Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Stem Cells ; 37(2): 202-215, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30376208

RESUMEN

Anterior-posterior (A-P) specification of the neural tube involves initial acquisition of anterior fate followed by the induction of posterior characteristics in the primitive anterior neuroectoderm. Several morphogens have been implicated in the regulation of A-P neural patterning; however, our understanding of the upstream regulators of these morphogens remains incomplete. Here, we show that the Krüppel-like zinc finger transcription factor GLI-Similar 3 (GLIS3) can direct differentiation of human embryonic stem cells (hESCs) into posterior neural progenitor cells in lieu of the default anterior pathway. Transcriptomic analyses reveal that this switch in cell fate is due to rapid activation of Wingless/Integrated (WNT) signaling pathway. Mechanistically, through genome-wide RNA-Seq, ChIP-Seq, and functional analyses, we show that GLIS3 binds to and directly regulates the transcription of several WNT genes, including the strong posteriorizing factor WNT3A, and that inhibition of WNT signaling is sufficient to abrogate GLIS3-induced posterior specification. Our findings suggest a potential role for GLIS3 in the regulation of A-P specification through direct transcriptional activation of WNT genes. Stem Cells 2018 Stem Cells 2019;37:202-215.


Asunto(s)
Proteínas de Unión al ADN/genética , Células Madre Embrionarias Humanas/citología , Células-Madre Neurales/citología , Proteínas Represoras/genética , Transactivadores/genética , Diferenciación Celular/fisiología , Células Cultivadas , Células Madre Embrionarias Humanas/metabolismo , Humanos , Células-Madre Neurales/metabolismo , Activación Transcripcional , Vía de Señalización Wnt
2.
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
3.
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.

4.
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.

5.
Stem Cells ; 30(5): 997-1007, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22378611

RESUMEN

Recent studies suggested that induced pluripotent stem cells (iPSCs) retain a residual donor cell gene expression, which may impact their capacity to differentiate into cell of origin. Here, we addressed a contribution of a lineage stage-specific donor cell memory in modulating the functional properties of iPSCs. iPSCs were generated from hepatic lineage cells at an early (hepatoblast-derived, HB-iPSCs) and end stage (adult hepatocyte, AH-iPSCs) of hepatocyte differentiation as well as from mouse embryonic fibroblasts (MEFs-iPSCs) using a lentiviral vector encoding four pluripotency-inducing factors Oct4, Sox2, Klf4, and c-Myc. All resulting iPSC lines acquired iPSCs phenotype as judged by the accepted criteria including morphology, expression of pluripotency markers, silencing of transducing factors, capacity of multilineage differentiation in teratoma assay, and normal diploid karyotype. However, HB-iPSCs were more efficient in directed differentiation toward hepatocytic lineage as compared to AH-iPSCs, MEF-iPSCs, or mouse embryonic stem cells (mESCs). Extensive comparative transcriptome analyses of the early passage iPSCs, donor cells, and mESCs revealed that despite global similarities in gene expression patterns between generated iPSCs and mESCs, HB-iPSCs retained a transcriptional memory (seven upregulated and 17 downregulated genes) typical of the original cells. Continuous passaging of HB-iPSCs erased most of these differences including a superior capacity for hepatic redifferentiation. These results suggest that retention of lineage stage-specific donor memory in iPSCs may facilitate differentiation into donor cell type. The identified gene set may help to improve hepatic differentiation for therapeutic applications and contribute to the better understanding of liver development.


Asunto(s)
Desdiferenciación Celular , Hepatocitos/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Hígado/metabolismo , Factores de Transcripción/biosíntesis , Animales , Células HEK293 , Hepatocitos/citología , Humanos , Células Madre Pluripotentes Inducidas/citología , Factor 4 Similar a Kruppel , Lentivirus , Hígado/citología , Ratones , Factores de Transcripción/genética , Transducción Genética
6.
Stem Cells ; 30(10): 2175-87, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22887864

RESUMEN

The expression and function of several multidrug transporters (including ABCB1 and ABCG2) have been studied in human cancer cells and in mouse and human adult stem cells. However, the expression of ABCG2 in human embryonic stem cells (hESCs) remains unclear. Limited and contradictory results in the literature from two research groups have raised questions regarding its expression and function. In this study, we used quantitative real-time PCR, Northern blots, whole genome RNA sequencing, Western blots, and immunofluorescence microscopy to study ABCG2 expression in hESCs. We found that full-length ABCG2 mRNA transcripts are expressed in undifferentiated hESC lines. However, ABCG2 protein was undetectable even under embryoid body differentiation or cytotoxic drug induction. Moreover, surface ABCG2 protein was coexpressed with the differentiation marker stage-specific embryonic antigen-1 of hESCs, following constant BMP-4 signaling at days 4 and 6. This expression was tightly correlated with the downregulation of two microRNAs (miRNAs) (i.e., hsa-miR-519c and hsa-miR-520h). Transfection of miRNA mimics and inhibitors of these two miRNAs confirmed their direct involvement in the regulation ABCG2 translation. Our findings clarify the controversy regarding the expression of the ABCG2 gene and also provide new insights into translational control of the expression of membrane transporter mRNAs by miRNAs in hESCs.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Células Madre Embrionarias/metabolismo , Regulación de la Expresión Génica , MicroARNs/genética , Proteínas de Neoplasias/genética , ARN Mensajero/biosíntesis , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 4/metabolismo , Diferenciación Celular , Células Cultivadas , Células Madre Embrionarias/citología , Células Nutrientes , Fibroblastos , Humanos , Antígeno Lewis X/genética , Antígeno Lewis X/metabolismo , Ratones , MicroARNs/metabolismo , Proteínas de Neoplasias/metabolismo , Biosíntesis de Proteínas , Transfección
7.
Mol Vis ; 18: 920-36, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22539871

RESUMEN

PURPOSE: To examine the potential of NIH-maintained human embryonic stem cell (hESC) lines TE03 and UC06 to differentiate into retinal progenitor cells (hESC-RPCs) using the noggin/Dkk-1/IGF-1/FGF9 protocol. An additional goal is to examine the in vivo dynamics of maturation and retinal integration of subretinal and epiretinal (vitreous space) hESC-RPC grafts without immunosuppression. METHODS: hESCs were neuralized in vitro with noggin for 2 weeks and expanded to derive neuroepithelial cells (hESC-neural precursors, NPs). Wnt (Integration 1 and wingless) blocking morphogens Dickkopf-1 (Dkk-1) and Insulin-like growth factor 1 (IGF-1) were used to direct NPs to a rostral neural fate, and fibroblast growth factor 9 (FGF9)/fibroblast growth factor-basic (bFGF) were added to bias the differentiation of developing anterior neuroectoderm cells to neural retina (NR) rather than retinal pigment epithelium (RPE). Cells were dissociated and grafted into the subretinal and epiretinal space of young adult (4-6-week-old) mice (C57BL/6J x129/Sv mixed background). Remaining cells were replated for (i) immunocytochemical analysis and (ii) used for quantitative reverse transcription polymerase chain reaction (qRT-PCR) analysis. Mice were sacrificed 3 weeks or 3 months after grafting, and the grafts were examined by histology and immunohistochemistry for survival of hESC-RPCs, presence of mature neuronal and retinal markers, and the dynamics of in vivo maturation and integration into the host retina. RESULTS: At the time of grafting, hESC-RPCs exhibited immature neural/neuronal immunophenotypes represented by nestin and neuronal class III ß-tubulin, with about half of the cells positive for cell proliferation marker Kiel University -raised antibody number 67 (Ki67), and no recoverin-positive (recoverin [+]) cells. The grafted cells expressed eye field markers paired box 6 (PAX6), retina and anterior neural fold homeobox (RAX), sine oculis homeobox homolog 6 (SIX6), LIM homeobox 2 (LHX2), early NR markers (Ceh-10 homeodomain containing homolog [CHX10], achaete-scute complex homolog 1 [MASH1], mouse atonal homolog 5 [MATH5], neurogenic differentiation 1 [NEUROD1]), and some retinal cell fate markers (brain-specific homeobox/POU domain transcription factor 3B [BRN3B], prospero homeobox 1 [PROX1], and recoverin). The cells in the subretinal grafts matured to predominantly recoverin [+] phenotype by 3 months and survived in a xenogenic environment without immunosuppression as long as the blood-retinal barrier was not breached by the transplantation procedure. The epiretinal grafts survived but did not express markers of mature retinal cells. Retinal integration into the retinal ganglion cell (RGC) layer and the inner nuclear layer (INL) was efficient from the epiretinal but not subretinal grafts. The subretinal grafts showed limited ability to structurally integrate into the host retina and only in cases when NR was damaged during grafting. Only limited synaptogenesis and no tumorigenicity was observed in grafts. CONCLUSIONS: Our studies show that (i) immunosuppression is not mandatory to xenogenic graft survival in the retina, (ii) the subretinal but not the epiretinal niche can promote maturation of hESC-RPCs to photoreceptors, and (iii) the hESC-RPCs from epiretinal but not subretinal grafts can efficiently integrate into the RGC layer and INL. The latter could be of value for long-lasting neuroprotection of retina in some degenerative conditions and glaucoma. Overall, our results provide new insights into the technical aspects associated with cell-based therapy in the retina.


Asunto(s)
Células Madre Embrionarias/citología , Células Fotorreceptoras/citología , Retina/trasplante , Neuronas Retinianas/citología , Animales , Biomarcadores/análisis , Proteínas Portadoras/farmacología , Diferenciación Celular/efectos de los fármacos , Línea Celular , Supervivencia Celular , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Factor 9 de Crecimiento de Fibroblastos/farmacología , Humanos , Inmunocompetencia , Factor I del Crecimiento Similar a la Insulina/farmacología , Péptidos y Proteínas de Señalización Intercelular/farmacología , Ratones , Células Fotorreceptoras/efectos de los fármacos , Células Fotorreceptoras/metabolismo , Retina/citología , Retina/metabolismo , Neuronas Retinianas/efectos de los fármacos , Neuronas Retinianas/metabolismo , Trasplante Heterólogo
8.
J Surg Res ; 170(2): e253-61, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21816427

RESUMEN

OBJECTIVE: The derivation of hepatocytes from human embryonic stem (hES) cells is of value both in the study of early human liver organogenesis and in the creation of an unlimited source of donor cells for hepatocyte transplantation therapy. Here, we report the generation of hepatocyte-like cells derived from hES cells. METHODS: Hepatic endoderm cells were generated by adding activin A for 5 d- to 1-d-old embryoid bodies formed from hES cells. The hepatic endoderm cells were cocultured with mitomycin treated 3T3-J2 feeder cells. RESULTS: After co-culture with mitomycin treated 3T3-J2 feeder cells, these hepatic endodermal cells yielded hepatocyte-like cell colonies, which possessed the proliferation potential to be cultured for an extended period of more than 30 d. With extensive expansion, they co-expressed the hepatic marker AFP and albumin, indicating that they were hepatocyte-like cells. CONCLUSIONS: We report the generation of proliferative hepatocyte-like cells from hES cells. These hES cell derived hepatic cells can effectively be used as in vitro model for studying the mechanisms of hepatic stem/progenitor cell origin, self-renewal and differentiation.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Embrionarias/citología , Hepatocitos/citología , Células del Estroma/citología , Células 3T3 , Animales , Biomarcadores/metabolismo , Técnicas de Cultivo de Célula/métodos , División Celular/fisiología , Células Cultivadas , Técnicas de Cocultivo , Cuerpos Embrioides/fisiología , Endodermo/citología , Proteínas Fluorescentes Verdes/genética , Humanos , Ratones , Mitomicina/farmacología , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Células del Estroma/efectos de los fármacos
9.
Nat Cell Biol ; 23(8): 822-833, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34341531

RESUMEN

Clinical management of patients with severe complications of COVID-19 has been hindered by a lack of effective drugs and a failure to capture the extensive heterogeneity of the disease with conventional methods. Here we review the emerging roles of complex organoids in the study of SARS-CoV-2 infection, modelling of COVID-19 disease pathology and in drug, antibody and vaccine development. We discuss opportunities for COVID-19 research and remaining challenges in the application of organoids.


Asunto(s)
COVID-19/inmunología , COVID-19/metabolismo , Organoides/metabolismo , SARS-CoV-2/patogenicidad , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/metabolismo , COVID-19/prevención & control , Vacunas contra la COVID-19/inmunología , Vacunas contra la COVID-19/uso terapéutico , Humanos , SARS-CoV-2/inmunología
10.
Mol Cell Biol ; 27(9): 3499-510, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17339341

RESUMEN

Transcriptional insulators are specialized cis-acting elements that protect promoters from inappropriate activation by distal enhancers. The H19 imprinting control region (ICR) functions as a CTCF-dependent, methylation-sensitive transcriptional insulator. We analyzed several insertional mutations and demonstrate that the ICR can function as a methylation-regulated maternal chromosome-specific insulator in novel chromosomal contexts. We used chromosome conformation capture and chromatin immunoprecipitation assays to investigate the configuration of cis-acting elements at these several insertion sites. By comparing maternal and paternal organizations on wild-type and mutant chromosomes, we hoped to identify mechanisms for ICR insulator function. We found that promoter and enhancer elements invariably associate to form DNA loop domains at transcriptionally active loci. Conversely, active insulators always prevent these promoter-enhancer interactions. Instead, the ICR insulator forms novel loop domains by associating with the blocked promoters and enhancers. We propose that these associations are fundamental to insulator function.


Asunto(s)
Cromosomas de los Mamíferos/genética , Elementos Aisladores/genética , Animales , Factor de Unión a CCCTC , Inmunoprecipitación de Cromatina , Metilación de ADN , Proteínas de Unión al ADN/genética , Elementos de Facilitación Genéticos/genética , Regulación de la Expresión Génica , Hígado/embriología , Hígado/metabolismo , Ratones , Madres , Células Musculares/metabolismo , Regiones Promotoras Genéticas/genética , Proteínas/genética , Proteínas Represoras/genética , alfa-Fetoproteínas/genética
11.
J Vis Exp ; (155)2020 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-32009654

RESUMEN

In vitro differentiation of human embryonic stem cells (hESCs) has transformed the ability to study human development on both biological and molecular levels and provided cells for use in regenerative applications. Standard approaches for hESC culture using colony type culture to maintain undifferentiated hESCs and embryoid body (EB) and rosette formation for differentiation into different germ layers are inefficient and time-consuming. Presented here is a single-cell culture method using hESCs instead of a colony-type culture. This method allows maintenance of the characteristic features of undifferentiated hESCs, including expression of hESC markers at levels comparable to colony type hESCs. In addition, the protocol presents an efficient method for neural progenitor cell (NPC) generation from single-cell type hESCs that produces NPCs within 1 week. These cells highly express several NPC marker genes and can differentiate into various neural cell types, including dopaminergic neurons and astrocytes. This single-cell culture system for hESCs will be useful in investigating the molecular mechanisms of these processes, studies of certain diseases, and drug discovery screens.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Células Madre Embrionarias Humanas/metabolismo , Diferenciación Celular , Línea Celular , Humanos
12.
13.
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
14.
Mol Cell Biol ; 24(9): 3588-95, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15082756

RESUMEN

Igf2 and H19 are coordinately regulated imprinted genes physically linked on the distal end of mouse chromosome 7. Genetic analyses demonstrate that the differentially methylated region (DMR) upstream of the H19 gene is necessary for three distinct functions: transcriptional insulation of the maternal Igf2 allele, transcriptional silencing of paternal H19 allele, and marking of the parental origin of the two chromosomes. To test the sufficiency of the DMR for the third function, we inserted DMR at two heterologous positions in the genome, downstream of H19 and at the alpha-fetoprotein locus on chromosome 5. Our results demonstrate that the DMR alone is sufficient to act as a mark of parental origin. Moreover, this activity is not dependent on germ line differences in DMR methylation. Thus, the DMR can mark its parental origin by a mechanism independent of its own DNA methylation.


Asunto(s)
Cromosomas/metabolismo , Metilación de ADN , Impresión Genómica , Proteínas/genética , ARN no Traducido/genética , Animales , Cromosomas/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Silenciador del Gen , Humanos , Masculino , Ratones , Biosíntesis de Proteínas , Proteínas/metabolismo , ARN Largo no Codificante
15.
Cell Death Differ ; 24(6): 1017-1028, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28362428

RESUMEN

p53 functions to induce cellular senescence, which is incompatible with self-renewal of pluripotent stem cells such as induced pluripotent stem cells (iPSC) and embryonic stem cells (ESC). However, p53 also has essential roles in these cells through DNA damage repair for maintaining genomic integrity and high sensitivity to apoptosis for eliminating severely damaged cells. We hypothesized that Δ133p53, a physiological inhibitory p53 isoform, is involved in the balanced regulation of self-renewing capacity, DNA damage repair and apoptosis. We examined 12 lines of human iPSC and their original fibroblasts, as well as three ESC lines, for endogenous protein levels of Δ133p53 and full-length p53 (FL-p53), and mRNA levels of various p53 target genes. While FL-p53 levels in iPSC and ESC widely ranged from below to above those in the fibroblasts, all iPSC and ESC lines expressed elevated levels of Δ133p53. The p53-inducible genes that mediate cellular senescence (p21WAF1, miR-34a, PAI-1 and IGFBP7), but not those for apoptosis (BAX and PUMA) and DNA damage repair (p53R2), were downregulated in iPSC and ESC. Consistent with these endogenous expression profiles, overexpression of Δ133p53 in human fibroblasts preferentially repressed the p53-inducible senescence mediators and significantly enhanced their reprogramming to iPSC. The iPSC lines derived from Δ133p53-overexpressing fibroblasts formed well-differentiated, benign teratomas in immunodeficient mice and had fewer numbers of somatic mutations than an iPSC derived from p53-knocked-down fibroblasts, suggesting that Δ133p53 overexpression is non- or less oncogenic and mutagenic than total inhibition of p53 activities. Overexpressed Δ133p53 prevented FL-p53 from binding to the regulatory regions of p21WAF1 and miR-34a promoters, providing a mechanistic basis for its dominant-negative inhibition of a subset of p53 target genes. This study supports the hypothesis that upregulation of Δ133p53 is an endogenous mechanism that facilitates human somatic cells to become self-renewing pluripotent stem cells with maintained apoptotic and DNA repair activities.


Asunto(s)
Desdiferenciación Celular , Fibroblastos/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Aminoácidos , Animales , Línea Celular , Senescencia Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Fibroblastos/fisiología , Regulación de la Expresión Génica , Humanos , Células Madre Pluripotentes Inducidas , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/genética , Ratones , Ratones Endogámicos NOD , Ratones SCID , MicroARNs/genética , Inhibidor 1 de Activador Plasminogénico/genética , Isoformas de Proteínas , Eliminación de Secuencia , Proteína p53 Supresora de Tumor/genética
16.
Int J Biochem Cell Biol ; 38(7): 1063-75, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16469522

RESUMEN

The culture of human embryonic stem cells (hESCs) is limited, both technically and with respect to clinical potential, by the use of mouse embryonic fibroblasts (MEFs) as a feeder layer. The concern over xenogeneic contaminants from the mouse feeder cells may restrict transplantation to humans and the variability in MEFs from batch-to-batch and laboratory-to-laboratory may contribute to some of the variability in experimental results. Finally, use of any feeder layer increases the work load and subsequently limits the large-scale culture of human ES cells. Thus, the development of feeder-free cultures will allow more reproducible culture conditions, facilitate scale-up and potentiate the clinical use of cells differentiated from hESC cultures. In this review, we describe various methods tested to culture cells in the absence of MEF feeder layers and other advances in eliminating xenogeneic products from the culture system.


Asunto(s)
Técnicas de Cocultivo/métodos , Embrión de Mamíferos/citología , Células Madre/citología , Animales , Antígenos Heterófilos/análisis , Antígenos Heterófilos/inmunología , Diferenciación Celular/efectos de los fármacos , Línea Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Medios de Cultivo Condicionados/análisis , Medios de Cultivo Condicionados/farmacología , Fibroblastos/metabolismo , Humanos , Ratones , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/efectos de los fármacos , Células Madre Pluripotentes/metabolismo , Proteínas Recombinantes/química , Células Madre/efectos de los fármacos
17.
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
18.
PLoS One ; 10(1): e0116032, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25587899

RESUMEN

Targeted genome engineering to robustly express transgenes is an essential methodology for stem cell-based research and therapy. Although designer nucleases have been used to drastically enhance gene editing efficiency, targeted addition and stable expression of transgenes to date is limited at single gene/locus and mostly PPP1R12C/AAVS1 in human stem cells. Here we constructed transcription activator-like effector nucleases (TALENs) targeting the safe-harbor like gene CLYBL to mediate reporter gene integration at 38%-58% efficiency, and used both AAVS1-TALENs and CLYBL-TALENs to simultaneously knock-in multiple reporter genes at dual safe-harbor loci in human induced pluripotent stem cells (iPSCs) and neural stem cells (NSCs). The CLYBL-TALEN engineered cell lines maintained robust reporter expression during self-renewal and differentiation, and revealed that CLYBL targeting resulted in stronger transgene expression and less perturbation on local gene expression than PPP1R12C/AAVS1. TALEN-mediated CLYBL engineering provides improved transgene expression and options for multiple genetic modification in human stem cells.


Asunto(s)
Endonucleasas/genética , Marcación de Gen/métodos , Células Madre Pluripotentes Inducidas/citología , Células-Madre Neurales/citología , Línea Celular , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células-Madre Neurales/metabolismo , Transgenes/genética
19.
Neuromuscul Disord ; 13(3): 252-8, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12609507

RESUMEN

Desmin myopathy is a familial or sporadic disorder characterized by the presence of desmin mutations that cause skeletal muscle weakness associated with cardiac conduction block, arrhythmia and heart failure. Distinctive histopathologic features include intracytoplasmic accumulation of desmin-reactive deposits and electron-dense granular aggregates in skeletal and cardiac muscle cells. We describe two families with features of adult-onset slowly progressive skeletal myopathy without cardiomyopathy. N342D point mutation was present in the desmin helical rod domain in patients of family 1, and I451M mutation was found in the non-helical tail domain in patients of family 2. Of interest, the same I451M mutation has previously been reported in patients with cardiomyopathy and no signs of skeletal myopathy. Some carriers of the I451M mutation did not develop any disease, suggesting incomplete penetrance. Expression studies demonstrated inability of the N342D mutant desmin to form cellular filamentous network, confirming the pathogenic role of this mutation, but the network was not affected by the tail-domain I451M mutation. Progressive skeletal myopathy is a rare phenotypic variant of desmin myopathy allelic to the more frequent cardio-skeletal form.


Asunto(s)
Desmina/genética , Enfermedades Musculares/genética , Mutación Puntual , Alanina/genética , Animales , Carcinoma/metabolismo , Línea Celular , Cisteína/genética , Análisis Mutacional de ADN , Desmina/metabolismo , Femenino , Técnica del Anticuerpo Fluorescente/métodos , Glicina/genética , Humanos , Masculino , Metionina/genética , Ratones , Datos de Secuencia Molecular , Enfermedades Musculares/etiología , Enfermedades Musculares/patología , Mioblastos/metabolismo , Linaje , Fenotipo , Transfección/métodos
20.
J Neurol ; 251(2): 143-9, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14991347

RESUMEN

Desminopathy is a familial or sporadic cardiac and skeletal muscular dystrophy associated with mutations in desmin. We have previously characterized a de novo desmin R406W mutation in a patient of European origin with early onset muscle weakness in the lower extremities and atrioventricular conduction block requiring a permanent pacemaker. The disease relentlessly progressed resulting in severe incapacity within 5 years after onset. We have now identified three other patients with early onset rapidly progressive cardiac and skeletal myopathy caused by this same desmin R406W mutation. The mutation was present in each studied patient, but not in their parents or other unaffected family members, indicating that the mutation in all four cases was generated de novo. The patients' mutation-carrying chromosomes showed no similarity, suggesting that the R406W mutation has occurred independently. These observations strongly confirm that the de novo R406W desmin mutation is the genetic basis for early-onset cardiac and skeletal myopathy in patients with sporadic disease and indicate that desmin position 406 is a hot spot for spontaneous mutations. The high pathogenic potential of this mutation can be explained by its location in the highly conserved YRKLLEGEE motif at the C-terminal end of the 2B helix that has a critical role in the process of desmin filament assembly.


Asunto(s)
Cardiomiopatías/genética , Desmina/genética , Músculo Esquelético/fisiopatología , Enfermedades Musculares/genética , Mutación/genética , Miocardio/patología , Adolescente , Adulto , Secuencias de Aminoácidos/genética , Sustitución de Aminoácidos , Secuencia de Bases/genética , Cardiomiopatías/fisiopatología , Secuencia Conservada , Progresión de la Enfermedad , Europa (Continente) , Femenino , Bloqueo Cardíaco/genética , Bloqueo Cardíaco/fisiopatología , Humanos , Masculino , Modelos Moleculares , Debilidad Muscular/genética , Debilidad Muscular/fisiopatología , Músculo Esquelético/patología , Enfermedades Musculares/fisiopatología , Linaje , Estructura Secundaria de Proteína/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA