Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros











Intervalo de año de publicación
1.
Biomaterials ; 136: 1-11, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28505596

RESUMEN

Cell replacement therapies have broad biomedical potential; however, low cell survival and poor functional integration post-transplantation are major hurdles that hamper clinical benefit. For example, following striatal transplantation of midbrain dopaminergic (mDA) neurons for the treatment of Parkinson's disease (PD), only 1-5% of the neurons typically survive in preclinical models and in clinical trials. In general, resource-intensive generation and implantation of larger numbers of cells are used to compensate for the low post-transplantation cell-survival. Poor graft survival is often attributed to adverse biochemical, mechanical, and/or immunological stress that cells experience during and after implantation. To address these challenges, we developed a functionalized hyaluronic acid (HA)-based hydrogel for in vitro maturation and central nervous system (CNS) transplantation of human pluripotent stem cell (hPSC)-derived neural progenitors. Specifically, we functionalized the HA hydrogel with RGD and heparin (hep) via click-chemistry and tailored its stiffness to encourage neuronal maturation, survival, and long-term maintenance of the desired mDA phenotype. Importantly, ∼5 times more hydrogel-encapsulated mDA neurons survived after transplantation in the rat striatum, compared to unencapsulated neurons harvested from commonly used 2D surfaces. This engineered biomaterial may therefore increase the therapeutic potential and reduce the manufacturing burden for successful neuronal implantation.


Asunto(s)
Neuronas Dopaminérgicas/citología , Neuronas Dopaminérgicas/trasplante , Células Madre Embrionarias/citología , Ácido Hialurónico/química , Hidrogeles/química , Andamios del Tejido/química , Animales , Línea Celular , Supervivencia Celular , Células Cultivadas , Femenino , Heparina/química , Humanos , Mesencéfalo/citología , Células-Madre Neurales/citología , Células-Madre Neurales/trasplante , Neurogénesis , Oligopéptidos/química , Ratas Endogámicas F344
2.
Theranostics ; 6(3): 291-301, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26909106

RESUMEN

Stem cell therapies have enormous potential for treating many debilitating diseases, including heart failure, stroke and traumatic brain injury. For maximal efficacy, these therapies require targeted cell delivery to specific tissues followed by successful cell engraftment. However, targeted delivery remains an open challenge. As one example, it is common for intravenous deliveries of mesenchymal stem cells (MSCs) to become entrapped in lung microvasculature instead of the target tissue. Hence, a robust, quantitative imaging method would be essential for developing efficacious cell therapies. Here we show that Magnetic Particle Imaging (MPI), a novel technique that directly images iron-oxide nanoparticle-tagged cells, can longitudinally monitor and quantify MSC administration in vivo. MPI offers near-ideal image contrast, depth penetration, and robustness; these properties make MPI both ultra-sensitive and linearly quantitative. Here, we imaged, for the first time, the dynamic trafficking of intravenous MSC administrations using MPI. Our results indicate that labeled MSC injections are immediately entrapped in lung tissue and then clear to the liver within one day, whereas standard iron oxide particle (Resovist) injections are immediately taken up by liver and spleen. Longitudinal MPI-CT imaging also indicated a clearance half-life of MSC iron oxide labels in the liver at 4.6 days. Finally, our ex vivo MPI biodistribution measurements of iron in liver, spleen, heart, and lungs after injection showed excellent agreement (R(2) = 0.943) with measurements from induction coupled plasma spectrometry. These results demonstrate that MPI offers strong utility for noninvasively imaging and quantifying the systemic distribution of cell therapies and other therapeutic agents.


Asunto(s)
Diagnóstico por Imagen/métodos , Compuestos Férricos/análisis , Magnetismo , Trasplante de Células Madre Mesenquimatosas , Administración Intravenosa , Animales , Femenino , Humanos , Ratones , Nanopartículas/análisis , Ratas Endogámicas F344 , Coloración y Etiquetado , Distribución Tisular
3.
Development ; 142(10): 1885-92, 2015 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-25968319

RESUMEN

Neural stem cells (NSCs) are defined by their ability to self-renew and to differentiate into mature neuronal and glial cell types. NSCs are the subject of intense investigation, owing to their crucial roles in neural development and adult brain function and because they present potential targets for gene and cell replacement therapies following injury or disease. Approaches to specifically genetically perturb or modulate NSC function would be valuable for either motivation. Unfortunately, most gene delivery vectors are incapable of efficient or specific gene delivery to NSCs in vivo. Vectors based on adeno-associated virus (AAV) present a number of advantages and have proven increasingly successful in clinical trials. However, natural AAV variants are inefficient in transducing NSCs. We previously engineered a novel AAV variant (AAV r3.45) capable of efficient transduction of adult NSCs in vitro. Here, to build upon the initial promise of this variant, we investigated its in vitro and in vivo infectivity. AAV r3.45 was more selective for NSCs than mature neurons in a human embryonic stem cell-derived culture containing a mixture of cell types, including NSCs and neurons. It was capable of more efficient and selective transduction of rat and mouse NSCs in vivo than natural AAV serotypes following intracranial vector administration. Delivery of constitutively active ß-catenin yielded insights into mechanisms by which this key regulator modulates NSC function, indicating that this engineered AAV variant can be harnessed for preferential modulation of adult NSCs in the hippocampus. The capacity to rapidly genetically modify these cells might greatly accelerate in vivo investigations of adult neurogenesis.


Asunto(s)
Dependovirus/genética , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Animales , Línea Celular , Técnicas de Transferencia de Gen , Humanos , Ratones , Ratas , beta Catenina/metabolismo
4.
Mol Ther ; 20(2): 329-38, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22108859

RESUMEN

Efficient approaches for the precise genetic engineering of human pluripotent stem cells (hPSCs) can enhance both basic and applied stem cell research. Adeno- associated virus (AAV) vectors are of particular interest for their capacity to mediate efficient gene delivery to and gene targeting in various cells. However, natural AAV serotypes offer only modest transduction of human embryonic and induced pluripotent stem cells (hESCs and hiPSCs), which limits their utility for efficiently manipulating the hPSC genome. Directed evolution is a powerful means to generate viral vectors with novel capabilities, and we have applied this approach to create a novel AAV variant with high gene delivery efficiencies (~50%) to hPSCs, which are importantly accompanied by a considerable increase in gene-targeting frequencies, up to 0.12%. While this level is likely sufficient for numerous applications, we also show that the gene-targeting efficiency mediated by an evolved AAV variant can be further enhanced (>1%) in the presence of targeted double- stranded breaks (DSBs) generated by the co-delivery of artificial zinc finger nucleases (ZFNs). Thus, this study demonstrates that under appropriate selective pressures, AAV vectors can be created to mediate efficient gene targeting in hPSCs, alone or in the presence of ZFN- mediated double-stranded DNA breaks.


Asunto(s)
Dependovirus/genética , Evolución Molecular Dirigida , Marcación de Gen , Vectores Genéticos/genética , Células Madre Pluripotentes/metabolismo , Transducción Genética , Secuencia de Bases , Proteínas de la Cápside/genética , Línea Celular , Roturas del ADN de Doble Cadena , Dependovirus/fisiología , Endonucleasas/genética , Expresión Génica , Biblioteca de Genes , Orden Génico , Técnicas de Transferencia de Gen , Humanos , Datos de Secuencia Molecular , Mutación , Tropismo Viral , Dedos de Zinc/genética
5.
Mol Ther ; 19(4): 667-75, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21224831

RESUMEN

Gene delivery to, and gene targeting in, stem cells would be a highly enabling technology for basic science and biomedical application. Adeno-associated viral (AAV) vectors have demonstrated the capacity for efficient delivery to numerous cells, but their application to stem cells has been limited by low transduction efficiency. Due to their considerable advantages, however, engineering AAV delivery systems to enhance gene delivery to stem cells may have an impact in stem cell biology and therapy. Therefore, using several diverse AAV capsid libraries-including randomly mutagenized, DNA shuffled, and random peptide insertion variants-we applied directed evolution to create a "designer" AAV vector with enhanced delivery efficiency for neural stem cells (NSCs). A novel AAV variant, carrying an insertion of a selected peptide sequence on the surface of the threefold spike within the heparin-binding site, emerged from this evolution. Importantly, this evolved AAV variant mediated efficient gene delivery to rat, mouse, and human NSCs, as well as efficient gene targeting within adult NSCs, and it is thus promising for applications ranging from basic stem cell biology to clinical translation.


Asunto(s)
Dependovirus/genética , Células-Madre Neurales/virología , Animales , Células Cultivadas , Cromatografía , Femenino , Técnica del Anticuerpo Fluorescente , Técnicas de Transferencia de Gen , Reacción en Cadena de la Polimerasa , Ratas , Ratas Endogámicas F344
6.
Restor Neurol Neurosci ; 28(4): 589-603, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20714081

RESUMEN

The greatest therapeutic promise of human embryonic stem cells (hESC) is to generate specialized cells to replace damaged tissue in patients suffering from various degenerative diseases. However, the signaling mechanisms involved in lineage restriction of ESC to adopt various cellular phenotypes are still under investigation. Furthermore, for progression of hESC-based therapies towards clinical applications, appropriate culture conditions must be developed to generate genetically stable homogenous populations of cells, to hinder possible adverse effects following transplantation. Other critical challenges that must be addressed for successful cell implantation include problems related to survival and functional efficacy of the grafted cells. This review initially describes the derivation of hESC and focuses on recent advances in generation, characterization, and maintenance of these cells. We also give an overview of original and emerging differentiation strategies used to convert hESC to different cell types. Finally, we will discuss transplantation studies of hESC-derived cells with respect to safety and functional recovery.


Asunto(s)
Células Madre Embrionarias/fisiología , Diferenciación Celular , Linaje de la Célula , Células Cultivadas , Células Madre Embrionarias/trasplante , Humanos , Células Madre Pluripotentes/fisiología
7.
Restor Neurol Neurosci ; 27(4): 359-70, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19738328

RESUMEN

PURPOSE: Human embryonic stem cells (hESCs) which express a reporter gene consistently during all phases of differentiation would be valuable for basic research on cell transplantation. In this study, we describe karyotypically-abnormal variant hESCs, BGO1V2-EFG, which express hrGFP driven by the EF1 promoter. METHODS: BGO1V2-EFG cells were analyzed by using immunocytochemistry, single cell-based confocal image, and in vitro differentiation, including dopaminergic differentiation. RESULTS: Undifferentiated BGO1V2-EFG cells expressed pluripotent ESC markers and retained the ability to differentiate into cell types of all three germ layers. BGO1V2-EFG cells maintained stable and robust hrGFP expression in vitro in the undifferentiated state and during differentiation. The EF1 promoter retained activity during dopaminergic differentiation, as 76% of tyrosine hydroxlase (TH)-positive cells co-expressed hrGFP by confocal analysis. Treated with sodium butyrate (0.02 mM to 2.0 mM), an inhibitor of histone deacetylase (HDAC), during differentiation did not affect hrGFP expression, although TH expression was reduced by higher concentrations of sodium butyrate. CONCLUSION: BGO1V2-EFG cells maintain stable and robust hrGFP expression in the undifferentiated state and during neural differentiation. Especially, the EF1 promoter was effective in driving hrGFP expression during dopaminergic differentiation. BGO1V2-EFG cells may be useful for transplantation studies in Parkinson disease animal models.


Asunto(s)
Diferenciación Celular/fisiología , Dopamina/metabolismo , Células Madre Embrionarias/fisiología , Proteínas Fluorescentes Verdes/genética , Butiratos/farmacología , Diferenciación Celular/efectos de los fármacos , Línea Celular , Dopamina/genética , Relación Dosis-Respuesta a Droga , Células Madre Embrionarias/efectos de los fármacos , Inhibidores Enzimáticos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/genética , Humanos , Microscopía Confocal/métodos , Factor 1 de Elongación Peptídica/genética , Regiones Promotoras Genéticas , Transfección/métodos , Tirosina 3-Monooxigenasa/metabolismo
8.
PLoS One ; 3(1): e1422, 2008 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-18183302

RESUMEN

BACKGROUND: We initiated differentiation of human embryonic stem cells (hESCs) into dopamine neurons, obtained a purified population of neuronal precursor cells by cell sorting, and determined patterns of gene transcription. METHODOLOGY: Dopaminergic differentiation of hESCs was initiated by culturing hESCs with a feeder layer of PA6 cells. Differentiating cells were then sorted to obtain a pure population of PSA-NCAM-expressing neuronal precursors, which were then analyzed for gene expression using Massive Parallel Signature Sequencing (MPSS). Individual genes as well as regions of the genome which were activated were determined. PRINCIPAL FINDINGS: A number of genes known to be involved in the specification of dopaminergic neurons, including MSX1, CDKN1C, Pitx1 and Pitx2, as well as several novel genes not previously associated with dopaminergic differentiation, were expressed. Notably, we found that a specific region of the genome located on chromosome 11p15.5 was highly activated. This region contains several genes which have previously been associated with the function of dopaminergic neurons, including the gene for tyrosine hydroxylase (TH), the rate-limiting enzyme in catecholamine biosynthesis, IGF2, and CDKN1C, which cooperates with Nurr1 in directing the differentiation of dopaminergic neurons. Other genes in this region not previously recognized as being involved in the functions of dopaminergic neurons were also activated, including H19, TSSC4, and HBG2. IGF2 and CDKN1C were also found to be highly expressed in mature human TH-positive dopamine neurons isolated from human brain samples by laser capture. CONCLUSIONS: The present data suggest that the H19-IGF2 imprinting region on chromosome 11p15.5 is involved in the process through which undifferentiated cells are specified to become neuronal precursors and/or dopaminergic neurons.


Asunto(s)
Cromosomas Humanos Par 11 , Dopamina/metabolismo , Células Madre Embrionarias/citología , Perfilación de la Expresión Génica , Neuronas/citología , Diferenciación Celular/genética , Línea Celular , Separación Celular , Citometría de Flujo , Humanos , Inmunohistoquímica , Neuronas/metabolismo , Reacción en Cadena de la Polimerasa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA