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










Base de dados
Intervalo de ano de publicação
1.
Reprod Fertil Dev ; 33(12): 725-735, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34488937

RESUMO

Programmed cell death plays a key role in mammalian development because the morphological events of an organism's formation are dependent on apoptosis. In the mouse development, the first apoptotic waves occur physiologically at the blastocyst stage. Cell number and the mean nucleus to cytoplasm (N/C) ratio increase exponentially throughout subsequent embryo cleavages, while cell volume concurrently decreases from the zygote to blastocyst stage. In this study we tested the hypothesis that reorganisation of the embryo structure by manipulating cell number, the N/C ratio and the cell volume of 2-cell embryos may result in the earlier and more frequent occurrence of apoptosis. The results indicate that doubling ('Aggregates' group) or halving ('Embryos 1/2' group) the initial cell number and modifying embryo volume, ploidy ('Embryos 4n' group) and the N/C ratio ('Embryos 2/1' group) reduce the probability of apoptosis in the resulting embryos. There was a higher probability of apoptosis in the inner cell mass of the blastocyst, but apoptotic cells were never observed at the morula stage in any of the experimental groups. Thus, manipulation of cell number, embryo volume, the N/C ratio and ploidy cause subtle changes in the occurrence of apoptosis, although these are mostly dependent on embryo stage and cell lineage (trophectoderm or inner cell mass), which have the greatest effect on the probability of apoptosis.


Assuntos
Apoptose/fisiologia , Blastocisto/fisiologia , Desenvolvimento Embrionário/fisiologia , Animais , Blastocisto/citologia , Contagem de Células , Técnicas de Cultura Embrionária , Camundongos
2.
Cells ; 10(3)2021 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-33668852

RESUMO

The phenomenon of the reprogramming of terminally differentiated cells can be achieved by various means, like somatic cell nuclear transfer, cell fusion with a pluripotent cell, or the introduction of pluripotency genes. Here, we present the evidence that somatic cells can attain the expression of pluripotency markers after their introduction into early embryos. Mouse embryonic fibroblasts introduced between blastomeres of cleaving embryos, within two days of in vitro culture, express transcription factors specific to blastocyst lineages, including pluripotency factors. Analysis of donor tissue marker DNA has revealed that the progeny of introduced cells are found in somatic tissues of foetuses and adult chimaeras, providing evidence for cell reprogramming. Analysis of ploidy has shown that in the chimaeras, the progeny of introduced cells are either diploid or tetraploid, the latter indicating cell fusion. The presence of donor DNA in diploid cells from chimaeric embryos proved that the non-fused progeny of introduced fibroblasts persisted in chimaeras, which is evidence of reprogramming by embryonic niche. When adult somatic (cumulus) cells were introduced into early cleavage embryos, the extent of integration was limited and only cell fusion-mediated reprogramming was observed. These results show that both cell fusion and cell interactions with the embryonic niche reprogrammed somatic cells towards pluripotency.


Assuntos
Envelhecimento/fisiologia , Biomarcadores/metabolismo , Reprogramação Celular , Quimera/fisiologia , Embrião de Mamíferos/citologia , Células-Tronco Pluripotentes/metabolismo , Animais , Blastocisto/citologia , Blastômeros/citologia , Fusão Celular , Linhagem Celular , Células do Cúmulo/citologia , Diploide , Técnicas de Cultura Embrionária , Desenvolvimento Embrionário , Feminino , Feto/citologia , Corantes Fluorescentes/metabolismo , Camundongos , Mórula/citologia , Células-Tronco Pluripotentes/citologia , Gravidez , Tetraploidia
3.
Postepy Biochem ; 67(4): 370-382, 2021 12 31.
Artigo em Polonês | MEDLINE | ID: mdl-35107955

RESUMO

The embryonic development of placental mammals takes place inside the mother's womb, which requires the formation of appropriate supportive structures by both the mother's organism and the developing embryo. The first stages of mammalian embryonic development, preceding implantation, are the period of differentiation of the first cell lineages ­ epiblast (which will give rise to the embryo proper), and extra-embryonic lineages: trophectoderm (responsible for implantation and formation of the placenta) and primitive endoderm (giving rise to the yolk sac). Their differentiation is necessary for further development, and is a common feature of the development of all placental mammals, but the timing and molecular mechanisms responsible for these processes differ between mammalian species.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Placenta , Animais , Diferenciação Celular , Linhagem da Célula , Embrião de Mamíferos , Feminino , Gravidez
4.
Development ; 147(14)2020 07 22.
Artigo em Inglês | MEDLINE | ID: mdl-32699138

RESUMO

Pre-implantation mammalian development unites extreme plasticity with a robust outcome: the formation of a blastocyst, an organised multi-layered structure ready for implantation. The process of blastocyst formation is one of the best-known examples of self-organisation. The first three cell lineages in mammalian development specify and arrange themselves during the morphogenic process based on cell-cell interactions. Despite decades of research, the unifying principles driving early mammalian development are still not fully defined. Here, we discuss the role of physical forces, and molecular and cellular mechanisms, in driving self-organisation and lineage formation that are shared between eutherian mammals.


Assuntos
Embrião de Mamíferos/metabolismo , Animais , Blastocisto/metabolismo , Diferenciação Celular , Linhagem da Célula , Desenvolvimento Embrionário , Regulação da Expressão Gênica no Desenvolvimento , Camadas Germinativas/metabolismo , Humanos , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
5.
Int J Dev Biol ; 63(3-4-5): 187-201, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31058296

RESUMO

The preimplantation development of mammals generally follows the same plan. It starts with the formation of a totipotent zygote, and through consecutive cleavage divisions and differentiation events leads to blastocyst formation. However, the intervening events may differ between species. The regulation of these processes has been extensively studied in the mouse, which displays some unique features among eutherian mammals. Farm animals such as pigs, cattle, sheep and rabbits share several similarities with one another, and with the human developmental plan. These include the timing of epigenetic reprogramming, the moment of embryonic genome activation and the developmental time-frame. Recently, efficient techniques for genetic modification have been established for large domestic animals. Genome sequences and gene manipulation tools are now available for cattle, pigs, sheep and goats, and a larger number of genetically engineered livestock is now accessible for biomedical research. Yet, these animals still make up less than 0.5% of animals in research, mainly due to our inadequate knowledge of the processes responsible for pluripotency maintenance (to date no stable naïve embryonic stem cell lines have been established) and early development. In this review, we highlight our present knowledge of the key preimplantation events in the 3 non-rodent species which present the highest potential for biomedical research related to early embryonic development: cattle, which offer an excellent model to study human in vitro embryo development, pigs which emerge as models to study the long-term effects of gene-based therapies and rabbits, which in many aspects of embryology resemble the human.


Assuntos
Blastocisto/fisiologia , Células-Tronco Pluripotentes/metabolismo , Zigoto/metabolismo , Animais , Pesquisa Biomédica , Bovinos , Embrião de Mamíferos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Modelos Animais , Partenogênese , Coelhos , Ovinos/embriologia , Transdução de Sinais/fisiologia , Suínos/embriologia
6.
Methods Mol Biol ; 1920: 163-182, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30737692

RESUMO

Mouse genetic approaches when combined with live imaging tools are revolutionizing our current understanding of mammalian developmental biology. The availability and improvement of a wide variety of genetically encoded fluorescent proteins have provided indispensable tools to visualize cells and subcellular features in living organisms. It is now possible to generate genetically modified mouse lines expressing several spectrally distinct fluorescent proteins in a tissue-specific or -inducible manner. Such reporter-expressing lines make it possible to image dynamic cellular behaviors in the context of living embryos undergoing normal or aberrant development. As with all viviparous mammals, mouse embryos develop within the uterus, and so live imaging experiments require culture conditions that closely mimic the in vivo environment. Over the past decades, significant advances have been made in developing conditions for culturing both pre- and postimplantation-stage mouse embryos. In this chapter, we discuss routine methods for ex utero culture of preimplantation- and postimplantation-stage mouse embryos. In particular, we describe protocols for collecting mouse embryos of various stages, setting up culture conditions for their ex utero culture and imaging, and using laser scanning confocal microscopy to visualize live processes in mouse embryos expressing fluorescent reporters.


Assuntos
Técnicas de Cultura Embrionária , Embrião de Mamíferos , Desenvolvimento Embrionário , Imagem Molecular/métodos , Animais , Desenvolvimento Embrionário/genética , Feminino , Expressão Gênica , Genes Reporter , Proteínas de Fluorescência Verde/genética , Humanos , Camundongos , Microscopia Confocal/métodos , Imagem com Lapso de Tempo
7.
PLoS One ; 14(2): e0212109, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30735538

RESUMO

During preimplantation mouse development stages, emerging pluripotent epiblast (Epi) and extraembryonic primitive endoderm (PrE) cells are first distributed in the blastocyst in a "salt-and-pepper" manner before they segregate into separate layers. As a result of segregation, PrE cells become localised on the surface of the inner cell mass (ICM), and the Epi is enclosed by the PrE on one side and by the trophectoderm on the other. During later development, a subpopulation of PrE cells migrates away from the ICM and forms the parietal endoderm (PE), while cells remaining in contact with the Epi form the visceral endoderm (VE). Here, we asked: what are the mechanisms mediating Epi and PrE cell segregation and the subsequent VE vs PE specification? Differences in cell adhesion have been proposed; however, we demonstrate that the levels of plasma membrane-bound E-cadherin (CDH1, cadherin 1) in Epi and PrE cells only differ after the segregation of these lineages within the ICM. Moreover, manipulating E-cadherin levels did not affect lineage specification or segregation, thus failing to confirm its role during these processes. Rather, we report changes in E-cadherin localisation during later PrE-to-PE transition which are accompanied by the presence of Vimentin and Twist, supporting the hypothesis that an epithelial-to-mesenchymal transition process occurs in the mouse peri-implantation blastocyst.


Assuntos
Blastocisto/citologia , Blastocisto/metabolismo , Caderinas/metabolismo , Endoderma/citologia , Células-Tronco Pluripotentes/citologia , Animais , Morte Celular , Linhagem da Célula , Membrana Celular/metabolismo , Implantação do Embrião , Transição Epitelial-Mesenquimal , Feminino , Camundongos , Transporte Proteico
8.
Curr Top Dev Biol ; 128: 267-294, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29477166

RESUMO

During the first days following fertilization, cells of mammalian embryo gradually lose totipotency, acquiring distinct identity. The first three lineages specified in the mammalian embryo are pluripotent epiblast, which later gives rise to the embryo proper, and two extraembryonic lineages, hypoblast (also known as primitive endoderm) and trophectoderm, which form tissues supporting development of the fetus in utero. Most of our knowledge regarding the mechanisms of early lineage specification in mammals comes from studies in the mouse. However, the growing body of evidence points to both similarities and species-specific differences. Understanding molecular and cellular mechanisms of early embryonic development in nonrodent mammals expands our understanding of basic mechanisms of differentiation and is essential for the development of effective protocols for assisted reproduction in agriculture, veterinary medicine, and for biomedical research. This review summarizes the current state of knowledge on key events in epiblast, hypoblast, and trophoblast differentiation in domestic mammals.


Assuntos
Animais Domésticos/embriologia , Desenvolvimento Embrionário , Animais , Animais Domésticos/genética , Linhagem da Célula , Embrião de Mamíferos/citologia , Regulação da Expressão Gênica no Desenvolvimento , Transdução de Sinais/genética
9.
Development ; 144(20): 3719-3730, 2017 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-28935706

RESUMO

Formation of epiblast (EPI) - the founder line of all embryonic lineages - and extra-embryonic supportive tissues is one of the key events in mammalian development. The prevailing model of early mammalian development is based almost exclusively on the mouse. Here, we provide a comprehensive, stage-by-stage analysis of EPI and extra-embryonic primitive endoderm (PrE) formation during preimplantation development of the rabbit. Although we observed that rabbit embryos have several features in common with mouse embryos, including a stage-related initiation of lineage specification, our results demonstrate the existence of some key differences in lineage specification among mammals. Contrary to the current view, our data suggest that reciprocal repression of GATA6 and NANOG is not fundamental for the initial stages of PrE versus EPI specification in mammals. Furthermore, our results provide insight into the observed discrepancies relating to the role of FGF/ERK signalling in PrE versus EPI specification between mouse and other mammals.


Assuntos
Endoderma/citologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Sistema de Sinalização das MAP Quinases , Animais , Blastocisto/citologia , Diferenciação Celular , Linhagem da Célula , Feminino , Fator de Transcrição GATA6/metabolismo , Perfilação da Expressão Gênica , Camadas Germinativas/citologia , Proteínas HMGB/metabolismo , Camundongos , Proteína Homeobox Nanog/metabolismo , Coelhos , Fatores de Transcrição SOXB1/metabolismo , Fatores de Transcrição SOXF/metabolismo
10.
Mol Hum Reprod ; 22(10): 681-690, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-26769259

RESUMO

Understanding the mechanisms underlying the first cell differentiation events in human preimplantation development is fundamental for defining the optimal conditions for IVF techniques and selecting the most viable embryos for further development. However, our comprehension of the very early events in development is still very limited. Moreover, our knowledge on early lineage specification comes primarily from studying the mouse model. It is important to recognize that although mammalian embryos share similar morphological landmarks, the timing and molecular control of developmental events may vary substantially between species. Mammalian blastocysts comprise three cell types that arise through two sequential rounds of binary cell fate decisions. During the first decision, cells located on the outside of the developing embryo form a precursor lineage for the embryonic part of the placenta: the trophectoderm and cells positioned inside the embryo become the inner cell mass (ICM). Subsequently, ICM cells differentiate into embryonic lineages that give rise to a variety of tissues in the developing foetus: either the epiblast or extraembryonic primitive endoderm. Successful formation of all three lineages is a prerequisite for implantation and development to term. A comprehensive understanding of the lineage specification processes in mammals is therefore necessary to shed light on the causes of early miscarriages and early pregnancy pathologies in humans.


Assuntos
Blastocisto/citologia , Diferenciação Celular/fisiologia , Sobrevivência Celular/fisiologia , Embrião de Mamíferos/citologia , Animais , Blastocisto/metabolismo , Diferenciação Celular/genética , Sobrevivência Celular/genética , Embrião de Mamíferos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Humanos , Camundongos
11.
Genes Dev ; 27(9): 997-1002, 2013 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-23651855

RESUMO

Reciprocal inductive interactions between the embryonic and extraembryonic tissues establish the anterior-posterior (AP) axis of the early mouse embryo. The anterior visceral endoderm (AVE) signaling center emerges at the distal tip of the embryo at embryonic day 5.5 and translocates to the prospective anterior side of the embryo. The process of AVE induction and migration are poorly understood. Here we demonstrate that the T-box gene Eomesodermin (Eomes) plays an essential role in AVE recruitment, in part by directly activating the homeobox transcription factor Lhx1. Thus, Eomes function in the visceral endoderm (VE) initiates an instructive transcriptional program controlling AP identity.


Assuntos
Endoderma/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas com Domínio T/metabolismo , Animais , Padronização Corporal/genética , Linhagem Celular , Embrião de Mamíferos , Proteínas com Homeodomínio LIM/genética , Proteínas com Homeodomínio LIM/metabolismo , Camundongos , Mutação , Proteínas com Domínio T/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
12.
Genesis ; 51(4): 219-33, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23349011

RESUMO

The preimplantation period of mouse early embryonic development is devoted to the specification of two extraembryonic tissues and their spatial segregation from the pluripotent epiblast. During this period two cell fate decisions are made while cells gradually lose their totipotency. The first fate decision involves the segregation of the extraembryonic trophectoderm (TE) lineage from the inner cell mass (ICM); the second occurs within the ICM and involves the segregation of the extraembryonic primitive endoderm (PrE) lineage from the pluripotent epiblast (EPI) lineage, which eventually gives rise to the embryo proper. Multiple determinants, such as differential cellular properties, signaling cues and the activity of transcriptional regulators, influence lineage choice in the early embryo. Here, we provide an overview of our current understanding of the mechanisms governing these cell fate decisions ensuring proper lineage allocation and segregation, while at the same time providing the embryo with an inherent flexibility to adjust when perturbed.


Assuntos
Blastocisto/citologia , Diferenciação Celular , Camundongos/embriologia , Morfogênese , Animais , Linhagem da Célula , Células-Tronco Embrionárias/citologia
13.
Development ; 140(2): 267-79, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-23193166

RESUMO

The emergence of pluripotent epiblast (EPI) and primitive endoderm (PrE) lineages within the inner cell mass (ICM) of the mouse blastocyst involves initial co-expression of lineage-associated markers followed by mutual exclusion and salt-and-pepper distribution of lineage-biased cells. Precisely how EPI and PrE cell fate commitment occurs is not entirely clear; however, previous studies in mice have implicated FGF/ERK signaling in this process. Here, we investigated the phenotype resulting from zygotic and maternal/zygotic inactivation of Fgf4. Fgf4 heterozygous blastocysts exhibited increased numbers of NANOG-positive EPI cells and reduced numbers of GATA6-positive PrE cells, suggesting that FGF signaling is tightly regulated to ensure specification of the appropriate numbers of cells for each lineage. Although the size of the ICM was unaffected in Fgf4 null mutant embryos, it entirely lacked a PrE layer and exclusively comprised NANOG-expressing cells at the time of implantation. An initial period of widespread EPI and PrE marker co-expression was however established even in the absence of FGF4. Thus, Fgf4 mutant embryos initiated the PrE program but exhibited defects in its restriction phase, when lineage bias is acquired. Consistent with this, XEN cells could be derived from Fgf4 mutant embryos in which PrE had been restored and these cells appeared indistinguishable from wild-type cells. Sustained exogenous FGF failed to rescue the mutant phenotype. Instead, depending on concentration, we noted no effect or conversion of all ICM cells to GATA6-positive PrE. We propose that heterogeneities in the availability of FGF produce the salt-and-pepper distribution of lineage-biased cells.


Assuntos
Endoderma/fisiologia , Fator 4 de Crescimento de Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Animais , Blastocisto/citologia , Técnicas de Cultura de Células/métodos , Diferenciação Celular , Linhagem da Célula , Fator de Transcrição GATA6/metabolismo , Marcadores Genéticos , Camadas Germinativas/citologia , Heterozigoto , Proteínas de Homeodomínio/metabolismo , Processamento de Imagem Assistida por Computador , Camundongos , Mutação , Proteína Homeobox Nanog , Transdução de Sinais
14.
Development ; 139(1): 129-39, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22096072

RESUMO

Cell differentiation during pre-implantation mammalian development involves the formation of two extra-embryonic lineages: trophoblast and primitive endoderm (PrE). A subset of cells within the inner cell mass (ICM) of the blastocyst does not respond to differentiation signals and forms the pluripotent epiblast, which gives rise to all of the tissues in the adult body. How this group of cells is set aside remains unknown. Recent studies documented distinct sequential phases of marker expression during the segregation of epiblast and PrE within the ICM. However, the connection between marker expression and lineage commitment remains unclear. Using a fluorescent reporter for PrE, we investigated the plasticity of epiblast and PrE precursors. Our observations reveal that loss of plasticity does not coincide directly with lineage restriction of epiblast and PrE markers, but rather with exclusion of the pluripotency marker Oct4 from the PrE. We note that individual ICM cells can contribute to all three lineages of the blastocyst until peri-implantation. However, epiblast precursors exhibit less plasticity than precursors of PrE, probably owing to differences in responsiveness to extracellular signalling. We therefore propose that the early embryo environment restricts the fate choice of epiblast but not PrE precursors, thus ensuring the formation and preservation of the pluripotent foetal lineage.


Assuntos
Massa Celular Interna do Blastocisto/fisiologia , Diferenciação Celular/fisiologia , Linhagem da Célula/fisiologia , Embrião de Mamíferos/embriologia , Desenvolvimento Embrionário/fisiologia , Endoderma/fisiologia , Animais , Imuno-Histoquímica , Camundongos , Microscopia Confocal , Fator 3 de Transcrição de Octâmero/metabolismo
15.
Dev Biol ; 361(2): 245-62, 2012 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-22051107

RESUMO

The visceral endoderm (VE) is an epithelial tissue in the early postimplantation mouse embryo that encapsulates the pluripotent epiblast distally and the extraembryonic ectoderm proximally. In addition to facilitating nutrient exchange before the establishment of a circulation, the VE is critical for patterning the epiblast. Since VE is derived from the primitive endoderm (PrE) of the blastocyst, and PrE-derived eXtraembryonic ENdoderm (XEN) cells can be propagated in vitro, XEN cells should provide an important tool for identifying factors that direct VE differentiation. In this study, we demonstrated that BMP4 signaling induces the formation of a polarized epithelium in XEN cells. This morphological transition was reversible, and was associated with the acquisition of a molecular signature comparable to extraembryonic (ex) VE. Resembling exVE which will form the endoderm of the visceral yolk sac, BMP4-treated XEN cells regulated hematopoiesis by stimulating the expansion of primitive erythroid progenitors. We also observed that LIF exerted an antagonistic effect on BMP4-induced XEN cell differentiation, thereby impacting the extrinsic conditions used for the isolation and maintenance of XEN cells in an undifferentiated state. Taken together, our data suggest that XEN cells can be differentiated towards an exVE identity upon BMP4 stimulation and therefore represent a valuable tool for investigating PrE lineage differentiation.


Assuntos
Padronização Corporal/efeitos dos fármacos , Proteína Morfogenética Óssea 4/farmacologia , Endoderma/efeitos dos fármacos , Endoderma/embriologia , Membranas Extraembrionárias/citologia , Transdução de Sinais/efeitos dos fármacos , Vísceras/embriologia , Animais , Padronização Corporal/genética , Polaridade Celular/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Endoderma/citologia , Epitélio/efeitos dos fármacos , Epitélio/embriologia , Epitélio/metabolismo , Células Precursoras Eritroides/citologia , Células Precursoras Eritroides/efeitos dos fármacos , Células Precursoras Eritroides/metabolismo , Membranas Extraembrionárias/efeitos dos fármacos , Membranas Extraembrionárias/embriologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Proteínas de Fluorescência Verde/metabolismo , Fator Inibidor de Leucemia/farmacologia , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Transdução de Sinais/genética , Regulação para Cima/efeitos dos fármacos , Vísceras/citologia , Vísceras/efeitos dos fármacos
16.
Methods Mol Biol ; 770: 243-57, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21805267

RESUMO

Mouse genetic approaches when combined with live imaging tools have the potential to revolutionize our current understanding of mammalian biology. The availability and improvement of a wide variety of fluorescent proteins have provided indispensable tools to visualize cells in living organisms. It is now possible to generate genetically modified mouse strains expressing fluorescent proteins in a tissue-specific manner. These reporter-expressing strains make it possible to image dynamic cell behaviors in the context of a living embryo. Since mouse embryos develop within the uterus, live imaging experiments require culture conditions that closely mimic those in vivo. Over the past few decades, significant advances have been made in developing conditions for culturing both pre- and postimplantation stage embryos. In this chapter, we will discuss methods for ex utero culture of preimplantation and postimplantation stage mouse embryos. In particular, we will describe protocols for collecting embryos at various stages, setting up culture conditions for imaging and using laser scanning confocal microscopy to visualize live processes in mouse embryos expressing fluorescent reporters.


Assuntos
Técnicas de Cultura Embrionária/métodos , Embrião de Mamíferos/citologia , Camundongos/embriologia , Imagem Molecular/métodos , Útero , Animais , Técnicas de Cultura Embrionária/instrumentação , Implantação do Embrião , Embrião de Mamíferos/fisiologia , Feminino , Masculino , Microscopia , Imagem Molecular/instrumentação , Fenômenos Ópticos
17.
Dev Biol ; 350(2): 393-404, 2011 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-21146513

RESUMO

Cells of the primitive endoderm (PrE) and the pluripotent epiblast (EPI), the two lineages specified within the inner cell mass (ICM) of the mouse blastocyst stage embryo, are segregated into adjacent tissue layers by the end of the preimplantation period. The PrE layer which emerges as a polarized epithelium adjacent to the blastocoel, with a basement membrane separating it from the EPI, has two derivatives, the visceral and parietal endoderm. In this study we have investigated the localization of two transcriptional regulators of the SOX family, SOX17 and SOX7, within the PrE and its derivatives. We noted that SOX17 was first detected in a salt-and-pepper distribution within the ICM, subsequently becoming restricted to the nascent PrE epithelium. This dynamic distribution of SOX17 resembled the localization of GATA6 and GATA4, two other PrE lineage-specific transcription factors. By contrast, SOX7 was only detected in PrE cells positioned in contact with the blastocoel, raising the possibility that these cells are molecularly distinct. Our observations support a model of sequential GATA6 > SOX17 > GATA4 > SOX7 transcription factor activation within the PrE lineage, perhaps correlating with the consecutive periods of cell lineage 'naïvete', commitment and sorting. Furthermore our data suggest that co-expression of SOX17 and SOX7 within sorted PrE cells could account for the absence of a detectable phenotype of Sox17 mutant blastocysts. However, analysis of implantation-delayed blastocysts, revealed a role for SOX17 in the maintenance of PrE epithelial integrity, with the absence of SOX17 leading to premature delamination and migration of parietal endoderm.


Assuntos
Blastocisto/fisiologia , Diferenciação Celular , Linhagem da Célula , Endoderma/fisiologia , Proteínas HMGB/fisiologia , Fatores de Transcrição SOXF/fisiologia , Fatores de Transcrição/fisiologia , Animais , Blastocisto/citologia , Movimento Celular , Polaridade Celular , Endoderma/citologia , Feminino , Proteínas HMGB/análise , Masculino , Camundongos , Camundongos Endogâmicos ICR , Fatores de Transcrição SOXF/análise , Fatores de Transcrição SOXF/genética
18.
BMC Dev Biol ; 10: 121, 2010 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-21176145

RESUMO

BACKGROUND: Understanding the dynamic cellular behaviors and underlying molecular mechanisms that drive morphogenesis is an ongoing challenge in biology. Live imaging provides the necessary methodology to unravel the synergistic and stereotypical cell and molecular events that shape the embryo. Genetically-encoded reporters represent an essential tool for live imaging. Reporter strains can be engineered by placing cis-regulatory elements of interest to direct the expression of a desired reporter gene. In the case of canonical Wnt signaling, also referred to as Wnt/ß-catenin signaling, since the downstream transcriptional response is well understood, reporters can be designed that reflect sites of active Wnt signaling, as opposed to sites of gene transcription, as is the case with many fluorescent reporters. However, even though several transgenic Wnt/ß-catenin reporter strains have been generated, to date, none provides the single-cell resolution favored for live imaging studies. RESULTS: We have placed six copies of a TCF/Lef responsive element and an hsp68 minimal promoter in front of a fluorescent protein fusion comprising human histone H2B to GFP and used it to generate a strain of mice that would report Wnt/ß-catenin signaling activity. Characterization of developmental and adult stages of the resulting TCF/Lef:H2B-GFP strain revealed discrete and specific expression of the transgene at previously characterized sites of Wnt/ß-catenin signaling. In support of the increased sensitivity of the TCF/Lef:H2B-GFP reporter, additional sites of Wnt/ß-catenin signaling not documented with other reporters but identified through genetic and embryological analysis were observed. Furthermore, the sub-cellular localization of the reporter minimized reporter perdurance, and allowed visualization and tracking of individual cells within a cohort, so facilitating the detailed analysis of cell behaviors and signaling activity during morphogenesis. CONCLUSION: By combining the Wnt activity read-out efficiency of multimerized TCF/Lef DNA binding sites, together with the high-resolution imaging afforded by subcellularly-localized fluorescent fusion proteins such as H2B-GFP, we have created a mouse transgenic line that faithfully recapitulates Wnt signaling activity at single-cell resolution. The TCF/Lef:H2B-GFP reporter represents a unique tool for live imaging the in vivo processes triggered by Wnt/ß-catenin signaling, and thus should help the formulation of a high-resolution understanding of the serial events that define the morphogenetic process regulated by this signaling pathway.


Assuntos
Camundongos/embriologia , Transdução de Sinais , Análise de Célula Única/métodos , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Animais , Endoderma/metabolismo , Gastrulação , Proteínas de Fluorescência Verde/metabolismo , Camundongos Transgênicos , Linha Primitiva/metabolismo , Fatores de Transcrição TCF/metabolismo
19.
Development ; 135(18): 3081-91, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18725515

RESUMO

The first two lineages to differentiate from a pluripotent cell population during mammalian development are the extraembryonic trophectoderm (TE) and the primitive endoderm (PrE). Whereas the mechanisms of TE specification have been extensively studied, segregation of PrE and the pluripotent epiblast (EPI) has received comparatively little attention. A current model of PrE specification suggests PrE precursors exhibit an apparently random distribution within the inner cell mass of the early blastocyst and then segregate to their final position lining the cavity by the late blastocyst. We have identified platelet-derived growth factor receptor alpha (Pdgfralpha) as an early-expressed protein that is also a marker of the later PrE lineage. By combining live imaging of embryos expressing a histone H2B-GFP fusion protein reporter under the control of Pdgfra regulatory elements with the analysis of lineage-specific markers, we investigated the events leading to PrE and EPI lineage segregation in the mouse, and correlated our findings using an embryo staging system based on total cell number. Before blastocyst formation, lineage-specific factors are expressed in an overlapping manner. Subsequently, a gradual progression towards a mutually exclusive expression of PrE- and EPI-specific markers occurs. Finally, cell sorting is achieved by a variety of cell behaviours and by selective apoptosis.


Assuntos
Blastocisto/metabolismo , Linhagem da Célula/genética , Células/metabolismo , Endoderma/metabolismo , Animais , Blastocisto/citologia , Feminino , Genes Reporter , Marcadores Genéticos/genética , Proteínas de Fluorescência Verde/metabolismo , Histonas/genética , Histonas/metabolismo , Camundongos , Modelos Biológicos , Gravidez , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Proteínas Recombinantes de Fusão/metabolismo
20.
Reproduction ; 133(1): 207-18, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17244747

RESUMO

Foetal fibroblasts (FFs) labelled with vital fluorescent dye were microsurgically introduced into eight-cell mouse embryos, three cells to each embryo. FFs were first identified in the inner cell mass (ICM) in about one-third of embryos, whereas in three quarters of embryos FFs were located among trophoblast cells. Some elimination of FFs from trophoblast occurred later on. Eventually, in blastocysts' outgrowths, an equally high contribution from FFs progeny (60%) was found in both ICM and trophoblast. Three days after manipulation, FFs resumed proliferation in vitro. More than three FFs were found in 46.2% of embryos on day 4. On the 7th day in vitro in 70% of embryos more than 12 FFs were found, proving at least three cell divisions. To study postimplantation development, the embryos with FFs were transferred to pseudopregnant recipients a day after manipulation. After implantation, FFs were identified by electrophoresis for isozymes of glucose phosphate isomerase (GPI). A single 11-day embryo delayed to day 8 proved chimeric by expressing both donor isozyme GPI-1B and recipient GPI-1A. Similar chimerism was found in the extraembryonic lineage of 11% of embryos by day 12. Starting from day 11 onwards, in 32% of normal embryos and in 57% of foetal membranes, hybrid GPI-1AB isozyme, as well as recipient isozyme, was present. Hybrid GPI-1AB can only be produced in hybrid cells derived by cell fusion, therefore, we suggest that during postimplantation development, FFs are rescued by fusion with recipient cells. In the mice born, hybrid isozyme was found in several tissues, including brain, lung, gut and kidney. We conclude that somatic cells (FFs) can proliferate in early embryonic environment until early postimplantation stages. Foetuses and the mice born are chimeras between recipient cells and hybrid cells with contributions from the donor FFs. Transdifferentiation as opposed to reprogramming by cell fusion can be considered as underlying cellular processes in these chimeras.


Assuntos
Fase de Clivagem do Zigoto/fisiologia , Desenvolvimento Fetal/fisiologia , Fibroblastos/transplante , Animais , Biomarcadores/análise , Massa Celular Interna do Blastocisto/citologia , Diferenciação Celular , Proliferação de Células , Quimera , Fibroblastos/citologia , Glucose-6-Fosfato Isomerase/análise , Isoenzimas/análise , Camundongos , Microscopia de Fluorescência , Trofoblastos/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...