Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Development ; 147(22)2020 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-33033120

RESUMEN

The path from a fertilised egg to an embryo involves the coordinated formation of cell types, tissues and organs. Developmental modules comprise discrete units specified by self-sufficient genetic programs that can interact with each other during embryogenesis. Here, we have taken advantage of the different span of embryonic development between two distantly related teleosts, zebrafish (Danio rerio) and medaka (Oryzias latipes) (3 and 9 days, respectively), to explore modularity principles. We report that inter-species blastula transplantations result in the ectopic formation of a retina formed by donor cells - a module. We show that the time taken for the retina to develop follows a genetic program: an ectopic zebrafish retina in medaka develops with zebrafish dynamics. Heterologous transplantation results in a temporal decoupling between the donor retina and host organism, illustrated by two paradigms that require retina-host interactions: lens recruitment and retino-tectal projections. Our results uncover a new experimental system for addressing temporal decoupling along embryonic development, and highlight the presence of largely autonomous but interconnected developmental modules that orchestrate organogenesis.


Asunto(s)
Blástula , Oryzias/embriología , Retina/embriología , Quimera por Trasplante/embriología , Pez Cebra/embriología , Animales , Blástula/embriología , Blástula/trasplante , Xenoinjertos , Retina/citología
2.
Curr Biol ; 30(21): 4227-4237.e5, 2020 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-32888487

RESUMEN

A hallmark of the evolutionary expansion of the neocortex is a specific increase in the number of neurons generated for the upper neocortical layers during development. The cause underlying this increase is unknown. Here, we show that lengthening the neurogenic period during neocortical development is sufficient to specifically increase upper-layer neuron generation. Thus, embryos of mouse strains with longer gestation exhibited a longer neurogenic period and generated more upper-layer, but not more deep-layer, neurons than embryos with shorter gestation. Accordingly, long-gestation embryos showed a greater abundance of neurogenic progenitors in the subventricular zone than short-gestation embryos at late stages of cortical neurogenesis. Analysis of a mouse-rat chimeric embryo, developing inside a rat mother, pointed to factors in the rat environment that influenced the upper-layer neuron generation by the mouse progenitors. Exploring a potential maternal source of such factors, short-gestation strain mouse embryos transferred to long-gestation strain mothers exhibited an increase in the length of the neurogenic period and upper-layer neuron generation. The opposite was the case for long-gestation strain mouse embryos transferred to short-gestation strain mothers, indicating a dominant maternal influence on the length of the neurogenic period and hence upper-layer neuron generation. In summary, our study uncovers a hitherto unknown link between embryonic cortical neurogenesis and the maternal gestational environment and provides experimental evidence that lengthening the neurogenic period during neocortical development underlies a key aspect of neocortical expansion.


Asunto(s)
Evolución Biológica , Desarrollo Embrionario/fisiología , Neocórtex/embriología , Neurogénesis/fisiología , Animales , Animales Recién Nacidos , Línea Celular , Proliferación Celular , Transferencia de Embrión , Embrión de Mamíferos , Femenino , Masculino , Ratones , Neocórtex/citología , Células-Madre Neurales/fisiología , Células-Madre Neurales/trasplante , Neuronas/fisiología , Embarazo , Ratas , Factores de Tiempo , Quimera por Trasplante/embriología
3.
Methods Mol Biol ; 2005: 101-124, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31175649

RESUMEN

Interspecies chimera formation provides a unique platform for studying donor cell developmental potential, modeling disease in vivo, as well as in vivo production of tissues and organs. The derivation of human pluripotent stem cells (hPSC) from either human embryos or somatic cell reprogramming facilitates our understanding of human development, as well as accelerates our exploration of regenerative medicine for human health. Due to similar organ size, close anatomy, and physiology between pig and human, human-Pig interspecies chimeric model in which pig serves as the host species may open new avenues for studying human embryogenesis, disease pathogenesis, and generation of human organ for transplantation to solve the worldwide donor organ shortage. Our previous study demonstrated chimeric competency of different types of human PSCs in pig host. In this chapter, we introduce our workflow for the generation of human PSCs and analysis of its chimeric contribution to pre- and postimplantation pig embryos.


Asunto(s)
Blastocisto/metabolismo , Modelos Biológicos , Células Madre Pluripotentes/metabolismo , Trasplante de Células Madre , Quimera por Trasplante/embriología , Animales , Blastocisto/citología , Xenoinjertos , Humanos , Células Madre Pluripotentes/citología , Porcinos
4.
Xenotransplantation ; 24(3)2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28397982

RESUMEN

BACKGROUND: Blastocyst complementation is an important technique for generating chimeric organs in organ-deficient pigs, which holds great promise for solving the problem of a shortage of organs for human transplantation procedures. Porcine chimeras have been generated using embryonic germ cells, embryonic stem cells, and induced pluripotent stem cells; however, there are no authentic pluripotent stem cells for pigs. In previous studies, blastomeres from 4- to 8-cell-stage parthenogenetic embryos were able to generate chimeric fetuses efficiently, but the resulting fetuses did not produce live-born young. Here, we used early-stage embryos from somatic cell nuclear transfer (SCNT) to generate chimeric piglets by the aggregation method. Then, the distribution of chimerism in various tissues and organs was observed through the expression of enhanced green fluorescent protein (EGFP). METHODS: Initially, we determined whether 4- to 8- or 8- to 16-cell-stage embryos were more suitable to generate chimeric piglets. Chimeras were produced by aggregating two EGFP-tagged Wuzhishan minipig (WZSP) SCNT embryos and two Bama minipig (BMP) SCNT embryos. The chimeric piglets were identified by coat color and microsatellite and swine leukocyte antigen analyses. Moreover, the distribution of chimerism in various tissues and organs of the piglets was evaluated by EGFP expression. RESULTS: We found that more aggregated embryos were produced using 4- to 8-cell-stage embryos (157/657, 23.9%) than 8- to 16-cell-stage embryos (100/499, 20.0%). Thus, 4- to 8-cell-stage embryos were used for the generation of chimeras. The rate of blastocysts development after aggregating WZSP with BMP embryos was 50.6%. Transfer of 391 blastocysts developed from 4- to 8-cell-stage embryos to five recipients gave rise to 18 piglets, of which two (11.1%) were confirmed to be chimeric by their coat color and microsatellite examination of the skin. One of the chimeric piglets died at 35 days and was subsequently autopsied, whereas the other piglet was maintained for the following observations. The heart and kidneys of the dead piglet showed chimerism, whereas the spinal cord, stomach, pancreas, intestines, muscle, ovary, and brain had no chimerism. CONCLUSIONS: To our knowledge, this is the first report of porcine chimeras generated by aggregating 4- to 8-cell-stage blastomeres from SCNT. We detected chimerism only in the skin, heart, and kidneys. Collectively, these results indicate that aggregation using 4- to 8-cell-stage SCNT embryos offers a practical approach for producing chimeric minipigs. Furthermore, it also provides a potential platform for generating interspecific chimeras between pigs and non-human primates for xenotransplantation.


Asunto(s)
Blastómeros/citología , Técnicas de Transferencia Nuclear , Porcinos Enanos/embriología , Porcinos Enanos/genética , Quimera por Trasplante/embriología , Quimera por Trasplante/genética , Animales , Animales Modificados Genéticamente , Agregación Celular , Técnicas de Cultivo de Embriones/métodos , Transferencia de Embrión/métodos , Femenino , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Antígenos de Histocompatibilidad Clase I/genética , Repeticiones de Microsatélite , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Pigmentación de la Piel/genética , Porcinos , Quimera por Trasplante/metabolismo
5.
Cell Stem Cell ; 17(1): 116-24, 2015 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-26119236

RESUMEN

Because of their similarity to humans, non-human primates are important models for studying human disease and developing therapeutic strategies. Establishment of chimeric animals using embryonic stem cells (ESCs) could help with these investigations, but has not so far been achieved. Here, we show that cynomolgus monkey ESCs (cESCs) grown in adjusted culture conditions are able to incorporate into host embryos and develop into chimeras with contribution in all three germ layers and in germ cell progenitors. Under the optimized culture conditions, which are based on an approach developed previously for naive human ESCs, the cESCs displayed altered growth properties, gene expression profiles, and self-renewal signaling pathways, suggestive of an altered naive-like cell state. Thus our findings show that it is feasible to generate chimeric monkeys using ESCs and open up new avenues for the use of non-human primate models to study both pluripotency and human disease.


Asunto(s)
Células Madre Embrionarias/citología , Macaca fascicularis/embriología , Quimera por Trasplante/embriología , Animales , Blastocisto/citología , Diferenciación Celular , Células Cultivadas , Células Madre Embrionarias/metabolismo , Células Madre Embrionarias/trasplante , Femenino , Humanos , Masculino , Modelos Animales , Mórula/citología , Embarazo , Testículo/citología , Testículo/embriología , Transcriptoma
6.
Genesis ; 52(5): 424-30, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24599776

RESUMEN

Grafting and transplantation experiments in embryology require proper distinction between host and donor tissues. For the avian model this has traditionally been achieved by using two closely related species (e.g., chick and quail) followed by species-specific antibody staining. Here, we show that an in situ hybridization probe against the HINTW gene is a robust and reliable marker for female-derived chicken cells. At all pre-circulation stages tested, all cells in female embryos, independently confirmed by PCR analysis, were strongly positive for HINTW, whereas all male embryos were negative. This probe is broadly applicable in intra-specific chick/chick chimera studies, and as a proof of principle, we utilized this probe to detect female cells in three experimental settings: (1) to mark female donor cells in a node transplantation assay; (2) to distinguish female cells in male/female twins generated by the Cornish pasty culture; and (3) to detect female half of the embryo in artificially generated bilateral gynandromorphs. A rapid, PCR based pre-screening step increases the efficiency of obtaining desired donor/host sex combination from 25% to 100%. For most avian chimera studies, this female-specific in situ probe is a low cost alternative to the commonly used QCPN antibody and to ubiquitous-GFP chicken strains which are not widely available to the research community.


Asunto(s)
Quimera/genética , Hidrolasas/genética , Hidrolasas/metabolismo , Quimera por Trasplante/embriología , Animales , Embrión de Pollo , Pollos , Femenino , Marcadores Genéticos , Masculino , Reacción en Cadena de la Polimerasa , Cromosomas Sexuales , Factores Sexuales
7.
Development ; 140(3): 552-61, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23293287

RESUMEN

Faithful progression through the cell cycle is crucial to the maintenance and developmental potential of stem cells. Here, we demonstrate that neural stem cells (NSCs) and intermediate neural progenitor cells (NPCs) employ a zinc-finger transcription factor specificity protein 2 (Sp2) as a cell cycle regulator in two temporally and spatially distinct progenitor domains. Differential conditional deletion of Sp2 in early embryonic cerebral cortical progenitors, and perinatal olfactory bulb progenitors disrupted transitions through G1, G2 and M phases, whereas DNA synthesis appeared intact. Cell-autonomous function of Sp2 was identified by deletion of Sp2 using mosaic analysis with double markers, which clearly established that conditional Sp2-null NSCs and NPCs are M phase arrested in vivo. Importantly, conditional deletion of Sp2 led to a decline in the generation of NPCs and neurons in the developing and postnatal brains. Our findings implicate Sp2-dependent mechanisms as novel regulators of cell cycle progression, the absence of which disrupts neurogenesis in the embryonic and postnatal brain.


Asunto(s)
Ciclo Celular , Células-Madre Neurales/metabolismo , Neurogénesis , Factor de Transcripción Sp2/metabolismo , Animales , Encéfalo/citología , Encéfalo/embriología , Encéfalo/metabolismo , Recuento de Células , Proliferación Celular , Cruzamientos Genéticos , Implantación del Embrión , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Femenino , Marcadores Genéticos , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Recombinación Homóloga , Proteínas de Filamentos Intermediarios/genética , Proteínas de Filamentos Intermediarios/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Nestina , Células-Madre Neurales/citología , Neuronas/citología , Neuronas/metabolismo , Factor de Transcripción PAX6 , Factores de Transcripción Paired Box/genética , Factores de Transcripción Paired Box/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Factor de Transcripción Sp2/genética , Nicho de Células Madre , Quimera por Trasplante/embriología , Quimera por Trasplante/metabolismo
8.
J Exp Biol ; 216(Pt 10): 1786-92, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23348946

RESUMEN

Chimeras are useful models for studies of developmental biology and cell differentiation. Intraspecies and interspecies germline chimeras have been produced in previous studies, but the feasibility of producing chimeras between animals of two different classes remains unclear. To address this issue, we attempted to produce chimeras between the Chinese soft-shelled turtle and the Peking duck by transferring stage X blastoderm cells to recipient embryos. We then examined the survival and development of the PKH26-labeled donor cells in the heterologous embryos. At early embryonic stages, both turtle and duck donor cells that were labeled with PKH26 were readily observed in the brain, neural tube, heart and gonads of the respective recipient embryos. Movement of turtle donor-derived cells was observed in the duck host embryos after 48 h of incubation. Although none of the hatchlings presented a chimeric phenotype, duck donor-derived cells were detected in a variety of organs in the hatchling turtles, particularly in the gonads. Moreover, in the hatched turtles, mRNA expression of tissue-specific duck genes MEF2a and MEF2c was detected in many tissues, including the muscle, heart, small and large intestines, stomach and kidney. Similarly, SPAG6 mRNA was detected in a subset of turtle tissues, including the gonad and the small and large intestines. These results suggest that duck donor-derived cells can survive and differentiate in recipient turtles; however, no turtle-derived cells were detected in the hatched ducks. Our findings indicate that chimeras can be produced between animals of two different classes.


Asunto(s)
Exoesqueleto/embriología , Blastodermo/citología , Patos/embriología , Transferencia de Embrión , Quimera por Trasplante/embriología , Tortugas/embriología , Animales , Vasos Sanguíneos/citología , Movimiento Celular , China , Embrión no Mamífero/irrigación sanguínea , Embrión no Mamífero/citología , Fluorescencia , Perfilación de la Expresión Génica , Gónadas/citología , Especificidad de Órganos/genética , Reacción en Cadena de la Polimerasa , Tripsina/metabolismo
9.
Dev Biol ; 370(1): 98-109, 2012 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-22824426

RESUMEN

The presence of germ cells in the early gonad is important for sexual fate determination and gonadal development in vertebrates. Recent studies in zebrafish and medaka have shown that a lack of germ cells in the early gonad induces sex reversal in favor of a male phenotype. However, it is uncertain whether the gonadal somatic cells or the germ cells are predominant in determining gonadal fate in other vertebrate. Here, we investigated the role of germ cells in gonadal differentiation in goldfish, a gonochoristic species that possesses an XX-XY genetic sex determination system. The primordial germ cells (PGCs) of the fish were eliminated during embryogenesis by injection of a morpholino oligonucleotide against the dead end gene. Fish without germ cells showed two types of gonadal morphology: one with an ovarian cavity; the other with seminiferous tubules. Next, we tested whether function could be restored to these empty gonads by transplantation of a single PGC into each embryo, and also determined the gonadal sex of the resulting germline chimeras. Transplantation of a single GFP-labeled PGC successfully produced a germline chimera in 42.7% of the embryos. Some of the adult germline chimeras had a developed gonad on one side that contained donor derived germ cells, while the contralateral gonad lacked any early germ cell stages. Female germline chimeras possessed a normal ovary and a germ-cell free ovary-like structure on the contralateral side; this structure was similar to those seen in female morphants. Male germline chimeras possessed a testis and a contralateral empty testis that contained some sperm in the tubular lumens. Analysis of aromatase, foxl2 and amh expression in gonads of morphants and germline chimeras suggested that somatic transdifferentiation did not occur. The offspring of fertile germline chimeras all had the donor-derived phenotype, indicating that germline replacement had occurred and that the transplanted PGC had rescued both female and male gonadal function. These findings suggest that the absence of germ cells did not affect the pathway for ovary or testis development and that phenotypic sex in goldfish is determined by somatic cells under genetic sex control rather than an interaction between the germ cells and somatic cells.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/fisiología , Células Germinativas/fisiología , Carpa Dorada/embriología , Gónadas/embriología , Procesos de Determinación del Sexo/fisiología , Animales , Aromatasa/genética , Aromatasa/metabolismo , Cartilla de ADN/genética , Femenino , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Gónadas/citología , Hibridación in Situ , Masculino , Morfolinos/genética , Receptores de Péptidos/genética , Receptores de Péptidos/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Procesos de Determinación del Sexo/genética , Quimera por Trasplante/embriología
10.
Development ; 139(16): 2926-34, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22764055

RESUMEN

Mesothelium is the surface layer of all coelomic organs and is crucial for the generation of their vasculature. Still, our understanding of the genesis of this essential cell type is restricted to the heart where a localized exogenous population of cells, the proepicardium, migrates to and envelops the myocardium supplying mesothelial, vascular and stromal cell lineages. Currently it is not known whether this pattern of development is specific to the heart or applies broadly to other coelomic organs. Using two independent long-term lineage-tracing studies, we demonstrate that mesothelial progenitors of the intestine are intrinsic to the gut tube anlage. Furthermore, a novel chick-quail chimera model of gut morphogenesis reveals these mesothelial progenitors are broadly distributed throughout the gut primordium and are not derived from a localized and exogenous proepicardium-like source of cells. These data demonstrate an intrinsic origin of mesothelial cells to a coelomic organ and provide a novel mechanism for the generation of mesothelial cells.


Asunto(s)
Epitelio/embriología , Intestinos/embriología , Animales , Animales Modificados Genéticamente , Linaje de la Célula , Embrión de Pollo , Coturnix , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Epitelio/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Mucosa Intestinal/metabolismo , Intestinos/citología , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Quimera por Trasplante/embriología , Quimera por Trasplante/genética , Quimera por Trasplante/metabolismo
11.
J Vis Exp ; (60)2012 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-22349214

RESUMEN

Avian embryos provide a unique platform for studying many vertebrate developmental processes, due to the easy access of the embryos within the egg. Chimeric avian embryos, in which quail donor tissue is transplanted into a chick embryo in ovo, combine the power of indelible genetic labeling of cell populations with the ease of manipulation presented by the avian embryo. Quail-chick chimeras are a classical tool for tracing migratory neural crest cells (NCCs). NCCs are a transient migratory population of cells in the embryo, which originate in the dorsal region of the developing neural tube. They undergo an epithelial to mesenchymal transition and subsequently migrate to other regions of the embryo, where they differentiate into various cell types including cartilage, melanocytes, neurons and glia. NCCs are multipotent, and their ultimate fate is influenced by 1) the region of the neural tube in which they originate along the rostro-caudal axis of the embryo, 2) signals from neighboring cells as they migrate, and 3) the microenvironment of their ultimate destination within the embryo. Tracing these cells from their point of origin at the neural tube, to their final position and fate within the embryo, provides important insight into the developmental processes that regulate patterning and organogenesis. Transplantation of complementary regions of donor neural tube (homotopic grafting) or different regions of donor neural tube (heterotopic grafting) can reveal differences in pre-specification of NCCs along the rostro-caudal axis. This technique can be further adapted to transplant a unilateral compartment of the neural tube, such that one side is derived from donor tissue, and the contralateral side remains unperturbed in the host embryo, yielding an internal control within the same sample. It can also be adapted for transplantation of brain segments in later embryos, after HH10, when the anterior neural tube has closed. Here we report techniques for generating quail-chick chimeras via neural tube transplantation, which allow for tracing of migratory NCCs derived from a discrete segment of the neural tube. Species-specific labeling of the donor-derived cells with the quail-specific QCPN antibody allows the researcher to distinguish donor and host cells at the experimental end point. This technique is straightforward, inexpensive, and has many applications, including fate-mapping, cell lineage tracing, and identifying pre-patterning events along the rostro-caudal axis. Because of the ease of access to the avian embryo, the quail-chick graft technique may be combined with other manipulations, including but not limited to lens ablation, injection of inhibitory molecules, or genetic manipulation via electroporation of expression plasmids, to identify the response of particular migratory streams of NCCs to perturbations in the embryo's developmental program. Furthermore, this grafting technique may also be used to generate other interspecific chimeric embryos such as quail-duck chimeras to study NCC contribution to craniofacial morphogenesis, or mouse-chick chimeras to combine the power of mouse genetics with the ease of manipulation of the avian embryo.


Asunto(s)
Diferenciación Celular/fisiología , Movimiento Celular/fisiología , Cresta Neural/citología , Cresta Neural/trasplante , Quimera por Trasplante/embriología , Trasplante Heterólogo/métodos , Animales , Rastreo Celular/métodos , Embrión de Pollo , Codorniz
12.
Biol Reprod ; 84(6): 1190-7, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21248287

RESUMEN

High frequency production of zebrafish germline chimeras was achieved by transplanting ovarian germ cells into sterile Danio hybrid recipients. Ovarian germ cells were obtained from 3-mo-old adult Tg(vasa:DsRed2-vasa);Tg(bactin:EGFP) double transgenic zebrafish by discontinuous Percoll gradient centrifugation. An average of 755 ± 108 DsRed-positive germ cells was recovered from each female. For transplantations, a total of approximately 620 ± 242 EGFP-positive cells of which 12 ± 4.7 were DsRed-positive germ cells were introduced into the abdominal cavity under the swim bladder of 2-wk-old sterile hybrid larvae. Six weeks after transplantation, a total of 10 recipients, obtained from 2 different transplantations, were examined, and 2 individuals (20%) were identified that possessed a large number of DsRed- and EGFP-positive cells in the gonadal region. The transplanted ovarian germ cells successfully colonized the gonads and differentiated into sperm in the male hybrid recipients. Of 67 adult recipients, 12 (18%) male chimeric fish reproduced and generated normal offspring when paired with wild-type zebrafish females. The fertilization efficiency ranged from 23% to 56%. Although the fertile male chimeras were generated by transplantation of ovarian germ cells, the F1 generation produced by the male chimeras contained both male and female progeny, indicating that male sex determination in zebrafish is not controlled by sex chromosome heterogamy. Our findings indicate that a population of ovarian germ cells that are present in the ovary of adult zebrafish can function as germline stem cells, able to proliferate and differentiate into testicular germ cells and functional sperm in male recipients. The high frequency of germline chimera formation achieved with the ovarian germ cells and the convenience of identifying the chimeras in the sterile host background should make this transplantation system useful for performing genetic manipulations in zebrafish.


Asunto(s)
Células Germinativas/trasplante , Diferenciación Sexual/fisiología , Quimera por Trasplante/embriología , Pez Cebra , Animales , Femenino , Larva/fisiología , Masculino
13.
Development ; 137(17): 2961-71, 2010 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-20699298

RESUMEN

In vertebrates, body musculature originates from somites, whereas head muscles originate from the cranial mesoderm. Neck muscles are located in the transition between these regions. We show that the chick occipital lateral plate mesoderm has myogenic capacity and gives rise to large muscles located in the neck and thorax. We present molecular and genetic evidence to show that these muscles not only have a unique origin, but additionally display a distinct temporal development, forming later than any other muscle group described to date. We further report that these muscles, found in the body of the animal, develop like head musculature rather than deploying the programme used by the trunk muscles. Using mouse genetics we reveal that these muscles are formed in trunk muscle mutants but are absent in head muscle mutants. In concordance with this conclusion, their connective tissue is neural crest in origin. Finally, we provide evidence that the mechanism by which these neck muscles develop is conserved in vertebrates.


Asunto(s)
Mesodermo/embriología , Desarrollo de Músculos , Músculos del Cuello/embriología , Animales , Animales Modificados Genéticamente , Proteínas Aviares/genética , Evolución Biológica , Embrión de Pollo , Coturnix , Regulación del Desarrollo de la Expresión Génica , Ratones , Desarrollo de Músculos/genética , Mutación , Cresta Neural/embriología , Factores de Transcripción Paired Box/genética , Somitos/embriología , Quimera por Trasplante/embriología , Quimera por Trasplante/genética
14.
J Vis Exp ; (46)2010 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-21206472

RESUMEN

Medaka is a small egg-laying freshwater fish that allows both genetic and embryological analyses and is one of the three vertebrate model organisms in which genome-wide phenotype-driven mutant screens were carried out (1). Divergence of functional overlap of related genes between medaka and zebrafish allows identification of novel phenotypes that are unidentifiable in a single species (2), thus medaka and zebrafish are complementary for genetic dissection of the vertebrate genome functions. Manipulation of medaka embryos, such as dechorionation, mounting embryos for imaging and cell transplantation, are key procedures to work on both medaka and zebrafish in a laboratory. Cell transplantation examines cell autonomy of medaka mutations. Chimeras are generated by transplanting labeled cells from donor embryos into unlabeled recipient embryos. Donor cells can be transplanted to specific areas of the recipient embryos based on the fate maps (3) so that clones from transplanted cells can be integrated in the tissue of interest during development. Due to the hard chorion and soft embryos, manipulation of medaka embryos is more involved than in zebrafish. In this video, we show detailed procedures to manipulate medaka embryos.


Asunto(s)
Trasplante de Células/métodos , Corion/cirugía , Oryzias/embriología , Oryzias/cirugía , Quimera por Trasplante/embriología , Animales , Oryzias/genética , Quimera por Trasplante/genética , Pez Cebra/embriología , Pez Cebra/genética
15.
J Vis Exp ; (31)2009 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-19749688

RESUMEN

Certain fundamental questions in the field of developmental biology can only be answered when cells are placed in novel environments or when small groups of cells in a larger context are altered. Watching how one cell interacts with and behaves in a unique environment is essential to characterizing cell functions. Determining how the localized misexpression of a specific protein influences surrounding cells provides insightful information on the roles that protein plays in a variety of developmental processes. Our lab uses the zebrafish model system to uniquely combine genetic approaches with classical transplantation techniques to generate genotypic or phenotypic chimeras. We study neuron-glial cell interactions during the formation of forebrain commissures in zebrafish. This video describes a method that allows our lab to investigate the role of astroglial populations in the diencephalon and the roles of specific guidance cues that influence projecting axons as they cross the midline. Due to their transparency zebrafish embryos are ideal models for this type of ectopic cell placement or localized gene misexpression. Tracking transplanted cells can be accomplished using a vital dye or a transgenic fish line expressing a fluorescent protein. We demonstrate here how to prepare donor embryos with a vital dye tracer for transplantation, as well as how to extract and transplant cells from one gastrula staged embryo to another. We present data showing ectopic GFP+ transgenic cells within the forebrain of zebrafish embryos and characterize the location of these cells with respect to forebrain commissures. In addition, we show laser scanning confocal timelapse microscopy of Alexa 594 labeled cells transplanted into a GFP+ transgenic host embryo. These data provide evidence that gastrula staged transplantation enables the targeted positioning of ectopic cells to address a variety of questions in Developmental Biology.


Asunto(s)
Trasplante de Células/métodos , Transferencia de Embrión/métodos , Gástrula/citología , Pez Cebra/embriología , Animales , Embrión no Mamífero/trasplante , Colorantes Fluorescentes/química , Colorantes Fluorescentes/metabolismo , Gástrula/metabolismo , Microinyecciones/métodos , Microscopía Confocal , Compuestos Orgánicos/química , Compuestos Orgánicos/metabolismo , Quimera por Trasplante/embriología
16.
J Vis Exp ; (29)2009 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-19617875

RESUMEN

One of the most powerful tools used to gain insight into complex developmental processes is the analysis of chimeric embryos. A chimera is defined as an organism that contains cells from more than one animal; mosaics are one type of chimera in which cells from more than one genotype are mixed, usually wild-type and mutant. In the zebrafish, chimeras can be readily made by transplantation of cells from a donor embryo into a host embryo at the appropriate embryonic stage. Labeled donor cells are generated by injection of a lineage marker, such as a fluorescent dye, into the one-cell stage embryo. Labeled donor cells are removed from donor embryos and introduced into unlabeled host embryos using an oil-controlled glass pipette mounted on either a compound or dissecting microscope. Donor cells can in some cases be targeted to a specific region or tissue of the developing blastula or gastrula stage host embryo by choosing a transplantation site in the host embryo based on well-established fate maps.


Asunto(s)
Trasplante de Células/métodos , Quimera por Trasplante/embriología , Pez Cebra/embriología , Animales , Blástula/trasplante , Transferencia de Embrión , Embrión no Mamífero/citología , Femenino , Gástrula/trasplante , Pez Cebra/genética
17.
Cell Prolif ; 42(2): 132-40, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19236382

RESUMEN

OBJECTIVES: In this study, we aimed at determining whether human immature dental pulp stem cells (hIDPSC) would be able to contribute to different cell types in mouse blastocysts without damaging them. Also, we analysed whether these blastocysts would progress further into embryogenesis when implanted to the uterus of foster mice, and develop human/mouse chimaera with retention of hIDPSC derivates and their differentiation. MATERIALS AND METHODS: hIDPSC and mouse blastocysts were used in this study. Fluorescence staining of hIDPSC and injection into mouse blastocysts, was performed. Histology, immunohistochemistry, fluorescence in situ hybridization and confocal microscopy were carried out. RESULTS AND CONCLUSION: hIDPSC showed biological compatibility with the mouse host environment and could survive, proliferate and contribute to the inner cell mass as well as to the trophoblast cell layer after introduction into early mouse embryos (n = 28), which achieved the hatching stage following 24 and 48 h in culture. When transferred to foster mice (n = 5), these blastocysts with hIDPSC (n = 57) yielded embryos (n = 3) and foetuses (n = 6); demonstrating presence of human cells in various organs, such as brain, liver, intestine and hearts, of the human/mouse chimaeras. We verified whether hIDPSC would also be able to differentiate into specific cell types in the mouse environment. Contribution of hIDPSC in at least two types of tissues (muscles and epithelial), was confirmed. We showed that hIDPSC survived, proliferated and differentiated in mouse developing blastocysts and were capable of producing human/mouse chimaeras.


Asunto(s)
Células Madre Adultas/citología , Pulpa Dental/citología , Embrión de Mamíferos/citología , Desarrollo Embrionario/fisiología , Feto/citología , Quimera por Trasplante/embriología , Células Madre Adultas/trasplante , Estructuras Animales/citología , Estructuras Animales/embriología , Estructuras Animales/metabolismo , Animales , Blastocisto/citología , Diferenciación Celular/fisiología , Cromosomas Humanos Y/química , Transferencia de Embrión , Embrión de Mamíferos/embriología , Células Epiteliales/citología , Células Epiteliales/metabolismo , Epitelio/embriología , Epitelio/metabolismo , Femenino , Feto/embriología , Feto/metabolismo , Humanos , Hibridación Fluorescente in Situ , Masculino , Ratones , Ratones Endogámicos , Células Musculares/citología , Células Musculares/metabolismo , Músculos/citología , Músculos/embriología , Músculos/metabolismo , Quimera por Trasplante/metabolismo
18.
Development ; 135(24): 4113-22, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19004859

RESUMEN

Neural crest cells that become pigment cells migrate along a dorsolateral route between the ectoderm and the somite, whereas most other neural crest cells are inhibited from entering this space. This pathway choice has been attributed to unique, cell-autonomous migratory properties acquired by neural crest cells when they become specified as melanoblasts. By shRNA knockdown and overexpression experiments, we investigated the roles of three transmembrane receptors in regulating dorsolateral pathfinding in the chick trunk. We show that Endothelin receptor B2 (EDNRB2) and EphB2 are both determinants in this process, and that, unlike in other species, c-KIT is not. We demonstrate that the overexpression of EDNRB2 can maintain normal dorsolateral migration of melanoblasts in the absence of EphB2, and vice versa, suggesting that changes in receptor expression levels regulate the invasion of this pathway. Furthermore, by heterotopic grafting, we show that neural crest cell populations that do not rely on the activation of these receptors can migrate dorsolaterally only if this path is free of inhibitory molecules. We conclude that the requirement for EDNRB2 and EphB2 expression by melanoblasts is to support their migration by helping them to overcome repulsive or non-permissive cues in the dorsolateral environment.


Asunto(s)
Cresta Neural/embriología , Receptor EphB2/fisiología , Receptores de Endotelina/fisiología , Animales , Animales Modificados Genéticamente , Secuencia de Bases , Tipificación del Cuerpo/genética , Tipificación del Cuerpo/fisiología , Movimiento Celular , Supervivencia Celular , Embrión de Pollo , Coturnix , Cartilla de ADN/genética , Regulación del Desarrollo de la Expresión Génica , Melanocitos/citología , Melanocitos/fisiología , Modelos Neurológicos , Cresta Neural/citología , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Proto-Oncogénicas c-kit/fisiología , ARN Interferente Pequeño/genética , Receptor EphB2/genética , Receptores de Endotelina/genética , Transducción de Señal , Quimera por Trasplante/embriología
19.
Cell Transplant ; 17(5): 495-506, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18714669

RESUMEN

It was reported that the dose of self-antigens can determine the consequence of deletional tolerance and donor T cells are critical for tolerance induction in mixed chimeras. This study aimed at assessing the effect of cell doses and marrow T cells on engraftment and tolerance induction after prenatal bone marrow transplantation. Intraperitoneal cell transplantation was performed in FVB/N (H-2K(q)) mice at gestational day 14 with escalating doses of adult C57BL/6 (H-2K(b)) marrows. Peripheral chimerism was examined postnatally by flow cytometry and tolerance was tested by skin transplantation. Transplantation of light-density marrow cells showed a dose response. High-level chimerism emerged with a threshold dose of 5.0 x 10(6) and host leukocytes could be nearly replaced at a dose of 7.5-10.0 x 10(6). High-dose transplants conferred a steady long-lasting donor-specific tolerance but were accompanied by >50% incidence of graft-versus-host disease. Depletion of marrow T cells lessened graft-versus-host disease to the detriment of engraftment. With low-level chimerism, tolerance was a graded phenomenon dependent upon the level of chimerism. Durable chimerism within 6 months required a threshold of > or = 2% chimerism at 1 month of age and predicted a 50% chance of long-term tolerance, whereas transient chimerism (<2%) only caused hyporesponsiveness to the donor. Tolerance induction did not succeed without peripheral chimerism even if a large amount of injected donor cells persisted in the peritoneum. Neither did an increase in cell doses or donor T-cell contents benefit skin graft survivals unless it had substantially improved peripheral chimerism. Thus, peripheral chimerism level can be a simple and straightforward test to predict the degree of prenatal immune tolerance.


Asunto(s)
Trasplante de Médula Ósea/inmunología , Tolerancia Inmunológica , Trasplante de Piel/inmunología , Linfocitos T/inmunología , Quimera por Trasplante/inmunología , Acondicionamiento Pretrasplante , Animales , Recuento de Células , Supresión Clonal , Femenino , Supervivencia de Injerto/inmunología , Depleción Linfocítica , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Valor Predictivo de las Pruebas , Embarazo , Linfocitos T/citología , Quimera por Trasplante/embriología , Inmunología del Trasplante
20.
Biol Reprod ; 79(5): 931-7, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18685127

RESUMEN

The present study was conducted to apply an interspecies germ cell transfer technique to wild bird reproduction. Pheasant (Phasianus colchicus) primordial germ cells (PGCs) retrieved from the gonads of 7-day-old embryos were transferred to the bloodstream of 2.5-day-old chicken (Gallus gallus) embryos. Pheasant-to-chicken germline chimeras hatched from the recipient embryos, and 10 pheasants were derived from testcross reproduction of the male chimeras with female pheasants. Gonadal migration of the transferred PGCs, their involvement in spermatogenesis, and production of chimeric semen were confirmed. The phenotype of pheasant progenies derived from the interspecies transfer was identical to that of wild pheasants. The average efficiency of reproduction estimated from the percentage of pheasants to total progenies was 17.5%. In conclusion, interspecies germ cell transfer into a developing embryo can be used for wild bird reproduction, and this reproductive technology may be applicable in conserving endangered bird species.


Asunto(s)
Cruzamiento/métodos , Galliformes/fisiología , Células Germinativas/trasplante , Técnicas Reproductivas Asistidas , Quimera por Trasplante/embriología , Animales , Animales Salvajes , Embrión de Pollo , Embrión no Mamífero/citología , Femenino , Galliformes/embriología , Masculino , Espermatozoides/citología , Trasplante Heterólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...