Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Bio Protoc ; 13(22): e4878, 2023 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-38023790

RESUMEN

The blastocysts consist of dozens of cells of three distinct lineages: epiblast (Epi), trophoblast (TB), and primitive endoderm (PrE). All embryonic and extraembryonic tissues are derived from Epi, TB, and PrE. Stem cell lines representing preimplantation Epi and TB have been established and are known as embryonic stem cells (ESCs) and trophoblast stem cells (TSCs). Extraembryonic endoderm cells (XENCs) constitute a cell line that has been established from PrE. Although in vivo, PrE gives rise to visceral endoderm (VE), parietal endoderm (PE), and marginal zone endoderm (MZE); XENCs, on blastocyst injection into chimeras, primarily contribute to the distal region of PE. Here, we provide a comprehensive protocol for the establishment of fully potent primitive endoderm stem cell (PrESC) lines. PrESCs are established and maintained on mouse embryonic fibroblast (MEF) feeder cells in a serum-free medium supplemented with fibroblast growth factor 4 (FGF4), heparin, CHIR99021, and platelet-derived growth factor-AA (PDGF-AA). PrESCs co-express markers indicative of pluripotency and endoderm lineage commitment, exhibiting characteristics akin to those of PrE. On transplantation of PrESCs into blastocysts, they demonstrate a high efficiency in contributing to VE, PE, and MZE. PrESCs serve as a valuable model for studying PrE, sharing similarities in gene expression profiles and differentiation potential. PrESCs constitute a pivotal cornerstone for in vitro analysis of early developmental mechanisms and for studies of embryo reconstitution in vitro, particularly in conjunction with ESCs and TSCs. Key features • Establishment and maintenance of primitive endoderm stem cell (PrESCs) capable of recapitulating the developmental prowess inherent to PrE. • Offering a source of PrE lineage for embryo-like organoid reconstitution studies.

2.
Methods Mol Biol ; 2529: 207-228, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35733017

RESUMEN

Among the most important histone methyltransferases for metazoan development are EZH1/2 and their homologs, which methylate histone H3 lysine 27 and act as part of a highly conserved set of chromatin regulators called Polycomb Group (PcG) proteins. Reaching a precise understanding of the roles of PcG proteins in the orchestration of differentiation and the maintenance of cell identity requires a variety of genetic and molecular approaches. Here, we present a full suite of methods for the study of PcG proteins in early murine development, including mutant strain generation, embryonic stem cell derivation, epigenomic profiling, and immunofluorescence and in situ hybridization.


Asunto(s)
Cromatina , Epigenómica , Animales , Diferenciación Celular/genética , Cromatina/genética , Epigénesis Genética , Ratones , Proteínas del Grupo Polycomb/genética
3.
Science ; 375(6580): 574-578, 2022 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-35113719

RESUMEN

The mammalian blastocyst consists of three distinct cell types: epiblast, trophoblast (TB), and primitive endoderm (PrE). Although embryonic stem cells (ESCs) and trophoblast stem cells (TSCs) retain the functional properties of epiblast and TB, respectively, stem cells that fully recapitulate the developmental potential of PrE have not been established. Here, we report derivation of primitive endoderm stem cells (PrESCs) in mice. PrESCs recapitulate properties of embryonic day 4.5 founder PrE, are efficiently incorporated into PrE upon blastocyst injection, generate functionally competent PrE-derived tissues, and support fetal development of PrE-depleted blastocysts in chimeras. Furthermore, PrESCs can establish interactions with ESCs and TSCs and generate descendants with yolk sac-like structures in utero. Establishment of PrESCs will enable the elucidation of the mechanisms for PrE specification and subsequent pre- and postimplantation development.


Asunto(s)
Células Madre Embrionarias/citología , Células Madre Embrionarias/fisiología , Endodermo/citología , Endodermo/embriología , Animales , Blastocisto/citología , Blastocisto/fisiología , Diferenciación Celular , Línea Celular , Linaje de la Célula , Quimera , Desarrollo Embrionario , Endodermo/crecimiento & desarrollo , Desarrollo Fetal , Estratos Germinativos/citología , Estratos Germinativos/embriología , Ratones , Ratones Endogámicos C57BL , Trofoblastos/citología , Trofoblastos/fisiología
4.
Nat Commun ; 12(1): 5341, 2021 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-34504070

RESUMEN

Polycomb repressive complexes-1 and -2 (PRC1 and 2) silence developmental genes in a spatiotemporal manner during embryogenesis. How Polycomb group (PcG) proteins orchestrate down-regulation of target genes upon differentiation, however, remains elusive. Here, by differentiating embryonic stem cells into embryoid bodies, we reveal a crucial role for the PCGF1-containing variant PRC1 complex (PCGF1-PRC1) to mediate differentiation-associated down-regulation of a group of genes. Upon differentiation cues, transcription is down-regulated at these genes, in association with PCGF1-PRC1-mediated deposition of histone H2AK119 mono-ubiquitination (H2AK119ub1) and PRC2 recruitment. In the absence of PCGF1-PRC1, both H2AK119ub1 deposition and PRC2 recruitment are disrupted, leading to aberrant expression of target genes. PCGF1-PRC1 is, therefore, required for initiation and consolidation of PcG-mediated gene repression during differentiation.


Asunto(s)
Cuerpos Embrioides/metabolismo , Regulación del Desarrollo de la Expresión Génica , Histonas/genética , Células Madre Embrionarias de Ratones/metabolismo , Complejo Represivo Polycomb 1/genética , Complejo Represivo Polycomb 2/genética , Animales , Diferenciación Celular , Embrión de Mamíferos , Cuerpos Embrioides/citología , Histonas/metabolismo , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , Ratones Transgénicos , Células Madre Embrionarias de Ratones/citología , Factor de Crecimiento Derivado de Plaquetas/genética , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Complejo Represivo Polycomb 1/metabolismo , Complejo Represivo Polycomb 2/metabolismo , Cultivo Primario de Células , Factores de Transcripción SOXC/genética , Factores de Transcripción SOXC/metabolismo , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcripción Genética , Ubiquitinación
5.
Sci Adv ; 3(5): e1602179, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28508054

RESUMEN

In mammals, the Y chromosome strictly influences the maintenance of male germ cells. Almost all mammalian species require genetic contributors to generate testes. An endangered species, Tokudaia osimensis, has a unique sex chromosome composition XO/XO, and genetic differences between males and females have not been confirmed. Although a distinctive sex-determining mechanism may exist in T. osimensis, it has been difficult to examine thoroughly in this rare animal species. To elucidate the discriminative sex-determining mechanism in T. osimensis and to find a strategy to prevent its possible extinction, we have established induced pluripotent stem cells (iPSCs) and derived interspecific chimeras using mice as the hosts and recipients. Generated iPSCs are considered to be in the so-called "true naïve" state, and T. osimensis iPSCs may contribute as interspecific chimeras to several different tissues and cells in live animals. Surprisingly, female T. osimensis iPSCs not only contributed to the female germ line in the interspecific mouse ovary but also differentiated into spermatocytes and spermatids that survived in the adult interspecific mouse testes. Thus, T. osimensis cells have high sexual plasticity through which female somatic cells can be converted to male germline cells. These findings suggest flexibility in T. osimensis cells, which can adapt their germ cell sex to the gonadal niche. The probable reduction of the extinction risk of an endangered species through the use of iPSCs is indicated by this study.


Asunto(s)
Cromosomas de los Mamíferos , Especies en Peligro de Extinción , Células Germinativas/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Murinae , Procesos de Determinación del Sexo/genética , Testículo/metabolismo , Cromosoma X , Animales , Cromosomas de los Mamíferos/genética , Cromosomas de los Mamíferos/metabolismo , Femenino , Células Germinativas/citología , Masculino , Murinae/genética , Murinae/metabolismo , Testículo/citología , Cromosoma X/genética , Cromosoma X/metabolismo
6.
Biol Reprod ; 94(6): 122, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27122635

RESUMEN

Mouse trophoblast stem cells (TSCs) proliferate indefinitely in vitro, despite their highly heterogeneous nature. In this study, we sought to characterize TSC colony types by using methods based on cell biology and biochemistry for a better understanding of how TSCs are maintained over multiple passages. Colonies of TSCs could be classified into four major types: type 1 is compact and dome-shaped, type 4 is flattened but with a large multilayered cell cluster, and types 2 and 3 are their intermediates. A time-lapse analysis indicated that type 1 colonies predominantly appeared after passaging, and a single type 1 colony gave rise to all other types. These colony transitions were irreversible, but at least some type 1 colonies persisted throughout culture. The typical cells comprising type 1 colonies were small and highly motile, and they aggregated together to form primary colonies. A hierarchical clustering based on global gene expression profiles suggested that a TSC line containing more type 1 colony cells was similar to in vivo extraembryonic tissues. Among the known TSC genes examined, Elf5 showed a differential expression pattern according to colony type, indicating that this gene might be a reliable marker of undifferentiated TSCs. When aggregated with fertilized embryos, cells from types 1 and 2, but not from type 4, distributed to the polar trophectoderm in blastocysts. These findings indicate that cells typically found in type 1 colonies can persist indefinitely as stem cells and are responsible for the maintenance of TSC lines. They may provide key information for future improvements in the quality of TSC lines.


Asunto(s)
Células Madre/citología , Trofoblastos/citología , Animales , Diferenciación Celular , Línea Celular , Proteínas de Unión al ADN/metabolismo , Femenino , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Células Madre/metabolismo , Factores de Transcripción/metabolismo , Trofoblastos/metabolismo
7.
Sci Rep ; 6: 23808, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-27033801

RESUMEN

Cloning animals by nuclear transfer provides the opportunity to preserve endangered mammalian species. However, there are risks associated with the collection of donor cells from the body such as accidental injury to or death of the animal. Here, we report the production of cloned mice from urine-derived cells collected noninvasively. Most of the urine-derived cells survived and were available as donors for nuclear transfer without any pretreatment. After nuclear transfer, 38-77% of the reconstructed embryos developed to the morula/blastocyst, in which the cell numbers in the inner cell mass and trophectoderm were similar to those of controls. Male and female cloned mice were delivered from cloned embryos transferred to recipient females, and these cloned animals grew to adulthood and delivered pups naturally when mated with each other. The results suggest that these cloned mice had normal fertility. In additional experiments, 26 nuclear transfer embryonic stem cell lines were established from 108 cloned blastocysts derived from four mouse strains including inbreds and F1 hybrids with relatively high success rates. Thus, cells derived from urine, which can be collected noninvasively, may be used in the rescue of endangered mammalian species by using nuclear transfer without causing injury to the animal.


Asunto(s)
Clonación de Organismos/métodos , Técnicas de Transferencia Nuclear , Orina/citología , Animales , Blastocisto/citología , Línea Celular , Transferencia de Embrión , Células Madre Embrionarias/citología , Femenino , Fertilidad , Proteínas Fluorescentes Verdes , Masculino , Ratones , Ratones Endogámicos , Ratones Transgénicos
8.
PLoS One ; 9(9): e107308, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25203285

RESUMEN

The inner cell mass (ICM) and trophoblast cell lineages duet early embryonic development in mammals. After implantation, the ICM forms the embryo proper as well as some extraembryonic tissues, whereas the trophoectoderm (TE) exclusively forms the fetal portion of the placenta and the trophoblast giant cells. Although embryonic stem (ES) cells can be derived from ICM in cultures of mouse blastocysts in the presence of LIF and/or combinations of small-molecule chemical compounds, and the undifferentiated pluripotent state can be stably maintained without use of serum and feeder cells, defined culture conditions for derivation and maintenance of undifferentiated trophoblast stem (TS) cells have not been established. Here, we report that addition of FGF2, activin A, XAV939, and Y27632 are necessary and sufficient for derivation of TS cells from both of E3.5 blastocysts and E6.5 early postimplantation extraembryonic ectoderm. Moreover, the undifferentiated TS cell state can be stably maintained in chemically defined culture conditions. Cells derived in this manner expressed TS cell marker genes, including Eomes, Elf5, Cdx2, Klf5, Cdh1, Esrrb, Sox2, and Tcfap2c; differentiated into all trophoblast subtypes (trophoblast giant cells, spongiotrophoblast, and labyrinthine trophoblast) in vitro; and exclusively contributed to trophoblast lineages in chimeric animals. This delineation of minimal requirements for derivation and self-renewal provides a defined platform for precise description and dissection of the molecular state of TS cells.


Asunto(s)
Medios de Cultivo/metabolismo , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/fisiología , Trofoblastos/efectos de los fármacos , Trofoblastos/fisiología , Activinas/farmacología , Amidas/farmacología , Animales , Blastocisto/efectos de los fármacos , Blastocisto/metabolismo , Blastocisto/fisiología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Linaje de la Célula/efectos de los fármacos , Linaje de la Célula/fisiología , Ectodermo/efectos de los fármacos , Ectodermo/metabolismo , Ectodermo/fisiología , Implantación del Embrión/efectos de los fármacos , Implantación del Embrión/fisiología , Desarrollo Embrionario/efectos de los fármacos , Desarrollo Embrionario/fisiología , Células Madre Embrionarias/metabolismo , Femenino , Factor 2 de Crecimiento de Fibroblastos/farmacología , Compuestos Heterocíclicos con 3 Anillos/farmacología , Humanos , Ratones , Placenta/metabolismo , Placenta/fisiología , Embarazo , Piridinas/farmacología , Trofoblastos/metabolismo
9.
PLoS One ; 9(8): e104957, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25126862

RESUMEN

Viral vectors have been used for hemophilia A gene therapy. However, due to its large size, full-length Factor VIII (FVIII) cDNA has not been successfully delivered using conventional viral vectors. Moreover, viral vectors may pose safety risks, e.g., adverse immunological reactions or virus-mediated cytotoxicity. Here, we took advantages of the non-viral vector gene delivery system based on piggyBac DNA transposon to transfer the full-length FVIII cDNA, for the purpose of treating hemophilia A. We tested the efficiency of this new vector system in human 293T cells and iPS cells, and confirmed the expression of the full-length FVIII in culture media using activity-sensitive coagulation assays. Hydrodynamic injection of the piggyBac vectors into hemophilia A mice temporally treated with an immunosuppressant resulted in stable production of circulating FVIII for over 300 days without development of anti-FVIII antibodies. Furthermore, tail-clip assay revealed significant improvement of blood coagulation time in the treated mice. piggyBac transposon vectors can facilitate the long-term expression of therapeutic transgenes in vitro and in vivo. This novel gene transfer strategy should provide safe and efficient delivery of FVIII.


Asunto(s)
ADN Complementario/uso terapéutico , Factor VIII/genética , Vectores Genéticos/uso terapéutico , Hemofilia A/terapia , Animales , Elementos Transponibles de ADN , ADN Complementario/administración & dosificación , ADN Complementario/genética , Modelos Animales de Enfermedad , Factor VIII/análisis , Expresión Génica , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Células HEK293 , Hemofilia A/sangre , Hemofilia A/genética , Humanos , Ratones
10.
PLoS One ; 9(4): e95329, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24736627

RESUMEN

Embryonic stem cells (ESCs) can contribute to the tissues of chimeric animals, including the germline. By contrast, epiblast stem cells (EpiSCs) barely contribute to chimeras. These two types of cells are established and maintained under different culture conditions. Here, we show that a modified EpiSC culture condition containing the GSK3 inhibitor CHIR99021 can support a germline-competent pluripotent state that is intermediate between ESCs and EpiSCs. When ESCs were cultured under a modified condition containing bFGF, Activin A, and CHIR99021, they converted to intermediate pluripotent stem cells (INTPSCs). These INTPSCs were able to form teratomas in vivo and contribute to chimeras by blastocyst injection. We also induced formation of INTPSCs (iINTPSCs) from mouse embryonic fibroblasts by exogenous expression of four reprogramming factors, Oct3/4, Sox2, Klf4, and c-Myc, under the INTPSC culture condition. These iINTPSCs contributed efficiently to chimeras, including the germline, by blastocyst injection. The INTPSCs exhibited several characteristic properties of both ESCs and EpiSCs. Our results suggest that the modified EpiSC culture condition can support growth of cells that meet the most stringent criteria for pluripotency, and that germline-competent pluripotency may depend on the activation state of Wnt signaling.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Estratos Germinativos/citología , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Óvulo/citología , Células Madre Pluripotentes/citología , Inhibidores de Proteínas Quinasas/farmacología , Espermatozoides/citología , Animales , Estratos Germinativos/efectos de los fármacos , Factor 4 Similar a Kruppel , Masculino , Ratones , Piridinas/farmacología , Pirimidinas/farmacología , Seguridad
11.
PLoS One ; 9(3): e92973, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24667806

RESUMEN

The most stringent criterion for evaluating pluripotency is generation of chimeric animals with germline transmission ability. Because the quality of induced pluripotent stem cell (iPSC) lines is heterogeneous, an easy and accurate system to evaluate these abilities would be useful. In this study, we describe a simple but comprehensive system for generating and evaluating iPSCs by single transfection of multiple piggyBac (PB) plasmid vectors encoding Tet-inducible polycistronic reprogramming factors, a pluripotent-cell-specific reporter, a constitutively active reporter, and a sperm-specific reporter. Using this system, we reprogrammed 129 and NOD mouse embryonic fibroblasts into iPSCs, and then evaluated the molecular and functional properties of the resultant iPSCs by quantitative RT-PCR analysis and chimera formation assays. The iPSCs contributed extensively to chimeras, as indicated by the constitutively active TagRFP reporter, and also differentiated into sperm, as indicated by the late-spermatogenesis-specific Acr (acrosin)-EGFP reporter. Next, we established secondary MEFs from E13.5 chimeric embryos and efficiently generated secondary iPSCs by simple addition of doxycycline. Finally, we applied this system to establishment and evaluation of rat iPSCs and production of rat sperm in mouse-rat interspecific chimeras. By monitoring the fluorescence of Acr-EGFP reporter, we could easily detect seminiferous tubules containing rat iPSC-derived spermatids and sperm. And, we succeeded to obtain viable offspring by intracytoplasmic sperm injection (ICSI) using these haploid male germ cells. We propose that this system will enable robust strategies for induction and evaluation of iPSCs, not only in rodents but also in other mammals. Such strategies will be especially valuable in non-rodent species, in which verification of germline transmission by mating is inefficient and time-consuming.


Asunto(s)
Reprogramación Celular/genética , Vectores Genéticos/genética , Células Madre Pluripotentes Inducidas/citología , Transfección/métodos , Animales , Masculino , Ratones , Plásmidos/genética , Ratas , Espermátides/citología , Espermátides/metabolismo
12.
Genes Cells ; 16(7): 815-25, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21658168

RESUMEN

PiggyBac (PB) transposition of reprogramming factors (Oct3/4 (O), Sox2 (S), Klf4 (K) and c-Myc) is a safe, nonviral method for generating induced pluripotent stem cells (iPSCs). However, compared with retroviral methods, the reprogramming efficiency of the PB-mediated methods is relatively low. In this study, we describe a simple and efficient system for generating high-quality iPSCs by a single transfection of multiple plasmids that does not require the use of a virus, special instruments or skilled techniques. To improve reprogramming efficiency, we modified the components of the polycistronic 2A vectors used in this study and also investigated the combination of another reprogramming-related factor (L-Myc). By simultaneous transposition of multiple PB vectors containing an EOS (early transposon promoter and Oct3/4 and Sox2 enhancers) reporter and modified polycistronic doxycycline (Dox)-inducible factors, we reprogrammed mouse somatic cells with an efficiency higher than is usually obtained with retroviral methods and we established some iPSC lines that contributed highly to chimeras. By using the Dox-inducible system, we also showed that the appropriate elimination of exogenous-factor expression at appropriate time accelerated the induction of Oct3/4 when a combination of OKS and c-Myc vectors were used.


Asunto(s)
Elementos Transponibles de ADN/genética , Doxiciclina/farmacología , Genes Reporteros/genética , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Animales , Diferenciación Celular , Reprogramación Celular/genética , Vectores Genéticos/genética , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Factor 4 Similar a Kruppel , Ratones , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Regiones Promotoras Genéticas/genética , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo
13.
Biol Reprod ; 81(6): 1147-53, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19726737

RESUMEN

The nonobese diabetic (NOD) mouse is a valuable model for human type 1 diabetes and the development of humanized mice. Although the importance of this mouse strain is widely recognized, its usefulness is constrained by the absence of NOD embryonic stem (ES) lines with adequate germline transmission competence. In the present study, we established two germline transmission-competent types of cell lines from NOD mice; these cell lines, male germline stem (GS) cells and ES cells, were derived from NOD spermatogonia and blastocysts, respectively. NOD-GS cells proliferated in vitro and differentiated into mature sperm after transplantation into testis. NOD-ES cell lines were effectively established from NOD blastocysts using culture medium containing inhibitors for fibroblast growth receptor, MEK, and GSK3. Both the NOD-GS and NOD-ES cell lines transmitted their haplotypes to progeny, revealing a novel strategy for gene modification in a pure NOD genetic background. Our results also suggest that the establishment of GS cells is an effective procedure in nonpermissive mouse strains or other species for ES cell derivation.


Asunto(s)
Proliferación Celular , Células Madre Embrionarias/citología , Espermatogénesis/fisiología , Espermatozoides/citología , Animales , Blastocisto/citología , Técnicas de Cultivo de Célula , Línea Celular , Femenino , Técnica del Anticuerpo Fluorescente , Genotipo , Inseminación Artificial , Masculino , Metilación , Ratones , Ratones Endogámicos NOD , Trasplante de Células Madre
14.
Cell ; 137(3): 571-84, 2009 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-19410550

RESUMEN

Specification of the germ cell lineage is vital to development and heredity. In mice, the germ cell fate is induced in pluripotent epiblast cells by signaling molecules, yet the underlying mechanism remains unknown. Here we demonstrate that germ cell fate in the epiblast is a direct consequence of Bmp4 signaling from the extraembryonic ectoderm (ExE), which is antagonized by the anterior visceral endoderm (AVE). Strikingly, Bmp8b from the ExE restricts AVE development, thereby contributing to Bmp4 signaling. Furthermore, Wnt3 in the epiblast ensures its responsiveness to Bmp4. Serum-free, defined cultures revealed that, in response to Bmp4, competent epiblast cells uniformly expressed key transcriptional regulators Blimp1 and Prdm14 and acquired germ-cell properties, including genome-wide epigenetic reprogramming, in an orderly fashion. Notably, the induced cells contributed to both spermatogenesis and fertility of offspring. By identifying a signaling principle in germ cell specification, our study establishes a robust strategy for reconstituting the mammalian germ cell lineage in vitro.


Asunto(s)
Proteína Morfogenética Ósea 4/fisiología , Linaje de la Célula/fisiología , Células Germinativas , Transducción de Señal/fisiología , Animales , Proteínas Morfogenéticas Óseas/fisiología , Diferenciación Celular/fisiología , Proteínas de Unión al ADN , Embrión de Mamíferos/citología , Embrión de Mamíferos/embriología , Embrión de Mamíferos/fisiología , Células Germinativas/citología , Células Germinativas/fisiología , Masculino , Mesodermo/citología , Mesodermo/fisiología , Ratones , Ratones Noqueados , Ratones Transgénicos , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Proteínas de Unión al ARN , Células Madre/citología , Células Madre/fisiología , Testículo/citología , Testículo/fisiología , Factores de Transcripción/fisiología , Proteínas Wnt/fisiología
15.
Nat Genet ; 40(8): 1016-22, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18622394

RESUMEN

Specification of germ cell fate is fundamental in development and heredity. Recent evidence indicates that in mice, specification of primordial germ cells (PGCs), the common source of both oocytes and spermatozoa, occurs through the integration of three key events: repression of the somatic program, reacquisition of potential pluripotency and ensuing genome-wide epigenetic reprogramming. Here we provide genetic evidence that Prdm14, a PR domain-containing transcriptional regulator with exclusive expression in the germ cell lineage and pluripotent cell lines, is critical in two of these events, the reacquisition of potential pluripotency and successful epigenetic reprogramming. In Prdm14 mutants, the failure of these two events manifests even in the presence of Prdm1 (also known as Blimp1), a key transcriptional regulator for PGC specification. Our combined evidence demonstrates that Prdm14 defines a previously unknown genetic pathway, initiating independently from Prdm1, for ensuring the launching of the mammalian germ cell lineage.


Asunto(s)
Células Germinativas/citología , Factores de Transcripción/metabolismo , Animales , Linaje de la Célula , Proteínas de Unión al ADN , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Femenino , Redes Reguladoras de Genes , Células Germinativas/metabolismo , Masculino , Ratones , Proteínas de Unión al ARN , Factores de Transcripción/genética
16.
Genes Dev ; 22(12): 1617-35, 2008 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-18559478

RESUMEN

Specification of germ cell fate is fundamental in development. With a highly representative single-cell microarray and rigorous quantitative PCR analysis, we defined the genome-wide transcription dynamics that create primordial germ cells (PGCs) from the epiblast, a process that exclusively segregates them from their somatic neighbors. We also analyzed the effect of the loss of Blimp1, a key transcriptional regulator, on these dynamics. Our analysis revealed that PGC specification involves complex, yet highly ordered regulation of a large number of genes, proceeding under the strong influence of mesoderm induction but specifically avoiding developmental programs such as the epithelial-mesenchymal transition, Hox cluster activation, cell cycle progression, and DNA methyltransferase machinery. Remarkably, Blimp1 is essential for repressing nearly all the genes normally down-regulated in PGCs relative to their somatic neighbors. In contrast, it is dispensable for the activation of approximately half of the genes up-regulated in PGCs, uncovering the Blimp1-independent events for PGC specification. Notably, however, highly PGC-specific genes exhibited distinct correlations to Blimp1 in wild-type embryos, and these correlations faithfully predicted their expression impairments in Blimp1 mutants. Moreover, their expression overlaps within single cells were severely damaged without Blimp1, demonstrating that Blimp1 exerts positive influence on their concerted activation. Thus, Blimp1 is not a single initiator but a dominant coordinator of the transcriptional program for the establishment of the germ cell fate in mice.


Asunto(s)
Tipificación del Cuerpo/genética , Linaje de la Célula/genética , Regulación del Desarrollo de la Expresión Génica , Factores de Transcripción/fisiología , Animales , Análisis por Conglomerados , Embrión de Mamíferos , Perfilación de la Expresión Génica , Genes del Desarrollo , Genoma , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Transducción de Señal/genética , Transcripción Genética
17.
Reproduction ; 136(4): 503-14, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18583473

RESUMEN

The ability to monitor the development of a given cell lineage in a non-invasive manner by fluorescent markers both in vivo and in vitro provides a great advantage for the analysis of the lineage of interest. To date, a number of transgenic or knock-in mouse strains, in which developing germ cells are marked with fluorescent reporters, have been generated. We here describe a novel double transgenic reporter mouse strain that expresses membrane-targeted Venus (mVenus), a brighter variant of yellow fluorescent protein (YFP), under the control of Prdm1 (Blimp1) regulatory elements and enhanced cyan fluorescent protein (ECFP) under the control of Dppa3 (Stella/Pgc7). The double transgenic strain unambiguously marked Prdm1 expression in the lineage-restricted precursors of primordial germ cells (PGCs) in the proximal epiblast at embryonic day (E) 6.25 and specifically illuminated Prdm1- and Dppa3-positive migrating PGCs after E8.5. The double transgenic reporter also precisely recapitulated dynamic embryonic expression of Prdm1 outside the germ cell lineage. Moreover, we derived ES cells that bore both transgenes. These cells made a robust contribution both to the germ and somatic cell lineages in chimeras with accurate Prdm1-mVenus and Dppa3-ECFP expression. The transgenic strain and the ES cells will serve as valuable experimental materials not only for analyzing the origin and properties of the germ cell lineage in vivo, but also for establishing a culture system to efficiently induce proper germ cells with temporally coordinated Prdm1 and Dppa3 expression in vitro.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Células Germinativas/citología , Proteínas Represoras/genética , Factores de Transcripción/genética , Animales , Células Cultivadas , Proteínas Cromosómicas no Histona , Técnica del Anticuerpo Fluorescente , Expresión Génica , Proteínas Fluorescentes Verdes , Hibridación Fluorescente in Situ , Proteínas Luminiscentes , Ratones , Ratones Transgénicos , Modelos Animales , Factor 1 de Unión al Dominio 1 de Regulación Positiva
18.
Nat Protoc ; 2(3): 739-52, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17406636

RESUMEN

We describe here a protocol for the representative amplification of global mRNAs from typical single mammalian cells to provide a template for high-density oligonucleotide microarray analysis. A single cell is lysed in a tube without purification and first-strand cDNAs are synthesized using a poly(dT)-tailed primer. Unreacted primer is specifically eliminated by exonuclease treatment and second strands are generated with a second poly(dT)-tailed primer after poly(dA) tailing of the first-strand cDNAs. The cDNAs are split into four tubes, which are independently directionally amplified by PCR, and then recombined. The amplified products (approximately 100 ng) show superior representation and reproducibility of original gene expression, especially for genes expressed in more than 20 copies per cell, compared with those obtained by a conventional PCR protocol, and can effectively be used for quantitative PCR and EST analyses. The cDNAs are then subjected to another PCR amplification with primers bearing the T7 promoter sequence. The resultant cDNA products are gel purified, amplified by one final cycle and used for isothermal linear amplification by T7 RNA polymerase to synthesize cRNAs for microarray hybridization. This protocol yields cDNA templates sufficient for more than 80 microarray hybridizations from a single cell, and can be completed in 5-6 days.


Asunto(s)
ADN Complementario/genética , Perfilación de la Expresión Génica/métodos , Técnicas de Amplificación de Ácido Nucleico/métodos , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , ARN Mensajero/genética , Animales , Cartilla de ADN , Ratones , Reacción en Cadena de la Polimerasa
20.
Cell ; 126(3): 597-609, 2006 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-16901790

RESUMEN

Epidermal lineage commitment occurs when multipotent stem cells are specified to three lineages: the epidermis, the hair follicle, and the sebaceous gland (SG). How and when a lineage becomes specified remains unknown. Here, we report the existence of a population of unipotent progenitor cells that reside in the SG and express the transcriptional repressor Blimp1. Using cell-culture studies and genetic lineage tracing, we demonstrate that Blimp1-expressing cells are upstream from other cells of the SG lineage. Blimp1 appears to govern cellular input into the gland since its loss leads to elevated c-myc expression, augmented cell proliferation, and SG hyperplasia. Finally, BrdU labeling experiments demonstrate that the SG defects associated with loss of Blimp1 lead to enhanced bulge stem cell activity, suggesting that when normal SG homeostasis is perturbed, multipotent stem cells in the bulge can be mobilized to correct this imbalance.


Asunto(s)
Diferenciación Celular/genética , Linaje de la Célula/genética , Células Epiteliales/metabolismo , Proteínas Represoras/metabolismo , Glándulas Sebáceas/embriología , Glándulas Sebáceas/crecimiento & desarrollo , Células Madre/metabolismo , Factores de Transcripción/metabolismo , Animales , Animales Recién Nacidos , Bromodesoxiuridina , Recuento de Células , Movimiento Celular/genética , Proliferación Celular , Células Cultivadas , Células Epiteliales/citología , Regulación del Desarrollo de la Expresión Génica/genética , Folículo Piloso/citología , Folículo Piloso/embriología , Folículo Piloso/crecimiento & desarrollo , Hiperplasia/genética , Hiperplasia/metabolismo , Hiperplasia/fisiopatología , Ratones , Ratones Noqueados , Ratones Transgénicos , Células Madre Multipotentes/citología , Células Madre Multipotentes/metabolismo , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas Represoras/genética , Glándulas Sebáceas/citología , Células Madre/citología , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...