English | MEDLINE | ID: mdl-23583175

ABSTRACT

Melanocytes are pigment-producing cells of neural crest (NC) origin that are responsible for protecting the skin against UV irradiation. Pluripotent stem cell (PSC) technology offers a promising approach for studying human melanocyte development and disease. Here, we report that timed exposure to activators of WNT, BMP, and EDN3 signaling triggers the sequential induction of NC and melanocyte precursor fates under dual-SMAD-inhibition conditions. Using a SOX10::GFP human embryonic stem cell (hESC) reporter line, we demonstrate that the temporal onset of WNT activation is particularly critical for human NC induction. Subsequent maturation of hESC-derived melanocytes yields pure populations that match the molecular and functional properties of adult melanocytes. Melanocytes from Hermansky-Pudlak syndrome and Chediak-Higashi syndrome patient-specific induced PSCs (iPSCs) faithfully reproduce the ultrastructural features of disease-associated pigmentation defects. Our data define a highly specific requirement for WNT signaling during NC induction and enable the generation of pure populations of human iPSC-derived melanocytes for faithful modeling of pigmentation disorders.


Subject(s)
Embryonic Stem Cells/cytology , Induced Pluripotent Stem Cells/cytology , Melanocytes/cytology , Models, Biological , Neural Crest/cytology , Bone Morphogenetic Proteins/metabolism , Cell Differentiation , Cell Lineage , Chediak-Higashi Syndrome/metabolism , Chediak-Higashi Syndrome/pathology , Embryonic Stem Cells/metabolism , Endothelin-3/metabolism , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hermanski-Pudlak Syndrome/metabolism , Hermanski-Pudlak Syndrome/pathology , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/ultrastructure , Melanocytes/metabolism , Neural Crest/metabolism , Pigmentation , SOXE Transcription Factors/genetics , SOXE Transcription Factors/metabolism , Signal Transduction , Wnt Proteins/metabolism
14.
PLoS One ; 7(8): e42302, 2012.
Article in English | MEDLINE | ID: mdl-22879936

ABSTRACT

Embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) offer great promise in regenerative medicine and disease modeling due to their unlimited self-renewal and broad differentiation capacity. There is evidence that the growth properties and critical signaling pathways differ between murine and human ESCs; therefore, it is essential to perform functional studies to test the putatively conserved mechanisms of pluripotent stem cell self-renewal between species. Previously, we identified the transcription factor Zfx as a key regulator of self-renewal in murine ESCs. Here we extend those findings to human ESCs. ZFX knockdown in hESCs hindered clonal growth and decreased colony size after serial replating. ZFX overexpression enhanced clone formation in the presence of Y-27632, increased colony size at low density and decreased expression of differentiation-related genes in human ESCs. ZFX-overexpressing hESCs resisted spontaneous differentiation but could be directed to differentiate into endodermal and neural cell fates when provided with the appropriate cues. Thus, ZFX acts as a molecular rheostat regulating the balance between self-renewal and differentiation in hESCs, revealing the close evolutionary conservation of the self-renewal mechanisms in murine and human ESCs.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Kruppel-Like Transcription Factors/metabolism , Animals , Cell Differentiation/genetics , Cell Line , Cell Proliferation , Cell Size , Chromosomes, Artificial, Bacterial/genetics , Clone Cells , Endoderm/cytology , Endoderm/metabolism , Gene Expression Profiling , Gene Expression Regulation , Gene Knockdown Techniques , Humans , Kruppel-Like Transcription Factors/genetics , Mice , Transgenes/genetics
15.
J Clin Invest ; 122(8): 2928-39, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22751106

ABSTRACT

Embryonic stem cells (ESCs) represent a promising source of midbrain dopaminergic (DA) neurons for applications in Parkinson disease. However, ESC-based transplantation paradigms carry a risk of introducing inappropriate or tumorigenic cells. Cell purification before transplantation may alleviate these concerns and enable identification of the specific DA neuron stage most suitable for cell therapy. Here, we used 3 transgenic mouse ESC reporter lines to mark DA neurons at 3 stages of differentiation (early, middle, and late) following induction of differentiation using Hes5::GFP, Nurr1::GFP, and Pitx3::YFP transgenes, respectively. Transplantation of FACS-purified cells from each line resulted in DA neuron engraftment, with the mid-stage and late-stage neuron grafts being composed almost exclusively of midbrain DA neurons. Mid-stage neuron cell grafts had the greatest amount of DA neuron survival and robustly induced recovery of motor deficits in hemiparkinsonian mice. Our data suggest that the Nurr1+ stage (middle stage) of neuronal differentiation is particularly suitable for grafting ESC-derived DA neurons. Moreover, global transcriptome analysis of progeny from each of the ESC reporter lines revealed expression of known midbrain DA neuron genes and also uncovered previously uncharacterized midbrain genes. These data demonstrate remarkable fate specificity of ESC-derived DA neurons and outline a sequential stage-specific ESC reporter line paradigm for in vivo gene discovery.


Subject(s)
Dopaminergic Neurons/transplantation , Embryonic Stem Cells/transplantation , Neural Stem Cells/transplantation , Animals , Cell Differentiation , Cell Line , Cell Separation/methods , Cell Survival , Dopaminergic Neurons/cytology , Dopaminergic Neurons/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Genes, Reporter , Graft Survival , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mesencephalon/cytology , Mesencephalon/metabolism , Mice , Mice, Transgenic , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transcriptome
16.
Stem Cells Dev ; 21(12): 2312-21, 2012 Aug 10.
Article in English | MEDLINE | ID: mdl-22224974

ABSTRACT

Endothelial cells line blood vessels and coordinate many aspects of vascular biology. More recent work has shown that endothelial cells provide a key niche in vivo for neural stem cells. In vitro, endothelial cells secrete a factor that expands neural stem cells while inhibiting their differentiation. Here, we show that a transformed mouse endothelial cell line (bEnd.3) maintains human pluripotent stem cells in an undifferentiated state. bEnd.3 cells have a practical advantage over mouse embryonic fibroblasts for pluripotent stem cell maintenance since they can be expanded in vitro and engineered to express genes of interest. We demonstrate this capability by producing fluorescent and drug-resistant feeder cells. Further, we show that bEnd.3 secretes an activity that maintains human embryonic stem cells without direct contact.


Subject(s)
Endothelial Cells/physiology , Feeder Cells/physiology , Induced Pluripotent Stem Cells/physiology , Animals , Cell Line , Cell Proliferation , Cell Shape , Cells, Cultured , Coculture Techniques , Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Endothelial Cells/cytology , Endothelial Cells/metabolism , Feeder Cells/cytology , Genetic Engineering , Humans , Induced Pluripotent Stem Cells/cytology , Mice , Neurogenesis
17.
Methods Mol Biol ; 793: 87-97, 2011.
Article in English | MEDLINE | ID: mdl-21913095

ABSTRACT

Human embryonic stem cells (hESCs) and the related induced pluripotent stem cells (hiPSCs) have attracted considerable attention since they can provide an unlimited source of many different tissue types. One challenge of using pluripotent cells is directing their broad differentiation potential into one specific tissue or cell fate. The cell fate choices of extraembryonic, endoderm, mesoderm, and ectoderm (including neural) lineages represent the earliest decisions. We found that pluripotent cells efficiently neuralize by blocking the signaling pathways required for alternative cell fate decisions. In this chapter, we detail methods to direct hESCs or hiPSCs into early neural cells and subsequently postmitotic neurons.


Subject(s)
Cell Differentiation , Cytological Techniques/methods , Embryonic Stem Cells/cytology , Induced Pluripotent Stem Cells/cytology , Models, Biological , Neurodegenerative Diseases/pathology , Neurons/cytology , Animals , Cell Line , Humans , Mice
18.
Nat Protoc ; 5(4): 688-701, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20360764

ABSTRACT

Human pluripotent stem cell (hPSC)-derived neural crest (NC) cells present a valuable tool for modeling aspects of human NC development, including cell fate specification, multipotency and cell migration. hPSC-derived NC cells are also suitable for modeling human disease and as a renewable cell source for applications in regenerative medicine. Here we provide protocols for the step-wise differentiation of human embryonic stem cells (hESCs) or human induced pluripotent stem cells (hiPSCs) into neuroectodermal and NC cells using either the MS5 coculture system or a novel defined culture method based on pharmacological inhibition of bone morphogenetic protein and transforming growth factor-beta signaling pathways. Furthermore, we present protocols for the purification and propagation of hPSC-NC cells using flow cytometry and defined in vitro culture conditions. Our protocol has been validated in multiple independent hESC and hiPSC lines. The average time required for generating purified hPSC-NC precursors using this protocol is 2-5 weeks.


Subject(s)
Cell Culture Techniques/methods , Neural Crest/cytology , Pluripotent Stem Cells/cytology , Cell Differentiation , Cell Proliferation , Cell Separation , Chromosomes, Artificial, Bacterial/genetics , Coculture Techniques , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Flow Cytometry , Humans , Models, Neurological , Neural Crest/metabolism , Pluripotent Stem Cells/metabolism , Schwann Cells/cytology
19.
Cell Stem Cell ; 6(4): 336-347, 2010 Apr 02.
Article in English | MEDLINE | ID: mdl-20362538

ABSTRACT

The floor plate (FP) is a critical signaling center during neural development located along the ventral midline of the embryo. Little is known about human FP development because of the lack of tissue accessibility. Here we report the efficient derivation of human embryonic stem cell (hESC)-derived FP tissue capable of secreting Netrin-1 and SHH and patterning primary and hESC derived tissues. FP induction in hESCs is dependent on early SHH exposure and occurs at the expense of anterior neurectoderm (AN). Global gene expression and functional studies identify SHH-mediated inhibition of Dkk-1 as key factor in FP versus AN specification. hESC-derived FP tissue is shown to be of anterior SIX6+ character but is responsive to caudalizing factors suppressing SIX6 expression and inducing a shift in usage of region-specific SHH enhancers. These data define the early signals that drive human FP versus AN specification and determine regional identity in hESC-derived FP.


Subject(s)
Embryonic Stem Cells/cytology , Nervous System/cytology , Animals , Body Patterning/genetics , Cell Differentiation/genetics , Cell Lineage/genetics , Embryonic Stem Cells/metabolism , Forkhead Transcription Factors/metabolism , Gene Expression Profiling , Gene Expression Regulation, Developmental , Hedgehog Proteins/metabolism , Hepatocyte Nuclear Factor 3-beta/metabolism , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Nerve Tissue Proteins/metabolism , Nervous System/metabolism , Neural Plate/embryology , Neural Plate/metabolism , Time Factors , Transcription, Genetic
20.
Nature ; 461(7262): 402-6, 2009 Sep 17.
Article in English | MEDLINE | ID: mdl-19693009

ABSTRACT

The isolation of human induced pluripotent stem cells (iPSCs) offers a new strategy for modelling human disease. Recent studies have reported the derivation and differentiation of disease-specific human iPSCs. However, a key challenge in the field is the demonstration of disease-related phenotypes and the ability to model pathogenesis and treatment of disease in iPSCs. Familial dysautonomia (FD) is a rare but fatal peripheral neuropathy, caused by a point mutation in the IKBKAP gene involved in transcriptional elongation. The disease is characterized by the depletion of autonomic and sensory neurons. The specificity to the peripheral nervous system and the mechanism of neuron loss in FD are poorly understood owing to the lack of an appropriate model system. Here we report the derivation of patient-specific FD-iPSCs and the directed differentiation into cells of all three germ layers including peripheral neurons. Gene expression analysis in purified FD-iPSC-derived lineages demonstrates tissue-specific mis-splicing of IKBKAP in vitro. Patient-specific neural crest precursors express particularly low levels of normal IKBKAP transcript, suggesting a mechanism for disease specificity. FD pathogenesis is further characterized by transcriptome analysis and cell-based assays revealing marked defects in neurogenic differentiation and migration behaviour. Furthermore, we use FD-iPSCs for validating the potency of candidate drugs in reversing aberrant splicing and ameliorating neuronal differentiation and migration. Our study illustrates the promise of iPSC technology for gaining new insights into human disease pathogenesis and treatment.


Subject(s)
Dysautonomia, Familial/pathology , Dysautonomia, Familial/therapy , Models, Biological , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/transplantation , Adolescent , Alternative Splicing/drug effects , Alternative Splicing/genetics , Animals , Carrier Proteins/genetics , Cell Dedifferentiation , Cell Differentiation , Cell Lineage , Cell Movement , Cells, Cultured , Child , Dysautonomia, Familial/drug therapy , Dysautonomia, Familial/genetics , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Profiling , Humans , Kinetin/pharmacology , Kinetin/therapeutic use , Male , Mice , Neural Crest/cytology , Neural Crest/drug effects , Organ Specificity , Phenotype , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/drug effects , Transcriptional Elongation Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...