Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
bioRxiv ; 2024 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-38496581

RESUMEN

One of the most important properties of human embryonic stem cells (hESCs) is related to their pluripotent states. In our recent study, we identified a previously unrecognized pluripotent state induced by RSeT medium. This state makes primed hESCs resistant to conversion to naïve pluripotent state. In this study, we have further characterized the metabolic features in these RSeT hESCs, including metabolic gene expression, metabolomic analysis, and various functional assays. The commonly reported metabolic modes include glycolysis or both glycolysis and oxidative phosphorylation (i.e., metabolic bivalency) in pluripotent stem cells. However, besides the presence of metabolic bivalency, RSeT hESCs exhibited a unique metabolome with additional fatty acid oxidation and imbalanced nucleotide metabolism. This metabolic quadrivalency is linked to hESC growth independent of oxygen tension and restricted capacity for naïve reprogramming in these cells. Thus, this study provides new insights into pluripotent state transitions and metabolic stress-associated hPSC growth in vitro.

2.
bioRxiv ; 2024 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-38410444

RESUMEN

One of the most important properties of human embryonic stem cells (hESCs) is related to their primed and naïve pluripotent states. Our previous meta-analysis indicates the existence of heterogeneous pluripotent states derived from diverse naïve protocols. In this study, we have characterized a commercial medium (RSeT)-based pluripotent state under various growth conditions. Notably, RSeT hESCs can circumvent hypoxic growth conditions as required by naïve hESCs, in which some RSeT cells (e.g., H1 cells) exhibit much lower single cell plating efficiency, having altered or much retarded cell growth under both normoxia and hypoxia. Evidently, hPSCs lack many transcriptomic hallmarks of naïve and formative pluripotency (a phase between naive and primed states). Integrative transcriptome analysis suggests our primed and RSeT hESCs are close to the early stage of post-implantation embryos, similar to the previously reported primary hESCs and early hESC cultures. Moreover, RSeT hESCs did not express naïve surface markers such as CD75, SUSD2, and CD130 at a significant level. Biochemically, RSeT hESCs exhibit a differential dependency of FGF2 and co-independency of both Janus kinase (JAK) and TGFß signaling in a cell-line-specific manner. Thus, RSeT hESCs represent a previously unrecognized pluripotent state downstream of formative pluripotency. Our data suggest that human naïve pluripotent potentials may be restricted in RSeT medium. Hence, this study provides new insights into pluripotent state transitions in vitro.

3.
Stem Cell Reports ; 17(2): 397-412, 2022 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-35063131

RESUMEN

Inhibition of PIKfyve phosphoinositide kinase selectively kills autophagy-dependent cancer cells by disrupting lysosome homeostasis. Here, we show that PIKfyve inhibitors can also selectively eliminate pluripotent embryonal carcinoma cells (ECCs), embryonic stem cells, and induced pluripotent stem cells under conditions where differentiated cells remain viable. PIKfyve inhibitors prevented lysosome fission, induced autophagosome accumulation, and reduced cell proliferation in both pluripotent and differentiated cells, but they induced death only in pluripotent cells. The ability of PIKfyve inhibitors to distinguish between pluripotent and differentiated cells was confirmed with xenografts derived from ECCs. Pretreatment of ECCs with the PIKfyve specific inhibitor WX8 suppressed their ability to form teratocarcinomas in mice, and intraperitoneal injections of WX8 into mice harboring teratocarcinoma xenografts selectively eliminated pluripotent cells. Differentiated cells continued to proliferate, but at a reduced rate. These results provide a proof of principle that PIKfyve specific inhibitors can selectively eliminate pluripotent stem cells in vivo as well as in vitro.


Asunto(s)
Apoptosis/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Fosfatidilinositol 3-Quinasas/química , Animales , Autofagia , Línea Celular , Supervivencia Celular/efectos de los fármacos , ADN/metabolismo , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Inhibidores Enzimáticos/uso terapéutico , Femenino , Fase G1 , Humanos , Hidrazinas/química , Hidrazinas/farmacología , Hidrazinas/uso terapéutico , Ratones , Ratones Desnudos , Fosfatidilinositol 3-Quinasas/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/efectos de los fármacos , Células Madre Pluripotentes/metabolismo , Teratocarcinoma/tratamiento farmacológico , Teratocarcinoma/patología , Trasplante Heterólogo
4.
PLoS One ; 16(5): e0251461, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33984026

RESUMEN

The ground or naive pluripotent state of human pluripotent stem cells (hPSCs), which was initially established in mouse embryonic stem cells (mESCs), is an emerging and tentative concept. To verify this vital concept in hPSCs, we performed a multivariate meta-analysis of major hPSC datasets via the combined analytic powers of percentile normalization, principal component analysis (PCA), t-distributed stochastic neighbor embedding (t-SNE), and SC3 consensus clustering. This robust bioinformatics approach has significantly improved the predictive values of our meta-analysis. Accordingly, we revealed various similarities or dissimilarities between some naive-like hPSCs (NLPs) generated from different laboratories. Our analysis confirms some previous studies and provides new evidence concerning the existence of three distinct naive-like pluripotent states. Moreover, our study offers global transcriptomic markers that define diverse pluripotent states under various hPSC growth protocols.


Asunto(s)
Genómica/métodos , Células Madre Pluripotentes/metabolismo , Transcriptoma , Diferenciación Celular , Proliferación Celular , Humanos , Células Madre Pluripotentes/citología
5.
Stem Cell Res ; 46: 101823, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32505898

RESUMEN

Muenke syndrome is the leading genetic cause of craniosynostosis and results in a variety of disabling clinical phenotypes. To model the disease and study the pathogenic mechanisms, a human induced pluripotent stem cell (hiPSC) line was generated from a patient diagnosed with Muenke syndrome. Successful reprogramming was validated by morphological features, karyotyping, loss of reprogramming factors, expression of pluripotency markers, mutation analysis and teratoma formation.


Asunto(s)
Craneosinostosis , Células Madre Pluripotentes Inducidas , Craneosinostosis/genética , Humanos , Mutación , Fenotipo , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/genética
6.
Dis Model Mech ; 13(6)2020 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-32423971

RESUMEN

The basal lamina is a specialized sheet of dense extracellular matrix (ECM) linked to the plasma membrane of specific cell types in their tissue context, which serves as a structural scaffold for organ genesis and maintenance. Disruption of the basal lamina and its functions is central to many disease processes, including cancer metastasis, kidney disease, eye disease, muscular dystrophies and specific types of brain malformation. The latter three pathologies occur in the α-dystroglycanopathies, which are caused by dysfunction of the ECM receptor α-dystroglycan. However, opportunities to study the basal lamina in various human disease tissues are restricted owing to its limited accessibility. Here, we report the generation of embryoid bodies from human induced pluripotent stem cells that model the basal lamina. Embryoid bodies cultured via this protocol mimic pre-gastrulation embryonic development, consisting of an epithelial core surrounded by a basal lamina and a peripheral layer of ECM-secreting endoderm. In α-dystroglycanopathy patient embryoid bodies, electron and fluorescence microscopy reveal ultrastructural basal lamina defects and reduced ECM accumulation. By starting from patient-derived cells, these results establish a method for the in vitro synthesis of patient-specific basal lamina and recapitulate disease-relevant ECM defects seen in the α-dystroglycanopathies. Finally, we apply this system to evaluate an experimental ribitol supplement therapy on genetically diverse α-dystroglycanopathy patient samples.This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Membrana Basal/metabolismo , Cuerpos Embrioides/metabolismo , Matriz Extracelular/metabolismo , Células Madre Embrionarias Humanas/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Síndrome de Walker-Warburg/metabolismo , Membrana Basal/efectos de los fármacos , Membrana Basal/ultraestructura , Estudios de Casos y Controles , Técnicas de Cultivo de Célula , Células Cultivadas , Niño , Preescolar , Distroglicanos/genética , Distroglicanos/metabolismo , Cuerpos Embrioides/efectos de los fármacos , Cuerpos Embrioides/ultraestructura , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/ultraestructura , Femenino , Regulación del Desarrollo de la Expresión Génica , Células Madre Embrionarias Humanas/efectos de los fármacos , Células Madre Embrionarias Humanas/ultraestructura , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/ultraestructura , Recién Nacido , Masculino , Persona de Mediana Edad , Ribitol/farmacología , Síndrome de Walker-Warburg/tratamiento farmacológico , Síndrome de Walker-Warburg/genética , Síndrome de Walker-Warburg/patología
7.
Mol Psychiatry ; 24(4): 613-624, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30135510

RESUMEN

Biological characterization of genetic variants identified in genome-wide association studies (GWAS) remains a substantial challenge. Here we used human-induced pluripotent stem cells (iPSC) and their neural derivatives to characterize common variants on chromosome 3p22 that have been associated by GWAS with major mental illnesses. IPSC-derived neural progenitor cells carrying the risk allele of the single nucleotide polymorphism (SNP), rs9834970, displayed lower baseline TRANK1 expression that was rescued by chronic treatment with therapeutic dosages of valproic acid (VPA). VPA had the greatest effects on TRANK1 expression in iPSC, NPC, and astrocytes. Although rs9834970 has no known function, we demonstrated that a nearby SNP, rs906482, strongly affects binding by the transcription factor, CTCF, and that the high-affinity allele usually occurs on haplotypes carrying the rs9834970 risk allele. Decreased expression of TRANK1 perturbed expression of many genes involved in neural development and differentiation. These findings have important implications for the pathophysiology of major mental illnesses and the development of novel therapeutics.


Asunto(s)
Citocinas/genética , Células-Madre Neurales/efectos de los fármacos , Ácido Valproico/farmacología , Alelos , Astrocitos/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Citocinas/efectos de los fármacos , Citocinas/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Frecuencia de los Genes/genética , Estudio de Asociación del Genoma Completo , Genotipo , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Neurogénesis/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Polimorfismo de Nucleótido Simple/genética , Ácido Valproico/metabolismo
8.
Trends Mol Med ; 24(9): 805-820, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30006147

RESUMEN

Use of human pluripotent stem cells (hPSCs) and their differentiated derivatives have led to recent proof-of-principle drug discoveries, defining a pathway to the implementation of hPSC-based drug discovery (hPDD). Current hPDD strategies, however, have inevitable conceptual biases and technological limitations, including the dimensionality of cell-culture methods, cell maturity and functionality, experimental variability, and data reproducibility. In this review, we dissect representative hPDD systems via analysis of hPSC-based 2D-monolayers, 3D culture, and organoids. We discuss mechanisms of drug discovery and drug repurposing, and roles of membrane drug transporters in tissue maturation and hPDD using the example of drugs that target various mutations of CFTR, the cystic fibrosis transmembrane conductance regulator gene, in patients with cystic fibrosis.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Desarrollo de Medicamentos/métodos , Descubrimiento de Drogas/métodos , Células Madre Pluripotentes/efectos de los fármacos , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Técnicas de Cultivo de Célula/instrumentación , Fibrosis Quística/tratamiento farmacológico , Fibrosis Quística/genética , Fibrosis Quística/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Desarrollo de Medicamentos/instrumentación , Descubrimiento de Drogas/instrumentación , Humanos , Terapia Molecular Dirigida/métodos , Organoides/citología , Organoides/efectos de los fármacos , Organoides/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo
9.
Stem Cells ; 36(10): 1501-1513, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29873142

RESUMEN

Human induced pluripotent stem cells (iPSCs) have great potential as source cells for therapeutic uses. However, reports indicate that iPSCs carry genetic abnormalities, which may impede their medical use. Little is known about mechanisms contributing to intrinsic DNA damage in iPSCs that could lead to genomic instability. In this report, we investigated the level of DNA damage in human iPSC lines compared with their founder fibroblast line and derived mesenchymal stromal cell (MSC) lines using the phosphorylated histone variant, γH2AX, as a marker of DNA damage. We show that human iPSCs have elevated basal levels of γH2AX, which correlate with markers of DNA replication: 5-ethynyl-2'-deoxyuridine and the single-stranded binding protein, replication protein A. γH2AX foci in iPSCs also colocalize to BRCA1 and RAD51, proteins in the homologous repair pathway, implying γH2AX in iPSCs marks sites of double strand breaks. Our study demonstrates an association between increased basal levels of γH2AX and the rapid replication of iPSCs. Stem Cells 2018;36:1501-1513.


Asunto(s)
Daño del ADN , Reparación del ADN , Histonas/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Animales , Diferenciación Celular/fisiología , Línea Celular , Replicación del ADN , Fibroblastos/metabolismo , Histonas/genética , Humanos , Ratones
10.
Cell ; 173(4): 851-863.e16, 2018 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-29576452

RESUMEN

Hibernating mammals survive hypothermia (<10°C) without injury, a remarkable feat of cellular preservation that bears significance for potential medical applications. However, mechanisms imparting cold resistance, such as cytoskeleton stability, remain elusive. Using the first iPSC line from a hibernating mammal (13-lined ground squirrel), we uncovered cellular pathways critical for cold tolerance. Comparison between human and ground squirrel iPSC-derived neurons revealed differential mitochondrial and protein quality control responses to cold. In human iPSC-neurons, cold triggered mitochondrial stress, resulting in reactive oxygen species overproduction and lysosomal membrane permeabilization, contributing to microtubule destruction. Manipulations of these pathways endowed microtubule cold stability upon human iPSC-neurons and rat (a non-hibernator) retina, preserving its light responsiveness after prolonged cold exposure. Furthermore, these treatments significantly improved microtubule integrity in cold-stored kidneys, demonstrating the potential for prolonging shelf-life of organ transplants. Thus, ground squirrel iPSCs offer a unique platform for bringing cold-adaptive strategies from hibernators to humans in clinical applications. VIDEO ABSTRACT.


Asunto(s)
Adaptación Fisiológica , Células Madre Pluripotentes Inducidas/metabolismo , Neuronas/metabolismo , Animales , Diferenciación Celular , Frío , Humanos , Células Madre Pluripotentes Inducidas/citología , Riñón/efectos de los fármacos , Riñón/metabolismo , Lisosomas/metabolismo , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Neuronas/citología , Estrés Oxidativo , Inhibidores de Proteasas/farmacología , Ratas , Especies Reactivas de Oxígeno/metabolismo , Retina/metabolismo , Sciuridae , Transcriptoma , Tubulina (Proteína)/química , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo
11.
Neuropsychopharmacology ; 42(3): 774-784, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27534267

RESUMEN

Because of unavoidable confounding variables in the direct study of human subjects, it has been difficult to unravel the effects of prenatal cocaine exposure on the human fetal brain, as well as the cellular and biochemical mechanisms involved. Here, we propose a novel approach using a human pluripotent stem cell (hPSC)-based 3D neocortical organoid model. This model retains essential features of human neocortical development by encompassing a single self-organized neocortical structure, without including an animal-derived gelatinous matrix. We reported previously that prenatal cocaine exposure to rats during the most active period of neural progenitor proliferation induces cytoarchitectural changes in the embryonic neocortex. We also identified a role of CYP450 and consequent oxidative ER stress signaling in these effects. However, because of differences between humans and rodents in neocorticogenesis and brain CYP metabolism, translation of the research findings from the rodent model to human brain development is uncertain. Using hPSC 3D neocortical organoids, we demonstrate that the effects of cocaine are mediated through CYP3A5-induced generation of reactive oxygen species, inhibition of neocortical progenitor cell proliferation, induction of premature neuronal differentiation, and interruption of neural tissue development. Furthermore, knockdown of CYP3A5 reversed these cocaine-induced pathological phenotypes, suggesting CYP3A5 as a therapeutic target to mitigate the deleterious neurodevelopmental effects of prenatal cocaine exposure in humans. Moreover, 3D organoid methodology provides an innovative platform for identifying adverse effects of abused psychostimulants and pharmaceutical agents, and can be adapted for use in neurodevelopmental disorders with genetic etiologies.


Asunto(s)
Cocaína/farmacología , Citocromo P-450 CYP3A/metabolismo , Inhibidores de Captación de Dopamina/farmacología , Neocórtex/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Células Madre Pluripotentes/efectos de los fármacos , Línea Celular , Humanos
12.
Stem Cell Res ; 17(1): 122-9, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27286574

RESUMEN

Identification and quantification of the characteristics of stem cell preparations is critical for understanding stem cell biology and for the development and manufacturing of stem cell based therapies. We have developed image analysis and visualization software that allows effective use of time-lapse microscopy to provide spatial and dynamic information from large numbers of human embryonic stem cell colonies. To achieve statistically relevant sampling, we examined >680 colonies from 3 different preparations of cells over 5days each, generating a total experimental dataset of 0.9 terabyte (TB). The 0.5 Giga-pixel images at each time point were represented by multi-resolution pyramids and visualized using the Deep Zoom Javascript library extended to support viewing Giga-pixel images over time and extracting data on individual colonies. We present a methodology that enables quantification of variations in nominally-identical preparations and between colonies, correlation of colony characteristics with Oct4 expression, and identification of rare events.


Asunto(s)
Células Madre Embrionarias Humanas/citología , Procesamiento de Imagen Asistido por Computador , Microscopía Fluorescente , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Imagen de Lapso de Tiempo , Línea Celular , Células Madre Embrionarias Humanas/metabolismo , Humanos , Programas Informáticos
13.
Blood ; 126(5): 629-39, 2015 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-26041741

RESUMEN

Adoptive transfer of T cells genetically modified to express chimeric antigen receptors (CARs) targeting the CD19 B cell-associated protein have demonstrated potent activity against relapsed/refractory B-lineage acute lymphoblastic leukemia (B-ALL). Not all patients respond, and CD19-negative relapses have been observed. Overexpression of the thymic stromal lymphopoietin receptor (TSLPR; encoded by CRLF2) occurs in a subset of adults and children with B-ALL and confers a high risk of relapse. Recent data suggest the TSLPR signaling axis is functionally important, suggesting that TSLPR would be an ideal immunotherapeutic target. We constructed short and long CARs targeting TSLPR and tested efficacy against CRLF2-overexpressing B-ALL. Both CARs demonstrated activity in vitro, but only short TSLPR CAR T cells mediated leukemia regression. In vivo activity of the short CAR was also associated with long-term persistence of CAR-expressing T cells. Short TSLPR CAR treatment of mice engrafted with a TSLPR-expressing ALL cell line induced leukemia cytotoxicity with efficacy comparable with that of CD19 CAR T cells. Short TSLPR CAR T cells also eradicated leukemia in 4 xenograft models of human CRLF2-overexpressing ALL. Finally, TSLPR has limited surface expression on normal tissues. TSLPR-targeted CAR T cells thus represent a potent oncoprotein-targeted immunotherapy for high-risk ALL.


Asunto(s)
Inmunoterapia Adoptiva/métodos , Leucemia-Linfoma Linfoblástico de Células Precursoras B/terapia , Receptores de Citocinas/antagonistas & inhibidores , Linfocitos T/inmunología , Animales , Antígenos CD19/metabolismo , Línea Celular Tumoral , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Leucemia-Linfoma Linfoblástico de Células Precursoras B/inmunología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Cell Rep ; 10(4): 616-32, 2015 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-25640183

RESUMEN

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


Asunto(s)
Neuronas/citología , Neuronas/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Proteínas Wnt/metabolismo , Proteína Wnt3/metabolismo , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Línea Celular , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Transducción de Señal , Proteínas Wnt/genética , Proteína Wnt3/genética
15.
J Vis Exp ; (89)2014 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-25077932

RESUMEN

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


Asunto(s)
Técnicas Citológicas/métodos , Inhibidores Enzimáticos/farmacología , Células Madre Pluripotentes/efectos de los fármacos , Células Madre Pluripotentes/fisiología , Quinasas Asociadas a rho/antagonistas & inhibidores , Amidas/farmacología , Animales , Dioxanos/farmacología , Fibroblastos/citología , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/fisiología , Laminina/farmacología , Ratones , Células Madre Pluripotentes/citología , Piridinas/farmacología , Pirimidinas/farmacología , Tiazoles/farmacología , Transfección
16.
Stem Cells Transl Med ; 3(7): 867-78, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24855277

RESUMEN

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


Asunto(s)
Bioensayo/métodos , Diferenciación Celular , Condrocitos/metabolismo , Condrogénesis , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Mesenquimatosas/metabolismo , Osteoblastos/metabolismo , Osteogénesis , Anciano , Anciano de 80 o más Años , Animales , Línea Celular , Reprogramación Celular , Condrocitos/trasplante , Femenino , Regulación del Desarrollo de la Expresión Génica , Humanos , Células Madre Pluripotentes Inducidas/trasplante , Masculino , Trasplante de Células Madre Mesenquimatosas , Ratones , Osteoblastos/trasplante , Fenotipo , Transfección
17.
Stem Cell Res ; 12(3): 610-21, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24603366

RESUMEN

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


Asunto(s)
Diferenciación Celular , Proliferación Celular , Mamíferos/embriología , Células Madre Pluripotentes/citología , Transducción de Señal , Animales , Humanos , Mamíferos/genética , Mamíferos/metabolismo , Células Madre Pluripotentes/metabolismo
18.
Cell Stem Cell ; 14(1): 13-26, 2014 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-24388173

RESUMEN

Human pluripotent stem cells (hPSCs) provide powerful resources for application in regenerative medicine and pharmaceutical development. In the past decade, various methods have been developed for large-scale hPSC culture that rely on combined use of multiple growth components, including media containing various growth factors, extracellular matrices, 3D environmental cues, and modes of multicellular association. In this Protocol Review, we dissect these growth components by comparing cell culture methods and identifying the benefits and pitfalls associated with each one. We further provide criteria, considerations, and suggestions to achieve optimal cell growth for hPSC expansion, differentiation, and use in future therapeutic applications.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/trasplante , Diferenciación Celular , Humanos
19.
Stem Cell Res ; 12(2): 376-86, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24374290

RESUMEN

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


Asunto(s)
Células Madre Pluripotentes Inducidas/fisiología , Células-Madre Neurales/fisiología , Animales , Diferenciación Celular/fisiología , Metilación de ADN , Femenino , Impresión Genómica , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Masculino , Proteínas de la Membrana/genética , Ratones , Proteínas del Tejido Nervioso/genética , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Óxido Nítrico/metabolismo , Transcriptoma
20.
Stem Cell Res ; 10(1): 57-66, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23117585

RESUMEN

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


Asunto(s)
Bases de Datos Factuales , Células Madre Pluripotentes/metabolismo , Animales , Línea Celular , Perfilación de la Expresión Génica , Humanos , Ratones , National Institutes of Health (U.S.) , Células Madre Pluripotentes/citología , Análisis de Componente Principal , Estados Unidos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA