Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS Biol ; 19(7): e3001345, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34310594

RESUMO

Architectural changes at the cellular and organism level are integral and necessary to successful development and growth. During mammalian preimplantation development, cells reduce in size and the architecture of the embryo changes significantly. Such changes must be coordinated correctly to ensure continued development of the embryo and, ultimately, a successful pregnancy. However, the nature of such transformations is poorly defined during mammalian preimplantation development. In order to quantitatively describe changes in cell environment and organism architecture, we designed Internal Versus External Neighbourhood (IVEN). IVEN is a user-interactive, open-source pipeline that classifies cells into different populations based on their position and quantifies the number of neighbours of every cell within a dataset in a 3D environment. Through IVEN-driven analyses, we show how transformations in cell environment, defined here as changes in cell neighbourhood, are related to changes in embryo geometry and major developmental events during preimplantation mammalian development. Moreover, we demonstrate that modulation of the FGF pathway alters spatial relations of inner cells and neighbourhood distributions, leading to overall changes in embryo architecture. In conjunction with IVEN-driven analyses, we uncover differences in the dynamic of cell size changes over the preimplantation period and determine that cells within the mammalian embryo initiate growth phase only at the time of implantation.


Assuntos
Blastocisto/citologia , Animais , Tamanho Celular , Desenvolvimento Embrionário , Feminino , Camundongos , Gravidez
2.
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
3.
Int J Dev Biol ; 63(3-4-5): 77-82, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31058304

RESUMO

In recent decades we have witnessed unprecedented progress in the field of the developmental biology of mammals. Building on 20th century discoveries, we have managed to increase our understanding of the molecular and cellular mechanisms governing early mammalian embryogenesis and link them to other biological questions, such as stem cells, regeneration, cancer, or tissue and organ formation. Consequently, it has also led to a creation of a completely new branch of reproductive medicine, i.e. assisted reproductive technology (ART). In this Special Issue of The International Journal of Developmental Biology (Int. J. Dev. Biol.) we wished to review state-of-the-art research regarding early mammalian development, from fertilization up to the implantation stage, and discuss its potential meaning for practical applications, including ART. As an introduction to the issue we present a compilation of short essays written by the most renowned scientists in the field, working both in basic and clinical research. The essays are dedicated to the greatest breakthroughs and challenges of 21st century developmental biology and reproductive medicine.


Assuntos
Biologia do Desenvolvimento/história , Medicina Reprodutiva/história , Animais , Linhagem da Célula , Biologia do Desenvolvimento/tendências , Implantação do Embrião , Células-Tronco Embrionárias , Fertilização , História do Século XX , História do Século XXI , Humanos , Medicina Reprodutiva/tendências , Técnicas de Reprodução Assistida/história , Técnicas de Reprodução Assistida/tendências
4.
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
6.
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
8.
Cell Rep ; 15(10): 2185-2199, 2016 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-27239041

RESUMO

It is now well-established that hematopoietic stem cells (HSCs) and progenitor cells originate from a specialized subset of endothelium, termed hemogenic endothelium (HE), via an endothelial-to-hematopoietic transition. However, the molecular mechanisms determining which endothelial progenitors possess this hemogenic potential are currently unknown. Here, we investigated the changes in hemogenic potential in endothelial progenitors at the early stages of embryonic development. Using an ETV2::GFP reporter mouse to isolate emerging endothelial progenitors, we observed a dramatic decrease in hemogenic potential between embryonic day (E)7.5 and E8.5. At the molecular level, Runx1 is expressed at much lower levels in E8.5 intra-embryonic progenitors, while Bmi1 expression is increased. Remarkably, the ectopic expression of Runx1 in these progenitors fully restores their hemogenic potential, as does the suppression of BMI1 function. Altogether, our data demonstrate that hemogenic competency in recently specified endothelial progenitors is restrained through the active silencing of Runx1 expression.


Assuntos
Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Desenvolvimento Embrionário , Células Progenitoras Endoteliais/metabolismo , Inativação Gênica , Hemangioblastos/citologia , Animais , Proteína Morfogenética Óssea 4/metabolismo , Embrião de Mamíferos/metabolismo , Desenvolvimento Embrionário/genética , Células Progenitoras Endoteliais/citologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Fluorescência Verde/metabolismo , Hemangioblastos/metabolismo , Hematopoese/genética , Imunofenotipagem , Masculino , Camundongos Endogâmicos ICR , Análise de Sequência com Séries de Oligonucleotídeos , Complexo Repressor Polycomb 1/antagonistas & inibidores , Complexo Repressor Polycomb 1/metabolismo , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/metabolismo , Análise de Célula Única , Proteínas Smad/metabolismo , Fatores de Transcrição/metabolismo
9.
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
10.
Semin Cell Dev Biol ; 47-48: 92-100, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26183190

RESUMO

Embryonic development is a complex and highly dynamic process during which individual cells interact with one another, adopt different identities and organize themselves in three-dimensional space to generate an entire organism. Recent technical developments in genomics and high-resolution quantitative imaging are making it possible to study cellular populations at single-cell resolution and begin to integrate different inputs, for example genetic, physical and chemical factors, that affect cell differentiation over spatial and temporal scales. The preimplantation mouse embryo allows the analysis of cell fate decisions in vivo with high spatiotemporal resolution. In this review we highlight how the application of live imaging and single-cell resolution analysis pipelines is providing an unprecedented level of insight on the processes that shape the earliest stages of mammalian development.


Assuntos
Blastocisto/fisiologia , Comunicação Celular/fisiologia , Diferenciação Celular/fisiologia , Desenvolvimento Embrionário/fisiologia , Animais , Blastocisto/citologia , Blastocisto/metabolismo , Comunicação Celular/genética , Diferenciação Celular/genética , Desenvolvimento Embrionário/genética , Regulação da Expressão Gênica no Desenvolvimento , Hibridização in Situ Fluorescente/métodos , Camundongos , Reprodutibilidade dos Testes , Análise de Célula Única/métodos
11.
Nat Cell Biol ; 16(6): 502-4, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24875737

RESUMO

Pluripotent embryonic stem cells (ESCs) can be derived from blastocyst-stage mouse embryos. However, the exact in vivo counterpart of ESCs has remained elusive. A combination of expression profiling and stem cell derivation identifies epiblast cells from late-stage blastocysts as the source, and functional equivalent, of ESCs.


Assuntos
Massa Celular Interna do Blastocisto/metabolismo , Diferenciação Celular , Linhagem da Célula , Proliferação de Células , Células-Tronco Embrionárias/metabolismo , Camadas Germinativas/enzimologia , Células-Tronco Pluripotentes/metabolismo , Fatores de Transcrição/metabolismo , Animais
12.
Development ; 140(21): 4311-22, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24067354

RESUMO

During mouse pre-implantation development, extra-embryonic primitive endoderm (PrE) and pluripotent epiblast precursors are specified in the inner cell mass (ICM) of the early blastocyst in a 'salt and pepper' manner, and are subsequently sorted into two distinct layers. Positional cues provided by the blastocyst cavity are thought to be instrumental for cell sorting; however, the sequence of events and the mechanisms that control this segregation remain unknown. Here, we show that atypical protein kinase C (aPKC), a protein associated with apicobasal polarity, is specifically enriched in PrE precursors in the ICM prior to cell sorting and prior to overt signs of cell polarisation. aPKC adopts a polarised localisation in PrE cells only after they reach the blastocyst cavity and form a mature epithelium, in a process that is dependent on FGF signalling. To assess the role of aPKC in PrE formation, we interfered with its activity using either chemical inhibition or RNAi knockdown. We show that inhibition of aPKC from the mid blastocyst stage not only prevents sorting of PrE precursors into a polarised monolayer but concomitantly affects the maturation of PrE precursors. Our results suggest that the processes of PrE and epiblast segregation, and cell fate progression are interdependent, and place aPKC as a central player in the segregation of epiblast and PrE progenitors in the mouse blastocyst.


Assuntos
Massa Celular Interna do Blastocisto/citologia , Blastocisto/enzimologia , Blastocisto/fisiologia , Células-Tronco Embrionárias/metabolismo , Endoderma/fisiologia , Proteína Quinase C/metabolismo , Animais , Linhagem da Célula/fisiologia , Polaridade Celular/fisiologia , Primers do DNA/genética , Endoderma/citologia , Fatores de Crescimento de Fibroblastos/metabolismo , Imunofluorescência , Processamento de Imagem Assistida por Computador , Camundongos , Microscopia Confocal , Proteína Quinase C/genética , Interferência de RNA
13.
BMC Dev Biol ; 13: 32, 2013 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-23941255

RESUMO

BACKGROUND: Preimplantation bovine development is emerging as an attractive experimental model, yet little is known about the mechanisms underlying trophoblast (TE)/inner cell mass (ICM) segregation in cattle. To gain an insight into these processes we have studied protein and mRNA distribution during the crucial stages of bovine development. Protein distribution of lineage specific markers OCT4, NANOG, CDX2 were analysed in 5-cell, 8-16 cell, morula and blastocyst stage embryos. ICM/TE mRNA levels were compared in hatched blastocysts and included: OCT4, NANOG, FN-1, KLF4, c-MYC, REX1, CDX2, KRT-18 and GATA6. RESULTS: At the mRNA level the observed distribution patterns agree with the mouse model. CDX2 and OCT4 proteins were first detected in 5-cell stage embryos. NANOG appeared at the morula stage and was located in the cytoplasm forming characteristic rings around the nuclei. Changes in sub-cellular localisation of OCT4, NANOG and CDX2 were noted from the 8-16 cell onwards. CDX2 initially co-localised with OCT4, but at the blastocyst stage a clear lineage segregation could be observed. Interestingly, we have observed in a small proportion of embryos (2%) that CDX2 immunolabelling overlapped with mitotic chromosomes. CONCLUSIONS: Cell fate specification in cattle become evident earlier than presently anticipated - around the time of bovine embryonic genome activation. There is an intriguing possibility that for proper lineage determination certain transcription factors (such as CDX2) may need to occupy specific regions of chromatin prior to its activation in the interphase nucleus. Our observation suggests a possible role of CDX2 in the process of epigenetic regulation of embryonic cell fate.


Assuntos
Blastocisto , Frações Subcelulares/metabolismo , Fatores de Transcrição/metabolismo , Trofoblastos/metabolismo , Animais , Sequência de Bases , Bovinos , Primers do DNA , Regulação da Expressão Gênica no Desenvolvimento , Fator 4 Semelhante a Kruppel , Reação em Cadeia da Polimerase em Tempo Real
14.
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
15.
Reproduction ; 145(3): R65-80, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23221010

RESUMO

During mammalian preimplantation development, the fertilised egg gives rise to a group of pluripotent embryonic cells, the epiblast, and to the extraembryonic lineages that support the development of the foetus during subsequent phases of development. This preimplantation period not only accommodates the first cell fate decisions in a mammal's life but also the transition from a totipotent cell, the zygote, capable of producing any cell type in the animal, to cells with a restricted developmental potential. The cellular and molecular mechanisms governing the balance between developmental potential and lineage specification have intrigued developmental biologists for decades. The preimplantation mouse embryo offers an invaluable system to study cell differentiation as well as the emergence and maintenance of pluripotency in the embryo. Here we review the most recent findings on the mechanisms controlling these early cell fate decisions. The model that emerges from the current evidence indicates that cell differentiation in the preimplantation embryo depends on cellular interaction and intercellular communication. This strategy underlies the plasticity of the early mouse embryo and ensures the correct specification of the first mammalian cell lineages.


Assuntos
Blastocisto/fisiologia , Diferenciação Celular , Linhagem da Célula , Células-Tronco Embrionárias/fisiologia , Células-Tronco Pluripotentes/fisiologia , Animais , Blastocisto/citologia , Blastocisto/metabolismo , Massa Celular Interna do Blastocisto/fisiologia , Comunicação Celular , Células-Tronco Embrionárias/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Morfogênese , Células-Tronco Pluripotentes/metabolismo , Transdução de Sinais , Fatores de Transcrição/metabolismo
16.
Methods Mol Biol ; 916: 275-85, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22914948

RESUMO

The separation of two populations of cells-primitive endoderm and epiblast-within the inner cell mass (ICM) of the mammalian blastocyst is a crucial event during preimplantation development. However, many aspects of this process are still not very well understood. Recently, the identification of platelet derived growth factor receptor alpha (Pdgfrα) as an early-expressed protein that is also a marker of the later primitive endoderm lineage, together with the availability of the Pdgfra(H2B-GFP) mouse strain (Hamilton et al. Mol Cell Biol 23:4013-4025, 2003), has made in vivo imaging of primitive endoderm formation possible. In this chapter we present two different approaches that can be used to follow the behavior of primitive endoderm cells within the mouse blastocyst in real time.


Assuntos
Blastocisto/citologia , Endoderma/citologia , Imagem Molecular/métodos , Sobrevivência de Tecidos , Animais , Feminino , Masculino , Camundongos , Fatores de Tempo
17.
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
18.
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
19.
Nature ; 434(7031): 391-5, 2005 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-15772664

RESUMO

One of the unanswered questions in mammalian development is how the embryonic-abembryonic axis of the blastocyst is first established. It is possible that the first cleavage division contributes to this process, because in most mouse embryos the progeny of one two-cell blastomere primarily populate the embryonic part of the blastocyst and the progeny of its sister populate the abembryonic part. However, it is not known whether the embryonic-abembryonic axis is set up by the first cleavage itself, by polarity in the oocyte that then sets the first cleavage plane with respect to the animal pole, or indeed whether it can be divorced entirely from the first cleavage and established in relation to the animal pole. Here we test the importance of the orientation of the first cleavage by imposing an elongated shape on the zygote so that the division no longer passes close to the animal pole, marked by the second polar body. Non-invasive lineage tracing shows that even when the first cleavage occurs along the short axis imposed by this experimental treatment, the progeny of the resulting two-cell blastomeres tend to populate the respective embryonic and abembryonic parts of the blastocyst. Thus, the first cleavage contributes to breaking the symmetry of the embryo, generating blastomeres with different developmental characteristics.


Assuntos
Blastocisto/citologia , Padronização Corporal , Zigoto/citologia , Zigoto/crescimento & desenvolvimento , Animais , Blastocisto/efeitos dos fármacos , Blastômeros/citologia , Blastômeros/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Cromatina/metabolismo , Citocalasina D/farmacologia , Feminino , Fertilização , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Zigoto/efeitos dos fármacos
20.
Mol Reprod Dev ; 70(4): 429-37, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15685637

RESUMO

The aim of this study was to investigate the fate of an additional female genome introduced to a dividing zygote. Maternal chromatin in the form of karyoplasts containing a metaphase II spindle were fused to zygotes blocked in anaphase or telophase of the first cleavage. Permanent preparations made 20-40 min after fusion at anaphase revealed that the donor maternal chromosomes had entered anaphase or telophase in 16 out of 18 cases. A further two groups of embryos that were fused at either anaphase or anaphase/telophase were cultured to the first division. Division occurred 50 min after fusion in both groups of embryos (86 and 85.1%, respectively), of which most divided to two cells (80 and 71.6% of total) and the remainder divided to three cells. About two thirds of two-cell embryos contained an extra nucleus in one blastomere. Nuclei containing donor maternal chromosomes reached a similar size to recipient nuclei in 68% of embryos derived from anaphase-blocked zygotes, in contrast to 31.1% of embryos derived from anaphase/telophase-blocked embryos. Replication of DNA in donor nuclei closely followed the timing and intensity of that in control embryos. When fixed 24 hr after fusion, one third of embryos were still at the two-cell stage, with one or both blastomeres showing a single metaphase plate of the second cleavage. In the remaining embryos, three or four cells were present, some containing two nuclei. Blastocysts developed in 50% of fused embryos and three young were born after transfer of cleaving hybrid embryos to recipients. Chromosome preparations from bone marrow of the young contained 3-4 tetraploid metaphase plates per several hundred plates counted compared with none in control embryos. In conclusion, additional maternal chromosomes can be introduced at the late-dividing zygote and join the embryonic cell cycles during subsequent divisions. This method may provide a useful approach for studying changes specific to the maternal genome during early cell cycles of the mammalian embryo.


Assuntos
Cromossomos/fisiologia , Fase de Clivagem do Zigoto/fisiologia , Meiose/fisiologia , Zigoto/fisiologia , Animais , Feminino , Fluoresceína-5-Isotiocianato , Corantes Fluorescentes , Masculino , Meiose/genética , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Mitose/genética , Mitose/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...