Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Annu Rev Cell Dev Biol ; 34: 405-426, 2018 10 06.
Article in English | MEDLINE | ID: mdl-30095292

ABSTRACT

We present an overview of symmetry breaking in early mammalian development as a continuous process from compaction to specification of the body axes. While earlier studies have focused on individual symmetry-breaking events, recent advances enable us to explore progressive symmetry breaking during early mammalian development. Although we primarily discuss embryonic development of the mouse, as it is the best-studied mammalian model system to date, we also highlight the shared and distinct aspects between different mammalian species. Finally, we discuss how insights gained from studying mammalian development can be generalized in light of self-organization principles. With this review, we hope to highlight new perspectives in studying symmetry breaking and self-organization in multicellular systems.


Subject(s)
Blastocyst/cytology , Body Patterning/genetics , Embryo, Mammalian , Embryonic Development/genetics , Cell Lineage/genetics , Humans
2.
EMBO J ; 42(17): e113280, 2023 09 04.
Article in English | MEDLINE | ID: mdl-37522872

ABSTRACT

Embryo implantation into the uterus marks a key transition in mammalian development. In mice, implantation is mediated by the trophoblast and is accompanied by a morphological transition from the blastocyst to the egg cylinder. However, the roles of trophoblast-uterine interactions in embryo morphogenesis during implantation are poorly understood due to inaccessibility in utero and the remaining challenges to recapitulate it ex vivo from the blastocyst. Here, we engineer a uterus-like microenvironment to recapitulate peri-implantation development of the whole mouse embryo ex vivo and reveal essential roles of the physical embryo-uterine interaction. We demonstrate that adhesion between the trophoblast and the uterine matrix is required for in utero-like transition of the blastocyst to the egg cylinder. Modeling the implanting embryo as a wetting droplet links embryo shape dynamics to the underlying changes in trophoblast adhesion and suggests that the adhesion-mediated tension release facilitates egg cylinder formation. Light-sheet live imaging and the experimental control of the engineered uterine geometry and trophoblast velocity uncovers the coordination between trophoblast motility and embryo growth, where the trophoblast delineates space for embryo morphogenesis.


Subject(s)
Blastocyst , Embryo Implantation , Female , Mice , Animals , Trophoblasts , Uterus , Embryonic Development , Mammals
3.
Development ; 149(1)2022 01 01.
Article in English | MEDLINE | ID: mdl-34908109

ABSTRACT

Development entails patterned emergence of diverse cell types within the embryo. In mammals, cells positioned inside the embryo give rise to the inner cell mass (ICM), which eventually forms the embryo itself. Yet, the molecular basis of how these cells recognise their 'inside' position to instruct their fate is unknown. Here, we show that provision of extracellular matrix (ECM) to isolated embryonic cells induces ICM specification and alters the subsequent spatial arrangement between epiblast (EPI) and primitive endoderm (PrE) cells that emerge within the ICM. Notably, this effect is dependent on integrin ß1 activity and involves apical-to-basal conversion of cell polarity. We demonstrate that ECM-integrin activity is sufficient for 'inside' positional signalling and is required for correct EPI/PrE patterning. Thus, our findings highlight the significance of ECM-integrin adhesion in enabling position sensing by cells to achieve tissue patterning.


Subject(s)
Body Patterning , Ectoderm/metabolism , Endoderm/metabolism , Extracellular Matrix/metabolism , Integrin beta1/metabolism , Signal Transduction , Animals , Cell Differentiation , Cell Polarity , Cells, Cultured , Ectoderm/cytology , Endoderm/cytology , Mice , Mice, Inbred C57BL , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism
4.
Nat Rev Mol Cell Biol ; 14(7): 452-9, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23778971

ABSTRACT

The mechanisms underlying the appearance of asymmetry between cells in the early embryo and consequently the specification of distinct cell lineages during mammalian development remain elusive. Recent experimental advances have revealed unexpected dynamics of and new complexity in this process. These findings can be integrated in a new unified framework that regards the early mammalian embryo as a self-organizing system.


Subject(s)
Cell Lineage , Embryo, Mammalian/cytology , Animals , Body Patterning , Cell Polarity , Computer Simulation , Embryonic Development , Gene Expression Regulation, Developmental , Humans , Models, Biological
5.
Nature ; 571(7763): 112-116, 2019 07.
Article in English | MEDLINE | ID: mdl-31189957

ABSTRACT

Size control is fundamental in tissue development and homeostasis1,2. Although the role of cell proliferation in these processes has been widely studied, the mechanisms that control embryo size-and how these mechanisms affect cell fate-remain unknown. Here we use the mouse blastocyst as a model to unravel a key role of fluid-filled lumen in the control of embryo size and specification of cell fate. We find that there is a twofold increase in lumenal pressure during blastocyst development, which translates into a concomitant increase in cell cortical tension and tissue stiffness of the trophectoderm that lines the lumen. Increased cortical tension leads to vinculin mechanosensing and maturation of functional tight junctions, which establishes a positive feedback loop to accommodate lumen growth. When the cortical tension reaches a critical threshold, cell-cell adhesion cannot be sustained during mitotic entry, which leads to trophectoderm rupture and blastocyst collapse. A simple theory of hydraulically gated oscillations recapitulates the observed dynamics of size oscillations, and predicts the scaling of embryo size with tissue volume. This theory further predicts that disrupted tight junctions or increased tissue stiffness lead to a smaller embryo size, which we verified by biophysical, embryological, pharmacological and genetic perturbations. Changes in lumenal pressure and size can influence the cell division pattern of the trophectoderm, and thereby affect cell allocation and fate. Our study reveals how lumenal pressure and tissue mechanics control embryo size at the tissue scale, which is coupled to cell position and fate at the cellular scale.


Subject(s)
Cell Differentiation , Cell Lineage , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Embryonic Development , Mechanotransduction, Cellular/physiology , Animals , Blastocyst/cytology , Cell Adhesion , Cell Division , Cell Shape , Embryo, Mammalian/anatomy & histology , Female , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Tight Junctions , Vinculin/metabolism
6.
Development ; 148(18)2021 03 12.
Article in English | MEDLINE | ID: mdl-33712442

ABSTRACT

Recognizing the crucial role of mechanical regulation and forces in tissue development and homeostasis has stirred a demand for in situ measurement of forces and stresses. Among emerging techniques, the use of cell geometry to infer cell junction tensions, cell pressures and tissue stress has gained popularity owing to the development of computational analyses. This approach is non-destructive and fast, and statistically validated based on comparisons with other techniques. However, its qualitative and quantitative limitations, in theory as well as in practice, should be examined with care. In this Primer, we summarize the underlying principles and assumptions behind stress inference, discuss its validity criteria and provide guidance to help beginners make the appropriate choice of its variants. We extend our discussion from two-dimensional stress inference to three dimensional, using the early mouse embryo as an example, and list a few possible extensions. We hope to make stress inference more accessible to the scientific community and trigger a broader interest in using this technique to study mechanics in development.


Subject(s)
Intercellular Junctions/physiology , Animals , Embryo, Mammalian/physiology , Mechanical Phenomena , Pressure , Stress, Mechanical
7.
Development ; 147(5)2020 03 02.
Article in English | MEDLINE | ID: mdl-32122910

ABSTRACT

Many developmental processes involve the emergence of intercellular fluid-filled lumina. This process of luminogenesis results in a build up of hydrostatic pressure and signalling molecules in the lumen. However, the potential roles of lumina in cellular functions, tissue morphogenesis and patterning have yet to be fully explored. In this Review, we discuss recent findings that describe how pressurized fluid expansion can provide both mechanical and biochemical cues to influence cell proliferation, migration and differentiation. We also review emerging techniques that allow for precise quantification of fluid pressure in vivo and in situ Finally, we discuss the intricate interplay between luminogenesis, tissue mechanics and signalling, which provide a new dimension for understanding the principles governing tissue self-organization in embryonic development.


Subject(s)
Embryonic Development/physiology , Mechanotransduction, Cellular/physiology , Osmotic Pressure/physiology , Stress, Mechanical , Animals , Biomechanical Phenomena , Cell Differentiation/physiology , Humans , Hydrostatic Pressure , Models, Biological
8.
Nature ; 536(7616): 344-348, 2016 08 18.
Article in English | MEDLINE | ID: mdl-27487217

ABSTRACT

During pre-implantation development, the mammalian embryo self-organizes into the blastocyst, which consists of an epithelial layer encapsulating the inner-cell mass (ICM) giving rise to all embryonic tissues. In mice, oriented cell division, apicobasal polarity and actomyosin contractility are thought to contribute to the formation of the ICM. However, how these processes work together remains unclear. Here we show that asymmetric segregation of the apical domain generates blastomeres with different contractilities, which triggers their sorting into inner and outer positions. Three-dimensional physical modelling of embryo morphogenesis reveals that cells internalize only when differences in surface contractility exceed a predictable threshold. We validate this prediction using biophysical measurements, and successfully redirect cell sorting within the developing blastocyst using maternal myosin (Myh9)-knockout chimaeric embryos. Finally, we find that loss of contractility causes blastomeres to show ICM-like markers, regardless of their position. In particular, contractility controls Yap subcellular localization, raising the possibility that mechanosensing occurs during blastocyst lineage specification. We conclude that contractility couples the positioning and fate specification of blastomeres. We propose that this ensures the robust self-organization of blastomeres into the blastocyst, which confers remarkable regulative capacities to mammalian embryos.


Subject(s)
Blastocyst Inner Cell Mass/cytology , Cell Differentiation , Cell Division , Cell Movement , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Blastocyst/cytology , Blastomeres/cytology , Cell Cycle Proteins , Cell Lineage , Cell Polarity , Embryonic Development , Female , Male , Mice , Phosphoproteins/metabolism , Protein Transport , Reproducibility of Results , YAP-Signaling Proteins
9.
Genes Dev ; 26(13): 1445-58, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22713603

ABSTRACT

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


Subject(s)
Endoderm/metabolism , GATA6 Transcription Factor/metabolism , Nuclear Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism , Animals , Cell Lineage , Endoderm/cytology , GATA6 Transcription Factor/genetics , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice , Nanog Homeobox Protein , Nuclear Proteins/genetics , Pluripotent Stem Cells/metabolism , Polycomb Repressive Complex 1 , Proto-Oncogene Proteins/genetics , Repressor Proteins/genetics , Transcription, Genetic
10.
Nat Methods ; 13(2): 139-42, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26657559

ABSTRACT

Despite its importance for understanding human infertility and congenital diseases, early mammalian development has remained inaccessible to in toto imaging. We developed an inverted light-sheet microscope that enabled us to image mouse embryos from zygote to blastocyst, computationally track all cells and reconstruct a complete lineage tree of mouse pre-implantation development. We used this unique data set to show that the first cell fate specification occurs at the 16-cell stage.


Subject(s)
Blastocyst/cytology , Microscopy/instrumentation , Microscopy/methods , Animals , Imaging, Three-Dimensional/instrumentation , Imaging, Three-Dimensional/methods , Mice , Time-Lapse Imaging/instrumentation , Time-Lapse Imaging/methods
11.
EMBO Rep ; 16(8): 1005-21, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26142281

ABSTRACT

Mammalian development begins with the segregation of embryonic and extra-embryonic lineages in the blastocyst. Recent studies revealed cell-to-cell gene expression heterogeneity and dynamic cell rearrangements during mouse blastocyst formation. Thus, mechanistic understanding of lineage specification requires quantitative description of gene expression dynamics at a single-cell resolution in living embryos. However, only a few fluorescent gene expression reporter mice are available and quantitative live image analysis is limited so far. Here, we carried out a fluorescence gene-trap screen and established reporter mice expressing Venus specifically in the first lineages. Lineage tracking, quantitative gene expression and cell position analyses allowed us to build a comprehensive lineage map of mouse pre-implantation development. Our systematic analysis revealed that, contrary to the available models, the timing and mechanism of lineage specification may be distinct between the trophectoderm and the inner cell mass. While expression of our trophectoderm-specific lineage marker is upregulated in outside cells upon asymmetric divisions at 8- and 16-cell stages, the inside-specific upregulation of the inner-cell-mass marker only becomes evident at the 64-cell stage. This study thus provides a framework toward systems-level understanding of embryogenesis marked by high dynamicity and stochastic variability.


Subject(s)
Blastocyst/physiology , Cell Lineage , Embryonic Development , Animals , Embryo Implantation , Gene Expression Regulation, Developmental , Genes, Reporter , Intravital Microscopy , Mice
12.
Nat Cell Biol ; 26(4): 530-541, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38499770

ABSTRACT

Embryonic induction is a key mechanism in development that corresponds to an interaction between a signalling and a responding tissue, causing a change in the direction of differentiation by the responding tissue. Considerable progress has been achieved in identifying inductive signals, yet how tissues control their responsiveness to these signals, known as competence, remains poorly understood. While the role of molecular signals in competence has been studied, how tissue mechanics influence competence remains unexplored. Here we investigate the role of hydrostatic pressure in controlling competence in neural crest cells, an embryonic cell population. We show that neural crest competence decreases concomitantly with an increase in the hydrostatic pressure of the blastocoel, an embryonic cavity in contact with the prospective neural crest. By manipulating hydrostatic pressure in vivo, we show that this increase leads to the inhibition of Yap signalling and impairs Wnt activation in the responding tissue, which would be required for neural crest induction. We further show that hydrostatic pressure controls neural crest induction in amphibian and mouse embryos and in human cells, suggesting a conserved mechanism across vertebrates. Our work sets out how tissue mechanics can interplay with signalling pathways to regulate embryonic competence.


Subject(s)
Embryonic Induction , Neural Crest , Animals , Humans , Mice , Hydrostatic Pressure , Neural Crest/metabolism , Prospective Studies , Wnt Proteins/metabolism
13.
J Biol Chem ; 287(30): 25361-9, 2012 Jul 20.
Article in English | MEDLINE | ID: mdl-22605343

ABSTRACT

The study of chromatin and its regulators is key to understanding and manipulating transcription. We previously exploited the Krüppel-associated box (KRAB) transcriptional repressor domain, present in hundreds of vertebrate-specific zinc finger proteins, to assess the effect of its binding to gene bodies. These experiments revealed that the ectopic and doxycycline (dox)-controlled tet repressor KRAB fusion protein (tTRKRAB) can induce reversible and long-range silencing of cellular promoters. Here, we extend this system to in vivo applications and use tTRKRAB to achieve externally controllable repression of an endogenous mouse locus. We employed lentiviral-mediated transgenesis with promoterless TetO-containing gene traps to engineer a mouse line where the endogenous kinesin family member 2A (Kif2A) promoter drives a YFP reporter gene. When these mice were crossed to animals expressing the TetO-binding tTRKRAB repressor, this regulator was recruited to the Kif2A locus, and YFP expression was reduced. This effect was reversed when dox was given to embryos or adult mice, demonstrating that the cellular Kif2A promoter was only silenced upon repressor binding. Molecular analyses confirmed that tTRKRAB induced transcriptional repression through the spread of H3K9me3-containing heterochromatin, without DNA methylation of the trapped Kif2A promoter. Therefore, we demonstrate that targeting of tTRKRAB to a gene body in vivo results in reversible transcriptional repression through the spreading of facultative heterochromatin. This finding not only sheds light on KRAB-mediated transcriptional processes, but also suggests approaches for the externally controllable and reversible modulation of chromatin and transcription in vivo.


Subject(s)
Carrier Proteins/metabolism , Chromatin Assembly and Disassembly/physiology , Genetic Loci/physiology , Nuclear Proteins/metabolism , Promoter Regions, Genetic/physiology , Repressor Proteins/metabolism , Transcription, Genetic/physiology , Animals , Carrier Proteins/genetics , Heterochromatin/genetics , Heterochromatin/metabolism , Kinesins/biosynthesis , Kinesins/genetics , Mice , Mice, Transgenic , Nuclear Proteins/genetics , Protein Structure, Tertiary , Repressor Proteins/biosynthesis , Repressor Proteins/genetics
14.
Nature ; 442(7099): E3-4; discussion E4, 2006 Jul 13.
Article in English | MEDLINE | ID: mdl-16837972

ABSTRACT

A recurring question in developmental biology has been whether localized determinants play any role in mammalian preimplantation development. This is a controversial issue that brings back the idea of prepatterning and is explored further by Plusa et al., who claim it is the first cleavage of the mouse zygote that predicts the blastocyst axis, rather than the animal pole or sperm entry point, as previously suggested. However, other evidence indicates that the blasotcyst axis is not predetermined and there is no prepatterning in the mouse egg. Here we investigate the origin of these different views and conclude that they arise from differences in the data themselves and in their interpretation.


Subject(s)
Blastocyst/cytology , Body Patterning , Embryonic Development , Zygote/growth & development , Animals , Blastomeres/cytology , Cell Lineage , Cell Polarity , Cleavage Stage, Ovum/cytology , Mice , Models, Biological , Zygote/cytology
15.
Dev Cell ; 57(11): 1311-1313, 2022 06 06.
Article in English | MEDLINE | ID: mdl-35671703

ABSTRACT

Embryonic cells of the early mouse embryo become hypersensitive to apoptotic stimuli before gastrulation. In this issue of Developmental Cell, Pernaute et al. show that this switch in sensitivity is a result of a change in mitochondrial dynamics and mitophagy levels controlled by DRP1, a regulator of mitochondrial fission.


Subject(s)
Dynamins , Mitochondrial Dynamics , Animals , Mice , Mitochondrial Proteins , Mitophagy
16.
Dev Cell ; 57(3): 373-386.e9, 2022 02 07.
Article in English | MEDLINE | ID: mdl-35063082

ABSTRACT

Upon implantation, mammalian embryos undergo major morphogenesis and key developmental processes such as body axis specification and gastrulation. However, limited accessibility obscures the study of these crucial processes. Here, we develop an ex vivo Matrigel-collagen-based culture to recapitulate mouse development from E4.5 to E6.0. Our system not only recapitulates embryonic growth, axis initiation, and overall 3D architecture in 49% of the cases, but its compatibility with light-sheet microscopy also enables the study of cellular dynamics through automatic cell segmentation. We find that, upon implantation, release of the increasing tension in the polar trophectoderm is necessary for its constriction and invagination. The resulting extra-embryonic ectoderm plays a key role in growth, morphogenesis, and patterning of the neighboring epiblast, which subsequently gives rise to all embryonic tissues. This 3D ex vivo system thus offers unprecedented access to peri-implantation development for in toto monitoring, measurement, and spatiotemporally controlled perturbation, revealing a mechano-chemical interplay between extra-embryonic and embryonic tissues.


Subject(s)
Embryo Implantation , Embryo, Mammalian/cytology , Embryonic Development , Animals , Body Patterning , Ectoderm/cytology , Machine Learning , Mice, Inbred C57BL , Microsurgery , Morphogenesis , Trophoblasts/cytology
17.
Curr Biol ; 32(21): 4707-4718.e8, 2022 11 07.
Article in English | MEDLINE | ID: mdl-36115340

ABSTRACT

Development is a highly dynamic process in which organisms often experience changes in both form and behavior, which are typically coupled to each other. However, little is known about how organismal-scale behaviors such as body contractility and motility impact morphogenesis. Here, we use the cnidarian Nematostella vectensis as a developmental model to uncover a mechanistic link between organismal size, shape, and behavior. Using quantitative live imaging in a large population of developing animals, combined with molecular and biophysical experiments, we demonstrate that the muscular-hydraulic machinery that controls body movement also drives larva-polyp morphogenesis. We show that organismal size largely depends on cavity inflation through fluid uptake, whereas body shape is constrained by the organization of the muscular system. The generation of ethograms identifies different trajectories of size and shape development in sessile and motile animals, which display distinct patterns of body contractions. With a simple theoretical model, we conceptualize how pressures generated by muscular hydraulics can act as a global mechanical regulator that coordinates tissue remodeling. Altogether, our findings illustrate how organismal contractility and motility behaviors can influence morphogenesis.


Subject(s)
Sea Anemones , Animals , Larva , Morphogenesis
18.
Nat Cell Biol ; 23(7): 733-744, 2021 07.
Article in English | MEDLINE | ID: mdl-34155381

ABSTRACT

Intestinal organoids derived from single cells undergo complex crypt-villus patterning and morphogenesis. However, the nature and coordination of the underlying forces remains poorly characterized. Here, using light-sheet microscopy and large-scale imaging quantification, we demonstrate that crypt formation coincides with a stark reduction in lumen volume. We develop a 3D biophysical model to computationally screen different mechanical scenarios of crypt morphogenesis. Combining this with live-imaging data and multiple mechanical perturbations, we show that actomyosin-driven crypt apical contraction and villus basal tension work synergistically with lumen volume reduction to drive crypt morphogenesis, and demonstrate the existence of a critical point in differential tensions above which crypt morphology becomes robust to volume changes. Finally, we identified a sodium/glucose cotransporter that is specific to differentiated enterocytes that modulates lumen volume reduction through cell swelling in the villus region. Together, our study uncovers the cellular basis of how cell fate modulates osmotic and actomyosin forces to coordinate robust morphogenesis.


Subject(s)
Cell Differentiation , Cell Lineage , Intestinal Mucosa/physiology , Mechanotransduction, Cellular , Osmoregulation , Paneth Cells/physiology , Stem Cells/physiology , Animals , Cell Movement , Cells, Cultured , Computer Simulation , Female , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Confocal , Microscopy, Video , Models, Biological , Morphogenesis , Myosin Type II/genetics , Myosin Type II/metabolism , Organoids , Osmotic Pressure , Paneth Cells/metabolism , Sodium-Glucose Transport Proteins/genetics , Sodium-Glucose Transport Proteins/metabolism , Stem Cells/metabolism , Stress, Mechanical , Time Factors
19.
Dev Cell ; 56(23): 3185-3191, 2021 12 06.
Article in English | MEDLINE | ID: mdl-34875224

ABSTRACT

In our 20th anniversary year, we reflect on how fields have changed since our first issue and here look to the future. In this collection of Voices, our writers speculate on the future: in terms of philosophy, cell states, cell processes, and then how to model cell systems.


Subject(s)
Cell Biology , Developmental Biology , Periodicals as Topic/statistics & numerical data , Humans , Time Factors
20.
Nature ; 430(6997): 360-4, 2004 Jul 15.
Article in English | MEDLINE | ID: mdl-15254539

ABSTRACT

Studies of experimentally manipulated embryos have led to the long-held conclusion that the polarity of the mouse embryo remains undetermined until the blastocyst stage. However, recent studies reporting that the embryonic-abembryonic axis of the blastocyst arises perpendicular to the first cleavage plane, and hence to the animal-vegetal axis of the zygote, have led to the claim that the axis of the mouse embryo is already specified in the egg. Here we show that there is no specification of the axis in the egg. Time-lapse recordings show that the second polar body does not mark a stationary animal pole, but instead, in half of the embryos, moves towards a first cleavage plane. The first cleavage plane coincides with the plane defined by the two apposing pronuclei once they have moved to the centre of the egg. Pronuclear transfer experiments confirm that the first cleavage plane is not determined in early interphase but rather is specified by the newly formed topology of the two pronuclei. The microtubule networks that allow mixing of parental chromosomes before dividing into two may be involved in these processes.


Subject(s)
Cell Nucleus/physiology , Cell Polarity , Cellular Structures/physiology , Zygote/cytology , Actins/metabolism , Animals , Cell Division/drug effects , Chorionic Gonadotropin/pharmacology , Chromosomes, Mammalian/drug effects , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Embryo, Mammalian/cytology , Embryo, Mammalian/drug effects , Embryo, Mammalian/metabolism , Female , Humans , Interphase/drug effects , Male , Meiosis/physiology , Mice , Zygote/drug effects , Zygote/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL