Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Braz. j. med. biol. res ; 51(5): e7183, 2018. graf
Artículo en Inglés | LILACS | ID: biblio-889088

RESUMEN

Human pluripotent stem cells (hPSCs)/OP9 coculture system is a widely used hematopoietic differentiation approach. The limited understanding of this process leads to its low efficiency. Thus, we used single-cell qPCR to reveal the gene expression profiles of individual CD34+ cells from different stages of differentiation. According to the dynamic gene expression of hematopoietic transcription factors, we overexpressed specific hematopoietic transcription factors (Gata2, Lmo2, Etv2, ERG, and SCL) at an early stage of hematopoietic differentiation. After overexpression, we generated more CD34+ cells with normal expression level of CD43 and CD31, which are used to define various hematopoietic progenitors. Furthermore, these CD34+ cells possessed normal differentiation potency in colony-forming unit assays and normal gene expression profiles. In this study, we demonstrated that single-cell qPCR can provide guidance for optimization of hematopoietic differentiation and transient overexpression of selected hematopoietic transcription factors can enhance hematopoietic differentiation.


Asunto(s)
Humanos , Células Madre Hematopoyéticas/citología , Diferenciación Celular , Técnicas de Cocultivo/métodos , Células Madre Pluripotentes/citología , Fenotipo , Expresión Génica , Reacción en Cadena de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de la Célula Individual/métodos , Citometría de Flujo
2.
Biol. Res ; 50: 14, 2017. tab, graf
Artículo en Inglés | LILACS | ID: biblio-838965

RESUMEN

BACKGROUND: Kidney diseases are a global health problem. Currently, over 2 million people require dialysis or transplant which are associated with high morbidity and mortality; therefore, new researches focused on regenerative medicine have been developed, including the use of stem cells. RESULTS: In this research, we generate differentiated kidney cells (DKCs) from mouse pluripotent stem cells (mPSCs) analyzing their morphological, genetic, phenotypic, and spectroscopic characteristics along differentiation, highlighting that there are no reports of the use of Fourier transform infrared (FTIR) spectroscopy to characterize the directed differentiation of mPSCs to DKCs. The genetic and protein experiments proved the obtention of DKCs that passed through the chronological stages of embryonic kidney development. Regarding vibrational spectroscopy analysis by FTIR, bands related with biomolecules were shown on mPSCs and DKCs spectra, observing distinct differences between cell lineages and maturation stages. The second derivative of DKCs spectra showed changes in the protein bands compared to mPSCs. Finally, the principal components analysis obtained from FTIR spectra allowed to characterize chemical and structurally mPSCs and their differentiation process to DKCs in a rapid and non-invasive way. CONCLUSION: Our results indicated that we obtained DKCs from mPSCs, which passed through the chronological stages of embryonic kidney development. Moreover, FTIR spectroscopy resulted in a non-invasive, rapid and precise technic that together with principal component analysis allows to characterize chemical and structurally both kind of cells and also discriminate and determine different stages along the cell differentiation process.


Asunto(s)
Animales , Ratones , Diferenciación Celular/fisiología , Espectroscopía Infrarroja por Transformada de Fourier/métodos , Células Madre Pluripotentes/fisiología , Riñón/citología , Inmunohistoquímica , Expresión Génica , Células Cultivadas , Técnica del Anticuerpo Fluorescente , Análisis de Componente Principal , Células Madre Pluripotentes/citología , Reacción en Cadena en Tiempo Real de la Polimerasa
3.
Journal of Veterinary Science ; : 519-528, 2014.
Artículo en Inglés | WPRIM | ID: wpr-24548

RESUMEN

The present study was conducted to develop an effective method for establishment of porcine parthenogenetic embryonic stem cells (ppESCs) from parthenogenetically activated oocyte-derived blastocysts. The addition of 10% fetal bovine serum (FBS) to the medium on the 3rd day of oocyte culturing improved the development of blastocysts, attachment of inner cell masses (ICMs) onto feeder cells, and formation of primitive ppESC colonies. ICM attachment was further enhanced by basic fibroblast growth factor, stem cell factor, and leukemia inhibitory factor. From these attached ICMs, seven ppESC lines were established. ppESC pluripotency was verified by strong enzymatic alkaline phosphatase activity and the expression of pluripotent markers OCT3/4, Nanog, and SSEA4. Moreover, the ppESCs were induced to form an embryoid body and teratoma. Differentiation into three germ layers (ectoderm, mesoderm, and endoderm) was confirmed by the expression of specific markers for the layers and histological analysis. In conclusion, data from the present study suggested that our modified culture conditions using FBS and cytokines are highly useful for improving the generation of pluripotent ppESCs.


Asunto(s)
Animales , Blastocisto/citología , Técnicas de Cultivo de Célula/veterinaria , Diferenciación Celular , Citocinas/metabolismo , Células Madre Embrionarias/citología , Partenogénesis , Células Madre Pluripotentes/citología , Porcinos/fisiología
4.
Indian J Exp Biol ; 2013 Feb; 51(2): 107-115
Artículo en Inglés | IMSEAR | ID: sea-147573

RESUMEN

Skin stem cells are very important in cosmetics, pharmacological and regenerative medicine and burn cases. Foreskin samples surgically removed after circumcision from boys below 7 years were collected and primary epidermal cells were prepared by enzymatic and mechanical tituration method. Selecting CD133 (prominin-1) multipotent stem cell marker, enriched stem cells were analyzed by MACS using CD133 antibodies conjugated with magnetic beads. CD133 positive and negative cells with specific skin stem cells markers like - CD34 (Universal stem cells marker), CD29 (integrin beta-1) and CD49f (integrin alpha-6) immunophenotypes were screened and sorted in flowcytometer. Further the expression of four embryonic genes or transcription factors of pluripotent stem cells were analyzed for pluripotent character of sorted cells. It was found that skin stem cell markers associated with CD133 cells, differentially expressed CD34, CD29 and CD49f immunophenotyes on both positive and negative CD133 cells in FACS analysis. The embryonic stem cell markers (induced pluripotent stem cell markers) like Oct4, SOX2, Notch-2 and K19 genes were expressed in CD133 positive epidermal cells. It is therefore evident that foreskin derived epidermal stem cells showed pluripotent or multipotent nature. This finding opens up avenues for new uses of these stem cells for direct cell seeding in wound healing, surgical suturing and drug screening.


Asunto(s)
Antígenos CD/metabolismo , Biomarcadores/metabolismo , Linaje de la Célula/genética , Separación Celular , Supervivencia Celular/genética , Niño , Epidermis/citología , Citometría de Flujo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Glicoproteínas/metabolismo , Humanos , Inmunofenotipificación , Masculino , Péptidos/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Propidio/metabolismo , Piel/citología , Coloración y Etiquetado
5.
Experimental & Molecular Medicine ; : e56-2013.
Artículo en Inglés | WPRIM | ID: wpr-152453

RESUMEN

As the theory of stem cell plasticity was first proposed, we have explored an alternative hypothesis for this phenomenon: namely that adult bone marrow (BM) and umbilical cord blood (UCB) contain more developmentally primitive cells than hematopoietic stem cells (HSCs). In support of this notion, using multiparameter sorting we were able to isolate small Sca1+Lin-CD45- cells and CD133+Lin-CD45- cells from murine BM and human UCB, respectively, which were further enriched for the detection of various early developmental markers such as the SSEA antigen on the surface and the Oct4 and Nanog transcription factors in the nucleus. Similar populations of cells have been found in various organs by our team and others, including the heart, brain and gonads. Owing to their primitive cellular features, such as the high nuclear/cytoplasm ratio and the presence of euchromatin, they are called very small embryonic-like stem cells (VSELs). In the appropriate in vivo models, VSELs differentiate into long-term repopulating HSCs, mesenchymal stem cells (MSCs), lung epithelial cells, cardiomyocytes and gametes. In this review, we discuss the most recent data from our laboratory and other groups regarding the optimal isolation procedures and describe the updated molecular characteristics of VSELs.


Asunto(s)
Animales , Humanos , Linaje de la Célula , Separación Celular/métodos , Células Madre Embrionarias/citología , Células Madre Hematopoyéticas/citología , Células Madre Mesenquimatosas/citología , Células Madre Pluripotentes/citología
6.
Artículo en Inglés | IMSEAR | ID: sea-144143

RESUMEN

Stem cells of the dental pulp are a population of postnatal stem cells with multilineage differentiation potential. These cells are derived from the neural ectomesenchyme, similar to most craniofacial tissues, and specific niches in the pulp have been identified. Since the isolation of dental pulp stem cells (DPSC) and stem cells from exfoliating deciduous teeth (SHED), numerous studies have attempted to define and characterize these cells, and embryonic stem cell features have been reported in both DPSC and SHED. These cells have a vast repertoire of differentiation - osteogenic, odontogenic, myogenic, adipogenic, neurogenic, and melanocytic, and have even demonstrated transdifferentiation to corneal cells and islet cells of pancreas. The combined advantages of multipotency/pluripotency and the relative ease of access of pulp tissue for autologous use render DPSC/ SHED attractive options in regenerative dentistry and medicine. This review gives a bird's eye view of current knowledge with respect to stem cells from the dental pulp.


Asunto(s)
Pulpa Dental , Humanos , Células Madre Multipotentes/citología , Células Madre Pluripotentes/citología , Células Madre , Diente Primario/citología
7.
Arq. neuropsiquiatr ; 70(7): 540-546, July 2012. ilus
Artículo en Inglés | LILACS | ID: lil-642981

RESUMEN

Cell therapies, based on transplantation of immature cells, are being considered as a promising tool in the treatment of neurological disorders. Many efforts are being concentrated on the development of safe and effective stem cell lines. Nevertheless, the neurogenic potential of some cell lines, i.e., the ability to generate mature neurons either in vitro or in vivo, is largely unknown. Recent evidence indicate that this potential might be distinct among different cell lines, therefore limiting their broad use as replacement cells in the central nervous system. Here, we have reviewed the latest advancements regarding the electrophysiological maturation of stem cells, focusing our attention on fetal-derived-, embryonic-, and induced pluripotent stem cells. In summary, a large body of evidence supports the biological safety, high neurogenic potential, and in some diseases probable clinical efficiency related to fetal-derived cells. By contrast, reliable data regarding embryonic and induced pluripotent stem cells are still missing.


Terapias celulares, baseadas no transplante de células imaturas, têm sido consideradas ferramentas promissoras no tratamento de doenças neurológicas. Muitos esforços têm sido concentrados no desenvolvimento de linhas de células-tronco seguras e eficazes. No entanto, o potencial neurogênico de algumas linhagens celulares, ou seja, a habilidade de gerar neurônios maduros, in vitro ou in vivo, ainda é altamente desconhecida. Dados recentes sugerem que esse potencial é distinto entre diversos tipos celulares, o que limitaria o largo emprego como células restauradoras no sistema nervoso central. Neste relato, revisaram-se os avanços recentes relacionados à maturação eletrofisiológica de células-tronco, com foco em células derivadas de tecido fetal, células embrionárias e células pluripotentes induzidas. Em resumo, há evidências que apontam para segurança biológica de células fetais, com alto potencial neurogênico e, em se tratando de algumas doenças, provável eficiência clínica. Ao contrário, ainda não há dados confiáveis acerca de células embrionárias e pluripotentes induzidas.


Asunto(s)
Humanos , Células Madre Embrionarias/citología , Enfermedades del Sistema Nervioso/cirugía , Células-Madre Neurales/citología , Células Madre Pluripotentes/citología , Células Madre Embrionarias/trasplante , Células-Madre Neurales/trasplante , Células Madre Pluripotentes/trasplante
8.
Experimental & Molecular Medicine ; : 303-309, 2012.
Artículo en Inglés | WPRIM | ID: wpr-186639

RESUMEN

We previously reported the successful establishment of embryonic stem cell (ESC)-like multipotent spermatogonial stem cells (mSSCs) from neonatal mouse testis. Here, we examined the ability of mSSCs to differentiate into vascular endothelial cells and smooth muscle cells, and compared to that of mouse ESCs. We used real-time reverse transcriptase polymerase chain reaction and immunohistochemistry to examine gene expression profiles of mSSCs and ESCs during in vitro vascular differentiation. Both mSSCs and ESCs exhibited substantial increase in the expression of mesodermal markers, such as Brachyury, Flk1, Mesp1, Nkx2.5, and Islet1, and a decrease in the expression of pluripotency markers, such as Oct3/4 and Nanog during the early stage of differentiation. The mRNA levels of vascular endothelial (VE)-cadherin and CD31 gradually increased in both differentiated mSSCs and ESCs. VE-cadherin- or CD31-positive cells formed sprouting branch-like structures, as observed during embryonic vascular development. At the same time, vascular smooth muscle cell-specific markers, such as myocardin and alpha-smooth muscle actin (SMA), were also highly expressed in differentiated mSSCs and ESCs. Immunocytochemical analysis revealed that the differentiated cells expressed both alpha-SMA and SM22-alpha proteins, and exhibited the intracellular fibril structure typical of smooth muscle cells. Overall, our findings showed that mSSCs have similar vascular differentiation abilities to those of ESCs, suggesting that mSSCs may be an alternative source of autologous pluripotent stem cells for vascular regeneration.


Asunto(s)
Animales , Humanos , Masculino , Ratones , Animales Recién Nacidos , Biomarcadores/metabolismo , Diferenciación Celular/fisiología , Células Madre Embrionarias/citología , Células Endoteliales/citología , Expresión Génica , Perfilación de la Expresión Génica , Inmunohistoquímica , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/citología , Células Madre Pluripotentes/citología , Reacción en Cadena en Tiempo Real de la Polimerasa , Espermatogonias/citología , Testículo/citología
9.
Experimental & Molecular Medicine ; : 175-186, 2010.
Artículo en Inglés | WPRIM | ID: wpr-203595

RESUMEN

Recently, reactive oxygen species (ROS) have been studied as a regulator of differentiation into specific cell types in embryonic stem cells (ESCs). However, ROS role in human ESCs (hESCs) is unknown because mouse ESCs have been used mainly for most studies. Herein we suggest that ROS generation may play a critical role in differentiation of hESCs; ROS enhances differentiation of hESCs into bi-potent mesendodermal cell lineage via ROS-involved signaling pathways. In ROS-inducing conditions, expression of pluripotency markers (Oct4, Tra 1-60, Nanog, and Sox2) of hESCs was decreased, while expression of mesodermal and endodermal markers was increased. Moreover, these differentiation events of hESCs in ROS-inducing conditions were decreased by free radical scavenger treatment. hESC-derived embryoid bodies (EBs) also showed similar differentiation patterns by ROS induction. In ROS-related signaling pathway, some of the MAPKs family members in hESCs were also affected by ROS induction. p38 MAPK and AKT (protein kinases B, PKB) were inactivated significantly by buthionine sulfoximine (BSO) treatment. JNK and ERK phosphorylation levels were increased at early time of BSO treatment but not at late time point. Moreover, MAPKs family-specific inhibitors could prevent the mesendodermal differentiation of hESCs by ROS induction. Our results demonstrate that stemness and differentiation of hESCs can be regulated by environmental factors such as ROS.


Asunto(s)
Humanos , Biomarcadores/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular , Linaje de la Célula/efectos de los fármacos , Células Cultivadas , Regulación hacia Abajo/efectos de los fármacos , Embrión de Mamíferos/citología , Células Madre Embrionarias/citología , Endodermo/citología , Activación Enzimática/efectos de los fármacos , Depuradores de Radicales Libres/farmacología , Mesodermo/citología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Células Madre Pluripotentes/citología , Especies Reactivas de Oxígeno/metabolismo , Regulación hacia Arriba/efectos de los fármacos
10.
Int. j. morphol ; 27(4): 1325-1333, dic. 2009. ilus
Artículo en Inglés | LILACS | ID: lil-582091

RESUMEN

Adult stem cells are great promise to the future of regenerative therapy, and understanding of its embryonic origin permit the discrimination of stem cell sources. Embryonic stem cells derived from inner cell mass of blastocyst originate the primordial germ cells, and pericyte stem cell associated to vessels endothelium in yolk sac. Currently, it is being proposed that embryonic primordial germ cell could originate hematopoietic stem cells based on the detection of germ cell markers (SSEA-1/TEC-1, Oct-4 and Nanog) in stem cell harvested from fetal liver and bone marrow. However, different experimental evidence points at two separate differentiation routes toward primordial germ cells, and hematopoietic stem cell with the same embryonic origin. The expression of undifferentiated stem cell markers in umbilical cord and placental vessels, such CD34, CXCR4, c-kit and OCT4 demonstrates the intimate relation between pericyte stem cells, endothelium, haematopoiesis, and primordial germ cells, which all originate from embryonic stem cell from the inner cell mass epiblast.


Las células madre adultas son una gran promesa para el futuro de la terapia regenerativa, y la comprensión de su origen embrionario permite la discriminación de las fuentes de células madre. Las células madre embrionarias derivadas del macizo celular interno del blastocisto originan las células germinales primordiales, y células madre pericíticas asociadas al endotelio de los vasos del saco vitelino. En la actualidad, se propone que las células germinales primordiales embrionarias podrían originar a las células madre hematopoyéticas sobre la base de la detección de marcadores de células germinales (SSEA-1/TEC-1 oct-4 y Nanog) en células madre extraídas de hígado fetal y médula ósea. Sin embargo, diferentes evidencias experimentales apuntan hacia dos vías separadas de diferenciación en células germinales primordiales, y en células madre hematopoyéticas con el mismo origen embrionario. La expresión de marcadores de células madre no diferenciadas en el cordón umbilical y los vasos de la placenta, como CD34, CXCR4, c-kit y OcT4 demuestra la íntima relación entre las células madre pericíticas, el endotelio y las células germinales primordiales, las que se originan en células madre embrionarias a partir del epiblasto del macizo celular interno.


Asunto(s)
Células Germinativas/citología , Células Madre Embrionarias/citología , Células Madre Hematopoyéticas/citología , Células Madre Pluripotentes/citología , Células Germinativas/fisiología , Células Madre Embrionarias/fisiología , Células Madre Hematopoyéticas/fisiología , Células Madre Pluripotentes/fisiología , Diferenciación Celular/fisiología , Embrión de Mamíferos/citología , Cordón Umbilical
13.
Yonsei Medical Journal ; : 819-827, 2008.
Artículo en Inglés | WPRIM | ID: wpr-153694

RESUMEN

PURPOSE: Human embryonic stem cells (hESCs) can proliferate for a prolonged period and differentiate into cardiomyocytes in vitro. Recent studies used bone morphogenetic protein 2 (BMP2) to generate cardiomyocytes from hESCs, however, all those studies used early embryoid bodies (EBs) and did not retrieve cardiomyocytes with a high yield. In this study, we treated long-term cultured EBs with BMP2 in order to promote differentiation into cardiomyocytes from hESCs. MATERIALS AND METHODS: hESC lines, including SNUhES3 and SNUhES4, were used in this study. Undifferentiated hESC colonies were detached to form EBs and cultured for up to 30 days. These long-term cultured EBs were differentiated into cardiomyocytes in serum-containing media. In our protocol, BMP2 was applied for 5 days after attachment of EBs. Cardiac specific markers, beating of differentiated cells and electron microscopic (EM) ultrastructures were evaluated and analyzed. RESULTS: Compared to 10-day or 20-day EBs, 30-day EBs showed a higher expression level of cardiac specific markers, Nkx2.5 and a-myosin heavy chain (alphaMHC). Treatment of BMP2 increased expression of cardiac troponin (cTn) I and a-actinin when evaluated at 20 days after attachment of 30-day EBs. Beating of differentiated cells was observed from 7 to 20 days after attachment. Moreover, EM findings demonstrated fine structures such as Z bands in these differentiated cardiomyocytes. These long-term cultured EBs yielded cardiomyocytes with an efficiency of as high as 73.6% when assessed by FACS. CONCLUSION: We demonstrated that the use of long-term cultured EBs may enhance differentiation into cardiomyocytes from hESCs when treated with BMP2.


Asunto(s)
Humanos , Proteína Morfogenética Ósea 2/farmacología , Técnicas de Cultivo de Célula , Diferenciación Celular , Línea Celular , Proliferación Celular , Células Madre Embrionarias/citología , Miocitos Cardíacos/citología , Células Madre Pluripotentes/citología , Transducción de Señal
14.
Artículo en Inglés | IMSEAR | ID: sea-38243

RESUMEN

Cell therapy is the promising therapeutic tool for the next decade. "Regenerative Medicine" based on cell and tissue replacement therapy is proposed as a revolutionary approach to various chronic and incurable conditions. The first key step for successful cell therapy is the establishment of clinical grade human Embryonic Stem Cell (hESC) lines. This article provides a concise summary on conventional and novel methods for hESC line derivation. There is also discussion on progression, future direction and problems in hESC line development. In Thailand, more advance knowledge, skill, and technology are required to develop the first human embryonic stem cell line and step forward to make cell therapy a reality.


Asunto(s)
Blastómeros/citología , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular , Línea Celular , Células Cultivadas , Humanos , Células Madre Pluripotentes/citología , Trasplante de Células Madre , Tailandia
15.
Medicina (B.Aires) ; 64(6): 543-549, 2005. ilus
Artículo en Español | LILACS | ID: lil-444256

RESUMEN

Embryonic stem cells are a population of cells located in the blastocyst, committed to specific differentiation according to spatial and temporal factors such as age and place of final location. Despite the final fate of hematic cells, hemopoietic cells retain a relative degree of plasticity dependent on environmental factors. Mesenchymal cells are a well differentiated population of bone marrow derived non hemopoietic cells with totipotential properties. The medical interest of such totipotentiality rests in the potential of such cells to repair damaged tissues. Particularly neuronal differentiation from progenitors obtained from mesenchymae non hemopoietic cells offers a new possibility in the field of neural transplantation and tissue engineering to repair functional entities in the nervous system.


Las células troncales embrionarias son totipotentes y se encuentran en pequeño número en el blastocisto donde pueden expandirse en forma indiferenciada durante un corto tiempo y de acuerdoal sitio donde se alojan, ellas adquirirán determinados fenotipos de diferenciación. Las células hemopoyéticas troncales se caracterizan por poseer un gran potencial proliferativo, cuyo modelo de regulación es jerárquico. Ellas retienen, a lo largo de su existencia, un cierto grado de plasticidad, lo cual hace que se puedan diferenciar en distintos tipos de células o de tejidos no hemopoyéticos, de acuerdo al microambiente donde se encuentran o bien a la presencia de ciertos factores estimulantes. En trabajos recientes se han podido aislar, por adherencia al plástico, en cultivo in vitro de médula ósea, células no hemopoyéticas que fueron llamadas mesenquimales por su semejanza con el tejido mesenquimal del embrión. Estas células, in vitro, pueden ser inducidas hacia nuevas líneas celulares, que se diferenciarán en nuevos tejidos. Las expectativas del beneficio terapéutico del transplante de médula ósea como el de células mesenquimales en enfermedades no hemopoyéticas son grandes, porque al utilizar tejidos o células autólogas, se evitan los graves problemas del rechazo inmunológico. En el caso del tejido nervioso la problemática de una fuente adecuada de donantes hace el tema especialmente interesante.


Asunto(s)
Humanos , Diferenciación Celular , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Tejido Nervioso/trasplante , Trasplante de Médula Ósea , Células Madre Pluripotentes/citología
16.
Experimental & Molecular Medicine ; : 399-407, 2005.
Artículo en Inglés | WPRIM | ID: wpr-207080

RESUMEN

Human embryonic stem cells (hESCs) need feeder cells for their maintenance in an undifferentiated state. In conventional culture systems, mouse embryonic fibroblasts (MEFs) serve as feeder cells to maintain hESCs. However, the use of MEFs elevates the risk of transmitting mouse pathogens and thus limits the potential of hESCs in cell replacement therapy. Consequently, the use of human feeder cells would be an important step forward in this in vitro technology. To address this issue, we used fibroblast-like cells differentiated from the Miz-hES6 hESC line (Diff (Miz-hES6)) as feeder cells to support the in vitro growth of three hESC lines. Immunofluorescence microscopy and reverse transcription-PCR assessing the expression of undifferentiated hESC markers revealed all three hESC lines were maintained in an undifferentiated state. In vitro proliferation proceeded as efficiently as when the hESCs were cultured on MEFS. Moreover, karyotype analysis revealed the chromosomal normality of the hESC lines and the Diff (Miz-hES6) feeders themselves after even 50 passages. Furthermore, the hESC lines maintained their pluripotency since they remained capable of forming embryoid bodies (EBs) in vitro. Thus, hESC-derived fibroblast-like cells successfully support in vitro hESC propagation.


Asunto(s)
Humanos , Biomarcadores/análisis , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Estructuras Embrionarias/citología , Fibroblastos/citología , Cariotipificación , Células Madre Pluripotentes/citología , Células Madre/citología , Factores de Tiempo
17.
Journal of Veterinary Science ; : 87-96, 2005.
Artículo en Inglés | WPRIM | ID: wpr-184698

RESUMEN

The remarkable potential of embryonic stem (ES) cells is their ability to develop into many different cell types. ES cells make it possible to treat patients by transplanting specialized healthy cells derived from them to repair damaged and diseased cells or tissues, known as "stem cell therapy". However, the issue of immunocompatibility is one of considerable significance in ES cell transplantation. One approach to overcome transplant rejection of human ES (hES) cells is to derive hES cells from nuclear transfer of the patient's own cells. This concept is known as "therapeutic cloning". In this review, we describe the derivations of ES cells and cloned ES cells by somatic cell nuclear transfer, and their potential applications in transplantation medicine.


Asunto(s)
Animales , Humanos , Técnicas de Cultivo de Célula/métodos , Clonación de Organismos/métodos , Estructuras Embrionarias/citología , Técnicas de Cultivo de Embriones , Células Madre Pluripotentes/citología , Trasplante de Células Madre/métodos
18.
Experimental & Molecular Medicine ; : 36-44, 2005.
Artículo en Inglés | WPRIM | ID: wpr-18133

RESUMEN

Human embryonic stem (hES) cells are capable of differentiating into pluralistic cell types, however, spontaneous differentiation generally gives rise to a limited number of specific differentiated cell types and a large degree of cell heterogeneity. In an effort to increase the efficiency of specified hES cell differentiation, we performed a series of transient transfection of hES cells with EGFP expression vectors driven by different promoter systems, including human cellular polypeptide chain elongation factor 1 alpha (hEF1alpha), human cytomegalo-virus, and chicken beta-actin. All these promoters were found to lead reporter gene expression in undifferentiated hES cells, but very few drug-selectable transfectants were obtained and failed to maintain stable expression of the transgene with either chemical or electroporation methods. In an attempt to increase transfection efficiency and obtain stable transgene expression, differentiated hES cells expressing both mesodermal and ectodermal markers were derived using a defined medium. Differentiated hES cells were electroporated with a hEF1alpha promoter-driven EGFP or human noggin expression vector. Using RT-PCR, immunocytochemistry and fluorescence microscopy, the differentiated hES cells transfected with foreign genes were confirmed to retain stable gene and protein expression during prolonged culture. These results may provide a new tool for introducing exogenous genes readily into hES cells, thereby facilitating more directed differentiation into specific and homogenous cell populations.


Asunto(s)
Animales , Humanos , Actinas/genética , Proteínas Morfogenéticas Óseas/genética , Diferenciación Celular , Pollos , Citomegalovirus/genética , Sistemas de Liberación de Medicamentos , Estructuras Embrionarias/citología , Terapia Genética , Proteínas Fluorescentes Verdes/genética , Técnicas para Inmunoenzimas , Microscopía Fluorescente , Factor 1 de Elongación Peptídica/genética , Células Madre Pluripotentes/citología , Regiones Promotoras Genéticas/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transcripción Genética/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA