Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 444(7118): 481-5, 2006 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-16929302

RESUMEN

The derivation of human embryonic stem (hES) cells currently requires the destruction of ex utero embryos. A previous study in mice indicates that it might be possible to generate embryonic stem (ES) cells using a single-cell biopsy similar to that used in preimplantation genetic diagnosis (PGD), which does not interfere with the embryo's developmental potential. By growing the single blastomere overnight, the resulting cells could be used for both genetic testing and stem cell derivation without affecting the clinical outcome of the procedure. Here we report a series of ten separate experiments demonstrating that hES cells can be derived from single blastomeres. In this proof-of-principle study, multiple biopsies were taken from each embryo using micromanipulation techniques and none of the biopsied embryos were allowed to develop in culture. Nineteen ES-cell-like outgrowths and two stable hES cell lines were obtained. The latter hES cell lines maintained undifferentiated proliferation for more than eight months, and showed normal karyotype and expression of markers of pluripotency, including Oct-4, SSEA-3, SSEA-4, TRA-1-60, TRA-1-81, nanog and alkaline phosphatase. These cells retained the potential to form derivatives of all three embryonic germ layers both in vitro and in teratomas. The ability to create new stem cell lines and therapies without destroying embryos would address the ethical concerns of many, and allow the generation of matched tissue for children and siblings born from transferred PGD embryos.


Asunto(s)
Blastómeros/citología , Línea Celular , Células Madre Embrionarias/citología , Diferenciación Celular , Técnicas de Cocultivo , Humanos
2.
Nature ; 439(7073): 216-9, 2006 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-16227970

RESUMEN

The most basic objection to human embryonic stem (ES) cell research is rooted in the fact that ES cell derivation deprives embryos of any further potential to develop into a complete human being. ES cell lines are conventionally isolated from the inner cell mass of blastocysts and, in a few instances, from cleavage stage embryos. So far, there have been no reports in the literature of stem cell lines derived using an approach that does not require embryo destruction. Here we report an alternative method of establishing ES cell lines-using a technique of single-cell embryo biopsy similar to that used in pre-implantation genetic diagnosis of genetic defects-that does not interfere with the developmental potential of embryos. Five putative ES and seven trophoblast stem (TS) cell lines were produced from single blastomeres, which maintained normal karyotype and markers of pluripotency or TS cells for up to more than 50 passages. The ES cells differentiated into derivatives of all three germ layers in vitro and in teratomas, and showed germ line transmission. Single-blastomere-biopsied embryos developed to term without a reduction in their developmental capacity. The ability to generate human ES cells without the destruction of ex utero embryos would reduce or eliminate the ethical concerns of many.


Asunto(s)
Blastómeros/citología , Diferenciación Celular , Separación Celular/métodos , Investigaciones con Embriones , Células Madre/citología , Animales , Biopsia , Técnicas de Cultivo de Célula , Células Cultivadas , Cariotipificación , Ratones , Teratoma , Trofoblastos/citología
3.
Stem Cells ; 28(4): 704-12, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20155819

RESUMEN

Human induced pluripotent stem cells (hiPSC) have been shown to differentiate into a variety of replacement cell types. Detailed evaluation and comparison with their human embryonic stem cell (hESC) counterparts is critical for assessment of their therapeutic potential. Using established methods, we demonstrate here that hiPSCs are capable of generating hemangioblasts/blast cells (BCs), endothelial cells, and hematopoietic cells with phenotypic and morphologic characteristics similar to those derived from hESCs, but with a dramatic decreased efficiency. Furthermore, in distinct contrast with the hESC derivatives, functional differences were observed in BCs derived from hiPSCs, including significantly increased apoptosis, severely limited growth and expansion capability, and a substantially decreased hematopoietic colony-forming capability. After further differentiation into erythroid cells, >1,000-fold difference in expansion capability was observed in hiPSC-BCs versus hESC-BCs. Although endothelial cells derived from hiPSCs were capable of taking up acetylated low-density lipoprotein and forming capillary-vascular-like structures on Matrigel, these cells also demonstrated early cellular senescence (most of the endothelial cells senesced after one passage). Similarly, retinal pigmented epithelium cells derived from hiPSCs began senescing in the first passage. Before clinical application, it will be necessary to determine the cause and extent of such abnormalities and whether they also occur in hiPSCs generated using different reprogramming methods.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Diferenciación Celular , Senescencia Celular , Hemangioblastos/citología , Células Madre Pluripotentes Inducidas/citología , Línea Celular , Proliferación Celular , Humanos , Fenotipo , Factores de Tiempo
4.
Stem Cells Transl Med ; 10 Suppl 2: S10-S17, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34724715

RESUMEN

Clinical success of adoptive cell therapy with chimeric antigen receptor (CAR) T cells for treating hematological malignancies has revolutionized the field of cellular immunotherapy. However, due to the nature of utilizing autologous T cells, affordability and availability are major hurdles, in addition to scientific challenges relating to CAR-T therapy optimization. Natural killer (NK) cell is a specialized immune effector cell type that recognizes and kills targets without human leukocyte antigen (HLA) restriction and prior sensitization. CAR-NK cells do not cause graft vs host disease and can be obtained from unrelated donors as well as pluripotent stem cells (PSC), representing an ideal off-the-shelf therapeutics readily available for patients. Furthermore, unlike cytotoxic T cells, NK cells specifically target and eliminate cancer stem cells, which are the cells causing relapse and metastasis. PSCs can be genetically manipulated and engineered with CARs at the pluripotent stage, which allows the establishment of permanent, stable, and clonal PSC-CAR lines for the manufacture of unlimited homogenous CAR-NK cells. Multiple master PSC-CAR cell banks targeting a variety of antigens for cancer, viral infection, and autoimmune diseases provide inexhaustible cell sources for all patients. Development of a next-generation 3D bioreactor platform for PSC expansion and NK cell production overcomes major barriers related to cost and scalability for CAR-NK product.


Asunto(s)
Neoplasias , Células Madre Pluripotentes , Receptores Quiméricos de Antígenos , Humanos , Inmunoterapia Adoptiva , Células Asesinas Naturales , Neoplasias/terapia , Células Madre Pluripotentes/metabolismo , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/metabolismo
5.
Transl Vis Sci Technol ; 10(9): 6, 2021 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-34347033

RESUMEN

Purpose: Retinitis pigmentosa (RP) is caused by mutations in more than 60 genes. Mutation-independent approaches to its treatment by exogeneous administration of neurotrophic factors that will preserve existing retinal anatomy and visual function are a rational strategy. Ciliary neurotrophic factor (CNTF) and oncostatin M (OSM) are two potent survival factors for neurons. However, growth factors degrade rapidly if administered directly. A sustained delivery of growth factors is required for translating their potential therapeutic benefit into patients. Methods: Stable and biocompatible nanoparticles (NP) that incorporated with CNTF and OSM (CNTF- and OSM-NP) were formulated. Both NP-trophic factors were tested in vitro using photoreceptor progenitor cells (PPC) and retinal ganglion progenitor cells (RGPC) derived from induced pluripotent stem cells and in vivo using an optic nerve crush model for glaucoma and the Royal College of Surgeons rat, model of RP (n = 8/treatment) by intravitreal delivery. Efficacy was evaluated by electroretinography and optokinetic response. Retinal histology and a whole mount analysis were performed at the end of experiments. Results: Significant prosurvival and pro-proliferation effects of both complexes were observed in both photoreceptor progenitor cells and RGPC in vitro. Importantly, significant RGC survival and preservation of vision and photoreceptors in both complex-treated animals were observed compared with control groups. Conclusions: These results demonstrate that NP-trophic factors are neuroprotective both in vitro and in vivo. A single intravitreal delivery of both NP-trophic factors offered neuroprotection in animal models of retinal degeneration. Translational Relevance: Sustained nanoparticle delivery of neurotrophic factors may offer beneficial effects in slowing down progressive retinal degenerative conditions, including retinitis pigmentosa, age-related macular degeneration, and glaucoma.


Asunto(s)
Nanopartículas , Degeneración Retiniana , Animales , Factor Neurotrófico Ciliar , Humanos , Oncostatina M , Ratas , Degeneración Retiniana/tratamiento farmacológico , Células Ganglionares de la Retina , Roedores
6.
Blood ; 112(12): 4475-84, 2008 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-18713948

RESUMEN

Human erythropoiesis is a complex multistep process that involves the differentiation of early erythroid progenitors to mature erythrocytes. Here we show that it is feasible to differentiate and mature human embryonic stem cells (hESCs) into functional oxygen-carrying erythrocytes on a large scale (10(10)-10(11) cells/6-well plate hESCs). We also show for the first time that the oxygen equilibrium curves of the hESC-derived cells are comparable with normal red blood cells and respond to changes in pH and 2,3-diphosphoglyerate. Although these cells mainly expressed fetal and embryonic globins, they also possessed the capacity to express the adult beta-globin chain on further maturation in vitro. Polymerase chain reaction and globin chain specific immunofluorescent analysis showed that the cells increased expression of beta-globin (from 0% to > 16%) after in vitro culture. Importantly, the cells underwent multiple maturation events, including a progressive decrease in size, increase in glycophorin A expression, and chromatin and nuclear condensation. This process resulted in extrusion of the pycnotic nuclei in up to more than 60% of the cells generating red blood cells with a diameter of approximately 6 to 8 mum. The results show that it is feasible to differentiate and mature hESCs into functional oxygen-carrying erythrocytes on a large scale.


Asunto(s)
Núcleo Celular/fisiología , Células Madre Embrionarias/fisiología , Eritrocitos/fisiología , Animales , Diferenciación Celular/fisiología , Fraccionamiento Celular , Células Cultivadas , Células Madre Embrionarias/citología , Eritrocitos/citología , Eritrocitos/metabolismo , Células Eritroides/citología , Células Eritroides/metabolismo , Citometría de Flujo , Humanos , Ratones , Sistema del Grupo Sanguíneo Rh-Hr/metabolismo , Ingeniería de Tejidos/métodos
7.
Hemoglobin ; 34(2): 145-50, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20353349

RESUMEN

Under culture conditions that promote hematopoietic differentiation, human embryonic stem cells (huESC) give rise to primitive erythroid cells that closely resemble the nucleated erythrocytes of early-stage human embryos. The globin chain distribution of these cells is similar to that seen during the embryonic and fetal stages of development. Here we show that huESC-derived erythroid cells produce substantial quantities of homotetrameric hemoglobin (Hb) composed exclusively of gamma-globin-containing subunits. The globin synthesis of these erythroid cells was also significantly unbalanced, with a substantial decrease of alpha-like globin chain synthesis in relation to that of their beta-like globins, a pattern characteristically associated with alpha-thalassemia (alpha-thal). This pattern of unbalanced globin synthesis appears to be an inherent feature of human erythroid cells that synthesize predominantly embryonic-stage globins.


Asunto(s)
Células Madre Embrionarias/citología , Eritroblastos/metabolismo , Eritropoyesis/genética , Regulación del Desarrollo de la Expresión Génica , Globinas alfa/biosíntesis , Talasemia alfa/genética , Globinas beta/biosíntesis , gamma-Globinas/biosíntesis , Células Cultivadas/citología , Células Cultivadas/metabolismo , Hemoglobinas Anormales/biosíntesis , Hemoglobinas Anormales/genética , Humanos , Globinas alfa/genética , Globinas beta/genética , gamma-Globinas/genética , Globinas zeta/biosíntesis , Globinas zeta/genética
8.
Am J Obstet Gynecol ; 200(1): 93.e1-8, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19121660

RESUMEN

OBJECTIVE: We used a mouse model of infection-induced preterm delivery to examine the roles of 2 adaptor proteins with central functions in Toll-like receptor signaling: MyD88 (myeloid differentiation primary-response gene 88) and TRIF (Toll/IL-1 receptor (TIR)-domain-containing adaptor protein-inducing IFN-beta). STUDY DESIGN: Mice deficient (KO) for MyD88, TRIF, both (DKO) or neither (WT) were inoculated into the uterus with killed Escherichia coli. Delivery outcomes, fetal status, serum progesterone, and nuclear translocation of the transcription factor nuclear factor kappa B (NFkappaB) were determined. RESULTS: Preterm birth (delivery in less than 48 hours) occurred in WT and TRIF-KO animals in a dose-dependent fashion, reaching 100% with 5-10 x 10(9) bacteria, while MyD88-KO and DKO animals were completely protected from delivery. Intrauterine fetal survival, maintenance of circulating progesterone levels, and nuclear translocation of NFkappaB were also dependent upon MyD88 but not TRIF. In contrast, induction of uterine interleukin (IL)-1beta and tumor necrosis factor alpha (TNF-alpha) depends upon actions of both MyD88 and TRIF. CONCLUSION: E coli-induced preterm delivery in the mouse is completely dependent upon MyD88 but not TRIF.


Asunto(s)
Infecciones por Escherichia coli/complicaciones , Escherichia coli/aislamiento & purificación , Factor 88 de Diferenciación Mieloide/metabolismo , Complicaciones Infecciosas del Embarazo/metabolismo , Nacimiento Prematuro/microbiología , Animales , Femenino , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , FN-kappa B/metabolismo , Embarazo , Complicaciones Infecciosas del Embarazo/microbiología , Nacimiento Prematuro/metabolismo , Progesterona/metabolismo , ARN/química , ARN/genética , Receptores de Interleucina/genética , Receptores de Interleucina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Estadísticas no Paramétricas
9.
Stem Cells Dev ; 16(4): 547-59, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17784829

RESUMEN

Enforced expression of the HoxB4 gene promotes expansion of hematopoietic stem cells (HSCs) and enhances hematopoietic development of both murine and human embryonic stem (ES) cells. HoxB4- expanded HSCs have also been shown to retain their normal potential for differentiation and longterm self-renewal in vivo without the development of leukemia, suggesting that manipulation of HoxB4 expression might represent an effective way to expand functional HSCs for use in transplantation medicine. However, the genetic modification of cells poses clinical concerns, including a potentially increased risk of tumor genicity. Constitutive high-level ectopic viral expression of HoxB4 can also produce perturbations in the lineage differentiation of HSCs, an indication that uncontrolled HoxB4 manipulation may not be a satisfactory therapeutic strategy. Here we demonstrate that recombinant HoxB4 protein fused with a triple protein transduction domain (tPTD) promotes hematopoietic development of hES cells. The tPTD-HoxB4 protein enhanced the development of erythroid, myeloid, and multipotential progenitors in both early- and late-stage embryoid bodies (EBs). This effect varied considerably between different hES cell lines. Addition of the tPTD-HoxB4 protein did not alter the globin gene expression pattern; progeny derived from hES cells expressed high levels of embryonic (epsilon) and fetal (gamma) globin genes with or without tPTD-HoxB4 treatment. CD34+ cells derived from hES cells engrafted in bone marrow when transplanted into fetal CD1 mice, although supplementation of the differentiation medium with tPTD-HoxB4 protein did not result in increased repopulating capacity. This suggests that other gene(s), together with HoxB4, are required for generating more competitive HSCs. In summary, our study demonstrates that the tPTD-HoxB4 protein can be used with other recombinant proteins to efficiently generate transplantable HSCs from human ES cells.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Embrionarias/fisiología , Hematopoyesis/fisiología , Proteínas de Homeodominio/genética , Factores de Transcripción/genética , Secuencia de Bases , Cartilla de ADN , ADN sin Sentido , Células Madre Embrionarias/citología , Proteínas de Homeodominio/metabolismo , Humanos , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , Regiones Promotoras Genéticas , Proteínas Recombinantes de Fusión/metabolismo , Factores de Transcripción/metabolismo
10.
Acta Ophthalmol ; 95(6): e468-e476, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28636206

RESUMEN

OBJECTIVE: To investigate whether intravitreally applied haemangioblasts (HB) derived from human embryonic stem cells (hESCs) are helpful for the repair of vascular damage caused in animals by an oxygen-induced retinopathy (OIR), by an induced diabetic retinopathy (DR) or by an induced retinal ischaemia with subsequent reperfusion. METHODS: Human embryonic stem cell-derived HBs were transplanted intravitreally into C57BL/6J mice (OIR model), into male Wistar rats with an induced DR and into male Wistar rats undergoing induced retinal ischaemia with subsequent reperfusion. Control groups of animals received an intravitreal injection of endothelial cells (ECs) or phosphate-buffered saline (PBS). We examined the vasculature integrity in the mice with OIR, the blood-retina barrier in the rats with induced DR, and retinal thickness and retinal ganglion cell density in retina flat mounts of the rats with the retinal ischaemic-reperfusion retinopathy. RESULTS: In the OIR model, the study group versus control groups showed a significantly (p < 0.001) smaller retinal avascular area [5.1 ± 2.7%;n = 18 animals versus 12.2 ± 2.8% (PBS group; n = 10 animals) and versus 11.8 ± 3.7% (EC group; n = 8 animals)] and less retinal neovascularization [6.3 ± 2.5%;n = 18 versus 15.2 ± 6.3% (n = 10; PBS group) and versus 15.8 ± 3.3% (n = 8; EC group)]. On retinal flat mounts, hESC-HBs were integrated into damaged retinal vessels and stained positive for PECAM (CD31) as EC marker. In the DR model, the study group versus the EC control group showed a significantly (p = 0.001) better blood-retina barrier function as measured at 2 days after the intravitreal injections [study group: 20.2 ± 12.8 µl/(g × hr); n = 6; versus EC control group: 52.9 ± 9.9 µl/(g × hr; n = 6)]. In the retinal ischaemia-reperfusion model, the groups did not differ significantly in retinal thickness and retinal ganglion cell density at 2, 5 and 7 days after baseline. CONCLUSION: By integrating into damaged retinal vessels and differentiating into ECs, intravitreally administered hESC-HBs may have partially repaired a retinal vascular injury caused by OIR model and DR.


Asunto(s)
Células Madre Embrionarias/trasplante , Hemangioblastos/trasplante , Enfermedades de la Retina/cirugía , Trasplante de Células Madre/métodos , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Inyecciones Intravítreas , Masculino , Ratones , Ratones Endogámicos C57BL , Oxígeno/toxicidad , Ratas , Ratas Wistar , Daño por Reperfusión/complicaciones , Enfermedades de la Retina/etiología , Enfermedades de la Retina/patología , Vasos Retinianos/patología
11.
Epigenomics ; 9(12): 1543-1558, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29135282

RESUMEN

AIM: To investigate the role of DNA methylation during erythrocyte production by human embryonic stem cells (hESCs). METHODS: We employed an erythroid differentiation model from hESCs, and then tracked the genome-wide DNA methylation maps and gene expression patterns through an Infinium HumanMethylation450K BeadChip and an Ilumina Human HT-12 v4 Expression Beadchip, respectively. RESULTS: A negative correlation between DNA methylation and gene expression was substantially enriched during the later differentiation stage and was present in both the promoter and the gene body. Moreover, erythropoietic genes with differentially methylated CpG sites that were primarily enriched in nonisland regions were upregulated, and demethylation of their gene bodies was associated with the presence of enhancers and DNase I hypersensitive sites. Finally, the components of JAK-STAT-NF-κB signaling were DNA hypomethylated and upregulated, which targets the key genes for erythropoiesis. CONCLUSION: Erythroid lineage commitment by hESCs requires genome-wide DNA methylation modifications to remodel gene expression dynamics.


Asunto(s)
Metilación de ADN , Células Madre Embrionarias/citología , Eritropoyesis , Genoma Humano , Línea Celular , Islas de CpG , Células Madre Embrionarias/metabolismo , Regulación del Desarrollo de la Expresión Génica , Humanos , Quinasas Janus/genética , FN-kappa B/genética , Factores de Transcripción STAT/genética , Transducción de Señal
12.
Methods Enzymol ; 418: 243-51, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17141039

RESUMEN

Embryonic stem (ES) cells, derived from early stage embryos, are pluripotent precursors of all of the tissues and organs of the body. ES cells from the mouse have been shown to undergo differentiation in vitro to form a variety of different cell types, including the differentiated progeny of hematopoietic precursors. These hematopoietic cells, however, exhibit numerous differences from those of human cells, and it has become increasingly clear that mouse ES cell differentiation has significant limitations as a model of human developmental biology. The more recent isolation and characterization of nonhuman primate ES cell lines have made available an experimental model with characteristics considerably more close to human biology. We have developed experimental conditions that promote efficient differentiation of these cells to produce progeny cells with considerable similarity to hematopoietic precursors harvested from bone marrow of adult animals.


Asunto(s)
Células Madre Embrionarias/citología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/fisiología , Animales , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular , Línea Celular , Células Madre Embrionarias/fisiología , Fibroblastos/citología , Fibroblastos/fisiología , Trasplante de Células Madre Hematopoyéticas , Humanos , Macaca mulatta , Ratones , Primates , Trasplante de Células Madre
13.
Sci Rep ; 6: 29784, 2016 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-27405580

RESUMEN

Photoreceptor degeneration due to retinitis pigmentosa (RP) is a primary cause of inherited retinal blindness. Photoreceptor cell-replacement may hold the potential for repair in a completely degenerate retina by reinstating light sensitive cells to form connections that relay information to downstream retinal layers. This study assessed the therapeutic potential of photoreceptor progenitors derived from human embryonic and induced pluripotent stem cells (ESCs and iPSCs) using a protocol that is suitable for future clinical trials. ESCs and iPSCs were cultured in four specific stages under defined conditions, resulting in generation of a near-homogeneous population of photoreceptor-like progenitors. Following transplantation into mice with end-stage retinal degeneration, these cells differentiated into photoreceptors and formed a cell layer connected with host retinal neurons. Visual function was partially restored in treated animals, as evidenced by two visual behavioral tests. Furthermore, the magnitude of functional improvement was positively correlated with the number of engrafted cells. Similar efficacy was observed using either ESCs or iPSCs as source material. These data validate the potential of human pluripotent stem cells for photoreceptor replacement therapies aimed at photoreceptor regeneration in retinal disease.


Asunto(s)
Ceguera , Diferenciación Celular , Células Madre Embrionarias Humanas , Células Madre Pluripotentes Inducidas , Células Fotorreceptoras de Vertebrados , Retinitis Pigmentosa , Animales , Ceguera/metabolismo , Ceguera/patología , Ceguera/terapia , Xenoinjertos , Células Madre Embrionarias Humanas/metabolismo , Células Madre Embrionarias Humanas/patología , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Ratones , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/patología , Células Fotorreceptoras de Vertebrados/trasplante , Retinitis Pigmentosa/metabolismo , Retinitis Pigmentosa/patología , Retinitis Pigmentosa/terapia
15.
Exp Hematol ; 30(1): 58-66, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11823038

RESUMEN

OBJECTIVE: The aim of this study was to characterize at the molecular level the hematopoietic progenitor cells derived from rhesus monkey embryonic stem (ES) cell differentiation. MATERIALS AND METHODS: We purified CD34(+) and CD34(+)CD38(-) cells from rhesus monkey ES cell cultures and examined the expression of a variety of genes associated with hematopoietic development, by semiquantitative polymerase chain reaction analysis. For comparison, we examined cell preparations from fresh or cultured rhesus monkey bone marrow (BM) and from mouse ES cells and BM. RESULTS: We observed a high degree of similarity in the expression patterns of these genes, with only a few exceptions. Most notably, the message of the flt3 gene was undetectable in rhesus monkey ES cell-derived CD34(+) and CD34(+)CD38(-) cells, whereas substantial flt3 expression was observed in the corresponding cells from fresh BM and in CD34(+) cells from cultured BM. The integrin alphaL and interleukin-6 (IL-6) receptor genes also were expressed in CD34(+)CD38(-) cells from BM, but there was little or no expression of these genes in CD34(+)CD38(-) cells derived from ES cells. Parallel analyses, using CD34(+)Lin(-) cells derived from murine ES cell cultures, showed no apparent expression of flt3, integrin alphaL, or IL-6 receptor, whereas corresponding cell preparations isolated from mouse BM expressed high levels of all of these genes. CONCLUSIONS: ES cell-derived hematopoietic progenitors, both from the rhesus monkey and from the mouse, exhibited the same alterations in gene expression compared with BM-derived cells from these animals. These observations could reflect the presence of different subpopulations in the cell fractions that were compared, or they may represent altered biologic properties of ES cell-derived hematopoietic stem cells.


Asunto(s)
Antígenos CD , Diferenciación Celular/genética , Células Madre Hematopoyéticas/fisiología , Células Madre/fisiología , ADP-Ribosil Ciclasa , ADP-Ribosil Ciclasa 1 , Animales , Antígenos CD34 , Antígenos de Diferenciación , Linaje de la Célula/genética , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Macaca mulatta , NAD+ Nucleosidasa , Reacción en Cadena de la Polimerasa
16.
Stem Cells Int ; 2015: 389628, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25755671

RESUMEN

Shortage of red blood cells (RBCs, erythrocytes) can have potentially life-threatening consequences for rare or unusual blood type patients with massive blood loss resulting from various conditions. Erythrocytes have been derived from human pluripotent stem cells (PSCs), but the risk of potential tumorigenicity cannot be ignored, and a majority of these cells produced from PSCs express embryonic ε- and fetal γ-globins with little or no adult ß-globin and remain nucleated. Here we report a method to generate erythrocytes from human hair follicle mesenchymal stem cells (hHFMSCs) by enforcing OCT4 gene expression and cytokine stimulation. Cells generated from hHFMSCs expressed mainly the adult ß-globin chain with minimum level of the fetal γ-globin chain. Furthermore, these cells also underwent multiple maturation events and formed enucleated erythrocytes with a biconcave disc shape. Gene expression analyses showed that OCT4 regulated the expression of genes associated with both pluripotency and erythroid development during hHFMSC transdifferentiation toward erythroid cells. These findings show that mature erythrocytes can be generated from adult somatic cells, which may serve as an alternative source of RBCs for potential autologous transfusion.

17.
Cancer Lett ; 186(2): 165-75, 2002 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-12213286

RESUMEN

The Copenhagen (COP) rat is extremely resistant to mammary cancer induction by carcinogens. Multiple genetic loci have been linked to the resistant phenotype, but the mechanisms underlying the resistance still remain unknown. Evidence has shown that the acquisition of angiogenic capacity is critical for tumor development. We, therefore, decided to investigate whether administration of angiogenic factor would enhance mammary carcinogenesis in the COP rat. Vascular endothelial growth factor (VEGF) was administered in a sustained releasing formula to pubescent female COP rats 2 weeks after N-nitroso-N-methylurea (NMU) treatment. Six months after NMU exposure, we found no difference in mammary tumor incidence between VEGF treated animals and controls. Analysis of VEGF expression, however, revealed different expression patterns in mammary epithelial cells of various origins. Mammary epithelial cells from pubescent susceptible Buffalo (BUF) and COP rats expressed substantial levels of VEGF messages, whereas cells prepared from 230-day-old rats showed negligible levels of VEGF mRNA. We also demonstrated that mammary epithelial cells from tumors developed in susceptible BUF rats expressed VEGF, whereas VEGF messages were barely detectable in tumors induced in COP rats. Furthermore, enlargement of the intramammary lymph nodes with prominent mast cells was observed in NMU treated COP rats, but not in NMU treated BUF rats. These results suggest that down regulation of VEGF expression is insufficient for resistance to mammary carcinogenesis, and that enhanced immune response, as evidenced by intramammary lymph node enlargement with mast cell accumulation, may also play a role in conferring resistance in the COP rat.


Asunto(s)
Factores de Crecimiento Endotelial/fisiología , Inmunidad Innata , Péptidos y Proteínas de Señalización Intercelular/fisiología , Linfocinas/fisiología , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/prevención & control , Mastocitos/fisiología , Animales , Carcinógenos , Células Cultivadas , Femenino , Genes ras , Ganglios Linfáticos/patología , Metilnitrosourea , Mutación , Fenotipo , ARN Mensajero/metabolismo , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
18.
Stem Cell Reports ; 3(5): 817-31, 2014 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-25418726

RESUMEN

Human induced pluripotent stem cells (iPSCs) provide a potentially replenishable source for the production of transfusable platelets. Here, we describe a method to generate megakaryocytes (MKs) and functional platelets from iPSCs in a scalable manner under serum/feeder-free conditions. The method also permits the cryopreservation of MK progenitors, enabling a rapid "surge" capacity when large numbers of platelets are needed. Ultrastructural/morphological analyses show no major differences between iPSC platelets and human blood platelets. iPSC platelets form aggregates, lamellipodia, and filopodia after activation and circulate in macrophage-depleted animals and incorporate into developing mouse thrombi in a manner identical to human platelets. By knocking out the ß2-microglobulin gene, we have generated platelets that are negative for the major histocompatibility antigens. The scalable generation of HLA-ABC-negative platelets from a renewable cell source represents an important step toward generating universal platelets for transfusion as well as a potential strategy for the management of platelet refractoriness.


Asunto(s)
Plaquetas/citología , Diferenciación Celular , Células Madre Pluripotentes Inducidas/citología , Megacariocitos/citología , Animales , Antígenos CD34/metabolismo , Plaquetas/metabolismo , Plaquetas/ultraestructura , Técnicas de Cultivo de Célula/métodos , Proliferación Celular , Células Cultivadas , Técnicas de Inactivación de Genes , Antígenos HLA/genética , Antígenos HLA/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/ultraestructura , Leucosialina/metabolismo , Masculino , Megacariocitos/metabolismo , Megacariocitos/ultraestructura , Ratones Endogámicos NOD , Ratones SCID , Microscopía Electrónica , Microscopía Fluorescente , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Transfusión de Plaquetas/métodos , Reproducibilidad de los Resultados , Trasplante Heterólogo , Microglobulina beta-2/genética , Microglobulina beta-2/metabolismo
19.
Stem Cell Reports ; 3(1): 115-30, 2014 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-25068126

RESUMEN

Current therapies for multiple sclerosis (MS) are largely palliative, not curative. Mesenchymal stem cells (MSCs) harbor regenerative and immunosuppressive functions, indicating a potential therapy for MS, yet the variability and low potency of MSCs from adult sources hinder their therapeutic potential. MSCs derived from human embryonic stem cells (hES-MSCs) may be better suited for clinical treatment of MS because of their unlimited and stable supply. Here, we show that hES-MSCs significantly reduce clinical symptoms and prevent neuronal demyelination in a mouse experimental autoimmune encephalitis (EAE) model of MS, and that the EAE disease-modifying effect of hES-MSCs is significantly greater than that of human bone-marrow-derived MSCs (BM-MSCs). Our evidence also suggests that increased IL-6 expression by BM-MSCs contributes to the reduced anti-EAE therapeutic activity of these cells. A distinct ability to extravasate and migrate into inflamed CNS tissues may also be associated with the robust therapeutic effects of hES-MSCs on EAE.


Asunto(s)
Células de la Médula Ósea/citología , Células Madre Embrionarias/citología , Encefalomielitis Autoinmune Experimental/terapia , Células Madre Mesenquimatosas/citología , Esclerosis Múltiple/patología , Esclerosis Múltiple/terapia , Animales , Sistema Nervioso Central/patología , Modelos Animales de Enfermedad , Humanos , Trasplante de Células Madre Mesenquimatosas , Ratones
20.
Regen Med ; 8(4): 413-24, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23826696

RESUMEN

BACKGROUND: Human embryonic stem cells (hESCs) have been derived and maintained on mouse embryonic fibroblast feeders to keep their undifferentiated status. To realize their clinical potential, a feeder-free and scalable system for large scale production of hESCs and their differentiated derivatives is required. MATERIALS & METHODS: hESCs were cultured and passaged on serum/feeder-free 3D microcarriers for five passages. For embryoid body (EB) formation and hemangioblast differentiation, the medium for 3D microcarriers was directly switched to EB medium. RESULTS: hESCs on 3D microcarriers maintained pluripotency and formed EBs, which were ten-times more efficient than hESCs cultured under 2D feeder-free conditions (0.11 ± 0.03 EB cells/hESC input 2D vs 1.19 ± 0.32 EB cells/hESC input 3D). After replating, EB cells from 3D culture readily developed into hemangioblasts with the potential to differentiate into hematopoietic and endothelial cells. Furthermore, this 3D system can also be adapted to human induced pluripotent stem cells, which generate functional hemangioblasts with high efficiency. CONCLUSION: This 3D serum- and stromal-free microcarrier system is important for future clinical applications, with the potential of developing to a GMP-compatible scalable system.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Diferenciación Celular , Medio de Cultivo Libre de Suero , DEAE-Celulosa/química , Células Nutrientes , Hemangioblastos/citología , Células Madre Hematopoyéticas/citología , Células Madre Pluripotentes/citología , Animales , Células Cultivadas , Colágeno/metabolismo , Combinación de Medicamentos , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Hemangioblastos/metabolismo , Células Madre Hematopoyéticas/metabolismo , Humanos , Laminina/metabolismo , Ratones , Microesferas , Células Madre Pluripotentes/metabolismo , Proteoglicanos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA