Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 184
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Stem Cells ; 36(10): 1552-1566, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30004605

RESUMEN

Although autologous induced pluripotent stem cells (iPSCs) can potentially be useful for treating patients without immune rejection, in reality it will be extremely expensive and labor-intensive to make iPSCs to realize personalized medicine. An alternative approach is to make use of human leukocyte antigen (HLA) haplotype homozygous donors to provide HLA matched iPSC products to significant numbers of patients. To establish a haplobank of iPSCs, we repurposed the cord blood bank by screening ∼4,200 high resolution HLA typed cord blood samples, and selected those homozygous for the 10 most frequent HLA-A,-B,-DRB1 haplotypes in the Korean population. Following the generation of 10 iPSC lines, we conducted a comprehensive characterization, including morphology, expression of pluripotent markers and cell surface antigens, three-germ layer formation, vector clearance, mycoplasma/microbiological/viral contamination, endotoxin, and short tandem repeat (STR) assays. Various genomic analyses using microarray and comparative genomic hybridization (aCGH)-based single nucleotide polymorphism (SNP) and copy number variation (CNV) were also conducted. These 10 HLA-homozygous iPSC lines match 41.07% of the Korean population. Comparative analysis of HLA population data shows that they are also of use in other Asian populations, such as Japan, with some limited utility in ethnically diverse populations, such as the UK. Taken together, the generation of the 10 most frequent Korean HLA-homozygous iPSC lines serves as a useful pointer for the development of optimal methods for iPSC generation and quality control and indicates the benefits and limitations of collaborative HLA driven selection of donors for future stocking of worldwide iPSC haplobanks. Stem Cells 2018;36:1552-1566.


Asunto(s)
Almacenamiento de Sangre/métodos , Inestabilidad Genómica/genética , Antígenos HLA/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Haplotipos , Antígenos de Histocompatibilidad Clase II , Humanos
2.
Cytotherapy ; 20(6): 861-872, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29793831

RESUMEN

BACKGROUND AIMS: We have previously reported the generation of a current Good Manufacture Practice (cGMP)-compliant induced pluripotent stem cell (iPSC) line for clinical applications. Here we show that multiple cellular products currently being considered for therapy can be generated from a single master cell bank of this or any other clinically compliant iPSC line METHODS: Using a stock at passage 20 prepared from the cGMP-compliant working cell bank (WCB), we tested differentiation into therapeutically relevant cell types of the three germ layers using standardized but generic protocols. Cells that we generated include (i) neural stem cells, dopaminergic neurons and astrocytes; (ii) retinal cells (retinal pigment epithelium and photoreceptors); and (iii) hepatocyte, endothelial and mesenchymal cells. To confirm that these generic protocols can also be used for other iPSC lines, we tested the reproducibility of our methodology with a second clinically compliant line RESULTS: Our results confirmed that well-characterized iPSC lines have broad potency, and, despite allelic variability, the same protocols could be used with minimal modifications with multiple qualified lines. In addition, we introduced a constitutively expressed GFP cassette in Chr13 safe harbor site using a standardized previously described method and observed no significant difference in growth and differentiation between the engineered line and the control line indicating that engineered products can be made using a standardized methodology CONCLUSIONS: We believe that our demonstration that multiple products can be made from the same WCB and that the same protocols can be used with multiple lines offers a path to a cost-effective strategy for developing cellular products from iPSC lines.


Asunto(s)
Ingeniería Celular/métodos , Ingeniería Celular/normas , Linaje de la Célula , Adhesión a Directriz , Células Madre Pluripotentes Inducidas/citología , Astrocitos/citología , Astrocitos/fisiología , Diferenciación Celular , Línea Celular , Neuronas Dopaminérgicas/citología , Neuronas Dopaminérgicas/fisiología , Células Endoteliales/citología , Células Endoteliales/fisiología , Adhesión a Directriz/normas , Hepatocitos/citología , Hepatocitos/fisiología , Humanos , Células Madre Pluripotentes Inducidas/fisiología , Mesodermo/citología , Mesodermo/fisiología , Células-Madre Neurales/citología , Células-Madre Neurales/fisiología , Guías de Práctica Clínica como Asunto/normas , Estándares de Referencia , Reproducibilidad de los Resultados , Retina/citología , Bancos de Tejidos/normas
3.
Cytotherapy ; 17(6): 756-764, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25770678

RESUMEN

The discovery of induced pluripotent stem cells (iPSCs) and the rapid evolution of clinically compliant protocols to generate such lines from a variety of tissue sources has raised the possibility that personalized medicine may be achievable in the near future. Several strategies to deliver iPSCs for iPSC-derived cell-based therapy have been proposed: one such model has been the cell-banking model, using processes developed by the cord blood industry. The cord blood industry has evolved primarily as a banking model in which units of cord blood harvested from discarded placenta are stored either in a public or a private cord blood bank for future use. The consideration of a cord blood--like banking model has been further spurred by the realization that this population of cells is an ideal starting sample to generate pluripotent cells. Spurred by these technological advances, major efforts are underway to develop a current Good Manufacturing Practice--compliant protocol to generate iPSCs from cord blood and to develop a haplobanking strategy. In this article, we discuss the issues that may affect such an effort.


Asunto(s)
Bancos de Sangre , Sangre Fetal/citología , Células Madre Pluripotentes Inducidas/citología , Humanos , Trasplante de Células Madre
4.
Cytotherapy ; 17(9): 1169-77, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26276001

RESUMEN

Mesenchymal stem/stromal cells (MSCs) have been extensively investigated for their regenerative, immune-modulatory, and wound healing properties. While the laboratory studies have suggested that MSC's have a unique potential for modulating the etiopathology of multiple diseases, the results from clinical trials have not been encouraging or reproducible. One of the explanations for such variability is explained by the "art" of isolating and propagating MSCs. Therefore, establishing more than minimal criteria to define MSC would help understand best protocols to isolate, propagate and deliver MSCs. Developing a calibration standard, a database and a set of functional tests would be a better quality metric for MSCs. In this review, we discuss the importance of selecting a standard, issues associated with coming up with such a standard and how these issues can be mitigated.


Asunto(s)
Separación Celular/normas , Células Madre Mesenquimatosas/citología , Técnicas de Cultivo de Célula , Células Cultivadas , Humanos , Trasplante de Células Madre Mesenquimatosas/métodos , Estándares de Referencia
5.
Stem Cells ; 31(5): 941-52, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23341249

RESUMEN

Astrocytes can be generated from various tissue sources including human pluripotent stem cells (PSC). In this manuscript, we describe a chemically defined xeno-free medium culture system for rapidly generating astrocytes from neural stem cells derived from PSC. We show that astrocyte development in vitro, mimics normal development in vivo, and also passes through a CD44(+) astrocyte precursor stage. Astrocytes generated by our method display similar gene expression patterns, morphological characteristics and functional properties to primary astrocytes, and they survive and integrate after xenotransplantation. Whole genome expression profiling of astrocyte differentiation was performed at several time points of differentiation, and the results indicate the importance of known regulators and identify potential novel regulators and stage-specific lineage markers.


Asunto(s)
Astrocitos/citología , Células Madre Pluripotentes Inducidas/citología , Células-Madre Neurales/citología , Animales , Astrocitos/metabolismo , Diferenciación Celular/fisiología , Células Cultivadas , Criopreservación , Técnicas Citológicas/métodos , Expresión Génica , Perfilación de la Expresión Génica , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Células-Madre Neurales/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Trasplante Heterólogo
6.
Cytotherapy ; 16(9): 1305-12, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25065637

RESUMEN

BACKGROUND AIMS: We have previously reported a Good Manufacturing Practice (GMP)-compatible process for generating authentic dopaminergic neurons in defined media from human pluripotent stem cells and determined the time point at which dopaminergic precursors/neurons (day 14 after neuronal stem cell [NSC] stage) can be frozen, shipped and thawed without compromising their viability and ability to mature in vitro. One important issue we wished to address is whether dopaminergic precursors/neurons manufactured by our GMP-compatible process can be cryopreserved and engrafted in animal Parkinson disease (PD) models. METHODS: In this study, we evaluated the efficacy of freshly prepared and cryopreserved dopaminergic neurons in the 6-hydroxydopamine-lesioned rat PD model. RESULTS: We showed functional recovery up to 6 months post-transplantation in rats transplanted with our cells, whether freshly prepared or cryopreserved. In contrast, no motor improvement was observed in two control groups receiving either medium or cells at a slightly earlier stage (day 10 after NSC stage). Histologic analysis at the end point of the study (6 months post-transplantation) showed robust long-term survival of donor-derived tyrosine hydroxylase (TH)(+) dopaminergic neurons in rats transplanted with day 14 dopaminergic neurons. Moreover, TH(+) fibers emanated from the graft core into the surrounding host striatum. Consistent with the behavioral analysis, no or few TH(+) neurons were detected in animals receiving day 10 cells, although human cells were present in the graft. Importantly, no tumors were detected in any grafted rats, but long-term tumorigenic studies will need to determine the safety of our products. CONCLUSIONS: Dopaminergic neurons manufactured by a GMP-compatible process from human ESC survived and engrafted efficiently in the 6-OHDA PD rat model.


Asunto(s)
Criopreservación/métodos , Neuronas Dopaminérgicas/fisiología , Células-Madre Neurales/fisiología , Enfermedad de Parkinson/terapia , Trasplante de Células Madre , Animales , Biotecnología , Línea Celular , Supervivencia Celular , Cuerpo Estriado/patología , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/patología , Femenino , Humanos , Industria Manufacturera , Fibras Nerviosas/patología , Células-Madre Neurales/patología , Neurogénesis , Oxidopamina/administración & dosificación , Oxidopamina/efectos adversos , Enfermedad de Parkinson/etiología , Guías de Práctica Clínica como Asunto , Ratas , Ratas Sprague-Dawley , Recuperación de la Función , Trasplante Heterólogo
7.
Nature ; 455(7211): 401-5, 2008 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-18724358

RESUMEN

Stem cells are defined as self-renewing cell populations that can differentiate into multiple distinct cell types. However, hundreds of different human cell lines from embryonic, fetal and adult sources have been called stem cells, even though they range from pluripotent cells-typified by embryonic stem cells, which are capable of virtually unlimited proliferation and differentiation-to adult stem cell lines, which can generate a far more limited repertoire of differentiated cell types. The rapid increase in reports of new sources of stem cells and their anticipated value to regenerative medicine has highlighted the need for a general, reproducible method for classification of these cells. We report here the creation and analysis of a database of global gene expression profiles (which we call the 'stem cell matrix') that enables the classification of cultured human stem cells in the context of a wide variety of pluripotent, multipotent and differentiated cell types. Using an unsupervised clustering method to categorize a collection of approximately 150 cell samples, we discovered that pluripotent stem cell lines group together, whereas other cell types, including brain-derived neural stem cell lines, are very diverse. Using further bioinformatic analysis we uncovered a protein-protein network (PluriNet) that is shared by the pluripotent cells (embryonic stem cells, embryonal carcinomas and induced pluripotent cells). Analysis of published data showed that the PluriNet seems to be a common characteristic of pluripotent cells, including mouse embryonic stem and induced pluripotent cells and human oocytes. Our results offer a new strategy for classifying stem cells and support the idea that pluripotency and self-renewal are under tight control by specific molecular networks.


Asunto(s)
Perfilación de la Expresión Génica , Células Madre/clasificación , Células Madre/metabolismo , Algoritmos , Animales , Inteligencia Artificial , Diferenciación Celular , Línea Celular , Biología Computacional , Bases de Datos Factuales , Células Madre Embrionarias/clasificación , Células Madre Embrionarias/metabolismo , Humanos , Ratones , Células Madre Multipotentes/clasificación , Células Madre Multipotentes/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Oocitos/clasificación , Oocitos/metabolismo , Fenotipo , Células Madre Pluripotentes/clasificación , Células Madre Pluripotentes/metabolismo , Unión Proteica
8.
Cytotherapy ; 15(8): 999-1010, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23664011

RESUMEN

BACKGROUND AIMS: We have previously described a xeno-free scalable system to generate transplantable dopaminergic neurons from human pluripotent stem cells. However, several important questions remain to be answered about our cell therapy efforts. These include determining the exact time at which cells should be transplanted and whether cells at this stage can be frozen, shipped, thawed and injected without compromising their ability to mature and survive the transplantation procedure. We also needed to determine whether further optimization of the culture process could shorten the development time and reduce variability and whether a current Good Manufacture Practice (CGMP) facility could manufacture cells with fidelity. METHODS: We developed an optimized protocol that included modulating the sonic hedgehog homolog gradient with bone morphogenetic proteins (BMP2) and addition of activin to the culture medium, which shortened the time to generate Lmx1A and FoxA2 immunoreactive cells by 4-6 days. RESULTS: We showed that cells at this stage could be safely frozen and thawed while retaining an excellent ability to continue to mature in vitro and survive transplant in vivo. Importantly, we successfully adapted this process to a CGMP facility and manufactured two lots of transplant-ready dopaminergic neurons (>250 vials) under CGMP-compatible conditions. In vitro characterization, including viability/recovery on thawing, whole genome expression as well as expression of midbrain/dopaminergic markers, showed that the cells manufactured under GMP-compatible conditions were similar to cells produced at lab scale. CONCLUSIONS: Our results suggest that this optimized protocol can be used to generate dopaminergic neurons for Investigational New Drug enabling studies.


Asunto(s)
Técnicas de Cultivo de Célula , Neuronas Dopaminérgicas/citología , Neuronas Dopaminérgicas/trasplante , Células Madre Pluripotentes Inducidas/metabolismo , Neurogénesis , Células Madre Pluripotentes/citología , Activinas , Animales , Proteína Morfogenética Ósea 2/metabolismo , Proliferación Celular , Supervivencia Celular , Tratamiento Basado en Trasplante de Células y Tejidos , Células Cultivadas , Criopreservación/métodos , Dopamina/análisis , Dopamina/biosíntesis , Descubrimiento de Drogas/métodos , Proteínas Hedgehog/metabolismo , Factor Nuclear 3-beta del Hepatocito/inmunología , Humanos , Proteínas con Homeodominio LIM/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Factores de Transcripción/inmunología
9.
J Cell Biochem ; 113(10): 3061-8, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22573568

RESUMEN

The discovery of the ability to induce somatic cells to a pluripotent state through the overexpression of specific transcription factors has the potential to transform the ways in which pharmaceutical agents and cellular transplantation therapies are developed. Proper utilization of the technology to generate induced pluripotent stem cells (iPSCs) requires that researchers select the appropriate reprogramming method for generating iPSCs so that the resulting iPSCs can be transitioned towards clinical applications effectively. This article reviews all of the currently available reprogramming techniques with a focus on critiquing them on the basis of their utility in translational medicine.


Asunto(s)
Diferenciación Celular , Ingeniería Celular/métodos , Células Madre Pluripotentes Inducidas/citología , Investigación Biomédica Traslacional/métodos , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Lentivirus/genética , Lentivirus/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Plásmidos/genética , Plásmidos/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Medicina Regenerativa , Reproducibilidad de los Resultados , Transfección , Transgenes
10.
J Neurosci Res ; 90(7): 1367-81, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22535492

RESUMEN

Human embryonic stem cell (hESC)-derived dopaminergic (DA) neurons hold potential for treating Parkinson's disease (PD) through cell replacement therapy. Generation of DA neurons from hESCs has been achieved by coculture with the stromal cell line PA6, a source of stromal cell-derived inducing activity (SDIA). However, the factors produced by stromal cells that result in SDIA are largely undefined. We previously reported that medium conditioned by PA6 cells can generate functional DA neurons from NTera2 human embryonal carcinoma stem cells. Here we show that PA6-conditioned medium can induce DA neuronal differentiation in both NTera2 cells and the hESC I6 cell line. To identify the factor(s) responsible for SDIA, we used large-scale microarray analysis of gene expression combined with mass spectrometric analysis of PA6-conditioned medium (CM). The candidate factors, hepatocyte growth factor (HGF), stromal cell-derived factor-1 α (SDF1α), secreted frizzled-related protein 1 (sFRP1), and vascular endothelial growth factor D (VEGFD) were identified, and their concentrations in PA6 CM were established by immunoaffinity capillary electrophoresis. Upon addition of SDF1α, sFRP1, and VEGFD to the culture medium, we observed an increase in the number of cells expressing tyrosine hydroxylase (a marker for DA neurons) and ßIII-tubulin (a marker for immature neurons) in both the NTera2 and I6 cell lines. These results indicate that SDF1α, sFRP1, and VEGFD are major components of SDIA and suggest the potential use of these defined factors to elicit DA differentiation of pluripotent human stem cells for therapeutic intervention in PD.


Asunto(s)
Neuronas Dopaminérgicas/citología , Factores de Crecimiento Nervioso/biosíntesis , Células-Madre Neurales/efectos de los fármacos , Neurogénesis/fisiología , Células Madre Pluripotentes/efectos de los fármacos , Animales , Biomarcadores/metabolismo , Técnicas de Cultivo de Célula , Línea Celular , Línea Celular Tumoral , Quimiocina CXCL12/biosíntesis , Quimiocina CXCL12/fisiología , Neuronas Dopaminérgicas/metabolismo , Células Madre de Carcinoma Embrionario/citología , Células Madre de Carcinoma Embrionario/efectos de los fármacos , Células Madre de Carcinoma Embrionario/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular/farmacología , Péptidos y Proteínas de Señalización Intercelular/fisiología , Proteínas de la Membrana/farmacología , Proteínas de la Membrana/fisiología , Ratones , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Células del Estroma/metabolismo , Tubulina (Proteína)/biosíntesis , Tirosina 3-Monooxigenasa/biosíntesis , Factor D de Crecimiento Endotelial Vascular/biosíntesis , Factor D de Crecimiento Endotelial Vascular/fisiología
11.
Stem Cells ; 29(7): 1013-20, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21544901

RESUMEN

The reprogramming of somatic cells to a pluripotent state by the expression of a defined set of exogenous transcription factors represents a significant breakthrough for the use of stem cells in regenerative medicine. It has the potential to make autologous stem cell therapy practical and promote better understanding of the disease processes by generating patient specific stem cells. Several strategies have been used to generate induced pluripotent stem cells (iPSCs) that include nucleic acid and non-nucleic acid-based approaches, with and without epigenetic modifications. The purpose of these different approaches for generating iPSCs, besides understanding the underlying mechanism, is to develop a facile method for reprogramming without genetic alteration, suitable for clinical use. Here, we discuss different strategies for generating iPSCs, with an emphasis on a recent non-cell autonomous approach to reprogram somatic progenitors that regenerate cornea to a pluripotent state through the recruitment of endogenous transcription factors.


Asunto(s)
Reprogramación Celular , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes/citología , Células Madre Adultas/citología , Células Madre Adultas/metabolismo , Animales , Humanos , Ácidos Nucleicos/genética , Ácidos Nucleicos/metabolismo , Células Madre Pluripotentes/metabolismo
12.
Cytotherapy ; 14(8): 902-16, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22731756

RESUMEN

Critical limb ischemia (CLI) is a syndrome manifested by ischemic rest pain, non-healing ulcers and tissue loss. CLI patients are at very high risk of amputation and experience poor physical function, leading to severe morbidity and mortality. The fundamental goal for CLI treatment is to relieve ischemic rest pain, heal ulcers, prevent limb loss and improve the quality of life, thereby extending the survival of the patient. Surgical or endovascular revascularization aimed at increasing blood flow is currently available for limb salvage in CLI. However, up to 30% of CLI patients are not suitable for such interventions because of high operative risk or unfavorable vascular anatomy. Therefore exploring new and more effective strategies for revascularization of ischemic limbs is imperative for the establishment of a viable therapeutic alternative. With the emergence of new approaches, this review describes up-to-date progress and developments in cell-based therapy as a novel and promising alternative for CLI treatment. Preliminary clinical data have established the safety, feasibility and efficacy of stem cells, and numerous studies are underway to consolidate this evidence further. However, significant hurdles remain to be addressed before this research can be responsibly translated to the bedside. In particular, we need better understanding of the behavior of cells post-transplantation and to learn how to control their survival and migration proliferation/differentiation in the hostile pathologic environment. Future research should focus on methods of isolation, optimal dosage, appropriate cell type, route of administration, role of tissue-derived factors and supportive endogenous stimulation.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos , Extremidades/fisiopatología , Isquemia/terapia , Células Madre , Diferenciación Celular , Proliferación Celular , Extremidades/irrigación sanguínea , Humanos , Isquemia/fisiopatología , Neovascularización Fisiológica , Trasplante de Células Madre , Células Madre/clasificación , Células Madre/citología
13.
PLoS Biol ; 7(8): e1000176, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19688041

RESUMEN

During embryogenesis, the neural stem cells (NSC) of the developing cerebral cortex are located in the ventricular zone (VZ) lining the cerebral ventricles. They exhibit apical and basal processes that contact the ventricular surface and the pial basement membrane, respectively. This unique architecture is important for VZ physical integrity and fate determination of NSC daughter cells. In addition, the shorter apical process is critical for interkinetic nuclear migration (INM), which enables VZ cell mitoses at the ventricular surface. Despite their importance, the mechanisms required for NSC adhesion to the ventricle are poorly understood. We have shown previously that one class of candidate adhesion molecules, laminins, are present in the ventricular region and that their integrin receptors are expressed by NSC. However, prior studies only demonstrate a role for their interaction in the attachment of the basal process to the overlying pial basement membrane. Here we use antibody-blocking and genetic experiments to reveal an additional and novel requirement for laminin/integrin interactions in apical process adhesion and NSC regulation. Transient abrogation of integrin binding and signalling using blocking antibodies to specifically target the ventricular region in utero results in abnormal INM and alterations in the orientation of NSC divisions. We found that these defects were also observed in laminin alpha2 deficient mice. More detailed analyses using a multidisciplinary approach to analyse stem cell behaviour by expression of fluorescent transgenes and multiphoton time-lapse imaging revealed that the transient embryonic disruption of laminin/integrin signalling at the VZ surface resulted in apical process detachment from the ventricular surface, dystrophic radial glia fibers, and substantial layering defects in the postnatal neocortex. Collectively, these data reveal novel roles for the laminin/integrin interaction in anchoring embryonic NSCs to the ventricular surface and maintaining the physical integrity of the neocortical niche, with even transient perturbations resulting in long-lasting cortical defects.


Asunto(s)
Ventrículos Cerebrales , Regulación del Desarrollo de la Expresión Génica , Cadenas beta de Integrinas/metabolismo , Neocórtex/embriología , Transducción de Señal , Células Madre/citología , Animales , Adhesión Celular , Diferenciación Celular , Ventrículos Cerebrales/citología , Ventrículos Cerebrales/embriología , Ventrículos Cerebrales/fisiología , Embrión de Mamíferos , Procesamiento de Imagen Asistido por Computador , Cadenas beta de Integrinas/genética , Laminina/genética , Laminina/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Neocórtex/citología , Neocórtex/metabolismo , Neuronas/citología , Neuronas/metabolismo
14.
Nat Med ; 11(9): 966-72, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16086023

RESUMEN

Demyelination is the hallmark of numerous neurodegenerative conditions, including multiple sclerosis. Oligodendrocyte progenitors (OPCs), which normally mature into myelin-forming oligodendrocytes, are typically present around demyelinated lesions but do not remyelinate affected axons. Here, we find that the glycosaminoglycan hyaluronan accumulates in demyelinated lesions from individuals with multiple sclerosis and in mice with experimental autoimmune encephalomyelitis. A high molecular weight (HMW) form of hyaluronan synthesized by astrocytes accumulates in chronic demyelinated lesions. This form of hyaluronan inhibits remyelination after lysolecithin-induced white matter demyelination. OPCs accrue and do not mature into myelin-forming cells in demyelinating lesions where HMW hyaluronan is present. Furthermore, the addition of HMW hyaluronan to OPC cultures reversibly inhibits progenitor-cell maturation, whereas degrading hyaluronan in astrocyte-OPC cocultures promotes oligodendrocyte maturation. HMW hyaluronan may therefore contribute substantially to remyelination failure by preventing the maturation of OPCs that are recruited to demyelinating lesions.


Asunto(s)
Astrocitos/fisiología , Enfermedades Desmielinizantes/fisiopatología , Receptores de Hialuranos/metabolismo , Ácido Hialurónico/metabolismo , Oligodendroglía/fisiología , Animales , Encefalomielitis Autoinmune Experimental/fisiopatología , Humanos , Ratones , Esclerosis Múltiple/fisiopatología , Células Madre/fisiología
15.
Stem Cells ; 28(10): 1893-904, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20715183

RESUMEN

Human induced pluripotent stem cells (iPSCs) reprogrammed from somatic cells represent a promising unlimited cell source for generating patient-specific cells for biomedical research and personalized medicine. As a first step, critical to clinical applications, we attempted to develop defined culture conditions to expand and differentiate human iPSCs into functional progeny such as dopaminergic neurons for treating or modeling Parkinson's disease (PD). We used a completely defined (xeno-free) system that we previously developed for efficient generation of authentic dopaminergic neurons from human embryonic stem cells (hESCs), and applied it to iPSCs. First, we adapted two human iPSC lines derived from different somatic cell types for the defined expansion medium and showed that the iPSCs grew similarly as hESCs in the same medium regarding pluripotency and genomic stability. Second, by using these two independent adapted iPSC lines, we showed that the process of differentiation into committed neural stem cells (NSCs) and subsequently into dopaminergic neurons was also similar to hESCs. Importantly, iPSC-derived dopaminergic neurons were functional as they survived and improved behavioral deficits in 6-hydroxydopamine-leasioned rats after transplantation. In addition, iPSC-derived NSCs and neurons could be efficiently transduced by a baculoviral vector delivering episomal DNA for future gene function study and disease modeling using iPSCs. We also performed genome-wide microarray comparisons between iPSCs and hESCs, and we derived NSC and dopaminergic neurons. Our data revealed overall similarity and visible differences at a molecular level. Efficient generation of functional dopaminergic neurons under defined conditions will facilitate research and applications using PD patient-specific iPSCs.


Asunto(s)
Dopamina/metabolismo , Células Madre Pluripotentes Inducidas/citología , Neuronas/citología , Neuronas/metabolismo , Diferenciación Celular , Línea Celular , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Humanos , Inmunohistoquímica , Células Madre Pluripotentes Inducidas/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa
16.
Blood ; 112(2): 295-307, 2008 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-18332228

RESUMEN

We compared the transcriptomes of marrow-derived mesenchymal stem cells (MSCs) with differentiated adipocytes, osteocytes, and chondrocytes derived from these MSCs. Using global gene-expression profiling arrays to detect RNA transcripts, we have identified markers that are specific for MSCs and their differentiated progeny. Further, we have also identified pathways that MSCs use to differentiate into adipogenic, chondrogenic, and osteogenic lineages. We identified activin-mediated transforming growth factor (TGF)-beta signaling, platelet-derived growth factor (PDGF) signaling and fibroblast growth factor (FGF) signaling as the key pathways involved in MSC differentiation. The differentiation of MSCs into these lineages is affected when these pathways are perturbed by inhibitors of cell surface receptor function. Since growth and differentiation are tightly linked processes, we also examined the importance of these 3 pathways in MSC growth. These 3 pathways were necessary and sufficient for MSC growth. Inhibiting any of these pathways slowed MSC growth, whereas a combination of TGF-beta, PDGF, and beta-FGF was sufficient to grow MSCs in a serum-free medium up to 5 passages. Thus, this study illustrates it is possible to predict signaling pathways active in cellular differentiation and growth using microarray data and experimentally verify these predictions.


Asunto(s)
Perfilación de la Expresión Génica , Péptidos y Proteínas de Señalización Intercelular/fisiología , Células Madre Mesenquimatosas/citología , Transducción de Señal , Adipocitos/citología , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Condrocitos/citología , Factores de Crecimiento de Fibroblastos/fisiología , Humanos , Osteoblastos/citología , Factor de Crecimiento Derivado de Plaquetas/fisiología , Factor de Crecimiento Transformador beta/fisiología
17.
Stem Cells ; 27(8): 1836-46, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19544414

RESUMEN

In this study, we targeted Olig2, a basic helix-loop-helix transcription factor that plays an important role in motoneuron and oligodendrocyte development, in human embryonic stem cell (hESC) line BG01 by homologous recombination. One allele of Olig2 locus was replaced by a green fluorescent protein (GFP) cassette with a targeting efficiency of 5.7%. Targeted clone R-Olig2 (like the other clones) retained pluripotency, typical hESC morphology, and a normal parental karyotype 46,XY. Most importantly, GFP expression recapitulated endogenous Olig2 expression when R-Olig2 was induced by sonic hedgehog and retinoic acid, and GFP-positive cells could be purified by fluorescence-activated cell sorting. Consistent with previous reports on rodents, early GFP-expressing cells appeared biased to a neuronal fate, whereas late GFP-expressing cells appeared biased to an oligodendrocytic fate. This was corroborated by myoblast coculture, transplantation into the rat spinal cords, and whole genome expression profiling. The present work reports an hESC reporter line generated by homologous recombination targeting a neural lineage-specific gene, which can be differentiated and sorted to obtain pure neural progenitor populations.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Células Madre Embrionarias/fisiología , Proteínas del Tejido Nervioso/genética , Neuroglía/fisiología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Línea Celular , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Perfilación de la Expresión Génica , Técnicas de Sustitución del Gen , Marcación de Gen , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Humanos , Inmunohistoquímica , Masculino , Proteínas del Tejido Nervioso/metabolismo , Neuroglía/citología , Neuroglía/metabolismo , Factor de Transcripción 2 de los Oligodendrocitos , Ratas , Ratas Sprague-Dawley , Recombinación Genética , Transfección
18.
BMC Cell Biol ; 10: 44, 2009 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-19500347

RESUMEN

BACKGROUND: A unique and essential property of embryonic stem cells is the ability to self-renew and differentiate into multiple cell lineages. However, the possible differences in proliferation and differentiation capabilities among independently-derived human embryonic stem cells (hESCs) are not well known because of insufficient characterization. To address this question, a side-by-side comparison of 1) the ability to maintain an undifferentiated state and to self-renew under standard conditions; 2) the ability to spontaneously differentiate into three primary embryonic germ lineages in differentiating embryoid bodies; and 3) the responses to directed neural differentiation was made between three NIH registered hES cell lines I3 (TE03), I6 (TE06) and BG01V. Lines I3 and I6 possess normal XX and a normal XY karyotype while BG01V is a variant cell line with an abnormal karyotype derived from the karyotypically normal cell line BG01. RESULTS: Using immunocytochemistry, flow cytometry, qRT-PCR and MPSS, we found that all three cell lines actively proliferated and expressed similar "stemness" markers including transcription factors POU5F1/Oct3/4 and NANOG, glycolipids SSEA4 and TRA-1-81, and alkaline phosphatase activity. All cell lines differentiated into three embryonic germ lineages in embryoid bodies and into neural cell lineages when cultured in neural differentiation medium. However, a profound variation in colony morphology, growth rate, BrdU incorporation, and relative abundance of gene expression in undifferentiated and differentiated states of the cell lines was observed. Undifferentiated I3 cells grew significantly slower but their differentiation potential was greater than I6 and BG01V. Under the same neural differentiation-promoting conditions, the ability of each cell line to differentiate into neural progenitors varied. CONCLUSION: Our comparative analysis provides further evidence for similarities and differences between three hESC lines in self-renewal, and spontaneous and directed differentiation. These differences may be associated with inherited variation in the sex, stage, quality and genetic background of embryos used for hESC line derivation, and/or changes acquired during passaging in culture.


Asunto(s)
Línea Celular , Células Madre Embrionarias/citología , Células Madre Embrionarias/fisiología , Fosfatasa Alcalina/metabolismo , Antígenos de Superficie/metabolismo , Biomarcadores/metabolismo , Bromodesoxiuridina/metabolismo , Técnicas de Cultivo de Célula/normas , Diferenciación Celular , Proliferación Celular , Citometría de Flujo , Proteínas de Homeodominio/metabolismo , Humanos , Inmunohistoquímica , Proteína Homeótica Nanog , Neuroglía/citología , Neuroglía/metabolismo , Neuronas/citología , Neuronas/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Células Madre Pluripotentes/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Antígenos Embrionarios Específico de Estadio/metabolismo
19.
Stem Cells ; 26(1): 119-26, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17962703

RESUMEN

It has previously been shown that the phage-derived phiC31 integrase can efficiently target native pseudo-attachment sites in the genome of various species in cultured cells, as well as in vivo. To demonstrate its utility in human embryonic stem cells (hESC), we have created hESC-derived clones containing expression constructs. Variant human embryonic stem cell lines BG01v and SA002 were used to derive lines expressing a green fluorescent protein (GFP) marker under control of either the human Oct4 promoter or the EF1alpha promoter. Stable clones were selected by antibiotic resistance and further characterized. The frequency of integration suggested candidate hot spots in the genome, which were mapped using a plasmid rescue strategy. The pseudo-attP profile in hESC differed from those reported earlier in differentiated cells. Clones derived using this method retained the ability to differentiate into all three germ layers, and fidelity of expression of GFP was verified in differentiation assays. GFP expression driven by the Oct4 promoter recapitulated endogenous Oct4 expression, whereas persistent stable expression of GFP expression driven by the EF1alpha promoter was seen. Our results demonstrate the utility of phiC31 integrase to target pseudo-attP sites in hESC and show that integrase-mediated site-specific integration can efficiently create stably expressing engineered human embryonic stem cell clones.


Asunto(s)
Células Madre Embrionarias/fisiología , Técnicas de Transferencia de Gen , Ingeniería Genética/métodos , Integrasas/metabolismo , Sitios de Ligazón Microbiológica/genética , Bacteriófagos , Diferenciación Celular/fisiología , Línea Celular , Clonación Molecular , Células Madre Embrionarias/citología , Vectores Genéticos , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 1 de Elongación Peptídica/genética , Plásmidos/genética , Células Madre Pluripotentes/fisiología , Regiones Promotoras Genéticas , Transfección
20.
Methods Mol Biol ; 549: 3-16, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19378192

RESUMEN

Recent results have raised important questions on our ability to amplify stem cell populations in sufficient numbers as to be useful for therapy. Several reports have indicated that human stem cell populations harvested from the adult have low or undetectable telomerase levels, age in culture, and may not be propagated indefinitely. Other groups have shown that stem cells age and as such, their properties will have changed depending on the age of the individual from which they are harvested, and the time for which they are propagated in culture. Other groups have shown that cells maintained in culture may undergo alterations as they are propagated, and that these alterations may alter the predicted behavior of stem cells. Yet others have shown that human cells differ from their counterparts in other species in significant ways and have identified important difficulties in assessing cells in a xeno environment. Clinical colleagues have identified issues of variability and difficulties in the long-term follow-up that is being requested. Researchers in the stem cell field focused on translational work need to develop a practical plan that takes into account such difficulties while developing manufacturing protocols, designing animal studies, or developing trial protocols. Such proactive planning will be critical in ensuring a successful transition from the bench to the clinic.


Asunto(s)
Neuronas , Trasplante de Células Madre , Adulto , Animales , Diferenciación Celular , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Ensayos Clínicos Fase I como Asunto , Células Madre Embrionarias/fisiología , Humanos , Neuronas/citología , Neuronas/fisiología , Selección de Paciente , Roedores , Trasplante de Células Madre/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA