Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Stem Cell Reports ; 19(1): 1-10, 2024 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-38157849

RESUMEN

The expression of one or more of a small number of molecules, typically cell surface-associated antigens, or transcription factors, is widely used for identifying pluripotent stem cells (PSCs) or for monitoring their differentiation. However, none of these marker molecules are uniquely expressed by PSCs and all are expressed by stem cells that have lost the ability to differentiate. Consequently, none are indicators of pluripotency, per se. Here we summarize the nature and characteristics of several markers that are in wide use, including the cell surface antigens, stage-specific embryonic antigen (SSEA)-1, SSEA-3, SSEA-4, TRA-1-60, TRA-1-81, GCTM2, and the transcription factors POUF5/OCT4, NANOG, and SOX2, highlighting issues that must be considered when interpreting data about their expression on putative PSCs.


Asunto(s)
Células Madre Pluripotentes , Células Madre Pluripotentes/metabolismo , Antígeno Lewis X/metabolismo , Diferenciación Celular , Factores de Transcripción/genética , Antígenos de Superficie/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/metabolismo
2.
Stem Cell Reports ; 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38964325

RESUMEN

Culture-acquired variants in human pluripotent stem cells (hPSCs) hinder their applications in research and clinic. However, the mechanisms that underpin selection of variants remain unclear. Here, through analysis of comprehensive karyotyping datasets from over 23,000 hPSC cultures of more than 1,500 lines, we explored how culture conditions shape variant selection. Strikingly, we identified an association of chromosome 1q gains with feeder-free cultures and noted a rise in its prevalence in recent years, coinciding with increased usage of feeder-free regimens. Competition experiments of multiple isogenic lines with and without a chromosome 1q gain confirmed that 1q variants have an advantage in feeder-free (E8/vitronectin), but not feeder-based, culture. Mechanistically, we show that overexpression of MDM4, located on chromosome 1q, drives variants' advantage in E8/vitronectin by alleviating genome damage-induced apoptosis, which is lower in feeder-based conditions. Our study explains condition-dependent patterns of hPSC aberrations and offers insights into the mechanisms of variant selection.

3.
Stem Cells ; 30(9): 1901-10, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22821732

RESUMEN

Human embryonic stem cells (hESCs) tend to lose genomic integrity during long periods of culture in vitro and to acquire a cancer-like phenotype. In this study, we aim at understanding the contribution of point mutations to the adaptation process and at providing a mechanistic explanation for their accumulation. We observed that, due to the absence of p21/Waf1/Cip1, cultured hESCs lack proper cell cycle checkpoints and are vulnerable to the kind of DNA damage usually repaired by the highly versatile nucleotide excision repair (NER) pathway. In response to UV-induced DNA damage, the majority of hESCs succumb to apoptosis; however, a subpopulation continues to proliferate, carrying damaged DNA and accumulating point mutations with a typical UV-induced signature. The UV-resistant cells retain their proliferative capacity and potential for pluripotent differentiation and are markedly less apoptotic to subsequent UV exposure. These findings demonstrate that, due to deficient DNA damage response, the modest NER activity in hESCs is insufficient to prevent increased mutagenesis. This provides for the appearance of genetically aberrant hESCs, paving the way for further major genetic changes.


Asunto(s)
Puntos de Control del Ciclo Celular/genética , Daño del ADN , Reparación del ADN , Células Madre Embrionarias/fisiología , Mutación Puntual , Apoptosis/genética , Procesos de Crecimiento Celular/genética , Células Cultivadas , Células Madre Embrionarias/citología , Humanos
4.
Stem Cells ; 28(5): 863-73, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20235236

RESUMEN

Transforming growth factor (TGF)-beta superfamily proteins play a key role in the regulation of human embryonic stem cells (hESCs). Those of the TGFbeta/activin/nodal branch seem to support self-renewal and pluripotency, whereas those of the bone morphogenic protein (BMP) branch induce differentiation. In contrast to this generalization, we found that hESC remained undifferentiated after knockdown of SMAD4 with inducible short hairpin RNA interference, although the knockdown inhibited TGFbeta signaling and rendered the cells nonresponsive to BMP-induced differentiation. Moreover, the rapid differentiation of hESC after pharmacological inhibition of TGFbeta/activin/nodal receptor signaling was restricted after SMAD4 knockdown. These results suggest that TGFbeta/activin/nodal signaling supports the undifferentiated phenotype of hESC by suppressing BMP activity. During long-term culture, SMAD4 knockdown cell populations became less stable and more permissive to neural induction, a situation that was rescued by re-establishment of SMAD4 expression. These results suggest that SMAD4 is not required for maintenance of the undifferentiated state of hESC, but rather to stabilize that state.


Asunto(s)
División Celular/genética , Linaje de la Célula/genética , Células Madre Embrionarias/metabolismo , Proteína Smad4/genética , Activinas/metabolismo , Diferenciación Celular/genética , Células Cultivadas , Regulación hacia Abajo/fisiología , Células Madre Embrionarias/citología , Humanos , Proteína Nodal/metabolismo , Interferencia de ARN/fisiología , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/metabolismo
5.
Dev Cell ; 56(17): 2455-2470.e10, 2021 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-34407428

RESUMEN

The appearance of genetic changes in human pluripotent stem cells (hPSCs) presents a concern for their use in research and regenerative medicine. Variant hPSCs that harbor recurrent culture-acquired aneuploidies display growth advantages over wild-type diploid cells, but the mechanisms that yield a drift from predominantly wild-type to variant cell populations remain poorly understood. Here, we show that the dominance of variant clones in mosaic cultures is enhanced through competitive interactions that result in the elimination of wild-type cells. This elimination occurs through corralling and mechanical compression by faster-growing variants, causing a redistribution of F-actin and sequestration of yes-associated protein (YAP) in the cytoplasm that induces apoptosis in wild-type cells. YAP overexpression or promotion of YAP nuclear localization in wild-type cells alleviates their "loser" phenotype. Our results demonstrate that hPSC fate is coupled to mechanical cues imposed by neighboring cells and reveal that hijacking this mechanism allows variants to achieve clonal dominance in cultures.


Asunto(s)
Competencia Celular/genética , Diferenciación Celular/genética , Proliferación Celular/genética , Células Madre Pluripotentes/citología , Proteínas Señalizadoras YAP/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Células Cultivadas , Citoplasma/metabolismo , Humanos , Factores de Transcripción/metabolismo
6.
Biochem Soc Trans ; 38(4): 1046-50, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20659001

RESUMEN

Human ES (embryonic stem) cells and iPS (induced pluripotent stem) cells have been heralded as a source of differentiated cells that could be used in the treatment of degenerative diseases, such as Parkinson's disease or diabetes. Despite the great potential for their use in regenerative therapy, the challenge remains to understand the basic biology of these remarkable cells, in order to differentiate them into any functional cell type. Given the scale of the task, high-throughput screening of agents and culture conditions offers one way to accelerate these studies. The screening of small-compound libraries is particularly amenable to such high-throughput methods. Coupled with high-content screening technology that enables simultaneous assessment of multiple cellular features in an automated and quantitative way, this approach is proving powerful in identifying both small molecules as tools for manipulating stem cell fates and novel mechanisms of differentiation not previously associated with stem cell biology. Such screens performed on human ES cells also demonstrate the usefulness of human ES/iPS cells as cellular models for pharmacological testing of drug efficacy and toxicity, possibly a more imminent use of these cells than in regenerative medicine.


Asunto(s)
Células Madre Embrionarias/efectos de los fármacos , Ensayos Analíticos de Alto Rendimiento/métodos , Bibliotecas de Moléculas Pequeñas/análisis , Algoritmos , Diferenciación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Descubrimiento de Drogas/métodos , Células Madre Embrionarias/fisiología , Ensayos Analíticos de Alto Rendimiento/instrumentación , Humanos , Modelos Biológicos , Bibliotecas de Moléculas Pequeñas/farmacología
7.
Stem Cells ; 27(5): 1057-65, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19415777

RESUMEN

Human embryonic stem cells undergo adaptive changes that can increase their growth capacity upon prolonged culture in vitro. This is frequently associated with nonrandom karyotypic changes, commonly involving amplification of genetic material from chromosomes 12, 17, and X. A recent study suggested that the karyotypically abnormal cells can be identified by their expression of CD30, which confers resistance to apoptosis. We have now investigated CD30 expression and apoptosis in karyotypically normal and abnormal sublines of the human ES cell line, H7, but our results were contrary to those previously observed. In this cell line, CD30 expression did not segregate the normal and abnormal cells, and abnormal cells were not protected from apoptosis. These data suggest that culture adaptation can occur through a variety of mechanisms.


Asunto(s)
Adaptación Biológica , Células Madre Embrionarias/metabolismo , Antígeno Ki-1/metabolismo , Apoptosis , Carcinoma Embrionario/metabolismo , Carcinoma Embrionario/patología , Supervivencia Celular , Células Cultivadas , Citometría de Flujo , Humanos , Cariotipificación
8.
Nat Biotechnol ; 25(7): 803-16, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17572666

RESUMEN

The International Stem Cell Initiative characterized 59 human embryonic stem cell lines from 17 laboratories worldwide. Despite diverse genotypes and different techniques used for derivation and maintenance, all lines exhibited similar expression patterns for several markers of human embryonic stem cells. They expressed the glycolipid antigens SSEA3 and SSEA4, the keratan sulfate antigens TRA-1-60, TRA-1-81, GCTM2 and GCT343, and the protein antigens CD9, Thy1 (also known as CD90), tissue-nonspecific alkaline phosphatase and class 1 HLA, as well as the strongly developmentally regulated genes NANOG, POU5F1 (formerly known as OCT4), TDGF1, DNMT3B, GABRB3 and GDF3. Nevertheless, the lines were not identical: differences in expression of several lineage markers were evident, and several imprinted genes showed generally similar allele-specific expression patterns, but some gene-dependent variation was observed. Also, some female lines expressed readily detectable levels of XIST whereas others did not. No significant contamination of the lines with mycoplasma, bacteria or cytopathic viruses was detected.


Asunto(s)
Células Madre Embrionarias/citología , Regulación del Desarrollo de la Expresión Génica , Fosfatasa Alcalina/metabolismo , Antígenos CD/biosíntesis , Biotecnología/métodos , Diferenciación Celular , Linaje de la Célula , Membrana Celular/metabolismo , Células Cultivadas , Análisis por Conglomerados , Femenino , Perfilación de la Expresión Génica , Genotipo , Glucolípidos/química , Humanos , Glicoproteínas de Membrana/biosíntesis , Tetraspanina 29
9.
Stem Cell Reports ; 14(6): 1009-1017, 2020 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-32413278

RESUMEN

Human pluripotent stem cells (PSCs) are subject to the appearance of recurrent genetic variants on prolonged culture. We have now found that, compared with isogenic differentiated cells, PSCs exhibit evidence of considerably more DNA damage during the S phase of the cell cycle, apparently as a consequence of DNA replication stress marked by slower progression of DNA replication, activation of latent origins of replication, and collapse of replication forks. As in many cancers, which, like PSCs, exhibit a shortened G1 phase and DNA replication stress, the resulting DNA damage may underlie the higher incidence of abnormal and abortive mitoses in PSCs, resulting in chromosomal non-dysjunction or cell death. However, we have found that the extent of DNA replication stress, DNA damage, and consequent aberrant mitoses can be substantially reduced by culturing PSCs in the presence of exogenous nucleosides, resulting in improved survival, clonogenicity, and population growth.


Asunto(s)
Replicación del ADN , Inestabilidad Genómica , Nucleósidos/farmacología , Células Madre Pluripotentes/efectos de los fármacos , Línea Celular , Medios de Cultivo/química , Medios de Cultivo/farmacología , Citoprotección , Daño del ADN , Humanos , Mitosis , Nucleósidos/análisis , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo
10.
Nat Commun ; 11(1): 1528, 2020 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-32251294

RESUMEN

The occurrence of repetitive genomic changes that provide a selective growth advantage in pluripotent stem cells is of concern for their clinical application. However, the effect of different culture conditions on the underlying mutation rate is unknown. Here we show that the mutation rate in two human embryonic stem cell lines derived and banked for clinical application is low and not substantially affected by culture with Rho Kinase inhibitor, commonly used in their routine maintenance. However, the mutation rate is reduced by >50% in cells cultured under 5% oxygen, when we also found alterations in imprint methylation and reversible DNA hypomethylation. Mutations are evenly distributed across the chromosomes, except for a slight increase on the X-chromosome, and an elevation in intergenic regions suggesting that chromatin structure may affect mutation rate. Overall the results suggest that pluripotent stem cells are not subject to unusually high rates of genetic or epigenetic alterations.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Cromosomas Humanos X/genética , ADN Intergénico/genética , Tasa de Mutación , Células Madre Pluripotentes/fisiología , Línea Celular , Medios de Cultivo/farmacología , Metilación de ADN , Análisis Mutacional de ADN , Epigénesis Genética , Humanos , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/genética , Oxígeno/química , Oxígeno/farmacología , Análisis de Secuencia de ARN , Secuenciación Completa del Genoma
12.
Lab Invest ; 89(3): 259-62, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19153556

RESUMEN

Substantial advances in the biology of human embryonic stem (ES) cells, and the technology for working with them, have been made over the past 10 years. Regulatory frameworks for their study are well developed, although some countries remain particularly restrictive. Markers and criteria for characterising human ES cells are also generally agreed, and protocols for promoting their differentiation are being established, providing the groundwork for the development of applications over the next 10 years. The recent appearance of technology to convert somatic cells to 'induced Pluripotent Stem Cells' closely resembling ES cells will certainly speed up these developments.


Asunto(s)
Diferenciación Celular , Células Madre Embrionarias/fisiología , Células Madre Pluripotentes/fisiología , Animales , Biomarcadores , Células Madre Embrionarias/citología , Humanos , Ratones , Microscopía de Contraste de Fase , Células Madre Pluripotentes/citología
13.
Stem Cells ; 26(12): 3068-74, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18787205

RESUMEN

OCT4 is a master regulator of self-renewal in embryonic stem cells and can potentially encode two spliced variants, designated OCT4A and OCT4B. We have examined the expression pattern of these OCT4 isoforms in various human pluripotent and nonpluripotent cells. Our data revealed that whereas OCT4A expression is restricted to embryonic stem (ES) and embryonal carcinoma (EC) cells, OCT4B can be detected in various nonpluripotent cell types. Furthermore, we detected a novel OCT4 spliced variant, designated OCT4B1, that is expressed primarily in human ES and EC cells and is downregulated following their differentiation. We also found a significantly higher level of OCT4B1 expression in stage-specific embryonic antigen-3 (SSEA3)(+) compared with SSEA3(-) subpopulations of cultured ES cells. Taken together, our data demonstrated a distinctive expression pattern for OCT4 spliced variants in different cell types and highlight the necessity of defining the type of OCT4 when addressing the expression of this gene in different human cells.


Asunto(s)
Empalme Alternativo , Regulación de la Expresión Génica , Factor 3 de Transcripción de Unión a Octámeros/biosíntesis , Factor 3 de Transcripción de Unión a Octámeros/genética , Células Madre Pluripotentes/metabolismo , Células Madre/metabolismo , Antígenos de Carbohidratos Asociados a Tumores/biosíntesis , Diferenciación Celular , Línea Celular Tumoral , Células Madre Embrionarias/citología , Exones , Células HeLa , Humanos , Intrones , Células K562 , Isoformas de Proteínas , Antígenos Embrionarios Específico de Estadio/biosíntesis
14.
Stem Cells ; 26(3): 715-23, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18055449

RESUMEN

Unlike pluripotent mouse embryonic stem (ES) cells, human ES cells and their malignant equivalents, embryonal carcinoma (EC) cells, require close cell-cell contact for efficient growth. Signaling through the NOTCH receptor, initiated by interaction with ligands of the DELTA/JAGGED family expressed on neighboring cells, plays a role in regulating the self-renewal of several stem cell systems. Members of the NOTCH and DELTA/JAGGED families are expressed by human EC and ES cells, and we have therefore investigated the possible role of NOTCH in the maintenance of these cells. Cleavage of both NOTCH1 and NOTCH2 to yield the intracellular domain responsible for the canonical signaling pathway of NOTCH was detected in several human EC and ES cell lines, suggesting that NOTCH signaling is active. Furthermore, the proliferation of human EC cells, as well as the expression of several downstream NOTCH target genes, was markedly reduced after small interfering RNA knockdown of NOTCH1, NOTCH2, and the canonical effector CBF-1 or after blocking NOTCH signaling with the gamma-secretase inhibitor L-685,458. The inhibitor also caused a reduction in the growth of human ES cells, although without evidence of differentiation. The results indicate that cell-cell signaling through the NOTCH system provides a critical cue for the proliferation of human EC and ES cell in vitro.


Asunto(s)
Comunicación Celular , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Receptores Notch/metabolismo , Transducción de Señal , Animales , Anticuerpos Monoclonales , Antígenos de Superficie/metabolismo , Apoptosis/efectos de los fármacos , Carcinoma Embrionario/metabolismo , Carcinoma Embrionario/patología , Comunicación Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Células Madre Embrionarias/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Humanos , Ratones , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/efectos de los fármacos , Células Madre Pluripotentes/metabolismo , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Estructura Terciaria de Proteína , Transporte de Proteínas/efectos de los fármacos , Receptores Notch/química , Transducción de Señal/efectos de los fármacos
15.
Semin Reprod Med ; 24(5): 289-97, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17123223

RESUMEN

Stem cell research has stimulated considerable recent interest, but the concepts are old. Nevertheless, our understanding of the basic biology of different stem cell systems is poor. Many questions remain to be answered: How can we recognize stem cells? Are the underlying control mechanisms common to different types of stem cell, the so-called stemness concept, or is the control of self-renewal and commitment distinct in different stem cell types? What is the significance of differences in stem cells from different species? Do stem cells from somatic tissues really show plasticity with an ability to generate cells from distinct lineages, or are the observed examples consequences of experimental artifact, or rare events of no physiological significance? Do genetic mutations in the genes controlling stem cell self-renewal and differentiation lie at the heart of carcinogenesis? Answers to these and related questions now offer exciting future possibilities for both basic biology and medicine.


Asunto(s)
Investigación/tendencias , Células Madre/citología , Adaptación Biológica , Células Madre Adultas/citología , Animales , Biomarcadores , Proliferación Celular , Células Cultivadas , Progresión de la Enfermedad , Células Madre Embrionarias/citología , Humanos , Células Madre Multipotentes/citología , Neoplasias/patología , Células Madre/fisiología
16.
Cell Stem Cell ; 19(5): 653-662, 2016 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-27545503

RESUMEN

Adult neural stem cells (NSCs) are defined by their inherent capacity to self-renew and give rise to neurons, astrocytes, and oligodendrocytes. In vivo, however, hippocampal NSCs do not generate oligodendrocytes for reasons that have remained enigmatic. Here, we report that deletion of Drosha in adult dentate gyrus NSCs activates oligodendrogenesis and reduces neurogenesis at the expense of gliogenesis. We further find that Drosha directly targets NFIB to repress its expression independently of Dicer and microRNAs. Knockdown of NFIB in Drosha-deficient hippocampal NSCs restores neurogenesis, suggesting that the Drosha/NFIB mechanism robustly prevents oligodendrocyte fate acquisition in vivo. Taken together, our findings establish that adult hippocampal NSCs inherently possess multilineage potential but that Drosha functions as a molecular barrier preventing oligodendrogenesis.


Asunto(s)
Células Madre Adultas/citología , Envejecimiento/metabolismo , Hipocampo/citología , Células Madre Multipotentes/citología , Factores de Transcripción NFI/metabolismo , Células-Madre Neurales/citología , Ribonucleasa III/metabolismo , Células Madre Adultas/metabolismo , Animales , Secuencia de Bases , Diferenciación Celular , Giro Dentado/citología , Eliminación de Gen , Técnicas de Silenciamiento del Gen , Ratones , Ratones Noqueados , Células Madre Multipotentes/metabolismo , Factores de Transcripción NFI/genética , Células-Madre Neurales/metabolismo , Neurogénesis/genética , Oligodendroglía/citología , Oligodendroglía/metabolismo , Unión Proteica , ARN Mensajero/genética , ARN Mensajero/metabolismo
17.
Stem Cell Reports ; 7(5): 998-1012, 2016 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-27829140

RESUMEN

Genetic changes in human pluripotent stem cells (hPSCs) gained during culture can confound experimental results and potentially jeopardize the outcome of clinical therapies. Particularly common changes in hPSCs are trisomies of chromosomes 1, 12, 17, and 20. Thus, hPSCs should be regularly screened for such aberrations. Although a number of methods are used to assess hPSC genotypes, there has been no systematic evaluation of the sensitivity of the commonly used techniques in detecting low-level mosaicism in hPSC cultures. We have performed mixing experiments to mimic the naturally occurring mosaicism and have assessed the sensitivity of chromosome banding, qPCR, fluorescence in situ hybridization, and digital droplet PCR in detecting variants. Our analysis highlights the limits of mosaicism detection by the commonly employed methods, a pivotal requirement for interpreting the genetic status of hPSCs and for setting standards for safe applications of hPSCs in regenerative medicine.


Asunto(s)
Variación Genética , Mosaicismo , Células Madre Pluripotentes/metabolismo , Técnicas de Cultivo de Célula , Línea Celular , Cromosomas Humanos , Cromosomas Humanos Par 17 , Cromosomas Humanos Par 20 , Variaciones en el Número de Copia de ADN , Humanos , Hibridación Fluorescente in Situ , Cariotipo , Células Madre Pluripotentes/citología , Reacción en Cadena de la Polimerasa , Trisomía
18.
Sci Rep ; 5: 11694, 2015 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-26170169

RESUMEN

Maternal exposure during pregnancy to toxins can occasionally lead to miscarriage and malformation. It is currently thought that toxins pass through the placental barrier, albeit bi-layered in the first trimester, and damage the fetus directly, albeit at low concentration. Here we examined the responses of human embryonic stem (hES) cells in tissue culture to two metals at low concentration. We compared direct exposures with indirect exposures across a bi-layered model of the placenta cell barrier. Direct exposure caused increased DNA damage without apoptosis or a loss of cell number but with some evidence of altered differentiation. Indirect exposure caused increased DNA damage and apoptosis but without loss of pluripotency. This was not caused by metal ions passing through the barrier. Instead the hES cells responded to signalling molecules (including TNF-α) secreted by the barrier cells. This mechanism was dependent on connexin 43 mediated intercellular 'bystander signalling' both within and between the trophoblast barrier and the hES colonies. These results highlight key differences between direct and indirect exposure of hES cells across a trophoblast barrier to metal toxins. It offers a theoretical possibility that an indirectly mediated toxicity of hES cells might have biological relevance to fetal development.


Asunto(s)
Comunicación Celular , Células Madre Embrionarias Humanas/metabolismo , Transducción de Señal , Trofoblastos/metabolismo , Apoptosis/efectos de los fármacos , Apoptosis/genética , Puntos de Control del Ciclo Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Conexina 43/metabolismo , Citocinas/biosíntesis , Daño del ADN/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Uniones Comunicantes/metabolismo , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/efectos de los fármacos , Humanos , Metales Pesados/toxicidad , Trofoblastos/citología , Trofoblastos/efectos de los fármacos , Factor de Necrosis Tumoral alfa/biosíntesis
19.
PLoS One ; 10(4): e0123467, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25875838

RESUMEN

We have used single cell transcriptome analysis to re-examine the substates of early passage, karyotypically Normal, and late passage, karyotypically Abnormal ('Culture Adapted') human embryonic stem cells characterized by differential expression of the cell surface marker antigen, SSEA3. The results confirmed that culture adaptation is associated with alterations to the dynamics of the SSEA3(+) and SSEA3(-) substates of these cells, with SSEA3(-) Adapted cells remaining within the stem cell compartment whereas the SSEA3(-) Normal cells appear to have differentiated. However, the single cell data reveal that these substates are characterized by further heterogeneity that changes on culture adaptation. Notably the Adapted population includes cells with a transcriptome substate suggestive of a shift to a more naïve-like phenotype in contrast to the cells of the Normal population. Further, a subset of the Normal SSEA3(+) cells expresses genes typical of endoderm differentiation, despite also expressing the undifferentiated stem cell genes, POU5F1 (OCT4) and NANOG, whereas such apparently lineage-primed cells are absent from the Adapted population. These results suggest that the selective growth advantage gained by genetically variant, culture adapted human embryonic stem cells may derive in part from a changed substate structure that influences their propensity for differentiation.


Asunto(s)
Antígenos de Carbohidratos Asociados a Tumores/genética , Diferenciación Celular , Antígenos Embrionarios Específico de Estadio/genética , Antígenos de Carbohidratos Asociados a Tumores/metabolismo , Células Cultivadas , Análisis por Conglomerados , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Células Madre Embrionarias Humanas , Humanos , Proteína Homeótica Nanog , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Antígenos Embrionarios Específico de Estadio/metabolismo , Transcriptoma
20.
Stem Cells Dev ; 13(4): 325-36, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15345125

RESUMEN

Human embryonic stem (ES) cells offer substantial opportunities for providing well-defined differentiated cells for drug discovery, toxicology, and regenerative medicine, but the development of efficient techniques for their large-scale culture under defined conditions, and for controlling and directing their differentiation, presents a substantial challenge. Markers for defining the undifferentiated cells are well established, based upon previous studies of embryonal carcinoma (EC) cells, their malignant counterparts from teratocarcinomas. These provide valuable tools for monitoring human ES cultures and their state of differentiation. However, current culture techniques are suboptimal and involve the use of poorly defined culture media and the use of feeder cells. Over time, the cells may also acquire karyotypic changes, reflecting genetic selection and adaptation to in vitro culture conditions. Nevertheless, progress is being made. Originally, human ES cells were derived and maintained in medium containing fetal calf serum. They are now widely cultured in a proprietary serum-free formulation (serum replacement from Invitrogen Corp., Carlsbad, CA), and recently we have derived a new human ES line in this medium without fetal calf serum. Human fibroblasts can also be used to replace mouse embryo fibroblasts as feeder cells. We have now found it possible to culture a subline of human ES cells on Matrigel, or purified collagen type IV, laminin, and fibronectin, without feeders or feeder-conditioned medium. These cells nevertheless retain the features of undifferentiated human ES cells, including a capacity for differentiation. Although these cells also carried karyotypic changes, further research focused upon understanding the mechanisms that control self-renewal, apoptosis, and commitment to differentiation will facilitate the development of defined culture conditions that minimize genetic change and optimize the maintenance of the undifferentiated stem cells.


Asunto(s)
Células Madre/citología , Células Madre/fisiología , Antígenos de Superficie/análisis , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular , Línea Celular , Embrión de Mamíferos , Humanos , Células Madre/ultraestructura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA