Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Cell Sci ; 137(7)2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38469748

RESUMEN

Equal cell division relies upon astral microtubule-based centering mechanisms, yet how the interplay between mitotic entry, cortical force generation and long astral microtubules leads to symmetric cell division is not resolved. We report that a cortically located sperm aster displaying long astral microtubules that penetrate the whole zygote does not undergo centration until mitotic entry. At mitotic entry, we find that microtubule-based cortical pulling is lost. Quantitative measurements of cortical pulling and cytoplasmic pulling together with physical simulations suggested that a wavelike loss of cortical pulling at mitotic entry leads to aster centration based on cytoplasmic pulling. Cortical actin is lost from the cortex at mitotic entry coincident with a fall in cortical tension from ∼300pN/µm to ∼100pN/µm. Following the loss of cortical force generators at mitotic entry, long microtubule-based cytoplasmic pulling is sufficient to displace the aster towards the cell center. These data reveal how mitotic aster centration is coordinated with mitotic entry in chordate zygotes.


Asunto(s)
Semen , Huso Acromático , Masculino , Humanos , Microtúbulos , Citoplasma , División Celular
2.
Development ; 150(1)2023 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-36515557

RESUMEN

The spindle assembly checkpoint (SAC) is a surveillance system that preserves genome integrity by delaying anaphase onset until all chromosomes are correctly attached to spindle microtubules. Recruitment of SAC proteins to unattached kinetochores generates an inhibitory signal that prolongs mitotic duration. Chordate embryos are atypical in that spindle defects do not delay mitotic progression during early development, implying that either the SAC is inactive or the cell-cycle target machinery is unresponsive. Here, we show that in embryos of the chordate Phallusia mammillata, the SAC delays mitotic progression from the 8th cleavage divisions. Unattached kinetochores are not recognized by the SAC machinery until the 7th cell cycle, when the SAC is acquired. After acquisition, SAC strength, which manifests as the degree of mitotic lengthening induced by spindle perturbations, is specific to different cell types and is modulated by cell size, showing similarity to SAC control in early Caenorhabditis elegans embryos. We conclude that SAC acquisition is a process that is likely specific to chordate embryos, while modulation of SAC efficiency in SAC proficient stages depends on cell fate and cell size, which is similar to non-chordate embryos.


Asunto(s)
Puntos de Control de la Fase M del Ciclo Celular , Huso Acromático , Animales , Huso Acromático/metabolismo , Cinetocoros/metabolismo , Microtúbulos/metabolismo , Caenorhabditis elegans/metabolismo , Tamaño de la Célula , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo
3.
Nat Methods ; 20(12): 1989-1999, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38057527

RESUMEN

Tissue morphogenesis results from a tight interplay between gene expression, biochemical signaling and mechanics. Although sequencing methods allow the generation of cell-resolved spatiotemporal maps of gene expression, creating similar maps of cell mechanics in three-dimensional (3D) developing tissues has remained a real challenge. Exploiting the foam-like arrangement of cells, we propose a robust end-to-end computational method called 'foambryo' to infer spatiotemporal atlases of cellular forces from fluorescence microscopy images of cell membranes. Our method generates precise 3D meshes of cells' geometry and successively predicts relative cell surface tensions and pressures. We validate it with 3D foam simulations, study its noise sensitivity and prove its biological relevance in mouse, ascidian and worm embryos. 3D force inference allows us to recover mechanical features identified previously, but also predicts new ones, unveiling potential new insights on the spatiotemporal regulation of cell mechanics in developing embryos. Our code is freely available and paves the way for unraveling the unknown mechanochemical feedbacks that control embryo and tissue morphogenesis.


Asunto(s)
Embrión de Mamíferos , Transducción de Señal , Animales , Ratones , Morfogénesis , Membrana Celular , Microscopía Fluorescente
4.
Mol Reprod Dev ; 86(10): 1333-1347, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31215734

RESUMEN

Endocrine Disrupting Chemicals (EDCs) are molecules able to interfere with the vertebrate hormonal system in different ways, a major one being the modification of the activity of nuclear receptors (NRs). Several NRs are expressed in the vertebrate brain during embryonic development and these NRs are suspected to be responsible for the neurodevelopmental defects induced by exposure to EDCs in fishes or amphibians and to participate in several neurodevelopmental disorders observed in humans. Known EDCs exert toxicity not only on vertebrate forms of marine life but also on marine invertebrates. However, because hormonal systems of invertebrates are poorly understood, it is not clear whether the teratogenic effects of known EDCs are because of endocrine disruption. The most conserved actors of endocrine systems are the NRs which are present in all metazoan genomes but their functions in invertebrate organisms are still insufficiently characterized. EDCs like bisphenol A have recently been shown to affect neurodevelopment in marine invertebrate chordates called ascidians. Because such phenotypes can be mediated by NRs expressed in the ascidian embryo, we review all the information available about NRs expression during ascidian embryogenesis and discuss their possible involvement in the neurodevelopmental phenotypes induced by EDCs.


Asunto(s)
Disruptores Endocrinos/toxicidad , Sistema Nervioso , Neurotoxinas/toxicidad , Receptores Citoplasmáticos y Nucleares/metabolismo , Urocordados , Animales , Embrión no Mamífero/efectos de los fármacos , Desarrollo Embrionario/efectos de los fármacos , Modelos Biológicos , Sistema Nervioso/efectos de los fármacos , Sistema Nervioso/embriología , Sistema Nervioso/crecimiento & desarrollo , Urocordados/efectos de los fármacos , Urocordados/embriología , Urocordados/crecimiento & desarrollo
5.
Nucleic Acids Res ; 44(D1): D808-18, 2016 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-26420834

RESUMEN

Ascidians belong to the tunicates, the sister group of vertebrates and are recognized model organisms in the field of embryonic development, regeneration and stem cells. ANISEED is the main information system in the field of ascidian developmental biology. This article reports the development of the system since its initial publication in 2010. Over the past five years, we refactored the system from an initial custom schema to an extended version of the Chado schema and redesigned all user and back end interfaces. This new architecture was used to improve and enrich the description of Ciona intestinalis embryonic development, based on an improved genome assembly and gene model set, refined functional gene annotation, and anatomical ontologies, and a new collection of full ORF cDNAs. The genomes of nine ascidian species have been sequenced since the release of the C. intestinalis genome. In ANISEED 2015, all nine new ascidian species can be explored via dedicated genome browsers, and searched by Blast. In addition, ANISEED provides full functional gene annotation, anatomical ontologies and some gene expression data for the six species with highest quality genomes. ANISEED is publicly available at: http://www.aniseed.cnrs.fr.


Asunto(s)
Ciona intestinalis/embriología , Ciona intestinalis/genética , Bases de Datos Genéticas , Urocordados/embriología , Urocordados/genética , Animales , Desarrollo Embrionario/genética , Genómica , Urocordados/anatomía & histología
6.
Adv Exp Med Biol ; 1029: 15-24, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29542077

RESUMEN

Phallusia mammillata has recently emerged as a new ascidian model. Its unique characteristics, including the optical transparency of eggs and embryos and efficient translation of exogenously introduced mRNA in eggs, make the Phallusia system suitable for fluorescent protein (FP)-based imaging approaches. In addition, genomic and transcriptomic resources are readily available for this ascidian species, facilitating functional gene studies. Microinjection is probably the most versatile technique for introducing exogenous molecules such as plasmids, mRNAs, and proteins into ascidian eggs/embryos. However, it is not practiced widely within the community; presumably, because the system is rather laborious to set up and it requires practice. Here, we describe in as much detail as possible two microinjection methods that we use daily in the laboratory: one based on an inverted microscope and the other on a stereomicroscope. Along the stepwise description of system setup and injection procedure, we provide practical tips in the hope that this chapter might be a useful guide for introducing or improving a microinjection setup.


Asunto(s)
Animales Modificados Genéticamente , Técnicas de Transferencia de Gen , Microinyecciones/métodos , ARN Mensajero/administración & dosificación , Urocordados/genética , Animales , Animales Modificados Genéticamente/embriología , Animales Modificados Genéticamente/genética , Animales Modificados Genéticamente/crecimiento & desarrollo , Técnicas de Cultivo de Embriones/instrumentación , Técnicas de Cultivo de Embriones/métodos , Embrión no Mamífero/ultraestructura , Femenino , Fertilización In Vitro/instrumentación , Fertilización In Vitro/métodos , Técnicas de Transferencia de Gen/instrumentación , Larva , Masculino , Microinyecciones/instrumentación , Microscopía/instrumentación , Óvulo , ARN Mensajero/genética , Transgenes , Urocordados/embriología , Urocordados/crecimiento & desarrollo
7.
Development ; 140(22): 4583-93, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24194472

RESUMEN

The fertilising sperm triggers a transient Ca(2+) increase that releases eggs from cell cycle arrest in the vast majority of animal eggs. In vertebrate eggs, Erp1, an APC/C(cdc20) inhibitor, links release from metaphase II arrest with the Ca(2+) transient and its degradation is triggered by the Ca(2+)-induced activation of CaMKII. By contrast, many invertebrate groups have mature eggs that arrest at metaphase I, and these species do not possess the CaMKII target Erp1 in their genomes. As a consequence, it is unknown exactly how cell cycle arrest at metaphase I is achieved and how the fertilisation Ca(2+) transient overcomes the arrest in the vast majority of animal species. Using live-cell imaging with a novel cyclin reporter to study cell cycle arrest and its release in urochordate ascidians, the closest living invertebrate group to the vertebrates, we have identified a new signalling pathway for cell cycle resumption in which CaMKII plays no part. Instead, we find that the Ca(2+)-activated phosphatase calcineurin (CN) is required for egg activation. Moreover, we demonstrate that parthenogenetic activation of metaphase I-arrested eggs by MEK inhibition, independent of a Ca(2+) increase, requires the activity of a second egg phosphatase: PP2A. Furthermore, PP2A activity, together with CN, is required for normal egg activation during fertilisation. As ascidians are a sister group of the vertebrates, we discuss these findings in relation to cell cycle arrest and egg activation in chordates.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Puntos de Control del Ciclo Celular , Meiosis , Óvulo/citología , Fosfoproteínas Fosfatasas/metabolismo , Urocordados/citología , Urocordados/enzimología , Ciclosoma-Complejo Promotor de la Anafase/antagonistas & inhibidores , Ciclosoma-Complejo Promotor de la Anafase/metabolismo , Animales , Antígenos Transformadores de Poliomavirus/metabolismo , Calcineurina/metabolismo , Inhibidores de la Calcineurina , Calcio/farmacología , Señalización del Calcio/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Ciclina B/metabolismo , Activación Enzimática/efectos de los fármacos , Fertilización/efectos de los fármacos , Mamíferos/metabolismo , Meiosis/efectos de los fármacos , Metafase/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Modelos Biológicos , Óvulo/enzimología , Proteína Fosfatasa 2/metabolismo , Ratas , Especificidad por Sustrato/efectos de los fármacos , Urocordados/efectos de los fármacos
8.
Dev Dyn ; 244(12): 1469-84, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26293695

RESUMEN

BACKGROUND: Establishment and maintenance of cell polarity is critical for normal embryonic development. Previously, it was thought that the echinoderm embryo remained relatively unpolarized until the first asymmetric division at the 16-cell stage. Here, we analyzed roles of the cell polarity regulators, the PAR complex proteins, and how their disruption in early development affects later developmental milestones. RESULTS: We found that PAR6, aPKC, and CDC42 localize to the apical cortex as early as the 2-cell stage and that this localization is retained through the gastrula stage. Of interest, PAR1 also colocalizes with these apical markers through the gastrula stage. Additionally, PAR1 was found to be in complex with aPKC, but not PAR6. PAR6, aPKC, and CDC42 are anchored in the cortical actin cytoskeleton by assembled myosin. Furthermore, assembled myosin was found to be necessary to maintain proper PAR6 localization through subsequent cleavage divisions. Interference with myosin assembly prevented the embryos from reaching the blastula stage, while transient disruptions of either actin or microtubules did not have this effect. CONCLUSIONS: These observations suggest that disruptions of the polarity in the early embryo can have a significant impact on the ability of the embryo to reach later critical stages in development.


Asunto(s)
Polaridad Celular/fisiología , Desarrollo Embrionario/fisiología , Erizos de Mar/embriología , Actinas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Proteínas de Ciclo Celular/metabolismo , Miosinas/metabolismo , Proteína Quinasa C/metabolismo , Erizos de Mar/metabolismo
9.
Dev Biol ; 384(2): 331-42, 2013 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-24140189

RESUMEN

During the transition from maternal to zygotic control of development, cell cycle length varies in different lineages, and this is important for their fates and functions. The maternal to zygotic transition (MZT) in metazoan embryos involves a profound remodeling of the cell cycle: S phase length increases then G2 is introduced. Although ß-catenin is the master regulator of endomesoderm patterning at MZT in all metazoans, the influence of maternal ß-catenin on the cell cycle at MZT remains poorly understood. By studying urochordate embryogenesis we found that cell cycle remodeling during MZT begins with the formation of 3 mitotic domains at the 16-cell stage arising from differential S phase lengthening, when endomesoderm is specified. Then, at the 64-cell stage, a G2 phase is introduced in the endoderm lineage during its specification. Strikingly, these two phases of cell cycle remodeling are patterned by ß-catenin-dependent transcription. Functional analysis revealed that, at the 16-cell stage, ß-catenin speeds up S phase in the endomesoderm. In contrast, two cell cycles later at gastrulation, nuclear ß-catenin induces endoderm fate and delays cell division. Such interphase lengthening in invaginating cells is known to be a requisite for gastrulation movements. Therefore, in basal chordates ß-catenin has a dual role to specify germ layers and remodel the cell cycle.


Asunto(s)
Ciclo Celular , Urocordados/embriología , Cigoto/metabolismo , beta Catenina/metabolismo , Animales , Secuencia de Bases , Cartilla de ADN , Femenino , Microscopía Fluorescente , Mitosis , Fase S , Cigoto/citología
10.
Biochem Biophys Res Commun ; 450(3): 1175-81, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24721426

RESUMEN

Like most metazoans, eggs of echinoderms and tunicates (marine deuterostomes, there is no data for the cephalochordates) arrest awaiting fertilization due to the activity of the Mos/MEK/MAPK cascade and are released from this cell cycle arrest by sperm-triggered Ca2+ signals. Invertebrate deuterostome eggs display mainly three distinct types of cell cycle arrest before fertilization mediated by potentially different cytostatic factors (CSF): one CSF causes arrest during meiotic metaphase I (MI-CSF in tunicates and some starfishes), another CSF likely causes arrest during meiotic metaphase II (amphioxus), and yet another form of CSF causes arrest to occur after meiotic exit during G1 of the first mitotic cycle (G1-CSF). In tunicates and echinoderms these different CSF activities have been shown to rely on the Mos//MAPK pathway for establishment and on Ca2+ signals for their inactivation. Despite these molecular similarities, release of MI-CSF arrest is caused by APC/C activation (to destroy cyclin B) whereas release from G1-CSF is caused by stimulating S phase and the synthesis of cyclins. Further research is needed to understand how both the Mos//MAPK cascade and Ca2+ achieve these tasks in different marine invertebrate deuterostomes. Another conserved feature of eggs is that protein synthesis of specific mRNAs is necessary to proceed through oocyte maturation and to maintain CSF-induced cell cycle arrest. Then activation of development at fertilization is accompanied by an increase in the rate of protein synthesis but the mechanisms involved are still largely unknown in most of the marine deuterostomes. How the sperm-triggered Ca2+ signals cause an increase in protein synthesis has been studied mainly in sea urchin eggs. Here we review these conserved features of eggs (arrest, activation and protein synthesis) focusing on the non-vertebrate deuterostomes.


Asunto(s)
Puntos de Control del Ciclo Celular/fisiología , Equinodermos/citología , Equinodermos/crecimiento & desarrollo , Urocordados/citología , Urocordados/crecimiento & desarrollo , Animales , Señalización del Calcio/fisiología , Equinodermos/fisiología , Femenino , Fertilización/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Oocitos/citología , Oocitos/crecimiento & desarrollo , Oocitos/fisiología , Biosíntesis de Proteínas/fisiología , Proteínas Proto-Oncogénicas c-mos/fisiología , Urocordados/fisiología , Cigoto/citología , Cigoto/crecimiento & desarrollo , Cigoto/fisiología
11.
Development ; 138(5): 885-95, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21303846

RESUMEN

Mos kinase is a universal mediator of oocyte meiotic maturation and is produced during oogenesis and destroyed after fertilization. The hallmark of maternal meiosis is that two successive M phases (meiosis I and II) drive two rounds of asymmetric cell division (ACD). However, how the egg limits the number of meioses to just two, thereby preventing gross aneuploidy, is poorly characterized. Here, in urochordate eggs, we show that loss of Mos/MAPK activity is necessary to prevent entry into meiosis III. Remarkably, maintaining the Mos/MAPK pathway active after fertilization at near physiological levels induces additional rounds of meiotic M phase (meiosis III, IV and V). During these additional rounds of meiosis, the spindle is positioned asymmetrically resulting in further rounds of ACD. In addition, inhibiting meiotic exit with Mos prevents pronuclear formation, cyclin A accumulation and maintains sperm-triggered Ca(2+) oscillations, all of which are hallmarks of the meiotic cell cycle in ascidians. It will be interesting to determine whether Mos availability in mammals can also control the number of meioses as it does in the urochordates. Our results demonstrate the power of urochordate eggs as a model to dissect the egg-to-embryo transition.


Asunto(s)
Meiosis , Óvulo/citología , Proteínas Proto-Oncogénicas c-mos/fisiología , Urocordados/citología , Animales , División Celular , Ciona intestinalis , Embrión no Mamífero , Sistema de Señalización de MAP Quinasas , Urocordados/embriología , Cigoto
12.
Methods Mol Biol ; 2740: 187-210, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38393477

RESUMEN

During eukaryotic cell division a microtubule-based structure, the mitotic spindle, aligns and segregates chromosomes between daughter cells. Understanding how this cellular structure is assembled and coordinated in space and in time requires measuring microtubule dynamics and visualizing spindle assembly with high temporal and spatial resolution. Visualization is often achieved by the introduction and the detection of molecular probes and fluorescence microscopy. Microtubules and mitotic spindles are highly conserved across eukaryotes; however, several technical limitations have restricted these investigations to only a few species. The ability to monitor microtubule and chromosome choreography in a wide range of species is fundamental to reveal conserved mechanisms or unravel unconventional strategies that certain forms of life have developed to ensure faithful partitioning of chromosomes during cell division. Here, we describe a technique based on injection of purified proteins that enables the visualization of microtubules and chromosomes with a high contrast in several divergent marine embryos. We also provide analysis methods and tools to extract microtubule dynamics and monitor spindle assembly. These techniques can be adapted to a wide variety of species in order to measure microtubule dynamics and spindle assembly kinetics when genetic tools are not available or in parallel to the development of such techniques in non-model organisms.


Asunto(s)
Microtúbulos , Huso Acromático , Huso Acromático/metabolismo , Microtúbulos/metabolismo , Ciclo Celular , División Celular , Cromosomas/metabolismo , Tubulina (Proteína)/metabolismo , Mitosis
13.
Development ; 137(12): 2011-21, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20463032

RESUMEN

Mitotic spindle orientation with respect to cortical polarity cues generates molecularly distinct daughter cells during asymmetric cell division (ACD). However, during ACD it remains unknown how the orientation of the mitotic spindle is regulated by cortical polarity cues until furrowing begins. In ascidians, the cortical centrosome-attracting body (CAB) generates three successive unequal cleavages and the asymmetric segregation of 40 localized postplasmic/PEM RNAs in germ cell precursors from the 8-64 cell stage. By combining fast 4D confocal fluorescence imaging with gene-silencing and classical blastomere isolation experiments, we show that spindle repositioning mechanisms are active from prometaphase until anaphase, when furrowing is initiated in B5.2 cells. We show that the vegetal-most spindle pole/centrosome is attracted towards the CAB during prometaphase, causing the spindle to position asymmetrically near the cortex. Next, during anaphase, the opposite spindle pole/centrosome is attracted towards the border with neighbouring B5.1 blastomeres, causing the spindle to rotate (10 degrees /minute) and migrate (3 microm/minute). Dynamic 4D fluorescence imaging of filamentous actin and plasma membrane shows that precise orientation of the cleavage furrow is determined by this second phase of rotational spindle displacement. Furthermore, in pairs of isolated B5.2 blastomeres, the second phase of rotational spindle displacement was lost. Finally, knockdown of PEM1, a protein localized in the CAB and required for unequal cleavage in B5.2 cells, completely randomizes spindle orientation. Together these data show that two separate mechanisms active during mitosis are responsible for spindle positioning, leading to precise orientation of the cleavage furrow during ACD in the cells that give rise to the germ lineage in ascidians.


Asunto(s)
Blastómeros/metabolismo , Centrosoma/metabolismo , Citoesqueleto/metabolismo , Huso Acromático/metabolismo , Urocordados/metabolismo , Actinas/genética , Actinas/metabolismo , Anafase , Animales , Blastómeros/citología , Ciclo Celular/genética , División Celular , Citoesqueleto/genética , Células Germinativas/metabolismo , Mitosis , Prometafase , Proteínas/genética , Proteínas/metabolismo , Huso Acromático/genética , Urocordados/citología
14.
Dev Growth Differ ; 55(4): 446-53, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23611302

RESUMEN

Decades of studies on ascidian embryogenesis have culminated in deciphering the first gene regulatory "blueprint" for the generation of all major larval tissue types in chordates. However, the current gene regulatory network (GRN) is not well integrated with the morphogenetic and cellular processes that are also taking place during embryogenesis. Describing these processes represents a major on-going challenge, aided by recent advances in imaging and fluorescent protein (FP) technologies. In this report, we describe the application of these technologies to the developmental biology of ascidians and provide a detailed practical guide on the preparation of ascidian embryos for imaging.


Asunto(s)
Biología Evolutiva/métodos , Urocordados/embriología , Animales , Ciona intestinalis/metabolismo , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes , Proteínas Fluorescentes Verdes/metabolismo , Microscopía Confocal/métodos , Imagen Molecular/métodos , Urocordados/genética
15.
Nat Cell Biol ; 5(11): 1023-5, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14593421

RESUMEN

Disjunction of pairs of homologous chromosomes during the first meiotic division (MI) requires anaphase-promoting complex (APC)-mediated activation of separase in budding yeast and Caenorhabditis elegans, but not Xenopus laevis. It is not clear which model best fits the mammalian system. Here we show that homologue disjunction in mouse oocytes is dependent on proteolysis of the separase inhibitor securin and the Cdk1 regulatory sub-unit cyclin B1. Proteolysis of both proteins was entirely dependent on their conserved destruction box (D-box) motifs, through which they are targeted to the APC. These data indicate that the mechanisms regulating homologue disjunction in mammalian oocytes are similar to those of budding yeast and C.elegans.


Asunto(s)
Ciclina B/metabolismo , Proteínas de Neoplasias/metabolismo , Oocitos/metabolismo , Animales , Ciclina B1 , Hidrólisis , Ratones , Securina
16.
Methods Mol Biol ; 2219: 217-230, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33074543

RESUMEN

Functional approaches for studying embryonic development have greatly advanced thanks to the CRISPR-Cas9 gene editing technique. Previously practiced in just a few organisms, these knockout techniques are now widely applied. Here we describe simple techniques for applying the CRISPR-Cas9 system to study the development of the nerve cord in the ascidian Phallusia mammillata.


Asunto(s)
Sistemas CRISPR-Cas , Edición Génica/métodos , Urocordados/embriología , Urocordados/genética , Animales , Microinyecciones , Urocordados/ultraestructura
17.
Elife ; 102021 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-34889186

RESUMEN

Cell division orientation is thought to result from a competition between cell geometry and polarity domains controlling the position of the mitotic spindle during mitosis. Depending on the level of cell shape anisotropy or the strength of the polarity domain, one dominates the other and determines the orientation of the spindle. Whether and how such competition is also at work to determine unequal cell division (UCD), producing daughter cells of different size, remains unclear. Here, we show that cell geometry and polarity domains cooperate, rather than compete, in positioning the cleavage plane during UCDs in early ascidian embryos. We found that the UCDs and their orientation at the ascidian third cleavage rely on the spindle tilting in an anisotropic cell shape, and cortical polarity domains exerting different effects on spindle astral microtubules. By systematically varying mitotic cell shape, we could modulate the effect of attractive and repulsive polarity domains and consequently generate predicted daughter cell size asymmetries and position. We therefore propose that the spindle position during UCD is set by the combined activities of cell geometry and polarity domains, where cell geometry modulates the effect of cortical polarity domain(s).


Asunto(s)
División Celular/fisiología , Polaridad Celular/fisiología , Forma de la Célula/fisiología , Embrión no Mamífero/fisiología , Desarrollo Embrionario/fisiología , Urocordados/fisiología , Animales
18.
Aquat Toxicol ; 232: 105768, 2021 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-33592501

RESUMEN

In recent years, pollution of surface waters with xenobiotic compounds became an issue of concern in society and has been the object of numerous studies. Most of these xenobiotic compounds are man-made molecules and some of them are qualified as endocrine disrupting chemicals (EDCs) when they interfere with hormones actions. Several studies have investigated the teratogenic impacts of EDCs in vertebrates (including marine vertebrates). However, the impact of such EDCs on marine invertebrates is much debated and still largely obscure. In addition, DNA-altering genotoxicants can induce embryonic malformations. The goal of this study is to develop a reliable and effective test for assessing toxicity of chemicals using embryos of the ascidian (Phallusia mammillata) in order to find phenotypic signatures associated with xenobiotics. We evaluated embryonic malformations with high-content analysis of larval phenotypes by scoring several quantitative and qualitative morphometric endpoints on a single image of Phallusia tadpole larvae with semi-automated image analysis. Using this approach we screened different classes of toxicants including genotoxicants, known or suspected EDCs and nuclear receptors (NRs) ligands. The screen presented here reveals a specific phenotypic signature for ligands of retinoic acid receptor/retinoid X receptor. Analysis of larval morphology combined with DNA staining revealed that embryos with DNA aberrations displayed severe malformations affecting multiple aspects of embryonic development. In contrast EDCs exposure induced no or little DNA aberrations and affected mainly neural development. Therefore the ascidian embryo/larval assay presented here can allow to distinguish the type of teratogenicity induced by different classes of toxicants.

19.
J Cell Biol ; 171(1): 47-59, 2005 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-16216922

RESUMEN

Cell cycle calcium signals are generated by the inositol trisphosphate (InsP3)-mediated release of calcium from internal stores (Ciapa, B., D. Pesando, M. Wilding, and M. Whitaker. 1994. Nature. 368:875-878; Groigno, L., and M. Whitaker. 1998. Cell. 92:193-204). The major internal calcium store is the endoplasmic reticulum (ER); thus, the spatial organization of the ER during mitosis may be important in shaping and defining calcium signals. In early Drosophila melanogaster embryos, ER surrounds the nucleus and mitotic spindle during mitosis, offering an opportunity to determine whether perinuclear localization of ER conditions calcium signaling during mitosis. We establish that the nuclear divisions in syncytial Drosophila embryos are accompanied by both cortical and nuclear localized calcium transients. Constructs that chelate InsP3 also prevent nuclear division. An analysis of nuclear calcium concentrations demonstrates that they are differentially regulated. These observations demonstrate that mitotic calcium signals in Drosophila embryos are confined to mitotic microdomains and offer an explanation for the apparent absence of detectable global calcium signals during mitosis in some cell types.


Asunto(s)
Señalización del Calcio , Ciclo Celular/fisiología , Drosophila/embriología , Retículo Endoplásmico/metabolismo , Microdominios de Membrana/fisiología , Membrana Nuclear/metabolismo , Animales , Drosophila/citología , Embrión no Mamífero , Retículo Endoplásmico/química , Microdominios de Membrana/metabolismo , Microscopía Confocal , Mitosis/fisiología , Membrana Nuclear/química
20.
Genes (Basel) ; 11(12)2020 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-33255457

RESUMEN

Polar body (PB) formation is an extreme form of unequal cell division that occurs in oocytes due to the eccentric position of the small meiotic spindle near the oocyte cortex. Prior to PB formation, a chromatin-centered process causes the cortex overlying the meiotic chromosomes to become polarized. This polarized cortical subdomain marks the site where a cortical protrusion or outpocket forms at the oocyte surface creating the future PBs. Using ascidians, we observed that PB1 becomes tethered to the fertilized egg via PB2, indicating that the site of PB1 cytokinesis directed the precise site for PB2 emission. We therefore studied whether the midbody remnant left behind following PB1 emission was involved, together with the egg chromatin, in defining the precise cortical site for PB2 emission. During outpocketing of PB2 in ascidians, we discovered that a small structure around 1 µm in diameter protruded from the cortical outpocket that will form the future PB2, which we define as the "polar corps". As emission of PB2 progressed, this small polar corps became localized between PB2 and PB1 and appeared to link PB2 to PB1. We tested the hypothesis that this small polar corps on the surface of the forming PB2 outpocket was the midbody remnant from the previous round of PB1 cytokinesis. We had previously discovered that Plk1::Ven labeled midbody remnants in ascidian embryos. We therefore used Plk1::Ven to follow the dynamics of the PB1 midbody remnant during meiosis II. Plk1::Ven strongly labeled the small polar corps that formed on the surface of the cortical outpocket that created PB2. Following emission of PB2, this polar corps was rich in Plk1::Ven and linked PB2 to PB1. By labelling actin (with TRITC-Phalloidin) we also demonstrated that actin accumulates at the midbody remnant and also forms a cortical cap around the midbody remnant in meiosis II that prefigured the precise site of cortical outpocketing during PB2 emission. Phalloidin staining of actin and immunolabelling of anti-phospho aPKC during meiosis II in fertilized eggs that had PB1 removed suggested that the midbody remnant remained within the fertilized egg following emission of PB1. Dynamic imaging of microtubules labelled with Ens::3GFP, MAP7::GFP or EB3::3GFP showed that one pole of the second meiotic spindle was located near the midbody remnant while the other pole rotated away from the cortex during outpocketing. Finally, we report that failure of the second meiotic spindle to rotate can lead to the formation of two cortical outpockets at anaphase II, one above each set of chromatids. It is not known whether the midbody remnant of PB1 is involved in directing the precise location of PB2 since our data are correlative in ascidians. However, a review of the literature indicates that PB1 is tethered to the egg surface via PB2 in several species including members of the cnidarians, lophotrochozoa and echinoids, suggesting that the midbody remnant formed during PB1 emission may be involved in directing the precise site of PB2 emission throughout the invertebrates.


Asunto(s)
Meiosis/fisiología , Cuerpos Polares/fisiología , Actinas/metabolismo , Animales , Bivalvos/metabolismo , Bivalvos/fisiología , Cromatina/metabolismo , Cromatina/fisiología , Cromosomas/metabolismo , Cromosomas/fisiología , Citocinesis/fisiología , Oocitos/metabolismo , Oocitos/fisiología , Cuerpos Polares/metabolismo , Huso Acromático/metabolismo , Huso Acromático/fisiología , Urocordados/metabolismo , Urocordados/fisiología , Cigoto/metabolismo , Cigoto/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA