Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 101
Filtrar
1.
Annu Rev Cell Dev Biol ; 30: 465-502, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25000993

RESUMO

Neural stem and progenitor cells have a central role in the development and evolution of the mammalian neocortex. In this review, we first provide a set of criteria to classify the various types of cortical stem and progenitor cells. We then discuss the issue of cell polarity, as well as specific subcellular features of these cells that are relevant for their modes of division and daughter cell fate. In addition, cortical stem and progenitor cell behavior is placed into a tissue context, with consideration of extracellular signals and cell-cell interactions. Finally, the differences across species regarding cortical stem and progenitor cells are dissected to gain insight into key developmental and evolutionary mechanisms underlying neocortex expansion.


Assuntos
Neocórtex/crescimento & desenvolvimento , Neurogênese/fisiologia , Animais , Divisão Celular Assimétrica , Compartimento Celular , Linhagem da Célula , Membrana Celular/fisiologia , Núcleo Celular/fisiologia , Polaridade Celular , Líquido Cefalorraquidiano/fisiologia , Humanos , Junções Intercelulares/fisiologia , Ventrículos Laterais/embriologia , Lipídeos de Membrana/metabolismo , Microglia/fisiologia , Mitose , Neocórtex/citologia , Neocórtex/embriologia , Células-Tronco Neurais/classificação , Células-Tronco Neurais/fisiologia , Células Neuroepiteliais/citologia , Células Neuroepiteliais/fisiologia , Neurônios/fisiologia , Organelas/fisiologia , Especificidade da Espécie
2.
J Cell Sci ; 137(7)2024 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-38469748

RESUMO

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.


Assuntos
Sêmen , Fuso Acromático , Masculino , Humanos , Microtúbulos , Citoplasma , Divisão Celular
3.
Development ; 150(19)2023 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-37756590

RESUMO

Successful nuclear migration through constricted spaces between cells or in the extracellular matrix relies on the ability of the nucleus to deform. Little is known about how this takes place in vivo. We have studied confined nuclear migration in Caenorhabditis elegans larval P cells, which is mediated by the LINC complex to pull nuclei towards the minus ends of microtubules. Null mutations of the LINC component unc-84 lead to a temperature-dependent phenotype, suggesting a parallel pathway for P-cell nuclear migration. A forward genetic screen for enhancers of unc-84 identified cgef-1 (CDC-42 guanine nucleotide exchange factor). Knockdown of CDC-42 in the absence of the LINC complex led to a P-cell nuclear migration defect. Expression of constitutively active CDC-42 partially rescued nuclear migration in cgef-1; unc-84 double mutants, suggesting that CDC-42 functions downstream of CGEF-1. The Arp2/3 complex and non-muscle myosin II (NMY-2) were also found to function parallel to the LINC pathway. In our model, CGEF-1 activates CDC-42, which induces actin polymerization through the Arp2/3 complex to deform the nucleus during nuclear migration, and NMY-2 helps to push the nucleus through confined spaces.


Assuntos
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Animais , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Actinas/metabolismo , Núcleo Celular/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Membrana Nuclear/metabolismo
4.
Dev Dyn ; 2024 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-38924277

RESUMO

BACKGROUND: Sex-specific morphogenesis occurs in Caenorhabditis elegans in the vulva of the hermaphrodite and in the male tail during the last larval stage. Temporal progression of vulva morphogenesis has been described in fine detail. However, a similar precise description of male tail morphogenesis was lacking. RESULTS: We here describe morphogenesis of the male tail at time points matching vulva development with special focus on morphogenesis of the tail tip. Using fluorescent reporters, we follow changes in cell shapes, cell fusions, nuclear migration, modifications in the basement membrane, and formation of a new apical extracellular matrix at the end of the tail. CONCLUSION: Our analysis answers two open questions about tail tip morphogenesis (TTM) by showing that one of the four tail tip cells, hyp11, remains largely separate, while the other cells fully fuse with each other and with two additional tail cells to form a ventral tail syncytium. This merger of cells begins at the apical surface early during TTM but is only completed toward the end of the process. This work provides a framework for future investigations of cell biological factors that drive male tail morphogenesis.

5.
Dev Biol ; 494: 60-70, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36509125

RESUMO

Neuroepithelial cells balance tissue growth requirement with the morphogenetic imperative of closing the neural tube. They apically constrict to generate mechanical forces which elevate the neural folds, but are thought to apically dilate during mitosis. However, we previously reported that mitotic neuroepithelial cells in the mouse posterior neuropore have smaller apical surfaces than non-mitotic cells. Here, we document progressive apical enrichment of non-muscle myosin-II in mitotic, but not non-mitotic, neuroepithelial cells with smaller apical areas. Live-imaging of the chick posterior neuropore confirms apical constriction synchronised with mitosis, reaching maximal constriction by anaphase, before division and re-dilation. Mitotic apical constriction amplitude is significantly greater than interphase constrictions. To investigate conservation in humans, we characterised early stages of iPSC differentiation through dual SMAD-inhibition to robustly produce pseudostratified neuroepithelia with apically enriched actomyosin. These cultured neuroepithelial cells achieve an equivalent apical area to those in mouse embryos. iPSC-derived neuroepithelial cells have large apical areas in G2 which constrict in M phase and retain this constriction in G1/S. Given that this differentiation method produces anterior neural identities, we studied the anterior neuroepithelium of the elevating mouse mid-brain neural tube. Instead of constricting, mid-brain mitotic neuroepithelial cells have larger apical areas than interphase cells. Tissue geometry differs between the apically convex early midbrain and flat posterior neuropore. Culturing human neuroepithelia on equivalently convex surfaces prevents mitotic apical constriction. Thus, neuroepithelial cells undergo high-amplitude apical constriction synchronised with cell cycle progression but the timing of their constriction if influenced by tissue geometry.


Assuntos
Mitose , Sistema Nervoso , Humanos , Animais , Camundongos , Constrição , Ciclo Celular , Diferenciação Celular/fisiologia
6.
Acta Neuropathol ; 147(1): 13, 2024 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-38194050

RESUMO

The development of the cerebral cortex involves a series of dynamic events, including cell proliferation and migration, which rely on the motor protein dynein and its regulators NDE1 and NDEL1. While the loss of function in NDE1 leads to microcephaly-related malformations of cortical development (MCDs), NDEL1 variants have not been detected in MCD patients. Here, we identified two patients with pachygyria, with or without subcortical band heterotopia (SBH), carrying the same de novo somatic mosaic NDEL1 variant, p.Arg105Pro (p.R105P). Through single-cell RNA sequencing and spatial transcriptomic analysis, we observed complementary expression of Nde1/NDE1 and Ndel1/NDEL1 in neural progenitors and post-mitotic neurons, respectively. Ndel1 knockdown by in utero electroporation resulted in impaired neuronal migration, a phenotype that could not be rescued by p.R105P. Remarkably, p.R105P expression alone strongly disrupted neuronal migration, increased the length of the leading process, and impaired nucleus-centrosome coupling, suggesting a failure in nucleokinesis. Mechanistically, p.R105P disrupted NDEL1 binding to the dynein regulator LIS1. This study identifies the first lissencephaly-associated NDEL1 variant and sheds light on the distinct roles of NDE1 and NDEL1 in nucleokinesis and MCD pathogenesis.


Assuntos
Lisencefalia , Humanos , Lisencefalia/genética , Movimento Celular/genética , Proliferação de Células , Córtex Cerebral , Dineínas/genética , Proteínas de Transporte , Proteínas Associadas aos Microtúbulos/genética
7.
Plant Cell Physiol ; 64(9): 1106-1117, 2023 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-37421143

RESUMO

Plant cells lack centrosomes and instead utilize acentrosomal microtubule organizing centers (MTOCs) to rapidly increase the number of microtubules at the onset of spindle assembly. Although several proteins required for MTOC formation have been identified, how the MTOC is positioned at the right place is not known. Here, we show that the inner nuclear membrane protein SUN2 is required for MTOC association with the nuclear envelope (NE) during mitotic prophase in the moss Physcomitrium patens. In actively dividing protonemal cells, microtubules accumulate around the NE during prophase. In particular, regional MTOC is formed at the apical surface of the nucleus. However, microtubule accumulation around the NE was impaired and apical MTOCs were mislocalized in sun2 knockout cells. Upon NE breakdown, the mitotic spindle was assembled with mislocalized MTOCs. However, completion of chromosome alignment in the spindle was delayed; in severe cases, the chromosome was transiently detached from the spindle body. SUN2 tended to localize to the apical surface of the nucleus during prophase in a microtubule-dependent manner. Based on these results, we propose that SUN2 facilitates the attachment of microtubules to chromosomes during spindle assembly by localizing microtubules to the NE. MTOC mispositioning was also observed during the first division of the gametophore tissue. Thus, this study suggests that microtubule-nucleus linking, a well-known function of SUN in animals and yeast, is conserved in plants.


Assuntos
Bryopsida , Membrana Nuclear , Animais , Membrana Nuclear/metabolismo , Centro Organizador dos Microtúbulos/metabolismo , Microtúbulos/metabolismo , Fuso Acromático/metabolismo , Cromossomos , Bryopsida/genética
8.
Development ; 147(20)2020 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-32994164

RESUMO

Between embryonic days 10.5 and 14.5, active proliferation drives rapid elongation of the murine midgut epithelial tube. Within this pseudostratified epithelium, nuclei synthesize DNA near the basal surface and move apically to divide. After mitosis, the majority of daughter cells extend a long, basally oriented filopodial protrusion, building a de novo path along which their nuclei can return to the basal side. WNT5A, which is secreted by surrounding mesenchymal cells, acts as a guidance cue to orchestrate this epithelial pathfinding behavior, but how this signal is received by epithelial cells is unknown. Here, we have investigated two known WNT5A receptors: ROR2 and RYK. We found that epithelial ROR2 is dispensable for midgut elongation. However, loss of Ryk phenocopies the Wnt5a-/- phenotype, perturbing post-mitotic pathfinding and leading to apoptosis. These studies reveal that the ligand-receptor pair WNT5A-RYK acts as a navigation system to instruct filopodial pathfinding, a process that is crucial for continuous cell cycling to fuel rapid midgut elongation.


Assuntos
Sistema Digestório/crescimento & desenvolvimento , Sistema Digestório/metabolismo , Pseudópodes/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Animais , Apoptose , Núcleo Celular/metabolismo , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Epitélio/metabolismo , Feminino , Masculino , Mesoderma/metabolismo , Camundongos Endogâmicos C57BL , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/metabolismo
9.
Proc Natl Acad Sci U S A ; 117(51): 32757-32763, 2020 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-33288691

RESUMO

After eukaryotic fertilization, gamete nuclei migrate to fuse parental genomes in order to initiate development of the next generation. In most animals, microtubules control female and male pronuclear migration in the zygote. Flowering plants, on the other hand, have evolved actin filament (F-actin)-based sperm nuclear migration systems for karyogamy. Flowering plants have also evolved a unique double-fertilization process: two female gametophytic cells, the egg and central cells, are each fertilized by a sperm cell. The molecular and cellular mechanisms of how flowering plants utilize and control F-actin for double-fertilization events are largely unknown. Using confocal microscopy live-cell imaging with a combination of pharmacological and genetic approaches, we identified factors involved in F-actin dynamics and sperm nuclear migration in Arabidopsis thaliana (Arabidopsis) and Nicotiana tabacum (tobacco). We demonstrate that the F-actin regulator, SCAR2, but not the ARP2/3 protein complex, controls the coordinated active F-actin movement. These results imply that an ARP2/3-independent WAVE/SCAR-signaling pathway regulates F-actin dynamics in female gametophytic cells for fertilization. We also identify that the class XI myosin XI-G controls active F-actin movement in the Arabidopsis central cell. XI-G is not a simple transporter, moving cargos along F-actin, but can generate forces that control the dynamic movement of F-actin for fertilization. Our results provide insights into the mechanisms that control gamete nuclear migration and reveal regulatory pathways for dynamic F-actin movement in flowering plants.


Assuntos
Complexo 2-3 de Proteínas Relacionadas à Actina/metabolismo , Proteínas de Arabidopsis/metabolismo , Arabidopsis/metabolismo , Miosinas/metabolismo , Nicotiana/metabolismo , Actinas/metabolismo , Arabidopsis/genética , Proteínas de Arabidopsis/genética , Núcleo Celular/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Magnoliopsida/metabolismo , Miosinas/genética , Óvulo Vegetal/metabolismo , Plantas Geneticamente Modificadas , Pólen/metabolismo
10.
J Cell Sci ; 132(13)2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-31182644

RESUMO

Cellular generation of mechanical forces required to close the presumptive spinal neural tube, the 'posterior neuropore' (PNP), involves interkinetic nuclear migration (INM) and apical constriction. Both processes change the apical surface area of neuroepithelial cells, but how they are biomechanically integrated is unknown. Rho kinase (Rock; herein referring to both ROCK1 and ROCK2) inhibition in mouse whole embryo culture progressively widens the PNP. PNP widening is not caused by increased mechanical tension opposing closure, as evidenced by diminished recoil following laser ablation. Rather, Rock inhibition diminishes neuroepithelial apical constriction, producing increased apical areas in neuroepithelial cells despite diminished tension. Neuroepithelial apices are also dynamically related to INM progression, with the smallest dimensions achieved in cells positive for the pan-M phase marker Rb phosphorylated at S780 (pRB-S780). A brief (2 h) Rock inhibition selectively increases the apical area of pRB-S780-positive cells, but not pre-anaphase cells positive for phosphorylated histone 3 (pHH3+). Longer inhibition (8 h, more than one cell cycle) increases apical areas in pHH3+ cells, suggesting cell cycle-dependent accumulation of cells with larger apical surfaces during PNP widening. Consequently, arresting cell cycle progression with hydroxyurea prevents PNP widening following Rock inhibition. Thus, Rock-dependent apical constriction compensates for the PNP-widening effects of INM to enable progression of closure.This article has an associated First Person interview with the first authors of the paper.


Assuntos
Divisão Celular , Tubo Neural/citologia , Tubo Neural/metabolismo , Quinases Associadas a rho/metabolismo , Actomiosina/metabolismo , Animais , Ciclo Celular/efeitos dos fármacos , Embrião de Mamíferos/citologia , Camundongos , Células Neuroepiteliais/citologia , Células Neuroepiteliais/efeitos dos fármacos , Células Neuroepiteliais/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Quinases Associadas a rho/antagonistas & inibidores
11.
Development ; 145(23)2018 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-30413561

RESUMO

The caspase-mediated regulation of many cellular processes, including apoptosis, justifies the substantial interest in understanding all of the biological features of these enzymes. To complement functional assays, it is crucial to identify caspase-activating cells in live tissues. Our work describes novel initiator caspase reporters that, for the first time, provide direct information concerning the initial steps of the caspase activation cascade in Drosophila tissues. One of our caspase sensors capitalises on the rapid subcellular localisation change of a fluorescent marker to uncover novel cellular apoptotic events relating to the actin-mediated positioning of the nucleus before cell delamination. The other construct benefits from caspase-induced nuclear translocation of a QF transcription factor. This feature enables the genetic manipulation of caspase-activating cells and reveals the spatiotemporal patterns of initiator caspase activity. Collectively, our sensors offer experimental opportunities not available by using previous reporters and have proven useful to illuminate previously unknown aspects of caspase-dependent processes in apoptotic and non-apoptotic cellular scenarios.


Assuntos
Caspases/metabolismo , Drosophila melanogaster/citologia , Drosophila melanogaster/metabolismo , Genes Reporter , Animais , Apoptose , Movimento Celular , Núcleo Celular/metabolismo , Sobrevivência Celular , Rastreamento de Células , Sistema Digestório/metabolismo , Proteínas de Drosophila/metabolismo , Ativação Enzimática , Feminino , Fatores de Tempo , Imagem com Lapso de Tempo , Asas de Animais/citologia
12.
Development ; 145(14)2018 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-29945866

RESUMO

Many proliferative epithelia are pseudostratified because of cell cycle-dependent interkinetic nuclear migration (IKNM, basal during G1 and apical during G2). Although most epithelia, including early embryonic neuroepithelia (≤100 µm thick), undergo IKNM over the entire apicobasal extent, more apicobasally elongated (300 µm) neural progenitor cells (radial glial cells) in the mid-embryonic mouse cerebral wall move their nuclei only within its apical (100 µm) compartment, leaving the remaining basal region nucleus-free (fiber-like). How this IKNM range [i.e. the thickness of a pseudostratified ventricular zone (VZ)] is determined remains unknown. Here, we report external fencing of IKNM and the VZ by differentiating cells. When a tight stack of multipolar cells immediately basal to the VZ was 'drilled' via acute neuron-directed expression of diphtheria toxin, IKNM of apicobasally connected progenitor cells continued further towards the basal region of the cell (200 µm). The unfencing-induced basally overshot nuclei stay in S phase for too long and do not move apically, suggesting that external limitation of IKNM is necessary for progenitors to undergo normal cytogenetic behaviors. Thus, physical collaboration between progenitors and differentiating cells, including neurons, underlies brain development.


Assuntos
Diferenciação Celular/fisiologia , Núcleo Celular/metabolismo , Proliferação de Células/fisiologia , Neuroglia/metabolismo , Células-Tronco/metabolismo , Animais , Núcleo Celular/genética , Camundongos , Camundongos Endogâmicos ICR , Neuroglia/citologia , Células-Tronco/citologia
13.
Development ; 145(21)2018 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-30401784

RESUMO

Embryonic development of the central nervous system (CNS) requires the proliferation of neural progenitor cells to be tightly regulated, allowing the formation of an organ with the right size and shape. This includes regulation of both the spatial distribution of mitosis and the mode of cell division. The centrosome, which is the main microtubule-organizing centre of animal cells, contributes to both of these processes. Here, we discuss the impact that centrosome-mediated control of cell division has on the shape of the overall growing CNS. We also review the intrinsic properties of the centrosome, both in terms of its molecular composition and its signalling capabilities, and discuss the fascinating notion that intrinsic centrosomal asymmetries in dividing neural progenitor cells are instructive for neurogenesis. Finally, we discuss the genetic links between centrosome dysfunction during development and the aetiology of microcephaly.


Assuntos
Sistema Nervoso Central/crescimento & desenvolvimento , Sistema Nervoso Central/metabolismo , Centrossomo/metabolismo , Animais , Humanos , Microcefalia/patologia , Mitose , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Neurogênese
14.
J Exp Zool B Mol Dev Evol ; 336(8): 642-651, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-32638525

RESUMO

Flowering plants (angiosperms) perform a unique double fertilization in which two sperm cells fuse with two female gamete cells in the embryo sac to develop a seed. Furthermore, during land plant evolution, the mode of sexual reproduction has been modified dramatically from motile sperm in the early-diverging land plants, such as mosses and ferns as well as some gymnosperms (Ginkgo and cycads) to nonmotile sperm that are delivered to female gametes by the pollen tube in flowering plants. Recent studies have revealed the cellular dynamics and molecular mechanisms for the complex series of double fertilization processes and elucidated differences and similarities between animals and plants. Here, together with a brief comparison with animals, we review the current understanding of flowering plant zygote dynamics, covering from gamete nuclear migration, karyogamy, and polyspermy block, to zygotic genome activation as well as asymmetrical division of the zygote. Further analyses of the detailed molecular and cellular mechanisms of flowering plant fertilization should shed light on the evolution of the unique sexual reproduction of flowering plants.


Assuntos
Magnoliopsida , Sementes/crescimento & desenvolvimento , Animais , Fertilização , Células Germinativas , Magnoliopsida/embriologia , Zigoto
15.
Semin Cell Dev Biol ; 82: 3-16, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29241689

RESUMO

Nuclear movement within a cell occurs in a variety of eukaryotic organisms including yeasts and filamentous fungi. Fungal molecular genetic studies identified the minus-end-directed microtubule motor cytoplasmic dynein as a critical protein for nuclear movement or orientation of the mitotic spindle contained in the nucleus. Studies in the budding yeast first indicated that dynein anchored at the cortex via its anchoring protein Num1 exerts pulling force on an astral microtubule to orient the anaphase spindle across the mother-daughter axis before nuclear division. Prior to anaphase, myosin V interacts with the plus end of an astral microtubule via Kar9-Bim1/EB1 and pulls the plus end along the actin cables to move the nucleus/spindle close to the bud neck. In addition, pushing or pulling forces generated from cortex-linked polymerization or depolymerization of microtubules drive nuclear movements in yeasts and possibly also in filamentous fungi. In filamentous fungi, multiple nuclei within a hyphal segment undergo dynein-dependent back-and-forth movements and their positioning is also influenced by cytoplasmic streaming toward the hyphal tip. In addition, nuclear movement occurs at various stages of fungal development and fungal infection of plant tissues. This review discusses our current understanding on the mechanisms of nuclear movement in fungal organisms, the importance of nuclear positioning and the regulatory strategies that ensure the proper positioning of nucleus/spindle.


Assuntos
Núcleo Celular/metabolismo , Fungos/patogenicidade
16.
J Cell Sci ; 131(16)2018 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-30054381

RESUMO

Pericentrin (Pcnt) is a multifunctional scaffold protein and mutations in the human PCNT gene are associated with several diseases, including ciliopathies. Pcnt plays a crucial role in ciliary development in olfactory receptor neurons, but its function in the photoreceptor-connecting cilium is unknown. We downregulated Pcnt in the retina ex vivo and in vivo via a virus-based RNA interference approach to study Pcnt function in photoreceptors. ShRNA-mediated knockdown of Pcnt impaired the development of the connecting cilium and the outer segment of photoreceptors, and caused a nuclear migration defect. In protein interaction screens, we found that the outer nuclear membrane protein Syne-2 (also known as Nesprin-2) is an interaction partner of Pcnt in photoreceptors. Syne-2 is important for positioning murine photoreceptor cell nuclei and for centrosomal migration during early ciliogenesis. CRISPR/Cas9-mediated knockout of Syne-2 in cell culture led to an overexpression and mislocalization of Pcnt and to ciliogenesis defects. Our findings suggest that the Pcnt-Syne-2 complex is important for ciliogenesis and outer segment formation during retinal development and plays a role in nuclear migration.


Assuntos
Antígenos/fisiologia , Cílios/fisiologia , Proteínas dos Microfilamentos/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Proteínas Nucleares/fisiologia , Organogênese/genética , Animais , Antígenos/genética , Sistemas CRISPR-Cas , Células Cultivadas , Cílios/genética , Feminino , Técnicas de Inativação de Genes , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas dos Microfilamentos/genética , Células NIH 3T3 , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Retina/embriologia , Retina/metabolismo
17.
Cell Biol Int ; 44(6): 1262-1266, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31486567

RESUMO

This survey paper contains a brief analysis of publications included in the special issue of the scientific journal Cell Biology International titled "Plant Cytoskeleton Structure, Dynamics and Functions". The manuscripts in this special issue reflect some new aspects of plant cytoskeleton organization, signaling and functioning, and results from different Ukrainian research groups, and focuses on bringing together scientists working across different instrumental scales.


Assuntos
Citoesqueleto/metabolismo , Microtúbulos/metabolismo , Plantas/ultraestrutura , Tubulina (Proteína)/metabolismo , Tubulina (Proteína)/genética
18.
J Cell Sci ; 130(22): 3862-3877, 2017 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-28982714

RESUMO

Homeostasis of renewing tissues requires balanced proliferation, differentiation and movement. This is particularly important in the intestinal epithelium where lineage tracing suggests that stochastic differentiation choices are intricately coupled to the position of a cell relative to a niche. To determine how position is achieved, we followed proliferating cells in intestinal organoids and discovered that the behaviour of mitotic sisters predicted long-term positioning. We found that, normally, 70% of sisters remain neighbours, while 30% lose contact and separate after cytokinesis. These post-mitotic placements predict longer term differences in positions assumed by sisters: adjacent sisters reach similar positions over time; in a pair of separating sisters, one remains close to its birthplace while the other is displaced upward. Computationally modelling crypt dynamics confirmed that post-mitotic separation leads to sisters reaching different compartments. We show that interkinetic nuclear migration, cell size and asymmetric tethering by a process extending from the basal side of cells contribute to separations. These processes are altered in adenomatous polyposis coli (Apc) mutant epithelia where separation is lost. We conclude that post-mitotic placement contributes to stochastic niche exit and, when defective, supports the clonal expansion of Apc mutant cells.


Assuntos
Núcleo Celular/fisiologia , Mucosa Intestinal/citologia , Animais , Transporte Biológico , Adesão Celular , Humanos , Cinética , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mitose , Organoides/citologia , Técnicas de Cultura de Tecidos
19.
Biochem Biophys Res Commun ; 519(4): 783-789, 2019 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-31551150

RESUMO

Nuclear migration in Arabidopsis root hairs is bidirectional and relies on actin filaments. However, how actin filaments regulate the bidirectional movement of nuclei remains unclear. Here, we discovered that nuclei migrate forward and backward according to the developmental stage of the hair cells. In addition, the migration direction of nuclei was not constant but reversed occasionally, accompanied by nuclear shape changes. Confocal microscopic analysis revealed that perinuclear actin bundles were closely related to the migration and shape of hair cell nuclei. Pharmacological studies showed that SMIFH2, an inhibitor of the actin nucleator-formin, inhibited nuclear backward migration probably by impairing the perinuclear actin filaments. These data indicate that nuclear migration in hair cells is likely motivated by the competition of mechanical forces acting on the nucleus. Furthermore, the perinuclear actin filaments are closely related to the migration direction of hair cell nuclei.


Assuntos
Citoesqueleto de Actina/metabolismo , Arabidopsis/citologia , Núcleo Celular/metabolismo , Movimento , Citoesqueleto de Actina/efeitos dos fármacos , Arabidopsis/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Núcleo Celular/efeitos dos fármacos , Movimento/efeitos dos fármacos , Raízes de Plantas/citologia , Raízes de Plantas/efeitos dos fármacos , Tionas/farmacologia , Uracila/análogos & derivados , Uracila/farmacologia
20.
Development ; 143(22): 4193-4202, 2016 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-27697906

RESUMO

Cellular migrations through constricted spaces are a crucial aspect of many developmental and disease processes including hematopoiesis, inflammation and metastasis. A limiting factor in these events is nuclear deformation. Here, we establish an in vivo model in which nuclei can be visualized while moving through constrictions and use it to elucidate mechanisms for nuclear migration. C. elegans hypodermal P-cell larval nuclei traverse a narrow space that is about 5% their width. This constriction is blocked by fibrous organelles, structures that pass through P cells to connect the muscles to cuticle. Fibrous organelles are removed just prior to nuclear migration, when nuclei and lamins undergo extreme morphological changes to squeeze through the space. Both actin and microtubule networks are organized to mediate nuclear migration. The LINC complex, consisting of the SUN protein UNC-84 and the KASH protein UNC-83, recruits dynein and kinesin-1 to the nuclear surface. Both motors function in P-cell nuclear migration, but dynein, functioning through UNC-83, plays a more central role as nuclei migrate towards minus ends of polarized microtubule networks. Thus, the nucleoskeleton and cytoskeleton are coordinated to move nuclei through constricted spaces.


Assuntos
Citoesqueleto de Actina/metabolismo , Caenorhabditis elegans , Núcleo Celular/metabolismo , Derme/embriologia , Derme/metabolismo , Microtúbulos/metabolismo , Actinas/metabolismo , Animais , Animais Geneticamente Modificados , Transporte Biológico , Padronização Corporal , Caenorhabditis elegans/citologia , Caenorhabditis elegans/embriologia , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Citoesqueleto/metabolismo , Derme/ultraestrutura , Embrião não Mamífero
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa