Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 115(4): 708-713, 2018 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-29311328

RESUMEN

Genetically engineered pigs play an indispensable role in the study of rare monogenic diseases. Pigs harboring a gene responsible for a specific disease can be efficiently generated via somatic cell cloning. The generation of somatic cell-cloned pigs from male cells with mutation(s) in an X chromosomal gene is a reliable and straightforward method for reproducing X-linked genetic diseases (XLGDs) in pigs. However, the severe symptoms of XLGDs are often accompanied by impaired growth and reproductive disorders, which hinder the reproduction of these valuable model animals. Here, we generated unique chimeric boars composed of mutant cells harboring a lethal XLGD and normal cells. The chimeric boars exhibited the cured phenotype with fertility while carrying and transmitting the genotype of the XLGD. This unique reproduction system permits routine production of XLGD model pigs through the male-based breeding, thereby opening an avenue for translational research using disease model pigs.


Asunto(s)
Técnicas de Cultivo de Embriones/métodos , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Reproducción/genética , Animales , Animales Modificados Genéticamente/genética , Cruzamiento , Quimera , Clonación de Organismos/métodos , Modelos Animales de Enfermedad , Fertilidad , Técnicas de Inactivación de Genes/métodos , Ingeniería Genética/métodos , Masculino , Técnicas de Transferencia Nuclear , Porcinos/genética
2.
Genesis ; 51(11): 763-76, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23913699

RESUMEN

Porcine induced pluripotent stem cells (iPSCs) provide useful information for translational research. The quality of iPSCs can be assessed by their ability to differentiate into various cell types after chimera formation. However, analysis of chimera formation in pigs is a labor-intensive and costly process, necessitating a simple evaluation method for porcine iPSCs. Our previous study identified mouse embryonic stem cell (ESC)-specific hypomethylated loci (EShypo-T-DMRs), and, in this study, 36 genes selected from these were used to evaluate porcine iPSC lines. Based on the methylation profiles of the 36 genes, the iPSC line, Porco Rosso-4, was found closest to mouse pluripotent stem cells among 5 porcine iPSCs. Moreover, Porco Rosso-4 more efficiently contributed to the inner cell mass (ICM) of blastocysts than the iPSC line showing the lowest reprogramming of the 36 genes (Porco Rosso-622-14), indicating that the DNA methylation profile correlates with efficiency of ICM contribution. Furthermore, factors known to enhance iPSC quality (serum-free medium with PD0325901 and CHIR99021) improved the methylation status at the 36 genes. Thus, the DNA methylation profile of these 36 genes is a viable index for evaluation of porcine iPSCs.


Asunto(s)
Metilación de ADN , Células Madre Embrionarias/metabolismo , Sitios Genéticos , Células Madre Pluripotentes Inducidas/metabolismo , Porcinos Enanos/embriología , Porcinos/embriología , Animales , Masa Celular Interna del Blastocisto/metabolismo , Línea Celular , Quimera , Embrión de Mamíferos , Expresión Génica , Genes , Ratones , Investigación Biomédica Traslacional
3.
Methods Mol Biol ; 2637: 213-221, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36773149

RESUMEN

Knock-in therapy, in which an insertion site can be controlled, would be more suitable for the treatment of genetic blood disorders as compared to conventional gene therapy with lentivirus vectors that introduce genes into the genome randomly. Recent advancements in genome editing technology have substantially improved the knock-in efficiency, making it a reality. We present the details of a virus-free CRISPR/Cas9-based genome editing method for bona fide mouse hematopoietic stem cells.


Asunto(s)
Sistemas CRISPR-Cas , Edición Génica , Ratones , Animales , Edición Génica/métodos , Terapia Genética/métodos , Lentivirus/genética
4.
Commun Med (Lond) ; 3(1): 56, 2023 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-37076593

RESUMEN

BACKGROUND: Base editing via CRISPR-Cas9 has garnered attention as a method for correcting disease-specific mutations without causing double-strand breaks, thereby avoiding large deletions and translocations in the host chromosome. However, its reliance on the protospacer adjacent motif (PAM) can limit its use. We aimed to restore a disease mutation in a patient with severe hemophilia B using base editing with SpCas9-NG, a modified Cas9 with the board PAM flexibility. METHODS: We generated induced pluripotent stem cells (iPSCs) from a patient with hemophilia B (c.947T>C; I316T) and established HEK293 cells and knock-in mice expressing the patient's F9 cDNA. We transduced the cytidine base editor (C>T), including the nickase version of Cas9 (wild-type SpCas9 or SpCas9-NG), into the HEK293 cells and knock-in mice through plasmid transfection and an adeno-associated virus vector, respectively. RESULTS: Here we demonstrate the broad PAM flexibility of SpCas9-NG near the mutation site. The base-editing approach using SpCas9-NG but not wild-type SpCas9 successfully converts C to T at the mutation in the iPSCs. Gene-corrected iPSCs differentiate into hepatocyte-like cells in vitro and express substantial levels of F9 mRNA after subrenal capsule transplantation into immunodeficient mice. Additionally, SpCas9-NG-mediated base editing corrects the mutation in both HEK293 cells and knock-in mice, thereby restoring the production of the coagulation factor. CONCLUSION: A base-editing approach utilizing the broad PAM flexibility of SpCas9-NG can provide a solution for the treatment of genetic diseases, including hemophilia B.


In patients with hemophilia B, the blood does not clot properly, leading to excessive bruising and bleeding. Hemophilia B is caused by an error in a gene called coagulation factor IX (F9). To treat patients with hemophilia B, we might be able to use a technology called CRISPR-Cas9 to edit and correct this genetic error, restoring factor IX function and improving clotting. Here, we test a specific CRISPR-Cas9 approach in cells and animals. We show that we are able to correct the genetic error in F9 in cells isolated from a patient with severe hemophilia B. We also show that we can fix the error in mice and that this increases levels of factor IX in the blood of the mice. With further testing, this gene-editing approach may be a viable therapy for patients with hemophilia B or similar genetic disorders.

5.
Nat Commun ; 14(1): 8031, 2023 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-38052804

RESUMEN

Cancer cells inevitably interact with neighboring host tissue-resident cells during the process of metastatic colonization, establishing a metastatic niche to fuel their survival, growth, and invasion. However, the underlying mechanisms in the metastatic niche are yet to be fully elucidated owing to the lack of methodologies for comprehensively studying the mechanisms of cell-cell interactions in the niche. Here, we improve a split green fluorescent protein (GFP)-based genetically encoded system to develop secretory glycosylphosphatidylinositol-anchored reconstitution-activated proteins to highlight intercellular connections (sGRAPHIC) for efficient fluorescent labeling of tissue-resident cells that neighbor on and putatively interact with cancer cells in deep tissues. The sGRAPHIC system enables the isolation of metastatic niche-associated tissue-resident cells for their characterization using a single-cell RNA sequencing platform. We use this sGRAPHIC-leveraged transcriptomic platform to uncover gene expression patterns in metastatic niche-associated hepatocytes in a murine model of liver metastasis. Among the marker genes of metastatic niche-associated hepatocytes, we identify Lgals3, encoding galectin-3, as a potential pro-metastatic factor that accelerates metastatic growth and invasion.


Asunto(s)
Neoplasias Hepáticas , Humanos , Ratones , Animales , Neoplasias Hepáticas/metabolismo , Hepatocitos/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Matriz Extracelular/metabolismo , Comunicación Celular
6.
Biol Reprod ; 87(6): 133, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23053438

RESUMEN

In vitro matured (IVM) oocytes have been used to create genetically modified pigs for various biomedical purposes. However, porcine embryos derived from IVM oocytes are very cryosensitive. Developing improved cryopreservation methods would facilitate the production of genetically modified pigs and also accelerate the conservation of genetic resources. We recently developed a novel hollow fiber vitrification (HFV) method; the present study was initiated to determine whether this new method permits the cryopreservation of IVM oocyte-derived porcine embryos. Embryos were created from the in vitro fertilization of IVM oocytes with frozen-thawed sperm derived from a transgenic pig carrying a humanized Kusabira-Orange (huKO) gene. Morula-stage embryos were assigned to vitrification and nonvitrification groups to compare their in vitro and in vivo developmental abilities. Vitrified morulae developed to the blastocyst stage at a rate similar to that of nonvitrified embryos (66/85, 77.6% vs. 67/84, 79.8%). Eighty-eight blastocysts that developed from vitrified morulae were transferred into the uteri of three recipient gilts. All three became pregnant and produced a total of 17 piglets (19.3%). This piglet production was slightly lower, albeit not significantly, than that of the nonvitrification group (27/88, 30.7%). Approximately half of the piglets in the vitrification (10/17, 58.8%) and nonvitrification (15/27, 55.6%) groups were transgenic. There was no significant difference in the growth rates among the piglets in the two groups. These results indicate that the HFV method is an extremely effective method for preserving cryosensitive embryos such as porcine in vitro maturation/fertilization-derived morulae.


Asunto(s)
Animales Modificados Genéticamente/fisiología , Criopreservación/veterinaria , Fertilización In Vitro/veterinaria , Técnicas de Maduración In Vitro de los Oocitos/veterinaria , Mórula , Sus scrofa/fisiología , Vitrificación , Animales , Animales Modificados Genéticamente/genética , Animales Modificados Genéticamente/crecimiento & desarrollo , Blastocisto/metabolismo , Cruzamientos Genéticos , Criopreservación/instrumentación , Ectogénesis , Implantación del Embrión , Pérdida del Embrión/prevención & control , Transferencia de Embrión/efectos adversos , Transferencia de Embrión/veterinaria , Femenino , Fertilización In Vitro/efectos adversos , Nacimiento Vivo , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Masculino , Mórula/citología , Mórula/metabolismo , Embarazo , Distribución Aleatoria , Preservación de Semen/efectos adversos , Sus scrofa/genética , Sus scrofa/crecimiento & desarrollo , Aumento de Peso , Proteína Fluorescente Roja
7.
Exp Hematol ; 95: 46-57.e8, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33395577

RESUMEN

We report that a sheep fetal liver provides a microenvironment for generating hematopoietic cells with long-term engrafting capacity and multilineage differentiation potential from human induced pluripotent stem cell (iPSC)-derived hemogenic endothelial cells (HEs). Despite the promise of iPSCs for making any cell types, generating hematopoietic stem and progenitor cells (HSPCs) is still a challenge. We hypothesized that the hematopoietic microenvironment, which exists in fetal liver but is lacking in vitro, turns iPSC-HEs into HSPCs. To test this, we transplanted CD45-negative iPSC-HEs into fetal sheep liver, in which HSPCs first grow. Within 2 months, the transplanted cells became CD45 positive and differentiated into multilineage blood cells in the fetal liver. Then, CD45-positive cells translocated to the bone marrow and were maintained there for 3 years with the capability of multilineage differentiation, indicating that hematopoietic cells with long-term engraftment potential were generated. Moreover, human hematopoietic cells were temporally enriched by xenogeneic donor-lymphocyte infusion into the sheep. This study could serve as a foundation to generate HSPCs from iPSCs.


Asunto(s)
Células Madre Hematopoyéticas/citología , Células Madre Pluripotentes Inducidas/trasplante , Ovinos/embriología , Animales , Células de la Médula Ósea/citología , Diferenciación Celular , Linaje de la Célula , Movimiento Celular , Microambiente Celular , Ensayo de Unidades Formadoras de Colonias , Trasplante de Células Madre de Sangre del Cordón Umbilical , Femenino , Técnicas Genéticas , Supervivencia de Injerto , Hemangioblastos/citología , Xenoinjertos , Humanos , Células Madre Pluripotentes Inducidas/citología , Antígenos Comunes de Leucocito/análisis , Hígado/embriología , Subgrupos Linfocitarios , Embarazo , Especificidad de la Especie
8.
Mol Ther Methods Clin Dev ; 20: 451-462, 2021 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-33614821

RESUMEN

We conducted two lines of genome-editing experiments of mouse hematopoietic stem cells (HSCs) with the clustered regularly interspaced short palindromic repeat (CRISPR) and CRISPR-associated protein 9 (Cas9). First, to evaluate the genome-editing efficiency in mouse bona fide HSCs, we knocked out integrin alpha 2b (Itga2b) with Cas9 ribonucleoprotein (Cas9/RNP) and performed serial transplantation in mice. The knockout efficiency was estimated at approximately 15%. Second, giving an example of X-linked severe combined immunodeficiency (X-SCID) as a target genetic disease, we showed a proof-of-concept of universal gene correction, allowing rescue of most of X-SCID mutations, in a completely non-viral setting. We inserted partial cDNA of interleukin-2 receptor gamma chain (Il2rg) into intron 1 of Il2rg via non-homologous end-joining (NHEJ) with Cas9/RNP and a homology-independent targeted integration (HITI)-based construct. Repaired HSCs reconstituted T lymphocytes and thymuses in SCID mice. Our results show that a non-viral genome-editing of HSCs with CRISPR/Cas9 will help cure genetic diseases.

9.
Methods Mol Biol ; 2320: 247-259, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34302663

RESUMEN

A knock-in can generate fluorescent or Cre-reporter under the control of an endogenous promoter. It also generates knock-out or tagged-protein with fluorescent protein and short tags for tracking and purification. Recent advances in genome editing with clustered regularly interspaced short palindromic repeat (CRISPR) and CRISPR-associated protein 9 (Cas9) significantly increased the efficiencies of making knock-in cells. Here we describe the detailed protocols of generating knock-in mouse and human pluripotent stem cells (PSCs) by electroporation and lipofection, respectively.


Asunto(s)
Sistemas CRISPR-Cas , Técnicas de Sustitución del Gen/métodos , Células Madre Pluripotentes Inducidas/citología , Animales , Células Cultivadas , Células Clonales , Medios de Cultivo , Cartilla de ADN , Resistencia a Medicamentos/genética , Electroporación , Células Madre Embrionarias/citología , Edición Génica/métodos , Genes Reporteros , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Ratones Endogámicos C57BL , Puromicina/farmacología , ARN Guía de Kinetoplastida/genética , Reparación del ADN por Recombinación/genética
10.
J Vis Exp ; (169)2021 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-33749676

RESUMEN

Pluripotent stem cell-derived cardiomyocytes (PSC-CMs) can be produced from both embryonic and induced pluripotent stem (ES/iPS) cells. These cells provide promising sources for cardiac disease modeling. For cardiomyopathies, sarcomere shortening is one of the standard physiological assessments that are used with adult cardiomyocytes to examine their disease phenotypes. However, the available methods are not appropriate to assess the contractility of PSC-CMs, as these cells have underdeveloped sarcomeres that are invisible under phase-contrast microscopy. To address this issue and to perform sarcomere shortening with PSC-CMs, fluorescent-tagged sarcomere proteins and fluorescent live-imaging were used. Thin Z-lines and an M-line reside at both ends and the center of a sarcomere, respectively. Z-line proteins - α-Actinin (ACTN2), Telethonin (TCAP), and actin-associated LIM protein (PDLIM3) - and one M-line protein - Myomesin-2 (Myom2) - were tagged with fluorescent proteins. These tagged proteins can be expressed from endogenous alleles as knock-ins or from adeno-associated viruses (AAVs). Here, we introduce the methods to differentiate mouse and human pluripotent stem cells to cardiomyocytes, to produce AAVs, and to perform and analyze live-imaging. We also describe the methods for producing polydimethylsiloxane (PDMS) stamps for a patterned culture of PSC-CMs, which facilitates the analysis of sarcomere shortening with fluorescent-tagged proteins. To assess sarcomere shortening, time-lapse images of the beating cells were recorded at a high framerate (50-100 frames per second) under electrical stimulation (0.5-1 Hz). To analyze sarcomere length over the course of cell contraction, the recorded time-lapse images were subjected to SarcOptiM, a plug-in for ImageJ/Fiji. Our strategy provides a simple platform for investigating cardiac disease phenotypes in PSC-CMs.


Asunto(s)
Colorantes Fluorescentes/metabolismo , Miocitos Cardíacos/metabolismo , Células Madre Pluripotentes/citología , Sarcómeros/metabolismo , Animales , Diferenciación Celular , Células Cultivadas , Dependovirus/metabolismo , Cuerpos Embrioides/citología , Humanos , Ratones , Células Madre Embrionarias de Ratones/citología , Miocitos Cardíacos/citología , Coloración y Etiquetado , Imagen de Lapso de Tiempo
11.
Nihon Rinsho ; 73 Suppl 5: 360-4, 2015 Jun.
Artículo en Japonés | MEDLINE | ID: mdl-30458079
12.
Sci Rep ; 10(1): 4249, 2020 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-32144297

RESUMEN

Pluripotent stem cell-derived cardiomyocytes (PSC-CMs) hold great promise for disease modeling and drug discovery. However, PSC-CMs exhibit immature phenotypes in culture, and the lack of maturity limits their broad applications. While physical and functional analyses are generally used to determine the status of cardiomyocyte maturation, they could be time-consuming and often present challenges in comparing maturation-enhancing strategies. Therefore, there is a demand for a method to assess cardiomyocyte maturation rapidly and reproducibly. In this study, we found that Myomesin-2 (Myom2), encoding M-protein, is upregulated postnatally, and based on this, we targeted TagRFP to the Myom2 locus in mouse embryonic stem cells. Myom2-RFP+ PSC-CMs exhibited more mature phenotypes than RFP- cells in morphology, function and transcriptionally, conductive to sarcomere shortening assays. Using this system, we screened extracellular matrices (ECMs) and identified laminin-511/521 as potent enhancers of cardiomyocyte maturation. Together, we developed and characterized a novel fluorescent reporter system for the assessment of cardiomyocyte maturation and identified potent maturation-enhancing ECMs through this simple and rapid assay. This system is expected to facilitate use of PSC-CMs in a variety of scientific and medical investigations.


Asunto(s)
Biomarcadores , Diferenciación Celular , Expresión Génica , Genes Reporteros , Laminina/genética , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Calcio , Diferenciación Celular/genética , Biología Computacional/métodos , Matriz Extracelular/metabolismo , Perfilación de la Expresión Génica , Laminina/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Sarcómeros/metabolismo , Transcripción Genética , Transcriptoma
13.
Exp Anim ; 69(2): 189-198, 2020 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-31801915

RESUMEN

X-linked severe combined immunodeficiency (X-SCID) is an inherited genetic disorder. A majority of X-SCID subjects carries point mutations in the Interleukin-2 receptor gamma chain (IL2RG) gene. In contrast, Il2rg-knockout mice recapitulating X-SCID phenotype lack a large part of Il2rg instead of point mutations. In this study, we generated novel X-SCID mouse strains with small insertion and deletion (InDel) mutations in Il2rg by using clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9. To this end, we injected Streptococcus pyogenes Cas9 (SpCas9) mRNA and single guide RNA targeting the exon 2, 3 or 4 of Il2rg into mouse zygotes. In the F0 generation, we obtained 35 pups and 25 out of them were positive for Surveyor assay, and most of mutants displayed dramatic reductions of T and B lymphocytes in the peripheral blood. By amplicon sequencing, 15 out of 31 founder mice were determined as monoallelic mutants with possible minor mosaicisms while 10 mice were mosaic. Finally, we established new strains with 7-nucleotide deletion and 1-nucleotide insertions in the exon 2 and the exons 3 and 4, respectively. Although no IL2RG protein was detected on T cells of exons 3 and 4 mutants, IL2RG protein was unexpectedly detected in the exon 2 mutants. These data indicated that CRISPR/Cas9 targeting Il2rg causes InDel mutations effectively and generates genetically X-SCID mice. Genetic mutations, however, did not necessarily grant phenotypical alteration, which requires an intensive analysis after establishing a strain to confirm their phenotypes.


Asunto(s)
Proteína 9 Asociada a CRISPR/genética , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Animales , Modelos Animales de Enfermedad , Edición Génica , Subunidad gamma Común de Receptores de Interleucina/genética , Ratones , Ratones Noqueados , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X
14.
Cancer Sci ; 100(12): 2444-50, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19775286

RESUMEN

Constitutive activation of Notch signaling is required for the proliferation of a subgroup of human T-cell acute lymphoblastic leukemias (T-ALL). Previous in vitro studies have demonstrated the therapeutic potential of Notch signaling inhibitors for treating T-ALL. To further examine this possibility, we applied a gamma-secretase inhibitor (GSI) to T-ALL xenograft models. Treatment of established subcutaneous tumors with GSI resulted in partial or complete regression of tumors arising from four T-ALL cell lines that were also sensitive to GSI in vitro. To elucidate the mechanism of action, we transduced DND-41 cells with the active form of Notch1 (aN1), which conferred resistance to in vitro GSI treatment. Nevertheless, in vivo treatment with GSI induced a partial but significant regression of subcutaneous tumors that developed from aN1-transduced DND-41 cells, whereas it induced complete regression of tumors that developed from mock-transduced DND-41 cells. These findings indicate that the remarkable efficacy of GSI might be attributable to dual mechanisms, directly via apoptosis of DND-41 cells through the inhibition of cell-autonomous Notch signaling, and indirectly via disturbance of tumor angiogenesis through the inhibition of non-cell-autonomous Notch signaling.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Antineoplásicos/farmacología , Leucemia-Linfoma Linfoblástico de Células T Precursoras/tratamiento farmacológico , Receptores Notch/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Animales , Línea Celular Tumoral , Humanos , Ratones , Ratones SCID , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Cell Transplant ; 18(4): 381-9, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19622226

RESUMEN

The ERas gene promotes the proliferation of and formation of teratomas by mouse embryonic stem (ES) cells. However, its human orthologue is not expressed in human ES cells. This implies that the behavior of transplanted mouse ES cells would not accurately reflect the behavior of transplanted human ES cells and that the use of nonhuman primate models might be more appropriate to demonstrate the safety of human ES cell-based therapies. However, the expression of the ERas gene has not been examined in nonhuman primate ES cells. In this study, we cloned the cynomolgus homologue and showed that the ERas gene is expressed in cynomolgus ES cells. Notably, it is also expressed in cynomolgus ES cell-derived differentiated progeny as well as cynomolgus adult tissues. The ERas protein is detectable in various cynomolgus tissues as assessed by immunohistochemisty. Cynomolgus ES cell-derived teratoma cells, which also expressed the ERas gene at higher levels than the undifferentiated cynomolgus ES cells, did not develop tumors in NOD/Shi-scid, IL-2Rgamma(null) (NOG) mice. Even when the ERas gene was overexpressed in cynomolgus stromal cells, only the plating efficiency was improved and the proliferation was not promoted. Thus, it is unlikely that ERas contributes to the tumorigenicity of cynomolgus cells. Therefore, cynomolgus ES cells are more similar to human than mouse ES cells despite that ERas is expressed in cynomolgus and mouse ES cells but not in human ES cells.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Células Madre Embrionarias/metabolismo , Proteína Oncogénica p21(ras)/metabolismo , Teratoma/metabolismo , Secuencia de Aminoácidos , Animales , Transformación Celular Neoplásica/patología , Células Cultivadas , Células Madre Embrionarias/patología , Humanos , Macaca fascicularis , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Trasplante de Neoplasias , Especificidad de Órganos , Especificidad de la Especie , Teratoma/patología , Trasplante Heterólogo
17.
Stem Cells Dev ; 17(2): 367-81, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18447651

RESUMEN

Because embryonic stem (ES) cells are able to proliferate indefinitely and differentiate into any type of cell, they have the potential for providing an inexhaustible supply of transplantable cells or tissues. However, methods for the in vitro differentiation of human ES cells are still quite limited. One possible strategy would be to generate differentiated cells in vivo. In view of future clinical application, we investigated the possibility of using xenogeneic large animals for this purpose. We transplanted nonhuman primate cynomolgus ES cells into fetal sheep at 43-67 gestational days (full term 147 days, n=15). After birth, cynomolgus tissues, which were mature teratomas, had been engrafted in sheep when more than 1 x 10(6) ES cells were transplanted at <50 gestational days. Despite the sustained engraftment, both cellular and humoral immune responses against the ES cells were detected, and additional transplantation was not successful in the animals. At 2 weeks post-transplantation, the ES cell progeny proliferated when transplanted at 48 (<50) gestational days, whereas they were cleared away when transplanted at 60 (>50) gestational days. These results support the rapid development of the xenogeneic immunological barrier in fetal sheep after 50 gestational days. Notably, a large number of Foxp3(+) regulatory T cells were present around the ES cell progeny, but macrophages were absent when the transplant was conducted at <50 gestational days, implying that regulatory T cells and premature innate immunity might have contributed to the sustained engraftment. In conclusion, long-term macroscopic engraftment of primate ES cells in sheep is feasible despite the xenogeneic immunological barrier.


Asunto(s)
Transferencia de Embrión , Células Madre Embrionarias/trasplante , Supervivencia de Injerto , Macaca fascicularis , Ovinos , Útero , Adaptación Biológica/genética , Adaptación Biológica/inmunología , Secuencia de Aminoácidos , Animales , Animales Recién Nacidos , Células Cultivadas , Transferencia de Embrión/métodos , Embrión de Mamíferos , Desarrollo Embrionario/genética , Células Madre Embrionarias/fisiología , Femenino , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Inmunidad Innata/genética , Inmunidad Innata/fisiología , Datos de Secuencia Molecular , Embarazo , Ovinos/embriología , Quimera por Trasplante , Inmunología del Trasplante , Trasplante Heterólogo , Útero/fisiología
18.
Cloning Stem Cells ; 10(3): 307-12, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18729764

RESUMEN

Sendai virus (SeV) vectors can introduce foreign genes efficiently and stably into primate embryonic stem (ES) cells. For the application of these cells, the control of transgene expression is important. Cynomolgus ES cells transduced with a SeV vector expressing the green fluorescent protein (GFP) gene were propagated in Knockout serum replacement (KSR)-supplemented medium, used widely for the serum-free culture of ES cells, and growth and transgene expression were evaluated. The SeV vector-mediated GFP expression was suppressed in the KSR-supplemented medium, although it was stable in regular fetal bovine serum (FBS)-supplemented medium. Propagation in the KSR-supplemented medium eventually resulted in a complete suppression of GFP expression and eradication of the SeV genome. The inhibitory effect of KSR on the transduction was attributable to the positive selection of untransduced ES cells in addition to the removal of the SeV vector from transduced cells. KSR also reduced the efficiency of the transduction. SeV vector-mediated transgene expression in ES cells was suppressed in the KSR-supplemented medium. Although the suppression is limited in specified cells such as ES cells, these findings will help elucidate how to control transgene expression.


Asunto(s)
Medios de Cultivo/química , Células Madre Embrionarias/fisiología , Vectores Genéticos , Macaca fascicularis , Virus Sendai/metabolismo , Transducción Genética , Animales , Bovinos , Células Cultivadas , Células Madre Embrionarias/citología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Ratones , Ratones Endogámicos BALB C , Virus Sendai/genética , Transgenes
19.
Cell Transplant ; 17(9): 1095-102, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19177845

RESUMEN

Embryonic stem (ES) cells have the ability to generate teratomas when transplanted into immunodeficient mice, but conditions affecting the generation remain to be elucidated. Nonhuman primate cynomolgus ES cells were transplanted into immunodeficient mice under different conditions; the number of transplanted cells, physical state (clumps or single dissociated cells), transplant site, differentiation state, and immunological state of recipient mice were all varied. The tumorigenicity was then evaluated. When cynomolgus ES cells were transplanted as clumps into the lower limb muscle in either nonobese diabetic/severe combined immunodeficiency (NOD/SCID) or NOD/SCID/gammac(null) (NOG) mice, teratomas developed in all the animals transplanted with 1 x 10(5) or more cells, but were not observed in any mouse transplanted with 1 x 10(5) cells. However, when the cells were transplanted as dissociated cells, the number of cells necessary for teratomas to form in all mice increased to 5 x 10(5). When the clump cells were injected subcutaneously (instead of intramuscularly), the number also increased to 5 x 10(5). When cynomolgus ES cell-derived progenitor cells (1 x 10(6)), which included residual pluripotent cells, were transplanted into the lower limb muscle of NOG or NOD/SCID mice, the incidence of teratomas differed between the strains; teratomas developed in five of five NOG mice but in only two of five NOD/SCID mice. The incidence of teratomas varied substantially depending on the transplanted cells and recipient mice. Thus, considerable care must be taken as to tumorigenicity.


Asunto(s)
Células Madre Embrionarias/trasplante , Trasplante de Células Madre/efectos adversos , Teratoma/etiología , Trasplante Heterólogo/efectos adversos , Animales , Células Madre Embrionarias/patología , Inyecciones Intramusculares , Inyecciones Subcutáneas , Macaca fascicularis , Ratones , Ratones Endogámicos NOD , Ratones SCID , Músculo Esquelético , Teratoma/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA