Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Genes Dev ; 26(13): 1445-58, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22713603

RESUMEN

The transcription factors Nanog and Gata6 are critical to specify the epiblast versus primitive endoderm (PrE) lineages. However, little is known about the mechanisms that regulate the protein stability and activity of these factors in the developing embryo. Here we uncover an early developmental function for the Polycomb group member Bmi1 in supporting PrE lineage formation through Gata6 protein stabilization. We show that Bmi1 is enriched in the extraembryonic (endoderm [XEN] and trophectodermal stem [TS]) compartment and repressed by Nanog in pluripotent embryonic stem (ES) cells. In vivo, Bmi1 overlaps with the nascent Gata6 and Nanog protein from the eight-cell stage onward before it preferentially cosegregates with Gata6 in PrE progenitors. Mechanistically, we demonstrate that Bmi1 interacts with Gata6 in a Ring finger-dependent manner to confer protection against Gata6 ubiquitination and proteasomal degradation. A direct role for Bmi1 in cell fate allocation is established by loss-of-function experiments in chimeric embryoid bodies. We thus propose a novel regulatory pathway by which Bmi1 action on Gata6 stability could alter the balance between Gata6 and Nanog protein levels to introduce a bias toward a PrE identity in a cell-autonomous manner.


Asunto(s)
Endodermo/metabolismo , Factor de Transcripción GATA6/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Represoras/metabolismo , Animales , Linaje de la Célula , Endodermo/citología , Factor de Transcripción GATA6/genética , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Ratones , Proteína Homeótica Nanog , Proteínas Nucleares/genética , Células Madre Pluripotentes/metabolismo , Complejo Represivo Polycomb 1 , Proteínas Proto-Oncogénicas/genética , Proteínas Represoras/genética , Transcripción Genética
2.
Arch Toxicol ; 92(10): 3117-3129, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30155720

RESUMEN

Liver disease is an escalating global health issue. While liver transplantation is an effective mode of therapy, patient mortality has increased due to the shortage of donor organs. Developing renewable sources of human liver tissue is therefore attractive. Pluripotent stem cell-derived liver tissue represents a potential alternative to cadaver derived hepatocytes and whole organ transplant. At present, two-dimensional differentiation procedures deliver tissue lacking certain functions and long-term stability. Efforts to overcome these limiting factors have led to the building of three-dimensional (3D) cellular aggregates. Although enabling for the field, their widespread application is limited due to their reliance on variable biological components. Our studies focused on the development of 3D liver tissue under defined conditions. In vitro generated 3D tissues exhibited stable phenotype for over 1 year in culture, providing an attractive resource for long-term in vitro studies. Moreover, 3D derived tissue provided critical liver support in two animal models, including immunocompetent recipients. Therefore, we believe that our study provides stable human tissue to better model liver biology 'in the dish', and in the future may permit the support of compromised liver function in humans.


Asunto(s)
Trasplante de Hígado/métodos , Hígado/citología , Células Madre Pluripotentes/citología , Ingeniería de Tejidos/métodos , Animales , Técnicas de Cultivo de Célula , Diferenciación Celular , Endodermo/citología , Femenino , Hepatectomía , Humanos , Hígado/fisiología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Células Madre Pluripotentes/fisiología , Esferoides Celulares/citología , Factores de Tiempo , Andamios del Tejido
3.
Cytometry A ; 89(5): 443-50, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27077783

RESUMEN

The clinical potential of multipotent mesenchymal stem cells (MSCs) has led to the essential development of analytical tools such as antibodies against membrane-bound proteins for the immunophenotypic characterization of human and rodent cells. Such tools are frequently lacking for emerging large animal models like the sheep that have greater relevance for the study of human musculoskeletal diseases. The present study identified a set of commercial nonspecies specific monoclonal antibodies for the immunophenotypic characterization of ovine MSCs. A protocol combining the less destructive proteolytic activity of accutase and EDTA was initially developed for the detachment of cells from plastic with minimum loss of cell surface antigens. A range of commercially available antibodies against human or rodent MSC antigens were then tested in single and multistain-based assays for their cross-reactivity to bone marrow derived ovine MSCs. Antibody clones cross-reactive to ovine CD73 (96.9% ± 5.9), CD90 (99.6% ± 0.3), CD105 (99.1 ± 1.5), CD271 (97.7 ± 2.0), and MHC1 (94.0% ± 7.2) antigens were identified using previously reported CD29, CD44, and CD166 as positive controls. Multistaining analysis indicated the colocalization of these antigens on MSCs. Furthermore, antibody clones identified to cross-react against white blood cell antigens exhibited either negative (CD117 (0.1% ± 0.1)) or low (MHCII (10.5% ± 16.0); CD31 (14.6% ± 4.2), and CD45 (39.4% ± 31.8)) cross-reactivity with ovine MSCs. The validation of these antibody clones to sheep MSC antigens is essential for studies utilizing this large animal model for stem cell-based therapies. © 2016 International Society for Advancement of Cytometry.


Asunto(s)
Anticuerpos Monoclonales/química , Células de la Médula Ósea/citología , Citometría de Flujo/métodos , Inmunofenotipificación/métodos , Células Madre Mesenquimatosas/citología , Adipocitos/citología , Adipocitos/inmunología , Animales , Anticuerpos Monoclonales/aislamiento & purificación , Antígenos CD/genética , Antígenos CD/inmunología , Biomarcadores/metabolismo , Células de la Médula Ósea/inmunología , Diferenciación Celular , Condrocitos/citología , Condrocitos/inmunología , Colagenasas/química , Reacciones Cruzadas , Ácido Edético/química , Expresión Génica , Humanos , Células Madre Mesenquimatosas/inmunología , Osteoblastos/citología , Osteoblastos/inmunología , Péptido Hidrolasas/química , Cultivo Primario de Células , Roedores , Ovinos
4.
Nucleic Acids Res ; 42(14): 9424-35, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25056318

RESUMEN

We have sequenced miRNA libraries from human embryonic, neural and foetal mesenchymal stem cells. We report that the majority of miRNA genes encode mature isomers that vary in size by one or more bases at the 3' and/or 5' end of the miRNA. Northern blotting for individual miRNAs showed that the proportions of isomiRs expressed by a single miRNA gene often differ between cell and tissue types. IsomiRs were readily co-immunoprecipitated with Argonaute proteins in vivo and were active in luciferase assays, indicating that they are functional. Bioinformatics analysis predicts substantial differences in targeting between miRNAs with minor 5' differences and in support of this we report that a 5' isomiR-9-1 gained the ability to inhibit the expression of DNMT3B and NCAM2 but lost the ability to inhibit CDH1 in vitro. This result was confirmed by the use of isomiR-specific sponges. Our analysis of the miRGator database indicates that a small percentage of human miRNA genes express isomiRs as the dominant transcript in certain cell types and analysis of miRBase shows that 5' isomiRs have replaced canonical miRNAs many times during evolution. This strongly indicates that isomiRs are of functional importance and have contributed to the evolution of miRNA genes.


Asunto(s)
MicroARNs/metabolismo , Animales , Proteínas Argonautas/metabolismo , Línea Celular , Evolución Molecular , Humanos , Ratones , MicroARNs/química , MicroARNs/genética , Precursores del ARN/química , ARN Mensajero/metabolismo , Células Madre/metabolismo
5.
FASEB J ; 28(2): 569-76, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24297703

RESUMEN

Transgenic technologies conventionally rely on the oocyte as a substrate for genetic modification. Owing to their accessibility, however, male germ cells, including mature sperm, have material advantages for use in transgenesis. Here we have exploited lentiviruses to generate transgenic animals via the male germline. When pseudotyped lentiviral vectors encoding green fluorescent protein (GFP) were incubated with mouse spermatozoa, these sperm were highly successful in producing transgenics. Lentivirally transduced mouse spermatozoa were used in in vitro fertilization (IVF) studies, and when followed by embryo transfer, ≥ 42% of founders were found to be transgenic for GFP. Inverse PCR strategy for integration site analysis demonstrated integration of at least 1 or 2 copies of GFP in the transgenics, mapping to different chromosomes. GFP expression was detected in a wide range of murine tissues, including testis and the transgene was stably transmitted to a third generation of transgenic animals. This relatively simple, yet highly efficient, technique for generating transgenic animals by transducing spermatozoa with lentiviral vectors in vitro is a powerful tool for the study of fertilization/preimplantation development, vertical viral gene transmission, gene function and regulation, and epigenetic inheritance.


Asunto(s)
Lentivirus/genética , Espermatozoides/metabolismo , Animales , Fertilización In Vitro , Células Germinativas/metabolismo , Masculino , Ratones , Ratones Transgénicos
6.
Development ; 137(15): 2483-92, 2010 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-20573702

RESUMEN

Pluripotent cells develop within the inner cell mass of blastocysts, a mosaic of cells surrounded by an extra-embryonic layer, the trophectoderm. We show that a set of somatic lineage regulators (including Hox, Gata and Sox factors) that carry bivalent chromatin enriched in H3K27me3 and H3K4me2 are selectively targeted by Suv39h1-mediated H3K9me3 and de novo DNA methylation in extra-embryonic versus embryonic (pluripotent) lineages, as assessed both in blastocyst-derived stem cells and in vivo. This stably repressed state is linked with a loss of gene priming for transcription through the exclusion of PRC1 (Ring1B) and RNA polymerase II complexes at bivalent, lineage-inappropriate genes upon trophoblast lineage commitment. Collectively, our results suggest a mutually exclusive role for Ring1B and Suv39h1 in regulating distinct chromatin states at key developmental genes and propose a novel mechanism by which lineage specification can be reinforced during early development.


Asunto(s)
Cromatina/química , Regulación del Desarrollo de la Expresión Génica , Metiltransferasas/fisiología , Proteínas Represoras/fisiología , Animales , Blastocisto , Linaje de la Célula , Cromatina/metabolismo , Metilación de ADN , Perfilación de la Expresión Génica , Silenciador del Gen , Metiltransferasas/metabolismo , Ratones , Modelos Biológicos , Complejo Represivo Polycomb 1 , Interferencia de ARN , ARN Polimerasa II/metabolismo , Proteínas Represoras/metabolismo , Trofoblastos/metabolismo , Ubiquitina-Proteína Ligasas
7.
Artículo en Inglés | MEDLINE | ID: mdl-29786563

RESUMEN

Liver transplantation represents the standard treatment for people with an end-stage liver disease and some liver-based metabolic disorders; however, shortage of liver donor tissues limits its availability. Furthermore, whole liver replacement eliminates the possibility of using native liver as a possible target for future gene therapy in case of liver-based metabolic defects. Cell therapy has emerged as a potential alternative, as cells can provide the hepatic functions and engraft in the liver parenchyma. Various options have been proposed, including human or other species hepatocytes, hepatocyte-like cells derived from stem cells or more futuristic alternatives, such as combination therapies with different cell types, organoids and cell-biomaterial combinations. In this review, we aim to give an overview of the cell therapies developed so far, highlighting preclinical and/or clinical achievements as well as the limitations that need to be overcome to make them fully effective and safe for clinical applications.This article is part of the theme issue 'Designer human tissue: coming to a lab near you'.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Hepatocitos/trasplante , Hepatopatías/terapia , Trasplante de Células Madre , Hepatocitos/fisiología , Humanos , Células Madre/fisiología
8.
Methods Mol Biol ; 1448: 95-106, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27317176

RESUMEN

Most transgenic technologies rely on the oocyte as a substrate for genetic modification. Transgenics animals are usually generated by the injection of the gene constructs (including lentiviruses encoding gene constructs or modified embryonic stem cells) into the pronucleus of a fertilized egg followed by the transfer of the injected embryos into the uterus of a foster mother. Male germ cells also have potential as templates for transgenic development. We have previously shown that mature sperm can be utilized as template for lentiviral transduction and as such used to generate transgenic mice efficiently with germ line capabilities. We provide here a detailed protocol that is relatively simple, to establish transgenic mice using lentivirally transduced spermatozoa. This protocol employs a well-established lentiviral gene delivery system (usual for somatic cells) delivering a variety of transgenes to be directly used with sperm, and the subsequent use of these modified sperm in in vitro fertilization studies and embryo transfer into foster female mice, for the establishment of transgenic mice.


Asunto(s)
Vectores Genéticos , Lentivirus/genética , Ratones Transgénicos/genética , Transducción Genética/métodos , Animales , Transferencia de Embrión/métodos , Células Madre Embrionarias/citología , Fertilización In Vitro/métodos , Técnicas de Transferencia de Gen , Masculino , Ratones , Oocitos/crecimiento & desarrollo , Espermatozoides/crecimiento & desarrollo
9.
Exp Hematol ; 30(12): 1428-35, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12482505

RESUMEN

Bone marrow from wild-type mice and mice with mutated Fas (lpr) or mutated Fas ligand (gld) was used to investigate the role of the Fas/FasL system in the regulation of myeloid progenitor cell kinetics.Granulocyte-macrophage colony-forming cells (CFU-GM) were measured by a standard colony assay and the proliferative activity of CFU-GM was measured by replating primary colonies and observing secondary colony formation. Fas expression was restored to lpr mouse bone marrow cells by retrovirus-mediated gene transfer and gld mouse marrow cells were treated with soluble FasL. Wild-type marrow cells were treated with YVAD (a caspase inhibitor) or anti-Fas monoclonal antibodies. There were greater frequencies of myeloid progenitor cells (CFU-GM) in lpr and gld mouse marrow compared to wild-type (WT) marrow (p = 0.0008). The proliferative capacity of CFU-GM was also significantly greater for lpr and gld CFU-GM compared to WT CFU-GM (p = 0.0003 and 0.0001, respectively). Retrovirus-mediated restoration of Fas into lpr marrow, and provision of soluble FasL (sFasL) to gld CFU-GM reduced CFU-GM proliferation to WT levels. Treatment of WT CFU-GM with YVAD or anti-FasL monoclonal antibody increased CFU-GM proliferation to the levels found in lpr and gld CFU-GM. YVAD significantly increased and anti-Fas significantly reduced the proliferative capacity of human CFU-GM (p = 0.015 and 0.04, respectively).Fas, FasL, and caspase activation may play an important role in regulating myeloid progenitor cell kinetics.


Asunto(s)
Apoptosis , Glicoproteínas de Membrana/metabolismo , Células Progenitoras Mieloides/citología , Receptor fas/metabolismo , Animales , Células de la Médula Ósea/citología , Caspasas/metabolismo , División Celular/efectos de los fármacos , Proteína Ligando Fas , Humanos , Cinética , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/farmacología , Ratones , Ratones Noqueados , Células Progenitoras Mieloides/metabolismo , Transducción Genética , Receptor fas/genética
10.
Cancer Res ; 74(5): 1319-28, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-24590807

RESUMEN

The high rate of glucose uptake to fuel the bioenergetic and anabolic demands of proliferating cancer cells is well recognized and is exploited with (18)F-2-fluoro-2-deoxy-d-glucose positron emission tomography ((18)F-FDG-PET) to image tumors clinically. In contrast, enhanced glucose storage as glycogen (glycogenesis) in cancer is less well understood and the availability of a noninvasive method to image glycogen in vivo could provide important biologic insights. Here, we demonstrate that (18)F-N-(methyl-(2-fluoroethyl)-1H-[1,2,3]triazole-4-yl)glucosamine ((18)F-NFTG) annotates glycogenesis in cancer cells and tumors in vivo, measured by PET. Specificity of glycogen labeling was demonstrated by isolating (18)F-NFTG-associated glycogen and with stable knockdown of glycogen synthase 1, which inhibited (18)F-NFTG uptake, whereas oncogene (Rab25) activation-associated glycogen synthesis led to increased uptake. We further show that the rate of glycogenesis is cell-cycle regulated, enhanced during the nonproliferative state of cancer cells. We demonstrate that glycogen levels, (18)F-NFTG, but not (18)F-FDG uptake, increase proportionally with cell density and G1-G0 arrest, with potential application in the assessment of activation of oncogenic pathways related to glycogenesis and the detection of posttreatment tumor quiescence.


Asunto(s)
Glucógeno/metabolismo , Neoplasias/diagnóstico , Neoplasias/metabolismo , Puntos de Control del Ciclo Celular/fisiología , Línea Celular Tumoral , Fluorodesoxiglucosa F18/metabolismo , Fase G1/fisiología , Humanos , Tomografía de Emisión de Positrones/métodos , Radiofármacos/metabolismo , Fase de Descanso del Ciclo Celular/fisiología , Proteínas de Unión al GTP rab/genética
11.
FEBS Open Bio ; 4: 266-75, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24918038

RESUMEN

Spermatozoa and lentiviruses are two of nature's most efficient gene delivery vehicles. Both can be genetically modified and used independently for the generation of transgenic animals or gene transfer/therapy of inherited disorders. Here we show that mature spermatozoa can be directly transduced with various pseudotyped lentiviral vectors and used in in vitro fertilisation studies. Lentiviral vectors encoding Green Fluorescent Protein (GFP) were shown to be efficiently processed and expressed in sperm. When these transduced sperm were used in in vitro fertilisation studies, GFP expression was observed in arising blastocysts. This simple technique of directly transducing spermatozoa has potential to be a powerful tool for the study of early and pre-implantation development and could be used as a technique in transgenic development and vertical viral transmission studies.

12.
Methods Mol Biol ; 891: 85-107, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22648769

RESUMEN

Vectors derived from the Retroviridae family have several attributes required for successful gene delivery. Retroviral vectors have an adequate payload size for the coding regions of most genes; they are safe to handle and simple to produce. These vectors can be manipulated to target different cell types with low immunogenicity and can permanently insert genetic information into the host cells' genome. Retroviral vectors have been used in gene therapy clinical trials and successfully applied experimentally in vitro, in vivo, and in utero.


Asunto(s)
Terapia Genética/métodos , Vectores Genéticos/biosíntesis , Vectores Genéticos/genética , Atención Prenatal/métodos , Retroviridae/genética , Ensayo de Inmunoadsorción Enzimática , Humanos , Reacción en Cadena de la Polimerasa , Volumetría
13.
Blood ; 104(9): 2697-703, 2004 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-15256424

RESUMEN

Gene therapy for a wide variety of disorders would be greatly enhanced by the development of vectors that could be targeted for gene delivery to specific populations of cells. We describe here high-efficiency targeted transduction based on a novel targeting strategy that exploits the ability of retroviruses to incorporate host cell proteins into the surface of the viral particle as they bud through the plasma membrane. Ecotropic retroviral particles produced in cells engineered to express the membrane-bound form of stem cell factor (mbSCF) transduce both human cell lines and primary cells with high efficiency in a strictly c-kit (SCF receptor)-dependent fashion. The availability of efficient targeted vectors provides a platform for the development of a new generation of therapies using in vivo gene delivery.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Células Madre Hematopoyéticas/metabolismo , Proteínas Proto-Oncogénicas c-kit , Retroviridae/genética , Transducción Genética/métodos , Proteínas de la Cápside , Línea Celular Tumoral , Proteínas Fluorescentes Verdes/genética , Células Madre Hematopoyéticas/química , Humanos , Ligandos , Ingeniería de Proteínas , Factor de Células Madre , Ensamble de Virus
14.
J Gene Med ; 6(11): 1189-96, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15468194

RESUMEN

BACKGROUND: The therapeutic potential of retroviruses can be significantly enhanced by display of specific molecules on the retroviral surface. This has been conventionally achieved by the manipulation of retroviral envelope proteins. In this report we have tested whether the natural budding mechanism of the retrovirus could be exploited to incorporate a specific molecule into the retroviral surface. METHODS: Retroviral packaging cells were engineered to express the membrane-bound form of human stem cell factor (mbSCF). Surface expression of mbSCF on retroviral packaging cells was confirmed by immunofluorescence and flow cytometry. Incorporation of mbSCF into retroviral particles was demonstrated by virus-binding assay and immunomagnetic capture of virus using antibody to SCF. Retroviral supernatants were tested for activity of the incorporated cytokine by proliferation assays on factor-dependent cells. Amphotropic retrovirus displaying surface mbSCF was used to transduce SCF receptor-positive haematopoietic cells. RESULTS: Retroviruses incorporating surface SCF showed increased levels of binding to cells (MO7e) expressing the SCF receptor, c-kit. mbSCF displayed on the viral surface retained levels of biological activity comparable with those of soluble recombinant growth factor. Transduction of c-kit-positive target cells with viruses displaying mbSCF showed enhanced levels of transduction in comparison with unmodified viruses. CONCLUSIONS: Expression of the membrane-bound form of human stem cell factor (mbSCF) on the surface of retroviral packaging cells allows its efficient incorporation into retrovirus particles in a biologically active form, opening up the possibility for the use of retroviral display in many therapeutic areas, such as in gene therapy, drug delivery and in the development of novel vaccines.


Asunto(s)
Retroviridae/metabolismo , Factor de Células Madre/biosíntesis , Proteínas del Envoltorio Viral/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Ratones , Proteínas Proto-Oncogénicas c-kit/biosíntesis , Retroviridae/genética , Factor de Células Madre/genética , Proteínas del Envoltorio Viral/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA